title
stringlengths
1
322
pmid
stringlengths
7
8
background_abstract
stringlengths
7
2.19k
background_abstract_label
stringlengths
7
30
methods_abstract
stringlengths
18
2.32k
methods_abstract_label
stringlengths
5
43
results_abstract
stringlengths
35
2.65k
results_abstract_label
stringlengths
6
30
conclusions_abstract
stringlengths
33
1.83k
conclusions_abstract_label
stringlengths
6
45
mesh_descriptor_names
list
pmcid
stringlengths
5
8
background_title
stringlengths
10
95
background_text
stringlengths
107
140k
methods_title
stringlengths
4
129
methods_text
stringlengths
99
196k
results_title
stringlengths
6
172
results_text
stringlengths
112
685k
conclusions_title
stringlengths
8
58
conclusions_text
stringlengths
64
66.1k
other_sections_titles
list
other_sections_texts
list
other_sections_sec_types
list
all_sections_titles
list
all_sections_texts
list
all_sections_sec_types
list
keywords
list
The safety of esophageal cancer surgery during COVID-19.
36254035
The ongoing coronavirus disease 2019 (COVID-19) pandemic has drastically challenged the safety of on esophageal cancer (EC) surgery during COVID-19. The study aimed to evaluate the safety of EC surgery during the COVID-19 pandemic.
BACKGROUND
This systematic review was performed in accordance with the PRISMA-P 2015 guidelines and registered in PROSPERO (registration number: CRD42022335164). A systematic search of PubMed, Embase, Cochrane Library, Web of Science, Medline, Chinese National Knowledge Infrastructure database, Chinese Scientific Journal database, and Wan Fang database was conducted to identify potentially relevant publications from January 2020 to May 2022. All data were independently extracted by two researchers. We will apply a fixed-effect model or random effect model basis on the heterogeneity test and employ with RevMan 5.4.1 software for data synthesis. The dichotomous surgical outcomes used risk ratios or risk differences, and for continuous surgical outcomes, mean differences (MD) or standardized MD, both with 95% confidence intervals were used. The primary outcomes were postoperative complications, anastomotic leaks, and mortality. The secondary outcomes were total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, number of patients needing ICU stay, and 30-day readmission.
METHODS
This study will comprehensively summarize the high-quality trials to determine the safety of EC surgery during COVID-19.
RESULTS
Our systematic review and meta-analysis will present evidence for the safety of EC surgery during COVID-19.
CONCLUSION
[ "COVID-19", "Esophageal Neoplasms", "Humans", "Meta-Analysis as Topic", "Pandemics", "Systematic Reviews as Topic" ]
9575394
1. Introduction
The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has posed a serious public health threat worldwide, with millions of people at risk in a growing number of countries.[1]Up to May 22, 2022, over 522 million confirmed cases and over 6 million confirmed deaths have been reported to the World Health Organization from different countries, areas and territories. During the pandemic, the management of patients with cancer has been affected at multiple stages, including the triage decisions, surgery, and neoadjuvant therapy as a bridge to reduce admissions and preserve health-care resources.[2] COVID Surg Collaborative estimated that 28,404,603 elective operations were canceled or postponed worldwide during the 12 weeks of peak disruption, with 38% being for cancer.[3] Esophageal cancer (EC) is the 8th most common type of cancer worldwide, constitutes the sixth leading cause of cancer deaths.[4] EC remains a global health concern with a dismal prognosis and an estimated 5-year survival rate of approximately 10% to 15%.[5] Surgery plays an important role in the treatment strategies for EC. Recent advances in surgical techniques and perioperative management have dramatically improved the mortality rate; however, esophagectomy remains a highly invasive procedure that can lead to severe postoperative complications.[6] Besides, the rapid global spread of COVID-19 presents an unprecedented crisis for esophagectomy.[7] Recent reports suggest that patients with cancer might have a higher risk of COVID-19 than individuals without cancer and patients with cancer had poorer outcomes from COVID-19.[8] In particular, for patients with EC, additional precautions have been taken, as surgery for EC alone has higher morbidity and mortality rates compared to that for other oncological surgeries.[9] The complexity of the multidisciplinary approach to EC patients and the high morbidity rates of esophageal surgery have challenged the treatment pathways of these patients, especially during the COVID-19 pandemic. Overall, 55% of scheduled endoscopic resections for gastrointestinal neoplastic lesions were deferred globally after the lockdown period, which was 11 times higher than in the previous year, and the majority of postponements (80%) occurred in severely affected countries.[10] For patients with cancer, delay of surgery has the potential to increase the likelihood of metastatic disease, with some patients’ tumors progressing from being curable (with near-normal life expectancy) to noncurable (with limited life expectancy).[11] Delay in time to esophagectomy for EC has been shown to have worse perioperative and long-term outcomes. Therefore, general guidance from health ministries and national surgical associations supported that time-dependent surgery should continue.[12] Despite outbreaks, cancer surgery must continue to prevent an overwhelming number of delayed operations, a possible increase in emergency procedures, and a significant decline in population health.[3] However, there have been conflicting results regarding the safety of EC surgery during the COVID-19 pandemic. Some studies have demonstrated that EC surgical procedures may be safely performed during the pandemic.[13–17] while another study reported that one of the esophago-gastric junction cancer patients developed COVID-19 pneumonia on post-operative day two, leading to impaired respiratory function and increased pleural fluid collection from the chest tube, resulting in a prolonged hospital stay.[18] And the other study reported the case of a young man who underwent thoracoscopic subtotal esophagectomy for distal esophageal adenocarcinoma who developed COVID-19 with severe clinical presentation.[19] Modeling the impact of delaying surgery for early EC in the era of COVID-19 showed that as the risk of infection with COVID-19 increased above 7%, delaying operations for 3 months has an improved long-term survival.[20] While other study showed that the EC time to surgery over 8 weeks is associated with lower survival.[21] So it is necessary to evaluate the safety of EC surgery during COVID-19. To the best of our knowledge, there have been no meta-analyses on the safety of EC surgery during COVID-19. Therefore, we performed a meta-analysis of cohort studies to evaluate the safety of EC surgery during the COVID-19 pandemic.
2. Methods
[SUBTITLE] 2.1. Study registration [SUBSECTION] The present study was conducted in accordance with the preferred reporting items for systematic reviews and meta-analysis protocols statement guidelines[22] and has been registered with PROSPERO under registration number CRD42022335164. The present study was conducted in accordance with the preferred reporting items for systematic reviews and meta-analysis protocols statement guidelines[22] and has been registered with PROSPERO under registration number CRD42022335164. [SUBTITLE] 2.2. Selection criteria [SUBSECTION] [SUBTITLE] 2.2.1. Type of studies. [SUBSECTION] The present study is a cohort studies of the safety of EC surgery during COVID-19. The present study is a cohort studies of the safety of EC surgery during COVID-19. [SUBTITLE] 2.2.2. Types of participants. [SUBSECTION] The sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery. The sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery. [SUBTITLE] 2.2.3. Types of interventions and comparisons. [SUBSECTION] However, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19. However, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19. [SUBTITLE] 2.2.4. Language. [SUBSECTION] There is restriction on Chinese or English. There is restriction on Chinese or English. [SUBTITLE] 2.2.1. Type of studies. [SUBSECTION] The present study is a cohort studies of the safety of EC surgery during COVID-19. The present study is a cohort studies of the safety of EC surgery during COVID-19. [SUBTITLE] 2.2.2. Types of participants. [SUBSECTION] The sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery. The sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery. [SUBTITLE] 2.2.3. Types of interventions and comparisons. [SUBSECTION] However, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19. However, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19. [SUBTITLE] 2.2.4. Language. [SUBSECTION] There is restriction on Chinese or English. There is restriction on Chinese or English. [SUBTITLE] 2.3. Exclusion criteria [SUBSECTION] Studies that met the following criteria were excluded: duplicate studies; articles published as case reports, case series, review articles, letters, editorials, and commentaries will be excluded; studies involving data that cannot be extracted or inadequate are lacking; there is no control group that assesses the safety of EC surgery during the pre-COVID-19 period; no outcome measures of interest are reported; Newcastle-Ottawa Scale (NOS) scores <5 points; studies written in languages other than Chinese or English. Studies that met the following criteria were excluded: duplicate studies; articles published as case reports, case series, review articles, letters, editorials, and commentaries will be excluded; studies involving data that cannot be extracted or inadequate are lacking; there is no control group that assesses the safety of EC surgery during the pre-COVID-19 period; no outcome measures of interest are reported; Newcastle-Ottawa Scale (NOS) scores <5 points; studies written in languages other than Chinese or English. [SUBTITLE] 2.4. Types of outcome measures [SUBSECTION] [SUBTITLE] 2.4.1. Primary outcomes. [SUBSECTION] The primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19. The primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19. [SUBTITLE] 2.4.2. Secondary outcomes. [SUBSECTION] The secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19. The secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19. [SUBTITLE] 2.4.1. Primary outcomes. [SUBSECTION] The primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19. The primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19. [SUBTITLE] 2.4.2. Secondary outcomes. [SUBSECTION] The secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19. The secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19. [SUBTITLE] 2.5. Search strategy [SUBSECTION] A systematic search plan will be performed in the following eight databases with a time restriction from January 2020 to May 2022 to filter eligible studies: PubMed, Embase, the Cochrane Library, Web of Science, Medline, Chinese National Knowledge Infrastructure database, Chinese Scientific Journal database, and Wan Fang database. We also will search journal articles, conference papers, and academic papers. The search keywords included terms related to COVID-19 pandemic, terms related to EC and terms related to surgery. Cross references were checked to assess if any relevant studies were missed. We considered a specific search strategy in PubMed as a typical example; the specific steps of the retrieval are shown in Table 1. Search strategy in PubMed database. A systematic search plan will be performed in the following eight databases with a time restriction from January 2020 to May 2022 to filter eligible studies: PubMed, Embase, the Cochrane Library, Web of Science, Medline, Chinese National Knowledge Infrastructure database, Chinese Scientific Journal database, and Wan Fang database. We also will search journal articles, conference papers, and academic papers. The search keywords included terms related to COVID-19 pandemic, terms related to EC and terms related to surgery. Cross references were checked to assess if any relevant studies were missed. We considered a specific search strategy in PubMed as a typical example; the specific steps of the retrieval are shown in Table 1. Search strategy in PubMed database. [SUBTITLE] 2.6. Data collection and analysis [SUBSECTION] [SUBTITLE] 2.6.1. Study selection. [SUBSECTION] We will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1). The guideline flow diagram. We will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1). The guideline flow diagram. [SUBTITLE] 2.6.2. Data extraction. [SUBSECTION] Two reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission. Two reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission. [SUBTITLE] 2.6.3. Assessment of risk of bias. [SUBSECTION] NOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion. NOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion. [SUBTITLE] 2.6.4. Measures of perioperative outcomes. [SUBSECTION] For dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05. For dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05. [SUBTITLE] 2.6.5. Data analysis and heterogeneity processing. [SUBSECTION] We will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible. We will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible. [SUBTITLE] 2.6.6. Subgroup analysis. [SUBSECTION] Subgroup analyses were conducted based on different type of surgical methods. Subgroup analyses were conducted based on different type of surgical methods. [SUBTITLE] 2.6.7. Sensitivity analysis. [SUBSECTION] The stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis. The stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis. [SUBTITLE] 2.6.8. Assessment of reporting bias. [SUBSECTION] When we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32] When we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32] [SUBTITLE] 2.6.9. Ethics and dissemination. [SUBSECTION] This systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences. This systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences. [SUBTITLE] 2.6.10. Grading the quality of evidence. [SUBSECTION] The Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively. The Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively. [SUBTITLE] 2.6.1. Study selection. [SUBSECTION] We will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1). The guideline flow diagram. We will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1). The guideline flow diagram. [SUBTITLE] 2.6.2. Data extraction. [SUBSECTION] Two reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission. Two reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission. [SUBTITLE] 2.6.3. Assessment of risk of bias. [SUBSECTION] NOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion. NOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion. [SUBTITLE] 2.6.4. Measures of perioperative outcomes. [SUBSECTION] For dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05. For dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05. [SUBTITLE] 2.6.5. Data analysis and heterogeneity processing. [SUBSECTION] We will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible. We will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible. [SUBTITLE] 2.6.6. Subgroup analysis. [SUBSECTION] Subgroup analyses were conducted based on different type of surgical methods. Subgroup analyses were conducted based on different type of surgical methods. [SUBTITLE] 2.6.7. Sensitivity analysis. [SUBSECTION] The stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis. The stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis. [SUBTITLE] 2.6.8. Assessment of reporting bias. [SUBSECTION] When we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32] When we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32] [SUBTITLE] 2.6.9. Ethics and dissemination. [SUBSECTION] This systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences. This systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences. [SUBTITLE] 2.6.10. Grading the quality of evidence. [SUBSECTION] The Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively. The Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively.
null
null
null
null
[ "2.1. Study registration", "2.2. Selection criteria", "2.2.1. Type of studies.", "2.2.2. Types of participants.", "2.2.3. Types of interventions and comparisons.", "2.2.4. Language.", "2.3. Exclusion criteria", "2.4. Types of outcome measures", "2.4.1. Primary outcomes.", "2.4.2. Secondary outcomes.", "2.5. Search strategy", "2.6. Data collection and analysis", "2.6.1. Study selection.", "2.6.2. Data extraction.", "2.6.3. Assessment of risk of bias.", "2.6.4. Measures of perioperative outcomes.", "2.6.5. Data analysis and heterogeneity processing.", "2.6.6. Subgroup analysis.", "2.6.7. Sensitivity analysis.", "2.6.8. Assessment of reporting bias.", "2.6.9. Ethics and dissemination.", "2.6.10. Grading the quality of evidence.", "Author contributions" ]
[ "The present study was conducted in accordance with the preferred reporting items for systematic reviews and meta-analysis protocols statement guidelines[22] and has been registered with PROSPERO under registration number CRD42022335164.", "[SUBTITLE] 2.2.1. Type of studies. [SUBSECTION] The present study is a cohort studies of the safety of EC surgery during COVID-19.\nThe present study is a cohort studies of the safety of EC surgery during COVID-19.\n[SUBTITLE] 2.2.2. Types of participants. [SUBSECTION] The sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery.\nThe sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery.\n[SUBTITLE] 2.2.3. Types of interventions and comparisons. [SUBSECTION] However, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19.\nHowever, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19.\n[SUBTITLE] 2.2.4. Language. [SUBSECTION] There is restriction on Chinese or English.\nThere is restriction on Chinese or English.", "The present study is a cohort studies of the safety of EC surgery during COVID-19.", "The sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery.", "However, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19.", "There is restriction on Chinese or English.", "Studies that met the following criteria were excluded: duplicate studies; articles published as case reports, case series, review articles, letters, editorials, and commentaries will be excluded; studies involving data that cannot be extracted or inadequate are lacking; there is no control group that assesses the safety of EC surgery during the pre-COVID-19 period; no outcome measures of interest are reported; Newcastle-Ottawa Scale (NOS) scores <5 points; studies written in languages other than Chinese or English.", "[SUBTITLE] 2.4.1. Primary outcomes. [SUBSECTION] The primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19.\nThe primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19.\n[SUBTITLE] 2.4.2. Secondary outcomes. [SUBSECTION] The secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19.\nThe secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19.", "The primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19.", "The secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19.", "A systematic search plan will be performed in the following eight databases with a time restriction from January 2020 to May 2022 to filter eligible studies: PubMed, Embase, the Cochrane Library, Web of Science, Medline, Chinese National Knowledge Infrastructure database, Chinese Scientific Journal database, and Wan Fang database. We also will search journal articles, conference papers, and academic papers. The search keywords included terms related to COVID-19 pandemic, terms related to EC and terms related to surgery. Cross references were checked to assess if any relevant studies were missed. We considered a specific search strategy in PubMed as a typical example; the specific steps of the retrieval are shown in Table 1.\nSearch strategy in PubMed database.", "[SUBTITLE] 2.6.1. Study selection. [SUBSECTION] We will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1).\nThe guideline flow diagram.\nWe will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1).\nThe guideline flow diagram.\n[SUBTITLE] 2.6.2. Data extraction. [SUBSECTION] Two reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission.\nTwo reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission.\n[SUBTITLE] 2.6.3. Assessment of risk of bias. [SUBSECTION] NOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion.\nNOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion.\n[SUBTITLE] 2.6.4. Measures of perioperative outcomes. [SUBSECTION] For dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05.\nFor dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05.\n[SUBTITLE] 2.6.5. Data analysis and heterogeneity processing. [SUBSECTION] We will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible.\nWe will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible.\n[SUBTITLE] 2.6.6. Subgroup analysis. [SUBSECTION] Subgroup analyses were conducted based on different type of surgical methods.\nSubgroup analyses were conducted based on different type of surgical methods.\n[SUBTITLE] 2.6.7. Sensitivity analysis. [SUBSECTION] The stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis.\nThe stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis.\n[SUBTITLE] 2.6.8. Assessment of reporting bias. [SUBSECTION] When we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32]\nWhen we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32]\n[SUBTITLE] 2.6.9. Ethics and dissemination. [SUBSECTION] This systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences.\nThis systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences.\n[SUBTITLE] 2.6.10. Grading the quality of evidence. [SUBSECTION] The Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively.\nThe Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively.", "We will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1).\nThe guideline flow diagram.", "Two reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission.", "NOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion.", "For dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05.", "We will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible.", "Subgroup analyses were conducted based on different type of surgical methods.", "The stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis.", "When we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32]", "This systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences.", "The Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively.", "Conceptualization: Qiuxiang Wang.\nData curation: Qiuxiang Wang, Chengjiao Yao, Yilin Li, Ruike Wu.\nFormal analysis: Qiuxiang Wang, Chengjiao Yao.\nFunding acquisition: Lihong Luo, Qin Xiong.\nInvestigation: Qiuxiang Wang, Yilin Li.\nMethodology: Qiuxiang Wang, Chengjiao Yao, Fengjiao Xie.\nSupervision: Peimin Feng.\nWriting – original draft: Qiuxiang Wang.\nWriting – review & editing: Qiuxiang Wang, Chengjiao Yao, Juan Wang." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Methods", "2.1. Study registration", "2.2. Selection criteria", "2.2.1. Type of studies.", "2.2.2. Types of participants.", "2.2.3. Types of interventions and comparisons.", "2.2.4. Language.", "2.3. Exclusion criteria", "2.4. Types of outcome measures", "2.4.1. Primary outcomes.", "2.4.2. Secondary outcomes.", "2.5. Search strategy", "2.6. Data collection and analysis", "2.6.1. Study selection.", "2.6.2. Data extraction.", "2.6.3. Assessment of risk of bias.", "2.6.4. Measures of perioperative outcomes.", "2.6.5. Data analysis and heterogeneity processing.", "2.6.6. Subgroup analysis.", "2.6.7. Sensitivity analysis.", "2.6.8. Assessment of reporting bias.", "2.6.9. Ethics and dissemination.", "2.6.10. Grading the quality of evidence.", "3. Discussion", "Author contributions" ]
[ "The ongoing coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has posed a serious public health threat worldwide, with millions of people at risk in a growing number of countries.[1]Up to May 22, 2022, over 522 million confirmed cases and over 6 million confirmed deaths have been reported to the World Health Organization from different countries, areas and territories. During the pandemic, the management of patients with cancer has been affected at multiple stages, including the triage decisions, surgery, and neoadjuvant therapy as a bridge to reduce admissions and preserve health-care resources.[2] COVID Surg Collaborative estimated that 28,404,603 elective operations were canceled or postponed worldwide during the 12 weeks of peak disruption, with 38% being for cancer.[3]\nEsophageal cancer (EC) is the 8th most common type of cancer worldwide, constitutes the sixth leading cause of cancer deaths.[4] EC remains a global health concern with a dismal prognosis and an estimated 5-year survival rate of approximately 10% to 15%.[5] Surgery plays an important role in the treatment strategies for EC. Recent advances in surgical techniques and perioperative management have dramatically improved the mortality rate; however, esophagectomy remains a highly invasive procedure that can lead to severe postoperative complications.[6] Besides, the rapid global spread of COVID-19 presents an unprecedented crisis for esophagectomy.[7] Recent reports suggest that patients with cancer might have a higher risk of COVID-19 than individuals without cancer and patients with cancer had poorer outcomes from COVID-19.[8] In particular, for patients with EC, additional precautions have been taken, as surgery for EC alone has higher morbidity and mortality rates compared to that for other oncological surgeries.[9] The complexity of the multidisciplinary approach to EC patients and the high morbidity rates of esophageal surgery have challenged the treatment pathways of these patients, especially during the COVID-19 pandemic. Overall, 55% of scheduled endoscopic resections for gastrointestinal neoplastic lesions were deferred globally after the lockdown period, which was 11 times higher than in the previous year, and the majority of postponements (80%) occurred in severely affected countries.[10] For patients with cancer, delay of surgery has the potential to increase the likelihood of metastatic disease, with some patients’ tumors progressing from being curable (with near-normal life expectancy) to noncurable (with limited life expectancy).[11] Delay in time to esophagectomy for EC has been shown to have worse perioperative and long-term outcomes. Therefore, general guidance from health ministries and national surgical associations supported that time-dependent surgery should continue.[12] Despite outbreaks, cancer surgery must continue to prevent an overwhelming number of delayed operations, a possible increase in emergency procedures, and a significant decline in population health.[3]\nHowever, there have been conflicting results regarding the safety of EC surgery during the COVID-19 pandemic. Some studies have demonstrated that EC surgical procedures may be safely performed during the pandemic.[13–17] while another study reported that one of the esophago-gastric junction cancer patients developed COVID-19 pneumonia on post-operative day two, leading to impaired respiratory function and increased pleural fluid collection from the chest tube, resulting in a prolonged hospital stay.[18] And the other study reported the case of a young man who underwent thoracoscopic subtotal esophagectomy for distal esophageal adenocarcinoma who developed COVID-19 with severe clinical presentation.[19] Modeling the impact of delaying surgery for early EC in the era of COVID-19 showed that as the risk of infection with COVID-19 increased above 7%, delaying operations for 3 months has an improved long-term survival.[20] While other study showed that the EC time to surgery over 8 weeks is associated with lower survival.[21] So it is necessary to evaluate the safety of EC surgery during COVID-19. To the best of our knowledge, there have been no meta-analyses on the safety of EC surgery during COVID-19. Therefore, we performed a meta-analysis of cohort studies to evaluate the safety of EC surgery during the COVID-19 pandemic.", "[SUBTITLE] 2.1. Study registration [SUBSECTION] The present study was conducted in accordance with the preferred reporting items for systematic reviews and meta-analysis protocols statement guidelines[22] and has been registered with PROSPERO under registration number CRD42022335164.\nThe present study was conducted in accordance with the preferred reporting items for systematic reviews and meta-analysis protocols statement guidelines[22] and has been registered with PROSPERO under registration number CRD42022335164.\n[SUBTITLE] 2.2. Selection criteria [SUBSECTION] [SUBTITLE] 2.2.1. Type of studies. [SUBSECTION] The present study is a cohort studies of the safety of EC surgery during COVID-19.\nThe present study is a cohort studies of the safety of EC surgery during COVID-19.\n[SUBTITLE] 2.2.2. Types of participants. [SUBSECTION] The sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery.\nThe sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery.\n[SUBTITLE] 2.2.3. Types of interventions and comparisons. [SUBSECTION] However, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19.\nHowever, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19.\n[SUBTITLE] 2.2.4. Language. [SUBSECTION] There is restriction on Chinese or English.\nThere is restriction on Chinese or English.\n[SUBTITLE] 2.2.1. Type of studies. [SUBSECTION] The present study is a cohort studies of the safety of EC surgery during COVID-19.\nThe present study is a cohort studies of the safety of EC surgery during COVID-19.\n[SUBTITLE] 2.2.2. Types of participants. [SUBSECTION] The sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery.\nThe sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery.\n[SUBTITLE] 2.2.3. Types of interventions and comparisons. [SUBSECTION] However, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19.\nHowever, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19.\n[SUBTITLE] 2.2.4. Language. [SUBSECTION] There is restriction on Chinese or English.\nThere is restriction on Chinese or English.\n[SUBTITLE] 2.3. Exclusion criteria [SUBSECTION] Studies that met the following criteria were excluded: duplicate studies; articles published as case reports, case series, review articles, letters, editorials, and commentaries will be excluded; studies involving data that cannot be extracted or inadequate are lacking; there is no control group that assesses the safety of EC surgery during the pre-COVID-19 period; no outcome measures of interest are reported; Newcastle-Ottawa Scale (NOS) scores <5 points; studies written in languages other than Chinese or English.\nStudies that met the following criteria were excluded: duplicate studies; articles published as case reports, case series, review articles, letters, editorials, and commentaries will be excluded; studies involving data that cannot be extracted or inadequate are lacking; there is no control group that assesses the safety of EC surgery during the pre-COVID-19 period; no outcome measures of interest are reported; Newcastle-Ottawa Scale (NOS) scores <5 points; studies written in languages other than Chinese or English.\n[SUBTITLE] 2.4. Types of outcome measures [SUBSECTION] [SUBTITLE] 2.4.1. Primary outcomes. [SUBSECTION] The primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19.\nThe primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19.\n[SUBTITLE] 2.4.2. Secondary outcomes. [SUBSECTION] The secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19.\nThe secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19.\n[SUBTITLE] 2.4.1. Primary outcomes. [SUBSECTION] The primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19.\nThe primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19.\n[SUBTITLE] 2.4.2. Secondary outcomes. [SUBSECTION] The secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19.\nThe secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19.\n[SUBTITLE] 2.5. Search strategy [SUBSECTION] A systematic search plan will be performed in the following eight databases with a time restriction from January 2020 to May 2022 to filter eligible studies: PubMed, Embase, the Cochrane Library, Web of Science, Medline, Chinese National Knowledge Infrastructure database, Chinese Scientific Journal database, and Wan Fang database. We also will search journal articles, conference papers, and academic papers. The search keywords included terms related to COVID-19 pandemic, terms related to EC and terms related to surgery. Cross references were checked to assess if any relevant studies were missed. We considered a specific search strategy in PubMed as a typical example; the specific steps of the retrieval are shown in Table 1.\nSearch strategy in PubMed database.\nA systematic search plan will be performed in the following eight databases with a time restriction from January 2020 to May 2022 to filter eligible studies: PubMed, Embase, the Cochrane Library, Web of Science, Medline, Chinese National Knowledge Infrastructure database, Chinese Scientific Journal database, and Wan Fang database. We also will search journal articles, conference papers, and academic papers. The search keywords included terms related to COVID-19 pandemic, terms related to EC and terms related to surgery. Cross references were checked to assess if any relevant studies were missed. We considered a specific search strategy in PubMed as a typical example; the specific steps of the retrieval are shown in Table 1.\nSearch strategy in PubMed database.\n[SUBTITLE] 2.6. Data collection and analysis [SUBSECTION] [SUBTITLE] 2.6.1. Study selection. [SUBSECTION] We will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1).\nThe guideline flow diagram.\nWe will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1).\nThe guideline flow diagram.\n[SUBTITLE] 2.6.2. Data extraction. [SUBSECTION] Two reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission.\nTwo reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission.\n[SUBTITLE] 2.6.3. Assessment of risk of bias. [SUBSECTION] NOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion.\nNOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion.\n[SUBTITLE] 2.6.4. Measures of perioperative outcomes. [SUBSECTION] For dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05.\nFor dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05.\n[SUBTITLE] 2.6.5. Data analysis and heterogeneity processing. [SUBSECTION] We will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible.\nWe will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible.\n[SUBTITLE] 2.6.6. Subgroup analysis. [SUBSECTION] Subgroup analyses were conducted based on different type of surgical methods.\nSubgroup analyses were conducted based on different type of surgical methods.\n[SUBTITLE] 2.6.7. Sensitivity analysis. [SUBSECTION] The stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis.\nThe stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis.\n[SUBTITLE] 2.6.8. Assessment of reporting bias. [SUBSECTION] When we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32]\nWhen we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32]\n[SUBTITLE] 2.6.9. Ethics and dissemination. [SUBSECTION] This systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences.\nThis systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences.\n[SUBTITLE] 2.6.10. Grading the quality of evidence. [SUBSECTION] The Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively.\nThe Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively.\n[SUBTITLE] 2.6.1. Study selection. [SUBSECTION] We will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1).\nThe guideline flow diagram.\nWe will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1).\nThe guideline flow diagram.\n[SUBTITLE] 2.6.2. Data extraction. [SUBSECTION] Two reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission.\nTwo reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission.\n[SUBTITLE] 2.6.3. Assessment of risk of bias. [SUBSECTION] NOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion.\nNOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion.\n[SUBTITLE] 2.6.4. Measures of perioperative outcomes. [SUBSECTION] For dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05.\nFor dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05.\n[SUBTITLE] 2.6.5. Data analysis and heterogeneity processing. [SUBSECTION] We will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible.\nWe will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible.\n[SUBTITLE] 2.6.6. Subgroup analysis. [SUBSECTION] Subgroup analyses were conducted based on different type of surgical methods.\nSubgroup analyses were conducted based on different type of surgical methods.\n[SUBTITLE] 2.6.7. Sensitivity analysis. [SUBSECTION] The stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis.\nThe stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis.\n[SUBTITLE] 2.6.8. Assessment of reporting bias. [SUBSECTION] When we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32]\nWhen we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32]\n[SUBTITLE] 2.6.9. Ethics and dissemination. [SUBSECTION] This systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences.\nThis systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences.\n[SUBTITLE] 2.6.10. Grading the quality of evidence. [SUBSECTION] The Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively.\nThe Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively.", "The present study was conducted in accordance with the preferred reporting items for systematic reviews and meta-analysis protocols statement guidelines[22] and has been registered with PROSPERO under registration number CRD42022335164.", "[SUBTITLE] 2.2.1. Type of studies. [SUBSECTION] The present study is a cohort studies of the safety of EC surgery during COVID-19.\nThe present study is a cohort studies of the safety of EC surgery during COVID-19.\n[SUBTITLE] 2.2.2. Types of participants. [SUBSECTION] The sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery.\nThe sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery.\n[SUBTITLE] 2.2.3. Types of interventions and comparisons. [SUBSECTION] However, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19.\nHowever, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19.\n[SUBTITLE] 2.2.4. Language. [SUBSECTION] There is restriction on Chinese or English.\nThere is restriction on Chinese or English.", "The present study is a cohort studies of the safety of EC surgery during COVID-19.", "The sample population included patients diagnosed with EC. The subjects enrolled were EC patients undergoing surgery before COVID-19 and during COVID-19, and there were no restrictions on the type of surgery.", "However, the intervention is not applicable. This meta-analysis will evaluate the safety of colorectal cancer surgery during COVID-19. Postoperative complications, anastomotic leak, mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative intensive care unit (ICU) stay, the number of patients needing ICU stay, and 30-day readmission were compared to evaluate the safety of EC surgery before and during COVID-19.", "There is restriction on Chinese or English.", "Studies that met the following criteria were excluded: duplicate studies; articles published as case reports, case series, review articles, letters, editorials, and commentaries will be excluded; studies involving data that cannot be extracted or inadequate are lacking; there is no control group that assesses the safety of EC surgery during the pre-COVID-19 period; no outcome measures of interest are reported; Newcastle-Ottawa Scale (NOS) scores <5 points; studies written in languages other than Chinese or English.", "[SUBTITLE] 2.4.1. Primary outcomes. [SUBSECTION] The primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19.\nThe primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19.\n[SUBTITLE] 2.4.2. Secondary outcomes. [SUBSECTION] The secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19.\nThe secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19.", "The primary outcomes of this study were to evaluate the postoperative complications, mortality, and anastomotic leak after EC surgery before COVID-19 and during COVID-19.", "The secondary outcomes of this study were to evaluate the total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU stay, and 30-day readmission of EC surgery before COVID-19 and during COVID-19.", "A systematic search plan will be performed in the following eight databases with a time restriction from January 2020 to May 2022 to filter eligible studies: PubMed, Embase, the Cochrane Library, Web of Science, Medline, Chinese National Knowledge Infrastructure database, Chinese Scientific Journal database, and Wan Fang database. We also will search journal articles, conference papers, and academic papers. The search keywords included terms related to COVID-19 pandemic, terms related to EC and terms related to surgery. Cross references were checked to assess if any relevant studies were missed. We considered a specific search strategy in PubMed as a typical example; the specific steps of the retrieval are shown in Table 1.\nSearch strategy in PubMed database.", "[SUBTITLE] 2.6.1. Study selection. [SUBSECTION] We will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1).\nThe guideline flow diagram.\nWe will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1).\nThe guideline flow diagram.\n[SUBTITLE] 2.6.2. Data extraction. [SUBSECTION] Two reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission.\nTwo reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission.\n[SUBTITLE] 2.6.3. Assessment of risk of bias. [SUBSECTION] NOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion.\nNOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion.\n[SUBTITLE] 2.6.4. Measures of perioperative outcomes. [SUBSECTION] For dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05.\nFor dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05.\n[SUBTITLE] 2.6.5. Data analysis and heterogeneity processing. [SUBSECTION] We will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible.\nWe will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible.\n[SUBTITLE] 2.6.6. Subgroup analysis. [SUBSECTION] Subgroup analyses were conducted based on different type of surgical methods.\nSubgroup analyses were conducted based on different type of surgical methods.\n[SUBTITLE] 2.6.7. Sensitivity analysis. [SUBSECTION] The stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis.\nThe stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis.\n[SUBTITLE] 2.6.8. Assessment of reporting bias. [SUBSECTION] When we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32]\nWhen we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32]\n[SUBTITLE] 2.6.9. Ethics and dissemination. [SUBSECTION] This systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences.\nThis systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences.\n[SUBTITLE] 2.6.10. Grading the quality of evidence. [SUBSECTION] The Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively.\nThe Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively.", "We will rely on two independent authors (QW and RW) to screen and select the eligible studies. The literature obtained will be imported into NoteExpress to screen the title and abstract. Subsequently, we will obtain full-text articles from relevant studies. After reading the full text of the remaining studies, the final number of included studies will be determined. Disagreements will be resolved by a third reviewer (PF). The entire study selection process is presented in the guideline flow diagram (Fig. 1).\nThe guideline flow diagram.", "Two reviewers (QW and JW) will independently extract data from the included studies and enter the extracted data into Excel sheets. Disagreements will be resolved through negotiation and discussion. Further controversy will be arbitrated by a third reviewer (PF). The following information was extracted from each included study: study baseline (the first author’s name, published year, country, study design, study size, age, sex, operative methods); primary surgical outcomes: postoperative complications, anastomotic leak, mortality; secondary surgical outcomes: total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, postoperative ICU stay, the number of patients needing ICU and 30-day readmission.", "NOS was used for the quality assessment of the included studies.[23] NOS provides a checklist of items for evaluating the quality of reporting and the risk of bias of the included studies based on three broad evaluation categories: selection, comparability, and exposure/outcomes.[24] The scale has three parameters and eight items with a total score of 9, scores ≤ 3 are usually considered low quality, scores of 4 or 5 are considered medium quality, and scores ≥ 6 are usually considered high quality.[25] Two reviewers (FX and RW) independently performed the quality assessment of the included studies, and the third team member (PF) performed the verification. Discrepancies were resolved through discussion.", "For dichotomous data, such as postoperative complications, anastomotic leak, mortality, number of patients requiring ICU admission, and 30-day readmission, we plan to present the results as risk ratios (RR) with 95% confidence intervals (CIs). For continuous data, such as total hospital stay, postoperative stay, preoperative waiting, operation time, blood loss, transfusion, and postoperative ICU stay, we will use the MD with a 95% CIs. Statistical significance was set at P < .05.", "We will use the Review Manager (RevMan) software (Version 5.4, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboation,s 2020) for the meta-analysis and statistical analysis. Egger’s test was performed using the Stata software (version 16.0, Stata Corp LP, College Station). Heterogeneity among the studies was evaluated using chi-square tests and inconsistency statistic.[26] If the included article reported outcomes in medians and interquartile ranges, the method described by Wan et al was used to calculate the mean and standard deviation (SD).[27,28] If the included articles reported outcomes as medians, maximum and minimum, the method described by Hozo et al was used to calculate the mean and SD.[29]It indicated that there was significant heterogeneity if I2 > 50% and/or P < .1, the fixed-effects model (Mantel–Haensze) was used to analyze the data if no heterogeneity was present, whereas the random-effects model was used if I2 > 50%.[30] The levels of heterogeneity assessed using I2 were as follows: 0% to 25%, homogeneity; 25% to 50%, low heterogeneity; 50% to 75%,moderate heterogeneity; and > 75% meant high heterogeneity.[31] Possible reasons for heterogeneity will be determined using sensitivity analysis or subgroup analysis. A descriptive analysis of the results will be performed when considerable heterogeneity makes the analysis infeasible.", "Subgroup analyses were conducted based on different type of surgical methods.", "The stability of the meta-analysis results was tested by changing effect model method in sensitivity analysis.", "When we select > 10 studies consistent with these conditions, a funnel plot was used to detect publication bias, and the Egger test of bias was used as a supplement.[32]", "This systematic review and meta-analysis was based on published data. As the researchers did not access any information that could lead to the identification of an individual patient, no ethical issues were raised in this study. Therefore, the requirement for ethical approval and consent from participants was waived. The research results may be published in peer-reviewed journals or disseminated at relevant conferences.", "The Grading of Recommendations Assessment, Development and Evaluation guidelines will be used to grade the quality of evidence as very low, low, moderate, or high, respectively.", "As COVID-19 prevention and control have become normal, the situation of COVID-19 is still not optimistic. During the COVID-19 pandemic, patients with EC are at a higher risk of developing COVID-19 due to frequent hospital visits and an increased risk of developing severe disease after the infecting COVID-19. Surgery is the main treatment modality for solid cancers.[33] Therefore, whether EC surgery can be performed safely during the COVID-19 pandemic has become an important topic of clinical concern.\nEsophagectomy is a complex surgical procedure and is associated with substantial morbidity, particularly postoperative pneumonia and consecutive respiratory failure.[34–37] An added risk of COVID-19 may be on increasing the risk of anastomotic leaks, as has been reported for colorectal cancer resections, as anastomotic leaks are reported in between 5% and 25% of EC cases and may have fatal consequences.[34,35,38] Moreover, patients with EC have poor nutrition and low resistance, which are associated with a high risk of anastomotic leaks. The shortage of ICU facilities and the potential increased risk of mortality related to perioperative COVID-19 infection in cancer patients have raised several concerns about the most appropriate surgical management of patients with EC.[15] So it is necessary to evaluate the safety of EC surgery during the COVID-19 pandemic.\nHowever, no systematic review or meta-analysis has been conducted on the safety of EC surgery during COVID-19. This study systematically evaluated the safety of EC surgery during COVID-19, in order to provide evidence-based evidence for the safety EC surgery during COVID-19. In this systematic review and meta-analysis, we will investigate clinical studies on the safety of EC surgery during COVID-19 by assessing the perioperative results, including: postoperative complications, anastomotic leak and mortality, total hospital stay, postoperative stay, preoperative waiting time, operation time, blood loss, transfusion, postoperative ICU stay, number of patients needing ICU, and 30-day readmission. We expect that our findings will be a useful resource for clinical practitioners and patients to suggest optimized clinical surgical strategies for EC during COVID-19.", "Conceptualization: Qiuxiang Wang.\nData curation: Qiuxiang Wang, Chengjiao Yao, Yilin Li, Ruike Wu.\nFormal analysis: Qiuxiang Wang, Chengjiao Yao.\nFunding acquisition: Lihong Luo, Qin Xiong.\nInvestigation: Qiuxiang Wang, Yilin Li.\nMethodology: Qiuxiang Wang, Chengjiao Yao, Fengjiao Xie.\nSupervision: Peimin Feng.\nWriting – original draft: Qiuxiang Wang.\nWriting – review & editing: Qiuxiang Wang, Chengjiao Yao, Juan Wang." ]
[ "intro", "methods", null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, "discussion", null ]
[ "COVID-19 pandemic", "esophageal cancer", "meta-analysis", "protocol", "surgery", "systematic review" ]
The effectiveness of massage interventions on procedural pain in neonates: A systematic review and meta-analysis.
36254036
The painful procedures experienced by neonates during hospitalization have short-term or long-term effects on neonates. While the limitations of previous interventions make it imperative to explore effective interventions that are readily available. This systematic review and meta-analysis was conducted to evaluate the safety and effectiveness of massage for pain management in neonates.
BACKGROUND
This systematic review was registered in PROSPER. PubMed, Embase, Cochrane Library, and the Clinical Trials Registry were searched to December 2021. Two reviewers independently carried out study selection, data extraction, bias risk assessment. Continuous data were analyzed by mean differences (MD). Dichotomous data were reported using relative risk. If at least two studies reported identical results by the same pain assessment tool, a meta-analysis was conducted using random effect model and inverse variance.
METHODS
Total 11 included studies involving 755 neonates investigated the effects of massage on neonatal pain response compared to standard care. The meta-analysis showed that massage could effectively improve pain response in neonates compared to standard care no matter whether neonatal infant pain scale (NIPS) or premature infant pain profile (PIPP) was used as an assessment tool. Besides, massage was also effective for crying duration, blood oxygen saturation both during and after the procedure, but non-effective for the variation of respiratory rate after the procedure, and heart rate both during and after the procedure.
RESULTS
Massage may have a positive effect on pain relief of neonate, and rigorous trials are needed in the future to determine the most effective massage method.
CONCLUSIONS
[ "Humans", "Infant", "Infant, Newborn", "Massage", "Pain", "Pain Management", "Pain, Procedural", "Punctures" ]
9575769
1. Introduction
Neonates undergo many painful examinations in hospital. Each infant undergoes 7.5 to 17.3 painful examinations every day, such as heel-pricks, venipunctures, etc.[1] Which undoubtedly brings great pain to newborns. It is reported that repeated pain examinations will cause short-term or long-term adverse consequences for neonates,[2–5] such as reducing cerebral blood flow, hinder the development of neurological or motor functions and even cause hypersensitivity to pain.[6–8] This highlights the significance of pain management for infants who experience painful procedures during hospitalization. Although there are many pharmaceutical interventions that could be able to alleviate the pain of infants, the safety of long-term use remains to be studied.[9] As a safe and reliable pain management method, non-pharmacological intervention is increasingly favored by parents of neonates.[10,11] There are a growing body of researches on non-pharmacological interventions, such as, non-nutritive sucking, skin-to-skin contact, and breastfeeding before painful procedures. These interventions have been proved to be effective in alleviating pain to newborn babies.[12–14] But these interventions require the presence of a mother or one of the parents, which is not always appropriate in a variety of clinical settings. So, we need to explore innovative interventions that can be used anytime in any setting and are effective for pain relief in newborns. Effective interventions would ideally be inexpensive, noninvasive, and be rapidly applied to improve pediatric pain control.[13] Massage, as an effective non-pharmacological intervention, has been gradually applied in various areas of clinical practice. It is a method applied by stimulating the acupuncture points and meridians in body using hands or special tools.[15,16] Studies have shown that massage can relieve stress and improve blood circulation by reducing the levels of cortisol and increasing the levels of serotonin and dopamine.[17,18] By stimulating the release of endorphins and serotonin, massage can relieve the pain of neonates, improve their sleep, and have a positive impact on the growth and development of neonates.[19] Besides, massage can also activate the parasympathetic nervous system as a result of stimulation of the vagal nerves and provide calm and rest in the body.[20] To our knowledge, only one systematic review on the non-pharmacological interventions[11] that reported the effectiveness of massage in procedural pain relief of infants previously. However, in this review, the massage was found to be effective in alleviating the pain response of premature infants, but not for neonates. Other recent reviews reported that the massage was safe and effective for neonates’ pain relief.[21] Moreover, a lot of studies have been conducted on the effects of massage for neonatal pain management in recent years, but the results were either controversial or partially effective.[22–24] Therefore, a more comprehensive systematic review and meta-analysis is needed to assess the safety and effectiveness of this intervention in the pain management of preterm and full-term infants. The purpose of this study was to comprehensively evaluate the effectiveness of massage on pain relief and other secondary outcomes (the variation of heart rate, respiration, blood oxygen saturation, crying time, cortisol levels, and adverse events) in premature and full-term infants.
2. Methods
[SUBTITLE] 2.1. Protocol and registration [SUBSECTION] This systematic review protocol was registered in PROSPERO database (CRD42022302115). We conducted this systematic review according to the recommendations of preferred reporting items for systematic reviews and meta-analyses guidelines.[25] This systematic review protocol was registered in PROSPERO database (CRD42022302115). We conducted this systematic review according to the recommendations of preferred reporting items for systematic reviews and meta-analyses guidelines.[25] [SUBTITLE] 2.2. Search strategy and selection criteria [SUBSECTION] We conducted electronic searches under the guidance of a library search specialist in the following databases to December 2021: PubMed, Embase, the Cochrane Library and the Clinical Trials Registry. Relevant articles were retrieved by combining the following medical subject headlines (MeSH) and keywords: (Infants OR Premature OR Preterm OR Neonatal OR Prematurity OR Newborn OR Neonate) AND (massage OR touch OR pain management). Eligible studies included had to fulfill the following criteria: randomized controlled/clinical trials that conducted on the neonates with gestation between 24 and 42 weeks; studies comparing massage interventions (massage, or therapeutic touch) to comparator groups (offering standard care); with outcome including one of the ten most common painful procedures experienced by infants such as heel-prick, venipuncture et al[26] Articles were excluded, if: reviews and case reports; studies not in English; studies without valid data or with improper data. All standardized measurement scales and tools related to neonates’ painful evaluation would be considered, including: premature infant pain profile (PIPP)[27,28] suitable for newborns between 28 and 40 weeks of gestation (WG); Neonatal Infant Pain Scale (NIPS) from 26 to 47 WG newborns[29]; neonatal facial coding system for 26 to 47 WG newborns.[30] We combined different tools in our systematic review for pain assessment. The main outcome was the neonates’ painful response, according to the suggestion of Pillai Riddell.[11] Secondary outcomes included the variations of heart rate, blood oxygen saturation, respiration rate during and after painful examination and variations of the crying time, cortisol levels, occurrence of adverse events between before and after the painful procedure. Besides, the measurements taken within five minutes after the painful procedure were considered. As for measurements taken before discharge, we included the closest one to discharge. We conducted electronic searches under the guidance of a library search specialist in the following databases to December 2021: PubMed, Embase, the Cochrane Library and the Clinical Trials Registry. Relevant articles were retrieved by combining the following medical subject headlines (MeSH) and keywords: (Infants OR Premature OR Preterm OR Neonatal OR Prematurity OR Newborn OR Neonate) AND (massage OR touch OR pain management). Eligible studies included had to fulfill the following criteria: randomized controlled/clinical trials that conducted on the neonates with gestation between 24 and 42 weeks; studies comparing massage interventions (massage, or therapeutic touch) to comparator groups (offering standard care); with outcome including one of the ten most common painful procedures experienced by infants such as heel-prick, venipuncture et al[26] Articles were excluded, if: reviews and case reports; studies not in English; studies without valid data or with improper data. All standardized measurement scales and tools related to neonates’ painful evaluation would be considered, including: premature infant pain profile (PIPP)[27,28] suitable for newborns between 28 and 40 weeks of gestation (WG); Neonatal Infant Pain Scale (NIPS) from 26 to 47 WG newborns[29]; neonatal facial coding system for 26 to 47 WG newborns.[30] We combined different tools in our systematic review for pain assessment. The main outcome was the neonates’ painful response, according to the suggestion of Pillai Riddell.[11] Secondary outcomes included the variations of heart rate, blood oxygen saturation, respiration rate during and after painful examination and variations of the crying time, cortisol levels, occurrence of adverse events between before and after the painful procedure. Besides, the measurements taken within five minutes after the painful procedure were considered. As for measurements taken before discharge, we included the closest one to discharge. [SUBTITLE] 2.3. Study selection and data extraction [SUBSECTION] We managed all the references in EndNote X9. After removing the duplication, two authors (LNG&TTX) independently reviewed the included studies according to the title and keywords, and then all eligible studies were retained for full-text assessment to determine whether suitable for inclusion in this systematic review. The reasons for the excluded references were recorded. To ensure the consistency of data, two reviewers extracted data independently and stored the data in Review Manager software. Before data analysis, two reviewers checked the data to avoid errors. The above process required two reviewers to reach a consensus, any dissenting opinions about the inclusion were resolved by consulting other reviewers (SRF&YXW). We managed all the references in EndNote X9. After removing the duplication, two authors (LNG&TTX) independently reviewed the included studies according to the title and keywords, and then all eligible studies were retained for full-text assessment to determine whether suitable for inclusion in this systematic review. The reasons for the excluded references were recorded. To ensure the consistency of data, two reviewers extracted data independently and stored the data in Review Manager software. Before data analysis, two reviewers checked the data to avoid errors. The above process required two reviewers to reach a consensus, any dissenting opinions about the inclusion were resolved by consulting other reviewers (SRF&YXW). [SUBTITLE] 2.4. Quality assessment [SUBSECTION] The Risk of Bias in the included studies was independently assessed by two reviewers (LNG&TTX) according to version 2 of the Cochrane risk-of-bias tool for randomized trials (RoB2) recommended by Cochrane Handbook for Systematic Reviews of Interventions, including bias arising from the randomization process, bias due to deviations, bias due to incomplete data, bias in measurement of the outcome, bias in selection of the reported result.[31] The risk of bias for each study was classified as low risk of bias, high risk of bias, and some concerns. The Risk of Bias in the included studies was independently assessed by two reviewers (LNG&TTX) according to version 2 of the Cochrane risk-of-bias tool for randomized trials (RoB2) recommended by Cochrane Handbook for Systematic Reviews of Interventions, including bias arising from the randomization process, bias due to deviations, bias due to incomplete data, bias in measurement of the outcome, bias in selection of the reported result.[31] The risk of bias for each study was classified as low risk of bias, high risk of bias, and some concerns. [SUBTITLE] 2.5. Statistical analysis [SUBSECTION] Statistical analysis was conducted in the Review Manager software by using a random-effect model with a 95% confidence interval (CI). Since neonatal pain was assessed using different tools, we treated each tool separately to ensure data consistency. Continuous data were analyzed using mean differences (MD). Dichotomous data were reported using relative risk. If at least two studies reported identical results using the same pain assessment tool, a meta-analysis was conducted by using the random effect model and inverse variance. Subgroup analysis would be conducted according to the different massage method to provide further guidance for clinical practice. Using the chi-square test with a significance level of 0.1 to assess the heterogeneity of included studies. According to the suggestions of preferred reporting items for systematic reviews and meta-analyses-P,[32] we classified I² as not important heterogeneity (0%–40%); moderate heterogeneity (30%–60%); substantial heterogeneity (50%–90%) and considerable heterogeneity (75%–100%). Statistical analysis was conducted in the Review Manager software by using a random-effect model with a 95% confidence interval (CI). Since neonatal pain was assessed using different tools, we treated each tool separately to ensure data consistency. Continuous data were analyzed using mean differences (MD). Dichotomous data were reported using relative risk. If at least two studies reported identical results using the same pain assessment tool, a meta-analysis was conducted by using the random effect model and inverse variance. Subgroup analysis would be conducted according to the different massage method to provide further guidance for clinical practice. Using the chi-square test with a significance level of 0.1 to assess the heterogeneity of included studies. According to the suggestions of preferred reporting items for systematic reviews and meta-analyses-P,[32] we classified I² as not important heterogeneity (0%–40%); moderate heterogeneity (30%–60%); substantial heterogeneity (50%–90%) and considerable heterogeneity (75%–100%).
3. Results
[SUBTITLE] 3.1. Study selection [SUBSECTION] The study selection process was showed by Flow Diagram (Fig. 1). A total of 5635 articles were identified through database searching, and 7 related studies were also included through snowballing, gray literature review and other methods. After the removal of 521 duplicated articles, a total of 5121 articles were retained. We eliminated 5089 results in the first screening by reading questions and keywords, and 32 studies were evaluated to be eligible for final selection (full-texts). In total, 21 articles were excluded for different reasons: 2 articles were not randomized controlled trials; 4 articles were not eligible population; 4 articles were not eligible interventions; 1 article was excluded for measurement; 6 articles were excluded because the full text could not be obtained even after contacting the authors; 1 article was not considered for language reasons; 3 articles’ dates could not be used. Finally, we conducted a meta-analysis of 11 studies, including 755 neonates investigating the effects of massage on neonatal pain response compared to standard care.[17,33–42] Preferred reporting items for systematic reviews and meta-analyses (PRISMA) Flow diagram of this meta-analysis. The study selection process was showed by Flow Diagram (Fig. 1). A total of 5635 articles were identified through database searching, and 7 related studies were also included through snowballing, gray literature review and other methods. After the removal of 521 duplicated articles, a total of 5121 articles were retained. We eliminated 5089 results in the first screening by reading questions and keywords, and 32 studies were evaluated to be eligible for final selection (full-texts). In total, 21 articles were excluded for different reasons: 2 articles were not randomized controlled trials; 4 articles were not eligible population; 4 articles were not eligible interventions; 1 article was excluded for measurement; 6 articles were excluded because the full text could not be obtained even after contacting the authors; 1 article was not considered for language reasons; 3 articles’ dates could not be used. Finally, we conducted a meta-analysis of 11 studies, including 755 neonates investigating the effects of massage on neonatal pain response compared to standard care.[17,33–42] Preferred reporting items for systematic reviews and meta-analyses (PRISMA) Flow diagram of this meta-analysis. [SUBTITLE] 3.2. Characteristics of included studies [SUBSECTION] The 11 studies were published between 2006 and 2021 written in English, and the characteristics were summarized in Table 1. They were carried out in 5 different countries, 2 in China,[33,40] 4 in Iran,[37,39,41,42] 1 in Korea,[34] 2 in Canada,[17,35] and 2 in Turkey.[36,38] Five studies[34,36–38,41] included neonates gestational age older than 37 weeks. Five studies[17,35,39,40,42] included premature younger than 37 weeks of gestational age, and one study[33] included newborns between 30 and 40 weeks of gestational age. All of the studies were randomized controlled trials, and the interventions studied were touch or massage. Seven of the studies were on massage[17,33,36–39,41] and four were on therapeutic touching,[34,35,40,42] but they were all included because the interventions were the same or similar. Moreover, three of the studies were cross-designed[17,33,40] and the rest were parallel designed. Characteristics of included studies. NIPS = neonatal infant pain scale, PIPP = premature infant pain profile, RCT = randomized controlled trial, RR = risk ratios. Included studies evaluated different painful procedures during hospitalization including venipuncture (n = 1),[33] hypothyroidism screening (n = 1),[37] heel-prick (n = 7),[17,34–36,38,39,41] screening for retinopathy (n = 1),[40] and endotracheal suctioning (n = 1).[42] Different assessment tools were used in the included studies, but all were reliable and valid standard assessment scales. Six used the Neonatal Infant Pain Scale[17,34,36–39] and five used Premature Infant Pain Profile.[33,35,40–42] In addition to pain assessments, several second outcomes were also included in the study, including heart rate, respiration, oxygen saturation and crying time. Moreover, there were some differences in the assessment time between studies. Three studies[24,43,44] were removed from the meta-analysis since the data was not able to be obtained, we reported their results through systematic synthesis. The 11 studies were published between 2006 and 2021 written in English, and the characteristics were summarized in Table 1. They were carried out in 5 different countries, 2 in China,[33,40] 4 in Iran,[37,39,41,42] 1 in Korea,[34] 2 in Canada,[17,35] and 2 in Turkey.[36,38] Five studies[34,36–38,41] included neonates gestational age older than 37 weeks. Five studies[17,35,39,40,42] included premature younger than 37 weeks of gestational age, and one study[33] included newborns between 30 and 40 weeks of gestational age. All of the studies were randomized controlled trials, and the interventions studied were touch or massage. Seven of the studies were on massage[17,33,36–39,41] and four were on therapeutic touching,[34,35,40,42] but they were all included because the interventions were the same or similar. Moreover, three of the studies were cross-designed[17,33,40] and the rest were parallel designed. Characteristics of included studies. NIPS = neonatal infant pain scale, PIPP = premature infant pain profile, RCT = randomized controlled trial, RR = risk ratios. Included studies evaluated different painful procedures during hospitalization including venipuncture (n = 1),[33] hypothyroidism screening (n = 1),[37] heel-prick (n = 7),[17,34–36,38,39,41] screening for retinopathy (n = 1),[40] and endotracheal suctioning (n = 1).[42] Different assessment tools were used in the included studies, but all were reliable and valid standard assessment scales. Six used the Neonatal Infant Pain Scale[17,34,36–39] and five used Premature Infant Pain Profile.[33,35,40–42] In addition to pain assessments, several second outcomes were also included in the study, including heart rate, respiration, oxygen saturation and crying time. Moreover, there were some differences in the assessment time between studies. Three studies[24,43,44] were removed from the meta-analysis since the data was not able to be obtained, we reported their results through systematic synthesis. [SUBTITLE] 3.3. Evaluation of evidence quality [SUBSECTION] The risk on bias of each included study was presented in Fig. 2 and the detail was also summarized (see Supplemental Table S1, http://links.lww.com/MD/H461, Supplemental Content, which summarized the risk of bias in studies). Seven studies reported the methods of random-sequence generation detailedly,[17,33,36–39,42] while others were unclear since insufficient information. Six of the studies described the detail of allocation concealment method appropriately,[17,34–36,38,39] but the rest were assessed as unclear bias risk. All but one study were unclear risks since the nature of the intervention and the difficulty of blinding participants.[33–42] For the blinding of outcome assessment, there were five studies describing it,[17,33,35,38,40] while the rest were uncertain. The risk of incomplete outcome data was high in only one study,[42] because the missing date of different groups was unbalanced and the reasons were different. For selective reporting, four studies[36,37,39,42] were judged low but one[40] was high risk since not all of the prespecified outcomes were reported. Of all the studies, we considered only four[33,38,41,42] to be free from the bias of other sources, and others unclear for insufficient evidence provided. Risk of bias summary. The risk on bias of each included study was presented in Fig. 2 and the detail was also summarized (see Supplemental Table S1, http://links.lww.com/MD/H461, Supplemental Content, which summarized the risk of bias in studies). Seven studies reported the methods of random-sequence generation detailedly,[17,33,36–39,42] while others were unclear since insufficient information. Six of the studies described the detail of allocation concealment method appropriately,[17,34–36,38,39] but the rest were assessed as unclear bias risk. All but one study were unclear risks since the nature of the intervention and the difficulty of blinding participants.[33–42] For the blinding of outcome assessment, there were five studies describing it,[17,33,35,38,40] while the rest were uncertain. The risk of incomplete outcome data was high in only one study,[42] because the missing date of different groups was unbalanced and the reasons were different. For selective reporting, four studies[36,37,39,42] were judged low but one[40] was high risk since not all of the prespecified outcomes were reported. Of all the studies, we considered only four[33,38,41,42] to be free from the bias of other sources, and others unclear for insufficient evidence provided. Risk of bias summary. [SUBTITLE] 3.4. Pain response score [SUBSECTION] Six studies[17,34,36–39] including 460 neonates investigated neonatal pain response using NIPS and found that the massage was effective in improving neonatal pain response compared to standard care (MD −2.02; 95% CI −2.63 to −1.42; I2 = 74%; P < .01) (Fig. 3A). Forest plot displaying the results of pain response score. According to subgroup analysis of age difference of neonates, massage could effectively improve their pain response regardless of whether the intervention group was full-term infants or premature infants. The differences between subgroups were not statistically significant (P = .89, I2 = 0%) (Fig. 4). Forest plot displaying the results of subgroup analysis by gestational age of neonates. Four studies[33,35,40,42] including 235 neonates investigated the pain response of neonates using the PIPP scale and showed that the pain response scores of neonates who received massage prior to the pain procedure were significantly lower than those who received routine care (MD −3.43; 95% CI −6.05 to −0.80; I2 = 93%; P = .01) (Fig. 3B). According to different intervention measures, subgroup analysis found that the differences between subgroups were statistically significant (P = .05 I2 = 75%) (Fig. 5), and the massage was effective for neonates, while therapeutic touch was not effective for neonates. Forest plot displaying the results of subgroup analysis by different interventions. Six studies[17,34,36–39] including 460 neonates investigated neonatal pain response using NIPS and found that the massage was effective in improving neonatal pain response compared to standard care (MD −2.02; 95% CI −2.63 to −1.42; I2 = 74%; P < .01) (Fig. 3A). Forest plot displaying the results of pain response score. According to subgroup analysis of age difference of neonates, massage could effectively improve their pain response regardless of whether the intervention group was full-term infants or premature infants. The differences between subgroups were not statistically significant (P = .89, I2 = 0%) (Fig. 4). Forest plot displaying the results of subgroup analysis by gestational age of neonates. Four studies[33,35,40,42] including 235 neonates investigated the pain response of neonates using the PIPP scale and showed that the pain response scores of neonates who received massage prior to the pain procedure were significantly lower than those who received routine care (MD −3.43; 95% CI −6.05 to −0.80; I2 = 93%; P = .01) (Fig. 3B). According to different intervention measures, subgroup analysis found that the differences between subgroups were statistically significant (P = .05 I2 = 75%) (Fig. 5), and the massage was effective for neonates, while therapeutic touch was not effective for neonates. Forest plot displaying the results of subgroup analysis by different interventions. [SUBTITLE] 3.5. Variation of heart rate [SUBSECTION] Three studies (n = 249) investigated the variation of heart rate during the painful procedure,[36,37,42] and there was no significant difference between the massage group and the standard care group (MD 3.39; 95% CI −1.14–7.92; I2 = 45%; P = .14) (Fig. 6A). Forest plot displaying the results of heart rate. Five studies with 332 neonates[17,34,36,37,41] analyzed the variations of heart rate after the painful examination, and the result showed that massage did not lower the heart rate of neonates after procedure compared to routine care (MD −2.15; 95% CI −8.82 to 4.52; I2 = 82%; P = .53) (Fig. 6B). Three studies (n = 249) investigated the variation of heart rate during the painful procedure,[36,37,42] and there was no significant difference between the massage group and the standard care group (MD 3.39; 95% CI −1.14–7.92; I2 = 45%; P = .14) (Fig. 6A). Forest plot displaying the results of heart rate. Five studies with 332 neonates[17,34,36,37,41] analyzed the variations of heart rate after the painful examination, and the result showed that massage did not lower the heart rate of neonates after procedure compared to routine care (MD −2.15; 95% CI −8.82 to 4.52; I2 = 82%; P = .53) (Fig. 6B). [SUBTITLE] 3.6. Variation of blood oxygen saturation [SUBSECTION] Three studies involving 249 neonates[36,37,42] explored the effects of massage on the change of blood oxygen saturation during painful examination, and the merged result showed that massage was more effective than standard care in improving neonate blood oxygen saturation during the examination (MD 2.25; 95% CI 0.28–4.76; I2 = 71%; P = .03) (Fig. 7A). Forest plot displaying the results of blood oxygen saturation. Five studies including 332 participants[17,34,36,37,41] revealed the massage group had a significant advantage over the control group in improving the blood oxygen saturation after the examination (MD 1.05; 95% CI 0.51–1.58; I2 = 0%; P < .01) (Fig. 7B). Three studies involving 249 neonates[36,37,42] explored the effects of massage on the change of blood oxygen saturation during painful examination, and the merged result showed that massage was more effective than standard care in improving neonate blood oxygen saturation during the examination (MD 2.25; 95% CI 0.28–4.76; I2 = 71%; P = .03) (Fig. 7A). Forest plot displaying the results of blood oxygen saturation. Five studies including 332 participants[17,34,36,37,41] revealed the massage group had a significant advantage over the control group in improving the blood oxygen saturation after the examination (MD 1.05; 95% CI 0.51–1.58; I2 = 0%; P < .01) (Fig. 7B). [SUBTITLE] 3.7. Respiratory rate variation and duration of crying [SUBSECTION] Only two studies[17,37] involving 113 neonates measured the effect of massage on respiration rate after neonatal procedure, and the results showed no effect on respiration rate (MD 0.11; 95% CI −2.61–2.84; I2 = 0%; P = .94) (Fig. 8A). Forest plot displaying the results of respiratory rate and duration of crying. Three studies including 238 neonates[35–37] revealed the effect of massage on crying time during painful procedures, and the merged result showed that neonates in the experimental group spent less time crying than those in the standard care group (MD −48.6; 95% CI −64.32 to −32.88; I ² = 8%; P < .01) (Fig. 8B). Only two studies[17,37] involving 113 neonates measured the effect of massage on respiration rate after neonatal procedure, and the results showed no effect on respiration rate (MD 0.11; 95% CI −2.61–2.84; I2 = 0%; P = .94) (Fig. 8A). Forest plot displaying the results of respiratory rate and duration of crying. Three studies including 238 neonates[35–37] revealed the effect of massage on crying time during painful procedures, and the merged result showed that neonates in the experimental group spent less time crying than those in the standard care group (MD −48.6; 95% CI −64.32 to −32.88; I ² = 8%; P < .01) (Fig. 8B).
5. Conclusions
In general, massage intervention plays a positive role in the relief of painful procedures in neonates, and we recommend it be used in clinical practice. Adverse events of massage intervention should be reported in future studies to guide clinical study and ensure the massage is carried out safely. In hospitals, especially in the NICU, neonates undergo a variety of painful procedures. So, it is imperative to identify the most effective interventions to manage procedural pain during hospitalization of neonate.
[ "2.1. Protocol and registration", "2.2. Search strategy and selection criteria", "2.3. Study selection and data extraction", "2.4. Quality assessment", "2.5. Statistical analysis", "3.1. Study selection", "3.2. Characteristics of included studies", "3.3. Evaluation of evidence quality", "3.4. Pain response score", "3.5. Variation of heart rate", "3.6. Variation of blood oxygen saturation", "3.7. Respiratory rate variation and duration of crying", "4.1. Limitations", "Acknowledgments", "Author contributions" ]
[ "This systematic review protocol was registered in PROSPERO database (CRD42022302115). We conducted this systematic review according to the recommendations of preferred reporting items for systematic reviews and meta-analyses guidelines.[25]", "We conducted electronic searches under the guidance of a library search specialist in the following databases to December 2021: PubMed, Embase, the Cochrane Library and the Clinical Trials Registry. Relevant articles were retrieved by combining the following medical subject headlines (MeSH) and keywords: (Infants OR Premature OR Preterm OR Neonatal OR Prematurity OR Newborn OR Neonate) AND (massage OR touch OR pain management).\nEligible studies included had to fulfill the following criteria: randomized controlled/clinical trials that conducted on the neonates with gestation between 24 and 42 weeks; studies comparing massage interventions (massage, or therapeutic touch) to comparator groups (offering standard care); with outcome including one of the ten most common painful procedures experienced by infants such as heel-prick, venipuncture et al[26] Articles were excluded, if: reviews and case reports; studies not in English; studies without valid data or with improper data.\nAll standardized measurement scales and tools related to neonates’ painful evaluation would be considered, including: premature infant pain profile (PIPP)[27,28] suitable for newborns between 28 and 40 weeks of gestation (WG); Neonatal Infant Pain Scale (NIPS) from 26 to 47 WG newborns[29]; neonatal facial coding system for 26 to 47 WG newborns.[30] We combined different tools in our systematic review for pain assessment.\nThe main outcome was the neonates’ painful response, according to the suggestion of Pillai Riddell.[11] Secondary outcomes included the variations of heart rate, blood oxygen saturation, respiration rate during and after painful examination and variations of the crying time, cortisol levels, occurrence of adverse events between before and after the painful procedure. Besides, the measurements taken within five minutes after the painful procedure were considered. As for measurements taken before discharge, we included the closest one to discharge.", "We managed all the references in EndNote X9. After removing the duplication, two authors (LNG&TTX) independently reviewed the included studies according to the title and keywords, and then all eligible studies were retained for full-text assessment to determine whether suitable for inclusion in this systematic review. The reasons for the excluded references were recorded. To ensure the consistency of data, two reviewers extracted data independently and stored the data in Review Manager software. Before data analysis, two reviewers checked the data to avoid errors. The above process required two reviewers to reach a consensus, any dissenting opinions about the inclusion were resolved by consulting other reviewers (SRF&YXW).", "The Risk of Bias in the included studies was independently assessed by two reviewers (LNG&TTX) according to version 2 of the Cochrane risk-of-bias tool for randomized trials (RoB2) recommended by Cochrane Handbook for Systematic Reviews of Interventions, including bias arising from the randomization process, bias due to deviations, bias due to incomplete data, bias in measurement of the outcome, bias in selection of the reported result.[31] The risk of bias for each study was classified as low risk of bias, high risk of bias, and some concerns.", "Statistical analysis was conducted in the Review Manager software by using a random-effect model with a 95% confidence interval (CI). Since neonatal pain was assessed using different tools, we treated each tool separately to ensure data consistency. Continuous data were analyzed using mean differences (MD). Dichotomous data were reported using relative risk. If at least two studies reported identical results using the same pain assessment tool, a meta-analysis was conducted by using the random effect model and inverse variance. Subgroup analysis would be conducted according to the different massage method to provide further guidance for clinical practice. Using the chi-square test with a significance level of 0.1 to assess the heterogeneity of included studies. According to the suggestions of preferred reporting items for systematic reviews and meta-analyses-P,[32] we classified I² as not important heterogeneity (0%–40%); moderate heterogeneity (30%–60%); substantial heterogeneity (50%–90%) and considerable heterogeneity (75%–100%).", "The study selection process was showed by Flow Diagram (Fig. 1). A total of 5635 articles were identified through database searching, and 7 related studies were also included through snowballing, gray literature review and other methods. After the removal of 521 duplicated articles, a total of 5121 articles were retained. We eliminated 5089 results in the first screening by reading questions and keywords, and 32 studies were evaluated to be eligible for final selection (full-texts). In total, 21 articles were excluded for different reasons: 2 articles were not randomized controlled trials; 4 articles were not eligible population; 4 articles were not eligible interventions; 1 article was excluded for measurement; 6 articles were excluded because the full text could not be obtained even after contacting the authors; 1 article was not considered for language reasons; 3 articles’ dates could not be used. Finally, we conducted a meta-analysis of 11 studies, including 755 neonates investigating the effects of massage on neonatal pain response compared to standard care.[17,33–42]\nPreferred reporting items for systematic reviews and meta-analyses (PRISMA) Flow diagram of this meta-analysis.", "The 11 studies were published between 2006 and 2021 written in English, and the characteristics were summarized in Table 1. They were carried out in 5 different countries, 2 in China,[33,40] 4 in Iran,[37,39,41,42] 1 in Korea,[34] 2 in Canada,[17,35] and 2 in Turkey.[36,38] Five studies[34,36–38,41] included neonates gestational age older than 37 weeks. Five studies[17,35,39,40,42] included premature younger than 37 weeks of gestational age, and one study[33] included newborns between 30 and 40 weeks of gestational age. All of the studies were randomized controlled trials, and the interventions studied were touch or massage. Seven of the studies were on massage[17,33,36–39,41] and four were on therapeutic touching,[34,35,40,42] but they were all included because the interventions were the same or similar. Moreover, three of the studies were cross-designed[17,33,40] and the rest were parallel designed.\nCharacteristics of included studies.\nNIPS = neonatal infant pain scale, PIPP = premature infant pain profile, RCT = randomized controlled trial, RR = risk ratios.\nIncluded studies evaluated different painful procedures during hospitalization including venipuncture (n = 1),[33] hypothyroidism screening (n = 1),[37] heel-prick (n = 7),[17,34–36,38,39,41] screening for retinopathy (n = 1),[40] and endotracheal suctioning (n = 1).[42]\nDifferent assessment tools were used in the included studies, but all were reliable and valid standard assessment scales. Six used the Neonatal Infant Pain Scale[17,34,36–39] and five used Premature Infant Pain Profile.[33,35,40–42] In addition to pain assessments, several second outcomes were also included in the study, including heart rate, respiration, oxygen saturation and crying time. Moreover, there were some differences in the assessment time between studies. Three studies[24,43,44] were removed from the meta-analysis since the data was not able to be obtained, we reported their results through systematic synthesis.", "The risk on bias of each included study was presented in Fig. 2 and the detail was also summarized (see Supplemental Table S1, http://links.lww.com/MD/H461, Supplemental Content, which summarized the risk of bias in studies). Seven studies reported the methods of random-sequence generation detailedly,[17,33,36–39,42] while others were unclear since insufficient information. Six of the studies described the detail of allocation concealment method appropriately,[17,34–36,38,39] but the rest were assessed as unclear bias risk. All but one study were unclear risks since the nature of the intervention and the difficulty of blinding participants.[33–42] For the blinding of outcome assessment, there were five studies describing it,[17,33,35,38,40] while the rest were uncertain. The risk of incomplete outcome data was high in only one study,[42] because the missing date of different groups was unbalanced and the reasons were different. For selective reporting, four studies[36,37,39,42] were judged low but one[40] was high risk since not all of the prespecified outcomes were reported. Of all the studies, we considered only four[33,38,41,42] to be free from the bias of other sources, and others unclear for insufficient evidence provided.\nRisk of bias summary.", "Six studies[17,34,36–39] including 460 neonates investigated neonatal pain response using NIPS and found that the massage was effective in improving neonatal pain response compared to standard care (MD −2.02; 95% CI −2.63 to −1.42; I2 = 74%; P < .01) (Fig. 3A).\nForest plot displaying the results of pain response score.\nAccording to subgroup analysis of age difference of neonates, massage could effectively improve their pain response regardless of whether the intervention group was full-term infants or premature infants. The differences between subgroups were not statistically significant (P = .89, I2 = 0%) (Fig. 4).\nForest plot displaying the results of subgroup analysis by gestational age of neonates.\nFour studies[33,35,40,42] including 235 neonates investigated the pain response of neonates using the PIPP scale and showed that the pain response scores of neonates who received massage prior to the pain procedure were significantly lower than those who received routine care (MD −3.43; 95% CI −6.05 to −0.80; I2 = 93%; P = .01) (Fig. 3B). According to different intervention measures, subgroup analysis found that the differences between subgroups were statistically significant (P = .05 I2 = 75%) (Fig. 5), and the massage was effective for neonates, while therapeutic touch was not effective for neonates.\nForest plot displaying the results of subgroup analysis by different interventions.", "Three studies (n = 249) investigated the variation of heart rate during the painful procedure,[36,37,42] and there was no significant difference between the massage group and the standard care group (MD 3.39; 95% CI −1.14–7.92; I2 = 45%; P = .14) (Fig. 6A).\nForest plot displaying the results of heart rate.\nFive studies with 332 neonates[17,34,36,37,41] analyzed the variations of heart rate after the painful examination, and the result showed that massage did not lower the heart rate of neonates after procedure compared to routine care (MD −2.15; 95% CI −8.82 to 4.52; I2 = 82%; P = .53) (Fig. 6B).", "Three studies involving 249 neonates[36,37,42] explored the effects of massage on the change of blood oxygen saturation during painful examination, and the merged result showed that massage was more effective than standard care in improving neonate blood oxygen saturation during the examination (MD 2.25; 95% CI 0.28–4.76; I2 = 71%; P = .03) (Fig. 7A).\nForest plot displaying the results of blood oxygen saturation.\nFive studies including 332 participants[17,34,36,37,41] revealed the massage group had a significant advantage over the control group in improving the blood oxygen saturation after the examination (MD 1.05; 95% CI 0.51–1.58; I2 = 0%; P < .01) (Fig. 7B).", "Only two studies[17,37] involving 113 neonates measured the effect of massage on respiration rate after neonatal procedure, and the results showed no effect on respiration rate (MD 0.11; 95% CI −2.61–2.84; I2 = 0%; P = .94) (Fig. 8A).\nForest plot displaying the results of respiratory rate and duration of crying.\nThree studies including 238 neonates[35–37] revealed the effect of massage on crying time during painful procedures, and the merged result showed that neonates in the experimental group spent less time crying than those in the standard care group (MD −48.6; 95% CI −64.32 to −32.88; I ² = 8%; P < .01) (Fig. 8B).", "As fewer than 10 studies were included in our meta-analysis, we were unable to test the symmetry of funnel plot as originally planned. Our retrieval strategy only considered English literature. Although we contacted the authors in different ways, the missing data prevented us from conducting subgroup analysis. Moreover, there may be differences in the standard of care, which may affect our comparisons between studies.", "The authors thank Weifang Health Commission Research Project for the funding on this review.", "Conceptualization: Jiang Liu, Yuxiu Liu.\nData curation: Jiang Liu, Shirong Fang, Lunan Gao, Tingting Xin.\nFormal analysis: Jiang Liu, Lunan Gao.\nFunding acquisition: Yuxia Wang, Yuxiu Liu.\nInvestigation: Shirong Fang, Yuxia Wang.\nMethodology: Jiang Liu, Shirong Fang, Yuxia Wang, Lunan Gao.\nResources: Lunan Gao, Yuxiu Liu.\nSoftware: Jiang Liu, Lunan Gao, Yuxiu Liu.\nSupervision: Shirong Fang, Yuxia Wang, Yuxiu Liu.\nValidation: Shirong Fang, Yuxia Wang, Tingting Xin.\nVisualization: Shirong Fang, Yuxia Wang, Tingting Xin.\nWriting – original draft: Jiang Liu.\nWriting – review & editing: Yuxiu Liu." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Methods", "2.1. Protocol and registration", "2.2. Search strategy and selection criteria", "2.3. Study selection and data extraction", "2.4. Quality assessment", "2.5. Statistical analysis", "3. Results", "3.1. Study selection", "3.2. Characteristics of included studies", "3.3. Evaluation of evidence quality", "3.4. Pain response score", "3.5. Variation of heart rate", "3.6. Variation of blood oxygen saturation", "3.7. Respiratory rate variation and duration of crying", "4. Discussion", "4.1. Limitations", "5. Conclusions", "Disclosure", "Acknowledgments", "Author contributions", "Supplementary Material" ]
[ "Neonates undergo many painful examinations in hospital. Each infant undergoes 7.5 to 17.3 painful examinations every day, such as heel-pricks, venipunctures, etc.[1] Which undoubtedly brings great pain to newborns. It is reported that repeated pain examinations will cause short-term or long-term adverse consequences for neonates,[2–5] such as reducing cerebral blood flow, hinder the development of neurological or motor functions and even cause hypersensitivity to pain.[6–8] This highlights the significance of pain management for infants who experience painful procedures during hospitalization.\nAlthough there are many pharmaceutical interventions that could be able to alleviate the pain of infants, the safety of long-term use remains to be studied.[9] As a safe and reliable pain management method, non-pharmacological intervention is increasingly favored by parents of neonates.[10,11] There are a growing body of researches on non-pharmacological interventions, such as, non-nutritive sucking, skin-to-skin contact, and breastfeeding before painful procedures. These interventions have been proved to be effective in alleviating pain to newborn babies.[12–14] But these interventions require the presence of a mother or one of the parents, which is not always appropriate in a variety of clinical settings. So, we need to explore innovative interventions that can be used anytime in any setting and are effective for pain relief in newborns.\nEffective interventions would ideally be inexpensive, noninvasive, and be rapidly applied to improve pediatric pain control.[13] Massage, as an effective non-pharmacological intervention, has been gradually applied in various areas of clinical practice. It is a method applied by stimulating the acupuncture points and meridians in body using hands or special tools.[15,16] Studies have shown that massage can relieve stress and improve blood circulation by reducing the levels of cortisol and increasing the levels of serotonin and dopamine.[17,18] By stimulating the release of endorphins and serotonin, massage can relieve the pain of neonates, improve their sleep, and have a positive impact on the growth and development of neonates.[19] Besides, massage can also activate the parasympathetic nervous system as a result of stimulation of the vagal nerves and provide calm and rest in the body.[20]\nTo our knowledge, only one systematic review on the non-pharmacological interventions[11] that reported the effectiveness of massage in procedural pain relief of infants previously. However, in this review, the massage was found to be effective in alleviating the pain response of premature infants, but not for neonates. Other recent reviews reported that the massage was safe and effective for neonates’ pain relief.[21] Moreover, a lot of studies have been conducted on the effects of massage for neonatal pain management in recent years, but the results were either controversial or partially effective.[22–24]\nTherefore, a more comprehensive systematic review and meta-analysis is needed to assess the safety and effectiveness of this intervention in the pain management of preterm and full-term infants. The purpose of this study was to comprehensively evaluate the effectiveness of massage on pain relief and other secondary outcomes (the variation of heart rate, respiration, blood oxygen saturation, crying time, cortisol levels, and adverse events) in premature and full-term infants.", "[SUBTITLE] 2.1. Protocol and registration [SUBSECTION] This systematic review protocol was registered in PROSPERO database (CRD42022302115). We conducted this systematic review according to the recommendations of preferred reporting items for systematic reviews and meta-analyses guidelines.[25]\nThis systematic review protocol was registered in PROSPERO database (CRD42022302115). We conducted this systematic review according to the recommendations of preferred reporting items for systematic reviews and meta-analyses guidelines.[25]\n[SUBTITLE] 2.2. Search strategy and selection criteria [SUBSECTION] We conducted electronic searches under the guidance of a library search specialist in the following databases to December 2021: PubMed, Embase, the Cochrane Library and the Clinical Trials Registry. Relevant articles were retrieved by combining the following medical subject headlines (MeSH) and keywords: (Infants OR Premature OR Preterm OR Neonatal OR Prematurity OR Newborn OR Neonate) AND (massage OR touch OR pain management).\nEligible studies included had to fulfill the following criteria: randomized controlled/clinical trials that conducted on the neonates with gestation between 24 and 42 weeks; studies comparing massage interventions (massage, or therapeutic touch) to comparator groups (offering standard care); with outcome including one of the ten most common painful procedures experienced by infants such as heel-prick, venipuncture et al[26] Articles were excluded, if: reviews and case reports; studies not in English; studies without valid data or with improper data.\nAll standardized measurement scales and tools related to neonates’ painful evaluation would be considered, including: premature infant pain profile (PIPP)[27,28] suitable for newborns between 28 and 40 weeks of gestation (WG); Neonatal Infant Pain Scale (NIPS) from 26 to 47 WG newborns[29]; neonatal facial coding system for 26 to 47 WG newborns.[30] We combined different tools in our systematic review for pain assessment.\nThe main outcome was the neonates’ painful response, according to the suggestion of Pillai Riddell.[11] Secondary outcomes included the variations of heart rate, blood oxygen saturation, respiration rate during and after painful examination and variations of the crying time, cortisol levels, occurrence of adverse events between before and after the painful procedure. Besides, the measurements taken within five minutes after the painful procedure were considered. As for measurements taken before discharge, we included the closest one to discharge.\nWe conducted electronic searches under the guidance of a library search specialist in the following databases to December 2021: PubMed, Embase, the Cochrane Library and the Clinical Trials Registry. Relevant articles were retrieved by combining the following medical subject headlines (MeSH) and keywords: (Infants OR Premature OR Preterm OR Neonatal OR Prematurity OR Newborn OR Neonate) AND (massage OR touch OR pain management).\nEligible studies included had to fulfill the following criteria: randomized controlled/clinical trials that conducted on the neonates with gestation between 24 and 42 weeks; studies comparing massage interventions (massage, or therapeutic touch) to comparator groups (offering standard care); with outcome including one of the ten most common painful procedures experienced by infants such as heel-prick, venipuncture et al[26] Articles were excluded, if: reviews and case reports; studies not in English; studies without valid data or with improper data.\nAll standardized measurement scales and tools related to neonates’ painful evaluation would be considered, including: premature infant pain profile (PIPP)[27,28] suitable for newborns between 28 and 40 weeks of gestation (WG); Neonatal Infant Pain Scale (NIPS) from 26 to 47 WG newborns[29]; neonatal facial coding system for 26 to 47 WG newborns.[30] We combined different tools in our systematic review for pain assessment.\nThe main outcome was the neonates’ painful response, according to the suggestion of Pillai Riddell.[11] Secondary outcomes included the variations of heart rate, blood oxygen saturation, respiration rate during and after painful examination and variations of the crying time, cortisol levels, occurrence of adverse events between before and after the painful procedure. Besides, the measurements taken within five minutes after the painful procedure were considered. As for measurements taken before discharge, we included the closest one to discharge.\n[SUBTITLE] 2.3. Study selection and data extraction [SUBSECTION] We managed all the references in EndNote X9. After removing the duplication, two authors (LNG&TTX) independently reviewed the included studies according to the title and keywords, and then all eligible studies were retained for full-text assessment to determine whether suitable for inclusion in this systematic review. The reasons for the excluded references were recorded. To ensure the consistency of data, two reviewers extracted data independently and stored the data in Review Manager software. Before data analysis, two reviewers checked the data to avoid errors. The above process required two reviewers to reach a consensus, any dissenting opinions about the inclusion were resolved by consulting other reviewers (SRF&YXW).\nWe managed all the references in EndNote X9. After removing the duplication, two authors (LNG&TTX) independently reviewed the included studies according to the title and keywords, and then all eligible studies were retained for full-text assessment to determine whether suitable for inclusion in this systematic review. The reasons for the excluded references were recorded. To ensure the consistency of data, two reviewers extracted data independently and stored the data in Review Manager software. Before data analysis, two reviewers checked the data to avoid errors. The above process required two reviewers to reach a consensus, any dissenting opinions about the inclusion were resolved by consulting other reviewers (SRF&YXW).\n[SUBTITLE] 2.4. Quality assessment [SUBSECTION] The Risk of Bias in the included studies was independently assessed by two reviewers (LNG&TTX) according to version 2 of the Cochrane risk-of-bias tool for randomized trials (RoB2) recommended by Cochrane Handbook for Systematic Reviews of Interventions, including bias arising from the randomization process, bias due to deviations, bias due to incomplete data, bias in measurement of the outcome, bias in selection of the reported result.[31] The risk of bias for each study was classified as low risk of bias, high risk of bias, and some concerns.\nThe Risk of Bias in the included studies was independently assessed by two reviewers (LNG&TTX) according to version 2 of the Cochrane risk-of-bias tool for randomized trials (RoB2) recommended by Cochrane Handbook for Systematic Reviews of Interventions, including bias arising from the randomization process, bias due to deviations, bias due to incomplete data, bias in measurement of the outcome, bias in selection of the reported result.[31] The risk of bias for each study was classified as low risk of bias, high risk of bias, and some concerns.\n[SUBTITLE] 2.5. Statistical analysis [SUBSECTION] Statistical analysis was conducted in the Review Manager software by using a random-effect model with a 95% confidence interval (CI). Since neonatal pain was assessed using different tools, we treated each tool separately to ensure data consistency. Continuous data were analyzed using mean differences (MD). Dichotomous data were reported using relative risk. If at least two studies reported identical results using the same pain assessment tool, a meta-analysis was conducted by using the random effect model and inverse variance. Subgroup analysis would be conducted according to the different massage method to provide further guidance for clinical practice. Using the chi-square test with a significance level of 0.1 to assess the heterogeneity of included studies. According to the suggestions of preferred reporting items for systematic reviews and meta-analyses-P,[32] we classified I² as not important heterogeneity (0%–40%); moderate heterogeneity (30%–60%); substantial heterogeneity (50%–90%) and considerable heterogeneity (75%–100%).\nStatistical analysis was conducted in the Review Manager software by using a random-effect model with a 95% confidence interval (CI). Since neonatal pain was assessed using different tools, we treated each tool separately to ensure data consistency. Continuous data were analyzed using mean differences (MD). Dichotomous data were reported using relative risk. If at least two studies reported identical results using the same pain assessment tool, a meta-analysis was conducted by using the random effect model and inverse variance. Subgroup analysis would be conducted according to the different massage method to provide further guidance for clinical practice. Using the chi-square test with a significance level of 0.1 to assess the heterogeneity of included studies. According to the suggestions of preferred reporting items for systematic reviews and meta-analyses-P,[32] we classified I² as not important heterogeneity (0%–40%); moderate heterogeneity (30%–60%); substantial heterogeneity (50%–90%) and considerable heterogeneity (75%–100%).", "This systematic review protocol was registered in PROSPERO database (CRD42022302115). We conducted this systematic review according to the recommendations of preferred reporting items for systematic reviews and meta-analyses guidelines.[25]", "We conducted electronic searches under the guidance of a library search specialist in the following databases to December 2021: PubMed, Embase, the Cochrane Library and the Clinical Trials Registry. Relevant articles were retrieved by combining the following medical subject headlines (MeSH) and keywords: (Infants OR Premature OR Preterm OR Neonatal OR Prematurity OR Newborn OR Neonate) AND (massage OR touch OR pain management).\nEligible studies included had to fulfill the following criteria: randomized controlled/clinical trials that conducted on the neonates with gestation between 24 and 42 weeks; studies comparing massage interventions (massage, or therapeutic touch) to comparator groups (offering standard care); with outcome including one of the ten most common painful procedures experienced by infants such as heel-prick, venipuncture et al[26] Articles were excluded, if: reviews and case reports; studies not in English; studies without valid data or with improper data.\nAll standardized measurement scales and tools related to neonates’ painful evaluation would be considered, including: premature infant pain profile (PIPP)[27,28] suitable for newborns between 28 and 40 weeks of gestation (WG); Neonatal Infant Pain Scale (NIPS) from 26 to 47 WG newborns[29]; neonatal facial coding system for 26 to 47 WG newborns.[30] We combined different tools in our systematic review for pain assessment.\nThe main outcome was the neonates’ painful response, according to the suggestion of Pillai Riddell.[11] Secondary outcomes included the variations of heart rate, blood oxygen saturation, respiration rate during and after painful examination and variations of the crying time, cortisol levels, occurrence of adverse events between before and after the painful procedure. Besides, the measurements taken within five minutes after the painful procedure were considered. As for measurements taken before discharge, we included the closest one to discharge.", "We managed all the references in EndNote X9. After removing the duplication, two authors (LNG&TTX) independently reviewed the included studies according to the title and keywords, and then all eligible studies were retained for full-text assessment to determine whether suitable for inclusion in this systematic review. The reasons for the excluded references were recorded. To ensure the consistency of data, two reviewers extracted data independently and stored the data in Review Manager software. Before data analysis, two reviewers checked the data to avoid errors. The above process required two reviewers to reach a consensus, any dissenting opinions about the inclusion were resolved by consulting other reviewers (SRF&YXW).", "The Risk of Bias in the included studies was independently assessed by two reviewers (LNG&TTX) according to version 2 of the Cochrane risk-of-bias tool for randomized trials (RoB2) recommended by Cochrane Handbook for Systematic Reviews of Interventions, including bias arising from the randomization process, bias due to deviations, bias due to incomplete data, bias in measurement of the outcome, bias in selection of the reported result.[31] The risk of bias for each study was classified as low risk of bias, high risk of bias, and some concerns.", "Statistical analysis was conducted in the Review Manager software by using a random-effect model with a 95% confidence interval (CI). Since neonatal pain was assessed using different tools, we treated each tool separately to ensure data consistency. Continuous data were analyzed using mean differences (MD). Dichotomous data were reported using relative risk. If at least two studies reported identical results using the same pain assessment tool, a meta-analysis was conducted by using the random effect model and inverse variance. Subgroup analysis would be conducted according to the different massage method to provide further guidance for clinical practice. Using the chi-square test with a significance level of 0.1 to assess the heterogeneity of included studies. According to the suggestions of preferred reporting items for systematic reviews and meta-analyses-P,[32] we classified I² as not important heterogeneity (0%–40%); moderate heterogeneity (30%–60%); substantial heterogeneity (50%–90%) and considerable heterogeneity (75%–100%).", "[SUBTITLE] 3.1. Study selection [SUBSECTION] The study selection process was showed by Flow Diagram (Fig. 1). A total of 5635 articles were identified through database searching, and 7 related studies were also included through snowballing, gray literature review and other methods. After the removal of 521 duplicated articles, a total of 5121 articles were retained. We eliminated 5089 results in the first screening by reading questions and keywords, and 32 studies were evaluated to be eligible for final selection (full-texts). In total, 21 articles were excluded for different reasons: 2 articles were not randomized controlled trials; 4 articles were not eligible population; 4 articles were not eligible interventions; 1 article was excluded for measurement; 6 articles were excluded because the full text could not be obtained even after contacting the authors; 1 article was not considered for language reasons; 3 articles’ dates could not be used. Finally, we conducted a meta-analysis of 11 studies, including 755 neonates investigating the effects of massage on neonatal pain response compared to standard care.[17,33–42]\nPreferred reporting items for systematic reviews and meta-analyses (PRISMA) Flow diagram of this meta-analysis.\nThe study selection process was showed by Flow Diagram (Fig. 1). A total of 5635 articles were identified through database searching, and 7 related studies were also included through snowballing, gray literature review and other methods. After the removal of 521 duplicated articles, a total of 5121 articles were retained. We eliminated 5089 results in the first screening by reading questions and keywords, and 32 studies were evaluated to be eligible for final selection (full-texts). In total, 21 articles were excluded for different reasons: 2 articles were not randomized controlled trials; 4 articles were not eligible population; 4 articles were not eligible interventions; 1 article was excluded for measurement; 6 articles were excluded because the full text could not be obtained even after contacting the authors; 1 article was not considered for language reasons; 3 articles’ dates could not be used. Finally, we conducted a meta-analysis of 11 studies, including 755 neonates investigating the effects of massage on neonatal pain response compared to standard care.[17,33–42]\nPreferred reporting items for systematic reviews and meta-analyses (PRISMA) Flow diagram of this meta-analysis.\n[SUBTITLE] 3.2. Characteristics of included studies [SUBSECTION] The 11 studies were published between 2006 and 2021 written in English, and the characteristics were summarized in Table 1. They were carried out in 5 different countries, 2 in China,[33,40] 4 in Iran,[37,39,41,42] 1 in Korea,[34] 2 in Canada,[17,35] and 2 in Turkey.[36,38] Five studies[34,36–38,41] included neonates gestational age older than 37 weeks. Five studies[17,35,39,40,42] included premature younger than 37 weeks of gestational age, and one study[33] included newborns between 30 and 40 weeks of gestational age. All of the studies were randomized controlled trials, and the interventions studied were touch or massage. Seven of the studies were on massage[17,33,36–39,41] and four were on therapeutic touching,[34,35,40,42] but they were all included because the interventions were the same or similar. Moreover, three of the studies were cross-designed[17,33,40] and the rest were parallel designed.\nCharacteristics of included studies.\nNIPS = neonatal infant pain scale, PIPP = premature infant pain profile, RCT = randomized controlled trial, RR = risk ratios.\nIncluded studies evaluated different painful procedures during hospitalization including venipuncture (n = 1),[33] hypothyroidism screening (n = 1),[37] heel-prick (n = 7),[17,34–36,38,39,41] screening for retinopathy (n = 1),[40] and endotracheal suctioning (n = 1).[42]\nDifferent assessment tools were used in the included studies, but all were reliable and valid standard assessment scales. Six used the Neonatal Infant Pain Scale[17,34,36–39] and five used Premature Infant Pain Profile.[33,35,40–42] In addition to pain assessments, several second outcomes were also included in the study, including heart rate, respiration, oxygen saturation and crying time. Moreover, there were some differences in the assessment time between studies. Three studies[24,43,44] were removed from the meta-analysis since the data was not able to be obtained, we reported their results through systematic synthesis.\nThe 11 studies were published between 2006 and 2021 written in English, and the characteristics were summarized in Table 1. They were carried out in 5 different countries, 2 in China,[33,40] 4 in Iran,[37,39,41,42] 1 in Korea,[34] 2 in Canada,[17,35] and 2 in Turkey.[36,38] Five studies[34,36–38,41] included neonates gestational age older than 37 weeks. Five studies[17,35,39,40,42] included premature younger than 37 weeks of gestational age, and one study[33] included newborns between 30 and 40 weeks of gestational age. All of the studies were randomized controlled trials, and the interventions studied were touch or massage. Seven of the studies were on massage[17,33,36–39,41] and four were on therapeutic touching,[34,35,40,42] but they were all included because the interventions were the same or similar. Moreover, three of the studies were cross-designed[17,33,40] and the rest were parallel designed.\nCharacteristics of included studies.\nNIPS = neonatal infant pain scale, PIPP = premature infant pain profile, RCT = randomized controlled trial, RR = risk ratios.\nIncluded studies evaluated different painful procedures during hospitalization including venipuncture (n = 1),[33] hypothyroidism screening (n = 1),[37] heel-prick (n = 7),[17,34–36,38,39,41] screening for retinopathy (n = 1),[40] and endotracheal suctioning (n = 1).[42]\nDifferent assessment tools were used in the included studies, but all were reliable and valid standard assessment scales. Six used the Neonatal Infant Pain Scale[17,34,36–39] and five used Premature Infant Pain Profile.[33,35,40–42] In addition to pain assessments, several second outcomes were also included in the study, including heart rate, respiration, oxygen saturation and crying time. Moreover, there were some differences in the assessment time between studies. Three studies[24,43,44] were removed from the meta-analysis since the data was not able to be obtained, we reported their results through systematic synthesis.\n[SUBTITLE] 3.3. Evaluation of evidence quality [SUBSECTION] The risk on bias of each included study was presented in Fig. 2 and the detail was also summarized (see Supplemental Table S1, http://links.lww.com/MD/H461, Supplemental Content, which summarized the risk of bias in studies). Seven studies reported the methods of random-sequence generation detailedly,[17,33,36–39,42] while others were unclear since insufficient information. Six of the studies described the detail of allocation concealment method appropriately,[17,34–36,38,39] but the rest were assessed as unclear bias risk. All but one study were unclear risks since the nature of the intervention and the difficulty of blinding participants.[33–42] For the blinding of outcome assessment, there were five studies describing it,[17,33,35,38,40] while the rest were uncertain. The risk of incomplete outcome data was high in only one study,[42] because the missing date of different groups was unbalanced and the reasons were different. For selective reporting, four studies[36,37,39,42] were judged low but one[40] was high risk since not all of the prespecified outcomes were reported. Of all the studies, we considered only four[33,38,41,42] to be free from the bias of other sources, and others unclear for insufficient evidence provided.\nRisk of bias summary.\nThe risk on bias of each included study was presented in Fig. 2 and the detail was also summarized (see Supplemental Table S1, http://links.lww.com/MD/H461, Supplemental Content, which summarized the risk of bias in studies). Seven studies reported the methods of random-sequence generation detailedly,[17,33,36–39,42] while others were unclear since insufficient information. Six of the studies described the detail of allocation concealment method appropriately,[17,34–36,38,39] but the rest were assessed as unclear bias risk. All but one study were unclear risks since the nature of the intervention and the difficulty of blinding participants.[33–42] For the blinding of outcome assessment, there were five studies describing it,[17,33,35,38,40] while the rest were uncertain. The risk of incomplete outcome data was high in only one study,[42] because the missing date of different groups was unbalanced and the reasons were different. For selective reporting, four studies[36,37,39,42] were judged low but one[40] was high risk since not all of the prespecified outcomes were reported. Of all the studies, we considered only four[33,38,41,42] to be free from the bias of other sources, and others unclear for insufficient evidence provided.\nRisk of bias summary.\n[SUBTITLE] 3.4. Pain response score [SUBSECTION] Six studies[17,34,36–39] including 460 neonates investigated neonatal pain response using NIPS and found that the massage was effective in improving neonatal pain response compared to standard care (MD −2.02; 95% CI −2.63 to −1.42; I2 = 74%; P < .01) (Fig. 3A).\nForest plot displaying the results of pain response score.\nAccording to subgroup analysis of age difference of neonates, massage could effectively improve their pain response regardless of whether the intervention group was full-term infants or premature infants. The differences between subgroups were not statistically significant (P = .89, I2 = 0%) (Fig. 4).\nForest plot displaying the results of subgroup analysis by gestational age of neonates.\nFour studies[33,35,40,42] including 235 neonates investigated the pain response of neonates using the PIPP scale and showed that the pain response scores of neonates who received massage prior to the pain procedure were significantly lower than those who received routine care (MD −3.43; 95% CI −6.05 to −0.80; I2 = 93%; P = .01) (Fig. 3B). According to different intervention measures, subgroup analysis found that the differences between subgroups were statistically significant (P = .05 I2 = 75%) (Fig. 5), and the massage was effective for neonates, while therapeutic touch was not effective for neonates.\nForest plot displaying the results of subgroup analysis by different interventions.\nSix studies[17,34,36–39] including 460 neonates investigated neonatal pain response using NIPS and found that the massage was effective in improving neonatal pain response compared to standard care (MD −2.02; 95% CI −2.63 to −1.42; I2 = 74%; P < .01) (Fig. 3A).\nForest plot displaying the results of pain response score.\nAccording to subgroup analysis of age difference of neonates, massage could effectively improve their pain response regardless of whether the intervention group was full-term infants or premature infants. The differences between subgroups were not statistically significant (P = .89, I2 = 0%) (Fig. 4).\nForest plot displaying the results of subgroup analysis by gestational age of neonates.\nFour studies[33,35,40,42] including 235 neonates investigated the pain response of neonates using the PIPP scale and showed that the pain response scores of neonates who received massage prior to the pain procedure were significantly lower than those who received routine care (MD −3.43; 95% CI −6.05 to −0.80; I2 = 93%; P = .01) (Fig. 3B). According to different intervention measures, subgroup analysis found that the differences between subgroups were statistically significant (P = .05 I2 = 75%) (Fig. 5), and the massage was effective for neonates, while therapeutic touch was not effective for neonates.\nForest plot displaying the results of subgroup analysis by different interventions.\n[SUBTITLE] 3.5. Variation of heart rate [SUBSECTION] Three studies (n = 249) investigated the variation of heart rate during the painful procedure,[36,37,42] and there was no significant difference between the massage group and the standard care group (MD 3.39; 95% CI −1.14–7.92; I2 = 45%; P = .14) (Fig. 6A).\nForest plot displaying the results of heart rate.\nFive studies with 332 neonates[17,34,36,37,41] analyzed the variations of heart rate after the painful examination, and the result showed that massage did not lower the heart rate of neonates after procedure compared to routine care (MD −2.15; 95% CI −8.82 to 4.52; I2 = 82%; P = .53) (Fig. 6B).\nThree studies (n = 249) investigated the variation of heart rate during the painful procedure,[36,37,42] and there was no significant difference between the massage group and the standard care group (MD 3.39; 95% CI −1.14–7.92; I2 = 45%; P = .14) (Fig. 6A).\nForest plot displaying the results of heart rate.\nFive studies with 332 neonates[17,34,36,37,41] analyzed the variations of heart rate after the painful examination, and the result showed that massage did not lower the heart rate of neonates after procedure compared to routine care (MD −2.15; 95% CI −8.82 to 4.52; I2 = 82%; P = .53) (Fig. 6B).\n[SUBTITLE] 3.6. Variation of blood oxygen saturation [SUBSECTION] Three studies involving 249 neonates[36,37,42] explored the effects of massage on the change of blood oxygen saturation during painful examination, and the merged result showed that massage was more effective than standard care in improving neonate blood oxygen saturation during the examination (MD 2.25; 95% CI 0.28–4.76; I2 = 71%; P = .03) (Fig. 7A).\nForest plot displaying the results of blood oxygen saturation.\nFive studies including 332 participants[17,34,36,37,41] revealed the massage group had a significant advantage over the control group in improving the blood oxygen saturation after the examination (MD 1.05; 95% CI 0.51–1.58; I2 = 0%; P < .01) (Fig. 7B).\nThree studies involving 249 neonates[36,37,42] explored the effects of massage on the change of blood oxygen saturation during painful examination, and the merged result showed that massage was more effective than standard care in improving neonate blood oxygen saturation during the examination (MD 2.25; 95% CI 0.28–4.76; I2 = 71%; P = .03) (Fig. 7A).\nForest plot displaying the results of blood oxygen saturation.\nFive studies including 332 participants[17,34,36,37,41] revealed the massage group had a significant advantage over the control group in improving the blood oxygen saturation after the examination (MD 1.05; 95% CI 0.51–1.58; I2 = 0%; P < .01) (Fig. 7B).\n[SUBTITLE] 3.7. Respiratory rate variation and duration of crying [SUBSECTION] Only two studies[17,37] involving 113 neonates measured the effect of massage on respiration rate after neonatal procedure, and the results showed no effect on respiration rate (MD 0.11; 95% CI −2.61–2.84; I2 = 0%; P = .94) (Fig. 8A).\nForest plot displaying the results of respiratory rate and duration of crying.\nThree studies including 238 neonates[35–37] revealed the effect of massage on crying time during painful procedures, and the merged result showed that neonates in the experimental group spent less time crying than those in the standard care group (MD −48.6; 95% CI −64.32 to −32.88; I ² = 8%; P < .01) (Fig. 8B).\nOnly two studies[17,37] involving 113 neonates measured the effect of massage on respiration rate after neonatal procedure, and the results showed no effect on respiration rate (MD 0.11; 95% CI −2.61–2.84; I2 = 0%; P = .94) (Fig. 8A).\nForest plot displaying the results of respiratory rate and duration of crying.\nThree studies including 238 neonates[35–37] revealed the effect of massage on crying time during painful procedures, and the merged result showed that neonates in the experimental group spent less time crying than those in the standard care group (MD −48.6; 95% CI −64.32 to −32.88; I ² = 8%; P < .01) (Fig. 8B).", "The study selection process was showed by Flow Diagram (Fig. 1). A total of 5635 articles were identified through database searching, and 7 related studies were also included through snowballing, gray literature review and other methods. After the removal of 521 duplicated articles, a total of 5121 articles were retained. We eliminated 5089 results in the first screening by reading questions and keywords, and 32 studies were evaluated to be eligible for final selection (full-texts). In total, 21 articles were excluded for different reasons: 2 articles were not randomized controlled trials; 4 articles were not eligible population; 4 articles were not eligible interventions; 1 article was excluded for measurement; 6 articles were excluded because the full text could not be obtained even after contacting the authors; 1 article was not considered for language reasons; 3 articles’ dates could not be used. Finally, we conducted a meta-analysis of 11 studies, including 755 neonates investigating the effects of massage on neonatal pain response compared to standard care.[17,33–42]\nPreferred reporting items for systematic reviews and meta-analyses (PRISMA) Flow diagram of this meta-analysis.", "The 11 studies were published between 2006 and 2021 written in English, and the characteristics were summarized in Table 1. They were carried out in 5 different countries, 2 in China,[33,40] 4 in Iran,[37,39,41,42] 1 in Korea,[34] 2 in Canada,[17,35] and 2 in Turkey.[36,38] Five studies[34,36–38,41] included neonates gestational age older than 37 weeks. Five studies[17,35,39,40,42] included premature younger than 37 weeks of gestational age, and one study[33] included newborns between 30 and 40 weeks of gestational age. All of the studies were randomized controlled trials, and the interventions studied were touch or massage. Seven of the studies were on massage[17,33,36–39,41] and four were on therapeutic touching,[34,35,40,42] but they were all included because the interventions were the same or similar. Moreover, three of the studies were cross-designed[17,33,40] and the rest were parallel designed.\nCharacteristics of included studies.\nNIPS = neonatal infant pain scale, PIPP = premature infant pain profile, RCT = randomized controlled trial, RR = risk ratios.\nIncluded studies evaluated different painful procedures during hospitalization including venipuncture (n = 1),[33] hypothyroidism screening (n = 1),[37] heel-prick (n = 7),[17,34–36,38,39,41] screening for retinopathy (n = 1),[40] and endotracheal suctioning (n = 1).[42]\nDifferent assessment tools were used in the included studies, but all were reliable and valid standard assessment scales. Six used the Neonatal Infant Pain Scale[17,34,36–39] and five used Premature Infant Pain Profile.[33,35,40–42] In addition to pain assessments, several second outcomes were also included in the study, including heart rate, respiration, oxygen saturation and crying time. Moreover, there were some differences in the assessment time between studies. Three studies[24,43,44] were removed from the meta-analysis since the data was not able to be obtained, we reported their results through systematic synthesis.", "The risk on bias of each included study was presented in Fig. 2 and the detail was also summarized (see Supplemental Table S1, http://links.lww.com/MD/H461, Supplemental Content, which summarized the risk of bias in studies). Seven studies reported the methods of random-sequence generation detailedly,[17,33,36–39,42] while others were unclear since insufficient information. Six of the studies described the detail of allocation concealment method appropriately,[17,34–36,38,39] but the rest were assessed as unclear bias risk. All but one study were unclear risks since the nature of the intervention and the difficulty of blinding participants.[33–42] For the blinding of outcome assessment, there were five studies describing it,[17,33,35,38,40] while the rest were uncertain. The risk of incomplete outcome data was high in only one study,[42] because the missing date of different groups was unbalanced and the reasons were different. For selective reporting, four studies[36,37,39,42] were judged low but one[40] was high risk since not all of the prespecified outcomes were reported. Of all the studies, we considered only four[33,38,41,42] to be free from the bias of other sources, and others unclear for insufficient evidence provided.\nRisk of bias summary.", "Six studies[17,34,36–39] including 460 neonates investigated neonatal pain response using NIPS and found that the massage was effective in improving neonatal pain response compared to standard care (MD −2.02; 95% CI −2.63 to −1.42; I2 = 74%; P < .01) (Fig. 3A).\nForest plot displaying the results of pain response score.\nAccording to subgroup analysis of age difference of neonates, massage could effectively improve their pain response regardless of whether the intervention group was full-term infants or premature infants. The differences between subgroups were not statistically significant (P = .89, I2 = 0%) (Fig. 4).\nForest plot displaying the results of subgroup analysis by gestational age of neonates.\nFour studies[33,35,40,42] including 235 neonates investigated the pain response of neonates using the PIPP scale and showed that the pain response scores of neonates who received massage prior to the pain procedure were significantly lower than those who received routine care (MD −3.43; 95% CI −6.05 to −0.80; I2 = 93%; P = .01) (Fig. 3B). According to different intervention measures, subgroup analysis found that the differences between subgroups were statistically significant (P = .05 I2 = 75%) (Fig. 5), and the massage was effective for neonates, while therapeutic touch was not effective for neonates.\nForest plot displaying the results of subgroup analysis by different interventions.", "Three studies (n = 249) investigated the variation of heart rate during the painful procedure,[36,37,42] and there was no significant difference between the massage group and the standard care group (MD 3.39; 95% CI −1.14–7.92; I2 = 45%; P = .14) (Fig. 6A).\nForest plot displaying the results of heart rate.\nFive studies with 332 neonates[17,34,36,37,41] analyzed the variations of heart rate after the painful examination, and the result showed that massage did not lower the heart rate of neonates after procedure compared to routine care (MD −2.15; 95% CI −8.82 to 4.52; I2 = 82%; P = .53) (Fig. 6B).", "Three studies involving 249 neonates[36,37,42] explored the effects of massage on the change of blood oxygen saturation during painful examination, and the merged result showed that massage was more effective than standard care in improving neonate blood oxygen saturation during the examination (MD 2.25; 95% CI 0.28–4.76; I2 = 71%; P = .03) (Fig. 7A).\nForest plot displaying the results of blood oxygen saturation.\nFive studies including 332 participants[17,34,36,37,41] revealed the massage group had a significant advantage over the control group in improving the blood oxygen saturation after the examination (MD 1.05; 95% CI 0.51–1.58; I2 = 0%; P < .01) (Fig. 7B).", "Only two studies[17,37] involving 113 neonates measured the effect of massage on respiration rate after neonatal procedure, and the results showed no effect on respiration rate (MD 0.11; 95% CI −2.61–2.84; I2 = 0%; P = .94) (Fig. 8A).\nForest plot displaying the results of respiratory rate and duration of crying.\nThree studies including 238 neonates[35–37] revealed the effect of massage on crying time during painful procedures, and the merged result showed that neonates in the experimental group spent less time crying than those in the standard care group (MD −48.6; 95% CI −64.32 to −32.88; I ² = 8%; P < .01) (Fig. 8B).", "To our knowledge, this is the first systematic review and meta-analysis to examine the effectiveness of massage in relieving procedural pain in neonates during hospitalizations. We conducted all major and second outcomes of neonates’ pain response including neonates’ pain behaviors, heart rate, respiration, blood oxygen saturation, crying time, cortisol level and adverse events. Findings of 11 studies were synthesized in this review. For studies with the same measurement tools, they were combined for meta-analysis, if not, we conducted a separate evaluation in the form of narration. In the meta-analysis, we synthesized the results of 6 studies[17,34,36–39] using NIPS, and found that massage had a significant effect on neonates’ pain relief compared to standard care. Subgroup analysis found that massage was effective for both full-term and premature neonate. There were 4 studies[33,35,40,42] using PIPP to evaluate neonates for pain, and the meta-analysis results were the same. Subgroup analysis based on the different massage method showed that massage was more effective in relieving procedural pain than therapeutic touch.\nIn addition, we also found, through meta-analysis, massage had no effect on heart rate changes of neonates (during or after the procedure) compared with the control group. However, it had positive effects on blood oxygen saturation during and after the procedure. For crying time, neonates receiving massage had less crying time than neonates receiving standard care. However, in terms of respiration, we found massage had no significant effect on neonates.\nFor cortisol level, only one study reported that cortisol level in the experimental group had no significant differences compared to the control group.[17] We could not conduct a meta-analysis on the occurrence of adverse events, because only a few authors explored the safety of massage, and relevant outcomes were not reported in all studies.\nIn the studies using NIPS, although the meta-analysis results showed that massage could effectively relieve the pain of neonates, the quality of evidence was not relatively high, which may be due to the clinical heterogeneity caused by differences in the different massage method and gestational age of neonates. We performed subgroup analysis based on whether the gestational age of neonates was greater than 37 weeks. The study found differences in gestational age did not influence the neonatal pain response (Fig. 4). While, in the study of neonates with gestational age less than 37 weeks, we consider the results stable and reliable, since the heterogeneity is small (P < .01, I² = 0%). In addition, we also found differences in the massage method across studies, which may cause clinical heterogeneity. But due to the lack of original studies, we were unable to do subgroup analyses based on differences in the massage method.\nThe results of subgroup analyses in studies using PIPP suggested the differences in the massage method (P = .05, I² = 75%) were likely to be the main source of heterogeneity. We also found the massage was more effective than therapeutic touch on neonatal pain relief. However, because of the insufficient number of original studies, we were unable to conduct subgroup analysis based on gestational age differences. There was still to be further strengthened in the original research. Noticeably, the different assessment tools were analyzed separately to reduce methodological heterogeneity caused by differences in assessment methods and improved the quality of evidence. In addition, analysis based on gender differences in future studies could help develop knowledge of the effectiveness of interventions.\nAs a non-pharmacological intervention, massage could be easily implemented at low cost and had few side effects.[22] Although it may not be readily accessible for all NICUs, principles guiding non-pharmacological interventions, as massage, should still be encouraged to relieve painful reactions of neonate. The American Academy of Pediatrics considers it is necessary to combine various interventions of non-pharmacological to reinforce their effectiveness,[9] and recent studies have found that multisensory stimulation had a positive effect on neonates in procedural pain relief.[45] Massage can also be used in conjunction with other non-pharmacological interventions,[46] such as olfactory stimulation and music therapy. Recent research found that multisensory stimulation had a significant effect on reducing procedural pain of neonate.[47,48] Future studies may consider combining massage with other sensory interventions to evaluate the effectiveness of pain relief in neonates.\nOur systematic review considered different pain assessment tools to evaluate massage for neonatal pain management during hospitalization. Although they are standard tools, there are some differences in measurement. In future studies, the combination of different tools is conducive to a more comprehensive assessment of the effectiveness of the intervention. One of the obvious problems for the pain assessment of neonate is the absence of a “gold standard”, and sometimes pain scales also can’t reflect painful response very well. Recently, one study showed that skin conductance (SC), as a physiological method, was an effective method in pain assessment of neonates. When pain occurs in neonates, the sympathetic nerve will respond to the stimulus. Skin conductance can evaluate pain by detecting changes in skin electrical activity and calculate SC values.[49] Therefore, we suggest the measurement of SC should be combined with other standardized devices to be used in future neonatal pain measures.\n[SUBTITLE] 4.1. Limitations [SUBSECTION] As fewer than 10 studies were included in our meta-analysis, we were unable to test the symmetry of funnel plot as originally planned. Our retrieval strategy only considered English literature. Although we contacted the authors in different ways, the missing data prevented us from conducting subgroup analysis. Moreover, there may be differences in the standard of care, which may affect our comparisons between studies.\nAs fewer than 10 studies were included in our meta-analysis, we were unable to test the symmetry of funnel plot as originally planned. Our retrieval strategy only considered English literature. Although we contacted the authors in different ways, the missing data prevented us from conducting subgroup analysis. Moreover, there may be differences in the standard of care, which may affect our comparisons between studies.", "As fewer than 10 studies were included in our meta-analysis, we were unable to test the symmetry of funnel plot as originally planned. Our retrieval strategy only considered English literature. Although we contacted the authors in different ways, the missing data prevented us from conducting subgroup analysis. Moreover, there may be differences in the standard of care, which may affect our comparisons between studies.", "In general, massage intervention plays a positive role in the relief of painful procedures in neonates, and we recommend it be used in clinical practice. Adverse events of massage intervention should be reported in future studies to guide clinical study and ensure the massage is carried out safely. In hospitals, especially in the NICU, neonates undergo a variety of painful procedures. So, it is imperative to identify the most effective interventions to manage procedural pain during hospitalization of neonate.", "The authors report no conflicts of interest in this work.", "The authors thank Weifang Health Commission Research Project for the funding on this review.", "Conceptualization: Jiang Liu, Yuxiu Liu.\nData curation: Jiang Liu, Shirong Fang, Lunan Gao, Tingting Xin.\nFormal analysis: Jiang Liu, Lunan Gao.\nFunding acquisition: Yuxia Wang, Yuxiu Liu.\nInvestigation: Shirong Fang, Yuxia Wang.\nMethodology: Jiang Liu, Shirong Fang, Yuxia Wang, Lunan Gao.\nResources: Lunan Gao, Yuxiu Liu.\nSoftware: Jiang Liu, Lunan Gao, Yuxiu Liu.\nSupervision: Shirong Fang, Yuxia Wang, Yuxiu Liu.\nValidation: Shirong Fang, Yuxia Wang, Tingting Xin.\nVisualization: Shirong Fang, Yuxia Wang, Tingting Xin.\nWriting – original draft: Jiang Liu.\nWriting – review & editing: Yuxiu Liu.", "" ]
[ "intro", "methods", null, null, null, null, null, "results", null, null, null, null, null, null, null, "discussion", null, "conclusions", "COI-Statement", null, null, "supplementary-material" ]
[ "massage", "neonate", "pain, non-", "pharmacological interventions" ]
Cost-effectiveness of home care services versus hospital care for pediatric patients worldwide: A protocol for systematic review and meta-analysis.
36254039
Despite the expansion of home care services (HCS) in several countries, there is still a need to systematically investigate the available evidence on the cost-effectiveness of this type of service compared to hospital care in the world, particularly for the pediatric population. Hence, we aimed to systematically synthesize and critically evaluate the evidence on the cost-effectiveness of HCS versus in-hospital services worldwide.
BACKGROUND
A systematic review and meta-analysis protocol guided by Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols. Ten databases will be searched: MEDLINE/PubMed, Cochrane Library, Excerpta Medica database, cummulative index to nursing and allied health literature (CINAHL), Web of Science, SCOPUS, Science Direct, PsycINFO, Latin American and Caribbean Health Sciences Literature and Chinese national knowledge infrastructure with no restrictions on publication date or languages. A checklist for assessing the quality of reporting of economic evaluation studies will be applied. To assess the methodological quality of evidence from observational research on comparative effectiveness, the Good Research for Comparative Effectiveness Checklist v5.0 will be used. The heterogeneity among the studies will be assessed using the I2 statistic test. According to the results of this test, we will verify whether a meta-analysis is feasible. If feasibility is confirmed, a random-effect model analysis will be carried out. For data analysis, the calculation of the pooled effect estimates will consider a 95% CI and alpha will be set in 0.05 using the R statistical software, v.4.0.4. In addition, we will rate the certainty of evidence based on Grading of Recommendations Assessment, Development and Evaluation. All methodological steps of this review will be performed independently and paired by 2 reviewers and conducted and managed in the EPPI-Reviewer Software™.
METHODS
The results may have relevance for the basis of public health policies, regarding the forms of organization of HCS, especially in terms of complete economic evaluations through cost-effectiveness analysis in relation to hospital care.
RESULTS
To the best of our knowledge this will be the first systematic review and metanalysis to synthesize and critically evaluate the evidence on the cost-effectiveness of HCS versus in-hospital services worldwide. The review will adopt a rigorous approach, adhering to PRISMA Statement 2020, using a comprehensive and systematic search strategy in 10 databases, further the gray literature, pre-prints, with no time period or language restrictions.
CONCLUSION
[ "Child", "Cost-Benefit Analysis", "Home Care Services", "Hospitals", "Humans", "Meta-Analysis as Topic", "Review Literature as Topic", "Systematic Reviews as Topic" ]
9575704
1. Introduction
Worldwide, Synchronously, the changes in the demographic and epidemiological transition that are taking place in most countries regardless of income level, pointing to a situation of triple burden of disease with the hegemonic presence of chronic conditions,[1] the need to adapt the health care model has emerged, leading many countries to think of Home Care Services (HCS) as a strategic point of care for health care.[2] The increase of HCS in several countries follows, in parallel, the interest of health systems in the process of de-hospitalization, rationalization of the use of hospital beds, cost reduction and organization of patient-centered care. The demand for HCS appears, therefore, as another challenge for health systems, contributing to change the focus of care and the environment in which care is provided.[2,3] In addition, continued care in the home environment is accompanied by other equally relevant health needs, such as the aging population, as well as the care provided to premature babies, to children with special needs and chronic diseases, to adults with multiple chronic-degenerative diseases, to individuals in palliative care, life support and rehabilitation. Thus, the relevance of the need to implement HCS stands out in the current and future health agenda of all health systems, aiming to contribute to the configuration of substitutive health networks and the transformation of health practice.[2–4] Researchers already question the real need for hospitalization for certain health problems, considering some reasons for hospitalization as dispensable or unnecessarily prolonged, and that they can be replaced or complemented by an HCS.[2–5] In addition to cost reduction, the HCS has been representing in the world scenario the connotation of offering quality care, providing welfare and comfort by allowing the patient to stay in their home environment, integrated into their life context.[5] It is important to emphasize that health financing is a topic that always raises debates in order to better define the use and allocation of resources for society as a whole. Most countries face increasing health care costs, both in absolute and relative terms, regardless of whether the financing model adopted is public, private, based on tax collection, or through direct user fees. The growth of health expenditures, with the need to seek efficiency in resource allocation, has occupied an important role in public policy discussions.[6] Economic evaluation studies, such as those of cost-effectiveness, are adopted in order to consider the cost factor in decision-making regarding the new technologies or models of health care, since financial resources are scarce and finite. In the evaluation of health care models, cost-effectiveness analysis is the most suitable method to compare 2 or more alternatives because it allows the combined analysis of clinical benefits and associated costs, providing objective and explicit data for decision making.[6] Cost-effectiveness studies can be understood as a tool to analyze the value of health interventions, since the method seeks to fill a gap between preferences and science. On one hand, there is the subjectivity of the preference that an individual or society has when faced with 2 options. On the other, there is the objectivity and the reproducibility of science, considering that the cost of a new technology or model of care needs to be manage.[7] In cost-effectiveness evaluation, costs are confronted with clinical outcomes in an attempt to understand the impact of different alternatives, identifying those with better intervention effects, in general, in exchange for a lower cost to health systems.[6–8] It is known that several countries are in the process of adapting to this new demand for care, due especially to issues of economic viability.[2,3] This adaptation has strengthened the emergence of new strategies and mechanisms for health care, such as HCS, which combines technological and scientific resources present in the hospital with the family environment.[9] The home has emerged, added to its humanizing characteristic and the demographic and epidemiological profile of the population, as a place with potential to expand and qualify the care processes.[10] Moreover, the development of HCS on the world scene has been following demographic and epidemiological changes[11] and is related to reducing the risk of infections[12] the humanization of care and quality of life, greater involvement of family members with the patient’s illness, closer relationship between the health team, patient and family,[13] cost reduction, increased hospital bed turnover with bed management, de-hospitalization[14] lower rates of clinical worsening and acute complications, less demand for urgency and emergency services, and lower readmission rates,[15] implementation of palliative care[3] and effective actions for prevention, promotion and recovery of health.[4] A systematic review and meta-analysis aimed at evaluating the effect of “hospital at home” services in adults >16 years that significantly replace inpatient time on health outcomes, showed that hospital at home is associated with reductions in mortality, re-admission rates and costs, and increases in patients and caregiver satisfaction, but no change in caregiver burden.[16] Despite the expansion of HCS in several countries, there is still a need to systematically investigate the available evidence on the cost-effectiveness of this type of service in relation to hospital care in the world, particularly for the pediatric population. Previous systematic review published in 2012 on the costs and effectiveness of pediatric home care pointed out that it can provide equivalent clinical outcomes for children and does not impose a greater burden on families.[17] In fact, in some cases, there is evidence of reduced burden and costs to families compared to hospital care. There is also growing evidence, although based on weaker evidence, that pediatric home care can reduce health care costs, particularly for children with complex and long-term needs.[18] It is noteworthy that this review was limited to the period from 1990 to 2007, and used simple methodological assessment tools available at the time. Since there is an increasing use of decision analysis models in economic evaluations of technology-assisted care models, the use of specific methodological guidelines with the main aspects of modeling is necessary,[18] since they can indicate how credible and relevant the result obtained is to inform decision makers. In this sense, systematic reviews of economic evaluations are important to synthesize the best evidence already available, in order to contribute to the formulation of guidelines for a new analysis, especially taking into account the most recent period (until 2022), in a context of contemporary transformations in health systems, and to identify the most relevant evaluations to inform a particular issue.[19] Hence, the aim of this study is to systematically synthesize and critically evaluate the evidence on cost-effectiveness of home care versus in-hospital services in pediatric patients worldwide.
2. Methods
[SUBTITLE] 2.1. Study design [SUBSECTION] This is a systematic review and meta-analysis protocol guided by the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols.[20] To ensure the reliability of the data and methodological transparency of this review, the protocol was submitted to the International Prospective Register of Systematic Reviews (NHS) for registration (Registration Number: CRD42022329687). This is a systematic review and meta-analysis protocol guided by the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols.[20] To ensure the reliability of the data and methodological transparency of this review, the protocol was submitted to the International Prospective Register of Systematic Reviews (NHS) for registration (Registration Number: CRD42022329687). [SUBTITLE] 2.2. Research question [SUBSECTION] In order to formulate the research question, the PECOS strategy was used[21] (P - Population or Patients; E - Exposure; C - Comparison; O - outcomes; S - study design), where P = Population = evidence of complete economic evaluations in pediatric patients, E = home care services, C = Comparison = hospital care (in-hospital services), O = Outcomes = mortality, readmission rates, patient and caregiver satisfaction, and cost; S = Study type: cost-effectiveness, cost-utility and cost-effectiveness evaluation studies. In this study our population was stated in line with the World Health Organization definition, that is, “child” as a person under 19 years of age, an “adolescent” as a person aged 10 to 19 years, an “infant” as a person aged 0 to 11 months, and a “newborn” as a person aged 0 to 28 days.[22] The acronym PECOS guided the structuring of the formulation of the research question: “What is the cost-effectiveness of home care compared to hospital care for pediatric patients in the world?" In order to formulate the research question, the PECOS strategy was used[21] (P - Population or Patients; E - Exposure; C - Comparison; O - outcomes; S - study design), where P = Population = evidence of complete economic evaluations in pediatric patients, E = home care services, C = Comparison = hospital care (in-hospital services), O = Outcomes = mortality, readmission rates, patient and caregiver satisfaction, and cost; S = Study type: cost-effectiveness, cost-utility and cost-effectiveness evaluation studies. In this study our population was stated in line with the World Health Organization definition, that is, “child” as a person under 19 years of age, an “adolescent” as a person aged 10 to 19 years, an “infant” as a person aged 0 to 11 months, and a “newborn” as a person aged 0 to 28 days.[22] The acronym PECOS guided the structuring of the formulation of the research question: “What is the cost-effectiveness of home care compared to hospital care for pediatric patients in the world?" [SUBTITLE] 2.3. Search strategy [SUBSECTION] The search for studies will be conducted systematically in ten electronic data-bases: Medical Literature Analysis and Retrieval System Online (MEDLINE) via PubMed, Excerpta Medica database, Cochrane Library, Web of Science, SCOPUS, Science Direct, Latin American and Caribbean Health Sciences Literature, Psychology Information (PsycINFO), cummulative index to nursing and allied health literature (CINAHL), and the Chinese National Knowledge Infrastructure. The strategy for seeking the studies will consist of a combination of controlled descriptors (indexers in the respective databases), synonyms, and keywords, as indicated in each electronic database. Thus, to search for articles in MEDLINE, we will use the Medical Subject Headings (MeSH) as controlled descriptors; the Emtree terms will be consulted in excerpta medica database; the PsycINFO Thesaurus will be consulted for the PsycINFO database; the DeCS (Health Sciences Descriptors) in the Latin American and Caribbean Health Sciences Literature database, and the CINAHL headings in the CINAHL database. It should be noted that there will be no date or language restriction in the search strategy to be performed. In addition to the electronic databases cited above, searches will be held on Clinical Trial Registry sites such as ClinicalTrials. gov (National Institutes of Health, NIH); World Health Organization International Clinical Trials Registry Platform, and the Brazilian Registry of Clinical Trials, as well as additional searches in sites of organizations and websites search, such as: The British Library, SciELO, Google Scholar, ProQuest Dissertations and Theses Global, Public Health Gray Literature Sources and Health Evidence and in the preprints for Health Sciences (medRxiv). Additionally, the list of final references from the included primary studies will be reviewed manually to find relevant studies to be added. The search strategy will be carried out by 2 researchers independently (LCLJ and RMP) according to the recommendations of the Cochrane Handbook.[23] Initially, the existence of an index of specific subject headings in each database (such as MeSH terms, Emtree terms, PsycINFO Thesaurus, DeCS and CINAHL headings and their synonyms and keywords will be identified. Subsequently, the search terms will be combined using the Boolean operators “AND” and “OR”[24,25] in order to obtain restrictive and additive combinations, respectively. In addition, the search will be performed using the identified descriptors and with expanded meaning, without the use of database filters to preserve significant samples and ensure lower risk of loss. The search strategy combining the MeSH controlled descriptors and keywords that will be used in MEDLINE will be adjusted to the other electronic databases as described in Table 1. Preliminary search strategy in the MEDLINE via PubMed. MEDLINE = medical literature analysis and retrieval system online. In this search strategy phase the EndNote™ reference manager will be used to store, organize and delete duplicates to ensure a systematic, comprehensive and manageable search. The search for studies will be conducted systematically in ten electronic data-bases: Medical Literature Analysis and Retrieval System Online (MEDLINE) via PubMed, Excerpta Medica database, Cochrane Library, Web of Science, SCOPUS, Science Direct, Latin American and Caribbean Health Sciences Literature, Psychology Information (PsycINFO), cummulative index to nursing and allied health literature (CINAHL), and the Chinese National Knowledge Infrastructure. The strategy for seeking the studies will consist of a combination of controlled descriptors (indexers in the respective databases), synonyms, and keywords, as indicated in each electronic database. Thus, to search for articles in MEDLINE, we will use the Medical Subject Headings (MeSH) as controlled descriptors; the Emtree terms will be consulted in excerpta medica database; the PsycINFO Thesaurus will be consulted for the PsycINFO database; the DeCS (Health Sciences Descriptors) in the Latin American and Caribbean Health Sciences Literature database, and the CINAHL headings in the CINAHL database. It should be noted that there will be no date or language restriction in the search strategy to be performed. In addition to the electronic databases cited above, searches will be held on Clinical Trial Registry sites such as ClinicalTrials. gov (National Institutes of Health, NIH); World Health Organization International Clinical Trials Registry Platform, and the Brazilian Registry of Clinical Trials, as well as additional searches in sites of organizations and websites search, such as: The British Library, SciELO, Google Scholar, ProQuest Dissertations and Theses Global, Public Health Gray Literature Sources and Health Evidence and in the preprints for Health Sciences (medRxiv). Additionally, the list of final references from the included primary studies will be reviewed manually to find relevant studies to be added. The search strategy will be carried out by 2 researchers independently (LCLJ and RMP) according to the recommendations of the Cochrane Handbook.[23] Initially, the existence of an index of specific subject headings in each database (such as MeSH terms, Emtree terms, PsycINFO Thesaurus, DeCS and CINAHL headings and their synonyms and keywords will be identified. Subsequently, the search terms will be combined using the Boolean operators “AND” and “OR”[24,25] in order to obtain restrictive and additive combinations, respectively. In addition, the search will be performed using the identified descriptors and with expanded meaning, without the use of database filters to preserve significant samples and ensure lower risk of loss. The search strategy combining the MeSH controlled descriptors and keywords that will be used in MEDLINE will be adjusted to the other electronic databases as described in Table 1. Preliminary search strategy in the MEDLINE via PubMed. MEDLINE = medical literature analysis and retrieval system online. In this search strategy phase the EndNote™ reference manager will be used to store, organize and delete duplicates to ensure a systematic, comprehensive and manageable search. [SUBTITLE] 2.4. Eligibility [SUBSECTION] A summary of the inclusion and exclusion criteria of this systematic review is depicted in Table 2 in line with the acronym PECOS. Inclusion and exclusion criteria. Regarding the study design, we will include analytical observational study designs and experimental studies, as well as gray literature, as recommended by the Cochrane Handbook.[23] Thus, studies that investigated epidemiological and clinical aspects on the cost-effectiveness of HCS in pediatric patients (<19 years) in relation to in-hospital services will be included in this systematic review. Studies that do not present complete economic evaluations (evidence without cost, benefit and effectiveness analysis), and studies involving adult and/or elderly patients will be excluded. The selection of studies will also be performed by 2 reviewers independently (LCLJ and RMP) and blinded. After this selection, a third reviewer (RAGL) will be responsible for analyzing and deciding on the inclusion or exclusion of each article, especially those containing conflicting decisions. In this step of inclusion and exclusion of articles that will compose the final sample, the Rayyan™-Qatar CRI,[26] as a tool to assist in the eligibility/selection of articles. A summary of the inclusion and exclusion criteria of this systematic review is depicted in Table 2 in line with the acronym PECOS. Inclusion and exclusion criteria. Regarding the study design, we will include analytical observational study designs and experimental studies, as well as gray literature, as recommended by the Cochrane Handbook.[23] Thus, studies that investigated epidemiological and clinical aspects on the cost-effectiveness of HCS in pediatric patients (<19 years) in relation to in-hospital services will be included in this systematic review. Studies that do not present complete economic evaluations (evidence without cost, benefit and effectiveness analysis), and studies involving adult and/or elderly patients will be excluded. The selection of studies will also be performed by 2 reviewers independently (LCLJ and RMP) and blinded. After this selection, a third reviewer (RAGL) will be responsible for analyzing and deciding on the inclusion or exclusion of each article, especially those containing conflicting decisions. In this step of inclusion and exclusion of articles that will compose the final sample, the Rayyan™-Qatar CRI,[26] as a tool to assist in the eligibility/selection of articles. [SUBTITLE] 2.5. Data extraction [SUBSECTION] First, the screening of the studies will be based on the information contained in their titles and abstracts and will be performed by the same 2 researchers (LCLJ and RMP). When the reviewers disagree, the article will be evaluated, and if the disagreement persists, a third reviewer (RAGL) will make a final decision. Once consensus is reached on the selected studies, a specific standardized form for extracting previously published full economic evaluation studies will be used.[6,27] The form is divided into 6 sections, according to the types of information made available by the studies: General information about the selected studies; Information about the study design, the population included and the comparators used; Information about the details of the economic model, time horizon and assumptions used and about the sensitivity analyses performed; Information about the costs; Sources of the data used in the study: epidemiological, intervention, effectiveness, cost and utility; Outcomes assessed in the studies and their measures and details of the sensitivity analysis.[27] First, the screening of the studies will be based on the information contained in their titles and abstracts and will be performed by the same 2 researchers (LCLJ and RMP). When the reviewers disagree, the article will be evaluated, and if the disagreement persists, a third reviewer (RAGL) will make a final decision. Once consensus is reached on the selected studies, a specific standardized form for extracting previously published full economic evaluation studies will be used.[6,27] The form is divided into 6 sections, according to the types of information made available by the studies: General information about the selected studies; Information about the study design, the population included and the comparators used; Information about the details of the economic model, time horizon and assumptions used and about the sensitivity analyses performed; Information about the costs; Sources of the data used in the study: epidemiological, intervention, effectiveness, cost and utility; Outcomes assessed in the studies and their measures and details of the sensitivity analysis.[27] [SUBTITLE] 2.6. Methodological evaluation [SUBSECTION] The assessment of the methodological quality of the studies will be defined as an essential process to establish internal validity, checking for possible biases and reliability of the evidence identified. Initially, the level of evidence will be identified and classified according to the scale developed by the Oxford Centre for Evidence-Based Medicine that is based on study design and classifies them into 1A, 1B, 1C, 2A, 2B, 2C, 3A, 3B, 4, and 5 (Table 3).[28] Classification and hierarchy of evidence. RCT = randomized controlled trials. *From the Centre for Evidence-Based Medicine, http://www.cebm.net Subsequently, the checklist for assessing the quality of reporting of economic evaluation studies proposed by The British Medical Journal will be applied[29] that allows the evaluation of the items of an economic evaluation. This checklist is divided into 3 blocks of questions: study design; data collection; and analysis and interpretation of results. The internal validity and risk of bias of randomized controlled trials will be assessed using the Cochrane Risk-Of-Bias tool revised for randomized controlled trials.[30] To assess the risk of bias in quasi-experimental studies, we will use the Risk of Bias In Non-randomized Studies of Interventions.[31] In addition, the Newcastle-Ottawa Scale[32] will be used to assess the internal validity and risk of bias of cohort studies. Cross-sectional studies will be evaluated using the Agency for Healthcare Research and Quality tool.[33] The same 2 reviewers (LCLJ and RMP) will perform the critical appraisal independently. To assess the methodological quality of the comparative effectiveness observational research evidence we will use the Good Research for Comparative Effectiveness Checklist v5.0.[34] The assessment of the methodological quality of the studies will be defined as an essential process to establish internal validity, checking for possible biases and reliability of the evidence identified. Initially, the level of evidence will be identified and classified according to the scale developed by the Oxford Centre for Evidence-Based Medicine that is based on study design and classifies them into 1A, 1B, 1C, 2A, 2B, 2C, 3A, 3B, 4, and 5 (Table 3).[28] Classification and hierarchy of evidence. RCT = randomized controlled trials. *From the Centre for Evidence-Based Medicine, http://www.cebm.net Subsequently, the checklist for assessing the quality of reporting of economic evaluation studies proposed by The British Medical Journal will be applied[29] that allows the evaluation of the items of an economic evaluation. This checklist is divided into 3 blocks of questions: study design; data collection; and analysis and interpretation of results. The internal validity and risk of bias of randomized controlled trials will be assessed using the Cochrane Risk-Of-Bias tool revised for randomized controlled trials.[30] To assess the risk of bias in quasi-experimental studies, we will use the Risk of Bias In Non-randomized Studies of Interventions.[31] In addition, the Newcastle-Ottawa Scale[32] will be used to assess the internal validity and risk of bias of cohort studies. Cross-sectional studies will be evaluated using the Agency for Healthcare Research and Quality tool.[33] The same 2 reviewers (LCLJ and RMP) will perform the critical appraisal independently. To assess the methodological quality of the comparative effectiveness observational research evidence we will use the Good Research for Comparative Effectiveness Checklist v5.0.[34] [SUBTITLE] 2.7. Data analysis and evidence synthesis [SUBSECTION] Heterogeneity among studies will be measured by the I2 statistic to estimate the percentage of variation among studies, where I2 = 0% to 40% indicates low heterogeneity; I2 = 30% to 60% moderate heterogeneity; I2 = 50% to 90% substantial heterogeneity; and I2 = 75% to 100% high heterogeneity.[35,36] According to the I2 statistic, we will determine whether a meta-analysis is feasible. In this case, we will check the statistical model to be used to gather the study-specific estimates, i.e., fixed-effects model or random-effects model analysis.[37–43] For data analysis, the calculation of pooled effect estimates will consider a CI = 95% and α = 0.05 using EPPI-Reviewer™ Software (UK). We will also assess publication bias if sufficient studies are identified per endpoint analyzed.[44] In addition, we will rate the certainty of the evidence based on the Cochrane methods and according to Grading of Recommendations Assessment, Development and Evaluation.[45] The evaluation of the quality of evidence will be performed independently and paired by 2 reviewers (LCLJ and EB). Disagreements will be handled by a 3rd reviewer (RAGL). All steps of this review will be conducted in EPPI-Reviewer™ Software (UK). The study results will be presented in PRISMA 2020[46] flowchart (Fig. 1), tables or graphs in the same way as the summaries are reported, in order to facilitate comparison of similarities and differences in the different study designs and outcomes between studies. The results will be presented and stratified in the subgroup analysis according to the health system models and in line with the income classification of the countries (high, medium-high, medium-low, and low) based on The World Bank Classification using the Gross National In-come per capita.[47] PRISMA flow diagram. Heterogeneity among studies will be measured by the I2 statistic to estimate the percentage of variation among studies, where I2 = 0% to 40% indicates low heterogeneity; I2 = 30% to 60% moderate heterogeneity; I2 = 50% to 90% substantial heterogeneity; and I2 = 75% to 100% high heterogeneity.[35,36] According to the I2 statistic, we will determine whether a meta-analysis is feasible. In this case, we will check the statistical model to be used to gather the study-specific estimates, i.e., fixed-effects model or random-effects model analysis.[37–43] For data analysis, the calculation of pooled effect estimates will consider a CI = 95% and α = 0.05 using EPPI-Reviewer™ Software (UK). We will also assess publication bias if sufficient studies are identified per endpoint analyzed.[44] In addition, we will rate the certainty of the evidence based on the Cochrane methods and according to Grading of Recommendations Assessment, Development and Evaluation.[45] The evaluation of the quality of evidence will be performed independently and paired by 2 reviewers (LCLJ and EB). Disagreements will be handled by a 3rd reviewer (RAGL). All steps of this review will be conducted in EPPI-Reviewer™ Software (UK). The study results will be presented in PRISMA 2020[46] flowchart (Fig. 1), tables or graphs in the same way as the summaries are reported, in order to facilitate comparison of similarities and differences in the different study designs and outcomes between studies. The results will be presented and stratified in the subgroup analysis according to the health system models and in line with the income classification of the countries (high, medium-high, medium-low, and low) based on The World Bank Classification using the Gross National In-come per capita.[47] PRISMA flow diagram. [SUBTITLE] 2.8. Patient and public involvement [SUBSECTION] This study protocol analyses existing research studies, and therefore involves no patients or members of the public. This study protocol analyses existing research studies, and therefore involves no patients or members of the public. [SUBTITLE] 2.9. Ethics and dissemination [SUBSECTION] This study involves neither human participants nor unpublished primary data. As such, ethics approval from a human research ethics committee is not required. Plans for the dissemination of this study comprise peer-reviewed publication and conference presentations. This study involves neither human participants nor unpublished primary data. As such, ethics approval from a human research ethics committee is not required. Plans for the dissemination of this study comprise peer-reviewed publication and conference presentations.
null
null
null
null
[ "2.1. Study design", "2.2. Research question", "2.3. Search strategy", "2.4. Eligibility", "2.5. Data extraction", "2.6. Methodological evaluation", "2.7. Data analysis and evidence synthesis", "2.9. Ethics and dissemination", "Author contributions" ]
[ "This is a systematic review and meta-analysis protocol guided by the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols.[20] To ensure the reliability of the data and methodological transparency of this review, the protocol was submitted to the International Prospective Register of Systematic Reviews (NHS) for registration (Registration Number: CRD42022329687).", "In order to formulate the research question, the PECOS strategy was used[21] (P - Population or Patients; E - Exposure; C - Comparison; O - outcomes; S - study design), where P = Population = evidence of complete economic evaluations in pediatric patients, E = home care services, C = Comparison = hospital care (in-hospital services), O = Outcomes = mortality, readmission rates, patient and caregiver satisfaction, and cost; S = Study type: cost-effectiveness, cost-utility and cost-effectiveness evaluation studies.\nIn this study our population was stated in line with the World Health Organization definition, that is, “child” as a person under 19 years of age, an “adolescent” as a person aged 10 to 19 years, an “infant” as a person aged 0 to 11 months, and a “newborn” as a person aged 0 to 28 days.[22] The acronym PECOS guided the structuring of the formulation of the research question: “What is the cost-effectiveness of home care compared to hospital care for pediatric patients in the world?\"", "The search for studies will be conducted systematically in ten electronic data-bases: Medical Literature Analysis and Retrieval System Online (MEDLINE) via PubMed, Excerpta Medica database, Cochrane Library, Web of Science, SCOPUS, Science Direct, Latin American and Caribbean Health Sciences Literature, Psychology Information (PsycINFO), cummulative index to nursing and allied health literature (CINAHL), and the Chinese National Knowledge Infrastructure. The strategy for seeking the studies will consist of a combination of controlled descriptors (indexers in the respective databases), synonyms, and keywords, as indicated in each electronic database. Thus, to search for articles in MEDLINE, we will use the Medical Subject Headings (MeSH) as controlled descriptors; the Emtree terms will be consulted in excerpta medica database; the PsycINFO Thesaurus will be consulted for the PsycINFO database; the DeCS (Health Sciences Descriptors) in the Latin American and Caribbean Health Sciences Literature database, and the CINAHL headings in the CINAHL database. It should be noted that there will be no date or language restriction in the search strategy to be performed. In addition to the electronic databases cited above, searches will be held on Clinical Trial Registry sites such as ClinicalTrials. gov (National Institutes of Health, NIH); World Health Organization International Clinical Trials Registry Platform, and the Brazilian Registry of Clinical Trials, as well as additional searches in sites of organizations and websites search, such as: The British Library, SciELO, Google Scholar, ProQuest Dissertations and Theses Global, Public Health Gray Literature Sources and Health Evidence and in the preprints for Health Sciences (medRxiv). Additionally, the list of final references from the included primary studies will be reviewed manually to find relevant studies to be added.\nThe search strategy will be carried out by 2 researchers independently (LCLJ and RMP) according to the recommendations of the Cochrane Handbook.[23] Initially, the existence of an index of specific subject headings in each database (such as MeSH terms, Emtree terms, PsycINFO Thesaurus, DeCS and CINAHL headings and their synonyms and keywords will be identified. Subsequently, the search terms will be combined using the Boolean operators “AND” and “OR”[24,25] in order to obtain restrictive and additive combinations, respectively. In addition, the search will be performed using the identified descriptors and with expanded meaning, without the use of database filters to preserve significant samples and ensure lower risk of loss. The search strategy combining the MeSH controlled descriptors and keywords that will be used in MEDLINE will be adjusted to the other electronic databases as described in Table 1.\nPreliminary search strategy in the MEDLINE via PubMed.\nMEDLINE = medical literature analysis and retrieval system online.\nIn this search strategy phase the EndNote™ reference manager will be used to store, organize and delete duplicates to ensure a systematic, comprehensive and manageable search.", "A summary of the inclusion and exclusion criteria of this systematic review is depicted in Table 2 in line with the acronym PECOS.\nInclusion and exclusion criteria.\nRegarding the study design, we will include analytical observational study designs and experimental studies, as well as gray literature, as recommended by the Cochrane Handbook.[23] Thus, studies that investigated epidemiological and clinical aspects on the cost-effectiveness of HCS in pediatric patients (<19 years) in relation to in-hospital services will be included in this systematic review. Studies that do not present complete economic evaluations (evidence without cost, benefit and effectiveness analysis), and studies involving adult and/or elderly patients will be excluded. The selection of studies will also be performed by 2 reviewers independently (LCLJ and RMP) and blinded. After this selection, a third reviewer (RAGL) will be responsible for analyzing and deciding on the inclusion or exclusion of each article, especially those containing conflicting decisions. In this step of inclusion and exclusion of articles that will compose the final sample, the Rayyan™-Qatar CRI,[26] as a tool to assist in the eligibility/selection of articles.", "First, the screening of the studies will be based on the information contained in their titles and abstracts and will be performed by the same 2 researchers (LCLJ and RMP). When the reviewers disagree, the article will be evaluated, and if the disagreement persists, a third reviewer (RAGL) will make a final decision. Once consensus is reached on the selected studies, a specific standardized form for extracting previously published full economic evaluation studies will be used.[6,27] The form is divided into 6 sections, according to the types of information made available by the studies: General information about the selected studies; Information about the study design, the population included and the comparators used; Information about the details of the economic model, time horizon and assumptions used and about the sensitivity analyses performed; Information about the costs; Sources of the data used in the study: epidemiological, intervention, effectiveness, cost and utility; Outcomes assessed in the studies and their measures and details of the sensitivity analysis.[27]", "The assessment of the methodological quality of the studies will be defined as an essential process to establish internal validity, checking for possible biases and reliability of the evidence identified. Initially, the level of evidence will be identified and classified according to the scale developed by the Oxford Centre for Evidence-Based Medicine that is based on study design and classifies them into 1A, 1B, 1C, 2A, 2B, 2C, 3A, 3B, 4, and 5 (Table 3).[28]\nClassification and hierarchy of evidence.\nRCT = randomized controlled trials.\n*From the Centre for Evidence-Based Medicine, http://www.cebm.net\nSubsequently, the checklist for assessing the quality of reporting of economic evaluation studies proposed by The British Medical Journal will be applied[29] that allows the evaluation of the items of an economic evaluation. This checklist is divided into 3 blocks of questions: study design; data collection; and analysis and interpretation of results. The internal validity and risk of bias of randomized controlled trials will be assessed using the Cochrane Risk-Of-Bias tool revised for randomized controlled trials.[30] To assess the risk of bias in quasi-experimental studies, we will use the Risk of Bias In Non-randomized Studies of Interventions.[31] In addition, the Newcastle-Ottawa Scale[32] will be used to assess the internal validity and risk of bias of cohort studies. Cross-sectional studies will be evaluated using the Agency for Healthcare Research and Quality tool.[33] The same 2 reviewers (LCLJ and RMP) will perform the critical appraisal independently. To assess the methodological quality of the comparative effectiveness observational research evidence we will use the Good Research for Comparative Effectiveness Checklist v5.0.[34]", "Heterogeneity among studies will be measured by the I2 statistic to estimate the percentage of variation among studies, where I2 = 0% to 40% indicates low heterogeneity; I2 = 30% to 60% moderate heterogeneity; I2 = 50% to 90% substantial heterogeneity; and I2 = 75% to 100% high heterogeneity.[35,36] According to the I2 statistic, we will determine whether a meta-analysis is feasible. In this case, we will check the statistical model to be used to gather the study-specific estimates, i.e., fixed-effects model or random-effects model analysis.[37–43] For data analysis, the calculation of pooled effect estimates will consider a CI = 95% and α = 0.05 using EPPI-Reviewer™ Software (UK). We will also assess publication bias if sufficient studies are identified per endpoint analyzed.[44] In addition, we will rate the certainty of the evidence based on the Cochrane methods and according to Grading of Recommendations Assessment, Development and Evaluation.[45] The evaluation of the quality of evidence will be performed independently and paired by 2 reviewers (LCLJ and EB). Disagreements will be handled by a 3rd reviewer (RAGL). All steps of this review will be conducted in EPPI-Reviewer™ Software (UK).\nThe study results will be presented in PRISMA 2020[46] flowchart (Fig. 1), tables or graphs in the same way as the summaries are reported, in order to facilitate comparison of similarities and differences in the different study designs and outcomes between studies. The results will be presented and stratified in the subgroup analysis according to the health system models and in line with the income classification of the countries (high, medium-high, medium-low, and low) based on The World Bank Classification using the Gross National In-come per capita.[47]\nPRISMA flow diagram.", "This study involves neither human participants nor unpublished primary data. As such, ethics approval from a human research ethics committee is not required. Plans for the dissemination of this study comprise peer-reviewed publication and conference presentations.", "LCLJ conceived the idea and planned and designed the study protocol. LCLJ wrote the first draft. LCLJ, RMP, EB and RAGL planned the data extraction and statistical analysis. LCLJ and RAGL provided critical insights. RMP and EB critically reviewed and modified the manuscript. All authors have reviewed and approved the manuscript. LCLJ is responsible for the overall content as guarantor.\nConceptualization: Luís Carlos Lopes-Júnior.\nData curation: Luís Carlos Lopes-Júnior, Emiliana Bomfim, Regina Aparecida Garcia de Lima.\nFormal analysis: Luís Carlos Lopes-Júnior, Raphael Manhães Pessanha, Emiliana Bomfim, Regina Aparecida Garcia de Lima.\nFunding acquisition: Luís Carlos Lopes-Júnior.\nInvestigation: Luís Carlos Lopes-Júnior.\nMethodology: Luís Carlos Lopes-Júnior.\nProject administration: Luís Carlos Lopes-Júnior.\nResources: Luís Carlos Lopes-Júnior.\nSoftware: Luís Carlos Lopes-Júnior.\nSupervision: Luís Carlos Lopes-Júnior.\nValidation: Luís Carlos Lopes-Júnior, Raphael Manhães Pessanha, Emiliana Bomfim, Regina Aparecida Garcia de Lima.\nVisualization: Luís Carlos Lopes-Júnior, Raphael Manhães Pessanha, Emiliana Bomfim, Regina Aparecida Garcia de Lima.\nWriting – original draft: Luís Carlos Lopes-Júnior, Raphael Manhães Pessanha, Emiliana Bomfim, Regina Aparecida Garcia de Lima.\nWriting – review & editing: Luís Carlos Lopes-Júnior, Raphael Manhães Pessanha, Emiliana Bomfim, Regina Aparecida Garcia de Lima." ]
[ null, null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Methods", "2.1. Study design", "2.2. Research question", "2.3. Search strategy", "2.4. Eligibility", "2.5. Data extraction", "2.6. Methodological evaluation", "2.7. Data analysis and evidence synthesis", "2.8. Patient and public involvement", "2.9. Ethics and dissemination", "3. Discussion", "Author contributions" ]
[ "Worldwide, Synchronously, the changes in the demographic and epidemiological transition that are taking place in most countries regardless of income level, pointing to a situation of triple burden of disease with the hegemonic presence of chronic conditions,[1] the need to adapt the health care model has emerged, leading many countries to think of Home Care Services (HCS) as a strategic point of care for health care.[2] The increase of HCS in several countries follows, in parallel, the interest of health systems in the process of de-hospitalization, rationalization of the use of hospital beds, cost reduction and organization of patient-centered care.\nThe demand for HCS appears, therefore, as another challenge for health systems, contributing to change the focus of care and the environment in which care is provided.[2,3] In addition, continued care in the home environment is accompanied by other equally relevant health needs, such as the aging population, as well as the care provided to premature babies, to children with special needs and chronic diseases, to adults with multiple chronic-degenerative diseases, to individuals in palliative care, life support and rehabilitation.\nThus, the relevance of the need to implement HCS stands out in the current and future health agenda of all health systems, aiming to contribute to the configuration of substitutive health networks and the transformation of health practice.[2–4] Researchers already question the real need for hospitalization for certain health problems, considering some reasons for hospitalization as dispensable or unnecessarily prolonged, and that they can be replaced or complemented by an HCS.[2–5] In addition to cost reduction, the HCS has been representing in the world scenario the connotation of offering quality care, providing welfare and comfort by allowing the patient to stay in their home environment, integrated into their life context.[5]\nIt is important to emphasize that health financing is a topic that always raises debates in order to better define the use and allocation of resources for society as a whole. Most countries face increasing health care costs, both in absolute and relative terms, regardless of whether the financing model adopted is public, private, based on tax collection, or through direct user fees. The growth of health expenditures, with the need to seek efficiency in resource allocation, has occupied an important role in public policy discussions.[6]\nEconomic evaluation studies, such as those of cost-effectiveness, are adopted in order to consider the cost factor in decision-making regarding the new technologies or models of health care, since financial resources are scarce and finite. In the evaluation of health care models, cost-effectiveness analysis is the most suitable method to compare 2 or more alternatives because it allows the combined analysis of clinical benefits and associated costs, providing objective and explicit data for decision making.[6] Cost-effectiveness studies can be understood as a tool to analyze the value of health interventions, since the method seeks to fill a gap between preferences and science. On one hand, there is the subjectivity of the preference that an individual or society has when faced with 2 options. On the other, there is the objectivity and the reproducibility of science, considering that the cost of a new technology or model of care needs to be manage.[7] In cost-effectiveness evaluation, costs are confronted with clinical outcomes in an attempt to understand the impact of different alternatives, identifying those with better intervention effects, in general, in exchange for a lower cost to health systems.[6–8]\nIt is known that several countries are in the process of adapting to this new demand for care, due especially to issues of economic viability.[2,3] This adaptation has strengthened the emergence of new strategies and mechanisms for health care, such as HCS, which combines technological and scientific resources present in the hospital with the family environment.[9] The home has emerged, added to its humanizing characteristic and the demographic and epidemiological profile of the population, as a place with potential to expand and qualify the care processes.[10] Moreover, the development of HCS on the world scene has been following demographic and epidemiological changes[11] and is related to reducing the risk of infections[12] the humanization of care and quality of life, greater involvement of family members with the patient’s illness, closer relationship between the health team, patient and family,[13] cost reduction, increased hospital bed turnover with bed management, de-hospitalization[14] lower rates of clinical worsening and acute complications, less demand for urgency and emergency services, and lower readmission rates,[15] implementation of palliative care[3] and effective actions for prevention, promotion and recovery of health.[4]\nA systematic review and meta-analysis aimed at evaluating the effect of “hospital at home” services in adults >16 years that significantly replace inpatient time on health outcomes, showed that hospital at home is associated with reductions in mortality, re-admission rates and costs, and increases in patients and caregiver satisfaction, but no change in caregiver burden.[16] Despite the expansion of HCS in several countries, there is still a need to systematically investigate the available evidence on the cost-effectiveness of this type of service in relation to hospital care in the world, particularly for the pediatric population.\nPrevious systematic review published in 2012 on the costs and effectiveness of pediatric home care pointed out that it can provide equivalent clinical outcomes for children and does not impose a greater burden on families.[17] In fact, in some cases, there is evidence of reduced burden and costs to families compared to hospital care. There is also growing evidence, although based on weaker evidence, that pediatric home care can reduce health care costs, particularly for children with complex and long-term needs.[18] It is noteworthy that this review was limited to the period from 1990 to 2007, and used simple methodological assessment tools available at the time.\nSince there is an increasing use of decision analysis models in economic evaluations of technology-assisted care models, the use of specific methodological guidelines with the main aspects of modeling is necessary,[18] since they can indicate how credible and relevant the result obtained is to inform decision makers. In this sense, systematic reviews of economic evaluations are important to synthesize the best evidence already available, in order to contribute to the formulation of guidelines for a new analysis, especially taking into account the most recent period (until 2022), in a context of contemporary transformations in health systems, and to identify the most relevant evaluations to inform a particular issue.[19] Hence, the aim of this study is to systematically synthesize and critically evaluate the evidence on cost-effectiveness of home care versus in-hospital services in pediatric patients worldwide.", "[SUBTITLE] 2.1. Study design [SUBSECTION] This is a systematic review and meta-analysis protocol guided by the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols.[20] To ensure the reliability of the data and methodological transparency of this review, the protocol was submitted to the International Prospective Register of Systematic Reviews (NHS) for registration (Registration Number: CRD42022329687).\nThis is a systematic review and meta-analysis protocol guided by the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols.[20] To ensure the reliability of the data and methodological transparency of this review, the protocol was submitted to the International Prospective Register of Systematic Reviews (NHS) for registration (Registration Number: CRD42022329687).\n[SUBTITLE] 2.2. Research question [SUBSECTION] In order to formulate the research question, the PECOS strategy was used[21] (P - Population or Patients; E - Exposure; C - Comparison; O - outcomes; S - study design), where P = Population = evidence of complete economic evaluations in pediatric patients, E = home care services, C = Comparison = hospital care (in-hospital services), O = Outcomes = mortality, readmission rates, patient and caregiver satisfaction, and cost; S = Study type: cost-effectiveness, cost-utility and cost-effectiveness evaluation studies.\nIn this study our population was stated in line with the World Health Organization definition, that is, “child” as a person under 19 years of age, an “adolescent” as a person aged 10 to 19 years, an “infant” as a person aged 0 to 11 months, and a “newborn” as a person aged 0 to 28 days.[22] The acronym PECOS guided the structuring of the formulation of the research question: “What is the cost-effectiveness of home care compared to hospital care for pediatric patients in the world?\"\nIn order to formulate the research question, the PECOS strategy was used[21] (P - Population or Patients; E - Exposure; C - Comparison; O - outcomes; S - study design), where P = Population = evidence of complete economic evaluations in pediatric patients, E = home care services, C = Comparison = hospital care (in-hospital services), O = Outcomes = mortality, readmission rates, patient and caregiver satisfaction, and cost; S = Study type: cost-effectiveness, cost-utility and cost-effectiveness evaluation studies.\nIn this study our population was stated in line with the World Health Organization definition, that is, “child” as a person under 19 years of age, an “adolescent” as a person aged 10 to 19 years, an “infant” as a person aged 0 to 11 months, and a “newborn” as a person aged 0 to 28 days.[22] The acronym PECOS guided the structuring of the formulation of the research question: “What is the cost-effectiveness of home care compared to hospital care for pediatric patients in the world?\"\n[SUBTITLE] 2.3. Search strategy [SUBSECTION] The search for studies will be conducted systematically in ten electronic data-bases: Medical Literature Analysis and Retrieval System Online (MEDLINE) via PubMed, Excerpta Medica database, Cochrane Library, Web of Science, SCOPUS, Science Direct, Latin American and Caribbean Health Sciences Literature, Psychology Information (PsycINFO), cummulative index to nursing and allied health literature (CINAHL), and the Chinese National Knowledge Infrastructure. The strategy for seeking the studies will consist of a combination of controlled descriptors (indexers in the respective databases), synonyms, and keywords, as indicated in each electronic database. Thus, to search for articles in MEDLINE, we will use the Medical Subject Headings (MeSH) as controlled descriptors; the Emtree terms will be consulted in excerpta medica database; the PsycINFO Thesaurus will be consulted for the PsycINFO database; the DeCS (Health Sciences Descriptors) in the Latin American and Caribbean Health Sciences Literature database, and the CINAHL headings in the CINAHL database. It should be noted that there will be no date or language restriction in the search strategy to be performed. In addition to the electronic databases cited above, searches will be held on Clinical Trial Registry sites such as ClinicalTrials. gov (National Institutes of Health, NIH); World Health Organization International Clinical Trials Registry Platform, and the Brazilian Registry of Clinical Trials, as well as additional searches in sites of organizations and websites search, such as: The British Library, SciELO, Google Scholar, ProQuest Dissertations and Theses Global, Public Health Gray Literature Sources and Health Evidence and in the preprints for Health Sciences (medRxiv). Additionally, the list of final references from the included primary studies will be reviewed manually to find relevant studies to be added.\nThe search strategy will be carried out by 2 researchers independently (LCLJ and RMP) according to the recommendations of the Cochrane Handbook.[23] Initially, the existence of an index of specific subject headings in each database (such as MeSH terms, Emtree terms, PsycINFO Thesaurus, DeCS and CINAHL headings and their synonyms and keywords will be identified. Subsequently, the search terms will be combined using the Boolean operators “AND” and “OR”[24,25] in order to obtain restrictive and additive combinations, respectively. In addition, the search will be performed using the identified descriptors and with expanded meaning, without the use of database filters to preserve significant samples and ensure lower risk of loss. The search strategy combining the MeSH controlled descriptors and keywords that will be used in MEDLINE will be adjusted to the other electronic databases as described in Table 1.\nPreliminary search strategy in the MEDLINE via PubMed.\nMEDLINE = medical literature analysis and retrieval system online.\nIn this search strategy phase the EndNote™ reference manager will be used to store, organize and delete duplicates to ensure a systematic, comprehensive and manageable search.\nThe search for studies will be conducted systematically in ten electronic data-bases: Medical Literature Analysis and Retrieval System Online (MEDLINE) via PubMed, Excerpta Medica database, Cochrane Library, Web of Science, SCOPUS, Science Direct, Latin American and Caribbean Health Sciences Literature, Psychology Information (PsycINFO), cummulative index to nursing and allied health literature (CINAHL), and the Chinese National Knowledge Infrastructure. The strategy for seeking the studies will consist of a combination of controlled descriptors (indexers in the respective databases), synonyms, and keywords, as indicated in each electronic database. Thus, to search for articles in MEDLINE, we will use the Medical Subject Headings (MeSH) as controlled descriptors; the Emtree terms will be consulted in excerpta medica database; the PsycINFO Thesaurus will be consulted for the PsycINFO database; the DeCS (Health Sciences Descriptors) in the Latin American and Caribbean Health Sciences Literature database, and the CINAHL headings in the CINAHL database. It should be noted that there will be no date or language restriction in the search strategy to be performed. In addition to the electronic databases cited above, searches will be held on Clinical Trial Registry sites such as ClinicalTrials. gov (National Institutes of Health, NIH); World Health Organization International Clinical Trials Registry Platform, and the Brazilian Registry of Clinical Trials, as well as additional searches in sites of organizations and websites search, such as: The British Library, SciELO, Google Scholar, ProQuest Dissertations and Theses Global, Public Health Gray Literature Sources and Health Evidence and in the preprints for Health Sciences (medRxiv). Additionally, the list of final references from the included primary studies will be reviewed manually to find relevant studies to be added.\nThe search strategy will be carried out by 2 researchers independently (LCLJ and RMP) according to the recommendations of the Cochrane Handbook.[23] Initially, the existence of an index of specific subject headings in each database (such as MeSH terms, Emtree terms, PsycINFO Thesaurus, DeCS and CINAHL headings and their synonyms and keywords will be identified. Subsequently, the search terms will be combined using the Boolean operators “AND” and “OR”[24,25] in order to obtain restrictive and additive combinations, respectively. In addition, the search will be performed using the identified descriptors and with expanded meaning, without the use of database filters to preserve significant samples and ensure lower risk of loss. The search strategy combining the MeSH controlled descriptors and keywords that will be used in MEDLINE will be adjusted to the other electronic databases as described in Table 1.\nPreliminary search strategy in the MEDLINE via PubMed.\nMEDLINE = medical literature analysis and retrieval system online.\nIn this search strategy phase the EndNote™ reference manager will be used to store, organize and delete duplicates to ensure a systematic, comprehensive and manageable search.\n[SUBTITLE] 2.4. Eligibility [SUBSECTION] A summary of the inclusion and exclusion criteria of this systematic review is depicted in Table 2 in line with the acronym PECOS.\nInclusion and exclusion criteria.\nRegarding the study design, we will include analytical observational study designs and experimental studies, as well as gray literature, as recommended by the Cochrane Handbook.[23] Thus, studies that investigated epidemiological and clinical aspects on the cost-effectiveness of HCS in pediatric patients (<19 years) in relation to in-hospital services will be included in this systematic review. Studies that do not present complete economic evaluations (evidence without cost, benefit and effectiveness analysis), and studies involving adult and/or elderly patients will be excluded. The selection of studies will also be performed by 2 reviewers independently (LCLJ and RMP) and blinded. After this selection, a third reviewer (RAGL) will be responsible for analyzing and deciding on the inclusion or exclusion of each article, especially those containing conflicting decisions. In this step of inclusion and exclusion of articles that will compose the final sample, the Rayyan™-Qatar CRI,[26] as a tool to assist in the eligibility/selection of articles.\nA summary of the inclusion and exclusion criteria of this systematic review is depicted in Table 2 in line with the acronym PECOS.\nInclusion and exclusion criteria.\nRegarding the study design, we will include analytical observational study designs and experimental studies, as well as gray literature, as recommended by the Cochrane Handbook.[23] Thus, studies that investigated epidemiological and clinical aspects on the cost-effectiveness of HCS in pediatric patients (<19 years) in relation to in-hospital services will be included in this systematic review. Studies that do not present complete economic evaluations (evidence without cost, benefit and effectiveness analysis), and studies involving adult and/or elderly patients will be excluded. The selection of studies will also be performed by 2 reviewers independently (LCLJ and RMP) and blinded. After this selection, a third reviewer (RAGL) will be responsible for analyzing and deciding on the inclusion or exclusion of each article, especially those containing conflicting decisions. In this step of inclusion and exclusion of articles that will compose the final sample, the Rayyan™-Qatar CRI,[26] as a tool to assist in the eligibility/selection of articles.\n[SUBTITLE] 2.5. Data extraction [SUBSECTION] First, the screening of the studies will be based on the information contained in their titles and abstracts and will be performed by the same 2 researchers (LCLJ and RMP). When the reviewers disagree, the article will be evaluated, and if the disagreement persists, a third reviewer (RAGL) will make a final decision. Once consensus is reached on the selected studies, a specific standardized form for extracting previously published full economic evaluation studies will be used.[6,27] The form is divided into 6 sections, according to the types of information made available by the studies: General information about the selected studies; Information about the study design, the population included and the comparators used; Information about the details of the economic model, time horizon and assumptions used and about the sensitivity analyses performed; Information about the costs; Sources of the data used in the study: epidemiological, intervention, effectiveness, cost and utility; Outcomes assessed in the studies and their measures and details of the sensitivity analysis.[27]\nFirst, the screening of the studies will be based on the information contained in their titles and abstracts and will be performed by the same 2 researchers (LCLJ and RMP). When the reviewers disagree, the article will be evaluated, and if the disagreement persists, a third reviewer (RAGL) will make a final decision. Once consensus is reached on the selected studies, a specific standardized form for extracting previously published full economic evaluation studies will be used.[6,27] The form is divided into 6 sections, according to the types of information made available by the studies: General information about the selected studies; Information about the study design, the population included and the comparators used; Information about the details of the economic model, time horizon and assumptions used and about the sensitivity analyses performed; Information about the costs; Sources of the data used in the study: epidemiological, intervention, effectiveness, cost and utility; Outcomes assessed in the studies and their measures and details of the sensitivity analysis.[27]\n[SUBTITLE] 2.6. Methodological evaluation [SUBSECTION] The assessment of the methodological quality of the studies will be defined as an essential process to establish internal validity, checking for possible biases and reliability of the evidence identified. Initially, the level of evidence will be identified and classified according to the scale developed by the Oxford Centre for Evidence-Based Medicine that is based on study design and classifies them into 1A, 1B, 1C, 2A, 2B, 2C, 3A, 3B, 4, and 5 (Table 3).[28]\nClassification and hierarchy of evidence.\nRCT = randomized controlled trials.\n*From the Centre for Evidence-Based Medicine, http://www.cebm.net\nSubsequently, the checklist for assessing the quality of reporting of economic evaluation studies proposed by The British Medical Journal will be applied[29] that allows the evaluation of the items of an economic evaluation. This checklist is divided into 3 blocks of questions: study design; data collection; and analysis and interpretation of results. The internal validity and risk of bias of randomized controlled trials will be assessed using the Cochrane Risk-Of-Bias tool revised for randomized controlled trials.[30] To assess the risk of bias in quasi-experimental studies, we will use the Risk of Bias In Non-randomized Studies of Interventions.[31] In addition, the Newcastle-Ottawa Scale[32] will be used to assess the internal validity and risk of bias of cohort studies. Cross-sectional studies will be evaluated using the Agency for Healthcare Research and Quality tool.[33] The same 2 reviewers (LCLJ and RMP) will perform the critical appraisal independently. To assess the methodological quality of the comparative effectiveness observational research evidence we will use the Good Research for Comparative Effectiveness Checklist v5.0.[34]\nThe assessment of the methodological quality of the studies will be defined as an essential process to establish internal validity, checking for possible biases and reliability of the evidence identified. Initially, the level of evidence will be identified and classified according to the scale developed by the Oxford Centre for Evidence-Based Medicine that is based on study design and classifies them into 1A, 1B, 1C, 2A, 2B, 2C, 3A, 3B, 4, and 5 (Table 3).[28]\nClassification and hierarchy of evidence.\nRCT = randomized controlled trials.\n*From the Centre for Evidence-Based Medicine, http://www.cebm.net\nSubsequently, the checklist for assessing the quality of reporting of economic evaluation studies proposed by The British Medical Journal will be applied[29] that allows the evaluation of the items of an economic evaluation. This checklist is divided into 3 blocks of questions: study design; data collection; and analysis and interpretation of results. The internal validity and risk of bias of randomized controlled trials will be assessed using the Cochrane Risk-Of-Bias tool revised for randomized controlled trials.[30] To assess the risk of bias in quasi-experimental studies, we will use the Risk of Bias In Non-randomized Studies of Interventions.[31] In addition, the Newcastle-Ottawa Scale[32] will be used to assess the internal validity and risk of bias of cohort studies. Cross-sectional studies will be evaluated using the Agency for Healthcare Research and Quality tool.[33] The same 2 reviewers (LCLJ and RMP) will perform the critical appraisal independently. To assess the methodological quality of the comparative effectiveness observational research evidence we will use the Good Research for Comparative Effectiveness Checklist v5.0.[34]\n[SUBTITLE] 2.7. Data analysis and evidence synthesis [SUBSECTION] Heterogeneity among studies will be measured by the I2 statistic to estimate the percentage of variation among studies, where I2 = 0% to 40% indicates low heterogeneity; I2 = 30% to 60% moderate heterogeneity; I2 = 50% to 90% substantial heterogeneity; and I2 = 75% to 100% high heterogeneity.[35,36] According to the I2 statistic, we will determine whether a meta-analysis is feasible. In this case, we will check the statistical model to be used to gather the study-specific estimates, i.e., fixed-effects model or random-effects model analysis.[37–43] For data analysis, the calculation of pooled effect estimates will consider a CI = 95% and α = 0.05 using EPPI-Reviewer™ Software (UK). We will also assess publication bias if sufficient studies are identified per endpoint analyzed.[44] In addition, we will rate the certainty of the evidence based on the Cochrane methods and according to Grading of Recommendations Assessment, Development and Evaluation.[45] The evaluation of the quality of evidence will be performed independently and paired by 2 reviewers (LCLJ and EB). Disagreements will be handled by a 3rd reviewer (RAGL). All steps of this review will be conducted in EPPI-Reviewer™ Software (UK).\nThe study results will be presented in PRISMA 2020[46] flowchart (Fig. 1), tables or graphs in the same way as the summaries are reported, in order to facilitate comparison of similarities and differences in the different study designs and outcomes between studies. The results will be presented and stratified in the subgroup analysis according to the health system models and in line with the income classification of the countries (high, medium-high, medium-low, and low) based on The World Bank Classification using the Gross National In-come per capita.[47]\nPRISMA flow diagram.\nHeterogeneity among studies will be measured by the I2 statistic to estimate the percentage of variation among studies, where I2 = 0% to 40% indicates low heterogeneity; I2 = 30% to 60% moderate heterogeneity; I2 = 50% to 90% substantial heterogeneity; and I2 = 75% to 100% high heterogeneity.[35,36] According to the I2 statistic, we will determine whether a meta-analysis is feasible. In this case, we will check the statistical model to be used to gather the study-specific estimates, i.e., fixed-effects model or random-effects model analysis.[37–43] For data analysis, the calculation of pooled effect estimates will consider a CI = 95% and α = 0.05 using EPPI-Reviewer™ Software (UK). We will also assess publication bias if sufficient studies are identified per endpoint analyzed.[44] In addition, we will rate the certainty of the evidence based on the Cochrane methods and according to Grading of Recommendations Assessment, Development and Evaluation.[45] The evaluation of the quality of evidence will be performed independently and paired by 2 reviewers (LCLJ and EB). Disagreements will be handled by a 3rd reviewer (RAGL). All steps of this review will be conducted in EPPI-Reviewer™ Software (UK).\nThe study results will be presented in PRISMA 2020[46] flowchart (Fig. 1), tables or graphs in the same way as the summaries are reported, in order to facilitate comparison of similarities and differences in the different study designs and outcomes between studies. The results will be presented and stratified in the subgroup analysis according to the health system models and in line with the income classification of the countries (high, medium-high, medium-low, and low) based on The World Bank Classification using the Gross National In-come per capita.[47]\nPRISMA flow diagram.\n[SUBTITLE] 2.8. Patient and public involvement [SUBSECTION] This study protocol analyses existing research studies, and therefore involves no patients or members of the public.\nThis study protocol analyses existing research studies, and therefore involves no patients or members of the public.\n[SUBTITLE] 2.9. Ethics and dissemination [SUBSECTION] This study involves neither human participants nor unpublished primary data. As such, ethics approval from a human research ethics committee is not required. Plans for the dissemination of this study comprise peer-reviewed publication and conference presentations.\nThis study involves neither human participants nor unpublished primary data. As such, ethics approval from a human research ethics committee is not required. Plans for the dissemination of this study comprise peer-reviewed publication and conference presentations.", "This is a systematic review and meta-analysis protocol guided by the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols.[20] To ensure the reliability of the data and methodological transparency of this review, the protocol was submitted to the International Prospective Register of Systematic Reviews (NHS) for registration (Registration Number: CRD42022329687).", "In order to formulate the research question, the PECOS strategy was used[21] (P - Population or Patients; E - Exposure; C - Comparison; O - outcomes; S - study design), where P = Population = evidence of complete economic evaluations in pediatric patients, E = home care services, C = Comparison = hospital care (in-hospital services), O = Outcomes = mortality, readmission rates, patient and caregiver satisfaction, and cost; S = Study type: cost-effectiveness, cost-utility and cost-effectiveness evaluation studies.\nIn this study our population was stated in line with the World Health Organization definition, that is, “child” as a person under 19 years of age, an “adolescent” as a person aged 10 to 19 years, an “infant” as a person aged 0 to 11 months, and a “newborn” as a person aged 0 to 28 days.[22] The acronym PECOS guided the structuring of the formulation of the research question: “What is the cost-effectiveness of home care compared to hospital care for pediatric patients in the world?\"", "The search for studies will be conducted systematically in ten electronic data-bases: Medical Literature Analysis and Retrieval System Online (MEDLINE) via PubMed, Excerpta Medica database, Cochrane Library, Web of Science, SCOPUS, Science Direct, Latin American and Caribbean Health Sciences Literature, Psychology Information (PsycINFO), cummulative index to nursing and allied health literature (CINAHL), and the Chinese National Knowledge Infrastructure. The strategy for seeking the studies will consist of a combination of controlled descriptors (indexers in the respective databases), synonyms, and keywords, as indicated in each electronic database. Thus, to search for articles in MEDLINE, we will use the Medical Subject Headings (MeSH) as controlled descriptors; the Emtree terms will be consulted in excerpta medica database; the PsycINFO Thesaurus will be consulted for the PsycINFO database; the DeCS (Health Sciences Descriptors) in the Latin American and Caribbean Health Sciences Literature database, and the CINAHL headings in the CINAHL database. It should be noted that there will be no date or language restriction in the search strategy to be performed. In addition to the electronic databases cited above, searches will be held on Clinical Trial Registry sites such as ClinicalTrials. gov (National Institutes of Health, NIH); World Health Organization International Clinical Trials Registry Platform, and the Brazilian Registry of Clinical Trials, as well as additional searches in sites of organizations and websites search, such as: The British Library, SciELO, Google Scholar, ProQuest Dissertations and Theses Global, Public Health Gray Literature Sources and Health Evidence and in the preprints for Health Sciences (medRxiv). Additionally, the list of final references from the included primary studies will be reviewed manually to find relevant studies to be added.\nThe search strategy will be carried out by 2 researchers independently (LCLJ and RMP) according to the recommendations of the Cochrane Handbook.[23] Initially, the existence of an index of specific subject headings in each database (such as MeSH terms, Emtree terms, PsycINFO Thesaurus, DeCS and CINAHL headings and their synonyms and keywords will be identified. Subsequently, the search terms will be combined using the Boolean operators “AND” and “OR”[24,25] in order to obtain restrictive and additive combinations, respectively. In addition, the search will be performed using the identified descriptors and with expanded meaning, without the use of database filters to preserve significant samples and ensure lower risk of loss. The search strategy combining the MeSH controlled descriptors and keywords that will be used in MEDLINE will be adjusted to the other electronic databases as described in Table 1.\nPreliminary search strategy in the MEDLINE via PubMed.\nMEDLINE = medical literature analysis and retrieval system online.\nIn this search strategy phase the EndNote™ reference manager will be used to store, organize and delete duplicates to ensure a systematic, comprehensive and manageable search.", "A summary of the inclusion and exclusion criteria of this systematic review is depicted in Table 2 in line with the acronym PECOS.\nInclusion and exclusion criteria.\nRegarding the study design, we will include analytical observational study designs and experimental studies, as well as gray literature, as recommended by the Cochrane Handbook.[23] Thus, studies that investigated epidemiological and clinical aspects on the cost-effectiveness of HCS in pediatric patients (<19 years) in relation to in-hospital services will be included in this systematic review. Studies that do not present complete economic evaluations (evidence without cost, benefit and effectiveness analysis), and studies involving adult and/or elderly patients will be excluded. The selection of studies will also be performed by 2 reviewers independently (LCLJ and RMP) and blinded. After this selection, a third reviewer (RAGL) will be responsible for analyzing and deciding on the inclusion or exclusion of each article, especially those containing conflicting decisions. In this step of inclusion and exclusion of articles that will compose the final sample, the Rayyan™-Qatar CRI,[26] as a tool to assist in the eligibility/selection of articles.", "First, the screening of the studies will be based on the information contained in their titles and abstracts and will be performed by the same 2 researchers (LCLJ and RMP). When the reviewers disagree, the article will be evaluated, and if the disagreement persists, a third reviewer (RAGL) will make a final decision. Once consensus is reached on the selected studies, a specific standardized form for extracting previously published full economic evaluation studies will be used.[6,27] The form is divided into 6 sections, according to the types of information made available by the studies: General information about the selected studies; Information about the study design, the population included and the comparators used; Information about the details of the economic model, time horizon and assumptions used and about the sensitivity analyses performed; Information about the costs; Sources of the data used in the study: epidemiological, intervention, effectiveness, cost and utility; Outcomes assessed in the studies and their measures and details of the sensitivity analysis.[27]", "The assessment of the methodological quality of the studies will be defined as an essential process to establish internal validity, checking for possible biases and reliability of the evidence identified. Initially, the level of evidence will be identified and classified according to the scale developed by the Oxford Centre for Evidence-Based Medicine that is based on study design and classifies them into 1A, 1B, 1C, 2A, 2B, 2C, 3A, 3B, 4, and 5 (Table 3).[28]\nClassification and hierarchy of evidence.\nRCT = randomized controlled trials.\n*From the Centre for Evidence-Based Medicine, http://www.cebm.net\nSubsequently, the checklist for assessing the quality of reporting of economic evaluation studies proposed by The British Medical Journal will be applied[29] that allows the evaluation of the items of an economic evaluation. This checklist is divided into 3 blocks of questions: study design; data collection; and analysis and interpretation of results. The internal validity and risk of bias of randomized controlled trials will be assessed using the Cochrane Risk-Of-Bias tool revised for randomized controlled trials.[30] To assess the risk of bias in quasi-experimental studies, we will use the Risk of Bias In Non-randomized Studies of Interventions.[31] In addition, the Newcastle-Ottawa Scale[32] will be used to assess the internal validity and risk of bias of cohort studies. Cross-sectional studies will be evaluated using the Agency for Healthcare Research and Quality tool.[33] The same 2 reviewers (LCLJ and RMP) will perform the critical appraisal independently. To assess the methodological quality of the comparative effectiveness observational research evidence we will use the Good Research for Comparative Effectiveness Checklist v5.0.[34]", "Heterogeneity among studies will be measured by the I2 statistic to estimate the percentage of variation among studies, where I2 = 0% to 40% indicates low heterogeneity; I2 = 30% to 60% moderate heterogeneity; I2 = 50% to 90% substantial heterogeneity; and I2 = 75% to 100% high heterogeneity.[35,36] According to the I2 statistic, we will determine whether a meta-analysis is feasible. In this case, we will check the statistical model to be used to gather the study-specific estimates, i.e., fixed-effects model or random-effects model analysis.[37–43] For data analysis, the calculation of pooled effect estimates will consider a CI = 95% and α = 0.05 using EPPI-Reviewer™ Software (UK). We will also assess publication bias if sufficient studies are identified per endpoint analyzed.[44] In addition, we will rate the certainty of the evidence based on the Cochrane methods and according to Grading of Recommendations Assessment, Development and Evaluation.[45] The evaluation of the quality of evidence will be performed independently and paired by 2 reviewers (LCLJ and EB). Disagreements will be handled by a 3rd reviewer (RAGL). All steps of this review will be conducted in EPPI-Reviewer™ Software (UK).\nThe study results will be presented in PRISMA 2020[46] flowchart (Fig. 1), tables or graphs in the same way as the summaries are reported, in order to facilitate comparison of similarities and differences in the different study designs and outcomes between studies. The results will be presented and stratified in the subgroup analysis according to the health system models and in line with the income classification of the countries (high, medium-high, medium-low, and low) based on The World Bank Classification using the Gross National In-come per capita.[47]\nPRISMA flow diagram.", "This study protocol analyses existing research studies, and therefore involves no patients or members of the public.", "This study involves neither human participants nor unpublished primary data. As such, ethics approval from a human research ethics committee is not required. Plans for the dissemination of this study comprise peer-reviewed publication and conference presentations.", "The results of this research will bring systematic and with high methodological rigor the available evidence on the cost-effectiveness of home care in pediatric patients compared to in-hospital services, exploring different outcomes available in the literature in the various models of health systems in the world. These results may have relevance for the basis of public health policies, regarding the forms of organization of HCS, especially in terms of complete economic evaluations through cost-effectiveness analysis in relation to hospital care. It is expected that from this product, it will be possible to obtain the necessary elements to support decision making for changes in management, practice and training of health professionals, impacting on the quality of home care.", "LCLJ conceived the idea and planned and designed the study protocol. LCLJ wrote the first draft. LCLJ, RMP, EB and RAGL planned the data extraction and statistical analysis. LCLJ and RAGL provided critical insights. RMP and EB critically reviewed and modified the manuscript. All authors have reviewed and approved the manuscript. LCLJ is responsible for the overall content as guarantor.\nConceptualization: Luís Carlos Lopes-Júnior.\nData curation: Luís Carlos Lopes-Júnior, Emiliana Bomfim, Regina Aparecida Garcia de Lima.\nFormal analysis: Luís Carlos Lopes-Júnior, Raphael Manhães Pessanha, Emiliana Bomfim, Regina Aparecida Garcia de Lima.\nFunding acquisition: Luís Carlos Lopes-Júnior.\nInvestigation: Luís Carlos Lopes-Júnior.\nMethodology: Luís Carlos Lopes-Júnior.\nProject administration: Luís Carlos Lopes-Júnior.\nResources: Luís Carlos Lopes-Júnior.\nSoftware: Luís Carlos Lopes-Júnior.\nSupervision: Luís Carlos Lopes-Júnior.\nValidation: Luís Carlos Lopes-Júnior, Raphael Manhães Pessanha, Emiliana Bomfim, Regina Aparecida Garcia de Lima.\nVisualization: Luís Carlos Lopes-Júnior, Raphael Manhães Pessanha, Emiliana Bomfim, Regina Aparecida Garcia de Lima.\nWriting – original draft: Luís Carlos Lopes-Júnior, Raphael Manhães Pessanha, Emiliana Bomfim, Regina Aparecida Garcia de Lima.\nWriting – review & editing: Luís Carlos Lopes-Júnior, Raphael Manhães Pessanha, Emiliana Bomfim, Regina Aparecida Garcia de Lima." ]
[ "intro", "methods", null, null, null, null, null, null, null, "subjects", null, "discussion", null ]
[ "cost-effectiveness", "health systems", "home care", "public health", "systematic review" ]
Potential pathogenic mechanism of type 1 X-linked lymphoproliferative syndrome caused by a mutation of SH2D1A gene in an infant: A case report.
36254040
X-linked lymphoproliferative syndrome (XLP) is a rare X-linked recessive inborn errors of immunity. The pathogenesis of XLP might be related to phophatidylinositol-3-kinase (PI3K)-associated pathways but insight details remain unclear. This study was to study an infant XLP-1 case caused by a mutation in SH2D1A gene, investigate the structural and functional alteration of mutant SAP protein, and explore the potential role of PI3K-associated pathways in the progression of XLP-1.
BACKGROUND
The proband's condition was monitored by laboratory and imagological examinations. Whole exome sequencing and Sanger sequencing were performed to detect the genetic disorder. Bioinformatics tools including PolyPhen-2, SWISS-MODEL and SWISS-PDB Viewer were used to predict the pathogenicity and estimate structural change of mutant protein. Flow cytometry was used to investigate expression of SAP and PI3K-associated proteins.
METHODS
The proband was diagnosed with XLP-1 caused by a hemizygous mutation c.96G > T in SH2D1A gene resulting in a missense substitution of Arginine to Serine at the site of amino acid 32 (p.R32S). The mutant protein contained a hydrogen bond turnover at the site of mutation and was predicted to be highly pathogenic. Expression of SH2D1A encoded protein SAP was downregulated in proband. The PI3K-AKT-mTOR signaling pathway was fully activated in XLP-1 patients, but it was inactive or only partially activated in healthy people or HLH patients.
RESULTS
The mutation c.96G > T in SH2D1A gene caused structural and functional changes in the SAP protein, resulting in XLP-1. The PI3K-AKT-mTOR signaling pathway may play a role in XLP-1 pathogenesis.
CONCLUSIONS
[ "Amino Acids", "Arginine", "Humans", "Infant", "Intracellular Signaling Peptides and Proteins", "Lymphoproliferative Disorders", "Mutant Proteins", "Mutation", "Phosphatidylinositol 3-Kinases", "Proto-Oncogene Proteins c-akt", "Serine", "Signaling Lymphocytic Activation Molecule Associated Protein", "TOR Serine-Threonine Kinases" ]
9575725
1. Introduction
X-linked lymphoproliferative syndrome (XLP) is an extremely rare X-linked recessive inborn errors of immunity caused by genetic variations, with a 1/1000000 incidence rate,[12]. XLP is currently classified into 2 forms based on mutant genes: Type 1 XLP (XLP-1) and Type 2 XLP (XLP-2) caused by mutations in SH2D1A gene and XIAP gene, respectively[3,4]. Common clinical characteristics of XLP-1 and XLP-2 including Epstein-Barr virus (EBV) infection, hemophagocytic lymphohistiocytosis (HLH), lymphoproliferative disorder, and dysgammaglobulinemia, but sometimes they also manifested differently. According to published studies, EBV infection and associated HLH were more frequently observed in patients with XLP-1, and sometimes even accompanied by catastrophic neurologic disorders. Development of lymphoma is only reported in XLP-1 patients[5]. Moreover, Natural killer cells (NK) and T lymphocytes are more likely to proliferate and infiltrate organs, including liver, spleen, and lymph nodes, in EBV-infected XLP-1 patients[6]. Persistent hypogammaglobulinemia appeared more often in XLP-1 patients while transient hypogammaglobulinemia was more frequently observed in XLP-2[7]. Inflammatory bowel disease, such as Crohn’s disease, was exclusively discovered in XLP-2 patients, and roughly 25% to 30% of XLP-2 patients were impacted[8]. Additionally, fever, subcutaneous petechia, hematemesis, hematochezia, pistaxis, jaundice, hepatosplenomegaly, lymphadenopathy, lymphocytic vasculitis, aplastic anemia, and lymphomatoid granulomatosis may also be signs and symptoms of XLP[9–11]. Due to the rapid progression and complicated symptoms of XLP, it is commonly misdiagnosed clinically. XLP has a poor prognosis and high mortality, which have been reported to be 75%, of which 70% died before the age of 10[12]. At present, the only promising treatment is allogeneic hematopoietic stem cell transplantation (HSCT) before the onset of typical symptoms or EBV infection[13]. The SH2D1A gene is found on chromosome Xq25 and has 4 exons, and mutations in this gene can result in SAP protein deficiency. SAP is a 128-amino acids signaling lymphocyte activating molecule (SLAM)-associated protein that contains 1 Src homology 2 (SH2) domain and is primarily expressed in T cells, NK cells, and some EBV-positive Burkitt lymphoma-derived B cells.[14] SAP can competitively bind to SLAMs via the SH2 domain and regulate a variety of processes, including the development and function of invariant natural killer T cells (iNKT), the clearance of EBV-infected B cells with cytotoxic T lymphocytes (CTLs) and NK cells, the development of germinal centers, the production of immunoglobulin, T cell restimulation-induced cell death, and the maintenance of T cell homeostasis.[15] In XLP-1 patients, SAP deficiency can prevent CTLs and NK cells from killing EBV-infected B cells.[16] The XIAP gene is also located on chromosome Xq25 and includes 7 exons, its encoding protein XIAP is a novel member of the inhibitor of apoptosis proteins family, which inhibits caspases directly and regulates apoptosis through several routes. XIAP gene is overexpressed in many tumor cell lines, and its expression is closely related to tumor progression, recurrence, prognosis, and treatment resistance.[17] Phophatidylinositol-3-kinase (PI3K) is a crucial signaling center in immune cells that catalyzes the conversion of PIP2 to PIP3 and thus facilitates membrane recruitment of molecules containing PIP3-binding Pleckstrin homology domains such as the AKT, PDK1, Tec family kinases, adapter molecules, guanine nucleotide exchange factors, and GTPase-activating proteins. These subsequently activate a number of important downstream pathways including mTOR, FOXO1 and BACH2-related pathways.[18] Too little or too much activity in the PI3K downstream pathway is harmful even pathogenic. Previous research discovered that SAP protein was downregulated in CTLs while the SLAM protein 2B4 was upregulated in patients with congenital PI3K deficiency disorder such as activated PI3Kδ syndrome, implying that SAP may interact with PI3K-associated pathways and XLP-1 may be related to PI3K signaling failure.[19] Insight details of the interaction patterns between SAP and PI3K, as well as the role of PI3K-associated pathways in pathogenesis of XLP-1 still need to be investigated further. In this study, we will describe an infant with XLP-1, assess the proband’s clinical features and genetic variants, analyze the structural and functional changes in SAP protein caused by gene mutation, and investigate the probable role of PI3K-associated pathways in XLP-1 pathogenesis.
2. Methods
[SUBTITLE] 2.1. Editorial policies and ethical considerations [SUBSECTION] The study was approved by the Ethics Committee of Kunming Children’s Hospital. All experiments were performed in compliance with the Helsinki Declaration. Informed written consent was obtained from the parents of the proband for the collection of clinical information, blood samples, DNA and presentation of patient’s materials. The study was approved by the Ethics Committee of Kunming Children’s Hospital. All experiments were performed in compliance with the Helsinki Declaration. Informed written consent was obtained from the parents of the proband for the collection of clinical information, blood samples, DNA and presentation of patient’s materials. [SUBTITLE] 2.2. Proband [SUBSECTION] The proband, a 7-month-old male, was admitted to the hospital due to “fever with rash.” Fever was up to 40°C and not relieved after treatment. Physical examination of the proband on admission observed listless and poor mental response, scattered rashes on the trunk and limbs particularly on both forearms and lower legs, eyelid edema, pharyngeal congestion, white pustules attached to the pharyngeal tonsils, and several palpable enlarged lymph nodes in the neck up to about 1 × 1 cm in size with medium hardness and poor range of motion. The proband was initially treated with ganciclovir, sodium succinate and intravenous IgG to against viral infection, reduce inflammation and adjust immunity, respectively. With the progression of the disease, the proband had recurring fevers and icteric sclera indicating aggravated liver damage. The proband’s liver and spleen were progressively enlarged, and the rash became more widespread and fusing into patches in some area. Staphylococcus aureus were tested positive in sputum. Cefperazone-Sulbactam, meropenem and vancomycin were successively given against infection. Bone marrow examination showed hemophagocytosis. Etoposide was then given, during the treatment, the proband experienced progressive pancytopenia and was interfered with the infusion of granulocyte colony-stimulating factor, interleukin-11, and apheresis platelets. Unfortunately, the proband’s condition kept aggravating despite accepting treatment and finally died due to multiple organ dysfunctions. Parents of the proband were healthy, and no other remarkable diseases history was identified in the family. The proband, a 7-month-old male, was admitted to the hospital due to “fever with rash.” Fever was up to 40°C and not relieved after treatment. Physical examination of the proband on admission observed listless and poor mental response, scattered rashes on the trunk and limbs particularly on both forearms and lower legs, eyelid edema, pharyngeal congestion, white pustules attached to the pharyngeal tonsils, and several palpable enlarged lymph nodes in the neck up to about 1 × 1 cm in size with medium hardness and poor range of motion. The proband was initially treated with ganciclovir, sodium succinate and intravenous IgG to against viral infection, reduce inflammation and adjust immunity, respectively. With the progression of the disease, the proband had recurring fevers and icteric sclera indicating aggravated liver damage. The proband’s liver and spleen were progressively enlarged, and the rash became more widespread and fusing into patches in some area. Staphylococcus aureus were tested positive in sputum. Cefperazone-Sulbactam, meropenem and vancomycin were successively given against infection. Bone marrow examination showed hemophagocytosis. Etoposide was then given, during the treatment, the proband experienced progressive pancytopenia and was interfered with the infusion of granulocyte colony-stimulating factor, interleukin-11, and apheresis platelets. Unfortunately, the proband’s condition kept aggravating despite accepting treatment and finally died due to multiple organ dysfunctions. Parents of the proband were healthy, and no other remarkable diseases history was identified in the family. [SUBTITLE] 2.3. Whole exome sequencing [SUBSECTION] Peripheral blood samples were collected from the proband and parents. Genomic DNA was extracted from peripheral blood using the DNA Extraction kit (CWBIO, China). The isolated DNA was quantified by agarose gel electrophoresis and Nanodrop 2000 (Thermal Fisher Scientific). Libraries were prepared using Illumina standard protocol and a minimum of 3 μg DNA was employed for the indexed Illumina libraries. To capture targeted genes, the biotinylated capture probes were designed to tile all of the exons with non-repeated regions. The resulting captured DNA was amplified by PCR and the PCR product was purified with SPRI beads (Beckman Coulter). The enriched libraries were sequenced on an Illumina NextSeq 500 sequencer for paired-end reads of 150 bp. Quality control criteria were applied to the raw sequencing data before being mapped to the UCSC hg19 human reference genome using BWA (0.7.10). Picard tools (1.119) were used to eliminate duplicated reads. SNP and InDels were detected and filtered by GATK (Genome Analysis TK-3.3.0). Variants were further annotated by ANNOVAR. All mutations identified were confirmed by Sanger sequencing. Sites of variation were identified through a comparison of DNA sequences with the corresponding GenBank (www.ncbi.nlm.nih.gov) reference sequences. Exon 1 of SH2D1A (NM_002351) were amplified using 2 pair primers (Forward: 5’-GCTCGATCGAACCAAGCTAC-3’; Reverse: 5’-GGAGCGAAGGTAAACTGTGG-3’). The PCR samples were visualized on agarose gels, purified and sequenced using the terminator cycle sequencing method on an ABI PRISM 3730 genetic analyzer (Thermo Fisher Scientific). The sequencing results were analyzed using the DNASTAR (Madison) software. Peripheral blood samples were collected from the proband and parents. Genomic DNA was extracted from peripheral blood using the DNA Extraction kit (CWBIO, China). The isolated DNA was quantified by agarose gel electrophoresis and Nanodrop 2000 (Thermal Fisher Scientific). Libraries were prepared using Illumina standard protocol and a minimum of 3 μg DNA was employed for the indexed Illumina libraries. To capture targeted genes, the biotinylated capture probes were designed to tile all of the exons with non-repeated regions. The resulting captured DNA was amplified by PCR and the PCR product was purified with SPRI beads (Beckman Coulter). The enriched libraries were sequenced on an Illumina NextSeq 500 sequencer for paired-end reads of 150 bp. Quality control criteria were applied to the raw sequencing data before being mapped to the UCSC hg19 human reference genome using BWA (0.7.10). Picard tools (1.119) were used to eliminate duplicated reads. SNP and InDels were detected and filtered by GATK (Genome Analysis TK-3.3.0). Variants were further annotated by ANNOVAR. All mutations identified were confirmed by Sanger sequencing. Sites of variation were identified through a comparison of DNA sequences with the corresponding GenBank (www.ncbi.nlm.nih.gov) reference sequences. Exon 1 of SH2D1A (NM_002351) were amplified using 2 pair primers (Forward: 5’-GCTCGATCGAACCAAGCTAC-3’; Reverse: 5’-GGAGCGAAGGTAAACTGTGG-3’). The PCR samples were visualized on agarose gels, purified and sequenced using the terminator cycle sequencing method on an ABI PRISM 3730 genetic analyzer (Thermo Fisher Scientific). The sequencing results were analyzed using the DNASTAR (Madison) software. [SUBTITLE] 2.4. Flow cytometry [SUBSECTION] Peripheral blood samples were collected from the proband and parents, as well as a familial HLH patient and a healthy child as control. Peripheral blood mononuclear cells were isolated using lymphocyte separation solution. Protein expression in peripheral blood mononuclear cells was determined via flow cytometry. Briefly, cells were fixed and permeabilized sequentially with an IntraPrep Permeabilizaton Reagent kit (Beckman Coulter,A07803) following manufacturer’s instruction. Specific antibodies were then added for incubation overnight, followed by 30 mins incubation with a fluorescently labeled secondary antibody. The flow cytometry tests were carried out with a BD FACSCanto II flow cytometer, and the data was processed with CytExpert 2.0 (Beckman Coulter) software. Antibodies used were listed as follows: Anti-SH2D1A/SAP (Abcam, ab109120); Anti-PI3 Kinase p110δ (Abcam, ab109006); Anti-PI3 Kinase p85α (Abcam, ab191606); Anti-PTEN (Abcam, ab32199); Anti-AKT (CST, 9272s); Anti-phospho-AKT (CST, 9271s); Anti-mTOR (Abcam, ab2732). Peripheral blood samples were collected from the proband and parents, as well as a familial HLH patient and a healthy child as control. Peripheral blood mononuclear cells were isolated using lymphocyte separation solution. Protein expression in peripheral blood mononuclear cells was determined via flow cytometry. Briefly, cells were fixed and permeabilized sequentially with an IntraPrep Permeabilizaton Reagent kit (Beckman Coulter,A07803) following manufacturer’s instruction. Specific antibodies were then added for incubation overnight, followed by 30 mins incubation with a fluorescently labeled secondary antibody. The flow cytometry tests were carried out with a BD FACSCanto II flow cytometer, and the data was processed with CytExpert 2.0 (Beckman Coulter) software. Antibodies used were listed as follows: Anti-SH2D1A/SAP (Abcam, ab109120); Anti-PI3 Kinase p110δ (Abcam, ab109006); Anti-PI3 Kinase p85α (Abcam, ab191606); Anti-PTEN (Abcam, ab32199); Anti-AKT (CST, 9272s); Anti-phospho-AKT (CST, 9271s); Anti-mTOR (Abcam, ab2732). [SUBTITLE] 2.5. Prediction of the pathogenicity and structure of mutant protein [SUBSECTION] Pathogenicity of mutant protein was predicted by PolyPhen-2 online scoring tool (http://genetics.bwh.harvard.edu/pph2/index.shtml). The greater the pathogenicity, the closer the score is to 1.0. The structure of wild-type protein was created by SWISS-MODEL online tool (http://swissmodel.expasy.org/) and the structure of mutant protein was estimated by SWISS-PDB Viewer 4.1.0 software. Pathogenicity of mutant protein was predicted by PolyPhen-2 online scoring tool (http://genetics.bwh.harvard.edu/pph2/index.shtml). The greater the pathogenicity, the closer the score is to 1.0. The structure of wild-type protein was created by SWISS-MODEL online tool (http://swissmodel.expasy.org/) and the structure of mutant protein was estimated by SWISS-PDB Viewer 4.1.0 software.
3. Results
[SUBTITLE] 3.1 Laboratory and imagological examinations indicated that the proband’s condition continued to deteriorate despite receiving treatment [SUBSECTION] To access detailed conditions of the proband, multiple laboratory and imagological examinations were conducted during hospitalization. Results showed that peripheral blood EBV nucleic acid load continued to rise and the antibodies to EBV capsid antigen (EBVCA) IgG and IgG antibodies to EBV nuclear antigen became positive as the disease progressed (Fig. 1A). The percentage of CD19 + B cells was slightly decreased (Fig. 1B), and humoral immunity test indicated increased levels of serum IgG, IgM, IgA, and decreased C3 (Fig. 1C). Hematological parameters of the proband pre- and during-hospitalization showed that the counts of leukocyte, lymphocyte, neutrophil, monocyte, platelet, erythrocyte and hemoglobin were declined with fluctuation and generally lower than reference values. The percentages of lymphocyte, neutrophil and monocyte were all fluctuated outside the reference value (Fig. 1D). Besides, indicators for liver, renal and cardiac function of proband were abnormal as disease progressed. The levels of alanine aminotransferase, aspartate aminotransferase, γ-glutamyltransferase (γ-GGT), alkaline phosphatase, total bilirubin, direct bilirubin, total bile acid, lactate dehydrogenase and α-hydroxybutyrate dehydrogenase (α-HBDH) were all extremely higher than reference values (Fig. 1E–G). Additionally, the levels of ferritin and triglycerides elevated dramatically indicated the development of HLH (Fig. 2A). Occurrence of HLH was also confirmed by hemophagocytes in bone marrow (Fig. 2B) and increased expression of serum IL-6 and IL-10, with IL-10 being 12 times higher than the reference values (Fig. 2C). Abdominal color ultrasound revealed hepatomegaly, gallbladder wall thickening, mild splenomegaly and abdominal effusion (Fig. 3A). Neck color ultrasound revealed slightly enlarged lymph nodes on both sides of neck and jaw (Fig. 3B). Chest X-ray revealed increased bilateral pleural effusion with deterioration of disease (Fig. 3C). Overall, with the progression of disease, the condition of the proband kept deteriorating, especially the infection was aggravated, immunity responses were deficient, and multiple organs were progressively disabled. The clinical features including EBV-infection, bone marrow hemophagocytosis, dysgammaglobulinemia, hepatosplenomegaly and lymphadenopathy observed suggested that the proband may suffer from XLP. Laboratory examinations of the proband with the progression of disease. (A) Results of peripheral blood EBV nucleic acid and antibodies test. (B) Results of lymphocyte subsets test. (C) Results of humoral immunity test. (D) Results of whole blood counts. There were ten times of testing results during hospitalization listed in chronological order from left to right. (E) Results of liver function test. There were 10 times of testing results during hospitalization listed in chronological order from left to right. (F) Results of renal function test. There were 8 times of testing results during hospitalization listed in chronological order from left to right. (G) Results of cardiac function test. There were 7 times of testing results during hospitalization listed in chronological order from left to right. EBV = Epstein-Barr virus. Occurrence of hemophagocytic lymphohistiocytosis was detected in the proband. (A) Results of ferritin and triglycerides tests. There were 4 times of testing results during hospitalization listed in chronological order from left to right. (B) Results of cytopathology detection of bone marrow. (C) Results of serum cytokine test. Imageological examinations of the proband with the progression of disease. (A) Results of abdominal color ultrasound. (B) Results of neck color ultrasound. (C) Results of chest X-ray. There were 5 times of examine results during hospitalization listed in chronological order from left to right. To access detailed conditions of the proband, multiple laboratory and imagological examinations were conducted during hospitalization. Results showed that peripheral blood EBV nucleic acid load continued to rise and the antibodies to EBV capsid antigen (EBVCA) IgG and IgG antibodies to EBV nuclear antigen became positive as the disease progressed (Fig. 1A). The percentage of CD19 + B cells was slightly decreased (Fig. 1B), and humoral immunity test indicated increased levels of serum IgG, IgM, IgA, and decreased C3 (Fig. 1C). Hematological parameters of the proband pre- and during-hospitalization showed that the counts of leukocyte, lymphocyte, neutrophil, monocyte, platelet, erythrocyte and hemoglobin were declined with fluctuation and generally lower than reference values. The percentages of lymphocyte, neutrophil and monocyte were all fluctuated outside the reference value (Fig. 1D). Besides, indicators for liver, renal and cardiac function of proband were abnormal as disease progressed. The levels of alanine aminotransferase, aspartate aminotransferase, γ-glutamyltransferase (γ-GGT), alkaline phosphatase, total bilirubin, direct bilirubin, total bile acid, lactate dehydrogenase and α-hydroxybutyrate dehydrogenase (α-HBDH) were all extremely higher than reference values (Fig. 1E–G). Additionally, the levels of ferritin and triglycerides elevated dramatically indicated the development of HLH (Fig. 2A). Occurrence of HLH was also confirmed by hemophagocytes in bone marrow (Fig. 2B) and increased expression of serum IL-6 and IL-10, with IL-10 being 12 times higher than the reference values (Fig. 2C). Abdominal color ultrasound revealed hepatomegaly, gallbladder wall thickening, mild splenomegaly and abdominal effusion (Fig. 3A). Neck color ultrasound revealed slightly enlarged lymph nodes on both sides of neck and jaw (Fig. 3B). Chest X-ray revealed increased bilateral pleural effusion with deterioration of disease (Fig. 3C). Overall, with the progression of disease, the condition of the proband kept deteriorating, especially the infection was aggravated, immunity responses were deficient, and multiple organs were progressively disabled. The clinical features including EBV-infection, bone marrow hemophagocytosis, dysgammaglobulinemia, hepatosplenomegaly and lymphadenopathy observed suggested that the proband may suffer from XLP. Laboratory examinations of the proband with the progression of disease. (A) Results of peripheral blood EBV nucleic acid and antibodies test. (B) Results of lymphocyte subsets test. (C) Results of humoral immunity test. (D) Results of whole blood counts. There were ten times of testing results during hospitalization listed in chronological order from left to right. (E) Results of liver function test. There were 10 times of testing results during hospitalization listed in chronological order from left to right. (F) Results of renal function test. There were 8 times of testing results during hospitalization listed in chronological order from left to right. (G) Results of cardiac function test. There were 7 times of testing results during hospitalization listed in chronological order from left to right. EBV = Epstein-Barr virus. Occurrence of hemophagocytic lymphohistiocytosis was detected in the proband. (A) Results of ferritin and triglycerides tests. There were 4 times of testing results during hospitalization listed in chronological order from left to right. (B) Results of cytopathology detection of bone marrow. (C) Results of serum cytokine test. Imageological examinations of the proband with the progression of disease. (A) Results of abdominal color ultrasound. (B) Results of neck color ultrasound. (C) Results of chest X-ray. There were 5 times of examine results during hospitalization listed in chronological order from left to right. [SUBTITLE] 3.2. The proband was diagnosed with XLP-1 caused by a hemizygous mutation c.96G > T in SH2D1A gene [SUBSECTION] In order to confirm whether the proband was suffered from genetic disorders, the peripheral blood DNA of the proband and parents were extracted, and whole exome sequencing was performed. The results revealed a hemizygous mutation in exon 1 of SH2D1A gene, which substituted Guanine to Thymine at the site of nucleotide 96 (c.96G > T) (Fig. 4A), resulting in a missense substitution of Arginine to Serine at the site of amino acid 32 (p.R32S). According to the ACMG guidelines, this mutation was identified as a pathogenic mutation. It was located in the mutation hotspot region with a low-frequency in the normal population database. The mutation with the same amino acid change as this proband had been reported previously but with different nucleotide change.[20] Sanger sequencing validation of this mutation was performed on the parents, and the results identified no variation at this site in the father and a heterozygous variation at this site in the mother (Fig. 4B and C). As XLP-1 inherited in a recessive pattern, the parents of the proband were all appeared healthy. Therefore, the diagnosis of XLP-1 was confirmed based on the proband’s typical clinical manifestations and genetic analysis results. Identification of a hemizygous mutation c.96G > T in SH2D1A gene of the proband. (A) Schematic diagram of SH2D1A gene. The mutation site was highlighted in red. (B) Results of Sanger sequencing. The arrows indicated the sites of mutation. (C) Genetic pedigree of the family. In order to confirm whether the proband was suffered from genetic disorders, the peripheral blood DNA of the proband and parents were extracted, and whole exome sequencing was performed. The results revealed a hemizygous mutation in exon 1 of SH2D1A gene, which substituted Guanine to Thymine at the site of nucleotide 96 (c.96G > T) (Fig. 4A), resulting in a missense substitution of Arginine to Serine at the site of amino acid 32 (p.R32S). According to the ACMG guidelines, this mutation was identified as a pathogenic mutation. It was located in the mutation hotspot region with a low-frequency in the normal population database. The mutation with the same amino acid change as this proband had been reported previously but with different nucleotide change.[20] Sanger sequencing validation of this mutation was performed on the parents, and the results identified no variation at this site in the father and a heterozygous variation at this site in the mother (Fig. 4B and C). As XLP-1 inherited in a recessive pattern, the parents of the proband were all appeared healthy. Therefore, the diagnosis of XLP-1 was confirmed based on the proband’s typical clinical manifestations and genetic analysis results. Identification of a hemizygous mutation c.96G > T in SH2D1A gene of the proband. (A) Schematic diagram of SH2D1A gene. The mutation site was highlighted in red. (B) Results of Sanger sequencing. The arrows indicated the sites of mutation. (C) Genetic pedigree of the family. [SUBTITLE] 3.3. The mutant SAP protein was highly pathogenic with structural and functional change [SUBSECTION] In order to explore whether the c.96G > T mutation in SH2D1A gene caused functional or structural deficiency in SAP protein, we first analyzed pathogenicity of mutant protein by PolyPhen-2 online scoring tool. As the greater the pathogenicity, the closer the score is to 1.0, the mutation was predicted to be highly pathogenic with a score of 1.0 (Fig. 5A). In addition, we analyzed the structural change of mutant SAP protein. As shown in Figure 5B, the secondary structure of SAP protein contained 2 α-helices and 8 β-strands and the mutation site was located at the end of the second β-strand. By comparing the structures of wild-type and mutant SAP proteins, we found that the substitution of amino acid Arginine to Serine causing a turnover of a hydrogen bond (Fig. 5B), suggesting the structure of SAP protein was changed. We also detected the expression of SAP in proband and the parents. As expected, flow cytometry detected that SAP protein was significantly downregulated in proband compared to which in parents. As shown in Figure 5C, SAP expression level in the proband, father and mother were 10.28%, 87.28%, and 80.31%, respectively, suggesting the function of SAP in proband was deficient. Overall, c.96G > T mutation of SH2D1A gene in the proband led to significant structural and functional deficiency of SAP protein, which was highly pathogenic. The mutant SAP protein was highly pathogenic with structural and functional. (A) The pathogenicity of mutant SAP protein was predicted by PolyPhen-2. The greater the pathogenicity, the closer the score is to 1.0. (B) The secondary structure of SAP protein and a structural comparison between wild-type and mutant SAP protein. The site and labels of mutation were highlighted in magenta. The green dotted lines illustrated hydrogen bonds. The magenta arrows indicated the turnover of a hydrogen bond. (C) Expression of SAP in the patient and parents detected by flow cytometry. Peaks in red were negative controls. Peaks in green illustrated expression of SAP. In order to explore whether the c.96G > T mutation in SH2D1A gene caused functional or structural deficiency in SAP protein, we first analyzed pathogenicity of mutant protein by PolyPhen-2 online scoring tool. As the greater the pathogenicity, the closer the score is to 1.0, the mutation was predicted to be highly pathogenic with a score of 1.0 (Fig. 5A). In addition, we analyzed the structural change of mutant SAP protein. As shown in Figure 5B, the secondary structure of SAP protein contained 2 α-helices and 8 β-strands and the mutation site was located at the end of the second β-strand. By comparing the structures of wild-type and mutant SAP proteins, we found that the substitution of amino acid Arginine to Serine causing a turnover of a hydrogen bond (Fig. 5B), suggesting the structure of SAP protein was changed. We also detected the expression of SAP in proband and the parents. As expected, flow cytometry detected that SAP protein was significantly downregulated in proband compared to which in parents. As shown in Figure 5C, SAP expression level in the proband, father and mother were 10.28%, 87.28%, and 80.31%, respectively, suggesting the function of SAP in proband was deficient. Overall, c.96G > T mutation of SH2D1A gene in the proband led to significant structural and functional deficiency of SAP protein, which was highly pathogenic. The mutant SAP protein was highly pathogenic with structural and functional. (A) The pathogenicity of mutant SAP protein was predicted by PolyPhen-2. The greater the pathogenicity, the closer the score is to 1.0. (B) The secondary structure of SAP protein and a structural comparison between wild-type and mutant SAP protein. The site and labels of mutation were highlighted in magenta. The green dotted lines illustrated hydrogen bonds. The magenta arrows indicated the turnover of a hydrogen bond. (C) Expression of SAP in the patient and parents detected by flow cytometry. Peaks in red were negative controls. Peaks in green illustrated expression of SAP. [SUBTITLE] 3.4. Activation of PI3K-AKT-mTOR signaling pathway in the proband [SUBSECTION] As previously stated, PI3K downstream signaling pathways may be altered in XLP-1 patients, thus we evaluated the expression of PI3K subunits p110δ and p85α, as well as downstream key proteins including AKT, p-AKT and mTOR in both the proband and the parents. To ensure that observed differences were due to XLP-1 rather than HLH, we also evaluated the expression of proteins in a familial HLH patient and a healthy child. As shown in Figure 6, PI3K-AKT-mTOR pathway was inactivated in the healthy control, as evidenced by high expression of the negative regulatory subunit p85α, which inhibit the activity of the functional subunit p110δ. Moreover, under normal circumstances, the downstream protein AKT was unphosphorylated and mTOR was not expressed. The proband had p85α downregulation and p110δ upregulation, AKT phosphorylation, and mTOR activation, indicating that the PI3K-AKT-mTOR pathway was considerably activated. The expression patterns of p85α, p110δ, AKT, and p-AKT were identical in the father and mother, but mTOR expression was muted. Although p85α and p110δ were excessively high in the familial HLH control, downstream AKT was unphosphorylated and mTOR was only expressed, in contrast to the fully activated PI3K-AKT-mTOR pathway in the XLP-1 proband. Overall, the findings showed that the PI3K-AKT-mTOR signaling pathway was fully activated in XLP-1 patients, but it was inactive or only partially activated in healthy people or HLH patients, implying that the PI3K-AKT-mTOR signaling pathway may play a role in XLP-1 pathogenesis. Expression of key proteins in PI3K downstream pathways. Peaks in red were negative controls. Peaks in green illustrated expression of proteins. PI3K = phophatidylinositol-3-kinase. As previously stated, PI3K downstream signaling pathways may be altered in XLP-1 patients, thus we evaluated the expression of PI3K subunits p110δ and p85α, as well as downstream key proteins including AKT, p-AKT and mTOR in both the proband and the parents. To ensure that observed differences were due to XLP-1 rather than HLH, we also evaluated the expression of proteins in a familial HLH patient and a healthy child. As shown in Figure 6, PI3K-AKT-mTOR pathway was inactivated in the healthy control, as evidenced by high expression of the negative regulatory subunit p85α, which inhibit the activity of the functional subunit p110δ. Moreover, under normal circumstances, the downstream protein AKT was unphosphorylated and mTOR was not expressed. The proband had p85α downregulation and p110δ upregulation, AKT phosphorylation, and mTOR activation, indicating that the PI3K-AKT-mTOR pathway was considerably activated. The expression patterns of p85α, p110δ, AKT, and p-AKT were identical in the father and mother, but mTOR expression was muted. Although p85α and p110δ were excessively high in the familial HLH control, downstream AKT was unphosphorylated and mTOR was only expressed, in contrast to the fully activated PI3K-AKT-mTOR pathway in the XLP-1 proband. Overall, the findings showed that the PI3K-AKT-mTOR signaling pathway was fully activated in XLP-1 patients, but it was inactive or only partially activated in healthy people or HLH patients, implying that the PI3K-AKT-mTOR signaling pathway may play a role in XLP-1 pathogenesis. Expression of key proteins in PI3K downstream pathways. Peaks in red were negative controls. Peaks in green illustrated expression of proteins. PI3K = phophatidylinositol-3-kinase.
5. Conclusions
In this study, we provided a detailed description of the clinical features of an XLP-1 patient and detected that the proband was caused by a hemizygous mutation c.96G > T in SH2D1A gene resulting in a missense substitution of Arginine to Serine (p.R32S). The mutant protein contained a hydrogen bond turnover at the site of mutation and the mutation resulted in downregulation of SH2D1A encoded protein SAP. The PI3K-AKT-mTOR signaling pathway was fully activated in XLP-1 patients, but it was inactive or only partially activated in healthy people or HLH patients, implying that the PI3K-AKT-mTOR signaling pathway may play a role in XLP-1 pathogenesis. This study would be helpful for subsequent research related to the diagnosis and pathogenesis of XLP-1.
[ "2.1. Editorial policies and ethical considerations", "2.2. Proband", "2.3. Whole exome sequencing", "2.4. Flow cytometry", "2.5. Prediction of the pathogenicity and structure of mutant protein", "3.1 Laboratory and imagological examinations indicated that the proband’s condition continued to deteriorate despite receiving treatment", "3.2. The proband was diagnosed with XLP-1 caused by a hemizygous mutation c.96G > T in SH2D1A gene", "3.3. The mutant SAP protein was highly pathogenic with structural and functional change", "3.4. Activation of PI3K-AKT-mTOR signaling pathway in the proband", "Acknowledgments", "Author contributions" ]
[ "The study was approved by the Ethics Committee of Kunming Children’s Hospital. All experiments were performed in compliance with the Helsinki Declaration. Informed written consent was obtained from the parents of the proband for the collection of clinical information, blood samples, DNA and presentation of patient’s materials.", "The proband, a 7-month-old male, was admitted to the hospital due to “fever with rash.” Fever was up to 40°C and not relieved after treatment. Physical examination of the proband on admission observed listless and poor mental response, scattered rashes on the trunk and limbs particularly on both forearms and lower legs, eyelid edema, pharyngeal congestion, white pustules attached to the pharyngeal tonsils, and several palpable enlarged lymph nodes in the neck up to about 1 × 1 cm in size with medium hardness and poor range of motion. The proband was initially treated with ganciclovir, sodium succinate and intravenous IgG to against viral infection, reduce inflammation and adjust immunity, respectively. With the progression of the disease, the proband had recurring fevers and icteric sclera indicating aggravated liver damage. The proband’s liver and spleen were progressively enlarged, and the rash became more widespread and fusing into patches in some area. Staphylococcus aureus were tested positive in sputum. Cefperazone-Sulbactam, meropenem and vancomycin were successively given against infection. Bone marrow examination showed hemophagocytosis. Etoposide was then given, during the treatment, the proband experienced progressive pancytopenia and was interfered with the infusion of granulocyte colony-stimulating factor, interleukin-11, and apheresis platelets. Unfortunately, the proband’s condition kept aggravating despite accepting treatment and finally died due to multiple organ dysfunctions. Parents of the proband were healthy, and no other remarkable diseases history was identified in the family.", "Peripheral blood samples were collected from the proband and parents. Genomic DNA was extracted from peripheral blood using the DNA Extraction kit (CWBIO, China). The isolated DNA was quantified by agarose gel electrophoresis and Nanodrop 2000 (Thermal Fisher Scientific). Libraries were prepared using Illumina standard protocol and a minimum of 3 μg DNA was employed for the indexed Illumina libraries. To capture targeted genes, the biotinylated capture probes were designed to tile all of the exons with non-repeated regions. The resulting captured DNA was amplified by PCR and the PCR product was purified with SPRI beads (Beckman Coulter). The enriched libraries were sequenced on an Illumina NextSeq 500 sequencer for paired-end reads of 150 bp. Quality control criteria were applied to the raw sequencing data before being mapped to the UCSC hg19 human reference genome using BWA (0.7.10). Picard tools (1.119) were used to eliminate duplicated reads. SNP and InDels were detected and filtered by GATK (Genome Analysis TK-3.3.0). Variants were further annotated by ANNOVAR. All mutations identified were confirmed by Sanger sequencing. Sites of variation were identified through a comparison of DNA sequences with the corresponding GenBank (www.ncbi.nlm.nih.gov) reference sequences. Exon 1 of SH2D1A (NM_002351) were amplified using 2 pair primers (Forward: 5’-GCTCGATCGAACCAAGCTAC-3’; Reverse: 5’-GGAGCGAAGGTAAACTGTGG-3’). The PCR samples were visualized on agarose gels, purified and sequenced using the terminator cycle sequencing method on an ABI PRISM 3730 genetic analyzer (Thermo Fisher Scientific). The sequencing results were analyzed using the DNASTAR (Madison) software.", "Peripheral blood samples were collected from the proband and parents, as well as a familial HLH patient and a healthy child as control. Peripheral blood mononuclear cells were isolated using lymphocyte separation solution. Protein expression in peripheral blood mononuclear cells was determined via flow cytometry. Briefly, cells were fixed and permeabilized sequentially with an IntraPrep Permeabilizaton Reagent kit (Beckman Coulter,A07803) following manufacturer’s instruction. Specific antibodies were then added for incubation overnight, followed by 30 mins incubation with a fluorescently labeled secondary antibody. The flow cytometry tests were carried out with a BD FACSCanto II flow cytometer, and the data was processed with CytExpert 2.0 (Beckman Coulter) software. Antibodies used were listed as follows: Anti-SH2D1A/SAP (Abcam, ab109120); Anti-PI3 Kinase p110δ (Abcam, ab109006); Anti-PI3 Kinase p85α (Abcam, ab191606); Anti-PTEN (Abcam, ab32199); Anti-AKT (CST, 9272s); Anti-phospho-AKT (CST, 9271s); Anti-mTOR (Abcam, ab2732).", "Pathogenicity of mutant protein was predicted by PolyPhen-2 online scoring tool (http://genetics.bwh.harvard.edu/pph2/index.shtml). The greater the pathogenicity, the closer the score is to 1.0. The structure of wild-type protein was created by SWISS-MODEL online tool (http://swissmodel.expasy.org/) and the structure of mutant protein was estimated by SWISS-PDB Viewer 4.1.0 software.", "To access detailed conditions of the proband, multiple laboratory and imagological examinations were conducted during hospitalization. Results showed that peripheral blood EBV nucleic acid load continued to rise and the antibodies to EBV capsid antigen (EBVCA) IgG and IgG antibodies to EBV nuclear antigen became positive as the disease progressed (Fig. 1A). The percentage of CD19 + B cells was slightly decreased (Fig. 1B), and humoral immunity test indicated increased levels of serum IgG, IgM, IgA, and decreased C3 (Fig. 1C). Hematological parameters of the proband pre- and during-hospitalization showed that the counts of leukocyte, lymphocyte, neutrophil, monocyte, platelet, erythrocyte and hemoglobin were declined with fluctuation and generally lower than reference values. The percentages of lymphocyte, neutrophil and monocyte were all fluctuated outside the reference value (Fig. 1D). Besides, indicators for liver, renal and cardiac function of proband were abnormal as disease progressed. The levels of alanine aminotransferase, aspartate aminotransferase, γ-glutamyltransferase (γ-GGT), alkaline phosphatase, total bilirubin, direct bilirubin, total bile acid, lactate dehydrogenase and α-hydroxybutyrate dehydrogenase (α-HBDH) were all extremely higher than reference values (Fig. 1E–G). Additionally, the levels of ferritin and triglycerides elevated dramatically indicated the development of HLH (Fig. 2A). Occurrence of HLH was also confirmed by hemophagocytes in bone marrow (Fig. 2B) and increased expression of serum IL-6 and IL-10, with IL-10 being 12 times higher than the reference values (Fig. 2C). Abdominal color ultrasound revealed hepatomegaly, gallbladder wall thickening, mild splenomegaly and abdominal effusion (Fig. 3A). Neck color ultrasound revealed slightly enlarged lymph nodes on both sides of neck and jaw (Fig. 3B). Chest X-ray revealed increased bilateral pleural effusion with deterioration of disease (Fig. 3C). Overall, with the progression of disease, the condition of the proband kept deteriorating, especially the infection was aggravated, immunity responses were deficient, and multiple organs were progressively disabled. The clinical features including EBV-infection, bone marrow hemophagocytosis, dysgammaglobulinemia, hepatosplenomegaly and lymphadenopathy observed suggested that the proband may suffer from XLP.\nLaboratory examinations of the proband with the progression of disease. (A) Results of peripheral blood EBV nucleic acid and antibodies test. (B) Results of lymphocyte subsets test. (C) Results of humoral immunity test. (D) Results of whole blood counts. There were ten times of testing results during hospitalization listed in chronological order from left to right. (E) Results of liver function test. There were 10 times of testing results during hospitalization listed in chronological order from left to right. (F) Results of renal function test. There were 8 times of testing results during hospitalization listed in chronological order from left to right. (G) Results of cardiac function test. There were 7 times of testing results during hospitalization listed in chronological order from left to right. EBV = Epstein-Barr virus.\nOccurrence of hemophagocytic lymphohistiocytosis was detected in the proband. (A) Results of ferritin and triglycerides tests. There were 4 times of testing results during hospitalization listed in chronological order from left to right. (B) Results of cytopathology detection of bone marrow. (C) Results of serum cytokine test.\nImageological examinations of the proband with the progression of disease. (A) Results of abdominal color ultrasound. (B) Results of neck color ultrasound. (C) Results of chest X-ray. There were 5 times of examine results during hospitalization listed in chronological order from left to right.", "In order to confirm whether the proband was suffered from genetic disorders, the peripheral blood DNA of the proband and parents were extracted, and whole exome sequencing was performed. The results revealed a hemizygous mutation in exon 1 of SH2D1A gene, which substituted Guanine to Thymine at the site of nucleotide 96 (c.96G > T) (Fig. 4A), resulting in a missense substitution of Arginine to Serine at the site of amino acid 32 (p.R32S). According to the ACMG guidelines, this mutation was identified as a pathogenic mutation. It was located in the mutation hotspot region with a low-frequency in the normal population database. The mutation with the same amino acid change as this proband had been reported previously but with different nucleotide change.[20] Sanger sequencing validation of this mutation was performed on the parents, and the results identified no variation at this site in the father and a heterozygous variation at this site in the mother (Fig. 4B and C). As XLP-1 inherited in a recessive pattern, the parents of the proband were all appeared healthy. Therefore, the diagnosis of XLP-1 was confirmed based on the proband’s typical clinical manifestations and genetic analysis results.\nIdentification of a hemizygous mutation c.96G > T in SH2D1A gene of the proband. (A) Schematic diagram of SH2D1A gene. The mutation site was highlighted in red. (B) Results of Sanger sequencing. The arrows indicated the sites of mutation. (C) Genetic pedigree of the family.", "In order to explore whether the c.96G > T mutation in SH2D1A gene caused functional or structural deficiency in SAP protein, we first analyzed pathogenicity of mutant protein by PolyPhen-2 online scoring tool. As the greater the pathogenicity, the closer the score is to 1.0, the mutation was predicted to be highly pathogenic with a score of 1.0 (Fig. 5A). In addition, we analyzed the structural change of mutant SAP protein. As shown in Figure 5B, the secondary structure of SAP protein contained 2 α-helices and 8 β-strands and the mutation site was located at the end of the second β-strand. By comparing the structures of wild-type and mutant SAP proteins, we found that the substitution of amino acid Arginine to Serine causing a turnover of a hydrogen bond (Fig. 5B), suggesting the structure of SAP protein was changed. We also detected the expression of SAP in proband and the parents. As expected, flow cytometry detected that SAP protein was significantly downregulated in proband compared to which in parents. As shown in Figure 5C, SAP expression level in the proband, father and mother were 10.28%, 87.28%, and 80.31%, respectively, suggesting the function of SAP in proband was deficient. Overall, c.96G > T mutation of SH2D1A gene in the proband led to significant structural and functional deficiency of SAP protein, which was highly pathogenic.\nThe mutant SAP protein was highly pathogenic with structural and functional. (A) The pathogenicity of mutant SAP protein was predicted by PolyPhen-2. The greater the pathogenicity, the closer the score is to 1.0. (B) The secondary structure of SAP protein and a structural comparison between wild-type and mutant SAP protein. The site and labels of mutation were highlighted in magenta. The green dotted lines illustrated hydrogen bonds. The magenta arrows indicated the turnover of a hydrogen bond. (C) Expression of SAP in the patient and parents detected by flow cytometry. Peaks in red were negative controls. Peaks in green illustrated expression of SAP.", "As previously stated, PI3K downstream signaling pathways may be altered in XLP-1 patients, thus we evaluated the expression of PI3K subunits p110δ and p85α, as well as downstream key proteins including AKT, p-AKT and mTOR in both the proband and the parents. To ensure that observed differences were due to XLP-1 rather than HLH, we also evaluated the expression of proteins in a familial HLH patient and a healthy child. As shown in Figure 6, PI3K-AKT-mTOR pathway was inactivated in the healthy control, as evidenced by high expression of the negative regulatory subunit p85α, which inhibit the activity of the functional subunit p110δ. Moreover, under normal circumstances, the downstream protein AKT was unphosphorylated and mTOR was not expressed. The proband had p85α downregulation and p110δ upregulation, AKT phosphorylation, and mTOR activation, indicating that the PI3K-AKT-mTOR pathway was considerably activated. The expression patterns of p85α, p110δ, AKT, and p-AKT were identical in the father and mother, but mTOR expression was muted. Although p85α and p110δ were excessively high in the familial HLH control, downstream AKT was unphosphorylated and mTOR was only expressed, in contrast to the fully activated PI3K-AKT-mTOR pathway in the XLP-1 proband. Overall, the findings showed that the PI3K-AKT-mTOR signaling pathway was fully activated in XLP-1 patients, but it was inactive or only partially activated in healthy people or HLH patients, implying that the PI3K-AKT-mTOR signaling pathway may play a role in XLP-1 pathogenesis.\nExpression of key proteins in PI3K downstream pathways. Peaks in red were negative controls. Peaks in green illustrated expression of proteins. PI3K = phophatidylinositol-3-kinase.", "The authors sincerely thank the patient and his family members for their participation and support.\nYW, YW, WL, and LT contributed equally to this work.", "Conceptualization: Yanchun Wang, Yan Wang, Weimin Lu, Lvyan Tao.\nData curation: Yang Xiao, Yuantao Zhou, Xiaoli He.\nFormal analysis: Yanchun Wang, Yan Wang, Weimin Lu, Lvyan Tao.\nInvestigation: Yang Xiao, Yuantao Zhou, Xiaoli He.\nMethodology: Yanchun Wang, Yan Wang, Weimin Lu, Lvyan Tao.\nResources: Yanchun Wang, Yan Wang, Weimin Lu, Lvyan Tao.\nSupervision: Yu Zhang, Li Li.\nVisualization: Yanchun Wang, Yan Wang, Weimin Lu, Lvyan Tao.\nWriting – original draft: Yu Zhang, Li Li.\nWriting – review & editing: Yu Zhang, Li Li." ]
[ null, null, null, null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Methods", "2.1. Editorial policies and ethical considerations", "2.2. Proband", "2.3. Whole exome sequencing", "2.4. Flow cytometry", "2.5. Prediction of the pathogenicity and structure of mutant protein", "3. Results", "3.1 Laboratory and imagological examinations indicated that the proband’s condition continued to deteriorate despite receiving treatment", "3.2. The proband was diagnosed with XLP-1 caused by a hemizygous mutation c.96G > T in SH2D1A gene", "3.3. The mutant SAP protein was highly pathogenic with structural and functional change", "3.4. Activation of PI3K-AKT-mTOR signaling pathway in the proband", "4. Discussion", "5. Conclusions", "Acknowledgments", "Author contributions" ]
[ "X-linked lymphoproliferative syndrome (XLP) is an extremely rare X-linked recessive inborn errors of immunity caused by genetic variations, with a 1/1000000 incidence rate,[12]. XLP is currently classified into 2 forms based on mutant genes: Type 1 XLP (XLP-1) and Type 2 XLP (XLP-2) caused by mutations in SH2D1A gene and XIAP gene, respectively[3,4]. Common clinical characteristics of XLP-1 and XLP-2 including Epstein-Barr virus (EBV) infection, hemophagocytic lymphohistiocytosis (HLH), lymphoproliferative disorder, and dysgammaglobulinemia, but sometimes they also manifested differently. According to published studies, EBV infection and associated HLH were more frequently observed in patients with XLP-1, and sometimes even accompanied by catastrophic neurologic disorders. Development of lymphoma is only reported in XLP-1 patients[5]. Moreover, Natural killer cells (NK) and T lymphocytes are more likely to proliferate and infiltrate organs, including liver, spleen, and lymph nodes, in EBV-infected XLP-1 patients[6]. Persistent hypogammaglobulinemia appeared more often in XLP-1 patients while transient hypogammaglobulinemia was more frequently observed in XLP-2[7]. Inflammatory bowel disease, such as Crohn’s disease, was exclusively discovered in XLP-2 patients, and roughly 25% to 30% of XLP-2 patients were impacted[8]. Additionally, fever, subcutaneous petechia, hematemesis, hematochezia, pistaxis, jaundice, hepatosplenomegaly, lymphadenopathy, lymphocytic vasculitis, aplastic anemia, and lymphomatoid granulomatosis may also be signs and symptoms of XLP[9–11]. Due to the rapid progression and complicated symptoms of XLP, it is commonly misdiagnosed clinically. XLP has a poor prognosis and high mortality, which have been reported to be 75%, of which 70% died before the age of 10[12]. At present, the only promising treatment is allogeneic hematopoietic stem cell transplantation (HSCT) before the onset of typical symptoms or EBV infection[13].\nThe SH2D1A gene is found on chromosome Xq25 and has 4 exons, and mutations in this gene can result in SAP protein deficiency. SAP is a 128-amino acids signaling lymphocyte activating molecule (SLAM)-associated protein that contains 1 Src homology 2 (SH2) domain and is primarily expressed in T cells, NK cells, and some EBV-positive Burkitt lymphoma-derived B cells.[14] SAP can competitively bind to SLAMs via the SH2 domain and regulate a variety of processes, including the development and function of invariant natural killer T cells (iNKT), the clearance of EBV-infected B cells with cytotoxic T lymphocytes (CTLs) and NK cells, the development of germinal centers, the production of immunoglobulin, T cell restimulation-induced cell death, and the maintenance of T cell homeostasis.[15] In XLP-1 patients, SAP deficiency can prevent CTLs and NK cells from killing EBV-infected B cells.[16] The XIAP gene is also located on chromosome Xq25 and includes 7 exons, its encoding protein XIAP is a novel member of the inhibitor of apoptosis proteins family, which inhibits caspases directly and regulates apoptosis through several routes. XIAP gene is overexpressed in many tumor cell lines, and its expression is closely related to tumor progression, recurrence, prognosis, and treatment resistance.[17] Phophatidylinositol-3-kinase (PI3K) is a crucial signaling center in immune cells that catalyzes the conversion of PIP2 to PIP3 and thus facilitates membrane recruitment of molecules containing PIP3-binding Pleckstrin homology domains such as the AKT, PDK1, Tec family kinases, adapter molecules, guanine nucleotide exchange factors, and GTPase-activating proteins. These subsequently activate a number of important downstream pathways including mTOR, FOXO1 and BACH2-related pathways.[18] Too little or too much activity in the PI3K downstream pathway is harmful even pathogenic. Previous research discovered that SAP protein was downregulated in CTLs while the SLAM protein 2B4 was upregulated in patients with congenital PI3K deficiency disorder such as activated PI3Kδ syndrome, implying that SAP may interact with PI3K-associated pathways and XLP-1 may be related to PI3K signaling failure.[19] Insight details of the interaction patterns between SAP and PI3K, as well as the role of PI3K-associated pathways in pathogenesis of XLP-1 still need to be investigated further.\nIn this study, we will describe an infant with XLP-1, assess the proband’s clinical features and genetic variants, analyze the structural and functional changes in SAP protein caused by gene mutation, and investigate the probable role of PI3K-associated pathways in XLP-1 pathogenesis.", "[SUBTITLE] 2.1. Editorial policies and ethical considerations [SUBSECTION] The study was approved by the Ethics Committee of Kunming Children’s Hospital. All experiments were performed in compliance with the Helsinki Declaration. Informed written consent was obtained from the parents of the proband for the collection of clinical information, blood samples, DNA and presentation of patient’s materials.\nThe study was approved by the Ethics Committee of Kunming Children’s Hospital. All experiments were performed in compliance with the Helsinki Declaration. Informed written consent was obtained from the parents of the proband for the collection of clinical information, blood samples, DNA and presentation of patient’s materials.\n[SUBTITLE] 2.2. Proband [SUBSECTION] The proband, a 7-month-old male, was admitted to the hospital due to “fever with rash.” Fever was up to 40°C and not relieved after treatment. Physical examination of the proband on admission observed listless and poor mental response, scattered rashes on the trunk and limbs particularly on both forearms and lower legs, eyelid edema, pharyngeal congestion, white pustules attached to the pharyngeal tonsils, and several palpable enlarged lymph nodes in the neck up to about 1 × 1 cm in size with medium hardness and poor range of motion. The proband was initially treated with ganciclovir, sodium succinate and intravenous IgG to against viral infection, reduce inflammation and adjust immunity, respectively. With the progression of the disease, the proband had recurring fevers and icteric sclera indicating aggravated liver damage. The proband’s liver and spleen were progressively enlarged, and the rash became more widespread and fusing into patches in some area. Staphylococcus aureus were tested positive in sputum. Cefperazone-Sulbactam, meropenem and vancomycin were successively given against infection. Bone marrow examination showed hemophagocytosis. Etoposide was then given, during the treatment, the proband experienced progressive pancytopenia and was interfered with the infusion of granulocyte colony-stimulating factor, interleukin-11, and apheresis platelets. Unfortunately, the proband’s condition kept aggravating despite accepting treatment and finally died due to multiple organ dysfunctions. Parents of the proband were healthy, and no other remarkable diseases history was identified in the family.\nThe proband, a 7-month-old male, was admitted to the hospital due to “fever with rash.” Fever was up to 40°C and not relieved after treatment. Physical examination of the proband on admission observed listless and poor mental response, scattered rashes on the trunk and limbs particularly on both forearms and lower legs, eyelid edema, pharyngeal congestion, white pustules attached to the pharyngeal tonsils, and several palpable enlarged lymph nodes in the neck up to about 1 × 1 cm in size with medium hardness and poor range of motion. The proband was initially treated with ganciclovir, sodium succinate and intravenous IgG to against viral infection, reduce inflammation and adjust immunity, respectively. With the progression of the disease, the proband had recurring fevers and icteric sclera indicating aggravated liver damage. The proband’s liver and spleen were progressively enlarged, and the rash became more widespread and fusing into patches in some area. Staphylococcus aureus were tested positive in sputum. Cefperazone-Sulbactam, meropenem and vancomycin were successively given against infection. Bone marrow examination showed hemophagocytosis. Etoposide was then given, during the treatment, the proband experienced progressive pancytopenia and was interfered with the infusion of granulocyte colony-stimulating factor, interleukin-11, and apheresis platelets. Unfortunately, the proband’s condition kept aggravating despite accepting treatment and finally died due to multiple organ dysfunctions. Parents of the proband were healthy, and no other remarkable diseases history was identified in the family.\n[SUBTITLE] 2.3. Whole exome sequencing [SUBSECTION] Peripheral blood samples were collected from the proband and parents. Genomic DNA was extracted from peripheral blood using the DNA Extraction kit (CWBIO, China). The isolated DNA was quantified by agarose gel electrophoresis and Nanodrop 2000 (Thermal Fisher Scientific). Libraries were prepared using Illumina standard protocol and a minimum of 3 μg DNA was employed for the indexed Illumina libraries. To capture targeted genes, the biotinylated capture probes were designed to tile all of the exons with non-repeated regions. The resulting captured DNA was amplified by PCR and the PCR product was purified with SPRI beads (Beckman Coulter). The enriched libraries were sequenced on an Illumina NextSeq 500 sequencer for paired-end reads of 150 bp. Quality control criteria were applied to the raw sequencing data before being mapped to the UCSC hg19 human reference genome using BWA (0.7.10). Picard tools (1.119) were used to eliminate duplicated reads. SNP and InDels were detected and filtered by GATK (Genome Analysis TK-3.3.0). Variants were further annotated by ANNOVAR. All mutations identified were confirmed by Sanger sequencing. Sites of variation were identified through a comparison of DNA sequences with the corresponding GenBank (www.ncbi.nlm.nih.gov) reference sequences. Exon 1 of SH2D1A (NM_002351) were amplified using 2 pair primers (Forward: 5’-GCTCGATCGAACCAAGCTAC-3’; Reverse: 5’-GGAGCGAAGGTAAACTGTGG-3’). The PCR samples were visualized on agarose gels, purified and sequenced using the terminator cycle sequencing method on an ABI PRISM 3730 genetic analyzer (Thermo Fisher Scientific). The sequencing results were analyzed using the DNASTAR (Madison) software.\nPeripheral blood samples were collected from the proband and parents. Genomic DNA was extracted from peripheral blood using the DNA Extraction kit (CWBIO, China). The isolated DNA was quantified by agarose gel electrophoresis and Nanodrop 2000 (Thermal Fisher Scientific). Libraries were prepared using Illumina standard protocol and a minimum of 3 μg DNA was employed for the indexed Illumina libraries. To capture targeted genes, the biotinylated capture probes were designed to tile all of the exons with non-repeated regions. The resulting captured DNA was amplified by PCR and the PCR product was purified with SPRI beads (Beckman Coulter). The enriched libraries were sequenced on an Illumina NextSeq 500 sequencer for paired-end reads of 150 bp. Quality control criteria were applied to the raw sequencing data before being mapped to the UCSC hg19 human reference genome using BWA (0.7.10). Picard tools (1.119) were used to eliminate duplicated reads. SNP and InDels were detected and filtered by GATK (Genome Analysis TK-3.3.0). Variants were further annotated by ANNOVAR. All mutations identified were confirmed by Sanger sequencing. Sites of variation were identified through a comparison of DNA sequences with the corresponding GenBank (www.ncbi.nlm.nih.gov) reference sequences. Exon 1 of SH2D1A (NM_002351) were amplified using 2 pair primers (Forward: 5’-GCTCGATCGAACCAAGCTAC-3’; Reverse: 5’-GGAGCGAAGGTAAACTGTGG-3’). The PCR samples were visualized on agarose gels, purified and sequenced using the terminator cycle sequencing method on an ABI PRISM 3730 genetic analyzer (Thermo Fisher Scientific). The sequencing results were analyzed using the DNASTAR (Madison) software.\n[SUBTITLE] 2.4. Flow cytometry [SUBSECTION] Peripheral blood samples were collected from the proband and parents, as well as a familial HLH patient and a healthy child as control. Peripheral blood mononuclear cells were isolated using lymphocyte separation solution. Protein expression in peripheral blood mononuclear cells was determined via flow cytometry. Briefly, cells were fixed and permeabilized sequentially with an IntraPrep Permeabilizaton Reagent kit (Beckman Coulter,A07803) following manufacturer’s instruction. Specific antibodies were then added for incubation overnight, followed by 30 mins incubation with a fluorescently labeled secondary antibody. The flow cytometry tests were carried out with a BD FACSCanto II flow cytometer, and the data was processed with CytExpert 2.0 (Beckman Coulter) software. Antibodies used were listed as follows: Anti-SH2D1A/SAP (Abcam, ab109120); Anti-PI3 Kinase p110δ (Abcam, ab109006); Anti-PI3 Kinase p85α (Abcam, ab191606); Anti-PTEN (Abcam, ab32199); Anti-AKT (CST, 9272s); Anti-phospho-AKT (CST, 9271s); Anti-mTOR (Abcam, ab2732).\nPeripheral blood samples were collected from the proband and parents, as well as a familial HLH patient and a healthy child as control. Peripheral blood mononuclear cells were isolated using lymphocyte separation solution. Protein expression in peripheral blood mononuclear cells was determined via flow cytometry. Briefly, cells were fixed and permeabilized sequentially with an IntraPrep Permeabilizaton Reagent kit (Beckman Coulter,A07803) following manufacturer’s instruction. Specific antibodies were then added for incubation overnight, followed by 30 mins incubation with a fluorescently labeled secondary antibody. The flow cytometry tests were carried out with a BD FACSCanto II flow cytometer, and the data was processed with CytExpert 2.0 (Beckman Coulter) software. Antibodies used were listed as follows: Anti-SH2D1A/SAP (Abcam, ab109120); Anti-PI3 Kinase p110δ (Abcam, ab109006); Anti-PI3 Kinase p85α (Abcam, ab191606); Anti-PTEN (Abcam, ab32199); Anti-AKT (CST, 9272s); Anti-phospho-AKT (CST, 9271s); Anti-mTOR (Abcam, ab2732).\n[SUBTITLE] 2.5. Prediction of the pathogenicity and structure of mutant protein [SUBSECTION] Pathogenicity of mutant protein was predicted by PolyPhen-2 online scoring tool (http://genetics.bwh.harvard.edu/pph2/index.shtml). The greater the pathogenicity, the closer the score is to 1.0. The structure of wild-type protein was created by SWISS-MODEL online tool (http://swissmodel.expasy.org/) and the structure of mutant protein was estimated by SWISS-PDB Viewer 4.1.0 software.\nPathogenicity of mutant protein was predicted by PolyPhen-2 online scoring tool (http://genetics.bwh.harvard.edu/pph2/index.shtml). The greater the pathogenicity, the closer the score is to 1.0. The structure of wild-type protein was created by SWISS-MODEL online tool (http://swissmodel.expasy.org/) and the structure of mutant protein was estimated by SWISS-PDB Viewer 4.1.0 software.", "The study was approved by the Ethics Committee of Kunming Children’s Hospital. All experiments were performed in compliance with the Helsinki Declaration. Informed written consent was obtained from the parents of the proband for the collection of clinical information, blood samples, DNA and presentation of patient’s materials.", "The proband, a 7-month-old male, was admitted to the hospital due to “fever with rash.” Fever was up to 40°C and not relieved after treatment. Physical examination of the proband on admission observed listless and poor mental response, scattered rashes on the trunk and limbs particularly on both forearms and lower legs, eyelid edema, pharyngeal congestion, white pustules attached to the pharyngeal tonsils, and several palpable enlarged lymph nodes in the neck up to about 1 × 1 cm in size with medium hardness and poor range of motion. The proband was initially treated with ganciclovir, sodium succinate and intravenous IgG to against viral infection, reduce inflammation and adjust immunity, respectively. With the progression of the disease, the proband had recurring fevers and icteric sclera indicating aggravated liver damage. The proband’s liver and spleen were progressively enlarged, and the rash became more widespread and fusing into patches in some area. Staphylococcus aureus were tested positive in sputum. Cefperazone-Sulbactam, meropenem and vancomycin were successively given against infection. Bone marrow examination showed hemophagocytosis. Etoposide was then given, during the treatment, the proband experienced progressive pancytopenia and was interfered with the infusion of granulocyte colony-stimulating factor, interleukin-11, and apheresis platelets. Unfortunately, the proband’s condition kept aggravating despite accepting treatment and finally died due to multiple organ dysfunctions. Parents of the proband were healthy, and no other remarkable diseases history was identified in the family.", "Peripheral blood samples were collected from the proband and parents. Genomic DNA was extracted from peripheral blood using the DNA Extraction kit (CWBIO, China). The isolated DNA was quantified by agarose gel electrophoresis and Nanodrop 2000 (Thermal Fisher Scientific). Libraries were prepared using Illumina standard protocol and a minimum of 3 μg DNA was employed for the indexed Illumina libraries. To capture targeted genes, the biotinylated capture probes were designed to tile all of the exons with non-repeated regions. The resulting captured DNA was amplified by PCR and the PCR product was purified with SPRI beads (Beckman Coulter). The enriched libraries were sequenced on an Illumina NextSeq 500 sequencer for paired-end reads of 150 bp. Quality control criteria were applied to the raw sequencing data before being mapped to the UCSC hg19 human reference genome using BWA (0.7.10). Picard tools (1.119) were used to eliminate duplicated reads. SNP and InDels were detected and filtered by GATK (Genome Analysis TK-3.3.0). Variants were further annotated by ANNOVAR. All mutations identified were confirmed by Sanger sequencing. Sites of variation were identified through a comparison of DNA sequences with the corresponding GenBank (www.ncbi.nlm.nih.gov) reference sequences. Exon 1 of SH2D1A (NM_002351) were amplified using 2 pair primers (Forward: 5’-GCTCGATCGAACCAAGCTAC-3’; Reverse: 5’-GGAGCGAAGGTAAACTGTGG-3’). The PCR samples were visualized on agarose gels, purified and sequenced using the terminator cycle sequencing method on an ABI PRISM 3730 genetic analyzer (Thermo Fisher Scientific). The sequencing results were analyzed using the DNASTAR (Madison) software.", "Peripheral blood samples were collected from the proband and parents, as well as a familial HLH patient and a healthy child as control. Peripheral blood mononuclear cells were isolated using lymphocyte separation solution. Protein expression in peripheral blood mononuclear cells was determined via flow cytometry. Briefly, cells were fixed and permeabilized sequentially with an IntraPrep Permeabilizaton Reagent kit (Beckman Coulter,A07803) following manufacturer’s instruction. Specific antibodies were then added for incubation overnight, followed by 30 mins incubation with a fluorescently labeled secondary antibody. The flow cytometry tests were carried out with a BD FACSCanto II flow cytometer, and the data was processed with CytExpert 2.0 (Beckman Coulter) software. Antibodies used were listed as follows: Anti-SH2D1A/SAP (Abcam, ab109120); Anti-PI3 Kinase p110δ (Abcam, ab109006); Anti-PI3 Kinase p85α (Abcam, ab191606); Anti-PTEN (Abcam, ab32199); Anti-AKT (CST, 9272s); Anti-phospho-AKT (CST, 9271s); Anti-mTOR (Abcam, ab2732).", "Pathogenicity of mutant protein was predicted by PolyPhen-2 online scoring tool (http://genetics.bwh.harvard.edu/pph2/index.shtml). The greater the pathogenicity, the closer the score is to 1.0. The structure of wild-type protein was created by SWISS-MODEL online tool (http://swissmodel.expasy.org/) and the structure of mutant protein was estimated by SWISS-PDB Viewer 4.1.0 software.", "[SUBTITLE] 3.1 Laboratory and imagological examinations indicated that the proband’s condition continued to deteriorate despite receiving treatment [SUBSECTION] To access detailed conditions of the proband, multiple laboratory and imagological examinations were conducted during hospitalization. Results showed that peripheral blood EBV nucleic acid load continued to rise and the antibodies to EBV capsid antigen (EBVCA) IgG and IgG antibodies to EBV nuclear antigen became positive as the disease progressed (Fig. 1A). The percentage of CD19 + B cells was slightly decreased (Fig. 1B), and humoral immunity test indicated increased levels of serum IgG, IgM, IgA, and decreased C3 (Fig. 1C). Hematological parameters of the proband pre- and during-hospitalization showed that the counts of leukocyte, lymphocyte, neutrophil, monocyte, platelet, erythrocyte and hemoglobin were declined with fluctuation and generally lower than reference values. The percentages of lymphocyte, neutrophil and monocyte were all fluctuated outside the reference value (Fig. 1D). Besides, indicators for liver, renal and cardiac function of proband were abnormal as disease progressed. The levels of alanine aminotransferase, aspartate aminotransferase, γ-glutamyltransferase (γ-GGT), alkaline phosphatase, total bilirubin, direct bilirubin, total bile acid, lactate dehydrogenase and α-hydroxybutyrate dehydrogenase (α-HBDH) were all extremely higher than reference values (Fig. 1E–G). Additionally, the levels of ferritin and triglycerides elevated dramatically indicated the development of HLH (Fig. 2A). Occurrence of HLH was also confirmed by hemophagocytes in bone marrow (Fig. 2B) and increased expression of serum IL-6 and IL-10, with IL-10 being 12 times higher than the reference values (Fig. 2C). Abdominal color ultrasound revealed hepatomegaly, gallbladder wall thickening, mild splenomegaly and abdominal effusion (Fig. 3A). Neck color ultrasound revealed slightly enlarged lymph nodes on both sides of neck and jaw (Fig. 3B). Chest X-ray revealed increased bilateral pleural effusion with deterioration of disease (Fig. 3C). Overall, with the progression of disease, the condition of the proband kept deteriorating, especially the infection was aggravated, immunity responses were deficient, and multiple organs were progressively disabled. The clinical features including EBV-infection, bone marrow hemophagocytosis, dysgammaglobulinemia, hepatosplenomegaly and lymphadenopathy observed suggested that the proband may suffer from XLP.\nLaboratory examinations of the proband with the progression of disease. (A) Results of peripheral blood EBV nucleic acid and antibodies test. (B) Results of lymphocyte subsets test. (C) Results of humoral immunity test. (D) Results of whole blood counts. There were ten times of testing results during hospitalization listed in chronological order from left to right. (E) Results of liver function test. There were 10 times of testing results during hospitalization listed in chronological order from left to right. (F) Results of renal function test. There were 8 times of testing results during hospitalization listed in chronological order from left to right. (G) Results of cardiac function test. There were 7 times of testing results during hospitalization listed in chronological order from left to right. EBV = Epstein-Barr virus.\nOccurrence of hemophagocytic lymphohistiocytosis was detected in the proband. (A) Results of ferritin and triglycerides tests. There were 4 times of testing results during hospitalization listed in chronological order from left to right. (B) Results of cytopathology detection of bone marrow. (C) Results of serum cytokine test.\nImageological examinations of the proband with the progression of disease. (A) Results of abdominal color ultrasound. (B) Results of neck color ultrasound. (C) Results of chest X-ray. There were 5 times of examine results during hospitalization listed in chronological order from left to right.\nTo access detailed conditions of the proband, multiple laboratory and imagological examinations were conducted during hospitalization. Results showed that peripheral blood EBV nucleic acid load continued to rise and the antibodies to EBV capsid antigen (EBVCA) IgG and IgG antibodies to EBV nuclear antigen became positive as the disease progressed (Fig. 1A). The percentage of CD19 + B cells was slightly decreased (Fig. 1B), and humoral immunity test indicated increased levels of serum IgG, IgM, IgA, and decreased C3 (Fig. 1C). Hematological parameters of the proband pre- and during-hospitalization showed that the counts of leukocyte, lymphocyte, neutrophil, monocyte, platelet, erythrocyte and hemoglobin were declined with fluctuation and generally lower than reference values. The percentages of lymphocyte, neutrophil and monocyte were all fluctuated outside the reference value (Fig. 1D). Besides, indicators for liver, renal and cardiac function of proband were abnormal as disease progressed. The levels of alanine aminotransferase, aspartate aminotransferase, γ-glutamyltransferase (γ-GGT), alkaline phosphatase, total bilirubin, direct bilirubin, total bile acid, lactate dehydrogenase and α-hydroxybutyrate dehydrogenase (α-HBDH) were all extremely higher than reference values (Fig. 1E–G). Additionally, the levels of ferritin and triglycerides elevated dramatically indicated the development of HLH (Fig. 2A). Occurrence of HLH was also confirmed by hemophagocytes in bone marrow (Fig. 2B) and increased expression of serum IL-6 and IL-10, with IL-10 being 12 times higher than the reference values (Fig. 2C). Abdominal color ultrasound revealed hepatomegaly, gallbladder wall thickening, mild splenomegaly and abdominal effusion (Fig. 3A). Neck color ultrasound revealed slightly enlarged lymph nodes on both sides of neck and jaw (Fig. 3B). Chest X-ray revealed increased bilateral pleural effusion with deterioration of disease (Fig. 3C). Overall, with the progression of disease, the condition of the proband kept deteriorating, especially the infection was aggravated, immunity responses were deficient, and multiple organs were progressively disabled. The clinical features including EBV-infection, bone marrow hemophagocytosis, dysgammaglobulinemia, hepatosplenomegaly and lymphadenopathy observed suggested that the proband may suffer from XLP.\nLaboratory examinations of the proband with the progression of disease. (A) Results of peripheral blood EBV nucleic acid and antibodies test. (B) Results of lymphocyte subsets test. (C) Results of humoral immunity test. (D) Results of whole blood counts. There were ten times of testing results during hospitalization listed in chronological order from left to right. (E) Results of liver function test. There were 10 times of testing results during hospitalization listed in chronological order from left to right. (F) Results of renal function test. There were 8 times of testing results during hospitalization listed in chronological order from left to right. (G) Results of cardiac function test. There were 7 times of testing results during hospitalization listed in chronological order from left to right. EBV = Epstein-Barr virus.\nOccurrence of hemophagocytic lymphohistiocytosis was detected in the proband. (A) Results of ferritin and triglycerides tests. There were 4 times of testing results during hospitalization listed in chronological order from left to right. (B) Results of cytopathology detection of bone marrow. (C) Results of serum cytokine test.\nImageological examinations of the proband with the progression of disease. (A) Results of abdominal color ultrasound. (B) Results of neck color ultrasound. (C) Results of chest X-ray. There were 5 times of examine results during hospitalization listed in chronological order from left to right.\n[SUBTITLE] 3.2. The proband was diagnosed with XLP-1 caused by a hemizygous mutation c.96G > T in SH2D1A gene [SUBSECTION] In order to confirm whether the proband was suffered from genetic disorders, the peripheral blood DNA of the proband and parents were extracted, and whole exome sequencing was performed. The results revealed a hemizygous mutation in exon 1 of SH2D1A gene, which substituted Guanine to Thymine at the site of nucleotide 96 (c.96G > T) (Fig. 4A), resulting in a missense substitution of Arginine to Serine at the site of amino acid 32 (p.R32S). According to the ACMG guidelines, this mutation was identified as a pathogenic mutation. It was located in the mutation hotspot region with a low-frequency in the normal population database. The mutation with the same amino acid change as this proband had been reported previously but with different nucleotide change.[20] Sanger sequencing validation of this mutation was performed on the parents, and the results identified no variation at this site in the father and a heterozygous variation at this site in the mother (Fig. 4B and C). As XLP-1 inherited in a recessive pattern, the parents of the proband were all appeared healthy. Therefore, the diagnosis of XLP-1 was confirmed based on the proband’s typical clinical manifestations and genetic analysis results.\nIdentification of a hemizygous mutation c.96G > T in SH2D1A gene of the proband. (A) Schematic diagram of SH2D1A gene. The mutation site was highlighted in red. (B) Results of Sanger sequencing. The arrows indicated the sites of mutation. (C) Genetic pedigree of the family.\nIn order to confirm whether the proband was suffered from genetic disorders, the peripheral blood DNA of the proband and parents were extracted, and whole exome sequencing was performed. The results revealed a hemizygous mutation in exon 1 of SH2D1A gene, which substituted Guanine to Thymine at the site of nucleotide 96 (c.96G > T) (Fig. 4A), resulting in a missense substitution of Arginine to Serine at the site of amino acid 32 (p.R32S). According to the ACMG guidelines, this mutation was identified as a pathogenic mutation. It was located in the mutation hotspot region with a low-frequency in the normal population database. The mutation with the same amino acid change as this proband had been reported previously but with different nucleotide change.[20] Sanger sequencing validation of this mutation was performed on the parents, and the results identified no variation at this site in the father and a heterozygous variation at this site in the mother (Fig. 4B and C). As XLP-1 inherited in a recessive pattern, the parents of the proband were all appeared healthy. Therefore, the diagnosis of XLP-1 was confirmed based on the proband’s typical clinical manifestations and genetic analysis results.\nIdentification of a hemizygous mutation c.96G > T in SH2D1A gene of the proband. (A) Schematic diagram of SH2D1A gene. The mutation site was highlighted in red. (B) Results of Sanger sequencing. The arrows indicated the sites of mutation. (C) Genetic pedigree of the family.\n[SUBTITLE] 3.3. The mutant SAP protein was highly pathogenic with structural and functional change [SUBSECTION] In order to explore whether the c.96G > T mutation in SH2D1A gene caused functional or structural deficiency in SAP protein, we first analyzed pathogenicity of mutant protein by PolyPhen-2 online scoring tool. As the greater the pathogenicity, the closer the score is to 1.0, the mutation was predicted to be highly pathogenic with a score of 1.0 (Fig. 5A). In addition, we analyzed the structural change of mutant SAP protein. As shown in Figure 5B, the secondary structure of SAP protein contained 2 α-helices and 8 β-strands and the mutation site was located at the end of the second β-strand. By comparing the structures of wild-type and mutant SAP proteins, we found that the substitution of amino acid Arginine to Serine causing a turnover of a hydrogen bond (Fig. 5B), suggesting the structure of SAP protein was changed. We also detected the expression of SAP in proband and the parents. As expected, flow cytometry detected that SAP protein was significantly downregulated in proband compared to which in parents. As shown in Figure 5C, SAP expression level in the proband, father and mother were 10.28%, 87.28%, and 80.31%, respectively, suggesting the function of SAP in proband was deficient. Overall, c.96G > T mutation of SH2D1A gene in the proband led to significant structural and functional deficiency of SAP protein, which was highly pathogenic.\nThe mutant SAP protein was highly pathogenic with structural and functional. (A) The pathogenicity of mutant SAP protein was predicted by PolyPhen-2. The greater the pathogenicity, the closer the score is to 1.0. (B) The secondary structure of SAP protein and a structural comparison between wild-type and mutant SAP protein. The site and labels of mutation were highlighted in magenta. The green dotted lines illustrated hydrogen bonds. The magenta arrows indicated the turnover of a hydrogen bond. (C) Expression of SAP in the patient and parents detected by flow cytometry. Peaks in red were negative controls. Peaks in green illustrated expression of SAP.\nIn order to explore whether the c.96G > T mutation in SH2D1A gene caused functional or structural deficiency in SAP protein, we first analyzed pathogenicity of mutant protein by PolyPhen-2 online scoring tool. As the greater the pathogenicity, the closer the score is to 1.0, the mutation was predicted to be highly pathogenic with a score of 1.0 (Fig. 5A). In addition, we analyzed the structural change of mutant SAP protein. As shown in Figure 5B, the secondary structure of SAP protein contained 2 α-helices and 8 β-strands and the mutation site was located at the end of the second β-strand. By comparing the structures of wild-type and mutant SAP proteins, we found that the substitution of amino acid Arginine to Serine causing a turnover of a hydrogen bond (Fig. 5B), suggesting the structure of SAP protein was changed. We also detected the expression of SAP in proband and the parents. As expected, flow cytometry detected that SAP protein was significantly downregulated in proband compared to which in parents. As shown in Figure 5C, SAP expression level in the proband, father and mother were 10.28%, 87.28%, and 80.31%, respectively, suggesting the function of SAP in proband was deficient. Overall, c.96G > T mutation of SH2D1A gene in the proband led to significant structural and functional deficiency of SAP protein, which was highly pathogenic.\nThe mutant SAP protein was highly pathogenic with structural and functional. (A) The pathogenicity of mutant SAP protein was predicted by PolyPhen-2. The greater the pathogenicity, the closer the score is to 1.0. (B) The secondary structure of SAP protein and a structural comparison between wild-type and mutant SAP protein. The site and labels of mutation were highlighted in magenta. The green dotted lines illustrated hydrogen bonds. The magenta arrows indicated the turnover of a hydrogen bond. (C) Expression of SAP in the patient and parents detected by flow cytometry. Peaks in red were negative controls. Peaks in green illustrated expression of SAP.\n[SUBTITLE] 3.4. Activation of PI3K-AKT-mTOR signaling pathway in the proband [SUBSECTION] As previously stated, PI3K downstream signaling pathways may be altered in XLP-1 patients, thus we evaluated the expression of PI3K subunits p110δ and p85α, as well as downstream key proteins including AKT, p-AKT and mTOR in both the proband and the parents. To ensure that observed differences were due to XLP-1 rather than HLH, we also evaluated the expression of proteins in a familial HLH patient and a healthy child. As shown in Figure 6, PI3K-AKT-mTOR pathway was inactivated in the healthy control, as evidenced by high expression of the negative regulatory subunit p85α, which inhibit the activity of the functional subunit p110δ. Moreover, under normal circumstances, the downstream protein AKT was unphosphorylated and mTOR was not expressed. The proband had p85α downregulation and p110δ upregulation, AKT phosphorylation, and mTOR activation, indicating that the PI3K-AKT-mTOR pathway was considerably activated. The expression patterns of p85α, p110δ, AKT, and p-AKT were identical in the father and mother, but mTOR expression was muted. Although p85α and p110δ were excessively high in the familial HLH control, downstream AKT was unphosphorylated and mTOR was only expressed, in contrast to the fully activated PI3K-AKT-mTOR pathway in the XLP-1 proband. Overall, the findings showed that the PI3K-AKT-mTOR signaling pathway was fully activated in XLP-1 patients, but it was inactive or only partially activated in healthy people or HLH patients, implying that the PI3K-AKT-mTOR signaling pathway may play a role in XLP-1 pathogenesis.\nExpression of key proteins in PI3K downstream pathways. Peaks in red were negative controls. Peaks in green illustrated expression of proteins. PI3K = phophatidylinositol-3-kinase.\nAs previously stated, PI3K downstream signaling pathways may be altered in XLP-1 patients, thus we evaluated the expression of PI3K subunits p110δ and p85α, as well as downstream key proteins including AKT, p-AKT and mTOR in both the proband and the parents. To ensure that observed differences were due to XLP-1 rather than HLH, we also evaluated the expression of proteins in a familial HLH patient and a healthy child. As shown in Figure 6, PI3K-AKT-mTOR pathway was inactivated in the healthy control, as evidenced by high expression of the negative regulatory subunit p85α, which inhibit the activity of the functional subunit p110δ. Moreover, under normal circumstances, the downstream protein AKT was unphosphorylated and mTOR was not expressed. The proband had p85α downregulation and p110δ upregulation, AKT phosphorylation, and mTOR activation, indicating that the PI3K-AKT-mTOR pathway was considerably activated. The expression patterns of p85α, p110δ, AKT, and p-AKT were identical in the father and mother, but mTOR expression was muted. Although p85α and p110δ were excessively high in the familial HLH control, downstream AKT was unphosphorylated and mTOR was only expressed, in contrast to the fully activated PI3K-AKT-mTOR pathway in the XLP-1 proband. Overall, the findings showed that the PI3K-AKT-mTOR signaling pathway was fully activated in XLP-1 patients, but it was inactive or only partially activated in healthy people or HLH patients, implying that the PI3K-AKT-mTOR signaling pathway may play a role in XLP-1 pathogenesis.\nExpression of key proteins in PI3K downstream pathways. Peaks in red were negative controls. Peaks in green illustrated expression of proteins. PI3K = phophatidylinositol-3-kinase.", "To access detailed conditions of the proband, multiple laboratory and imagological examinations were conducted during hospitalization. Results showed that peripheral blood EBV nucleic acid load continued to rise and the antibodies to EBV capsid antigen (EBVCA) IgG and IgG antibodies to EBV nuclear antigen became positive as the disease progressed (Fig. 1A). The percentage of CD19 + B cells was slightly decreased (Fig. 1B), and humoral immunity test indicated increased levels of serum IgG, IgM, IgA, and decreased C3 (Fig. 1C). Hematological parameters of the proband pre- and during-hospitalization showed that the counts of leukocyte, lymphocyte, neutrophil, monocyte, platelet, erythrocyte and hemoglobin were declined with fluctuation and generally lower than reference values. The percentages of lymphocyte, neutrophil and monocyte were all fluctuated outside the reference value (Fig. 1D). Besides, indicators for liver, renal and cardiac function of proband were abnormal as disease progressed. The levels of alanine aminotransferase, aspartate aminotransferase, γ-glutamyltransferase (γ-GGT), alkaline phosphatase, total bilirubin, direct bilirubin, total bile acid, lactate dehydrogenase and α-hydroxybutyrate dehydrogenase (α-HBDH) were all extremely higher than reference values (Fig. 1E–G). Additionally, the levels of ferritin and triglycerides elevated dramatically indicated the development of HLH (Fig. 2A). Occurrence of HLH was also confirmed by hemophagocytes in bone marrow (Fig. 2B) and increased expression of serum IL-6 and IL-10, with IL-10 being 12 times higher than the reference values (Fig. 2C). Abdominal color ultrasound revealed hepatomegaly, gallbladder wall thickening, mild splenomegaly and abdominal effusion (Fig. 3A). Neck color ultrasound revealed slightly enlarged lymph nodes on both sides of neck and jaw (Fig. 3B). Chest X-ray revealed increased bilateral pleural effusion with deterioration of disease (Fig. 3C). Overall, with the progression of disease, the condition of the proband kept deteriorating, especially the infection was aggravated, immunity responses were deficient, and multiple organs were progressively disabled. The clinical features including EBV-infection, bone marrow hemophagocytosis, dysgammaglobulinemia, hepatosplenomegaly and lymphadenopathy observed suggested that the proband may suffer from XLP.\nLaboratory examinations of the proband with the progression of disease. (A) Results of peripheral blood EBV nucleic acid and antibodies test. (B) Results of lymphocyte subsets test. (C) Results of humoral immunity test. (D) Results of whole blood counts. There were ten times of testing results during hospitalization listed in chronological order from left to right. (E) Results of liver function test. There were 10 times of testing results during hospitalization listed in chronological order from left to right. (F) Results of renal function test. There were 8 times of testing results during hospitalization listed in chronological order from left to right. (G) Results of cardiac function test. There were 7 times of testing results during hospitalization listed in chronological order from left to right. EBV = Epstein-Barr virus.\nOccurrence of hemophagocytic lymphohistiocytosis was detected in the proband. (A) Results of ferritin and triglycerides tests. There were 4 times of testing results during hospitalization listed in chronological order from left to right. (B) Results of cytopathology detection of bone marrow. (C) Results of serum cytokine test.\nImageological examinations of the proband with the progression of disease. (A) Results of abdominal color ultrasound. (B) Results of neck color ultrasound. (C) Results of chest X-ray. There were 5 times of examine results during hospitalization listed in chronological order from left to right.", "In order to confirm whether the proband was suffered from genetic disorders, the peripheral blood DNA of the proband and parents were extracted, and whole exome sequencing was performed. The results revealed a hemizygous mutation in exon 1 of SH2D1A gene, which substituted Guanine to Thymine at the site of nucleotide 96 (c.96G > T) (Fig. 4A), resulting in a missense substitution of Arginine to Serine at the site of amino acid 32 (p.R32S). According to the ACMG guidelines, this mutation was identified as a pathogenic mutation. It was located in the mutation hotspot region with a low-frequency in the normal population database. The mutation with the same amino acid change as this proband had been reported previously but with different nucleotide change.[20] Sanger sequencing validation of this mutation was performed on the parents, and the results identified no variation at this site in the father and a heterozygous variation at this site in the mother (Fig. 4B and C). As XLP-1 inherited in a recessive pattern, the parents of the proband were all appeared healthy. Therefore, the diagnosis of XLP-1 was confirmed based on the proband’s typical clinical manifestations and genetic analysis results.\nIdentification of a hemizygous mutation c.96G > T in SH2D1A gene of the proband. (A) Schematic diagram of SH2D1A gene. The mutation site was highlighted in red. (B) Results of Sanger sequencing. The arrows indicated the sites of mutation. (C) Genetic pedigree of the family.", "In order to explore whether the c.96G > T mutation in SH2D1A gene caused functional or structural deficiency in SAP protein, we first analyzed pathogenicity of mutant protein by PolyPhen-2 online scoring tool. As the greater the pathogenicity, the closer the score is to 1.0, the mutation was predicted to be highly pathogenic with a score of 1.0 (Fig. 5A). In addition, we analyzed the structural change of mutant SAP protein. As shown in Figure 5B, the secondary structure of SAP protein contained 2 α-helices and 8 β-strands and the mutation site was located at the end of the second β-strand. By comparing the structures of wild-type and mutant SAP proteins, we found that the substitution of amino acid Arginine to Serine causing a turnover of a hydrogen bond (Fig. 5B), suggesting the structure of SAP protein was changed. We also detected the expression of SAP in proband and the parents. As expected, flow cytometry detected that SAP protein was significantly downregulated in proband compared to which in parents. As shown in Figure 5C, SAP expression level in the proband, father and mother were 10.28%, 87.28%, and 80.31%, respectively, suggesting the function of SAP in proband was deficient. Overall, c.96G > T mutation of SH2D1A gene in the proband led to significant structural and functional deficiency of SAP protein, which was highly pathogenic.\nThe mutant SAP protein was highly pathogenic with structural and functional. (A) The pathogenicity of mutant SAP protein was predicted by PolyPhen-2. The greater the pathogenicity, the closer the score is to 1.0. (B) The secondary structure of SAP protein and a structural comparison between wild-type and mutant SAP protein. The site and labels of mutation were highlighted in magenta. The green dotted lines illustrated hydrogen bonds. The magenta arrows indicated the turnover of a hydrogen bond. (C) Expression of SAP in the patient and parents detected by flow cytometry. Peaks in red were negative controls. Peaks in green illustrated expression of SAP.", "As previously stated, PI3K downstream signaling pathways may be altered in XLP-1 patients, thus we evaluated the expression of PI3K subunits p110δ and p85α, as well as downstream key proteins including AKT, p-AKT and mTOR in both the proband and the parents. To ensure that observed differences were due to XLP-1 rather than HLH, we also evaluated the expression of proteins in a familial HLH patient and a healthy child. As shown in Figure 6, PI3K-AKT-mTOR pathway was inactivated in the healthy control, as evidenced by high expression of the negative regulatory subunit p85α, which inhibit the activity of the functional subunit p110δ. Moreover, under normal circumstances, the downstream protein AKT was unphosphorylated and mTOR was not expressed. The proband had p85α downregulation and p110δ upregulation, AKT phosphorylation, and mTOR activation, indicating that the PI3K-AKT-mTOR pathway was considerably activated. The expression patterns of p85α, p110δ, AKT, and p-AKT were identical in the father and mother, but mTOR expression was muted. Although p85α and p110δ were excessively high in the familial HLH control, downstream AKT was unphosphorylated and mTOR was only expressed, in contrast to the fully activated PI3K-AKT-mTOR pathway in the XLP-1 proband. Overall, the findings showed that the PI3K-AKT-mTOR signaling pathway was fully activated in XLP-1 patients, but it was inactive or only partially activated in healthy people or HLH patients, implying that the PI3K-AKT-mTOR signaling pathway may play a role in XLP-1 pathogenesis.\nExpression of key proteins in PI3K downstream pathways. Peaks in red were negative controls. Peaks in green illustrated expression of proteins. PI3K = phophatidylinositol-3-kinase.", "Due to complex clinical characteristics, XLP-1 is easily confused with other diseases such as jaundice, hyperbilirubinemia, hypogammaglobulinemia, and lymphoma at the time of diagnosis. Through whole exome sequencing, we identified a hemizygous mutation c.96G > T (p.R32S) in exon 1 of SH2D1A gene in the proband of our study, which was inherited from the mother who was a healthy heterozygous carrier of this mutation. Although the sequencing technology greatly assists in the clinical diagnosis and classification of XLP, there is an urgent need for a way to rapidly diagnose XLP due to the acute onset and rapid progression of XLP, especially in cases accompanied by life-threatened HLH. Flow cytometry can be used to quickly detect the expression of SAP or XIAP protein and can also be used to identify the phenotypic and functional deficiency characteristics of lymphocytes in XLP patients, supporting the rapid screening need of XLP. Rapid detection by flow cytometry allows for more treatment time for critical XLP patients and more preparation time for patients suitable for HSCT, which will help improve XLP survival. In this study, we confirmed the SAP protein deficiency in the proband and analyzed the characteristics of peripheral blood lymphocyte subsets via flow cytometry. Based on the clinical manifestations, sequencing and protein expression findings of the proband, we confirmed the diagnosis of XLP-1. Apart from this, flow cytometry can also be used to detect the level of NKT cells which can assist in XLP-1 diagnosis. Previous studies have found that iNKT cells have a constant TCRα receptor and they cannot develop without SAP protein, thus iNKT cells will not be detected in XLP-1 patients when SAP protein is deficient.[21,22] Additionally, iNKT can be labeled with CD3, TCRVα24 and TCRVβ11 antibodies that offer convenience in the detection.[23] Overall, combination of sequencing technology and flow cytometry provides a more efficient and accurate way in the diagnosis and classification of XLP.\nSAP protein is an adaptor molecule, which competitively binds to the intracellular region of the SLAM receptor proteins such as 2B4 (SLAMF4) and Ly108 (NTB-A or SLAMF6).[24] The SLAM proteins are particularly important in the generation of cytotoxic effects. In the lack of SAP, the SLAM proteins play inhibitory roles by binding to their ligands and recruiting phosphatases including SHP-1, SHP-2 and SHIP1.[25] Those inhibitory activities are easily triggered in B cells since high levels of SLAM proteins and their ligands are expressed in B cells.[19] Previous research found that the typical cytological characteristics of XLP-1 are functional deficiencies in CTLs and NK cells, which preventing them from killing EBV-infected B cells but retaining the ability to kill many other targets.[16] Moreover, T cell restimulation-induced cell death was disabled possibly due to the inhibitory effect produced by SLAM proteins and their ligands in XLP-1 patients, hence catastrophic lymphoproliferation and HLH were very likely to be induced in the presence of EBV infection.[26] The pathogenic mechanisms of XLP-1 have not been fully understood so far.\nInterestingly, we found that the PI3K-AKT-mTOR signaling pathway was fully activated in XLP-1 patients, but it was inactive or only partially activated in healthy people or HLH patients. It is known that PI3K signaling pathways are closely related to lymphocyte development and differentiation.[27] In congenital PI3K deficiency disorders such as activated PI3Kδ syndrome, its over-activation can block B cell development and defect class switching of antibodies, resulting in the production of low affinity antibodies and high expression of IgM.[28] In this study, the proportion of CD19 + B cells of the proband was slightly lower than the reference value, suggesting that the differentiation and amplification of mature B cells may be defective after the activation of PI3K-AKT-mTOR pathway triggered by SH2D1A gene mutation. Meanwhile, the IgM level of the patient was significantly higher than the normal reference value, indicating activation of PI3K-AKT-mTOR signaling pathway might be the potential pathogenic mechanisms of XLP-1. In general, phosphorylation of AKT can directly cause the activation of mTOR, but in this study, we first reported that mTOR was inactivated though PI3K and AKT were activated in the parents of proband with healthy phenotype. The specific reason behind this is unknown yet, there were published studies indicated that mTOR may be activated independently of PI3K in both T and B cells,[29] which might explain the inactivation of mTOR. In addition, studies of PI3K mutant mouse models have shown that at the absence of p85α or p110δ, GC formation was impaired, proliferation and differentiation of mature B cells was decreased, and humoral immunity showed a cliff decline.[18] As for B cell subsets, the proportion of follicular B cells was less than 50%, while proportions of marginal zone B and B1 cells were increased sharply.[30] Follicular B cells are mainly developed into plasma cells, which can produce a large number of antibodies with high affinity. Marginal zone B cells can quickly recognize T1 and TD antigens through their surface BCR and produce antibodies with low affinity. B1 cells contribute to autoantibody reactions and antibody polyreactions and can also produce a large number of antibodies with low affinity including natural antibody IgM and mucosal antibody IgA. Overall, excessive activation of PI3K signal can inhibit the proliferation and differentiation of B cells and lead to the decrease of humoral immunity, which may be the potential reason for the onset of disease in our study. The molecular pathogenic mechanisms of XLP-1 may involve other types of lymphocytes and their internal signaling pathways which needs to be explored by more studies. Also, the correlation and interaction patterns of SAP and PI3K pathways still need to be clarified by further studies.\nXLP-1 presents in patients at an average age of 2.5 years, but the proband in this study was only 7 months old at onset. Such a young age with severe liver injury and HLH was very rare in previously reported XLP-1 cases, thus greatly increasing the difficulty of diagnosis and treatment. The only effective treatment for XLP-1 at present is HSCT. However, HSCT treatment requires fulfillment of strict matching and screening criteria and comes with a high surgical risk, so it is not suitable for all XLP-1 patients. In addition to HSCT, gene therapy has developed rapidly in recent years in animal models and is expected to be applied in the treatment of human gene-deficient diseases including XLP-1 in the future.", "In this study, we provided a detailed description of the clinical features of an XLP-1 patient and detected that the proband was caused by a hemizygous mutation c.96G > T in SH2D1A gene resulting in a missense substitution of Arginine to Serine (p.R32S). The mutant protein contained a hydrogen bond turnover at the site of mutation and the mutation resulted in downregulation of SH2D1A encoded protein SAP. The PI3K-AKT-mTOR signaling pathway was fully activated in XLP-1 patients, but it was inactive or only partially activated in healthy people or HLH patients, implying that the PI3K-AKT-mTOR signaling pathway may play a role in XLP-1 pathogenesis. This study would be helpful for subsequent research related to the diagnosis and pathogenesis of XLP-1.", "The authors sincerely thank the patient and his family members for their participation and support.\nYW, YW, WL, and LT contributed equally to this work.", "Conceptualization: Yanchun Wang, Yan Wang, Weimin Lu, Lvyan Tao.\nData curation: Yang Xiao, Yuantao Zhou, Xiaoli He.\nFormal analysis: Yanchun Wang, Yan Wang, Weimin Lu, Lvyan Tao.\nInvestigation: Yang Xiao, Yuantao Zhou, Xiaoli He.\nMethodology: Yanchun Wang, Yan Wang, Weimin Lu, Lvyan Tao.\nResources: Yanchun Wang, Yan Wang, Weimin Lu, Lvyan Tao.\nSupervision: Yu Zhang, Li Li.\nVisualization: Yanchun Wang, Yan Wang, Weimin Lu, Lvyan Tao.\nWriting – original draft: Yu Zhang, Li Li.\nWriting – review & editing: Yu Zhang, Li Li." ]
[ "intro", "methods", null, null, null, null, null, "results", null, null, null, null, "discussion", "conclusions", null, null ]
[ "AKT", "mTOR", "PI3K", "SAP", "\nSH2D1A\n", "X-linked lymphoproliferative syndrome" ]
Effectiveness and safety of Tai Chi for anxiety disorder of COVID-19: A protocol of systematic review and meta-analysis.
36254045
Anxiety disorders pose a significant threat to the clinical rehabilitation of patients with coronavirus disease 2019 (COVID-19). Tai Chi is a therapeutic exercise that can be used to treat anxiety disorders. We aim to conduct a systematic review and meta-analysis to evaluate the effectiveness and safety of Tai Chi for treating patients with anxiety disorders caused by COVID-19.
BACKGROUND
The PubMed, EMBASE, Cochrane Library, China National Knowledge Infrastructure, Chinese Biomedical Literature, Wan Fang, and Chinese Clinical Trial Registry databases will be searched for reports of randomized controlled trials on Tai Chi for the treatment of anxiety disorders caused by COVID-19, published from December 1, 2019, to August 22, 2022. Two researchers will screen the articles and extract the relevant information.
METHODS
The results will provide a systematic overview of the current evidence on the use of Tai Chi to treat anxiety disorders caused by COVID-19 among patients.
RESULTS
The conclusions of this study will help clarify whether Tai Chi is effective and safe for treating anxiety disorders caused by COVID-19.
CONCLUSION
[ "Anxiety Disorders", "COVID-19", "China", "Humans", "Meta-Analysis as Topic", "Systematic Reviews as Topic", "Tai Ji" ]
9575401
1. Introduction
The COVID-19 pandemic is a major global public health challenge. Since the outbreak of COVID-19 in Wuhan, China, in December 2019, the number of confirmed cases and deaths due to COVID-19 worldwide has exceeded 525 million and 6.2 million (as of August 2022), respectively, and continues to increase.[1] The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes COVID-19, is a novel virus; thus, the general population is highly susceptible and has no specific immunity against it. In addition, SARS-CoV-2 is mutating frequently and rapidly, making the development of established treatment regimens for COVID-19 challenging. Alpha, beta, gamma, delta,[2] and omicron variants of SARS-CoV-2 have been identified so far.[3] Therefore, although the existing therapeutic regimens for COVID-19 generally alleviate the symptoms of the disease, the rapid and frequent development of new variants of SARS-CoV-2[4,5] results in the recurrence of COVID-19. The survey of patients with COVID-19 showed that more than 40% of the respondents experienced some degree of anxiety and that 18.6% of those who experienced anxiety also had moderate to severe depression or stress.[6] Patients with COVID-19 generally show signs of emotional distress, including anxiety,[7] memory and attention loss, sleep disorders, communication disorders, post-traumatic stress disorder, and other mental health problems.[8] Regarding social activities, patients with COVID-19 often experience anxiety due to the adverse effects of the COVID-19 pandemic, such as reduced social contact, loneliness, incomplete recovery of physical health, or unemployment caused by the disease.[9] Post-COVID-19 anxiety disorder is a health condition that usually requires long-term treatment, which has a long-term negative impact on the physiological and psychological health of patients.[10] With the steady increase in the number of patients with COVID-19, the negative impact of post-COVID-19 anxiety disorders on individuals, families, and the society is emerging,[11] and has become a health problem that cannot be ignored. Thus, post-COVID-19 anxiety disorder has become a hot topic in the COVID-19 discourse globally. It is important to understand the symptoms and effects of anxiety disorders in patients after COVID-19 to reduce and avoid negative effects and improve symptom management in such patients. Research has shown that Tai Chi can be used to treat anxiety, depression, traumatic stress disorders, and other mental health conditions.[12,13] Tai Chi is characterized by soft movements, concentrated thoughts, command of movements with thoughts, coordinated breathing, and control of hand-eye movements with breathing. It requires coordination of the hands, feet, head, and eyes, and internal and external cultivation.[14] Tai Chi cultivates the mind; seeks harmony between man and nature; and has a good effect on the adjustment of the states of the respiratory system, nervous system, and internal organs.[15,16] It focuses on the internal essence and spirit, thus playing a role in cultivating the mind, relieving anxiety and depression,[17] and promoting a healthy state of mind. Therefore, we will perform a systematic review and meta-analysis to explore the effectiveness and safety of Tai Chi in treating anxiety disorders caused by COVID-19 to provide a reference for treating health crises caused by post-COVID-19 anxiety disorders.
2. Methods
[SUBTITLE] 2.1. Protocol registration [SUBSECTION] This research program was registered in PROSPERO (registration number: CRD42022320766). The protocol report was based on the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols 2015 statement.[18] This research program was registered in PROSPERO (registration number: CRD42022320766). The protocol report was based on the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols 2015 statement.[18] [SUBTITLE] 2.2. Inclusion criteria [SUBSECTION] [SUBTITLE] 2.2.1. Participants. [SUBSECTION] COVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants. COVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants. [SUBTITLE] 2.2.2. Interventions. [SUBSECTION] The experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi. The experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi. [SUBTITLE] 2.2.3. Outcomes. [SUBSECTION] The primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22] Data on adverse events and the safety of the included studies will be utilized for safety analysis. The primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22] Data on adverse events and the safety of the included studies will be utilized for safety analysis. [SUBTITLE] 2.2.4. Studies. [SUBSECTION] We will include randomized controlled trials (RCTs). We will include randomized controlled trials (RCTs). [SUBTITLE] 2.2.1. Participants. [SUBSECTION] COVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants. COVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants. [SUBTITLE] 2.2.2. Interventions. [SUBSECTION] The experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi. The experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi. [SUBTITLE] 2.2.3. Outcomes. [SUBSECTION] The primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22] Data on adverse events and the safety of the included studies will be utilized for safety analysis. The primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22] Data on adverse events and the safety of the included studies will be utilized for safety analysis. [SUBTITLE] 2.2.4. Studies. [SUBSECTION] We will include randomized controlled trials (RCTs). We will include randomized controlled trials (RCTs). [SUBTITLE] 2.3. Search strategy [SUBSECTION] We will search the electronic databases of PubMed, EMBASE, Cochrane Library, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, Wan Fang, and the Chinese Clinical Trial Registry for RCTs on Tai Chi for the treatment of anxiety disorders caused by COVID-19. Relevant articles published from December 1, 2019, to August 22, 2022, will be included. The search strategy for PubMed is presented in Table 1. PubMed search strategy. COVID-19 = coronavirus disease 2019. We will search the electronic databases of PubMed, EMBASE, Cochrane Library, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, Wan Fang, and the Chinese Clinical Trial Registry for RCTs on Tai Chi for the treatment of anxiety disorders caused by COVID-19. Relevant articles published from December 1, 2019, to August 22, 2022, will be included. The search strategy for PubMed is presented in Table 1. PubMed search strategy. COVID-19 = coronavirus disease 2019. [SUBTITLE] 2.4. Selections of studies [SUBSECTION] Two reviewers (S-QZ and XY) will independently review and screen the studies according to the inclusion and exclusion criteria of the review. First, the reviewers will utilize EndNote X9 software to exclude duplicate articles. Thereafter, they will exclude obviously irrelevant literature by reading the titles and abstracts of the articles. They will then screen the remaining articles by reading their full texts and provide the reasons for excluding ineligible studies. The selection process is shown in Figure 1. The study selection process. Two reviewers (S-QZ and XY) will independently review and screen the studies according to the inclusion and exclusion criteria of the review. First, the reviewers will utilize EndNote X9 software to exclude duplicate articles. Thereafter, they will exclude obviously irrelevant literature by reading the titles and abstracts of the articles. They will then screen the remaining articles by reading their full texts and provide the reasons for excluding ineligible studies. The selection process is shown in Figure 1. The study selection process. [SUBTITLE] 2.5. Data collection and management [SUBSECTION] Two reviewers (H-YL and X-YM) will independently screen the literature and extract the data from the included studies. The collected data will include the information of each study (year of publication, first author, sample size, age and sex of participants, course of the disease, intervention, control, course of treatment, primary outcome, and secondary outcome) and its results (results of outcomes, adverse events, and safety). If there is any disagreement between the two reviewers during the screening, they will consult a third reviewer (QT) for the final decision. Two reviewers (H-YL and X-YM) will independently screen the literature and extract the data from the included studies. The collected data will include the information of each study (year of publication, first author, sample size, age and sex of participants, course of the disease, intervention, control, course of treatment, primary outcome, and secondary outcome) and its results (results of outcomes, adverse events, and safety). If there is any disagreement between the two reviewers during the screening, they will consult a third reviewer (QT) for the final decision. [SUBTITLE] 2.6. Assessment of risk of bias [SUBSECTION] Two researchers (R-YL and R-YW) will separately evaluate the methodological qualities of the literature using the Cochrane Collaboration’s Risk of Bias tool, which includes 7 items. The risk of bias in each aspect will be assessed, and the results will be categorized into three grades: low risk, unclear risk, and high risk. Two researchers (R-YL and R-YW) will separately evaluate the methodological qualities of the literature using the Cochrane Collaboration’s Risk of Bias tool, which includes 7 items. The risk of bias in each aspect will be assessed, and the results will be categorized into three grades: low risk, unclear risk, and high risk. [SUBTITLE] 2.7. Data syntheses [SUBSECTION] [SUBTITLE] 2.7.1. Data synthesis and assessment of heterogeneity. [SUBSECTION] RevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity. RevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity. [SUBTITLE] 2.7.2. Assessment of reporting bias. [SUBSECTION] We will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias. We will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias. [SUBTITLE] 2.7.3. Subgroup analysis. [SUBSECTION] If significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi. If significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi. [SUBTITLE] 2.7.4. Sensitivity analysis. [SUBSECTION] We will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data. We will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data. [SUBTITLE] 2.7.5. Grading the quality of evidence. [SUBSECTION] We will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence. We will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence. [SUBTITLE] 2.7.6. Dealing with missing data. [SUBSECTION] We will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed. We will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed. [SUBTITLE] 2.7.7. Ethics and dissemination. [SUBSECTION] No ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal. No ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal. [SUBTITLE] 2.7.1. Data synthesis and assessment of heterogeneity. [SUBSECTION] RevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity. RevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity. [SUBTITLE] 2.7.2. Assessment of reporting bias. [SUBSECTION] We will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias. We will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias. [SUBTITLE] 2.7.3. Subgroup analysis. [SUBSECTION] If significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi. If significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi. [SUBTITLE] 2.7.4. Sensitivity analysis. [SUBSECTION] We will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data. We will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data. [SUBTITLE] 2.7.5. Grading the quality of evidence. [SUBSECTION] We will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence. We will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence. [SUBTITLE] 2.7.6. Dealing with missing data. [SUBSECTION] We will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed. We will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed. [SUBTITLE] 2.7.7. Ethics and dissemination. [SUBSECTION] No ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal. No ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal.
null
null
null
null
[ "2.1. Protocol registration", "2.2. Inclusion criteria", "2.2.1. Participants.", "2.2.2. Interventions.", "2.2.3. Outcomes.", "2.2.4. Studies.", "2.3. Search strategy", "2.4. Selections of studies", "2.5. Data collection and management", "2.6. Assessment of risk of bias", "2.7. Data syntheses", "2.7.1. Data synthesis and assessment of heterogeneity.", "2.7.2. Assessment of reporting bias.", "2.7.3. Subgroup analysis.", "2.7.4. Sensitivity analysis.", "2.7.5. Grading the quality of evidence.", "2.7.6. Dealing with missing data.", "2.7.7. Ethics and dissemination.", "Author contributions" ]
[ "This research program was registered in PROSPERO (registration number: CRD42022320766). The protocol report was based on the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols 2015 statement.[18]", "[SUBTITLE] 2.2.1. Participants. [SUBSECTION] COVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants.\nCOVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants.\n[SUBTITLE] 2.2.2. Interventions. [SUBSECTION] The experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi.\nThe experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi.\n[SUBTITLE] 2.2.3. Outcomes. [SUBSECTION] The primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22]\nData on adverse events and the safety of the included studies will be utilized for safety analysis.\nThe primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22]\nData on adverse events and the safety of the included studies will be utilized for safety analysis.\n[SUBTITLE] 2.2.4. Studies. [SUBSECTION] We will include randomized controlled trials (RCTs).\nWe will include randomized controlled trials (RCTs).", "COVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants.", "The experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi.", "The primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22]\nData on adverse events and the safety of the included studies will be utilized for safety analysis.", "We will include randomized controlled trials (RCTs).", "We will search the electronic databases of PubMed, EMBASE, Cochrane Library, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, Wan Fang, and the Chinese Clinical Trial Registry for RCTs on Tai Chi for the treatment of anxiety disorders caused by COVID-19. Relevant articles published from December 1, 2019, to August 22, 2022, will be included. The search strategy for PubMed is presented in Table 1.\nPubMed search strategy.\nCOVID-19 = coronavirus disease 2019.", "Two reviewers (S-QZ and XY) will independently review and screen the studies according to the inclusion and exclusion criteria of the review. First, the reviewers will utilize EndNote X9 software to exclude duplicate articles. Thereafter, they will exclude obviously irrelevant literature by reading the titles and abstracts of the articles. They will then screen the remaining articles by reading their full texts and provide the reasons for excluding ineligible studies. The selection process is shown in Figure 1.\nThe study selection process.", "Two reviewers (H-YL and X-YM) will independently screen the literature and extract the data from the included studies. The collected data will include the information of each study (year of publication, first author, sample size, age and sex of participants, course of the disease, intervention, control, course of treatment, primary outcome, and secondary outcome) and its results (results of outcomes, adverse events, and safety). If there is any disagreement between the two reviewers during the screening, they will consult a third reviewer (QT) for the final decision.", "Two researchers (R-YL and R-YW) will separately evaluate the methodological qualities of the literature using the Cochrane Collaboration’s Risk of Bias tool, which includes 7 items. The risk of bias in each aspect will be assessed, and the results will be categorized into three grades: low risk, unclear risk, and high risk.", "[SUBTITLE] 2.7.1. Data synthesis and assessment of heterogeneity. [SUBSECTION] RevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity.\nRevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity.\n[SUBTITLE] 2.7.2. Assessment of reporting bias. [SUBSECTION] We will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias.\nWe will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias.\n[SUBTITLE] 2.7.3. Subgroup analysis. [SUBSECTION] If significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi.\nIf significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi.\n[SUBTITLE] 2.7.4. Sensitivity analysis. [SUBSECTION] We will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data.\nWe will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data.\n[SUBTITLE] 2.7.5. Grading the quality of evidence. [SUBSECTION] We will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence.\nWe will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence.\n[SUBTITLE] 2.7.6. Dealing with missing data. [SUBSECTION] We will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed.\nWe will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed.\n[SUBTITLE] 2.7.7. Ethics and dissemination. [SUBSECTION] No ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal.\nNo ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal.", "RevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity.", "We will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias.", "If significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi.", "We will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data.", "We will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence.", "We will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed.", "No ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal.", "Assessment of risk bias: Runyu Liang, Ruoyu Wang.\nConceptualization: Shiqiang Zhang, Luwen Zhu.\nData curation: Hongyu Li, Xiyuan Ma.\nFormal analysis: Shiqiang Zhang, Hongyu Li.\nFunding: Luwen Zhu.\nInvestigation: Shiqiang Zhang, Xia Yin.\nProject administration: Qiang Tang.\nResources: Shiqiang Zhang.\nSoftware: Shiqiang Zhang.\nWriting – original draft: Hongyu Li, Shiqiang Zhang.\nWriting – review & editing: Hongyu Li, Qiang Tang." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Methods", "2.1. Protocol registration", "2.2. Inclusion criteria", "2.2.1. Participants.", "2.2.2. Interventions.", "2.2.3. Outcomes.", "2.2.4. Studies.", "2.3. Search strategy", "2.4. Selections of studies", "2.5. Data collection and management", "2.6. Assessment of risk of bias", "2.7. Data syntheses", "2.7.1. Data synthesis and assessment of heterogeneity.", "2.7.2. Assessment of reporting bias.", "2.7.3. Subgroup analysis.", "2.7.4. Sensitivity analysis.", "2.7.5. Grading the quality of evidence.", "2.7.6. Dealing with missing data.", "2.7.7. Ethics and dissemination.", "3. Discussion", "Author contributions" ]
[ "The COVID-19 pandemic is a major global public health challenge. Since the outbreak of COVID-19 in Wuhan, China, in December 2019, the number of confirmed cases and deaths due to COVID-19 worldwide has exceeded 525 million and 6.2 million (as of August 2022), respectively, and continues to increase.[1] The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes COVID-19, is a novel virus; thus, the general population is highly susceptible and has no specific immunity against it. In addition, SARS-CoV-2 is mutating frequently and rapidly, making the development of established treatment regimens for COVID-19 challenging. Alpha, beta, gamma, delta,[2] and omicron variants of SARS-CoV-2 have been identified so far.[3] Therefore, although the existing therapeutic regimens for COVID-19 generally alleviate the symptoms of the disease, the rapid and frequent development of new variants of SARS-CoV-2[4,5] results in the recurrence of COVID-19.\nThe survey of patients with COVID-19 showed that more than 40% of the respondents experienced some degree of anxiety and that 18.6% of those who experienced anxiety also had moderate to severe depression or stress.[6] Patients with COVID-19 generally show signs of emotional distress, including anxiety,[7] memory and attention loss, sleep disorders, communication disorders, post-traumatic stress disorder, and other mental health problems.[8] Regarding social activities, patients with COVID-19 often experience anxiety due to the adverse effects of the COVID-19 pandemic, such as reduced social contact, loneliness, incomplete recovery of physical health, or unemployment caused by the disease.[9]\nPost-COVID-19 anxiety disorder is a health condition that usually requires long-term treatment, which has a long-term negative impact on the physiological and psychological health of patients.[10] With the steady increase in the number of patients with COVID-19, the negative impact of post-COVID-19 anxiety disorders on individuals, families, and the society is emerging,[11] and has become a health problem that cannot be ignored. Thus, post-COVID-19 anxiety disorder has become a hot topic in the COVID-19 discourse globally.\nIt is important to understand the symptoms and effects of anxiety disorders in patients after COVID-19 to reduce and avoid negative effects and improve symptom management in such patients. Research has shown that Tai Chi can be used to treat anxiety, depression, traumatic stress disorders, and other mental health conditions.[12,13] Tai Chi is characterized by soft movements, concentrated thoughts, command of movements with thoughts, coordinated breathing, and control of hand-eye movements with breathing. It requires coordination of the hands, feet, head, and eyes, and internal and external cultivation.[14] Tai Chi cultivates the mind; seeks harmony between man and nature; and has a good effect on the adjustment of the states of the respiratory system, nervous system, and internal organs.[15,16] It focuses on the internal essence and spirit, thus playing a role in cultivating the mind, relieving anxiety and depression,[17] and promoting a healthy state of mind. Therefore, we will perform a systematic review and meta-analysis to explore the effectiveness and safety of Tai Chi in treating anxiety disorders caused by COVID-19 to provide a reference for treating health crises caused by post-COVID-19 anxiety disorders.", "[SUBTITLE] 2.1. Protocol registration [SUBSECTION] This research program was registered in PROSPERO (registration number: CRD42022320766). The protocol report was based on the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols 2015 statement.[18]\nThis research program was registered in PROSPERO (registration number: CRD42022320766). The protocol report was based on the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols 2015 statement.[18]\n[SUBTITLE] 2.2. Inclusion criteria [SUBSECTION] [SUBTITLE] 2.2.1. Participants. [SUBSECTION] COVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants.\nCOVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants.\n[SUBTITLE] 2.2.2. Interventions. [SUBSECTION] The experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi.\nThe experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi.\n[SUBTITLE] 2.2.3. Outcomes. [SUBSECTION] The primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22]\nData on adverse events and the safety of the included studies will be utilized for safety analysis.\nThe primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22]\nData on adverse events and the safety of the included studies will be utilized for safety analysis.\n[SUBTITLE] 2.2.4. Studies. [SUBSECTION] We will include randomized controlled trials (RCTs).\nWe will include randomized controlled trials (RCTs).\n[SUBTITLE] 2.2.1. Participants. [SUBSECTION] COVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants.\nCOVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants.\n[SUBTITLE] 2.2.2. Interventions. [SUBSECTION] The experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi.\nThe experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi.\n[SUBTITLE] 2.2.3. Outcomes. [SUBSECTION] The primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22]\nData on adverse events and the safety of the included studies will be utilized for safety analysis.\nThe primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22]\nData on adverse events and the safety of the included studies will be utilized for safety analysis.\n[SUBTITLE] 2.2.4. Studies. [SUBSECTION] We will include randomized controlled trials (RCTs).\nWe will include randomized controlled trials (RCTs).\n[SUBTITLE] 2.3. Search strategy [SUBSECTION] We will search the electronic databases of PubMed, EMBASE, Cochrane Library, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, Wan Fang, and the Chinese Clinical Trial Registry for RCTs on Tai Chi for the treatment of anxiety disorders caused by COVID-19. Relevant articles published from December 1, 2019, to August 22, 2022, will be included. The search strategy for PubMed is presented in Table 1.\nPubMed search strategy.\nCOVID-19 = coronavirus disease 2019.\nWe will search the electronic databases of PubMed, EMBASE, Cochrane Library, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, Wan Fang, and the Chinese Clinical Trial Registry for RCTs on Tai Chi for the treatment of anxiety disorders caused by COVID-19. Relevant articles published from December 1, 2019, to August 22, 2022, will be included. The search strategy for PubMed is presented in Table 1.\nPubMed search strategy.\nCOVID-19 = coronavirus disease 2019.\n[SUBTITLE] 2.4. Selections of studies [SUBSECTION] Two reviewers (S-QZ and XY) will independently review and screen the studies according to the inclusion and exclusion criteria of the review. First, the reviewers will utilize EndNote X9 software to exclude duplicate articles. Thereafter, they will exclude obviously irrelevant literature by reading the titles and abstracts of the articles. They will then screen the remaining articles by reading their full texts and provide the reasons for excluding ineligible studies. The selection process is shown in Figure 1.\nThe study selection process.\nTwo reviewers (S-QZ and XY) will independently review and screen the studies according to the inclusion and exclusion criteria of the review. First, the reviewers will utilize EndNote X9 software to exclude duplicate articles. Thereafter, they will exclude obviously irrelevant literature by reading the titles and abstracts of the articles. They will then screen the remaining articles by reading their full texts and provide the reasons for excluding ineligible studies. The selection process is shown in Figure 1.\nThe study selection process.\n[SUBTITLE] 2.5. Data collection and management [SUBSECTION] Two reviewers (H-YL and X-YM) will independently screen the literature and extract the data from the included studies. The collected data will include the information of each study (year of publication, first author, sample size, age and sex of participants, course of the disease, intervention, control, course of treatment, primary outcome, and secondary outcome) and its results (results of outcomes, adverse events, and safety). If there is any disagreement between the two reviewers during the screening, they will consult a third reviewer (QT) for the final decision.\nTwo reviewers (H-YL and X-YM) will independently screen the literature and extract the data from the included studies. The collected data will include the information of each study (year of publication, first author, sample size, age and sex of participants, course of the disease, intervention, control, course of treatment, primary outcome, and secondary outcome) and its results (results of outcomes, adverse events, and safety). If there is any disagreement between the two reviewers during the screening, they will consult a third reviewer (QT) for the final decision.\n[SUBTITLE] 2.6. Assessment of risk of bias [SUBSECTION] Two researchers (R-YL and R-YW) will separately evaluate the methodological qualities of the literature using the Cochrane Collaboration’s Risk of Bias tool, which includes 7 items. The risk of bias in each aspect will be assessed, and the results will be categorized into three grades: low risk, unclear risk, and high risk.\nTwo researchers (R-YL and R-YW) will separately evaluate the methodological qualities of the literature using the Cochrane Collaboration’s Risk of Bias tool, which includes 7 items. The risk of bias in each aspect will be assessed, and the results will be categorized into three grades: low risk, unclear risk, and high risk.\n[SUBTITLE] 2.7. Data syntheses [SUBSECTION] [SUBTITLE] 2.7.1. Data synthesis and assessment of heterogeneity. [SUBSECTION] RevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity.\nRevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity.\n[SUBTITLE] 2.7.2. Assessment of reporting bias. [SUBSECTION] We will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias.\nWe will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias.\n[SUBTITLE] 2.7.3. Subgroup analysis. [SUBSECTION] If significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi.\nIf significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi.\n[SUBTITLE] 2.7.4. Sensitivity analysis. [SUBSECTION] We will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data.\nWe will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data.\n[SUBTITLE] 2.7.5. Grading the quality of evidence. [SUBSECTION] We will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence.\nWe will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence.\n[SUBTITLE] 2.7.6. Dealing with missing data. [SUBSECTION] We will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed.\nWe will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed.\n[SUBTITLE] 2.7.7. Ethics and dissemination. [SUBSECTION] No ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal.\nNo ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal.\n[SUBTITLE] 2.7.1. Data synthesis and assessment of heterogeneity. [SUBSECTION] RevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity.\nRevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity.\n[SUBTITLE] 2.7.2. Assessment of reporting bias. [SUBSECTION] We will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias.\nWe will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias.\n[SUBTITLE] 2.7.3. Subgroup analysis. [SUBSECTION] If significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi.\nIf significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi.\n[SUBTITLE] 2.7.4. Sensitivity analysis. [SUBSECTION] We will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data.\nWe will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data.\n[SUBTITLE] 2.7.5. Grading the quality of evidence. [SUBSECTION] We will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence.\nWe will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence.\n[SUBTITLE] 2.7.6. Dealing with missing data. [SUBSECTION] We will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed.\nWe will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed.\n[SUBTITLE] 2.7.7. Ethics and dissemination. [SUBSECTION] No ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal.\nNo ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal.", "This research program was registered in PROSPERO (registration number: CRD42022320766). The protocol report was based on the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols 2015 statement.[18]", "[SUBTITLE] 2.2.1. Participants. [SUBSECTION] COVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants.\nCOVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants.\n[SUBTITLE] 2.2.2. Interventions. [SUBSECTION] The experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi.\nThe experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi.\n[SUBTITLE] 2.2.3. Outcomes. [SUBSECTION] The primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22]\nData on adverse events and the safety of the included studies will be utilized for safety analysis.\nThe primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22]\nData on adverse events and the safety of the included studies will be utilized for safety analysis.\n[SUBTITLE] 2.2.4. Studies. [SUBSECTION] We will include randomized controlled trials (RCTs).\nWe will include randomized controlled trials (RCTs).", "COVID-19 patients with anxiety disorders will be included. There will be no restriction on the age, race, and country of the participants.", "The experimental group will include patients who underwent Tai Chi, including modified Tai Chi. The control group will include patients who received Western or Chinese herbal medicine, except Tai Chi.", "The primary outcome will be changes in Hamilton Anxiety Scale[19] and Self-Rating Anxiety Scale[20] scores after treatment. The secondary outcome indicators will be Liebowitz Social Anxiety Scale[21] and Short Form 36 questionnaire scores. SF-36 is used to measure an individual’s quality of life.[22]\nData on adverse events and the safety of the included studies will be utilized for safety analysis.", "We will include randomized controlled trials (RCTs).", "We will search the electronic databases of PubMed, EMBASE, Cochrane Library, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, Wan Fang, and the Chinese Clinical Trial Registry for RCTs on Tai Chi for the treatment of anxiety disorders caused by COVID-19. Relevant articles published from December 1, 2019, to August 22, 2022, will be included. The search strategy for PubMed is presented in Table 1.\nPubMed search strategy.\nCOVID-19 = coronavirus disease 2019.", "Two reviewers (S-QZ and XY) will independently review and screen the studies according to the inclusion and exclusion criteria of the review. First, the reviewers will utilize EndNote X9 software to exclude duplicate articles. Thereafter, they will exclude obviously irrelevant literature by reading the titles and abstracts of the articles. They will then screen the remaining articles by reading their full texts and provide the reasons for excluding ineligible studies. The selection process is shown in Figure 1.\nThe study selection process.", "Two reviewers (H-YL and X-YM) will independently screen the literature and extract the data from the included studies. The collected data will include the information of each study (year of publication, first author, sample size, age and sex of participants, course of the disease, intervention, control, course of treatment, primary outcome, and secondary outcome) and its results (results of outcomes, adverse events, and safety). If there is any disagreement between the two reviewers during the screening, they will consult a third reviewer (QT) for the final decision.", "Two researchers (R-YL and R-YW) will separately evaluate the methodological qualities of the literature using the Cochrane Collaboration’s Risk of Bias tool, which includes 7 items. The risk of bias in each aspect will be assessed, and the results will be categorized into three grades: low risk, unclear risk, and high risk.", "[SUBTITLE] 2.7.1. Data synthesis and assessment of heterogeneity. [SUBSECTION] RevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity.\nRevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity.\n[SUBTITLE] 2.7.2. Assessment of reporting bias. [SUBSECTION] We will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias.\nWe will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias.\n[SUBTITLE] 2.7.3. Subgroup analysis. [SUBSECTION] If significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi.\nIf significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi.\n[SUBTITLE] 2.7.4. Sensitivity analysis. [SUBSECTION] We will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data.\nWe will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data.\n[SUBTITLE] 2.7.5. Grading the quality of evidence. [SUBSECTION] We will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence.\nWe will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence.\n[SUBTITLE] 2.7.6. Dealing with missing data. [SUBSECTION] We will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed.\nWe will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed.\n[SUBTITLE] 2.7.7. Ethics and dissemination. [SUBSECTION] No ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal.\nNo ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal.", "RevMan software 5.4 will be used for statistical analyses. We will analyze continuous variables using mean differences or standard mean differences with 95% confidence intervals. I2 is used to evaluate the heterogeneity of literature statistics. When I2 < 50% indicates low heterogeneity, the fixed effect model will be used for analysis. When I2 ≥ 50%, indicating high heterogeneity, it is necessary to determine the cause of heterogeneity and select appropriate methods to reduce heterogeneity.", "We will utilize a funnel plot to assess reporting bias if the included studies are more than ten. An asymmetrical funnel plot indicates the existence of publishing bias.", "If significant heterogeneity exists, subgroup analysis will be performed according to different courses of treatment, levels of anxiety, or types of Tai Chi.", "We will conduct a sensitivity analysis to assess the robustness and reliability of the results by excluding low-quality studies and focusing on missing data.", "We will use the Grading of Recommendations Assessment, Development, and Evaluation Reliability Study system to assess the quality of the obtained evidence.", "We will try to contact the corresponding author of any study with missing data to obtain the missing information. If the corresponding author cannot be reached, we will use the available data for synthesis, and the potential impact of the missing information will be reviewed.", "No ethical approval is needed for this type of systematic review as the data is reviewed retrospectively. This systematic review and meta-analysis will be published in a peer-reviewed journal.", "The COVID-19 pandemic has had a significant impact on the mental health of patients and healthcare workers. Patients diagnosed with COVID-19 require isolation, and during isolation, the patients tend to experience fear, loneliness, sadness, and other negative emotions, which result in anxiety.[23] Although several patients with COVID-19 recover, they experience serious psychological effects of the disease, including anxiety.[24] Tai Chi is a traditional Chinese rehabilitation therapy with a long history. It focuses on self-cultivation, mood improvement, and strengthening of the body.[25] It has been found that Tai Chi training can not only strengthen the body and improve the body’s immunity and lung function but also improve anxiety and depression.[26] Our systematic review and meta-analysis can serve as a reference for the clinical treatment and improvement of the psychological status of patients with COVID-19.", "Assessment of risk bias: Runyu Liang, Ruoyu Wang.\nConceptualization: Shiqiang Zhang, Luwen Zhu.\nData curation: Hongyu Li, Xiyuan Ma.\nFormal analysis: Shiqiang Zhang, Hongyu Li.\nFunding: Luwen Zhu.\nInvestigation: Shiqiang Zhang, Xia Yin.\nProject administration: Qiang Tang.\nResources: Shiqiang Zhang.\nSoftware: Shiqiang Zhang.\nWriting – original draft: Hongyu Li, Shiqiang Zhang.\nWriting – review & editing: Hongyu Li, Qiang Tang." ]
[ "intro", "methods", null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, "discussion", null ]
[ "anxiety disorders", "COVID-19", "meta-analysis", "protocol", "Tai Chi" ]
PD-1 and CTLA-4 inhibitors in combination vs. alone for the treatment of advanced melanoma: A systematic review and meta-analysis.
36254050
Metastatic melanoma treatment has drastically changed during the past decade with the advent of immunotherapy. We conducted a meta-analysis, to assess PD-1 and CTLA-4 inhibitors in combination vs. alone for the treatment of advanced melanoma.
BACKGROUND
The EMBASE, Medline via PubMed, Scopus, Cochrane Central, and Web of Science databases were searched. The records retrieved were screened for eligibility. Odds ratio (OR) was applied to compare dichotomous variables. All the results were reported with 95% confidence intervals (CI). Mantel-Haenszel method was used to estimate pooled OR and 95% confidence intervals for dichotomous data.
METHODS
We retrieved 3092 citations of which we included 3 randomized controlled trials and 2 retrospective, cohort studies. The pooled OR was 2.144 (95% CI: 1.650-2.786, I2 = 80.38% P = .000) for overall response and 2.117 (95% CI: 1.578-2.841, I2 = 70.17% P = .000) for the complete response (CR). Subgroup analysis in nivolumab category showed that the pooled OR was 1.766 (95% CI: 1.324-2.355, I2 = 0.0% P = .000) for the overall response and was 1.284 (95% CI: 0.889-1.855, I2 = 0.0% P = .182) for the CR and in the ipilimumab category the pooled OR was 5.440 (95% CI: 2.896-10.220, I2 = 70.89% P = .001) for the overall response and was 5.169 (95% CI: 3.163-8.446, I2 = 0.0% P = .000) for the CR. The incidence of any treatment-related adverse events was significantly higher in the combination group than that of the nivolumab monotherapy 4.044 (95% CI: 1.740-9.403, I2 = 91.64% P = .001) or the ipilimumab monotherapy 2.465 (95% CI: 0.839-7.236, I2 = 93.02 % P = .101).
RESULTS
Combination therapy with ipilimumab plus nivolumab is a promising strategy in the treatment of patients with advanced melanoma with superior overall and complete responses over either monotherapies.
CONCLUSION
[ "Humans", "Antineoplastic Combined Chemotherapy Protocols", "Immune Checkpoint Inhibitors", "Ipilimumab", "Melanoma", "Nivolumab", "Programmed Cell Death 1 Receptor", "Retrospective Studies" ]
9575742
1. Introduction
Metastatic melanoma treatment has drastically changed during the past decade with the advent of immunotherapy and then molecular targeted therapy. Today, 5-year survival is achievable in almost 50% of the patients with metastatic melanoma when treated with combination immunotherapy.[1] This is in contrast to 10 years ago when metastatic melanoma was considered unvaryingly lethal with an overall survival rate of less than 5%.[2] The last decade has observed a significant change in the treatment of metastatic or unresectable melanoma patients, with the advent of immune checkpoint blockade proteins including ipilimumab, nivolumab, pembrolizumab, and combination ipilimumab-nivolumab. These antibodies are coinhibitory protein receptors (PD-1 and CTLA-4 coinhibitory receptors) which are located on the surface of the lymphocytes.[3] Their ligands (e.g., PD-L1/PD-L2 and B7, respectively), on the other hand, are expressed on the tumor cells and restrain T-cells function rendering them unable to mount a response against cancer cells and causing resistance of malignant melanoma in many patients to conventional anticancer therapy.[4] In 2010, Hodi et al, showed, for the first time, that overall survival in patients with metastatic melanoma improved with the treatment via the anti–CTLA-4 antibody (ipilimumab).[5] Also, data on 1861 patients across 12 trials treated with ipilimumab revealed a 3-year survival rate of approximately 20% (plateaued afterward, supporting the durability of response to CTLA-4 blockade).[6] Accordingly, monoclonal antibodies targeting PD-1 were developed which demonstrated clinical activity in melanoma[7] with an even higher overall response rate (30%–40% at 5 years) compared to CTLA-4 blockade and ongoing durable responses in 70%–80% of responding patients.[8] Subsequently, dual immune checkpoint blockade (ICB) with ipilimumab-nivolumab was introduced and demonstrated considerable enhancements in response rate (58%) compared with ipilimumab or nivolumab alone in patients with advanced melanoma. Besides, both nivolumab-containing arms demonstrated superior overall survival (OS) compared with ipilimumab alone.[9] More data emerging from five-year follow-up verified a substantial OS where more than half of patients (52%) in the ICB group were still alive at the time of assessment. The median treatment-free interval was also demonstrably high reaching 18.1 months, highlighting the durability of these responses.[1] Nevertheless, this was accompanied by significant toxicity from dual ICB resulting in treatment interruption or discontinuation in more than 50% of the patients.[10] Nivolumab plus ipilimumab showed a high survival rate and safety outcomes in other studies, further backing up the application of this combination for advanced melanoma in multiple subgroups.[11] A meta-analysis of two studies showed that nivolumab-plus ipilimumab combination therapy had an obvious significant advantage over the ipilimumab monotherapy in patients with advanced melanoma.[12] Considering the increase in the number of studies performed in the field, we conducted a meta-analysis, to assess PD-1 and CTLA-4 inhibitors in combination Vs. alone for the treatment of advanced melanoma.
2. Materials and Methods
This review was conducted according to a predetermined protocol based on the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement.[13] [SUBTITLE] 2.1. Literature search [SUBSECTION] The EMBASE, Medline via PubMed, Scopus, Cochrane Central, and Web of Science databases were electronically searched using the following terms “anti-PD-1” “nivolumab,” “pembrolizumab,” “anti-CTLA-4,” “’ ipilimumab,” and “melanoma” and the following search strategy ((((((PD-1[Title/Abstract]) OR (pembrolizumab [Title/Abstract])) OR (nivolumab [Title/Abstract])) AND (ctla-4[Title/Abstract])) OR (ipilimumab [Title/Abstract]))) AND (melanoma [Title/Abstract]). Two independent reviewers screened the titles and abstracts of retrieved citations. Accordingly, the full texts of the retrieved papers were screened and included only when they fulfilled our criteria. The electronic search was accompanied by manual searches for references to the included studies and related citations. Discrepancies were resolved by discussion between reviewers or by a third senior author. Only original research studies in English were considered. The search had no time restriction. The EMBASE, Medline via PubMed, Scopus, Cochrane Central, and Web of Science databases were electronically searched using the following terms “anti-PD-1” “nivolumab,” “pembrolizumab,” “anti-CTLA-4,” “’ ipilimumab,” and “melanoma” and the following search strategy ((((((PD-1[Title/Abstract]) OR (pembrolizumab [Title/Abstract])) OR (nivolumab [Title/Abstract])) AND (ctla-4[Title/Abstract])) OR (ipilimumab [Title/Abstract]))) AND (melanoma [Title/Abstract]). Two independent reviewers screened the titles and abstracts of retrieved citations. Accordingly, the full texts of the retrieved papers were screened and included only when they fulfilled our criteria. The electronic search was accompanied by manual searches for references to the included studies and related citations. Discrepancies were resolved by discussion between reviewers or by a third senior author. Only original research studies in English were considered. The search had no time restriction. [SUBTITLE] 2.2. Inclusion and exclusion criteria [SUBSECTION] English full-text randomized controlled trials (RCTs) that fulfilled the following criteria were included in this meta-analysis: patients with advanced stage III or stage IV melanoma, in whom nivolumab or ipilimumab was administered alone (control) or in combination (intervention), and assessed progression-free survival (PFS) rate, overall survival (OS; the time from the initiation of treatment until death), complete response rate, partial response rate, objective response rate (ORR), stable disease rate, and safety measures as their outcomes. On the other hand, non-English articles, nonoriginal articles, thesis or conference papers that were never subsequently published, and studies on animals (in vivo) and cell lines (in vitro) were excluded from this study. English full-text randomized controlled trials (RCTs) that fulfilled the following criteria were included in this meta-analysis: patients with advanced stage III or stage IV melanoma, in whom nivolumab or ipilimumab was administered alone (control) or in combination (intervention), and assessed progression-free survival (PFS) rate, overall survival (OS; the time from the initiation of treatment until death), complete response rate, partial response rate, objective response rate (ORR), stable disease rate, and safety measures as their outcomes. On the other hand, non-English articles, nonoriginal articles, thesis or conference papers that were never subsequently published, and studies on animals (in vivo) and cell lines (in vitro) were excluded from this study. [SUBTITLE] 2.3. Data extraction [SUBSECTION] Data extraction was independently conducted by two reviewers using a standardized approach. Disagreements were resolved by discussion with a third senior reviewer. Data on authors’ names, year of publication, journal name, study phase, the sample size in each arm, immunotherapy regimen used, the mean age of patients, and information regarding study design (randomization, allocation concealment, description of withdrawals per arm, and blinding) for the trials included in the study. Data extraction was independently conducted by two reviewers using a standardized approach. Disagreements were resolved by discussion with a third senior reviewer. Data on authors’ names, year of publication, journal name, study phase, the sample size in each arm, immunotherapy regimen used, the mean age of patients, and information regarding study design (randomization, allocation concealment, description of withdrawals per arm, and blinding) for the trials included in the study. [SUBTITLE] 2.4. Quality assessment [SUBSECTION] Cochrane Collaboration’s tool for risk of bias (ROB) assessment was used to assess the quality of the included studies. The items of this tool were as follows; allocation concealment, selective outcome reporting, blinding of participants, masking of outcome assessors, generation of allocation sequence, incomplete follow-up, and other potential sources of bias. A senior author judged any disagreements between the 2 reviewers who performed the assessment. For each element, the risk of bias was considered as low, unclear, or high. If the reviewer could find information on all the parameters mentioned in the tool or no information at all, then the study was allocated to one of the low bias or high bias categories, respectively. If the information retrieved by the reviewer was partial or unclear, the risk of bias was considered to be unclear.[14] Cochrane Collaboration’s tool for risk of bias (ROB) assessment was used to assess the quality of the included studies. The items of this tool were as follows; allocation concealment, selective outcome reporting, blinding of participants, masking of outcome assessors, generation of allocation sequence, incomplete follow-up, and other potential sources of bias. A senior author judged any disagreements between the 2 reviewers who performed the assessment. For each element, the risk of bias was considered as low, unclear, or high. If the reviewer could find information on all the parameters mentioned in the tool or no information at all, then the study was allocated to one of the low bias or high bias categories, respectively. If the information retrieved by the reviewer was partial or unclear, the risk of bias was considered to be unclear.[14] [SUBTITLE] 2.5. Statistics [SUBSECTION] The Comprehensive Meta-Analysis Software (CMA) software version 2.0 was used to analyze the data. Odds ratio (OR) was applied to compare dichotomous variables. All the results were reported with 95% confidence intervals (CI). Mantel–Haenszel method was used to estimate pooled OR and 95% confidence intervals for dichotomous data. Heterogeneity of the data was assessed using I-square (I2) test and considered high if I2>50%. In this case, the random effect model was chosen; otherwise, the fixed effect model was used. Subgroup analyses were conducted based on the intervention of the study design: anti-PD-1 plus anti-CLTA-4 versus anti-PD-1 (nivolumab), and anti-PD-1 plus anti-CLTA-4 versus anti-CLTA-4 (ipilimumab). Funnel plotting, Egger’s regression, and trim and fill were not used for the assessment of publication bias in this literature as the number of studies was less than 10. P < .05 is considered statistically significant. The Comprehensive Meta-Analysis Software (CMA) software version 2.0 was used to analyze the data. Odds ratio (OR) was applied to compare dichotomous variables. All the results were reported with 95% confidence intervals (CI). Mantel–Haenszel method was used to estimate pooled OR and 95% confidence intervals for dichotomous data. Heterogeneity of the data was assessed using I-square (I2) test and considered high if I2>50%. In this case, the random effect model was chosen; otherwise, the fixed effect model was used. Subgroup analyses were conducted based on the intervention of the study design: anti-PD-1 plus anti-CLTA-4 versus anti-PD-1 (nivolumab), and anti-PD-1 plus anti-CLTA-4 versus anti-CLTA-4 (ipilimumab). Funnel plotting, Egger’s regression, and trim and fill were not used for the assessment of publication bias in this literature as the number of studies was less than 10. P < .05 is considered statistically significant.
3. Results
[SUBTITLE] 3.1. General characteristics of the included studies [SUBSECTION] We retrieved 3092 citations, including 3088 publications by electronic search of databases, and 4 studies by a manual search of websites and checking reference lists of the included studies. After the removal of duplicate records, a total of 2839 titles were screened, during which 2779 articles were excluded. Accordingly, and after abstract and full-text screening, 3 randomized controlled trials and 2 retrospective, cohort studies were included in this meta-analysis. The detailed information regarding the number of identified studies, and the stages of evaluation and exclusion is presented in Figure 1 (see PRISMA flow diagram). PRISMA flowchart of the included studies in the systematic review and meta-analysis. The characteristics of the included studies are presented in Table 1. Of the included RCTs, one publication was from phase III and two citations were from phase II. A total of 1605 patients were included in this meta-analysis of which 675 were in the combination therapy group, 359 in the nivolumab monotherapy group, and 571 in the ipilimumab monotherapy group. PFS was the main endpoint in 4 of the included citations and OS was the primary outcome in 4 of the included papers. In all of the included studies, PFS and OS were higher in the combination therapy group (nivolumab plus ipilimumab) than those of the monotherapy groups (nivolumab or ipilimumab). Characteristics of included studies in the meta-analysis. Ipi = ipilimumab, ORR =objective response rate, OS = overall survival, PFS = progression-free survival. We retrieved 3092 citations, including 3088 publications by electronic search of databases, and 4 studies by a manual search of websites and checking reference lists of the included studies. After the removal of duplicate records, a total of 2839 titles were screened, during which 2779 articles were excluded. Accordingly, and after abstract and full-text screening, 3 randomized controlled trials and 2 retrospective, cohort studies were included in this meta-analysis. The detailed information regarding the number of identified studies, and the stages of evaluation and exclusion is presented in Figure 1 (see PRISMA flow diagram). PRISMA flowchart of the included studies in the systematic review and meta-analysis. The characteristics of the included studies are presented in Table 1. Of the included RCTs, one publication was from phase III and two citations were from phase II. A total of 1605 patients were included in this meta-analysis of which 675 were in the combination therapy group, 359 in the nivolumab monotherapy group, and 571 in the ipilimumab monotherapy group. PFS was the main endpoint in 4 of the included citations and OS was the primary outcome in 4 of the included papers. In all of the included studies, PFS and OS were higher in the combination therapy group (nivolumab plus ipilimumab) than those of the monotherapy groups (nivolumab or ipilimumab). Characteristics of included studies in the meta-analysis. Ipi = ipilimumab, ORR =objective response rate, OS = overall survival, PFS = progression-free survival. [SUBTITLE] 3.2. Efficacy [SUBSECTION] The main endpoints of the included studies were ORR, PFS, and OS for efficacy. However, not all of the included studies reported all of these endpoints. For example, ORR was reported only by one of the included studies. Subgroup analyses were conducted based on the intervention of the study design. The heterogeneity of the included studies was found to be high thus random effect model was used for the analyses. The PFS and/or OS of these trials are presented in Table 1. Overall analysis showed that the pooled OR was 2.144 (95% CI: 1.650–2.786, I2 = 80.38% P = .000; Fig. 2) for overall response and 2.117 (95% CI: 1.578–2.841, I2 = 70.17% P = .000; Fig. 3) for the complete response (CR). Forest plot of standardized mean difference (SMD) for overall response for combination therapy vs. both ipilimumab and nivolumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Red squares show pooled effect in each subgroup. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI). Forest plot of standardized mean difference (SMD) for complete response for combination therapy vs. both ipilimumab and nivolumab therapy in patients with advanced melanoma. Green square shows overall pooled effect. Red squares show pooled effect in each subgroup. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI). The main endpoints of the included studies were ORR, PFS, and OS for efficacy. However, not all of the included studies reported all of these endpoints. For example, ORR was reported only by one of the included studies. Subgroup analyses were conducted based on the intervention of the study design. The heterogeneity of the included studies was found to be high thus random effect model was used for the analyses. The PFS and/or OS of these trials are presented in Table 1. Overall analysis showed that the pooled OR was 2.144 (95% CI: 1.650–2.786, I2 = 80.38% P = .000; Fig. 2) for overall response and 2.117 (95% CI: 1.578–2.841, I2 = 70.17% P = .000; Fig. 3) for the complete response (CR). Forest plot of standardized mean difference (SMD) for overall response for combination therapy vs. both ipilimumab and nivolumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Red squares show pooled effect in each subgroup. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI). Forest plot of standardized mean difference (SMD) for complete response for combination therapy vs. both ipilimumab and nivolumab therapy in patients with advanced melanoma. Green square shows overall pooled effect. Red squares show pooled effect in each subgroup. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI). [SUBTITLE] 3.3. Nivolumab plus ipilimumab vs. nivolumab [SUBSECTION] Subgroup analysis in this category showed that the pooled OR was 1.766 (95% CI: 1.324–2.355, I2 = 0.0% P = .000; Fig. 2) for the overall response and was 1.284 (95% CI: 0.889–1.855, I2 = 0.0% P = .182; Fig. 3) for the CR. These findings indicated a significantly higher overall response but not CR and thus beneficial for the patients in the combination therapy group compared with the nivolumab group in the included population. Also, in the combination therapy group, significantly longer OS and PFS were observed in each of the included studies. However, analysis was not possible for these variables (Table 1). Subgroup analysis in this category showed that the pooled OR was 1.766 (95% CI: 1.324–2.355, I2 = 0.0% P = .000; Fig. 2) for the overall response and was 1.284 (95% CI: 0.889–1.855, I2 = 0.0% P = .182; Fig. 3) for the CR. These findings indicated a significantly higher overall response but not CR and thus beneficial for the patients in the combination therapy group compared with the nivolumab group in the included population. Also, in the combination therapy group, significantly longer OS and PFS were observed in each of the included studies. However, analysis was not possible for these variables (Table 1). [SUBTITLE] 3.4. Nivolumab plus ipilimumab vs. ipilimumab [SUBSECTION] Subgroup analysis in this category revealed that the pooled OR was 5.440 (95% CI: 2.896–10.220, I2 = 70.89% P = .001; Fig. 2) for the overall response and was 5.169 (95% CI: 3.163–8.446, I2 = 0.0% P = .000; Fig. 3) for the complete response (CR). These findings indicated a significantly higher overall response, CR, and thus beneficial for the patients in the combination therapy group compared with the ipilimumab group in the included population. Also, in the combination therapy group, significantly longer OS and PFS were observed in each of the included studies. However, analysis was not possible for these variables (Table 1). Subgroup analysis in this category revealed that the pooled OR was 5.440 (95% CI: 2.896–10.220, I2 = 70.89% P = .001; Fig. 2) for the overall response and was 5.169 (95% CI: 3.163–8.446, I2 = 0.0% P = .000; Fig. 3) for the complete response (CR). These findings indicated a significantly higher overall response, CR, and thus beneficial for the patients in the combination therapy group compared with the ipilimumab group in the included population. Also, in the combination therapy group, significantly longer OS and PFS were observed in each of the included studies. However, analysis was not possible for these variables (Table 1). [SUBTITLE] 3.5. Safety analysis [SUBSECTION] Larkin et al showed that 59%, 23%, and 28% of patients in the nivolumab-plus-ipilimumab, nivolumab, and ipilimumab groups, respectively, had grade 3 or 4 treatment-related side effects.[1] Hodi et al showed that at the time of the most recent data lock, the rates of treatment-related adverse events of any grade were 92% (86 of 94 patients) and 94 percent (43 of 46 patients), respectively. The most common adverse reactions to therapy were diarrhea, rash, fatigue, and pruritus in both groups. Consistent with Larkin et al, study, Hodi et al, showed that the frequency of grade 3–4 adverse events was higher in the combination therapy group.[15] However, da Silva et al, found that the rate of grade 3–4 adverse events was similar between the groups. Diarrhea or colitis was the most frequent grade 3–5 treatment-related adverse event, followed by a rise in alanine aminotransferase or aspartate aminotransferase in this study.[16] Our analysis revealed that the incidence of any treatment-related adverse events was significantly higher in the combination group than that of the nivolumab monotherapy 4.044 (95% CI: 1.740–9.403, I2 = 91.64% P = .001; Fig. 4). However, the heterogeneity of the included studies was high. Also, the incidence of any treatment-related adverse events was higher in the combination group than that of the ipilimumab monotherapy 2.465 (95% CI: 0.839–7.236, I2 = 93.02% P = .101; Fig. 5). However, this did not reach a statistical significance and the heterogeneity of the included studies was modest. Forest plot of standardized mean difference (SMD) for treatment related-adverse event for combination therapy vs. nivolumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI). Forest plot of standardized mean difference (SMD) for treatment related-adverse event for combination therapy vs. ipilimumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI). Larkin et al showed that 59%, 23%, and 28% of patients in the nivolumab-plus-ipilimumab, nivolumab, and ipilimumab groups, respectively, had grade 3 or 4 treatment-related side effects.[1] Hodi et al showed that at the time of the most recent data lock, the rates of treatment-related adverse events of any grade were 92% (86 of 94 patients) and 94 percent (43 of 46 patients), respectively. The most common adverse reactions to therapy were diarrhea, rash, fatigue, and pruritus in both groups. Consistent with Larkin et al, study, Hodi et al, showed that the frequency of grade 3–4 adverse events was higher in the combination therapy group.[15] However, da Silva et al, found that the rate of grade 3–4 adverse events was similar between the groups. Diarrhea or colitis was the most frequent grade 3–5 treatment-related adverse event, followed by a rise in alanine aminotransferase or aspartate aminotransferase in this study.[16] Our analysis revealed that the incidence of any treatment-related adverse events was significantly higher in the combination group than that of the nivolumab monotherapy 4.044 (95% CI: 1.740–9.403, I2 = 91.64% P = .001; Fig. 4). However, the heterogeneity of the included studies was high. Also, the incidence of any treatment-related adverse events was higher in the combination group than that of the ipilimumab monotherapy 2.465 (95% CI: 0.839–7.236, I2 = 93.02% P = .101; Fig. 5). However, this did not reach a statistical significance and the heterogeneity of the included studies was modest. Forest plot of standardized mean difference (SMD) for treatment related-adverse event for combination therapy vs. nivolumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI). Forest plot of standardized mean difference (SMD) for treatment related-adverse event for combination therapy vs. ipilimumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI). [SUBTITLE] 3.6. Publication bias assessment [SUBSECTION] The quality of included studies was evaluated based on the standards of RCT quality assessment of the Cochrane Reviewer handbook.[14] There was no attrition bias and reporting bias in the included studies. However, detection, performance, and selection (allocation concealment) biases were found in three of the included citations.[16–18] Also, random sequence generation was not implemented in two of the included publications[16,18] (Fig. 6). Different levels of risk of bias for each item in included studies. The Cochrane risk of bias tool was used for the detection of publication bias. The quality of included studies was evaluated based on the standards of RCT quality assessment of the Cochrane Reviewer handbook.[14] There was no attrition bias and reporting bias in the included studies. However, detection, performance, and selection (allocation concealment) biases were found in three of the included citations.[16–18] Also, random sequence generation was not implemented in two of the included publications[16,18] (Fig. 6). Different levels of risk of bias for each item in included studies. The Cochrane risk of bias tool was used for the detection of publication bias.
5. Conclusions
Data emerging from this study showed that combination therapy with ipilimumab plus nivolumab meaningfully increased the overall response rate, including the CR, compared with monotherapy with ipilimumab or nivolumab in patients with advanced melanoma regardless of the patients untreated or after anti-CTLA-4 treatment. This was accompanied by higher incidences of potential IrAEs in combination therapy. In this light, combination therapy with ipilimumab plus nivolumab could be a promising strategy in the treatment of patients with advanced melanoma.
[ "2.1. Literature search", "2.2. Inclusion and exclusion criteria", "2.3. Data extraction", "2.4. Quality assessment", "2.5. Statistics", "3.1. General characteristics of the included studies", "3.2. Efficacy", "3.3. Nivolumab plus ipilimumab vs. nivolumab", "3.4. Nivolumab plus ipilimumab vs. ipilimumab", "3.5. Safety analysis", "3.6. Publication bias assessment", "4. Discussions", "4.1. A closer look at individual studies", "4.2. Safety", "4.3. Limitations", "Author contributions" ]
[ "The EMBASE, Medline via PubMed, Scopus, Cochrane Central, and Web of Science databases were electronically searched using the following terms “anti-PD-1” “nivolumab,” “pembrolizumab,” “anti-CTLA-4,” “’ ipilimumab,” and “melanoma” and the following search strategy ((((((PD-1[Title/Abstract]) OR (pembrolizumab [Title/Abstract])) OR (nivolumab [Title/Abstract])) AND (ctla-4[Title/Abstract])) OR (ipilimumab [Title/Abstract]))) AND (melanoma [Title/Abstract]). Two independent reviewers screened the titles and abstracts of retrieved citations. Accordingly, the full texts of the retrieved papers were screened and included only when they fulfilled our criteria. The electronic search was accompanied by manual searches for references to the included studies and related citations. Discrepancies were resolved by discussion between reviewers or by a third senior author. Only original research studies in English were considered. The search had no time restriction.", "English full-text randomized controlled trials (RCTs) that fulfilled the following criteria were included in this meta-analysis: patients with advanced stage III or stage IV melanoma, in whom nivolumab or ipilimumab was administered alone (control) or in combination (intervention), and assessed progression-free survival (PFS) rate, overall survival (OS; the time from the initiation of treatment until death), complete response rate, partial response rate, objective response rate (ORR), stable disease rate, and safety measures as their outcomes. On the other hand, non-English articles, nonoriginal articles, thesis or conference papers that were never subsequently published, and studies on animals (in vivo) and cell lines (in vitro) were excluded from this study.", "Data extraction was independently conducted by two reviewers using a standardized approach. Disagreements were resolved by discussion with a third senior reviewer. Data on authors’ names, year of publication, journal name, study phase, the sample size in each arm, immunotherapy regimen used, the mean age of patients, and information regarding study design (randomization, allocation concealment, description of withdrawals per arm, and blinding) for the trials included in the study.", "Cochrane Collaboration’s tool for risk of bias (ROB) assessment was used to assess the quality of the included studies. The items of this tool were as follows; allocation concealment, selective outcome reporting, blinding of participants, masking of outcome assessors, generation of allocation sequence, incomplete follow-up, and other potential sources of bias. A senior author judged any disagreements between the 2 reviewers who performed the assessment. For each element, the risk of bias was considered as low, unclear, or high. If the reviewer could find information on all the parameters mentioned in the tool or no information at all, then the study was allocated to one of the low bias or high bias categories, respectively. If the information retrieved by the reviewer was partial or unclear, the risk of bias was considered to be unclear.[14]", "The Comprehensive Meta-Analysis Software (CMA) software version 2.0 was used to analyze the data. Odds ratio (OR) was applied to compare dichotomous variables. All the results were reported with 95% confidence intervals (CI). Mantel–Haenszel method was used to estimate pooled OR and 95% confidence intervals for dichotomous data. Heterogeneity of the data was assessed using I-square (I2) test and considered high if I2>50%. In this case, the random effect model was chosen; otherwise, the fixed effect model was used. Subgroup analyses were conducted based on the intervention of the study design: anti-PD-1 plus anti-CLTA-4 versus anti-PD-1 (nivolumab), and anti-PD-1 plus anti-CLTA-4 versus anti-CLTA-4 (ipilimumab). Funnel plotting, Egger’s regression, and trim and fill were not used for the assessment of publication bias in this literature as the number of studies was less than 10. P < .05 is considered statistically significant.", "We retrieved 3092 citations, including 3088 publications by electronic search of databases, and 4 studies by a manual search of websites and checking reference lists of the included studies. After the removal of duplicate records, a total of 2839 titles were screened, during which 2779 articles were excluded. Accordingly, and after abstract and full-text screening, 3 randomized controlled trials and 2 retrospective, cohort studies were included in this meta-analysis. The detailed information regarding the number of identified studies, and the stages of evaluation and exclusion is presented in Figure 1 (see PRISMA flow diagram).\nPRISMA flowchart of the included studies in the systematic review and meta-analysis.\nThe characteristics of the included studies are presented in Table 1. Of the included RCTs, one publication was from phase III and two citations were from phase II. A total of 1605 patients were included in this meta-analysis of which 675 were in the combination therapy group, 359 in the nivolumab monotherapy group, and 571 in the ipilimumab monotherapy group. PFS was the main endpoint in 4 of the included citations and OS was the primary outcome in 4 of the included papers. In all of the included studies, PFS and OS were higher in the combination therapy group (nivolumab plus ipilimumab) than those of the monotherapy groups (nivolumab or ipilimumab).\nCharacteristics of included studies in the meta-analysis.\nIpi = ipilimumab, ORR =objective response rate, OS = overall survival, PFS = progression-free survival.", "The main endpoints of the included studies were ORR, PFS, and OS for efficacy. However, not all of the included studies reported all of these endpoints. For example, ORR was reported only by one of the included studies. Subgroup analyses were conducted based on the intervention of the study design. The heterogeneity of the included studies was found to be high thus random effect model was used for the analyses. The PFS and/or OS of these trials are presented in Table 1. Overall analysis showed that the pooled OR was 2.144 (95% CI: 1.650–2.786, I2 = 80.38% P = .000; Fig. 2) for overall response and 2.117 (95% CI: 1.578–2.841, I2 = 70.17% P = .000; Fig. 3) for the complete response (CR).\nForest plot of standardized mean difference (SMD) for overall response for combination therapy vs. both ipilimumab and nivolumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Red squares show pooled effect in each subgroup. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).\nForest plot of standardized mean difference (SMD) for complete response for combination therapy vs. both ipilimumab and nivolumab therapy in patients with advanced melanoma. Green square shows overall pooled effect. Red squares show pooled effect in each subgroup. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).", "Subgroup analysis in this category showed that the pooled OR was 1.766 (95% CI: 1.324–2.355, I2 = 0.0% P = .000; Fig. 2) for the overall response and was 1.284 (95% CI: 0.889–1.855, I2 = 0.0% P = .182; Fig. 3) for the CR. These findings indicated a significantly higher overall response but not CR and thus beneficial for the patients in the combination therapy group compared with the nivolumab group in the included population. Also, in the combination therapy group, significantly longer OS and PFS were observed in each of the included studies. However, analysis was not possible for these variables (Table 1).", "Subgroup analysis in this category revealed that the pooled OR was 5.440 (95% CI: 2.896–10.220, I2 = 70.89% P = .001; Fig. 2) for the overall response and was 5.169 (95% CI: 3.163–8.446, I2 = 0.0% P = .000; Fig. 3) for the complete response (CR). These findings indicated a significantly higher overall response, CR, and thus beneficial for the patients in the combination therapy group compared with the ipilimumab group in the included population. Also, in the combination therapy group, significantly longer OS and PFS were observed in each of the included studies. However, analysis was not possible for these variables (Table 1).", "Larkin et al showed that 59%, 23%, and 28% of patients in the nivolumab-plus-ipilimumab, nivolumab, and ipilimumab groups, respectively, had grade 3 or 4 treatment-related side effects.[1] Hodi et al showed that at the time of the most recent data lock, the rates of treatment-related adverse events of any grade were 92% (86 of 94 patients) and 94 percent (43 of 46 patients), respectively. The most common adverse reactions to therapy were diarrhea, rash, fatigue, and pruritus in both groups. Consistent with Larkin et al, study, Hodi et al, showed that the frequency of grade 3–4 adverse events was higher in the combination therapy group.[15] However, da Silva et al, found that the rate of grade 3–4 adverse events was similar between the groups. Diarrhea or colitis was the most frequent grade 3–5 treatment-related adverse event, followed by a rise in alanine aminotransferase or aspartate aminotransferase in this study.[16]\nOur analysis revealed that the incidence of any treatment-related adverse events was significantly higher in the combination group than that of the nivolumab monotherapy 4.044 (95% CI: 1.740–9.403, I2 = 91.64% P = .001; Fig. 4). However, the heterogeneity of the included studies was high. Also, the incidence of any treatment-related adverse events was higher in the combination group than that of the ipilimumab monotherapy 2.465 (95% CI: 0.839–7.236, I2 = 93.02% P = .101; Fig. 5). However, this did not reach a statistical significance and the heterogeneity of the included studies was modest.\nForest plot of standardized mean difference (SMD) for treatment related-adverse event for combination therapy vs. nivolumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).\nForest plot of standardized mean difference (SMD) for treatment related-adverse event for combination therapy vs. ipilimumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).", "The quality of included studies was evaluated based on the standards of RCT quality assessment of the Cochrane Reviewer handbook.[14] There was no attrition bias and reporting bias in the included studies. However, detection, performance, and selection (allocation concealment) biases were found in three of the included citations.[16–18] Also, random sequence generation was not implemented in two of the included publications[16,18] (Fig. 6).\nDifferent levels of risk of bias for each item in included studies. The Cochrane risk of bias tool was used for the detection of publication bias.", "The results emerging from this systematic review and meta-analysis showed that combination therapy with nivolumab plus ipilimumab was more effective than nivolumab or ipilimumab alone in the treatment of advanced metastatic melanoma (treated or untreated). This was suggested by higher overall response and CR of the combination therapy compared with either of the monotherapies. However, this was accompanied by a higher incidence of grade 3/4/5 adverse events in the combination therapy than in either of the monotherapies.\n[SUBTITLE] 4.1. A closer look at individual studies [SUBSECTION] Our pooled analysis was in total agreement with the results of individual trials included in this study. In that light, Larkin et al, showed (CheckMate 067) that the median OS was over 60.0 months in the combination therapy group as opposed to 36.9 and 19.9 months in the nivolumab and ipilimumab groups, respectively. Also, the hazard ratio (HR) for death was found to be lower in the combination therapy group than in ipilimumab alone (nivolumab plus ipilimumab vs. ipilimumab, 0.52; HR for death with nivolumab vs. ipilimumab, 0.63). Five-year follow-up of the patients showed a 52% OS in the combination therapy group as compared with nivolumab or ipilimumab alone (44% and 26%, respectively). The authors reported no new late toxic effects nor sustained deterioration of health-related quality of life in the combination group compared with the monotherapies.[1] Similarly, in another study by Hodi et al, (CheckMate 069) it was found that OS rates in all randomized patients were 63·8% (95% CI: 53.3–72.6) for the combination therapy group vs 53·6% (95% CI: 38.1–66.8) for ipilimumab alone at a median follow-up of 24 months. However, grade 3–4 adverse events associated with combination therapy were observed in 51 [54%] of 94 patients vs 9 [20%] of 46 patients linked to ipilimumab alone. The results of this study indicated that the combination of nivolumab plus ipilimumab may result in a higher OS rate vs ipilimumab in patients with advanced melanoma.[19] In line with previous studies, in a multicenter retrospective cohort study Zimmer et al, found that OS rates for the monotherapy and the combination groups were 16% and 21%, respectively. The disease control rate was 42% for the monotherapy and 33% for the combination groups. One-year OS rates for the monotherapy and the combination groups were 54% and 55%, respectively. The authors, however, stated that the combination therapy with nivolumab and ipilimumab was significantly less effective in patients with advanced melanoma with previous anti-PD failure compared with treatment-naïve melanoma patients.[18] This was, in contrast, to da Silva et al findings, which showed that in advanced melanoma patients who were resistant to anti-PD-1 monotherapy, combination therapy had higher efficacy than monotherapy with a higher ORR (60 [31%] of 193 patients vs 21 [13%] of 162 patients; P < .0001), longer PFS (median 3.0 months [95% CI: 2.6–3.6] vs 2.6 months [2.4–2.9]; HR, 0·69, 95% CI: 0·55–0·87; P = .0019), and longer OS (median overall survival 20·4 months [95% CI: 12·7–34·8] vs 8·8 months [6·1–11·3]; HR, 0·50, 95% CI: 0.38–0.66; P < .0001), with a similar rate of grade 3–5 toxicity. The results of this study suggested combination therapy with ipilimumab and nivolumab as a superior option over ipilimumab monotherapy in advanced melanoma patients.[16] Long et al found that combination therapy with nivolumab plus ipilimumab results in higher an intracranial response than ipilimumab monotherapy (16 (46%; 95% CI: 29–63) of 35 patients, 5 (20%; 7–41) of 25, and 1 (6%; 0–30) of 16, respectively) in patients with metastatic melanoma to the brain.[17]\nA meta-analysis by Menshawy et al[20] performed on 1910 patients (nivolumab group, n = 1207 and control group, n = 703) showed that combination therapy with nivolumab plus ipilimumab had higher ORR [RR: 3.58, 95% CI: 2.08–6.14], complete response rate (RR: 5.93, 95% CI: 2.45–14.37), partial response rate (RR: 2.80, 95% CI: 2.16–3.64), stable disease rate (RR: 0.56, 95% CI: 0.41–0.76), and PFS (hazard ratio: 0.67, 95% CI: 0.60–0.74) compared with ipilimumab monotherapy. However, this meta-analysis is rather old and did not include the 5-year update from the CheckMate 067 study by Larkin et al,[1] and also recent studies by da Silva et al[16] and Long et al.[17] On the other hand, this study included a study by Postow et al,[21] which is an earlier version of the CheckMate 069 study by Hodi et al[19] and should not be included in the meta-analysis as a separate entity. Another meta-analysis by Hao et al performed on two studies showed that combination therapy with nivolumab plus ipilimumab had a visible significant advantage over the ipilimumab monotherapy.[12] But the number of studies was too low to decide on anything.\nOur pooled analysis was in total agreement with the results of individual trials included in this study. In that light, Larkin et al, showed (CheckMate 067) that the median OS was over 60.0 months in the combination therapy group as opposed to 36.9 and 19.9 months in the nivolumab and ipilimumab groups, respectively. Also, the hazard ratio (HR) for death was found to be lower in the combination therapy group than in ipilimumab alone (nivolumab plus ipilimumab vs. ipilimumab, 0.52; HR for death with nivolumab vs. ipilimumab, 0.63). Five-year follow-up of the patients showed a 52% OS in the combination therapy group as compared with nivolumab or ipilimumab alone (44% and 26%, respectively). The authors reported no new late toxic effects nor sustained deterioration of health-related quality of life in the combination group compared with the monotherapies.[1] Similarly, in another study by Hodi et al, (CheckMate 069) it was found that OS rates in all randomized patients were 63·8% (95% CI: 53.3–72.6) for the combination therapy group vs 53·6% (95% CI: 38.1–66.8) for ipilimumab alone at a median follow-up of 24 months. However, grade 3–4 adverse events associated with combination therapy were observed in 51 [54%] of 94 patients vs 9 [20%] of 46 patients linked to ipilimumab alone. The results of this study indicated that the combination of nivolumab plus ipilimumab may result in a higher OS rate vs ipilimumab in patients with advanced melanoma.[19] In line with previous studies, in a multicenter retrospective cohort study Zimmer et al, found that OS rates for the monotherapy and the combination groups were 16% and 21%, respectively. The disease control rate was 42% for the monotherapy and 33% for the combination groups. One-year OS rates for the monotherapy and the combination groups were 54% and 55%, respectively. The authors, however, stated that the combination therapy with nivolumab and ipilimumab was significantly less effective in patients with advanced melanoma with previous anti-PD failure compared with treatment-naïve melanoma patients.[18] This was, in contrast, to da Silva et al findings, which showed that in advanced melanoma patients who were resistant to anti-PD-1 monotherapy, combination therapy had higher efficacy than monotherapy with a higher ORR (60 [31%] of 193 patients vs 21 [13%] of 162 patients; P < .0001), longer PFS (median 3.0 months [95% CI: 2.6–3.6] vs 2.6 months [2.4–2.9]; HR, 0·69, 95% CI: 0·55–0·87; P = .0019), and longer OS (median overall survival 20·4 months [95% CI: 12·7–34·8] vs 8·8 months [6·1–11·3]; HR, 0·50, 95% CI: 0.38–0.66; P < .0001), with a similar rate of grade 3–5 toxicity. The results of this study suggested combination therapy with ipilimumab and nivolumab as a superior option over ipilimumab monotherapy in advanced melanoma patients.[16] Long et al found that combination therapy with nivolumab plus ipilimumab results in higher an intracranial response than ipilimumab monotherapy (16 (46%; 95% CI: 29–63) of 35 patients, 5 (20%; 7–41) of 25, and 1 (6%; 0–30) of 16, respectively) in patients with metastatic melanoma to the brain.[17]\nA meta-analysis by Menshawy et al[20] performed on 1910 patients (nivolumab group, n = 1207 and control group, n = 703) showed that combination therapy with nivolumab plus ipilimumab had higher ORR [RR: 3.58, 95% CI: 2.08–6.14], complete response rate (RR: 5.93, 95% CI: 2.45–14.37), partial response rate (RR: 2.80, 95% CI: 2.16–3.64), stable disease rate (RR: 0.56, 95% CI: 0.41–0.76), and PFS (hazard ratio: 0.67, 95% CI: 0.60–0.74) compared with ipilimumab monotherapy. However, this meta-analysis is rather old and did not include the 5-year update from the CheckMate 067 study by Larkin et al,[1] and also recent studies by da Silva et al[16] and Long et al.[17] On the other hand, this study included a study by Postow et al,[21] which is an earlier version of the CheckMate 069 study by Hodi et al[19] and should not be included in the meta-analysis as a separate entity. Another meta-analysis by Hao et al performed on two studies showed that combination therapy with nivolumab plus ipilimumab had a visible significant advantage over the ipilimumab monotherapy.[12] But the number of studies was too low to decide on anything.\n[SUBTITLE] 4.2. Safety [SUBSECTION] One of the main concerns with immune checkpoint inhibitors is the risk of immune-related adverse events (IrAEs). Nivolumab was shown to be well tolerated by patients with advanced melanoma. On the one hand, it is speculated that combination therapy with immunotherapies elevates the incidence of potential IrAEs. In a systematic review and meta-analysis by Almutairi et al., it was found that the incidences of potential IrAEs in combination therapies vs. monotherapies were higher for most types of IrAEs. This caused hyperglycemia, thyroid, hepatic, and musculoskeletal disorders in the patients receiving these medications.[22] On the other hand, it was argued that the emergence of both early and late IrAEs could be regarded as a favorable prognostic parameter in patients receiving immunotherapy and immunoradiotherapy for solid tumors especially if these events are delayed; as they are accompanied by increased overall response rate and improved OS and PFS in these patients.[23,24] In any case, the oncologist should take into consideration the occurrence of IrAEs in patients with advanced melanoma especially those with a history of autoimmune disease, poor kidney function of grade 3 or greater, and use of CTLA-4 inhibitors.[25]\nOne of the main concerns with immune checkpoint inhibitors is the risk of immune-related adverse events (IrAEs). Nivolumab was shown to be well tolerated by patients with advanced melanoma. On the one hand, it is speculated that combination therapy with immunotherapies elevates the incidence of potential IrAEs. In a systematic review and meta-analysis by Almutairi et al., it was found that the incidences of potential IrAEs in combination therapies vs. monotherapies were higher for most types of IrAEs. This caused hyperglycemia, thyroid, hepatic, and musculoskeletal disorders in the patients receiving these medications.[22] On the other hand, it was argued that the emergence of both early and late IrAEs could be regarded as a favorable prognostic parameter in patients receiving immunotherapy and immunoradiotherapy for solid tumors especially if these events are delayed; as they are accompanied by increased overall response rate and improved OS and PFS in these patients.[23,24] In any case, the oncologist should take into consideration the occurrence of IrAEs in patients with advanced melanoma especially those with a history of autoimmune disease, poor kidney function of grade 3 or greater, and use of CTLA-4 inhibitors.[25]\n[SUBTITLE] 4.3. Limitations [SUBSECTION] Our study had several shortcomings that should be taken into consideration when interpreting the results. We based our analysis on unadjusted data. This means that confounders such as age, BRAF mutation status, prior systemic therapy, PD-L1 status, and gender were not considered in this meta-analysis. This might be the result of the low number of studies existing in the field and included in this meta-analysis. An increase in the number of studies published in this field and also adjustments for confounders mentioned above might result in more accurate outcomes. Besides, this study was limited to English full-text original papers. This results in missing data emerging from conferences and also other languages. Further, the existence of an open-labeled clinical trial, and also pharmaceutical companies-funded studies increased the ROB in our study.\nOur study had several shortcomings that should be taken into consideration when interpreting the results. We based our analysis on unadjusted data. This means that confounders such as age, BRAF mutation status, prior systemic therapy, PD-L1 status, and gender were not considered in this meta-analysis. This might be the result of the low number of studies existing in the field and included in this meta-analysis. An increase in the number of studies published in this field and also adjustments for confounders mentioned above might result in more accurate outcomes. Besides, this study was limited to English full-text original papers. This results in missing data emerging from conferences and also other languages. Further, the existence of an open-labeled clinical trial, and also pharmaceutical companies-funded studies increased the ROB in our study.", "Our pooled analysis was in total agreement with the results of individual trials included in this study. In that light, Larkin et al, showed (CheckMate 067) that the median OS was over 60.0 months in the combination therapy group as opposed to 36.9 and 19.9 months in the nivolumab and ipilimumab groups, respectively. Also, the hazard ratio (HR) for death was found to be lower in the combination therapy group than in ipilimumab alone (nivolumab plus ipilimumab vs. ipilimumab, 0.52; HR for death with nivolumab vs. ipilimumab, 0.63). Five-year follow-up of the patients showed a 52% OS in the combination therapy group as compared with nivolumab or ipilimumab alone (44% and 26%, respectively). The authors reported no new late toxic effects nor sustained deterioration of health-related quality of life in the combination group compared with the monotherapies.[1] Similarly, in another study by Hodi et al, (CheckMate 069) it was found that OS rates in all randomized patients were 63·8% (95% CI: 53.3–72.6) for the combination therapy group vs 53·6% (95% CI: 38.1–66.8) for ipilimumab alone at a median follow-up of 24 months. However, grade 3–4 adverse events associated with combination therapy were observed in 51 [54%] of 94 patients vs 9 [20%] of 46 patients linked to ipilimumab alone. The results of this study indicated that the combination of nivolumab plus ipilimumab may result in a higher OS rate vs ipilimumab in patients with advanced melanoma.[19] In line with previous studies, in a multicenter retrospective cohort study Zimmer et al, found that OS rates for the monotherapy and the combination groups were 16% and 21%, respectively. The disease control rate was 42% for the monotherapy and 33% for the combination groups. One-year OS rates for the monotherapy and the combination groups were 54% and 55%, respectively. The authors, however, stated that the combination therapy with nivolumab and ipilimumab was significantly less effective in patients with advanced melanoma with previous anti-PD failure compared with treatment-naïve melanoma patients.[18] This was, in contrast, to da Silva et al findings, which showed that in advanced melanoma patients who were resistant to anti-PD-1 monotherapy, combination therapy had higher efficacy than monotherapy with a higher ORR (60 [31%] of 193 patients vs 21 [13%] of 162 patients; P < .0001), longer PFS (median 3.0 months [95% CI: 2.6–3.6] vs 2.6 months [2.4–2.9]; HR, 0·69, 95% CI: 0·55–0·87; P = .0019), and longer OS (median overall survival 20·4 months [95% CI: 12·7–34·8] vs 8·8 months [6·1–11·3]; HR, 0·50, 95% CI: 0.38–0.66; P < .0001), with a similar rate of grade 3–5 toxicity. The results of this study suggested combination therapy with ipilimumab and nivolumab as a superior option over ipilimumab monotherapy in advanced melanoma patients.[16] Long et al found that combination therapy with nivolumab plus ipilimumab results in higher an intracranial response than ipilimumab monotherapy (16 (46%; 95% CI: 29–63) of 35 patients, 5 (20%; 7–41) of 25, and 1 (6%; 0–30) of 16, respectively) in patients with metastatic melanoma to the brain.[17]\nA meta-analysis by Menshawy et al[20] performed on 1910 patients (nivolumab group, n = 1207 and control group, n = 703) showed that combination therapy with nivolumab plus ipilimumab had higher ORR [RR: 3.58, 95% CI: 2.08–6.14], complete response rate (RR: 5.93, 95% CI: 2.45–14.37), partial response rate (RR: 2.80, 95% CI: 2.16–3.64), stable disease rate (RR: 0.56, 95% CI: 0.41–0.76), and PFS (hazard ratio: 0.67, 95% CI: 0.60–0.74) compared with ipilimumab monotherapy. However, this meta-analysis is rather old and did not include the 5-year update from the CheckMate 067 study by Larkin et al,[1] and also recent studies by da Silva et al[16] and Long et al.[17] On the other hand, this study included a study by Postow et al,[21] which is an earlier version of the CheckMate 069 study by Hodi et al[19] and should not be included in the meta-analysis as a separate entity. Another meta-analysis by Hao et al performed on two studies showed that combination therapy with nivolumab plus ipilimumab had a visible significant advantage over the ipilimumab monotherapy.[12] But the number of studies was too low to decide on anything.", "One of the main concerns with immune checkpoint inhibitors is the risk of immune-related adverse events (IrAEs). Nivolumab was shown to be well tolerated by patients with advanced melanoma. On the one hand, it is speculated that combination therapy with immunotherapies elevates the incidence of potential IrAEs. In a systematic review and meta-analysis by Almutairi et al., it was found that the incidences of potential IrAEs in combination therapies vs. monotherapies were higher for most types of IrAEs. This caused hyperglycemia, thyroid, hepatic, and musculoskeletal disorders in the patients receiving these medications.[22] On the other hand, it was argued that the emergence of both early and late IrAEs could be regarded as a favorable prognostic parameter in patients receiving immunotherapy and immunoradiotherapy for solid tumors especially if these events are delayed; as they are accompanied by increased overall response rate and improved OS and PFS in these patients.[23,24] In any case, the oncologist should take into consideration the occurrence of IrAEs in patients with advanced melanoma especially those with a history of autoimmune disease, poor kidney function of grade 3 or greater, and use of CTLA-4 inhibitors.[25]", "Our study had several shortcomings that should be taken into consideration when interpreting the results. We based our analysis on unadjusted data. This means that confounders such as age, BRAF mutation status, prior systemic therapy, PD-L1 status, and gender were not considered in this meta-analysis. This might be the result of the low number of studies existing in the field and included in this meta-analysis. An increase in the number of studies published in this field and also adjustments for confounders mentioned above might result in more accurate outcomes. Besides, this study was limited to English full-text original papers. This results in missing data emerging from conferences and also other languages. Further, the existence of an open-labeled clinical trial, and also pharmaceutical companies-funded studies increased the ROB in our study.", "RZH, XLZ, JML, YJZ, XCZ, and FC performed the statistical analysis and wrote the manuscript. RZH, XLZ, JML, and YJZ conceived the study design and participated in the manuscript writing. XCZ and FC revised and edited the manuscript. All authors read the final manuscript." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Materials and Methods", "2.1. Literature search", "2.2. Inclusion and exclusion criteria", "2.3. Data extraction", "2.4. Quality assessment", "2.5. Statistics", "3. Results", "3.1. General characteristics of the included studies", "3.2. Efficacy", "3.3. Nivolumab plus ipilimumab vs. nivolumab", "3.4. Nivolumab plus ipilimumab vs. ipilimumab", "3.5. Safety analysis", "3.6. Publication bias assessment", "4. Discussions", "4.1. A closer look at individual studies", "4.2. Safety", "4.3. Limitations", "5. Conclusions", "Author contributions" ]
[ "Metastatic melanoma treatment has drastically changed during the past decade with the advent of immunotherapy and then molecular targeted therapy. Today, 5-year survival is achievable in almost 50% of the patients with metastatic melanoma when treated with combination immunotherapy.[1] This is in contrast to 10 years ago when metastatic melanoma was considered unvaryingly lethal with an overall survival rate of less than 5%.[2]\nThe last decade has observed a significant change in the treatment of metastatic or unresectable melanoma patients, with the advent of immune checkpoint blockade proteins including ipilimumab, nivolumab, pembrolizumab, and combination ipilimumab-nivolumab. These antibodies are coinhibitory protein receptors (PD-1 and CTLA-4 coinhibitory receptors) which are located on the surface of the lymphocytes.[3] Their ligands (e.g., PD-L1/PD-L2 and B7, respectively), on the other hand, are expressed on the tumor cells and restrain T-cells function rendering them unable to mount a response against cancer cells and causing resistance of malignant melanoma in many patients to conventional anticancer therapy.[4]\nIn 2010, Hodi et al, showed, for the first time, that overall survival in patients with metastatic melanoma improved with the treatment via the anti–CTLA-4 antibody (ipilimumab).[5] Also, data on 1861 patients across 12 trials treated with ipilimumab revealed a 3-year survival rate of approximately 20% (plateaued afterward, supporting the durability of response to CTLA-4 blockade).[6] Accordingly, monoclonal antibodies targeting PD-1 were developed which demonstrated clinical activity in melanoma[7] with an even higher overall response rate (30%–40% at 5 years) compared to CTLA-4 blockade and ongoing durable responses in 70%–80% of responding patients.[8] Subsequently, dual immune checkpoint blockade (ICB) with ipilimumab-nivolumab was introduced and demonstrated considerable enhancements in response rate (58%) compared with ipilimumab or nivolumab alone in patients with advanced melanoma. Besides, both nivolumab-containing arms demonstrated superior overall survival (OS) compared with ipilimumab alone.[9] More data emerging from five-year follow-up verified a substantial OS where more than half of patients (52%) in the ICB group were still alive at the time of assessment. The median treatment-free interval was also demonstrably high reaching 18.1 months, highlighting the durability of these responses.[1] Nevertheless, this was accompanied by significant toxicity from dual ICB resulting in treatment interruption or discontinuation in more than 50% of the patients.[10] Nivolumab plus ipilimumab showed a high survival rate and safety outcomes in other studies, further backing up the application of this combination for advanced melanoma in multiple subgroups.[11]\nA meta-analysis of two studies showed that nivolumab-plus ipilimumab combination therapy had an obvious significant advantage over the ipilimumab monotherapy in patients with advanced melanoma.[12] Considering the increase in the number of studies performed in the field, we conducted a meta-analysis, to assess PD-1 and CTLA-4 inhibitors in combination Vs. alone for the treatment of advanced melanoma.", "This review was conducted according to a predetermined protocol based on the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement.[13]\n[SUBTITLE] 2.1. Literature search [SUBSECTION] The EMBASE, Medline via PubMed, Scopus, Cochrane Central, and Web of Science databases were electronically searched using the following terms “anti-PD-1” “nivolumab,” “pembrolizumab,” “anti-CTLA-4,” “’ ipilimumab,” and “melanoma” and the following search strategy ((((((PD-1[Title/Abstract]) OR (pembrolizumab [Title/Abstract])) OR (nivolumab [Title/Abstract])) AND (ctla-4[Title/Abstract])) OR (ipilimumab [Title/Abstract]))) AND (melanoma [Title/Abstract]). Two independent reviewers screened the titles and abstracts of retrieved citations. Accordingly, the full texts of the retrieved papers were screened and included only when they fulfilled our criteria. The electronic search was accompanied by manual searches for references to the included studies and related citations. Discrepancies were resolved by discussion between reviewers or by a third senior author. Only original research studies in English were considered. The search had no time restriction.\nThe EMBASE, Medline via PubMed, Scopus, Cochrane Central, and Web of Science databases were electronically searched using the following terms “anti-PD-1” “nivolumab,” “pembrolizumab,” “anti-CTLA-4,” “’ ipilimumab,” and “melanoma” and the following search strategy ((((((PD-1[Title/Abstract]) OR (pembrolizumab [Title/Abstract])) OR (nivolumab [Title/Abstract])) AND (ctla-4[Title/Abstract])) OR (ipilimumab [Title/Abstract]))) AND (melanoma [Title/Abstract]). Two independent reviewers screened the titles and abstracts of retrieved citations. Accordingly, the full texts of the retrieved papers were screened and included only when they fulfilled our criteria. The electronic search was accompanied by manual searches for references to the included studies and related citations. Discrepancies were resolved by discussion between reviewers or by a third senior author. Only original research studies in English were considered. The search had no time restriction.\n[SUBTITLE] 2.2. Inclusion and exclusion criteria [SUBSECTION] English full-text randomized controlled trials (RCTs) that fulfilled the following criteria were included in this meta-analysis: patients with advanced stage III or stage IV melanoma, in whom nivolumab or ipilimumab was administered alone (control) or in combination (intervention), and assessed progression-free survival (PFS) rate, overall survival (OS; the time from the initiation of treatment until death), complete response rate, partial response rate, objective response rate (ORR), stable disease rate, and safety measures as their outcomes. On the other hand, non-English articles, nonoriginal articles, thesis or conference papers that were never subsequently published, and studies on animals (in vivo) and cell lines (in vitro) were excluded from this study.\nEnglish full-text randomized controlled trials (RCTs) that fulfilled the following criteria were included in this meta-analysis: patients with advanced stage III or stage IV melanoma, in whom nivolumab or ipilimumab was administered alone (control) or in combination (intervention), and assessed progression-free survival (PFS) rate, overall survival (OS; the time from the initiation of treatment until death), complete response rate, partial response rate, objective response rate (ORR), stable disease rate, and safety measures as their outcomes. On the other hand, non-English articles, nonoriginal articles, thesis or conference papers that were never subsequently published, and studies on animals (in vivo) and cell lines (in vitro) were excluded from this study.\n[SUBTITLE] 2.3. Data extraction [SUBSECTION] Data extraction was independently conducted by two reviewers using a standardized approach. Disagreements were resolved by discussion with a third senior reviewer. Data on authors’ names, year of publication, journal name, study phase, the sample size in each arm, immunotherapy regimen used, the mean age of patients, and information regarding study design (randomization, allocation concealment, description of withdrawals per arm, and blinding) for the trials included in the study.\nData extraction was independently conducted by two reviewers using a standardized approach. Disagreements were resolved by discussion with a third senior reviewer. Data on authors’ names, year of publication, journal name, study phase, the sample size in each arm, immunotherapy regimen used, the mean age of patients, and information regarding study design (randomization, allocation concealment, description of withdrawals per arm, and blinding) for the trials included in the study.\n[SUBTITLE] 2.4. Quality assessment [SUBSECTION] Cochrane Collaboration’s tool for risk of bias (ROB) assessment was used to assess the quality of the included studies. The items of this tool were as follows; allocation concealment, selective outcome reporting, blinding of participants, masking of outcome assessors, generation of allocation sequence, incomplete follow-up, and other potential sources of bias. A senior author judged any disagreements between the 2 reviewers who performed the assessment. For each element, the risk of bias was considered as low, unclear, or high. If the reviewer could find information on all the parameters mentioned in the tool or no information at all, then the study was allocated to one of the low bias or high bias categories, respectively. If the information retrieved by the reviewer was partial or unclear, the risk of bias was considered to be unclear.[14]\nCochrane Collaboration’s tool for risk of bias (ROB) assessment was used to assess the quality of the included studies. The items of this tool were as follows; allocation concealment, selective outcome reporting, blinding of participants, masking of outcome assessors, generation of allocation sequence, incomplete follow-up, and other potential sources of bias. A senior author judged any disagreements between the 2 reviewers who performed the assessment. For each element, the risk of bias was considered as low, unclear, or high. If the reviewer could find information on all the parameters mentioned in the tool or no information at all, then the study was allocated to one of the low bias or high bias categories, respectively. If the information retrieved by the reviewer was partial or unclear, the risk of bias was considered to be unclear.[14]\n[SUBTITLE] 2.5. Statistics [SUBSECTION] The Comprehensive Meta-Analysis Software (CMA) software version 2.0 was used to analyze the data. Odds ratio (OR) was applied to compare dichotomous variables. All the results were reported with 95% confidence intervals (CI). Mantel–Haenszel method was used to estimate pooled OR and 95% confidence intervals for dichotomous data. Heterogeneity of the data was assessed using I-square (I2) test and considered high if I2>50%. In this case, the random effect model was chosen; otherwise, the fixed effect model was used. Subgroup analyses were conducted based on the intervention of the study design: anti-PD-1 plus anti-CLTA-4 versus anti-PD-1 (nivolumab), and anti-PD-1 plus anti-CLTA-4 versus anti-CLTA-4 (ipilimumab). Funnel plotting, Egger’s regression, and trim and fill were not used for the assessment of publication bias in this literature as the number of studies was less than 10. P < .05 is considered statistically significant.\nThe Comprehensive Meta-Analysis Software (CMA) software version 2.0 was used to analyze the data. Odds ratio (OR) was applied to compare dichotomous variables. All the results were reported with 95% confidence intervals (CI). Mantel–Haenszel method was used to estimate pooled OR and 95% confidence intervals for dichotomous data. Heterogeneity of the data was assessed using I-square (I2) test and considered high if I2>50%. In this case, the random effect model was chosen; otherwise, the fixed effect model was used. Subgroup analyses were conducted based on the intervention of the study design: anti-PD-1 plus anti-CLTA-4 versus anti-PD-1 (nivolumab), and anti-PD-1 plus anti-CLTA-4 versus anti-CLTA-4 (ipilimumab). Funnel plotting, Egger’s regression, and trim and fill were not used for the assessment of publication bias in this literature as the number of studies was less than 10. P < .05 is considered statistically significant.", "The EMBASE, Medline via PubMed, Scopus, Cochrane Central, and Web of Science databases were electronically searched using the following terms “anti-PD-1” “nivolumab,” “pembrolizumab,” “anti-CTLA-4,” “’ ipilimumab,” and “melanoma” and the following search strategy ((((((PD-1[Title/Abstract]) OR (pembrolizumab [Title/Abstract])) OR (nivolumab [Title/Abstract])) AND (ctla-4[Title/Abstract])) OR (ipilimumab [Title/Abstract]))) AND (melanoma [Title/Abstract]). Two independent reviewers screened the titles and abstracts of retrieved citations. Accordingly, the full texts of the retrieved papers were screened and included only when they fulfilled our criteria. The electronic search was accompanied by manual searches for references to the included studies and related citations. Discrepancies were resolved by discussion between reviewers or by a third senior author. Only original research studies in English were considered. The search had no time restriction.", "English full-text randomized controlled trials (RCTs) that fulfilled the following criteria were included in this meta-analysis: patients with advanced stage III or stage IV melanoma, in whom nivolumab or ipilimumab was administered alone (control) or in combination (intervention), and assessed progression-free survival (PFS) rate, overall survival (OS; the time from the initiation of treatment until death), complete response rate, partial response rate, objective response rate (ORR), stable disease rate, and safety measures as their outcomes. On the other hand, non-English articles, nonoriginal articles, thesis or conference papers that were never subsequently published, and studies on animals (in vivo) and cell lines (in vitro) were excluded from this study.", "Data extraction was independently conducted by two reviewers using a standardized approach. Disagreements were resolved by discussion with a third senior reviewer. Data on authors’ names, year of publication, journal name, study phase, the sample size in each arm, immunotherapy regimen used, the mean age of patients, and information regarding study design (randomization, allocation concealment, description of withdrawals per arm, and blinding) for the trials included in the study.", "Cochrane Collaboration’s tool for risk of bias (ROB) assessment was used to assess the quality of the included studies. The items of this tool were as follows; allocation concealment, selective outcome reporting, blinding of participants, masking of outcome assessors, generation of allocation sequence, incomplete follow-up, and other potential sources of bias. A senior author judged any disagreements between the 2 reviewers who performed the assessment. For each element, the risk of bias was considered as low, unclear, or high. If the reviewer could find information on all the parameters mentioned in the tool or no information at all, then the study was allocated to one of the low bias or high bias categories, respectively. If the information retrieved by the reviewer was partial or unclear, the risk of bias was considered to be unclear.[14]", "The Comprehensive Meta-Analysis Software (CMA) software version 2.0 was used to analyze the data. Odds ratio (OR) was applied to compare dichotomous variables. All the results were reported with 95% confidence intervals (CI). Mantel–Haenszel method was used to estimate pooled OR and 95% confidence intervals for dichotomous data. Heterogeneity of the data was assessed using I-square (I2) test and considered high if I2>50%. In this case, the random effect model was chosen; otherwise, the fixed effect model was used. Subgroup analyses were conducted based on the intervention of the study design: anti-PD-1 plus anti-CLTA-4 versus anti-PD-1 (nivolumab), and anti-PD-1 plus anti-CLTA-4 versus anti-CLTA-4 (ipilimumab). Funnel plotting, Egger’s regression, and trim and fill were not used for the assessment of publication bias in this literature as the number of studies was less than 10. P < .05 is considered statistically significant.", "[SUBTITLE] 3.1. General characteristics of the included studies [SUBSECTION] We retrieved 3092 citations, including 3088 publications by electronic search of databases, and 4 studies by a manual search of websites and checking reference lists of the included studies. After the removal of duplicate records, a total of 2839 titles were screened, during which 2779 articles were excluded. Accordingly, and after abstract and full-text screening, 3 randomized controlled trials and 2 retrospective, cohort studies were included in this meta-analysis. The detailed information regarding the number of identified studies, and the stages of evaluation and exclusion is presented in Figure 1 (see PRISMA flow diagram).\nPRISMA flowchart of the included studies in the systematic review and meta-analysis.\nThe characteristics of the included studies are presented in Table 1. Of the included RCTs, one publication was from phase III and two citations were from phase II. A total of 1605 patients were included in this meta-analysis of which 675 were in the combination therapy group, 359 in the nivolumab monotherapy group, and 571 in the ipilimumab monotherapy group. PFS was the main endpoint in 4 of the included citations and OS was the primary outcome in 4 of the included papers. In all of the included studies, PFS and OS were higher in the combination therapy group (nivolumab plus ipilimumab) than those of the monotherapy groups (nivolumab or ipilimumab).\nCharacteristics of included studies in the meta-analysis.\nIpi = ipilimumab, ORR =objective response rate, OS = overall survival, PFS = progression-free survival.\nWe retrieved 3092 citations, including 3088 publications by electronic search of databases, and 4 studies by a manual search of websites and checking reference lists of the included studies. After the removal of duplicate records, a total of 2839 titles were screened, during which 2779 articles were excluded. Accordingly, and after abstract and full-text screening, 3 randomized controlled trials and 2 retrospective, cohort studies were included in this meta-analysis. The detailed information regarding the number of identified studies, and the stages of evaluation and exclusion is presented in Figure 1 (see PRISMA flow diagram).\nPRISMA flowchart of the included studies in the systematic review and meta-analysis.\nThe characteristics of the included studies are presented in Table 1. Of the included RCTs, one publication was from phase III and two citations were from phase II. A total of 1605 patients were included in this meta-analysis of which 675 were in the combination therapy group, 359 in the nivolumab monotherapy group, and 571 in the ipilimumab monotherapy group. PFS was the main endpoint in 4 of the included citations and OS was the primary outcome in 4 of the included papers. In all of the included studies, PFS and OS were higher in the combination therapy group (nivolumab plus ipilimumab) than those of the monotherapy groups (nivolumab or ipilimumab).\nCharacteristics of included studies in the meta-analysis.\nIpi = ipilimumab, ORR =objective response rate, OS = overall survival, PFS = progression-free survival.\n[SUBTITLE] 3.2. Efficacy [SUBSECTION] The main endpoints of the included studies were ORR, PFS, and OS for efficacy. However, not all of the included studies reported all of these endpoints. For example, ORR was reported only by one of the included studies. Subgroup analyses were conducted based on the intervention of the study design. The heterogeneity of the included studies was found to be high thus random effect model was used for the analyses. The PFS and/or OS of these trials are presented in Table 1. Overall analysis showed that the pooled OR was 2.144 (95% CI: 1.650–2.786, I2 = 80.38% P = .000; Fig. 2) for overall response and 2.117 (95% CI: 1.578–2.841, I2 = 70.17% P = .000; Fig. 3) for the complete response (CR).\nForest plot of standardized mean difference (SMD) for overall response for combination therapy vs. both ipilimumab and nivolumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Red squares show pooled effect in each subgroup. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).\nForest plot of standardized mean difference (SMD) for complete response for combination therapy vs. both ipilimumab and nivolumab therapy in patients with advanced melanoma. Green square shows overall pooled effect. Red squares show pooled effect in each subgroup. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).\nThe main endpoints of the included studies were ORR, PFS, and OS for efficacy. However, not all of the included studies reported all of these endpoints. For example, ORR was reported only by one of the included studies. Subgroup analyses were conducted based on the intervention of the study design. The heterogeneity of the included studies was found to be high thus random effect model was used for the analyses. The PFS and/or OS of these trials are presented in Table 1. Overall analysis showed that the pooled OR was 2.144 (95% CI: 1.650–2.786, I2 = 80.38% P = .000; Fig. 2) for overall response and 2.117 (95% CI: 1.578–2.841, I2 = 70.17% P = .000; Fig. 3) for the complete response (CR).\nForest plot of standardized mean difference (SMD) for overall response for combination therapy vs. both ipilimumab and nivolumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Red squares show pooled effect in each subgroup. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).\nForest plot of standardized mean difference (SMD) for complete response for combination therapy vs. both ipilimumab and nivolumab therapy in patients with advanced melanoma. Green square shows overall pooled effect. Red squares show pooled effect in each subgroup. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).\n[SUBTITLE] 3.3. Nivolumab plus ipilimumab vs. nivolumab [SUBSECTION] Subgroup analysis in this category showed that the pooled OR was 1.766 (95% CI: 1.324–2.355, I2 = 0.0% P = .000; Fig. 2) for the overall response and was 1.284 (95% CI: 0.889–1.855, I2 = 0.0% P = .182; Fig. 3) for the CR. These findings indicated a significantly higher overall response but not CR and thus beneficial for the patients in the combination therapy group compared with the nivolumab group in the included population. Also, in the combination therapy group, significantly longer OS and PFS were observed in each of the included studies. However, analysis was not possible for these variables (Table 1).\nSubgroup analysis in this category showed that the pooled OR was 1.766 (95% CI: 1.324–2.355, I2 = 0.0% P = .000; Fig. 2) for the overall response and was 1.284 (95% CI: 0.889–1.855, I2 = 0.0% P = .182; Fig. 3) for the CR. These findings indicated a significantly higher overall response but not CR and thus beneficial for the patients in the combination therapy group compared with the nivolumab group in the included population. Also, in the combination therapy group, significantly longer OS and PFS were observed in each of the included studies. However, analysis was not possible for these variables (Table 1).\n[SUBTITLE] 3.4. Nivolumab plus ipilimumab vs. ipilimumab [SUBSECTION] Subgroup analysis in this category revealed that the pooled OR was 5.440 (95% CI: 2.896–10.220, I2 = 70.89% P = .001; Fig. 2) for the overall response and was 5.169 (95% CI: 3.163–8.446, I2 = 0.0% P = .000; Fig. 3) for the complete response (CR). These findings indicated a significantly higher overall response, CR, and thus beneficial for the patients in the combination therapy group compared with the ipilimumab group in the included population. Also, in the combination therapy group, significantly longer OS and PFS were observed in each of the included studies. However, analysis was not possible for these variables (Table 1).\nSubgroup analysis in this category revealed that the pooled OR was 5.440 (95% CI: 2.896–10.220, I2 = 70.89% P = .001; Fig. 2) for the overall response and was 5.169 (95% CI: 3.163–8.446, I2 = 0.0% P = .000; Fig. 3) for the complete response (CR). These findings indicated a significantly higher overall response, CR, and thus beneficial for the patients in the combination therapy group compared with the ipilimumab group in the included population. Also, in the combination therapy group, significantly longer OS and PFS were observed in each of the included studies. However, analysis was not possible for these variables (Table 1).\n[SUBTITLE] 3.5. Safety analysis [SUBSECTION] Larkin et al showed that 59%, 23%, and 28% of patients in the nivolumab-plus-ipilimumab, nivolumab, and ipilimumab groups, respectively, had grade 3 or 4 treatment-related side effects.[1] Hodi et al showed that at the time of the most recent data lock, the rates of treatment-related adverse events of any grade were 92% (86 of 94 patients) and 94 percent (43 of 46 patients), respectively. The most common adverse reactions to therapy were diarrhea, rash, fatigue, and pruritus in both groups. Consistent with Larkin et al, study, Hodi et al, showed that the frequency of grade 3–4 adverse events was higher in the combination therapy group.[15] However, da Silva et al, found that the rate of grade 3–4 adverse events was similar between the groups. Diarrhea or colitis was the most frequent grade 3–5 treatment-related adverse event, followed by a rise in alanine aminotransferase or aspartate aminotransferase in this study.[16]\nOur analysis revealed that the incidence of any treatment-related adverse events was significantly higher in the combination group than that of the nivolumab monotherapy 4.044 (95% CI: 1.740–9.403, I2 = 91.64% P = .001; Fig. 4). However, the heterogeneity of the included studies was high. Also, the incidence of any treatment-related adverse events was higher in the combination group than that of the ipilimumab monotherapy 2.465 (95% CI: 0.839–7.236, I2 = 93.02% P = .101; Fig. 5). However, this did not reach a statistical significance and the heterogeneity of the included studies was modest.\nForest plot of standardized mean difference (SMD) for treatment related-adverse event for combination therapy vs. nivolumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).\nForest plot of standardized mean difference (SMD) for treatment related-adverse event for combination therapy vs. ipilimumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).\nLarkin et al showed that 59%, 23%, and 28% of patients in the nivolumab-plus-ipilimumab, nivolumab, and ipilimumab groups, respectively, had grade 3 or 4 treatment-related side effects.[1] Hodi et al showed that at the time of the most recent data lock, the rates of treatment-related adverse events of any grade were 92% (86 of 94 patients) and 94 percent (43 of 46 patients), respectively. The most common adverse reactions to therapy were diarrhea, rash, fatigue, and pruritus in both groups. Consistent with Larkin et al, study, Hodi et al, showed that the frequency of grade 3–4 adverse events was higher in the combination therapy group.[15] However, da Silva et al, found that the rate of grade 3–4 adverse events was similar between the groups. Diarrhea or colitis was the most frequent grade 3–5 treatment-related adverse event, followed by a rise in alanine aminotransferase or aspartate aminotransferase in this study.[16]\nOur analysis revealed that the incidence of any treatment-related adverse events was significantly higher in the combination group than that of the nivolumab monotherapy 4.044 (95% CI: 1.740–9.403, I2 = 91.64% P = .001; Fig. 4). However, the heterogeneity of the included studies was high. Also, the incidence of any treatment-related adverse events was higher in the combination group than that of the ipilimumab monotherapy 2.465 (95% CI: 0.839–7.236, I2 = 93.02% P = .101; Fig. 5). However, this did not reach a statistical significance and the heterogeneity of the included studies was modest.\nForest plot of standardized mean difference (SMD) for treatment related-adverse event for combination therapy vs. nivolumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).\nForest plot of standardized mean difference (SMD) for treatment related-adverse event for combination therapy vs. ipilimumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).\n[SUBTITLE] 3.6. Publication bias assessment [SUBSECTION] The quality of included studies was evaluated based on the standards of RCT quality assessment of the Cochrane Reviewer handbook.[14] There was no attrition bias and reporting bias in the included studies. However, detection, performance, and selection (allocation concealment) biases were found in three of the included citations.[16–18] Also, random sequence generation was not implemented in two of the included publications[16,18] (Fig. 6).\nDifferent levels of risk of bias for each item in included studies. The Cochrane risk of bias tool was used for the detection of publication bias.\nThe quality of included studies was evaluated based on the standards of RCT quality assessment of the Cochrane Reviewer handbook.[14] There was no attrition bias and reporting bias in the included studies. However, detection, performance, and selection (allocation concealment) biases were found in three of the included citations.[16–18] Also, random sequence generation was not implemented in two of the included publications[16,18] (Fig. 6).\nDifferent levels of risk of bias for each item in included studies. The Cochrane risk of bias tool was used for the detection of publication bias.", "We retrieved 3092 citations, including 3088 publications by electronic search of databases, and 4 studies by a manual search of websites and checking reference lists of the included studies. After the removal of duplicate records, a total of 2839 titles were screened, during which 2779 articles were excluded. Accordingly, and after abstract and full-text screening, 3 randomized controlled trials and 2 retrospective, cohort studies were included in this meta-analysis. The detailed information regarding the number of identified studies, and the stages of evaluation and exclusion is presented in Figure 1 (see PRISMA flow diagram).\nPRISMA flowchart of the included studies in the systematic review and meta-analysis.\nThe characteristics of the included studies are presented in Table 1. Of the included RCTs, one publication was from phase III and two citations were from phase II. A total of 1605 patients were included in this meta-analysis of which 675 were in the combination therapy group, 359 in the nivolumab monotherapy group, and 571 in the ipilimumab monotherapy group. PFS was the main endpoint in 4 of the included citations and OS was the primary outcome in 4 of the included papers. In all of the included studies, PFS and OS were higher in the combination therapy group (nivolumab plus ipilimumab) than those of the monotherapy groups (nivolumab or ipilimumab).\nCharacteristics of included studies in the meta-analysis.\nIpi = ipilimumab, ORR =objective response rate, OS = overall survival, PFS = progression-free survival.", "The main endpoints of the included studies were ORR, PFS, and OS for efficacy. However, not all of the included studies reported all of these endpoints. For example, ORR was reported only by one of the included studies. Subgroup analyses were conducted based on the intervention of the study design. The heterogeneity of the included studies was found to be high thus random effect model was used for the analyses. The PFS and/or OS of these trials are presented in Table 1. Overall analysis showed that the pooled OR was 2.144 (95% CI: 1.650–2.786, I2 = 80.38% P = .000; Fig. 2) for overall response and 2.117 (95% CI: 1.578–2.841, I2 = 70.17% P = .000; Fig. 3) for the complete response (CR).\nForest plot of standardized mean difference (SMD) for overall response for combination therapy vs. both ipilimumab and nivolumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Red squares show pooled effect in each subgroup. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).\nForest plot of standardized mean difference (SMD) for complete response for combination therapy vs. both ipilimumab and nivolumab therapy in patients with advanced melanoma. Green square shows overall pooled effect. Red squares show pooled effect in each subgroup. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).", "Subgroup analysis in this category showed that the pooled OR was 1.766 (95% CI: 1.324–2.355, I2 = 0.0% P = .000; Fig. 2) for the overall response and was 1.284 (95% CI: 0.889–1.855, I2 = 0.0% P = .182; Fig. 3) for the CR. These findings indicated a significantly higher overall response but not CR and thus beneficial for the patients in the combination therapy group compared with the nivolumab group in the included population. Also, in the combination therapy group, significantly longer OS and PFS were observed in each of the included studies. However, analysis was not possible for these variables (Table 1).", "Subgroup analysis in this category revealed that the pooled OR was 5.440 (95% CI: 2.896–10.220, I2 = 70.89% P = .001; Fig. 2) for the overall response and was 5.169 (95% CI: 3.163–8.446, I2 = 0.0% P = .000; Fig. 3) for the complete response (CR). These findings indicated a significantly higher overall response, CR, and thus beneficial for the patients in the combination therapy group compared with the ipilimumab group in the included population. Also, in the combination therapy group, significantly longer OS and PFS were observed in each of the included studies. However, analysis was not possible for these variables (Table 1).", "Larkin et al showed that 59%, 23%, and 28% of patients in the nivolumab-plus-ipilimumab, nivolumab, and ipilimumab groups, respectively, had grade 3 or 4 treatment-related side effects.[1] Hodi et al showed that at the time of the most recent data lock, the rates of treatment-related adverse events of any grade were 92% (86 of 94 patients) and 94 percent (43 of 46 patients), respectively. The most common adverse reactions to therapy were diarrhea, rash, fatigue, and pruritus in both groups. Consistent with Larkin et al, study, Hodi et al, showed that the frequency of grade 3–4 adverse events was higher in the combination therapy group.[15] However, da Silva et al, found that the rate of grade 3–4 adverse events was similar between the groups. Diarrhea or colitis was the most frequent grade 3–5 treatment-related adverse event, followed by a rise in alanine aminotransferase or aspartate aminotransferase in this study.[16]\nOur analysis revealed that the incidence of any treatment-related adverse events was significantly higher in the combination group than that of the nivolumab monotherapy 4.044 (95% CI: 1.740–9.403, I2 = 91.64% P = .001; Fig. 4). However, the heterogeneity of the included studies was high. Also, the incidence of any treatment-related adverse events was higher in the combination group than that of the ipilimumab monotherapy 2.465 (95% CI: 0.839–7.236, I2 = 93.02% P = .101; Fig. 5). However, this did not reach a statistical significance and the heterogeneity of the included studies was modest.\nForest plot of standardized mean difference (SMD) for treatment related-adverse event for combination therapy vs. nivolumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).\nForest plot of standardized mean difference (SMD) for treatment related-adverse event for combination therapy vs. ipilimumab therapy in patients with advanced melanoma. The green square shows the overall pooled effect. Black squares indicate the SMD in each study. Horizontal lines represent a 95% confidence interval (CI).", "The quality of included studies was evaluated based on the standards of RCT quality assessment of the Cochrane Reviewer handbook.[14] There was no attrition bias and reporting bias in the included studies. However, detection, performance, and selection (allocation concealment) biases were found in three of the included citations.[16–18] Also, random sequence generation was not implemented in two of the included publications[16,18] (Fig. 6).\nDifferent levels of risk of bias for each item in included studies. The Cochrane risk of bias tool was used for the detection of publication bias.", "The results emerging from this systematic review and meta-analysis showed that combination therapy with nivolumab plus ipilimumab was more effective than nivolumab or ipilimumab alone in the treatment of advanced metastatic melanoma (treated or untreated). This was suggested by higher overall response and CR of the combination therapy compared with either of the monotherapies. However, this was accompanied by a higher incidence of grade 3/4/5 adverse events in the combination therapy than in either of the monotherapies.\n[SUBTITLE] 4.1. A closer look at individual studies [SUBSECTION] Our pooled analysis was in total agreement with the results of individual trials included in this study. In that light, Larkin et al, showed (CheckMate 067) that the median OS was over 60.0 months in the combination therapy group as opposed to 36.9 and 19.9 months in the nivolumab and ipilimumab groups, respectively. Also, the hazard ratio (HR) for death was found to be lower in the combination therapy group than in ipilimumab alone (nivolumab plus ipilimumab vs. ipilimumab, 0.52; HR for death with nivolumab vs. ipilimumab, 0.63). Five-year follow-up of the patients showed a 52% OS in the combination therapy group as compared with nivolumab or ipilimumab alone (44% and 26%, respectively). The authors reported no new late toxic effects nor sustained deterioration of health-related quality of life in the combination group compared with the monotherapies.[1] Similarly, in another study by Hodi et al, (CheckMate 069) it was found that OS rates in all randomized patients were 63·8% (95% CI: 53.3–72.6) for the combination therapy group vs 53·6% (95% CI: 38.1–66.8) for ipilimumab alone at a median follow-up of 24 months. However, grade 3–4 adverse events associated with combination therapy were observed in 51 [54%] of 94 patients vs 9 [20%] of 46 patients linked to ipilimumab alone. The results of this study indicated that the combination of nivolumab plus ipilimumab may result in a higher OS rate vs ipilimumab in patients with advanced melanoma.[19] In line with previous studies, in a multicenter retrospective cohort study Zimmer et al, found that OS rates for the monotherapy and the combination groups were 16% and 21%, respectively. The disease control rate was 42% for the monotherapy and 33% for the combination groups. One-year OS rates for the monotherapy and the combination groups were 54% and 55%, respectively. The authors, however, stated that the combination therapy with nivolumab and ipilimumab was significantly less effective in patients with advanced melanoma with previous anti-PD failure compared with treatment-naïve melanoma patients.[18] This was, in contrast, to da Silva et al findings, which showed that in advanced melanoma patients who were resistant to anti-PD-1 monotherapy, combination therapy had higher efficacy than monotherapy with a higher ORR (60 [31%] of 193 patients vs 21 [13%] of 162 patients; P < .0001), longer PFS (median 3.0 months [95% CI: 2.6–3.6] vs 2.6 months [2.4–2.9]; HR, 0·69, 95% CI: 0·55–0·87; P = .0019), and longer OS (median overall survival 20·4 months [95% CI: 12·7–34·8] vs 8·8 months [6·1–11·3]; HR, 0·50, 95% CI: 0.38–0.66; P < .0001), with a similar rate of grade 3–5 toxicity. The results of this study suggested combination therapy with ipilimumab and nivolumab as a superior option over ipilimumab monotherapy in advanced melanoma patients.[16] Long et al found that combination therapy with nivolumab plus ipilimumab results in higher an intracranial response than ipilimumab monotherapy (16 (46%; 95% CI: 29–63) of 35 patients, 5 (20%; 7–41) of 25, and 1 (6%; 0–30) of 16, respectively) in patients with metastatic melanoma to the brain.[17]\nA meta-analysis by Menshawy et al[20] performed on 1910 patients (nivolumab group, n = 1207 and control group, n = 703) showed that combination therapy with nivolumab plus ipilimumab had higher ORR [RR: 3.58, 95% CI: 2.08–6.14], complete response rate (RR: 5.93, 95% CI: 2.45–14.37), partial response rate (RR: 2.80, 95% CI: 2.16–3.64), stable disease rate (RR: 0.56, 95% CI: 0.41–0.76), and PFS (hazard ratio: 0.67, 95% CI: 0.60–0.74) compared with ipilimumab monotherapy. However, this meta-analysis is rather old and did not include the 5-year update from the CheckMate 067 study by Larkin et al,[1] and also recent studies by da Silva et al[16] and Long et al.[17] On the other hand, this study included a study by Postow et al,[21] which is an earlier version of the CheckMate 069 study by Hodi et al[19] and should not be included in the meta-analysis as a separate entity. Another meta-analysis by Hao et al performed on two studies showed that combination therapy with nivolumab plus ipilimumab had a visible significant advantage over the ipilimumab monotherapy.[12] But the number of studies was too low to decide on anything.\nOur pooled analysis was in total agreement with the results of individual trials included in this study. In that light, Larkin et al, showed (CheckMate 067) that the median OS was over 60.0 months in the combination therapy group as opposed to 36.9 and 19.9 months in the nivolumab and ipilimumab groups, respectively. Also, the hazard ratio (HR) for death was found to be lower in the combination therapy group than in ipilimumab alone (nivolumab plus ipilimumab vs. ipilimumab, 0.52; HR for death with nivolumab vs. ipilimumab, 0.63). Five-year follow-up of the patients showed a 52% OS in the combination therapy group as compared with nivolumab or ipilimumab alone (44% and 26%, respectively). The authors reported no new late toxic effects nor sustained deterioration of health-related quality of life in the combination group compared with the monotherapies.[1] Similarly, in another study by Hodi et al, (CheckMate 069) it was found that OS rates in all randomized patients were 63·8% (95% CI: 53.3–72.6) for the combination therapy group vs 53·6% (95% CI: 38.1–66.8) for ipilimumab alone at a median follow-up of 24 months. However, grade 3–4 adverse events associated with combination therapy were observed in 51 [54%] of 94 patients vs 9 [20%] of 46 patients linked to ipilimumab alone. The results of this study indicated that the combination of nivolumab plus ipilimumab may result in a higher OS rate vs ipilimumab in patients with advanced melanoma.[19] In line with previous studies, in a multicenter retrospective cohort study Zimmer et al, found that OS rates for the monotherapy and the combination groups were 16% and 21%, respectively. The disease control rate was 42% for the monotherapy and 33% for the combination groups. One-year OS rates for the monotherapy and the combination groups were 54% and 55%, respectively. The authors, however, stated that the combination therapy with nivolumab and ipilimumab was significantly less effective in patients with advanced melanoma with previous anti-PD failure compared with treatment-naïve melanoma patients.[18] This was, in contrast, to da Silva et al findings, which showed that in advanced melanoma patients who were resistant to anti-PD-1 monotherapy, combination therapy had higher efficacy than monotherapy with a higher ORR (60 [31%] of 193 patients vs 21 [13%] of 162 patients; P < .0001), longer PFS (median 3.0 months [95% CI: 2.6–3.6] vs 2.6 months [2.4–2.9]; HR, 0·69, 95% CI: 0·55–0·87; P = .0019), and longer OS (median overall survival 20·4 months [95% CI: 12·7–34·8] vs 8·8 months [6·1–11·3]; HR, 0·50, 95% CI: 0.38–0.66; P < .0001), with a similar rate of grade 3–5 toxicity. The results of this study suggested combination therapy with ipilimumab and nivolumab as a superior option over ipilimumab monotherapy in advanced melanoma patients.[16] Long et al found that combination therapy with nivolumab plus ipilimumab results in higher an intracranial response than ipilimumab monotherapy (16 (46%; 95% CI: 29–63) of 35 patients, 5 (20%; 7–41) of 25, and 1 (6%; 0–30) of 16, respectively) in patients with metastatic melanoma to the brain.[17]\nA meta-analysis by Menshawy et al[20] performed on 1910 patients (nivolumab group, n = 1207 and control group, n = 703) showed that combination therapy with nivolumab plus ipilimumab had higher ORR [RR: 3.58, 95% CI: 2.08–6.14], complete response rate (RR: 5.93, 95% CI: 2.45–14.37), partial response rate (RR: 2.80, 95% CI: 2.16–3.64), stable disease rate (RR: 0.56, 95% CI: 0.41–0.76), and PFS (hazard ratio: 0.67, 95% CI: 0.60–0.74) compared with ipilimumab monotherapy. However, this meta-analysis is rather old and did not include the 5-year update from the CheckMate 067 study by Larkin et al,[1] and also recent studies by da Silva et al[16] and Long et al.[17] On the other hand, this study included a study by Postow et al,[21] which is an earlier version of the CheckMate 069 study by Hodi et al[19] and should not be included in the meta-analysis as a separate entity. Another meta-analysis by Hao et al performed on two studies showed that combination therapy with nivolumab plus ipilimumab had a visible significant advantage over the ipilimumab monotherapy.[12] But the number of studies was too low to decide on anything.\n[SUBTITLE] 4.2. Safety [SUBSECTION] One of the main concerns with immune checkpoint inhibitors is the risk of immune-related adverse events (IrAEs). Nivolumab was shown to be well tolerated by patients with advanced melanoma. On the one hand, it is speculated that combination therapy with immunotherapies elevates the incidence of potential IrAEs. In a systematic review and meta-analysis by Almutairi et al., it was found that the incidences of potential IrAEs in combination therapies vs. monotherapies were higher for most types of IrAEs. This caused hyperglycemia, thyroid, hepatic, and musculoskeletal disorders in the patients receiving these medications.[22] On the other hand, it was argued that the emergence of both early and late IrAEs could be regarded as a favorable prognostic parameter in patients receiving immunotherapy and immunoradiotherapy for solid tumors especially if these events are delayed; as they are accompanied by increased overall response rate and improved OS and PFS in these patients.[23,24] In any case, the oncologist should take into consideration the occurrence of IrAEs in patients with advanced melanoma especially those with a history of autoimmune disease, poor kidney function of grade 3 or greater, and use of CTLA-4 inhibitors.[25]\nOne of the main concerns with immune checkpoint inhibitors is the risk of immune-related adverse events (IrAEs). Nivolumab was shown to be well tolerated by patients with advanced melanoma. On the one hand, it is speculated that combination therapy with immunotherapies elevates the incidence of potential IrAEs. In a systematic review and meta-analysis by Almutairi et al., it was found that the incidences of potential IrAEs in combination therapies vs. monotherapies were higher for most types of IrAEs. This caused hyperglycemia, thyroid, hepatic, and musculoskeletal disorders in the patients receiving these medications.[22] On the other hand, it was argued that the emergence of both early and late IrAEs could be regarded as a favorable prognostic parameter in patients receiving immunotherapy and immunoradiotherapy for solid tumors especially if these events are delayed; as they are accompanied by increased overall response rate and improved OS and PFS in these patients.[23,24] In any case, the oncologist should take into consideration the occurrence of IrAEs in patients with advanced melanoma especially those with a history of autoimmune disease, poor kidney function of grade 3 or greater, and use of CTLA-4 inhibitors.[25]\n[SUBTITLE] 4.3. Limitations [SUBSECTION] Our study had several shortcomings that should be taken into consideration when interpreting the results. We based our analysis on unadjusted data. This means that confounders such as age, BRAF mutation status, prior systemic therapy, PD-L1 status, and gender were not considered in this meta-analysis. This might be the result of the low number of studies existing in the field and included in this meta-analysis. An increase in the number of studies published in this field and also adjustments for confounders mentioned above might result in more accurate outcomes. Besides, this study was limited to English full-text original papers. This results in missing data emerging from conferences and also other languages. Further, the existence of an open-labeled clinical trial, and also pharmaceutical companies-funded studies increased the ROB in our study.\nOur study had several shortcomings that should be taken into consideration when interpreting the results. We based our analysis on unadjusted data. This means that confounders such as age, BRAF mutation status, prior systemic therapy, PD-L1 status, and gender were not considered in this meta-analysis. This might be the result of the low number of studies existing in the field and included in this meta-analysis. An increase in the number of studies published in this field and also adjustments for confounders mentioned above might result in more accurate outcomes. Besides, this study was limited to English full-text original papers. This results in missing data emerging from conferences and also other languages. Further, the existence of an open-labeled clinical trial, and also pharmaceutical companies-funded studies increased the ROB in our study.", "Our pooled analysis was in total agreement with the results of individual trials included in this study. In that light, Larkin et al, showed (CheckMate 067) that the median OS was over 60.0 months in the combination therapy group as opposed to 36.9 and 19.9 months in the nivolumab and ipilimumab groups, respectively. Also, the hazard ratio (HR) for death was found to be lower in the combination therapy group than in ipilimumab alone (nivolumab plus ipilimumab vs. ipilimumab, 0.52; HR for death with nivolumab vs. ipilimumab, 0.63). Five-year follow-up of the patients showed a 52% OS in the combination therapy group as compared with nivolumab or ipilimumab alone (44% and 26%, respectively). The authors reported no new late toxic effects nor sustained deterioration of health-related quality of life in the combination group compared with the monotherapies.[1] Similarly, in another study by Hodi et al, (CheckMate 069) it was found that OS rates in all randomized patients were 63·8% (95% CI: 53.3–72.6) for the combination therapy group vs 53·6% (95% CI: 38.1–66.8) for ipilimumab alone at a median follow-up of 24 months. However, grade 3–4 adverse events associated with combination therapy were observed in 51 [54%] of 94 patients vs 9 [20%] of 46 patients linked to ipilimumab alone. The results of this study indicated that the combination of nivolumab plus ipilimumab may result in a higher OS rate vs ipilimumab in patients with advanced melanoma.[19] In line with previous studies, in a multicenter retrospective cohort study Zimmer et al, found that OS rates for the monotherapy and the combination groups were 16% and 21%, respectively. The disease control rate was 42% for the monotherapy and 33% for the combination groups. One-year OS rates for the monotherapy and the combination groups were 54% and 55%, respectively. The authors, however, stated that the combination therapy with nivolumab and ipilimumab was significantly less effective in patients with advanced melanoma with previous anti-PD failure compared with treatment-naïve melanoma patients.[18] This was, in contrast, to da Silva et al findings, which showed that in advanced melanoma patients who were resistant to anti-PD-1 monotherapy, combination therapy had higher efficacy than monotherapy with a higher ORR (60 [31%] of 193 patients vs 21 [13%] of 162 patients; P < .0001), longer PFS (median 3.0 months [95% CI: 2.6–3.6] vs 2.6 months [2.4–2.9]; HR, 0·69, 95% CI: 0·55–0·87; P = .0019), and longer OS (median overall survival 20·4 months [95% CI: 12·7–34·8] vs 8·8 months [6·1–11·3]; HR, 0·50, 95% CI: 0.38–0.66; P < .0001), with a similar rate of grade 3–5 toxicity. The results of this study suggested combination therapy with ipilimumab and nivolumab as a superior option over ipilimumab monotherapy in advanced melanoma patients.[16] Long et al found that combination therapy with nivolumab plus ipilimumab results in higher an intracranial response than ipilimumab monotherapy (16 (46%; 95% CI: 29–63) of 35 patients, 5 (20%; 7–41) of 25, and 1 (6%; 0–30) of 16, respectively) in patients with metastatic melanoma to the brain.[17]\nA meta-analysis by Menshawy et al[20] performed on 1910 patients (nivolumab group, n = 1207 and control group, n = 703) showed that combination therapy with nivolumab plus ipilimumab had higher ORR [RR: 3.58, 95% CI: 2.08–6.14], complete response rate (RR: 5.93, 95% CI: 2.45–14.37), partial response rate (RR: 2.80, 95% CI: 2.16–3.64), stable disease rate (RR: 0.56, 95% CI: 0.41–0.76), and PFS (hazard ratio: 0.67, 95% CI: 0.60–0.74) compared with ipilimumab monotherapy. However, this meta-analysis is rather old and did not include the 5-year update from the CheckMate 067 study by Larkin et al,[1] and also recent studies by da Silva et al[16] and Long et al.[17] On the other hand, this study included a study by Postow et al,[21] which is an earlier version of the CheckMate 069 study by Hodi et al[19] and should not be included in the meta-analysis as a separate entity. Another meta-analysis by Hao et al performed on two studies showed that combination therapy with nivolumab plus ipilimumab had a visible significant advantage over the ipilimumab monotherapy.[12] But the number of studies was too low to decide on anything.", "One of the main concerns with immune checkpoint inhibitors is the risk of immune-related adverse events (IrAEs). Nivolumab was shown to be well tolerated by patients with advanced melanoma. On the one hand, it is speculated that combination therapy with immunotherapies elevates the incidence of potential IrAEs. In a systematic review and meta-analysis by Almutairi et al., it was found that the incidences of potential IrAEs in combination therapies vs. monotherapies were higher for most types of IrAEs. This caused hyperglycemia, thyroid, hepatic, and musculoskeletal disorders in the patients receiving these medications.[22] On the other hand, it was argued that the emergence of both early and late IrAEs could be regarded as a favorable prognostic parameter in patients receiving immunotherapy and immunoradiotherapy for solid tumors especially if these events are delayed; as they are accompanied by increased overall response rate and improved OS and PFS in these patients.[23,24] In any case, the oncologist should take into consideration the occurrence of IrAEs in patients with advanced melanoma especially those with a history of autoimmune disease, poor kidney function of grade 3 or greater, and use of CTLA-4 inhibitors.[25]", "Our study had several shortcomings that should be taken into consideration when interpreting the results. We based our analysis on unadjusted data. This means that confounders such as age, BRAF mutation status, prior systemic therapy, PD-L1 status, and gender were not considered in this meta-analysis. This might be the result of the low number of studies existing in the field and included in this meta-analysis. An increase in the number of studies published in this field and also adjustments for confounders mentioned above might result in more accurate outcomes. Besides, this study was limited to English full-text original papers. This results in missing data emerging from conferences and also other languages. Further, the existence of an open-labeled clinical trial, and also pharmaceutical companies-funded studies increased the ROB in our study.", "Data emerging from this study showed that combination therapy with ipilimumab plus nivolumab meaningfully increased the overall response rate, including the CR, compared with monotherapy with ipilimumab or nivolumab in patients with advanced melanoma regardless of the patients untreated or after anti-CTLA-4 treatment. This was accompanied by higher incidences of potential IrAEs in combination therapy. In this light, combination therapy with ipilimumab plus nivolumab could be a promising strategy in the treatment of patients with advanced melanoma.", "RZH, XLZ, JML, YJZ, XCZ, and FC performed the statistical analysis and wrote the manuscript. RZH, XLZ, JML, and YJZ conceived the study design and participated in the manuscript writing. XCZ and FC revised and edited the manuscript. All authors read the final manuscript." ]
[ "intro", "methods", null, null, null, null, null, "results", null, null, null, null, null, null, null, null, null, null, "conclusions", null ]
[ "immunotherapy", "ipilimumab", "melanoma", "meta-analysis", "nivolumab", "systematic review" ]
The efficacy of Yiqi Huoxue method in treating coronary artery disease after percutaneous coronary intervention: A meta-analysis in accordance with PRISMA guideline.
36254054
Percutaneous coronary intervention (PCI), the most common method in treating coronary artery disease (CAD), has a variety of side effects. Yiqi Huoxue therapy (YQHX) can effectively alleviate the symptoms of patients and reduce the side effects. However, a reliable and systematic assessment of the methodologies is not available.
BACKGROUND
Seven electronic databases were searched to identify randomized controlled trials of YQHX method for CAD after PCI. The quality assessment of the trials included was performed by employing the Cochrane Risk of Bias tool.
METHODS
One thousand eight hundred sixty-eight patients from 23 randomized controlled trials were included in this review. The aggregated results showed that the experimental group got better effect in increasing ORR, TCMSRR, ECG, HDL-C, and in lowering the level of CRP, TC, and MACE in comparison with the control group.
RESULTS
YQHX method is a valid complementary and alternative therapy in the management of CAD after PCI, and is an effective and safe therapy for CAD.
CONCLUSION
[ "Humans", "Coronary Artery Disease", "Percutaneous Coronary Intervention", "Research Design", "Randomized Controlled Trials as Topic" ]
9575818
1. Introduction
Coronary artery disease (CAD) is one of the most serious cardiovascular diseases threatening human health. Yiqi Huoxue therapy (YQHX) means invigorating Qi and activating blood circulation, and has already been widely used to treat cardiovascular diseases.[1–3] Presently, percutaneous coronary intervention (PCI) is the most common method in treating CAD and can reduce the mortality of CAD significantly.[3] However, PCI cannot root out the underlying causes or pathological basis of coronary stenosis and may damage blood vessels and endothelial cells, thus inducing platelet aggregation and thrombosis.[4] Furthermore, the patients may also be complicated with coronary embolism and thrombosis, myocardial ischemia-reperfusion injury, coronary microcirculation disorder, no reflux, in-stent restenosis and other pathological conditions.[5] Routine dual antiplatelet therapy after PCI also has toxic and side effects.[6] In addition, PCI is an exogenous injury and is normally costly. Therefore, alternative and complementary therapies in CAD after PCI treatment are becoming more and more imminent. According to the basic theories of Traditional Chinese Medicine (TCM), CAD after PCI is equivalent to the term of “Xiong Bi.” The etiology and pathogenesis of CAD after PCI are related to Qi deficiency and blood stasis referring to the 5 elements in TCM. When Qi deficiency occurs in the organism, blood circulation will become, to a different degree, retarded, which may lead to a blockage of the heart vessels.[7] Therefore, invigorating Qi and activating circulation to remove blood stasis, the term for which in Chinese Pinyin is “Yiqi Huoxue,” are important therapies for CAD after PCI.[8] Clinical studies have proven that YQHX can effectively alleviate the symptoms of the patients and reduce the side effects of drugs.[5] However, either the evidence of the effect of YQHX on CAD after PCI is so far insufficient or the current information available is not systematic. Therefore, a meta-analysis of clinical randomized controlled trials (RCTs) was conducted to evaluate the efficacy and safety of YQHX on patients with CAD after PCI.
2.5. Statistical method
Meta-analyses of RCTs were performed by using RevMan 5.3. Data were summarized by using risk ratio (RR) with 95% confidence intervals (CI) for discontinuous outcomes, or standard mean difference (SMD) with 95% CI for continuous outcomes. The data were assessed by both fixed effect model and random effect model. Publication bias was assessed by funnel plot analysis if the group included more than 10 trials.[12]
3. Results
[SUBTITLE] 3.1. Study selection [SUBSECTION] The search of 7 databases (English or Chinese) identified 283 records for further evaluation (Fig. 1). 23 RCTs of them were eligible.[4–26] All studies involved patient consent, and the informed consent was given. Flow diagram of study selection and identification. The search of 7 databases (English or Chinese) identified 283 records for further evaluation (Fig. 1). 23 RCTs of them were eligible.[4–26] All studies involved patient consent, and the informed consent was given. Flow diagram of study selection and identification. [SUBTITLE] 3.2. Study characteristics [SUBSECTION] All of the 23 trials included were conducted in China and published in Chinese. All studies were performed in China, with a total of 1868 patients involved (936 the in control group, and 932 in the experimental group). In addition, all the studies exhibited comparable baseline patient characteristics, including age and gender (1053 male, and 815 female). The characteristics of the selected studies are shown in Table 2. Characteristics of included studies. CRP = C-reactive protein, CWM = conventional western medicine, ECG = electrocardiogram, HDL-C = high-density lipoprotein cholesterol, LDL-C = low density lipoprotein-cholesterol, MACE = major adverse cardiovascular events, NA = not applicable, ORR = overall response rate, TC = total cholesterol, TCMSRR = TCM syndrome response rate, TG = triglycerides, YQHX = Yiqi Huoxue. All of the 23 trials included were conducted in China and published in Chinese. All studies were performed in China, with a total of 1868 patients involved (936 the in control group, and 932 in the experimental group). In addition, all the studies exhibited comparable baseline patient characteristics, including age and gender (1053 male, and 815 female). The characteristics of the selected studies are shown in Table 2. Characteristics of included studies. CRP = C-reactive protein, CWM = conventional western medicine, ECG = electrocardiogram, HDL-C = high-density lipoprotein cholesterol, LDL-C = low density lipoprotein-cholesterol, MACE = major adverse cardiovascular events, NA = not applicable, ORR = overall response rate, TC = total cholesterol, TCMSRR = TCM syndrome response rate, TG = triglycerides, YQHX = Yiqi Huoxue. [SUBTITLE] 3.3. Study quality [SUBSECTION] Among the trials, 14 studies[5–17,25] stated the method of the sequence generation with random number table and drawing, while none of the 22 studies reported details for sample size calculations and none was double-blind or placebo controlled study. Additionally, none mentioned allocation concealment or blinding methods. Fourteen trials included[5–9,12–15,17–20,22] were assessed as low risk of bias in incomplete outcome data. Fourteen of the trials included[5,6,8,10,11,13,14,16–22] were assessed as low risk of reporting bias, and the other 4,[7,9,12,15] as unclear risk of reporting bias. The details of the risk of bias of each trial are presented in Figures 2 and 3. Risk of bias: reviewing authors’ judgments about each risk of bias item for each included study. Summary of the risk of bias assessment for included trials. Among the trials, 14 studies[5–17,25] stated the method of the sequence generation with random number table and drawing, while none of the 22 studies reported details for sample size calculations and none was double-blind or placebo controlled study. Additionally, none mentioned allocation concealment or blinding methods. Fourteen trials included[5–9,12–15,17–20,22] were assessed as low risk of bias in incomplete outcome data. Fourteen of the trials included[5,6,8,10,11,13,14,16–22] were assessed as low risk of reporting bias, and the other 4,[7,9,12,15] as unclear risk of reporting bias. The details of the risk of bias of each trial are presented in Figures 2 and 3. Risk of bias: reviewing authors’ judgments about each risk of bias item for each included study. Summary of the risk of bias assessment for included trials. [SUBTITLE] 3.4. Effects of the interventions [SUBSECTION] The outcomes, ORR (13 trials), TCMSRR (8 trials), ECG (9 trials), MACE (9 trials), blood lipid (TC, TG) level (7 trials), (HDL-C, LDL-C) level (6 trials), and CRP (6 trials) were analyzed. [SUBTITLE] 3.4.1. Overall response rate [SUBSECTION] Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4). Forest plot of overall response rate. Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4). Forest plot of overall response rate. [SUBTITLE] 3.4.2. ECG improvement [SUBSECTION] Nine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5). Forest plot of improvement of ECG. ECG = electrocardiogram. Nine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5). Forest plot of improvement of ECG. ECG = electrocardiogram. [SUBTITLE] 3.4.3. TCM syndrome response rate [SUBSECTION] Eight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6). Forest plot of TCM syndrome response rate. TCM = traditional Chinese medicine. Eight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6). Forest plot of TCM syndrome response rate. TCM = traditional Chinese medicine. [SUBTITLE] 3.4.4. Major adverse cardiovascular events [SUBSECTION] Nine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7). Forest plot of improvement of MACE. MACE = major adverse cardiovascular events. Nine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7). Forest plot of improvement of MACE. MACE = major adverse cardiovascular events. [SUBTITLE] 3.4.5. GRADE assessment [SUBSECTION] However, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3). Summary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI. There were serious limitations of methodological quality of included trials according to the risk of bias assessment. There were serious publication bias between the studies. CAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine. However, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3). Summary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI. There were serious limitations of methodological quality of included trials according to the risk of bias assessment. There were serious publication bias between the studies. CAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine. [SUBTITLE] 3.4.6. CRP [SUBSECTION] Six RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8). Forest plot of improvement of CRP. CRP = C-reactive protein. Six RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8). Forest plot of improvement of CRP. CRP = C-reactive protein. [SUBTITLE] 3.4.7. Blood lipid [SUBSECTION] (1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9). (2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10). (3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11). (4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12). Forest plot of improvement of TC. TC = total cholesterol. Forest plot of improvement of TG. TG = triglycerides. Forest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol. Forest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol. (1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9). (2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10). (3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11). (4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12). Forest plot of improvement of TC. TC = total cholesterol. Forest plot of improvement of TG. TG = triglycerides. Forest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol. Forest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol. The outcomes, ORR (13 trials), TCMSRR (8 trials), ECG (9 trials), MACE (9 trials), blood lipid (TC, TG) level (7 trials), (HDL-C, LDL-C) level (6 trials), and CRP (6 trials) were analyzed. [SUBTITLE] 3.4.1. Overall response rate [SUBSECTION] Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4). Forest plot of overall response rate. Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4). Forest plot of overall response rate. [SUBTITLE] 3.4.2. ECG improvement [SUBSECTION] Nine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5). Forest plot of improvement of ECG. ECG = electrocardiogram. Nine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5). Forest plot of improvement of ECG. ECG = electrocardiogram. [SUBTITLE] 3.4.3. TCM syndrome response rate [SUBSECTION] Eight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6). Forest plot of TCM syndrome response rate. TCM = traditional Chinese medicine. Eight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6). Forest plot of TCM syndrome response rate. TCM = traditional Chinese medicine. [SUBTITLE] 3.4.4. Major adverse cardiovascular events [SUBSECTION] Nine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7). Forest plot of improvement of MACE. MACE = major adverse cardiovascular events. Nine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7). Forest plot of improvement of MACE. MACE = major adverse cardiovascular events. [SUBTITLE] 3.4.5. GRADE assessment [SUBSECTION] However, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3). Summary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI. There were serious limitations of methodological quality of included trials according to the risk of bias assessment. There were serious publication bias between the studies. CAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine. However, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3). Summary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI. There were serious limitations of methodological quality of included trials according to the risk of bias assessment. There were serious publication bias between the studies. CAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine. [SUBTITLE] 3.4.6. CRP [SUBSECTION] Six RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8). Forest plot of improvement of CRP. CRP = C-reactive protein. Six RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8). Forest plot of improvement of CRP. CRP = C-reactive protein. [SUBTITLE] 3.4.7. Blood lipid [SUBSECTION] (1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9). (2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10). (3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11). (4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12). Forest plot of improvement of TC. TC = total cholesterol. Forest plot of improvement of TG. TG = triglycerides. Forest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol. Forest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol. (1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9). (2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10). (3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11). (4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12). Forest plot of improvement of TC. TC = total cholesterol. Forest plot of improvement of TG. TG = triglycerides. Forest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol. Forest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol. [SUBTITLE] 3.5. Adverse reactions [SUBSECTION] Five cases in the treatment group and 9 cases in the control group had adverse reactions, with an incidence of 4.1%, 7.5%, respectively. The incidence of adverse reactions in the treatment of PCI patients with YQHX was relatively lower. Five cases in the treatment group and 9 cases in the control group had adverse reactions, with an incidence of 4.1%, 7.5%, respectively. The incidence of adverse reactions in the treatment of PCI patients with YQHX was relatively lower. [SUBTITLE] 3.6. Publication bias [SUBSECTION] In order to detect possible publication bias, the 13 trials of ORR were analyzed with a fixed effects model. The funnel plot of ORR was asymmetrical, indicating the presence of publication bias (Fig. 13). Funnel plot of the overall response rate. In order to detect possible publication bias, the 13 trials of ORR were analyzed with a fixed effects model. The funnel plot of ORR was asymmetrical, indicating the presence of publication bias (Fig. 13). Funnel plot of the overall response rate.
null
null
[ "2. Methods", "2.1. Search strategy", "2.2. Study selection", "2.3. Data abstraction", "2.4. Quality assessment", "3.1. Study selection", "3.2. Study characteristics", "3.3. Study quality", "3.4. Effects of the interventions", "3.4.1. Overall response rate", "3.4.2. ECG improvement", "3.4.3. TCM syndrome response rate", "3.4.4. Major adverse cardiovascular events", "3.4.5. GRADE assessment", "3.4.6. CRP", "3.4.7. Blood lipid", "3.5. Adverse reactions", "3.6. Publication bias", "Acknowledgments", "Author contributions" ]
[ "[SUBTITLE] 2.1. Search strategy [SUBSECTION] The Cochrane Library, PubMed, EMBASE, the China National Knowledge Infrastructure database, the Chinese Biomedical Literature database, the VIP database and the Wanfang database were searched. The search terms used were (Chinese medicine OR herbs OR herbal formula OR Yiqi OR Huoxue OR Supplementing Qi OR activating blood circulation) AND (CAD) AND (PCI OR intervention therapy) AND (RCT). The last search was finished on December 31, 2021. No limit was placed on the language.\nThe Cochrane Library, PubMed, EMBASE, the China National Knowledge Infrastructure database, the Chinese Biomedical Literature database, the VIP database and the Wanfang database were searched. The search terms used were (Chinese medicine OR herbs OR herbal formula OR Yiqi OR Huoxue OR Supplementing Qi OR activating blood circulation) AND (CAD) AND (PCI OR intervention therapy) AND (RCT). The last search was finished on December 31, 2021. No limit was placed on the language.\n[SUBTITLE] 2.2. Study selection [SUBSECTION] Studies were selected according to the Cochrane Handbook for Systematic Reviews of Interventions.[9]\nInclusion Criteria. The studies were performed as RCTs; patients were diagnosed with CAD and received PCI; dual antiplatelet therapy plus other western medicine was permitted to be taken according to individual symptoms; YQHX formula composed of classic herbs (Astragalus or Salvia miltiorrhiza or Codonopsis pilosula or Ginseng with clear dose) with conventional western medicine (CWM) was used for the experimental group and CWM alone for the control group. The primary outcomes included overall response rate (ORR) and TCM syndrome response rate (TCMSRR) by referring to the evaluation criteria of Guidelines for clinical research on Chinese new herbal medicines (Table 1), electrocardiogram (ECG) improvement (ST segment rose more than 0.05 MV after treatment, in the main lead, the change of T wave became more than 25% shallow or T wave changed from flat to upright), and major adverse cardiovascular events (MACE); secondary outcomes included levels of C-reactive protein (CRP) and blood lipid index (total cholesterol [TC], TG, high-density lipoprotein cholesterol [HDL-C], LDL-C).\nEvaluation criteria on the efficacy of clinical symptoms and TCM syndromes recommended by GCRNDTCM\nGCRNDTCM = guidelines of clinical research of new drugs of traditional Chinese medicine, TCM = traditional Chinese medicine.\nExclusion Criteria. The target population was incongruent with diagnostic criteria of CAD and received PCI; the main intervention was mixed with too many measures; the studies were allocated neither with appropriate control nor with randomization; the studies had data missed or duplicate publication.\nStudies were selected according to the Cochrane Handbook for Systematic Reviews of Interventions.[9]\nInclusion Criteria. The studies were performed as RCTs; patients were diagnosed with CAD and received PCI; dual antiplatelet therapy plus other western medicine was permitted to be taken according to individual symptoms; YQHX formula composed of classic herbs (Astragalus or Salvia miltiorrhiza or Codonopsis pilosula or Ginseng with clear dose) with conventional western medicine (CWM) was used for the experimental group and CWM alone for the control group. The primary outcomes included overall response rate (ORR) and TCM syndrome response rate (TCMSRR) by referring to the evaluation criteria of Guidelines for clinical research on Chinese new herbal medicines (Table 1), electrocardiogram (ECG) improvement (ST segment rose more than 0.05 MV after treatment, in the main lead, the change of T wave became more than 25% shallow or T wave changed from flat to upright), and major adverse cardiovascular events (MACE); secondary outcomes included levels of C-reactive protein (CRP) and blood lipid index (total cholesterol [TC], TG, high-density lipoprotein cholesterol [HDL-C], LDL-C).\nEvaluation criteria on the efficacy of clinical symptoms and TCM syndromes recommended by GCRNDTCM\nGCRNDTCM = guidelines of clinical research of new drugs of traditional Chinese medicine, TCM = traditional Chinese medicine.\nExclusion Criteria. The target population was incongruent with diagnostic criteria of CAD and received PCI; the main intervention was mixed with too many measures; the studies were allocated neither with appropriate control nor with randomization; the studies had data missed or duplicate publication.\n[SUBTITLE] 2.3. Data abstraction [SUBSECTION] Two authors (Miao Z and Mingyue S) independently screened the titles and abstracts of the achieved citations from primary searching. Full text of the articles of potential interest was downloaded for further evaluation. Those meeting inclusion criteria were included in the final review. The discrepancies in the process of selection were resolved by the third author (Huijun Y).\nTwo authors (Miao Z and Mingyue S) independently screened the titles and abstracts of the achieved citations from primary searching. Full text of the articles of potential interest was downloaded for further evaluation. Those meeting inclusion criteria were included in the final review. The discrepancies in the process of selection were resolved by the third author (Huijun Y).\n[SUBTITLE] 2.4. Quality assessment [SUBSECTION] The methodological quality of trials was assessed independently by 2 authors (Miao Z and Mingyue S) referring to criteria from the Cochrane Handbook for Systematic Review of Interventions. The items included random sequence generation, allocation concealment, blinding, incomplete outcome data, selective outcome reporting, and other bias (defined as baseline data comparability).\nThe methodological quality of trials was assessed independently by 2 authors (Miao Z and Mingyue S) referring to criteria from the Cochrane Handbook for Systematic Review of Interventions. The items included random sequence generation, allocation concealment, blinding, incomplete outcome data, selective outcome reporting, and other bias (defined as baseline data comparability).\n[SUBTITLE] 2.5. Statistical method [SUBSECTION] Meta-analyses of RCTs were performed by using RevMan 5.3. Data were summarized by using risk ratio (RR) with 95% confidence intervals (CI) for discontinuous outcomes, or standard mean difference (SMD) with 95% CI for continuous outcomes. The data were assessed by both fixed effect model and random effect model. Publication bias was assessed by funnel plot analysis if the group included more than 10 trials.[12]\nMeta-analyses of RCTs were performed by using RevMan 5.3. Data were summarized by using risk ratio (RR) with 95% confidence intervals (CI) for discontinuous outcomes, or standard mean difference (SMD) with 95% CI for continuous outcomes. The data were assessed by both fixed effect model and random effect model. Publication bias was assessed by funnel plot analysis if the group included more than 10 trials.[12]", "The Cochrane Library, PubMed, EMBASE, the China National Knowledge Infrastructure database, the Chinese Biomedical Literature database, the VIP database and the Wanfang database were searched. The search terms used were (Chinese medicine OR herbs OR herbal formula OR Yiqi OR Huoxue OR Supplementing Qi OR activating blood circulation) AND (CAD) AND (PCI OR intervention therapy) AND (RCT). The last search was finished on December 31, 2021. No limit was placed on the language.", "Studies were selected according to the Cochrane Handbook for Systematic Reviews of Interventions.[9]\nInclusion Criteria. The studies were performed as RCTs; patients were diagnosed with CAD and received PCI; dual antiplatelet therapy plus other western medicine was permitted to be taken according to individual symptoms; YQHX formula composed of classic herbs (Astragalus or Salvia miltiorrhiza or Codonopsis pilosula or Ginseng with clear dose) with conventional western medicine (CWM) was used for the experimental group and CWM alone for the control group. The primary outcomes included overall response rate (ORR) and TCM syndrome response rate (TCMSRR) by referring to the evaluation criteria of Guidelines for clinical research on Chinese new herbal medicines (Table 1), electrocardiogram (ECG) improvement (ST segment rose more than 0.05 MV after treatment, in the main lead, the change of T wave became more than 25% shallow or T wave changed from flat to upright), and major adverse cardiovascular events (MACE); secondary outcomes included levels of C-reactive protein (CRP) and blood lipid index (total cholesterol [TC], TG, high-density lipoprotein cholesterol [HDL-C], LDL-C).\nEvaluation criteria on the efficacy of clinical symptoms and TCM syndromes recommended by GCRNDTCM\nGCRNDTCM = guidelines of clinical research of new drugs of traditional Chinese medicine, TCM = traditional Chinese medicine.\nExclusion Criteria. The target population was incongruent with diagnostic criteria of CAD and received PCI; the main intervention was mixed with too many measures; the studies were allocated neither with appropriate control nor with randomization; the studies had data missed or duplicate publication.", "Two authors (Miao Z and Mingyue S) independently screened the titles and abstracts of the achieved citations from primary searching. Full text of the articles of potential interest was downloaded for further evaluation. Those meeting inclusion criteria were included in the final review. The discrepancies in the process of selection were resolved by the third author (Huijun Y).", "The methodological quality of trials was assessed independently by 2 authors (Miao Z and Mingyue S) referring to criteria from the Cochrane Handbook for Systematic Review of Interventions. The items included random sequence generation, allocation concealment, blinding, incomplete outcome data, selective outcome reporting, and other bias (defined as baseline data comparability).", "The search of 7 databases (English or Chinese) identified 283 records for further evaluation (Fig. 1). 23 RCTs of them were eligible.[4–26] All studies involved patient consent, and the informed consent was given.\nFlow diagram of study selection and identification.", "All of the 23 trials included were conducted in China and published in Chinese. All studies were performed in China, with a total of 1868 patients involved (936 the in control group, and 932 in the experimental group). In addition, all the studies exhibited comparable baseline patient characteristics, including age and gender (1053 male, and 815 female). The characteristics of the selected studies are shown in Table 2.\nCharacteristics of included studies.\nCRP = C-reactive protein, CWM = conventional western medicine, ECG = electrocardiogram, HDL-C = high-density lipoprotein cholesterol, LDL-C = low density lipoprotein-cholesterol, MACE = major adverse cardiovascular events, NA = not applicable, ORR = overall response rate, TC = total cholesterol, TCMSRR = TCM syndrome response rate, TG = triglycerides, YQHX = Yiqi Huoxue.", "Among the trials, 14 studies[5–17,25] stated the method of the sequence generation with random number table and drawing, while none of the 22 studies reported details for sample size calculations and none was double-blind or placebo controlled study. Additionally, none mentioned allocation concealment or blinding methods. Fourteen trials included[5–9,12–15,17–20,22] were assessed as low risk of bias in incomplete outcome data. Fourteen of the trials included[5,6,8,10,11,13,14,16–22] were assessed as low risk of reporting bias, and the other 4,[7,9,12,15] as unclear risk of reporting bias. The details of the risk of bias of each trial are presented in Figures 2 and 3.\nRisk of bias: reviewing authors’ judgments about each risk of bias item for each included study.\nSummary of the risk of bias assessment for included trials.", "The outcomes, ORR (13 trials), TCMSRR (8 trials), ECG (9 trials), MACE (9 trials), blood lipid (TC, TG) level (7 trials), (HDL-C, LDL-C) level (6 trials), and CRP (6 trials) were analyzed.\n[SUBTITLE] 3.4.1. Overall response rate [SUBSECTION] Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4).\nForest plot of overall response rate.\n Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4).\nForest plot of overall response rate.\n[SUBTITLE] 3.4.2. ECG improvement [SUBSECTION] Nine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5).\nForest plot of improvement of ECG. ECG = electrocardiogram.\nNine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5).\nForest plot of improvement of ECG. ECG = electrocardiogram.\n[SUBTITLE] 3.4.3. TCM syndrome response rate [SUBSECTION] Eight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6).\nForest plot of TCM syndrome response rate. TCM = traditional Chinese medicine.\nEight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6).\nForest plot of TCM syndrome response rate. TCM = traditional Chinese medicine.\n[SUBTITLE] 3.4.4. Major adverse cardiovascular events [SUBSECTION] Nine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7).\nForest plot of improvement of MACE. MACE = major adverse cardiovascular events.\nNine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7).\nForest plot of improvement of MACE. MACE = major adverse cardiovascular events.\n[SUBTITLE] 3.4.5. GRADE assessment [SUBSECTION] However, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3).\nSummary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI.\n There were serious limitations of methodological quality of included trials according to the risk of bias assessment.\n There were serious publication bias between the studies.\nCAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine.\nHowever, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3).\nSummary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI.\n There were serious limitations of methodological quality of included trials according to the risk of bias assessment.\n There were serious publication bias between the studies.\nCAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine.\n[SUBTITLE] 3.4.6. CRP [SUBSECTION] Six RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8).\nForest plot of improvement of CRP. CRP = C-reactive protein.\nSix RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8).\nForest plot of improvement of CRP. CRP = C-reactive protein.\n[SUBTITLE] 3.4.7. Blood lipid [SUBSECTION] (1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9).\n(2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10).\n(3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11).\n(4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12).\nForest plot of improvement of TC. TC = total cholesterol.\nForest plot of improvement of TG. TG = triglycerides.\nForest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol.\nForest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol.\n(1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9).\n(2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10).\n(3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11).\n(4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12).\nForest plot of improvement of TC. TC = total cholesterol.\nForest plot of improvement of TG. TG = triglycerides.\nForest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol.\nForest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol.", " Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4).\nForest plot of overall response rate.", "Nine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5).\nForest plot of improvement of ECG. ECG = electrocardiogram.", "Eight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6).\nForest plot of TCM syndrome response rate. TCM = traditional Chinese medicine.", "Nine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7).\nForest plot of improvement of MACE. MACE = major adverse cardiovascular events.", "However, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3).\nSummary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI.\n There were serious limitations of methodological quality of included trials according to the risk of bias assessment.\n There were serious publication bias between the studies.\nCAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine.", "Six RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8).\nForest plot of improvement of CRP. CRP = C-reactive protein.", "(1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9).\n(2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10).\n(3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11).\n(4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12).\nForest plot of improvement of TC. TC = total cholesterol.\nForest plot of improvement of TG. TG = triglycerides.\nForest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol.\nForest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol.", "Five cases in the treatment group and 9 cases in the control group had adverse reactions, with an incidence of 4.1%, 7.5%, respectively. The incidence of adverse reactions in the treatment of PCI patients with YQHX was relatively lower.", "In order to detect possible publication bias, the 13 trials of ORR were analyzed with a fixed effects model. The funnel plot of ORR was asymmetrical, indicating the presence of publication bias (Fig. 13).\nFunnel plot of the overall response rate.", "Thank all the authors for their contributions to this article.", "Data curation: Miao Zhang, Qiting Chen.\nFormal analysis: Ming-Yue Sun, Qiting Chen.\nFunding acquisition: Miao Zhang, Feng-Qin Xu.\nInvestigation: Hui-Jun Yin.\nMethodology: Qiting Chen.\nProject administration: Wenbo Wei.\nSoftware: Miao Zhang, Qiting Chen.\nSupervision: Zongzheng Chen, Hui-Jun Yin.\nValidation: Zongzheng Chen.\nVisualization: Ruiting Wang.\nWriting – original draft: Miao Zhang, Ming-Yue Sun.\nWriting – review & editing: Miao Zhang, Wenbo Wei, Feng-Qin Xu, Hui-Jun Yin." ]
[ "methods", null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Methods", "2.1. Search strategy", "2.2. Study selection", "2.3. Data abstraction", "2.4. Quality assessment", "2.5. Statistical method", "3. Results", "3.1. Study selection", "3.2. Study characteristics", "3.3. Study quality", "3.4. Effects of the interventions", "3.4.1. Overall response rate", "3.4.2. ECG improvement", "3.4.3. TCM syndrome response rate", "3.4.4. Major adverse cardiovascular events", "3.4.5. GRADE assessment", "3.4.6. CRP", "3.4.7. Blood lipid", "3.5. Adverse reactions", "3.6. Publication bias", "4. Discussion", "Acknowledgments", "Author contributions" ]
[ "Coronary artery disease (CAD) is one of the most serious cardiovascular diseases threatening human health. Yiqi Huoxue therapy (YQHX) means invigorating Qi and activating blood circulation, and has already been widely used to treat cardiovascular diseases.[1–3] Presently, percutaneous coronary intervention (PCI) is the most common method in treating CAD and can reduce the mortality of CAD significantly.[3] However, PCI cannot root out the underlying causes or pathological basis of coronary stenosis and may damage blood vessels and endothelial cells, thus inducing platelet aggregation and thrombosis.[4] Furthermore, the patients may also be complicated with coronary embolism and thrombosis, myocardial ischemia-reperfusion injury, coronary microcirculation disorder, no reflux, in-stent restenosis and other pathological conditions.[5] Routine dual antiplatelet therapy after PCI also has toxic and side effects.[6] In addition, PCI is an exogenous injury and is normally costly. Therefore, alternative and complementary therapies in CAD after PCI treatment are becoming more and more imminent.\nAccording to the basic theories of Traditional Chinese Medicine (TCM), CAD after PCI is equivalent to the term of “Xiong Bi.” The etiology and pathogenesis of CAD after PCI are related to Qi deficiency and blood stasis referring to the 5 elements in TCM. When Qi deficiency occurs in the organism, blood circulation will become, to a different degree, retarded, which may lead to a blockage of the heart vessels.[7] Therefore, invigorating Qi and activating circulation to remove blood stasis, the term for which in Chinese Pinyin is “Yiqi Huoxue,” are important therapies for CAD after PCI.[8] Clinical studies have proven that YQHX can effectively alleviate the symptoms of the patients and reduce the side effects of drugs.[5] However, either the evidence of the effect of YQHX on CAD after PCI is so far insufficient or the current information available is not systematic. Therefore, a meta-analysis of clinical randomized controlled trials (RCTs) was conducted to evaluate the efficacy and safety of YQHX on patients with CAD after PCI.", "[SUBTITLE] 2.1. Search strategy [SUBSECTION] The Cochrane Library, PubMed, EMBASE, the China National Knowledge Infrastructure database, the Chinese Biomedical Literature database, the VIP database and the Wanfang database were searched. The search terms used were (Chinese medicine OR herbs OR herbal formula OR Yiqi OR Huoxue OR Supplementing Qi OR activating blood circulation) AND (CAD) AND (PCI OR intervention therapy) AND (RCT). The last search was finished on December 31, 2021. No limit was placed on the language.\nThe Cochrane Library, PubMed, EMBASE, the China National Knowledge Infrastructure database, the Chinese Biomedical Literature database, the VIP database and the Wanfang database were searched. The search terms used were (Chinese medicine OR herbs OR herbal formula OR Yiqi OR Huoxue OR Supplementing Qi OR activating blood circulation) AND (CAD) AND (PCI OR intervention therapy) AND (RCT). The last search was finished on December 31, 2021. No limit was placed on the language.\n[SUBTITLE] 2.2. Study selection [SUBSECTION] Studies were selected according to the Cochrane Handbook for Systematic Reviews of Interventions.[9]\nInclusion Criteria. The studies were performed as RCTs; patients were diagnosed with CAD and received PCI; dual antiplatelet therapy plus other western medicine was permitted to be taken according to individual symptoms; YQHX formula composed of classic herbs (Astragalus or Salvia miltiorrhiza or Codonopsis pilosula or Ginseng with clear dose) with conventional western medicine (CWM) was used for the experimental group and CWM alone for the control group. The primary outcomes included overall response rate (ORR) and TCM syndrome response rate (TCMSRR) by referring to the evaluation criteria of Guidelines for clinical research on Chinese new herbal medicines (Table 1), electrocardiogram (ECG) improvement (ST segment rose more than 0.05 MV after treatment, in the main lead, the change of T wave became more than 25% shallow or T wave changed from flat to upright), and major adverse cardiovascular events (MACE); secondary outcomes included levels of C-reactive protein (CRP) and blood lipid index (total cholesterol [TC], TG, high-density lipoprotein cholesterol [HDL-C], LDL-C).\nEvaluation criteria on the efficacy of clinical symptoms and TCM syndromes recommended by GCRNDTCM\nGCRNDTCM = guidelines of clinical research of new drugs of traditional Chinese medicine, TCM = traditional Chinese medicine.\nExclusion Criteria. The target population was incongruent with diagnostic criteria of CAD and received PCI; the main intervention was mixed with too many measures; the studies were allocated neither with appropriate control nor with randomization; the studies had data missed or duplicate publication.\nStudies were selected according to the Cochrane Handbook for Systematic Reviews of Interventions.[9]\nInclusion Criteria. The studies were performed as RCTs; patients were diagnosed with CAD and received PCI; dual antiplatelet therapy plus other western medicine was permitted to be taken according to individual symptoms; YQHX formula composed of classic herbs (Astragalus or Salvia miltiorrhiza or Codonopsis pilosula or Ginseng with clear dose) with conventional western medicine (CWM) was used for the experimental group and CWM alone for the control group. The primary outcomes included overall response rate (ORR) and TCM syndrome response rate (TCMSRR) by referring to the evaluation criteria of Guidelines for clinical research on Chinese new herbal medicines (Table 1), electrocardiogram (ECG) improvement (ST segment rose more than 0.05 MV after treatment, in the main lead, the change of T wave became more than 25% shallow or T wave changed from flat to upright), and major adverse cardiovascular events (MACE); secondary outcomes included levels of C-reactive protein (CRP) and blood lipid index (total cholesterol [TC], TG, high-density lipoprotein cholesterol [HDL-C], LDL-C).\nEvaluation criteria on the efficacy of clinical symptoms and TCM syndromes recommended by GCRNDTCM\nGCRNDTCM = guidelines of clinical research of new drugs of traditional Chinese medicine, TCM = traditional Chinese medicine.\nExclusion Criteria. The target population was incongruent with diagnostic criteria of CAD and received PCI; the main intervention was mixed with too many measures; the studies were allocated neither with appropriate control nor with randomization; the studies had data missed or duplicate publication.\n[SUBTITLE] 2.3. Data abstraction [SUBSECTION] Two authors (Miao Z and Mingyue S) independently screened the titles and abstracts of the achieved citations from primary searching. Full text of the articles of potential interest was downloaded for further evaluation. Those meeting inclusion criteria were included in the final review. The discrepancies in the process of selection were resolved by the third author (Huijun Y).\nTwo authors (Miao Z and Mingyue S) independently screened the titles and abstracts of the achieved citations from primary searching. Full text of the articles of potential interest was downloaded for further evaluation. Those meeting inclusion criteria were included in the final review. The discrepancies in the process of selection were resolved by the third author (Huijun Y).\n[SUBTITLE] 2.4. Quality assessment [SUBSECTION] The methodological quality of trials was assessed independently by 2 authors (Miao Z and Mingyue S) referring to criteria from the Cochrane Handbook for Systematic Review of Interventions. The items included random sequence generation, allocation concealment, blinding, incomplete outcome data, selective outcome reporting, and other bias (defined as baseline data comparability).\nThe methodological quality of trials was assessed independently by 2 authors (Miao Z and Mingyue S) referring to criteria from the Cochrane Handbook for Systematic Review of Interventions. The items included random sequence generation, allocation concealment, blinding, incomplete outcome data, selective outcome reporting, and other bias (defined as baseline data comparability).\n[SUBTITLE] 2.5. Statistical method [SUBSECTION] Meta-analyses of RCTs were performed by using RevMan 5.3. Data were summarized by using risk ratio (RR) with 95% confidence intervals (CI) for discontinuous outcomes, or standard mean difference (SMD) with 95% CI for continuous outcomes. The data were assessed by both fixed effect model and random effect model. Publication bias was assessed by funnel plot analysis if the group included more than 10 trials.[12]\nMeta-analyses of RCTs were performed by using RevMan 5.3. Data were summarized by using risk ratio (RR) with 95% confidence intervals (CI) for discontinuous outcomes, or standard mean difference (SMD) with 95% CI for continuous outcomes. The data were assessed by both fixed effect model and random effect model. Publication bias was assessed by funnel plot analysis if the group included more than 10 trials.[12]", "The Cochrane Library, PubMed, EMBASE, the China National Knowledge Infrastructure database, the Chinese Biomedical Literature database, the VIP database and the Wanfang database were searched. The search terms used were (Chinese medicine OR herbs OR herbal formula OR Yiqi OR Huoxue OR Supplementing Qi OR activating blood circulation) AND (CAD) AND (PCI OR intervention therapy) AND (RCT). The last search was finished on December 31, 2021. No limit was placed on the language.", "Studies were selected according to the Cochrane Handbook for Systematic Reviews of Interventions.[9]\nInclusion Criteria. The studies were performed as RCTs; patients were diagnosed with CAD and received PCI; dual antiplatelet therapy plus other western medicine was permitted to be taken according to individual symptoms; YQHX formula composed of classic herbs (Astragalus or Salvia miltiorrhiza or Codonopsis pilosula or Ginseng with clear dose) with conventional western medicine (CWM) was used for the experimental group and CWM alone for the control group. The primary outcomes included overall response rate (ORR) and TCM syndrome response rate (TCMSRR) by referring to the evaluation criteria of Guidelines for clinical research on Chinese new herbal medicines (Table 1), electrocardiogram (ECG) improvement (ST segment rose more than 0.05 MV after treatment, in the main lead, the change of T wave became more than 25% shallow or T wave changed from flat to upright), and major adverse cardiovascular events (MACE); secondary outcomes included levels of C-reactive protein (CRP) and blood lipid index (total cholesterol [TC], TG, high-density lipoprotein cholesterol [HDL-C], LDL-C).\nEvaluation criteria on the efficacy of clinical symptoms and TCM syndromes recommended by GCRNDTCM\nGCRNDTCM = guidelines of clinical research of new drugs of traditional Chinese medicine, TCM = traditional Chinese medicine.\nExclusion Criteria. The target population was incongruent with diagnostic criteria of CAD and received PCI; the main intervention was mixed with too many measures; the studies were allocated neither with appropriate control nor with randomization; the studies had data missed or duplicate publication.", "Two authors (Miao Z and Mingyue S) independently screened the titles and abstracts of the achieved citations from primary searching. Full text of the articles of potential interest was downloaded for further evaluation. Those meeting inclusion criteria were included in the final review. The discrepancies in the process of selection were resolved by the third author (Huijun Y).", "The methodological quality of trials was assessed independently by 2 authors (Miao Z and Mingyue S) referring to criteria from the Cochrane Handbook for Systematic Review of Interventions. The items included random sequence generation, allocation concealment, blinding, incomplete outcome data, selective outcome reporting, and other bias (defined as baseline data comparability).", "Meta-analyses of RCTs were performed by using RevMan 5.3. Data were summarized by using risk ratio (RR) with 95% confidence intervals (CI) for discontinuous outcomes, or standard mean difference (SMD) with 95% CI for continuous outcomes. The data were assessed by both fixed effect model and random effect model. Publication bias was assessed by funnel plot analysis if the group included more than 10 trials.[12]", "[SUBTITLE] 3.1. Study selection [SUBSECTION] The search of 7 databases (English or Chinese) identified 283 records for further evaluation (Fig. 1). 23 RCTs of them were eligible.[4–26] All studies involved patient consent, and the informed consent was given.\nFlow diagram of study selection and identification.\nThe search of 7 databases (English or Chinese) identified 283 records for further evaluation (Fig. 1). 23 RCTs of them were eligible.[4–26] All studies involved patient consent, and the informed consent was given.\nFlow diagram of study selection and identification.\n[SUBTITLE] 3.2. Study characteristics [SUBSECTION] All of the 23 trials included were conducted in China and published in Chinese. All studies were performed in China, with a total of 1868 patients involved (936 the in control group, and 932 in the experimental group). In addition, all the studies exhibited comparable baseline patient characteristics, including age and gender (1053 male, and 815 female). The characteristics of the selected studies are shown in Table 2.\nCharacteristics of included studies.\nCRP = C-reactive protein, CWM = conventional western medicine, ECG = electrocardiogram, HDL-C = high-density lipoprotein cholesterol, LDL-C = low density lipoprotein-cholesterol, MACE = major adverse cardiovascular events, NA = not applicable, ORR = overall response rate, TC = total cholesterol, TCMSRR = TCM syndrome response rate, TG = triglycerides, YQHX = Yiqi Huoxue.\nAll of the 23 trials included were conducted in China and published in Chinese. All studies were performed in China, with a total of 1868 patients involved (936 the in control group, and 932 in the experimental group). In addition, all the studies exhibited comparable baseline patient characteristics, including age and gender (1053 male, and 815 female). The characteristics of the selected studies are shown in Table 2.\nCharacteristics of included studies.\nCRP = C-reactive protein, CWM = conventional western medicine, ECG = electrocardiogram, HDL-C = high-density lipoprotein cholesterol, LDL-C = low density lipoprotein-cholesterol, MACE = major adverse cardiovascular events, NA = not applicable, ORR = overall response rate, TC = total cholesterol, TCMSRR = TCM syndrome response rate, TG = triglycerides, YQHX = Yiqi Huoxue.\n[SUBTITLE] 3.3. Study quality [SUBSECTION] Among the trials, 14 studies[5–17,25] stated the method of the sequence generation with random number table and drawing, while none of the 22 studies reported details for sample size calculations and none was double-blind or placebo controlled study. Additionally, none mentioned allocation concealment or blinding methods. Fourteen trials included[5–9,12–15,17–20,22] were assessed as low risk of bias in incomplete outcome data. Fourteen of the trials included[5,6,8,10,11,13,14,16–22] were assessed as low risk of reporting bias, and the other 4,[7,9,12,15] as unclear risk of reporting bias. The details of the risk of bias of each trial are presented in Figures 2 and 3.\nRisk of bias: reviewing authors’ judgments about each risk of bias item for each included study.\nSummary of the risk of bias assessment for included trials.\nAmong the trials, 14 studies[5–17,25] stated the method of the sequence generation with random number table and drawing, while none of the 22 studies reported details for sample size calculations and none was double-blind or placebo controlled study. Additionally, none mentioned allocation concealment or blinding methods. Fourteen trials included[5–9,12–15,17–20,22] were assessed as low risk of bias in incomplete outcome data. Fourteen of the trials included[5,6,8,10,11,13,14,16–22] were assessed as low risk of reporting bias, and the other 4,[7,9,12,15] as unclear risk of reporting bias. The details of the risk of bias of each trial are presented in Figures 2 and 3.\nRisk of bias: reviewing authors’ judgments about each risk of bias item for each included study.\nSummary of the risk of bias assessment for included trials.\n[SUBTITLE] 3.4. Effects of the interventions [SUBSECTION] The outcomes, ORR (13 trials), TCMSRR (8 trials), ECG (9 trials), MACE (9 trials), blood lipid (TC, TG) level (7 trials), (HDL-C, LDL-C) level (6 trials), and CRP (6 trials) were analyzed.\n[SUBTITLE] 3.4.1. Overall response rate [SUBSECTION] Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4).\nForest plot of overall response rate.\n Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4).\nForest plot of overall response rate.\n[SUBTITLE] 3.4.2. ECG improvement [SUBSECTION] Nine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5).\nForest plot of improvement of ECG. ECG = electrocardiogram.\nNine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5).\nForest plot of improvement of ECG. ECG = electrocardiogram.\n[SUBTITLE] 3.4.3. TCM syndrome response rate [SUBSECTION] Eight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6).\nForest plot of TCM syndrome response rate. TCM = traditional Chinese medicine.\nEight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6).\nForest plot of TCM syndrome response rate. TCM = traditional Chinese medicine.\n[SUBTITLE] 3.4.4. Major adverse cardiovascular events [SUBSECTION] Nine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7).\nForest plot of improvement of MACE. MACE = major adverse cardiovascular events.\nNine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7).\nForest plot of improvement of MACE. MACE = major adverse cardiovascular events.\n[SUBTITLE] 3.4.5. GRADE assessment [SUBSECTION] However, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3).\nSummary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI.\n There were serious limitations of methodological quality of included trials according to the risk of bias assessment.\n There were serious publication bias between the studies.\nCAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine.\nHowever, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3).\nSummary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI.\n There were serious limitations of methodological quality of included trials according to the risk of bias assessment.\n There were serious publication bias between the studies.\nCAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine.\n[SUBTITLE] 3.4.6. CRP [SUBSECTION] Six RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8).\nForest plot of improvement of CRP. CRP = C-reactive protein.\nSix RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8).\nForest plot of improvement of CRP. CRP = C-reactive protein.\n[SUBTITLE] 3.4.7. Blood lipid [SUBSECTION] (1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9).\n(2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10).\n(3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11).\n(4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12).\nForest plot of improvement of TC. TC = total cholesterol.\nForest plot of improvement of TG. TG = triglycerides.\nForest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol.\nForest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol.\n(1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9).\n(2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10).\n(3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11).\n(4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12).\nForest plot of improvement of TC. TC = total cholesterol.\nForest plot of improvement of TG. TG = triglycerides.\nForest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol.\nForest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol.\nThe outcomes, ORR (13 trials), TCMSRR (8 trials), ECG (9 trials), MACE (9 trials), blood lipid (TC, TG) level (7 trials), (HDL-C, LDL-C) level (6 trials), and CRP (6 trials) were analyzed.\n[SUBTITLE] 3.4.1. Overall response rate [SUBSECTION] Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4).\nForest plot of overall response rate.\n Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4).\nForest plot of overall response rate.\n[SUBTITLE] 3.4.2. ECG improvement [SUBSECTION] Nine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5).\nForest plot of improvement of ECG. ECG = electrocardiogram.\nNine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5).\nForest plot of improvement of ECG. ECG = electrocardiogram.\n[SUBTITLE] 3.4.3. TCM syndrome response rate [SUBSECTION] Eight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6).\nForest plot of TCM syndrome response rate. TCM = traditional Chinese medicine.\nEight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6).\nForest plot of TCM syndrome response rate. TCM = traditional Chinese medicine.\n[SUBTITLE] 3.4.4. Major adverse cardiovascular events [SUBSECTION] Nine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7).\nForest plot of improvement of MACE. MACE = major adverse cardiovascular events.\nNine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7).\nForest plot of improvement of MACE. MACE = major adverse cardiovascular events.\n[SUBTITLE] 3.4.5. GRADE assessment [SUBSECTION] However, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3).\nSummary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI.\n There were serious limitations of methodological quality of included trials according to the risk of bias assessment.\n There were serious publication bias between the studies.\nCAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine.\nHowever, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3).\nSummary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI.\n There were serious limitations of methodological quality of included trials according to the risk of bias assessment.\n There were serious publication bias between the studies.\nCAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine.\n[SUBTITLE] 3.4.6. CRP [SUBSECTION] Six RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8).\nForest plot of improvement of CRP. CRP = C-reactive protein.\nSix RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8).\nForest plot of improvement of CRP. CRP = C-reactive protein.\n[SUBTITLE] 3.4.7. Blood lipid [SUBSECTION] (1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9).\n(2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10).\n(3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11).\n(4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12).\nForest plot of improvement of TC. TC = total cholesterol.\nForest plot of improvement of TG. TG = triglycerides.\nForest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol.\nForest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol.\n(1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9).\n(2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10).\n(3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11).\n(4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12).\nForest plot of improvement of TC. TC = total cholesterol.\nForest plot of improvement of TG. TG = triglycerides.\nForest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol.\nForest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol.\n[SUBTITLE] 3.5. Adverse reactions [SUBSECTION] Five cases in the treatment group and 9 cases in the control group had adverse reactions, with an incidence of 4.1%, 7.5%, respectively. The incidence of adverse reactions in the treatment of PCI patients with YQHX was relatively lower.\nFive cases in the treatment group and 9 cases in the control group had adverse reactions, with an incidence of 4.1%, 7.5%, respectively. The incidence of adverse reactions in the treatment of PCI patients with YQHX was relatively lower.\n[SUBTITLE] 3.6. Publication bias [SUBSECTION] In order to detect possible publication bias, the 13 trials of ORR were analyzed with a fixed effects model. The funnel plot of ORR was asymmetrical, indicating the presence of publication bias (Fig. 13).\nFunnel plot of the overall response rate.\nIn order to detect possible publication bias, the 13 trials of ORR were analyzed with a fixed effects model. The funnel plot of ORR was asymmetrical, indicating the presence of publication bias (Fig. 13).\nFunnel plot of the overall response rate.", "The search of 7 databases (English or Chinese) identified 283 records for further evaluation (Fig. 1). 23 RCTs of them were eligible.[4–26] All studies involved patient consent, and the informed consent was given.\nFlow diagram of study selection and identification.", "All of the 23 trials included were conducted in China and published in Chinese. All studies were performed in China, with a total of 1868 patients involved (936 the in control group, and 932 in the experimental group). In addition, all the studies exhibited comparable baseline patient characteristics, including age and gender (1053 male, and 815 female). The characteristics of the selected studies are shown in Table 2.\nCharacteristics of included studies.\nCRP = C-reactive protein, CWM = conventional western medicine, ECG = electrocardiogram, HDL-C = high-density lipoprotein cholesterol, LDL-C = low density lipoprotein-cholesterol, MACE = major adverse cardiovascular events, NA = not applicable, ORR = overall response rate, TC = total cholesterol, TCMSRR = TCM syndrome response rate, TG = triglycerides, YQHX = Yiqi Huoxue.", "Among the trials, 14 studies[5–17,25] stated the method of the sequence generation with random number table and drawing, while none of the 22 studies reported details for sample size calculations and none was double-blind or placebo controlled study. Additionally, none mentioned allocation concealment or blinding methods. Fourteen trials included[5–9,12–15,17–20,22] were assessed as low risk of bias in incomplete outcome data. Fourteen of the trials included[5,6,8,10,11,13,14,16–22] were assessed as low risk of reporting bias, and the other 4,[7,9,12,15] as unclear risk of reporting bias. The details of the risk of bias of each trial are presented in Figures 2 and 3.\nRisk of bias: reviewing authors’ judgments about each risk of bias item for each included study.\nSummary of the risk of bias assessment for included trials.", "The outcomes, ORR (13 trials), TCMSRR (8 trials), ECG (9 trials), MACE (9 trials), blood lipid (TC, TG) level (7 trials), (HDL-C, LDL-C) level (6 trials), and CRP (6 trials) were analyzed.\n[SUBTITLE] 3.4.1. Overall response rate [SUBSECTION] Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4).\nForest plot of overall response rate.\n Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4).\nForest plot of overall response rate.\n[SUBTITLE] 3.4.2. ECG improvement [SUBSECTION] Nine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5).\nForest plot of improvement of ECG. ECG = electrocardiogram.\nNine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5).\nForest plot of improvement of ECG. ECG = electrocardiogram.\n[SUBTITLE] 3.4.3. TCM syndrome response rate [SUBSECTION] Eight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6).\nForest plot of TCM syndrome response rate. TCM = traditional Chinese medicine.\nEight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6).\nForest plot of TCM syndrome response rate. TCM = traditional Chinese medicine.\n[SUBTITLE] 3.4.4. Major adverse cardiovascular events [SUBSECTION] Nine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7).\nForest plot of improvement of MACE. MACE = major adverse cardiovascular events.\nNine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7).\nForest plot of improvement of MACE. MACE = major adverse cardiovascular events.\n[SUBTITLE] 3.4.5. GRADE assessment [SUBSECTION] However, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3).\nSummary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI.\n There were serious limitations of methodological quality of included trials according to the risk of bias assessment.\n There were serious publication bias between the studies.\nCAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine.\nHowever, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3).\nSummary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI.\n There were serious limitations of methodological quality of included trials according to the risk of bias assessment.\n There were serious publication bias between the studies.\nCAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine.\n[SUBTITLE] 3.4.6. CRP [SUBSECTION] Six RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8).\nForest plot of improvement of CRP. CRP = C-reactive protein.\nSix RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8).\nForest plot of improvement of CRP. CRP = C-reactive protein.\n[SUBTITLE] 3.4.7. Blood lipid [SUBSECTION] (1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9).\n(2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10).\n(3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11).\n(4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12).\nForest plot of improvement of TC. TC = total cholesterol.\nForest plot of improvement of TG. TG = triglycerides.\nForest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol.\nForest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol.\n(1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9).\n(2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10).\n(3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11).\n(4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12).\nForest plot of improvement of TC. TC = total cholesterol.\nForest plot of improvement of TG. TG = triglycerides.\nForest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol.\nForest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol.", " Thirteen RCTs[5,6,8–10,12,14,15,17,19,21,23,25] reported ORR and found an obvious difference (RR = 1.24, 95% CI 1.17–1.32, 1092 participants, P < .00001), which meant that YQHX plus CWM was significantly better than CWM. No heterogeneity was found (I2 = 0%, P = .69) (Fig. 4).\nForest plot of overall response rate.", "Nine RCTs[9,10,12,13,15,18,19,21,22] evaluated the effect of ECG improvement and found an significant difference (RR = 1.33, 95% CI 1.21–1.46, 699 participants, P < .0001). The result indicated that YQHX plus CWM was significantly better than CWM in ECG improvement, and there was significant homogeneity (I2 = 0%, P = .86) (Fig. 5).\nForest plot of improvement of ECG. ECG = electrocardiogram.", "Eight RCTs[9,11–14,18,21,22] reported TCMSRR and found an obvious difference (RR = 1.26, 95% CI 1.17–1.36, 508 participants, P < .00001). The result indicated that YQHX combined with CWM was significantly better than CWM in the TCMSRR and there was significant homogeneity (I2 = 0%, P = .96) (Fig. 6).\nForest plot of TCM syndrome response rate. TCM = traditional Chinese medicine.", "Nine RCTs[4,6,10,11,13,16,17,19,20] reported MACE and found an obvious difference (RR = 0.26, 95% CI 0.16–0.42, 723 participants, P < .00001). The result indicated that YQHX combined with CWM were significantly better than CWM in the MACE and there was significant homogeneity (I2 = 0%, P = .99) (Fig. 7).\nForest plot of improvement of MACE. MACE = major adverse cardiovascular events.", "However, due to the poor methodology of the studies included and the obvious statistical heterogeneity among trials, quality of the evidence for all 4 outcomes (ORR, TCMSRR, ECG, MACE) were “low” and “very low,” according to the GRADE assessment (see Table 3).\nSummary of finding table of Yiqi Huoxue formula with conventional western medicine for patients were diagnosed with CAD and received PCI.\n There were serious limitations of methodological quality of included trials according to the risk of bias assessment.\n There were serious publication bias between the studies.\nCAD = coronary artery disease, CI = confidence interval, ECG = electrocardiogram, PCI = percutaneous coronary intervention, RR = risk ratio, TCM = Traditional Chinese Medicine.", "Six RCTs[9,12,15–17,24] reported CRP and found an obvious difference (SMD = −4.08, 95% CI − 5.78–2.37, 407 participants, P < .00001). The trials were divided into 4 subgroups which were 14d, 28d, 56d and 6m by course of intervention. The meta-analysis of 3 subgroups of CRP assessment showed obvious differences (28d: SMD = -2.00, 95% CI -4.79 to 0.78, 151 participants, P < .00001; 6m: SMD = -3.34, 95% CI -5.32 to -1.35, 136 participants, P = .001) with significant heterogeneity (28d: I2 = 98%, P = .16; 6m: I2 = 93%, P < .00001). There was significant difference between subgroups (I2 = 94.2%, P < .00001) (Fig. 8).\nForest plot of improvement of CRP. CRP = C-reactive protein.", "(1) TC 8 RCTs[5,7,11,12,15,21,24,26] reported TC and found an obvious difference (SMD = −1.34, 95% CI − 2.20 to − 0.47, 563 participants, P = .003). The trials were divided into 4 subgroups which were 15d, 28d, 56d, and 3-6m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = -0.19, 95% CI -0.70 to 0.32, 60 participants, P = .47; 28d: SMD = -1.15, 95% CI -3.45 to 1.15, 151 participants, P = .33; 3-6m: SMD = -0.33, 95% CI -0.69 to 0.03, 120 participants, P = .08). In contrast, the 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = -2.55, 95% CI -3.24 to -1.85, 232 participants, P < .00001) with low heterogeneity (I2 = 34%, P = .22). There was significant difference between subgroups (I2 = 91.9%, P < .00001) (Fig. 9).\n(2) TG 8 RCTs[5,7,11,12,15,21,24,26] reported TG and found no obvious difference (SMD = −0.67, 95% CI − 1.41 to 0.07, 563 participants, P = .07). In TG group, the trials were divided into 4 subgroups which were 15d, 28d, 56d and 3-6m. The meta-analysis of subgroups showed no significant differences (15d: SMD = -0.08, 95% CI -0.59 to 0.43, 60 participants, P = .76; 28d: SMD = -0.24, 95% CI -0.56 to 0.08, 151 participants, P = .14; 56d: SMD = -1.55, 95% CI -1.41 to 0.07, 232 participants, P = .15; 3-6m: SMD = -0.10, 95% CI -0.46 to 0.26, 120 participants, P = .57). There was no significant difference between subgroups (I2 = 0%, P = .55) (Fig. 10).\n(3) HDL-C 6 RCTs[5,11,12,15,21,24] reported HDL-C and found an significant difference (SMD = 0.56, 95% CI 0.15–0.98, 399 participants, P = .008). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (15d: SMD = 0.08, 95% CI -0.43 to 0.58, 60 participants, P = .76; 28d: SMD = 0.52, 95% CI -0.06 to 0.10, 151 participants, P = .08; 3m: SMD = 0.11, 95% CI -0.40 to 0.61, 60 participants, P = .68). The 56d subgroup[21,24] showed that 2 groups had obvious difference (SMD = 1.08, 95% CI 0.36 to 1.81, 128 participants, P < .05) with heterogeneity (I2 = 73%, P = .05). There was significant difference between subgroups (I2 = 51.9%, P = .10) (Fig. 11).\n(4) LDL-C 7 RCTs[5,11,12,15,21,24,26] reported LDL-C and found an obvious difference (SMD = −1.44, 95% CI -2.53 to -0.35, 503 participants, P = .0009). The trials were divided into 4 subgroups which were 15d, 28d, 56d and 3m. The meta-analysis of 3 subgroups showed no significant differences (28d: SMD = -0.32, 95% CI -1.07 to 0.42, 151 participants, P = .40; 56d: SMD = -2.38, 95% CI -5.04 to 0.28, 232 participants, P = .08; 3m: SMD = -0.39, 95% CI -0.90 to 0.12, 60 participants, P = .14). The 15d group[14] showed that 2 groups had obvious difference (SMD = -2.03, 95% CI -2.67 to -1.40, 60 participants, P < .00001). There was significant difference between subgroups (I2 = 84.8%, P = .0002) (Fig. 12).\nForest plot of improvement of TC. TC = total cholesterol.\nForest plot of improvement of TG. TG = triglycerides.\nForest plot of improvement of HDL-C. HDL-C = high-density lipoprotein cholesterol.\nForest plot of improvement of LDL-C. LDL-C = Low density lipoprotein-cholesterol.", "Five cases in the treatment group and 9 cases in the control group had adverse reactions, with an incidence of 4.1%, 7.5%, respectively. The incidence of adverse reactions in the treatment of PCI patients with YQHX was relatively lower.", "In order to detect possible publication bias, the 13 trials of ORR were analyzed with a fixed effects model. The funnel plot of ORR was asymmetrical, indicating the presence of publication bias (Fig. 13).\nFunnel plot of the overall response rate.", "In this study, we systematically evaluated the RCT of the control group treated with CWM, while the experimental group were added YQHX. The ORR, ECG, TCMSRR, and MACE were the important indicators of clinical efficacy and quality of life of the patients.[27,28] The results showed that these 4 outcomes of CAD patients after PCI treated with CWM alone were inferior to CWM plus YQHX herbs, which had higher safety and fewer side effects.\nInflammatory response is an important factor affecting plaque progression.[35] CRP is a kind of protein secreted in the blood during acute inflammation and reflects the inflammatory status of the organism.[29] The concentration of CRP in serum is positively correlated with the formation of atherosclerosis and the severity of CAD.[30,31] Therefore, CRP can be used as an important index to measure the risk of CAD after PCI.[37] The results showed that the combination of YQHX with CWM was better than CWM alone in reducing CRP.\nHyperlipidemia is one of the risk factors of atherosclerosis.[32] Elevated blood lipids can damage endothelial cells, increase vascular permeability, cause cholesterol deposition, plaque formation, and thrombosis, and lead to coronary stenosis.[33,34] Therefore, abnormal blood lipid level is an independent risk factor for restenosis after PCI. The results indicated that the combination of YQHX with CWM was better than CWM alone in reducing TC and increasing HDL-C.\nPCI is currently recognized as the most effective and safe way to restore myocardial reperfusion when it meets the indications. PCI is an exogenous injury, and the persistent blood stasis is not relieved by the operation. The mechanical injury of PCI can also induce the formation of new blood stasis, then damage the local tissue, leading to the aggravation of blood stasis.[35,36] PCI, having the function of “breaking blood,” is easy to consume the healthy Qi of the injured organism. The etiology and pathogenesis of PCI after CAD are Qi deficiency and blood stasis, for which YQHX is the basic therapy. Results from previous studies showed that YQHX, as a complementary treatment, may alleviate the clinical symptoms, reduce the onset of angina pectoris and the side effects of drugs, increase the exercise endurance, prevent in-stent restenosis, and improve the quality of life of CAD after PCI patients.[28,37]\nBased on the above results, it can be concluded that the combination of YQHX with CWM is superior to CWM alone in the treatment of CAD after PCI. This integrated therapy has fewer adverse reactions and higher safety. However, the quality of the methodologies being used is not high enough. As all the studies were conducted in Chinese population, the results cannot necessarily be extrapolated to other populations. Therefore, more high-quality, multi-center, and largely sampled RCTs need to be carried out to provide more reliable evidence-based medical basis for clinical guidance.", "Thank all the authors for their contributions to this article.", "Data curation: Miao Zhang, Qiting Chen.\nFormal analysis: Ming-Yue Sun, Qiting Chen.\nFunding acquisition: Miao Zhang, Feng-Qin Xu.\nInvestigation: Hui-Jun Yin.\nMethodology: Qiting Chen.\nProject administration: Wenbo Wei.\nSoftware: Miao Zhang, Qiting Chen.\nSupervision: Zongzheng Chen, Hui-Jun Yin.\nValidation: Zongzheng Chen.\nVisualization: Ruiting Wang.\nWriting – original draft: Miao Zhang, Ming-Yue Sun.\nWriting – review & editing: Miao Zhang, Wenbo Wei, Feng-Qin Xu, Hui-Jun Yin." ]
[ "intro", "methods", null, null, null, null, "methods", "results", null, null, null, null, null, null, null, null, null, null, null, null, null, "discussion", null, null ]
[ "coronary artery disease", "meta-analysis", "percutaneous coronary intervention", "Yiqi Huoxue" ]
Systematic review and meta-analysis of acupuncture in the treatment of cognitive impairment after stroke.
36254056
We aim to make a systematic evaluation of the clinical efficacy of acupuncture in the treatment of cognitive impairment after stroke, to provide evidence-based medical evidence for clinical practice.
BACKGROUND
We searched all the randomized controlled trials of China National Knowledge Infrastructure, Wan fang data knowledge service platform, VIP Chinese periodical service platform full-text Journal Database, Chinese Biomedical Literature Database, Cochrane Library Database, and PubMed Database about acupuncture treatment of post-stroke cognitive impairment (PSCI). Two researchers independently screened the literature and extracted the data according to the inclusion and exclusion criteria. The bias risk assessment manual of Cochrane collaboration Network was used to evaluate the bias risk, and all data were analyzed by Stata16.0.
METHODS
Fourteen articles were included, with a total of 2402 patients. Meta-analysis showed that acupuncture combined with routine therapy could significantly reduce the score of cognitive impairment symptoms compared with the control group. The mini-mental state examination scale (MMSE) score (weighted mean difference [WMD] = 3.23, 95% confidence interval [CI]: 1.89-4.56, P < .01), Montreal cognitive assessment scale (MoCA) score (WMD = 3.41, 95% CI: 0.93-5.89, P < .01), Barthel index of activities of daily living (MBI) score (WMD = 4.59, 95% CI: 1.43-7.75, P < .01), and Lowenstein assessment scale (LOTCA) score (WMD = 8.60, 95% CI: 6.32-10.89, P = .00) were significantly improved in the patients receiving group acupuncture combined with routine therapy.
RESULTS
Acupuncture combined with routine therapy seems to be more effective than conventional therapy alone in the treatment of PSCI. However, the differences between different acupuncture types need to be clarified in more high-quality randomized controlled trials.
CONCLUSION
[ "Activities of Daily Living", "Acupuncture Therapy", "Cognitive Dysfunction", "Humans", "Stroke", "Treatment Outcome" ]
9575739
1. Introduction
Stroke is a common disease that often occurs in people over 50 years old and has the characteristics of high incidence, high mortality, and high disability rate. Its clinical manifestation is transient or permanent brain dysfunction, which occupies the first place in the cause of death of urban residents. Stroke can be divided into 2 categories: ischemic stroke and hemorrhagic stroke.[1] From mild cognitive impairment to severe post-stroke dementia, the mortality, disability, and high nursing costs of post-stroke cognitive impairment (PSCI) increased step by step.[2,3] PSCI has a significant impact on patients, their families, and medical resources. Due to the huge and increasing global burden of stroke, PSCI has become an increasingly serious public health care challenge.[4] Acupuncture is currently recognized as one of the first choices for the treatment of cognitive dysfunction after stroke in China and is often used in combination with other conventional therapies.[5] In addition, acupuncture has been accepted in stroke rehabilitation in many countries, as this treatment is relatively safe and effective in improving post-stroke chronic symptoms, such as disability, dysphagia, and insomnia.[6–9] In the United States, approximately half the stroke survivors engage in some form of acupuncture therapy.[10] Patients with stroke also have a higher TCM utilization rate than people without stroke.[11] According to a modern understanding of acupuncture mechanisms, acupuncture may elicit vegetative reflexes, thereby changing the flow of blood and enhancing the functional properties of connective tissue and organs. Acupuncture signals are recognized as a potent form of sensory stimulation that ascends mainly through the spinal ventrolateral funiculus to the brain.[12] The mechanisms of acupuncture-mediated neuroplasticity have recently attracted increased interest. Accordingly, acupuncture modulation over several cognition- or aging-related gene expressions,[13] plasticity signaling pathways,[14,15] and brain functional connectivities[16] has been studied. The evidence for acupuncture in clinical practice is currently inconsistent; therefore, the purpose of this systematic review is to analyze the randomized controlled trials of acupuncture in the treatment of PSCI to examine whether conventional treatment combined with acupuncture has the additional benefit and provide an evidence-based medicine basis for the clinical application of acupuncture.
2. Materials and Methods
[SUBTITLE] 2.1. Search strategy [SUBSECTION] By using the combination of subject words and free words, 4 Chinese databases were searched, including the Chinese periodical full-text database, Wanfang data knowledge service platform, Chinese biomedical literature database, VIP Chinese periodical service platform, PubMed, and Cochrane Library. The retrieval time is from January 1, 2016, to January 10, 2021. English keywords and corresponding Chinese keywords: acupuncture, acupuncture therapy, stroke, infarction, hemorrhage cerebrovascular, cerebrovascular disease, electroacupuncture, electro-acupuncture, cognitive dysfunction, cognitive impairment, cognitive function, etc. No filters are used. By using the combination of subject words and free words, 4 Chinese databases were searched, including the Chinese periodical full-text database, Wanfang data knowledge service platform, Chinese biomedical literature database, VIP Chinese periodical service platform, PubMed, and Cochrane Library. The retrieval time is from January 1, 2016, to January 10, 2021. English keywords and corresponding Chinese keywords: acupuncture, acupuncture therapy, stroke, infarction, hemorrhage cerebrovascular, cerebrovascular disease, electroacupuncture, electro-acupuncture, cognitive dysfunction, cognitive impairment, cognitive function, etc. No filters are used.
4.3. Meta-analysis results
[SUBTITLE] 4.3.1.1. MMSE score. [SUBSECTION] Nine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved. MMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale. Nine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved. MMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale. [SUBTITLE] 4.3.1.2. MoCA score [SUBSECTION] Eight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved. MoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale. Eight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved. MoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale. [SUBTITLE] 4.3.1.3. MBI score [SUBSECTION] Four studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc. MBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index. Four studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc. MBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index. [SUBTITLE] 4.3.1.4. LOTCA score [SUBSECTION] Two studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly. LOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale. Two studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly. LOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale.
null
null
[ "2.1. Search strategy", "3. Literature inclusion and Exclusion criteria", "3.1. Literature inclusion criteria", "3.2. Literature exclusion criteria", "3.3. Literature screening", "3.4. Data extraction", "3.5. Document quality evaluation", "3.6. Statistical analysis", "3.7. Ethical approval", "4. Results", "4.1. Basic characteristics of the included literature", "4.2. Quality evaluation", "4.3.1.1. MMSE score.", "4.3.1.2. MoCA score", "4.3.1.3. MBI score", "4.3.1.4. LOTCA score", "4.4. Sensitivity analysis", "4.5. Publication bias analysis", "Author contributions" ]
[ "By using the combination of subject words and free words, 4 Chinese databases were searched, including the Chinese periodical full-text database, Wanfang data knowledge service platform, Chinese biomedical literature database, VIP Chinese periodical service platform, PubMed, and Cochrane Library. The retrieval time is from January 1, 2016, to January 10, 2021. English keywords and corresponding Chinese keywords: acupuncture, acupuncture therapy, stroke, infarction, hemorrhage cerebrovascular, cerebrovascular disease, electroacupuncture, electro-acupuncture, cognitive dysfunction, cognitive impairment, cognitive function, etc. No filters are used.", "[SUBTITLE] 3.1. Literature inclusion criteria [SUBSECTION] We formulated inclusion criteria by the principles of PICOS, and only documents that meet all of the following criteria would be included. Population: clear diagnostic criteria of a stroke complicated with cognitive impairment; intervention: experimental group received routine treatment and acupuncture treatment without restriction on mode and course; comparison: control group received routine treatment; outcome indicators: mini-mental state examination scale (MMSE) (A standardized mental state examination tool for simple screening of temporal orientation, place orientation, immediate memory, attention and computation, delayed memory, language, and visual space),[17] Montreal cognitive assessment scale (MoCA) (An assessment tool for rapid screening of cognitive dysfunction in terms of orientation, short-term memory, executive function, language, abstraction, animal naming, attention, clock-drawing test),[17] Barthel index of activities of daily living (MBI) (A scale used to measure changes in independent living skills such as eating, dressing, continence, and mobility in older people before and after treatment),[18] Lowenstein assessment scale (LOTCA) (A standardized neuropsychological test that assesses orientation, perception, visual motor organization, and mental functioning).[19] Study type: Randomized controlled trials. The language was Chinese or English.\nWe formulated inclusion criteria by the principles of PICOS, and only documents that meet all of the following criteria would be included. Population: clear diagnostic criteria of a stroke complicated with cognitive impairment; intervention: experimental group received routine treatment and acupuncture treatment without restriction on mode and course; comparison: control group received routine treatment; outcome indicators: mini-mental state examination scale (MMSE) (A standardized mental state examination tool for simple screening of temporal orientation, place orientation, immediate memory, attention and computation, delayed memory, language, and visual space),[17] Montreal cognitive assessment scale (MoCA) (An assessment tool for rapid screening of cognitive dysfunction in terms of orientation, short-term memory, executive function, language, abstraction, animal naming, attention, clock-drawing test),[17] Barthel index of activities of daily living (MBI) (A scale used to measure changes in independent living skills such as eating, dressing, continence, and mobility in older people before and after treatment),[18] Lowenstein assessment scale (LOTCA) (A standardized neuropsychological test that assesses orientation, perception, visual motor organization, and mental functioning).[19] Study type: Randomized controlled trials. The language was Chinese or English.\n[SUBTITLE] 3.2. Literature exclusion criteria [SUBSECTION] Acupuncture therapy was included in the control group; the experimental group did not combine acupuncture therapy with conventional therapy; the course of treatment was <4 weeks; reviews, case reports, guidelines, or, comment; data was incomplete and could not be available.\nAcupuncture therapy was included in the control group; the experimental group did not combine acupuncture therapy with conventional therapy; the course of treatment was <4 weeks; reviews, case reports, guidelines, or, comment; data was incomplete and could not be available.\n[SUBTITLE] 3.3. Literature screening [SUBSECTION] First of all, duplicate documents were eliminated. Two researchers excluded literature that did not meet the inclusion criteria by reading the title and abstract. The possible literature was read in full text to further judge whether it was included or not. After completion of the preliminary screening, any differences will be resolved through discussion among all authors.\nFirst of all, duplicate documents were eliminated. Two researchers excluded literature that did not meet the inclusion criteria by reading the title and abstract. The possible literature was read in full text to further judge whether it was included or not. After completion of the preliminary screening, any differences will be resolved through discussion among all authors.\n[SUBTITLE] 3.4. Data extraction [SUBSECTION] After numbering the final included documents, the data was extracted using the “data extraction Table” designed by Excel, and the extracted contents included: basic information: literature name, author, year of publication, etc. Research characteristics: intervention measures, sample size, course of treatment, outcome index, adverse reaction report, etc.\nAfter numbering the final included documents, the data was extracted using the “data extraction Table” designed by Excel, and the extracted contents included: basic information: literature name, author, year of publication, etc. Research characteristics: intervention measures, sample size, course of treatment, outcome index, adverse reaction report, etc.\n[SUBTITLE] 3.5. Document quality evaluation [SUBSECTION] The literature quality evaluation method evaluates the risk bias of the included literature quality according to the Cochrane systematic evaluation manual, and if there are differences in the evaluation results, it will be discussed and resolved with the third researcher. The evaluation contents include random allocation method, blind method, allocation concealment, data integrity of research results, selective reporting, and other biases. The degree of risk bias, it can be divided into 3 levels: low risk, high risk, and unclear.\nThe literature quality evaluation method evaluates the risk bias of the included literature quality according to the Cochrane systematic evaluation manual, and if there are differences in the evaluation results, it will be discussed and resolved with the third researcher. The evaluation contents include random allocation method, blind method, allocation concealment, data integrity of research results, selective reporting, and other biases. The degree of risk bias, it can be divided into 3 levels: low risk, high risk, and unclear.\n[SUBTITLE] 3.6. Statistical analysis [SUBSECTION] The data was analyzed by Stata16 software, the weighted mean difference (WMD) was used for the measurement data, and the corresponding confidence interval (CI) of each effect was calculated by 95%. If P < .01, the difference was statistically significant. The heterogeneity of the included research was tested, and the corresponding effect model was selected according to the values of the Q test and I2 test. If P ≥ .05 and I2 ≤ 50%, the statistical heterogeneity among the studies is small, then the “fixed effect model” is adopted for the meta-analysis of the curative effect; if P < .05, I2 > 50%, it indicates that the heterogeneity between the studies is large. The “random effect model” was adopted to analyze the curative effect. In addition, the sources of heterogeneity were analyzed by subgroup analysis or sensitivity analysis. We use the funnel chart to detect publication bias, and Begg and Egger tests were used to determine that. When the funnel chart shows a symmetrical inverted triangle and the P values of Begg and Egger are >.05, we believe that there is no potential publication bias, otherwise, there may be publication bias.\nThe data was analyzed by Stata16 software, the weighted mean difference (WMD) was used for the measurement data, and the corresponding confidence interval (CI) of each effect was calculated by 95%. If P < .01, the difference was statistically significant. The heterogeneity of the included research was tested, and the corresponding effect model was selected according to the values of the Q test and I2 test. If P ≥ .05 and I2 ≤ 50%, the statistical heterogeneity among the studies is small, then the “fixed effect model” is adopted for the meta-analysis of the curative effect; if P < .05, I2 > 50%, it indicates that the heterogeneity between the studies is large. The “random effect model” was adopted to analyze the curative effect. In addition, the sources of heterogeneity were analyzed by subgroup analysis or sensitivity analysis. We use the funnel chart to detect publication bias, and Begg and Egger tests were used to determine that. When the funnel chart shows a symmetrical inverted triangle and the P values of Begg and Egger are >.05, we believe that there is no potential publication bias, otherwise, there may be publication bias.\n[SUBTITLE] 3.7. Ethical approval [SUBSECTION] Not applicable.\nThis paper is a kind of quantitative comprehensive evaluation based on literature data of multiple types independent research studies.\nNot applicable.\nThis paper is a kind of quantitative comprehensive evaluation based on literature data of multiple types independent research studies.", "We formulated inclusion criteria by the principles of PICOS, and only documents that meet all of the following criteria would be included. Population: clear diagnostic criteria of a stroke complicated with cognitive impairment; intervention: experimental group received routine treatment and acupuncture treatment without restriction on mode and course; comparison: control group received routine treatment; outcome indicators: mini-mental state examination scale (MMSE) (A standardized mental state examination tool for simple screening of temporal orientation, place orientation, immediate memory, attention and computation, delayed memory, language, and visual space),[17] Montreal cognitive assessment scale (MoCA) (An assessment tool for rapid screening of cognitive dysfunction in terms of orientation, short-term memory, executive function, language, abstraction, animal naming, attention, clock-drawing test),[17] Barthel index of activities of daily living (MBI) (A scale used to measure changes in independent living skills such as eating, dressing, continence, and mobility in older people before and after treatment),[18] Lowenstein assessment scale (LOTCA) (A standardized neuropsychological test that assesses orientation, perception, visual motor organization, and mental functioning).[19] Study type: Randomized controlled trials. The language was Chinese or English.", "Acupuncture therapy was included in the control group; the experimental group did not combine acupuncture therapy with conventional therapy; the course of treatment was <4 weeks; reviews, case reports, guidelines, or, comment; data was incomplete and could not be available.", "First of all, duplicate documents were eliminated. Two researchers excluded literature that did not meet the inclusion criteria by reading the title and abstract. The possible literature was read in full text to further judge whether it was included or not. After completion of the preliminary screening, any differences will be resolved through discussion among all authors.", "After numbering the final included documents, the data was extracted using the “data extraction Table” designed by Excel, and the extracted contents included: basic information: literature name, author, year of publication, etc. Research characteristics: intervention measures, sample size, course of treatment, outcome index, adverse reaction report, etc.", "The literature quality evaluation method evaluates the risk bias of the included literature quality according to the Cochrane systematic evaluation manual, and if there are differences in the evaluation results, it will be discussed and resolved with the third researcher. The evaluation contents include random allocation method, blind method, allocation concealment, data integrity of research results, selective reporting, and other biases. The degree of risk bias, it can be divided into 3 levels: low risk, high risk, and unclear.", "The data was analyzed by Stata16 software, the weighted mean difference (WMD) was used for the measurement data, and the corresponding confidence interval (CI) of each effect was calculated by 95%. If P < .01, the difference was statistically significant. The heterogeneity of the included research was tested, and the corresponding effect model was selected according to the values of the Q test and I2 test. If P ≥ .05 and I2 ≤ 50%, the statistical heterogeneity among the studies is small, then the “fixed effect model” is adopted for the meta-analysis of the curative effect; if P < .05, I2 > 50%, it indicates that the heterogeneity between the studies is large. The “random effect model” was adopted to analyze the curative effect. In addition, the sources of heterogeneity were analyzed by subgroup analysis or sensitivity analysis. We use the funnel chart to detect publication bias, and Begg and Egger tests were used to determine that. When the funnel chart shows a symmetrical inverted triangle and the P values of Begg and Egger are >.05, we believe that there is no potential publication bias, otherwise, there may be publication bias.", "Not applicable.\nThis paper is a kind of quantitative comprehensive evaluation based on literature data of multiple types independent research studies.", "According to the retrieval strategy, 1464 articles were retrieved, 273 repetitive articles were excluded, and 1191 articles were excluded after reading titles/abstracts manually. Re-screening and reading the remaining 100 full-text articles, and finally including 14 articles. The literature screening process and results are shown in Figure 1.\nFlow chart of literature screening.\n[SUBTITLE] 4.1. Basic characteristics of the included literature [SUBSECTION] The basic characteristics of the literature were included in 14 articles, with a total of 2402 patients. The basic characteristics of each study are shown in Table 1.\nThe characteristics of included studies.\nLOTCA = Lowenstein assessment scale, MBI = activities of daily living Barthel index, MMSE = mini-mental state scale, MOCA = Montreal cognitive assessment scale, rTMS = transcranial magnetic stimulations.\nThe basic characteristics of the literature were included in 14 articles, with a total of 2402 patients. The basic characteristics of each study are shown in Table 1.\nThe characteristics of included studies.\nLOTCA = Lowenstein assessment scale, MBI = activities of daily living Barthel index, MMSE = mini-mental state scale, MOCA = Montreal cognitive assessment scale, rTMS = transcranial magnetic stimulations.\n[SUBTITLE] 4.2. Quality evaluation [SUBSECTION] The risk bias was assessed according to the Cochrane guideline. Ten articles[20–22,24,25,28,29,31–33] were grouped by the random number table method, 1 article[27] used the computer random grouping method, and the other 3 articles[23,26,30] did not mention the specific distribution scheme. In the aspect of data collection and evaluation, 1 article[27] mentioned that the implementation of the blind method was single-blind, the other 11 articles did not mention the blind method. In terms of other biases, the diagnostic criteria of 8 articles[20,21,25,27,29–31,33] are not specified, which may affect the authenticity of the study, so they are evaluated as high risk. The quality evaluation of the included literature is shown in Table 2.\nQuality assessment of included studies.\nThe risk bias was assessed according to the Cochrane guideline. Ten articles[20–22,24,25,28,29,31–33] were grouped by the random number table method, 1 article[27] used the computer random grouping method, and the other 3 articles[23,26,30] did not mention the specific distribution scheme. In the aspect of data collection and evaluation, 1 article[27] mentioned that the implementation of the blind method was single-blind, the other 11 articles did not mention the blind method. In terms of other biases, the diagnostic criteria of 8 articles[20,21,25,27,29–31,33] are not specified, which may affect the authenticity of the study, so they are evaluated as high risk. The quality evaluation of the included literature is shown in Table 2.\nQuality assessment of included studies.\n[SUBTITLE] 4.3. Meta-analysis results [SUBSECTION] [SUBTITLE] 4.3.1.1. MMSE score. [SUBSECTION] Nine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved.\nMMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale.\nNine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved.\nMMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale.\n[SUBTITLE] 4.3.1.2. MoCA score [SUBSECTION] Eight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved.\nMoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale.\nEight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved.\nMoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale.\n[SUBTITLE] 4.3.1.3. MBI score [SUBSECTION] Four studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc.\nMBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index.\nFour studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc.\nMBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index.\n[SUBTITLE] 4.3.1.4. LOTCA score [SUBSECTION] Two studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly.\nLOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale.\nTwo studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly.\nLOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale.\n[SUBTITLE] 4.3.1.1. MMSE score. [SUBSECTION] Nine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved.\nMMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale.\nNine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved.\nMMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale.\n[SUBTITLE] 4.3.1.2. MoCA score [SUBSECTION] Eight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved.\nMoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale.\nEight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved.\nMoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale.\n[SUBTITLE] 4.3.1.3. MBI score [SUBSECTION] Four studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc.\nMBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index.\nFour studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc.\nMBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index.\n[SUBTITLE] 4.3.1.4. LOTCA score [SUBSECTION] Two studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly.\nLOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale.\nTwo studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly.\nLOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale.\n[SUBTITLE] 4.4. Sensitivity analysis [SUBSECTION] Sensitivity analysis was conducted by excluding 1 study at a time. We find that the results were robust and did not change the conclusions due to the exclusion of any study.\nSensitivity analysis was conducted by excluding 1 study at a time. We find that the results were robust and did not change the conclusions due to the exclusion of any study.\n[SUBTITLE] 4.5. Publication bias analysis [SUBSECTION] Because the number of articles included in the MBI score and LOTCA score index groups was <5, it is not suitable for funnel chart analysis. The MMSE score and MoCA score index groups were evaluated for the existence of publication bias. The results showed that although individual subjects deviate from the CI of the funnel diagram, they are roughly symmetrical. In addition, the P values of Begg and Egger tests of all indicators were >.05, indicating that there was no potential publication bias.\nBecause the number of articles included in the MBI score and LOTCA score index groups was <5, it is not suitable for funnel chart analysis. The MMSE score and MoCA score index groups were evaluated for the existence of publication bias. The results showed that although individual subjects deviate from the CI of the funnel diagram, they are roughly symmetrical. In addition, the P values of Begg and Egger tests of all indicators were >.05, indicating that there was no potential publication bias.", "The basic characteristics of the literature were included in 14 articles, with a total of 2402 patients. The basic characteristics of each study are shown in Table 1.\nThe characteristics of included studies.\nLOTCA = Lowenstein assessment scale, MBI = activities of daily living Barthel index, MMSE = mini-mental state scale, MOCA = Montreal cognitive assessment scale, rTMS = transcranial magnetic stimulations.", "The risk bias was assessed according to the Cochrane guideline. Ten articles[20–22,24,25,28,29,31–33] were grouped by the random number table method, 1 article[27] used the computer random grouping method, and the other 3 articles[23,26,30] did not mention the specific distribution scheme. In the aspect of data collection and evaluation, 1 article[27] mentioned that the implementation of the blind method was single-blind, the other 11 articles did not mention the blind method. In terms of other biases, the diagnostic criteria of 8 articles[20,21,25,27,29–31,33] are not specified, which may affect the authenticity of the study, so they are evaluated as high risk. The quality evaluation of the included literature is shown in Table 2.\nQuality assessment of included studies.", "Nine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved.\nMMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale.", "Eight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved.\nMoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale.", "Four studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc.\nMBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index.", "Two studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly.\nLOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale.", "Sensitivity analysis was conducted by excluding 1 study at a time. We find that the results were robust and did not change the conclusions due to the exclusion of any study.", "Because the number of articles included in the MBI score and LOTCA score index groups was <5, it is not suitable for funnel chart analysis. The MMSE score and MoCA score index groups were evaluated for the existence of publication bias. The results showed that although individual subjects deviate from the CI of the funnel diagram, they are roughly symmetrical. In addition, the P values of Begg and Egger tests of all indicators were >.05, indicating that there was no potential publication bias.", "Methodology: Kang Xiong.\nSoftware: Mei-Ling Zhang.\nSupervision: Bin Wang.\nWriting – original draft: Zhen-Zhi Wang, Zhen Sun.\nWriting – review & editing: Zhen-Zhi Wang, Zhen Sun." ]
[ null, null, null, null, null, null, null, null, null, "results", null, null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Materials and Methods", "2.1. Search strategy", "3. Literature inclusion and Exclusion criteria", "3.1. Literature inclusion criteria", "3.2. Literature exclusion criteria", "3.3. Literature screening", "3.4. Data extraction", "3.5. Document quality evaluation", "3.6. Statistical analysis", "3.7. Ethical approval", "4. Results", "4.1. Basic characteristics of the included literature", "4.2. Quality evaluation", "4.3. Meta-analysis results", "4.3.1.1. MMSE score.", "4.3.1.2. MoCA score", "4.3.1.3. MBI score", "4.3.1.4. LOTCA score", "4.4. Sensitivity analysis", "4.5. Publication bias analysis", "5. Discussion", "Author contributions" ]
[ "Stroke is a common disease that often occurs in people over 50 years old and has the characteristics of high incidence, high mortality, and high disability rate. Its clinical manifestation is transient or permanent brain dysfunction, which occupies the first place in the cause of death of urban residents. Stroke can be divided into 2 categories: ischemic stroke and hemorrhagic stroke.[1] From mild cognitive impairment to severe post-stroke dementia, the mortality, disability, and high nursing costs of post-stroke cognitive impairment (PSCI) increased step by step.[2,3] PSCI has a significant impact on patients, their families, and medical resources. Due to the huge and increasing global burden of stroke, PSCI has become an increasingly serious public health care challenge.[4]\nAcupuncture is currently recognized as one of the first choices for the treatment of cognitive dysfunction after stroke in China and is often used in combination with other conventional therapies.[5] In addition, acupuncture has been accepted in stroke rehabilitation in many countries, as this treatment is relatively safe and effective in improving post-stroke chronic symptoms, such as disability, dysphagia, and insomnia.[6–9] In the United States, approximately half the stroke survivors engage in some form of acupuncture therapy.[10] Patients with stroke also have a higher TCM utilization rate than people without stroke.[11]\nAccording to a modern understanding of acupuncture mechanisms, acupuncture may elicit vegetative reflexes, thereby changing the flow of blood and enhancing the functional properties of connective tissue and organs. Acupuncture signals are recognized as a potent form of sensory stimulation that ascends mainly through the spinal ventrolateral funiculus to the brain.[12] The mechanisms of acupuncture-mediated neuroplasticity have recently attracted increased interest. Accordingly, acupuncture modulation over several cognition- or aging-related gene expressions,[13] plasticity signaling pathways,[14,15] and brain functional connectivities[16] has been studied.\nThe evidence for acupuncture in clinical practice is currently inconsistent; therefore, the purpose of this systematic review is to analyze the randomized controlled trials of acupuncture in the treatment of PSCI to examine whether conventional treatment combined with acupuncture has the additional benefit and provide an evidence-based medicine basis for the clinical application of acupuncture.", "[SUBTITLE] 2.1. Search strategy [SUBSECTION] By using the combination of subject words and free words, 4 Chinese databases were searched, including the Chinese periodical full-text database, Wanfang data knowledge service platform, Chinese biomedical literature database, VIP Chinese periodical service platform, PubMed, and Cochrane Library. The retrieval time is from January 1, 2016, to January 10, 2021. English keywords and corresponding Chinese keywords: acupuncture, acupuncture therapy, stroke, infarction, hemorrhage cerebrovascular, cerebrovascular disease, electroacupuncture, electro-acupuncture, cognitive dysfunction, cognitive impairment, cognitive function, etc. No filters are used.\nBy using the combination of subject words and free words, 4 Chinese databases were searched, including the Chinese periodical full-text database, Wanfang data knowledge service platform, Chinese biomedical literature database, VIP Chinese periodical service platform, PubMed, and Cochrane Library. The retrieval time is from January 1, 2016, to January 10, 2021. English keywords and corresponding Chinese keywords: acupuncture, acupuncture therapy, stroke, infarction, hemorrhage cerebrovascular, cerebrovascular disease, electroacupuncture, electro-acupuncture, cognitive dysfunction, cognitive impairment, cognitive function, etc. No filters are used.", "By using the combination of subject words and free words, 4 Chinese databases were searched, including the Chinese periodical full-text database, Wanfang data knowledge service platform, Chinese biomedical literature database, VIP Chinese periodical service platform, PubMed, and Cochrane Library. The retrieval time is from January 1, 2016, to January 10, 2021. English keywords and corresponding Chinese keywords: acupuncture, acupuncture therapy, stroke, infarction, hemorrhage cerebrovascular, cerebrovascular disease, electroacupuncture, electro-acupuncture, cognitive dysfunction, cognitive impairment, cognitive function, etc. No filters are used.", "[SUBTITLE] 3.1. Literature inclusion criteria [SUBSECTION] We formulated inclusion criteria by the principles of PICOS, and only documents that meet all of the following criteria would be included. Population: clear diagnostic criteria of a stroke complicated with cognitive impairment; intervention: experimental group received routine treatment and acupuncture treatment without restriction on mode and course; comparison: control group received routine treatment; outcome indicators: mini-mental state examination scale (MMSE) (A standardized mental state examination tool for simple screening of temporal orientation, place orientation, immediate memory, attention and computation, delayed memory, language, and visual space),[17] Montreal cognitive assessment scale (MoCA) (An assessment tool for rapid screening of cognitive dysfunction in terms of orientation, short-term memory, executive function, language, abstraction, animal naming, attention, clock-drawing test),[17] Barthel index of activities of daily living (MBI) (A scale used to measure changes in independent living skills such as eating, dressing, continence, and mobility in older people before and after treatment),[18] Lowenstein assessment scale (LOTCA) (A standardized neuropsychological test that assesses orientation, perception, visual motor organization, and mental functioning).[19] Study type: Randomized controlled trials. The language was Chinese or English.\nWe formulated inclusion criteria by the principles of PICOS, and only documents that meet all of the following criteria would be included. Population: clear diagnostic criteria of a stroke complicated with cognitive impairment; intervention: experimental group received routine treatment and acupuncture treatment without restriction on mode and course; comparison: control group received routine treatment; outcome indicators: mini-mental state examination scale (MMSE) (A standardized mental state examination tool for simple screening of temporal orientation, place orientation, immediate memory, attention and computation, delayed memory, language, and visual space),[17] Montreal cognitive assessment scale (MoCA) (An assessment tool for rapid screening of cognitive dysfunction in terms of orientation, short-term memory, executive function, language, abstraction, animal naming, attention, clock-drawing test),[17] Barthel index of activities of daily living (MBI) (A scale used to measure changes in independent living skills such as eating, dressing, continence, and mobility in older people before and after treatment),[18] Lowenstein assessment scale (LOTCA) (A standardized neuropsychological test that assesses orientation, perception, visual motor organization, and mental functioning).[19] Study type: Randomized controlled trials. The language was Chinese or English.\n[SUBTITLE] 3.2. Literature exclusion criteria [SUBSECTION] Acupuncture therapy was included in the control group; the experimental group did not combine acupuncture therapy with conventional therapy; the course of treatment was <4 weeks; reviews, case reports, guidelines, or, comment; data was incomplete and could not be available.\nAcupuncture therapy was included in the control group; the experimental group did not combine acupuncture therapy with conventional therapy; the course of treatment was <4 weeks; reviews, case reports, guidelines, or, comment; data was incomplete and could not be available.\n[SUBTITLE] 3.3. Literature screening [SUBSECTION] First of all, duplicate documents were eliminated. Two researchers excluded literature that did not meet the inclusion criteria by reading the title and abstract. The possible literature was read in full text to further judge whether it was included or not. After completion of the preliminary screening, any differences will be resolved through discussion among all authors.\nFirst of all, duplicate documents were eliminated. Two researchers excluded literature that did not meet the inclusion criteria by reading the title and abstract. The possible literature was read in full text to further judge whether it was included or not. After completion of the preliminary screening, any differences will be resolved through discussion among all authors.\n[SUBTITLE] 3.4. Data extraction [SUBSECTION] After numbering the final included documents, the data was extracted using the “data extraction Table” designed by Excel, and the extracted contents included: basic information: literature name, author, year of publication, etc. Research characteristics: intervention measures, sample size, course of treatment, outcome index, adverse reaction report, etc.\nAfter numbering the final included documents, the data was extracted using the “data extraction Table” designed by Excel, and the extracted contents included: basic information: literature name, author, year of publication, etc. Research characteristics: intervention measures, sample size, course of treatment, outcome index, adverse reaction report, etc.\n[SUBTITLE] 3.5. Document quality evaluation [SUBSECTION] The literature quality evaluation method evaluates the risk bias of the included literature quality according to the Cochrane systematic evaluation manual, and if there are differences in the evaluation results, it will be discussed and resolved with the third researcher. The evaluation contents include random allocation method, blind method, allocation concealment, data integrity of research results, selective reporting, and other biases. The degree of risk bias, it can be divided into 3 levels: low risk, high risk, and unclear.\nThe literature quality evaluation method evaluates the risk bias of the included literature quality according to the Cochrane systematic evaluation manual, and if there are differences in the evaluation results, it will be discussed and resolved with the third researcher. The evaluation contents include random allocation method, blind method, allocation concealment, data integrity of research results, selective reporting, and other biases. The degree of risk bias, it can be divided into 3 levels: low risk, high risk, and unclear.\n[SUBTITLE] 3.6. Statistical analysis [SUBSECTION] The data was analyzed by Stata16 software, the weighted mean difference (WMD) was used for the measurement data, and the corresponding confidence interval (CI) of each effect was calculated by 95%. If P < .01, the difference was statistically significant. The heterogeneity of the included research was tested, and the corresponding effect model was selected according to the values of the Q test and I2 test. If P ≥ .05 and I2 ≤ 50%, the statistical heterogeneity among the studies is small, then the “fixed effect model” is adopted for the meta-analysis of the curative effect; if P < .05, I2 > 50%, it indicates that the heterogeneity between the studies is large. The “random effect model” was adopted to analyze the curative effect. In addition, the sources of heterogeneity were analyzed by subgroup analysis or sensitivity analysis. We use the funnel chart to detect publication bias, and Begg and Egger tests were used to determine that. When the funnel chart shows a symmetrical inverted triangle and the P values of Begg and Egger are >.05, we believe that there is no potential publication bias, otherwise, there may be publication bias.\nThe data was analyzed by Stata16 software, the weighted mean difference (WMD) was used for the measurement data, and the corresponding confidence interval (CI) of each effect was calculated by 95%. If P < .01, the difference was statistically significant. The heterogeneity of the included research was tested, and the corresponding effect model was selected according to the values of the Q test and I2 test. If P ≥ .05 and I2 ≤ 50%, the statistical heterogeneity among the studies is small, then the “fixed effect model” is adopted for the meta-analysis of the curative effect; if P < .05, I2 > 50%, it indicates that the heterogeneity between the studies is large. The “random effect model” was adopted to analyze the curative effect. In addition, the sources of heterogeneity were analyzed by subgroup analysis or sensitivity analysis. We use the funnel chart to detect publication bias, and Begg and Egger tests were used to determine that. When the funnel chart shows a symmetrical inverted triangle and the P values of Begg and Egger are >.05, we believe that there is no potential publication bias, otherwise, there may be publication bias.\n[SUBTITLE] 3.7. Ethical approval [SUBSECTION] Not applicable.\nThis paper is a kind of quantitative comprehensive evaluation based on literature data of multiple types independent research studies.\nNot applicable.\nThis paper is a kind of quantitative comprehensive evaluation based on literature data of multiple types independent research studies.", "We formulated inclusion criteria by the principles of PICOS, and only documents that meet all of the following criteria would be included. Population: clear diagnostic criteria of a stroke complicated with cognitive impairment; intervention: experimental group received routine treatment and acupuncture treatment without restriction on mode and course; comparison: control group received routine treatment; outcome indicators: mini-mental state examination scale (MMSE) (A standardized mental state examination tool for simple screening of temporal orientation, place orientation, immediate memory, attention and computation, delayed memory, language, and visual space),[17] Montreal cognitive assessment scale (MoCA) (An assessment tool for rapid screening of cognitive dysfunction in terms of orientation, short-term memory, executive function, language, abstraction, animal naming, attention, clock-drawing test),[17] Barthel index of activities of daily living (MBI) (A scale used to measure changes in independent living skills such as eating, dressing, continence, and mobility in older people before and after treatment),[18] Lowenstein assessment scale (LOTCA) (A standardized neuropsychological test that assesses orientation, perception, visual motor organization, and mental functioning).[19] Study type: Randomized controlled trials. The language was Chinese or English.", "Acupuncture therapy was included in the control group; the experimental group did not combine acupuncture therapy with conventional therapy; the course of treatment was <4 weeks; reviews, case reports, guidelines, or, comment; data was incomplete and could not be available.", "First of all, duplicate documents were eliminated. Two researchers excluded literature that did not meet the inclusion criteria by reading the title and abstract. The possible literature was read in full text to further judge whether it was included or not. After completion of the preliminary screening, any differences will be resolved through discussion among all authors.", "After numbering the final included documents, the data was extracted using the “data extraction Table” designed by Excel, and the extracted contents included: basic information: literature name, author, year of publication, etc. Research characteristics: intervention measures, sample size, course of treatment, outcome index, adverse reaction report, etc.", "The literature quality evaluation method evaluates the risk bias of the included literature quality according to the Cochrane systematic evaluation manual, and if there are differences in the evaluation results, it will be discussed and resolved with the third researcher. The evaluation contents include random allocation method, blind method, allocation concealment, data integrity of research results, selective reporting, and other biases. The degree of risk bias, it can be divided into 3 levels: low risk, high risk, and unclear.", "The data was analyzed by Stata16 software, the weighted mean difference (WMD) was used for the measurement data, and the corresponding confidence interval (CI) of each effect was calculated by 95%. If P < .01, the difference was statistically significant. The heterogeneity of the included research was tested, and the corresponding effect model was selected according to the values of the Q test and I2 test. If P ≥ .05 and I2 ≤ 50%, the statistical heterogeneity among the studies is small, then the “fixed effect model” is adopted for the meta-analysis of the curative effect; if P < .05, I2 > 50%, it indicates that the heterogeneity between the studies is large. The “random effect model” was adopted to analyze the curative effect. In addition, the sources of heterogeneity were analyzed by subgroup analysis or sensitivity analysis. We use the funnel chart to detect publication bias, and Begg and Egger tests were used to determine that. When the funnel chart shows a symmetrical inverted triangle and the P values of Begg and Egger are >.05, we believe that there is no potential publication bias, otherwise, there may be publication bias.", "Not applicable.\nThis paper is a kind of quantitative comprehensive evaluation based on literature data of multiple types independent research studies.", "According to the retrieval strategy, 1464 articles were retrieved, 273 repetitive articles were excluded, and 1191 articles were excluded after reading titles/abstracts manually. Re-screening and reading the remaining 100 full-text articles, and finally including 14 articles. The literature screening process and results are shown in Figure 1.\nFlow chart of literature screening.\n[SUBTITLE] 4.1. Basic characteristics of the included literature [SUBSECTION] The basic characteristics of the literature were included in 14 articles, with a total of 2402 patients. The basic characteristics of each study are shown in Table 1.\nThe characteristics of included studies.\nLOTCA = Lowenstein assessment scale, MBI = activities of daily living Barthel index, MMSE = mini-mental state scale, MOCA = Montreal cognitive assessment scale, rTMS = transcranial magnetic stimulations.\nThe basic characteristics of the literature were included in 14 articles, with a total of 2402 patients. The basic characteristics of each study are shown in Table 1.\nThe characteristics of included studies.\nLOTCA = Lowenstein assessment scale, MBI = activities of daily living Barthel index, MMSE = mini-mental state scale, MOCA = Montreal cognitive assessment scale, rTMS = transcranial magnetic stimulations.\n[SUBTITLE] 4.2. Quality evaluation [SUBSECTION] The risk bias was assessed according to the Cochrane guideline. Ten articles[20–22,24,25,28,29,31–33] were grouped by the random number table method, 1 article[27] used the computer random grouping method, and the other 3 articles[23,26,30] did not mention the specific distribution scheme. In the aspect of data collection and evaluation, 1 article[27] mentioned that the implementation of the blind method was single-blind, the other 11 articles did not mention the blind method. In terms of other biases, the diagnostic criteria of 8 articles[20,21,25,27,29–31,33] are not specified, which may affect the authenticity of the study, so they are evaluated as high risk. The quality evaluation of the included literature is shown in Table 2.\nQuality assessment of included studies.\nThe risk bias was assessed according to the Cochrane guideline. Ten articles[20–22,24,25,28,29,31–33] were grouped by the random number table method, 1 article[27] used the computer random grouping method, and the other 3 articles[23,26,30] did not mention the specific distribution scheme. In the aspect of data collection and evaluation, 1 article[27] mentioned that the implementation of the blind method was single-blind, the other 11 articles did not mention the blind method. In terms of other biases, the diagnostic criteria of 8 articles[20,21,25,27,29–31,33] are not specified, which may affect the authenticity of the study, so they are evaluated as high risk. The quality evaluation of the included literature is shown in Table 2.\nQuality assessment of included studies.\n[SUBTITLE] 4.3. Meta-analysis results [SUBSECTION] [SUBTITLE] 4.3.1.1. MMSE score. [SUBSECTION] Nine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved.\nMMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale.\nNine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved.\nMMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale.\n[SUBTITLE] 4.3.1.2. MoCA score [SUBSECTION] Eight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved.\nMoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale.\nEight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved.\nMoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale.\n[SUBTITLE] 4.3.1.3. MBI score [SUBSECTION] Four studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc.\nMBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index.\nFour studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc.\nMBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index.\n[SUBTITLE] 4.3.1.4. LOTCA score [SUBSECTION] Two studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly.\nLOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale.\nTwo studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly.\nLOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale.\n[SUBTITLE] 4.3.1.1. MMSE score. [SUBSECTION] Nine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved.\nMMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale.\nNine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved.\nMMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale.\n[SUBTITLE] 4.3.1.2. MoCA score [SUBSECTION] Eight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved.\nMoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale.\nEight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved.\nMoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale.\n[SUBTITLE] 4.3.1.3. MBI score [SUBSECTION] Four studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc.\nMBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index.\nFour studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc.\nMBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index.\n[SUBTITLE] 4.3.1.4. LOTCA score [SUBSECTION] Two studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly.\nLOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale.\nTwo studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly.\nLOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale.\n[SUBTITLE] 4.4. Sensitivity analysis [SUBSECTION] Sensitivity analysis was conducted by excluding 1 study at a time. We find that the results were robust and did not change the conclusions due to the exclusion of any study.\nSensitivity analysis was conducted by excluding 1 study at a time. We find that the results were robust and did not change the conclusions due to the exclusion of any study.\n[SUBTITLE] 4.5. Publication bias analysis [SUBSECTION] Because the number of articles included in the MBI score and LOTCA score index groups was <5, it is not suitable for funnel chart analysis. The MMSE score and MoCA score index groups were evaluated for the existence of publication bias. The results showed that although individual subjects deviate from the CI of the funnel diagram, they are roughly symmetrical. In addition, the P values of Begg and Egger tests of all indicators were >.05, indicating that there was no potential publication bias.\nBecause the number of articles included in the MBI score and LOTCA score index groups was <5, it is not suitable for funnel chart analysis. The MMSE score and MoCA score index groups were evaluated for the existence of publication bias. The results showed that although individual subjects deviate from the CI of the funnel diagram, they are roughly symmetrical. In addition, the P values of Begg and Egger tests of all indicators were >.05, indicating that there was no potential publication bias.", "The basic characteristics of the literature were included in 14 articles, with a total of 2402 patients. The basic characteristics of each study are shown in Table 1.\nThe characteristics of included studies.\nLOTCA = Lowenstein assessment scale, MBI = activities of daily living Barthel index, MMSE = mini-mental state scale, MOCA = Montreal cognitive assessment scale, rTMS = transcranial magnetic stimulations.", "The risk bias was assessed according to the Cochrane guideline. Ten articles[20–22,24,25,28,29,31–33] were grouped by the random number table method, 1 article[27] used the computer random grouping method, and the other 3 articles[23,26,30] did not mention the specific distribution scheme. In the aspect of data collection and evaluation, 1 article[27] mentioned that the implementation of the blind method was single-blind, the other 11 articles did not mention the blind method. In terms of other biases, the diagnostic criteria of 8 articles[20,21,25,27,29–31,33] are not specified, which may affect the authenticity of the study, so they are evaluated as high risk. The quality evaluation of the included literature is shown in Table 2.\nQuality assessment of included studies.", "[SUBTITLE] 4.3.1.1. MMSE score. [SUBSECTION] Nine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved.\nMMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale.\nNine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved.\nMMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale.\n[SUBTITLE] 4.3.1.2. MoCA score [SUBSECTION] Eight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved.\nMoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale.\nEight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved.\nMoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale.\n[SUBTITLE] 4.3.1.3. MBI score [SUBSECTION] Four studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc.\nMBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index.\nFour studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc.\nMBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index.\n[SUBTITLE] 4.3.1.4. LOTCA score [SUBSECTION] Two studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly.\nLOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale.\nTwo studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly.\nLOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale.", "Nine studies[22–29,32] used the MMSE score as the outcome index, and there was statistical heterogeneity among the results (I2 = 80.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the improvement of MMSE score in the treatment group was better than that in the control group, and the difference was statistically significant (WMD = 3.23, 95% CI: 1.89–4.56, P < .01) (Fig. 2). It indicates that the cognitive dysfunction of stroke patients has been significantly improved.\nMMSE score of patients with stroke treated with acupuncture. MMSE = mini-mental state scale.", "Eight studies[21–27,33] used the MoCA score as the outcome index, and there was statistical heterogeneity among the results (I2 = 95.8%, P < .001), so the random effect model was used for meta-analysis. The results showed that the intervention group was superior to the control group in improving the MoCA score, and the difference was statistically significant (WMD = 3.41, 95% CI: 0.93–5.89, P < .01) (Fig. 3). It indicates that the cognitive function of patients with mild cognitive impairment has been significantly improved.\nMoCA score of patients with stroke treated with acupuncture. MoCA = Montreal cognitive assessment scale.", "Four studies[20,23,30,31] used the MBI score as the outcome index. Heterogeneity analysis showed that there was a large heterogeneity among the studies (I2 > 50%), so the random effect model was used for Meta-analysis. The results of the combined effect of clinical effective rate showed that there was a significant difference in MBI score between the control group and the treatment group, and the effect value was combined with WMD = 4.59 (95% CI: 1.43–7.75, P < .01) (Fig. 4). It shows that the self-care ability of stroke patients is improved obviously, which include Feeding, bathing, grooming, dressing, controlling urine and feces, going to the toilet, transferring beds and chairs, etc.\nMBI score of patients with stroke treated with acupuncture. MBI = activities of daily living Barthel index.", "Two studies[24,31] used the LOTCA score as the outcome index, and there was no statistical heterogeneity among the studies (I2 = 0.0%, P = .360), so the fixed effect model was used for meta-analysis. The combined effect value of WMD was 8.60 (95% CI: 6.32–10.89, P < .01). It shows that the clinical efficacy of scalp acupuncture in the treatment of cognitive impairment after stroke is better than that of the control group (Fig. 5). It showed that the patient’s orientation, perception, visual movement, thinking, and other aspects of the situation improved significantly.\nLOTCA score of patients with stroke treated with acupuncture. LOTCA = Lowenstein assessment scale.", "Sensitivity analysis was conducted by excluding 1 study at a time. We find that the results were robust and did not change the conclusions due to the exclusion of any study.", "Because the number of articles included in the MBI score and LOTCA score index groups was <5, it is not suitable for funnel chart analysis. The MMSE score and MoCA score index groups were evaluated for the existence of publication bias. The results showed that although individual subjects deviate from the CI of the funnel diagram, they are roughly symmetrical. In addition, the P values of Begg and Egger tests of all indicators were >.05, indicating that there was no potential publication bias.", "The 14 studies included in this study included 2402 cases. Meta-analysis results of 4 observational indicators showed that acupuncture combined with other therapies had significant advantages in treating PSCI, improving MMSE score, MoCA score, MBI index, and LOTCA scale compared with other therapies. MMSE score (WMD = 3.23, 95% CI: 1.89–4.56, P < .01), MoCA score (WMD = 3.41, 95% CI: 0.93–5.89, P < .01), MBI score (WMD = 4.59, 95% CI: 1.43–7.75, P < .01) and LOTCA score (WMD = 8.60, 95% CI: 6.32–10.89, P < .01). It can be concluded that acupuncture has a good effect on improving cognitive function and promoting the rehabilitation of PSCI patients, which provides a theoretical basis for the therapeutic advantage of acupuncture combined with conventional therapy in treating PSCI.\nTraditional acupuncture has a long history in the treatment of stroke, and traditional Chinese medicine has a comprehensive understanding of it.[19] Stroke is also called “dementia” in the Internal Classic of Huangdi. So far, it is considered that “wind, fire, phlegm, qi, blood stasis, deficiency” is its etiology and pathogenesis. [34] Cognitive impairment often occurs in stroke, vascular diseases, and other diseases. The therapeutic effect of western medicine is limited and the side effects are obvious. Some studies have shown that cognitive impairment belongs to neurodegenerative disease, which is due to the functional degradation of central nervous cells in patients, especially in the cerebral cortex and hippocampus.[35] This leads to cognitive impairment in patients. Personality change, decreased living ability, memory loss and slow response are the common clinical manifestations. After the occurrence of PSCI, the quality of life of patients decreased sharply, and looking for effective ways to improve patients’ cognitive function is of great significance to improve patients’ cognitive function and promote their prognosis.[36]\nIn recent years, acupuncture has been widely used in the clinical treatment of PSCI. Many studies have shown that acupuncture may improve the cognitive function of PSCI patients by regulating enzyme activity, improving synaptic plasticity, and inhibiting the expression of inflammatory cytokines.[37–39] Some studies have found that acupuncture can significantly reduce the incidence of stroke sequelae, has a positive effect on the treatment of cognitive impairment, and reveals that acupuncture can regulate hemorheology and neuron electrophysiology and protect neurons.[40–42] Acupuncture has the characteristics of economy, safety, simplicity, and so on, so it is more and more widely used in clinical practice.\nMore and more studies have demonstrated the neuroprotective activities of acupuncture therapy. Most of these studies have proposed that acupuncture exerts its therapeutic effects via regulating various signaling pathways. [43] Such as, some studies have confirmed that acupuncture exerts several beneficial effects on the central nervous system via the activation of brain-derived neurotrophic factor and its downstream signaling pathway.[44–46] Ryffel et al suggested that protection of dopamine neuronal degeneration by acupuncture was due to enhancement of cyclophilin A (CypA) levels. Based on recently published studies, CypA is ubiquitously expressed in the brain[46] and is predominantly localized in neurons.[47] Another study shows that acupuncture possesses the ability to ameliorate mutant α-syn-induced motor abnormalities. This ability may be due to the that EA enhances both anti-inflammatory and antioxidant activities and suppresses aberrant glial activation in the diseased sites of brains.[48]\nIn a word, acupuncture treatments in animal experiments have generated valuable mechanistic insights into the pathology of Parkinson and have provided evidence that acupuncture therapy is neuroprotective and can increase various neuroprotective agents such as brain-derived neurotrophic factor, glial cell- derived neurotrophic factor, and cyclophilin A, enhancing both anti-inflammatory and antioxidant activities.\nTraditional Chinese medicine has developed a relatively rich way, such as scalp acupuncture, body acupuncture, electroacupuncture, moxibustion, and acupuncture combined with other therapies and so on. The above methods have achieved remarkable results in improving patients’ cognition and activities of daily life.\nIt is worth noting that this study has some limitations. First of all, due to the low overall quality of the literature on acupuncture treatment of cognitive impairment after stroke, the final included literature is of low quality; in addition, there is little data related to adverse reactions and it is difficult to evaluate. Thirdly, most of the included subjects were aged between 45 and 70, while other age groups were underrepresented. Certain conditions that may be encountered in these age groups may lead to bias in the interpretation of the results. furthermore, none of the included studies considered the effect of gender. After that, further research in this area is needed. Due to the small amount of literature related to different types of acupuncture therapy, we did not conduct further subgroup analysis. Finally, it is suggested that a more scientific and rigorous research scheme should be designed in the future. It is hopeful to carry out the study through large samples and high-quality randomized controlled trials, to explore the differences between different types of acupuncture, and to provide a more scientific clinical basis for the popularization and application of acupuncture treatment for PSCI.\nIn conclusion, available evidence suggests that acupuncture combined with conventional therapy has a potential clinical advantage over conventional therapy in improving cognitive impairment after stroke. However, the differences between different types of acupuncture need to be clarified in more high-quality studies for further comparison.", "Methodology: Kang Xiong.\nSoftware: Mei-Ling Zhang.\nSupervision: Bin Wang.\nWriting – original draft: Zhen-Zhi Wang, Zhen Sun.\nWriting – review & editing: Zhen-Zhi Wang, Zhen Sun." ]
[ "intro", "methods", null, null, null, null, null, null, null, null, null, "results", null, null, "results", null, null, null, null, null, null, "discussion", null ]
[ "acupuncture", "cognitive impairment", "meta-analysis", "stroke", "systematic review" ]
Effects of percutaneous neuromodulation in neuromusculoskeletal pathologies: A systematic review.
36254060
Percutaneous neuromodulation (PNM) consists in using electrical stimulation on a peripheral nerve by using a needle as an electrode in order to lessen the pain and restore both neuromuscular and nervous system functions. The aims of the present study were to evaluate the current scientific evidence of the effects of PNM on pain and physical capabilities in neuromusculoskeletal injuries.
BACKGROUND
Data sources: There was used the PRISMA protocol. In order to do the literature research, there were used the PubMed, Cochrane, Scopus, and Web of Science databases. Study selection or eligibility criteria: There were also included experimental clinical trials published between 2010 and nowadays, tested on humans, which feature treatment based on needles with electrical stimulation in order to treat neuromusculoskeletal injuries. Study appraisal and synthesis methods: A quality assessment was performed according to the PEDro scale and reviewed the impact factor and quartile of the journal.
METHODS
The treatment resulted in significant improvement in terms of pain intensity, pressure pain threshold, balance, muscular endurance, functionality/disability, subjective improvement, function of the descending pain modulatory system, and intake of drugs. Limitations: the lack of previous research studies on the subject and the lack of data on opioid intake in the selected studies.
RESULTS
Treatment based on PNM may be an alternative when treating injuries in soft tissues without significant side effects. However, there are few articles investigating the effects of PNM so more evidence is needed to draw solid conclusions.
CONCLUSION
[ "Analgesics, Opioid", "Humans", "Pain", "Pain Measurement" ]
9575779
1. Introduction
Neuromodulation is a quality of the nervous system that regulates or modifies electrical impulses, by enhancing or inhibiting them.[1] Even though its mechanism of action is yet to be fully determined, it is known that it lies on the ascending and descending pathways and the supraspinal regions of the central nervous system. The chronic effect of said neurophysiological process may cause central nervous system plasticity, with lasting clinical effects.[2,3] Neuromodulation is part of the treatment for multiple pathologies with few side effects.[4] Nowadays, there are described the following neuromodulation procedures, classified according to the area of application[5,6]: at the brain level, cortical stimulation[7] and deep brain stimulation,[8] at the spinal level, stimulation of the dorsal column of the spinal cord,[9] nerve root stimulation[10] and dorsal root ganglion stimulation[11] and at the level of the peripheral nervous system, the stimulation of the peripheral nerve,[12] the stimulation of the receptive field of the peripheral nerve[13] and the stimulation of the muscular motor point.[5] Peripheral nerve and muscle level neuromodulation (percutaneous neuromodulation [PNM]) consists in percutaneous electrical stimulation of a peripheral nerve, along its pathway or in a muscle, through a puncture needle with electric current of low or medium frequency. Even though the mechanism of action is not fully known, the PNM spans the ascending and descending pathways, as well as the supraspinal regions of the central nervous system, regulating or modifying the electrical impulses transmitted through said pathways, and thus inhibiting or exciting them.[3,14] The main objective of the application of PNM is to provoke a motor and/or sensitive response that eases pain and restores the usual functioning of the nervous system. Thus, on the one hand, it may relieve central sensitization and hyperexcitability (hyperalgesia or allodynia) related chronic pain and neuropathic pain, and, on the other hand, it may improve the neuromuscular function, muscle recruitment patterns and motor control.[5,14,15] In this line, recent studies suggests PNM could relieve central sensitization by improving conditioned pain modulation, reducing motor-evoked potential and increasing intracortical inhibition.[16–18] Additionally, the effects of this technique are being investigated for the improvement of some physical qualities such as elasticity, strength, balance and muscular endurance.[15,19–25] An injury does not only produce changes in an anatomical level; it also may unfold changes within the nervous system, such as increase in the motoneuron excitability, decrease in the sense of position, movement and strength, decrease in the acetylcholine receptors, decrease in the cholinergic/non-cholinergic muscle stimulation, decrease in voluntary activation, decrease in corticospinal excitability, and central sensitization.[26] Central sensitization, on the other hand, represents an intensification when it comes to the activity of circuits and neurons in the nociceptive pathways, due to the enhancement of the excitability of the membrane and the synapses. In patients with central sensitization, the cortex suffers an aberrant reorganization, and the pain increases drastically. In addition, the perception of any other sensory experience reaches a higher amplitude, duration, and spatial extent, which results in a reduced excitation-inhibition balance.[16] Therefore, considering the changes that take place in the nervous system after an injury, and the mechanism of action of PNM, some authors used said technique to treat soft tissue injuries. The objective of this study was to evaluate the effects of PNM on pain and physical capabilities when it comes to neuromusculoskeletal injuries.
null
null
3. Results
[SUBTITLE] 3.1. Selection of articles [SUBSECTION] Through the literature search in said four databases, we found 176 potential articles for this review by all authors (I.F.-M., J.J.R.-Á., R.M.-L., and E.S.R-L). Among those, we selected 24 and narrowed it down to 15,[14,16–25,30–33] since 9 were duplicates. After reading the 15 articles, we did not discard a single one and we used them to make the bibliographical review (Fig. 1) Flowchart. Through the literature search in said four databases, we found 176 potential articles for this review by all authors (I.F.-M., J.J.R.-Á., R.M.-L., and E.S.R-L). Among those, we selected 24 and narrowed it down to 15,[14,16–25,30–33] since 9 were duplicates. After reading the 15 articles, we did not discard a single one and we used them to make the bibliographical review (Fig. 1) Flowchart. [SUBTITLE] 3.2. Types of study [SUBSECTION] Among the selected 15 articles, 14 were randomized studies[16–25,30–33] and the last one was a prospective observational study.[14] Among the selected 15 articles, 14 were randomized studies[16–25,30–33] and the last one was a prospective observational study.[14] [SUBTITLE] 3.3. Methodological quality of the selected articles [SUBSECTION] The articles obtained an average score of 7.06 ± 1.84 on the PEDro scale[29] (Table 1). Methodological quality according to PEDro scale. Eligibility criteria were specified. Subjects were randomly allocated to groups (in a crossover study, subjects were randomly allocated an order in which treatments were received). Allocation was concealed. The groups were similar at baseline regarding the most important prognostic indicators. There was blinding of all subjects. There was blinding of all therapists who administered the therapy. There was blinding of all assessors who measured at least one key outcome. Measures of at least one key outcome were obtained from >85% of the subjects initially allocated to groups. All subjects for whom outcome measures were available received the treatment or control condition as allocated or, where this was not the case, data for at least one key outcome was analyzed by “intention to treat”. The results of between-group statistical comparisons are reported for at least one key outcome. The study provides both point measures and measures of variability for at least one key outcome. The articles obtained an average score of 7.06 ± 1.84 on the PEDro scale[29] (Table 1). Methodological quality according to PEDro scale. Eligibility criteria were specified. Subjects were randomly allocated to groups (in a crossover study, subjects were randomly allocated an order in which treatments were received). Allocation was concealed. The groups were similar at baseline regarding the most important prognostic indicators. There was blinding of all subjects. There was blinding of all therapists who administered the therapy. There was blinding of all assessors who measured at least one key outcome. Measures of at least one key outcome were obtained from >85% of the subjects initially allocated to groups. All subjects for whom outcome measures were available received the treatment or control condition as allocated or, where this was not the case, data for at least one key outcome was analyzed by “intention to treat”. The results of between-group statistical comparisons are reported for at least one key outcome. The study provides both point measures and measures of variability for at least one key outcome. [SUBTITLE] 3.4. Pain intensity [SUBSECTION] All articles that analyzed the effect of PNM on pain found significant improvements. The studies made by Hadizadeh et al[23] in patients with trigger points in upper trapezius, Botelho et al[16] in patients with myofascial pain syndrome, da Graca-Tarragó et al 2016[17] in patients with knee osteoarthritis, and Raphael et al[33] in patients with chronic pain and hyperalgesia obtained a significant improvement when it came to pain intensity (P ≤ .048; P < .001; P = .001, and P < .0005, respectively). Likewise, García-Bermejo et al 2020 found a significant improvement on PNM based treatment when treating unilateral anterior knee pain, regardless using it in a contralateral or homolateral manner (P = .01 in both groups).[22] On the other hand, Rossi et al[14] observed a significant improvement in patients with neuropathic pain (P < .001), which began 60 minutes post-treatment and lasted for 6 months. Some studies compared the effects of PNM to other techniques. Pérez-Palomares et al found no significant difference (P = .94) between PNM and DN in patients with chronic low back pain when it came to mid-term effect.[32] Nonetheless, León-Hernández et al did find significant differences when combining PNM with DN against only using DN on patients with chronic neck pain (P < .05), with a significant decrease in the post-needling soreness intensity 72 hours after using PNM (P < .05).[24] Dunning et al[30,31] observed a significant decrease in pain thanks to the PNM treatment, compared to manual therapy, in patients with knee osteoarthritis and plantar fasciitis. Said decrease lasted for 3 months. Sumen et al[25] compared PNM treatment to low-level laser therapy in patients with an active trigger point in upper trapezius and diagnosed with myofascial pain syndrome. Both treatments improved symptomatology (P = .016 and P = .001, respectively).[25] Finally, da Graca-Tarragó et al 2019 observed that patients with knee osteoarthritis that received transcranial direct currents and PNM, regardless of being simulated or not, showed a significant improvement (P < .03), with a higher reduction of the pain intensity when combining both techniques.[18] All articles that analyzed the effect of PNM on pain found significant improvements. The studies made by Hadizadeh et al[23] in patients with trigger points in upper trapezius, Botelho et al[16] in patients with myofascial pain syndrome, da Graca-Tarragó et al 2016[17] in patients with knee osteoarthritis, and Raphael et al[33] in patients with chronic pain and hyperalgesia obtained a significant improvement when it came to pain intensity (P ≤ .048; P < .001; P = .001, and P < .0005, respectively). Likewise, García-Bermejo et al 2020 found a significant improvement on PNM based treatment when treating unilateral anterior knee pain, regardless using it in a contralateral or homolateral manner (P = .01 in both groups).[22] On the other hand, Rossi et al[14] observed a significant improvement in patients with neuropathic pain (P < .001), which began 60 minutes post-treatment and lasted for 6 months. Some studies compared the effects of PNM to other techniques. Pérez-Palomares et al found no significant difference (P = .94) between PNM and DN in patients with chronic low back pain when it came to mid-term effect.[32] Nonetheless, León-Hernández et al did find significant differences when combining PNM with DN against only using DN on patients with chronic neck pain (P < .05), with a significant decrease in the post-needling soreness intensity 72 hours after using PNM (P < .05).[24] Dunning et al[30,31] observed a significant decrease in pain thanks to the PNM treatment, compared to manual therapy, in patients with knee osteoarthritis and plantar fasciitis. Said decrease lasted for 3 months. Sumen et al[25] compared PNM treatment to low-level laser therapy in patients with an active trigger point in upper trapezius and diagnosed with myofascial pain syndrome. Both treatments improved symptomatology (P = .016 and P = .001, respectively).[25] Finally, da Graca-Tarragó et al 2019 observed that patients with knee osteoarthritis that received transcranial direct currents and PNM, regardless of being simulated or not, showed a significant improvement (P < .03), with a higher reduction of the pain intensity when combining both techniques.[18] [SUBTITLE] 3.5. Pressure pain threshold [SUBSECTION] All articles found a significant improvement in the pressure pain threshold when using PNM or DN, with no significant differences between them.[17,18,24,25,32] All articles found a significant improvement in the pressure pain threshold when using PNM or DN, with no significant differences between them.[17,18,24,25,32] [SUBTITLE] 3.6. Range of motion [SUBSECTION] There was a significant improvement in three out of six articles that analyzed this parameter. In patients with trigger points in upper trapezius (P = .048),[23] patients with bilateral reduced hamstring syndrome (P < .01),[21] and patients with anterior knee pain (P = .001).[22] Nonetheless, de la Cruz et al 2019 found no significant improvement in ballet dancers,[19,21] nor did Sumen et al in patients with trigger points in upper trapezius.[25] There was a significant improvement in three out of six articles that analyzed this parameter. In patients with trigger points in upper trapezius (P = .048),[23] patients with bilateral reduced hamstring syndrome (P < .01),[21] and patients with anterior knee pain (P = .001).[22] Nonetheless, de la Cruz et al 2019 found no significant improvement in ballet dancers,[19,21] nor did Sumen et al in patients with trigger points in upper trapezius.[25] [SUBTITLE] 3.7. Balance and muscle endurance [SUBSECTION] Two studies evaluated balance and muscle endurance and observed a significant improvement in ballet dancers (P < .001 in both cases).[19,21] Two studies evaluated balance and muscle endurance and observed a significant improvement in ballet dancers (P < .001 in both cases).[19,21] [SUBTITLE] 3.8. Muscle contractile properties [SUBSECTION] Only one study evaluated the effects of PNM on muscle contractile properties through tensiomyography and found no significant differences (P > .05).[20] Only one study evaluated the effects of PNM on muscle contractile properties through tensiomyography and found no significant differences (P > .05).[20] [SUBTITLE] 3.9. Functionality/disability [SUBSECTION] Five studies analyzed the effects of PNM on functionality/disability[18,22,24,30,31] and all of them found significant improvements. In patients with knee osteoarthritis (P < .001 and P = .03)[18,31] in patients with plantar fasciitis (P < .001),[30] in patients with anterior knee pain (P = .001),[22] and in patients with neck pain, if they combined the treatment with DN.[24] Five studies analyzed the effects of PNM on functionality/disability[18,22,24,30,31] and all of them found significant improvements. In patients with knee osteoarthritis (P < .001 and P = .03)[18,31] in patients with plantar fasciitis (P < .001),[30] in patients with anterior knee pain (P = .001),[22] and in patients with neck pain, if they combined the treatment with DN.[24] [SUBTITLE] 3.10. Quality of life [SUBSECTION] The only study that analyzed the quality of life compared PNM and DN, and found no significant differences.[32] The only study that analyzed the quality of life compared PNM and DN, and found no significant differences.[32] [SUBTITLE] 3.11. Intake of drugs [SUBSECTION] The 5 studies that analyzed intake of drugs reported that there was a reduction in the use of analgesics and non-steroidal anti-inflammatory drugs after PNM based treatment. In chronic neuropathic pain,[14] in plantar fasciitis (P = .001),[30] in knee osteoarthritis (P = .023 and P < .019)[18,31] and in myofascial pain syndrome.[16] The 5 studies that analyzed intake of drugs reported that there was a reduction in the use of analgesics and non-steroidal anti-inflammatory drugs after PNM based treatment. In chronic neuropathic pain,[14] in plantar fasciitis (P = .001),[30] in knee osteoarthritis (P = .023 and P < .019)[18,31] and in myofascial pain syndrome.[16] [SUBTITLE] 3.12. Sleep quality [SUBSECTION] Two studies evaluated the sleep quality. In the myofascial pain syndrome, Botelho et al found an improvement in said parameter (P = .04).[16] Nonetheless, Pérez-Palomares et al[32] found no major differences when it came to chronic low back pain (P = .68). Two studies evaluated the sleep quality. In the myofascial pain syndrome, Botelho et al found an improvement in said parameter (P = .04).[16] Nonetheless, Pérez-Palomares et al[32] found no major differences when it came to chronic low back pain (P = .68). [SUBTITLE] 3.13. Subjective improvement [SUBSECTION] The three articles that analyze subjective improvement after PNM found a significant improvement in patients with neuropathic pain,[14] knee osteoarthritis,[31] and plantar fasciitis[30] (P < .001 in all three cases). The three articles that analyze subjective improvement after PNM found a significant improvement in patients with neuropathic pain,[14] knee osteoarthritis,[31] and plantar fasciitis[30] (P < .001 in all three cases). [SUBTITLE] 3.14. Function of the descending pain modulatory system [SUBSECTION] Three articles analyze the effect of PNM on the function of the descending pain modulatory system, obtaining significant improvements. Those three articles made the analysis based on myofascial pain (P = .01)[16] and osteoarthritis (P = .01).[17,18] Three articles analyze the effect of PNM on the function of the descending pain modulatory system, obtaining significant improvements. Those three articles made the analysis based on myofascial pain (P = .01)[16] and osteoarthritis (P = .01).[17,18] [SUBTITLE] 3.15. Cortical excitability [SUBSECTION] Two articles analyzed the effects of PNM on cortical excitability and found a significant decrease in the motor evoked potential in myofascial pain (P = .02)[16] and osteoarthritis (P = .03).[17] In terms of the cortical silent period, both studies agree on the significant increase in said parameter (P = .005 and P = .001, respectively). Finally, none of those studies found significant differences when it came to short intracortical inhibition. Two articles analyzed the effects of PNM on cortical excitability and found a significant decrease in the motor evoked potential in myofascial pain (P = .02)[16] and osteoarthritis (P = .03).[17] In terms of the cortical silent period, both studies agree on the significant increase in said parameter (P = .005 and P = .001, respectively). Finally, none of those studies found significant differences when it came to short intracortical inhibition. [SUBTITLE] 3.16. Brain derived neurotrophic factor [SUBSECTION] Three articles studied the effect of PNM on BDNF, with conflicting results. The BDNF is a protein associated with nerve growth factor who is secreted by astrocytes and glial cells and produces spinal cord neurons sensitization, facilitates the activation of N-Methyl-d-asparyaye and increases the excitability of gamma-aminobutyric acid-ergic neurons.[16] Botelho et al obtained significantly higher results of BDNF (P < .01) than the placebo when it came to myofascial pain.[16] Nonetheless, da Graca-Tarragó et al 2019 found no effect on BDNF when treating knee osteoarthritis.[18] On the other hand, in the study by da Graca-Tarragó et al 2016, they observed that BDNF had no correlation with the motor evoked potential, but it had a negative and marginal correlation (P = .05) with the pressure pain threshold.[17] Three articles studied the effect of PNM on BDNF, with conflicting results. The BDNF is a protein associated with nerve growth factor who is secreted by astrocytes and glial cells and produces spinal cord neurons sensitization, facilitates the activation of N-Methyl-d-asparyaye and increases the excitability of gamma-aminobutyric acid-ergic neurons.[16] Botelho et al obtained significantly higher results of BDNF (P < .01) than the placebo when it came to myofascial pain.[16] Nonetheless, da Graca-Tarragó et al 2019 found no effect on BDNF when treating knee osteoarthritis.[18] On the other hand, in the study by da Graca-Tarragó et al 2016, they observed that BDNF had no correlation with the motor evoked potential, but it had a negative and marginal correlation (P = .05) with the pressure pain threshold.[17] [SUBTITLE] 3.17. Adverse effects [SUBSECTION] Nine articles informed of the possible adverse effects of the PNM; two of them found no such effects after applying treatment[19,33] and three of them had no unspecified significant, severe or moderate adverse effects.[16–18] In three articles, patients reported frequent post-needling soreness and/or hematomas,[14,30,31] and two studies reported occasional somnolence, headache, and nausea.[30,31] All articles that showed there were adverse effects after treatment coincide that said effects disappeared spontaneously within 1 to 4 days. Nine articles informed of the possible adverse effects of the PNM; two of them found no such effects after applying treatment[19,33] and three of them had no unspecified significant, severe or moderate adverse effects.[16–18] In three articles, patients reported frequent post-needling soreness and/or hematomas,[14,30,31] and two studies reported occasional somnolence, headache, and nausea.[30,31] All articles that showed there were adverse effects after treatment coincide that said effects disappeared spontaneously within 1 to 4 days.
null
null
[ "2. Methodology", "2.1. Design", "2.2. Identification and selection of articles", "2.3. Inclusion criteria", "2.4. Exclusion criteria", "2.5. Evaluation of the characteristics of the articles", "3.1. Selection of articles", "3.2. Types of study", "3.3. Methodological quality of the selected articles", "3.4. Pain intensity", "3.5. Pressure pain threshold", "3.6. Range of motion", "3.7. Balance and muscle endurance", "3.8. Muscle contractile properties", "3.9. Functionality/disability", "3.10. Quality of life", "3.11. Intake of drugs", "3.12. Sleep quality", "3.13. Subjective improvement", "3.14. Function of the descending pain modulatory system", "3.15. Cortical excitability", "3.16. Brain derived neurotrophic factor", "3.17. Adverse effects", "5. Limits of the study", "6. Conclusion", "Author contributions" ]
[ "[SUBTITLE] 2.1. Design [SUBSECTION] Systematic review on the effects of PNM. There was used the PRISMA protocol[27] and the research question was stated according to the PICO strategy.[28] This article does not contain any studies with human participants or animals performed by any of the authors. The medical ethics committee approval is not required for this type of study.\nSystematic review on the effects of PNM. There was used the PRISMA protocol[27] and the research question was stated according to the PICO strategy.[28] This article does not contain any studies with human participants or animals performed by any of the authors. The medical ethics committee approval is not required for this type of study.\n[SUBTITLE] 2.2. Identification and selection of articles [SUBSECTION] The literature research was performed between April and June 2020. In order to do so, there were used the PubMed, Cochrane, Scopus, and Web of Science databases. The searching phrase was created by combining keywords and Medical Subject Headings that defined the use of electrically stimulated needles and treatments or therapies were not wanted in the search, by using them with the Boolean operators OR and NOT. (“percutaneous electric nerve stimulation” OR “PNM therapy” OR “PNM therapies” OR “percutaneous electrical neuromodulation”) OR (“electrical dry needling [DN]”) OR (“intramuscular electrical stimulation”) NOT (“spinal cord stimulation”) NOT (“transcutaneous electric nerve stimulation”) NOT (“transcranial direct current stimulation [tDCS]”) NOT (“implanted” OR “surgery” OR “surgical”). The search was limited to articles published between 2010 and 2020, both in English and Spanish. The first selection of articles was screened by removing those that were not related to the topic. After fully reading the articles, a second screening was performed. Finally, the selected articles had to comply with some inclusion and exclusion criteria:\nThe literature research was performed between April and June 2020. In order to do so, there were used the PubMed, Cochrane, Scopus, and Web of Science databases. The searching phrase was created by combining keywords and Medical Subject Headings that defined the use of electrically stimulated needles and treatments or therapies were not wanted in the search, by using them with the Boolean operators OR and NOT. (“percutaneous electric nerve stimulation” OR “PNM therapy” OR “PNM therapies” OR “percutaneous electrical neuromodulation”) OR (“electrical dry needling [DN]”) OR (“intramuscular electrical stimulation”) NOT (“spinal cord stimulation”) NOT (“transcutaneous electric nerve stimulation”) NOT (“transcranial direct current stimulation [tDCS]”) NOT (“implanted” OR “surgery” OR “surgical”). The search was limited to articles published between 2010 and 2020, both in English and Spanish. The first selection of articles was screened by removing those that were not related to the topic. After fully reading the articles, a second screening was performed. Finally, the selected articles had to comply with some inclusion and exclusion criteria:\n[SUBTITLE] 2.3. Inclusion criteria [SUBSECTION] Clinical trial tested on humans; and at least one of the experimental groups should receive treatment based on DN or acupuncture with electrical stimulation.\nClinical trial tested on humans; and at least one of the experimental groups should receive treatment based on DN or acupuncture with electrical stimulation.\n[SUBTITLE] 2.4. Exclusion criteria [SUBSECTION] Use of the PNM in non-neuromusculoskeletal pathologies; treatments based on surgery of electrostimulation devices; not using needles as a conducting electrode; treatment based on tDCS; treatments based on Chinese traditional medicine, which treats another region, far from the region in pain and with no direct relationship between the region receiving the treatment and the pathological region: auriculotherapy, digit puncture, acupuncture…\nUse of the PNM in non-neuromusculoskeletal pathologies; treatments based on surgery of electrostimulation devices; not using needles as a conducting electrode; treatment based on tDCS; treatments based on Chinese traditional medicine, which treats another region, far from the region in pain and with no direct relationship between the region receiving the treatment and the pathological region: auriculotherapy, digit puncture, acupuncture…\n[SUBTITLE] 2.5. Evaluation of the characteristics of the articles [SUBSECTION] A quality assessment was performed by two authors (I.F.-M., J.J.R.-Á.) according to the PEDro scale[29] and reviewed the impact factor and quartile of the journal.\nThe independent variables of the study were the following: target tissue; treatment time; number of sessions; current type; current frequency; pulse width, and current intensity. On the other hand, the dependent variables were: pain intensity; range of motion (ROM); maximal isometric strength; balance; muscular endurance; muscle contractile properties; functionality/disability; quality of life; intake of drugs; sleep quality; subjective improvement; adverse effects; cortical excitability, and brain-derived neurotrophic factor (BDNF).\nA quality assessment was performed by two authors (I.F.-M., J.J.R.-Á.) according to the PEDro scale[29] and reviewed the impact factor and quartile of the journal.\nThe independent variables of the study were the following: target tissue; treatment time; number of sessions; current type; current frequency; pulse width, and current intensity. On the other hand, the dependent variables were: pain intensity; range of motion (ROM); maximal isometric strength; balance; muscular endurance; muscle contractile properties; functionality/disability; quality of life; intake of drugs; sleep quality; subjective improvement; adverse effects; cortical excitability, and brain-derived neurotrophic factor (BDNF).", "Systematic review on the effects of PNM. There was used the PRISMA protocol[27] and the research question was stated according to the PICO strategy.[28] This article does not contain any studies with human participants or animals performed by any of the authors. The medical ethics committee approval is not required for this type of study.", "The literature research was performed between April and June 2020. In order to do so, there were used the PubMed, Cochrane, Scopus, and Web of Science databases. The searching phrase was created by combining keywords and Medical Subject Headings that defined the use of electrically stimulated needles and treatments or therapies were not wanted in the search, by using them with the Boolean operators OR and NOT. (“percutaneous electric nerve stimulation” OR “PNM therapy” OR “PNM therapies” OR “percutaneous electrical neuromodulation”) OR (“electrical dry needling [DN]”) OR (“intramuscular electrical stimulation”) NOT (“spinal cord stimulation”) NOT (“transcutaneous electric nerve stimulation”) NOT (“transcranial direct current stimulation [tDCS]”) NOT (“implanted” OR “surgery” OR “surgical”). The search was limited to articles published between 2010 and 2020, both in English and Spanish. The first selection of articles was screened by removing those that were not related to the topic. After fully reading the articles, a second screening was performed. Finally, the selected articles had to comply with some inclusion and exclusion criteria:", "Clinical trial tested on humans; and at least one of the experimental groups should receive treatment based on DN or acupuncture with electrical stimulation.", "Use of the PNM in non-neuromusculoskeletal pathologies; treatments based on surgery of electrostimulation devices; not using needles as a conducting electrode; treatment based on tDCS; treatments based on Chinese traditional medicine, which treats another region, far from the region in pain and with no direct relationship between the region receiving the treatment and the pathological region: auriculotherapy, digit puncture, acupuncture…", "A quality assessment was performed by two authors (I.F.-M., J.J.R.-Á.) according to the PEDro scale[29] and reviewed the impact factor and quartile of the journal.\nThe independent variables of the study were the following: target tissue; treatment time; number of sessions; current type; current frequency; pulse width, and current intensity. On the other hand, the dependent variables were: pain intensity; range of motion (ROM); maximal isometric strength; balance; muscular endurance; muscle contractile properties; functionality/disability; quality of life; intake of drugs; sleep quality; subjective improvement; adverse effects; cortical excitability, and brain-derived neurotrophic factor (BDNF).", "Through the literature search in said four databases, we found 176 potential articles for this review by all authors (I.F.-M., J.J.R.-Á., R.M.-L., and E.S.R-L). Among those, we selected 24 and narrowed it down to 15,[14,16–25,30–33] since 9 were duplicates. After reading the 15 articles, we did not discard a single one and we used them to make the bibliographical review (Fig. 1)\nFlowchart.", "Among the selected 15 articles, 14 were randomized studies[16–25,30–33] and the last one was a prospective observational study.[14]", "The articles obtained an average score of 7.06 ± 1.84 on the PEDro scale[29] (Table 1).\nMethodological quality according to PEDro scale.\nEligibility criteria were specified.\nSubjects were randomly allocated to groups (in a crossover study, subjects were randomly allocated an order in which treatments were received).\nAllocation was concealed.\nThe groups were similar at baseline regarding the most important prognostic indicators.\nThere was blinding of all subjects.\nThere was blinding of all therapists who administered the therapy.\nThere was blinding of all assessors who measured at least one key outcome.\nMeasures of at least one key outcome were obtained from >85% of the subjects initially allocated to groups.\nAll subjects for whom outcome measures were available received the treatment or control condition as allocated or, where this was not the case, data for at least one key outcome was analyzed by “intention to treat”.\nThe results of between-group statistical comparisons are reported for at least one key outcome.\nThe study provides both point measures and measures of variability for at least one key outcome.", "All articles that analyzed the effect of PNM on pain found significant improvements. The studies made by Hadizadeh et al[23] in patients with trigger points in upper trapezius, Botelho et al[16] in patients with myofascial pain syndrome, da Graca-Tarragó et al 2016[17] in patients with knee osteoarthritis, and Raphael et al[33] in patients with chronic pain and hyperalgesia obtained a significant improvement when it came to pain intensity (P ≤ .048; P < .001; P = .001, and P < .0005, respectively). Likewise, García-Bermejo et al 2020 found a significant improvement on PNM based treatment when treating unilateral anterior knee pain, regardless using it in a contralateral or homolateral manner (P = .01 in both groups).[22]\nOn the other hand, Rossi et al[14] observed a significant improvement in patients with neuropathic pain (P < .001), which began 60 minutes post-treatment and lasted for 6 months.\nSome studies compared the effects of PNM to other techniques. Pérez-Palomares et al found no significant difference (P = .94) between PNM and DN in patients with chronic low back pain when it came to mid-term effect.[32] Nonetheless, León-Hernández et al did find significant differences when combining PNM with DN against only using DN on patients with chronic neck pain (P < .05), with a significant decrease in the post-needling soreness intensity 72 hours after using PNM (P < .05).[24] Dunning et al[30,31] observed a significant decrease in pain thanks to the PNM treatment, compared to manual therapy, in patients with knee osteoarthritis and plantar fasciitis. Said decrease lasted for 3 months. Sumen et al[25] compared PNM treatment to low-level laser therapy in patients with an active trigger point in upper trapezius and diagnosed with myofascial pain syndrome. Both treatments improved symptomatology (P = .016 and P = .001, respectively).[25]\nFinally, da Graca-Tarragó et al 2019 observed that patients with knee osteoarthritis that received transcranial direct currents and PNM, regardless of being simulated or not, showed a significant improvement (P < .03), with a higher reduction of the pain intensity when combining both techniques.[18]", "All articles found a significant improvement in the pressure pain threshold when using PNM or DN, with no significant differences between them.[17,18,24,25,32]", "There was a significant improvement in three out of six articles that analyzed this parameter. In patients with trigger points in upper trapezius (P = .048),[23] patients with bilateral reduced hamstring syndrome (P < .01),[21] and patients with anterior knee pain (P = .001).[22] Nonetheless, de la Cruz et al 2019 found no significant improvement in ballet dancers,[19,21] nor did Sumen et al in patients with trigger points in upper trapezius.[25]", "Two studies evaluated balance and muscle endurance and observed a significant improvement in ballet dancers (P < .001 in both cases).[19,21]", "Only one study evaluated the effects of PNM on muscle contractile properties through tensiomyography and found no significant differences (P > .05).[20]", "Five studies analyzed the effects of PNM on functionality/disability[18,22,24,30,31] and all of them found significant improvements. In patients with knee osteoarthritis (P < .001 and P = .03)[18,31] in patients with plantar fasciitis (P < .001),[30] in patients with anterior knee pain (P = .001),[22] and in patients with neck pain, if they combined the treatment with DN.[24]", "The only study that analyzed the quality of life compared PNM and DN, and found no significant differences.[32]", "The 5 studies that analyzed intake of drugs reported that there was a reduction in the use of analgesics and non-steroidal anti-inflammatory drugs after PNM based treatment. In chronic neuropathic pain,[14] in plantar fasciitis (P = .001),[30] in knee osteoarthritis (P = .023 and P < .019)[18,31] and in myofascial pain syndrome.[16]", "Two studies evaluated the sleep quality. In the myofascial pain syndrome, Botelho et al found an improvement in said parameter (P = .04).[16] Nonetheless, Pérez-Palomares et al[32] found no major differences when it came to chronic low back pain (P = .68).", "The three articles that analyze subjective improvement after PNM found a significant improvement in patients with neuropathic pain,[14] knee osteoarthritis,[31] and plantar fasciitis[30] (P < .001 in all three cases).", "Three articles analyze the effect of PNM on the function of the descending pain modulatory system, obtaining significant improvements. Those three articles made the analysis based on myofascial pain (P = .01)[16] and osteoarthritis (P = .01).[17,18]", "Two articles analyzed the effects of PNM on cortical excitability and found a significant decrease in the motor evoked potential in myofascial pain (P = .02)[16] and osteoarthritis (P = .03).[17] In terms of the cortical silent period, both studies agree on the significant increase in said parameter (P = .005 and P = .001, respectively). Finally, none of those studies found significant differences when it came to short intracortical inhibition.", "Three articles studied the effect of PNM on BDNF, with conflicting results. The BDNF is a protein associated with nerve growth factor who is secreted by astrocytes and glial cells and produces spinal cord neurons sensitization, facilitates the activation of N-Methyl-d-asparyaye and increases the excitability of gamma-aminobutyric acid-ergic neurons.[16] Botelho et al obtained significantly higher results of BDNF (P < .01) than the placebo when it came to myofascial pain.[16] Nonetheless, da Graca-Tarragó et al 2019 found no effect on BDNF when treating knee osteoarthritis.[18] On the other hand, in the study by da Graca-Tarragó et al 2016, they observed that BDNF had no correlation with the motor evoked potential, but it had a negative and marginal correlation (P = .05) with the pressure pain threshold.[17]", "Nine articles informed of the possible adverse effects of the PNM; two of them found no such effects after applying treatment[19,33] and three of them had no unspecified significant, severe or moderate adverse effects.[16–18] In three articles, patients reported frequent post-needling soreness and/or hematomas,[14,30,31] and two studies reported occasional somnolence, headache, and nausea.[30,31] All articles that showed there were adverse effects after treatment coincide that said effects disappeared spontaneously within 1 to 4 days.", "The main limitations of the study were the lack of previous research studies on the subject and the lack of data on opioid intake in the selected studies.", "The use of PNM seems useful when treating neuromusculoskeletal injuries. However, results may not be conclusive, since there are few articles published currently and we cannot confirm that the results obtained with PNM to be applicable to all neuromusculoskeletal injuries.\nType of study, subjects, target tissue, duration of the treatment, current parameters, and groups.\na/s = active/shame, CONTR = contralateral, DN = dry needling, E = exercise, Ecc = eccentric, G = group, HOMO = homolateral, LLLT = low-level laser therapy, MPS = myofascial pain syndrome, MT = manual therapy, MTP = myofascial trigger point, N = nerve, PNM = percutaneous neuromodulation, S = stretching, tDCS = transcranial direct current stimulation, TENS = transcutaneous electrical nerve stimulation, US = ultrasounds.\nResults.\na/s = active/shame, BDNF = brain derived neurotrophic factor, CPM = conditioned pain modulation, DN = dry needling, E = exercise, G = group, MEP = motor evoked potential, MT = manual therapy, PNM = percutaneous neuromodulation, S = stretching, tDCS = transcranial direct current stimulation, WOMAC = Western Ontario and McMaster Universities Arthritis Index.", "Conceptualization: Ibon Fidalgo-Martin, Juan José Ramos-Álvarez.\nFormal analysis: Juan José Ramos-Álvarez, Elena Sonsoles Rodríguez-López.\nFunding acquisition: Elena Sonsoles Rodríguez-López.\nInvestigation: Ibon Fidalgo-Martin, Juan José Ramos-Álvarez, Roberto Murias-Lozano, Elena Sonsoles Rodríguez-López.\nMethodology: Ibon Fidalgo-Martin, Juan José Ramos-Álvarez, Roberto Murias-Lozano, Elena Sonsoles Rodríguez-López.\nResources: Ibon Fidalgo-Martin, Roberto Murias-Lozano.\nSoftware: Ibon Fidalgo-Martin.\nSupervision: Ibon Fidalgo-Martin, Juan José Ramos-Álvarez, Roberto Murias-Lozano, Elena Sonsoles Rodríguez-López.\nValidation: Juan José Ramos-Álvarez.\nWriting – original draft: Ibon Fidalgo-Martin, Juan José Ramos-Álvarez.\nWriting – review & editing: Juan José Ramos-Álvarez, Roberto Murias-Lozano, Elena Sonsoles Rodríguez-López." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Methodology", "2.1. Design", "2.2. Identification and selection of articles", "2.3. Inclusion criteria", "2.4. Exclusion criteria", "2.5. Evaluation of the characteristics of the articles", "3. Results", "3.1. Selection of articles", "3.2. Types of study", "3.3. Methodological quality of the selected articles", "3.4. Pain intensity", "3.5. Pressure pain threshold", "3.6. Range of motion", "3.7. Balance and muscle endurance", "3.8. Muscle contractile properties", "3.9. Functionality/disability", "3.10. Quality of life", "3.11. Intake of drugs", "3.12. Sleep quality", "3.13. Subjective improvement", "3.14. Function of the descending pain modulatory system", "3.15. Cortical excitability", "3.16. Brain derived neurotrophic factor", "3.17. Adverse effects", "4. Discussion", "5. Limits of the study", "6. Conclusion", "Author contributions" ]
[ "Neuromodulation is a quality of the nervous system that regulates or modifies electrical impulses, by enhancing or inhibiting them.[1] Even though its mechanism of action is yet to be fully determined, it is known that it lies on the ascending and descending pathways and the supraspinal regions of the central nervous system. The chronic effect of said neurophysiological process may cause central nervous system plasticity, with lasting clinical effects.[2,3] Neuromodulation is part of the treatment for multiple pathologies with few side effects.[4]\nNowadays, there are described the following neuromodulation procedures, classified according to the area of application[5,6]: at the brain level, cortical stimulation[7] and deep brain stimulation,[8] at the spinal level, stimulation of the dorsal column of the spinal cord,[9] nerve root stimulation[10] and dorsal root ganglion stimulation[11] and at the level of the peripheral nervous system, the stimulation of the peripheral nerve,[12] the stimulation of the receptive field of the peripheral nerve[13] and the stimulation of the muscular motor point.[5]\nPeripheral nerve and muscle level neuromodulation (percutaneous neuromodulation [PNM]) consists in percutaneous electrical stimulation of a peripheral nerve, along its pathway or in a muscle, through a puncture needle with electric current of low or medium frequency. Even though the mechanism of action is not fully known, the PNM spans the ascending and descending pathways, as well as the supraspinal regions of the central nervous system, regulating or modifying the electrical impulses transmitted through said pathways, and thus inhibiting or exciting them.[3,14]\nThe main objective of the application of PNM is to provoke a motor and/or sensitive response that eases pain and restores the usual functioning of the nervous system. Thus, on the one hand, it may relieve central sensitization and hyperexcitability (hyperalgesia or allodynia) related chronic pain and neuropathic pain, and, on the other hand, it may improve the neuromuscular function, muscle recruitment patterns and motor control.[5,14,15] In this line, recent studies suggests PNM could relieve central sensitization by improving conditioned pain modulation, reducing motor-evoked potential and increasing intracortical inhibition.[16–18] Additionally, the effects of this technique are being investigated for the improvement of some physical qualities such as elasticity, strength, balance and muscular endurance.[15,19–25]\nAn injury does not only produce changes in an anatomical level; it also may unfold changes within the nervous system, such as increase in the motoneuron excitability, decrease in the sense of position, movement and strength, decrease in the acetylcholine receptors, decrease in the cholinergic/non-cholinergic muscle stimulation, decrease in voluntary activation, decrease in corticospinal excitability, and central sensitization.[26] Central sensitization, on the other hand, represents an intensification when it comes to the activity of circuits and neurons in the nociceptive pathways, due to the enhancement of the excitability of the membrane and the synapses. In patients with central sensitization, the cortex suffers an aberrant reorganization, and the pain increases drastically. In addition, the perception of any other sensory experience reaches a higher amplitude, duration, and spatial extent, which results in a reduced excitation-inhibition balance.[16]\nTherefore, considering the changes that take place in the nervous system after an injury, and the mechanism of action of PNM, some authors used said technique to treat soft tissue injuries.\nThe objective of this study was to evaluate the effects of PNM on pain and physical capabilities when it comes to neuromusculoskeletal injuries.", "[SUBTITLE] 2.1. Design [SUBSECTION] Systematic review on the effects of PNM. There was used the PRISMA protocol[27] and the research question was stated according to the PICO strategy.[28] This article does not contain any studies with human participants or animals performed by any of the authors. The medical ethics committee approval is not required for this type of study.\nSystematic review on the effects of PNM. There was used the PRISMA protocol[27] and the research question was stated according to the PICO strategy.[28] This article does not contain any studies with human participants or animals performed by any of the authors. The medical ethics committee approval is not required for this type of study.\n[SUBTITLE] 2.2. Identification and selection of articles [SUBSECTION] The literature research was performed between April and June 2020. In order to do so, there were used the PubMed, Cochrane, Scopus, and Web of Science databases. The searching phrase was created by combining keywords and Medical Subject Headings that defined the use of electrically stimulated needles and treatments or therapies were not wanted in the search, by using them with the Boolean operators OR and NOT. (“percutaneous electric nerve stimulation” OR “PNM therapy” OR “PNM therapies” OR “percutaneous electrical neuromodulation”) OR (“electrical dry needling [DN]”) OR (“intramuscular electrical stimulation”) NOT (“spinal cord stimulation”) NOT (“transcutaneous electric nerve stimulation”) NOT (“transcranial direct current stimulation [tDCS]”) NOT (“implanted” OR “surgery” OR “surgical”). The search was limited to articles published between 2010 and 2020, both in English and Spanish. The first selection of articles was screened by removing those that were not related to the topic. After fully reading the articles, a second screening was performed. Finally, the selected articles had to comply with some inclusion and exclusion criteria:\nThe literature research was performed between April and June 2020. In order to do so, there were used the PubMed, Cochrane, Scopus, and Web of Science databases. The searching phrase was created by combining keywords and Medical Subject Headings that defined the use of electrically stimulated needles and treatments or therapies were not wanted in the search, by using them with the Boolean operators OR and NOT. (“percutaneous electric nerve stimulation” OR “PNM therapy” OR “PNM therapies” OR “percutaneous electrical neuromodulation”) OR (“electrical dry needling [DN]”) OR (“intramuscular electrical stimulation”) NOT (“spinal cord stimulation”) NOT (“transcutaneous electric nerve stimulation”) NOT (“transcranial direct current stimulation [tDCS]”) NOT (“implanted” OR “surgery” OR “surgical”). The search was limited to articles published between 2010 and 2020, both in English and Spanish. The first selection of articles was screened by removing those that were not related to the topic. After fully reading the articles, a second screening was performed. Finally, the selected articles had to comply with some inclusion and exclusion criteria:\n[SUBTITLE] 2.3. Inclusion criteria [SUBSECTION] Clinical trial tested on humans; and at least one of the experimental groups should receive treatment based on DN or acupuncture with electrical stimulation.\nClinical trial tested on humans; and at least one of the experimental groups should receive treatment based on DN or acupuncture with electrical stimulation.\n[SUBTITLE] 2.4. Exclusion criteria [SUBSECTION] Use of the PNM in non-neuromusculoskeletal pathologies; treatments based on surgery of electrostimulation devices; not using needles as a conducting electrode; treatment based on tDCS; treatments based on Chinese traditional medicine, which treats another region, far from the region in pain and with no direct relationship between the region receiving the treatment and the pathological region: auriculotherapy, digit puncture, acupuncture…\nUse of the PNM in non-neuromusculoskeletal pathologies; treatments based on surgery of electrostimulation devices; not using needles as a conducting electrode; treatment based on tDCS; treatments based on Chinese traditional medicine, which treats another region, far from the region in pain and with no direct relationship between the region receiving the treatment and the pathological region: auriculotherapy, digit puncture, acupuncture…\n[SUBTITLE] 2.5. Evaluation of the characteristics of the articles [SUBSECTION] A quality assessment was performed by two authors (I.F.-M., J.J.R.-Á.) according to the PEDro scale[29] and reviewed the impact factor and quartile of the journal.\nThe independent variables of the study were the following: target tissue; treatment time; number of sessions; current type; current frequency; pulse width, and current intensity. On the other hand, the dependent variables were: pain intensity; range of motion (ROM); maximal isometric strength; balance; muscular endurance; muscle contractile properties; functionality/disability; quality of life; intake of drugs; sleep quality; subjective improvement; adverse effects; cortical excitability, and brain-derived neurotrophic factor (BDNF).\nA quality assessment was performed by two authors (I.F.-M., J.J.R.-Á.) according to the PEDro scale[29] and reviewed the impact factor and quartile of the journal.\nThe independent variables of the study were the following: target tissue; treatment time; number of sessions; current type; current frequency; pulse width, and current intensity. On the other hand, the dependent variables were: pain intensity; range of motion (ROM); maximal isometric strength; balance; muscular endurance; muscle contractile properties; functionality/disability; quality of life; intake of drugs; sleep quality; subjective improvement; adverse effects; cortical excitability, and brain-derived neurotrophic factor (BDNF).", "Systematic review on the effects of PNM. There was used the PRISMA protocol[27] and the research question was stated according to the PICO strategy.[28] This article does not contain any studies with human participants or animals performed by any of the authors. The medical ethics committee approval is not required for this type of study.", "The literature research was performed between April and June 2020. In order to do so, there were used the PubMed, Cochrane, Scopus, and Web of Science databases. The searching phrase was created by combining keywords and Medical Subject Headings that defined the use of electrically stimulated needles and treatments or therapies were not wanted in the search, by using them with the Boolean operators OR and NOT. (“percutaneous electric nerve stimulation” OR “PNM therapy” OR “PNM therapies” OR “percutaneous electrical neuromodulation”) OR (“electrical dry needling [DN]”) OR (“intramuscular electrical stimulation”) NOT (“spinal cord stimulation”) NOT (“transcutaneous electric nerve stimulation”) NOT (“transcranial direct current stimulation [tDCS]”) NOT (“implanted” OR “surgery” OR “surgical”). The search was limited to articles published between 2010 and 2020, both in English and Spanish. The first selection of articles was screened by removing those that were not related to the topic. After fully reading the articles, a second screening was performed. Finally, the selected articles had to comply with some inclusion and exclusion criteria:", "Clinical trial tested on humans; and at least one of the experimental groups should receive treatment based on DN or acupuncture with electrical stimulation.", "Use of the PNM in non-neuromusculoskeletal pathologies; treatments based on surgery of electrostimulation devices; not using needles as a conducting electrode; treatment based on tDCS; treatments based on Chinese traditional medicine, which treats another region, far from the region in pain and with no direct relationship between the region receiving the treatment and the pathological region: auriculotherapy, digit puncture, acupuncture…", "A quality assessment was performed by two authors (I.F.-M., J.J.R.-Á.) according to the PEDro scale[29] and reviewed the impact factor and quartile of the journal.\nThe independent variables of the study were the following: target tissue; treatment time; number of sessions; current type; current frequency; pulse width, and current intensity. On the other hand, the dependent variables were: pain intensity; range of motion (ROM); maximal isometric strength; balance; muscular endurance; muscle contractile properties; functionality/disability; quality of life; intake of drugs; sleep quality; subjective improvement; adverse effects; cortical excitability, and brain-derived neurotrophic factor (BDNF).", "[SUBTITLE] 3.1. Selection of articles [SUBSECTION] Through the literature search in said four databases, we found 176 potential articles for this review by all authors (I.F.-M., J.J.R.-Á., R.M.-L., and E.S.R-L). Among those, we selected 24 and narrowed it down to 15,[14,16–25,30–33] since 9 were duplicates. After reading the 15 articles, we did not discard a single one and we used them to make the bibliographical review (Fig. 1)\nFlowchart.\nThrough the literature search in said four databases, we found 176 potential articles for this review by all authors (I.F.-M., J.J.R.-Á., R.M.-L., and E.S.R-L). Among those, we selected 24 and narrowed it down to 15,[14,16–25,30–33] since 9 were duplicates. After reading the 15 articles, we did not discard a single one and we used them to make the bibliographical review (Fig. 1)\nFlowchart.\n[SUBTITLE] 3.2. Types of study [SUBSECTION] Among the selected 15 articles, 14 were randomized studies[16–25,30–33] and the last one was a prospective observational study.[14]\nAmong the selected 15 articles, 14 were randomized studies[16–25,30–33] and the last one was a prospective observational study.[14]\n[SUBTITLE] 3.3. Methodological quality of the selected articles [SUBSECTION] The articles obtained an average score of 7.06 ± 1.84 on the PEDro scale[29] (Table 1).\nMethodological quality according to PEDro scale.\nEligibility criteria were specified.\nSubjects were randomly allocated to groups (in a crossover study, subjects were randomly allocated an order in which treatments were received).\nAllocation was concealed.\nThe groups were similar at baseline regarding the most important prognostic indicators.\nThere was blinding of all subjects.\nThere was blinding of all therapists who administered the therapy.\nThere was blinding of all assessors who measured at least one key outcome.\nMeasures of at least one key outcome were obtained from >85% of the subjects initially allocated to groups.\nAll subjects for whom outcome measures were available received the treatment or control condition as allocated or, where this was not the case, data for at least one key outcome was analyzed by “intention to treat”.\nThe results of between-group statistical comparisons are reported for at least one key outcome.\nThe study provides both point measures and measures of variability for at least one key outcome.\nThe articles obtained an average score of 7.06 ± 1.84 on the PEDro scale[29] (Table 1).\nMethodological quality according to PEDro scale.\nEligibility criteria were specified.\nSubjects were randomly allocated to groups (in a crossover study, subjects were randomly allocated an order in which treatments were received).\nAllocation was concealed.\nThe groups were similar at baseline regarding the most important prognostic indicators.\nThere was blinding of all subjects.\nThere was blinding of all therapists who administered the therapy.\nThere was blinding of all assessors who measured at least one key outcome.\nMeasures of at least one key outcome were obtained from >85% of the subjects initially allocated to groups.\nAll subjects for whom outcome measures were available received the treatment or control condition as allocated or, where this was not the case, data for at least one key outcome was analyzed by “intention to treat”.\nThe results of between-group statistical comparisons are reported for at least one key outcome.\nThe study provides both point measures and measures of variability for at least one key outcome.\n[SUBTITLE] 3.4. Pain intensity [SUBSECTION] All articles that analyzed the effect of PNM on pain found significant improvements. The studies made by Hadizadeh et al[23] in patients with trigger points in upper trapezius, Botelho et al[16] in patients with myofascial pain syndrome, da Graca-Tarragó et al 2016[17] in patients with knee osteoarthritis, and Raphael et al[33] in patients with chronic pain and hyperalgesia obtained a significant improvement when it came to pain intensity (P ≤ .048; P < .001; P = .001, and P < .0005, respectively). Likewise, García-Bermejo et al 2020 found a significant improvement on PNM based treatment when treating unilateral anterior knee pain, regardless using it in a contralateral or homolateral manner (P = .01 in both groups).[22]\nOn the other hand, Rossi et al[14] observed a significant improvement in patients with neuropathic pain (P < .001), which began 60 minutes post-treatment and lasted for 6 months.\nSome studies compared the effects of PNM to other techniques. Pérez-Palomares et al found no significant difference (P = .94) between PNM and DN in patients with chronic low back pain when it came to mid-term effect.[32] Nonetheless, León-Hernández et al did find significant differences when combining PNM with DN against only using DN on patients with chronic neck pain (P < .05), with a significant decrease in the post-needling soreness intensity 72 hours after using PNM (P < .05).[24] Dunning et al[30,31] observed a significant decrease in pain thanks to the PNM treatment, compared to manual therapy, in patients with knee osteoarthritis and plantar fasciitis. Said decrease lasted for 3 months. Sumen et al[25] compared PNM treatment to low-level laser therapy in patients with an active trigger point in upper trapezius and diagnosed with myofascial pain syndrome. Both treatments improved symptomatology (P = .016 and P = .001, respectively).[25]\nFinally, da Graca-Tarragó et al 2019 observed that patients with knee osteoarthritis that received transcranial direct currents and PNM, regardless of being simulated or not, showed a significant improvement (P < .03), with a higher reduction of the pain intensity when combining both techniques.[18]\nAll articles that analyzed the effect of PNM on pain found significant improvements. The studies made by Hadizadeh et al[23] in patients with trigger points in upper trapezius, Botelho et al[16] in patients with myofascial pain syndrome, da Graca-Tarragó et al 2016[17] in patients with knee osteoarthritis, and Raphael et al[33] in patients with chronic pain and hyperalgesia obtained a significant improvement when it came to pain intensity (P ≤ .048; P < .001; P = .001, and P < .0005, respectively). Likewise, García-Bermejo et al 2020 found a significant improvement on PNM based treatment when treating unilateral anterior knee pain, regardless using it in a contralateral or homolateral manner (P = .01 in both groups).[22]\nOn the other hand, Rossi et al[14] observed a significant improvement in patients with neuropathic pain (P < .001), which began 60 minutes post-treatment and lasted for 6 months.\nSome studies compared the effects of PNM to other techniques. Pérez-Palomares et al found no significant difference (P = .94) between PNM and DN in patients with chronic low back pain when it came to mid-term effect.[32] Nonetheless, León-Hernández et al did find significant differences when combining PNM with DN against only using DN on patients with chronic neck pain (P < .05), with a significant decrease in the post-needling soreness intensity 72 hours after using PNM (P < .05).[24] Dunning et al[30,31] observed a significant decrease in pain thanks to the PNM treatment, compared to manual therapy, in patients with knee osteoarthritis and plantar fasciitis. Said decrease lasted for 3 months. Sumen et al[25] compared PNM treatment to low-level laser therapy in patients with an active trigger point in upper trapezius and diagnosed with myofascial pain syndrome. Both treatments improved symptomatology (P = .016 and P = .001, respectively).[25]\nFinally, da Graca-Tarragó et al 2019 observed that patients with knee osteoarthritis that received transcranial direct currents and PNM, regardless of being simulated or not, showed a significant improvement (P < .03), with a higher reduction of the pain intensity when combining both techniques.[18]\n[SUBTITLE] 3.5. Pressure pain threshold [SUBSECTION] All articles found a significant improvement in the pressure pain threshold when using PNM or DN, with no significant differences between them.[17,18,24,25,32]\nAll articles found a significant improvement in the pressure pain threshold when using PNM or DN, with no significant differences between them.[17,18,24,25,32]\n[SUBTITLE] 3.6. Range of motion [SUBSECTION] There was a significant improvement in three out of six articles that analyzed this parameter. In patients with trigger points in upper trapezius (P = .048),[23] patients with bilateral reduced hamstring syndrome (P < .01),[21] and patients with anterior knee pain (P = .001).[22] Nonetheless, de la Cruz et al 2019 found no significant improvement in ballet dancers,[19,21] nor did Sumen et al in patients with trigger points in upper trapezius.[25]\nThere was a significant improvement in three out of six articles that analyzed this parameter. In patients with trigger points in upper trapezius (P = .048),[23] patients with bilateral reduced hamstring syndrome (P < .01),[21] and patients with anterior knee pain (P = .001).[22] Nonetheless, de la Cruz et al 2019 found no significant improvement in ballet dancers,[19,21] nor did Sumen et al in patients with trigger points in upper trapezius.[25]\n[SUBTITLE] 3.7. Balance and muscle endurance [SUBSECTION] Two studies evaluated balance and muscle endurance and observed a significant improvement in ballet dancers (P < .001 in both cases).[19,21]\nTwo studies evaluated balance and muscle endurance and observed a significant improvement in ballet dancers (P < .001 in both cases).[19,21]\n[SUBTITLE] 3.8. Muscle contractile properties [SUBSECTION] Only one study evaluated the effects of PNM on muscle contractile properties through tensiomyography and found no significant differences (P > .05).[20]\nOnly one study evaluated the effects of PNM on muscle contractile properties through tensiomyography and found no significant differences (P > .05).[20]\n[SUBTITLE] 3.9. Functionality/disability [SUBSECTION] Five studies analyzed the effects of PNM on functionality/disability[18,22,24,30,31] and all of them found significant improvements. In patients with knee osteoarthritis (P < .001 and P = .03)[18,31] in patients with plantar fasciitis (P < .001),[30] in patients with anterior knee pain (P = .001),[22] and in patients with neck pain, if they combined the treatment with DN.[24]\nFive studies analyzed the effects of PNM on functionality/disability[18,22,24,30,31] and all of them found significant improvements. In patients with knee osteoarthritis (P < .001 and P = .03)[18,31] in patients with plantar fasciitis (P < .001),[30] in patients with anterior knee pain (P = .001),[22] and in patients with neck pain, if they combined the treatment with DN.[24]\n[SUBTITLE] 3.10. Quality of life [SUBSECTION] The only study that analyzed the quality of life compared PNM and DN, and found no significant differences.[32]\nThe only study that analyzed the quality of life compared PNM and DN, and found no significant differences.[32]\n[SUBTITLE] 3.11. Intake of drugs [SUBSECTION] The 5 studies that analyzed intake of drugs reported that there was a reduction in the use of analgesics and non-steroidal anti-inflammatory drugs after PNM based treatment. In chronic neuropathic pain,[14] in plantar fasciitis (P = .001),[30] in knee osteoarthritis (P = .023 and P < .019)[18,31] and in myofascial pain syndrome.[16]\nThe 5 studies that analyzed intake of drugs reported that there was a reduction in the use of analgesics and non-steroidal anti-inflammatory drugs after PNM based treatment. In chronic neuropathic pain,[14] in plantar fasciitis (P = .001),[30] in knee osteoarthritis (P = .023 and P < .019)[18,31] and in myofascial pain syndrome.[16]\n[SUBTITLE] 3.12. Sleep quality [SUBSECTION] Two studies evaluated the sleep quality. In the myofascial pain syndrome, Botelho et al found an improvement in said parameter (P = .04).[16] Nonetheless, Pérez-Palomares et al[32] found no major differences when it came to chronic low back pain (P = .68).\nTwo studies evaluated the sleep quality. In the myofascial pain syndrome, Botelho et al found an improvement in said parameter (P = .04).[16] Nonetheless, Pérez-Palomares et al[32] found no major differences when it came to chronic low back pain (P = .68).\n[SUBTITLE] 3.13. Subjective improvement [SUBSECTION] The three articles that analyze subjective improvement after PNM found a significant improvement in patients with neuropathic pain,[14] knee osteoarthritis,[31] and plantar fasciitis[30] (P < .001 in all three cases).\nThe three articles that analyze subjective improvement after PNM found a significant improvement in patients with neuropathic pain,[14] knee osteoarthritis,[31] and plantar fasciitis[30] (P < .001 in all three cases).\n[SUBTITLE] 3.14. Function of the descending pain modulatory system [SUBSECTION] Three articles analyze the effect of PNM on the function of the descending pain modulatory system, obtaining significant improvements. Those three articles made the analysis based on myofascial pain (P = .01)[16] and osteoarthritis (P = .01).[17,18]\nThree articles analyze the effect of PNM on the function of the descending pain modulatory system, obtaining significant improvements. Those three articles made the analysis based on myofascial pain (P = .01)[16] and osteoarthritis (P = .01).[17,18]\n[SUBTITLE] 3.15. Cortical excitability [SUBSECTION] Two articles analyzed the effects of PNM on cortical excitability and found a significant decrease in the motor evoked potential in myofascial pain (P = .02)[16] and osteoarthritis (P = .03).[17] In terms of the cortical silent period, both studies agree on the significant increase in said parameter (P = .005 and P = .001, respectively). Finally, none of those studies found significant differences when it came to short intracortical inhibition.\nTwo articles analyzed the effects of PNM on cortical excitability and found a significant decrease in the motor evoked potential in myofascial pain (P = .02)[16] and osteoarthritis (P = .03).[17] In terms of the cortical silent period, both studies agree on the significant increase in said parameter (P = .005 and P = .001, respectively). Finally, none of those studies found significant differences when it came to short intracortical inhibition.\n[SUBTITLE] 3.16. Brain derived neurotrophic factor [SUBSECTION] Three articles studied the effect of PNM on BDNF, with conflicting results. The BDNF is a protein associated with nerve growth factor who is secreted by astrocytes and glial cells and produces spinal cord neurons sensitization, facilitates the activation of N-Methyl-d-asparyaye and increases the excitability of gamma-aminobutyric acid-ergic neurons.[16] Botelho et al obtained significantly higher results of BDNF (P < .01) than the placebo when it came to myofascial pain.[16] Nonetheless, da Graca-Tarragó et al 2019 found no effect on BDNF when treating knee osteoarthritis.[18] On the other hand, in the study by da Graca-Tarragó et al 2016, they observed that BDNF had no correlation with the motor evoked potential, but it had a negative and marginal correlation (P = .05) with the pressure pain threshold.[17]\nThree articles studied the effect of PNM on BDNF, with conflicting results. The BDNF is a protein associated with nerve growth factor who is secreted by astrocytes and glial cells and produces spinal cord neurons sensitization, facilitates the activation of N-Methyl-d-asparyaye and increases the excitability of gamma-aminobutyric acid-ergic neurons.[16] Botelho et al obtained significantly higher results of BDNF (P < .01) than the placebo when it came to myofascial pain.[16] Nonetheless, da Graca-Tarragó et al 2019 found no effect on BDNF when treating knee osteoarthritis.[18] On the other hand, in the study by da Graca-Tarragó et al 2016, they observed that BDNF had no correlation with the motor evoked potential, but it had a negative and marginal correlation (P = .05) with the pressure pain threshold.[17]\n[SUBTITLE] 3.17. Adverse effects [SUBSECTION] Nine articles informed of the possible adverse effects of the PNM; two of them found no such effects after applying treatment[19,33] and three of them had no unspecified significant, severe or moderate adverse effects.[16–18] In three articles, patients reported frequent post-needling soreness and/or hematomas,[14,30,31] and two studies reported occasional somnolence, headache, and nausea.[30,31] All articles that showed there were adverse effects after treatment coincide that said effects disappeared spontaneously within 1 to 4 days.\nNine articles informed of the possible adverse effects of the PNM; two of them found no such effects after applying treatment[19,33] and three of them had no unspecified significant, severe or moderate adverse effects.[16–18] In three articles, patients reported frequent post-needling soreness and/or hematomas,[14,30,31] and two studies reported occasional somnolence, headache, and nausea.[30,31] All articles that showed there were adverse effects after treatment coincide that said effects disappeared spontaneously within 1 to 4 days.", "Through the literature search in said four databases, we found 176 potential articles for this review by all authors (I.F.-M., J.J.R.-Á., R.M.-L., and E.S.R-L). Among those, we selected 24 and narrowed it down to 15,[14,16–25,30–33] since 9 were duplicates. After reading the 15 articles, we did not discard a single one and we used them to make the bibliographical review (Fig. 1)\nFlowchart.", "Among the selected 15 articles, 14 were randomized studies[16–25,30–33] and the last one was a prospective observational study.[14]", "The articles obtained an average score of 7.06 ± 1.84 on the PEDro scale[29] (Table 1).\nMethodological quality according to PEDro scale.\nEligibility criteria were specified.\nSubjects were randomly allocated to groups (in a crossover study, subjects were randomly allocated an order in which treatments were received).\nAllocation was concealed.\nThe groups were similar at baseline regarding the most important prognostic indicators.\nThere was blinding of all subjects.\nThere was blinding of all therapists who administered the therapy.\nThere was blinding of all assessors who measured at least one key outcome.\nMeasures of at least one key outcome were obtained from >85% of the subjects initially allocated to groups.\nAll subjects for whom outcome measures were available received the treatment or control condition as allocated or, where this was not the case, data for at least one key outcome was analyzed by “intention to treat”.\nThe results of between-group statistical comparisons are reported for at least one key outcome.\nThe study provides both point measures and measures of variability for at least one key outcome.", "All articles that analyzed the effect of PNM on pain found significant improvements. The studies made by Hadizadeh et al[23] in patients with trigger points in upper trapezius, Botelho et al[16] in patients with myofascial pain syndrome, da Graca-Tarragó et al 2016[17] in patients with knee osteoarthritis, and Raphael et al[33] in patients with chronic pain and hyperalgesia obtained a significant improvement when it came to pain intensity (P ≤ .048; P < .001; P = .001, and P < .0005, respectively). Likewise, García-Bermejo et al 2020 found a significant improvement on PNM based treatment when treating unilateral anterior knee pain, regardless using it in a contralateral or homolateral manner (P = .01 in both groups).[22]\nOn the other hand, Rossi et al[14] observed a significant improvement in patients with neuropathic pain (P < .001), which began 60 minutes post-treatment and lasted for 6 months.\nSome studies compared the effects of PNM to other techniques. Pérez-Palomares et al found no significant difference (P = .94) between PNM and DN in patients with chronic low back pain when it came to mid-term effect.[32] Nonetheless, León-Hernández et al did find significant differences when combining PNM with DN against only using DN on patients with chronic neck pain (P < .05), with a significant decrease in the post-needling soreness intensity 72 hours after using PNM (P < .05).[24] Dunning et al[30,31] observed a significant decrease in pain thanks to the PNM treatment, compared to manual therapy, in patients with knee osteoarthritis and plantar fasciitis. Said decrease lasted for 3 months. Sumen et al[25] compared PNM treatment to low-level laser therapy in patients with an active trigger point in upper trapezius and diagnosed with myofascial pain syndrome. Both treatments improved symptomatology (P = .016 and P = .001, respectively).[25]\nFinally, da Graca-Tarragó et al 2019 observed that patients with knee osteoarthritis that received transcranial direct currents and PNM, regardless of being simulated or not, showed a significant improvement (P < .03), with a higher reduction of the pain intensity when combining both techniques.[18]", "All articles found a significant improvement in the pressure pain threshold when using PNM or DN, with no significant differences between them.[17,18,24,25,32]", "There was a significant improvement in three out of six articles that analyzed this parameter. In patients with trigger points in upper trapezius (P = .048),[23] patients with bilateral reduced hamstring syndrome (P < .01),[21] and patients with anterior knee pain (P = .001).[22] Nonetheless, de la Cruz et al 2019 found no significant improvement in ballet dancers,[19,21] nor did Sumen et al in patients with trigger points in upper trapezius.[25]", "Two studies evaluated balance and muscle endurance and observed a significant improvement in ballet dancers (P < .001 in both cases).[19,21]", "Only one study evaluated the effects of PNM on muscle contractile properties through tensiomyography and found no significant differences (P > .05).[20]", "Five studies analyzed the effects of PNM on functionality/disability[18,22,24,30,31] and all of them found significant improvements. In patients with knee osteoarthritis (P < .001 and P = .03)[18,31] in patients with plantar fasciitis (P < .001),[30] in patients with anterior knee pain (P = .001),[22] and in patients with neck pain, if they combined the treatment with DN.[24]", "The only study that analyzed the quality of life compared PNM and DN, and found no significant differences.[32]", "The 5 studies that analyzed intake of drugs reported that there was a reduction in the use of analgesics and non-steroidal anti-inflammatory drugs after PNM based treatment. In chronic neuropathic pain,[14] in plantar fasciitis (P = .001),[30] in knee osteoarthritis (P = .023 and P < .019)[18,31] and in myofascial pain syndrome.[16]", "Two studies evaluated the sleep quality. In the myofascial pain syndrome, Botelho et al found an improvement in said parameter (P = .04).[16] Nonetheless, Pérez-Palomares et al[32] found no major differences when it came to chronic low back pain (P = .68).", "The three articles that analyze subjective improvement after PNM found a significant improvement in patients with neuropathic pain,[14] knee osteoarthritis,[31] and plantar fasciitis[30] (P < .001 in all three cases).", "Three articles analyze the effect of PNM on the function of the descending pain modulatory system, obtaining significant improvements. Those three articles made the analysis based on myofascial pain (P = .01)[16] and osteoarthritis (P = .01).[17,18]", "Two articles analyzed the effects of PNM on cortical excitability and found a significant decrease in the motor evoked potential in myofascial pain (P = .02)[16] and osteoarthritis (P = .03).[17] In terms of the cortical silent period, both studies agree on the significant increase in said parameter (P = .005 and P = .001, respectively). Finally, none of those studies found significant differences when it came to short intracortical inhibition.", "Three articles studied the effect of PNM on BDNF, with conflicting results. The BDNF is a protein associated with nerve growth factor who is secreted by astrocytes and glial cells and produces spinal cord neurons sensitization, facilitates the activation of N-Methyl-d-asparyaye and increases the excitability of gamma-aminobutyric acid-ergic neurons.[16] Botelho et al obtained significantly higher results of BDNF (P < .01) than the placebo when it came to myofascial pain.[16] Nonetheless, da Graca-Tarragó et al 2019 found no effect on BDNF when treating knee osteoarthritis.[18] On the other hand, in the study by da Graca-Tarragó et al 2016, they observed that BDNF had no correlation with the motor evoked potential, but it had a negative and marginal correlation (P = .05) with the pressure pain threshold.[17]", "Nine articles informed of the possible adverse effects of the PNM; two of them found no such effects after applying treatment[19,33] and three of them had no unspecified significant, severe or moderate adverse effects.[16–18] In three articles, patients reported frequent post-needling soreness and/or hematomas,[14,30,31] and two studies reported occasional somnolence, headache, and nausea.[30,31] All articles that showed there were adverse effects after treatment coincide that said effects disappeared spontaneously within 1 to 4 days.", "Among the articles selected for this review, there is a consensus on the fact that PNM produced significant improvements in pain intensity,[14,16–18,22–24,30,31,33] pressure pain threshold,[17,18,25,33] balance and muscle endurance,[19,21] functionality/disability,[18,22,30,31] subjective improvement,[14,30,31] and the function of the descending pain modulatory system.[16–18] Some articles also agree on the effect it has on the intake of drugs after treating with PNM,[14,16,18,30,31] producing a significant change in three of them.[1–3,16,30,31]\nOn the other hand, there was no significant improvement when it came to muscle contractile properties[20] or the quality of life of the patients.[32] The study by de la Cruz et al in 2020[20] was the only one that showed the effect of said technique on muscle contractile properties. Thus, it is hard to draw solid conclusions on the matter. Nonetheless, not finding any significant changes leads us to think that we can use this technique before physical exercise or sport, in case of confirming said findings.\nOn the other hand, the study that analyzed the quality of life of patients after receiving PNM treatment[32] compared its effect to DN, from which we can conclude that none of them obtained better results. In order to establish a direct cause-effect between PNM and the improvement of the quality of life, the study should have a control group.\nIn terms of the ROM, only 50% of the articles that studied this parameter found a significant improvement thanks to the PNM.[20,22,23] The adverse results on the ROM may be conditioned because of the characteristics of the studied sample. In two of the articles, they studied the ROM of ballet dancers, since their activity involves a lot of ROM and, therefore, it is unlikely to improve due to this intervention.[19,21] In the third article, they used PNM only in the trapezius.[25] Evidently, this is not the only muscle involved in neck ROM, which is way the intervention would be insufficient to evaluate said parameter.\nThere is little research when it comes to the improvement of strength by using PNM; as far as we know only one article evaluates this parameter,[15] and even though it obtained good results, it had no control group and it is not published at an indexed journal. It would be necessary to have more studies to confirm that PNM is useful when it comes to restoring strength. We obtain the same conclusion in parameters such as sleep quality, since only two articles have studied it, with conflicting results.[16,32]\nWhen it comes to the effects of PNM on cortical excitability, the two articles that analyzed it[16,18] obtained a significant decrease in the motor evoked potential, and a significant increase in the cortical silent period, with no significant differences on the intracortical inhibition. Intracortical facilitation seemed to improve only when combined with tDCS, a form of top-down neuromodulation through constant, low and direct current delivered via electrodes on the head that modulates the thalamocortical synapses within pain pathways and it might change in thalamic inhibitory pathways, cingulate cortex, and periaqueductal gray matter.[18] Thus, said effect may be the result of the last technique or because of the combination of both. When it comes to BDNF, the results obtained are unalike[16–18] which is why we cannot draw conclusions and need more research. The crossed effect observed by de la Cruz et al 2020[20] and García-Bermejo et al 2020[22]on the untreated limb suggests that PNM does not only produce distal-level effects on the homolateral region, but also may act on the central nervous system, causing bilateral effects.\nSome studies compared the effect of PNM with DN,[24,32] resulting in both being equally effective techniques in short-term, with DN having a better benefit-cost ratio. Nonetheless, the worst post-needling soreness of DN went together with the highest rate of treatment abandonment.[32] However, the combination of both techniques obtained better short-term results, improving post-needling soreness.[4,24] This suggests that it may be a good idea to combine both techniques, but there should be more studies to prove that suggestion. Likewise, combining PNM and tDCS may improve the results rather than using those techniques separately.[18] All the same, only one study considers that possibility.\nFinally, choosing the optimal current parameters was a controversial topic, due to the bibliographical variety. However, it seems that frequency is the most important parameter and with more options in terms of application.[5]\nThere is a huge disparity between the frequency used in the articles within this bibliographical review, with a range between 2 and 100 Hertz (Hz).[14–25,30–33] In relation to the frequency, there are various hypotheses to explain the action mechanisms of the technique. The first hypothesis suggests that electrical impulses modulate in a peripheral manner. According to the second hypothesis, it may be possible to explain their effect by stimulating the release of endogenous opioids, through electric current. Finally, the last hypothesis has observed mechanisms of synaptic plasticity.[14,34,35]\nPaying attention to the mechanism of release of endogenous opioids, they found out that low frequencies (2–5 Hz) activate small-diameter motor fibers. Consequently, they cause a release of enkephalins and endorphins, which cause extra-segmental analgesia, with longer lasting effects. On the other hand, higher frequencies (50–100 Hz) may selectively activate big diameter nonnociceptive Aβ fibers, which cause a release of dynorphins and thus produce a decrease in the activity and sensitization of the nociceptive cells on the segment-level in the central nervous system. The analgesia mechanism produced by high frequencies produce shorter-lasting effects. Nonetheless, its application seems interesting in patients with tolerance towards opioids, since the main mechanism of low frequencies is the release of endogenous opioids, as opposed to the higher frequencies, which seem to be segmental.[14,35–38]\nThe best choice of frequencies may be a combination of both high and low frequencies, since it seems that the therapeutic effect of the combination is more effective than applying them separately.[39]\nThe best application time seems to be 30 minutes, since Hamza et al used 15, 30, and 45 minute intervals and obtained best results when using it for 30 minutes instead of 15 minutes. There was no difference between applying it for 30 or 45 minutes.[40]\nMoreover, there are two types of applications when it comes to the effect of PNM on synaptic plasticity. According to said classification, there are long-term potentiation and long-term depression processes. Long-term potentiation is a form of use-dependent plasticity which can be produced using high frequency stimulation trains of 100 Hz for 1 second repeated five times at 10 seconds intervals. As a result, the synaptic transmission improves persistently. On the other hand, long-term depression, using a low frequency, non-interrupted and long lasted stimulation (17 minutes at 1 Hz) reduces the effectiveness of the synaptic transmission.[9,41,42] Repairing to this, we could adjust the parameters of the application to potentiate Aβ fibers or depress C fibers.\nBesides, recent studies suggests that spinal cord stimulation could be effective for treating allodynia and neuropathic pain by attenuating wide dynamic range neurons hyperexcitability, releasing gamma-aminobutyric acid and decreasing neurotransmitters as glutamate and activating dorsal column fibers in an orthodromic manner, furthermore these authors propose as a promising therapeutic target the dorsal root ganglion.[11,43,44] This finding evidences a new possible therapeutic target in PNM.\nMost of the articles selected in this review have notified adverse effects, even though they were mild and disappeared in 4 days at most. These facts imply that the technique is safe.\nFor this review, we established a minimum score of 5 in the PEDro scale in order to include the articles. Nonetheless, we chose one article with a score of 4, since it was the only ones that evaluated the effects of PNM on neuropathic pain.[14]", "The main limitations of the study were the lack of previous research studies on the subject and the lack of data on opioid intake in the selected studies.", "The use of PNM seems useful when treating neuromusculoskeletal injuries. However, results may not be conclusive, since there are few articles published currently and we cannot confirm that the results obtained with PNM to be applicable to all neuromusculoskeletal injuries.\nType of study, subjects, target tissue, duration of the treatment, current parameters, and groups.\na/s = active/shame, CONTR = contralateral, DN = dry needling, E = exercise, Ecc = eccentric, G = group, HOMO = homolateral, LLLT = low-level laser therapy, MPS = myofascial pain syndrome, MT = manual therapy, MTP = myofascial trigger point, N = nerve, PNM = percutaneous neuromodulation, S = stretching, tDCS = transcranial direct current stimulation, TENS = transcutaneous electrical nerve stimulation, US = ultrasounds.\nResults.\na/s = active/shame, BDNF = brain derived neurotrophic factor, CPM = conditioned pain modulation, DN = dry needling, E = exercise, G = group, MEP = motor evoked potential, MT = manual therapy, PNM = percutaneous neuromodulation, S = stretching, tDCS = transcranial direct current stimulation, WOMAC = Western Ontario and McMaster Universities Arthritis Index.", "Conceptualization: Ibon Fidalgo-Martin, Juan José Ramos-Álvarez.\nFormal analysis: Juan José Ramos-Álvarez, Elena Sonsoles Rodríguez-López.\nFunding acquisition: Elena Sonsoles Rodríguez-López.\nInvestigation: Ibon Fidalgo-Martin, Juan José Ramos-Álvarez, Roberto Murias-Lozano, Elena Sonsoles Rodríguez-López.\nMethodology: Ibon Fidalgo-Martin, Juan José Ramos-Álvarez, Roberto Murias-Lozano, Elena Sonsoles Rodríguez-López.\nResources: Ibon Fidalgo-Martin, Roberto Murias-Lozano.\nSoftware: Ibon Fidalgo-Martin.\nSupervision: Ibon Fidalgo-Martin, Juan José Ramos-Álvarez, Roberto Murias-Lozano, Elena Sonsoles Rodríguez-López.\nValidation: Juan José Ramos-Álvarez.\nWriting – original draft: Ibon Fidalgo-Martin, Juan José Ramos-Álvarez.\nWriting – review & editing: Juan José Ramos-Álvarez, Roberto Murias-Lozano, Elena Sonsoles Rodríguez-López." ]
[ "intro", null, null, null, null, null, null, "results", null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, "discussion", null, null, null ]
[ "invasive physiotherapy", "percutaneous electrical nerve stimulation", "percutaneous neuromodulation", "PNM" ]
MRI changes of adjacent segments after transforaminal lumbar interbody fusion (TLIF) and foraminal endoscopy: A case-control study.
36254062
Intervertebral foramen endoscopy has developed rapidly, but compared with transforaminal lumbar interbody fusion (TLIF), the progress of degeneration is unknown. We aim to compare the changes of intervertebral disc and intervertebral foramen in adjacent segments after TLIF and endoscopic discectomy for patients with lumbar disc herniation (LDH).
BACKGROUND
From 2014 to 2017, 87 patients who were diagnosed with single-level LDH and received surgery of TLIF (group T, n = 43) or endoscopic discectomy (group F, n = 44) were retrospectively analyzed. X-ray, MRI, CT and clinical symptoms were recorded before operation and at the last follow-up (FU). The neurological function was originally evaluated by the Japanese Orthopaedic Association (JOA) scores. Radiological evaluation included the height of intervertebral space (HIS), intervertebral foramen height (FH), intervertebral foramen area (FA), lumbar lordosis (CA) and intervertebral disc degeneration Pfirrmann scores.
METHODS
There was no significant difference in baseline characteristics, JOA improvement rate, reoperation rate and complications between the two groups. The age, average blood loss, average hospital stays and average operation time in group F were lower than those in group T. During the last FU, HIS, CA and FA decreased in both groups, and the changes in group T were more significant than those in group F (P < .05). There was no significant difference in FH changes between the two groups (P > .05).
RESULTS
Both TLIF and endoscopic surgery can achieve good results in the treatment of LDH, but the risk of lumbar disc height loss and intervertebral foramina reduction in the adjacent segment after endoscopic surgery is lower.
CONCLUSION
[ "Humans", "Case-Control Studies", "Endoscopy", "Intervertebral Disc Degeneration", "Intervertebral Disc Displacement", "Lumbar Vertebrae", "Magnetic Resonance Imaging", "Retrospective Studies", "Spinal Fusion", "Treatment Outcome" ]
9575806
1. Introduction
Disc changes in the adjacent segment after lumbar fusion has aroused full attention, and the relationship between the two has been reported in many literatures.[1] adjacent segmental degeneration (ASD) is defined as degeneration of adjacent intervertebral disc after surgery by observation on imaging, regardless of symptoms.[2] In recent years, endoscopic technology has been widely used in clinic. Compared with open surgery, for example transforaminal lumbar interbody fusion (TLIF), endoscopic surgery has advantages of less trauma, less postoperative pain and a more rapid recovery, and the curative effect is equivalent to that of open surgery.[3,4] However, there is a lack of systematic comparative study focusing on ASD after endoscopic surgery and TLIF. Although various novel operations may help to attenuate ASD (especially for motion-preserving surgery),[1,5] their efficacies are still controversial. The purpose of this study was to compare the effects of endoscopic discectomy and TLIF on the changes of adjacent segmental disc height and foraminal changes, to review the literature to analyze its risk factors, and to raise awareness of ASD.
2.2. Surgical procedure for LDH
TLIF (T group): endotracheal intubation under general anesthesia, the patient took a prone position and raised the waist bridge. A 10‐12 cm median incision was performed and peeled off layer by layer along the bilateral sub-periosteum of the spinous process. Fully expose the lamina and articular process. The inferior articular process and part of the upper joint. Chisel off the process and hyperplastic osteophyte and bite off the ligamentum flavum, such as bilateral symptoms. If it is heavy, the contralateral decompression is carried out in the same way. Cage rack placement and connection of titanium. Place cage rack and connect titanium. Indwelling 1 negative pressure drainage tube. Suture incision layer by layer. Foraminal endoscopic discectomy (F group): prone position, chest and ilium cushion soft pillow raised to make the abdomen empty, fully expand the intervertebral foramen and reduce the intervertebral foramen plastic operation. Determine the puncture path: Mark the outline of the ilium and determine the hand under fluoroscopy. The operative segment, and then determine the puncture distance according to the patient’s body size, in order to match the vertebrae. The horizontal gap is marked by a diagonal line with an angle of about 30°, and the puncture point is the line and the distance. The point of intersection of parallel lines at a predetermined distance from the rear median line. Disinfect and spread towels. After that, the local anesthetic diluted to 1% was applied to the skin and subcutaneous of the puncture point. Fascia infiltration anesthesia, and then the 18G puncture needle was punctured slowly until there was obvious obstruction force, that is, at the fascia of the lumbar dorsal muscle, the puncture needle is slightly retracted and blocked by local anesthesia. Continue to deepen the puncture needle to the tip of the superior articular process and replace 0.5% lidocaine. Due to the anesthesia of the facet joint, the puncture needle was withdrawn slightly to increase and deepen the tilt of the head. Through the safety triangle puncture along the direction of the spinal canal, it is confirmed that the needle tip is located in the right position. The midline of the spinous process is connected with the posterior edge of the vertebral body laterally. After cutting the skin with a sharp knife, insert it. Enter the guide wire, then use the step-by-step sleeve to expand the soft tissue, and then the fourth-stage ring. Saw to enlarge the intervertebral foramen step by step (each step is done under fluoroscopy, ring saw. Do not exceed the inner edge of the pedicle), and finally the working sleeve is placed smoothly and the fluoroscopy is accurate. It is recognized that it is located at the predetermined target position. Turn on the imaging system and carefully identify. Microscopic structure, separation and adhesion, removal of protruding nucleus pulposus tissue, surrounding. Decompression of the walking nerve root and detection of the pressure of the superior exit nerve root. Until the nerve root pulses with the pulse can be seen under the microscope, and the fibers are treated by radiofrequency thermocoagulation. The ring is formed, and the skin is sutured after careful hemostasis.
3. Results
A total of 87 patients were enrolled in our study the patients were divided into F group (n = 44) and T group(n = 43), including 24 males and 20 females in Foraminal group with an average age of 51.75 ± 3.65 years and 25 males and 18 females in TLIF group with an average age of 53.89 ± 5.21 years. The demographic characteristics of patients were summarized in Table 1 and baseline characteristics were well balanced between the two groups, including gender, and basic physical condition. But the age, blood loss, operation time, length of stays in group F were lower than those in group T(P < .05). Baseline demographic information of patients with LDH. LDH = lumbar disc herniation. The date of the height of intervertebral space (HIS) is summarized in Table 2. Before surgery, there was no statistical significance between group F and group T regarding AH, MH, and PH. but the mean HIS was significantly higher in group T. At the final FU the mean HIS decreased in two group (P < .05). Noticeably, the change of AH, MH, PH and average height in group F was all lower than that in group T (all P < .05) (Fig. 1). Change of the HIS of patients in the two groups. ADH = anterior disc height, MDH = middle disc height, PDH = posterior disc height; the average value of the three heights. #Comparison of parameters between the two groups. P < .05, comparison of parameters within the same groups before surgery and final FU. Change of the HIS of patients in the two groups HIS = height of intervertebral space. Table 3 demonstrates the changes of imaging outcomes between the two groups. We did not find any statistical difference in the comparison of FH, Pfirrmann scores, CA and FA within the two groups before surgery (all P > .05). However, the change of Pfirrmann scores, CA, and FA was significantly larger in group T at the last FU (P < .05). Although there is a difference of the reduction of FH was no significant difference (Fig. 2). Imaging outcomes of patients in the two groups. PS = Pfirrmann score, CA = Cobb angle, FH = foraminal height, FA = foraminal area. Comparison of parameters between the two groups. P < .05, comparison of parameters within the same groups before surgery and final FU. Imaging outcomes of patients in the two groups. The clinical results were summarized in Table 4. The JOA scores improved from 14.90 ± 2.20 to 24.12 ± 2.40 in F group, and 14.55 ± 1.98 to 25.71 ± 2.12 in T group. No significant difference was observed in JOA scores between the two groups at the final FU. There were 40 (90.90%) patients who acquired significant alleviation of original symptoms in group F, whereas in group T, 41(95.35%) patients had symptom alleviation. No difference was observed between the peri-operative complications, three patients in group T experienced surgery-related complications: one with lumbar hematoma, one with surgery site infection and one with cerebrospinal fluid leakage. In group F, the number of patients with complications was one: one with Cerebrospinal fluid leakage. all patients received timely symptomatic treatment and all were cured. The re-operation rates were 4.55% (2/44) in F group (two patients underwent open surgery because of the protruding of the operative segment.), and 2.33% (1/43) in T group (One patient underwent endoscopic revision because of ASD) at the final FU. Typical case: Figure 3. Clinical outcomes of patients in the two groups. Typical case: A 50-year-old male patient presented with recurrent low back pain for 1 year, which was aggravated by numbness, pain and fatigue of the right lower limb for more than 1 month. Diagnosis: lumbar disc herniation (L5/S1). The pain symptoms after L5/S1 lumbar intervertebral discectomy under foraminal endoscopy were significantly relieved, and the improvement rate of JOA in the last FU was 79.23%. The changes of intervertebral disc height, intervertebral foramen height and area were not obvious. Preoperative magnetic resonance (ACE) final follow-up (BDF)).
null
null
[ "2. Methods", "2.1. Patients’ population", "2.3. Clinical and radiological assessment", "2.4. Statistical analysis", "4.1. Limitations", "5. Conclusion", "Author contributions" ]
[ "The study has been approved by the hospital’s board of directors and informed consent through institutional review, including details of the operation, including treatment mechanisms, predicted outcomes, and potential risks and adverse effects.\n[SUBTITLE] 2.1. Patients’ population [SUBSECTION] Patients diagnosed with single-segment LDH and undergoing TLIF or endoscopic discectomy at our institution from June 2014 to June 2017 were included in this study. Inclusion: clinical diagnosis of L5/S1 LDH and complete imaging data. Exclusion: multi-segmental LDH, scoliosis, fracture, slippage and other lumbar spine diseases. (2) Unable to undergo surgery. (3) History of previous lumbar spine surgery. Group F: nerve compression leading to symptoms such as low back pain and intermittent claudication; ineffective conservative treatment: lumbar disc herniation (LDH), prolapse, free: symptoms cannot be relieved and continue to worsen: LDH with lateral saphenous fossa or local spinal stenosis, etc. Group T: the above criteria were accompanied by severe muscle weakness, foot drop, cauda equina syndrome, etc. endoscopic discectomy was difficult. Patients with osteoporosis in both groups continued pharmacological treatment.\nPatients diagnosed with single-segment LDH and undergoing TLIF or endoscopic discectomy at our institution from June 2014 to June 2017 were included in this study. Inclusion: clinical diagnosis of L5/S1 LDH and complete imaging data. Exclusion: multi-segmental LDH, scoliosis, fracture, slippage and other lumbar spine diseases. (2) Unable to undergo surgery. (3) History of previous lumbar spine surgery. Group F: nerve compression leading to symptoms such as low back pain and intermittent claudication; ineffective conservative treatment: lumbar disc herniation (LDH), prolapse, free: symptoms cannot be relieved and continue to worsen: LDH with lateral saphenous fossa or local spinal stenosis, etc. Group T: the above criteria were accompanied by severe muscle weakness, foot drop, cauda equina syndrome, etc. endoscopic discectomy was difficult. Patients with osteoporosis in both groups continued pharmacological treatment.\n[SUBTITLE] 2.2. Surgical procedure for LDH [SUBSECTION] TLIF (T group): endotracheal intubation under general anesthesia, the patient took a prone position and raised the waist bridge. A 10‐12 cm median incision was performed and peeled off layer by layer along the bilateral sub-periosteum of the spinous process.\nFully expose the lamina and articular process. The inferior articular process and part of the upper joint. Chisel off the process and hyperplastic osteophyte and bite off the ligamentum flavum, such as bilateral symptoms. If it is heavy, the contralateral decompression is carried out in the same way. Cage rack placement and connection of titanium. Place cage rack and connect titanium. Indwelling 1 negative pressure drainage tube. Suture incision layer by layer.\nForaminal endoscopic discectomy (F group): prone position, chest and ilium cushion soft pillow raised to make the abdomen empty, fully expand the intervertebral foramen and reduce the intervertebral foramen plastic operation. Determine the puncture path: Mark the outline of the ilium and determine the hand under fluoroscopy. The operative segment, and then determine the puncture distance according to the patient’s body size, in order to match the vertebrae. The horizontal gap is marked by a diagonal line with an angle of about 30°, and the puncture point is the line and the distance. The point of intersection of parallel lines at a predetermined distance from the rear median line. Disinfect and spread towels. After that, the local anesthetic diluted to 1% was applied to the skin and subcutaneous of the puncture point. Fascia infiltration anesthesia, and then the 18G puncture needle was punctured slowly until there was obvious obstruction force, that is, at the fascia of the lumbar dorsal muscle, the puncture needle is slightly retracted and blocked by local anesthesia. Continue to deepen the puncture needle to the tip of the superior articular process and replace 0.5% lidocaine. Due to the anesthesia of the facet joint, the puncture needle was withdrawn slightly to increase and deepen the tilt of the head. Through the safety triangle puncture along the direction of the spinal canal, it is confirmed that the needle tip is located in the right position. The midline of the spinous process is connected with the posterior edge of the vertebral body laterally. After cutting the skin with a sharp knife, insert it. Enter the guide wire, then use the step-by-step sleeve to expand the soft tissue, and then the fourth-stage ring. Saw to enlarge the intervertebral foramen step by step (each step is done under fluoroscopy, ring saw. Do not exceed the inner edge of the pedicle), and finally the working sleeve is placed smoothly and the fluoroscopy is accurate. It is recognized that it is located at the predetermined target position. Turn on the imaging system and carefully identify. Microscopic structure, separation and adhesion, removal of protruding nucleus pulposus tissue, surrounding. Decompression of the walking nerve root and detection of the pressure of the superior exit nerve root. Until the nerve root pulses with the pulse can be seen under the microscope, and the fibers are treated by radiofrequency thermocoagulation. The ring is formed, and the skin is sutured after careful hemostasis.\nTLIF (T group): endotracheal intubation under general anesthesia, the patient took a prone position and raised the waist bridge. A 10‐12 cm median incision was performed and peeled off layer by layer along the bilateral sub-periosteum of the spinous process.\nFully expose the lamina and articular process. The inferior articular process and part of the upper joint. Chisel off the process and hyperplastic osteophyte and bite off the ligamentum flavum, such as bilateral symptoms. If it is heavy, the contralateral decompression is carried out in the same way. Cage rack placement and connection of titanium. Place cage rack and connect titanium. Indwelling 1 negative pressure drainage tube. Suture incision layer by layer.\nForaminal endoscopic discectomy (F group): prone position, chest and ilium cushion soft pillow raised to make the abdomen empty, fully expand the intervertebral foramen and reduce the intervertebral foramen plastic operation. Determine the puncture path: Mark the outline of the ilium and determine the hand under fluoroscopy. The operative segment, and then determine the puncture distance according to the patient’s body size, in order to match the vertebrae. The horizontal gap is marked by a diagonal line with an angle of about 30°, and the puncture point is the line and the distance. The point of intersection of parallel lines at a predetermined distance from the rear median line. Disinfect and spread towels. After that, the local anesthetic diluted to 1% was applied to the skin and subcutaneous of the puncture point. Fascia infiltration anesthesia, and then the 18G puncture needle was punctured slowly until there was obvious obstruction force, that is, at the fascia of the lumbar dorsal muscle, the puncture needle is slightly retracted and blocked by local anesthesia. Continue to deepen the puncture needle to the tip of the superior articular process and replace 0.5% lidocaine. Due to the anesthesia of the facet joint, the puncture needle was withdrawn slightly to increase and deepen the tilt of the head. Through the safety triangle puncture along the direction of the spinal canal, it is confirmed that the needle tip is located in the right position. The midline of the spinous process is connected with the posterior edge of the vertebral body laterally. After cutting the skin with a sharp knife, insert it. Enter the guide wire, then use the step-by-step sleeve to expand the soft tissue, and then the fourth-stage ring. Saw to enlarge the intervertebral foramen step by step (each step is done under fluoroscopy, ring saw. Do not exceed the inner edge of the pedicle), and finally the working sleeve is placed smoothly and the fluoroscopy is accurate. It is recognized that it is located at the predetermined target position. Turn on the imaging system and carefully identify. Microscopic structure, separation and adhesion, removal of protruding nucleus pulposus tissue, surrounding. Decompression of the walking nerve root and detection of the pressure of the superior exit nerve root. Until the nerve root pulses with the pulse can be seen under the microscope, and the fibers are treated by radiofrequency thermocoagulation. The ring is formed, and the skin is sutured after careful hemostasis.\n[SUBTITLE] 2.3. Clinical and radiological assessment [SUBSECTION] Demographics information including age, gender, duration of symptoms, body mass index, osteoporosis, blood loss, operation time and length of stay were evaluated to between groups. Patients were followed up for at least 36 months after surgery.\nRadiologic data include the following parameters:\nThe Cobb angle (CA) of the whole lumbar lordosis: the angle between the line at the upper endplate of L1 and the upper endplate of S1.\nCross-sectional area (FA) and height (FH) of intervertebral foramen: on the sagittal section of the intervertebral foramen, the line around the corresponding intervertebral foramen on the sagittal section forms an area and the height of the upper and lower edges.[6]\nThe height of the anterior intervertebral space (AH).\nThe height of the middle intervertebral space (MH).\nThe height of the posterior intervertebral space (PH).\nPfirrmann grade: intervertebral disc degeneration was evaluated by Pfirrmann grade.\nNervous system function was obtained using Japanese Orthopedic Association Scores System (JOA Scores).\nAlleviation of original symptoms, re-operation rates, complications were counted.\nThe data of preoperative and last follow-up (FU) were measured in all patients.\nDemographics information including age, gender, duration of symptoms, body mass index, osteoporosis, blood loss, operation time and length of stay were evaluated to between groups. Patients were followed up for at least 36 months after surgery.\nRadiologic data include the following parameters:\nThe Cobb angle (CA) of the whole lumbar lordosis: the angle between the line at the upper endplate of L1 and the upper endplate of S1.\nCross-sectional area (FA) and height (FH) of intervertebral foramen: on the sagittal section of the intervertebral foramen, the line around the corresponding intervertebral foramen on the sagittal section forms an area and the height of the upper and lower edges.[6]\nThe height of the anterior intervertebral space (AH).\nThe height of the middle intervertebral space (MH).\nThe height of the posterior intervertebral space (PH).\nPfirrmann grade: intervertebral disc degeneration was evaluated by Pfirrmann grade.\nNervous system function was obtained using Japanese Orthopedic Association Scores System (JOA Scores).\nAlleviation of original symptoms, re-operation rates, complications were counted.\nThe data of preoperative and last follow-up (FU) were measured in all patients.\n[SUBTITLE] 2.4. Statistical analysis [SUBSECTION] Statistical analysis was performed using the Statistical Package for the Social Sciences version 23.0 (IBM Armonls, NY, USA). Continuous variables were recorded as mean values ± standard deviation (SD), and categorical variables were expressed by proportions (%). The unpaired 2-tailed Student t test or Mann–Whitney U test were performed to compare the mean values or data distribution of continuous variables. And categorical variables were compared with the χ2 (Chi-square) test or Fisher exact test, as appropriate. The data measured before and after the last FU were statistically analyzed. Paired t-test was used for the last comparison before operation, and independent sample t-test was used for the comparison between groups. A P value of <.05 was considered statistically significant.\nStatistical analysis was performed using the Statistical Package for the Social Sciences version 23.0 (IBM Armonls, NY, USA). Continuous variables were recorded as mean values ± standard deviation (SD), and categorical variables were expressed by proportions (%). The unpaired 2-tailed Student t test or Mann–Whitney U test were performed to compare the mean values or data distribution of continuous variables. And categorical variables were compared with the χ2 (Chi-square) test or Fisher exact test, as appropriate. The data measured before and after the last FU were statistically analyzed. Paired t-test was used for the last comparison before operation, and independent sample t-test was used for the comparison between groups. A P value of <.05 was considered statistically significant.", "Patients diagnosed with single-segment LDH and undergoing TLIF or endoscopic discectomy at our institution from June 2014 to June 2017 were included in this study. Inclusion: clinical diagnosis of L5/S1 LDH and complete imaging data. Exclusion: multi-segmental LDH, scoliosis, fracture, slippage and other lumbar spine diseases. (2) Unable to undergo surgery. (3) History of previous lumbar spine surgery. Group F: nerve compression leading to symptoms such as low back pain and intermittent claudication; ineffective conservative treatment: lumbar disc herniation (LDH), prolapse, free: symptoms cannot be relieved and continue to worsen: LDH with lateral saphenous fossa or local spinal stenosis, etc. Group T: the above criteria were accompanied by severe muscle weakness, foot drop, cauda equina syndrome, etc. endoscopic discectomy was difficult. Patients with osteoporosis in both groups continued pharmacological treatment.", "Demographics information including age, gender, duration of symptoms, body mass index, osteoporosis, blood loss, operation time and length of stay were evaluated to between groups. Patients were followed up for at least 36 months after surgery.\nRadiologic data include the following parameters:\nThe Cobb angle (CA) of the whole lumbar lordosis: the angle between the line at the upper endplate of L1 and the upper endplate of S1.\nCross-sectional area (FA) and height (FH) of intervertebral foramen: on the sagittal section of the intervertebral foramen, the line around the corresponding intervertebral foramen on the sagittal section forms an area and the height of the upper and lower edges.[6]\nThe height of the anterior intervertebral space (AH).\nThe height of the middle intervertebral space (MH).\nThe height of the posterior intervertebral space (PH).\nPfirrmann grade: intervertebral disc degeneration was evaluated by Pfirrmann grade.\nNervous system function was obtained using Japanese Orthopedic Association Scores System (JOA Scores).\nAlleviation of original symptoms, re-operation rates, complications were counted.\nThe data of preoperative and last follow-up (FU) were measured in all patients.", "Statistical analysis was performed using the Statistical Package for the Social Sciences version 23.0 (IBM Armonls, NY, USA). Continuous variables were recorded as mean values ± standard deviation (SD), and categorical variables were expressed by proportions (%). The unpaired 2-tailed Student t test or Mann–Whitney U test were performed to compare the mean values or data distribution of continuous variables. And categorical variables were compared with the χ2 (Chi-square) test or Fisher exact test, as appropriate. The data measured before and after the last FU were statistically analyzed. Paired t-test was used for the last comparison before operation, and independent sample t-test was used for the comparison between groups. A P value of <.05 was considered statistically significant.", "There may be differences in baseline characteristics of the preoperative population that affect the accuracy of this study. In addition, this is a small sample and short-term FU imaging measurement study that requires a large population, prospective study to analyze changes in adjacent segments with conservatively treated controls. Finally, the same team of physicians responsible for standardized procedures to assess FU symptoms and independent measurements of imaging parameters was blinded to this study.", "Both TLIF and endoscopic surgery can achieve good results in the treatment of LDH, but the risk of lumbar disc height loss and intervertebral foramina reduction in the adjacent segment after intervertebral foraminal surgery is lower.", "SW analyzed and interpreted the patient data, and was a major contributor in writing the manuscript. DY and GZ performed the examination of the data, and substantively revised the manuscript, conducted the acquisition of data. JC and FZ proposed the idea of study design. JS and RY finished the final assessment of the manuscript. All authors read and approved the final manuscript.\nData curation: Deyu Yang, Feng Zhao, Shunmin Wang.\nFormal analysis: Shunmin Wang.\nFunding acquisition: Gengyang Zheng, Jiangang Shi, Ruijin You.\nInvestigation: Gengyang Zheng, Shunmin Wang.\nMethodology: Ruijin You, Shunmin Wang.\nSoftware: Ruijin You.\nSupervision: Deyu Yang, Jiangang Shi, Jie Cao.\nProject administration: Shunmin Wang.\nValidation: Gengyang Zheng, Jie Cao, Ruijin You.\nWriting – original draft: Shunmin Wang.\nWriting – review &amp; editing: Jiangang Shi, Jie Cao, Shunmin Wang." ]
[ "methods", null, null, null, null, null, null ]
[ "1. Introduction", "2. Methods", "2.1. Patients’ population", "2.2. Surgical procedure for LDH", "2.3. Clinical and radiological assessment", "2.4. Statistical analysis", "3. Results", "4. Discussion", "4.1. Limitations", "5. Conclusion", "Author contributions" ]
[ "Disc changes in the adjacent segment after lumbar fusion has aroused full attention, and the relationship between the two has been reported in many literatures.[1] adjacent segmental degeneration (ASD) is defined as degeneration of adjacent intervertebral disc after surgery by observation on imaging, regardless of symptoms.[2] In recent years, endoscopic technology has been widely used in clinic. Compared with open surgery, for example transforaminal lumbar interbody fusion (TLIF), endoscopic surgery has advantages of less trauma, less postoperative pain and a more rapid recovery, and the curative effect is equivalent to that of open surgery.[3,4] However, there is a lack of systematic comparative study focusing on ASD after endoscopic surgery and TLIF. Although various novel operations may help to attenuate ASD (especially for motion-preserving surgery),[1,5] their efficacies are still controversial. The purpose of this study was to compare the effects of endoscopic discectomy and TLIF on the changes of adjacent segmental disc height and foraminal changes, to review the literature to analyze its risk factors, and to raise awareness of ASD.", "The study has been approved by the hospital’s board of directors and informed consent through institutional review, including details of the operation, including treatment mechanisms, predicted outcomes, and potential risks and adverse effects.\n[SUBTITLE] 2.1. Patients’ population [SUBSECTION] Patients diagnosed with single-segment LDH and undergoing TLIF or endoscopic discectomy at our institution from June 2014 to June 2017 were included in this study. Inclusion: clinical diagnosis of L5/S1 LDH and complete imaging data. Exclusion: multi-segmental LDH, scoliosis, fracture, slippage and other lumbar spine diseases. (2) Unable to undergo surgery. (3) History of previous lumbar spine surgery. Group F: nerve compression leading to symptoms such as low back pain and intermittent claudication; ineffective conservative treatment: lumbar disc herniation (LDH), prolapse, free: symptoms cannot be relieved and continue to worsen: LDH with lateral saphenous fossa or local spinal stenosis, etc. Group T: the above criteria were accompanied by severe muscle weakness, foot drop, cauda equina syndrome, etc. endoscopic discectomy was difficult. Patients with osteoporosis in both groups continued pharmacological treatment.\nPatients diagnosed with single-segment LDH and undergoing TLIF or endoscopic discectomy at our institution from June 2014 to June 2017 were included in this study. Inclusion: clinical diagnosis of L5/S1 LDH and complete imaging data. Exclusion: multi-segmental LDH, scoliosis, fracture, slippage and other lumbar spine diseases. (2) Unable to undergo surgery. (3) History of previous lumbar spine surgery. Group F: nerve compression leading to symptoms such as low back pain and intermittent claudication; ineffective conservative treatment: lumbar disc herniation (LDH), prolapse, free: symptoms cannot be relieved and continue to worsen: LDH with lateral saphenous fossa or local spinal stenosis, etc. Group T: the above criteria were accompanied by severe muscle weakness, foot drop, cauda equina syndrome, etc. endoscopic discectomy was difficult. Patients with osteoporosis in both groups continued pharmacological treatment.\n[SUBTITLE] 2.2. Surgical procedure for LDH [SUBSECTION] TLIF (T group): endotracheal intubation under general anesthesia, the patient took a prone position and raised the waist bridge. A 10‐12 cm median incision was performed and peeled off layer by layer along the bilateral sub-periosteum of the spinous process.\nFully expose the lamina and articular process. The inferior articular process and part of the upper joint. Chisel off the process and hyperplastic osteophyte and bite off the ligamentum flavum, such as bilateral symptoms. If it is heavy, the contralateral decompression is carried out in the same way. Cage rack placement and connection of titanium. Place cage rack and connect titanium. Indwelling 1 negative pressure drainage tube. Suture incision layer by layer.\nForaminal endoscopic discectomy (F group): prone position, chest and ilium cushion soft pillow raised to make the abdomen empty, fully expand the intervertebral foramen and reduce the intervertebral foramen plastic operation. Determine the puncture path: Mark the outline of the ilium and determine the hand under fluoroscopy. The operative segment, and then determine the puncture distance according to the patient’s body size, in order to match the vertebrae. The horizontal gap is marked by a diagonal line with an angle of about 30°, and the puncture point is the line and the distance. The point of intersection of parallel lines at a predetermined distance from the rear median line. Disinfect and spread towels. After that, the local anesthetic diluted to 1% was applied to the skin and subcutaneous of the puncture point. Fascia infiltration anesthesia, and then the 18G puncture needle was punctured slowly until there was obvious obstruction force, that is, at the fascia of the lumbar dorsal muscle, the puncture needle is slightly retracted and blocked by local anesthesia. Continue to deepen the puncture needle to the tip of the superior articular process and replace 0.5% lidocaine. Due to the anesthesia of the facet joint, the puncture needle was withdrawn slightly to increase and deepen the tilt of the head. Through the safety triangle puncture along the direction of the spinal canal, it is confirmed that the needle tip is located in the right position. The midline of the spinous process is connected with the posterior edge of the vertebral body laterally. After cutting the skin with a sharp knife, insert it. Enter the guide wire, then use the step-by-step sleeve to expand the soft tissue, and then the fourth-stage ring. Saw to enlarge the intervertebral foramen step by step (each step is done under fluoroscopy, ring saw. Do not exceed the inner edge of the pedicle), and finally the working sleeve is placed smoothly and the fluoroscopy is accurate. It is recognized that it is located at the predetermined target position. Turn on the imaging system and carefully identify. Microscopic structure, separation and adhesion, removal of protruding nucleus pulposus tissue, surrounding. Decompression of the walking nerve root and detection of the pressure of the superior exit nerve root. Until the nerve root pulses with the pulse can be seen under the microscope, and the fibers are treated by radiofrequency thermocoagulation. The ring is formed, and the skin is sutured after careful hemostasis.\nTLIF (T group): endotracheal intubation under general anesthesia, the patient took a prone position and raised the waist bridge. A 10‐12 cm median incision was performed and peeled off layer by layer along the bilateral sub-periosteum of the spinous process.\nFully expose the lamina and articular process. The inferior articular process and part of the upper joint. Chisel off the process and hyperplastic osteophyte and bite off the ligamentum flavum, such as bilateral symptoms. If it is heavy, the contralateral decompression is carried out in the same way. Cage rack placement and connection of titanium. Place cage rack and connect titanium. Indwelling 1 negative pressure drainage tube. Suture incision layer by layer.\nForaminal endoscopic discectomy (F group): prone position, chest and ilium cushion soft pillow raised to make the abdomen empty, fully expand the intervertebral foramen and reduce the intervertebral foramen plastic operation. Determine the puncture path: Mark the outline of the ilium and determine the hand under fluoroscopy. The operative segment, and then determine the puncture distance according to the patient’s body size, in order to match the vertebrae. The horizontal gap is marked by a diagonal line with an angle of about 30°, and the puncture point is the line and the distance. The point of intersection of parallel lines at a predetermined distance from the rear median line. Disinfect and spread towels. After that, the local anesthetic diluted to 1% was applied to the skin and subcutaneous of the puncture point. Fascia infiltration anesthesia, and then the 18G puncture needle was punctured slowly until there was obvious obstruction force, that is, at the fascia of the lumbar dorsal muscle, the puncture needle is slightly retracted and blocked by local anesthesia. Continue to deepen the puncture needle to the tip of the superior articular process and replace 0.5% lidocaine. Due to the anesthesia of the facet joint, the puncture needle was withdrawn slightly to increase and deepen the tilt of the head. Through the safety triangle puncture along the direction of the spinal canal, it is confirmed that the needle tip is located in the right position. The midline of the spinous process is connected with the posterior edge of the vertebral body laterally. After cutting the skin with a sharp knife, insert it. Enter the guide wire, then use the step-by-step sleeve to expand the soft tissue, and then the fourth-stage ring. Saw to enlarge the intervertebral foramen step by step (each step is done under fluoroscopy, ring saw. Do not exceed the inner edge of the pedicle), and finally the working sleeve is placed smoothly and the fluoroscopy is accurate. It is recognized that it is located at the predetermined target position. Turn on the imaging system and carefully identify. Microscopic structure, separation and adhesion, removal of protruding nucleus pulposus tissue, surrounding. Decompression of the walking nerve root and detection of the pressure of the superior exit nerve root. Until the nerve root pulses with the pulse can be seen under the microscope, and the fibers are treated by radiofrequency thermocoagulation. The ring is formed, and the skin is sutured after careful hemostasis.\n[SUBTITLE] 2.3. Clinical and radiological assessment [SUBSECTION] Demographics information including age, gender, duration of symptoms, body mass index, osteoporosis, blood loss, operation time and length of stay were evaluated to between groups. Patients were followed up for at least 36 months after surgery.\nRadiologic data include the following parameters:\nThe Cobb angle (CA) of the whole lumbar lordosis: the angle between the line at the upper endplate of L1 and the upper endplate of S1.\nCross-sectional area (FA) and height (FH) of intervertebral foramen: on the sagittal section of the intervertebral foramen, the line around the corresponding intervertebral foramen on the sagittal section forms an area and the height of the upper and lower edges.[6]\nThe height of the anterior intervertebral space (AH).\nThe height of the middle intervertebral space (MH).\nThe height of the posterior intervertebral space (PH).\nPfirrmann grade: intervertebral disc degeneration was evaluated by Pfirrmann grade.\nNervous system function was obtained using Japanese Orthopedic Association Scores System (JOA Scores).\nAlleviation of original symptoms, re-operation rates, complications were counted.\nThe data of preoperative and last follow-up (FU) were measured in all patients.\nDemographics information including age, gender, duration of symptoms, body mass index, osteoporosis, blood loss, operation time and length of stay were evaluated to between groups. Patients were followed up for at least 36 months after surgery.\nRadiologic data include the following parameters:\nThe Cobb angle (CA) of the whole lumbar lordosis: the angle between the line at the upper endplate of L1 and the upper endplate of S1.\nCross-sectional area (FA) and height (FH) of intervertebral foramen: on the sagittal section of the intervertebral foramen, the line around the corresponding intervertebral foramen on the sagittal section forms an area and the height of the upper and lower edges.[6]\nThe height of the anterior intervertebral space (AH).\nThe height of the middle intervertebral space (MH).\nThe height of the posterior intervertebral space (PH).\nPfirrmann grade: intervertebral disc degeneration was evaluated by Pfirrmann grade.\nNervous system function was obtained using Japanese Orthopedic Association Scores System (JOA Scores).\nAlleviation of original symptoms, re-operation rates, complications were counted.\nThe data of preoperative and last follow-up (FU) were measured in all patients.\n[SUBTITLE] 2.4. Statistical analysis [SUBSECTION] Statistical analysis was performed using the Statistical Package for the Social Sciences version 23.0 (IBM Armonls, NY, USA). Continuous variables were recorded as mean values ± standard deviation (SD), and categorical variables were expressed by proportions (%). The unpaired 2-tailed Student t test or Mann–Whitney U test were performed to compare the mean values or data distribution of continuous variables. And categorical variables were compared with the χ2 (Chi-square) test or Fisher exact test, as appropriate. The data measured before and after the last FU were statistically analyzed. Paired t-test was used for the last comparison before operation, and independent sample t-test was used for the comparison between groups. A P value of <.05 was considered statistically significant.\nStatistical analysis was performed using the Statistical Package for the Social Sciences version 23.0 (IBM Armonls, NY, USA). Continuous variables were recorded as mean values ± standard deviation (SD), and categorical variables were expressed by proportions (%). The unpaired 2-tailed Student t test or Mann–Whitney U test were performed to compare the mean values or data distribution of continuous variables. And categorical variables were compared with the χ2 (Chi-square) test or Fisher exact test, as appropriate. The data measured before and after the last FU were statistically analyzed. Paired t-test was used for the last comparison before operation, and independent sample t-test was used for the comparison between groups. A P value of <.05 was considered statistically significant.", "Patients diagnosed with single-segment LDH and undergoing TLIF or endoscopic discectomy at our institution from June 2014 to June 2017 were included in this study. Inclusion: clinical diagnosis of L5/S1 LDH and complete imaging data. Exclusion: multi-segmental LDH, scoliosis, fracture, slippage and other lumbar spine diseases. (2) Unable to undergo surgery. (3) History of previous lumbar spine surgery. Group F: nerve compression leading to symptoms such as low back pain and intermittent claudication; ineffective conservative treatment: lumbar disc herniation (LDH), prolapse, free: symptoms cannot be relieved and continue to worsen: LDH with lateral saphenous fossa or local spinal stenosis, etc. Group T: the above criteria were accompanied by severe muscle weakness, foot drop, cauda equina syndrome, etc. endoscopic discectomy was difficult. Patients with osteoporosis in both groups continued pharmacological treatment.", "TLIF (T group): endotracheal intubation under general anesthesia, the patient took a prone position and raised the waist bridge. A 10‐12 cm median incision was performed and peeled off layer by layer along the bilateral sub-periosteum of the spinous process.\nFully expose the lamina and articular process. The inferior articular process and part of the upper joint. Chisel off the process and hyperplastic osteophyte and bite off the ligamentum flavum, such as bilateral symptoms. If it is heavy, the contralateral decompression is carried out in the same way. Cage rack placement and connection of titanium. Place cage rack and connect titanium. Indwelling 1 negative pressure drainage tube. Suture incision layer by layer.\nForaminal endoscopic discectomy (F group): prone position, chest and ilium cushion soft pillow raised to make the abdomen empty, fully expand the intervertebral foramen and reduce the intervertebral foramen plastic operation. Determine the puncture path: Mark the outline of the ilium and determine the hand under fluoroscopy. The operative segment, and then determine the puncture distance according to the patient’s body size, in order to match the vertebrae. The horizontal gap is marked by a diagonal line with an angle of about 30°, and the puncture point is the line and the distance. The point of intersection of parallel lines at a predetermined distance from the rear median line. Disinfect and spread towels. After that, the local anesthetic diluted to 1% was applied to the skin and subcutaneous of the puncture point. Fascia infiltration anesthesia, and then the 18G puncture needle was punctured slowly until there was obvious obstruction force, that is, at the fascia of the lumbar dorsal muscle, the puncture needle is slightly retracted and blocked by local anesthesia. Continue to deepen the puncture needle to the tip of the superior articular process and replace 0.5% lidocaine. Due to the anesthesia of the facet joint, the puncture needle was withdrawn slightly to increase and deepen the tilt of the head. Through the safety triangle puncture along the direction of the spinal canal, it is confirmed that the needle tip is located in the right position. The midline of the spinous process is connected with the posterior edge of the vertebral body laterally. After cutting the skin with a sharp knife, insert it. Enter the guide wire, then use the step-by-step sleeve to expand the soft tissue, and then the fourth-stage ring. Saw to enlarge the intervertebral foramen step by step (each step is done under fluoroscopy, ring saw. Do not exceed the inner edge of the pedicle), and finally the working sleeve is placed smoothly and the fluoroscopy is accurate. It is recognized that it is located at the predetermined target position. Turn on the imaging system and carefully identify. Microscopic structure, separation and adhesion, removal of protruding nucleus pulposus tissue, surrounding. Decompression of the walking nerve root and detection of the pressure of the superior exit nerve root. Until the nerve root pulses with the pulse can be seen under the microscope, and the fibers are treated by radiofrequency thermocoagulation. The ring is formed, and the skin is sutured after careful hemostasis.", "Demographics information including age, gender, duration of symptoms, body mass index, osteoporosis, blood loss, operation time and length of stay were evaluated to between groups. Patients were followed up for at least 36 months after surgery.\nRadiologic data include the following parameters:\nThe Cobb angle (CA) of the whole lumbar lordosis: the angle between the line at the upper endplate of L1 and the upper endplate of S1.\nCross-sectional area (FA) and height (FH) of intervertebral foramen: on the sagittal section of the intervertebral foramen, the line around the corresponding intervertebral foramen on the sagittal section forms an area and the height of the upper and lower edges.[6]\nThe height of the anterior intervertebral space (AH).\nThe height of the middle intervertebral space (MH).\nThe height of the posterior intervertebral space (PH).\nPfirrmann grade: intervertebral disc degeneration was evaluated by Pfirrmann grade.\nNervous system function was obtained using Japanese Orthopedic Association Scores System (JOA Scores).\nAlleviation of original symptoms, re-operation rates, complications were counted.\nThe data of preoperative and last follow-up (FU) were measured in all patients.", "Statistical analysis was performed using the Statistical Package for the Social Sciences version 23.0 (IBM Armonls, NY, USA). Continuous variables were recorded as mean values ± standard deviation (SD), and categorical variables were expressed by proportions (%). The unpaired 2-tailed Student t test or Mann–Whitney U test were performed to compare the mean values or data distribution of continuous variables. And categorical variables were compared with the χ2 (Chi-square) test or Fisher exact test, as appropriate. The data measured before and after the last FU were statistically analyzed. Paired t-test was used for the last comparison before operation, and independent sample t-test was used for the comparison between groups. A P value of <.05 was considered statistically significant.", "A total of 87 patients were enrolled in our study the patients were divided into F group (n = 44) and T group(n = 43), including 24 males and 20 females in Foraminal group with an average age of 51.75 ± 3.65 years and 25 males and 18 females in TLIF group with an average age of 53.89 ± 5.21 years. The demographic characteristics of patients were summarized in Table 1 and baseline characteristics were well balanced between the two groups, including gender, and basic physical condition. But the age, blood loss, operation time, length of stays in group F were lower than those in group T(P < .05).\nBaseline demographic information of patients with LDH. LDH = lumbar disc herniation.\nThe date of the height of intervertebral space (HIS) is summarized in Table 2. Before surgery, there was no statistical significance between group F and group T regarding AH, MH, and PH. but the mean HIS was significantly higher in group T. At the final FU the mean HIS decreased in two group (P < .05). Noticeably, the change of AH, MH, PH and average height in group F was all lower than that in group T (all P < .05) (Fig. 1).\nChange of the HIS of patients in the two groups.\nADH = anterior disc height, MDH = middle disc height, PDH = posterior disc height; the average value of the three heights.\n#Comparison of parameters between the two groups.\nP < .05, comparison of parameters within the same groups before surgery and final FU.\nChange of the HIS of patients in the two groups HIS = height of intervertebral space.\nTable 3 demonstrates the changes of imaging outcomes between the two groups. We did not find any statistical difference in the comparison of FH, Pfirrmann scores, CA and FA within the two groups before surgery (all P > .05). However, the change of Pfirrmann scores, CA, and FA was significantly larger in group T at the last FU (P < .05). Although there is a difference of the reduction of FH was no significant difference (Fig. 2).\nImaging outcomes of patients in the two groups.\nPS = Pfirrmann score, CA = Cobb angle, FH = foraminal height, FA = foraminal area.\nComparison of parameters between the two groups.\nP < .05, comparison of parameters within the same groups before surgery and final FU.\nImaging outcomes of patients in the two groups.\nThe clinical results were summarized in Table 4. The JOA scores improved from 14.90 ± 2.20 to 24.12 ± 2.40 in F group, and 14.55 ± 1.98 to 25.71 ± 2.12 in T group. No significant difference was observed in JOA scores between the two groups at the final FU. There were 40 (90.90%) patients who acquired significant alleviation of original symptoms in group F, whereas in group T, 41(95.35%) patients had symptom alleviation. No difference was observed between the peri-operative complications, three patients in group T experienced surgery-related complications: one with lumbar hematoma, one with surgery site infection and one with cerebrospinal fluid leakage. In group F, the number of patients with complications was one: one with Cerebrospinal fluid leakage. all patients received timely symptomatic treatment and all were cured. The re-operation rates were 4.55% (2/44) in F group (two patients underwent open surgery because of the protruding of the operative segment.), and 2.33% (1/43) in T group (One patient underwent endoscopic revision because of ASD) at the final FU. Typical case: Figure 3.\nClinical outcomes of patients in the two groups.\nTypical case: A 50-year-old male patient presented with recurrent low back pain for 1 year, which was aggravated by numbness, pain and fatigue of the right lower limb for more than 1 month. Diagnosis: lumbar disc herniation (L5/S1). The pain symptoms after L5/S1 lumbar intervertebral discectomy under foraminal endoscopy were significantly relieved, and the improvement rate of JOA in the last FU was 79.23%. The changes of intervertebral disc height, intervertebral foramen height and area were not obvious. Preoperative magnetic resonance (ACE) final follow-up (BDF)).", "According to previous reports,[7] the incidence of ASD after lumbar fusion surgery and non-fusion surgery was 5‐77% (mean 26.6%) and 10% respectively. However, the[8,9] ASD after spinal fusion is considered to be multifactorial. Many literatures have reported that the degree of intervertebral disc degeneration was closely related to age (over 60 years old), genetic factors, high body mass index, preexisting stenosis or degeneration of adjacent segments, lumbar insufficiency, multi-segmental lumbar fixation and fusion.[10–12] Therefore, this study aimed to eliminating interference of these factors, and there is no statistical difference of these factors between two groups before operation. In addition, the operation itself is also one of the important reasons resulting in ASD. Ekman et al found that lumbar fusion accelerated ASD. after long-term FU.[13] Some scholars reported that the incidence of cephalic ASD examined by X-ray 2‐3 years after lumbar fixation and fusion was 38.5%.[14] This study also focuses on the cephalic adjacent intervertebral disc. Radcliff et al pointed out that the rate of ASD after fusion was significantly higher than that in patients without decompression,[15] and concluded that excessive distracting by the fusion cage to the intervertebral space was an important risk factor for ASD.[16] In a retrospective study, Biden et al suggested that floating fusion, in which the lower end of the fusion vertebra located at L5, is a risk factor for ASD.[9] In addition, floating fusion was more likely to develop ASD in patients with posterolateral lumbar fixation.[17]\nAlthough various reasons were attributed to ASD from the different views of many studies, the author speculates that surgery-related biomechanical changes of the spine are one of the most reasonable mechanisms.\nIn 1983, Kirkaldy‐Willis put forward the theory of three-joint complex (composed of intervertebral disc and two posterior facet joints), and believed that this structure plays an important role in maintaining the stability of the spine.[18] Liu et al, after six-year FU of patients accepting L4-5 fusion, found that the incidence of ASD was the highest in patients undergoing laminectomy.[19] Imagama et al followed up 52 patients after L4-5 laminectomy or L4-5 fenestration fusion for five years, revealing that patients with fenestration were less likely to develop ASD.[20] The results showed that the preservation of the structure of the posterior column of the lumbar spine is an important factor to avoid ASD. Lumbar fusion requires extensive peeling off of paraspinal muscles, removal of part of ligaments and bony structure, destruction of the stability of the three-joint complex, resulting in abnormal load distribution of the whole spine, prone to vertebral spondylolisthesis or fracture and other diseases.[21] Therefore, it changes the original equilibrium relationship between the diseased vertebral body and the adjacent vertebral body, and aggravates the postoperative ASD.[21–23] Ma et al found in the human cadaver model that the increase in stress on the facet joints after fusion may affect the degeneration of adjacent segments.[24] Through the analysis of three-dimensional finite element model, the biomechanical load of the adjacent vertebral facet joint above the fusion segment is obviously abnormal.[25,26]\nMakino at al reported that the incidence of ASD in 41 L4-5 PLIF patients with minimum intervertebral space distraction (12.2%) was significantly lower than that of previous ASD with PLIF distraction (31.8%).[27] It is considered that the use of a smaller fusion cage to minimize the opening of the intervertebral space may prevent ASD. In a biomechanical study of a finite element model fused at the L4/5 level, stress on the L3/4 endplate and intervertebral disc increased during flexion/extension movement.[28] In addition, in the cadaveric L3/4 fixation model, Cunningham et al observed an increase of pressure in the L2/3 intervertebral disc by 45% during flexion/straightening.[29] It can be seen that the cadaveric experiment showed that the pressure in the proximal intervertebral disc of the adjacent intervertebral disc increased to a fixed level.[29,30]\nTherefore, we think that the occurrence of ASD after fusion may be related to mechanical factors, the destruction and disorder of local structure, the range of motion of its upper adjacent segments and the compensatory load of facet joints.\nBecause the nucleus pulposus tissue is a colloidal semi-liquid substance with flow characteristics, the volume of the intervertebral disc will be further degraded and absorbed over time after nucleus pulposus resection.[31] Therefore, the removal of the nucleus pulposus of PTED (percutaneous transforaminal endoscopic discectomy) leads to the decrease of the bearing capacity of intervertebral disc, which in turn leads to the decrease of the upper vertebral body. At the last FU, the height of the intervertebral space in the upper adjacent segment was lower than that before operation, and there was statistical significance (P < .05). It may be related to the natural process of aging. However, compared with TLIF, PTED can not only retain more spinal range of motion, but also retain as much intervertebral disc tissue as possible on the basis of ensuring the curative effect, which provides a pathological basis for self-repair and secondary stability in the later stage, and may reduce the incidence of ASD or delay the occurrence of ASD.\nMany studies have shown that the decrease, disappearance or kyphosis of lumbar physiological curvature is closely related to the degeneration of intervertebral disc. Studies have shown that lumbar physiological curvature changes in patients with LDH may be the result of lumbar mechanical structural imbalance caused by lumbar degeneration.[32] Hypolordosis in the instrumented segment increased the load on the posterior pedicle device, posterior shear, and strain on the vertebral plate at the adjacent level. Biomechanical effects may explain the long-term consequences of degenerative changes after lumbar fusion.[33] The fusion of the lumbar spine in abnormal sagittal alignment and the loss of lumbar anterior convexity pre dispose the patient to degeneration of adjacent segments.[34] In this study, the changes of adjacent segments and CA in group F were significantly lower than those in group T, indicating that PTED can maintain physiological curvature and mechanical balance of spinal structure to some extent, and reduce the incidence of lumbar disc height loss.\nIntervertebral disc degeneration can directly and indirectly affect the area of intervertebral foramen. Cinotti et al found that intervertebral disc height loss can lead to intervertebral foramen stenosis by measuring 160 intervertebral foramen in dry cadaver specimens and 50 intervertebral foramina in fresh cadaveric spine.[35] In this study, the cross-sectional area of intervertebral foramen decreased before operation and at the last FU, and the change in group F was lower than that in group T. The stenosis of intervertebral space caused by intervertebral disc degeneration can significantly reduce the height of intervertebral foramen, especially the minimum sagittal diameter of intervertebral foramen. It may be due to the natural degeneration of the intervertebral disc or the change of posture during the examination of the patient.\nIn the past, many scholars have shown that the foraminal endoscope had a definite effect for LDH in early stage, and could significantly improve the pain symptoms.[36] In 588 patients with LDH treated by intervertebral foramen endoscopy and followed up for more than 2 years, the excellent and good recovery rate was 95.3%, and the recurrence rate was 3.6%.[37] Studies have reported that a small number of ASD patients can progress to symptomatic ASD, and their imaging findings are not necessarily correlated with symptoms after spinal fusion. Therefore, there was no significant difference in the JOA scores between the two groups at the last FU in this study.[38–40] At the same time, compared with open surgery, foraminal endoscopic surgery was performed under local anesthesia, the operator can observe patient’s feedback well, and there is no need to expose the herniated intervertebral disc by pulling the nerve root and dural sac during the operation, which reduces the risk of nerve injury.[41] Therefore, this study also found that minimally invasive surgery has a low complication rate and a low recurrence rate.\n[SUBTITLE] 4.1. Limitations [SUBSECTION] There may be differences in baseline characteristics of the preoperative population that affect the accuracy of this study. In addition, this is a small sample and short-term FU imaging measurement study that requires a large population, prospective study to analyze changes in adjacent segments with conservatively treated controls. Finally, the same team of physicians responsible for standardized procedures to assess FU symptoms and independent measurements of imaging parameters was blinded to this study.\nThere may be differences in baseline characteristics of the preoperative population that affect the accuracy of this study. In addition, this is a small sample and short-term FU imaging measurement study that requires a large population, prospective study to analyze changes in adjacent segments with conservatively treated controls. Finally, the same team of physicians responsible for standardized procedures to assess FU symptoms and independent measurements of imaging parameters was blinded to this study.", "There may be differences in baseline characteristics of the preoperative population that affect the accuracy of this study. In addition, this is a small sample and short-term FU imaging measurement study that requires a large population, prospective study to analyze changes in adjacent segments with conservatively treated controls. Finally, the same team of physicians responsible for standardized procedures to assess FU symptoms and independent measurements of imaging parameters was blinded to this study.", "Both TLIF and endoscopic surgery can achieve good results in the treatment of LDH, but the risk of lumbar disc height loss and intervertebral foramina reduction in the adjacent segment after intervertebral foraminal surgery is lower.", "SW analyzed and interpreted the patient data, and was a major contributor in writing the manuscript. DY and GZ performed the examination of the data, and substantively revised the manuscript, conducted the acquisition of data. JC and FZ proposed the idea of study design. JS and RY finished the final assessment of the manuscript. All authors read and approved the final manuscript.\nData curation: Deyu Yang, Feng Zhao, Shunmin Wang.\nFormal analysis: Shunmin Wang.\nFunding acquisition: Gengyang Zheng, Jiangang Shi, Ruijin You.\nInvestigation: Gengyang Zheng, Shunmin Wang.\nMethodology: Ruijin You, Shunmin Wang.\nSoftware: Ruijin You.\nSupervision: Deyu Yang, Jiangang Shi, Jie Cao.\nProject administration: Shunmin Wang.\nValidation: Gengyang Zheng, Jie Cao, Ruijin You.\nWriting – original draft: Shunmin Wang.\nWriting – review &amp; editing: Jiangang Shi, Jie Cao, Shunmin Wang." ]
[ "intro", "methods", null, "methods", null, null, "results", "discussion", null, null, null ]
[ "adjacent segment degeneration", "endoscopic surgery", "LDH", "TLIF" ]
The clinical efficacy of Shenmai injection in the prophylaxis and treatment of intradialytic hypotension: A protocol for systematic review and meta-analysis.
36254066
Intradialytic hypotension (IDH) is a common complication in hemodialysis. IDH can induce vomiting, chest tightness and syncope, and hemodialysis shall be discontinued in patients with severe IDH. As is revealed in related studies, Shenmai injection (SMI) can be used in the prophylaxis and treatment of IDH. However, there is still a lack of consensus about the efficacy among reported studies, which cannot provide compelling evidence. Therefore, a meta-analysis was conducted in this study to further investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH.
BACKGROUND
PubMed, Web of Science, Scopus, Cochrane Library, Embase, China Scientific Journal Database, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, and Wanfang Data were systematically retrieved from their establishment to June 2022. Subsequently, literature screening, data extraction, quality evaluation and cross-checking of results were performed according to the Cochrane Handbook. Besides, a meta-analysis was performed with the assistance of Revman 5.3 software.
METHODS
This study will evaluate whether SMI is effective in the prophylaxis and treatment of IDH.
RESULTS
The latest evidence for the efficacy and safety of SMI in the prevention and treatment of IDH can be provided through this study.
CONCLUSIONS
[ "Drug Combinations", "Drugs, Chinese Herbal", "Humans", "Hypotension", "Meta-Analysis as Topic", "Research Design", "Systematic Reviews as Topic", "Treatment Outcome" ]
9575838
1. Introduction
Intradialytic hypotension (IDH) is one of the common clinical complications in patients receiving maintenance hemodialysis, with a high incidence of about 20% to 30%.[1–3] IDH can be induced by multiple factors, mainly including the imbalance of the ultrafiltration rate and capillary refill rate, as well as the decrease in cardiac function, electrolyte disturbance, high dialysate temperature, and immune response to hemodialysis.[4] This disease often occurs in the middle and late stages of hemodialysis. Patients mainly present with nausea, vomiting, palpitation, sweating and paleness, and hypotensive shock and sudden death may occur in severe cases.[5,6] Therefore, it is of great significance to conduct early prophylaxis and timely management of IDH is important. Shenmai injection (SMI) is a compound of effective saponins extracted from Ginseng (Panax ginseng) and Radix Ophiopogonis (Ophiopogon japonicus).[7] This formula originates from Qianjin Pharmacopoeia and has the effects of rescuing rebellion and returning Yang, consolidating deficiency, nourishing Yin and clearing the heart, restoring the pulse and benefiting Qi, and benefiting Qi and generating fluid.[8,9] It is mainly employed in the treatment of “deficiency labor” and “syncope” in clinical practice.[10] According to modern pharmacological research, ginsenosides, maitake saponins and maitake flavonoids in Ginseng and Radix Ophiopogonis can improve the adaptability of myocardial cells to hypoxia, enhance myocardial metabolism, protect the lipid structure of myocardial cell membranes, stabilize cell membranes, promote the synthesis and release of prostaglandins from myocardial cells, and regulate vascular endothelial function.[7,11–13] It has been found that giving an appropriate amount of SMI to patients receiving hemodialysis can effectively reduce the incidence of IDH. As is revealed in many randomized controlled trials,[14–16] SMI could achieve significant efficacy in the prophylaxis and treatment of IDH. However, there are some limitations due to varying effect sizes and small sample sizes, as well as a lack of relevant systematic evaluation and conclusions. Based on that, the efficacy and safety of SMI in the prophylaxis and treatment of IDH were evaluated by a meta-analysis, with the aim of providing a basis for its clinical application.
2. Methods
[SUBTITLE] 2.1. Study registration [SUBSECTION] The protocol of this review was registered in PROSPERO (CRD42022328480). Meanwhile, it was reported as per the statement guidelines of preferred reporting items for the systematic review and meta-analysis protocol.[17] The protocol of this review was registered in PROSPERO (CRD42022328480). Meanwhile, it was reported as per the statement guidelines of preferred reporting items for the systematic review and meta-analysis protocol.[17] [SUBTITLE] 2.2. Inclusion criteria [SUBSECTION] [SUBTITLE] 2.2.1. Types of this study [SUBSECTION] In this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH. In this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH. [SUBTITLE] 2.2.2. Types of participants [SUBSECTION] The diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness; Patients with an age ≥18 years; Patients with a regular hemodialysis period >90 days; Patients with the hemodialysis frequency being 3 times a week. The diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness; Patients with an age ≥18 years; Patients with a regular hemodialysis period >90 days; Patients with the hemodialysis frequency being 3 times a week. [SUBTITLE] 2.2.3. Types of interventions [SUBSECTION] In the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs. In the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs. [SUBTITLE] 2.2.4. Types of outcome indexes [SUBSECTION] [SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome. The incidence of IDH was the primary outcome. [SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events. The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events. [SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome. The incidence of IDH was the primary outcome. [SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events. The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events. [SUBTITLE] 2.2.1. Types of this study [SUBSECTION] In this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH. In this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH. [SUBTITLE] 2.2.2. Types of participants [SUBSECTION] The diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness; Patients with an age ≥18 years; Patients with a regular hemodialysis period >90 days; Patients with the hemodialysis frequency being 3 times a week. The diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness; Patients with an age ≥18 years; Patients with a regular hemodialysis period >90 days; Patients with the hemodialysis frequency being 3 times a week. [SUBTITLE] 2.2.3. Types of interventions [SUBSECTION] In the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs. In the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs. [SUBTITLE] 2.2.4. Types of outcome indexes [SUBSECTION] [SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome. The incidence of IDH was the primary outcome. [SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events. The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events. [SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome. The incidence of IDH was the primary outcome. [SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events. The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events. [SUBTITLE] 2.3. Exclusion criteria [SUBSECTION] Non-randomized controlled trials (studies related to animal experiments, theoretical discussion, review and experience summary, case reports, analytical articles and so forth); Duplicate publications; Articles without complete outcome indicators. Non-randomized controlled trials (studies related to animal experiments, theoretical discussion, review and experience summary, case reports, analytical articles and so forth); Duplicate publications; Articles without complete outcome indicators. [SUBTITLE] 2.4. Information sources and retrieval strategy [SUBSECTION] The foreign and domestic articles on the prophylaxis and treatment of IDH with SMI were retrieved from PubMed, Web of Science, Scopus, Cochrane Library, Embase, China Scientific Journal Database, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, and Wanfang Data. Besides, relevant articles were also obtained from the Internet as supplements. In addition, the references included in the article were also be consulted, and other relevant articles were manually searched to obtain relevant information that was not found during the above retrieval. All retrieval strategies were determined after multiple searches. The retrieval period was determined from the establishment of the database to June 2022. The research strategy for PubMed is shown in Table 1. Search strategy in PubMed database. The foreign and domestic articles on the prophylaxis and treatment of IDH with SMI were retrieved from PubMed, Web of Science, Scopus, Cochrane Library, Embase, China Scientific Journal Database, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, and Wanfang Data. Besides, relevant articles were also obtained from the Internet as supplements. In addition, the references included in the article were also be consulted, and other relevant articles were manually searched to obtain relevant information that was not found during the above retrieval. All retrieval strategies were determined after multiple searches. The retrieval period was determined from the establishment of the database to June 2022. The research strategy for PubMed is shown in Table 1. Search strategy in PubMed database. [SUBTITLE] 2.5. Data collection and analysis [SUBSECTION] [SUBTITLE] 2.5.1. Data extraction and management [SUBSECTION] The acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1. Flow diagram of study selection process. The acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1. Flow diagram of study selection process. [SUBTITLE] 2.5.2. Assessment of risk of bias [SUBSECTION] The risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it. The risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it. [SUBTITLE] 2.5.3. Measures of treatment effects [SUBSECTION] For dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval. For dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval. [SUBTITLE] 2.5.4. Management of missing data [SUBSECTION] If there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data. If there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data. [SUBTITLE] 2.5.5. Assessment of heterogeneity and data synthesis [SUBSECTION] A meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis. A meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis. [SUBTITLE] 2.5.6. Assessment of reporting biases [SUBSECTION] A funnel plot was utilized to test the potential publication bias.[19] A funnel plot was utilized to test the potential publication bias.[19] [SUBTITLE] 2.5.7. Subgroup analysis [SUBSECTION] A subgroup analysis was conducted based on the primary intervention in the control group. A subgroup analysis was conducted based on the primary intervention in the control group. [SUBTITLE] 2.5.8. Sensitivity analysis [SUBSECTION] A sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results. A sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results. [SUBTITLE] 2.5.9. Ethics and dissemination [SUBSECTION] Ethical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences. Ethical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences. [SUBTITLE] 2.5.1. Data extraction and management [SUBSECTION] The acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1. Flow diagram of study selection process. The acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1. Flow diagram of study selection process. [SUBTITLE] 2.5.2. Assessment of risk of bias [SUBSECTION] The risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it. The risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it. [SUBTITLE] 2.5.3. Measures of treatment effects [SUBSECTION] For dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval. For dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval. [SUBTITLE] 2.5.4. Management of missing data [SUBSECTION] If there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data. If there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data. [SUBTITLE] 2.5.5. Assessment of heterogeneity and data synthesis [SUBSECTION] A meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis. A meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis. [SUBTITLE] 2.5.6. Assessment of reporting biases [SUBSECTION] A funnel plot was utilized to test the potential publication bias.[19] A funnel plot was utilized to test the potential publication bias.[19] [SUBTITLE] 2.5.7. Subgroup analysis [SUBSECTION] A subgroup analysis was conducted based on the primary intervention in the control group. A subgroup analysis was conducted based on the primary intervention in the control group. [SUBTITLE] 2.5.8. Sensitivity analysis [SUBSECTION] A sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results. A sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results. [SUBTITLE] 2.5.9. Ethics and dissemination [SUBSECTION] Ethical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences. Ethical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences.
null
null
null
null
[ "2.1. Study registration", "2.2. Inclusion criteria", "2.2.1. Types of this study", "2.2.2. Types of participants", "2.2.3. Types of interventions", "2.2.4. Types of outcome indexes", "2.2.4.1. Primary outcomes", "2.2.4.2. Secondary outcomes", "2.3. Exclusion criteria", "2.4. Information sources and retrieval strategy", "2.5. Data collection and analysis", "2.5.1. Data extraction and management", "2.5.2. Assessment of risk of bias", "2.5.3. Measures of treatment effects", "2.5.4. Management of missing data", "2.5.5. Assessment of heterogeneity and data synthesis", "2.5.6. Assessment of reporting biases", "2.5.7. Subgroup analysis", "2.5.8. Sensitivity analysis", "2.5.9. Ethics and dissemination", "Author contributions" ]
[ "The protocol of this review was registered in PROSPERO (CRD42022328480). Meanwhile, it was reported as per the statement guidelines of preferred reporting items for the systematic review and meta-analysis protocol.[17]", "[SUBTITLE] 2.2.1. Types of this study [SUBSECTION] In this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH.\nIn this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH.\n[SUBTITLE] 2.2.2. Types of participants [SUBSECTION] The diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness;\nPatients with an age ≥18 years;\nPatients with a regular hemodialysis period >90 days;\nPatients with the hemodialysis frequency being 3 times a week.\nThe diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness;\nPatients with an age ≥18 years;\nPatients with a regular hemodialysis period >90 days;\nPatients with the hemodialysis frequency being 3 times a week.\n[SUBTITLE] 2.2.3. Types of interventions [SUBSECTION] In the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs.\nIn the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs.\n[SUBTITLE] 2.2.4. Types of outcome indexes [SUBSECTION] [SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome.\nThe incidence of IDH was the primary outcome.\n[SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\nThe secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\n[SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome.\nThe incidence of IDH was the primary outcome.\n[SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\nThe secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.", "In this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH.", "The diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness;\nPatients with an age ≥18 years;\nPatients with a regular hemodialysis period >90 days;\nPatients with the hemodialysis frequency being 3 times a week.", "In the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs.", "[SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome.\nThe incidence of IDH was the primary outcome.\n[SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\nThe secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.", "The incidence of IDH was the primary outcome.", "The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.", "Non-randomized controlled trials (studies related to animal experiments, theoretical discussion, review and experience summary, case reports, analytical articles and so forth);\nDuplicate publications;\nArticles without complete outcome indicators.", "The foreign and domestic articles on the prophylaxis and treatment of IDH with SMI were retrieved from PubMed, Web of Science, Scopus, Cochrane Library, Embase, China Scientific Journal Database, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, and Wanfang Data. Besides, relevant articles were also obtained from the Internet as supplements. In addition, the references included in the article were also be consulted, and other relevant articles were manually searched to obtain relevant information that was not found during the above retrieval. All retrieval strategies were determined after multiple searches. The retrieval period was determined from the establishment of the database to June 2022. The research strategy for PubMed is shown in Table 1.\nSearch strategy in PubMed database.", "[SUBTITLE] 2.5.1. Data extraction and management [SUBSECTION] The acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1.\nFlow diagram of study selection process.\nThe acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1.\nFlow diagram of study selection process.\n[SUBTITLE] 2.5.2. Assessment of risk of bias [SUBSECTION] The risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it.\nThe risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it.\n[SUBTITLE] 2.5.3. Measures of treatment effects [SUBSECTION] For dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval.\nFor dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval.\n[SUBTITLE] 2.5.4. Management of missing data [SUBSECTION] If there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data.\nIf there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data.\n[SUBTITLE] 2.5.5. Assessment of heterogeneity and data synthesis [SUBSECTION] A meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis.\nA meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis.\n[SUBTITLE] 2.5.6. Assessment of reporting biases [SUBSECTION] A funnel plot was utilized to test the potential publication bias.[19]\nA funnel plot was utilized to test the potential publication bias.[19]\n[SUBTITLE] 2.5.7. Subgroup analysis [SUBSECTION] A subgroup analysis was conducted based on the primary intervention in the control group.\nA subgroup analysis was conducted based on the primary intervention in the control group.\n[SUBTITLE] 2.5.8. Sensitivity analysis [SUBSECTION] A sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results.\nA sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results.\n[SUBTITLE] 2.5.9. Ethics and dissemination [SUBSECTION] Ethical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences.\nEthical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences.", "The acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1.\nFlow diagram of study selection process.", "The risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it.", "For dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval.", "If there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data.", "A meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis.", "A funnel plot was utilized to test the potential publication bias.[19]", "A subgroup analysis was conducted based on the primary intervention in the control group.", "A sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results.", "Ethical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences.", "Conceptualization: Zhen Xiang, Guodan Yu.\nData curation: Zhen Xiang, Xin Lin.\nFunding acquisition: Guodan Yu.\nMethodology: Xin Lin.\nResources: Xin Lin, Jun Wang.\nSupervision: Guodan Yu.\nValidation: Jun Wang.\nVisualization: Jun Wang.\nWriting – original draft: Zhen Xiang, Guodan Yu.\nWriting – review & editing: Zhen Xiang, Guodan Yu." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Methods", "2.1. Study registration", "2.2. Inclusion criteria", "2.2.1. Types of this study", "2.2.2. Types of participants", "2.2.3. Types of interventions", "2.2.4. Types of outcome indexes", "2.2.4.1. Primary outcomes", "2.2.4.2. Secondary outcomes", "2.3. Exclusion criteria", "2.4. Information sources and retrieval strategy", "2.5. Data collection and analysis", "2.5.1. Data extraction and management", "2.5.2. Assessment of risk of bias", "2.5.3. Measures of treatment effects", "2.5.4. Management of missing data", "2.5.5. Assessment of heterogeneity and data synthesis", "2.5.6. Assessment of reporting biases", "2.5.7. Subgroup analysis", "2.5.8. Sensitivity analysis", "2.5.9. Ethics and dissemination", "3. Discussion", "Author contributions" ]
[ "Intradialytic hypotension (IDH) is one of the common clinical complications in patients receiving maintenance hemodialysis, with a high incidence of about 20% to 30%.[1–3] IDH can be induced by multiple factors, mainly including the imbalance of the ultrafiltration rate and capillary refill rate, as well as the decrease in cardiac function, electrolyte disturbance, high dialysate temperature, and immune response to hemodialysis.[4] This disease often occurs in the middle and late stages of hemodialysis. Patients mainly present with nausea, vomiting, palpitation, sweating and paleness, and hypotensive shock and sudden death may occur in severe cases.[5,6] Therefore, it is of great significance to conduct early prophylaxis and timely management of IDH is important.\nShenmai injection (SMI) is a compound of effective saponins extracted from Ginseng (Panax ginseng) and Radix Ophiopogonis (Ophiopogon japonicus).[7] This formula originates from Qianjin Pharmacopoeia and has the effects of rescuing rebellion and returning Yang, consolidating deficiency, nourishing Yin and clearing the heart, restoring the pulse and benefiting Qi, and benefiting Qi and generating fluid.[8,9] It is mainly employed in the treatment of “deficiency labor” and “syncope” in clinical practice.[10] According to modern pharmacological research, ginsenosides, maitake saponins and maitake flavonoids in Ginseng and Radix Ophiopogonis can improve the adaptability of myocardial cells to hypoxia, enhance myocardial metabolism, protect the lipid structure of myocardial cell membranes, stabilize cell membranes, promote the synthesis and release of prostaglandins from myocardial cells, and regulate vascular endothelial function.[7,11–13] It has been found that giving an appropriate amount of SMI to patients receiving hemodialysis can effectively reduce the incidence of IDH.\nAs is revealed in many randomized controlled trials,[14–16] SMI could achieve significant efficacy in the prophylaxis and treatment of IDH. However, there are some limitations due to varying effect sizes and small sample sizes, as well as a lack of relevant systematic evaluation and conclusions. Based on that, the efficacy and safety of SMI in the prophylaxis and treatment of IDH were evaluated by a meta-analysis, with the aim of providing a basis for its clinical application.", "[SUBTITLE] 2.1. Study registration [SUBSECTION] The protocol of this review was registered in PROSPERO (CRD42022328480). Meanwhile, it was reported as per the statement guidelines of preferred reporting items for the systematic review and meta-analysis protocol.[17]\nThe protocol of this review was registered in PROSPERO (CRD42022328480). Meanwhile, it was reported as per the statement guidelines of preferred reporting items for the systematic review and meta-analysis protocol.[17]\n[SUBTITLE] 2.2. Inclusion criteria [SUBSECTION] [SUBTITLE] 2.2.1. Types of this study [SUBSECTION] In this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH.\nIn this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH.\n[SUBTITLE] 2.2.2. Types of participants [SUBSECTION] The diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness;\nPatients with an age ≥18 years;\nPatients with a regular hemodialysis period >90 days;\nPatients with the hemodialysis frequency being 3 times a week.\nThe diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness;\nPatients with an age ≥18 years;\nPatients with a regular hemodialysis period >90 days;\nPatients with the hemodialysis frequency being 3 times a week.\n[SUBTITLE] 2.2.3. Types of interventions [SUBSECTION] In the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs.\nIn the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs.\n[SUBTITLE] 2.2.4. Types of outcome indexes [SUBSECTION] [SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome.\nThe incidence of IDH was the primary outcome.\n[SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\nThe secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\n[SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome.\nThe incidence of IDH was the primary outcome.\n[SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\nThe secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\n[SUBTITLE] 2.2.1. Types of this study [SUBSECTION] In this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH.\nIn this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH.\n[SUBTITLE] 2.2.2. Types of participants [SUBSECTION] The diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness;\nPatients with an age ≥18 years;\nPatients with a regular hemodialysis period >90 days;\nPatients with the hemodialysis frequency being 3 times a week.\nThe diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness;\nPatients with an age ≥18 years;\nPatients with a regular hemodialysis period >90 days;\nPatients with the hemodialysis frequency being 3 times a week.\n[SUBTITLE] 2.2.3. Types of interventions [SUBSECTION] In the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs.\nIn the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs.\n[SUBTITLE] 2.2.4. Types of outcome indexes [SUBSECTION] [SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome.\nThe incidence of IDH was the primary outcome.\n[SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\nThe secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\n[SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome.\nThe incidence of IDH was the primary outcome.\n[SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\nThe secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\n[SUBTITLE] 2.3. Exclusion criteria [SUBSECTION] Non-randomized controlled trials (studies related to animal experiments, theoretical discussion, review and experience summary, case reports, analytical articles and so forth);\nDuplicate publications;\nArticles without complete outcome indicators.\nNon-randomized controlled trials (studies related to animal experiments, theoretical discussion, review and experience summary, case reports, analytical articles and so forth);\nDuplicate publications;\nArticles without complete outcome indicators.\n[SUBTITLE] 2.4. Information sources and retrieval strategy [SUBSECTION] The foreign and domestic articles on the prophylaxis and treatment of IDH with SMI were retrieved from PubMed, Web of Science, Scopus, Cochrane Library, Embase, China Scientific Journal Database, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, and Wanfang Data. Besides, relevant articles were also obtained from the Internet as supplements. In addition, the references included in the article were also be consulted, and other relevant articles were manually searched to obtain relevant information that was not found during the above retrieval. All retrieval strategies were determined after multiple searches. The retrieval period was determined from the establishment of the database to June 2022. The research strategy for PubMed is shown in Table 1.\nSearch strategy in PubMed database.\nThe foreign and domestic articles on the prophylaxis and treatment of IDH with SMI were retrieved from PubMed, Web of Science, Scopus, Cochrane Library, Embase, China Scientific Journal Database, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, and Wanfang Data. Besides, relevant articles were also obtained from the Internet as supplements. In addition, the references included in the article were also be consulted, and other relevant articles were manually searched to obtain relevant information that was not found during the above retrieval. All retrieval strategies were determined after multiple searches. The retrieval period was determined from the establishment of the database to June 2022. The research strategy for PubMed is shown in Table 1.\nSearch strategy in PubMed database.\n[SUBTITLE] 2.5. Data collection and analysis [SUBSECTION] [SUBTITLE] 2.5.1. Data extraction and management [SUBSECTION] The acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1.\nFlow diagram of study selection process.\nThe acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1.\nFlow diagram of study selection process.\n[SUBTITLE] 2.5.2. Assessment of risk of bias [SUBSECTION] The risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it.\nThe risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it.\n[SUBTITLE] 2.5.3. Measures of treatment effects [SUBSECTION] For dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval.\nFor dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval.\n[SUBTITLE] 2.5.4. Management of missing data [SUBSECTION] If there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data.\nIf there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data.\n[SUBTITLE] 2.5.5. Assessment of heterogeneity and data synthesis [SUBSECTION] A meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis.\nA meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis.\n[SUBTITLE] 2.5.6. Assessment of reporting biases [SUBSECTION] A funnel plot was utilized to test the potential publication bias.[19]\nA funnel plot was utilized to test the potential publication bias.[19]\n[SUBTITLE] 2.5.7. Subgroup analysis [SUBSECTION] A subgroup analysis was conducted based on the primary intervention in the control group.\nA subgroup analysis was conducted based on the primary intervention in the control group.\n[SUBTITLE] 2.5.8. Sensitivity analysis [SUBSECTION] A sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results.\nA sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results.\n[SUBTITLE] 2.5.9. Ethics and dissemination [SUBSECTION] Ethical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences.\nEthical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences.\n[SUBTITLE] 2.5.1. Data extraction and management [SUBSECTION] The acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1.\nFlow diagram of study selection process.\nThe acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1.\nFlow diagram of study selection process.\n[SUBTITLE] 2.5.2. Assessment of risk of bias [SUBSECTION] The risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it.\nThe risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it.\n[SUBTITLE] 2.5.3. Measures of treatment effects [SUBSECTION] For dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval.\nFor dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval.\n[SUBTITLE] 2.5.4. Management of missing data [SUBSECTION] If there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data.\nIf there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data.\n[SUBTITLE] 2.5.5. Assessment of heterogeneity and data synthesis [SUBSECTION] A meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis.\nA meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis.\n[SUBTITLE] 2.5.6. Assessment of reporting biases [SUBSECTION] A funnel plot was utilized to test the potential publication bias.[19]\nA funnel plot was utilized to test the potential publication bias.[19]\n[SUBTITLE] 2.5.7. Subgroup analysis [SUBSECTION] A subgroup analysis was conducted based on the primary intervention in the control group.\nA subgroup analysis was conducted based on the primary intervention in the control group.\n[SUBTITLE] 2.5.8. Sensitivity analysis [SUBSECTION] A sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results.\nA sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results.\n[SUBTITLE] 2.5.9. Ethics and dissemination [SUBSECTION] Ethical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences.\nEthical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences.", "The protocol of this review was registered in PROSPERO (CRD42022328480). Meanwhile, it was reported as per the statement guidelines of preferred reporting items for the systematic review and meta-analysis protocol.[17]", "[SUBTITLE] 2.2.1. Types of this study [SUBSECTION] In this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH.\nIn this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH.\n[SUBTITLE] 2.2.2. Types of participants [SUBSECTION] The diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness;\nPatients with an age ≥18 years;\nPatients with a regular hemodialysis period >90 days;\nPatients with the hemodialysis frequency being 3 times a week.\nThe diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness;\nPatients with an age ≥18 years;\nPatients with a regular hemodialysis period >90 days;\nPatients with the hemodialysis frequency being 3 times a week.\n[SUBTITLE] 2.2.3. Types of interventions [SUBSECTION] In the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs.\nIn the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs.\n[SUBTITLE] 2.2.4. Types of outcome indexes [SUBSECTION] [SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome.\nThe incidence of IDH was the primary outcome.\n[SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\nThe secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\n[SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome.\nThe incidence of IDH was the primary outcome.\n[SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\nThe secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.", "In this study, randomized controlled trials would be conducted to investigate the efficacy and safety of SMI in the prophylaxis and treatment of IDH.", "The diagnostic criteria of Kidney Disease Outcomes Quality Initiative (KDOQI) were adopted during the inclusion process. KDOQI guidelines for hemodialysis hypotension diagnosis, that is, a decrease in mean arterial pressure (MAP) ≥10 mm Hg or a decrease in systolic blood pressure (SBP) ≥20 mm Hg, were adopted during the inclusion process, which may be accompanied by the symptoms of hypotension, such as dizziness, fatigue and weakness;\nPatients with an age ≥18 years;\nPatients with a regular hemodialysis period >90 days;\nPatients with the hemodialysis frequency being 3 times a week.", "In the experimental group, patients were treated with SMI, regardless of the dose, timing and duration of administration, or combined with conventional treatment. In the control group, patients were treated with conventional treatment alone, including reducing blood flow rate, controlling ultrafiltration, and applying vasoactive drugs.", "[SUBTITLE] 2.2.4.1. Primary outcomes [SUBSECTION] The incidence of IDH was the primary outcome.\nThe incidence of IDH was the primary outcome.\n[SUBTITLE] 2.2.4.2. Secondary outcomes [SUBSECTION] The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.\nThe secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.", "The incidence of IDH was the primary outcome.", "The secondary outcomes were composed of the total effective rate, the mean arterial pressure after hemodialysis and adverse events.", "Non-randomized controlled trials (studies related to animal experiments, theoretical discussion, review and experience summary, case reports, analytical articles and so forth);\nDuplicate publications;\nArticles without complete outcome indicators.", "The foreign and domestic articles on the prophylaxis and treatment of IDH with SMI were retrieved from PubMed, Web of Science, Scopus, Cochrane Library, Embase, China Scientific Journal Database, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, and Wanfang Data. Besides, relevant articles were also obtained from the Internet as supplements. In addition, the references included in the article were also be consulted, and other relevant articles were manually searched to obtain relevant information that was not found during the above retrieval. All retrieval strategies were determined after multiple searches. The retrieval period was determined from the establishment of the database to June 2022. The research strategy for PubMed is shown in Table 1.\nSearch strategy in PubMed database.", "[SUBTITLE] 2.5.1. Data extraction and management [SUBSECTION] The acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1.\nFlow diagram of study selection process.\nThe acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1.\nFlow diagram of study selection process.\n[SUBTITLE] 2.5.2. Assessment of risk of bias [SUBSECTION] The risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it.\nThe risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it.\n[SUBTITLE] 2.5.3. Measures of treatment effects [SUBSECTION] For dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval.\nFor dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval.\n[SUBTITLE] 2.5.4. Management of missing data [SUBSECTION] If there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data.\nIf there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data.\n[SUBTITLE] 2.5.5. Assessment of heterogeneity and data synthesis [SUBSECTION] A meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis.\nA meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis.\n[SUBTITLE] 2.5.6. Assessment of reporting biases [SUBSECTION] A funnel plot was utilized to test the potential publication bias.[19]\nA funnel plot was utilized to test the potential publication bias.[19]\n[SUBTITLE] 2.5.7. Subgroup analysis [SUBSECTION] A subgroup analysis was conducted based on the primary intervention in the control group.\nA subgroup analysis was conducted based on the primary intervention in the control group.\n[SUBTITLE] 2.5.8. Sensitivity analysis [SUBSECTION] A sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results.\nA sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results.\n[SUBTITLE] 2.5.9. Ethics and dissemination [SUBSECTION] Ethical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences.\nEthical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences.", "The acquired articles were independently screened and cross-checked by two investigators according to the inclusion and exclusion criteria. The title of the article was read first. If it was in line with the inclusion criteria, the abstract and full text were further read and included if they were consistent with the inclusion criteria. In case of disagreement, a third researcher was consulted to assist in the determination or experts were consulted to resolve it. The data extraction table was prepared by two researchers independently based on the method recommended by the Cochrane Collaboration. The contents mainly included: basic information of the study, such as the first author of the included article and date of publication; types of study design, interventions, and controls; clinical indicators, including the incidence of IDH, the total effective rate, and the mean arterial pressure after hemodialysis. The specific screening process is shown in Figure 1.\nFlow diagram of study selection process.", "The risk of bias in the included articles was assessed according to the Risk of Bias Assessment Tool in Cochrane Handbook of Systematic Reviews (5th edition).[18] Two investigators evaluated the risk of bias for each item as unclear, low risk, and high risk of bias. In case of disagreement, the third investigator was responsible for resolving it.", "For dichotomous outcomes, the risk ratio was used in the meta-analysis. The continuous data were expressed as the standardized mean difference. All of these data were summarized with a 95% confidence interval.", "If there are missing data in the included articles, the authors would be contacted by email to obtain relevant information. If the complete data cannot be obtained, it would be recorded in the bias risk assessment. Meanwhile, data analysis would be performed based on the existing data.", "A meta-analysis was performed with the assistance of RevMan 5.3 software (The Cochrane Collaboration, Oxford, UK). The included data were tested in terms of the heterogeneity with I2 <50% indicates a good homogeneity of the study, and a fixed-effects model would be used to conduct a meta-analysis. I2 >50% indicates a larger heterogeneity of the study, and the source of heterogeneity would be explored. If the significant clinical heterogeneity and the source of heterogeneity cannot be found, a random-effects model would be selected for analysis.", "A funnel plot was utilized to test the potential publication bias.[19]", "A subgroup analysis was conducted based on the primary intervention in the control group.", "A sensitivity analysis was conducted with the fixed-effects and random-effects models, in an attempt to test the stability of the results.", "Ethical approval is not required for this protocol due to the fact that the systematic evaluation is not involved with the data of individual patients. The results will be published in peer-reviewed journals and presented at related conferences.", "SMI is commonly used in the prophylaxis and treatment of IDH owing to its functions of benefiting Qi and strengthening the heart, generating fluid and restoring the blood vessels, activating blood circulation and removing blood stasis, and strengthening healthy qi to eliminate pathogens.[11,20–22] However, there is no meta-analysis on the prophylaxis and treatment of IDH with SMI at home and abroad. In this study, the incidence of hemodialysis-related hypotension, the total effective rate and the mean arterial pressure after hemodialysis were analyzed based on the results of clinical studies on the prophylaxis and treatment of IDH with SMI. These findings could provide the evidence-based medical basis for the application of SMI in the prophylaxis and treatment of IDH.", "Conceptualization: Zhen Xiang, Guodan Yu.\nData curation: Zhen Xiang, Xin Lin.\nFunding acquisition: Guodan Yu.\nMethodology: Xin Lin.\nResources: Xin Lin, Jun Wang.\nSupervision: Guodan Yu.\nValidation: Jun Wang.\nVisualization: Jun Wang.\nWriting – original draft: Zhen Xiang, Guodan Yu.\nWriting – review & editing: Zhen Xiang, Guodan Yu." ]
[ "intro", "methods", null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, "discussion", null ]
[ "intradialytic hypotension", "meta-analysis", "protocol", "Shenmai injection" ]
Multiplex PCR: Aid to more-timely and directed therapeutic intervention for patients with infectious gastroenteritis.
36254068
Multiplex PCR is a sensitive and rapid method compared with conventional methods. Therefore, we use multiplex PCR for the rapid detection of the four major intestinal pathogens causing gastroenteritis (Shigella spp., Campylobacter spp., Aeromonas spp. and Enterohemorrhagic Escherichia coli [EHEC]) in stool specimens.
BACKGROUND
A prospective randomized study using 200 stool samples obtained from patients presented with acute gastroenteritis during the study period (between February 2019 and December 2021). Bacteria in stool samples were identified using conventional culture methods and multiplex PCR for stool samples.
MATERIALS AND METHODS
The identified organisms using conventional cultures; were Shigella (27%), Aeromonas species (10%) and EHEC (O157) (8%). Using multiplex PCR. Shigella spp. was the most commonly identified pathogen (detected in 40.5% of positive samples), followed by Aeromonas spp. (30%), EHEC (20%) and Campylobacter species was only detected in (1%) of positive samples. The diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Shigella, EHEC and Aeromonas showed, sensitivity of 100% (for each), specificity of 88.5%, 92.4%, 77.8% respectively. However, the diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Campylobacter showed specificity of 99% and NPV of 100%.
RESULTS
Multiplex PCR is an accurate and rapid method for detection of common intestinal pathogens causing severe gastroenteritis. a rapid method that could be used in outbreaks for diagnosis of the common enteric pathogens causing fatal gastroenteritis.
CONCLUSIONS
[ "Diarrhea", "Feces", "Gastroenteritis", "Humans", "Molecular Diagnostic Techniques", "Multiplex Polymerase Chain Reaction", "Prospective Studies", "Sensitivity and Specificity" ]
9575839
1. Introduction
Infectious gastroenteritis (IG) is one of the most common diseases worldwide, killing millions of individuals each year. In industrialized countries, IG remains a major public health burden, although mortality is low. However, mortality can be found in developing countries, where epidemics of bacterial gastroenteritis may develop.[1] Shigella, Campylobacter and enterohemorrhagic E coli are three common intestinal pathogens involved in gastroenteritis and are important to study because of the frequency and severity of symptoms they cause. EHEC identification would also be critical in the event of food-borne illness outbreaks or agroterrorism, thus the accurate and rapid diagnosis is a must in outbreaks to direct therapeutic intervention.[2] Aeromonas causes diarrheal infections, most commonly in children (especially those under 5 years) and in immune-compromised patients and the incidence of gastroenteritis tends to be higher in summer than other seasons. The organism has been also isolated with high frequency from patients with traveler’s diarrhea.[3] Acute self-limiting diarrhea occurs in children, and chronic gastroenteritis or enterocolitis may occur in children and the elderly. The presentation of gastroenteritis caused by Aeromonas includes various combinations of fever, vomiting, and increased fecal leucocytes or erythrocytes.[4] Most medical microbiology laboratories use traditional and conventional methods for detection of infectious cause of diarrhea, among direct microscopic examination, the use of conventional cultures and identification of organisms using biochemical reactions, which take a long time; results can release after 3 to 4 days.[2] Furthermore, misidentification of culture increases hands-on time and delay in reporting of a definite negative result. Other problems are the viable but non-culturable state of Campylobacter jejuni[5] and the limited viability of Shigella outside the human body. These may compromise the sensitivity of culture.[6] Another value of the accurate and rapid diagnosis of gastroenteritis is the rapid start of therapeutic intervention as just mentioned before. It is known that bacterial gastroenteritis is a self-limiting disease, and antimicrobial therapy is not particularly required. Clinically, it is not possible to distinguish pathogens that will respond to antibiotics from those that will not respond. Antibiotics add to the cost of treatment, put the patient at risk for adverse events, and can encourage development of resistant bacteria.[7] However, in certain infections e.g. dysentery, cholera, and for certain cases of persistent diarrhea particularly in infants, elderly people, and immunocompromised individuals who are at higher risk of developing more severe complications, treatment can decrease the duration and severity of illness if it is initiated early in the course of infection.[8] Molecular methods are sensitive and rapid methods compared to conventional methods. However, broad application remains limited due to their assumed high costs, inhibition caused by fecal constituents, and the need for specialized laboratories. Due to the high throughput of stool screening and the number of possible enteric pathogens, implementation of a molecular approach which uses multiplexing of targets is mandatory.[9] The objective of this study is to evaluate the multiplex PCR for the rapid detection of four major intestinal pathogens causing gastroenteritis (Shigella spp., Campylobacter spp., Aeromonas spp. and Enterohemorrhagic Escherichia coli (EHEC) in stool specimens compared to other conventional culture methods.
3.4. Diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of studied pathogen
The majority of infections were caused by a single pathogen. However, multiple enteric pathogens in individuals’ samples were detected by the multiplex PCR not by conventional methods. Only three samples contain mixed infections with the four studied pathogen. The diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Shigella, EHEC and Aeromonas showed, sensitivity of 100% (for each), specificity of 88.5%, 92.4%, 77.8% respectively, PPV of 66.7%, 53.3%, and 33.3%, respectively and NPV of 100% (for each). However, the diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Campylobacter showed specificity of 99% and NPV of 100%; (sensitivity and PPV cannot be calculated as no Campylobacter isolate identified by conventional method; which considered as gold standard for diagnosis of bacterial infection) Table 4. Diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of studied pathogen. NPV = negative predictive value, PCR = polymerase chain reaction, PPV = positive predictive value.
3. Results
[SUBTITLE] 3.1. Demographic and clinical presentation of patients with gastroenteritis [SUBSECTION] In this prospective randomized cohort study, between February 2019 and December 2021. a total of 200 stool samples were collected from patients presented with acute gastroenteritis; including 120 female samples and 80 male samples with their age ranging from 18 years to 55 years. One hundred eighty (90%) of patients suffered from watery diarrhea while (10%) had bloody diarrhea. ninety patients (45%) had associated abdominal cramps/colic, (50/200, 25%) had vomiting, (40/200, 20%) had dehydration, (20/200, 10%) had steatorrhea and (4/200, 2%) complained from weight loss Table 1. Demographic and clinical presentation of patients with gastroenteritis. In this prospective randomized cohort study, between February 2019 and December 2021. a total of 200 stool samples were collected from patients presented with acute gastroenteritis; including 120 female samples and 80 male samples with their age ranging from 18 years to 55 years. One hundred eighty (90%) of patients suffered from watery diarrhea while (10%) had bloody diarrhea. ninety patients (45%) had associated abdominal cramps/colic, (50/200, 25%) had vomiting, (40/200, 20%) had dehydration, (20/200, 10%) had steatorrhea and (4/200, 2%) complained from weight loss Table 1. Demographic and clinical presentation of patients with gastroenteritis. [SUBTITLE] 3.2. Macroscopic and microscopic examination of the studied stool samples of all included patients [SUBSECTION] The macroscopic examination in the studied stool samples showed that (160/200, 80%) of samples were watery in consistency whereas (24/200, 12%) were semi-formed. Regarding the stool color (66%) of the studied samples were brownish, (20/200, 10%) were whitish, (22/200, 11%) were greenish, (12/200, 6%) were reddish, and (14/200, 7%) yellowish. Out of the 200 studied samples, 136 samples (68%) contained visible mucous and 12 samples (6%) were bloody. As regard to the microscopic examination, (40/200, 20%) of the studied samples had a WBCs count < 10 cells/HPF, (90/200, 45%) had a WBCs count 10 to 49 cells/HPF, (40/200, 20%) had a WBCs count 50 to 99 cells/HPF, and (36/200, 18%) had a WBCs count > 100 cells/HPF. Three out of the 200 studied samples (5.5%) contained red blood cells > 100/HPF Table 2. Macroscopic and microscopic examination of the studied stool samples of all included patients. HPF = high power field, RBCs = red blood cells, WBC = white blood cells. The macroscopic examination in the studied stool samples showed that (160/200, 80%) of samples were watery in consistency whereas (24/200, 12%) were semi-formed. Regarding the stool color (66%) of the studied samples were brownish, (20/200, 10%) were whitish, (22/200, 11%) were greenish, (12/200, 6%) were reddish, and (14/200, 7%) yellowish. Out of the 200 studied samples, 136 samples (68%) contained visible mucous and 12 samples (6%) were bloody. As regard to the microscopic examination, (40/200, 20%) of the studied samples had a WBCs count < 10 cells/HPF, (90/200, 45%) had a WBCs count 10 to 49 cells/HPF, (40/200, 20%) had a WBCs count 50 to 99 cells/HPF, and (36/200, 18%) had a WBCs count > 100 cells/HPF. Three out of the 200 studied samples (5.5%) contained red blood cells > 100/HPF Table 2. Macroscopic and microscopic examination of the studied stool samples of all included patients. HPF = high power field, RBCs = red blood cells, WBC = white blood cells. [SUBTITLE] 3.3. Comparison between conventional methods and multiplex-PCR with regard to the recovery of the studied pathogens in the 200 included patients [SUBSECTION] Using conventional culture and identification methods, (192 out of 200, 96%) studied stool samples gave positive results. The most commonly identified organisms were E coli (118/200, 59%), Shigella (54/200, 27%) and Aeromonas species (20/200, 10%). Campylobacter was not revealed in cultures. Using specific E coli O157 anti-sera, 16 isolates out of 118 identified E coli (13.56%) were found to belong to the O157 serotype, and the remaining isolates (102/118, 86.4%) were the normal gut E coli microbiota. Thus, the EHEC represented 8% (16/200) of the total identified pathogens by conventional culture method. Using multiplex PCR, 173/ 200 fecal samples (86.5%) were positive. Shigella spp. was the most commonly identified pathogen as it was detected in (81/200, 40.5%) of the PCR-positive samples; Out of the 81 multiplex PCR-positive Shigella spp., 27 samples (27/81, 33.3%) were not detected by conventional methods. Aeromonas spp. was detected in 60 samples (60/200, 30%); out of the 60 positive samples., forty samples (40/60, 66.67%) were missed by the conventional culture methods. EHEC were detected in (30/200, 15%) samples. Of them 14 samples were missed by conventional culture method. Campylobacter species were also detected in only 2 samples (2/200, 1%); Campylobacter spp. isolate was only detected by the multiplex PCR Figure 3. Comparison between conventional culture methods and multiplex-PCR as regard to the recovery of the pathogens in the 200 included patients. EHEC = Enterohemorrhagic Escherichia coli, PCR = polymerase chain reaction. Finally, Comparison between conventional methods and multiplex-PCR as regard to the recovery of the pathogens in the 200 included patients. Multiplex-PCR appeared to be more advantageous to conventional detection methods regarding the detection of Shigella, EHEC and Aeromonas isolates with a statistically significant difference (P < .05). Although, no significant difference was detected between conventional culture methods and Multiplex-PCR regarding detection of Campylobacter, but this pathogen was only detected by Multiplex-PCR method Table 3. Comparison between conventional methods and multiplex-PCR with regard to the recovery of the studied pathogens in the 200 included patients. PCR = polymerase chain reaction. χ2: Chi-square test. S: P value < .05 is considered significant. HS: P value < .001 is considered highly significant. NS: P value > .05 is considered non-significant. Using conventional culture and identification methods, (192 out of 200, 96%) studied stool samples gave positive results. The most commonly identified organisms were E coli (118/200, 59%), Shigella (54/200, 27%) and Aeromonas species (20/200, 10%). Campylobacter was not revealed in cultures. Using specific E coli O157 anti-sera, 16 isolates out of 118 identified E coli (13.56%) were found to belong to the O157 serotype, and the remaining isolates (102/118, 86.4%) were the normal gut E coli microbiota. Thus, the EHEC represented 8% (16/200) of the total identified pathogens by conventional culture method. Using multiplex PCR, 173/ 200 fecal samples (86.5%) were positive. Shigella spp. was the most commonly identified pathogen as it was detected in (81/200, 40.5%) of the PCR-positive samples; Out of the 81 multiplex PCR-positive Shigella spp., 27 samples (27/81, 33.3%) were not detected by conventional methods. Aeromonas spp. was detected in 60 samples (60/200, 30%); out of the 60 positive samples., forty samples (40/60, 66.67%) were missed by the conventional culture methods. EHEC were detected in (30/200, 15%) samples. Of them 14 samples were missed by conventional culture method. Campylobacter species were also detected in only 2 samples (2/200, 1%); Campylobacter spp. isolate was only detected by the multiplex PCR Figure 3. Comparison between conventional culture methods and multiplex-PCR as regard to the recovery of the pathogens in the 200 included patients. EHEC = Enterohemorrhagic Escherichia coli, PCR = polymerase chain reaction. Finally, Comparison between conventional methods and multiplex-PCR as regard to the recovery of the pathogens in the 200 included patients. Multiplex-PCR appeared to be more advantageous to conventional detection methods regarding the detection of Shigella, EHEC and Aeromonas isolates with a statistically significant difference (P < .05). Although, no significant difference was detected between conventional culture methods and Multiplex-PCR regarding detection of Campylobacter, but this pathogen was only detected by Multiplex-PCR method Table 3. Comparison between conventional methods and multiplex-PCR with regard to the recovery of the studied pathogens in the 200 included patients. PCR = polymerase chain reaction. χ2: Chi-square test. S: P value < .05 is considered significant. HS: P value < .001 is considered highly significant. NS: P value > .05 is considered non-significant. [SUBTITLE] 3.4. Diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of studied pathogen [SUBSECTION] The majority of infections were caused by a single pathogen. However, multiple enteric pathogens in individuals’ samples were detected by the multiplex PCR not by conventional methods. Only three samples contain mixed infections with the four studied pathogen. The diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Shigella, EHEC and Aeromonas showed, sensitivity of 100% (for each), specificity of 88.5%, 92.4%, 77.8% respectively, PPV of 66.7%, 53.3%, and 33.3%, respectively and NPV of 100% (for each). However, the diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Campylobacter showed specificity of 99% and NPV of 100%; (sensitivity and PPV cannot be calculated as no Campylobacter isolate identified by conventional method; which considered as gold standard for diagnosis of bacterial infection) Table 4. Diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of studied pathogen. NPV = negative predictive value, PCR = polymerase chain reaction, PPV = positive predictive value. The majority of infections were caused by a single pathogen. However, multiple enteric pathogens in individuals’ samples were detected by the multiplex PCR not by conventional methods. Only three samples contain mixed infections with the four studied pathogen. The diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Shigella, EHEC and Aeromonas showed, sensitivity of 100% (for each), specificity of 88.5%, 92.4%, 77.8% respectively, PPV of 66.7%, 53.3%, and 33.3%, respectively and NPV of 100% (for each). However, the diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Campylobacter showed specificity of 99% and NPV of 100%; (sensitivity and PPV cannot be calculated as no Campylobacter isolate identified by conventional method; which considered as gold standard for diagnosis of bacterial infection) Table 4. Diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of studied pathogen. NPV = negative predictive value, PCR = polymerase chain reaction, PPV = positive predictive value.
null
null
[ "2. Materials and Methods", "2.1. Patient selection", "2.2. Microbiological processing of stool samples", "2.2.1. Sample processing.", "2.2.2. Sample culture.", "2.3. Multiplex PCR extraction and amplification", "2.4. Statistical analysis", "2.4.1. Ethical approval and consent to participate.", "2.4.2. Conflict of interest.", "3.1. Demographic and clinical presentation of patients with gastroenteritis", "3.2. Macroscopic and microscopic examination of the studied stool samples of all included patients", "5. Conclusion", "5.1. Study limitation", "Author contributions" ]
[ "[SUBTITLE] 2.1. Patient selection [SUBSECTION] A total of 200 stool samples were collected randemly from patients attended Helwan, South Valley and Tanta Universities outpatient clinics with clinical manifestations of acute gastroenteritis; between February 2019 and December 2021. For each stool sample, a portion of the sample was transferred into a sterile tube and stored at −20°C for molecular diagnostics. The rest was used for stool culture process within 1 hours of receipt. All patients were subjected to full history taking, lying stress on manifestations of gastroenteritis (diarrhea, vomiting, fever and abdominal discomfort), bleeding tendency, hematuria and history of antibiotic intake. All patients aged from 18 to 60 years with clinical manifestations of acute gastroenteritis were included in the study. Patients with history of antibiotics intake, chronic diarrhea of any cause, cancer anywhere in the body including cancer colon, drug intake and immunosuppression due to any cause were excluded from the study.\nA total of 200 stool samples were collected randemly from patients attended Helwan, South Valley and Tanta Universities outpatient clinics with clinical manifestations of acute gastroenteritis; between February 2019 and December 2021. For each stool sample, a portion of the sample was transferred into a sterile tube and stored at −20°C for molecular diagnostics. The rest was used for stool culture process within 1 hours of receipt. All patients were subjected to full history taking, lying stress on manifestations of gastroenteritis (diarrhea, vomiting, fever and abdominal discomfort), bleeding tendency, hematuria and history of antibiotic intake. All patients aged from 18 to 60 years with clinical manifestations of acute gastroenteritis were included in the study. Patients with history of antibiotics intake, chronic diarrhea of any cause, cancer anywhere in the body including cancer colon, drug intake and immunosuppression due to any cause were excluded from the study.\n[SUBTITLE] 2.2. Microbiological processing of stool samples [SUBSECTION] [SUBTITLE] 2.2.1. Sample processing. [SUBSECTION] Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.\n Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.\n[SUBTITLE] 2.2.2. Sample culture. [SUBSECTION] Using a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]\nUsing a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]\n[SUBTITLE] 2.2.1. Sample processing. [SUBSECTION] Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.\n Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.\n[SUBTITLE] 2.2.2. Sample culture. [SUBSECTION] Using a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]\nUsing a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]\n[SUBTITLE] 2.3. Multiplex PCR extraction and amplification [SUBSECTION] A multiplex PCR was performed (by 2 clinical pathology doctors with more than 5 year experience) on all stool specimens for the detection of Shigella spp., Campylobacter spp.., Aeromonas spp. and E coli O157 DNA. After thawing the samples, 1 mL (for liquid stool) or 1 gram (for solid feces) was used for the extraction of bacterial DNA using the QIA amp DAN Mini Kit supplied by (Qiagen, Germantown, MD) according to the manufacturer’s instructions. The bacterial genomes were amplified by multiplex real-time PCR using the Seeplex® Diarrhea ACE Detection kits (Seegene, Republic of [South Korea]) were used for the multiplex PCR test. The bacterial panels included:\na) Diarrhea B1 ACE detection is a multiplex assay that permits the simultaneous amplification of target DNA of: Shigella spp. (S flexneri, S boydii, S sonnei and S dysenteriae), Campylobacter spp. (C jejuni and C coli) and Internal Control (IC) Figure 1.\nMultiplex PCR using the ACE B1 diarrhea kit (A) ACEB1 marker (B) Combined Shigella and Campylobacter (bp: 330 and 227 respectively), (C) Negative control. PCR = polymerase chain reaction.\nb) Diarrhea B2 ACE Detection is a multiplex assay that permits the simultaneous amplification of target DNA of: Aeromonas spp. (A salmonicida, A sobria, A bivalvium, A hydrophila), E coli O157:H7, VTEC family and Internal Control (IC). The target genes were respectively IpaH, Rfb, Hip and Hly for Shigella, EHEC, Campylobacter and Aeromonas Figure 2.\nMultiplex PCR using the ACE B2 diarrhea kit showing (A) and (B) Aeromonas spp. (bp: 217), (C) ACEB2 marker. PCR = polymerase chain reaction.\nShigella dysenteriae (Shiga) (ATCC® 13313) was used as a positive control and Staphylococcus aureus subsp. aureus Rosenbach (ATCC® 25923™) was used as a negative control\nA multiplex PCR was performed (by 2 clinical pathology doctors with more than 5 year experience) on all stool specimens for the detection of Shigella spp., Campylobacter spp.., Aeromonas spp. and E coli O157 DNA. After thawing the samples, 1 mL (for liquid stool) or 1 gram (for solid feces) was used for the extraction of bacterial DNA using the QIA amp DAN Mini Kit supplied by (Qiagen, Germantown, MD) according to the manufacturer’s instructions. The bacterial genomes were amplified by multiplex real-time PCR using the Seeplex® Diarrhea ACE Detection kits (Seegene, Republic of [South Korea]) were used for the multiplex PCR test. The bacterial panels included:\na) Diarrhea B1 ACE detection is a multiplex assay that permits the simultaneous amplification of target DNA of: Shigella spp. (S flexneri, S boydii, S sonnei and S dysenteriae), Campylobacter spp. (C jejuni and C coli) and Internal Control (IC) Figure 1.\nMultiplex PCR using the ACE B1 diarrhea kit (A) ACEB1 marker (B) Combined Shigella and Campylobacter (bp: 330 and 227 respectively), (C) Negative control. PCR = polymerase chain reaction.\nb) Diarrhea B2 ACE Detection is a multiplex assay that permits the simultaneous amplification of target DNA of: Aeromonas spp. (A salmonicida, A sobria, A bivalvium, A hydrophila), E coli O157:H7, VTEC family and Internal Control (IC). The target genes were respectively IpaH, Rfb, Hip and Hly for Shigella, EHEC, Campylobacter and Aeromonas Figure 2.\nMultiplex PCR using the ACE B2 diarrhea kit showing (A) and (B) Aeromonas spp. (bp: 217), (C) ACEB2 marker. PCR = polymerase chain reaction.\nShigella dysenteriae (Shiga) (ATCC® 13313) was used as a positive control and Staphylococcus aureus subsp. aureus Rosenbach (ATCC® 25923™) was used as a negative control\n[SUBTITLE] 2.4. Statistical analysis [SUBSECTION] Analysis was done using the IBM SPSS statistics (V. 20.0, IBM Corp., Chicago, USA, 2011). Descriptive Statistics: Qualitative data were presented as counts and percentage. The association between each 2 variables or comparison between 2 independent groups as regards the categorized data was done using the Chi square test or the Fisher exact test. The probability of error at > .05 was considered non-significant, while ≤ .05 was considered significant and < .01 was considered highly significant.\n[SUBTITLE] 2.4.1. Ethical approval and consent to participate. [SUBSECTION] The study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).\nThe study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).\n[SUBTITLE] 2.4.2. Conflict of interest. [SUBSECTION] We don’t receive any financial support or relationships that may pose conflict of interest in this work.\nWe don’t receive any financial support or relationships that may pose conflict of interest in this work.\nAnalysis was done using the IBM SPSS statistics (V. 20.0, IBM Corp., Chicago, USA, 2011). Descriptive Statistics: Qualitative data were presented as counts and percentage. The association between each 2 variables or comparison between 2 independent groups as regards the categorized data was done using the Chi square test or the Fisher exact test. The probability of error at > .05 was considered non-significant, while ≤ .05 was considered significant and < .01 was considered highly significant.\n[SUBTITLE] 2.4.1. Ethical approval and consent to participate. [SUBSECTION] The study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).\nThe study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).\n[SUBTITLE] 2.4.2. Conflict of interest. [SUBSECTION] We don’t receive any financial support or relationships that may pose conflict of interest in this work.\nWe don’t receive any financial support or relationships that may pose conflict of interest in this work.", "A total of 200 stool samples were collected randemly from patients attended Helwan, South Valley and Tanta Universities outpatient clinics with clinical manifestations of acute gastroenteritis; between February 2019 and December 2021. For each stool sample, a portion of the sample was transferred into a sterile tube and stored at −20°C for molecular diagnostics. The rest was used for stool culture process within 1 hours of receipt. All patients were subjected to full history taking, lying stress on manifestations of gastroenteritis (diarrhea, vomiting, fever and abdominal discomfort), bleeding tendency, hematuria and history of antibiotic intake. All patients aged from 18 to 60 years with clinical manifestations of acute gastroenteritis were included in the study. Patients with history of antibiotics intake, chronic diarrhea of any cause, cancer anywhere in the body including cancer colon, drug intake and immunosuppression due to any cause were excluded from the study.", "[SUBTITLE] 2.2.1. Sample processing. [SUBSECTION] Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.\n Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.\n[SUBTITLE] 2.2.2. Sample culture. [SUBSECTION] Using a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]\nUsing a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]", " Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.", "Using a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]", "A multiplex PCR was performed (by 2 clinical pathology doctors with more than 5 year experience) on all stool specimens for the detection of Shigella spp., Campylobacter spp.., Aeromonas spp. and E coli O157 DNA. After thawing the samples, 1 mL (for liquid stool) or 1 gram (for solid feces) was used for the extraction of bacterial DNA using the QIA amp DAN Mini Kit supplied by (Qiagen, Germantown, MD) according to the manufacturer’s instructions. The bacterial genomes were amplified by multiplex real-time PCR using the Seeplex® Diarrhea ACE Detection kits (Seegene, Republic of [South Korea]) were used for the multiplex PCR test. The bacterial panels included:\na) Diarrhea B1 ACE detection is a multiplex assay that permits the simultaneous amplification of target DNA of: Shigella spp. (S flexneri, S boydii, S sonnei and S dysenteriae), Campylobacter spp. (C jejuni and C coli) and Internal Control (IC) Figure 1.\nMultiplex PCR using the ACE B1 diarrhea kit (A) ACEB1 marker (B) Combined Shigella and Campylobacter (bp: 330 and 227 respectively), (C) Negative control. PCR = polymerase chain reaction.\nb) Diarrhea B2 ACE Detection is a multiplex assay that permits the simultaneous amplification of target DNA of: Aeromonas spp. (A salmonicida, A sobria, A bivalvium, A hydrophila), E coli O157:H7, VTEC family and Internal Control (IC). The target genes were respectively IpaH, Rfb, Hip and Hly for Shigella, EHEC, Campylobacter and Aeromonas Figure 2.\nMultiplex PCR using the ACE B2 diarrhea kit showing (A) and (B) Aeromonas spp. (bp: 217), (C) ACEB2 marker. PCR = polymerase chain reaction.\nShigella dysenteriae (Shiga) (ATCC® 13313) was used as a positive control and Staphylococcus aureus subsp. aureus Rosenbach (ATCC® 25923™) was used as a negative control", "Analysis was done using the IBM SPSS statistics (V. 20.0, IBM Corp., Chicago, USA, 2011). Descriptive Statistics: Qualitative data were presented as counts and percentage. The association between each 2 variables or comparison between 2 independent groups as regards the categorized data was done using the Chi square test or the Fisher exact test. The probability of error at > .05 was considered non-significant, while ≤ .05 was considered significant and < .01 was considered highly significant.\n[SUBTITLE] 2.4.1. Ethical approval and consent to participate. [SUBSECTION] The study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).\nThe study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).\n[SUBTITLE] 2.4.2. Conflict of interest. [SUBSECTION] We don’t receive any financial support or relationships that may pose conflict of interest in this work.\nWe don’t receive any financial support or relationships that may pose conflict of interest in this work.", "The study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).", "We don’t receive any financial support or relationships that may pose conflict of interest in this work.", "In this prospective randomized cohort study, between February 2019 and December 2021. a total of 200 stool samples were collected from patients presented with acute gastroenteritis; including 120 female samples and 80 male samples with their age ranging from 18 years to 55 years. One hundred eighty (90%) of patients suffered from watery diarrhea while (10%) had bloody diarrhea. ninety patients (45%) had associated abdominal cramps/colic, (50/200, 25%) had vomiting, (40/200, 20%) had dehydration, (20/200, 10%) had steatorrhea and (4/200, 2%) complained from weight loss Table 1.\nDemographic and clinical presentation of patients with gastroenteritis.", "The macroscopic examination in the studied stool samples showed that (160/200, 80%) of samples were watery in consistency whereas (24/200, 12%) were semi-formed. Regarding the stool color (66%) of the studied samples were brownish, (20/200, 10%) were whitish, (22/200, 11%) were greenish, (12/200, 6%) were reddish, and (14/200, 7%) yellowish. Out of the 200 studied samples, 136 samples (68%) contained visible mucous and 12 samples (6%) were bloody. As regard to the microscopic examination, (40/200, 20%) of the studied samples had a WBCs count < 10 cells/HPF, (90/200, 45%) had a WBCs count 10 to 49 cells/HPF, (40/200, 20%) had a WBCs count 50 to 99 cells/HPF, and (36/200, 18%) had a WBCs count > 100 cells/HPF. Three out of the 200 studied samples (5.5%) contained red blood cells > 100/HPF Table 2.\nMacroscopic and microscopic examination of the studied stool samples of all included patients.\nHPF = high power field, RBCs = red blood cells, WBC = white blood cells.", "Multiplex PCR can overcome limitations of culture-based methods as it is a more rapid and to some extent more accurate method with higher sensitivity compared to the traditional cultured based methods for detection of common intestinal pathogens.\n[SUBTITLE] 5.1. Study limitation [SUBSECTION] No reevaluation of patients after antimicrobial therapy as multiplex PCR panels can’t differentiate between viable and dead organisms, especially in patients with multiple stools detected organisms, which could be related to colonic colonization with asymptomatic organisms.\nSince no controls were included in this study and enteropathogens have often been detected in healthy controls, the detection didn’t necessarily mean disease association.\nNo reevaluation of patients after antimicrobial therapy as multiplex PCR panels can’t differentiate between viable and dead organisms, especially in patients with multiple stools detected organisms, which could be related to colonic colonization with asymptomatic organisms.\nSince no controls were included in this study and enteropathogens have often been detected in healthy controls, the detection didn’t necessarily mean disease association.", "No reevaluation of patients after antimicrobial therapy as multiplex PCR panels can’t differentiate between viable and dead organisms, especially in patients with multiple stools detected organisms, which could be related to colonic colonization with asymptomatic organisms.\nSince no controls were included in this study and enteropathogens have often been detected in healthy controls, the detection didn’t necessarily mean disease association.", "Conceptualization: Karim Montasser, Heba Ahmed Osman, and Abeer M. M. Sabry.\nData curation: Karim Montasser, Heba Ahmed Osman, Abeer M. M. Sabry, Haidy S. Khalil, Hanan Abozaid, and Wesam Hatem Amer.\nFormal analysis: Karim Montasser\nInvestigation: Karim Montasser, Heba Ahmed Osman, Hanan Abozaid, Haidy S. Khalil, Wesam Hatem Amer, and Abeer M. M. Sabry.\nMethodology: Karim Montasser, Heba Ahmed Osman, Abeer M. M. Sabry, Haidy S. Khalil, Hanan Abozaid, and Wesam Hatem Amer.\nProject administration: Karim Montasser.\nWriting – original draft: Wesam Hatem Amer, Hanan Abozaid, and Haidy S. Khalil.\nWriting – review & editing: Karim Montasser, Heba Ahmed Osman, and Abeer M. M. Sabry" ]
[ "methods", "subjects", null, null, null, null, null, null, null, "subjects", "subjects", null, null, null ]
[ "1. Introduction", "2. Materials and Methods", "2.1. Patient selection", "2.2. Microbiological processing of stool samples", "2.2.1. Sample processing.", "2.2.2. Sample culture.", "2.3. Multiplex PCR extraction and amplification", "2.4. Statistical analysis", "2.4.1. Ethical approval and consent to participate.", "2.4.2. Conflict of interest.", "3. Results", "3.1. Demographic and clinical presentation of patients with gastroenteritis", "3.2. Macroscopic and microscopic examination of the studied stool samples of all included patients", "3.3. Comparison between conventional methods and multiplex-PCR with regard to the recovery of the studied pathogens in the 200 included patients", "3.4. Diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of studied pathogen", "4. Discussion", "5. Conclusion", "5.1. Study limitation", "Author contributions" ]
[ "Infectious gastroenteritis (IG) is one of the most common diseases worldwide, killing millions of individuals each year. In industrialized countries, IG remains a major public health burden, although mortality is low. However, mortality can be found in developing countries, where epidemics of bacterial gastroenteritis may develop.[1]\nShigella, Campylobacter and enterohemorrhagic E coli are three common intestinal pathogens involved in gastroenteritis and are important to study because of the frequency and severity of symptoms they cause. EHEC identification would also be critical in the event of food-borne illness outbreaks or agroterrorism, thus the accurate and rapid diagnosis is a must in outbreaks to direct therapeutic intervention.[2]\nAeromonas causes diarrheal infections, most commonly in children (especially those under 5 years) and in immune-compromised patients and the incidence of gastroenteritis tends to be higher in summer than other seasons. The organism has been also isolated with high frequency from patients with traveler’s diarrhea.[3] Acute self-limiting diarrhea occurs in children, and chronic gastroenteritis or enterocolitis may occur in children and the elderly. The presentation of gastroenteritis caused by Aeromonas includes various combinations of fever, vomiting, and increased fecal leucocytes or erythrocytes.[4]\nMost medical microbiology laboratories use traditional and conventional methods for detection of infectious cause of diarrhea, among direct microscopic examination, the use of conventional cultures and identification of organisms using biochemical reactions, which take a long time; results can release after 3 to 4 days.[2] Furthermore, misidentification of culture increases hands-on time and delay in reporting of a definite negative result. Other problems are the viable but non-culturable state of Campylobacter jejuni[5] and the limited viability of Shigella outside the human body. These may compromise the sensitivity of culture.[6] Another value of the accurate and rapid diagnosis of gastroenteritis is the rapid start of therapeutic intervention as just mentioned before. It is known that bacterial gastroenteritis is a self-limiting disease, and antimicrobial therapy is not particularly required. Clinically, it is not possible to distinguish pathogens that will respond to antibiotics from those that will not respond. Antibiotics add to the cost of treatment, put the patient at risk for adverse events, and can encourage development of resistant bacteria.[7] However, in certain infections e.g. dysentery, cholera, and for certain cases of persistent diarrhea particularly in infants, elderly people, and immunocompromised individuals who are at higher risk of developing more severe complications, treatment can decrease the duration and severity of illness if it is initiated early in the course of infection.[8]\nMolecular methods are sensitive and rapid methods compared to conventional methods. However, broad application remains limited due to their assumed high costs, inhibition caused by fecal constituents, and the need for specialized laboratories. Due to the high throughput of stool screening and the number of possible enteric pathogens, implementation of a molecular approach which uses multiplexing of targets is mandatory.[9] The objective of this study is to evaluate the multiplex PCR for the rapid detection of four major intestinal pathogens causing gastroenteritis (Shigella spp., Campylobacter spp., Aeromonas spp. and Enterohemorrhagic Escherichia coli (EHEC) in stool specimens compared to other conventional culture methods.", "[SUBTITLE] 2.1. Patient selection [SUBSECTION] A total of 200 stool samples were collected randemly from patients attended Helwan, South Valley and Tanta Universities outpatient clinics with clinical manifestations of acute gastroenteritis; between February 2019 and December 2021. For each stool sample, a portion of the sample was transferred into a sterile tube and stored at −20°C for molecular diagnostics. The rest was used for stool culture process within 1 hours of receipt. All patients were subjected to full history taking, lying stress on manifestations of gastroenteritis (diarrhea, vomiting, fever and abdominal discomfort), bleeding tendency, hematuria and history of antibiotic intake. All patients aged from 18 to 60 years with clinical manifestations of acute gastroenteritis were included in the study. Patients with history of antibiotics intake, chronic diarrhea of any cause, cancer anywhere in the body including cancer colon, drug intake and immunosuppression due to any cause were excluded from the study.\nA total of 200 stool samples were collected randemly from patients attended Helwan, South Valley and Tanta Universities outpatient clinics with clinical manifestations of acute gastroenteritis; between February 2019 and December 2021. For each stool sample, a portion of the sample was transferred into a sterile tube and stored at −20°C for molecular diagnostics. The rest was used for stool culture process within 1 hours of receipt. All patients were subjected to full history taking, lying stress on manifestations of gastroenteritis (diarrhea, vomiting, fever and abdominal discomfort), bleeding tendency, hematuria and history of antibiotic intake. All patients aged from 18 to 60 years with clinical manifestations of acute gastroenteritis were included in the study. Patients with history of antibiotics intake, chronic diarrhea of any cause, cancer anywhere in the body including cancer colon, drug intake and immunosuppression due to any cause were excluded from the study.\n[SUBTITLE] 2.2. Microbiological processing of stool samples [SUBSECTION] [SUBTITLE] 2.2.1. Sample processing. [SUBSECTION] Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.\n Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.\n[SUBTITLE] 2.2.2. Sample culture. [SUBSECTION] Using a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]\nUsing a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]\n[SUBTITLE] 2.2.1. Sample processing. [SUBSECTION] Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.\n Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.\n[SUBTITLE] 2.2.2. Sample culture. [SUBSECTION] Using a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]\nUsing a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]\n[SUBTITLE] 2.3. Multiplex PCR extraction and amplification [SUBSECTION] A multiplex PCR was performed (by 2 clinical pathology doctors with more than 5 year experience) on all stool specimens for the detection of Shigella spp., Campylobacter spp.., Aeromonas spp. and E coli O157 DNA. After thawing the samples, 1 mL (for liquid stool) or 1 gram (for solid feces) was used for the extraction of bacterial DNA using the QIA amp DAN Mini Kit supplied by (Qiagen, Germantown, MD) according to the manufacturer’s instructions. The bacterial genomes were amplified by multiplex real-time PCR using the Seeplex® Diarrhea ACE Detection kits (Seegene, Republic of [South Korea]) were used for the multiplex PCR test. The bacterial panels included:\na) Diarrhea B1 ACE detection is a multiplex assay that permits the simultaneous amplification of target DNA of: Shigella spp. (S flexneri, S boydii, S sonnei and S dysenteriae), Campylobacter spp. (C jejuni and C coli) and Internal Control (IC) Figure 1.\nMultiplex PCR using the ACE B1 diarrhea kit (A) ACEB1 marker (B) Combined Shigella and Campylobacter (bp: 330 and 227 respectively), (C) Negative control. PCR = polymerase chain reaction.\nb) Diarrhea B2 ACE Detection is a multiplex assay that permits the simultaneous amplification of target DNA of: Aeromonas spp. (A salmonicida, A sobria, A bivalvium, A hydrophila), E coli O157:H7, VTEC family and Internal Control (IC). The target genes were respectively IpaH, Rfb, Hip and Hly for Shigella, EHEC, Campylobacter and Aeromonas Figure 2.\nMultiplex PCR using the ACE B2 diarrhea kit showing (A) and (B) Aeromonas spp. (bp: 217), (C) ACEB2 marker. PCR = polymerase chain reaction.\nShigella dysenteriae (Shiga) (ATCC® 13313) was used as a positive control and Staphylococcus aureus subsp. aureus Rosenbach (ATCC® 25923™) was used as a negative control\nA multiplex PCR was performed (by 2 clinical pathology doctors with more than 5 year experience) on all stool specimens for the detection of Shigella spp., Campylobacter spp.., Aeromonas spp. and E coli O157 DNA. After thawing the samples, 1 mL (for liquid stool) or 1 gram (for solid feces) was used for the extraction of bacterial DNA using the QIA amp DAN Mini Kit supplied by (Qiagen, Germantown, MD) according to the manufacturer’s instructions. The bacterial genomes were amplified by multiplex real-time PCR using the Seeplex® Diarrhea ACE Detection kits (Seegene, Republic of [South Korea]) were used for the multiplex PCR test. The bacterial panels included:\na) Diarrhea B1 ACE detection is a multiplex assay that permits the simultaneous amplification of target DNA of: Shigella spp. (S flexneri, S boydii, S sonnei and S dysenteriae), Campylobacter spp. (C jejuni and C coli) and Internal Control (IC) Figure 1.\nMultiplex PCR using the ACE B1 diarrhea kit (A) ACEB1 marker (B) Combined Shigella and Campylobacter (bp: 330 and 227 respectively), (C) Negative control. PCR = polymerase chain reaction.\nb) Diarrhea B2 ACE Detection is a multiplex assay that permits the simultaneous amplification of target DNA of: Aeromonas spp. (A salmonicida, A sobria, A bivalvium, A hydrophila), E coli O157:H7, VTEC family and Internal Control (IC). The target genes were respectively IpaH, Rfb, Hip and Hly for Shigella, EHEC, Campylobacter and Aeromonas Figure 2.\nMultiplex PCR using the ACE B2 diarrhea kit showing (A) and (B) Aeromonas spp. (bp: 217), (C) ACEB2 marker. PCR = polymerase chain reaction.\nShigella dysenteriae (Shiga) (ATCC® 13313) was used as a positive control and Staphylococcus aureus subsp. aureus Rosenbach (ATCC® 25923™) was used as a negative control\n[SUBTITLE] 2.4. Statistical analysis [SUBSECTION] Analysis was done using the IBM SPSS statistics (V. 20.0, IBM Corp., Chicago, USA, 2011). Descriptive Statistics: Qualitative data were presented as counts and percentage. The association between each 2 variables or comparison between 2 independent groups as regards the categorized data was done using the Chi square test or the Fisher exact test. The probability of error at > .05 was considered non-significant, while ≤ .05 was considered significant and < .01 was considered highly significant.\n[SUBTITLE] 2.4.1. Ethical approval and consent to participate. [SUBSECTION] The study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).\nThe study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).\n[SUBTITLE] 2.4.2. Conflict of interest. [SUBSECTION] We don’t receive any financial support or relationships that may pose conflict of interest in this work.\nWe don’t receive any financial support or relationships that may pose conflict of interest in this work.\nAnalysis was done using the IBM SPSS statistics (V. 20.0, IBM Corp., Chicago, USA, 2011). Descriptive Statistics: Qualitative data were presented as counts and percentage. The association between each 2 variables or comparison between 2 independent groups as regards the categorized data was done using the Chi square test or the Fisher exact test. The probability of error at > .05 was considered non-significant, while ≤ .05 was considered significant and < .01 was considered highly significant.\n[SUBTITLE] 2.4.1. Ethical approval and consent to participate. [SUBSECTION] The study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).\nThe study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).\n[SUBTITLE] 2.4.2. Conflict of interest. [SUBSECTION] We don’t receive any financial support or relationships that may pose conflict of interest in this work.\nWe don’t receive any financial support or relationships that may pose conflict of interest in this work.", "A total of 200 stool samples were collected randemly from patients attended Helwan, South Valley and Tanta Universities outpatient clinics with clinical manifestations of acute gastroenteritis; between February 2019 and December 2021. For each stool sample, a portion of the sample was transferred into a sterile tube and stored at −20°C for molecular diagnostics. The rest was used for stool culture process within 1 hours of receipt. All patients were subjected to full history taking, lying stress on manifestations of gastroenteritis (diarrhea, vomiting, fever and abdominal discomfort), bleeding tendency, hematuria and history of antibiotic intake. All patients aged from 18 to 60 years with clinical manifestations of acute gastroenteritis were included in the study. Patients with history of antibiotics intake, chronic diarrhea of any cause, cancer anywhere in the body including cancer colon, drug intake and immunosuppression due to any cause were excluded from the study.", "[SUBTITLE] 2.2.1. Sample processing. [SUBSECTION] Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.\n Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.\n[SUBTITLE] 2.2.2. Sample culture. [SUBSECTION] Using a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]\nUsing a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]", " Two Microbiology doctors with more than 5-year experience were responsible for stool samples processing as a routine test in Microbiology department of Helwan, South Valley and Tanta Universities.\nStool specimen in a wide—necked container was processed within 1 hour from its arrival to the lab. Macroscopy of stool samples will be done to check for characteristic color, presence of blood and mucous. Wet smear of stool samples was examined microscopically for presence of pus, RBCs and parasitic causes as protozoa trophozoite, ameba cyst. Basic fuchsine stain (10 gm/L) for 10 to 20 seconds was used for rapid examination of Campylobacter spp. which appears as small, S-shaped, gram-negative, non-spore forming rods.", "Using a sterile inoculation loop, a loopful from the stool specimen (from areas with blood or mucous, if present) was inoculated onto a plate of MacConkey agar (Oxoid Discs, Oxoid, North Shore, United Kingdom) for Enterobacteriaceae, a plate of XLD agar selective media for Salmonella and Shigella (Oxoid) and a plate of Skirrow’s agar (Oxoid) for Campylobacter spp. isolation. The inoculated XLD agar and MacConkey agar were then incubated at 36°C ± 1°C for 18 to 48 hours under aerobic conditions. The inoculated Skirrow’s agar medium was incubated at 42°C ± 1°C under microaerophilic conditions for 48 to 72 hours. A loopful of the stool specimen was additionally inoculated onto a tube of Selenite F broth, for maximum recovery of Salmonella and Shigella is obtained when inoculating an enrichment broth in addition to primary direct plating of specimens, and was incubated at 36°C ± 1°C under aerobic conditions for 18 hours. After 18 hours incubation, a subculture was done from the surface of the Selenite F broth on XLD and MacConkey agar media and the plates were incubated for 48 hours at 36°C ± 1°C under aerobic conditions. Except for the Skirrow’s agar plate, which was left to be examined at the end of the incubation period, other plates were examined daily for the presence of growing microorganisms. Growing colonies were further identified by traditional biochemical reactions (triple sugar iron agar, citrate, urase, oxidase, lysine iron agar and motility indole ornithine) and sub cultured on a blood agar plate for further identification by API 10S (Biomerieux, Marcy-l'Étoile, France). When cases of severe bloody diarrhea were implicated, sorbitol MacConkey media (Liofilchem, Roseto degli Abruzzi, Italy) was used to detect E coli O157:H7 (most O157:H7 E coli are sorbitol-negative so appear colorless).[10]", "A multiplex PCR was performed (by 2 clinical pathology doctors with more than 5 year experience) on all stool specimens for the detection of Shigella spp., Campylobacter spp.., Aeromonas spp. and E coli O157 DNA. After thawing the samples, 1 mL (for liquid stool) or 1 gram (for solid feces) was used for the extraction of bacterial DNA using the QIA amp DAN Mini Kit supplied by (Qiagen, Germantown, MD) according to the manufacturer’s instructions. The bacterial genomes were amplified by multiplex real-time PCR using the Seeplex® Diarrhea ACE Detection kits (Seegene, Republic of [South Korea]) were used for the multiplex PCR test. The bacterial panels included:\na) Diarrhea B1 ACE detection is a multiplex assay that permits the simultaneous amplification of target DNA of: Shigella spp. (S flexneri, S boydii, S sonnei and S dysenteriae), Campylobacter spp. (C jejuni and C coli) and Internal Control (IC) Figure 1.\nMultiplex PCR using the ACE B1 diarrhea kit (A) ACEB1 marker (B) Combined Shigella and Campylobacter (bp: 330 and 227 respectively), (C) Negative control. PCR = polymerase chain reaction.\nb) Diarrhea B2 ACE Detection is a multiplex assay that permits the simultaneous amplification of target DNA of: Aeromonas spp. (A salmonicida, A sobria, A bivalvium, A hydrophila), E coli O157:H7, VTEC family and Internal Control (IC). The target genes were respectively IpaH, Rfb, Hip and Hly for Shigella, EHEC, Campylobacter and Aeromonas Figure 2.\nMultiplex PCR using the ACE B2 diarrhea kit showing (A) and (B) Aeromonas spp. (bp: 217), (C) ACEB2 marker. PCR = polymerase chain reaction.\nShigella dysenteriae (Shiga) (ATCC® 13313) was used as a positive control and Staphylococcus aureus subsp. aureus Rosenbach (ATCC® 25923™) was used as a negative control", "Analysis was done using the IBM SPSS statistics (V. 20.0, IBM Corp., Chicago, USA, 2011). Descriptive Statistics: Qualitative data were presented as counts and percentage. The association between each 2 variables or comparison between 2 independent groups as regards the categorized data was done using the Chi square test or the Fisher exact test. The probability of error at > .05 was considered non-significant, while ≤ .05 was considered significant and < .01 was considered highly significant.\n[SUBTITLE] 2.4.1. Ethical approval and consent to participate. [SUBSECTION] The study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).\nThe study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).\n[SUBTITLE] 2.4.2. Conflict of interest. [SUBSECTION] We don’t receive any financial support or relationships that may pose conflict of interest in this work.\nWe don’t receive any financial support or relationships that may pose conflict of interest in this work.", "The study has been performed in accordance with the Declaration of Helsinki. The study protocol was approved by South Valley Faculty of Medicine Ethical Committee code number (SVU-MED-GIT023-4-22-9-447).", "We don’t receive any financial support or relationships that may pose conflict of interest in this work.", "[SUBTITLE] 3.1. Demographic and clinical presentation of patients with gastroenteritis [SUBSECTION] In this prospective randomized cohort study, between February 2019 and December 2021. a total of 200 stool samples were collected from patients presented with acute gastroenteritis; including 120 female samples and 80 male samples with their age ranging from 18 years to 55 years. One hundred eighty (90%) of patients suffered from watery diarrhea while (10%) had bloody diarrhea. ninety patients (45%) had associated abdominal cramps/colic, (50/200, 25%) had vomiting, (40/200, 20%) had dehydration, (20/200, 10%) had steatorrhea and (4/200, 2%) complained from weight loss Table 1.\nDemographic and clinical presentation of patients with gastroenteritis.\nIn this prospective randomized cohort study, between February 2019 and December 2021. a total of 200 stool samples were collected from patients presented with acute gastroenteritis; including 120 female samples and 80 male samples with their age ranging from 18 years to 55 years. One hundred eighty (90%) of patients suffered from watery diarrhea while (10%) had bloody diarrhea. ninety patients (45%) had associated abdominal cramps/colic, (50/200, 25%) had vomiting, (40/200, 20%) had dehydration, (20/200, 10%) had steatorrhea and (4/200, 2%) complained from weight loss Table 1.\nDemographic and clinical presentation of patients with gastroenteritis.\n[SUBTITLE] 3.2. Macroscopic and microscopic examination of the studied stool samples of all included patients [SUBSECTION] The macroscopic examination in the studied stool samples showed that (160/200, 80%) of samples were watery in consistency whereas (24/200, 12%) were semi-formed. Regarding the stool color (66%) of the studied samples were brownish, (20/200, 10%) were whitish, (22/200, 11%) were greenish, (12/200, 6%) were reddish, and (14/200, 7%) yellowish. Out of the 200 studied samples, 136 samples (68%) contained visible mucous and 12 samples (6%) were bloody. As regard to the microscopic examination, (40/200, 20%) of the studied samples had a WBCs count < 10 cells/HPF, (90/200, 45%) had a WBCs count 10 to 49 cells/HPF, (40/200, 20%) had a WBCs count 50 to 99 cells/HPF, and (36/200, 18%) had a WBCs count > 100 cells/HPF. Three out of the 200 studied samples (5.5%) contained red blood cells > 100/HPF Table 2.\nMacroscopic and microscopic examination of the studied stool samples of all included patients.\nHPF = high power field, RBCs = red blood cells, WBC = white blood cells.\nThe macroscopic examination in the studied stool samples showed that (160/200, 80%) of samples were watery in consistency whereas (24/200, 12%) were semi-formed. Regarding the stool color (66%) of the studied samples were brownish, (20/200, 10%) were whitish, (22/200, 11%) were greenish, (12/200, 6%) were reddish, and (14/200, 7%) yellowish. Out of the 200 studied samples, 136 samples (68%) contained visible mucous and 12 samples (6%) were bloody. As regard to the microscopic examination, (40/200, 20%) of the studied samples had a WBCs count < 10 cells/HPF, (90/200, 45%) had a WBCs count 10 to 49 cells/HPF, (40/200, 20%) had a WBCs count 50 to 99 cells/HPF, and (36/200, 18%) had a WBCs count > 100 cells/HPF. Three out of the 200 studied samples (5.5%) contained red blood cells > 100/HPF Table 2.\nMacroscopic and microscopic examination of the studied stool samples of all included patients.\nHPF = high power field, RBCs = red blood cells, WBC = white blood cells.\n[SUBTITLE] 3.3. Comparison between conventional methods and multiplex-PCR with regard to the recovery of the studied pathogens in the 200 included patients [SUBSECTION] Using conventional culture and identification methods, (192 out of 200, 96%) studied stool samples gave positive results. The most commonly identified organisms were E coli (118/200, 59%), Shigella (54/200, 27%) and Aeromonas species (20/200, 10%). Campylobacter was not revealed in cultures.\nUsing specific E coli O157 anti-sera, 16 isolates out of 118 identified E coli (13.56%) were found to belong to the O157 serotype, and the remaining isolates (102/118, 86.4%) were the normal gut E coli microbiota.\nThus, the EHEC represented 8% (16/200) of the total identified pathogens by conventional culture method.\nUsing multiplex PCR, 173/ 200 fecal samples (86.5%) were positive. Shigella spp. was the most commonly identified pathogen as it was detected in (81/200, 40.5%) of the PCR-positive samples; Out of the 81 multiplex PCR-positive Shigella spp., 27 samples (27/81, 33.3%) were not detected by conventional methods.\nAeromonas spp. was detected in 60 samples (60/200, 30%); out of the 60 positive samples., forty samples (40/60, 66.67%) were missed by the conventional culture methods.\nEHEC were detected in (30/200, 15%) samples. Of them 14 samples were missed by conventional culture method.\nCampylobacter species were also detected in only 2 samples (2/200, 1%); Campylobacter spp. isolate was only detected by the multiplex PCR Figure 3.\nComparison between conventional culture methods and multiplex-PCR as regard to the recovery of the pathogens in the 200 included patients. EHEC = Enterohemorrhagic Escherichia coli, PCR = polymerase chain reaction.\nFinally, Comparison between conventional methods and multiplex-PCR as regard to the recovery of the pathogens in the 200 included patients. Multiplex-PCR appeared to be more advantageous to conventional detection methods regarding the detection of Shigella, EHEC and Aeromonas isolates with a statistically significant difference (P < .05).\nAlthough, no significant difference was detected between conventional culture methods and Multiplex-PCR regarding detection of Campylobacter, but this pathogen was only detected by Multiplex-PCR method Table 3.\nComparison between conventional methods and multiplex-PCR with regard to the recovery of the studied pathogens in the 200 included patients.\nPCR = polymerase chain reaction.\nχ2: Chi-square test.\nS: P value < .05 is considered significant.\nHS: P value < .001 is considered highly significant.\nNS: P value > .05 is considered non-significant.\nUsing conventional culture and identification methods, (192 out of 200, 96%) studied stool samples gave positive results. The most commonly identified organisms were E coli (118/200, 59%), Shigella (54/200, 27%) and Aeromonas species (20/200, 10%). Campylobacter was not revealed in cultures.\nUsing specific E coli O157 anti-sera, 16 isolates out of 118 identified E coli (13.56%) were found to belong to the O157 serotype, and the remaining isolates (102/118, 86.4%) were the normal gut E coli microbiota.\nThus, the EHEC represented 8% (16/200) of the total identified pathogens by conventional culture method.\nUsing multiplex PCR, 173/ 200 fecal samples (86.5%) were positive. Shigella spp. was the most commonly identified pathogen as it was detected in (81/200, 40.5%) of the PCR-positive samples; Out of the 81 multiplex PCR-positive Shigella spp., 27 samples (27/81, 33.3%) were not detected by conventional methods.\nAeromonas spp. was detected in 60 samples (60/200, 30%); out of the 60 positive samples., forty samples (40/60, 66.67%) were missed by the conventional culture methods.\nEHEC were detected in (30/200, 15%) samples. Of them 14 samples were missed by conventional culture method.\nCampylobacter species were also detected in only 2 samples (2/200, 1%); Campylobacter spp. isolate was only detected by the multiplex PCR Figure 3.\nComparison between conventional culture methods and multiplex-PCR as regard to the recovery of the pathogens in the 200 included patients. EHEC = Enterohemorrhagic Escherichia coli, PCR = polymerase chain reaction.\nFinally, Comparison between conventional methods and multiplex-PCR as regard to the recovery of the pathogens in the 200 included patients. Multiplex-PCR appeared to be more advantageous to conventional detection methods regarding the detection of Shigella, EHEC and Aeromonas isolates with a statistically significant difference (P < .05).\nAlthough, no significant difference was detected between conventional culture methods and Multiplex-PCR regarding detection of Campylobacter, but this pathogen was only detected by Multiplex-PCR method Table 3.\nComparison between conventional methods and multiplex-PCR with regard to the recovery of the studied pathogens in the 200 included patients.\nPCR = polymerase chain reaction.\nχ2: Chi-square test.\nS: P value < .05 is considered significant.\nHS: P value < .001 is considered highly significant.\nNS: P value > .05 is considered non-significant.\n[SUBTITLE] 3.4. Diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of studied pathogen [SUBSECTION] The majority of infections were caused by a single pathogen. However, multiple enteric pathogens in individuals’ samples were detected by the multiplex PCR not by conventional methods.\nOnly three samples contain mixed infections with the four studied pathogen.\nThe diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Shigella, EHEC and Aeromonas showed, sensitivity of 100% (for each), specificity of 88.5%, 92.4%, 77.8% respectively, PPV of 66.7%, 53.3%, and 33.3%, respectively and NPV of 100% (for each). However, the diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Campylobacter showed specificity of 99% and NPV of 100%; (sensitivity and PPV cannot be calculated as no Campylobacter isolate identified by conventional method; which considered as gold standard for diagnosis of bacterial infection) Table 4.\nDiagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of studied pathogen.\nNPV = negative predictive value, PCR = polymerase chain reaction, PPV = positive predictive value.\nThe majority of infections were caused by a single pathogen. However, multiple enteric pathogens in individuals’ samples were detected by the multiplex PCR not by conventional methods.\nOnly three samples contain mixed infections with the four studied pathogen.\nThe diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Shigella, EHEC and Aeromonas showed, sensitivity of 100% (for each), specificity of 88.5%, 92.4%, 77.8% respectively, PPV of 66.7%, 53.3%, and 33.3%, respectively and NPV of 100% (for each). However, the diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Campylobacter showed specificity of 99% and NPV of 100%; (sensitivity and PPV cannot be calculated as no Campylobacter isolate identified by conventional method; which considered as gold standard for diagnosis of bacterial infection) Table 4.\nDiagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of studied pathogen.\nNPV = negative predictive value, PCR = polymerase chain reaction, PPV = positive predictive value.", "In this prospective randomized cohort study, between February 2019 and December 2021. a total of 200 stool samples were collected from patients presented with acute gastroenteritis; including 120 female samples and 80 male samples with their age ranging from 18 years to 55 years. One hundred eighty (90%) of patients suffered from watery diarrhea while (10%) had bloody diarrhea. ninety patients (45%) had associated abdominal cramps/colic, (50/200, 25%) had vomiting, (40/200, 20%) had dehydration, (20/200, 10%) had steatorrhea and (4/200, 2%) complained from weight loss Table 1.\nDemographic and clinical presentation of patients with gastroenteritis.", "The macroscopic examination in the studied stool samples showed that (160/200, 80%) of samples were watery in consistency whereas (24/200, 12%) were semi-formed. Regarding the stool color (66%) of the studied samples were brownish, (20/200, 10%) were whitish, (22/200, 11%) were greenish, (12/200, 6%) were reddish, and (14/200, 7%) yellowish. Out of the 200 studied samples, 136 samples (68%) contained visible mucous and 12 samples (6%) were bloody. As regard to the microscopic examination, (40/200, 20%) of the studied samples had a WBCs count < 10 cells/HPF, (90/200, 45%) had a WBCs count 10 to 49 cells/HPF, (40/200, 20%) had a WBCs count 50 to 99 cells/HPF, and (36/200, 18%) had a WBCs count > 100 cells/HPF. Three out of the 200 studied samples (5.5%) contained red blood cells > 100/HPF Table 2.\nMacroscopic and microscopic examination of the studied stool samples of all included patients.\nHPF = high power field, RBCs = red blood cells, WBC = white blood cells.", "Using conventional culture and identification methods, (192 out of 200, 96%) studied stool samples gave positive results. The most commonly identified organisms were E coli (118/200, 59%), Shigella (54/200, 27%) and Aeromonas species (20/200, 10%). Campylobacter was not revealed in cultures.\nUsing specific E coli O157 anti-sera, 16 isolates out of 118 identified E coli (13.56%) were found to belong to the O157 serotype, and the remaining isolates (102/118, 86.4%) were the normal gut E coli microbiota.\nThus, the EHEC represented 8% (16/200) of the total identified pathogens by conventional culture method.\nUsing multiplex PCR, 173/ 200 fecal samples (86.5%) were positive. Shigella spp. was the most commonly identified pathogen as it was detected in (81/200, 40.5%) of the PCR-positive samples; Out of the 81 multiplex PCR-positive Shigella spp., 27 samples (27/81, 33.3%) were not detected by conventional methods.\nAeromonas spp. was detected in 60 samples (60/200, 30%); out of the 60 positive samples., forty samples (40/60, 66.67%) were missed by the conventional culture methods.\nEHEC were detected in (30/200, 15%) samples. Of them 14 samples were missed by conventional culture method.\nCampylobacter species were also detected in only 2 samples (2/200, 1%); Campylobacter spp. isolate was only detected by the multiplex PCR Figure 3.\nComparison between conventional culture methods and multiplex-PCR as regard to the recovery of the pathogens in the 200 included patients. EHEC = Enterohemorrhagic Escherichia coli, PCR = polymerase chain reaction.\nFinally, Comparison between conventional methods and multiplex-PCR as regard to the recovery of the pathogens in the 200 included patients. Multiplex-PCR appeared to be more advantageous to conventional detection methods regarding the detection of Shigella, EHEC and Aeromonas isolates with a statistically significant difference (P < .05).\nAlthough, no significant difference was detected between conventional culture methods and Multiplex-PCR regarding detection of Campylobacter, but this pathogen was only detected by Multiplex-PCR method Table 3.\nComparison between conventional methods and multiplex-PCR with regard to the recovery of the studied pathogens in the 200 included patients.\nPCR = polymerase chain reaction.\nχ2: Chi-square test.\nS: P value < .05 is considered significant.\nHS: P value < .001 is considered highly significant.\nNS: P value > .05 is considered non-significant.", "The majority of infections were caused by a single pathogen. However, multiple enteric pathogens in individuals’ samples were detected by the multiplex PCR not by conventional methods.\nOnly three samples contain mixed infections with the four studied pathogen.\nThe diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Shigella, EHEC and Aeromonas showed, sensitivity of 100% (for each), specificity of 88.5%, 92.4%, 77.8% respectively, PPV of 66.7%, 53.3%, and 33.3%, respectively and NPV of 100% (for each). However, the diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Campylobacter showed specificity of 99% and NPV of 100%; (sensitivity and PPV cannot be calculated as no Campylobacter isolate identified by conventional method; which considered as gold standard for diagnosis of bacterial infection) Table 4.\nDiagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of studied pathogen.\nNPV = negative predictive value, PCR = polymerase chain reaction, PPV = positive predictive value.", "In Africa, Asia, and South America, diarrhea accounts for 13% of deaths annually.[11]\nSymptomatic treatment is the aim of therapy of acute gastroenteritis; antibiotic therapy required in specific critical situation; however, one of the great problems is a long duration needed till stool culture results become available, so its impact on treatment is minimal.[12,13]\nStool culture is the gold standard for identification of bacteria present in the stool. However, its time consuming and require specific culture agars followed by biochemical, morphologic, and serologic testing to confirm the culture isolate.[14–18]\nIt’s known that multiplex PCR, is time saving allow early and multiple gastrointestinal infections identification, this is of great important for early and accurate treatment of acute bacterial gastroenteritis especially in critically ill patients.[19]\nTherefore, we aim in this study to evaluate efficacy of multiplex PCR method, as a rabid and accurate method for diagnosis of the four common enteropathogenic bacteria responsible for fatal gastroenteritis; that could be missed when using conventional laboratory methods.\nTo our knowledge, this study is the first one done in Egypt using multiplex-PCR for accurate diagnosis of the four common bacterial pathogens responsible for adults’ infectious gastroenteritis.\nIn this study, using conventional culture; E coli were the most commonly identified organisms it is detected in more than half of samples, followed by Shigella (27%), finally, Aeromonas species which detected in only 10% of samples.\nThis come in agreement with El Hassan et al,[20] who reported that, E coli was the most commonly isolated enteropathogen from diarrheal stool; being responsible for about 72% of cases of diarrhea. Followed by Shigella spp. which detected in 8% of stool samples.\nYet, our results are much higher than those found in Saudi Arabia by El-Sheikh and El-Assouli[21] who detected EHEC in about 2% of cases presenting with diarrhea. This may be related to his study on the prevalence of different causes (viral, bacterial, and parasitic) of acute diarrhea among children with in Jeddah. In addition to different worldwide prevalence of pathogens responsible for infectious gastroenteritis.\nBoth Shigella spp. and Enterohemorrhagic E-Coli (EHEC) are intestinal pathogen that presented with dysentery; however, development of fatal complications is commonly reported with shigella spp.[22,23] So, discrimination between Shigella spp. and EHEC is of clinical and epidemiological importance, for rapid and early outbreak control.[24]\nIn our study, using multiplex PCR, 86.5% of fecal samples were positive. Shigella spp., was the most commonly identified pathogen as it was detected in about 40% of the PCR-positive samples, with 27 samples detected by using multiplex-PCR and missed by conventional culture methods. Also, Aeromonas spp. and EHEC were detected by multiplex-PCR in 60 (30%) and 30 (15%) samples respectively with 40 samples and 14 samples respectively were missed by the conventional culture methods.\nOur results are in agreement with O’Leary et al,[2] who compared between multiplex PCR and conventional diagnostic methods for accurate detection of bacterial pathogen from feces of patients with acute gastroenteritis; they found that, all positive results by conventional methods were matched by positive results with multiplex PCR with additional 17 positive results detected by multiplex PCR.\nOur results come in agreement with Zaki and El-Adrosy[25] at Mansoura, Egypt who reported that EHEC was positive by PCR in 25% of patients affected in an outbreak of diarrhea.\nAlso, Gray et al,[26] reported that using multiplex-PCR provide both improved recovery of pathogens and detection of pathogens unable to be tested by conventional tests.\nAt the same time, Dixit et al,[27] by using the same multiplex PCR commercial kit as in our study, Shigella spp. was the most detected organism, followed by Campylobacter and E coli. Also, Mota et al,[28] who investigated the bacterial causes of bloody diarrhea by both conventional and molecular methods, Shigella was recovered from most cases, while EHEC was detected in 1% of cases.\nLikewise, Guan et al and Thiem et al,[29,30] who found that multiplex PCR has a higher detection rate for EHEC and Shigella spp. than conventional culture methods. Also, two previous studies in India[31] and Thailand[32] suggest that stool culture has a lower sensitivity than PCR in the diagnosis of Shigella spp.\nShigella spp. frequently escape detection by traditional culture methods. However, detection of the four species of Shigella by real-time PCR, targeting the invasion plasmid antigen H (ipaH) gene; can mask the EIEC detection in stool by real-time PCR, which carry the same target gene. IpaH is carried by all four Shigella species as well as by enteroinvasive Escherichia coli (EIEC). Because EIEC is rare in fecal specimens from patients with diarrhea, it is thought that most organisms detected by ipaH-specific PCR are Shigella spp.[30] Multiplex PCR can be modified to identify EIEC in cultures from ipaH-positive samples by an assay with two sets of multiplex PCR reactions that differentiates Shigella and EIEC based on the presence/absence of at least two out of six loci, the majority of Shigella genomes lacked all six loci, while at least two loci were present in most EIEC genomes.[33]\nDifferent studies done to confirm the volubility of molecular technique over conventual culture method in discriminating between Shigella spp. and diarrheagenic E-Coli.\nLikewise, Islam et al,[34] in Bangladesh, who found that conventional culture methods for the isolation of Shigella spp. had a sensitivity of 72% however, when the PCR technique targeting the ipaH gene it was considered as the gold standard. They recommended PCR to be employed in routine diagnosis of dysentery in clinical centers as well as in epidemiologic studies. Also, Dutta et al,[31] agreed with that of Islam et al,[34] and added that the PCR assays can further identify a number of non-typable Shigella strains, which would have remained undiagnosed if PCR had not been used.[31,34]\nMoreover, Thiem et al and Wang et al[30,35] found that real-time PCR targeting the ipaH gene; detected Shigella spp. in 58% of randomly selected Shigella culture negative specimens and in 97% of Shigella culture positive specimens. The authors concluded that the high detection rate of ipaH gene in culture negative specimens through use of real-time PCR suggests that earlier estimates of shigellosis burden measured by conventional culture may have underestimated the true disease burden.\nCampylobacter species isolate in Egypt was more frequent than Salmonella, Shigella and other enteric bacterial pathogens.[36] Also, WHO reported that Campylobacter was the most common bacterial cause of human gastroenteritis in the world; about 3 to 4 times more frequent than Salmonella and E coli.[37]\nOne of the great problems of Campylobacter infection is its serious long-term complications, including the peripheral neuropathy (Guillain–Barré syndrome and Miller Fisher syndrome) temporary paralysis, arthritis and irritable bowel syndrome.[38]\nIn our study Campylobacter spp., was isolated only by using multiplex-PCR in two male patients presented with bloody diarrhea.\nThis comes with White et al,[39] who found that 4% of patients presented with bloody diarrhea, had campylobacteriosis, and considered older patients are more likely to have severe illness and be hospitalized.\nAlso, Barakat et al,[40] reported that multiplex PCR is a valuable method for detection of virulent and resistant genes of Campylobacter bacteria.\nIn this study, the diagnostic evaluation of multiplex PCR in relation to conventional method in diagnosis of Shigella, EHEC and Aeromonas showed sensitivity of 100% each and specificity of 88.5%, 92.4%, and 77.8% respectively. However, the diagnostic evaluation of multiplex PCR for Campylobacter showed specificity of 99% but sensitivity cannot be calculated (as no cases detected by conventional culture method).\nThis nearly come in agreement with Rundell et al who found in his study that, multiplex PCR had a sensitivity of for detection of Campylobacter, EHEC and Shigella spp. of 100%, 96.8%, and 97.6% respectively; and specificity of 100%, 99.5%, and 100% respectively depending on the causative bacteria.[41] Also, Our results are in agreement with O’Leary et al,[2] who compared between multiplex PCR and conventional diagnostic methods for the simultaneous detection of Campylobacter spp., Shigella spp., and E coli O157 from feces. The sensitivity for multiplex PCR was found to be 100%, the specificity was 99.3%, the positive predictive value was 91.5%, and the negative predictive value was 100%.", "Multiplex PCR can overcome limitations of culture-based methods as it is a more rapid and to some extent more accurate method with higher sensitivity compared to the traditional cultured based methods for detection of common intestinal pathogens.\n[SUBTITLE] 5.1. Study limitation [SUBSECTION] No reevaluation of patients after antimicrobial therapy as multiplex PCR panels can’t differentiate between viable and dead organisms, especially in patients with multiple stools detected organisms, which could be related to colonic colonization with asymptomatic organisms.\nSince no controls were included in this study and enteropathogens have often been detected in healthy controls, the detection didn’t necessarily mean disease association.\nNo reevaluation of patients after antimicrobial therapy as multiplex PCR panels can’t differentiate between viable and dead organisms, especially in patients with multiple stools detected organisms, which could be related to colonic colonization with asymptomatic organisms.\nSince no controls were included in this study and enteropathogens have often been detected in healthy controls, the detection didn’t necessarily mean disease association.", "No reevaluation of patients after antimicrobial therapy as multiplex PCR panels can’t differentiate between viable and dead organisms, especially in patients with multiple stools detected organisms, which could be related to colonic colonization with asymptomatic organisms.\nSince no controls were included in this study and enteropathogens have often been detected in healthy controls, the detection didn’t necessarily mean disease association.", "Conceptualization: Karim Montasser, Heba Ahmed Osman, and Abeer M. M. Sabry.\nData curation: Karim Montasser, Heba Ahmed Osman, Abeer M. M. Sabry, Haidy S. Khalil, Hanan Abozaid, and Wesam Hatem Amer.\nFormal analysis: Karim Montasser\nInvestigation: Karim Montasser, Heba Ahmed Osman, Hanan Abozaid, Haidy S. Khalil, Wesam Hatem Amer, and Abeer M. M. Sabry.\nMethodology: Karim Montasser, Heba Ahmed Osman, Abeer M. M. Sabry, Haidy S. Khalil, Hanan Abozaid, and Wesam Hatem Amer.\nProject administration: Karim Montasser.\nWriting – original draft: Wesam Hatem Amer, Hanan Abozaid, and Haidy S. Khalil.\nWriting – review & editing: Karim Montasser, Heba Ahmed Osman, and Abeer M. M. Sabry" ]
[ "intro", "methods", "subjects", null, null, null, null, null, null, null, "results", "subjects", "subjects", "subjects", "methods", "discussion", null, null, null ]
[ "Enterohemorrhagic E coli O157 spp", "multiplex PCR", "Shigella spp", "stool culture" ]
Risk factors of postoperative hydrocephalus following decompressive craniectomy for spontaneous intracranial hemorrhages and intraventricular hemorrhage.
36254070
Hydrocephalus is a complication of spontaneous intracerebral hemorrhage; however, its predictive relationship with hydrocephalus in this patient cohort is not understood. Here, we evaluated the incidence and risk factors of hydrocephalus after craniectomy.
INTRODUCTION
Retrospectively studied data from 39 patients in the same hospital from 2016/01 to 2020/12 and analyzed risk factors for hydrocephalus. The clinical data recorded included patient age, sex, timing of surgery, initial Glasgow Coma Scale score, intracerebral hemorrhage (ICH) score, alcohol consumption, cigarette smoking, medical comorbidity, and blood data. Predictors of patient outcomes were determined using Student t test, chi-square test, and logistic regression.
METHODS
We recruited 39 patients with cerebral herniation who underwent craniectomy for spontaneous supratentorial hemorrhage. Persistent hydrocephalus was observed in 17 patients. The development of hydrocephalus was significantly associated with the timing of operation, cigarette smoking, and alcohol consumption according to the Student t test and chi-square test. Univariate and multivariate analyses suggested that postoperative hydrocephalus was significantly associated with the timing of surgery (P = .031) and cigarette smoking (P = .041).
RESULTS
The incidence of hydrocephalus in patients who underwent delayed operation (more than 4 hours) was lower than that in patients who underwent an operation after less than 4 hours. nonsmoking groups also have lower incidence of hydrocephalus. Among patients who suffered from spontaneous supratentorial hemorrhage and need to receive emergent craniectomy, physicians should be reminded that postoperative hydrocephalus followed by ventriculoperitoneal shunting may be necessary in the future.
DISCUSSION
[ "Cerebral Hemorrhage", "Decompressive Craniectomy", "Humans", "Hydrocephalus", "Intracranial Hemorrhages", "Postoperative Complications", "Retrospective Studies", "Risk Factors", "Treatment Outcome" ]
9575832
1. Introduction
Spontaneous non-traumatic intracranial hemorrhage (ICH) is the second most common form of stroke (approximately 15–30% of all strokes). It is also the deadliest disease and has high morbidity and mortality rates. The main causes of spontaneous ICH include poorly controlled hypertension, acutely increased cerebral blood flow, vascular anomalies, and coagulopathies, such as antiplatelet agents. Neurological deterioration after the initial hemorrhage is usually due to a combination of rebleeding, cerebral edema, seizures, increased intracranial pressure (ICP), and hydrocephalus [1,2]; these are related to poor functional outcomes and morbidity. The initial management of spontaneous ICH includes blood pressure management, anti-epileptic drugs, and hemostasis. If the patient has coagulopathy or is undergoing anticoagulant therapy, correction with coagulopathy is necessary. Evaluation of surgical intervention is important, including in patients with a Glasgow Coma Scale (GCS) score ≤8, evidence of transtentorial herniation, or significant intraventricular hemorrhage or hydrocephalus. These factors should be considered in ICP monitoring and further surgical treatment. The removal of intracranial hematomas has many clinical benefits, such as the prevention of damage to the brain stem, cerebral herniation, controlled ICP management, and a decrease in excitotoxicity and neurotoxicity of blood products.[3] Decompressive craniectomy with hematoma evacuation may play a role in comatose patients with significant midline shift and large hematomas on brain computerized tomography (CT) scans or patients with refractory increased ICP,[4] which is defined as ICP > 20 mm Hg for >15 minutes in a 1-hour period refractory to first-tier therapies, surgical decompression is suggested. External ventricular drainage (EVD) is the procedure of choice for the treatment of acute hydrocephalus and increased ICP in patients with intracerebral hemorrhage and intraventricular hemorrhage.[5] After initial operative management, the patient was admitted to an intensive care unit. There are many complications of spontaneous ICH, such as cerebral edema, rebleeding, seizure attacks, and hydrocephalus. Hydrocephalus is a common condition after spontaneous ICH, and post-hemorrhage is the second most common cause of non-obstructive hydrocephalus, especially in patients who undergo decompressive craniectomy (risk factor for hydrocephalus in patients with brain injury).[6] The patients with intraventricular hemorrhage have a higher risk of developing permanent hydrocephalus and requiring shunting operations.[7] However, there are no efficient risk factors for predicting the incidence of hydrocephalus following spontaneous ICH status after decompressive craniectomy. Other risk factors for hydrocephalus in patients who have undergone decompressive craniectomy for spontaneous ICH have not been reported. Here, we retrospectively analyzed data from patients who underwent decompressive craniectomy for spontaneous ICH to identify the risk factors for postoperative hydrocephalus.
2. Materials and Methods
We studied patients with spontaneous intracranial hematoma with intraventricular hemorrhage and ventricular extension, suspicious acute hydrocephalus and mass effect who received decompressive craniectomy and EVD for medically refractory increased ICP at Tri-Service General hospital, Taipei, Taiwan from January 2016 to December 2020; 39 patients were included. Due to different pathophysiologies, pressure dynamics, and neurologic symptoms and signs of hydrocephalus in supra- and infra-tentorial hemorrhage, we only included patients with supratentorial hemorrhage. A flow chart of the study design is shown in Figure 1. Patient data were collected in accordance with the tenets of the Declaration of Helsinki. This retrospective study was approved by the institutional review committee of the Tri-Service General Hospital. Other patients were excluded for the following reasons: tumor bleeding, post-infarct hemorrhagic transformation, or death due to cardiopulmonary disorders. All patients were monitored in the intensive care unit and received medical treatment and management for ICP control (head elevation by 30°, anti-epileptic medication, sedation, etc). Decompressive craniectomy was performed when the patients had increased ICP and signs of brain stem herniation and intraventricular hemorrhage on brain CT. The intracerebral blood clot was almost completely removed during the operation. Ventriculoperitoneal (VP) shunting was performed after the patients showed signs of hydrocephalus during hospitalization (failed weaning of external ventricular draining, conscious disturbance after removal of EVD, etc). Evidence of hydrocephalus included failed weaning from EVD. During hospitalization, we tried to clamp the EVD and intensively monitored the patient’s clinical feature and conscious status.[8] Under the critical care of post decompressive craniectomy, we set the EVD at level of 10 cm H2O over the foramen of Monroe; in order to force cerebrospinal fluid (CSF) flow through EVD in patients with ICP elevations, EVD could temporarily be lowered to the levels between 0 and 5 cm H2O. We will try to wean the EVD based on the relative vital signs, neurological function, and stable intracranial pressure after 1 week. The EVD systems were gradually raised in 5 cm steps every 24 hour up to a final level of 25 cm H2O, provided that it was clinically well tolerated. In case of successful weaning, EVD was subsequently closed for 48 hour. If the patient cannot tolerate the treatment, they may suffer from conscious disturbance, seizure, or other neurological deficits, and a VP shunt is suggested. The weaning period was approximately 2 weeks.[9] Radiographic data on serial brain CT of ventricular dilation included the frontal horns, temporal horns, and third ventricle. A combination of unilateral ventricular dilation due to encephalomalacia and a normal-sized contralateral ventricle was defined as ventriculomegaly versus hydrocephalus. An Evans ratio of at least 0.3 may be consistent with a diagnosis of hydrocephalus (Fig. 2).[10] The case of presentation of our clinical courses is in Figure 3. Clinical data and a series of brain CT scans for each patient were collected, as shown. The clinical data included patient age, sex, timing of operation, initial GCS, ICH score, alcohol consumption, cigarette smoking, hemodialysis, anticoagulant agent usage, history of cancer, previous stroke, heart valve diseases, type 2 diabetes mellitus, C-reactive protein (CRP), and albumin levels. The flow chart of study designs. Evans ratios, the ratio of maximum width of the frontal horn to the maximum width of the inner table of the cranium recorded on the side contralateral to the decompressive craniectomy. (A) The 20th patient in our study, is a 66-year-old male, had a GCS of E2M4V2 when he was arrived to hospital. He suffered from spontaneous ICH with brain stem herniation over the left basal ganglion and intraventricular hemorrhage. (B) He received decompressive craniectomy and EVD after he was transferred to our hospital or about 445 min later. Brain CT scan on the 7th day after operation showed nearly complete hematoma evacuation and no hydrocephalus after we clamped the EVD. We then removed the EVD on the 13th day after operation. The GCS showed E3M6VT (T: tracheostomy) when he was transferred to the ordinary ward; the patient didn’t have symptoms of hydrocephalus on further follow up. CT = computerized tomography, EVD = external ventricular drainage, ICH = intracranial hemorrhage, GCS = Glasgow Coma Scale. Univariate analysis was conducted to identify the association of each explanatory factor with the condition of hydrocephalus after intracranial hemorrhage (i.e., with or without hydrocephalus) using Student t test and the chi-square test with Fisher exact test for continuous and categorical data, respectively. Logistic regression was employed to determine predictors in the multivariate analysis, as described previously. A P value of .05 or less indicated a significant statistical difference.
3. Result
A total of 39 patients underwent decompressive craniectomy for spontaneous ICH with cerebral herniation. Indications for decompressive craniectomy in our study included emergency conditions with herniation or deteriorating symptoms. This study included 14 women and 25 men with spontaneous ICH. The baseline patient characteristics are presented in Table 1. The patients’ ages ranged from 35 to 79 years (mean: 58.56 years). The mean spontaneous ICH volume was 66 mL. The delay from the emergency department to craniectomy varied: the hydrocephalus group was 112 to 552 minutes (mean: 232.12 minute), and the non-hydrocephalus group was 107 to 969 minutes (mean: 371.5 minute). Postoperative hydrocephalus developed on serial brain CT scans in 17 patients after decompressive craniectomy. The duration from craniectomy to ventriculoperitoneal shunting (VP shunting) ranged from 6 to 30 days (mean: 14.5 days). The demographic data of the 39 patients who underwent decompressive craniectomy due to spontaneous ICH are summarized in Table 2. The average follow-up period of our patients was approximately 12 weeks, and we plan to arrange cranioplasty after adequate clinical conditions. No patient developed hydrocephalus at a later time point. The baseline characteristics of the patients. CRP = C-reactive protein, VP shunting = ventriculoperitoneal shunting. Demographic data of 39 patients with decompressive craniectomy due to spontaneous ICH. Drink = alcohol drinking, GCS = Glasgow Coma Scale, ICH = intracranial hemorrhage, MLS = midline shift, OP = operation, Pre-DC = pre-decompressive craniectomy, smoke = cigarette smoking. Comorbidity, including hemodialysis, hypertension, Anticoagulant usage, heart valve diseases, previous stroke, type 2 diabetes mellitus, hyperlipidemia. Y: yes, N: no. The development of postoperative hydrocephalus was not significantly associated with patient age, sex, GCS score on admission, ICH score, hemodialysis, anticoagulant agent usage, history of cancer, previous stroke, heart valve diseases, type 2 diabetes mellitus, CRP, and albumin (Table 3). However, the timing of surgery, cigarette smoking, and alcohol consumption were significantly associated with postoperative hydrocephalus (Table 2). Univariate and multivariate logistic regression analyses were used to evaluate the data presented in Table 4. Patients who underwent ultra-early operation (less than 4 hours) were likely to have a higher incidence of hydrocephalus than those who underwent an operation more than 4 hours ago (P valve: .038; odds ratio (OR) 6.79, 95% confidence ratio (CI) 1.19–38.57). Patients who smoked cigarettes were associated with postoperative hydrocephalus (P valve: .021; OR, 14.27; 95% CI:1.12–181.79). The distribution of demography and clinical characteristic by treatment. CT = computerized tomography, CRP = C-reactive protein, GCS = Glasgow Coma Scale, M ± SD = mean ± deviation, spontaneous ICH = spontaneous intracranial hemorrhage. Mann–Whitney U test or chi-square test. Fisher exact test. Univariate and multivariate of logistic regression analysis. CI = confidence interval, OR = odds ratio, ref = reference group.
null
null
[ "5. Conclusion", "Author contributions" ]
[ "Hydrocephalus is a complication in patients who are suffered from spontaneous ICH with brain stem herniation. It most commonly occurs at the onset of spontaneous ICH. Ultra-delayed operation (more than 4 hours) and non-cigarette smoking showed a relatively lower risk of postoperative hydrocephalus. Physicians should remember the higher incidence of postoperative hydrocephalus before ultra-early operation for spontaneous ICH. Patients suffering from spontaneous ICH with brain stem herniation need to go to the operating room as soon as possible for decompressive craniectomy, but physicians should remind patients and their families with a higher incidence of postoperative hydrocephalus. Therefore, VP shunting may be necessary in the future.", "Yi-Chieh Wu, Jang-Chun Lin, Yu-Ching Chou and Wei-Hsiu Liu was responsible for designing this research. Hsiang-Chih Liao, Da-Tong Ju, Dueng-Yuan Hueng, Chi-Tun Tang, Kuan-Yin Tseng, Kuan-Nien Chou, Bon-Jour Lin and Shao-Wei Feng extracted the data and conducted the statistical analysis. Yi- An Chen, Ming-Hsuan Chung and Peng- Wei Wang drafted the manuscript." ]
[ null, null ]
[ "1. Introduction", "2. Materials and Methods", "3. Result", "4. Discussion", "5. Conclusion", "Author contributions" ]
[ "Spontaneous non-traumatic intracranial hemorrhage (ICH) is the second most common form of stroke (approximately 15–30% of all strokes). It is also the deadliest disease and has high morbidity and mortality rates. The main causes of spontaneous ICH include poorly controlled hypertension, acutely increased cerebral blood flow, vascular anomalies, and coagulopathies, such as antiplatelet agents. Neurological deterioration after the initial hemorrhage is usually due to a combination of rebleeding, cerebral edema, seizures, increased intracranial pressure (ICP), and hydrocephalus [1,2]; these are related to poor functional outcomes and morbidity.\nThe initial management of spontaneous ICH includes blood pressure management, anti-epileptic drugs, and hemostasis. If the patient has coagulopathy or is undergoing anticoagulant therapy, correction with coagulopathy is necessary. Evaluation of surgical intervention is important, including in patients with a Glasgow Coma Scale (GCS) score ≤8, evidence of transtentorial herniation, or significant intraventricular hemorrhage or hydrocephalus. These factors should be considered in ICP monitoring and further surgical treatment. The removal of intracranial hematomas has many clinical benefits, such as the prevention of damage to the brain stem, cerebral herniation, controlled ICP management, and a decrease in excitotoxicity and neurotoxicity of blood products.[3] Decompressive craniectomy with hematoma evacuation may play a role in comatose patients with significant midline shift and large hematomas on brain computerized tomography (CT) scans or patients with refractory increased ICP,[4] which is defined as ICP > 20 mm Hg for >15 minutes in a 1-hour period refractory to first-tier therapies, surgical decompression is suggested. External ventricular drainage (EVD) is the procedure of choice for the treatment of acute hydrocephalus and increased ICP in patients with intracerebral hemorrhage and intraventricular hemorrhage.[5] After initial operative management, the patient was admitted to an intensive care unit. There are many complications of spontaneous ICH, such as cerebral edema, rebleeding, seizure attacks, and hydrocephalus. Hydrocephalus is a common condition after spontaneous ICH, and post-hemorrhage is the second most common cause of non-obstructive hydrocephalus, especially in patients who undergo decompressive craniectomy (risk factor for hydrocephalus in patients with brain injury).[6] The patients with intraventricular hemorrhage have a higher risk of developing permanent hydrocephalus and requiring shunting operations.[7] However, there are no efficient risk factors for predicting the incidence of hydrocephalus following spontaneous ICH status after decompressive craniectomy.\nOther risk factors for hydrocephalus in patients who have undergone decompressive craniectomy for spontaneous ICH have not been reported. Here, we retrospectively analyzed data from patients who underwent decompressive craniectomy for spontaneous ICH to identify the risk factors for postoperative hydrocephalus.", "We studied patients with spontaneous intracranial hematoma with intraventricular hemorrhage and ventricular extension, suspicious acute hydrocephalus and mass effect who received decompressive craniectomy and EVD for medically refractory increased ICP at Tri-Service General hospital, Taipei, Taiwan from January 2016 to December 2020; 39 patients were included. Due to different pathophysiologies, pressure dynamics, and neurologic symptoms and signs of hydrocephalus in supra- and infra-tentorial hemorrhage, we only included patients with supratentorial hemorrhage. A flow chart of the study design is shown in Figure 1. Patient data were collected in accordance with the tenets of the Declaration of Helsinki. This retrospective study was approved by the institutional review committee of the Tri-Service General Hospital. Other patients were excluded for the following reasons: tumor bleeding, post-infarct hemorrhagic transformation, or death due to cardiopulmonary disorders. All patients were monitored in the intensive care unit and received medical treatment and management for ICP control (head elevation by 30°, anti-epileptic medication, sedation, etc). Decompressive craniectomy was performed when the patients had increased ICP and signs of brain stem herniation and intraventricular hemorrhage on brain CT. The intracerebral blood clot was almost completely removed during the operation. Ventriculoperitoneal (VP) shunting was performed after the patients showed signs of hydrocephalus during hospitalization (failed weaning of external ventricular draining, conscious disturbance after removal of EVD, etc). Evidence of hydrocephalus included failed weaning from EVD. During hospitalization, we tried to clamp the EVD and intensively monitored the patient’s clinical feature and conscious status.[8] Under the critical care of post decompressive craniectomy, we set the EVD at level of 10 cm H2O over the foramen of Monroe; in order to force cerebrospinal fluid (CSF) flow through EVD in patients with ICP elevations, EVD could temporarily be lowered to the levels between 0 and 5 cm H2O. We will try to wean the EVD based on the relative vital signs, neurological function, and stable intracranial pressure after 1 week. The EVD systems were gradually raised in 5 cm steps every 24 hour up to a final level of 25 cm H2O, provided that it was clinically well tolerated. In case of successful weaning, EVD was subsequently closed for 48 hour. If the patient cannot tolerate the treatment, they may suffer from conscious disturbance, seizure, or other neurological deficits, and a VP shunt is suggested. The weaning period was approximately 2 weeks.[9] Radiographic data on serial brain CT of ventricular dilation included the frontal horns, temporal horns, and third ventricle. A combination of unilateral ventricular dilation due to encephalomalacia and a normal-sized contralateral ventricle was defined as ventriculomegaly versus hydrocephalus. An Evans ratio of at least 0.3 may be consistent with a diagnosis of hydrocephalus (Fig. 2).[10] The case of presentation of our clinical courses is in Figure 3. Clinical data and a series of brain CT scans for each patient were collected, as shown. The clinical data included patient age, sex, timing of operation, initial GCS, ICH score, alcohol consumption, cigarette smoking, hemodialysis, anticoagulant agent usage, history of cancer, previous stroke, heart valve diseases, type 2 diabetes mellitus, C-reactive protein (CRP), and albumin levels.\nThe flow chart of study designs.\nEvans ratios, the ratio of maximum width of the frontal horn to the maximum width of the inner table of the cranium recorded on the side contralateral to the decompressive craniectomy.\n(A) The 20th patient in our study, is a 66-year-old male, had a GCS of E2M4V2 when he was arrived to hospital. He suffered from spontaneous ICH with brain stem herniation over the left basal ganglion and intraventricular hemorrhage. (B) He received decompressive craniectomy and EVD after he was transferred to our hospital or about 445 min later. Brain CT scan on the 7th day after operation showed nearly complete hematoma evacuation and no hydrocephalus after we clamped the EVD. We then removed the EVD on the 13th day after operation. The GCS showed E3M6VT (T: tracheostomy) when he was transferred to the ordinary ward; the patient didn’t have symptoms of hydrocephalus on further follow up. CT = computerized tomography, EVD = external ventricular drainage, ICH = intracranial hemorrhage, GCS = Glasgow Coma Scale.\nUnivariate analysis was conducted to identify the association of each explanatory factor with the condition of hydrocephalus after intracranial hemorrhage (i.e., with or without hydrocephalus) using Student t test and the chi-square test with Fisher exact test for continuous and categorical data, respectively. Logistic regression was employed to determine predictors in the multivariate analysis, as described previously. A P value of .05 or less indicated a significant statistical difference.", "A total of 39 patients underwent decompressive craniectomy for spontaneous ICH with cerebral herniation. Indications for decompressive craniectomy in our study included emergency conditions with herniation or deteriorating symptoms. This study included 14 women and 25 men with spontaneous ICH. The baseline patient characteristics are presented in Table 1. The patients’ ages ranged from 35 to 79 years (mean: 58.56 years). The mean spontaneous ICH volume was 66 mL. The delay from the emergency department to craniectomy varied: the hydrocephalus group was 112 to 552 minutes (mean: 232.12 minute), and the non-hydrocephalus group was 107 to 969 minutes (mean: 371.5 minute). Postoperative hydrocephalus developed on serial brain CT scans in 17 patients after decompressive craniectomy. The duration from craniectomy to ventriculoperitoneal shunting (VP shunting) ranged from 6 to 30 days (mean: 14.5 days). The demographic data of the 39 patients who underwent decompressive craniectomy due to spontaneous ICH are summarized in Table 2. The average follow-up period of our patients was approximately 12 weeks, and we plan to arrange cranioplasty after adequate clinical conditions. No patient developed hydrocephalus at a later time point.\nThe baseline characteristics of the patients.\nCRP = C-reactive protein, VP shunting = ventriculoperitoneal shunting.\nDemographic data of 39 patients with decompressive craniectomy due to spontaneous ICH.\nDrink = alcohol drinking, GCS = Glasgow Coma Scale, ICH = intracranial hemorrhage, MLS = midline shift, OP = operation, Pre-DC = pre-decompressive craniectomy, smoke = cigarette smoking. Comorbidity, including hemodialysis, hypertension, Anticoagulant usage, heart valve diseases, previous stroke, type 2 diabetes mellitus, hyperlipidemia. Y: yes, N: no.\nThe development of postoperative hydrocephalus was not significantly associated with patient age, sex, GCS score on admission, ICH score, hemodialysis, anticoagulant agent usage, history of cancer, previous stroke, heart valve diseases, type 2 diabetes mellitus, CRP, and albumin (Table 3). However, the timing of surgery, cigarette smoking, and alcohol consumption were significantly associated with postoperative hydrocephalus (Table 2). Univariate and multivariate logistic regression analyses were used to evaluate the data presented in Table 4. Patients who underwent ultra-early operation (less than 4 hours) were likely to have a higher incidence of hydrocephalus than those who underwent an operation more than 4 hours ago (P valve: .038; odds ratio (OR) 6.79, 95% confidence ratio (CI) 1.19–38.57). Patients who smoked cigarettes were associated with postoperative hydrocephalus (P valve: .021; OR, 14.27; 95% CI:1.12–181.79).\nThe distribution of demography and clinical characteristic by treatment.\nCT = computerized tomography, CRP = C-reactive protein, GCS = Glasgow Coma Scale, M ± SD = mean ± deviation, spontaneous ICH = spontaneous intracranial hemorrhage.\nMann–Whitney U test or chi-square test.\nFisher exact test.\nUnivariate and multivariate of logistic regression analysis.\nCI = confidence interval, OR = odds ratio, ref = reference group.", "Our study aimed to determine the predictors of hydrocephalus in patients with spontaneous ICH. We found at least 4 predictors for a high risk of hydrocephalus: timing of operation, cigarette smoking, and alcohol drinking. These factors were statistically significant in predicting the incidence of hydrocephalus.\nWe reviewed the literature on hydrocephalus and decompressive craniectomy. Some studies hypothesized that the abnormal collections of hygromas, hydrocephalus, and subgaleal hygromas after decompressive craniectomy are caused by altered brain pulsatility, CSF hydrodynamics, decreased cerebral blood flow, and impaired brain glymphatic clearance in a vulnerable subset of patients..[11] Extension to the ventricles was the only independent risk factor for hydrocephalus (4–13 days), while extension to ventricles, decompressive craniotomy, and intracranial infection were independent predictors of hydrocephalus (≥14 days).[12] However, we found that some patients who underwent decompressive craniectomy due to spontaneous ICH had a higher risk of hydrocephalus. Another study suggested a link between decompressive craniectomy and hydrocephalus in the setting of traumatic brain centers on the intracranial dura-arachnoid interface, where shearing forces from the primary injury may critically interrupt CSF resorption systems.[13] When such a disruption is followed by the characteristically large craniectomy required for trauma management, the abnormal resulting transcerebral and intracranial pressure gradients allow for a marked expansion of all the subdural spaces given the pressure-dependent nature of the proposed mechanism.[13]\nWe attempted to explain the association between cigarette smoking or alcohol consumption and the higher risk of postoperative hydrocephalus. There is no strong evidence that smoking history or alcohol consumption is associated with a higher risk of hydrocephalus following decompressive craniectomy in spontaneous ICH. This may be associated with the side effects of ethanol, nicotine, or tar oil. However, further studies are required for further evaluation. According to the literature, smoking is associated with cerebrospinal fluid shunt in patients with idiopathic intracranial hypertension (Investigative Ophthalmology & Visual Science, 2013), It was also found that smokers had a greater odds of undergoing cerebrospinal fluid shunt compared to nonsmokers. The reason for this remains unclear. Although the mechanism of smoking and hydrocephalus is still unclear, we can also inform patients that smoking may be a risk factor for hydrocephalus after intracranial hemorrhage and encourage them to quit smoking.\nThe group that required more than four hours after spontaneous ICH symptoms to reach the operating room had a lower incidence of hydrocephalus. We sometimes need to arrange the operation as soon as possible because of significant brain stem herniation or refractory increased ICP, but there is higher evidence of hydrocephalus in this group. According to the journal, rebleeding leads to worse outcomes in ultra-early craniotomy for spontaneous ICH.[14] Rebleeding was more common in patients who underwent surgery within four hours compared with 12 hours.[14] The 22th patient is a case who experienced rebleeding and underwent reoperation for hematoma removal due to significant mass effect and neurological deficit (Fig. 4). There was a relationship between rebleeding and mortality in the 4-hour surgery group. More rebleeding might influence hydrocephalus in follow-up studies; we surveyed the residual intracranial blood volume postoperatively following brain CT. More residual hematoma volumes after surgery have a significant association with hydrocephalus following decompressive craniectomy, which may be linked to a higher rebleeding rate after surgery. However, further evidence is required for further evaluation.\n(A) The 22nd patient in our study, is a 73-year-old male with GCS of E2M4V1 when he was arrived to hospital, suffered from spontaneous right putamen ICH. (B) He received decompressive craniectomy and EVD, and the timing of the operation was about 122 min. However, he was still in a coma, and repeated brain CT data showed a rebleeding hematoma on day 3. (C) The patient was received reoperation of removal of hematoma and check bleeding. After the operation, his GCS showed improved (E1M2VT to E2M4VT). The patient suffered from worsening neurological symptoms after we clamp the EVD tube. The following brain CT revealed persistent hydrocephalus with dilated lateral ventricle; he then received the VP shunting operation. CT = computerized tomography, EVD = external ventricular drainage, ICH = intracranial hemorrhage, GCS = Glasgow Coma Scale, VP shunting = ventriculoperitoneal shunting.\nOur study has some limitations. A major limitation of our study is that it was retrospective non-randomized and single. These factors can lead to information bias owing to unclear data collection. Second, we did not explain all of the results; they need to be evaluated further to understand mechanisms such as rebleeding with hydrocephalus and the influence of ethanol, nicotine, and tar oil. Therefore, more clinical studies are needed to explore the effectiveness of this treatment in patients who undergo decompressive craniectomy. Another limitation is that we excluded patients who died after decompressive craniectomy. The patient who expired during the postoperative care may have multiple possible factors, such as infections, infarction, cerebrovascular failure, or the effect of hydrocephalus. We cannot easily distinguish these complex possibilities, and we need to research more evidence and modify our study to improve it. However, we did not discuss the relationship between hydrocephalus and postoperative complications of decompressive craniectomy, such as infection, pneumocephalus, or sinking flap syndrome. Rebleeding was the only post-operative complication that we studied. This is our limitation and we will evaluate this issue in future studies.", "Hydrocephalus is a complication in patients who are suffered from spontaneous ICH with brain stem herniation. It most commonly occurs at the onset of spontaneous ICH. Ultra-delayed operation (more than 4 hours) and non-cigarette smoking showed a relatively lower risk of postoperative hydrocephalus. Physicians should remember the higher incidence of postoperative hydrocephalus before ultra-early operation for spontaneous ICH. Patients suffering from spontaneous ICH with brain stem herniation need to go to the operating room as soon as possible for decompressive craniectomy, but physicians should remind patients and their families with a higher incidence of postoperative hydrocephalus. Therefore, VP shunting may be necessary in the future.", "Yi-Chieh Wu, Jang-Chun Lin, Yu-Ching Chou and Wei-Hsiu Liu was responsible for designing this research. Hsiang-Chih Liao, Da-Tong Ju, Dueng-Yuan Hueng, Chi-Tun Tang, Kuan-Yin Tseng, Kuan-Nien Chou, Bon-Jour Lin and Shao-Wei Feng extracted the data and conducted the statistical analysis. Yi- An Chen, Ming-Hsuan Chung and Peng- Wei Wang drafted the manuscript." ]
[ "intro", "methods", "results", "discussion", null, null ]
[ "decompressive craniectomy", "postoperative hydrocephalus", "spontaneous intracranial hemorrhage" ]
The efficacy of irinotecan supplementation for colorectal cancer: A meta-analysis of randomized controlled studies.
36254072
The efficacy of irinotecan as the adjunctive therapy to fluorouracil and leucovorin remains controversial in patients with colorectal cancer. We conduct this meta-analysis to explore the efficacy of irinotecan supplementation for colorectal cancer.
BACKGROUND
We have searched PubMed, EMBASE, Web of science, EBSCO, and Cochrane library databases through March 19, 2020, and included randomized controlled trials assessing the efficacy of irinotecan plus fluorouracil and leucovorin for colorectal cancer.
METHODS
Five randomized controlled trials were included in the meta-analysis. Compared with fluorouracil and leucovorin for colorectal cancer, irinotecan supplementation could significantly improve progression-free survival rate (hazard ratio = 0.72; 95% confidence interval [CI] = 0.58-0.90; P = .003), median progression-free survival (standard mean difference = -0.30; 95% CI = -0.44 to -0.15; P < .0001), overall survival rate (hazard ratio = 0.77; 95% CI = 0.66-0.90; P = .001), and objective response (risk ratio [RR] = 0.57; 95% CI = 0.49-0.66; P < .00001) and decrease progressive disease (RR = 2.10; 95% CI = 1.40-3.14; P = .0003), but revealed no obvious effect on complete response (RR = 0.88; 95% CI = 0.33-2.29; P = .79). The incidence of grade ≥3 adverse events in irinotecan supplementation group was increased compared to control group (RR = 0.67; 95% CI = 0.57-0.79; P < .00001).
RESULTS
Irinotecan as the adjunctive therapy to fluorouracil and leucovorin can increase the survival and objective response of patients with colorectal cancer, but the incidence of grade ≥3 adverse events is found to be increased after irinotecan supplementation.
CONCLUSIONS
[ "Humans", "Antineoplastic Combined Chemotherapy Protocols", "Camptothecin", "Colorectal Neoplasms", "Dietary Supplements", "Fluorouracil", "Irinotecan", "Leucovorin", "Randomized Controlled Trials as Topic" ]
9575722
1. Introduction
Colorectal cancer is regarded as a significant cause of mortality.[1–3] The prognosis of these patients is determined by the stages of colorectal cancer, and 5-year survival rates of stage I, II, and III after surgical intervention are 93.2%, 82.5%, and 59.5%, respectively. Especially, 5-year survival rate of stage IV is only 8.1%.[4] Many patients with resected cancer may suffer from recurrence.[5,6] Fluorouracil and leucovorin have been widely used for colorectal cancer,[7] and are reported to reduce the recurrence rate and improve survival.[8] In order to improve the treatment efficacy, irinotecan or oxaliplatin is used combined with fluorouracil and leucovorin, and these combinations are generally regarded as the effective approach for advanced colorectal cancer.[9,10] In elderly patients, irinotecan or oxaliplatin in combination with fluorouracil is well tolerated and shows similar efficacy between elderly and younger patients.[11] In metastatic colorectal cancer, combining irinotecan with fluorouracil results in a remarkable increase in progression-free survival and overall survival than fluorouracil alone.[12] However, current evidence is insufficient for routine use of irinotecan supplementation for colorectal cancer, and several studies have reported the conflicting results of irinotecan supplementation for colorectal cancer.[4,9,13,14] This meta-analysis aims to assess the efficacy and safety of irinotecan in combination with fluorouracil and leucovorin for colorectal cancer.
2. Materials and methods
This meta-analysis was performed based on the Preferred Reporting Items for Systematic Reviews and Meta-analysis statement and Cochrane Handbook for Systematic Reviews of Interventions.[15,16] No ethical approval and patient consent were required because all analyses were based on previously published studies. [SUBTITLE] 2.1. Literature search [SUBSECTION] We have systematically searched several databases including PubMed, EMBASE, Web of science, EBSCO, and the Cochrane library from inception to March 19, 2020 with the following keywords: “irinotecan” AND “fluorouracil” AND “leucovorin” AND “colorectal cancer” OR “colon cancer” OR “rectal cancer”. The inclusion criteria were as follows: study design was RCT, patients were diagnosed with colorectal cancer, and intervention treatments were irinotecan plus fluorouracil and leucovorin versus only fluorouracil and leucovorin. Patients who previously received pelvic radiotherapy were excluded. We have systematically searched several databases including PubMed, EMBASE, Web of science, EBSCO, and the Cochrane library from inception to March 19, 2020 with the following keywords: “irinotecan” AND “fluorouracil” AND “leucovorin” AND “colorectal cancer” OR “colon cancer” OR “rectal cancer”. The inclusion criteria were as follows: study design was RCT, patients were diagnosed with colorectal cancer, and intervention treatments were irinotecan plus fluorouracil and leucovorin versus only fluorouracil and leucovorin. Patients who previously received pelvic radiotherapy were excluded. [SUBTITLE] 2.2. Data extraction and outcome measures [SUBSECTION] Some baseline information was extracted, and they included first author, number of patients, age, sex, performance status, primary tumor site (colon/ rectum/both), and detail methods in 2 groups. Data were extracted independently by 2 investigators, and discrepancies were resolved by consensus. The primary outcomes were progression-free survival rate, median progression-free survival, and overall survival rate. Secondary outcomes included objective response, progressive disease, complete response, and grade ≥3 adverse events. Some baseline information was extracted, and they included first author, number of patients, age, sex, performance status, primary tumor site (colon/ rectum/both), and detail methods in 2 groups. Data were extracted independently by 2 investigators, and discrepancies were resolved by consensus. The primary outcomes were progression-free survival rate, median progression-free survival, and overall survival rate. Secondary outcomes included objective response, progressive disease, complete response, and grade ≥3 adverse events. [SUBTITLE] 2.3. Assessment for risk of bias [SUBSECTION] The risk of bias tool was used to assess the quality of individual studies according to the Cochrane Handbook for Systematic Reviews of Interventions,[16] and the sources of bias were divided into selection bias, performance bias, attrition bias, detection bias, reporting bias, and other potential sources of bias. The overall risk of bias for each study was evaluated and rated: low, unclear, and high.[17] Two investigators independently assessed the quality of included studies, and any discrepancy was solved by consensus. The risk of bias tool was used to assess the quality of individual studies according to the Cochrane Handbook for Systematic Reviews of Interventions,[16] and the sources of bias were divided into selection bias, performance bias, attrition bias, detection bias, reporting bias, and other potential sources of bias. The overall risk of bias for each study was evaluated and rated: low, unclear, and high.[17] Two investigators independently assessed the quality of included studies, and any discrepancy was solved by consensus. [SUBTITLE] 2.4. Statistical analysis [SUBSECTION] We assessed hazard ratio (HR) or risk ratio (RR) with 95% confidence interval (CI) for dichotomous outcomes (progression-free survival rate, overall survival rate, objective response, progressive disease, complete response, and grade ≥3 adverse events) and standard mean difference with 95% CI for continuous outcome (median progression-free survival). Heterogeneity was evaluated by the I2 statistic, and I2 > 50% indicated significant heterogeneity.[18] The random-effects model was used when encountering significant heterogeneity, while fixed-effects model was applied when no significant heterogeneity was found. We searched for potential sources of heterogeneity, and sensitivity analysis was performed to detect the influence of a single study on the overall estimate via omitting 1 study in turn or conducting the subgroup analysis. Owing to the limited number (<10) of included studies, publication bias was not assessed. A P value of <.05 was indicated to be statistically significant. All statistical analyses were performed using Review Manager Version 5.3 (The Cochrane Collaboration, Software Update, Oxford, United Kingdom). We assessed hazard ratio (HR) or risk ratio (RR) with 95% confidence interval (CI) for dichotomous outcomes (progression-free survival rate, overall survival rate, objective response, progressive disease, complete response, and grade ≥3 adverse events) and standard mean difference with 95% CI for continuous outcome (median progression-free survival). Heterogeneity was evaluated by the I2 statistic, and I2 > 50% indicated significant heterogeneity.[18] The random-effects model was used when encountering significant heterogeneity, while fixed-effects model was applied when no significant heterogeneity was found. We searched for potential sources of heterogeneity, and sensitivity analysis was performed to detect the influence of a single study on the overall estimate via omitting 1 study in turn or conducting the subgroup analysis. Owing to the limited number (<10) of included studies, publication bias was not assessed. A P value of <.05 was indicated to be statistically significant. All statistical analyses were performed using Review Manager Version 5.3 (The Cochrane Collaboration, Software Update, Oxford, United Kingdom).
3. Results
[SUBTITLE] 3.1. Literature search, study characteristics, and quality assessment [SUBSECTION] Figure 1 shows the detail flowchart of the search and selection results. Five hundred ninety-eight potentially relevant articles were initially identified. Two hundred twenty-three duplicates and 366 papers after checking the titles/abstracts were excluded. Four studies were removed because of different combination drugs, and 5 randomized controlled trials (RCTs) were finally included in the meta-analysis.[4,9,13,14,19] Flow diagram of study searching and selection process. The baseline characteristics of 5 included RCTs are shown in Table 1. These studies were published between 2000 and 2015, and the total sample size was 4536. All included RCTs reported irinotecan as the adjunctive therapy to fluorouracil and leucovorin, and the methods between irinotecan group and control group were different in each RCT, detailed in Table 1. In the study by Saltz,[14] we just extracted the data of study 2 (Douillard) for this meta-analysis in order to avoid the duplicated data of Saltz.[19] Characteristics of included studies. CI = continuous infusion, IV = intravenous. Four studies reported progression-free survival rate,[4,9,13,19] 2 studies reported median progression-free survival,[9,13] 4 studies reported overall survival rate and objective response,[9,13,14,19] 2 studies reported progressive disease and complete response,[9,13] and 3 studies reported grade ≥3 adverse events.[9,13,19] Figure 1 shows the detail flowchart of the search and selection results. Five hundred ninety-eight potentially relevant articles were initially identified. Two hundred twenty-three duplicates and 366 papers after checking the titles/abstracts were excluded. Four studies were removed because of different combination drugs, and 5 randomized controlled trials (RCTs) were finally included in the meta-analysis.[4,9,13,14,19] Flow diagram of study searching and selection process. The baseline characteristics of 5 included RCTs are shown in Table 1. These studies were published between 2000 and 2015, and the total sample size was 4536. All included RCTs reported irinotecan as the adjunctive therapy to fluorouracil and leucovorin, and the methods between irinotecan group and control group were different in each RCT, detailed in Table 1. In the study by Saltz,[14] we just extracted the data of study 2 (Douillard) for this meta-analysis in order to avoid the duplicated data of Saltz.[19] Characteristics of included studies. CI = continuous infusion, IV = intravenous. Four studies reported progression-free survival rate,[4,9,13,19] 2 studies reported median progression-free survival,[9,13] 4 studies reported overall survival rate and objective response,[9,13,14,19] 2 studies reported progressive disease and complete response,[9,13] and 3 studies reported grade ≥3 adverse events.[9,13,19] [SUBTITLE] 3.2. Assessment of risk of bias [SUBSECTION] Risk of bias analysis is presented in Figure 2. These 5 included RCTs generally had high quality although 4 studies had high risk of bias due to their nonblindness.[4,9,14,19] Risk of bias assessment. (A) Authors’ judgments about each risk of bias item for each included study. (B) Authors’ judgments about each risk of bias item are presented as percentages across all included studies. Risk of bias analysis is presented in Figure 2. These 5 included RCTs generally had high quality although 4 studies had high risk of bias due to their nonblindness.[4,9,14,19] Risk of bias assessment. (A) Authors’ judgments about each risk of bias item for each included study. (B) Authors’ judgments about each risk of bias item are presented as percentages across all included studies. [SUBTITLE] 3.3. Primary outcomes: progression-free survival rate, median progression-free survival, and overall survival rate [SUBSECTION] Compared to control group for colorectal cancer, irinotecan supplementation was associated with substantially improved progression-free survival rate (HR = 0.72; 95% CI = 0.58–0.90; P = .003) with significant heterogeneity among the studies (I2 = 88%, heterogeneity P < .0001; Fig. 3), median progression-free survival (standard mean difference = –0.30; 95% CI = –0.44 to –0.15; P < .0001) with no heterogeneity among the studies (I2 = 0%, heterogeneity P = .79; Fig. 4), and overall survival rate (HR = 0.77; 95% CI = 0.66–0.90; P = .001) with no heterogeneity among the studies (I2 = 0%, heterogeneity P = .98; Fig. 5). Forest plot for the meta-analysis of progression-free survival rate. CI = confidence interval, IV = intravenous, SE = standard error. Forest plot for the meta-analysis of median progression-free survival (month). CI = confidence interval, IV = intravenous, SE = standard error. Forest plot for the meta-analysis of overall survival rate. CI = confidence interval, IV = intravenous, SE = standard error. Compared to control group for colorectal cancer, irinotecan supplementation was associated with substantially improved progression-free survival rate (HR = 0.72; 95% CI = 0.58–0.90; P = .003) with significant heterogeneity among the studies (I2 = 88%, heterogeneity P < .0001; Fig. 3), median progression-free survival (standard mean difference = –0.30; 95% CI = –0.44 to –0.15; P < .0001) with no heterogeneity among the studies (I2 = 0%, heterogeneity P = .79; Fig. 4), and overall survival rate (HR = 0.77; 95% CI = 0.66–0.90; P = .001) with no heterogeneity among the studies (I2 = 0%, heterogeneity P = .98; Fig. 5). Forest plot for the meta-analysis of progression-free survival rate. CI = confidence interval, IV = intravenous, SE = standard error. Forest plot for the meta-analysis of median progression-free survival (month). CI = confidence interval, IV = intravenous, SE = standard error. Forest plot for the meta-analysis of overall survival rate. CI = confidence interval, IV = intravenous, SE = standard error. [SUBTITLE] 3.4. Sensitivity analysis [SUBSECTION] There was significant heterogeneity for progression-free survival rate, but no heterogeneity was observed for median progression-free survival or overall survival rate. As shown in Figure 3, the study conducted by Van Cutsem et al[4] showed the results that were almost completely out of range of the others and probably contributed to the heterogeneity. After excluding that study, the results suggested that irinotecan supplementation could also improve progression-free survival rate for colorectal cancer than control intervention (HR = 0.65; 95% CI = 0.63–0.67; P < .00001). No evidence of heterogeneity was observed among the remaining studies (I2 = 0%). There was significant heterogeneity for progression-free survival rate, but no heterogeneity was observed for median progression-free survival or overall survival rate. As shown in Figure 3, the study conducted by Van Cutsem et al[4] showed the results that were almost completely out of range of the others and probably contributed to the heterogeneity. After excluding that study, the results suggested that irinotecan supplementation could also improve progression-free survival rate for colorectal cancer than control intervention (HR = 0.65; 95% CI = 0.63–0.67; P < .00001). No evidence of heterogeneity was observed among the remaining studies (I2 = 0%). [SUBTITLE] 3.5. Secondary outcomes [SUBSECTION] In comparison with control group for colorectal cancer, irinotecan supplementation showed the obvious increase in objective response (RR = 0.57; 95% CI = 0.49–0.66; P < .00001; Fig. 6) and the decrease in progressive disease (RR = 2.10; 95% CI = 1.40–3.14; P = .0003; Fig. 7), but had no substantial impact on complete response (RR = 0.89; 95% CI = 0.37–2.13; P = .79; Fig. 8). In addition, the incidence of grade ≥3 adverse events in irinotecan supplementation group was higher than that in control group (RR = 0.67; 95% CI = 0.57–0.79; P < .00001; Fig. 9). Forest plot for the meta-analysis of objective response. CI = confidence interval, IV = intravenous. Forest plot for the meta-analysis of progressive disease. CI = confidence interval, IV = intravenous. Forest plot for the meta-analysis of complete response. CI = confidence interval, IV = intravenous. Forest plot for the meta-analysis of grade ≥3 adverse events. CI = confidence interval, IV = intravenous. In comparison with control group for colorectal cancer, irinotecan supplementation showed the obvious increase in objective response (RR = 0.57; 95% CI = 0.49–0.66; P < .00001; Fig. 6) and the decrease in progressive disease (RR = 2.10; 95% CI = 1.40–3.14; P = .0003; Fig. 7), but had no substantial impact on complete response (RR = 0.89; 95% CI = 0.37–2.13; P = .79; Fig. 8). In addition, the incidence of grade ≥3 adverse events in irinotecan supplementation group was higher than that in control group (RR = 0.67; 95% CI = 0.57–0.79; P < .00001; Fig. 9). Forest plot for the meta-analysis of objective response. CI = confidence interval, IV = intravenous. Forest plot for the meta-analysis of progressive disease. CI = confidence interval, IV = intravenous. Forest plot for the meta-analysis of complete response. CI = confidence interval, IV = intravenous. Forest plot for the meta-analysis of grade ≥3 adverse events. CI = confidence interval, IV = intravenous.
null
null
[ "2.1. Literature search", "2.2. Data extraction and outcome measures", "2.3. Assessment for risk of bias", "2.4. Statistical analysis", "3.1. Literature search, study characteristics, and quality assessment", "3.2. Assessment of risk of bias", "3.3. Primary outcomes: progression-free survival rate, median progression-free survival, and overall survival rate", "3.4. Sensitivity analysis", "3.5. Secondary outcomes", "5. Conclusion" ]
[ "We have systematically searched several databases including PubMed, EMBASE, Web of science, EBSCO, and the Cochrane library from inception to March 19, 2020 with the following keywords: “irinotecan” AND “fluorouracil” AND “leucovorin” AND “colorectal cancer” OR “colon cancer” OR “rectal cancer”.\nThe inclusion criteria were as follows: study design was RCT, patients were diagnosed with colorectal cancer, and intervention treatments were irinotecan plus fluorouracil and leucovorin versus only fluorouracil and leucovorin. Patients who previously received pelvic radiotherapy were excluded.", "Some baseline information was extracted, and they included first author, number of patients, age, sex, performance status, primary tumor site (colon/ rectum/both), and detail methods in 2 groups. Data were extracted independently by 2 investigators, and discrepancies were resolved by consensus. The primary outcomes were progression-free survival rate, median progression-free survival, and overall survival rate. Secondary outcomes included objective response, progressive disease, complete response, and grade ≥3 adverse events.", "The risk of bias tool was used to assess the quality of individual studies according to the Cochrane Handbook for Systematic Reviews of Interventions,[16] and the sources of bias were divided into selection bias, performance bias, attrition bias, detection bias, reporting bias, and other potential sources of bias. The overall risk of bias for each study was evaluated and rated: low, unclear, and high.[17] Two investigators independently assessed the quality of included studies, and any discrepancy was solved by consensus.", "We assessed hazard ratio (HR) or risk ratio (RR) with 95% confidence interval (CI) for dichotomous outcomes (progression-free survival rate, overall survival rate, objective response, progressive disease, complete response, and grade ≥3 adverse events) and standard mean difference with 95% CI for continuous outcome (median progression-free survival). Heterogeneity was evaluated by the I2 statistic, and I2 > 50% indicated significant heterogeneity.[18] The random-effects model was used when encountering significant heterogeneity, while fixed-effects model was applied when no significant heterogeneity was found. We searched for potential sources of heterogeneity, and sensitivity analysis was performed to detect the influence of a single study on the overall estimate via omitting 1 study in turn or conducting the subgroup analysis. Owing to the limited number (<10) of included studies, publication bias was not assessed. A P value of <.05 was indicated to be statistically significant. All statistical analyses were performed using Review Manager Version 5.3 (The Cochrane Collaboration, Software Update, Oxford, United Kingdom).", "Figure 1 shows the detail flowchart of the search and selection results. Five hundred ninety-eight potentially relevant articles were initially identified. Two hundred twenty-three duplicates and 366 papers after checking the titles/abstracts were excluded. Four studies were removed because of different combination drugs, and 5 randomized controlled trials (RCTs) were finally included in the meta-analysis.[4,9,13,14,19]\nFlow diagram of study searching and selection process.\nThe baseline characteristics of 5 included RCTs are shown in Table 1. These studies were published between 2000 and 2015, and the total sample size was 4536. All included RCTs reported irinotecan as the adjunctive therapy to fluorouracil and leucovorin, and the methods between irinotecan group and control group were different in each RCT, detailed in Table 1. In the study by Saltz,[14] we just extracted the data of study 2 (Douillard) for this meta-analysis in order to avoid the duplicated data of Saltz.[19]\nCharacteristics of included studies.\nCI = continuous infusion, IV = intravenous.\nFour studies reported progression-free survival rate,[4,9,13,19] 2 studies reported median progression-free survival,[9,13] 4 studies reported overall survival rate and objective response,[9,13,14,19] 2 studies reported progressive disease and complete response,[9,13] and 3 studies reported grade ≥3 adverse events.[9,13,19]", "Risk of bias analysis is presented in Figure 2. These 5 included RCTs generally had high quality although 4 studies had high risk of bias due to their nonblindness.[4,9,14,19]\nRisk of bias assessment. (A) Authors’ judgments about each risk of bias item for each included study. (B) Authors’ judgments about each risk of bias item are presented as percentages across all included studies.", "Compared to control group for colorectal cancer, irinotecan supplementation was associated with substantially improved progression-free survival rate (HR = 0.72; 95% CI = 0.58–0.90; P = .003) with significant heterogeneity among the studies (I2 = 88%, heterogeneity P < .0001; Fig. 3), median progression-free survival (standard mean difference = –0.30; 95% CI = –0.44 to –0.15; P < .0001) with no heterogeneity among the studies (I2 = 0%, heterogeneity P = .79; Fig. 4), and overall survival rate (HR = 0.77; 95% CI = 0.66–0.90; P = .001) with no heterogeneity among the studies (I2 = 0%, heterogeneity P = .98; Fig. 5).\nForest plot for the meta-analysis of progression-free survival rate. CI = confidence interval, IV = intravenous, SE = standard error.\nForest plot for the meta-analysis of median progression-free survival (month). CI = confidence interval, IV = intravenous, SE = standard error.\nForest plot for the meta-analysis of overall survival rate. CI = confidence interval, IV = intravenous, SE = standard error.", "There was significant heterogeneity for progression-free survival rate, but no heterogeneity was observed for median progression-free survival or overall survival rate. As shown in Figure 3, the study conducted by Van Cutsem et al[4] showed the results that were almost completely out of range of the others and probably contributed to the heterogeneity. After excluding that study, the results suggested that irinotecan supplementation could also improve progression-free survival rate for colorectal cancer than control intervention (HR = 0.65; 95% CI = 0.63–0.67; P < .00001). No evidence of heterogeneity was observed among the remaining studies (I2 = 0%).", "In comparison with control group for colorectal cancer, irinotecan supplementation showed the obvious increase in objective response (RR = 0.57; 95% CI = 0.49–0.66; P < .00001; Fig. 6) and the decrease in progressive disease (RR = 2.10; 95% CI = 1.40–3.14; P = .0003; Fig. 7), but had no substantial impact on complete response (RR = 0.89; 95% CI = 0.37–2.13; P = .79; Fig. 8). In addition, the incidence of grade ≥3 adverse events in irinotecan supplementation group was higher than that in control group (RR = 0.67; 95% CI = 0.57–0.79; P < .00001; Fig. 9).\nForest plot for the meta-analysis of objective response. CI = confidence interval, IV = intravenous.\nForest plot for the meta-analysis of progressive disease. CI = confidence interval, IV = intravenous.\nForest plot for the meta-analysis of complete response. CI = confidence interval, IV = intravenous.\nForest plot for the meta-analysis of grade ≥3 adverse events. CI = confidence interval, IV = intravenous.", "Irinotecan supplementation can improve the survival and objective response of colorectal cancer patients receiving fluorouracil and leucovorin, but with the increase in grade ≥3 adverse events." ]
[ null, null, null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Materials and methods", "2.1. Literature search", "2.2. Data extraction and outcome measures", "2.3. Assessment for risk of bias", "2.4. Statistical analysis", "3. Results", "3.1. Literature search, study characteristics, and quality assessment", "3.2. Assessment of risk of bias", "3.3. Primary outcomes: progression-free survival rate, median progression-free survival, and overall survival rate", "3.4. Sensitivity analysis", "3.5. Secondary outcomes", "4. Discussion", "5. Conclusion" ]
[ "Colorectal cancer is regarded as a significant cause of mortality.[1–3] The prognosis of these patients is determined by the stages of colorectal cancer, and 5-year survival rates of stage I, II, and III after surgical intervention are 93.2%, 82.5%, and 59.5%, respectively. Especially, 5-year survival rate of stage IV is only 8.1%.[4] Many patients with resected cancer may suffer from recurrence.[5,6] Fluorouracil and leucovorin have been widely used for colorectal cancer,[7] and are reported to reduce the recurrence rate and improve survival.[8]\nIn order to improve the treatment efficacy, irinotecan or oxaliplatin is used combined with fluorouracil and leucovorin, and these combinations are generally regarded as the effective approach for advanced colorectal cancer.[9,10] In elderly patients, irinotecan or oxaliplatin in combination with fluorouracil is well tolerated and shows similar efficacy between elderly and younger patients.[11] In metastatic colorectal cancer, combining irinotecan with fluorouracil results in a remarkable increase in progression-free survival and overall survival than fluorouracil alone.[12]\nHowever, current evidence is insufficient for routine use of irinotecan supplementation for colorectal cancer, and several studies have reported the conflicting results of irinotecan supplementation for colorectal cancer.[4,9,13,14] This meta-analysis aims to assess the efficacy and safety of irinotecan in combination with fluorouracil and leucovorin for colorectal cancer.", "This meta-analysis was performed based on the Preferred Reporting Items for Systematic Reviews and Meta-analysis statement and Cochrane Handbook for Systematic Reviews of Interventions.[15,16] No ethical approval and patient consent were required because all analyses were based on previously published studies.\n[SUBTITLE] 2.1. Literature search [SUBSECTION] We have systematically searched several databases including PubMed, EMBASE, Web of science, EBSCO, and the Cochrane library from inception to March 19, 2020 with the following keywords: “irinotecan” AND “fluorouracil” AND “leucovorin” AND “colorectal cancer” OR “colon cancer” OR “rectal cancer”.\nThe inclusion criteria were as follows: study design was RCT, patients were diagnosed with colorectal cancer, and intervention treatments were irinotecan plus fluorouracil and leucovorin versus only fluorouracil and leucovorin. Patients who previously received pelvic radiotherapy were excluded.\nWe have systematically searched several databases including PubMed, EMBASE, Web of science, EBSCO, and the Cochrane library from inception to March 19, 2020 with the following keywords: “irinotecan” AND “fluorouracil” AND “leucovorin” AND “colorectal cancer” OR “colon cancer” OR “rectal cancer”.\nThe inclusion criteria were as follows: study design was RCT, patients were diagnosed with colorectal cancer, and intervention treatments were irinotecan plus fluorouracil and leucovorin versus only fluorouracil and leucovorin. Patients who previously received pelvic radiotherapy were excluded.\n[SUBTITLE] 2.2. Data extraction and outcome measures [SUBSECTION] Some baseline information was extracted, and they included first author, number of patients, age, sex, performance status, primary tumor site (colon/ rectum/both), and detail methods in 2 groups. Data were extracted independently by 2 investigators, and discrepancies were resolved by consensus. The primary outcomes were progression-free survival rate, median progression-free survival, and overall survival rate. Secondary outcomes included objective response, progressive disease, complete response, and grade ≥3 adverse events.\nSome baseline information was extracted, and they included first author, number of patients, age, sex, performance status, primary tumor site (colon/ rectum/both), and detail methods in 2 groups. Data were extracted independently by 2 investigators, and discrepancies were resolved by consensus. The primary outcomes were progression-free survival rate, median progression-free survival, and overall survival rate. Secondary outcomes included objective response, progressive disease, complete response, and grade ≥3 adverse events.\n[SUBTITLE] 2.3. Assessment for risk of bias [SUBSECTION] The risk of bias tool was used to assess the quality of individual studies according to the Cochrane Handbook for Systematic Reviews of Interventions,[16] and the sources of bias were divided into selection bias, performance bias, attrition bias, detection bias, reporting bias, and other potential sources of bias. The overall risk of bias for each study was evaluated and rated: low, unclear, and high.[17] Two investigators independently assessed the quality of included studies, and any discrepancy was solved by consensus.\nThe risk of bias tool was used to assess the quality of individual studies according to the Cochrane Handbook for Systematic Reviews of Interventions,[16] and the sources of bias were divided into selection bias, performance bias, attrition bias, detection bias, reporting bias, and other potential sources of bias. The overall risk of bias for each study was evaluated and rated: low, unclear, and high.[17] Two investigators independently assessed the quality of included studies, and any discrepancy was solved by consensus.\n[SUBTITLE] 2.4. Statistical analysis [SUBSECTION] We assessed hazard ratio (HR) or risk ratio (RR) with 95% confidence interval (CI) for dichotomous outcomes (progression-free survival rate, overall survival rate, objective response, progressive disease, complete response, and grade ≥3 adverse events) and standard mean difference with 95% CI for continuous outcome (median progression-free survival). Heterogeneity was evaluated by the I2 statistic, and I2 > 50% indicated significant heterogeneity.[18] The random-effects model was used when encountering significant heterogeneity, while fixed-effects model was applied when no significant heterogeneity was found. We searched for potential sources of heterogeneity, and sensitivity analysis was performed to detect the influence of a single study on the overall estimate via omitting 1 study in turn or conducting the subgroup analysis. Owing to the limited number (<10) of included studies, publication bias was not assessed. A P value of <.05 was indicated to be statistically significant. All statistical analyses were performed using Review Manager Version 5.3 (The Cochrane Collaboration, Software Update, Oxford, United Kingdom).\nWe assessed hazard ratio (HR) or risk ratio (RR) with 95% confidence interval (CI) for dichotomous outcomes (progression-free survival rate, overall survival rate, objective response, progressive disease, complete response, and grade ≥3 adverse events) and standard mean difference with 95% CI for continuous outcome (median progression-free survival). Heterogeneity was evaluated by the I2 statistic, and I2 > 50% indicated significant heterogeneity.[18] The random-effects model was used when encountering significant heterogeneity, while fixed-effects model was applied when no significant heterogeneity was found. We searched for potential sources of heterogeneity, and sensitivity analysis was performed to detect the influence of a single study on the overall estimate via omitting 1 study in turn or conducting the subgroup analysis. Owing to the limited number (<10) of included studies, publication bias was not assessed. A P value of <.05 was indicated to be statistically significant. All statistical analyses were performed using Review Manager Version 5.3 (The Cochrane Collaboration, Software Update, Oxford, United Kingdom).", "We have systematically searched several databases including PubMed, EMBASE, Web of science, EBSCO, and the Cochrane library from inception to March 19, 2020 with the following keywords: “irinotecan” AND “fluorouracil” AND “leucovorin” AND “colorectal cancer” OR “colon cancer” OR “rectal cancer”.\nThe inclusion criteria were as follows: study design was RCT, patients were diagnosed with colorectal cancer, and intervention treatments were irinotecan plus fluorouracil and leucovorin versus only fluorouracil and leucovorin. Patients who previously received pelvic radiotherapy were excluded.", "Some baseline information was extracted, and they included first author, number of patients, age, sex, performance status, primary tumor site (colon/ rectum/both), and detail methods in 2 groups. Data were extracted independently by 2 investigators, and discrepancies were resolved by consensus. The primary outcomes were progression-free survival rate, median progression-free survival, and overall survival rate. Secondary outcomes included objective response, progressive disease, complete response, and grade ≥3 adverse events.", "The risk of bias tool was used to assess the quality of individual studies according to the Cochrane Handbook for Systematic Reviews of Interventions,[16] and the sources of bias were divided into selection bias, performance bias, attrition bias, detection bias, reporting bias, and other potential sources of bias. The overall risk of bias for each study was evaluated and rated: low, unclear, and high.[17] Two investigators independently assessed the quality of included studies, and any discrepancy was solved by consensus.", "We assessed hazard ratio (HR) or risk ratio (RR) with 95% confidence interval (CI) for dichotomous outcomes (progression-free survival rate, overall survival rate, objective response, progressive disease, complete response, and grade ≥3 adverse events) and standard mean difference with 95% CI for continuous outcome (median progression-free survival). Heterogeneity was evaluated by the I2 statistic, and I2 > 50% indicated significant heterogeneity.[18] The random-effects model was used when encountering significant heterogeneity, while fixed-effects model was applied when no significant heterogeneity was found. We searched for potential sources of heterogeneity, and sensitivity analysis was performed to detect the influence of a single study on the overall estimate via omitting 1 study in turn or conducting the subgroup analysis. Owing to the limited number (<10) of included studies, publication bias was not assessed. A P value of <.05 was indicated to be statistically significant. All statistical analyses were performed using Review Manager Version 5.3 (The Cochrane Collaboration, Software Update, Oxford, United Kingdom).", "[SUBTITLE] 3.1. Literature search, study characteristics, and quality assessment [SUBSECTION] Figure 1 shows the detail flowchart of the search and selection results. Five hundred ninety-eight potentially relevant articles were initially identified. Two hundred twenty-three duplicates and 366 papers after checking the titles/abstracts were excluded. Four studies were removed because of different combination drugs, and 5 randomized controlled trials (RCTs) were finally included in the meta-analysis.[4,9,13,14,19]\nFlow diagram of study searching and selection process.\nThe baseline characteristics of 5 included RCTs are shown in Table 1. These studies were published between 2000 and 2015, and the total sample size was 4536. All included RCTs reported irinotecan as the adjunctive therapy to fluorouracil and leucovorin, and the methods between irinotecan group and control group were different in each RCT, detailed in Table 1. In the study by Saltz,[14] we just extracted the data of study 2 (Douillard) for this meta-analysis in order to avoid the duplicated data of Saltz.[19]\nCharacteristics of included studies.\nCI = continuous infusion, IV = intravenous.\nFour studies reported progression-free survival rate,[4,9,13,19] 2 studies reported median progression-free survival,[9,13] 4 studies reported overall survival rate and objective response,[9,13,14,19] 2 studies reported progressive disease and complete response,[9,13] and 3 studies reported grade ≥3 adverse events.[9,13,19]\nFigure 1 shows the detail flowchart of the search and selection results. Five hundred ninety-eight potentially relevant articles were initially identified. Two hundred twenty-three duplicates and 366 papers after checking the titles/abstracts were excluded. Four studies were removed because of different combination drugs, and 5 randomized controlled trials (RCTs) were finally included in the meta-analysis.[4,9,13,14,19]\nFlow diagram of study searching and selection process.\nThe baseline characteristics of 5 included RCTs are shown in Table 1. These studies were published between 2000 and 2015, and the total sample size was 4536. All included RCTs reported irinotecan as the adjunctive therapy to fluorouracil and leucovorin, and the methods between irinotecan group and control group were different in each RCT, detailed in Table 1. In the study by Saltz,[14] we just extracted the data of study 2 (Douillard) for this meta-analysis in order to avoid the duplicated data of Saltz.[19]\nCharacteristics of included studies.\nCI = continuous infusion, IV = intravenous.\nFour studies reported progression-free survival rate,[4,9,13,19] 2 studies reported median progression-free survival,[9,13] 4 studies reported overall survival rate and objective response,[9,13,14,19] 2 studies reported progressive disease and complete response,[9,13] and 3 studies reported grade ≥3 adverse events.[9,13,19]\n[SUBTITLE] 3.2. Assessment of risk of bias [SUBSECTION] Risk of bias analysis is presented in Figure 2. These 5 included RCTs generally had high quality although 4 studies had high risk of bias due to their nonblindness.[4,9,14,19]\nRisk of bias assessment. (A) Authors’ judgments about each risk of bias item for each included study. (B) Authors’ judgments about each risk of bias item are presented as percentages across all included studies.\nRisk of bias analysis is presented in Figure 2. These 5 included RCTs generally had high quality although 4 studies had high risk of bias due to their nonblindness.[4,9,14,19]\nRisk of bias assessment. (A) Authors’ judgments about each risk of bias item for each included study. (B) Authors’ judgments about each risk of bias item are presented as percentages across all included studies.\n[SUBTITLE] 3.3. Primary outcomes: progression-free survival rate, median progression-free survival, and overall survival rate [SUBSECTION] Compared to control group for colorectal cancer, irinotecan supplementation was associated with substantially improved progression-free survival rate (HR = 0.72; 95% CI = 0.58–0.90; P = .003) with significant heterogeneity among the studies (I2 = 88%, heterogeneity P < .0001; Fig. 3), median progression-free survival (standard mean difference = –0.30; 95% CI = –0.44 to –0.15; P < .0001) with no heterogeneity among the studies (I2 = 0%, heterogeneity P = .79; Fig. 4), and overall survival rate (HR = 0.77; 95% CI = 0.66–0.90; P = .001) with no heterogeneity among the studies (I2 = 0%, heterogeneity P = .98; Fig. 5).\nForest plot for the meta-analysis of progression-free survival rate. CI = confidence interval, IV = intravenous, SE = standard error.\nForest plot for the meta-analysis of median progression-free survival (month). CI = confidence interval, IV = intravenous, SE = standard error.\nForest plot for the meta-analysis of overall survival rate. CI = confidence interval, IV = intravenous, SE = standard error.\nCompared to control group for colorectal cancer, irinotecan supplementation was associated with substantially improved progression-free survival rate (HR = 0.72; 95% CI = 0.58–0.90; P = .003) with significant heterogeneity among the studies (I2 = 88%, heterogeneity P < .0001; Fig. 3), median progression-free survival (standard mean difference = –0.30; 95% CI = –0.44 to –0.15; P < .0001) with no heterogeneity among the studies (I2 = 0%, heterogeneity P = .79; Fig. 4), and overall survival rate (HR = 0.77; 95% CI = 0.66–0.90; P = .001) with no heterogeneity among the studies (I2 = 0%, heterogeneity P = .98; Fig. 5).\nForest plot for the meta-analysis of progression-free survival rate. CI = confidence interval, IV = intravenous, SE = standard error.\nForest plot for the meta-analysis of median progression-free survival (month). CI = confidence interval, IV = intravenous, SE = standard error.\nForest plot for the meta-analysis of overall survival rate. CI = confidence interval, IV = intravenous, SE = standard error.\n[SUBTITLE] 3.4. Sensitivity analysis [SUBSECTION] There was significant heterogeneity for progression-free survival rate, but no heterogeneity was observed for median progression-free survival or overall survival rate. As shown in Figure 3, the study conducted by Van Cutsem et al[4] showed the results that were almost completely out of range of the others and probably contributed to the heterogeneity. After excluding that study, the results suggested that irinotecan supplementation could also improve progression-free survival rate for colorectal cancer than control intervention (HR = 0.65; 95% CI = 0.63–0.67; P < .00001). No evidence of heterogeneity was observed among the remaining studies (I2 = 0%).\nThere was significant heterogeneity for progression-free survival rate, but no heterogeneity was observed for median progression-free survival or overall survival rate. As shown in Figure 3, the study conducted by Van Cutsem et al[4] showed the results that were almost completely out of range of the others and probably contributed to the heterogeneity. After excluding that study, the results suggested that irinotecan supplementation could also improve progression-free survival rate for colorectal cancer than control intervention (HR = 0.65; 95% CI = 0.63–0.67; P < .00001). No evidence of heterogeneity was observed among the remaining studies (I2 = 0%).\n[SUBTITLE] 3.5. Secondary outcomes [SUBSECTION] In comparison with control group for colorectal cancer, irinotecan supplementation showed the obvious increase in objective response (RR = 0.57; 95% CI = 0.49–0.66; P < .00001; Fig. 6) and the decrease in progressive disease (RR = 2.10; 95% CI = 1.40–3.14; P = .0003; Fig. 7), but had no substantial impact on complete response (RR = 0.89; 95% CI = 0.37–2.13; P = .79; Fig. 8). In addition, the incidence of grade ≥3 adverse events in irinotecan supplementation group was higher than that in control group (RR = 0.67; 95% CI = 0.57–0.79; P < .00001; Fig. 9).\nForest plot for the meta-analysis of objective response. CI = confidence interval, IV = intravenous.\nForest plot for the meta-analysis of progressive disease. CI = confidence interval, IV = intravenous.\nForest plot for the meta-analysis of complete response. CI = confidence interval, IV = intravenous.\nForest plot for the meta-analysis of grade ≥3 adverse events. CI = confidence interval, IV = intravenous.\nIn comparison with control group for colorectal cancer, irinotecan supplementation showed the obvious increase in objective response (RR = 0.57; 95% CI = 0.49–0.66; P < .00001; Fig. 6) and the decrease in progressive disease (RR = 2.10; 95% CI = 1.40–3.14; P = .0003; Fig. 7), but had no substantial impact on complete response (RR = 0.89; 95% CI = 0.37–2.13; P = .79; Fig. 8). In addition, the incidence of grade ≥3 adverse events in irinotecan supplementation group was higher than that in control group (RR = 0.67; 95% CI = 0.57–0.79; P < .00001; Fig. 9).\nForest plot for the meta-analysis of objective response. CI = confidence interval, IV = intravenous.\nForest plot for the meta-analysis of progressive disease. CI = confidence interval, IV = intravenous.\nForest plot for the meta-analysis of complete response. CI = confidence interval, IV = intravenous.\nForest plot for the meta-analysis of grade ≥3 adverse events. CI = confidence interval, IV = intravenous.", "Figure 1 shows the detail flowchart of the search and selection results. Five hundred ninety-eight potentially relevant articles were initially identified. Two hundred twenty-three duplicates and 366 papers after checking the titles/abstracts were excluded. Four studies were removed because of different combination drugs, and 5 randomized controlled trials (RCTs) were finally included in the meta-analysis.[4,9,13,14,19]\nFlow diagram of study searching and selection process.\nThe baseline characteristics of 5 included RCTs are shown in Table 1. These studies were published between 2000 and 2015, and the total sample size was 4536. All included RCTs reported irinotecan as the adjunctive therapy to fluorouracil and leucovorin, and the methods between irinotecan group and control group were different in each RCT, detailed in Table 1. In the study by Saltz,[14] we just extracted the data of study 2 (Douillard) for this meta-analysis in order to avoid the duplicated data of Saltz.[19]\nCharacteristics of included studies.\nCI = continuous infusion, IV = intravenous.\nFour studies reported progression-free survival rate,[4,9,13,19] 2 studies reported median progression-free survival,[9,13] 4 studies reported overall survival rate and objective response,[9,13,14,19] 2 studies reported progressive disease and complete response,[9,13] and 3 studies reported grade ≥3 adverse events.[9,13,19]", "Risk of bias analysis is presented in Figure 2. These 5 included RCTs generally had high quality although 4 studies had high risk of bias due to their nonblindness.[4,9,14,19]\nRisk of bias assessment. (A) Authors’ judgments about each risk of bias item for each included study. (B) Authors’ judgments about each risk of bias item are presented as percentages across all included studies.", "Compared to control group for colorectal cancer, irinotecan supplementation was associated with substantially improved progression-free survival rate (HR = 0.72; 95% CI = 0.58–0.90; P = .003) with significant heterogeneity among the studies (I2 = 88%, heterogeneity P < .0001; Fig. 3), median progression-free survival (standard mean difference = –0.30; 95% CI = –0.44 to –0.15; P < .0001) with no heterogeneity among the studies (I2 = 0%, heterogeneity P = .79; Fig. 4), and overall survival rate (HR = 0.77; 95% CI = 0.66–0.90; P = .001) with no heterogeneity among the studies (I2 = 0%, heterogeneity P = .98; Fig. 5).\nForest plot for the meta-analysis of progression-free survival rate. CI = confidence interval, IV = intravenous, SE = standard error.\nForest plot for the meta-analysis of median progression-free survival (month). CI = confidence interval, IV = intravenous, SE = standard error.\nForest plot for the meta-analysis of overall survival rate. CI = confidence interval, IV = intravenous, SE = standard error.", "There was significant heterogeneity for progression-free survival rate, but no heterogeneity was observed for median progression-free survival or overall survival rate. As shown in Figure 3, the study conducted by Van Cutsem et al[4] showed the results that were almost completely out of range of the others and probably contributed to the heterogeneity. After excluding that study, the results suggested that irinotecan supplementation could also improve progression-free survival rate for colorectal cancer than control intervention (HR = 0.65; 95% CI = 0.63–0.67; P < .00001). No evidence of heterogeneity was observed among the remaining studies (I2 = 0%).", "In comparison with control group for colorectal cancer, irinotecan supplementation showed the obvious increase in objective response (RR = 0.57; 95% CI = 0.49–0.66; P < .00001; Fig. 6) and the decrease in progressive disease (RR = 2.10; 95% CI = 1.40–3.14; P = .0003; Fig. 7), but had no substantial impact on complete response (RR = 0.89; 95% CI = 0.37–2.13; P = .79; Fig. 8). In addition, the incidence of grade ≥3 adverse events in irinotecan supplementation group was higher than that in control group (RR = 0.67; 95% CI = 0.57–0.79; P < .00001; Fig. 9).\nForest plot for the meta-analysis of objective response. CI = confidence interval, IV = intravenous.\nForest plot for the meta-analysis of progressive disease. CI = confidence interval, IV = intravenous.\nForest plot for the meta-analysis of complete response. CI = confidence interval, IV = intravenous.\nForest plot for the meta-analysis of grade ≥3 adverse events. CI = confidence interval, IV = intravenous.", "Irinotecan was documented to be an effective topoisomerase I inhibitor with antitumor properties and its combination with fluorouracil/leucovorin was found to improve the outcomes of patients with metastatic colorectal cancer.[20,21] In contrast, in another trial involving patients with stage III colon cancer, irinotecan plus fluorouracil/leucovorin did not improve overall survival compared with fluorouracil/leucovorin alone.[4] Considering these inconsistence, our meta-analysis was performed and confirmed that irinotecan in combination with fluorouracil and leucovorin could substantially improve progression-free survival rate, median progression-free survival, overall survival rate, and objective response and reduce the incidence of progressive disease for colorectal cancer compared to only fluorouracil and leucovorin, but revealed no obvious influence on complete response.\nRegarding the sensitivity analysis, significant heterogeneity remains for progression-free survival rate. Three studies reported metastatic colorectal cancer,[9,13,19] while the remaining study conducted by Van Cutsem et al[4] reported colon cancer with stage III. After excluding that study, there was no heterogeneity found. Irinotecan supplementation can also improve progression-free survival rate for colorectal cancer (P < .00001) than control intervention. These indicated that irinotecan plus fluorouracil and leucovorin may have better efficacy to improve progression-free survival rate in stage IV colorectal cancer than that in stage III colorectal cancer.\nIn addition, patient populations with different age ranges may have some impact on the efficacy of irinotecan supplementation. For instance, adding irinotecan to fluorouracil for metastatic colorectal cancer showed no significant impact on progression-free survival in patients aged ≥75.[13] In contrast, a post hoc analysis demonstrated that irinotecan plus fluorouracil can improve progression-free survival than fluorouracil alone in patients only aged 70 to 75 years, but this efficacy was not observed in patients aged >75 years.[13]\nFluorouracil/leucovorin in combination with irinotecan was found to have the advantage of reduced toxicity compared with fluorouracil/leucovorin.[4,13] However, irinotecan supplementation was found to increase the incidence of grade ≥3 adverse events than control group in colorectal cancer based on the results of this meta-analysis. These side effects mainly included diarrhea and neutropenia and were generally manageable and acceptable.[4,9,11,19] Several limitations exist in this meta-analysis. First, our analysis was based on only 5 RCTs, and more RCTs with large sample size should be conducted to explore this issue. Next, there is significant heterogeneity, and these sources of heterogeneity should be assessed by subgroup analysis (different stages of colorectal cancer, patients with various age range, and methods of drug combination). However, it is not possible due to the small number of included studies. Finally, genetic variants such as the expression of metadherin and carcinoembryonic antigen may affect therapeutic response and prognosis of colorectal cancer and produce some bias.[22]", "Irinotecan supplementation can improve the survival and objective response of colorectal cancer patients receiving fluorouracil and leucovorin, but with the increase in grade ≥3 adverse events." ]
[ "intro", "methods", null, null, null, null, "results", null, null, null, null, null, "discussion", null ]
[ "colorectal cancer", "fluorouracil", "irinotecan", "leucovorin", "randomized controlled trials" ]
Comparison of regional cerebral oxygen saturation during one-lung ventilation under desflurane or propofol anesthesia: A randomized trial.
36254073
During one-lung ventilation (OLV), deterioration of pulmonary oxygenation reduces arterial oxygen saturation and cerebral oxygen saturation (rSO2). However, oxidative stress during OLV causes lung injury, so the fraction of inspiratory oxygen (FiO2) should be kept as low as possible. We investigated the changes in rSO2 under propofol or desflurane anesthesia while percutaneous oxygen saturation (SpO2) was kept as low as possible during OLV.
BACKGROUND
Thirty-six patients scheduled for thoracic surgery under OLV in the lateral decubitus position were randomly assigned to propofol (n = 19) or desflurane (n = 17) anesthesia. FiO2 was set to 0.4 at the start of surgery under two-lung ventilation (measurement point: T3) and then adjusted to maintain an SpO2 of 92% to 94% after the initiation of OLV. The primary outcome was the difference in the absolute value of the decrease in rSO2 from T3 to 30 minutes after the initiation of OLV (T5), which was analyzed by an analysis of covariance adjusted for the rSO2 value at T3.
METHODS
The mean rSO2 values were 61.5% ± 5.1% at T3 and 57.1% ± 5.3% at T5 in the propofol group and 62.2% ± 6.0% at T3 and 58.6% ± 5.3% at T5 in the desflurane group. The difference in the absolute value of decrease between groups (propofol group - desflurane group) was 0.95 (95% confidence interval, [-0.32, 2.2]; P = .152).
RESULTS
Both propofol and desflurane anesthesia maintain comparable cerebral oxygenation and can be used safely, even when the SpO2 is kept as low as possible during OLV.
CONCLUSIONS
[ "Anesthesia", "Desflurane", "Humans", "One-Lung Ventilation", "Oxygen", "Oxygen Saturation", "Propofol" ]
9575834
1. Introduction
One-lung ventilation (OLV) is an indispensable anesthetic procedure for thoracic surgery. During OLV, the collapsed and unventilated lung still has perfusion that can cause intrapulmonary shunting, leading to impaired oxygenation and hypoxemia. Although a high fraction of inspiratory oxygen (FiO2) tends to be set to avoid hypoxemia during OLV, high FiO2 levels also increase the risk of postoperative respiratory complications and 30-day mortality.[1,2] Furthermore, oxidative stress during OLV causes lung injury, which is exacerbated by a higher FiO2.[3] The FiO2 should thus be kept as low as possible while maintaining the percutaneous oxygen saturation (SpO2) level within the appropriate range.[4] There is some concern about an insufficient oxygen supply to vital organs, especially the central nervous system, during OLV. Cerebral oxygenation is evaluated by the regional cerebral oxygen saturation (rSO2) measurement. It has been reported that the rSO2 declines during OLV despite the SpO2 being above 90%, which may induce postoperative cognitive dysfunction.[5–9] Sungur et al[8] reported that preventing cerebral desaturation might reduce postoperative cognitive dysfunction. Recently, for adult patients, desflurane has been more commonly used as an anesthetic agent than sevoflurane because of its lower in vivo metabolic rate and faster emergence from general anesthesia. Desflurane has similar oxygenation, shunt fraction, and hemodynamics to sevoflurane during OLV.[10] Although arterial oxygenation during OLV has been reported to be reduced with desflurane-remifentanil anesthesia in comparison to propofol-remifentanil anesthesia, desflurane is nevertheless still frequently used as an anesthetic agent.[11] In general, both propofol and desflurane reduce the cerebral metabolic rate for oxygen (CMRO2), although their effects on the cerebral blood flow (CBF) are not the same. Propofol has a cerebrovascular contractile effect and reduces both the CMRO2 and CBF.[12] Of note, the reduction in CBF outweighs the reduction in CMRO2 due to propofol, resulting in an overall reduction in the CBF/CMRO2 ratio.[13–15] Conversely, volatile anesthetic agents, such as desflurane, have a dose-dependent cerebrovascular dilator effect, which causes an increased CBF at concentrations >1 minimal alveolar concentration (MAC).[16–18] As such, the cerebral oxygen balance during OLV can be impaired under propofol anesthesia in comparison to volatile anesthetics. A previous study further showed that cerebral oxygen desaturation evaluated based on the jugular bulb venous oxygen saturation (SjO2) occurred more frequently under propofol anesthesia than under sevoflurane anesthesia during OLV, with an FiO2 of 0.5 and SpO2 of approximately 95%.[19] To our knowledge, no previous study has compared cerebral oxygenation during OLV under desflurane and propofol anesthesia. We hypothesize that cerebral oxygenation is more markedly impaired during OLV under propofol anesthesia than under desflurane anesthesia. The present study therefore compared the effects of propofol and desflurane on cerebral oxygenation measured by rSO2 during OLV to avoid a high FiO2 level in patients undergoing thoracic surgery.
2.3. Procedures
The 36 total patients were randomly allocated to the desflurane or propofol group. After arriving in the operating room, a cerebral oximeter probe and electroencephalograph sensor were placed on the forehead of the subject and connected to a regional cerebral oximetry system (O3 regional oximetry; Masimo, Irvine, CA) and electroencephalograph monitor (SedLine sedation monitor; Masimo), respectively. The O3 regional oximetry recorded bilateral rSO2 by near-infrared spectroscopy, and the SedLine sedation monitor recorded the Patient State index. A 22-gauge catheter was placed into a radial artery and connected to a pressure transducer to monitor the arterial blood pressure, stroke volume variation and continuous cardiac index using the Vigileo system (Edwards Lifescience, Irvine, CA) with an arterial pressure waveform analysis sensor (FloTrac Sensor; Edwards Lifesciences). [SUBTITLE] 2.3.1. Anesthetic management. [SUBSECTION] Before the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids. Before the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids. [SUBTITLE] 2.3.2. Respiratory settings. [SUBSECTION] The subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study. The subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study. [SUBTITLE] 2.3.3. Measurement values and time points. [SUBSECTION] The measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined. Measurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation. All complications during hospitalization were recorded. The measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined. Measurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation. All complications during hospitalization were recorded.
3. Results
Between March and October, a total of 99 participants were screened for eligibility. Four patients scheduled for surgery at the same time on the same day were excluded. Seven patients were excluded because of a past medical history of postoperative nausea and vomiting, and 9 patients were excluded for other reasons (chronic kidney disease in 3, atrial fibrillation in 2, thoracic empyema in 2, sequelae after resection of a metastatic brain tumor in 1, and another clinical trial in progress in 1). Of the remaining 39 participants, 20 patients were assigned to the propofol group and 19 to the desflurane group (CONSORT Flow Diagram, Figure 2). One patient in the propofol group and 2 in the desflurane group were excluded after randomization (canceled surgery, n = 1; diagnosis of interstitial pneumonia after the induction of anesthesia, n = 1; and severe desaturation event occurring immediately after the induction of anesthesia, n = 1). Thus, a total of 36 patients (propofol group, n = 19; desflurane group, n = 17) thus completed the study protocol, and their datasets were available for the analysis. Figure 2. CONSORT diagram of the study flow. ASA-PSC = American Society of Anesthesiologists physical status classification. The baseline characteristics and intraoperative variables of these 36 patients are shown in Table 1. Figure 3A shows the mean rSO2 values at each measurement point: 61.5% ± 5.1% in the propofol group and 62.2% ± 6.0% in the desflurane group at T3, and 57.1% ± 5.3% in the propofol group and 58.6% ± 5.3% in the desflurane group at T5. There was no significant difference between the groups in the absolute value of the decrease in rSO2 adjusted by the rSO2 value at T3 (Table 2). Characteristics of the patients. Statistics presented: median (interquartile range) for continuous variables; n (%) for categorical variables. %FEV1.0 = percent predicted forced expiratory volume in 1 second, ASA-PSC = American Society of Anesthesiologists physical status classification, FVC = forced vital capacity, FEV1.0 = forced expiratory volume in one second, OLV = one-lung ventilation, SpO2 = percutaneous oxygen saturation. Primary and secondary outcomes: ANCOVA adjusted for the value of rSO2 at the T3. is the coefficient obtained from ANCOVA. If β of group is >0, this indicate that the absolute value of the decrease in rSO2 from T3 to T5 is greater in the Propofol group than Desflurane group, with adjustment for rSO2 at T3. CI = confidence interval, rSO2 = regional cerebral oxygen saturation, T3 = just before the start of surgery, T5 = 30 minutes after initiation of one-lung ventilation. Changes in (A) mean rSO2, (B) dependent lung side rSO2, and (C) dominant side rSO2. Each value represents the mean ± standard deviation. The solid circle and solid line represent the propofol group, and the open circle and interrupted line represent the desflurane group. See Figure 1. for the definitions of the measurement time points. dependent lung side rSO2 = rSO2 values on the dependent lung side, dominant side rSO2 = rSO2 values on the dominant side, mean rSO2 = mean of the left and right rSO2, rSO2 = regional cerebral oxygen saturation. After adjustment by the rSO2 value at T3, the absolute value of the decrease in rSO2 was significantly greater in the propofol group than in the desflurane group on both the dependent lung and the dominant sides (Table 2). Figure 3B and C shows the rSO2 values on the dependent lung and dominant lung sides, respectively, in both groups. The PaO2 value at T3 was significantly higher in the desflurane group. There were no significant differences in the MAP, continuous cardiac index, SpO2, PaCO2, any other hemodynamic parameters or the arterial blood gas data between the groups at T3 and T5 (Table 3). Furthermore, there were no significant differences in the incidence of cerebral desaturation events between the groups (propofol group, n = 5; desflurane group, n = 2; P = .41). One patient in each group showed a decrease in SpO2 immediately after the start of one-lung ventilation due to malposition of the double lumen tube, and the FiO2 was temporarily increased to 1.0 to adjust the tube position using a fiberscope. No cases required the adjustment of FiO2 in response to decreased rSO2. Complications occurred in 2 subjects in each group: paroxysmal atrial fibrillation and postoperative respiratory failure requiring admission to the intensive care unit in one patient each in the propofol group and prolonged pulmonary leakage and continuous atrial fibrillation requiring electrical ablation in the desflurane group. Intraoperative anesthetic variables and arterial blood gas data. Satistics presented: mean (SD). There were not significant different in any values between the 2 groups at T3 and T5, respectively. BT = body temperature, Ce = effect site concentration, FiO2 = fraction of inspiratory oxygen, HR = heart rate, MAP = mean arterial blood pressure, PaCO2 = arterial partial pressure of carbon dioxide, PaO2 = arterial partial pressure of oxygen, SpO2 = percutaneous oxygen saturation, T3 = just before the start of surgery, T5 = 30 minutes after initiation of OLV. All patients were discharged with no central neurological complications.
null
null
[ "2. Materials and Methods", "2.1. Study design and participants", "2.2. Randomization and masking", "2.3.1. Anesthetic management.", "2.3.2. Respiratory settings.", "2.3.3. Measurement values and time points.", "2.4. Outcomes", "2.5. Statistical analyses", "Author contributions" ]
[ "[SUBTITLE] 2.1. Study design and participants [SUBSECTION] This single-center, single-blinded, randomized clinical trial with parallel groups took place in Gifu University Hospital. The study protocol was approved by the Institutional Review Board of The Gifu University Hospital (decision number: 2019-274, approval date: March 16, 2020) and was registered at University hospital Medical Information Network Center (identifier: UMIN000039403, resister date: March 4, 2020).\nWe evaluated patients of ≥20 years of age who were scheduled for elective thoracic surgery requiring OLV in the lateral decubitus position at Gifu University Hospital from March to October 2020. Cases involving esophagectomy, robot-assisted thoracic surgery, lung resection for pneumothorax, surgery with carbon dioxide gas insufflation for pneumothorax and surgery with an expected duration of OLV < 30 minutes were not included in the analysis.\nParticipants who did not meet the exclusion criteria were asked to give their written informed consent before enrollment. The exclusion criteria were a medical history of lobectomy or segmentectomy, cerebrovascular disease, coronary artery disease, heart failure, or interstitial pneumonia (including suspicious cases); an American Society of Anesthesiologists physical status classification of ≥Ⅲ for any other reason; contraindication of study drugs; and any other reason deemed to render the participant inappropriate for intervention.\nThis study was conducted in accordance with the Consolidated Standards of Reporting Trials (CONSORT) 2010 statement.\nThis single-center, single-blinded, randomized clinical trial with parallel groups took place in Gifu University Hospital. The study protocol was approved by the Institutional Review Board of The Gifu University Hospital (decision number: 2019-274, approval date: March 16, 2020) and was registered at University hospital Medical Information Network Center (identifier: UMIN000039403, resister date: March 4, 2020).\nWe evaluated patients of ≥20 years of age who were scheduled for elective thoracic surgery requiring OLV in the lateral decubitus position at Gifu University Hospital from March to October 2020. Cases involving esophagectomy, robot-assisted thoracic surgery, lung resection for pneumothorax, surgery with carbon dioxide gas insufflation for pneumothorax and surgery with an expected duration of OLV < 30 minutes were not included in the analysis.\nParticipants who did not meet the exclusion criteria were asked to give their written informed consent before enrollment. The exclusion criteria were a medical history of lobectomy or segmentectomy, cerebrovascular disease, coronary artery disease, heart failure, or interstitial pneumonia (including suspicious cases); an American Society of Anesthesiologists physical status classification of ≥Ⅲ for any other reason; contraindication of study drugs; and any other reason deemed to render the participant inappropriate for intervention.\nThis study was conducted in accordance with the Consolidated Standards of Reporting Trials (CONSORT) 2010 statement.\n[SUBTITLE] 2.2. Randomization and masking [SUBSECTION] The data manager used the R software program, version 3.6.3 (www.r-project.org), to create a randomization sequence with 1:1 allocation, a mixed block size of 2 or 4, and no stratification. Researchers were not informed of the block size throughout the trial. The researchers assigned patients to either the propofol or desflurane group using sealed envelopes prepared according to a pregenerated allocation table. Sequence generation and randomization envelope preparation were performed by the data manager, independent of the researchers who had no further role in the trial. The patients were not informed of the treatment allocation.\nThe data manager used the R software program, version 3.6.3 (www.r-project.org), to create a randomization sequence with 1:1 allocation, a mixed block size of 2 or 4, and no stratification. Researchers were not informed of the block size throughout the trial. The researchers assigned patients to either the propofol or desflurane group using sealed envelopes prepared according to a pregenerated allocation table. Sequence generation and randomization envelope preparation were performed by the data manager, independent of the researchers who had no further role in the trial. The patients were not informed of the treatment allocation.\n[SUBTITLE] 2.3. Procedures [SUBSECTION] The 36 total patients were randomly allocated to the desflurane or propofol group. After arriving in the operating room, a cerebral oximeter probe and electroencephalograph sensor were placed on the forehead of the subject and connected to a regional cerebral oximetry system (O3 regional oximetry; Masimo, Irvine, CA) and electroencephalograph monitor (SedLine sedation monitor; Masimo), respectively. The O3 regional oximetry recorded bilateral rSO2 by near-infrared spectroscopy, and the SedLine sedation monitor recorded the Patient State index. A 22-gauge catheter was placed into a radial artery and connected to a pressure transducer to monitor the arterial blood pressure, stroke volume variation and continuous cardiac index using the Vigileo system (Edwards Lifescience, Irvine, CA) with an arterial pressure waveform analysis sensor (FloTrac Sensor; Edwards Lifesciences).\n[SUBTITLE] 2.3.1. Anesthetic management. [SUBSECTION] Before the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids.\nBefore the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids.\n[SUBTITLE] 2.3.2. Respiratory settings. [SUBSECTION] The subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study.\nThe subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study.\n[SUBTITLE] 2.3.3. Measurement values and time points. [SUBSECTION] The measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined.\nMeasurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation.\nAll complications during hospitalization were recorded.\nThe measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined.\nMeasurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation.\nAll complications during hospitalization were recorded.\nThe 36 total patients were randomly allocated to the desflurane or propofol group. After arriving in the operating room, a cerebral oximeter probe and electroencephalograph sensor were placed on the forehead of the subject and connected to a regional cerebral oximetry system (O3 regional oximetry; Masimo, Irvine, CA) and electroencephalograph monitor (SedLine sedation monitor; Masimo), respectively. The O3 regional oximetry recorded bilateral rSO2 by near-infrared spectroscopy, and the SedLine sedation monitor recorded the Patient State index. A 22-gauge catheter was placed into a radial artery and connected to a pressure transducer to monitor the arterial blood pressure, stroke volume variation and continuous cardiac index using the Vigileo system (Edwards Lifescience, Irvine, CA) with an arterial pressure waveform analysis sensor (FloTrac Sensor; Edwards Lifesciences).\n[SUBTITLE] 2.3.1. Anesthetic management. [SUBSECTION] Before the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids.\nBefore the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids.\n[SUBTITLE] 2.3.2. Respiratory settings. [SUBSECTION] The subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study.\nThe subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study.\n[SUBTITLE] 2.3.3. Measurement values and time points. [SUBSECTION] The measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined.\nMeasurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation.\nAll complications during hospitalization were recorded.\nThe measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined.\nMeasurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation.\nAll complications during hospitalization were recorded.\n[SUBTITLE] 2.4. Outcomes [SUBSECTION] The primary outcome was the absolute value of the decrease in the mean left and right rSO2 values from T3 to T5. The secondary outcomes were the absolute value of the decrease in the rSO2 value of the dependent and nondependent lung side from T3 to T5, the absolute value of the decrease in the rSO2 value on the dominant and nondominant side of the subject from T3 to T5, the number of cerebral desaturations and complications occurring until discharge from the hospital.\nThe primary outcome was the absolute value of the decrease in the mean left and right rSO2 values from T3 to T5. The secondary outcomes were the absolute value of the decrease in the rSO2 value of the dependent and nondependent lung side from T3 to T5, the absolute value of the decrease in the rSO2 value on the dominant and nondominant side of the subject from T3 to T5, the number of cerebral desaturations and complications occurring until discharge from the hospital.\n[SUBTITLE] 2.5. Statistical analyses [SUBSECTION] Prior to this study, a pilot study (decision number: 2018-210) was conducted to determine the sample size and collect results to select appropriate measurement values. In the pilot study, the absolute value of the decrease in the mean left and right rSO2 values from T3 to T5 was 14.0% in the propofol group and 9.8% in the desflurane group. Because the standard deviations (SDs) were 4.14 and 3.15, respectively, the SD was conservatively set at 4.14 in both groups. To detect a difference of 4.2% in the absolute value of the decrease in rSO2 between the propofol and desflurane groups by a t test with a detection power of 80% and at a 0.05 two-sided significance level, 16 cases per group (32 cases in total) were deemed necessary. We estimated that 10% of cases for per group would have unusable data, so we determined that 36 participants in whom measurements could be performed at least once during OLV were required. The full analysis set included all patients who received the protocol treatment at least once, except for those cases that were never evaluated for efficacy.\nBaseline characteristics and aggregated data are presented as the median (interquartile range: IQR) or mean ± SD for continuous variables and numbers (%) for categorical variables. The patient hemodynamic parameters and results of arterial blood gas analyses at T3 and T5 were compared between the 2 groups using Student t test. The primary outcome was analyzed according to the intent-to-treat principle with an analysis of covariance (ANCOVA) adjusted for the mean left and right rSO2 value at T3 (baseline). The secondary outcomes were analyzed with adjustment for the baseline value using a similar model to that used for the primary outcome. All outcomes at each measurement point were described using the mean, median, SD and 25th and 75th percentiles, and the 95% confidence interval was calculated. The Fisher exact test was used to compare the incidence of cerebral desaturation. Imputation was not used for missing data for any outcomes, as no data were missing. Subgroup and post hoc analyses were not performed.\nAll statistical analyses were performed with the R software program, version 4.0.3 (www.r-project.org). Two-sided P value of <.05 were considered statistically significant. There was no adjustment for multiple comparisons, as the analyses of secondary endpoints were interpreted as exploratory.\nPrior to this study, a pilot study (decision number: 2018-210) was conducted to determine the sample size and collect results to select appropriate measurement values. In the pilot study, the absolute value of the decrease in the mean left and right rSO2 values from T3 to T5 was 14.0% in the propofol group and 9.8% in the desflurane group. Because the standard deviations (SDs) were 4.14 and 3.15, respectively, the SD was conservatively set at 4.14 in both groups. To detect a difference of 4.2% in the absolute value of the decrease in rSO2 between the propofol and desflurane groups by a t test with a detection power of 80% and at a 0.05 two-sided significance level, 16 cases per group (32 cases in total) were deemed necessary. We estimated that 10% of cases for per group would have unusable data, so we determined that 36 participants in whom measurements could be performed at least once during OLV were required. The full analysis set included all patients who received the protocol treatment at least once, except for those cases that were never evaluated for efficacy.\nBaseline characteristics and aggregated data are presented as the median (interquartile range: IQR) or mean ± SD for continuous variables and numbers (%) for categorical variables. The patient hemodynamic parameters and results of arterial blood gas analyses at T3 and T5 were compared between the 2 groups using Student t test. The primary outcome was analyzed according to the intent-to-treat principle with an analysis of covariance (ANCOVA) adjusted for the mean left and right rSO2 value at T3 (baseline). The secondary outcomes were analyzed with adjustment for the baseline value using a similar model to that used for the primary outcome. All outcomes at each measurement point were described using the mean, median, SD and 25th and 75th percentiles, and the 95% confidence interval was calculated. The Fisher exact test was used to compare the incidence of cerebral desaturation. Imputation was not used for missing data for any outcomes, as no data were missing. Subgroup and post hoc analyses were not performed.\nAll statistical analyses were performed with the R software program, version 4.0.3 (www.r-project.org). Two-sided P value of <.05 were considered statistically significant. There was no adjustment for multiple comparisons, as the analyses of secondary endpoints were interpreted as exploratory.", "This single-center, single-blinded, randomized clinical trial with parallel groups took place in Gifu University Hospital. The study protocol was approved by the Institutional Review Board of The Gifu University Hospital (decision number: 2019-274, approval date: March 16, 2020) and was registered at University hospital Medical Information Network Center (identifier: UMIN000039403, resister date: March 4, 2020).\nWe evaluated patients of ≥20 years of age who were scheduled for elective thoracic surgery requiring OLV in the lateral decubitus position at Gifu University Hospital from March to October 2020. Cases involving esophagectomy, robot-assisted thoracic surgery, lung resection for pneumothorax, surgery with carbon dioxide gas insufflation for pneumothorax and surgery with an expected duration of OLV < 30 minutes were not included in the analysis.\nParticipants who did not meet the exclusion criteria were asked to give their written informed consent before enrollment. The exclusion criteria were a medical history of lobectomy or segmentectomy, cerebrovascular disease, coronary artery disease, heart failure, or interstitial pneumonia (including suspicious cases); an American Society of Anesthesiologists physical status classification of ≥Ⅲ for any other reason; contraindication of study drugs; and any other reason deemed to render the participant inappropriate for intervention.\nThis study was conducted in accordance with the Consolidated Standards of Reporting Trials (CONSORT) 2010 statement.", "The data manager used the R software program, version 3.6.3 (www.r-project.org), to create a randomization sequence with 1:1 allocation, a mixed block size of 2 or 4, and no stratification. Researchers were not informed of the block size throughout the trial. The researchers assigned patients to either the propofol or desflurane group using sealed envelopes prepared according to a pregenerated allocation table. Sequence generation and randomization envelope preparation were performed by the data manager, independent of the researchers who had no further role in the trial. The patients were not informed of the treatment allocation.", "Before the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids.", "The subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study.", "The measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined.\nMeasurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation.\nAll complications during hospitalization were recorded.", "The primary outcome was the absolute value of the decrease in the mean left and right rSO2 values from T3 to T5. The secondary outcomes were the absolute value of the decrease in the rSO2 value of the dependent and nondependent lung side from T3 to T5, the absolute value of the decrease in the rSO2 value on the dominant and nondominant side of the subject from T3 to T5, the number of cerebral desaturations and complications occurring until discharge from the hospital.", "Prior to this study, a pilot study (decision number: 2018-210) was conducted to determine the sample size and collect results to select appropriate measurement values. In the pilot study, the absolute value of the decrease in the mean left and right rSO2 values from T3 to T5 was 14.0% in the propofol group and 9.8% in the desflurane group. Because the standard deviations (SDs) were 4.14 and 3.15, respectively, the SD was conservatively set at 4.14 in both groups. To detect a difference of 4.2% in the absolute value of the decrease in rSO2 between the propofol and desflurane groups by a t test with a detection power of 80% and at a 0.05 two-sided significance level, 16 cases per group (32 cases in total) were deemed necessary. We estimated that 10% of cases for per group would have unusable data, so we determined that 36 participants in whom measurements could be performed at least once during OLV were required. The full analysis set included all patients who received the protocol treatment at least once, except for those cases that were never evaluated for efficacy.\nBaseline characteristics and aggregated data are presented as the median (interquartile range: IQR) or mean ± SD for continuous variables and numbers (%) for categorical variables. The patient hemodynamic parameters and results of arterial blood gas analyses at T3 and T5 were compared between the 2 groups using Student t test. The primary outcome was analyzed according to the intent-to-treat principle with an analysis of covariance (ANCOVA) adjusted for the mean left and right rSO2 value at T3 (baseline). The secondary outcomes were analyzed with adjustment for the baseline value using a similar model to that used for the primary outcome. All outcomes at each measurement point were described using the mean, median, SD and 25th and 75th percentiles, and the 95% confidence interval was calculated. The Fisher exact test was used to compare the incidence of cerebral desaturation. Imputation was not used for missing data for any outcomes, as no data were missing. Subgroup and post hoc analyses were not performed.\nAll statistical analyses were performed with the R software program, version 4.0.3 (www.r-project.org). Two-sided P value of <.05 were considered statistically significant. There was no adjustment for multiple comparisons, as the analyses of secondary endpoints were interpreted as exploratory.", "Conceptualization: Keishu Hayashi, Kumiko Tanabe, Hiroki Iida.\nData curation: Keishu Hayashi.\nFormal analysis: Takuma Ishihara.\nFunding acquisition: Hiroki Iida.\nInvestigation: Keishu Hayashi, Yuko Yamada.\nMethodology: Kumiko Tanabe, Hiroki Iida.\nProject administration: Keishu Hayashi, Hiroki Iida.\nSupervision:Hiroki Iida.\nWriting-original draft: Keishu Hayashi, Yuko Yamada, Takuma Ishihara, Kumiko Tanabe.\nWriting-review and editing: Kumiko Tanabe, Hiroki Iida.\nAll authors read and approved the final articles." ]
[ "methods", null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Materials and Methods", "2.1. Study design and participants", "2.2. Randomization and masking", "2.3. Procedures", "2.3.1. Anesthetic management.", "2.3.2. Respiratory settings.", "2.3.3. Measurement values and time points.", "2.4. Outcomes", "2.5. Statistical analyses", "3. Results", "4. Discussion", "Author contributions" ]
[ "One-lung ventilation (OLV) is an indispensable anesthetic procedure for thoracic surgery. During OLV, the collapsed and unventilated lung still has perfusion that can cause intrapulmonary shunting, leading to impaired oxygenation and hypoxemia. Although a high fraction of inspiratory oxygen (FiO2) tends to be set to avoid hypoxemia during OLV, high FiO2 levels also increase the risk of postoperative respiratory complications and 30-day mortality.[1,2] Furthermore, oxidative stress during OLV causes lung injury, which is exacerbated by a higher FiO2.[3] The FiO2 should thus be kept as low as possible while maintaining the percutaneous oxygen saturation (SpO2) level within the appropriate range.[4]\nThere is some concern about an insufficient oxygen supply to vital organs, especially the central nervous system, during OLV. Cerebral oxygenation is evaluated by the regional cerebral oxygen saturation (rSO2) measurement. It has been reported that the rSO2 declines during OLV despite the SpO2 being above 90%, which may induce postoperative cognitive dysfunction.[5–9] Sungur et al[8] reported that preventing cerebral desaturation might reduce postoperative cognitive dysfunction.\nRecently, for adult patients, desflurane has been more commonly used as an anesthetic agent than sevoflurane because of its lower in vivo metabolic rate and faster emergence from general anesthesia. Desflurane has similar oxygenation, shunt fraction, and hemodynamics to sevoflurane during OLV.[10] Although arterial oxygenation during OLV has been reported to be reduced with desflurane-remifentanil anesthesia in comparison to propofol-remifentanil anesthesia, desflurane is nevertheless still frequently used as an anesthetic agent.[11]\nIn general, both propofol and desflurane reduce the cerebral metabolic rate for oxygen (CMRO2), although their effects on the cerebral blood flow (CBF) are not the same. Propofol has a cerebrovascular contractile effect and reduces both the CMRO2 and CBF.[12] Of note, the reduction in CBF outweighs the reduction in CMRO2 due to propofol, resulting in an overall reduction in the CBF/CMRO2 ratio.[13–15] Conversely, volatile anesthetic agents, such as desflurane, have a dose-dependent cerebrovascular dilator effect, which causes an increased CBF at concentrations >1 minimal alveolar concentration (MAC).[16–18] As such, the cerebral oxygen balance during OLV can be impaired under propofol anesthesia in comparison to volatile anesthetics. A previous study further showed that cerebral oxygen desaturation evaluated based on the jugular bulb venous oxygen saturation (SjO2) occurred more frequently under propofol anesthesia than under sevoflurane anesthesia during OLV, with an FiO2 of 0.5 and SpO2 of approximately 95%.[19] To our knowledge, no previous study has compared cerebral oxygenation during OLV under desflurane and propofol anesthesia.\nWe hypothesize that cerebral oxygenation is more markedly impaired during OLV under propofol anesthesia than under desflurane anesthesia. The present study therefore compared the effects of propofol and desflurane on cerebral oxygenation measured by rSO2 during OLV to avoid a high FiO2 level in patients undergoing thoracic surgery.", "[SUBTITLE] 2.1. Study design and participants [SUBSECTION] This single-center, single-blinded, randomized clinical trial with parallel groups took place in Gifu University Hospital. The study protocol was approved by the Institutional Review Board of The Gifu University Hospital (decision number: 2019-274, approval date: March 16, 2020) and was registered at University hospital Medical Information Network Center (identifier: UMIN000039403, resister date: March 4, 2020).\nWe evaluated patients of ≥20 years of age who were scheduled for elective thoracic surgery requiring OLV in the lateral decubitus position at Gifu University Hospital from March to October 2020. Cases involving esophagectomy, robot-assisted thoracic surgery, lung resection for pneumothorax, surgery with carbon dioxide gas insufflation for pneumothorax and surgery with an expected duration of OLV < 30 minutes were not included in the analysis.\nParticipants who did not meet the exclusion criteria were asked to give their written informed consent before enrollment. The exclusion criteria were a medical history of lobectomy or segmentectomy, cerebrovascular disease, coronary artery disease, heart failure, or interstitial pneumonia (including suspicious cases); an American Society of Anesthesiologists physical status classification of ≥Ⅲ for any other reason; contraindication of study drugs; and any other reason deemed to render the participant inappropriate for intervention.\nThis study was conducted in accordance with the Consolidated Standards of Reporting Trials (CONSORT) 2010 statement.\nThis single-center, single-blinded, randomized clinical trial with parallel groups took place in Gifu University Hospital. The study protocol was approved by the Institutional Review Board of The Gifu University Hospital (decision number: 2019-274, approval date: March 16, 2020) and was registered at University hospital Medical Information Network Center (identifier: UMIN000039403, resister date: March 4, 2020).\nWe evaluated patients of ≥20 years of age who were scheduled for elective thoracic surgery requiring OLV in the lateral decubitus position at Gifu University Hospital from March to October 2020. Cases involving esophagectomy, robot-assisted thoracic surgery, lung resection for pneumothorax, surgery with carbon dioxide gas insufflation for pneumothorax and surgery with an expected duration of OLV < 30 minutes were not included in the analysis.\nParticipants who did not meet the exclusion criteria were asked to give their written informed consent before enrollment. The exclusion criteria were a medical history of lobectomy or segmentectomy, cerebrovascular disease, coronary artery disease, heart failure, or interstitial pneumonia (including suspicious cases); an American Society of Anesthesiologists physical status classification of ≥Ⅲ for any other reason; contraindication of study drugs; and any other reason deemed to render the participant inappropriate for intervention.\nThis study was conducted in accordance with the Consolidated Standards of Reporting Trials (CONSORT) 2010 statement.\n[SUBTITLE] 2.2. Randomization and masking [SUBSECTION] The data manager used the R software program, version 3.6.3 (www.r-project.org), to create a randomization sequence with 1:1 allocation, a mixed block size of 2 or 4, and no stratification. Researchers were not informed of the block size throughout the trial. The researchers assigned patients to either the propofol or desflurane group using sealed envelopes prepared according to a pregenerated allocation table. Sequence generation and randomization envelope preparation were performed by the data manager, independent of the researchers who had no further role in the trial. The patients were not informed of the treatment allocation.\nThe data manager used the R software program, version 3.6.3 (www.r-project.org), to create a randomization sequence with 1:1 allocation, a mixed block size of 2 or 4, and no stratification. Researchers were not informed of the block size throughout the trial. The researchers assigned patients to either the propofol or desflurane group using sealed envelopes prepared according to a pregenerated allocation table. Sequence generation and randomization envelope preparation were performed by the data manager, independent of the researchers who had no further role in the trial. The patients were not informed of the treatment allocation.\n[SUBTITLE] 2.3. Procedures [SUBSECTION] The 36 total patients were randomly allocated to the desflurane or propofol group. After arriving in the operating room, a cerebral oximeter probe and electroencephalograph sensor were placed on the forehead of the subject and connected to a regional cerebral oximetry system (O3 regional oximetry; Masimo, Irvine, CA) and electroencephalograph monitor (SedLine sedation monitor; Masimo), respectively. The O3 regional oximetry recorded bilateral rSO2 by near-infrared spectroscopy, and the SedLine sedation monitor recorded the Patient State index. A 22-gauge catheter was placed into a radial artery and connected to a pressure transducer to monitor the arterial blood pressure, stroke volume variation and continuous cardiac index using the Vigileo system (Edwards Lifescience, Irvine, CA) with an arterial pressure waveform analysis sensor (FloTrac Sensor; Edwards Lifesciences).\n[SUBTITLE] 2.3.1. Anesthetic management. [SUBSECTION] Before the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids.\nBefore the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids.\n[SUBTITLE] 2.3.2. Respiratory settings. [SUBSECTION] The subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study.\nThe subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study.\n[SUBTITLE] 2.3.3. Measurement values and time points. [SUBSECTION] The measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined.\nMeasurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation.\nAll complications during hospitalization were recorded.\nThe measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined.\nMeasurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation.\nAll complications during hospitalization were recorded.\nThe 36 total patients were randomly allocated to the desflurane or propofol group. After arriving in the operating room, a cerebral oximeter probe and electroencephalograph sensor were placed on the forehead of the subject and connected to a regional cerebral oximetry system (O3 regional oximetry; Masimo, Irvine, CA) and electroencephalograph monitor (SedLine sedation monitor; Masimo), respectively. The O3 regional oximetry recorded bilateral rSO2 by near-infrared spectroscopy, and the SedLine sedation monitor recorded the Patient State index. A 22-gauge catheter was placed into a radial artery and connected to a pressure transducer to monitor the arterial blood pressure, stroke volume variation and continuous cardiac index using the Vigileo system (Edwards Lifescience, Irvine, CA) with an arterial pressure waveform analysis sensor (FloTrac Sensor; Edwards Lifesciences).\n[SUBTITLE] 2.3.1. Anesthetic management. [SUBSECTION] Before the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids.\nBefore the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids.\n[SUBTITLE] 2.3.2. Respiratory settings. [SUBSECTION] The subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study.\nThe subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study.\n[SUBTITLE] 2.3.3. Measurement values and time points. [SUBSECTION] The measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined.\nMeasurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation.\nAll complications during hospitalization were recorded.\nThe measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined.\nMeasurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation.\nAll complications during hospitalization were recorded.\n[SUBTITLE] 2.4. Outcomes [SUBSECTION] The primary outcome was the absolute value of the decrease in the mean left and right rSO2 values from T3 to T5. The secondary outcomes were the absolute value of the decrease in the rSO2 value of the dependent and nondependent lung side from T3 to T5, the absolute value of the decrease in the rSO2 value on the dominant and nondominant side of the subject from T3 to T5, the number of cerebral desaturations and complications occurring until discharge from the hospital.\nThe primary outcome was the absolute value of the decrease in the mean left and right rSO2 values from T3 to T5. The secondary outcomes were the absolute value of the decrease in the rSO2 value of the dependent and nondependent lung side from T3 to T5, the absolute value of the decrease in the rSO2 value on the dominant and nondominant side of the subject from T3 to T5, the number of cerebral desaturations and complications occurring until discharge from the hospital.\n[SUBTITLE] 2.5. Statistical analyses [SUBSECTION] Prior to this study, a pilot study (decision number: 2018-210) was conducted to determine the sample size and collect results to select appropriate measurement values. In the pilot study, the absolute value of the decrease in the mean left and right rSO2 values from T3 to T5 was 14.0% in the propofol group and 9.8% in the desflurane group. Because the standard deviations (SDs) were 4.14 and 3.15, respectively, the SD was conservatively set at 4.14 in both groups. To detect a difference of 4.2% in the absolute value of the decrease in rSO2 between the propofol and desflurane groups by a t test with a detection power of 80% and at a 0.05 two-sided significance level, 16 cases per group (32 cases in total) were deemed necessary. We estimated that 10% of cases for per group would have unusable data, so we determined that 36 participants in whom measurements could be performed at least once during OLV were required. The full analysis set included all patients who received the protocol treatment at least once, except for those cases that were never evaluated for efficacy.\nBaseline characteristics and aggregated data are presented as the median (interquartile range: IQR) or mean ± SD for continuous variables and numbers (%) for categorical variables. The patient hemodynamic parameters and results of arterial blood gas analyses at T3 and T5 were compared between the 2 groups using Student t test. The primary outcome was analyzed according to the intent-to-treat principle with an analysis of covariance (ANCOVA) adjusted for the mean left and right rSO2 value at T3 (baseline). The secondary outcomes were analyzed with adjustment for the baseline value using a similar model to that used for the primary outcome. All outcomes at each measurement point were described using the mean, median, SD and 25th and 75th percentiles, and the 95% confidence interval was calculated. The Fisher exact test was used to compare the incidence of cerebral desaturation. Imputation was not used for missing data for any outcomes, as no data were missing. Subgroup and post hoc analyses were not performed.\nAll statistical analyses were performed with the R software program, version 4.0.3 (www.r-project.org). Two-sided P value of <.05 were considered statistically significant. There was no adjustment for multiple comparisons, as the analyses of secondary endpoints were interpreted as exploratory.\nPrior to this study, a pilot study (decision number: 2018-210) was conducted to determine the sample size and collect results to select appropriate measurement values. In the pilot study, the absolute value of the decrease in the mean left and right rSO2 values from T3 to T5 was 14.0% in the propofol group and 9.8% in the desflurane group. Because the standard deviations (SDs) were 4.14 and 3.15, respectively, the SD was conservatively set at 4.14 in both groups. To detect a difference of 4.2% in the absolute value of the decrease in rSO2 between the propofol and desflurane groups by a t test with a detection power of 80% and at a 0.05 two-sided significance level, 16 cases per group (32 cases in total) were deemed necessary. We estimated that 10% of cases for per group would have unusable data, so we determined that 36 participants in whom measurements could be performed at least once during OLV were required. The full analysis set included all patients who received the protocol treatment at least once, except for those cases that were never evaluated for efficacy.\nBaseline characteristics and aggregated data are presented as the median (interquartile range: IQR) or mean ± SD for continuous variables and numbers (%) for categorical variables. The patient hemodynamic parameters and results of arterial blood gas analyses at T3 and T5 were compared between the 2 groups using Student t test. The primary outcome was analyzed according to the intent-to-treat principle with an analysis of covariance (ANCOVA) adjusted for the mean left and right rSO2 value at T3 (baseline). The secondary outcomes were analyzed with adjustment for the baseline value using a similar model to that used for the primary outcome. All outcomes at each measurement point were described using the mean, median, SD and 25th and 75th percentiles, and the 95% confidence interval was calculated. The Fisher exact test was used to compare the incidence of cerebral desaturation. Imputation was not used for missing data for any outcomes, as no data were missing. Subgroup and post hoc analyses were not performed.\nAll statistical analyses were performed with the R software program, version 4.0.3 (www.r-project.org). Two-sided P value of <.05 were considered statistically significant. There was no adjustment for multiple comparisons, as the analyses of secondary endpoints were interpreted as exploratory.", "This single-center, single-blinded, randomized clinical trial with parallel groups took place in Gifu University Hospital. The study protocol was approved by the Institutional Review Board of The Gifu University Hospital (decision number: 2019-274, approval date: March 16, 2020) and was registered at University hospital Medical Information Network Center (identifier: UMIN000039403, resister date: March 4, 2020).\nWe evaluated patients of ≥20 years of age who were scheduled for elective thoracic surgery requiring OLV in the lateral decubitus position at Gifu University Hospital from March to October 2020. Cases involving esophagectomy, robot-assisted thoracic surgery, lung resection for pneumothorax, surgery with carbon dioxide gas insufflation for pneumothorax and surgery with an expected duration of OLV < 30 minutes were not included in the analysis.\nParticipants who did not meet the exclusion criteria were asked to give their written informed consent before enrollment. The exclusion criteria were a medical history of lobectomy or segmentectomy, cerebrovascular disease, coronary artery disease, heart failure, or interstitial pneumonia (including suspicious cases); an American Society of Anesthesiologists physical status classification of ≥Ⅲ for any other reason; contraindication of study drugs; and any other reason deemed to render the participant inappropriate for intervention.\nThis study was conducted in accordance with the Consolidated Standards of Reporting Trials (CONSORT) 2010 statement.", "The data manager used the R software program, version 3.6.3 (www.r-project.org), to create a randomization sequence with 1:1 allocation, a mixed block size of 2 or 4, and no stratification. Researchers were not informed of the block size throughout the trial. The researchers assigned patients to either the propofol or desflurane group using sealed envelopes prepared according to a pregenerated allocation table. Sequence generation and randomization envelope preparation were performed by the data manager, independent of the researchers who had no further role in the trial. The patients were not informed of the treatment allocation.", "The 36 total patients were randomly allocated to the desflurane or propofol group. After arriving in the operating room, a cerebral oximeter probe and electroencephalograph sensor were placed on the forehead of the subject and connected to a regional cerebral oximetry system (O3 regional oximetry; Masimo, Irvine, CA) and electroencephalograph monitor (SedLine sedation monitor; Masimo), respectively. The O3 regional oximetry recorded bilateral rSO2 by near-infrared spectroscopy, and the SedLine sedation monitor recorded the Patient State index. A 22-gauge catheter was placed into a radial artery and connected to a pressure transducer to monitor the arterial blood pressure, stroke volume variation and continuous cardiac index using the Vigileo system (Edwards Lifescience, Irvine, CA) with an arterial pressure waveform analysis sensor (FloTrac Sensor; Edwards Lifesciences).\n[SUBTITLE] 2.3.1. Anesthetic management. [SUBSECTION] Before the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids.\nBefore the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids.\n[SUBTITLE] 2.3.2. Respiratory settings. [SUBSECTION] The subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study.\nThe subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study.\n[SUBTITLE] 2.3.3. Measurement values and time points. [SUBSECTION] The measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined.\nMeasurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation.\nAll complications during hospitalization were recorded.\nThe measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined.\nMeasurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation.\nAll complications during hospitalization were recorded.", "Before the induction of anesthesia, an epidural catheter was placed in patients planning to undergo lobectomy or segmentectomy. General anesthesia was induced with remifentanil and propofol (1.0–2.0 mg/kg) in the desflurane group and with remifentanil and target-controlled infusion of 2.0 to 5.0 µg/mL plasma concentration of propofol, calculated with Diprifusor (Astra-Zeneca Pharmaceuticals, Macclesfield, UK) in the propofol group. After muscle blockade with rocuronium, a left-sided double lumen tube was intubated endotracheally and placed using fiberscope. The patients were placed in the lateral decubitus position while bending the surgical table to widen the intercostal space. Anesthesia was maintained with remifentanil and either propofol or desflurane. The doses of propofol and desflurane were gradually adjusted to a Patient State index of 25 to 50. Hemodynamic management was performed based on the Vigileo monitor, and the mean arterial blood pressure (MAP) was maintained within 20% of the preinduction value by the administration of phenylephrine, ephedrine, and fluids.", "The subjects were mechanically ventilated with a mixture of air and oxygen at FiO2 0.4. An anesthesia workstation was used (Dräger Perseus A500; Dräger, Lübeck, Germany), and the subject was ventilated with the AutoFlow-volume control ventilation mode. The tidal volume was set at 8 mL/kg (predicted body weight), and the positive end-expiratory pressure was set at 5 cmH2O. The respiration rate was adjusted to maintain the arterial partial pressure of carbon dioxide (PaCO2) at approximately 40 mmHg. After the pleural incision was made, OLV was initiated with an FiO2 of 0.4, and the tidal volume was not changed unless the peak airway pressure exceeded 30 cmH2O. The FiO2 was adjusted to maintain an SpO2 of 92% to 94%, and the respiration rate was adjusted to maintain PaCO2 at 40 mmHg. When the SpO2 dropped to <90%, the FiO2 was temporarily increased to 1.0 and then decreased after the cause of the drop was corrected. Unless a left-right difference in rSO2 or a sudden decrease in rSO2 was observed, FiO2 did not increase in response to a decrease in rSO2. Epidural bolus was not administered until the end of the study.", "The measurement values included FiO2, heart rate, MAP, SpO2, body temperature, stroke volume variation and continuous cardiac index, Patient State index, left rSO2 (Lt. rSO2), right rSO2 (Rt. rSO2), concentration of propofol, end-tidal desflurane, concentration of remifentanil, and arterial blood gas sampling data (pH, arterial partial pressure of oxygen: PaO2, PaCO2, base excess, hematocrit). The measurements were performed at the following time points: after arrival in the operating room (T1); during mechanical two-lung ventilation just before position change (T2); just before the start of surgery (T3); 15 minutes after the initiation of OLV (T4); 30 minutes after the initiation of OLV (T5); during two-lung ventilation at the end of surgery (T6) (Fig. 1). We set the rSO2 value of T3 as the baseline value. Cerebral desaturation was defined as a relative decrease of ≥20% from the baseline rSO2 value or an absolute rSO2 value of <50%. The patient’s dominant hand was confirmed at the time of informed consent, and the dominant and nondominant sides were defined.\nMeasurement time points. T1: After arrival in the operating room. T2: Mechanical two-lung ventilation just before position change. T3 (baseline): Just before the start of surgery. T4: 15 minutes after the initiation of one-lung ventilation. T5: 30 minutes after the initiation of one-lung ventilation. T6: Two-lung ventilation at the end of surgery. FiO2 = fraction of inspiratory oxygen, OLV = one-lung ventilation, TLV = two-lung ventilation.\nAll complications during hospitalization were recorded.", "The primary outcome was the absolute value of the decrease in the mean left and right rSO2 values from T3 to T5. The secondary outcomes were the absolute value of the decrease in the rSO2 value of the dependent and nondependent lung side from T3 to T5, the absolute value of the decrease in the rSO2 value on the dominant and nondominant side of the subject from T3 to T5, the number of cerebral desaturations and complications occurring until discharge from the hospital.", "Prior to this study, a pilot study (decision number: 2018-210) was conducted to determine the sample size and collect results to select appropriate measurement values. In the pilot study, the absolute value of the decrease in the mean left and right rSO2 values from T3 to T5 was 14.0% in the propofol group and 9.8% in the desflurane group. Because the standard deviations (SDs) were 4.14 and 3.15, respectively, the SD was conservatively set at 4.14 in both groups. To detect a difference of 4.2% in the absolute value of the decrease in rSO2 between the propofol and desflurane groups by a t test with a detection power of 80% and at a 0.05 two-sided significance level, 16 cases per group (32 cases in total) were deemed necessary. We estimated that 10% of cases for per group would have unusable data, so we determined that 36 participants in whom measurements could be performed at least once during OLV were required. The full analysis set included all patients who received the protocol treatment at least once, except for those cases that were never evaluated for efficacy.\nBaseline characteristics and aggregated data are presented as the median (interquartile range: IQR) or mean ± SD for continuous variables and numbers (%) for categorical variables. The patient hemodynamic parameters and results of arterial blood gas analyses at T3 and T5 were compared between the 2 groups using Student t test. The primary outcome was analyzed according to the intent-to-treat principle with an analysis of covariance (ANCOVA) adjusted for the mean left and right rSO2 value at T3 (baseline). The secondary outcomes were analyzed with adjustment for the baseline value using a similar model to that used for the primary outcome. All outcomes at each measurement point were described using the mean, median, SD and 25th and 75th percentiles, and the 95% confidence interval was calculated. The Fisher exact test was used to compare the incidence of cerebral desaturation. Imputation was not used for missing data for any outcomes, as no data were missing. Subgroup and post hoc analyses were not performed.\nAll statistical analyses were performed with the R software program, version 4.0.3 (www.r-project.org). Two-sided P value of <.05 were considered statistically significant. There was no adjustment for multiple comparisons, as the analyses of secondary endpoints were interpreted as exploratory.", "Between March and October, a total of 99 participants were screened for eligibility. Four patients scheduled for surgery at the same time on the same day were excluded. Seven patients were excluded because of a past medical history of postoperative nausea and vomiting, and 9 patients were excluded for other reasons (chronic kidney disease in 3, atrial fibrillation in 2, thoracic empyema in 2, sequelae after resection of a metastatic brain tumor in 1, and another clinical trial in progress in 1).\nOf the remaining 39 participants, 20 patients were assigned to the propofol group and 19 to the desflurane group (CONSORT Flow Diagram, Figure 2). One patient in the propofol group and 2 in the desflurane group were excluded after randomization (canceled surgery, n = 1; diagnosis of interstitial pneumonia after the induction of anesthesia, n = 1; and severe desaturation event occurring immediately after the induction of anesthesia, n = 1). Thus, a total of 36 patients (propofol group, n = 19; desflurane group, n = 17) thus completed the study protocol, and their datasets were available for the analysis.\nFigure 2. CONSORT diagram of the study flow. ASA-PSC = American Society of Anesthesiologists physical status classification.\nThe baseline characteristics and intraoperative variables of these 36 patients are shown in Table 1. Figure 3A shows the mean rSO2 values at each measurement point: 61.5% ± 5.1% in the propofol group and 62.2% ± 6.0% in the desflurane group at T3, and 57.1% ± 5.3% in the propofol group and 58.6% ± 5.3% in the desflurane group at T5. There was no significant difference between the groups in the absolute value of the decrease in rSO2 adjusted by the rSO2 value at T3 (Table 2).\nCharacteristics of the patients.\nStatistics presented: median (interquartile range) for continuous variables; n (%) for categorical variables.\n%FEV1.0 = percent predicted forced expiratory volume in 1 second, ASA-PSC = American Society of Anesthesiologists physical status classification, FVC = forced vital capacity, FEV1.0 = forced expiratory volume in one second, OLV = one-lung ventilation, SpO2 = percutaneous oxygen saturation.\nPrimary and secondary outcomes: ANCOVA adjusted for the value of rSO2 at the T3.\n is the coefficient obtained from ANCOVA. If β of group is >0, this indicate that the absolute value of the decrease in rSO2 from T3 to T5 is greater in the Propofol group than Desflurane group, with adjustment for rSO2 at T3.\nCI = confidence interval, rSO2 = regional cerebral oxygen saturation, T3 = just before the start of surgery, T5 = 30 minutes after initiation of one-lung ventilation.\nChanges in (A) mean rSO2, (B) dependent lung side rSO2, and (C) dominant side rSO2. Each value represents the mean ± standard deviation. The solid circle and solid line represent the propofol group, and the open circle and interrupted line represent the desflurane group. See Figure 1. for the definitions of the measurement time points. dependent lung side rSO2 = rSO2 values on the dependent lung side, dominant side rSO2 = rSO2 values on the dominant side, mean rSO2 = mean of the left and right rSO2, rSO2 = regional cerebral oxygen saturation.\nAfter adjustment by the rSO2 value at T3, the absolute value of the decrease in rSO2 was significantly greater in the propofol group than in the desflurane group on both the dependent lung and the dominant sides (Table 2). Figure 3B and C shows the rSO2 values on the dependent lung and dominant lung sides, respectively, in both groups. The PaO2 value at T3 was significantly higher in the desflurane group. There were no significant differences in the MAP, continuous cardiac index, SpO2, PaCO2, any other hemodynamic parameters or the arterial blood gas data between the groups at T3 and T5 (Table 3). Furthermore, there were no significant differences in the incidence of cerebral desaturation events between the groups (propofol group, n = 5; desflurane group, n = 2; P = .41). One patient in each group showed a decrease in SpO2 immediately after the start of one-lung ventilation due to malposition of the double lumen tube, and the FiO2 was temporarily increased to 1.0 to adjust the tube position using a fiberscope. No cases required the adjustment of FiO2 in response to decreased rSO2. Complications occurred in 2 subjects in each group: paroxysmal atrial fibrillation and postoperative respiratory failure requiring admission to the intensive care unit in one patient each in the propofol group and prolonged pulmonary leakage and continuous atrial fibrillation requiring electrical ablation in the desflurane group.\nIntraoperative anesthetic variables and arterial blood gas data.\nSatistics presented: mean (SD).\nThere were not significant different in any values between the 2 groups at T3 and T5, respectively.\nBT = body temperature, Ce = effect site concentration, FiO2 = fraction of inspiratory oxygen, HR = heart rate, MAP = mean arterial blood pressure, PaCO2 = arterial partial pressure of carbon dioxide, PaO2 = arterial partial pressure of oxygen, SpO2 = percutaneous oxygen saturation, T3 = just before the start of surgery, T5 = 30 minutes after initiation of OLV.\nAll patients were discharged with no central neurological complications.", "The present study was performed to compare the effects of propofol and desflurane on cerebral oxygenation evaluated by rSO2 during OLV while avoiding a high FiO2 level. There were no significant differences between the 2 groups in the absolute value of the decrease in the mean rSO2 up to 30 minutes with an SpO2 of 92% to 94% after the initiation of OLV. We hypothesized that cerebral oxygenation would be more markedly impaired in the propofol group than in the desflurane group because of the greater reduction in the CBF/CMRO2 ratio by propofol. However, the present study indicated that propofol anesthesia was comparable to desflurane anesthesia with regard to cerebral oxygenation during OLV. Thus, both anesthetic agents can be safely used in routine clinical practice in patients who do not have symptomatic cerebrovascular or cardiovascular complications.\nDesaturation events during OLV can easily occur due to tube misalignment and tube obstruction with sputum. Thus, mechanical ventilation tends to be maintained at a higher FiO2 level to avoid desaturation events during OLV. However, a high FiO2 causes oxidative stress and is dose-dependently associated with respiratory complications and 30-day mortality.[2,3] Furthermore, hyperoxemia can carry a risk of myocardial ischemia, reduced cardiac output, reduced coronary blood flow, and reduced renal blood flow.[20] The optimal ranges of target FiO2 and SpO2 during general anesthesia for major surgery are unclear.[21] To prevent lung injury during OLV due to a high FiO2, the FiO2 should be kept as low as possible to maintain an SpO2 of ≥ 90%. In the present study, the SpO2 was maintained at 92% to 94% during OLV without the need for a high FiO2 (FiO2 values at T5: 0.33 ± 0.21 in the propofol group and 0.31 ± 0.12 in the desflurane group; SpO2 values at T5: approximately 93% in both groups). In general, the MAP, PaO2, and PaCO2 are closely related to cerebral perfusion and oxygenation. In the present study, the PaO2 values in the desflurane group were significantly higher than those in the propofol group at T3. However, the MAP and PaCO2 values did not differ to a statistically significant extent, and PaCO2 was maintained at approximately 40 mmHg in both groups at T3 and T5. In general, CBF is maintained when PaO2 is ≥60 mmHg. Therefore, the difference at T3 is not expected to affect CBF. This suggests that maintaining normocapnia and other hemodynamic parameters without significant fluctuations may prevent a significant reduction in rSO2, even under propofol anesthesia.\nAlthough both volatile anesthetics and propofol reduce the CMRO2 value, the effects on CBF differ between the 2 agents. Propofol reduces the CBF more markedly than the CMRO2, resulting in an overall reduction in the CBF/CMRO2.[12–15] In contrast, volatile anesthetics reduce the CMRO2 in a dose-dependent manner, and the CBF increases, depending on the MAC after maximal cerebral metabolic suppression, which mainly occurs after exceeding 1 MAC.[16–18,22] However, the effect on the CBF is unchanged or even slightly reduced with a low MAC.[22] We previously demonstrated that 0.5 MAC of desflurane had no significant vasodilatory effect on the diameter of rats’ cerebral pial arterioles.[23] Furthermore, Mielck et al[24] reported that 1 MAC desflurane significantly reduced the CBF in comparison to an awake state in healthy humans. In the present study, the end-expiratory desflurane concentration was 3.9 ± 0.2%, which was <1 MAC, even considering the age of the participants. Therefore, cerebrovascular dilation might not have occurred as expected, and no marked difference in the rSO2 reduction was noted between the desflurane and propofol groups.\nImportant factors associated with the cerebral oxygen supply are arterial oxygen saturation (SaO2), hemoglobin, cardiac output, and CBF. The cerebral oxygen demand during general anesthesia depends on the body temperature, seizure, type of anesthetic agent, and depth of anesthesia. Although oxygen dissolves directly in plasma depending on the PaO2 value, the amount is very small, and its role in the oxygen supply is minimal. The rSO2 is calculated by measuring the hemoglobin in arterial, venous, and capillary blood, assuming that 70% of the venous blood and 30% of the arterial blood are included in the cerebral blood volume.[25,26] Masimo O3 also calculates the rSO2 under this assumption, providing a close correlation with the reference cerebral oxygen saturation (0.3 SaO2 + 0.7 SjO2), as measured by blood gas sampling.[26] Although the rSO2 reflects the cerebral tissue oxygenation and blood flow, the rSO2 is more closely associated with SjO2 than SaO2 due to the high proportion of venous components.[25] Our study showed no marked changes in hemodynamic parameters from T3 to T5, suggesting that a decrease in rSO2 reflected a decrease in SaO2.\nThe present study showed that the absolute value of the decrease in rSO2 was significantly larger in the propofol group than in the desflurane group on both the dependent lung and dominant sides. The bilateral SjO2 values did not differ to a statistically significant extent in healthy patients in the supine position; however, postural change during anesthesia, such as adopting the Trendelenburg or beach chair positions, generally affects cerebral circulation.[27,28] In the present study, patients were placed in the lateral decubitus position with bending of the surgical table. The cerebral blood volume is presumed to increase on the dependent lung side while in the lateral decubitus position.[27] Blauenstein et al[29] reported that in healthy right-hand dominant patients, the CBF in the inferior frontal region is higher in the right hemisphere than in the left hemisphere. Although it has been reported that midazolam decreases the CBF in the left prefrontal cortex, we found no evidence that propofol or desflurane affects the laterality of the cerebral blood volume.[30] The significant difference in the rSO2 reduction between the 2 groups on both the dependent lung and dominant sides may be induced an increase in the cerebral blood volume or CBF because propofol has no vasodilatory effect. However, evidence regarding the laterality of cerebral oxygenation measured by the near-infrared spectroscopy system is insufficient to explain our results at present, and the degree of difference between the 2 groups is not an issue in clinical practice.\nThe present study was associated with several limitations. First, our study did not measure the CBF or CMRO2, so we were unable to determine whether the change in rSO2 was due to a change in the blood flow or a decrease in SaO2. While the intracranial pressure may also influence the CBF, this impact cannot be ruled out, as it was not measured in this study. Second, we did not strictly protocolize the phenylephrine and ephedrine doses, although the MAP was controlled. Because phenylephrine reduced the rSO2 value due to extracranial blood flow, our results might not solely reflect frontal lobe oxygenation.[31] Third, since postoperative cognitive dysfunction was not evaluated, the effect of a decreased rSO2 on the neurological prognosis was unclear.\nIn conclusion, our present study suggests that both propofol and desflurane anesthesia can maintain comparable cerebral oxygenation during OLV under an SpO2 of 92% to 94% and normocapnia settings in patients with no serious complications. As only patients with a healthy brain function were included in this study, cerebral oxygenation monitoring may be necessary in patients at risk for cerebrovascular disorders, such as cerebrovascular disease and cardiovascular disease.", "Conceptualization: Keishu Hayashi, Kumiko Tanabe, Hiroki Iida.\nData curation: Keishu Hayashi.\nFormal analysis: Takuma Ishihara.\nFunding acquisition: Hiroki Iida.\nInvestigation: Keishu Hayashi, Yuko Yamada.\nMethodology: Kumiko Tanabe, Hiroki Iida.\nProject administration: Keishu Hayashi, Hiroki Iida.\nSupervision:Hiroki Iida.\nWriting-original draft: Keishu Hayashi, Yuko Yamada, Takuma Ishihara, Kumiko Tanabe.\nWriting-review and editing: Kumiko Tanabe, Hiroki Iida.\nAll authors read and approved the final articles." ]
[ "intro", "methods", null, null, "methods", null, null, null, null, null, "results", "discussion", null ]
[ "cerebral blood flow", "cerebral oxygen saturation", "near-infrared spectroscopy", "one-lung ventilation", "oxidative stress", "thoracic surgery" ]
Comparative efficacy and safety of pharmacological interventions for severe COVID-19 patients: An updated network meta-analysis of 48 randomized controlled trials.
36254081
To date, there has been little agreement on what drug is the "best" drug for treating severe COVID-19 patients. This study aimed to assess the efficacy and safety of different medications available at present for severe COVID-19.
BACKGROUND
We searched databases for randomized controlled trials (RCTs) published up to February 28, 2022, with no language restrictions, of medications recommended for patients (aged 16 years or older) with severe COVID-19 infection. We extracted data on trials and patient characteristics, and the following primary outcomes: all-cause mortality (ACM), and treatment-emergent adverse events (TEAEs).
METHODS
We identified 4021 abstracts and of these included 48 RCTs comprising 9147 participants through database searches and other sources. For decrease in ACM, we found that ivermectin/doxycycline, C-IVIG (i.e., a hyperimmune anti-COVID-19 intravenous immunoglobulin), methylprednisolone, interferon-beta/standard-of-care (SOC), interferon-beta-1b, convalescent plasma, remdesivir, lopinavir/ritonavir, immunoglobulin gamma, high dosage sarilumab (HS), auxora, and imatinib were effective when compared with placebo or SOC group. We found that colchicine and interferon-beta/SOC were only associated with the TEAEs of severe COVID-19 patients.
RESULTS
This study suggested that ivermectin/doxycycline, C-IVIG, methylprednisolone, interferon-beta/SOC, interferon-beta-1b, convalescent plasma (CP), remdesivir, lopinavir/ritonavir, immunoglobulin gamma, HS, auxora, and imatinib were efficacious for treating severe COVID-19 patients. We found that most medications were safe in treating severe COVID-19. More large-scale RCTs are still needed to confirm the results of this study.
CONCLUSION
[ "COVID-19", "Colchicine", "Coronavirus Infections", "Doxycycline", "Humans", "Imatinib Mesylate", "Immunization, Passive", "Immunoglobulins, Intravenous", "Interferon beta-1b", "Ivermectin", "Lopinavir", "Methylprednisolone", "Network Meta-Analysis", "Pandemics", "Pneumonia, Viral", "Randomized Controlled Trials as Topic", "Ritonavir", "COVID-19 Serotherapy", "COVID-19 Drug Treatment" ]
9575403
1. Introduction
To date, the World Health Organization has confirmed over 480 million cases.[1] The mortality in patients with COVID-19 was estimated at 1.28%.[1] For the current analyses, COVID-19 infection is the leading cause of the global burden of disease and public health, which has increased significantly since 2019, driven mainly by high morbidity, high impact, and mortality.[2] Analysis of the data showed many pharmacologic interventions have been used to treat COVID-19 patients, which have been effective against COVID-19 infection.[3–5] However, there were many compounds that differ in efficacy and safety, and it was not clear which drug was the “best” drug for treating severe COVID-19 patients. Due to the small sample size and time-updated lagged in previous meta-analyses and studies, there has been debate about the effectiveness and safety of drugs, with some findings contradicting each other for COVID-19 infection.[6,7] Of primary concern was the medications on COVID-19 with all infection levels (i.e., mild, moderate and severe infection) in prior studies.[8] Here, we had a critical shortage that we lacked an analysis stratified by different infection levels in medications of COVID-19. It is well known that the disease varies in infection, which could lead to individual treatment of different infection subgroups in patients.[9] Though the network meta analysis (NMA) has been performed in prior reports,[7] there was little published data on the network study for the therapy of severe COVID-19 patients. Additionally, we found that there have been further randomized controlled trials (RCTs) of some other drugs for the treatment of COVID-19 through the literature search.[10] Some large-scale RCTs have been completed in the treatment of COVID-19 infection. We urgently need an updated assessment of the available evidence to support clinical decision-making. How do we select pharmacological interventions for severe COVID-19 patients in clinical practice? In order to fill this gap, we did an updated NMA of RCTs for drug interventions of severe COVID-19 infection. The purpose of this study is to evaluate the efficacy and safety of medications based on RCTs of the available evidence for severe COVID-19 infection.
2. Materials and Methods
[SUBTITLE] 2.1. Protocol and registration [SUBSECTION] We performed the present NMA in accordance with the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement.[11] This study was a review article and did not involve a research protocol requiring approval by a relevant institutional review board or ethics committee. Informed consent was also not applicable. We registered the protocol in the International Prospective Register of Systematic Reviews (PROSPERO) (CRD42021293879). We performed the present NMA in accordance with the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement.[11] This study was a review article and did not involve a research protocol requiring approval by a relevant institutional review board or ethics committee. Informed consent was also not applicable. We registered the protocol in the International Prospective Register of Systematic Reviews (PROSPERO) (CRD42021293879). [SUBTITLE] 2.2. Search strategy and selection criteria [SUBSECTION] We conducted a systematic literature search in 8 electronic databases (PubMed, Elsevier Science Direct, Cochrane Library, Google Scholar, Springer Link, MedRxiv, China National Knowledge Infrastructure, and Wanfangdata) for RCTs published, with no language restrictions from their inception to February 28, 2022. We included the RCTs on the treatment of severe COVID-19 patients (aged 16 years or older). The appendix had full search strategies listed (Supplemental Appendix 1, http://links.lww.com/MD/H533). We extracted data on RCTs, patient and medications characteristics (Table 1). Characteristics of randomized controlled trials of pharmacological interventions in severe patients with COVID-19. ACM = all-cause mortality, DRPCTs = double-blind, randomized placebo-controlled trials, COVID-19 = coronavirus disease 2019, MDRCTs = multicenter, double-blind, randomized controlled trials, MDRPCTs = multicenter, double-blind, randomized placebo-controlled trials, MRCTs = multicenter, randomized controlled trials, RCTs = randomized controlled trials, RPCTs = randomized placebo-controlled trials, RT-PCR = reverse transcription-polymerase chain reaction, SOC = standard of care, TEAEs = treatment-emergent adverse events, TRPCTs = three-blind, randomized placebo-controlled trials, UC-MSCs = human umbilical cord-derived mesenchymal stem cells. If the total number of gender is not equal to the number of participants, it is due to loss to follow-up from participants. To determine their eligibility, we reviewed the abstracts and full-texts of potentially relevant articles. We selected articles for the evaluation based on the criterion: at least one statistical analysis of the association between severe COVID-19 and medications was presented and described as an assessment for efficacy or safety. All RCTs that measured the efficacy or safety between drug interventions and severe COVID-19 infection were considered for inclusion. We listed full inclusion and exclusion criteria in the appendix (Supplemental Appendix 2, http://links.lww.com/MD/H534). During the selection of qualified studies, we resolved any ambiguity through mutual discussion and consensus. We conducted a systematic literature search in 8 electronic databases (PubMed, Elsevier Science Direct, Cochrane Library, Google Scholar, Springer Link, MedRxiv, China National Knowledge Infrastructure, and Wanfangdata) for RCTs published, with no language restrictions from their inception to February 28, 2022. We included the RCTs on the treatment of severe COVID-19 patients (aged 16 years or older). The appendix had full search strategies listed (Supplemental Appendix 1, http://links.lww.com/MD/H533). We extracted data on RCTs, patient and medications characteristics (Table 1). Characteristics of randomized controlled trials of pharmacological interventions in severe patients with COVID-19. ACM = all-cause mortality, DRPCTs = double-blind, randomized placebo-controlled trials, COVID-19 = coronavirus disease 2019, MDRCTs = multicenter, double-blind, randomized controlled trials, MDRPCTs = multicenter, double-blind, randomized placebo-controlled trials, MRCTs = multicenter, randomized controlled trials, RCTs = randomized controlled trials, RPCTs = randomized placebo-controlled trials, RT-PCR = reverse transcription-polymerase chain reaction, SOC = standard of care, TEAEs = treatment-emergent adverse events, TRPCTs = three-blind, randomized placebo-controlled trials, UC-MSCs = human umbilical cord-derived mesenchymal stem cells. If the total number of gender is not equal to the number of participants, it is due to loss to follow-up from participants. To determine their eligibility, we reviewed the abstracts and full-texts of potentially relevant articles. We selected articles for the evaluation based on the criterion: at least one statistical analysis of the association between severe COVID-19 and medications was presented and described as an assessment for efficacy or safety. All RCTs that measured the efficacy or safety between drug interventions and severe COVID-19 infection were considered for inclusion. We listed full inclusion and exclusion criteria in the appendix (Supplemental Appendix 2, http://links.lww.com/MD/H534). During the selection of qualified studies, we resolved any ambiguity through mutual discussion and consensus. [SUBTITLE] 2.3. Data extraction [SUBSECTION] At least 2 independent investigators (JQJ, CJF, and FZJ) extracted and entered onto all data through a standardized data extraction form. The main data extracted was the assessments of efficacy and safety. We collected the following information: basic characteristics, including author name, publication year, country/countries of origin, study design, method of COVID-19 testing, patient population, age, gender, sample size, interventions, treatment medication dose, controls, control medication dose, follow-up time, and primary outcomes; Primary outcomes, including all-cause mortality (ACM) and rate of treatment-emergent adverse events (TEAEs). We also contacted the authors if they did not report the above data information in the published article. One independent investigator undertook a preliminary extraction of studies, and another investigator reviewed the extraction. Differences were discussed, and a third researcher participated (CQL) if they reached no agreement. At least 2 independent investigators (JQJ, CJF, and FZJ) extracted and entered onto all data through a standardized data extraction form. The main data extracted was the assessments of efficacy and safety. We collected the following information: basic characteristics, including author name, publication year, country/countries of origin, study design, method of COVID-19 testing, patient population, age, gender, sample size, interventions, treatment medication dose, controls, control medication dose, follow-up time, and primary outcomes; Primary outcomes, including all-cause mortality (ACM) and rate of treatment-emergent adverse events (TEAEs). We also contacted the authors if they did not report the above data information in the published article. One independent investigator undertook a preliminary extraction of studies, and another investigator reviewed the extraction. Differences were discussed, and a third researcher participated (CQL) if they reached no agreement. [SUBTITLE] 2.4. Quality assessment [SUBSECTION] At least 2 investigators (ZG, CQL, JQJ, CJF, and FZJ) evaluated the risk of bias for all studies. We estimated the risk of bias with the Cochrane Risk-of-Bias Tool.[12] We assessed the certainty of evidence by using the Grading of Recommendations Assessment, Development, and Evaluation approach for the NMA.[13] At least 2 investigators (ZG, CQL, JQJ, CJF, and FZJ) evaluated the risk of bias for all studies. We estimated the risk of bias with the Cochrane Risk-of-Bias Tool.[12] We assessed the certainty of evidence by using the Grading of Recommendations Assessment, Development, and Evaluation approach for the NMA.[13] [SUBTITLE] 2.5. Outcome measures and definitions [SUBSECTION] The severe COVID-19 infection represented patients with fever or suspected respiratory infection, plus one of the following: respiratory rate > 30 breaths/min, severe respiratory distress, or SpO2 ≤ 93% on room air.[14,15] The primary outcomes were the ACM and TEAEs for severe COVID-19 patients, from the beginning of treatment to the end of follow-up. The definitions of ACM, TEAEs, and severe COVID-19 patients can refer to our previous study.[14] The severe COVID-19 infection represented patients with fever or suspected respiratory infection, plus one of the following: respiratory rate > 30 breaths/min, severe respiratory distress, or SpO2 ≤ 93% on room air.[14,15] The primary outcomes were the ACM and TEAEs for severe COVID-19 patients, from the beginning of treatment to the end of follow-up. The definitions of ACM, TEAEs, and severe COVID-19 patients can refer to our previous study.[14] [SUBTITLE] 2.6. Data analysis [SUBSECTION] [SUBTITLE] 2.6.1. Assessment of the transitivity assumption. [SUBSECTION] Transitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB). Transitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB). [SUBTITLE] 2.6.2. Network meta-analysis. [SUBSECTION] We used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05. We merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa. We used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05. We merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa. [SUBTITLE] 2.6.3. Assessment of heterogeneity and inconsistency. [SUBSECTION] We used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05. We used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05. [SUBTITLE] 2.6.4. Assessment of the risk of bias. [SUBSECTION] We assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17] We assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17] [SUBTITLE] 2.6.5. Sensitivity analysis. [SUBSECTION] The inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB. We used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes. The inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB. We used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes. [SUBTITLE] 2.6.1. Assessment of the transitivity assumption. [SUBSECTION] Transitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB). Transitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB). [SUBTITLE] 2.6.2. Network meta-analysis. [SUBSECTION] We used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05. We merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa. We used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05. We merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa. [SUBTITLE] 2.6.3. Assessment of heterogeneity and inconsistency. [SUBSECTION] We used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05. We used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05. [SUBTITLE] 2.6.4. Assessment of the risk of bias. [SUBSECTION] We assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17] We assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17] [SUBTITLE] 2.6.5. Sensitivity analysis. [SUBSECTION] The inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB. We used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes. The inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB. We used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes.
3. Results
[SUBTITLE] 3.1. Description of included studies [SUBSECTION] The search identified 4021 citations, out of which 3670 were excluded for duplications, wrong study design or population (i.e., non-severe COVID-19 infection), inappropriate intervention, not outcome/drug of interest, non-clinical studies, non-RCTs, review articles, commentaries, guidelines, and meta-analysis by checking titles and abstracts. The full texts of 351 articles were obtained to check eligibility, in which we excluded 308 articles for non- fulfilling eligibility criteria, unable to check eligibility and duplications. Finally, 43 studies[10,18–59] were included in our NMA. Figure 1 shows the selection process for the included studies. PRISMA flow-chart for study selection. Forty-eight RCTs, including 9147 patients, were included (Fig. 1) and described in Table 1. This analysis had a mean sample size of 106 [interquartile range 58–238]. The age of subjects was greater than or equal to 16 years. The median duration of follow-up treatment was 28 days (interquartile range 27–30). The search identified 4021 citations, out of which 3670 were excluded for duplications, wrong study design or population (i.e., non-severe COVID-19 infection), inappropriate intervention, not outcome/drug of interest, non-clinical studies, non-RCTs, review articles, commentaries, guidelines, and meta-analysis by checking titles and abstracts. The full texts of 351 articles were obtained to check eligibility, in which we excluded 308 articles for non- fulfilling eligibility criteria, unable to check eligibility and duplications. Finally, 43 studies[10,18–59] were included in our NMA. Figure 1 shows the selection process for the included studies. PRISMA flow-chart for study selection. Forty-eight RCTs, including 9147 patients, were included (Fig. 1) and described in Table 1. This analysis had a mean sample size of 106 [interquartile range 58–238]. The age of subjects was greater than or equal to 16 years. The median duration of follow-up treatment was 28 days (interquartile range 27–30). [SUBTITLE] 3.2. Quality appraisal [SUBSECTION] The included RCTs were of good quality (Fig. 2). Figure 2 also showed the risk of bias summary. The quality of the included randomized controlled trials. (A) Risk of bias summary (Note: The yellow circle with question mark represents “unclear risk of bias”, the red one with minus sign represents “high risk of bias” and the green one with plus sign represents “low risk of bias”). (B) Risk of bias graph. The included RCTs were of good quality (Fig. 2). Figure 2 also showed the risk of bias summary. The quality of the included randomized controlled trials. (A) Risk of bias summary (Note: The yellow circle with question mark represents “unclear risk of bias”, the red one with minus sign represents “high risk of bias” and the green one with plus sign represents “low risk of bias”). (B) Risk of bias graph. [SUBTITLE] 3.3. Network of evidence [SUBSECTION] In the network of connected RCTs (Fig. 3), the width of the lines corresponded to the number of trials included each treatment comparison. From Figure 3, we could see that the result was well connected. As shown in Figure 3A, the standard of care (SOC) was the most well-connected treatment, with chloroquine (CQ), hydroxychloroquine (HCQ), CQ/HCQ, convalescent plasma (CP), CP/SOC, remdesivir, remdesivir/SOC, lopinavir/ritonavir (LPV/r), interferon-beta-1b (IFN-β-1b), C-IVIG (i.e., a hyperimmune anti-COVID-19 intravenous immunoglobulin), ivermectin/doxycycline, IFN-β/SOC, tocilizumab, ruxolitinib/SOC, methylprednisolone, azithromycin/SOC, and auxora directly connected to it. Sarilumab such as high dosage sarilumab (HS) and low dosage sarilumab (LS), CQ/HCQ, ivermectin, canakinumab, colchicine, baricitinib, immunoglobulin gamma (IG), α-Lipoic acid, imatinib, mavrilimumab, lenzilumab, mycobacterium-w, otilimab, N-acetylcysteine, tocilizumab, high-dose intravenous vitamin C, hydrocortisone, and CP were directly connected to placebo in this network plot. Several sources of indirect evidence were available to inform comparisons between ivermectin, tocilizumab, HCQ, CP, auxora, placebo, and SOC (Fig. 3A). In Figure 3B, there was also a direct connection between SOC and C-IVIG, CP, LPV/r, ruxolitinib/SOC, ivermectin, IFN-β/SOC, and tocilizumab, or between placebo and HS, LS, mycobacterium-w, mavrilimumab, colchicine, canakinumab, baricitinib, human umbilical cord-derived mesenchymal stem cells, HCQ, lenzilumab, CP or tocilizumab. Several indirect connections were available between SOC and CP, tocilizumab, placebo (Fig. 3B). Network plot of eligible comparisons for all-cause mortality (A), and the ratio of treatment-emergent adverse events (B) for medications in patients with severe COVID-19. ALA= α-Lipoic acid, C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, IFN-β= interferon-beta, IG = immunoglobulin gamma, HDIVC = high-dose intravenous vitamin C, HS = high dosage sarilumab, LS = low dosage sarilumab, SOC = standard-of-care, UC-MSCs = human umbilical cord-derived mesenchymal stem cells. In the network of connected RCTs (Fig. 3), the width of the lines corresponded to the number of trials included each treatment comparison. From Figure 3, we could see that the result was well connected. As shown in Figure 3A, the standard of care (SOC) was the most well-connected treatment, with chloroquine (CQ), hydroxychloroquine (HCQ), CQ/HCQ, convalescent plasma (CP), CP/SOC, remdesivir, remdesivir/SOC, lopinavir/ritonavir (LPV/r), interferon-beta-1b (IFN-β-1b), C-IVIG (i.e., a hyperimmune anti-COVID-19 intravenous immunoglobulin), ivermectin/doxycycline, IFN-β/SOC, tocilizumab, ruxolitinib/SOC, methylprednisolone, azithromycin/SOC, and auxora directly connected to it. Sarilumab such as high dosage sarilumab (HS) and low dosage sarilumab (LS), CQ/HCQ, ivermectin, canakinumab, colchicine, baricitinib, immunoglobulin gamma (IG), α-Lipoic acid, imatinib, mavrilimumab, lenzilumab, mycobacterium-w, otilimab, N-acetylcysteine, tocilizumab, high-dose intravenous vitamin C, hydrocortisone, and CP were directly connected to placebo in this network plot. Several sources of indirect evidence were available to inform comparisons between ivermectin, tocilizumab, HCQ, CP, auxora, placebo, and SOC (Fig. 3A). In Figure 3B, there was also a direct connection between SOC and C-IVIG, CP, LPV/r, ruxolitinib/SOC, ivermectin, IFN-β/SOC, and tocilizumab, or between placebo and HS, LS, mycobacterium-w, mavrilimumab, colchicine, canakinumab, baricitinib, human umbilical cord-derived mesenchymal stem cells, HCQ, lenzilumab, CP or tocilizumab. Several indirect connections were available between SOC and CP, tocilizumab, placebo (Fig. 3B). Network plot of eligible comparisons for all-cause mortality (A), and the ratio of treatment-emergent adverse events (B) for medications in patients with severe COVID-19. ALA= α-Lipoic acid, C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, IFN-β= interferon-beta, IG = immunoglobulin gamma, HDIVC = high-dose intravenous vitamin C, HS = high dosage sarilumab, LS = low dosage sarilumab, SOC = standard-of-care, UC-MSCs = human umbilical cord-derived mesenchymal stem cells. [SUBTITLE] 3.4. Efficacy outcomes [SUBSECTION] Forty-one studies[10,18–38,40–51,53–59] reported ACM as outcome measurement (Supplemental Table S1, http://links.lww.com/MD/H536). We found that C-IVIG (OR 0.22, 95% CI 0.05-0.95), methylprednisolone (OR 0.27, 95% CI 0.09-0.77), IFN-β/SOC (OR 0.30, 95% CI 0.11-0.83), CP (OR 0.49, 95% CI 0.26-0.94), remdesivir (OR 0.58, 95% CI 0.37-0.93), and HS (OR 0.45, 95% CI 0.20-0.99) were associated with the decrease of ACM when compared with the SOC group (Fig. 4). Unfortunately, there was no significant difference in other 27 medications (e.g., ivermectin/doxycycline, IFN-β-1b, LPV/r, IG, LPV/r, and imatinib) or placebo for ACM when compared with SOC. Network meta-analyses of the relative efficacy and safety of medications for all-cause mortality among patients with severe COVID-19 infection. The red font and asterisk indicated comparisons that were statistically significant. ALA= α-Lipoic acid, C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, CI = confidence interval, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, HDIVC = high-dose intravenous vitamin C, HS = high dosage sarilumab, IFN-β=interferon-beta, IG = immunoglobulin gamma, LS = low dosage sarilumab, OR = odds ratio, SOC = standard-of-care. For a decrease in ACM, ivermectin/doxycycline (OR 0.06, 95% CI 0.00-0.88), C-IVIG (OR 0.12, 95% CI 0.02-0.61), methylprednisolone (OR 0.15, 95% CI 0.04-0.52), IG (OR 0.27, 95% CI 0.08-0.85), IFN-β/SOC (OR 0.17, 95% CI 0.05-0.56), IFN-β-1b (OR 0.16, 95% CI 0.03-0.98), auxora (OR 0.37, 95% CI 0.18-0.78), imatinib (OR 0.49, 95% CI 0.25-0.96), CP (OR 0.27, 95% CI 0.11-0.64), remdesivir (OR 0.32, 95% CI 0.14-0.74), and LPV/r (OR 0.32, 95% CI 0.12-0.85) were effective when compared with the placebo group (Fig. 4). We did not identify that there was a difference between placebo and other 22 medications (e.g., ruxolitinib/SOC, ivermectin, tocilizumab, HS, and mavrilimumab) or SOC for the ACM of severe COVID-19 infection (Fig. 4). The Supplemental Figure S1, http://links.lww.com/MD/H538 presented the ranking for the ACM of medications in severe COVID-19 patients based on cumulative probability plots and SUCRA. Forty-one studies[10,18–38,40–51,53–59] reported ACM as outcome measurement (Supplemental Table S1, http://links.lww.com/MD/H536). We found that C-IVIG (OR 0.22, 95% CI 0.05-0.95), methylprednisolone (OR 0.27, 95% CI 0.09-0.77), IFN-β/SOC (OR 0.30, 95% CI 0.11-0.83), CP (OR 0.49, 95% CI 0.26-0.94), remdesivir (OR 0.58, 95% CI 0.37-0.93), and HS (OR 0.45, 95% CI 0.20-0.99) were associated with the decrease of ACM when compared with the SOC group (Fig. 4). Unfortunately, there was no significant difference in other 27 medications (e.g., ivermectin/doxycycline, IFN-β-1b, LPV/r, IG, LPV/r, and imatinib) or placebo for ACM when compared with SOC. Network meta-analyses of the relative efficacy and safety of medications for all-cause mortality among patients with severe COVID-19 infection. The red font and asterisk indicated comparisons that were statistically significant. ALA= α-Lipoic acid, C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, CI = confidence interval, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, HDIVC = high-dose intravenous vitamin C, HS = high dosage sarilumab, IFN-β=interferon-beta, IG = immunoglobulin gamma, LS = low dosage sarilumab, OR = odds ratio, SOC = standard-of-care. For a decrease in ACM, ivermectin/doxycycline (OR 0.06, 95% CI 0.00-0.88), C-IVIG (OR 0.12, 95% CI 0.02-0.61), methylprednisolone (OR 0.15, 95% CI 0.04-0.52), IG (OR 0.27, 95% CI 0.08-0.85), IFN-β/SOC (OR 0.17, 95% CI 0.05-0.56), IFN-β-1b (OR 0.16, 95% CI 0.03-0.98), auxora (OR 0.37, 95% CI 0.18-0.78), imatinib (OR 0.49, 95% CI 0.25-0.96), CP (OR 0.27, 95% CI 0.11-0.64), remdesivir (OR 0.32, 95% CI 0.14-0.74), and LPV/r (OR 0.32, 95% CI 0.12-0.85) were effective when compared with the placebo group (Fig. 4). We did not identify that there was a difference between placebo and other 22 medications (e.g., ruxolitinib/SOC, ivermectin, tocilizumab, HS, and mavrilimumab) or SOC for the ACM of severe COVID-19 infection (Fig. 4). The Supplemental Figure S1, http://links.lww.com/MD/H538 presented the ranking for the ACM of medications in severe COVID-19 patients based on cumulative probability plots and SUCRA. [SUBTITLE] 3.5. Safety outcomes [SUBSECTION] In the safety outcome, data from 19 studies[18,20,21,23,26–29,34,38–40,48,50–54,56]were merged for analysis (Supplemental Table S2, http://links.lww.com/MD/H537). We found that colchicine (OR 2.77, 95% CI 1.03-7.42) and IFN-β/SOC (OR 18.98, 95% CI 1.70-211.84) seemed to increase the risk of TEAEs when compared with the SOC group. Moreover, IFN-β/SOC (OR 19.00, 95% CI 2.36-153.10) were associated with the increase of TEAEs when compared with placebo. We found no significant difference in TEAEs in other 15 medications for severe COVID-19 patients when compared with SOC or placebo (Fig. 5). Network meta-analyses of the relative efficacy and safety of medications for the ratio of treatment-emergent adverse events among patients with severe COVID-19 infection. The red font and asterisk indicated comparisons that were statistically significant. C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, CI = confidence interval, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, HS = high dosage sarilumab, IFN-β=interferon-beta, LS = low dosage sarilumab, OR = odds ratio, SOC = standard-of-care, UC-MSCs = human umbilical cord-derived mesenchymal stem cells. The figure of SUCRA showed that baricitinib had the highest cumulative probability (SUCRA: 88.3%) becoming the best intervention in TEAEs, followed by LPV/r (SUCRA: 78.8%), SOC (SUCRA: 76.0%), lenzilumab (SUCRA: 72.6%), C-IVIG (SUCRA: 68.5%), tocilizumab (SUCRA: 63.6%), placebo (SUCRA: 53.1%), ivermectin (SUCRA: 52.3%), canakinumab (SUCRA: 51.9%), mycobacterium-w (SUCRA: 50.1%), LS (SUCRA: 49.3%), mavrilimumab (SUCRA: 47.1%), ruxolitinib/SOC (SUCRA: 46.8%), CP (SUCRA: 44.5%), HS (SUCRA: 31.0%), HCQ (SUCRA: 29.5%), human umbilical cord-derived mesenchymal stem cells (SUCRA: 29.1%), colchicine (SUCRA: 14.2%), and IFN-β/SOC (SUCRA: 3.2%) (Supplemental Figure S2, http://links.lww.com/MD/H539). In the safety outcome, data from 19 studies[18,20,21,23,26–29,34,38–40,48,50–54,56]were merged for analysis (Supplemental Table S2, http://links.lww.com/MD/H537). We found that colchicine (OR 2.77, 95% CI 1.03-7.42) and IFN-β/SOC (OR 18.98, 95% CI 1.70-211.84) seemed to increase the risk of TEAEs when compared with the SOC group. Moreover, IFN-β/SOC (OR 19.00, 95% CI 2.36-153.10) were associated with the increase of TEAEs when compared with placebo. We found no significant difference in TEAEs in other 15 medications for severe COVID-19 patients when compared with SOC or placebo (Fig. 5). Network meta-analyses of the relative efficacy and safety of medications for the ratio of treatment-emergent adverse events among patients with severe COVID-19 infection. The red font and asterisk indicated comparisons that were statistically significant. C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, CI = confidence interval, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, HS = high dosage sarilumab, IFN-β=interferon-beta, LS = low dosage sarilumab, OR = odds ratio, SOC = standard-of-care, UC-MSCs = human umbilical cord-derived mesenchymal stem cells. The figure of SUCRA showed that baricitinib had the highest cumulative probability (SUCRA: 88.3%) becoming the best intervention in TEAEs, followed by LPV/r (SUCRA: 78.8%), SOC (SUCRA: 76.0%), lenzilumab (SUCRA: 72.6%), C-IVIG (SUCRA: 68.5%), tocilizumab (SUCRA: 63.6%), placebo (SUCRA: 53.1%), ivermectin (SUCRA: 52.3%), canakinumab (SUCRA: 51.9%), mycobacterium-w (SUCRA: 50.1%), LS (SUCRA: 49.3%), mavrilimumab (SUCRA: 47.1%), ruxolitinib/SOC (SUCRA: 46.8%), CP (SUCRA: 44.5%), HS (SUCRA: 31.0%), HCQ (SUCRA: 29.5%), human umbilical cord-derived mesenchymal stem cells (SUCRA: 29.1%), colchicine (SUCRA: 14.2%), and IFN-β/SOC (SUCRA: 3.2%) (Supplemental Figure S2, http://links.lww.com/MD/H539). [SUBTITLE] 3.6. Evaluation of inconsistency [SUBSECTION] According to the inconsistency test (Table 2), no significant inconsistency or qualitative difference was available for the ACM and TEAEs. Thus, the consistency hypothesis was accepted in this NMA. The evaluation of inconsistency for the efficacy and safety of medications among severe COVID-19 patients. COVID-19 = coronavirus disease 2019. According to the inconsistency test (Table 2), no significant inconsistency or qualitative difference was available for the ACM and TEAEs. Thus, the consistency hypothesis was accepted in this NMA. The evaluation of inconsistency for the efficacy and safety of medications among severe COVID-19 patients. COVID-19 = coronavirus disease 2019. [SUBTITLE] 3.7. Sensitivity analysis and publication bias [SUBSECTION] We analyzed the potential sources of heterogeneity or inconsistency by using subgroup and meta-regression analyses. Univariable meta-regression and subgroup analyses indicated that there were heterogeneous sources (such as DS, blinding and RRB) for the ACM (P < .05) (Fig. 6A). Whilst the SS and RRB were the heterogeneity source of TEAEs based on the sensitivity analysis (P < .05) (Fig. 6B). Meta-regression and sensitivity analyses for the efficacy and safety of medications in patients with severe COVID-19 infection. (A) all-cause mortality. (B) the ratio of treatment-emergent adverse events. CD = crossover design, COVID-19 = coronavirus disease 2019, DS = duration of study, IS = industry sponsorship, MS = multicenter study, RRB = risk of reported bias, SS = sample size. None of the funnel plots of outcomes (ACM and TEAEs) indicated a significant asymmetry (Supplemental Figure S3, http://links.lww.com/MD/H540). We analyzed the potential sources of heterogeneity or inconsistency by using subgroup and meta-regression analyses. Univariable meta-regression and subgroup analyses indicated that there were heterogeneous sources (such as DS, blinding and RRB) for the ACM (P < .05) (Fig. 6A). Whilst the SS and RRB were the heterogeneity source of TEAEs based on the sensitivity analysis (P < .05) (Fig. 6B). Meta-regression and sensitivity analyses for the efficacy and safety of medications in patients with severe COVID-19 infection. (A) all-cause mortality. (B) the ratio of treatment-emergent adverse events. CD = crossover design, COVID-19 = coronavirus disease 2019, DS = duration of study, IS = industry sponsorship, MS = multicenter study, RRB = risk of reported bias, SS = sample size. None of the funnel plots of outcomes (ACM and TEAEs) indicated a significant asymmetry (Supplemental Figure S3, http://links.lww.com/MD/H540).
5. Conclusions
In conclusion, this NMA demonstrated that ivermectin/doxycycline, C-IVIG, methylprednisolone, IFN-β/SOC, IFN-β-1b, CP, remdesivir, LPV/r, IG, HS, auxora, and imatinib were effective for treating severe COVID-19 patients. There may be a difference in the findings of medications for severe COVID-19 patients from different control conditions (i.e., placebo and SOC) in RCTs. We found that most medications were safe in treating severe COVID-19. The present NMA reported uncertain estimates on the efficacy and safety of medications in the severe COVID-19 treatment. Maybe it’s because there was inadequate evidence of a reduction in ACM and the absence of TEAEs. However, this study had 2 strengths. One was that a comprehensive meta-analysis strategy was used to reduce the risk of publication bias. The other was that the SUCRA was used to assess possibly the best intervention. Despite these limitations, to date, the present findings might represent the most comprehensive meta-analysis of the available evidence for severe COVID-19 infection. Future guidelines and decision-making treatment plan should consider these results for the severe COVID-19 treatment. Importantly, the government, academia and researchers should collaborate to develop more large-scale RCTs studies and further estimate the efficacy and safety of treatment interventions on mortality, virological and clinical outcomes for different levels of infection with COVID-19.
[ "2.1. Protocol and registration", "2.2. Search strategy and selection criteria", "2.3. Data extraction", "2.4. Quality assessment", "2.5. Outcome measures and definitions", "2.6. Data analysis", "2.6.1. Assessment of the transitivity assumption.", "2.6.2. Network meta-analysis.", "2.6.3. Assessment of heterogeneity and inconsistency.", "2.6.4. Assessment of the risk of bias.", "2.6.5. Sensitivity analysis.", "3.1. Description of included studies", "3.2. Quality appraisal", "3.3. Network of evidence", "3.4. Efficacy outcomes", "3.5. Safety outcomes", "3.6. Evaluation of inconsistency", "3.7. Sensitivity analysis and publication bias", "4.1. Efficacy of current medications", "4.2. Safety of current medications", "Author contributions", "Acknowledgments" ]
[ "We performed the present NMA in accordance with the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement.[11] This study was a review article and did not involve a research protocol requiring approval by a relevant institutional review board or ethics committee. Informed consent was also not applicable. We registered the protocol in the International Prospective Register of Systematic Reviews (PROSPERO) (CRD42021293879).", "We conducted a systematic literature search in 8 electronic databases (PubMed, Elsevier Science Direct, Cochrane Library, Google Scholar, Springer Link, MedRxiv, China National Knowledge Infrastructure, and Wanfangdata) for RCTs published, with no language restrictions from their inception to February 28, 2022. We included the RCTs on the treatment of severe COVID-19 patients (aged 16 years or older). The appendix had full search strategies listed (Supplemental Appendix 1, http://links.lww.com/MD/H533). We extracted data on RCTs, patient and medications characteristics (Table 1).\nCharacteristics of randomized controlled trials of pharmacological interventions in severe patients with COVID-19.\nACM = all-cause mortality, DRPCTs = double-blind, randomized placebo-controlled trials, COVID-19 = coronavirus disease 2019, MDRCTs = multicenter, double-blind, randomized controlled trials, MDRPCTs = multicenter, double-blind, randomized placebo-controlled trials, MRCTs = multicenter, randomized controlled trials, RCTs = randomized controlled trials, RPCTs = randomized placebo-controlled trials, RT-PCR = reverse transcription-polymerase chain reaction, SOC = standard of care, TEAEs = treatment-emergent adverse events, TRPCTs = three-blind, randomized placebo-controlled trials, UC-MSCs = human umbilical cord-derived mesenchymal stem cells.\nIf the total number of gender is not equal to the number of participants, it is due to loss to follow-up from participants.\nTo determine their eligibility, we reviewed the abstracts and full-texts of potentially relevant articles. We selected articles for the evaluation based on the criterion: at least one statistical analysis of the association between severe COVID-19 and medications was presented and described as an assessment for efficacy or safety.\nAll RCTs that measured the efficacy or safety between drug interventions and severe COVID-19 infection were considered for inclusion. We listed full inclusion and exclusion criteria in the appendix (Supplemental Appendix 2, http://links.lww.com/MD/H534). During the selection of qualified studies, we resolved any ambiguity through mutual discussion and consensus.", "At least 2 independent investigators (JQJ, CJF, and FZJ) extracted and entered onto all data through a standardized data extraction form. The main data extracted was the assessments of efficacy and safety. We collected the following information: basic characteristics, including author name, publication year, country/countries of origin, study design, method of COVID-19 testing, patient population, age, gender, sample size, interventions, treatment medication dose, controls, control medication dose, follow-up time, and primary outcomes; Primary outcomes, including all-cause mortality (ACM) and rate of treatment-emergent adverse events (TEAEs). We also contacted the authors if they did not report the above data information in the published article. One independent investigator undertook a preliminary extraction of studies, and another investigator reviewed the extraction. Differences were discussed, and a third researcher participated (CQL) if they reached no agreement.", "At least 2 investigators (ZG, CQL, JQJ, CJF, and FZJ) evaluated the risk of bias for all studies. We estimated the risk of bias with the Cochrane Risk-of-Bias Tool.[12] We assessed the certainty of evidence by using the Grading of Recommendations Assessment, Development, and Evaluation approach for the NMA.[13]", "The severe COVID-19 infection represented patients with fever or suspected respiratory infection, plus one of the following: respiratory rate > 30 breaths/min, severe respiratory distress, or SpO2 ≤ 93% on room air.[14,15] The primary outcomes were the ACM and TEAEs for severe COVID-19 patients, from the beginning of treatment to the end of follow-up. The definitions of ACM, TEAEs, and severe COVID-19 patients can refer to our previous study.[14]", "[SUBTITLE] 2.6.1. Assessment of the transitivity assumption. [SUBSECTION] Transitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB).\nTransitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB).\n[SUBTITLE] 2.6.2. Network meta-analysis. [SUBSECTION] We used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05.\nWe merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa.\nWe used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05.\nWe merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa.\n[SUBTITLE] 2.6.3. Assessment of heterogeneity and inconsistency. [SUBSECTION] We used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05.\nWe used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05.\n[SUBTITLE] 2.6.4. Assessment of the risk of bias. [SUBSECTION] We assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17]\nWe assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17]\n[SUBTITLE] 2.6.5. Sensitivity analysis. [SUBSECTION] The inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB.\nWe used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes.\nThe inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB.\nWe used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes.", "Transitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB).", "We used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05.\nWe merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa.", "We used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05.", "We assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17]", "The inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB.\nWe used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes.", "The search identified 4021 citations, out of which 3670 were excluded for duplications, wrong study design or population (i.e., non-severe COVID-19 infection), inappropriate intervention, not outcome/drug of interest, non-clinical studies, non-RCTs, review articles, commentaries, guidelines, and meta-analysis by checking titles and abstracts. The full texts of 351 articles were obtained to check eligibility, in which we excluded 308 articles for non- fulfilling eligibility criteria, unable to check eligibility and duplications. Finally, 43 studies[10,18–59] were included in our NMA. Figure 1 shows the selection process for the included studies.\nPRISMA flow-chart for study selection.\nForty-eight RCTs, including 9147 patients, were included (Fig. 1) and described in Table 1. This analysis had a mean sample size of 106 [interquartile range 58–238]. The age of subjects was greater than or equal to 16 years. The median duration of follow-up treatment was 28 days (interquartile range 27–30).", "The included RCTs were of good quality (Fig. 2). Figure 2 also showed the risk of bias summary.\nThe quality of the included randomized controlled trials. (A) Risk of bias summary (Note: The yellow circle with question mark represents “unclear risk of bias”, the red one with minus sign represents “high risk of bias” and the green one with plus sign represents “low risk of bias”). (B) Risk of bias graph.", "In the network of connected RCTs (Fig. 3), the width of the lines corresponded to the number of trials included each treatment comparison. From Figure 3, we could see that the result was well connected. As shown in Figure 3A, the standard of care (SOC) was the most well-connected treatment, with chloroquine (CQ), hydroxychloroquine (HCQ), CQ/HCQ, convalescent plasma (CP), CP/SOC, remdesivir, remdesivir/SOC, lopinavir/ritonavir (LPV/r), interferon-beta-1b (IFN-β-1b), C-IVIG (i.e., a hyperimmune anti-COVID-19 intravenous immunoglobulin), ivermectin/doxycycline, IFN-β/SOC, tocilizumab, ruxolitinib/SOC, methylprednisolone, azithromycin/SOC, and auxora directly connected to it. Sarilumab such as high dosage sarilumab (HS) and low dosage sarilumab (LS), CQ/HCQ, ivermectin, canakinumab, colchicine, baricitinib, immunoglobulin gamma (IG), α-Lipoic acid, imatinib, mavrilimumab, lenzilumab, mycobacterium-w, otilimab, N-acetylcysteine, tocilizumab, high-dose intravenous vitamin C, hydrocortisone, and CP were directly connected to placebo in this network plot. Several sources of indirect evidence were available to inform comparisons between ivermectin, tocilizumab, HCQ, CP, auxora, placebo, and SOC (Fig. 3A). In Figure 3B, there was also a direct connection between SOC and C-IVIG, CP, LPV/r, ruxolitinib/SOC, ivermectin, IFN-β/SOC, and tocilizumab, or between placebo and HS, LS, mycobacterium-w, mavrilimumab, colchicine, canakinumab, baricitinib, human umbilical cord-derived mesenchymal stem cells, HCQ, lenzilumab, CP or tocilizumab. Several indirect connections were available between SOC and CP, tocilizumab, placebo (Fig. 3B).\nNetwork plot of eligible comparisons for all-cause mortality (A), and the ratio of treatment-emergent adverse events (B) for medications in patients with severe COVID-19. ALA= α-Lipoic acid, C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, IFN-β= interferon-beta, IG = immunoglobulin gamma, HDIVC = high-dose intravenous vitamin C, HS = high dosage sarilumab, LS = low dosage sarilumab, SOC = standard-of-care, UC-MSCs = human umbilical cord-derived mesenchymal stem cells.", "Forty-one studies[10,18–38,40–51,53–59] reported ACM as outcome measurement (Supplemental Table S1, http://links.lww.com/MD/H536). We found that C-IVIG (OR 0.22, 95% CI 0.05-0.95), methylprednisolone (OR 0.27, 95% CI 0.09-0.77), IFN-β/SOC (OR 0.30, 95% CI 0.11-0.83), CP (OR 0.49, 95% CI 0.26-0.94), remdesivir (OR 0.58, 95% CI 0.37-0.93), and HS (OR 0.45, 95% CI 0.20-0.99) were associated with the decrease of ACM when compared with the SOC group (Fig. 4). Unfortunately, there was no significant difference in other 27 medications (e.g., ivermectin/doxycycline, IFN-β-1b, LPV/r, IG, LPV/r, and imatinib) or placebo for ACM when compared with SOC.\nNetwork meta-analyses of the relative efficacy and safety of medications for all-cause mortality among patients with severe COVID-19 infection. The red font and asterisk indicated comparisons that were statistically significant. ALA= α-Lipoic acid, C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, CI = confidence interval, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, HDIVC = high-dose intravenous vitamin C, HS = high dosage sarilumab, IFN-β=interferon-beta, IG = immunoglobulin gamma, LS = low dosage sarilumab, OR = odds ratio, SOC = standard-of-care.\nFor a decrease in ACM, ivermectin/doxycycline (OR 0.06, 95% CI 0.00-0.88), C-IVIG (OR 0.12, 95% CI 0.02-0.61), methylprednisolone (OR 0.15, 95% CI 0.04-0.52), IG (OR 0.27, 95% CI 0.08-0.85), IFN-β/SOC (OR 0.17, 95% CI 0.05-0.56), IFN-β-1b (OR 0.16, 95% CI 0.03-0.98), auxora (OR 0.37, 95% CI 0.18-0.78), imatinib (OR 0.49, 95% CI 0.25-0.96), CP (OR 0.27, 95% CI 0.11-0.64), remdesivir (OR 0.32, 95% CI 0.14-0.74), and LPV/r (OR 0.32, 95% CI 0.12-0.85) were effective when compared with the placebo group (Fig. 4). We did not identify that there was a difference between placebo and other 22 medications (e.g., ruxolitinib/SOC, ivermectin, tocilizumab, HS, and mavrilimumab) or SOC for the ACM of severe COVID-19 infection (Fig. 4).\nThe Supplemental Figure S1, http://links.lww.com/MD/H538 presented the ranking for the ACM of medications in severe COVID-19 patients based on cumulative probability plots and SUCRA.", "In the safety outcome, data from 19 studies[18,20,21,23,26–29,34,38–40,48,50–54,56]were merged for analysis (Supplemental Table S2, http://links.lww.com/MD/H537). We found that colchicine (OR 2.77, 95% CI 1.03-7.42) and IFN-β/SOC (OR 18.98, 95% CI 1.70-211.84) seemed to increase the risk of TEAEs when compared with the SOC group. Moreover, IFN-β/SOC (OR 19.00, 95% CI 2.36-153.10) were associated with the increase of TEAEs when compared with placebo. We found no significant difference in TEAEs in other 15 medications for severe COVID-19 patients when compared with SOC or placebo (Fig. 5).\nNetwork meta-analyses of the relative efficacy and safety of medications for the ratio of treatment-emergent adverse events among patients with severe COVID-19 infection. The red font and asterisk indicated comparisons that were statistically significant. C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, CI = confidence interval, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, HS = high dosage sarilumab, IFN-β=interferon-beta, LS = low dosage sarilumab, OR = odds ratio, SOC = standard-of-care, UC-MSCs = human umbilical cord-derived mesenchymal stem cells.\nThe figure of SUCRA showed that baricitinib had the highest cumulative probability (SUCRA: 88.3%) becoming the best intervention in TEAEs, followed by LPV/r (SUCRA: 78.8%), SOC (SUCRA: 76.0%), lenzilumab (SUCRA: 72.6%), C-IVIG (SUCRA: 68.5%), tocilizumab (SUCRA: 63.6%), placebo (SUCRA: 53.1%), ivermectin (SUCRA: 52.3%), canakinumab (SUCRA: 51.9%), mycobacterium-w (SUCRA: 50.1%), LS (SUCRA: 49.3%), mavrilimumab (SUCRA: 47.1%), ruxolitinib/SOC (SUCRA: 46.8%), CP (SUCRA: 44.5%), HS (SUCRA: 31.0%), HCQ (SUCRA: 29.5%), human umbilical cord-derived mesenchymal stem cells (SUCRA: 29.1%), colchicine (SUCRA: 14.2%), and IFN-β/SOC (SUCRA: 3.2%) (Supplemental Figure S2, http://links.lww.com/MD/H539).", "According to the inconsistency test (Table 2), no significant inconsistency or qualitative difference was available for the ACM and TEAEs. Thus, the consistency hypothesis was accepted in this NMA.\nThe evaluation of inconsistency for the efficacy and safety of medications among severe COVID-19 patients.\nCOVID-19 = coronavirus disease 2019.", "We analyzed the potential sources of heterogeneity or inconsistency by using subgroup and meta-regression analyses. Univariable meta-regression and subgroup analyses indicated that there were heterogeneous sources (such as DS, blinding and RRB) for the ACM (P < .05) (Fig. 6A). Whilst the SS and RRB were the heterogeneity source of TEAEs based on the sensitivity analysis (P < .05) (Fig. 6B).\nMeta-regression and sensitivity analyses for the efficacy and safety of medications in patients with severe COVID-19 infection. (A) all-cause mortality. (B) the ratio of treatment-emergent adverse events. CD = crossover design, COVID-19 = coronavirus disease 2019, DS = duration of study, IS = industry sponsorship, MS = multicenter study, RRB = risk of reported bias, SS = sample size.\nNone of the funnel plots of outcomes (ACM and TEAEs) indicated a significant asymmetry (Supplemental Figure S3, http://links.lww.com/MD/H540).", "This study supported evidence from previous observations.[72,73] As we know, intravenous immunoglobulin has already been validated as an effective antiviral drug for treating COVID-19, SARS and Middle East respiratory syndrome.[20,74,75] This is consistent with our results (i.e., C-IVIG, HS, and IG). Recent evidence suggested that anti-cytokine effects, inhibition of complement activation, and down-regulation of B and T cells’ functions by C-IVIG can prevent organ failure and subsequent mortality in severe COVID-19 patients.[20] Our findings also confirmed that sarilumab could prevent patients with COVID-19 from progressing to death. Sarilumab is a fully human antibody against the interleukin (IL)-6 receptor, which can rapidly lower C-reactive protein and mediate COVID-19 clearance.[76] Prolonged glucocorticoid treatment is associated with improved outcomes of acute respiratory distress syndrome.[77] Several reports have also shown that treatment with methylprednisolone could significantly reduce the risk of death among patients with acute respiratory distress syndrome.[78] This also accords with our observations, which suggested that methylprednisolone was associated with decreased ACM in severe COVID-19 patients.\nThe SUCRA indicated that ivermectin/doxycycline was the highest ranked intervention with a SUCRA of 0.821. Previous studies showed that ivermectin or doxycycline is effective in treating COVID-19 patients.[79,80] We attribute this to a couple of reasons. First, ivermectin or doxycycline might possess antiviral as well as immunomodulatory activity.[79,80] Second, they might have anti-inflammatory and immunomodulatory agents and can curb over-reacting innate and cellular immune responses.[81] One possible implication may be that the ivermectin/doxycycline is an excellent selection for treating severe COVID-19 patients. Of note, based on the result of meta-regression analysis on the heterogeneity (such as DS, blinding, and RRB), the present result may need further verification. Thus, statistical indications of clinical superiority in this network analysis required careful interpretation.\nInterestingly, we found that small-molecule protein kinase inhibitors (i.e., auxora and imatinib) were efficacious for the therapy of severe COVID-19 infection. Recent research has established that a calcium release-activated calcium channel inhibitor, such as auxora, can reduce the occurrence of COVID-19 death due to blocking the release of multiple pro-inflammatory cytokines, including IL-6. Additionally, evidence showed that imatinib might play its potentially antiviral and beneficial immunomodulatory role in severe COVID-19 patients.[82] These results further support those of previous studies.[25,83] Collectively, the present findings indicate that the small-molecule protein kinase inhibitors provide a new, and perhaps superior, avenue for the severe COVID-19 treatment. In addition, our results may provide a robust strategy for clinical combination therapy among severe COVID-19 patients.\nWe have demonstrated that IFN-β/SOC, IFN-β-1b, remdesivir, LPV/r and CP were associated with a reduction of ACM in severe COVID-19 infection. Several lines of evidence suggested that IFN deficiency was a hallmark of severe COVID-19.[84] IFN might treat severe COVID-19 infection through adapted anti-inflammatory therapies that target IL-6 or TNF-β.[84,85] Data from prior studies suggested that remdesivir was a broad-spectrum antiviral activity against RNA viruses, which could improve the survival of COVID-19 infection due to inhibiting viral replication.[86] Recent studies reported that LPV/r displayed inhibitory activities against SARS-CoV-2 main protease and inhibited SARS-CoV-2 replication in Vero E6 cells.[87] Similarly to other antibodies therapy, CP can prevent the death of severe COVID-19 patients because of alleviating the inflammation and overreaction of the immune system through antibodies.[88] These findings further support the idea of our NMA study.\nConversely, no significant difference was found in other 21 medications (e.g., HCQ, α-Lipoic acid, hydrocortisone, otilimab, and mavrilimumab) for the ACM of severe COVID-19 infection when compared with SOC or placebo. This finding was contrary to previous studies which have suggested that some antiviral drugs (hydrocortisone, mavrilimumab, etc) were associated with decreased ACM in patients with COVID-19.[29,32,89,90] It is difficult to explain this result, but it might be related to the difference of participants’ selection in differential studies. For instance, the findings of this NMA were based on a larger sample size (i.e., more participants were included). Correspondingly, most of the previous studies presented a smaller sample size.[29,32] The present study differed from earlier studies,[90,91] which selected subjects with all infection levels (i.e., mild, moderate and severe infections). Furthermore, prior studies were inadequate for the analyses stratified by different infection levels (i.e., non-severe and severe infection) in medications of COVID-19.[29,32,89,90] Prior studies suggested that COVID-19 patients at different infection levels often led to different outcomes of treatment.[92] Another possible explanation for this was that we compared the efficacy and safety of SOC, which existed the bias due to the differential SOC of every country (i.e., the SOC is not standardized) except for the reasons given above.[21,23,45] The present study raised the possibility that our findings might be beneficial to guiding the selection of drug interventions for clinicians in severe COVID-19 patients.", "In terms of safety, we summarized the TEAEs. We found that colchicine and IFN-β/SOC were only associated with the TEAEs of severe COVID-19 patients in this study. Recent meta-analysis studies and large-scale RCTs[45,46,92] seemed to be consistent with our findings, which identified most pharmacological treatments had a good safety in treating severe COVID-19. However, as mentioned in the present study, colchicine and IFN-β/SOC should be chosen cautiously in treating severe COVID-19 patients based on safety. Colchicine might increase the TEAEs in treating patients with severe COVID-19 infection. It is difficult to explain this result, but it might be related to the toxicity (e.g., gastrointestinal mucosal damage) in the case of colchicine treatment.[93] In addition, it is possible that the use of IFN-β in combination with SOC was associated with increased TEAEs in treating severe COVID-19, which should be weighed in all future IFN-β studies.[94]\nIn summary, clinicians might need to select treatment regimens based on the ranks of efficacy and safety (i.e., SUCRA) when medications are used in treating severe COVID-19 patients. Additionally, it should be reminded that further studies, which reduce the effects of SS and RRB, will need to be undertaken based on the result of sensitivity analysis.\nSome limitations constrained this study. Firstly, included studies might be small in this NMA, which should be considered when interpreting the findings. Secondly, the published data we extracted included only 2 types of outcomes, some important outcomes such as discharge ratio, clinical improvement ratio, and the ratio of virological cure, were not analyzed. Thirdly, although we did our best to include all available RCTs, we cannot eliminate the possibility of missing data. Fourthly, some nodes in our network included only a few trials. The sample size of actual head-to-head trials was very small. Hence, comparative efficacy and safety between interventions was frequently based on indirect comparisons. Finally, the sensitivity analysis showed that there were several heterogeneity sources, which may conceal or exaggerate the effect size of this network analysis. Further large-scale RCTs, which control these confounding factors, will need to be undertaken to verify our findings. Though there are still many shortcomings in our research, it is certain that the prevention and therapy of COVID-19 is set to change for the better in the future.", "Conceptualization: Cheng Qing Lin, Zhao Gang.\nData curation: Chen Jun Fang, Jia Qing Jun, Fang Zi Jian.\nFormal analysis: Cheng Qing Lin, Zhao Gang.\nFunding acquisition: Cheng Qing Lin.\nMethodology: Cheng Qing Lin, Chen Jun Fang.\nSupervision: Jia Qing Jun, Fang Zi Jian.\nWriting – original draft: Cheng Qing Lin.\nWriting – review & editing: Cheng Qing Lin, Zhao Gang.", "We acknowledge with gratitude all study authors who responded to our data requests. We thank many researchers who sent information for our previous reviews on which this report was built. This work is supported by the Basic Public Welfare Research Project of Zhejiang Province (grant number: LGF21H260007), the Medical Science and Technology Project of Zhejiang Province (grant numbers: 2020PY064 and 2021PY065), and the Key Medical Discipline construction project (Disinfection and Vector Control) of Hangzhou." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Materials and Methods", "2.1. Protocol and registration", "2.2. Search strategy and selection criteria", "2.3. Data extraction", "2.4. Quality assessment", "2.5. Outcome measures and definitions", "2.6. Data analysis", "2.6.1. Assessment of the transitivity assumption.", "2.6.2. Network meta-analysis.", "2.6.3. Assessment of heterogeneity and inconsistency.", "2.6.4. Assessment of the risk of bias.", "2.6.5. Sensitivity analysis.", "3. Results", "3.1. Description of included studies", "3.2. Quality appraisal", "3.3. Network of evidence", "3.4. Efficacy outcomes", "3.5. Safety outcomes", "3.6. Evaluation of inconsistency", "3.7. Sensitivity analysis and publication bias", "4. Discussion", "4.1. Efficacy of current medications", "4.2. Safety of current medications", "5. Conclusions", "Author contributions", "Acknowledgments", "Supplementary Material" ]
[ "To date, the World Health Organization has confirmed over 480 million cases.[1] The mortality in patients with COVID-19 was estimated at 1.28%.[1] For the current analyses, COVID-19 infection is the leading cause of the global burden of disease and public health, which has increased significantly since 2019, driven mainly by high morbidity, high impact, and mortality.[2]\nAnalysis of the data showed many pharmacologic interventions have been used to treat COVID-19 patients, which have been effective against COVID-19 infection.[3–5] However, there were many compounds that differ in efficacy and safety, and it was not clear which drug was the “best” drug for treating severe COVID-19 patients. Due to the small sample size and time-updated lagged in previous meta-analyses and studies, there has been debate about the effectiveness and safety of drugs, with some findings contradicting each other for COVID-19 infection.[6,7]\nOf primary concern was the medications on COVID-19 with all infection levels (i.e., mild, moderate and severe infection) in prior studies.[8] Here, we had a critical shortage that we lacked an analysis stratified by different infection levels in medications of COVID-19. It is well known that the disease varies in infection, which could lead to individual treatment of different infection subgroups in patients.[9]\nThough the network meta analysis (NMA) has been performed in prior reports,[7] there was little published data on the network study for the therapy of severe COVID-19 patients. Additionally, we found that there have been further randomized controlled trials (RCTs) of some other drugs for the treatment of COVID-19 through the literature search.[10] Some large-scale RCTs have been completed in the treatment of COVID-19 infection. We urgently need an updated assessment of the available evidence to support clinical decision-making.\nHow do we select pharmacological interventions for severe COVID-19 patients in clinical practice? In order to fill this gap, we did an updated NMA of RCTs for drug interventions of severe COVID-19 infection. The purpose of this study is to evaluate the efficacy and safety of medications based on RCTs of the available evidence for severe COVID-19 infection.", "[SUBTITLE] 2.1. Protocol and registration [SUBSECTION] We performed the present NMA in accordance with the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement.[11] This study was a review article and did not involve a research protocol requiring approval by a relevant institutional review board or ethics committee. Informed consent was also not applicable. We registered the protocol in the International Prospective Register of Systematic Reviews (PROSPERO) (CRD42021293879).\nWe performed the present NMA in accordance with the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement.[11] This study was a review article and did not involve a research protocol requiring approval by a relevant institutional review board or ethics committee. Informed consent was also not applicable. We registered the protocol in the International Prospective Register of Systematic Reviews (PROSPERO) (CRD42021293879).\n[SUBTITLE] 2.2. Search strategy and selection criteria [SUBSECTION] We conducted a systematic literature search in 8 electronic databases (PubMed, Elsevier Science Direct, Cochrane Library, Google Scholar, Springer Link, MedRxiv, China National Knowledge Infrastructure, and Wanfangdata) for RCTs published, with no language restrictions from their inception to February 28, 2022. We included the RCTs on the treatment of severe COVID-19 patients (aged 16 years or older). The appendix had full search strategies listed (Supplemental Appendix 1, http://links.lww.com/MD/H533). We extracted data on RCTs, patient and medications characteristics (Table 1).\nCharacteristics of randomized controlled trials of pharmacological interventions in severe patients with COVID-19.\nACM = all-cause mortality, DRPCTs = double-blind, randomized placebo-controlled trials, COVID-19 = coronavirus disease 2019, MDRCTs = multicenter, double-blind, randomized controlled trials, MDRPCTs = multicenter, double-blind, randomized placebo-controlled trials, MRCTs = multicenter, randomized controlled trials, RCTs = randomized controlled trials, RPCTs = randomized placebo-controlled trials, RT-PCR = reverse transcription-polymerase chain reaction, SOC = standard of care, TEAEs = treatment-emergent adverse events, TRPCTs = three-blind, randomized placebo-controlled trials, UC-MSCs = human umbilical cord-derived mesenchymal stem cells.\nIf the total number of gender is not equal to the number of participants, it is due to loss to follow-up from participants.\nTo determine their eligibility, we reviewed the abstracts and full-texts of potentially relevant articles. We selected articles for the evaluation based on the criterion: at least one statistical analysis of the association between severe COVID-19 and medications was presented and described as an assessment for efficacy or safety.\nAll RCTs that measured the efficacy or safety between drug interventions and severe COVID-19 infection were considered for inclusion. We listed full inclusion and exclusion criteria in the appendix (Supplemental Appendix 2, http://links.lww.com/MD/H534). During the selection of qualified studies, we resolved any ambiguity through mutual discussion and consensus.\nWe conducted a systematic literature search in 8 electronic databases (PubMed, Elsevier Science Direct, Cochrane Library, Google Scholar, Springer Link, MedRxiv, China National Knowledge Infrastructure, and Wanfangdata) for RCTs published, with no language restrictions from their inception to February 28, 2022. We included the RCTs on the treatment of severe COVID-19 patients (aged 16 years or older). The appendix had full search strategies listed (Supplemental Appendix 1, http://links.lww.com/MD/H533). We extracted data on RCTs, patient and medications characteristics (Table 1).\nCharacteristics of randomized controlled trials of pharmacological interventions in severe patients with COVID-19.\nACM = all-cause mortality, DRPCTs = double-blind, randomized placebo-controlled trials, COVID-19 = coronavirus disease 2019, MDRCTs = multicenter, double-blind, randomized controlled trials, MDRPCTs = multicenter, double-blind, randomized placebo-controlled trials, MRCTs = multicenter, randomized controlled trials, RCTs = randomized controlled trials, RPCTs = randomized placebo-controlled trials, RT-PCR = reverse transcription-polymerase chain reaction, SOC = standard of care, TEAEs = treatment-emergent adverse events, TRPCTs = three-blind, randomized placebo-controlled trials, UC-MSCs = human umbilical cord-derived mesenchymal stem cells.\nIf the total number of gender is not equal to the number of participants, it is due to loss to follow-up from participants.\nTo determine their eligibility, we reviewed the abstracts and full-texts of potentially relevant articles. We selected articles for the evaluation based on the criterion: at least one statistical analysis of the association between severe COVID-19 and medications was presented and described as an assessment for efficacy or safety.\nAll RCTs that measured the efficacy or safety between drug interventions and severe COVID-19 infection were considered for inclusion. We listed full inclusion and exclusion criteria in the appendix (Supplemental Appendix 2, http://links.lww.com/MD/H534). During the selection of qualified studies, we resolved any ambiguity through mutual discussion and consensus.\n[SUBTITLE] 2.3. Data extraction [SUBSECTION] At least 2 independent investigators (JQJ, CJF, and FZJ) extracted and entered onto all data through a standardized data extraction form. The main data extracted was the assessments of efficacy and safety. We collected the following information: basic characteristics, including author name, publication year, country/countries of origin, study design, method of COVID-19 testing, patient population, age, gender, sample size, interventions, treatment medication dose, controls, control medication dose, follow-up time, and primary outcomes; Primary outcomes, including all-cause mortality (ACM) and rate of treatment-emergent adverse events (TEAEs). We also contacted the authors if they did not report the above data information in the published article. One independent investigator undertook a preliminary extraction of studies, and another investigator reviewed the extraction. Differences were discussed, and a third researcher participated (CQL) if they reached no agreement.\nAt least 2 independent investigators (JQJ, CJF, and FZJ) extracted and entered onto all data through a standardized data extraction form. The main data extracted was the assessments of efficacy and safety. We collected the following information: basic characteristics, including author name, publication year, country/countries of origin, study design, method of COVID-19 testing, patient population, age, gender, sample size, interventions, treatment medication dose, controls, control medication dose, follow-up time, and primary outcomes; Primary outcomes, including all-cause mortality (ACM) and rate of treatment-emergent adverse events (TEAEs). We also contacted the authors if they did not report the above data information in the published article. One independent investigator undertook a preliminary extraction of studies, and another investigator reviewed the extraction. Differences were discussed, and a third researcher participated (CQL) if they reached no agreement.\n[SUBTITLE] 2.4. Quality assessment [SUBSECTION] At least 2 investigators (ZG, CQL, JQJ, CJF, and FZJ) evaluated the risk of bias for all studies. We estimated the risk of bias with the Cochrane Risk-of-Bias Tool.[12] We assessed the certainty of evidence by using the Grading of Recommendations Assessment, Development, and Evaluation approach for the NMA.[13]\nAt least 2 investigators (ZG, CQL, JQJ, CJF, and FZJ) evaluated the risk of bias for all studies. We estimated the risk of bias with the Cochrane Risk-of-Bias Tool.[12] We assessed the certainty of evidence by using the Grading of Recommendations Assessment, Development, and Evaluation approach for the NMA.[13]\n[SUBTITLE] 2.5. Outcome measures and definitions [SUBSECTION] The severe COVID-19 infection represented patients with fever or suspected respiratory infection, plus one of the following: respiratory rate > 30 breaths/min, severe respiratory distress, or SpO2 ≤ 93% on room air.[14,15] The primary outcomes were the ACM and TEAEs for severe COVID-19 patients, from the beginning of treatment to the end of follow-up. The definitions of ACM, TEAEs, and severe COVID-19 patients can refer to our previous study.[14]\nThe severe COVID-19 infection represented patients with fever or suspected respiratory infection, plus one of the following: respiratory rate > 30 breaths/min, severe respiratory distress, or SpO2 ≤ 93% on room air.[14,15] The primary outcomes were the ACM and TEAEs for severe COVID-19 patients, from the beginning of treatment to the end of follow-up. The definitions of ACM, TEAEs, and severe COVID-19 patients can refer to our previous study.[14]\n[SUBTITLE] 2.6. Data analysis [SUBSECTION] [SUBTITLE] 2.6.1. Assessment of the transitivity assumption. [SUBSECTION] Transitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB).\nTransitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB).\n[SUBTITLE] 2.6.2. Network meta-analysis. [SUBSECTION] We used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05.\nWe merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa.\nWe used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05.\nWe merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa.\n[SUBTITLE] 2.6.3. Assessment of heterogeneity and inconsistency. [SUBSECTION] We used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05.\nWe used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05.\n[SUBTITLE] 2.6.4. Assessment of the risk of bias. [SUBSECTION] We assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17]\nWe assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17]\n[SUBTITLE] 2.6.5. Sensitivity analysis. [SUBSECTION] The inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB.\nWe used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes.\nThe inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB.\nWe used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes.\n[SUBTITLE] 2.6.1. Assessment of the transitivity assumption. [SUBSECTION] Transitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB).\nTransitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB).\n[SUBTITLE] 2.6.2. Network meta-analysis. [SUBSECTION] We used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05.\nWe merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa.\nWe used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05.\nWe merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa.\n[SUBTITLE] 2.6.3. Assessment of heterogeneity and inconsistency. [SUBSECTION] We used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05.\nWe used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05.\n[SUBTITLE] 2.6.4. Assessment of the risk of bias. [SUBSECTION] We assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17]\nWe assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17]\n[SUBTITLE] 2.6.5. Sensitivity analysis. [SUBSECTION] The inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB.\nWe used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes.\nThe inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB.\nWe used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes.", "We performed the present NMA in accordance with the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement.[11] This study was a review article and did not involve a research protocol requiring approval by a relevant institutional review board or ethics committee. Informed consent was also not applicable. We registered the protocol in the International Prospective Register of Systematic Reviews (PROSPERO) (CRD42021293879).", "We conducted a systematic literature search in 8 electronic databases (PubMed, Elsevier Science Direct, Cochrane Library, Google Scholar, Springer Link, MedRxiv, China National Knowledge Infrastructure, and Wanfangdata) for RCTs published, with no language restrictions from their inception to February 28, 2022. We included the RCTs on the treatment of severe COVID-19 patients (aged 16 years or older). The appendix had full search strategies listed (Supplemental Appendix 1, http://links.lww.com/MD/H533). We extracted data on RCTs, patient and medications characteristics (Table 1).\nCharacteristics of randomized controlled trials of pharmacological interventions in severe patients with COVID-19.\nACM = all-cause mortality, DRPCTs = double-blind, randomized placebo-controlled trials, COVID-19 = coronavirus disease 2019, MDRCTs = multicenter, double-blind, randomized controlled trials, MDRPCTs = multicenter, double-blind, randomized placebo-controlled trials, MRCTs = multicenter, randomized controlled trials, RCTs = randomized controlled trials, RPCTs = randomized placebo-controlled trials, RT-PCR = reverse transcription-polymerase chain reaction, SOC = standard of care, TEAEs = treatment-emergent adverse events, TRPCTs = three-blind, randomized placebo-controlled trials, UC-MSCs = human umbilical cord-derived mesenchymal stem cells.\nIf the total number of gender is not equal to the number of participants, it is due to loss to follow-up from participants.\nTo determine their eligibility, we reviewed the abstracts and full-texts of potentially relevant articles. We selected articles for the evaluation based on the criterion: at least one statistical analysis of the association between severe COVID-19 and medications was presented and described as an assessment for efficacy or safety.\nAll RCTs that measured the efficacy or safety between drug interventions and severe COVID-19 infection were considered for inclusion. We listed full inclusion and exclusion criteria in the appendix (Supplemental Appendix 2, http://links.lww.com/MD/H534). During the selection of qualified studies, we resolved any ambiguity through mutual discussion and consensus.", "At least 2 independent investigators (JQJ, CJF, and FZJ) extracted and entered onto all data through a standardized data extraction form. The main data extracted was the assessments of efficacy and safety. We collected the following information: basic characteristics, including author name, publication year, country/countries of origin, study design, method of COVID-19 testing, patient population, age, gender, sample size, interventions, treatment medication dose, controls, control medication dose, follow-up time, and primary outcomes; Primary outcomes, including all-cause mortality (ACM) and rate of treatment-emergent adverse events (TEAEs). We also contacted the authors if they did not report the above data information in the published article. One independent investigator undertook a preliminary extraction of studies, and another investigator reviewed the extraction. Differences were discussed, and a third researcher participated (CQL) if they reached no agreement.", "At least 2 investigators (ZG, CQL, JQJ, CJF, and FZJ) evaluated the risk of bias for all studies. We estimated the risk of bias with the Cochrane Risk-of-Bias Tool.[12] We assessed the certainty of evidence by using the Grading of Recommendations Assessment, Development, and Evaluation approach for the NMA.[13]", "The severe COVID-19 infection represented patients with fever or suspected respiratory infection, plus one of the following: respiratory rate > 30 breaths/min, severe respiratory distress, or SpO2 ≤ 93% on room air.[14,15] The primary outcomes were the ACM and TEAEs for severe COVID-19 patients, from the beginning of treatment to the end of follow-up. The definitions of ACM, TEAEs, and severe COVID-19 patients can refer to our previous study.[14]", "[SUBTITLE] 2.6.1. Assessment of the transitivity assumption. [SUBSECTION] Transitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB).\nTransitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB).\n[SUBTITLE] 2.6.2. Network meta-analysis. [SUBSECTION] We used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05.\nWe merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa.\nWe used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05.\nWe merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa.\n[SUBTITLE] 2.6.3. Assessment of heterogeneity and inconsistency. [SUBSECTION] We used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05.\nWe used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05.\n[SUBTITLE] 2.6.4. Assessment of the risk of bias. [SUBSECTION] We assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17]\nWe assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17]\n[SUBTITLE] 2.6.5. Sensitivity analysis. [SUBSECTION] The inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB.\nWe used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes.\nThe inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB.\nWe used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes.", "Transitivity is the fundamental assumption of indirect comparisons and NMA. We investigated the distribution of potential effect modifiers to assess the transitivity assumption. Potential effect modifiers included multicenter study, duration of study (DS), blinding, crossover design, sample size (SS), industry sponsorship, and risk of reported bias (RRB).", "We used STATA statistical software (Version 15, Stata Corporation, and College Station, Texas, USA) to conduct NMAs. We analyzed binary variables (ACM and TEAEs) using odds ratio (OR) with 95% confidence interval (CI). The appendix reported additional details (Supplemental Appendix 3, http://links.lww.com/MD/H535). We defined statistical significance as the P value < .05.\nWe merged simultaneously the direct evidence and indirect evidence or different indirect evidence through an NMA. In NMA, we analyzed the effect of intervention using group-level data. A fixed-effect model (i.e., I2 ≤ 50%) or a random-effect model (i.e., I2 > 50%) was used to summarize the effect sizes of NMA. We ranked the therapeutic effect for each outcome using the surface under the cumulative ranking area (SUCRA) curve and mean ranks.[16] In addition, the endpoints which lower was better would show rank 1 was the best and rank N was the worst based on figures and vice versa.", "We used the node splitting method (i.e., split evidence on a specific comparison into direct and indirect evidence) to estimate the inconsistency of NMAs.[17] There was no significant inconsistency in outcomes if P > .05.", "We assessed the risk of bias of included studies using the Cochrane Collaboration’s Tool for Assessing Risk of Bias,[12] classifying the risk of bias as high, unclear, or low. We evaluated the small-study effect using comparison adjusted funnel plots.[17]", "The inclusion of various study designs and populations might contribute to heterogeneity and inconsistency. We used sensitivity analysis to evaluate the effect of our conclusions. We analyzed the data using the following restrictions: multicenter study, DS, blinding, crossover design, SS, industry sponsorship, and RRB.\nWe used the “netmeta” package in R (version 4.0.5) to duplicate the NMAs of primary outcomes.", "[SUBTITLE] 3.1. Description of included studies [SUBSECTION] The search identified 4021 citations, out of which 3670 were excluded for duplications, wrong study design or population (i.e., non-severe COVID-19 infection), inappropriate intervention, not outcome/drug of interest, non-clinical studies, non-RCTs, review articles, commentaries, guidelines, and meta-analysis by checking titles and abstracts. The full texts of 351 articles were obtained to check eligibility, in which we excluded 308 articles for non- fulfilling eligibility criteria, unable to check eligibility and duplications. Finally, 43 studies[10,18–59] were included in our NMA. Figure 1 shows the selection process for the included studies.\nPRISMA flow-chart for study selection.\nForty-eight RCTs, including 9147 patients, were included (Fig. 1) and described in Table 1. This analysis had a mean sample size of 106 [interquartile range 58–238]. The age of subjects was greater than or equal to 16 years. The median duration of follow-up treatment was 28 days (interquartile range 27–30).\nThe search identified 4021 citations, out of which 3670 were excluded for duplications, wrong study design or population (i.e., non-severe COVID-19 infection), inappropriate intervention, not outcome/drug of interest, non-clinical studies, non-RCTs, review articles, commentaries, guidelines, and meta-analysis by checking titles and abstracts. The full texts of 351 articles were obtained to check eligibility, in which we excluded 308 articles for non- fulfilling eligibility criteria, unable to check eligibility and duplications. Finally, 43 studies[10,18–59] were included in our NMA. Figure 1 shows the selection process for the included studies.\nPRISMA flow-chart for study selection.\nForty-eight RCTs, including 9147 patients, were included (Fig. 1) and described in Table 1. This analysis had a mean sample size of 106 [interquartile range 58–238]. The age of subjects was greater than or equal to 16 years. The median duration of follow-up treatment was 28 days (interquartile range 27–30).\n[SUBTITLE] 3.2. Quality appraisal [SUBSECTION] The included RCTs were of good quality (Fig. 2). Figure 2 also showed the risk of bias summary.\nThe quality of the included randomized controlled trials. (A) Risk of bias summary (Note: The yellow circle with question mark represents “unclear risk of bias”, the red one with minus sign represents “high risk of bias” and the green one with plus sign represents “low risk of bias”). (B) Risk of bias graph.\nThe included RCTs were of good quality (Fig. 2). Figure 2 also showed the risk of bias summary.\nThe quality of the included randomized controlled trials. (A) Risk of bias summary (Note: The yellow circle with question mark represents “unclear risk of bias”, the red one with minus sign represents “high risk of bias” and the green one with plus sign represents “low risk of bias”). (B) Risk of bias graph.\n[SUBTITLE] 3.3. Network of evidence [SUBSECTION] In the network of connected RCTs (Fig. 3), the width of the lines corresponded to the number of trials included each treatment comparison. From Figure 3, we could see that the result was well connected. As shown in Figure 3A, the standard of care (SOC) was the most well-connected treatment, with chloroquine (CQ), hydroxychloroquine (HCQ), CQ/HCQ, convalescent plasma (CP), CP/SOC, remdesivir, remdesivir/SOC, lopinavir/ritonavir (LPV/r), interferon-beta-1b (IFN-β-1b), C-IVIG (i.e., a hyperimmune anti-COVID-19 intravenous immunoglobulin), ivermectin/doxycycline, IFN-β/SOC, tocilizumab, ruxolitinib/SOC, methylprednisolone, azithromycin/SOC, and auxora directly connected to it. Sarilumab such as high dosage sarilumab (HS) and low dosage sarilumab (LS), CQ/HCQ, ivermectin, canakinumab, colchicine, baricitinib, immunoglobulin gamma (IG), α-Lipoic acid, imatinib, mavrilimumab, lenzilumab, mycobacterium-w, otilimab, N-acetylcysteine, tocilizumab, high-dose intravenous vitamin C, hydrocortisone, and CP were directly connected to placebo in this network plot. Several sources of indirect evidence were available to inform comparisons between ivermectin, tocilizumab, HCQ, CP, auxora, placebo, and SOC (Fig. 3A). In Figure 3B, there was also a direct connection between SOC and C-IVIG, CP, LPV/r, ruxolitinib/SOC, ivermectin, IFN-β/SOC, and tocilizumab, or between placebo and HS, LS, mycobacterium-w, mavrilimumab, colchicine, canakinumab, baricitinib, human umbilical cord-derived mesenchymal stem cells, HCQ, lenzilumab, CP or tocilizumab. Several indirect connections were available between SOC and CP, tocilizumab, placebo (Fig. 3B).\nNetwork plot of eligible comparisons for all-cause mortality (A), and the ratio of treatment-emergent adverse events (B) for medications in patients with severe COVID-19. ALA= α-Lipoic acid, C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, IFN-β= interferon-beta, IG = immunoglobulin gamma, HDIVC = high-dose intravenous vitamin C, HS = high dosage sarilumab, LS = low dosage sarilumab, SOC = standard-of-care, UC-MSCs = human umbilical cord-derived mesenchymal stem cells.\nIn the network of connected RCTs (Fig. 3), the width of the lines corresponded to the number of trials included each treatment comparison. From Figure 3, we could see that the result was well connected. As shown in Figure 3A, the standard of care (SOC) was the most well-connected treatment, with chloroquine (CQ), hydroxychloroquine (HCQ), CQ/HCQ, convalescent plasma (CP), CP/SOC, remdesivir, remdesivir/SOC, lopinavir/ritonavir (LPV/r), interferon-beta-1b (IFN-β-1b), C-IVIG (i.e., a hyperimmune anti-COVID-19 intravenous immunoglobulin), ivermectin/doxycycline, IFN-β/SOC, tocilizumab, ruxolitinib/SOC, methylprednisolone, azithromycin/SOC, and auxora directly connected to it. Sarilumab such as high dosage sarilumab (HS) and low dosage sarilumab (LS), CQ/HCQ, ivermectin, canakinumab, colchicine, baricitinib, immunoglobulin gamma (IG), α-Lipoic acid, imatinib, mavrilimumab, lenzilumab, mycobacterium-w, otilimab, N-acetylcysteine, tocilizumab, high-dose intravenous vitamin C, hydrocortisone, and CP were directly connected to placebo in this network plot. Several sources of indirect evidence were available to inform comparisons between ivermectin, tocilizumab, HCQ, CP, auxora, placebo, and SOC (Fig. 3A). In Figure 3B, there was also a direct connection between SOC and C-IVIG, CP, LPV/r, ruxolitinib/SOC, ivermectin, IFN-β/SOC, and tocilizumab, or between placebo and HS, LS, mycobacterium-w, mavrilimumab, colchicine, canakinumab, baricitinib, human umbilical cord-derived mesenchymal stem cells, HCQ, lenzilumab, CP or tocilizumab. Several indirect connections were available between SOC and CP, tocilizumab, placebo (Fig. 3B).\nNetwork plot of eligible comparisons for all-cause mortality (A), and the ratio of treatment-emergent adverse events (B) for medications in patients with severe COVID-19. ALA= α-Lipoic acid, C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, IFN-β= interferon-beta, IG = immunoglobulin gamma, HDIVC = high-dose intravenous vitamin C, HS = high dosage sarilumab, LS = low dosage sarilumab, SOC = standard-of-care, UC-MSCs = human umbilical cord-derived mesenchymal stem cells.\n[SUBTITLE] 3.4. Efficacy outcomes [SUBSECTION] Forty-one studies[10,18–38,40–51,53–59] reported ACM as outcome measurement (Supplemental Table S1, http://links.lww.com/MD/H536). We found that C-IVIG (OR 0.22, 95% CI 0.05-0.95), methylprednisolone (OR 0.27, 95% CI 0.09-0.77), IFN-β/SOC (OR 0.30, 95% CI 0.11-0.83), CP (OR 0.49, 95% CI 0.26-0.94), remdesivir (OR 0.58, 95% CI 0.37-0.93), and HS (OR 0.45, 95% CI 0.20-0.99) were associated with the decrease of ACM when compared with the SOC group (Fig. 4). Unfortunately, there was no significant difference in other 27 medications (e.g., ivermectin/doxycycline, IFN-β-1b, LPV/r, IG, LPV/r, and imatinib) or placebo for ACM when compared with SOC.\nNetwork meta-analyses of the relative efficacy and safety of medications for all-cause mortality among patients with severe COVID-19 infection. The red font and asterisk indicated comparisons that were statistically significant. ALA= α-Lipoic acid, C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, CI = confidence interval, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, HDIVC = high-dose intravenous vitamin C, HS = high dosage sarilumab, IFN-β=interferon-beta, IG = immunoglobulin gamma, LS = low dosage sarilumab, OR = odds ratio, SOC = standard-of-care.\nFor a decrease in ACM, ivermectin/doxycycline (OR 0.06, 95% CI 0.00-0.88), C-IVIG (OR 0.12, 95% CI 0.02-0.61), methylprednisolone (OR 0.15, 95% CI 0.04-0.52), IG (OR 0.27, 95% CI 0.08-0.85), IFN-β/SOC (OR 0.17, 95% CI 0.05-0.56), IFN-β-1b (OR 0.16, 95% CI 0.03-0.98), auxora (OR 0.37, 95% CI 0.18-0.78), imatinib (OR 0.49, 95% CI 0.25-0.96), CP (OR 0.27, 95% CI 0.11-0.64), remdesivir (OR 0.32, 95% CI 0.14-0.74), and LPV/r (OR 0.32, 95% CI 0.12-0.85) were effective when compared with the placebo group (Fig. 4). We did not identify that there was a difference between placebo and other 22 medications (e.g., ruxolitinib/SOC, ivermectin, tocilizumab, HS, and mavrilimumab) or SOC for the ACM of severe COVID-19 infection (Fig. 4).\nThe Supplemental Figure S1, http://links.lww.com/MD/H538 presented the ranking for the ACM of medications in severe COVID-19 patients based on cumulative probability plots and SUCRA.\nForty-one studies[10,18–38,40–51,53–59] reported ACM as outcome measurement (Supplemental Table S1, http://links.lww.com/MD/H536). We found that C-IVIG (OR 0.22, 95% CI 0.05-0.95), methylprednisolone (OR 0.27, 95% CI 0.09-0.77), IFN-β/SOC (OR 0.30, 95% CI 0.11-0.83), CP (OR 0.49, 95% CI 0.26-0.94), remdesivir (OR 0.58, 95% CI 0.37-0.93), and HS (OR 0.45, 95% CI 0.20-0.99) were associated with the decrease of ACM when compared with the SOC group (Fig. 4). Unfortunately, there was no significant difference in other 27 medications (e.g., ivermectin/doxycycline, IFN-β-1b, LPV/r, IG, LPV/r, and imatinib) or placebo for ACM when compared with SOC.\nNetwork meta-analyses of the relative efficacy and safety of medications for all-cause mortality among patients with severe COVID-19 infection. The red font and asterisk indicated comparisons that were statistically significant. ALA= α-Lipoic acid, C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, CI = confidence interval, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, HDIVC = high-dose intravenous vitamin C, HS = high dosage sarilumab, IFN-β=interferon-beta, IG = immunoglobulin gamma, LS = low dosage sarilumab, OR = odds ratio, SOC = standard-of-care.\nFor a decrease in ACM, ivermectin/doxycycline (OR 0.06, 95% CI 0.00-0.88), C-IVIG (OR 0.12, 95% CI 0.02-0.61), methylprednisolone (OR 0.15, 95% CI 0.04-0.52), IG (OR 0.27, 95% CI 0.08-0.85), IFN-β/SOC (OR 0.17, 95% CI 0.05-0.56), IFN-β-1b (OR 0.16, 95% CI 0.03-0.98), auxora (OR 0.37, 95% CI 0.18-0.78), imatinib (OR 0.49, 95% CI 0.25-0.96), CP (OR 0.27, 95% CI 0.11-0.64), remdesivir (OR 0.32, 95% CI 0.14-0.74), and LPV/r (OR 0.32, 95% CI 0.12-0.85) were effective when compared with the placebo group (Fig. 4). We did not identify that there was a difference between placebo and other 22 medications (e.g., ruxolitinib/SOC, ivermectin, tocilizumab, HS, and mavrilimumab) or SOC for the ACM of severe COVID-19 infection (Fig. 4).\nThe Supplemental Figure S1, http://links.lww.com/MD/H538 presented the ranking for the ACM of medications in severe COVID-19 patients based on cumulative probability plots and SUCRA.\n[SUBTITLE] 3.5. Safety outcomes [SUBSECTION] In the safety outcome, data from 19 studies[18,20,21,23,26–29,34,38–40,48,50–54,56]were merged for analysis (Supplemental Table S2, http://links.lww.com/MD/H537). We found that colchicine (OR 2.77, 95% CI 1.03-7.42) and IFN-β/SOC (OR 18.98, 95% CI 1.70-211.84) seemed to increase the risk of TEAEs when compared with the SOC group. Moreover, IFN-β/SOC (OR 19.00, 95% CI 2.36-153.10) were associated with the increase of TEAEs when compared with placebo. We found no significant difference in TEAEs in other 15 medications for severe COVID-19 patients when compared with SOC or placebo (Fig. 5).\nNetwork meta-analyses of the relative efficacy and safety of medications for the ratio of treatment-emergent adverse events among patients with severe COVID-19 infection. The red font and asterisk indicated comparisons that were statistically significant. C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, CI = confidence interval, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, HS = high dosage sarilumab, IFN-β=interferon-beta, LS = low dosage sarilumab, OR = odds ratio, SOC = standard-of-care, UC-MSCs = human umbilical cord-derived mesenchymal stem cells.\nThe figure of SUCRA showed that baricitinib had the highest cumulative probability (SUCRA: 88.3%) becoming the best intervention in TEAEs, followed by LPV/r (SUCRA: 78.8%), SOC (SUCRA: 76.0%), lenzilumab (SUCRA: 72.6%), C-IVIG (SUCRA: 68.5%), tocilizumab (SUCRA: 63.6%), placebo (SUCRA: 53.1%), ivermectin (SUCRA: 52.3%), canakinumab (SUCRA: 51.9%), mycobacterium-w (SUCRA: 50.1%), LS (SUCRA: 49.3%), mavrilimumab (SUCRA: 47.1%), ruxolitinib/SOC (SUCRA: 46.8%), CP (SUCRA: 44.5%), HS (SUCRA: 31.0%), HCQ (SUCRA: 29.5%), human umbilical cord-derived mesenchymal stem cells (SUCRA: 29.1%), colchicine (SUCRA: 14.2%), and IFN-β/SOC (SUCRA: 3.2%) (Supplemental Figure S2, http://links.lww.com/MD/H539).\nIn the safety outcome, data from 19 studies[18,20,21,23,26–29,34,38–40,48,50–54,56]were merged for analysis (Supplemental Table S2, http://links.lww.com/MD/H537). We found that colchicine (OR 2.77, 95% CI 1.03-7.42) and IFN-β/SOC (OR 18.98, 95% CI 1.70-211.84) seemed to increase the risk of TEAEs when compared with the SOC group. Moreover, IFN-β/SOC (OR 19.00, 95% CI 2.36-153.10) were associated with the increase of TEAEs when compared with placebo. We found no significant difference in TEAEs in other 15 medications for severe COVID-19 patients when compared with SOC or placebo (Fig. 5).\nNetwork meta-analyses of the relative efficacy and safety of medications for the ratio of treatment-emergent adverse events among patients with severe COVID-19 infection. The red font and asterisk indicated comparisons that were statistically significant. C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, CI = confidence interval, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, HS = high dosage sarilumab, IFN-β=interferon-beta, LS = low dosage sarilumab, OR = odds ratio, SOC = standard-of-care, UC-MSCs = human umbilical cord-derived mesenchymal stem cells.\nThe figure of SUCRA showed that baricitinib had the highest cumulative probability (SUCRA: 88.3%) becoming the best intervention in TEAEs, followed by LPV/r (SUCRA: 78.8%), SOC (SUCRA: 76.0%), lenzilumab (SUCRA: 72.6%), C-IVIG (SUCRA: 68.5%), tocilizumab (SUCRA: 63.6%), placebo (SUCRA: 53.1%), ivermectin (SUCRA: 52.3%), canakinumab (SUCRA: 51.9%), mycobacterium-w (SUCRA: 50.1%), LS (SUCRA: 49.3%), mavrilimumab (SUCRA: 47.1%), ruxolitinib/SOC (SUCRA: 46.8%), CP (SUCRA: 44.5%), HS (SUCRA: 31.0%), HCQ (SUCRA: 29.5%), human umbilical cord-derived mesenchymal stem cells (SUCRA: 29.1%), colchicine (SUCRA: 14.2%), and IFN-β/SOC (SUCRA: 3.2%) (Supplemental Figure S2, http://links.lww.com/MD/H539).\n[SUBTITLE] 3.6. Evaluation of inconsistency [SUBSECTION] According to the inconsistency test (Table 2), no significant inconsistency or qualitative difference was available for the ACM and TEAEs. Thus, the consistency hypothesis was accepted in this NMA.\nThe evaluation of inconsistency for the efficacy and safety of medications among severe COVID-19 patients.\nCOVID-19 = coronavirus disease 2019.\nAccording to the inconsistency test (Table 2), no significant inconsistency or qualitative difference was available for the ACM and TEAEs. Thus, the consistency hypothesis was accepted in this NMA.\nThe evaluation of inconsistency for the efficacy and safety of medications among severe COVID-19 patients.\nCOVID-19 = coronavirus disease 2019.\n[SUBTITLE] 3.7. Sensitivity analysis and publication bias [SUBSECTION] We analyzed the potential sources of heterogeneity or inconsistency by using subgroup and meta-regression analyses. Univariable meta-regression and subgroup analyses indicated that there were heterogeneous sources (such as DS, blinding and RRB) for the ACM (P < .05) (Fig. 6A). Whilst the SS and RRB were the heterogeneity source of TEAEs based on the sensitivity analysis (P < .05) (Fig. 6B).\nMeta-regression and sensitivity analyses for the efficacy and safety of medications in patients with severe COVID-19 infection. (A) all-cause mortality. (B) the ratio of treatment-emergent adverse events. CD = crossover design, COVID-19 = coronavirus disease 2019, DS = duration of study, IS = industry sponsorship, MS = multicenter study, RRB = risk of reported bias, SS = sample size.\nNone of the funnel plots of outcomes (ACM and TEAEs) indicated a significant asymmetry (Supplemental Figure S3, http://links.lww.com/MD/H540).\nWe analyzed the potential sources of heterogeneity or inconsistency by using subgroup and meta-regression analyses. Univariable meta-regression and subgroup analyses indicated that there were heterogeneous sources (such as DS, blinding and RRB) for the ACM (P < .05) (Fig. 6A). Whilst the SS and RRB were the heterogeneity source of TEAEs based on the sensitivity analysis (P < .05) (Fig. 6B).\nMeta-regression and sensitivity analyses for the efficacy and safety of medications in patients with severe COVID-19 infection. (A) all-cause mortality. (B) the ratio of treatment-emergent adverse events. CD = crossover design, COVID-19 = coronavirus disease 2019, DS = duration of study, IS = industry sponsorship, MS = multicenter study, RRB = risk of reported bias, SS = sample size.\nNone of the funnel plots of outcomes (ACM and TEAEs) indicated a significant asymmetry (Supplemental Figure S3, http://links.lww.com/MD/H540).", "The search identified 4021 citations, out of which 3670 were excluded for duplications, wrong study design or population (i.e., non-severe COVID-19 infection), inappropriate intervention, not outcome/drug of interest, non-clinical studies, non-RCTs, review articles, commentaries, guidelines, and meta-analysis by checking titles and abstracts. The full texts of 351 articles were obtained to check eligibility, in which we excluded 308 articles for non- fulfilling eligibility criteria, unable to check eligibility and duplications. Finally, 43 studies[10,18–59] were included in our NMA. Figure 1 shows the selection process for the included studies.\nPRISMA flow-chart for study selection.\nForty-eight RCTs, including 9147 patients, were included (Fig. 1) and described in Table 1. This analysis had a mean sample size of 106 [interquartile range 58–238]. The age of subjects was greater than or equal to 16 years. The median duration of follow-up treatment was 28 days (interquartile range 27–30).", "The included RCTs were of good quality (Fig. 2). Figure 2 also showed the risk of bias summary.\nThe quality of the included randomized controlled trials. (A) Risk of bias summary (Note: The yellow circle with question mark represents “unclear risk of bias”, the red one with minus sign represents “high risk of bias” and the green one with plus sign represents “low risk of bias”). (B) Risk of bias graph.", "In the network of connected RCTs (Fig. 3), the width of the lines corresponded to the number of trials included each treatment comparison. From Figure 3, we could see that the result was well connected. As shown in Figure 3A, the standard of care (SOC) was the most well-connected treatment, with chloroquine (CQ), hydroxychloroquine (HCQ), CQ/HCQ, convalescent plasma (CP), CP/SOC, remdesivir, remdesivir/SOC, lopinavir/ritonavir (LPV/r), interferon-beta-1b (IFN-β-1b), C-IVIG (i.e., a hyperimmune anti-COVID-19 intravenous immunoglobulin), ivermectin/doxycycline, IFN-β/SOC, tocilizumab, ruxolitinib/SOC, methylprednisolone, azithromycin/SOC, and auxora directly connected to it. Sarilumab such as high dosage sarilumab (HS) and low dosage sarilumab (LS), CQ/HCQ, ivermectin, canakinumab, colchicine, baricitinib, immunoglobulin gamma (IG), α-Lipoic acid, imatinib, mavrilimumab, lenzilumab, mycobacterium-w, otilimab, N-acetylcysteine, tocilizumab, high-dose intravenous vitamin C, hydrocortisone, and CP were directly connected to placebo in this network plot. Several sources of indirect evidence were available to inform comparisons between ivermectin, tocilizumab, HCQ, CP, auxora, placebo, and SOC (Fig. 3A). In Figure 3B, there was also a direct connection between SOC and C-IVIG, CP, LPV/r, ruxolitinib/SOC, ivermectin, IFN-β/SOC, and tocilizumab, or between placebo and HS, LS, mycobacterium-w, mavrilimumab, colchicine, canakinumab, baricitinib, human umbilical cord-derived mesenchymal stem cells, HCQ, lenzilumab, CP or tocilizumab. Several indirect connections were available between SOC and CP, tocilizumab, placebo (Fig. 3B).\nNetwork plot of eligible comparisons for all-cause mortality (A), and the ratio of treatment-emergent adverse events (B) for medications in patients with severe COVID-19. ALA= α-Lipoic acid, C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, IFN-β= interferon-beta, IG = immunoglobulin gamma, HDIVC = high-dose intravenous vitamin C, HS = high dosage sarilumab, LS = low dosage sarilumab, SOC = standard-of-care, UC-MSCs = human umbilical cord-derived mesenchymal stem cells.", "Forty-one studies[10,18–38,40–51,53–59] reported ACM as outcome measurement (Supplemental Table S1, http://links.lww.com/MD/H536). We found that C-IVIG (OR 0.22, 95% CI 0.05-0.95), methylprednisolone (OR 0.27, 95% CI 0.09-0.77), IFN-β/SOC (OR 0.30, 95% CI 0.11-0.83), CP (OR 0.49, 95% CI 0.26-0.94), remdesivir (OR 0.58, 95% CI 0.37-0.93), and HS (OR 0.45, 95% CI 0.20-0.99) were associated with the decrease of ACM when compared with the SOC group (Fig. 4). Unfortunately, there was no significant difference in other 27 medications (e.g., ivermectin/doxycycline, IFN-β-1b, LPV/r, IG, LPV/r, and imatinib) or placebo for ACM when compared with SOC.\nNetwork meta-analyses of the relative efficacy and safety of medications for all-cause mortality among patients with severe COVID-19 infection. The red font and asterisk indicated comparisons that were statistically significant. ALA= α-Lipoic acid, C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, CI = confidence interval, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, HDIVC = high-dose intravenous vitamin C, HS = high dosage sarilumab, IFN-β=interferon-beta, IG = immunoglobulin gamma, LS = low dosage sarilumab, OR = odds ratio, SOC = standard-of-care.\nFor a decrease in ACM, ivermectin/doxycycline (OR 0.06, 95% CI 0.00-0.88), C-IVIG (OR 0.12, 95% CI 0.02-0.61), methylprednisolone (OR 0.15, 95% CI 0.04-0.52), IG (OR 0.27, 95% CI 0.08-0.85), IFN-β/SOC (OR 0.17, 95% CI 0.05-0.56), IFN-β-1b (OR 0.16, 95% CI 0.03-0.98), auxora (OR 0.37, 95% CI 0.18-0.78), imatinib (OR 0.49, 95% CI 0.25-0.96), CP (OR 0.27, 95% CI 0.11-0.64), remdesivir (OR 0.32, 95% CI 0.14-0.74), and LPV/r (OR 0.32, 95% CI 0.12-0.85) were effective when compared with the placebo group (Fig. 4). We did not identify that there was a difference between placebo and other 22 medications (e.g., ruxolitinib/SOC, ivermectin, tocilizumab, HS, and mavrilimumab) or SOC for the ACM of severe COVID-19 infection (Fig. 4).\nThe Supplemental Figure S1, http://links.lww.com/MD/H538 presented the ranking for the ACM of medications in severe COVID-19 patients based on cumulative probability plots and SUCRA.", "In the safety outcome, data from 19 studies[18,20,21,23,26–29,34,38–40,48,50–54,56]were merged for analysis (Supplemental Table S2, http://links.lww.com/MD/H537). We found that colchicine (OR 2.77, 95% CI 1.03-7.42) and IFN-β/SOC (OR 18.98, 95% CI 1.70-211.84) seemed to increase the risk of TEAEs when compared with the SOC group. Moreover, IFN-β/SOC (OR 19.00, 95% CI 2.36-153.10) were associated with the increase of TEAEs when compared with placebo. We found no significant difference in TEAEs in other 15 medications for severe COVID-19 patients when compared with SOC or placebo (Fig. 5).\nNetwork meta-analyses of the relative efficacy and safety of medications for the ratio of treatment-emergent adverse events among patients with severe COVID-19 infection. The red font and asterisk indicated comparisons that were statistically significant. C-IVIG = hyperimmune anti-COVID-19 intravenous immunoglobulin, CI = confidence interval, COVID-19 = coronavirus disease 2019, CP = convalescent plasma, HS = high dosage sarilumab, IFN-β=interferon-beta, LS = low dosage sarilumab, OR = odds ratio, SOC = standard-of-care, UC-MSCs = human umbilical cord-derived mesenchymal stem cells.\nThe figure of SUCRA showed that baricitinib had the highest cumulative probability (SUCRA: 88.3%) becoming the best intervention in TEAEs, followed by LPV/r (SUCRA: 78.8%), SOC (SUCRA: 76.0%), lenzilumab (SUCRA: 72.6%), C-IVIG (SUCRA: 68.5%), tocilizumab (SUCRA: 63.6%), placebo (SUCRA: 53.1%), ivermectin (SUCRA: 52.3%), canakinumab (SUCRA: 51.9%), mycobacterium-w (SUCRA: 50.1%), LS (SUCRA: 49.3%), mavrilimumab (SUCRA: 47.1%), ruxolitinib/SOC (SUCRA: 46.8%), CP (SUCRA: 44.5%), HS (SUCRA: 31.0%), HCQ (SUCRA: 29.5%), human umbilical cord-derived mesenchymal stem cells (SUCRA: 29.1%), colchicine (SUCRA: 14.2%), and IFN-β/SOC (SUCRA: 3.2%) (Supplemental Figure S2, http://links.lww.com/MD/H539).", "According to the inconsistency test (Table 2), no significant inconsistency or qualitative difference was available for the ACM and TEAEs. Thus, the consistency hypothesis was accepted in this NMA.\nThe evaluation of inconsistency for the efficacy and safety of medications among severe COVID-19 patients.\nCOVID-19 = coronavirus disease 2019.", "We analyzed the potential sources of heterogeneity or inconsistency by using subgroup and meta-regression analyses. Univariable meta-regression and subgroup analyses indicated that there were heterogeneous sources (such as DS, blinding and RRB) for the ACM (P < .05) (Fig. 6A). Whilst the SS and RRB were the heterogeneity source of TEAEs based on the sensitivity analysis (P < .05) (Fig. 6B).\nMeta-regression and sensitivity analyses for the efficacy and safety of medications in patients with severe COVID-19 infection. (A) all-cause mortality. (B) the ratio of treatment-emergent adverse events. CD = crossover design, COVID-19 = coronavirus disease 2019, DS = duration of study, IS = industry sponsorship, MS = multicenter study, RRB = risk of reported bias, SS = sample size.\nNone of the funnel plots of outcomes (ACM and TEAEs) indicated a significant asymmetry (Supplemental Figure S3, http://links.lww.com/MD/H540).", "This study was based on 48 RCTs, which included 9147 severe patients randomly assigned to 35 medications or SOC or placebo. Our NMA indicated that 12 medications (i.e., ivermectin/doxycycline, C-IVIG, methylprednisolone, IFN-β/SOC, IFN-β-1b, CP, remdesivir, LPV/r, HS, IG, auxora, and imatinib) were efficacious for the therapy of severe COVID-19 infection based on the controls of SOC or placebo. For a decrease in ACM, on the one hand, C-IVIG, methylprednisolone, IFN-β/SOC, HS, remdesivir, and CP were more efficacious than SOC; on the other hand, the efficacy of ivermectin/doxycycline, C-IVIG, methylprednisolone, IFN-β/SOC, IFN-β-1b, CP, remdesivir, LPV/r, IG, auxora, and imatinib were superior to placebo in all 35 drug interventions. From this, we can infer that there is a difference in the findings of medications for severe COVID-19 patients from different control conditions in RCTs.\nCuriously, our findings differed from our earlier pilot studies,[14,60] which have only indicated 2 effective medications (i.e., IG and methylprednisolone) in the decreased ACM for severe COVID-19 patients. In addition, the CP group showed lower TEAEs than the placebo for severe COVID-19 infection in our earlier NMAs.[14,60] These inconsistencies may be due to an increase in sample size in the updated NMA. The study extended the previous work that provided a reference for selecting the medication for patients with severe COVID-19 infection.[61] The present study raises the possibility that we cannot completely discount the efficacy of other medications for severe COVID-19 infection in clinic. So far, maybe it’s because there was inadequate evidence of a benefit in ACM for them. In either case, it is noteworthy that we must fully consider its efficacy and safety when the medications are used in treating severe COVID-19 patients. Some medications (e.g., tocilizumab, otilimab, and mavrilimumab) might be a selection of combination treatment for COVID-19 infection.[62,63]\nIt is particularly worth mentioning that host factors, including age, prior-medical history, demographics, and medical facility, may be key determinants of disease severity and progression.[64] Similarly, the above factors are associated with the drug treatment effect of COVID-19 infection. Several studies have reported that aging and prior-medical history (such as cancer or chronic kidney/liver/lung diseases) might be 2 prominent impact factors for medical therapy from COVID-19.[65,66] Kopel et al suggested that some demographics factors (such as gender, ethnic group and socioeconomic status) were associated with the therapy of COVID-19 infection.[67,68] In another study in India, Sarkar et al[69] reported that different strains of SARS-CoV-2 were associated with the efficacy of antiviral drugs. It has also been suggested that medical facilities may be an impact factor for the COVID-19 treatment.[70] We have considered these impact factors in the overall therapy for COVID-19 patients during this NMA design. To reduce the impact of age, we included all of studies based on patient age greater than or equal to 16 years. As is well known, the design principles of RCTs are randomization, control, blinding, and repetition.[71] According to the 48 RCTs data analysis of this NMA, we confirmed that age, prior-medical history, gender and racial ratios for participants were balanced between experimental and control groups (P > .05). In view of this, we suggest that the effects of age, prior-medical history, gender and racial ratios in the findings are small in this NMA study. This might present one of the main strengthens of this NMA. Unfortunately, the description of most studies was insufficient in the gender-specific, different strains and medical facilities for COVID-19 therapy in previous RCTs. We could not explore the efficacy of drugs treatment from the gender-specific or different strains as well as the impact from medical facilities for COVID-19 therapy.\n[SUBTITLE] 4.1. Efficacy of current medications [SUBSECTION] This study supported evidence from previous observations.[72,73] As we know, intravenous immunoglobulin has already been validated as an effective antiviral drug for treating COVID-19, SARS and Middle East respiratory syndrome.[20,74,75] This is consistent with our results (i.e., C-IVIG, HS, and IG). Recent evidence suggested that anti-cytokine effects, inhibition of complement activation, and down-regulation of B and T cells’ functions by C-IVIG can prevent organ failure and subsequent mortality in severe COVID-19 patients.[20] Our findings also confirmed that sarilumab could prevent patients with COVID-19 from progressing to death. Sarilumab is a fully human antibody against the interleukin (IL)-6 receptor, which can rapidly lower C-reactive protein and mediate COVID-19 clearance.[76] Prolonged glucocorticoid treatment is associated with improved outcomes of acute respiratory distress syndrome.[77] Several reports have also shown that treatment with methylprednisolone could significantly reduce the risk of death among patients with acute respiratory distress syndrome.[78] This also accords with our observations, which suggested that methylprednisolone was associated with decreased ACM in severe COVID-19 patients.\nThe SUCRA indicated that ivermectin/doxycycline was the highest ranked intervention with a SUCRA of 0.821. Previous studies showed that ivermectin or doxycycline is effective in treating COVID-19 patients.[79,80] We attribute this to a couple of reasons. First, ivermectin or doxycycline might possess antiviral as well as immunomodulatory activity.[79,80] Second, they might have anti-inflammatory and immunomodulatory agents and can curb over-reacting innate and cellular immune responses.[81] One possible implication may be that the ivermectin/doxycycline is an excellent selection for treating severe COVID-19 patients. Of note, based on the result of meta-regression analysis on the heterogeneity (such as DS, blinding, and RRB), the present result may need further verification. Thus, statistical indications of clinical superiority in this network analysis required careful interpretation.\nInterestingly, we found that small-molecule protein kinase inhibitors (i.e., auxora and imatinib) were efficacious for the therapy of severe COVID-19 infection. Recent research has established that a calcium release-activated calcium channel inhibitor, such as auxora, can reduce the occurrence of COVID-19 death due to blocking the release of multiple pro-inflammatory cytokines, including IL-6. Additionally, evidence showed that imatinib might play its potentially antiviral and beneficial immunomodulatory role in severe COVID-19 patients.[82] These results further support those of previous studies.[25,83] Collectively, the present findings indicate that the small-molecule protein kinase inhibitors provide a new, and perhaps superior, avenue for the severe COVID-19 treatment. In addition, our results may provide a robust strategy for clinical combination therapy among severe COVID-19 patients.\nWe have demonstrated that IFN-β/SOC, IFN-β-1b, remdesivir, LPV/r and CP were associated with a reduction of ACM in severe COVID-19 infection. Several lines of evidence suggested that IFN deficiency was a hallmark of severe COVID-19.[84] IFN might treat severe COVID-19 infection through adapted anti-inflammatory therapies that target IL-6 or TNF-β.[84,85] Data from prior studies suggested that remdesivir was a broad-spectrum antiviral activity against RNA viruses, which could improve the survival of COVID-19 infection due to inhibiting viral replication.[86] Recent studies reported that LPV/r displayed inhibitory activities against SARS-CoV-2 main protease and inhibited SARS-CoV-2 replication in Vero E6 cells.[87] Similarly to other antibodies therapy, CP can prevent the death of severe COVID-19 patients because of alleviating the inflammation and overreaction of the immune system through antibodies.[88] These findings further support the idea of our NMA study.\nConversely, no significant difference was found in other 21 medications (e.g., HCQ, α-Lipoic acid, hydrocortisone, otilimab, and mavrilimumab) for the ACM of severe COVID-19 infection when compared with SOC or placebo. This finding was contrary to previous studies which have suggested that some antiviral drugs (hydrocortisone, mavrilimumab, etc) were associated with decreased ACM in patients with COVID-19.[29,32,89,90] It is difficult to explain this result, but it might be related to the difference of participants’ selection in differential studies. For instance, the findings of this NMA were based on a larger sample size (i.e., more participants were included). Correspondingly, most of the previous studies presented a smaller sample size.[29,32] The present study differed from earlier studies,[90,91] which selected subjects with all infection levels (i.e., mild, moderate and severe infections). Furthermore, prior studies were inadequate for the analyses stratified by different infection levels (i.e., non-severe and severe infection) in medications of COVID-19.[29,32,89,90] Prior studies suggested that COVID-19 patients at different infection levels often led to different outcomes of treatment.[92] Another possible explanation for this was that we compared the efficacy and safety of SOC, which existed the bias due to the differential SOC of every country (i.e., the SOC is not standardized) except for the reasons given above.[21,23,45] The present study raised the possibility that our findings might be beneficial to guiding the selection of drug interventions for clinicians in severe COVID-19 patients.\nThis study supported evidence from previous observations.[72,73] As we know, intravenous immunoglobulin has already been validated as an effective antiviral drug for treating COVID-19, SARS and Middle East respiratory syndrome.[20,74,75] This is consistent with our results (i.e., C-IVIG, HS, and IG). Recent evidence suggested that anti-cytokine effects, inhibition of complement activation, and down-regulation of B and T cells’ functions by C-IVIG can prevent organ failure and subsequent mortality in severe COVID-19 patients.[20] Our findings also confirmed that sarilumab could prevent patients with COVID-19 from progressing to death. Sarilumab is a fully human antibody against the interleukin (IL)-6 receptor, which can rapidly lower C-reactive protein and mediate COVID-19 clearance.[76] Prolonged glucocorticoid treatment is associated with improved outcomes of acute respiratory distress syndrome.[77] Several reports have also shown that treatment with methylprednisolone could significantly reduce the risk of death among patients with acute respiratory distress syndrome.[78] This also accords with our observations, which suggested that methylprednisolone was associated with decreased ACM in severe COVID-19 patients.\nThe SUCRA indicated that ivermectin/doxycycline was the highest ranked intervention with a SUCRA of 0.821. Previous studies showed that ivermectin or doxycycline is effective in treating COVID-19 patients.[79,80] We attribute this to a couple of reasons. First, ivermectin or doxycycline might possess antiviral as well as immunomodulatory activity.[79,80] Second, they might have anti-inflammatory and immunomodulatory agents and can curb over-reacting innate and cellular immune responses.[81] One possible implication may be that the ivermectin/doxycycline is an excellent selection for treating severe COVID-19 patients. Of note, based on the result of meta-regression analysis on the heterogeneity (such as DS, blinding, and RRB), the present result may need further verification. Thus, statistical indications of clinical superiority in this network analysis required careful interpretation.\nInterestingly, we found that small-molecule protein kinase inhibitors (i.e., auxora and imatinib) were efficacious for the therapy of severe COVID-19 infection. Recent research has established that a calcium release-activated calcium channel inhibitor, such as auxora, can reduce the occurrence of COVID-19 death due to blocking the release of multiple pro-inflammatory cytokines, including IL-6. Additionally, evidence showed that imatinib might play its potentially antiviral and beneficial immunomodulatory role in severe COVID-19 patients.[82] These results further support those of previous studies.[25,83] Collectively, the present findings indicate that the small-molecule protein kinase inhibitors provide a new, and perhaps superior, avenue for the severe COVID-19 treatment. In addition, our results may provide a robust strategy for clinical combination therapy among severe COVID-19 patients.\nWe have demonstrated that IFN-β/SOC, IFN-β-1b, remdesivir, LPV/r and CP were associated with a reduction of ACM in severe COVID-19 infection. Several lines of evidence suggested that IFN deficiency was a hallmark of severe COVID-19.[84] IFN might treat severe COVID-19 infection through adapted anti-inflammatory therapies that target IL-6 or TNF-β.[84,85] Data from prior studies suggested that remdesivir was a broad-spectrum antiviral activity against RNA viruses, which could improve the survival of COVID-19 infection due to inhibiting viral replication.[86] Recent studies reported that LPV/r displayed inhibitory activities against SARS-CoV-2 main protease and inhibited SARS-CoV-2 replication in Vero E6 cells.[87] Similarly to other antibodies therapy, CP can prevent the death of severe COVID-19 patients because of alleviating the inflammation and overreaction of the immune system through antibodies.[88] These findings further support the idea of our NMA study.\nConversely, no significant difference was found in other 21 medications (e.g., HCQ, α-Lipoic acid, hydrocortisone, otilimab, and mavrilimumab) for the ACM of severe COVID-19 infection when compared with SOC or placebo. This finding was contrary to previous studies which have suggested that some antiviral drugs (hydrocortisone, mavrilimumab, etc) were associated with decreased ACM in patients with COVID-19.[29,32,89,90] It is difficult to explain this result, but it might be related to the difference of participants’ selection in differential studies. For instance, the findings of this NMA were based on a larger sample size (i.e., more participants were included). Correspondingly, most of the previous studies presented a smaller sample size.[29,32] The present study differed from earlier studies,[90,91] which selected subjects with all infection levels (i.e., mild, moderate and severe infections). Furthermore, prior studies were inadequate for the analyses stratified by different infection levels (i.e., non-severe and severe infection) in medications of COVID-19.[29,32,89,90] Prior studies suggested that COVID-19 patients at different infection levels often led to different outcomes of treatment.[92] Another possible explanation for this was that we compared the efficacy and safety of SOC, which existed the bias due to the differential SOC of every country (i.e., the SOC is not standardized) except for the reasons given above.[21,23,45] The present study raised the possibility that our findings might be beneficial to guiding the selection of drug interventions for clinicians in severe COVID-19 patients.\n[SUBTITLE] 4.2. Safety of current medications [SUBSECTION] In terms of safety, we summarized the TEAEs. We found that colchicine and IFN-β/SOC were only associated with the TEAEs of severe COVID-19 patients in this study. Recent meta-analysis studies and large-scale RCTs[45,46,92] seemed to be consistent with our findings, which identified most pharmacological treatments had a good safety in treating severe COVID-19. However, as mentioned in the present study, colchicine and IFN-β/SOC should be chosen cautiously in treating severe COVID-19 patients based on safety. Colchicine might increase the TEAEs in treating patients with severe COVID-19 infection. It is difficult to explain this result, but it might be related to the toxicity (e.g., gastrointestinal mucosal damage) in the case of colchicine treatment.[93] In addition, it is possible that the use of IFN-β in combination with SOC was associated with increased TEAEs in treating severe COVID-19, which should be weighed in all future IFN-β studies.[94]\nIn summary, clinicians might need to select treatment regimens based on the ranks of efficacy and safety (i.e., SUCRA) when medications are used in treating severe COVID-19 patients. Additionally, it should be reminded that further studies, which reduce the effects of SS and RRB, will need to be undertaken based on the result of sensitivity analysis.\nSome limitations constrained this study. Firstly, included studies might be small in this NMA, which should be considered when interpreting the findings. Secondly, the published data we extracted included only 2 types of outcomes, some important outcomes such as discharge ratio, clinical improvement ratio, and the ratio of virological cure, were not analyzed. Thirdly, although we did our best to include all available RCTs, we cannot eliminate the possibility of missing data. Fourthly, some nodes in our network included only a few trials. The sample size of actual head-to-head trials was very small. Hence, comparative efficacy and safety between interventions was frequently based on indirect comparisons. Finally, the sensitivity analysis showed that there were several heterogeneity sources, which may conceal or exaggerate the effect size of this network analysis. Further large-scale RCTs, which control these confounding factors, will need to be undertaken to verify our findings. Though there are still many shortcomings in our research, it is certain that the prevention and therapy of COVID-19 is set to change for the better in the future.\nIn terms of safety, we summarized the TEAEs. We found that colchicine and IFN-β/SOC were only associated with the TEAEs of severe COVID-19 patients in this study. Recent meta-analysis studies and large-scale RCTs[45,46,92] seemed to be consistent with our findings, which identified most pharmacological treatments had a good safety in treating severe COVID-19. However, as mentioned in the present study, colchicine and IFN-β/SOC should be chosen cautiously in treating severe COVID-19 patients based on safety. Colchicine might increase the TEAEs in treating patients with severe COVID-19 infection. It is difficult to explain this result, but it might be related to the toxicity (e.g., gastrointestinal mucosal damage) in the case of colchicine treatment.[93] In addition, it is possible that the use of IFN-β in combination with SOC was associated with increased TEAEs in treating severe COVID-19, which should be weighed in all future IFN-β studies.[94]\nIn summary, clinicians might need to select treatment regimens based on the ranks of efficacy and safety (i.e., SUCRA) when medications are used in treating severe COVID-19 patients. Additionally, it should be reminded that further studies, which reduce the effects of SS and RRB, will need to be undertaken based on the result of sensitivity analysis.\nSome limitations constrained this study. Firstly, included studies might be small in this NMA, which should be considered when interpreting the findings. Secondly, the published data we extracted included only 2 types of outcomes, some important outcomes such as discharge ratio, clinical improvement ratio, and the ratio of virological cure, were not analyzed. Thirdly, although we did our best to include all available RCTs, we cannot eliminate the possibility of missing data. Fourthly, some nodes in our network included only a few trials. The sample size of actual head-to-head trials was very small. Hence, comparative efficacy and safety between interventions was frequently based on indirect comparisons. Finally, the sensitivity analysis showed that there were several heterogeneity sources, which may conceal or exaggerate the effect size of this network analysis. Further large-scale RCTs, which control these confounding factors, will need to be undertaken to verify our findings. Though there are still many shortcomings in our research, it is certain that the prevention and therapy of COVID-19 is set to change for the better in the future.", "This study supported evidence from previous observations.[72,73] As we know, intravenous immunoglobulin has already been validated as an effective antiviral drug for treating COVID-19, SARS and Middle East respiratory syndrome.[20,74,75] This is consistent with our results (i.e., C-IVIG, HS, and IG). Recent evidence suggested that anti-cytokine effects, inhibition of complement activation, and down-regulation of B and T cells’ functions by C-IVIG can prevent organ failure and subsequent mortality in severe COVID-19 patients.[20] Our findings also confirmed that sarilumab could prevent patients with COVID-19 from progressing to death. Sarilumab is a fully human antibody against the interleukin (IL)-6 receptor, which can rapidly lower C-reactive protein and mediate COVID-19 clearance.[76] Prolonged glucocorticoid treatment is associated with improved outcomes of acute respiratory distress syndrome.[77] Several reports have also shown that treatment with methylprednisolone could significantly reduce the risk of death among patients with acute respiratory distress syndrome.[78] This also accords with our observations, which suggested that methylprednisolone was associated with decreased ACM in severe COVID-19 patients.\nThe SUCRA indicated that ivermectin/doxycycline was the highest ranked intervention with a SUCRA of 0.821. Previous studies showed that ivermectin or doxycycline is effective in treating COVID-19 patients.[79,80] We attribute this to a couple of reasons. First, ivermectin or doxycycline might possess antiviral as well as immunomodulatory activity.[79,80] Second, they might have anti-inflammatory and immunomodulatory agents and can curb over-reacting innate and cellular immune responses.[81] One possible implication may be that the ivermectin/doxycycline is an excellent selection for treating severe COVID-19 patients. Of note, based on the result of meta-regression analysis on the heterogeneity (such as DS, blinding, and RRB), the present result may need further verification. Thus, statistical indications of clinical superiority in this network analysis required careful interpretation.\nInterestingly, we found that small-molecule protein kinase inhibitors (i.e., auxora and imatinib) were efficacious for the therapy of severe COVID-19 infection. Recent research has established that a calcium release-activated calcium channel inhibitor, such as auxora, can reduce the occurrence of COVID-19 death due to blocking the release of multiple pro-inflammatory cytokines, including IL-6. Additionally, evidence showed that imatinib might play its potentially antiviral and beneficial immunomodulatory role in severe COVID-19 patients.[82] These results further support those of previous studies.[25,83] Collectively, the present findings indicate that the small-molecule protein kinase inhibitors provide a new, and perhaps superior, avenue for the severe COVID-19 treatment. In addition, our results may provide a robust strategy for clinical combination therapy among severe COVID-19 patients.\nWe have demonstrated that IFN-β/SOC, IFN-β-1b, remdesivir, LPV/r and CP were associated with a reduction of ACM in severe COVID-19 infection. Several lines of evidence suggested that IFN deficiency was a hallmark of severe COVID-19.[84] IFN might treat severe COVID-19 infection through adapted anti-inflammatory therapies that target IL-6 or TNF-β.[84,85] Data from prior studies suggested that remdesivir was a broad-spectrum antiviral activity against RNA viruses, which could improve the survival of COVID-19 infection due to inhibiting viral replication.[86] Recent studies reported that LPV/r displayed inhibitory activities against SARS-CoV-2 main protease and inhibited SARS-CoV-2 replication in Vero E6 cells.[87] Similarly to other antibodies therapy, CP can prevent the death of severe COVID-19 patients because of alleviating the inflammation and overreaction of the immune system through antibodies.[88] These findings further support the idea of our NMA study.\nConversely, no significant difference was found in other 21 medications (e.g., HCQ, α-Lipoic acid, hydrocortisone, otilimab, and mavrilimumab) for the ACM of severe COVID-19 infection when compared with SOC or placebo. This finding was contrary to previous studies which have suggested that some antiviral drugs (hydrocortisone, mavrilimumab, etc) were associated with decreased ACM in patients with COVID-19.[29,32,89,90] It is difficult to explain this result, but it might be related to the difference of participants’ selection in differential studies. For instance, the findings of this NMA were based on a larger sample size (i.e., more participants were included). Correspondingly, most of the previous studies presented a smaller sample size.[29,32] The present study differed from earlier studies,[90,91] which selected subjects with all infection levels (i.e., mild, moderate and severe infections). Furthermore, prior studies were inadequate for the analyses stratified by different infection levels (i.e., non-severe and severe infection) in medications of COVID-19.[29,32,89,90] Prior studies suggested that COVID-19 patients at different infection levels often led to different outcomes of treatment.[92] Another possible explanation for this was that we compared the efficacy and safety of SOC, which existed the bias due to the differential SOC of every country (i.e., the SOC is not standardized) except for the reasons given above.[21,23,45] The present study raised the possibility that our findings might be beneficial to guiding the selection of drug interventions for clinicians in severe COVID-19 patients.", "In terms of safety, we summarized the TEAEs. We found that colchicine and IFN-β/SOC were only associated with the TEAEs of severe COVID-19 patients in this study. Recent meta-analysis studies and large-scale RCTs[45,46,92] seemed to be consistent with our findings, which identified most pharmacological treatments had a good safety in treating severe COVID-19. However, as mentioned in the present study, colchicine and IFN-β/SOC should be chosen cautiously in treating severe COVID-19 patients based on safety. Colchicine might increase the TEAEs in treating patients with severe COVID-19 infection. It is difficult to explain this result, but it might be related to the toxicity (e.g., gastrointestinal mucosal damage) in the case of colchicine treatment.[93] In addition, it is possible that the use of IFN-β in combination with SOC was associated with increased TEAEs in treating severe COVID-19, which should be weighed in all future IFN-β studies.[94]\nIn summary, clinicians might need to select treatment regimens based on the ranks of efficacy and safety (i.e., SUCRA) when medications are used in treating severe COVID-19 patients. Additionally, it should be reminded that further studies, which reduce the effects of SS and RRB, will need to be undertaken based on the result of sensitivity analysis.\nSome limitations constrained this study. Firstly, included studies might be small in this NMA, which should be considered when interpreting the findings. Secondly, the published data we extracted included only 2 types of outcomes, some important outcomes such as discharge ratio, clinical improvement ratio, and the ratio of virological cure, were not analyzed. Thirdly, although we did our best to include all available RCTs, we cannot eliminate the possibility of missing data. Fourthly, some nodes in our network included only a few trials. The sample size of actual head-to-head trials was very small. Hence, comparative efficacy and safety between interventions was frequently based on indirect comparisons. Finally, the sensitivity analysis showed that there were several heterogeneity sources, which may conceal or exaggerate the effect size of this network analysis. Further large-scale RCTs, which control these confounding factors, will need to be undertaken to verify our findings. Though there are still many shortcomings in our research, it is certain that the prevention and therapy of COVID-19 is set to change for the better in the future.", "In conclusion, this NMA demonstrated that ivermectin/doxycycline, C-IVIG, methylprednisolone, IFN-β/SOC, IFN-β-1b, CP, remdesivir, LPV/r, IG, HS, auxora, and imatinib were effective for treating severe COVID-19 patients. There may be a difference in the findings of medications for severe COVID-19 patients from different control conditions (i.e., placebo and SOC) in RCTs. We found that most medications were safe in treating severe COVID-19. The present NMA reported uncertain estimates on the efficacy and safety of medications in the severe COVID-19 treatment. Maybe it’s because there was inadequate evidence of a reduction in ACM and the absence of TEAEs. However, this study had 2 strengths. One was that a comprehensive meta-analysis strategy was used to reduce the risk of publication bias. The other was that the SUCRA was used to assess possibly the best intervention.\nDespite these limitations, to date, the present findings might represent the most comprehensive meta-analysis of the available evidence for severe COVID-19 infection. Future guidelines and decision-making treatment plan should consider these results for the severe COVID-19 treatment. Importantly, the government, academia and researchers should collaborate to develop more large-scale RCTs studies and further estimate the efficacy and safety of treatment interventions on mortality, virological and clinical outcomes for different levels of infection with COVID-19.", "Conceptualization: Cheng Qing Lin, Zhao Gang.\nData curation: Chen Jun Fang, Jia Qing Jun, Fang Zi Jian.\nFormal analysis: Cheng Qing Lin, Zhao Gang.\nFunding acquisition: Cheng Qing Lin.\nMethodology: Cheng Qing Lin, Chen Jun Fang.\nSupervision: Jia Qing Jun, Fang Zi Jian.\nWriting – original draft: Cheng Qing Lin.\nWriting – review & editing: Cheng Qing Lin, Zhao Gang.", "We acknowledge with gratitude all study authors who responded to our data requests. We thank many researchers who sent information for our previous reviews on which this report was built. This work is supported by the Basic Public Welfare Research Project of Zhejiang Province (grant number: LGF21H260007), the Medical Science and Technology Project of Zhejiang Province (grant numbers: 2020PY064 and 2021PY065), and the Key Medical Discipline construction project (Disinfection and Vector Control) of Hangzhou.", "" ]
[ "intro", "methods", null, null, null, null, null, null, null, null, null, null, null, "results", null, null, null, null, null, null, null, "discussion", null, null, "conclusions", null, null, "supplementary-material" ]
[ "efficacy", "network meta-analysis", "randomized controlled trials", "safety", "severe COVID-19" ]
Development of a values-based decision aid to determine discharge destination: Case reports of older stroke survivors and their families.
36254089
Older adults affected by stroke must face a difficult choice between receiving post-discharge care at home or in a facility. This study aimed to develop a decision aid (DA) to help older adults and their families choose the place of post-discharge care based on their values.
BACKGROUND
Values and data for designing the DA were obtained through interviews with older stroke patients and their families, a questionnaire survey of various health professionals, and a review of patients' medical records. Next, a prototypic DA was prepared and tested for comprehensibility and usability using the 12-item International Patient Decision Aid Standards instrument.
METHODS
The DA was developed based on the following 6 values that were common among older stroke patients and their families: "activities of daily living," "services and fees," "emergencies," "family support," "environment," and "home renovation." The prototype met the criteria in the comprehensibility and usability tests.
RESULTS
Older stroke patients can use the DA to think through the evidence-based information matching their own values to make a more satisfactory decision. The effectiveness of this DA should further be investigated in clinical settings.
CONCLUSION
[ "Activities of Daily Living", "Aftercare", "Aged", "Decision Support Techniques", "Humans", "Patient Discharge", "Stroke", "Survivors" ]
9575811
1. Introduction
Cerebral stroke is the second leading cause of death worldwide.[1] The morbidity and recurrence of stroke increase with age, and stroke is prone to aggravation. Therefore, older adults affected by stroke are faced with the difficult decision of whether to live at home as before or receive care elsewhere after discharge. Self-determination is not a written or codified right in Japan, and the majority of older patients make the decision with their families. Furthermore, as families are often responsible for taking care of the older members, the family’s say is prioritized in the decision-making process.[2] In fact, it is not all rare that decisions about an older patient are made by other family members and medical and/or welfare professionals without the patient’s participation. For this reason, studies on discharge support for older adults in Japan have mainly consisted of case studies and surveys from the perspective of families or professionals, such as identification screening items and developing screening tools for promoting early discharge,[3] factors affecting the discharge destination,[4,5] and nurses’ coordination abilities or decision-making support.[6] There are almost no studies or assessments from the viewpoint of older adults. Decision-making support about discharge destination is one of the most difficult challenges in hospitals. Indeed, it has been reported that the greatest challenge is coordination to find a compromise between different intentions of older adults and their families and even professionals.[7] However, an established method of decision-making support is not available for use in healthcare settings in Japan. A very large number of randomized controlled trials have been included in reviews on discharge support for older adults in other countries.[8–10] The benefits of discharge support evaluated in these studies were mainly related to costs, such as shortened hospital stays, lower readmission rates, decreased mortality, and increased satisfaction and quality of life of patients and caregivers. Pioneering studies on decision-making support have been conducted in the US, Canada, the UK, and Germany, and have reported that the involvement of the patient in decision-making[11] and clarification of values are keys to better decision-making. Decision aids (DAs) are being developed actively as means of supporting decision-making and have been demonstrated to be effective for increasing knowledge, reducing conflicts and unclarity of values, and increasing participation in the decision-making process.[12] van Weert et al[13] showed that DAs are effective even in older adults for increasing knowledge, accurately perceiving risks, reducing conflicts in decision-making, and increasing patient participation. Evidence and the effectiveness of DAs for decision-making on where older adults should receive care have not yet been demonstrated because such decisions are particularly difficult to make due to the communication difficulties older adults face and the various social systems and people/places involved.[14,15] To date, only Garvelink et al[14] have developed a DA for selecting care at home or at a long-term care facility for frail older adults from the perspective of caregivers. However, participation by older adults in the development of this DA was difficult because they were frail or in the terminal stage. In Japan, information brochures are commonly used for decision-making on discharge destinations, but they often contain more information than necessary and confuse many older adults and their families. This problem prompted us to develop a DA in which the information is arranged to help older adults and their families compare the pros and cons of each option and make choices matching their values. However, only a small number of DAs have been developed previously in Japan, and none of them have been developed based on assessments by professionals in various disciplines. In addition, frail subjects such as older adults have very rarely been involved in the development of DAs anywhere in the world. This study thus aimed to develop a DA to help older adults and their families to make decisions on the place for post-discharge care based on their values.
2. Methods
[SUBTITLE] 2.1. Study design and process of development [SUBSECTION] This study consisted of a 3-cycle mixed quantitative/qualitative study involving older stroke patients, the ultimate users of the DA, families, and multidisciplinary clinicians (physicians, nurses, physical therapist [PT] occupational therapists [OT], and medical social worker [MSW]) to develop a decision-making guide in line with the values of older stroke patients and their families. Specifically, the steps consisted of Cycle 1: Selection of items, Cycle 2: The development prototype, and Cycle 3: Validity assessment. The process of development followed the structural process of Coulter et al,[16] who performed a systemic review of articles on DA (Fig. 1), with references to the International Patient Decision Aid Standards instrument (IPDASi),[17] Ottawa Personal Decision Guide,[18] and DA for older adults[14,19] as well as brochures issued by the Japanese local government units, such as cities, towns, and villages. Model development process for decision aid. This study consisted of a 3-cycle mixed quantitative/qualitative study involving older stroke patients, the ultimate users of the DA, families, and multidisciplinary clinicians (physicians, nurses, physical therapist [PT] occupational therapists [OT], and medical social worker [MSW]) to develop a decision-making guide in line with the values of older stroke patients and their families. Specifically, the steps consisted of Cycle 1: Selection of items, Cycle 2: The development prototype, and Cycle 3: Validity assessment. The process of development followed the structural process of Coulter et al,[16] who performed a systemic review of articles on DA (Fig. 1), with references to the International Patient Decision Aid Standards instrument (IPDASi),[17] Ottawa Personal Decision Guide,[18] and DA for older adults[14,19] as well as brochures issued by the Japanese local government units, such as cities, towns, and villages. Model development process for decision aid. [SUBTITLE] 2.2. Cycle 1: Selection of items [SUBSECTION] [SUBTITLE] 2.2..1. Approach 1. Assess patients’ views on decisional needs. [SUBSECTION] Convenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20] Convenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20] [SUBTITLE] 2.2..2. Approach 2. Assess clinicians’ views on patients’ needs. [SUBSECTION] In total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all. In total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all. [SUBTITLE] 2.2..3. Approach 3. Determine format and distribution. [SUBSECTION] Effective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia. Effective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia. [SUBTITLE] 2.2..4. Approach 4. Review and synthesize evidence. [SUBSECTION] To incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded. To incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded. [SUBTITLE] 2.2..1. Approach 1. Assess patients’ views on decisional needs. [SUBSECTION] Convenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20] Convenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20] [SUBTITLE] 2.2..2. Approach 2. Assess clinicians’ views on patients’ needs. [SUBSECTION] In total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all. In total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all. [SUBTITLE] 2.2..3. Approach 3. Determine format and distribution. [SUBSECTION] Effective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia. Effective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia. [SUBTITLE] 2.2..4. Approach 4. Review and synthesize evidence. [SUBSECTION] To incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded. To incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded. [SUBTITLE] 2.3. Cycle 2: The development prototype [SUBSECTION] After accomplishing the Approach 1, 2, 3, and 4 stages, a prototype DA consisting of 12 A4-sized pages was prepared. Previous DA studies relied on the generic Ottawa Personal Decision Guide[18] founded on the Ottawa Decision Support Framework (ODSF) as reference and also met the conditions of the IPDASi. DA derived from the ODSF can enhance knowledge and the accurate awareness of risk, promote decision-making matching personal values, and reduce the percentage of patients facing difficulty in making the decision; thus, using the ODSF is reported to be a valid approach for developing a DA.[22] Furthermore, the Ottawa Personal Decision Guide incorporates data specialized for decision-making related to options, risks, and benefits, meeting the minimal quality standards of a DA.[18] Therefore, we decided to apply the structure of introduction, facts and information, pros and cons of various options, expressing and weighing values, and checking preparedness for decision-making. After accomplishing the Approach 1, 2, 3, and 4 stages, a prototype DA consisting of 12 A4-sized pages was prepared. Previous DA studies relied on the generic Ottawa Personal Decision Guide[18] founded on the Ottawa Decision Support Framework (ODSF) as reference and also met the conditions of the IPDASi. DA derived from the ODSF can enhance knowledge and the accurate awareness of risk, promote decision-making matching personal values, and reduce the percentage of patients facing difficulty in making the decision; thus, using the ODSF is reported to be a valid approach for developing a DA.[22] Furthermore, the Ottawa Personal Decision Guide incorporates data specialized for decision-making related to options, risks, and benefits, meeting the minimal quality standards of a DA.[18] Therefore, we decided to apply the structure of introduction, facts and information, pros and cons of various options, expressing and weighing values, and checking preparedness for decision-making. [SUBTITLE] 2.4. Cycle 3: Validity assessment [SUBSECTION] To meet the minimal scientific standards of a DA, 11 experts (5 researchers in decision-making and 6 graduate students who were nurses and public health nurses) were recruited to perform the comprehensibility test questionnaire to evaluate whether the DA met all the 12 minimal criteria of the IPDASi (Table 1), which entailed 6 items on qualifying criteria that determine that an intervention is a form of DA and 6 items on criteria to evaluate the risk of any detrimental biases, in the case that the qualifying criteria are not met. The qualifying criteria were evaluated binarily by a “yes” or “no.” The certifying criteria were evaluated on a scale from “not at all (1 point)” to “very much (4 points).” All qualifying criteria must be obligatorily met, but it is recommended that the certifying criteria be met as well. Comprehensibility test. IPDAS minimal qualifying and certification criteria for decision aids (Garvelink, 2016). DA = decision aid. Furthermore, a usability test (Table 2) was conducted with a questionnaire to evaluate the ease of using the DA on 20 subjects, including 1 older stroke patient aged ≥ 65 years, 1 family member, clinicians (1 physician, 1 nurse, 1 PT, 1 OT, and 1 MSW), 1 discharge support nurse, 1 older adult participant from the general population, and 11 experts in decision-making. The questionnaire included 7 items evaluated on a scale of “Not at all (0 points)” to “very much (3 points).” The questionnaire was created with reference to a previous study[14] and the User Manual for Acceptability,[23] and the respondents were encouraged to offer opinions for a draft DA in the free response section of the questionnaire. Usability test (N = 21). Reference: Garvelink (2016). DA = decision aid. To meet the minimal scientific standards of a DA, 11 experts (5 researchers in decision-making and 6 graduate students who were nurses and public health nurses) were recruited to perform the comprehensibility test questionnaire to evaluate whether the DA met all the 12 minimal criteria of the IPDASi (Table 1), which entailed 6 items on qualifying criteria that determine that an intervention is a form of DA and 6 items on criteria to evaluate the risk of any detrimental biases, in the case that the qualifying criteria are not met. The qualifying criteria were evaluated binarily by a “yes” or “no.” The certifying criteria were evaluated on a scale from “not at all (1 point)” to “very much (4 points).” All qualifying criteria must be obligatorily met, but it is recommended that the certifying criteria be met as well. Comprehensibility test. IPDAS minimal qualifying and certification criteria for decision aids (Garvelink, 2016). DA = decision aid. Furthermore, a usability test (Table 2) was conducted with a questionnaire to evaluate the ease of using the DA on 20 subjects, including 1 older stroke patient aged ≥ 65 years, 1 family member, clinicians (1 physician, 1 nurse, 1 PT, 1 OT, and 1 MSW), 1 discharge support nurse, 1 older adult participant from the general population, and 11 experts in decision-making. The questionnaire included 7 items evaluated on a scale of “Not at all (0 points)” to “very much (3 points).” The questionnaire was created with reference to a previous study[14] and the User Manual for Acceptability,[23] and the respondents were encouraged to offer opinions for a draft DA in the free response section of the questionnaire. Usability test (N = 21). Reference: Garvelink (2016). DA = decision aid. [SUBTITLE] 2.5. Steering: Review by steering group; redraft and redesign, if necessary [SUBSECTION] The content was corrected and the layout was modified according to the results of the comprehensibility and usability tests to revise the draft DA. The layout was prepared by a graphic designer. Finally, the content of the completed DA was evaluated by older stroke patients, their families, and clinicians. The content was corrected and the layout was modified according to the results of the comprehensibility and usability tests to revise the draft DA. The layout was prepared by a graphic designer. Finally, the content of the completed DA was evaluated by older stroke patients, their families, and clinicians. [SUBTITLE] 2.6. Ethics approval and consent to participate [SUBSECTION] Written informed consent to participate in the interview survey was obtained after explaining the nature of the study orally and in writing. Returned questionnaires were interpreted as consent for questionnaires distributed to clinicians and for evaluating content validity. This study was approved by the institutional review boards of St. Luke’s International University (16-A024), Toyama Prefectural Rehabilitation Hospital & Support Center for Children with Disabilities (No.31), and University of Toyama (28-63). Written informed consent to participate in the interview survey was obtained after explaining the nature of the study orally and in writing. Returned questionnaires were interpreted as consent for questionnaires distributed to clinicians and for evaluating content validity. This study was approved by the institutional review boards of St. Luke’s International University (16-A024), Toyama Prefectural Rehabilitation Hospital & Support Center for Children with Disabilities (No.31), and University of Toyama (28-63).
3. Results
[SUBTITLE] 3.1. Cycle 1: Selection of items [SUBSECTION] In cycle 1, the selection of items was conducted. First, as a result of the literature review, we found a total of 127 references, 72 in Japanese and 55 in English. There were 2 existing DAs on where to receive post-discharge care.[14,24] However, they targeted end-of-life and frail older adults, and none addressed stroke patients. There were 2 Cochrane reviews on the location of post-discharge care for older stroke patients. Ward et al[25] compared the benefits of home and hospital for the rehabilitation of older adults, and Boland et al[26] compared home with other locations for the care of older adults, but the pros and cons of the location for post-discharge care of older adults were not mentioned clearly. That is, the 2 existing DAs did not provide evidence related to the place of convalescence, so rather than presenting the advantages and disadvantages, they had incorporated things the user should know and made references to preferences and values. Through this review, we found evidence that research on where older stroke patients decide to receive post-discharge care is not established, and we thus decided to review the medical records of 103 older stroke patients aged ≥ 65 years in the participating facilities to investigate the factors influencing discharge destinations. The results showed that the factors that influenced the discharge destination were “ADL,” “eating disorder,” and “family’s preference for discharge destination.” Five values were identified: “Degree of independence in ADL,” “environment for convalescence,” “family relationship,” “disease management,” and “social resources and fees.” These were common values that affected the decision-making of older stroke patients and their families regarding the discharge destination (Table 3). Qualitative synthesis. () Number of codes. In addition, we selected items for the DA that would help clarify these 5 values and that we considered to be important information from the professional viewpoint. The contents were selected to include the following: contents of long-term care insurance services, consultation services, costs, home repairs, prospects during hospitalization, family wishes, ADL, and responses to illness. The layout of the DA was designed to enhance legibility for older adults by using large font sizes, easy-to-read fonts, and clear contrasts. Furthermore, the DA had a 2-page format printed on a single folded sheet to make it user friendly for users who may have disabilities, such as memory impairment and aphasia, and was printed on a brochure format so that it could be used with family and clinicians. Kanji characters and illustrations were used with preference over hiragana and katakana phonetic letters, and items that required taking memos or writing were minimized. In cycle 1, the selection of items was conducted. First, as a result of the literature review, we found a total of 127 references, 72 in Japanese and 55 in English. There were 2 existing DAs on where to receive post-discharge care.[14,24] However, they targeted end-of-life and frail older adults, and none addressed stroke patients. There were 2 Cochrane reviews on the location of post-discharge care for older stroke patients. Ward et al[25] compared the benefits of home and hospital for the rehabilitation of older adults, and Boland et al[26] compared home with other locations for the care of older adults, but the pros and cons of the location for post-discharge care of older adults were not mentioned clearly. That is, the 2 existing DAs did not provide evidence related to the place of convalescence, so rather than presenting the advantages and disadvantages, they had incorporated things the user should know and made references to preferences and values. Through this review, we found evidence that research on where older stroke patients decide to receive post-discharge care is not established, and we thus decided to review the medical records of 103 older stroke patients aged ≥ 65 years in the participating facilities to investigate the factors influencing discharge destinations. The results showed that the factors that influenced the discharge destination were “ADL,” “eating disorder,” and “family’s preference for discharge destination.” Five values were identified: “Degree of independence in ADL,” “environment for convalescence,” “family relationship,” “disease management,” and “social resources and fees.” These were common values that affected the decision-making of older stroke patients and their families regarding the discharge destination (Table 3). Qualitative synthesis. () Number of codes. In addition, we selected items for the DA that would help clarify these 5 values and that we considered to be important information from the professional viewpoint. The contents were selected to include the following: contents of long-term care insurance services, consultation services, costs, home repairs, prospects during hospitalization, family wishes, ADL, and responses to illness. The layout of the DA was designed to enhance legibility for older adults by using large font sizes, easy-to-read fonts, and clear contrasts. Furthermore, the DA had a 2-page format printed on a single folded sheet to make it user friendly for users who may have disabilities, such as memory impairment and aphasia, and was printed on a brochure format so that it could be used with family and clinicians. Kanji characters and illustrations were used with preference over hiragana and katakana phonetic letters, and items that required taking memos or writing were minimized. [SUBTITLE] 3.2. Cycle 2: Prototype [SUBSECTION] In Introduction of DA prototype Version 1, 3 discharge destinations options, that is, “home,” “residence/facility (for independent people),” and “hospital/facility (for people who need long-term care),” were listed, and how to use the DA, flow from admission to discharge, wishes and level of preparation, and the types of clinicians and distinction of their roles were described. In Facts and information, stroke, recurrence prophylaxis, long-term insurance, and features of the discharge destinations were described in detail. In Pros and cons of various options and clear expression of values and Weighing, the pros and cons of the 3 discharge destination options were printed next to the options to enable easy comparison as they related to the values that were commonly observed in older stroke patients and their families. Furthermore, a scale of “Not important 1 point” to “Very important 5 points” was used and placed vertically next to each item so that the level of importance could be compared easily. Items were added to Checking preparedness for decision-making to identify anybody other than the respondent who would help with their decision-making and who they wanted to do it with. The DA prototype Version 1 was completed on November 17, 2017, and the development group conducted a discussion to confirm that it met all the minimal criteria, that is, 12 items of the IPDASi. In Introduction of DA prototype Version 1, 3 discharge destinations options, that is, “home,” “residence/facility (for independent people),” and “hospital/facility (for people who need long-term care),” were listed, and how to use the DA, flow from admission to discharge, wishes and level of preparation, and the types of clinicians and distinction of their roles were described. In Facts and information, stroke, recurrence prophylaxis, long-term insurance, and features of the discharge destinations were described in detail. In Pros and cons of various options and clear expression of values and Weighing, the pros and cons of the 3 discharge destination options were printed next to the options to enable easy comparison as they related to the values that were commonly observed in older stroke patients and their families. Furthermore, a scale of “Not important 1 point” to “Very important 5 points” was used and placed vertically next to each item so that the level of importance could be compared easily. Items were added to Checking preparedness for decision-making to identify anybody other than the respondent who would help with their decision-making and who they wanted to do it with. The DA prototype Version 1 was completed on November 17, 2017, and the development group conducted a discussion to confirm that it met all the minimal criteria, that is, 12 items of the IPDASi. [SUBTITLE] 3.3. Cycle 3 [SUBSECTION] To be classified as a DA according to the 6 qualifying criteria of the IPDASi in the comprehensibility test (Table 4), all criteria must be met. The only criterion that all respondents said they met was “Available options.” In particular, 2 respondents (18.2%) answered no for “positive characteristics (pros),” “negative characteristics (cons),” and “experience of the consequence of the options.” The 6 certifying criteria of the IPDASi must be answered by “Applicable” or higher on the 4-level scale of evaluation to meet the certifying criteria. The only items for which all respondents answered “Applicable” or higher were “Production or publication date” and “Information about the funding source used for development.” The certifying criteria were met the least for “Information about the levels of uncertainty associated with event or outcome probabilities,” with 8 respondents (72.7%) answering that it was not applicable, and 2 respondents (18.2%) answering that it was not at all applicable. The outcomes of the usability test (Table 5) of evaluation by component of the DA draft 1) ~ 5) were “Normal” or above for 90% of the respondents or higher for almost all items. Further, ≥80% respondents gave good evaluations for “understandable language,” “length,” and “content bias.” However, respondents gave the lowest evaluations for the item on “clarity of how the DA should be used.” The result of comprehensibility test (N = 11). Number of respondents/total number of respondents. No. 7 was unanswered by one. The result of usability test (N = 20). Questions 2 and 4 are reversed, number of respondents/total number of people. DA = decision aid. In addition, in the free response section, the need for a table of contents, ingenuity to make it easier for patients and their families to imagine post-discharge daily life, weak expression of pros and cons, layout font, font size, and colors used were pointed out. There were also requests for providing additional information on the per-month costs of living in facilities and the types and roles of staff involved during hospital stay or after discharge. Furthermore, other respondents provided opinions such as “I believe that I will be able to make the best judgment for myself with staff support and explanations based on this DA,” “Wouldn’t the gap between values and reality cause problems?,” and “It would be hard to just be handed the DA and read and understand it by myself, so it would be better to look at it while somebody is explaining it to me.” To be classified as a DA according to the 6 qualifying criteria of the IPDASi in the comprehensibility test (Table 4), all criteria must be met. The only criterion that all respondents said they met was “Available options.” In particular, 2 respondents (18.2%) answered no for “positive characteristics (pros),” “negative characteristics (cons),” and “experience of the consequence of the options.” The 6 certifying criteria of the IPDASi must be answered by “Applicable” or higher on the 4-level scale of evaluation to meet the certifying criteria. The only items for which all respondents answered “Applicable” or higher were “Production or publication date” and “Information about the funding source used for development.” The certifying criteria were met the least for “Information about the levels of uncertainty associated with event or outcome probabilities,” with 8 respondents (72.7%) answering that it was not applicable, and 2 respondents (18.2%) answering that it was not at all applicable. The outcomes of the usability test (Table 5) of evaluation by component of the DA draft 1) ~ 5) were “Normal” or above for 90% of the respondents or higher for almost all items. Further, ≥80% respondents gave good evaluations for “understandable language,” “length,” and “content bias.” However, respondents gave the lowest evaluations for the item on “clarity of how the DA should be used.” The result of comprehensibility test (N = 11). Number of respondents/total number of respondents. No. 7 was unanswered by one. The result of usability test (N = 20). Questions 2 and 4 are reversed, number of respondents/total number of people. DA = decision aid. In addition, in the free response section, the need for a table of contents, ingenuity to make it easier for patients and their families to imagine post-discharge daily life, weak expression of pros and cons, layout font, font size, and colors used were pointed out. There were also requests for providing additional information on the per-month costs of living in facilities and the types and roles of staff involved during hospital stay or after discharge. Furthermore, other respondents provided opinions such as “I believe that I will be able to make the best judgment for myself with staff support and explanations based on this DA,” “Wouldn’t the gap between values and reality cause problems?,” and “It would be hard to just be handed the DA and read and understand it by myself, so it would be better to look at it while somebody is explaining it to me.” [SUBTITLE] 3.4. Steering [SUBSECTION] To clarify the methods of use, an explanation on the instructions for use was added to the DA. The table of the pros and cons of the options was reorganized and improved. As the options were difficult to differentiate, the 2 options were named “Same place as before admission” and “Different place from before admission.” To make it easier for older adults to understand, the layout, organization, and terms used for the information were further modified into succinct and easily comprehensible expressions. Moreover, home renovations were especially important for people who wished to return to their homes to recover, so it was placed as a separate item to clearly present the values. Finally, a DA based on the 6 values of “ADL,” “Services and fees,” “Emergencies,” “Family support,” “Environment,” and “Home renovation” was created. The DA that was revised again was distributed to one of each of the following persons: an older stroke patient, family member, physician, nurse, PT, OT, and MSW. They were then asked to check the entire document. However, only minor additions and revisions of wording were needed, and the DA was ultimately developed as a 12-page, A4 paper-sized document (Table 6). Comments and revision regarding the prototype content. ADL = activities of daily living, DA = decision aid. To clarify the methods of use, an explanation on the instructions for use was added to the DA. The table of the pros and cons of the options was reorganized and improved. As the options were difficult to differentiate, the 2 options were named “Same place as before admission” and “Different place from before admission.” To make it easier for older adults to understand, the layout, organization, and terms used for the information were further modified into succinct and easily comprehensible expressions. Moreover, home renovations were especially important for people who wished to return to their homes to recover, so it was placed as a separate item to clearly present the values. Finally, a DA based on the 6 values of “ADL,” “Services and fees,” “Emergencies,” “Family support,” “Environment,” and “Home renovation” was created. The DA that was revised again was distributed to one of each of the following persons: an older stroke patient, family member, physician, nurse, PT, OT, and MSW. They were then asked to check the entire document. However, only minor additions and revisions of wording were needed, and the DA was ultimately developed as a 12-page, A4 paper-sized document (Table 6). Comments and revision regarding the prototype content. ADL = activities of daily living, DA = decision aid.
null
null
[ "2.1. Study design and process of development", "2.2. Cycle 1: Selection of items", "2.2..1. Approach 1. Assess patients’ views on decisional needs.", "2.2..2. Approach 2. Assess clinicians’ views on patients’ needs.", "2.2..3. Approach 3. Determine format and distribution.", "2.2..4. Approach 4. Review and synthesize evidence.", "2.3. Cycle 2: The development prototype", "2.4. Cycle 3: Validity assessment", "2.5. Steering: Review by steering group; redraft and redesign, if necessary", "2.6. Ethics approval and consent to participate", "3.1. Cycle 1: Selection of items", "3.2. Cycle 2: Prototype", "3.3. Cycle 3", "3.4. Steering", "4.1. Study limitations and future prospects", "Acknowledgments", "Author contribution" ]
[ "This study consisted of a 3-cycle mixed quantitative/qualitative study involving older stroke patients, the ultimate users of the DA, families, and multidisciplinary clinicians (physicians, nurses, physical therapist [PT] occupational therapists [OT], and medical social worker [MSW]) to develop a decision-making guide in line with the values of older stroke patients and their families. Specifically, the steps consisted of Cycle 1: Selection of items, Cycle 2: The development prototype, and Cycle 3: Validity assessment. The process of development followed the structural process of Coulter et al,[16] who performed a systemic review of articles on DA (Fig. 1), with references to the International Patient Decision Aid Standards instrument (IPDASi),[17] Ottawa Personal Decision Guide,[18] and DA for older adults[14,19] as well as brochures issued by the Japanese local government units, such as cities, towns, and villages.\nModel development process for decision aid.", "[SUBTITLE] 2.2..1. Approach 1. Assess patients’ views on decisional needs. [SUBSECTION] Convenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20]\nConvenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20]\n[SUBTITLE] 2.2..2. Approach 2. Assess clinicians’ views on patients’ needs. [SUBSECTION] In total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all.\nIn total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all.\n[SUBTITLE] 2.2..3. Approach 3. Determine format and distribution. [SUBSECTION] Effective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia.\nEffective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia.\n[SUBTITLE] 2.2..4. Approach 4. Review and synthesize evidence. [SUBSECTION] To incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded.\nTo incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded.", "Convenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20]", "In total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all.", "Effective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia.", "To incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded.", "After accomplishing the Approach 1, 2, 3, and 4 stages, a prototype DA consisting of 12 A4-sized pages was prepared. Previous DA studies relied on the generic Ottawa Personal Decision Guide[18] founded on the Ottawa Decision Support Framework (ODSF) as reference and also met the conditions of the IPDASi. DA derived from the ODSF can enhance knowledge and the accurate awareness of risk, promote decision-making matching personal values, and reduce the percentage of patients facing difficulty in making the decision; thus, using the ODSF is reported to be a valid approach for developing a DA.[22] Furthermore, the Ottawa Personal Decision Guide incorporates data specialized for decision-making related to options, risks, and benefits, meeting the minimal quality standards of a DA.[18] Therefore, we decided to apply the structure of introduction, facts and information, pros and cons of various options, expressing and weighing values, and checking preparedness for decision-making.", "To meet the minimal scientific standards of a DA, 11 experts (5 researchers in decision-making and 6 graduate students who were nurses and public health nurses) were recruited to perform the comprehensibility test questionnaire to evaluate whether the DA met all the 12 minimal criteria of the IPDASi (Table 1), which entailed 6 items on qualifying criteria that determine that an intervention is a form of DA and 6 items on criteria to evaluate the risk of any detrimental biases, in the case that the qualifying criteria are not met. The qualifying criteria were evaluated binarily by a “yes” or “no.” The certifying criteria were evaluated on a scale from “not at all (1 point)” to “very much (4 points).” All qualifying criteria must be obligatorily met, but it is recommended that the certifying criteria be met as well.\nComprehensibility test.\nIPDAS minimal qualifying and certification criteria for decision aids (Garvelink, 2016).\nDA = decision aid.\nFurthermore, a usability test (Table 2) was conducted with a questionnaire to evaluate the ease of using the DA on 20 subjects, including 1 older stroke patient aged ≥ 65 years, 1 family member, clinicians (1 physician, 1 nurse, 1 PT, 1 OT, and 1 MSW), 1 discharge support nurse, 1 older adult participant from the general population, and 11 experts in decision-making. The questionnaire included 7 items evaluated on a scale of “Not at all (0 points)” to “very much (3 points).” The questionnaire was created with reference to a previous study[14] and the User Manual for Acceptability,[23] and the respondents were encouraged to offer opinions for a draft DA in the free response section of the questionnaire.\nUsability test (N = 21).\nReference: Garvelink (2016).\nDA = decision aid.", "The content was corrected and the layout was modified according to the results of the comprehensibility and usability tests to revise the draft DA. The layout was prepared by a graphic designer. Finally, the content of the completed DA was evaluated by older stroke patients, their families, and clinicians.", "Written informed consent to participate in the interview survey was obtained after explaining the nature of the study orally and in writing. Returned questionnaires were interpreted as consent for questionnaires distributed to clinicians and for evaluating content validity. This study was approved by the institutional review boards of St. Luke’s International University (16-A024), Toyama Prefectural Rehabilitation Hospital & Support Center for Children with Disabilities (No.31), and University of Toyama (28-63).", "In cycle 1, the selection of items was conducted. First, as a result of the literature review, we found a total of 127 references, 72 in Japanese and 55 in English. There were 2 existing DAs on where to receive post-discharge care.[14,24] However, they targeted end-of-life and frail older adults, and none addressed stroke patients. There were 2 Cochrane reviews on the location of post-discharge care for older stroke patients. Ward et al[25] compared the benefits of home and hospital for the rehabilitation of older adults, and Boland et al[26] compared home with other locations for the care of older adults, but the pros and cons of the location for post-discharge care of older adults were not mentioned clearly. That is, the 2 existing DAs did not provide evidence related to the place of convalescence, so rather than presenting the advantages and disadvantages, they had incorporated things the user should know and made references to preferences and values. Through this review, we found evidence that research on where older stroke patients decide to receive post-discharge care is not established, and we thus decided to review the medical records of 103 older stroke patients aged ≥ 65 years in the participating facilities to investigate the factors influencing discharge destinations. The results showed that the factors that influenced the discharge destination were “ADL,” “eating disorder,” and “family’s preference for discharge destination.”\nFive values were identified: “Degree of independence in ADL,” “environment for convalescence,” “family relationship,” “disease management,” and “social resources and fees.” These were common values that affected the decision-making of older stroke patients and their families regarding the discharge destination (Table 3).\nQualitative synthesis.\n() Number of codes.\nIn addition, we selected items for the DA that would help clarify these 5 values and that we considered to be important information from the professional viewpoint. The contents were selected to include the following: contents of long-term care insurance services, consultation services, costs, home repairs, prospects during hospitalization, family wishes, ADL, and responses to illness.\nThe layout of the DA was designed to enhance legibility for older adults by using large font sizes, easy-to-read fonts, and clear contrasts. Furthermore, the DA had a 2-page format printed on a single folded sheet to make it user friendly for users who may have disabilities, such as memory impairment and aphasia, and was printed on a brochure format so that it could be used with family and clinicians. Kanji characters and illustrations were used with preference over hiragana and katakana phonetic letters, and items that required taking memos or writing were minimized.", "In Introduction of DA prototype Version 1, 3 discharge destinations options, that is, “home,” “residence/facility (for independent people),” and “hospital/facility (for people who need long-term care),” were listed, and how to use the DA, flow from admission to discharge, wishes and level of preparation, and the types of clinicians and distinction of their roles were described. In Facts and information, stroke, recurrence prophylaxis, long-term insurance, and features of the discharge destinations were described in detail. In Pros and cons of various options and clear expression of values and Weighing, the pros and cons of the 3 discharge destination options were printed next to the options to enable easy comparison as they related to the values that were commonly observed in older stroke patients and their families. Furthermore, a scale of “Not important 1 point” to “Very important 5 points” was used and placed vertically next to each item so that the level of importance could be compared easily. Items were added to Checking preparedness for decision-making to identify anybody other than the respondent who would help with their decision-making and who they wanted to do it with. The DA prototype Version 1 was completed on November 17, 2017, and the development group conducted a discussion to confirm that it met all the minimal criteria, that is, 12 items of the IPDASi.", "To be classified as a DA according to the 6 qualifying criteria of the IPDASi in the comprehensibility test (Table 4), all criteria must be met. The only criterion that all respondents said they met was “Available options.” In particular, 2 respondents (18.2%) answered no for “positive characteristics (pros),” “negative characteristics (cons),” and “experience of the consequence of the options.” The 6 certifying criteria of the IPDASi must be answered by “Applicable” or higher on the 4-level scale of evaluation to meet the certifying criteria. The only items for which all respondents answered “Applicable” or higher were “Production or publication date” and “Information about the funding source used for development.” The certifying criteria were met the least for “Information about the levels of uncertainty associated with event or outcome probabilities,” with 8 respondents (72.7%) answering that it was not applicable, and 2 respondents (18.2%) answering that it was not at all applicable. The outcomes of the usability test (Table 5) of evaluation by component of the DA draft 1) ~ 5) were “Normal” or above for 90% of the respondents or higher for almost all items. Further, ≥80% respondents gave good evaluations for “understandable language,” “length,” and “content bias.” However, respondents gave the lowest evaluations for the item on “clarity of how the DA should be used.”\nThe result of comprehensibility test (N = 11).\nNumber of respondents/total number of respondents. No. 7 was unanswered by one.\nThe result of usability test (N = 20).\nQuestions 2 and 4 are reversed, number of respondents/total number of people.\nDA = decision aid.\nIn addition, in the free response section, the need for a table of contents, ingenuity to make it easier for patients and their families to imagine post-discharge daily life, weak expression of pros and cons, layout font, font size, and colors used were pointed out. There were also requests for providing additional information on the per-month costs of living in facilities and the types and roles of staff involved during hospital stay or after discharge. Furthermore, other respondents provided opinions such as “I believe that I will be able to make the best judgment for myself with staff support and explanations based on this DA,” “Wouldn’t the gap between values and reality cause problems?,” and “It would be hard to just be handed the DA and read and understand it by myself, so it would be better to look at it while somebody is explaining it to me.”", "To clarify the methods of use, an explanation on the instructions for use was added to the DA. The table of the pros and cons of the options was reorganized and improved. As the options were difficult to differentiate, the 2 options were named “Same place as before admission” and “Different place from before admission.” To make it easier for older adults to understand, the layout, organization, and terms used for the information were further modified into succinct and easily comprehensible expressions. Moreover, home renovations were especially important for people who wished to return to their homes to recover, so it was placed as a separate item to clearly present the values. Finally, a DA based on the 6 values of “ADL,” “Services and fees,” “Emergencies,” “Family support,” “Environment,” and “Home renovation” was created.\nThe DA that was revised again was distributed to one of each of the following persons: an older stroke patient, family member, physician, nurse, PT, OT, and MSW. They were then asked to check the entire document. However, only minor additions and revisions of wording were needed, and the DA was ultimately developed as a 12-page, A4 paper-sized document (Table 6).\nComments and revision regarding the prototype content.\nADL = activities of daily living, DA = decision aid.", "There were several limitations to this study. First, because feedback from so many different people, such as older stroke patients, their families, and various clinicians, was needed in the process of development, only feedback from a small number of each could be evaluated. Furthermore, as there were little data on where older adults receive post-discharge care and this data were supplemented by a review of medical records from a single institution, the data were not versatile and lacked generalizability. However, to the best of our knowledge, this is the first study that involved the participation of families, clinicians, and, most remarkably, older stroke patients themselves in the process of developing the DA from the preliminary survey of needs. In future, we plan to assign participants into 2 groups, one that is provided this DA and one that is not, to evaluate it in a randomized controlled trial.", "We express our sincerest gratitude to the older adults, their families, and clinicians in the hospital for their participation, which made this study possible. This work was supported by KAKENHI Grant-in-Aid for Young Scientists (B) (15K20759) and Grant-in-Aid for Scientific Research (C) (19K11244). The authors of this work have nothing to disclose.", "YA designed the study and drafted the manuscript. KN provided advice on the analysis and study overall, as well as offering suggestions. All authors have consented to publication and have checked the final document.\nConceptualization: Yoriko Aoki, Kazuhiro Nakayama.\nData curation: Yoriko Aoki, Kazuhiro Nakayama.\nFormal analysis: Yoriko Aoki, Kazuhiro Nakayama.\nWriting – original draft: Yoriko Aoki.\nWriting – review & editing: Yoriko Aoki." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "1. Introduction", "2. Methods", "2.1. Study design and process of development", "2.2. Cycle 1: Selection of items", "2.2..1. Approach 1. Assess patients’ views on decisional needs.", "2.2..2. Approach 2. Assess clinicians’ views on patients’ needs.", "2.2..3. Approach 3. Determine format and distribution.", "2.2..4. Approach 4. Review and synthesize evidence.", "2.3. Cycle 2: The development prototype", "2.4. Cycle 3: Validity assessment", "2.5. Steering: Review by steering group; redraft and redesign, if necessary", "2.6. Ethics approval and consent to participate", "3. Results", "3.1. Cycle 1: Selection of items", "3.2. Cycle 2: Prototype", "3.3. Cycle 3", "3.4. Steering", "4. Discussion", "4.1. Study limitations and future prospects", "Acknowledgments", "Author contribution" ]
[ "Cerebral stroke is the second leading cause of death worldwide.[1] The morbidity and recurrence of stroke increase with age, and stroke is prone to aggravation. Therefore, older adults affected by stroke are faced with the difficult decision of whether to live at home as before or receive care elsewhere after discharge. Self-determination is not a written or codified right in Japan, and the majority of older patients make the decision with their families. Furthermore, as families are often responsible for taking care of the older members, the family’s say is prioritized in the decision-making process.[2] In fact, it is not all rare that decisions about an older patient are made by other family members and medical and/or welfare professionals without the patient’s participation.\nFor this reason, studies on discharge support for older adults in Japan have mainly consisted of case studies and surveys from the perspective of families or professionals, such as identification screening items and developing screening tools for promoting early discharge,[3] factors affecting the discharge destination,[4,5] and nurses’ coordination abilities or decision-making support.[6] There are almost no studies or assessments from the viewpoint of older adults. Decision-making support about discharge destination is one of the most difficult challenges in hospitals. Indeed, it has been reported that the greatest challenge is coordination to find a compromise between different intentions of older adults and their families and even professionals.[7] However, an established method of decision-making support is not available for use in healthcare settings in Japan.\nA very large number of randomized controlled trials have been included in reviews on discharge support for older adults in other countries.[8–10] The benefits of discharge support evaluated in these studies were mainly related to costs, such as shortened hospital stays, lower readmission rates, decreased mortality, and increased satisfaction and quality of life of patients and caregivers. Pioneering studies on decision-making support have been conducted in the US, Canada, the UK, and Germany, and have reported that the involvement of the patient in decision-making[11] and clarification of values are keys to better decision-making. Decision aids (DAs) are being developed actively as means of supporting decision-making and have been demonstrated to be effective for increasing knowledge, reducing conflicts and unclarity of values, and increasing participation in the decision-making process.[12] van Weert et al[13] showed that DAs are effective even in older adults for increasing knowledge, accurately perceiving risks, reducing conflicts in decision-making, and increasing patient participation. Evidence and the effectiveness of DAs for decision-making on where older adults should receive care have not yet been demonstrated because such decisions are particularly difficult to make due to the communication difficulties older adults face and the various social systems and people/places involved.[14,15] To date, only Garvelink et al[14] have developed a DA for selecting care at home or at a long-term care facility for frail older adults from the perspective of caregivers. However, participation by older adults in the development of this DA was difficult because they were frail or in the terminal stage.\nIn Japan, information brochures are commonly used for decision-making on discharge destinations, but they often contain more information than necessary and confuse many older adults and their families. This problem prompted us to develop a DA in which the information is arranged to help older adults and their families compare the pros and cons of each option and make choices matching their values. However, only a small number of DAs have been developed previously in Japan, and none of them have been developed based on assessments by professionals in various disciplines. In addition, frail subjects such as older adults have very rarely been involved in the development of DAs anywhere in the world. This study thus aimed to develop a DA to help older adults and their families to make decisions on the place for post-discharge care based on their values.", "[SUBTITLE] 2.1. Study design and process of development [SUBSECTION] This study consisted of a 3-cycle mixed quantitative/qualitative study involving older stroke patients, the ultimate users of the DA, families, and multidisciplinary clinicians (physicians, nurses, physical therapist [PT] occupational therapists [OT], and medical social worker [MSW]) to develop a decision-making guide in line with the values of older stroke patients and their families. Specifically, the steps consisted of Cycle 1: Selection of items, Cycle 2: The development prototype, and Cycle 3: Validity assessment. The process of development followed the structural process of Coulter et al,[16] who performed a systemic review of articles on DA (Fig. 1), with references to the International Patient Decision Aid Standards instrument (IPDASi),[17] Ottawa Personal Decision Guide,[18] and DA for older adults[14,19] as well as brochures issued by the Japanese local government units, such as cities, towns, and villages.\nModel development process for decision aid.\nThis study consisted of a 3-cycle mixed quantitative/qualitative study involving older stroke patients, the ultimate users of the DA, families, and multidisciplinary clinicians (physicians, nurses, physical therapist [PT] occupational therapists [OT], and medical social worker [MSW]) to develop a decision-making guide in line with the values of older stroke patients and their families. Specifically, the steps consisted of Cycle 1: Selection of items, Cycle 2: The development prototype, and Cycle 3: Validity assessment. The process of development followed the structural process of Coulter et al,[16] who performed a systemic review of articles on DA (Fig. 1), with references to the International Patient Decision Aid Standards instrument (IPDASi),[17] Ottawa Personal Decision Guide,[18] and DA for older adults[14,19] as well as brochures issued by the Japanese local government units, such as cities, towns, and villages.\nModel development process for decision aid.\n[SUBTITLE] 2.2. Cycle 1: Selection of items [SUBSECTION] [SUBTITLE] 2.2..1. Approach 1. Assess patients’ views on decisional needs. [SUBSECTION] Convenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20]\nConvenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20]\n[SUBTITLE] 2.2..2. Approach 2. Assess clinicians’ views on patients’ needs. [SUBSECTION] In total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all.\nIn total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all.\n[SUBTITLE] 2.2..3. Approach 3. Determine format and distribution. [SUBSECTION] Effective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia.\nEffective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia.\n[SUBTITLE] 2.2..4. Approach 4. Review and synthesize evidence. [SUBSECTION] To incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded.\nTo incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded.\n[SUBTITLE] 2.2..1. Approach 1. Assess patients’ views on decisional needs. [SUBSECTION] Convenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20]\nConvenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20]\n[SUBTITLE] 2.2..2. Approach 2. Assess clinicians’ views on patients’ needs. [SUBSECTION] In total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all.\nIn total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all.\n[SUBTITLE] 2.2..3. Approach 3. Determine format and distribution. [SUBSECTION] Effective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia.\nEffective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia.\n[SUBTITLE] 2.2..4. Approach 4. Review and synthesize evidence. [SUBSECTION] To incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded.\nTo incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded.\n[SUBTITLE] 2.3. Cycle 2: The development prototype [SUBSECTION] After accomplishing the Approach 1, 2, 3, and 4 stages, a prototype DA consisting of 12 A4-sized pages was prepared. Previous DA studies relied on the generic Ottawa Personal Decision Guide[18] founded on the Ottawa Decision Support Framework (ODSF) as reference and also met the conditions of the IPDASi. DA derived from the ODSF can enhance knowledge and the accurate awareness of risk, promote decision-making matching personal values, and reduce the percentage of patients facing difficulty in making the decision; thus, using the ODSF is reported to be a valid approach for developing a DA.[22] Furthermore, the Ottawa Personal Decision Guide incorporates data specialized for decision-making related to options, risks, and benefits, meeting the minimal quality standards of a DA.[18] Therefore, we decided to apply the structure of introduction, facts and information, pros and cons of various options, expressing and weighing values, and checking preparedness for decision-making.\nAfter accomplishing the Approach 1, 2, 3, and 4 stages, a prototype DA consisting of 12 A4-sized pages was prepared. Previous DA studies relied on the generic Ottawa Personal Decision Guide[18] founded on the Ottawa Decision Support Framework (ODSF) as reference and also met the conditions of the IPDASi. DA derived from the ODSF can enhance knowledge and the accurate awareness of risk, promote decision-making matching personal values, and reduce the percentage of patients facing difficulty in making the decision; thus, using the ODSF is reported to be a valid approach for developing a DA.[22] Furthermore, the Ottawa Personal Decision Guide incorporates data specialized for decision-making related to options, risks, and benefits, meeting the minimal quality standards of a DA.[18] Therefore, we decided to apply the structure of introduction, facts and information, pros and cons of various options, expressing and weighing values, and checking preparedness for decision-making.\n[SUBTITLE] 2.4. Cycle 3: Validity assessment [SUBSECTION] To meet the minimal scientific standards of a DA, 11 experts (5 researchers in decision-making and 6 graduate students who were nurses and public health nurses) were recruited to perform the comprehensibility test questionnaire to evaluate whether the DA met all the 12 minimal criteria of the IPDASi (Table 1), which entailed 6 items on qualifying criteria that determine that an intervention is a form of DA and 6 items on criteria to evaluate the risk of any detrimental biases, in the case that the qualifying criteria are not met. The qualifying criteria were evaluated binarily by a “yes” or “no.” The certifying criteria were evaluated on a scale from “not at all (1 point)” to “very much (4 points).” All qualifying criteria must be obligatorily met, but it is recommended that the certifying criteria be met as well.\nComprehensibility test.\nIPDAS minimal qualifying and certification criteria for decision aids (Garvelink, 2016).\nDA = decision aid.\nFurthermore, a usability test (Table 2) was conducted with a questionnaire to evaluate the ease of using the DA on 20 subjects, including 1 older stroke patient aged ≥ 65 years, 1 family member, clinicians (1 physician, 1 nurse, 1 PT, 1 OT, and 1 MSW), 1 discharge support nurse, 1 older adult participant from the general population, and 11 experts in decision-making. The questionnaire included 7 items evaluated on a scale of “Not at all (0 points)” to “very much (3 points).” The questionnaire was created with reference to a previous study[14] and the User Manual for Acceptability,[23] and the respondents were encouraged to offer opinions for a draft DA in the free response section of the questionnaire.\nUsability test (N = 21).\nReference: Garvelink (2016).\nDA = decision aid.\nTo meet the minimal scientific standards of a DA, 11 experts (5 researchers in decision-making and 6 graduate students who were nurses and public health nurses) were recruited to perform the comprehensibility test questionnaire to evaluate whether the DA met all the 12 minimal criteria of the IPDASi (Table 1), which entailed 6 items on qualifying criteria that determine that an intervention is a form of DA and 6 items on criteria to evaluate the risk of any detrimental biases, in the case that the qualifying criteria are not met. The qualifying criteria were evaluated binarily by a “yes” or “no.” The certifying criteria were evaluated on a scale from “not at all (1 point)” to “very much (4 points).” All qualifying criteria must be obligatorily met, but it is recommended that the certifying criteria be met as well.\nComprehensibility test.\nIPDAS minimal qualifying and certification criteria for decision aids (Garvelink, 2016).\nDA = decision aid.\nFurthermore, a usability test (Table 2) was conducted with a questionnaire to evaluate the ease of using the DA on 20 subjects, including 1 older stroke patient aged ≥ 65 years, 1 family member, clinicians (1 physician, 1 nurse, 1 PT, 1 OT, and 1 MSW), 1 discharge support nurse, 1 older adult participant from the general population, and 11 experts in decision-making. The questionnaire included 7 items evaluated on a scale of “Not at all (0 points)” to “very much (3 points).” The questionnaire was created with reference to a previous study[14] and the User Manual for Acceptability,[23] and the respondents were encouraged to offer opinions for a draft DA in the free response section of the questionnaire.\nUsability test (N = 21).\nReference: Garvelink (2016).\nDA = decision aid.\n[SUBTITLE] 2.5. Steering: Review by steering group; redraft and redesign, if necessary [SUBSECTION] The content was corrected and the layout was modified according to the results of the comprehensibility and usability tests to revise the draft DA. The layout was prepared by a graphic designer. Finally, the content of the completed DA was evaluated by older stroke patients, their families, and clinicians.\nThe content was corrected and the layout was modified according to the results of the comprehensibility and usability tests to revise the draft DA. The layout was prepared by a graphic designer. Finally, the content of the completed DA was evaluated by older stroke patients, their families, and clinicians.\n[SUBTITLE] 2.6. Ethics approval and consent to participate [SUBSECTION] Written informed consent to participate in the interview survey was obtained after explaining the nature of the study orally and in writing. Returned questionnaires were interpreted as consent for questionnaires distributed to clinicians and for evaluating content validity. This study was approved by the institutional review boards of St. Luke’s International University (16-A024), Toyama Prefectural Rehabilitation Hospital & Support Center for Children with Disabilities (No.31), and University of Toyama (28-63).\nWritten informed consent to participate in the interview survey was obtained after explaining the nature of the study orally and in writing. Returned questionnaires were interpreted as consent for questionnaires distributed to clinicians and for evaluating content validity. This study was approved by the institutional review boards of St. Luke’s International University (16-A024), Toyama Prefectural Rehabilitation Hospital & Support Center for Children with Disabilities (No.31), and University of Toyama (28-63).", "This study consisted of a 3-cycle mixed quantitative/qualitative study involving older stroke patients, the ultimate users of the DA, families, and multidisciplinary clinicians (physicians, nurses, physical therapist [PT] occupational therapists [OT], and medical social worker [MSW]) to develop a decision-making guide in line with the values of older stroke patients and their families. Specifically, the steps consisted of Cycle 1: Selection of items, Cycle 2: The development prototype, and Cycle 3: Validity assessment. The process of development followed the structural process of Coulter et al,[16] who performed a systemic review of articles on DA (Fig. 1), with references to the International Patient Decision Aid Standards instrument (IPDASi),[17] Ottawa Personal Decision Guide,[18] and DA for older adults[14,19] as well as brochures issued by the Japanese local government units, such as cities, towns, and villages.\nModel development process for decision aid.", "[SUBTITLE] 2.2..1. Approach 1. Assess patients’ views on decisional needs. [SUBSECTION] Convenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20]\nConvenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20]\n[SUBTITLE] 2.2..2. Approach 2. Assess clinicians’ views on patients’ needs. [SUBSECTION] In total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all.\nIn total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all.\n[SUBTITLE] 2.2..3. Approach 3. Determine format and distribution. [SUBSECTION] Effective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia.\nEffective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia.\n[SUBTITLE] 2.2..4. Approach 4. Review and synthesize evidence. [SUBSECTION] To incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded.\nTo incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded.", "Convenience sampling was conducted on 10 older stroke patients aged ≥ 65 years and 10 key persons in their families. Those who could not clearly state their own intentions due to severe dementia or aphasia were excluded. We interviewed them about their values and needs associated with decision-making, and the data obtained from the interviews were analyzed.[20]", "In total, 39 clinicians (7 physicians, 11 nurses, 8 PT, 4 OT, and 9 MSW) who were in charge of 10 older stroke patients who had been hospitalized for 1 week were surveyed. A questionnaire was developed to examine information items that patients and their families should know when deciding where to be discharged from the hospital, based on previous studies.[3–5] We analyzed whether the information items were necessary or unnecessary by simple tabulation. Specifically, the survey inquired on items related to medical care (neurovascular disease): that is, disease, physical function (abilities and disabilities in activities of daily living [ADL]), cognitive decline, chronic medical care, medication management, family’s long-term care-giving capacities, and items related to public health and welfare: that is, service points, procedures of application for the physical disability certificate, need for and details of long-term care insurance (steps from application to certification, details and conditions for available services, fees, and list of providers) in a free response format. Respondents were further asked to select from one of the options as to who needed the above information for deciding discharge destination: patient only, family only, patient and the family, and not needed at all.", "Effective layout and format of the brochure were investigated by taking into account the advanced age of the older stroke patients and the characteristics of their common disorders and disabilities, such as motor disability, memory impairment, attention deficit, and aphasia.", "To incorporate the benefits and risks of each place for post-discharge care for older stroke patients in the DA, we referred to the 278-item Ottawa Hospital Research Institute A to Z Inventory[21] to first research about existing and general DA. DAs with unclear details were ordered by contacting the authors directly via email, and a literature review was conducted using international databases, such as the Cochrane Library, Medline, CINAHL, and JAMAS, for Japanese articles only. The literature review was limited to academic papers, and a full-year search was conducted. The keywords used included “decision aids,” “location of care,” and “discharge planning,” and the age category was set to ≥ 65 years. In addition, “Stoke” was multiplied, and content related to mental illness and acute conditions was excluded.", "After accomplishing the Approach 1, 2, 3, and 4 stages, a prototype DA consisting of 12 A4-sized pages was prepared. Previous DA studies relied on the generic Ottawa Personal Decision Guide[18] founded on the Ottawa Decision Support Framework (ODSF) as reference and also met the conditions of the IPDASi. DA derived from the ODSF can enhance knowledge and the accurate awareness of risk, promote decision-making matching personal values, and reduce the percentage of patients facing difficulty in making the decision; thus, using the ODSF is reported to be a valid approach for developing a DA.[22] Furthermore, the Ottawa Personal Decision Guide incorporates data specialized for decision-making related to options, risks, and benefits, meeting the minimal quality standards of a DA.[18] Therefore, we decided to apply the structure of introduction, facts and information, pros and cons of various options, expressing and weighing values, and checking preparedness for decision-making.", "To meet the minimal scientific standards of a DA, 11 experts (5 researchers in decision-making and 6 graduate students who were nurses and public health nurses) were recruited to perform the comprehensibility test questionnaire to evaluate whether the DA met all the 12 minimal criteria of the IPDASi (Table 1), which entailed 6 items on qualifying criteria that determine that an intervention is a form of DA and 6 items on criteria to evaluate the risk of any detrimental biases, in the case that the qualifying criteria are not met. The qualifying criteria were evaluated binarily by a “yes” or “no.” The certifying criteria were evaluated on a scale from “not at all (1 point)” to “very much (4 points).” All qualifying criteria must be obligatorily met, but it is recommended that the certifying criteria be met as well.\nComprehensibility test.\nIPDAS minimal qualifying and certification criteria for decision aids (Garvelink, 2016).\nDA = decision aid.\nFurthermore, a usability test (Table 2) was conducted with a questionnaire to evaluate the ease of using the DA on 20 subjects, including 1 older stroke patient aged ≥ 65 years, 1 family member, clinicians (1 physician, 1 nurse, 1 PT, 1 OT, and 1 MSW), 1 discharge support nurse, 1 older adult participant from the general population, and 11 experts in decision-making. The questionnaire included 7 items evaluated on a scale of “Not at all (0 points)” to “very much (3 points).” The questionnaire was created with reference to a previous study[14] and the User Manual for Acceptability,[23] and the respondents were encouraged to offer opinions for a draft DA in the free response section of the questionnaire.\nUsability test (N = 21).\nReference: Garvelink (2016).\nDA = decision aid.", "The content was corrected and the layout was modified according to the results of the comprehensibility and usability tests to revise the draft DA. The layout was prepared by a graphic designer. Finally, the content of the completed DA was evaluated by older stroke patients, their families, and clinicians.", "Written informed consent to participate in the interview survey was obtained after explaining the nature of the study orally and in writing. Returned questionnaires were interpreted as consent for questionnaires distributed to clinicians and for evaluating content validity. This study was approved by the institutional review boards of St. Luke’s International University (16-A024), Toyama Prefectural Rehabilitation Hospital & Support Center for Children with Disabilities (No.31), and University of Toyama (28-63).", "[SUBTITLE] 3.1. Cycle 1: Selection of items [SUBSECTION] In cycle 1, the selection of items was conducted. First, as a result of the literature review, we found a total of 127 references, 72 in Japanese and 55 in English. There were 2 existing DAs on where to receive post-discharge care.[14,24] However, they targeted end-of-life and frail older adults, and none addressed stroke patients. There were 2 Cochrane reviews on the location of post-discharge care for older stroke patients. Ward et al[25] compared the benefits of home and hospital for the rehabilitation of older adults, and Boland et al[26] compared home with other locations for the care of older adults, but the pros and cons of the location for post-discharge care of older adults were not mentioned clearly. That is, the 2 existing DAs did not provide evidence related to the place of convalescence, so rather than presenting the advantages and disadvantages, they had incorporated things the user should know and made references to preferences and values. Through this review, we found evidence that research on where older stroke patients decide to receive post-discharge care is not established, and we thus decided to review the medical records of 103 older stroke patients aged ≥ 65 years in the participating facilities to investigate the factors influencing discharge destinations. The results showed that the factors that influenced the discharge destination were “ADL,” “eating disorder,” and “family’s preference for discharge destination.”\nFive values were identified: “Degree of independence in ADL,” “environment for convalescence,” “family relationship,” “disease management,” and “social resources and fees.” These were common values that affected the decision-making of older stroke patients and their families regarding the discharge destination (Table 3).\nQualitative synthesis.\n() Number of codes.\nIn addition, we selected items for the DA that would help clarify these 5 values and that we considered to be important information from the professional viewpoint. The contents were selected to include the following: contents of long-term care insurance services, consultation services, costs, home repairs, prospects during hospitalization, family wishes, ADL, and responses to illness.\nThe layout of the DA was designed to enhance legibility for older adults by using large font sizes, easy-to-read fonts, and clear contrasts. Furthermore, the DA had a 2-page format printed on a single folded sheet to make it user friendly for users who may have disabilities, such as memory impairment and aphasia, and was printed on a brochure format so that it could be used with family and clinicians. Kanji characters and illustrations were used with preference over hiragana and katakana phonetic letters, and items that required taking memos or writing were minimized.\nIn cycle 1, the selection of items was conducted. First, as a result of the literature review, we found a total of 127 references, 72 in Japanese and 55 in English. There were 2 existing DAs on where to receive post-discharge care.[14,24] However, they targeted end-of-life and frail older adults, and none addressed stroke patients. There were 2 Cochrane reviews on the location of post-discharge care for older stroke patients. Ward et al[25] compared the benefits of home and hospital for the rehabilitation of older adults, and Boland et al[26] compared home with other locations for the care of older adults, but the pros and cons of the location for post-discharge care of older adults were not mentioned clearly. That is, the 2 existing DAs did not provide evidence related to the place of convalescence, so rather than presenting the advantages and disadvantages, they had incorporated things the user should know and made references to preferences and values. Through this review, we found evidence that research on where older stroke patients decide to receive post-discharge care is not established, and we thus decided to review the medical records of 103 older stroke patients aged ≥ 65 years in the participating facilities to investigate the factors influencing discharge destinations. The results showed that the factors that influenced the discharge destination were “ADL,” “eating disorder,” and “family’s preference for discharge destination.”\nFive values were identified: “Degree of independence in ADL,” “environment for convalescence,” “family relationship,” “disease management,” and “social resources and fees.” These were common values that affected the decision-making of older stroke patients and their families regarding the discharge destination (Table 3).\nQualitative synthesis.\n() Number of codes.\nIn addition, we selected items for the DA that would help clarify these 5 values and that we considered to be important information from the professional viewpoint. The contents were selected to include the following: contents of long-term care insurance services, consultation services, costs, home repairs, prospects during hospitalization, family wishes, ADL, and responses to illness.\nThe layout of the DA was designed to enhance legibility for older adults by using large font sizes, easy-to-read fonts, and clear contrasts. Furthermore, the DA had a 2-page format printed on a single folded sheet to make it user friendly for users who may have disabilities, such as memory impairment and aphasia, and was printed on a brochure format so that it could be used with family and clinicians. Kanji characters and illustrations were used with preference over hiragana and katakana phonetic letters, and items that required taking memos or writing were minimized.\n[SUBTITLE] 3.2. Cycle 2: Prototype [SUBSECTION] In Introduction of DA prototype Version 1, 3 discharge destinations options, that is, “home,” “residence/facility (for independent people),” and “hospital/facility (for people who need long-term care),” were listed, and how to use the DA, flow from admission to discharge, wishes and level of preparation, and the types of clinicians and distinction of their roles were described. In Facts and information, stroke, recurrence prophylaxis, long-term insurance, and features of the discharge destinations were described in detail. In Pros and cons of various options and clear expression of values and Weighing, the pros and cons of the 3 discharge destination options were printed next to the options to enable easy comparison as they related to the values that were commonly observed in older stroke patients and their families. Furthermore, a scale of “Not important 1 point” to “Very important 5 points” was used and placed vertically next to each item so that the level of importance could be compared easily. Items were added to Checking preparedness for decision-making to identify anybody other than the respondent who would help with their decision-making and who they wanted to do it with. The DA prototype Version 1 was completed on November 17, 2017, and the development group conducted a discussion to confirm that it met all the minimal criteria, that is, 12 items of the IPDASi.\nIn Introduction of DA prototype Version 1, 3 discharge destinations options, that is, “home,” “residence/facility (for independent people),” and “hospital/facility (for people who need long-term care),” were listed, and how to use the DA, flow from admission to discharge, wishes and level of preparation, and the types of clinicians and distinction of their roles were described. In Facts and information, stroke, recurrence prophylaxis, long-term insurance, and features of the discharge destinations were described in detail. In Pros and cons of various options and clear expression of values and Weighing, the pros and cons of the 3 discharge destination options were printed next to the options to enable easy comparison as they related to the values that were commonly observed in older stroke patients and their families. Furthermore, a scale of “Not important 1 point” to “Very important 5 points” was used and placed vertically next to each item so that the level of importance could be compared easily. Items were added to Checking preparedness for decision-making to identify anybody other than the respondent who would help with their decision-making and who they wanted to do it with. The DA prototype Version 1 was completed on November 17, 2017, and the development group conducted a discussion to confirm that it met all the minimal criteria, that is, 12 items of the IPDASi.\n[SUBTITLE] 3.3. Cycle 3 [SUBSECTION] To be classified as a DA according to the 6 qualifying criteria of the IPDASi in the comprehensibility test (Table 4), all criteria must be met. The only criterion that all respondents said they met was “Available options.” In particular, 2 respondents (18.2%) answered no for “positive characteristics (pros),” “negative characteristics (cons),” and “experience of the consequence of the options.” The 6 certifying criteria of the IPDASi must be answered by “Applicable” or higher on the 4-level scale of evaluation to meet the certifying criteria. The only items for which all respondents answered “Applicable” or higher were “Production or publication date” and “Information about the funding source used for development.” The certifying criteria were met the least for “Information about the levels of uncertainty associated with event or outcome probabilities,” with 8 respondents (72.7%) answering that it was not applicable, and 2 respondents (18.2%) answering that it was not at all applicable. The outcomes of the usability test (Table 5) of evaluation by component of the DA draft 1) ~ 5) were “Normal” or above for 90% of the respondents or higher for almost all items. Further, ≥80% respondents gave good evaluations for “understandable language,” “length,” and “content bias.” However, respondents gave the lowest evaluations for the item on “clarity of how the DA should be used.”\nThe result of comprehensibility test (N = 11).\nNumber of respondents/total number of respondents. No. 7 was unanswered by one.\nThe result of usability test (N = 20).\nQuestions 2 and 4 are reversed, number of respondents/total number of people.\nDA = decision aid.\nIn addition, in the free response section, the need for a table of contents, ingenuity to make it easier for patients and their families to imagine post-discharge daily life, weak expression of pros and cons, layout font, font size, and colors used were pointed out. There were also requests for providing additional information on the per-month costs of living in facilities and the types and roles of staff involved during hospital stay or after discharge. Furthermore, other respondents provided opinions such as “I believe that I will be able to make the best judgment for myself with staff support and explanations based on this DA,” “Wouldn’t the gap between values and reality cause problems?,” and “It would be hard to just be handed the DA and read and understand it by myself, so it would be better to look at it while somebody is explaining it to me.”\nTo be classified as a DA according to the 6 qualifying criteria of the IPDASi in the comprehensibility test (Table 4), all criteria must be met. The only criterion that all respondents said they met was “Available options.” In particular, 2 respondents (18.2%) answered no for “positive characteristics (pros),” “negative characteristics (cons),” and “experience of the consequence of the options.” The 6 certifying criteria of the IPDASi must be answered by “Applicable” or higher on the 4-level scale of evaluation to meet the certifying criteria. The only items for which all respondents answered “Applicable” or higher were “Production or publication date” and “Information about the funding source used for development.” The certifying criteria were met the least for “Information about the levels of uncertainty associated with event or outcome probabilities,” with 8 respondents (72.7%) answering that it was not applicable, and 2 respondents (18.2%) answering that it was not at all applicable. The outcomes of the usability test (Table 5) of evaluation by component of the DA draft 1) ~ 5) were “Normal” or above for 90% of the respondents or higher for almost all items. Further, ≥80% respondents gave good evaluations for “understandable language,” “length,” and “content bias.” However, respondents gave the lowest evaluations for the item on “clarity of how the DA should be used.”\nThe result of comprehensibility test (N = 11).\nNumber of respondents/total number of respondents. No. 7 was unanswered by one.\nThe result of usability test (N = 20).\nQuestions 2 and 4 are reversed, number of respondents/total number of people.\nDA = decision aid.\nIn addition, in the free response section, the need for a table of contents, ingenuity to make it easier for patients and their families to imagine post-discharge daily life, weak expression of pros and cons, layout font, font size, and colors used were pointed out. There were also requests for providing additional information on the per-month costs of living in facilities and the types and roles of staff involved during hospital stay or after discharge. Furthermore, other respondents provided opinions such as “I believe that I will be able to make the best judgment for myself with staff support and explanations based on this DA,” “Wouldn’t the gap between values and reality cause problems?,” and “It would be hard to just be handed the DA and read and understand it by myself, so it would be better to look at it while somebody is explaining it to me.”\n[SUBTITLE] 3.4. Steering [SUBSECTION] To clarify the methods of use, an explanation on the instructions for use was added to the DA. The table of the pros and cons of the options was reorganized and improved. As the options were difficult to differentiate, the 2 options were named “Same place as before admission” and “Different place from before admission.” To make it easier for older adults to understand, the layout, organization, and terms used for the information were further modified into succinct and easily comprehensible expressions. Moreover, home renovations were especially important for people who wished to return to their homes to recover, so it was placed as a separate item to clearly present the values. Finally, a DA based on the 6 values of “ADL,” “Services and fees,” “Emergencies,” “Family support,” “Environment,” and “Home renovation” was created.\nThe DA that was revised again was distributed to one of each of the following persons: an older stroke patient, family member, physician, nurse, PT, OT, and MSW. They were then asked to check the entire document. However, only minor additions and revisions of wording were needed, and the DA was ultimately developed as a 12-page, A4 paper-sized document (Table 6).\nComments and revision regarding the prototype content.\nADL = activities of daily living, DA = decision aid.\nTo clarify the methods of use, an explanation on the instructions for use was added to the DA. The table of the pros and cons of the options was reorganized and improved. As the options were difficult to differentiate, the 2 options were named “Same place as before admission” and “Different place from before admission.” To make it easier for older adults to understand, the layout, organization, and terms used for the information were further modified into succinct and easily comprehensible expressions. Moreover, home renovations were especially important for people who wished to return to their homes to recover, so it was placed as a separate item to clearly present the values. Finally, a DA based on the 6 values of “ADL,” “Services and fees,” “Emergencies,” “Family support,” “Environment,” and “Home renovation” was created.\nThe DA that was revised again was distributed to one of each of the following persons: an older stroke patient, family member, physician, nurse, PT, OT, and MSW. They were then asked to check the entire document. However, only minor additions and revisions of wording were needed, and the DA was ultimately developed as a 12-page, A4 paper-sized document (Table 6).\nComments and revision regarding the prototype content.\nADL = activities of daily living, DA = decision aid.", "In cycle 1, the selection of items was conducted. First, as a result of the literature review, we found a total of 127 references, 72 in Japanese and 55 in English. There were 2 existing DAs on where to receive post-discharge care.[14,24] However, they targeted end-of-life and frail older adults, and none addressed stroke patients. There were 2 Cochrane reviews on the location of post-discharge care for older stroke patients. Ward et al[25] compared the benefits of home and hospital for the rehabilitation of older adults, and Boland et al[26] compared home with other locations for the care of older adults, but the pros and cons of the location for post-discharge care of older adults were not mentioned clearly. That is, the 2 existing DAs did not provide evidence related to the place of convalescence, so rather than presenting the advantages and disadvantages, they had incorporated things the user should know and made references to preferences and values. Through this review, we found evidence that research on where older stroke patients decide to receive post-discharge care is not established, and we thus decided to review the medical records of 103 older stroke patients aged ≥ 65 years in the participating facilities to investigate the factors influencing discharge destinations. The results showed that the factors that influenced the discharge destination were “ADL,” “eating disorder,” and “family’s preference for discharge destination.”\nFive values were identified: “Degree of independence in ADL,” “environment for convalescence,” “family relationship,” “disease management,” and “social resources and fees.” These were common values that affected the decision-making of older stroke patients and their families regarding the discharge destination (Table 3).\nQualitative synthesis.\n() Number of codes.\nIn addition, we selected items for the DA that would help clarify these 5 values and that we considered to be important information from the professional viewpoint. The contents were selected to include the following: contents of long-term care insurance services, consultation services, costs, home repairs, prospects during hospitalization, family wishes, ADL, and responses to illness.\nThe layout of the DA was designed to enhance legibility for older adults by using large font sizes, easy-to-read fonts, and clear contrasts. Furthermore, the DA had a 2-page format printed on a single folded sheet to make it user friendly for users who may have disabilities, such as memory impairment and aphasia, and was printed on a brochure format so that it could be used with family and clinicians. Kanji characters and illustrations were used with preference over hiragana and katakana phonetic letters, and items that required taking memos or writing were minimized.", "In Introduction of DA prototype Version 1, 3 discharge destinations options, that is, “home,” “residence/facility (for independent people),” and “hospital/facility (for people who need long-term care),” were listed, and how to use the DA, flow from admission to discharge, wishes and level of preparation, and the types of clinicians and distinction of their roles were described. In Facts and information, stroke, recurrence prophylaxis, long-term insurance, and features of the discharge destinations were described in detail. In Pros and cons of various options and clear expression of values and Weighing, the pros and cons of the 3 discharge destination options were printed next to the options to enable easy comparison as they related to the values that were commonly observed in older stroke patients and their families. Furthermore, a scale of “Not important 1 point” to “Very important 5 points” was used and placed vertically next to each item so that the level of importance could be compared easily. Items were added to Checking preparedness for decision-making to identify anybody other than the respondent who would help with their decision-making and who they wanted to do it with. The DA prototype Version 1 was completed on November 17, 2017, and the development group conducted a discussion to confirm that it met all the minimal criteria, that is, 12 items of the IPDASi.", "To be classified as a DA according to the 6 qualifying criteria of the IPDASi in the comprehensibility test (Table 4), all criteria must be met. The only criterion that all respondents said they met was “Available options.” In particular, 2 respondents (18.2%) answered no for “positive characteristics (pros),” “negative characteristics (cons),” and “experience of the consequence of the options.” The 6 certifying criteria of the IPDASi must be answered by “Applicable” or higher on the 4-level scale of evaluation to meet the certifying criteria. The only items for which all respondents answered “Applicable” or higher were “Production or publication date” and “Information about the funding source used for development.” The certifying criteria were met the least for “Information about the levels of uncertainty associated with event or outcome probabilities,” with 8 respondents (72.7%) answering that it was not applicable, and 2 respondents (18.2%) answering that it was not at all applicable. The outcomes of the usability test (Table 5) of evaluation by component of the DA draft 1) ~ 5) were “Normal” or above for 90% of the respondents or higher for almost all items. Further, ≥80% respondents gave good evaluations for “understandable language,” “length,” and “content bias.” However, respondents gave the lowest evaluations for the item on “clarity of how the DA should be used.”\nThe result of comprehensibility test (N = 11).\nNumber of respondents/total number of respondents. No. 7 was unanswered by one.\nThe result of usability test (N = 20).\nQuestions 2 and 4 are reversed, number of respondents/total number of people.\nDA = decision aid.\nIn addition, in the free response section, the need for a table of contents, ingenuity to make it easier for patients and their families to imagine post-discharge daily life, weak expression of pros and cons, layout font, font size, and colors used were pointed out. There were also requests for providing additional information on the per-month costs of living in facilities and the types and roles of staff involved during hospital stay or after discharge. Furthermore, other respondents provided opinions such as “I believe that I will be able to make the best judgment for myself with staff support and explanations based on this DA,” “Wouldn’t the gap between values and reality cause problems?,” and “It would be hard to just be handed the DA and read and understand it by myself, so it would be better to look at it while somebody is explaining it to me.”", "To clarify the methods of use, an explanation on the instructions for use was added to the DA. The table of the pros and cons of the options was reorganized and improved. As the options were difficult to differentiate, the 2 options were named “Same place as before admission” and “Different place from before admission.” To make it easier for older adults to understand, the layout, organization, and terms used for the information were further modified into succinct and easily comprehensible expressions. Moreover, home renovations were especially important for people who wished to return to their homes to recover, so it was placed as a separate item to clearly present the values. Finally, a DA based on the 6 values of “ADL,” “Services and fees,” “Emergencies,” “Family support,” “Environment,” and “Home renovation” was created.\nThe DA that was revised again was distributed to one of each of the following persons: an older stroke patient, family member, physician, nurse, PT, OT, and MSW. They were then asked to check the entire document. However, only minor additions and revisions of wording were needed, and the DA was ultimately developed as a 12-page, A4 paper-sized document (Table 6).\nComments and revision regarding the prototype content.\nADL = activities of daily living, DA = decision aid.", "This study aimed to develop a DA based on the values of older adults and their families to help older stroke patients choose where they wanted to receive post-discharge care. First, older stroke patients and their families were surveyed via an interview, multidisciplinary clinicians were surveyed via a questionnaire, and medical records were reviewed to extract the values and other data to base the DA design on. Next, a prototype was created, the IPDASi 12-item comprehensibility test and usability test were performed, and repeated revisions were made to perfect the DA. Herein, the process of development and evaluation of the DA using comprehensibility and usability tests are discussed.\nFirst, the ultimate users of the DA, that is, older stroke patients, families, and clinicians, all repeatedly participated in the process of development. The most challenging task in Japanese healthcare settings is decision support regarding discharge destinations, which is reported to consist of adjusting the “gaps in intentions” between old older adults and their families and even medical staff.[7] This can be attributed to the fact that older stroke patients are not participating much in the decision-making and that their values are instead being conveyed by their families or clinicians. Although their participation in the process of development is important, it is often difficult to involve frail older adults in practice, and it is recognized that there are many disagreements as to which stage is the most suitable for them to be involved in.[27] However, Dugas et al[11] stated the importance of involving the patients in the process of development to avoid stigmas and explained how it is helpful for highlighting the problems in the real world. The present DA incorporated the perspectives of older stroke patients, their families, and clinicians in the process of development, and the participation of the older stroke patients themselves is noteworthy, particularly in Japan. Japanese people value communication in indirect ways, such as the use of metaphors and hinting,[28] and believe that they do not need to express themselves to be understood by others, for example, older adults themselves often forego expressing their wishes clearly. Therefore, getting older stroke patients involved and having them participate repeatedly in the development allowed creating a DA that promotes mutual understanding with families and clinicians and is easy to accept for all parties. Moreover, having multidisciplinary clinicians participate repeatedly in the process of development was also notable. Discharge care in multidisciplinary teamwork in Japan is reported to be challenging due to disparities between staff in their discharge support skills and the lack of understanding of the functions and roles of partner facilities.[7] In addition, it has become clear that physicians, nurses, rehabilitation staff, and welfare staff all share this feeling of difficulty.[29] In this study, there was no difference in the answers of individuals with different occupations in the questionnaire survey for clinicians. Further, no particular discrepancy in the subsequent evaluation and confirmation was observed. This suggested that having a clear goal of decision-making on discharge destination and sharing opinions and compromising in each scenario would also increase the mutual understanding of various professional roles.\nSecond, the DA was developed based on the common values of older stroke patients and their families. The values related to discharge destination decision-making common to older stroke patients and their families in this study were “ADL,” “Services and fees,” “Emergencies,” “Family support,” “Environment,” and “Home renovation,” showing that there were actually many commonalities between the values of older stroke patients and their families. Garvelink et al[14] reported that “friends, family, or community members’ support,” “home renovations,” and “fees that the home, daily life, and services will cost” were important elements of frail older adults’ decisions on where to receive care. Murray[24] reported “Privacy,” “Self-management,” and “Relationships with family and pets” as important values for terminal-phase older adults in deciding where to receive additional care. The outcomes of our study were similar in that our respondents valued connections with family and the community. The damage caused by relocation is especially serious for older adults and is reported to comprise physical, mental, and social damages.[30] For older adults who have lived for a long time in environments that they have become acclimatized to, the discharge destination can have a greater impact than the change of location per se, so we considered that minimizing change and preventing isolation would ensure the smoothest path for the older stroke patients and their families to the lifestyle they envisioned. Furthermore, as a characteristic of the present case, ADL and responses to emergencies were also viewed as important. ADL[31] and medical acts[32] have already been known to affect older stroke patients’ discharge destination, which were consistent with the findings from the clinicians’ viewpoint. Niiyama[33] reported older stroke patients’ experiences related to changes to self-image and needing to depend on the help of others, such as “unable to move,” “receiving the help of others,” and “living with disability.” When suffering from a stroke, the anxiety that the acquired ability is impaired and may recur has a great impact on patients’ post-discharge life plans, suggesting that it is necessary to consider the characteristics of the disease.\nThird, although the majority of the respondents left positive responses in the evaluation of the internal validity of the DA, the evidence for the older stroke patients’ discharge destination remains vague. Evidence supporting that home or a facility is better for older adults’ convalescence, either in Japan or abroad, is lacking.[26] Therefore, IPDASi evaluations for positive and negative characteristics of the options, experience of the consequences, and information about the levels of uncertainty associated with event or outcome probabilities were low, even after identifying the factors associated with discharge destination from the medical record review. Furthermore, a DA that guides a rational decision-making process needs to present information on the positive and negative evidence for the options equally. However, our findings also suggested that the evaluators of this DA were not familiar with the DA itself as there are no established DA methods for Japanese healthcare settings. It is difficult for older adults who have suddenly suffered a stroke to read, understand, and make decisions based on the DA alone. Therefore, by using the DA, patients can organize evidence-based information that suits their values and make more satisfactory decisions in a short hospital stay. In the future, it is hoped that DAs will be used together with families and specialists. The quality and quantity of information and how to utilize it also warrant further research.\nA 12-page, A4-sized DA based on the 6 common values between older stroke patients and their families, that is, “ADL,” “Services and fees,” “Emergencies,” “Family support,” “Environment,” and “Home renovation,” was developed, and its internal validity was confirmed. The DA allows patients to sort out evidence-based information that matches their values and make more satisfying decisions within a short hospital stay. In the future, it is necessary to verify the effectiveness of the DA in clinical practice.\n[SUBTITLE] 4.1. Study limitations and future prospects [SUBSECTION] There were several limitations to this study. First, because feedback from so many different people, such as older stroke patients, their families, and various clinicians, was needed in the process of development, only feedback from a small number of each could be evaluated. Furthermore, as there were little data on where older adults receive post-discharge care and this data were supplemented by a review of medical records from a single institution, the data were not versatile and lacked generalizability. However, to the best of our knowledge, this is the first study that involved the participation of families, clinicians, and, most remarkably, older stroke patients themselves in the process of developing the DA from the preliminary survey of needs. In future, we plan to assign participants into 2 groups, one that is provided this DA and one that is not, to evaluate it in a randomized controlled trial.\nThere were several limitations to this study. First, because feedback from so many different people, such as older stroke patients, their families, and various clinicians, was needed in the process of development, only feedback from a small number of each could be evaluated. Furthermore, as there were little data on where older adults receive post-discharge care and this data were supplemented by a review of medical records from a single institution, the data were not versatile and lacked generalizability. However, to the best of our knowledge, this is the first study that involved the participation of families, clinicians, and, most remarkably, older stroke patients themselves in the process of developing the DA from the preliminary survey of needs. In future, we plan to assign participants into 2 groups, one that is provided this DA and one that is not, to evaluate it in a randomized controlled trial.", "There were several limitations to this study. First, because feedback from so many different people, such as older stroke patients, their families, and various clinicians, was needed in the process of development, only feedback from a small number of each could be evaluated. Furthermore, as there were little data on where older adults receive post-discharge care and this data were supplemented by a review of medical records from a single institution, the data were not versatile and lacked generalizability. However, to the best of our knowledge, this is the first study that involved the participation of families, clinicians, and, most remarkably, older stroke patients themselves in the process of developing the DA from the preliminary survey of needs. In future, we plan to assign participants into 2 groups, one that is provided this DA and one that is not, to evaluate it in a randomized controlled trial.", "We express our sincerest gratitude to the older adults, their families, and clinicians in the hospital for their participation, which made this study possible. This work was supported by KAKENHI Grant-in-Aid for Young Scientists (B) (15K20759) and Grant-in-Aid for Scientific Research (C) (19K11244). The authors of this work have nothing to disclose.", "YA designed the study and drafted the manuscript. KN provided advice on the analysis and study overall, as well as offering suggestions. All authors have consented to publication and have checked the final document.\nConceptualization: Yoriko Aoki, Kazuhiro Nakayama.\nData curation: Yoriko Aoki, Kazuhiro Nakayama.\nFormal analysis: Yoriko Aoki, Kazuhiro Nakayama.\nWriting – original draft: Yoriko Aoki.\nWriting – review & editing: Yoriko Aoki." ]
[ "intro", "methods", null, null, null, null, null, null, null, null, null, null, "results", null, null, null, null, "discussion", null, null, null ]
[ "case report", "decision aid", "discharge destination", "older stroke survivor" ]
Nationwide Survey for Pediatric Gastrostomy Tube Placement in Korea.
36254529
Various methods have been implemented for pediatric gastrostomy tube placement. We aimed to investigate the performance status of pediatric gastrostomy in South Korea and to present indications and appropriate methods for domestic situations.
BACKGROUND
A survey was conducted among pediatric endoscopists who performed upper gastrointestinal endoscopy in Korea. The questionnaire consisted of 16 questions on gastrostomy performance status.
METHODS
Among the 48 institutions where the survey was applied, 36 (75%) responded. Of the 36 institutions, gastrostomy was performed in 31 (86.1%). The departments in which gastrostomy was performed were pediatrics at 26 institutions (81.3%), surgery at 24 institutions (75.0%), internal medicine at 9 institutions (28.1%), and radiology at 7 institutions (21.9%). There were 18 institutions (66.7%) using the pull method for percutaneous endoscopic gastrostomy (PEG) and nine institutions (33.3%) using the push method. When performing gastrostomies, fundoplication procedures were performed in 19 institutions (61.3%), if deemed necessary. However, 12 institutions (38.7%) answered that gastrostomy was always implemented alone. Complications after gastrostomy included buried bumper syndrome, wound infection, leakage, tube migration, and incorrect opening site in the stomach, but the number of cases with complications was very small.
RESULTS
In Korea, a pediatric gastrostomy is implemented in various ways depending on the institution. Clinicians are concerned about choosing the most effective methods with fewer complications after the procedure. In our study, we reported only a few complications. Korea has good accessibility for pediatric gastrointestinal endoscopy, and this survey showed that it is a safe procedure that can be considered initially in pediatric gastrostomy. This study is expected to help to create optimal pediatric PEG guidelines in Korea.
CONCLUSION
[ "Child", "Enteral Nutrition", "Gastrostomy", "Humans", "Pediatrics", "Republic of Korea", "Surveys and Questionnaires" ]
9577357
INTRODUCTION
A gastrostomy tube is used when a nasogastric tube is required for more than 1 month to provide sufficient nutrition when oral feeding is impossible.1 It is already well known that nutrition through gastrostomy tubes improves malnutrition status of children with neurodisabilities or oncologic conditions, has a positive effect on prognosis, and induces longer survival.2 Gastrostomy tube placement can be performed with upper gastrointestinal (UGI) endoscopy or surgical methods. Some centers have implemented radiological methods that use fluoroscopy instead of UGI endoscopy. Recently, pediatric endoscopists in Korea have been performing percutaneous endoscopic gastrostomy (PEG) using UGI endoscopy. In the past, PEG was mainly performed using the pull method, and it was recommended to use it initially. Complications such as infection and dislodgement have been reported to be higher than with the surgical method. Recently, some institutions choose to use the push method in children.34 With the implementation of laparoscopy, the disadvantages of conventional open surgery have greatly improved.5 To date, PEG methods using UGI endoscopes have been recommended as a better way to implement them without complications and are easier than other methods for pediatric gastrostomy tube placement. However, if PEG procedures are difficult due to structural problems of the stomach or if fundoplication is required due to gastroesophageal reflux disease (GERD), a surgical method is required.2 Although various methods for gastrostomy tube replacement such as the pull-method PEG, push-method PEG, open surgery, laparoscopic surgery, and radiological methods have been implemented, large complications are likely associated with the individual characteristics of patients rather than the procedure itself. In this study, we aimed to investigate the performance status of pediatric PEG in South Korea and to report indications and appropriate methods for domestic situations. We also aimed to compare questionnaire variables according to different groups in order to examine associations between variables.
METHODS
[SUBTITLE] Survey [SUBSECTION] In October 2021, a survey was conducted at 48 institutions that conduct UGI endoscopy in South Korea. In the questionnaire, the availability of pediatric gastrostomies, the average number of procedures per year, and the method of implementation were investigated. In the case of PEG, the method of PEG (pull vs. push), the type of tube used, and the implementation method according to the weight of the child were also investigated. Further questions included whether replacement of the gastrostomy tube was possible at each institution, which department performed the replacement, whether gastroscopy was performed after gastrostomy, and whether fundoplication was performed with gastrostomy. Lastly, complications that occurred after gastrostomy, when the gastrostomy was removed, when re-surgery was conducted, whether the nutritional status of children actually improved, and whether the re-hospitalization rate was reduced by gastrostomy were investigated. We also compared questionnaire variables between groups divided according to the number of annually performed pediatric gastrostomies at each center, the specialist performing pediatric gastrostomies, and recently used PEG method. In October 2021, a survey was conducted at 48 institutions that conduct UGI endoscopy in South Korea. In the questionnaire, the availability of pediatric gastrostomies, the average number of procedures per year, and the method of implementation were investigated. In the case of PEG, the method of PEG (pull vs. push), the type of tube used, and the implementation method according to the weight of the child were also investigated. Further questions included whether replacement of the gastrostomy tube was possible at each institution, which department performed the replacement, whether gastroscopy was performed after gastrostomy, and whether fundoplication was performed with gastrostomy. Lastly, complications that occurred after gastrostomy, when the gastrostomy was removed, when re-surgery was conducted, whether the nutritional status of children actually improved, and whether the re-hospitalization rate was reduced by gastrostomy were investigated. We also compared questionnaire variables between groups divided according to the number of annually performed pediatric gastrostomies at each center, the specialist performing pediatric gastrostomies, and recently used PEG method. [SUBTITLE] Statistical analysis [SUBSECTION] For statistical comparisons between groups, the χ2 test or Fisher’s exact test was used for categorical variables. Comparative data for categorical variables were reported as numbers and percentage. Statistical significance was defined as a P value ≤ 0.05. All statistical analyses were performed using R version 3.2.3 (R Foundation, Vienna, Austria). For statistical comparisons between groups, the χ2 test or Fisher’s exact test was used for categorical variables. Comparative data for categorical variables were reported as numbers and percentage. Statistical significance was defined as a P value ≤ 0.05. All statistical analyses were performed using R version 3.2.3 (R Foundation, Vienna, Austria). [SUBTITLE] Ethics statement [SUBSECTION] This study was approved by the Institutional Review Board (IRB) of Jeonbuk National University Hospital, and informed consent was waived because the study was performed with an online questionnaire survey (IRB Number 2021-12-022). This study was approved by the Institutional Review Board (IRB) of Jeonbuk National University Hospital, and informed consent was waived because the study was performed with an online questionnaire survey (IRB Number 2021-12-022).
RESULTS
[SUBTITLE] Questionnaire answers [SUBSECTION] Of the 48 institutions where the survey was requested to be applied, 36 (75.0%) responded. The geographic locations of the 36 institutions are shown in Fig. 1. Of the 36 institutions, gastrostomy was performed in 31 (86.1%). Regarding the number of gastrostomies performed annually, 19 institutions (61.3%) answered that they performed fewer than five gastrostomies/year (Table 1). The gastrostomy was performed by only pediatric endoscopists at 7 institutions (22.6%), by other specialist only at 6 institutions (19.4%), and by pediatric endoscopists or other specialists at 18 institutions (58.1%). PEGs were performed in 27 institutions (87.1%). When performing PEGs, 13 institutions (48.1%) performed deep sedation, 7 institutions (25.9%) performed general anesthesia, and 7 institutions (25.9%) performed either deep sedation or general anesthesia. There were 18 institutions (66.7%) using the pull method for PEG and 9 institutions (33.3%) using the push method. High-profile balloon-based tubes were used in 6 institutions (22.2%), and 19 institutions (70.4%) used high-profile plastic-based tubes. Two institutions (7.4%) used low-profile balloon-based tubes (Table 1). PEG = percutaneous endoscopic gastrostomy, SG = surgical gastrostomy, RG = radiologic gastrostomy, EGD = esophagogastroduodenoscopy. When asked if children under 10 kg underwent gastrostomies, 13 institutions (48.1%) answered yes. The replacement of gastrostomy tubes was performed by only pediatric endoscopists at 15 institutions (48.4%), by other specialist only at 7 institutions (22.6%), and by pediatric endoscopists or other specialists at 9 institutions (29.0%). When asked whether esophagogastroduodenoscopy (EGD) was conducted at least once after gastrostomy, 21 institutions (67.7%) answered that they had never performed it. When performing gastrostomies, fundoplication procedures to prevent GERD were performed in 19 institutions (61.3%) if deemed necessary. However, 12 institutions (38.7%) answered that gastrostomy was always implemented alone (Table 1). When asked whether they experienced a situation in which the gastrostomy tube had to be removed after gastrostomy, 3 institutions (9.7%) answered yes. The reasons for gastrostomy tube removal were: oral nutrition became possible and therefore, the tube was no longer necessary, buried bumper syndrome, and the general rigidity became too severe to use the gastrostomy tube. When asked whether they experienced reoperation after gastrostomy, 5 institutions (16.1%) answered yes. The reasons for these were buried bumper syndrome in 2 cases, site infection in 1 case, and tube migration in 1 case, and incorrect tube location after open surgery, leading to feeding problems in 1 case. When asked whether gastrostomy tube treatment was thought to improve patients’ nutritional status, 31 institutions (96.8%) answered yes. When asked whether gastrostomy tube treatment seemed to reduce the patient’s hospitalization rate, 27 institutions (87.1%) answered yes (Table 1). Of the 48 institutions where the survey was requested to be applied, 36 (75.0%) responded. The geographic locations of the 36 institutions are shown in Fig. 1. Of the 36 institutions, gastrostomy was performed in 31 (86.1%). Regarding the number of gastrostomies performed annually, 19 institutions (61.3%) answered that they performed fewer than five gastrostomies/year (Table 1). The gastrostomy was performed by only pediatric endoscopists at 7 institutions (22.6%), by other specialist only at 6 institutions (19.4%), and by pediatric endoscopists or other specialists at 18 institutions (58.1%). PEGs were performed in 27 institutions (87.1%). When performing PEGs, 13 institutions (48.1%) performed deep sedation, 7 institutions (25.9%) performed general anesthesia, and 7 institutions (25.9%) performed either deep sedation or general anesthesia. There were 18 institutions (66.7%) using the pull method for PEG and 9 institutions (33.3%) using the push method. High-profile balloon-based tubes were used in 6 institutions (22.2%), and 19 institutions (70.4%) used high-profile plastic-based tubes. Two institutions (7.4%) used low-profile balloon-based tubes (Table 1). PEG = percutaneous endoscopic gastrostomy, SG = surgical gastrostomy, RG = radiologic gastrostomy, EGD = esophagogastroduodenoscopy. When asked if children under 10 kg underwent gastrostomies, 13 institutions (48.1%) answered yes. The replacement of gastrostomy tubes was performed by only pediatric endoscopists at 15 institutions (48.4%), by other specialist only at 7 institutions (22.6%), and by pediatric endoscopists or other specialists at 9 institutions (29.0%). When asked whether esophagogastroduodenoscopy (EGD) was conducted at least once after gastrostomy, 21 institutions (67.7%) answered that they had never performed it. When performing gastrostomies, fundoplication procedures to prevent GERD were performed in 19 institutions (61.3%) if deemed necessary. However, 12 institutions (38.7%) answered that gastrostomy was always implemented alone (Table 1). When asked whether they experienced a situation in which the gastrostomy tube had to be removed after gastrostomy, 3 institutions (9.7%) answered yes. The reasons for gastrostomy tube removal were: oral nutrition became possible and therefore, the tube was no longer necessary, buried bumper syndrome, and the general rigidity became too severe to use the gastrostomy tube. When asked whether they experienced reoperation after gastrostomy, 5 institutions (16.1%) answered yes. The reasons for these were buried bumper syndrome in 2 cases, site infection in 1 case, and tube migration in 1 case, and incorrect tube location after open surgery, leading to feeding problems in 1 case. When asked whether gastrostomy tube treatment was thought to improve patients’ nutritional status, 31 institutions (96.8%) answered yes. When asked whether gastrostomy tube treatment seemed to reduce the patient’s hospitalization rate, 27 institutions (87.1%) answered yes (Table 1). [SUBTITLE] Comparison between questionnaire variables between groups [SUBSECTION] According to the comparison between the groups divided according to the number of to the number of annually performed pediatric gastrostomies at each center, the rate of PEG performance in children less than 10 kg was significantly higher in centers that had answered that they were conducting ≥ 5 gastrostomies per year than those that had answered that they were conducting < 5 gastrostomies per year (77.8% vs. 33.3%, P = 0.046). Complications related with gastrostomies were also significantly higher in in centers that had answered that they were conducting ≥ 5 gastrostomies per year than those that had answered that they were conducting < 5 gastrostomies per year (41.7% vs. 5.3%, P = 0.022) (Table 2). Values are presented as number (%). PEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy. According to the comparison between groups divided according to the specialist performing pediatric gastrostomies, no significant difference in questionnaire variables were observed between the “pediatric endoscopist only” group and the “other specialists" group (Table 3). Values are presented as number (%). PEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy. According to the comparison between groups divided according to the recently used PEG method, PEG tube preference was statistically significant between the ‘pull method’ group and the ‘push method’ group (P = 0.006). Those using the pull method preferred the high profile plastic based tube (88.9%), while those using the push method preferred the high profile balloon based tube (44.4%) (Table 4). Values are presented as number (%). PEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy. According to the comparison between the groups divided according to the number of to the number of annually performed pediatric gastrostomies at each center, the rate of PEG performance in children less than 10 kg was significantly higher in centers that had answered that they were conducting ≥ 5 gastrostomies per year than those that had answered that they were conducting < 5 gastrostomies per year (77.8% vs. 33.3%, P = 0.046). Complications related with gastrostomies were also significantly higher in in centers that had answered that they were conducting ≥ 5 gastrostomies per year than those that had answered that they were conducting < 5 gastrostomies per year (41.7% vs. 5.3%, P = 0.022) (Table 2). Values are presented as number (%). PEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy. According to the comparison between groups divided according to the specialist performing pediatric gastrostomies, no significant difference in questionnaire variables were observed between the “pediatric endoscopist only” group and the “other specialists" group (Table 3). Values are presented as number (%). PEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy. According to the comparison between groups divided according to the recently used PEG method, PEG tube preference was statistically significant between the ‘pull method’ group and the ‘push method’ group (P = 0.006). Those using the pull method preferred the high profile plastic based tube (88.9%), while those using the push method preferred the high profile balloon based tube (44.4%) (Table 4). Values are presented as number (%). PEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy.
null
null
[ "Survey", "Statistical analysis", "Questionnaire answers", "Comparison between questionnaire variables between groups" ]
[ "In October 2021, a survey was conducted at 48 institutions that conduct UGI endoscopy in South Korea. In the questionnaire, the availability of pediatric gastrostomies, the average number of procedures per year, and the method of implementation were investigated. In the case of PEG, the method of PEG (pull vs. push), the type of tube used, and the implementation method according to the weight of the child were also investigated. Further questions included whether replacement of the gastrostomy tube was possible at each institution, which department performed the replacement, whether gastroscopy was performed after gastrostomy, and whether fundoplication was performed with gastrostomy. Lastly, complications that occurred after gastrostomy, when the gastrostomy was removed, when re-surgery was conducted, whether the nutritional status of children actually improved, and whether the re-hospitalization rate was reduced by gastrostomy were investigated.\nWe also compared questionnaire variables between groups divided according to the number of annually performed pediatric gastrostomies at each center, the specialist performing pediatric gastrostomies, and recently used PEG method.", "For statistical comparisons between groups, the χ2 test or Fisher’s exact test was used for categorical variables. Comparative data for categorical variables were reported as numbers and percentage. Statistical significance was defined as a P value ≤ 0.05. All statistical analyses were performed using R version 3.2.3 (R Foundation, Vienna, Austria).", "Of the 48 institutions where the survey was requested to be applied, 36 (75.0%) responded. The geographic locations of the 36 institutions are shown in Fig. 1. Of the 36 institutions, gastrostomy was performed in 31 (86.1%). Regarding the number of gastrostomies performed annually, 19 institutions (61.3%) answered that they performed fewer than five gastrostomies/year (Table 1). The gastrostomy was performed by only pediatric endoscopists at 7 institutions (22.6%), by other specialist only at 6 institutions (19.4%), and by pediatric endoscopists or other specialists at 18 institutions (58.1%). PEGs were performed in 27 institutions (87.1%). When performing PEGs, 13 institutions (48.1%) performed deep sedation, 7 institutions (25.9%) performed general anesthesia, and 7 institutions (25.9%) performed either deep sedation or general anesthesia. There were 18 institutions (66.7%) using the pull method for PEG and 9 institutions (33.3%) using the push method. High-profile balloon-based tubes were used in 6 institutions (22.2%), and 19 institutions (70.4%) used high-profile plastic-based tubes. Two institutions (7.4%) used low-profile balloon-based tubes (Table 1).\nPEG = percutaneous endoscopic gastrostomy, SG = surgical gastrostomy, RG = radiologic gastrostomy, EGD = esophagogastroduodenoscopy.\nWhen asked if children under 10 kg underwent gastrostomies, 13 institutions (48.1%) answered yes. The replacement of gastrostomy tubes was performed by only pediatric endoscopists at 15 institutions (48.4%), by other specialist only at 7 institutions (22.6%), and by pediatric endoscopists or other specialists at 9 institutions (29.0%). When asked whether esophagogastroduodenoscopy (EGD) was conducted at least once after gastrostomy, 21 institutions (67.7%) answered that they had never performed it. When performing gastrostomies, fundoplication procedures to prevent GERD were performed in 19 institutions (61.3%) if deemed necessary. However, 12 institutions (38.7%) answered that gastrostomy was always implemented alone (Table 1).\nWhen asked whether they experienced a situation in which the gastrostomy tube had to be removed after gastrostomy, 3 institutions (9.7%) answered yes. The reasons for gastrostomy tube removal were: oral nutrition became possible and therefore, the tube was no longer necessary, buried bumper syndrome, and the general rigidity became too severe to use the gastrostomy tube. When asked whether they experienced reoperation after gastrostomy, 5 institutions (16.1%) answered yes. The reasons for these were buried bumper syndrome in 2 cases, site infection in 1 case, and tube migration in 1 case, and incorrect tube location after open surgery, leading to feeding problems in 1 case. When asked whether gastrostomy tube treatment was thought to improve patients’ nutritional status, 31 institutions (96.8%) answered yes. When asked whether gastrostomy tube treatment seemed to reduce the patient’s hospitalization rate, 27 institutions (87.1%) answered yes (Table 1).", "According to the comparison between the groups divided according to the number of to the number of annually performed pediatric gastrostomies at each center, the rate of PEG performance in children less than 10 kg was significantly higher in centers that had answered that they were conducting ≥ 5 gastrostomies per year than those that had answered that they were conducting < 5 gastrostomies per year (77.8% vs. 33.3%, P = 0.046). Complications related with gastrostomies were also significantly higher in in centers that had answered that they were conducting ≥ 5 gastrostomies per year than those that had answered that they were conducting < 5 gastrostomies per year (41.7% vs. 5.3%, P = 0.022) (Table 2).\nValues are presented as number (%).\nPEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy.\nAccording to the comparison between groups divided according to the specialist performing pediatric gastrostomies, no significant difference in questionnaire variables were observed between the “pediatric endoscopist only” group and the “other specialists\" group (Table 3).\nValues are presented as number (%).\nPEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy.\nAccording to the comparison between groups divided according to the recently used PEG method, PEG tube preference was statistically significant between the ‘pull method’ group and the ‘push method’ group (P = 0.006). Those using the pull method preferred the high profile plastic based tube (88.9%), while those using the push method preferred the high profile balloon based tube (44.4%) (Table 4).\nValues are presented as number (%).\nPEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy." ]
[ null, null, null, null ]
[ "INTRODUCTION", "METHODS", "Survey", "Statistical analysis", "Ethics statement", "RESULTS", "Questionnaire answers", "Comparison between questionnaire variables between groups", "DISCUSSION" ]
[ "A gastrostomy tube is used when a nasogastric tube is required for more than 1 month to provide sufficient nutrition when oral feeding is impossible.1 It is already well known that nutrition through gastrostomy tubes improves malnutrition status of children with neurodisabilities or oncologic conditions, has a positive effect on prognosis, and induces longer survival.2 Gastrostomy tube placement can be performed with upper gastrointestinal (UGI) endoscopy or surgical methods. Some centers have implemented radiological methods that use fluoroscopy instead of UGI endoscopy. Recently, pediatric endoscopists in Korea have been performing percutaneous endoscopic gastrostomy (PEG) using UGI endoscopy. In the past, PEG was mainly performed using the pull method, and it was recommended to use it initially. Complications such as infection and dislodgement have been reported to be higher than with the surgical method. Recently, some institutions choose to use the push method in children.34\nWith the implementation of laparoscopy, the disadvantages of conventional open surgery have greatly improved.5 To date, PEG methods using UGI endoscopes have been recommended as a better way to implement them without complications and are easier than other methods for pediatric gastrostomy tube placement. However, if PEG procedures are difficult due to structural problems of the stomach or if fundoplication is required due to gastroesophageal reflux disease (GERD), a surgical method is required.2 Although various methods for gastrostomy tube replacement such as the pull-method PEG, push-method PEG, open surgery, laparoscopic surgery, and radiological methods have been implemented, large complications are likely associated with the individual characteristics of patients rather than the procedure itself.\nIn this study, we aimed to investigate the performance status of pediatric PEG in South Korea and to report indications and appropriate methods for domestic situations. We also aimed to compare questionnaire variables according to different groups in order to examine associations between variables.", "[SUBTITLE] Survey [SUBSECTION] In October 2021, a survey was conducted at 48 institutions that conduct UGI endoscopy in South Korea. In the questionnaire, the availability of pediatric gastrostomies, the average number of procedures per year, and the method of implementation were investigated. In the case of PEG, the method of PEG (pull vs. push), the type of tube used, and the implementation method according to the weight of the child were also investigated. Further questions included whether replacement of the gastrostomy tube was possible at each institution, which department performed the replacement, whether gastroscopy was performed after gastrostomy, and whether fundoplication was performed with gastrostomy. Lastly, complications that occurred after gastrostomy, when the gastrostomy was removed, when re-surgery was conducted, whether the nutritional status of children actually improved, and whether the re-hospitalization rate was reduced by gastrostomy were investigated.\nWe also compared questionnaire variables between groups divided according to the number of annually performed pediatric gastrostomies at each center, the specialist performing pediatric gastrostomies, and recently used PEG method.\nIn October 2021, a survey was conducted at 48 institutions that conduct UGI endoscopy in South Korea. In the questionnaire, the availability of pediatric gastrostomies, the average number of procedures per year, and the method of implementation were investigated. In the case of PEG, the method of PEG (pull vs. push), the type of tube used, and the implementation method according to the weight of the child were also investigated. Further questions included whether replacement of the gastrostomy tube was possible at each institution, which department performed the replacement, whether gastroscopy was performed after gastrostomy, and whether fundoplication was performed with gastrostomy. Lastly, complications that occurred after gastrostomy, when the gastrostomy was removed, when re-surgery was conducted, whether the nutritional status of children actually improved, and whether the re-hospitalization rate was reduced by gastrostomy were investigated.\nWe also compared questionnaire variables between groups divided according to the number of annually performed pediatric gastrostomies at each center, the specialist performing pediatric gastrostomies, and recently used PEG method.\n[SUBTITLE] Statistical analysis [SUBSECTION] For statistical comparisons between groups, the χ2 test or Fisher’s exact test was used for categorical variables. Comparative data for categorical variables were reported as numbers and percentage. Statistical significance was defined as a P value ≤ 0.05. All statistical analyses were performed using R version 3.2.3 (R Foundation, Vienna, Austria).\nFor statistical comparisons between groups, the χ2 test or Fisher’s exact test was used for categorical variables. Comparative data for categorical variables were reported as numbers and percentage. Statistical significance was defined as a P value ≤ 0.05. All statistical analyses were performed using R version 3.2.3 (R Foundation, Vienna, Austria).\n[SUBTITLE] Ethics statement [SUBSECTION] This study was approved by the Institutional Review Board (IRB) of Jeonbuk National University Hospital, and informed consent was waived because the study was performed with an online questionnaire survey (IRB Number 2021-12-022).\nThis study was approved by the Institutional Review Board (IRB) of Jeonbuk National University Hospital, and informed consent was waived because the study was performed with an online questionnaire survey (IRB Number 2021-12-022).", "In October 2021, a survey was conducted at 48 institutions that conduct UGI endoscopy in South Korea. In the questionnaire, the availability of pediatric gastrostomies, the average number of procedures per year, and the method of implementation were investigated. In the case of PEG, the method of PEG (pull vs. push), the type of tube used, and the implementation method according to the weight of the child were also investigated. Further questions included whether replacement of the gastrostomy tube was possible at each institution, which department performed the replacement, whether gastroscopy was performed after gastrostomy, and whether fundoplication was performed with gastrostomy. Lastly, complications that occurred after gastrostomy, when the gastrostomy was removed, when re-surgery was conducted, whether the nutritional status of children actually improved, and whether the re-hospitalization rate was reduced by gastrostomy were investigated.\nWe also compared questionnaire variables between groups divided according to the number of annually performed pediatric gastrostomies at each center, the specialist performing pediatric gastrostomies, and recently used PEG method.", "For statistical comparisons between groups, the χ2 test or Fisher’s exact test was used for categorical variables. Comparative data for categorical variables were reported as numbers and percentage. Statistical significance was defined as a P value ≤ 0.05. All statistical analyses were performed using R version 3.2.3 (R Foundation, Vienna, Austria).", "This study was approved by the Institutional Review Board (IRB) of Jeonbuk National University Hospital, and informed consent was waived because the study was performed with an online questionnaire survey (IRB Number 2021-12-022).", "[SUBTITLE] Questionnaire answers [SUBSECTION] Of the 48 institutions where the survey was requested to be applied, 36 (75.0%) responded. The geographic locations of the 36 institutions are shown in Fig. 1. Of the 36 institutions, gastrostomy was performed in 31 (86.1%). Regarding the number of gastrostomies performed annually, 19 institutions (61.3%) answered that they performed fewer than five gastrostomies/year (Table 1). The gastrostomy was performed by only pediatric endoscopists at 7 institutions (22.6%), by other specialist only at 6 institutions (19.4%), and by pediatric endoscopists or other specialists at 18 institutions (58.1%). PEGs were performed in 27 institutions (87.1%). When performing PEGs, 13 institutions (48.1%) performed deep sedation, 7 institutions (25.9%) performed general anesthesia, and 7 institutions (25.9%) performed either deep sedation or general anesthesia. There were 18 institutions (66.7%) using the pull method for PEG and 9 institutions (33.3%) using the push method. High-profile balloon-based tubes were used in 6 institutions (22.2%), and 19 institutions (70.4%) used high-profile plastic-based tubes. Two institutions (7.4%) used low-profile balloon-based tubes (Table 1).\nPEG = percutaneous endoscopic gastrostomy, SG = surgical gastrostomy, RG = radiologic gastrostomy, EGD = esophagogastroduodenoscopy.\nWhen asked if children under 10 kg underwent gastrostomies, 13 institutions (48.1%) answered yes. The replacement of gastrostomy tubes was performed by only pediatric endoscopists at 15 institutions (48.4%), by other specialist only at 7 institutions (22.6%), and by pediatric endoscopists or other specialists at 9 institutions (29.0%). When asked whether esophagogastroduodenoscopy (EGD) was conducted at least once after gastrostomy, 21 institutions (67.7%) answered that they had never performed it. When performing gastrostomies, fundoplication procedures to prevent GERD were performed in 19 institutions (61.3%) if deemed necessary. However, 12 institutions (38.7%) answered that gastrostomy was always implemented alone (Table 1).\nWhen asked whether they experienced a situation in which the gastrostomy tube had to be removed after gastrostomy, 3 institutions (9.7%) answered yes. The reasons for gastrostomy tube removal were: oral nutrition became possible and therefore, the tube was no longer necessary, buried bumper syndrome, and the general rigidity became too severe to use the gastrostomy tube. When asked whether they experienced reoperation after gastrostomy, 5 institutions (16.1%) answered yes. The reasons for these were buried bumper syndrome in 2 cases, site infection in 1 case, and tube migration in 1 case, and incorrect tube location after open surgery, leading to feeding problems in 1 case. When asked whether gastrostomy tube treatment was thought to improve patients’ nutritional status, 31 institutions (96.8%) answered yes. When asked whether gastrostomy tube treatment seemed to reduce the patient’s hospitalization rate, 27 institutions (87.1%) answered yes (Table 1).\nOf the 48 institutions where the survey was requested to be applied, 36 (75.0%) responded. The geographic locations of the 36 institutions are shown in Fig. 1. Of the 36 institutions, gastrostomy was performed in 31 (86.1%). Regarding the number of gastrostomies performed annually, 19 institutions (61.3%) answered that they performed fewer than five gastrostomies/year (Table 1). The gastrostomy was performed by only pediatric endoscopists at 7 institutions (22.6%), by other specialist only at 6 institutions (19.4%), and by pediatric endoscopists or other specialists at 18 institutions (58.1%). PEGs were performed in 27 institutions (87.1%). When performing PEGs, 13 institutions (48.1%) performed deep sedation, 7 institutions (25.9%) performed general anesthesia, and 7 institutions (25.9%) performed either deep sedation or general anesthesia. There were 18 institutions (66.7%) using the pull method for PEG and 9 institutions (33.3%) using the push method. High-profile balloon-based tubes were used in 6 institutions (22.2%), and 19 institutions (70.4%) used high-profile plastic-based tubes. Two institutions (7.4%) used low-profile balloon-based tubes (Table 1).\nPEG = percutaneous endoscopic gastrostomy, SG = surgical gastrostomy, RG = radiologic gastrostomy, EGD = esophagogastroduodenoscopy.\nWhen asked if children under 10 kg underwent gastrostomies, 13 institutions (48.1%) answered yes. The replacement of gastrostomy tubes was performed by only pediatric endoscopists at 15 institutions (48.4%), by other specialist only at 7 institutions (22.6%), and by pediatric endoscopists or other specialists at 9 institutions (29.0%). When asked whether esophagogastroduodenoscopy (EGD) was conducted at least once after gastrostomy, 21 institutions (67.7%) answered that they had never performed it. When performing gastrostomies, fundoplication procedures to prevent GERD were performed in 19 institutions (61.3%) if deemed necessary. However, 12 institutions (38.7%) answered that gastrostomy was always implemented alone (Table 1).\nWhen asked whether they experienced a situation in which the gastrostomy tube had to be removed after gastrostomy, 3 institutions (9.7%) answered yes. The reasons for gastrostomy tube removal were: oral nutrition became possible and therefore, the tube was no longer necessary, buried bumper syndrome, and the general rigidity became too severe to use the gastrostomy tube. When asked whether they experienced reoperation after gastrostomy, 5 institutions (16.1%) answered yes. The reasons for these were buried bumper syndrome in 2 cases, site infection in 1 case, and tube migration in 1 case, and incorrect tube location after open surgery, leading to feeding problems in 1 case. When asked whether gastrostomy tube treatment was thought to improve patients’ nutritional status, 31 institutions (96.8%) answered yes. When asked whether gastrostomy tube treatment seemed to reduce the patient’s hospitalization rate, 27 institutions (87.1%) answered yes (Table 1).\n[SUBTITLE] Comparison between questionnaire variables between groups [SUBSECTION] According to the comparison between the groups divided according to the number of to the number of annually performed pediatric gastrostomies at each center, the rate of PEG performance in children less than 10 kg was significantly higher in centers that had answered that they were conducting ≥ 5 gastrostomies per year than those that had answered that they were conducting < 5 gastrostomies per year (77.8% vs. 33.3%, P = 0.046). Complications related with gastrostomies were also significantly higher in in centers that had answered that they were conducting ≥ 5 gastrostomies per year than those that had answered that they were conducting < 5 gastrostomies per year (41.7% vs. 5.3%, P = 0.022) (Table 2).\nValues are presented as number (%).\nPEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy.\nAccording to the comparison between groups divided according to the specialist performing pediatric gastrostomies, no significant difference in questionnaire variables were observed between the “pediatric endoscopist only” group and the “other specialists\" group (Table 3).\nValues are presented as number (%).\nPEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy.\nAccording to the comparison between groups divided according to the recently used PEG method, PEG tube preference was statistically significant between the ‘pull method’ group and the ‘push method’ group (P = 0.006). Those using the pull method preferred the high profile plastic based tube (88.9%), while those using the push method preferred the high profile balloon based tube (44.4%) (Table 4).\nValues are presented as number (%).\nPEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy.\nAccording to the comparison between the groups divided according to the number of to the number of annually performed pediatric gastrostomies at each center, the rate of PEG performance in children less than 10 kg was significantly higher in centers that had answered that they were conducting ≥ 5 gastrostomies per year than those that had answered that they were conducting < 5 gastrostomies per year (77.8% vs. 33.3%, P = 0.046). Complications related with gastrostomies were also significantly higher in in centers that had answered that they were conducting ≥ 5 gastrostomies per year than those that had answered that they were conducting < 5 gastrostomies per year (41.7% vs. 5.3%, P = 0.022) (Table 2).\nValues are presented as number (%).\nPEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy.\nAccording to the comparison between groups divided according to the specialist performing pediatric gastrostomies, no significant difference in questionnaire variables were observed between the “pediatric endoscopist only” group and the “other specialists\" group (Table 3).\nValues are presented as number (%).\nPEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy.\nAccording to the comparison between groups divided according to the recently used PEG method, PEG tube preference was statistically significant between the ‘pull method’ group and the ‘push method’ group (P = 0.006). Those using the pull method preferred the high profile plastic based tube (88.9%), while those using the push method preferred the high profile balloon based tube (44.4%) (Table 4).\nValues are presented as number (%).\nPEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy.", "Of the 48 institutions where the survey was requested to be applied, 36 (75.0%) responded. The geographic locations of the 36 institutions are shown in Fig. 1. Of the 36 institutions, gastrostomy was performed in 31 (86.1%). Regarding the number of gastrostomies performed annually, 19 institutions (61.3%) answered that they performed fewer than five gastrostomies/year (Table 1). The gastrostomy was performed by only pediatric endoscopists at 7 institutions (22.6%), by other specialist only at 6 institutions (19.4%), and by pediatric endoscopists or other specialists at 18 institutions (58.1%). PEGs were performed in 27 institutions (87.1%). When performing PEGs, 13 institutions (48.1%) performed deep sedation, 7 institutions (25.9%) performed general anesthesia, and 7 institutions (25.9%) performed either deep sedation or general anesthesia. There were 18 institutions (66.7%) using the pull method for PEG and 9 institutions (33.3%) using the push method. High-profile balloon-based tubes were used in 6 institutions (22.2%), and 19 institutions (70.4%) used high-profile plastic-based tubes. Two institutions (7.4%) used low-profile balloon-based tubes (Table 1).\nPEG = percutaneous endoscopic gastrostomy, SG = surgical gastrostomy, RG = radiologic gastrostomy, EGD = esophagogastroduodenoscopy.\nWhen asked if children under 10 kg underwent gastrostomies, 13 institutions (48.1%) answered yes. The replacement of gastrostomy tubes was performed by only pediatric endoscopists at 15 institutions (48.4%), by other specialist only at 7 institutions (22.6%), and by pediatric endoscopists or other specialists at 9 institutions (29.0%). When asked whether esophagogastroduodenoscopy (EGD) was conducted at least once after gastrostomy, 21 institutions (67.7%) answered that they had never performed it. When performing gastrostomies, fundoplication procedures to prevent GERD were performed in 19 institutions (61.3%) if deemed necessary. However, 12 institutions (38.7%) answered that gastrostomy was always implemented alone (Table 1).\nWhen asked whether they experienced a situation in which the gastrostomy tube had to be removed after gastrostomy, 3 institutions (9.7%) answered yes. The reasons for gastrostomy tube removal were: oral nutrition became possible and therefore, the tube was no longer necessary, buried bumper syndrome, and the general rigidity became too severe to use the gastrostomy tube. When asked whether they experienced reoperation after gastrostomy, 5 institutions (16.1%) answered yes. The reasons for these were buried bumper syndrome in 2 cases, site infection in 1 case, and tube migration in 1 case, and incorrect tube location after open surgery, leading to feeding problems in 1 case. When asked whether gastrostomy tube treatment was thought to improve patients’ nutritional status, 31 institutions (96.8%) answered yes. When asked whether gastrostomy tube treatment seemed to reduce the patient’s hospitalization rate, 27 institutions (87.1%) answered yes (Table 1).", "According to the comparison between the groups divided according to the number of to the number of annually performed pediatric gastrostomies at each center, the rate of PEG performance in children less than 10 kg was significantly higher in centers that had answered that they were conducting ≥ 5 gastrostomies per year than those that had answered that they were conducting < 5 gastrostomies per year (77.8% vs. 33.3%, P = 0.046). Complications related with gastrostomies were also significantly higher in in centers that had answered that they were conducting ≥ 5 gastrostomies per year than those that had answered that they were conducting < 5 gastrostomies per year (41.7% vs. 5.3%, P = 0.022) (Table 2).\nValues are presented as number (%).\nPEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy.\nAccording to the comparison between groups divided according to the specialist performing pediatric gastrostomies, no significant difference in questionnaire variables were observed between the “pediatric endoscopist only” group and the “other specialists\" group (Table 3).\nValues are presented as number (%).\nPEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy.\nAccording to the comparison between groups divided according to the recently used PEG method, PEG tube preference was statistically significant between the ‘pull method’ group and the ‘push method’ group (P = 0.006). Those using the pull method preferred the high profile plastic based tube (88.9%), while those using the push method preferred the high profile balloon based tube (44.4%) (Table 4).\nValues are presented as number (%).\nPEG = percutaneous endoscopic gastrostomy, EGD = esophagogastroduodenoscopy.", "There are very few endoscopy centers in Korea where pediatric endoscopy is possible. The demand for pediatric gastrostomy is also not high compared to that of adults, but it is an important procedure from the perspective of the nutritional treatment of patients. Through this survey, we observed that although there are not many cases of pediatric gastrostomy in Korea, pediatric PEG and pediatric surgical gastrostomy (SG) are performed to a similar extent. Some centers perform PEG with the help of endoscopists from the Department of Internal Medicine, and very rarely perform gastrostomy by imaging. Gastrostomy can be performed in various ways because there is no treatment method that has enough advantages to give absolute priority. With the development of gastrostomy methods, there are fewer complications, and it can be performed without difficulty with less time and cost. Some studies have reported that gastrostomy was performed safely with the collaboration of endoscopists and surgeons, and that could decrease the risks for each method.6\nGastrostomy was performed using only SG until 1980. PEG was initiated by Ponsky, a surgeon specialized in adults, who developed the pull method. It is referred to as the pull method because the thread from the abdominal skin is removed from the mouth using an endoscope, and the gastrostomy tube is tied and then pulled outward from the abdomen and held in position.7 Although the procedure time is short owing to the simplicity of the method, it cannot be used if the esophagus is narrow because the gastrostomy tube passes through the mouth to the esophagus. In addition, it is associated with a risk of infection. Therefore, if a patient has a narrow esophagus or a high risk of infection, other methods should be considered first.28 In addition, the pull method requires the use of a gastrostomy tube made of a plastic bumper. In this survey, 70.4% of institutions were found to use high-profile plastic-based tubes, where the pull method was performed, and buried bumper syndrome occurred only in institutions using the pull method. Buried bumper syndrome occurs when the bumper digs into the abdominal wall and dislodgement occurs and is thought to be a consequence of the plastic bumper part of the tube rather than the procedure method.3\nThe push method uses an introducer that performs gastropexy from the abdominal skin and inserts and mounts the gastrostomy tube directly. EGD is performed, but because the gastrostomy tube does not pass through the mouth and esophagus, it can be performed even if the esophagus is narrow, and reduce the risk of infection, compared to the pull method.89 In this study, among the 7 out of 9 institutions that answered that the main method of implementation was the push method using an introducer, 7 institutions used the high-profile balloon-based tube from the time of the procedure, while 2 institutions used the low-profile balloon-based tube from the beginning. Although rare, using a balloon-based tube is more appropriate for preventing buried bumper syndrome.9 Some studies have reported that the bleeding rate is high in the push method, includes procedures such as gastropexy or introducers on the abdomen.10 However, a small-scale study comparing the pull and push methods in adults in Korea reported that there was no significant difference in infection or bleeding, and no buried bumper syndrome was reported.11 Another study comparing outcomes between primary gastrostomy tubes and buttons (G-tube and G-button) in pediatric patients reported that primary G-tube offers no significant advantage in overall, minor or major complications when compared to primary G-button.12 Large-scale studies comparing pull and push methods are required.\nIn this study, 61.3% of the procedures were performed together with fundoplication when required. SG is conducted when fundoplication is performed too or when PEG is expected to be difficult due to structural problems.2 SG may be advantageous for the operator because it is performed under general anesthesia. However, the duration of gastrostomy is relatively longer than that of PEG, and general anesthesia may cause dangerous situations depending on the patient’s general condition. In addition, the cost and recovery time after the procedure also has disadvantages compared with those of PEG.13 As in this survey, the priority consideration for pediatric gastrostomy procedures in Korea is PEG, and SG is selected in situations where PEG is difficult. SG has also been developed in a way that compensates for its limitations. Recently, laparoscopy has been used instead of open surgery, which leaves a large scar. However, fundoplication or gastrostomy using laparoscopy cannot shorten the procedure time or eliminate the need for general anesthesia, although a recent study reported that laparoscopic PEG may be safer than conventional PEG in high-risk pediatric patients with severe thoracoabdominal deformity, previous abdominal surgery, ventriculoperitoneal shunt, and abdominal tumors.1415 In addition, the difference in the procedures time depends more on the ability of the operator than on the procedure method. In this study, among the cases in which gastrostomy was performed again, there was a case in which the stormy site was placed too close to the pylorus, causing ballooning to interfere with gastric emptying. This may occur more often when open surgery is performed for gastrostomy in young children. Laparoscopy is more advantageous for site selection than open surgery. However, this complication can never occur in PEGs that focus on the stomach. Although the laparoscopic method is a recent trend in surgical procedures, it has inevitable disadvantages in terms of fundoplication. Fundoplication is a procedure in which the lower esophagus is wound around the muscle and tightened by the operator. Depending on the proficiency of the operator, reflux may continue because of tightening too loosely, and there may be cases where the liquids cannot be swallowed because of the tightening was too hard. Therefore, a recent study published in Korea reported that concomitant EGD during fundoplication was better than fundoplication alone regarding optimal tightness inside the esophagus.16\nCompared to other countries, Korea has better access to pediatric endoscopy, and many pediatric endoscopy specialists are active in various parts of the country.17 According to the questionnaire data obtained in this study, it was observed that each center has implemented methods according to each hospital’s situation to reduce complications, procedure time, and costs. Thus, it was found that most centers did not have frequent complications and maintained the procedure method. Most of them recognized gastrostomy as an essential technique for children to improve the nutritional status of the patient, reduce the hospitalization rate, and impose great advantages on the clinical course.1819 Approximately 50% of the institutions used general anesthesia, which could be related to the sedation method of pediatric endoscopy. Most pediatric endoscopists are responsible for managing PEG and gastrostomy tubes. A 38.7% of the institutions did not consider fundoplication, and initially implemented PEG gastrostomy. For pediatric endoscopists, this survey showed that PEG is a safe procedure that should be considered as a first-line treatment. In addition, based on the results of this survey, a multicenter chart review of patients undergoing these procedures should be conducted." ]
[ "intro", "methods", null, null, "ethics-statement", "results", null, null, "discussion" ]
[ "Gastrostomy", "Pediatrics", "Endoscopic Surgical Procedure", "Buried Bumper" ]
Neutrophil-Lymphocyte Ratio and Monocyte-Lymphocyte Ratio According to the Radiologic Severity of
36254530
To date, no study has investigated whether the neutrophil-lymphocyte ratio (NLR) and monocyte-lymphocyte ratio (MLR) have a clinical value in Mycobacterium avium complex (MAC)-pulmonary disease (PD).
BACKGROUND
We aimed to assess whether the baseline NLR and MLR were different according to the severity of MAC-PD based on the radiologic classification by retrospectively analyzing 549 patients treated in a tertiary referral center in South Korea.
METHODS
Both NLR and MLR were significantly higher as 3.33 and 0.43 respectively in the fibrocavitary type, followed by 2.34 and 0.27 in the cavitary nodular bronchiectatic type and significantly lower as 1.88 and 0.23 in the non-cavitary nodular bronchiectatic type.
RESULTS
The baseline NLR and MLR showed a distinct difference in accordance with the radiologic severity of MAC-PD.
CONCLUSION
[ "Humans", "Lung Diseases", "Lymphocytes", "Monocytes", "Mycobacterium avium Complex", "Mycobacterium avium-intracellulare Infection", "Neutrophils", "Retrospective Studies" ]
9577355
Introduction
The neutrophil-lymphocyte ratio (NLR) and monocyte-lymphocyte ratio (MLR) are readily available laboratory markers calculated from a complete blood count (CBC). Previous studies have reported that both NLR and MLR were useful laboratory markers of the severity, treatment response, or recurrence of pulmonary or extrapulmonary tuberculosis.12345 Although the clinical characteristics of Mycobacterium avium complex (MAC)-pulmonary disease (PD) are similar to those of tuberculosis in many aspects, such as symptoms, radiologic findings, or treatment regimen,6 to date, no study has investigated the value of NLR and MLR in MAC-PD. Therefore, we aimed to assess this issue in patients with MAC-PD. Considering that, notably, NLR and MLR were associated with tuberculosis severity, we investigated whether the baseline NLR and MLR were different according to the severity of MAC-PD based on the radiologic classification.
Methods
[SUBTITLE] Study subjects [SUBSECTION] Patients were enrolled in the Asan Medical Center in Seoul, South Korea. From 2001 to 2019, 1,148 patients with MAC-PD who had initiated treatment with a macrolide-containing regimen were identified. We excluded those 1) who did not undergo CBC examination within 90 days prior to treatment initiation (n = 384), 2) whose computed tomography (CT) findings were categorized as unclassifiable type (n = 150), and 3) who had comorbidities that could affect the results of white blood cell differential count, including liver disease (n = 24), infectious disease such as pneumonia (n = 21), were receiving calcineurin inhibitor (n = 7) or anti-cancer chemotherapy (n = 6), and others (n = 7). The medical records of the remaining patients were retrospectively analyzed in November 2021. Patients were enrolled in the Asan Medical Center in Seoul, South Korea. From 2001 to 2019, 1,148 patients with MAC-PD who had initiated treatment with a macrolide-containing regimen were identified. We excluded those 1) who did not undergo CBC examination within 90 days prior to treatment initiation (n = 384), 2) whose computed tomography (CT) findings were categorized as unclassifiable type (n = 150), and 3) who had comorbidities that could affect the results of white blood cell differential count, including liver disease (n = 24), infectious disease such as pneumonia (n = 21), were receiving calcineurin inhibitor (n = 7) or anti-cancer chemotherapy (n = 6), and others (n = 7). The medical records of the remaining patients were retrospectively analyzed in November 2021. [SUBTITLE] Radiologic classification and baseline NLR and MLR [SUBSECTION] Radiologic findings on chest CT were classified into one of the three major types: fibrocavitary (FC), cavitary nodular bronchiectatic (C-NB), and non-cavitary nodular bronchiectatic (NC-NB) types.7 The baseline NLR and MLR were compared according to each radiologic type. NLR and MLR were calculated from peripheral CBC. An automatic blood count device was employed for the analysis of CBC. NLR and MLR were determined as follows: NLR = Absolute Neutrophil Count/Absolute Lymphocyte Count, MLR = Absolute Monocyte Count/Absolute Lymphocyte Count.4 Radiologic findings on chest CT were classified into one of the three major types: fibrocavitary (FC), cavitary nodular bronchiectatic (C-NB), and non-cavitary nodular bronchiectatic (NC-NB) types.7 The baseline NLR and MLR were compared according to each radiologic type. NLR and MLR were calculated from peripheral CBC. An automatic blood count device was employed for the analysis of CBC. NLR and MLR were determined as follows: NLR = Absolute Neutrophil Count/Absolute Lymphocyte Count, MLR = Absolute Monocyte Count/Absolute Lymphocyte Count.4 [SUBTITLE] Statistical analysis [SUBSECTION] All data are presented as means ± standard deviation or as medians (interquartile range [IQR]) for continuous variables and number (%) for categorical variables. The baseline NLR and MLR values according to radiologic classification were compared using the Kruskal-Wallis test. For statistical analysis, an IBM SPSS Statistics for Windows, version 26.0 (IBM Corp., Armonk, NY, USA) software was used. All data are presented as means ± standard deviation or as medians (interquartile range [IQR]) for continuous variables and number (%) for categorical variables. The baseline NLR and MLR values according to radiologic classification were compared using the Kruskal-Wallis test. For statistical analysis, an IBM SPSS Statistics for Windows, version 26.0 (IBM Corp., Armonk, NY, USA) software was used. [SUBTITLE] Ethics statement [SUBSECTION] The study protocol was approved by the Institutional Review Board (IRB) of the Asan Medical Center (IRB No. 2021-1210). The requirement for informed consent was waived by the board because of the retrospective nature of this study. The study protocol was approved by the Institutional Review Board (IRB) of the Asan Medical Center (IRB No. 2021-1210). The requirement for informed consent was waived by the board because of the retrospective nature of this study.
Results
[SUBTITLE] Study participants [SUBSECTION] The eligibility screening identified 549 patients with MAC-PD, including 95, 176, and 278 patients with FC, C-NB, and NC-NB types, respectively. Their median age was 64.0 (IQR, 56.0–72.0) years, and women were predominant (61.4%). The mean body mass index was 20.3 ± 2.9 kg/m2. Sputum acid-fast bacillus (AFB) smear positivity was noted in 49.0% of the patients. As shown in Table 1, a statistically significant difference was noted in terms of age, gender, body mass index, smoking history, etiologic organism, and sputum AFB smear positivity. Data are presented as mean ± standard deviation, median (interquartile range), or frequencies (%). FC = fibrocavitary, C-NB = cavitary nodular bronchiectatic, NC-NB = non-cavitary nodular bronchiectatic. aThe extent of the involved lobe was assessed according to how many of the six lung lobes were involved, considering the lingular segment to be a separate lobe. The eligibility screening identified 549 patients with MAC-PD, including 95, 176, and 278 patients with FC, C-NB, and NC-NB types, respectively. Their median age was 64.0 (IQR, 56.0–72.0) years, and women were predominant (61.4%). The mean body mass index was 20.3 ± 2.9 kg/m2. Sputum acid-fast bacillus (AFB) smear positivity was noted in 49.0% of the patients. As shown in Table 1, a statistically significant difference was noted in terms of age, gender, body mass index, smoking history, etiologic organism, and sputum AFB smear positivity. Data are presented as mean ± standard deviation, median (interquartile range), or frequencies (%). FC = fibrocavitary, C-NB = cavitary nodular bronchiectatic, NC-NB = non-cavitary nodular bronchiectatic. aThe extent of the involved lobe was assessed according to how many of the six lung lobes were involved, considering the lingular segment to be a separate lobe. [SUBTITLE] Baseline NLR and MLR according to radiologic classification [SUBSECTION] The baseline NLR of the 549 patients was 2.16 (IQR, 1.47–3.25). As shown in Fig. 1A, NLR was significantly higher in the FC type (3.33 [IQR, 2.49–4.50]), followed by that in the C-NB type (2.34 [IQR, 1.60–3.55]), and significantly lower in the NC-NB type (1.88 [IQR, 1.30–2.44]) (P < 0.001). In addition, the baseline MLR of the 549 patients was 0.27 (IQR, 0.20–0.39), and it was significantly higher in the FC type (0.43 [IQR, 0.30–0.57]), followed by that in the C-NB type (0.27 [IQR, 0.21–0.40]), and NC-NB type (0.23 [IQR, 0.18–0.31]) (P < 0.001), as shown in Fig. 1B. FC = fibrocavitary, C-NB = cavitary nodular bronchiectatic, NC-NB = non-cavitary nodular bronchiectatic. The baseline NLR of the 549 patients was 2.16 (IQR, 1.47–3.25). As shown in Fig. 1A, NLR was significantly higher in the FC type (3.33 [IQR, 2.49–4.50]), followed by that in the C-NB type (2.34 [IQR, 1.60–3.55]), and significantly lower in the NC-NB type (1.88 [IQR, 1.30–2.44]) (P < 0.001). In addition, the baseline MLR of the 549 patients was 0.27 (IQR, 0.20–0.39), and it was significantly higher in the FC type (0.43 [IQR, 0.30–0.57]), followed by that in the C-NB type (0.27 [IQR, 0.21–0.40]), and NC-NB type (0.23 [IQR, 0.18–0.31]) (P < 0.001), as shown in Fig. 1B. FC = fibrocavitary, C-NB = cavitary nodular bronchiectatic, NC-NB = non-cavitary nodular bronchiectatic.
null
null
[ "Study subjects", "Radiologic classification and baseline NLR and MLR", "Statistical analysis", "Study participants", "Baseline NLR and MLR according to radiologic classification" ]
[ "Patients were enrolled in the Asan Medical Center in Seoul, South Korea. From 2001 to 2019, 1,148 patients with MAC-PD who had initiated treatment with a macrolide-containing regimen were identified. We excluded those 1) who did not undergo CBC examination within 90 days prior to treatment initiation (n = 384), 2) whose computed tomography (CT) findings were categorized as unclassifiable type (n = 150), and 3) who had comorbidities that could affect the results of white blood cell differential count, including liver disease (n = 24), infectious disease such as pneumonia (n = 21), were receiving calcineurin inhibitor (n = 7) or anti-cancer chemotherapy (n = 6), and others (n = 7). The medical records of the remaining patients were retrospectively analyzed in November 2021.", "Radiologic findings on chest CT were classified into one of the three major types: fibrocavitary (FC), cavitary nodular bronchiectatic (C-NB), and non-cavitary nodular bronchiectatic (NC-NB) types.7 The baseline NLR and MLR were compared according to each radiologic type. NLR and MLR were calculated from peripheral CBC. An automatic blood count device was employed for the analysis of CBC. NLR and MLR were determined as follows: NLR = Absolute Neutrophil Count/Absolute Lymphocyte Count, MLR = Absolute Monocyte Count/Absolute Lymphocyte Count.4", "All data are presented as means ± standard deviation or as medians (interquartile range [IQR]) for continuous variables and number (%) for categorical variables. The baseline NLR and MLR values according to radiologic classification were compared using the Kruskal-Wallis test. For statistical analysis, an IBM SPSS Statistics for Windows, version 26.0 (IBM Corp., Armonk, NY, USA) software was used.", "The eligibility screening identified 549 patients with MAC-PD, including 95, 176, and 278 patients with FC, C-NB, and NC-NB types, respectively. Their median age was 64.0 (IQR, 56.0–72.0) years, and women were predominant (61.4%). The mean body mass index was 20.3 ± 2.9 kg/m2. Sputum acid-fast bacillus (AFB) smear positivity was noted in 49.0% of the patients. As shown in Table 1, a statistically significant difference was noted in terms of age, gender, body mass index, smoking history, etiologic organism, and sputum AFB smear positivity.\nData are presented as mean ± standard deviation, median (interquartile range), or frequencies (%).\nFC = fibrocavitary, C-NB = cavitary nodular bronchiectatic, NC-NB = non-cavitary nodular bronchiectatic.\naThe extent of the involved lobe was assessed according to how many of the six lung lobes were involved, considering the lingular segment to be a separate lobe.", "The baseline NLR of the 549 patients was 2.16 (IQR, 1.47–3.25). As shown in Fig. 1A, NLR was significantly higher in the FC type (3.33 [IQR, 2.49–4.50]), followed by that in the C-NB type (2.34 [IQR, 1.60–3.55]), and significantly lower in the NC-NB type (1.88 [IQR, 1.30–2.44]) (P < 0.001). In addition, the baseline MLR of the 549 patients was 0.27 (IQR, 0.20–0.39), and it was significantly higher in the FC type (0.43 [IQR, 0.30–0.57]), followed by that in the C-NB type (0.27 [IQR, 0.21–0.40]), and NC-NB type (0.23 [IQR, 0.18–0.31]) (P < 0.001), as shown in Fig. 1B.\nFC = fibrocavitary, C-NB = cavitary nodular bronchiectatic, NC-NB = non-cavitary nodular bronchiectatic." ]
[ null, null, null, null, null ]
[ "Introduction", "Methods", "Study subjects", "Radiologic classification and baseline NLR and MLR", "Statistical analysis", "Ethics statement", "Results", "Study participants", "Baseline NLR and MLR according to radiologic classification", "Discussion" ]
[ "The neutrophil-lymphocyte ratio (NLR) and monocyte-lymphocyte ratio (MLR) are readily available laboratory markers calculated from a complete blood count (CBC). Previous studies have reported that both NLR and MLR were useful laboratory markers of the severity, treatment response, or recurrence of pulmonary or extrapulmonary tuberculosis.12345 Although the clinical characteristics of Mycobacterium avium complex (MAC)-pulmonary disease (PD) are similar to those of tuberculosis in many aspects, such as symptoms, radiologic findings, or treatment regimen,6 to date, no study has investigated the value of NLR and MLR in MAC-PD. Therefore, we aimed to assess this issue in patients with MAC-PD. Considering that, notably, NLR and MLR were associated with tuberculosis severity, we investigated whether the baseline NLR and MLR were different according to the severity of MAC-PD based on the radiologic classification.", "[SUBTITLE] Study subjects [SUBSECTION] Patients were enrolled in the Asan Medical Center in Seoul, South Korea. From 2001 to 2019, 1,148 patients with MAC-PD who had initiated treatment with a macrolide-containing regimen were identified. We excluded those 1) who did not undergo CBC examination within 90 days prior to treatment initiation (n = 384), 2) whose computed tomography (CT) findings were categorized as unclassifiable type (n = 150), and 3) who had comorbidities that could affect the results of white blood cell differential count, including liver disease (n = 24), infectious disease such as pneumonia (n = 21), were receiving calcineurin inhibitor (n = 7) or anti-cancer chemotherapy (n = 6), and others (n = 7). The medical records of the remaining patients were retrospectively analyzed in November 2021.\nPatients were enrolled in the Asan Medical Center in Seoul, South Korea. From 2001 to 2019, 1,148 patients with MAC-PD who had initiated treatment with a macrolide-containing regimen were identified. We excluded those 1) who did not undergo CBC examination within 90 days prior to treatment initiation (n = 384), 2) whose computed tomography (CT) findings were categorized as unclassifiable type (n = 150), and 3) who had comorbidities that could affect the results of white blood cell differential count, including liver disease (n = 24), infectious disease such as pneumonia (n = 21), were receiving calcineurin inhibitor (n = 7) or anti-cancer chemotherapy (n = 6), and others (n = 7). The medical records of the remaining patients were retrospectively analyzed in November 2021.\n[SUBTITLE] Radiologic classification and baseline NLR and MLR [SUBSECTION] Radiologic findings on chest CT were classified into one of the three major types: fibrocavitary (FC), cavitary nodular bronchiectatic (C-NB), and non-cavitary nodular bronchiectatic (NC-NB) types.7 The baseline NLR and MLR were compared according to each radiologic type. NLR and MLR were calculated from peripheral CBC. An automatic blood count device was employed for the analysis of CBC. NLR and MLR were determined as follows: NLR = Absolute Neutrophil Count/Absolute Lymphocyte Count, MLR = Absolute Monocyte Count/Absolute Lymphocyte Count.4\nRadiologic findings on chest CT were classified into one of the three major types: fibrocavitary (FC), cavitary nodular bronchiectatic (C-NB), and non-cavitary nodular bronchiectatic (NC-NB) types.7 The baseline NLR and MLR were compared according to each radiologic type. NLR and MLR were calculated from peripheral CBC. An automatic blood count device was employed for the analysis of CBC. NLR and MLR were determined as follows: NLR = Absolute Neutrophil Count/Absolute Lymphocyte Count, MLR = Absolute Monocyte Count/Absolute Lymphocyte Count.4\n[SUBTITLE] Statistical analysis [SUBSECTION] All data are presented as means ± standard deviation or as medians (interquartile range [IQR]) for continuous variables and number (%) for categorical variables. The baseline NLR and MLR values according to radiologic classification were compared using the Kruskal-Wallis test. For statistical analysis, an IBM SPSS Statistics for Windows, version 26.0 (IBM Corp., Armonk, NY, USA) software was used.\nAll data are presented as means ± standard deviation or as medians (interquartile range [IQR]) for continuous variables and number (%) for categorical variables. The baseline NLR and MLR values according to radiologic classification were compared using the Kruskal-Wallis test. For statistical analysis, an IBM SPSS Statistics for Windows, version 26.0 (IBM Corp., Armonk, NY, USA) software was used.\n[SUBTITLE] Ethics statement [SUBSECTION] The study protocol was approved by the Institutional Review Board (IRB) of the Asan Medical Center (IRB No. 2021-1210). The requirement for informed consent was waived by the board because of the retrospective nature of this study.\nThe study protocol was approved by the Institutional Review Board (IRB) of the Asan Medical Center (IRB No. 2021-1210). The requirement for informed consent was waived by the board because of the retrospective nature of this study.", "Patients were enrolled in the Asan Medical Center in Seoul, South Korea. From 2001 to 2019, 1,148 patients with MAC-PD who had initiated treatment with a macrolide-containing regimen were identified. We excluded those 1) who did not undergo CBC examination within 90 days prior to treatment initiation (n = 384), 2) whose computed tomography (CT) findings were categorized as unclassifiable type (n = 150), and 3) who had comorbidities that could affect the results of white blood cell differential count, including liver disease (n = 24), infectious disease such as pneumonia (n = 21), were receiving calcineurin inhibitor (n = 7) or anti-cancer chemotherapy (n = 6), and others (n = 7). The medical records of the remaining patients were retrospectively analyzed in November 2021.", "Radiologic findings on chest CT were classified into one of the three major types: fibrocavitary (FC), cavitary nodular bronchiectatic (C-NB), and non-cavitary nodular bronchiectatic (NC-NB) types.7 The baseline NLR and MLR were compared according to each radiologic type. NLR and MLR were calculated from peripheral CBC. An automatic blood count device was employed for the analysis of CBC. NLR and MLR were determined as follows: NLR = Absolute Neutrophil Count/Absolute Lymphocyte Count, MLR = Absolute Monocyte Count/Absolute Lymphocyte Count.4", "All data are presented as means ± standard deviation or as medians (interquartile range [IQR]) for continuous variables and number (%) for categorical variables. The baseline NLR and MLR values according to radiologic classification were compared using the Kruskal-Wallis test. For statistical analysis, an IBM SPSS Statistics for Windows, version 26.0 (IBM Corp., Armonk, NY, USA) software was used.", "The study protocol was approved by the Institutional Review Board (IRB) of the Asan Medical Center (IRB No. 2021-1210). The requirement for informed consent was waived by the board because of the retrospective nature of this study.", "[SUBTITLE] Study participants [SUBSECTION] The eligibility screening identified 549 patients with MAC-PD, including 95, 176, and 278 patients with FC, C-NB, and NC-NB types, respectively. Their median age was 64.0 (IQR, 56.0–72.0) years, and women were predominant (61.4%). The mean body mass index was 20.3 ± 2.9 kg/m2. Sputum acid-fast bacillus (AFB) smear positivity was noted in 49.0% of the patients. As shown in Table 1, a statistically significant difference was noted in terms of age, gender, body mass index, smoking history, etiologic organism, and sputum AFB smear positivity.\nData are presented as mean ± standard deviation, median (interquartile range), or frequencies (%).\nFC = fibrocavitary, C-NB = cavitary nodular bronchiectatic, NC-NB = non-cavitary nodular bronchiectatic.\naThe extent of the involved lobe was assessed according to how many of the six lung lobes were involved, considering the lingular segment to be a separate lobe.\nThe eligibility screening identified 549 patients with MAC-PD, including 95, 176, and 278 patients with FC, C-NB, and NC-NB types, respectively. Their median age was 64.0 (IQR, 56.0–72.0) years, and women were predominant (61.4%). The mean body mass index was 20.3 ± 2.9 kg/m2. Sputum acid-fast bacillus (AFB) smear positivity was noted in 49.0% of the patients. As shown in Table 1, a statistically significant difference was noted in terms of age, gender, body mass index, smoking history, etiologic organism, and sputum AFB smear positivity.\nData are presented as mean ± standard deviation, median (interquartile range), or frequencies (%).\nFC = fibrocavitary, C-NB = cavitary nodular bronchiectatic, NC-NB = non-cavitary nodular bronchiectatic.\naThe extent of the involved lobe was assessed according to how many of the six lung lobes were involved, considering the lingular segment to be a separate lobe.\n[SUBTITLE] Baseline NLR and MLR according to radiologic classification [SUBSECTION] The baseline NLR of the 549 patients was 2.16 (IQR, 1.47–3.25). As shown in Fig. 1A, NLR was significantly higher in the FC type (3.33 [IQR, 2.49–4.50]), followed by that in the C-NB type (2.34 [IQR, 1.60–3.55]), and significantly lower in the NC-NB type (1.88 [IQR, 1.30–2.44]) (P < 0.001). In addition, the baseline MLR of the 549 patients was 0.27 (IQR, 0.20–0.39), and it was significantly higher in the FC type (0.43 [IQR, 0.30–0.57]), followed by that in the C-NB type (0.27 [IQR, 0.21–0.40]), and NC-NB type (0.23 [IQR, 0.18–0.31]) (P < 0.001), as shown in Fig. 1B.\nFC = fibrocavitary, C-NB = cavitary nodular bronchiectatic, NC-NB = non-cavitary nodular bronchiectatic.\nThe baseline NLR of the 549 patients was 2.16 (IQR, 1.47–3.25). As shown in Fig. 1A, NLR was significantly higher in the FC type (3.33 [IQR, 2.49–4.50]), followed by that in the C-NB type (2.34 [IQR, 1.60–3.55]), and significantly lower in the NC-NB type (1.88 [IQR, 1.30–2.44]) (P < 0.001). In addition, the baseline MLR of the 549 patients was 0.27 (IQR, 0.20–0.39), and it was significantly higher in the FC type (0.43 [IQR, 0.30–0.57]), followed by that in the C-NB type (0.27 [IQR, 0.21–0.40]), and NC-NB type (0.23 [IQR, 0.18–0.31]) (P < 0.001), as shown in Fig. 1B.\nFC = fibrocavitary, C-NB = cavitary nodular bronchiectatic, NC-NB = non-cavitary nodular bronchiectatic.", "The eligibility screening identified 549 patients with MAC-PD, including 95, 176, and 278 patients with FC, C-NB, and NC-NB types, respectively. Their median age was 64.0 (IQR, 56.0–72.0) years, and women were predominant (61.4%). The mean body mass index was 20.3 ± 2.9 kg/m2. Sputum acid-fast bacillus (AFB) smear positivity was noted in 49.0% of the patients. As shown in Table 1, a statistically significant difference was noted in terms of age, gender, body mass index, smoking history, etiologic organism, and sputum AFB smear positivity.\nData are presented as mean ± standard deviation, median (interquartile range), or frequencies (%).\nFC = fibrocavitary, C-NB = cavitary nodular bronchiectatic, NC-NB = non-cavitary nodular bronchiectatic.\naThe extent of the involved lobe was assessed according to how many of the six lung lobes were involved, considering the lingular segment to be a separate lobe.", "The baseline NLR of the 549 patients was 2.16 (IQR, 1.47–3.25). As shown in Fig. 1A, NLR was significantly higher in the FC type (3.33 [IQR, 2.49–4.50]), followed by that in the C-NB type (2.34 [IQR, 1.60–3.55]), and significantly lower in the NC-NB type (1.88 [IQR, 1.30–2.44]) (P < 0.001). In addition, the baseline MLR of the 549 patients was 0.27 (IQR, 0.20–0.39), and it was significantly higher in the FC type (0.43 [IQR, 0.30–0.57]), followed by that in the C-NB type (0.27 [IQR, 0.21–0.40]), and NC-NB type (0.23 [IQR, 0.18–0.31]) (P < 0.001), as shown in Fig. 1B.\nFC = fibrocavitary, C-NB = cavitary nodular bronchiectatic, NC-NB = non-cavitary nodular bronchiectatic.", "Although previous studies have shown that NLR and MLR are related to the treatment outcomes or severity of tuberculosis,1245 whether these two laboratory markers would also have a clinical value in MAC-PD has not been assessed, so far. This study investigated this aspect by retrospectively analyzing 549 patients with MAC-PD in a tertiary referral center in South Korea. We found a distinct difference in the baseline NLR and MLR according to the radiologic severity of MAC-PD; both NLR and MLR showed higher values when the radiologic findings of MAC-PD were more severe.\nRadiologic classification has vital importance in MAC-PD regarding various aspects including the treatment regimen, outcome, and prognosis. In terms of drug regimen composition, an intermittent oral-drug therapy is recommended for patients with NC-NB type, whereas an aminoglycoside-containing daily regimen is recommended for those with cavitary type (i.e., FC and C-NB types).6 Notably, the treatment success rate for cavitary MAC-PD was reported to be significantly lower than that for NC-NB type despite receiving standard treatment.7 In addition, significant difference in the mortality rate was noted according to the radiologic type of MAC-PD. A recent study reported that the overall survival rates were higher in patients with NC-NB type than in those with a cavitary disease, and those patients with C-NB type showed mortality rates intermediate between those of NC-NB and FC forms.8\nThese clinical differences according to radiologic classification could be explained by cavitation, which indicates a higher mycobacterial burden in MAC-PD.9 Therefore, it can be predicted that the presence of cavity is accompanied by a severe overall inflammatory response. In general, the immune responses of circulating leukocytes to various inflammatory events are characterized by an increased number of neutrophils in the peripheral blood.10 In the case of tuberculosis, extensive neutrophilic response is a sign of severity and has been particularly related to tissue destruction.11 Neutrophilia is independently associated with a higher risk of cavity formation.12 Additionally, an increase in matrix metalloproteinase-8-expressing neutrophil was related to cavitation in tuberculosis.9 Moreover, monocytes are one of the essential components of the innate immune responses and act as a link to the adaptive immune system.4 Therefore, as the disease progresses, the number of monocytes derived from the hematopoietic stem cells could increase to phagocytize and restrict mycobacterium.13 An ongoing immune response against the cavitary lesion could result in further increase in the percentage of blood monocytes.141516 Moreover, lymphopenia could result from the accumulation of lymphocytes at the infection site, leading to their decreased number in the peripheral blood.17 These theoretical changes in neutrophils, lymphocytes, and monocytes can reasonably explain why NLR and MLR were increased in accordance with the radiologic severity in MAC-PD.\nOnly those patients who received treatment were included in this study. This is because patients with cavitary form MAC-PD without treatment are rarely encountered, as immediate treatment initiation is recommended for this type.18 When we performed the same analysis in patients with MAC-PD who did not receive treatment using the data of the patients enrolled in our previous study,19 we found a statistical difference in NLR and MLR between the cavitary vs. non-cavitary type (Supplementary Fig. 1). However, the number of patients with cavitary type was too small to analyze.\nThere may be other laboratory test results that can be related to the radiologic severity of MAC-PD including erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), or platelet count considering that previous studies have shown these inflammatory markers were associated with severity or the immune response of mycobacterial disease.202122 We compared the ESR, CRP, or platelet count according to the radiologic classification among a portion of patients in this study for whom these inflammatory markers were measured, the results of which are shown in Supplementary Fig. 2. As the Figure denotes, statistically significant differences in CRP and platelet count were noted among the three groups. Besides laboratory markers, the extent of radiologic involvement could also reflect the severity of MAC-PD. Additional analysis and results regarding whether the baseline NLR and MLR differ according to the number of involved lobes in each radiologic type, are summarized in Supplementary Fig. 3.\nThis study had some limitations; the most significant limitation being that it was conducted at a single referral center, and it had a retrospective design. Second, although NLR and MLR were measured in all the enrolled patients within 90 days of treatment initiation, the measured time points were slightly different for each patient. It was unclear whether the slightly different measurement time points for each patient affected the overall results of NLR and MLR. Finally, whether NLR and MLR could be used as a marker to predict treatment outcomes was not assessed. Further studies are needed to determine whether these values at baseline or during treatment are related to treatment outcomes in terms of treatment success or mortality.\nIn conclusion, this study showed that the baseline NLR and MLR had a distinct difference in accordance with the radiologic severity of MAC-PD." ]
[ "intro", "methods", null, null, null, "ethics-statement", "results", null, null, "discussion" ]
[ "Mycobacterium avium Complex", "Complete Blood Count", "Neutrophil", "Lymphocyte", "Monocyte" ]
The First Systematic Gastroscopy Training Program for Surgeons in Korea.
36254531
Endoscopic evaluation of the stomach is essential for preoperative planning and post-surgical surveillance for various diseases of the stomach, including malignancy. The gastroscopy education program for surgeons is currently in its infancy and is not systematically organized in Korea. This study aimed to introduce the first systematic gastroscopy education program for surgeons in Korea.
BACKGROUND
The gastroscopy education program entitled "Gastroscopy School for Surgeons (GSS)" comprised of theoretical education, dry lab hands-on training, and clinical practice. All participants were beginners without any gastroscopy experience. Clinical practice started after the completion of the theoretical and dry lab training. The gastroscopy practices utilized simple luminal observation, biopsy, localization using clips or dye injection, and limited therapeutic gastroscopy. The educational performances and surveys from 33 participants were analyzed.
METHODS
The participants consisted of surgical residents, general surgeons, gastrointestinal-specialized surgeons, and physicians. Participants performed a total of 2,272 gastroscopies, 2,008 of which were post-gastrectomy cases. Currently, of the 33 participants, 7 (21.2%) of the participants performed gastroscopy regularly, and 7 (21.2%) occasionally. According to the self-reported survey, one participant assessed their current gastroscopic technique to be at the expert level, and 25 (75.8%) at a proficient level. All participants considered gastroscopy education for surgeons to be necessary, and 28 (84.8%) stated that systematic education is not currently provided in Korea.
RESULTS
We introduced the first systematic gastroscopy education program for surgeons in Korea, namely the GSS, which is practical and meets clinical needs. More training centers are needed to expand gastroscopy training among Korean surgeons.
CONCLUSION
[ "Gastrectomy", "Gastroscopy", "Humans", "Republic of Korea", "Surgeons", "Surgical Instruments" ]
9577353
INTRODUCTION
Evaluation of the stomach before and after surgery using gastroscopy is essential for treating various diseases of the stomach, including malignancy.12 In particular, gastroscopy can be used for clinical staging, tumor localization, treatment of early gastric cancer, complication management, and post-gastrectomy surveillance.3456 In Korea, most gastroscopies are performed by a gastroenterologist, and they are involved in all aspects of a broad endoscopic area.7 Gastroenterologists usually focus on diagnosing and treating a wide range of diseases using endoscopy, not only malignancy but also benign diseases such as motility disorders or functional diseases. Therefore, the endoscopy education system for gastroenterologists is more systematic, detailed, and strict to produce endoscopy experts.89 In contrast, gastrointestinal (GI) surgeons are more interested in the narrow area of endoscopy, such as localization for gastrectomy, intraoperative gastroscopy, and post-gastrectomy surveillance.1011 In addition, gastroscopy education for GI surgeons was not much developed due to surgeons’ low interest. Currently, the necessity of surgeons’ endoscopy is being raised because minimal invasive approach is adopted and performed actively in every field of surgery. In particular, intraoperative endoscopy has an important role as an assistant of minimal invasive surgery. Several studies reported the advantages of intraoperative gastroscopy in terms of precise localization of the tumor during laparoscopic surgery, and image-guided surgery using indocyanine green (ICG).2121314 In such situation, education system of endoscopy for surgeon is needed to increase the surgeon’s understanding of gastroscopy, meet the increasing demands of intraoperative gastroscopy, and to make a smooth communication between gastroenterologists and GI surgeons.1516 However, there is a lack of a well-organized gastroscopy education program for surgeons in Korea. This study aimed to describe the first systematic gastroscopy education program for surgeons in Korea.
METHODS
[SUBTITLE] Education program [SUBSECTION] The surgeon’s gastroscopy education program at Seoul St. Mary’s Hospital started in 2010. Initially, it was conducted for senior surgical residents and clinical fellows who trained at the Division of Gastrointestinal Surgery, Department of Surgery of Seoul St. Mary’s Hospital. In August 2017, a systematic gastroscopy education program, entitled the Gastroscopy School for Surgeons (GSS), was launched. The GSS is divided into 12 and 20 weeks, depending on the breadth and depth of training. The 12-week course is aimed at senior surgical residents and general surgeons (including colorectal, hepatobiliopancreatic, breast, general surgeons, but excluding GI surgeons). Training before clinical practice consists of an introductory lecture, dry lab hands-on training, and simulations. The introductory lecture includes 1) sedation and patient monitoring; 2) indications, limitations, and contraindications of gastroscopic procedure; 3) complications and management; 4) preparation and informed consent with the ethical issue; 5) various gastroscopic findings; 6) writing reports of gastroscopic findings.8 During the clinical practice component, gastroscope insertion and luminal observation, post-gastrectomy surveillance, biopsy, clipping, etc., are taught. Approximately 30 to 100 cases of gastroscopy on actual patients are performed. The 20-week course is specifically designed for the clinical fellows of GI surgery and GI surgeons. In addition to content from the 12-week course, advanced techniques such as intraoperative gastroscopy, tumor localization with dye injection, polypectomy, and balloon dilatation are included, and approximately 100 to 180 cases are performed. All trainers are GI surgeons, with at least several years of experience in gastroscopy. More than 1,000 cases are performed by each trainer per year. Trainer skills were standardized through meetings and consensus before teaching in the GSS. The surgeon’s gastroscopy education program at Seoul St. Mary’s Hospital started in 2010. Initially, it was conducted for senior surgical residents and clinical fellows who trained at the Division of Gastrointestinal Surgery, Department of Surgery of Seoul St. Mary’s Hospital. In August 2017, a systematic gastroscopy education program, entitled the Gastroscopy School for Surgeons (GSS), was launched. The GSS is divided into 12 and 20 weeks, depending on the breadth and depth of training. The 12-week course is aimed at senior surgical residents and general surgeons (including colorectal, hepatobiliopancreatic, breast, general surgeons, but excluding GI surgeons). Training before clinical practice consists of an introductory lecture, dry lab hands-on training, and simulations. The introductory lecture includes 1) sedation and patient monitoring; 2) indications, limitations, and contraindications of gastroscopic procedure; 3) complications and management; 4) preparation and informed consent with the ethical issue; 5) various gastroscopic findings; 6) writing reports of gastroscopic findings.8 During the clinical practice component, gastroscope insertion and luminal observation, post-gastrectomy surveillance, biopsy, clipping, etc., are taught. Approximately 30 to 100 cases of gastroscopy on actual patients are performed. The 20-week course is specifically designed for the clinical fellows of GI surgery and GI surgeons. In addition to content from the 12-week course, advanced techniques such as intraoperative gastroscopy, tumor localization with dye injection, polypectomy, and balloon dilatation are included, and approximately 100 to 180 cases are performed. All trainers are GI surgeons, with at least several years of experience in gastroscopy. More than 1,000 cases are performed by each trainer per year. Trainer skills were standardized through meetings and consensus before teaching in the GSS. [SUBTITLE] Assessment [SUBSECTION] The actual clinical practice included scope handling, luminal observation, scope insertion, mouth to mouth, and additional procedures. During the training, the trainer evaluates the trainee’s skill using Miller’s Prism, and each step must be performed excellently before progressing to the next step.17 Self-assessment of the learner’s skill level before and one year after training was recorded using the Kirkpatrick-Phillips model.18 The current status of the surgeon’s gastroscopy experience in each participant’s institution and their perception of gastroscopy education in Korea were recorded through a post-training survey.18 The survey form is shown in Supplementary Data 1. The actual clinical practice included scope handling, luminal observation, scope insertion, mouth to mouth, and additional procedures. During the training, the trainer evaluates the trainee’s skill using Miller’s Prism, and each step must be performed excellently before progressing to the next step.17 Self-assessment of the learner’s skill level before and one year after training was recorded using the Kirkpatrick-Phillips model.18 The current status of the surgeon’s gastroscopy experience in each participant’s institution and their perception of gastroscopy education in Korea were recorded through a post-training survey.18 The survey form is shown in Supplementary Data 1. [SUBTITLE] Participants and data collection [SUBSECTION] Participants who registered for the GSS and trained between August 2017 and November 2019 were enrolled in this study. The participants were recruited through a brochure and promotion through the Korean Surgical Society, the Korean Gastric Cancer Association, and the Korean Association of Surgeons. To ensure high-quality education, the trainer and trainee were matched 1:1, and the trainee’s performance was evaluated by the trainer during the training. Gastroscopy was performed on patients who underwent outpatient gastroscopy at the Division of Gastrointestinal Surgery of Seoul St. Mary’s Hospital. Prospectively collected data on the condition of the stomach, reconstruction method, and procedures during gastroscopy were analyzed. Participants who registered for the GSS and trained between August 2017 and November 2019 were enrolled in this study. The participants were recruited through a brochure and promotion through the Korean Surgical Society, the Korean Gastric Cancer Association, and the Korean Association of Surgeons. To ensure high-quality education, the trainer and trainee were matched 1:1, and the trainee’s performance was evaluated by the trainer during the training. Gastroscopy was performed on patients who underwent outpatient gastroscopy at the Division of Gastrointestinal Surgery of Seoul St. Mary’s Hospital. Prospectively collected data on the condition of the stomach, reconstruction method, and procedures during gastroscopy were analyzed. [SUBTITLE] Statistical analysis [SUBSECTION] Descriptive statistics were reported as numbers (percentages) and medians (interquartile ranges). Categorical variables were analyzed using the χ2 test and Fisher’s exact test to compare the groups. Statistical significance was set at P < 0.05. All statistical analyses were performed using SPSS for Windows (ver. 21.0; SPSS, Inc., Chicago, IL, USA). Descriptive statistics were reported as numbers (percentages) and medians (interquartile ranges). Categorical variables were analyzed using the χ2 test and Fisher’s exact test to compare the groups. Statistical significance was set at P < 0.05. All statistical analyses were performed using SPSS for Windows (ver. 21.0; SPSS, Inc., Chicago, IL, USA). [SUBTITLE] Ethics statement [SUBSECTION] The present study was approved by the Institutional Review Board of Seoul St. Mary’s Hospital (KC20QISI0077). Written informed consent was obtained from all the patients and participants. The present study was approved by the Institutional Review Board of Seoul St. Mary’s Hospital (KC20QISI0077). Written informed consent was obtained from all the patients and participants.
RESULTS
[SUBTITLE] Characteristics of the participants [SUBSECTION] In total, 33 participants were included in this study. The mean age was 35 years old. Twenty-five participants completed the 12-week course, and eight the 20-week course. The median number of gastroscopy cases was 62. The trainees consisted of three GI surgeons, nine general surgeons, 20 surgical residents, and one physician (Table 1). Values are presented as number (%) or number (interquartile range). GI= gastrointestinal. In total, 33 participants were included in this study. The mean age was 35 years old. Twenty-five participants completed the 12-week course, and eight the 20-week course. The median number of gastroscopy cases was 62. The trainees consisted of three GI surgeons, nine general surgeons, 20 surgical residents, and one physician (Table 1). Values are presented as number (%) or number (interquartile range). GI= gastrointestinal. [SUBTITLE] Details of the gastroscopy cases [SUBSECTION] Of the 2,272 cases of gastroscopy, 264 (11.6%) cases had no history of gastrectomy, and 2,008 (88.4%) cases had a history of any type of gastrectomy. Among the gastrectomy cases, the most common history of gastrectomy was distal gastrectomy (60.7%), followed by total gastrectomy (23.1%). Proximal gastrectomy, gastric wedge resection, and other types of gastrectomy were reported at 0.4%, 4.1%, and 0.1%, respectively. The reconstruction method after 1,378 distal gastrectomies consisted of Billroth I gastroduodenostomy (12.6%), Billroth II gastrojejunostomy (78.4%), and Roux-en-Y gastrojejunostomy (9.1%). Roux-en-Y esophagojejunostomy was performed for all gastrectomy cases. After proximal gastrectomy, double-tract reconstruction was performed in all patients (Fig. 1). Among the 2,272 cases of gastroscopy, additional procedures (other than luminal observation) performed were biopsy (18.0%), clipping (0.6%), injection (0.2%), dye spreading (0.4%), and balloon dilatation (0.6%). Severe gag reflex was the most common adverse event during a gastroscopy (29 cases). Mucosal injury, minor bleeding, and incomplete luminal observation were observed in one, nine, and four cases, respectively. All adverse events were resolved without special management (Supplementary Table 1). TG = total gastrectomy, DG = distal gastrectomy, PG = proximal gastrectomy, WR = wedge resection, R-Y = Roux-en-Y. Of the 2,272 cases of gastroscopy, 264 (11.6%) cases had no history of gastrectomy, and 2,008 (88.4%) cases had a history of any type of gastrectomy. Among the gastrectomy cases, the most common history of gastrectomy was distal gastrectomy (60.7%), followed by total gastrectomy (23.1%). Proximal gastrectomy, gastric wedge resection, and other types of gastrectomy were reported at 0.4%, 4.1%, and 0.1%, respectively. The reconstruction method after 1,378 distal gastrectomies consisted of Billroth I gastroduodenostomy (12.6%), Billroth II gastrojejunostomy (78.4%), and Roux-en-Y gastrojejunostomy (9.1%). Roux-en-Y esophagojejunostomy was performed for all gastrectomy cases. After proximal gastrectomy, double-tract reconstruction was performed in all patients (Fig. 1). Among the 2,272 cases of gastroscopy, additional procedures (other than luminal observation) performed were biopsy (18.0%), clipping (0.6%), injection (0.2%), dye spreading (0.4%), and balloon dilatation (0.6%). Severe gag reflex was the most common adverse event during a gastroscopy (29 cases). Mucosal injury, minor bleeding, and incomplete luminal observation were observed in one, nine, and four cases, respectively. All adverse events were resolved without special management (Supplementary Table 1). TG = total gastrectomy, DG = distal gastrectomy, PG = proximal gastrectomy, WR = wedge resection, R-Y = Roux-en-Y. [SUBTITLE] Survey’s results I: participant’s gastroscopy experience after education [SUBSECTION] After more than one year of training, a self-assessment of gastroscopy performance survey was completed by all 33 participants. One year after the training, the participants’ positions were seven GI surgeons, twenty-three general surgeons, two surgical residents, and one physician (Table 1). Currently, three participants performed gastroscopy as their main work (3 or more times/week), and seven performed gastroscopies weekly or more. In contrast, 16 participants have not performed gastroscopy in their work (Fig. 2A). One participant self-assessed their gastroscopy skills to be at the expert level (able to train others), and 25 evaluated themselves as good performers (without the need for supervision) or better. Notably, not a single participant evaluated themselves as incompetent in gastroscopy (Fig. 2B). All participants were able to perform basic procedures, such as luminal observation and biopsy. A total of 19 patients underwent minor procedures, such as clipping, injection, dye spreading, or balloon dilatation, and four underwent major procedures, such as stent insertion, polypectomy, or endoscopic submucosal dissection. Moreover, ten participants were able to perform intraoperative gastroscopy, and one was able to perform advanced procedures, such as endoscopic retrograde cholangiopancreatography (ERCP) or enteroscopy (Fig. 2). There was no significant difference in the skills and confidence level of the participants regardless of the training course (12-week vs. 20-week) completed. However, the skills level was relatively lower among participants who were trained in less than 60 cases (Supplementary Table 2). After more than one year of training, a self-assessment of gastroscopy performance survey was completed by all 33 participants. One year after the training, the participants’ positions were seven GI surgeons, twenty-three general surgeons, two surgical residents, and one physician (Table 1). Currently, three participants performed gastroscopy as their main work (3 or more times/week), and seven performed gastroscopies weekly or more. In contrast, 16 participants have not performed gastroscopy in their work (Fig. 2A). One participant self-assessed their gastroscopy skills to be at the expert level (able to train others), and 25 evaluated themselves as good performers (without the need for supervision) or better. Notably, not a single participant evaluated themselves as incompetent in gastroscopy (Fig. 2B). All participants were able to perform basic procedures, such as luminal observation and biopsy. A total of 19 patients underwent minor procedures, such as clipping, injection, dye spreading, or balloon dilatation, and four underwent major procedures, such as stent insertion, polypectomy, or endoscopic submucosal dissection. Moreover, ten participants were able to perform intraoperative gastroscopy, and one was able to perform advanced procedures, such as endoscopic retrograde cholangiopancreatography (ERCP) or enteroscopy (Fig. 2). There was no significant difference in the skills and confidence level of the participants regardless of the training course (12-week vs. 20-week) completed. However, the skills level was relatively lower among participants who were trained in less than 60 cases (Supplementary Table 2). [SUBTITLE] Survey’s results II: current status of surgeons’ gastroscopy [SUBSECTION] The survey also collected information on the gastroscopies performed by surgeons at the participant’s institution. At the time of the survey, 20 participants worked at seven tertiary university hospitals, seven were in private clinics, and six were in military service for their duty. The role of the surgeon in performing gastroscopy was diversely distributed by institutions, from all procedures were performed for all the cases to impossible to perform any procedures at all (Fig. 3A). There was no systematic gastroscopy education program for surgeons in any of the institutions except Seoul St. Mary’s Hospital (our institution). There were a few smaller training sessions conducted by surgeons or gastroenterologists (Fig. 3B). Despite the lack of formal gastroscopy training, surgeons reported that more than half of intraoperative gastroscopy procedures were performed by surgeons (Fig. 3C). The survey also collected information on the gastroscopies performed by surgeons at the participant’s institution. At the time of the survey, 20 participants worked at seven tertiary university hospitals, seven were in private clinics, and six were in military service for their duty. The role of the surgeon in performing gastroscopy was diversely distributed by institutions, from all procedures were performed for all the cases to impossible to perform any procedures at all (Fig. 3A). There was no systematic gastroscopy education program for surgeons in any of the institutions except Seoul St. Mary’s Hospital (our institution). There were a few smaller training sessions conducted by surgeons or gastroenterologists (Fig. 3B). Despite the lack of formal gastroscopy training, surgeons reported that more than half of intraoperative gastroscopy procedures were performed by surgeons (Fig. 3C). [SUBTITLE] Survey’s results III: subjective opinions of the participants [SUBSECTION] All participants recognized that gastroscopy was important to surgeons (Fig. 4A). The lack of a systemic education system was considered the primary barrier to surgeons performing gastroscopy. Additionally, conflict with the other departments was also recognized as a major hurdle. Furthermore, there was a perceived lack of interest among surgeons and the excessive workload was also problematic (Fig. 4B). Most participants stated that the gastroscopy education system for surgeons in Korea was insufficient (Fig. 4C). The participants provided their subjective opinions on the surgeon’s gastroscopy in the survey. A total of 87.9% identified the current problems associated with gastroscopy and highlighted the limitations of preoperative information and postoperative complication management when gastroscopy is performed by a non-surgeon, such as a gastroenterologist. The lack of a gastroscopy training system was also articulated. Second, 90.9% responded to the question regarding the merits of surgeons performing gastroscopies. The participants said that surgeons’ gastroscopy is useful in determining the detailed surgical plan preoperatively, and various reconstruction methods can be easily determined during postoperative surveillance. Finally, 54.5% provided suggestions for the development of the surgeon’s gastroscopy. Participants confirmed that it is necessary to increase the quality and quantity of gastroscopy education, with more training centers, and official support from professional associations and societies (Supplementary Table 3). All participants recognized that gastroscopy was important to surgeons (Fig. 4A). The lack of a systemic education system was considered the primary barrier to surgeons performing gastroscopy. Additionally, conflict with the other departments was also recognized as a major hurdle. Furthermore, there was a perceived lack of interest among surgeons and the excessive workload was also problematic (Fig. 4B). Most participants stated that the gastroscopy education system for surgeons in Korea was insufficient (Fig. 4C). The participants provided their subjective opinions on the surgeon’s gastroscopy in the survey. A total of 87.9% identified the current problems associated with gastroscopy and highlighted the limitations of preoperative information and postoperative complication management when gastroscopy is performed by a non-surgeon, such as a gastroenterologist. The lack of a gastroscopy training system was also articulated. Second, 90.9% responded to the question regarding the merits of surgeons performing gastroscopies. The participants said that surgeons’ gastroscopy is useful in determining the detailed surgical plan preoperatively, and various reconstruction methods can be easily determined during postoperative surveillance. Finally, 54.5% provided suggestions for the development of the surgeon’s gastroscopy. Participants confirmed that it is necessary to increase the quality and quantity of gastroscopy education, with more training centers, and official support from professional associations and societies (Supplementary Table 3).
null
null
[ "Education program", "Assessment", "Participants and data collection", "Statistical analysis", "Characteristics of the participants", "Details of the gastroscopy cases", "Survey’s results I: participant’s gastroscopy experience after education", "Survey’s results II: current status of surgeons’ gastroscopy", "Survey’s results III: subjective opinions of the participants" ]
[ "The surgeon’s gastroscopy education program at Seoul St. Mary’s Hospital started in 2010. Initially, it was conducted for senior surgical residents and clinical fellows who trained at the Division of Gastrointestinal Surgery, Department of Surgery of Seoul St. Mary’s Hospital. In August 2017, a systematic gastroscopy education program, entitled the Gastroscopy School for Surgeons (GSS), was launched.\nThe GSS is divided into 12 and 20 weeks, depending on the breadth and depth of training. The 12-week course is aimed at senior surgical residents and general surgeons (including colorectal, hepatobiliopancreatic, breast, general surgeons, but excluding GI surgeons). Training before clinical practice consists of an introductory lecture, dry lab hands-on training, and simulations. The introductory lecture includes 1) sedation and patient monitoring; 2) indications, limitations, and contraindications of gastroscopic procedure; 3) complications and management; 4) preparation and informed consent with the ethical issue; 5) various gastroscopic findings; 6) writing reports of gastroscopic findings.8 During the clinical practice component, gastroscope insertion and luminal observation, post-gastrectomy surveillance, biopsy, clipping, etc., are taught. Approximately 30 to 100 cases of gastroscopy on actual patients are performed. The 20-week course is specifically designed for the clinical fellows of GI surgery and GI surgeons. In addition to content from the 12-week course, advanced techniques such as intraoperative gastroscopy, tumor localization with dye injection, polypectomy, and balloon dilatation are included, and approximately 100 to 180 cases are performed.\nAll trainers are GI surgeons, with at least several years of experience in gastroscopy. More than 1,000 cases are performed by each trainer per year. Trainer skills were standardized through meetings and consensus before teaching in the GSS.", "The actual clinical practice included scope handling, luminal observation, scope insertion, mouth to mouth, and additional procedures. During the training, the trainer evaluates the trainee’s skill using Miller’s Prism, and each step must be performed excellently before progressing to the next step.17 Self-assessment of the learner’s skill level before and one year after training was recorded using the Kirkpatrick-Phillips model.18 The current status of the surgeon’s gastroscopy experience in each participant’s institution and their perception of gastroscopy education in Korea were recorded through a post-training survey.18 The survey form is shown in Supplementary Data 1.", "Participants who registered for the GSS and trained between August 2017 and November 2019 were enrolled in this study. The participants were recruited through a brochure and promotion through the Korean Surgical Society, the Korean Gastric Cancer Association, and the Korean Association of Surgeons. To ensure high-quality education, the trainer and trainee were matched 1:1, and the trainee’s performance was evaluated by the trainer during the training.\nGastroscopy was performed on patients who underwent outpatient gastroscopy at the Division of Gastrointestinal Surgery of Seoul St. Mary’s Hospital. Prospectively collected data on the condition of the stomach, reconstruction method, and procedures during gastroscopy were analyzed.", "Descriptive statistics were reported as numbers (percentages) and medians (interquartile ranges). Categorical variables were analyzed using the χ2 test and Fisher’s exact test to compare the groups. Statistical significance was set at P < 0.05. All statistical analyses were performed using SPSS for Windows (ver. 21.0; SPSS, Inc., Chicago, IL, USA).", "In total, 33 participants were included in this study. The mean age was 35 years old. Twenty-five participants completed the 12-week course, and eight the 20-week course. The median number of gastroscopy cases was 62. The trainees consisted of three GI surgeons, nine general surgeons, 20 surgical residents, and one physician (Table 1).\nValues are presented as number (%) or number (interquartile range).\nGI= gastrointestinal.", "Of the 2,272 cases of gastroscopy, 264 (11.6%) cases had no history of gastrectomy, and 2,008 (88.4%) cases had a history of any type of gastrectomy. Among the gastrectomy cases, the most common history of gastrectomy was distal gastrectomy (60.7%), followed by total gastrectomy (23.1%). Proximal gastrectomy, gastric wedge resection, and other types of gastrectomy were reported at 0.4%, 4.1%, and 0.1%, respectively. The reconstruction method after 1,378 distal gastrectomies consisted of Billroth I gastroduodenostomy (12.6%), Billroth II gastrojejunostomy (78.4%), and Roux-en-Y gastrojejunostomy (9.1%). Roux-en-Y esophagojejunostomy was performed for all gastrectomy cases. After proximal gastrectomy, double-tract reconstruction was performed in all patients (Fig. 1). Among the 2,272 cases of gastroscopy, additional procedures (other than luminal observation) performed were biopsy (18.0%), clipping (0.6%), injection (0.2%), dye spreading (0.4%), and balloon dilatation (0.6%). Severe gag reflex was the most common adverse event during a gastroscopy (29 cases). Mucosal injury, minor bleeding, and incomplete luminal observation were observed in one, nine, and four cases, respectively. All adverse events were resolved without special management (Supplementary Table 1).\nTG = total gastrectomy, DG = distal gastrectomy, PG = proximal gastrectomy, WR = wedge resection, R-Y = Roux-en-Y.", "After more than one year of training, a self-assessment of gastroscopy performance survey was completed by all 33 participants. One year after the training, the participants’ positions were seven GI surgeons, twenty-three general surgeons, two surgical residents, and one physician (Table 1). Currently, three participants performed gastroscopy as their main work (3 or more times/week), and seven performed gastroscopies weekly or more. In contrast, 16 participants have not performed gastroscopy in their work (Fig. 2A). One participant self-assessed their gastroscopy skills to be at the expert level (able to train others), and 25 evaluated themselves as good performers (without the need for supervision) or better. Notably, not a single participant evaluated themselves as incompetent in gastroscopy (Fig. 2B). All participants were able to perform basic procedures, such as luminal observation and biopsy. A total of 19 patients underwent minor procedures, such as clipping, injection, dye spreading, or balloon dilatation, and four underwent major procedures, such as stent insertion, polypectomy, or endoscopic submucosal dissection. Moreover, ten participants were able to perform intraoperative gastroscopy, and one was able to perform advanced procedures, such as endoscopic retrograde cholangiopancreatography (ERCP) or enteroscopy (Fig. 2). There was no significant difference in the skills and confidence level of the participants regardless of the training course (12-week vs. 20-week) completed. However, the skills level was relatively lower among participants who were trained in less than 60 cases (Supplementary Table 2).", "The survey also collected information on the gastroscopies performed by surgeons at the participant’s institution. At the time of the survey, 20 participants worked at seven tertiary university hospitals, seven were in private clinics, and six were in military service for their duty. The role of the surgeon in performing gastroscopy was diversely distributed by institutions, from all procedures were performed for all the cases to impossible to perform any procedures at all (Fig. 3A). There was no systematic gastroscopy education program for surgeons in any of the institutions except Seoul St. Mary’s Hospital (our institution). There were a few smaller training sessions conducted by surgeons or gastroenterologists (Fig. 3B). Despite the lack of formal gastroscopy training, surgeons reported that more than half of intraoperative gastroscopy procedures were performed by surgeons (Fig. 3C).", "All participants recognized that gastroscopy was important to surgeons (Fig. 4A). The lack of a systemic education system was considered the primary barrier to surgeons performing gastroscopy. Additionally, conflict with the other departments was also recognized as a major hurdle. Furthermore, there was a perceived lack of interest among surgeons and the excessive workload was also problematic (Fig. 4B). Most participants stated that the gastroscopy education system for surgeons in Korea was insufficient (Fig. 4C).\nThe participants provided their subjective opinions on the surgeon’s gastroscopy in the survey. A total of 87.9% identified the current problems associated with gastroscopy and highlighted the limitations of preoperative information and postoperative complication management when gastroscopy is performed by a non-surgeon, such as a gastroenterologist. The lack of a gastroscopy training system was also articulated. Second, 90.9% responded to the question regarding the merits of surgeons performing gastroscopies. The participants said that surgeons’ gastroscopy is useful in determining the detailed surgical plan preoperatively, and various reconstruction methods can be easily determined during postoperative surveillance. Finally, 54.5% provided suggestions for the development of the surgeon’s gastroscopy. Participants confirmed that it is necessary to increase the quality and quantity of gastroscopy education, with more training centers, and official support from professional associations and societies (Supplementary Table 3)." ]
[ null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "METHODS", "Education program", "Assessment", "Participants and data collection", "Statistical analysis", "Ethics statement", "RESULTS", "Characteristics of the participants", "Details of the gastroscopy cases", "Survey’s results I: participant’s gastroscopy experience after education", "Survey’s results II: current status of surgeons’ gastroscopy", "Survey’s results III: subjective opinions of the participants", "DISCUSSION" ]
[ "Evaluation of the stomach before and after surgery using gastroscopy is essential for treating various diseases of the stomach, including malignancy.12 In particular, gastroscopy can be used for clinical staging, tumor localization, treatment of early gastric cancer, complication management, and post-gastrectomy surveillance.3456\nIn Korea, most gastroscopies are performed by a gastroenterologist, and they are involved in all aspects of a broad endoscopic area.7 Gastroenterologists usually focus on diagnosing and treating a wide range of diseases using endoscopy, not only malignancy but also benign diseases such as motility disorders or functional diseases. Therefore, the endoscopy education system for gastroenterologists is more systematic, detailed, and strict to produce endoscopy experts.89 In contrast, gastrointestinal (GI) surgeons are more interested in the narrow area of endoscopy, such as localization for gastrectomy, intraoperative gastroscopy, and post-gastrectomy surveillance.1011 In addition, gastroscopy education for GI surgeons was not much developed due to surgeons’ low interest.\nCurrently, the necessity of surgeons’ endoscopy is being raised because minimal invasive approach is adopted and performed actively in every field of surgery. In particular, intraoperative endoscopy has an important role as an assistant of minimal invasive surgery. Several studies reported the advantages of intraoperative gastroscopy in terms of precise localization of the tumor during laparoscopic surgery, and image-guided surgery using indocyanine green (ICG).2121314 In such situation, education system of endoscopy for surgeon is needed to increase the surgeon’s understanding of gastroscopy, meet the increasing demands of intraoperative gastroscopy, and to make a smooth communication between gastroenterologists and GI surgeons.1516 However, there is a lack of a well-organized gastroscopy education program for surgeons in Korea.\nThis study aimed to describe the first systematic gastroscopy education program for surgeons in Korea.", "[SUBTITLE] Education program [SUBSECTION] The surgeon’s gastroscopy education program at Seoul St. Mary’s Hospital started in 2010. Initially, it was conducted for senior surgical residents and clinical fellows who trained at the Division of Gastrointestinal Surgery, Department of Surgery of Seoul St. Mary’s Hospital. In August 2017, a systematic gastroscopy education program, entitled the Gastroscopy School for Surgeons (GSS), was launched.\nThe GSS is divided into 12 and 20 weeks, depending on the breadth and depth of training. The 12-week course is aimed at senior surgical residents and general surgeons (including colorectal, hepatobiliopancreatic, breast, general surgeons, but excluding GI surgeons). Training before clinical practice consists of an introductory lecture, dry lab hands-on training, and simulations. The introductory lecture includes 1) sedation and patient monitoring; 2) indications, limitations, and contraindications of gastroscopic procedure; 3) complications and management; 4) preparation and informed consent with the ethical issue; 5) various gastroscopic findings; 6) writing reports of gastroscopic findings.8 During the clinical practice component, gastroscope insertion and luminal observation, post-gastrectomy surveillance, biopsy, clipping, etc., are taught. Approximately 30 to 100 cases of gastroscopy on actual patients are performed. The 20-week course is specifically designed for the clinical fellows of GI surgery and GI surgeons. In addition to content from the 12-week course, advanced techniques such as intraoperative gastroscopy, tumor localization with dye injection, polypectomy, and balloon dilatation are included, and approximately 100 to 180 cases are performed.\nAll trainers are GI surgeons, with at least several years of experience in gastroscopy. More than 1,000 cases are performed by each trainer per year. Trainer skills were standardized through meetings and consensus before teaching in the GSS.\nThe surgeon’s gastroscopy education program at Seoul St. Mary’s Hospital started in 2010. Initially, it was conducted for senior surgical residents and clinical fellows who trained at the Division of Gastrointestinal Surgery, Department of Surgery of Seoul St. Mary’s Hospital. In August 2017, a systematic gastroscopy education program, entitled the Gastroscopy School for Surgeons (GSS), was launched.\nThe GSS is divided into 12 and 20 weeks, depending on the breadth and depth of training. The 12-week course is aimed at senior surgical residents and general surgeons (including colorectal, hepatobiliopancreatic, breast, general surgeons, but excluding GI surgeons). Training before clinical practice consists of an introductory lecture, dry lab hands-on training, and simulations. The introductory lecture includes 1) sedation and patient monitoring; 2) indications, limitations, and contraindications of gastroscopic procedure; 3) complications and management; 4) preparation and informed consent with the ethical issue; 5) various gastroscopic findings; 6) writing reports of gastroscopic findings.8 During the clinical practice component, gastroscope insertion and luminal observation, post-gastrectomy surveillance, biopsy, clipping, etc., are taught. Approximately 30 to 100 cases of gastroscopy on actual patients are performed. The 20-week course is specifically designed for the clinical fellows of GI surgery and GI surgeons. In addition to content from the 12-week course, advanced techniques such as intraoperative gastroscopy, tumor localization with dye injection, polypectomy, and balloon dilatation are included, and approximately 100 to 180 cases are performed.\nAll trainers are GI surgeons, with at least several years of experience in gastroscopy. More than 1,000 cases are performed by each trainer per year. Trainer skills were standardized through meetings and consensus before teaching in the GSS.\n[SUBTITLE] Assessment [SUBSECTION] The actual clinical practice included scope handling, luminal observation, scope insertion, mouth to mouth, and additional procedures. During the training, the trainer evaluates the trainee’s skill using Miller’s Prism, and each step must be performed excellently before progressing to the next step.17 Self-assessment of the learner’s skill level before and one year after training was recorded using the Kirkpatrick-Phillips model.18 The current status of the surgeon’s gastroscopy experience in each participant’s institution and their perception of gastroscopy education in Korea were recorded through a post-training survey.18 The survey form is shown in Supplementary Data 1.\nThe actual clinical practice included scope handling, luminal observation, scope insertion, mouth to mouth, and additional procedures. During the training, the trainer evaluates the trainee’s skill using Miller’s Prism, and each step must be performed excellently before progressing to the next step.17 Self-assessment of the learner’s skill level before and one year after training was recorded using the Kirkpatrick-Phillips model.18 The current status of the surgeon’s gastroscopy experience in each participant’s institution and their perception of gastroscopy education in Korea were recorded through a post-training survey.18 The survey form is shown in Supplementary Data 1.\n[SUBTITLE] Participants and data collection [SUBSECTION] Participants who registered for the GSS and trained between August 2017 and November 2019 were enrolled in this study. The participants were recruited through a brochure and promotion through the Korean Surgical Society, the Korean Gastric Cancer Association, and the Korean Association of Surgeons. To ensure high-quality education, the trainer and trainee were matched 1:1, and the trainee’s performance was evaluated by the trainer during the training.\nGastroscopy was performed on patients who underwent outpatient gastroscopy at the Division of Gastrointestinal Surgery of Seoul St. Mary’s Hospital. Prospectively collected data on the condition of the stomach, reconstruction method, and procedures during gastroscopy were analyzed.\nParticipants who registered for the GSS and trained between August 2017 and November 2019 were enrolled in this study. The participants were recruited through a brochure and promotion through the Korean Surgical Society, the Korean Gastric Cancer Association, and the Korean Association of Surgeons. To ensure high-quality education, the trainer and trainee were matched 1:1, and the trainee’s performance was evaluated by the trainer during the training.\nGastroscopy was performed on patients who underwent outpatient gastroscopy at the Division of Gastrointestinal Surgery of Seoul St. Mary’s Hospital. Prospectively collected data on the condition of the stomach, reconstruction method, and procedures during gastroscopy were analyzed.\n[SUBTITLE] Statistical analysis [SUBSECTION] Descriptive statistics were reported as numbers (percentages) and medians (interquartile ranges). Categorical variables were analyzed using the χ2 test and Fisher’s exact test to compare the groups. Statistical significance was set at P < 0.05. All statistical analyses were performed using SPSS for Windows (ver. 21.0; SPSS, Inc., Chicago, IL, USA).\nDescriptive statistics were reported as numbers (percentages) and medians (interquartile ranges). Categorical variables were analyzed using the χ2 test and Fisher’s exact test to compare the groups. Statistical significance was set at P < 0.05. All statistical analyses were performed using SPSS for Windows (ver. 21.0; SPSS, Inc., Chicago, IL, USA).\n[SUBTITLE] Ethics statement [SUBSECTION] The present study was approved by the Institutional Review Board of Seoul St. Mary’s Hospital (KC20QISI0077). Written informed consent was obtained from all the patients and participants.\nThe present study was approved by the Institutional Review Board of Seoul St. Mary’s Hospital (KC20QISI0077). Written informed consent was obtained from all the patients and participants.", "The surgeon’s gastroscopy education program at Seoul St. Mary’s Hospital started in 2010. Initially, it was conducted for senior surgical residents and clinical fellows who trained at the Division of Gastrointestinal Surgery, Department of Surgery of Seoul St. Mary’s Hospital. In August 2017, a systematic gastroscopy education program, entitled the Gastroscopy School for Surgeons (GSS), was launched.\nThe GSS is divided into 12 and 20 weeks, depending on the breadth and depth of training. The 12-week course is aimed at senior surgical residents and general surgeons (including colorectal, hepatobiliopancreatic, breast, general surgeons, but excluding GI surgeons). Training before clinical practice consists of an introductory lecture, dry lab hands-on training, and simulations. The introductory lecture includes 1) sedation and patient monitoring; 2) indications, limitations, and contraindications of gastroscopic procedure; 3) complications and management; 4) preparation and informed consent with the ethical issue; 5) various gastroscopic findings; 6) writing reports of gastroscopic findings.8 During the clinical practice component, gastroscope insertion and luminal observation, post-gastrectomy surveillance, biopsy, clipping, etc., are taught. Approximately 30 to 100 cases of gastroscopy on actual patients are performed. The 20-week course is specifically designed for the clinical fellows of GI surgery and GI surgeons. In addition to content from the 12-week course, advanced techniques such as intraoperative gastroscopy, tumor localization with dye injection, polypectomy, and balloon dilatation are included, and approximately 100 to 180 cases are performed.\nAll trainers are GI surgeons, with at least several years of experience in gastroscopy. More than 1,000 cases are performed by each trainer per year. Trainer skills were standardized through meetings and consensus before teaching in the GSS.", "The actual clinical practice included scope handling, luminal observation, scope insertion, mouth to mouth, and additional procedures. During the training, the trainer evaluates the trainee’s skill using Miller’s Prism, and each step must be performed excellently before progressing to the next step.17 Self-assessment of the learner’s skill level before and one year after training was recorded using the Kirkpatrick-Phillips model.18 The current status of the surgeon’s gastroscopy experience in each participant’s institution and their perception of gastroscopy education in Korea were recorded through a post-training survey.18 The survey form is shown in Supplementary Data 1.", "Participants who registered for the GSS and trained between August 2017 and November 2019 were enrolled in this study. The participants were recruited through a brochure and promotion through the Korean Surgical Society, the Korean Gastric Cancer Association, and the Korean Association of Surgeons. To ensure high-quality education, the trainer and trainee were matched 1:1, and the trainee’s performance was evaluated by the trainer during the training.\nGastroscopy was performed on patients who underwent outpatient gastroscopy at the Division of Gastrointestinal Surgery of Seoul St. Mary’s Hospital. Prospectively collected data on the condition of the stomach, reconstruction method, and procedures during gastroscopy were analyzed.", "Descriptive statistics were reported as numbers (percentages) and medians (interquartile ranges). Categorical variables were analyzed using the χ2 test and Fisher’s exact test to compare the groups. Statistical significance was set at P < 0.05. All statistical analyses were performed using SPSS for Windows (ver. 21.0; SPSS, Inc., Chicago, IL, USA).", "The present study was approved by the Institutional Review Board of Seoul St. Mary’s Hospital (KC20QISI0077). Written informed consent was obtained from all the patients and participants.", "[SUBTITLE] Characteristics of the participants [SUBSECTION] In total, 33 participants were included in this study. The mean age was 35 years old. Twenty-five participants completed the 12-week course, and eight the 20-week course. The median number of gastroscopy cases was 62. The trainees consisted of three GI surgeons, nine general surgeons, 20 surgical residents, and one physician (Table 1).\nValues are presented as number (%) or number (interquartile range).\nGI= gastrointestinal.\nIn total, 33 participants were included in this study. The mean age was 35 years old. Twenty-five participants completed the 12-week course, and eight the 20-week course. The median number of gastroscopy cases was 62. The trainees consisted of three GI surgeons, nine general surgeons, 20 surgical residents, and one physician (Table 1).\nValues are presented as number (%) or number (interquartile range).\nGI= gastrointestinal.\n[SUBTITLE] Details of the gastroscopy cases [SUBSECTION] Of the 2,272 cases of gastroscopy, 264 (11.6%) cases had no history of gastrectomy, and 2,008 (88.4%) cases had a history of any type of gastrectomy. Among the gastrectomy cases, the most common history of gastrectomy was distal gastrectomy (60.7%), followed by total gastrectomy (23.1%). Proximal gastrectomy, gastric wedge resection, and other types of gastrectomy were reported at 0.4%, 4.1%, and 0.1%, respectively. The reconstruction method after 1,378 distal gastrectomies consisted of Billroth I gastroduodenostomy (12.6%), Billroth II gastrojejunostomy (78.4%), and Roux-en-Y gastrojejunostomy (9.1%). Roux-en-Y esophagojejunostomy was performed for all gastrectomy cases. After proximal gastrectomy, double-tract reconstruction was performed in all patients (Fig. 1). Among the 2,272 cases of gastroscopy, additional procedures (other than luminal observation) performed were biopsy (18.0%), clipping (0.6%), injection (0.2%), dye spreading (0.4%), and balloon dilatation (0.6%). Severe gag reflex was the most common adverse event during a gastroscopy (29 cases). Mucosal injury, minor bleeding, and incomplete luminal observation were observed in one, nine, and four cases, respectively. All adverse events were resolved without special management (Supplementary Table 1).\nTG = total gastrectomy, DG = distal gastrectomy, PG = proximal gastrectomy, WR = wedge resection, R-Y = Roux-en-Y.\nOf the 2,272 cases of gastroscopy, 264 (11.6%) cases had no history of gastrectomy, and 2,008 (88.4%) cases had a history of any type of gastrectomy. Among the gastrectomy cases, the most common history of gastrectomy was distal gastrectomy (60.7%), followed by total gastrectomy (23.1%). Proximal gastrectomy, gastric wedge resection, and other types of gastrectomy were reported at 0.4%, 4.1%, and 0.1%, respectively. The reconstruction method after 1,378 distal gastrectomies consisted of Billroth I gastroduodenostomy (12.6%), Billroth II gastrojejunostomy (78.4%), and Roux-en-Y gastrojejunostomy (9.1%). Roux-en-Y esophagojejunostomy was performed for all gastrectomy cases. After proximal gastrectomy, double-tract reconstruction was performed in all patients (Fig. 1). Among the 2,272 cases of gastroscopy, additional procedures (other than luminal observation) performed were biopsy (18.0%), clipping (0.6%), injection (0.2%), dye spreading (0.4%), and balloon dilatation (0.6%). Severe gag reflex was the most common adverse event during a gastroscopy (29 cases). Mucosal injury, minor bleeding, and incomplete luminal observation were observed in one, nine, and four cases, respectively. All adverse events were resolved without special management (Supplementary Table 1).\nTG = total gastrectomy, DG = distal gastrectomy, PG = proximal gastrectomy, WR = wedge resection, R-Y = Roux-en-Y.\n[SUBTITLE] Survey’s results I: participant’s gastroscopy experience after education [SUBSECTION] After more than one year of training, a self-assessment of gastroscopy performance survey was completed by all 33 participants. One year after the training, the participants’ positions were seven GI surgeons, twenty-three general surgeons, two surgical residents, and one physician (Table 1). Currently, three participants performed gastroscopy as their main work (3 or more times/week), and seven performed gastroscopies weekly or more. In contrast, 16 participants have not performed gastroscopy in their work (Fig. 2A). One participant self-assessed their gastroscopy skills to be at the expert level (able to train others), and 25 evaluated themselves as good performers (without the need for supervision) or better. Notably, not a single participant evaluated themselves as incompetent in gastroscopy (Fig. 2B). All participants were able to perform basic procedures, such as luminal observation and biopsy. A total of 19 patients underwent minor procedures, such as clipping, injection, dye spreading, or balloon dilatation, and four underwent major procedures, such as stent insertion, polypectomy, or endoscopic submucosal dissection. Moreover, ten participants were able to perform intraoperative gastroscopy, and one was able to perform advanced procedures, such as endoscopic retrograde cholangiopancreatography (ERCP) or enteroscopy (Fig. 2). There was no significant difference in the skills and confidence level of the participants regardless of the training course (12-week vs. 20-week) completed. However, the skills level was relatively lower among participants who were trained in less than 60 cases (Supplementary Table 2).\nAfter more than one year of training, a self-assessment of gastroscopy performance survey was completed by all 33 participants. One year after the training, the participants’ positions were seven GI surgeons, twenty-three general surgeons, two surgical residents, and one physician (Table 1). Currently, three participants performed gastroscopy as their main work (3 or more times/week), and seven performed gastroscopies weekly or more. In contrast, 16 participants have not performed gastroscopy in their work (Fig. 2A). One participant self-assessed their gastroscopy skills to be at the expert level (able to train others), and 25 evaluated themselves as good performers (without the need for supervision) or better. Notably, not a single participant evaluated themselves as incompetent in gastroscopy (Fig. 2B). All participants were able to perform basic procedures, such as luminal observation and biopsy. A total of 19 patients underwent minor procedures, such as clipping, injection, dye spreading, or balloon dilatation, and four underwent major procedures, such as stent insertion, polypectomy, or endoscopic submucosal dissection. Moreover, ten participants were able to perform intraoperative gastroscopy, and one was able to perform advanced procedures, such as endoscopic retrograde cholangiopancreatography (ERCP) or enteroscopy (Fig. 2). There was no significant difference in the skills and confidence level of the participants regardless of the training course (12-week vs. 20-week) completed. However, the skills level was relatively lower among participants who were trained in less than 60 cases (Supplementary Table 2).\n[SUBTITLE] Survey’s results II: current status of surgeons’ gastroscopy [SUBSECTION] The survey also collected information on the gastroscopies performed by surgeons at the participant’s institution. At the time of the survey, 20 participants worked at seven tertiary university hospitals, seven were in private clinics, and six were in military service for their duty. The role of the surgeon in performing gastroscopy was diversely distributed by institutions, from all procedures were performed for all the cases to impossible to perform any procedures at all (Fig. 3A). There was no systematic gastroscopy education program for surgeons in any of the institutions except Seoul St. Mary’s Hospital (our institution). There were a few smaller training sessions conducted by surgeons or gastroenterologists (Fig. 3B). Despite the lack of formal gastroscopy training, surgeons reported that more than half of intraoperative gastroscopy procedures were performed by surgeons (Fig. 3C).\nThe survey also collected information on the gastroscopies performed by surgeons at the participant’s institution. At the time of the survey, 20 participants worked at seven tertiary university hospitals, seven were in private clinics, and six were in military service for their duty. The role of the surgeon in performing gastroscopy was diversely distributed by institutions, from all procedures were performed for all the cases to impossible to perform any procedures at all (Fig. 3A). There was no systematic gastroscopy education program for surgeons in any of the institutions except Seoul St. Mary’s Hospital (our institution). There were a few smaller training sessions conducted by surgeons or gastroenterologists (Fig. 3B). Despite the lack of formal gastroscopy training, surgeons reported that more than half of intraoperative gastroscopy procedures were performed by surgeons (Fig. 3C).\n[SUBTITLE] Survey’s results III: subjective opinions of the participants [SUBSECTION] All participants recognized that gastroscopy was important to surgeons (Fig. 4A). The lack of a systemic education system was considered the primary barrier to surgeons performing gastroscopy. Additionally, conflict with the other departments was also recognized as a major hurdle. Furthermore, there was a perceived lack of interest among surgeons and the excessive workload was also problematic (Fig. 4B). Most participants stated that the gastroscopy education system for surgeons in Korea was insufficient (Fig. 4C).\nThe participants provided their subjective opinions on the surgeon’s gastroscopy in the survey. A total of 87.9% identified the current problems associated with gastroscopy and highlighted the limitations of preoperative information and postoperative complication management when gastroscopy is performed by a non-surgeon, such as a gastroenterologist. The lack of a gastroscopy training system was also articulated. Second, 90.9% responded to the question regarding the merits of surgeons performing gastroscopies. The participants said that surgeons’ gastroscopy is useful in determining the detailed surgical plan preoperatively, and various reconstruction methods can be easily determined during postoperative surveillance. Finally, 54.5% provided suggestions for the development of the surgeon’s gastroscopy. Participants confirmed that it is necessary to increase the quality and quantity of gastroscopy education, with more training centers, and official support from professional associations and societies (Supplementary Table 3).\nAll participants recognized that gastroscopy was important to surgeons (Fig. 4A). The lack of a systemic education system was considered the primary barrier to surgeons performing gastroscopy. Additionally, conflict with the other departments was also recognized as a major hurdle. Furthermore, there was a perceived lack of interest among surgeons and the excessive workload was also problematic (Fig. 4B). Most participants stated that the gastroscopy education system for surgeons in Korea was insufficient (Fig. 4C).\nThe participants provided their subjective opinions on the surgeon’s gastroscopy in the survey. A total of 87.9% identified the current problems associated with gastroscopy and highlighted the limitations of preoperative information and postoperative complication management when gastroscopy is performed by a non-surgeon, such as a gastroenterologist. The lack of a gastroscopy training system was also articulated. Second, 90.9% responded to the question regarding the merits of surgeons performing gastroscopies. The participants said that surgeons’ gastroscopy is useful in determining the detailed surgical plan preoperatively, and various reconstruction methods can be easily determined during postoperative surveillance. Finally, 54.5% provided suggestions for the development of the surgeon’s gastroscopy. Participants confirmed that it is necessary to increase the quality and quantity of gastroscopy education, with more training centers, and official support from professional associations and societies (Supplementary Table 3).", "In total, 33 participants were included in this study. The mean age was 35 years old. Twenty-five participants completed the 12-week course, and eight the 20-week course. The median number of gastroscopy cases was 62. The trainees consisted of three GI surgeons, nine general surgeons, 20 surgical residents, and one physician (Table 1).\nValues are presented as number (%) or number (interquartile range).\nGI= gastrointestinal.", "Of the 2,272 cases of gastroscopy, 264 (11.6%) cases had no history of gastrectomy, and 2,008 (88.4%) cases had a history of any type of gastrectomy. Among the gastrectomy cases, the most common history of gastrectomy was distal gastrectomy (60.7%), followed by total gastrectomy (23.1%). Proximal gastrectomy, gastric wedge resection, and other types of gastrectomy were reported at 0.4%, 4.1%, and 0.1%, respectively. The reconstruction method after 1,378 distal gastrectomies consisted of Billroth I gastroduodenostomy (12.6%), Billroth II gastrojejunostomy (78.4%), and Roux-en-Y gastrojejunostomy (9.1%). Roux-en-Y esophagojejunostomy was performed for all gastrectomy cases. After proximal gastrectomy, double-tract reconstruction was performed in all patients (Fig. 1). Among the 2,272 cases of gastroscopy, additional procedures (other than luminal observation) performed were biopsy (18.0%), clipping (0.6%), injection (0.2%), dye spreading (0.4%), and balloon dilatation (0.6%). Severe gag reflex was the most common adverse event during a gastroscopy (29 cases). Mucosal injury, minor bleeding, and incomplete luminal observation were observed in one, nine, and four cases, respectively. All adverse events were resolved without special management (Supplementary Table 1).\nTG = total gastrectomy, DG = distal gastrectomy, PG = proximal gastrectomy, WR = wedge resection, R-Y = Roux-en-Y.", "After more than one year of training, a self-assessment of gastroscopy performance survey was completed by all 33 participants. One year after the training, the participants’ positions were seven GI surgeons, twenty-three general surgeons, two surgical residents, and one physician (Table 1). Currently, three participants performed gastroscopy as their main work (3 or more times/week), and seven performed gastroscopies weekly or more. In contrast, 16 participants have not performed gastroscopy in their work (Fig. 2A). One participant self-assessed their gastroscopy skills to be at the expert level (able to train others), and 25 evaluated themselves as good performers (without the need for supervision) or better. Notably, not a single participant evaluated themselves as incompetent in gastroscopy (Fig. 2B). All participants were able to perform basic procedures, such as luminal observation and biopsy. A total of 19 patients underwent minor procedures, such as clipping, injection, dye spreading, or balloon dilatation, and four underwent major procedures, such as stent insertion, polypectomy, or endoscopic submucosal dissection. Moreover, ten participants were able to perform intraoperative gastroscopy, and one was able to perform advanced procedures, such as endoscopic retrograde cholangiopancreatography (ERCP) or enteroscopy (Fig. 2). There was no significant difference in the skills and confidence level of the participants regardless of the training course (12-week vs. 20-week) completed. However, the skills level was relatively lower among participants who were trained in less than 60 cases (Supplementary Table 2).", "The survey also collected information on the gastroscopies performed by surgeons at the participant’s institution. At the time of the survey, 20 participants worked at seven tertiary university hospitals, seven were in private clinics, and six were in military service for their duty. The role of the surgeon in performing gastroscopy was diversely distributed by institutions, from all procedures were performed for all the cases to impossible to perform any procedures at all (Fig. 3A). There was no systematic gastroscopy education program for surgeons in any of the institutions except Seoul St. Mary’s Hospital (our institution). There were a few smaller training sessions conducted by surgeons or gastroenterologists (Fig. 3B). Despite the lack of formal gastroscopy training, surgeons reported that more than half of intraoperative gastroscopy procedures were performed by surgeons (Fig. 3C).", "All participants recognized that gastroscopy was important to surgeons (Fig. 4A). The lack of a systemic education system was considered the primary barrier to surgeons performing gastroscopy. Additionally, conflict with the other departments was also recognized as a major hurdle. Furthermore, there was a perceived lack of interest among surgeons and the excessive workload was also problematic (Fig. 4B). Most participants stated that the gastroscopy education system for surgeons in Korea was insufficient (Fig. 4C).\nThe participants provided their subjective opinions on the surgeon’s gastroscopy in the survey. A total of 87.9% identified the current problems associated with gastroscopy and highlighted the limitations of preoperative information and postoperative complication management when gastroscopy is performed by a non-surgeon, such as a gastroenterologist. The lack of a gastroscopy training system was also articulated. Second, 90.9% responded to the question regarding the merits of surgeons performing gastroscopies. The participants said that surgeons’ gastroscopy is useful in determining the detailed surgical plan preoperatively, and various reconstruction methods can be easily determined during postoperative surveillance. Finally, 54.5% provided suggestions for the development of the surgeon’s gastroscopy. Participants confirmed that it is necessary to increase the quality and quantity of gastroscopy education, with more training centers, and official support from professional associations and societies (Supplementary Table 3).", "Historically, endoscopy was initiated by surgeons. After Mikulicz performed the first practical gastroscopy in 1880, the first sigmoidoscopy in 1895, the first ERCP in 1968, and the first colonoscopic polypectomy in 1969, were all performed by surgeons.1920 Currently, the clinician responsible for performing endoscopy differ among countries. In some countries, both gastroenterologists and surgeons are major providers. In other countries, gastroenterologists perform most of the endoscopy.2122 In Korea, most endoscopies are performed by gastroenterologists. Surgeons or physicians perform endoscopies only in a few hospitals. Therefore, access to endoscopy among surgeons is not very common. Several studies have reported the importance of providing tailored treatment by determining the extent of surgery based on precise preoperative or intraoperative localization.2111223 Moreover, studies on intraoperative gastroscopy, such as fluorescence image-guided surgery using ICG or laparoscopic-endoscopic cooperative surgery, have been actively reported.1424252627 Gastroscopy is also widely used for post-gastrectomy complication management.52829 Currently, reconstruction after gastrectomy is becoming increasingly complex and diversified. Hence, it is critical for the clinician performing gastroscopic surveillance to have an excellent understanding of post-gastrectomy anatomy.303132 As the use of endoscopy during and after surgery has increased, the importance of the surgeon’s understanding of endoscopy has been further emphasized. Furthermore, as shown in the present study, surgeons are increasingly interested in learning and performing gastroscopies.\nPractical and systematic education is essential for proper and safe endoscopy. There have been various reports of this in the gastroenterology field. The American Society for Gastrointestinal Endoscopy presented the principles for safe and effective endoscopy.3334 The Korean Society of Gastrointestinal Endoscopy also reported the status of endoscopy training for gastroenterologists.89 In contrast, there are limited reports on endoscopy training for surgeons. There are a few reports from professional associations and societies in Canada and the United States.1635363738 In contrast, there is only a single report in Korea: the initial gastroscopy training program for senior surgical residents and clinical fellows in GI surgery and the results related to the learning curve in 2012.39 After then, the importance of gastroscopy for surgeons and the need for gastroscopy training at the professional association and society level has increased. In response, the GSS, a systematic education program, was launched.\nThe GSS is a program without long history and has not been externally verified. Also, it is insufficient in terms of quantity and detail compared to the endoscopy education program implemented by the Korean Society of Gastrointestinal Endoscopy.8 However, the GSS is not designed to train at the same level as expert gastroenterologists or endoscopy specialists. The purpose of the GSS is to increase the understanding of gastroscopy and enable smooth communication with a gastroenterologist by learning basic gastroscopic techniques such as localization before or during surgery and routine surveillance after surgery as a GI surgeon. In addition, maximizing the advantages of intraoperative gastroscopy and improving surgical outcomes is another goal. Although the quantity of clinical practice of the GSS seems to be insufficient, several reports suggested the minimum quantity of clinical practice required to overcome the learning curve in limited endoscopic training for surgeons.163940 Moreover, the GSS consists of various educational methods, such as basic theory classes, dry lab hands-on practices, and simulations used for safety training. Trainers were all GI surgeons with extensive gastroscopy experience and matched 1:1 with trainees to conduct training. This allowed immediate and appropriate feedback for learning, and tailored training could also be provided. As a result, most of the participants could perform gastroscopy independently without a supervisor, and some had reached a level where they could train others. This result corresponds to the “Return On Investment” in the Kirkpatrick-Phillips model. Therefore, the results of this study revealed the effectiveness of a systematic gastroscopy education program for surgeons.18 Based on this report, the GSS should be developed more systematically and receive validation from an external judge of endoscopy experts in the future.\nIn Korea, surgeon’s gastroscopy is actively performed in a few institutions, while most institutions do not. In particular, only our institution has a systematic educational program for surgeons for gastroscopy education. Recognizing the importance of gastroscopy in surgery, several surgical societies, such as the Korean Surgical Society and the Korean Gastric Cancer Association, have widely promoted gastroscopy education programs in every aspect. As mentioned by the participants, available training programs are still insufficient at present. Surgeons’ gastroscopy education programs need to be developed through various routes, with the support and policy promotion at the professional association and society level. Every surgeon and surgical institution should have an interest in developing the surgeon’s gastroscopy skills and knowledge, including intra- or perioperative gastroscopy. Furthermore, it is critical to develop more training centers and reach a consensus with the gastroenterologists at the professional body level.\nThe present study has several limitations. First, the quantity of clinical practice of the GSS is insufficient compared to education program for the gastroenterologist. However, as mentioned above, since the final goal of the two educations is different, the GSS could be considered as consisting of the minimum requirement of educational content, duration, and quantity to achieve its final goal. The development of more educational programs from more training centers could provide a higher-quality education for surgeons in the future. Second, the survey was completed only by participants who sympathized with the importance of surgeons’ gastroscopy and felt the need for gastroscopy education. Also, a few questions tend to reflect only the subjective opinions of respondents. Although the selection bias and lack of objectivity, we believe it would be minimal, considering that surgeons’ interest and desire for gastroscopy training have already been voiced in many congresses and reports.15 In addition, one of the most critical purposes of this report is to awaken surgeons’ awareness of the importance of the development and expansion of the surgeon’s gastroscopy education program. Consequently, although the survey results could be subjective, such opinions would also be necessary. Third, the gastroscopy cases performed by participants in this study were largely limited to post-gastrectomy. For surgeons who have a better understanding of post-gastrectomy status than gastroenterologists, gastroscopy for post-gastrectomy status is important. In addition, to expand the surgeon’s scope of practice relating to gastroscopy, it is essential to focus on post-gastrectomy surveillance.\nIn conclusion, the present study introduced the first systematic gastroscopy education program for surgeons in Korea. Our systematic education could meet the clinical needs of surgeons. We also suggested a future direction for gastroscopy training among surgeons. For the expansion and development of gastroscopy among surgeons, the efforts of individual surgeons must be accompanied by the endeavor to expand infrastructure and training facilities for gastroscopy by surgeons at the society and institutional level." ]
[ "intro", "methods", null, null, null, null, "ethics-statement", "results", null, null, null, null, null, "discussion" ]
[ "Gastroscopy", "Education", "Surgery", "Gastrectomy" ]
Impact of COVID-19 Pandemic on Biomedical Publications and Their Citation Frequency.
36254532
The coronavirus disease 2019 (COVID-19) pandemic has resulted in enormous related publications. However, the citation frequency of these documents and their influence on the journal impact factor (JIF) are not well examined. We aimed to evaluate the impact of COVID-19 on biomedical research publications and their citation frequency.
BACKGROUND
We searched publications on biomedical research in the Web of Science using the search terms "COVID-19," "SARS-Cov-2," "2019 corona*," "corona virus disease 2019," "coronavirus disease 2019," "novel coronavirus infection" and "2019-ncov." The top 200 journals were defined as those with a higher number of COVID-19 publications than other journals in 2020. The COVID-19 impact ratio was calculated as the ratio of the average number of citations per item in 2021 to the JIF for 2020.
METHODS
The average number of citations for the top 200 journals in 2021, per item published in 2020, was 25.7 (range, 0-270). The average COVID-19 impact ratio was 3.84 (range, 0.26-16.58) for 197 journals that recorded the JIF for 2020. The average JIF ratio for the top 197 journals including the JIFs for 2020 and 2021 was 1.77 (range, 0.68-8.89). The COVID-19 impact ratio significantly correlated with the JIF ratio (r = 0.403, P = 0.010). Twenty-five Korean journals with a COVID-19 impact ratio > 1.5 demonstrated a higher JIF ratio (1.31 ± 0.39 vs. 1.01 ± 0.18, P < 0.001) than 33 Korean journals with a lower COVID-19 impact ratio.
RESULTS
COVID-19 pandemic infection has significantly impacted the trends in biomedical research and the citation of related publications.
CONCLUSION
[ "COVID-19", "Humans", "Journal Impact Factor", "Pandemics", "Publications", "SARS-CoV-2" ]
9577356
INTRODUCTION
The coronavirus disease 2019 (COVID-19) pandemic has evoked an unprecedented global health crisis.1 The prevention, diagnosis, and treatment of the new viral disease have been a major concern of biomedical societies in both academia and industry.23 Therefore, urgent and rapid communication of biomedical information is necessary. As a result, publications on COVID-19 contributed to a dramatic increase in the total number of biomedical publications, as well as their citation. More than 260,000 COVID-19-related articles have been published to date and listed in PubMed. We aimed to evaluate the impact of COVID-19 on biomedical research publications and their citation frequency.
METHODS
We searched publications on biomedical research in PubMed and the Web of Science (Clarivate Analytics) with the following search terms: “COVID-19,” “SARS-Cov-2,” “2019 corona*,” “corona virus disease 2019,” “coronavirus disease 2019” “novel coronavirus infection,” and “2019-ncov.” The top 200 journals were defined as those with a higher number of COVID-19 publications in the Web of Science than in other journals in 2020. The COVID-19 impact ratio was calculated as the ratio of the average citations per item in 2021 to the journal impact factor (JIF) for 2020. The JIF ratio was calculated as the ratio of the JIF for 2021 to that for 2020. The correlation between the COVID-19 impact ratio and JIF ratio was evaluated using the Pearson correlation test (SPSS version 27.0; IBM SPSS Statistics, Armonk, NY, USA). In addition, the COVID-19 impact ratio and JIF ratio were evaluated for Korean journals listed in the Web of Science. [SUBTITLE] Ethics statement [SUBSECTION] Institutional Review Board approval was waived for this study, because this study was based on publication data of the PubMed and Web of Science. Institutional Review Board approval was waived for this study, because this study was based on publication data of the PubMed and Web of Science.
RESULTS
As of June 30, 2022, 267,549 documents regarding COVID-19 have been published and listed in PubMed (National Library of Medicine) (50 in 2019, 91,548 in 2020, 136,961 in 2021, 67,372 in 2022 until June 30). Among these, 58,344 documents published in 2020 and 90,642 documents published in 2021 are listed in the Web of Science. Regarding the document types published in 2020, articles comprised 49.0% of all documents, review articles 11.3%, editorial materials 18.8%, and letters 20.9% (Table 1). Values are presented as number (%). COVID-19 = coronavirus disease 2019. According to the categories of the Web of Science, 7,444 documents (12.8%) in 2020 belonged to general medicine, followed by public environmental occupational health (5,013, 8.6%), infectious diseases (3,991, 6.8%), surgery (2,877, 4.9%), and immunology (2,797, 4.7%) (Table 2). They received 232,696 total citations (average citation per item, 31.3; top citation, 7,016) in 2021 in general medicine; 85,602 total citations (average citation per item, 17.1; top citation, 1,570) in 2021 in public environmental occupational health; 109,518 total citations (average citation per item, 27.4; top citation, 2,821) in 2021 in infectious diseases; 24,511 total citations (average citation per item, 8.5; top citation, 1,019) in 2021 in the surgery; and 86,393 total citations (average citations per item, 30.9; top citations, 2,821) in 2021 in immunology. Documents according to the categories recorded the highest of 73.3 citations per item. COVID-19 = coronavirus disease 2019. The average number of publications in 2020 in the top 200 journals was 126 (range, 57–778) (Table 3). The average number of total citations in 2021 for publications in 2020 was 3,671 (range, 4–54,682) for the top 200 journals. The average number of citations for the top 200 journals in 2021 per item published in 2020 was 25.7 (range, 0–270.0). The average COVID-19 impact ratio was 3.84 (range, 0.26–16.58) for 197 journals that recorded JIFs for 2020. The average JIF ratio for the top 197 journals that had JIFs for 2020 and 2021 was 1.77 (range, 0.68–8.89). The COVID-19 impact ratio significantly correlated with the JIF ratio (r = 0.403, P = 0.010; Fig. 1). For these journals, the JIFs for 2020 and 2021 strongly correlated (r = 0.939, P = 0.010). Journal of Biomolecular Structure Dynamics was excluded, as the journal had no record of JIF for 2020. Journal of Biomolecular Structure Dynamics had 262 COVID-19 publications in 2020, 3,961 total citations in 2021, and 15.1 citations per item on average in 2021. COVID-19 = coronavirus disease 2019, JIF = journal impact factor, JCR = Journal Citation Report. aThe JIF ratio was calculated as the ratio of the JIF for 2021 to the JIF for 2020. COVID-19 = coronavirus disease 2019, JIF = journal impact factor, JCR = Journal Citation Report. For the Korean journals, the average number of COVID-19 publications in 2020 in 58 Korean journals, with both JIF 2020 and JIF 2021 and listed in the Web of Science, was 5.50 (range, 0–126). They received 8.8 citations (range, 0–61.0) per item in 2021 (Table 4). The COVID-19 impact ratio was 2.97 on average (range, 0–24.67) for the Korean journals. The average JIF ratio for Korean journals was 1.14 (range, 0.50–2.49). Twenty-five Korean journals with a COVID-19 impact ratio > 1.5 demonstrated a higher JIF ratio (1.31 ± 0.39 vs. 1.01 ± 0.18, P < 0.001) than 33 Korean journals with a lower COVID-19 impact ratio. Journals with more than five COVID-19-related publications in 2020 or more than 100 citations in 2021 for COVID-19-related publications in 2020 were included. Korean Journal of Anesthesiology was excluded, as the journal had no record of JIF for 2020. Korean Journal of Anesthesiology had seven COVID-19 publications in 2020, 46 citations in 2021, and 6.6 citations per item on average in 2021. COVID-19 = coronavirus disease 2019, JIF = journal impact factor, JCR = Journal Citation Report. aThe JIF ratio was calculated as the ratio of the JIF for 2021 to the JIF for 2020. Among the journals in the category of “medicine general internal” of the Web of Science, top 20 journals, that recorded JIFs for 2020, showed 199 (range, 95–597) COVID-19 publications in average, average total citations of 9,805.0 (281–54,682), 36.2 average citations per item in 2021 (range, 2.4–164.7), COVID-19 impact ratio of 2.69 (range, 0.62–6.36) and JIF ratio of 1.86 (range, 0.96–4.62) (Supplementary Table 1). The Journal of Korean Medical Science showed the highest COVID-19 impact ratio (6.36) among the 20 journals in this category.
null
null
[]
[]
[]
[ "INTRODUCTION", "METHODS", "Ethics statement", "RESULTS", "DISCUSSION" ]
[ "The coronavirus disease 2019 (COVID-19) pandemic has evoked an unprecedented global health crisis.1 The prevention, diagnosis, and treatment of the new viral disease have been a major concern of biomedical societies in both academia and industry.23 Therefore, urgent and rapid communication of biomedical information is necessary. As a result, publications on COVID-19 contributed to a dramatic increase in the total number of biomedical publications, as well as their citation. More than 260,000 COVID-19-related articles have been published to date and listed in PubMed. We aimed to evaluate the impact of COVID-19 on biomedical research publications and their citation frequency.", "We searched publications on biomedical research in PubMed and the Web of Science (Clarivate Analytics) with the following search terms: “COVID-19,” “SARS-Cov-2,” “2019 corona*,” “corona virus disease 2019,” “coronavirus disease 2019” “novel coronavirus infection,” and “2019-ncov.” The top 200 journals were defined as those with a higher number of COVID-19 publications in the Web of Science than in other journals in 2020. The COVID-19 impact ratio was calculated as the ratio of the average citations per item in 2021 to the journal impact factor (JIF) for 2020. The JIF ratio was calculated as the ratio of the JIF for 2021 to that for 2020. The correlation between the COVID-19 impact ratio and JIF ratio was evaluated using the Pearson correlation test (SPSS version 27.0; IBM SPSS Statistics, Armonk, NY, USA). In addition, the COVID-19 impact ratio and JIF ratio were evaluated for Korean journals listed in the Web of Science.\n[SUBTITLE] Ethics statement [SUBSECTION] Institutional Review Board approval was waived for this study, because this study was based on publication data of the PubMed and Web of Science.\nInstitutional Review Board approval was waived for this study, because this study was based on publication data of the PubMed and Web of Science.", "Institutional Review Board approval was waived for this study, because this study was based on publication data of the PubMed and Web of Science.", "As of June 30, 2022, 267,549 documents regarding COVID-19 have been published and listed in PubMed (National Library of Medicine) (50 in 2019, 91,548 in 2020, 136,961 in 2021, 67,372 in 2022 until June 30). Among these, 58,344 documents published in 2020 and 90,642 documents published in 2021 are listed in the Web of Science. Regarding the document types published in 2020, articles comprised 49.0% of all documents, review articles 11.3%, editorial materials 18.8%, and letters 20.9% (Table 1).\nValues are presented as number (%).\nCOVID-19 = coronavirus disease 2019.\nAccording to the categories of the Web of Science, 7,444 documents (12.8%) in 2020 belonged to general medicine, followed by public environmental occupational health (5,013, 8.6%), infectious diseases (3,991, 6.8%), surgery (2,877, 4.9%), and immunology (2,797, 4.7%) (Table 2). They received 232,696 total citations (average citation per item, 31.3; top citation, 7,016) in 2021 in general medicine; 85,602 total citations (average citation per item, 17.1; top citation, 1,570) in 2021 in public environmental occupational health; 109,518 total citations (average citation per item, 27.4; top citation, 2,821) in 2021 in infectious diseases; 24,511 total citations (average citation per item, 8.5; top citation, 1,019) in 2021 in the surgery; and 86,393 total citations (average citations per item, 30.9; top citations, 2,821) in 2021 in immunology. Documents according to the categories recorded the highest of 73.3 citations per item.\nCOVID-19 = coronavirus disease 2019.\nThe average number of publications in 2020 in the top 200 journals was 126 (range, 57–778) (Table 3). The average number of total citations in 2021 for publications in 2020 was 3,671 (range, 4–54,682) for the top 200 journals. The average number of citations for the top 200 journals in 2021 per item published in 2020 was 25.7 (range, 0–270.0). The average COVID-19 impact ratio was 3.84 (range, 0.26–16.58) for 197 journals that recorded JIFs for 2020. The average JIF ratio for the top 197 journals that had JIFs for 2020 and 2021 was 1.77 (range, 0.68–8.89). The COVID-19 impact ratio significantly correlated with the JIF ratio (r = 0.403, P = 0.010; Fig. 1). For these journals, the JIFs for 2020 and 2021 strongly correlated (r = 0.939, P = 0.010).\n\nJournal of Biomolecular Structure Dynamics was excluded, as the journal had no record of JIF for 2020. Journal of Biomolecular Structure Dynamics had 262 COVID-19 publications in 2020, 3,961 total citations in 2021, and 15.1 citations per item on average in 2021.\nCOVID-19 = coronavirus disease 2019, JIF = journal impact factor, JCR = Journal Citation Report.\naThe JIF ratio was calculated as the ratio of the JIF for 2021 to the JIF for 2020.\nCOVID-19 = coronavirus disease 2019, JIF = journal impact factor, JCR = Journal Citation Report.\nFor the Korean journals, the average number of COVID-19 publications in 2020 in 58 Korean journals, with both JIF 2020 and JIF 2021 and listed in the Web of Science, was 5.50 (range, 0–126). They received 8.8 citations (range, 0–61.0) per item in 2021 (Table 4). The COVID-19 impact ratio was 2.97 on average (range, 0–24.67) for the Korean journals. The average JIF ratio for Korean journals was 1.14 (range, 0.50–2.49). Twenty-five Korean journals with a COVID-19 impact ratio > 1.5 demonstrated a higher JIF ratio (1.31 ± 0.39 vs. 1.01 ± 0.18, P < 0.001) than 33 Korean journals with a lower COVID-19 impact ratio.\nJournals with more than five COVID-19-related publications in 2020 or more than 100 citations in 2021 for COVID-19-related publications in 2020 were included. Korean Journal of Anesthesiology was excluded, as the journal had no record of JIF for 2020. Korean Journal of Anesthesiology had seven COVID-19 publications in 2020, 46 citations in 2021, and 6.6 citations per item on average in 2021.\nCOVID-19 = coronavirus disease 2019, JIF = journal impact factor, JCR = Journal Citation Report.\naThe JIF ratio was calculated as the ratio of the JIF for 2021 to the JIF for 2020.\nAmong the journals in the category of “medicine general internal” of the Web of Science, top 20 journals, that recorded JIFs for 2020, showed 199 (range, 95–597) COVID-19 publications in average, average total citations of 9,805.0 (281–54,682), 36.2 average citations per item in 2021 (range, 2.4–164.7), COVID-19 impact ratio of 2.69 (range, 0.62–6.36) and JIF ratio of 1.86 (range, 0.96–4.62) (Supplementary Table 1). The Journal of Korean Medical Science showed the highest COVID-19 impact ratio (6.36) among the 20 journals in this category.", "Our study showed that COVID-19 pandemic influenced the citation frequency of publications and that a 3.8-fold increase in citations in 2021 for the documents published in 2020 as compared with JIF 2020 was observed in the top 197 journals with a higher number of COVID-19 publications. The average JIF ratio for the top 197 journals with JIFs for 2020 and 2021 was 1.77, and the COVID-19 impact ratio significantly correlated with the JIF ratio. Korean journals with a COVID-19 impact ratio > 1.5 showed a higher JIF ratio than those with a lower COVID-19 impact ratio. General medicine in the Web of Science category, had the most documents (12.8%) published in 2020, followed by public environmental occupational health (8.6%), infectious diseases (6.8%), surgery (4.9%), and immunology (4.7%).\nGong et al.4 found that the number of COVID-19-related publications demonstrated a high growth trend in the first 10 days of February 2020, and that China published the largest number of studies, as the country was the most affected by the pandemic in its early stages. The Scopus database search for the first year of the COVID-19 pandemic between January 1, 2020, and December 31, 2020, revealed that, among the 20 highest-ranked countries by the gross domestic product, the United States of America was the most productive country (n = 13,491) in the COVID-19 and COVID-19-related domains, with one and a half times or more publications than any other country.5\nAnother bibliometric analysis of COVID-19 during the early stages of the outbreak (December 2019 to June 19, 2020) indicated that articles (9,140 of 19,044 publications, 48.0%) were the most prevalent document type, followed by letters (4,192, 22.0%) and reviews (1,797, 9.4%).6 According to another analysis of the early phase publications, in the 16,670 relevant articles dated between February 14, 2020, and June 1, 2020, the most common topics were health care responses (2,812, 16.9%) and clinical manifestations (1,828, 10.9%).7 Research on clinical manifestations and protective measures has shown an increasing trend, whereas research on disease transmission, epidemiology, health care response, and radiology demonstrates a decreasing trend.7 Artificial intelligence-based bibliometric analysis has shown the potential of exploring many academic publications during a public health crisis. The dominant topics related to the spread of COVID-19 were public health response, clinical care practices, clinical characteristics, risk factors, and epidemic models.8\nAn analysis of the recent documents published between January 1, 2019 and January 1, 2021, in 10 high-impact medical and infectious disease journals (New England Journal of Medicine, Lancet, Journal of the American Medical Association, Nature Medicine, British Medical Journal, Annals of Internal Medicine, Lancet Global Health, Lancet Public Health, Lancet Infectious Disease, and Clinical Infectious Disease) showed that 1,022 (16.2%) of 6,319 studies were related to COVID-19.9 In this meta-analysis, the authors estimated that the COVID pandemic was associated with an 18% decrease in the production of non-COVID-19 documents owing to editorial strategies using simulation models. They also found a significant decrease in the number of original articles for COVID-19 research compared with non-COVID-19 research (47.9% vs. 71.3%, P <  0.001). More authors were associated with COVID-19 publications, especially case reports, than with non-COVID-19 publications (median, 9.0 authors ([interquartile range {IQR}, 6.0–13.0] vs. 4.0 [IQR, 3.0–6.0], P <  0.001). The scientific quality of COVID-19-themed research was estimated to be below average with lower levels of evidence during the early period of the COVID-19 pandemic (from March 12 to April 12, 2020) in the three highest-ranked journals (New England Journal of Medicine, Journal of the American Medical Association, Lancet).10\nA study reported an increase in the retraction rate related to an upsurge in the publication rate from January 1, 2020 to October 10, 2021.11 The data from the Retraction Watch database (http://retractiondatabase.org/) showed a total of 157 withdrawn articles on COVID-19. “No information” was a reason for retraction in the half (50%) of the articles, while other reasons for retraction included concern/issues about data, duplication, journal error, lack of approval from a third party, plagiarism, etc. Another cross-sectional study reported that more than half (59%, 27/46) of retracted COVID-19 articles remained available as original unmarked electronic documents (33% as full text and 26% as an abstract only) and that sources of articles after retraction were preprint servers, ResearchGate and, less commonly, websites including PubMed Central and the World Health Organization.12\nOne study showed that the top 100 highly cited COVID-19 articles in 2020 had citations ranging from 1,147 to 20,440. The median number of citations was 1,970 (IQR, 1,456–2,939).13 Most of the first authors were from China (58%), followed by the United States of America (16%), and the United Kingdom (7%). New England Journal of Medicine, Journal of the American Medical Association, and Lancet comprised 37% of these documents. Most of the top 100 highly cited COVID-19 documents were descriptive studies focusing on the epidemiology (48%) and clinical course (60%) of COVID-19.13\nBrandt et al.14 compared citation rates between COVID-19 and non-COVID-19 articles published over a period of 24 months (from January 1, 2020 to December 31, 2021) in 24 major scientific journals in eight selected fields. The results showed that a COVID-19:non-COVID-19 per-article citation ratio was 5.58. COVID-19 documents have showed more than 80% increase in citations relative to non-COVID-19 documents, when the influence of other variables were minimized with negative binomial regression.\nSocial media has the potential as a vehicle for disseminating scientific information during a public health crisis.15 According to Taneja et al.,15 during the first six months of the pandemic, from December 2019 through May 2020, strong correlations were identified between Twitter activity trends and preprint and publication activity (P < 0.001 for both). Sharing and spreading information on COVID-19 in a timely manner on social media was achieved at a much faster pace. According to a study involving an artificial neural network model, the Pearson correlation coefficient between the JIF of papers with COVID-19 as a topic and its Altmetric scores was 0.185 with P < 0.01.16 However, Altmetric score was not precise to describe the immediacy of citations of academic publication on COVID-19 research.16\nCOVID-19 continues to disrupt scientific and clinical studies and to shift research, publishing ethics, and the dynamics of the peer reviewing process to a new normal.1718 However, research quality and integrity should not be compromised despite the innovations in research methodology and situations related to the current health crisis.\nIn conclusion, COVID-19 pandemic significantly impacted the trends in biomedical publications and their citations." ]
[ "intro", "methods", "ethics-statement", "results", "discussion" ]
[ "COVID-19", "Journal", "Publication", "Impact Factor", "Citation", "Research" ]
The association of chronic liver disorders with exacerbation of symptoms and complications related to COVID-19: A systematic review and meta-analysis of cohort studies.
36254683
The aim of this review was to combine the results of published cohort studies to determine the exact association between chronic liver disorders, and the severe form of COVID-19, and its associated complications.
INTRODUCTION
This meta-analysis employed a keyword search (COVID-19 and chronic liver disorders) using PubMed (Medline), Scopus, Web of Sciences, and Embase (Elsevier). All articles related from January 2019 to May 2022 were reviewed. The STATA software was used for analysis.
METHODS
The risk of death in COVID-19 patients with chronic liver disorders was higher than in ones without the chronic liver disease (RR: 1.52; CI 95%: 1.46-1.57; I2 : 86.14%). Also, the risk of acute respiratory distress syndrome (ARDS) and hospitalization in COVID-19 patients with chronic liver disorders was higher than in ones without the chronic liver disease ([RR: 1.65; CI 95%: 1.09-2.50; I2 : 0.00%] and [RR: 1.39; CI 95%: 1.23-1.58; I2 : 0.20%]). Also, the meta-analysis showed cough, headache, myalgia, nausea, diarrhea, and fatigue were 1.37 (CI 95%: 1.20-1.55), 1.23 (CI 95%: 1.09-1.38), 1.25 (CI 95%: 1.04-1.50), 1.19 (CI 95%: 1.02-1.40), 1.89 (CI 95%: 1.30-2.75), 1.49 (CI 95%: 1.07-2.09), and 1.14 (CI 95%: 0.98-1.33), respectively, whereas the risk of all these symptoms was higher in COVID-19 patients with chronic liver diseases than ones without chronic liver disorders.
RESULTS
The mortality and complications due to COVID-19 were significantly different between patients with the chronic liver disease and the general population.
CONCLUSION
[ "Humans", "COVID-19", "SARS-CoV-2", "Liver Diseases", "Cohort Studies" ]
9716710
null
null
METHODS
The guideline of Preferred Reporting Items for Systematic Reviews and Meta‐Analyses (PRISMA) was used to review meta‐analyses, and systematic reviews. 27 Also, the study protocol was registered in PROSPER with the code CRD42022327806. [SUBTITLE] Search strategy and screening [SUBSECTION] The search was performed without language restrictions. The search strategy consisted of the following main keywords extracted from Mesh: “COVID 19,” “Liver Disease,” and “Chronic Liver Dysfunction.” Search databases included PubMed (Medline), Scopus, Web of Sciences, and Embase (Elsevier). The search deadline was from January 1, 2019, to January 1, 2022. Duplicate published articles were removed considering their titles, authors, and publication years using Endnote software version 9. Then, the remaining studies were evaluated by reviewing their titles, abstracts, and full texts, considering the inclusion criteria. In addition to searching the mentioned databases, gray literature was searched by reviewing articles in the first 10 pages of Google scholar, and manual search was performed by reviewing references of related studies. Two authors (MA, MA) independently screened articles based on their titles, abstracts, and full texts, and disputes were resolved by the third one (YM). After screening, the studies were finally selected by evaluating their full texts. The search was performed without language restrictions. The search strategy consisted of the following main keywords extracted from Mesh: “COVID 19,” “Liver Disease,” and “Chronic Liver Dysfunction.” Search databases included PubMed (Medline), Scopus, Web of Sciences, and Embase (Elsevier). The search deadline was from January 1, 2019, to January 1, 2022. Duplicate published articles were removed considering their titles, authors, and publication years using Endnote software version 9. Then, the remaining studies were evaluated by reviewing their titles, abstracts, and full texts, considering the inclusion criteria. In addition to searching the mentioned databases, gray literature was searched by reviewing articles in the first 10 pages of Google scholar, and manual search was performed by reviewing references of related studies. Two authors (MA, MA) independently screened articles based on their titles, abstracts, and full texts, and disputes were resolved by the third one (YM). After screening, the studies were finally selected by evaluating their full texts. [SUBTITLE] Inclusion and exclusion criteria [SUBSECTION] The inclusion criteria were defined based on the PECOT structure as follows: Population included the whole population, whether people with the CLD or healthy ones, exposure was considered as the presence of chronic liver disorders, comparison included comparing healthy people with those with chronic liver disorders, and outcomes included the COVID‐19 symptoms and its associated complications, such as hospitalization in the intensive care unit, death, and acute respiratory distress syndrome (ARDS). The intended studies to conduct meta‐analysis were cohort ones because this type of observational studies is much more important than others to examine the casual association. Systematic reviews, case reports, case series, case controls, cross sectionals, clinical trials, and other interventional studies as well as letters to the editor were excluded from the present research. Also, studies that met the inclusion criteria but their full texts were not available, first by sending an email to their authors, the full texts were requested and if the authors did not respond, they were removed. The inclusion criteria were defined based on the PECOT structure as follows: Population included the whole population, whether people with the CLD or healthy ones, exposure was considered as the presence of chronic liver disorders, comparison included comparing healthy people with those with chronic liver disorders, and outcomes included the COVID‐19 symptoms and its associated complications, such as hospitalization in the intensive care unit, death, and acute respiratory distress syndrome (ARDS). The intended studies to conduct meta‐analysis were cohort ones because this type of observational studies is much more important than others to examine the casual association. Systematic reviews, case reports, case series, case controls, cross sectionals, clinical trials, and other interventional studies as well as letters to the editor were excluded from the present research. Also, studies that met the inclusion criteria but their full texts were not available, first by sending an email to their authors, the full texts were requested and if the authors did not respond, they were removed. [SUBTITLE] Data extraction [SUBSECTION] After three stages of evaluation of titles, abstracts, and full texts, the selected articles were retrieved for detailed analysis. Data were collected using a checklist included authors' names, country, publication year, study type, study population, sample size, data source, age, number of patients with COVID‐19, hospitalization and mortality due to COVID‐19, ARDS, need for ventilation, ICU hospitalization, and evaluation of COVID‐19 symptoms (fever, cough, weakness, chest pain, abdominal pain, CT scan, Chest X‐ray, respiratory problems and shortness of breath, decreased sense of smell decreased sense of taste, fatigue, headache, dizziness, myalgia, diarrhea, nausea, and vomiting). After three stages of evaluation of titles, abstracts, and full texts, the selected articles were retrieved for detailed analysis. Data were collected using a checklist included authors' names, country, publication year, study type, study population, sample size, data source, age, number of patients with COVID‐19, hospitalization and mortality due to COVID‐19, ARDS, need for ventilation, ICU hospitalization, and evaluation of COVID‐19 symptoms (fever, cough, weakness, chest pain, abdominal pain, CT scan, Chest X‐ray, respiratory problems and shortness of breath, decreased sense of smell decreased sense of taste, fatigue, headache, dizziness, myalgia, diarrhea, nausea, and vomiting). [SUBTITLE] Risk of bias [SUBSECTION] Two of the authors (MA and PM) conducted a qualitative evaluation of the studies based on the Newcastle–Ottawa Quality Assessment Scale (NOS) checklist designed to evaluate the quality of observational studies. This tool examines each research with eight items in three groups, including how to select study samples, how to compare and analyze study groups, and how to measure and analyze the desired outcome. Each of these items is given a score of one if it is observed in the studies, and the maximum score for each study is 9 points. In case of discrepancies in the score assigned to the published articles, the discussion method and the third researcher were applied to reach an agreement. Two of the authors (MA and PM) conducted a qualitative evaluation of the studies based on the Newcastle–Ottawa Quality Assessment Scale (NOS) checklist designed to evaluate the quality of observational studies. This tool examines each research with eight items in three groups, including how to select study samples, how to compare and analyze study groups, and how to measure and analyze the desired outcome. Each of these items is given a score of one if it is observed in the studies, and the maximum score for each study is 9 points. In case of discrepancies in the score assigned to the published articles, the discussion method and the third researcher were applied to reach an agreement. [SUBTITLE] Statistical analysis [SUBSECTION] To calculate the association, cumulative risk ratio (RR) with the 95% confidence interval and the meta set command were used considering logarithm and logarithm standard deviation of the RR. Heterogeneity was assessed between studies using the I 2 and Q Cochrane tests. Egger test was used to evaluate the publication bias. Statistical analysis was performed using STATA 16.0 and P‐value < 0.05 was considered. To calculate the association, cumulative risk ratio (RR) with the 95% confidence interval and the meta set command were used considering logarithm and logarithm standard deviation of the RR. Heterogeneity was assessed between studies using the I 2 and Q Cochrane tests. Egger test was used to evaluate the publication bias. Statistical analysis was performed using STATA 16.0 and P‐value < 0.05 was considered.
RESULTS
First, 2251 studies were collected by searching based on the search strategy in the desired databases, of which 491 studies were duplicated, and 1760 ones remained. After reviewing the remained articles based on their titles, abstracts and full texts, 17 studies were selected for analysis. All articles were conducted in 2020 and 2021 and were cohorts (Figure 1 and Tables 1 and 2). The flowchart of search strategy and syntax Characteristics of included cohort studies (items related to COVID‐19 complications) Bahardoust, M. (2021) (Iran) Liver disease + Covid‐19 Covid‐19 Liver disease = 81 Non‐liver disease = 921 Liver disease = 81 Non‐liver disease = 921 Liver disease = 81 Non‐liver disease = 921 Liver disease = 76 non‐liver disease = 598 >7 day Liver disease = 10 non‐liver disease = 65 Covid‐19 alone Covid‐19 + Cirrhosis Cirrhosis alone COVID‐19 alone = 108 COV + Cirrhosis = 37 Cirrhosis alone = 127 COVID‐19 alone = 108 COVID‐19 + cirrhosis = 37 COVID‐19 alone = 108 COVID‐19 + cirrhosis = 37 Cirrhosis alone = 127 Covid‐19 = 15 Covid‐19 + Cirrhosis = 11 Cirrhosis = 24 Covid‐19 = 41 Covid‐19 + Cirrhosis = 16 Cirrhosis = 31 Covid‐19 = 41 COVID‐19 + Cirrhosis = 14 Cirrhosis = 18 Davidov‐Derevynko, Y. (2021) (Israel) COVID + Liver COVID + non‐Liver Covid‐19 = 323 Covid‐19 + Liver = 59 COVID‐19 = 323 COVID‐19 + Liver = 59 COVID‐19 = 323 COVID‐19 + Liver = 59 COVID‐19 = 22 COVID‐19 + Liver = 10 COVID‐19 = 36 COVID‐19 + Liver = 9 COVID‐19 = 35 COVID‐19 + Liver = 9 NAFLD + Covid‐19 Non‐NAFLD + Covid‐19 NAFLD + Covid‐19 = 61 Covid‐19 = 132 NAFLD +Covid‐19 = 61 Covid‐19 = 132 NAFLD +Covid‐19 = 61 Covid‐19 = 132 NAFLD + Covid‐19 = 18 Covid‐19 = 41 NAFLD + Covid‐19 = 11 Covid‐19 = 27 Covid‐19 id Covid‐19 + Liver Covid‐19 = 2895 Covid‐19 + Liver = 457 64.8 Covid‐19 = 2895 Covid‐19 + Liver = 457 Covid‐19 = 2895 Covid‐19 + Liver = 457 Covid‐19 = 769 Covid‐19 + Liver = 135 Covid‐19 = 522 Covid‐19 + Liver = 108 Covid‐19 id Covid‐19 + Liver Covid‐19 = 303 Covid‐19 + Liver = 14 70.5 Covid‐19 = 303 Covid‐19 + Liver = 14 Covid‐19 = 303 Covid‐19 + Liver = 14 Covid‐19 = 67 Covid‐19 + Liver = 4 Covid‐19 = 18 Covid‐19 + Liver = 1 Covid‐19 Covid‐19 + Liver Covid‐19 = 419 Covid‐19 + Liver = 28 40.8 Covid‐19 = 419 Covid‐19 + Liver = 28 Covid‐19 = 200 Covid‐19 + Liver = 26 Covid‐19 = 39 Covid‐19 + Liver = 8 Covid‐19 = 28 Covid‐19 + Liver = 3 Covid‐19 = 19 Covid‐19 + Liver = 2 Covid‐19 Covid‐19 + Liver Covid‐19 = 294 Covid‐19 + Liver = 69 Covid‐19 = 294 Covid‐19 + Liver = 69 Covid‐19 = 294 Covid‐19 + Liver = 69 Covid‐19 = 39 Covid‐19 + Liver = 16 Covid‐19 = 103 Covid‐19 + Liver = 34 Covid‐19 = 89 Covid‐19 + Liver = 33 Non‐NAFLD + Covid‐19 NAFLD + Covid‐19 Covid‐19 = 194 Covid‐19 + Liver = 86 Covid‐19 = 194 Covid‐19 + Liver = 86 Covid‐19 = 194 Covid‐19 + Liver = 86 Covid‐19 = 0 Covid‐19 + Liver = 0 Covid‐19 = 2 Covid‐19 + Liver = 2 Covid‐19 = 13 Covid‐19 + Liver = 5 Covid‐19 Covid‐19 + Liver Covid‐19 = 118 Covid‐19 + Liver = 22 Covid‐19 = 118 Covid‐19 + Liver = 22 Covid‐19 = 118 Covid‐19 + Liver = 22 Covid‐19 = 0 Covid‐19 + Liver = 1 Covid‐19 Covid‐19 + Liver COVID‐19 + Liver = 47 COVID‐19 = 958 COVID‐19 + Liver = 47 COVID‐19 = 958 COVID‐19 + Liver = 47 COVID‐19 = 958 COVID‐19 + Liver = 7 COVID‐19 = 70 COVID‐19 + Liver = 8 COVID‐19 = 105 COVID‐19 + Liver = 8 COVID‐19 = 89 Invasive mechanical ventilation COVID‐19 + Liver = 4 COVID‐19 = 66 Invasive mechanical ventilation and ECMO COVID‐19 + Liver = 1 COVID‐19 = 17 Covid‐19 Covid‐19 + Liver COVID‐19 + Liver = 52 Covid‐19 = 52 COVID‐19 + Liver = 52 COVID‐19 = 52 COVID‐19 + Liver = 52 COVID‐19 = 52 COVID‐19 + Liver = 9 COVID‐19 = 0 Non‐invasive ventilation COVID‐19 = 2 COVID‐19 + Liver = 5 Invasive mechanical ventilation COVID‐19 = 1 COVID‐19 + Liver = 5 NO ventilation COVID‐19 = 49 COVID‐19 + Liver = 42 COVID‐19 COVID‐19 + Liver COVID‐19 = 105 COVID‐19 + Liver = 34 COVID‐19 = 105 COVID‐19 + Liver = 34 COVID‐19 = 105 COVID‐19 + Liver = 34 COVID‐19 = 29 COVID‐19 + Liver = 9 COVID‐19 = 41 COVID‐19 + Liver = 19 COVID‐19 = 25 COVID‐19 + Liver = 14 COVID‐19 = 11 COVID‐19 + Liver = 7 COVID‐19 COVID‐19 + Liver COVID‐19 = 243 634 COVID‐19 + Liver = 15 476 COVID‐19 = 243 634 COVID‐19 + Liver = 15 476 COVID‐19 = 243 634 COVID‐19 + Liver = 15 476 COVID‐19 = 35 262 COVID‐19 + Liver = 2941 COVID‐19 = 16 449 COVID‐19 + Liver = 1600 1. Noncirrhosis/negative 2. Non‐cirrhosis/positive; 3. Cirrhosis/negative; 4. Cirrhosis/positive 1. Healthy = 128 864 2. COVID‐19 = 29 446 3. Cirrhosis = 53 476 4. Cirrhosis + Covid‐19 = 8941 2. COVID‐19 = 29 446 4. Cirrhosis + Covid‐19 = 8941 Covid‐19 + Liver Covid‐19 All = 2780 Covid‐19 + Liver = 250 Covid‐19 = 2530 All = 2780 Covid‐19 + Liver = 250 Covid‐19 = 2530 Covid‐19 + Liver = 130 Covid‐19 = 760 Covid‐19 + Liver = 30 Covid‐19 = 110 NAFLD + Covid‐19 Non‐NAFLD + Covid‐19 NAFLD = 553 Non‐NAFLD = 2736 NAFLD = 553 Non‐NAFLD = 2736 NAFLD = 553 Non‐NAFLD = 2736 NAFLD = 60 Non‐NAFLD = 239 NAFLD = 196 Non‐NAFLD = 726 NAFLD = 76 Non‐NAFLD = 221 Characteristics of included cohort studies (items related to signs and symptoms of COVID‐19) Liver disease ≥38.5 = 35 <38.5 = 46 Non‐liver disease ≥38.5 = 175 <38.5 = 746 Liver disease = 47 Non‐liver disease = 552 Liver disease = 20 Non‐liver disease = 221 Liver disease = 20 Non‐liver disease = 313 Liver disease = 26 Non‐liver disease = 275 Liver disease = 49 Non‐liver disease = 485 Liver disease = 39 Non‐liver disease = 230 Liver disease = 37 Non‐liver disease = 276 Liver disease = 13 Non‐liver disease = 166 Covid‐19 = 75 Covid‐19 + Cirrhosis = 20 Covid‐19 = 70 Covid‐19 + Cirrhosis = 26 Covid‐19 = 19 Covid‐19 + Cirrhosis = 3 Covid‐19 = 8 Covid‐19 + Cirrhosis = 4 Covid‐19 = 15 Covid‐19 + Cirrhosis = 2 Covid‐19 = 31 Covid‐19 + Cirrhosis = 4 Covid‐19 = 29 Covid‐19 + Cirrhosis = 3 Covid‐19 = 29 Covid‐19 + Cirrhosis = 4 Covid‐19 = 74 Covid‐19 + Cirrhosis = 23 Covid‐19 = 17 Covid‐19 + Cirrhosis = 9 Covid‐19 = 44 Covid‐19 + Cirrhosis = 12 Covid‐19 = 174 Covid‐19 + Liver = 8 Covid‐19 = 185 Covid‐19 + Liver = 10 Covid‐19 = 46 Covid‐19 + Liver = 0 Covid‐19 = 69 Covid‐19 + Liver = 2 Covid‐19 = 38 Covid‐19 + Liver = 2 Covid‐19 = 23 Covid‐19 + Liver = 1 Covid‐19 = 17 Covid‐19 + Liver = 2 Covid‐19 = 82 Covid‐19 + Liver = 3 Covid‐19 = 108 Covid‐19 + Liver = 6 Covid‐19 = 132 Covid‐19 + Liver = 55 Covid‐19 = 109 Covid‐19 + Liver = 47 Covid‐19 = 16 Covid‐19 + Liver = 3 Covid‐19 = 21 Covid‐19 + Liver = 7 Covid‐19 = 16 Covid‐19 + Liver = 7 Covid‐19 = 42 Covid‐19 + Liver = 16 COVID‐19 + Liver = 24 COVID‐19 = 425 COVID‐19 + Liver = 23 COVID‐19 = 544 COVID‐19 + Liver = 8 COVID‐19 = 245 COVID‐19 + Liver = 9 COVID‐19 = 316 COVID‐19 + Liver = 15 COVID‐19 = 232 NAFLD = 319 Non‐NAFLD = 1283 NAFLD = 318 Non‐NAFLD = 1273 NAFLD = 23 Non‐NAFLD = 72 NAFLD = 74 Non‐NAFLD = 240 NAFLD = 101 Non‐NAFLD = 363 NAFLD = 23 Non‐NAFLD = 72 NAFLD = 316 Non‐NAFLD = 1519 NAFLD = 144 Non‐NAFLD = 588 The risk of death in COVID‐19 patients with chronic liver disorders was compared with that in ones without the CLD in 15 studies. 19 , 26 , 28 , 29 , 30 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , 38 , 39 , 40 , 42 In all of these studies, the mortality rate in patients with COVID‐19, and the CLD was higher than that in patients with COVID‐19, and without the CLD. 19 , 26 , 28 , 30 , 32 , 33 , 34 , 36 , 37 , 38 , 39 , 40 The cumulative RR after combining these studies was 1.52 (CI 95%: 1.46–1.57; I 2: 86.14%) (Figure 2). The pooled effect (risk ratio, RR) of chronic liver disorders on the mortality and admission to ICU related to COVID‐19 The risk of ARDS in patients with COVID‐19 who had chronic liver disorders was compared in three studies with those without chronic liver disorders. 35 , 37 , 39 The cumulative RR after combining these studies was 1.65 (CI 95%: 1.09–2.50; I 2: 0.00%) (Table 3). Results of meta‐analysis and reports pooled odds ratio based on symptoms and complications related to COVID‐19 Note: A: COVID‐19 individual with CLD; B: Healthy individual with CLD; C: COVID‐19 individual without CLD; D: Healthy individual without CLD. The risk of hospitalization of COVID‐19 patients who had chronic liver disorders in the intensive care unit was compared with that of ones without the CLD in nine studies, 26 , 29 , 30 , 31 , 34 , 35 , 37 , 39 , 42 , 43 in all of which the risk of hospitalization in the ICU was higher in the group of COVID‐19 patients with the CLD. 26 , 29 , 30 , 31 , 34 , 35 , 37 , 39 The cumulative RR after combining these studies was 1.39 (CI 95%: 1.23–1.58) with heterogeneity (I 2) of 0.2% (Figure 2). The risk of need for ventilation in COVID‐19 patients who had the CLD was evaluated in 11 studies compared with those who did not have chronic liver disorders. 26 , 29 , 30 , 32 , 33 , 34 , 37 , 38 , 39 , 40 , 42 In all of these studies, the need for ventilation was higher in the group with the CLD 26 , 29 , 30 , 32 , 33 , 34 , 37 , 38 , 39 , 40 The frequency of need for ventilation was higher in the group of patients with COVID‐19 and liver disorders. 42 The cumulative RR after combining these studies was 1.53 (CI 95%: 1.46–1.60) with heterogeneity (I 2) equal to 0.2% (Figure 3). The pooled effect (risk ratio, RR) of chronic liver disorders on the ventilation needs, fever, and cough related to COVID‐19 To determine the association between the COVID‐19 symptoms and chronic liver disorders, patients with COVID‐19 who had chronic liver disorders were compared with those who did not have the CLD, and the results of the meta‐analysis showed cough, headache, myalgia, nausea, diarrhea and fatigue were 1.37 (CI 95%: 1.20–1.55), 1.23 (CI 95%: 1.09–1.38), 1.25 (CI 95%: 1.04–1.50), 1.19 (CI 95%: 1.02–1.40), 1.89 (CI 95%: 1.30–2.75), 1.49 (CI 95%: 1.07–2.09), and 1.14 (CI 95%: 0.98–1.33), respectively, while the risk of all these symptoms was higher in COVID‐19 patients with underlying liver diseases than ones without chronic liver disorders. Other symptoms have been reported in the table (Figure 3 and Table 3). To examine the publication bias, the Eggers test was performed, which was not significant in all variables except myalgia and headache. That is, the publication bias has not occurred. [SUBTITLE] Subgroups analysis [SUBSECTION] The results of subgroups analysis according to age and continents for symptoms and complications related to COVID‐19 were reported in Table 4. The results show that complications related to COVID‐19 are more significant and severe in patients with CLD with age <60 and live in Asia (Table 4). The subgroups analysis of signs, symptoms, and complications related to COVID 19 in CLD patients based on continents and age The results of subgroups analysis according to age and continents for symptoms and complications related to COVID‐19 were reported in Table 4. The results show that complications related to COVID‐19 are more significant and severe in patients with CLD with age <60 and live in Asia (Table 4). The subgroups analysis of signs, symptoms, and complications related to COVID 19 in CLD patients based on continents and age [SUBTITLE] Discussion [SUBSECTION] The RR of complications and consequences of COVID‐19 in patients with the CLD during the pandemic period was examined in this study and the results showed the need for ventilation and ARDS had higher risks than other outcomes related to COVID‐19 in patients with chronic liver disorders. Exacerbation of alveolar epithelial damages, increased vascular permeability, and systemic inflammation are among the factors that predispose to increased ARDS in patients with the CLD. 44 , 45 Also, because these patients had a higher BMI and obesity was more prevalent in them, their need for ventilation was higher when they were infected with COVID‐19. 46 Due to the direct and significant effect of SARS‐CoV‐2 virus on ACE‐2 receptors in liver cells, patients with chronic liver disorders will have a more severe form of the disease if they develop COVID‐19, which causes their hospitalization in the intensive care unit of hospitals due to COVID‐19. According to the results of previous studies, patients with chronic liver disorders have a higher risk of being admitted to the intensive care unit than others in the community if they develop COVID‐19. In the present meta‐analysis, the same result was obtained, which was in line with the results of preliminary studies published in the world. 47 However, the results of some studies in the world have not shown this association, for example, a study by Huang, R. et al. showed the risk of hospitalization in intensive care unit due to COVID‐19 in patients with chronic liver disorders was not significantly different from other people in the community. 35 The reason for this inconsistency can be attributed to differences in the study method, how to collect information, and the type of patients studied in these articles. Also, the risk of death due to COVID‐19 in the present meta‐analysis was significantly higher in patients with chronic liver disorders than other individuals. The reason for this can be attributed to the increase in inflammatory cytokines such as IL‐6, Ferritin and TNF‐alpha. 48 In patients with COVID‐19, inflammatory cytokines increase, eventually leading to a cytokine storm in more severe forms of the disease. On the other hand, for patients with severe forms of COVID‐19, drugs that aggravate cytokines and cause cytokine storms are prescribed, which may also exacerbate death in COVID‐19 patients with chronic liver disorders. Coincidence of chronic liver disorders and the use of anti‐COVID‐19 drugs significantly increases the risk of mortality in COVID‐19 patients with the CLD. Also, worsening liver function reduces the number and disrupt the function of neutrophils, monocytes and innate immune proteins, and ultimately the number of both B and T lymphocytes involved in acquired immunity decreases and eventually immune dysfunction increases. 49 , 50 In this study, in addition to the mentioned cases, the symptoms associated with COVID‐19 including fever, cough, headache, myalgia, nausea, diarrhea, and fatigue were also evaluated in patients with the CLD. The results of this meta‐analysis showed among these symptoms, nausea, diarrhea and abdominal pain were more common than other ones in patients with chronic liver disorders. Increased gastrointestinal symptoms may be due to liver dysfunction. In addition, other symptoms such as fever, cough, headache, myalgia, and fatigue were more common in patients with the underlying liver disease than in healthy individuals while cytokines were effective in worsening these symptoms. 48 The severity of various symptoms in patients with chronic liver disorders can affect outcomes of COVID‐19 and, as a result, increase the outcome and mortality in this group of patients. Some studies on the effect of COVID‐19 on the incidence of liver disorders published results, which suggested in the case of COVID‐19 due to the widespread distribution of the main virus receptor called the angiotensin‐converting enzyme 2 (ACE2), the virus could cause a widespread disease with more involvement of extra‐pulmonary organs, especially the liver. ACE2 receptors are also expressed in the gastrointestinal tract, vascular endothelium, and hepatic cholangiocytes. 51 , 52 Various studies showed elevated liver enzymes indicated liver damages and were common in COVID‐19 patients with chronic and non‐CLDs. 52 On the other hand, initial clinical studies in this area also confirmed a significant increase in liver enzymes such as ALT, and AST due to SARS‐CoV‐2 infection. 53 , 54 , 55 , 56 The results of a meta‐analysis also showed the levels of ALP, and γ‐GT enzymes were significant as a result of cholangiocellular damages. 57 , 58 , 59 As the study results show, it is not yet clear how much an increase in liver enzyme levels can exacerbate the complications of COVID‐19 or the disease progression. In patients with COVID‐19 who have not had a chronic liver disorder or liver damage before infection, a slight liver disorder is found after recovery. 60 , 61 Abnormal results of liver tests were associated with more severe forms of COVID‐19 and mortality. Although RNAs of SARS‐CoV‐2 have been detected in the liver of patients with COVID‐19, it is not yet exactly clear how much SARS‐CoV‐2 infects the liver and multiplies in its cells. 17 , 51 The range of liver damages in the COVID‐19 disease may be direct infection by SARS‐CoV‐2, indirect involvement by systemic inflammation, and hypoxic changes. However, due to the major role of the liver in endobiotic and xenobiotic drug metabolism, coagulation, albumin, and production of acute phase reactants, liver dysfunction may affect the pathophysiology of the COVID‐19 disease. 62 , 63 The results of the present meta‐analysis along with other published results can be useful in finding many of these answers. They are also useful in updating treatment and prevention guidelines. This study was the first meta‐analysis to compare the complications and consequences associated with COVID‐19 in two groups of patients with the CLD and healthy ones. On the other hand, the results of subgroup analyses and overall results had a high homogeneity, which indicated the homogeneous and correct selection of initial studies in order to perform this meta‐analysis. The heterogeneity in this meta‐analysis only was higher at two outcomes (Fever and Mortality). For detecting sources of this heterogeneity, all primary cohort studies were reviewed, and extracted related variables that reported in selected cohort studies completely. Between reported variables, age and continents were extracted. Other variables like type of underlying diseases, such as diabetes, coronary heart disease (CHD), and cancers, type of study population, type of measures tools for outcomes measure were not reported in selected cohort studies. The results of subgroup analysis based on age and continents show that the heterogeneity was decreased in many of categories, but in fever and mortality not decreased. This heterogeneity rate confirms the difference between the combined studies. This difference may be due to differences in some section of studies such as the methods of measuring the outcomes and the tools used, the methods of sampling, presence of important underlying diseases (comorbidities) like diabetes, CHD, cancers, and chronic obstructive pulmonary disease (COPD). These variables and factors not reported in selected cohort studies in this meta‐analysis, so authors could not subgroup analysis based on its. The results of this research are also based on cohort studies, which are one of the most important observational studies in order to find a causal association, but the overall results of this study have not yet determined the exact association between SARS‐CoV‐2 infection and chronic liver disorders. In other words, according to the results of published studies, it can be claimed that there is a kind of causal association between these two factors, and in order to find it, studies based on genetics or molecular science such as Mendelian randomization are needed. The RR of complications and consequences of COVID‐19 in patients with the CLD during the pandemic period was examined in this study and the results showed the need for ventilation and ARDS had higher risks than other outcomes related to COVID‐19 in patients with chronic liver disorders. Exacerbation of alveolar epithelial damages, increased vascular permeability, and systemic inflammation are among the factors that predispose to increased ARDS in patients with the CLD. 44 , 45 Also, because these patients had a higher BMI and obesity was more prevalent in them, their need for ventilation was higher when they were infected with COVID‐19. 46 Due to the direct and significant effect of SARS‐CoV‐2 virus on ACE‐2 receptors in liver cells, patients with chronic liver disorders will have a more severe form of the disease if they develop COVID‐19, which causes their hospitalization in the intensive care unit of hospitals due to COVID‐19. According to the results of previous studies, patients with chronic liver disorders have a higher risk of being admitted to the intensive care unit than others in the community if they develop COVID‐19. In the present meta‐analysis, the same result was obtained, which was in line with the results of preliminary studies published in the world. 47 However, the results of some studies in the world have not shown this association, for example, a study by Huang, R. et al. showed the risk of hospitalization in intensive care unit due to COVID‐19 in patients with chronic liver disorders was not significantly different from other people in the community. 35 The reason for this inconsistency can be attributed to differences in the study method, how to collect information, and the type of patients studied in these articles. Also, the risk of death due to COVID‐19 in the present meta‐analysis was significantly higher in patients with chronic liver disorders than other individuals. The reason for this can be attributed to the increase in inflammatory cytokines such as IL‐6, Ferritin and TNF‐alpha. 48 In patients with COVID‐19, inflammatory cytokines increase, eventually leading to a cytokine storm in more severe forms of the disease. On the other hand, for patients with severe forms of COVID‐19, drugs that aggravate cytokines and cause cytokine storms are prescribed, which may also exacerbate death in COVID‐19 patients with chronic liver disorders. Coincidence of chronic liver disorders and the use of anti‐COVID‐19 drugs significantly increases the risk of mortality in COVID‐19 patients with the CLD. Also, worsening liver function reduces the number and disrupt the function of neutrophils, monocytes and innate immune proteins, and ultimately the number of both B and T lymphocytes involved in acquired immunity decreases and eventually immune dysfunction increases. 49 , 50 In this study, in addition to the mentioned cases, the symptoms associated with COVID‐19 including fever, cough, headache, myalgia, nausea, diarrhea, and fatigue were also evaluated in patients with the CLD. The results of this meta‐analysis showed among these symptoms, nausea, diarrhea and abdominal pain were more common than other ones in patients with chronic liver disorders. Increased gastrointestinal symptoms may be due to liver dysfunction. In addition, other symptoms such as fever, cough, headache, myalgia, and fatigue were more common in patients with the underlying liver disease than in healthy individuals while cytokines were effective in worsening these symptoms. 48 The severity of various symptoms in patients with chronic liver disorders can affect outcomes of COVID‐19 and, as a result, increase the outcome and mortality in this group of patients. Some studies on the effect of COVID‐19 on the incidence of liver disorders published results, which suggested in the case of COVID‐19 due to the widespread distribution of the main virus receptor called the angiotensin‐converting enzyme 2 (ACE2), the virus could cause a widespread disease with more involvement of extra‐pulmonary organs, especially the liver. ACE2 receptors are also expressed in the gastrointestinal tract, vascular endothelium, and hepatic cholangiocytes. 51 , 52 Various studies showed elevated liver enzymes indicated liver damages and were common in COVID‐19 patients with chronic and non‐CLDs. 52 On the other hand, initial clinical studies in this area also confirmed a significant increase in liver enzymes such as ALT, and AST due to SARS‐CoV‐2 infection. 53 , 54 , 55 , 56 The results of a meta‐analysis also showed the levels of ALP, and γ‐GT enzymes were significant as a result of cholangiocellular damages. 57 , 58 , 59 As the study results show, it is not yet clear how much an increase in liver enzyme levels can exacerbate the complications of COVID‐19 or the disease progression. In patients with COVID‐19 who have not had a chronic liver disorder or liver damage before infection, a slight liver disorder is found after recovery. 60 , 61 Abnormal results of liver tests were associated with more severe forms of COVID‐19 and mortality. Although RNAs of SARS‐CoV‐2 have been detected in the liver of patients with COVID‐19, it is not yet exactly clear how much SARS‐CoV‐2 infects the liver and multiplies in its cells. 17 , 51 The range of liver damages in the COVID‐19 disease may be direct infection by SARS‐CoV‐2, indirect involvement by systemic inflammation, and hypoxic changes. However, due to the major role of the liver in endobiotic and xenobiotic drug metabolism, coagulation, albumin, and production of acute phase reactants, liver dysfunction may affect the pathophysiology of the COVID‐19 disease. 62 , 63 The results of the present meta‐analysis along with other published results can be useful in finding many of these answers. They are also useful in updating treatment and prevention guidelines. This study was the first meta‐analysis to compare the complications and consequences associated with COVID‐19 in two groups of patients with the CLD and healthy ones. On the other hand, the results of subgroup analyses and overall results had a high homogeneity, which indicated the homogeneous and correct selection of initial studies in order to perform this meta‐analysis. The heterogeneity in this meta‐analysis only was higher at two outcomes (Fever and Mortality). For detecting sources of this heterogeneity, all primary cohort studies were reviewed, and extracted related variables that reported in selected cohort studies completely. Between reported variables, age and continents were extracted. Other variables like type of underlying diseases, such as diabetes, coronary heart disease (CHD), and cancers, type of study population, type of measures tools for outcomes measure were not reported in selected cohort studies. The results of subgroup analysis based on age and continents show that the heterogeneity was decreased in many of categories, but in fever and mortality not decreased. This heterogeneity rate confirms the difference between the combined studies. This difference may be due to differences in some section of studies such as the methods of measuring the outcomes and the tools used, the methods of sampling, presence of important underlying diseases (comorbidities) like diabetes, CHD, cancers, and chronic obstructive pulmonary disease (COPD). These variables and factors not reported in selected cohort studies in this meta‐analysis, so authors could not subgroup analysis based on its. The results of this research are also based on cohort studies, which are one of the most important observational studies in order to find a causal association, but the overall results of this study have not yet determined the exact association between SARS‐CoV‐2 infection and chronic liver disorders. In other words, according to the results of published studies, it can be claimed that there is a kind of causal association between these two factors, and in order to find it, studies based on genetics or molecular science such as Mendelian randomization are needed.
CONCLUSION
The results of this study showed the mortality and consequences due to COVID‐19 were significantly different between patients with the CLD and the general population. Also, the COVID‐19 symptoms in people with liver disorders were significantly more severe than those in healthy people. So, taking measures is necessary to manage them. It is recommended to reduce the risk of mortality and other consequences of COVID‐19 through screening and treating people with liver disorders in the lower stages of the CLD.
[ "INTRODUCTION", "Search strategy and screening", "Inclusion and exclusion criteria", "Data extraction", "Risk of bias", "Statistical analysis", "Subgroups analysis", "Discussion", "AUTHOR CONTRIBUTIONS" ]
[ "The chronic liver disease (CLD) is a progressive deterioration of liver function over 6 months, which slowly progresses, leaving the liver unable to synthesize coagulation factors, and proteins, detoxify harmful metabolic products, and excrete bile.\n1\n, \n2\n It is a continuous process of inflammation, destruction, and regeneration of the liver parenchyma, which leads to fibrosis and cirrhosis.\n1\n, \n3\n CLD is caused by a wide range of causes, including toxins, long‐term alcohol abuse, infection, autoimmune diseases, genetic disorders, and metabolic disorders.\n1\n, \n4\n Cirrhosis is the final stage of CLD, which leads to liver dysfunction, extensive nodule formation, vascular reorganization, neo‐angiogenesis, and extracellular matrix deposition.\n1\n, \n5\n, \n6\n The underlying mechanism of fibrosis and cirrhosis at the cellular level is the uptake of stellate cells and fibroblasts, which leads to fibrosis while parenchymal regeneration relies on liver stem cells.\n1\n, \n7\n The CLD is a very common clinical condition and the focus is more on its common causes, clinical manifestations and management.\n1\n, \n8\n, \n9\n, \n10\n, \n11\n About 1.5 billion people worldwide have CLD, which causes more than 2 million deaths a year.\n9\n, \n12\n With the rapid spread of COVID‐19, there are considerable concerns that patients with CLD are a vulnerable population and at higher risks for the more severe form of COVID‐19 and its associated complications.\n13\n Early on, patients with underlying diseases such as diabetes, chronic lung diseases, cardiovascular diseases, hypertension, and cancer were labeled as those at high risks for severe COVID‐19.\n14\n Although the virus mainly affects the lungs, experiences of China and the United States suggest SARS‐CoV‐2 may affect extra‐pulmonary systems, including the gastrointestinal and hepato‐biliary systems.\n15\n, \n16\n\n\nHowever, it was not initially clear whether patients with the CLD were more susceptible to COVID‐19. Data from some recent studies showed the CLD in the absence of immunosuppressive therapy was not associated with an increased risk of COVID‐19,\n17\n whereas results from other studies showed the CLD, including patients with cirrhosis and non‐cirrhosis, was associated with higher rates of mortality because of COVID‐19.\n18\n, \n19\n, \n20\n Also, various reports from different countries showed more than half of the hospitalized adults due to COVID‐19 had abnormal aminotransferase levels and 2% to 11% of them had underlying liver diseases.\n17\n, \n21\n, \n22\n, \n23\n, \n24\n, \n25\n However, there are limited and conflicting reports on the liver disease nature among COVID‐19 patients, and it is unclear how underlying CLD affects liver injuries and clinical outcomes in these patients.\n26\n Due to the discrepancies in the results of previous studies and the importance of determining the association between various underlying diseases, especially liver disorders, and COVID‐19 and its severe form, the authors in this meta‐analysis decided to combine the results of published cohort studies to determine the exact association between chronic liver disorders, and the severe form of COVID‐19, and its associated complications.", "The search was performed without language restrictions. The search strategy consisted of the following main keywords extracted from Mesh:\n“COVID 19,” “Liver Disease,” and “Chronic Liver Dysfunction.”\nSearch databases included PubMed (Medline), Scopus, Web of Sciences, and Embase (Elsevier).\nThe search deadline was from January 1, 2019, to January 1, 2022. Duplicate published articles were removed considering their titles, authors, and publication years using Endnote software version 9. Then, the remaining studies were evaluated by reviewing their titles, abstracts, and full texts, considering the inclusion criteria. In addition to searching the mentioned databases, gray literature was searched by reviewing articles in the first 10 pages of Google scholar, and manual search was performed by reviewing references of related studies. Two authors (MA, MA) independently screened articles based on their titles, abstracts, and full texts, and disputes were resolved by the third one (YM). After screening, the studies were finally selected by evaluating their full texts.", "The inclusion criteria were defined based on the PECOT structure as follows:\nPopulation included the whole population, whether people with the CLD or healthy ones, exposure was considered as the presence of chronic liver disorders, comparison included comparing healthy people with those with chronic liver disorders, and outcomes included the COVID‐19 symptoms and its associated complications, such as hospitalization in the intensive care unit, death, and acute respiratory distress syndrome (ARDS). The intended studies to conduct meta‐analysis were cohort ones because this type of observational studies is much more important than others to examine the casual association.\nSystematic reviews, case reports, case series, case controls, cross sectionals, clinical trials, and other interventional studies as well as letters to the editor were excluded from the present research. Also, studies that met the inclusion criteria but their full texts were not available, first by sending an email to their authors, the full texts were requested and if the authors did not respond, they were removed.", "After three stages of evaluation of titles, abstracts, and full texts, the selected articles were retrieved for detailed analysis. Data were collected using a checklist included authors' names, country, publication year, study type, study population, sample size, data source, age, number of patients with COVID‐19, hospitalization and mortality due to COVID‐19, ARDS, need for ventilation, ICU hospitalization, and evaluation of COVID‐19 symptoms (fever, cough, weakness, chest pain, abdominal pain, CT scan, Chest X‐ray, respiratory problems and shortness of breath, decreased sense of smell decreased sense of taste, fatigue, headache, dizziness, myalgia, diarrhea, nausea, and vomiting).", "Two of the authors (MA and PM) conducted a qualitative evaluation of the studies based on the Newcastle–Ottawa Quality Assessment Scale (NOS) checklist designed to evaluate the quality of observational studies. This tool examines each research with eight items in three groups, including how to select study samples, how to compare and analyze study groups, and how to measure and analyze the desired outcome. Each of these items is given a score of one if it is observed in the studies, and the maximum score for each study is 9 points. In case of discrepancies in the score assigned to the published articles, the discussion method and the third researcher were applied to reach an agreement.", "To calculate the association, cumulative risk ratio (RR) with the 95% confidence interval and the meta set command were used considering logarithm and logarithm standard deviation of the RR. Heterogeneity was assessed between studies using the I\n2 and Q Cochrane tests. Egger test was used to evaluate the publication bias. Statistical analysis was performed using STATA 16.0 and P‐value < 0.05 was considered.", "The results of subgroups analysis according to age and continents for symptoms and complications related to COVID‐19 were reported in Table 4. The results show that complications related to COVID‐19 are more significant and severe in patients with CLD with age <60 and live in Asia (Table 4).\nThe subgroups analysis of signs, symptoms, and complications related to COVID 19 in CLD patients based on continents and age", "The RR of complications and consequences of COVID‐19 in patients with the CLD during the pandemic period was examined in this study and the results showed the need for ventilation and ARDS had higher risks than other outcomes related to COVID‐19 in patients with chronic liver disorders. Exacerbation of alveolar epithelial damages, increased vascular permeability, and systemic inflammation are among the factors that predispose to increased ARDS in patients with the CLD.\n44\n, \n45\n Also, because these patients had a higher BMI and obesity was more prevalent in them, their need for ventilation was higher when they were infected with COVID‐19.\n46\n\n\nDue to the direct and significant effect of SARS‐CoV‐2 virus on ACE‐2 receptors in liver cells, patients with chronic liver disorders will have a more severe form of the disease if they develop COVID‐19, which causes their hospitalization in the intensive care unit of hospitals due to COVID‐19. According to the results of previous studies, patients with chronic liver disorders have a higher risk of being admitted to the intensive care unit than others in the community if they develop COVID‐19. In the present meta‐analysis, the same result was obtained, which was in line with the results of preliminary studies published in the world.\n47\n However, the results of some studies in the world have not shown this association, for example, a study by Huang, R. et al. showed the risk of hospitalization in intensive care unit due to COVID‐19 in patients with chronic liver disorders was not significantly different from other people in the community.\n35\n The reason for this inconsistency can be attributed to differences in the study method, how to collect information, and the type of patients studied in these articles.\nAlso, the risk of death due to COVID‐19 in the present meta‐analysis was significantly higher in patients with chronic liver disorders than other individuals. The reason for this can be attributed to the increase in inflammatory cytokines such as IL‐6, Ferritin and TNF‐alpha.\n48\n In patients with COVID‐19, inflammatory cytokines increase, eventually leading to a cytokine storm in more severe forms of the disease. On the other hand, for patients with severe forms of COVID‐19, drugs that aggravate cytokines and cause cytokine storms are prescribed, which may also exacerbate death in COVID‐19 patients with chronic liver disorders. Coincidence of chronic liver disorders and the use of anti‐COVID‐19 drugs significantly increases the risk of mortality in COVID‐19 patients with the CLD. Also, worsening liver function reduces the number and disrupt the function of neutrophils, monocytes and innate immune proteins, and ultimately the number of both B and T lymphocytes involved in acquired immunity decreases and eventually immune dysfunction increases.\n49\n, \n50\n\n\nIn this study, in addition to the mentioned cases, the symptoms associated with COVID‐19 including fever, cough, headache, myalgia, nausea, diarrhea, and fatigue were also evaluated in patients with the CLD. The results of this meta‐analysis showed among these symptoms, nausea, diarrhea and abdominal pain were more common than other ones in patients with chronic liver disorders. Increased gastrointestinal symptoms may be due to liver dysfunction. In addition, other symptoms such as fever, cough, headache, myalgia, and fatigue were more common in patients with the underlying liver disease than in healthy individuals while cytokines were effective in worsening these symptoms.\n48\n The severity of various symptoms in patients with chronic liver disorders can affect outcomes of COVID‐19 and, as a result, increase the outcome and mortality in this group of patients.\nSome studies on the effect of COVID‐19 on the incidence of liver disorders published results, which suggested in the case of COVID‐19 due to the widespread distribution of the main virus receptor called the angiotensin‐converting enzyme 2 (ACE2), the virus could cause a widespread disease with more involvement of extra‐pulmonary organs, especially the liver. ACE2 receptors are also expressed in the gastrointestinal tract, vascular endothelium, and hepatic cholangiocytes.\n51\n, \n52\n Various studies showed elevated liver enzymes indicated liver damages and were common in COVID‐19 patients with chronic and non‐CLDs.\n52\n On the other hand, initial clinical studies in this area also confirmed a significant increase in liver enzymes such as ALT, and AST due to SARS‐CoV‐2 infection.\n53\n, \n54\n, \n55\n, \n56\n The results of a meta‐analysis also showed the levels of ALP, and γ‐GT enzymes were significant as a result of cholangiocellular damages.\n57\n, \n58\n, \n59\n As the study results show, it is not yet clear how much an increase in liver enzyme levels can exacerbate the complications of COVID‐19 or the disease progression. In patients with COVID‐19 who have not had a chronic liver disorder or liver damage before infection, a slight liver disorder is found after recovery.\n60\n, \n61\n\n\nAbnormal results of liver tests were associated with more severe forms of COVID‐19 and mortality. Although RNAs of SARS‐CoV‐2 have been detected in the liver of patients with COVID‐19, it is not yet exactly clear how much SARS‐CoV‐2 infects the liver and multiplies in its cells.\n17\n, \n51\n The range of liver damages in the COVID‐19 disease may be direct infection by SARS‐CoV‐2, indirect involvement by systemic inflammation, and hypoxic changes. However, due to the major role of the liver in endobiotic and xenobiotic drug metabolism, coagulation, albumin, and production of acute phase reactants, liver dysfunction may affect the pathophysiology of the COVID‐19 disease.\n62\n, \n63\n The results of the present meta‐analysis along with other published results can be useful in finding many of these answers. They are also useful in updating treatment and prevention guidelines. This study was the first meta‐analysis to compare the complications and consequences associated with COVID‐19 in two groups of patients with the CLD and healthy ones. On the other hand, the results of subgroup analyses and overall results had a high homogeneity, which indicated the homogeneous and correct selection of initial studies in order to perform this meta‐analysis.\nThe heterogeneity in this meta‐analysis only was higher at two outcomes (Fever and Mortality). For detecting sources of this heterogeneity, all primary cohort studies were reviewed, and extracted related variables that reported in selected cohort studies completely. Between reported variables, age and continents were extracted. Other variables like type of underlying diseases, such as diabetes, coronary heart disease (CHD), and cancers, type of study population, type of measures tools for outcomes measure were not reported in selected cohort studies. The results of subgroup analysis based on age and continents show that the heterogeneity was decreased in many of categories, but in fever and mortality not decreased. This heterogeneity rate confirms the difference between the combined studies. This difference may be due to differences in some section of studies such as the methods of measuring the outcomes and the tools used, the methods of sampling, presence of important underlying diseases (comorbidities) like diabetes, CHD, cancers, and chronic obstructive pulmonary disease (COPD). These variables and factors not reported in selected cohort studies in this meta‐analysis, so authors could not subgroup analysis based on its.\nThe results of this research are also based on cohort studies, which are one of the most important observational studies in order to find a causal association, but the overall results of this study have not yet determined the exact association between SARS‐CoV‐2 infection and chronic liver disorders. In other words, according to the results of published studies, it can be claimed that there is a kind of causal association between these two factors, and in order to find it, studies based on genetics or molecular science such as Mendelian randomization are needed.", "YM: concept development (provided idea for the research). MA and PM: search strategy. MA, PM, and KZ: data extraction. YM: supervision. FM, SK, and MA: analysis/interpretation. All authors: writing (responsible for writing a substantive part of the manuscript)." ]
[ null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "METHODS", "Search strategy and screening", "Inclusion and exclusion criteria", "Data extraction", "Risk of bias", "Statistical analysis", "RESULTS", "Subgroups analysis", "Discussion", "CONCLUSION", "CONFLICT OF INTEREST", "AUTHOR CONTRIBUTIONS" ]
[ "The chronic liver disease (CLD) is a progressive deterioration of liver function over 6 months, which slowly progresses, leaving the liver unable to synthesize coagulation factors, and proteins, detoxify harmful metabolic products, and excrete bile.\n1\n, \n2\n It is a continuous process of inflammation, destruction, and regeneration of the liver parenchyma, which leads to fibrosis and cirrhosis.\n1\n, \n3\n CLD is caused by a wide range of causes, including toxins, long‐term alcohol abuse, infection, autoimmune diseases, genetic disorders, and metabolic disorders.\n1\n, \n4\n Cirrhosis is the final stage of CLD, which leads to liver dysfunction, extensive nodule formation, vascular reorganization, neo‐angiogenesis, and extracellular matrix deposition.\n1\n, \n5\n, \n6\n The underlying mechanism of fibrosis and cirrhosis at the cellular level is the uptake of stellate cells and fibroblasts, which leads to fibrosis while parenchymal regeneration relies on liver stem cells.\n1\n, \n7\n The CLD is a very common clinical condition and the focus is more on its common causes, clinical manifestations and management.\n1\n, \n8\n, \n9\n, \n10\n, \n11\n About 1.5 billion people worldwide have CLD, which causes more than 2 million deaths a year.\n9\n, \n12\n With the rapid spread of COVID‐19, there are considerable concerns that patients with CLD are a vulnerable population and at higher risks for the more severe form of COVID‐19 and its associated complications.\n13\n Early on, patients with underlying diseases such as diabetes, chronic lung diseases, cardiovascular diseases, hypertension, and cancer were labeled as those at high risks for severe COVID‐19.\n14\n Although the virus mainly affects the lungs, experiences of China and the United States suggest SARS‐CoV‐2 may affect extra‐pulmonary systems, including the gastrointestinal and hepato‐biliary systems.\n15\n, \n16\n\n\nHowever, it was not initially clear whether patients with the CLD were more susceptible to COVID‐19. Data from some recent studies showed the CLD in the absence of immunosuppressive therapy was not associated with an increased risk of COVID‐19,\n17\n whereas results from other studies showed the CLD, including patients with cirrhosis and non‐cirrhosis, was associated with higher rates of mortality because of COVID‐19.\n18\n, \n19\n, \n20\n Also, various reports from different countries showed more than half of the hospitalized adults due to COVID‐19 had abnormal aminotransferase levels and 2% to 11% of them had underlying liver diseases.\n17\n, \n21\n, \n22\n, \n23\n, \n24\n, \n25\n However, there are limited and conflicting reports on the liver disease nature among COVID‐19 patients, and it is unclear how underlying CLD affects liver injuries and clinical outcomes in these patients.\n26\n Due to the discrepancies in the results of previous studies and the importance of determining the association between various underlying diseases, especially liver disorders, and COVID‐19 and its severe form, the authors in this meta‐analysis decided to combine the results of published cohort studies to determine the exact association between chronic liver disorders, and the severe form of COVID‐19, and its associated complications.", "The guideline of Preferred Reporting Items for Systematic Reviews and Meta‐Analyses (PRISMA) was used to review meta‐analyses, and systematic reviews.\n27\n Also, the study protocol was registered in PROSPER with the code CRD42022327806.\n[SUBTITLE] Search strategy and screening [SUBSECTION] The search was performed without language restrictions. The search strategy consisted of the following main keywords extracted from Mesh:\n“COVID 19,” “Liver Disease,” and “Chronic Liver Dysfunction.”\nSearch databases included PubMed (Medline), Scopus, Web of Sciences, and Embase (Elsevier).\nThe search deadline was from January 1, 2019, to January 1, 2022. Duplicate published articles were removed considering their titles, authors, and publication years using Endnote software version 9. Then, the remaining studies were evaluated by reviewing their titles, abstracts, and full texts, considering the inclusion criteria. In addition to searching the mentioned databases, gray literature was searched by reviewing articles in the first 10 pages of Google scholar, and manual search was performed by reviewing references of related studies. Two authors (MA, MA) independently screened articles based on their titles, abstracts, and full texts, and disputes were resolved by the third one (YM). After screening, the studies were finally selected by evaluating their full texts.\nThe search was performed without language restrictions. The search strategy consisted of the following main keywords extracted from Mesh:\n“COVID 19,” “Liver Disease,” and “Chronic Liver Dysfunction.”\nSearch databases included PubMed (Medline), Scopus, Web of Sciences, and Embase (Elsevier).\nThe search deadline was from January 1, 2019, to January 1, 2022. Duplicate published articles were removed considering their titles, authors, and publication years using Endnote software version 9. Then, the remaining studies were evaluated by reviewing their titles, abstracts, and full texts, considering the inclusion criteria. In addition to searching the mentioned databases, gray literature was searched by reviewing articles in the first 10 pages of Google scholar, and manual search was performed by reviewing references of related studies. Two authors (MA, MA) independently screened articles based on their titles, abstracts, and full texts, and disputes were resolved by the third one (YM). After screening, the studies were finally selected by evaluating their full texts.\n[SUBTITLE] Inclusion and exclusion criteria [SUBSECTION] The inclusion criteria were defined based on the PECOT structure as follows:\nPopulation included the whole population, whether people with the CLD or healthy ones, exposure was considered as the presence of chronic liver disorders, comparison included comparing healthy people with those with chronic liver disorders, and outcomes included the COVID‐19 symptoms and its associated complications, such as hospitalization in the intensive care unit, death, and acute respiratory distress syndrome (ARDS). The intended studies to conduct meta‐analysis were cohort ones because this type of observational studies is much more important than others to examine the casual association.\nSystematic reviews, case reports, case series, case controls, cross sectionals, clinical trials, and other interventional studies as well as letters to the editor were excluded from the present research. Also, studies that met the inclusion criteria but their full texts were not available, first by sending an email to their authors, the full texts were requested and if the authors did not respond, they were removed.\nThe inclusion criteria were defined based on the PECOT structure as follows:\nPopulation included the whole population, whether people with the CLD or healthy ones, exposure was considered as the presence of chronic liver disorders, comparison included comparing healthy people with those with chronic liver disorders, and outcomes included the COVID‐19 symptoms and its associated complications, such as hospitalization in the intensive care unit, death, and acute respiratory distress syndrome (ARDS). The intended studies to conduct meta‐analysis were cohort ones because this type of observational studies is much more important than others to examine the casual association.\nSystematic reviews, case reports, case series, case controls, cross sectionals, clinical trials, and other interventional studies as well as letters to the editor were excluded from the present research. Also, studies that met the inclusion criteria but their full texts were not available, first by sending an email to their authors, the full texts were requested and if the authors did not respond, they were removed.\n[SUBTITLE] Data extraction [SUBSECTION] After three stages of evaluation of titles, abstracts, and full texts, the selected articles were retrieved for detailed analysis. Data were collected using a checklist included authors' names, country, publication year, study type, study population, sample size, data source, age, number of patients with COVID‐19, hospitalization and mortality due to COVID‐19, ARDS, need for ventilation, ICU hospitalization, and evaluation of COVID‐19 symptoms (fever, cough, weakness, chest pain, abdominal pain, CT scan, Chest X‐ray, respiratory problems and shortness of breath, decreased sense of smell decreased sense of taste, fatigue, headache, dizziness, myalgia, diarrhea, nausea, and vomiting).\nAfter three stages of evaluation of titles, abstracts, and full texts, the selected articles were retrieved for detailed analysis. Data were collected using a checklist included authors' names, country, publication year, study type, study population, sample size, data source, age, number of patients with COVID‐19, hospitalization and mortality due to COVID‐19, ARDS, need for ventilation, ICU hospitalization, and evaluation of COVID‐19 symptoms (fever, cough, weakness, chest pain, abdominal pain, CT scan, Chest X‐ray, respiratory problems and shortness of breath, decreased sense of smell decreased sense of taste, fatigue, headache, dizziness, myalgia, diarrhea, nausea, and vomiting).\n[SUBTITLE] Risk of bias [SUBSECTION] Two of the authors (MA and PM) conducted a qualitative evaluation of the studies based on the Newcastle–Ottawa Quality Assessment Scale (NOS) checklist designed to evaluate the quality of observational studies. This tool examines each research with eight items in three groups, including how to select study samples, how to compare and analyze study groups, and how to measure and analyze the desired outcome. Each of these items is given a score of one if it is observed in the studies, and the maximum score for each study is 9 points. In case of discrepancies in the score assigned to the published articles, the discussion method and the third researcher were applied to reach an agreement.\nTwo of the authors (MA and PM) conducted a qualitative evaluation of the studies based on the Newcastle–Ottawa Quality Assessment Scale (NOS) checklist designed to evaluate the quality of observational studies. This tool examines each research with eight items in three groups, including how to select study samples, how to compare and analyze study groups, and how to measure and analyze the desired outcome. Each of these items is given a score of one if it is observed in the studies, and the maximum score for each study is 9 points. In case of discrepancies in the score assigned to the published articles, the discussion method and the third researcher were applied to reach an agreement.\n[SUBTITLE] Statistical analysis [SUBSECTION] To calculate the association, cumulative risk ratio (RR) with the 95% confidence interval and the meta set command were used considering logarithm and logarithm standard deviation of the RR. Heterogeneity was assessed between studies using the I\n2 and Q Cochrane tests. Egger test was used to evaluate the publication bias. Statistical analysis was performed using STATA 16.0 and P‐value < 0.05 was considered.\nTo calculate the association, cumulative risk ratio (RR) with the 95% confidence interval and the meta set command were used considering logarithm and logarithm standard deviation of the RR. Heterogeneity was assessed between studies using the I\n2 and Q Cochrane tests. Egger test was used to evaluate the publication bias. Statistical analysis was performed using STATA 16.0 and P‐value < 0.05 was considered.", "The search was performed without language restrictions. The search strategy consisted of the following main keywords extracted from Mesh:\n“COVID 19,” “Liver Disease,” and “Chronic Liver Dysfunction.”\nSearch databases included PubMed (Medline), Scopus, Web of Sciences, and Embase (Elsevier).\nThe search deadline was from January 1, 2019, to January 1, 2022. Duplicate published articles were removed considering their titles, authors, and publication years using Endnote software version 9. Then, the remaining studies were evaluated by reviewing their titles, abstracts, and full texts, considering the inclusion criteria. In addition to searching the mentioned databases, gray literature was searched by reviewing articles in the first 10 pages of Google scholar, and manual search was performed by reviewing references of related studies. Two authors (MA, MA) independently screened articles based on their titles, abstracts, and full texts, and disputes were resolved by the third one (YM). After screening, the studies were finally selected by evaluating their full texts.", "The inclusion criteria were defined based on the PECOT structure as follows:\nPopulation included the whole population, whether people with the CLD or healthy ones, exposure was considered as the presence of chronic liver disorders, comparison included comparing healthy people with those with chronic liver disorders, and outcomes included the COVID‐19 symptoms and its associated complications, such as hospitalization in the intensive care unit, death, and acute respiratory distress syndrome (ARDS). The intended studies to conduct meta‐analysis were cohort ones because this type of observational studies is much more important than others to examine the casual association.\nSystematic reviews, case reports, case series, case controls, cross sectionals, clinical trials, and other interventional studies as well as letters to the editor were excluded from the present research. Also, studies that met the inclusion criteria but their full texts were not available, first by sending an email to their authors, the full texts were requested and if the authors did not respond, they were removed.", "After three stages of evaluation of titles, abstracts, and full texts, the selected articles were retrieved for detailed analysis. Data were collected using a checklist included authors' names, country, publication year, study type, study population, sample size, data source, age, number of patients with COVID‐19, hospitalization and mortality due to COVID‐19, ARDS, need for ventilation, ICU hospitalization, and evaluation of COVID‐19 symptoms (fever, cough, weakness, chest pain, abdominal pain, CT scan, Chest X‐ray, respiratory problems and shortness of breath, decreased sense of smell decreased sense of taste, fatigue, headache, dizziness, myalgia, diarrhea, nausea, and vomiting).", "Two of the authors (MA and PM) conducted a qualitative evaluation of the studies based on the Newcastle–Ottawa Quality Assessment Scale (NOS) checklist designed to evaluate the quality of observational studies. This tool examines each research with eight items in three groups, including how to select study samples, how to compare and analyze study groups, and how to measure and analyze the desired outcome. Each of these items is given a score of one if it is observed in the studies, and the maximum score for each study is 9 points. In case of discrepancies in the score assigned to the published articles, the discussion method and the third researcher were applied to reach an agreement.", "To calculate the association, cumulative risk ratio (RR) with the 95% confidence interval and the meta set command were used considering logarithm and logarithm standard deviation of the RR. Heterogeneity was assessed between studies using the I\n2 and Q Cochrane tests. Egger test was used to evaluate the publication bias. Statistical analysis was performed using STATA 16.0 and P‐value < 0.05 was considered.", "First, 2251 studies were collected by searching based on the search strategy in the desired databases, of which 491 studies were duplicated, and 1760 ones remained. After reviewing the remained articles based on their titles, abstracts and full texts, 17 studies were selected for analysis. All articles were conducted in 2020 and 2021 and were cohorts (Figure 1 and Tables 1 and 2).\nThe flowchart of search strategy and syntax\nCharacteristics of included cohort studies (items related to COVID‐19 complications)\nBahardoust, M.\n(2021) (Iran)\nLiver disease + Covid‐19\nCovid‐19\nLiver disease = 81\nNon‐liver disease = 921\nLiver disease = 81\nNon‐liver disease = 921\nLiver disease = 81\nNon‐liver disease = 921\nLiver disease = 76\nnon‐liver disease = 598\n>7 day\nLiver disease = 10\nnon‐liver disease = 65\nCovid‐19 alone\nCovid‐19 + Cirrhosis\nCirrhosis alone\nCOVID‐19 alone = 108\nCOV + Cirrhosis = 37\nCirrhosis alone = 127\nCOVID‐19 alone = 108\nCOVID‐19 + cirrhosis = 37\nCOVID‐19 alone = 108\nCOVID‐19 + cirrhosis = 37\nCirrhosis alone = 127\nCovid‐19 = 15\nCovid‐19 + Cirrhosis = 11\nCirrhosis = 24\nCovid‐19 = 41\nCovid‐19 + Cirrhosis = 16\nCirrhosis = 31\nCovid‐19 = 41\nCOVID‐19 + Cirrhosis = 14\nCirrhosis = 18\nDavidov‐Derevynko, Y. (2021)\n(Israel)\nCOVID + Liver\nCOVID + non‐Liver\nCovid‐19 = 323\nCovid‐19 + Liver = 59\nCOVID‐19 = 323\nCOVID‐19 + Liver = 59\nCOVID‐19 = 323\nCOVID‐19 + Liver = 59\nCOVID‐19 = 22\nCOVID‐19 + Liver = 10\nCOVID‐19 = 36\nCOVID‐19 + Liver = 9\nCOVID‐19 = 35\nCOVID‐19 + Liver = 9\nNAFLD + Covid‐19\nNon‐NAFLD + Covid‐19\nNAFLD + Covid‐19 = 61\nCovid‐19 = 132\nNAFLD +Covid‐19 = 61\nCovid‐19 = 132\nNAFLD +Covid‐19 = 61\nCovid‐19 = 132\nNAFLD + Covid‐19 = 18\nCovid‐19 = 41\nNAFLD + Covid‐19 = 11\nCovid‐19 = 27\nCovid‐19 id\nCovid‐19 + Liver\nCovid‐19 = 2895\nCovid‐19 + Liver = 457\n64.8\nCovid‐19 = 2895\nCovid‐19 + Liver = 457\nCovid‐19 = 2895\nCovid‐19 + Liver = 457\nCovid‐19 = 769\nCovid‐19 + Liver = 135\nCovid‐19 = 522\nCovid‐19 + Liver = 108\nCovid‐19 id\nCovid‐19 + Liver\nCovid‐19 = 303\nCovid‐19 + Liver = 14\n70.5\nCovid‐19 = 303\nCovid‐19 + Liver = 14\nCovid‐19 = 303\nCovid‐19 + Liver = 14\nCovid‐19 = 67\nCovid‐19 + Liver = 4\nCovid‐19 = 18\nCovid‐19 + Liver = 1\nCovid‐19\nCovid‐19 + Liver\nCovid‐19 = 419\nCovid‐19 + Liver = 28\n40.8\nCovid‐19 = 419\nCovid‐19 + Liver = 28\nCovid‐19 = 200\nCovid‐19 + Liver = 26\nCovid‐19 = 39\nCovid‐19 + Liver = 8\nCovid‐19 = 28\nCovid‐19 + Liver = 3\nCovid‐19 = 19\nCovid‐19 + Liver = 2\nCovid‐19\nCovid‐19 + Liver\nCovid‐19 = 294\nCovid‐19 + Liver = 69\nCovid‐19 = 294\nCovid‐19 + Liver = 69\nCovid‐19 = 294\nCovid‐19 + Liver = 69\nCovid‐19 = 39\nCovid‐19 + Liver = 16\nCovid‐19 = 103\nCovid‐19 + Liver = 34\nCovid‐19 = 89\nCovid‐19 + Liver = 33\nNon‐NAFLD + Covid‐19\nNAFLD + Covid‐19\nCovid‐19 = 194\nCovid‐19 + Liver = 86\nCovid‐19 = 194\nCovid‐19 + Liver = 86\nCovid‐19 = 194\nCovid‐19 + Liver = 86\nCovid‐19 = 0\nCovid‐19 + Liver = 0\nCovid‐19 = 2\nCovid‐19 + Liver = 2\nCovid‐19 = 13\nCovid‐19 + Liver = 5\nCovid‐19\nCovid‐19 + Liver\nCovid‐19 = 118\nCovid‐19 + Liver = 22\nCovid‐19 = 118\nCovid‐19 + Liver = 22\nCovid‐19 = 118\nCovid‐19 + Liver = 22\nCovid‐19 = 0\nCovid‐19 + Liver = 1\nCovid‐19\nCovid‐19 + Liver\nCOVID‐19 + Liver = 47\nCOVID‐19 = 958\nCOVID‐19 + Liver = 47\nCOVID‐19 = 958\nCOVID‐19 + Liver = 47\nCOVID‐19 = 958\nCOVID‐19 + Liver = 7\nCOVID‐19 = 70\nCOVID‐19 + Liver = 8\nCOVID‐19 = 105\nCOVID‐19 + Liver = 8\nCOVID‐19 = 89\nInvasive mechanical ventilation\nCOVID‐19 + Liver = 4\nCOVID‐19 = 66\nInvasive mechanical ventilation and ECMO\nCOVID‐19 + Liver = 1\nCOVID‐19 = 17\nCovid‐19\nCovid‐19 + Liver\nCOVID‐19 + Liver = 52\nCovid‐19 = 52\nCOVID‐19 + Liver = 52\nCOVID‐19 = 52\nCOVID‐19 + Liver = 52\nCOVID‐19 = 52\nCOVID‐19 + Liver = 9\nCOVID‐19 = 0\nNon‐invasive ventilation\nCOVID‐19 = 2\nCOVID‐19 + Liver = 5\nInvasive mechanical ventilation\nCOVID‐19 = 1\nCOVID‐19 + Liver = 5\nNO ventilation\nCOVID‐19 = 49\nCOVID‐19 + Liver = 42\nCOVID‐19\nCOVID‐19 + Liver\nCOVID‐19 = 105\nCOVID‐19 + Liver = 34\nCOVID‐19 = 105\nCOVID‐19 + Liver = 34\nCOVID‐19 = 105\nCOVID‐19 + Liver = 34\nCOVID‐19 = 29\nCOVID‐19 + Liver = 9\nCOVID‐19 = 41\nCOVID‐19 + Liver = 19\nCOVID‐19 = 25\nCOVID‐19 + Liver = 14\nCOVID‐19 = 11\nCOVID‐19 + Liver = 7\nCOVID‐19\nCOVID‐19 + Liver\nCOVID‐19 = 243 634\nCOVID‐19 + Liver = 15 476\nCOVID‐19 = 243 634\nCOVID‐19 + Liver = 15 476\nCOVID‐19 = 243 634\nCOVID‐19 + Liver = 15 476\nCOVID‐19 = 35 262\nCOVID‐19 + Liver = 2941\nCOVID‐19 = 16 449\nCOVID‐19 + Liver = 1600\n1. Noncirrhosis/negative\n2. Non‐cirrhosis/positive;\n3. Cirrhosis/negative;\n4. Cirrhosis/positive\n1. Healthy = 128 864\n2. COVID‐19 = 29 446\n3. Cirrhosis = 53 476\n4. Cirrhosis + Covid‐19 = 8941\n2. COVID‐19 = 29 446\n4. Cirrhosis + Covid‐19 = 8941\nCovid‐19 + Liver\nCovid‐19\nAll = 2780\nCovid‐19 + Liver = 250\nCovid‐19 = 2530\nAll = 2780\nCovid‐19 + Liver = 250\nCovid‐19 = 2530\nCovid‐19 + Liver = 130\nCovid‐19 = 760\nCovid‐19 + Liver = 30\nCovid‐19 = 110\nNAFLD + Covid‐19\nNon‐NAFLD + Covid‐19\nNAFLD = 553\nNon‐NAFLD = 2736\nNAFLD = 553\nNon‐NAFLD = 2736\nNAFLD = 553\nNon‐NAFLD = 2736\nNAFLD = 60\nNon‐NAFLD = 239\nNAFLD = 196\nNon‐NAFLD = 726\nNAFLD = 76\nNon‐NAFLD = 221\nCharacteristics of included cohort studies (items related to signs and symptoms of COVID‐19)\nLiver disease\n≥38.5 = 35\n<38.5 = 46\nNon‐liver disease\n≥38.5 = 175\n<38.5 = 746\nLiver disease = 47\nNon‐liver disease = 552\nLiver disease = 20\nNon‐liver disease = 221\nLiver disease = 20\nNon‐liver disease = 313\nLiver disease = 26\nNon‐liver disease =\n275\nLiver disease = 49\nNon‐liver disease = 485\nLiver disease = 39\nNon‐liver disease = 230\nLiver disease = 37\nNon‐liver disease = 276\nLiver disease = 13\nNon‐liver disease = 166\nCovid‐19 = 75\nCovid‐19 + Cirrhosis = 20\nCovid‐19 = 70\nCovid‐19 + Cirrhosis = 26\nCovid‐19 = 19\nCovid‐19 + Cirrhosis = 3\nCovid‐19 = 8\nCovid‐19 + Cirrhosis = 4\nCovid‐19 = 15\nCovid‐19 + Cirrhosis = 2\nCovid‐19 = 31\nCovid‐19 + Cirrhosis = 4\nCovid‐19 = 29\nCovid‐19 + Cirrhosis = 3\nCovid‐19 = 29\nCovid‐19 + Cirrhosis = 4\nCovid‐19 = 74\nCovid‐19 + Cirrhosis = 23\nCovid‐19 = 17\nCovid‐19 + Cirrhosis = 9\nCovid‐19 = 44\nCovid‐19 + Cirrhosis = 12\nCovid‐19 = 174\nCovid‐19 + Liver = 8\nCovid‐19 = 185\nCovid‐19 + Liver = 10\nCovid‐19 = 46\nCovid‐19 + Liver = 0\nCovid‐19 = 69\nCovid‐19 + Liver = 2\nCovid‐19 = 38\nCovid‐19 + Liver = 2\nCovid‐19 = 23\nCovid‐19 + Liver = 1\nCovid‐19 = 17\nCovid‐19 + Liver = 2\nCovid‐19 = 82\nCovid‐19 + Liver = 3\nCovid‐19 = 108\nCovid‐19 + Liver = 6\nCovid‐19 = 132\nCovid‐19 + Liver = 55\nCovid‐19 = 109\nCovid‐19 + Liver = 47\nCovid‐19 = 16\nCovid‐19 + Liver = 3\nCovid‐19 = 21\nCovid‐19 + Liver = 7\nCovid‐19 = 16\nCovid‐19 + Liver = 7\nCovid‐19 = 42\nCovid‐19 + Liver = 16\nCOVID‐19 + Liver = 24\nCOVID‐19 = 425\nCOVID‐19 + Liver = 23\nCOVID‐19 = 544\nCOVID‐19 + Liver = 8\nCOVID‐19 = 245\nCOVID‐19 + Liver = 9\nCOVID‐19 = 316\nCOVID‐19 + Liver = 15\nCOVID‐19 = 232\nNAFLD = 319\nNon‐NAFLD = 1283\nNAFLD = 318\nNon‐NAFLD = 1273\nNAFLD = 23\nNon‐NAFLD = 72\nNAFLD = 74\nNon‐NAFLD = 240\nNAFLD = 101\nNon‐NAFLD = 363\nNAFLD = 23\nNon‐NAFLD = 72\nNAFLD = 316\nNon‐NAFLD = 1519\nNAFLD = 144\nNon‐NAFLD = 588\nThe risk of death in COVID‐19 patients with chronic liver disorders was compared with that in ones without the CLD in 15 studies.\n19\n, \n26\n, \n28\n, \n29\n, \n30\n, \n31\n, \n32\n, \n33\n, \n34\n, \n35\n, \n36\n, \n37\n, \n38\n, \n39\n, \n40\n, \n42\n In all of these studies, the mortality rate in patients with COVID‐19, and the CLD was higher than that in patients with COVID‐19, and without the CLD.\n19\n, \n26\n, \n28\n, \n30\n, \n32\n, \n33\n, \n34\n, \n36\n, \n37\n, \n38\n, \n39\n, \n40\n The cumulative RR after combining these studies was 1.52 (CI 95%: 1.46–1.57; I\n2: 86.14%) (Figure 2).\nThe pooled effect (risk ratio, RR) of chronic liver disorders on the mortality and admission to ICU related to COVID‐19\nThe risk of ARDS in patients with COVID‐19 who had chronic liver disorders was compared in three studies with those without chronic liver disorders.\n35\n, \n37\n, \n39\n The cumulative RR after combining these studies was 1.65 (CI 95%: 1.09–2.50; I\n2: 0.00%) (Table 3).\nResults of meta‐analysis and reports pooled odds ratio based on symptoms and complications related to COVID‐19\n\nNote: A: COVID‐19 individual with CLD; B: Healthy individual with CLD; C: COVID‐19 individual without CLD; D: Healthy individual without CLD.\nThe risk of hospitalization of COVID‐19 patients who had chronic liver disorders in the intensive care unit was compared with that of ones without the CLD in nine studies,\n26\n, \n29\n, \n30\n, \n31\n, \n34\n, \n35\n, \n37\n, \n39\n, \n42\n, \n43\n in all of which the risk of hospitalization in the ICU was higher in the group of COVID‐19 patients with the CLD.\n26\n, \n29\n, \n30\n, \n31\n, \n34\n, \n35\n, \n37\n, \n39\n The cumulative RR after combining these studies was 1.39 (CI 95%: 1.23–1.58) with heterogeneity (I\n2) of 0.2% (Figure 2).\nThe risk of need for ventilation in COVID‐19 patients who had the CLD was evaluated in 11 studies compared with those who did not have chronic liver disorders.\n26\n, \n29\n, \n30\n, \n32\n, \n33\n, \n34\n, \n37\n, \n38\n, \n39\n, \n40\n, \n42\n In all of these studies, the need for ventilation was higher in the group with the CLD\n26\n, \n29\n, \n30\n, \n32\n, \n33\n, \n34\n, \n37\n, \n38\n, \n39\n, \n40\n The frequency of need for ventilation was higher in the group of patients with COVID‐19 and liver disorders.\n42\n The cumulative RR after combining these studies was 1.53 (CI 95%: 1.46–1.60) with heterogeneity (I\n2) equal to 0.2% (Figure 3).\nThe pooled effect (risk ratio, RR) of chronic liver disorders on the ventilation needs, fever, and cough related to COVID‐19\nTo determine the association between the COVID‐19 symptoms and chronic liver disorders, patients with COVID‐19 who had chronic liver disorders were compared with those who did not have the CLD, and the results of the meta‐analysis showed cough, headache, myalgia, nausea, diarrhea and fatigue were 1.37 (CI 95%: 1.20–1.55), 1.23 (CI 95%: 1.09–1.38), 1.25 (CI 95%: 1.04–1.50), 1.19 (CI 95%: 1.02–1.40), 1.89 (CI 95%: 1.30–2.75), 1.49 (CI 95%: 1.07–2.09), and 1.14 (CI 95%: 0.98–1.33), respectively, while the risk of all these symptoms was higher in COVID‐19 patients with underlying liver diseases than ones without chronic liver disorders. Other symptoms have been reported in the table (Figure 3 and Table 3).\nTo examine the publication bias, the Eggers test was performed, which was not significant in all variables except myalgia and headache. That is, the publication bias has not occurred.\n[SUBTITLE] Subgroups analysis [SUBSECTION] The results of subgroups analysis according to age and continents for symptoms and complications related to COVID‐19 were reported in Table 4. The results show that complications related to COVID‐19 are more significant and severe in patients with CLD with age <60 and live in Asia (Table 4).\nThe subgroups analysis of signs, symptoms, and complications related to COVID 19 in CLD patients based on continents and age\nThe results of subgroups analysis according to age and continents for symptoms and complications related to COVID‐19 were reported in Table 4. The results show that complications related to COVID‐19 are more significant and severe in patients with CLD with age <60 and live in Asia (Table 4).\nThe subgroups analysis of signs, symptoms, and complications related to COVID 19 in CLD patients based on continents and age\n[SUBTITLE] Discussion [SUBSECTION] The RR of complications and consequences of COVID‐19 in patients with the CLD during the pandemic period was examined in this study and the results showed the need for ventilation and ARDS had higher risks than other outcomes related to COVID‐19 in patients with chronic liver disorders. Exacerbation of alveolar epithelial damages, increased vascular permeability, and systemic inflammation are among the factors that predispose to increased ARDS in patients with the CLD.\n44\n, \n45\n Also, because these patients had a higher BMI and obesity was more prevalent in them, their need for ventilation was higher when they were infected with COVID‐19.\n46\n\n\nDue to the direct and significant effect of SARS‐CoV‐2 virus on ACE‐2 receptors in liver cells, patients with chronic liver disorders will have a more severe form of the disease if they develop COVID‐19, which causes their hospitalization in the intensive care unit of hospitals due to COVID‐19. According to the results of previous studies, patients with chronic liver disorders have a higher risk of being admitted to the intensive care unit than others in the community if they develop COVID‐19. In the present meta‐analysis, the same result was obtained, which was in line with the results of preliminary studies published in the world.\n47\n However, the results of some studies in the world have not shown this association, for example, a study by Huang, R. et al. showed the risk of hospitalization in intensive care unit due to COVID‐19 in patients with chronic liver disorders was not significantly different from other people in the community.\n35\n The reason for this inconsistency can be attributed to differences in the study method, how to collect information, and the type of patients studied in these articles.\nAlso, the risk of death due to COVID‐19 in the present meta‐analysis was significantly higher in patients with chronic liver disorders than other individuals. The reason for this can be attributed to the increase in inflammatory cytokines such as IL‐6, Ferritin and TNF‐alpha.\n48\n In patients with COVID‐19, inflammatory cytokines increase, eventually leading to a cytokine storm in more severe forms of the disease. On the other hand, for patients with severe forms of COVID‐19, drugs that aggravate cytokines and cause cytokine storms are prescribed, which may also exacerbate death in COVID‐19 patients with chronic liver disorders. Coincidence of chronic liver disorders and the use of anti‐COVID‐19 drugs significantly increases the risk of mortality in COVID‐19 patients with the CLD. Also, worsening liver function reduces the number and disrupt the function of neutrophils, monocytes and innate immune proteins, and ultimately the number of both B and T lymphocytes involved in acquired immunity decreases and eventually immune dysfunction increases.\n49\n, \n50\n\n\nIn this study, in addition to the mentioned cases, the symptoms associated with COVID‐19 including fever, cough, headache, myalgia, nausea, diarrhea, and fatigue were also evaluated in patients with the CLD. The results of this meta‐analysis showed among these symptoms, nausea, diarrhea and abdominal pain were more common than other ones in patients with chronic liver disorders. Increased gastrointestinal symptoms may be due to liver dysfunction. In addition, other symptoms such as fever, cough, headache, myalgia, and fatigue were more common in patients with the underlying liver disease than in healthy individuals while cytokines were effective in worsening these symptoms.\n48\n The severity of various symptoms in patients with chronic liver disorders can affect outcomes of COVID‐19 and, as a result, increase the outcome and mortality in this group of patients.\nSome studies on the effect of COVID‐19 on the incidence of liver disorders published results, which suggested in the case of COVID‐19 due to the widespread distribution of the main virus receptor called the angiotensin‐converting enzyme 2 (ACE2), the virus could cause a widespread disease with more involvement of extra‐pulmonary organs, especially the liver. ACE2 receptors are also expressed in the gastrointestinal tract, vascular endothelium, and hepatic cholangiocytes.\n51\n, \n52\n Various studies showed elevated liver enzymes indicated liver damages and were common in COVID‐19 patients with chronic and non‐CLDs.\n52\n On the other hand, initial clinical studies in this area also confirmed a significant increase in liver enzymes such as ALT, and AST due to SARS‐CoV‐2 infection.\n53\n, \n54\n, \n55\n, \n56\n The results of a meta‐analysis also showed the levels of ALP, and γ‐GT enzymes were significant as a result of cholangiocellular damages.\n57\n, \n58\n, \n59\n As the study results show, it is not yet clear how much an increase in liver enzyme levels can exacerbate the complications of COVID‐19 or the disease progression. In patients with COVID‐19 who have not had a chronic liver disorder or liver damage before infection, a slight liver disorder is found after recovery.\n60\n, \n61\n\n\nAbnormal results of liver tests were associated with more severe forms of COVID‐19 and mortality. Although RNAs of SARS‐CoV‐2 have been detected in the liver of patients with COVID‐19, it is not yet exactly clear how much SARS‐CoV‐2 infects the liver and multiplies in its cells.\n17\n, \n51\n The range of liver damages in the COVID‐19 disease may be direct infection by SARS‐CoV‐2, indirect involvement by systemic inflammation, and hypoxic changes. However, due to the major role of the liver in endobiotic and xenobiotic drug metabolism, coagulation, albumin, and production of acute phase reactants, liver dysfunction may affect the pathophysiology of the COVID‐19 disease.\n62\n, \n63\n The results of the present meta‐analysis along with other published results can be useful in finding many of these answers. They are also useful in updating treatment and prevention guidelines. This study was the first meta‐analysis to compare the complications and consequences associated with COVID‐19 in two groups of patients with the CLD and healthy ones. On the other hand, the results of subgroup analyses and overall results had a high homogeneity, which indicated the homogeneous and correct selection of initial studies in order to perform this meta‐analysis.\nThe heterogeneity in this meta‐analysis only was higher at two outcomes (Fever and Mortality). For detecting sources of this heterogeneity, all primary cohort studies were reviewed, and extracted related variables that reported in selected cohort studies completely. Between reported variables, age and continents were extracted. Other variables like type of underlying diseases, such as diabetes, coronary heart disease (CHD), and cancers, type of study population, type of measures tools for outcomes measure were not reported in selected cohort studies. The results of subgroup analysis based on age and continents show that the heterogeneity was decreased in many of categories, but in fever and mortality not decreased. This heterogeneity rate confirms the difference between the combined studies. This difference may be due to differences in some section of studies such as the methods of measuring the outcomes and the tools used, the methods of sampling, presence of important underlying diseases (comorbidities) like diabetes, CHD, cancers, and chronic obstructive pulmonary disease (COPD). These variables and factors not reported in selected cohort studies in this meta‐analysis, so authors could not subgroup analysis based on its.\nThe results of this research are also based on cohort studies, which are one of the most important observational studies in order to find a causal association, but the overall results of this study have not yet determined the exact association between SARS‐CoV‐2 infection and chronic liver disorders. In other words, according to the results of published studies, it can be claimed that there is a kind of causal association between these two factors, and in order to find it, studies based on genetics or molecular science such as Mendelian randomization are needed.\nThe RR of complications and consequences of COVID‐19 in patients with the CLD during the pandemic period was examined in this study and the results showed the need for ventilation and ARDS had higher risks than other outcomes related to COVID‐19 in patients with chronic liver disorders. Exacerbation of alveolar epithelial damages, increased vascular permeability, and systemic inflammation are among the factors that predispose to increased ARDS in patients with the CLD.\n44\n, \n45\n Also, because these patients had a higher BMI and obesity was more prevalent in them, their need for ventilation was higher when they were infected with COVID‐19.\n46\n\n\nDue to the direct and significant effect of SARS‐CoV‐2 virus on ACE‐2 receptors in liver cells, patients with chronic liver disorders will have a more severe form of the disease if they develop COVID‐19, which causes their hospitalization in the intensive care unit of hospitals due to COVID‐19. According to the results of previous studies, patients with chronic liver disorders have a higher risk of being admitted to the intensive care unit than others in the community if they develop COVID‐19. In the present meta‐analysis, the same result was obtained, which was in line with the results of preliminary studies published in the world.\n47\n However, the results of some studies in the world have not shown this association, for example, a study by Huang, R. et al. showed the risk of hospitalization in intensive care unit due to COVID‐19 in patients with chronic liver disorders was not significantly different from other people in the community.\n35\n The reason for this inconsistency can be attributed to differences in the study method, how to collect information, and the type of patients studied in these articles.\nAlso, the risk of death due to COVID‐19 in the present meta‐analysis was significantly higher in patients with chronic liver disorders than other individuals. The reason for this can be attributed to the increase in inflammatory cytokines such as IL‐6, Ferritin and TNF‐alpha.\n48\n In patients with COVID‐19, inflammatory cytokines increase, eventually leading to a cytokine storm in more severe forms of the disease. On the other hand, for patients with severe forms of COVID‐19, drugs that aggravate cytokines and cause cytokine storms are prescribed, which may also exacerbate death in COVID‐19 patients with chronic liver disorders. Coincidence of chronic liver disorders and the use of anti‐COVID‐19 drugs significantly increases the risk of mortality in COVID‐19 patients with the CLD. Also, worsening liver function reduces the number and disrupt the function of neutrophils, monocytes and innate immune proteins, and ultimately the number of both B and T lymphocytes involved in acquired immunity decreases and eventually immune dysfunction increases.\n49\n, \n50\n\n\nIn this study, in addition to the mentioned cases, the symptoms associated with COVID‐19 including fever, cough, headache, myalgia, nausea, diarrhea, and fatigue were also evaluated in patients with the CLD. The results of this meta‐analysis showed among these symptoms, nausea, diarrhea and abdominal pain were more common than other ones in patients with chronic liver disorders. Increased gastrointestinal symptoms may be due to liver dysfunction. In addition, other symptoms such as fever, cough, headache, myalgia, and fatigue were more common in patients with the underlying liver disease than in healthy individuals while cytokines were effective in worsening these symptoms.\n48\n The severity of various symptoms in patients with chronic liver disorders can affect outcomes of COVID‐19 and, as a result, increase the outcome and mortality in this group of patients.\nSome studies on the effect of COVID‐19 on the incidence of liver disorders published results, which suggested in the case of COVID‐19 due to the widespread distribution of the main virus receptor called the angiotensin‐converting enzyme 2 (ACE2), the virus could cause a widespread disease with more involvement of extra‐pulmonary organs, especially the liver. ACE2 receptors are also expressed in the gastrointestinal tract, vascular endothelium, and hepatic cholangiocytes.\n51\n, \n52\n Various studies showed elevated liver enzymes indicated liver damages and were common in COVID‐19 patients with chronic and non‐CLDs.\n52\n On the other hand, initial clinical studies in this area also confirmed a significant increase in liver enzymes such as ALT, and AST due to SARS‐CoV‐2 infection.\n53\n, \n54\n, \n55\n, \n56\n The results of a meta‐analysis also showed the levels of ALP, and γ‐GT enzymes were significant as a result of cholangiocellular damages.\n57\n, \n58\n, \n59\n As the study results show, it is not yet clear how much an increase in liver enzyme levels can exacerbate the complications of COVID‐19 or the disease progression. In patients with COVID‐19 who have not had a chronic liver disorder or liver damage before infection, a slight liver disorder is found after recovery.\n60\n, \n61\n\n\nAbnormal results of liver tests were associated with more severe forms of COVID‐19 and mortality. Although RNAs of SARS‐CoV‐2 have been detected in the liver of patients with COVID‐19, it is not yet exactly clear how much SARS‐CoV‐2 infects the liver and multiplies in its cells.\n17\n, \n51\n The range of liver damages in the COVID‐19 disease may be direct infection by SARS‐CoV‐2, indirect involvement by systemic inflammation, and hypoxic changes. However, due to the major role of the liver in endobiotic and xenobiotic drug metabolism, coagulation, albumin, and production of acute phase reactants, liver dysfunction may affect the pathophysiology of the COVID‐19 disease.\n62\n, \n63\n The results of the present meta‐analysis along with other published results can be useful in finding many of these answers. They are also useful in updating treatment and prevention guidelines. This study was the first meta‐analysis to compare the complications and consequences associated with COVID‐19 in two groups of patients with the CLD and healthy ones. On the other hand, the results of subgroup analyses and overall results had a high homogeneity, which indicated the homogeneous and correct selection of initial studies in order to perform this meta‐analysis.\nThe heterogeneity in this meta‐analysis only was higher at two outcomes (Fever and Mortality). For detecting sources of this heterogeneity, all primary cohort studies were reviewed, and extracted related variables that reported in selected cohort studies completely. Between reported variables, age and continents were extracted. Other variables like type of underlying diseases, such as diabetes, coronary heart disease (CHD), and cancers, type of study population, type of measures tools for outcomes measure were not reported in selected cohort studies. The results of subgroup analysis based on age and continents show that the heterogeneity was decreased in many of categories, but in fever and mortality not decreased. This heterogeneity rate confirms the difference between the combined studies. This difference may be due to differences in some section of studies such as the methods of measuring the outcomes and the tools used, the methods of sampling, presence of important underlying diseases (comorbidities) like diabetes, CHD, cancers, and chronic obstructive pulmonary disease (COPD). These variables and factors not reported in selected cohort studies in this meta‐analysis, so authors could not subgroup analysis based on its.\nThe results of this research are also based on cohort studies, which are one of the most important observational studies in order to find a causal association, but the overall results of this study have not yet determined the exact association between SARS‐CoV‐2 infection and chronic liver disorders. In other words, according to the results of published studies, it can be claimed that there is a kind of causal association between these two factors, and in order to find it, studies based on genetics or molecular science such as Mendelian randomization are needed.", "The results of subgroups analysis according to age and continents for symptoms and complications related to COVID‐19 were reported in Table 4. The results show that complications related to COVID‐19 are more significant and severe in patients with CLD with age <60 and live in Asia (Table 4).\nThe subgroups analysis of signs, symptoms, and complications related to COVID 19 in CLD patients based on continents and age", "The RR of complications and consequences of COVID‐19 in patients with the CLD during the pandemic period was examined in this study and the results showed the need for ventilation and ARDS had higher risks than other outcomes related to COVID‐19 in patients with chronic liver disorders. Exacerbation of alveolar epithelial damages, increased vascular permeability, and systemic inflammation are among the factors that predispose to increased ARDS in patients with the CLD.\n44\n, \n45\n Also, because these patients had a higher BMI and obesity was more prevalent in them, their need for ventilation was higher when they were infected with COVID‐19.\n46\n\n\nDue to the direct and significant effect of SARS‐CoV‐2 virus on ACE‐2 receptors in liver cells, patients with chronic liver disorders will have a more severe form of the disease if they develop COVID‐19, which causes their hospitalization in the intensive care unit of hospitals due to COVID‐19. According to the results of previous studies, patients with chronic liver disorders have a higher risk of being admitted to the intensive care unit than others in the community if they develop COVID‐19. In the present meta‐analysis, the same result was obtained, which was in line with the results of preliminary studies published in the world.\n47\n However, the results of some studies in the world have not shown this association, for example, a study by Huang, R. et al. showed the risk of hospitalization in intensive care unit due to COVID‐19 in patients with chronic liver disorders was not significantly different from other people in the community.\n35\n The reason for this inconsistency can be attributed to differences in the study method, how to collect information, and the type of patients studied in these articles.\nAlso, the risk of death due to COVID‐19 in the present meta‐analysis was significantly higher in patients with chronic liver disorders than other individuals. The reason for this can be attributed to the increase in inflammatory cytokines such as IL‐6, Ferritin and TNF‐alpha.\n48\n In patients with COVID‐19, inflammatory cytokines increase, eventually leading to a cytokine storm in more severe forms of the disease. On the other hand, for patients with severe forms of COVID‐19, drugs that aggravate cytokines and cause cytokine storms are prescribed, which may also exacerbate death in COVID‐19 patients with chronic liver disorders. Coincidence of chronic liver disorders and the use of anti‐COVID‐19 drugs significantly increases the risk of mortality in COVID‐19 patients with the CLD. Also, worsening liver function reduces the number and disrupt the function of neutrophils, monocytes and innate immune proteins, and ultimately the number of both B and T lymphocytes involved in acquired immunity decreases and eventually immune dysfunction increases.\n49\n, \n50\n\n\nIn this study, in addition to the mentioned cases, the symptoms associated with COVID‐19 including fever, cough, headache, myalgia, nausea, diarrhea, and fatigue were also evaluated in patients with the CLD. The results of this meta‐analysis showed among these symptoms, nausea, diarrhea and abdominal pain were more common than other ones in patients with chronic liver disorders. Increased gastrointestinal symptoms may be due to liver dysfunction. In addition, other symptoms such as fever, cough, headache, myalgia, and fatigue were more common in patients with the underlying liver disease than in healthy individuals while cytokines were effective in worsening these symptoms.\n48\n The severity of various symptoms in patients with chronic liver disorders can affect outcomes of COVID‐19 and, as a result, increase the outcome and mortality in this group of patients.\nSome studies on the effect of COVID‐19 on the incidence of liver disorders published results, which suggested in the case of COVID‐19 due to the widespread distribution of the main virus receptor called the angiotensin‐converting enzyme 2 (ACE2), the virus could cause a widespread disease with more involvement of extra‐pulmonary organs, especially the liver. ACE2 receptors are also expressed in the gastrointestinal tract, vascular endothelium, and hepatic cholangiocytes.\n51\n, \n52\n Various studies showed elevated liver enzymes indicated liver damages and were common in COVID‐19 patients with chronic and non‐CLDs.\n52\n On the other hand, initial clinical studies in this area also confirmed a significant increase in liver enzymes such as ALT, and AST due to SARS‐CoV‐2 infection.\n53\n, \n54\n, \n55\n, \n56\n The results of a meta‐analysis also showed the levels of ALP, and γ‐GT enzymes were significant as a result of cholangiocellular damages.\n57\n, \n58\n, \n59\n As the study results show, it is not yet clear how much an increase in liver enzyme levels can exacerbate the complications of COVID‐19 or the disease progression. In patients with COVID‐19 who have not had a chronic liver disorder or liver damage before infection, a slight liver disorder is found after recovery.\n60\n, \n61\n\n\nAbnormal results of liver tests were associated with more severe forms of COVID‐19 and mortality. Although RNAs of SARS‐CoV‐2 have been detected in the liver of patients with COVID‐19, it is not yet exactly clear how much SARS‐CoV‐2 infects the liver and multiplies in its cells.\n17\n, \n51\n The range of liver damages in the COVID‐19 disease may be direct infection by SARS‐CoV‐2, indirect involvement by systemic inflammation, and hypoxic changes. However, due to the major role of the liver in endobiotic and xenobiotic drug metabolism, coagulation, albumin, and production of acute phase reactants, liver dysfunction may affect the pathophysiology of the COVID‐19 disease.\n62\n, \n63\n The results of the present meta‐analysis along with other published results can be useful in finding many of these answers. They are also useful in updating treatment and prevention guidelines. This study was the first meta‐analysis to compare the complications and consequences associated with COVID‐19 in two groups of patients with the CLD and healthy ones. On the other hand, the results of subgroup analyses and overall results had a high homogeneity, which indicated the homogeneous and correct selection of initial studies in order to perform this meta‐analysis.\nThe heterogeneity in this meta‐analysis only was higher at two outcomes (Fever and Mortality). For detecting sources of this heterogeneity, all primary cohort studies were reviewed, and extracted related variables that reported in selected cohort studies completely. Between reported variables, age and continents were extracted. Other variables like type of underlying diseases, such as diabetes, coronary heart disease (CHD), and cancers, type of study population, type of measures tools for outcomes measure were not reported in selected cohort studies. The results of subgroup analysis based on age and continents show that the heterogeneity was decreased in many of categories, but in fever and mortality not decreased. This heterogeneity rate confirms the difference between the combined studies. This difference may be due to differences in some section of studies such as the methods of measuring the outcomes and the tools used, the methods of sampling, presence of important underlying diseases (comorbidities) like diabetes, CHD, cancers, and chronic obstructive pulmonary disease (COPD). These variables and factors not reported in selected cohort studies in this meta‐analysis, so authors could not subgroup analysis based on its.\nThe results of this research are also based on cohort studies, which are one of the most important observational studies in order to find a causal association, but the overall results of this study have not yet determined the exact association between SARS‐CoV‐2 infection and chronic liver disorders. In other words, according to the results of published studies, it can be claimed that there is a kind of causal association between these two factors, and in order to find it, studies based on genetics or molecular science such as Mendelian randomization are needed.", "The results of this study showed the mortality and consequences due to COVID‐19 were significantly different between patients with the CLD and the general population. Also, the COVID‐19 symptoms in people with liver disorders were significantly more severe than those in healthy people. So, taking measures is necessary to manage them. It is recommended to reduce the risk of mortality and other consequences of COVID‐19 through screening and treating people with liver disorders in the lower stages of the CLD.", "The authors declare that they have no competing interests.", "YM: concept development (provided idea for the research). MA and PM: search strategy. MA, PM, and KZ: data extraction. YM: supervision. FM, SK, and MA: analysis/interpretation. All authors: writing (responsible for writing a substantive part of the manuscript)." ]
[ null, "methods", null, null, null, null, null, "results", null, null, "conclusions", "COI-statement", null ]
[ "chronic liver disease", "complications", "COVID‐19", "meta‐analysis", "mortality" ]
Characteristics and Outcomes of Thrombolysis-Treated Stroke Patients With and Without Saccular Intracranial Aneurysms.
36254706
Intravenous thrombolysis seems safe in acute ischemic stroke patients with saccular, unruptured intracranial aneurysms (UIAs), but little is known about the differences in cardiovascular risk factors and outcomes between intravenous thrombolysis-treated stroke patients with and without UIAs. We hypothesized that UIA patients would have a higher burden of cardiovascular risk factors and, therefore, a higher risk of an unfavorable outcome.
BACKGROUND
In this prospective cohort study conducted in Helsinki University Hospital, we identified intravenous thrombolysis-treated patients with concurrent saccular UIAs admitted to a comprehensive stroke center between 2005 and 2019 using 2 overlapping methods. For each UIA patient, a control patient was identified and matched (1:1) for age, sex, admission year, and stroke severity. The primary outcome was an unfavorable outcome at 3 months, defined as a modified Rankin Scale (mRS) score 3 to 6. The secondary outcomes were an excellent outcome (mRS score 0-1) at 3 months and mRS difference in shift analysis.
METHODS
In total, 118 UIA patients and 118 matched control patients were identified. The UIA patients were more often current smokers, and their admission systolic blood pressure was higher. The rate of hemorrhagic complications did not differ between the groups. UIAs were not associated with an unfavorable outcome in the conditional logistic regression analysis (odds ratio, 1.41 [95% CI, 0.79-2.54]; P=0.25). However, the UIA patients were less likely to have excellent outcomes (odds ratio for non-excellent outcome, 2.09 [95% CI, 1.13-3.85]; P=0.02). In shift analysis, UIAs were associated with higher mRS (odds ratio, 1.61 [95% CI, 1.03-2.49]; P=0.04).
RESULTS
The intravenous thrombolysis-treated stroke patients with UIAs were more often current smokers and had higher systolic blood pressure than the matched patients without UIAs. They were as likely to have unfavorable outcomes at 3 months but seemed less likely to achieve excellent outcomes and were more likely to have higher mRS in shift analysis.
CONCLUSIONS
[ "Humans", "Intracranial Aneurysm", "Prospective Studies", "Ischemic Stroke", "Retrospective Studies", "Stroke", "Thrombolytic Therapy", "Treatment Outcome" ]
9698101
null
null
Methods
[SUBTITLE] Research Materials Transparency [SUBSECTION] Anonymized data that support the findings of this study are available from the corresponding author upon reasonable request. Anonymized data that support the findings of this study are available from the corresponding author upon reasonable request. [SUBTITLE] Study Cohort [SUBSECTION] We have previously described our cohort of UIA patients treated with IVT for ischemic stroke.1 We used the prospectively collected Helsinki Stroke Thrombolysis Registry2 to identify IVT-treated patients at Helsinki University Hospital between 2005 and 2019. We employed 2 overlapping methods to retrospectively identify patients with UIAs from the registry. First, we screened the radiology reports of all the patients in the registry for mentions of UIAs. The reports were limited to studies performed 1 day before or within 7 days of the IVT. Second, for all the patients in the thrombolysis registry, we searched the nationwide Finnish Care Register for Health Care for a UIA diagnosis made any time before or within 1 year after the IVT or for a diagnosis of subarachnoid hemorrhage dated within 1 year after the IVT. We then scrutinized the imaging studies of all patients identified through either method to confirm the presence of UIAs. A flowchart of the study is shown in Figure S1. Only patients with saccular UIAs were included in the study, as we previously found that 6 of 14 (43%) patients with fusiform UIAs died due to aneurysm rupture between 1 day and 5 months after IVT.1 In the current study, each UIA patient was matched with a control patient without a UIA. The UIA patients and controls were matched on age (±3 years), sex, admission year (±1 year), and the National Institutes of Health Stroke Scale (NIHSS)3 category reflecting symptom severity (0–4, mild; 5–15, moderate; 16–20, severe; >20, extremely severe) on admission. Regarding the selection of the matching parameters, older age and severe symptoms are associated with worse outcomes after IVT. The admission year was included as a matching parameter because acute stroke treatment has evolved during the cohort’s collection period (eg, thrombectomy has become the standard treatment in large vessel occlusions). The matching based on the aforementioned parameters was performed randomly. The local institutional research committee approved the study and waived the requirement for patient consent (HUS/203/2020 §14). The corresponding author had full access to all the data in the study and takes responsibility for its integrity and the data analysis. The study was conducted according to the Strengthening the Reporting of Observational studies in Epidemiology (STROBE) Statement. We have previously described our cohort of UIA patients treated with IVT for ischemic stroke.1 We used the prospectively collected Helsinki Stroke Thrombolysis Registry2 to identify IVT-treated patients at Helsinki University Hospital between 2005 and 2019. We employed 2 overlapping methods to retrospectively identify patients with UIAs from the registry. First, we screened the radiology reports of all the patients in the registry for mentions of UIAs. The reports were limited to studies performed 1 day before or within 7 days of the IVT. Second, for all the patients in the thrombolysis registry, we searched the nationwide Finnish Care Register for Health Care for a UIA diagnosis made any time before or within 1 year after the IVT or for a diagnosis of subarachnoid hemorrhage dated within 1 year after the IVT. We then scrutinized the imaging studies of all patients identified through either method to confirm the presence of UIAs. A flowchart of the study is shown in Figure S1. Only patients with saccular UIAs were included in the study, as we previously found that 6 of 14 (43%) patients with fusiform UIAs died due to aneurysm rupture between 1 day and 5 months after IVT.1 In the current study, each UIA patient was matched with a control patient without a UIA. The UIA patients and controls were matched on age (±3 years), sex, admission year (±1 year), and the National Institutes of Health Stroke Scale (NIHSS)3 category reflecting symptom severity (0–4, mild; 5–15, moderate; 16–20, severe; >20, extremely severe) on admission. Regarding the selection of the matching parameters, older age and severe symptoms are associated with worse outcomes after IVT. The admission year was included as a matching parameter because acute stroke treatment has evolved during the cohort’s collection period (eg, thrombectomy has become the standard treatment in large vessel occlusions). The matching based on the aforementioned parameters was performed randomly. The local institutional research committee approved the study and waived the requirement for patient consent (HUS/203/2020 §14). The corresponding author had full access to all the data in the study and takes responsibility for its integrity and the data analysis. The study was conducted according to the Strengthening the Reporting of Observational studies in Epidemiology (STROBE) Statement. [SUBTITLE] Data Collection [SUBSECTION] The following variables were available from the Helsinki Stroke Thrombolysis Registry: date and time of the IVT, age, NIHSS score, systolic and diastolic blood pressure, weight on admission, and modified Rankin Scale (mRS)4 at 3 months after the IVT. Weight was based on the patients’ reports, except for the patients with aphasia, severe dysarthria, or lowered level of consciousness, for whom a treating physician estimated their weight. Blood tests on admission were available for plasma glucose and plasma lipids. We also retrospectively collected data from the hospital’s electronic records on the use of antithrombotic medication at the time of the stroke (antiplatelet or anticoagulant), history of smoking (never, ex-smoker, or current), and history of hypertension or diabetes (preexisting diagnosis of hypertension/diabetes or use of antihypertensive/antidiabetic medication at the time of the stroke). We analyzed all the post-IVT computed tomography and magnetic resonance images taken during hospitalization to identify hemorrhagic complications following IVT. All the patients underwent routine control imaging at least once ≈24 hours after IVT. The following variables were available from the Helsinki Stroke Thrombolysis Registry: date and time of the IVT, age, NIHSS score, systolic and diastolic blood pressure, weight on admission, and modified Rankin Scale (mRS)4 at 3 months after the IVT. Weight was based on the patients’ reports, except for the patients with aphasia, severe dysarthria, or lowered level of consciousness, for whom a treating physician estimated their weight. Blood tests on admission were available for plasma glucose and plasma lipids. We also retrospectively collected data from the hospital’s electronic records on the use of antithrombotic medication at the time of the stroke (antiplatelet or anticoagulant), history of smoking (never, ex-smoker, or current), and history of hypertension or diabetes (preexisting diagnosis of hypertension/diabetes or use of antihypertensive/antidiabetic medication at the time of the stroke). We analyzed all the post-IVT computed tomography and magnetic resonance images taken during hospitalization to identify hemorrhagic complications following IVT. All the patients underwent routine control imaging at least once ≈24 hours after IVT. [SUBTITLE] Outcomes [SUBSECTION] Our primary outcome was favorable (mRS score 0–2) versus unfavorable (mRS score 3–6) outcome at 3 months after IVT. The secondary outcomes were excellent (mRS score 0–1) versus non-excellent (mRS score 2–6) outcome at 3 months after IVT, mRS difference in shift analysis (described below), in-hospital symptomatic intracranial hemorrhage (ICH) following IVT, according to the European–Australian Cooperative Acute Stroke Study 2 criteria5 (an NIHSS score increase of ≥4 points and any ICH), and any in-hospital ICH following IVT. Our primary outcome was favorable (mRS score 0–2) versus unfavorable (mRS score 3–6) outcome at 3 months after IVT. The secondary outcomes were excellent (mRS score 0–1) versus non-excellent (mRS score 2–6) outcome at 3 months after IVT, mRS difference in shift analysis (described below), in-hospital symptomatic intracranial hemorrhage (ICH) following IVT, according to the European–Australian Cooperative Acute Stroke Study 2 criteria5 (an NIHSS score increase of ≥4 points and any ICH), and any in-hospital ICH following IVT. [SUBTITLE] Statistical Analyses [SUBSECTION] For the categorical variables, we report frequencies and proportions. For the continuous variables, we report means with SDs and medians with interquartile ranges. For the 1:1 matched case-control comparisons, we used the paired t test for the continuous variables and McNemar test for the categorical variables.6 To test whether the matched variables (age, sex, admission year, and NIHSS score at admission) were associated with an unfavorable outcome, we used a logistic regression model that included age, sex, admission year, and admission NIHSS score. Moreover, we used conditional logistic regression analysis to test if UIAs were associated with an unfavorable outcome. Systolic blood pressure, plasma glucose, and weight on admission have been associated with outcome after IVT in previous studies.7–11 Therefore, in addition to the univariate model, we tested a multivariate conditional regression model that included these variables as covariates. For an additional shift analysis, we conducted an ordinal logistic regression analysis using 3-month mRS as an ordinal variable. Both univariate and multivariate models were tested, and we used robust estimators of variance and clustering to account for the matched structure of the study population.12 Since the UIA patients were less likely to have excellent outcomes (see results) and they were more often current smokers, we conducted a post hoc conditional regression analysis to test whether smoking status was associated with outcome. There were few missing values, so we excluded subjects with missing values from the regression analyses and analyses comparing the groups. We report odds ratios (ORs) with 95% CIs for all regression analyses. A P<0.05 was considered statistically significant. We used Stata 17.0 (StataCorp, TX) for the statistical analyses. For the categorical variables, we report frequencies and proportions. For the continuous variables, we report means with SDs and medians with interquartile ranges. For the 1:1 matched case-control comparisons, we used the paired t test for the continuous variables and McNemar test for the categorical variables.6 To test whether the matched variables (age, sex, admission year, and NIHSS score at admission) were associated with an unfavorable outcome, we used a logistic regression model that included age, sex, admission year, and admission NIHSS score. Moreover, we used conditional logistic regression analysis to test if UIAs were associated with an unfavorable outcome. Systolic blood pressure, plasma glucose, and weight on admission have been associated with outcome after IVT in previous studies.7–11 Therefore, in addition to the univariate model, we tested a multivariate conditional regression model that included these variables as covariates. For an additional shift analysis, we conducted an ordinal logistic regression analysis using 3-month mRS as an ordinal variable. Both univariate and multivariate models were tested, and we used robust estimators of variance and clustering to account for the matched structure of the study population.12 Since the UIA patients were less likely to have excellent outcomes (see results) and they were more often current smokers, we conducted a post hoc conditional regression analysis to test whether smoking status was associated with outcome. There were few missing values, so we excluded subjects with missing values from the regression analyses and analyses comparing the groups. We report odds ratios (ORs) with 95% CIs for all regression analyses. A P<0.05 was considered statistically significant. We used Stata 17.0 (StataCorp, TX) for the statistical analyses.
Results
[SUBTITLE] Patient Characteristics [SUBSECTION] Of 3953 patients treated with IVT between 2005 and 2019, we identified 118 patients with saccular UIAs (Figure S1). As stated above, an additional 14 patients with fusiform UIAs were excluded from the study. Compared with the 3821 patients without UIAs, the UIA patients were more often female (42.8% versus 57.6%, respectively; P=0.001) but did not differ in age (68.0±13.8 years versus 69.7±12.0 years [mean±SD]; P=0.10) or NIHSS score (9.3±6.8 versus 9.5±8.2; P=0.76). Our final cohort consisted of 118 patients with saccular UIAs and 118 matched control patients without UIAs. The mean age of the cohort was 68.7±11.9 years, the mean NIHSS score was 9.5±7.8, and 144 (61%) were female. The mean admission year of the matched patients was slightly earlier (2013.0±4.2 and 2013.8±4.2; P<0.01). Otherwise, the UIA patients and matched patients did not differ in any matched variable (Table 1). Characteristics of the 118 Patients With Aneurysms and 118 Matched Patients Without Aneurysms Included in the Analysis The 118 UIA patients had a total of 137 saccular UIAs. The mean aneurysm diameter was 3.9±2.7 mm and 5 (4 %) were at least 10 mm in diameter. Eighteen (15%) UIA patients eventually underwent aneurysm treatment (surgical treatment in 7 patients and endovascular treatment in 11 patients), but none was treated during the 3-month follow-up period. Fourteen UIA patients died within 3 months after IVT. The cause of death was available for 13 of these patients, and none of them died due to a subarachnoid hemorrhage. Of 3953 patients treated with IVT between 2005 and 2019, we identified 118 patients with saccular UIAs (Figure S1). As stated above, an additional 14 patients with fusiform UIAs were excluded from the study. Compared with the 3821 patients without UIAs, the UIA patients were more often female (42.8% versus 57.6%, respectively; P=0.001) but did not differ in age (68.0±13.8 years versus 69.7±12.0 years [mean±SD]; P=0.10) or NIHSS score (9.3±6.8 versus 9.5±8.2; P=0.76). Our final cohort consisted of 118 patients with saccular UIAs and 118 matched control patients without UIAs. The mean age of the cohort was 68.7±11.9 years, the mean NIHSS score was 9.5±7.8, and 144 (61%) were female. The mean admission year of the matched patients was slightly earlier (2013.0±4.2 and 2013.8±4.2; P<0.01). Otherwise, the UIA patients and matched patients did not differ in any matched variable (Table 1). Characteristics of the 118 Patients With Aneurysms and 118 Matched Patients Without Aneurysms Included in the Analysis The 118 UIA patients had a total of 137 saccular UIAs. The mean aneurysm diameter was 3.9±2.7 mm and 5 (4 %) were at least 10 mm in diameter. Eighteen (15%) UIA patients eventually underwent aneurysm treatment (surgical treatment in 7 patients and endovascular treatment in 11 patients), but none was treated during the 3-month follow-up period. Fourteen UIA patients died within 3 months after IVT. The cause of death was available for 13 of these patients, and none of them died due to a subarachnoid hemorrhage. [SUBTITLE] Vascular Risk Factors and Outcome [SUBSECTION] Table 1 shows the characteristics of the 118 UIA patients and the 118 matched patients. The UIA patients were more often current smokers and had slightly higher systolic blood pressure on admission. Otherwise, the groups did not differ in their vascular risk factor profiles. Five (4%) of the matched patients and 8 (7%) of the UIA patients had symptomatic ICH following IVT (P=0.37). Likewise, 20 (17%) of the matched patients and 22 (19%) of the UIA patients had any ICH following IVT (P=0.74). Overall, 91 (39%) patients had unfavorable outcomes at 3 months. Older age, an earlier admission year, and a higher admission NIHSS score were associated with an unfavorable outcome (Table S1). The Figure shows the 3-month mRS values for the UIA and the matched patients. Sixty-eight (58%) and 77 (65%) of the UIA and matched patients, respectively, had a favorable outcome (mRS score 0–2) at 3 months. In the univariate conditional logistic regression analysis, the presence of a UIA was not associated with an unfavorable (mRS score 3–6) outcome (OR, 1.45 [95% CI, 0.82–2.56]; P=0.20; Table 2). Similarly, UIA status was not associated with an unfavorable outcome in the multivariate conditional logistic regression analysis (Table 2). However, the UIA patients were less likely to have an excellent outcome (mRS score 0–1) at 3 months (35% versus 49%), with an OR of 2.06 ([95% CI, 1.14–3.75]; P=0.02) for a non-excellent (mRS score 2–6) outcome (Table S2). Results of the Univariate and Multivariate Conditional Logistic Regression Analyses Three-month outcomes following intravenous thrombolysis for patients with unruptured intracranial aneurysms (UIA) and matched control patients. mRS indicates modified Rankin Scale. In the shift analysis, UIA status increased the odds of higher 3-months mRS in both univariate (OR, 1.56 [95% CI, 1.03–2.35]; P=0.04) and multivariate models (OR, 1.61 [95% CI, 1.03–2.49]; P=0.04). In a post-hoc analysis, the current smokers were not likelier to have unfavorable outcomes at 3 months compared with never smokers (OR, 1.33 [95% CI, 0.42–4.26]; P=0.63). However, the current smokers had suggestive evidence for higher odds of non-excellent outcomes at 3 months compared with never smokers (OR, 3.61 [95% CI, 0.97–13.38]; P=0.06). As the control patients had their strokes slightly earlier (by a few months) than the UIA patients, we also performed the analyses controlling for admission year, which did not affect the findings (results not shown). Table 1 shows the characteristics of the 118 UIA patients and the 118 matched patients. The UIA patients were more often current smokers and had slightly higher systolic blood pressure on admission. Otherwise, the groups did not differ in their vascular risk factor profiles. Five (4%) of the matched patients and 8 (7%) of the UIA patients had symptomatic ICH following IVT (P=0.37). Likewise, 20 (17%) of the matched patients and 22 (19%) of the UIA patients had any ICH following IVT (P=0.74). Overall, 91 (39%) patients had unfavorable outcomes at 3 months. Older age, an earlier admission year, and a higher admission NIHSS score were associated with an unfavorable outcome (Table S1). The Figure shows the 3-month mRS values for the UIA and the matched patients. Sixty-eight (58%) and 77 (65%) of the UIA and matched patients, respectively, had a favorable outcome (mRS score 0–2) at 3 months. In the univariate conditional logistic regression analysis, the presence of a UIA was not associated with an unfavorable (mRS score 3–6) outcome (OR, 1.45 [95% CI, 0.82–2.56]; P=0.20; Table 2). Similarly, UIA status was not associated with an unfavorable outcome in the multivariate conditional logistic regression analysis (Table 2). However, the UIA patients were less likely to have an excellent outcome (mRS score 0–1) at 3 months (35% versus 49%), with an OR of 2.06 ([95% CI, 1.14–3.75]; P=0.02) for a non-excellent (mRS score 2–6) outcome (Table S2). Results of the Univariate and Multivariate Conditional Logistic Regression Analyses Three-month outcomes following intravenous thrombolysis for patients with unruptured intracranial aneurysms (UIA) and matched control patients. mRS indicates modified Rankin Scale. In the shift analysis, UIA status increased the odds of higher 3-months mRS in both univariate (OR, 1.56 [95% CI, 1.03–2.35]; P=0.04) and multivariate models (OR, 1.61 [95% CI, 1.03–2.49]; P=0.04). In a post-hoc analysis, the current smokers were not likelier to have unfavorable outcomes at 3 months compared with never smokers (OR, 1.33 [95% CI, 0.42–4.26]; P=0.63). However, the current smokers had suggestive evidence for higher odds of non-excellent outcomes at 3 months compared with never smokers (OR, 3.61 [95% CI, 0.97–13.38]; P=0.06). As the control patients had their strokes slightly earlier (by a few months) than the UIA patients, we also performed the analyses controlling for admission year, which did not affect the findings (results not shown).
null
null
[ "Study Cohort", "Data Collection", "Outcomes", "Statistical Analyses", "Vascular Risk Factors and Outcome", "Article Information", "Sources of Funding", "Supplemental Material" ]
[ "We have previously described our cohort of UIA patients treated with IVT for ischemic stroke.1 We used the prospectively collected Helsinki Stroke Thrombolysis Registry2 to identify IVT-treated patients at Helsinki University Hospital between 2005 and 2019. We employed 2 overlapping methods to retrospectively identify patients with UIAs from the registry. First, we screened the radiology reports of all the patients in the registry for mentions of UIAs. The reports were limited to studies performed 1 day before or within 7 days of the IVT. Second, for all the patients in the thrombolysis registry, we searched the nationwide Finnish Care Register for Health Care for a UIA diagnosis made any time before or within 1 year after the IVT or for a diagnosis of subarachnoid hemorrhage dated within 1 year after the IVT. We then scrutinized the imaging studies of all patients identified through either method to confirm the presence of UIAs. A flowchart of the study is shown in Figure S1.\nOnly patients with saccular UIAs were included in the study, as we previously found that 6 of 14 (43%) patients with fusiform UIAs died due to aneurysm rupture between 1 day and 5 months after IVT.1\nIn the current study, each UIA patient was matched with a control patient without a UIA. The UIA patients and controls were matched on age (±3 years), sex, admission year (±1 year), and the National Institutes of Health Stroke Scale (NIHSS)3 category reflecting symptom severity (0–4, mild; 5–15, moderate; 16–20, severe; >20, extremely severe) on admission. Regarding the selection of the matching parameters, older age and severe symptoms are associated with worse outcomes after IVT. The admission year was included as a matching parameter because acute stroke treatment has evolved during the cohort’s collection period (eg, thrombectomy has become the standard treatment in large vessel occlusions). The matching based on the aforementioned parameters was performed randomly.\nThe local institutional research committee approved the study and waived the requirement for patient consent (HUS/203/2020 §14). The corresponding author had full access to all the data in the study and takes responsibility for its integrity and the data analysis. The study was conducted according to the Strengthening the Reporting of Observational studies in Epidemiology (STROBE) Statement.", "The following variables were available from the Helsinki Stroke Thrombolysis Registry: date and time of the IVT, age, NIHSS score, systolic and diastolic blood pressure, weight on admission, and modified Rankin Scale (mRS)4 at 3 months after the IVT. Weight was based on the patients’ reports, except for the patients with aphasia, severe dysarthria, or lowered level of consciousness, for whom a treating physician estimated their weight. Blood tests on admission were available for plasma glucose and plasma lipids.\nWe also retrospectively collected data from the hospital’s electronic records on the use of antithrombotic medication at the time of the stroke (antiplatelet or anticoagulant), history of smoking (never, ex-smoker, or current), and history of hypertension or diabetes (preexisting diagnosis of hypertension/diabetes or use of antihypertensive/antidiabetic medication at the time of the stroke).\nWe analyzed all the post-IVT computed tomography and magnetic resonance images taken during hospitalization to identify hemorrhagic complications following IVT. All the patients underwent routine control imaging at least once ≈24 hours after IVT.", "Our primary outcome was favorable (mRS score 0–2) versus unfavorable (mRS score 3–6) outcome at 3 months after IVT. The secondary outcomes were excellent (mRS score 0–1) versus non-excellent (mRS score 2–6) outcome at 3 months after IVT, mRS difference in shift analysis (described below), in-hospital symptomatic intracranial hemorrhage (ICH) following IVT, according to the European–Australian Cooperative Acute Stroke Study 2 criteria5 (an NIHSS score increase of ≥4 points and any ICH), and any in-hospital ICH following IVT.", "For the categorical variables, we report frequencies and proportions. For the continuous variables, we report means with SDs and medians with interquartile ranges.\nFor the 1:1 matched case-control comparisons, we used the paired t test for the continuous variables and McNemar test for the categorical variables.6 To test whether the matched variables (age, sex, admission year, and NIHSS score at admission) were associated with an unfavorable outcome, we used a logistic regression model that included age, sex, admission year, and admission NIHSS score. Moreover, we used conditional logistic regression analysis to test if UIAs were associated with an unfavorable outcome. Systolic blood pressure, plasma glucose, and weight on admission have been associated with outcome after IVT in previous studies.7–11 Therefore, in addition to the univariate model, we tested a multivariate conditional regression model that included these variables as covariates.\nFor an additional shift analysis, we conducted an ordinal logistic regression analysis using 3-month mRS as an ordinal variable. Both univariate and multivariate models were tested, and we used robust estimators of variance and clustering to account for the matched structure of the study population.12\nSince the UIA patients were less likely to have excellent outcomes (see results) and they were more often current smokers, we conducted a post hoc conditional regression analysis to test whether smoking status was associated with outcome.\nThere were few missing values, so we excluded subjects with missing values from the regression analyses and analyses comparing the groups. We report odds ratios (ORs) with 95% CIs for all regression analyses. A P<0.05 was considered statistically significant. We used Stata 17.0 (StataCorp, TX) for the statistical analyses.", "Table 1 shows the characteristics of the 118 UIA patients and the 118 matched patients. The UIA patients were more often current smokers and had slightly higher systolic blood pressure on admission. Otherwise, the groups did not differ in their vascular risk factor profiles. Five (4%) of the matched patients and 8 (7%) of the UIA patients had symptomatic ICH following IVT (P=0.37). Likewise, 20 (17%) of the matched patients and 22 (19%) of the UIA patients had any ICH following IVT (P=0.74).\nOverall, 91 (39%) patients had unfavorable outcomes at 3 months. Older age, an earlier admission year, and a higher admission NIHSS score were associated with an unfavorable outcome (Table S1).\nThe Figure shows the 3-month mRS values for the UIA and the matched patients. Sixty-eight (58%) and 77 (65%) of the UIA and matched patients, respectively, had a favorable outcome (mRS score 0–2) at 3 months. In the univariate conditional logistic regression analysis, the presence of a UIA was not associated with an unfavorable (mRS score 3–6) outcome (OR, 1.45 [95% CI, 0.82–2.56]; P=0.20; Table 2). Similarly, UIA status was not associated with an unfavorable outcome in the multivariate conditional logistic regression analysis (Table 2). However, the UIA patients were less likely to have an excellent outcome (mRS score 0–1) at 3 months (35% versus 49%), with an OR of 2.06 ([95% CI, 1.14–3.75]; P=0.02) for a non-excellent (mRS score 2–6) outcome (Table S2).\nResults of the Univariate and Multivariate Conditional Logistic Regression Analyses\nThree-month outcomes following intravenous thrombolysis for patients with unruptured intracranial aneurysms (UIA) and matched control patients. mRS indicates modified Rankin Scale.\nIn the shift analysis, UIA status increased the odds of higher 3-months mRS in both univariate (OR, 1.56 [95% CI, 1.03–2.35]; P=0.04) and multivariate models (OR, 1.61 [95% CI, 1.03–2.49]; P=0.04).\nIn a post-hoc analysis, the current smokers were not likelier to have unfavorable outcomes at 3 months compared with never smokers (OR, 1.33 [95% CI, 0.42–4.26]; P=0.63). However, the current smokers had suggestive evidence for higher odds of non-excellent outcomes at 3 months compared with never smokers (OR, 3.61 [95% CI, 0.97–13.38]; P=0.06).\nAs the control patients had their strokes slightly earlier (by a few months) than the UIA patients, we also performed the analyses controlling for admission year, which did not affect the findings (results not shown).", "[SUBTITLE] Sources of Funding [SUBSECTION] Funding was provided by Helsinki University Hospital, Y124930064. Dr Kaprio has received funding from the Academy of Finland.\nFunding was provided by Helsinki University Hospital, Y124930064. Dr Kaprio has received funding from the Academy of Finland.\n[SUBTITLE] Disclosures [SUBSECTION] Dr Kaprio has received funding from the Academy of Finland. Dr Putaala has taken part in stroke studies TASTE (Tenecteplase Versus Alteplase for Stroke Thrombolysis Evaluation Trial), TEMPO-2 (A Randomized Controlled Trial of TNK-tPA Versus Standard of Care for Minor Ischemic Stroke With Proven Occlusion), and TWIST (Tenecteplase in Wake-up Ischaemic Stroke Trial). The other authors report no conflicts.\nDr Kaprio has received funding from the Academy of Finland. Dr Putaala has taken part in stroke studies TASTE (Tenecteplase Versus Alteplase for Stroke Thrombolysis Evaluation Trial), TEMPO-2 (A Randomized Controlled Trial of TNK-tPA Versus Standard of Care for Minor Ischemic Stroke With Proven Occlusion), and TWIST (Tenecteplase in Wake-up Ischaemic Stroke Trial). The other authors report no conflicts.\n[SUBTITLE] Supplemental Material [SUBSECTION] Figure S1\nTables S1–S2\nFigure S1\nTables S1–S2", "Funding was provided by Helsinki University Hospital, Y124930064. Dr Kaprio has received funding from the Academy of Finland.", "Figure S1\nTables S1–S2" ]
[ null, null, null, null, null, null, null, null ]
[ "Methods", "Research Materials Transparency", "Study Cohort", "Data Collection", "Outcomes", "Statistical Analyses", "Results", "Patient Characteristics", "Vascular Risk Factors and Outcome", "Discussion", "Article Information", "Sources of Funding", "Disclosures", "Supplemental Material", "Supplementary Material" ]
[ "[SUBTITLE] Research Materials Transparency [SUBSECTION] Anonymized data that support the findings of this study are available from the corresponding author upon reasonable request.\nAnonymized data that support the findings of this study are available from the corresponding author upon reasonable request.\n[SUBTITLE] Study Cohort [SUBSECTION] We have previously described our cohort of UIA patients treated with IVT for ischemic stroke.1 We used the prospectively collected Helsinki Stroke Thrombolysis Registry2 to identify IVT-treated patients at Helsinki University Hospital between 2005 and 2019. We employed 2 overlapping methods to retrospectively identify patients with UIAs from the registry. First, we screened the radiology reports of all the patients in the registry for mentions of UIAs. The reports were limited to studies performed 1 day before or within 7 days of the IVT. Second, for all the patients in the thrombolysis registry, we searched the nationwide Finnish Care Register for Health Care for a UIA diagnosis made any time before or within 1 year after the IVT or for a diagnosis of subarachnoid hemorrhage dated within 1 year after the IVT. We then scrutinized the imaging studies of all patients identified through either method to confirm the presence of UIAs. A flowchart of the study is shown in Figure S1.\nOnly patients with saccular UIAs were included in the study, as we previously found that 6 of 14 (43%) patients with fusiform UIAs died due to aneurysm rupture between 1 day and 5 months after IVT.1\nIn the current study, each UIA patient was matched with a control patient without a UIA. The UIA patients and controls were matched on age (±3 years), sex, admission year (±1 year), and the National Institutes of Health Stroke Scale (NIHSS)3 category reflecting symptom severity (0–4, mild; 5–15, moderate; 16–20, severe; >20, extremely severe) on admission. Regarding the selection of the matching parameters, older age and severe symptoms are associated with worse outcomes after IVT. The admission year was included as a matching parameter because acute stroke treatment has evolved during the cohort’s collection period (eg, thrombectomy has become the standard treatment in large vessel occlusions). The matching based on the aforementioned parameters was performed randomly.\nThe local institutional research committee approved the study and waived the requirement for patient consent (HUS/203/2020 §14). The corresponding author had full access to all the data in the study and takes responsibility for its integrity and the data analysis. The study was conducted according to the Strengthening the Reporting of Observational studies in Epidemiology (STROBE) Statement.\nWe have previously described our cohort of UIA patients treated with IVT for ischemic stroke.1 We used the prospectively collected Helsinki Stroke Thrombolysis Registry2 to identify IVT-treated patients at Helsinki University Hospital between 2005 and 2019. We employed 2 overlapping methods to retrospectively identify patients with UIAs from the registry. First, we screened the radiology reports of all the patients in the registry for mentions of UIAs. The reports were limited to studies performed 1 day before or within 7 days of the IVT. Second, for all the patients in the thrombolysis registry, we searched the nationwide Finnish Care Register for Health Care for a UIA diagnosis made any time before or within 1 year after the IVT or for a diagnosis of subarachnoid hemorrhage dated within 1 year after the IVT. We then scrutinized the imaging studies of all patients identified through either method to confirm the presence of UIAs. A flowchart of the study is shown in Figure S1.\nOnly patients with saccular UIAs were included in the study, as we previously found that 6 of 14 (43%) patients with fusiform UIAs died due to aneurysm rupture between 1 day and 5 months after IVT.1\nIn the current study, each UIA patient was matched with a control patient without a UIA. The UIA patients and controls were matched on age (±3 years), sex, admission year (±1 year), and the National Institutes of Health Stroke Scale (NIHSS)3 category reflecting symptom severity (0–4, mild; 5–15, moderate; 16–20, severe; >20, extremely severe) on admission. Regarding the selection of the matching parameters, older age and severe symptoms are associated with worse outcomes after IVT. The admission year was included as a matching parameter because acute stroke treatment has evolved during the cohort’s collection period (eg, thrombectomy has become the standard treatment in large vessel occlusions). The matching based on the aforementioned parameters was performed randomly.\nThe local institutional research committee approved the study and waived the requirement for patient consent (HUS/203/2020 §14). The corresponding author had full access to all the data in the study and takes responsibility for its integrity and the data analysis. The study was conducted according to the Strengthening the Reporting of Observational studies in Epidemiology (STROBE) Statement.\n[SUBTITLE] Data Collection [SUBSECTION] The following variables were available from the Helsinki Stroke Thrombolysis Registry: date and time of the IVT, age, NIHSS score, systolic and diastolic blood pressure, weight on admission, and modified Rankin Scale (mRS)4 at 3 months after the IVT. Weight was based on the patients’ reports, except for the patients with aphasia, severe dysarthria, or lowered level of consciousness, for whom a treating physician estimated their weight. Blood tests on admission were available for plasma glucose and plasma lipids.\nWe also retrospectively collected data from the hospital’s electronic records on the use of antithrombotic medication at the time of the stroke (antiplatelet or anticoagulant), history of smoking (never, ex-smoker, or current), and history of hypertension or diabetes (preexisting diagnosis of hypertension/diabetes or use of antihypertensive/antidiabetic medication at the time of the stroke).\nWe analyzed all the post-IVT computed tomography and magnetic resonance images taken during hospitalization to identify hemorrhagic complications following IVT. All the patients underwent routine control imaging at least once ≈24 hours after IVT.\nThe following variables were available from the Helsinki Stroke Thrombolysis Registry: date and time of the IVT, age, NIHSS score, systolic and diastolic blood pressure, weight on admission, and modified Rankin Scale (mRS)4 at 3 months after the IVT. Weight was based on the patients’ reports, except for the patients with aphasia, severe dysarthria, or lowered level of consciousness, for whom a treating physician estimated their weight. Blood tests on admission were available for plasma glucose and plasma lipids.\nWe also retrospectively collected data from the hospital’s electronic records on the use of antithrombotic medication at the time of the stroke (antiplatelet or anticoagulant), history of smoking (never, ex-smoker, or current), and history of hypertension or diabetes (preexisting diagnosis of hypertension/diabetes or use of antihypertensive/antidiabetic medication at the time of the stroke).\nWe analyzed all the post-IVT computed tomography and magnetic resonance images taken during hospitalization to identify hemorrhagic complications following IVT. All the patients underwent routine control imaging at least once ≈24 hours after IVT.\n[SUBTITLE] Outcomes [SUBSECTION] Our primary outcome was favorable (mRS score 0–2) versus unfavorable (mRS score 3–6) outcome at 3 months after IVT. The secondary outcomes were excellent (mRS score 0–1) versus non-excellent (mRS score 2–6) outcome at 3 months after IVT, mRS difference in shift analysis (described below), in-hospital symptomatic intracranial hemorrhage (ICH) following IVT, according to the European–Australian Cooperative Acute Stroke Study 2 criteria5 (an NIHSS score increase of ≥4 points and any ICH), and any in-hospital ICH following IVT.\nOur primary outcome was favorable (mRS score 0–2) versus unfavorable (mRS score 3–6) outcome at 3 months after IVT. The secondary outcomes were excellent (mRS score 0–1) versus non-excellent (mRS score 2–6) outcome at 3 months after IVT, mRS difference in shift analysis (described below), in-hospital symptomatic intracranial hemorrhage (ICH) following IVT, according to the European–Australian Cooperative Acute Stroke Study 2 criteria5 (an NIHSS score increase of ≥4 points and any ICH), and any in-hospital ICH following IVT.\n[SUBTITLE] Statistical Analyses [SUBSECTION] For the categorical variables, we report frequencies and proportions. For the continuous variables, we report means with SDs and medians with interquartile ranges.\nFor the 1:1 matched case-control comparisons, we used the paired t test for the continuous variables and McNemar test for the categorical variables.6 To test whether the matched variables (age, sex, admission year, and NIHSS score at admission) were associated with an unfavorable outcome, we used a logistic regression model that included age, sex, admission year, and admission NIHSS score. Moreover, we used conditional logistic regression analysis to test if UIAs were associated with an unfavorable outcome. Systolic blood pressure, plasma glucose, and weight on admission have been associated with outcome after IVT in previous studies.7–11 Therefore, in addition to the univariate model, we tested a multivariate conditional regression model that included these variables as covariates.\nFor an additional shift analysis, we conducted an ordinal logistic regression analysis using 3-month mRS as an ordinal variable. Both univariate and multivariate models were tested, and we used robust estimators of variance and clustering to account for the matched structure of the study population.12\nSince the UIA patients were less likely to have excellent outcomes (see results) and they were more often current smokers, we conducted a post hoc conditional regression analysis to test whether smoking status was associated with outcome.\nThere were few missing values, so we excluded subjects with missing values from the regression analyses and analyses comparing the groups. We report odds ratios (ORs) with 95% CIs for all regression analyses. A P<0.05 was considered statistically significant. We used Stata 17.0 (StataCorp, TX) for the statistical analyses.\nFor the categorical variables, we report frequencies and proportions. For the continuous variables, we report means with SDs and medians with interquartile ranges.\nFor the 1:1 matched case-control comparisons, we used the paired t test for the continuous variables and McNemar test for the categorical variables.6 To test whether the matched variables (age, sex, admission year, and NIHSS score at admission) were associated with an unfavorable outcome, we used a logistic regression model that included age, sex, admission year, and admission NIHSS score. Moreover, we used conditional logistic regression analysis to test if UIAs were associated with an unfavorable outcome. Systolic blood pressure, plasma glucose, and weight on admission have been associated with outcome after IVT in previous studies.7–11 Therefore, in addition to the univariate model, we tested a multivariate conditional regression model that included these variables as covariates.\nFor an additional shift analysis, we conducted an ordinal logistic regression analysis using 3-month mRS as an ordinal variable. Both univariate and multivariate models were tested, and we used robust estimators of variance and clustering to account for the matched structure of the study population.12\nSince the UIA patients were less likely to have excellent outcomes (see results) and they were more often current smokers, we conducted a post hoc conditional regression analysis to test whether smoking status was associated with outcome.\nThere were few missing values, so we excluded subjects with missing values from the regression analyses and analyses comparing the groups. We report odds ratios (ORs) with 95% CIs for all regression analyses. A P<0.05 was considered statistically significant. We used Stata 17.0 (StataCorp, TX) for the statistical analyses.", "Anonymized data that support the findings of this study are available from the corresponding author upon reasonable request.", "We have previously described our cohort of UIA patients treated with IVT for ischemic stroke.1 We used the prospectively collected Helsinki Stroke Thrombolysis Registry2 to identify IVT-treated patients at Helsinki University Hospital between 2005 and 2019. We employed 2 overlapping methods to retrospectively identify patients with UIAs from the registry. First, we screened the radiology reports of all the patients in the registry for mentions of UIAs. The reports were limited to studies performed 1 day before or within 7 days of the IVT. Second, for all the patients in the thrombolysis registry, we searched the nationwide Finnish Care Register for Health Care for a UIA diagnosis made any time before or within 1 year after the IVT or for a diagnosis of subarachnoid hemorrhage dated within 1 year after the IVT. We then scrutinized the imaging studies of all patients identified through either method to confirm the presence of UIAs. A flowchart of the study is shown in Figure S1.\nOnly patients with saccular UIAs were included in the study, as we previously found that 6 of 14 (43%) patients with fusiform UIAs died due to aneurysm rupture between 1 day and 5 months after IVT.1\nIn the current study, each UIA patient was matched with a control patient without a UIA. The UIA patients and controls were matched on age (±3 years), sex, admission year (±1 year), and the National Institutes of Health Stroke Scale (NIHSS)3 category reflecting symptom severity (0–4, mild; 5–15, moderate; 16–20, severe; >20, extremely severe) on admission. Regarding the selection of the matching parameters, older age and severe symptoms are associated with worse outcomes after IVT. The admission year was included as a matching parameter because acute stroke treatment has evolved during the cohort’s collection period (eg, thrombectomy has become the standard treatment in large vessel occlusions). The matching based on the aforementioned parameters was performed randomly.\nThe local institutional research committee approved the study and waived the requirement for patient consent (HUS/203/2020 §14). The corresponding author had full access to all the data in the study and takes responsibility for its integrity and the data analysis. The study was conducted according to the Strengthening the Reporting of Observational studies in Epidemiology (STROBE) Statement.", "The following variables were available from the Helsinki Stroke Thrombolysis Registry: date and time of the IVT, age, NIHSS score, systolic and diastolic blood pressure, weight on admission, and modified Rankin Scale (mRS)4 at 3 months after the IVT. Weight was based on the patients’ reports, except for the patients with aphasia, severe dysarthria, or lowered level of consciousness, for whom a treating physician estimated their weight. Blood tests on admission were available for plasma glucose and plasma lipids.\nWe also retrospectively collected data from the hospital’s electronic records on the use of antithrombotic medication at the time of the stroke (antiplatelet or anticoagulant), history of smoking (never, ex-smoker, or current), and history of hypertension or diabetes (preexisting diagnosis of hypertension/diabetes or use of antihypertensive/antidiabetic medication at the time of the stroke).\nWe analyzed all the post-IVT computed tomography and magnetic resonance images taken during hospitalization to identify hemorrhagic complications following IVT. All the patients underwent routine control imaging at least once ≈24 hours after IVT.", "Our primary outcome was favorable (mRS score 0–2) versus unfavorable (mRS score 3–6) outcome at 3 months after IVT. The secondary outcomes were excellent (mRS score 0–1) versus non-excellent (mRS score 2–6) outcome at 3 months after IVT, mRS difference in shift analysis (described below), in-hospital symptomatic intracranial hemorrhage (ICH) following IVT, according to the European–Australian Cooperative Acute Stroke Study 2 criteria5 (an NIHSS score increase of ≥4 points and any ICH), and any in-hospital ICH following IVT.", "For the categorical variables, we report frequencies and proportions. For the continuous variables, we report means with SDs and medians with interquartile ranges.\nFor the 1:1 matched case-control comparisons, we used the paired t test for the continuous variables and McNemar test for the categorical variables.6 To test whether the matched variables (age, sex, admission year, and NIHSS score at admission) were associated with an unfavorable outcome, we used a logistic regression model that included age, sex, admission year, and admission NIHSS score. Moreover, we used conditional logistic regression analysis to test if UIAs were associated with an unfavorable outcome. Systolic blood pressure, plasma glucose, and weight on admission have been associated with outcome after IVT in previous studies.7–11 Therefore, in addition to the univariate model, we tested a multivariate conditional regression model that included these variables as covariates.\nFor an additional shift analysis, we conducted an ordinal logistic regression analysis using 3-month mRS as an ordinal variable. Both univariate and multivariate models were tested, and we used robust estimators of variance and clustering to account for the matched structure of the study population.12\nSince the UIA patients were less likely to have excellent outcomes (see results) and they were more often current smokers, we conducted a post hoc conditional regression analysis to test whether smoking status was associated with outcome.\nThere were few missing values, so we excluded subjects with missing values from the regression analyses and analyses comparing the groups. We report odds ratios (ORs) with 95% CIs for all regression analyses. A P<0.05 was considered statistically significant. We used Stata 17.0 (StataCorp, TX) for the statistical analyses.", "[SUBTITLE] Patient Characteristics [SUBSECTION] Of 3953 patients treated with IVT between 2005 and 2019, we identified 118 patients with saccular UIAs (Figure S1). As stated above, an additional 14 patients with fusiform UIAs were excluded from the study. Compared with the 3821 patients without UIAs, the UIA patients were more often female (42.8% versus 57.6%, respectively; P=0.001) but did not differ in age (68.0±13.8 years versus 69.7±12.0 years [mean±SD]; P=0.10) or NIHSS score (9.3±6.8 versus 9.5±8.2; P=0.76).\nOur final cohort consisted of 118 patients with saccular UIAs and 118 matched control patients without UIAs. The mean age of the cohort was 68.7±11.9 years, the mean NIHSS score was 9.5±7.8, and 144 (61%) were female. The mean admission year of the matched patients was slightly earlier (2013.0±4.2 and 2013.8±4.2; P<0.01). Otherwise, the UIA patients and matched patients did not differ in any matched variable (Table 1).\nCharacteristics of the 118 Patients With Aneurysms and 118 Matched Patients Without Aneurysms Included in the Analysis\nThe 118 UIA patients had a total of 137 saccular UIAs. The mean aneurysm diameter was 3.9±2.7 mm and 5 (4 %) were at least 10 mm in diameter. Eighteen (15%) UIA patients eventually underwent aneurysm treatment (surgical treatment in 7 patients and endovascular treatment in 11 patients), but none was treated during the 3-month follow-up period. Fourteen UIA patients died within 3 months after IVT. The cause of death was available for 13 of these patients, and none of them died due to a subarachnoid hemorrhage.\nOf 3953 patients treated with IVT between 2005 and 2019, we identified 118 patients with saccular UIAs (Figure S1). As stated above, an additional 14 patients with fusiform UIAs were excluded from the study. Compared with the 3821 patients without UIAs, the UIA patients were more often female (42.8% versus 57.6%, respectively; P=0.001) but did not differ in age (68.0±13.8 years versus 69.7±12.0 years [mean±SD]; P=0.10) or NIHSS score (9.3±6.8 versus 9.5±8.2; P=0.76).\nOur final cohort consisted of 118 patients with saccular UIAs and 118 matched control patients without UIAs. The mean age of the cohort was 68.7±11.9 years, the mean NIHSS score was 9.5±7.8, and 144 (61%) were female. The mean admission year of the matched patients was slightly earlier (2013.0±4.2 and 2013.8±4.2; P<0.01). Otherwise, the UIA patients and matched patients did not differ in any matched variable (Table 1).\nCharacteristics of the 118 Patients With Aneurysms and 118 Matched Patients Without Aneurysms Included in the Analysis\nThe 118 UIA patients had a total of 137 saccular UIAs. The mean aneurysm diameter was 3.9±2.7 mm and 5 (4 %) were at least 10 mm in diameter. Eighteen (15%) UIA patients eventually underwent aneurysm treatment (surgical treatment in 7 patients and endovascular treatment in 11 patients), but none was treated during the 3-month follow-up period. Fourteen UIA patients died within 3 months after IVT. The cause of death was available for 13 of these patients, and none of them died due to a subarachnoid hemorrhage.\n[SUBTITLE] Vascular Risk Factors and Outcome [SUBSECTION] Table 1 shows the characteristics of the 118 UIA patients and the 118 matched patients. The UIA patients were more often current smokers and had slightly higher systolic blood pressure on admission. Otherwise, the groups did not differ in their vascular risk factor profiles. Five (4%) of the matched patients and 8 (7%) of the UIA patients had symptomatic ICH following IVT (P=0.37). Likewise, 20 (17%) of the matched patients and 22 (19%) of the UIA patients had any ICH following IVT (P=0.74).\nOverall, 91 (39%) patients had unfavorable outcomes at 3 months. Older age, an earlier admission year, and a higher admission NIHSS score were associated with an unfavorable outcome (Table S1).\nThe Figure shows the 3-month mRS values for the UIA and the matched patients. Sixty-eight (58%) and 77 (65%) of the UIA and matched patients, respectively, had a favorable outcome (mRS score 0–2) at 3 months. In the univariate conditional logistic regression analysis, the presence of a UIA was not associated with an unfavorable (mRS score 3–6) outcome (OR, 1.45 [95% CI, 0.82–2.56]; P=0.20; Table 2). Similarly, UIA status was not associated with an unfavorable outcome in the multivariate conditional logistic regression analysis (Table 2). However, the UIA patients were less likely to have an excellent outcome (mRS score 0–1) at 3 months (35% versus 49%), with an OR of 2.06 ([95% CI, 1.14–3.75]; P=0.02) for a non-excellent (mRS score 2–6) outcome (Table S2).\nResults of the Univariate and Multivariate Conditional Logistic Regression Analyses\nThree-month outcomes following intravenous thrombolysis for patients with unruptured intracranial aneurysms (UIA) and matched control patients. mRS indicates modified Rankin Scale.\nIn the shift analysis, UIA status increased the odds of higher 3-months mRS in both univariate (OR, 1.56 [95% CI, 1.03–2.35]; P=0.04) and multivariate models (OR, 1.61 [95% CI, 1.03–2.49]; P=0.04).\nIn a post-hoc analysis, the current smokers were not likelier to have unfavorable outcomes at 3 months compared with never smokers (OR, 1.33 [95% CI, 0.42–4.26]; P=0.63). However, the current smokers had suggestive evidence for higher odds of non-excellent outcomes at 3 months compared with never smokers (OR, 3.61 [95% CI, 0.97–13.38]; P=0.06).\nAs the control patients had their strokes slightly earlier (by a few months) than the UIA patients, we also performed the analyses controlling for admission year, which did not affect the findings (results not shown).\nTable 1 shows the characteristics of the 118 UIA patients and the 118 matched patients. The UIA patients were more often current smokers and had slightly higher systolic blood pressure on admission. Otherwise, the groups did not differ in their vascular risk factor profiles. Five (4%) of the matched patients and 8 (7%) of the UIA patients had symptomatic ICH following IVT (P=0.37). Likewise, 20 (17%) of the matched patients and 22 (19%) of the UIA patients had any ICH following IVT (P=0.74).\nOverall, 91 (39%) patients had unfavorable outcomes at 3 months. Older age, an earlier admission year, and a higher admission NIHSS score were associated with an unfavorable outcome (Table S1).\nThe Figure shows the 3-month mRS values for the UIA and the matched patients. Sixty-eight (58%) and 77 (65%) of the UIA and matched patients, respectively, had a favorable outcome (mRS score 0–2) at 3 months. In the univariate conditional logistic regression analysis, the presence of a UIA was not associated with an unfavorable (mRS score 3–6) outcome (OR, 1.45 [95% CI, 0.82–2.56]; P=0.20; Table 2). Similarly, UIA status was not associated with an unfavorable outcome in the multivariate conditional logistic regression analysis (Table 2). However, the UIA patients were less likely to have an excellent outcome (mRS score 0–1) at 3 months (35% versus 49%), with an OR of 2.06 ([95% CI, 1.14–3.75]; P=0.02) for a non-excellent (mRS score 2–6) outcome (Table S2).\nResults of the Univariate and Multivariate Conditional Logistic Regression Analyses\nThree-month outcomes following intravenous thrombolysis for patients with unruptured intracranial aneurysms (UIA) and matched control patients. mRS indicates modified Rankin Scale.\nIn the shift analysis, UIA status increased the odds of higher 3-months mRS in both univariate (OR, 1.56 [95% CI, 1.03–2.35]; P=0.04) and multivariate models (OR, 1.61 [95% CI, 1.03–2.49]; P=0.04).\nIn a post-hoc analysis, the current smokers were not likelier to have unfavorable outcomes at 3 months compared with never smokers (OR, 1.33 [95% CI, 0.42–4.26]; P=0.63). However, the current smokers had suggestive evidence for higher odds of non-excellent outcomes at 3 months compared with never smokers (OR, 3.61 [95% CI, 0.97–13.38]; P=0.06).\nAs the control patients had their strokes slightly earlier (by a few months) than the UIA patients, we also performed the analyses controlling for admission year, which did not affect the findings (results not shown).", "Of 3953 patients treated with IVT between 2005 and 2019, we identified 118 patients with saccular UIAs (Figure S1). As stated above, an additional 14 patients with fusiform UIAs were excluded from the study. Compared with the 3821 patients without UIAs, the UIA patients were more often female (42.8% versus 57.6%, respectively; P=0.001) but did not differ in age (68.0±13.8 years versus 69.7±12.0 years [mean±SD]; P=0.10) or NIHSS score (9.3±6.8 versus 9.5±8.2; P=0.76).\nOur final cohort consisted of 118 patients with saccular UIAs and 118 matched control patients without UIAs. The mean age of the cohort was 68.7±11.9 years, the mean NIHSS score was 9.5±7.8, and 144 (61%) were female. The mean admission year of the matched patients was slightly earlier (2013.0±4.2 and 2013.8±4.2; P<0.01). Otherwise, the UIA patients and matched patients did not differ in any matched variable (Table 1).\nCharacteristics of the 118 Patients With Aneurysms and 118 Matched Patients Without Aneurysms Included in the Analysis\nThe 118 UIA patients had a total of 137 saccular UIAs. The mean aneurysm diameter was 3.9±2.7 mm and 5 (4 %) were at least 10 mm in diameter. Eighteen (15%) UIA patients eventually underwent aneurysm treatment (surgical treatment in 7 patients and endovascular treatment in 11 patients), but none was treated during the 3-month follow-up period. Fourteen UIA patients died within 3 months after IVT. The cause of death was available for 13 of these patients, and none of them died due to a subarachnoid hemorrhage.", "Table 1 shows the characteristics of the 118 UIA patients and the 118 matched patients. The UIA patients were more often current smokers and had slightly higher systolic blood pressure on admission. Otherwise, the groups did not differ in their vascular risk factor profiles. Five (4%) of the matched patients and 8 (7%) of the UIA patients had symptomatic ICH following IVT (P=0.37). Likewise, 20 (17%) of the matched patients and 22 (19%) of the UIA patients had any ICH following IVT (P=0.74).\nOverall, 91 (39%) patients had unfavorable outcomes at 3 months. Older age, an earlier admission year, and a higher admission NIHSS score were associated with an unfavorable outcome (Table S1).\nThe Figure shows the 3-month mRS values for the UIA and the matched patients. Sixty-eight (58%) and 77 (65%) of the UIA and matched patients, respectively, had a favorable outcome (mRS score 0–2) at 3 months. In the univariate conditional logistic regression analysis, the presence of a UIA was not associated with an unfavorable (mRS score 3–6) outcome (OR, 1.45 [95% CI, 0.82–2.56]; P=0.20; Table 2). Similarly, UIA status was not associated with an unfavorable outcome in the multivariate conditional logistic regression analysis (Table 2). However, the UIA patients were less likely to have an excellent outcome (mRS score 0–1) at 3 months (35% versus 49%), with an OR of 2.06 ([95% CI, 1.14–3.75]; P=0.02) for a non-excellent (mRS score 2–6) outcome (Table S2).\nResults of the Univariate and Multivariate Conditional Logistic Regression Analyses\nThree-month outcomes following intravenous thrombolysis for patients with unruptured intracranial aneurysms (UIA) and matched control patients. mRS indicates modified Rankin Scale.\nIn the shift analysis, UIA status increased the odds of higher 3-months mRS in both univariate (OR, 1.56 [95% CI, 1.03–2.35]; P=0.04) and multivariate models (OR, 1.61 [95% CI, 1.03–2.49]; P=0.04).\nIn a post-hoc analysis, the current smokers were not likelier to have unfavorable outcomes at 3 months compared with never smokers (OR, 1.33 [95% CI, 0.42–4.26]; P=0.63). However, the current smokers had suggestive evidence for higher odds of non-excellent outcomes at 3 months compared with never smokers (OR, 3.61 [95% CI, 0.97–13.38]; P=0.06).\nAs the control patients had their strokes slightly earlier (by a few months) than the UIA patients, we also performed the analyses controlling for admission year, which did not affect the findings (results not shown).", "Although the patients with ischemic stroke with and without UIAs in this study were as likely to have favorable (mRS score 0–2) outcomes at 3 months, the patients with UIAs did not achieve excellent outcomes (mRS score 0–1) as often. UIA patients also had an increased odds of higher mRS in shift analysis. The IVT-treated patients with stroke with saccular UIAs did not differ significantly in their cardiovascular risk factor profile, including in terms of diabetes, from the patients without UIAs matched for age, sex, and stroke severity. However, they were more often current smokers, and their admission systolic blood pressure was slightly higher. We found no differences in hemorrhagic complications following IVT in the patients with and without UIAs.\nOur finding that smoking and hypertension are more common in UIA patients—even among patients with ischemic stroke, who are considered to have more cardiovascular risk factors compared with the general population—is in accordance with the known fact that these 2 modifiable risk factors contribute strongly to UIA formation, as, for example, a recent Mendelian randomization study has suggested.13 Few studies have compared the outcomes14–18 and/or characteristics15–20 of patients with stroke with and without UIAs. Most studies have included a small number of UIA patients and, hence, have been limited to unadjusted analyses. These studies mostly found no differences in sex, age, or vascular risk factors between patients with and without UIAs; however, in some studies, UIA patients were older,15,16 more often female,15,16 or likelier to have hypertension.19 Studies analyzing IVT complications15,17,18,21,22 or outcomes14,15,17,19 have not found any differences between patients with and without UIAs. The only study that was large enough to conduct multivariate analyses included 95 patients with minor stroke or transient ischemic attack and UIAs and found that the patients with UIAs were more often female, smokers, and likelier to have hypertension than the patients without UIAs.20 These results are in accordance with our results.\nTo the best of our knowledge, our study is the first to report that IVT-treated stroke patients with UIAs were less likely to have excellent outcomes after their stroke than patients without UIAs. UIA patients are more often hypertensive and smokers, both of which are associated with reduced white matter integrity.23,24 It is plausible that UIA patients therefore have reduced cerebral plasticity, rendering them less likely to make an excellent recovery.\nOur study included a large cohort of patients with ischemic stroke with UIAs, and we conducted a matched comparison of patients with stroke with and without UIAs. The strengths of our study also include its prospective nature and that we were able to reliably record post-IVT complications, as all patients had at least 1 follow-up brain imaging scan. The major limitations of our study are that smoking status, diabetes, and hypertension were retrospectively recorded based on medical records. Moreover, weight was based on self-reports or clinical estimates. Our results are applicable only to stroke patients eligible for IVT, as the cohort did not represent all patients with ischemic stroke. We included patients who were treated with IVT but did not use any other criteria for stroke (eg, symptom duration or post-IVT imaging studies). Hence, it is possible that our cohort included a few stroke mimic patients, but this number is presumably rather small, and the proportion of such patients should not differ significantly between the 2 groups (UIA patients and matched controls). As we limited the imaging studies to the day before and 7 days post-IVT, it is possible that we excluded a small number of patients who were admitted and imaged due to transient ischemic attack a few days before IVT. We did not have data on prestroke functional status (eg, mRS scale) and, therefore, we could not assess changes in the patients’ functional status following stroke.\nThe findings of this study suggest that even among patients with ischemic stroke, smoking and hypertension are more common in patients with UIAs. In contrast, this association was not seen for diabetes or hypercholesterolemia. Also, IVT-treated ischemic stroke patients with UIAs may reach excellent outcomes less frequently than matched patients without UIAs. This finding needs to be confirmed in further studies.", "[SUBTITLE] Sources of Funding [SUBSECTION] Funding was provided by Helsinki University Hospital, Y124930064. Dr Kaprio has received funding from the Academy of Finland.\nFunding was provided by Helsinki University Hospital, Y124930064. Dr Kaprio has received funding from the Academy of Finland.\n[SUBTITLE] Disclosures [SUBSECTION] Dr Kaprio has received funding from the Academy of Finland. Dr Putaala has taken part in stroke studies TASTE (Tenecteplase Versus Alteplase for Stroke Thrombolysis Evaluation Trial), TEMPO-2 (A Randomized Controlled Trial of TNK-tPA Versus Standard of Care for Minor Ischemic Stroke With Proven Occlusion), and TWIST (Tenecteplase in Wake-up Ischaemic Stroke Trial). The other authors report no conflicts.\nDr Kaprio has received funding from the Academy of Finland. Dr Putaala has taken part in stroke studies TASTE (Tenecteplase Versus Alteplase for Stroke Thrombolysis Evaluation Trial), TEMPO-2 (A Randomized Controlled Trial of TNK-tPA Versus Standard of Care for Minor Ischemic Stroke With Proven Occlusion), and TWIST (Tenecteplase in Wake-up Ischaemic Stroke Trial). The other authors report no conflicts.\n[SUBTITLE] Supplemental Material [SUBSECTION] Figure S1\nTables S1–S2\nFigure S1\nTables S1–S2", "Funding was provided by Helsinki University Hospital, Y124930064. Dr Kaprio has received funding from the Academy of Finland.", "Dr Kaprio has received funding from the Academy of Finland. Dr Putaala has taken part in stroke studies TASTE (Tenecteplase Versus Alteplase for Stroke Thrombolysis Evaluation Trial), TEMPO-2 (A Randomized Controlled Trial of TNK-tPA Versus Standard of Care for Minor Ischemic Stroke With Proven Occlusion), and TWIST (Tenecteplase in Wake-up Ischaemic Stroke Trial). The other authors report no conflicts.", "Figure S1\nTables S1–S2", "" ]
[ "methods", "materials", null, null, null, null, "results", "subjects", null, "discussion", null, null, "COI-Statement", null, "supplementary-material" ]
[ "cardiovascular disease", "hospital", "intracranial aneurysm", "ischemic stroke", "risk factors" ]
Multisystem inflammatory syndrome associated with SARS-CoV-2 infection in children: update and new insights from the second report of an Iranian referral hospital.
36254726
Here, we are sharing our second report about children affected by Multisystem Inflammatory Syndrome in Children (MIS-C). The aim of the present study was to update our knowledge about children with MIS-C. Furthermore, we tried to compare clinical manifestations, laboratory features and final outcome of patients based on disease severity, in order to better understanding of the nature of this novel syndrome.
INTRODUCTION
This retrospective study was conducted at Children's Medical Center Hospital, the hub of excellence in paediatrics in Iran, located in Tehran, Iran. We reviewed medical records of children admitted to the hospital with the diagnosis of MIS-C from July 2020 to October 2021.
METHODS
One hundred and twenty-two patients enrolled the study. Ninety-seven (79.5%) patients had mild to moderate MIS-C (MIS-C without overlap with KD (n = 80); MIS-C overlapping with KD (n = 17)) and 25 (20.5%) patients showed severe MIS-C. The mean age of all patients was 6.4 ± 4.0 years. Nausea and vomiting (53.3%), skin rash (49.6%), abdominal pain (46.7%) and conjunctivitis (41.8%) were also frequently seen Headache, chest pain, tachypnea and respiratory distress were significantly more common in patients with severe MIS-C (P < 0.0001, P = 0.021, P < 0.0001 and P < 0.0001, respectively). Positive anti-N severe acute respiratory syndrome coronavirus 2 IgM and IgG were detected in 14 (33.3%) and 23 (46.9%) tested patients, respectively. Albumin, and vitamin D levels in children with severe MISC were significantly lower than children with mild to moderate MIS-C (P < 0.0001, P = 0.05). Unfortunately, 2 (1.6%) of 122 patients died and both had severe MIS-C.
RESULTS
Patients with MIS-C in our region suffer from wide range of signs and symptoms. Among laboratory parameters, hypoalbuminemia and low vitamin D levels may predict a more severe course of the disease. Coronary artery dilation is frequently seen among all patients, regardless of disease severity.
CONCLUSION
[ "Humans", "Child", "Child, Preschool", "COVID-19", "Iran", "SARS-CoV-2", "Retrospective Studies", "Hospitals", "Referral and Consultation", "Vitamin D" ]
9671882
null
null
null
null
Results
One hundred and twenty-two patients enrolled the study. Ninety-seven (79.5%) patients had mild to moderate MIS-C (MIS-C without overlap with KD (n = 80); MIS-C overlapping with KD (n = 17)) and 25 (20.5%) patients showed severe MIS-C. Forty-eight (39.3%) patients were female and 74 (60.7%) patients were male. There was no significant difference in sex distribution between three groups (P = 0.837). The mean age of all patients was 6.4 ± 4.0 years. The mean age of patients with severe MIS-C (8.2 ± 4.6 years) was significantly higher than MIS-C group without overlap with KD (5.7 ± 3.7 years) and MIS-C group overlapping with KD (6.75 ± 4.4 years), respectively (P = 0.029). Fifteen (12.3%) patients had underlying disease; 11 (11.3%) patients with mild and moderate MIS-C and 4 (16%) patients with severe MIS-C. This was not a noticeable difference (P = 0.507). Totally, 74 patients (60.7%) had the history of close contact with an individual with illness clinically compatible with COVID-19 disease who had close contact with an individual with laboratory-confirmed SARS-CoV-2 infection; however, there was no significant difference between two groups (P = 0.172). This was found in 62 cases with mild and moderate MIS-C (64%). We also studied serologic tests as a footprint of COVID-19 in some of our patients as indicated. Positive anti-N SARS-CoV-2 IgM and IgG were detected in 14 (33.3%) and 23 (46.9%) tested patients, respectively. There was no significant difference between mild to moderate and severe disease (P = 1.000 and P = 0.734 respectively). We also obtained a nasopharyngeal SARS-CoV-2 RT-PCR test for all of the patients: 36.9% of samples were positive; 37.1% in patients with mild and moderate disease and 36.9% in patients with severe disease (P = 1.000). The most common clinical manifestation at the presentation was fever which was documented in all patients. Nausea and vomiting (53.3%), skin rash (49.6%), abdominal pain (46.7%) and conjunctivitis (41.8%) were also frequently seen. We also compared clinical manifestations between three groups (MIS-C overlapping with KD, MIS-C without overlap with KD and severe MIS-C). Headache, chest pain, tachypnea and respiratory distress were significantly more common in patients with severe MIS-C (P < 0.0001, P = 0.021, P < 0.0001 and P < 0.0001, respectively). Table 2 shows the clinical manifestations in detail. Table 2.Clinical characteristics of patients with MIS-C in different groups at presentationVariables N (%)Disease severityTotal (n = 122)MIS-C overlapping with KD (n = 17)MIS-C without overlap with KD (n = 80)Severe MIS-C (n = 25)P valueConstitutional symptomsHeadache23 (18.9)1 (5.9)10 (12.8)12 (48.0)0.000Myalgia24 (20.3)3 (17.6)18 (23.4)3 (12.5)0.49DermatologicConjunctivitis51 (41.8)7 (41.2)34 (42.5)10 (40.0)0.97Skin rash59 (49.6)9 (52.9)40 (51.3)10 (41.7)0.61Mucus membrane involvement18 (15.1)3 (17.6)11 (14.1)4 (16.7)0.9RespiratoryRhinorrhea3 (2.5)1 (5.9)2 (2.6)0 (0)0.49Cough35 (29.2)4 (23.5)21 (26.9)10 (40)0.39Chest pain4 (3.3)0 (0)1 (1.0)3 (12.0)0.021Tachypnea28 (23.0)0 (0)14 (14.4)14 (56.0)0.000Respiratory distress25 (20.8)0 (0)11 (14.1)14 (56.0)0.000GastrointestinalHepatomegaly7 (6.2)0 (0)5 (6.7)2 (9.1)0.496Splenomegaly9 (8.0)0 (0)7 (9.3)2 (9.1)0.446Ascites11 (9.6)3 (18.8)6 (8)2 (8.7)0.41Abdominal pain56 (46.7)9 (52.9)35 (44.3)12 (50)0.76Nausea and vomiting65 (53.3)5 (29.4)45 (56.3)15 (60.0)0.082Diarrhoea44 (36.7)5 (29.4)33 (41.3)6 (26.1)0.33Ileocolitis5 (4.3)0 (0)4 (5.3)1 (4.3)0.64Colitis2 (1.8)0 (0)2 (2.7)0 (0)0.6Lymphadenopathy6 (5)1 (5.9)3 (3.8)2 (8.0)0.688Sore throat7 (5.8)0 (0)4 (5.1)3 (12.0)0.236Edema25 (21.0)6 (35.3)14 (19.7)5 (20.8)0.282 Clinical characteristics of patients with MIS-C in different groups at presentation We also studied some laboratory parameters in our patients including complete blood count (CBC), biochemistry, liver tests (aspartate transaminase (AST) and alanine transaminase (ALT)), prothrombin time (PT), thromboplastin time (PTT), albumin, inflammatory markers (erythrocyte sedimentation rate (ESR), C-reactive protein (CRP)), Ferritin, IL-6, Fibrinogen, D-dimer, cardiac and other enzymes (Creatine phosphokinase (CPK), CPK-MB, troponin, Lactate dehydrogenase (LDH), amylase, lipase), triglyceride and vitamin D level. We found that vitamin D levels in children with severe MISC were significantly lower than children with mild to moderate MIS-C (P = 0.05). Table 3 shows laboratory information in details. Table 3.Laboratory findings of the patients with mild to moderate and severe MIS-CLaboratory characteristics Median (IQR 25–75)Disease severityMild and moderate MIS-CSevere MIS-CP valueCBC WBC ( × 10⁹ cells per L)8.350 (6.198–12.178)10.11 (7.255–15.780)0.163Neutrophils ( × 10⁹ cells per L)5.700 (3.190–8.035)8.230 (4.625–12.25)0.370Lymphocyte ( × 10⁹ cells per L)1.520 (0.985–3.105)1.400 (0.665–1.920)0.178Hemoglobin (g/dl)11.4 (10.4–12.6)11.6 (10.6–12.8)0.645Platelet ( × 10⁹ cells per L)235 (129–311)182 (113–247)0.178BiochemistryBUN (mg/dl)11 (8–15)15 (11–19)0.002Creatinine (mg/dl)0.6 (0.5–0.7)0.7 (0.5–0.8)0.160Calcium (mg/dl)8.6 (8.1–9.2)8.4 (7.7–8.9)0.816Phosphorus (mg/dl)3.9 (3.0–4.7)3.7 (2.9–4.3)1.000Magnesium (mg/dl)2.1 (1.9–2.3)2.0 (1.7–2.2)0.397Sodium (mEq/l)134 (130–138)134 (130–1137)0.889Potassium (mEq/l)3.9 (3.6–4.3)3.9 (3.6–4.3)0.831Liver testsAST (U/L)31 (22–59)41 (26–101)0.233ALT (U/L)24 (14–51)27 (16–74)0.711PT (sec)14 (13–16)15 (13–16)1.000PTT (sec)36 (32–40)34 (31–39)0.722Inflammatory markersAlbumin (g/dl)4.1 (3.6–4.4)3.3 (2.9–3.6)<0.0001ESR (mm/h)27 (12–49)30 (15–53)0.493CRP (mg/l)28 (6–54)57 (24–119)0.346Ferritin (ng/ml)193 (95–441)457 (170–1462)0.428Interleukin-6 (pg/ml)76 (11–184)3 (0- 3.3)0.167Fibrinogen (mg/dl)377 (255–508)417 (254–541)0.379D-dimer (ng/ml)2.8 (0.8–316)4.2 (1.0–420.2)0.765EnzymesCPK (U/l)55 (36–105)72 (31–337)0.458CPK-MB (μg/l)16 (10–20)20 (11–37)0.778Troponin0.1 (0.09–0.1)0.1 (0.1–4.2)0.294LDH (U/l)537 (436–670)558 (416–855)0.780Amylase (U/l)65 (28–232)43 (6–117)1.000Lipase (U/l)32 (20–266)13 (3–25)0.598OthersVitamin D (ng/ml)20.5 (8.2–33.7)8.5 (3.0–14.2)0.05Triglyceride (mg/dl)192 (132–283)92 (142–142)1.000WBC, White blood cell count; ALT, Alanine aminotransferase; AST, Aspartate aminotransferase; CRP, C-reactive protein; ESR, Erythrocyte sedimentation rate; PT, Prothrombin time; PTT, Partial thromboplastin time; CPK, Creatine phosphokinase; LDH, Lactate dehydrogenase. Laboratory findings of the patients with mild to moderate and severe MIS-C WBC, White blood cell count; ALT, Alanine aminotransferase; AST, Aspartate aminotransferase; CRP, C-reactive protein; ESR, Erythrocyte sedimentation rate; PT, Prothrombin time; PTT, Partial thromboplastin time; CPK, Creatine phosphokinase; LDH, Lactate dehydrogenase. We also studied clinical manifestation in the course of the disease. Cardiac involvement was among one of the considerable organ involvements among patients. The most common cardiac presentation was coronary arteries dilation which was detected in 42 (34.4) patients (41.2% (n = 7) of the MIS-C group overlapping with KD, 33.8% (n = 27) in MIS-C group without overlap with KD, and in 32% (n = 8) of cases with severe MIS-C (P = 0.809), respectively). Hypotension, myocarditis and valvulitis were also seen. Hypotension and myocarditis were significantly more common among patients with severe MIS-C (P < 0.000 and P = 0.041, respectively), while valvulitis was only seen in 2 patients with mild and moderate disease (P = 1.000). Renal involvement was seen in 4 patients, and was significantly more common among patients with severe MIS-C (P = 0.027). In the course of admission, 25 (20.5%) patients required supplemental oxygen, 11 (11.3%) patients with mild and moderate MIS-C and 14 (56.0%) patients with severe MIS-C. This difference was statistically significant (P < 0.0001). Twenty-five (20.5) patients experienced PICU admission and 2 (8.0) patients were intubated. All of these patients had severe MIS-C. In contrast, no patients with mild and moderate disease required PICU care or intubation. These differences were significant (P < 0.0001for both). Unfortunately, 2 (1.6%) of 122 patients died and both had severe MIS-C. The mortality rate was 8% among patients with severe MIS-C, while it was not found in cases with mild and moderate disease. As a result, severe MIS-C significantly increased mortality among patients compared to mild and moderate disease (P = 0.041). Details are shown in Table 3. The median duration of hospital stay in mild and moderate disease was 6 days and in severe disease was 11 days. This finding was also significant (P = 0.000). Given the current national shortage of IVIG in Iran and effectiveness of corticosteroid based on literature, we used pulse glucocorticoids for patients with moderate or severe illness and high-dose glucocorticoids for patients with mild illness. Corticosteroid was used to treat 92 patients (77.3%); 14 (82.4%) MIS-C subjects overlapping with KD, 57 (74.0%) cases without overlap with KD, and 21 (84.0%) subjects with severe disease (P = 0.507). We observed clinical improvement in the way of symptom resolution with the use of steroids in our population. IVIG was also required in 20 patients (18.3%). Totally, 45.5% of patients with severe disease required this treatment, while it was only prescribed in 2 (12.5%) MIS-C subjects overlapping with KD and 8 (11.3%) cases without overlap with KD, respectively (P = 0.001). Table 4 shows the clinical characteristic in the course of the disease and also the applied management and final outcome of the patients. Table 4.Clinical characteristic in the course of the disease, management and outcomeDiseaseSeverity variableaTotal (122)Mild and Moderate MIS-C (97)Severe MIS-C (25)P-valueICU Admission25 (20.5)0 (0)25 (100.0)0.000RespiratoryO2 supplement25 (20.5)11 (11.3)14 (56.0)0.000Non-invasive mechanical ventilation7 (5.7)1 (1.0)6 (24.0)0.000Intubation2 (1.6)0 (0)2 (8.0)0.041CardiacHypotension25 (20.5%)11 (11.3)14 (56.0)0.000Myocarditis2 (1.6)0 (0)2 (8)0.041Valvulitis2 (1.6)2 (2.1)0 (0)1.000Coronary Arteries dilation42 (34.4)34 (35.1)8 (32)0.818Renal involvement4 (3.3)1 (1.0)3 (12)0.027Final outcomeHospital Stay in survived patients (day).6 (5–10)6 (4–8)11 (8–13)0.000Death2 (1.6)0 (0)2 (8.0)0.041aall of the values are in Number (%), except for Hospital stay which is median (IQ 25–75). Clinical characteristic in the course of the disease, management and outcome all of the values are in Number (%), except for Hospital stay which is median (IQ 25–75).
Conclusion
In conclusion, patients with MIS-C in our region suffer from wide range of signs and symptoms. Among laboratory parameters, hypoalbuminemia and low vitamin D levels may predict a more severe course of the disease. Coronary artery dilation is frequently seen among all patients, regardless of disease severity. Mortality rate was 1.6% in our study, all in patients with severe disease.
[ "Introduction", "Case definition", "Therapeutic team" ]
[ "Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first detected in December 2019, in Wuhan, China and rapidly spread throughout the world [1]. World Health Organization (WHO) introduced COVID-19 as a pandemic on March 2020.\nCOVID-19 disease results in spectrum of signs and symptoms, ranging from asymptomatic to severe pneumonia and death [2, 3]. Children are also affected by COVID-19, but they have less severe disease and better outcome than adults [2, 4].\nOn spring 2020, authors announce that a remarkable increase in Kawasaki disease (KD)-like syndrome outbreak in children, is happening, which results in a multisystem inflammatory phenomenon, with footprint of COVID-19 [5–8]. They called this new entity: Multisystem Inflammatory Syndrome in Children (MIS-C). The signs and symptoms of this cytokine mediated syndrome are fever and multiple organ involvement, which results in critically illness requiring hospitalisation and subsequent morbidities in many of cases [9]. Among them, cardiac involvements, including coronary arteries dilation are of particular importance [10, 11]. The association with COVID-19 can be demonstrated by positive SARS-CoV-2 RT-PCR or more common positive SARS-CoV-2 serology; and even a history of exposure to a COVID-19 patient [12]. Many authors studied the demographic features, clinical manifestation and therapeutic approaches of this syndrome [13–15]. We also published our first experience in 45 children affected by MIS-C on August 2020 [16]. Here, we are sharing our second report about children affected by MIS-C. The aim of the present study was to update our knowledge about children with MIS-C in our region. Furthermore, we tried to compare clinical manifestations, laboratory features and final outcome of patients based on disease severity, in order to better understanding of the nature of this novel syndrome.", "Several MIS-C case definitions have been described by national and international committees. We use the CDC's criteria to diagnose the disease in our centre [17]. Characteristics principles for MISC diagnosis as follows:\nAge under 21 yearsFever lasting 24 h or more (documented or subjective)Presence of inflammatory markers (increased ESR, CRP, fibrinogen, procalcitonin, D-dimer, ferritin, LDH, IL-6 and neutrophil; as well as lymphocytopenia and hypoalbuminemia)At least two organ involvement (cardiovascular, respiratory, renal, neurologic, haematologic, gastrointestinal and dermatologic)Severe illness requiring hospital admissionNo other probable diagnosisFootprint of SARS-CoV-2 infection or exposure (positive SARS-CoV-2 RT-PCR, positive SARS-CoV-2 serology, positive SARS-CoV-2 antigen test and exposure to a patient with COVID-19 within last four weeks)\nAge under 21 years\nFever lasting 24 h or more (documented or subjective)\nPresence of inflammatory markers (increased ESR, CRP, fibrinogen, procalcitonin, D-dimer, ferritin, LDH, IL-6 and neutrophil; as well as lymphocytopenia and hypoalbuminemia)\nAt least two organ involvement (cardiovascular, respiratory, renal, neurologic, haematologic, gastrointestinal and dermatologic)\nSevere illness requiring hospital admission\nNo other probable diagnosis\nFootprint of SARS-CoV-2 infection or exposure (positive SARS-CoV-2 RT-PCR, positive SARS-CoV-2 serology, positive SARS-CoV-2 antigen test and exposure to a patient with COVID-19 within last four weeks)\nWe also studied patients in two distinct groups (mild/moderate, and severe) based on the Vasoactive-Inotropic Score (VIS), degree of respiratory support and evidence of organ injury [18] (Table 1).\nTable 1.Disease severity assessment in children diagnosed with MIS-CVariablesDisease severityMildModerateSevereSupplemental oxygen /respiratory supportNoMask or nasal canulaHigh flow oxygen and/or mechanical ventilationVasoactive drugs requirementNoYesYesOrgans involvementMinorMinor or limited Or coronary arteries involvementModerate to severe organ involvement such as ventricular dysfunction and coronary artery aneurysmProgressive diseaseNoYesYes\nDisease severity assessment in children diagnosed with MIS-C\nAlthough distinguishing patients with KD-like MIS-C from those with true KD is usually difficult, the classification of MIS-C vs. KD is based on SARS-CoV-2 testing and exposure history. Patients with positive SARS-CoV-2 testing (or with an exposure to an individual with COVID-19) who also fulfil criteria for complete or incomplete KD are considered to have MIS-C overlapping with KD.", "Our main therapeutic team consisted of general paediatrician, paediatric infectious disease specialist, paediatric rheumatologist, paediatric intensivist, paediatric cardiologist and pharmacologist. As this hospital is a tertiary referral hospital with all subspecialist available, we consulted with other physicians when needed." ]
[ "other", "other", "other" ]
[ "Introduction", "Materials and methods", "Case definition", "Therapeutic team", "Data analysis", "Results", "Discussion", "Conclusion" ]
[ "Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first detected in December 2019, in Wuhan, China and rapidly spread throughout the world [1]. World Health Organization (WHO) introduced COVID-19 as a pandemic on March 2020.\nCOVID-19 disease results in spectrum of signs and symptoms, ranging from asymptomatic to severe pneumonia and death [2, 3]. Children are also affected by COVID-19, but they have less severe disease and better outcome than adults [2, 4].\nOn spring 2020, authors announce that a remarkable increase in Kawasaki disease (KD)-like syndrome outbreak in children, is happening, which results in a multisystem inflammatory phenomenon, with footprint of COVID-19 [5–8]. They called this new entity: Multisystem Inflammatory Syndrome in Children (MIS-C). The signs and symptoms of this cytokine mediated syndrome are fever and multiple organ involvement, which results in critically illness requiring hospitalisation and subsequent morbidities in many of cases [9]. Among them, cardiac involvements, including coronary arteries dilation are of particular importance [10, 11]. The association with COVID-19 can be demonstrated by positive SARS-CoV-2 RT-PCR or more common positive SARS-CoV-2 serology; and even a history of exposure to a COVID-19 patient [12]. Many authors studied the demographic features, clinical manifestation and therapeutic approaches of this syndrome [13–15]. We also published our first experience in 45 children affected by MIS-C on August 2020 [16]. Here, we are sharing our second report about children affected by MIS-C. The aim of the present study was to update our knowledge about children with MIS-C in our region. Furthermore, we tried to compare clinical manifestations, laboratory features and final outcome of patients based on disease severity, in order to better understanding of the nature of this novel syndrome.", "The Research deputy and Ethics committee of Tehran University of Medical Sciences approved the study (IR.TUMS.VCR.REC.1399.057). Since the study was done retrospectively, no further intervention was applied and all of the diagnostic and therapeutic measures were performed based on physician's preference in the duration of hospitalisation; nonetheless, we obtaine a general written informed consent from parents of all patients at the time of admission.\nThis retrospective study was conducted at Children's Medical Center Hospital, the hub of excellence in paediatrics in Iran, located in Tehran, Iran. All patients with suspected MIS-C have paediatric cardiology, paediatric infectious disease and paediatric rheumatology consults. Patients were evaluated for demographic characteristics, history of contact with confirmed or suspected cases of COVID-19, presenting symptoms, clinical and laboratory features, cardiac involvement, treatment modalities and outcomes. The echocardiography (ECHO) and ECG findings for the patients were recorded at the time of admission with follow-up within 2 weeks post discharge with paediatric cardiology.\nRenal involvement was defined according to the kidney function, serum Blood Urea Nitrogen, creatinine sodium, potassium, calcium and glomerular filtration rate according to Schwartz formula.\nWe reviewed medical records of children admitted to the hospital with the diagnosis of MIS-C from July 2020 to October 2021. We have published our first experience in children with MIS-C on August 2020 [16].", "Several MIS-C case definitions have been described by national and international committees. We use the CDC's criteria to diagnose the disease in our centre [17]. Characteristics principles for MISC diagnosis as follows:\nAge under 21 yearsFever lasting 24 h or more (documented or subjective)Presence of inflammatory markers (increased ESR, CRP, fibrinogen, procalcitonin, D-dimer, ferritin, LDH, IL-6 and neutrophil; as well as lymphocytopenia and hypoalbuminemia)At least two organ involvement (cardiovascular, respiratory, renal, neurologic, haematologic, gastrointestinal and dermatologic)Severe illness requiring hospital admissionNo other probable diagnosisFootprint of SARS-CoV-2 infection or exposure (positive SARS-CoV-2 RT-PCR, positive SARS-CoV-2 serology, positive SARS-CoV-2 antigen test and exposure to a patient with COVID-19 within last four weeks)\nAge under 21 years\nFever lasting 24 h or more (documented or subjective)\nPresence of inflammatory markers (increased ESR, CRP, fibrinogen, procalcitonin, D-dimer, ferritin, LDH, IL-6 and neutrophil; as well as lymphocytopenia and hypoalbuminemia)\nAt least two organ involvement (cardiovascular, respiratory, renal, neurologic, haematologic, gastrointestinal and dermatologic)\nSevere illness requiring hospital admission\nNo other probable diagnosis\nFootprint of SARS-CoV-2 infection or exposure (positive SARS-CoV-2 RT-PCR, positive SARS-CoV-2 serology, positive SARS-CoV-2 antigen test and exposure to a patient with COVID-19 within last four weeks)\nWe also studied patients in two distinct groups (mild/moderate, and severe) based on the Vasoactive-Inotropic Score (VIS), degree of respiratory support and evidence of organ injury [18] (Table 1).\nTable 1.Disease severity assessment in children diagnosed with MIS-CVariablesDisease severityMildModerateSevereSupplemental oxygen /respiratory supportNoMask or nasal canulaHigh flow oxygen and/or mechanical ventilationVasoactive drugs requirementNoYesYesOrgans involvementMinorMinor or limited Or coronary arteries involvementModerate to severe organ involvement such as ventricular dysfunction and coronary artery aneurysmProgressive diseaseNoYesYes\nDisease severity assessment in children diagnosed with MIS-C\nAlthough distinguishing patients with KD-like MIS-C from those with true KD is usually difficult, the classification of MIS-C vs. KD is based on SARS-CoV-2 testing and exposure history. Patients with positive SARS-CoV-2 testing (or with an exposure to an individual with COVID-19) who also fulfil criteria for complete or incomplete KD are considered to have MIS-C overlapping with KD.", "Our main therapeutic team consisted of general paediatrician, paediatric infectious disease specialist, paediatric rheumatologist, paediatric intensivist, paediatric cardiologist and pharmacologist. As this hospital is a tertiary referral hospital with all subspecialist available, we consulted with other physicians when needed.", "The statistical analyses were performed using Statistical Package for the Social Sciences (SPSS) version 13.0 software (SPSS Inc.). Categorical variables were reported as frequency and percentages. Normally distributed continuous variables were presented as means with standard deviations (s.d.) and not normally distributed variables were described using median and interquartile range (IQR) values. For comparing laboratory parameters between the groups, the nonparametric tests (Mann–Whitney) or parametric tests (non-paired Student t test) were performed. P < 0.05 was considered as statistically significant.", "One hundred and twenty-two patients enrolled the study. Ninety-seven (79.5%) patients had mild to moderate MIS-C (MIS-C without overlap with KD (n = 80); MIS-C overlapping with KD (n = 17)) and 25 (20.5%) patients showed severe MIS-C.\nForty-eight (39.3%) patients were female and 74 (60.7%) patients were male. There was no significant difference in sex distribution between three groups (P = 0.837). The mean age of all patients was 6.4 ± 4.0 years. The mean age of patients with severe MIS-C (8.2 ± 4.6 years) was significantly higher than MIS-C group without overlap with KD (5.7 ± 3.7 years) and MIS-C group overlapping with KD (6.75 ± 4.4 years), respectively (P = 0.029). Fifteen (12.3%) patients had underlying disease; 11 (11.3%) patients with mild and moderate MIS-C and 4 (16%) patients with severe MIS-C. This was not a noticeable difference (P = 0.507).\nTotally, 74 patients (60.7%) had the history of close contact with an individual with illness clinically compatible with COVID-19 disease who had close contact with an individual with laboratory-confirmed SARS-CoV-2 infection; however, there was no significant difference between two groups (P = 0.172). This was found in 62 cases with mild and moderate MIS-C (64%).\nWe also studied serologic tests as a footprint of COVID-19 in some of our patients as indicated. Positive anti-N SARS-CoV-2 IgM and IgG were detected in 14 (33.3%) and 23 (46.9%) tested patients, respectively. There was no significant difference between mild to moderate and severe disease (P = 1.000 and P = 0.734 respectively). We also obtained a nasopharyngeal SARS-CoV-2 RT-PCR test for all of the patients: 36.9% of samples were positive; 37.1% in patients with mild and moderate disease and 36.9% in patients with severe disease (P = 1.000).\nThe most common clinical manifestation at the presentation was fever which was documented in all patients. Nausea and vomiting (53.3%), skin rash (49.6%), abdominal pain (46.7%) and conjunctivitis (41.8%) were also frequently seen. We also compared clinical manifestations between three groups (MIS-C overlapping with KD, MIS-C without overlap with KD and severe MIS-C). Headache, chest pain, tachypnea and respiratory distress were significantly more common in patients with severe MIS-C (P < 0.0001, P = 0.021, P < 0.0001 and P < 0.0001, respectively). Table 2 shows the clinical manifestations in detail.\nTable 2.Clinical characteristics of patients with MIS-C in different groups at presentationVariables N (%)Disease severityTotal (n = 122)MIS-C overlapping with KD (n = 17)MIS-C without overlap with KD (n = 80)Severe MIS-C (n = 25)P valueConstitutional symptomsHeadache23 (18.9)1 (5.9)10 (12.8)12 (48.0)0.000Myalgia24 (20.3)3 (17.6)18 (23.4)3 (12.5)0.49DermatologicConjunctivitis51 (41.8)7 (41.2)34 (42.5)10 (40.0)0.97Skin rash59 (49.6)9 (52.9)40 (51.3)10 (41.7)0.61Mucus membrane involvement18 (15.1)3 (17.6)11 (14.1)4 (16.7)0.9RespiratoryRhinorrhea3 (2.5)1 (5.9)2 (2.6)0 (0)0.49Cough35 (29.2)4 (23.5)21 (26.9)10 (40)0.39Chest pain4 (3.3)0 (0)1 (1.0)3 (12.0)0.021Tachypnea28 (23.0)0 (0)14 (14.4)14 (56.0)0.000Respiratory distress25 (20.8)0 (0)11 (14.1)14 (56.0)0.000GastrointestinalHepatomegaly7 (6.2)0 (0)5 (6.7)2 (9.1)0.496Splenomegaly9 (8.0)0 (0)7 (9.3)2 (9.1)0.446Ascites11 (9.6)3 (18.8)6 (8)2 (8.7)0.41Abdominal pain56 (46.7)9 (52.9)35 (44.3)12 (50)0.76Nausea and vomiting65 (53.3)5 (29.4)45 (56.3)15 (60.0)0.082Diarrhoea44 (36.7)5 (29.4)33 (41.3)6 (26.1)0.33Ileocolitis5 (4.3)0 (0)4 (5.3)1 (4.3)0.64Colitis2 (1.8)0 (0)2 (2.7)0 (0)0.6Lymphadenopathy6 (5)1 (5.9)3 (3.8)2 (8.0)0.688Sore throat7 (5.8)0 (0)4 (5.1)3 (12.0)0.236Edema25 (21.0)6 (35.3)14 (19.7)5 (20.8)0.282\nClinical characteristics of patients with MIS-C in different groups at presentation\nWe also studied some laboratory parameters in our patients including complete blood count (CBC), biochemistry, liver tests (aspartate transaminase (AST) and alanine transaminase (ALT)), prothrombin time (PT), thromboplastin time (PTT), albumin, inflammatory markers (erythrocyte sedimentation rate (ESR), C-reactive protein (CRP)), Ferritin, IL-6, Fibrinogen, D-dimer, cardiac and other enzymes (Creatine phosphokinase (CPK), CPK-MB, troponin, Lactate dehydrogenase (LDH), amylase, lipase), triglyceride and vitamin D level. We found that vitamin D levels in children with severe MISC were significantly lower than children with mild to moderate MIS-C (P = 0.05). Table 3 shows laboratory information in details.\nTable 3.Laboratory findings of the patients with mild to moderate and severe MIS-CLaboratory characteristics Median (IQR 25–75)Disease severityMild and moderate MIS-CSevere MIS-CP valueCBC\nWBC ( × 10⁹ cells per L)8.350 (6.198–12.178)10.11 (7.255–15.780)0.163Neutrophils ( × 10⁹ cells per L)5.700 (3.190–8.035)8.230 (4.625–12.25)0.370Lymphocyte ( × 10⁹ cells per L)1.520 (0.985–3.105)1.400 (0.665–1.920)0.178Hemoglobin (g/dl)11.4 (10.4–12.6)11.6 (10.6–12.8)0.645Platelet ( × 10⁹ cells per L)235 (129–311)182 (113–247)0.178BiochemistryBUN (mg/dl)11 (8–15)15 (11–19)0.002Creatinine (mg/dl)0.6 (0.5–0.7)0.7 (0.5–0.8)0.160Calcium (mg/dl)8.6 (8.1–9.2)8.4 (7.7–8.9)0.816Phosphorus (mg/dl)3.9 (3.0–4.7)3.7 (2.9–4.3)1.000Magnesium (mg/dl)2.1 (1.9–2.3)2.0 (1.7–2.2)0.397Sodium (mEq/l)134 (130–138)134 (130–1137)0.889Potassium (mEq/l)3.9 (3.6–4.3)3.9 (3.6–4.3)0.831Liver testsAST (U/L)31 (22–59)41 (26–101)0.233ALT (U/L)24 (14–51)27 (16–74)0.711PT (sec)14 (13–16)15 (13–16)1.000PTT (sec)36 (32–40)34 (31–39)0.722Inflammatory markersAlbumin (g/dl)4.1 (3.6–4.4)3.3 (2.9–3.6)<0.0001ESR (mm/h)27 (12–49)30 (15–53)0.493CRP (mg/l)28 (6–54)57 (24–119)0.346Ferritin (ng/ml)193 (95–441)457 (170–1462)0.428Interleukin-6 (pg/ml)76 (11–184)3 (0- 3.3)0.167Fibrinogen (mg/dl)377 (255–508)417 (254–541)0.379D-dimer (ng/ml)2.8 (0.8–316)4.2 (1.0–420.2)0.765EnzymesCPK (U/l)55 (36–105)72 (31–337)0.458CPK-MB (μg/l)16 (10–20)20 (11–37)0.778Troponin0.1 (0.09–0.1)0.1 (0.1–4.2)0.294LDH (U/l)537 (436–670)558 (416–855)0.780Amylase (U/l)65 (28–232)43 (6–117)1.000Lipase (U/l)32 (20–266)13 (3–25)0.598OthersVitamin D (ng/ml)20.5 (8.2–33.7)8.5 (3.0–14.2)0.05Triglyceride (mg/dl)192 (132–283)92 (142–142)1.000WBC, White blood cell count; ALT, Alanine aminotransferase; AST, Aspartate aminotransferase; CRP, C-reactive protein; ESR, Erythrocyte sedimentation rate; PT, Prothrombin time; PTT, Partial thromboplastin time; CPK, Creatine phosphokinase; LDH, Lactate dehydrogenase.\nLaboratory findings of the patients with mild to moderate and severe MIS-C\nWBC, White blood cell count; ALT, Alanine aminotransferase; AST, Aspartate aminotransferase; CRP, C-reactive protein; ESR, Erythrocyte sedimentation rate; PT, Prothrombin time; PTT, Partial thromboplastin time; CPK, Creatine phosphokinase; LDH, Lactate dehydrogenase.\nWe also studied clinical manifestation in the course of the disease. Cardiac involvement was among one of the considerable organ involvements among patients. The most common cardiac presentation was coronary arteries dilation which was detected in 42 (34.4) patients (41.2% (n = 7) of the MIS-C group overlapping with KD, 33.8% (n = 27) in MIS-C group without overlap with KD, and in 32% (n = 8) of cases with severe MIS-C (P = 0.809), respectively). Hypotension, myocarditis and valvulitis were also seen. Hypotension and myocarditis were significantly more common among patients with severe MIS-C (P < 0.000 and P = 0.041, respectively), while valvulitis was only seen in 2 patients with mild and moderate disease (P = 1.000). Renal involvement was seen in 4 patients, and was significantly more common among patients with severe MIS-C (P = 0.027). In the course of admission, 25 (20.5%) patients required supplemental oxygen, 11 (11.3%) patients with mild and moderate MIS-C and 14 (56.0%) patients with severe MIS-C. This difference was statistically significant (P < 0.0001). Twenty-five (20.5) patients experienced PICU admission and 2 (8.0) patients were intubated. All of these patients had severe MIS-C. In contrast, no patients with mild and moderate disease required PICU care or intubation. These differences were significant (P < 0.0001for both).\nUnfortunately, 2 (1.6%) of 122 patients died and both had severe MIS-C. The mortality rate was 8% among patients with severe MIS-C, while it was not found in cases with mild and moderate disease. As a result, severe MIS-C significantly increased mortality among patients compared to mild and moderate disease (P = 0.041). Details are shown in Table 3. The median duration of hospital stay in mild and moderate disease was 6 days and in severe disease was 11 days. This finding was also significant (P = 0.000).\nGiven the current national shortage of IVIG in Iran and effectiveness of corticosteroid based on literature, we used pulse glucocorticoids for patients with moderate or severe illness and high-dose glucocorticoids for patients with mild illness. Corticosteroid was used to treat 92 patients (77.3%); 14 (82.4%) MIS-C subjects overlapping with KD, 57 (74.0%) cases without overlap with KD, and 21 (84.0%) subjects with severe disease (P = 0.507). We observed clinical improvement in the way of symptom resolution with the use of steroids in our population.\nIVIG was also required in 20 patients (18.3%). Totally, 45.5% of patients with severe disease required this treatment, while it was only prescribed in 2 (12.5%) MIS-C subjects overlapping with KD and 8 (11.3%) cases without overlap with KD, respectively (P = 0.001).\nTable 4 shows the clinical characteristic in the course of the disease and also the applied management and final outcome of the patients.\nTable 4.Clinical characteristic in the course of the disease, management and outcomeDiseaseSeverity variableaTotal (122)Mild and Moderate MIS-C (97)Severe MIS-C (25)P-valueICU Admission25 (20.5)0 (0)25 (100.0)0.000RespiratoryO2 supplement25 (20.5)11 (11.3)14 (56.0)0.000Non-invasive mechanical ventilation7 (5.7)1 (1.0)6 (24.0)0.000Intubation2 (1.6)0 (0)2 (8.0)0.041CardiacHypotension25 (20.5%)11 (11.3)14 (56.0)0.000Myocarditis2 (1.6)0 (0)2 (8)0.041Valvulitis2 (1.6)2 (2.1)0 (0)1.000Coronary Arteries dilation42 (34.4)34 (35.1)8 (32)0.818Renal involvement4 (3.3)1 (1.0)3 (12)0.027Final outcomeHospital Stay in survived patients (day).6 (5–10)6 (4–8)11 (8–13)0.000Death2 (1.6)0 (0)2 (8.0)0.041aall of the values are in Number (%), except for Hospital stay which is median (IQ 25–75).\nClinical characteristic in the course of the disease, management and outcome\nall of the values are in Number (%), except for Hospital stay which is median (IQ 25–75).", "Herein, we studied the clinical and laboratory characteristic of one hundred and twenty-two patients admitted to our hospital with the confirmed diagnosis of MIS-C. As mentioned before, our first experience of 45 patients with MIS-C has published earlier, on August 2020 [16] and this manuscript is an update of our centre. Furthermore, in the current study, we categorised patients based on disease severity into two subgroups and we compared them with each other: mild to moderate MIS-C and severe MIS-C.\nWith the exception of fever, which is the cornerstone of the diagnosis, the most observed signs and symptoms at presentation were nausea and vomiting (53.3%), skin rash (49.6%), abdominal pain (46.7%), conjunctivitis (41.8%) and diarrhoea (36.7%). It is almost equal to our previous findings. On the other hand, rhinorrhea, chest pain, hepatomegaly, splenomegaly, ascites, ileitis, colitis, lymphadenopathy and sore throat were seen infrequently, each of them in less than 10% of patients. Among them, headache, chest pain, tachypnea and respiratory distress were significantly more common in patients with severe MIS-C.\nIn a systematic review conducted by Radia et al., gastrointestinal symptoms were the most common clinical presentation (71%), including abdominal pain, diarrhoea and vomiting; 42% of studied patients suffered from skin rash and cough. They reported respiratory tract symptoms as the least observed manifestations (cough and sore throat) [19]. In a case series of 58 children with MIS-C from 8 hospitals in England, abdominal pain, diarrhoea, rash, shock, vomiting and conjunctivitis were frequently seen (more than 40%). Lymphadenopathy, swollen extremities, sore throat and confusion were the least common clinical features [9].\nIn a study of MIS-C in Mumbai-India, diarrhoea/vomiting, pain in abdomen, rash and conjunctivitis were observed in more than half of the patients. They also categorised the patients into MIS-C with shock and MIS-C without shock, based on inotrope requirement and/or more than 20cc/kg fluid resuscitation. They found that patients with shock were older than patients without shock [20]; similarly, we found that our patient with severe MIS-C were older than patients with mild to moderate disease. Findings of above studies, about distribution pattern of clinical manifestations at the beginning of the hospitalisation, were almost consistent with our results.\nElevated COVID-19 IgM and IgG were detected in 33.3% and 46.9% of patients respectively. In addition, 36.9% of subjects were SARS-CoV-2 RT-PCR positive. There was no significant difference between two groups in terms of COVID-19 SARS-CoV-2 serology or SARS-CoV-2 RT-PCR. In other studies, the percentage of patients with evidence of past or current COVID-19 infection was reported higher [9, 19, 21].\nWe studied inflammatory markers and compared them between two groups. ESR, CRP, D-dimer and Ferritin were higher among patients with severe MIS-C, however none of them were statistically significant. Similarly, albumin, as a marker of inflammation, was markedly lower in patients with severe MIS-C. In a systematic review of patients with MIS-C authors found that inflammatory markers were raised in most of the cases, including neutrophilia, elevated CRP level and lymphopenia [19]. In a retrospective case series of children who were hospitalised with the diagnosis of MIS-C in Atlanta, all 11 patients developed hypoalbuminemia and 83% of them received intravenous albumin infusion [22]. In the study of Toreres et al. from Santiago de Chile, patients with severe disease had lower albumin and higher D-dimer levels [23]. Our study also confirmed that hypoalbuminemia might be a predictor of severe disease.\nAnother finding was that vitamin D levels were low in most of the patients and was significantly lower in patients with severe disease. In a review conducted by Feketae et al., vitamin D was introduced as a predictor of MIS-C severity. They also made a hypothesis that correction of vitamin D deficiency may possibly improve the clinical course of the disease [24]. The role of vitamin D in COVID-19 has been well studied by several authors previously [25–27].\nIn our study, 20.5% of patients required ICU admission, all with severe MIS-C. The need for supplemental oxygen, non-invasive ventilation and intubation were also significantly higher in patients with severe MIS-C.\nAs a consequence of cytokine storm, MIS-C results in multi-organ failure [28]. We found that hypotension and myocarditis were significantly more frequent in patients with severe MIS-C; surprisingly, the rate of coronary artery dilation was almost 35% in each two groups; and it was reported 29% in our previous report. Renal involvement was a rare complication and was markedly more frequent in patient with severe MIS-C. It was in consistent with Pouletty et al.'s study. They found that myocarditis found in 86% of patients with severe disease (requiring intensive care) and 55% of patients with non-severe disease; a meaningful finding [29].\nIn the multi-centre observational study conducted in India, 23 patients with MIS-C were included [20]. They found that LV systolic dysfunction was significantly higher in patients with shock. They also reported that coronary artery dilation was seen in 26% of patients, with no significant difference in patient with or without shock, a finding consistent with our results. In another single centre study done in Philadelphia, Corwin et al. defined 3 subgroups for their patients with MIS-C: patient with critical, moderate and mild illness. They found that coronary artery dilation was seen only in 2 of 5 patients with severe illness and none of patients with moderate and mild disease; nonetheless this finding was not statistically significant [30]. So, we can conclude that coronary artery dilation is a relatively common finding in patients with MIS-C, regardless of disease severity. Although there is no clear aetiology for coronary artery dilation, it can be secondary to vasculitis or systemic hyperinflammation, and this abnormality usually resolves within 30 days [31].\nUnfortunately, 2 of the patients with severe MIS-C died, while all children with mild to moderate MIS-C survived. In a study from England [9], the rate of intubation was 43%, which is clearly higher in comparison to our patients. They reported that 14% of patients experienced coronary artery aneurysm (35% in our study). The mortality rate of their patients was almost similar to our study (2% in mentioned study vs 1.6% in our study). In our first report, the mortality rate was 11% [16]. This decrease may be attributed to the increased experience and publication of reliable guidelines in the management of such a novel syndrome.\nIn another study from the USA, 28 children with MIS-C were reviewed retrospectively. Sixty-one per cent of cases required ICU admission, 43% received supplemental oxygen and 25% required noninvasive ventilation; acute kidney injury was seen in 21% of patients [21]. All of these complications were higher among their patients compare to ours, however they reported no death. These results might be explained in part by racial difference, better care and more equipped facilities in a developed country.\nThe initial steps in the management of critically ill children with COVID-19 are supportive cares, including oxygen supplement and hemodynamic support. The goals of treatment are decreasing systemic inflammation in order to lessen morbidity and mortality. For these purposes, IVIG, corticosteroid and tocilizumab have been suggested by authors [21, 30]. In our centre, IVIG and corticosteroid are commonly prescribed. Totally, corticosteroid and IVIG was prescribed in 77.3% and 18.3% of our patients respectively and IVIG was significantly used more frequently in patients with severe disease. In other studies, IVIG was prescribed clearly more frequently; it can be well explained by the fact that IVIG is among the scarce drugs in Iran, mainly due to high cost and insurance policies; so, the prescription is completely strict and only in definite indications with approved efficacy.\nTo our knowledge, we reported a relatively large numbers of MIS-C. However, it has some limitations. Our centre is a referral hospital and several children with complex complicated diseases have been referred to our hospital. Our study may therefore represent a more severe form of disease, and the results should be interpreted with caution.", "In conclusion, patients with MIS-C in our region suffer from wide range of signs and symptoms. Among laboratory parameters, hypoalbuminemia and low vitamin D levels may predict a more severe course of the disease. Coronary artery dilation is frequently seen among all patients, regardless of disease severity. Mortality rate was 1.6% in our study, all in patients with severe disease." ]
[ "other", "materials", "other", "other", "other", "results", "discussion", "conclusions" ]
[ "COVID-19", "MIS-C", "SARS-CoV-2" ]
Systematic review of shared decision-making in guidelines about colorectal cancer screening.
36254840
We aimed to systematically evaluate quality of shared decision-making (SDM) in colorectal cancer (CRC) screening clinical practice guidelines (CPGs) and consensus statements (CSs).
INTRODUCTION
Search for CRC screening guidances was from 2010 to November 2021 in EMBASE, Web of Science, MEDLINE, Scopus and CDSR, and the World Wide Web. Three independent reviewers and an arbitrator rated the quality of each guidance using a SDM quality assessment tool (maximum score: 31). Reviewer agreement was 0.88.
METHODS
SDM appeared in 41/83 (49.4%) CPGs and 9/19 (47.4%) CSs. None met all the quality criteria, and 51.0% (52/102) failed to meet any quality items. Overall compliance was low (mean 1.63, IQR 0-2). Quality was better in guidances published after 2015 (mean 1, IQR 0-3 vs. mean 0.5, IQR 0-1.5; p = 0.048) and when the term SDM was specifically reported (mean 4.5, IQR 2.5-4.5 vs. mean 0.5, IQR 0-1.5; p < 0.001). CPGs underpinned by systematic reviews showed better SDM quality than consensus (mean 1, IQR 0-3 vs. mean 0, IQR 0-2, p = 0.040).
RESULTS
SDM quality was suboptimal and mentioned in less than half of the guidances, and recommendations were scarce. Guideline developers should incorporate evidence-based SDM recommendations in guidances to underpin the translation of evidence into practice.
CONCLUSION
[ "Humans", "Colorectal Neoplasms", "Decision Making", "Decision Making, Shared", "Early Detection of Cancer", "Patient Participation" ]
9786598
null
null
METHODS
Following prospective registration (Prospero no: CRD42021286156), this systematic review was conducted following advocated methods for search, assessment and reporting of guidelines using the Preferred Reporting Items for Systematic Reviews and Meta‐Analysis (PRISMA) statement (see Appendix S1) (Liberati et al., 2009; Moher et al., 2009). [SUBTITLE] Search strategy, data sources, inclusion and exclusion criteria [SUBSECTION] A search for relevant publications covering major electronic databases (EMBASE, Web of Science, MEDLINE, Scopus and CDSR) was developed without language restrictions to capture peer‐reviewed and grey literature from 2010 until November 2021. We also searched 59 websites of important professional societies and eight guidance‐specific databases. We explored the World Wide Web to include professional societies from countries with global CRC scientific production bigger than 0.5% (Maes‐Carballo et al., 2021, 2022; Maes‐Carballo, Mignini, et al., 2020). A total of 85,932 ‘Colorectal Cancer and Health’ records were scrutinised from Scopus on 10 March 2022 to calculate the scientific production of each country. This decision is adhered to rules followed by other previous peer‐reviewed published systematic reviews (Maes‐Carballo et al., 2021, 2022; Maes‐Carballo, Mignini, et al., 2020; Maes‐Carballo, Munoz‐Nunez, et al., 2020). We revised references from the guidances retrieved to search for conceivable additional CPGs or CSs. The final search integrated MeSH terms ‘practice guidelines’, ‘guidelines’, ‘consensus’, ‘colorectal neoplasms’, ‘colorectal cancer’, ‘screening’ and including word alternatives. Details of the search strategy were documented in Appendix S2. Endnote X9 software was employed to handle the searches downloaded. Selection criteria captured eligible guidances about CRC screening produced by national or international professional organisations and societies or governmental agencies. Those guidances in which screening was the central issue and those in which there was a section dedicated to screening or prevention were included in our systematic review. We excluded protocols or screening programme documents, CPGs and CSs about diagnosis and treatment, out‐of‐date guidelines replaced by updates from the same organisation, and CPG and CSs for education and information purposes, randomised controlled trials, observational studies, narrative reviews, scientific reports, discussion papers, conference abstracts, and posters. Studies were chosen through a multi‐step approach, including deleting duplicates, reading titles and abstracts, and assessing full texts. Initially, titles and abstracts were considered for eligibility by two reviewers (CR‐E and AI‐A). Then, full texts were appraised for eligibility also by these two reviewers. Potential disagreements or inconsistencies were decided by arbitration of another reviewer (MM‐C). Articles in duplication were recognised and excluded. When several versions of the same guidance were discovered, the most updated version was incorporated. Data were extracted independently and duplicated by three reviewers (YG‐F, CR‐E and AI‐A). Disagreements were solved by consensus or arbitration (MM‐C). A search for relevant publications covering major electronic databases (EMBASE, Web of Science, MEDLINE, Scopus and CDSR) was developed without language restrictions to capture peer‐reviewed and grey literature from 2010 until November 2021. We also searched 59 websites of important professional societies and eight guidance‐specific databases. We explored the World Wide Web to include professional societies from countries with global CRC scientific production bigger than 0.5% (Maes‐Carballo et al., 2021, 2022; Maes‐Carballo, Mignini, et al., 2020). A total of 85,932 ‘Colorectal Cancer and Health’ records were scrutinised from Scopus on 10 March 2022 to calculate the scientific production of each country. This decision is adhered to rules followed by other previous peer‐reviewed published systematic reviews (Maes‐Carballo et al., 2021, 2022; Maes‐Carballo, Mignini, et al., 2020; Maes‐Carballo, Munoz‐Nunez, et al., 2020). We revised references from the guidances retrieved to search for conceivable additional CPGs or CSs. The final search integrated MeSH terms ‘practice guidelines’, ‘guidelines’, ‘consensus’, ‘colorectal neoplasms’, ‘colorectal cancer’, ‘screening’ and including word alternatives. Details of the search strategy were documented in Appendix S2. Endnote X9 software was employed to handle the searches downloaded. Selection criteria captured eligible guidances about CRC screening produced by national or international professional organisations and societies or governmental agencies. Those guidances in which screening was the central issue and those in which there was a section dedicated to screening or prevention were included in our systematic review. We excluded protocols or screening programme documents, CPGs and CSs about diagnosis and treatment, out‐of‐date guidelines replaced by updates from the same organisation, and CPG and CSs for education and information purposes, randomised controlled trials, observational studies, narrative reviews, scientific reports, discussion papers, conference abstracts, and posters. Studies were chosen through a multi‐step approach, including deleting duplicates, reading titles and abstracts, and assessing full texts. Initially, titles and abstracts were considered for eligibility by two reviewers (CR‐E and AI‐A). Then, full texts were appraised for eligibility also by these two reviewers. Potential disagreements or inconsistencies were decided by arbitration of another reviewer (MM‐C). Articles in duplication were recognised and excluded. When several versions of the same guidance were discovered, the most updated version was incorporated. Data were extracted independently and duplicated by three reviewers (YG‐F, CR‐E and AI‐A). Disagreements were solved by consensus or arbitration (MM‐C). [SUBTITLE] Quality appraisal of guidances and data extraction [SUBSECTION] The included guidance's characteristics and quality were extracted into a piloted electronic data extraction sheet. The methodological quality assessment was estimated using an already published appraisal tool consisting of a 31‐item checklist grouped into 11 domains (Maes‐Carballo, Munoz‐Nunez, et al., 2020). Three reviewers (YG‐F, CRE‐L and AI‐A) evaluated the quality of SDM in CRC screening guidances (Appendix S3). The 11 domains were basic information (items 1–4), background (items 5–7), selection criteria (items 8–9), strengths and limitations (items 10–14), recommendations about SDM (items 15–17), facilitators and barriers (items 18–19), implementation (items 20–21), resource implications (items 22–24), monitoring and auditing criteria (items 25–27), recommendations for further research and limitations about these recommendations described (items 28–29), and editorial independence and declaration of interest (items 30–31). In those general guidances on the management of CRC, SDM was only considered if it was covered in the screening section. The questions were designed for a binary ‘yes/no’ answer: ‘Yes’ or 1 if the item was met and ‘no’ or ‘0’ if the criterion was not accomplished. The high quality was related to a higher quantity of items completed in the CPG or CS evaluated. No formal score or cut‐off point was specified to determine the quality (Maes‐Carballo, Munoz‐Nunez, et al., 2020). All three reviewers (YG‐F, CRE‐L and AI‐A) had a prior meeting receiving a training seminar and workshop from the arbitrator and creator of the tool (MM‐C) that included education on SDM and the process and application of the SDM tool. The included guidance's characteristics and quality were extracted into a piloted electronic data extraction sheet. The methodological quality assessment was estimated using an already published appraisal tool consisting of a 31‐item checklist grouped into 11 domains (Maes‐Carballo, Munoz‐Nunez, et al., 2020). Three reviewers (YG‐F, CRE‐L and AI‐A) evaluated the quality of SDM in CRC screening guidances (Appendix S3). The 11 domains were basic information (items 1–4), background (items 5–7), selection criteria (items 8–9), strengths and limitations (items 10–14), recommendations about SDM (items 15–17), facilitators and barriers (items 18–19), implementation (items 20–21), resource implications (items 22–24), monitoring and auditing criteria (items 25–27), recommendations for further research and limitations about these recommendations described (items 28–29), and editorial independence and declaration of interest (items 30–31). In those general guidances on the management of CRC, SDM was only considered if it was covered in the screening section. The questions were designed for a binary ‘yes/no’ answer: ‘Yes’ or 1 if the item was met and ‘no’ or ‘0’ if the criterion was not accomplished. The high quality was related to a higher quantity of items completed in the CPG or CS evaluated. No formal score or cut‐off point was specified to determine the quality (Maes‐Carballo, Munoz‐Nunez, et al., 2020). All three reviewers (YG‐F, CRE‐L and AI‐A) had a prior meeting receiving a training seminar and workshop from the arbitrator and creator of the tool (MM‐C) that included education on SDM and the process and application of the SDM tool. [SUBTITLE] Evidence synthesis, investigation of heterogeneity and data analysis [SUBSECTION] A descriptive analysis of the characteristics and quality of the selected guidances was conducted. Statistical data analysis was completed using Stata 16. The Kruskal–Wallis test was utilised to compare scores and stratify for factors or characteristics that may influence the quality of SDM in CPGs and CSs. Values were assessed statistically significant when p < 0.05. Inter‐rater reliability between reviewers in data extraction was calculated using the intra‐class correlation coefficient (ICC). A result of more than 0.75 was considered good (Koo & Li, 2016). A descriptive analysis of the characteristics and quality of the selected guidances was conducted. Statistical data analysis was completed using Stata 16. The Kruskal–Wallis test was utilised to compare scores and stratify for factors or characteristics that may influence the quality of SDM in CPGs and CSs. Values were assessed statistically significant when p < 0.05. Inter‐rater reliability between reviewers in data extraction was calculated using the intra‐class correlation coefficient (ICC). A result of more than 0.75 was considered good (Koo & Li, 2016).
RESULTS
[SUBTITLE] Study selection [SUBSECTION] The study selection process is illustrated in the flow diagram in Figure 1. The initial search identified 8229 citations. We removed 439 duplicates and 7676 records for not meeting the selection criteria (inappropriate population, publication, development group or outdated guidance). A total of 114 records were filtered through reviews of titles and abstracts. Later, we obtained the full text of 114 citations for eligibility assessment, and finally, 102 guidances (83 CPGs) (Alberta, 2020; Alsanea et al., 2015; Aranda et al., 2015; Aranda‐Hernandez et al., 2016; Atkin et al., 2012; Austoker et al., 2012; Benton et al., 2016; Bo In Lee et al., 2012; Brenner et al., 2017; Brouwers et al., 2011; Canadian task Force on preventive health C, 2016; Clarke & Feuerstein, 2019; Cubiella et al., 2018; Day et al., 2011; Del Giudice et al., 2014; Duffy et al., 2014; European Colorectal Cancer Screening Guidelines Working Group et al., 2013; European Commission, 2010; Fabio Leonel Gil Parada et al., 2015; Gupta et al., 2019; Halloran et al., 2012; Hassan et al., 2013; Helsingen et al., 2019; Hospital provincial Neuquén, 2016; Instituto Mexicano del Seguro Social. Guía de Práctica Clínica, 2010; Instituto Nacional del Cáncer, 2015; Jenkins et al., 2018; Jenkinson & Steele, 2010; Jover et al., 2012; Kwaan & Jones‐Webb, 2018; Lam et al., 2018; Lansdorp‐Vogelaar et al., 2012; Leddin et al., 2013; Lee et al., 2012; Leong et al., 2017; Lieberman, 2012; Lopes et al., 2018; Malila et al., 2012; Ministry of Health, 2010, 2016; Minozzi et al., 2012; Monahan et al., 2019; Moreno et al., 2018; Moss et al., 2012; Network NCC, 2021; New Brunswick Colon Cancer Screening, 2013; NHS, 2021; Ong et al., 2014; Provenzale et al., 2016; Qaseem et al., 2019; Quirke et al., 2011, 2012; Recommended Cancer Screenings, 2013; Regula & Kaminski, 2010; Rex et al., 2017; Rubeca et al., 2017; Salzman et al., 2016; SemFYC AEdG, 2018; Seppälä et al., 2021; Shaukat et al., 2021; Society AC, 2018; Spada et al., 2014; Steele, Pox et al., 2012; Steele, Rey et al., 2012; Steinwachs et al., 2010; Stoffel et al., 2015; Tanaka et al., 2015; Telford, 2011; Tinmouth et al., 2014; Uruguay MdSd. Ministerio de Salud de Uruguay, 2018; US Preventive Services Task Force et al., 2021; Valori et al., 2012; Vasen et al., 2014; Vieth et al., 2012; von Karsa et al., 2012; Washington KFHPo, 2021; Wilkins et al., 2018; Wilkinson et al., 2019; Wolf et al., 2018; Wong et al., 2015; Yang et al., 2020; Zeimet et al., 2017; 손대경 김, 박윤희, 서민아, 신애선, 이희영, 임종필, 조현민, 홍성필, 김백희, 김용수, 김정욱, 김현수, 남정모, 박동일, 엄준원, 오순남, 임환섭, 장희진, 함상근, 정지혜, 김수영, 김열, 이원철, 정승용, 2015) and 19 CSs (Asociación Mexicana de Endoscopia Gastrointestinal y Colegio de Profesionistas, 2016; ANM. Programa Nacional de Consensos Inter‐Sociedades, 2010; Basu et al., 2021; García‐Carbonero et al., 2017; Giardiello et al., 2014; Hadjiliadis et al., 2018; Heresbach et al., 2016; Hyer et al., 2019; Johnson et al., 2014; Leddin et al., 2010, 2018; Lieberman et al., 2012; Rembacken et al., 2012; Robertson et al., 2017; Schmiegel et al., 2010; Schmoll et al., 2012; Sollano et al., 2017; Sung et al., 2015; 中国抗癌协会肿瘤内镜学专业委员会 国上国中医中中消中中中金国, 2019) were included in our study for the last appraisal. The flow diagram detailing the study selection The study selection process is illustrated in the flow diagram in Figure 1. The initial search identified 8229 citations. We removed 439 duplicates and 7676 records for not meeting the selection criteria (inappropriate population, publication, development group or outdated guidance). A total of 114 records were filtered through reviews of titles and abstracts. Later, we obtained the full text of 114 citations for eligibility assessment, and finally, 102 guidances (83 CPGs) (Alberta, 2020; Alsanea et al., 2015; Aranda et al., 2015; Aranda‐Hernandez et al., 2016; Atkin et al., 2012; Austoker et al., 2012; Benton et al., 2016; Bo In Lee et al., 2012; Brenner et al., 2017; Brouwers et al., 2011; Canadian task Force on preventive health C, 2016; Clarke & Feuerstein, 2019; Cubiella et al., 2018; Day et al., 2011; Del Giudice et al., 2014; Duffy et al., 2014; European Colorectal Cancer Screening Guidelines Working Group et al., 2013; European Commission, 2010; Fabio Leonel Gil Parada et al., 2015; Gupta et al., 2019; Halloran et al., 2012; Hassan et al., 2013; Helsingen et al., 2019; Hospital provincial Neuquén, 2016; Instituto Mexicano del Seguro Social. Guía de Práctica Clínica, 2010; Instituto Nacional del Cáncer, 2015; Jenkins et al., 2018; Jenkinson & Steele, 2010; Jover et al., 2012; Kwaan & Jones‐Webb, 2018; Lam et al., 2018; Lansdorp‐Vogelaar et al., 2012; Leddin et al., 2013; Lee et al., 2012; Leong et al., 2017; Lieberman, 2012; Lopes et al., 2018; Malila et al., 2012; Ministry of Health, 2010, 2016; Minozzi et al., 2012; Monahan et al., 2019; Moreno et al., 2018; Moss et al., 2012; Network NCC, 2021; New Brunswick Colon Cancer Screening, 2013; NHS, 2021; Ong et al., 2014; Provenzale et al., 2016; Qaseem et al., 2019; Quirke et al., 2011, 2012; Recommended Cancer Screenings, 2013; Regula & Kaminski, 2010; Rex et al., 2017; Rubeca et al., 2017; Salzman et al., 2016; SemFYC AEdG, 2018; Seppälä et al., 2021; Shaukat et al., 2021; Society AC, 2018; Spada et al., 2014; Steele, Pox et al., 2012; Steele, Rey et al., 2012; Steinwachs et al., 2010; Stoffel et al., 2015; Tanaka et al., 2015; Telford, 2011; Tinmouth et al., 2014; Uruguay MdSd. Ministerio de Salud de Uruguay, 2018; US Preventive Services Task Force et al., 2021; Valori et al., 2012; Vasen et al., 2014; Vieth et al., 2012; von Karsa et al., 2012; Washington KFHPo, 2021; Wilkins et al., 2018; Wilkinson et al., 2019; Wolf et al., 2018; Wong et al., 2015; Yang et al., 2020; Zeimet et al., 2017; 손대경 김, 박윤희, 서민아, 신애선, 이희영, 임종필, 조현민, 홍성필, 김백희, 김용수, 김정욱, 김현수, 남정모, 박동일, 엄준원, 오순남, 임환섭, 장희진, 함상근, 정지혜, 김수영, 김열, 이원철, 정승용, 2015) and 19 CSs (Asociación Mexicana de Endoscopia Gastrointestinal y Colegio de Profesionistas, 2016; ANM. Programa Nacional de Consensos Inter‐Sociedades, 2010; Basu et al., 2021; García‐Carbonero et al., 2017; Giardiello et al., 2014; Hadjiliadis et al., 2018; Heresbach et al., 2016; Hyer et al., 2019; Johnson et al., 2014; Leddin et al., 2010, 2018; Lieberman et al., 2012; Rembacken et al., 2012; Robertson et al., 2017; Schmiegel et al., 2010; Schmoll et al., 2012; Sollano et al., 2017; Sung et al., 2015; 中国抗癌协会肿瘤内镜学专业委员会 国上国中医中中消中中中金国, 2019) were included in our study for the last appraisal. The flow diagram detailing the study selection [SUBTITLE] Characteristics of guidances and quality appraisal [SUBSECTION] The guidance's characteristics (type of document, entity, country, year, journal of publication, version and evidence analysis) were reported in Table 1. The guidances' mean number of items related to SDM was 1.63 (IQR 0–2). The quality assessment results using the SDM instrument are shown in Figure 2 and Appendix S4. No significant differences were obtained between CPGs and CSs concerning the SDM quality (p = 0.959). A total of 50 guidances (49.0%), 41/83 (49.4%) CPGs and 9/19 (47.4%) CSs, reported something about SDM. None of the guidelines met all the quality criteria, and 51.0% of the guidelines accomplished 0 items, and only 5.9% of them accomplished more than 25% items (8/31). When the SDM term was specifically cited in the guidance (n = 13), the quality of the CPG or CSs concerning SDM was better than when it did not appear (n = 89) (mean 4.5, IQR 2.5–4.5 vs. mean 0.5, IQR 0–1.5; p < 0.001). Description of the screening clinical practice guidelines (CPGs) and consensus (CSs) (n=102) selected for the systematic review on the quality of reporting for SDM The compliance of the items with the SDM quality and reporting analysis tool The best‐scored domains were basic information (domain 1) with a range of guidances accomplishing items from 3 to 50, background (domain 2) with a range from 1 to 15, and recommendations (domain 5) with a range from 5 to 13. Resource implications (domain 8), monitoring and auditing criteria (domain 9), and independence and conflict of interest (domain 11) did not appraise any of the items in any guidance. Only 13/102 (12.7%), 10/102 (9.8%) and 3/102 (2.9%) guidances informed SDM in their executive summary, table of content, glossary, abbreviations, acronyms or topic indexes. SDM concept was only explained in one (1.0%) guidance. Both the primary population and patient subgroups with special consideration were characterised in 15/102 (14.7%). The search strategy and the study design and methodology limitations were reported in 2/102 (2.0%), respectively. The importance of the outcomes and the consistency of the results were detailed in only one guidance (1.0%) each. No PICO question was identified in any guidance. The benefits vs. harms of SDM in CRC screening were considered only in 3/102 (2.9%). The evidence of using SDM was exhibited in 2/102 (2.0%). Recommendations about SDM use were clear, precise and reliable in only 7/102 (6.9%) guidance documents, and these recommendations were well‐reported regarding specific subgroups in 5/102 (4.9%) guidances. Facilitators and barriers for SDM application were well‐described in 4/102 (3.9%), advice on applying SDM in clinical routine in 7/102 (6.9%), and additional materials were provided in other 7/102 (6.9%). Suggestions for further research were located in 2/102 (2.0%), and limitations about SDM recommendations were also described in 2/102 (2.0%). No information about SDM implementation cost, any criteria to assess and measure SDM adherence, conflict of interest regarding SDM or a declaration of the value of SDM in clinical practice. The European Commission (Austoker et al., 2012; European Commission, 2010) guidances and the American Cancer Society (ACS) (Wolf et al., 2018) CPG achieved the highest number of quality SDM items completed (Appendix S4). The guidance's characteristics (type of document, entity, country, year, journal of publication, version and evidence analysis) were reported in Table 1. The guidances' mean number of items related to SDM was 1.63 (IQR 0–2). The quality assessment results using the SDM instrument are shown in Figure 2 and Appendix S4. No significant differences were obtained between CPGs and CSs concerning the SDM quality (p = 0.959). A total of 50 guidances (49.0%), 41/83 (49.4%) CPGs and 9/19 (47.4%) CSs, reported something about SDM. None of the guidelines met all the quality criteria, and 51.0% of the guidelines accomplished 0 items, and only 5.9% of them accomplished more than 25% items (8/31). When the SDM term was specifically cited in the guidance (n = 13), the quality of the CPG or CSs concerning SDM was better than when it did not appear (n = 89) (mean 4.5, IQR 2.5–4.5 vs. mean 0.5, IQR 0–1.5; p < 0.001). Description of the screening clinical practice guidelines (CPGs) and consensus (CSs) (n=102) selected for the systematic review on the quality of reporting for SDM The compliance of the items with the SDM quality and reporting analysis tool The best‐scored domains were basic information (domain 1) with a range of guidances accomplishing items from 3 to 50, background (domain 2) with a range from 1 to 15, and recommendations (domain 5) with a range from 5 to 13. Resource implications (domain 8), monitoring and auditing criteria (domain 9), and independence and conflict of interest (domain 11) did not appraise any of the items in any guidance. Only 13/102 (12.7%), 10/102 (9.8%) and 3/102 (2.9%) guidances informed SDM in their executive summary, table of content, glossary, abbreviations, acronyms or topic indexes. SDM concept was only explained in one (1.0%) guidance. Both the primary population and patient subgroups with special consideration were characterised in 15/102 (14.7%). The search strategy and the study design and methodology limitations were reported in 2/102 (2.0%), respectively. The importance of the outcomes and the consistency of the results were detailed in only one guidance (1.0%) each. No PICO question was identified in any guidance. The benefits vs. harms of SDM in CRC screening were considered only in 3/102 (2.9%). The evidence of using SDM was exhibited in 2/102 (2.0%). Recommendations about SDM use were clear, precise and reliable in only 7/102 (6.9%) guidance documents, and these recommendations were well‐reported regarding specific subgroups in 5/102 (4.9%) guidances. Facilitators and barriers for SDM application were well‐described in 4/102 (3.9%), advice on applying SDM in clinical routine in 7/102 (6.9%), and additional materials were provided in other 7/102 (6.9%). Suggestions for further research were located in 2/102 (2.0%), and limitations about SDM recommendations were also described in 2/102 (2.0%). No information about SDM implementation cost, any criteria to assess and measure SDM adherence, conflict of interest regarding SDM or a declaration of the value of SDM in clinical practice. The European Commission (Austoker et al., 2012; European Commission, 2010) guidances and the American Cancer Society (ACS) (Wolf et al., 2018) CPG achieved the highest number of quality SDM items completed (Appendix S4). [SUBTITLE] Analysis of guidances' characteristics [SUBSECTION] The countries' distribution concerning SDM was erratic. Most of the guidances were from Europe (41/102; %) or North America (40/102; %). Table 2 shows factors than may influence the SDM quality of the guidances. Two CPGs or CSs were from Africa or Oceania (2/102; %). Asia and South America had 12/102 (%) and 5/12 (%), respectively. The quality of SDM did not vary between continents (p = 0.233). Factors that may influence the SDM quality and reporting of the CRC screening guidances A greater tendency to introduce and recommend SDM was observed in the most recent guidances (Figure 3). The publication year after 2015 had an important influence on the quality than older publications (mean 1, IQR 0–3 vs. mean 0.5, IQR 0–1.5; p = 0.048). The publication in a journal (p = 0.131), and the version number (p = 0.416). The specific quality tool referral increased the quality and reporting of SDM on guidances (p < 0.001). CPGs following systematic reviews had better quality than consensus or literature reviews or when it was not reported (mean 1, IQR 0–3 vs. mean 0, IQR 0–2 vs. mean 0, IQR 0–1; p = 0.040). Appearance of SDM concerning the published year of the guidance document The countries' distribution concerning SDM was erratic. Most of the guidances were from Europe (41/102; %) or North America (40/102; %). Table 2 shows factors than may influence the SDM quality of the guidances. Two CPGs or CSs were from Africa or Oceania (2/102; %). Asia and South America had 12/102 (%) and 5/12 (%), respectively. The quality of SDM did not vary between continents (p = 0.233). Factors that may influence the SDM quality and reporting of the CRC screening guidances A greater tendency to introduce and recommend SDM was observed in the most recent guidances (Figure 3). The publication year after 2015 had an important influence on the quality than older publications (mean 1, IQR 0–3 vs. mean 0.5, IQR 0–1.5; p = 0.048). The publication in a journal (p = 0.131), and the version number (p = 0.416). The specific quality tool referral increased the quality and reporting of SDM on guidances (p < 0.001). CPGs following systematic reviews had better quality than consensus or literature reviews or when it was not reported (mean 1, IQR 0–3 vs. mean 0, IQR 0–2 vs. mean 0, IQR 0–1; p = 0.040). Appearance of SDM concerning the published year of the guidance document
Conclusions
This systematic review demonstrated that SDM quality in guidance documents was suboptimal as it did not appear in half of the guidances analyzed. SDM recommendations were scarce and unclear. Recent guidances following systematic reviews and referring to quality tools (e.g. AGREE II or RIGHT) had better SDM quality. Therefore, guideline developers, professional institutions, medical journals and policymakers should consider including evidence‐based SDM recommendations in trustworthy and well‐developed guidances to ensure proper translation of evidence into practice.
[ "INTRODUCTION", "Search strategy, data sources, inclusion and exclusion criteria", "Quality appraisal of guidances and data extraction", "Evidence synthesis, investigation of heterogeneity and data analysis", "Study selection", "Characteristics of guidances and quality appraisal", "Analysis of guidances' characteristics", "Main findings", "Strengths and weaknesses", "Implications" ]
[ "Colorectal cancer (CRC) incidence and mortality have decreased due to screening programmes, removing precancerous polyps with colonoscopy, and advances in management (Cotton et al., 1996; Zauber et al., 2012). In screening programmes, the patient's overall health, prior screening history, and preferences and values must be considered in selecting an individualised approach adapted to the patient's risk of acquiring CRC (Hoffmann et al., 2020; US Preventive Services Task Force et al., 2021). Shared decision‐making (SDM) is essential for decision‐making, weighing up risks vs. benefits for each individual patient (Schrager & Burnside, 2017). In recent years, health‐care policymaking has emphasised SDM as a cornerstone of evidence‐based and patient‐centred care (Barry & Edgman‐Levitan, 2012; Elwyn et al., 2010). Institutional promotion is fundamental for improving SDM application (Senate and House of Representatives, 2010), and clinical practice guidelines (CPGs) and consensus statements (CSs) should promote it and advise about its execution (Maes‐Carballo, Munoz‐Nunez, et al., 2020). Although CPGs and CSs increasingly support it (Gärtner et al., 2019), they remain unclear on accomplishing SDM in routine practice (Elwyn et al., 2012). Therefore, the analysis of the quality of SDM in guidances is critically essential. To the best of our knowledge, no systematic review has investigated SDM in CRC screening guidances.\nConsidering this background, this systematic review aimed to analyze SDM in CRC screening CPGs and CSs, evaluating the quality of recommendations about SDM.", "A search for relevant publications covering major electronic databases (EMBASE, Web of Science, MEDLINE, Scopus and CDSR) was developed without language restrictions to capture peer‐reviewed and grey literature from 2010 until November 2021. We also searched 59 websites of important professional societies and eight guidance‐specific databases. We explored the World Wide Web to include professional societies from countries with global CRC scientific production bigger than 0.5% (Maes‐Carballo et al., 2021, 2022; Maes‐Carballo, Mignini, et al., 2020). A total of 85,932 ‘Colorectal Cancer and Health’ records were scrutinised from Scopus on 10 March 2022 to calculate the scientific production of each country. This decision is adhered to rules followed by other previous peer‐reviewed published systematic reviews (Maes‐Carballo et al., 2021, 2022; Maes‐Carballo, Mignini, et al., 2020; Maes‐Carballo, Munoz‐Nunez, et al., 2020). We revised references from the guidances retrieved to search for conceivable additional CPGs or CSs. The final search integrated MeSH terms ‘practice guidelines’, ‘guidelines’, ‘consensus’, ‘colorectal neoplasms’, ‘colorectal cancer’, ‘screening’ and including word alternatives. Details of the search strategy were documented in Appendix S2. Endnote X9 software was employed to handle the searches downloaded.\nSelection criteria captured eligible guidances about CRC screening produced by national or international professional organisations and societies or governmental agencies. Those guidances in which screening was the central issue and those in which there was a section dedicated to screening or prevention were included in our systematic review. We excluded protocols or screening programme documents, CPGs and CSs about diagnosis and treatment, out‐of‐date guidelines replaced by updates from the same organisation, and CPG and CSs for education and information purposes, randomised controlled trials, observational studies, narrative reviews, scientific reports, discussion papers, conference abstracts, and posters.\nStudies were chosen through a multi‐step approach, including deleting duplicates, reading titles and abstracts, and assessing full texts. Initially, titles and abstracts were considered for eligibility by two reviewers (CR‐E and AI‐A). Then, full texts were appraised for eligibility also by these two reviewers. Potential disagreements or inconsistencies were decided by arbitration of another reviewer (MM‐C). Articles in duplication were recognised and excluded. When several versions of the same guidance were discovered, the most updated version was incorporated. Data were extracted independently and duplicated by three reviewers (YG‐F, CR‐E and AI‐A). Disagreements were solved by consensus or arbitration (MM‐C).", "The included guidance's characteristics and quality were extracted into a piloted electronic data extraction sheet. The methodological quality assessment was estimated using an already published appraisal tool consisting of a 31‐item checklist grouped into 11 domains (Maes‐Carballo, Munoz‐Nunez, et al., 2020). Three reviewers (YG‐F, CRE‐L and AI‐A) evaluated the quality of SDM in CRC screening guidances (Appendix S3). The 11 domains were basic information (items 1–4), background (items 5–7), selection criteria (items 8–9), strengths and limitations (items 10–14), recommendations about SDM (items 15–17), facilitators and barriers (items 18–19), implementation (items 20–21), resource implications (items 22–24), monitoring and auditing criteria (items 25–27), recommendations for further research and limitations about these recommendations described (items 28–29), and editorial independence and declaration of interest (items 30–31). In those general guidances on the management of CRC, SDM was only considered if it was covered in the screening section. The questions were designed for a binary ‘yes/no’ answer: ‘Yes’ or 1 if the item was met and ‘no’ or ‘0’ if the criterion was not accomplished. The high quality was related to a higher quantity of items completed in the CPG or CS evaluated. No formal score or cut‐off point was specified to determine the quality (Maes‐Carballo, Munoz‐Nunez, et al., 2020). All three reviewers (YG‐F, CRE‐L and AI‐A) had a prior meeting receiving a training seminar and workshop from the arbitrator and creator of the tool (MM‐C) that included education on SDM and the process and application of the SDM tool.", "A descriptive analysis of the characteristics and quality of the selected guidances was conducted. Statistical data analysis was completed using Stata 16. The Kruskal–Wallis test was utilised to compare scores and stratify for factors or characteristics that may influence the quality of SDM in CPGs and CSs. Values were assessed statistically significant when p < 0.05. Inter‐rater reliability between reviewers in data extraction was calculated using the intra‐class correlation coefficient (ICC). A result of more than 0.75 was considered good (Koo & Li, 2016).", "The study selection process is illustrated in the flow diagram in Figure 1. The initial search identified 8229 citations. We removed 439 duplicates and 7676 records for not meeting the selection criteria (inappropriate population, publication, development group or outdated guidance). A total of 114 records were filtered through reviews of titles and abstracts. Later, we obtained the full text of 114 citations for eligibility assessment, and finally, 102 guidances (83 CPGs) (Alberta, 2020; Alsanea et al., 2015; Aranda et al., 2015; Aranda‐Hernandez et al., 2016; Atkin et al., 2012; Austoker et al., 2012; Benton et al., 2016; Bo In Lee et al., 2012; Brenner et al., 2017; Brouwers et al., 2011; Canadian task Force on preventive health C, 2016; Clarke & Feuerstein, 2019; Cubiella et al., 2018; Day et al., 2011; Del Giudice et al., 2014; Duffy et al., 2014; European Colorectal Cancer Screening Guidelines Working Group et al., 2013; European Commission, 2010; Fabio Leonel Gil Parada et al., 2015; Gupta et al., 2019; Halloran et al., 2012; Hassan et al., 2013; Helsingen et al., 2019; Hospital provincial Neuquén, 2016; Instituto Mexicano del Seguro Social. Guía de Práctica Clínica, 2010; Instituto Nacional del Cáncer, 2015; Jenkins et al., 2018; Jenkinson & Steele, 2010; Jover et al., 2012; Kwaan & Jones‐Webb, 2018; Lam et al., 2018; Lansdorp‐Vogelaar et al., 2012; Leddin et al., 2013; Lee et al., 2012; Leong et al., 2017; Lieberman, 2012; Lopes et al., 2018; Malila et al., 2012; Ministry of Health, 2010, 2016; Minozzi et al., 2012; Monahan et al., 2019; Moreno et al., 2018; Moss et al., 2012; Network NCC, 2021; New Brunswick Colon Cancer Screening, 2013; NHS, 2021; Ong et al., 2014; Provenzale et al., 2016; Qaseem et al., 2019; Quirke et al., 2011, 2012; Recommended Cancer Screenings, 2013; Regula & Kaminski, 2010; Rex et al., 2017; Rubeca et al., 2017; Salzman et al., 2016; SemFYC AEdG, 2018; Seppälä et al., 2021; Shaukat et al., 2021; Society AC, 2018; Spada et al., 2014; Steele, Pox et al., 2012; Steele, Rey et al., 2012; Steinwachs et al., 2010; Stoffel et al., 2015; Tanaka et al., 2015; Telford, 2011; Tinmouth et al., 2014; Uruguay MdSd. Ministerio de Salud de Uruguay, 2018; US Preventive Services Task Force et al., 2021; Valori et al., 2012; Vasen et al., 2014; Vieth et al., 2012; von Karsa et al., 2012; Washington KFHPo, 2021; Wilkins et al., 2018; Wilkinson et al., 2019; Wolf et al., 2018; Wong et al., 2015; Yang et al., 2020; Zeimet et al., 2017; 손대경 김, 박윤희, 서민아, 신애선, 이희영, 임종필, 조현민, 홍성필, 김백희, 김용수, 김정욱, 김현수, 남정모, 박동일, 엄준원, 오순남, 임환섭, 장희진, 함상근, 정지혜, 김수영, 김열, 이원철, 정승용, 2015) and 19 CSs (Asociación Mexicana de Endoscopia Gastrointestinal y Colegio de Profesionistas, 2016; ANM. Programa Nacional de Consensos Inter‐Sociedades, 2010; Basu et al., 2021; García‐Carbonero et al., 2017; Giardiello et al., 2014; Hadjiliadis et al., 2018; Heresbach et al., 2016; Hyer et al., 2019; Johnson et al., 2014; Leddin et al., 2010, 2018; Lieberman et al., 2012; Rembacken et al., 2012; Robertson et al., 2017; Schmiegel et al., 2010; Schmoll et al., 2012; Sollano et al., 2017; Sung et al., 2015; 中国抗癌协会肿瘤内镜学专业委员会 国上国中医中中消中中中金国, 2019) were included in our study for the last appraisal.\nThe flow diagram detailing the study selection", "The guidance's characteristics (type of document, entity, country, year, journal of publication, version and evidence analysis) were reported in Table 1. The guidances' mean number of items related to SDM was 1.63 (IQR 0–2). The quality assessment results using the SDM instrument are shown in Figure 2 and Appendix S4. No significant differences were obtained between CPGs and CSs concerning the SDM quality (p = 0.959). A total of 50 guidances (49.0%), 41/83 (49.4%) CPGs and 9/19 (47.4%) CSs, reported something about SDM. None of the guidelines met all the quality criteria, and 51.0% of the guidelines accomplished 0 items, and only 5.9% of them accomplished more than 25% items (8/31). When the SDM term was specifically cited in the guidance (n = 13), the quality of the CPG or CSs concerning SDM was better than when it did not appear (n = 89) (mean 4.5, IQR 2.5–4.5 vs. mean 0.5, IQR 0–1.5; p < 0.001).\nDescription of the screening clinical practice guidelines (CPGs) and consensus (CSs) (n=102) selected for the systematic review on the quality of reporting for SDM\nThe compliance of the items with the SDM quality and reporting analysis tool\nThe best‐scored domains were basic information (domain 1) with a range of guidances accomplishing items from 3 to 50, background (domain 2) with a range from 1 to 15, and recommendations (domain 5) with a range from 5 to 13. Resource implications (domain 8), monitoring and auditing criteria (domain 9), and independence and conflict of interest (domain 11) did not appraise any of the items in any guidance. Only 13/102 (12.7%), 10/102 (9.8%) and 3/102 (2.9%) guidances informed SDM in their executive summary, table of content, glossary, abbreviations, acronyms or topic indexes. SDM concept was only explained in one (1.0%) guidance. Both the primary population and patient subgroups with special consideration were characterised in 15/102 (14.7%). The search strategy and the study design and methodology limitations were reported in 2/102 (2.0%), respectively. The importance of the outcomes and the consistency of the results were detailed in only one guidance (1.0%) each. No PICO question was identified in any guidance. The benefits vs. harms of SDM in CRC screening were considered only in 3/102 (2.9%). The evidence of using SDM was exhibited in 2/102 (2.0%). Recommendations about SDM use were clear, precise and reliable in only 7/102 (6.9%) guidance documents, and these recommendations were well‐reported regarding specific subgroups in 5/102 (4.9%) guidances. Facilitators and barriers for SDM application were well‐described in 4/102 (3.9%), advice on applying SDM in clinical routine in 7/102 (6.9%), and additional materials were provided in other 7/102 (6.9%). Suggestions for further research were located in 2/102 (2.0%), and limitations about SDM recommendations were also described in 2/102 (2.0%). No information about SDM implementation cost, any criteria to assess and measure SDM adherence, conflict of interest regarding SDM or a declaration of the value of SDM in clinical practice. The European Commission (Austoker et al., 2012; European Commission, 2010) guidances and the American Cancer Society (ACS) (Wolf et al., 2018) CPG achieved the highest number of quality SDM items completed (Appendix S4).", "The countries' distribution concerning SDM was erratic. Most of the guidances were from Europe (41/102; %) or North America (40/102; %). Table 2 shows factors than may influence the SDM quality of the guidances. Two CPGs or CSs were from Africa or Oceania (2/102; %). Asia and South America had 12/102 (%) and 5/12 (%), respectively. The quality of SDM did not vary between continents (p = 0.233).\nFactors that may influence the SDM quality and reporting of the CRC screening guidances\nA greater tendency to introduce and recommend SDM was observed in the most recent guidances (Figure 3). The publication year after 2015 had an important influence on the quality than older publications (mean 1, IQR 0–3 vs. mean 0.5, IQR 0–1.5; p = 0.048). The publication in a journal (p = 0.131), and the version number (p = 0.416). The specific quality tool referral increased the quality and reporting of SDM on guidances (p < 0.001). CPGs following systematic reviews had better quality than consensus or literature reviews or when it was not reported (mean 1, IQR 0–3 vs. mean 0, IQR 0–2 vs. mean 0, IQR 0–1; p = 0.040).\nAppearance of SDM concerning the published year of the guidance document", "Our results showed that CPGs and CSs for SDM in CRC screening were of low quality, with variation between guidances and across domains. Recent guidances had better quality, but there is extensive room for improvement. CPGs based on systematic reviews scored better than CSs or guidances that did not report any of it for evidence analysis. Guidances that contained a description of the use of a specific quality tool such as AGREE II or RIGHT demonstrated higher quality.", "To the best of our knowledge, this systematic review is the first to investigate the quality of SDM in CRC screening guidances. One of the main strengths of our review was its comprehensive search based on a broad conceptual framework with no language barriers. SDM is a trendy term of relatively recent appearance. Most methodological recommendation manuals remark a 2‐ to 3‐year window for guidance renovation (Vernooij et al., 2014). However, for studying it, we included more than 10 years of published guidance documents to scrutinise the situation of SDM through time. Our study contained guidance documents of professional organisations from scientifically active nations with more than 0.5% of the global CRC research production.\nA rigorous methodology in conducting systematic reviews is mandatory to guarantee reliable results. In this concern, the trustworthiness of the study selection and the data extraction process is critical. As in similar investigations that use this SDM instrument, there is a possibility of empirical limitations related to the subjectivity of quality data extraction. To minimise this inconvenience, four reviewers performed a preliminary meeting to explain and understand the tool where doubts were solved; and three reviewers worked independently and in duplicate, with double checks included throughout the work. Arbitration was accomplished by a fourth experienced reviewer and creator of the SDM instrument used. The reviewer agreement was good (ICC = 0.88), implying reliable results (Appendix S5).\nOur systematic review aimed to study the quality of SDM. We are conscious that ‘not all the items can have the same relevance and weight’ (Maes‐Carballo, Munoz‐Nunez, et al., 2020). This procedure involving a quality assessment instrument specifies if SDM was cited and which aspects were often considered. Further studies should focus on rating quality.", "CRC early diagnosis could decrease morbimortality by discovering less invasive lesions and permitting more efficient treatments. Furthermore, the debate about the effectiveness and overtreatment due to false‐positive results has appeared on the scene. CRC screening is costly and annoying and could increase the risk of false positives or negatives, which may incur unnecessary stress or procedures and a false sense of security. The mortality reduction is not statistically significant at all ages, and the benefit vs. harm balance is unknown. So, screening should be tailored to the characteristics (age, genetic factors, race, etc.), desires and values of women. Screening programmes are an excellent area for SDM practice as there are different options with similar benefits and harms, and option choice might depend on the patient's values and preferences (Wieringa et al., 2017). The practice of SDM by clinicians could support evidence‐based decisions (Heen et al., 2021) and increase patient satisfaction and treatment engagement (Baca‐Dietz et al., 2020).\nOur systematic review showed that SDM had gained notoriety over the years, with an increasing tendency of SDM presence and recommendations related to them. However, it has also demonstrated that SDM advice merits improvement in all the areas but specifically urgently in the reporting of the SDM resource implications and conflict of interest and the explanation of monitoring and auditing SDM use. It is essential its presence in CPGs and CSs, which hold the potential to influence the care delivered by health‐care providers and the outcomes for patients. Guidances should provide clear and reasonable recommendations for SDM applicability (Rabi et al., 2020; Woolf et al., 1999). More efforts should be made in SDM (Keating & Pace, 2018), and future guidelines should play a more important role in SDM implementation (Gärtner et al., 2019).\nSDM is a new trend, and recent guidances are starting to increase recommendations about basic concepts, evidence and applicability. However, it merits consideration to keep working in that direction. The evidence analysis showed that guidances underpinned by systematic reviews had better quality than consensus or not reported. The referral to SDM term in guidances has shown an improvement in SDM quality, which seems logical as normally improved precision and clearance of recommendations." ]
[ null, null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "METHODS", "Search strategy, data sources, inclusion and exclusion criteria", "Quality appraisal of guidances and data extraction", "Evidence synthesis, investigation of heterogeneity and data analysis", "RESULTS", "Study selection", "Characteristics of guidances and quality appraisal", "Analysis of guidances' characteristics", "DISCUSSION", "Main findings", "Strengths and weaknesses", "Implications", "Conclusions", "CONFLICTS OF INTEREST", "Supporting information" ]
[ "Colorectal cancer (CRC) incidence and mortality have decreased due to screening programmes, removing precancerous polyps with colonoscopy, and advances in management (Cotton et al., 1996; Zauber et al., 2012). In screening programmes, the patient's overall health, prior screening history, and preferences and values must be considered in selecting an individualised approach adapted to the patient's risk of acquiring CRC (Hoffmann et al., 2020; US Preventive Services Task Force et al., 2021). Shared decision‐making (SDM) is essential for decision‐making, weighing up risks vs. benefits for each individual patient (Schrager & Burnside, 2017). In recent years, health‐care policymaking has emphasised SDM as a cornerstone of evidence‐based and patient‐centred care (Barry & Edgman‐Levitan, 2012; Elwyn et al., 2010). Institutional promotion is fundamental for improving SDM application (Senate and House of Representatives, 2010), and clinical practice guidelines (CPGs) and consensus statements (CSs) should promote it and advise about its execution (Maes‐Carballo, Munoz‐Nunez, et al., 2020). Although CPGs and CSs increasingly support it (Gärtner et al., 2019), they remain unclear on accomplishing SDM in routine practice (Elwyn et al., 2012). Therefore, the analysis of the quality of SDM in guidances is critically essential. To the best of our knowledge, no systematic review has investigated SDM in CRC screening guidances.\nConsidering this background, this systematic review aimed to analyze SDM in CRC screening CPGs and CSs, evaluating the quality of recommendations about SDM.", "Following prospective registration (Prospero no: CRD42021286156), this systematic review was conducted following advocated methods for search, assessment and reporting of guidelines using the Preferred Reporting Items for Systematic Reviews and Meta‐Analysis (PRISMA) statement (see Appendix S1) (Liberati et al., 2009; Moher et al., 2009).\n[SUBTITLE] Search strategy, data sources, inclusion and exclusion criteria [SUBSECTION] A search for relevant publications covering major electronic databases (EMBASE, Web of Science, MEDLINE, Scopus and CDSR) was developed without language restrictions to capture peer‐reviewed and grey literature from 2010 until November 2021. We also searched 59 websites of important professional societies and eight guidance‐specific databases. We explored the World Wide Web to include professional societies from countries with global CRC scientific production bigger than 0.5% (Maes‐Carballo et al., 2021, 2022; Maes‐Carballo, Mignini, et al., 2020). A total of 85,932 ‘Colorectal Cancer and Health’ records were scrutinised from Scopus on 10 March 2022 to calculate the scientific production of each country. This decision is adhered to rules followed by other previous peer‐reviewed published systematic reviews (Maes‐Carballo et al., 2021, 2022; Maes‐Carballo, Mignini, et al., 2020; Maes‐Carballo, Munoz‐Nunez, et al., 2020). We revised references from the guidances retrieved to search for conceivable additional CPGs or CSs. The final search integrated MeSH terms ‘practice guidelines’, ‘guidelines’, ‘consensus’, ‘colorectal neoplasms’, ‘colorectal cancer’, ‘screening’ and including word alternatives. Details of the search strategy were documented in Appendix S2. Endnote X9 software was employed to handle the searches downloaded.\nSelection criteria captured eligible guidances about CRC screening produced by national or international professional organisations and societies or governmental agencies. Those guidances in which screening was the central issue and those in which there was a section dedicated to screening or prevention were included in our systematic review. We excluded protocols or screening programme documents, CPGs and CSs about diagnosis and treatment, out‐of‐date guidelines replaced by updates from the same organisation, and CPG and CSs for education and information purposes, randomised controlled trials, observational studies, narrative reviews, scientific reports, discussion papers, conference abstracts, and posters.\nStudies were chosen through a multi‐step approach, including deleting duplicates, reading titles and abstracts, and assessing full texts. Initially, titles and abstracts were considered for eligibility by two reviewers (CR‐E and AI‐A). Then, full texts were appraised for eligibility also by these two reviewers. Potential disagreements or inconsistencies were decided by arbitration of another reviewer (MM‐C). Articles in duplication were recognised and excluded. When several versions of the same guidance were discovered, the most updated version was incorporated. Data were extracted independently and duplicated by three reviewers (YG‐F, CR‐E and AI‐A). Disagreements were solved by consensus or arbitration (MM‐C).\nA search for relevant publications covering major electronic databases (EMBASE, Web of Science, MEDLINE, Scopus and CDSR) was developed without language restrictions to capture peer‐reviewed and grey literature from 2010 until November 2021. We also searched 59 websites of important professional societies and eight guidance‐specific databases. We explored the World Wide Web to include professional societies from countries with global CRC scientific production bigger than 0.5% (Maes‐Carballo et al., 2021, 2022; Maes‐Carballo, Mignini, et al., 2020). A total of 85,932 ‘Colorectal Cancer and Health’ records were scrutinised from Scopus on 10 March 2022 to calculate the scientific production of each country. This decision is adhered to rules followed by other previous peer‐reviewed published systematic reviews (Maes‐Carballo et al., 2021, 2022; Maes‐Carballo, Mignini, et al., 2020; Maes‐Carballo, Munoz‐Nunez, et al., 2020). We revised references from the guidances retrieved to search for conceivable additional CPGs or CSs. The final search integrated MeSH terms ‘practice guidelines’, ‘guidelines’, ‘consensus’, ‘colorectal neoplasms’, ‘colorectal cancer’, ‘screening’ and including word alternatives. Details of the search strategy were documented in Appendix S2. Endnote X9 software was employed to handle the searches downloaded.\nSelection criteria captured eligible guidances about CRC screening produced by national or international professional organisations and societies or governmental agencies. Those guidances in which screening was the central issue and those in which there was a section dedicated to screening or prevention were included in our systematic review. We excluded protocols or screening programme documents, CPGs and CSs about diagnosis and treatment, out‐of‐date guidelines replaced by updates from the same organisation, and CPG and CSs for education and information purposes, randomised controlled trials, observational studies, narrative reviews, scientific reports, discussion papers, conference abstracts, and posters.\nStudies were chosen through a multi‐step approach, including deleting duplicates, reading titles and abstracts, and assessing full texts. Initially, titles and abstracts were considered for eligibility by two reviewers (CR‐E and AI‐A). Then, full texts were appraised for eligibility also by these two reviewers. Potential disagreements or inconsistencies were decided by arbitration of another reviewer (MM‐C). Articles in duplication were recognised and excluded. When several versions of the same guidance were discovered, the most updated version was incorporated. Data were extracted independently and duplicated by three reviewers (YG‐F, CR‐E and AI‐A). Disagreements were solved by consensus or arbitration (MM‐C).\n[SUBTITLE] Quality appraisal of guidances and data extraction [SUBSECTION] The included guidance's characteristics and quality were extracted into a piloted electronic data extraction sheet. The methodological quality assessment was estimated using an already published appraisal tool consisting of a 31‐item checklist grouped into 11 domains (Maes‐Carballo, Munoz‐Nunez, et al., 2020). Three reviewers (YG‐F, CRE‐L and AI‐A) evaluated the quality of SDM in CRC screening guidances (Appendix S3). The 11 domains were basic information (items 1–4), background (items 5–7), selection criteria (items 8–9), strengths and limitations (items 10–14), recommendations about SDM (items 15–17), facilitators and barriers (items 18–19), implementation (items 20–21), resource implications (items 22–24), monitoring and auditing criteria (items 25–27), recommendations for further research and limitations about these recommendations described (items 28–29), and editorial independence and declaration of interest (items 30–31). In those general guidances on the management of CRC, SDM was only considered if it was covered in the screening section. The questions were designed for a binary ‘yes/no’ answer: ‘Yes’ or 1 if the item was met and ‘no’ or ‘0’ if the criterion was not accomplished. The high quality was related to a higher quantity of items completed in the CPG or CS evaluated. No formal score or cut‐off point was specified to determine the quality (Maes‐Carballo, Munoz‐Nunez, et al., 2020). All three reviewers (YG‐F, CRE‐L and AI‐A) had a prior meeting receiving a training seminar and workshop from the arbitrator and creator of the tool (MM‐C) that included education on SDM and the process and application of the SDM tool.\nThe included guidance's characteristics and quality were extracted into a piloted electronic data extraction sheet. The methodological quality assessment was estimated using an already published appraisal tool consisting of a 31‐item checklist grouped into 11 domains (Maes‐Carballo, Munoz‐Nunez, et al., 2020). Three reviewers (YG‐F, CRE‐L and AI‐A) evaluated the quality of SDM in CRC screening guidances (Appendix S3). The 11 domains were basic information (items 1–4), background (items 5–7), selection criteria (items 8–9), strengths and limitations (items 10–14), recommendations about SDM (items 15–17), facilitators and barriers (items 18–19), implementation (items 20–21), resource implications (items 22–24), monitoring and auditing criteria (items 25–27), recommendations for further research and limitations about these recommendations described (items 28–29), and editorial independence and declaration of interest (items 30–31). In those general guidances on the management of CRC, SDM was only considered if it was covered in the screening section. The questions were designed for a binary ‘yes/no’ answer: ‘Yes’ or 1 if the item was met and ‘no’ or ‘0’ if the criterion was not accomplished. The high quality was related to a higher quantity of items completed in the CPG or CS evaluated. No formal score or cut‐off point was specified to determine the quality (Maes‐Carballo, Munoz‐Nunez, et al., 2020). All three reviewers (YG‐F, CRE‐L and AI‐A) had a prior meeting receiving a training seminar and workshop from the arbitrator and creator of the tool (MM‐C) that included education on SDM and the process and application of the SDM tool.\n[SUBTITLE] Evidence synthesis, investigation of heterogeneity and data analysis [SUBSECTION] A descriptive analysis of the characteristics and quality of the selected guidances was conducted. Statistical data analysis was completed using Stata 16. The Kruskal–Wallis test was utilised to compare scores and stratify for factors or characteristics that may influence the quality of SDM in CPGs and CSs. Values were assessed statistically significant when p < 0.05. Inter‐rater reliability between reviewers in data extraction was calculated using the intra‐class correlation coefficient (ICC). A result of more than 0.75 was considered good (Koo & Li, 2016).\nA descriptive analysis of the characteristics and quality of the selected guidances was conducted. Statistical data analysis was completed using Stata 16. The Kruskal–Wallis test was utilised to compare scores and stratify for factors or characteristics that may influence the quality of SDM in CPGs and CSs. Values were assessed statistically significant when p < 0.05. Inter‐rater reliability between reviewers in data extraction was calculated using the intra‐class correlation coefficient (ICC). A result of more than 0.75 was considered good (Koo & Li, 2016).", "A search for relevant publications covering major electronic databases (EMBASE, Web of Science, MEDLINE, Scopus and CDSR) was developed without language restrictions to capture peer‐reviewed and grey literature from 2010 until November 2021. We also searched 59 websites of important professional societies and eight guidance‐specific databases. We explored the World Wide Web to include professional societies from countries with global CRC scientific production bigger than 0.5% (Maes‐Carballo et al., 2021, 2022; Maes‐Carballo, Mignini, et al., 2020). A total of 85,932 ‘Colorectal Cancer and Health’ records were scrutinised from Scopus on 10 March 2022 to calculate the scientific production of each country. This decision is adhered to rules followed by other previous peer‐reviewed published systematic reviews (Maes‐Carballo et al., 2021, 2022; Maes‐Carballo, Mignini, et al., 2020; Maes‐Carballo, Munoz‐Nunez, et al., 2020). We revised references from the guidances retrieved to search for conceivable additional CPGs or CSs. The final search integrated MeSH terms ‘practice guidelines’, ‘guidelines’, ‘consensus’, ‘colorectal neoplasms’, ‘colorectal cancer’, ‘screening’ and including word alternatives. Details of the search strategy were documented in Appendix S2. Endnote X9 software was employed to handle the searches downloaded.\nSelection criteria captured eligible guidances about CRC screening produced by national or international professional organisations and societies or governmental agencies. Those guidances in which screening was the central issue and those in which there was a section dedicated to screening or prevention were included in our systematic review. We excluded protocols or screening programme documents, CPGs and CSs about diagnosis and treatment, out‐of‐date guidelines replaced by updates from the same organisation, and CPG and CSs for education and information purposes, randomised controlled trials, observational studies, narrative reviews, scientific reports, discussion papers, conference abstracts, and posters.\nStudies were chosen through a multi‐step approach, including deleting duplicates, reading titles and abstracts, and assessing full texts. Initially, titles and abstracts were considered for eligibility by two reviewers (CR‐E and AI‐A). Then, full texts were appraised for eligibility also by these two reviewers. Potential disagreements or inconsistencies were decided by arbitration of another reviewer (MM‐C). Articles in duplication were recognised and excluded. When several versions of the same guidance were discovered, the most updated version was incorporated. Data were extracted independently and duplicated by three reviewers (YG‐F, CR‐E and AI‐A). Disagreements were solved by consensus or arbitration (MM‐C).", "The included guidance's characteristics and quality were extracted into a piloted electronic data extraction sheet. The methodological quality assessment was estimated using an already published appraisal tool consisting of a 31‐item checklist grouped into 11 domains (Maes‐Carballo, Munoz‐Nunez, et al., 2020). Three reviewers (YG‐F, CRE‐L and AI‐A) evaluated the quality of SDM in CRC screening guidances (Appendix S3). The 11 domains were basic information (items 1–4), background (items 5–7), selection criteria (items 8–9), strengths and limitations (items 10–14), recommendations about SDM (items 15–17), facilitators and barriers (items 18–19), implementation (items 20–21), resource implications (items 22–24), monitoring and auditing criteria (items 25–27), recommendations for further research and limitations about these recommendations described (items 28–29), and editorial independence and declaration of interest (items 30–31). In those general guidances on the management of CRC, SDM was only considered if it was covered in the screening section. The questions were designed for a binary ‘yes/no’ answer: ‘Yes’ or 1 if the item was met and ‘no’ or ‘0’ if the criterion was not accomplished. The high quality was related to a higher quantity of items completed in the CPG or CS evaluated. No formal score or cut‐off point was specified to determine the quality (Maes‐Carballo, Munoz‐Nunez, et al., 2020). All three reviewers (YG‐F, CRE‐L and AI‐A) had a prior meeting receiving a training seminar and workshop from the arbitrator and creator of the tool (MM‐C) that included education on SDM and the process and application of the SDM tool.", "A descriptive analysis of the characteristics and quality of the selected guidances was conducted. Statistical data analysis was completed using Stata 16. The Kruskal–Wallis test was utilised to compare scores and stratify for factors or characteristics that may influence the quality of SDM in CPGs and CSs. Values were assessed statistically significant when p < 0.05. Inter‐rater reliability between reviewers in data extraction was calculated using the intra‐class correlation coefficient (ICC). A result of more than 0.75 was considered good (Koo & Li, 2016).", "[SUBTITLE] Study selection [SUBSECTION] The study selection process is illustrated in the flow diagram in Figure 1. The initial search identified 8229 citations. We removed 439 duplicates and 7676 records for not meeting the selection criteria (inappropriate population, publication, development group or outdated guidance). A total of 114 records were filtered through reviews of titles and abstracts. Later, we obtained the full text of 114 citations for eligibility assessment, and finally, 102 guidances (83 CPGs) (Alberta, 2020; Alsanea et al., 2015; Aranda et al., 2015; Aranda‐Hernandez et al., 2016; Atkin et al., 2012; Austoker et al., 2012; Benton et al., 2016; Bo In Lee et al., 2012; Brenner et al., 2017; Brouwers et al., 2011; Canadian task Force on preventive health C, 2016; Clarke & Feuerstein, 2019; Cubiella et al., 2018; Day et al., 2011; Del Giudice et al., 2014; Duffy et al., 2014; European Colorectal Cancer Screening Guidelines Working Group et al., 2013; European Commission, 2010; Fabio Leonel Gil Parada et al., 2015; Gupta et al., 2019; Halloran et al., 2012; Hassan et al., 2013; Helsingen et al., 2019; Hospital provincial Neuquén, 2016; Instituto Mexicano del Seguro Social. Guía de Práctica Clínica, 2010; Instituto Nacional del Cáncer, 2015; Jenkins et al., 2018; Jenkinson & Steele, 2010; Jover et al., 2012; Kwaan & Jones‐Webb, 2018; Lam et al., 2018; Lansdorp‐Vogelaar et al., 2012; Leddin et al., 2013; Lee et al., 2012; Leong et al., 2017; Lieberman, 2012; Lopes et al., 2018; Malila et al., 2012; Ministry of Health, 2010, 2016; Minozzi et al., 2012; Monahan et al., 2019; Moreno et al., 2018; Moss et al., 2012; Network NCC, 2021; New Brunswick Colon Cancer Screening, 2013; NHS, 2021; Ong et al., 2014; Provenzale et al., 2016; Qaseem et al., 2019; Quirke et al., 2011, 2012; Recommended Cancer Screenings, 2013; Regula & Kaminski, 2010; Rex et al., 2017; Rubeca et al., 2017; Salzman et al., 2016; SemFYC AEdG, 2018; Seppälä et al., 2021; Shaukat et al., 2021; Society AC, 2018; Spada et al., 2014; Steele, Pox et al., 2012; Steele, Rey et al., 2012; Steinwachs et al., 2010; Stoffel et al., 2015; Tanaka et al., 2015; Telford, 2011; Tinmouth et al., 2014; Uruguay MdSd. Ministerio de Salud de Uruguay, 2018; US Preventive Services Task Force et al., 2021; Valori et al., 2012; Vasen et al., 2014; Vieth et al., 2012; von Karsa et al., 2012; Washington KFHPo, 2021; Wilkins et al., 2018; Wilkinson et al., 2019; Wolf et al., 2018; Wong et al., 2015; Yang et al., 2020; Zeimet et al., 2017; 손대경 김, 박윤희, 서민아, 신애선, 이희영, 임종필, 조현민, 홍성필, 김백희, 김용수, 김정욱, 김현수, 남정모, 박동일, 엄준원, 오순남, 임환섭, 장희진, 함상근, 정지혜, 김수영, 김열, 이원철, 정승용, 2015) and 19 CSs (Asociación Mexicana de Endoscopia Gastrointestinal y Colegio de Profesionistas, 2016; ANM. Programa Nacional de Consensos Inter‐Sociedades, 2010; Basu et al., 2021; García‐Carbonero et al., 2017; Giardiello et al., 2014; Hadjiliadis et al., 2018; Heresbach et al., 2016; Hyer et al., 2019; Johnson et al., 2014; Leddin et al., 2010, 2018; Lieberman et al., 2012; Rembacken et al., 2012; Robertson et al., 2017; Schmiegel et al., 2010; Schmoll et al., 2012; Sollano et al., 2017; Sung et al., 2015; 中国抗癌协会肿瘤内镜学专业委员会 国上国中医中中消中中中金国, 2019) were included in our study for the last appraisal.\nThe flow diagram detailing the study selection\nThe study selection process is illustrated in the flow diagram in Figure 1. The initial search identified 8229 citations. We removed 439 duplicates and 7676 records for not meeting the selection criteria (inappropriate population, publication, development group or outdated guidance). A total of 114 records were filtered through reviews of titles and abstracts. Later, we obtained the full text of 114 citations for eligibility assessment, and finally, 102 guidances (83 CPGs) (Alberta, 2020; Alsanea et al., 2015; Aranda et al., 2015; Aranda‐Hernandez et al., 2016; Atkin et al., 2012; Austoker et al., 2012; Benton et al., 2016; Bo In Lee et al., 2012; Brenner et al., 2017; Brouwers et al., 2011; Canadian task Force on preventive health C, 2016; Clarke & Feuerstein, 2019; Cubiella et al., 2018; Day et al., 2011; Del Giudice et al., 2014; Duffy et al., 2014; European Colorectal Cancer Screening Guidelines Working Group et al., 2013; European Commission, 2010; Fabio Leonel Gil Parada et al., 2015; Gupta et al., 2019; Halloran et al., 2012; Hassan et al., 2013; Helsingen et al., 2019; Hospital provincial Neuquén, 2016; Instituto Mexicano del Seguro Social. Guía de Práctica Clínica, 2010; Instituto Nacional del Cáncer, 2015; Jenkins et al., 2018; Jenkinson & Steele, 2010; Jover et al., 2012; Kwaan & Jones‐Webb, 2018; Lam et al., 2018; Lansdorp‐Vogelaar et al., 2012; Leddin et al., 2013; Lee et al., 2012; Leong et al., 2017; Lieberman, 2012; Lopes et al., 2018; Malila et al., 2012; Ministry of Health, 2010, 2016; Minozzi et al., 2012; Monahan et al., 2019; Moreno et al., 2018; Moss et al., 2012; Network NCC, 2021; New Brunswick Colon Cancer Screening, 2013; NHS, 2021; Ong et al., 2014; Provenzale et al., 2016; Qaseem et al., 2019; Quirke et al., 2011, 2012; Recommended Cancer Screenings, 2013; Regula & Kaminski, 2010; Rex et al., 2017; Rubeca et al., 2017; Salzman et al., 2016; SemFYC AEdG, 2018; Seppälä et al., 2021; Shaukat et al., 2021; Society AC, 2018; Spada et al., 2014; Steele, Pox et al., 2012; Steele, Rey et al., 2012; Steinwachs et al., 2010; Stoffel et al., 2015; Tanaka et al., 2015; Telford, 2011; Tinmouth et al., 2014; Uruguay MdSd. Ministerio de Salud de Uruguay, 2018; US Preventive Services Task Force et al., 2021; Valori et al., 2012; Vasen et al., 2014; Vieth et al., 2012; von Karsa et al., 2012; Washington KFHPo, 2021; Wilkins et al., 2018; Wilkinson et al., 2019; Wolf et al., 2018; Wong et al., 2015; Yang et al., 2020; Zeimet et al., 2017; 손대경 김, 박윤희, 서민아, 신애선, 이희영, 임종필, 조현민, 홍성필, 김백희, 김용수, 김정욱, 김현수, 남정모, 박동일, 엄준원, 오순남, 임환섭, 장희진, 함상근, 정지혜, 김수영, 김열, 이원철, 정승용, 2015) and 19 CSs (Asociación Mexicana de Endoscopia Gastrointestinal y Colegio de Profesionistas, 2016; ANM. Programa Nacional de Consensos Inter‐Sociedades, 2010; Basu et al., 2021; García‐Carbonero et al., 2017; Giardiello et al., 2014; Hadjiliadis et al., 2018; Heresbach et al., 2016; Hyer et al., 2019; Johnson et al., 2014; Leddin et al., 2010, 2018; Lieberman et al., 2012; Rembacken et al., 2012; Robertson et al., 2017; Schmiegel et al., 2010; Schmoll et al., 2012; Sollano et al., 2017; Sung et al., 2015; 中国抗癌协会肿瘤内镜学专业委员会 国上国中医中中消中中中金国, 2019) were included in our study for the last appraisal.\nThe flow diagram detailing the study selection\n[SUBTITLE] Characteristics of guidances and quality appraisal [SUBSECTION] The guidance's characteristics (type of document, entity, country, year, journal of publication, version and evidence analysis) were reported in Table 1. The guidances' mean number of items related to SDM was 1.63 (IQR 0–2). The quality assessment results using the SDM instrument are shown in Figure 2 and Appendix S4. No significant differences were obtained between CPGs and CSs concerning the SDM quality (p = 0.959). A total of 50 guidances (49.0%), 41/83 (49.4%) CPGs and 9/19 (47.4%) CSs, reported something about SDM. None of the guidelines met all the quality criteria, and 51.0% of the guidelines accomplished 0 items, and only 5.9% of them accomplished more than 25% items (8/31). When the SDM term was specifically cited in the guidance (n = 13), the quality of the CPG or CSs concerning SDM was better than when it did not appear (n = 89) (mean 4.5, IQR 2.5–4.5 vs. mean 0.5, IQR 0–1.5; p < 0.001).\nDescription of the screening clinical practice guidelines (CPGs) and consensus (CSs) (n=102) selected for the systematic review on the quality of reporting for SDM\nThe compliance of the items with the SDM quality and reporting analysis tool\nThe best‐scored domains were basic information (domain 1) with a range of guidances accomplishing items from 3 to 50, background (domain 2) with a range from 1 to 15, and recommendations (domain 5) with a range from 5 to 13. Resource implications (domain 8), monitoring and auditing criteria (domain 9), and independence and conflict of interest (domain 11) did not appraise any of the items in any guidance. Only 13/102 (12.7%), 10/102 (9.8%) and 3/102 (2.9%) guidances informed SDM in their executive summary, table of content, glossary, abbreviations, acronyms or topic indexes. SDM concept was only explained in one (1.0%) guidance. Both the primary population and patient subgroups with special consideration were characterised in 15/102 (14.7%). The search strategy and the study design and methodology limitations were reported in 2/102 (2.0%), respectively. The importance of the outcomes and the consistency of the results were detailed in only one guidance (1.0%) each. No PICO question was identified in any guidance. The benefits vs. harms of SDM in CRC screening were considered only in 3/102 (2.9%). The evidence of using SDM was exhibited in 2/102 (2.0%). Recommendations about SDM use were clear, precise and reliable in only 7/102 (6.9%) guidance documents, and these recommendations were well‐reported regarding specific subgroups in 5/102 (4.9%) guidances. Facilitators and barriers for SDM application were well‐described in 4/102 (3.9%), advice on applying SDM in clinical routine in 7/102 (6.9%), and additional materials were provided in other 7/102 (6.9%). Suggestions for further research were located in 2/102 (2.0%), and limitations about SDM recommendations were also described in 2/102 (2.0%). No information about SDM implementation cost, any criteria to assess and measure SDM adherence, conflict of interest regarding SDM or a declaration of the value of SDM in clinical practice. The European Commission (Austoker et al., 2012; European Commission, 2010) guidances and the American Cancer Society (ACS) (Wolf et al., 2018) CPG achieved the highest number of quality SDM items completed (Appendix S4).\nThe guidance's characteristics (type of document, entity, country, year, journal of publication, version and evidence analysis) were reported in Table 1. The guidances' mean number of items related to SDM was 1.63 (IQR 0–2). The quality assessment results using the SDM instrument are shown in Figure 2 and Appendix S4. No significant differences were obtained between CPGs and CSs concerning the SDM quality (p = 0.959). A total of 50 guidances (49.0%), 41/83 (49.4%) CPGs and 9/19 (47.4%) CSs, reported something about SDM. None of the guidelines met all the quality criteria, and 51.0% of the guidelines accomplished 0 items, and only 5.9% of them accomplished more than 25% items (8/31). When the SDM term was specifically cited in the guidance (n = 13), the quality of the CPG or CSs concerning SDM was better than when it did not appear (n = 89) (mean 4.5, IQR 2.5–4.5 vs. mean 0.5, IQR 0–1.5; p < 0.001).\nDescription of the screening clinical practice guidelines (CPGs) and consensus (CSs) (n=102) selected for the systematic review on the quality of reporting for SDM\nThe compliance of the items with the SDM quality and reporting analysis tool\nThe best‐scored domains were basic information (domain 1) with a range of guidances accomplishing items from 3 to 50, background (domain 2) with a range from 1 to 15, and recommendations (domain 5) with a range from 5 to 13. Resource implications (domain 8), monitoring and auditing criteria (domain 9), and independence and conflict of interest (domain 11) did not appraise any of the items in any guidance. Only 13/102 (12.7%), 10/102 (9.8%) and 3/102 (2.9%) guidances informed SDM in their executive summary, table of content, glossary, abbreviations, acronyms or topic indexes. SDM concept was only explained in one (1.0%) guidance. Both the primary population and patient subgroups with special consideration were characterised in 15/102 (14.7%). The search strategy and the study design and methodology limitations were reported in 2/102 (2.0%), respectively. The importance of the outcomes and the consistency of the results were detailed in only one guidance (1.0%) each. No PICO question was identified in any guidance. The benefits vs. harms of SDM in CRC screening were considered only in 3/102 (2.9%). The evidence of using SDM was exhibited in 2/102 (2.0%). Recommendations about SDM use were clear, precise and reliable in only 7/102 (6.9%) guidance documents, and these recommendations were well‐reported regarding specific subgroups in 5/102 (4.9%) guidances. Facilitators and barriers for SDM application were well‐described in 4/102 (3.9%), advice on applying SDM in clinical routine in 7/102 (6.9%), and additional materials were provided in other 7/102 (6.9%). Suggestions for further research were located in 2/102 (2.0%), and limitations about SDM recommendations were also described in 2/102 (2.0%). No information about SDM implementation cost, any criteria to assess and measure SDM adherence, conflict of interest regarding SDM or a declaration of the value of SDM in clinical practice. The European Commission (Austoker et al., 2012; European Commission, 2010) guidances and the American Cancer Society (ACS) (Wolf et al., 2018) CPG achieved the highest number of quality SDM items completed (Appendix S4).\n[SUBTITLE] Analysis of guidances' characteristics [SUBSECTION] The countries' distribution concerning SDM was erratic. Most of the guidances were from Europe (41/102; %) or North America (40/102; %). Table 2 shows factors than may influence the SDM quality of the guidances. Two CPGs or CSs were from Africa or Oceania (2/102; %). Asia and South America had 12/102 (%) and 5/12 (%), respectively. The quality of SDM did not vary between continents (p = 0.233).\nFactors that may influence the SDM quality and reporting of the CRC screening guidances\nA greater tendency to introduce and recommend SDM was observed in the most recent guidances (Figure 3). The publication year after 2015 had an important influence on the quality than older publications (mean 1, IQR 0–3 vs. mean 0.5, IQR 0–1.5; p = 0.048). The publication in a journal (p = 0.131), and the version number (p = 0.416). The specific quality tool referral increased the quality and reporting of SDM on guidances (p < 0.001). CPGs following systematic reviews had better quality than consensus or literature reviews or when it was not reported (mean 1, IQR 0–3 vs. mean 0, IQR 0–2 vs. mean 0, IQR 0–1; p = 0.040).\nAppearance of SDM concerning the published year of the guidance document\nThe countries' distribution concerning SDM was erratic. Most of the guidances were from Europe (41/102; %) or North America (40/102; %). Table 2 shows factors than may influence the SDM quality of the guidances. Two CPGs or CSs were from Africa or Oceania (2/102; %). Asia and South America had 12/102 (%) and 5/12 (%), respectively. The quality of SDM did not vary between continents (p = 0.233).\nFactors that may influence the SDM quality and reporting of the CRC screening guidances\nA greater tendency to introduce and recommend SDM was observed in the most recent guidances (Figure 3). The publication year after 2015 had an important influence on the quality than older publications (mean 1, IQR 0–3 vs. mean 0.5, IQR 0–1.5; p = 0.048). The publication in a journal (p = 0.131), and the version number (p = 0.416). The specific quality tool referral increased the quality and reporting of SDM on guidances (p < 0.001). CPGs following systematic reviews had better quality than consensus or literature reviews or when it was not reported (mean 1, IQR 0–3 vs. mean 0, IQR 0–2 vs. mean 0, IQR 0–1; p = 0.040).\nAppearance of SDM concerning the published year of the guidance document", "The study selection process is illustrated in the flow diagram in Figure 1. The initial search identified 8229 citations. We removed 439 duplicates and 7676 records for not meeting the selection criteria (inappropriate population, publication, development group or outdated guidance). A total of 114 records were filtered through reviews of titles and abstracts. Later, we obtained the full text of 114 citations for eligibility assessment, and finally, 102 guidances (83 CPGs) (Alberta, 2020; Alsanea et al., 2015; Aranda et al., 2015; Aranda‐Hernandez et al., 2016; Atkin et al., 2012; Austoker et al., 2012; Benton et al., 2016; Bo In Lee et al., 2012; Brenner et al., 2017; Brouwers et al., 2011; Canadian task Force on preventive health C, 2016; Clarke & Feuerstein, 2019; Cubiella et al., 2018; Day et al., 2011; Del Giudice et al., 2014; Duffy et al., 2014; European Colorectal Cancer Screening Guidelines Working Group et al., 2013; European Commission, 2010; Fabio Leonel Gil Parada et al., 2015; Gupta et al., 2019; Halloran et al., 2012; Hassan et al., 2013; Helsingen et al., 2019; Hospital provincial Neuquén, 2016; Instituto Mexicano del Seguro Social. Guía de Práctica Clínica, 2010; Instituto Nacional del Cáncer, 2015; Jenkins et al., 2018; Jenkinson & Steele, 2010; Jover et al., 2012; Kwaan & Jones‐Webb, 2018; Lam et al., 2018; Lansdorp‐Vogelaar et al., 2012; Leddin et al., 2013; Lee et al., 2012; Leong et al., 2017; Lieberman, 2012; Lopes et al., 2018; Malila et al., 2012; Ministry of Health, 2010, 2016; Minozzi et al., 2012; Monahan et al., 2019; Moreno et al., 2018; Moss et al., 2012; Network NCC, 2021; New Brunswick Colon Cancer Screening, 2013; NHS, 2021; Ong et al., 2014; Provenzale et al., 2016; Qaseem et al., 2019; Quirke et al., 2011, 2012; Recommended Cancer Screenings, 2013; Regula & Kaminski, 2010; Rex et al., 2017; Rubeca et al., 2017; Salzman et al., 2016; SemFYC AEdG, 2018; Seppälä et al., 2021; Shaukat et al., 2021; Society AC, 2018; Spada et al., 2014; Steele, Pox et al., 2012; Steele, Rey et al., 2012; Steinwachs et al., 2010; Stoffel et al., 2015; Tanaka et al., 2015; Telford, 2011; Tinmouth et al., 2014; Uruguay MdSd. Ministerio de Salud de Uruguay, 2018; US Preventive Services Task Force et al., 2021; Valori et al., 2012; Vasen et al., 2014; Vieth et al., 2012; von Karsa et al., 2012; Washington KFHPo, 2021; Wilkins et al., 2018; Wilkinson et al., 2019; Wolf et al., 2018; Wong et al., 2015; Yang et al., 2020; Zeimet et al., 2017; 손대경 김, 박윤희, 서민아, 신애선, 이희영, 임종필, 조현민, 홍성필, 김백희, 김용수, 김정욱, 김현수, 남정모, 박동일, 엄준원, 오순남, 임환섭, 장희진, 함상근, 정지혜, 김수영, 김열, 이원철, 정승용, 2015) and 19 CSs (Asociación Mexicana de Endoscopia Gastrointestinal y Colegio de Profesionistas, 2016; ANM. Programa Nacional de Consensos Inter‐Sociedades, 2010; Basu et al., 2021; García‐Carbonero et al., 2017; Giardiello et al., 2014; Hadjiliadis et al., 2018; Heresbach et al., 2016; Hyer et al., 2019; Johnson et al., 2014; Leddin et al., 2010, 2018; Lieberman et al., 2012; Rembacken et al., 2012; Robertson et al., 2017; Schmiegel et al., 2010; Schmoll et al., 2012; Sollano et al., 2017; Sung et al., 2015; 中国抗癌协会肿瘤内镜学专业委员会 国上国中医中中消中中中金国, 2019) were included in our study for the last appraisal.\nThe flow diagram detailing the study selection", "The guidance's characteristics (type of document, entity, country, year, journal of publication, version and evidence analysis) were reported in Table 1. The guidances' mean number of items related to SDM was 1.63 (IQR 0–2). The quality assessment results using the SDM instrument are shown in Figure 2 and Appendix S4. No significant differences were obtained between CPGs and CSs concerning the SDM quality (p = 0.959). A total of 50 guidances (49.0%), 41/83 (49.4%) CPGs and 9/19 (47.4%) CSs, reported something about SDM. None of the guidelines met all the quality criteria, and 51.0% of the guidelines accomplished 0 items, and only 5.9% of them accomplished more than 25% items (8/31). When the SDM term was specifically cited in the guidance (n = 13), the quality of the CPG or CSs concerning SDM was better than when it did not appear (n = 89) (mean 4.5, IQR 2.5–4.5 vs. mean 0.5, IQR 0–1.5; p < 0.001).\nDescription of the screening clinical practice guidelines (CPGs) and consensus (CSs) (n=102) selected for the systematic review on the quality of reporting for SDM\nThe compliance of the items with the SDM quality and reporting analysis tool\nThe best‐scored domains were basic information (domain 1) with a range of guidances accomplishing items from 3 to 50, background (domain 2) with a range from 1 to 15, and recommendations (domain 5) with a range from 5 to 13. Resource implications (domain 8), monitoring and auditing criteria (domain 9), and independence and conflict of interest (domain 11) did not appraise any of the items in any guidance. Only 13/102 (12.7%), 10/102 (9.8%) and 3/102 (2.9%) guidances informed SDM in their executive summary, table of content, glossary, abbreviations, acronyms or topic indexes. SDM concept was only explained in one (1.0%) guidance. Both the primary population and patient subgroups with special consideration were characterised in 15/102 (14.7%). The search strategy and the study design and methodology limitations were reported in 2/102 (2.0%), respectively. The importance of the outcomes and the consistency of the results were detailed in only one guidance (1.0%) each. No PICO question was identified in any guidance. The benefits vs. harms of SDM in CRC screening were considered only in 3/102 (2.9%). The evidence of using SDM was exhibited in 2/102 (2.0%). Recommendations about SDM use were clear, precise and reliable in only 7/102 (6.9%) guidance documents, and these recommendations were well‐reported regarding specific subgroups in 5/102 (4.9%) guidances. Facilitators and barriers for SDM application were well‐described in 4/102 (3.9%), advice on applying SDM in clinical routine in 7/102 (6.9%), and additional materials were provided in other 7/102 (6.9%). Suggestions for further research were located in 2/102 (2.0%), and limitations about SDM recommendations were also described in 2/102 (2.0%). No information about SDM implementation cost, any criteria to assess and measure SDM adherence, conflict of interest regarding SDM or a declaration of the value of SDM in clinical practice. The European Commission (Austoker et al., 2012; European Commission, 2010) guidances and the American Cancer Society (ACS) (Wolf et al., 2018) CPG achieved the highest number of quality SDM items completed (Appendix S4).", "The countries' distribution concerning SDM was erratic. Most of the guidances were from Europe (41/102; %) or North America (40/102; %). Table 2 shows factors than may influence the SDM quality of the guidances. Two CPGs or CSs were from Africa or Oceania (2/102; %). Asia and South America had 12/102 (%) and 5/12 (%), respectively. The quality of SDM did not vary between continents (p = 0.233).\nFactors that may influence the SDM quality and reporting of the CRC screening guidances\nA greater tendency to introduce and recommend SDM was observed in the most recent guidances (Figure 3). The publication year after 2015 had an important influence on the quality than older publications (mean 1, IQR 0–3 vs. mean 0.5, IQR 0–1.5; p = 0.048). The publication in a journal (p = 0.131), and the version number (p = 0.416). The specific quality tool referral increased the quality and reporting of SDM on guidances (p < 0.001). CPGs following systematic reviews had better quality than consensus or literature reviews or when it was not reported (mean 1, IQR 0–3 vs. mean 0, IQR 0–2 vs. mean 0, IQR 0–1; p = 0.040).\nAppearance of SDM concerning the published year of the guidance document", "[SUBTITLE] Main findings [SUBSECTION] Our results showed that CPGs and CSs for SDM in CRC screening were of low quality, with variation between guidances and across domains. Recent guidances had better quality, but there is extensive room for improvement. CPGs based on systematic reviews scored better than CSs or guidances that did not report any of it for evidence analysis. Guidances that contained a description of the use of a specific quality tool such as AGREE II or RIGHT demonstrated higher quality.\nOur results showed that CPGs and CSs for SDM in CRC screening were of low quality, with variation between guidances and across domains. Recent guidances had better quality, but there is extensive room for improvement. CPGs based on systematic reviews scored better than CSs or guidances that did not report any of it for evidence analysis. Guidances that contained a description of the use of a specific quality tool such as AGREE II or RIGHT demonstrated higher quality.\n[SUBTITLE] Strengths and weaknesses [SUBSECTION] To the best of our knowledge, this systematic review is the first to investigate the quality of SDM in CRC screening guidances. One of the main strengths of our review was its comprehensive search based on a broad conceptual framework with no language barriers. SDM is a trendy term of relatively recent appearance. Most methodological recommendation manuals remark a 2‐ to 3‐year window for guidance renovation (Vernooij et al., 2014). However, for studying it, we included more than 10 years of published guidance documents to scrutinise the situation of SDM through time. Our study contained guidance documents of professional organisations from scientifically active nations with more than 0.5% of the global CRC research production.\nA rigorous methodology in conducting systematic reviews is mandatory to guarantee reliable results. In this concern, the trustworthiness of the study selection and the data extraction process is critical. As in similar investigations that use this SDM instrument, there is a possibility of empirical limitations related to the subjectivity of quality data extraction. To minimise this inconvenience, four reviewers performed a preliminary meeting to explain and understand the tool where doubts were solved; and three reviewers worked independently and in duplicate, with double checks included throughout the work. Arbitration was accomplished by a fourth experienced reviewer and creator of the SDM instrument used. The reviewer agreement was good (ICC = 0.88), implying reliable results (Appendix S5).\nOur systematic review aimed to study the quality of SDM. We are conscious that ‘not all the items can have the same relevance and weight’ (Maes‐Carballo, Munoz‐Nunez, et al., 2020). This procedure involving a quality assessment instrument specifies if SDM was cited and which aspects were often considered. Further studies should focus on rating quality.\nTo the best of our knowledge, this systematic review is the first to investigate the quality of SDM in CRC screening guidances. One of the main strengths of our review was its comprehensive search based on a broad conceptual framework with no language barriers. SDM is a trendy term of relatively recent appearance. Most methodological recommendation manuals remark a 2‐ to 3‐year window for guidance renovation (Vernooij et al., 2014). However, for studying it, we included more than 10 years of published guidance documents to scrutinise the situation of SDM through time. Our study contained guidance documents of professional organisations from scientifically active nations with more than 0.5% of the global CRC research production.\nA rigorous methodology in conducting systematic reviews is mandatory to guarantee reliable results. In this concern, the trustworthiness of the study selection and the data extraction process is critical. As in similar investigations that use this SDM instrument, there is a possibility of empirical limitations related to the subjectivity of quality data extraction. To minimise this inconvenience, four reviewers performed a preliminary meeting to explain and understand the tool where doubts were solved; and three reviewers worked independently and in duplicate, with double checks included throughout the work. Arbitration was accomplished by a fourth experienced reviewer and creator of the SDM instrument used. The reviewer agreement was good (ICC = 0.88), implying reliable results (Appendix S5).\nOur systematic review aimed to study the quality of SDM. We are conscious that ‘not all the items can have the same relevance and weight’ (Maes‐Carballo, Munoz‐Nunez, et al., 2020). This procedure involving a quality assessment instrument specifies if SDM was cited and which aspects were often considered. Further studies should focus on rating quality.\n[SUBTITLE] Implications [SUBSECTION] CRC early diagnosis could decrease morbimortality by discovering less invasive lesions and permitting more efficient treatments. Furthermore, the debate about the effectiveness and overtreatment due to false‐positive results has appeared on the scene. CRC screening is costly and annoying and could increase the risk of false positives or negatives, which may incur unnecessary stress or procedures and a false sense of security. The mortality reduction is not statistically significant at all ages, and the benefit vs. harm balance is unknown. So, screening should be tailored to the characteristics (age, genetic factors, race, etc.), desires and values of women. Screening programmes are an excellent area for SDM practice as there are different options with similar benefits and harms, and option choice might depend on the patient's values and preferences (Wieringa et al., 2017). The practice of SDM by clinicians could support evidence‐based decisions (Heen et al., 2021) and increase patient satisfaction and treatment engagement (Baca‐Dietz et al., 2020).\nOur systematic review showed that SDM had gained notoriety over the years, with an increasing tendency of SDM presence and recommendations related to them. However, it has also demonstrated that SDM advice merits improvement in all the areas but specifically urgently in the reporting of the SDM resource implications and conflict of interest and the explanation of monitoring and auditing SDM use. It is essential its presence in CPGs and CSs, which hold the potential to influence the care delivered by health‐care providers and the outcomes for patients. Guidances should provide clear and reasonable recommendations for SDM applicability (Rabi et al., 2020; Woolf et al., 1999). More efforts should be made in SDM (Keating & Pace, 2018), and future guidelines should play a more important role in SDM implementation (Gärtner et al., 2019).\nSDM is a new trend, and recent guidances are starting to increase recommendations about basic concepts, evidence and applicability. However, it merits consideration to keep working in that direction. The evidence analysis showed that guidances underpinned by systematic reviews had better quality than consensus or not reported. The referral to SDM term in guidances has shown an improvement in SDM quality, which seems logical as normally improved precision and clearance of recommendations.\nCRC early diagnosis could decrease morbimortality by discovering less invasive lesions and permitting more efficient treatments. Furthermore, the debate about the effectiveness and overtreatment due to false‐positive results has appeared on the scene. CRC screening is costly and annoying and could increase the risk of false positives or negatives, which may incur unnecessary stress or procedures and a false sense of security. The mortality reduction is not statistically significant at all ages, and the benefit vs. harm balance is unknown. So, screening should be tailored to the characteristics (age, genetic factors, race, etc.), desires and values of women. Screening programmes are an excellent area for SDM practice as there are different options with similar benefits and harms, and option choice might depend on the patient's values and preferences (Wieringa et al., 2017). The practice of SDM by clinicians could support evidence‐based decisions (Heen et al., 2021) and increase patient satisfaction and treatment engagement (Baca‐Dietz et al., 2020).\nOur systematic review showed that SDM had gained notoriety over the years, with an increasing tendency of SDM presence and recommendations related to them. However, it has also demonstrated that SDM advice merits improvement in all the areas but specifically urgently in the reporting of the SDM resource implications and conflict of interest and the explanation of monitoring and auditing SDM use. It is essential its presence in CPGs and CSs, which hold the potential to influence the care delivered by health‐care providers and the outcomes for patients. Guidances should provide clear and reasonable recommendations for SDM applicability (Rabi et al., 2020; Woolf et al., 1999). More efforts should be made in SDM (Keating & Pace, 2018), and future guidelines should play a more important role in SDM implementation (Gärtner et al., 2019).\nSDM is a new trend, and recent guidances are starting to increase recommendations about basic concepts, evidence and applicability. However, it merits consideration to keep working in that direction. The evidence analysis showed that guidances underpinned by systematic reviews had better quality than consensus or not reported. The referral to SDM term in guidances has shown an improvement in SDM quality, which seems logical as normally improved precision and clearance of recommendations.\n[SUBTITLE] Conclusions [SUBSECTION] This systematic review demonstrated that SDM quality in guidance documents was suboptimal as it did not appear in half of the guidances analyzed. SDM recommendations were scarce and unclear. Recent guidances following systematic reviews and referring to quality tools (e.g. AGREE II or RIGHT) had better SDM quality. Therefore, guideline developers, professional institutions, medical journals and policymakers should consider including evidence‐based SDM recommendations in trustworthy and well‐developed guidances to ensure proper translation of evidence into practice.\nThis systematic review demonstrated that SDM quality in guidance documents was suboptimal as it did not appear in half of the guidances analyzed. SDM recommendations were scarce and unclear. Recent guidances following systematic reviews and referring to quality tools (e.g. AGREE II or RIGHT) had better SDM quality. Therefore, guideline developers, professional institutions, medical journals and policymakers should consider including evidence‐based SDM recommendations in trustworthy and well‐developed guidances to ensure proper translation of evidence into practice.", "Our results showed that CPGs and CSs for SDM in CRC screening were of low quality, with variation between guidances and across domains. Recent guidances had better quality, but there is extensive room for improvement. CPGs based on systematic reviews scored better than CSs or guidances that did not report any of it for evidence analysis. Guidances that contained a description of the use of a specific quality tool such as AGREE II or RIGHT demonstrated higher quality.", "To the best of our knowledge, this systematic review is the first to investigate the quality of SDM in CRC screening guidances. One of the main strengths of our review was its comprehensive search based on a broad conceptual framework with no language barriers. SDM is a trendy term of relatively recent appearance. Most methodological recommendation manuals remark a 2‐ to 3‐year window for guidance renovation (Vernooij et al., 2014). However, for studying it, we included more than 10 years of published guidance documents to scrutinise the situation of SDM through time. Our study contained guidance documents of professional organisations from scientifically active nations with more than 0.5% of the global CRC research production.\nA rigorous methodology in conducting systematic reviews is mandatory to guarantee reliable results. In this concern, the trustworthiness of the study selection and the data extraction process is critical. As in similar investigations that use this SDM instrument, there is a possibility of empirical limitations related to the subjectivity of quality data extraction. To minimise this inconvenience, four reviewers performed a preliminary meeting to explain and understand the tool where doubts were solved; and three reviewers worked independently and in duplicate, with double checks included throughout the work. Arbitration was accomplished by a fourth experienced reviewer and creator of the SDM instrument used. The reviewer agreement was good (ICC = 0.88), implying reliable results (Appendix S5).\nOur systematic review aimed to study the quality of SDM. We are conscious that ‘not all the items can have the same relevance and weight’ (Maes‐Carballo, Munoz‐Nunez, et al., 2020). This procedure involving a quality assessment instrument specifies if SDM was cited and which aspects were often considered. Further studies should focus on rating quality.", "CRC early diagnosis could decrease morbimortality by discovering less invasive lesions and permitting more efficient treatments. Furthermore, the debate about the effectiveness and overtreatment due to false‐positive results has appeared on the scene. CRC screening is costly and annoying and could increase the risk of false positives or negatives, which may incur unnecessary stress or procedures and a false sense of security. The mortality reduction is not statistically significant at all ages, and the benefit vs. harm balance is unknown. So, screening should be tailored to the characteristics (age, genetic factors, race, etc.), desires and values of women. Screening programmes are an excellent area for SDM practice as there are different options with similar benefits and harms, and option choice might depend on the patient's values and preferences (Wieringa et al., 2017). The practice of SDM by clinicians could support evidence‐based decisions (Heen et al., 2021) and increase patient satisfaction and treatment engagement (Baca‐Dietz et al., 2020).\nOur systematic review showed that SDM had gained notoriety over the years, with an increasing tendency of SDM presence and recommendations related to them. However, it has also demonstrated that SDM advice merits improvement in all the areas but specifically urgently in the reporting of the SDM resource implications and conflict of interest and the explanation of monitoring and auditing SDM use. It is essential its presence in CPGs and CSs, which hold the potential to influence the care delivered by health‐care providers and the outcomes for patients. Guidances should provide clear and reasonable recommendations for SDM applicability (Rabi et al., 2020; Woolf et al., 1999). More efforts should be made in SDM (Keating & Pace, 2018), and future guidelines should play a more important role in SDM implementation (Gärtner et al., 2019).\nSDM is a new trend, and recent guidances are starting to increase recommendations about basic concepts, evidence and applicability. However, it merits consideration to keep working in that direction. The evidence analysis showed that guidances underpinned by systematic reviews had better quality than consensus or not reported. The referral to SDM term in guidances has shown an improvement in SDM quality, which seems logical as normally improved precision and clearance of recommendations.", "This systematic review demonstrated that SDM quality in guidance documents was suboptimal as it did not appear in half of the guidances analyzed. SDM recommendations were scarce and unclear. Recent guidances following systematic reviews and referring to quality tools (e.g. AGREE II or RIGHT) had better SDM quality. Therefore, guideline developers, professional institutions, medical journals and policymakers should consider including evidence‐based SDM recommendations in trustworthy and well‐developed guidances to ensure proper translation of evidence into practice.", "There are no conflicts of interest.", "\nAppendix S0: PRISMA 2020 Checklist\nClick here for additional data file.\n\nAppendix S1: Data sources and search strategy.\nClick here for additional data file.\n\nAppendix S2: Quality assessment tool for Shared Decision Making (SDM) recommendations in Breast Cancer Management Clinical Practice Guidelines (CPG) and consensus (CS).\nClick here for additional data file.\n\nAppendix S3: Quality assessment tool for Shared Decision Making (SDM) recommendations in Breast Cancer Management Clinical Practice Guidelines (CPG) and consensus (CS).\nClick here for additional data file.\n\nAppendix S4: Data extraction analysis.\nClick here for additional data file.\n\nAppendix S5: The reviewer agreement calculated using the intra‐class correlation coefficient (corr).\nClick here for additional data file." ]
[ null, "methods", null, null, null, "results", null, null, null, "discussion", null, null, null, "conclusions", "COI-statement", "supplementary-material" ]
[ "‘clinical practice guidelines’", "‘colorectal cancer screening’", "‘consensus’", "‘quality of guidelines’", "‘shared decision‐making’" ]
Prevalence and prognostic impact of hepatopulmonary syndrome in patients with unresectable hepatocellular carcinoma undergoing transarterial chemoembolization: a prospective cohort study.
36255217
To determine the prevalence and prognostic impact of hepatopulmonary syndrome (HPS) in patients with unresectable hepatocellular carcinoma (HCC) undergoing transarterial chemoembolization (TACE).
BACKGROUND
Fifty-four patients with unresectable HCC undergoing TACE between December 2014 and December 2015 were prospectively screened for HPS and were followed up for a maximum of 2 years or until the end of this prospective study.
METHODS
Nineteen of the 54 (35.2%) patients were considered to have HPS, including one (5.3%) with severe HPS, nine (47.4%) with moderate HPS, and nine (47.4%) with mild HPS. The median overall survival (OS) was 10.1 (95% confidence interval [CI], 3.9-16.3) months for patients with HPS and 15.1 (95% CI, 7.3-22.9) months for patients without HPS, which is not a significant difference ( P  = 0.100). The median progression-free survival was also not significantly different between patients with and without HPS (5.2 [95% CI, 0-12.8] vs. 8.4 [95% CI, 3.6-13.1] months; P  = 0.537). In the multivariable Cox regression analyses, carbon monoxide diffusing capacity (hazard ratio [HR] = 1.033 [95% CI, 1.003-1.064]; P  = 0.028) and Child-Pugh class (HR = 1.815 [95% CI, 1.011-3.260]; P  = 0.046) were identified to be the independent prognostic factors of OS.
RESULTS
Mild or moderate HPS is common in patients with unresectable HCC undergoing TACE, but it does not seem to have a significant prognostic impact.
CONCLUSION
[ "Humans", "Carcinoma, Hepatocellular", "Chemoembolization, Therapeutic", "Prospective Studies", "Liver Neoplasms", "Prognosis", "Hepatopulmonary Syndrome", "Prevalence", "Treatment Outcome", "Retrospective Studies" ]
9746741
Introduction
Hepatocellular carcinoma (HCC) is the seventh most common malignancy and the fourth leading cause of cancer-related mortality worldwide.[1,2] Approximately 80% to 85% of patients with HCC have unresectable disease at present and therefore, curative treatment options are available to only 15% to 20% of patients.[3,4] Transarterial chemoembolization (TACE) is the most widely used palliative treatment option for unresectable HCC.[5] TACE is effective in controlling disease progression as the blood supply for HCC preferentially originates from the hepatic artery.[6,7] However, since HCC usually arises in the setting of chronic liver disease, it is therefore not uncommon for HCC patients to present with major complications of liver dysfunction and/or portal hypertension (eg, ascites, variceal bleeding, and hepatic encephalopathy), which may negatively affect prognosis and treatment outcomes.[7] Hepatopulmonary syndrome (HPS) is a common pulmonary complication of liver disease and/or portal hypertension and is defined as an arterial oxygenation defect induced by intra-pulmonary vascular dilation (IPVD).[8] HPS has been mostly reported in cirrhotic patients, with a prevalence of 12% to 32% in those evaluated for liver transplantation (LT) or transjugular intra-hepatic portosystemic shunt (TIPS) creation,[8] and negatively affects the prognosis of patients with cirrhosis.[9] HPS has also been reported in patients with unresectable HCC, but the prevalence and prognostic impact of HPS in this population remain unknown.[10] Consequently, whether HPS screening and treatment are necessary for patients with unresectable HCC undergoing TACE is still unclear. The purpose of this study was to assess the prevalence and the prognostic impact of HPS in patients with unresectable HCC undergoing TACE.
TACE procedure
With the patient under local anesthesia, right femoral access was obtained, and angiography of the superior mesenteric and common hepatic arteries was performed using a 5-Fr Rösch hepatic catheter (Radiofocus; Terumo, Tokyo, Japan). A 3-Fr microcatheter (MicroFerret; Cook, Indiana, USA) was coaxially placed into the proper, lobar, segmental, or subsegmental hepatic artery, and 500 to 1000 mg 5-fluorouracil was infused through the microcatheter for 10 to 15 min. Subsequently, 20 to 40 mg doxorubicin mixed at a 1:1 ratio in an emulsion of iodized oil (Lipiodol; Guerbet, France) was infused, which was followed by embolization with gelatin sponge pledgets (Gelfoam; Upjohn, Missouri, USA) until stasis or near stasis of arterial flow was achieved. Hemostasis at the access site was achieved by manual compression.
Results
[SUBTITLE] Prevalence of HPS and clinical characteristics of the patients [SUBSECTION] A total of 60 patients with HCC undergoing TACE were eligible for inclusion in this study [Figure 1]. Six of these patients were excluded from the study due to previous TIPS creation (n = 2), discontinued treatment (n = 1), a poor echocardiographic window (n = 1), and intra-cardiac shunt (n = 2). The remaining 54 patients were included in the study and were screened for HPS. Among these patients, 31 (57.4%) had a positive CEE result. Of these patients, 19 (61.3%) had an elevated AaO2. These patients were considered to have HPS, including one (5.3%) with severe HPS, nine (47.4%) with moderate HPS, and nine (47.4%) with mild HPS. Therefore, the prevalence of HPS was 35.2% (19/54). Flow chart of the patient selection and HPS screening processes. AaO2: Alveolar-arterial oxygen gradient; CEE: Contrast-enhanced echocardiography; HPS: Hepatopulmonary syndrome; TIPS: Transjugular intra-hepatic portosystemic shunt. The demographics, etiology of liver disease, cirrhosis status, Child-Pugh class, performance status, tumor characteristics, and history of prior treatments were not significantly different between those with and without HPS (all P > 0.050) [Table 1]. The number of patients who complained of dyspnea was significantly higher in the group with HPS than in the group without HPS (8/19 vs. 4/35; P = 0.016). However, the number of patients who had cyanosis (3/19 vs. 2/35; P = 0.332) and digital clubbing (2/19 vs. 2/35; P = 0.607) was not significantly different between those with and without HPS. Significantly more patients with HPS had spider angioma than those without HPS (8/19 vs. 3/35; P = 0.010). Clinical characteristics of patients with and without HPS. Data are presented as mean ± standard deviation or n(%). Student's t test. Fisher's exact test. Pearson chi-square. AaO2: Alveolar-arterial oxygen gradient; ABG: Arterial blood gas; AFP: Alpha-fetoprotein; DLCO: Carbon monoxide diffusing capacity; ECOG: European Cooperative Oncology Group; FEV1: Forced expiratory volume in 1 s; FVC: Forced vital capacity; HBV: Hepatitis V virus; HCV: Hepatitis C virus; HE: Hepatic encephalopathy; HPS: Hepatopulmonary syndrome; PaCO2: Partial arterial pressure of carbon dioxide; PaO2: Partial arterial pressure of oxygen; SaO2: Arterial oxygen saturation; TACE: Transcatheter arterial chemoembolization; TLC: Total lung capacity. Patients with HPS had significantly higher AaO2 values (26.1 ± 9.3 vs. 15.5 ± 11.6 mmHg; P = 0.001), significantly lower arterial oxygen saturation (95.4 ± 2.5 vs. 96.8 ± 1.7; P = 0.019), and PaO2 values (76.8 ± 10.0 vs. 85.0 ± 12.0 mmHg; P = 0.015) than those without HPS. However, the partial arterial pressure of carbon dioxide was not significantly different between patients with and without HPS (37.4 ± 4.1 vs. 39.4 ± 4.2 mmHg; P = 0.104). In addition, the carbon monoxide diffusing capacity (DLCO) was the only pulmonary function test result that was significantly different between patients with and without HPS (83.3 ± 14.2% predicted vs. 93.1 ± 15.2% predicted; P = 0.032). A total of 60 patients with HCC undergoing TACE were eligible for inclusion in this study [Figure 1]. Six of these patients were excluded from the study due to previous TIPS creation (n = 2), discontinued treatment (n = 1), a poor echocardiographic window (n = 1), and intra-cardiac shunt (n = 2). The remaining 54 patients were included in the study and were screened for HPS. Among these patients, 31 (57.4%) had a positive CEE result. Of these patients, 19 (61.3%) had an elevated AaO2. These patients were considered to have HPS, including one (5.3%) with severe HPS, nine (47.4%) with moderate HPS, and nine (47.4%) with mild HPS. Therefore, the prevalence of HPS was 35.2% (19/54). Flow chart of the patient selection and HPS screening processes. AaO2: Alveolar-arterial oxygen gradient; CEE: Contrast-enhanced echocardiography; HPS: Hepatopulmonary syndrome; TIPS: Transjugular intra-hepatic portosystemic shunt. The demographics, etiology of liver disease, cirrhosis status, Child-Pugh class, performance status, tumor characteristics, and history of prior treatments were not significantly different between those with and without HPS (all P > 0.050) [Table 1]. The number of patients who complained of dyspnea was significantly higher in the group with HPS than in the group without HPS (8/19 vs. 4/35; P = 0.016). However, the number of patients who had cyanosis (3/19 vs. 2/35; P = 0.332) and digital clubbing (2/19 vs. 2/35; P = 0.607) was not significantly different between those with and without HPS. Significantly more patients with HPS had spider angioma than those without HPS (8/19 vs. 3/35; P = 0.010). Clinical characteristics of patients with and without HPS. Data are presented as mean ± standard deviation or n(%). Student's t test. Fisher's exact test. Pearson chi-square. AaO2: Alveolar-arterial oxygen gradient; ABG: Arterial blood gas; AFP: Alpha-fetoprotein; DLCO: Carbon monoxide diffusing capacity; ECOG: European Cooperative Oncology Group; FEV1: Forced expiratory volume in 1 s; FVC: Forced vital capacity; HBV: Hepatitis V virus; HCV: Hepatitis C virus; HE: Hepatic encephalopathy; HPS: Hepatopulmonary syndrome; PaCO2: Partial arterial pressure of carbon dioxide; PaO2: Partial arterial pressure of oxygen; SaO2: Arterial oxygen saturation; TACE: Transcatheter arterial chemoembolization; TLC: Total lung capacity. Patients with HPS had significantly higher AaO2 values (26.1 ± 9.3 vs. 15.5 ± 11.6 mmHg; P = 0.001), significantly lower arterial oxygen saturation (95.4 ± 2.5 vs. 96.8 ± 1.7; P = 0.019), and PaO2 values (76.8 ± 10.0 vs. 85.0 ± 12.0 mmHg; P = 0.015) than those without HPS. However, the partial arterial pressure of carbon dioxide was not significantly different between patients with and without HPS (37.4 ± 4.1 vs. 39.4 ± 4.2 mmHg; P = 0.104). In addition, the carbon monoxide diffusing capacity (DLCO) was the only pulmonary function test result that was significantly different between patients with and without HPS (83.3 ± 14.2% predicted vs. 93.1 ± 15.2% predicted; P = 0.032). [SUBTITLE] Clinical outcome of TACE for HCC according to HPS status [SUBSECTION] The median interval from HCC diagnosis to study entry was not significantly different between patients with and without HPS (8.5 [95% CI, 3.2–13.8] vs. 3.9 [95% CI, 0–9.9] months; P = 0.613). A total of 172 TACE sessions were performed for the patients during the study, with no significant difference between those with and without HPS (3.1 ± 1.5 sessions per patient vs. 3.3 ± 1.6 sessions per patient; P = 0.549). No major complications, defined according to the Society of Interventional Radiology clinical practice guidelines, were observed in any patients.[18] Five (9.3%) patients, including one with HPS and four without HPS, were lost to or refused the follow-up after 3.3, 2.5, 3.8, 5.2, and 5.3 months. The median overall survival (OS) was not significantly different between patients with and without HPS (12.0 [95% CI, 8.2–15.7] vs. 14.9 [95% CI, 11.9–17.9] months; P = 0.223). The tumor response was not significantly different between patients with and without HPS (P = 0.748). For patients with HPS, complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD) were observed in 2 (10.5%), 8 (42.1%), 1 (5.3%), and 8 (42.1%) patients, respectively. For patients without HPS, CR, PR, SD, and PD were observed in 7 (20.0%), 15 (42.9%), 1 (2.9%), and 12 (17.1%) patients, respectively. Of the patients with PD, 18 (90%, 18/20) died before the 6-month evaluation, including 6 of the 19 (31.6%) patients with HPS and 12 of the 35 (34.3%) patients without HPS. The 6-month overall response rates (ie, the percentage of patients in whom CR or PR was observed) for patients with and without HPS were 53.8% (7/13) and 78.3% (18/23), respectively, and the difference was not significantly different (P = 0.153). A total of 32 (59.3%) patients died at the end of the study, mainly due to disease progression (87.5% [28/32]), with no significant difference of mortality between patients with and without HPS (73.7% [14/19] vs. 51.4% [18/35]; P = 0.112). The median OS was 10.1 (95% CI, 3.9–16.3) months for patients with HPS and 15.1 (95% CI, 7.3–22.9) months for patients without HPS, which was not a significant difference (P = 0.100) [Figure 2]. The median PFS was also not significantly different between patients with and without HPS (5.2 [95% CI, 0–12.8] vs. 8.4 [95% CI, 3.6–13.1] months; P = 0.537) [Figure 3]. In the multivariate Cox regression analyses, DLCO (hazard ratio [HR] = 1.033 [95% CI, 1.003–1.064]; P = 0.028) and Child-Pugh class (HR = 1.815 [95% CI, 1.011–3.260]; P = 0.046) were identified to be the independent prognostic factors of OS [Table 2]. Kaplan-Meier curve of OS according to HPS status. HPS: Hepatopulmonary syndrome; OS: Overall survival. Kaplan-Meier curve of PFS according to HPS status. HPS: Hepatopulmonary syndrome; PFS: Progression-free survival. Multivariate cox regression analyses to identify the prognostic factors of OS. AaO2: Alveolar-arterial oxygen gradient; DLCO: Carbon monoxide diffusing capacity; HR: Hazard ratio; HPS: Hepatopulmonary syndrome; OS: Overall survival. The median interval from HCC diagnosis to study entry was not significantly different between patients with and without HPS (8.5 [95% CI, 3.2–13.8] vs. 3.9 [95% CI, 0–9.9] months; P = 0.613). A total of 172 TACE sessions were performed for the patients during the study, with no significant difference between those with and without HPS (3.1 ± 1.5 sessions per patient vs. 3.3 ± 1.6 sessions per patient; P = 0.549). No major complications, defined according to the Society of Interventional Radiology clinical practice guidelines, were observed in any patients.[18] Five (9.3%) patients, including one with HPS and four without HPS, were lost to or refused the follow-up after 3.3, 2.5, 3.8, 5.2, and 5.3 months. The median overall survival (OS) was not significantly different between patients with and without HPS (12.0 [95% CI, 8.2–15.7] vs. 14.9 [95% CI, 11.9–17.9] months; P = 0.223). The tumor response was not significantly different between patients with and without HPS (P = 0.748). For patients with HPS, complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD) were observed in 2 (10.5%), 8 (42.1%), 1 (5.3%), and 8 (42.1%) patients, respectively. For patients without HPS, CR, PR, SD, and PD were observed in 7 (20.0%), 15 (42.9%), 1 (2.9%), and 12 (17.1%) patients, respectively. Of the patients with PD, 18 (90%, 18/20) died before the 6-month evaluation, including 6 of the 19 (31.6%) patients with HPS and 12 of the 35 (34.3%) patients without HPS. The 6-month overall response rates (ie, the percentage of patients in whom CR or PR was observed) for patients with and without HPS were 53.8% (7/13) and 78.3% (18/23), respectively, and the difference was not significantly different (P = 0.153). A total of 32 (59.3%) patients died at the end of the study, mainly due to disease progression (87.5% [28/32]), with no significant difference of mortality between patients with and without HPS (73.7% [14/19] vs. 51.4% [18/35]; P = 0.112). The median OS was 10.1 (95% CI, 3.9–16.3) months for patients with HPS and 15.1 (95% CI, 7.3–22.9) months for patients without HPS, which was not a significant difference (P = 0.100) [Figure 2]. The median PFS was also not significantly different between patients with and without HPS (5.2 [95% CI, 0–12.8] vs. 8.4 [95% CI, 3.6–13.1] months; P = 0.537) [Figure 3]. In the multivariate Cox regression analyses, DLCO (hazard ratio [HR] = 1.033 [95% CI, 1.003–1.064]; P = 0.028) and Child-Pugh class (HR = 1.815 [95% CI, 1.011–3.260]; P = 0.046) were identified to be the independent prognostic factors of OS [Table 2]. Kaplan-Meier curve of OS according to HPS status. HPS: Hepatopulmonary syndrome; OS: Overall survival. Kaplan-Meier curve of PFS according to HPS status. HPS: Hepatopulmonary syndrome; PFS: Progression-free survival. Multivariate cox regression analyses to identify the prognostic factors of OS. AaO2: Alveolar-arterial oxygen gradient; DLCO: Carbon monoxide diffusing capacity; HR: Hazard ratio; HPS: Hepatopulmonary syndrome; OS: Overall survival.
null
null
[ "Methods", "Study design", "Follow-up", "Statistical analysis", "Clinical outcome of TACE for HCC according to HPS status", "Funding" ]
[ "[SUBTITLE] Study design [SUBSECTION] This prospective cohort study was approved by the institutional review board of West China Hospital (Identifier, 2014–249), and written informed consent was obtained from all patients. All patients with unresectable HCC undergoing TACE at a single institution between December 2014 and December 2015 were eligible for inclusion in this study and were screened for HPS. The diagnosis of HCC was established by clinical, laboratory, imaging, and when necessary, histologic findings. The diagnosis of HPS in patients with HCC was established by a positive contrast-enhanced echocardiography (CEE) result (ie, the appearance of microbubbles in the left heart chambers within 3 to 6 heartbeats after their initial appearance in the right heart chambers) and an elevated alveolar-arterial oxygen gradient (AaO2)(≥15 or ≥20 mmHg if age ≥65 years).[8,11] CEE was performed using 10 mL agitated saline and transthoracic echocardiography in the parasternal four-chamber view as described previously.[12,13] The appearance of any microbubble in the left heart chambers <3 heartbeats after their initial appearance in the right heart chambers indicates the presence of an intra-cardiac shunt. Arterial blood gas sampling was performed while the patient was in an upright position and was breathing room air. The severity of HPS was categorized as very severe (partial arterial pressure of oxygen [PaO2] <50 mmHg), severe (PaO2 50–59 mmHg), moderate (PaO2 60–79 mmHg), or mild (PaO2 ≥80 mmHg), as described previously.[14,15] The exclusion criteria were age <18 years, uncontrolled infection, other known malignancies, intracardiac shunts, heart failure, pulmonary hypertension, pregnancy, and previous TIPS creation.\nThis prospective cohort study was approved by the institutional review board of West China Hospital (Identifier, 2014–249), and written informed consent was obtained from all patients. All patients with unresectable HCC undergoing TACE at a single institution between December 2014 and December 2015 were eligible for inclusion in this study and were screened for HPS. The diagnosis of HCC was established by clinical, laboratory, imaging, and when necessary, histologic findings. The diagnosis of HPS in patients with HCC was established by a positive contrast-enhanced echocardiography (CEE) result (ie, the appearance of microbubbles in the left heart chambers within 3 to 6 heartbeats after their initial appearance in the right heart chambers) and an elevated alveolar-arterial oxygen gradient (AaO2)(≥15 or ≥20 mmHg if age ≥65 years).[8,11] CEE was performed using 10 mL agitated saline and transthoracic echocardiography in the parasternal four-chamber view as described previously.[12,13] The appearance of any microbubble in the left heart chambers <3 heartbeats after their initial appearance in the right heart chambers indicates the presence of an intra-cardiac shunt. Arterial blood gas sampling was performed while the patient was in an upright position and was breathing room air. The severity of HPS was categorized as very severe (partial arterial pressure of oxygen [PaO2] <50 mmHg), severe (PaO2 50–59 mmHg), moderate (PaO2 60–79 mmHg), or mild (PaO2 ≥80 mmHg), as described previously.[14,15] The exclusion criteria were age <18 years, uncontrolled infection, other known malignancies, intracardiac shunts, heart failure, pulmonary hypertension, pregnancy, and previous TIPS creation.\n[SUBTITLE] TACE procedure [SUBSECTION] With the patient under local anesthesia, right femoral access was obtained, and angiography of the superior mesenteric and common hepatic arteries was performed using a 5-Fr Rösch hepatic catheter (Radiofocus; Terumo, Tokyo, Japan). A 3-Fr microcatheter (MicroFerret; Cook, Indiana, USA) was coaxially placed into the proper, lobar, segmental, or subsegmental hepatic artery, and 500 to 1000 mg 5-fluorouracil was infused through the microcatheter for 10 to 15 min. Subsequently, 20 to 40 mg doxorubicin mixed at a 1:1 ratio in an emulsion of iodized oil (Lipiodol; Guerbet, France) was infused, which was followed by embolization with gelatin sponge pledgets (Gelfoam; Upjohn, Missouri, USA) until stasis or near stasis of arterial flow was achieved. Hemostasis at the access site was achieved by manual compression.\nWith the patient under local anesthesia, right femoral access was obtained, and angiography of the superior mesenteric and common hepatic arteries was performed using a 5-Fr Rösch hepatic catheter (Radiofocus; Terumo, Tokyo, Japan). A 3-Fr microcatheter (MicroFerret; Cook, Indiana, USA) was coaxially placed into the proper, lobar, segmental, or subsegmental hepatic artery, and 500 to 1000 mg 5-fluorouracil was infused through the microcatheter for 10 to 15 min. Subsequently, 20 to 40 mg doxorubicin mixed at a 1:1 ratio in an emulsion of iodized oil (Lipiodol; Guerbet, France) was infused, which was followed by embolization with gelatin sponge pledgets (Gelfoam; Upjohn, Missouri, USA) until stasis or near stasis of arterial flow was achieved. Hemostasis at the access site was achieved by manual compression.\n[SUBTITLE] Follow-up [SUBSECTION] Follow-up visits were scheduled every 4 to 12 weeks until death up to a maximum of 2 years or until the end of the study (June 2017). Contrast-enhanced computed tomography or magnetic resonance imaging was performed at each follow-up visit to evaluate the tumor response (ie, the best overall response) by modified Response Evaluation Criteria in Solid Tumors Criteria.[16] Progression-free survival (PFS) was defined as the interval from study entry to disease progression, death from any cause, or when the patient was lost to or refused follow-up, up to a maximum of 2 years or until the end of the study.[17] Overall survival (OS) was defined as the interval from study entry to death from any cause or when the patient was lost to or refused follow-up, up to a maximum of 2 years or until the end of the study. Repeat TACE was performed every 4 to 8 weeks as needed if residual or new tumor tissue was detected at follow-up.\nFollow-up visits were scheduled every 4 to 12 weeks until death up to a maximum of 2 years or until the end of the study (June 2017). Contrast-enhanced computed tomography or magnetic resonance imaging was performed at each follow-up visit to evaluate the tumor response (ie, the best overall response) by modified Response Evaluation Criteria in Solid Tumors Criteria.[16] Progression-free survival (PFS) was defined as the interval from study entry to disease progression, death from any cause, or when the patient was lost to or refused follow-up, up to a maximum of 2 years or until the end of the study.[17] Overall survival (OS) was defined as the interval from study entry to death from any cause or when the patient was lost to or refused follow-up, up to a maximum of 2 years or until the end of the study. Repeat TACE was performed every 4 to 8 weeks as needed if residual or new tumor tissue was detected at follow-up.\n[SUBTITLE] Statistical analysis [SUBSECTION] Continuous variables were compared using Student's t-test or Mann–Whitney U test, as appropriate. Categorical variables were compared using the chi-squared or Fisher's exact test. The time-to-event distributions were estimated using the Kaplan–Meier method and compared using the log-rank test. According to the results of univariate analyses and clinical relevance, variables were included in the multivariate Cox regression analyses to identify independent prognostic factors. All analyses were performed using SPSS software (version 19.0; SPSS, Chicago, IL, USA). A two-sided P value <0.05 was considered statistically significant.\nContinuous variables were compared using Student's t-test or Mann–Whitney U test, as appropriate. Categorical variables were compared using the chi-squared or Fisher's exact test. The time-to-event distributions were estimated using the Kaplan–Meier method and compared using the log-rank test. According to the results of univariate analyses and clinical relevance, variables were included in the multivariate Cox regression analyses to identify independent prognostic factors. All analyses were performed using SPSS software (version 19.0; SPSS, Chicago, IL, USA). A two-sided P value <0.05 was considered statistically significant.", "This prospective cohort study was approved by the institutional review board of West China Hospital (Identifier, 2014–249), and written informed consent was obtained from all patients. All patients with unresectable HCC undergoing TACE at a single institution between December 2014 and December 2015 were eligible for inclusion in this study and were screened for HPS. The diagnosis of HCC was established by clinical, laboratory, imaging, and when necessary, histologic findings. The diagnosis of HPS in patients with HCC was established by a positive contrast-enhanced echocardiography (CEE) result (ie, the appearance of microbubbles in the left heart chambers within 3 to 6 heartbeats after their initial appearance in the right heart chambers) and an elevated alveolar-arterial oxygen gradient (AaO2)(≥15 or ≥20 mmHg if age ≥65 years).[8,11] CEE was performed using 10 mL agitated saline and transthoracic echocardiography in the parasternal four-chamber view as described previously.[12,13] The appearance of any microbubble in the left heart chambers <3 heartbeats after their initial appearance in the right heart chambers indicates the presence of an intra-cardiac shunt. Arterial blood gas sampling was performed while the patient was in an upright position and was breathing room air. The severity of HPS was categorized as very severe (partial arterial pressure of oxygen [PaO2] <50 mmHg), severe (PaO2 50–59 mmHg), moderate (PaO2 60–79 mmHg), or mild (PaO2 ≥80 mmHg), as described previously.[14,15] The exclusion criteria were age <18 years, uncontrolled infection, other known malignancies, intracardiac shunts, heart failure, pulmonary hypertension, pregnancy, and previous TIPS creation.", "Follow-up visits were scheduled every 4 to 12 weeks until death up to a maximum of 2 years or until the end of the study (June 2017). Contrast-enhanced computed tomography or magnetic resonance imaging was performed at each follow-up visit to evaluate the tumor response (ie, the best overall response) by modified Response Evaluation Criteria in Solid Tumors Criteria.[16] Progression-free survival (PFS) was defined as the interval from study entry to disease progression, death from any cause, or when the patient was lost to or refused follow-up, up to a maximum of 2 years or until the end of the study.[17] Overall survival (OS) was defined as the interval from study entry to death from any cause or when the patient was lost to or refused follow-up, up to a maximum of 2 years or until the end of the study. Repeat TACE was performed every 4 to 8 weeks as needed if residual or new tumor tissue was detected at follow-up.", "Continuous variables were compared using Student's t-test or Mann–Whitney U test, as appropriate. Categorical variables were compared using the chi-squared or Fisher's exact test. The time-to-event distributions were estimated using the Kaplan–Meier method and compared using the log-rank test. According to the results of univariate analyses and clinical relevance, variables were included in the multivariate Cox regression analyses to identify independent prognostic factors. All analyses were performed using SPSS software (version 19.0; SPSS, Chicago, IL, USA). A two-sided P value <0.05 was considered statistically significant.", "The median interval from HCC diagnosis to study entry was not significantly different between patients with and without HPS (8.5 [95% CI, 3.2–13.8] vs. 3.9 [95% CI, 0–9.9] months; P = 0.613). A total of 172 TACE sessions were performed for the patients during the study, with no significant difference between those with and without HPS (3.1 ± 1.5 sessions per patient vs. 3.3 ± 1.6 sessions per patient; P = 0.549). No major complications, defined according to the Society of Interventional Radiology clinical practice guidelines, were observed in any patients.[18] Five (9.3%) patients, including one with HPS and four without HPS, were lost to or refused the follow-up after 3.3, 2.5, 3.8, 5.2, and 5.3 months. The median overall survival (OS) was not significantly different between patients with and without HPS (12.0 [95% CI, 8.2–15.7] vs. 14.9 [95% CI, 11.9–17.9] months; P = 0.223).\nThe tumor response was not significantly different between patients with and without HPS (P = 0.748). For patients with HPS, complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD) were observed in 2 (10.5%), 8 (42.1%), 1 (5.3%), and 8 (42.1%) patients, respectively. For patients without HPS, CR, PR, SD, and PD were observed in 7 (20.0%), 15 (42.9%), 1 (2.9%), and 12 (17.1%) patients, respectively. Of the patients with PD, 18 (90%, 18/20) died before the 6-month evaluation, including 6 of the 19 (31.6%) patients with HPS and 12 of the 35 (34.3%) patients without HPS. The 6-month overall response rates (ie, the percentage of patients in whom CR or PR was observed) for patients with and without HPS were 53.8% (7/13) and 78.3% (18/23), respectively, and the difference was not significantly different (P = 0.153).\nA total of 32 (59.3%) patients died at the end of the study, mainly due to disease progression (87.5% [28/32]), with no significant difference of mortality between patients with and without HPS (73.7% [14/19] vs. 51.4% [18/35]; P = 0.112). The median OS was 10.1 (95% CI, 3.9–16.3) months for patients with HPS and 15.1 (95% CI, 7.3–22.9) months for patients without HPS, which was not a significant difference (P = 0.100) [Figure 2]. The median PFS was also not significantly different between patients with and without HPS (5.2 [95% CI, 0–12.8] vs. 8.4 [95% CI, 3.6–13.1] months; P = 0.537) [Figure 3]. In the multivariate Cox regression analyses, DLCO (hazard ratio [HR] = 1.033 [95% CI, 1.003–1.064]; P = 0.028) and Child-Pugh class (HR = 1.815 [95% CI, 1.011–3.260]; P = 0.046) were identified to be the independent prognostic factors of OS [Table 2].\nKaplan-Meier curve of OS according to HPS status. HPS: Hepatopulmonary syndrome; OS: Overall survival.\nKaplan-Meier curve of PFS according to HPS status. HPS: Hepatopulmonary syndrome; PFS: Progression-free survival.\nMultivariate cox regression analyses to identify the prognostic factors of OS.\nAaO2: Alveolar-arterial oxygen gradient; DLCO: Carbon monoxide diffusing capacity; HR: Hazard ratio; HPS: Hepatopulmonary syndrome; OS: Overall survival.", "This work was supported by the National Natural Science Fund of China (No. 82001937) and the CAMS Initiative for Innovative Medicine (No. 2021-I2M-1-015)." ]
[ "methods", null, null, null, null, null ]
[ "Introduction", "Methods", "Study design", "TACE procedure", "Follow-up", "Statistical analysis", "Results", "Prevalence of HPS and clinical characteristics of the patients", "Clinical outcome of TACE for HCC according to HPS status", "Discussion", "Funding", "Conflicts of interest" ]
[ "Hepatocellular carcinoma (HCC) is the seventh most common malignancy and the fourth leading cause of cancer-related mortality worldwide.[1,2] Approximately 80% to 85% of patients with HCC have unresectable disease at present and therefore, curative treatment options are available to only 15% to 20% of patients.[3,4] Transarterial chemoembolization (TACE) is the most widely used palliative treatment option for unresectable HCC.[5] TACE is effective in controlling disease progression as the blood supply for HCC preferentially originates from the hepatic artery.[6,7] However, since HCC usually arises in the setting of chronic liver disease, it is therefore not uncommon for HCC patients to present with major complications of liver dysfunction and/or portal hypertension (eg, ascites, variceal bleeding, and hepatic encephalopathy), which may negatively affect prognosis and treatment outcomes.[7]\nHepatopulmonary syndrome (HPS) is a common pulmonary complication of liver disease and/or portal hypertension and is defined as an arterial oxygenation defect induced by intra-pulmonary vascular dilation (IPVD).[8] HPS has been mostly reported in cirrhotic patients, with a prevalence of 12% to 32% in those evaluated for liver transplantation (LT) or transjugular intra-hepatic portosystemic shunt (TIPS) creation,[8] and negatively affects the prognosis of patients with cirrhosis.[9] HPS has also been reported in patients with unresectable HCC, but the prevalence and prognostic impact of HPS in this population remain unknown.[10] Consequently, whether HPS screening and treatment are necessary for patients with unresectable HCC undergoing TACE is still unclear. The purpose of this study was to assess the prevalence and the prognostic impact of HPS in patients with unresectable HCC undergoing TACE.", "[SUBTITLE] Study design [SUBSECTION] This prospective cohort study was approved by the institutional review board of West China Hospital (Identifier, 2014–249), and written informed consent was obtained from all patients. All patients with unresectable HCC undergoing TACE at a single institution between December 2014 and December 2015 were eligible for inclusion in this study and were screened for HPS. The diagnosis of HCC was established by clinical, laboratory, imaging, and when necessary, histologic findings. The diagnosis of HPS in patients with HCC was established by a positive contrast-enhanced echocardiography (CEE) result (ie, the appearance of microbubbles in the left heart chambers within 3 to 6 heartbeats after their initial appearance in the right heart chambers) and an elevated alveolar-arterial oxygen gradient (AaO2)(≥15 or ≥20 mmHg if age ≥65 years).[8,11] CEE was performed using 10 mL agitated saline and transthoracic echocardiography in the parasternal four-chamber view as described previously.[12,13] The appearance of any microbubble in the left heart chambers <3 heartbeats after their initial appearance in the right heart chambers indicates the presence of an intra-cardiac shunt. Arterial blood gas sampling was performed while the patient was in an upright position and was breathing room air. The severity of HPS was categorized as very severe (partial arterial pressure of oxygen [PaO2] <50 mmHg), severe (PaO2 50–59 mmHg), moderate (PaO2 60–79 mmHg), or mild (PaO2 ≥80 mmHg), as described previously.[14,15] The exclusion criteria were age <18 years, uncontrolled infection, other known malignancies, intracardiac shunts, heart failure, pulmonary hypertension, pregnancy, and previous TIPS creation.\nThis prospective cohort study was approved by the institutional review board of West China Hospital (Identifier, 2014–249), and written informed consent was obtained from all patients. All patients with unresectable HCC undergoing TACE at a single institution between December 2014 and December 2015 were eligible for inclusion in this study and were screened for HPS. The diagnosis of HCC was established by clinical, laboratory, imaging, and when necessary, histologic findings. The diagnosis of HPS in patients with HCC was established by a positive contrast-enhanced echocardiography (CEE) result (ie, the appearance of microbubbles in the left heart chambers within 3 to 6 heartbeats after their initial appearance in the right heart chambers) and an elevated alveolar-arterial oxygen gradient (AaO2)(≥15 or ≥20 mmHg if age ≥65 years).[8,11] CEE was performed using 10 mL agitated saline and transthoracic echocardiography in the parasternal four-chamber view as described previously.[12,13] The appearance of any microbubble in the left heart chambers <3 heartbeats after their initial appearance in the right heart chambers indicates the presence of an intra-cardiac shunt. Arterial blood gas sampling was performed while the patient was in an upright position and was breathing room air. The severity of HPS was categorized as very severe (partial arterial pressure of oxygen [PaO2] <50 mmHg), severe (PaO2 50–59 mmHg), moderate (PaO2 60–79 mmHg), or mild (PaO2 ≥80 mmHg), as described previously.[14,15] The exclusion criteria were age <18 years, uncontrolled infection, other known malignancies, intracardiac shunts, heart failure, pulmonary hypertension, pregnancy, and previous TIPS creation.\n[SUBTITLE] TACE procedure [SUBSECTION] With the patient under local anesthesia, right femoral access was obtained, and angiography of the superior mesenteric and common hepatic arteries was performed using a 5-Fr Rösch hepatic catheter (Radiofocus; Terumo, Tokyo, Japan). A 3-Fr microcatheter (MicroFerret; Cook, Indiana, USA) was coaxially placed into the proper, lobar, segmental, or subsegmental hepatic artery, and 500 to 1000 mg 5-fluorouracil was infused through the microcatheter for 10 to 15 min. Subsequently, 20 to 40 mg doxorubicin mixed at a 1:1 ratio in an emulsion of iodized oil (Lipiodol; Guerbet, France) was infused, which was followed by embolization with gelatin sponge pledgets (Gelfoam; Upjohn, Missouri, USA) until stasis or near stasis of arterial flow was achieved. Hemostasis at the access site was achieved by manual compression.\nWith the patient under local anesthesia, right femoral access was obtained, and angiography of the superior mesenteric and common hepatic arteries was performed using a 5-Fr Rösch hepatic catheter (Radiofocus; Terumo, Tokyo, Japan). A 3-Fr microcatheter (MicroFerret; Cook, Indiana, USA) was coaxially placed into the proper, lobar, segmental, or subsegmental hepatic artery, and 500 to 1000 mg 5-fluorouracil was infused through the microcatheter for 10 to 15 min. Subsequently, 20 to 40 mg doxorubicin mixed at a 1:1 ratio in an emulsion of iodized oil (Lipiodol; Guerbet, France) was infused, which was followed by embolization with gelatin sponge pledgets (Gelfoam; Upjohn, Missouri, USA) until stasis or near stasis of arterial flow was achieved. Hemostasis at the access site was achieved by manual compression.\n[SUBTITLE] Follow-up [SUBSECTION] Follow-up visits were scheduled every 4 to 12 weeks until death up to a maximum of 2 years or until the end of the study (June 2017). Contrast-enhanced computed tomography or magnetic resonance imaging was performed at each follow-up visit to evaluate the tumor response (ie, the best overall response) by modified Response Evaluation Criteria in Solid Tumors Criteria.[16] Progression-free survival (PFS) was defined as the interval from study entry to disease progression, death from any cause, or when the patient was lost to or refused follow-up, up to a maximum of 2 years or until the end of the study.[17] Overall survival (OS) was defined as the interval from study entry to death from any cause or when the patient was lost to or refused follow-up, up to a maximum of 2 years or until the end of the study. Repeat TACE was performed every 4 to 8 weeks as needed if residual or new tumor tissue was detected at follow-up.\nFollow-up visits were scheduled every 4 to 12 weeks until death up to a maximum of 2 years or until the end of the study (June 2017). Contrast-enhanced computed tomography or magnetic resonance imaging was performed at each follow-up visit to evaluate the tumor response (ie, the best overall response) by modified Response Evaluation Criteria in Solid Tumors Criteria.[16] Progression-free survival (PFS) was defined as the interval from study entry to disease progression, death from any cause, or when the patient was lost to or refused follow-up, up to a maximum of 2 years or until the end of the study.[17] Overall survival (OS) was defined as the interval from study entry to death from any cause or when the patient was lost to or refused follow-up, up to a maximum of 2 years or until the end of the study. Repeat TACE was performed every 4 to 8 weeks as needed if residual or new tumor tissue was detected at follow-up.\n[SUBTITLE] Statistical analysis [SUBSECTION] Continuous variables were compared using Student's t-test or Mann–Whitney U test, as appropriate. Categorical variables were compared using the chi-squared or Fisher's exact test. The time-to-event distributions were estimated using the Kaplan–Meier method and compared using the log-rank test. According to the results of univariate analyses and clinical relevance, variables were included in the multivariate Cox regression analyses to identify independent prognostic factors. All analyses were performed using SPSS software (version 19.0; SPSS, Chicago, IL, USA). A two-sided P value <0.05 was considered statistically significant.\nContinuous variables were compared using Student's t-test or Mann–Whitney U test, as appropriate. Categorical variables were compared using the chi-squared or Fisher's exact test. The time-to-event distributions were estimated using the Kaplan–Meier method and compared using the log-rank test. According to the results of univariate analyses and clinical relevance, variables were included in the multivariate Cox regression analyses to identify independent prognostic factors. All analyses were performed using SPSS software (version 19.0; SPSS, Chicago, IL, USA). A two-sided P value <0.05 was considered statistically significant.", "This prospective cohort study was approved by the institutional review board of West China Hospital (Identifier, 2014–249), and written informed consent was obtained from all patients. All patients with unresectable HCC undergoing TACE at a single institution between December 2014 and December 2015 were eligible for inclusion in this study and were screened for HPS. The diagnosis of HCC was established by clinical, laboratory, imaging, and when necessary, histologic findings. The diagnosis of HPS in patients with HCC was established by a positive contrast-enhanced echocardiography (CEE) result (ie, the appearance of microbubbles in the left heart chambers within 3 to 6 heartbeats after their initial appearance in the right heart chambers) and an elevated alveolar-arterial oxygen gradient (AaO2)(≥15 or ≥20 mmHg if age ≥65 years).[8,11] CEE was performed using 10 mL agitated saline and transthoracic echocardiography in the parasternal four-chamber view as described previously.[12,13] The appearance of any microbubble in the left heart chambers <3 heartbeats after their initial appearance in the right heart chambers indicates the presence of an intra-cardiac shunt. Arterial blood gas sampling was performed while the patient was in an upright position and was breathing room air. The severity of HPS was categorized as very severe (partial arterial pressure of oxygen [PaO2] <50 mmHg), severe (PaO2 50–59 mmHg), moderate (PaO2 60–79 mmHg), or mild (PaO2 ≥80 mmHg), as described previously.[14,15] The exclusion criteria were age <18 years, uncontrolled infection, other known malignancies, intracardiac shunts, heart failure, pulmonary hypertension, pregnancy, and previous TIPS creation.", "With the patient under local anesthesia, right femoral access was obtained, and angiography of the superior mesenteric and common hepatic arteries was performed using a 5-Fr Rösch hepatic catheter (Radiofocus; Terumo, Tokyo, Japan). A 3-Fr microcatheter (MicroFerret; Cook, Indiana, USA) was coaxially placed into the proper, lobar, segmental, or subsegmental hepatic artery, and 500 to 1000 mg 5-fluorouracil was infused through the microcatheter for 10 to 15 min. Subsequently, 20 to 40 mg doxorubicin mixed at a 1:1 ratio in an emulsion of iodized oil (Lipiodol; Guerbet, France) was infused, which was followed by embolization with gelatin sponge pledgets (Gelfoam; Upjohn, Missouri, USA) until stasis or near stasis of arterial flow was achieved. Hemostasis at the access site was achieved by manual compression.", "Follow-up visits were scheduled every 4 to 12 weeks until death up to a maximum of 2 years or until the end of the study (June 2017). Contrast-enhanced computed tomography or magnetic resonance imaging was performed at each follow-up visit to evaluate the tumor response (ie, the best overall response) by modified Response Evaluation Criteria in Solid Tumors Criteria.[16] Progression-free survival (PFS) was defined as the interval from study entry to disease progression, death from any cause, or when the patient was lost to or refused follow-up, up to a maximum of 2 years or until the end of the study.[17] Overall survival (OS) was defined as the interval from study entry to death from any cause or when the patient was lost to or refused follow-up, up to a maximum of 2 years or until the end of the study. Repeat TACE was performed every 4 to 8 weeks as needed if residual or new tumor tissue was detected at follow-up.", "Continuous variables were compared using Student's t-test or Mann–Whitney U test, as appropriate. Categorical variables were compared using the chi-squared or Fisher's exact test. The time-to-event distributions were estimated using the Kaplan–Meier method and compared using the log-rank test. According to the results of univariate analyses and clinical relevance, variables were included in the multivariate Cox regression analyses to identify independent prognostic factors. All analyses were performed using SPSS software (version 19.0; SPSS, Chicago, IL, USA). A two-sided P value <0.05 was considered statistically significant.", "[SUBTITLE] Prevalence of HPS and clinical characteristics of the patients [SUBSECTION] A total of 60 patients with HCC undergoing TACE were eligible for inclusion in this study [Figure 1]. Six of these patients were excluded from the study due to previous TIPS creation (n = 2), discontinued treatment (n = 1), a poor echocardiographic window (n = 1), and intra-cardiac shunt (n = 2). The remaining 54 patients were included in the study and were screened for HPS. Among these patients, 31 (57.4%) had a positive CEE result. Of these patients, 19 (61.3%) had an elevated AaO2. These patients were considered to have HPS, including one (5.3%) with severe HPS, nine (47.4%) with moderate HPS, and nine (47.4%) with mild HPS. Therefore, the prevalence of HPS was 35.2% (19/54).\nFlow chart of the patient selection and HPS screening processes. AaO2: Alveolar-arterial oxygen gradient; CEE: Contrast-enhanced echocardiography; HPS: Hepatopulmonary syndrome; TIPS: Transjugular intra-hepatic portosystemic shunt.\nThe demographics, etiology of liver disease, cirrhosis status, Child-Pugh class, performance status, tumor characteristics, and history of prior treatments were not significantly different between those with and without HPS (all P > 0.050) [Table 1]. The number of patients who complained of dyspnea was significantly higher in the group with HPS than in the group without HPS (8/19 vs. 4/35; P = 0.016). However, the number of patients who had cyanosis (3/19 vs. 2/35; P = 0.332) and digital clubbing (2/19 vs. 2/35; P = 0.607) was not significantly different between those with and without HPS. Significantly more patients with HPS had spider angioma than those without HPS (8/19 vs. 3/35; P = 0.010).\nClinical characteristics of patients with and without HPS.\nData are presented as mean ± standard deviation or n(%).\nStudent's t test.\nFisher's exact test.\nPearson chi-square. AaO2: Alveolar-arterial oxygen gradient; ABG: Arterial blood gas; AFP: Alpha-fetoprotein; DLCO: Carbon monoxide diffusing capacity; ECOG: European Cooperative Oncology Group; FEV1: Forced expiratory volume in 1 s; FVC: Forced vital capacity; HBV: Hepatitis V virus; HCV: Hepatitis C virus; HE: Hepatic encephalopathy; HPS: Hepatopulmonary syndrome; PaCO2: Partial arterial pressure of carbon dioxide; PaO2: Partial arterial pressure of oxygen; SaO2: Arterial oxygen saturation; TACE: Transcatheter arterial chemoembolization; TLC: Total lung capacity.\nPatients with HPS had significantly higher AaO2 values (26.1 ± 9.3 vs. 15.5 ± 11.6 mmHg; P = 0.001), significantly lower arterial oxygen saturation (95.4 ± 2.5 vs. 96.8 ± 1.7; P = 0.019), and PaO2 values (76.8 ± 10.0 vs. 85.0 ± 12.0 mmHg; P = 0.015) than those without HPS. However, the partial arterial pressure of carbon dioxide was not significantly different between patients with and without HPS (37.4 ± 4.1 vs. 39.4 ± 4.2 mmHg; P = 0.104). In addition, the carbon monoxide diffusing capacity (DLCO) was the only pulmonary function test result that was significantly different between patients with and without HPS (83.3 ± 14.2% predicted vs. 93.1 ± 15.2% predicted; P = 0.032).\nA total of 60 patients with HCC undergoing TACE were eligible for inclusion in this study [Figure 1]. Six of these patients were excluded from the study due to previous TIPS creation (n = 2), discontinued treatment (n = 1), a poor echocardiographic window (n = 1), and intra-cardiac shunt (n = 2). The remaining 54 patients were included in the study and were screened for HPS. Among these patients, 31 (57.4%) had a positive CEE result. Of these patients, 19 (61.3%) had an elevated AaO2. These patients were considered to have HPS, including one (5.3%) with severe HPS, nine (47.4%) with moderate HPS, and nine (47.4%) with mild HPS. Therefore, the prevalence of HPS was 35.2% (19/54).\nFlow chart of the patient selection and HPS screening processes. AaO2: Alveolar-arterial oxygen gradient; CEE: Contrast-enhanced echocardiography; HPS: Hepatopulmonary syndrome; TIPS: Transjugular intra-hepatic portosystemic shunt.\nThe demographics, etiology of liver disease, cirrhosis status, Child-Pugh class, performance status, tumor characteristics, and history of prior treatments were not significantly different between those with and without HPS (all P > 0.050) [Table 1]. The number of patients who complained of dyspnea was significantly higher in the group with HPS than in the group without HPS (8/19 vs. 4/35; P = 0.016). However, the number of patients who had cyanosis (3/19 vs. 2/35; P = 0.332) and digital clubbing (2/19 vs. 2/35; P = 0.607) was not significantly different between those with and without HPS. Significantly more patients with HPS had spider angioma than those without HPS (8/19 vs. 3/35; P = 0.010).\nClinical characteristics of patients with and without HPS.\nData are presented as mean ± standard deviation or n(%).\nStudent's t test.\nFisher's exact test.\nPearson chi-square. AaO2: Alveolar-arterial oxygen gradient; ABG: Arterial blood gas; AFP: Alpha-fetoprotein; DLCO: Carbon monoxide diffusing capacity; ECOG: European Cooperative Oncology Group; FEV1: Forced expiratory volume in 1 s; FVC: Forced vital capacity; HBV: Hepatitis V virus; HCV: Hepatitis C virus; HE: Hepatic encephalopathy; HPS: Hepatopulmonary syndrome; PaCO2: Partial arterial pressure of carbon dioxide; PaO2: Partial arterial pressure of oxygen; SaO2: Arterial oxygen saturation; TACE: Transcatheter arterial chemoembolization; TLC: Total lung capacity.\nPatients with HPS had significantly higher AaO2 values (26.1 ± 9.3 vs. 15.5 ± 11.6 mmHg; P = 0.001), significantly lower arterial oxygen saturation (95.4 ± 2.5 vs. 96.8 ± 1.7; P = 0.019), and PaO2 values (76.8 ± 10.0 vs. 85.0 ± 12.0 mmHg; P = 0.015) than those without HPS. However, the partial arterial pressure of carbon dioxide was not significantly different between patients with and without HPS (37.4 ± 4.1 vs. 39.4 ± 4.2 mmHg; P = 0.104). In addition, the carbon monoxide diffusing capacity (DLCO) was the only pulmonary function test result that was significantly different between patients with and without HPS (83.3 ± 14.2% predicted vs. 93.1 ± 15.2% predicted; P = 0.032).\n[SUBTITLE] Clinical outcome of TACE for HCC according to HPS status [SUBSECTION] The median interval from HCC diagnosis to study entry was not significantly different between patients with and without HPS (8.5 [95% CI, 3.2–13.8] vs. 3.9 [95% CI, 0–9.9] months; P = 0.613). A total of 172 TACE sessions were performed for the patients during the study, with no significant difference between those with and without HPS (3.1 ± 1.5 sessions per patient vs. 3.3 ± 1.6 sessions per patient; P = 0.549). No major complications, defined according to the Society of Interventional Radiology clinical practice guidelines, were observed in any patients.[18] Five (9.3%) patients, including one with HPS and four without HPS, were lost to or refused the follow-up after 3.3, 2.5, 3.8, 5.2, and 5.3 months. The median overall survival (OS) was not significantly different between patients with and without HPS (12.0 [95% CI, 8.2–15.7] vs. 14.9 [95% CI, 11.9–17.9] months; P = 0.223).\nThe tumor response was not significantly different between patients with and without HPS (P = 0.748). For patients with HPS, complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD) were observed in 2 (10.5%), 8 (42.1%), 1 (5.3%), and 8 (42.1%) patients, respectively. For patients without HPS, CR, PR, SD, and PD were observed in 7 (20.0%), 15 (42.9%), 1 (2.9%), and 12 (17.1%) patients, respectively. Of the patients with PD, 18 (90%, 18/20) died before the 6-month evaluation, including 6 of the 19 (31.6%) patients with HPS and 12 of the 35 (34.3%) patients without HPS. The 6-month overall response rates (ie, the percentage of patients in whom CR or PR was observed) for patients with and without HPS were 53.8% (7/13) and 78.3% (18/23), respectively, and the difference was not significantly different (P = 0.153).\nA total of 32 (59.3%) patients died at the end of the study, mainly due to disease progression (87.5% [28/32]), with no significant difference of mortality between patients with and without HPS (73.7% [14/19] vs. 51.4% [18/35]; P = 0.112). The median OS was 10.1 (95% CI, 3.9–16.3) months for patients with HPS and 15.1 (95% CI, 7.3–22.9) months for patients without HPS, which was not a significant difference (P = 0.100) [Figure 2]. The median PFS was also not significantly different between patients with and without HPS (5.2 [95% CI, 0–12.8] vs. 8.4 [95% CI, 3.6–13.1] months; P = 0.537) [Figure 3]. In the multivariate Cox regression analyses, DLCO (hazard ratio [HR] = 1.033 [95% CI, 1.003–1.064]; P = 0.028) and Child-Pugh class (HR = 1.815 [95% CI, 1.011–3.260]; P = 0.046) were identified to be the independent prognostic factors of OS [Table 2].\nKaplan-Meier curve of OS according to HPS status. HPS: Hepatopulmonary syndrome; OS: Overall survival.\nKaplan-Meier curve of PFS according to HPS status. HPS: Hepatopulmonary syndrome; PFS: Progression-free survival.\nMultivariate cox regression analyses to identify the prognostic factors of OS.\nAaO2: Alveolar-arterial oxygen gradient; DLCO: Carbon monoxide diffusing capacity; HR: Hazard ratio; HPS: Hepatopulmonary syndrome; OS: Overall survival.\nThe median interval from HCC diagnosis to study entry was not significantly different between patients with and without HPS (8.5 [95% CI, 3.2–13.8] vs. 3.9 [95% CI, 0–9.9] months; P = 0.613). A total of 172 TACE sessions were performed for the patients during the study, with no significant difference between those with and without HPS (3.1 ± 1.5 sessions per patient vs. 3.3 ± 1.6 sessions per patient; P = 0.549). No major complications, defined according to the Society of Interventional Radiology clinical practice guidelines, were observed in any patients.[18] Five (9.3%) patients, including one with HPS and four without HPS, were lost to or refused the follow-up after 3.3, 2.5, 3.8, 5.2, and 5.3 months. The median overall survival (OS) was not significantly different between patients with and without HPS (12.0 [95% CI, 8.2–15.7] vs. 14.9 [95% CI, 11.9–17.9] months; P = 0.223).\nThe tumor response was not significantly different between patients with and without HPS (P = 0.748). For patients with HPS, complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD) were observed in 2 (10.5%), 8 (42.1%), 1 (5.3%), and 8 (42.1%) patients, respectively. For patients without HPS, CR, PR, SD, and PD were observed in 7 (20.0%), 15 (42.9%), 1 (2.9%), and 12 (17.1%) patients, respectively. Of the patients with PD, 18 (90%, 18/20) died before the 6-month evaluation, including 6 of the 19 (31.6%) patients with HPS and 12 of the 35 (34.3%) patients without HPS. The 6-month overall response rates (ie, the percentage of patients in whom CR or PR was observed) for patients with and without HPS were 53.8% (7/13) and 78.3% (18/23), respectively, and the difference was not significantly different (P = 0.153).\nA total of 32 (59.3%) patients died at the end of the study, mainly due to disease progression (87.5% [28/32]), with no significant difference of mortality between patients with and without HPS (73.7% [14/19] vs. 51.4% [18/35]; P = 0.112). The median OS was 10.1 (95% CI, 3.9–16.3) months for patients with HPS and 15.1 (95% CI, 7.3–22.9) months for patients without HPS, which was not a significant difference (P = 0.100) [Figure 2]. The median PFS was also not significantly different between patients with and without HPS (5.2 [95% CI, 0–12.8] vs. 8.4 [95% CI, 3.6–13.1] months; P = 0.537) [Figure 3]. In the multivariate Cox regression analyses, DLCO (hazard ratio [HR] = 1.033 [95% CI, 1.003–1.064]; P = 0.028) and Child-Pugh class (HR = 1.815 [95% CI, 1.011–3.260]; P = 0.046) were identified to be the independent prognostic factors of OS [Table 2].\nKaplan-Meier curve of OS according to HPS status. HPS: Hepatopulmonary syndrome; OS: Overall survival.\nKaplan-Meier curve of PFS according to HPS status. HPS: Hepatopulmonary syndrome; PFS: Progression-free survival.\nMultivariate cox regression analyses to identify the prognostic factors of OS.\nAaO2: Alveolar-arterial oxygen gradient; DLCO: Carbon monoxide diffusing capacity; HR: Hazard ratio; HPS: Hepatopulmonary syndrome; OS: Overall survival.", "A total of 60 patients with HCC undergoing TACE were eligible for inclusion in this study [Figure 1]. Six of these patients were excluded from the study due to previous TIPS creation (n = 2), discontinued treatment (n = 1), a poor echocardiographic window (n = 1), and intra-cardiac shunt (n = 2). The remaining 54 patients were included in the study and were screened for HPS. Among these patients, 31 (57.4%) had a positive CEE result. Of these patients, 19 (61.3%) had an elevated AaO2. These patients were considered to have HPS, including one (5.3%) with severe HPS, nine (47.4%) with moderate HPS, and nine (47.4%) with mild HPS. Therefore, the prevalence of HPS was 35.2% (19/54).\nFlow chart of the patient selection and HPS screening processes. AaO2: Alveolar-arterial oxygen gradient; CEE: Contrast-enhanced echocardiography; HPS: Hepatopulmonary syndrome; TIPS: Transjugular intra-hepatic portosystemic shunt.\nThe demographics, etiology of liver disease, cirrhosis status, Child-Pugh class, performance status, tumor characteristics, and history of prior treatments were not significantly different between those with and without HPS (all P > 0.050) [Table 1]. The number of patients who complained of dyspnea was significantly higher in the group with HPS than in the group without HPS (8/19 vs. 4/35; P = 0.016). However, the number of patients who had cyanosis (3/19 vs. 2/35; P = 0.332) and digital clubbing (2/19 vs. 2/35; P = 0.607) was not significantly different between those with and without HPS. Significantly more patients with HPS had spider angioma than those without HPS (8/19 vs. 3/35; P = 0.010).\nClinical characteristics of patients with and without HPS.\nData are presented as mean ± standard deviation or n(%).\nStudent's t test.\nFisher's exact test.\nPearson chi-square. AaO2: Alveolar-arterial oxygen gradient; ABG: Arterial blood gas; AFP: Alpha-fetoprotein; DLCO: Carbon monoxide diffusing capacity; ECOG: European Cooperative Oncology Group; FEV1: Forced expiratory volume in 1 s; FVC: Forced vital capacity; HBV: Hepatitis V virus; HCV: Hepatitis C virus; HE: Hepatic encephalopathy; HPS: Hepatopulmonary syndrome; PaCO2: Partial arterial pressure of carbon dioxide; PaO2: Partial arterial pressure of oxygen; SaO2: Arterial oxygen saturation; TACE: Transcatheter arterial chemoembolization; TLC: Total lung capacity.\nPatients with HPS had significantly higher AaO2 values (26.1 ± 9.3 vs. 15.5 ± 11.6 mmHg; P = 0.001), significantly lower arterial oxygen saturation (95.4 ± 2.5 vs. 96.8 ± 1.7; P = 0.019), and PaO2 values (76.8 ± 10.0 vs. 85.0 ± 12.0 mmHg; P = 0.015) than those without HPS. However, the partial arterial pressure of carbon dioxide was not significantly different between patients with and without HPS (37.4 ± 4.1 vs. 39.4 ± 4.2 mmHg; P = 0.104). In addition, the carbon monoxide diffusing capacity (DLCO) was the only pulmonary function test result that was significantly different between patients with and without HPS (83.3 ± 14.2% predicted vs. 93.1 ± 15.2% predicted; P = 0.032).", "The median interval from HCC diagnosis to study entry was not significantly different between patients with and without HPS (8.5 [95% CI, 3.2–13.8] vs. 3.9 [95% CI, 0–9.9] months; P = 0.613). A total of 172 TACE sessions were performed for the patients during the study, with no significant difference between those with and without HPS (3.1 ± 1.5 sessions per patient vs. 3.3 ± 1.6 sessions per patient; P = 0.549). No major complications, defined according to the Society of Interventional Radiology clinical practice guidelines, were observed in any patients.[18] Five (9.3%) patients, including one with HPS and four without HPS, were lost to or refused the follow-up after 3.3, 2.5, 3.8, 5.2, and 5.3 months. The median overall survival (OS) was not significantly different between patients with and without HPS (12.0 [95% CI, 8.2–15.7] vs. 14.9 [95% CI, 11.9–17.9] months; P = 0.223).\nThe tumor response was not significantly different between patients with and without HPS (P = 0.748). For patients with HPS, complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD) were observed in 2 (10.5%), 8 (42.1%), 1 (5.3%), and 8 (42.1%) patients, respectively. For patients without HPS, CR, PR, SD, and PD were observed in 7 (20.0%), 15 (42.9%), 1 (2.9%), and 12 (17.1%) patients, respectively. Of the patients with PD, 18 (90%, 18/20) died before the 6-month evaluation, including 6 of the 19 (31.6%) patients with HPS and 12 of the 35 (34.3%) patients without HPS. The 6-month overall response rates (ie, the percentage of patients in whom CR or PR was observed) for patients with and without HPS were 53.8% (7/13) and 78.3% (18/23), respectively, and the difference was not significantly different (P = 0.153).\nA total of 32 (59.3%) patients died at the end of the study, mainly due to disease progression (87.5% [28/32]), with no significant difference of mortality between patients with and without HPS (73.7% [14/19] vs. 51.4% [18/35]; P = 0.112). The median OS was 10.1 (95% CI, 3.9–16.3) months for patients with HPS and 15.1 (95% CI, 7.3–22.9) months for patients without HPS, which was not a significant difference (P = 0.100) [Figure 2]. The median PFS was also not significantly different between patients with and without HPS (5.2 [95% CI, 0–12.8] vs. 8.4 [95% CI, 3.6–13.1] months; P = 0.537) [Figure 3]. In the multivariate Cox regression analyses, DLCO (hazard ratio [HR] = 1.033 [95% CI, 1.003–1.064]; P = 0.028) and Child-Pugh class (HR = 1.815 [95% CI, 1.011–3.260]; P = 0.046) were identified to be the independent prognostic factors of OS [Table 2].\nKaplan-Meier curve of OS according to HPS status. HPS: Hepatopulmonary syndrome; OS: Overall survival.\nKaplan-Meier curve of PFS according to HPS status. HPS: Hepatopulmonary syndrome; PFS: Progression-free survival.\nMultivariate cox regression analyses to identify the prognostic factors of OS.\nAaO2: Alveolar-arterial oxygen gradient; DLCO: Carbon monoxide diffusing capacity; HR: Hazard ratio; HPS: Hepatopulmonary syndrome; OS: Overall survival.", "The prevalence of HPS in cirrhotic patients evaluated for LT or TIPS creation ranges from 12% to 32%, and those with HPS have double the risk of death compared to those without HPS.[9] Therefore, patients with HPS are eligible to receive higher priority on the transplant waitlist than patients without HPS.[19] In patients with unresectable HCC, however, the prevalence and prognostic impact of HPS remain unknown. In this study, we found that: (1) the prevalence of HPS in patients with HCC undergoing TACE was very common, (2) HPS was mostly mild or moderate in patients with HCC; and (3) the median OS and PFS were not significantly different between patients with and without HPS.\nApproximately 50% to 60% of cirrhotic patients evaluated for LT or TIPS creation have IPVD, as demonstrated by a positive CEE result; depending on the cut-off value of AaO2 or PaO2 used to define arterial oxygenation defects, 27% to 94% of these patients could meet the diagnostic criteria for HPS, resulting in a prevalence of 12% to 32% for HPS in this population.[8,15] In this study, 35.2% (19/54) of patients met the diagnostic criteria provided by the clinical practice guidelines for HPS.[8,11] These results suggest that the prevalence of HPS is comparable between cirrhotic patients evaluated for LT or TIPS creation and HCC patients undergoing TACE. Notably, however, the majority of the patients in this study had only mild to moderate HPS, which also explains why many patients had no apparent symptoms. Similarly, in two recent prospective studies including cirrhotic and Budd-Chiari syndrome patients undergoing TIPS creation or balloon angioplasty, where severe to very severe HPS was observed in only 0% to 8% of the study population.[13,15] This indicates that while HPS is common, the majority of cases is mild to moderate in severity with no apparent symptom.\nHPS substantially worsens the prognosis of cirrhosis.[9] Swanson et al[20] showed a median survival of approximately 25 months and a 5-year survival rate of 23% in cirrhotic patients with HPS, compared to a median survival of 87 months and a 5-year survival rate of 63% in cirrhotic patients without HPS matched for etiology and severity of the liver disease. In addition, mortality was mainly related to the major complications of liver dysfunction and/or portal hypertension, as opposed to those of a primary respiratory event. However, these findings are unlikely to be generalizable to patients with unresectable HCC since they have a much shorter survival and higher risk of death by disease progression.[21] In this study, the median OS and PFS were not significantly different between patients with and without HPS. In addition, disease progression, rather than major complications of liver dysfunction and/or portal hypertension, was the cause of discontinuation in 87.5% (28/32) of the patients. These findings suggest that mild or moderate HPS may not have a prognostic impact in patients with unresectable HCC undergoing TACE, and therefore, HPS screening and treatment may be unwarranted in this population. However, for severe and very severe HPS in patients with unresectable HCC, screening (with pulse oximetry) and treatment (with oxygenation therapy and garlic) may be advisable,[9,22] because mortality in HPS is associated with the severity of hypoxemia,[21,23] and HPS could worsen patient functional status and quality of life.[9]\nThis study has several limitations. First, although this is the largest clinical cohort for patients with HCC and HPS to our acknowledge, the sample size of the current study is still relatively small.[11,24] Second, the HPS status was not reevaluated during the study to confirm that no changes had occurred since study entry, although HPS seldom resolves spontaneously and most patients with subclinical HPS seem to have stable arterial oxygenation over time.[23,25] Third, the majority of the patients had only mild to moderate HPS, and the results may not be applicable for patients with severe and very severe HPS.\nIn conclusion, mild or moderate HPS is common in patients with unresectable HCC undergoing TACE but it does not seem to have a prognostic impact in patients with unresectable HCC. Therefore, HPS screening and treatment seem to be unwarranted in patients with HCC undergoing TACE and should, perhaps, be considered for those with only severe or very severe HPS.", "This work was supported by the National Natural Science Fund of China (No. 82001937) and the CAMS Initiative for Innovative Medicine (No. 2021-I2M-1-015).", "None." ]
[ "intro", "methods", null, "methods", null, null, "results", "subjects", null, "discussion", null, "COI-statement" ]
[ "Hepatocellular carcinoma", "Hepatopulmonary syndrome", "Chemoembolization", "Therapeutic", "Prognosis", "Prevalence" ]
Genetic and environmental etiology of drinking motives in college students.
36256465
Drinking motives are robust proximal predictors of alcohol use behaviors and may mediate distinct etiological pathways in the development of alcohol misuse. However, little is known about the genetic and environmental etiology of drinking motives themselves and their potential utility as endophenotypes.
BACKGROUND
Here, we leverage a longitudinal study of college students from diverse racial/ethnic backgrounds (phenotypic N = 9889, genotypic N = 4855) to investigate the temporal stability and demographic and environmental predictors of four types of drinking motives (enhancement, social, coping, and conformity). Using genome-wide association study (GWAS) and in silico tools, we characterize their associated genes and genetic variants (single nucleotide polymorphisms or SNPs).
METHODS
Drinking motives were stable across four years of college (ICC >0.74). Some robust environmental predictors of alcohol misuse (parental autonomy granting and peer deviance) were broadly associated with multiple types of drinking motives, while others (e.g., trauma exposure) were type specific. Genome-wide analyses indicated modest SNP-based heritability (14-22%, n.s.) and several suggestive genomic loci that corroborate findings from previous molecular genetic studies (e.g., PECR and SIRT4 genes), indicating possible differences in the genetic etiology of positive versus negative reinforcement drinking motives that align with an internalizing/externalizing typology of alcohol misuse. Coping motives were significantly genetically correlated with alcohol use disorder diagnoses (rg  = 0.71, p = 0.001). However, results from the genetic analyses were largely underpowered to detect significant associations.
RESULTS
Drinking motives show promise as endophenotypes but require further investigation in larger samples to further our understanding of the etiology of alcohol misuse.
CONCLUSIONS
[ "Humans", "Genome-Wide Association Study", "Alcoholism", "Longitudinal Studies", "Students", "Motivation", "Universities", "Alcohol Drinking", "Adaptation, Psychological", "Alcohol Drinking in College" ]
9828131
null
null
null
null
RESULTS
[SUBTITLE] Phenotypic etiology [SUBSECTION] Descriptive statistics for drinking motives across survey waves are shown in Table 1. Mean levels for all four motives increased slightly but remained relatively stable across college. Correlations between each of the motives across time are shown in Table 2. Although there was some decay in the strength of correlation between longer periods of time (e.g., year 1 with year 4), each pair of adjacent time points was moderately correlated (Pearson r = 0.37 to 0.56, p < 5 × 10−20), with excellent overall stability (ICC = 0.75 to 0.81). Conformity motives showed the weakest correlations across time but also had the lowest levels of endorsement. Consistent with previous research, there were significant positive cross‐sectional and longitudinal correlations between motives and alcohol use outcomes, particularly between coping motives and AUDsx and between enhancement/social motives and both consumption and AUDsx (Table 3). Cross‐time correlations for drinking motives across five waves of assessment. Note: *p < 5e‐10, **p < 5e‐20, ***p < 5e‐100. Cross‐sectional and prospective correlations between drinking motives and alcohol use outcomes. Note: Bolded values are significant after multiple testing correction for 56 tests, adjusted alpha = 0.0009. Abbreviations: AUDsx, alcohol use disorder symptom count; Y1F‐Y4S, measurement time from Year 1 Fall to Year 4 Spring. Results from the linear models of predictors of drinking motives are displayed in Table 4. Male gender, older age, and recent trauma exposure were associated with higher levels of conformity motives. Students from racial and ethnic minorities generally had lower or not significantly different levels of all four drinking motives when compared with White students, with the exception that self‐reported Asian ethnicity was associated with higher levels of conformity motives. Parental autonomy granting was associated with lower levels of all drinking motives, while higher parental involvement was associated with higher levels of positive reinforcement (social and enhancement) motives. Peer deviance was associated with higher levels of all motives except conformity. Lifetime (pre‐college) trauma exposure was simultaneously associated with both higher coping motives and lower social motives. Linear regression results of demographic and environmental factors predicting drinking motives. Note: Reference category for the Ethnicity measure is White. Bolded values are significant after multiple testing correction for 8 predictor variables, adjusted alpha = 0.0062. Descriptive statistics for drinking motives across survey waves are shown in Table 1. Mean levels for all four motives increased slightly but remained relatively stable across college. Correlations between each of the motives across time are shown in Table 2. Although there was some decay in the strength of correlation between longer periods of time (e.g., year 1 with year 4), each pair of adjacent time points was moderately correlated (Pearson r = 0.37 to 0.56, p < 5 × 10−20), with excellent overall stability (ICC = 0.75 to 0.81). Conformity motives showed the weakest correlations across time but also had the lowest levels of endorsement. Consistent with previous research, there were significant positive cross‐sectional and longitudinal correlations between motives and alcohol use outcomes, particularly between coping motives and AUDsx and between enhancement/social motives and both consumption and AUDsx (Table 3). Cross‐time correlations for drinking motives across five waves of assessment. Note: *p < 5e‐10, **p < 5e‐20, ***p < 5e‐100. Cross‐sectional and prospective correlations between drinking motives and alcohol use outcomes. Note: Bolded values are significant after multiple testing correction for 56 tests, adjusted alpha = 0.0009. Abbreviations: AUDsx, alcohol use disorder symptom count; Y1F‐Y4S, measurement time from Year 1 Fall to Year 4 Spring. Results from the linear models of predictors of drinking motives are displayed in Table 4. Male gender, older age, and recent trauma exposure were associated with higher levels of conformity motives. Students from racial and ethnic minorities generally had lower or not significantly different levels of all four drinking motives when compared with White students, with the exception that self‐reported Asian ethnicity was associated with higher levels of conformity motives. Parental autonomy granting was associated with lower levels of all drinking motives, while higher parental involvement was associated with higher levels of positive reinforcement (social and enhancement) motives. Peer deviance was associated with higher levels of all motives except conformity. Lifetime (pre‐college) trauma exposure was simultaneously associated with both higher coping motives and lower social motives. Linear regression results of demographic and environmental factors predicting drinking motives. Note: Reference category for the Ethnicity measure is White. Bolded values are significant after multiple testing correction for 8 predictor variables, adjusted alpha = 0.0062. [SUBTITLE] Genetic etiology [SUBSECTION] [SUBTITLE] SNP Heritability [SUBSECTION] Heritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample. Heritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample. [SUBTITLE] GWAS [SUBSECTION] Manhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3. Manhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives. Genomic annotation for top loci associated with drinking motives. Note: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci. The genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r 2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07). For the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7. For the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS. GWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives. Manhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3. Manhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives. Genomic annotation for top loci associated with drinking motives. Note: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci. The genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r 2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07). For the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7. For the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS. GWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives. [SUBTITLE] SNP Heritability [SUBSECTION] Heritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample. Heritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample. [SUBTITLE] GWAS [SUBSECTION] Manhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3. Manhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives. Genomic annotation for top loci associated with drinking motives. Note: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci. The genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r 2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07). For the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7. For the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS. GWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives. Manhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3. Manhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives. Genomic annotation for top loci associated with drinking motives. Note: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci. The genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r 2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07). For the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7. For the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS. GWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives. [SUBTITLE] Genetic correlation [SUBSECTION] LDSC indicated no significant genome‐wide heritability based on the SNP association signals, and a mean χ2 < 1 for social, enhancement, and conformity motives, making them unsuitable for further LDSC analyses. However, there was sufficient SNP heritability for coping motives (h 2 SNP = 0.56, SE = 0.21, mean χ2 = 1.20) to conduct genetic correlation analyses with external GWAS of alcohol‐related phenotypes. After Bonferroni correction for three tests (alpha = 0.05/3 = 0.017), these showed a significant correlation between coping motives and AUD diagnoses (r g = 0.71, p = 0.001), but not alcohol consumption (r g = 0.16, p = 0.069) or AUDIT‐T scores (r g = 0.05, p = 0.644). Local genetic correlation was carried out to obtain a more fine‐grained view of the genetic relationship between drinking motives and alcohol phenotypes. Examining 98 known alcohol‐related loci, we saw local enrichment (p < 0.0001) for heritability in 13, 20, 44, and 11 of the 98 loci for social, enhancement, coping, and conformity motives, respectively (Table S6). These all exceed the number of loci expected by chance at an alpha of 0.05 (binomial test p = 3.76 × 10−04, 1.43 × 10−04, 3.68 × 10−32 and 0.004, respectively). Local genetic correlations suggested some interesting findings, for example, negative genetic correlations between coping motives and (1) consumption (r g = −0.27, p = 0.004), (2) AUDIT‐T (r g = −0.24, p = 0.041), and (3) AUD (r g = −0.34, p = 0.013) at the ADH locus (chr4:96,764,066‐101,983,024). However, no local genetic correlations survived multiple testing correction. Trans‐ancestral genetic correlation analysis using Popcorn was limited by the low SNP heritability, so estimates were not calculable for many pairs of GWAS summary statistics (Table S7). For the correlations that could be calculated, there was no evidence that the SNP effects on drinking motives differed between ancestry groups: the correlation between ancestry groups did not significantly differ from r = 1.0 (all p's > 0.19). LDSC indicated no significant genome‐wide heritability based on the SNP association signals, and a mean χ2 < 1 for social, enhancement, and conformity motives, making them unsuitable for further LDSC analyses. However, there was sufficient SNP heritability for coping motives (h 2 SNP = 0.56, SE = 0.21, mean χ2 = 1.20) to conduct genetic correlation analyses with external GWAS of alcohol‐related phenotypes. After Bonferroni correction for three tests (alpha = 0.05/3 = 0.017), these showed a significant correlation between coping motives and AUD diagnoses (r g = 0.71, p = 0.001), but not alcohol consumption (r g = 0.16, p = 0.069) or AUDIT‐T scores (r g = 0.05, p = 0.644). Local genetic correlation was carried out to obtain a more fine‐grained view of the genetic relationship between drinking motives and alcohol phenotypes. Examining 98 known alcohol‐related loci, we saw local enrichment (p < 0.0001) for heritability in 13, 20, 44, and 11 of the 98 loci for social, enhancement, coping, and conformity motives, respectively (Table S6). These all exceed the number of loci expected by chance at an alpha of 0.05 (binomial test p = 3.76 × 10−04, 1.43 × 10−04, 3.68 × 10−32 and 0.004, respectively). Local genetic correlations suggested some interesting findings, for example, negative genetic correlations between coping motives and (1) consumption (r g = −0.27, p = 0.004), (2) AUDIT‐T (r g = −0.24, p = 0.041), and (3) AUD (r g = −0.34, p = 0.013) at the ADH locus (chr4:96,764,066‐101,983,024). However, no local genetic correlations survived multiple testing correction. Trans‐ancestral genetic correlation analysis using Popcorn was limited by the low SNP heritability, so estimates were not calculable for many pairs of GWAS summary statistics (Table S7). For the correlations that could be calculated, there was no evidence that the SNP effects on drinking motives differed between ancestry groups: the correlation between ancestry groups did not significantly differ from r = 1.0 (all p's > 0.19).
null
null
[ "INTRODUCTION", "Participants", "Measures", "Drinking motives", "Alcohol use", "Environmental risk and protective factors", "Genotyping", "Data analysis", "Phenotypic etiology", "Genetic etiology", "\nSNP Heritability", "GWAS", "Genetic correlation" ]
[ "Alcohol misuse, including heavy consumption and alcohol use disorder (AUD), is one of the leading contributors to the global burden of disease (Rehm et al., 2009). It is well established that not all individuals are at equal risk for alcohol misuse, with genetic factors responsible for about 50% of individual differences in liability to developing AUDs (Verhulst et al., 2015). Aside from robust links to alcohol metabolism candidate genes (the ADH and ALDH gene families), early efforts to pinpoint the specific genes underlying this heritability were largely unsuccessful (Hart & Kranzler, 2015). This led researchers to conclude that the genetic etiology of alcohol misuse is characterized by high polygenicity: an aggregate impact of thousands of genetic variants (single nucleotide polymorphisms or SNPs), each with very small effects. Recent work capitalizing on enormous sample sizes through data‐sharing consortia has propelled forward progress in this area, resulting in nearly 100 genomic loci robustly associated with alcohol consumption and/or pathological use (Kranzler et al., 2019; Liu et al., 2019; Sanchez‐Roige et al., 2019; Walters et al., 2018; Zhou et al., 2020). However, significantly associated variants explain less than 10% of the inter‐individual variation in consumption and liability to AUD, leaving much of the heritability to be explained. Further, with the exception of alcohol metabolism genes, the mechanisms linking these genes to differences in alcohol use behaviors are largely still unclear.\nAn extensive literature indicates a diverse array of interpersonal characteristics and psychiatric disorders associated with alcohol use/misuse (Kessler et al., 2005; Krueger & Markon, 2006; Lynam & Miller, 2004; Whelan et al., 2014). There are also well‐supported divergent etiological pathways to alcohol misuse, most notably the “internalizing” and “externalizing” pathways (Conrod et al., 2006; Mezquita et al., 2014; Zucker, 2008), in which internalizing‐related alcohol misuse is driven by drinking to cope with negative affect (depression, anxiety) and externalizing‐related alcohol misuse stems from a core deficit in impulse control shared with other substance and behavioral addictions, typically with an earlier onset and more severe manifestation (Cloninger et al., 1988). Given the long, winding biological pathways connecting genetic variants in a cell's DNA to the manifestation of a high‐level behavioral outcome, it is likely that genetic variants associated with alcohol misuse have a more direct impact on the intermediate steps of these heterogeneous pathways.\nA potential tool to validate and investigate these pathways is the use of “endophenotypes,” factors which sit in the mediational pathway between a genetic predisposition and the distal manifestation of a trait or disorder (Gottesman & Gould, 2003; Hines et al., 2005). There is some debate over the specific definition of an endophenotype, but here we use the recommendations of Lenzenweger (2013) to refer to endophenotypes as measurable constructs located within the causal path between gene(s) and behavior. Fundamental criteria for an endophenotype include heritability, an association with the target phenotype, and shared genetic variance with the target phenotype, which are necessary conditions for mediation of a genetic etiological pathway. Iacono et al. (2017) further recommend that a useful endophenotype should demonstrate strong associations with specific genetic variants that are also linked to the target phenotype, and should be able to illuminate underlying developmental and/or mechanistic processes.\nDrinking motives, the reasons why people consume alcohol and what they hope to achieve by drinking, present a clear mechanism by which internalizing and externalizing pathways may lead to alcohol misuse and elucidate the intermediate mechanisms by which such pathways may unfold. As alcohol is a psychoactive drug with both stimulant and sedative effects (Kreusch et al., 2013), motivations for drinking are likely to differ between individuals and, potentially, even between the same individual in different situations. The foremost model of drinking motives, developed by Cooper (1994), proposes four distinct types of drinking motives that fall under two dimensions: valence (negative versus positive reinforcement) and source (internal versus external). Negative reinforcement motives include coping (internal) and conformity (external) motives, which reflect drinking to obtain relief from negative emotions or escape unpleasant affective states. Positive reinforcement motives, on the other hand, underlie drinking to obtain positive affective states or enjoy the pleasurable aspects of alcohol, and are subclassified into enhancement (internal) and social (external) motives. Internal motives are driven by one's own desires or feelings, while external motives are driven by social or environmental influences. Drinking motives, particularly enhancement and coping motives, have repeatedly demonstrated robust, proximal associations with measures of alcohol consumption and alcohol problems (Carpenter & Hasin, 1998; Kuntsche et al., 2005).\nDrinking motives are therefore plausible candidate endophenotypes for alcohol misuse, fulfilling the criterion of association with the target phenotype, and the utility of providing mechanistic insight. Despite their strong association with alcohol misuse, there has been little research on the etiology of drinking motives themselves, and even less on their genetic etiology. Moreover, research on the genetic etiology of drinking motives (and of alcohol‐related phenotypes in general) is especially limited among diverse ancestry cohorts. As potential endophenotypes, knowledge of some of the basic epidemiological aspects of drinking motives is needed: how reliable are they, how do they (or do they not) change across time, and what genetic and environmental factors are associated with their development—and might thus be used to modify them or predict individual risk?\nDrinking motives have primarily been studied in relation to alcohol, where studies consistently demonstrate that coping and enhancement motives serve as important mediators of the relationship between temperament (neuroticism and impulsivity, respectively) and alcohol use/misuse outcomes (Adams et al., 2012; Mezquita et al., 2010). Coping motives also have strong evidence for mediating the effects of environmental exposures—in particular, trauma—on problematic alcohol use (Hawn et al., 2020). Social drinking motives are the most highly endorsed type but are generally related to normative drinking rather than problematic use or negative consequences of use, while conformity motives have less consistent, and usually transient or weaker associations to alcohol outcomes (Bresin & Mekawi, 2021; Kuntsche et al., 2005). Overall higher motivations are generally associated with higher levels of alcohol use and problems (Bresin & Mekawi, 2021; Cho et al., 2015). The existing research on drinking motives has primarily been carried out in European ancestry individuals, although the existing evidence indicates that similar processes are at play across various ethnic groups and cultures (Ertl et al., 2018; Mezquita et al., 2018; Wicki et al., 2017). Cultural norms surrounding drinking behaviors do, though, play a role in cross‐cultural differences in mean levels of drinking motives (Kuntsche et al., 2015; Wicki et al., 2017).\nDrinking motives show moderate stability in young adulthood, with correlations of 0.38–0.64 across a 1‐year period from age 19–20 (Labhart et al., 2016), and standardized loadings of 0.68–0.87 on a latent trait factor across ages 23–33 (Windle & Windle, 2018). However, their stability during the critical period of emerging adulthood in which drinking behaviors are typically established has not yet been well‐characterized.\nOnly a few studies have directly examined predictors of drinking motives themselves, but these have provided initial evidence that factors associated with (adolescent) alcohol use, such as stressful life events and peer group alcohol use, are associated with higher levels of all types of drinking motives (Windle & Windle, 2018). Drinking motives, particularly enhancement motives, tend to resemble those of classroom peers (Temmen & Crockett, 2018) and drinking buddies (Kehayes et al., 2021), who seem to set the norms in which one's early drinking motives develop. Van Damme et al. (2015) further demonstrated an effect of parental drinking on enhancement/social motives, although it is unclear whether this is a result of parenting behaviors or an inherited predisposition towards alcohol use that is shared with positive reinforcement motives.\nA few twin studies in European ancestry samples have been conducted to estimate the heritability of drinking motives (Agrawal et al., 2008; Kristjansson et al., 2011; Mackie et al., 2011; Prescott et al., 2004; Young‐Wolff et al., 2009), fulfilling a requisite criterion for an endophenotype. These studies have examined Cooper's (1994) Drinking Motives Questionnaire (DMQ) and the related Alcohol Use Inventory (Wanberg & Horn, 1983) subscales and estimated that their heritability ranges from 11% to 40%. There is also some evidence from twin studies that drinking motives mediate the latent genetic overlap between depression and AUD via coping motives (Young‐Wolff et al., 2009). The latent genetic association between the personality traits of neuroticism/impulsivity and AUD are also mediated via coping and enhancement motives, respectively (Littlefield et al., 2011; Prescott et al., 2004), mirroring the relationship observed at the phenotypic level (Adams et al., 2012; Mezquita et al., 2010). However, to date, no robust molecular genetic studies have yet investigated the genetic etiology of drinking motives or their genetic relationship to alcohol misuse.\nThe aims of this project are thus twofold: (1) to investigate the phenotypic etiology of drinking motives and characterize their stability and associated environmental factors and (2) to investigate the genetic etiology of drinking motives in individuals from diverse ancestry groups using multiple molecular genetic methods. These analyses provide insight into the shared and distinct etiologies of different types of drinking motives and the utility of drinking motives to serve as endophenotypes for alcohol misuse.", "Data comes from the parent study “Spit for Science” (S4S) (Dick et al., 2014) a longitudinal, prospective study investigating genetic and environmental influences on mental health and substance use in college students at a large, urban, public university. The study enrolled four cohorts of incoming students between 2011 and 2015, and all first‐time freshmen aged 18 years and older were eligible to complete a self‐report survey and provide a saliva sample for DNA collection. Each subsequent spring that participants were enrolled at the university, they were invited to participate in a follow‐up survey. All participants provided informed consent and the S4S study was approved by the university Institutional Review Board.\nA total of N = 9889 students from four cohorts were enrolled in the study at the time of analysis. Participation rates have been consistently high across cohorts, with 63–68% of the eligible incoming students enrolling in the study each year and retention rates of 48–75% across follow‐up surveys. The demographic characteristics of the sample are consistent with those of the overall university student population: 61.5% female, with self‐reported race/ethnicity of 0.5% American Indian/Native Alaskan, 16.3% Asian, 18.9% African American, 49.4% Caucasian, 6.0% Hispanic/Latino, 6.2% multiracial, 0.7% Native Hawaiian/Pacific Islander, and 1.9% unknown/unreported. Nearly all participants, 91% of the total sample, also provided a DNA sample, and genotyping was completed for the first three cohorts at the time of analysis, with n = 6325 samples passing quality control.", "All measures were collected via a confidential online self‐report survey. Participants were emailed an individual link to this survey and could complete it at a time and location of their choosing. The surveys assessed a wide range of behaviors, characteristics, and environmental exposures. Data was collected and managed by the secure, web‐based REDCap system of electronic data capture tools (Harris et al., 2009). Measures were largely drawn from psychometrically validated scales administered in an abbreviated version to reduce participant burden. The first survey in the fall of the freshman year (Y1F) was considered a baseline metric and thus assessed lifetime measures of psychopathology and environmental exposures up to the beginning of college, while later assessments in the spring semester of each year (Y1S, Y2S, Y3S, and Y4S) focused on the experience of such factors in the time since the previous survey.\n[SUBTITLE] Drinking motives [SUBSECTION] In each survey, participants who had initiated drinking completed an abbreviated version of the Drinking Motives Questionnaire—Revised (Cooper, 1994). This scale contains four subscales whose items are summed to create scores: Coping (e.g., “because it helps me when I feel depressed or nervous”), Enhancement (e.g., “because it gives me a pleasant feeling”), Conformity (e.g., “to get in with a group I like”), and Social (e.g., “because it makes social gatherings more fun”). Responses are on a Likert‐like scale from 1 = Strongly Agree to 4 = Strongly Disagree (reverse coded). Four items per each subscale were assessed in the Y1F, Y1S, and Y2S surveys, and the one best‐performing item (based on factor loadings) from each subscale was included in the Y3S and Y4S surveys due to space limitations. However, descriptive statistics (see Table 1) and correlations across waves showed that the single‐item scores performed similarly to the multi‐item scale scores.\nDescriptive statistics for drinking motive scores.\nIn each survey, participants who had initiated drinking completed an abbreviated version of the Drinking Motives Questionnaire—Revised (Cooper, 1994). This scale contains four subscales whose items are summed to create scores: Coping (e.g., “because it helps me when I feel depressed or nervous”), Enhancement (e.g., “because it gives me a pleasant feeling”), Conformity (e.g., “to get in with a group I like”), and Social (e.g., “because it makes social gatherings more fun”). Responses are on a Likert‐like scale from 1 = Strongly Agree to 4 = Strongly Disagree (reverse coded). Four items per each subscale were assessed in the Y1F, Y1S, and Y2S surveys, and the one best‐performing item (based on factor loadings) from each subscale was included in the Y3S and Y4S surveys due to space limitations. However, descriptive statistics (see Table 1) and correlations across waves showed that the single‐item scores performed similarly to the multi‐item scale scores.\nDescriptive statistics for drinking motive scores.\n[SUBTITLE] Alcohol use [SUBSECTION] At each wave, participants were asked if they had initiated alcohol use, and, if so, about alcohol use behaviors including typical consumption (gm EtOH/month) and symptoms of DSM‐5 Alcohol Use Disorder (AUDsx), which were measured using the Semi‐Structured Assessment for the Genetics of Alcoholism (SSAGA; Bucholz et al., 1994) and summed.\nAt each wave, participants were asked if they had initiated alcohol use, and, if so, about alcohol use behaviors including typical consumption (gm EtOH/month) and symptoms of DSM‐5 Alcohol Use Disorder (AUDsx), which were measured using the Semi‐Structured Assessment for the Genetics of Alcoholism (SSAGA; Bucholz et al., 1994) and summed.\n[SUBTITLE] Environmental risk and protective factors [SUBSECTION] The S4S surveys assess numerous environmental and psychosocial constructs that have demonstrated associations with alcohol use and psychopathology. Among these are parenting behaviors, specifically the Involvement and Autonomy Granting subscales of Steinberg's Parenting Style scale (Steinberg et al., 1992), peer deviance (the proportion of one's friends who engage in deviant behaviors such as getting drunk and cutting school, as described by Kendler et al., 2008), and exposure to traumatic events such as an assault or natural disaster, measured by the Life Events Checklist (Gray et al., 2004).\nThe S4S surveys assess numerous environmental and psychosocial constructs that have demonstrated associations with alcohol use and psychopathology. Among these are parenting behaviors, specifically the Involvement and Autonomy Granting subscales of Steinberg's Parenting Style scale (Steinberg et al., 1992), peer deviance (the proportion of one's friends who engage in deviant behaviors such as getting drunk and cutting school, as described by Kendler et al., 2008), and exposure to traumatic events such as an assault or natural disaster, measured by the Life Events Checklist (Gray et al., 2004).", "In each survey, participants who had initiated drinking completed an abbreviated version of the Drinking Motives Questionnaire—Revised (Cooper, 1994). This scale contains four subscales whose items are summed to create scores: Coping (e.g., “because it helps me when I feel depressed or nervous”), Enhancement (e.g., “because it gives me a pleasant feeling”), Conformity (e.g., “to get in with a group I like”), and Social (e.g., “because it makes social gatherings more fun”). Responses are on a Likert‐like scale from 1 = Strongly Agree to 4 = Strongly Disagree (reverse coded). Four items per each subscale were assessed in the Y1F, Y1S, and Y2S surveys, and the one best‐performing item (based on factor loadings) from each subscale was included in the Y3S and Y4S surveys due to space limitations. However, descriptive statistics (see Table 1) and correlations across waves showed that the single‐item scores performed similarly to the multi‐item scale scores.\nDescriptive statistics for drinking motive scores.", "At each wave, participants were asked if they had initiated alcohol use, and, if so, about alcohol use behaviors including typical consumption (gm EtOH/month) and symptoms of DSM‐5 Alcohol Use Disorder (AUDsx), which were measured using the Semi‐Structured Assessment for the Genetics of Alcoholism (SSAGA; Bucholz et al., 1994) and summed.", "The S4S surveys assess numerous environmental and psychosocial constructs that have demonstrated associations with alcohol use and psychopathology. Among these are parenting behaviors, specifically the Involvement and Autonomy Granting subscales of Steinberg's Parenting Style scale (Steinberg et al., 1992), peer deviance (the proportion of one's friends who engage in deviant behaviors such as getting drunk and cutting school, as described by Kendler et al., 2008), and exposure to traumatic events such as an assault or natural disaster, measured by the Life Events Checklist (Gray et al., 2004).", "Information about genotyping for this sample has been described in detail elsewhere (Dick et al., 2014; Webb et al., 2017). Briefly, samples were genotyped on the Axiom BioBank Array, Catalog Version 2 (Affymetrix Inc., Santa Clara, CA) and imputed to the 1000 Genomes phase 3 all‐ancestries reference panel (The 1000 Genomes Project Consortium, 2015) after removing poor quality SNPs (missingness >5%, Hardy–Weinberg equilibrium [HWE] p values <5 × 10−6) and individual samples (genotyping rate < 98%, heterozygosity outliers, phenotypic/genotypic sex discordance, excess relatedness). Genetic ancestry principal components (PCs) were derived from the 1000 Genomes (phase 3) full reference population and projected onto the S4S samples to identify genetically homogenous ancestral groups for analysis, as described by Peterson et al. (2017). After this procedure, individuals from five continental ancestral super‐populations were available for analysis: Africa (AFR), the Americas (AMR), East Asia (EAS), Europe (EUR), and South Asia (SAS). Within‐group ancestry PCs were then calculated within each of these super‐populations in order to capture fine‐grained differences in allele frequencies that could contribute to residual population stratification. Within groups, additional quality control steps were taken to remove reference population outliers, those with excess relatedness (pi‐hat > 0.1), and SNPs with low ancestry‐specific minor allele frequency (MAF)/HWE.", "Data analysis involved two parts. First, we examined drinking motives at a phenotypic level in the full sample. We estimated the Pearson and intraclass correlations between motives across survey waves to estimate their temporal stability, calculated the Pearson correlations between motives and alcohol consumption/AUDsx cross‐sectionally and prospectively (Y1F motives with Y1F‐Y4S alcohol measures), and conducted linear regression analyses to examine how demographic and environmental characteristics predicted mean levels of each type of drinking motive.\nSecond, we conducted a series of analyses on the genotypic data to investigate the genetic etiology of drinking motives. The genotyped sample (n = 6325) was restricted to include only unrelated individuals with nonmissing phenotype information—i.e., those who initiated alcohol use during the time the measures were collected and chose to answer the relevant survey questions (analytic n = 4855). We used the mean of all available scores across waves (Table 1) for the four drinking motives subscales as the outcome phenotypes. We also utilized relevant covariates to control for possible confounding effects on genetic associations with the phenotypes, which included sex, age (mean across waves where participants had nonmissing drinking motives scores), and within‐ancestry PCs.\nWe estimated the heritability of drinking motives that could be attributable to measured genetic variants (SNP heritability; h\n\n2\n\nSNP) through the use of genome‐wide complex trait analysis (GCTA; Yang et al., 2011). Genotyped SNPs were filtered for a MAF >0.01 before calculating the genomic relatedness matrix between individuals. GCTA was then run separately in each ancestry using the first 10 within‐ancestry PCs, age, and sex as covariates, and the resulting heritability estimates were meta‐analyzed with a fixed‐effects, inverse variance‐weighted scheme.\nNext, we conducted a genome‐wide association (GWAS) of the imputed genetic variants. SNPs were filtered for imputation quality (INFO score >0.5) and ancestry‐specific MAF corresponding to a minor allele count (MAC) > 100. Previous work has established that a MAC > 40 allows for reliable statistical estimation (Bigdeli et al., 2014); we use a more conservative threshold because the somewhat skewed distribution of the phenotypes. Within‐ancestry PCs to include as covariates were decided based on their association with the phenotype in a stepwise linear regression to avoid overfitting (c.f. Webb et al., 2017). The specific PCs used in each analysis are shown in Table S1. GWAS was conducted using SNPTEST (Marchini et al., 2007), again running separately in each ancestry group and meta‐analyzing the association test for each SNP using inverse‐variance weighting in METAL (Abecasis et al., 2012). We present only the results for SNPs that were available in at least 1000 individuals (meaning that the SNP had to pass quality control filters in either the AFR, EUR, or a combination of two or more ancestry sub‐groups), to be certain that spurious results from small samples were not given undue consideration. Multiple testing correction was performed by using a Bonferroni‐corrected threshold for genome‐wide significance of 5 × 10−8 and calculating false discovery rates (FDR) using the qvalue package for R/Bioconductor (Storey et al., 2015). Given the small size of our sample and its corresponding statistical power (see Discussion), we also examine “suggestive” signals (p < 5 × 10−6) and ancestry‐specific results in the largest subgroup (EUR; n = 2537), although these are given lower credibility. Regional association plots for associated loci were visualized using LocusZoom (Pruim et al., 2010). Significant and suggestive signals were uploaded to FUMA (Watanabe et al., 2017) to define candidate genomic loci based on LD (variants with r\n\n2\n > 0.6 with the lead SNP, using the 1000 Genomes all‐ancestries reference panel for the LD structure [The 1000 Genomes Project Consortium, 2015]), annotate functional consequences of SNPs in the loci, and map candidate genes to these association signals based on position (within 10 kb), known expression quantitative trait locus (eQTL) effects, or 3‐dimensional chromatin interactions.\nAfter conducting GWAS at the individual variant level, we applied gene‐based and pathway‐based association analyses to test whether the SNP association signal was enriched at these aggregate levels, using MAGMA v.1.08 (de Leeuw et al., 2015). We used the 1000 Genomes as reference panels for the LD structure of each of the five ancestral continental superpopulations. After annotation of the SNPs to genes, genes were also grouped into pathways based on the curated hallmark and canonical pathways gene‐sets from the Molecular Signatures Database (Liberzon et al., 2015). These pathways represent groups of genes whose products are involved in known metabolic and regulatory biochemical processes. Gene‐based analyses were conducted on the ancestry‐specific SNPTEST results and meta‐analyzed with MAGMA, using an inverse variance‐weighted Stouffer's Z test. Pathway analyses were conducted on the meta‐analyzed, transancestral gene‐based results. We also used Popcorn (Brown et al., 2016) to estimate the genetic correlation of the GWAS results across ancestry groups, using the 1000 Genomes as a reference for LD patterns in each ancestry group and calculating the genetic impact correlation (the similarity between SNP effect sizes across ancestry groups, while accounting for differences in allele frequencies).\nFinally, we investigated a key endophenotype criterion for drinking motives by estimating their genetic overlap with alcohol‐related phenotypes. Using LD score regression (LDSC; Bulik‐Sullivan et al., 2015), we estimated h\n\n2\n\nSNP from the GWAS summary statistics and, if there was sufficient signal (mean χ2 > 1), we used LDSC to estimate the genome‐wide genetic correlations with three alcohol phenotypes from publicly available GWAS results: consumption (Liu et al., 2019), AUDIT total scores (Sanchez‐Roige et al., 2019), and AUD diagnoses (Walters et al., 2018). As LDSC assumptions rest on ancestry‐specific LD patterns, we used only results of the drinking motives GWAS from the largest group, the EUR ancestry subset (n = 2537), and conducted analyses using EUR LD scores provided with the software and recommended analysis settings (INFO >0.9, MAF >0.01, HapMap3 SNPs). LDSC provides only a global, genome‐wide estimate of the correlation, so we additionally conducted local genetic correlation for a more fine‐grained approach. We curated a list of 98 known alcohol‐related loci based on genome‐wide significant findings from well‐powered GWASs (Liu et al., 2019; Sanchez‐Roige et al., 2019; Walters et al., 2018; Zhou et al., 2020), using a 250 kb window around the lead SNP if a single position rather than a region was provided and merging overlapping loci (within 250 kb) within and across studies. Using LAVA (Werme et al., 2022), we investigated the local genetic correlation between drinking motives (EUR ancestry results) and the same three alcohol‐related phenotypes as for the genome‐wide correlations (consumption, AUDIT‐T, and AUD) within these curated loci. Bivariate correlations were calculated only when there was enrichment (p < 0.0001) in the univariate heritability for both phenotypes at a locus.", "Descriptive statistics for drinking motives across survey waves are shown in Table 1. Mean levels for all four motives increased slightly but remained relatively stable across college. Correlations between each of the motives across time are shown in Table 2. Although there was some decay in the strength of correlation between longer periods of time (e.g., year 1 with year 4), each pair of adjacent time points was moderately correlated (Pearson r = 0.37 to 0.56, p < 5 × 10−20), with excellent overall stability (ICC = 0.75 to 0.81). Conformity motives showed the weakest correlations across time but also had the lowest levels of endorsement. Consistent with previous research, there were significant positive cross‐sectional and longitudinal correlations between motives and alcohol use outcomes, particularly between coping motives and AUDsx and between enhancement/social motives and both consumption and AUDsx (Table 3).\nCross‐time correlations for drinking motives across five waves of assessment.\n\nNote: *p < 5e‐10, **p < 5e‐20, ***p < 5e‐100.\nCross‐sectional and prospective correlations between drinking motives and alcohol use outcomes.\n\nNote: Bolded values are significant after multiple testing correction for 56 tests, adjusted alpha = 0.0009.\nAbbreviations: AUDsx, alcohol use disorder symptom count; Y1F‐Y4S, measurement time from Year 1 Fall to Year 4 Spring.\nResults from the linear models of predictors of drinking motives are displayed in Table 4. Male gender, older age, and recent trauma exposure were associated with higher levels of conformity motives. Students from racial and ethnic minorities generally had lower or not significantly different levels of all four drinking motives when compared with White students, with the exception that self‐reported Asian ethnicity was associated with higher levels of conformity motives. Parental autonomy granting was associated with lower levels of all drinking motives, while higher parental involvement was associated with higher levels of positive reinforcement (social and enhancement) motives. Peer deviance was associated with higher levels of all motives except conformity. Lifetime (pre‐college) trauma exposure was simultaneously associated with both higher coping motives and lower social motives.\nLinear regression results of demographic and environmental factors predicting drinking motives.\n\nNote: Reference category for the Ethnicity measure is White. Bolded values are significant after multiple testing correction for 8 predictor variables, adjusted alpha = 0.0062.", "[SUBTITLE] \nSNP Heritability [SUBSECTION] Heritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample.\nHeritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample.\n[SUBTITLE] GWAS [SUBSECTION] Manhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3.\nManhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives.\nGenomic annotation for top loci associated with drinking motives.\n\nNote: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci.\nThe genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r\n2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07).\nFor the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7.\nFor the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS.\nGWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives.\nManhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3.\nManhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives.\nGenomic annotation for top loci associated with drinking motives.\n\nNote: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci.\nThe genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r\n2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07).\nFor the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7.\nFor the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS.\nGWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives.", "Heritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample.", "Manhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3.\nManhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives.\nGenomic annotation for top loci associated with drinking motives.\n\nNote: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci.\nThe genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r\n2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07).\nFor the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7.\nFor the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS.\nGWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives.", "LDSC indicated no significant genome‐wide heritability based on the SNP association signals, and a mean χ2 < 1 for social, enhancement, and conformity motives, making them unsuitable for further LDSC analyses. However, there was sufficient SNP heritability for coping motives (h\n\n2\n\nSNP = 0.56, SE = 0.21, mean χ2 = 1.20) to conduct genetic correlation analyses with external GWAS of alcohol‐related phenotypes. After Bonferroni correction for three tests (alpha = 0.05/3 = 0.017), these showed a significant correlation between coping motives and AUD diagnoses (r\ng = 0.71, p = 0.001), but not alcohol consumption (r\ng = 0.16, p = 0.069) or AUDIT‐T scores (r\ng = 0.05, p = 0.644).\nLocal genetic correlation was carried out to obtain a more fine‐grained view of the genetic relationship between drinking motives and alcohol phenotypes. Examining 98 known alcohol‐related loci, we saw local enrichment (p < 0.0001) for heritability in 13, 20, 44, and 11 of the 98 loci for social, enhancement, coping, and conformity motives, respectively (Table S6). These all exceed the number of loci expected by chance at an alpha of 0.05 (binomial test p = 3.76 × 10−04, 1.43 × 10−04, 3.68 × 10−32 and 0.004, respectively). Local genetic correlations suggested some interesting findings, for example, negative genetic correlations between coping motives and (1) consumption (r\ng = −0.27, p = 0.004), (2) AUDIT‐T (r\ng = −0.24, p = 0.041), and (3) AUD (r\ng = −0.34, p = 0.013) at the ADH locus (chr4:96,764,066‐101,983,024). However, no local genetic correlations survived multiple testing correction.\nTrans‐ancestral genetic correlation analysis using Popcorn was limited by the low SNP heritability, so estimates were not calculable for many pairs of GWAS summary statistics (Table S7). For the correlations that could be calculated, there was no evidence that the SNP effects on drinking motives differed between ancestry groups: the correlation between ancestry groups did not significantly differ from r = 1.0 (all p's > 0.19)." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "MATERIALS AND METHODS", "Participants", "Measures", "Drinking motives", "Alcohol use", "Environmental risk and protective factors", "Genotyping", "Data analysis", "RESULTS", "Phenotypic etiology", "Genetic etiology", "\nSNP Heritability", "GWAS", "Genetic correlation", "DISCUSSION", "CONFLICT OF INTEREST", "Supporting information" ]
[ "Alcohol misuse, including heavy consumption and alcohol use disorder (AUD), is one of the leading contributors to the global burden of disease (Rehm et al., 2009). It is well established that not all individuals are at equal risk for alcohol misuse, with genetic factors responsible for about 50% of individual differences in liability to developing AUDs (Verhulst et al., 2015). Aside from robust links to alcohol metabolism candidate genes (the ADH and ALDH gene families), early efforts to pinpoint the specific genes underlying this heritability were largely unsuccessful (Hart & Kranzler, 2015). This led researchers to conclude that the genetic etiology of alcohol misuse is characterized by high polygenicity: an aggregate impact of thousands of genetic variants (single nucleotide polymorphisms or SNPs), each with very small effects. Recent work capitalizing on enormous sample sizes through data‐sharing consortia has propelled forward progress in this area, resulting in nearly 100 genomic loci robustly associated with alcohol consumption and/or pathological use (Kranzler et al., 2019; Liu et al., 2019; Sanchez‐Roige et al., 2019; Walters et al., 2018; Zhou et al., 2020). However, significantly associated variants explain less than 10% of the inter‐individual variation in consumption and liability to AUD, leaving much of the heritability to be explained. Further, with the exception of alcohol metabolism genes, the mechanisms linking these genes to differences in alcohol use behaviors are largely still unclear.\nAn extensive literature indicates a diverse array of interpersonal characteristics and psychiatric disorders associated with alcohol use/misuse (Kessler et al., 2005; Krueger & Markon, 2006; Lynam & Miller, 2004; Whelan et al., 2014). There are also well‐supported divergent etiological pathways to alcohol misuse, most notably the “internalizing” and “externalizing” pathways (Conrod et al., 2006; Mezquita et al., 2014; Zucker, 2008), in which internalizing‐related alcohol misuse is driven by drinking to cope with negative affect (depression, anxiety) and externalizing‐related alcohol misuse stems from a core deficit in impulse control shared with other substance and behavioral addictions, typically with an earlier onset and more severe manifestation (Cloninger et al., 1988). Given the long, winding biological pathways connecting genetic variants in a cell's DNA to the manifestation of a high‐level behavioral outcome, it is likely that genetic variants associated with alcohol misuse have a more direct impact on the intermediate steps of these heterogeneous pathways.\nA potential tool to validate and investigate these pathways is the use of “endophenotypes,” factors which sit in the mediational pathway between a genetic predisposition and the distal manifestation of a trait or disorder (Gottesman & Gould, 2003; Hines et al., 2005). There is some debate over the specific definition of an endophenotype, but here we use the recommendations of Lenzenweger (2013) to refer to endophenotypes as measurable constructs located within the causal path between gene(s) and behavior. Fundamental criteria for an endophenotype include heritability, an association with the target phenotype, and shared genetic variance with the target phenotype, which are necessary conditions for mediation of a genetic etiological pathway. Iacono et al. (2017) further recommend that a useful endophenotype should demonstrate strong associations with specific genetic variants that are also linked to the target phenotype, and should be able to illuminate underlying developmental and/or mechanistic processes.\nDrinking motives, the reasons why people consume alcohol and what they hope to achieve by drinking, present a clear mechanism by which internalizing and externalizing pathways may lead to alcohol misuse and elucidate the intermediate mechanisms by which such pathways may unfold. As alcohol is a psychoactive drug with both stimulant and sedative effects (Kreusch et al., 2013), motivations for drinking are likely to differ between individuals and, potentially, even between the same individual in different situations. The foremost model of drinking motives, developed by Cooper (1994), proposes four distinct types of drinking motives that fall under two dimensions: valence (negative versus positive reinforcement) and source (internal versus external). Negative reinforcement motives include coping (internal) and conformity (external) motives, which reflect drinking to obtain relief from negative emotions or escape unpleasant affective states. Positive reinforcement motives, on the other hand, underlie drinking to obtain positive affective states or enjoy the pleasurable aspects of alcohol, and are subclassified into enhancement (internal) and social (external) motives. Internal motives are driven by one's own desires or feelings, while external motives are driven by social or environmental influences. Drinking motives, particularly enhancement and coping motives, have repeatedly demonstrated robust, proximal associations with measures of alcohol consumption and alcohol problems (Carpenter & Hasin, 1998; Kuntsche et al., 2005).\nDrinking motives are therefore plausible candidate endophenotypes for alcohol misuse, fulfilling the criterion of association with the target phenotype, and the utility of providing mechanistic insight. Despite their strong association with alcohol misuse, there has been little research on the etiology of drinking motives themselves, and even less on their genetic etiology. Moreover, research on the genetic etiology of drinking motives (and of alcohol‐related phenotypes in general) is especially limited among diverse ancestry cohorts. As potential endophenotypes, knowledge of some of the basic epidemiological aspects of drinking motives is needed: how reliable are they, how do they (or do they not) change across time, and what genetic and environmental factors are associated with their development—and might thus be used to modify them or predict individual risk?\nDrinking motives have primarily been studied in relation to alcohol, where studies consistently demonstrate that coping and enhancement motives serve as important mediators of the relationship between temperament (neuroticism and impulsivity, respectively) and alcohol use/misuse outcomes (Adams et al., 2012; Mezquita et al., 2010). Coping motives also have strong evidence for mediating the effects of environmental exposures—in particular, trauma—on problematic alcohol use (Hawn et al., 2020). Social drinking motives are the most highly endorsed type but are generally related to normative drinking rather than problematic use or negative consequences of use, while conformity motives have less consistent, and usually transient or weaker associations to alcohol outcomes (Bresin & Mekawi, 2021; Kuntsche et al., 2005). Overall higher motivations are generally associated with higher levels of alcohol use and problems (Bresin & Mekawi, 2021; Cho et al., 2015). The existing research on drinking motives has primarily been carried out in European ancestry individuals, although the existing evidence indicates that similar processes are at play across various ethnic groups and cultures (Ertl et al., 2018; Mezquita et al., 2018; Wicki et al., 2017). Cultural norms surrounding drinking behaviors do, though, play a role in cross‐cultural differences in mean levels of drinking motives (Kuntsche et al., 2015; Wicki et al., 2017).\nDrinking motives show moderate stability in young adulthood, with correlations of 0.38–0.64 across a 1‐year period from age 19–20 (Labhart et al., 2016), and standardized loadings of 0.68–0.87 on a latent trait factor across ages 23–33 (Windle & Windle, 2018). However, their stability during the critical period of emerging adulthood in which drinking behaviors are typically established has not yet been well‐characterized.\nOnly a few studies have directly examined predictors of drinking motives themselves, but these have provided initial evidence that factors associated with (adolescent) alcohol use, such as stressful life events and peer group alcohol use, are associated with higher levels of all types of drinking motives (Windle & Windle, 2018). Drinking motives, particularly enhancement motives, tend to resemble those of classroom peers (Temmen & Crockett, 2018) and drinking buddies (Kehayes et al., 2021), who seem to set the norms in which one's early drinking motives develop. Van Damme et al. (2015) further demonstrated an effect of parental drinking on enhancement/social motives, although it is unclear whether this is a result of parenting behaviors or an inherited predisposition towards alcohol use that is shared with positive reinforcement motives.\nA few twin studies in European ancestry samples have been conducted to estimate the heritability of drinking motives (Agrawal et al., 2008; Kristjansson et al., 2011; Mackie et al., 2011; Prescott et al., 2004; Young‐Wolff et al., 2009), fulfilling a requisite criterion for an endophenotype. These studies have examined Cooper's (1994) Drinking Motives Questionnaire (DMQ) and the related Alcohol Use Inventory (Wanberg & Horn, 1983) subscales and estimated that their heritability ranges from 11% to 40%. There is also some evidence from twin studies that drinking motives mediate the latent genetic overlap between depression and AUD via coping motives (Young‐Wolff et al., 2009). The latent genetic association between the personality traits of neuroticism/impulsivity and AUD are also mediated via coping and enhancement motives, respectively (Littlefield et al., 2011; Prescott et al., 2004), mirroring the relationship observed at the phenotypic level (Adams et al., 2012; Mezquita et al., 2010). However, to date, no robust molecular genetic studies have yet investigated the genetic etiology of drinking motives or their genetic relationship to alcohol misuse.\nThe aims of this project are thus twofold: (1) to investigate the phenotypic etiology of drinking motives and characterize their stability and associated environmental factors and (2) to investigate the genetic etiology of drinking motives in individuals from diverse ancestry groups using multiple molecular genetic methods. These analyses provide insight into the shared and distinct etiologies of different types of drinking motives and the utility of drinking motives to serve as endophenotypes for alcohol misuse.", "[SUBTITLE] Participants [SUBSECTION] Data comes from the parent study “Spit for Science” (S4S) (Dick et al., 2014) a longitudinal, prospective study investigating genetic and environmental influences on mental health and substance use in college students at a large, urban, public university. The study enrolled four cohorts of incoming students between 2011 and 2015, and all first‐time freshmen aged 18 years and older were eligible to complete a self‐report survey and provide a saliva sample for DNA collection. Each subsequent spring that participants were enrolled at the university, they were invited to participate in a follow‐up survey. All participants provided informed consent and the S4S study was approved by the university Institutional Review Board.\nA total of N = 9889 students from four cohorts were enrolled in the study at the time of analysis. Participation rates have been consistently high across cohorts, with 63–68% of the eligible incoming students enrolling in the study each year and retention rates of 48–75% across follow‐up surveys. The demographic characteristics of the sample are consistent with those of the overall university student population: 61.5% female, with self‐reported race/ethnicity of 0.5% American Indian/Native Alaskan, 16.3% Asian, 18.9% African American, 49.4% Caucasian, 6.0% Hispanic/Latino, 6.2% multiracial, 0.7% Native Hawaiian/Pacific Islander, and 1.9% unknown/unreported. Nearly all participants, 91% of the total sample, also provided a DNA sample, and genotyping was completed for the first three cohorts at the time of analysis, with n = 6325 samples passing quality control.\nData comes from the parent study “Spit for Science” (S4S) (Dick et al., 2014) a longitudinal, prospective study investigating genetic and environmental influences on mental health and substance use in college students at a large, urban, public university. The study enrolled four cohorts of incoming students between 2011 and 2015, and all first‐time freshmen aged 18 years and older were eligible to complete a self‐report survey and provide a saliva sample for DNA collection. Each subsequent spring that participants were enrolled at the university, they were invited to participate in a follow‐up survey. All participants provided informed consent and the S4S study was approved by the university Institutional Review Board.\nA total of N = 9889 students from four cohorts were enrolled in the study at the time of analysis. Participation rates have been consistently high across cohorts, with 63–68% of the eligible incoming students enrolling in the study each year and retention rates of 48–75% across follow‐up surveys. The demographic characteristics of the sample are consistent with those of the overall university student population: 61.5% female, with self‐reported race/ethnicity of 0.5% American Indian/Native Alaskan, 16.3% Asian, 18.9% African American, 49.4% Caucasian, 6.0% Hispanic/Latino, 6.2% multiracial, 0.7% Native Hawaiian/Pacific Islander, and 1.9% unknown/unreported. Nearly all participants, 91% of the total sample, also provided a DNA sample, and genotyping was completed for the first three cohorts at the time of analysis, with n = 6325 samples passing quality control.\n[SUBTITLE] Measures [SUBSECTION] All measures were collected via a confidential online self‐report survey. Participants were emailed an individual link to this survey and could complete it at a time and location of their choosing. The surveys assessed a wide range of behaviors, characteristics, and environmental exposures. Data was collected and managed by the secure, web‐based REDCap system of electronic data capture tools (Harris et al., 2009). Measures were largely drawn from psychometrically validated scales administered in an abbreviated version to reduce participant burden. The first survey in the fall of the freshman year (Y1F) was considered a baseline metric and thus assessed lifetime measures of psychopathology and environmental exposures up to the beginning of college, while later assessments in the spring semester of each year (Y1S, Y2S, Y3S, and Y4S) focused on the experience of such factors in the time since the previous survey.\n[SUBTITLE] Drinking motives [SUBSECTION] In each survey, participants who had initiated drinking completed an abbreviated version of the Drinking Motives Questionnaire—Revised (Cooper, 1994). This scale contains four subscales whose items are summed to create scores: Coping (e.g., “because it helps me when I feel depressed or nervous”), Enhancement (e.g., “because it gives me a pleasant feeling”), Conformity (e.g., “to get in with a group I like”), and Social (e.g., “because it makes social gatherings more fun”). Responses are on a Likert‐like scale from 1 = Strongly Agree to 4 = Strongly Disagree (reverse coded). Four items per each subscale were assessed in the Y1F, Y1S, and Y2S surveys, and the one best‐performing item (based on factor loadings) from each subscale was included in the Y3S and Y4S surveys due to space limitations. However, descriptive statistics (see Table 1) and correlations across waves showed that the single‐item scores performed similarly to the multi‐item scale scores.\nDescriptive statistics for drinking motive scores.\nIn each survey, participants who had initiated drinking completed an abbreviated version of the Drinking Motives Questionnaire—Revised (Cooper, 1994). This scale contains four subscales whose items are summed to create scores: Coping (e.g., “because it helps me when I feel depressed or nervous”), Enhancement (e.g., “because it gives me a pleasant feeling”), Conformity (e.g., “to get in with a group I like”), and Social (e.g., “because it makes social gatherings more fun”). Responses are on a Likert‐like scale from 1 = Strongly Agree to 4 = Strongly Disagree (reverse coded). Four items per each subscale were assessed in the Y1F, Y1S, and Y2S surveys, and the one best‐performing item (based on factor loadings) from each subscale was included in the Y3S and Y4S surveys due to space limitations. However, descriptive statistics (see Table 1) and correlations across waves showed that the single‐item scores performed similarly to the multi‐item scale scores.\nDescriptive statistics for drinking motive scores.\n[SUBTITLE] Alcohol use [SUBSECTION] At each wave, participants were asked if they had initiated alcohol use, and, if so, about alcohol use behaviors including typical consumption (gm EtOH/month) and symptoms of DSM‐5 Alcohol Use Disorder (AUDsx), which were measured using the Semi‐Structured Assessment for the Genetics of Alcoholism (SSAGA; Bucholz et al., 1994) and summed.\nAt each wave, participants were asked if they had initiated alcohol use, and, if so, about alcohol use behaviors including typical consumption (gm EtOH/month) and symptoms of DSM‐5 Alcohol Use Disorder (AUDsx), which were measured using the Semi‐Structured Assessment for the Genetics of Alcoholism (SSAGA; Bucholz et al., 1994) and summed.\n[SUBTITLE] Environmental risk and protective factors [SUBSECTION] The S4S surveys assess numerous environmental and psychosocial constructs that have demonstrated associations with alcohol use and psychopathology. Among these are parenting behaviors, specifically the Involvement and Autonomy Granting subscales of Steinberg's Parenting Style scale (Steinberg et al., 1992), peer deviance (the proportion of one's friends who engage in deviant behaviors such as getting drunk and cutting school, as described by Kendler et al., 2008), and exposure to traumatic events such as an assault or natural disaster, measured by the Life Events Checklist (Gray et al., 2004).\nThe S4S surveys assess numerous environmental and psychosocial constructs that have demonstrated associations with alcohol use and psychopathology. Among these are parenting behaviors, specifically the Involvement and Autonomy Granting subscales of Steinberg's Parenting Style scale (Steinberg et al., 1992), peer deviance (the proportion of one's friends who engage in deviant behaviors such as getting drunk and cutting school, as described by Kendler et al., 2008), and exposure to traumatic events such as an assault or natural disaster, measured by the Life Events Checklist (Gray et al., 2004).\nAll measures were collected via a confidential online self‐report survey. Participants were emailed an individual link to this survey and could complete it at a time and location of their choosing. The surveys assessed a wide range of behaviors, characteristics, and environmental exposures. Data was collected and managed by the secure, web‐based REDCap system of electronic data capture tools (Harris et al., 2009). Measures were largely drawn from psychometrically validated scales administered in an abbreviated version to reduce participant burden. The first survey in the fall of the freshman year (Y1F) was considered a baseline metric and thus assessed lifetime measures of psychopathology and environmental exposures up to the beginning of college, while later assessments in the spring semester of each year (Y1S, Y2S, Y3S, and Y4S) focused on the experience of such factors in the time since the previous survey.\n[SUBTITLE] Drinking motives [SUBSECTION] In each survey, participants who had initiated drinking completed an abbreviated version of the Drinking Motives Questionnaire—Revised (Cooper, 1994). This scale contains four subscales whose items are summed to create scores: Coping (e.g., “because it helps me when I feel depressed or nervous”), Enhancement (e.g., “because it gives me a pleasant feeling”), Conformity (e.g., “to get in with a group I like”), and Social (e.g., “because it makes social gatherings more fun”). Responses are on a Likert‐like scale from 1 = Strongly Agree to 4 = Strongly Disagree (reverse coded). Four items per each subscale were assessed in the Y1F, Y1S, and Y2S surveys, and the one best‐performing item (based on factor loadings) from each subscale was included in the Y3S and Y4S surveys due to space limitations. However, descriptive statistics (see Table 1) and correlations across waves showed that the single‐item scores performed similarly to the multi‐item scale scores.\nDescriptive statistics for drinking motive scores.\nIn each survey, participants who had initiated drinking completed an abbreviated version of the Drinking Motives Questionnaire—Revised (Cooper, 1994). This scale contains four subscales whose items are summed to create scores: Coping (e.g., “because it helps me when I feel depressed or nervous”), Enhancement (e.g., “because it gives me a pleasant feeling”), Conformity (e.g., “to get in with a group I like”), and Social (e.g., “because it makes social gatherings more fun”). Responses are on a Likert‐like scale from 1 = Strongly Agree to 4 = Strongly Disagree (reverse coded). Four items per each subscale were assessed in the Y1F, Y1S, and Y2S surveys, and the one best‐performing item (based on factor loadings) from each subscale was included in the Y3S and Y4S surveys due to space limitations. However, descriptive statistics (see Table 1) and correlations across waves showed that the single‐item scores performed similarly to the multi‐item scale scores.\nDescriptive statistics for drinking motive scores.\n[SUBTITLE] Alcohol use [SUBSECTION] At each wave, participants were asked if they had initiated alcohol use, and, if so, about alcohol use behaviors including typical consumption (gm EtOH/month) and symptoms of DSM‐5 Alcohol Use Disorder (AUDsx), which were measured using the Semi‐Structured Assessment for the Genetics of Alcoholism (SSAGA; Bucholz et al., 1994) and summed.\nAt each wave, participants were asked if they had initiated alcohol use, and, if so, about alcohol use behaviors including typical consumption (gm EtOH/month) and symptoms of DSM‐5 Alcohol Use Disorder (AUDsx), which were measured using the Semi‐Structured Assessment for the Genetics of Alcoholism (SSAGA; Bucholz et al., 1994) and summed.\n[SUBTITLE] Environmental risk and protective factors [SUBSECTION] The S4S surveys assess numerous environmental and psychosocial constructs that have demonstrated associations with alcohol use and psychopathology. Among these are parenting behaviors, specifically the Involvement and Autonomy Granting subscales of Steinberg's Parenting Style scale (Steinberg et al., 1992), peer deviance (the proportion of one's friends who engage in deviant behaviors such as getting drunk and cutting school, as described by Kendler et al., 2008), and exposure to traumatic events such as an assault or natural disaster, measured by the Life Events Checklist (Gray et al., 2004).\nThe S4S surveys assess numerous environmental and psychosocial constructs that have demonstrated associations with alcohol use and psychopathology. Among these are parenting behaviors, specifically the Involvement and Autonomy Granting subscales of Steinberg's Parenting Style scale (Steinberg et al., 1992), peer deviance (the proportion of one's friends who engage in deviant behaviors such as getting drunk and cutting school, as described by Kendler et al., 2008), and exposure to traumatic events such as an assault or natural disaster, measured by the Life Events Checklist (Gray et al., 2004).\n[SUBTITLE] Genotyping [SUBSECTION] Information about genotyping for this sample has been described in detail elsewhere (Dick et al., 2014; Webb et al., 2017). Briefly, samples were genotyped on the Axiom BioBank Array, Catalog Version 2 (Affymetrix Inc., Santa Clara, CA) and imputed to the 1000 Genomes phase 3 all‐ancestries reference panel (The 1000 Genomes Project Consortium, 2015) after removing poor quality SNPs (missingness >5%, Hardy–Weinberg equilibrium [HWE] p values <5 × 10−6) and individual samples (genotyping rate < 98%, heterozygosity outliers, phenotypic/genotypic sex discordance, excess relatedness). Genetic ancestry principal components (PCs) were derived from the 1000 Genomes (phase 3) full reference population and projected onto the S4S samples to identify genetically homogenous ancestral groups for analysis, as described by Peterson et al. (2017). After this procedure, individuals from five continental ancestral super‐populations were available for analysis: Africa (AFR), the Americas (AMR), East Asia (EAS), Europe (EUR), and South Asia (SAS). Within‐group ancestry PCs were then calculated within each of these super‐populations in order to capture fine‐grained differences in allele frequencies that could contribute to residual population stratification. Within groups, additional quality control steps were taken to remove reference population outliers, those with excess relatedness (pi‐hat > 0.1), and SNPs with low ancestry‐specific minor allele frequency (MAF)/HWE.\nInformation about genotyping for this sample has been described in detail elsewhere (Dick et al., 2014; Webb et al., 2017). Briefly, samples were genotyped on the Axiom BioBank Array, Catalog Version 2 (Affymetrix Inc., Santa Clara, CA) and imputed to the 1000 Genomes phase 3 all‐ancestries reference panel (The 1000 Genomes Project Consortium, 2015) after removing poor quality SNPs (missingness >5%, Hardy–Weinberg equilibrium [HWE] p values <5 × 10−6) and individual samples (genotyping rate < 98%, heterozygosity outliers, phenotypic/genotypic sex discordance, excess relatedness). Genetic ancestry principal components (PCs) were derived from the 1000 Genomes (phase 3) full reference population and projected onto the S4S samples to identify genetically homogenous ancestral groups for analysis, as described by Peterson et al. (2017). After this procedure, individuals from five continental ancestral super‐populations were available for analysis: Africa (AFR), the Americas (AMR), East Asia (EAS), Europe (EUR), and South Asia (SAS). Within‐group ancestry PCs were then calculated within each of these super‐populations in order to capture fine‐grained differences in allele frequencies that could contribute to residual population stratification. Within groups, additional quality control steps were taken to remove reference population outliers, those with excess relatedness (pi‐hat > 0.1), and SNPs with low ancestry‐specific minor allele frequency (MAF)/HWE.\n[SUBTITLE] Data analysis [SUBSECTION] Data analysis involved two parts. First, we examined drinking motives at a phenotypic level in the full sample. We estimated the Pearson and intraclass correlations between motives across survey waves to estimate their temporal stability, calculated the Pearson correlations between motives and alcohol consumption/AUDsx cross‐sectionally and prospectively (Y1F motives with Y1F‐Y4S alcohol measures), and conducted linear regression analyses to examine how demographic and environmental characteristics predicted mean levels of each type of drinking motive.\nSecond, we conducted a series of analyses on the genotypic data to investigate the genetic etiology of drinking motives. The genotyped sample (n = 6325) was restricted to include only unrelated individuals with nonmissing phenotype information—i.e., those who initiated alcohol use during the time the measures were collected and chose to answer the relevant survey questions (analytic n = 4855). We used the mean of all available scores across waves (Table 1) for the four drinking motives subscales as the outcome phenotypes. We also utilized relevant covariates to control for possible confounding effects on genetic associations with the phenotypes, which included sex, age (mean across waves where participants had nonmissing drinking motives scores), and within‐ancestry PCs.\nWe estimated the heritability of drinking motives that could be attributable to measured genetic variants (SNP heritability; h\n\n2\n\nSNP) through the use of genome‐wide complex trait analysis (GCTA; Yang et al., 2011). Genotyped SNPs were filtered for a MAF >0.01 before calculating the genomic relatedness matrix between individuals. GCTA was then run separately in each ancestry using the first 10 within‐ancestry PCs, age, and sex as covariates, and the resulting heritability estimates were meta‐analyzed with a fixed‐effects, inverse variance‐weighted scheme.\nNext, we conducted a genome‐wide association (GWAS) of the imputed genetic variants. SNPs were filtered for imputation quality (INFO score >0.5) and ancestry‐specific MAF corresponding to a minor allele count (MAC) > 100. Previous work has established that a MAC > 40 allows for reliable statistical estimation (Bigdeli et al., 2014); we use a more conservative threshold because the somewhat skewed distribution of the phenotypes. Within‐ancestry PCs to include as covariates were decided based on their association with the phenotype in a stepwise linear regression to avoid overfitting (c.f. Webb et al., 2017). The specific PCs used in each analysis are shown in Table S1. GWAS was conducted using SNPTEST (Marchini et al., 2007), again running separately in each ancestry group and meta‐analyzing the association test for each SNP using inverse‐variance weighting in METAL (Abecasis et al., 2012). We present only the results for SNPs that were available in at least 1000 individuals (meaning that the SNP had to pass quality control filters in either the AFR, EUR, or a combination of two or more ancestry sub‐groups), to be certain that spurious results from small samples were not given undue consideration. Multiple testing correction was performed by using a Bonferroni‐corrected threshold for genome‐wide significance of 5 × 10−8 and calculating false discovery rates (FDR) using the qvalue package for R/Bioconductor (Storey et al., 2015). Given the small size of our sample and its corresponding statistical power (see Discussion), we also examine “suggestive” signals (p < 5 × 10−6) and ancestry‐specific results in the largest subgroup (EUR; n = 2537), although these are given lower credibility. Regional association plots for associated loci were visualized using LocusZoom (Pruim et al., 2010). Significant and suggestive signals were uploaded to FUMA (Watanabe et al., 2017) to define candidate genomic loci based on LD (variants with r\n\n2\n > 0.6 with the lead SNP, using the 1000 Genomes all‐ancestries reference panel for the LD structure [The 1000 Genomes Project Consortium, 2015]), annotate functional consequences of SNPs in the loci, and map candidate genes to these association signals based on position (within 10 kb), known expression quantitative trait locus (eQTL) effects, or 3‐dimensional chromatin interactions.\nAfter conducting GWAS at the individual variant level, we applied gene‐based and pathway‐based association analyses to test whether the SNP association signal was enriched at these aggregate levels, using MAGMA v.1.08 (de Leeuw et al., 2015). We used the 1000 Genomes as reference panels for the LD structure of each of the five ancestral continental superpopulations. After annotation of the SNPs to genes, genes were also grouped into pathways based on the curated hallmark and canonical pathways gene‐sets from the Molecular Signatures Database (Liberzon et al., 2015). These pathways represent groups of genes whose products are involved in known metabolic and regulatory biochemical processes. Gene‐based analyses were conducted on the ancestry‐specific SNPTEST results and meta‐analyzed with MAGMA, using an inverse variance‐weighted Stouffer's Z test. Pathway analyses were conducted on the meta‐analyzed, transancestral gene‐based results. We also used Popcorn (Brown et al., 2016) to estimate the genetic correlation of the GWAS results across ancestry groups, using the 1000 Genomes as a reference for LD patterns in each ancestry group and calculating the genetic impact correlation (the similarity between SNP effect sizes across ancestry groups, while accounting for differences in allele frequencies).\nFinally, we investigated a key endophenotype criterion for drinking motives by estimating their genetic overlap with alcohol‐related phenotypes. Using LD score regression (LDSC; Bulik‐Sullivan et al., 2015), we estimated h\n\n2\n\nSNP from the GWAS summary statistics and, if there was sufficient signal (mean χ2 > 1), we used LDSC to estimate the genome‐wide genetic correlations with three alcohol phenotypes from publicly available GWAS results: consumption (Liu et al., 2019), AUDIT total scores (Sanchez‐Roige et al., 2019), and AUD diagnoses (Walters et al., 2018). As LDSC assumptions rest on ancestry‐specific LD patterns, we used only results of the drinking motives GWAS from the largest group, the EUR ancestry subset (n = 2537), and conducted analyses using EUR LD scores provided with the software and recommended analysis settings (INFO >0.9, MAF >0.01, HapMap3 SNPs). LDSC provides only a global, genome‐wide estimate of the correlation, so we additionally conducted local genetic correlation for a more fine‐grained approach. We curated a list of 98 known alcohol‐related loci based on genome‐wide significant findings from well‐powered GWASs (Liu et al., 2019; Sanchez‐Roige et al., 2019; Walters et al., 2018; Zhou et al., 2020), using a 250 kb window around the lead SNP if a single position rather than a region was provided and merging overlapping loci (within 250 kb) within and across studies. Using LAVA (Werme et al., 2022), we investigated the local genetic correlation between drinking motives (EUR ancestry results) and the same three alcohol‐related phenotypes as for the genome‐wide correlations (consumption, AUDIT‐T, and AUD) within these curated loci. Bivariate correlations were calculated only when there was enrichment (p < 0.0001) in the univariate heritability for both phenotypes at a locus.\nData analysis involved two parts. First, we examined drinking motives at a phenotypic level in the full sample. We estimated the Pearson and intraclass correlations between motives across survey waves to estimate their temporal stability, calculated the Pearson correlations between motives and alcohol consumption/AUDsx cross‐sectionally and prospectively (Y1F motives with Y1F‐Y4S alcohol measures), and conducted linear regression analyses to examine how demographic and environmental characteristics predicted mean levels of each type of drinking motive.\nSecond, we conducted a series of analyses on the genotypic data to investigate the genetic etiology of drinking motives. The genotyped sample (n = 6325) was restricted to include only unrelated individuals with nonmissing phenotype information—i.e., those who initiated alcohol use during the time the measures were collected and chose to answer the relevant survey questions (analytic n = 4855). We used the mean of all available scores across waves (Table 1) for the four drinking motives subscales as the outcome phenotypes. We also utilized relevant covariates to control for possible confounding effects on genetic associations with the phenotypes, which included sex, age (mean across waves where participants had nonmissing drinking motives scores), and within‐ancestry PCs.\nWe estimated the heritability of drinking motives that could be attributable to measured genetic variants (SNP heritability; h\n\n2\n\nSNP) through the use of genome‐wide complex trait analysis (GCTA; Yang et al., 2011). Genotyped SNPs were filtered for a MAF >0.01 before calculating the genomic relatedness matrix between individuals. GCTA was then run separately in each ancestry using the first 10 within‐ancestry PCs, age, and sex as covariates, and the resulting heritability estimates were meta‐analyzed with a fixed‐effects, inverse variance‐weighted scheme.\nNext, we conducted a genome‐wide association (GWAS) of the imputed genetic variants. SNPs were filtered for imputation quality (INFO score >0.5) and ancestry‐specific MAF corresponding to a minor allele count (MAC) > 100. Previous work has established that a MAC > 40 allows for reliable statistical estimation (Bigdeli et al., 2014); we use a more conservative threshold because the somewhat skewed distribution of the phenotypes. Within‐ancestry PCs to include as covariates were decided based on their association with the phenotype in a stepwise linear regression to avoid overfitting (c.f. Webb et al., 2017). The specific PCs used in each analysis are shown in Table S1. GWAS was conducted using SNPTEST (Marchini et al., 2007), again running separately in each ancestry group and meta‐analyzing the association test for each SNP using inverse‐variance weighting in METAL (Abecasis et al., 2012). We present only the results for SNPs that were available in at least 1000 individuals (meaning that the SNP had to pass quality control filters in either the AFR, EUR, or a combination of two or more ancestry sub‐groups), to be certain that spurious results from small samples were not given undue consideration. Multiple testing correction was performed by using a Bonferroni‐corrected threshold for genome‐wide significance of 5 × 10−8 and calculating false discovery rates (FDR) using the qvalue package for R/Bioconductor (Storey et al., 2015). Given the small size of our sample and its corresponding statistical power (see Discussion), we also examine “suggestive” signals (p < 5 × 10−6) and ancestry‐specific results in the largest subgroup (EUR; n = 2537), although these are given lower credibility. Regional association plots for associated loci were visualized using LocusZoom (Pruim et al., 2010). Significant and suggestive signals were uploaded to FUMA (Watanabe et al., 2017) to define candidate genomic loci based on LD (variants with r\n\n2\n > 0.6 with the lead SNP, using the 1000 Genomes all‐ancestries reference panel for the LD structure [The 1000 Genomes Project Consortium, 2015]), annotate functional consequences of SNPs in the loci, and map candidate genes to these association signals based on position (within 10 kb), known expression quantitative trait locus (eQTL) effects, or 3‐dimensional chromatin interactions.\nAfter conducting GWAS at the individual variant level, we applied gene‐based and pathway‐based association analyses to test whether the SNP association signal was enriched at these aggregate levels, using MAGMA v.1.08 (de Leeuw et al., 2015). We used the 1000 Genomes as reference panels for the LD structure of each of the five ancestral continental superpopulations. After annotation of the SNPs to genes, genes were also grouped into pathways based on the curated hallmark and canonical pathways gene‐sets from the Molecular Signatures Database (Liberzon et al., 2015). These pathways represent groups of genes whose products are involved in known metabolic and regulatory biochemical processes. Gene‐based analyses were conducted on the ancestry‐specific SNPTEST results and meta‐analyzed with MAGMA, using an inverse variance‐weighted Stouffer's Z test. Pathway analyses were conducted on the meta‐analyzed, transancestral gene‐based results. We also used Popcorn (Brown et al., 2016) to estimate the genetic correlation of the GWAS results across ancestry groups, using the 1000 Genomes as a reference for LD patterns in each ancestry group and calculating the genetic impact correlation (the similarity between SNP effect sizes across ancestry groups, while accounting for differences in allele frequencies).\nFinally, we investigated a key endophenotype criterion for drinking motives by estimating their genetic overlap with alcohol‐related phenotypes. Using LD score regression (LDSC; Bulik‐Sullivan et al., 2015), we estimated h\n\n2\n\nSNP from the GWAS summary statistics and, if there was sufficient signal (mean χ2 > 1), we used LDSC to estimate the genome‐wide genetic correlations with three alcohol phenotypes from publicly available GWAS results: consumption (Liu et al., 2019), AUDIT total scores (Sanchez‐Roige et al., 2019), and AUD diagnoses (Walters et al., 2018). As LDSC assumptions rest on ancestry‐specific LD patterns, we used only results of the drinking motives GWAS from the largest group, the EUR ancestry subset (n = 2537), and conducted analyses using EUR LD scores provided with the software and recommended analysis settings (INFO >0.9, MAF >0.01, HapMap3 SNPs). LDSC provides only a global, genome‐wide estimate of the correlation, so we additionally conducted local genetic correlation for a more fine‐grained approach. We curated a list of 98 known alcohol‐related loci based on genome‐wide significant findings from well‐powered GWASs (Liu et al., 2019; Sanchez‐Roige et al., 2019; Walters et al., 2018; Zhou et al., 2020), using a 250 kb window around the lead SNP if a single position rather than a region was provided and merging overlapping loci (within 250 kb) within and across studies. Using LAVA (Werme et al., 2022), we investigated the local genetic correlation between drinking motives (EUR ancestry results) and the same three alcohol‐related phenotypes as for the genome‐wide correlations (consumption, AUDIT‐T, and AUD) within these curated loci. Bivariate correlations were calculated only when there was enrichment (p < 0.0001) in the univariate heritability for both phenotypes at a locus.", "Data comes from the parent study “Spit for Science” (S4S) (Dick et al., 2014) a longitudinal, prospective study investigating genetic and environmental influences on mental health and substance use in college students at a large, urban, public university. The study enrolled four cohorts of incoming students between 2011 and 2015, and all first‐time freshmen aged 18 years and older were eligible to complete a self‐report survey and provide a saliva sample for DNA collection. Each subsequent spring that participants were enrolled at the university, they were invited to participate in a follow‐up survey. All participants provided informed consent and the S4S study was approved by the university Institutional Review Board.\nA total of N = 9889 students from four cohorts were enrolled in the study at the time of analysis. Participation rates have been consistently high across cohorts, with 63–68% of the eligible incoming students enrolling in the study each year and retention rates of 48–75% across follow‐up surveys. The demographic characteristics of the sample are consistent with those of the overall university student population: 61.5% female, with self‐reported race/ethnicity of 0.5% American Indian/Native Alaskan, 16.3% Asian, 18.9% African American, 49.4% Caucasian, 6.0% Hispanic/Latino, 6.2% multiracial, 0.7% Native Hawaiian/Pacific Islander, and 1.9% unknown/unreported. Nearly all participants, 91% of the total sample, also provided a DNA sample, and genotyping was completed for the first three cohorts at the time of analysis, with n = 6325 samples passing quality control.", "All measures were collected via a confidential online self‐report survey. Participants were emailed an individual link to this survey and could complete it at a time and location of their choosing. The surveys assessed a wide range of behaviors, characteristics, and environmental exposures. Data was collected and managed by the secure, web‐based REDCap system of electronic data capture tools (Harris et al., 2009). Measures were largely drawn from psychometrically validated scales administered in an abbreviated version to reduce participant burden. The first survey in the fall of the freshman year (Y1F) was considered a baseline metric and thus assessed lifetime measures of psychopathology and environmental exposures up to the beginning of college, while later assessments in the spring semester of each year (Y1S, Y2S, Y3S, and Y4S) focused on the experience of such factors in the time since the previous survey.\n[SUBTITLE] Drinking motives [SUBSECTION] In each survey, participants who had initiated drinking completed an abbreviated version of the Drinking Motives Questionnaire—Revised (Cooper, 1994). This scale contains four subscales whose items are summed to create scores: Coping (e.g., “because it helps me when I feel depressed or nervous”), Enhancement (e.g., “because it gives me a pleasant feeling”), Conformity (e.g., “to get in with a group I like”), and Social (e.g., “because it makes social gatherings more fun”). Responses are on a Likert‐like scale from 1 = Strongly Agree to 4 = Strongly Disagree (reverse coded). Four items per each subscale were assessed in the Y1F, Y1S, and Y2S surveys, and the one best‐performing item (based on factor loadings) from each subscale was included in the Y3S and Y4S surveys due to space limitations. However, descriptive statistics (see Table 1) and correlations across waves showed that the single‐item scores performed similarly to the multi‐item scale scores.\nDescriptive statistics for drinking motive scores.\nIn each survey, participants who had initiated drinking completed an abbreviated version of the Drinking Motives Questionnaire—Revised (Cooper, 1994). This scale contains four subscales whose items are summed to create scores: Coping (e.g., “because it helps me when I feel depressed or nervous”), Enhancement (e.g., “because it gives me a pleasant feeling”), Conformity (e.g., “to get in with a group I like”), and Social (e.g., “because it makes social gatherings more fun”). Responses are on a Likert‐like scale from 1 = Strongly Agree to 4 = Strongly Disagree (reverse coded). Four items per each subscale were assessed in the Y1F, Y1S, and Y2S surveys, and the one best‐performing item (based on factor loadings) from each subscale was included in the Y3S and Y4S surveys due to space limitations. However, descriptive statistics (see Table 1) and correlations across waves showed that the single‐item scores performed similarly to the multi‐item scale scores.\nDescriptive statistics for drinking motive scores.\n[SUBTITLE] Alcohol use [SUBSECTION] At each wave, participants were asked if they had initiated alcohol use, and, if so, about alcohol use behaviors including typical consumption (gm EtOH/month) and symptoms of DSM‐5 Alcohol Use Disorder (AUDsx), which were measured using the Semi‐Structured Assessment for the Genetics of Alcoholism (SSAGA; Bucholz et al., 1994) and summed.\nAt each wave, participants were asked if they had initiated alcohol use, and, if so, about alcohol use behaviors including typical consumption (gm EtOH/month) and symptoms of DSM‐5 Alcohol Use Disorder (AUDsx), which were measured using the Semi‐Structured Assessment for the Genetics of Alcoholism (SSAGA; Bucholz et al., 1994) and summed.\n[SUBTITLE] Environmental risk and protective factors [SUBSECTION] The S4S surveys assess numerous environmental and psychosocial constructs that have demonstrated associations with alcohol use and psychopathology. Among these are parenting behaviors, specifically the Involvement and Autonomy Granting subscales of Steinberg's Parenting Style scale (Steinberg et al., 1992), peer deviance (the proportion of one's friends who engage in deviant behaviors such as getting drunk and cutting school, as described by Kendler et al., 2008), and exposure to traumatic events such as an assault or natural disaster, measured by the Life Events Checklist (Gray et al., 2004).\nThe S4S surveys assess numerous environmental and psychosocial constructs that have demonstrated associations with alcohol use and psychopathology. Among these are parenting behaviors, specifically the Involvement and Autonomy Granting subscales of Steinberg's Parenting Style scale (Steinberg et al., 1992), peer deviance (the proportion of one's friends who engage in deviant behaviors such as getting drunk and cutting school, as described by Kendler et al., 2008), and exposure to traumatic events such as an assault or natural disaster, measured by the Life Events Checklist (Gray et al., 2004).", "In each survey, participants who had initiated drinking completed an abbreviated version of the Drinking Motives Questionnaire—Revised (Cooper, 1994). This scale contains four subscales whose items are summed to create scores: Coping (e.g., “because it helps me when I feel depressed or nervous”), Enhancement (e.g., “because it gives me a pleasant feeling”), Conformity (e.g., “to get in with a group I like”), and Social (e.g., “because it makes social gatherings more fun”). Responses are on a Likert‐like scale from 1 = Strongly Agree to 4 = Strongly Disagree (reverse coded). Four items per each subscale were assessed in the Y1F, Y1S, and Y2S surveys, and the one best‐performing item (based on factor loadings) from each subscale was included in the Y3S and Y4S surveys due to space limitations. However, descriptive statistics (see Table 1) and correlations across waves showed that the single‐item scores performed similarly to the multi‐item scale scores.\nDescriptive statistics for drinking motive scores.", "At each wave, participants were asked if they had initiated alcohol use, and, if so, about alcohol use behaviors including typical consumption (gm EtOH/month) and symptoms of DSM‐5 Alcohol Use Disorder (AUDsx), which were measured using the Semi‐Structured Assessment for the Genetics of Alcoholism (SSAGA; Bucholz et al., 1994) and summed.", "The S4S surveys assess numerous environmental and psychosocial constructs that have demonstrated associations with alcohol use and psychopathology. Among these are parenting behaviors, specifically the Involvement and Autonomy Granting subscales of Steinberg's Parenting Style scale (Steinberg et al., 1992), peer deviance (the proportion of one's friends who engage in deviant behaviors such as getting drunk and cutting school, as described by Kendler et al., 2008), and exposure to traumatic events such as an assault or natural disaster, measured by the Life Events Checklist (Gray et al., 2004).", "Information about genotyping for this sample has been described in detail elsewhere (Dick et al., 2014; Webb et al., 2017). Briefly, samples were genotyped on the Axiom BioBank Array, Catalog Version 2 (Affymetrix Inc., Santa Clara, CA) and imputed to the 1000 Genomes phase 3 all‐ancestries reference panel (The 1000 Genomes Project Consortium, 2015) after removing poor quality SNPs (missingness >5%, Hardy–Weinberg equilibrium [HWE] p values <5 × 10−6) and individual samples (genotyping rate < 98%, heterozygosity outliers, phenotypic/genotypic sex discordance, excess relatedness). Genetic ancestry principal components (PCs) were derived from the 1000 Genomes (phase 3) full reference population and projected onto the S4S samples to identify genetically homogenous ancestral groups for analysis, as described by Peterson et al. (2017). After this procedure, individuals from five continental ancestral super‐populations were available for analysis: Africa (AFR), the Americas (AMR), East Asia (EAS), Europe (EUR), and South Asia (SAS). Within‐group ancestry PCs were then calculated within each of these super‐populations in order to capture fine‐grained differences in allele frequencies that could contribute to residual population stratification. Within groups, additional quality control steps were taken to remove reference population outliers, those with excess relatedness (pi‐hat > 0.1), and SNPs with low ancestry‐specific minor allele frequency (MAF)/HWE.", "Data analysis involved two parts. First, we examined drinking motives at a phenotypic level in the full sample. We estimated the Pearson and intraclass correlations between motives across survey waves to estimate their temporal stability, calculated the Pearson correlations between motives and alcohol consumption/AUDsx cross‐sectionally and prospectively (Y1F motives with Y1F‐Y4S alcohol measures), and conducted linear regression analyses to examine how demographic and environmental characteristics predicted mean levels of each type of drinking motive.\nSecond, we conducted a series of analyses on the genotypic data to investigate the genetic etiology of drinking motives. The genotyped sample (n = 6325) was restricted to include only unrelated individuals with nonmissing phenotype information—i.e., those who initiated alcohol use during the time the measures were collected and chose to answer the relevant survey questions (analytic n = 4855). We used the mean of all available scores across waves (Table 1) for the four drinking motives subscales as the outcome phenotypes. We also utilized relevant covariates to control for possible confounding effects on genetic associations with the phenotypes, which included sex, age (mean across waves where participants had nonmissing drinking motives scores), and within‐ancestry PCs.\nWe estimated the heritability of drinking motives that could be attributable to measured genetic variants (SNP heritability; h\n\n2\n\nSNP) through the use of genome‐wide complex trait analysis (GCTA; Yang et al., 2011). Genotyped SNPs were filtered for a MAF >0.01 before calculating the genomic relatedness matrix between individuals. GCTA was then run separately in each ancestry using the first 10 within‐ancestry PCs, age, and sex as covariates, and the resulting heritability estimates were meta‐analyzed with a fixed‐effects, inverse variance‐weighted scheme.\nNext, we conducted a genome‐wide association (GWAS) of the imputed genetic variants. SNPs were filtered for imputation quality (INFO score >0.5) and ancestry‐specific MAF corresponding to a minor allele count (MAC) > 100. Previous work has established that a MAC > 40 allows for reliable statistical estimation (Bigdeli et al., 2014); we use a more conservative threshold because the somewhat skewed distribution of the phenotypes. Within‐ancestry PCs to include as covariates were decided based on their association with the phenotype in a stepwise linear regression to avoid overfitting (c.f. Webb et al., 2017). The specific PCs used in each analysis are shown in Table S1. GWAS was conducted using SNPTEST (Marchini et al., 2007), again running separately in each ancestry group and meta‐analyzing the association test for each SNP using inverse‐variance weighting in METAL (Abecasis et al., 2012). We present only the results for SNPs that were available in at least 1000 individuals (meaning that the SNP had to pass quality control filters in either the AFR, EUR, or a combination of two or more ancestry sub‐groups), to be certain that spurious results from small samples were not given undue consideration. Multiple testing correction was performed by using a Bonferroni‐corrected threshold for genome‐wide significance of 5 × 10−8 and calculating false discovery rates (FDR) using the qvalue package for R/Bioconductor (Storey et al., 2015). Given the small size of our sample and its corresponding statistical power (see Discussion), we also examine “suggestive” signals (p < 5 × 10−6) and ancestry‐specific results in the largest subgroup (EUR; n = 2537), although these are given lower credibility. Regional association plots for associated loci were visualized using LocusZoom (Pruim et al., 2010). Significant and suggestive signals were uploaded to FUMA (Watanabe et al., 2017) to define candidate genomic loci based on LD (variants with r\n\n2\n > 0.6 with the lead SNP, using the 1000 Genomes all‐ancestries reference panel for the LD structure [The 1000 Genomes Project Consortium, 2015]), annotate functional consequences of SNPs in the loci, and map candidate genes to these association signals based on position (within 10 kb), known expression quantitative trait locus (eQTL) effects, or 3‐dimensional chromatin interactions.\nAfter conducting GWAS at the individual variant level, we applied gene‐based and pathway‐based association analyses to test whether the SNP association signal was enriched at these aggregate levels, using MAGMA v.1.08 (de Leeuw et al., 2015). We used the 1000 Genomes as reference panels for the LD structure of each of the five ancestral continental superpopulations. After annotation of the SNPs to genes, genes were also grouped into pathways based on the curated hallmark and canonical pathways gene‐sets from the Molecular Signatures Database (Liberzon et al., 2015). These pathways represent groups of genes whose products are involved in known metabolic and regulatory biochemical processes. Gene‐based analyses were conducted on the ancestry‐specific SNPTEST results and meta‐analyzed with MAGMA, using an inverse variance‐weighted Stouffer's Z test. Pathway analyses were conducted on the meta‐analyzed, transancestral gene‐based results. We also used Popcorn (Brown et al., 2016) to estimate the genetic correlation of the GWAS results across ancestry groups, using the 1000 Genomes as a reference for LD patterns in each ancestry group and calculating the genetic impact correlation (the similarity between SNP effect sizes across ancestry groups, while accounting for differences in allele frequencies).\nFinally, we investigated a key endophenotype criterion for drinking motives by estimating their genetic overlap with alcohol‐related phenotypes. Using LD score regression (LDSC; Bulik‐Sullivan et al., 2015), we estimated h\n\n2\n\nSNP from the GWAS summary statistics and, if there was sufficient signal (mean χ2 > 1), we used LDSC to estimate the genome‐wide genetic correlations with three alcohol phenotypes from publicly available GWAS results: consumption (Liu et al., 2019), AUDIT total scores (Sanchez‐Roige et al., 2019), and AUD diagnoses (Walters et al., 2018). As LDSC assumptions rest on ancestry‐specific LD patterns, we used only results of the drinking motives GWAS from the largest group, the EUR ancestry subset (n = 2537), and conducted analyses using EUR LD scores provided with the software and recommended analysis settings (INFO >0.9, MAF >0.01, HapMap3 SNPs). LDSC provides only a global, genome‐wide estimate of the correlation, so we additionally conducted local genetic correlation for a more fine‐grained approach. We curated a list of 98 known alcohol‐related loci based on genome‐wide significant findings from well‐powered GWASs (Liu et al., 2019; Sanchez‐Roige et al., 2019; Walters et al., 2018; Zhou et al., 2020), using a 250 kb window around the lead SNP if a single position rather than a region was provided and merging overlapping loci (within 250 kb) within and across studies. Using LAVA (Werme et al., 2022), we investigated the local genetic correlation between drinking motives (EUR ancestry results) and the same three alcohol‐related phenotypes as for the genome‐wide correlations (consumption, AUDIT‐T, and AUD) within these curated loci. Bivariate correlations were calculated only when there was enrichment (p < 0.0001) in the univariate heritability for both phenotypes at a locus.", "[SUBTITLE] Phenotypic etiology [SUBSECTION] Descriptive statistics for drinking motives across survey waves are shown in Table 1. Mean levels for all four motives increased slightly but remained relatively stable across college. Correlations between each of the motives across time are shown in Table 2. Although there was some decay in the strength of correlation between longer periods of time (e.g., year 1 with year 4), each pair of adjacent time points was moderately correlated (Pearson r = 0.37 to 0.56, p < 5 × 10−20), with excellent overall stability (ICC = 0.75 to 0.81). Conformity motives showed the weakest correlations across time but also had the lowest levels of endorsement. Consistent with previous research, there were significant positive cross‐sectional and longitudinal correlations between motives and alcohol use outcomes, particularly between coping motives and AUDsx and between enhancement/social motives and both consumption and AUDsx (Table 3).\nCross‐time correlations for drinking motives across five waves of assessment.\n\nNote: *p < 5e‐10, **p < 5e‐20, ***p < 5e‐100.\nCross‐sectional and prospective correlations between drinking motives and alcohol use outcomes.\n\nNote: Bolded values are significant after multiple testing correction for 56 tests, adjusted alpha = 0.0009.\nAbbreviations: AUDsx, alcohol use disorder symptom count; Y1F‐Y4S, measurement time from Year 1 Fall to Year 4 Spring.\nResults from the linear models of predictors of drinking motives are displayed in Table 4. Male gender, older age, and recent trauma exposure were associated with higher levels of conformity motives. Students from racial and ethnic minorities generally had lower or not significantly different levels of all four drinking motives when compared with White students, with the exception that self‐reported Asian ethnicity was associated with higher levels of conformity motives. Parental autonomy granting was associated with lower levels of all drinking motives, while higher parental involvement was associated with higher levels of positive reinforcement (social and enhancement) motives. Peer deviance was associated with higher levels of all motives except conformity. Lifetime (pre‐college) trauma exposure was simultaneously associated with both higher coping motives and lower social motives.\nLinear regression results of demographic and environmental factors predicting drinking motives.\n\nNote: Reference category for the Ethnicity measure is White. Bolded values are significant after multiple testing correction for 8 predictor variables, adjusted alpha = 0.0062.\nDescriptive statistics for drinking motives across survey waves are shown in Table 1. Mean levels for all four motives increased slightly but remained relatively stable across college. Correlations between each of the motives across time are shown in Table 2. Although there was some decay in the strength of correlation between longer periods of time (e.g., year 1 with year 4), each pair of adjacent time points was moderately correlated (Pearson r = 0.37 to 0.56, p < 5 × 10−20), with excellent overall stability (ICC = 0.75 to 0.81). Conformity motives showed the weakest correlations across time but also had the lowest levels of endorsement. Consistent with previous research, there were significant positive cross‐sectional and longitudinal correlations between motives and alcohol use outcomes, particularly between coping motives and AUDsx and between enhancement/social motives and both consumption and AUDsx (Table 3).\nCross‐time correlations for drinking motives across five waves of assessment.\n\nNote: *p < 5e‐10, **p < 5e‐20, ***p < 5e‐100.\nCross‐sectional and prospective correlations between drinking motives and alcohol use outcomes.\n\nNote: Bolded values are significant after multiple testing correction for 56 tests, adjusted alpha = 0.0009.\nAbbreviations: AUDsx, alcohol use disorder symptom count; Y1F‐Y4S, measurement time from Year 1 Fall to Year 4 Spring.\nResults from the linear models of predictors of drinking motives are displayed in Table 4. Male gender, older age, and recent trauma exposure were associated with higher levels of conformity motives. Students from racial and ethnic minorities generally had lower or not significantly different levels of all four drinking motives when compared with White students, with the exception that self‐reported Asian ethnicity was associated with higher levels of conformity motives. Parental autonomy granting was associated with lower levels of all drinking motives, while higher parental involvement was associated with higher levels of positive reinforcement (social and enhancement) motives. Peer deviance was associated with higher levels of all motives except conformity. Lifetime (pre‐college) trauma exposure was simultaneously associated with both higher coping motives and lower social motives.\nLinear regression results of demographic and environmental factors predicting drinking motives.\n\nNote: Reference category for the Ethnicity measure is White. Bolded values are significant after multiple testing correction for 8 predictor variables, adjusted alpha = 0.0062.\n[SUBTITLE] Genetic etiology [SUBSECTION] [SUBTITLE] \nSNP Heritability [SUBSECTION] Heritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample.\nHeritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample.\n[SUBTITLE] GWAS [SUBSECTION] Manhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3.\nManhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives.\nGenomic annotation for top loci associated with drinking motives.\n\nNote: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci.\nThe genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r\n2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07).\nFor the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7.\nFor the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS.\nGWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives.\nManhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3.\nManhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives.\nGenomic annotation for top loci associated with drinking motives.\n\nNote: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci.\nThe genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r\n2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07).\nFor the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7.\nFor the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS.\nGWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives.\n[SUBTITLE] \nSNP Heritability [SUBSECTION] Heritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample.\nHeritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample.\n[SUBTITLE] GWAS [SUBSECTION] Manhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3.\nManhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives.\nGenomic annotation for top loci associated with drinking motives.\n\nNote: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci.\nThe genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r\n2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07).\nFor the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7.\nFor the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS.\nGWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives.\nManhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3.\nManhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives.\nGenomic annotation for top loci associated with drinking motives.\n\nNote: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci.\nThe genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r\n2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07).\nFor the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7.\nFor the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS.\nGWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives.\n[SUBTITLE] Genetic correlation [SUBSECTION] LDSC indicated no significant genome‐wide heritability based on the SNP association signals, and a mean χ2 < 1 for social, enhancement, and conformity motives, making them unsuitable for further LDSC analyses. However, there was sufficient SNP heritability for coping motives (h\n\n2\n\nSNP = 0.56, SE = 0.21, mean χ2 = 1.20) to conduct genetic correlation analyses with external GWAS of alcohol‐related phenotypes. After Bonferroni correction for three tests (alpha = 0.05/3 = 0.017), these showed a significant correlation between coping motives and AUD diagnoses (r\ng = 0.71, p = 0.001), but not alcohol consumption (r\ng = 0.16, p = 0.069) or AUDIT‐T scores (r\ng = 0.05, p = 0.644).\nLocal genetic correlation was carried out to obtain a more fine‐grained view of the genetic relationship between drinking motives and alcohol phenotypes. Examining 98 known alcohol‐related loci, we saw local enrichment (p < 0.0001) for heritability in 13, 20, 44, and 11 of the 98 loci for social, enhancement, coping, and conformity motives, respectively (Table S6). These all exceed the number of loci expected by chance at an alpha of 0.05 (binomial test p = 3.76 × 10−04, 1.43 × 10−04, 3.68 × 10−32 and 0.004, respectively). Local genetic correlations suggested some interesting findings, for example, negative genetic correlations between coping motives and (1) consumption (r\ng = −0.27, p = 0.004), (2) AUDIT‐T (r\ng = −0.24, p = 0.041), and (3) AUD (r\ng = −0.34, p = 0.013) at the ADH locus (chr4:96,764,066‐101,983,024). However, no local genetic correlations survived multiple testing correction.\nTrans‐ancestral genetic correlation analysis using Popcorn was limited by the low SNP heritability, so estimates were not calculable for many pairs of GWAS summary statistics (Table S7). For the correlations that could be calculated, there was no evidence that the SNP effects on drinking motives differed between ancestry groups: the correlation between ancestry groups did not significantly differ from r = 1.0 (all p's > 0.19).\nLDSC indicated no significant genome‐wide heritability based on the SNP association signals, and a mean χ2 < 1 for social, enhancement, and conformity motives, making them unsuitable for further LDSC analyses. However, there was sufficient SNP heritability for coping motives (h\n\n2\n\nSNP = 0.56, SE = 0.21, mean χ2 = 1.20) to conduct genetic correlation analyses with external GWAS of alcohol‐related phenotypes. After Bonferroni correction for three tests (alpha = 0.05/3 = 0.017), these showed a significant correlation between coping motives and AUD diagnoses (r\ng = 0.71, p = 0.001), but not alcohol consumption (r\ng = 0.16, p = 0.069) or AUDIT‐T scores (r\ng = 0.05, p = 0.644).\nLocal genetic correlation was carried out to obtain a more fine‐grained view of the genetic relationship between drinking motives and alcohol phenotypes. Examining 98 known alcohol‐related loci, we saw local enrichment (p < 0.0001) for heritability in 13, 20, 44, and 11 of the 98 loci for social, enhancement, coping, and conformity motives, respectively (Table S6). These all exceed the number of loci expected by chance at an alpha of 0.05 (binomial test p = 3.76 × 10−04, 1.43 × 10−04, 3.68 × 10−32 and 0.004, respectively). Local genetic correlations suggested some interesting findings, for example, negative genetic correlations between coping motives and (1) consumption (r\ng = −0.27, p = 0.004), (2) AUDIT‐T (r\ng = −0.24, p = 0.041), and (3) AUD (r\ng = −0.34, p = 0.013) at the ADH locus (chr4:96,764,066‐101,983,024). However, no local genetic correlations survived multiple testing correction.\nTrans‐ancestral genetic correlation analysis using Popcorn was limited by the low SNP heritability, so estimates were not calculable for many pairs of GWAS summary statistics (Table S7). For the correlations that could be calculated, there was no evidence that the SNP effects on drinking motives differed between ancestry groups: the correlation between ancestry groups did not significantly differ from r = 1.0 (all p's > 0.19).", "Descriptive statistics for drinking motives across survey waves are shown in Table 1. Mean levels for all four motives increased slightly but remained relatively stable across college. Correlations between each of the motives across time are shown in Table 2. Although there was some decay in the strength of correlation between longer periods of time (e.g., year 1 with year 4), each pair of adjacent time points was moderately correlated (Pearson r = 0.37 to 0.56, p < 5 × 10−20), with excellent overall stability (ICC = 0.75 to 0.81). Conformity motives showed the weakest correlations across time but also had the lowest levels of endorsement. Consistent with previous research, there were significant positive cross‐sectional and longitudinal correlations between motives and alcohol use outcomes, particularly between coping motives and AUDsx and between enhancement/social motives and both consumption and AUDsx (Table 3).\nCross‐time correlations for drinking motives across five waves of assessment.\n\nNote: *p < 5e‐10, **p < 5e‐20, ***p < 5e‐100.\nCross‐sectional and prospective correlations between drinking motives and alcohol use outcomes.\n\nNote: Bolded values are significant after multiple testing correction for 56 tests, adjusted alpha = 0.0009.\nAbbreviations: AUDsx, alcohol use disorder symptom count; Y1F‐Y4S, measurement time from Year 1 Fall to Year 4 Spring.\nResults from the linear models of predictors of drinking motives are displayed in Table 4. Male gender, older age, and recent trauma exposure were associated with higher levels of conformity motives. Students from racial and ethnic minorities generally had lower or not significantly different levels of all four drinking motives when compared with White students, with the exception that self‐reported Asian ethnicity was associated with higher levels of conformity motives. Parental autonomy granting was associated with lower levels of all drinking motives, while higher parental involvement was associated with higher levels of positive reinforcement (social and enhancement) motives. Peer deviance was associated with higher levels of all motives except conformity. Lifetime (pre‐college) trauma exposure was simultaneously associated with both higher coping motives and lower social motives.\nLinear regression results of demographic and environmental factors predicting drinking motives.\n\nNote: Reference category for the Ethnicity measure is White. Bolded values are significant after multiple testing correction for 8 predictor variables, adjusted alpha = 0.0062.", "[SUBTITLE] \nSNP Heritability [SUBSECTION] Heritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample.\nHeritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample.\n[SUBTITLE] GWAS [SUBSECTION] Manhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3.\nManhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives.\nGenomic annotation for top loci associated with drinking motives.\n\nNote: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci.\nThe genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r\n2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07).\nFor the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7.\nFor the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS.\nGWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives.\nManhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3.\nManhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives.\nGenomic annotation for top loci associated with drinking motives.\n\nNote: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci.\nThe genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r\n2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07).\nFor the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7.\nFor the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS.\nGWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives.", "Heritability estimates from GCTA are displayed in Table S2, with meta‐analysis estimates ranging from 14% (coping) to 22% (enhancement). However, none of the meta‐analytic estimates were significantly differentiable from zero in this sample.", "Manhattan plots for the SNP‐based GWAS results are shown in Figure 1 (cross‐ancestry meta‐analysis) and Figures S1 and S2 (EUR ancestry subset). There was little evidence of inflation that could indicate bias or population stratification; the median chi‐square statistic (λ) was 0.948–1.021 and the scaled statistic (λ1000) was 1.000 for each meta‐analysis. For social motives, there was no evidence for any locus reaching genome‐wide significance (p < 5 × 10−08), and little evidence of even suggestive association peaks (p < 5 × 10−06). For the other three motive types, at least one genome‐wide significant SNP was identified in addition to several suggestive loci (regional locus plots in Figures S3–S8). The significant and suggestive loci are shown in Table 5. Because true association signals should be found in LD blocks, rather than lone SNPs, Table 5 shows suggestive loci only when three or more SNPs in the same position (±10 kb) pass the suggestive significance threshold. Additional FUMA annotation information on suggestive/significant loci can be found in Table S3.\nManhattan plot of genome‐wide association meta‐analysis results for (A) social, (B) enhancement, (C) coping, and (D) conformity drinking motives.\nGenomic annotation for top loci associated with drinking motives.\n\nNote: Loci are defined by the presence of genome‐wide significant (p < 5e‐08) SNPs or peaks with three or more suggestive (p < 5e‐06) SNPs. Position is the location of the lead SNP (build GRCh37). Local genes are defined by locus position within 10 kb of the gene. *eQTL and chromatin interaction (CI) mapping performed with FUMA only for genome‐wide significant loci.\nThe genome‐wide significant locus for enhancement motives included 2 SNPs found only in the EUR ancestry group, located on chromosome 3 in the FBLN2 (fibulin 2) gene, which codes for an extracellular matrix protein involved in organ development and differentiation. The regional association plot (Figure S3) showed little association enrichment for SNPs in high LD with the lead SNP, indicating this may be a spurious association. However, FUMA annotation of this region revealed a candidate nonsynonymous exonic variant in FBLN2, rs113265853, which was in LD (r\n2 = 0.73) with the lead genome‐wide significant SNP rs149867189. This exonic variant was not directly analyzed but was present in the LD reference panel. Though it may be a spurious signal, an alternate possibility is that the association signal may be tagging this rare (1000 Genomes MAF = 0.01) functional variant. Examining the EUR ancestry‐specific GWAS for enhancement motives, there was also a peak just below genome‐wide significance (p = 6.72 × 10−08), in the PECR (peroxisomal trans‐2‐enoyl‐CoA reductase) gene on chromosome 2 (Figures S1 and S4). This gene has been previously linked to alcohol dependence (Treutlein et al., 2009) and also had a suggestive association in the cross‐ancestry meta‐analysis (p = 8.24 × 10−07).\nFor the coping motives meta‐analysis, one genome‐wide significant SNP was found in a peak in an intergenic region on chromosome 5 with no nearby genes, although the gene CDH6 was mapped by chromatin interactions with this locus in nine tissues, including stem cells, heart, and liver. The EUR ancestry‐specific GWAS identified additional genome‐wide significant loci in peaks on chromosome 9 (Intergenic, 25 kb from the gene GRIN3A) and 15 (in the pseudogene LOC390617) and a lone significant SNP on chromosome 7 (intergenic), see Figures S5–S7.\nFor the conformity motives meta‐analysis, one genome‐wide significant SNP alongside a peak of 9 suggestive SNPs was found on chromosome 12 in the SIRT4 (sirtuin 4) gene (Figure S8). No additional significant loci were identified in the EUR ancestry‐specific GWAS.\nGWAS summary statistics from the individual ancestry groups were carried forward into gene‐based enrichment testing using MAGMA and meta‐analyzed, and the resulting (meta‐analyzed) gene‐based P values were tested for enrichment in biological pathways. Results from the gene‐based and pathway‐based association analyses are presented in Tables S4 and S5. After Bonferroni correction for the number of tested genes/pathways, there was no evidence for significant enrichment of the SNP association signal in any specific genes or pathways for any of the four motives.", "LDSC indicated no significant genome‐wide heritability based on the SNP association signals, and a mean χ2 < 1 for social, enhancement, and conformity motives, making them unsuitable for further LDSC analyses. However, there was sufficient SNP heritability for coping motives (h\n\n2\n\nSNP = 0.56, SE = 0.21, mean χ2 = 1.20) to conduct genetic correlation analyses with external GWAS of alcohol‐related phenotypes. After Bonferroni correction for three tests (alpha = 0.05/3 = 0.017), these showed a significant correlation between coping motives and AUD diagnoses (r\ng = 0.71, p = 0.001), but not alcohol consumption (r\ng = 0.16, p = 0.069) or AUDIT‐T scores (r\ng = 0.05, p = 0.644).\nLocal genetic correlation was carried out to obtain a more fine‐grained view of the genetic relationship between drinking motives and alcohol phenotypes. Examining 98 known alcohol‐related loci, we saw local enrichment (p < 0.0001) for heritability in 13, 20, 44, and 11 of the 98 loci for social, enhancement, coping, and conformity motives, respectively (Table S6). These all exceed the number of loci expected by chance at an alpha of 0.05 (binomial test p = 3.76 × 10−04, 1.43 × 10−04, 3.68 × 10−32 and 0.004, respectively). Local genetic correlations suggested some interesting findings, for example, negative genetic correlations between coping motives and (1) consumption (r\ng = −0.27, p = 0.004), (2) AUDIT‐T (r\ng = −0.24, p = 0.041), and (3) AUD (r\ng = −0.34, p = 0.013) at the ADH locus (chr4:96,764,066‐101,983,024). However, no local genetic correlations survived multiple testing correction.\nTrans‐ancestral genetic correlation analysis using Popcorn was limited by the low SNP heritability, so estimates were not calculable for many pairs of GWAS summary statistics (Table S7). For the correlations that could be calculated, there was no evidence that the SNP effects on drinking motives differed between ancestry groups: the correlation between ancestry groups did not significantly differ from r = 1.0 (all p's > 0.19).", "In this set of analyses, we have uncovered several insights about the nature of drinking motives in college students. First, drinking motives are stable throughout college and represent reliable psychosocial constructs. Second, some robust environmental predictors of alcohol misuse like parental autonomy granting and peer deviance have broad associations with all types of drinking motives, while others, like trauma exposure, have more specific associations. For example, parental autonomy granting predicted lower levels of all drinking motives and peer drinking predicted higher levels of all drinking motives, while lifetime trauma exposure was simultaneously associated with lower social motives and higher coping motives. Third, the investigation into the genetic etiology of drinking motives in college students identified some promising but, as of yet, largely inconclusive results. We found substantial SNP heritability estimates (positive reinforcement motives: 16–22%; negative reinforcement motives: 14–16%). However, these estimates were not significantly different from zero—not surprising given the current sample size—so the ability for inference is limited. In genetic association testing, several loci were identified with suggestive or marginally significant effects, although the results of the gene‐based and pathway‐based analyses showed little evidence of enrichment at the aggregate levels. Genetic correlation analyses indicated substantial sharing of genetic factors between coping motives and AUD that merits follow‐up, though large standard errors again point to the need for replication in more robust samples.\nThese findings have encouraging implications for the use of drinking motives as endophenotypes of internalizing and externalizing pathways to alcohol misuse. Their stability across time is important for establishing that these are trait‐like outcomes linked to enduring temperamental dimensions (e.g., personality), and thus are viable for aiding gene identification efforts (Gottesman & Gould, 2003). These results were consistent with estimates in a slightly older sample across 10 years (Windle & Windle, 2018) and somewhat higher than those of a similar aged sample that looked across a shorter (1 year) time frame (Labhart et al., 2016), indicating high stability when motives are considered in the long‐term. However, their moderate correlations across shorter intervals also point to their potential to be modified via environmental interventions.\nA few promising results from the genetic analyses are also worth considering. First, we found a suggestive association with enhancement motives in the PECR gene in Europeans. Although not quite reaching the threshold of genome‐wide significance, this signal showed a clear peak with enrichment of association signal in a large number of SNPs within a single locus, which bolsters confidence that it is a true effect. This gene is highly expressed in the liver and has been previously implicated in a GWAS of early onset alcohol dependence (Treutlein et al., 2009). Such evidence is consistent with the hypothesized connection between enhancement motives and an externalizing pathway/subtype of alcohol dependence characterized by early age of onset and stronger genetic influences (e.g., Cloninger et al., 1988). The PECR region (±10 kb, chr2:216893111‐216956539) has not been implicated in any larger GWAS of alcohol phenotypes since the Treutlein et al. (2009) study, although no such study has focused specifically on a severe/early‐onset subtype. This region had minimal association with alcohol consumption (p = 0.0015), AUDIT total scores (p = 0.0071), or AUD (p = 0.0061) in recent large‐scale GWAS analyses (Liu et al., 2019; Sanchez‐Roige et al., 2019; Walters et al., 2018). There was also evidence for a genome‐wide significant association in the SIRT4 gene with conformity motives. This gene is a close relative of the SIRT1 gene that was implicated in the genetic etiology of major depression (CONVERGE Consortium, 2015) and may suggest a common predisposition shared between internalizing psychopathology and this negative reinforcement motive. However, there is also a large number of other genes linked to this locus by positional, eQTL, and chromatin interaction mapping, any of which could be driving the identified association effect. The SIRT4 region (±10 kb, chr12:120730124‐120761045) had minimal association with alcohol consumption (p = 0.0026), AUDIT total scores (p = 0.0004), or AUD (p = 0.0370) in recent large‐scale GWAS analyses (Liu et al., 2019; Sanchez‐Roige et al., 2019; Walters et al., 2018).\nAn unusual result from the local genetic correlation analysis was the finding of a negative correlation (r\ng = −0.34) between coping motives and AUD at the ADH locus, despite an overall positive genome‐wide correlation (r\ng = 0.71). Although this result did not survive multiple testing correction, the strong biological evidence for ADH and the consistency of the negative genetic correlation across related measures (r\ng = −0.27 with consumption and r\ng = −0.24 with AUDIT scores) indicates that this may be worth further, cautious, consideration. We speculate that this may be a consequence of the relatively specific association of coping motives with alcohol problems/AUD, rather than with heaviness of consumption (Bresin & Mekawi, 2021; Kuntsche et al., 2005). Variants in the ADH genes are, of course, associated with multiple domains of alcohol use/misuse, but their foremost effect is on heaviness of consumption due to lowered sensitivity to the intoxicating effects of EtOH (Hart & Kranzler, 2015). Therefore while the aggregate genetic effects on AUD are positively correlated with coping motives, the specific effects of ADH variants are not. However, it's still surprising to see a negative genetic correlation rather than simply a null or weaker positive one. Another possible explanation is that coping motives primarily reflect a negative reinforcement mechanism while those with lower sensitivity to intoxication due to ADH variants are pursuing positive reinforcement, (i.e., chasing a high), and drinking more to achieve that reward state rather than to escape from a negative affective state.\nWe conclude that our findings at this stage thus provide only modest insight into the biology underlying drinking motives and their potential genetic pathways to alcohol misuse. This is perhaps unsurprising given trends in gene identification efforts for complex, and particularly psychiatric/behavioral traits. The emerging landscape of the field indicates that tens if not hundreds of thousands of samples may be needed for credible GWAS results, even for “less genetically complex” endophenotypes (Flint & Munafo, 2007). Power calculations with the Genetic Power Calculator (Purcell et al., 2003) indicated that we had 0.5–22% power to detect individual SNP associations with a typical effect size for a complex trait (MAF = 0.10, explaining 0.2–0.5% of the trait variance), and that a sample size of at least 8500–21,500 would be needed to detect SNP effects in that range. Our sample had >80% power to detect larger effect sizes (e.g., 1% of trait variance explained), which might be expected either from larger effects on endophenotypes that lie closer together in biological pathways, or from the aggregation of individual SNP effects into genes and pathways. Whether or not they prove to be simpler genetic targets, endophenotypes retain uniquely valuable roles, such as their ability to provide insight into the etiological mechanisms of a distal target phenotype and allow for prospective identification of individuals at risk (Iacono et al., 2017).\nIn the first reported GWAS of drinking motives, we already show promising results for an endophenotype—plausible candidate genes and robust genetic correlation with alcohol misuse measures—in a small sample of a few thousand participants, suggesting that GWAS samples for drinking motives may not need to be as large as for alcohol misuse measures to achieve similar gains in gene identification progress. Further, unlike many endophenotypes (e.g., brain imaging, neurophysiological measures) whose main drawback is their costliness to collect, drinking motives are reliably measured by a simple survey and could be collected at scale for GWAS in much the same way as alcohol misuse measures are now, possibly with a much greater return on investment. Recently developed statistical methods, such as genomic structural equation modeling (Grotzinger et al., 2019) might provide a means to investigate the overlapping and/or mediational role of drinking motives in the genetic etiology of alcohol misuse. The potential insights that drinking motives can provide into the mechanisms underlying alcohol misuse underscores their value for study with larger samples and more powerful study designs in the future.", "The authors report no financial disclosures or potential conflicts of interest.", "\nFigures S1–S8\n\nClick here for additional data file.\n\nTables S1–S7\n\nClick here for additional data file." ]
[ null, "materials-and-methods", null, null, null, null, null, null, null, "results", null, null, null, null, null, "discussion", "COI-statement", "supplementary-material" ]
[ "alcohol misuse", "college students", "drinking motives", "endophenotype", "GWAS" ]
Blocking connexin 43 hemichannel-mediated ATP release reduces communication within and between tubular epithelial cells and medullary fibroblasts in a model of diabetic nephropathy.
36256487
Fibrosis of renal tubules is the final common pathway in diabetic nephropathy and develops in the face of tubular injury and fibroblast activation. Aberrant connexin 43 (Cx43) hemichannel activity has been linked to this damage under euglycaemic conditions, however, its role in glycaemic injury is unknown. This study investigated the effect of a Cx43 blocker (Tonabersat) on hemichannel activity and cell-cell interactions within and between tubular epithelial cells and fibroblasts in an in vitro model of diabetic nephropathy.
INTRODUCTION
Human kidney (HK2) proximal tubule epithelial cells and medullary fibroblasts (TK173) were treated in low (5 mM) or high (25 mM) glucose ± transforming growth factor beta-1 (TGFβ1) ± Tonabersat in high glucose. Carboxyfluorescein dye uptake and ATPlite luminescence assessed changes in hemichannel-mediated ATP release, while immunoblotting determined protein expression. Co-incubation with the ATP-diphosphohydrolase apyrase or a P2X7R inhibitor (A438079) assessed ATP-P2X7R signalling. Indirect co-culture with conditioned media from the alternate cell type evaluated paracrine-mediated heterotypic interactions.
METHODS
Tonabersat partially negated glucose/TGFβ1-induced increases in Cx43 hemichannel-mediated ATP release and downstream changes in adherens junction and extracellular matrix (ECM) protein expression in HK2 and TK173 cells. Apyrase and A438079 highlighted the role for ATP-P2X7R in driving changes in protein expression in TK173 fibroblasts. Indirect co-culture studies suggest that epithelial cell secretome increases Tonabersat-sensitive hemichannel-mediated dye uptake in fibroblasts and downstream protein expression.
RESULTS
Tonabersat-sensitive hemichannel-mediated ATP release enhances TGFβ1-driven heterotypic cell-cell interaction and favours myofibroblast activation. The data supports the potential benefit of Cx43 inhibition in reducing tubulointerstitial fibrosis in late-stage diabetic nephropathy.
CONCLUSION
[ "Humans", "Adenosine Triphosphate", "Apyrase", "Communication", "Connexin 43", "Diabetes Mellitus", "Diabetic Nephropathies", "Epithelial Cells", "Fibroblasts", "Fibrosis", "Glucose" ]
9828766
null
null
METHODS
[SUBTITLE] Materials [SUBSECTION] Human kidney (HK2) proximal tubule epithelial cells were purchased from American Type Culture Collection (ATCC) (LGC Standards; Manassas, United States). Medullary renal fibroblasts (TK173) were derived from normal human kidneys and gifted from Dr. Isak Demirel (Örebro University, Sweden). Tissue culture plastic and supplies were from Sarstedt Inc (Leicester, UK), while tissue culture media and fetal calf serum (FCS) were from Fisher Scientific (Loughborough, UK). Penicillin/Streptomycin was from Sigma‐Aldrich (Dorset, UK), while epithelial growth factor (EGF) was purchased from ProSpec (Rotherham, UK). Glass‐bottomed fluorodishes for carboxyfluorescein dye uptake studies were from Thistle Scientific (Glasgow, UK). For western blotting, all membranes, buffers and equipment were obtained from Licor Biosciences (Lincoln, USA). Recombinant TGFβ1 (PHG9214) was from Fisher Scientific (Loughborough, UK), while Tonabersat was purchased from Cambridge Bioscience (Cambridge, UK). ATPlite luminescence assay was from PerkinElmer (Llantrisant, UK). Unless otherwise stated, all chemicals were from Sigma‐Aldrich (Dorset, UK). Human kidney (HK2) proximal tubule epithelial cells were purchased from American Type Culture Collection (ATCC) (LGC Standards; Manassas, United States). Medullary renal fibroblasts (TK173) were derived from normal human kidneys and gifted from Dr. Isak Demirel (Örebro University, Sweden). Tissue culture plastic and supplies were from Sarstedt Inc (Leicester, UK), while tissue culture media and fetal calf serum (FCS) were from Fisher Scientific (Loughborough, UK). Penicillin/Streptomycin was from Sigma‐Aldrich (Dorset, UK), while epithelial growth factor (EGF) was purchased from ProSpec (Rotherham, UK). Glass‐bottomed fluorodishes for carboxyfluorescein dye uptake studies were from Thistle Scientific (Glasgow, UK). For western blotting, all membranes, buffers and equipment were obtained from Licor Biosciences (Lincoln, USA). Recombinant TGFβ1 (PHG9214) was from Fisher Scientific (Loughborough, UK), while Tonabersat was purchased from Cambridge Bioscience (Cambridge, UK). ATPlite luminescence assay was from PerkinElmer (Llantrisant, UK). Unless otherwise stated, all chemicals were from Sigma‐Aldrich (Dorset, UK). [SUBTITLE] Cell culture and treatment [SUBSECTION] HK2 cells (passages 9–21) were maintained in Dulbecco's Modified Eagle's Medium (DMEM)/Ham's F12 medium, while TK173 (passages 4–10) were maintained in DMEM(1x) + pyruvate. HK2 media contained penicillin/streptomycin (2%) and EGF (5 ng/ml). TK173 media were supplemented with minimum essential medium non‐essential amino acids (MEM NEAA). All culture media contained FCS (10%). Cells were cultured at 37°C in a humidified environment with 5% CO2. For HK2 treatments, cells were cultured in low (5 mM) glucose DMEM/F12 (5 mmoL/L) for 48 h, followed by serum starvation overnight prior to treatment with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) at either 5 mM or 25 mM D‐glucose. For TK173 treatments, cells were seeded in culture medium for 24 h, followed by overnight serum starvation before treatment in DMEM(1x), containing 1% FCS with TGFβ1 (10 ng/ml) ± Tonabersat (10 μM) ± apyrase (100 IU/ml) ± P2X7R inhibitor A438079 (50 μM). For co‐culture experiments, cells were treated for 48 h in 5 mM or 25 mM D‐glucose ± Tonabersat (10 μM), before harvesting the media and freeze‐thawing 10x. Conditioned media were then used for treatment of the alternative cell type for 48 h. For this, both cell types were grown in DMEM (1x). HK2 cells (passages 9–21) were maintained in Dulbecco's Modified Eagle's Medium (DMEM)/Ham's F12 medium, while TK173 (passages 4–10) were maintained in DMEM(1x) + pyruvate. HK2 media contained penicillin/streptomycin (2%) and EGF (5 ng/ml). TK173 media were supplemented with minimum essential medium non‐essential amino acids (MEM NEAA). All culture media contained FCS (10%). Cells were cultured at 37°C in a humidified environment with 5% CO2. For HK2 treatments, cells were cultured in low (5 mM) glucose DMEM/F12 (5 mmoL/L) for 48 h, followed by serum starvation overnight prior to treatment with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) at either 5 mM or 25 mM D‐glucose. For TK173 treatments, cells were seeded in culture medium for 24 h, followed by overnight serum starvation before treatment in DMEM(1x), containing 1% FCS with TGFβ1 (10 ng/ml) ± Tonabersat (10 μM) ± apyrase (100 IU/ml) ± P2X7R inhibitor A438079 (50 μM). For co‐culture experiments, cells were treated for 48 h in 5 mM or 25 mM D‐glucose ± Tonabersat (10 μM), before harvesting the media and freeze‐thawing 10x. Conditioned media were then used for treatment of the alternative cell type for 48 h. For this, both cell types were grown in DMEM (1x). [SUBTITLE] Carboxyfluorescein dye uptake [SUBSECTION] Cells were cultured on glass‐bottomed fluorodishes before exposure to Ca2+‐free balanced salt solution (BSS) + carboxyfluorescein (200 μM) for 10 min and Ca2+‐containing BSS + carboxyfluorescein (200 μM) for 5 min. Cells were washed with Ca2+‐containing BSS before imaging with a Cool Snap HQ CCD camera (Roper Scientific, Gottingen, Germany) and Metamorph software (v7.75, Universal Imaging Corp., Marlow, UK). Approximately 10–12 images were taken per dish, and regions of interest (ROI) drawn around individual cells (~15 cell/image). Two separate dishes were imaged per condition for each N number. The integrated density was measured using Fiji software (v2.1.0, ImageJ, LOCI, Wisconsin, USA), as described in detail previously. 18 Percentage dye uptake was calculated using these values, where treatment was set to 100%. Cells were cultured on glass‐bottomed fluorodishes before exposure to Ca2+‐free balanced salt solution (BSS) + carboxyfluorescein (200 μM) for 10 min and Ca2+‐containing BSS + carboxyfluorescein (200 μM) for 5 min. Cells were washed with Ca2+‐containing BSS before imaging with a Cool Snap HQ CCD camera (Roper Scientific, Gottingen, Germany) and Metamorph software (v7.75, Universal Imaging Corp., Marlow, UK). Approximately 10–12 images were taken per dish, and regions of interest (ROI) drawn around individual cells (~15 cell/image). Two separate dishes were imaged per condition for each N number. The integrated density was measured using Fiji software (v2.1.0, ImageJ, LOCI, Wisconsin, USA), as described in detail previously. 18 Percentage dye uptake was calculated using these values, where treatment was set to 100%. [SUBTITLE] ATP luminescence assay [SUBSECTION] The ATP luminescence assay from PerkinElmer is a quantitative assay which measures the amount of ATP in the cell supernatant. Cells were seeded at 1 × 104 following the treatment protocol before being incubated with the substrate solution as provided in the kit. The foil‐covered plate was placed on a shaker for 5 min, before being dark‐adapted for 10 min. Luminescence was measured using a Chameleon plate reader and provided a direct measure of hemichannel‐mediated ATP release. The ATP luminescence assay from PerkinElmer is a quantitative assay which measures the amount of ATP in the cell supernatant. Cells were seeded at 1 × 104 following the treatment protocol before being incubated with the substrate solution as provided in the kit. The foil‐covered plate was placed on a shaker for 5 min, before being dark‐adapted for 10 min. Luminescence was measured using a Chameleon plate reader and provided a direct measure of hemichannel‐mediated ATP release. [SUBTITLE] Immunocytochemistry [SUBSECTION] HK2 and TK173 cells were stimulated with TGFβ1 (10 ng/ml) ± Tonabersat at 100 μM and 10 μM respectively for 48 h prior to fixing with paraformaldehyde (4%), and subsequent blocking with goat serum (10%) for 1 h at room temperature (RT). Antibodies against fibronectin (Santa Cruz sc‐271098), N‐cadherin (Abcam ab18203), Cx43 (Abcam ab11370), and vimentin (Cell Signaling Technology D21H3), (all 1:200) were used for incubation overnight at 4°C. After washing, cells were incubated with nuclear stain, 4′,6‐diamidino‐2‐phenylindole (DAPI) (1 mmoL/L) for 3 min. Cells were incubated with Alexa‐Fluor 488 for 1 h at RT before visualisation using a Leica TC SP8 confocal microscope (Wetzlar, Germany). HK2 and TK173 cells were stimulated with TGFβ1 (10 ng/ml) ± Tonabersat at 100 μM and 10 μM respectively for 48 h prior to fixing with paraformaldehyde (4%), and subsequent blocking with goat serum (10%) for 1 h at room temperature (RT). Antibodies against fibronectin (Santa Cruz sc‐271098), N‐cadherin (Abcam ab18203), Cx43 (Abcam ab11370), and vimentin (Cell Signaling Technology D21H3), (all 1:200) were used for incubation overnight at 4°C. After washing, cells were incubated with nuclear stain, 4′,6‐diamidino‐2‐phenylindole (DAPI) (1 mmoL/L) for 3 min. Cells were incubated with Alexa‐Fluor 488 for 1 h at RT before visualisation using a Leica TC SP8 confocal microscope (Wetzlar, Germany). [SUBTITLE] Western blotting [SUBSECTION] Whole cell lysates were prepared and separated using SDS‐PAGE gel electrophoresis at 125 volts (V) for 1.5 h, before transfer at 100 V for 1 h, as previously described. 19 Membranes were blocked before being probed overnight at 4°C with primary antibodies against Cx43, β‐catenin (Cell Signaling Technology D10A8), N‐cadherin, fibronectin, collagen I (Abcam ab34710), (all at 1:1000) and α‐tubulin (Sigma‐Aldrich T5168), at a dilution of 1:20,000, as a house‐keeping protein. After washing, membranes were probed with secondary antibody (goat anti‐rabbit 800 and/or goat anti‐mouse) at 1:20,000 for 1 h at RT. Bands were visualised using an Odyssey Fc imaging unit before semi‐quantification and analysis using ImageStudioLite software (5.2.5). Whole cell lysates were prepared and separated using SDS‐PAGE gel electrophoresis at 125 volts (V) for 1.5 h, before transfer at 100 V for 1 h, as previously described. 19 Membranes were blocked before being probed overnight at 4°C with primary antibodies against Cx43, β‐catenin (Cell Signaling Technology D10A8), N‐cadherin, fibronectin, collagen I (Abcam ab34710), (all at 1:1000) and α‐tubulin (Sigma‐Aldrich T5168), at a dilution of 1:20,000, as a house‐keeping protein. After washing, membranes were probed with secondary antibody (goat anti‐rabbit 800 and/or goat anti‐mouse) at 1:20,000 for 1 h at RT. Bands were visualised using an Odyssey Fc imaging unit before semi‐quantification and analysis using ImageStudioLite software (5.2.5). [SUBTITLE] Statistical analysis [SUBSECTION] All data are presented as mean value ± SEM. Statistical analysis was performed using ANOVA and Tukey post‐test; p ≤ 0.05 was considered statistically significant, with ‘n’ denoting sample number. All data are presented as mean value ± SEM. Statistical analysis was performed using ANOVA and Tukey post‐test; p ≤ 0.05 was considered statistically significant, with ‘n’ denoting sample number.
RESULTS
[SUBTITLE] Tonabersat blocks hemichannel‐mediated ATP release in tubular epithelial cells [SUBSECTION] Previous studies link increased Cx43 expression to inflammation and fibrosis in a model of renal disease, 6 , 20 while findings from our laboratory report that aberrant Cx43 hemichannel‐mediated ATP release from TGFβ1‐treated tubular epithelial cells evoked phenotypic and functional changes, characteristic of tubular damage. 12 , 21 , 22 Consequently, the current study determined if this effect was accentuated in high glucose and if the response could be decreased using Tonabersat, an efficient Cx43 hemichannel blocker. Human kidney (HK2) cells were cultured at low (5 mM) and high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) at high glucose for 48 h. Glucose (25 mM) increased dye uptake (25.3 ± 8.8%, p < 0.05), compared to low glucose (5 mM) control (Figure 1b,c). The response was amplified by 16.4% in the presence of TGFβ1 (p < 0.001). The combined effect of high glucose and TGFβ1 was reduced by 42.3% ± 5.3% when cells were preincubated with Tonabersat (Figure 1c; p < 0.001). In Figure 1d, TGFβ1 increased ATP release at low (32.3 ± 10.2%; p < 0.05) and high (55.7%, p < 0.001) glucose, with the response at 25 mM glucose significantly decreased by Tonabersat (36.9 ± 12.3%, p < 0.05). Tonabersat negates TGFβ1/glucose‐evoked increases in hemichannel activity in (HK2) proximal tubule epithelial cells. Cells were cultured for 48 h in either low (5 mM) or high (25 mM) glucose ± TGFβ1 (10 ng/mL) ± Tonabersat (Tb; 100 μM). Panel (a) outlines the protocol used to measure Cx43 hemichannel‐mediated carboxyfluorescein (CBF) dye uptake (BSS is balanced salt solution), while panel (b) provides representative images for dye uptake in each condition. Carboxyfluorescein studies (c) assessed changes in hemichannel mediate, and an ATPlite assay used luminescence as a direct measure of ATP release (d). Data represents mean ± SEM, n = 4–6, where *p < 0.05, **p < 0.01, ***p < 0.001. Previous studies link increased Cx43 expression to inflammation and fibrosis in a model of renal disease, 6 , 20 while findings from our laboratory report that aberrant Cx43 hemichannel‐mediated ATP release from TGFβ1‐treated tubular epithelial cells evoked phenotypic and functional changes, characteristic of tubular damage. 12 , 21 , 22 Consequently, the current study determined if this effect was accentuated in high glucose and if the response could be decreased using Tonabersat, an efficient Cx43 hemichannel blocker. Human kidney (HK2) cells were cultured at low (5 mM) and high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) at high glucose for 48 h. Glucose (25 mM) increased dye uptake (25.3 ± 8.8%, p < 0.05), compared to low glucose (5 mM) control (Figure 1b,c). The response was amplified by 16.4% in the presence of TGFβ1 (p < 0.001). The combined effect of high glucose and TGFβ1 was reduced by 42.3% ± 5.3% when cells were preincubated with Tonabersat (Figure 1c; p < 0.001). In Figure 1d, TGFβ1 increased ATP release at low (32.3 ± 10.2%; p < 0.05) and high (55.7%, p < 0.001) glucose, with the response at 25 mM glucose significantly decreased by Tonabersat (36.9 ± 12.3%, p < 0.05). Tonabersat negates TGFβ1/glucose‐evoked increases in hemichannel activity in (HK2) proximal tubule epithelial cells. Cells were cultured for 48 h in either low (5 mM) or high (25 mM) glucose ± TGFβ1 (10 ng/mL) ± Tonabersat (Tb; 100 μM). Panel (a) outlines the protocol used to measure Cx43 hemichannel‐mediated carboxyfluorescein (CBF) dye uptake (BSS is balanced salt solution), while panel (b) provides representative images for dye uptake in each condition. Carboxyfluorescein studies (c) assessed changes in hemichannel mediate, and an ATPlite assay used luminescence as a direct measure of ATP release (d). Data represents mean ± SEM, n = 4–6, where *p < 0.05, **p < 0.01, ***p < 0.001. [SUBTITLE] Blocking Cx43 hemichannels inhibits TGFβ1/glucose‐induced increases in expression of Cx43 and markers of tubular damage [SUBSECTION] We previously reported that UUO causes disassembly of the adherens junction complex 12 and that this is accompanied by increased interstitial fibrosis. 6 In the absence of glycaemic injury, this effect was partly restored in the Cx43+/− mouse model of UUO. Consequently, we evaluated the effect of TGFβ1 on Cx43, adherens junction (N‐cadherin and β‐catenin) and ECM (fibronectin and collagen I) protein expression in high glucose (Figure 2). Human kidney (HK2) cells were treated for 48 h in either low (5 mM) or high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) to block ATP release (Figure 2a). Compared to low glucose control, the cytokine increased expression of total Cx43 by 76.4 ± 10.2% (Figure 2b; p < 0.001), N‐cadherin by 39.8 ± 7.8% (Figure 2d; p < 0.01), fibronectin by 52.1 ± 6.7% (Figure 2 e; p < 0.001) and collagen I by 28.8 ± 8.6% (Figure 2f; p < 0.05). This effect was amplified in the presence of high (25 mM) glucose, where TGFβ1 increased N‐cadherin (51.3%, p < 0.001), fibronectin (67.7%, p < 0.001) and collagen I (88.5%, p < 0.001) expression. The effect of TGFβ1 on Cx43 was not accentuated under high (25 mM) glucose conditions (64.4%, p < 0.001), and there were no effects of glucose or TGFβ1 on β‐catenin expression (Figure 2c). Tonabersat inhibits TGFβ1/glucose‐evoked increases in Cx43, adherens junction and ECM protein expression. HK2 cells were cultured for 48 h in either low (5 mM) or high (25 mM) glucose ± TGFβ1 (10 ng/mL) ± Tonabersat (Tb; 100 μM). Tonabersat blocks Cx43 hemichannel‐mediated ATP release (a). Western blot examined whole cell protein expression (%) for Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f). Data shown illustrate a representative blot and mean data ± SEM (n = 4–6), where *p < 0.05, **p < 0.01, ***p < 0.001. Having determined that Tonabersat blocks TGFβ1‐induced changes in hemichannel activity (Figure 1), we assessed if TGFβ1‐evoked changes in protein expression under conditions of high glucose were hemichannel mediated. As shown in Figure 2, Tonabersat decreased high glucose and TGFβ1‐induced increases in Cx43 expression by 46 ± 10.3% (p < 0.01), while fibronectin and collagen I expression were decreased by 35.7 ± 4.8%, (p < 0.01) and 40.6 ± 5.4%, (p < 0.001), respectively. There was no effect on N‐cadherin. We previously reported that UUO causes disassembly of the adherens junction complex 12 and that this is accompanied by increased interstitial fibrosis. 6 In the absence of glycaemic injury, this effect was partly restored in the Cx43+/− mouse model of UUO. Consequently, we evaluated the effect of TGFβ1 on Cx43, adherens junction (N‐cadherin and β‐catenin) and ECM (fibronectin and collagen I) protein expression in high glucose (Figure 2). Human kidney (HK2) cells were treated for 48 h in either low (5 mM) or high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) to block ATP release (Figure 2a). Compared to low glucose control, the cytokine increased expression of total Cx43 by 76.4 ± 10.2% (Figure 2b; p < 0.001), N‐cadherin by 39.8 ± 7.8% (Figure 2d; p < 0.01), fibronectin by 52.1 ± 6.7% (Figure 2 e; p < 0.001) and collagen I by 28.8 ± 8.6% (Figure 2f; p < 0.05). This effect was amplified in the presence of high (25 mM) glucose, where TGFβ1 increased N‐cadherin (51.3%, p < 0.001), fibronectin (67.7%, p < 0.001) and collagen I (88.5%, p < 0.001) expression. The effect of TGFβ1 on Cx43 was not accentuated under high (25 mM) glucose conditions (64.4%, p < 0.001), and there were no effects of glucose or TGFβ1 on β‐catenin expression (Figure 2c). Tonabersat inhibits TGFβ1/glucose‐evoked increases in Cx43, adherens junction and ECM protein expression. HK2 cells were cultured for 48 h in either low (5 mM) or high (25 mM) glucose ± TGFβ1 (10 ng/mL) ± Tonabersat (Tb; 100 μM). Tonabersat blocks Cx43 hemichannel‐mediated ATP release (a). Western blot examined whole cell protein expression (%) for Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f). Data shown illustrate a representative blot and mean data ± SEM (n = 4–6), where *p < 0.05, **p < 0.01, ***p < 0.001. Having determined that Tonabersat blocks TGFβ1‐induced changes in hemichannel activity (Figure 1), we assessed if TGFβ1‐evoked changes in protein expression under conditions of high glucose were hemichannel mediated. As shown in Figure 2, Tonabersat decreased high glucose and TGFβ1‐induced increases in Cx43 expression by 46 ± 10.3% (p < 0.01), while fibronectin and collagen I expression were decreased by 35.7 ± 4.8%, (p < 0.01) and 40.6 ± 5.4%, (p < 0.001), respectively. There was no effect on N‐cadherin. [SUBTITLE] Tonabersat blocks hemichannel‐mediated ATP release in renal fibroblasts [SUBSECTION] Tubulointerstitial fibrosis (TIF) is the key underlying pathology of diabetic kidney disease and develops in response to activation of multiple cell types in and around the proximal tubules. 23 Medullary fibroblasts contribute to the onset and progression of TIF in diabetic nephropathy. With previous studies linking the heterozygous Cx43+/− UUO mouse to decreased fibroblast activation in the kidney cortex, 6 we wanted to assess (i) if renal fibroblasts (TK173) have functional hemichannels and (ii) if Tonabersat could block effects of glucose and TGFβ1 on Cx43 hemichannel‐mediated release of ATP. Renal fibroblasts were treated in low (5 mM) and high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (10 μM) at high glucose for 48 h. Carboxyfluorescein measured hemichannel‐mediated dye uptake and ATP luminescence assays quantified ATP release. As shown in Figure 3a,b, high glucose (25 mM) augmented TGFβ1‐evoked dye uptake by 28.1% (compared high glucose alone; p < 0.05). The combined effect of glucose and cytokine was completely negated when cells were co‐incubated with Tonabersat (49.8 ± 4.6% reduction; p < 0.001). The ability of Tonabersat to reduce dye uptake in response to TGFβ1 and high glucose was paralleled by a 43 ± 5.9% decrease in ATP release (Figure 3c; p < 0.05). Tonabersat negates TGFβ1/glucose‐evoked hemichannel mediated ATP release in (TK173) renal fibroblasts. Cells were treated with TGFβ1 (10 ng/ml) in low or high glucose ± Tonabersat (Tb; 10 μM) for 48 h. Panel (a) shows representative images for carboxyfluorescein dye uptake in each condition. Mean data (± SEM, n = 4–6) demonstrated that TGFβ1 increased dye uptake, an effect that was amplified when co‐incubated at high glucose (panel (b)). Examining ATP release, this high glucose TGFβ1‐induced response was reversed to near basal (5 mM glucose) levels when cells were pre‐incubated with Tonabersat (panel (c)). Significances are represented as *p < 0.05, **p < 0.01, ***p < 0.001. Tubulointerstitial fibrosis (TIF) is the key underlying pathology of diabetic kidney disease and develops in response to activation of multiple cell types in and around the proximal tubules. 23 Medullary fibroblasts contribute to the onset and progression of TIF in diabetic nephropathy. With previous studies linking the heterozygous Cx43+/− UUO mouse to decreased fibroblast activation in the kidney cortex, 6 we wanted to assess (i) if renal fibroblasts (TK173) have functional hemichannels and (ii) if Tonabersat could block effects of glucose and TGFβ1 on Cx43 hemichannel‐mediated release of ATP. Renal fibroblasts were treated in low (5 mM) and high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (10 μM) at high glucose for 48 h. Carboxyfluorescein measured hemichannel‐mediated dye uptake and ATP luminescence assays quantified ATP release. As shown in Figure 3a,b, high glucose (25 mM) augmented TGFβ1‐evoked dye uptake by 28.1% (compared high glucose alone; p < 0.05). The combined effect of glucose and cytokine was completely negated when cells were co‐incubated with Tonabersat (49.8 ± 4.6% reduction; p < 0.001). The ability of Tonabersat to reduce dye uptake in response to TGFβ1 and high glucose was paralleled by a 43 ± 5.9% decrease in ATP release (Figure 3c; p < 0.05). Tonabersat negates TGFβ1/glucose‐evoked hemichannel mediated ATP release in (TK173) renal fibroblasts. Cells were treated with TGFβ1 (10 ng/ml) in low or high glucose ± Tonabersat (Tb; 10 μM) for 48 h. Panel (a) shows representative images for carboxyfluorescein dye uptake in each condition. Mean data (± SEM, n = 4–6) demonstrated that TGFβ1 increased dye uptake, an effect that was amplified when co‐incubated at high glucose (panel (b)). Examining ATP release, this high glucose TGFβ1‐induced response was reversed to near basal (5 mM glucose) levels when cells were pre‐incubated with Tonabersat (panel (c)). Significances are represented as *p < 0.05, **p < 0.01, ***p < 0.001. [SUBTITLE] Tonabersat inhibits TGFβ1/glucose‐induced changes in Cx43, N‐cadherin and ECM localisation and expression in renal fibroblasts [SUBSECTION] Having ascertained that TGFβ1 stimulates Cx43 hemichannel activity in renal fibroblasts, an effect augmented in the presence of high glucose, we investigated if blocking this activity could confer any phenotypic protection. Immunocytochemistry (Figure 4a) and western blotting (Figure 4b–f) were used to determine TGFβ1‐induced changes in protein localisation and expression at low (5 mM) and high (25 mM) glucose, before assessing the ability of Tonabersat to dampen the combined effect of glycaemic and cytokine challenge in TK173 fibroblasts. Tonabersat reverses TGFβ1/glucose‐evoked changes in ECM and adherens protein expression and localisation in (TK173) renal fibroblasts. In panel (a) immunocytochemistry revealed the effect of the pro‐fibrotic cytokine TGFβ1 (10 ng/ml) on the localisation of Cx43, N‐cadherin, vimentin and fibronectin in low (5 mM) and high (25 mM) glucose ± Tonabersat (Tb; 10 μM) in TK173 fibroblasts. Western blot examined whole cell protein expression (%) of Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f). Representative blots and mean data ± SEM (n = 4–6) are shown, where *p < 0.05, **p < 0.01, ***p < 0.001. The pro‐fibrotic cytokine TGFβ1 had negligible effects on localisation of candidate proteins, but increased expression in both low (5mM) and high (25mM) glucose as determined by immunocytochemistry. This effect was particularly evident with ECM proteins fibronectin and vimentin (Figure 4a). As an intermediate filament protein, vimentin was used in place of collagen I which was poorly resolved using immunocytochemistry. Tonabersat reversed the combined effects of high glucose and cytokine. Semi‐quantification of protein changes using western blot determined that in the presence of high glucose, TGFβ1 increased expression of Cx43 (Figure 4b 66.1%, p < 0.001), β‐catenin (Figure 4c 32.7%, p < 0.05), N‐cadherin (Figure 4d 45%, p < 0.05), fibronectin (Figure 4e 47.6%, p < 0.01) and collagen I (Figure 4f 26.4%, p=NS). Tonabersat inhibited these effects for Cx43 (54.3 ± 12.6%, p < 0.01), N‐cadherin (40.9 ± 11.8%, p < 0.05), fibronectin (34.3 ± 4.2%, p < 0.05) and collagen I (30.8 ± 3%, p=NS). Having ascertained that TGFβ1 stimulates Cx43 hemichannel activity in renal fibroblasts, an effect augmented in the presence of high glucose, we investigated if blocking this activity could confer any phenotypic protection. Immunocytochemistry (Figure 4a) and western blotting (Figure 4b–f) were used to determine TGFβ1‐induced changes in protein localisation and expression at low (5 mM) and high (25 mM) glucose, before assessing the ability of Tonabersat to dampen the combined effect of glycaemic and cytokine challenge in TK173 fibroblasts. Tonabersat reverses TGFβ1/glucose‐evoked changes in ECM and adherens protein expression and localisation in (TK173) renal fibroblasts. In panel (a) immunocytochemistry revealed the effect of the pro‐fibrotic cytokine TGFβ1 (10 ng/ml) on the localisation of Cx43, N‐cadherin, vimentin and fibronectin in low (5 mM) and high (25 mM) glucose ± Tonabersat (Tb; 10 μM) in TK173 fibroblasts. Western blot examined whole cell protein expression (%) of Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f). Representative blots and mean data ± SEM (n = 4–6) are shown, where *p < 0.05, **p < 0.01, ***p < 0.001. The pro‐fibrotic cytokine TGFβ1 had negligible effects on localisation of candidate proteins, but increased expression in both low (5mM) and high (25mM) glucose as determined by immunocytochemistry. This effect was particularly evident with ECM proteins fibronectin and vimentin (Figure 4a). As an intermediate filament protein, vimentin was used in place of collagen I which was poorly resolved using immunocytochemistry. Tonabersat reversed the combined effects of high glucose and cytokine. Semi‐quantification of protein changes using western blot determined that in the presence of high glucose, TGFβ1 increased expression of Cx43 (Figure 4b 66.1%, p < 0.001), β‐catenin (Figure 4c 32.7%, p < 0.05), N‐cadherin (Figure 4d 45%, p < 0.05), fibronectin (Figure 4e 47.6%, p < 0.01) and collagen I (Figure 4f 26.4%, p=NS). Tonabersat inhibited these effects for Cx43 (54.3 ± 12.6%, p < 0.01), N‐cadherin (40.9 ± 11.8%, p < 0.05), fibronectin (34.3 ± 4.2%, p < 0.05) and collagen I (30.8 ± 3%, p=NS). [SUBTITLE] ATP mediates TGFβ1/glucose‐induced changes in Cx43, N‐cadherin and ECM expression in renal fibroblasts [SUBSECTION] Having determined that high glucose in combination with TGFβ1 evokes a Tonabersat‐sensitive increase in ATP release (Figure 3) and increased expression of tubular injury associated proteins (Figure 4), we further explored a downstream role for ATP‐mediated purinergic signalling in driving high glucose and TGFβ1‐induced changes. An ATP‐diphosphohydrolase, apyrase (apy) catalyses the sequential hydrolysis of ATP to ADP and ADP to AMP, releasing inorganic phosphate and reducing the extracellular concentration of ATP ([ATP]e) to decrease activation of P2‐purinergic receptors (Figure 5a). TK173 cells were treated in low or high glucose with TGFβ1 (10 ng/ml) ± apyrase (100 IU/ml) for 48 h. Western blot assessed changes in whole cell protein expression. Compared to high glucose alone, TGFβ1 plus 25 mM glucose increased expression of Cx43 (Figure 5b 66.1%, p < 0.01), β‐catenin (Figure 5c 32.7%, p < 0.05), N‐cadherin (Figure 5d 45%, p < 0.01), fibronectin (Figure 5e 35.2%, p < 0.05) and collagen I (Figure 5f 26.4%, p=NS). These changes decreased by 51.4 ± 14.2% (Cx43; p < 0.05), 34.0 ± 7.0%, (N‐cadherin; p < 0.05), 26.6 ± 1.3%, (fibronectin; p < 0.05), and 47.8 ± 12.3%, (collagen I; p < 0.05), when cells were co‐incubated with apyrase. Hydrolysis of ATP failed to significantly alter TGFβ1/glucose‐induced changes in β‐catenin. Reducing activation of the P2X7 receptor reverses TGFβ1/glucose‐induced changes in ECM and adherens junction protein expression in (TK173) renal fibroblasts. To reduce activation of the P2X7R, either the concentration of TGFβ1/glucose‐evoked extracellular ATP ([ATP]e) was reduced by apyrase (apy), which catalyses the sequential hydrolysis of ATP to ADP and AMP, or the P2X7 receptor was inhibited using A438079. Both strategies reduced markers of TGFβ1‐dependent inflammation and fibrosis (a). Western blot analysis determined whole cell protein expression (%) of Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f) in response to TGFβ1 (10 ng/ml) in low (5 mM) and high (25 mM) glucose ± apyrase (100 IU/ml) ± the P2X7R inhibitor (P2X7Ri: A438079 (50 μM)). Representative blots are shown above mean data ± SEM, n = 3–5, where *p < 0.05, **p < 0.01, ***p < 0.001. Known to mediate inflammation and downstream fibrosis, activation of the P2X7 receptor (P2X7R) has been associated with tubular injury in an in vivo model of diabetic nephropathy. 10 We used the P2X7R inhibitor A438079 (50 μM) to determine the role of this receptor subtype in glucose/cytokine‐induced changes in TK173 fibroblasts (Figure 5a). The P2X7R inhibitor reversed TGFβ1/high glucose‐induced increases in Cx43 (Figure 5b 48.7 ± 12.7%, p < 0.05), N‐cadherin (Figure 5d 53.1 ± 13.9%, p < 0.01) and collagen I (Figure 5f 62.6 ± 2.7%, P < 0.001), and marginally reduced expression of β‐catenin (Figure 5c 21.8 ± 14.2%) and fibronectin (Figure 5 e 26.3 ± 1.9%). Having determined that high glucose in combination with TGFβ1 evokes a Tonabersat‐sensitive increase in ATP release (Figure 3) and increased expression of tubular injury associated proteins (Figure 4), we further explored a downstream role for ATP‐mediated purinergic signalling in driving high glucose and TGFβ1‐induced changes. An ATP‐diphosphohydrolase, apyrase (apy) catalyses the sequential hydrolysis of ATP to ADP and ADP to AMP, releasing inorganic phosphate and reducing the extracellular concentration of ATP ([ATP]e) to decrease activation of P2‐purinergic receptors (Figure 5a). TK173 cells were treated in low or high glucose with TGFβ1 (10 ng/ml) ± apyrase (100 IU/ml) for 48 h. Western blot assessed changes in whole cell protein expression. Compared to high glucose alone, TGFβ1 plus 25 mM glucose increased expression of Cx43 (Figure 5b 66.1%, p < 0.01), β‐catenin (Figure 5c 32.7%, p < 0.05), N‐cadherin (Figure 5d 45%, p < 0.01), fibronectin (Figure 5e 35.2%, p < 0.05) and collagen I (Figure 5f 26.4%, p=NS). These changes decreased by 51.4 ± 14.2% (Cx43; p < 0.05), 34.0 ± 7.0%, (N‐cadherin; p < 0.05), 26.6 ± 1.3%, (fibronectin; p < 0.05), and 47.8 ± 12.3%, (collagen I; p < 0.05), when cells were co‐incubated with apyrase. Hydrolysis of ATP failed to significantly alter TGFβ1/glucose‐induced changes in β‐catenin. Reducing activation of the P2X7 receptor reverses TGFβ1/glucose‐induced changes in ECM and adherens junction protein expression in (TK173) renal fibroblasts. To reduce activation of the P2X7R, either the concentration of TGFβ1/glucose‐evoked extracellular ATP ([ATP]e) was reduced by apyrase (apy), which catalyses the sequential hydrolysis of ATP to ADP and AMP, or the P2X7 receptor was inhibited using A438079. Both strategies reduced markers of TGFβ1‐dependent inflammation and fibrosis (a). Western blot analysis determined whole cell protein expression (%) of Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f) in response to TGFβ1 (10 ng/ml) in low (5 mM) and high (25 mM) glucose ± apyrase (100 IU/ml) ± the P2X7R inhibitor (P2X7Ri: A438079 (50 μM)). Representative blots are shown above mean data ± SEM, n = 3–5, where *p < 0.05, **p < 0.01, ***p < 0.001. Known to mediate inflammation and downstream fibrosis, activation of the P2X7 receptor (P2X7R) has been associated with tubular injury in an in vivo model of diabetic nephropathy. 10 We used the P2X7R inhibitor A438079 (50 μM) to determine the role of this receptor subtype in glucose/cytokine‐induced changes in TK173 fibroblasts (Figure 5a). The P2X7R inhibitor reversed TGFβ1/high glucose‐induced increases in Cx43 (Figure 5b 48.7 ± 12.7%, p < 0.05), N‐cadherin (Figure 5d 53.1 ± 13.9%, p < 0.01) and collagen I (Figure 5f 62.6 ± 2.7%, P < 0.001), and marginally reduced expression of β‐catenin (Figure 5c 21.8 ± 14.2%) and fibronectin (Figure 5 e 26.3 ± 1.9%). [SUBTITLE] Tonabersat partially negates epithelial cell secretome‐mediated changes on fibroblast hemichannel number and protein expression [SUBSECTION] While tubular epithelial cells are often regarded as the initial drivers of renal injury, fibroblasts are considered the major ECM producing cell type. Importantly, they are recruited and activated in response to sustained tubular injury. Regulated by soluble ligands, for example, ATP, crosstalk between the tubular epithelia and fibroblasts can orchestrate many forms of disease progression 24 including diabetic nephropathy. 25 To investigate the role of the cell secretome in paracrine‐mediated signalling between each cell type, conditioned media transfer (indirect co‐culture) was performed between tubular epithelial cells and medullary fibroblasts. In doing so, we determined the downstream effects on hemichannel activity and protein expression on both HK2 cells and TK173 cells when incubated with conditioned media of the alternate cell type (Figure 6a). Conditioned media from high glucose‐treated proximal tubule epithelial cells (HK2) evoked Tonabersat‐sensitive changes in ECM and adherens junction protein expression in (TK173) renal fibroblasts. Indirect co‐culture utilised 10 times freeze‐thawed 48 h high glucose (25 mM)‐treated conditioned media (CM) ± Tonabersat (Tb; 10 μM) to determine the effect of cell secretome on the heterotypic cell type (a). Carboxyfluorescein studies assessed changes in dye uptake when fibroblast conditioned media were added to tubular epithelial cells (b) and (c) and vice versa (d) and (e). Determined by western blotting, tubular epithelial cell secretome examined Tonabersat‐sensitive changes in fibroblast whole cell protein expression (%) for Cx43 (f), β‐catenin (g), N‐cadherin (h), fibronectin (i) and collagen I (j). Data shown illustrate representative blots and mean data ± SEM (n = 3–4), where *p < 0.05, **p < 0.01, ***p < 0.001. Although the transfer of high glucose conditioned media from fibroblasts evoked a Tonabersat‐sensitive increase in dye uptake in tubular epithelial cells (55.9%, p < 0.001) as compared to 5 mM glucose control (Figure 6b,c), it had minimal effect on HK2 protein expression (see: Figure S1). Conditioned media from high glucose‐treated proximal tubule epithelial cells (HK2) evoked a Tonabersat‐sensitive increase in dye uptake in (TK173) renal fibroblasts of 54.5% (p < 0.001), compared to low glucose control (Figure 6d,e). Contrary to the lack of effect that fibroblast (TK173) conditioned media had on tubule epithelial (HK2) phenotype, the effects of HK2 conditioned media on TK173 dye uptake, were paralleled by changes in Cx43 expression and markers of the ECM and adherens junction. Western blot determined that high glucose conditioned media from epithelial cells increased expression of Cx43 (47.7%, Figure 6f; p < 0.001), N‐cadherin (24.1%, Figure 6h; p < 0.05) and fibronectin (60.5%, Figure 6i; p < 0.01) in fibroblasts compared to unconditioned 25 mM glucose media. Tonabersat negated this effect by 33.9 ± 11.4%, p < 0.01 (Cx43), 25.3 ± 8.9%, p < 0.05 (N‐cadherin) and 53.4 ± 7.7%, p < 0.05 (fibronectin). There were minimal effects on β‐catenin (Figure 6g) or collagen I (Figure 6j). While tubular epithelial cells are often regarded as the initial drivers of renal injury, fibroblasts are considered the major ECM producing cell type. Importantly, they are recruited and activated in response to sustained tubular injury. Regulated by soluble ligands, for example, ATP, crosstalk between the tubular epithelia and fibroblasts can orchestrate many forms of disease progression 24 including diabetic nephropathy. 25 To investigate the role of the cell secretome in paracrine‐mediated signalling between each cell type, conditioned media transfer (indirect co‐culture) was performed between tubular epithelial cells and medullary fibroblasts. In doing so, we determined the downstream effects on hemichannel activity and protein expression on both HK2 cells and TK173 cells when incubated with conditioned media of the alternate cell type (Figure 6a). Conditioned media from high glucose‐treated proximal tubule epithelial cells (HK2) evoked Tonabersat‐sensitive changes in ECM and adherens junction protein expression in (TK173) renal fibroblasts. Indirect co‐culture utilised 10 times freeze‐thawed 48 h high glucose (25 mM)‐treated conditioned media (CM) ± Tonabersat (Tb; 10 μM) to determine the effect of cell secretome on the heterotypic cell type (a). Carboxyfluorescein studies assessed changes in dye uptake when fibroblast conditioned media were added to tubular epithelial cells (b) and (c) and vice versa (d) and (e). Determined by western blotting, tubular epithelial cell secretome examined Tonabersat‐sensitive changes in fibroblast whole cell protein expression (%) for Cx43 (f), β‐catenin (g), N‐cadherin (h), fibronectin (i) and collagen I (j). Data shown illustrate representative blots and mean data ± SEM (n = 3–4), where *p < 0.05, **p < 0.01, ***p < 0.001. Although the transfer of high glucose conditioned media from fibroblasts evoked a Tonabersat‐sensitive increase in dye uptake in tubular epithelial cells (55.9%, p < 0.001) as compared to 5 mM glucose control (Figure 6b,c), it had minimal effect on HK2 protein expression (see: Figure S1). Conditioned media from high glucose‐treated proximal tubule epithelial cells (HK2) evoked a Tonabersat‐sensitive increase in dye uptake in (TK173) renal fibroblasts of 54.5% (p < 0.001), compared to low glucose control (Figure 6d,e). Contrary to the lack of effect that fibroblast (TK173) conditioned media had on tubule epithelial (HK2) phenotype, the effects of HK2 conditioned media on TK173 dye uptake, were paralleled by changes in Cx43 expression and markers of the ECM and adherens junction. Western blot determined that high glucose conditioned media from epithelial cells increased expression of Cx43 (47.7%, Figure 6f; p < 0.001), N‐cadherin (24.1%, Figure 6h; p < 0.05) and fibronectin (60.5%, Figure 6i; p < 0.01) in fibroblasts compared to unconditioned 25 mM glucose media. Tonabersat negated this effect by 33.9 ± 11.4%, p < 0.01 (Cx43), 25.3 ± 8.9%, p < 0.05 (N‐cadherin) and 53.4 ± 7.7%, p < 0.05 (fibronectin). There were minimal effects on β‐catenin (Figure 6g) or collagen I (Figure 6j).
null
null
[ "What is already known?", "What this study found?", "Implications?", "INTRODUCTION", "Materials", "Cell culture and treatment", "Carboxyfluorescein dye uptake", "\nATP luminescence assay", "Immunocytochemistry", "Western blotting", "Statistical analysis", "Tonabersat blocks hemichannel‐mediated ATP release in tubular epithelial cells", "Blocking Cx43 hemichannels inhibits TGFβ1/glucose‐induced increases in expression of Cx43 and markers of tubular damage", "Tonabersat blocks hemichannel‐mediated ATP release in renal fibroblasts", "Tonabersat inhibits TGFβ1/glucose‐induced changes in Cx43, N‐cadherin and ECM localisation and expression in renal fibroblasts", "\nATP mediates TGFβ1/glucose‐induced changes in Cx43, N‐cadherin and ECM expression in renal fibroblasts", "Tonabersat partially negates epithelial cell secretome‐mediated changes on fibroblast hemichannel number and protein expression", "AUTHOR CONTRIBUTIONS", "FUNDING INFORMATION" ]
[ "\nCx43 hemichannels release ATP in TGFβ1‐treated tubular epithelial cells.\n\nCx43 hemichannels release ATP in TGFβ1‐treated tubular epithelial cells.", "\nTGFβ1 increases Cx43 hemichannel‐mediated ATP release in TK173 renal fibroblasts and HK2 tubular epithelial cells, an effect amplified by high glucose.Aberrant ATP release initiates phenotypic changes via P2X7 receptors.Co‐culture of TK173 cells with high glucose conditioned HK2 media, increased Cx43 hemichannel‐mediated dye uptake and ECM protein expression.Characteristic of late‐stage kidney damage, effects were partly blocked with Tonabersat.\n\nTGFβ1 increases Cx43 hemichannel‐mediated ATP release in TK173 renal fibroblasts and HK2 tubular epithelial cells, an effect amplified by high glucose.\nAberrant ATP release initiates phenotypic changes via P2X7 receptors.\nCo‐culture of TK173 cells with high glucose conditioned HK2 media, increased Cx43 hemichannel‐mediated dye uptake and ECM protein expression.\nCharacteristic of late‐stage kidney damage, effects were partly blocked with Tonabersat.", "\nTonabersat blocks Cx43 hemichannel‐mediated communication within and between tubular epithelial cells (HK2) and medullary renal fibroblasts (TK173), protecting against phenotypic changes characteristic of diabetic nephropathy.\n\nTonabersat blocks Cx43 hemichannel‐mediated communication within and between tubular epithelial cells (HK2) and medullary renal fibroblasts (TK173), protecting against phenotypic changes characteristic of diabetic nephropathy.", "Worldwide, diabetic kidney disease develops in around 40% of those with diabetes and is the leading cause of chronic kidney disease (CKD).\n1\n In 2017, 1.2 million people died from CKD while 697.5 million cases were recorded globally.\n2\n In the United Kingdom, 30% of those entering maintenance renal replacement therapy have diabetic nephropathy as their primary renal disease.\n3\n Currently, there are no curative therapeutic options to prevent transition to end‐stage kidney disease and innovative approaches are urgently required.\nAs the main contributor to end‐stage renal failure, understanding mechanisms by which glucose alters renal function is critical to identifying future strategies for prevention and/or arrest of diabetic nephropathy (DN). Early damage in DN is characterised by persistent inflammation and fibrosis of the tubulointerstitium, where fibrosis develops in response to various morphological and phenotypic changes, including partial epithelial‐to‐mesenchymal transition (pEMT), and extracellular matrix (ECM) remodelling.\n4\n In the face of sustained glycaemic injury, soluble chemokines, adhesion molecules and cytokines, for example, transforming growth factor beta‐1 (TGFβ1), recruit and activate infiltrating immune cells and resident fibroblasts to mediate inflammation and fibrosis, severity of which dictates disease progression.\n5\n\n\nWork by Abed et al., reported that heterozygous connexin (Cx)43+/− mice exhibited decreased tubular macrophage infiltration and reduced numbers of active fibroblasts when surgically induced to exhibit interstitial inflammation and fibrosis via unilateral ureteral obstruction (UUO).\n6\n Moreover, our laboratory demonstrated that these mice exhibit reduced disassembly of adherens junction and tight junction proteins, the former of which is considered an initiating trigger for pEMT in the injured tubules.\n7\n\n\nConnexins oligomerise to form hexameric structures, before being transported to the cell membrane where they form hemichannels which allow for the local release of various danger‐associated molecular patterns (DAMPs), for example, adenosine triphosphate (ATP), in response to pathological triggers, including hyperglycaemia\n8\n and inflammation.\n9\n Excess ATP within the intercellular environment activates P2X7 purinergic receptors\n10\n to trigger inflammatory and fibrotic events in both resident tubule epithelial cells\n11\n and other associated cell types local to the proximal kidney, for example, macrophages.\n10\n We previously demonstrated that pharmacologically inhibiting Cx43‐mediated hemichannel activity using Cx43 hemichannel blocker Peptide 5, protects against early tubular injury in an in vitro model of experimental chronic kidney disease under euglycaemic conditions.\n12\n Similar to Peptide 5, Tonabersat; a benzopyran derivative originally reported to inhibit gap junction communication,\n13\n has recently been shown to be equivalent to Peptide 5 in its capacity to act as a blocker of Cx43 hemichannels.\n14\n, \n15\n, \n16\n Consequently, Tonabersat (Xiflam) has since been reported to confer protection in both in vivo models of age‐related macular degeneration\n14\n and diabetic retinopathy.\n14\n, \n17\n Currently entering Phase2b clinical trials for diabetic macular oedema and retinopathy, its ability to target hemichannels in the diabetic kidney and confer anti‐inflammatory protection are yet to be reported.\nIn this study, we tested whether blocking Cx43‐mediated hemichannel ATP release decreased P2X7R activation within and between tubule epithelial cells and renal fibroblasts in conditions designed to mimic diabetic nephropathy. We posit that a heterotypic pro‐inflammatory relationship exists between tubular epithelial cells and resident fibroblasts, and that blocking Cx43‐mediated ATP release can dampen this interaction. The data demonstrate the importance of Cx43 hemichannel‐mediated paracrine signalling in kidney damage, and suggest the potential benefit of using Cx43 hemichannel blockers in managing tubulointerstitial fibrosis in late‐stage DN.", "Human kidney (HK2) proximal tubule epithelial cells were purchased from American Type Culture Collection (ATCC) (LGC Standards; Manassas, United States). Medullary renal fibroblasts (TK173) were derived from normal human kidneys and gifted from Dr. Isak Demirel (Örebro University, Sweden). Tissue culture plastic and supplies were from Sarstedt Inc (Leicester, UK), while tissue culture media and fetal calf serum (FCS) were from Fisher Scientific (Loughborough, UK). Penicillin/Streptomycin was from Sigma‐Aldrich (Dorset, UK), while epithelial growth factor (EGF) was purchased from ProSpec (Rotherham, UK). Glass‐bottomed fluorodishes for carboxyfluorescein dye uptake studies were from Thistle Scientific (Glasgow, UK). For western blotting, all membranes, buffers and equipment were obtained from Licor Biosciences (Lincoln, USA). Recombinant TGFβ1 (PHG9214) was from Fisher Scientific (Loughborough, UK), while Tonabersat was purchased from Cambridge Bioscience (Cambridge, UK). ATPlite luminescence assay was from PerkinElmer (Llantrisant, UK). Unless otherwise stated, all chemicals were from Sigma‐Aldrich (Dorset, UK).", "HK2 cells (passages 9–21) were maintained in Dulbecco's Modified Eagle's Medium (DMEM)/Ham's F12 medium, while TK173 (passages 4–10) were maintained in DMEM(1x) + pyruvate. HK2 media contained penicillin/streptomycin (2%) and EGF (5 ng/ml). TK173 media were supplemented with minimum essential medium non‐essential amino acids (MEM NEAA). All culture media contained FCS (10%). Cells were cultured at 37°C in a humidified environment with 5% CO2.\nFor HK2 treatments, cells were cultured in low (5 mM) glucose DMEM/F12 (5 mmoL/L) for 48 h, followed by serum starvation overnight prior to treatment with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) at either 5 mM or 25 mM D‐glucose. For TK173 treatments, cells were seeded in culture medium for 24 h, followed by overnight serum starvation before treatment in DMEM(1x), containing 1% FCS with TGFβ1 (10 ng/ml) ± Tonabersat (10 μM) ± apyrase (100 IU/ml) ± P2X7R inhibitor A438079 (50 μM). For co‐culture experiments, cells were treated for 48 h in 5 mM or 25 mM D‐glucose ± Tonabersat (10 μM), before harvesting the media and freeze‐thawing 10x. Conditioned media were then used for treatment of the alternative cell type for 48 h. For this, both cell types were grown in DMEM (1x).", "Cells were cultured on glass‐bottomed fluorodishes before exposure to Ca2+‐free balanced salt solution (BSS) + carboxyfluorescein (200 μM) for 10 min and Ca2+‐containing BSS + carboxyfluorescein (200 μM) for 5 min. Cells were washed with Ca2+‐containing BSS before imaging with a Cool Snap HQ CCD camera (Roper Scientific, Gottingen, Germany) and Metamorph software (v7.75, Universal Imaging Corp., Marlow, UK). Approximately 10–12 images were taken per dish, and regions of interest (ROI) drawn around individual cells (~15 cell/image). Two separate dishes were imaged per condition for each N number. The integrated density was measured using Fiji software (v2.1.0, ImageJ, LOCI, Wisconsin, USA), as described in detail previously.\n18\n Percentage dye uptake was calculated using these values, where treatment was set to 100%.", "The ATP luminescence assay from PerkinElmer is a quantitative assay which measures the amount of ATP in the cell supernatant. Cells were seeded at 1 × 104 following the treatment protocol before being incubated with the substrate solution as provided in the kit. The foil‐covered plate was placed on a shaker for 5 min, before being dark‐adapted for 10 min. Luminescence was measured using a Chameleon plate reader and provided a direct measure of hemichannel‐mediated ATP release.", "HK2 and TK173 cells were stimulated with TGFβ1 (10 ng/ml) ± Tonabersat at 100 μM and 10 μM respectively for 48 h prior to fixing with paraformaldehyde (4%), and subsequent blocking with goat serum (10%) for 1 h at room temperature (RT). Antibodies against fibronectin (Santa Cruz sc‐271098), N‐cadherin (Abcam ab18203), Cx43 (Abcam ab11370), and vimentin (Cell Signaling Technology D21H3), (all 1:200) were used for incubation overnight at 4°C. After washing, cells were incubated with nuclear stain, 4′,6‐diamidino‐2‐phenylindole (DAPI) (1 mmoL/L) for 3 min. Cells were incubated with Alexa‐Fluor 488 for 1 h at RT before visualisation using a Leica TC SP8 confocal microscope (Wetzlar, Germany).", "Whole cell lysates were prepared and separated using SDS‐PAGE gel electrophoresis at 125 volts (V) for 1.5 h, before transfer at 100 V for 1 h, as previously described.\n19\n Membranes were blocked before being probed overnight at 4°C with primary antibodies against Cx43, β‐catenin (Cell Signaling Technology D10A8), N‐cadherin, fibronectin, collagen I (Abcam ab34710), (all at 1:1000) and α‐tubulin (Sigma‐Aldrich T5168), at a dilution of 1:20,000, as a house‐keeping protein. After washing, membranes were probed with secondary antibody (goat anti‐rabbit 800 and/or goat anti‐mouse) at 1:20,000 for 1 h at RT. Bands were visualised using an Odyssey Fc imaging unit before semi‐quantification and analysis using ImageStudioLite software (5.2.5).", "All data are presented as mean value ± SEM. Statistical analysis was performed using ANOVA and Tukey post‐test; p ≤ 0.05 was considered statistically significant, with ‘n’ denoting sample number.", "Previous studies link increased Cx43 expression to inflammation and fibrosis in a model of renal disease,\n6\n, \n20\n while findings from our laboratory report that aberrant Cx43 hemichannel‐mediated ATP release from TGFβ1‐treated tubular epithelial cells evoked phenotypic and functional changes, characteristic of tubular damage.\n12\n, \n21\n, \n22\n Consequently, the current study determined if this effect was accentuated in high glucose and if the response could be decreased using Tonabersat, an efficient Cx43 hemichannel blocker. Human kidney (HK2) cells were cultured at low (5 mM) and high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) at high glucose for 48 h. Glucose (25 mM) increased dye uptake (25.3 ± 8.8%, p < 0.05), compared to low glucose (5 mM) control (Figure 1b,c). The response was amplified by 16.4% in the presence of TGFβ1 (p < 0.001). The combined effect of high glucose and TGFβ1 was reduced by 42.3% ± 5.3% when cells were preincubated with Tonabersat (Figure 1c; p < 0.001). In Figure 1d, TGFβ1 increased ATP release at low (32.3 ± 10.2%; p < 0.05) and high (55.7%, p < 0.001) glucose, with the response at 25 mM glucose significantly decreased by Tonabersat (36.9 ± 12.3%, p < 0.05).\nTonabersat negates TGFβ1/glucose‐evoked increases in hemichannel activity in (HK2) proximal tubule epithelial cells. Cells were cultured for 48 h in either low (5 mM) or high (25 mM) glucose ± TGFβ1 (10 ng/mL) ± Tonabersat (Tb; 100 μM). Panel (a) outlines the protocol used to measure Cx43 hemichannel‐mediated carboxyfluorescein (CBF) dye uptake (BSS is balanced salt solution), while panel (b) provides representative images for dye uptake in each condition. Carboxyfluorescein studies (c) assessed changes in hemichannel mediate, and an ATPlite assay used luminescence as a direct measure of ATP release (d). Data represents mean ± SEM, n = 4–6, where *p < 0.05, **p < 0.01, ***p < 0.001.", "We previously reported that UUO causes disassembly of the adherens junction complex\n12\n and that this is accompanied by increased interstitial fibrosis.\n6\n In the absence of glycaemic injury, this effect was partly restored in the Cx43+/− mouse model of UUO. Consequently, we evaluated the effect of TGFβ1 on Cx43, adherens junction (N‐cadherin and β‐catenin) and ECM (fibronectin and collagen I) protein expression in high glucose (Figure 2). Human kidney (HK2) cells were treated for 48 h in either low (5 mM) or high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) to block ATP release (Figure 2a). Compared to low glucose control, the cytokine increased expression of total Cx43 by 76.4 ± 10.2% (Figure 2b; p < 0.001), N‐cadherin by 39.8 ± 7.8% (Figure 2d; p < 0.01), fibronectin by 52.1 ± 6.7% (Figure 2 e; p < 0.001) and collagen I by 28.8 ± 8.6% (Figure 2f; p < 0.05). This effect was amplified in the presence of high (25 mM) glucose, where TGFβ1 increased N‐cadherin (51.3%, p < 0.001), fibronectin (67.7%, p < 0.001) and collagen I (88.5%, p < 0.001) expression. The effect of TGFβ1 on Cx43 was not accentuated under high (25 mM) glucose conditions (64.4%, p < 0.001), and there were no effects of glucose or TGFβ1 on β‐catenin expression (Figure 2c).\nTonabersat inhibits TGFβ1/glucose‐evoked increases in Cx43, adherens junction and ECM protein expression. HK2 cells were cultured for 48 h in either low (5 mM) or high (25 mM) glucose ± TGFβ1 (10 ng/mL) ± Tonabersat (Tb; 100 μM). Tonabersat blocks Cx43 hemichannel‐mediated ATP release (a). Western blot examined whole cell protein expression (%) for Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f). Data shown illustrate a representative blot and mean data ± SEM (n = 4–6), where *p < 0.05, **p < 0.01, ***p < 0.001.\nHaving determined that Tonabersat blocks TGFβ1‐induced changes in hemichannel activity (Figure 1), we assessed if TGFβ1‐evoked changes in protein expression under conditions of high glucose were hemichannel mediated. As shown in Figure 2, Tonabersat decreased high glucose and TGFβ1‐induced increases in Cx43 expression by 46 ± 10.3% (p < 0.01), while fibronectin and collagen I expression were decreased by 35.7 ± 4.8%, (p < 0.01) and 40.6 ± 5.4%, (p < 0.001), respectively. There was no effect on N‐cadherin.", "Tubulointerstitial fibrosis (TIF) is the key underlying pathology of diabetic kidney disease and develops in response to activation of multiple cell types in and around the proximal tubules.\n23\n Medullary fibroblasts contribute to the onset and progression of TIF in diabetic nephropathy. With previous studies linking the heterozygous Cx43+/− UUO mouse to decreased fibroblast activation in the kidney cortex,\n6\n we wanted to assess (i) if renal fibroblasts (TK173) have functional hemichannels and (ii) if Tonabersat could block effects of glucose and TGFβ1 on Cx43 hemichannel‐mediated release of ATP. Renal fibroblasts were treated in low (5 mM) and high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (10 μM) at high glucose for 48 h. Carboxyfluorescein measured hemichannel‐mediated dye uptake and ATP luminescence assays quantified ATP release.\nAs shown in Figure 3a,b, high glucose (25 mM) augmented TGFβ1‐evoked dye uptake by 28.1% (compared high glucose alone; p < 0.05). The combined effect of glucose and cytokine was completely negated when cells were co‐incubated with Tonabersat (49.8 ± 4.6% reduction; p < 0.001). The ability of Tonabersat to reduce dye uptake in response to TGFβ1 and high glucose was paralleled by a 43 ± 5.9% decrease in ATP release (Figure 3c; p < 0.05).\nTonabersat negates TGFβ1/glucose‐evoked hemichannel mediated ATP release in (TK173) renal fibroblasts. Cells were treated with TGFβ1 (10 ng/ml) in low or high glucose ± Tonabersat (Tb; 10 μM) for 48 h. Panel (a) shows representative images for carboxyfluorescein dye uptake in each condition. Mean data (± SEM, n = 4–6) demonstrated that TGFβ1 increased dye uptake, an effect that was amplified when co‐incubated at high glucose (panel (b)). Examining ATP release, this high glucose TGFβ1‐induced response was reversed to near basal (5 mM glucose) levels when cells were pre‐incubated with Tonabersat (panel (c)). Significances are represented as *p < 0.05, **p < 0.01, ***p < 0.001.", "Having ascertained that TGFβ1 stimulates Cx43 hemichannel activity in renal fibroblasts, an effect augmented in the presence of high glucose, we investigated if blocking this activity could confer any phenotypic protection. Immunocytochemistry (Figure 4a) and western blotting (Figure 4b–f) were used to determine TGFβ1‐induced changes in protein localisation and expression at low (5 mM) and high (25 mM) glucose, before assessing the ability of Tonabersat to dampen the combined effect of glycaemic and cytokine challenge in TK173 fibroblasts.\nTonabersat reverses TGFβ1/glucose‐evoked changes in ECM and adherens protein expression and localisation in (TK173) renal fibroblasts. In panel (a) immunocytochemistry revealed the effect of the pro‐fibrotic cytokine TGFβ1 (10 ng/ml) on the localisation of Cx43, N‐cadherin, vimentin and fibronectin in low (5 mM) and high (25 mM) glucose ± Tonabersat (Tb; 10 μM) in TK173 fibroblasts. Western blot examined whole cell protein expression (%) of Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f). Representative blots and mean data ± SEM (n = 4–6) are shown, where *p < 0.05, **p < 0.01, ***p < 0.001.\nThe pro‐fibrotic cytokine TGFβ1 had negligible effects on localisation of candidate proteins, but increased expression in both low (5mM) and high (25mM) glucose as determined by immunocytochemistry. This effect was particularly evident with ECM proteins fibronectin and vimentin (Figure 4a). As an intermediate filament protein, vimentin was used in place of collagen I which was poorly resolved using immunocytochemistry. Tonabersat reversed the combined effects of high glucose and cytokine. Semi‐quantification of protein changes using western blot determined that in the presence of high glucose, TGFβ1 increased expression of Cx43 (Figure 4b 66.1%, p < 0.001), β‐catenin (Figure 4c 32.7%, p < 0.05), N‐cadherin (Figure 4d 45%, p < 0.05), fibronectin (Figure 4e 47.6%, p < 0.01) and collagen I (Figure 4f 26.4%, p=NS). Tonabersat inhibited these effects for Cx43 (54.3 ± 12.6%, p < 0.01), N‐cadherin (40.9 ± 11.8%, p < 0.05), fibronectin (34.3 ± 4.2%, p < 0.05) and collagen I (30.8 ± 3%, p=NS).", "Having determined that high glucose in combination with TGFβ1 evokes a Tonabersat‐sensitive increase in ATP release (Figure 3) and increased expression of tubular injury associated proteins (Figure 4), we further explored a downstream role for ATP‐mediated purinergic signalling in driving high glucose and TGFβ1‐induced changes. An ATP‐diphosphohydrolase, apyrase (apy) catalyses the sequential hydrolysis of ATP to ADP and ADP to AMP, releasing inorganic phosphate and reducing the extracellular concentration of ATP ([ATP]e) to decrease activation of P2‐purinergic receptors (Figure 5a). TK173 cells were treated in low or high glucose with TGFβ1 (10 ng/ml) ± apyrase (100 IU/ml) for 48 h. Western blot assessed changes in whole cell protein expression. Compared to high glucose alone, TGFβ1 plus 25 mM glucose increased expression of Cx43 (Figure 5b 66.1%, p < 0.01), β‐catenin (Figure 5c 32.7%, p < 0.05), N‐cadherin (Figure 5d 45%, p < 0.01), fibronectin (Figure 5e 35.2%, p < 0.05) and collagen I (Figure 5f 26.4%, p=NS). These changes decreased by 51.4 ± 14.2% (Cx43; p < 0.05), 34.0 ± 7.0%, (N‐cadherin; p < 0.05), 26.6 ± 1.3%, (fibronectin; p < 0.05), and 47.8 ± 12.3%, (collagen I; p < 0.05), when cells were co‐incubated with apyrase. Hydrolysis of ATP failed to significantly alter TGFβ1/glucose‐induced changes in β‐catenin.\nReducing activation of the P2X7 receptor reverses TGFβ1/glucose‐induced changes in ECM and adherens junction protein expression in (TK173) renal fibroblasts. To reduce activation of the P2X7R, either the concentration of TGFβ1/glucose‐evoked extracellular ATP ([ATP]e) was reduced by apyrase (apy), which catalyses the sequential hydrolysis of ATP to ADP and AMP, or the P2X7 receptor was inhibited using A438079. Both strategies reduced markers of TGFβ1‐dependent inflammation and fibrosis (a). Western blot analysis determined whole cell protein expression (%) of Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f) in response to TGFβ1 (10 ng/ml) in low (5 mM) and high (25 mM) glucose ± apyrase (100 IU/ml) ± the P2X7R inhibitor (P2X7Ri: A438079 (50 μM)). Representative blots are shown above mean data ± SEM, n = 3–5, where *p < 0.05, **p < 0.01, ***p < 0.001.\nKnown to mediate inflammation and downstream fibrosis, activation of the P2X7 receptor (P2X7R) has been associated with tubular injury in an in vivo model of diabetic nephropathy.\n10\n We used the P2X7R inhibitor A438079 (50 μM) to determine the role of this receptor subtype in glucose/cytokine‐induced changes in TK173 fibroblasts (Figure 5a). The P2X7R inhibitor reversed TGFβ1/high glucose‐induced increases in Cx43 (Figure 5b 48.7 ± 12.7%, p < 0.05), N‐cadherin (Figure 5d 53.1 ± 13.9%, p < 0.01) and collagen I (Figure 5f 62.6 ± 2.7%, P < 0.001), and marginally reduced expression of β‐catenin (Figure 5c 21.8 ± 14.2%) and fibronectin (Figure 5 e 26.3 ± 1.9%).", "While tubular epithelial cells are often regarded as the initial drivers of renal injury, fibroblasts are considered the major ECM producing cell type. Importantly, they are recruited and activated in response to sustained tubular injury. Regulated by soluble ligands, for example, ATP, crosstalk between the tubular epithelia and fibroblasts can orchestrate many forms of disease progression\n24\n including diabetic nephropathy.\n25\n To investigate the role of the cell secretome in paracrine‐mediated signalling between each cell type, conditioned media transfer (indirect co‐culture) was performed between tubular epithelial cells and medullary fibroblasts. In doing so, we determined the downstream effects on hemichannel activity and protein expression on both HK2 cells and TK173 cells when incubated with conditioned media of the alternate cell type (Figure 6a).\nConditioned media from high glucose‐treated proximal tubule epithelial cells (HK2) evoked Tonabersat‐sensitive changes in ECM and adherens junction protein expression in (TK173) renal fibroblasts. Indirect co‐culture utilised 10 times freeze‐thawed 48 h high glucose (25 mM)‐treated conditioned media (CM) ± Tonabersat (Tb; 10 μM) to determine the effect of cell secretome on the heterotypic cell type (a). Carboxyfluorescein studies assessed changes in dye uptake when fibroblast conditioned media were added to tubular epithelial cells (b) and (c) and vice versa (d) and (e). Determined by western blotting, tubular epithelial cell secretome examined Tonabersat‐sensitive changes in fibroblast whole cell protein expression (%) for Cx43 (f), β‐catenin (g), N‐cadherin (h), fibronectin (i) and collagen I (j). Data shown illustrate representative blots and mean data ± SEM (n = 3–4), where *p < 0.05, **p < 0.01, ***p < 0.001.\nAlthough the transfer of high glucose conditioned media from fibroblasts evoked a Tonabersat‐sensitive increase in dye uptake in tubular epithelial cells (55.9%, p < 0.001) as compared to 5 mM glucose control (Figure 6b,c), it had minimal effect on HK2 protein expression (see: Figure S1).\nConditioned media from high glucose‐treated proximal tubule epithelial cells (HK2) evoked a Tonabersat‐sensitive increase in dye uptake in (TK173) renal fibroblasts of 54.5% (p < 0.001), compared to low glucose control (Figure 6d,e). Contrary to the lack of effect that fibroblast (TK173) conditioned media had on tubule epithelial (HK2) phenotype, the effects of HK2 conditioned media on TK173 dye uptake, were paralleled by changes in Cx43 expression and markers of the ECM and adherens junction. Western blot determined that high glucose conditioned media from epithelial cells increased expression of Cx43 (47.7%, Figure 6f; p < 0.001), N‐cadherin (24.1%, Figure 6h; p < 0.05) and fibronectin (60.5%, Figure 6i; p < 0.01) in fibroblasts compared to unconditioned 25 mM glucose media. Tonabersat negated this effect by 33.9 ± 11.4%, p < 0.01 (Cx43), 25.3 ± 8.9%, p < 0.05 (N‐cadherin) and 53.4 ± 7.7%, p < 0.05 (fibronectin). There were minimal effects on β‐catenin (Figure 6g) or collagen I (Figure 6j).", "CEH and PES were involved in conceptualisation, supervision and project administration. BMW and CLC were involved in experimental studies. BMW, PES and CEH were involved in writing—original draft preparation. BMW, CLC, ID, PES and CEH were involved in writing. All authors have read and agreed to the published version of the manuscript.", "BMW is a recipient of a PhD studentship supported by Diabetes UK and the Masonic Charitable Foundation. (18/0005919; awarded to PES & CEH)." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Novelty statement", "What is already known?", "What this study found?", "Implications?", "INTRODUCTION", "METHODS", "Materials", "Cell culture and treatment", "Carboxyfluorescein dye uptake", "\nATP luminescence assay", "Immunocytochemistry", "Western blotting", "Statistical analysis", "RESULTS", "Tonabersat blocks hemichannel‐mediated ATP release in tubular epithelial cells", "Blocking Cx43 hemichannels inhibits TGFβ1/glucose‐induced increases in expression of Cx43 and markers of tubular damage", "Tonabersat blocks hemichannel‐mediated ATP release in renal fibroblasts", "Tonabersat inhibits TGFβ1/glucose‐induced changes in Cx43, N‐cadherin and ECM localisation and expression in renal fibroblasts", "\nATP mediates TGFβ1/glucose‐induced changes in Cx43, N‐cadherin and ECM expression in renal fibroblasts", "Tonabersat partially negates epithelial cell secretome‐mediated changes on fibroblast hemichannel number and protein expression", "DISCUSSION", "AUTHOR CONTRIBUTIONS", "FUNDING INFORMATION", "CONFLICT OF INTEREST", "Supporting information" ]
[ "", "\nCx43 hemichannels release ATP in TGFβ1‐treated tubular epithelial cells.\n\nCx43 hemichannels release ATP in TGFβ1‐treated tubular epithelial cells.", "\nTGFβ1 increases Cx43 hemichannel‐mediated ATP release in TK173 renal fibroblasts and HK2 tubular epithelial cells, an effect amplified by high glucose.Aberrant ATP release initiates phenotypic changes via P2X7 receptors.Co‐culture of TK173 cells with high glucose conditioned HK2 media, increased Cx43 hemichannel‐mediated dye uptake and ECM protein expression.Characteristic of late‐stage kidney damage, effects were partly blocked with Tonabersat.\n\nTGFβ1 increases Cx43 hemichannel‐mediated ATP release in TK173 renal fibroblasts and HK2 tubular epithelial cells, an effect amplified by high glucose.\nAberrant ATP release initiates phenotypic changes via P2X7 receptors.\nCo‐culture of TK173 cells with high glucose conditioned HK2 media, increased Cx43 hemichannel‐mediated dye uptake and ECM protein expression.\nCharacteristic of late‐stage kidney damage, effects were partly blocked with Tonabersat.", "\nTonabersat blocks Cx43 hemichannel‐mediated communication within and between tubular epithelial cells (HK2) and medullary renal fibroblasts (TK173), protecting against phenotypic changes characteristic of diabetic nephropathy.\n\nTonabersat blocks Cx43 hemichannel‐mediated communication within and between tubular epithelial cells (HK2) and medullary renal fibroblasts (TK173), protecting against phenotypic changes characteristic of diabetic nephropathy.", "Worldwide, diabetic kidney disease develops in around 40% of those with diabetes and is the leading cause of chronic kidney disease (CKD).\n1\n In 2017, 1.2 million people died from CKD while 697.5 million cases were recorded globally.\n2\n In the United Kingdom, 30% of those entering maintenance renal replacement therapy have diabetic nephropathy as their primary renal disease.\n3\n Currently, there are no curative therapeutic options to prevent transition to end‐stage kidney disease and innovative approaches are urgently required.\nAs the main contributor to end‐stage renal failure, understanding mechanisms by which glucose alters renal function is critical to identifying future strategies for prevention and/or arrest of diabetic nephropathy (DN). Early damage in DN is characterised by persistent inflammation and fibrosis of the tubulointerstitium, where fibrosis develops in response to various morphological and phenotypic changes, including partial epithelial‐to‐mesenchymal transition (pEMT), and extracellular matrix (ECM) remodelling.\n4\n In the face of sustained glycaemic injury, soluble chemokines, adhesion molecules and cytokines, for example, transforming growth factor beta‐1 (TGFβ1), recruit and activate infiltrating immune cells and resident fibroblasts to mediate inflammation and fibrosis, severity of which dictates disease progression.\n5\n\n\nWork by Abed et al., reported that heterozygous connexin (Cx)43+/− mice exhibited decreased tubular macrophage infiltration and reduced numbers of active fibroblasts when surgically induced to exhibit interstitial inflammation and fibrosis via unilateral ureteral obstruction (UUO).\n6\n Moreover, our laboratory demonstrated that these mice exhibit reduced disassembly of adherens junction and tight junction proteins, the former of which is considered an initiating trigger for pEMT in the injured tubules.\n7\n\n\nConnexins oligomerise to form hexameric structures, before being transported to the cell membrane where they form hemichannels which allow for the local release of various danger‐associated molecular patterns (DAMPs), for example, adenosine triphosphate (ATP), in response to pathological triggers, including hyperglycaemia\n8\n and inflammation.\n9\n Excess ATP within the intercellular environment activates P2X7 purinergic receptors\n10\n to trigger inflammatory and fibrotic events in both resident tubule epithelial cells\n11\n and other associated cell types local to the proximal kidney, for example, macrophages.\n10\n We previously demonstrated that pharmacologically inhibiting Cx43‐mediated hemichannel activity using Cx43 hemichannel blocker Peptide 5, protects against early tubular injury in an in vitro model of experimental chronic kidney disease under euglycaemic conditions.\n12\n Similar to Peptide 5, Tonabersat; a benzopyran derivative originally reported to inhibit gap junction communication,\n13\n has recently been shown to be equivalent to Peptide 5 in its capacity to act as a blocker of Cx43 hemichannels.\n14\n, \n15\n, \n16\n Consequently, Tonabersat (Xiflam) has since been reported to confer protection in both in vivo models of age‐related macular degeneration\n14\n and diabetic retinopathy.\n14\n, \n17\n Currently entering Phase2b clinical trials for diabetic macular oedema and retinopathy, its ability to target hemichannels in the diabetic kidney and confer anti‐inflammatory protection are yet to be reported.\nIn this study, we tested whether blocking Cx43‐mediated hemichannel ATP release decreased P2X7R activation within and between tubule epithelial cells and renal fibroblasts in conditions designed to mimic diabetic nephropathy. We posit that a heterotypic pro‐inflammatory relationship exists between tubular epithelial cells and resident fibroblasts, and that blocking Cx43‐mediated ATP release can dampen this interaction. The data demonstrate the importance of Cx43 hemichannel‐mediated paracrine signalling in kidney damage, and suggest the potential benefit of using Cx43 hemichannel blockers in managing tubulointerstitial fibrosis in late‐stage DN.", "[SUBTITLE] Materials [SUBSECTION] Human kidney (HK2) proximal tubule epithelial cells were purchased from American Type Culture Collection (ATCC) (LGC Standards; Manassas, United States). Medullary renal fibroblasts (TK173) were derived from normal human kidneys and gifted from Dr. Isak Demirel (Örebro University, Sweden). Tissue culture plastic and supplies were from Sarstedt Inc (Leicester, UK), while tissue culture media and fetal calf serum (FCS) were from Fisher Scientific (Loughborough, UK). Penicillin/Streptomycin was from Sigma‐Aldrich (Dorset, UK), while epithelial growth factor (EGF) was purchased from ProSpec (Rotherham, UK). Glass‐bottomed fluorodishes for carboxyfluorescein dye uptake studies were from Thistle Scientific (Glasgow, UK). For western blotting, all membranes, buffers and equipment were obtained from Licor Biosciences (Lincoln, USA). Recombinant TGFβ1 (PHG9214) was from Fisher Scientific (Loughborough, UK), while Tonabersat was purchased from Cambridge Bioscience (Cambridge, UK). ATPlite luminescence assay was from PerkinElmer (Llantrisant, UK). Unless otherwise stated, all chemicals were from Sigma‐Aldrich (Dorset, UK).\nHuman kidney (HK2) proximal tubule epithelial cells were purchased from American Type Culture Collection (ATCC) (LGC Standards; Manassas, United States). Medullary renal fibroblasts (TK173) were derived from normal human kidneys and gifted from Dr. Isak Demirel (Örebro University, Sweden). Tissue culture plastic and supplies were from Sarstedt Inc (Leicester, UK), while tissue culture media and fetal calf serum (FCS) were from Fisher Scientific (Loughborough, UK). Penicillin/Streptomycin was from Sigma‐Aldrich (Dorset, UK), while epithelial growth factor (EGF) was purchased from ProSpec (Rotherham, UK). Glass‐bottomed fluorodishes for carboxyfluorescein dye uptake studies were from Thistle Scientific (Glasgow, UK). For western blotting, all membranes, buffers and equipment were obtained from Licor Biosciences (Lincoln, USA). Recombinant TGFβ1 (PHG9214) was from Fisher Scientific (Loughborough, UK), while Tonabersat was purchased from Cambridge Bioscience (Cambridge, UK). ATPlite luminescence assay was from PerkinElmer (Llantrisant, UK). Unless otherwise stated, all chemicals were from Sigma‐Aldrich (Dorset, UK).\n[SUBTITLE] Cell culture and treatment [SUBSECTION] HK2 cells (passages 9–21) were maintained in Dulbecco's Modified Eagle's Medium (DMEM)/Ham's F12 medium, while TK173 (passages 4–10) were maintained in DMEM(1x) + pyruvate. HK2 media contained penicillin/streptomycin (2%) and EGF (5 ng/ml). TK173 media were supplemented with minimum essential medium non‐essential amino acids (MEM NEAA). All culture media contained FCS (10%). Cells were cultured at 37°C in a humidified environment with 5% CO2.\nFor HK2 treatments, cells were cultured in low (5 mM) glucose DMEM/F12 (5 mmoL/L) for 48 h, followed by serum starvation overnight prior to treatment with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) at either 5 mM or 25 mM D‐glucose. For TK173 treatments, cells were seeded in culture medium for 24 h, followed by overnight serum starvation before treatment in DMEM(1x), containing 1% FCS with TGFβ1 (10 ng/ml) ± Tonabersat (10 μM) ± apyrase (100 IU/ml) ± P2X7R inhibitor A438079 (50 μM). For co‐culture experiments, cells were treated for 48 h in 5 mM or 25 mM D‐glucose ± Tonabersat (10 μM), before harvesting the media and freeze‐thawing 10x. Conditioned media were then used for treatment of the alternative cell type for 48 h. For this, both cell types were grown in DMEM (1x).\nHK2 cells (passages 9–21) were maintained in Dulbecco's Modified Eagle's Medium (DMEM)/Ham's F12 medium, while TK173 (passages 4–10) were maintained in DMEM(1x) + pyruvate. HK2 media contained penicillin/streptomycin (2%) and EGF (5 ng/ml). TK173 media were supplemented with minimum essential medium non‐essential amino acids (MEM NEAA). All culture media contained FCS (10%). Cells were cultured at 37°C in a humidified environment with 5% CO2.\nFor HK2 treatments, cells were cultured in low (5 mM) glucose DMEM/F12 (5 mmoL/L) for 48 h, followed by serum starvation overnight prior to treatment with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) at either 5 mM or 25 mM D‐glucose. For TK173 treatments, cells were seeded in culture medium for 24 h, followed by overnight serum starvation before treatment in DMEM(1x), containing 1% FCS with TGFβ1 (10 ng/ml) ± Tonabersat (10 μM) ± apyrase (100 IU/ml) ± P2X7R inhibitor A438079 (50 μM). For co‐culture experiments, cells were treated for 48 h in 5 mM or 25 mM D‐glucose ± Tonabersat (10 μM), before harvesting the media and freeze‐thawing 10x. Conditioned media were then used for treatment of the alternative cell type for 48 h. For this, both cell types were grown in DMEM (1x).\n[SUBTITLE] Carboxyfluorescein dye uptake [SUBSECTION] Cells were cultured on glass‐bottomed fluorodishes before exposure to Ca2+‐free balanced salt solution (BSS) + carboxyfluorescein (200 μM) for 10 min and Ca2+‐containing BSS + carboxyfluorescein (200 μM) for 5 min. Cells were washed with Ca2+‐containing BSS before imaging with a Cool Snap HQ CCD camera (Roper Scientific, Gottingen, Germany) and Metamorph software (v7.75, Universal Imaging Corp., Marlow, UK). Approximately 10–12 images were taken per dish, and regions of interest (ROI) drawn around individual cells (~15 cell/image). Two separate dishes were imaged per condition for each N number. The integrated density was measured using Fiji software (v2.1.0, ImageJ, LOCI, Wisconsin, USA), as described in detail previously.\n18\n Percentage dye uptake was calculated using these values, where treatment was set to 100%.\nCells were cultured on glass‐bottomed fluorodishes before exposure to Ca2+‐free balanced salt solution (BSS) + carboxyfluorescein (200 μM) for 10 min and Ca2+‐containing BSS + carboxyfluorescein (200 μM) for 5 min. Cells were washed with Ca2+‐containing BSS before imaging with a Cool Snap HQ CCD camera (Roper Scientific, Gottingen, Germany) and Metamorph software (v7.75, Universal Imaging Corp., Marlow, UK). Approximately 10–12 images were taken per dish, and regions of interest (ROI) drawn around individual cells (~15 cell/image). Two separate dishes were imaged per condition for each N number. The integrated density was measured using Fiji software (v2.1.0, ImageJ, LOCI, Wisconsin, USA), as described in detail previously.\n18\n Percentage dye uptake was calculated using these values, where treatment was set to 100%.\n[SUBTITLE] \nATP luminescence assay [SUBSECTION] The ATP luminescence assay from PerkinElmer is a quantitative assay which measures the amount of ATP in the cell supernatant. Cells were seeded at 1 × 104 following the treatment protocol before being incubated with the substrate solution as provided in the kit. The foil‐covered plate was placed on a shaker for 5 min, before being dark‐adapted for 10 min. Luminescence was measured using a Chameleon plate reader and provided a direct measure of hemichannel‐mediated ATP release.\nThe ATP luminescence assay from PerkinElmer is a quantitative assay which measures the amount of ATP in the cell supernatant. Cells were seeded at 1 × 104 following the treatment protocol before being incubated with the substrate solution as provided in the kit. The foil‐covered plate was placed on a shaker for 5 min, before being dark‐adapted for 10 min. Luminescence was measured using a Chameleon plate reader and provided a direct measure of hemichannel‐mediated ATP release.\n[SUBTITLE] Immunocytochemistry [SUBSECTION] HK2 and TK173 cells were stimulated with TGFβ1 (10 ng/ml) ± Tonabersat at 100 μM and 10 μM respectively for 48 h prior to fixing with paraformaldehyde (4%), and subsequent blocking with goat serum (10%) for 1 h at room temperature (RT). Antibodies against fibronectin (Santa Cruz sc‐271098), N‐cadherin (Abcam ab18203), Cx43 (Abcam ab11370), and vimentin (Cell Signaling Technology D21H3), (all 1:200) were used for incubation overnight at 4°C. After washing, cells were incubated with nuclear stain, 4′,6‐diamidino‐2‐phenylindole (DAPI) (1 mmoL/L) for 3 min. Cells were incubated with Alexa‐Fluor 488 for 1 h at RT before visualisation using a Leica TC SP8 confocal microscope (Wetzlar, Germany).\nHK2 and TK173 cells were stimulated with TGFβ1 (10 ng/ml) ± Tonabersat at 100 μM and 10 μM respectively for 48 h prior to fixing with paraformaldehyde (4%), and subsequent blocking with goat serum (10%) for 1 h at room temperature (RT). Antibodies against fibronectin (Santa Cruz sc‐271098), N‐cadherin (Abcam ab18203), Cx43 (Abcam ab11370), and vimentin (Cell Signaling Technology D21H3), (all 1:200) were used for incubation overnight at 4°C. After washing, cells were incubated with nuclear stain, 4′,6‐diamidino‐2‐phenylindole (DAPI) (1 mmoL/L) for 3 min. Cells were incubated with Alexa‐Fluor 488 for 1 h at RT before visualisation using a Leica TC SP8 confocal microscope (Wetzlar, Germany).\n[SUBTITLE] Western blotting [SUBSECTION] Whole cell lysates were prepared and separated using SDS‐PAGE gel electrophoresis at 125 volts (V) for 1.5 h, before transfer at 100 V for 1 h, as previously described.\n19\n Membranes were blocked before being probed overnight at 4°C with primary antibodies against Cx43, β‐catenin (Cell Signaling Technology D10A8), N‐cadherin, fibronectin, collagen I (Abcam ab34710), (all at 1:1000) and α‐tubulin (Sigma‐Aldrich T5168), at a dilution of 1:20,000, as a house‐keeping protein. After washing, membranes were probed with secondary antibody (goat anti‐rabbit 800 and/or goat anti‐mouse) at 1:20,000 for 1 h at RT. Bands were visualised using an Odyssey Fc imaging unit before semi‐quantification and analysis using ImageStudioLite software (5.2.5).\nWhole cell lysates were prepared and separated using SDS‐PAGE gel electrophoresis at 125 volts (V) for 1.5 h, before transfer at 100 V for 1 h, as previously described.\n19\n Membranes were blocked before being probed overnight at 4°C with primary antibodies against Cx43, β‐catenin (Cell Signaling Technology D10A8), N‐cadherin, fibronectin, collagen I (Abcam ab34710), (all at 1:1000) and α‐tubulin (Sigma‐Aldrich T5168), at a dilution of 1:20,000, as a house‐keeping protein. After washing, membranes were probed with secondary antibody (goat anti‐rabbit 800 and/or goat anti‐mouse) at 1:20,000 for 1 h at RT. Bands were visualised using an Odyssey Fc imaging unit before semi‐quantification and analysis using ImageStudioLite software (5.2.5).\n[SUBTITLE] Statistical analysis [SUBSECTION] All data are presented as mean value ± SEM. Statistical analysis was performed using ANOVA and Tukey post‐test; p ≤ 0.05 was considered statistically significant, with ‘n’ denoting sample number.\nAll data are presented as mean value ± SEM. Statistical analysis was performed using ANOVA and Tukey post‐test; p ≤ 0.05 was considered statistically significant, with ‘n’ denoting sample number.", "Human kidney (HK2) proximal tubule epithelial cells were purchased from American Type Culture Collection (ATCC) (LGC Standards; Manassas, United States). Medullary renal fibroblasts (TK173) were derived from normal human kidneys and gifted from Dr. Isak Demirel (Örebro University, Sweden). Tissue culture plastic and supplies were from Sarstedt Inc (Leicester, UK), while tissue culture media and fetal calf serum (FCS) were from Fisher Scientific (Loughborough, UK). Penicillin/Streptomycin was from Sigma‐Aldrich (Dorset, UK), while epithelial growth factor (EGF) was purchased from ProSpec (Rotherham, UK). Glass‐bottomed fluorodishes for carboxyfluorescein dye uptake studies were from Thistle Scientific (Glasgow, UK). For western blotting, all membranes, buffers and equipment were obtained from Licor Biosciences (Lincoln, USA). Recombinant TGFβ1 (PHG9214) was from Fisher Scientific (Loughborough, UK), while Tonabersat was purchased from Cambridge Bioscience (Cambridge, UK). ATPlite luminescence assay was from PerkinElmer (Llantrisant, UK). Unless otherwise stated, all chemicals were from Sigma‐Aldrich (Dorset, UK).", "HK2 cells (passages 9–21) were maintained in Dulbecco's Modified Eagle's Medium (DMEM)/Ham's F12 medium, while TK173 (passages 4–10) were maintained in DMEM(1x) + pyruvate. HK2 media contained penicillin/streptomycin (2%) and EGF (5 ng/ml). TK173 media were supplemented with minimum essential medium non‐essential amino acids (MEM NEAA). All culture media contained FCS (10%). Cells were cultured at 37°C in a humidified environment with 5% CO2.\nFor HK2 treatments, cells were cultured in low (5 mM) glucose DMEM/F12 (5 mmoL/L) for 48 h, followed by serum starvation overnight prior to treatment with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) at either 5 mM or 25 mM D‐glucose. For TK173 treatments, cells were seeded in culture medium for 24 h, followed by overnight serum starvation before treatment in DMEM(1x), containing 1% FCS with TGFβ1 (10 ng/ml) ± Tonabersat (10 μM) ± apyrase (100 IU/ml) ± P2X7R inhibitor A438079 (50 μM). For co‐culture experiments, cells were treated for 48 h in 5 mM or 25 mM D‐glucose ± Tonabersat (10 μM), before harvesting the media and freeze‐thawing 10x. Conditioned media were then used for treatment of the alternative cell type for 48 h. For this, both cell types were grown in DMEM (1x).", "Cells were cultured on glass‐bottomed fluorodishes before exposure to Ca2+‐free balanced salt solution (BSS) + carboxyfluorescein (200 μM) for 10 min and Ca2+‐containing BSS + carboxyfluorescein (200 μM) for 5 min. Cells were washed with Ca2+‐containing BSS before imaging with a Cool Snap HQ CCD camera (Roper Scientific, Gottingen, Germany) and Metamorph software (v7.75, Universal Imaging Corp., Marlow, UK). Approximately 10–12 images were taken per dish, and regions of interest (ROI) drawn around individual cells (~15 cell/image). Two separate dishes were imaged per condition for each N number. The integrated density was measured using Fiji software (v2.1.0, ImageJ, LOCI, Wisconsin, USA), as described in detail previously.\n18\n Percentage dye uptake was calculated using these values, where treatment was set to 100%.", "The ATP luminescence assay from PerkinElmer is a quantitative assay which measures the amount of ATP in the cell supernatant. Cells were seeded at 1 × 104 following the treatment protocol before being incubated with the substrate solution as provided in the kit. The foil‐covered plate was placed on a shaker for 5 min, before being dark‐adapted for 10 min. Luminescence was measured using a Chameleon plate reader and provided a direct measure of hemichannel‐mediated ATP release.", "HK2 and TK173 cells were stimulated with TGFβ1 (10 ng/ml) ± Tonabersat at 100 μM and 10 μM respectively for 48 h prior to fixing with paraformaldehyde (4%), and subsequent blocking with goat serum (10%) for 1 h at room temperature (RT). Antibodies against fibronectin (Santa Cruz sc‐271098), N‐cadherin (Abcam ab18203), Cx43 (Abcam ab11370), and vimentin (Cell Signaling Technology D21H3), (all 1:200) were used for incubation overnight at 4°C. After washing, cells were incubated with nuclear stain, 4′,6‐diamidino‐2‐phenylindole (DAPI) (1 mmoL/L) for 3 min. Cells were incubated with Alexa‐Fluor 488 for 1 h at RT before visualisation using a Leica TC SP8 confocal microscope (Wetzlar, Germany).", "Whole cell lysates were prepared and separated using SDS‐PAGE gel electrophoresis at 125 volts (V) for 1.5 h, before transfer at 100 V for 1 h, as previously described.\n19\n Membranes were blocked before being probed overnight at 4°C with primary antibodies against Cx43, β‐catenin (Cell Signaling Technology D10A8), N‐cadherin, fibronectin, collagen I (Abcam ab34710), (all at 1:1000) and α‐tubulin (Sigma‐Aldrich T5168), at a dilution of 1:20,000, as a house‐keeping protein. After washing, membranes were probed with secondary antibody (goat anti‐rabbit 800 and/or goat anti‐mouse) at 1:20,000 for 1 h at RT. Bands were visualised using an Odyssey Fc imaging unit before semi‐quantification and analysis using ImageStudioLite software (5.2.5).", "All data are presented as mean value ± SEM. Statistical analysis was performed using ANOVA and Tukey post‐test; p ≤ 0.05 was considered statistically significant, with ‘n’ denoting sample number.", "[SUBTITLE] Tonabersat blocks hemichannel‐mediated ATP release in tubular epithelial cells [SUBSECTION] Previous studies link increased Cx43 expression to inflammation and fibrosis in a model of renal disease,\n6\n, \n20\n while findings from our laboratory report that aberrant Cx43 hemichannel‐mediated ATP release from TGFβ1‐treated tubular epithelial cells evoked phenotypic and functional changes, characteristic of tubular damage.\n12\n, \n21\n, \n22\n Consequently, the current study determined if this effect was accentuated in high glucose and if the response could be decreased using Tonabersat, an efficient Cx43 hemichannel blocker. Human kidney (HK2) cells were cultured at low (5 mM) and high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) at high glucose for 48 h. Glucose (25 mM) increased dye uptake (25.3 ± 8.8%, p < 0.05), compared to low glucose (5 mM) control (Figure 1b,c). The response was amplified by 16.4% in the presence of TGFβ1 (p < 0.001). The combined effect of high glucose and TGFβ1 was reduced by 42.3% ± 5.3% when cells were preincubated with Tonabersat (Figure 1c; p < 0.001). In Figure 1d, TGFβ1 increased ATP release at low (32.3 ± 10.2%; p < 0.05) and high (55.7%, p < 0.001) glucose, with the response at 25 mM glucose significantly decreased by Tonabersat (36.9 ± 12.3%, p < 0.05).\nTonabersat negates TGFβ1/glucose‐evoked increases in hemichannel activity in (HK2) proximal tubule epithelial cells. Cells were cultured for 48 h in either low (5 mM) or high (25 mM) glucose ± TGFβ1 (10 ng/mL) ± Tonabersat (Tb; 100 μM). Panel (a) outlines the protocol used to measure Cx43 hemichannel‐mediated carboxyfluorescein (CBF) dye uptake (BSS is balanced salt solution), while panel (b) provides representative images for dye uptake in each condition. Carboxyfluorescein studies (c) assessed changes in hemichannel mediate, and an ATPlite assay used luminescence as a direct measure of ATP release (d). Data represents mean ± SEM, n = 4–6, where *p < 0.05, **p < 0.01, ***p < 0.001.\nPrevious studies link increased Cx43 expression to inflammation and fibrosis in a model of renal disease,\n6\n, \n20\n while findings from our laboratory report that aberrant Cx43 hemichannel‐mediated ATP release from TGFβ1‐treated tubular epithelial cells evoked phenotypic and functional changes, characteristic of tubular damage.\n12\n, \n21\n, \n22\n Consequently, the current study determined if this effect was accentuated in high glucose and if the response could be decreased using Tonabersat, an efficient Cx43 hemichannel blocker. Human kidney (HK2) cells were cultured at low (5 mM) and high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) at high glucose for 48 h. Glucose (25 mM) increased dye uptake (25.3 ± 8.8%, p < 0.05), compared to low glucose (5 mM) control (Figure 1b,c). The response was amplified by 16.4% in the presence of TGFβ1 (p < 0.001). The combined effect of high glucose and TGFβ1 was reduced by 42.3% ± 5.3% when cells were preincubated with Tonabersat (Figure 1c; p < 0.001). In Figure 1d, TGFβ1 increased ATP release at low (32.3 ± 10.2%; p < 0.05) and high (55.7%, p < 0.001) glucose, with the response at 25 mM glucose significantly decreased by Tonabersat (36.9 ± 12.3%, p < 0.05).\nTonabersat negates TGFβ1/glucose‐evoked increases in hemichannel activity in (HK2) proximal tubule epithelial cells. Cells were cultured for 48 h in either low (5 mM) or high (25 mM) glucose ± TGFβ1 (10 ng/mL) ± Tonabersat (Tb; 100 μM). Panel (a) outlines the protocol used to measure Cx43 hemichannel‐mediated carboxyfluorescein (CBF) dye uptake (BSS is balanced salt solution), while panel (b) provides representative images for dye uptake in each condition. Carboxyfluorescein studies (c) assessed changes in hemichannel mediate, and an ATPlite assay used luminescence as a direct measure of ATP release (d). Data represents mean ± SEM, n = 4–6, where *p < 0.05, **p < 0.01, ***p < 0.001.\n[SUBTITLE] Blocking Cx43 hemichannels inhibits TGFβ1/glucose‐induced increases in expression of Cx43 and markers of tubular damage [SUBSECTION] We previously reported that UUO causes disassembly of the adherens junction complex\n12\n and that this is accompanied by increased interstitial fibrosis.\n6\n In the absence of glycaemic injury, this effect was partly restored in the Cx43+/− mouse model of UUO. Consequently, we evaluated the effect of TGFβ1 on Cx43, adherens junction (N‐cadherin and β‐catenin) and ECM (fibronectin and collagen I) protein expression in high glucose (Figure 2). Human kidney (HK2) cells were treated for 48 h in either low (5 mM) or high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) to block ATP release (Figure 2a). Compared to low glucose control, the cytokine increased expression of total Cx43 by 76.4 ± 10.2% (Figure 2b; p < 0.001), N‐cadherin by 39.8 ± 7.8% (Figure 2d; p < 0.01), fibronectin by 52.1 ± 6.7% (Figure 2 e; p < 0.001) and collagen I by 28.8 ± 8.6% (Figure 2f; p < 0.05). This effect was amplified in the presence of high (25 mM) glucose, where TGFβ1 increased N‐cadherin (51.3%, p < 0.001), fibronectin (67.7%, p < 0.001) and collagen I (88.5%, p < 0.001) expression. The effect of TGFβ1 on Cx43 was not accentuated under high (25 mM) glucose conditions (64.4%, p < 0.001), and there were no effects of glucose or TGFβ1 on β‐catenin expression (Figure 2c).\nTonabersat inhibits TGFβ1/glucose‐evoked increases in Cx43, adherens junction and ECM protein expression. HK2 cells were cultured for 48 h in either low (5 mM) or high (25 mM) glucose ± TGFβ1 (10 ng/mL) ± Tonabersat (Tb; 100 μM). Tonabersat blocks Cx43 hemichannel‐mediated ATP release (a). Western blot examined whole cell protein expression (%) for Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f). Data shown illustrate a representative blot and mean data ± SEM (n = 4–6), where *p < 0.05, **p < 0.01, ***p < 0.001.\nHaving determined that Tonabersat blocks TGFβ1‐induced changes in hemichannel activity (Figure 1), we assessed if TGFβ1‐evoked changes in protein expression under conditions of high glucose were hemichannel mediated. As shown in Figure 2, Tonabersat decreased high glucose and TGFβ1‐induced increases in Cx43 expression by 46 ± 10.3% (p < 0.01), while fibronectin and collagen I expression were decreased by 35.7 ± 4.8%, (p < 0.01) and 40.6 ± 5.4%, (p < 0.001), respectively. There was no effect on N‐cadherin.\nWe previously reported that UUO causes disassembly of the adherens junction complex\n12\n and that this is accompanied by increased interstitial fibrosis.\n6\n In the absence of glycaemic injury, this effect was partly restored in the Cx43+/− mouse model of UUO. Consequently, we evaluated the effect of TGFβ1 on Cx43, adherens junction (N‐cadherin and β‐catenin) and ECM (fibronectin and collagen I) protein expression in high glucose (Figure 2). Human kidney (HK2) cells were treated for 48 h in either low (5 mM) or high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) to block ATP release (Figure 2a). Compared to low glucose control, the cytokine increased expression of total Cx43 by 76.4 ± 10.2% (Figure 2b; p < 0.001), N‐cadherin by 39.8 ± 7.8% (Figure 2d; p < 0.01), fibronectin by 52.1 ± 6.7% (Figure 2 e; p < 0.001) and collagen I by 28.8 ± 8.6% (Figure 2f; p < 0.05). This effect was amplified in the presence of high (25 mM) glucose, where TGFβ1 increased N‐cadherin (51.3%, p < 0.001), fibronectin (67.7%, p < 0.001) and collagen I (88.5%, p < 0.001) expression. The effect of TGFβ1 on Cx43 was not accentuated under high (25 mM) glucose conditions (64.4%, p < 0.001), and there were no effects of glucose or TGFβ1 on β‐catenin expression (Figure 2c).\nTonabersat inhibits TGFβ1/glucose‐evoked increases in Cx43, adherens junction and ECM protein expression. HK2 cells were cultured for 48 h in either low (5 mM) or high (25 mM) glucose ± TGFβ1 (10 ng/mL) ± Tonabersat (Tb; 100 μM). Tonabersat blocks Cx43 hemichannel‐mediated ATP release (a). Western blot examined whole cell protein expression (%) for Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f). Data shown illustrate a representative blot and mean data ± SEM (n = 4–6), where *p < 0.05, **p < 0.01, ***p < 0.001.\nHaving determined that Tonabersat blocks TGFβ1‐induced changes in hemichannel activity (Figure 1), we assessed if TGFβ1‐evoked changes in protein expression under conditions of high glucose were hemichannel mediated. As shown in Figure 2, Tonabersat decreased high glucose and TGFβ1‐induced increases in Cx43 expression by 46 ± 10.3% (p < 0.01), while fibronectin and collagen I expression were decreased by 35.7 ± 4.8%, (p < 0.01) and 40.6 ± 5.4%, (p < 0.001), respectively. There was no effect on N‐cadherin.\n[SUBTITLE] Tonabersat blocks hemichannel‐mediated ATP release in renal fibroblasts [SUBSECTION] Tubulointerstitial fibrosis (TIF) is the key underlying pathology of diabetic kidney disease and develops in response to activation of multiple cell types in and around the proximal tubules.\n23\n Medullary fibroblasts contribute to the onset and progression of TIF in diabetic nephropathy. With previous studies linking the heterozygous Cx43+/− UUO mouse to decreased fibroblast activation in the kidney cortex,\n6\n we wanted to assess (i) if renal fibroblasts (TK173) have functional hemichannels and (ii) if Tonabersat could block effects of glucose and TGFβ1 on Cx43 hemichannel‐mediated release of ATP. Renal fibroblasts were treated in low (5 mM) and high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (10 μM) at high glucose for 48 h. Carboxyfluorescein measured hemichannel‐mediated dye uptake and ATP luminescence assays quantified ATP release.\nAs shown in Figure 3a,b, high glucose (25 mM) augmented TGFβ1‐evoked dye uptake by 28.1% (compared high glucose alone; p < 0.05). The combined effect of glucose and cytokine was completely negated when cells were co‐incubated with Tonabersat (49.8 ± 4.6% reduction; p < 0.001). The ability of Tonabersat to reduce dye uptake in response to TGFβ1 and high glucose was paralleled by a 43 ± 5.9% decrease in ATP release (Figure 3c; p < 0.05).\nTonabersat negates TGFβ1/glucose‐evoked hemichannel mediated ATP release in (TK173) renal fibroblasts. Cells were treated with TGFβ1 (10 ng/ml) in low or high glucose ± Tonabersat (Tb; 10 μM) for 48 h. Panel (a) shows representative images for carboxyfluorescein dye uptake in each condition. Mean data (± SEM, n = 4–6) demonstrated that TGFβ1 increased dye uptake, an effect that was amplified when co‐incubated at high glucose (panel (b)). Examining ATP release, this high glucose TGFβ1‐induced response was reversed to near basal (5 mM glucose) levels when cells were pre‐incubated with Tonabersat (panel (c)). Significances are represented as *p < 0.05, **p < 0.01, ***p < 0.001.\nTubulointerstitial fibrosis (TIF) is the key underlying pathology of diabetic kidney disease and develops in response to activation of multiple cell types in and around the proximal tubules.\n23\n Medullary fibroblasts contribute to the onset and progression of TIF in diabetic nephropathy. With previous studies linking the heterozygous Cx43+/− UUO mouse to decreased fibroblast activation in the kidney cortex,\n6\n we wanted to assess (i) if renal fibroblasts (TK173) have functional hemichannels and (ii) if Tonabersat could block effects of glucose and TGFβ1 on Cx43 hemichannel‐mediated release of ATP. Renal fibroblasts were treated in low (5 mM) and high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (10 μM) at high glucose for 48 h. Carboxyfluorescein measured hemichannel‐mediated dye uptake and ATP luminescence assays quantified ATP release.\nAs shown in Figure 3a,b, high glucose (25 mM) augmented TGFβ1‐evoked dye uptake by 28.1% (compared high glucose alone; p < 0.05). The combined effect of glucose and cytokine was completely negated when cells were co‐incubated with Tonabersat (49.8 ± 4.6% reduction; p < 0.001). The ability of Tonabersat to reduce dye uptake in response to TGFβ1 and high glucose was paralleled by a 43 ± 5.9% decrease in ATP release (Figure 3c; p < 0.05).\nTonabersat negates TGFβ1/glucose‐evoked hemichannel mediated ATP release in (TK173) renal fibroblasts. Cells were treated with TGFβ1 (10 ng/ml) in low or high glucose ± Tonabersat (Tb; 10 μM) for 48 h. Panel (a) shows representative images for carboxyfluorescein dye uptake in each condition. Mean data (± SEM, n = 4–6) demonstrated that TGFβ1 increased dye uptake, an effect that was amplified when co‐incubated at high glucose (panel (b)). Examining ATP release, this high glucose TGFβ1‐induced response was reversed to near basal (5 mM glucose) levels when cells were pre‐incubated with Tonabersat (panel (c)). Significances are represented as *p < 0.05, **p < 0.01, ***p < 0.001.\n[SUBTITLE] Tonabersat inhibits TGFβ1/glucose‐induced changes in Cx43, N‐cadherin and ECM localisation and expression in renal fibroblasts [SUBSECTION] Having ascertained that TGFβ1 stimulates Cx43 hemichannel activity in renal fibroblasts, an effect augmented in the presence of high glucose, we investigated if blocking this activity could confer any phenotypic protection. Immunocytochemistry (Figure 4a) and western blotting (Figure 4b–f) were used to determine TGFβ1‐induced changes in protein localisation and expression at low (5 mM) and high (25 mM) glucose, before assessing the ability of Tonabersat to dampen the combined effect of glycaemic and cytokine challenge in TK173 fibroblasts.\nTonabersat reverses TGFβ1/glucose‐evoked changes in ECM and adherens protein expression and localisation in (TK173) renal fibroblasts. In panel (a) immunocytochemistry revealed the effect of the pro‐fibrotic cytokine TGFβ1 (10 ng/ml) on the localisation of Cx43, N‐cadherin, vimentin and fibronectin in low (5 mM) and high (25 mM) glucose ± Tonabersat (Tb; 10 μM) in TK173 fibroblasts. Western blot examined whole cell protein expression (%) of Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f). Representative blots and mean data ± SEM (n = 4–6) are shown, where *p < 0.05, **p < 0.01, ***p < 0.001.\nThe pro‐fibrotic cytokine TGFβ1 had negligible effects on localisation of candidate proteins, but increased expression in both low (5mM) and high (25mM) glucose as determined by immunocytochemistry. This effect was particularly evident with ECM proteins fibronectin and vimentin (Figure 4a). As an intermediate filament protein, vimentin was used in place of collagen I which was poorly resolved using immunocytochemistry. Tonabersat reversed the combined effects of high glucose and cytokine. Semi‐quantification of protein changes using western blot determined that in the presence of high glucose, TGFβ1 increased expression of Cx43 (Figure 4b 66.1%, p < 0.001), β‐catenin (Figure 4c 32.7%, p < 0.05), N‐cadherin (Figure 4d 45%, p < 0.05), fibronectin (Figure 4e 47.6%, p < 0.01) and collagen I (Figure 4f 26.4%, p=NS). Tonabersat inhibited these effects for Cx43 (54.3 ± 12.6%, p < 0.01), N‐cadherin (40.9 ± 11.8%, p < 0.05), fibronectin (34.3 ± 4.2%, p < 0.05) and collagen I (30.8 ± 3%, p=NS).\nHaving ascertained that TGFβ1 stimulates Cx43 hemichannel activity in renal fibroblasts, an effect augmented in the presence of high glucose, we investigated if blocking this activity could confer any phenotypic protection. Immunocytochemistry (Figure 4a) and western blotting (Figure 4b–f) were used to determine TGFβ1‐induced changes in protein localisation and expression at low (5 mM) and high (25 mM) glucose, before assessing the ability of Tonabersat to dampen the combined effect of glycaemic and cytokine challenge in TK173 fibroblasts.\nTonabersat reverses TGFβ1/glucose‐evoked changes in ECM and adherens protein expression and localisation in (TK173) renal fibroblasts. In panel (a) immunocytochemistry revealed the effect of the pro‐fibrotic cytokine TGFβ1 (10 ng/ml) on the localisation of Cx43, N‐cadherin, vimentin and fibronectin in low (5 mM) and high (25 mM) glucose ± Tonabersat (Tb; 10 μM) in TK173 fibroblasts. Western blot examined whole cell protein expression (%) of Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f). Representative blots and mean data ± SEM (n = 4–6) are shown, where *p < 0.05, **p < 0.01, ***p < 0.001.\nThe pro‐fibrotic cytokine TGFβ1 had negligible effects on localisation of candidate proteins, but increased expression in both low (5mM) and high (25mM) glucose as determined by immunocytochemistry. This effect was particularly evident with ECM proteins fibronectin and vimentin (Figure 4a). As an intermediate filament protein, vimentin was used in place of collagen I which was poorly resolved using immunocytochemistry. Tonabersat reversed the combined effects of high glucose and cytokine. Semi‐quantification of protein changes using western blot determined that in the presence of high glucose, TGFβ1 increased expression of Cx43 (Figure 4b 66.1%, p < 0.001), β‐catenin (Figure 4c 32.7%, p < 0.05), N‐cadherin (Figure 4d 45%, p < 0.05), fibronectin (Figure 4e 47.6%, p < 0.01) and collagen I (Figure 4f 26.4%, p=NS). Tonabersat inhibited these effects for Cx43 (54.3 ± 12.6%, p < 0.01), N‐cadherin (40.9 ± 11.8%, p < 0.05), fibronectin (34.3 ± 4.2%, p < 0.05) and collagen I (30.8 ± 3%, p=NS).\n[SUBTITLE] \nATP mediates TGFβ1/glucose‐induced changes in Cx43, N‐cadherin and ECM expression in renal fibroblasts [SUBSECTION] Having determined that high glucose in combination with TGFβ1 evokes a Tonabersat‐sensitive increase in ATP release (Figure 3) and increased expression of tubular injury associated proteins (Figure 4), we further explored a downstream role for ATP‐mediated purinergic signalling in driving high glucose and TGFβ1‐induced changes. An ATP‐diphosphohydrolase, apyrase (apy) catalyses the sequential hydrolysis of ATP to ADP and ADP to AMP, releasing inorganic phosphate and reducing the extracellular concentration of ATP ([ATP]e) to decrease activation of P2‐purinergic receptors (Figure 5a). TK173 cells were treated in low or high glucose with TGFβ1 (10 ng/ml) ± apyrase (100 IU/ml) for 48 h. Western blot assessed changes in whole cell protein expression. Compared to high glucose alone, TGFβ1 plus 25 mM glucose increased expression of Cx43 (Figure 5b 66.1%, p < 0.01), β‐catenin (Figure 5c 32.7%, p < 0.05), N‐cadherin (Figure 5d 45%, p < 0.01), fibronectin (Figure 5e 35.2%, p < 0.05) and collagen I (Figure 5f 26.4%, p=NS). These changes decreased by 51.4 ± 14.2% (Cx43; p < 0.05), 34.0 ± 7.0%, (N‐cadherin; p < 0.05), 26.6 ± 1.3%, (fibronectin; p < 0.05), and 47.8 ± 12.3%, (collagen I; p < 0.05), when cells were co‐incubated with apyrase. Hydrolysis of ATP failed to significantly alter TGFβ1/glucose‐induced changes in β‐catenin.\nReducing activation of the P2X7 receptor reverses TGFβ1/glucose‐induced changes in ECM and adherens junction protein expression in (TK173) renal fibroblasts. To reduce activation of the P2X7R, either the concentration of TGFβ1/glucose‐evoked extracellular ATP ([ATP]e) was reduced by apyrase (apy), which catalyses the sequential hydrolysis of ATP to ADP and AMP, or the P2X7 receptor was inhibited using A438079. Both strategies reduced markers of TGFβ1‐dependent inflammation and fibrosis (a). Western blot analysis determined whole cell protein expression (%) of Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f) in response to TGFβ1 (10 ng/ml) in low (5 mM) and high (25 mM) glucose ± apyrase (100 IU/ml) ± the P2X7R inhibitor (P2X7Ri: A438079 (50 μM)). Representative blots are shown above mean data ± SEM, n = 3–5, where *p < 0.05, **p < 0.01, ***p < 0.001.\nKnown to mediate inflammation and downstream fibrosis, activation of the P2X7 receptor (P2X7R) has been associated with tubular injury in an in vivo model of diabetic nephropathy.\n10\n We used the P2X7R inhibitor A438079 (50 μM) to determine the role of this receptor subtype in glucose/cytokine‐induced changes in TK173 fibroblasts (Figure 5a). The P2X7R inhibitor reversed TGFβ1/high glucose‐induced increases in Cx43 (Figure 5b 48.7 ± 12.7%, p < 0.05), N‐cadherin (Figure 5d 53.1 ± 13.9%, p < 0.01) and collagen I (Figure 5f 62.6 ± 2.7%, P < 0.001), and marginally reduced expression of β‐catenin (Figure 5c 21.8 ± 14.2%) and fibronectin (Figure 5 e 26.3 ± 1.9%).\nHaving determined that high glucose in combination with TGFβ1 evokes a Tonabersat‐sensitive increase in ATP release (Figure 3) and increased expression of tubular injury associated proteins (Figure 4), we further explored a downstream role for ATP‐mediated purinergic signalling in driving high glucose and TGFβ1‐induced changes. An ATP‐diphosphohydrolase, apyrase (apy) catalyses the sequential hydrolysis of ATP to ADP and ADP to AMP, releasing inorganic phosphate and reducing the extracellular concentration of ATP ([ATP]e) to decrease activation of P2‐purinergic receptors (Figure 5a). TK173 cells were treated in low or high glucose with TGFβ1 (10 ng/ml) ± apyrase (100 IU/ml) for 48 h. Western blot assessed changes in whole cell protein expression. Compared to high glucose alone, TGFβ1 plus 25 mM glucose increased expression of Cx43 (Figure 5b 66.1%, p < 0.01), β‐catenin (Figure 5c 32.7%, p < 0.05), N‐cadherin (Figure 5d 45%, p < 0.01), fibronectin (Figure 5e 35.2%, p < 0.05) and collagen I (Figure 5f 26.4%, p=NS). These changes decreased by 51.4 ± 14.2% (Cx43; p < 0.05), 34.0 ± 7.0%, (N‐cadherin; p < 0.05), 26.6 ± 1.3%, (fibronectin; p < 0.05), and 47.8 ± 12.3%, (collagen I; p < 0.05), when cells were co‐incubated with apyrase. Hydrolysis of ATP failed to significantly alter TGFβ1/glucose‐induced changes in β‐catenin.\nReducing activation of the P2X7 receptor reverses TGFβ1/glucose‐induced changes in ECM and adherens junction protein expression in (TK173) renal fibroblasts. To reduce activation of the P2X7R, either the concentration of TGFβ1/glucose‐evoked extracellular ATP ([ATP]e) was reduced by apyrase (apy), which catalyses the sequential hydrolysis of ATP to ADP and AMP, or the P2X7 receptor was inhibited using A438079. Both strategies reduced markers of TGFβ1‐dependent inflammation and fibrosis (a). Western blot analysis determined whole cell protein expression (%) of Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f) in response to TGFβ1 (10 ng/ml) in low (5 mM) and high (25 mM) glucose ± apyrase (100 IU/ml) ± the P2X7R inhibitor (P2X7Ri: A438079 (50 μM)). Representative blots are shown above mean data ± SEM, n = 3–5, where *p < 0.05, **p < 0.01, ***p < 0.001.\nKnown to mediate inflammation and downstream fibrosis, activation of the P2X7 receptor (P2X7R) has been associated with tubular injury in an in vivo model of diabetic nephropathy.\n10\n We used the P2X7R inhibitor A438079 (50 μM) to determine the role of this receptor subtype in glucose/cytokine‐induced changes in TK173 fibroblasts (Figure 5a). The P2X7R inhibitor reversed TGFβ1/high glucose‐induced increases in Cx43 (Figure 5b 48.7 ± 12.7%, p < 0.05), N‐cadherin (Figure 5d 53.1 ± 13.9%, p < 0.01) and collagen I (Figure 5f 62.6 ± 2.7%, P < 0.001), and marginally reduced expression of β‐catenin (Figure 5c 21.8 ± 14.2%) and fibronectin (Figure 5 e 26.3 ± 1.9%).\n[SUBTITLE] Tonabersat partially negates epithelial cell secretome‐mediated changes on fibroblast hemichannel number and protein expression [SUBSECTION] While tubular epithelial cells are often regarded as the initial drivers of renal injury, fibroblasts are considered the major ECM producing cell type. Importantly, they are recruited and activated in response to sustained tubular injury. Regulated by soluble ligands, for example, ATP, crosstalk between the tubular epithelia and fibroblasts can orchestrate many forms of disease progression\n24\n including diabetic nephropathy.\n25\n To investigate the role of the cell secretome in paracrine‐mediated signalling between each cell type, conditioned media transfer (indirect co‐culture) was performed between tubular epithelial cells and medullary fibroblasts. In doing so, we determined the downstream effects on hemichannel activity and protein expression on both HK2 cells and TK173 cells when incubated with conditioned media of the alternate cell type (Figure 6a).\nConditioned media from high glucose‐treated proximal tubule epithelial cells (HK2) evoked Tonabersat‐sensitive changes in ECM and adherens junction protein expression in (TK173) renal fibroblasts. Indirect co‐culture utilised 10 times freeze‐thawed 48 h high glucose (25 mM)‐treated conditioned media (CM) ± Tonabersat (Tb; 10 μM) to determine the effect of cell secretome on the heterotypic cell type (a). Carboxyfluorescein studies assessed changes in dye uptake when fibroblast conditioned media were added to tubular epithelial cells (b) and (c) and vice versa (d) and (e). Determined by western blotting, tubular epithelial cell secretome examined Tonabersat‐sensitive changes in fibroblast whole cell protein expression (%) for Cx43 (f), β‐catenin (g), N‐cadherin (h), fibronectin (i) and collagen I (j). Data shown illustrate representative blots and mean data ± SEM (n = 3–4), where *p < 0.05, **p < 0.01, ***p < 0.001.\nAlthough the transfer of high glucose conditioned media from fibroblasts evoked a Tonabersat‐sensitive increase in dye uptake in tubular epithelial cells (55.9%, p < 0.001) as compared to 5 mM glucose control (Figure 6b,c), it had minimal effect on HK2 protein expression (see: Figure S1).\nConditioned media from high glucose‐treated proximal tubule epithelial cells (HK2) evoked a Tonabersat‐sensitive increase in dye uptake in (TK173) renal fibroblasts of 54.5% (p < 0.001), compared to low glucose control (Figure 6d,e). Contrary to the lack of effect that fibroblast (TK173) conditioned media had on tubule epithelial (HK2) phenotype, the effects of HK2 conditioned media on TK173 dye uptake, were paralleled by changes in Cx43 expression and markers of the ECM and adherens junction. Western blot determined that high glucose conditioned media from epithelial cells increased expression of Cx43 (47.7%, Figure 6f; p < 0.001), N‐cadherin (24.1%, Figure 6h; p < 0.05) and fibronectin (60.5%, Figure 6i; p < 0.01) in fibroblasts compared to unconditioned 25 mM glucose media. Tonabersat negated this effect by 33.9 ± 11.4%, p < 0.01 (Cx43), 25.3 ± 8.9%, p < 0.05 (N‐cadherin) and 53.4 ± 7.7%, p < 0.05 (fibronectin). There were minimal effects on β‐catenin (Figure 6g) or collagen I (Figure 6j).\nWhile tubular epithelial cells are often regarded as the initial drivers of renal injury, fibroblasts are considered the major ECM producing cell type. Importantly, they are recruited and activated in response to sustained tubular injury. Regulated by soluble ligands, for example, ATP, crosstalk between the tubular epithelia and fibroblasts can orchestrate many forms of disease progression\n24\n including diabetic nephropathy.\n25\n To investigate the role of the cell secretome in paracrine‐mediated signalling between each cell type, conditioned media transfer (indirect co‐culture) was performed between tubular epithelial cells and medullary fibroblasts. In doing so, we determined the downstream effects on hemichannel activity and protein expression on both HK2 cells and TK173 cells when incubated with conditioned media of the alternate cell type (Figure 6a).\nConditioned media from high glucose‐treated proximal tubule epithelial cells (HK2) evoked Tonabersat‐sensitive changes in ECM and adherens junction protein expression in (TK173) renal fibroblasts. Indirect co‐culture utilised 10 times freeze‐thawed 48 h high glucose (25 mM)‐treated conditioned media (CM) ± Tonabersat (Tb; 10 μM) to determine the effect of cell secretome on the heterotypic cell type (a). Carboxyfluorescein studies assessed changes in dye uptake when fibroblast conditioned media were added to tubular epithelial cells (b) and (c) and vice versa (d) and (e). Determined by western blotting, tubular epithelial cell secretome examined Tonabersat‐sensitive changes in fibroblast whole cell protein expression (%) for Cx43 (f), β‐catenin (g), N‐cadherin (h), fibronectin (i) and collagen I (j). Data shown illustrate representative blots and mean data ± SEM (n = 3–4), where *p < 0.05, **p < 0.01, ***p < 0.001.\nAlthough the transfer of high glucose conditioned media from fibroblasts evoked a Tonabersat‐sensitive increase in dye uptake in tubular epithelial cells (55.9%, p < 0.001) as compared to 5 mM glucose control (Figure 6b,c), it had minimal effect on HK2 protein expression (see: Figure S1).\nConditioned media from high glucose‐treated proximal tubule epithelial cells (HK2) evoked a Tonabersat‐sensitive increase in dye uptake in (TK173) renal fibroblasts of 54.5% (p < 0.001), compared to low glucose control (Figure 6d,e). Contrary to the lack of effect that fibroblast (TK173) conditioned media had on tubule epithelial (HK2) phenotype, the effects of HK2 conditioned media on TK173 dye uptake, were paralleled by changes in Cx43 expression and markers of the ECM and adherens junction. Western blot determined that high glucose conditioned media from epithelial cells increased expression of Cx43 (47.7%, Figure 6f; p < 0.001), N‐cadherin (24.1%, Figure 6h; p < 0.05) and fibronectin (60.5%, Figure 6i; p < 0.01) in fibroblasts compared to unconditioned 25 mM glucose media. Tonabersat negated this effect by 33.9 ± 11.4%, p < 0.01 (Cx43), 25.3 ± 8.9%, p < 0.05 (N‐cadherin) and 53.4 ± 7.7%, p < 0.05 (fibronectin). There were minimal effects on β‐catenin (Figure 6g) or collagen I (Figure 6j).", "Previous studies link increased Cx43 expression to inflammation and fibrosis in a model of renal disease,\n6\n, \n20\n while findings from our laboratory report that aberrant Cx43 hemichannel‐mediated ATP release from TGFβ1‐treated tubular epithelial cells evoked phenotypic and functional changes, characteristic of tubular damage.\n12\n, \n21\n, \n22\n Consequently, the current study determined if this effect was accentuated in high glucose and if the response could be decreased using Tonabersat, an efficient Cx43 hemichannel blocker. Human kidney (HK2) cells were cultured at low (5 mM) and high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) at high glucose for 48 h. Glucose (25 mM) increased dye uptake (25.3 ± 8.8%, p < 0.05), compared to low glucose (5 mM) control (Figure 1b,c). The response was amplified by 16.4% in the presence of TGFβ1 (p < 0.001). The combined effect of high glucose and TGFβ1 was reduced by 42.3% ± 5.3% when cells were preincubated with Tonabersat (Figure 1c; p < 0.001). In Figure 1d, TGFβ1 increased ATP release at low (32.3 ± 10.2%; p < 0.05) and high (55.7%, p < 0.001) glucose, with the response at 25 mM glucose significantly decreased by Tonabersat (36.9 ± 12.3%, p < 0.05).\nTonabersat negates TGFβ1/glucose‐evoked increases in hemichannel activity in (HK2) proximal tubule epithelial cells. Cells were cultured for 48 h in either low (5 mM) or high (25 mM) glucose ± TGFβ1 (10 ng/mL) ± Tonabersat (Tb; 100 μM). Panel (a) outlines the protocol used to measure Cx43 hemichannel‐mediated carboxyfluorescein (CBF) dye uptake (BSS is balanced salt solution), while panel (b) provides representative images for dye uptake in each condition. Carboxyfluorescein studies (c) assessed changes in hemichannel mediate, and an ATPlite assay used luminescence as a direct measure of ATP release (d). Data represents mean ± SEM, n = 4–6, where *p < 0.05, **p < 0.01, ***p < 0.001.", "We previously reported that UUO causes disassembly of the adherens junction complex\n12\n and that this is accompanied by increased interstitial fibrosis.\n6\n In the absence of glycaemic injury, this effect was partly restored in the Cx43+/− mouse model of UUO. Consequently, we evaluated the effect of TGFβ1 on Cx43, adherens junction (N‐cadherin and β‐catenin) and ECM (fibronectin and collagen I) protein expression in high glucose (Figure 2). Human kidney (HK2) cells were treated for 48 h in either low (5 mM) or high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (100 μM) to block ATP release (Figure 2a). Compared to low glucose control, the cytokine increased expression of total Cx43 by 76.4 ± 10.2% (Figure 2b; p < 0.001), N‐cadherin by 39.8 ± 7.8% (Figure 2d; p < 0.01), fibronectin by 52.1 ± 6.7% (Figure 2 e; p < 0.001) and collagen I by 28.8 ± 8.6% (Figure 2f; p < 0.05). This effect was amplified in the presence of high (25 mM) glucose, where TGFβ1 increased N‐cadherin (51.3%, p < 0.001), fibronectin (67.7%, p < 0.001) and collagen I (88.5%, p < 0.001) expression. The effect of TGFβ1 on Cx43 was not accentuated under high (25 mM) glucose conditions (64.4%, p < 0.001), and there were no effects of glucose or TGFβ1 on β‐catenin expression (Figure 2c).\nTonabersat inhibits TGFβ1/glucose‐evoked increases in Cx43, adherens junction and ECM protein expression. HK2 cells were cultured for 48 h in either low (5 mM) or high (25 mM) glucose ± TGFβ1 (10 ng/mL) ± Tonabersat (Tb; 100 μM). Tonabersat blocks Cx43 hemichannel‐mediated ATP release (a). Western blot examined whole cell protein expression (%) for Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f). Data shown illustrate a representative blot and mean data ± SEM (n = 4–6), where *p < 0.05, **p < 0.01, ***p < 0.001.\nHaving determined that Tonabersat blocks TGFβ1‐induced changes in hemichannel activity (Figure 1), we assessed if TGFβ1‐evoked changes in protein expression under conditions of high glucose were hemichannel mediated. As shown in Figure 2, Tonabersat decreased high glucose and TGFβ1‐induced increases in Cx43 expression by 46 ± 10.3% (p < 0.01), while fibronectin and collagen I expression were decreased by 35.7 ± 4.8%, (p < 0.01) and 40.6 ± 5.4%, (p < 0.001), respectively. There was no effect on N‐cadherin.", "Tubulointerstitial fibrosis (TIF) is the key underlying pathology of diabetic kidney disease and develops in response to activation of multiple cell types in and around the proximal tubules.\n23\n Medullary fibroblasts contribute to the onset and progression of TIF in diabetic nephropathy. With previous studies linking the heterozygous Cx43+/− UUO mouse to decreased fibroblast activation in the kidney cortex,\n6\n we wanted to assess (i) if renal fibroblasts (TK173) have functional hemichannels and (ii) if Tonabersat could block effects of glucose and TGFβ1 on Cx43 hemichannel‐mediated release of ATP. Renal fibroblasts were treated in low (5 mM) and high (25 mM) glucose with TGFβ1 (10 ng/ml) ± Tonabersat (10 μM) at high glucose for 48 h. Carboxyfluorescein measured hemichannel‐mediated dye uptake and ATP luminescence assays quantified ATP release.\nAs shown in Figure 3a,b, high glucose (25 mM) augmented TGFβ1‐evoked dye uptake by 28.1% (compared high glucose alone; p < 0.05). The combined effect of glucose and cytokine was completely negated when cells were co‐incubated with Tonabersat (49.8 ± 4.6% reduction; p < 0.001). The ability of Tonabersat to reduce dye uptake in response to TGFβ1 and high glucose was paralleled by a 43 ± 5.9% decrease in ATP release (Figure 3c; p < 0.05).\nTonabersat negates TGFβ1/glucose‐evoked hemichannel mediated ATP release in (TK173) renal fibroblasts. Cells were treated with TGFβ1 (10 ng/ml) in low or high glucose ± Tonabersat (Tb; 10 μM) for 48 h. Panel (a) shows representative images for carboxyfluorescein dye uptake in each condition. Mean data (± SEM, n = 4–6) demonstrated that TGFβ1 increased dye uptake, an effect that was amplified when co‐incubated at high glucose (panel (b)). Examining ATP release, this high glucose TGFβ1‐induced response was reversed to near basal (5 mM glucose) levels when cells were pre‐incubated with Tonabersat (panel (c)). Significances are represented as *p < 0.05, **p < 0.01, ***p < 0.001.", "Having ascertained that TGFβ1 stimulates Cx43 hemichannel activity in renal fibroblasts, an effect augmented in the presence of high glucose, we investigated if blocking this activity could confer any phenotypic protection. Immunocytochemistry (Figure 4a) and western blotting (Figure 4b–f) were used to determine TGFβ1‐induced changes in protein localisation and expression at low (5 mM) and high (25 mM) glucose, before assessing the ability of Tonabersat to dampen the combined effect of glycaemic and cytokine challenge in TK173 fibroblasts.\nTonabersat reverses TGFβ1/glucose‐evoked changes in ECM and adherens protein expression and localisation in (TK173) renal fibroblasts. In panel (a) immunocytochemistry revealed the effect of the pro‐fibrotic cytokine TGFβ1 (10 ng/ml) on the localisation of Cx43, N‐cadherin, vimentin and fibronectin in low (5 mM) and high (25 mM) glucose ± Tonabersat (Tb; 10 μM) in TK173 fibroblasts. Western blot examined whole cell protein expression (%) of Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f). Representative blots and mean data ± SEM (n = 4–6) are shown, where *p < 0.05, **p < 0.01, ***p < 0.001.\nThe pro‐fibrotic cytokine TGFβ1 had negligible effects on localisation of candidate proteins, but increased expression in both low (5mM) and high (25mM) glucose as determined by immunocytochemistry. This effect was particularly evident with ECM proteins fibronectin and vimentin (Figure 4a). As an intermediate filament protein, vimentin was used in place of collagen I which was poorly resolved using immunocytochemistry. Tonabersat reversed the combined effects of high glucose and cytokine. Semi‐quantification of protein changes using western blot determined that in the presence of high glucose, TGFβ1 increased expression of Cx43 (Figure 4b 66.1%, p < 0.001), β‐catenin (Figure 4c 32.7%, p < 0.05), N‐cadherin (Figure 4d 45%, p < 0.05), fibronectin (Figure 4e 47.6%, p < 0.01) and collagen I (Figure 4f 26.4%, p=NS). Tonabersat inhibited these effects for Cx43 (54.3 ± 12.6%, p < 0.01), N‐cadherin (40.9 ± 11.8%, p < 0.05), fibronectin (34.3 ± 4.2%, p < 0.05) and collagen I (30.8 ± 3%, p=NS).", "Having determined that high glucose in combination with TGFβ1 evokes a Tonabersat‐sensitive increase in ATP release (Figure 3) and increased expression of tubular injury associated proteins (Figure 4), we further explored a downstream role for ATP‐mediated purinergic signalling in driving high glucose and TGFβ1‐induced changes. An ATP‐diphosphohydrolase, apyrase (apy) catalyses the sequential hydrolysis of ATP to ADP and ADP to AMP, releasing inorganic phosphate and reducing the extracellular concentration of ATP ([ATP]e) to decrease activation of P2‐purinergic receptors (Figure 5a). TK173 cells were treated in low or high glucose with TGFβ1 (10 ng/ml) ± apyrase (100 IU/ml) for 48 h. Western blot assessed changes in whole cell protein expression. Compared to high glucose alone, TGFβ1 plus 25 mM glucose increased expression of Cx43 (Figure 5b 66.1%, p < 0.01), β‐catenin (Figure 5c 32.7%, p < 0.05), N‐cadherin (Figure 5d 45%, p < 0.01), fibronectin (Figure 5e 35.2%, p < 0.05) and collagen I (Figure 5f 26.4%, p=NS). These changes decreased by 51.4 ± 14.2% (Cx43; p < 0.05), 34.0 ± 7.0%, (N‐cadherin; p < 0.05), 26.6 ± 1.3%, (fibronectin; p < 0.05), and 47.8 ± 12.3%, (collagen I; p < 0.05), when cells were co‐incubated with apyrase. Hydrolysis of ATP failed to significantly alter TGFβ1/glucose‐induced changes in β‐catenin.\nReducing activation of the P2X7 receptor reverses TGFβ1/glucose‐induced changes in ECM and adherens junction protein expression in (TK173) renal fibroblasts. To reduce activation of the P2X7R, either the concentration of TGFβ1/glucose‐evoked extracellular ATP ([ATP]e) was reduced by apyrase (apy), which catalyses the sequential hydrolysis of ATP to ADP and AMP, or the P2X7 receptor was inhibited using A438079. Both strategies reduced markers of TGFβ1‐dependent inflammation and fibrosis (a). Western blot analysis determined whole cell protein expression (%) of Cx43 (b), β‐catenin (c), N‐cadherin (d), fibronectin (e) and collagen I (f) in response to TGFβ1 (10 ng/ml) in low (5 mM) and high (25 mM) glucose ± apyrase (100 IU/ml) ± the P2X7R inhibitor (P2X7Ri: A438079 (50 μM)). Representative blots are shown above mean data ± SEM, n = 3–5, where *p < 0.05, **p < 0.01, ***p < 0.001.\nKnown to mediate inflammation and downstream fibrosis, activation of the P2X7 receptor (P2X7R) has been associated with tubular injury in an in vivo model of diabetic nephropathy.\n10\n We used the P2X7R inhibitor A438079 (50 μM) to determine the role of this receptor subtype in glucose/cytokine‐induced changes in TK173 fibroblasts (Figure 5a). The P2X7R inhibitor reversed TGFβ1/high glucose‐induced increases in Cx43 (Figure 5b 48.7 ± 12.7%, p < 0.05), N‐cadherin (Figure 5d 53.1 ± 13.9%, p < 0.01) and collagen I (Figure 5f 62.6 ± 2.7%, P < 0.001), and marginally reduced expression of β‐catenin (Figure 5c 21.8 ± 14.2%) and fibronectin (Figure 5 e 26.3 ± 1.9%).", "While tubular epithelial cells are often regarded as the initial drivers of renal injury, fibroblasts are considered the major ECM producing cell type. Importantly, they are recruited and activated in response to sustained tubular injury. Regulated by soluble ligands, for example, ATP, crosstalk between the tubular epithelia and fibroblasts can orchestrate many forms of disease progression\n24\n including diabetic nephropathy.\n25\n To investigate the role of the cell secretome in paracrine‐mediated signalling between each cell type, conditioned media transfer (indirect co‐culture) was performed between tubular epithelial cells and medullary fibroblasts. In doing so, we determined the downstream effects on hemichannel activity and protein expression on both HK2 cells and TK173 cells when incubated with conditioned media of the alternate cell type (Figure 6a).\nConditioned media from high glucose‐treated proximal tubule epithelial cells (HK2) evoked Tonabersat‐sensitive changes in ECM and adherens junction protein expression in (TK173) renal fibroblasts. Indirect co‐culture utilised 10 times freeze‐thawed 48 h high glucose (25 mM)‐treated conditioned media (CM) ± Tonabersat (Tb; 10 μM) to determine the effect of cell secretome on the heterotypic cell type (a). Carboxyfluorescein studies assessed changes in dye uptake when fibroblast conditioned media were added to tubular epithelial cells (b) and (c) and vice versa (d) and (e). Determined by western blotting, tubular epithelial cell secretome examined Tonabersat‐sensitive changes in fibroblast whole cell protein expression (%) for Cx43 (f), β‐catenin (g), N‐cadherin (h), fibronectin (i) and collagen I (j). Data shown illustrate representative blots and mean data ± SEM (n = 3–4), where *p < 0.05, **p < 0.01, ***p < 0.001.\nAlthough the transfer of high glucose conditioned media from fibroblasts evoked a Tonabersat‐sensitive increase in dye uptake in tubular epithelial cells (55.9%, p < 0.001) as compared to 5 mM glucose control (Figure 6b,c), it had minimal effect on HK2 protein expression (see: Figure S1).\nConditioned media from high glucose‐treated proximal tubule epithelial cells (HK2) evoked a Tonabersat‐sensitive increase in dye uptake in (TK173) renal fibroblasts of 54.5% (p < 0.001), compared to low glucose control (Figure 6d,e). Contrary to the lack of effect that fibroblast (TK173) conditioned media had on tubule epithelial (HK2) phenotype, the effects of HK2 conditioned media on TK173 dye uptake, were paralleled by changes in Cx43 expression and markers of the ECM and adherens junction. Western blot determined that high glucose conditioned media from epithelial cells increased expression of Cx43 (47.7%, Figure 6f; p < 0.001), N‐cadherin (24.1%, Figure 6h; p < 0.05) and fibronectin (60.5%, Figure 6i; p < 0.01) in fibroblasts compared to unconditioned 25 mM glucose media. Tonabersat negated this effect by 33.9 ± 11.4%, p < 0.01 (Cx43), 25.3 ± 8.9%, p < 0.05 (N‐cadherin) and 53.4 ± 7.7%, p < 0.05 (fibronectin). There were minimal effects on β‐catenin (Figure 6g) or collagen I (Figure 6j).", "The current study demonstrates that the pro‐fibrotic cytokine TGFβ1 increases Cx43 hemichannel‐mediated ATP release in TK173 renal fibroblasts and HK2 tubular epithelial cells, events which lead to autocrine and paracrine effects and were amplified by high glucose. Local increases in ATP independently initiate P2X7R‐mediated changes in adherens junction and ECM protein expression on both resident epithelial cells and fibroblasts (see Figure 7). Our data support earlier studies under euglycaemic conditions demonstrating that TGFβ1 evokes Cx43‐mediated changes in expression of markers of tubular injury and fibrosis in human primary proximal tubule epithelial cells (hPTECs)\n7\n, \n12\n, \n21\n, \n22\n and further suggests that effects of the cytokine are compounded by high glucose. The importance of P2X7R in diabetic kidney disease was reported by Menzies et al., who showed that deficiency of P2X7R reduced macrophage infiltration and collagen IV deposition in a Type 1 mouse model of diabetic nephropathy.\n10\n Poorly expressed in healthy kidney tissue,\n26\n expression of P2X7R is markedly increased in renal biopsy material from people with diabetic nephropathy.\n10\n, \n12\n Pharmacologically blocking Cx43‐mediated ATP release using Tonabersat and reducing markers of tubular injury in the present study corroborates data from our normoglycaemic Cx43+/− UUO model\n12\n while demonstrating that this protection is still apparent under high glucose conditions associated with DN and likely involves Cx43 hemichannel‐mediated ATP release as an initiating trigger.\nCell–cell interactions between tubular epithelial cells and fibroblasts. Glucose and transforming growth factor beta1 (TGFβ1) increase Cx43 hemichannel‐mediated ATP release from both human proximal tubule epithelial cells (hPTECs) and fibroblasts. Local increases in extracellular ATP stimulate P2X7R on both cell types driving an altered cell phenotype of tubule cells through partial epithelial‐to‐mesenchymal transition and myofibroblast activation. Co‐culture experiments determine that an ATP‐P2X7R driven response in hPTECs triggers secretion of cytokines, for example, TGFβ1, which through paracrine mediated signalling activates neighbouring fibroblasts to further increase fibroblast‐derived deposition of extracellular matrix (ECM) and exacerbate increased fibrosis.\nLacking direct cell–cell contact, heterotypic cell‐to‐cell crosstalk between the tubular epithelia and fibroblasts is regulated by soluble mediators (e.g., cytokines) and has been associated with various forms of disease progression.\n24\n, \n27\n Although tubular epithelial cells are often perceived as the instigators of renal injury, inflammation, and downstream fibrosis in and around the tubules involves multiple cell types. Moreover, a 50% knockdown of Cx43 in the heterozygous Cx43+/− mouse model of unilateral ureteral obstruction, has been shown to decrease interstitial fibrosis (sirius red staining) and fibroblast accumulation (fibroblast specific protein‐1 [FSP‐1] staining).\n6\n Mediated by Tonabersat‐sensitive Cx43 hemichannel ATP release, indirect co‐culture studies utilising conditioned media from the alternative cell type demonstrate the degree of interdependence between tubular epithelial cells and fibroblasts. Driven by epithelial cell‐mediated release of cytokines and chemokines, for example, TGFβ1, we found that the association between tubular epithelial cells and TK173 medullary fibroblasts favoured epithelial regulation of fibroblast protein expression (Figure 7). This apparent hierarchy of control has previously been reported when describing the pro‐fibrotic effects of the tubular senescence‐associated secretory phenotype (SASP) and activation of fibroblasts by mouse tubular epithelial cells.\n28\n Moreover, studies by Zhou et al., determined that tubule‐derived Wnts have an essential role in promoting fibroblast activation\n27\n while injured tubular epithelial cells have been shown to initiate the activation and proliferation of fibroblasts via direct shuttling of miR‐150‐containing exosomes\n29\n or the miR‐21/PTEN/Akt pathway.\n30\n Building on these observations, data in the current study suggest that blocking Cx43‐mediated ATP release may not only dampen homotypic PTEC‐PTEC, fibroblast–fibroblast activation and ECM remodelling/deposition, but may importantly reduce epithelial–mesenchymal communication, events which predispose a profibrotic environment.\n21\n\n\nIn conclusion, while this in vitro study provides a minimalistic model for the complexities of what is a debilitating complication of diabetes, the model provides an accessible and reliable tool for the in vitro study of cellular mechanisms. This is supported by the fact that our data build upon previously published observations which have informed in vivo work using the UUO mouse model of advanced interstitial inflammation and fibrosis.\n12\n Moreover, although recently reported that Tonabersat has homology with some other alpha group connexin isoforms (e.g., Cx40),\n31\n previously published in vitro\n12\n data using Cx43 hemichannel blocker Peptide 5 and in vivo\n6\n observations using the Cx43+/− heterozygous mouse, strongly support a pathogenic role of Cx43 hemichannels in interstitial inflammation and fibrosis, while highlighting the potential future use of Tonabersat in dampening aberrant Cx43 hemichannel‐mediated purinergic communication in a model of diabetic nephropathy.", "CEH and PES were involved in conceptualisation, supervision and project administration. BMW and CLC were involved in experimental studies. BMW, PES and CEH were involved in writing—original draft preparation. BMW, CLC, ID, PES and CEH were involved in writing. All authors have read and agreed to the published version of the manuscript.", "BMW is a recipient of a PhD studentship supported by Diabetes UK and the Masonic Charitable Foundation. (18/0005919; awarded to PES & CEH).", "The authors confirm that there are no conflict‐of‐interest issues.", "\nFigure S1.\n\nClick here for additional data file." ]
[ null, null, null, null, null, "methods", null, null, null, null, null, null, null, "results", null, null, null, null, null, null, "discussion", null, null, "COI-statement", "supplementary-material" ]
[ "adenosine triphosphate", "connexin 43", "diabetic nephropathy", "epithelial cells", "fibroblasts", "fibrosis", "Tonabersat" ]
Mental and behavioral disorders in the prison context.
36256558
During the execution of sentences in prison units, there are deficiencies within the adaptation mechanisms of persons deprived of liberty, this varies depending on the place where they serve the sanction and the length of stay in it.
INTRODUCTION
Theoretical review. Primary sources of information: scientific articles from indexed journals, in specialized medical information search engines. Original publications on mental and behavioural disorders in the prison and prison context were included, from 2016 to April 2021, with an analytical, observational, prospective, retrospective, cross-sectional and randomized design, systematic reviews and meta-analyzes, complete articles, carried out in any country, with subjects over 15 years of age, of any sex or gender, in English and Spanish.
MATERIAL AND METHOD
16 articles were included in this theoretical review. The prevalence of mental disorders within the prison population is high; depression, anxiety, substance use and psychotic disorders predominate. The importance of having mental health programmes in prisons, with initial diagnosis and personalized interventions, was observed. The authors recommend psychopharmacological interventions and cognitive behavioural management.
RESULTS
The need to restructure the mental health approach in prisons was evidenced, and early diagnosis and personalized follow-up should be guaranteed. Pharmacological intervention and, to a greater degree, cognitive behavioural therapy seem to be effective for these types of patients.
DISCUSSION
[ "Humans", "Cross-Sectional Studies", "Mental Disorders", "Prisoners", "Prisons", "Prospective Studies", "Retrospective Studies", "Male", "Female", "Adult" ]
9578298
Introduction
The World Health Organisation states that problems related to mental health are up to seven times more likely to appear in the prison population than amongst the general public in Western societies. This upward trend in mental disorders coincides with a growing prison population. Another aggravating factor is substance use in prisons1. The behavioural changes that lead to mental disorders and prison violence often take place in contexts of inhumane confinement conditions that are common in Latin-American prisons. Inmates undergo a process of adaptation that can include injury and aggression, and in some cases, overcrowding, hunger and disease during imprisonment2. It has been shown that there are failures in inmates’ adaptation mechanisms during the confinement process, which vary according to there they are imprisoned and the length of sentence. There is a reduction in the individual’s strategies that enable them to act in reaction to being imprisoned; there is also a loss of ideas, actions and feelings that gradually affect an individual’s relationship with their surroundings3. Although changes are being made to improve the treatment of inmates in the prison system of several Latin American countries, there are prisons where the care of patients with previous psychiatric disorders, such as schizophrenia, is in the hand of general physicians. The profile for this group of patients is a complex one, and poor treatment adherence and drug consumption are very common4. The presence of psychiatric disorders in this context is a reality, as is the link between pre-existing mental disorders and the frequent exacerbation of symptoms and imbalances. It is therefore important to carry out this theoretical review, to highlight the need to identify and offer timely treatment for mental disorders in the prison population, with the active participation of the psychiatrist as a specialist in this field. We also seek to identify the possible causes behind the appearance of mental disorders. In this case it is essential to consider the emotional changes brought about by imprisonment and the inability to adapt to incarceration. There are few studies in Ecuador on this issue, which makes this review article even more relevant, as it will be of benefit not only to health professionals but to all those who work directly with prison inmates. This theoretical review was carried out to compile information about the prevalence of mental and behavioural disorders in the prison setting, and also to highlight the importance of early psychiatric intervention. The recommendations for managing this group in situations of confinement, with a focus on the role of the psychiatrist as a mental health professional, are also described. The results of this review may help psychiatric practice in prisons, since they offer a compendium of up to date information about prevalence, risk factors and therapeutic approaches. The results of this research also highlight the importance of restructuring mental health care for inmates, since psychiatric evaluation is often late and when it is adequately carried out, ongoing monitoring is infrequent, despite being regulated under the law. Another factor is the constant turnover in staff working at these institutions, given that there is a rotation system for professionals, who offer their services on a transient basis, while many professionals do not have experience in managing patients with mental disorders.
null
null
Resultats
A total of 29 articles on mental disorders amongst prison inmates were identified. They were published between January 2016 and April 2021. 3 of them were removed since they were published in German, 2 for using a qualitative design, 3 journal editorials, 2 articles that were duplicated and 3 for not having a clear and reproducible methodology. 16 articles were left at the end of the selection process, and these were included in the theoretical review (Table 1). Table 1Traceability.Author/yearCountry Journal Browser URL Adraro et al. (2019)EthiopiaBMC Public Health BMC https://bmcpublichealth.biomedcentral.com/articles/10.1186/s12889-019-7879-6 Baranyi et al. (2019)UK Lancet Glob HealthThe Lancet https://www.thelancet.com/journals/langlo/article/PIIS2214-109X(18)30539-4/fulltext Bartlett et al. (2018)UKBr J PsychiatryPubmed https://pubmed.ncbi.nlm.nih.gov/29486822/ Doyle et al. (2019)Australia Aust N Z J Public HealthPubmed https://pubmed.ncbi.nlm.nih.gov/30908856/ Eher et al. (2019) Austria Acta Psychiatr ScandWiley Online Library >https://onlinelibrary.wiley.com/doi/abs/10.1111/acps.13024 Facer et al. (2019) UK PLoS OnePubmed https://pubmed.ncbi.nlm.nih.gov/31557173/ Favril et al. (2020)New Zealand Soc Psychiatry Psychiatr EpidemiolSpringer Link https://link.springer.com/article/10.1007%2Fs00127-020-01851-7 Fazel et al. (2016) UK Lancet PsychiatryPubmed https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5008459/ Forry et al. (2019) Uganda BMC PsychiatryBMC >https://bmcpsychiatry.biomedcentral.com/articles/10.1186/s12888-019-2167-7 Fovet et al. (2020) FranceEur PsychiatryPubmed https://pubmed.ncbi.nlm.nih.gov/32336297/ Gottfried et al. (2017) USAJ Correct Health Care Pubmed https://pubmed.ncbi.nlm.nih.gov/28715985/ Jakobowitz et al. (2017) UK Soc Psychiatry Psychiatr EpidemiolSpringer Link https://link.springer.com/article/10.1007%2Fs00127-016-1313-5 Peters et al. (2017) USA Am J Drug Alcohol AbuseTaylor & Francis Online https://www.tandfonline.com/doi/full/10.1080/00952990.2017.1303838 Van Buitenen et al. (2020) Holland Eur Psychiatry Pubmed https://pubmed.ncbi.nlm.nih.gov/32522312/ Wills (2017)USA Int Rev PsychiatryPubmed https://pubmed.ncbi.nlm.nih.gov/27701919/ Wilton et al. (2017)Canada Psychiatr ServPubmed https://pubmed.ncbi.nlm.nih.gov/28292226/ [SUBTITLE] Prevalence of mental disorders and risk factors [SUBSECTION] Analysis of the reports on the prevalence of mental disorders in the prison population showed a range of figures. In research carried out by Adraro et al.5, a prevalence of 62.7% for common mental disorders such as anxiety, depression and somatic symptom disorder was found in a group of 300 prison inmates, which is considered to be a high figure, in which three out of every five inmates are affected. In another study by Baranyi et al.6, it was found that the prevalence of mental disorders was up to 15.8 times higher amongst inmates than they were in the general public, with a tendency to decrease with time spent in prison. The authors also commented that the inmates were often from a low socio-economic background, belonged to minority groups and had a history of child abuse and substance use, which made them vulnerable to psychiatric disorders. The poor living conditions, physical aggression and psychological abuse often encountered in prison can further aggravate mental disorders. According to Van Buitenen et al.7, the prevalence of schizophrenia was 56.7%; for substance abuse disorder, 43.1%; while up to 56.9% of the 5,247 inmates had other mental disorders, such as impulsiveness, poor social skills and disruptive behaviour. On the other hand, a meta-analysis by Facer et al.8 showed that post-traumatic stress disorder (PTSD) was associated with a number of mental disorders such as depression and anxiety disorders. Inmates with PTSD were also significantly more likely to suffer from substance use disorder. A research project by Jakobowitz et al.9 showed that the prevalence of mental disorders amongst inmates was 8.65% for psychosis; 39.2% for depression; 45.8% for substance abuse; 41.4% for alcohol abuse; 17.8% for personality disorders; 8.1% for anxiety disorders and PTSD; and 5.1% for adaptation disorders. According to Forry et al.10 and Fovet et al.11, the most common mental disorders in the prison population are severe depression (44% and 31.2%, respectively), followed by generalised anxiety disorder (30.9 and 44.4%, respectively). Favril et al.12 describe a high prevalence of mental disorders in the prison population of New Zealand: up to 90.7% of the inmates present at least one mental disorder; substance abuse was present in 87% of the inmates; suicidal ideation was present in 36.4%; and suicide attempts in 55.6%. Another study by Fazel et al.13 mentions that there is an important association between severe depression and psychotic illness (p<0.05), and states that one out of every seven inmates suffers from depression or psychosis. They also mention high rates of coincidence between mental illness and substance use, as well as suicide and self-harming, which are more common amongst inmates than the general public of the same age and sex. According to the authors, prison inmates with mental disorders are disproportionately involved in prison infractions and violent incidents, and are more likely to be accused of breaking prison rules and being injured in fights. Inmates with mental disorders and with a history of violence run an even higher risk in this regard, since psychiatric disorders are associated with violent incidents in prison (Table 2). Table 2Prevalence of mental disorders amongst inmates.Author/year PrevalenceAdraro et al. (2019)Common mental disorders: 62.7% (CI 95%: 65.7-68.5).Baranyi et al. (2019)Psychosis: 6.2%.Severe depression: 3.8%.Alcohol use: 3.8%.Substance use: 5.1%.Eher et al. (2019)Mental disorder: 92.9%.Alcohol abuse: 40%.Paraphilias: 43.3%.Personality disorders: 53.6%.Type B personality disorders: 47.8%.Favril et al. (2020)Substance abuse: 87%.Any mental disorder: 90.7%.PTSD: 23.9%.Panic disorder: 5.6%.Generalised anxiety disorder: 8.7%.Bipolar disorder: 11.1%.Suicidal ideas: 36.4%.Attempted suicide: 55.6%.Forry et al. (2019)Severe depression: 44%.Antisocial personality disorder: 20.5%.Bipolar disorder: 23.5%.Generalised anxiety disorder: 30.9%.Panic disorder: 32.6%.Substance abuse: 12.5%.Fovet et al. (2020)Affective disorders: 31.2%.Anxiety disorders: 44.4%.Panic disorders: 13%.Substance abuse: 53.5%.Risk of suicide: 31.4%.Jakobowitz et al. (2017)Psychosis: 8.65%.Depression: 39.2%.Substance abuse: 45.8%.Alcohol abuse: 41.4%.Personality disorder: 17.8%.Anxiety: 8.1%.PTSD: 8.1%.Adaptation disorder: 5.1%.Van Buitenen et al. (2020)Schizophrenia: 56.7%. Substance use: 43.1%.Co-morbid disorders (impulsiveness. poor social skills, disruptive behaviour): 56.9%.Note. CI: confidence interval; PTSD: post-traumatic stress disorder. Note. CI: confidence interval; PTSD: post-traumatic stress disorder. Analysis of the reports on the prevalence of mental disorders in the prison population showed a range of figures. In research carried out by Adraro et al.5, a prevalence of 62.7% for common mental disorders such as anxiety, depression and somatic symptom disorder was found in a group of 300 prison inmates, which is considered to be a high figure, in which three out of every five inmates are affected. In another study by Baranyi et al.6, it was found that the prevalence of mental disorders was up to 15.8 times higher amongst inmates than they were in the general public, with a tendency to decrease with time spent in prison. The authors also commented that the inmates were often from a low socio-economic background, belonged to minority groups and had a history of child abuse and substance use, which made them vulnerable to psychiatric disorders. The poor living conditions, physical aggression and psychological abuse often encountered in prison can further aggravate mental disorders. According to Van Buitenen et al.7, the prevalence of schizophrenia was 56.7%; for substance abuse disorder, 43.1%; while up to 56.9% of the 5,247 inmates had other mental disorders, such as impulsiveness, poor social skills and disruptive behaviour. On the other hand, a meta-analysis by Facer et al.8 showed that post-traumatic stress disorder (PTSD) was associated with a number of mental disorders such as depression and anxiety disorders. Inmates with PTSD were also significantly more likely to suffer from substance use disorder. A research project by Jakobowitz et al.9 showed that the prevalence of mental disorders amongst inmates was 8.65% for psychosis; 39.2% for depression; 45.8% for substance abuse; 41.4% for alcohol abuse; 17.8% for personality disorders; 8.1% for anxiety disorders and PTSD; and 5.1% for adaptation disorders. According to Forry et al.10 and Fovet et al.11, the most common mental disorders in the prison population are severe depression (44% and 31.2%, respectively), followed by generalised anxiety disorder (30.9 and 44.4%, respectively). Favril et al.12 describe a high prevalence of mental disorders in the prison population of New Zealand: up to 90.7% of the inmates present at least one mental disorder; substance abuse was present in 87% of the inmates; suicidal ideation was present in 36.4%; and suicide attempts in 55.6%. Another study by Fazel et al.13 mentions that there is an important association between severe depression and psychotic illness (p<0.05), and states that one out of every seven inmates suffers from depression or psychosis. They also mention high rates of coincidence between mental illness and substance use, as well as suicide and self-harming, which are more common amongst inmates than the general public of the same age and sex. According to the authors, prison inmates with mental disorders are disproportionately involved in prison infractions and violent incidents, and are more likely to be accused of breaking prison rules and being injured in fights. Inmates with mental disorders and with a history of violence run an even higher risk in this regard, since psychiatric disorders are associated with violent incidents in prison (Table 2). Table 2Prevalence of mental disorders amongst inmates.Author/year PrevalenceAdraro et al. (2019)Common mental disorders: 62.7% (CI 95%: 65.7-68.5).Baranyi et al. (2019)Psychosis: 6.2%.Severe depression: 3.8%.Alcohol use: 3.8%.Substance use: 5.1%.Eher et al. (2019)Mental disorder: 92.9%.Alcohol abuse: 40%.Paraphilias: 43.3%.Personality disorders: 53.6%.Type B personality disorders: 47.8%.Favril et al. (2020)Substance abuse: 87%.Any mental disorder: 90.7%.PTSD: 23.9%.Panic disorder: 5.6%.Generalised anxiety disorder: 8.7%.Bipolar disorder: 11.1%.Suicidal ideas: 36.4%.Attempted suicide: 55.6%.Forry et al. (2019)Severe depression: 44%.Antisocial personality disorder: 20.5%.Bipolar disorder: 23.5%.Generalised anxiety disorder: 30.9%.Panic disorder: 32.6%.Substance abuse: 12.5%.Fovet et al. (2020)Affective disorders: 31.2%.Anxiety disorders: 44.4%.Panic disorders: 13%.Substance abuse: 53.5%.Risk of suicide: 31.4%.Jakobowitz et al. (2017)Psychosis: 8.65%.Depression: 39.2%.Substance abuse: 45.8%.Alcohol abuse: 41.4%.Personality disorder: 17.8%.Anxiety: 8.1%.PTSD: 8.1%.Adaptation disorder: 5.1%.Van Buitenen et al. (2020)Schizophrenia: 56.7%. Substance use: 43.1%.Co-morbid disorders (impulsiveness. poor social skills, disruptive behaviour): 56.9%.Note. CI: confidence interval; PTSD: post-traumatic stress disorder. Note. CI: confidence interval; PTSD: post-traumatic stress disorder. [SUBTITLE] Recommendations for management [SUBSECTION] Adraro et al.5 consider that early examination of inmates when they arrive at prison is necessary, as are adequate treatment and monitoring. Jobs should also be created in prison to enable inmates to come together and develop positive relationships for social support and promote coping skills. Van Buitenen et al.7, consider that there is a possible causal relationship between psychopathology and criminal conduct, which should be borne in mind when developing treatment programmes for this sector of the population. According to Bartlet and Hollins14, mental health management of female inmates should be based on a system with sufficient resources to permit effective screening, individualised healthcare planning and primary and specialised medical care. A systematic review by Doyle et al.15 on treatment approaches for abuse of alcohol and other drugs amongst inmates, established that treatment is provided in different formats, including personalised consultations, group sessions, therapeutic communities and residential treatment programmes. Furthermore, the researchers observed that cognitive behavioural therapy of between 9 and 12 months is the most effective approach for treating inmates with this problem. According to Jakobowitz et al.9, there are specific and serious problems in psychiatric and psychological treatment of prison inmates. The limited availability of adequate premises often interrupts the continuity needed for care. Inmates can be released with no prior notice by the courts or transferred to other prisons, which is highly detrimental when planning subsequent treatment. Short periods of imprisonment are particularly likely to cause problems for mental health treatment, and can make access to prison psychiatric services difficult. Forry et al.10 also report a high prevalence of mental disorders in prisons, and state that it is necessary to develop the skills of health workers and other prison staff in detecting, evaluating and treating inmates with mental disorders. There is also the need for a clear referral process for person with such disorders, while improving the living conditions and standards of prison inmates would help to prevent mental disorders and their associated co-morbidities. Fazel et al.13 recommend that all prisons should have an identification system to identify persons with serious mental health problems, including case tracking systems when entering prison and assignment of adequate service levels. All prisons should also have a suicide prevention strategy that includes detection and close monitoring of risks, multi-disciplinary management of high-risk patients and staff training13. Gottfried et al.16 consider that mental health treatment needs for prison inmates are a vital concern, and highlight the importance of identifying strategies that can help to improve service provision and treat mental disorders more effectively. For Wilton and Stewart17, psychiatric services in prisons should detect substance use disorders, and if they are present, they should ensure that treatment is provided to improve the living conditions of the population, since this appears to be the key factor in contributing towards worse outcomes for inmates with mental disorders. According to Wills18, mental health professionals in prisons need to know the gender and age differences, along with the psychiatric history and background of inmates, to enable them to benefit from interviews with family members and friends, and to establish the security levels within the prison and the precautions necessary to prevent drug trafficking in prison. The author also considers that interventions based on cognitive behavioural therapy are very effective for prison inmates. Peters et al.19 consider that some mental health treatment and behavioural strategies in prisons should include integrated treatment of dual disorders, the risk-need-response model and cognitive behavioural therapy, and comment that the use of such strategies in the design of behavioural health services can significantly reduce criminal behaviours. In the case of substance use disorders, the aim of treatment in prison is to provide effective short-term services (1-3 months) to manage acute symptoms. The initial phases of treatment programmes include detoxification, psychiatric consultation to establish a psychotropic medication regimen, comprehensive evaluation and motivational strategies to involve inmates in treatment19. For Eher et al.20, the presence of a mental disorder is an important risk factor for criminal behaviour, not only for sexual crimes but for any type of infraction. Furthermore, existing disorders can worsen or new disorders may appear over the course of the inmate’s prison sentence. For this reason the authors consider that alongside a specific treatment to reduce risks, sex offenders with a mental disorder also often need interventions to manage the psychological and social consequences of this kind of disorder (Table 3). Table 3Therapeutic interventions for mental disorders amongst inmates .Author/year Therapeutic interventions Adraro et al. (2019)An early examination of inmates should be carried out when they arrive, with adequate treatment and follow up. Jobs should also be created that can bring inmates together and help to develop positive relationships, thus creating social support and helping with coping strategies. Bartlett et al. (2018)The management of female inmates' health should be based on a system with sufficient resources to enable screening, planning of individualised care and primary and specialised medical treatment, which makes it necessary to pat closer attention to the gender dimensions of women's previous experience, the nature of prison and the presence or absence of specific gender factors that help or impede mental wellbeing. It also states the mental health care should be based on respect for autonomy and self-determination, which are factors in up to 50% of the mental disorders amongst female inmates.Doyle et al. (2019)Behavioural cognitive therapy (9-12 months). Psycho-educational programmes. Motivational programmes.Eher et al. (2019)Alongside specific treatment for risk reduction, the psychological and social consequences of mental disorder should be considered in interventions to ensure effective and sustainable risk reduction. Fazel et al. (2016)The interventions to treat mental disorders in inmates include behavioural cognitive therapy, behavioural dialectic therapy and treatment based on meditation, in individual and group formats. Strategies to manage the risk of suicide include detection at reception, staff training, treatment, adequate supervision of inmates at risk, improved security of the physical setting and social support.Identification of mental problems.Suicide prevention strategies.Monitoring of substance dependence.Behavioural cognitive therapy.Methadone maintenance treatment.Gottfried et al. (2017)The importance of identifying strategies to help to improve service provision and more effectively treat mental illness, and the need to implement programmes designed to facilitate growth, maintain the treatment objectives and promote successful reintegration into society, would be of great help to this population. It is also necessary to implement programmes that bring about a successful transition of mentally ill inmates from the prison system to community-based mental health treatment services.Peters et al. (2017)Integrated treatment of dual disorders.Risk-need-response model.Behavioural cognitive therapy.Wills (2017)Mental health professionals working at prisons need to know the gender and age differences, the psychiatric record and background of the inmate, which can help in interviews with family and friends, the level of security assigned within the prison and the precautions required to prevent smuggling of drugs in prison. These authors consider interventions based on behavioural cognitive therapy to be highly effective. Adraro et al.5 consider that early examination of inmates when they arrive at prison is necessary, as are adequate treatment and monitoring. Jobs should also be created in prison to enable inmates to come together and develop positive relationships for social support and promote coping skills. Van Buitenen et al.7, consider that there is a possible causal relationship between psychopathology and criminal conduct, which should be borne in mind when developing treatment programmes for this sector of the population. According to Bartlet and Hollins14, mental health management of female inmates should be based on a system with sufficient resources to permit effective screening, individualised healthcare planning and primary and specialised medical care. A systematic review by Doyle et al.15 on treatment approaches for abuse of alcohol and other drugs amongst inmates, established that treatment is provided in different formats, including personalised consultations, group sessions, therapeutic communities and residential treatment programmes. Furthermore, the researchers observed that cognitive behavioural therapy of between 9 and 12 months is the most effective approach for treating inmates with this problem. According to Jakobowitz et al.9, there are specific and serious problems in psychiatric and psychological treatment of prison inmates. The limited availability of adequate premises often interrupts the continuity needed for care. Inmates can be released with no prior notice by the courts or transferred to other prisons, which is highly detrimental when planning subsequent treatment. Short periods of imprisonment are particularly likely to cause problems for mental health treatment, and can make access to prison psychiatric services difficult. Forry et al.10 also report a high prevalence of mental disorders in prisons, and state that it is necessary to develop the skills of health workers and other prison staff in detecting, evaluating and treating inmates with mental disorders. There is also the need for a clear referral process for person with such disorders, while improving the living conditions and standards of prison inmates would help to prevent mental disorders and their associated co-morbidities. Fazel et al.13 recommend that all prisons should have an identification system to identify persons with serious mental health problems, including case tracking systems when entering prison and assignment of adequate service levels. All prisons should also have a suicide prevention strategy that includes detection and close monitoring of risks, multi-disciplinary management of high-risk patients and staff training13. Gottfried et al.16 consider that mental health treatment needs for prison inmates are a vital concern, and highlight the importance of identifying strategies that can help to improve service provision and treat mental disorders more effectively. For Wilton and Stewart17, psychiatric services in prisons should detect substance use disorders, and if they are present, they should ensure that treatment is provided to improve the living conditions of the population, since this appears to be the key factor in contributing towards worse outcomes for inmates with mental disorders. According to Wills18, mental health professionals in prisons need to know the gender and age differences, along with the psychiatric history and background of inmates, to enable them to benefit from interviews with family members and friends, and to establish the security levels within the prison and the precautions necessary to prevent drug trafficking in prison. The author also considers that interventions based on cognitive behavioural therapy are very effective for prison inmates. Peters et al.19 consider that some mental health treatment and behavioural strategies in prisons should include integrated treatment of dual disorders, the risk-need-response model and cognitive behavioural therapy, and comment that the use of such strategies in the design of behavioural health services can significantly reduce criminal behaviours. In the case of substance use disorders, the aim of treatment in prison is to provide effective short-term services (1-3 months) to manage acute symptoms. The initial phases of treatment programmes include detoxification, psychiatric consultation to establish a psychotropic medication regimen, comprehensive evaluation and motivational strategies to involve inmates in treatment19. For Eher et al.20, the presence of a mental disorder is an important risk factor for criminal behaviour, not only for sexual crimes but for any type of infraction. Furthermore, existing disorders can worsen or new disorders may appear over the course of the inmate’s prison sentence. For this reason the authors consider that alongside a specific treatment to reduce risks, sex offenders with a mental disorder also often need interventions to manage the psychological and social consequences of this kind of disorder (Table 3). Table 3Therapeutic interventions for mental disorders amongst inmates .Author/year Therapeutic interventions Adraro et al. (2019)An early examination of inmates should be carried out when they arrive, with adequate treatment and follow up. Jobs should also be created that can bring inmates together and help to develop positive relationships, thus creating social support and helping with coping strategies. Bartlett et al. (2018)The management of female inmates' health should be based on a system with sufficient resources to enable screening, planning of individualised care and primary and specialised medical treatment, which makes it necessary to pat closer attention to the gender dimensions of women's previous experience, the nature of prison and the presence or absence of specific gender factors that help or impede mental wellbeing. It also states the mental health care should be based on respect for autonomy and self-determination, which are factors in up to 50% of the mental disorders amongst female inmates.Doyle et al. (2019)Behavioural cognitive therapy (9-12 months). Psycho-educational programmes. Motivational programmes.Eher et al. (2019)Alongside specific treatment for risk reduction, the psychological and social consequences of mental disorder should be considered in interventions to ensure effective and sustainable risk reduction. Fazel et al. (2016)The interventions to treat mental disorders in inmates include behavioural cognitive therapy, behavioural dialectic therapy and treatment based on meditation, in individual and group formats. Strategies to manage the risk of suicide include detection at reception, staff training, treatment, adequate supervision of inmates at risk, improved security of the physical setting and social support.Identification of mental problems.Suicide prevention strategies.Monitoring of substance dependence.Behavioural cognitive therapy.Methadone maintenance treatment.Gottfried et al. (2017)The importance of identifying strategies to help to improve service provision and more effectively treat mental illness, and the need to implement programmes designed to facilitate growth, maintain the treatment objectives and promote successful reintegration into society, would be of great help to this population. It is also necessary to implement programmes that bring about a successful transition of mentally ill inmates from the prison system to community-based mental health treatment services.Peters et al. (2017)Integrated treatment of dual disorders.Risk-need-response model.Behavioural cognitive therapy.Wills (2017)Mental health professionals working at prisons need to know the gender and age differences, the psychiatric record and background of the inmate, which can help in interviews with family and friends, the level of security assigned within the prison and the precautions required to prevent smuggling of drugs in prison. These authors consider interventions based on behavioural cognitive therapy to be highly effective.
null
null
[ "Prevalence of mental disorders and risk factors", "Recommendations for management" ]
[ "Analysis of the reports on the prevalence of mental disorders in the prison population showed a range of figures. In research carried out by Adraro et al.5, a prevalence of 62.7% for common mental disorders such as anxiety, depression and somatic symptom disorder was found in a group of 300 prison inmates, which is considered to be a high figure, in which three out of every five inmates are affected.\nIn another study by Baranyi et al.6, it was found that the prevalence of mental disorders was up to 15.8 times higher amongst inmates than they were in the general public, with a tendency to decrease with time spent in prison. The authors also commented that the inmates were often from a low socio-economic background, belonged to minority groups and had a history of child abuse and substance use, which made them vulnerable to psychiatric disorders. The poor living conditions, physical aggression and psychological abuse often encountered in prison can further aggravate mental disorders.\nAccording to Van Buitenen et al.7, the prevalence of schizophrenia was 56.7%; for substance abuse disorder, 43.1%; while up to 56.9% of the 5,247 inmates had other mental disorders, such as impulsiveness, poor social skills and disruptive behaviour.\nOn the other hand, a meta-analysis by Facer et al.8 showed that post-traumatic stress disorder (PTSD) was associated with a number of mental disorders such as depression and anxiety disorders. Inmates with PTSD were also significantly more likely to suffer from substance use disorder.\nA research project by Jakobowitz et al.9 showed that the prevalence of mental disorders amongst inmates was 8.65% for psychosis; 39.2% for depression; 45.8% for substance abuse; 41.4% for alcohol abuse; 17.8% for personality disorders; 8.1% for anxiety disorders and PTSD; and 5.1% for adaptation disorders. According to Forry et al.10 and Fovet et al.11, the most common mental disorders in the prison population are severe depression (44% and 31.2%, respectively), followed by generalised anxiety disorder (30.9 and 44.4%, respectively).\nFavril et al.12 describe a high prevalence of mental disorders in the prison population of New Zealand: up to 90.7% of the inmates present at least one mental disorder; substance abuse was present in 87% of the inmates; suicidal ideation was present in 36.4%; and suicide attempts in 55.6%.\nAnother study by Fazel et al.13 mentions that there is an important association between severe depression and psychotic illness (p<0.05), and states that one out of every seven inmates suffers from depression or psychosis. They also mention high rates of coincidence between mental illness and substance use, as well as suicide and self-harming, which are more common amongst inmates than the general public of the same age and sex. According to the authors, prison inmates with mental disorders are disproportionately involved in prison infractions and violent incidents, and are more likely to be accused of breaking prison rules and being injured in fights. Inmates with mental disorders and with a history of violence run an even higher risk in this regard, since psychiatric disorders are associated with violent incidents in prison (Table 2).\n\nTable 2Prevalence of mental disorders amongst inmates.Author/year PrevalenceAdraro et al. (2019)Common mental disorders: 62.7% (CI 95%: 65.7-68.5).Baranyi et al. (2019)Psychosis: 6.2%.Severe depression: 3.8%.Alcohol use: 3.8%.Substance use: 5.1%.Eher et al. (2019)Mental disorder: 92.9%.Alcohol abuse: 40%.Paraphilias: 43.3%.Personality disorders: 53.6%.Type B personality disorders: 47.8%.Favril et al. (2020)Substance abuse: 87%.Any mental disorder: 90.7%.PTSD: 23.9%.Panic disorder: 5.6%.Generalised anxiety disorder: 8.7%.Bipolar disorder: 11.1%.Suicidal ideas: 36.4%.Attempted suicide: 55.6%.Forry et al. (2019)Severe depression: 44%.Antisocial personality disorder: 20.5%.Bipolar disorder: 23.5%.Generalised anxiety disorder: 30.9%.Panic disorder: 32.6%.Substance abuse: 12.5%.Fovet et al. (2020)Affective disorders: 31.2%.Anxiety disorders: 44.4%.Panic disorders: 13%.Substance abuse: 53.5%.Risk of suicide: 31.4%.Jakobowitz et al. (2017)Psychosis: 8.65%.Depression: 39.2%.Substance abuse: 45.8%.Alcohol abuse: 41.4%.Personality disorder: 17.8%.Anxiety: 8.1%.PTSD: 8.1%.Adaptation disorder: 5.1%.Van Buitenen et al. (2020)Schizophrenia: 56.7%. Substance use: 43.1%.Co-morbid disorders (impulsiveness. poor social skills, disruptive behaviour): 56.9%.Note. CI: confidence interval; PTSD: post-traumatic stress disorder.\n\nNote. CI: confidence interval; PTSD: post-traumatic stress disorder.", "Adraro et al.5 consider that early examination of inmates when they arrive at prison is necessary, as are adequate treatment and monitoring. Jobs should also be created in prison to enable inmates to come together and develop positive relationships for social support and promote coping skills. Van Buitenen et al.7, consider that there is a possible causal relationship between psychopathology and criminal conduct, which should be borne in mind when developing treatment programmes for this sector of the population.\nAccording to Bartlet and Hollins14, mental health management of female inmates should be based on a system with sufficient resources to permit effective screening, individualised healthcare planning and primary and specialised medical care.\nA systematic review by Doyle et al.15 on treatment approaches for abuse of alcohol and other drugs amongst inmates, established that treatment is provided in different formats, including personalised consultations, group sessions, therapeutic communities and residential treatment programmes. Furthermore, the researchers observed that cognitive behavioural therapy of between 9 and 12 months is the most effective approach for treating inmates with this problem.\nAccording to Jakobowitz et al.9, there are specific and serious problems in psychiatric and psychological treatment of prison inmates. The limited availability of adequate premises often interrupts the continuity needed for care. Inmates can be released with no prior notice by the courts or transferred to other prisons, which is highly detrimental when planning subsequent treatment. Short periods of imprisonment are particularly likely to cause problems for mental health treatment, and can make access to prison psychiatric services difficult.\nForry et al.10 also report a high prevalence of mental disorders in prisons, and state that it is necessary to develop the skills of health workers and other prison staff in detecting, evaluating and treating inmates with mental disorders. There is also the need for a clear referral process for person with such disorders, while improving the living conditions and standards of prison inmates would help to prevent mental disorders and their associated co-morbidities.\nFazel et al.13 recommend that all prisons should have an identification system to identify persons with serious mental health problems, including case tracking systems when entering prison and assignment of adequate service levels. All prisons should also have a suicide prevention strategy that includes detection and close monitoring of risks, multi-disciplinary management of high-risk patients and staff training13.\nGottfried et al.16 consider that mental health treatment needs for prison inmates are a vital concern, and highlight the importance of identifying strategies that can help to improve service provision and treat mental disorders more effectively.\nFor Wilton and Stewart17, psychiatric services in prisons should detect substance use disorders, and if they are present, they should ensure that treatment is provided to improve the living conditions of the population, since this appears to be the key factor in contributing towards worse outcomes for inmates with mental disorders.\nAccording to Wills18, mental health professionals in prisons need to know the gender and age differences, along with the psychiatric history and background of inmates, to enable them to benefit from interviews with family members and friends, and to establish the security levels within the prison and the precautions necessary to prevent drug trafficking in prison. The author also considers that interventions based on cognitive behavioural therapy are very effective for prison inmates.\nPeters et al.19 consider that some mental health treatment and behavioural strategies in prisons should include integrated treatment of dual disorders, the risk-need-response model and cognitive behavioural therapy, and comment that the use of such strategies in the design of behavioural health services can significantly reduce criminal behaviours.\nIn the case of substance use disorders, the aim of treatment in prison is to provide effective short-term services (1-3 months) to manage acute symptoms. The initial phases of treatment programmes include detoxification, psychiatric consultation to establish a psychotropic medication regimen, comprehensive evaluation and motivational strategies to involve inmates in treatment19.\nFor Eher et al.20, the presence of a mental disorder is an important risk factor for criminal behaviour, not only for sexual crimes but for any type of infraction. Furthermore, existing disorders can worsen or new disorders may appear over the course of the inmate’s prison sentence. For this reason the authors consider that alongside a specific treatment to reduce risks, sex offenders with a mental disorder also often need interventions to manage the psychological and social consequences of this kind of disorder (Table 3).\n\nTable 3Therapeutic interventions for mental disorders amongst inmates .Author/year Therapeutic interventions Adraro et al. (2019)An early examination of inmates should be carried out when they arrive, with adequate treatment and follow up. Jobs should also be created that can bring inmates together and help to develop positive relationships, thus creating social support and helping with coping strategies. Bartlett et al. (2018)The management of female inmates' health should be based on a system with sufficient resources to enable screening, planning of individualised care and primary and specialised medical treatment, which makes it necessary to pat closer attention to the gender dimensions of women's previous experience, the nature of prison and the presence or absence of specific gender factors that help or impede mental wellbeing. It also states the mental health care should be based on respect for autonomy and self-determination, which are factors in up to 50% of the mental disorders amongst female inmates.Doyle et al. (2019)Behavioural cognitive therapy (9-12 months). Psycho-educational programmes. Motivational programmes.Eher et al. (2019)Alongside specific treatment for risk reduction, the psychological and social consequences of mental disorder should be considered in interventions to ensure effective and sustainable risk reduction. Fazel et al. (2016)The interventions to treat mental disorders in inmates include behavioural cognitive therapy, behavioural dialectic therapy and treatment based on meditation, in individual and group formats. Strategies to manage the risk of suicide include detection at reception, staff training, treatment, adequate supervision of inmates at risk, improved security of the physical setting and social support.Identification of mental problems.Suicide prevention strategies.Monitoring of substance dependence.Behavioural cognitive therapy.Methadone maintenance treatment.Gottfried et al. (2017)The importance of identifying strategies to help to improve service provision and more effectively treat mental illness, and the need to implement programmes designed to facilitate growth, maintain the treatment objectives and promote successful reintegration into society, would be of great help to this population. It is also necessary to implement programmes that bring about a successful transition of mentally ill inmates from the prison system to community-based mental health treatment services.Peters et al. (2017)Integrated treatment of dual disorders.Risk-need-response model.Behavioural cognitive therapy.Wills (2017)Mental health professionals working at prisons need to know the gender and age differences, the psychiatric record and background of the inmate, which can help in interviews with family and friends, the level of security assigned within the prison and the precautions required to prevent smuggling of drugs in prison. These authors consider interventions based on behavioural cognitive therapy to be highly effective.\n" ]
[ null, null ]
[ "Introduction", "Material and method", "Resultats", "Prevalence of mental disorders and risk factors", "Recommendations for management", "Discussion" ]
[ "The World Health Organisation states that problems related to mental health are up to seven times more likely to appear in the prison population than amongst the general public in Western societies. This upward trend in mental disorders coincides with a growing prison population. Another aggravating factor is substance use in prisons1.\nThe behavioural changes that lead to mental disorders and prison violence often take place in contexts of inhumane confinement conditions that are common in Latin-American prisons. Inmates undergo a process of adaptation that can include injury and aggression, and in some cases, overcrowding, hunger and disease during imprisonment2.\nIt has been shown that there are failures in inmates’ adaptation mechanisms during the confinement process, which vary according to there they are imprisoned and the length of sentence. There is a reduction in the individual’s strategies that enable them to act in reaction to being imprisoned; there is also a loss of ideas, actions and feelings that gradually affect an individual’s relationship with their surroundings3.\nAlthough changes are being made to improve the treatment of inmates in the prison system of several Latin American countries, there are prisons where the care of patients with previous psychiatric disorders, such as schizophrenia, is in the hand of general physicians. The profile for this group of patients is a complex one, and poor treatment adherence and drug consumption are very common4.\nThe presence of psychiatric disorders in this context is a reality, as is the link between pre-existing mental disorders and the frequent exacerbation of symptoms and imbalances. It is therefore important to carry out this theoretical review, to highlight the need to identify and offer timely treatment for mental disorders in the prison population, with the active participation of the psychiatrist as a specialist in this field. We also seek to identify the possible causes behind the appearance of mental disorders. In this case it is essential to consider the emotional changes brought about by imprisonment and the inability to adapt to incarceration.\nThere are few studies in Ecuador on this issue, which makes this review article even more relevant, as it will be of benefit not only to health professionals but to all those who work directly with prison inmates.\nThis theoretical review was carried out to compile information about the prevalence of mental and behavioural disorders in the prison setting, and also to highlight the importance of early psychiatric intervention. The recommendations for managing this group in situations of confinement, with a focus on the role of the psychiatrist as a mental health professional, are also described.\nThe results of this review may help psychiatric practice in prisons, since they offer a compendium of up to date information about prevalence, risk factors and therapeutic approaches. The results of this research also highlight the importance of restructuring mental health care for inmates, since psychiatric evaluation is often late and when it is adequately carried out, ongoing monitoring is infrequent, despite being regulated under the law. Another factor is the constant turnover in staff working at these institutions, given that there is a rotation system for professionals, who offer their services on a transient basis, while many professionals do not have experience in managing patients with mental disorders.", "A theoretical review of scientific articles was carried out, where articles from journals indexed as primary information sources were used. Secondary sources included specialised databases such as: PubMed, SCOPUS, SciELO, latindex, DOAJ and Google Scholar.\nThe use of MESH terms related to the review was established as the search strategy: mental disorders, prison, mental health, penitentiary, confinement centre, penitentiary system, social re-adaptation, mental pathology, psychiatry, mental illness, mental health in prison, prison psychiatry, mental illness in prisons.\nThe inclusion criteria consisted of articles about mental and behavioural disorders in the prison context, published between January 2016 and April 2021, original articles, with an analytical design, observational, prospective, retrospective, cross-sectional, randomised, systematic reviews and meta-analyses. Complete articles prepared in any country with subjects over 15 years of age, of any sex or gender and articles from indexed journals in English and Spanish were also included.\nThe exclusion criteria included duplicated articles, paid-access articles, articles with no declaration of conflicts of interest, digital or printed press articles, case reports, conference minutes, letters to the editor, graduate theses, articles with no bioethical considerations.\nThe PICOT research model was used for preparing the questions:\n\nPopulation (P): prison inmates over 18 years of age.Intervention (I): bibliographical review about mental and behavioural disorders, their prevalence and treatment strategies.Comparison (C): not applicable.Outcome (O): compendium of evidence available about the main mental and behavioural disorders in the prison population, their incidence and treatment.Time (T): from January 2016 to April 2021.\n\nPopulation (P): prison inmates over 18 years of age.\nIntervention (I): bibliographical review about mental and behavioural disorders, their prevalence and treatment strategies.\nComparison (C): not applicable.\nOutcome (O): compendium of evidence available about the main mental and behavioural disorders in the prison population, their incidence and treatment.\nTime (T): from January 2016 to April 2021.\nThe bibliographical review responded to the following questions: “What are the most common mental and behavioural disorders in the prison population?” and “What are the treatment strategies for mental and behavioural disorders in the prison population?”\nPart of the methodology used was a search in the specialised databases, using the key words as the main strategy (MESH descriptors). The inclusion and exclusion criteria were included in the search to obtain information more simply. The results of each article were then reviewed, and their theoretical and methodological approaches were considered. The Cochrane criteria were used for quality of evidence and risk of bias. Once this process was completed, the material was then analysed and the conclusions of the theoretical review were then drawn.", "A total of 29 articles on mental disorders amongst prison inmates were identified. They were published between January 2016 and April 2021. 3 of them were removed since they were published in German, 2 for using a qualitative design, 3 journal editorials, 2 articles that were duplicated and 3 for not having a clear and reproducible methodology. 16 articles were left at the end of the selection process, and these were included in the theoretical review (Table 1).\n\nTable 1Traceability.Author/yearCountry Journal Browser URL Adraro et al. (2019)EthiopiaBMC Public Health BMC\nhttps://bmcpublichealth.biomedcentral.com/articles/10.1186/s12889-019-7879-6\nBaranyi et al. (2019)UK Lancet Glob HealthThe Lancet\nhttps://www.thelancet.com/journals/langlo/article/PIIS2214-109X(18)30539-4/fulltext\nBartlett et al. (2018)UKBr J PsychiatryPubmed \nhttps://pubmed.ncbi.nlm.nih.gov/29486822/\nDoyle et al. (2019)Australia Aust N Z J Public HealthPubmed \nhttps://pubmed.ncbi.nlm.nih.gov/30908856/\nEher et al. (2019) Austria Acta Psychiatr ScandWiley Online Library \n>https://onlinelibrary.wiley.com/doi/abs/10.1111/acps.13024\nFacer et al. (2019) UK PLoS OnePubmed \nhttps://pubmed.ncbi.nlm.nih.gov/31557173/\nFavril et al. (2020)New Zealand Soc Psychiatry Psychiatr EpidemiolSpringer Link \nhttps://link.springer.com/article/10.1007%2Fs00127-020-01851-7\nFazel et al. (2016) UK Lancet PsychiatryPubmed \nhttps://www.ncbi.nlm.nih.gov/pmc/articles/PMC5008459/\nForry et al. (2019) Uganda BMC PsychiatryBMC \n>https://bmcpsychiatry.biomedcentral.com/articles/10.1186/s12888-019-2167-7\nFovet et al. (2020) FranceEur PsychiatryPubmed \nhttps://pubmed.ncbi.nlm.nih.gov/32336297/\nGottfried et al. (2017) USAJ Correct Health Care Pubmed \nhttps://pubmed.ncbi.nlm.nih.gov/28715985/\nJakobowitz et al. (2017) UK Soc Psychiatry Psychiatr EpidemiolSpringer Link\nhttps://link.springer.com/article/10.1007%2Fs00127-016-1313-5\nPeters et al. (2017) USA Am J Drug Alcohol AbuseTaylor & Francis Online \nhttps://www.tandfonline.com/doi/full/10.1080/00952990.2017.1303838\nVan Buitenen et al. (2020) Holland Eur Psychiatry Pubmed \nhttps://pubmed.ncbi.nlm.nih.gov/32522312/\nWills (2017)USA Int Rev PsychiatryPubmed \nhttps://pubmed.ncbi.nlm.nih.gov/27701919/\nWilton et al. (2017)Canada Psychiatr ServPubmed \nhttps://pubmed.ncbi.nlm.nih.gov/28292226/\n\n\n[SUBTITLE] Prevalence of mental disorders and risk factors [SUBSECTION] Analysis of the reports on the prevalence of mental disorders in the prison population showed a range of figures. In research carried out by Adraro et al.5, a prevalence of 62.7% for common mental disorders such as anxiety, depression and somatic symptom disorder was found in a group of 300 prison inmates, which is considered to be a high figure, in which three out of every five inmates are affected.\nIn another study by Baranyi et al.6, it was found that the prevalence of mental disorders was up to 15.8 times higher amongst inmates than they were in the general public, with a tendency to decrease with time spent in prison. The authors also commented that the inmates were often from a low socio-economic background, belonged to minority groups and had a history of child abuse and substance use, which made them vulnerable to psychiatric disorders. The poor living conditions, physical aggression and psychological abuse often encountered in prison can further aggravate mental disorders.\nAccording to Van Buitenen et al.7, the prevalence of schizophrenia was 56.7%; for substance abuse disorder, 43.1%; while up to 56.9% of the 5,247 inmates had other mental disorders, such as impulsiveness, poor social skills and disruptive behaviour.\nOn the other hand, a meta-analysis by Facer et al.8 showed that post-traumatic stress disorder (PTSD) was associated with a number of mental disorders such as depression and anxiety disorders. Inmates with PTSD were also significantly more likely to suffer from substance use disorder.\nA research project by Jakobowitz et al.9 showed that the prevalence of mental disorders amongst inmates was 8.65% for psychosis; 39.2% for depression; 45.8% for substance abuse; 41.4% for alcohol abuse; 17.8% for personality disorders; 8.1% for anxiety disorders and PTSD; and 5.1% for adaptation disorders. According to Forry et al.10 and Fovet et al.11, the most common mental disorders in the prison population are severe depression (44% and 31.2%, respectively), followed by generalised anxiety disorder (30.9 and 44.4%, respectively).\nFavril et al.12 describe a high prevalence of mental disorders in the prison population of New Zealand: up to 90.7% of the inmates present at least one mental disorder; substance abuse was present in 87% of the inmates; suicidal ideation was present in 36.4%; and suicide attempts in 55.6%.\nAnother study by Fazel et al.13 mentions that there is an important association between severe depression and psychotic illness (p<0.05), and states that one out of every seven inmates suffers from depression or psychosis. They also mention high rates of coincidence between mental illness and substance use, as well as suicide and self-harming, which are more common amongst inmates than the general public of the same age and sex. According to the authors, prison inmates with mental disorders are disproportionately involved in prison infractions and violent incidents, and are more likely to be accused of breaking prison rules and being injured in fights. Inmates with mental disorders and with a history of violence run an even higher risk in this regard, since psychiatric disorders are associated with violent incidents in prison (Table 2).\n\nTable 2Prevalence of mental disorders amongst inmates.Author/year PrevalenceAdraro et al. (2019)Common mental disorders: 62.7% (CI 95%: 65.7-68.5).Baranyi et al. (2019)Psychosis: 6.2%.Severe depression: 3.8%.Alcohol use: 3.8%.Substance use: 5.1%.Eher et al. (2019)Mental disorder: 92.9%.Alcohol abuse: 40%.Paraphilias: 43.3%.Personality disorders: 53.6%.Type B personality disorders: 47.8%.Favril et al. (2020)Substance abuse: 87%.Any mental disorder: 90.7%.PTSD: 23.9%.Panic disorder: 5.6%.Generalised anxiety disorder: 8.7%.Bipolar disorder: 11.1%.Suicidal ideas: 36.4%.Attempted suicide: 55.6%.Forry et al. (2019)Severe depression: 44%.Antisocial personality disorder: 20.5%.Bipolar disorder: 23.5%.Generalised anxiety disorder: 30.9%.Panic disorder: 32.6%.Substance abuse: 12.5%.Fovet et al. (2020)Affective disorders: 31.2%.Anxiety disorders: 44.4%.Panic disorders: 13%.Substance abuse: 53.5%.Risk of suicide: 31.4%.Jakobowitz et al. (2017)Psychosis: 8.65%.Depression: 39.2%.Substance abuse: 45.8%.Alcohol abuse: 41.4%.Personality disorder: 17.8%.Anxiety: 8.1%.PTSD: 8.1%.Adaptation disorder: 5.1%.Van Buitenen et al. (2020)Schizophrenia: 56.7%. Substance use: 43.1%.Co-morbid disorders (impulsiveness. poor social skills, disruptive behaviour): 56.9%.Note. CI: confidence interval; PTSD: post-traumatic stress disorder.\n\nNote. CI: confidence interval; PTSD: post-traumatic stress disorder.\nAnalysis of the reports on the prevalence of mental disorders in the prison population showed a range of figures. In research carried out by Adraro et al.5, a prevalence of 62.7% for common mental disorders such as anxiety, depression and somatic symptom disorder was found in a group of 300 prison inmates, which is considered to be a high figure, in which three out of every five inmates are affected.\nIn another study by Baranyi et al.6, it was found that the prevalence of mental disorders was up to 15.8 times higher amongst inmates than they were in the general public, with a tendency to decrease with time spent in prison. The authors also commented that the inmates were often from a low socio-economic background, belonged to minority groups and had a history of child abuse and substance use, which made them vulnerable to psychiatric disorders. The poor living conditions, physical aggression and psychological abuse often encountered in prison can further aggravate mental disorders.\nAccording to Van Buitenen et al.7, the prevalence of schizophrenia was 56.7%; for substance abuse disorder, 43.1%; while up to 56.9% of the 5,247 inmates had other mental disorders, such as impulsiveness, poor social skills and disruptive behaviour.\nOn the other hand, a meta-analysis by Facer et al.8 showed that post-traumatic stress disorder (PTSD) was associated with a number of mental disorders such as depression and anxiety disorders. Inmates with PTSD were also significantly more likely to suffer from substance use disorder.\nA research project by Jakobowitz et al.9 showed that the prevalence of mental disorders amongst inmates was 8.65% for psychosis; 39.2% for depression; 45.8% for substance abuse; 41.4% for alcohol abuse; 17.8% for personality disorders; 8.1% for anxiety disorders and PTSD; and 5.1% for adaptation disorders. According to Forry et al.10 and Fovet et al.11, the most common mental disorders in the prison population are severe depression (44% and 31.2%, respectively), followed by generalised anxiety disorder (30.9 and 44.4%, respectively).\nFavril et al.12 describe a high prevalence of mental disorders in the prison population of New Zealand: up to 90.7% of the inmates present at least one mental disorder; substance abuse was present in 87% of the inmates; suicidal ideation was present in 36.4%; and suicide attempts in 55.6%.\nAnother study by Fazel et al.13 mentions that there is an important association between severe depression and psychotic illness (p<0.05), and states that one out of every seven inmates suffers from depression or psychosis. They also mention high rates of coincidence between mental illness and substance use, as well as suicide and self-harming, which are more common amongst inmates than the general public of the same age and sex. According to the authors, prison inmates with mental disorders are disproportionately involved in prison infractions and violent incidents, and are more likely to be accused of breaking prison rules and being injured in fights. Inmates with mental disorders and with a history of violence run an even higher risk in this regard, since psychiatric disorders are associated with violent incidents in prison (Table 2).\n\nTable 2Prevalence of mental disorders amongst inmates.Author/year PrevalenceAdraro et al. (2019)Common mental disorders: 62.7% (CI 95%: 65.7-68.5).Baranyi et al. (2019)Psychosis: 6.2%.Severe depression: 3.8%.Alcohol use: 3.8%.Substance use: 5.1%.Eher et al. (2019)Mental disorder: 92.9%.Alcohol abuse: 40%.Paraphilias: 43.3%.Personality disorders: 53.6%.Type B personality disorders: 47.8%.Favril et al. (2020)Substance abuse: 87%.Any mental disorder: 90.7%.PTSD: 23.9%.Panic disorder: 5.6%.Generalised anxiety disorder: 8.7%.Bipolar disorder: 11.1%.Suicidal ideas: 36.4%.Attempted suicide: 55.6%.Forry et al. (2019)Severe depression: 44%.Antisocial personality disorder: 20.5%.Bipolar disorder: 23.5%.Generalised anxiety disorder: 30.9%.Panic disorder: 32.6%.Substance abuse: 12.5%.Fovet et al. (2020)Affective disorders: 31.2%.Anxiety disorders: 44.4%.Panic disorders: 13%.Substance abuse: 53.5%.Risk of suicide: 31.4%.Jakobowitz et al. (2017)Psychosis: 8.65%.Depression: 39.2%.Substance abuse: 45.8%.Alcohol abuse: 41.4%.Personality disorder: 17.8%.Anxiety: 8.1%.PTSD: 8.1%.Adaptation disorder: 5.1%.Van Buitenen et al. (2020)Schizophrenia: 56.7%. Substance use: 43.1%.Co-morbid disorders (impulsiveness. poor social skills, disruptive behaviour): 56.9%.Note. CI: confidence interval; PTSD: post-traumatic stress disorder.\n\nNote. CI: confidence interval; PTSD: post-traumatic stress disorder.\n[SUBTITLE] Recommendations for management [SUBSECTION] Adraro et al.5 consider that early examination of inmates when they arrive at prison is necessary, as are adequate treatment and monitoring. Jobs should also be created in prison to enable inmates to come together and develop positive relationships for social support and promote coping skills. Van Buitenen et al.7, consider that there is a possible causal relationship between psychopathology and criminal conduct, which should be borne in mind when developing treatment programmes for this sector of the population.\nAccording to Bartlet and Hollins14, mental health management of female inmates should be based on a system with sufficient resources to permit effective screening, individualised healthcare planning and primary and specialised medical care.\nA systematic review by Doyle et al.15 on treatment approaches for abuse of alcohol and other drugs amongst inmates, established that treatment is provided in different formats, including personalised consultations, group sessions, therapeutic communities and residential treatment programmes. Furthermore, the researchers observed that cognitive behavioural therapy of between 9 and 12 months is the most effective approach for treating inmates with this problem.\nAccording to Jakobowitz et al.9, there are specific and serious problems in psychiatric and psychological treatment of prison inmates. The limited availability of adequate premises often interrupts the continuity needed for care. Inmates can be released with no prior notice by the courts or transferred to other prisons, which is highly detrimental when planning subsequent treatment. Short periods of imprisonment are particularly likely to cause problems for mental health treatment, and can make access to prison psychiatric services difficult.\nForry et al.10 also report a high prevalence of mental disorders in prisons, and state that it is necessary to develop the skills of health workers and other prison staff in detecting, evaluating and treating inmates with mental disorders. There is also the need for a clear referral process for person with such disorders, while improving the living conditions and standards of prison inmates would help to prevent mental disorders and their associated co-morbidities.\nFazel et al.13 recommend that all prisons should have an identification system to identify persons with serious mental health problems, including case tracking systems when entering prison and assignment of adequate service levels. All prisons should also have a suicide prevention strategy that includes detection and close monitoring of risks, multi-disciplinary management of high-risk patients and staff training13.\nGottfried et al.16 consider that mental health treatment needs for prison inmates are a vital concern, and highlight the importance of identifying strategies that can help to improve service provision and treat mental disorders more effectively.\nFor Wilton and Stewart17, psychiatric services in prisons should detect substance use disorders, and if they are present, they should ensure that treatment is provided to improve the living conditions of the population, since this appears to be the key factor in contributing towards worse outcomes for inmates with mental disorders.\nAccording to Wills18, mental health professionals in prisons need to know the gender and age differences, along with the psychiatric history and background of inmates, to enable them to benefit from interviews with family members and friends, and to establish the security levels within the prison and the precautions necessary to prevent drug trafficking in prison. The author also considers that interventions based on cognitive behavioural therapy are very effective for prison inmates.\nPeters et al.19 consider that some mental health treatment and behavioural strategies in prisons should include integrated treatment of dual disorders, the risk-need-response model and cognitive behavioural therapy, and comment that the use of such strategies in the design of behavioural health services can significantly reduce criminal behaviours.\nIn the case of substance use disorders, the aim of treatment in prison is to provide effective short-term services (1-3 months) to manage acute symptoms. The initial phases of treatment programmes include detoxification, psychiatric consultation to establish a psychotropic medication regimen, comprehensive evaluation and motivational strategies to involve inmates in treatment19.\nFor Eher et al.20, the presence of a mental disorder is an important risk factor for criminal behaviour, not only for sexual crimes but for any type of infraction. Furthermore, existing disorders can worsen or new disorders may appear over the course of the inmate’s prison sentence. For this reason the authors consider that alongside a specific treatment to reduce risks, sex offenders with a mental disorder also often need interventions to manage the psychological and social consequences of this kind of disorder (Table 3).\n\nTable 3Therapeutic interventions for mental disorders amongst inmates .Author/year Therapeutic interventions Adraro et al. (2019)An early examination of inmates should be carried out when they arrive, with adequate treatment and follow up. Jobs should also be created that can bring inmates together and help to develop positive relationships, thus creating social support and helping with coping strategies. Bartlett et al. (2018)The management of female inmates' health should be based on a system with sufficient resources to enable screening, planning of individualised care and primary and specialised medical treatment, which makes it necessary to pat closer attention to the gender dimensions of women's previous experience, the nature of prison and the presence or absence of specific gender factors that help or impede mental wellbeing. It also states the mental health care should be based on respect for autonomy and self-determination, which are factors in up to 50% of the mental disorders amongst female inmates.Doyle et al. (2019)Behavioural cognitive therapy (9-12 months). Psycho-educational programmes. Motivational programmes.Eher et al. (2019)Alongside specific treatment for risk reduction, the psychological and social consequences of mental disorder should be considered in interventions to ensure effective and sustainable risk reduction. Fazel et al. (2016)The interventions to treat mental disorders in inmates include behavioural cognitive therapy, behavioural dialectic therapy and treatment based on meditation, in individual and group formats. Strategies to manage the risk of suicide include detection at reception, staff training, treatment, adequate supervision of inmates at risk, improved security of the physical setting and social support.Identification of mental problems.Suicide prevention strategies.Monitoring of substance dependence.Behavioural cognitive therapy.Methadone maintenance treatment.Gottfried et al. (2017)The importance of identifying strategies to help to improve service provision and more effectively treat mental illness, and the need to implement programmes designed to facilitate growth, maintain the treatment objectives and promote successful reintegration into society, would be of great help to this population. It is also necessary to implement programmes that bring about a successful transition of mentally ill inmates from the prison system to community-based mental health treatment services.Peters et al. (2017)Integrated treatment of dual disorders.Risk-need-response model.Behavioural cognitive therapy.Wills (2017)Mental health professionals working at prisons need to know the gender and age differences, the psychiatric record and background of the inmate, which can help in interviews with family and friends, the level of security assigned within the prison and the precautions required to prevent smuggling of drugs in prison. These authors consider interventions based on behavioural cognitive therapy to be highly effective.\n\nAdraro et al.5 consider that early examination of inmates when they arrive at prison is necessary, as are adequate treatment and monitoring. Jobs should also be created in prison to enable inmates to come together and develop positive relationships for social support and promote coping skills. Van Buitenen et al.7, consider that there is a possible causal relationship between psychopathology and criminal conduct, which should be borne in mind when developing treatment programmes for this sector of the population.\nAccording to Bartlet and Hollins14, mental health management of female inmates should be based on a system with sufficient resources to permit effective screening, individualised healthcare planning and primary and specialised medical care.\nA systematic review by Doyle et al.15 on treatment approaches for abuse of alcohol and other drugs amongst inmates, established that treatment is provided in different formats, including personalised consultations, group sessions, therapeutic communities and residential treatment programmes. Furthermore, the researchers observed that cognitive behavioural therapy of between 9 and 12 months is the most effective approach for treating inmates with this problem.\nAccording to Jakobowitz et al.9, there are specific and serious problems in psychiatric and psychological treatment of prison inmates. The limited availability of adequate premises often interrupts the continuity needed for care. Inmates can be released with no prior notice by the courts or transferred to other prisons, which is highly detrimental when planning subsequent treatment. Short periods of imprisonment are particularly likely to cause problems for mental health treatment, and can make access to prison psychiatric services difficult.\nForry et al.10 also report a high prevalence of mental disorders in prisons, and state that it is necessary to develop the skills of health workers and other prison staff in detecting, evaluating and treating inmates with mental disorders. There is also the need for a clear referral process for person with such disorders, while improving the living conditions and standards of prison inmates would help to prevent mental disorders and their associated co-morbidities.\nFazel et al.13 recommend that all prisons should have an identification system to identify persons with serious mental health problems, including case tracking systems when entering prison and assignment of adequate service levels. All prisons should also have a suicide prevention strategy that includes detection and close monitoring of risks, multi-disciplinary management of high-risk patients and staff training13.\nGottfried et al.16 consider that mental health treatment needs for prison inmates are a vital concern, and highlight the importance of identifying strategies that can help to improve service provision and treat mental disorders more effectively.\nFor Wilton and Stewart17, psychiatric services in prisons should detect substance use disorders, and if they are present, they should ensure that treatment is provided to improve the living conditions of the population, since this appears to be the key factor in contributing towards worse outcomes for inmates with mental disorders.\nAccording to Wills18, mental health professionals in prisons need to know the gender and age differences, along with the psychiatric history and background of inmates, to enable them to benefit from interviews with family members and friends, and to establish the security levels within the prison and the precautions necessary to prevent drug trafficking in prison. The author also considers that interventions based on cognitive behavioural therapy are very effective for prison inmates.\nPeters et al.19 consider that some mental health treatment and behavioural strategies in prisons should include integrated treatment of dual disorders, the risk-need-response model and cognitive behavioural therapy, and comment that the use of such strategies in the design of behavioural health services can significantly reduce criminal behaviours.\nIn the case of substance use disorders, the aim of treatment in prison is to provide effective short-term services (1-3 months) to manage acute symptoms. The initial phases of treatment programmes include detoxification, psychiatric consultation to establish a psychotropic medication regimen, comprehensive evaluation and motivational strategies to involve inmates in treatment19.\nFor Eher et al.20, the presence of a mental disorder is an important risk factor for criminal behaviour, not only for sexual crimes but for any type of infraction. Furthermore, existing disorders can worsen or new disorders may appear over the course of the inmate’s prison sentence. For this reason the authors consider that alongside a specific treatment to reduce risks, sex offenders with a mental disorder also often need interventions to manage the psychological and social consequences of this kind of disorder (Table 3).\n\nTable 3Therapeutic interventions for mental disorders amongst inmates .Author/year Therapeutic interventions Adraro et al. (2019)An early examination of inmates should be carried out when they arrive, with adequate treatment and follow up. Jobs should also be created that can bring inmates together and help to develop positive relationships, thus creating social support and helping with coping strategies. Bartlett et al. (2018)The management of female inmates' health should be based on a system with sufficient resources to enable screening, planning of individualised care and primary and specialised medical treatment, which makes it necessary to pat closer attention to the gender dimensions of women's previous experience, the nature of prison and the presence or absence of specific gender factors that help or impede mental wellbeing. It also states the mental health care should be based on respect for autonomy and self-determination, which are factors in up to 50% of the mental disorders amongst female inmates.Doyle et al. (2019)Behavioural cognitive therapy (9-12 months). Psycho-educational programmes. Motivational programmes.Eher et al. (2019)Alongside specific treatment for risk reduction, the psychological and social consequences of mental disorder should be considered in interventions to ensure effective and sustainable risk reduction. Fazel et al. (2016)The interventions to treat mental disorders in inmates include behavioural cognitive therapy, behavioural dialectic therapy and treatment based on meditation, in individual and group formats. Strategies to manage the risk of suicide include detection at reception, staff training, treatment, adequate supervision of inmates at risk, improved security of the physical setting and social support.Identification of mental problems.Suicide prevention strategies.Monitoring of substance dependence.Behavioural cognitive therapy.Methadone maintenance treatment.Gottfried et al. (2017)The importance of identifying strategies to help to improve service provision and more effectively treat mental illness, and the need to implement programmes designed to facilitate growth, maintain the treatment objectives and promote successful reintegration into society, would be of great help to this population. It is also necessary to implement programmes that bring about a successful transition of mentally ill inmates from the prison system to community-based mental health treatment services.Peters et al. (2017)Integrated treatment of dual disorders.Risk-need-response model.Behavioural cognitive therapy.Wills (2017)Mental health professionals working at prisons need to know the gender and age differences, the psychiatric record and background of the inmate, which can help in interviews with family and friends, the level of security assigned within the prison and the precautions required to prevent smuggling of drugs in prison. These authors consider interventions based on behavioural cognitive therapy to be highly effective.\n", "Analysis of the reports on the prevalence of mental disorders in the prison population showed a range of figures. In research carried out by Adraro et al.5, a prevalence of 62.7% for common mental disorders such as anxiety, depression and somatic symptom disorder was found in a group of 300 prison inmates, which is considered to be a high figure, in which three out of every five inmates are affected.\nIn another study by Baranyi et al.6, it was found that the prevalence of mental disorders was up to 15.8 times higher amongst inmates than they were in the general public, with a tendency to decrease with time spent in prison. The authors also commented that the inmates were often from a low socio-economic background, belonged to minority groups and had a history of child abuse and substance use, which made them vulnerable to psychiatric disorders. The poor living conditions, physical aggression and psychological abuse often encountered in prison can further aggravate mental disorders.\nAccording to Van Buitenen et al.7, the prevalence of schizophrenia was 56.7%; for substance abuse disorder, 43.1%; while up to 56.9% of the 5,247 inmates had other mental disorders, such as impulsiveness, poor social skills and disruptive behaviour.\nOn the other hand, a meta-analysis by Facer et al.8 showed that post-traumatic stress disorder (PTSD) was associated with a number of mental disorders such as depression and anxiety disorders. Inmates with PTSD were also significantly more likely to suffer from substance use disorder.\nA research project by Jakobowitz et al.9 showed that the prevalence of mental disorders amongst inmates was 8.65% for psychosis; 39.2% for depression; 45.8% for substance abuse; 41.4% for alcohol abuse; 17.8% for personality disorders; 8.1% for anxiety disorders and PTSD; and 5.1% for adaptation disorders. According to Forry et al.10 and Fovet et al.11, the most common mental disorders in the prison population are severe depression (44% and 31.2%, respectively), followed by generalised anxiety disorder (30.9 and 44.4%, respectively).\nFavril et al.12 describe a high prevalence of mental disorders in the prison population of New Zealand: up to 90.7% of the inmates present at least one mental disorder; substance abuse was present in 87% of the inmates; suicidal ideation was present in 36.4%; and suicide attempts in 55.6%.\nAnother study by Fazel et al.13 mentions that there is an important association between severe depression and psychotic illness (p<0.05), and states that one out of every seven inmates suffers from depression or psychosis. They also mention high rates of coincidence between mental illness and substance use, as well as suicide and self-harming, which are more common amongst inmates than the general public of the same age and sex. According to the authors, prison inmates with mental disorders are disproportionately involved in prison infractions and violent incidents, and are more likely to be accused of breaking prison rules and being injured in fights. Inmates with mental disorders and with a history of violence run an even higher risk in this regard, since psychiatric disorders are associated with violent incidents in prison (Table 2).\n\nTable 2Prevalence of mental disorders amongst inmates.Author/year PrevalenceAdraro et al. (2019)Common mental disorders: 62.7% (CI 95%: 65.7-68.5).Baranyi et al. (2019)Psychosis: 6.2%.Severe depression: 3.8%.Alcohol use: 3.8%.Substance use: 5.1%.Eher et al. (2019)Mental disorder: 92.9%.Alcohol abuse: 40%.Paraphilias: 43.3%.Personality disorders: 53.6%.Type B personality disorders: 47.8%.Favril et al. (2020)Substance abuse: 87%.Any mental disorder: 90.7%.PTSD: 23.9%.Panic disorder: 5.6%.Generalised anxiety disorder: 8.7%.Bipolar disorder: 11.1%.Suicidal ideas: 36.4%.Attempted suicide: 55.6%.Forry et al. (2019)Severe depression: 44%.Antisocial personality disorder: 20.5%.Bipolar disorder: 23.5%.Generalised anxiety disorder: 30.9%.Panic disorder: 32.6%.Substance abuse: 12.5%.Fovet et al. (2020)Affective disorders: 31.2%.Anxiety disorders: 44.4%.Panic disorders: 13%.Substance abuse: 53.5%.Risk of suicide: 31.4%.Jakobowitz et al. (2017)Psychosis: 8.65%.Depression: 39.2%.Substance abuse: 45.8%.Alcohol abuse: 41.4%.Personality disorder: 17.8%.Anxiety: 8.1%.PTSD: 8.1%.Adaptation disorder: 5.1%.Van Buitenen et al. (2020)Schizophrenia: 56.7%. Substance use: 43.1%.Co-morbid disorders (impulsiveness. poor social skills, disruptive behaviour): 56.9%.Note. CI: confidence interval; PTSD: post-traumatic stress disorder.\n\nNote. CI: confidence interval; PTSD: post-traumatic stress disorder.", "Adraro et al.5 consider that early examination of inmates when they arrive at prison is necessary, as are adequate treatment and monitoring. Jobs should also be created in prison to enable inmates to come together and develop positive relationships for social support and promote coping skills. Van Buitenen et al.7, consider that there is a possible causal relationship between psychopathology and criminal conduct, which should be borne in mind when developing treatment programmes for this sector of the population.\nAccording to Bartlet and Hollins14, mental health management of female inmates should be based on a system with sufficient resources to permit effective screening, individualised healthcare planning and primary and specialised medical care.\nA systematic review by Doyle et al.15 on treatment approaches for abuse of alcohol and other drugs amongst inmates, established that treatment is provided in different formats, including personalised consultations, group sessions, therapeutic communities and residential treatment programmes. Furthermore, the researchers observed that cognitive behavioural therapy of between 9 and 12 months is the most effective approach for treating inmates with this problem.\nAccording to Jakobowitz et al.9, there are specific and serious problems in psychiatric and psychological treatment of prison inmates. The limited availability of adequate premises often interrupts the continuity needed for care. Inmates can be released with no prior notice by the courts or transferred to other prisons, which is highly detrimental when planning subsequent treatment. Short periods of imprisonment are particularly likely to cause problems for mental health treatment, and can make access to prison psychiatric services difficult.\nForry et al.10 also report a high prevalence of mental disorders in prisons, and state that it is necessary to develop the skills of health workers and other prison staff in detecting, evaluating and treating inmates with mental disorders. There is also the need for a clear referral process for person with such disorders, while improving the living conditions and standards of prison inmates would help to prevent mental disorders and their associated co-morbidities.\nFazel et al.13 recommend that all prisons should have an identification system to identify persons with serious mental health problems, including case tracking systems when entering prison and assignment of adequate service levels. All prisons should also have a suicide prevention strategy that includes detection and close monitoring of risks, multi-disciplinary management of high-risk patients and staff training13.\nGottfried et al.16 consider that mental health treatment needs for prison inmates are a vital concern, and highlight the importance of identifying strategies that can help to improve service provision and treat mental disorders more effectively.\nFor Wilton and Stewart17, psychiatric services in prisons should detect substance use disorders, and if they are present, they should ensure that treatment is provided to improve the living conditions of the population, since this appears to be the key factor in contributing towards worse outcomes for inmates with mental disorders.\nAccording to Wills18, mental health professionals in prisons need to know the gender and age differences, along with the psychiatric history and background of inmates, to enable them to benefit from interviews with family members and friends, and to establish the security levels within the prison and the precautions necessary to prevent drug trafficking in prison. The author also considers that interventions based on cognitive behavioural therapy are very effective for prison inmates.\nPeters et al.19 consider that some mental health treatment and behavioural strategies in prisons should include integrated treatment of dual disorders, the risk-need-response model and cognitive behavioural therapy, and comment that the use of such strategies in the design of behavioural health services can significantly reduce criminal behaviours.\nIn the case of substance use disorders, the aim of treatment in prison is to provide effective short-term services (1-3 months) to manage acute symptoms. The initial phases of treatment programmes include detoxification, psychiatric consultation to establish a psychotropic medication regimen, comprehensive evaluation and motivational strategies to involve inmates in treatment19.\nFor Eher et al.20, the presence of a mental disorder is an important risk factor for criminal behaviour, not only for sexual crimes but for any type of infraction. Furthermore, existing disorders can worsen or new disorders may appear over the course of the inmate’s prison sentence. For this reason the authors consider that alongside a specific treatment to reduce risks, sex offenders with a mental disorder also often need interventions to manage the psychological and social consequences of this kind of disorder (Table 3).\n\nTable 3Therapeutic interventions for mental disorders amongst inmates .Author/year Therapeutic interventions Adraro et al. (2019)An early examination of inmates should be carried out when they arrive, with adequate treatment and follow up. Jobs should also be created that can bring inmates together and help to develop positive relationships, thus creating social support and helping with coping strategies. Bartlett et al. (2018)The management of female inmates' health should be based on a system with sufficient resources to enable screening, planning of individualised care and primary and specialised medical treatment, which makes it necessary to pat closer attention to the gender dimensions of women's previous experience, the nature of prison and the presence or absence of specific gender factors that help or impede mental wellbeing. It also states the mental health care should be based on respect for autonomy and self-determination, which are factors in up to 50% of the mental disorders amongst female inmates.Doyle et al. (2019)Behavioural cognitive therapy (9-12 months). Psycho-educational programmes. Motivational programmes.Eher et al. (2019)Alongside specific treatment for risk reduction, the psychological and social consequences of mental disorder should be considered in interventions to ensure effective and sustainable risk reduction. Fazel et al. (2016)The interventions to treat mental disorders in inmates include behavioural cognitive therapy, behavioural dialectic therapy and treatment based on meditation, in individual and group formats. Strategies to manage the risk of suicide include detection at reception, staff training, treatment, adequate supervision of inmates at risk, improved security of the physical setting and social support.Identification of mental problems.Suicide prevention strategies.Monitoring of substance dependence.Behavioural cognitive therapy.Methadone maintenance treatment.Gottfried et al. (2017)The importance of identifying strategies to help to improve service provision and more effectively treat mental illness, and the need to implement programmes designed to facilitate growth, maintain the treatment objectives and promote successful reintegration into society, would be of great help to this population. It is also necessary to implement programmes that bring about a successful transition of mentally ill inmates from the prison system to community-based mental health treatment services.Peters et al. (2017)Integrated treatment of dual disorders.Risk-need-response model.Behavioural cognitive therapy.Wills (2017)Mental health professionals working at prisons need to know the gender and age differences, the psychiatric record and background of the inmate, which can help in interviews with family and friends, the level of security assigned within the prison and the precautions required to prevent smuggling of drugs in prison. These authors consider interventions based on behavioural cognitive therapy to be highly effective.\n", "Several studies highlight the fact that the incidence of mental disorders in the prison population is higher than in the general public. The focal points of these studies include the underlying conditions of prison inmates, the fact that care for such populations may be an indicator of the willingness of society and the health system to extend medical care to vulnerable populations and the possibility that untreated mental disorders can be a risk factor for recidivism21.\nThe results obtained in this study coincide with this focus. What is more, almost all the authors consulted agree that the prevalence of mental disorders in the prison population - of whatever age and gender - is high, with a predominance of depressive disorders, anxiety, substance use and psychotic disorders6-11,13,20,21.\nThese results match those described by Gabrysh et al.22, who, in an analysis of the Chilean prison population, found that 64% of inmates had mental disorders when entering prison, with a predominance of depressive disorders (30%). However, the researchers also described a drop in the prevalence of such disorders after three years imprisonment, especially amongst those cases who participated in educational or occupational activities in prison.\nThe results published by Constantino et al.23 in a study in Rio de Janeiro (Brazil) on inmates likewise coincide in this regard. The study found a high prevalence of stress (35.8% in men and 57% in women), along with moderate and severe depressive symptoms (31.1% in men and 47.1% in women), which highlights the high level of psychopathological co-morbidity in the prison population and the links with humiliation and mistreatment.\nHigh levels of other mental disorders and problematic behaviours in the prison population have also been documented. The relationship between PTSD and these results is not clearly understood and may be perpetuating the under-diagnosis and poor treatment of PTSD in prisons24,25.\nInmates convicted of violent crimes are more likely to have at least one mental disorder, such as severe depression. Inmates diagnosed with a psychotic disorder are more likely to have committed a violent crime26.\nGates et al.27 also consider that there is a strong link between mental disorders and self-harming. This type of disorder is associated with suicidal ideation, attempted suicide and death from suicide, as well with substance use disorders, which significantly increase the risk of suicide, which may also support the results obtained in this review.\nIn this context, the current evidence indicates that inmates sentenced to full internment suffer significantly higher levels of depression, alcohol abuse and illegal substance use, than those sentenced to open regimes, which suggests that the association between imprisonment and mental health can vary substantially between centres; it also highlights the importance of opening up the research to go beyond the study of prisons28.\nIn this regard, it is accepted that the loss of liberty, autonomy and communication with family and friends has a major physical, social and psychological impact on the prison population, and makes the magnitude of mental and behavioural disorders in this sector of the population a significantly higher one. Furthermore, whatever mental disorders existed prior to sentencing greatly worsen with imprisonment29.\nA population group that merits special attention in this regard are inmates over 65 years of age, who appear to be especially vulnerable to mental and behavioural disorders, which are often masked by the development of neurodegenerative diseases, such as dementia or cerebrovascular diseases. Therefore, elderly inmates should be regarded as a high risk population and receive more frequent psychological and psychiatric care30,31.\nWhen analysing the psychiatric treatment strategies in the prison setting, a match of criteria was found amongst the authors in terms of the importance of having mental health programme in prison institutions, which should diagnose inmates as soon as they arrive and personalise interventions according to the type of disorder that is detected, their background, the risk of developing mental disorders, or aggravating factors related to the characteristics of the prison, and the type and length of sentence. Many authors recommend the use of behavioural cognitive therapy and pharmacological treatment when necessary13,15,18,19.\nThese results highlight the need to design pragmatic intervention programmes that have the potential to improve access to mental health services in prisons, which are often riddled with delays or are inadequate. They also highlight the lack of psychiatric professionals trained to treat and prevent mental disorders in the prison setting, which means that providing incentives for academic and professional training in this field would be a positive step, since the population in question has considerable mental health needs during their sentence and reinsertion into society.\nThese proposals are supported by the results of Hopkin et al.32, who stated that insurance coverage and contact with mental health professionals and other services can be improved through interventions in the period of transit from prison to the community.\nThe need to improve access to mental health in the prison system was also highlighted by Martin et al.33, who found that although mental health care was provided when inmates entered prison, only 8% of them received follow ups up to halfway through their sentence, while most inmates with mental disorders prior to sentencing received psychiatric care only up to 10% of their sentence. This means it is necessary to establish more effective mental health policies in prisons that can contribute towards better rehabilitation of inmates and reduce recidivism.\nThis study found that the qualitative design for a theoretical review can be a limitation, since there is no longer an evaluation of the quality of the evidence or the results, which makes it advisable to carry out a quantitative analysis (meta-analysis) of the information in future research studies." ]
[ "intro", "materials|methods", "results", null, null, "discussion" ]
[ "anxiety", "depression", "psychotic disorder", "prisoners", "prisons", "ansiedad", "depresión", "trastornos psicóticos", "prisioneros", "prisiones." ]
The Incidence Trend of Papillary Thyroid Carcinoma in the United States During 2003-2017.
36256588
The rapid increase in the detection rate of thyroid cancer over the past few decades has caused some unexpected economic burdens. However, that of papillary thyroid carcinoma (PTC) seems to have had the opposite trend, which is worthy of further comprehensive exploration.
BACKGROUND
The Surveillance, Epidemiology, and End Results 18 database was used to identify patients with PTC diagnosed during 2003-2017. The incidence trends were analyzed using joinpoint analysis and an age-period-cohort model.
METHODS
The overall PTC incidence rate increased from 9.9 to 16.1 per 100 000 between 2003 and 2017. The joinpoint analysis indicated that the incidence growth rate began to slow down in 2009 (annual percentage change [APC] = 3.1%, 95% confidence interval [CI] = 1.9%-4.4%). After reaching its peak in 2015, it began to decrease by 2.8% (95% CI = -4.6% to -1.0%) per year. The stratified analysis indicated that the incidence patterns of different sexes, age groups, races, and tumor stages and sizes had similar downward trends, including for the localized (APC = -4.5%, 95% CI = -7% to -1.9%) and distant (APC = -1.3%, 95% CI = -2.7% to -.1%) stages, and larger tumors (APC = -4%, 95% CI = -12% to 4.7%). The age-period-cohort model indicated a significant period effect on PTC, which gradually weakened after 2008-2012. The cohort effect indicates that the risk of late birth cohorts is gradually stabilizing and lower than that of early birth cohorts.
RESULTS
The analysis results of the recent downward trend and period effect for the incidence of each subgroup further support the important role of correcting overdiagnosis in reducing the prevalence of PTC. Future research needs to analyze more-recent data to verify these downward trends.
CONCLUSION
[ "Humans", "United States", "Thyroid Cancer, Papillary", "Incidence", "SEER Program", "Thyroid Neoplasms", "Racial Groups" ]
9583193
Introduction
The incidence of thyroid cancer (TC) has been increasing in recent decades, especially among females.1,2 Papillary thyroid carcinoma (PTC) is the most common thyroid cancer, accounting for more than 90% of cases.3,4 Previous studies have found that almost all worsened diagnoses are attributable to increases in PTC.5 The global incidence of TC is increasing much faster than other malignancies due to the widespread use of well-diagnosed thyroid ultrasonography and needle biopsy.4,6 This trend is particularly striking in the United States, where the PTC incidence in females increased from 4.6 to 17.9 per 100 000 between 1975 and 2017.3 Many researchers believe that the significant increase in TC incidence is due to overdiagnosis.6,7 Others believe that the important underlying factors include obesity and increased exposure to ionizing radiation.8,9 The sharp increase in TC incidence was recently observed to have slowed down and even reversed, especially in small TC tumors.10 Whether this transition is common and its reason are worthy of further investigation. Therefore, in order to systematically and comprehensively understand the latest evolutions in PTC, this study examined the period trend of PTC incidence according to age, sex, race, tumor size, and tumor stage. Based on the age-period-cohort model, we evaluated the effects of age, period, and birth cohort on time trends, which can provide clues for understanding the role of risk and protective factors in the etiology of PTC.
null
null
Results
The ASIRs of PTC overall and by sex during 2003-2017 are shown in Figure 1. The overall PTC incidence increased from 9.9 to 16.1 per 100 000 between 2003 and 2017. The joinpoint analysis indicated that the growth rate was the fastest before 2009, when it increased by 7.6% (95% CI = 6.9%–8.3%) per year, and then decreased by 3.1%. After reaching its peak in 2015, it began to decrease by 2.8% per year (Table 1). The incidence for female patients continued to increase from 2003 to 2015, with the fastest increase rate of 7.9% (95% CI = 7.1%–8.7%) per year from 2003 to 2009, which then slowed to 2.4% (95% CI = 1.5%–3.4%) during 2009-2015. Similarly, the incidence began to decline at a rate of 5.5% (95% CI = −9.4% to −1.4%) after 2015. Among male patients, the increase during 2003-2012 was comparable to that in female patients (APC = 6.5%, 95% CI = 5.7%–7.3%) (Table 1), with a stable and declining trend in subsequent years (Figure 1).Figure 1.Trends in the incidence rate of PTC by tumor sex. Rates are age adjusted to the 2000 US standard population.Table 1.Annual Percentage Change in Incidence of Papillary Thyroid Carcinoma, 2003-2017.Trend 1Trend 2Trend 3YearsAPC(95%CI)YearsAPC(95%CI)YearsAPC(95%CI)Overall2003-20097.6a(6.9,8.3)2009-20153.1a(1.9,4.4)2015-2017−2.8a(−4.6,−1)Sex Male2003-20126.5a(5.7,7.3)2012-2017−.1 (−1.8,1.8) Female2003-20097.9a(7.1,8.7)2009-20152.4a(1.5,3.4)2015-2017−5.5a(-9.4,-1.4)Age 20-242003-20154.4a(2.9,6)2015-2017−4.4 (−25.5,22.6) 25-292003-20154.1a(3.3,5)2015-2017−6.7 (−19,7.5) 30-342003-20087.3a(3.8,11)2008-20153.5a(1,6.2)2015-2017−5.7 (−18.8,9.6) 35-392003-20135.7a(4.7,6.7)2013-2017−1.8 (−5.5,1.9) 40-442003-20117.4a(6.1,8.6)2011-2017−.1 (−1.8,1.8) 45-492003-20069.0a(6.2,11.8)2006-20135.0a(4.1,5.9)2013-2017−2.8a(−4.3,−1.2) 50-542003-20108.2a(7.1,9.2)2010-20151.3 (−1.1,3.7)2015-2017−4 (−11,3.4) 55-592003-20098.2a(6.9,9.5)2009-20153.0a(1.4,4.7)2015-2017−6.2 (−12.8,0.9) 60-642003-20099.7a(6.6,12.8)2009-2017.8 (−1.1,2.6) 65-692003-20118.4a(6.9,10.1)2011-2017−1.2 (−3.4,1.1) 70-742003-20099.6a(8,11.3)2009-20132.3 (−2.2,7.1)2013-2017−4.4a(−7.1,−1.6) 75-792003-20117.3a(5.4,9.2)2011-2017−.9 (−3.6,1.8) 80-842003-20125.3a(2.6,8)2012-2017−1.3 (−7.3,5.2)Race White2003-20097.6a(6.2,9)2009-20153.0a(1.8,4.3)2015-2017−6.1a(−11.1,-.8) Black2003-20156.0a(5.4,6.5)2015-2017−14.6a(−20.9,−7.9) American Indian2003-20128.3a(4.7,12)2012-2017−.8 (−6.7,5.5) Asian2003-20107.1a(5.2,9.1)2010-20171 (−.4,2.4)Stage Localized2003-20097.2a(6.2,8.2)2009-20142.9a(1.1,4.6)2014-2017−4.5a(−7,−1.9) Regional2003-200910.2a(9.2,11.2)2009-20153.7a(2.4,4.9)2015-2017−1.2 (−6.5,4.4) Distant2003-2017−1.3 (−2.7,0.1)Size <1 cm2004-200910.7a(7.8,13.7)2009-20134 (−.2,8.3)2013-2017−3.8a(−6.3,−1.2) 1-2.9 cm2004-20098.5a(6.8,10.3)2009-20152.6a(1.4,3.9)2015-2017−3 (−7.8,2) ≥3 cm2004-20105.1a(3.1,7.2)2010-20153.9a(1,6.9)2015-2017−4 (−12,4.7)APC: annual percent change; CI: confidence interval.aThe value is statistically different from zero (2‐sided P < .05) based on joinpoint regression analysis. Trends in the incidence rate of PTC by tumor sex. Rates are age adjusted to the 2000 US standard population. Annual Percentage Change in Incidence of Papillary Thyroid Carcinoma, 2003-2017. APC: annual percent change; CI: confidence interval. aThe value is statistically different from zero (2‐sided P < .05) based on joinpoint regression analysis. The age-specific incidence curve is shown in Figure 2. During the study period, the incidence rates were lower for those aged <30 and 80-84 years than in the other age groups. Overall, before about 2009, those aged 30-34 and 40-79 years had the fastest growth rates, with an annual growth rate of more than 7%, of which those aged 60-64 and 70-74 years had a rate of nearly 10%. Each age group had a varying degree of decline after 2013 or 2015, although some APCs were not statistically significant (Table 1).Figure 2.Age-specific incidence trends of PTC. Age-specific incidence trends of PTC. Incidence trends were similar for whites and Asian Americans, who had consistently higher rates than blacks and American Indians (Figure 3). The incidence for whites increased by 7.6% (95% CI = 6.2-9.0%) per year during 2003-2009. The rate of increase slowed slightly during 2009-2015 (APC = 3.0%, 95% CI = 1.8%–4.3%) (Figure 3 and Table 1). Likewise, the incidence among whites declined at a rate of 6.1% (95% CI = −11.1% to −.8%) between 2015 and 2017. Asian Americans also had a fluctuating slight decline of 1% (95% CI = −.4% to −2.4%) during the most-recent period. The incidence for blacks declined significantly by 14.6% (95% CI = −20.9% to −7.9%) per year after 2015.Figure 3.Trends in the incidence rate of PTC by race. Rates are age adjusted to the 2000 US standard population. Trends in the incidence rate of PTC by race. Rates are age adjusted to the 2000 US standard population. PTC incidence curves and APCs by stage are presented in Figure 4 and Table 1. There was a significant difference in incidence among the three stages, with the highest and lowest in the localized and distant stages, respectively. The incidence rates for patients in the localized and regional stages increased rapidly at rates of 7.2% and 10.2% before 2009, respectively, and by 2.9% and 3.7% each year during 2009-2015. The incidence rates for patients in both stages decreased after 2015, with those in the localized stage having the greatest decrease (APC = −4.5%, 95% CI = −7% to −1.9%). The incidence for distant-stage patients has been stable for the past 15 years, and a slight downward trend was also recently observed (APC = −1.3% to 95% CI = −2.7% to .1%).Figure 4.Trends in the incidence rate of PTC by tumor size. Rates are age adjusted to the 2000 US standard population. Trends in the incidence rate of PTC by tumor size. Rates are age adjusted to the 2000 US standard population. We also examined the trend of PTC incidence according to tumor size (Figure 5). The incidence of cancers of all sizes had the fastest growth rate during 2004-2009, with smaller tumors having faster rates (Table 1). After 2009-2010, the incidence growth rate of three sizes of PTC slowed. We found that the incidence of tumors smaller than 1 cm was reversed from 2013, decreasing by 3.8% per year. Similarly, tumor of 1-2.9 cm and larger had downward trends after 2015 (3% [95% CI = −7.8% to 2%] and 4% [95% CI = −12% to 4.7%]).Figure 5.Trends in the incidence rate of PTC by stage. Rates are age adjusted to the 2000 US standard population. Trends in the incidence rate of PTC by stage. Rates are age adjusted to the 2000 US standard population. The age-period-cohort analysis revealed period and cohort effects on PTC incidence trends (Figure 6). There was a significant period effect during the study period. The effect size increased rapidly before 2008-2012, after which it slowed. This indicates that the rate of increase in risk has slowed down after 2008-2012, which was consistent with the results of the joinpoint analysis. Cohort effects and period effects had opposite trends. The cohort curve had a slight downward trend. Taking the 1960 birth cohort as a reference, the risk of patients born before was slightly higher, and the risk of the subsequent birth cohort gradually decreased and leveled off.Figure 6.Period and cohort effects in the incidence of papillary thyroid carcinoma. Period and cohort effects in the incidence of papillary thyroid carcinoma.
Conclusions
In conclusion, the present analysis has revealed that the PTC incidence, which has been increasing rapidly, has recently slowed down and even decreased, including for tumors that are larger and of distant stage. This further supports the importance of overdiagnosis regarding health policymakers, physicians, and patients, the revision of thyroid management guidelines, and the reclassification of low-risk indolent TC as significant contributors to the decreased PTC detection rate. The results for the period effect also suggest that changes in PCT incidence are closely related to overdiagnosis. Future studies will need to analyze more-recent data to confirm these trends.
[ "Data Source", "Statistical Analysis" ]
[ "First PTC diagnoses during 2003-2017 were selected from the Surveillance,\nEpidemiology, and End Results (SEER) 18 database, which contains data from 18\ncancer registries covering approximately 35% of the United States population.\nMulticenter, high-quality, public data provide reliable evidence support and\nfirst-hand information for evidence-based practice by clinicians and cancer\nresearchers.11,12 PTC cases were defined using International\nClassification of Diseases for Oncology (third edition) histological codes 8050,\n8052, 8130, 8260, 8340-8344, 8450, and 8452, and primary site code C73.9. Age at\ndiagnosis (20-84 years), race (white, black, American Indian/Alaska Native,\nAsian Americans, or Pacific Islander), sex (male, female), tumor size (<1 cm,\n1-2.9 cm, ≥3 cm) and SEER summary stage (localized, regional, and distant) were\nincluded in this study.\nAll the steps in this study follow the principles outlined in the 1964 Helsinki\nDeclaration and its subsequent amendments. This study was exempted the informed\nconsent and review approval by the Institutional Research Committee of School of\nBasic Medicine and Public Health of Jinan University, because the SEER data were\npublic and all patient information was de-identified.", "The SEER*Stat program of the National Cancer Institute was used to calculate\nage-standardized incidence rates (ASIR; standardized to the United States\npopulation in 2000) and 95% confidence intervals (CIs). ASIR was stratified by\nsex, age, race, and tumor stage and size.\nJoinpoint regression software was developed by the National Cancer Institute\n(NCI) and is used to capture changes in cancer trends.13 The Joinpoint regression\nsoftware also allows to find statistically best fits the trend data, utilizing\nwith the minimum number of joinpoints to the model with maximum number of\njoinpoints. Joinpoint regression analysis was used to describe the continuous\nincidence trend.14 At each optimal fitting point, the direction of the\nASIR trend was observed. The annual percentage change (APC) was the change rate\nat each interval. A two-sided probability value of P < .05\nwas considered statistically significant. Joinpoint regression software version\n4.9.1.0 was used in this study.\nThe age-period-cohort model was fitted to 5-year bands according to ages (20-24,\n25-29, …, 75-79, 80-84 years), periods (2003-2007, 2008-2012, 2013-2017), and\nbirth cohorts (1923-1927, 1928-1932, …, 1988-1992, 1993-1997). In this study,\nthe intrinsic estimator algorithm is used to solve the unrecognizable problem of\nage, period and cohort collinearity. The algorithm has been confirmed to be\nestimable, unbiased, and asymptotic.15 The Akaike information\ncriterion and Bayesian information criterion were used to test the goodness of\nfit of the model. These statistical analyses were performed using Stata\nsoftware." ]
[ null, null ]
[ "Introduction", "Materials and Methods", "Data Source", "Statistical Analysis", "Results", "Discussion", "Conclusions" ]
[ "The incidence of thyroid cancer (TC) has been increasing in recent decades,\nespecially among females.1,2\nPapillary thyroid carcinoma (PTC) is the most common thyroid cancer, accounting for\nmore than 90% of cases.3,4\nPrevious studies have found that almost all worsened diagnoses are attributable to\nincreases in PTC.5 The global incidence of TC is increasing much faster than other\nmalignancies due to the widespread use of well-diagnosed thyroid ultrasonography and\nneedle biopsy.4,6\nThis trend is particularly striking in the United States, where the PTC incidence in\nfemales increased from 4.6 to 17.9 per 100 000 between 1975 and 2017.3\nMany researchers believe that the significant increase in TC incidence is due to\noverdiagnosis.6,7\nOthers believe that the important underlying factors include obesity and increased\nexposure to ionizing radiation.8,9 The sharp increase in TC\nincidence was recently observed to have slowed down and even reversed, especially in\nsmall TC tumors.10 Whether this transition is common and its reason are worthy of\nfurther investigation.\nTherefore, in order to systematically and comprehensively understand the latest\nevolutions in PTC, this study examined the period trend of PTC incidence according\nto age, sex, race, tumor size, and tumor stage. Based on the age-period-cohort\nmodel, we evaluated the effects of age, period, and birth cohort on time trends,\nwhich can provide clues for understanding the role of risk and protective factors in\nthe etiology of PTC.", "[SUBTITLE] Data Source [SUBSECTION] First PTC diagnoses during 2003-2017 were selected from the Surveillance,\nEpidemiology, and End Results (SEER) 18 database, which contains data from 18\ncancer registries covering approximately 35% of the United States population.\nMulticenter, high-quality, public data provide reliable evidence support and\nfirst-hand information for evidence-based practice by clinicians and cancer\nresearchers.11,12 PTC cases were defined using International\nClassification of Diseases for Oncology (third edition) histological codes 8050,\n8052, 8130, 8260, 8340-8344, 8450, and 8452, and primary site code C73.9. Age at\ndiagnosis (20-84 years), race (white, black, American Indian/Alaska Native,\nAsian Americans, or Pacific Islander), sex (male, female), tumor size (<1 cm,\n1-2.9 cm, ≥3 cm) and SEER summary stage (localized, regional, and distant) were\nincluded in this study.\nAll the steps in this study follow the principles outlined in the 1964 Helsinki\nDeclaration and its subsequent amendments. This study was exempted the informed\nconsent and review approval by the Institutional Research Committee of School of\nBasic Medicine and Public Health of Jinan University, because the SEER data were\npublic and all patient information was de-identified.\nFirst PTC diagnoses during 2003-2017 were selected from the Surveillance,\nEpidemiology, and End Results (SEER) 18 database, which contains data from 18\ncancer registries covering approximately 35% of the United States population.\nMulticenter, high-quality, public data provide reliable evidence support and\nfirst-hand information for evidence-based practice by clinicians and cancer\nresearchers.11,12 PTC cases were defined using International\nClassification of Diseases for Oncology (third edition) histological codes 8050,\n8052, 8130, 8260, 8340-8344, 8450, and 8452, and primary site code C73.9. Age at\ndiagnosis (20-84 years), race (white, black, American Indian/Alaska Native,\nAsian Americans, or Pacific Islander), sex (male, female), tumor size (<1 cm,\n1-2.9 cm, ≥3 cm) and SEER summary stage (localized, regional, and distant) were\nincluded in this study.\nAll the steps in this study follow the principles outlined in the 1964 Helsinki\nDeclaration and its subsequent amendments. This study was exempted the informed\nconsent and review approval by the Institutional Research Committee of School of\nBasic Medicine and Public Health of Jinan University, because the SEER data were\npublic and all patient information was de-identified.\n[SUBTITLE] Statistical Analysis [SUBSECTION] The SEER*Stat program of the National Cancer Institute was used to calculate\nage-standardized incidence rates (ASIR; standardized to the United States\npopulation in 2000) and 95% confidence intervals (CIs). ASIR was stratified by\nsex, age, race, and tumor stage and size.\nJoinpoint regression software was developed by the National Cancer Institute\n(NCI) and is used to capture changes in cancer trends.13 The Joinpoint regression\nsoftware also allows to find statistically best fits the trend data, utilizing\nwith the minimum number of joinpoints to the model with maximum number of\njoinpoints. Joinpoint regression analysis was used to describe the continuous\nincidence trend.14 At each optimal fitting point, the direction of the\nASIR trend was observed. The annual percentage change (APC) was the change rate\nat each interval. A two-sided probability value of P < .05\nwas considered statistically significant. Joinpoint regression software version\n4.9.1.0 was used in this study.\nThe age-period-cohort model was fitted to 5-year bands according to ages (20-24,\n25-29, …, 75-79, 80-84 years), periods (2003-2007, 2008-2012, 2013-2017), and\nbirth cohorts (1923-1927, 1928-1932, …, 1988-1992, 1993-1997). In this study,\nthe intrinsic estimator algorithm is used to solve the unrecognizable problem of\nage, period and cohort collinearity. The algorithm has been confirmed to be\nestimable, unbiased, and asymptotic.15 The Akaike information\ncriterion and Bayesian information criterion were used to test the goodness of\nfit of the model. These statistical analyses were performed using Stata\nsoftware.\nThe SEER*Stat program of the National Cancer Institute was used to calculate\nage-standardized incidence rates (ASIR; standardized to the United States\npopulation in 2000) and 95% confidence intervals (CIs). ASIR was stratified by\nsex, age, race, and tumor stage and size.\nJoinpoint regression software was developed by the National Cancer Institute\n(NCI) and is used to capture changes in cancer trends.13 The Joinpoint regression\nsoftware also allows to find statistically best fits the trend data, utilizing\nwith the minimum number of joinpoints to the model with maximum number of\njoinpoints. Joinpoint regression analysis was used to describe the continuous\nincidence trend.14 At each optimal fitting point, the direction of the\nASIR trend was observed. The annual percentage change (APC) was the change rate\nat each interval. A two-sided probability value of P < .05\nwas considered statistically significant. Joinpoint regression software version\n4.9.1.0 was used in this study.\nThe age-period-cohort model was fitted to 5-year bands according to ages (20-24,\n25-29, …, 75-79, 80-84 years), periods (2003-2007, 2008-2012, 2013-2017), and\nbirth cohorts (1923-1927, 1928-1932, …, 1988-1992, 1993-1997). In this study,\nthe intrinsic estimator algorithm is used to solve the unrecognizable problem of\nage, period and cohort collinearity. The algorithm has been confirmed to be\nestimable, unbiased, and asymptotic.15 The Akaike information\ncriterion and Bayesian information criterion were used to test the goodness of\nfit of the model. These statistical analyses were performed using Stata\nsoftware.", "First PTC diagnoses during 2003-2017 were selected from the Surveillance,\nEpidemiology, and End Results (SEER) 18 database, which contains data from 18\ncancer registries covering approximately 35% of the United States population.\nMulticenter, high-quality, public data provide reliable evidence support and\nfirst-hand information for evidence-based practice by clinicians and cancer\nresearchers.11,12 PTC cases were defined using International\nClassification of Diseases for Oncology (third edition) histological codes 8050,\n8052, 8130, 8260, 8340-8344, 8450, and 8452, and primary site code C73.9. Age at\ndiagnosis (20-84 years), race (white, black, American Indian/Alaska Native,\nAsian Americans, or Pacific Islander), sex (male, female), tumor size (<1 cm,\n1-2.9 cm, ≥3 cm) and SEER summary stage (localized, regional, and distant) were\nincluded in this study.\nAll the steps in this study follow the principles outlined in the 1964 Helsinki\nDeclaration and its subsequent amendments. This study was exempted the informed\nconsent and review approval by the Institutional Research Committee of School of\nBasic Medicine and Public Health of Jinan University, because the SEER data were\npublic and all patient information was de-identified.", "The SEER*Stat program of the National Cancer Institute was used to calculate\nage-standardized incidence rates (ASIR; standardized to the United States\npopulation in 2000) and 95% confidence intervals (CIs). ASIR was stratified by\nsex, age, race, and tumor stage and size.\nJoinpoint regression software was developed by the National Cancer Institute\n(NCI) and is used to capture changes in cancer trends.13 The Joinpoint regression\nsoftware also allows to find statistically best fits the trend data, utilizing\nwith the minimum number of joinpoints to the model with maximum number of\njoinpoints. Joinpoint regression analysis was used to describe the continuous\nincidence trend.14 At each optimal fitting point, the direction of the\nASIR trend was observed. The annual percentage change (APC) was the change rate\nat each interval. A two-sided probability value of P < .05\nwas considered statistically significant. Joinpoint regression software version\n4.9.1.0 was used in this study.\nThe age-period-cohort model was fitted to 5-year bands according to ages (20-24,\n25-29, …, 75-79, 80-84 years), periods (2003-2007, 2008-2012, 2013-2017), and\nbirth cohorts (1923-1927, 1928-1932, …, 1988-1992, 1993-1997). In this study,\nthe intrinsic estimator algorithm is used to solve the unrecognizable problem of\nage, period and cohort collinearity. The algorithm has been confirmed to be\nestimable, unbiased, and asymptotic.15 The Akaike information\ncriterion and Bayesian information criterion were used to test the goodness of\nfit of the model. These statistical analyses were performed using Stata\nsoftware.", "The ASIRs of PTC overall and by sex during 2003-2017 are shown in Figure 1. The overall PTC\nincidence increased from 9.9 to 16.1 per 100 000 between 2003 and 2017. The\njoinpoint analysis indicated that the growth rate was the fastest before 2009, when\nit increased by 7.6% (95% CI = 6.9%–8.3%) per year, and then decreased by 3.1%.\nAfter reaching its peak in 2015, it began to decrease by 2.8% per year (Table 1). The incidence\nfor female patients continued to increase from 2003 to 2015, with the fastest\nincrease rate of 7.9% (95% CI = 7.1%–8.7%) per year from 2003 to 2009, which then\nslowed to 2.4% (95% CI = 1.5%–3.4%) during 2009-2015. Similarly, the incidence began\nto decline at a rate of 5.5% (95% CI = −9.4% to −1.4%) after 2015. Among male\npatients, the increase during 2003-2012 was comparable to that in female patients\n(APC = 6.5%, 95% CI = 5.7%–7.3%) (Table 1), with a stable and declining\ntrend in subsequent years (Figure\n1).Figure 1.Trends in the incidence rate of PTC by tumor sex. Rates are age adjusted\nto the 2000 US standard population.Table 1.Annual Percentage Change in Incidence of Papillary Thyroid Carcinoma,\n2003-2017.Trend 1Trend 2Trend 3YearsAPC(95%CI)YearsAPC(95%CI)YearsAPC(95%CI)Overall2003-20097.6a(6.9,8.3)2009-20153.1a(1.9,4.4)2015-2017−2.8a(−4.6,−1)Sex Male2003-20126.5a(5.7,7.3)2012-2017−.1 (−1.8,1.8) Female2003-20097.9a(7.1,8.7)2009-20152.4a(1.5,3.4)2015-2017−5.5a(-9.4,-1.4)Age 20-242003-20154.4a(2.9,6)2015-2017−4.4 (−25.5,22.6) 25-292003-20154.1a(3.3,5)2015-2017−6.7 (−19,7.5) 30-342003-20087.3a(3.8,11)2008-20153.5a(1,6.2)2015-2017−5.7 (−18.8,9.6) 35-392003-20135.7a(4.7,6.7)2013-2017−1.8 (−5.5,1.9) 40-442003-20117.4a(6.1,8.6)2011-2017−.1 (−1.8,1.8) 45-492003-20069.0a(6.2,11.8)2006-20135.0a(4.1,5.9)2013-2017−2.8a(−4.3,−1.2) 50-542003-20108.2a(7.1,9.2)2010-20151.3 (−1.1,3.7)2015-2017−4 (−11,3.4) 55-592003-20098.2a(6.9,9.5)2009-20153.0a(1.4,4.7)2015-2017−6.2 (−12.8,0.9) 60-642003-20099.7a(6.6,12.8)2009-2017.8 (−1.1,2.6) 65-692003-20118.4a(6.9,10.1)2011-2017−1.2 (−3.4,1.1) 70-742003-20099.6a(8,11.3)2009-20132.3 (−2.2,7.1)2013-2017−4.4a(−7.1,−1.6) 75-792003-20117.3a(5.4,9.2)2011-2017−.9 (−3.6,1.8) 80-842003-20125.3a(2.6,8)2012-2017−1.3 (−7.3,5.2)Race White2003-20097.6a(6.2,9)2009-20153.0a(1.8,4.3)2015-2017−6.1a(−11.1,-.8) Black2003-20156.0a(5.4,6.5)2015-2017−14.6a(−20.9,−7.9) American Indian2003-20128.3a(4.7,12)2012-2017−.8 (−6.7,5.5) Asian2003-20107.1a(5.2,9.1)2010-20171 (−.4,2.4)Stage Localized2003-20097.2a(6.2,8.2)2009-20142.9a(1.1,4.6)2014-2017−4.5a(−7,−1.9) Regional2003-200910.2a(9.2,11.2)2009-20153.7a(2.4,4.9)2015-2017−1.2 (−6.5,4.4) Distant2003-2017−1.3 (−2.7,0.1)Size <1 cm2004-200910.7a(7.8,13.7)2009-20134 (−.2,8.3)2013-2017−3.8a(−6.3,−1.2) 1-2.9 cm2004-20098.5a(6.8,10.3)2009-20152.6a(1.4,3.9)2015-2017−3 (−7.8,2) ≥3 cm2004-20105.1a(3.1,7.2)2010-20153.9a(1,6.9)2015-2017−4 (−12,4.7)APC: annual percent change; CI: confidence interval.aThe value is statistically different from zero (2‐sided\nP < .05) based on joinpoint regression\nanalysis.\nTrends in the incidence rate of PTC by tumor sex. Rates are age adjusted\nto the 2000 US standard population.\nAnnual Percentage Change in Incidence of Papillary Thyroid Carcinoma,\n2003-2017.\nAPC: annual percent change; CI: confidence interval.\naThe value is statistically different from zero (2‐sided\nP < .05) based on joinpoint regression\nanalysis.\nThe age-specific incidence curve is shown in Figure 2. During the study period, the\nincidence rates were lower for those aged <30 and 80-84 years than in the other\nage groups. Overall, before about 2009, those aged 30-34 and 40-79 years had the\nfastest growth rates, with an annual growth rate of more than 7%, of which those\naged 60-64 and 70-74 years had a rate of nearly 10%. Each age group had a varying\ndegree of decline after 2013 or 2015, although some APCs were not statistically\nsignificant (Table\n1).Figure 2.Age-specific incidence trends of PTC.\nAge-specific incidence trends of PTC.\nIncidence trends were similar for whites and Asian Americans, who had consistently\nhigher rates than blacks and American Indians (Figure 3). The incidence for whites\nincreased by 7.6% (95% CI = 6.2-9.0%) per year during 2003-2009. The rate of\nincrease slowed slightly during 2009-2015 (APC = 3.0%, 95% CI = 1.8%–4.3%) (Figure 3 and Table 1). Likewise, the\nincidence among whites declined at a rate of 6.1% (95% CI = −11.1% to −.8%) between\n2015 and 2017. Asian Americans also had a fluctuating slight decline of 1% (95% CI =\n−.4% to −2.4%) during the most-recent period. The incidence for blacks declined\nsignificantly by 14.6% (95% CI = −20.9% to −7.9%) per year after 2015.Figure 3.Trends in the incidence rate of PTC by race. Rates are age adjusted to\nthe 2000 US standard population.\nTrends in the incidence rate of PTC by race. Rates are age adjusted to\nthe 2000 US standard population.\nPTC incidence curves and APCs by stage are presented in Figure 4 and Table 1. There was a significant\ndifference in incidence among the three stages, with the highest and lowest in the\nlocalized and distant stages, respectively. The incidence rates for patients in the\nlocalized and regional stages increased rapidly at rates of 7.2% and 10.2% before\n2009, respectively, and by 2.9% and 3.7% each year during 2009-2015. The incidence\nrates for patients in both stages decreased after 2015, with those in the localized\nstage having the greatest decrease (APC = −4.5%, 95% CI = −7% to −1.9%). The\nincidence for distant-stage patients has been stable for the past 15 years, and a\nslight downward trend was also recently observed (APC = −1.3% to 95% CI = −2.7% to\n.1%).Figure 4.Trends in the incidence rate of PTC by tumor size. Rates are age adjusted\nto the 2000 US standard population.\nTrends in the incidence rate of PTC by tumor size. Rates are age adjusted\nto the 2000 US standard population.\nWe also examined the trend of PTC incidence according to tumor size (Figure 5). The incidence of\ncancers of all sizes had the fastest growth rate during 2004-2009, with smaller\ntumors having faster rates (Table 1). After 2009-2010, the incidence growth rate of three sizes of\nPTC slowed. We found that the incidence of tumors smaller than 1 cm was reversed\nfrom 2013, decreasing by 3.8% per year. Similarly, tumor of 1-2.9 cm and larger had\ndownward trends after 2015 (3% [95% CI = −7.8% to 2%] and 4% [95% CI = −12% to\n4.7%]).Figure 5.Trends in the incidence rate of PTC by stage. Rates are age adjusted to\nthe 2000 US standard population.\nTrends in the incidence rate of PTC by stage. Rates are age adjusted to\nthe 2000 US standard population.\nThe age-period-cohort analysis revealed period and cohort effects on PTC incidence\ntrends (Figure 6). There\nwas a significant period effect during the study period. The effect size increased\nrapidly before 2008-2012, after which it slowed. This indicates that the rate of\nincrease in risk has slowed down after 2008-2012, which was consistent with the\nresults of the joinpoint analysis. Cohort effects and period effects had opposite\ntrends. The cohort curve had a slight downward trend. Taking the 1960 birth cohort\nas a reference, the risk of patients born before was slightly higher, and the risk\nof the subsequent birth cohort gradually decreased and leveled off.Figure 6.Period and cohort effects in the incidence of papillary thyroid\ncarcinoma.\nPeriod and cohort effects in the incidence of papillary thyroid\ncarcinoma.", "Our joinpoint analysis of PTC incidence during 2003-2017 found that the overall\nincidence in the United States did actually increase rapidly during 2003-2009 (APC =\n7.6%, 95% CI = 6.9-8.3%), and then decreased from 2009 to 2015 at a rate of 3.1% per\nyear. Notably, the trend of the overall PTC incidence began to reverse after 2015,\nshowing a decrease of 2.8% per year. Similar downward trends were found when the\ndata were stratified by sex, age, race, and tumor stage and size. The significant\nperiod effect indicated that PTC risk increased slowly after 2008-2012, while the\ncohort effect showed an opposite trend. After the 1960s, PTC risk decreased slightly\nand leveled off, indicating that later birth cohorts have lower risks than earlier\nbirth cohorts.\nThe decrease in the detection rate of PTC and the downward trend may be caused by\nvarious factors. Since the late 1970s, when high-resolution imaging techniques were\nfirst used to assess thyroid nodules leading to an increased likelihood of detecting\nlocalized TC and low-risk small nodules early, the widespread use of ultrasonography\nand needle biopsy led to an increased detection rate of small tumors.16-18 There are multiple reports of\nthe detection rate of PTC increasing, but this was not associated with any benefit\nof a reduced mortality risk.19,20 The American Thyroid\nAssociation (ATA) therefore guidelines should not recommend biopsy of nodules of\n5 mm or less from 2009.21 The 2015 ATA guidelines pointed out that the maximum\nthreshold for needle aspiration biopsy was increased to 1 cm.22 For nodules\nless than 1 cm suspicious ultrasound, if there is no thyroid invasion or ultrasound\nsuspicious lymph nodes can be closely followed up. The present study indicated that\nthe incidence of tumors <1 cm decreased to 4% from an annual increase of 10.7%\nprior to 2009. At the same time, a decreasing trend was also observed for tumors\nsized 1-2.9 cm and larger than 3 cm. Compared with before the revision, the\nincreasing trend in the PTC incidence subsequently slowed, indicating that it played\na role in reducing the detection rate of PTC.23\nThe PTC incidence peaked in 2015, followed by a downward trend. In addition to\nchanges in screening and thyroid nodule and biopsy criteria, another important\nfactor contributing to the increased PTC incidence in the past 2 decades is the\nincrease in the diagnosis of the follicular variant of PTC.24-26 Previous research\ndemonstrated that patients with noninvasive encapsulated follicular variant of\npapillary thyroid carcinoma (EFVPTC) have a very low risk of adverse outcomes and\nshould be recoded to classify noninvasive EFVPTC from malignant to tumor in\nsitu.27 This reclassification can significantly reduce the clinical\nconsequences associated with diagnosis and helps to reduce the fear experienced by\npatients. A recent stratification of the histological subtypes of PTC also indicated\nthat the decline in the overall PTC incidence during 2015-2017 coincided with a\ndecline in the noninvasive EFVPTC incidence, which was an important reason for the\nrecent decline in the PTC incidence.24\nOther possible risk factors for PTC include ionizing radiation, genetic changes,\nobesity, lifestyle changes, etc.28,29 Some studies have linked\nobesity and overweight to an increased risk of thyroid cancer29,30 but some have\nhad the opposite result.31 Exposure of human thyroid cells to ionizing radiation can\ninduce chromosomal rearrangements that lead to carcinogenesis and are therefore\nthought to be closely associated with an increased risk of PTC.32 However, the\neffects of these lifestyle and environmental changes on reducing the incidence of\nPTC are still unclear.\nPeriod effects generally reflect changes in risk affecting all age groups over a time\nperiod due to changes in environmental factors, diagnostic criteria, disease\nclassification, and screening programs. The results for the period effects in this\nstudy also reflect recent trends in the PTC incidence associated with revisions of\nthe ATA guidelines and changes in disease classification, which was consistent with\nthe above analysis. An epidemiological study using data from the national cancer\nregistry of South Korea during 1999-2016 determined that after nationwide screening\nfor TC was introduced in 1999, the ASIR of TC increased 10-fold in just over a\ndecade (from 6.3 per 100 000 in 1999 to 63.4 per 100 000 in 2012).33 However, the\nTC incidence in South Korean females decreased by nearly 20-fold in just 4 years,\ndue to changes in thyroid nodule screening guidelines and debate among the South\nKorean public, epidemiologists, and TC doctors about TC overdiagnosis since\n2014.33 These observations demonstrate the importance of the period\neffect on changes in TC incidence. The dramatic increase in incidence has been\nreversed by the emphasis on overdiagnosis and overtreatment of indolent diseases in\nthe United States and the implementation of changes in clinical practice guidelines\nrelated to biopsy risk criteria. This decreasing trend was observed in all\nsubgroups, with significant decreases not only in those with small tumors at the\ndistant stage, but also for >1 cm tumors at the distant stage, which was not\nstatistically significant. This suggests that the measures taken contributed\nsignificantly to reducing the PTC prevalence; however, due to delays in data\nregistration, more data are needed to characterize recent trends.\nThe cohort effect showed that the risk of morbidity in late birth cohorts decreased\nand stabilized compared with early birth cohorts. This may be due to changes in\nlifestyle and disease awareness. The most-recent birth cohort was better educated\nand had a better understanding of cancer knowledge and disease prevention.34 In addition,\nbased on research into the long-term downward trend of global TC mortality and the\ndownward trend of period and cohort effects, it has been argued that there is a lack\nof evidence for “true” risk factors (ie, those other than overdiagnosis) that may\nsignificantly contribute to TC mortality and indirectly confirm a major role of\noverdiagnosis in TC morbidity.35\nThe study had some limitations. Firstly, due to delays in data registration, only\n2 years of decline were observed, and more up-to-date data are therefore needed to\nconfirm whether this trend can be maintained. Secondly, although the\nage-period-cohort model can provide clues to the etiology of emerging trends, it\ndoes not produce direct evidence. At the same time, due to the inherent limitations\nof collinearity in the age-period-cohort model, the corresponding results should be\ncautiously interpreted. Due to the lack of relevant data on some important\ninfluencing factors such as personal lifestyle and environmental exposures in the\nSEER database, it is difficult to assess their impact on changes in PTC risk.", "In conclusion, the present analysis has revealed that the PTC incidence, which has\nbeen increasing rapidly, has recently slowed down and even decreased, including for\ntumors that are larger and of distant stage. This further supports the importance of\noverdiagnosis regarding health policymakers, physicians, and patients, the revision\nof thyroid management guidelines, and the reclassification of low-risk indolent TC\nas significant contributors to the decreased PTC detection rate. The results for the\nperiod effect also suggest that changes in PCT incidence are closely related to\noverdiagnosis. Future studies will need to analyze more-recent data to confirm these\ntrends." ]
[ "intro", "materials|methods", null, null, "results", "discussion", "conclusions" ]
[ "papillary thyroid carcinoma", "incidence trend", "joinpoint analysis", "age-period-cohort model", "surveillance, epidemiology, and end results" ]
Moderators of an intervention on emotional and behavioural problems: household- and school-level parental education.
36256856
Children of lower-educated parents and children in schools with a relatively high percentage of peers with lower-educated parents (lower parental education schools) are more likely to develop emotional and behavioural problems compared to children in higher-educated households and schools. Universal school-based preventive interventions, such as the Good Behaviour Game (GBG), are generally effective in preventing the development of emotional and behavioural problems, but information about potential moderators is limited. This study examined whether the effectiveness of the GBG in preventing emotional and behavioural problems differs between children in lower- and higher-educated households and schools.
BACKGROUND
Using a longitudinal multi-level randomized controlled trial design, 731 children (Mage=6.02 towards the end of kindergarten) from 31 mainstream schools (intervention arm: 21 schools, 484 children; control arm: 10 schools, 247 children) were followed annually from kindergarten to second grade (2004-2006). The GBG was implemented in first and second grades.
METHODS
Overall, the GBG prevented the development of emotional and behavioural problems. However, for emotional problems, the GBG-effect was slightly more pronounced in higher parental education schools than in lower parental education schools (Bhigher parental education schools =-0.281, P <0.001; Blower parental education schools =-0.140, P = 0.016). No moderation by household-level parental education was found.
RESULTS
Studies into universal school-based preventive interventions, and in particular the GBG, should consider and incorporate school-level factors when studying the effectiveness of such interventions. More attention should be directed towards factors that may influence universal prevention effectiveness, particularly in lower parental education schools.
CONCLUSIONS
[ "Child", "Humans", "Schools", "Problem Behavior", "Emotions", "Peer Group", "Parents" ]
9713443
Introduction
Poor mental health among school-aged children, including emotional and behavioural problems, is a global public health concern.1 Without intervention, emotional and behavioural problems that develop during elementary school have been shown to increase the risk of many concurrent and future negative outcomes, such as mental disorders, physical health problems, academic failure, criminality and unemployment in adulthood.1–3 Mental health problems cause a large proportion of the global disease burden and are estimated to account for 32.4% of years lived with disability and 13% of disability adjusted life years.4 Therefore, early prevention of emotional and behavioural problems is an urgent matter. Elementary schools are accessible and practical settings for the implementation of preventive (universal) interventions. Universal school-based preventive interventions (i.e. those delivered to all children) may be key to effective preventive efforts. One such programme is the Good Behaviour Game (GBG),5 which has been proven effective in preventing the development of children’s behavioural and emotional problems.6–9 The GBG has previously been referred to as a ‘behavioural vaccine’ due to its cost-effectiveness and its ability to prevent mental health problems across diverse cultures and populations.6 It aims to prevent mental health problems in healthy children and in children at risk of developing mental health problems. When implemented on a large scale in early primary education, universal school-based interventions like the GBG have the capacity to reach large quantities of broad populations, including children who may be otherwise hard to reach. However, in more recent research, it has been shown that the GBG may differentially affect children with varying risk profiles and that its benefit may not equally extend to children with higher family-demographic risk profiles.10 This challenges the notion that the GBG is a ‘behavioural vaccine’ and should be further explored. Thus, we investigate whether the effect of the GBG is moderated by a well-established risk factor at both the household and school levels. Across nations, a robust risk factor of poor child mental health at both the household and school levels is low socioeconomic status (SES).11 In the Netherlands, where this study was conducted, school-level socioeconomic inequalities within and between schools are measured by children’s parents’ education levels.12 Children of lower-educated parents (and higher-educated parents) are likely to attend schools with children from similar parental education backgrounds.13 Already in elementary school, children of lower-educated parents and children in schools with a high percentage of students with lower-educated parents (lower parental education schools) are at a higher risk of developing emotional and behavioural problems.14 This may be due to the risk factors that are associated with lower-educated households (e.g. less resources at home and less cultivating parenting strategies) and with lower parental education schools (e.g. less effective school management and more teacher distress).15,16 On the one hand, interventions like the GBG may have the potential to decrease inequalities in the prevalence of mental health problems in children from lower- and higher-educated contexts. On the other, they may be less effective in decreasing inequalities owing to factors related to lower household- and school-level parental education because these factors may reduce the effectiveness of the intervention. However, it remains unknown whether the impact of the GBG indeed differs between children from lower- and higher-educated households and schools. The majority of the school-based intervention studies on children’s emotional and behavioural problems have not included household- or school-level parental education or only included SES as a descriptive or a study variable.17,18 Some of these studies examined either children from low SES households10,19 or low SES schools alone19–23 and thereby lack a comparison group. Additionally, studies that did use SES as a moderator did not account for SES at both the household and school levels.17,18,24–28 Not accounting for SES at both levels may lead to the misleading conclusion that the effects are explained solely by either household- or school-level SES.14 Therefore, this study provides a novel approach by allowing a more detailed examination of the moderating role of a well-established risk factor at both levels. Specifically, we examine whether household- and school-level parental education moderate the effectiveness of the GBG in preventing the development of Dutch children’s emotional and behavioural problems from kindergarten to second grade.
Methods
[SUBTITLE] Sample [SUBSECTION] Participants were recruited from the first 31 elementary schools in rural and urban areas of the Netherlands that agreed to participate in the research project. Schools could participate if they were willing to implement the GBG (if randomly selected in the intervention arm) or if they were willing to be on a waiting list (if randomly selected in the control arm). Children’s emotional and behavioural problems were annually assessed for 3 years, from kindergarten (Mage=6.02, SD= 0.46) to second grade (in spring). Inclusion criteria were (i) active parental consent, (ii) data on school-level parental education and (iii) at least two out of three completed waves of teacher-reported data on emotional and behavioural problems. In total, out of 825 children who were initially included in the study, 731 (50% girls) fulfilled these criteria (see the flowchart in figure 1). All children had complete data on school-level parental education, 18.5% had missing data on household-level parental education and 24% had missing data on emotional and behavioural problems for one wave. Flowchart of the cluster randomized participants included in the randomized control trial, adapted with permission from Witvliet and colleagues30 Participants were recruited from the first 31 elementary schools in rural and urban areas of the Netherlands that agreed to participate in the research project. Schools could participate if they were willing to implement the GBG (if randomly selected in the intervention arm) or if they were willing to be on a waiting list (if randomly selected in the control arm). Children’s emotional and behavioural problems were annually assessed for 3 years, from kindergarten (Mage=6.02, SD= 0.46) to second grade (in spring). Inclusion criteria were (i) active parental consent, (ii) data on school-level parental education and (iii) at least two out of three completed waves of teacher-reported data on emotional and behavioural problems. In total, out of 825 children who were initially included in the study, 731 (50% girls) fulfilled these criteria (see the flowchart in figure 1). All children had complete data on school-level parental education, 18.5% had missing data on household-level parental education and 24% had missing data on emotional and behavioural problems for one wave. Flowchart of the cluster randomized participants included in the randomized control trial, adapted with permission from Witvliet and colleagues30 [SUBTITLE] Design and procedure [SUBSECTION] Participating schools were randomly assigned, with an oversampling of intervention schools, to either the control (10 schools, n = 247 children) or the GBG intervention arm (21 schools, n = 484 children). See Supplementary appendix A for sample size determination. The first assessments of emotional and behavioural problems were conducted in the Spring of 2004 when participants were in kindergarten (pre-intervention). In first and second grades, the GBG intervention was implemented and the second and third assessments were conducted. Participating schools were randomly assigned, with an oversampling of intervention schools, to either the control (10 schools, n = 247 children) or the GBG intervention arm (21 schools, n = 484 children). See Supplementary appendix A for sample size determination. The first assessments of emotional and behavioural problems were conducted in the Spring of 2004 when participants were in kindergarten (pre-intervention). In first and second grades, the GBG intervention was implemented and the second and third assessments were conducted. [SUBTITLE] The GBG [SUBSECTION] The GBG is a classroom-based preventive intervention that aims to prevent disruptive behaviour by creating a positive and a predictable classroom environment where children work in teams and stimulate each other to show appropriate classroom behaviour. The GBG is implemented in classrooms by teachers for 15–60-min periods while students are working on regular school tasks. Before the GBG period, teachers and students select positively formulated classroom rules. Teachers then identify and assign children to teams of 4–5 students with an equal number of disruptive and non-disruptive children and give each team a set of cards. During the game, if a team member violates one of the preselected rules, teachers take a card from that team. Teams are rewarded at the end of the game period if at least one card remains. Teachers praise teams and children by complimenting appropriate behaviour and, aside from removing cards from teams that violate the rules, do not pay attention to disruptive behaviour. The GBG is implemented in three phases: introduction, expansion and generalization. In the introduction phase, the GBG is played three times a week. In the expansion and generalization phases, the duration (hours/days) is extended. More information regarding the intervention strategy, implementation and teacher training is described elsewhere.29 The GBG is a classroom-based preventive intervention that aims to prevent disruptive behaviour by creating a positive and a predictable classroom environment where children work in teams and stimulate each other to show appropriate classroom behaviour. The GBG is implemented in classrooms by teachers for 15–60-min periods while students are working on regular school tasks. Before the GBG period, teachers and students select positively formulated classroom rules. Teachers then identify and assign children to teams of 4–5 students with an equal number of disruptive and non-disruptive children and give each team a set of cards. During the game, if a team member violates one of the preselected rules, teachers take a card from that team. Teams are rewarded at the end of the game period if at least one card remains. Teachers praise teams and children by complimenting appropriate behaviour and, aside from removing cards from teams that violate the rules, do not pay attention to disruptive behaviour. The GBG is implemented in three phases: introduction, expansion and generalization. In the introduction phase, the GBG is played three times a week. In the expansion and generalization phases, the duration (hours/days) is extended. More information regarding the intervention strategy, implementation and teacher training is described elsewhere.29 [SUBTITLE] Measures [SUBSECTION] Household-level parental education was based on the highest education level per household, obtained by the (two) parent/caregiver(s). Parental education levels were ranked according to the Dutch Standard Education Classification,30 which corresponds to the International Standard Classification of Education (ISCED).31 Following the ISCED classifications, parental education levels were coded using an 8-point scale, with education levels ranging from 0= no education/early education, 1= primary education, 2= lower secondary education, 3= upper secondary education, 4= post-secondary non-tertiary education, 5= short-cycle tertiary education, 6 =bachelor’s degree or equivalent, to 7 =master’s degree, equivalent or higher. The household parental education levels were reverse coded so that higher scores indicated lower parental education levels. School-level parental education levels were determined by the per-school percentage of children of low-educated parents. In the Netherlands, school-level socioeconomic inequalities are measured by children’s parents’ education levels. The Netherlands Inspectorate of Education calculates the percentage of low parental education levels of each school to identify schools that qualify for additional governmental resources.12 Low education is defined as parent(s) completing no more than elementary school. Thus, in this study, school-level parental education was based on the percentage score of low parental education levels of the entire school population. The percentage scores can range from 0% to 100%, with higher percentage scores indicating schools with higher percentages of children of low-educated parents. This information is publicly available (www.duo.nl). Teacher ratings of individual children’s behavioural and emotional problems were assessed by the Problem Behaviour at School Interview (PBSI).32 The PBSI is a validated questionnaire conducted via interview that uses a 5-point Likert scale ranging from 0 (never applicable) to 4 (often applicable).33 Behavioural problem scores were assessed by conduct problems (12 items) and oppositional defiant problems (7 items), and calculated as the average of the mean scores of the two subscales. Emotional problem scores were assessed by depression (7 items) and anxiety (5 items) symptoms, and the same procedure was followed. Higher scores indicated higher levels of emotional and behavioural problems. See Supplementary appendix A for more information regarding the PBSI and the outcome variables. Intervention status was dummy-coded (0 =control, 1 = GBG). Covariates included gender (0 =girls, 1 =boys) and cluster size. Cluster size (i.e. number of participating children per school) was grand-mean centred and included to account for unequal cluster sizes (M=23, range =8–88; mode =14, median =20). Baseline differences in kindergarten were controlled for because—despite randomization—children in the GBG arm had moderately higher levels of emotional [MGBG=0.85, SD=0.57; Mcontrol=0.67, SD=0.55, t(647)=−3.85, P<0.001, Cohen’s d=0.32] and slightly higher levels of behavioural problems [MGBG=0.80, SD=0.67; Mcontrol=0.69, SD=0.65, t(650)=−2.08, P=0.038, Cohen’s d=0.17] than children in the control arm. Household-level parental education was based on the highest education level per household, obtained by the (two) parent/caregiver(s). Parental education levels were ranked according to the Dutch Standard Education Classification,30 which corresponds to the International Standard Classification of Education (ISCED).31 Following the ISCED classifications, parental education levels were coded using an 8-point scale, with education levels ranging from 0= no education/early education, 1= primary education, 2= lower secondary education, 3= upper secondary education, 4= post-secondary non-tertiary education, 5= short-cycle tertiary education, 6 =bachelor’s degree or equivalent, to 7 =master’s degree, equivalent or higher. The household parental education levels were reverse coded so that higher scores indicated lower parental education levels. School-level parental education levels were determined by the per-school percentage of children of low-educated parents. In the Netherlands, school-level socioeconomic inequalities are measured by children’s parents’ education levels. The Netherlands Inspectorate of Education calculates the percentage of low parental education levels of each school to identify schools that qualify for additional governmental resources.12 Low education is defined as parent(s) completing no more than elementary school. Thus, in this study, school-level parental education was based on the percentage score of low parental education levels of the entire school population. The percentage scores can range from 0% to 100%, with higher percentage scores indicating schools with higher percentages of children of low-educated parents. This information is publicly available (www.duo.nl). Teacher ratings of individual children’s behavioural and emotional problems were assessed by the Problem Behaviour at School Interview (PBSI).32 The PBSI is a validated questionnaire conducted via interview that uses a 5-point Likert scale ranging from 0 (never applicable) to 4 (often applicable).33 Behavioural problem scores were assessed by conduct problems (12 items) and oppositional defiant problems (7 items), and calculated as the average of the mean scores of the two subscales. Emotional problem scores were assessed by depression (7 items) and anxiety (5 items) symptoms, and the same procedure was followed. Higher scores indicated higher levels of emotional and behavioural problems. See Supplementary appendix A for more information regarding the PBSI and the outcome variables. Intervention status was dummy-coded (0 =control, 1 = GBG). Covariates included gender (0 =girls, 1 =boys) and cluster size. Cluster size (i.e. number of participating children per school) was grand-mean centred and included to account for unequal cluster sizes (M=23, range =8–88; mode =14, median =20). Baseline differences in kindergarten were controlled for because—despite randomization—children in the GBG arm had moderately higher levels of emotional [MGBG=0.85, SD=0.57; Mcontrol=0.67, SD=0.55, t(647)=−3.85, P<0.001, Cohen’s d=0.32] and slightly higher levels of behavioural problems [MGBG=0.80, SD=0.67; Mcontrol=0.69, SD=0.65, t(650)=−2.08, P=0.038, Cohen’s d=0.17] than children in the control arm. [SUBTITLE] Statistical analyses [SUBSECTION] A parallel latent growth curve (LGM) model with two-level time-nested-within-individual data structure (1 =variation across individual children, 2 =variation across schools), in which the development of emotional and behavioural problems was conceptualized by latent growth parameters (intercept and a linear slope), was used to test the main effects and potential moderation by household- and school-level parental education of the GBG in preventing the development of emotional and behavioural problems. The intercept represented the initial level in kindergarten (baseline) and the slope represented change over time (from kindergarten to second grade). The analyses were conducted in three steps. All models were fitted in Mplus version 8.0.34 We first computed design effects. Design effects larger than 2.0 indicate significant clustering of the data at the school level [Design effects =1+(nc−1)ICC].35 In the second step, we tested for main effects of the GBG intervention by regressing the outcome on the GBG intervention status, adjusting for the baseline differences in emotional and behavioural problems. In the third step, we tested moderation by household- and school-level parental education via a cross-level interaction and a between(school)-level interaction, respectively. Before examining cross-level interactions between household-level parental education and the GBG, we checked whether such interactions could be performed. To do this, we modelled a random slope at the (within)household-level and estimated its variance at the (between)school-level. This random slope represented the effect of household-level parental education on the growth parameters of children’s (individual-level) emotional or behavioural problems. Then, using Satorra Bentler chi-square difference tests, we checked whether adding a random slope improved the model fit of the main effect model in Step 2. If this was the case, the random slope parameter was regressed on the GBG at the between-level (i.e. cross-level interaction) to test the interaction between household-level parental education and the GBG on the development of individual-level emotional and behavioural problems. To test for moderation by school-level parental education at the between-level, an interaction term between school-level parental education and the GBG was added as a predictor of between-level emotional and behavioural problem development. Model fit indices for multi-level latent growth models were used to determine model fit at both the household and school levels. For specifics, see Supplementary appendix Btable S1. MLR estimators (maximum likelihood estimation with robust standard errors) were used to account for the possible non-normal distribution of data. Missing data were therefore handled using the default option in Mplus for MLR-estimation with missing at random data (i.e. Full Information Maximum Likelihood estimation). To ensure that the results were robust, two additional sensitivity tests were done: (i) by imputing the missing data in MPLUS (N = 25 imputed datasets) and (ii) by testing the models on a subsample (N=596) with complete household-level parental education data. A parallel latent growth curve (LGM) model with two-level time-nested-within-individual data structure (1 =variation across individual children, 2 =variation across schools), in which the development of emotional and behavioural problems was conceptualized by latent growth parameters (intercept and a linear slope), was used to test the main effects and potential moderation by household- and school-level parental education of the GBG in preventing the development of emotional and behavioural problems. The intercept represented the initial level in kindergarten (baseline) and the slope represented change over time (from kindergarten to second grade). The analyses were conducted in three steps. All models were fitted in Mplus version 8.0.34 We first computed design effects. Design effects larger than 2.0 indicate significant clustering of the data at the school level [Design effects =1+(nc−1)ICC].35 In the second step, we tested for main effects of the GBG intervention by regressing the outcome on the GBG intervention status, adjusting for the baseline differences in emotional and behavioural problems. In the third step, we tested moderation by household- and school-level parental education via a cross-level interaction and a between(school)-level interaction, respectively. Before examining cross-level interactions between household-level parental education and the GBG, we checked whether such interactions could be performed. To do this, we modelled a random slope at the (within)household-level and estimated its variance at the (between)school-level. This random slope represented the effect of household-level parental education on the growth parameters of children’s (individual-level) emotional or behavioural problems. Then, using Satorra Bentler chi-square difference tests, we checked whether adding a random slope improved the model fit of the main effect model in Step 2. If this was the case, the random slope parameter was regressed on the GBG at the between-level (i.e. cross-level interaction) to test the interaction between household-level parental education and the GBG on the development of individual-level emotional and behavioural problems. To test for moderation by school-level parental education at the between-level, an interaction term between school-level parental education and the GBG was added as a predictor of between-level emotional and behavioural problem development. Model fit indices for multi-level latent growth models were used to determine model fit at both the household and school levels. For specifics, see Supplementary appendix Btable S1. MLR estimators (maximum likelihood estimation with robust standard errors) were used to account for the possible non-normal distribution of data. Missing data were therefore handled using the default option in Mplus for MLR-estimation with missing at random data (i.e. Full Information Maximum Likelihood estimation). To ensure that the results were robust, two additional sensitivity tests were done: (i) by imputing the missing data in MPLUS (N = 25 imputed datasets) and (ii) by testing the models on a subsample (N=596) with complete household-level parental education data. [SUBTITLE] Ethics [SUBSECTION] This study was approved by the Medical Ethics Committee of the Vrije Universiteit Amsterdam Medical Center and was registered with the ‘Netherlands Trial Register’ [Trial NL470 (NTR512)] (www.trialregister.nl). Signed parental informed consent was obtained from parents. Parents and children could revoke participation at any time. This study was approved by the Medical Ethics Committee of the Vrije Universiteit Amsterdam Medical Center and was registered with the ‘Netherlands Trial Register’ [Trial NL470 (NTR512)] (www.trialregister.nl). Signed parental informed consent was obtained from parents. Parents and children could revoke participation at any time.
Results
[SUBTITLE] Descriptive statistics [SUBSECTION] Descriptive statistics of household- and school-level parental education of the whole sample are presented in table 1. The household-level parental education levels were slightly higher in the control arm than in the GBG arm, t(1)=2.75, P=0.006, Cohen’s d=0.24. Descriptive statistics of household- and school-level parental education of the whole sample The per-school percentage of children of low-educated parents was not significantly different between the schools in the control (M=18.61%, SD=23.97%) and intervention arms (M=15.35%, SD=17.02%), t(29)=0.44, P=0.666, Cohen’s d=0.17. The correlation between household- and school-level parental education in our sample was positive and of moderate magnitude (r=0.42, P<0.001). Descriptive statistics of household- and school-level parental education of the whole sample are presented in table 1. The household-level parental education levels were slightly higher in the control arm than in the GBG arm, t(1)=2.75, P=0.006, Cohen’s d=0.24. Descriptive statistics of household- and school-level parental education of the whole sample The per-school percentage of children of low-educated parents was not significantly different between the schools in the control (M=18.61%, SD=23.97%) and intervention arms (M=15.35%, SD=17.02%), t(29)=0.44, P=0.666, Cohen’s d=0.17. The correlation between household- and school-level parental education in our sample was positive and of moderate magnitude (r=0.42, P<0.001). [SUBTITLE] Model building, unconditional latent growth models per condition and the GBG main effects [SUBSECTION] Intra-class correlations, design effect values, model fit indices of the unconditional LGMs for the whole sample and model building testing results are presented in Supplementary appendix Btable S1. Design effects indicated the need to use a two-level structure to analyze the data. Model fit indices were acceptable for both outcomes. Adding the random slope improved the model fit of the main effect model of emotional problems only, which indicated that cross-level interaction testing can be performed for emotional but not for behavioural problems. Results from the unconditional LGMs (Supplementary appendix Btable S2) showed that in the GBG arm emotional and behavioural problems stayed stable over time, as indicated by the non-significant slopes (emotional problems: B=0.065, P=0.115; behavioural problems: B=−0.041, P=0.177). In the control arm, there was a significant yearly increase of emotional problems (B = 0.271, P < 0.001) and a borderline significant yearly increase of behavioural problems (B=0.100, P=0.057). This indicates that without the GBG, emotional (and to a lesser extent behavioural) problems tended to increase from kindergarten to second grade. Results of main effects (table 2) showed that the GBG was effective in preventing the increase in emotional problems that was found in the control group [B=−0.208, 95% CI (−0.345, −0.070), P = 0.003]. In addition, the GBG was also effective in preventing behavioural problems from kindergarten to second grade [B=−0.133, 95% CI (−0.256, −0.010), P=0.034]. Main effects of the GBG and moderation by household- and school-level parental education on children’s emotional and behavioural problems Note: *P < .05, **P < .01, ***P < .001; n.a. = estimate not available due to the use of Monte Carlo integration to estimate cross-level interaction. Note that the effect of school-level parental education is small because it represents the effect at 1% change in school-level parental education. Intra-class correlations, design effect values, model fit indices of the unconditional LGMs for the whole sample and model building testing results are presented in Supplementary appendix Btable S1. Design effects indicated the need to use a two-level structure to analyze the data. Model fit indices were acceptable for both outcomes. Adding the random slope improved the model fit of the main effect model of emotional problems only, which indicated that cross-level interaction testing can be performed for emotional but not for behavioural problems. Results from the unconditional LGMs (Supplementary appendix Btable S2) showed that in the GBG arm emotional and behavioural problems stayed stable over time, as indicated by the non-significant slopes (emotional problems: B=0.065, P=0.115; behavioural problems: B=−0.041, P=0.177). In the control arm, there was a significant yearly increase of emotional problems (B = 0.271, P < 0.001) and a borderline significant yearly increase of behavioural problems (B=0.100, P=0.057). This indicates that without the GBG, emotional (and to a lesser extent behavioural) problems tended to increase from kindergarten to second grade. Results of main effects (table 2) showed that the GBG was effective in preventing the increase in emotional problems that was found in the control group [B=−0.208, 95% CI (−0.345, −0.070), P = 0.003]. In addition, the GBG was also effective in preventing behavioural problems from kindergarten to second grade [B=−0.133, 95% CI (−0.256, −0.010), P=0.034]. Main effects of the GBG and moderation by household- and school-level parental education on children’s emotional and behavioural problems Note: *P < .05, **P < .01, ***P < .001; n.a. = estimate not available due to the use of Monte Carlo integration to estimate cross-level interaction. Note that the effect of school-level parental education is small because it represents the effect at 1% change in school-level parental education. [SUBTITLE] Moderation by household- and school-level parental education of the GBG impact [SUBSECTION] Household level: Results showed no significant cross-level interaction between household-level parental education and the GBG-effect on individual-level emotional problem development, B=0.010, 95% CI (−0.055, 0.074), P=0.765 (see table 2). The cross-level interaction for behavioural problems was not tested. School level: Results showed a significant interaction between school-level parental education and the GBG-effect on children’s emotional problems, B=0.007, 95% CI (0.002, 0.013), P = 0.005 (see table 2). That is, the GBG was more effective in preventing the development of emotional problems in higher parental education schools than in lower parental education schools. Figure 2A shows a visual representation of this interaction effect in which the effects were probed at 0.50 SD above [lower parental education schools; ∼26% of the total sample; B=−0.140, SE=0.059, 95% CI (−0.255, −0.026), P = 0.016] and at 0.50 SD below the mean of school-level parental education [higher parental education schools; ∼7% of the total sample; B=−0.281, SE=0.080, 95% CI (−0.438, −0.124), P < 0.001]. For behavioural problems, no moderation between school-level parental education and the GBG was found, B=0.002, 95% CI (−0.003, 0.007), P = 0.382 (see figure 2B). The two sensitivity tests showed no changes in interpretation of the results. For specifics, see Supplementary appendix Btables S3 and S4. School-level parental education effects on the development of emotional problems (A) and behavioural problems (B) in GBG vs. control arms Household level: Results showed no significant cross-level interaction between household-level parental education and the GBG-effect on individual-level emotional problem development, B=0.010, 95% CI (−0.055, 0.074), P=0.765 (see table 2). The cross-level interaction for behavioural problems was not tested. School level: Results showed a significant interaction between school-level parental education and the GBG-effect on children’s emotional problems, B=0.007, 95% CI (0.002, 0.013), P = 0.005 (see table 2). That is, the GBG was more effective in preventing the development of emotional problems in higher parental education schools than in lower parental education schools. Figure 2A shows a visual representation of this interaction effect in which the effects were probed at 0.50 SD above [lower parental education schools; ∼26% of the total sample; B=−0.140, SE=0.059, 95% CI (−0.255, −0.026), P = 0.016] and at 0.50 SD below the mean of school-level parental education [higher parental education schools; ∼7% of the total sample; B=−0.281, SE=0.080, 95% CI (−0.438, −0.124), P < 0.001]. For behavioural problems, no moderation between school-level parental education and the GBG was found, B=0.002, 95% CI (−0.003, 0.007), P = 0.382 (see figure 2B). The two sensitivity tests showed no changes in interpretation of the results. For specifics, see Supplementary appendix Btables S3 and S4. School-level parental education effects on the development of emotional problems (A) and behavioural problems (B) in GBG vs. control arms
null
null
[ "Sample", "Design and procedure", "The GBG", "Measures", "Statistical analyses", "Ethics", "Descriptive statistics", "Model building, unconditional latent growth models per condition and the GBG main effects", "Moderation by household- and school-level parental education of the GBG impact", "Supplementary data", "Funding" ]
[ "Participants were recruited from the first 31 elementary schools in rural and urban areas of the Netherlands that agreed to participate in the research project. Schools could participate if they were willing to implement the GBG (if randomly selected in the intervention arm) or if they were willing to be on a waiting list (if randomly selected in the control arm).\nChildren’s emotional and behavioural problems were annually assessed for 3 years, from kindergarten (Mage=6.02, SD= 0.46) to second grade (in spring). Inclusion criteria were (i) active parental consent, (ii) data on school-level parental education and (iii) at least two out of three completed waves of teacher-reported data on emotional and behavioural problems. In total, out of 825 children who were initially included in the study, 731 (50% girls) fulfilled these criteria (see the flowchart in figure 1). All children had complete data on school-level parental education, 18.5% had missing data on household-level parental education and 24% had missing data on emotional and behavioural problems for one wave.\nFlowchart of the cluster randomized participants included in the randomized control trial, adapted with permission from Witvliet and colleagues30", "Participating schools were randomly assigned, with an oversampling of intervention schools, to either the control (10 schools, n = 247 children) or the GBG intervention arm (21 schools, n = 484 children). See Supplementary appendix A for sample size determination. The first assessments of emotional and behavioural problems were conducted in the Spring of 2004 when participants were in kindergarten (pre-intervention). In first and second grades, the GBG intervention was implemented and the second and third assessments were conducted.", "The GBG is a classroom-based preventive intervention that aims to prevent disruptive behaviour by creating a positive and a predictable classroom environment where children work in teams and stimulate each other to show appropriate classroom behaviour. The GBG is implemented in classrooms by teachers for 15–60-min periods while students are working on regular school tasks. Before the GBG period, teachers and students select positively formulated classroom rules. Teachers then identify and assign children to teams of 4–5 students with an equal number of disruptive and non-disruptive children and give each team a set of cards. During the game, if a team member violates one of the preselected rules, teachers take a card from that team. Teams are rewarded at the end of the game period if at least one card remains. Teachers praise teams and children by complimenting appropriate behaviour and, aside from removing cards from teams that violate the rules, do not pay attention to disruptive behaviour. The GBG is implemented in three phases: introduction, expansion and generalization. In the introduction phase, the GBG is played three times a week. In the expansion and generalization phases, the duration (hours/days) is extended. More information regarding the intervention strategy, implementation and teacher training is described elsewhere.29", "\nHousehold-level parental education was based on the highest education level per household, obtained by the (two) parent/caregiver(s). Parental education levels were ranked according to the Dutch Standard Education Classification,30 which corresponds to the International Standard Classification of Education (ISCED).31 Following the ISCED classifications, parental education levels were coded using an 8-point scale, with education levels ranging from 0= no education/early education, 1= primary education, 2= lower secondary education, 3= upper secondary education, 4= post-secondary non-tertiary education, 5= short-cycle tertiary education, 6 =bachelor’s degree or equivalent, to 7 =master’s degree, equivalent or higher. The household parental education levels were reverse coded so that higher scores indicated lower parental education levels.\n\nSchool-level parental education levels were determined by the per-school percentage of children of low-educated parents. In the Netherlands, school-level socioeconomic inequalities are measured by children’s parents’ education levels. The Netherlands Inspectorate of Education calculates the percentage of low parental education levels of each school to identify schools that qualify for additional governmental resources.12 Low education is defined as parent(s) completing no more than elementary school. Thus, in this study, school-level parental education was based on the percentage score of low parental education levels of the entire school population. The percentage scores can range from 0% to 100%, with higher percentage scores indicating schools with higher percentages of children of low-educated parents. This information is publicly available (www.duo.nl).\n\nTeacher ratings of individual children’s behavioural and emotional problems were assessed by the Problem Behaviour at School Interview (PBSI).32 The PBSI is a validated questionnaire conducted via interview that uses a 5-point Likert scale ranging from 0 (never applicable) to 4 (often applicable).33 Behavioural problem scores were assessed by conduct problems (12 items) and oppositional defiant problems (7 items), and calculated as the average of the mean scores of the two subscales. Emotional problem scores were assessed by depression (7 items) and anxiety (5 items) symptoms, and the same procedure was followed. Higher scores indicated higher levels of emotional and behavioural problems. See Supplementary appendix A for more information regarding the PBSI and the outcome variables.\n\nIntervention status was dummy-coded (0 =control, 1 = GBG).\n\nCovariates included gender (0 =girls, 1 =boys) and cluster size. Cluster size (i.e. number of participating children per school) was grand-mean centred and included to account for unequal cluster sizes (M=23, range =8–88; mode =14, median =20). Baseline differences in kindergarten were controlled for because—despite randomization—children in the GBG arm had moderately higher levels of emotional [MGBG=0.85, SD=0.57; Mcontrol=0.67, SD=0.55, t(647)=−3.85, P<0.001, Cohen’s d=0.32] and slightly higher levels of behavioural problems [MGBG=0.80, SD=0.67; Mcontrol=0.69, SD=0.65, t(650)=−2.08, P=0.038, Cohen’s d=0.17] than children in the control arm.", "A parallel latent growth curve (LGM) model with two-level time-nested-within-individual data structure (1 =variation across individual children, 2 =variation across schools), in which the development of emotional and behavioural problems was conceptualized by latent growth parameters (intercept and a linear slope), was used to test the main effects and potential moderation by household- and school-level parental education of the GBG in preventing the development of emotional and behavioural problems. The intercept represented the initial level in kindergarten (baseline) and the slope represented change over time (from kindergarten to second grade).\nThe analyses were conducted in three steps. All models were fitted in Mplus version 8.0.34 We first computed design effects. Design effects larger than 2.0 indicate significant clustering of the data at the school level [Design effects =1+(nc−1)ICC].35 In the second step, we tested for main effects of the GBG intervention by regressing the outcome on the GBG intervention status, adjusting for the baseline differences in emotional and behavioural problems. In the third step, we tested moderation by household- and school-level parental education via a cross-level interaction and a between(school)-level interaction, respectively. Before examining cross-level interactions between household-level parental education and the GBG, we checked whether such interactions could be performed. To do this, we modelled a random slope at the (within)household-level and estimated its variance at the (between)school-level. This random slope represented the effect of household-level parental education on the growth parameters of children’s (individual-level) emotional or behavioural problems. Then, using Satorra Bentler chi-square difference tests, we checked whether adding a random slope improved the model fit of the main effect model in Step 2. If this was the case, the random slope parameter was regressed on the GBG at the between-level (i.e. cross-level interaction) to test the interaction between household-level parental education and the GBG on the development of individual-level emotional and behavioural problems. To test for moderation by school-level parental education at the between-level, an interaction term between school-level parental education and the GBG was added as a predictor of between-level emotional and behavioural problem development.\nModel fit indices for multi-level latent growth models were used to determine model fit at both the household and school levels. For specifics, see Supplementary appendix Btable S1. MLR estimators (maximum likelihood estimation with robust standard errors) were used to account for the possible non-normal distribution of data. Missing data were therefore handled using the default option in Mplus for MLR-estimation with missing at random data (i.e. Full Information Maximum Likelihood estimation). To ensure that the results were robust, two additional sensitivity tests were done: (i) by imputing the missing data in MPLUS (N = 25 imputed datasets) and (ii) by testing the models on a subsample (N=596) with complete household-level parental education data.", "This study was approved by the Medical Ethics Committee of the Vrije Universiteit Amsterdam Medical Center and was registered with the ‘Netherlands Trial Register’ [Trial NL470 (NTR512)] (www.trialregister.nl). Signed parental informed consent was obtained from parents. Parents and children could revoke participation at any time.", "Descriptive statistics of household- and school-level parental education of the whole sample are presented in table 1. The household-level parental education levels were slightly higher in the control arm than in the GBG arm, t(1)=2.75, P=0.006, Cohen’s d=0.24.\nDescriptive statistics of household- and school-level parental education of the whole sample\nThe per-school percentage of children of low-educated parents was not significantly different between the schools in the control (M=18.61%, SD=23.97%) and intervention arms (M=15.35%, SD=17.02%), t(29)=0.44, P=0.666, Cohen’s d=0.17. The correlation between household- and school-level parental education in our sample was positive and of moderate magnitude (r=0.42, P<0.001).", "Intra-class correlations, design effect values, model fit indices of the unconditional LGMs for the whole sample and model building testing results are presented in Supplementary appendix Btable S1. Design effects indicated the need to use a two-level structure to analyze the data. Model fit indices were acceptable for both outcomes. Adding the random slope improved the model fit of the main effect model of emotional problems only, which indicated that cross-level interaction testing can be performed for emotional but not for behavioural problems.\nResults from the unconditional LGMs (Supplementary appendix Btable S2) showed that in the GBG arm emotional and behavioural problems stayed stable over time, as indicated by the non-significant slopes (emotional problems: B=0.065, P=0.115; behavioural problems: B=−0.041, P=0.177). In the control arm, there was a significant yearly increase of emotional problems (B = 0.271, P < 0.001) and a borderline significant yearly increase of behavioural problems (B=0.100, P=0.057). This indicates that without the GBG, emotional (and to a lesser extent behavioural) problems tended to increase from kindergarten to second grade.\nResults of main effects (table 2) showed that the GBG was effective in preventing the increase in emotional problems that was found in the control group [B=−0.208, 95% CI (−0.345, −0.070), P = 0.003]. In addition, the GBG was also effective in preventing behavioural problems from kindergarten to second grade [B=−0.133, 95% CI (−0.256, −0.010), P=0.034].\nMain effects of the GBG and moderation by household- and school-level parental education on children’s emotional and behavioural problems\n\nNote: *P < .05, **P < .01, ***P < .001; n.a. = estimate not available due to the use of Monte Carlo integration to estimate cross-level interaction. Note that the effect of school-level parental education is small because it represents the effect at 1% change in school-level parental education.", "\nHousehold level: Results showed no significant cross-level interaction between household-level parental education and the GBG-effect on individual-level emotional problem development, B=0.010, 95% CI (−0.055, 0.074), P=0.765 (see table 2). The cross-level interaction for behavioural problems was not tested.\n\nSchool level: Results showed a significant interaction between school-level parental education and the GBG-effect on children’s emotional problems, B=0.007, 95% CI (0.002, 0.013), P = 0.005 (see table 2). That is, the GBG was more effective in preventing the development of emotional problems in higher parental education schools than in lower parental education schools. Figure 2A shows a visual representation of this interaction effect in which the effects were probed at 0.50 SD above [lower parental education schools; ∼26% of the total sample; B=−0.140, SE=0.059, 95% CI (−0.255, −0.026), P = 0.016] and at 0.50 SD below the mean of school-level parental education [higher parental education schools; ∼7% of the total sample; B=−0.281, SE=0.080, 95% CI (−0.438, −0.124), P < 0.001]. For behavioural problems, no moderation between school-level parental education and the GBG was found, B=0.002, 95% CI (−0.003, 0.007), P = 0.382 (see figure 2B). The two sensitivity tests showed no changes in interpretation of the results. For specifics, see Supplementary appendix Btables S3 and S4.\nSchool-level parental education effects on the development of emotional problems (A) and behavioural problems (B) in GBG vs. control arms", "\nSupplementary data are available at EURPUB online.", "This study was supported by a grant from the Netherlands Organization for Health Research and Development (ZonMw) (project No. 531003013) and by ZonMw Grants #26200002 and #120620029.T.A.J.H. was funded through a grant awarded by the Norwegian Research Council (project number 288638) to the Centre for Global Health Inequalities Research (CHAIN) at the Norwegian University for Science and Technology (NTNU).\n\nConflicts of interest: None declared.\nThe effectiveness of the GBG in preventing the development of behavioural and emotional problems did not differ between children of lower- and higher-educated parents from kindergarten to second grade.\nThe GBG is equally effective in preventing the development of behavioural problems in schools with higher and lower percentages of children with lower-educated parents, but less effective in preventing the development of emotional problems in lower parental education schools than in higher parental education schools.\nWhen testing intervention effectiveness, school-level variables as moderators should be included in study designs.\nMore attention should be directed towards schools with a higher percentage of children with lower-educated parents." ]
[ null, null, null, null, null, null, null, null, null, null, null ]
[ "Introduction", "Methods", "Sample", "Design and procedure", "The GBG", "Measures", "Statistical analyses", "Ethics", "Results", "Descriptive statistics", "Model building, unconditional latent growth models per condition and the GBG main effects", "Moderation by household- and school-level parental education of the GBG impact", "Discussion", "Supplementary data", "Funding", "Supplementary Material" ]
[ "Poor mental health among school-aged children, including emotional and behavioural problems, is a global public health concern.1 Without intervention, emotional and behavioural problems that develop during elementary school have been shown to increase the risk of many concurrent and future negative outcomes, such as mental disorders, physical health problems, academic failure, criminality and unemployment in adulthood.1–3 Mental health problems cause a large proportion of the global disease burden and are estimated to account for 32.4% of years lived with disability and 13% of disability adjusted life years.4 Therefore, early prevention of emotional and behavioural problems is an urgent matter. Elementary schools are accessible and practical settings for the implementation of preventive (universal) interventions. Universal school-based preventive interventions (i.e. those delivered to all children) may be key to effective preventive efforts. One such programme is the Good Behaviour Game (GBG),5 which has been proven effective in preventing the development of children’s behavioural and emotional problems.6–9\nThe GBG has previously been referred to as a ‘behavioural vaccine’ due to its cost-effectiveness and its ability to prevent mental health problems across diverse cultures and populations.6 It aims to prevent mental health problems in healthy children and in children at risk of developing mental health problems. When implemented on a large scale in early primary education, universal school-based interventions like the GBG have the capacity to reach large quantities of broad populations, including children who may be otherwise hard to reach. However, in more recent research, it has been shown that the GBG may differentially affect children with varying risk profiles and that its benefit may not equally extend to children with higher family-demographic risk profiles.10 This challenges the notion that the GBG is a ‘behavioural vaccine’ and should be further explored. Thus, we investigate whether the effect of the GBG is moderated by a well-established risk factor at both the household and school levels.\nAcross nations, a robust risk factor of poor child mental health at both the household and school levels is low socioeconomic status (SES).11 In the Netherlands, where this study was conducted, school-level socioeconomic inequalities within and between schools are measured by children’s parents’ education levels.12 Children of lower-educated parents (and higher-educated parents) are likely to attend schools with children from similar parental education backgrounds.13 Already in elementary school, children of lower-educated parents and children in schools with a high percentage of students with lower-educated parents (lower parental education schools) are at a higher risk of developing emotional and behavioural problems.14 This may be due to the risk factors that are associated with lower-educated households (e.g. less resources at home and less cultivating parenting strategies) and with lower parental education schools (e.g. less effective school management and more teacher distress).15,16 On the one hand, interventions like the GBG may have the potential to decrease inequalities in the prevalence of mental health problems in children from lower- and higher-educated contexts. On the other, they may be less effective in decreasing inequalities owing to factors related to lower household- and school-level parental education because these factors may reduce the effectiveness of the intervention. However, it remains unknown whether the impact of the GBG indeed differs between children from lower- and higher-educated households and schools.\nThe majority of the school-based intervention studies on children’s emotional and behavioural problems have not included household- or school-level parental education or only included SES as a descriptive or a study variable.17,18 Some of these studies examined either children from low SES households10,19 or low SES schools alone19–23 and thereby lack a comparison group. Additionally, studies that did use SES as a moderator did not account for SES at both the household and school levels.17,18,24–28 Not accounting for SES at both levels may lead to the misleading conclusion that the effects are explained solely by either household- or school-level SES.14 Therefore, this study provides a novel approach by allowing a more detailed examination of the moderating role of a well-established risk factor at both levels. Specifically, we examine whether household- and school-level parental education moderate the effectiveness of the GBG in preventing the development of Dutch children’s emotional and behavioural problems from kindergarten to second grade.", "[SUBTITLE] Sample [SUBSECTION] Participants were recruited from the first 31 elementary schools in rural and urban areas of the Netherlands that agreed to participate in the research project. Schools could participate if they were willing to implement the GBG (if randomly selected in the intervention arm) or if they were willing to be on a waiting list (if randomly selected in the control arm).\nChildren’s emotional and behavioural problems were annually assessed for 3 years, from kindergarten (Mage=6.02, SD= 0.46) to second grade (in spring). Inclusion criteria were (i) active parental consent, (ii) data on school-level parental education and (iii) at least two out of three completed waves of teacher-reported data on emotional and behavioural problems. In total, out of 825 children who were initially included in the study, 731 (50% girls) fulfilled these criteria (see the flowchart in figure 1). All children had complete data on school-level parental education, 18.5% had missing data on household-level parental education and 24% had missing data on emotional and behavioural problems for one wave.\nFlowchart of the cluster randomized participants included in the randomized control trial, adapted with permission from Witvliet and colleagues30\nParticipants were recruited from the first 31 elementary schools in rural and urban areas of the Netherlands that agreed to participate in the research project. Schools could participate if they were willing to implement the GBG (if randomly selected in the intervention arm) or if they were willing to be on a waiting list (if randomly selected in the control arm).\nChildren’s emotional and behavioural problems were annually assessed for 3 years, from kindergarten (Mage=6.02, SD= 0.46) to second grade (in spring). Inclusion criteria were (i) active parental consent, (ii) data on school-level parental education and (iii) at least two out of three completed waves of teacher-reported data on emotional and behavioural problems. In total, out of 825 children who were initially included in the study, 731 (50% girls) fulfilled these criteria (see the flowchart in figure 1). All children had complete data on school-level parental education, 18.5% had missing data on household-level parental education and 24% had missing data on emotional and behavioural problems for one wave.\nFlowchart of the cluster randomized participants included in the randomized control trial, adapted with permission from Witvliet and colleagues30\n[SUBTITLE] Design and procedure [SUBSECTION] Participating schools were randomly assigned, with an oversampling of intervention schools, to either the control (10 schools, n = 247 children) or the GBG intervention arm (21 schools, n = 484 children). See Supplementary appendix A for sample size determination. The first assessments of emotional and behavioural problems were conducted in the Spring of 2004 when participants were in kindergarten (pre-intervention). In first and second grades, the GBG intervention was implemented and the second and third assessments were conducted.\nParticipating schools were randomly assigned, with an oversampling of intervention schools, to either the control (10 schools, n = 247 children) or the GBG intervention arm (21 schools, n = 484 children). See Supplementary appendix A for sample size determination. The first assessments of emotional and behavioural problems were conducted in the Spring of 2004 when participants were in kindergarten (pre-intervention). In first and second grades, the GBG intervention was implemented and the second and third assessments were conducted.\n[SUBTITLE] The GBG [SUBSECTION] The GBG is a classroom-based preventive intervention that aims to prevent disruptive behaviour by creating a positive and a predictable classroom environment where children work in teams and stimulate each other to show appropriate classroom behaviour. The GBG is implemented in classrooms by teachers for 15–60-min periods while students are working on regular school tasks. Before the GBG period, teachers and students select positively formulated classroom rules. Teachers then identify and assign children to teams of 4–5 students with an equal number of disruptive and non-disruptive children and give each team a set of cards. During the game, if a team member violates one of the preselected rules, teachers take a card from that team. Teams are rewarded at the end of the game period if at least one card remains. Teachers praise teams and children by complimenting appropriate behaviour and, aside from removing cards from teams that violate the rules, do not pay attention to disruptive behaviour. The GBG is implemented in three phases: introduction, expansion and generalization. In the introduction phase, the GBG is played three times a week. In the expansion and generalization phases, the duration (hours/days) is extended. More information regarding the intervention strategy, implementation and teacher training is described elsewhere.29\nThe GBG is a classroom-based preventive intervention that aims to prevent disruptive behaviour by creating a positive and a predictable classroom environment where children work in teams and stimulate each other to show appropriate classroom behaviour. The GBG is implemented in classrooms by teachers for 15–60-min periods while students are working on regular school tasks. Before the GBG period, teachers and students select positively formulated classroom rules. Teachers then identify and assign children to teams of 4–5 students with an equal number of disruptive and non-disruptive children and give each team a set of cards. During the game, if a team member violates one of the preselected rules, teachers take a card from that team. Teams are rewarded at the end of the game period if at least one card remains. Teachers praise teams and children by complimenting appropriate behaviour and, aside from removing cards from teams that violate the rules, do not pay attention to disruptive behaviour. The GBG is implemented in three phases: introduction, expansion and generalization. In the introduction phase, the GBG is played three times a week. In the expansion and generalization phases, the duration (hours/days) is extended. More information regarding the intervention strategy, implementation and teacher training is described elsewhere.29\n[SUBTITLE] Measures [SUBSECTION] \nHousehold-level parental education was based on the highest education level per household, obtained by the (two) parent/caregiver(s). Parental education levels were ranked according to the Dutch Standard Education Classification,30 which corresponds to the International Standard Classification of Education (ISCED).31 Following the ISCED classifications, parental education levels were coded using an 8-point scale, with education levels ranging from 0= no education/early education, 1= primary education, 2= lower secondary education, 3= upper secondary education, 4= post-secondary non-tertiary education, 5= short-cycle tertiary education, 6 =bachelor’s degree or equivalent, to 7 =master’s degree, equivalent or higher. The household parental education levels were reverse coded so that higher scores indicated lower parental education levels.\n\nSchool-level parental education levels were determined by the per-school percentage of children of low-educated parents. In the Netherlands, school-level socioeconomic inequalities are measured by children’s parents’ education levels. The Netherlands Inspectorate of Education calculates the percentage of low parental education levels of each school to identify schools that qualify for additional governmental resources.12 Low education is defined as parent(s) completing no more than elementary school. Thus, in this study, school-level parental education was based on the percentage score of low parental education levels of the entire school population. The percentage scores can range from 0% to 100%, with higher percentage scores indicating schools with higher percentages of children of low-educated parents. This information is publicly available (www.duo.nl).\n\nTeacher ratings of individual children’s behavioural and emotional problems were assessed by the Problem Behaviour at School Interview (PBSI).32 The PBSI is a validated questionnaire conducted via interview that uses a 5-point Likert scale ranging from 0 (never applicable) to 4 (often applicable).33 Behavioural problem scores were assessed by conduct problems (12 items) and oppositional defiant problems (7 items), and calculated as the average of the mean scores of the two subscales. Emotional problem scores were assessed by depression (7 items) and anxiety (5 items) symptoms, and the same procedure was followed. Higher scores indicated higher levels of emotional and behavioural problems. See Supplementary appendix A for more information regarding the PBSI and the outcome variables.\n\nIntervention status was dummy-coded (0 =control, 1 = GBG).\n\nCovariates included gender (0 =girls, 1 =boys) and cluster size. Cluster size (i.e. number of participating children per school) was grand-mean centred and included to account for unequal cluster sizes (M=23, range =8–88; mode =14, median =20). Baseline differences in kindergarten were controlled for because—despite randomization—children in the GBG arm had moderately higher levels of emotional [MGBG=0.85, SD=0.57; Mcontrol=0.67, SD=0.55, t(647)=−3.85, P<0.001, Cohen’s d=0.32] and slightly higher levels of behavioural problems [MGBG=0.80, SD=0.67; Mcontrol=0.69, SD=0.65, t(650)=−2.08, P=0.038, Cohen’s d=0.17] than children in the control arm.\n\nHousehold-level parental education was based on the highest education level per household, obtained by the (two) parent/caregiver(s). Parental education levels were ranked according to the Dutch Standard Education Classification,30 which corresponds to the International Standard Classification of Education (ISCED).31 Following the ISCED classifications, parental education levels were coded using an 8-point scale, with education levels ranging from 0= no education/early education, 1= primary education, 2= lower secondary education, 3= upper secondary education, 4= post-secondary non-tertiary education, 5= short-cycle tertiary education, 6 =bachelor’s degree or equivalent, to 7 =master’s degree, equivalent or higher. The household parental education levels were reverse coded so that higher scores indicated lower parental education levels.\n\nSchool-level parental education levels were determined by the per-school percentage of children of low-educated parents. In the Netherlands, school-level socioeconomic inequalities are measured by children’s parents’ education levels. The Netherlands Inspectorate of Education calculates the percentage of low parental education levels of each school to identify schools that qualify for additional governmental resources.12 Low education is defined as parent(s) completing no more than elementary school. Thus, in this study, school-level parental education was based on the percentage score of low parental education levels of the entire school population. The percentage scores can range from 0% to 100%, with higher percentage scores indicating schools with higher percentages of children of low-educated parents. This information is publicly available (www.duo.nl).\n\nTeacher ratings of individual children’s behavioural and emotional problems were assessed by the Problem Behaviour at School Interview (PBSI).32 The PBSI is a validated questionnaire conducted via interview that uses a 5-point Likert scale ranging from 0 (never applicable) to 4 (often applicable).33 Behavioural problem scores were assessed by conduct problems (12 items) and oppositional defiant problems (7 items), and calculated as the average of the mean scores of the two subscales. Emotional problem scores were assessed by depression (7 items) and anxiety (5 items) symptoms, and the same procedure was followed. Higher scores indicated higher levels of emotional and behavioural problems. See Supplementary appendix A for more information regarding the PBSI and the outcome variables.\n\nIntervention status was dummy-coded (0 =control, 1 = GBG).\n\nCovariates included gender (0 =girls, 1 =boys) and cluster size. Cluster size (i.e. number of participating children per school) was grand-mean centred and included to account for unequal cluster sizes (M=23, range =8–88; mode =14, median =20). Baseline differences in kindergarten were controlled for because—despite randomization—children in the GBG arm had moderately higher levels of emotional [MGBG=0.85, SD=0.57; Mcontrol=0.67, SD=0.55, t(647)=−3.85, P<0.001, Cohen’s d=0.32] and slightly higher levels of behavioural problems [MGBG=0.80, SD=0.67; Mcontrol=0.69, SD=0.65, t(650)=−2.08, P=0.038, Cohen’s d=0.17] than children in the control arm.\n[SUBTITLE] Statistical analyses [SUBSECTION] A parallel latent growth curve (LGM) model with two-level time-nested-within-individual data structure (1 =variation across individual children, 2 =variation across schools), in which the development of emotional and behavioural problems was conceptualized by latent growth parameters (intercept and a linear slope), was used to test the main effects and potential moderation by household- and school-level parental education of the GBG in preventing the development of emotional and behavioural problems. The intercept represented the initial level in kindergarten (baseline) and the slope represented change over time (from kindergarten to second grade).\nThe analyses were conducted in three steps. All models were fitted in Mplus version 8.0.34 We first computed design effects. Design effects larger than 2.0 indicate significant clustering of the data at the school level [Design effects =1+(nc−1)ICC].35 In the second step, we tested for main effects of the GBG intervention by regressing the outcome on the GBG intervention status, adjusting for the baseline differences in emotional and behavioural problems. In the third step, we tested moderation by household- and school-level parental education via a cross-level interaction and a between(school)-level interaction, respectively. Before examining cross-level interactions between household-level parental education and the GBG, we checked whether such interactions could be performed. To do this, we modelled a random slope at the (within)household-level and estimated its variance at the (between)school-level. This random slope represented the effect of household-level parental education on the growth parameters of children’s (individual-level) emotional or behavioural problems. Then, using Satorra Bentler chi-square difference tests, we checked whether adding a random slope improved the model fit of the main effect model in Step 2. If this was the case, the random slope parameter was regressed on the GBG at the between-level (i.e. cross-level interaction) to test the interaction between household-level parental education and the GBG on the development of individual-level emotional and behavioural problems. To test for moderation by school-level parental education at the between-level, an interaction term between school-level parental education and the GBG was added as a predictor of between-level emotional and behavioural problem development.\nModel fit indices for multi-level latent growth models were used to determine model fit at both the household and school levels. For specifics, see Supplementary appendix Btable S1. MLR estimators (maximum likelihood estimation with robust standard errors) were used to account for the possible non-normal distribution of data. Missing data were therefore handled using the default option in Mplus for MLR-estimation with missing at random data (i.e. Full Information Maximum Likelihood estimation). To ensure that the results were robust, two additional sensitivity tests were done: (i) by imputing the missing data in MPLUS (N = 25 imputed datasets) and (ii) by testing the models on a subsample (N=596) with complete household-level parental education data.\nA parallel latent growth curve (LGM) model with two-level time-nested-within-individual data structure (1 =variation across individual children, 2 =variation across schools), in which the development of emotional and behavioural problems was conceptualized by latent growth parameters (intercept and a linear slope), was used to test the main effects and potential moderation by household- and school-level parental education of the GBG in preventing the development of emotional and behavioural problems. The intercept represented the initial level in kindergarten (baseline) and the slope represented change over time (from kindergarten to second grade).\nThe analyses were conducted in three steps. All models were fitted in Mplus version 8.0.34 We first computed design effects. Design effects larger than 2.0 indicate significant clustering of the data at the school level [Design effects =1+(nc−1)ICC].35 In the second step, we tested for main effects of the GBG intervention by regressing the outcome on the GBG intervention status, adjusting for the baseline differences in emotional and behavioural problems. In the third step, we tested moderation by household- and school-level parental education via a cross-level interaction and a between(school)-level interaction, respectively. Before examining cross-level interactions between household-level parental education and the GBG, we checked whether such interactions could be performed. To do this, we modelled a random slope at the (within)household-level and estimated its variance at the (between)school-level. This random slope represented the effect of household-level parental education on the growth parameters of children’s (individual-level) emotional or behavioural problems. Then, using Satorra Bentler chi-square difference tests, we checked whether adding a random slope improved the model fit of the main effect model in Step 2. If this was the case, the random slope parameter was regressed on the GBG at the between-level (i.e. cross-level interaction) to test the interaction between household-level parental education and the GBG on the development of individual-level emotional and behavioural problems. To test for moderation by school-level parental education at the between-level, an interaction term between school-level parental education and the GBG was added as a predictor of between-level emotional and behavioural problem development.\nModel fit indices for multi-level latent growth models were used to determine model fit at both the household and school levels. For specifics, see Supplementary appendix Btable S1. MLR estimators (maximum likelihood estimation with robust standard errors) were used to account for the possible non-normal distribution of data. Missing data were therefore handled using the default option in Mplus for MLR-estimation with missing at random data (i.e. Full Information Maximum Likelihood estimation). To ensure that the results were robust, two additional sensitivity tests were done: (i) by imputing the missing data in MPLUS (N = 25 imputed datasets) and (ii) by testing the models on a subsample (N=596) with complete household-level parental education data.\n[SUBTITLE] Ethics [SUBSECTION] This study was approved by the Medical Ethics Committee of the Vrije Universiteit Amsterdam Medical Center and was registered with the ‘Netherlands Trial Register’ [Trial NL470 (NTR512)] (www.trialregister.nl). Signed parental informed consent was obtained from parents. Parents and children could revoke participation at any time.\nThis study was approved by the Medical Ethics Committee of the Vrije Universiteit Amsterdam Medical Center and was registered with the ‘Netherlands Trial Register’ [Trial NL470 (NTR512)] (www.trialregister.nl). Signed parental informed consent was obtained from parents. Parents and children could revoke participation at any time.", "Participants were recruited from the first 31 elementary schools in rural and urban areas of the Netherlands that agreed to participate in the research project. Schools could participate if they were willing to implement the GBG (if randomly selected in the intervention arm) or if they were willing to be on a waiting list (if randomly selected in the control arm).\nChildren’s emotional and behavioural problems were annually assessed for 3 years, from kindergarten (Mage=6.02, SD= 0.46) to second grade (in spring). Inclusion criteria were (i) active parental consent, (ii) data on school-level parental education and (iii) at least two out of three completed waves of teacher-reported data on emotional and behavioural problems. In total, out of 825 children who were initially included in the study, 731 (50% girls) fulfilled these criteria (see the flowchart in figure 1). All children had complete data on school-level parental education, 18.5% had missing data on household-level parental education and 24% had missing data on emotional and behavioural problems for one wave.\nFlowchart of the cluster randomized participants included in the randomized control trial, adapted with permission from Witvliet and colleagues30", "Participating schools were randomly assigned, with an oversampling of intervention schools, to either the control (10 schools, n = 247 children) or the GBG intervention arm (21 schools, n = 484 children). See Supplementary appendix A for sample size determination. The first assessments of emotional and behavioural problems were conducted in the Spring of 2004 when participants were in kindergarten (pre-intervention). In first and second grades, the GBG intervention was implemented and the second and third assessments were conducted.", "The GBG is a classroom-based preventive intervention that aims to prevent disruptive behaviour by creating a positive and a predictable classroom environment where children work in teams and stimulate each other to show appropriate classroom behaviour. The GBG is implemented in classrooms by teachers for 15–60-min periods while students are working on regular school tasks. Before the GBG period, teachers and students select positively formulated classroom rules. Teachers then identify and assign children to teams of 4–5 students with an equal number of disruptive and non-disruptive children and give each team a set of cards. During the game, if a team member violates one of the preselected rules, teachers take a card from that team. Teams are rewarded at the end of the game period if at least one card remains. Teachers praise teams and children by complimenting appropriate behaviour and, aside from removing cards from teams that violate the rules, do not pay attention to disruptive behaviour. The GBG is implemented in three phases: introduction, expansion and generalization. In the introduction phase, the GBG is played three times a week. In the expansion and generalization phases, the duration (hours/days) is extended. More information regarding the intervention strategy, implementation and teacher training is described elsewhere.29", "\nHousehold-level parental education was based on the highest education level per household, obtained by the (two) parent/caregiver(s). Parental education levels were ranked according to the Dutch Standard Education Classification,30 which corresponds to the International Standard Classification of Education (ISCED).31 Following the ISCED classifications, parental education levels were coded using an 8-point scale, with education levels ranging from 0= no education/early education, 1= primary education, 2= lower secondary education, 3= upper secondary education, 4= post-secondary non-tertiary education, 5= short-cycle tertiary education, 6 =bachelor’s degree or equivalent, to 7 =master’s degree, equivalent or higher. The household parental education levels were reverse coded so that higher scores indicated lower parental education levels.\n\nSchool-level parental education levels were determined by the per-school percentage of children of low-educated parents. In the Netherlands, school-level socioeconomic inequalities are measured by children’s parents’ education levels. The Netherlands Inspectorate of Education calculates the percentage of low parental education levels of each school to identify schools that qualify for additional governmental resources.12 Low education is defined as parent(s) completing no more than elementary school. Thus, in this study, school-level parental education was based on the percentage score of low parental education levels of the entire school population. The percentage scores can range from 0% to 100%, with higher percentage scores indicating schools with higher percentages of children of low-educated parents. This information is publicly available (www.duo.nl).\n\nTeacher ratings of individual children’s behavioural and emotional problems were assessed by the Problem Behaviour at School Interview (PBSI).32 The PBSI is a validated questionnaire conducted via interview that uses a 5-point Likert scale ranging from 0 (never applicable) to 4 (often applicable).33 Behavioural problem scores were assessed by conduct problems (12 items) and oppositional defiant problems (7 items), and calculated as the average of the mean scores of the two subscales. Emotional problem scores were assessed by depression (7 items) and anxiety (5 items) symptoms, and the same procedure was followed. Higher scores indicated higher levels of emotional and behavioural problems. See Supplementary appendix A for more information regarding the PBSI and the outcome variables.\n\nIntervention status was dummy-coded (0 =control, 1 = GBG).\n\nCovariates included gender (0 =girls, 1 =boys) and cluster size. Cluster size (i.e. number of participating children per school) was grand-mean centred and included to account for unequal cluster sizes (M=23, range =8–88; mode =14, median =20). Baseline differences in kindergarten were controlled for because—despite randomization—children in the GBG arm had moderately higher levels of emotional [MGBG=0.85, SD=0.57; Mcontrol=0.67, SD=0.55, t(647)=−3.85, P<0.001, Cohen’s d=0.32] and slightly higher levels of behavioural problems [MGBG=0.80, SD=0.67; Mcontrol=0.69, SD=0.65, t(650)=−2.08, P=0.038, Cohen’s d=0.17] than children in the control arm.", "A parallel latent growth curve (LGM) model with two-level time-nested-within-individual data structure (1 =variation across individual children, 2 =variation across schools), in which the development of emotional and behavioural problems was conceptualized by latent growth parameters (intercept and a linear slope), was used to test the main effects and potential moderation by household- and school-level parental education of the GBG in preventing the development of emotional and behavioural problems. The intercept represented the initial level in kindergarten (baseline) and the slope represented change over time (from kindergarten to second grade).\nThe analyses were conducted in three steps. All models were fitted in Mplus version 8.0.34 We first computed design effects. Design effects larger than 2.0 indicate significant clustering of the data at the school level [Design effects =1+(nc−1)ICC].35 In the second step, we tested for main effects of the GBG intervention by regressing the outcome on the GBG intervention status, adjusting for the baseline differences in emotional and behavioural problems. In the third step, we tested moderation by household- and school-level parental education via a cross-level interaction and a between(school)-level interaction, respectively. Before examining cross-level interactions between household-level parental education and the GBG, we checked whether such interactions could be performed. To do this, we modelled a random slope at the (within)household-level and estimated its variance at the (between)school-level. This random slope represented the effect of household-level parental education on the growth parameters of children’s (individual-level) emotional or behavioural problems. Then, using Satorra Bentler chi-square difference tests, we checked whether adding a random slope improved the model fit of the main effect model in Step 2. If this was the case, the random slope parameter was regressed on the GBG at the between-level (i.e. cross-level interaction) to test the interaction between household-level parental education and the GBG on the development of individual-level emotional and behavioural problems. To test for moderation by school-level parental education at the between-level, an interaction term between school-level parental education and the GBG was added as a predictor of between-level emotional and behavioural problem development.\nModel fit indices for multi-level latent growth models were used to determine model fit at both the household and school levels. For specifics, see Supplementary appendix Btable S1. MLR estimators (maximum likelihood estimation with robust standard errors) were used to account for the possible non-normal distribution of data. Missing data were therefore handled using the default option in Mplus for MLR-estimation with missing at random data (i.e. Full Information Maximum Likelihood estimation). To ensure that the results were robust, two additional sensitivity tests were done: (i) by imputing the missing data in MPLUS (N = 25 imputed datasets) and (ii) by testing the models on a subsample (N=596) with complete household-level parental education data.", "This study was approved by the Medical Ethics Committee of the Vrije Universiteit Amsterdam Medical Center and was registered with the ‘Netherlands Trial Register’ [Trial NL470 (NTR512)] (www.trialregister.nl). Signed parental informed consent was obtained from parents. Parents and children could revoke participation at any time.", "[SUBTITLE] Descriptive statistics [SUBSECTION] Descriptive statistics of household- and school-level parental education of the whole sample are presented in table 1. The household-level parental education levels were slightly higher in the control arm than in the GBG arm, t(1)=2.75, P=0.006, Cohen’s d=0.24.\nDescriptive statistics of household- and school-level parental education of the whole sample\nThe per-school percentage of children of low-educated parents was not significantly different between the schools in the control (M=18.61%, SD=23.97%) and intervention arms (M=15.35%, SD=17.02%), t(29)=0.44, P=0.666, Cohen’s d=0.17. The correlation between household- and school-level parental education in our sample was positive and of moderate magnitude (r=0.42, P<0.001).\nDescriptive statistics of household- and school-level parental education of the whole sample are presented in table 1. The household-level parental education levels were slightly higher in the control arm than in the GBG arm, t(1)=2.75, P=0.006, Cohen’s d=0.24.\nDescriptive statistics of household- and school-level parental education of the whole sample\nThe per-school percentage of children of low-educated parents was not significantly different between the schools in the control (M=18.61%, SD=23.97%) and intervention arms (M=15.35%, SD=17.02%), t(29)=0.44, P=0.666, Cohen’s d=0.17. The correlation between household- and school-level parental education in our sample was positive and of moderate magnitude (r=0.42, P<0.001).\n[SUBTITLE] Model building, unconditional latent growth models per condition and the GBG main effects [SUBSECTION] Intra-class correlations, design effect values, model fit indices of the unconditional LGMs for the whole sample and model building testing results are presented in Supplementary appendix Btable S1. Design effects indicated the need to use a two-level structure to analyze the data. Model fit indices were acceptable for both outcomes. Adding the random slope improved the model fit of the main effect model of emotional problems only, which indicated that cross-level interaction testing can be performed for emotional but not for behavioural problems.\nResults from the unconditional LGMs (Supplementary appendix Btable S2) showed that in the GBG arm emotional and behavioural problems stayed stable over time, as indicated by the non-significant slopes (emotional problems: B=0.065, P=0.115; behavioural problems: B=−0.041, P=0.177). In the control arm, there was a significant yearly increase of emotional problems (B = 0.271, P < 0.001) and a borderline significant yearly increase of behavioural problems (B=0.100, P=0.057). This indicates that without the GBG, emotional (and to a lesser extent behavioural) problems tended to increase from kindergarten to second grade.\nResults of main effects (table 2) showed that the GBG was effective in preventing the increase in emotional problems that was found in the control group [B=−0.208, 95% CI (−0.345, −0.070), P = 0.003]. In addition, the GBG was also effective in preventing behavioural problems from kindergarten to second grade [B=−0.133, 95% CI (−0.256, −0.010), P=0.034].\nMain effects of the GBG and moderation by household- and school-level parental education on children’s emotional and behavioural problems\n\nNote: *P < .05, **P < .01, ***P < .001; n.a. = estimate not available due to the use of Monte Carlo integration to estimate cross-level interaction. Note that the effect of school-level parental education is small because it represents the effect at 1% change in school-level parental education.\nIntra-class correlations, design effect values, model fit indices of the unconditional LGMs for the whole sample and model building testing results are presented in Supplementary appendix Btable S1. Design effects indicated the need to use a two-level structure to analyze the data. Model fit indices were acceptable for both outcomes. Adding the random slope improved the model fit of the main effect model of emotional problems only, which indicated that cross-level interaction testing can be performed for emotional but not for behavioural problems.\nResults from the unconditional LGMs (Supplementary appendix Btable S2) showed that in the GBG arm emotional and behavioural problems stayed stable over time, as indicated by the non-significant slopes (emotional problems: B=0.065, P=0.115; behavioural problems: B=−0.041, P=0.177). In the control arm, there was a significant yearly increase of emotional problems (B = 0.271, P < 0.001) and a borderline significant yearly increase of behavioural problems (B=0.100, P=0.057). This indicates that without the GBG, emotional (and to a lesser extent behavioural) problems tended to increase from kindergarten to second grade.\nResults of main effects (table 2) showed that the GBG was effective in preventing the increase in emotional problems that was found in the control group [B=−0.208, 95% CI (−0.345, −0.070), P = 0.003]. In addition, the GBG was also effective in preventing behavioural problems from kindergarten to second grade [B=−0.133, 95% CI (−0.256, −0.010), P=0.034].\nMain effects of the GBG and moderation by household- and school-level parental education on children’s emotional and behavioural problems\n\nNote: *P < .05, **P < .01, ***P < .001; n.a. = estimate not available due to the use of Monte Carlo integration to estimate cross-level interaction. Note that the effect of school-level parental education is small because it represents the effect at 1% change in school-level parental education.\n[SUBTITLE] Moderation by household- and school-level parental education of the GBG impact [SUBSECTION] \nHousehold level: Results showed no significant cross-level interaction between household-level parental education and the GBG-effect on individual-level emotional problem development, B=0.010, 95% CI (−0.055, 0.074), P=0.765 (see table 2). The cross-level interaction for behavioural problems was not tested.\n\nSchool level: Results showed a significant interaction between school-level parental education and the GBG-effect on children’s emotional problems, B=0.007, 95% CI (0.002, 0.013), P = 0.005 (see table 2). That is, the GBG was more effective in preventing the development of emotional problems in higher parental education schools than in lower parental education schools. Figure 2A shows a visual representation of this interaction effect in which the effects were probed at 0.50 SD above [lower parental education schools; ∼26% of the total sample; B=−0.140, SE=0.059, 95% CI (−0.255, −0.026), P = 0.016] and at 0.50 SD below the mean of school-level parental education [higher parental education schools; ∼7% of the total sample; B=−0.281, SE=0.080, 95% CI (−0.438, −0.124), P < 0.001]. For behavioural problems, no moderation between school-level parental education and the GBG was found, B=0.002, 95% CI (−0.003, 0.007), P = 0.382 (see figure 2B). The two sensitivity tests showed no changes in interpretation of the results. For specifics, see Supplementary appendix Btables S3 and S4.\nSchool-level parental education effects on the development of emotional problems (A) and behavioural problems (B) in GBG vs. control arms\n\nHousehold level: Results showed no significant cross-level interaction between household-level parental education and the GBG-effect on individual-level emotional problem development, B=0.010, 95% CI (−0.055, 0.074), P=0.765 (see table 2). The cross-level interaction for behavioural problems was not tested.\n\nSchool level: Results showed a significant interaction between school-level parental education and the GBG-effect on children’s emotional problems, B=0.007, 95% CI (0.002, 0.013), P = 0.005 (see table 2). That is, the GBG was more effective in preventing the development of emotional problems in higher parental education schools than in lower parental education schools. Figure 2A shows a visual representation of this interaction effect in which the effects were probed at 0.50 SD above [lower parental education schools; ∼26% of the total sample; B=−0.140, SE=0.059, 95% CI (−0.255, −0.026), P = 0.016] and at 0.50 SD below the mean of school-level parental education [higher parental education schools; ∼7% of the total sample; B=−0.281, SE=0.080, 95% CI (−0.438, −0.124), P < 0.001]. For behavioural problems, no moderation between school-level parental education and the GBG was found, B=0.002, 95% CI (−0.003, 0.007), P = 0.382 (see figure 2B). The two sensitivity tests showed no changes in interpretation of the results. For specifics, see Supplementary appendix Btables S3 and S4.\nSchool-level parental education effects on the development of emotional problems (A) and behavioural problems (B) in GBG vs. control arms", "Descriptive statistics of household- and school-level parental education of the whole sample are presented in table 1. The household-level parental education levels were slightly higher in the control arm than in the GBG arm, t(1)=2.75, P=0.006, Cohen’s d=0.24.\nDescriptive statistics of household- and school-level parental education of the whole sample\nThe per-school percentage of children of low-educated parents was not significantly different between the schools in the control (M=18.61%, SD=23.97%) and intervention arms (M=15.35%, SD=17.02%), t(29)=0.44, P=0.666, Cohen’s d=0.17. The correlation between household- and school-level parental education in our sample was positive and of moderate magnitude (r=0.42, P<0.001).", "Intra-class correlations, design effect values, model fit indices of the unconditional LGMs for the whole sample and model building testing results are presented in Supplementary appendix Btable S1. Design effects indicated the need to use a two-level structure to analyze the data. Model fit indices were acceptable for both outcomes. Adding the random slope improved the model fit of the main effect model of emotional problems only, which indicated that cross-level interaction testing can be performed for emotional but not for behavioural problems.\nResults from the unconditional LGMs (Supplementary appendix Btable S2) showed that in the GBG arm emotional and behavioural problems stayed stable over time, as indicated by the non-significant slopes (emotional problems: B=0.065, P=0.115; behavioural problems: B=−0.041, P=0.177). In the control arm, there was a significant yearly increase of emotional problems (B = 0.271, P < 0.001) and a borderline significant yearly increase of behavioural problems (B=0.100, P=0.057). This indicates that without the GBG, emotional (and to a lesser extent behavioural) problems tended to increase from kindergarten to second grade.\nResults of main effects (table 2) showed that the GBG was effective in preventing the increase in emotional problems that was found in the control group [B=−0.208, 95% CI (−0.345, −0.070), P = 0.003]. In addition, the GBG was also effective in preventing behavioural problems from kindergarten to second grade [B=−0.133, 95% CI (−0.256, −0.010), P=0.034].\nMain effects of the GBG and moderation by household- and school-level parental education on children’s emotional and behavioural problems\n\nNote: *P < .05, **P < .01, ***P < .001; n.a. = estimate not available due to the use of Monte Carlo integration to estimate cross-level interaction. Note that the effect of school-level parental education is small because it represents the effect at 1% change in school-level parental education.", "\nHousehold level: Results showed no significant cross-level interaction between household-level parental education and the GBG-effect on individual-level emotional problem development, B=0.010, 95% CI (−0.055, 0.074), P=0.765 (see table 2). The cross-level interaction for behavioural problems was not tested.\n\nSchool level: Results showed a significant interaction between school-level parental education and the GBG-effect on children’s emotional problems, B=0.007, 95% CI (0.002, 0.013), P = 0.005 (see table 2). That is, the GBG was more effective in preventing the development of emotional problems in higher parental education schools than in lower parental education schools. Figure 2A shows a visual representation of this interaction effect in which the effects were probed at 0.50 SD above [lower parental education schools; ∼26% of the total sample; B=−0.140, SE=0.059, 95% CI (−0.255, −0.026), P = 0.016] and at 0.50 SD below the mean of school-level parental education [higher parental education schools; ∼7% of the total sample; B=−0.281, SE=0.080, 95% CI (−0.438, −0.124), P < 0.001]. For behavioural problems, no moderation between school-level parental education and the GBG was found, B=0.002, 95% CI (−0.003, 0.007), P = 0.382 (see figure 2B). The two sensitivity tests showed no changes in interpretation of the results. For specifics, see Supplementary appendix Btables S3 and S4.\nSchool-level parental education effects on the development of emotional problems (A) and behavioural problems (B) in GBG vs. control arms", "Overall, the GBG prevented the development of emotional and behavioural problems from kindergarten to second grade. Specifically, results showed that the effectiveness of the GBG in preventing emotional and behavioural problems did not differ between children of lower- and higher-educated parents. Nevertheless, the GBG was more effective in schools with a lower (compared to higher) percentage of children of lower-educated parents, albeit only for emotional problems.\nTo our knowledge, this study provides preliminary evidence that school-level parental education may impact the effectiveness of the GBG in reducing emotional problems. Previous studies mainly tested household/individual-level factors, such as gender, initial risk status and behaviour type as moderators of universal school-based programmes like the GBG.36,37 This study suggests that more attention needs to be directed towards lower parental education schools and that in addition to individual-level moderators, school-level moderators should be studied to better understand the potential differential impact of universal school-based interventions.\nThe characteristics of lower and higher parental education schools may explain why the GBG was less effective in lower parental education schools for emotional problems. Lower parental education schools may have fewer resources, less effective school management, less teacher support and teachers who are insufficiently prepared to deal with such schools’ diverse populations.16,38 Nevertheless, this study cannot explain why the school-level interaction effect was found for emotional but not for behavioural problems. It stands to reason that the GBG is more directed towards behavioural problems. Thus, it may be less affected by possible school-level factors that may attenuate its impact. However, we should be cautious in interpreting the results before replication studies with longer follow-up procedures are conducted.\nThe following limitations should be noted. First, and most importantly, we did not have implementation fidelity data. It is possible that there were no major differences in implementation fidelity based on school-level parental education since the interaction effect between school-level parental education and the GBG on behavioural and emotional problems differed. Our study should be considered as an effectiveness trial and an exploratory study meant to stimulate further investigation. It is important to study, for instance, whether the GBG’s weaker effect on emotional problems in lower parental education schools is due to (i) specific school-level factors, (ii) possible problems with implementation or (iii) to lack of components more directly targeting emotional problems. Second, our sample was not randomly drawn from the Dutch population of elementary schools. Third, we used teacher-reports and teachers were not blinded to condition. Self-reports and observational data, which could have provided additional insights, were not available. Fourth, sample size at the between-level was relatively small with 31 schools. Due to this we did not, for example, have enough power to test a three-way interaction of the GBG, household- and school-level parental education. Finally, we used parental education as an index of broader SES. Future replication studies are encouraged to use broader SES indices.\nDespite these limitations, our results have implications for research and practice. Testing implementation fidelity and school-level moderators that relate to lower parental education schools would result in determining the specific factors to be addressed, such as teacher support and training or implementation infrastructure in schools. Furthermore, if lower parental education schools need more support in preventing emotional problems, more intensified or selective interventions that target high-risk populations could be implemented in these schools. Nevertheless, it is noteworthy that for general prevention efforts the GBG was equally effective in preventing behavioural and emotional problems irrespective of household-level parental education and in preventing behavioural problems irrespective of school-level parental education. Although results suggested that the GBG was less effective in lower parental education schools, it still was an effective tool for preventing the development of emotional problems in these schools. School-based universal interventions reduce the potential that children who may be at risk of developing mental health problems or who may be otherwise difficult to reach will be overlooked. For instance, despite the need for mental health services, it has been shown that the majority of low SES children do not receive treatment.39 At a time in which SES-related inequalities are on the rise,40 this study shows that the GBG is effective in preventing the development of behavioural and emotional problems of children in lower- and higher-educated households and schools while suggesting that more attention should be directed towards lower parental education schools.", "\nSupplementary data are available at EURPUB online.", "This study was supported by a grant from the Netherlands Organization for Health Research and Development (ZonMw) (project No. 531003013) and by ZonMw Grants #26200002 and #120620029.T.A.J.H. was funded through a grant awarded by the Norwegian Research Council (project number 288638) to the Centre for Global Health Inequalities Research (CHAIN) at the Norwegian University for Science and Technology (NTNU).\n\nConflicts of interest: None declared.\nThe effectiveness of the GBG in preventing the development of behavioural and emotional problems did not differ between children of lower- and higher-educated parents from kindergarten to second grade.\nThe GBG is equally effective in preventing the development of behavioural problems in schools with higher and lower percentages of children with lower-educated parents, but less effective in preventing the development of emotional problems in lower parental education schools than in higher parental education schools.\nWhen testing intervention effectiveness, school-level variables as moderators should be included in study designs.\nMore attention should be directed towards schools with a higher percentage of children with lower-educated parents.", "Click here for additional data file." ]
[ "intro", "methods", null, null, null, null, null, null, "results", null, null, null, "discussion", null, null, "supplementary-material" ]
[]
Laparoscopic Versus Abdominal Radical Hysterectomy for Cervical Cancer: A Meta-analysis of Randomized Controlled Trials.
36256872
Laparoscopic radical hysterectomy (LRH) and open abdominal radical hysterectomy (ARH) have been used for cervical cancer treatment. We aimed to perform a meta-analysis to compare the efficacy and safety of LRH and ARH in the treatment of cervical cancer to provide reliable evidence to the clinical cervical cancer treatment.
BACKGROUND
Two investigators independently searched PubMed and other databases for randomized controlled trials (RCTs) comparing LRH and ARH for cervical cancer treatment up to May 31, 2022. The risk of bias assessment tool recommended by Cochrane library was used for quality assessment. RevMan 5.3 software was used for meta-analysis.
METHODS
Fourteen RCTs with a total of 1700 patients with cervical cancer were finally included. Meta-analyses indicated that compared with ARH, LRH reduced the intraoperative blood loss (mean difference [MD]=-58.08; 95% CI, -70.91, -45.24), the time to first passage of flatus (MD=-14.50; 95% CI, -16.55, -12.44) (all P <0.05), and increase the number of lymph nodes removed (MD=3.47; 95% CI, 0.51, 6.43; P =0.02). There were no significant differences in the duration of surgery (MD=27.62; 95% CI, -6.26, 61.49), intraoperative complications (odd ratio [OR]=1.10; 95% CI, 0.17, 7.32), postoperative complications (OR=0.78; 95% CI, 0.33, 1.86), relapse rate (OR=1.45; 95% CI, 0.56, 3.74), and survival rate (OR=0.75; 95% CI, 0.52, 1.08) between LRH group and ARH group (all P >0.05).
RESULTS
LRH has more advantages over ARH in the treatment of cervical cancer. Still, the long-term effects and safety of LRH and ARH need more high-quality, large-sample RCTs to be further verified.
CONCLUSIONS
[ "Female", "Humans", "Uterine Cervical Neoplasms", "Neoplasm Staging", "Retrospective Studies", "Neoplasm Recurrence, Local", "Randomized Controlled Trials as Topic", "Hysterectomy", "Laparoscopy" ]
9624383
null
null
METHODS
This meta-analysis and systematic review was conducted in comply with the statement of preferred reporting items for systematic reviews and meta-analyses13 (Supplemental Digital Content 1, http://links.lww.com/AJCO/A427 and Supplemental Digital Content 2, http://links.lww.com/AJCO/A428). [SUBTITLE] Search Strategy [SUBSECTION] The 2 authors independently searched PubMed, Web of Science, EMbase, Cochrane Library, China Biomedical Database, Chinese National Knowledge Infrastructure (CNKI), Wanfang, and Weipu databases for randomized controlled trials (RCTs) on the efficacy and safety of LRH and ARH for cervical cancer treatment. The retrieval time limit is from the establishment of the database to May 31, 2022. The language was limited to Chinese or English. The search terms used were as following: (“cervical cancer” OR “cervical carcinoma” OR “uterine cervical neoplasms” OR “invasive carcinoma of cervix uteri”) AND (“laparoscopy” OR “laparotomy” OR “surgery” OR “abdominal” OR “open”). The 2 authors independently searched PubMed, Web of Science, EMbase, Cochrane Library, China Biomedical Database, Chinese National Knowledge Infrastructure (CNKI), Wanfang, and Weipu databases for randomized controlled trials (RCTs) on the efficacy and safety of LRH and ARH for cervical cancer treatment. The retrieval time limit is from the establishment of the database to May 31, 2022. The language was limited to Chinese or English. The search terms used were as following: (“cervical cancer” OR “cervical carcinoma” OR “uterine cervical neoplasms” OR “invasive carcinoma of cervix uteri”) AND (“laparoscopy” OR “laparotomy” OR “surgery” OR “abdominal” OR “open”). [SUBTITLE] Inclusion and Exclusion Criteria [SUBSECTION] The inclusion criteria for this meta-analysis were as follows: (1) the type of study was a RCT, the language was limited to Chinese and English, (2) the patients were clinically diagnosed with cervical cancer confirmed by pathology and underwent radical hysterectomy (3) LRH was used for the intervention measures in the experimental group, and ARH was used for the intervention measures in the control group, (4) The article reported relevant outcome indicators, such as operation time, intraoperative blood loss, number of lymph node biopsies, recurrence rate, and survival rate. The exclusion criteria for this meta-analysis were as follows: (1) review, case, and basic experimental studies; (2) duplicate published literature reports; (3) research reports for which data could not be extracted. The inclusion criteria for this meta-analysis were as follows: (1) the type of study was a RCT, the language was limited to Chinese and English, (2) the patients were clinically diagnosed with cervical cancer confirmed by pathology and underwent radical hysterectomy (3) LRH was used for the intervention measures in the experimental group, and ARH was used for the intervention measures in the control group, (4) The article reported relevant outcome indicators, such as operation time, intraoperative blood loss, number of lymph node biopsies, recurrence rate, and survival rate. The exclusion criteria for this meta-analysis were as follows: (1) review, case, and basic experimental studies; (2) duplicate published literature reports; (3) research reports for which data could not be extracted. [SUBTITLE] Quality Assessment [SUBSECTION] The RCTs included in this study were assessed using the risk of bias assessment tool for RCTs recommended by Cochrane library.14 The evaluated items in this tool include (1) the method of generating random sequences; (2) whether the allocation scheme is hidden; (3)whether the personnel is blinded; (4) whether the outcome assessment is blinded; (5) whether the outcome data is complete; (6) whether the results are selectively reported; and (7) whether there is any other bias. For each study result, a choice was made on the above 7 items, including “Yes” (low bias), “No” (high bias), and “Unclear” (lack of relevant information or uncertain bias). The RCTs included in this study were assessed using the risk of bias assessment tool for RCTs recommended by Cochrane library.14 The evaluated items in this tool include (1) the method of generating random sequences; (2) whether the allocation scheme is hidden; (3)whether the personnel is blinded; (4) whether the outcome assessment is blinded; (5) whether the outcome data is complete; (6) whether the results are selectively reported; and (7) whether there is any other bias. For each study result, a choice was made on the above 7 items, including “Yes” (low bias), “No” (high bias), and “Unclear” (lack of relevant information or uncertain bias). [SUBTITLE] Literature Screening and Data Extraction [SUBSECTION] In this meta-analysis, 2 reviewers independently screened the literature and extracted data and cross-checked them. In case of disagreement, a third party was consulted to assist in judgment, and the first original authors would be contacted as much as possible to supplement the lack of data. We extracted the following data: basic information of included studies, including research title, first author, publication journal and time, etc.; basic characteristics of research subjects, including age, number of cases, FIGO stage; Key elements of study design type, and quality assessment; relevant outcome data. In this meta-analysis, 2 reviewers independently screened the literature and extracted data and cross-checked them. In case of disagreement, a third party was consulted to assist in judgment, and the first original authors would be contacted as much as possible to supplement the lack of data. We extracted the following data: basic information of included studies, including research title, first author, publication journal and time, etc.; basic characteristics of research subjects, including age, number of cases, FIGO stage; Key elements of study design type, and quality assessment; relevant outcome data. [SUBTITLE] Statistical Analysis [SUBSECTION] This study used RevMan 5.3 software for meta-analysis. For enumeration data, odd ratio (OR) and its 95% confidence interval (CI) were used as effect size. For measurement data, mean difference (MD) and its 95% CI were used as effect size. The χ2 test was used to analyze the heterogeneity between the results of the studies. If there was no statistical heterogeneity among the results of each study (P>0.10, I 2 ≤50%), a fixed-effects model was used for meta-analysis; otherwise, a random-effects model was used for meta-analysis. Publication bias was evaluated by the demonstration of funnel plots, and asymmetry was assessed by means of the Egger regression test. P<0.05 indicated that the difference between groups was statistically significant. This study used RevMan 5.3 software for meta-analysis. For enumeration data, odd ratio (OR) and its 95% confidence interval (CI) were used as effect size. For measurement data, mean difference (MD) and its 95% CI were used as effect size. The χ2 test was used to analyze the heterogeneity between the results of the studies. If there was no statistical heterogeneity among the results of each study (P>0.10, I 2 ≤50%), a fixed-effects model was used for meta-analysis; otherwise, a random-effects model was used for meta-analysis. Publication bias was evaluated by the demonstration of funnel plots, and asymmetry was assessed by means of the Egger regression test. P<0.05 indicated that the difference between groups was statistically significant.
RESULTS
[SUBTITLE] RCT Selection [SUBSECTION] A total of 238 literatures were initially detected in the first literature search. After reading the titles and abstracts, 58 papers were included after excluding the literatures that did not meet the inclusion criteria. After reading the full text of the literature, 14 RCTs15–28 were finally included (Fig. 1). Of the included 14 RCTs, a total of 1700 patients with cervical cancer were involved, 852 patients underwent LRH treatment, 848 patients underwent ARH treatment. The basic characteristics of the included studies are shown in Table 1. Preferred reporting items for systematic reviews and meta-analyses flow diagram of randomized controlled trialselection. The Characteristics of Included Randomized Controlled Trials ①duration of surgery; ②intraoperative blood loss; ③number of lymph nodes removed; ④intraoperative complications; ⑤postoperative complications; ⑥the time to first passage of flatus; ⑦relapse rate; ⑧survival rate. ARH indicates abdominal radical hysterectomy; LRH, laparoscopic radical hysterectomy; NA, not available. A total of 238 literatures were initially detected in the first literature search. After reading the titles and abstracts, 58 papers were included after excluding the literatures that did not meet the inclusion criteria. After reading the full text of the literature, 14 RCTs15–28 were finally included (Fig. 1). Of the included 14 RCTs, a total of 1700 patients with cervical cancer were involved, 852 patients underwent LRH treatment, 848 patients underwent ARH treatment. The basic characteristics of the included studies are shown in Table 1. Preferred reporting items for systematic reviews and meta-analyses flow diagram of randomized controlled trialselection. The Characteristics of Included Randomized Controlled Trials ①duration of surgery; ②intraoperative blood loss; ③number of lymph nodes removed; ④intraoperative complications; ⑤postoperative complications; ⑥the time to first passage of flatus; ⑦relapse rate; ⑧survival rate. ARH indicates abdominal radical hysterectomy; LRH, laparoscopic radical hysterectomy; NA, not available. [SUBTITLE] Literature Quality Evaluation [SUBSECTION] Of all the 14 included RCTs, 11 RCTs15–22,24–26 described specific methods for generating random sequences, and another 3 RCTs did not mention specific randomization methods. Only 6 RCTs reported specific allocation concealment methods. Blinding of participants and personnel and outcome assessment (detection bias) were not stated in the included RCTs. No loss to follow-up and selective outcomes were reported. The results of the literature quality evaluation are shown in Figures 2 and 3. Risk of bias graph. Risk of bias summary. Of all the 14 included RCTs, 11 RCTs15–22,24–26 described specific methods for generating random sequences, and another 3 RCTs did not mention specific randomization methods. Only 6 RCTs reported specific allocation concealment methods. Blinding of participants and personnel and outcome assessment (detection bias) were not stated in the included RCTs. No loss to follow-up and selective outcomes were reported. The results of the literature quality evaluation are shown in Figures 2 and 3. Risk of bias graph. Risk of bias summary. [SUBTITLE] Meta-analysis [SUBSECTION] [SUBTITLE] Duration of Surgery [SUBSECTION] Eleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I 2 = 99%). Forest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy. Eleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I 2 = 99%). Forest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy. [SUBTITLE] Intraoperative Blood Loss [SUBSECTION] Ten included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I 2 = 73%). Ten included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I 2 = 73%). [SUBTITLE] Number of Lymph Nodes Removed [SUBSECTION] Eight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I 2 = 97%). Eight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I 2 = 97%). [SUBTITLE] The Time to First Passage of Flatus [SUBSECTION] Seven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I 2 = 70%). Seven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I 2 = 70%). [SUBTITLE] Intraoperative Complications [SUBSECTION] Three included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I 2 =53%). Forest plots for synthesized outcomes. Three included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I 2 =53%). Forest plots for synthesized outcomes. [SUBTITLE] Postoperative Complications [SUBSECTION] Three included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I 2 =37%). Three included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I 2 =37%). [SUBTITLE] Relapse Rate [SUBSECTION] Five included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I 2=54%). Five included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I 2=54%). [SUBTITLE] Survival Rate [SUBSECTION] Six included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I 2 =48%). Six included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I 2 =48%). [SUBTITLE] Duration of Surgery [SUBSECTION] Eleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I 2 = 99%). Forest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy. Eleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I 2 = 99%). Forest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy. [SUBTITLE] Intraoperative Blood Loss [SUBSECTION] Ten included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I 2 = 73%). Ten included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I 2 = 73%). [SUBTITLE] Number of Lymph Nodes Removed [SUBSECTION] Eight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I 2 = 97%). Eight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I 2 = 97%). [SUBTITLE] The Time to First Passage of Flatus [SUBSECTION] Seven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I 2 = 70%). Seven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I 2 = 70%). [SUBTITLE] Intraoperative Complications [SUBSECTION] Three included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I 2 =53%). Forest plots for synthesized outcomes. Three included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I 2 =53%). Forest plots for synthesized outcomes. [SUBTITLE] Postoperative Complications [SUBSECTION] Three included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I 2 =37%). Three included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I 2 =37%). [SUBTITLE] Relapse Rate [SUBSECTION] Five included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I 2=54%). Five included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I 2=54%). [SUBTITLE] Survival Rate [SUBSECTION] Six included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I 2 =48%). Six included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I 2 =48%). [SUBTITLE] Publication Bias [SUBSECTION] As showed in Figures 6 and 7, the dots in the funnel plots for synthesized outcomes were evenly distributed, and the results of Egger regression tests indicated that there was no publication bias in the synthesized outcomes (all P>0.05). Funnel plots for synthesized outcomes. MD indicates mean difference; OD, odd ratio. Funnel plots for synthesized outcomes. OD indicates odd ratio. As showed in Figures 6 and 7, the dots in the funnel plots for synthesized outcomes were evenly distributed, and the results of Egger regression tests indicated that there was no publication bias in the synthesized outcomes (all P>0.05). Funnel plots for synthesized outcomes. MD indicates mean difference; OD, odd ratio. Funnel plots for synthesized outcomes. OD indicates odd ratio. [SUBTITLE] Sensitivity Analyses [SUBSECTION] Sensitivity analyses that assess the impact of single 1 study on the overall risk estimate by removing 1 study in each turn. No significant change of the overall risk estimates by removing any single study was found. Sensitivity analyses that assess the impact of single 1 study on the overall risk estimate by removing 1 study in each turn. No significant change of the overall risk estimates by removing any single study was found.
CONCLUSIONS
In conclusion, the results of this meta-analysis have found that LRH is beneficial to reduce the intraoperative blood loss, shorten the recovery time of postoperative gastrointestinal function, and increase the number of lymph nodes removed in patients with cervical cancer compared with ARH. Future research should be based on the patients’ safety and quality of life, focusing on postoperative overall survival time, tumor-free survival time, and quality of life of patients with cervical cancer. It is suggested that more RCTs should be carried out on the efficacy of LRH and ARH on postoperative quality of life and other outcomes with larger sample size and longer follow-up.
[ "BACKGROUND", "Search Strategy", "Inclusion and Exclusion Criteria", "Quality Assessment", "Literature Screening and Data Extraction", "Statistical Analysis", "RCT Selection", "Literature Quality Evaluation", "Meta-analysis", "Duration of Surgery", "Intraoperative Blood Loss", "Number of Lymph Nodes Removed", "The Time to First Passage of Flatus", "Intraoperative Complications", "Postoperative Complications", "Relapse Rate", "Survival Rate", "Publication Bias", "Sensitivity Analyses", "DISCUSSIONS" ]
[ "Cervical cancer is one of the most common malignant tumors that seriously threaten women’s health, and its incidence ranks third among female malignant tumors in the world, second only to breast cancer and colorectal cancer.1,2 There are about 500,000 new cases of cervical cancer every year in the world, 85% of which are in developing countries, and about 250,000 people die of cervical cancer each year.3,4 In China, 140,000 new cases are discovered every year, accounting for about one third of the global patients with cervical cancer, and about 50,000 patients die of cervical cancer every year.5,6 In recent years, the incidence of cervical cancer has a younger trend.7,8 Therefore, the treatment and care of cervical cancer is of great significance to the prognosis of patients.\nRadical hysterectomy and lymph node dissection are the surgical methods recommended by the International Federation of Obstetrics and Gynaecology (FIGO) for the treatment of early-stage cervical cancer.9 Since the first successful laparoscopic pelvic lymph node dissection for cervical cancer in 1989, minimally invasive techniques have developed rapidly. In 1991, laparoscopy was first used for pelvic lymph node dissection combined with vaginal hysterectomy in the treatment of early cervical cancer. Laparoscopy has been used for total hysterectomy and pelvic lymph node dissection in the treatment of cervical cancer. Since then, many studies10–12 have shown that laparoscopic surgery has the advantages of less trauma, faster recovery, and less pain for patients after surgery, which has been valued by the majority of obstetrics and gynecologists. In recent years, many researchers have conducted many studies on the clinical efficacy and safety of laparoscopic surgery for cervical cancer. However, because of the small number of independent research samples and uneven research quality limits the results promotion. So far, no large-sample, multicenter clinical trials have been reported. To this end, we aimed to conduct a meta-analysis to compare the efficacy and safety of laparoscopic radical hysterectomy (LRH) and open abdominal radical hysterectomy (ARH) in the treatment of cervical cancer to provide a reliable basis for the selection of clinical surgical methods for cervical cancer treatment.", "The 2 authors independently searched PubMed, Web of Science, EMbase, Cochrane Library, China Biomedical Database, Chinese National Knowledge Infrastructure (CNKI), Wanfang, and Weipu databases for randomized controlled trials (RCTs) on the efficacy and safety of LRH and ARH for cervical cancer treatment. The retrieval time limit is from the establishment of the database to May 31, 2022. The language was limited to Chinese or English. The search terms used were as following: (“cervical cancer” OR “cervical carcinoma” OR “uterine cervical neoplasms” OR “invasive carcinoma of cervix uteri”) AND (“laparoscopy” OR “laparotomy” OR “surgery” OR “abdominal” OR “open”).", "The inclusion criteria for this meta-analysis were as follows: (1) the type of study was a RCT, the language was limited to Chinese and English, (2) the patients were clinically diagnosed with cervical cancer confirmed by pathology and underwent radical hysterectomy (3) LRH was used for the intervention measures in the experimental group, and ARH was used for the intervention measures in the control group, (4) The article reported relevant outcome indicators, such as operation time, intraoperative blood loss, number of lymph node biopsies, recurrence rate, and survival rate. The exclusion criteria for this meta-analysis were as follows: (1) review, case, and basic experimental studies; (2) duplicate published literature reports; (3) research reports for which data could not be extracted.", "The RCTs included in this study were assessed using the risk of bias assessment tool for RCTs recommended by Cochrane library.14 The evaluated items in this tool include (1) the method of generating random sequences; (2) whether the allocation scheme is hidden; (3)whether the personnel is blinded; (4) whether the outcome assessment is blinded; (5) whether the outcome data is complete; (6) whether the results are selectively reported; and (7) whether there is any other bias. For each study result, a choice was made on the above 7 items, including “Yes” (low bias), “No” (high bias), and “Unclear” (lack of relevant information or uncertain bias).", "In this meta-analysis, 2 reviewers independently screened the literature and extracted data and cross-checked them. In case of disagreement, a third party was consulted to assist in judgment, and the first original authors would be contacted as much as possible to supplement the lack of data. We extracted the following data: basic information of included studies, including research title, first author, publication journal and time, etc.; basic characteristics of research subjects, including age, number of cases, FIGO stage; Key elements of study design type, and quality assessment; relevant outcome data.", "This study used RevMan 5.3 software for meta-analysis. For enumeration data, odd ratio (OR) and its 95% confidence interval (CI) were used as effect size. For measurement data, mean difference (MD) and its 95% CI were used as effect size. The χ2 test was used to analyze the heterogeneity between the results of the studies. If there was no statistical heterogeneity among the results of each study (P>0.10, I\n2 ≤50%), a fixed-effects model was used for meta-analysis; otherwise, a random-effects model was used for meta-analysis. Publication bias was evaluated by the demonstration of funnel plots, and asymmetry was assessed by means of the Egger regression test. P<0.05 indicated that the difference between groups was statistically significant.", "A total of 238 literatures were initially detected in the first literature search. After reading the titles and abstracts, 58 papers were included after excluding the literatures that did not meet the inclusion criteria. After reading the full text of the literature, 14 RCTs15–28 were finally included (Fig. 1). Of the included 14 RCTs, a total of 1700 patients with cervical cancer were involved, 852 patients underwent LRH treatment, 848 patients underwent ARH treatment. The basic characteristics of the included studies are shown in Table 1.\nPreferred reporting items for systematic reviews and meta-analyses flow diagram of randomized controlled trialselection.\nThe Characteristics of Included Randomized Controlled Trials\n①duration of surgery; ②intraoperative blood loss; ③number of lymph nodes removed; ④intraoperative complications; ⑤postoperative complications; ⑥the time to first passage of flatus; ⑦relapse rate; ⑧survival rate.\nARH indicates abdominal radical hysterectomy; LRH, laparoscopic radical hysterectomy; NA, not available.", "Of all the 14 included RCTs, 11 RCTs15–22,24–26 described specific methods for generating random sequences, and another 3 RCTs did not mention specific randomization methods. Only 6 RCTs reported specific allocation concealment methods. Blinding of participants and personnel and outcome assessment (detection bias) were not stated in the included RCTs. No loss to follow-up and selective outcomes were reported. The results of the literature quality evaluation are shown in Figures 2 and 3.\nRisk of bias graph.\nRisk of bias summary.", "[SUBTITLE] Duration of Surgery [SUBSECTION] Eleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I\n2 = 99%).\nForest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy.\nEleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I\n2 = 99%).\nForest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy.\n[SUBTITLE] Intraoperative Blood Loss [SUBSECTION] Ten included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I\n2 = 73%).\nTen included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I\n2 = 73%).\n[SUBTITLE] Number of Lymph Nodes Removed [SUBSECTION] Eight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I\n2 = 97%).\nEight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I\n2 = 97%).\n[SUBTITLE] The Time to First Passage of Flatus [SUBSECTION] Seven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I\n2 = 70%).\nSeven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I\n2 = 70%).\n[SUBTITLE] Intraoperative Complications [SUBSECTION] Three included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I\n2 =53%).\nForest plots for synthesized outcomes.\nThree included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I\n2 =53%).\nForest plots for synthesized outcomes.\n[SUBTITLE] Postoperative Complications [SUBSECTION] Three included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I\n2 =37%).\nThree included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I\n2 =37%).\n[SUBTITLE] Relapse Rate [SUBSECTION] Five included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I\n2=54%).\nFive included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I\n2=54%).\n[SUBTITLE] Survival Rate [SUBSECTION] Six included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I\n2 =48%).\nSix included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I\n2 =48%).", "Eleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I\n2 = 99%).\nForest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy.", "Ten included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I\n2 = 73%).", "Eight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I\n2 = 97%).", "Seven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I\n2 = 70%).", "Three included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I\n2 =53%).\nForest plots for synthesized outcomes.", "Three included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I\n2 =37%).", "Five included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I\n2=54%).", "Six included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I\n2 =48%).", "As showed in Figures 6 and 7, the dots in the funnel plots for synthesized outcomes were evenly distributed, and the results of Egger regression tests indicated that there was no publication bias in the synthesized outcomes (all P>0.05).\nFunnel plots for synthesized outcomes. MD indicates mean difference; OD, odd ratio.\nFunnel plots for synthesized outcomes. OD indicates odd ratio.", "Sensitivity analyses that assess the impact of single 1 study on the overall risk estimate by removing 1 study in each turn. No significant change of the overall risk estimates by removing any single study was found.", "The treatment of cervical cancer should be based on the patient’s age, general condition, tumor stage, histologic type, lymph node metastasis, and fertility requirements to formulate the best treatment plan.29,30 For the diagnosis and treatment of cervical cancer, not only we must follow the basic principles of diagnosis and treatment guidelines, but also pay attention to individualized treatment, so the choice of specific treatment plan often requires the comprehensive judgment of clinicians.31 The results of clinical studies32–34 so far have shown that for patients with early stage of cervical cancer FIGO clinical stage compared with radiotherapy alone, surgical treatment has the advantages of reducing the mortality rate of cervical cancer and the probability of tumor recurrence, and improving the quality of life of patients after surgery. At the same time, the possibility of permanent damage to ovarian function and other normal organs around the tumor by radiation therapy should be considered.35 Therefore, for patients with early-stage cervical cancer, surgery is still the first choice for treatment. This meta-analysis has included 14 RCTs comparing ARH and LRH in the treatment of cervical cancer. The results have showed that compared with the ARH, the LRH in the treatment of early cervical cancer can reduce the intraoperative blood loss, significantly increase the number of lymph nodes removed, and shorten the time to first passage of flatus. There are no significant differences between the ARH and LRH in terms of duration of surgery, intraoperative complications, postoperative complications, postoperative relapse rate, and survival rate.\nWe have not found the difference in the duration of surgery between ARH and LRH. On the one hand, the laparoscopic surgery is a new development technology, and the publication time of the included studies in this meta-analysis is different. In earlier studies, the laparoscopic surgery was carried out for a short period of time. ARH is a traditional operation, the surgeon may have higher technical level and richer surgical experience.36 On the other hand, the “learning curve” of the surgeon also affects the duration of surgery.37–39 As none of the included studies reported the surgeon’s proficiency in LRH and the number of previous surgeries, the learning curve of the surgeons also contributed to the statistical heterogeneity among the included studies. Finally, none of the included studies have reported whether the patients had a history of multiple operations in the lower abdomen, which may cause adhesions between the pelvic and abdominal organs and tissues, leading to longer duration of surgery.\nThe intraoperative blood loss of LRH is less than that of ARH, and the difference was statistically significant. LRH has a small incision, whereas ARH has a larger incision. The process of entering the abdominal cavity through the skin layer by layer damages more blood vessels and increases the bleeding of the incision.40 When laparoscopic ultrasonic scalpel is used for tissue separation during surgery, it can not only cut tissue but also stop bleeding in a timely and effective manner, reducing unnecessary bleeding.41,42 Because of the magnifying effect of laparoscopic equipment, it is easier and clearer for the operator to identify important tissue structures such as blood vessels and nerves and the local anatomical relationship between them during surgical operations.43–45 When the laparoscopic eyepiece is close to the observation target, the surgical details can be observed for better and finer operation, and the bleeding-prone tissue can be effectively and preventively avoided. When the laparoscopic eyepiece is retreated, the surgical field of view can be expanded, and the operator’s awareness of the operation can be improved. The overall grasp of the visual field avoids the omission of small active bleeding foci in the surgical field of view, and can effectively stop bleeding in time, thereby significantly reducing the amount of bleeding on the surgical wound and avoiding the occurrence of major bleeding.46–48\n\nThe results of this study have showed that the time of the time to the first passage of flatus in the LRH is earlier than that of ARH. The possible reason for the difference may be that laparoscopic surgery avoids the touch of the abdominal retractor, gauze and operator’s gloves on the intestine, and the less intestinal tract manipulations in the LRH reduce irritation to the patient’s gastrointestinal tract. In addition, laparoscopic surgery is performed in a relatively closed abdominal cavity, and the pelvic and abdominal organs do not need to be exposed for a long time, and the intestinal tract will not be dry and cold, so that the patient’s gastrointestinal motility function can be quickly recovered after surgery.49–51\n\nThere are no significant differences in recurrence and survival between LRH and ARH. A long-term follow-up study52 has found that the 5-year survival rate after LRH was basically the same as that of laparotomy. In a retrospective case-control study,53 2 groups of cervical cancer patients with different surgical procedures were followed up for 5 years. The results showed that compared with laparoscopic surgery, the 5-year overall survival rates were 95.2% and 96.4%, respectively, the 5-year recurrence-free survival rates were 92.8% and 94.4%, and the 5-year tumor-specific survival rates were 89.6% and 90.7%, respectively. In recent years, some researchers54–56 have compared the 5-year overall survival rate and 5-year disease-free survival rate of laparoscopic surgery and traditional laparotomy in the treatment of cervical cancer, and have found that there is no significant difference between the 2 groups. We have evaluated the data from English versus Chinese reports, there is a difference in the survival rate between the English versus Chinese reports. Heretofore, the predominance of English data suggests an adverse survival rate associated with a minimally invasive approach. For patients with cervical cancer, whether different surgical methods will improve their postoperative survival rate and which method is more advantageous, there is still no clear conclusion. Therefore, long-term follow-up results of multiple large-sample RCTs are still needed to confirm and evaluate the effect of LRH and ARH.\nThere are certain limitations in this study that must be considered. First, because surgery cannot be blinded, this may increase the measurement and performance bias of the study and increase the clinical heterogeneity among included RCTs. Second, some of the measurement indicators in this study were subject to the surgeon’s learning curve and clinical experience, and none of the included studies reported whether the surgeons were proficient in LRH and the number of previous surgeries. Third, we included patients not only limited to early stage, we tried to conduct subgroup analysis based on the disease stage, yet the data were limited for subgroup analysis. In addition, most of the included studies had small sample sizes, which may affect the internal validity of the results. It is recommended that randomization methods, allocation concealment, and blinding design should be reported in detail in future clinical trials to reduce selection bias and measurement bias." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "BACKGROUND", "METHODS", "Search Strategy", "Inclusion and Exclusion Criteria", "Quality Assessment", "Literature Screening and Data Extraction", "Statistical Analysis", "RESULTS", "RCT Selection", "Literature Quality Evaluation", "Meta-analysis", "Duration of Surgery", "Intraoperative Blood Loss", "Number of Lymph Nodes Removed", "The Time to First Passage of Flatus", "Intraoperative Complications", "Postoperative Complications", "Relapse Rate", "Survival Rate", "Publication Bias", "Sensitivity Analyses", "DISCUSSIONS", "CONCLUSIONS", "Supplementary Material" ]
[ "Cervical cancer is one of the most common malignant tumors that seriously threaten women’s health, and its incidence ranks third among female malignant tumors in the world, second only to breast cancer and colorectal cancer.1,2 There are about 500,000 new cases of cervical cancer every year in the world, 85% of which are in developing countries, and about 250,000 people die of cervical cancer each year.3,4 In China, 140,000 new cases are discovered every year, accounting for about one third of the global patients with cervical cancer, and about 50,000 patients die of cervical cancer every year.5,6 In recent years, the incidence of cervical cancer has a younger trend.7,8 Therefore, the treatment and care of cervical cancer is of great significance to the prognosis of patients.\nRadical hysterectomy and lymph node dissection are the surgical methods recommended by the International Federation of Obstetrics and Gynaecology (FIGO) for the treatment of early-stage cervical cancer.9 Since the first successful laparoscopic pelvic lymph node dissection for cervical cancer in 1989, minimally invasive techniques have developed rapidly. In 1991, laparoscopy was first used for pelvic lymph node dissection combined with vaginal hysterectomy in the treatment of early cervical cancer. Laparoscopy has been used for total hysterectomy and pelvic lymph node dissection in the treatment of cervical cancer. Since then, many studies10–12 have shown that laparoscopic surgery has the advantages of less trauma, faster recovery, and less pain for patients after surgery, which has been valued by the majority of obstetrics and gynecologists. In recent years, many researchers have conducted many studies on the clinical efficacy and safety of laparoscopic surgery for cervical cancer. However, because of the small number of independent research samples and uneven research quality limits the results promotion. So far, no large-sample, multicenter clinical trials have been reported. To this end, we aimed to conduct a meta-analysis to compare the efficacy and safety of laparoscopic radical hysterectomy (LRH) and open abdominal radical hysterectomy (ARH) in the treatment of cervical cancer to provide a reliable basis for the selection of clinical surgical methods for cervical cancer treatment.", "This meta-analysis and systematic review was conducted in comply with the statement of preferred reporting items for systematic reviews and meta-analyses13 (Supplemental Digital Content 1, http://links.lww.com/AJCO/A427 and Supplemental Digital Content 2, http://links.lww.com/AJCO/A428).\n[SUBTITLE] Search Strategy [SUBSECTION] The 2 authors independently searched PubMed, Web of Science, EMbase, Cochrane Library, China Biomedical Database, Chinese National Knowledge Infrastructure (CNKI), Wanfang, and Weipu databases for randomized controlled trials (RCTs) on the efficacy and safety of LRH and ARH for cervical cancer treatment. The retrieval time limit is from the establishment of the database to May 31, 2022. The language was limited to Chinese or English. The search terms used were as following: (“cervical cancer” OR “cervical carcinoma” OR “uterine cervical neoplasms” OR “invasive carcinoma of cervix uteri”) AND (“laparoscopy” OR “laparotomy” OR “surgery” OR “abdominal” OR “open”).\nThe 2 authors independently searched PubMed, Web of Science, EMbase, Cochrane Library, China Biomedical Database, Chinese National Knowledge Infrastructure (CNKI), Wanfang, and Weipu databases for randomized controlled trials (RCTs) on the efficacy and safety of LRH and ARH for cervical cancer treatment. The retrieval time limit is from the establishment of the database to May 31, 2022. The language was limited to Chinese or English. The search terms used were as following: (“cervical cancer” OR “cervical carcinoma” OR “uterine cervical neoplasms” OR “invasive carcinoma of cervix uteri”) AND (“laparoscopy” OR “laparotomy” OR “surgery” OR “abdominal” OR “open”).\n[SUBTITLE] Inclusion and Exclusion Criteria [SUBSECTION] The inclusion criteria for this meta-analysis were as follows: (1) the type of study was a RCT, the language was limited to Chinese and English, (2) the patients were clinically diagnosed with cervical cancer confirmed by pathology and underwent radical hysterectomy (3) LRH was used for the intervention measures in the experimental group, and ARH was used for the intervention measures in the control group, (4) The article reported relevant outcome indicators, such as operation time, intraoperative blood loss, number of lymph node biopsies, recurrence rate, and survival rate. The exclusion criteria for this meta-analysis were as follows: (1) review, case, and basic experimental studies; (2) duplicate published literature reports; (3) research reports for which data could not be extracted.\nThe inclusion criteria for this meta-analysis were as follows: (1) the type of study was a RCT, the language was limited to Chinese and English, (2) the patients were clinically diagnosed with cervical cancer confirmed by pathology and underwent radical hysterectomy (3) LRH was used for the intervention measures in the experimental group, and ARH was used for the intervention measures in the control group, (4) The article reported relevant outcome indicators, such as operation time, intraoperative blood loss, number of lymph node biopsies, recurrence rate, and survival rate. The exclusion criteria for this meta-analysis were as follows: (1) review, case, and basic experimental studies; (2) duplicate published literature reports; (3) research reports for which data could not be extracted.\n[SUBTITLE] Quality Assessment [SUBSECTION] The RCTs included in this study were assessed using the risk of bias assessment tool for RCTs recommended by Cochrane library.14 The evaluated items in this tool include (1) the method of generating random sequences; (2) whether the allocation scheme is hidden; (3)whether the personnel is blinded; (4) whether the outcome assessment is blinded; (5) whether the outcome data is complete; (6) whether the results are selectively reported; and (7) whether there is any other bias. For each study result, a choice was made on the above 7 items, including “Yes” (low bias), “No” (high bias), and “Unclear” (lack of relevant information or uncertain bias).\nThe RCTs included in this study were assessed using the risk of bias assessment tool for RCTs recommended by Cochrane library.14 The evaluated items in this tool include (1) the method of generating random sequences; (2) whether the allocation scheme is hidden; (3)whether the personnel is blinded; (4) whether the outcome assessment is blinded; (5) whether the outcome data is complete; (6) whether the results are selectively reported; and (7) whether there is any other bias. For each study result, a choice was made on the above 7 items, including “Yes” (low bias), “No” (high bias), and “Unclear” (lack of relevant information or uncertain bias).\n[SUBTITLE] Literature Screening and Data Extraction [SUBSECTION] In this meta-analysis, 2 reviewers independently screened the literature and extracted data and cross-checked them. In case of disagreement, a third party was consulted to assist in judgment, and the first original authors would be contacted as much as possible to supplement the lack of data. We extracted the following data: basic information of included studies, including research title, first author, publication journal and time, etc.; basic characteristics of research subjects, including age, number of cases, FIGO stage; Key elements of study design type, and quality assessment; relevant outcome data.\nIn this meta-analysis, 2 reviewers independently screened the literature and extracted data and cross-checked them. In case of disagreement, a third party was consulted to assist in judgment, and the first original authors would be contacted as much as possible to supplement the lack of data. We extracted the following data: basic information of included studies, including research title, first author, publication journal and time, etc.; basic characteristics of research subjects, including age, number of cases, FIGO stage; Key elements of study design type, and quality assessment; relevant outcome data.\n[SUBTITLE] Statistical Analysis [SUBSECTION] This study used RevMan 5.3 software for meta-analysis. For enumeration data, odd ratio (OR) and its 95% confidence interval (CI) were used as effect size. For measurement data, mean difference (MD) and its 95% CI were used as effect size. The χ2 test was used to analyze the heterogeneity between the results of the studies. If there was no statistical heterogeneity among the results of each study (P>0.10, I\n2 ≤50%), a fixed-effects model was used for meta-analysis; otherwise, a random-effects model was used for meta-analysis. Publication bias was evaluated by the demonstration of funnel plots, and asymmetry was assessed by means of the Egger regression test. P<0.05 indicated that the difference between groups was statistically significant.\nThis study used RevMan 5.3 software for meta-analysis. For enumeration data, odd ratio (OR) and its 95% confidence interval (CI) were used as effect size. For measurement data, mean difference (MD) and its 95% CI were used as effect size. The χ2 test was used to analyze the heterogeneity between the results of the studies. If there was no statistical heterogeneity among the results of each study (P>0.10, I\n2 ≤50%), a fixed-effects model was used for meta-analysis; otherwise, a random-effects model was used for meta-analysis. Publication bias was evaluated by the demonstration of funnel plots, and asymmetry was assessed by means of the Egger regression test. P<0.05 indicated that the difference between groups was statistically significant.", "The 2 authors independently searched PubMed, Web of Science, EMbase, Cochrane Library, China Biomedical Database, Chinese National Knowledge Infrastructure (CNKI), Wanfang, and Weipu databases for randomized controlled trials (RCTs) on the efficacy and safety of LRH and ARH for cervical cancer treatment. The retrieval time limit is from the establishment of the database to May 31, 2022. The language was limited to Chinese or English. The search terms used were as following: (“cervical cancer” OR “cervical carcinoma” OR “uterine cervical neoplasms” OR “invasive carcinoma of cervix uteri”) AND (“laparoscopy” OR “laparotomy” OR “surgery” OR “abdominal” OR “open”).", "The inclusion criteria for this meta-analysis were as follows: (1) the type of study was a RCT, the language was limited to Chinese and English, (2) the patients were clinically diagnosed with cervical cancer confirmed by pathology and underwent radical hysterectomy (3) LRH was used for the intervention measures in the experimental group, and ARH was used for the intervention measures in the control group, (4) The article reported relevant outcome indicators, such as operation time, intraoperative blood loss, number of lymph node biopsies, recurrence rate, and survival rate. The exclusion criteria for this meta-analysis were as follows: (1) review, case, and basic experimental studies; (2) duplicate published literature reports; (3) research reports for which data could not be extracted.", "The RCTs included in this study were assessed using the risk of bias assessment tool for RCTs recommended by Cochrane library.14 The evaluated items in this tool include (1) the method of generating random sequences; (2) whether the allocation scheme is hidden; (3)whether the personnel is blinded; (4) whether the outcome assessment is blinded; (5) whether the outcome data is complete; (6) whether the results are selectively reported; and (7) whether there is any other bias. For each study result, a choice was made on the above 7 items, including “Yes” (low bias), “No” (high bias), and “Unclear” (lack of relevant information or uncertain bias).", "In this meta-analysis, 2 reviewers independently screened the literature and extracted data and cross-checked them. In case of disagreement, a third party was consulted to assist in judgment, and the first original authors would be contacted as much as possible to supplement the lack of data. We extracted the following data: basic information of included studies, including research title, first author, publication journal and time, etc.; basic characteristics of research subjects, including age, number of cases, FIGO stage; Key elements of study design type, and quality assessment; relevant outcome data.", "This study used RevMan 5.3 software for meta-analysis. For enumeration data, odd ratio (OR) and its 95% confidence interval (CI) were used as effect size. For measurement data, mean difference (MD) and its 95% CI were used as effect size. The χ2 test was used to analyze the heterogeneity between the results of the studies. If there was no statistical heterogeneity among the results of each study (P>0.10, I\n2 ≤50%), a fixed-effects model was used for meta-analysis; otherwise, a random-effects model was used for meta-analysis. Publication bias was evaluated by the demonstration of funnel plots, and asymmetry was assessed by means of the Egger regression test. P<0.05 indicated that the difference between groups was statistically significant.", "[SUBTITLE] RCT Selection [SUBSECTION] A total of 238 literatures were initially detected in the first literature search. After reading the titles and abstracts, 58 papers were included after excluding the literatures that did not meet the inclusion criteria. After reading the full text of the literature, 14 RCTs15–28 were finally included (Fig. 1). Of the included 14 RCTs, a total of 1700 patients with cervical cancer were involved, 852 patients underwent LRH treatment, 848 patients underwent ARH treatment. The basic characteristics of the included studies are shown in Table 1.\nPreferred reporting items for systematic reviews and meta-analyses flow diagram of randomized controlled trialselection.\nThe Characteristics of Included Randomized Controlled Trials\n①duration of surgery; ②intraoperative blood loss; ③number of lymph nodes removed; ④intraoperative complications; ⑤postoperative complications; ⑥the time to first passage of flatus; ⑦relapse rate; ⑧survival rate.\nARH indicates abdominal radical hysterectomy; LRH, laparoscopic radical hysterectomy; NA, not available.\nA total of 238 literatures were initially detected in the first literature search. After reading the titles and abstracts, 58 papers were included after excluding the literatures that did not meet the inclusion criteria. After reading the full text of the literature, 14 RCTs15–28 were finally included (Fig. 1). Of the included 14 RCTs, a total of 1700 patients with cervical cancer were involved, 852 patients underwent LRH treatment, 848 patients underwent ARH treatment. The basic characteristics of the included studies are shown in Table 1.\nPreferred reporting items for systematic reviews and meta-analyses flow diagram of randomized controlled trialselection.\nThe Characteristics of Included Randomized Controlled Trials\n①duration of surgery; ②intraoperative blood loss; ③number of lymph nodes removed; ④intraoperative complications; ⑤postoperative complications; ⑥the time to first passage of flatus; ⑦relapse rate; ⑧survival rate.\nARH indicates abdominal radical hysterectomy; LRH, laparoscopic radical hysterectomy; NA, not available.\n[SUBTITLE] Literature Quality Evaluation [SUBSECTION] Of all the 14 included RCTs, 11 RCTs15–22,24–26 described specific methods for generating random sequences, and another 3 RCTs did not mention specific randomization methods. Only 6 RCTs reported specific allocation concealment methods. Blinding of participants and personnel and outcome assessment (detection bias) were not stated in the included RCTs. No loss to follow-up and selective outcomes were reported. The results of the literature quality evaluation are shown in Figures 2 and 3.\nRisk of bias graph.\nRisk of bias summary.\nOf all the 14 included RCTs, 11 RCTs15–22,24–26 described specific methods for generating random sequences, and another 3 RCTs did not mention specific randomization methods. Only 6 RCTs reported specific allocation concealment methods. Blinding of participants and personnel and outcome assessment (detection bias) were not stated in the included RCTs. No loss to follow-up and selective outcomes were reported. The results of the literature quality evaluation are shown in Figures 2 and 3.\nRisk of bias graph.\nRisk of bias summary.\n[SUBTITLE] Meta-analysis [SUBSECTION] [SUBTITLE] Duration of Surgery [SUBSECTION] Eleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I\n2 = 99%).\nForest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy.\nEleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I\n2 = 99%).\nForest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy.\n[SUBTITLE] Intraoperative Blood Loss [SUBSECTION] Ten included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I\n2 = 73%).\nTen included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I\n2 = 73%).\n[SUBTITLE] Number of Lymph Nodes Removed [SUBSECTION] Eight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I\n2 = 97%).\nEight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I\n2 = 97%).\n[SUBTITLE] The Time to First Passage of Flatus [SUBSECTION] Seven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I\n2 = 70%).\nSeven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I\n2 = 70%).\n[SUBTITLE] Intraoperative Complications [SUBSECTION] Three included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I\n2 =53%).\nForest plots for synthesized outcomes.\nThree included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I\n2 =53%).\nForest plots for synthesized outcomes.\n[SUBTITLE] Postoperative Complications [SUBSECTION] Three included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I\n2 =37%).\nThree included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I\n2 =37%).\n[SUBTITLE] Relapse Rate [SUBSECTION] Five included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I\n2=54%).\nFive included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I\n2=54%).\n[SUBTITLE] Survival Rate [SUBSECTION] Six included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I\n2 =48%).\nSix included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I\n2 =48%).\n[SUBTITLE] Duration of Surgery [SUBSECTION] Eleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I\n2 = 99%).\nForest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy.\nEleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I\n2 = 99%).\nForest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy.\n[SUBTITLE] Intraoperative Blood Loss [SUBSECTION] Ten included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I\n2 = 73%).\nTen included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I\n2 = 73%).\n[SUBTITLE] Number of Lymph Nodes Removed [SUBSECTION] Eight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I\n2 = 97%).\nEight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I\n2 = 97%).\n[SUBTITLE] The Time to First Passage of Flatus [SUBSECTION] Seven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I\n2 = 70%).\nSeven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I\n2 = 70%).\n[SUBTITLE] Intraoperative Complications [SUBSECTION] Three included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I\n2 =53%).\nForest plots for synthesized outcomes.\nThree included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I\n2 =53%).\nForest plots for synthesized outcomes.\n[SUBTITLE] Postoperative Complications [SUBSECTION] Three included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I\n2 =37%).\nThree included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I\n2 =37%).\n[SUBTITLE] Relapse Rate [SUBSECTION] Five included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I\n2=54%).\nFive included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I\n2=54%).\n[SUBTITLE] Survival Rate [SUBSECTION] Six included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I\n2 =48%).\nSix included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I\n2 =48%).\n[SUBTITLE] Publication Bias [SUBSECTION] As showed in Figures 6 and 7, the dots in the funnel plots for synthesized outcomes were evenly distributed, and the results of Egger regression tests indicated that there was no publication bias in the synthesized outcomes (all P>0.05).\nFunnel plots for synthesized outcomes. MD indicates mean difference; OD, odd ratio.\nFunnel plots for synthesized outcomes. OD indicates odd ratio.\nAs showed in Figures 6 and 7, the dots in the funnel plots for synthesized outcomes were evenly distributed, and the results of Egger regression tests indicated that there was no publication bias in the synthesized outcomes (all P>0.05).\nFunnel plots for synthesized outcomes. MD indicates mean difference; OD, odd ratio.\nFunnel plots for synthesized outcomes. OD indicates odd ratio.\n[SUBTITLE] Sensitivity Analyses [SUBSECTION] Sensitivity analyses that assess the impact of single 1 study on the overall risk estimate by removing 1 study in each turn. No significant change of the overall risk estimates by removing any single study was found.\nSensitivity analyses that assess the impact of single 1 study on the overall risk estimate by removing 1 study in each turn. No significant change of the overall risk estimates by removing any single study was found.", "A total of 238 literatures were initially detected in the first literature search. After reading the titles and abstracts, 58 papers were included after excluding the literatures that did not meet the inclusion criteria. After reading the full text of the literature, 14 RCTs15–28 were finally included (Fig. 1). Of the included 14 RCTs, a total of 1700 patients with cervical cancer were involved, 852 patients underwent LRH treatment, 848 patients underwent ARH treatment. The basic characteristics of the included studies are shown in Table 1.\nPreferred reporting items for systematic reviews and meta-analyses flow diagram of randomized controlled trialselection.\nThe Characteristics of Included Randomized Controlled Trials\n①duration of surgery; ②intraoperative blood loss; ③number of lymph nodes removed; ④intraoperative complications; ⑤postoperative complications; ⑥the time to first passage of flatus; ⑦relapse rate; ⑧survival rate.\nARH indicates abdominal radical hysterectomy; LRH, laparoscopic radical hysterectomy; NA, not available.", "Of all the 14 included RCTs, 11 RCTs15–22,24–26 described specific methods for generating random sequences, and another 3 RCTs did not mention specific randomization methods. Only 6 RCTs reported specific allocation concealment methods. Blinding of participants and personnel and outcome assessment (detection bias) were not stated in the included RCTs. No loss to follow-up and selective outcomes were reported. The results of the literature quality evaluation are shown in Figures 2 and 3.\nRisk of bias graph.\nRisk of bias summary.", "[SUBTITLE] Duration of Surgery [SUBSECTION] Eleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I\n2 = 99%).\nForest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy.\nEleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I\n2 = 99%).\nForest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy.\n[SUBTITLE] Intraoperative Blood Loss [SUBSECTION] Ten included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I\n2 = 73%).\nTen included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I\n2 = 73%).\n[SUBTITLE] Number of Lymph Nodes Removed [SUBSECTION] Eight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I\n2 = 97%).\nEight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I\n2 = 97%).\n[SUBTITLE] The Time to First Passage of Flatus [SUBSECTION] Seven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I\n2 = 70%).\nSeven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I\n2 = 70%).\n[SUBTITLE] Intraoperative Complications [SUBSECTION] Three included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I\n2 =53%).\nForest plots for synthesized outcomes.\nThree included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I\n2 =53%).\nForest plots for synthesized outcomes.\n[SUBTITLE] Postoperative Complications [SUBSECTION] Three included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I\n2 =37%).\nThree included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I\n2 =37%).\n[SUBTITLE] Relapse Rate [SUBSECTION] Five included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I\n2=54%).\nFive included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I\n2=54%).\n[SUBTITLE] Survival Rate [SUBSECTION] Six included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I\n2 =48%).\nSix included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I\n2 =48%).", "Eleven included RCTs16,18–23,25–28 reported the duration of surgery. Meta-analysis of random effects model showed that there was no significant difference in the duration of surgery between LRH group and ARH group (MD=27.62; 95% CI, −6.26, 61.49; P=0.11, Fig. 4A), with evidence of heterogeneity (P<0.001, I\n2 = 99%).\nForest plots for synthesized outcomes. ARH indicates abdominal radical hysterectomy; CI, confidence interval; LRH, laparoscopic radical hysterectomy.", "Ten included RCTs16,19–23,25–28 reported the intraoperative blood loss. Meta-analysis of random effects model showed that the intraoperative blood loss in the LRH group was significantly less than that of ARH group (MD=−58.08; 95% CI, −70.91, −45.24; P<0.001, Fig. 4B), with evidence of heterogeneity (P<0.001, I\n2 = 73%).", "Eight included RCTs16,18,19,21–23,25,26 reported the number of lymph nodes removed. Meta-analysis of random effects model showed that the number of lymph nodes removed in the LRH group was significantly more than that of ARH group (MD=3.47; 95% CI, 0.51, 6.43; P=0.02, Fig. 4C), with evidence of heterogeneity (P<0.001, I\n2 = 97%).", "Seven included RCTs16,19,20,22,25–27 reported the time to first passage of flatus. Meta-analysis of random effects model showed that the time to first passage of flatus in the LRH group was significantly less than that of ARH group (MD=−14.50; 95% CI, −16.55, −12.44; P<0.001, Fig. 4D), with evidence of heterogeneity (P =0.003, I\n2 = 70%).", "Three included RCTs18,21,22 reported the intraoperative complications. Meta-analysis of random effects model showed that there was no significant difference in intraoperative complications between LRH group and ARH group (OR=1.10; 95% CI, 0.17, 7.32; P=0.92, Fig. 5A), with evidence of heterogeneity (P =0.12, I\n2 =53%).\nForest plots for synthesized outcomes.", "Three included RCTs18,20,21 reported the postoperative complications. Meta-analysis of fixed effects model showed that there was no significant difference in postoperative complications between LRH group and ARH group (OR=0.78; 95% CI, 0.33, 1.86; P=0.57, Fig. 5B], with no evidence of heterogeneity (P =0.21, I\n2 =37%).", "Five included RCTs15,21,24,27,28 reported the relapse rate. The meta-analysis of random effects model showed that there was no significant difference in the relapse rate between LRH group and ARH group (OR=1.45; 95% CI, 0.56, 3.74; P=0.44, Fig. 5C), with evidence of heterogeneity (P=0.07, I\n2=54%).", "Six included RCTs15,19,20,22,24,27 reported the survival rate. The meta-analysis of fixed effects model showed that there was no significant difference in the survival rate between LRH group and ARH group (OR=0.75; 95% CI, 0.52, 1.08; P=0.12, Fig. 5D], with evidence of heterogeneity (P =0.08, I\n2 =48%).", "As showed in Figures 6 and 7, the dots in the funnel plots for synthesized outcomes were evenly distributed, and the results of Egger regression tests indicated that there was no publication bias in the synthesized outcomes (all P>0.05).\nFunnel plots for synthesized outcomes. MD indicates mean difference; OD, odd ratio.\nFunnel plots for synthesized outcomes. OD indicates odd ratio.", "Sensitivity analyses that assess the impact of single 1 study on the overall risk estimate by removing 1 study in each turn. No significant change of the overall risk estimates by removing any single study was found.", "The treatment of cervical cancer should be based on the patient’s age, general condition, tumor stage, histologic type, lymph node metastasis, and fertility requirements to formulate the best treatment plan.29,30 For the diagnosis and treatment of cervical cancer, not only we must follow the basic principles of diagnosis and treatment guidelines, but also pay attention to individualized treatment, so the choice of specific treatment plan often requires the comprehensive judgment of clinicians.31 The results of clinical studies32–34 so far have shown that for patients with early stage of cervical cancer FIGO clinical stage compared with radiotherapy alone, surgical treatment has the advantages of reducing the mortality rate of cervical cancer and the probability of tumor recurrence, and improving the quality of life of patients after surgery. At the same time, the possibility of permanent damage to ovarian function and other normal organs around the tumor by radiation therapy should be considered.35 Therefore, for patients with early-stage cervical cancer, surgery is still the first choice for treatment. This meta-analysis has included 14 RCTs comparing ARH and LRH in the treatment of cervical cancer. The results have showed that compared with the ARH, the LRH in the treatment of early cervical cancer can reduce the intraoperative blood loss, significantly increase the number of lymph nodes removed, and shorten the time to first passage of flatus. There are no significant differences between the ARH and LRH in terms of duration of surgery, intraoperative complications, postoperative complications, postoperative relapse rate, and survival rate.\nWe have not found the difference in the duration of surgery between ARH and LRH. On the one hand, the laparoscopic surgery is a new development technology, and the publication time of the included studies in this meta-analysis is different. In earlier studies, the laparoscopic surgery was carried out for a short period of time. ARH is a traditional operation, the surgeon may have higher technical level and richer surgical experience.36 On the other hand, the “learning curve” of the surgeon also affects the duration of surgery.37–39 As none of the included studies reported the surgeon’s proficiency in LRH and the number of previous surgeries, the learning curve of the surgeons also contributed to the statistical heterogeneity among the included studies. Finally, none of the included studies have reported whether the patients had a history of multiple operations in the lower abdomen, which may cause adhesions between the pelvic and abdominal organs and tissues, leading to longer duration of surgery.\nThe intraoperative blood loss of LRH is less than that of ARH, and the difference was statistically significant. LRH has a small incision, whereas ARH has a larger incision. The process of entering the abdominal cavity through the skin layer by layer damages more blood vessels and increases the bleeding of the incision.40 When laparoscopic ultrasonic scalpel is used for tissue separation during surgery, it can not only cut tissue but also stop bleeding in a timely and effective manner, reducing unnecessary bleeding.41,42 Because of the magnifying effect of laparoscopic equipment, it is easier and clearer for the operator to identify important tissue structures such as blood vessels and nerves and the local anatomical relationship between them during surgical operations.43–45 When the laparoscopic eyepiece is close to the observation target, the surgical details can be observed for better and finer operation, and the bleeding-prone tissue can be effectively and preventively avoided. When the laparoscopic eyepiece is retreated, the surgical field of view can be expanded, and the operator’s awareness of the operation can be improved. The overall grasp of the visual field avoids the omission of small active bleeding foci in the surgical field of view, and can effectively stop bleeding in time, thereby significantly reducing the amount of bleeding on the surgical wound and avoiding the occurrence of major bleeding.46–48\n\nThe results of this study have showed that the time of the time to the first passage of flatus in the LRH is earlier than that of ARH. The possible reason for the difference may be that laparoscopic surgery avoids the touch of the abdominal retractor, gauze and operator’s gloves on the intestine, and the less intestinal tract manipulations in the LRH reduce irritation to the patient’s gastrointestinal tract. In addition, laparoscopic surgery is performed in a relatively closed abdominal cavity, and the pelvic and abdominal organs do not need to be exposed for a long time, and the intestinal tract will not be dry and cold, so that the patient’s gastrointestinal motility function can be quickly recovered after surgery.49–51\n\nThere are no significant differences in recurrence and survival between LRH and ARH. A long-term follow-up study52 has found that the 5-year survival rate after LRH was basically the same as that of laparotomy. In a retrospective case-control study,53 2 groups of cervical cancer patients with different surgical procedures were followed up for 5 years. The results showed that compared with laparoscopic surgery, the 5-year overall survival rates were 95.2% and 96.4%, respectively, the 5-year recurrence-free survival rates were 92.8% and 94.4%, and the 5-year tumor-specific survival rates were 89.6% and 90.7%, respectively. In recent years, some researchers54–56 have compared the 5-year overall survival rate and 5-year disease-free survival rate of laparoscopic surgery and traditional laparotomy in the treatment of cervical cancer, and have found that there is no significant difference between the 2 groups. We have evaluated the data from English versus Chinese reports, there is a difference in the survival rate between the English versus Chinese reports. Heretofore, the predominance of English data suggests an adverse survival rate associated with a minimally invasive approach. For patients with cervical cancer, whether different surgical methods will improve their postoperative survival rate and which method is more advantageous, there is still no clear conclusion. Therefore, long-term follow-up results of multiple large-sample RCTs are still needed to confirm and evaluate the effect of LRH and ARH.\nThere are certain limitations in this study that must be considered. First, because surgery cannot be blinded, this may increase the measurement and performance bias of the study and increase the clinical heterogeneity among included RCTs. Second, some of the measurement indicators in this study were subject to the surgeon’s learning curve and clinical experience, and none of the included studies reported whether the surgeons were proficient in LRH and the number of previous surgeries. Third, we included patients not only limited to early stage, we tried to conduct subgroup analysis based on the disease stage, yet the data were limited for subgroup analysis. In addition, most of the included studies had small sample sizes, which may affect the internal validity of the results. It is recommended that randomization methods, allocation concealment, and blinding design should be reported in detail in future clinical trials to reduce selection bias and measurement bias.", "In conclusion, the results of this meta-analysis have found that LRH is beneficial to reduce the intraoperative blood loss, shorten the recovery time of postoperative gastrointestinal function, and increase the number of lymph nodes removed in patients with cervical cancer compared with ARH. Future research should be based on the patients’ safety and quality of life, focusing on postoperative overall survival time, tumor-free survival time, and quality of life of patients with cervical cancer. It is suggested that more RCTs should be carried out on the efficacy of LRH and ARH on postoperative quality of life and other outcomes with larger sample size and longer follow-up.", "" ]
[ null, "methods", null, null, null, null, null, "results", null, null, null, null, null, null, null, null, null, null, null, null, null, null, "conclusions", "supplementary-material" ]
[ "laparoscopic", "abdominal", "radical hysterectomy", "cervical cancer", "treatment", "care" ]
The evaluation of next-generation sequencing assisted pathogenic detection in immunocompromised hosts with pulmonary infection: A retrospective study.
36257289
Pulmonary infections are frequent in immunocompromised hosts (ICH), and microbial detection is difficult. As a new method, next-generation sequencing (NGS) may offer a solution.
INTRODUCTION
This study enrolled 356 patients with ICH complicated by pulmonary infection that were admitted to Zhongshan Hospital, Fudan University, from November 17, 2017, to November 23, 2018, including 102 and 254 in the NGS and non-NGS groups, respectively. Clinical characteristics, detection time, rough positive rate, effective positive rate, impact on anti-infective treatment plan, 30-day/60-day mortality, and in-hospital mortality were compared.
METHODS
NGS-assisted pathogenic detection reduced detection time (28.2 h [interquartile range (IQR) 25.9-29.83 h] vs. 50.50 h [IQR 47.90-90.91 h], P < 0.001), increased positive rate, rate of mixed infection detected, effective positive rate, and proportion of antibiotic treatment modification (45.28% vs. 89.22%, 4.72% vs. 51.96%, 21.65% vs. 64.71%, 16.54% vs. 46.08%, P < 0.001). The NGS group had a significantly lower 60-day mortality rate (18.63% vs. 33.07%, P = 0.007). The difference in the Kaplan-Meier survival curve was significant (P = 0.029). After multivariate logistic regression, NGS-assisted pathogenic detection remained a significant predictor of survival (OR 0.189, confidence interval [CI], 0.068-0.526).
RESULTS
NGS-assisted pathogenic detection may improve detection efficiency and is associated with better clinical outcomes in these patients.
CONCLUSION
[ "Humans", "Retrospective Studies", "High-Throughput Nucleotide Sequencing", "Immunocompromised Host" ]
9716706
null
null
null
null
RESULTS
[SUBTITLE] Study participants and baseline characteristics [SUBSECTION] Six‐hundred seventy‐eight patients met the enrollment criteria, and after, 322 patients were excluded (321 patients died before or within 48 h after microbial results and one HIV‐positive patient), and 356 patients were included in the final analysis (Figure 1). No significant differences in age, sex, APACHE II scores, proportion of patients with severe pneumonia, multidrug‐resistant bacterial infection, or invasive mechanical ventilation were detected between the two groups. However, the type of immunocompromise differed significantly between the two groups (P < 0.001). After further comparison using the Bonferroni method, we discovered the cause of this difference. The proportion of hematological neoplasms was lower in the non‐NGS group (16.93% vs. 36.27%, P < 0.05), and the proportion of chemotherapy or radiotherapy for solid tumors was higher (16.93% vs. 3.92%, P < 0.05) (Table 1). Patient selection. Abbreviations: NGS, next‐generation sequencing; HIV, human immunodeficiency virus Baseline characteristics Abbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; DM, diabetes mellitus; NGS, next‐generation sequencing; ns, the difference was not significant (by Bonferroni method); OR, odds ratio. Data are shown with median interquartile range (IQR). Renal failure or chronic kidney disease that requires glucocorticoid or immunosuppressant. Significant difference (Bonferroni's method). Six‐hundred seventy‐eight patients met the enrollment criteria, and after, 322 patients were excluded (321 patients died before or within 48 h after microbial results and one HIV‐positive patient), and 356 patients were included in the final analysis (Figure 1). No significant differences in age, sex, APACHE II scores, proportion of patients with severe pneumonia, multidrug‐resistant bacterial infection, or invasive mechanical ventilation were detected between the two groups. However, the type of immunocompromise differed significantly between the two groups (P < 0.001). After further comparison using the Bonferroni method, we discovered the cause of this difference. The proportion of hematological neoplasms was lower in the non‐NGS group (16.93% vs. 36.27%, P < 0.05), and the proportion of chemotherapy or radiotherapy for solid tumors was higher (16.93% vs. 3.92%, P < 0.05) (Table 1). Patient selection. Abbreviations: NGS, next‐generation sequencing; HIV, human immunodeficiency virus Baseline characteristics Abbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; DM, diabetes mellitus; NGS, next‐generation sequencing; ns, the difference was not significant (by Bonferroni method); OR, odds ratio. Data are shown with median interquartile range (IQR). Renal failure or chronic kidney disease that requires glucocorticoid or immunosuppressant. Significant difference (Bonferroni's method). [SUBTITLE] NGS performance [SUBSECTION] The detection time was shorter in the NGS group than in the non‐NGS group (28.2 h IQR 25.9–29.83 h vs. 50.50 h IQR 47.90–90.91 h, P < 0.001). The positive rate, effective positive rate, rate of mixed infection detected, and proportion of modified antibiotic treatment were significantly higher in the NGS group (89.22% vs. 45.28%, 51.96% vs. 4.72%, 64.71% vs. 21.65%, 46.08% vs. 17.71%, P < 0.001) (Data S2). In total, 123 samples from the NGS group were sent for NGS detection, including 79 blood samples, 28 BALF samples, 15 sputum samples, and 1 pleural effusion sample. The positive rate was not significantly different among the three sample types (blood, sputum, and BALF), and NGS had a higher positive rate than microbial culture in each sample type (86.08% vs. 11.54% in blood samples, 89.29% vs. 26.92% in BALF samples, 100% vs. 54.93% in sputum samples, P < 0.001). In total, 31 types of bacteria (only those with reported evidence of pathogenicity in the lungs), 9 types of fungi, 9 types of viruses, and 2 types of mycobacteria were identified in the NGS group. Pathogens that are frequently detected include Pneumocystis jeroveci (PJ) and cytomegalovirus (CMV). In the non‐NGS group, culture‐positive Candida albicans was the most common pathogen, which was clinically insignificant (important pathogens detected in the NGS group are listed in Figure 2, and the complete catalog is in Data S3). Important pathogens detected by NGS and traditional microbial method in the NGS group. Abbreviations: NTM, non‐tuberculous mycobacterial; MTB, Mycobacterium tuberculosis ; CMV, cytomegalovirus; NGS, next‐generation sequencing; PCR, polymerase chain reaction The detection time was shorter in the NGS group than in the non‐NGS group (28.2 h IQR 25.9–29.83 h vs. 50.50 h IQR 47.90–90.91 h, P < 0.001). The positive rate, effective positive rate, rate of mixed infection detected, and proportion of modified antibiotic treatment were significantly higher in the NGS group (89.22% vs. 45.28%, 51.96% vs. 4.72%, 64.71% vs. 21.65%, 46.08% vs. 17.71%, P < 0.001) (Data S2). In total, 123 samples from the NGS group were sent for NGS detection, including 79 blood samples, 28 BALF samples, 15 sputum samples, and 1 pleural effusion sample. The positive rate was not significantly different among the three sample types (blood, sputum, and BALF), and NGS had a higher positive rate than microbial culture in each sample type (86.08% vs. 11.54% in blood samples, 89.29% vs. 26.92% in BALF samples, 100% vs. 54.93% in sputum samples, P < 0.001). In total, 31 types of bacteria (only those with reported evidence of pathogenicity in the lungs), 9 types of fungi, 9 types of viruses, and 2 types of mycobacteria were identified in the NGS group. Pathogens that are frequently detected include Pneumocystis jeroveci (PJ) and cytomegalovirus (CMV). In the non‐NGS group, culture‐positive Candida albicans was the most common pathogen, which was clinically insignificant (important pathogens detected in the NGS group are listed in Figure 2, and the complete catalog is in Data S3). Important pathogens detected by NGS and traditional microbial method in the NGS group. Abbreviations: NTM, non‐tuberculous mycobacterial; MTB, Mycobacterium tuberculosis ; CMV, cytomegalovirus; NGS, next‐generation sequencing; PCR, polymerase chain reaction [SUBTITLE] Concordance between results of NGS and traditional methods [SUBSECTION] In the NGS group, results of the NGS and traditional methods were compared. NGS results were positive in 89.25% of cases, with 46.24% of cases being double‐positive (NGS positive and culture positive). The negative predictive value was 81.82% ± 40.45%. Subsequently, we compared NGS and culture results for these double‐positive cases. Match, partly matched, and mismatched cases accounted for 21.51%, 7.53%, and 9.67% of all cases, respectively. Mismatch cases were mostly caused by culture‐positive C. albicans, and after removing this interference, the proportion declined to 2.15% (Data S4 and S5). In the NGS group, results of the NGS and traditional methods were compared. NGS results were positive in 89.25% of cases, with 46.24% of cases being double‐positive (NGS positive and culture positive). The negative predictive value was 81.82% ± 40.45%. Subsequently, we compared NGS and culture results for these double‐positive cases. Match, partly matched, and mismatched cases accounted for 21.51%, 7.53%, and 9.67% of all cases, respectively. Mismatch cases were mostly caused by culture‐positive C. albicans, and after removing this interference, the proportion declined to 2.15% (Data S4 and S5). [SUBTITLE] Outcomes of the two groups [SUBSECTION] There was a significant difference in 30‐/60‐day and in‐hospital mortality between the NGS and traditional detection groups (16.67% vs. 28.35%, 18.63% vs. 33.07%, 18.63% vs. 33.07%, P < 0.05). This difference was also significant in Kaplan–Meier curve (Figure 3). Multivariate logistic regression analysis (Table 2) indicated that HAP/VAP (OR 6.41, P = 0.038), invasive ventilation (OR 17.271, P < 0.001), and APACHE II scores (OR 1.899, P < 0.001) were risk factors for 60‐day mortality, and NGS detection was a predictor of survival (OR 0.189, confidence interval [CI], 0.068–0.526). Subsequently, subgroup analysis was used to identify the appropriate population that benefitted from this detection method in terms of survival (Figure 4). Potential survival benefits included age >62 years (OR 0.342, P = 0.007), APACHE II score >10 (OR 0.358, P = 0.004), severe pneumonia (OR 0.047, P = 0.007), radiotherapy or chemotherapy for malignant tumors (OR 0.314, P = 0.047), and multidrug resistant infection (OR 0.052, P = 0.002). Survival curve of the NGS and non‐NGS groups Univariate and multivariate logistic regression analysis for 60‐day mortality Abbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; NGS, next‐generation sequencing; OR, odds ratio. Subgroup analysis for 60‐day mortality. Abbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; DM, diabetes mellitus; NGS, next‐generation sequencing; OR, odds ratio There was a significant difference in 30‐/60‐day and in‐hospital mortality between the NGS and traditional detection groups (16.67% vs. 28.35%, 18.63% vs. 33.07%, 18.63% vs. 33.07%, P < 0.05). This difference was also significant in Kaplan–Meier curve (Figure 3). Multivariate logistic regression analysis (Table 2) indicated that HAP/VAP (OR 6.41, P = 0.038), invasive ventilation (OR 17.271, P < 0.001), and APACHE II scores (OR 1.899, P < 0.001) were risk factors for 60‐day mortality, and NGS detection was a predictor of survival (OR 0.189, confidence interval [CI], 0.068–0.526). Subsequently, subgroup analysis was used to identify the appropriate population that benefitted from this detection method in terms of survival (Figure 4). Potential survival benefits included age >62 years (OR 0.342, P = 0.007), APACHE II score >10 (OR 0.358, P = 0.004), severe pneumonia (OR 0.047, P = 0.007), radiotherapy or chemotherapy for malignant tumors (OR 0.314, P = 0.047), and multidrug resistant infection (OR 0.052, P = 0.002). Survival curve of the NGS and non‐NGS groups Univariate and multivariate logistic regression analysis for 60‐day mortality Abbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; NGS, next‐generation sequencing; OR, odds ratio. Subgroup analysis for 60‐day mortality. Abbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; DM, diabetes mellitus; NGS, next‐generation sequencing; OR, odds ratio
null
null
[ "INTRODUCTION", "Patient population", "Study design and data collection", "Criteria for some follow‐up content", "Statistical analysis", "Study participants and baseline characteristics", "NGS performance", "Concordance between results of NGS and traditional methods", "Outcomes of the two groups", "ETHICS STATEMENT", "AUTHOR CONTRIBUTIONS" ]
[ "The number of hospitalized patients considered immunocompromised hosts (ICH) continues to increase because of the prevalent use of chemotherapy for cancer, organ transplantation, glucocorticoids, and other immunosuppressive therapies for various disease conditions. For these patients, pulmonary infection is one of the most frequent and fatal complications.\n1\n Respiratory infections and drug toxicity were major concerns in over two‐thirds of ICH cases, and mortality may reach 50–80% if accompanied by respiratory failure.\n2\n, \n3\n Traditional infectious and opportunistic pathogens account for a substantial proportion of infections in these patients. Danés et al.\n4\n discovered opportunistic fungal pneumonia in 38% of non‐human immunodeficiency virus (HIV) ICH patients with etiologically confirmed pneumonia. Therefore, accurate and rapid microbial detection can improve outcomes in these patients. However, the current standard microbial culture has two major drawbacks: low sensitivity and the amount of time it takes to return results.\n5\n\n\nMolecular diagnostic methods, such as polymerase chain reaction (PCR) and next‐generation sequencing (NGS), may provide a solution to this problem. Pathogen sequences can be sorted using deep sequencing and data mining without using a specific primer.\n6\n Several studies have demonstrated the medical benefits of NGS. Guo et al.\n7\n and Ai et al.\n8\n used NGS to monitor disease progression and therapeutic effectiveness. Long et al.\n9\n utilized NGS to detect bacteria in blood samples from patients with sepsis in the intensive care unit (ICU) and discovered greater sensitivity than culture. Pan et al.\n10\n used a case series to reveal the effectiveness of NGS in detecting pathogens in bronchoalveolar lavage fluid (BALF) samples of ICH with community‐acquired pneumonia. Miao et al.\n11\n analyzed 511 specimens (16 types of samples, BALF and sputum samples, approximately 60%, and blood samples, 7.4%) using NGS and traditional culture and discovered that NGS had higher sensitivity and was less likely to be affected by antibiotic exposure. However, obtaining good‐quality BALF or sputum samples remains difficult in many patients, especially in ICH with pulmonary infection; thus, blood specimens may be the main sample type. Therefore, developing a sensitive detection method utilizing NGS for various sample types would assist clinicians in the diagnosis and treatment of ICH with pulmonary infection; however, this has not been studied previously. Because NGS is more expensive than traditional diagnostic methods, it must be evaluated in an appropriate population, such as ICH, with subsequent clinical outcomes and detection efficiency. In this study, we aimed to assess the impact of NGS‐assisted pathogenic detection on the diagnosis, treatment, and outcomes of ICH with pulmonary infection and radiographic evidence of bilateral diffuse lesions.", "This study was conducted at Zhongshan Hospital, Fudan University, a tertiary medical center in Shanghai. Clinical Research Ethics Committee of Zhongshan Hospital, Fudan University (Shanghai, China) approved this study (B2017‐122, B2018‐205). This study enrolled consecutive immunocompromised patients between November 17, 2017, and November 23, 2018. Inclusion criteria involved patients: (1) aged ≥16 years old, (2) with pulmonary infection, (3) who required routine standard microbial detection (either blood samples or respiratory tract specimens), and (4) whose pulmonary imaging revealed bilateral diffuse lesions. Immunocompromise was defined in accordance with the hospital‐acquired pneumonia or ventilator‐associated pneumonia (HAP/VAP) guidelines of the Japanese respiratory society 2009.\n12\n Patients had been immunocompromised for at least 1 month before enrollment.\nAnti‐infection treatment usually takes 24–48 h to take effect, and it is difficult to determine whether the treatment was effective if patients died before the microbial results or less than 48 h after the microbial results. Therefore, we excluded those who died before the microbial results were reported after 48 h. When comparing the sequence of microbial results and patient death, mycobacteria or negative Aspergillus culture results were not considered. In our hospital, at least 168 h is necessary for a negative Aspergillus culture result, and mycobacteria culture may take approximately 6 weeks. In this study, ICH was required for the timely adjustment of anti‐infection treatment. Some patients may die before these two types of results are obtained, and these results had little effect. Only one HIV‐positive patient was transferred to the Shanghai Public Health Clinical Center after the test, and we could not follow‐up on the outcome. Thus, this patient was excluded.", "This was a retrospective study. During the research period, physicians screened patients who met the criteria and asked the research assistants to assess the patients as a group. After obtaining informed consent, patients received free NGS detection (limited by the detective capacity, only a portion of the patients who met the criteria received this extra detection). If the patient survived for at least 48 h after the microbial result, the patient was included in the NGS group. For the non‐NGS group, we screened medical records during the research period and selected all patients who met the study's criteria but did not receive NGS detection. Patient baseline data, including age, sex, acute physiology and chronic health evaluation II (APACHE II) score, type of immunosuppression, experimental examination, treatment, length of hospital stay, and survival data, were collected from medical records and electronic databases. In both groups, sputum, BALF, blood, lung tissue, throat swabs, and pleural effusion samples were collected before antibiotic use. In the non‐NGS group, only conventional microbiological methods, including smears and cultures of normal bacteria and fungi, were used. Subsequently, positive cultures were subjected to drug‐sensitivity tests. Samples from suspected cases of mycobacteria were also sent for acid‐fast bacillus testing and mycobacterial culture. Based on clinical features, serological tests and PCR were used in some cases. These tests included 1,3‐β‐D‐glucan, Cryptococcus capsular antigen, G‐lipopolysaccharide, antibodies against Mycoplasma pneumoniae and Legionella pneumophila, detection of nine main pathogens in the respiratory tract, CMV‐IgG, CMV‐IgM, CMV‐DNA, influenza A/B, and T‐SPOT. Empiric antibiotics were adjusted based on the results of the microbiological tests. For the NGS group, samples were sent to the BGISEQ‐500/100 NGS and conventional pathogenic detection platforms simultaneously, and empiric antibiotics were adjusted later. The follow‐up period was limited to 60 days. Data S1 contains the procedures for NGS detection.", "The detection time is the time duration from sample collection to result (Mycobacteria culture and negative Aspergillus culture not included). If there were more than one sample, we recorded the time of the first positive sample or the first sample (all samples were negative) for the non‐NGS group, and for the NGS group, we recorded the average time of all the samples sent for NGS detection.\nAn NGS report was considered positive if at least one pathogen was detected that met the following three criteria\n11\n, \n13\n: (i) pathogenicity was detected in the lungs; (ii) at least two copy reads were detected; and (iii) if the pathogen was detected in both the negative control sample and the test specimen, the amount of nucleic acid in the test specimen should be at least five times higher.\nEffective positive result is a result required overall consideration of NGS results, traditional microbial results, previous literature, other experiments or imagological examinations, and clinical characteristics by physicians. In this study, we considered an uncertain clinical microbial diagnosis as negative for ease of analysis.\nModification of antibiotic treatment (MAT): Antibiotic treatment was adjusted based on the results of microbial detection.", "Data were analyzed using SPSS 24.0 (SPSS, Chicago, IL, USA). For continuous variables, we used a two‐tailed independent Student's t‐test or the Wilcoxon rank‐sum test. Non‐parametrically distributed variables were assessed using chi‐square (χ2) test or Fisher's exact test. The R and C χ2 test and Bonferroni method were used to compare the proportions of different ICH types between the two groups. Univariate and multivariate analyses were performed using logistic regression. The Kaplan–Meier survival analysis was used to compare survival rate between different groups. Statistical significance was set at P < 0.05.", "Six‐hundred seventy‐eight patients met the enrollment criteria, and after, 322 patients were excluded (321 patients died before or within 48 h after microbial results and one HIV‐positive patient), and 356 patients were included in the final analysis (Figure 1). No significant differences in age, sex, APACHE II scores, proportion of patients with severe pneumonia, multidrug‐resistant bacterial infection, or invasive mechanical ventilation were detected between the two groups. However, the type of immunocompromise differed significantly between the two groups (P < 0.001). After further comparison using the Bonferroni method, we discovered the cause of this difference. The proportion of hematological neoplasms was lower in the non‐NGS group (16.93% vs. 36.27%, P < 0.05), and the proportion of chemotherapy or radiotherapy for solid tumors was higher (16.93% vs. 3.92%, P < 0.05) (Table 1).\nPatient selection. Abbreviations: NGS, next‐generation sequencing; HIV, human immunodeficiency virus\nBaseline characteristics\nAbbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; DM, diabetes mellitus; NGS, next‐generation sequencing; ns, the difference was not significant (by Bonferroni method); OR, odds ratio.\nData are shown with median interquartile range (IQR).\nRenal failure or chronic kidney disease that requires glucocorticoid or immunosuppressant.\nSignificant difference (Bonferroni's method).", "The detection time was shorter in the NGS group than in the non‐NGS group (28.2 h IQR 25.9–29.83 h vs. 50.50 h IQR 47.90–90.91 h, P < 0.001). The positive rate, effective positive rate, rate of mixed infection detected, and proportion of modified antibiotic treatment were significantly higher in the NGS group (89.22% vs. 45.28%, 51.96% vs. 4.72%, 64.71% vs. 21.65%, 46.08% vs. 17.71%, P < 0.001) (Data S2). In total, 123 samples from the NGS group were sent for NGS detection, including 79 blood samples, 28 BALF samples, 15 sputum samples, and 1 pleural effusion sample. The positive rate was not significantly different among the three sample types (blood, sputum, and BALF), and NGS had a higher positive rate than microbial culture in each sample type (86.08% vs. 11.54% in blood samples, 89.29% vs. 26.92% in BALF samples, 100% vs. 54.93% in sputum samples, P < 0.001). In total, 31 types of bacteria (only those with reported evidence of pathogenicity in the lungs), 9 types of fungi, 9 types of viruses, and 2 types of mycobacteria were identified in the NGS group. Pathogens that are frequently detected include Pneumocystis jeroveci (PJ) and cytomegalovirus (CMV). In the non‐NGS group, culture‐positive Candida albicans was the most common pathogen, which was clinically insignificant (important pathogens detected in the NGS group are listed in Figure 2, and the complete catalog is in Data S3).\nImportant pathogens detected by NGS and traditional microbial method in the NGS group. Abbreviations: NTM, non‐tuberculous mycobacterial; MTB, \nMycobacterium tuberculosis\n; CMV, cytomegalovirus; NGS, next‐generation sequencing; PCR, polymerase chain reaction", "In the NGS group, results of the NGS and traditional methods were compared. NGS results were positive in 89.25% of cases, with 46.24% of cases being double‐positive (NGS positive and culture positive). The negative predictive value was 81.82% ± 40.45%. Subsequently, we compared NGS and culture results for these double‐positive cases. Match, partly matched, and mismatched cases accounted for 21.51%, 7.53%, and 9.67% of all cases, respectively. Mismatch cases were mostly caused by culture‐positive C. albicans, and after removing this interference, the proportion declined to 2.15% (Data S4 and S5).", "There was a significant difference in 30‐/60‐day and in‐hospital mortality between the NGS and traditional detection groups (16.67% vs. 28.35%, 18.63% vs. 33.07%, 18.63% vs. 33.07%, P < 0.05). This difference was also significant in Kaplan–Meier curve (Figure 3). Multivariate logistic regression analysis (Table 2) indicated that HAP/VAP (OR 6.41, P = 0.038), invasive ventilation (OR 17.271, P < 0.001), and APACHE II scores (OR 1.899, P < 0.001) were risk factors for 60‐day mortality, and NGS detection was a predictor of survival (OR 0.189, confidence interval [CI], 0.068–0.526). Subsequently, subgroup analysis was used to identify the appropriate population that benefitted from this detection method in terms of survival (Figure 4). Potential survival benefits included age >62 years (OR 0.342, P = 0.007), APACHE II score >10 (OR 0.358, P = 0.004), severe pneumonia (OR 0.047, P = 0.007), radiotherapy or chemotherapy for malignant tumors (OR 0.314, P = 0.047), and multidrug resistant infection (OR 0.052, P = 0.002).\nSurvival curve of the NGS and non‐NGS groups\nUnivariate and multivariate logistic regression analysis for 60‐day mortality\nAbbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; NGS, next‐generation sequencing; OR, odds ratio.\nSubgroup analysis for 60‐day mortality. Abbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; DM, diabetes mellitus; NGS, next‐generation sequencing; OR, odds ratio", "This study was approved by the Clinical Research Ethics Committee of Zhongshan Hospital, Fudan University (Shanghai, China) (B2017‐122, B2018‐205) and informed consent was obtained from every participant. Informed consent was obtained from individual or guardian participants.", "DH Z, SJ C, JJ J, and YL S designed the study and the idea of this article; SJ C, DN H, DH Z, and JJ J enrolled patients; CC C, LL W, XJ T, XY C, L T, and YY Z collected data; DH Z, SJ C, and Y W conducted statistics and analysis of results; DH Z and Y W also conducted manuscript writing and editing; SJ C, JJ J, and YL S revised and corrected the manuscript." ]
[ null, null, null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "MATERIALS AND METHODS", "Patient population", "Study design and data collection", "Criteria for some follow‐up content", "Statistical analysis", "RESULTS", "Study participants and baseline characteristics", "NGS performance", "Concordance between results of NGS and traditional methods", "Outcomes of the two groups", "DISCUSSION", "CONFLICT OF INTEREST", "ETHICS STATEMENT", "AUTHOR CONTRIBUTIONS", "Supporting information" ]
[ "The number of hospitalized patients considered immunocompromised hosts (ICH) continues to increase because of the prevalent use of chemotherapy for cancer, organ transplantation, glucocorticoids, and other immunosuppressive therapies for various disease conditions. For these patients, pulmonary infection is one of the most frequent and fatal complications.\n1\n Respiratory infections and drug toxicity were major concerns in over two‐thirds of ICH cases, and mortality may reach 50–80% if accompanied by respiratory failure.\n2\n, \n3\n Traditional infectious and opportunistic pathogens account for a substantial proportion of infections in these patients. Danés et al.\n4\n discovered opportunistic fungal pneumonia in 38% of non‐human immunodeficiency virus (HIV) ICH patients with etiologically confirmed pneumonia. Therefore, accurate and rapid microbial detection can improve outcomes in these patients. However, the current standard microbial culture has two major drawbacks: low sensitivity and the amount of time it takes to return results.\n5\n\n\nMolecular diagnostic methods, such as polymerase chain reaction (PCR) and next‐generation sequencing (NGS), may provide a solution to this problem. Pathogen sequences can be sorted using deep sequencing and data mining without using a specific primer.\n6\n Several studies have demonstrated the medical benefits of NGS. Guo et al.\n7\n and Ai et al.\n8\n used NGS to monitor disease progression and therapeutic effectiveness. Long et al.\n9\n utilized NGS to detect bacteria in blood samples from patients with sepsis in the intensive care unit (ICU) and discovered greater sensitivity than culture. Pan et al.\n10\n used a case series to reveal the effectiveness of NGS in detecting pathogens in bronchoalveolar lavage fluid (BALF) samples of ICH with community‐acquired pneumonia. Miao et al.\n11\n analyzed 511 specimens (16 types of samples, BALF and sputum samples, approximately 60%, and blood samples, 7.4%) using NGS and traditional culture and discovered that NGS had higher sensitivity and was less likely to be affected by antibiotic exposure. However, obtaining good‐quality BALF or sputum samples remains difficult in many patients, especially in ICH with pulmonary infection; thus, blood specimens may be the main sample type. Therefore, developing a sensitive detection method utilizing NGS for various sample types would assist clinicians in the diagnosis and treatment of ICH with pulmonary infection; however, this has not been studied previously. Because NGS is more expensive than traditional diagnostic methods, it must be evaluated in an appropriate population, such as ICH, with subsequent clinical outcomes and detection efficiency. In this study, we aimed to assess the impact of NGS‐assisted pathogenic detection on the diagnosis, treatment, and outcomes of ICH with pulmonary infection and radiographic evidence of bilateral diffuse lesions.", "[SUBTITLE] Patient population [SUBSECTION] This study was conducted at Zhongshan Hospital, Fudan University, a tertiary medical center in Shanghai. Clinical Research Ethics Committee of Zhongshan Hospital, Fudan University (Shanghai, China) approved this study (B2017‐122, B2018‐205). This study enrolled consecutive immunocompromised patients between November 17, 2017, and November 23, 2018. Inclusion criteria involved patients: (1) aged ≥16 years old, (2) with pulmonary infection, (3) who required routine standard microbial detection (either blood samples or respiratory tract specimens), and (4) whose pulmonary imaging revealed bilateral diffuse lesions. Immunocompromise was defined in accordance with the hospital‐acquired pneumonia or ventilator‐associated pneumonia (HAP/VAP) guidelines of the Japanese respiratory society 2009.\n12\n Patients had been immunocompromised for at least 1 month before enrollment.\nAnti‐infection treatment usually takes 24–48 h to take effect, and it is difficult to determine whether the treatment was effective if patients died before the microbial results or less than 48 h after the microbial results. Therefore, we excluded those who died before the microbial results were reported after 48 h. When comparing the sequence of microbial results and patient death, mycobacteria or negative Aspergillus culture results were not considered. In our hospital, at least 168 h is necessary for a negative Aspergillus culture result, and mycobacteria culture may take approximately 6 weeks. In this study, ICH was required for the timely adjustment of anti‐infection treatment. Some patients may die before these two types of results are obtained, and these results had little effect. Only one HIV‐positive patient was transferred to the Shanghai Public Health Clinical Center after the test, and we could not follow‐up on the outcome. Thus, this patient was excluded.\nThis study was conducted at Zhongshan Hospital, Fudan University, a tertiary medical center in Shanghai. Clinical Research Ethics Committee of Zhongshan Hospital, Fudan University (Shanghai, China) approved this study (B2017‐122, B2018‐205). This study enrolled consecutive immunocompromised patients between November 17, 2017, and November 23, 2018. Inclusion criteria involved patients: (1) aged ≥16 years old, (2) with pulmonary infection, (3) who required routine standard microbial detection (either blood samples or respiratory tract specimens), and (4) whose pulmonary imaging revealed bilateral diffuse lesions. Immunocompromise was defined in accordance with the hospital‐acquired pneumonia or ventilator‐associated pneumonia (HAP/VAP) guidelines of the Japanese respiratory society 2009.\n12\n Patients had been immunocompromised for at least 1 month before enrollment.\nAnti‐infection treatment usually takes 24–48 h to take effect, and it is difficult to determine whether the treatment was effective if patients died before the microbial results or less than 48 h after the microbial results. Therefore, we excluded those who died before the microbial results were reported after 48 h. When comparing the sequence of microbial results and patient death, mycobacteria or negative Aspergillus culture results were not considered. In our hospital, at least 168 h is necessary for a negative Aspergillus culture result, and mycobacteria culture may take approximately 6 weeks. In this study, ICH was required for the timely adjustment of anti‐infection treatment. Some patients may die before these two types of results are obtained, and these results had little effect. Only one HIV‐positive patient was transferred to the Shanghai Public Health Clinical Center after the test, and we could not follow‐up on the outcome. Thus, this patient was excluded.\n[SUBTITLE] Study design and data collection [SUBSECTION] This was a retrospective study. During the research period, physicians screened patients who met the criteria and asked the research assistants to assess the patients as a group. After obtaining informed consent, patients received free NGS detection (limited by the detective capacity, only a portion of the patients who met the criteria received this extra detection). If the patient survived for at least 48 h after the microbial result, the patient was included in the NGS group. For the non‐NGS group, we screened medical records during the research period and selected all patients who met the study's criteria but did not receive NGS detection. Patient baseline data, including age, sex, acute physiology and chronic health evaluation II (APACHE II) score, type of immunosuppression, experimental examination, treatment, length of hospital stay, and survival data, were collected from medical records and electronic databases. In both groups, sputum, BALF, blood, lung tissue, throat swabs, and pleural effusion samples were collected before antibiotic use. In the non‐NGS group, only conventional microbiological methods, including smears and cultures of normal bacteria and fungi, were used. Subsequently, positive cultures were subjected to drug‐sensitivity tests. Samples from suspected cases of mycobacteria were also sent for acid‐fast bacillus testing and mycobacterial culture. Based on clinical features, serological tests and PCR were used in some cases. These tests included 1,3‐β‐D‐glucan, Cryptococcus capsular antigen, G‐lipopolysaccharide, antibodies against Mycoplasma pneumoniae and Legionella pneumophila, detection of nine main pathogens in the respiratory tract, CMV‐IgG, CMV‐IgM, CMV‐DNA, influenza A/B, and T‐SPOT. Empiric antibiotics were adjusted based on the results of the microbiological tests. For the NGS group, samples were sent to the BGISEQ‐500/100 NGS and conventional pathogenic detection platforms simultaneously, and empiric antibiotics were adjusted later. The follow‐up period was limited to 60 days. Data S1 contains the procedures for NGS detection.\nThis was a retrospective study. During the research period, physicians screened patients who met the criteria and asked the research assistants to assess the patients as a group. After obtaining informed consent, patients received free NGS detection (limited by the detective capacity, only a portion of the patients who met the criteria received this extra detection). If the patient survived for at least 48 h after the microbial result, the patient was included in the NGS group. For the non‐NGS group, we screened medical records during the research period and selected all patients who met the study's criteria but did not receive NGS detection. Patient baseline data, including age, sex, acute physiology and chronic health evaluation II (APACHE II) score, type of immunosuppression, experimental examination, treatment, length of hospital stay, and survival data, were collected from medical records and electronic databases. In both groups, sputum, BALF, blood, lung tissue, throat swabs, and pleural effusion samples were collected before antibiotic use. In the non‐NGS group, only conventional microbiological methods, including smears and cultures of normal bacteria and fungi, were used. Subsequently, positive cultures were subjected to drug‐sensitivity tests. Samples from suspected cases of mycobacteria were also sent for acid‐fast bacillus testing and mycobacterial culture. Based on clinical features, serological tests and PCR were used in some cases. These tests included 1,3‐β‐D‐glucan, Cryptococcus capsular antigen, G‐lipopolysaccharide, antibodies against Mycoplasma pneumoniae and Legionella pneumophila, detection of nine main pathogens in the respiratory tract, CMV‐IgG, CMV‐IgM, CMV‐DNA, influenza A/B, and T‐SPOT. Empiric antibiotics were adjusted based on the results of the microbiological tests. For the NGS group, samples were sent to the BGISEQ‐500/100 NGS and conventional pathogenic detection platforms simultaneously, and empiric antibiotics were adjusted later. The follow‐up period was limited to 60 days. Data S1 contains the procedures for NGS detection.\n[SUBTITLE] Criteria for some follow‐up content [SUBSECTION] The detection time is the time duration from sample collection to result (Mycobacteria culture and negative Aspergillus culture not included). If there were more than one sample, we recorded the time of the first positive sample or the first sample (all samples were negative) for the non‐NGS group, and for the NGS group, we recorded the average time of all the samples sent for NGS detection.\nAn NGS report was considered positive if at least one pathogen was detected that met the following three criteria\n11\n, \n13\n: (i) pathogenicity was detected in the lungs; (ii) at least two copy reads were detected; and (iii) if the pathogen was detected in both the negative control sample and the test specimen, the amount of nucleic acid in the test specimen should be at least five times higher.\nEffective positive result is a result required overall consideration of NGS results, traditional microbial results, previous literature, other experiments or imagological examinations, and clinical characteristics by physicians. In this study, we considered an uncertain clinical microbial diagnosis as negative for ease of analysis.\nModification of antibiotic treatment (MAT): Antibiotic treatment was adjusted based on the results of microbial detection.\nThe detection time is the time duration from sample collection to result (Mycobacteria culture and negative Aspergillus culture not included). If there were more than one sample, we recorded the time of the first positive sample or the first sample (all samples were negative) for the non‐NGS group, and for the NGS group, we recorded the average time of all the samples sent for NGS detection.\nAn NGS report was considered positive if at least one pathogen was detected that met the following three criteria\n11\n, \n13\n: (i) pathogenicity was detected in the lungs; (ii) at least two copy reads were detected; and (iii) if the pathogen was detected in both the negative control sample and the test specimen, the amount of nucleic acid in the test specimen should be at least five times higher.\nEffective positive result is a result required overall consideration of NGS results, traditional microbial results, previous literature, other experiments or imagological examinations, and clinical characteristics by physicians. In this study, we considered an uncertain clinical microbial diagnosis as negative for ease of analysis.\nModification of antibiotic treatment (MAT): Antibiotic treatment was adjusted based on the results of microbial detection.\n[SUBTITLE] Statistical analysis [SUBSECTION] Data were analyzed using SPSS 24.0 (SPSS, Chicago, IL, USA). For continuous variables, we used a two‐tailed independent Student's t‐test or the Wilcoxon rank‐sum test. Non‐parametrically distributed variables were assessed using chi‐square (χ2) test or Fisher's exact test. The R and C χ2 test and Bonferroni method were used to compare the proportions of different ICH types between the two groups. Univariate and multivariate analyses were performed using logistic regression. The Kaplan–Meier survival analysis was used to compare survival rate between different groups. Statistical significance was set at P < 0.05.\nData were analyzed using SPSS 24.0 (SPSS, Chicago, IL, USA). For continuous variables, we used a two‐tailed independent Student's t‐test or the Wilcoxon rank‐sum test. Non‐parametrically distributed variables were assessed using chi‐square (χ2) test or Fisher's exact test. The R and C χ2 test and Bonferroni method were used to compare the proportions of different ICH types between the two groups. Univariate and multivariate analyses were performed using logistic regression. The Kaplan–Meier survival analysis was used to compare survival rate between different groups. Statistical significance was set at P < 0.05.", "This study was conducted at Zhongshan Hospital, Fudan University, a tertiary medical center in Shanghai. Clinical Research Ethics Committee of Zhongshan Hospital, Fudan University (Shanghai, China) approved this study (B2017‐122, B2018‐205). This study enrolled consecutive immunocompromised patients between November 17, 2017, and November 23, 2018. Inclusion criteria involved patients: (1) aged ≥16 years old, (2) with pulmonary infection, (3) who required routine standard microbial detection (either blood samples or respiratory tract specimens), and (4) whose pulmonary imaging revealed bilateral diffuse lesions. Immunocompromise was defined in accordance with the hospital‐acquired pneumonia or ventilator‐associated pneumonia (HAP/VAP) guidelines of the Japanese respiratory society 2009.\n12\n Patients had been immunocompromised for at least 1 month before enrollment.\nAnti‐infection treatment usually takes 24–48 h to take effect, and it is difficult to determine whether the treatment was effective if patients died before the microbial results or less than 48 h after the microbial results. Therefore, we excluded those who died before the microbial results were reported after 48 h. When comparing the sequence of microbial results and patient death, mycobacteria or negative Aspergillus culture results were not considered. In our hospital, at least 168 h is necessary for a negative Aspergillus culture result, and mycobacteria culture may take approximately 6 weeks. In this study, ICH was required for the timely adjustment of anti‐infection treatment. Some patients may die before these two types of results are obtained, and these results had little effect. Only one HIV‐positive patient was transferred to the Shanghai Public Health Clinical Center after the test, and we could not follow‐up on the outcome. Thus, this patient was excluded.", "This was a retrospective study. During the research period, physicians screened patients who met the criteria and asked the research assistants to assess the patients as a group. After obtaining informed consent, patients received free NGS detection (limited by the detective capacity, only a portion of the patients who met the criteria received this extra detection). If the patient survived for at least 48 h after the microbial result, the patient was included in the NGS group. For the non‐NGS group, we screened medical records during the research period and selected all patients who met the study's criteria but did not receive NGS detection. Patient baseline data, including age, sex, acute physiology and chronic health evaluation II (APACHE II) score, type of immunosuppression, experimental examination, treatment, length of hospital stay, and survival data, were collected from medical records and electronic databases. In both groups, sputum, BALF, blood, lung tissue, throat swabs, and pleural effusion samples were collected before antibiotic use. In the non‐NGS group, only conventional microbiological methods, including smears and cultures of normal bacteria and fungi, were used. Subsequently, positive cultures were subjected to drug‐sensitivity tests. Samples from suspected cases of mycobacteria were also sent for acid‐fast bacillus testing and mycobacterial culture. Based on clinical features, serological tests and PCR were used in some cases. These tests included 1,3‐β‐D‐glucan, Cryptococcus capsular antigen, G‐lipopolysaccharide, antibodies against Mycoplasma pneumoniae and Legionella pneumophila, detection of nine main pathogens in the respiratory tract, CMV‐IgG, CMV‐IgM, CMV‐DNA, influenza A/B, and T‐SPOT. Empiric antibiotics were adjusted based on the results of the microbiological tests. For the NGS group, samples were sent to the BGISEQ‐500/100 NGS and conventional pathogenic detection platforms simultaneously, and empiric antibiotics were adjusted later. The follow‐up period was limited to 60 days. Data S1 contains the procedures for NGS detection.", "The detection time is the time duration from sample collection to result (Mycobacteria culture and negative Aspergillus culture not included). If there were more than one sample, we recorded the time of the first positive sample or the first sample (all samples were negative) for the non‐NGS group, and for the NGS group, we recorded the average time of all the samples sent for NGS detection.\nAn NGS report was considered positive if at least one pathogen was detected that met the following three criteria\n11\n, \n13\n: (i) pathogenicity was detected in the lungs; (ii) at least two copy reads were detected; and (iii) if the pathogen was detected in both the negative control sample and the test specimen, the amount of nucleic acid in the test specimen should be at least five times higher.\nEffective positive result is a result required overall consideration of NGS results, traditional microbial results, previous literature, other experiments or imagological examinations, and clinical characteristics by physicians. In this study, we considered an uncertain clinical microbial diagnosis as negative for ease of analysis.\nModification of antibiotic treatment (MAT): Antibiotic treatment was adjusted based on the results of microbial detection.", "Data were analyzed using SPSS 24.0 (SPSS, Chicago, IL, USA). For continuous variables, we used a two‐tailed independent Student's t‐test or the Wilcoxon rank‐sum test. Non‐parametrically distributed variables were assessed using chi‐square (χ2) test or Fisher's exact test. The R and C χ2 test and Bonferroni method were used to compare the proportions of different ICH types between the two groups. Univariate and multivariate analyses were performed using logistic regression. The Kaplan–Meier survival analysis was used to compare survival rate between different groups. Statistical significance was set at P < 0.05.", "[SUBTITLE] Study participants and baseline characteristics [SUBSECTION] Six‐hundred seventy‐eight patients met the enrollment criteria, and after, 322 patients were excluded (321 patients died before or within 48 h after microbial results and one HIV‐positive patient), and 356 patients were included in the final analysis (Figure 1). No significant differences in age, sex, APACHE II scores, proportion of patients with severe pneumonia, multidrug‐resistant bacterial infection, or invasive mechanical ventilation were detected between the two groups. However, the type of immunocompromise differed significantly between the two groups (P < 0.001). After further comparison using the Bonferroni method, we discovered the cause of this difference. The proportion of hematological neoplasms was lower in the non‐NGS group (16.93% vs. 36.27%, P < 0.05), and the proportion of chemotherapy or radiotherapy for solid tumors was higher (16.93% vs. 3.92%, P < 0.05) (Table 1).\nPatient selection. Abbreviations: NGS, next‐generation sequencing; HIV, human immunodeficiency virus\nBaseline characteristics\nAbbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; DM, diabetes mellitus; NGS, next‐generation sequencing; ns, the difference was not significant (by Bonferroni method); OR, odds ratio.\nData are shown with median interquartile range (IQR).\nRenal failure or chronic kidney disease that requires glucocorticoid or immunosuppressant.\nSignificant difference (Bonferroni's method).\nSix‐hundred seventy‐eight patients met the enrollment criteria, and after, 322 patients were excluded (321 patients died before or within 48 h after microbial results and one HIV‐positive patient), and 356 patients were included in the final analysis (Figure 1). No significant differences in age, sex, APACHE II scores, proportion of patients with severe pneumonia, multidrug‐resistant bacterial infection, or invasive mechanical ventilation were detected between the two groups. However, the type of immunocompromise differed significantly between the two groups (P < 0.001). After further comparison using the Bonferroni method, we discovered the cause of this difference. The proportion of hematological neoplasms was lower in the non‐NGS group (16.93% vs. 36.27%, P < 0.05), and the proportion of chemotherapy or radiotherapy for solid tumors was higher (16.93% vs. 3.92%, P < 0.05) (Table 1).\nPatient selection. Abbreviations: NGS, next‐generation sequencing; HIV, human immunodeficiency virus\nBaseline characteristics\nAbbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; DM, diabetes mellitus; NGS, next‐generation sequencing; ns, the difference was not significant (by Bonferroni method); OR, odds ratio.\nData are shown with median interquartile range (IQR).\nRenal failure or chronic kidney disease that requires glucocorticoid or immunosuppressant.\nSignificant difference (Bonferroni's method).\n[SUBTITLE] NGS performance [SUBSECTION] The detection time was shorter in the NGS group than in the non‐NGS group (28.2 h IQR 25.9–29.83 h vs. 50.50 h IQR 47.90–90.91 h, P < 0.001). The positive rate, effective positive rate, rate of mixed infection detected, and proportion of modified antibiotic treatment were significantly higher in the NGS group (89.22% vs. 45.28%, 51.96% vs. 4.72%, 64.71% vs. 21.65%, 46.08% vs. 17.71%, P < 0.001) (Data S2). In total, 123 samples from the NGS group were sent for NGS detection, including 79 blood samples, 28 BALF samples, 15 sputum samples, and 1 pleural effusion sample. The positive rate was not significantly different among the three sample types (blood, sputum, and BALF), and NGS had a higher positive rate than microbial culture in each sample type (86.08% vs. 11.54% in blood samples, 89.29% vs. 26.92% in BALF samples, 100% vs. 54.93% in sputum samples, P < 0.001). In total, 31 types of bacteria (only those with reported evidence of pathogenicity in the lungs), 9 types of fungi, 9 types of viruses, and 2 types of mycobacteria were identified in the NGS group. Pathogens that are frequently detected include Pneumocystis jeroveci (PJ) and cytomegalovirus (CMV). In the non‐NGS group, culture‐positive Candida albicans was the most common pathogen, which was clinically insignificant (important pathogens detected in the NGS group are listed in Figure 2, and the complete catalog is in Data S3).\nImportant pathogens detected by NGS and traditional microbial method in the NGS group. Abbreviations: NTM, non‐tuberculous mycobacterial; MTB, \nMycobacterium tuberculosis\n; CMV, cytomegalovirus; NGS, next‐generation sequencing; PCR, polymerase chain reaction\nThe detection time was shorter in the NGS group than in the non‐NGS group (28.2 h IQR 25.9–29.83 h vs. 50.50 h IQR 47.90–90.91 h, P < 0.001). The positive rate, effective positive rate, rate of mixed infection detected, and proportion of modified antibiotic treatment were significantly higher in the NGS group (89.22% vs. 45.28%, 51.96% vs. 4.72%, 64.71% vs. 21.65%, 46.08% vs. 17.71%, P < 0.001) (Data S2). In total, 123 samples from the NGS group were sent for NGS detection, including 79 blood samples, 28 BALF samples, 15 sputum samples, and 1 pleural effusion sample. The positive rate was not significantly different among the three sample types (blood, sputum, and BALF), and NGS had a higher positive rate than microbial culture in each sample type (86.08% vs. 11.54% in blood samples, 89.29% vs. 26.92% in BALF samples, 100% vs. 54.93% in sputum samples, P < 0.001). In total, 31 types of bacteria (only those with reported evidence of pathogenicity in the lungs), 9 types of fungi, 9 types of viruses, and 2 types of mycobacteria were identified in the NGS group. Pathogens that are frequently detected include Pneumocystis jeroveci (PJ) and cytomegalovirus (CMV). In the non‐NGS group, culture‐positive Candida albicans was the most common pathogen, which was clinically insignificant (important pathogens detected in the NGS group are listed in Figure 2, and the complete catalog is in Data S3).\nImportant pathogens detected by NGS and traditional microbial method in the NGS group. Abbreviations: NTM, non‐tuberculous mycobacterial; MTB, \nMycobacterium tuberculosis\n; CMV, cytomegalovirus; NGS, next‐generation sequencing; PCR, polymerase chain reaction\n[SUBTITLE] Concordance between results of NGS and traditional methods [SUBSECTION] In the NGS group, results of the NGS and traditional methods were compared. NGS results were positive in 89.25% of cases, with 46.24% of cases being double‐positive (NGS positive and culture positive). The negative predictive value was 81.82% ± 40.45%. Subsequently, we compared NGS and culture results for these double‐positive cases. Match, partly matched, and mismatched cases accounted for 21.51%, 7.53%, and 9.67% of all cases, respectively. Mismatch cases were mostly caused by culture‐positive C. albicans, and after removing this interference, the proportion declined to 2.15% (Data S4 and S5).\nIn the NGS group, results of the NGS and traditional methods were compared. NGS results were positive in 89.25% of cases, with 46.24% of cases being double‐positive (NGS positive and culture positive). The negative predictive value was 81.82% ± 40.45%. Subsequently, we compared NGS and culture results for these double‐positive cases. Match, partly matched, and mismatched cases accounted for 21.51%, 7.53%, and 9.67% of all cases, respectively. Mismatch cases were mostly caused by culture‐positive C. albicans, and after removing this interference, the proportion declined to 2.15% (Data S4 and S5).\n[SUBTITLE] Outcomes of the two groups [SUBSECTION] There was a significant difference in 30‐/60‐day and in‐hospital mortality between the NGS and traditional detection groups (16.67% vs. 28.35%, 18.63% vs. 33.07%, 18.63% vs. 33.07%, P < 0.05). This difference was also significant in Kaplan–Meier curve (Figure 3). Multivariate logistic regression analysis (Table 2) indicated that HAP/VAP (OR 6.41, P = 0.038), invasive ventilation (OR 17.271, P < 0.001), and APACHE II scores (OR 1.899, P < 0.001) were risk factors for 60‐day mortality, and NGS detection was a predictor of survival (OR 0.189, confidence interval [CI], 0.068–0.526). Subsequently, subgroup analysis was used to identify the appropriate population that benefitted from this detection method in terms of survival (Figure 4). Potential survival benefits included age >62 years (OR 0.342, P = 0.007), APACHE II score >10 (OR 0.358, P = 0.004), severe pneumonia (OR 0.047, P = 0.007), radiotherapy or chemotherapy for malignant tumors (OR 0.314, P = 0.047), and multidrug resistant infection (OR 0.052, P = 0.002).\nSurvival curve of the NGS and non‐NGS groups\nUnivariate and multivariate logistic regression analysis for 60‐day mortality\nAbbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; NGS, next‐generation sequencing; OR, odds ratio.\nSubgroup analysis for 60‐day mortality. Abbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; DM, diabetes mellitus; NGS, next‐generation sequencing; OR, odds ratio\nThere was a significant difference in 30‐/60‐day and in‐hospital mortality between the NGS and traditional detection groups (16.67% vs. 28.35%, 18.63% vs. 33.07%, 18.63% vs. 33.07%, P < 0.05). This difference was also significant in Kaplan–Meier curve (Figure 3). Multivariate logistic regression analysis (Table 2) indicated that HAP/VAP (OR 6.41, P = 0.038), invasive ventilation (OR 17.271, P < 0.001), and APACHE II scores (OR 1.899, P < 0.001) were risk factors for 60‐day mortality, and NGS detection was a predictor of survival (OR 0.189, confidence interval [CI], 0.068–0.526). Subsequently, subgroup analysis was used to identify the appropriate population that benefitted from this detection method in terms of survival (Figure 4). Potential survival benefits included age >62 years (OR 0.342, P = 0.007), APACHE II score >10 (OR 0.358, P = 0.004), severe pneumonia (OR 0.047, P = 0.007), radiotherapy or chemotherapy for malignant tumors (OR 0.314, P = 0.047), and multidrug resistant infection (OR 0.052, P = 0.002).\nSurvival curve of the NGS and non‐NGS groups\nUnivariate and multivariate logistic regression analysis for 60‐day mortality\nAbbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; NGS, next‐generation sequencing; OR, odds ratio.\nSubgroup analysis for 60‐day mortality. Abbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; DM, diabetes mellitus; NGS, next‐generation sequencing; OR, odds ratio", "Six‐hundred seventy‐eight patients met the enrollment criteria, and after, 322 patients were excluded (321 patients died before or within 48 h after microbial results and one HIV‐positive patient), and 356 patients were included in the final analysis (Figure 1). No significant differences in age, sex, APACHE II scores, proportion of patients with severe pneumonia, multidrug‐resistant bacterial infection, or invasive mechanical ventilation were detected between the two groups. However, the type of immunocompromise differed significantly between the two groups (P < 0.001). After further comparison using the Bonferroni method, we discovered the cause of this difference. The proportion of hematological neoplasms was lower in the non‐NGS group (16.93% vs. 36.27%, P < 0.05), and the proportion of chemotherapy or radiotherapy for solid tumors was higher (16.93% vs. 3.92%, P < 0.05) (Table 1).\nPatient selection. Abbreviations: NGS, next‐generation sequencing; HIV, human immunodeficiency virus\nBaseline characteristics\nAbbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; DM, diabetes mellitus; NGS, next‐generation sequencing; ns, the difference was not significant (by Bonferroni method); OR, odds ratio.\nData are shown with median interquartile range (IQR).\nRenal failure or chronic kidney disease that requires glucocorticoid or immunosuppressant.\nSignificant difference (Bonferroni's method).", "The detection time was shorter in the NGS group than in the non‐NGS group (28.2 h IQR 25.9–29.83 h vs. 50.50 h IQR 47.90–90.91 h, P < 0.001). The positive rate, effective positive rate, rate of mixed infection detected, and proportion of modified antibiotic treatment were significantly higher in the NGS group (89.22% vs. 45.28%, 51.96% vs. 4.72%, 64.71% vs. 21.65%, 46.08% vs. 17.71%, P < 0.001) (Data S2). In total, 123 samples from the NGS group were sent for NGS detection, including 79 blood samples, 28 BALF samples, 15 sputum samples, and 1 pleural effusion sample. The positive rate was not significantly different among the three sample types (blood, sputum, and BALF), and NGS had a higher positive rate than microbial culture in each sample type (86.08% vs. 11.54% in blood samples, 89.29% vs. 26.92% in BALF samples, 100% vs. 54.93% in sputum samples, P < 0.001). In total, 31 types of bacteria (only those with reported evidence of pathogenicity in the lungs), 9 types of fungi, 9 types of viruses, and 2 types of mycobacteria were identified in the NGS group. Pathogens that are frequently detected include Pneumocystis jeroveci (PJ) and cytomegalovirus (CMV). In the non‐NGS group, culture‐positive Candida albicans was the most common pathogen, which was clinically insignificant (important pathogens detected in the NGS group are listed in Figure 2, and the complete catalog is in Data S3).\nImportant pathogens detected by NGS and traditional microbial method in the NGS group. Abbreviations: NTM, non‐tuberculous mycobacterial; MTB, \nMycobacterium tuberculosis\n; CMV, cytomegalovirus; NGS, next‐generation sequencing; PCR, polymerase chain reaction", "In the NGS group, results of the NGS and traditional methods were compared. NGS results were positive in 89.25% of cases, with 46.24% of cases being double‐positive (NGS positive and culture positive). The negative predictive value was 81.82% ± 40.45%. Subsequently, we compared NGS and culture results for these double‐positive cases. Match, partly matched, and mismatched cases accounted for 21.51%, 7.53%, and 9.67% of all cases, respectively. Mismatch cases were mostly caused by culture‐positive C. albicans, and after removing this interference, the proportion declined to 2.15% (Data S4 and S5).", "There was a significant difference in 30‐/60‐day and in‐hospital mortality between the NGS and traditional detection groups (16.67% vs. 28.35%, 18.63% vs. 33.07%, 18.63% vs. 33.07%, P < 0.05). This difference was also significant in Kaplan–Meier curve (Figure 3). Multivariate logistic regression analysis (Table 2) indicated that HAP/VAP (OR 6.41, P = 0.038), invasive ventilation (OR 17.271, P < 0.001), and APACHE II scores (OR 1.899, P < 0.001) were risk factors for 60‐day mortality, and NGS detection was a predictor of survival (OR 0.189, confidence interval [CI], 0.068–0.526). Subsequently, subgroup analysis was used to identify the appropriate population that benefitted from this detection method in terms of survival (Figure 4). Potential survival benefits included age >62 years (OR 0.342, P = 0.007), APACHE II score >10 (OR 0.358, P = 0.004), severe pneumonia (OR 0.047, P = 0.007), radiotherapy or chemotherapy for malignant tumors (OR 0.314, P = 0.047), and multidrug resistant infection (OR 0.052, P = 0.002).\nSurvival curve of the NGS and non‐NGS groups\nUnivariate and multivariate logistic regression analysis for 60‐day mortality\nAbbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; NGS, next‐generation sequencing; OR, odds ratio.\nSubgroup analysis for 60‐day mortality. Abbreviations: APACHE, acute physiology and chronic health evaluation; CI, confidence interval; HAP/VAP, hospital‐acquired pneumonia or ventilator‐associated pneumonia; DM, diabetes mellitus; NGS, next‐generation sequencing; OR, odds ratio", "In this study, we discovered that NGS‐assisted pathogenic detection improved diagnosis and treatment and was associated with better clinical outcomes in immunocompromised pulmonary‐infected patients with radiographic evidence of bilateral diffuse lesions. This method reduced microbial detection time, increased detection efficiency, provided more evidence for modifying antibiotic treatment, and was associated with decreased mortality. To our knowledge, this is the first study to evaluate the detection performance, and the impact on treatment and clinical outcomes of NGS‐assisted pathogenic detection in ICH with pulmonary infection. This is also the first study to explore the relationship between molecular detection in blood samples and clinical outcomes of ICH.\nTimely and precise pathogenic diagnosis is essential in ICH. This aids the identification of opportunistic pathogens, and deescalates antibiotics over time.\n14\n, \n15\n, \n16\n Previous studies have indicated that conventional culture examination may require 3–5 days for the final result, and NGS only requires 2–3 days.\n3\n, \n16\n We discovered similar results when comparing detection times. The sequencing procedure only takes approximately 24 h; hence, the total time to obtaining results by improving submission process can be shortened in the future.\n17\n, \n18\n, \n19\n As expected, NGS produced a satisfactory positive rate compared to conventional methods. We used the concept of “effective positive rate” to determine the real pathogen, which required an overall consideration of NGS results, traditional microbial results, previous literature, other experimental or imageological examination, and clinical characteristics by physicians. Based on this concept, the NGS group had a higher rate of detected mixed infection and proportion of modified antibiotic treatment.\nThe advantage of NGS was further demonstrated by comparing the detection performance of the three main types of samples. Miao et al.\n11\n discovered no difference in the positive rate between the three types of samples, which may give clinicians more options. However, only a few blood samples were included, and only the non‐tuberculous mycobacterial‐positive rate was compared between groups.\n11\n In many cases, obtaining ICH sputum samples is difficult, leaving blood samples as the only viable option for diagnostic testing. CMV and PJ, which are often unidentified using traditional microbial culture methods, were prevalent in ICH using NGS. This is consistent with the results of a study by Pan et al.\n10\n In addition, Aspergillus, which was identified by NGS in approximately 10% of all samples, was unidentified using the traditional method. These results suggest the need to consider all possible pathogens in ICH. The negative predictive value was relatively high (81.82% ± 40.45%) was similar to the negative predictive value for NGS (89.2% ± 7.6%) in a study by Parize et al.\n18\n study that compared traditional culture and NGS in the microbial diagnosis of all types of ICH infections using NGS.\nShorter microbial detection times, higher detection efficiency, and timely and reasonable antibiotic treatment adjustments may explain the better outcomes of the NGS group. Several studies have demonstrated the positive impact of rapid diagnosis and antibiotic management in bloodstream infection cases.\n19\n, \n20\n, \n21\n Recently, Xie et al.\n22\n discovered that NGS pathogenic detection reduced mortality in ICU patients with severe pneumonia. This was a retrospective study with a relatively small sample size, and the detection time was not reported. Here, we attempt to confirm and explain this conclusion in a study using a larger sample size. We focused on ICH, in whom the infection diagnosis is more difficult using conventional diagnostic methods.\nAfter the multivariate logistic regression analysis, NGS remained a significant predictor of survival. In the subgroup analysis to determine the appropriate population for NGS detection, patients over 62 years of age or with high APACHE II scores received a survival benefit. Similar to the study by Xie et al.,\n22\n this detection method was associated with reduced mortality in patients with severe pneumonia in our study. Our study was underpowered to draw conclusions specific to patients with multidrug resistant infections.\nOur findings suggest that NGS‐assisted pathogenic detection works well in immunocompromised pulmonary infection patients with radiographic evidence of bilateral diffuse lesions, and that patients over 62 years or with high APACHE II scores (≥10) may benefit more. However, this study had several limitations. First, it was a non‐randomized clinical trial single‐center study, population size was not very large and patients were not enrolled prospectively. Second, not all patients in the NGS group underwent NGS detection for blood samples. Third, NGS detection in this study only covered DNA, thus eliminating potential viruses. This study provides strong evidence to justify further studies, including randomized trials, to determine whether NGS can improve clinical decision‐making and subsequent outcomes in ICH with pulmonary infection.", "The authors have no conflict of interest to declare.", "This study was approved by the Clinical Research Ethics Committee of Zhongshan Hospital, Fudan University (Shanghai, China) (B2017‐122, B2018‐205) and informed consent was obtained from every participant. Informed consent was obtained from individual or guardian participants.", "DH Z, SJ C, JJ J, and YL S designed the study and the idea of this article; SJ C, DN H, DH Z, and JJ J enrolled patients; CC C, LL W, XJ T, XY C, L T, and YY Z collected data; DH Z, SJ C, and Y W conducted statistics and analysis of results; DH Z and Y W also conducted manuscript writing and editing; SJ C, JJ J, and YL S revised and corrected the manuscript.", "\nData S1. Supporting Information\nClick here for additional data file.\n\nData S2. Supporting Information\nClick here for additional data file.\n\nData S3. Supporting Information\nClick here for additional data file.\n\nData S4. Supporting Information\nClick here for additional data file.\n\nData S5. Supporting Information\nClick here for additional data file." ]
[ null, "materials-and-methods", null, null, null, null, "results", null, null, null, null, "discussion", "COI-statement", null, null, "supplementary-material" ]
[ "immunocompromised host", "next generation sequencing", "outcome", "pulmonary infection" ]
Evaluation of large airway specimens obtained by transbronchial lung cryobiopsy in diffuse parenchymal lung diseases.
36258160
The difference in diagnostic yield between surgical lung biopsy and transbronchial lung cryobiopsy (TBLC) in diffuse parenchymal lung diseases (DPLD) has been reported to be due to differences in the rate of interpathologist agreement, specimen size, and specimen adequacy. In TBLC, the specimens containing large airway components are generally believed as inadequate specimens for histological evaluation, but the detailed characteristics of TBLC specimens including the large airway and the impact on histological diagnostic rates of DPLD have not been investigated.
BACKGROUND
We retrospectively reviewed the specimen characteristics of patients with DPLD who underwent TBLC.
METHODS
Between February 2018 and January 2020, 74 patients and 177 specimens were included. There were 85 (48.0%) large airway specimens (LAS) that contained bronchial gland or bronchial cartilage. The ideal specimen ratio was significantly lower in the LAS-positive group than that in the LAS-negative group (5.8% vs. 45.6%), and the proportion of bronchioles, alveoli, and perilobular area were similarly lower in the LAS-positive group. The presence of traction bronchiectasis and diaphragm overlap sign on high-resolution computed tomography (HRCT) were also significantly higher in the LAS-positive group than those in the LAS-negative group. We observed a statistically significant trend in histological diagnostic yield (40.7% in LAS positive group; 60.8% in LAS positive and negative group; 91.6% in LAS negative group) (Cochran-Armitage trend test).
RESULTS
LAS is a specimen often collected in TBLC and contains a low percentage of bronchioles, alveoli, and perilobular area. Since the histological diagnostic yield tends to be higher in cases that do not contain LAS, it may be important to determine the biopsy site that reduces the frequency of LAS collection by referring to the HRCT findings in TBLC.
CONCLUSION
[ "Humans", "Bronchoscopy", "Retrospective Studies", "Lung Diseases, Interstitial", "Lung", "Biopsy" ]
9578247
null
null
null
null
Results
[SUBTITLE] Patients [SUBSECTION] This study included 90 consecutive patients evaluated between February 2018 and January 2020. We excluded 16 patients: 9 were missing pulmonary function tests within 3 months of TBLC, 3 were biopsied for peripheral lung lesions using EBUS, 2 were biopsied for central-type lung cancer, and 2 had an inappropriate pathological evaluation. In total, 74 patients were available in this study, and their baseline characteristics are as indicated in Table 1 for the three groups: LAS-positive, LAS-positive and LAS-negative, LAS-negative. There were 50 LAS-positive cases (67.5%), of which all specimens were LAS in 27 cases and only some specimens were LAS in 23 cases. HRCT pattern accounted for approximately 90% of indeterminate for UIP and alternative diagnoses. All cases with HRCT pattern UIP were positive for some collagen-related autoantibodies or myeloperoxidase-antineutrophil cytoplasmic antibody. Table 1Baseline characteristics of patientsCharacteristicsAll patientsLAS-positiveLAS-positiveandLAS-negativeLAS-negative P valuePatients, n7427 (36.4)23 (31.0)24 (32.4)Male, n (%)40 (54.0)14 (51.8)15 (65.2)11 (45.8)0.698Age, years67.0 ± 11.569.1 ± 7.167.6 ± 8.664.0 ± 16.70.278Current or ex-smokers, n (%)44 (59.4)16 (59.2)17 (73.9)11 (45.8)0.358KL-6, U/mL1565.1 ± 1523.91473 ± 10511784 ± 20481455 ± 14240.713SP-D, ng/mL346.4 ± 293.0339.7 ± 279.9372.1 ± 339.5327.8 ± 267.40.873PFT %FVC81.8 ± 20.278.9 ± 18.480.1 ± 18.086.8 ± 23.80.347 FEV1/FVC ratio79.9 ± 10.280.4 ± 11.981.5 ± 8.977.9 ± 9.30.483 %DLCO64.1 ± 24.568.0 ± 27.263.9 ± 23.160.3 ± 23.30.584HRCT findings, n (%) Honeycomb19 (25.6)7 (25.9)5 (18.5)7 (29.1)0.805 Traction bronchiectasis50 (67.5)22 (81.4)15 (65.2)13 (54.1) < 0.05  Reticulation63 (85.1)25 (92.5)20 (86.9)18 (75.0)0.07 GGO55 (74.3)19 (70.3)16 (69.5)20 (83.3)0.3 Consolidation17 (22.9)5 (18.5)6 (26.0)6 (25.0)0.573 Nodule17 (22.9)7 (25.9)4 (17.3)6 (25.0)0.917 Cyst12 (16.2)4 (14.8)4 (17.3)4 (16.6)0.853HRCT pattern, n (%) UIP5 (6.7)1 (3.7)3 (13.0)1 (4.1)0.91 Probable UIP4 (5.4)2 (7.4)1 (4.3)1 (4.1)0.603 Indeterminate for UIP27 (36.4)14 (51.8)7 (30.4)6 (25.0) < 0.05  Alternative diagnosis38 (51.3)10 (37.0)12 (52.1)16 (66.6) < 0.05 Histological diagnosis, n (%)47 (63.5)11 (40.7)14 (60.8)22 (91.6) < 0.05 Number of specimens, n2.3 ± 0.72.1 ± 0.82.6 ± 0.52.4 ± 0.80.06Bronchial bleeding48 (64.8)15 (55.5)20 (86.9)12 (50.0)0.75 Mild27 (36.4)6 (22.2)13 (56.5)8 (33.3)0.367 Moderate21 (28.3)9 (33.3)7 (30.4)4 (16.6)0.186Pneumothorax5 (6.7)0 (0)0 (0)5 (20.8) < 0.05  Mild3 (4.0)0 (0)0 (0)3 (12.5) < 0.05  Moderate1 (1.3)0 (0)0 (0)1 (4.1)0.206 Severe1 (1.3)0 (0)0 (0)1 (4.1)0.206Highlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant differenceDLCO, diffusing capacity for carbon monoxide; FEV1, forced expiratory volume in 1 s; FVC, forced volume capacity; GGO, ground-glass opacities; HRCT, high-resolution computed tomography; KL-6, Krebs von den Lungen-6; LAS, large airway specimen; PFT, pulmonary function test; SP-D, surfactant protein D; UIP, usual interstitial pneumonia Baseline characteristics of patients Highlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant difference DLCO, diffusing capacity for carbon monoxide; FEV1, forced expiratory volume in 1 s; FVC, forced volume capacity; GGO, ground-glass opacities; HRCT, high-resolution computed tomography; KL-6, Krebs von den Lungen-6; LAS, large airway specimen; PFT, pulmonary function test; SP-D, surfactant protein D; UIP, usual interstitial pneumonia This study included 90 consecutive patients evaluated between February 2018 and January 2020. We excluded 16 patients: 9 were missing pulmonary function tests within 3 months of TBLC, 3 were biopsied for peripheral lung lesions using EBUS, 2 were biopsied for central-type lung cancer, and 2 had an inappropriate pathological evaluation. In total, 74 patients were available in this study, and their baseline characteristics are as indicated in Table 1 for the three groups: LAS-positive, LAS-positive and LAS-negative, LAS-negative. There were 50 LAS-positive cases (67.5%), of which all specimens were LAS in 27 cases and only some specimens were LAS in 23 cases. HRCT pattern accounted for approximately 90% of indeterminate for UIP and alternative diagnoses. All cases with HRCT pattern UIP were positive for some collagen-related autoantibodies or myeloperoxidase-antineutrophil cytoplasmic antibody. Table 1Baseline characteristics of patientsCharacteristicsAll patientsLAS-positiveLAS-positiveandLAS-negativeLAS-negative P valuePatients, n7427 (36.4)23 (31.0)24 (32.4)Male, n (%)40 (54.0)14 (51.8)15 (65.2)11 (45.8)0.698Age, years67.0 ± 11.569.1 ± 7.167.6 ± 8.664.0 ± 16.70.278Current or ex-smokers, n (%)44 (59.4)16 (59.2)17 (73.9)11 (45.8)0.358KL-6, U/mL1565.1 ± 1523.91473 ± 10511784 ± 20481455 ± 14240.713SP-D, ng/mL346.4 ± 293.0339.7 ± 279.9372.1 ± 339.5327.8 ± 267.40.873PFT %FVC81.8 ± 20.278.9 ± 18.480.1 ± 18.086.8 ± 23.80.347 FEV1/FVC ratio79.9 ± 10.280.4 ± 11.981.5 ± 8.977.9 ± 9.30.483 %DLCO64.1 ± 24.568.0 ± 27.263.9 ± 23.160.3 ± 23.30.584HRCT findings, n (%) Honeycomb19 (25.6)7 (25.9)5 (18.5)7 (29.1)0.805 Traction bronchiectasis50 (67.5)22 (81.4)15 (65.2)13 (54.1) < 0.05  Reticulation63 (85.1)25 (92.5)20 (86.9)18 (75.0)0.07 GGO55 (74.3)19 (70.3)16 (69.5)20 (83.3)0.3 Consolidation17 (22.9)5 (18.5)6 (26.0)6 (25.0)0.573 Nodule17 (22.9)7 (25.9)4 (17.3)6 (25.0)0.917 Cyst12 (16.2)4 (14.8)4 (17.3)4 (16.6)0.853HRCT pattern, n (%) UIP5 (6.7)1 (3.7)3 (13.0)1 (4.1)0.91 Probable UIP4 (5.4)2 (7.4)1 (4.3)1 (4.1)0.603 Indeterminate for UIP27 (36.4)14 (51.8)7 (30.4)6 (25.0) < 0.05  Alternative diagnosis38 (51.3)10 (37.0)12 (52.1)16 (66.6) < 0.05 Histological diagnosis, n (%)47 (63.5)11 (40.7)14 (60.8)22 (91.6) < 0.05 Number of specimens, n2.3 ± 0.72.1 ± 0.82.6 ± 0.52.4 ± 0.80.06Bronchial bleeding48 (64.8)15 (55.5)20 (86.9)12 (50.0)0.75 Mild27 (36.4)6 (22.2)13 (56.5)8 (33.3)0.367 Moderate21 (28.3)9 (33.3)7 (30.4)4 (16.6)0.186Pneumothorax5 (6.7)0 (0)0 (0)5 (20.8) < 0.05  Mild3 (4.0)0 (0)0 (0)3 (12.5) < 0.05  Moderate1 (1.3)0 (0)0 (0)1 (4.1)0.206 Severe1 (1.3)0 (0)0 (0)1 (4.1)0.206Highlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant differenceDLCO, diffusing capacity for carbon monoxide; FEV1, forced expiratory volume in 1 s; FVC, forced volume capacity; GGO, ground-glass opacities; HRCT, high-resolution computed tomography; KL-6, Krebs von den Lungen-6; LAS, large airway specimen; PFT, pulmonary function test; SP-D, surfactant protein D; UIP, usual interstitial pneumonia Baseline characteristics of patients Highlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant difference DLCO, diffusing capacity for carbon monoxide; FEV1, forced expiratory volume in 1 s; FVC, forced volume capacity; GGO, ground-glass opacities; HRCT, high-resolution computed tomography; KL-6, Krebs von den Lungen-6; LAS, large airway specimen; PFT, pulmonary function test; SP-D, surfactant protein D; UIP, usual interstitial pneumonia [SUBTITLE] Diagnostic yield and histological diagnosis [SUBSECTION] A specific histological diagnosis was obtained in 47/74 cases (63.5%). The histological diagnostic yield tended to be statistically significantly higher in the LAS-negative group (40.7% in LAS positive group; 60.8% in LAS positive and negative group; 91.6% in LAS negative group) (Cochran-Armitage trend test). The histological interpretations are shown in Table 2 and include 11 (14.8%) UIP, 6 (8.1) probable UIP, 16 (21.6%) indeterminate for UIP, 9 (12.1%) non-specific interstitial pneumonia, 6 (8.1%) organizing pneumonia and 6 (8.1%) hypersensitivity pneumonitis. The institutional diagnosis after MDD is shown in Table 3 and include 9 (12.1%) idiopathic pulmonary fibrosis, 6 (8.1%) non-specific interstitial pneumonia, 4 (5.4%) organizing pneumonia, 24 (32.4%) unclassifiable and 9 (12.1%) hypersensitivity pneumonitis. Table 2Histological diagnosisHistological diagnosisNo (%)UIP11 (14.8)Probable UIP6 (8.1)Indeterminate for UIP16 (21.6)– 14 NSIP/UIP– 1 NSIP/UIP/OP– 1 UIP/OPAlternative diagnosis NSIP9 (12.1) OP6 (8.1) HP6 (8.1) Smoking-related IP2 (2.7) Others7 (9.4)– 4 IgG4-related disease– 1 sarcoidosis– 1 cellular bronchiolitis– 1 malignancyN.D.11 (14.8)DPB, diffuse panbronchiolitis; HP, hypersensitivity pneumonitis; N.D., non-diagnostic; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; UIP, usual interstitial pneumoniaTable 3MDD diagnosisMDD diagnosisNo (%)IPF9 (12.1)NSIP6 (8.1)OP4 (5.4)Unclassifiable24 (32.4)Hypersensitivity pneumonitis9 (12.1)CTD-ILD (MCTD/RA/SjS/SLE/SSc)10 (13.5)MPA3 (4.0)Others9 (12.1)CTD, connective tissue disease; IPF, idiopathic pulmonary fibrosis; MCTD, mixed connective tissue disease; MPA, microscopic polyangiitis; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; RA, rheumatoid arthritis; SjS, Sjögren’s syndrome; SLE, systemic lupus erythematosus; SSc, systemic sclerosis Histological diagnosis DPB, diffuse panbronchiolitis; HP, hypersensitivity pneumonitis; N.D., non-diagnostic; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; UIP, usual interstitial pneumonia MDD diagnosis CTD, connective tissue disease; IPF, idiopathic pulmonary fibrosis; MCTD, mixed connective tissue disease; MPA, microscopic polyangiitis; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; RA, rheumatoid arthritis; SjS, Sjögren’s syndrome; SLE, systemic lupus erythematosus; SSc, systemic sclerosis A specific histological diagnosis was obtained in 47/74 cases (63.5%). The histological diagnostic yield tended to be statistically significantly higher in the LAS-negative group (40.7% in LAS positive group; 60.8% in LAS positive and negative group; 91.6% in LAS negative group) (Cochran-Armitage trend test). The histological interpretations are shown in Table 2 and include 11 (14.8%) UIP, 6 (8.1) probable UIP, 16 (21.6%) indeterminate for UIP, 9 (12.1%) non-specific interstitial pneumonia, 6 (8.1%) organizing pneumonia and 6 (8.1%) hypersensitivity pneumonitis. The institutional diagnosis after MDD is shown in Table 3 and include 9 (12.1%) idiopathic pulmonary fibrosis, 6 (8.1%) non-specific interstitial pneumonia, 4 (5.4%) organizing pneumonia, 24 (32.4%) unclassifiable and 9 (12.1%) hypersensitivity pneumonitis. Table 2Histological diagnosisHistological diagnosisNo (%)UIP11 (14.8)Probable UIP6 (8.1)Indeterminate for UIP16 (21.6)– 14 NSIP/UIP– 1 NSIP/UIP/OP– 1 UIP/OPAlternative diagnosis NSIP9 (12.1) OP6 (8.1) HP6 (8.1) Smoking-related IP2 (2.7) Others7 (9.4)– 4 IgG4-related disease– 1 sarcoidosis– 1 cellular bronchiolitis– 1 malignancyN.D.11 (14.8)DPB, diffuse panbronchiolitis; HP, hypersensitivity pneumonitis; N.D., non-diagnostic; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; UIP, usual interstitial pneumoniaTable 3MDD diagnosisMDD diagnosisNo (%)IPF9 (12.1)NSIP6 (8.1)OP4 (5.4)Unclassifiable24 (32.4)Hypersensitivity pneumonitis9 (12.1)CTD-ILD (MCTD/RA/SjS/SLE/SSc)10 (13.5)MPA3 (4.0)Others9 (12.1)CTD, connective tissue disease; IPF, idiopathic pulmonary fibrosis; MCTD, mixed connective tissue disease; MPA, microscopic polyangiitis; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; RA, rheumatoid arthritis; SjS, Sjögren’s syndrome; SLE, systemic lupus erythematosus; SSc, systemic sclerosis Histological diagnosis DPB, diffuse panbronchiolitis; HP, hypersensitivity pneumonitis; N.D., non-diagnostic; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; UIP, usual interstitial pneumonia MDD diagnosis CTD, connective tissue disease; IPF, idiopathic pulmonary fibrosis; MCTD, mixed connective tissue disease; MPA, microscopic polyangiitis; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; RA, rheumatoid arthritis; SjS, Sjögren’s syndrome; SLE, systemic lupus erythematosus; SSc, systemic sclerosis [SUBTITLE] Complications [SUBSECTION] In a comparison between the three groups, the LAS-negative group tended to have more pneumothorax complications than the other groups (0% in LAS positive group; 0% in LAS positive and negative group; 20.8% in LAS negative group) (Cochran-Armitage trend test). There was no clear difference in bronchial bleeding between the three groups (Table 1). In a comparison between the three groups, the LAS-negative group tended to have more pneumothorax complications than the other groups (0% in LAS positive group; 0% in LAS positive and negative group; 20.8% in LAS negative group) (Cochran-Armitage trend test). There was no clear difference in bronchial bleeding between the three groups (Table 1). [SUBTITLE] Specimen characteristics [SUBSECTION] In total, 177 specimens were collected in the 74 cases, with a mean number of specimens per case of 2.3 ± 0.7 (Tables 1 and 4). 175 were collected with 1.9 mm probes and only 2 were collected with 2.4 mm probes. The most common biopsy lobe was the right lower lobe (63.8%), followed by the right upper lobe (15.2%) and the left lower lobe (14.1%). There were 85 (48.0%) LAS containing bronchial gland or bronchial cartilage: 30 containing both bronchial gland and bronchial cartilage, 50 containing only bronchial cartilage, and 5 containing only bronchial gland. The anatomical structures observed except for the large airway were alveoli (91.5%), bronchioles (49.1%), and perilobular area (43.5%), with few pleural specimens (1.2%). Ideal specimens were obtained in 47 of the 177 (26.5%) specimens, about a quarter of the total. A comparison of specimen characteristics showed that the proportion of bronchioles, alveoli, and perilobular area were significantly lower in the LAS-positive group than in the LAS-negative group (Table 3). Similarly, the ideal specimen ratio and the frequency of specimens of high pathological confidence were significantly lower in the LAS-positive group. The ratio of specimens with a pathological confidence level A was 25 of the 47 (53.1%) for ideal specimens and 43 of the 130 (33.0%) for unideal specimens (P < 0.05). In terms of the HRCT findings at the sample collection site, the LAS-positive group had a significantly higher proportion of traction bronchiectasis (65.5% vs. 29.6%) and diaphragm overlap sign (54.1% vs. 33.6%) than the LAS-negative group. Table 4Characteristics of specimens obtained by TBLCCharacteristicsAll specimensLAS positiveLAS negative P valueSpecimens, n (%)17785 (48.0)92 (51.9)Maximum diameter, mm6.4 ± 2.97.5 ± 3.75.3 ± 1.3 < 0.05 Biopsy lobe, n (%) Right upper lobe27 (15.2)7 (8.2)20 (21.7) < 0.05  Right middle lobe6 (3.3)2 (2.3)4 (4.3)0.684 Right lower lobe113 (63.8)58 (68.2)55 (59.7)0.275 Left upper lobe6 (3.3)2 (2.3)4 (4.2)0.684 Left lower lobe25 (14,1)16 (18.8)9 (9.7)0.129Structures observed on the specimen Bronchiole87 (49.1)23 (27.0)64 (69.5) < 0.05  Alveoli162 (91.5)73 (85.8)89 (96.7) < 0.05  Perilobular area77 (43.5)15 (17.6)62 (67.3) < 0.05  Pleura1 (1.2)0 (0)1 (1.0)1Ideal specimen, n (%)47 (26.5)5 (5.8)42 (45.6) < 0.05 Pathological confidence level Level A68 (38.4)23 (27.0)45 (48.9) < 0.05  Level B84 (47.4)48 (56.4)36 (39.1) < 0.05  Level C10 (5.6)5 (5.8)5 (5.4)1 Not evaluated15 (8.4)9 (10.5)6 (6.5)HRCT findings in the biopsied lung segment, n (%) Honeycomb8 (4.5)4 (4.7)4 (4.3)1 Traction bronchiectasis101 (57.0)65 (76.4)36 (39.1) < 0.05  Reticulation130 (73.4)68 (80.0)62 (67.3)0.063 GGO60 (33.8)28 (32.9)32 (34.7)0.874 Consolidation16 (9.0)8 (9.4)8 (8.6)1 Nodule14 (7.9)4 (4.7)10 (10.8)0.167 Cyst5 (2.8)0 (0)1 (1.0)0.059Diaphragm overlap sign, n (%)77 (43.5)46 (54.1)31 (33.6) < 0.05 Highlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant differenceGGO, ground-glass opacities; HRCT, high-resolution computed tomography; LAS, large airway specimen Characteristics of specimens obtained by TBLC Highlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant difference GGO, ground-glass opacities; HRCT, high-resolution computed tomography; LAS, large airway specimen In total, 177 specimens were collected in the 74 cases, with a mean number of specimens per case of 2.3 ± 0.7 (Tables 1 and 4). 175 were collected with 1.9 mm probes and only 2 were collected with 2.4 mm probes. The most common biopsy lobe was the right lower lobe (63.8%), followed by the right upper lobe (15.2%) and the left lower lobe (14.1%). There were 85 (48.0%) LAS containing bronchial gland or bronchial cartilage: 30 containing both bronchial gland and bronchial cartilage, 50 containing only bronchial cartilage, and 5 containing only bronchial gland. The anatomical structures observed except for the large airway were alveoli (91.5%), bronchioles (49.1%), and perilobular area (43.5%), with few pleural specimens (1.2%). Ideal specimens were obtained in 47 of the 177 (26.5%) specimens, about a quarter of the total. A comparison of specimen characteristics showed that the proportion of bronchioles, alveoli, and perilobular area were significantly lower in the LAS-positive group than in the LAS-negative group (Table 3). Similarly, the ideal specimen ratio and the frequency of specimens of high pathological confidence were significantly lower in the LAS-positive group. The ratio of specimens with a pathological confidence level A was 25 of the 47 (53.1%) for ideal specimens and 43 of the 130 (33.0%) for unideal specimens (P < 0.05). In terms of the HRCT findings at the sample collection site, the LAS-positive group had a significantly higher proportion of traction bronchiectasis (65.5% vs. 29.6%) and diaphragm overlap sign (54.1% vs. 33.6%) than the LAS-negative group. Table 4Characteristics of specimens obtained by TBLCCharacteristicsAll specimensLAS positiveLAS negative P valueSpecimens, n (%)17785 (48.0)92 (51.9)Maximum diameter, mm6.4 ± 2.97.5 ± 3.75.3 ± 1.3 < 0.05 Biopsy lobe, n (%) Right upper lobe27 (15.2)7 (8.2)20 (21.7) < 0.05  Right middle lobe6 (3.3)2 (2.3)4 (4.3)0.684 Right lower lobe113 (63.8)58 (68.2)55 (59.7)0.275 Left upper lobe6 (3.3)2 (2.3)4 (4.2)0.684 Left lower lobe25 (14,1)16 (18.8)9 (9.7)0.129Structures observed on the specimen Bronchiole87 (49.1)23 (27.0)64 (69.5) < 0.05  Alveoli162 (91.5)73 (85.8)89 (96.7) < 0.05  Perilobular area77 (43.5)15 (17.6)62 (67.3) < 0.05  Pleura1 (1.2)0 (0)1 (1.0)1Ideal specimen, n (%)47 (26.5)5 (5.8)42 (45.6) < 0.05 Pathological confidence level Level A68 (38.4)23 (27.0)45 (48.9) < 0.05  Level B84 (47.4)48 (56.4)36 (39.1) < 0.05  Level C10 (5.6)5 (5.8)5 (5.4)1 Not evaluated15 (8.4)9 (10.5)6 (6.5)HRCT findings in the biopsied lung segment, n (%) Honeycomb8 (4.5)4 (4.7)4 (4.3)1 Traction bronchiectasis101 (57.0)65 (76.4)36 (39.1) < 0.05  Reticulation130 (73.4)68 (80.0)62 (67.3)0.063 GGO60 (33.8)28 (32.9)32 (34.7)0.874 Consolidation16 (9.0)8 (9.4)8 (8.6)1 Nodule14 (7.9)4 (4.7)10 (10.8)0.167 Cyst5 (2.8)0 (0)1 (1.0)0.059Diaphragm overlap sign, n (%)77 (43.5)46 (54.1)31 (33.6) < 0.05 Highlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant differenceGGO, ground-glass opacities; HRCT, high-resolution computed tomography; LAS, large airway specimen Characteristics of specimens obtained by TBLC Highlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant difference GGO, ground-glass opacities; HRCT, high-resolution computed tomography; LAS, large airway specimen
Conclusion
In conclusion, this study showed that LAS obtained in TBLC have a high frequency of unideal specimens that do not include the bronchiole or perilobular area. Since the histological diagnostic yield tends to be higher in cases that do not contain LAS, it may be important to determine the biopsy site that reduces the frequency of LAS collection by referring to the HRCT findings in TBLC.
[ "Background", "Patients", "Data collection", "Transbronchial cryobiopsy procedure", "Pathological evaluation", "Study design", "Statistical analysis", "Patients", "Diagnostic yield and histological diagnosis", "Complications", "Specimen characteristics" ]
[ "Diffuse parenchymal lung diseases (DPLD) comprise heterogeneous disorders with different prognostic and therapeutic implications. Many international guidelines have required confirmation of the pathological findings by surgical lung biopsy (SLB) to recognize histological patterns and provide an accurate diagnosis by obtaining a sufficient volume of specimens [1, 2]. SLB has a histological diagnostic yield of more than 90%, but it has disadvantages of certain morbidity and mortality and the presence of patients excluded from the indication due to advanced age, complications including pulmonary hypertension, and severe respiratory failure [3, 4]. Transbronchial lung cryobiopsy (TBLC) is a promising new bronchoscopic biopsy technique with lower mortality and fewer complications, but the diagnostic yield for DPLD is 70–80%, which is inferior to that of SLB [5]. This difference in diagnostic yield between SLB and TBLC has been reported to be due to the rate of interpathologist agreement, specimen size, and specimen adequacy [6–9].\nAs mentioned in previous TBLC reports for DPLD [10, 11], specimens containing large airway are considered unideal with little contribution to the histological diagnosis. However, the specific conditions under which these specimens are likely to be collected and their actual impact on diagnostic rates are not known. The purpose of this study was to examine the characteristics of TBLC specimens containing large airway and their impact on the rate of histological diagnosis in individual cases.", "This retrospective study was conducted in the Department of Respiratory Medicine of Saitama Red Cross Hospital between February 2018 and January 2020 and was approved from the institutional review board of Saitama Red Cross Hospital (approval number 19-C). The need for patient approval and/or informed consent was waived due to the retrospective nature of the study. The inclusion criteria were age > 20 years and patients with DPLD who underwent TBLC. Exclusion criteria were as follows: patients who underwent biopsy from a site directly visible by bronchoscopy, patients for whom biopsy was performed after confirmation by endobronchial ultrasonography (EBUS) at a peripheral site, patients who met the pulmonary function test criteria for relative contraindications to TBLC as indicated in a previous report [12]; %forced vital capacity < 50% and %diffusion capacity of the lung for carbon monoxide < 35%, and patients for whom pulmonary function test, high-resolution computed tomography (HRCT), or laboratory findings were missing within 3 months of TBLC.", "Baseline clinical measurements were obtained within 3 months of the bronchoscopy. Each patient’s HRCT scan was reviewed in a multi-disciplinary discussion and classified as presenting an HRCT pattern of usual interstitial pneumonia (UIP), probable UIP, indeterminate for UIP, or alternative diagnosis according to the American Thoracic Society (ATS)/European Respiratory Society (ERS)/Japanese Respiratory Society (JRS)/Latin American Thoracic Association (ALAT) guideline 2018 [13]. Definitions of terms related to the HRCT findings were based on the definitions of the Fleischer Society [14].", "Bronchoscopy was performed with a BF-1T290 flexible bronchoscope (Olympus, Tokyo, Japan). Patients were anesthetized with midazolam and fentanyl intravenously, and 2% lidocaine was added intratracheally as appropriate. The endotracheal tube (SACETT Suction Above Cuff Endotracheal Tube 8.0 mm; Smiths Medical International Ltd., Minneapolis, MN, USA) was inserted for airway control. An endobronchial balloon catheter (Fogarty catheter, E-080-4F; Edwards Life-sciences, Irvine, CA, USA) was routinely used for bronchial blockade. The flexible reusable cryoprobe, either 1.9 mm or 2.4 mm in diameter (ERBECRYO® 2 system; Erbe Elektromedizin GmbH, Tübingen, Germany) was inserted from a peripheral bronchus to just below the pleura under fluoroscopy. The probe was withdrawn for 1 cm proximally to prevent pneumothorax. After freezing the probe for 5–7 s, the bronchoscope was withdrawn along with the biopsy specimen and probe, and the endobronchial balloon catheter was inflated simultaneously. In accordance with previous reports [15], Complications were classified into four levels. “Serious” adverse events were defined as those that were life-threatening, and ‘severe’ adverse events were defined as those requiring surgical or radiological interventions or mechanical ventilation. “Moderate’ or ‘mild’ adverse events were defined according to the severity of individual types of adverse events.", "The biopsy specimen was fixed in 10% neutral buffered formalin, embedded in paraffin, and stained with hematoxylin and eosin. Additional staining and immunohistochemistry were performed when a more accurate evaluation was required. Each TBLC specimen was evaluated by a pulmonary pathology expert and classified according to the included anatomical structures (large airway, bronchioles, alveoli, perilobular area, and pleura).\nBased on the fact that the bronchi divide into membranous bronchioles of 2 mm diameter without bronchial glands or bronchial cartilage at the seventh branch [16], we defined LAS as biopsy specimens in which a bronchial gland or bronchial cartilage was present. The biopsy specimens were evaluated in terms of quality and confidence level of the pathological diagnosis. We defined an ideal specimen as a specimen that includes all of bronchioles, alveoli, and perilobular area. The confidence level was classified into three groups [17]. Level A specimens could be used to establish a definite pathological diagnosis; level B specimens could be used to suggest a probable diagnosis; and level C specimens showed only certain findings that were difficult to diagnose by themselves. When a definitive diagnosis such as hypersensitivity pneumonitis, sarcoidosis, or malignancy could not be made and the disease was considered idiopathic, the ATS/ERS/JRS/ALAT guideline 2018 was applied for histological diagnosis [13]. The clinical information, HRCT findings, and pathological diagnosis were then reviewed in a multidisciplinary discussion to make the final institutional diagnosis.", "We identified the anatomical structures in all TBLC specimens and classified them into specimens with or without LAS and compared patient characteristics and specimen characteristics. Patient characteristics included sex, age, smoking history, fibrosis biomarkers, pulmonary function tests, HRCT findings, HRCT pattern, histological diagnostic yield, complications, and the number of specimens collected. Specimen characteristics included maximum diameter, biopsy lobe, structures observed in the specimen, ideal specimen ratio, pathological confidence, HRCT findings in the biopsied lung segment, and diaphragm overlap sign. In this study, we have set a new HRCT sign called diaphragm overlap sign, which examines whether the biopsy site has overlap with the diaphragm in the anterior–posterior direction on HRCT (shown in Fig. 1). When the diaphragm overlaps the lung at the biopsy site during a TBLC procedure, the position of the cryoprobe tip in the lung and its distance from the pleura are more likely to be indistinct, increasing the likelihood of obtaining a centrally sided specimen.\nFig. 1Chest HRCT findings of diaphragm overlap sign. a Case positive for diaphragm overlap sign. Right B10 b + c branched into a subsegmental bronchus (arrows); the peripheral lesion included reticular shadows and ground-glass opacities with traction bronchiectasis (arrowheads). In the anterior–posterior direction, this S10b + c area and the diaphragm completely overlapped (inside of the green area) and were judged to be positive for diaphragm overlap sign. b The S10b + c lesion is located at the red line in the CT scout view, and the coronal section shows that it is completely subdiaphragmatic in height. c Case negative for diaphragm overlap sign. Right B8 branched into a subsegmental bronchus, and then B8a proceeded laterally (arrows); the peripheral lesion of B8b included a consolidation (arrowheads). In the anterior–posterior direction, S8b did not overlap with the diaphragm (outside of the green area) and was judged to be negative for diaphragm overlap sign. d The S8b lesion is located at the red line in the CT scout view, and the coronal section shows that it to be at a height above the diaphragm\nChest HRCT findings of diaphragm overlap sign. a Case positive for diaphragm overlap sign. Right B10 b + c branched into a subsegmental bronchus (arrows); the peripheral lesion included reticular shadows and ground-glass opacities with traction bronchiectasis (arrowheads). In the anterior–posterior direction, this S10b + c area and the diaphragm completely overlapped (inside of the green area) and were judged to be positive for diaphragm overlap sign. b The S10b + c lesion is located at the red line in the CT scout view, and the coronal section shows that it is completely subdiaphragmatic in height. c Case negative for diaphragm overlap sign. Right B8 branched into a subsegmental bronchus, and then B8a proceeded laterally (arrows); the peripheral lesion of B8b included a consolidation (arrowheads). In the anterior–posterior direction, S8b did not overlap with the diaphragm (outside of the green area) and was judged to be negative for diaphragm overlap sign. d The S8b lesion is located at the red line in the CT scout view, and the coronal section shows that it to be at a height above the diaphragm", "Categorical variables are expressed as frequencies and percentages, and continuous variables are expressed as means and standard deviations, as appropriate. Categorical variables were analyzed using the chi-squared test, Fisher’s exact test, and Cochran-Armitage trend test. Continuous variables were analyzed using Mann–Whitney U test and one-way analysis of variance (ANOVA) with post hoc Tukey’s tests. For all statistical tests, a P value < 0.05 was defined as significant. Statistical analysis was performed using EZR version 1.42 (CRAN: the Comprehensive R Archive Network at http://cran.r-project.org/).", "This study included 90 consecutive patients evaluated between February 2018 and January 2020. We excluded 16 patients: 9 were missing pulmonary function tests within 3 months of TBLC, 3 were biopsied for peripheral lung lesions using EBUS, 2 were biopsied for central-type lung cancer, and 2 had an inappropriate pathological evaluation. In total, 74 patients were available in this study, and their baseline characteristics are as indicated in Table 1 for the three groups: LAS-positive, LAS-positive and LAS-negative, LAS-negative. There were 50 LAS-positive cases (67.5%), of which all specimens were LAS in 27 cases and only some specimens were LAS in 23 cases. HRCT pattern accounted for approximately 90% of indeterminate for UIP and alternative diagnoses. All cases with HRCT pattern UIP were positive for some collagen-related autoantibodies or myeloperoxidase-antineutrophil cytoplasmic antibody.\nTable 1Baseline characteristics of patientsCharacteristicsAll patientsLAS-positiveLAS-positiveandLAS-negativeLAS-negative\nP valuePatients, n7427 (36.4)23 (31.0)24 (32.4)Male, n (%)40 (54.0)14 (51.8)15 (65.2)11 (45.8)0.698Age, years67.0 ± 11.569.1 ± 7.167.6 ± 8.664.0 ± 16.70.278Current or ex-smokers, n (%)44 (59.4)16 (59.2)17 (73.9)11 (45.8)0.358KL-6, U/mL1565.1 ± 1523.91473 ± 10511784 ± 20481455 ± 14240.713SP-D, ng/mL346.4 ± 293.0339.7 ± 279.9372.1 ± 339.5327.8 ± 267.40.873PFT %FVC81.8 ± 20.278.9 ± 18.480.1 ± 18.086.8 ± 23.80.347 FEV1/FVC ratio79.9 ± 10.280.4 ± 11.981.5 ± 8.977.9 ± 9.30.483 %DLCO64.1 ± 24.568.0 ± 27.263.9 ± 23.160.3 ± 23.30.584HRCT findings, n (%) Honeycomb19 (25.6)7 (25.9)5 (18.5)7 (29.1)0.805 Traction bronchiectasis50 (67.5)22 (81.4)15 (65.2)13 (54.1)\n< 0.05\n Reticulation63 (85.1)25 (92.5)20 (86.9)18 (75.0)0.07 GGO55 (74.3)19 (70.3)16 (69.5)20 (83.3)0.3 Consolidation17 (22.9)5 (18.5)6 (26.0)6 (25.0)0.573 Nodule17 (22.9)7 (25.9)4 (17.3)6 (25.0)0.917 Cyst12 (16.2)4 (14.8)4 (17.3)4 (16.6)0.853HRCT pattern, n (%) UIP5 (6.7)1 (3.7)3 (13.0)1 (4.1)0.91 Probable UIP4 (5.4)2 (7.4)1 (4.3)1 (4.1)0.603 Indeterminate for UIP27 (36.4)14 (51.8)7 (30.4)6 (25.0)\n< 0.05\n Alternative diagnosis38 (51.3)10 (37.0)12 (52.1)16 (66.6)\n< 0.05\nHistological diagnosis, n (%)47 (63.5)11 (40.7)14 (60.8)22 (91.6)\n< 0.05\nNumber of specimens, n2.3 ± 0.72.1 ± 0.82.6 ± 0.52.4 ± 0.80.06Bronchial bleeding48 (64.8)15 (55.5)20 (86.9)12 (50.0)0.75 Mild27 (36.4)6 (22.2)13 (56.5)8 (33.3)0.367 Moderate21 (28.3)9 (33.3)7 (30.4)4 (16.6)0.186Pneumothorax5 (6.7)0 (0)0 (0)5 (20.8)\n< 0.05\n Mild3 (4.0)0 (0)0 (0)3 (12.5)\n< 0.05\n Moderate1 (1.3)0 (0)0 (0)1 (4.1)0.206 Severe1 (1.3)0 (0)0 (0)1 (4.1)0.206Highlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant differenceDLCO, diffusing capacity for carbon monoxide; FEV1, forced expiratory volume in 1 s; FVC, forced volume capacity; GGO, ground-glass opacities; HRCT, high-resolution computed tomography; KL-6, Krebs von den Lungen-6; LAS, large airway specimen; PFT, pulmonary function test; SP-D, surfactant protein D; UIP, usual interstitial pneumonia\nBaseline characteristics of patients\nHighlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant difference\nDLCO, diffusing capacity for carbon monoxide; FEV1, forced expiratory volume in 1 s; FVC, forced volume capacity; GGO, ground-glass opacities; HRCT, high-resolution computed tomography; KL-6, Krebs von den Lungen-6; LAS, large airway specimen; PFT, pulmonary function test; SP-D, surfactant protein D; UIP, usual interstitial pneumonia", "A specific histological diagnosis was obtained in 47/74 cases (63.5%). The histological diagnostic yield tended to be statistically significantly higher in the LAS-negative group (40.7% in LAS positive group; 60.8% in LAS positive and negative group; 91.6% in LAS negative group) (Cochran-Armitage trend test). The histological interpretations are shown in Table 2 and include 11 (14.8%) UIP, 6 (8.1) probable UIP, 16 (21.6%) indeterminate for UIP, 9 (12.1%) non-specific interstitial pneumonia, 6 (8.1%) organizing pneumonia and 6 (8.1%) hypersensitivity pneumonitis. The institutional diagnosis after MDD is shown in Table 3 and include 9 (12.1%) idiopathic pulmonary fibrosis, 6 (8.1%) non-specific interstitial pneumonia, 4 (5.4%) organizing pneumonia, 24 (32.4%) unclassifiable and 9 (12.1%) hypersensitivity pneumonitis.\nTable 2Histological diagnosisHistological diagnosisNo (%)UIP11 (14.8)Probable UIP6 (8.1)Indeterminate for UIP16 (21.6)– 14 NSIP/UIP– 1 NSIP/UIP/OP– 1 UIP/OPAlternative diagnosis NSIP9 (12.1) OP6 (8.1) HP6 (8.1) Smoking-related IP2 (2.7) Others7 (9.4)– 4 IgG4-related disease– 1 sarcoidosis– 1 cellular bronchiolitis– 1 malignancyN.D.11 (14.8)DPB, diffuse panbronchiolitis; HP, hypersensitivity pneumonitis; N.D., non-diagnostic; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; UIP, usual interstitial pneumoniaTable 3MDD diagnosisMDD diagnosisNo (%)IPF9 (12.1)NSIP6 (8.1)OP4 (5.4)Unclassifiable24 (32.4)Hypersensitivity pneumonitis9 (12.1)CTD-ILD (MCTD/RA/SjS/SLE/SSc)10 (13.5)MPA3 (4.0)Others9 (12.1)CTD, connective tissue disease; IPF, idiopathic pulmonary fibrosis; MCTD, mixed connective tissue disease; MPA, microscopic polyangiitis; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; RA, rheumatoid arthritis; SjS, Sjögren’s syndrome; SLE, systemic lupus erythematosus; SSc, systemic sclerosis\nHistological diagnosis\nDPB, diffuse panbronchiolitis; HP, hypersensitivity pneumonitis; N.D., non-diagnostic; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; UIP, usual interstitial pneumonia\nMDD diagnosis\nCTD, connective tissue disease; IPF, idiopathic pulmonary fibrosis; MCTD, mixed connective tissue disease; MPA, microscopic polyangiitis; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; RA, rheumatoid arthritis; SjS, Sjögren’s syndrome; SLE, systemic lupus erythematosus; SSc, systemic sclerosis", "In a comparison between the three groups, the LAS-negative group tended to have more pneumothorax complications than the other groups (0% in LAS positive group; 0% in LAS positive and negative group; 20.8% in LAS negative group) (Cochran-Armitage trend test). There was no clear difference in bronchial bleeding between the three groups (Table 1).", "In total, 177 specimens were collected in the 74 cases, with a mean number of specimens per case of 2.3 ± 0.7 (Tables 1 and 4). 175 were collected with 1.9 mm probes and only 2 were collected with 2.4 mm probes. The most common biopsy lobe was the right lower lobe (63.8%), followed by the right upper lobe (15.2%) and the left lower lobe (14.1%). There were 85 (48.0%) LAS containing bronchial gland or bronchial cartilage: 30 containing both bronchial gland and bronchial cartilage, 50 containing only bronchial cartilage, and 5 containing only bronchial gland. The anatomical structures observed except for the large airway were alveoli (91.5%), bronchioles (49.1%), and perilobular area (43.5%), with few pleural specimens (1.2%). Ideal specimens were obtained in 47 of the 177 (26.5%) specimens, about a quarter of the total. A comparison of specimen characteristics showed that the proportion of bronchioles, alveoli, and perilobular area were significantly lower in the LAS-positive group than in the LAS-negative group (Table 3). Similarly, the ideal specimen ratio and the frequency of specimens of high pathological confidence were significantly lower in the LAS-positive group. The ratio of specimens with a pathological confidence level A was 25 of the 47 (53.1%) for ideal specimens and 43 of the 130 (33.0%) for unideal specimens (P < 0.05). In terms of the HRCT findings at the sample collection site, the LAS-positive group had a significantly higher proportion of traction bronchiectasis (65.5% vs. 29.6%) and diaphragm overlap sign (54.1% vs. 33.6%) than the LAS-negative group.\nTable 4Characteristics of specimens obtained by TBLCCharacteristicsAll specimensLAS positiveLAS negative\nP valueSpecimens, n (%)17785 (48.0)92 (51.9)Maximum diameter, mm6.4 ± 2.97.5 ± 3.75.3 ± 1.3\n< 0.05\nBiopsy lobe, n (%) Right upper lobe27 (15.2)7 (8.2)20 (21.7)\n< 0.05\n Right middle lobe6 (3.3)2 (2.3)4 (4.3)0.684 Right lower lobe113 (63.8)58 (68.2)55 (59.7)0.275 Left upper lobe6 (3.3)2 (2.3)4 (4.2)0.684 Left lower lobe25 (14,1)16 (18.8)9 (9.7)0.129Structures observed on the specimen Bronchiole87 (49.1)23 (27.0)64 (69.5)\n< 0.05\n Alveoli162 (91.5)73 (85.8)89 (96.7)\n< 0.05\n Perilobular area77 (43.5)15 (17.6)62 (67.3)\n< 0.05\n Pleura1 (1.2)0 (0)1 (1.0)1Ideal specimen, n (%)47 (26.5)5 (5.8)42 (45.6)\n< 0.05\nPathological confidence level Level A68 (38.4)23 (27.0)45 (48.9)\n< 0.05\n Level B84 (47.4)48 (56.4)36 (39.1)\n< 0.05\n Level C10 (5.6)5 (5.8)5 (5.4)1 Not evaluated15 (8.4)9 (10.5)6 (6.5)HRCT findings in the biopsied lung segment, n (%) Honeycomb8 (4.5)4 (4.7)4 (4.3)1 Traction bronchiectasis101 (57.0)65 (76.4)36 (39.1)\n< 0.05\n Reticulation130 (73.4)68 (80.0)62 (67.3)0.063 GGO60 (33.8)28 (32.9)32 (34.7)0.874 Consolidation16 (9.0)8 (9.4)8 (8.6)1 Nodule14 (7.9)4 (4.7)10 (10.8)0.167 Cyst5 (2.8)0 (0)1 (1.0)0.059Diaphragm overlap sign, n (%)77 (43.5)46 (54.1)31 (33.6)\n< 0.05\nHighlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant differenceGGO, ground-glass opacities; HRCT, high-resolution computed tomography; LAS, large airway specimen\nCharacteristics of specimens obtained by TBLC\nHighlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant difference\nGGO, ground-glass opacities; HRCT, high-resolution computed tomography; LAS, large airway specimen" ]
[ null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Materials and methods", "Patients", "Data collection", "Transbronchial cryobiopsy procedure", "Pathological evaluation", "Study design", "Statistical analysis", "Results", "Patients", "Diagnostic yield and histological diagnosis", "Complications", "Specimen characteristics", "Discussion", "Conclusion" ]
[ "Diffuse parenchymal lung diseases (DPLD) comprise heterogeneous disorders with different prognostic and therapeutic implications. Many international guidelines have required confirmation of the pathological findings by surgical lung biopsy (SLB) to recognize histological patterns and provide an accurate diagnosis by obtaining a sufficient volume of specimens [1, 2]. SLB has a histological diagnostic yield of more than 90%, but it has disadvantages of certain morbidity and mortality and the presence of patients excluded from the indication due to advanced age, complications including pulmonary hypertension, and severe respiratory failure [3, 4]. Transbronchial lung cryobiopsy (TBLC) is a promising new bronchoscopic biopsy technique with lower mortality and fewer complications, but the diagnostic yield for DPLD is 70–80%, which is inferior to that of SLB [5]. This difference in diagnostic yield between SLB and TBLC has been reported to be due to the rate of interpathologist agreement, specimen size, and specimen adequacy [6–9].\nAs mentioned in previous TBLC reports for DPLD [10, 11], specimens containing large airway are considered unideal with little contribution to the histological diagnosis. However, the specific conditions under which these specimens are likely to be collected and their actual impact on diagnostic rates are not known. The purpose of this study was to examine the characteristics of TBLC specimens containing large airway and their impact on the rate of histological diagnosis in individual cases.", "[SUBTITLE] Patients [SUBSECTION] This retrospective study was conducted in the Department of Respiratory Medicine of Saitama Red Cross Hospital between February 2018 and January 2020 and was approved from the institutional review board of Saitama Red Cross Hospital (approval number 19-C). The need for patient approval and/or informed consent was waived due to the retrospective nature of the study. The inclusion criteria were age > 20 years and patients with DPLD who underwent TBLC. Exclusion criteria were as follows: patients who underwent biopsy from a site directly visible by bronchoscopy, patients for whom biopsy was performed after confirmation by endobronchial ultrasonography (EBUS) at a peripheral site, patients who met the pulmonary function test criteria for relative contraindications to TBLC as indicated in a previous report [12]; %forced vital capacity < 50% and %diffusion capacity of the lung for carbon monoxide < 35%, and patients for whom pulmonary function test, high-resolution computed tomography (HRCT), or laboratory findings were missing within 3 months of TBLC.\nThis retrospective study was conducted in the Department of Respiratory Medicine of Saitama Red Cross Hospital between February 2018 and January 2020 and was approved from the institutional review board of Saitama Red Cross Hospital (approval number 19-C). The need for patient approval and/or informed consent was waived due to the retrospective nature of the study. The inclusion criteria were age > 20 years and patients with DPLD who underwent TBLC. Exclusion criteria were as follows: patients who underwent biopsy from a site directly visible by bronchoscopy, patients for whom biopsy was performed after confirmation by endobronchial ultrasonography (EBUS) at a peripheral site, patients who met the pulmonary function test criteria for relative contraindications to TBLC as indicated in a previous report [12]; %forced vital capacity < 50% and %diffusion capacity of the lung for carbon monoxide < 35%, and patients for whom pulmonary function test, high-resolution computed tomography (HRCT), or laboratory findings were missing within 3 months of TBLC.\n[SUBTITLE] Data collection [SUBSECTION] Baseline clinical measurements were obtained within 3 months of the bronchoscopy. Each patient’s HRCT scan was reviewed in a multi-disciplinary discussion and classified as presenting an HRCT pattern of usual interstitial pneumonia (UIP), probable UIP, indeterminate for UIP, or alternative diagnosis according to the American Thoracic Society (ATS)/European Respiratory Society (ERS)/Japanese Respiratory Society (JRS)/Latin American Thoracic Association (ALAT) guideline 2018 [13]. Definitions of terms related to the HRCT findings were based on the definitions of the Fleischer Society [14].\nBaseline clinical measurements were obtained within 3 months of the bronchoscopy. Each patient’s HRCT scan was reviewed in a multi-disciplinary discussion and classified as presenting an HRCT pattern of usual interstitial pneumonia (UIP), probable UIP, indeterminate for UIP, or alternative diagnosis according to the American Thoracic Society (ATS)/European Respiratory Society (ERS)/Japanese Respiratory Society (JRS)/Latin American Thoracic Association (ALAT) guideline 2018 [13]. Definitions of terms related to the HRCT findings were based on the definitions of the Fleischer Society [14].\n[SUBTITLE] Transbronchial cryobiopsy procedure [SUBSECTION] Bronchoscopy was performed with a BF-1T290 flexible bronchoscope (Olympus, Tokyo, Japan). Patients were anesthetized with midazolam and fentanyl intravenously, and 2% lidocaine was added intratracheally as appropriate. The endotracheal tube (SACETT Suction Above Cuff Endotracheal Tube 8.0 mm; Smiths Medical International Ltd., Minneapolis, MN, USA) was inserted for airway control. An endobronchial balloon catheter (Fogarty catheter, E-080-4F; Edwards Life-sciences, Irvine, CA, USA) was routinely used for bronchial blockade. The flexible reusable cryoprobe, either 1.9 mm or 2.4 mm in diameter (ERBECRYO® 2 system; Erbe Elektromedizin GmbH, Tübingen, Germany) was inserted from a peripheral bronchus to just below the pleura under fluoroscopy. The probe was withdrawn for 1 cm proximally to prevent pneumothorax. After freezing the probe for 5–7 s, the bronchoscope was withdrawn along with the biopsy specimen and probe, and the endobronchial balloon catheter was inflated simultaneously. In accordance with previous reports [15], Complications were classified into four levels. “Serious” adverse events were defined as those that were life-threatening, and ‘severe’ adverse events were defined as those requiring surgical or radiological interventions or mechanical ventilation. “Moderate’ or ‘mild’ adverse events were defined according to the severity of individual types of adverse events.\nBronchoscopy was performed with a BF-1T290 flexible bronchoscope (Olympus, Tokyo, Japan). Patients were anesthetized with midazolam and fentanyl intravenously, and 2% lidocaine was added intratracheally as appropriate. The endotracheal tube (SACETT Suction Above Cuff Endotracheal Tube 8.0 mm; Smiths Medical International Ltd., Minneapolis, MN, USA) was inserted for airway control. An endobronchial balloon catheter (Fogarty catheter, E-080-4F; Edwards Life-sciences, Irvine, CA, USA) was routinely used for bronchial blockade. The flexible reusable cryoprobe, either 1.9 mm or 2.4 mm in diameter (ERBECRYO® 2 system; Erbe Elektromedizin GmbH, Tübingen, Germany) was inserted from a peripheral bronchus to just below the pleura under fluoroscopy. The probe was withdrawn for 1 cm proximally to prevent pneumothorax. After freezing the probe for 5–7 s, the bronchoscope was withdrawn along with the biopsy specimen and probe, and the endobronchial balloon catheter was inflated simultaneously. In accordance with previous reports [15], Complications were classified into four levels. “Serious” adverse events were defined as those that were life-threatening, and ‘severe’ adverse events were defined as those requiring surgical or radiological interventions or mechanical ventilation. “Moderate’ or ‘mild’ adverse events were defined according to the severity of individual types of adverse events.\n[SUBTITLE] Pathological evaluation [SUBSECTION] The biopsy specimen was fixed in 10% neutral buffered formalin, embedded in paraffin, and stained with hematoxylin and eosin. Additional staining and immunohistochemistry were performed when a more accurate evaluation was required. Each TBLC specimen was evaluated by a pulmonary pathology expert and classified according to the included anatomical structures (large airway, bronchioles, alveoli, perilobular area, and pleura).\nBased on the fact that the bronchi divide into membranous bronchioles of 2 mm diameter without bronchial glands or bronchial cartilage at the seventh branch [16], we defined LAS as biopsy specimens in which a bronchial gland or bronchial cartilage was present. The biopsy specimens were evaluated in terms of quality and confidence level of the pathological diagnosis. We defined an ideal specimen as a specimen that includes all of bronchioles, alveoli, and perilobular area. The confidence level was classified into three groups [17]. Level A specimens could be used to establish a definite pathological diagnosis; level B specimens could be used to suggest a probable diagnosis; and level C specimens showed only certain findings that were difficult to diagnose by themselves. When a definitive diagnosis such as hypersensitivity pneumonitis, sarcoidosis, or malignancy could not be made and the disease was considered idiopathic, the ATS/ERS/JRS/ALAT guideline 2018 was applied for histological diagnosis [13]. The clinical information, HRCT findings, and pathological diagnosis were then reviewed in a multidisciplinary discussion to make the final institutional diagnosis.\nThe biopsy specimen was fixed in 10% neutral buffered formalin, embedded in paraffin, and stained with hematoxylin and eosin. Additional staining and immunohistochemistry were performed when a more accurate evaluation was required. Each TBLC specimen was evaluated by a pulmonary pathology expert and classified according to the included anatomical structures (large airway, bronchioles, alveoli, perilobular area, and pleura).\nBased on the fact that the bronchi divide into membranous bronchioles of 2 mm diameter without bronchial glands or bronchial cartilage at the seventh branch [16], we defined LAS as biopsy specimens in which a bronchial gland or bronchial cartilage was present. The biopsy specimens were evaluated in terms of quality and confidence level of the pathological diagnosis. We defined an ideal specimen as a specimen that includes all of bronchioles, alveoli, and perilobular area. The confidence level was classified into three groups [17]. Level A specimens could be used to establish a definite pathological diagnosis; level B specimens could be used to suggest a probable diagnosis; and level C specimens showed only certain findings that were difficult to diagnose by themselves. When a definitive diagnosis such as hypersensitivity pneumonitis, sarcoidosis, or malignancy could not be made and the disease was considered idiopathic, the ATS/ERS/JRS/ALAT guideline 2018 was applied for histological diagnosis [13]. The clinical information, HRCT findings, and pathological diagnosis were then reviewed in a multidisciplinary discussion to make the final institutional diagnosis.\n[SUBTITLE] Study design [SUBSECTION] We identified the anatomical structures in all TBLC specimens and classified them into specimens with or without LAS and compared patient characteristics and specimen characteristics. Patient characteristics included sex, age, smoking history, fibrosis biomarkers, pulmonary function tests, HRCT findings, HRCT pattern, histological diagnostic yield, complications, and the number of specimens collected. Specimen characteristics included maximum diameter, biopsy lobe, structures observed in the specimen, ideal specimen ratio, pathological confidence, HRCT findings in the biopsied lung segment, and diaphragm overlap sign. In this study, we have set a new HRCT sign called diaphragm overlap sign, which examines whether the biopsy site has overlap with the diaphragm in the anterior–posterior direction on HRCT (shown in Fig. 1). When the diaphragm overlaps the lung at the biopsy site during a TBLC procedure, the position of the cryoprobe tip in the lung and its distance from the pleura are more likely to be indistinct, increasing the likelihood of obtaining a centrally sided specimen.\nFig. 1Chest HRCT findings of diaphragm overlap sign. a Case positive for diaphragm overlap sign. Right B10 b + c branched into a subsegmental bronchus (arrows); the peripheral lesion included reticular shadows and ground-glass opacities with traction bronchiectasis (arrowheads). In the anterior–posterior direction, this S10b + c area and the diaphragm completely overlapped (inside of the green area) and were judged to be positive for diaphragm overlap sign. b The S10b + c lesion is located at the red line in the CT scout view, and the coronal section shows that it is completely subdiaphragmatic in height. c Case negative for diaphragm overlap sign. Right B8 branched into a subsegmental bronchus, and then B8a proceeded laterally (arrows); the peripheral lesion of B8b included a consolidation (arrowheads). In the anterior–posterior direction, S8b did not overlap with the diaphragm (outside of the green area) and was judged to be negative for diaphragm overlap sign. d The S8b lesion is located at the red line in the CT scout view, and the coronal section shows that it to be at a height above the diaphragm\nChest HRCT findings of diaphragm overlap sign. a Case positive for diaphragm overlap sign. Right B10 b + c branched into a subsegmental bronchus (arrows); the peripheral lesion included reticular shadows and ground-glass opacities with traction bronchiectasis (arrowheads). In the anterior–posterior direction, this S10b + c area and the diaphragm completely overlapped (inside of the green area) and were judged to be positive for diaphragm overlap sign. b The S10b + c lesion is located at the red line in the CT scout view, and the coronal section shows that it is completely subdiaphragmatic in height. c Case negative for diaphragm overlap sign. Right B8 branched into a subsegmental bronchus, and then B8a proceeded laterally (arrows); the peripheral lesion of B8b included a consolidation (arrowheads). In the anterior–posterior direction, S8b did not overlap with the diaphragm (outside of the green area) and was judged to be negative for diaphragm overlap sign. d The S8b lesion is located at the red line in the CT scout view, and the coronal section shows that it to be at a height above the diaphragm\nWe identified the anatomical structures in all TBLC specimens and classified them into specimens with or without LAS and compared patient characteristics and specimen characteristics. Patient characteristics included sex, age, smoking history, fibrosis biomarkers, pulmonary function tests, HRCT findings, HRCT pattern, histological diagnostic yield, complications, and the number of specimens collected. Specimen characteristics included maximum diameter, biopsy lobe, structures observed in the specimen, ideal specimen ratio, pathological confidence, HRCT findings in the biopsied lung segment, and diaphragm overlap sign. In this study, we have set a new HRCT sign called diaphragm overlap sign, which examines whether the biopsy site has overlap with the diaphragm in the anterior–posterior direction on HRCT (shown in Fig. 1). When the diaphragm overlaps the lung at the biopsy site during a TBLC procedure, the position of the cryoprobe tip in the lung and its distance from the pleura are more likely to be indistinct, increasing the likelihood of obtaining a centrally sided specimen.\nFig. 1Chest HRCT findings of diaphragm overlap sign. a Case positive for diaphragm overlap sign. Right B10 b + c branched into a subsegmental bronchus (arrows); the peripheral lesion included reticular shadows and ground-glass opacities with traction bronchiectasis (arrowheads). In the anterior–posterior direction, this S10b + c area and the diaphragm completely overlapped (inside of the green area) and were judged to be positive for diaphragm overlap sign. b The S10b + c lesion is located at the red line in the CT scout view, and the coronal section shows that it is completely subdiaphragmatic in height. c Case negative for diaphragm overlap sign. Right B8 branched into a subsegmental bronchus, and then B8a proceeded laterally (arrows); the peripheral lesion of B8b included a consolidation (arrowheads). In the anterior–posterior direction, S8b did not overlap with the diaphragm (outside of the green area) and was judged to be negative for diaphragm overlap sign. d The S8b lesion is located at the red line in the CT scout view, and the coronal section shows that it to be at a height above the diaphragm\nChest HRCT findings of diaphragm overlap sign. a Case positive for diaphragm overlap sign. Right B10 b + c branched into a subsegmental bronchus (arrows); the peripheral lesion included reticular shadows and ground-glass opacities with traction bronchiectasis (arrowheads). In the anterior–posterior direction, this S10b + c area and the diaphragm completely overlapped (inside of the green area) and were judged to be positive for diaphragm overlap sign. b The S10b + c lesion is located at the red line in the CT scout view, and the coronal section shows that it is completely subdiaphragmatic in height. c Case negative for diaphragm overlap sign. Right B8 branched into a subsegmental bronchus, and then B8a proceeded laterally (arrows); the peripheral lesion of B8b included a consolidation (arrowheads). In the anterior–posterior direction, S8b did not overlap with the diaphragm (outside of the green area) and was judged to be negative for diaphragm overlap sign. d The S8b lesion is located at the red line in the CT scout view, and the coronal section shows that it to be at a height above the diaphragm\n[SUBTITLE] Statistical analysis [SUBSECTION] Categorical variables are expressed as frequencies and percentages, and continuous variables are expressed as means and standard deviations, as appropriate. Categorical variables were analyzed using the chi-squared test, Fisher’s exact test, and Cochran-Armitage trend test. Continuous variables were analyzed using Mann–Whitney U test and one-way analysis of variance (ANOVA) with post hoc Tukey’s tests. For all statistical tests, a P value < 0.05 was defined as significant. Statistical analysis was performed using EZR version 1.42 (CRAN: the Comprehensive R Archive Network at http://cran.r-project.org/).\nCategorical variables are expressed as frequencies and percentages, and continuous variables are expressed as means and standard deviations, as appropriate. Categorical variables were analyzed using the chi-squared test, Fisher’s exact test, and Cochran-Armitage trend test. Continuous variables were analyzed using Mann–Whitney U test and one-way analysis of variance (ANOVA) with post hoc Tukey’s tests. For all statistical tests, a P value < 0.05 was defined as significant. Statistical analysis was performed using EZR version 1.42 (CRAN: the Comprehensive R Archive Network at http://cran.r-project.org/).", "This retrospective study was conducted in the Department of Respiratory Medicine of Saitama Red Cross Hospital between February 2018 and January 2020 and was approved from the institutional review board of Saitama Red Cross Hospital (approval number 19-C). The need for patient approval and/or informed consent was waived due to the retrospective nature of the study. The inclusion criteria were age > 20 years and patients with DPLD who underwent TBLC. Exclusion criteria were as follows: patients who underwent biopsy from a site directly visible by bronchoscopy, patients for whom biopsy was performed after confirmation by endobronchial ultrasonography (EBUS) at a peripheral site, patients who met the pulmonary function test criteria for relative contraindications to TBLC as indicated in a previous report [12]; %forced vital capacity < 50% and %diffusion capacity of the lung for carbon monoxide < 35%, and patients for whom pulmonary function test, high-resolution computed tomography (HRCT), or laboratory findings were missing within 3 months of TBLC.", "Baseline clinical measurements were obtained within 3 months of the bronchoscopy. Each patient’s HRCT scan was reviewed in a multi-disciplinary discussion and classified as presenting an HRCT pattern of usual interstitial pneumonia (UIP), probable UIP, indeterminate for UIP, or alternative diagnosis according to the American Thoracic Society (ATS)/European Respiratory Society (ERS)/Japanese Respiratory Society (JRS)/Latin American Thoracic Association (ALAT) guideline 2018 [13]. Definitions of terms related to the HRCT findings were based on the definitions of the Fleischer Society [14].", "Bronchoscopy was performed with a BF-1T290 flexible bronchoscope (Olympus, Tokyo, Japan). Patients were anesthetized with midazolam and fentanyl intravenously, and 2% lidocaine was added intratracheally as appropriate. The endotracheal tube (SACETT Suction Above Cuff Endotracheal Tube 8.0 mm; Smiths Medical International Ltd., Minneapolis, MN, USA) was inserted for airway control. An endobronchial balloon catheter (Fogarty catheter, E-080-4F; Edwards Life-sciences, Irvine, CA, USA) was routinely used for bronchial blockade. The flexible reusable cryoprobe, either 1.9 mm or 2.4 mm in diameter (ERBECRYO® 2 system; Erbe Elektromedizin GmbH, Tübingen, Germany) was inserted from a peripheral bronchus to just below the pleura under fluoroscopy. The probe was withdrawn for 1 cm proximally to prevent pneumothorax. After freezing the probe for 5–7 s, the bronchoscope was withdrawn along with the biopsy specimen and probe, and the endobronchial balloon catheter was inflated simultaneously. In accordance with previous reports [15], Complications were classified into four levels. “Serious” adverse events were defined as those that were life-threatening, and ‘severe’ adverse events were defined as those requiring surgical or radiological interventions or mechanical ventilation. “Moderate’ or ‘mild’ adverse events were defined according to the severity of individual types of adverse events.", "The biopsy specimen was fixed in 10% neutral buffered formalin, embedded in paraffin, and stained with hematoxylin and eosin. Additional staining and immunohistochemistry were performed when a more accurate evaluation was required. Each TBLC specimen was evaluated by a pulmonary pathology expert and classified according to the included anatomical structures (large airway, bronchioles, alveoli, perilobular area, and pleura).\nBased on the fact that the bronchi divide into membranous bronchioles of 2 mm diameter without bronchial glands or bronchial cartilage at the seventh branch [16], we defined LAS as biopsy specimens in which a bronchial gland or bronchial cartilage was present. The biopsy specimens were evaluated in terms of quality and confidence level of the pathological diagnosis. We defined an ideal specimen as a specimen that includes all of bronchioles, alveoli, and perilobular area. The confidence level was classified into three groups [17]. Level A specimens could be used to establish a definite pathological diagnosis; level B specimens could be used to suggest a probable diagnosis; and level C specimens showed only certain findings that were difficult to diagnose by themselves. When a definitive diagnosis such as hypersensitivity pneumonitis, sarcoidosis, or malignancy could not be made and the disease was considered idiopathic, the ATS/ERS/JRS/ALAT guideline 2018 was applied for histological diagnosis [13]. The clinical information, HRCT findings, and pathological diagnosis were then reviewed in a multidisciplinary discussion to make the final institutional diagnosis.", "We identified the anatomical structures in all TBLC specimens and classified them into specimens with or without LAS and compared patient characteristics and specimen characteristics. Patient characteristics included sex, age, smoking history, fibrosis biomarkers, pulmonary function tests, HRCT findings, HRCT pattern, histological diagnostic yield, complications, and the number of specimens collected. Specimen characteristics included maximum diameter, biopsy lobe, structures observed in the specimen, ideal specimen ratio, pathological confidence, HRCT findings in the biopsied lung segment, and diaphragm overlap sign. In this study, we have set a new HRCT sign called diaphragm overlap sign, which examines whether the biopsy site has overlap with the diaphragm in the anterior–posterior direction on HRCT (shown in Fig. 1). When the diaphragm overlaps the lung at the biopsy site during a TBLC procedure, the position of the cryoprobe tip in the lung and its distance from the pleura are more likely to be indistinct, increasing the likelihood of obtaining a centrally sided specimen.\nFig. 1Chest HRCT findings of diaphragm overlap sign. a Case positive for diaphragm overlap sign. Right B10 b + c branched into a subsegmental bronchus (arrows); the peripheral lesion included reticular shadows and ground-glass opacities with traction bronchiectasis (arrowheads). In the anterior–posterior direction, this S10b + c area and the diaphragm completely overlapped (inside of the green area) and were judged to be positive for diaphragm overlap sign. b The S10b + c lesion is located at the red line in the CT scout view, and the coronal section shows that it is completely subdiaphragmatic in height. c Case negative for diaphragm overlap sign. Right B8 branched into a subsegmental bronchus, and then B8a proceeded laterally (arrows); the peripheral lesion of B8b included a consolidation (arrowheads). In the anterior–posterior direction, S8b did not overlap with the diaphragm (outside of the green area) and was judged to be negative for diaphragm overlap sign. d The S8b lesion is located at the red line in the CT scout view, and the coronal section shows that it to be at a height above the diaphragm\nChest HRCT findings of diaphragm overlap sign. a Case positive for diaphragm overlap sign. Right B10 b + c branched into a subsegmental bronchus (arrows); the peripheral lesion included reticular shadows and ground-glass opacities with traction bronchiectasis (arrowheads). In the anterior–posterior direction, this S10b + c area and the diaphragm completely overlapped (inside of the green area) and were judged to be positive for diaphragm overlap sign. b The S10b + c lesion is located at the red line in the CT scout view, and the coronal section shows that it is completely subdiaphragmatic in height. c Case negative for diaphragm overlap sign. Right B8 branched into a subsegmental bronchus, and then B8a proceeded laterally (arrows); the peripheral lesion of B8b included a consolidation (arrowheads). In the anterior–posterior direction, S8b did not overlap with the diaphragm (outside of the green area) and was judged to be negative for diaphragm overlap sign. d The S8b lesion is located at the red line in the CT scout view, and the coronal section shows that it to be at a height above the diaphragm", "Categorical variables are expressed as frequencies and percentages, and continuous variables are expressed as means and standard deviations, as appropriate. Categorical variables were analyzed using the chi-squared test, Fisher’s exact test, and Cochran-Armitage trend test. Continuous variables were analyzed using Mann–Whitney U test and one-way analysis of variance (ANOVA) with post hoc Tukey’s tests. For all statistical tests, a P value < 0.05 was defined as significant. Statistical analysis was performed using EZR version 1.42 (CRAN: the Comprehensive R Archive Network at http://cran.r-project.org/).", "[SUBTITLE] Patients [SUBSECTION] This study included 90 consecutive patients evaluated between February 2018 and January 2020. We excluded 16 patients: 9 were missing pulmonary function tests within 3 months of TBLC, 3 were biopsied for peripheral lung lesions using EBUS, 2 were biopsied for central-type lung cancer, and 2 had an inappropriate pathological evaluation. In total, 74 patients were available in this study, and their baseline characteristics are as indicated in Table 1 for the three groups: LAS-positive, LAS-positive and LAS-negative, LAS-negative. There were 50 LAS-positive cases (67.5%), of which all specimens were LAS in 27 cases and only some specimens were LAS in 23 cases. HRCT pattern accounted for approximately 90% of indeterminate for UIP and alternative diagnoses. All cases with HRCT pattern UIP were positive for some collagen-related autoantibodies or myeloperoxidase-antineutrophil cytoplasmic antibody.\nTable 1Baseline characteristics of patientsCharacteristicsAll patientsLAS-positiveLAS-positiveandLAS-negativeLAS-negative\nP valuePatients, n7427 (36.4)23 (31.0)24 (32.4)Male, n (%)40 (54.0)14 (51.8)15 (65.2)11 (45.8)0.698Age, years67.0 ± 11.569.1 ± 7.167.6 ± 8.664.0 ± 16.70.278Current or ex-smokers, n (%)44 (59.4)16 (59.2)17 (73.9)11 (45.8)0.358KL-6, U/mL1565.1 ± 1523.91473 ± 10511784 ± 20481455 ± 14240.713SP-D, ng/mL346.4 ± 293.0339.7 ± 279.9372.1 ± 339.5327.8 ± 267.40.873PFT %FVC81.8 ± 20.278.9 ± 18.480.1 ± 18.086.8 ± 23.80.347 FEV1/FVC ratio79.9 ± 10.280.4 ± 11.981.5 ± 8.977.9 ± 9.30.483 %DLCO64.1 ± 24.568.0 ± 27.263.9 ± 23.160.3 ± 23.30.584HRCT findings, n (%) Honeycomb19 (25.6)7 (25.9)5 (18.5)7 (29.1)0.805 Traction bronchiectasis50 (67.5)22 (81.4)15 (65.2)13 (54.1)\n< 0.05\n Reticulation63 (85.1)25 (92.5)20 (86.9)18 (75.0)0.07 GGO55 (74.3)19 (70.3)16 (69.5)20 (83.3)0.3 Consolidation17 (22.9)5 (18.5)6 (26.0)6 (25.0)0.573 Nodule17 (22.9)7 (25.9)4 (17.3)6 (25.0)0.917 Cyst12 (16.2)4 (14.8)4 (17.3)4 (16.6)0.853HRCT pattern, n (%) UIP5 (6.7)1 (3.7)3 (13.0)1 (4.1)0.91 Probable UIP4 (5.4)2 (7.4)1 (4.3)1 (4.1)0.603 Indeterminate for UIP27 (36.4)14 (51.8)7 (30.4)6 (25.0)\n< 0.05\n Alternative diagnosis38 (51.3)10 (37.0)12 (52.1)16 (66.6)\n< 0.05\nHistological diagnosis, n (%)47 (63.5)11 (40.7)14 (60.8)22 (91.6)\n< 0.05\nNumber of specimens, n2.3 ± 0.72.1 ± 0.82.6 ± 0.52.4 ± 0.80.06Bronchial bleeding48 (64.8)15 (55.5)20 (86.9)12 (50.0)0.75 Mild27 (36.4)6 (22.2)13 (56.5)8 (33.3)0.367 Moderate21 (28.3)9 (33.3)7 (30.4)4 (16.6)0.186Pneumothorax5 (6.7)0 (0)0 (0)5 (20.8)\n< 0.05\n Mild3 (4.0)0 (0)0 (0)3 (12.5)\n< 0.05\n Moderate1 (1.3)0 (0)0 (0)1 (4.1)0.206 Severe1 (1.3)0 (0)0 (0)1 (4.1)0.206Highlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant differenceDLCO, diffusing capacity for carbon monoxide; FEV1, forced expiratory volume in 1 s; FVC, forced volume capacity; GGO, ground-glass opacities; HRCT, high-resolution computed tomography; KL-6, Krebs von den Lungen-6; LAS, large airway specimen; PFT, pulmonary function test; SP-D, surfactant protein D; UIP, usual interstitial pneumonia\nBaseline characteristics of patients\nHighlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant difference\nDLCO, diffusing capacity for carbon monoxide; FEV1, forced expiratory volume in 1 s; FVC, forced volume capacity; GGO, ground-glass opacities; HRCT, high-resolution computed tomography; KL-6, Krebs von den Lungen-6; LAS, large airway specimen; PFT, pulmonary function test; SP-D, surfactant protein D; UIP, usual interstitial pneumonia\nThis study included 90 consecutive patients evaluated between February 2018 and January 2020. We excluded 16 patients: 9 were missing pulmonary function tests within 3 months of TBLC, 3 were biopsied for peripheral lung lesions using EBUS, 2 were biopsied for central-type lung cancer, and 2 had an inappropriate pathological evaluation. In total, 74 patients were available in this study, and their baseline characteristics are as indicated in Table 1 for the three groups: LAS-positive, LAS-positive and LAS-negative, LAS-negative. There were 50 LAS-positive cases (67.5%), of which all specimens were LAS in 27 cases and only some specimens were LAS in 23 cases. HRCT pattern accounted for approximately 90% of indeterminate for UIP and alternative diagnoses. All cases with HRCT pattern UIP were positive for some collagen-related autoantibodies or myeloperoxidase-antineutrophil cytoplasmic antibody.\nTable 1Baseline characteristics of patientsCharacteristicsAll patientsLAS-positiveLAS-positiveandLAS-negativeLAS-negative\nP valuePatients, n7427 (36.4)23 (31.0)24 (32.4)Male, n (%)40 (54.0)14 (51.8)15 (65.2)11 (45.8)0.698Age, years67.0 ± 11.569.1 ± 7.167.6 ± 8.664.0 ± 16.70.278Current or ex-smokers, n (%)44 (59.4)16 (59.2)17 (73.9)11 (45.8)0.358KL-6, U/mL1565.1 ± 1523.91473 ± 10511784 ± 20481455 ± 14240.713SP-D, ng/mL346.4 ± 293.0339.7 ± 279.9372.1 ± 339.5327.8 ± 267.40.873PFT %FVC81.8 ± 20.278.9 ± 18.480.1 ± 18.086.8 ± 23.80.347 FEV1/FVC ratio79.9 ± 10.280.4 ± 11.981.5 ± 8.977.9 ± 9.30.483 %DLCO64.1 ± 24.568.0 ± 27.263.9 ± 23.160.3 ± 23.30.584HRCT findings, n (%) Honeycomb19 (25.6)7 (25.9)5 (18.5)7 (29.1)0.805 Traction bronchiectasis50 (67.5)22 (81.4)15 (65.2)13 (54.1)\n< 0.05\n Reticulation63 (85.1)25 (92.5)20 (86.9)18 (75.0)0.07 GGO55 (74.3)19 (70.3)16 (69.5)20 (83.3)0.3 Consolidation17 (22.9)5 (18.5)6 (26.0)6 (25.0)0.573 Nodule17 (22.9)7 (25.9)4 (17.3)6 (25.0)0.917 Cyst12 (16.2)4 (14.8)4 (17.3)4 (16.6)0.853HRCT pattern, n (%) UIP5 (6.7)1 (3.7)3 (13.0)1 (4.1)0.91 Probable UIP4 (5.4)2 (7.4)1 (4.3)1 (4.1)0.603 Indeterminate for UIP27 (36.4)14 (51.8)7 (30.4)6 (25.0)\n< 0.05\n Alternative diagnosis38 (51.3)10 (37.0)12 (52.1)16 (66.6)\n< 0.05\nHistological diagnosis, n (%)47 (63.5)11 (40.7)14 (60.8)22 (91.6)\n< 0.05\nNumber of specimens, n2.3 ± 0.72.1 ± 0.82.6 ± 0.52.4 ± 0.80.06Bronchial bleeding48 (64.8)15 (55.5)20 (86.9)12 (50.0)0.75 Mild27 (36.4)6 (22.2)13 (56.5)8 (33.3)0.367 Moderate21 (28.3)9 (33.3)7 (30.4)4 (16.6)0.186Pneumothorax5 (6.7)0 (0)0 (0)5 (20.8)\n< 0.05\n Mild3 (4.0)0 (0)0 (0)3 (12.5)\n< 0.05\n Moderate1 (1.3)0 (0)0 (0)1 (4.1)0.206 Severe1 (1.3)0 (0)0 (0)1 (4.1)0.206Highlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant differenceDLCO, diffusing capacity for carbon monoxide; FEV1, forced expiratory volume in 1 s; FVC, forced volume capacity; GGO, ground-glass opacities; HRCT, high-resolution computed tomography; KL-6, Krebs von den Lungen-6; LAS, large airway specimen; PFT, pulmonary function test; SP-D, surfactant protein D; UIP, usual interstitial pneumonia\nBaseline characteristics of patients\nHighlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant difference\nDLCO, diffusing capacity for carbon monoxide; FEV1, forced expiratory volume in 1 s; FVC, forced volume capacity; GGO, ground-glass opacities; HRCT, high-resolution computed tomography; KL-6, Krebs von den Lungen-6; LAS, large airway specimen; PFT, pulmonary function test; SP-D, surfactant protein D; UIP, usual interstitial pneumonia\n[SUBTITLE] Diagnostic yield and histological diagnosis [SUBSECTION] A specific histological diagnosis was obtained in 47/74 cases (63.5%). The histological diagnostic yield tended to be statistically significantly higher in the LAS-negative group (40.7% in LAS positive group; 60.8% in LAS positive and negative group; 91.6% in LAS negative group) (Cochran-Armitage trend test). The histological interpretations are shown in Table 2 and include 11 (14.8%) UIP, 6 (8.1) probable UIP, 16 (21.6%) indeterminate for UIP, 9 (12.1%) non-specific interstitial pneumonia, 6 (8.1%) organizing pneumonia and 6 (8.1%) hypersensitivity pneumonitis. The institutional diagnosis after MDD is shown in Table 3 and include 9 (12.1%) idiopathic pulmonary fibrosis, 6 (8.1%) non-specific interstitial pneumonia, 4 (5.4%) organizing pneumonia, 24 (32.4%) unclassifiable and 9 (12.1%) hypersensitivity pneumonitis.\nTable 2Histological diagnosisHistological diagnosisNo (%)UIP11 (14.8)Probable UIP6 (8.1)Indeterminate for UIP16 (21.6)– 14 NSIP/UIP– 1 NSIP/UIP/OP– 1 UIP/OPAlternative diagnosis NSIP9 (12.1) OP6 (8.1) HP6 (8.1) Smoking-related IP2 (2.7) Others7 (9.4)– 4 IgG4-related disease– 1 sarcoidosis– 1 cellular bronchiolitis– 1 malignancyN.D.11 (14.8)DPB, diffuse panbronchiolitis; HP, hypersensitivity pneumonitis; N.D., non-diagnostic; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; UIP, usual interstitial pneumoniaTable 3MDD diagnosisMDD diagnosisNo (%)IPF9 (12.1)NSIP6 (8.1)OP4 (5.4)Unclassifiable24 (32.4)Hypersensitivity pneumonitis9 (12.1)CTD-ILD (MCTD/RA/SjS/SLE/SSc)10 (13.5)MPA3 (4.0)Others9 (12.1)CTD, connective tissue disease; IPF, idiopathic pulmonary fibrosis; MCTD, mixed connective tissue disease; MPA, microscopic polyangiitis; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; RA, rheumatoid arthritis; SjS, Sjögren’s syndrome; SLE, systemic lupus erythematosus; SSc, systemic sclerosis\nHistological diagnosis\nDPB, diffuse panbronchiolitis; HP, hypersensitivity pneumonitis; N.D., non-diagnostic; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; UIP, usual interstitial pneumonia\nMDD diagnosis\nCTD, connective tissue disease; IPF, idiopathic pulmonary fibrosis; MCTD, mixed connective tissue disease; MPA, microscopic polyangiitis; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; RA, rheumatoid arthritis; SjS, Sjögren’s syndrome; SLE, systemic lupus erythematosus; SSc, systemic sclerosis\nA specific histological diagnosis was obtained in 47/74 cases (63.5%). The histological diagnostic yield tended to be statistically significantly higher in the LAS-negative group (40.7% in LAS positive group; 60.8% in LAS positive and negative group; 91.6% in LAS negative group) (Cochran-Armitage trend test). The histological interpretations are shown in Table 2 and include 11 (14.8%) UIP, 6 (8.1) probable UIP, 16 (21.6%) indeterminate for UIP, 9 (12.1%) non-specific interstitial pneumonia, 6 (8.1%) organizing pneumonia and 6 (8.1%) hypersensitivity pneumonitis. The institutional diagnosis after MDD is shown in Table 3 and include 9 (12.1%) idiopathic pulmonary fibrosis, 6 (8.1%) non-specific interstitial pneumonia, 4 (5.4%) organizing pneumonia, 24 (32.4%) unclassifiable and 9 (12.1%) hypersensitivity pneumonitis.\nTable 2Histological diagnosisHistological diagnosisNo (%)UIP11 (14.8)Probable UIP6 (8.1)Indeterminate for UIP16 (21.6)– 14 NSIP/UIP– 1 NSIP/UIP/OP– 1 UIP/OPAlternative diagnosis NSIP9 (12.1) OP6 (8.1) HP6 (8.1) Smoking-related IP2 (2.7) Others7 (9.4)– 4 IgG4-related disease– 1 sarcoidosis– 1 cellular bronchiolitis– 1 malignancyN.D.11 (14.8)DPB, diffuse panbronchiolitis; HP, hypersensitivity pneumonitis; N.D., non-diagnostic; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; UIP, usual interstitial pneumoniaTable 3MDD diagnosisMDD diagnosisNo (%)IPF9 (12.1)NSIP6 (8.1)OP4 (5.4)Unclassifiable24 (32.4)Hypersensitivity pneumonitis9 (12.1)CTD-ILD (MCTD/RA/SjS/SLE/SSc)10 (13.5)MPA3 (4.0)Others9 (12.1)CTD, connective tissue disease; IPF, idiopathic pulmonary fibrosis; MCTD, mixed connective tissue disease; MPA, microscopic polyangiitis; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; RA, rheumatoid arthritis; SjS, Sjögren’s syndrome; SLE, systemic lupus erythematosus; SSc, systemic sclerosis\nHistological diagnosis\nDPB, diffuse panbronchiolitis; HP, hypersensitivity pneumonitis; N.D., non-diagnostic; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; UIP, usual interstitial pneumonia\nMDD diagnosis\nCTD, connective tissue disease; IPF, idiopathic pulmonary fibrosis; MCTD, mixed connective tissue disease; MPA, microscopic polyangiitis; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; RA, rheumatoid arthritis; SjS, Sjögren’s syndrome; SLE, systemic lupus erythematosus; SSc, systemic sclerosis\n[SUBTITLE] Complications [SUBSECTION] In a comparison between the three groups, the LAS-negative group tended to have more pneumothorax complications than the other groups (0% in LAS positive group; 0% in LAS positive and negative group; 20.8% in LAS negative group) (Cochran-Armitage trend test). There was no clear difference in bronchial bleeding between the three groups (Table 1).\nIn a comparison between the three groups, the LAS-negative group tended to have more pneumothorax complications than the other groups (0% in LAS positive group; 0% in LAS positive and negative group; 20.8% in LAS negative group) (Cochran-Armitage trend test). There was no clear difference in bronchial bleeding between the three groups (Table 1).\n[SUBTITLE] Specimen characteristics [SUBSECTION] In total, 177 specimens were collected in the 74 cases, with a mean number of specimens per case of 2.3 ± 0.7 (Tables 1 and 4). 175 were collected with 1.9 mm probes and only 2 were collected with 2.4 mm probes. The most common biopsy lobe was the right lower lobe (63.8%), followed by the right upper lobe (15.2%) and the left lower lobe (14.1%). There were 85 (48.0%) LAS containing bronchial gland or bronchial cartilage: 30 containing both bronchial gland and bronchial cartilage, 50 containing only bronchial cartilage, and 5 containing only bronchial gland. The anatomical structures observed except for the large airway were alveoli (91.5%), bronchioles (49.1%), and perilobular area (43.5%), with few pleural specimens (1.2%). Ideal specimens were obtained in 47 of the 177 (26.5%) specimens, about a quarter of the total. A comparison of specimen characteristics showed that the proportion of bronchioles, alveoli, and perilobular area were significantly lower in the LAS-positive group than in the LAS-negative group (Table 3). Similarly, the ideal specimen ratio and the frequency of specimens of high pathological confidence were significantly lower in the LAS-positive group. The ratio of specimens with a pathological confidence level A was 25 of the 47 (53.1%) for ideal specimens and 43 of the 130 (33.0%) for unideal specimens (P < 0.05). In terms of the HRCT findings at the sample collection site, the LAS-positive group had a significantly higher proportion of traction bronchiectasis (65.5% vs. 29.6%) and diaphragm overlap sign (54.1% vs. 33.6%) than the LAS-negative group.\nTable 4Characteristics of specimens obtained by TBLCCharacteristicsAll specimensLAS positiveLAS negative\nP valueSpecimens, n (%)17785 (48.0)92 (51.9)Maximum diameter, mm6.4 ± 2.97.5 ± 3.75.3 ± 1.3\n< 0.05\nBiopsy lobe, n (%) Right upper lobe27 (15.2)7 (8.2)20 (21.7)\n< 0.05\n Right middle lobe6 (3.3)2 (2.3)4 (4.3)0.684 Right lower lobe113 (63.8)58 (68.2)55 (59.7)0.275 Left upper lobe6 (3.3)2 (2.3)4 (4.2)0.684 Left lower lobe25 (14,1)16 (18.8)9 (9.7)0.129Structures observed on the specimen Bronchiole87 (49.1)23 (27.0)64 (69.5)\n< 0.05\n Alveoli162 (91.5)73 (85.8)89 (96.7)\n< 0.05\n Perilobular area77 (43.5)15 (17.6)62 (67.3)\n< 0.05\n Pleura1 (1.2)0 (0)1 (1.0)1Ideal specimen, n (%)47 (26.5)5 (5.8)42 (45.6)\n< 0.05\nPathological confidence level Level A68 (38.4)23 (27.0)45 (48.9)\n< 0.05\n Level B84 (47.4)48 (56.4)36 (39.1)\n< 0.05\n Level C10 (5.6)5 (5.8)5 (5.4)1 Not evaluated15 (8.4)9 (10.5)6 (6.5)HRCT findings in the biopsied lung segment, n (%) Honeycomb8 (4.5)4 (4.7)4 (4.3)1 Traction bronchiectasis101 (57.0)65 (76.4)36 (39.1)\n< 0.05\n Reticulation130 (73.4)68 (80.0)62 (67.3)0.063 GGO60 (33.8)28 (32.9)32 (34.7)0.874 Consolidation16 (9.0)8 (9.4)8 (8.6)1 Nodule14 (7.9)4 (4.7)10 (10.8)0.167 Cyst5 (2.8)0 (0)1 (1.0)0.059Diaphragm overlap sign, n (%)77 (43.5)46 (54.1)31 (33.6)\n< 0.05\nHighlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant differenceGGO, ground-glass opacities; HRCT, high-resolution computed tomography; LAS, large airway specimen\nCharacteristics of specimens obtained by TBLC\nHighlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant difference\nGGO, ground-glass opacities; HRCT, high-resolution computed tomography; LAS, large airway specimen\nIn total, 177 specimens were collected in the 74 cases, with a mean number of specimens per case of 2.3 ± 0.7 (Tables 1 and 4). 175 were collected with 1.9 mm probes and only 2 were collected with 2.4 mm probes. The most common biopsy lobe was the right lower lobe (63.8%), followed by the right upper lobe (15.2%) and the left lower lobe (14.1%). There were 85 (48.0%) LAS containing bronchial gland or bronchial cartilage: 30 containing both bronchial gland and bronchial cartilage, 50 containing only bronchial cartilage, and 5 containing only bronchial gland. The anatomical structures observed except for the large airway were alveoli (91.5%), bronchioles (49.1%), and perilobular area (43.5%), with few pleural specimens (1.2%). Ideal specimens were obtained in 47 of the 177 (26.5%) specimens, about a quarter of the total. A comparison of specimen characteristics showed that the proportion of bronchioles, alveoli, and perilobular area were significantly lower in the LAS-positive group than in the LAS-negative group (Table 3). Similarly, the ideal specimen ratio and the frequency of specimens of high pathological confidence were significantly lower in the LAS-positive group. The ratio of specimens with a pathological confidence level A was 25 of the 47 (53.1%) for ideal specimens and 43 of the 130 (33.0%) for unideal specimens (P < 0.05). In terms of the HRCT findings at the sample collection site, the LAS-positive group had a significantly higher proportion of traction bronchiectasis (65.5% vs. 29.6%) and diaphragm overlap sign (54.1% vs. 33.6%) than the LAS-negative group.\nTable 4Characteristics of specimens obtained by TBLCCharacteristicsAll specimensLAS positiveLAS negative\nP valueSpecimens, n (%)17785 (48.0)92 (51.9)Maximum diameter, mm6.4 ± 2.97.5 ± 3.75.3 ± 1.3\n< 0.05\nBiopsy lobe, n (%) Right upper lobe27 (15.2)7 (8.2)20 (21.7)\n< 0.05\n Right middle lobe6 (3.3)2 (2.3)4 (4.3)0.684 Right lower lobe113 (63.8)58 (68.2)55 (59.7)0.275 Left upper lobe6 (3.3)2 (2.3)4 (4.2)0.684 Left lower lobe25 (14,1)16 (18.8)9 (9.7)0.129Structures observed on the specimen Bronchiole87 (49.1)23 (27.0)64 (69.5)\n< 0.05\n Alveoli162 (91.5)73 (85.8)89 (96.7)\n< 0.05\n Perilobular area77 (43.5)15 (17.6)62 (67.3)\n< 0.05\n Pleura1 (1.2)0 (0)1 (1.0)1Ideal specimen, n (%)47 (26.5)5 (5.8)42 (45.6)\n< 0.05\nPathological confidence level Level A68 (38.4)23 (27.0)45 (48.9)\n< 0.05\n Level B84 (47.4)48 (56.4)36 (39.1)\n< 0.05\n Level C10 (5.6)5 (5.8)5 (5.4)1 Not evaluated15 (8.4)9 (10.5)6 (6.5)HRCT findings in the biopsied lung segment, n (%) Honeycomb8 (4.5)4 (4.7)4 (4.3)1 Traction bronchiectasis101 (57.0)65 (76.4)36 (39.1)\n< 0.05\n Reticulation130 (73.4)68 (80.0)62 (67.3)0.063 GGO60 (33.8)28 (32.9)32 (34.7)0.874 Consolidation16 (9.0)8 (9.4)8 (8.6)1 Nodule14 (7.9)4 (4.7)10 (10.8)0.167 Cyst5 (2.8)0 (0)1 (1.0)0.059Diaphragm overlap sign, n (%)77 (43.5)46 (54.1)31 (33.6)\n< 0.05\nHighlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant differenceGGO, ground-glass opacities; HRCT, high-resolution computed tomography; LAS, large airway specimen\nCharacteristics of specimens obtained by TBLC\nHighlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant difference\nGGO, ground-glass opacities; HRCT, high-resolution computed tomography; LAS, large airway specimen", "This study included 90 consecutive patients evaluated between February 2018 and January 2020. We excluded 16 patients: 9 were missing pulmonary function tests within 3 months of TBLC, 3 were biopsied for peripheral lung lesions using EBUS, 2 were biopsied for central-type lung cancer, and 2 had an inappropriate pathological evaluation. In total, 74 patients were available in this study, and their baseline characteristics are as indicated in Table 1 for the three groups: LAS-positive, LAS-positive and LAS-negative, LAS-negative. There were 50 LAS-positive cases (67.5%), of which all specimens were LAS in 27 cases and only some specimens were LAS in 23 cases. HRCT pattern accounted for approximately 90% of indeterminate for UIP and alternative diagnoses. All cases with HRCT pattern UIP were positive for some collagen-related autoantibodies or myeloperoxidase-antineutrophil cytoplasmic antibody.\nTable 1Baseline characteristics of patientsCharacteristicsAll patientsLAS-positiveLAS-positiveandLAS-negativeLAS-negative\nP valuePatients, n7427 (36.4)23 (31.0)24 (32.4)Male, n (%)40 (54.0)14 (51.8)15 (65.2)11 (45.8)0.698Age, years67.0 ± 11.569.1 ± 7.167.6 ± 8.664.0 ± 16.70.278Current or ex-smokers, n (%)44 (59.4)16 (59.2)17 (73.9)11 (45.8)0.358KL-6, U/mL1565.1 ± 1523.91473 ± 10511784 ± 20481455 ± 14240.713SP-D, ng/mL346.4 ± 293.0339.7 ± 279.9372.1 ± 339.5327.8 ± 267.40.873PFT %FVC81.8 ± 20.278.9 ± 18.480.1 ± 18.086.8 ± 23.80.347 FEV1/FVC ratio79.9 ± 10.280.4 ± 11.981.5 ± 8.977.9 ± 9.30.483 %DLCO64.1 ± 24.568.0 ± 27.263.9 ± 23.160.3 ± 23.30.584HRCT findings, n (%) Honeycomb19 (25.6)7 (25.9)5 (18.5)7 (29.1)0.805 Traction bronchiectasis50 (67.5)22 (81.4)15 (65.2)13 (54.1)\n< 0.05\n Reticulation63 (85.1)25 (92.5)20 (86.9)18 (75.0)0.07 GGO55 (74.3)19 (70.3)16 (69.5)20 (83.3)0.3 Consolidation17 (22.9)5 (18.5)6 (26.0)6 (25.0)0.573 Nodule17 (22.9)7 (25.9)4 (17.3)6 (25.0)0.917 Cyst12 (16.2)4 (14.8)4 (17.3)4 (16.6)0.853HRCT pattern, n (%) UIP5 (6.7)1 (3.7)3 (13.0)1 (4.1)0.91 Probable UIP4 (5.4)2 (7.4)1 (4.3)1 (4.1)0.603 Indeterminate for UIP27 (36.4)14 (51.8)7 (30.4)6 (25.0)\n< 0.05\n Alternative diagnosis38 (51.3)10 (37.0)12 (52.1)16 (66.6)\n< 0.05\nHistological diagnosis, n (%)47 (63.5)11 (40.7)14 (60.8)22 (91.6)\n< 0.05\nNumber of specimens, n2.3 ± 0.72.1 ± 0.82.6 ± 0.52.4 ± 0.80.06Bronchial bleeding48 (64.8)15 (55.5)20 (86.9)12 (50.0)0.75 Mild27 (36.4)6 (22.2)13 (56.5)8 (33.3)0.367 Moderate21 (28.3)9 (33.3)7 (30.4)4 (16.6)0.186Pneumothorax5 (6.7)0 (0)0 (0)5 (20.8)\n< 0.05\n Mild3 (4.0)0 (0)0 (0)3 (12.5)\n< 0.05\n Moderate1 (1.3)0 (0)0 (0)1 (4.1)0.206 Severe1 (1.3)0 (0)0 (0)1 (4.1)0.206Highlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant differenceDLCO, diffusing capacity for carbon monoxide; FEV1, forced expiratory volume in 1 s; FVC, forced volume capacity; GGO, ground-glass opacities; HRCT, high-resolution computed tomography; KL-6, Krebs von den Lungen-6; LAS, large airway specimen; PFT, pulmonary function test; SP-D, surfactant protein D; UIP, usual interstitial pneumonia\nBaseline characteristics of patients\nHighlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant difference\nDLCO, diffusing capacity for carbon monoxide; FEV1, forced expiratory volume in 1 s; FVC, forced volume capacity; GGO, ground-glass opacities; HRCT, high-resolution computed tomography; KL-6, Krebs von den Lungen-6; LAS, large airway specimen; PFT, pulmonary function test; SP-D, surfactant protein D; UIP, usual interstitial pneumonia", "A specific histological diagnosis was obtained in 47/74 cases (63.5%). The histological diagnostic yield tended to be statistically significantly higher in the LAS-negative group (40.7% in LAS positive group; 60.8% in LAS positive and negative group; 91.6% in LAS negative group) (Cochran-Armitage trend test). The histological interpretations are shown in Table 2 and include 11 (14.8%) UIP, 6 (8.1) probable UIP, 16 (21.6%) indeterminate for UIP, 9 (12.1%) non-specific interstitial pneumonia, 6 (8.1%) organizing pneumonia and 6 (8.1%) hypersensitivity pneumonitis. The institutional diagnosis after MDD is shown in Table 3 and include 9 (12.1%) idiopathic pulmonary fibrosis, 6 (8.1%) non-specific interstitial pneumonia, 4 (5.4%) organizing pneumonia, 24 (32.4%) unclassifiable and 9 (12.1%) hypersensitivity pneumonitis.\nTable 2Histological diagnosisHistological diagnosisNo (%)UIP11 (14.8)Probable UIP6 (8.1)Indeterminate for UIP16 (21.6)– 14 NSIP/UIP– 1 NSIP/UIP/OP– 1 UIP/OPAlternative diagnosis NSIP9 (12.1) OP6 (8.1) HP6 (8.1) Smoking-related IP2 (2.7) Others7 (9.4)– 4 IgG4-related disease– 1 sarcoidosis– 1 cellular bronchiolitis– 1 malignancyN.D.11 (14.8)DPB, diffuse panbronchiolitis; HP, hypersensitivity pneumonitis; N.D., non-diagnostic; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; UIP, usual interstitial pneumoniaTable 3MDD diagnosisMDD diagnosisNo (%)IPF9 (12.1)NSIP6 (8.1)OP4 (5.4)Unclassifiable24 (32.4)Hypersensitivity pneumonitis9 (12.1)CTD-ILD (MCTD/RA/SjS/SLE/SSc)10 (13.5)MPA3 (4.0)Others9 (12.1)CTD, connective tissue disease; IPF, idiopathic pulmonary fibrosis; MCTD, mixed connective tissue disease; MPA, microscopic polyangiitis; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; RA, rheumatoid arthritis; SjS, Sjögren’s syndrome; SLE, systemic lupus erythematosus; SSc, systemic sclerosis\nHistological diagnosis\nDPB, diffuse panbronchiolitis; HP, hypersensitivity pneumonitis; N.D., non-diagnostic; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; UIP, usual interstitial pneumonia\nMDD diagnosis\nCTD, connective tissue disease; IPF, idiopathic pulmonary fibrosis; MCTD, mixed connective tissue disease; MPA, microscopic polyangiitis; NSIP, non-specific interstitial pneumonia; OP, organizing pneumonia; RA, rheumatoid arthritis; SjS, Sjögren’s syndrome; SLE, systemic lupus erythematosus; SSc, systemic sclerosis", "In a comparison between the three groups, the LAS-negative group tended to have more pneumothorax complications than the other groups (0% in LAS positive group; 0% in LAS positive and negative group; 20.8% in LAS negative group) (Cochran-Armitage trend test). There was no clear difference in bronchial bleeding between the three groups (Table 1).", "In total, 177 specimens were collected in the 74 cases, with a mean number of specimens per case of 2.3 ± 0.7 (Tables 1 and 4). 175 were collected with 1.9 mm probes and only 2 were collected with 2.4 mm probes. The most common biopsy lobe was the right lower lobe (63.8%), followed by the right upper lobe (15.2%) and the left lower lobe (14.1%). There were 85 (48.0%) LAS containing bronchial gland or bronchial cartilage: 30 containing both bronchial gland and bronchial cartilage, 50 containing only bronchial cartilage, and 5 containing only bronchial gland. The anatomical structures observed except for the large airway were alveoli (91.5%), bronchioles (49.1%), and perilobular area (43.5%), with few pleural specimens (1.2%). Ideal specimens were obtained in 47 of the 177 (26.5%) specimens, about a quarter of the total. A comparison of specimen characteristics showed that the proportion of bronchioles, alveoli, and perilobular area were significantly lower in the LAS-positive group than in the LAS-negative group (Table 3). Similarly, the ideal specimen ratio and the frequency of specimens of high pathological confidence were significantly lower in the LAS-positive group. The ratio of specimens with a pathological confidence level A was 25 of the 47 (53.1%) for ideal specimens and 43 of the 130 (33.0%) for unideal specimens (P < 0.05). In terms of the HRCT findings at the sample collection site, the LAS-positive group had a significantly higher proportion of traction bronchiectasis (65.5% vs. 29.6%) and diaphragm overlap sign (54.1% vs. 33.6%) than the LAS-negative group.\nTable 4Characteristics of specimens obtained by TBLCCharacteristicsAll specimensLAS positiveLAS negative\nP valueSpecimens, n (%)17785 (48.0)92 (51.9)Maximum diameter, mm6.4 ± 2.97.5 ± 3.75.3 ± 1.3\n< 0.05\nBiopsy lobe, n (%) Right upper lobe27 (15.2)7 (8.2)20 (21.7)\n< 0.05\n Right middle lobe6 (3.3)2 (2.3)4 (4.3)0.684 Right lower lobe113 (63.8)58 (68.2)55 (59.7)0.275 Left upper lobe6 (3.3)2 (2.3)4 (4.2)0.684 Left lower lobe25 (14,1)16 (18.8)9 (9.7)0.129Structures observed on the specimen Bronchiole87 (49.1)23 (27.0)64 (69.5)\n< 0.05\n Alveoli162 (91.5)73 (85.8)89 (96.7)\n< 0.05\n Perilobular area77 (43.5)15 (17.6)62 (67.3)\n< 0.05\n Pleura1 (1.2)0 (0)1 (1.0)1Ideal specimen, n (%)47 (26.5)5 (5.8)42 (45.6)\n< 0.05\nPathological confidence level Level A68 (38.4)23 (27.0)45 (48.9)\n< 0.05\n Level B84 (47.4)48 (56.4)36 (39.1)\n< 0.05\n Level C10 (5.6)5 (5.8)5 (5.4)1 Not evaluated15 (8.4)9 (10.5)6 (6.5)HRCT findings in the biopsied lung segment, n (%) Honeycomb8 (4.5)4 (4.7)4 (4.3)1 Traction bronchiectasis101 (57.0)65 (76.4)36 (39.1)\n< 0.05\n Reticulation130 (73.4)68 (80.0)62 (67.3)0.063 GGO60 (33.8)28 (32.9)32 (34.7)0.874 Consolidation16 (9.0)8 (9.4)8 (8.6)1 Nodule14 (7.9)4 (4.7)10 (10.8)0.167 Cyst5 (2.8)0 (0)1 (1.0)0.059Diaphragm overlap sign, n (%)77 (43.5)46 (54.1)31 (33.6)\n< 0.05\nHighlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant differenceGGO, ground-glass opacities; HRCT, high-resolution computed tomography; LAS, large airway specimen\nCharacteristics of specimens obtained by TBLC\nHighlighted in bold the areas where a p-value of less than 0.05 indicates a statistically significant difference\nGGO, ground-glass opacities; HRCT, high-resolution computed tomography; LAS, large airway specimen", "To our knowledge, this is the first retrospective study of LAS obtained in TBLC, and it has revealed three important points. First, LAS are specimens that can be routinely collected by TBLC in patients with DPLD and most LAS are unideal specimens that do not contain bronchioles or perilobular area. Second, LAS are more likely to be collected in the presence of traction bronchiectasis or diaphragm overlap sign on HRCT of the biopsied lung segment. Third, TBLC cases that do not contain LAS tend to have a higher histological diagnostic yield.\nPrevious reports comparing the diagnostic yield of TBLC and SLB in DPLD suggested that specimens containing large airway or pleura, specimens with non-specific pathological findings or hemorrhage artifacts, and normal lung tissue were recognized as unideal specimens that may not contribute to the histological diagnosis [6, 7, 9, 18]. However, in terms of the definition of an ideal specimen, most of the studies defined adequacy as the inclusion of alveoli in the specimen and did not define whether there were concomitant bronchioles, perilobular area, or pleura [13]. However, pathological evaluation of specimens largely occupied by alveoli may be unideal for some histological patterns in which lesions are located also in the bronchiole or perilobular area (e.g., UIP pattern, UIP/NSIP pattern, hypersensitivity pneumonitis, sarcoidosis). Therefore, we considered that the conventional definition of only the presence of alveoli as a requirement for an ideal specimen is insufficient and inaccurate. And we investigated the possibility that TBLC specimens containing all of bronchioles, alveoli, and perilobular area would be ideal for histological diagnosis. Based on this definition, we attempted to clarify the exact role of LAS in the present TBLC study.\nTo date, only two reports have referred to specimens from the large airway in TBLC. Ussavarungsi et al. [10] reported that specimens including large airway were found in 47% of TBLC cases, and Pajares et al. reported that specimens including bronchial wall were found in 8.6% of cases [11]. However, the definition of LAS was not rigorous, and we defined LAS as specimens that include bronchial gland or bronchial cartilage based on the results of a previous anatomical study [15]. In the present study, LAS were found in 67.5% of TBLC cases and 48.0% of TBLC specimens under the usual TBLC procedure, indicating that LAS are not infrequent and can be commonly collected in TBLC. Besides, most of the LAS were unideal specimens (94.2%), and the difference in the proportion of ideal specimens in the LAS-negative group was statistically significant (Table 4). We must also mention that there is uncertainty about whether the method of specimen evaluation described above is universal because no similar studies have been conducted previously, to our knowledge. However, we believe that these definitions will be helpful in creating a smoother and less misunderstood discussion of diagnostic yield and the adequacy of TBLC specimens.\nWe reviewed the HRCT findings in the biopsied lung segment where LAS were collected and obtained two interesting findings. First, the rate of LAS collection increased when traction bronchiectasis was present (Table 4). In addition to previous reports cautioning that dense honeycomb lesions or dense fibrotic lung parenchyma lesions should be avoided in TBLC sampling [12, 19], we speculate that optimal sampling sites in TBLC may be in areas mainly ground-glass opacities and reticular shadows where architectural distortion such as traction bronchiectasis or honeycomb is not evident [10]. Second, the presence of a diaphragm overlap sign increases the ratio of LAS collection. HRCT, which is usually performed before TBLC, is performed on deep inspiration with the patient in the awake state. However, during the actual TBLC procedure, deep sedation is often used to reduce pain, and patients rarely inhale deeply or stop breathing. In other words, it is common for TBLC to be performed in situations where the precise location of the biopsy is more difficult to determine than when HRCT is performed. Therefore, we believe that if the area to be sampled at the time of HRCT overlaps the sub-diaphragm area, there is a greater possibility that the area will be hidden by the diaphragm at the time of TBLC. If the biopsy site overlaps the sub-diaphragmatic area, determination of whether the cryobiopsy probe has reached just below the pleura can be inaccurate, leading to an inaccurate biopsy of the peripheral lung. To predict whether such a precarious situation would occur, we established the diaphragm overlap sign and found that the percentage of LAS collected increased when the sign was positive. Although the fluoroscopic guidance is widely used in bronchoscopy, its accuracy is limited to the lateral lung area [20]. The use of cone beam CT-guided TBLC (CBCT-guided TBLC) has recently been reported to reduce complications such as pneumothorax and hemorrhage and improve diagnostic accuracy because the distance between the probe and pleura can be more accurately determined [21–23]. CBCT-guided TBLC is not a universal technique because it is performed under general anesthesia in a hybrid operating room. However, we believe that these reports are significant in that they demonstrate the usefulness of multi-dimensional (e.g.; axial, coronal, sagittal) guidance. Therefore, for fluoroscopic TBLC performed under the guidance of coronal view only, it is advisable to confirm the expected position of the pleura-probe in the axial and sagittal images of HRCT in advance, and to avoid biopsy of the area with positive diaphragm overlap sign.\nAlthough the impact of the presence of LAS on the histological diagnostic yield has not been studied before, our study suggested that LAS had apparent effect on the rate of histological diagnosis (Table 1). On the other hand, about half of the biopsy specimens were LAS-positive, and we believe that the optimal number of specimens should be investigated. The average number of specimens collected was 2.7 in this study, and we collected a total of 2 or more specimens from different lung segments in 66/74 (89.1%) cases. 23 of the 51 LAS-positive cases had both LAS-positive and LAS-negative specimens in the same TBLC procedure. Therefore, it is important for pulmonologists to recognize that there is always the possibility of LAS in individual biopsy specimens and to perform TBLC so that multiple biopsies do not diminish the accuracy of histological diagnosis. Previous reports by Ravaglia et al. that obtaining two specimens from different segments within the same lobe may reduce sampling error and increase diagnostic yield would also support our current considerations [17, 24].\nThis study has several limitations. First, this is a retrospective observational study with a small number of cases from a single center. Second, the pathological evaluation was performed by a single pathologist. Considering the low rate of concordance between pathologists in the histological evaluation of TBLC specimens, it would be necessary for multiple pathologists to perform the histological evaluation. Third, several new pathological definitions (adequacy of a specimen and LAS) and new HRCT definition (diaphragm overlap sign) were used in the present study, but these are not generalized concepts or indicators, and their validity needs to be evaluated by pathologists and radiologists in the future. Finally, we did not compare the pathological findings of SLB in the same case. This study was conducted from the viewpoint of LAS as one of the factors involved in the diagnostic discrepancy between SLB and TBLC, and a comparative study of the characteristics of specimens and LAS collection ratios of both SLB and TBLC will further clarify the reasons behind the difference in diagnostic accuracy.", "In conclusion, this study showed that LAS obtained in TBLC have a high frequency of unideal specimens that do not include the bronchiole or perilobular area. Since the histological diagnostic yield tends to be higher in cases that do not contain LAS, it may be important to determine the biopsy site that reduces the frequency of LAS collection by referring to the HRCT findings in TBLC." ]
[ null, "materials|methods", null, null, null, null, null, null, "results", null, null, null, null, "discussion", "conclusion" ]
[ "Large airway specimen", "Histological diagnostic yield", "Specimen adequacy", "Surgical lung biopsy", "Transbronchial lung cryobiopsy" ]
The use of frozen embryos and frozen sperm have complementary IVF outcomes: a retrospective analysis in couples experiencing IVF/Donor and IVF/Husband.
36258180
The cryopreservation of sperm or embryos has been an important strategy in the treatment of infertility. Recently studies have revealed the outcomes after IVF (in vitro fertilization) treatment for single-factor exposure either to frozen sperm or embryos.
BACKGROUND
This retrospective study was to uncover the exposure to both frozen sperm and embryo effects using IVF/H (in vitro fertilization using husbands' fresh sperm) or IVF/D (in vitro fertilization using donors' frozen sperm) treatment.
METHODS
The results showed the clinical pregnancy rate (CPR), live birth rate (LBR) and low birth weight rate (LBW) increased to 63.2% (or 68.1%), 61.1% (or 66.4%) and 15.8% (or 16.2%) after using frozen embryo transfer within Group IVF/H (or Group IVF/D). After using frozen sperm, the high-quality embryo rate (HER) increased to 52% and baby with birth defect rate (BDR) reduced to 0% in subgroup D/ET comparing to subgroup H/ET. While the fertilization rate (FER), cleavage rate (CLR), HER and multiple pregnancy rate (MUR) reduced to 75%, 71%, 45% and 9.2% in subgroup D/FET comparing to subgroup H/FET. Finally, our study found accumulative frozen gamete effects, including both sperm and embryos, led to the significantly increasing in the HER (p < 0.05), CPR (p < 0.001), LBR (p < 0.001) and LBW (p < 0.05) in subgroup D/FET comparing to subgroup H/ET.
RESULTS
The use of frozen embryos and frozen sperm have complementary IVF outcomes. Our findings highlighted the parent's distinguished frozen effect not only for clinical studies but also for basic research on the mechanism of cellular response adaptations to cryopreservation.
CONCLUSION
[ "Pregnancy", "Female", "Male", "Humans", "Retrospective Studies", "Spouses", "Semen", "Fertilization in Vitro", "Pregnancy Rate", "Spermatozoa" ]
9578274
Introduction
To keep the division of embryo the same pace with the growth of the endometrium, the transferring of frozen embryos have been widely used in assisted reproductive technology (ART). Since the first successful report of frozen embryo transfer (FET) [1], the cryopreservation of embryos has been an important strategy in the treatment of infertility. FET strategies contribute an additional 25–50% chance of pregnancy for couples who have cryopreserved embryos [2–4]. However, FET is not free from the risk of a higher multipregnancy rate (MPR) and low birth weight rate (LBW), even though the live birth rate (LBR) of frozen–thawed embryos is usually higher than that of fresh transferred embryos [5–8]. On the other hand, male due to azoospermia or sperm retrieval difficulties on the day of egg retrieval in vitro fertilization with frozen spermatozoa is used to treat female who might also have tubal lesions or those experiencing failure of prior artificial insemination with donor semen (AID) cycles [9, 10]. LBW Meanwhile, there is a matter of debate on the clinical outcomes caused by alterations in the DNA integrity of semen following cryopreservation [11, 12]. Indeed, the baby with no birth defect rate following pregnancies after IVF/D was not different (97.3% vs. 97.4%) [13] after IVF with fresh husband spermatozoa, but the clinical pregnancy rate (CPR) per transfer was higher after using frozen donor semen than that after using the husbands’ semen (27.6% vs. 21.9%, respectively)[13] Above all, taking advantage of freezing sperm, eggs or embryos is just like a double-edged sword for IVF outcomes, so doctors who use it need to be cautious. Nowadays, the practical ART will also meet the clinical treatment of one couple not only need to freeze sperm, but also suitable for freezing embryos. There still have been less studies of this double-factors freezing on the IVF outcomes including fertilization, pregnancy, and child birth. Most of published studies are incomprehensive, they are retrospective or refer to cases involving either frozen embryo transfer or frozen donor spermatozoa. For this article, we cover the findings of both freezing embryos transfer (FET) and IVF/D. As far as ART procedures are concerned, FET and IVF/D lead to some specific questions requiring answers: (i) the effects on the pregnancy and neonate characteristics of single-factor exposure to frozen embryo transfer; (ii) the effects on the fertilization, pregnancy and neonate characteristics of single-factor exposure to frozen donor semen; and (iii) the influence of the duration of sperm and embryo cryopreservation on pregnancy and newborn health. Methods.
null
null
Results
[SUBTITLE] Participants and grouping [SUBSECTION] A total of 860 couples undergoing IVF-ET treatment were included in the analysis, including 573 couples with husband sperm (Group IVF/H) and 287 couples with frozen donor sperm (Group IVF/D). According to whether or not frozen embryo transfer was performed, the IVF/H group was divided into the H/ET subgroup (133 couples treated by IVF/H and fresh embryo transfer) and the H/FET subgroup (440 couples treated by IVF/H and frozen embryo transfer); moreover, the IVF/D group was also divided into the D/ET subgroup (49 couples treated by IVF/D and fresh embryo transfer) and the D/FET subgroup (238 couples treated by IVF/D and frozen embryo transfer) (detailed in Fig. 1). A total of 860 couples undergoing IVF-ET treatment were included in the analysis, including 573 couples with husband sperm (Group IVF/H) and 287 couples with frozen donor sperm (Group IVF/D). According to whether or not frozen embryo transfer was performed, the IVF/H group was divided into the H/ET subgroup (133 couples treated by IVF/H and fresh embryo transfer) and the H/FET subgroup (440 couples treated by IVF/H and frozen embryo transfer); moreover, the IVF/D group was also divided into the D/ET subgroup (49 couples treated by IVF/D and fresh embryo transfer) and the D/FET subgroup (238 couples treated by IVF/D and frozen embryo transfer) (detailed in Fig. 1). [SUBTITLE] The baseline characteristics of the couples [SUBSECTION] Except for the female BMI in the FN subgroup, the variables were nonnormally distributed among the other subgroups. Thus, using the nonparametric test (Kruskal–Wallis model), we found that the median endometrial thickness (10 cm vs. 9 cm, p = 0.015), oocytes retrieved (8 vs. 11, p = 0.000) and MII oocytes (6 vs. 10, p = 0.000) were differentiated within Group IVF/H, but we found that only female age (30 years vs. 28 years, p = 0.002) was differentiated within Group IVF/D. There were no significant differences (p > 0.05) within Group IVF/H or Group IVF/D, including the male age, semen parameters and female BMI (detailed in Table 1). Table 1The basic characteristics of the 860 couples in one IVF/H (or IVF/D) cycleBaseline ParametersGroup IVF/H P value (H/ET vs. H/FET)Group IVF/D P value (D/ET vs. D/FET)H/ET1 subgroupH/FET2 subgroupD/ET3 subgroupD/FET4 subgroupMedian [first quartile, third quartile]Median [first quartile, third quartile]Inclusion Samples (n)133440/49238/Male Age (husband or donor) a32 [29, 34]32 [29, 34]0.73123 [22, 26]23 [21, 27]0.967Semen Volume (Ml) a3 [2, 3]3 [3]0.3482 [2, 2]2 [2, 2]0.608Semen Concentration (10^6/ML) a56 [48, 64]54 [48, 62]0.73446 [43, 53]47 [43, 51]0.974Semen (PR + NP) (%)a56 [50, 60]54 [50, 60]0.93146 [43, 54]48 [43, 52]0.574Female Age a33 [30, 35]32 [30, 35]0.64430 [28, 35]28 [26, 32]0.002**Female BMI (kg/m2) a21 [24, 25]21 [26, 27]0.86222 [20, 23]21 [20, 24]0.862Endometrial Thickness (cm) a10 [8, 12]9 [7, 11]0.015*10 [8, 12]10 [8, 12]0.805Oocytes Retrieved (n) a8 [4, 11]11[8, 16]0.000***11 [8, 15]11 [8, 15]0.590MII Oocytes (n) a6 [4, 10]10 [6, 14]0.000***9 [7, 13]9 [6, 12]0.721MII Oocytes (%)a93 [78, 100]91 [82, 100]0.66081 [68, 96]86 [73, 96]0.503Note:1: H/ET subgroup = using fresh sperm and fresh embryo transfer;2: H/FET subgroup = using fresh sperm and frozen embryo transfer;3: D/ET subgroup = using frozen sperm and fresh embryo transfer;4: D/FET subgroup = using frozen sperm and frozen embryo transfer;a: Nonparametric test (Kruskal–Wallis Model)* p < 0.05 **p < 0.01 ***p < 0.001 The basic characteristics of the 860 couples in one IVF/H (or IVF/D) cycle Note: 1: H/ET subgroup = using fresh sperm and fresh embryo transfer; 2: H/FET subgroup = using fresh sperm and frozen embryo transfer; 3: D/ET subgroup = using frozen sperm and fresh embryo transfer; 4: D/FET subgroup = using frozen sperm and frozen embryo transfer; a: Nonparametric test (Kruskal–Wallis Model) * p < 0.05 **p < 0.01 ***p < 0.001 Except for the female BMI in the FN subgroup, the variables were nonnormally distributed among the other subgroups. Thus, using the nonparametric test (Kruskal–Wallis model), we found that the median endometrial thickness (10 cm vs. 9 cm, p = 0.015), oocytes retrieved (8 vs. 11, p = 0.000) and MII oocytes (6 vs. 10, p = 0.000) were differentiated within Group IVF/H, but we found that only female age (30 years vs. 28 years, p = 0.002) was differentiated within Group IVF/D. There were no significant differences (p > 0.05) within Group IVF/H or Group IVF/D, including the male age, semen parameters and female BMI (detailed in Table 1). Table 1The basic characteristics of the 860 couples in one IVF/H (or IVF/D) cycleBaseline ParametersGroup IVF/H P value (H/ET vs. H/FET)Group IVF/D P value (D/ET vs. D/FET)H/ET1 subgroupH/FET2 subgroupD/ET3 subgroupD/FET4 subgroupMedian [first quartile, third quartile]Median [first quartile, third quartile]Inclusion Samples (n)133440/49238/Male Age (husband or donor) a32 [29, 34]32 [29, 34]0.73123 [22, 26]23 [21, 27]0.967Semen Volume (Ml) a3 [2, 3]3 [3]0.3482 [2, 2]2 [2, 2]0.608Semen Concentration (10^6/ML) a56 [48, 64]54 [48, 62]0.73446 [43, 53]47 [43, 51]0.974Semen (PR + NP) (%)a56 [50, 60]54 [50, 60]0.93146 [43, 54]48 [43, 52]0.574Female Age a33 [30, 35]32 [30, 35]0.64430 [28, 35]28 [26, 32]0.002**Female BMI (kg/m2) a21 [24, 25]21 [26, 27]0.86222 [20, 23]21 [20, 24]0.862Endometrial Thickness (cm) a10 [8, 12]9 [7, 11]0.015*10 [8, 12]10 [8, 12]0.805Oocytes Retrieved (n) a8 [4, 11]11[8, 16]0.000***11 [8, 15]11 [8, 15]0.590MII Oocytes (n) a6 [4, 10]10 [6, 14]0.000***9 [7, 13]9 [6, 12]0.721MII Oocytes (%)a93 [78, 100]91 [82, 100]0.66081 [68, 96]86 [73, 96]0.503Note:1: H/ET subgroup = using fresh sperm and fresh embryo transfer;2: H/FET subgroup = using fresh sperm and frozen embryo transfer;3: D/ET subgroup = using frozen sperm and fresh embryo transfer;4: D/FET subgroup = using frozen sperm and frozen embryo transfer;a: Nonparametric test (Kruskal–Wallis Model)* p < 0.05 **p < 0.01 ***p < 0.001 The basic characteristics of the 860 couples in one IVF/H (or IVF/D) cycle Note: 1: H/ET subgroup = using fresh sperm and fresh embryo transfer; 2: H/FET subgroup = using fresh sperm and frozen embryo transfer; 3: D/ET subgroup = using frozen sperm and fresh embryo transfer; 4: D/FET subgroup = using frozen sperm and frozen embryo transfer; a: Nonparametric test (Kruskal–Wallis Model) * p < 0.05 **p < 0.01 ***p < 0.001 [SUBTITLE] Maternal factor exposure to frozen embryo transfer: the comparison of clinical outcomes within group IVF/H or group IVF/D [SUBSECTION] Using the nonparametric test (Kruskal–Wallis Model), the outcomes of fertilization and embryo transfer were compared within Group IVF/H and Group IVF/D (see Table 2). Before embryo transfer, the median of all the fertilization outcomes in the H/FET subgroup was higher than that in the H/ET subgroup. The median [first quartile, third quartile] fertilization rate (%), cleavage rate (%) and high-quality embryo rate (%) were 80 [67, 91], 100 [80, 100] and 57 [36, 78], respectively, in the H/FET subgroup. In contrast, there was no significant difference between the D/ET and D/FET subgroups. After frozen embryo transfer, the CPR and LBR were significantly higher in the H/FET subgroup than in the fresh embryo treatment subgroup and the H/ET subgroup (63.2 vs. 39.8, p = 0.000; 61.1 vs. 39.1, p = 0.000; 14.8 vs. 6.7, p = 0.016, respectively). However, the LBW was significantly higher in the H/FET subgroup than in the fresh embryo treatment subgroup in the H/ET subgroup (16.2 vs. 4.9, p = 0.021). In another IVF/D group, the CPR and LBR were significantly higher in the D/FET subgroup than in the fresh embryo treatment subgroup in the D/ET subgroup (68.1 vs. 49.0, p = 0.017; 66.4 vs. 47.0, p = 0.010, respectively). Otherwise, the BDR was significantly higher in the D/FET subgroup than in the fresh embryo treatment group in subgroup D/ET (1.0 vs. 0.0, p = 0.000). Table 2The IVF outcomes of the 860 couples in one IVF/H (or IVF/D) cycleOutcome IndicatorsGroup IVF/H (Χ 2 ) P value (H/ET vs. H/FET)Group IVF/D (Χ 2 ) P value (D/ET vs. D/FET)H/ET1 subgroupH/FET2 subgroupD/ET3 subgroupD/FET4 subgroupMedian [first quartile, third quartile]Median [first quartile, third quartile]After FertilizationFertilization Number (n)a5 [3, 8]8 [5, 11]0.000***8 [6, 12]8 [5, 11]0.342Fertilization Rate (%)a71 [50,89]80 [67,91]0.001**67 [57,88]75 [57,84]0.822Cleavage Number (n) a4 [2, 8]8 [5, 11]0.000***8 [6, 11]7 [4, 11]0.309Cleavage Rate (%)a67 [50,88]100 [80,100]0.000***67 [57,88]71 [55,82]0.667High-Quality Embryos (n) a2 [1, 5]4 [3, 7]0.000***6 [3, 8]4 [2, 7]0.105High-Quality Embryos Rate (%)a33 [10,54]57 [36,78]0.000***52 [33,67]45 [28,60]0.070After TransferBiochemical Pregnancy Rate (%)b0.01.4(2.377) 0.1232.00.4(1.542) 0.214Clinical Pregnancy Rate (%)b39.863.2(22.789) 0.000***49.068.1(5.734) 0.017*Live Birth Rate (%)b39.161.1(20.134) 0.000***47.066.4(6.598) 0.010*Multipregnancy Rate (%)b6.714.8(5.820) 0.016*14.39.2(1.137) 0.286Miscarriage Rate (%)b0.82.0(0.997) 0.3182.02.1(0.001) 0.979Total Baby Sex Ratio (%)b69.4114.1(3.129) 0.077121.482.0(1.036) 0.309Low Birth Weight Rate (%)b4.916.2(5.286) 0.021*19.415.8(0.029) 0.806Baby with Birth Defect Rate (%)b1.61.5(0.007) 0.9330.01.0(216.712) 0.000***Note:1: H/ET subgroup = using fresh sperm and fresh embryo transfer;2: H/FET subgroup = using fresh sperm and frozen embryo transfer;3: D/ET subgroup = using frozen sperm and fresh embryo transfer;4: D/FET subgroup = using frozen sperm and frozen embryo transfer;a: Nonparametric Test (Kruskal–Wallis Model)b: chi square test for independent samples* p < 0.05 **p < 0.01 ***p < 0.001 The IVF outcomes of the 860 couples in one IVF/H (or IVF/D) cycle Note: 1: H/ET subgroup = using fresh sperm and fresh embryo transfer; 2: H/FET subgroup = using fresh sperm and frozen embryo transfer; 3: D/ET subgroup = using frozen sperm and fresh embryo transfer; 4: D/FET subgroup = using frozen sperm and frozen embryo transfer; a: Nonparametric Test (Kruskal–Wallis Model) b: chi square test for independent samples * p < 0.05 **p < 0.01 ***p < 0.001 Using the nonparametric test (Kruskal–Wallis Model), the outcomes of fertilization and embryo transfer were compared within Group IVF/H and Group IVF/D (see Table 2). Before embryo transfer, the median of all the fertilization outcomes in the H/FET subgroup was higher than that in the H/ET subgroup. The median [first quartile, third quartile] fertilization rate (%), cleavage rate (%) and high-quality embryo rate (%) were 80 [67, 91], 100 [80, 100] and 57 [36, 78], respectively, in the H/FET subgroup. In contrast, there was no significant difference between the D/ET and D/FET subgroups. After frozen embryo transfer, the CPR and LBR were significantly higher in the H/FET subgroup than in the fresh embryo treatment subgroup and the H/ET subgroup (63.2 vs. 39.8, p = 0.000; 61.1 vs. 39.1, p = 0.000; 14.8 vs. 6.7, p = 0.016, respectively). However, the LBW was significantly higher in the H/FET subgroup than in the fresh embryo treatment subgroup in the H/ET subgroup (16.2 vs. 4.9, p = 0.021). In another IVF/D group, the CPR and LBR were significantly higher in the D/FET subgroup than in the fresh embryo treatment subgroup in the D/ET subgroup (68.1 vs. 49.0, p = 0.017; 66.4 vs. 47.0, p = 0.010, respectively). Otherwise, the BDR was significantly higher in the D/FET subgroup than in the fresh embryo treatment group in subgroup D/ET (1.0 vs. 0.0, p = 0.000). Table 2The IVF outcomes of the 860 couples in one IVF/H (or IVF/D) cycleOutcome IndicatorsGroup IVF/H (Χ 2 ) P value (H/ET vs. H/FET)Group IVF/D (Χ 2 ) P value (D/ET vs. D/FET)H/ET1 subgroupH/FET2 subgroupD/ET3 subgroupD/FET4 subgroupMedian [first quartile, third quartile]Median [first quartile, third quartile]After FertilizationFertilization Number (n)a5 [3, 8]8 [5, 11]0.000***8 [6, 12]8 [5, 11]0.342Fertilization Rate (%)a71 [50,89]80 [67,91]0.001**67 [57,88]75 [57,84]0.822Cleavage Number (n) a4 [2, 8]8 [5, 11]0.000***8 [6, 11]7 [4, 11]0.309Cleavage Rate (%)a67 [50,88]100 [80,100]0.000***67 [57,88]71 [55,82]0.667High-Quality Embryos (n) a2 [1, 5]4 [3, 7]0.000***6 [3, 8]4 [2, 7]0.105High-Quality Embryos Rate (%)a33 [10,54]57 [36,78]0.000***52 [33,67]45 [28,60]0.070After TransferBiochemical Pregnancy Rate (%)b0.01.4(2.377) 0.1232.00.4(1.542) 0.214Clinical Pregnancy Rate (%)b39.863.2(22.789) 0.000***49.068.1(5.734) 0.017*Live Birth Rate (%)b39.161.1(20.134) 0.000***47.066.4(6.598) 0.010*Multipregnancy Rate (%)b6.714.8(5.820) 0.016*14.39.2(1.137) 0.286Miscarriage Rate (%)b0.82.0(0.997) 0.3182.02.1(0.001) 0.979Total Baby Sex Ratio (%)b69.4114.1(3.129) 0.077121.482.0(1.036) 0.309Low Birth Weight Rate (%)b4.916.2(5.286) 0.021*19.415.8(0.029) 0.806Baby with Birth Defect Rate (%)b1.61.5(0.007) 0.9330.01.0(216.712) 0.000***Note:1: H/ET subgroup = using fresh sperm and fresh embryo transfer;2: H/FET subgroup = using fresh sperm and frozen embryo transfer;3: D/ET subgroup = using frozen sperm and fresh embryo transfer;4: D/FET subgroup = using frozen sperm and frozen embryo transfer;a: Nonparametric Test (Kruskal–Wallis Model)b: chi square test for independent samples* p < 0.05 **p < 0.01 ***p < 0.001 The IVF outcomes of the 860 couples in one IVF/H (or IVF/D) cycle Note: 1: H/ET subgroup = using fresh sperm and fresh embryo transfer; 2: H/FET subgroup = using fresh sperm and frozen embryo transfer; 3: D/ET subgroup = using frozen sperm and fresh embryo transfer; 4: D/FET subgroup = using frozen sperm and frozen embryo transfer; a: Nonparametric Test (Kruskal–Wallis Model) b: chi square test for independent samples * p < 0.05 **p < 0.01 ***p < 0.001 [SUBTITLE] Paternal factor exposure to frozen sperm fertilization: the comparison of clinical outcomes between Group IVF/H and Group IVF/D [SUBSECTION] Using the chi square test for independent samples, the comparison of clinical outcomes between Group IVF/H and Group IVF/D is shown in Figs. 2 and 3. Compared with the D/ET subgroup using frozen sperm (donor sperm) fertilization, the HER was higher than that of the H/ET subgroup using fresh husband sperm (p < 0.05). In contrast, the values of the FER, CLR and HER in the D/FET subgroup were lower than those of the H/FET subgroup using fresh husband sperm (all p < 0.05). After embryo transfer, the BDR in the D/ET subgroup was lower than that of the H/ET subgroup using fresh husband sperm (p < 0.001). Meanwhile, the MUR in the D/FET subgroup was lower than that of the H/FET subgroup using fresh husband sperm (p < 0.05). Fig. 2The comparison of fertilization outcomes between Group IVF/H and Group IVF/D. The values of the FER, CLR and HER in subgroups H/ET and H/FET are shown in the separated blue histogram and those of the D/ET and D/FET subgroups are shown in the red histogram. Note: FER = fertilization rate; CLR = cleavage rate; HER = high-quality embryo rate; * p < 0.05 **p < 0.01 ***p < 0.001 The comparison of fertilization outcomes between Group IVF/H and Group IVF/D. The values of the FER, CLR and HER in subgroups H/ET and H/FET are shown in the separated blue histogram and those of the D/ET and D/FET subgroups are shown in the red histogram. Note: FER = fertilization rate; CLR = cleavage rate; HER = high-quality embryo rate; * p < 0.05 **p < 0.01 ***p < 0.001 Fig. 3The comparison of embryo transfer outcomes between Group IVF/H and Group IVF/D. The proportions of the BPR, CPR, LBR, MUR, MSR, TBSR, LBW and BDR in the H/ET and H/FET subgroups are shown in the combined blue histogram and those of the D/ET or D/FET subgroups are shown in the combined red histogram. Note: BPR = biochemical pregnancy rate; CPR = clinical pregnancy rate; LBR = live birth rate; MUR = multipregnancy rate; MSR = miscarriage rate; TBSR = total baby sex ratio; LBW = low birth weight rate; BDR = baby with birth defect rate;* p < 0.05 **p < 0.01 ***p < 0.001 The comparison of embryo transfer outcomes between Group IVF/H and Group IVF/D. The proportions of the BPR, CPR, LBR, MUR, MSR, TBSR, LBW and BDR in the H/ET and H/FET subgroups are shown in the combined blue histogram and those of the D/ET or D/FET subgroups are shown in the combined red histogram. Note: BPR = biochemical pregnancy rate; CPR = clinical pregnancy rate; LBR = live birth rate; MUR = multipregnancy rate; MSR = miscarriage rate; TBSR = total baby sex ratio; LBW = low birth weight rate; BDR = baby with birth defect rate;* p < 0.05 **p < 0.01 ***p < 0.001 Using the chi square test for independent samples, the comparison of clinical outcomes between Group IVF/H and Group IVF/D is shown in Figs. 2 and 3. Compared with the D/ET subgroup using frozen sperm (donor sperm) fertilization, the HER was higher than that of the H/ET subgroup using fresh husband sperm (p < 0.05). In contrast, the values of the FER, CLR and HER in the D/FET subgroup were lower than those of the H/FET subgroup using fresh husband sperm (all p < 0.05). After embryo transfer, the BDR in the D/ET subgroup was lower than that of the H/ET subgroup using fresh husband sperm (p < 0.001). Meanwhile, the MUR in the D/FET subgroup was lower than that of the H/FET subgroup using fresh husband sperm (p < 0.05). Fig. 2The comparison of fertilization outcomes between Group IVF/H and Group IVF/D. The values of the FER, CLR and HER in subgroups H/ET and H/FET are shown in the separated blue histogram and those of the D/ET and D/FET subgroups are shown in the red histogram. Note: FER = fertilization rate; CLR = cleavage rate; HER = high-quality embryo rate; * p < 0.05 **p < 0.01 ***p < 0.001 The comparison of fertilization outcomes between Group IVF/H and Group IVF/D. The values of the FER, CLR and HER in subgroups H/ET and H/FET are shown in the separated blue histogram and those of the D/ET and D/FET subgroups are shown in the red histogram. Note: FER = fertilization rate; CLR = cleavage rate; HER = high-quality embryo rate; * p < 0.05 **p < 0.01 ***p < 0.001 Fig. 3The comparison of embryo transfer outcomes between Group IVF/H and Group IVF/D. The proportions of the BPR, CPR, LBR, MUR, MSR, TBSR, LBW and BDR in the H/ET and H/FET subgroups are shown in the combined blue histogram and those of the D/ET or D/FET subgroups are shown in the combined red histogram. Note: BPR = biochemical pregnancy rate; CPR = clinical pregnancy rate; LBR = live birth rate; MUR = multipregnancy rate; MSR = miscarriage rate; TBSR = total baby sex ratio; LBW = low birth weight rate; BDR = baby with birth defect rate;* p < 0.05 **p < 0.01 ***p < 0.001 The comparison of embryo transfer outcomes between Group IVF/H and Group IVF/D. The proportions of the BPR, CPR, LBR, MUR, MSR, TBSR, LBW and BDR in the H/ET and H/FET subgroups are shown in the combined blue histogram and those of the D/ET or D/FET subgroups are shown in the combined red histogram. Note: BPR = biochemical pregnancy rate; CPR = clinical pregnancy rate; LBR = live birth rate; MUR = multipregnancy rate; MSR = miscarriage rate; TBSR = total baby sex ratio; LBW = low birth weight rate; BDR = baby with birth defect rate;* p < 0.05 **p < 0.01 ***p < 0.001 [SUBTITLE] Parental factors exposure to frozen sperm fertilization and frozen embryo transfer: the comparison of clinical outcomes between Group IVF/H and Group IVF/D [SUBSECTION] When both frozen sperm and frozen embryo transfer were used in the D/FET group, the HER was higher than that for the use of both fresh sperm and fresh embryo transfer in the H/ET group (p < 0.05). After embryo transfer, the CPR and LBR values in the D/FET subgroup were higher than those in the H/ET group (p < 0.001). Meanwhile, the LBW in the D/FET subgroup was higher than that in the H/ET subgroup (p < 0.05). When both frozen sperm and frozen embryo transfer were used in the D/FET group, the HER was higher than that for the use of both fresh sperm and fresh embryo transfer in the H/ET group (p < 0.05). After embryo transfer, the CPR and LBR values in the D/FET subgroup were higher than those in the H/ET group (p < 0.001). Meanwhile, the LBW in the D/FET subgroup was higher than that in the H/ET subgroup (p < 0.05).
Conclusion
This study demonstrates that using frozen sperm or frozen embryo transfer have distinguished effects on the different IVF stages. Especially, the use of frozen embryos and frozen sperm have complementary IVF outcomes. Basic research on the mechanism of the cellular response adaptations to cryopreservation are needed to support our findings.
[ "Methods", "The processing of ovulation, fertilization and embryo transfer were as follows", "Grouping and statistical analysis", "Participants and grouping", "The baseline characteristics of the couples", "Maternal factor exposure to frozen embryo transfer: the comparison of clinical outcomes within group IVF/H or group IVF/D", "Paternal factor exposure to frozen sperm fertilization: the comparison of clinical outcomes between Group IVF/H and Group IVF/D", "Parental factors exposure to frozen sperm fertilization and frozen embryo transfer: the comparison of clinical outcomes between Group IVF/H and Group IVF/D" ]
[ "This study was approved by the Ethical Committee of the institutional review board of the Ethical Committee of the Obstetrics and Gynaecology Hospital of Fudan University and written informed consent was obtained from all couples. All methods were performed in accordance with the relevant guidelines and regulations.\nWe retrospectively analyzed the IVF treatment of couples experiencing infertility with frozen-thawed donor sperm (Group IVF/D) or fresh husband sperm (Group IVF/H) at Shanghai Ji Ai Genetics and IVF Institute (Jan. 2013-Feb. 2019), after which we followed up until the birth of the baby. For Group IVF/D, the donor’s inclusion criteria were normal sperm parameters above World Health Organization guidelines [14]. For Group IVF/H, the husband’s inclusion criteria were also normal sperm parameters according to World Health Organization guidelines [14]. The inclusion criteria for women in both groups were those undergoing their first IVF-ET cycle caused only by tubal obstruction factors. Women with premature ovarian failure, polycystic ovarian syndrome, chromosome abnormalities, habitual abortion and other diseases that cause infertility were all excluded from this study. In addition, male patients with anti-sperm antibodies, high DNA fragmentation index, non-ejaculation, retrograde ejaculation, chromosome abnormalities and other diseases that cause infertility were also excluded.\nThe processing of the semen sample treatments was as follows: According to the World Health Organization guidelines [14], donors’ semen samples were obtained by masturbation. After liquefaction of the fresh ejaculate, the specimens’ characteristics were evaluated, such as volume, count and motility. The qualified donors followed the National Sperm Bank reference for semen parameters (before freezing: volume ≥ 2.0 ml, concentration ≥ 60 million/ml, progressive motility ≥ 60%, normal sperm morphology ≥ 70%; after thawing: concentration ≥ 15 million/ml, progressive motility ≥ 32%, normal sperm morphology ≥ 4%). On the day of egg collection by uterine surgery, the semen samples were thawed in a water bath at 37 ℃. Then, the semen sample was treated by the density gradient method (45% and 90% gradient solution, Vitrolife, Gothenburg, Sweden), by which the samples were centrifuged at 500 g for 20 min. After removing the supernatant, the precipitate was washed with washing solution (Vitrolife, Gothenburg, Sweden) and centrifuged at 300 g for 10 min. For the husband, all the semen samples were obtained by masturbation on the day of egg collection by uterine surgery, and the fresh sperm were used in the IVF-ET cycles.\n[SUBTITLE] The processing of ovulation, fertilization and embryo transfer were as follows [SUBSECTION] controlled ovarian hyperstimulation (COH) used a gonadotropin-releasing hormone agonist protocol (the following was a shortened version of the long protocol) or a GnRH antagonist protocol, both of which are effective in blocking a premature LH surge [15, 16]. Generally, for the long protocol, a GnRH agonist (Triptorelin Acetate, Ipsen Pharma Biotech, France) was administered by subcutaneous injection daily starting from the luteal phase of the menstrual cycle for 10–14 days, and then ovarian stimulation with rFSH (Gn-F, Merck Serono SA Aubonne Branch, Switzerland) commenced. For the GnRH antagonist protocol, ovarian stimulation began on the second day of the menstrual cycle, and on the fifth day, antagonist (Cetrorelix Acetate, Cetrotide, Serono Labs Inc., Switzerland) administration started. Once the leading two follicles reached 18 mm or larger in diameter, the hCG administration was ejected as a trigger on the same day. Thirty-five to forty hours later, a doctor punctured the follicles and collected the eggs with the guidance of an ultrasound instrument. The sperm and oocytes (10000:1) were added to 0.1 ml of prebalanced embryo culture medium into four-well plates covered with mineral oil. The evaluation of embryo quality was performed on the 3rd day after fertilization. The embryos were divided into four levels according to the characteristics of blastomeres, such as the number, the morphology and fragments. Grade I and Grade II embryos were defined as high-quality embryos, and the other grades were defined as low-quality embryos [17, 18]. When the number of high-quality embryos was two or lower and the woman was younger than 35 years old, the embryos were frozen for transfer. All transferred embryos were 4-cell embryos. The frozen embryos were thawed according to the rapid recovery method of vitrification. Embryos with a recovery rate above 50% could be used for transfer. After embryo transfer, the patients were followed up until birth.\ncontrolled ovarian hyperstimulation (COH) used a gonadotropin-releasing hormone agonist protocol (the following was a shortened version of the long protocol) or a GnRH antagonist protocol, both of which are effective in blocking a premature LH surge [15, 16]. Generally, for the long protocol, a GnRH agonist (Triptorelin Acetate, Ipsen Pharma Biotech, France) was administered by subcutaneous injection daily starting from the luteal phase of the menstrual cycle for 10–14 days, and then ovarian stimulation with rFSH (Gn-F, Merck Serono SA Aubonne Branch, Switzerland) commenced. For the GnRH antagonist protocol, ovarian stimulation began on the second day of the menstrual cycle, and on the fifth day, antagonist (Cetrorelix Acetate, Cetrotide, Serono Labs Inc., Switzerland) administration started. Once the leading two follicles reached 18 mm or larger in diameter, the hCG administration was ejected as a trigger on the same day. Thirty-five to forty hours later, a doctor punctured the follicles and collected the eggs with the guidance of an ultrasound instrument. The sperm and oocytes (10000:1) were added to 0.1 ml of prebalanced embryo culture medium into four-well plates covered with mineral oil. The evaluation of embryo quality was performed on the 3rd day after fertilization. The embryos were divided into four levels according to the characteristics of blastomeres, such as the number, the morphology and fragments. Grade I and Grade II embryos were defined as high-quality embryos, and the other grades were defined as low-quality embryos [17, 18]. When the number of high-quality embryos was two or lower and the woman was younger than 35 years old, the embryos were frozen for transfer. All transferred embryos were 4-cell embryos. The frozen embryos were thawed according to the rapid recovery method of vitrification. Embryos with a recovery rate above 50% could be used for transfer. After embryo transfer, the patients were followed up until birth.\n[SUBTITLE] Grouping and statistical analysis [SUBSECTION] According to the treatment of embryo transfer, Group IVF/D was divided into the D/ET subgroup (treated with frozen donor sperm and Fresh embryo transfer) and the D/FET subgroup (treated with frozen donor sperm and frozen embryo transfer). Group IVF/H was also divided into the H/ET subgroup (treated with Fresh husband sperm and Fresh embryo transfer) and the H/FET subgroup (treated with Fresh husband sperm and frozen embryo transfer). The detail of the research grouping flow chat is shown in Fig. 1.\n\nFig. 1The research grouping flow chat\n\nThe research grouping flow chat\nThe female-related factors, including age, GnRH injection days and dosage, the estrogen secretion peak of egg collection, endometrium thickness, the number of eggs retrieved, the number of fertilizations, the number of effective embryos, the number of high-quality embryos, etc., were followed up and analyzed by SPSS 19.0 software (SPSS Inc., Cambridge, MA, USA). First, all the parameters were checked by a normal distribution test (Kolmogorov–Smirnov model and Shapiro–Wilk model). Then, the medians (first quartile, third quartile) of the continuous parameters were calculated. Finally, the variation in clinical outcomes within or between the IVF/D or IVF/H groups was compared using the nonparametric test (Kruskal–Wallis model) or chi square test for independent samples (p < 0.05).\nAccording to the treatment of embryo transfer, Group IVF/D was divided into the D/ET subgroup (treated with frozen donor sperm and Fresh embryo transfer) and the D/FET subgroup (treated with frozen donor sperm and frozen embryo transfer). Group IVF/H was also divided into the H/ET subgroup (treated with Fresh husband sperm and Fresh embryo transfer) and the H/FET subgroup (treated with Fresh husband sperm and frozen embryo transfer). The detail of the research grouping flow chat is shown in Fig. 1.\n\nFig. 1The research grouping flow chat\n\nThe research grouping flow chat\nThe female-related factors, including age, GnRH injection days and dosage, the estrogen secretion peak of egg collection, endometrium thickness, the number of eggs retrieved, the number of fertilizations, the number of effective embryos, the number of high-quality embryos, etc., were followed up and analyzed by SPSS 19.0 software (SPSS Inc., Cambridge, MA, USA). First, all the parameters were checked by a normal distribution test (Kolmogorov–Smirnov model and Shapiro–Wilk model). Then, the medians (first quartile, third quartile) of the continuous parameters were calculated. Finally, the variation in clinical outcomes within or between the IVF/D or IVF/H groups was compared using the nonparametric test (Kruskal–Wallis model) or chi square test for independent samples (p < 0.05).", "controlled ovarian hyperstimulation (COH) used a gonadotropin-releasing hormone agonist protocol (the following was a shortened version of the long protocol) or a GnRH antagonist protocol, both of which are effective in blocking a premature LH surge [15, 16]. Generally, for the long protocol, a GnRH agonist (Triptorelin Acetate, Ipsen Pharma Biotech, France) was administered by subcutaneous injection daily starting from the luteal phase of the menstrual cycle for 10–14 days, and then ovarian stimulation with rFSH (Gn-F, Merck Serono SA Aubonne Branch, Switzerland) commenced. For the GnRH antagonist protocol, ovarian stimulation began on the second day of the menstrual cycle, and on the fifth day, antagonist (Cetrorelix Acetate, Cetrotide, Serono Labs Inc., Switzerland) administration started. Once the leading two follicles reached 18 mm or larger in diameter, the hCG administration was ejected as a trigger on the same day. Thirty-five to forty hours later, a doctor punctured the follicles and collected the eggs with the guidance of an ultrasound instrument. The sperm and oocytes (10000:1) were added to 0.1 ml of prebalanced embryo culture medium into four-well plates covered with mineral oil. The evaluation of embryo quality was performed on the 3rd day after fertilization. The embryos were divided into four levels according to the characteristics of blastomeres, such as the number, the morphology and fragments. Grade I and Grade II embryos were defined as high-quality embryos, and the other grades were defined as low-quality embryos [17, 18]. When the number of high-quality embryos was two or lower and the woman was younger than 35 years old, the embryos were frozen for transfer. All transferred embryos were 4-cell embryos. The frozen embryos were thawed according to the rapid recovery method of vitrification. Embryos with a recovery rate above 50% could be used for transfer. After embryo transfer, the patients were followed up until birth.", "According to the treatment of embryo transfer, Group IVF/D was divided into the D/ET subgroup (treated with frozen donor sperm and Fresh embryo transfer) and the D/FET subgroup (treated with frozen donor sperm and frozen embryo transfer). Group IVF/H was also divided into the H/ET subgroup (treated with Fresh husband sperm and Fresh embryo transfer) and the H/FET subgroup (treated with Fresh husband sperm and frozen embryo transfer). The detail of the research grouping flow chat is shown in Fig. 1.\n\nFig. 1The research grouping flow chat\n\nThe research grouping flow chat\nThe female-related factors, including age, GnRH injection days and dosage, the estrogen secretion peak of egg collection, endometrium thickness, the number of eggs retrieved, the number of fertilizations, the number of effective embryos, the number of high-quality embryos, etc., were followed up and analyzed by SPSS 19.0 software (SPSS Inc., Cambridge, MA, USA). First, all the parameters were checked by a normal distribution test (Kolmogorov–Smirnov model and Shapiro–Wilk model). Then, the medians (first quartile, third quartile) of the continuous parameters were calculated. Finally, the variation in clinical outcomes within or between the IVF/D or IVF/H groups was compared using the nonparametric test (Kruskal–Wallis model) or chi square test for independent samples (p < 0.05).", "A total of 860 couples undergoing IVF-ET treatment were included in the analysis, including 573 couples with husband sperm (Group IVF/H) and 287 couples with frozen donor sperm (Group IVF/D). According to whether or not frozen embryo transfer was performed, the IVF/H group was divided into the H/ET subgroup (133 couples treated by IVF/H and fresh embryo transfer) and the H/FET subgroup (440 couples treated by IVF/H and frozen embryo transfer); moreover, the IVF/D group was also divided into the D/ET subgroup (49 couples treated by IVF/D and fresh embryo transfer) and the D/FET subgroup (238 couples treated by IVF/D and frozen embryo transfer) (detailed in Fig. 1).", "Except for the female BMI in the FN subgroup, the variables were nonnormally distributed among the other subgroups. Thus, using the nonparametric test (Kruskal–Wallis model), we found that the median endometrial thickness (10 cm vs. 9 cm, p = 0.015), oocytes retrieved (8 vs. 11, p = 0.000) and MII oocytes (6 vs. 10, p = 0.000) were differentiated within Group IVF/H, but we found that only female age (30 years vs. 28 years, p = 0.002) was differentiated within Group IVF/D. There were no significant differences (p > 0.05) within Group IVF/H or Group IVF/D, including the male age, semen parameters and female BMI (detailed in Table 1).\n\nTable 1The basic characteristics of the 860 couples in one IVF/H (or IVF/D) cycleBaseline ParametersGroup IVF/H\nP value\n(H/ET vs. H/FET)Group IVF/D\nP value\n(D/ET vs. D/FET)H/ET1 subgroupH/FET2 subgroupD/ET3 subgroupD/FET4 subgroupMedian [first quartile, third quartile]Median [first quartile, third quartile]Inclusion Samples (n)133440/49238/Male Age (husband or donor) a32 [29, 34]32 [29, 34]0.73123 [22, 26]23 [21, 27]0.967Semen Volume (Ml) a3 [2, 3]3 [3]0.3482 [2, 2]2 [2, 2]0.608Semen Concentration (10^6/ML) a56 [48, 64]54 [48, 62]0.73446 [43, 53]47 [43, 51]0.974Semen (PR + NP) (%)a56 [50, 60]54 [50, 60]0.93146 [43, 54]48 [43, 52]0.574Female Age a33 [30, 35]32 [30, 35]0.64430 [28, 35]28 [26, 32]0.002**Female BMI (kg/m2) a21 [24, 25]21 [26, 27]0.86222 [20, 23]21 [20, 24]0.862Endometrial Thickness (cm) a10 [8, 12]9 [7, 11]0.015*10 [8, 12]10 [8, 12]0.805Oocytes Retrieved (n) a8 [4, 11]11[8, 16]0.000***11 [8, 15]11 [8, 15]0.590MII Oocytes (n) a6 [4, 10]10 [6, 14]0.000***9 [7, 13]9 [6, 12]0.721MII Oocytes (%)a93 [78, 100]91 [82, 100]0.66081 [68, 96]86 [73, 96]0.503Note:1: H/ET subgroup = using fresh sperm and fresh embryo transfer;2: H/FET subgroup = using fresh sperm and frozen embryo transfer;3: D/ET subgroup = using frozen sperm and fresh embryo transfer;4: D/FET subgroup = using frozen sperm and frozen embryo transfer;a: Nonparametric test (Kruskal–Wallis Model)* p < 0.05 **p < 0.01 ***p < 0.001\n\nThe basic characteristics of the 860 couples in one IVF/H (or IVF/D) cycle\nNote:\n1: H/ET subgroup = using fresh sperm and fresh embryo transfer;\n2: H/FET subgroup = using fresh sperm and frozen embryo transfer;\n3: D/ET subgroup = using frozen sperm and fresh embryo transfer;\n4: D/FET subgroup = using frozen sperm and frozen embryo transfer;\na: Nonparametric test (Kruskal–Wallis Model)\n* p < 0.05 **p < 0.01 ***p < 0.001", "Using the nonparametric test (Kruskal–Wallis Model), the outcomes of fertilization and embryo transfer were compared within Group IVF/H and Group IVF/D (see Table 2). Before embryo transfer, the median of all the fertilization outcomes in the H/FET subgroup was higher than that in the H/ET subgroup. The median [first quartile, third quartile] fertilization rate (%), cleavage rate (%) and high-quality embryo rate (%) were 80 [67, 91], 100 [80, 100] and 57 [36, 78], respectively, in the H/FET subgroup. In contrast, there was no significant difference between the D/ET and D/FET subgroups. After frozen embryo transfer, the CPR and LBR were significantly higher in the H/FET subgroup than in the fresh embryo treatment subgroup and the H/ET subgroup (63.2 vs. 39.8, p = 0.000; 61.1 vs. 39.1, p = 0.000; 14.8 vs. 6.7, p = 0.016, respectively). However, the LBW was significantly higher in the H/FET subgroup than in the fresh embryo treatment subgroup in the H/ET subgroup (16.2 vs. 4.9, p = 0.021). In another IVF/D group, the CPR and LBR were significantly higher in the D/FET subgroup than in the fresh embryo treatment subgroup in the D/ET subgroup (68.1 vs. 49.0, p = 0.017; 66.4 vs. 47.0, p = 0.010, respectively). Otherwise, the BDR was significantly higher in the D/FET subgroup than in the fresh embryo treatment group in subgroup D/ET (1.0 vs. 0.0, p = 0.000).\n\nTable 2The IVF outcomes of the 860 couples in one IVF/H (or IVF/D) cycleOutcome IndicatorsGroup IVF/H\n(Χ\n2\n)\n\nP value\n(H/ET vs. H/FET)Group IVF/D\n(Χ\n2\n)\n\nP value\n(D/ET vs. D/FET)H/ET1 subgroupH/FET2 subgroupD/ET3 subgroupD/FET4 subgroupMedian [first quartile, third quartile]Median [first quartile, third quartile]After FertilizationFertilization Number (n)a5 [3, 8]8 [5, 11]0.000***8 [6, 12]8 [5, 11]0.342Fertilization Rate (%)a71 [50,89]80 [67,91]0.001**67 [57,88]75 [57,84]0.822Cleavage Number (n) a4 [2, 8]8 [5, 11]0.000***8 [6, 11]7 [4, 11]0.309Cleavage Rate (%)a67 [50,88]100 [80,100]0.000***67 [57,88]71 [55,82]0.667High-Quality Embryos (n) a2 [1, 5]4 [3, 7]0.000***6 [3, 8]4 [2, 7]0.105High-Quality Embryos Rate (%)a33 [10,54]57 [36,78]0.000***52 [33,67]45 [28,60]0.070After TransferBiochemical Pregnancy Rate (%)b0.01.4(2.377) 0.1232.00.4(1.542) 0.214Clinical Pregnancy Rate (%)b39.863.2(22.789) 0.000***49.068.1(5.734) 0.017*Live Birth Rate (%)b39.161.1(20.134) 0.000***47.066.4(6.598) 0.010*Multipregnancy Rate (%)b6.714.8(5.820) 0.016*14.39.2(1.137) 0.286Miscarriage Rate (%)b0.82.0(0.997) 0.3182.02.1(0.001) 0.979Total Baby Sex Ratio (%)b69.4114.1(3.129) 0.077121.482.0(1.036) 0.309Low Birth Weight Rate (%)b4.916.2(5.286) 0.021*19.415.8(0.029) 0.806Baby with Birth Defect Rate (%)b1.61.5(0.007) 0.9330.01.0(216.712) 0.000***Note:1: H/ET subgroup = using fresh sperm and fresh embryo transfer;2: H/FET subgroup = using fresh sperm and frozen embryo transfer;3: D/ET subgroup = using frozen sperm and fresh embryo transfer;4: D/FET subgroup = using frozen sperm and frozen embryo transfer;a: Nonparametric Test (Kruskal–Wallis Model)b: chi square test for independent samples* p < 0.05 **p < 0.01 ***p < 0.001\n\nThe IVF outcomes of the 860 couples in one IVF/H (or IVF/D) cycle\nNote:\n1: H/ET subgroup = using fresh sperm and fresh embryo transfer;\n2: H/FET subgroup = using fresh sperm and frozen embryo transfer;\n3: D/ET subgroup = using frozen sperm and fresh embryo transfer;\n4: D/FET subgroup = using frozen sperm and frozen embryo transfer;\na: Nonparametric Test (Kruskal–Wallis Model)\nb: chi square test for independent samples\n* p < 0.05 **p < 0.01 ***p < 0.001", "Using the chi square test for independent samples, the comparison of clinical outcomes between Group IVF/H and Group IVF/D is shown in Figs. 2 and 3. Compared with the D/ET subgroup using frozen sperm (donor sperm) fertilization, the HER was higher than that of the H/ET subgroup using fresh husband sperm (p < 0.05). In contrast, the values of the FER, CLR and HER in the D/FET subgroup were lower than those of the H/FET subgroup using fresh husband sperm (all p < 0.05). After embryo transfer, the BDR in the D/ET subgroup was lower than that of the H/ET subgroup using fresh husband sperm (p < 0.001). Meanwhile, the MUR in the D/FET subgroup was lower than that of the H/FET subgroup using fresh husband sperm (p < 0.05).\n\nFig. 2The comparison of fertilization outcomes between Group IVF/H and Group IVF/D. The values of the FER, CLR and HER in subgroups H/ET and H/FET are shown in the separated blue histogram and those of the D/ET and D/FET subgroups are shown in the red histogram. Note: FER = fertilization rate; CLR = cleavage rate; HER = high-quality embryo rate; * p < 0.05 **p < 0.01 ***p < 0.001\n\nThe comparison of fertilization outcomes between Group IVF/H and Group IVF/D. The values of the FER, CLR and HER in subgroups H/ET and H/FET are shown in the separated blue histogram and those of the D/ET and D/FET subgroups are shown in the red histogram. Note: FER = fertilization rate; CLR = cleavage rate; HER = high-quality embryo rate; * p < 0.05 **p < 0.01 ***p < 0.001\n\nFig. 3The comparison of embryo transfer outcomes between Group IVF/H and Group IVF/D. The proportions of the BPR, CPR, LBR, MUR, MSR, TBSR, LBW and BDR in the H/ET and H/FET subgroups are shown in the combined blue histogram and those of the D/ET or D/FET subgroups are shown in the combined red histogram. Note: BPR = biochemical pregnancy rate; CPR = clinical pregnancy rate; LBR = live birth rate; MUR = multipregnancy rate; MSR = miscarriage rate; TBSR = total baby sex ratio; LBW = low birth weight rate; BDR = baby with birth defect rate;* p < 0.05 **p < 0.01 ***p < 0.001\n\nThe comparison of embryo transfer outcomes between Group IVF/H and Group IVF/D. The proportions of the BPR, CPR, LBR, MUR, MSR, TBSR, LBW and BDR in the H/ET and H/FET subgroups are shown in the combined blue histogram and those of the D/ET or D/FET subgroups are shown in the combined red histogram. Note: BPR = biochemical pregnancy rate; CPR = clinical pregnancy rate; LBR = live birth rate; MUR = multipregnancy rate; MSR = miscarriage rate; TBSR = total baby sex ratio; LBW = low birth weight rate; BDR = baby with birth defect rate;* p < 0.05 **p < 0.01 ***p < 0.001", "When both frozen sperm and frozen embryo transfer were used in the D/FET group, the HER was higher than that for the use of both fresh sperm and fresh embryo transfer in the H/ET group (p < 0.05). After embryo transfer, the CPR and LBR values in the D/FET subgroup were higher than those in the H/ET group (p < 0.001). Meanwhile, the LBW in the D/FET subgroup was higher than that in the H/ET subgroup (p < 0.05)." ]
[ null, null, null, null, null, null, null, null ]
[ "Introduction", "Methods", "The processing of ovulation, fertilization and embryo transfer were as follows", "Grouping and statistical analysis", "Results", "Participants and grouping", "The baseline characteristics of the couples", "Maternal factor exposure to frozen embryo transfer: the comparison of clinical outcomes within group IVF/H or group IVF/D", "Paternal factor exposure to frozen sperm fertilization: the comparison of clinical outcomes between Group IVF/H and Group IVF/D", "Parental factors exposure to frozen sperm fertilization and frozen embryo transfer: the comparison of clinical outcomes between Group IVF/H and Group IVF/D", "Discussion", "Conclusion" ]
[ "To keep the division of embryo the same pace with the growth of the endometrium, the transferring of frozen embryos have been widely used in assisted reproductive technology (ART). Since the first successful report of frozen embryo transfer (FET) [1], the cryopreservation of embryos has been an important strategy in the treatment of infertility. FET strategies contribute an additional 25–50% chance of pregnancy for couples who have cryopreserved embryos [2–4]. However, FET is not free from the risk of a higher multipregnancy rate (MPR) and low birth weight rate (LBW), even though the live birth rate (LBR) of frozen–thawed embryos is usually higher than that of fresh transferred embryos [5–8]. On the other hand, male due to azoospermia or sperm retrieval difficulties on the day of egg retrieval in vitro fertilization with frozen spermatozoa is used to treat female who might also have tubal lesions or those experiencing failure of prior artificial insemination with donor semen (AID) cycles [9, 10]. LBW Meanwhile, there is a matter of debate on the clinical outcomes caused by alterations in the DNA integrity of semen following cryopreservation [11, 12]. Indeed, the baby with no birth defect rate following pregnancies after IVF/D was not different (97.3% vs. 97.4%) [13] after IVF with fresh husband spermatozoa, but the clinical pregnancy rate (CPR) per transfer was higher after using frozen donor semen than that after using the husbands’ semen (27.6% vs. 21.9%, respectively)[13] Above all, taking advantage of freezing sperm, eggs or embryos is just like a double-edged sword for IVF outcomes, so doctors who use it need to be cautious.\nNowadays, the practical ART will also meet the clinical treatment of one couple not only need to freeze sperm, but also suitable for freezing embryos. There still have been less studies of this double-factors freezing on the IVF outcomes including fertilization, pregnancy, and child birth. Most of published studies are incomprehensive, they are retrospective or refer to cases involving either frozen embryo transfer or frozen donor spermatozoa. For this article, we cover the findings of both freezing embryos transfer (FET) and IVF/D. As far as ART procedures are concerned, FET and IVF/D lead to some specific questions requiring answers: (i) the effects on the pregnancy and neonate characteristics of single-factor exposure to frozen embryo transfer; (ii) the effects on the fertilization, pregnancy and neonate characteristics of single-factor exposure to frozen donor semen; and (iii) the influence of the duration of sperm and embryo cryopreservation on pregnancy and newborn health. Methods.", "This study was approved by the Ethical Committee of the institutional review board of the Ethical Committee of the Obstetrics and Gynaecology Hospital of Fudan University and written informed consent was obtained from all couples. All methods were performed in accordance with the relevant guidelines and regulations.\nWe retrospectively analyzed the IVF treatment of couples experiencing infertility with frozen-thawed donor sperm (Group IVF/D) or fresh husband sperm (Group IVF/H) at Shanghai Ji Ai Genetics and IVF Institute (Jan. 2013-Feb. 2019), after which we followed up until the birth of the baby. For Group IVF/D, the donor’s inclusion criteria were normal sperm parameters above World Health Organization guidelines [14]. For Group IVF/H, the husband’s inclusion criteria were also normal sperm parameters according to World Health Organization guidelines [14]. The inclusion criteria for women in both groups were those undergoing their first IVF-ET cycle caused only by tubal obstruction factors. Women with premature ovarian failure, polycystic ovarian syndrome, chromosome abnormalities, habitual abortion and other diseases that cause infertility were all excluded from this study. In addition, male patients with anti-sperm antibodies, high DNA fragmentation index, non-ejaculation, retrograde ejaculation, chromosome abnormalities and other diseases that cause infertility were also excluded.\nThe processing of the semen sample treatments was as follows: According to the World Health Organization guidelines [14], donors’ semen samples were obtained by masturbation. After liquefaction of the fresh ejaculate, the specimens’ characteristics were evaluated, such as volume, count and motility. The qualified donors followed the National Sperm Bank reference for semen parameters (before freezing: volume ≥ 2.0 ml, concentration ≥ 60 million/ml, progressive motility ≥ 60%, normal sperm morphology ≥ 70%; after thawing: concentration ≥ 15 million/ml, progressive motility ≥ 32%, normal sperm morphology ≥ 4%). On the day of egg collection by uterine surgery, the semen samples were thawed in a water bath at 37 ℃. Then, the semen sample was treated by the density gradient method (45% and 90% gradient solution, Vitrolife, Gothenburg, Sweden), by which the samples were centrifuged at 500 g for 20 min. After removing the supernatant, the precipitate was washed with washing solution (Vitrolife, Gothenburg, Sweden) and centrifuged at 300 g for 10 min. For the husband, all the semen samples were obtained by masturbation on the day of egg collection by uterine surgery, and the fresh sperm were used in the IVF-ET cycles.\n[SUBTITLE] The processing of ovulation, fertilization and embryo transfer were as follows [SUBSECTION] controlled ovarian hyperstimulation (COH) used a gonadotropin-releasing hormone agonist protocol (the following was a shortened version of the long protocol) or a GnRH antagonist protocol, both of which are effective in blocking a premature LH surge [15, 16]. Generally, for the long protocol, a GnRH agonist (Triptorelin Acetate, Ipsen Pharma Biotech, France) was administered by subcutaneous injection daily starting from the luteal phase of the menstrual cycle for 10–14 days, and then ovarian stimulation with rFSH (Gn-F, Merck Serono SA Aubonne Branch, Switzerland) commenced. For the GnRH antagonist protocol, ovarian stimulation began on the second day of the menstrual cycle, and on the fifth day, antagonist (Cetrorelix Acetate, Cetrotide, Serono Labs Inc., Switzerland) administration started. Once the leading two follicles reached 18 mm or larger in diameter, the hCG administration was ejected as a trigger on the same day. Thirty-five to forty hours later, a doctor punctured the follicles and collected the eggs with the guidance of an ultrasound instrument. The sperm and oocytes (10000:1) were added to 0.1 ml of prebalanced embryo culture medium into four-well plates covered with mineral oil. The evaluation of embryo quality was performed on the 3rd day after fertilization. The embryos were divided into four levels according to the characteristics of blastomeres, such as the number, the morphology and fragments. Grade I and Grade II embryos were defined as high-quality embryos, and the other grades were defined as low-quality embryos [17, 18]. When the number of high-quality embryos was two or lower and the woman was younger than 35 years old, the embryos were frozen for transfer. All transferred embryos were 4-cell embryos. The frozen embryos were thawed according to the rapid recovery method of vitrification. Embryos with a recovery rate above 50% could be used for transfer. After embryo transfer, the patients were followed up until birth.\ncontrolled ovarian hyperstimulation (COH) used a gonadotropin-releasing hormone agonist protocol (the following was a shortened version of the long protocol) or a GnRH antagonist protocol, both of which are effective in blocking a premature LH surge [15, 16]. Generally, for the long protocol, a GnRH agonist (Triptorelin Acetate, Ipsen Pharma Biotech, France) was administered by subcutaneous injection daily starting from the luteal phase of the menstrual cycle for 10–14 days, and then ovarian stimulation with rFSH (Gn-F, Merck Serono SA Aubonne Branch, Switzerland) commenced. For the GnRH antagonist protocol, ovarian stimulation began on the second day of the menstrual cycle, and on the fifth day, antagonist (Cetrorelix Acetate, Cetrotide, Serono Labs Inc., Switzerland) administration started. Once the leading two follicles reached 18 mm or larger in diameter, the hCG administration was ejected as a trigger on the same day. Thirty-five to forty hours later, a doctor punctured the follicles and collected the eggs with the guidance of an ultrasound instrument. The sperm and oocytes (10000:1) were added to 0.1 ml of prebalanced embryo culture medium into four-well plates covered with mineral oil. The evaluation of embryo quality was performed on the 3rd day after fertilization. The embryos were divided into four levels according to the characteristics of blastomeres, such as the number, the morphology and fragments. Grade I and Grade II embryos were defined as high-quality embryos, and the other grades were defined as low-quality embryos [17, 18]. When the number of high-quality embryos was two or lower and the woman was younger than 35 years old, the embryos were frozen for transfer. All transferred embryos were 4-cell embryos. The frozen embryos were thawed according to the rapid recovery method of vitrification. Embryos with a recovery rate above 50% could be used for transfer. After embryo transfer, the patients were followed up until birth.\n[SUBTITLE] Grouping and statistical analysis [SUBSECTION] According to the treatment of embryo transfer, Group IVF/D was divided into the D/ET subgroup (treated with frozen donor sperm and Fresh embryo transfer) and the D/FET subgroup (treated with frozen donor sperm and frozen embryo transfer). Group IVF/H was also divided into the H/ET subgroup (treated with Fresh husband sperm and Fresh embryo transfer) and the H/FET subgroup (treated with Fresh husband sperm and frozen embryo transfer). The detail of the research grouping flow chat is shown in Fig. 1.\n\nFig. 1The research grouping flow chat\n\nThe research grouping flow chat\nThe female-related factors, including age, GnRH injection days and dosage, the estrogen secretion peak of egg collection, endometrium thickness, the number of eggs retrieved, the number of fertilizations, the number of effective embryos, the number of high-quality embryos, etc., were followed up and analyzed by SPSS 19.0 software (SPSS Inc., Cambridge, MA, USA). First, all the parameters were checked by a normal distribution test (Kolmogorov–Smirnov model and Shapiro–Wilk model). Then, the medians (first quartile, third quartile) of the continuous parameters were calculated. Finally, the variation in clinical outcomes within or between the IVF/D or IVF/H groups was compared using the nonparametric test (Kruskal–Wallis model) or chi square test for independent samples (p < 0.05).\nAccording to the treatment of embryo transfer, Group IVF/D was divided into the D/ET subgroup (treated with frozen donor sperm and Fresh embryo transfer) and the D/FET subgroup (treated with frozen donor sperm and frozen embryo transfer). Group IVF/H was also divided into the H/ET subgroup (treated with Fresh husband sperm and Fresh embryo transfer) and the H/FET subgroup (treated with Fresh husband sperm and frozen embryo transfer). The detail of the research grouping flow chat is shown in Fig. 1.\n\nFig. 1The research grouping flow chat\n\nThe research grouping flow chat\nThe female-related factors, including age, GnRH injection days and dosage, the estrogen secretion peak of egg collection, endometrium thickness, the number of eggs retrieved, the number of fertilizations, the number of effective embryos, the number of high-quality embryos, etc., were followed up and analyzed by SPSS 19.0 software (SPSS Inc., Cambridge, MA, USA). First, all the parameters were checked by a normal distribution test (Kolmogorov–Smirnov model and Shapiro–Wilk model). Then, the medians (first quartile, third quartile) of the continuous parameters were calculated. Finally, the variation in clinical outcomes within or between the IVF/D or IVF/H groups was compared using the nonparametric test (Kruskal–Wallis model) or chi square test for independent samples (p < 0.05).", "controlled ovarian hyperstimulation (COH) used a gonadotropin-releasing hormone agonist protocol (the following was a shortened version of the long protocol) or a GnRH antagonist protocol, both of which are effective in blocking a premature LH surge [15, 16]. Generally, for the long protocol, a GnRH agonist (Triptorelin Acetate, Ipsen Pharma Biotech, France) was administered by subcutaneous injection daily starting from the luteal phase of the menstrual cycle for 10–14 days, and then ovarian stimulation with rFSH (Gn-F, Merck Serono SA Aubonne Branch, Switzerland) commenced. For the GnRH antagonist protocol, ovarian stimulation began on the second day of the menstrual cycle, and on the fifth day, antagonist (Cetrorelix Acetate, Cetrotide, Serono Labs Inc., Switzerland) administration started. Once the leading two follicles reached 18 mm or larger in diameter, the hCG administration was ejected as a trigger on the same day. Thirty-five to forty hours later, a doctor punctured the follicles and collected the eggs with the guidance of an ultrasound instrument. The sperm and oocytes (10000:1) were added to 0.1 ml of prebalanced embryo culture medium into four-well plates covered with mineral oil. The evaluation of embryo quality was performed on the 3rd day after fertilization. The embryos were divided into four levels according to the characteristics of blastomeres, such as the number, the morphology and fragments. Grade I and Grade II embryos were defined as high-quality embryos, and the other grades were defined as low-quality embryos [17, 18]. When the number of high-quality embryos was two or lower and the woman was younger than 35 years old, the embryos were frozen for transfer. All transferred embryos were 4-cell embryos. The frozen embryos were thawed according to the rapid recovery method of vitrification. Embryos with a recovery rate above 50% could be used for transfer. After embryo transfer, the patients were followed up until birth.", "According to the treatment of embryo transfer, Group IVF/D was divided into the D/ET subgroup (treated with frozen donor sperm and Fresh embryo transfer) and the D/FET subgroup (treated with frozen donor sperm and frozen embryo transfer). Group IVF/H was also divided into the H/ET subgroup (treated with Fresh husband sperm and Fresh embryo transfer) and the H/FET subgroup (treated with Fresh husband sperm and frozen embryo transfer). The detail of the research grouping flow chat is shown in Fig. 1.\n\nFig. 1The research grouping flow chat\n\nThe research grouping flow chat\nThe female-related factors, including age, GnRH injection days and dosage, the estrogen secretion peak of egg collection, endometrium thickness, the number of eggs retrieved, the number of fertilizations, the number of effective embryos, the number of high-quality embryos, etc., were followed up and analyzed by SPSS 19.0 software (SPSS Inc., Cambridge, MA, USA). First, all the parameters were checked by a normal distribution test (Kolmogorov–Smirnov model and Shapiro–Wilk model). Then, the medians (first quartile, third quartile) of the continuous parameters were calculated. Finally, the variation in clinical outcomes within or between the IVF/D or IVF/H groups was compared using the nonparametric test (Kruskal–Wallis model) or chi square test for independent samples (p < 0.05).", "[SUBTITLE] Participants and grouping [SUBSECTION] A total of 860 couples undergoing IVF-ET treatment were included in the analysis, including 573 couples with husband sperm (Group IVF/H) and 287 couples with frozen donor sperm (Group IVF/D). According to whether or not frozen embryo transfer was performed, the IVF/H group was divided into the H/ET subgroup (133 couples treated by IVF/H and fresh embryo transfer) and the H/FET subgroup (440 couples treated by IVF/H and frozen embryo transfer); moreover, the IVF/D group was also divided into the D/ET subgroup (49 couples treated by IVF/D and fresh embryo transfer) and the D/FET subgroup (238 couples treated by IVF/D and frozen embryo transfer) (detailed in Fig. 1).\nA total of 860 couples undergoing IVF-ET treatment were included in the analysis, including 573 couples with husband sperm (Group IVF/H) and 287 couples with frozen donor sperm (Group IVF/D). According to whether or not frozen embryo transfer was performed, the IVF/H group was divided into the H/ET subgroup (133 couples treated by IVF/H and fresh embryo transfer) and the H/FET subgroup (440 couples treated by IVF/H and frozen embryo transfer); moreover, the IVF/D group was also divided into the D/ET subgroup (49 couples treated by IVF/D and fresh embryo transfer) and the D/FET subgroup (238 couples treated by IVF/D and frozen embryo transfer) (detailed in Fig. 1).\n[SUBTITLE] The baseline characteristics of the couples [SUBSECTION] Except for the female BMI in the FN subgroup, the variables were nonnormally distributed among the other subgroups. Thus, using the nonparametric test (Kruskal–Wallis model), we found that the median endometrial thickness (10 cm vs. 9 cm, p = 0.015), oocytes retrieved (8 vs. 11, p = 0.000) and MII oocytes (6 vs. 10, p = 0.000) were differentiated within Group IVF/H, but we found that only female age (30 years vs. 28 years, p = 0.002) was differentiated within Group IVF/D. There were no significant differences (p > 0.05) within Group IVF/H or Group IVF/D, including the male age, semen parameters and female BMI (detailed in Table 1).\n\nTable 1The basic characteristics of the 860 couples in one IVF/H (or IVF/D) cycleBaseline ParametersGroup IVF/H\nP value\n(H/ET vs. H/FET)Group IVF/D\nP value\n(D/ET vs. D/FET)H/ET1 subgroupH/FET2 subgroupD/ET3 subgroupD/FET4 subgroupMedian [first quartile, third quartile]Median [first quartile, third quartile]Inclusion Samples (n)133440/49238/Male Age (husband or donor) a32 [29, 34]32 [29, 34]0.73123 [22, 26]23 [21, 27]0.967Semen Volume (Ml) a3 [2, 3]3 [3]0.3482 [2, 2]2 [2, 2]0.608Semen Concentration (10^6/ML) a56 [48, 64]54 [48, 62]0.73446 [43, 53]47 [43, 51]0.974Semen (PR + NP) (%)a56 [50, 60]54 [50, 60]0.93146 [43, 54]48 [43, 52]0.574Female Age a33 [30, 35]32 [30, 35]0.64430 [28, 35]28 [26, 32]0.002**Female BMI (kg/m2) a21 [24, 25]21 [26, 27]0.86222 [20, 23]21 [20, 24]0.862Endometrial Thickness (cm) a10 [8, 12]9 [7, 11]0.015*10 [8, 12]10 [8, 12]0.805Oocytes Retrieved (n) a8 [4, 11]11[8, 16]0.000***11 [8, 15]11 [8, 15]0.590MII Oocytes (n) a6 [4, 10]10 [6, 14]0.000***9 [7, 13]9 [6, 12]0.721MII Oocytes (%)a93 [78, 100]91 [82, 100]0.66081 [68, 96]86 [73, 96]0.503Note:1: H/ET subgroup = using fresh sperm and fresh embryo transfer;2: H/FET subgroup = using fresh sperm and frozen embryo transfer;3: D/ET subgroup = using frozen sperm and fresh embryo transfer;4: D/FET subgroup = using frozen sperm and frozen embryo transfer;a: Nonparametric test (Kruskal–Wallis Model)* p < 0.05 **p < 0.01 ***p < 0.001\n\nThe basic characteristics of the 860 couples in one IVF/H (or IVF/D) cycle\nNote:\n1: H/ET subgroup = using fresh sperm and fresh embryo transfer;\n2: H/FET subgroup = using fresh sperm and frozen embryo transfer;\n3: D/ET subgroup = using frozen sperm and fresh embryo transfer;\n4: D/FET subgroup = using frozen sperm and frozen embryo transfer;\na: Nonparametric test (Kruskal–Wallis Model)\n* p < 0.05 **p < 0.01 ***p < 0.001\nExcept for the female BMI in the FN subgroup, the variables were nonnormally distributed among the other subgroups. Thus, using the nonparametric test (Kruskal–Wallis model), we found that the median endometrial thickness (10 cm vs. 9 cm, p = 0.015), oocytes retrieved (8 vs. 11, p = 0.000) and MII oocytes (6 vs. 10, p = 0.000) were differentiated within Group IVF/H, but we found that only female age (30 years vs. 28 years, p = 0.002) was differentiated within Group IVF/D. There were no significant differences (p > 0.05) within Group IVF/H or Group IVF/D, including the male age, semen parameters and female BMI (detailed in Table 1).\n\nTable 1The basic characteristics of the 860 couples in one IVF/H (or IVF/D) cycleBaseline ParametersGroup IVF/H\nP value\n(H/ET vs. H/FET)Group IVF/D\nP value\n(D/ET vs. D/FET)H/ET1 subgroupH/FET2 subgroupD/ET3 subgroupD/FET4 subgroupMedian [first quartile, third quartile]Median [first quartile, third quartile]Inclusion Samples (n)133440/49238/Male Age (husband or donor) a32 [29, 34]32 [29, 34]0.73123 [22, 26]23 [21, 27]0.967Semen Volume (Ml) a3 [2, 3]3 [3]0.3482 [2, 2]2 [2, 2]0.608Semen Concentration (10^6/ML) a56 [48, 64]54 [48, 62]0.73446 [43, 53]47 [43, 51]0.974Semen (PR + NP) (%)a56 [50, 60]54 [50, 60]0.93146 [43, 54]48 [43, 52]0.574Female Age a33 [30, 35]32 [30, 35]0.64430 [28, 35]28 [26, 32]0.002**Female BMI (kg/m2) a21 [24, 25]21 [26, 27]0.86222 [20, 23]21 [20, 24]0.862Endometrial Thickness (cm) a10 [8, 12]9 [7, 11]0.015*10 [8, 12]10 [8, 12]0.805Oocytes Retrieved (n) a8 [4, 11]11[8, 16]0.000***11 [8, 15]11 [8, 15]0.590MII Oocytes (n) a6 [4, 10]10 [6, 14]0.000***9 [7, 13]9 [6, 12]0.721MII Oocytes (%)a93 [78, 100]91 [82, 100]0.66081 [68, 96]86 [73, 96]0.503Note:1: H/ET subgroup = using fresh sperm and fresh embryo transfer;2: H/FET subgroup = using fresh sperm and frozen embryo transfer;3: D/ET subgroup = using frozen sperm and fresh embryo transfer;4: D/FET subgroup = using frozen sperm and frozen embryo transfer;a: Nonparametric test (Kruskal–Wallis Model)* p < 0.05 **p < 0.01 ***p < 0.001\n\nThe basic characteristics of the 860 couples in one IVF/H (or IVF/D) cycle\nNote:\n1: H/ET subgroup = using fresh sperm and fresh embryo transfer;\n2: H/FET subgroup = using fresh sperm and frozen embryo transfer;\n3: D/ET subgroup = using frozen sperm and fresh embryo transfer;\n4: D/FET subgroup = using frozen sperm and frozen embryo transfer;\na: Nonparametric test (Kruskal–Wallis Model)\n* p < 0.05 **p < 0.01 ***p < 0.001\n[SUBTITLE] Maternal factor exposure to frozen embryo transfer: the comparison of clinical outcomes within group IVF/H or group IVF/D [SUBSECTION] Using the nonparametric test (Kruskal–Wallis Model), the outcomes of fertilization and embryo transfer were compared within Group IVF/H and Group IVF/D (see Table 2). Before embryo transfer, the median of all the fertilization outcomes in the H/FET subgroup was higher than that in the H/ET subgroup. The median [first quartile, third quartile] fertilization rate (%), cleavage rate (%) and high-quality embryo rate (%) were 80 [67, 91], 100 [80, 100] and 57 [36, 78], respectively, in the H/FET subgroup. In contrast, there was no significant difference between the D/ET and D/FET subgroups. After frozen embryo transfer, the CPR and LBR were significantly higher in the H/FET subgroup than in the fresh embryo treatment subgroup and the H/ET subgroup (63.2 vs. 39.8, p = 0.000; 61.1 vs. 39.1, p = 0.000; 14.8 vs. 6.7, p = 0.016, respectively). However, the LBW was significantly higher in the H/FET subgroup than in the fresh embryo treatment subgroup in the H/ET subgroup (16.2 vs. 4.9, p = 0.021). In another IVF/D group, the CPR and LBR were significantly higher in the D/FET subgroup than in the fresh embryo treatment subgroup in the D/ET subgroup (68.1 vs. 49.0, p = 0.017; 66.4 vs. 47.0, p = 0.010, respectively). Otherwise, the BDR was significantly higher in the D/FET subgroup than in the fresh embryo treatment group in subgroup D/ET (1.0 vs. 0.0, p = 0.000).\n\nTable 2The IVF outcomes of the 860 couples in one IVF/H (or IVF/D) cycleOutcome IndicatorsGroup IVF/H\n(Χ\n2\n)\n\nP value\n(H/ET vs. H/FET)Group IVF/D\n(Χ\n2\n)\n\nP value\n(D/ET vs. D/FET)H/ET1 subgroupH/FET2 subgroupD/ET3 subgroupD/FET4 subgroupMedian [first quartile, third quartile]Median [first quartile, third quartile]After FertilizationFertilization Number (n)a5 [3, 8]8 [5, 11]0.000***8 [6, 12]8 [5, 11]0.342Fertilization Rate (%)a71 [50,89]80 [67,91]0.001**67 [57,88]75 [57,84]0.822Cleavage Number (n) a4 [2, 8]8 [5, 11]0.000***8 [6, 11]7 [4, 11]0.309Cleavage Rate (%)a67 [50,88]100 [80,100]0.000***67 [57,88]71 [55,82]0.667High-Quality Embryos (n) a2 [1, 5]4 [3, 7]0.000***6 [3, 8]4 [2, 7]0.105High-Quality Embryos Rate (%)a33 [10,54]57 [36,78]0.000***52 [33,67]45 [28,60]0.070After TransferBiochemical Pregnancy Rate (%)b0.01.4(2.377) 0.1232.00.4(1.542) 0.214Clinical Pregnancy Rate (%)b39.863.2(22.789) 0.000***49.068.1(5.734) 0.017*Live Birth Rate (%)b39.161.1(20.134) 0.000***47.066.4(6.598) 0.010*Multipregnancy Rate (%)b6.714.8(5.820) 0.016*14.39.2(1.137) 0.286Miscarriage Rate (%)b0.82.0(0.997) 0.3182.02.1(0.001) 0.979Total Baby Sex Ratio (%)b69.4114.1(3.129) 0.077121.482.0(1.036) 0.309Low Birth Weight Rate (%)b4.916.2(5.286) 0.021*19.415.8(0.029) 0.806Baby with Birth Defect Rate (%)b1.61.5(0.007) 0.9330.01.0(216.712) 0.000***Note:1: H/ET subgroup = using fresh sperm and fresh embryo transfer;2: H/FET subgroup = using fresh sperm and frozen embryo transfer;3: D/ET subgroup = using frozen sperm and fresh embryo transfer;4: D/FET subgroup = using frozen sperm and frozen embryo transfer;a: Nonparametric Test (Kruskal–Wallis Model)b: chi square test for independent samples* p < 0.05 **p < 0.01 ***p < 0.001\n\nThe IVF outcomes of the 860 couples in one IVF/H (or IVF/D) cycle\nNote:\n1: H/ET subgroup = using fresh sperm and fresh embryo transfer;\n2: H/FET subgroup = using fresh sperm and frozen embryo transfer;\n3: D/ET subgroup = using frozen sperm and fresh embryo transfer;\n4: D/FET subgroup = using frozen sperm and frozen embryo transfer;\na: Nonparametric Test (Kruskal–Wallis Model)\nb: chi square test for independent samples\n* p < 0.05 **p < 0.01 ***p < 0.001\nUsing the nonparametric test (Kruskal–Wallis Model), the outcomes of fertilization and embryo transfer were compared within Group IVF/H and Group IVF/D (see Table 2). Before embryo transfer, the median of all the fertilization outcomes in the H/FET subgroup was higher than that in the H/ET subgroup. The median [first quartile, third quartile] fertilization rate (%), cleavage rate (%) and high-quality embryo rate (%) were 80 [67, 91], 100 [80, 100] and 57 [36, 78], respectively, in the H/FET subgroup. In contrast, there was no significant difference between the D/ET and D/FET subgroups. After frozen embryo transfer, the CPR and LBR were significantly higher in the H/FET subgroup than in the fresh embryo treatment subgroup and the H/ET subgroup (63.2 vs. 39.8, p = 0.000; 61.1 vs. 39.1, p = 0.000; 14.8 vs. 6.7, p = 0.016, respectively). However, the LBW was significantly higher in the H/FET subgroup than in the fresh embryo treatment subgroup in the H/ET subgroup (16.2 vs. 4.9, p = 0.021). In another IVF/D group, the CPR and LBR were significantly higher in the D/FET subgroup than in the fresh embryo treatment subgroup in the D/ET subgroup (68.1 vs. 49.0, p = 0.017; 66.4 vs. 47.0, p = 0.010, respectively). Otherwise, the BDR was significantly higher in the D/FET subgroup than in the fresh embryo treatment group in subgroup D/ET (1.0 vs. 0.0, p = 0.000).\n\nTable 2The IVF outcomes of the 860 couples in one IVF/H (or IVF/D) cycleOutcome IndicatorsGroup IVF/H\n(Χ\n2\n)\n\nP value\n(H/ET vs. H/FET)Group IVF/D\n(Χ\n2\n)\n\nP value\n(D/ET vs. D/FET)H/ET1 subgroupH/FET2 subgroupD/ET3 subgroupD/FET4 subgroupMedian [first quartile, third quartile]Median [first quartile, third quartile]After FertilizationFertilization Number (n)a5 [3, 8]8 [5, 11]0.000***8 [6, 12]8 [5, 11]0.342Fertilization Rate (%)a71 [50,89]80 [67,91]0.001**67 [57,88]75 [57,84]0.822Cleavage Number (n) a4 [2, 8]8 [5, 11]0.000***8 [6, 11]7 [4, 11]0.309Cleavage Rate (%)a67 [50,88]100 [80,100]0.000***67 [57,88]71 [55,82]0.667High-Quality Embryos (n) a2 [1, 5]4 [3, 7]0.000***6 [3, 8]4 [2, 7]0.105High-Quality Embryos Rate (%)a33 [10,54]57 [36,78]0.000***52 [33,67]45 [28,60]0.070After TransferBiochemical Pregnancy Rate (%)b0.01.4(2.377) 0.1232.00.4(1.542) 0.214Clinical Pregnancy Rate (%)b39.863.2(22.789) 0.000***49.068.1(5.734) 0.017*Live Birth Rate (%)b39.161.1(20.134) 0.000***47.066.4(6.598) 0.010*Multipregnancy Rate (%)b6.714.8(5.820) 0.016*14.39.2(1.137) 0.286Miscarriage Rate (%)b0.82.0(0.997) 0.3182.02.1(0.001) 0.979Total Baby Sex Ratio (%)b69.4114.1(3.129) 0.077121.482.0(1.036) 0.309Low Birth Weight Rate (%)b4.916.2(5.286) 0.021*19.415.8(0.029) 0.806Baby with Birth Defect Rate (%)b1.61.5(0.007) 0.9330.01.0(216.712) 0.000***Note:1: H/ET subgroup = using fresh sperm and fresh embryo transfer;2: H/FET subgroup = using fresh sperm and frozen embryo transfer;3: D/ET subgroup = using frozen sperm and fresh embryo transfer;4: D/FET subgroup = using frozen sperm and frozen embryo transfer;a: Nonparametric Test (Kruskal–Wallis Model)b: chi square test for independent samples* p < 0.05 **p < 0.01 ***p < 0.001\n\nThe IVF outcomes of the 860 couples in one IVF/H (or IVF/D) cycle\nNote:\n1: H/ET subgroup = using fresh sperm and fresh embryo transfer;\n2: H/FET subgroup = using fresh sperm and frozen embryo transfer;\n3: D/ET subgroup = using frozen sperm and fresh embryo transfer;\n4: D/FET subgroup = using frozen sperm and frozen embryo transfer;\na: Nonparametric Test (Kruskal–Wallis Model)\nb: chi square test for independent samples\n* p < 0.05 **p < 0.01 ***p < 0.001\n[SUBTITLE] Paternal factor exposure to frozen sperm fertilization: the comparison of clinical outcomes between Group IVF/H and Group IVF/D [SUBSECTION] Using the chi square test for independent samples, the comparison of clinical outcomes between Group IVF/H and Group IVF/D is shown in Figs. 2 and 3. Compared with the D/ET subgroup using frozen sperm (donor sperm) fertilization, the HER was higher than that of the H/ET subgroup using fresh husband sperm (p < 0.05). In contrast, the values of the FER, CLR and HER in the D/FET subgroup were lower than those of the H/FET subgroup using fresh husband sperm (all p < 0.05). After embryo transfer, the BDR in the D/ET subgroup was lower than that of the H/ET subgroup using fresh husband sperm (p < 0.001). Meanwhile, the MUR in the D/FET subgroup was lower than that of the H/FET subgroup using fresh husband sperm (p < 0.05).\n\nFig. 2The comparison of fertilization outcomes between Group IVF/H and Group IVF/D. The values of the FER, CLR and HER in subgroups H/ET and H/FET are shown in the separated blue histogram and those of the D/ET and D/FET subgroups are shown in the red histogram. Note: FER = fertilization rate; CLR = cleavage rate; HER = high-quality embryo rate; * p < 0.05 **p < 0.01 ***p < 0.001\n\nThe comparison of fertilization outcomes between Group IVF/H and Group IVF/D. The values of the FER, CLR and HER in subgroups H/ET and H/FET are shown in the separated blue histogram and those of the D/ET and D/FET subgroups are shown in the red histogram. Note: FER = fertilization rate; CLR = cleavage rate; HER = high-quality embryo rate; * p < 0.05 **p < 0.01 ***p < 0.001\n\nFig. 3The comparison of embryo transfer outcomes between Group IVF/H and Group IVF/D. The proportions of the BPR, CPR, LBR, MUR, MSR, TBSR, LBW and BDR in the H/ET and H/FET subgroups are shown in the combined blue histogram and those of the D/ET or D/FET subgroups are shown in the combined red histogram. Note: BPR = biochemical pregnancy rate; CPR = clinical pregnancy rate; LBR = live birth rate; MUR = multipregnancy rate; MSR = miscarriage rate; TBSR = total baby sex ratio; LBW = low birth weight rate; BDR = baby with birth defect rate;* p < 0.05 **p < 0.01 ***p < 0.001\n\nThe comparison of embryo transfer outcomes between Group IVF/H and Group IVF/D. The proportions of the BPR, CPR, LBR, MUR, MSR, TBSR, LBW and BDR in the H/ET and H/FET subgroups are shown in the combined blue histogram and those of the D/ET or D/FET subgroups are shown in the combined red histogram. Note: BPR = biochemical pregnancy rate; CPR = clinical pregnancy rate; LBR = live birth rate; MUR = multipregnancy rate; MSR = miscarriage rate; TBSR = total baby sex ratio; LBW = low birth weight rate; BDR = baby with birth defect rate;* p < 0.05 **p < 0.01 ***p < 0.001\nUsing the chi square test for independent samples, the comparison of clinical outcomes between Group IVF/H and Group IVF/D is shown in Figs. 2 and 3. Compared with the D/ET subgroup using frozen sperm (donor sperm) fertilization, the HER was higher than that of the H/ET subgroup using fresh husband sperm (p < 0.05). In contrast, the values of the FER, CLR and HER in the D/FET subgroup were lower than those of the H/FET subgroup using fresh husband sperm (all p < 0.05). After embryo transfer, the BDR in the D/ET subgroup was lower than that of the H/ET subgroup using fresh husband sperm (p < 0.001). Meanwhile, the MUR in the D/FET subgroup was lower than that of the H/FET subgroup using fresh husband sperm (p < 0.05).\n\nFig. 2The comparison of fertilization outcomes between Group IVF/H and Group IVF/D. The values of the FER, CLR and HER in subgroups H/ET and H/FET are shown in the separated blue histogram and those of the D/ET and D/FET subgroups are shown in the red histogram. Note: FER = fertilization rate; CLR = cleavage rate; HER = high-quality embryo rate; * p < 0.05 **p < 0.01 ***p < 0.001\n\nThe comparison of fertilization outcomes between Group IVF/H and Group IVF/D. The values of the FER, CLR and HER in subgroups H/ET and H/FET are shown in the separated blue histogram and those of the D/ET and D/FET subgroups are shown in the red histogram. Note: FER = fertilization rate; CLR = cleavage rate; HER = high-quality embryo rate; * p < 0.05 **p < 0.01 ***p < 0.001\n\nFig. 3The comparison of embryo transfer outcomes between Group IVF/H and Group IVF/D. The proportions of the BPR, CPR, LBR, MUR, MSR, TBSR, LBW and BDR in the H/ET and H/FET subgroups are shown in the combined blue histogram and those of the D/ET or D/FET subgroups are shown in the combined red histogram. Note: BPR = biochemical pregnancy rate; CPR = clinical pregnancy rate; LBR = live birth rate; MUR = multipregnancy rate; MSR = miscarriage rate; TBSR = total baby sex ratio; LBW = low birth weight rate; BDR = baby with birth defect rate;* p < 0.05 **p < 0.01 ***p < 0.001\n\nThe comparison of embryo transfer outcomes between Group IVF/H and Group IVF/D. The proportions of the BPR, CPR, LBR, MUR, MSR, TBSR, LBW and BDR in the H/ET and H/FET subgroups are shown in the combined blue histogram and those of the D/ET or D/FET subgroups are shown in the combined red histogram. Note: BPR = biochemical pregnancy rate; CPR = clinical pregnancy rate; LBR = live birth rate; MUR = multipregnancy rate; MSR = miscarriage rate; TBSR = total baby sex ratio; LBW = low birth weight rate; BDR = baby with birth defect rate;* p < 0.05 **p < 0.01 ***p < 0.001\n[SUBTITLE] Parental factors exposure to frozen sperm fertilization and frozen embryo transfer: the comparison of clinical outcomes between Group IVF/H and Group IVF/D [SUBSECTION] When both frozen sperm and frozen embryo transfer were used in the D/FET group, the HER was higher than that for the use of both fresh sperm and fresh embryo transfer in the H/ET group (p < 0.05). After embryo transfer, the CPR and LBR values in the D/FET subgroup were higher than those in the H/ET group (p < 0.001). Meanwhile, the LBW in the D/FET subgroup was higher than that in the H/ET subgroup (p < 0.05).\nWhen both frozen sperm and frozen embryo transfer were used in the D/FET group, the HER was higher than that for the use of both fresh sperm and fresh embryo transfer in the H/ET group (p < 0.05). After embryo transfer, the CPR and LBR values in the D/FET subgroup were higher than those in the H/ET group (p < 0.001). Meanwhile, the LBW in the D/FET subgroup was higher than that in the H/ET subgroup (p < 0.05).", "A total of 860 couples undergoing IVF-ET treatment were included in the analysis, including 573 couples with husband sperm (Group IVF/H) and 287 couples with frozen donor sperm (Group IVF/D). According to whether or not frozen embryo transfer was performed, the IVF/H group was divided into the H/ET subgroup (133 couples treated by IVF/H and fresh embryo transfer) and the H/FET subgroup (440 couples treated by IVF/H and frozen embryo transfer); moreover, the IVF/D group was also divided into the D/ET subgroup (49 couples treated by IVF/D and fresh embryo transfer) and the D/FET subgroup (238 couples treated by IVF/D and frozen embryo transfer) (detailed in Fig. 1).", "Except for the female BMI in the FN subgroup, the variables were nonnormally distributed among the other subgroups. Thus, using the nonparametric test (Kruskal–Wallis model), we found that the median endometrial thickness (10 cm vs. 9 cm, p = 0.015), oocytes retrieved (8 vs. 11, p = 0.000) and MII oocytes (6 vs. 10, p = 0.000) were differentiated within Group IVF/H, but we found that only female age (30 years vs. 28 years, p = 0.002) was differentiated within Group IVF/D. There were no significant differences (p > 0.05) within Group IVF/H or Group IVF/D, including the male age, semen parameters and female BMI (detailed in Table 1).\n\nTable 1The basic characteristics of the 860 couples in one IVF/H (or IVF/D) cycleBaseline ParametersGroup IVF/H\nP value\n(H/ET vs. H/FET)Group IVF/D\nP value\n(D/ET vs. D/FET)H/ET1 subgroupH/FET2 subgroupD/ET3 subgroupD/FET4 subgroupMedian [first quartile, third quartile]Median [first quartile, third quartile]Inclusion Samples (n)133440/49238/Male Age (husband or donor) a32 [29, 34]32 [29, 34]0.73123 [22, 26]23 [21, 27]0.967Semen Volume (Ml) a3 [2, 3]3 [3]0.3482 [2, 2]2 [2, 2]0.608Semen Concentration (10^6/ML) a56 [48, 64]54 [48, 62]0.73446 [43, 53]47 [43, 51]0.974Semen (PR + NP) (%)a56 [50, 60]54 [50, 60]0.93146 [43, 54]48 [43, 52]0.574Female Age a33 [30, 35]32 [30, 35]0.64430 [28, 35]28 [26, 32]0.002**Female BMI (kg/m2) a21 [24, 25]21 [26, 27]0.86222 [20, 23]21 [20, 24]0.862Endometrial Thickness (cm) a10 [8, 12]9 [7, 11]0.015*10 [8, 12]10 [8, 12]0.805Oocytes Retrieved (n) a8 [4, 11]11[8, 16]0.000***11 [8, 15]11 [8, 15]0.590MII Oocytes (n) a6 [4, 10]10 [6, 14]0.000***9 [7, 13]9 [6, 12]0.721MII Oocytes (%)a93 [78, 100]91 [82, 100]0.66081 [68, 96]86 [73, 96]0.503Note:1: H/ET subgroup = using fresh sperm and fresh embryo transfer;2: H/FET subgroup = using fresh sperm and frozen embryo transfer;3: D/ET subgroup = using frozen sperm and fresh embryo transfer;4: D/FET subgroup = using frozen sperm and frozen embryo transfer;a: Nonparametric test (Kruskal–Wallis Model)* p < 0.05 **p < 0.01 ***p < 0.001\n\nThe basic characteristics of the 860 couples in one IVF/H (or IVF/D) cycle\nNote:\n1: H/ET subgroup = using fresh sperm and fresh embryo transfer;\n2: H/FET subgroup = using fresh sperm and frozen embryo transfer;\n3: D/ET subgroup = using frozen sperm and fresh embryo transfer;\n4: D/FET subgroup = using frozen sperm and frozen embryo transfer;\na: Nonparametric test (Kruskal–Wallis Model)\n* p < 0.05 **p < 0.01 ***p < 0.001", "Using the nonparametric test (Kruskal–Wallis Model), the outcomes of fertilization and embryo transfer were compared within Group IVF/H and Group IVF/D (see Table 2). Before embryo transfer, the median of all the fertilization outcomes in the H/FET subgroup was higher than that in the H/ET subgroup. The median [first quartile, third quartile] fertilization rate (%), cleavage rate (%) and high-quality embryo rate (%) were 80 [67, 91], 100 [80, 100] and 57 [36, 78], respectively, in the H/FET subgroup. In contrast, there was no significant difference between the D/ET and D/FET subgroups. After frozen embryo transfer, the CPR and LBR were significantly higher in the H/FET subgroup than in the fresh embryo treatment subgroup and the H/ET subgroup (63.2 vs. 39.8, p = 0.000; 61.1 vs. 39.1, p = 0.000; 14.8 vs. 6.7, p = 0.016, respectively). However, the LBW was significantly higher in the H/FET subgroup than in the fresh embryo treatment subgroup in the H/ET subgroup (16.2 vs. 4.9, p = 0.021). In another IVF/D group, the CPR and LBR were significantly higher in the D/FET subgroup than in the fresh embryo treatment subgroup in the D/ET subgroup (68.1 vs. 49.0, p = 0.017; 66.4 vs. 47.0, p = 0.010, respectively). Otherwise, the BDR was significantly higher in the D/FET subgroup than in the fresh embryo treatment group in subgroup D/ET (1.0 vs. 0.0, p = 0.000).\n\nTable 2The IVF outcomes of the 860 couples in one IVF/H (or IVF/D) cycleOutcome IndicatorsGroup IVF/H\n(Χ\n2\n)\n\nP value\n(H/ET vs. H/FET)Group IVF/D\n(Χ\n2\n)\n\nP value\n(D/ET vs. D/FET)H/ET1 subgroupH/FET2 subgroupD/ET3 subgroupD/FET4 subgroupMedian [first quartile, third quartile]Median [first quartile, third quartile]After FertilizationFertilization Number (n)a5 [3, 8]8 [5, 11]0.000***8 [6, 12]8 [5, 11]0.342Fertilization Rate (%)a71 [50,89]80 [67,91]0.001**67 [57,88]75 [57,84]0.822Cleavage Number (n) a4 [2, 8]8 [5, 11]0.000***8 [6, 11]7 [4, 11]0.309Cleavage Rate (%)a67 [50,88]100 [80,100]0.000***67 [57,88]71 [55,82]0.667High-Quality Embryos (n) a2 [1, 5]4 [3, 7]0.000***6 [3, 8]4 [2, 7]0.105High-Quality Embryos Rate (%)a33 [10,54]57 [36,78]0.000***52 [33,67]45 [28,60]0.070After TransferBiochemical Pregnancy Rate (%)b0.01.4(2.377) 0.1232.00.4(1.542) 0.214Clinical Pregnancy Rate (%)b39.863.2(22.789) 0.000***49.068.1(5.734) 0.017*Live Birth Rate (%)b39.161.1(20.134) 0.000***47.066.4(6.598) 0.010*Multipregnancy Rate (%)b6.714.8(5.820) 0.016*14.39.2(1.137) 0.286Miscarriage Rate (%)b0.82.0(0.997) 0.3182.02.1(0.001) 0.979Total Baby Sex Ratio (%)b69.4114.1(3.129) 0.077121.482.0(1.036) 0.309Low Birth Weight Rate (%)b4.916.2(5.286) 0.021*19.415.8(0.029) 0.806Baby with Birth Defect Rate (%)b1.61.5(0.007) 0.9330.01.0(216.712) 0.000***Note:1: H/ET subgroup = using fresh sperm and fresh embryo transfer;2: H/FET subgroup = using fresh sperm and frozen embryo transfer;3: D/ET subgroup = using frozen sperm and fresh embryo transfer;4: D/FET subgroup = using frozen sperm and frozen embryo transfer;a: Nonparametric Test (Kruskal–Wallis Model)b: chi square test for independent samples* p < 0.05 **p < 0.01 ***p < 0.001\n\nThe IVF outcomes of the 860 couples in one IVF/H (or IVF/D) cycle\nNote:\n1: H/ET subgroup = using fresh sperm and fresh embryo transfer;\n2: H/FET subgroup = using fresh sperm and frozen embryo transfer;\n3: D/ET subgroup = using frozen sperm and fresh embryo transfer;\n4: D/FET subgroup = using frozen sperm and frozen embryo transfer;\na: Nonparametric Test (Kruskal–Wallis Model)\nb: chi square test for independent samples\n* p < 0.05 **p < 0.01 ***p < 0.001", "Using the chi square test for independent samples, the comparison of clinical outcomes between Group IVF/H and Group IVF/D is shown in Figs. 2 and 3. Compared with the D/ET subgroup using frozen sperm (donor sperm) fertilization, the HER was higher than that of the H/ET subgroup using fresh husband sperm (p < 0.05). In contrast, the values of the FER, CLR and HER in the D/FET subgroup were lower than those of the H/FET subgroup using fresh husband sperm (all p < 0.05). After embryo transfer, the BDR in the D/ET subgroup was lower than that of the H/ET subgroup using fresh husband sperm (p < 0.001). Meanwhile, the MUR in the D/FET subgroup was lower than that of the H/FET subgroup using fresh husband sperm (p < 0.05).\n\nFig. 2The comparison of fertilization outcomes between Group IVF/H and Group IVF/D. The values of the FER, CLR and HER in subgroups H/ET and H/FET are shown in the separated blue histogram and those of the D/ET and D/FET subgroups are shown in the red histogram. Note: FER = fertilization rate; CLR = cleavage rate; HER = high-quality embryo rate; * p < 0.05 **p < 0.01 ***p < 0.001\n\nThe comparison of fertilization outcomes between Group IVF/H and Group IVF/D. The values of the FER, CLR and HER in subgroups H/ET and H/FET are shown in the separated blue histogram and those of the D/ET and D/FET subgroups are shown in the red histogram. Note: FER = fertilization rate; CLR = cleavage rate; HER = high-quality embryo rate; * p < 0.05 **p < 0.01 ***p < 0.001\n\nFig. 3The comparison of embryo transfer outcomes between Group IVF/H and Group IVF/D. The proportions of the BPR, CPR, LBR, MUR, MSR, TBSR, LBW and BDR in the H/ET and H/FET subgroups are shown in the combined blue histogram and those of the D/ET or D/FET subgroups are shown in the combined red histogram. Note: BPR = biochemical pregnancy rate; CPR = clinical pregnancy rate; LBR = live birth rate; MUR = multipregnancy rate; MSR = miscarriage rate; TBSR = total baby sex ratio; LBW = low birth weight rate; BDR = baby with birth defect rate;* p < 0.05 **p < 0.01 ***p < 0.001\n\nThe comparison of embryo transfer outcomes between Group IVF/H and Group IVF/D. The proportions of the BPR, CPR, LBR, MUR, MSR, TBSR, LBW and BDR in the H/ET and H/FET subgroups are shown in the combined blue histogram and those of the D/ET or D/FET subgroups are shown in the combined red histogram. Note: BPR = biochemical pregnancy rate; CPR = clinical pregnancy rate; LBR = live birth rate; MUR = multipregnancy rate; MSR = miscarriage rate; TBSR = total baby sex ratio; LBW = low birth weight rate; BDR = baby with birth defect rate;* p < 0.05 **p < 0.01 ***p < 0.001", "When both frozen sperm and frozen embryo transfer were used in the D/FET group, the HER was higher than that for the use of both fresh sperm and fresh embryo transfer in the H/ET group (p < 0.05). After embryo transfer, the CPR and LBR values in the D/FET subgroup were higher than those in the H/ET group (p < 0.001). Meanwhile, the LBW in the D/FET subgroup was higher than that in the H/ET subgroup (p < 0.05).", "The results of our study indicated that using frozen sperm or frozen embryo transfer have different effects on the different IVF stages, frozen sperm mainly increases fertilization rate and reduces birth defects, while cryopreservation of embryos increases pregnancy rate respectly. For example, in terms of the single factor comparison, the frozen embryo transfer method was more conductive to CPR and LBR, of which increased to 63.2% and 61.1% after using frozen embryo transfer in the H/FET subgroup within IVF/H group. The same results of IVF outcomes were also being found within IVF/D group. In recent years, there has been an accelerating trend toward the use of frozen embryo transfer. Several studies focusing on similar frozen strategies have been conducted in recent years[8, 9, 21, 24, 26]. Recently, Qianqian Z et al. collected a larger general population than other reports to explore the superiority of the freeze-embryo strategy in all IVF/H patients. The live birth rate (LBR) after the first complete IVF cycle was 50.74% in the freeze-embryo strategy. For women who were younger than 31 years old, the LBR of that treatment cycle was 63.81% (95% CI: 62.80–64.80%). The LBR of our results, after using frozen embryo transfer in the H/FET subgroup, was 61.1% (the median age of the treated women was 32 years old). Therefore, our report was extremely close to that of Qianqian Z et al. Although the clinical pregnancy rates and live birth rates after cryopreservation are now meta-analyzed to be close to or even higher than those of fresh cycles[7, 9, 10], singletons born after FET have a higher risk of LBW[19]. In our data, the LBW (including singletons, twins and more) was significantly higher in the H/FET subgroup than in the H/ET subgroup receiving embryo treatment (16.2 vs. 4.9, respectively, p = 0.021).\nOther wisely, the frozen sperm fertilization method had an advantage of the HER and BDR, of which increased to 52% and reduced to 0%, only in subgroup D/ET comparing to subgroup H/ET. The differentiated results of IVF outcomes were found in subgroup D/FET comparing to subgroup H/FET. Limited research has been performed on outcomes from IVF treatments with frozen donor sperm [5, 6, 25]. The French Sperm Bank network covers 22 centers of sperm cryopreservation working under the same rules: the CECOS. In this prospective study, 3689 pregnancies achieved after IVF with frozen donor spermatozoa (IVF/D) were followed and reported to the central CECOS between 1987 and 1994. In the prospective CECOS study, the miscarriage rate (MSR) was 21.5%, the multipregnancy rate (MUR) was 29% (including that of twins, triplets and more), the low birth weight rate (LBW) was 4.7%, and the baby with birth defect rate (BDR) was 2.7%. In our study, most of the neonatal characteristics achieved after IVF/D were better, showing a lower MSR (2-2.1%) and MUR (9.2–14.3%) and a higher BDR (0.0–1.0%). The gaps between these two studies are due to the younger donors (the median age was 23 years old) or the rapid development of IVF technology itself in these decades, i.e., using frozen embryo transfer to avoid the deleterious effects of controlled ovarian stimulation on the endometrium [8, 9, 27]. Controversially, the frozen spermatozoa are from donors with normal semen parameters whereas the fresh spermatozoa are from the men in the couples undergoing IVF. Although we improved the inclusion criteria for fresh spermatozoa with normal standard semen parameters. We still cannot excluded impaired sperm genome or proteomic quality by which the couples were diagnosed as infertility [20, 28].\nInterestingly, we first found that the exposure to both frozen sperm and embryo treatment had a complementary effect comparing to the use of fresh sperm and embryos. The combined data led to increases in the HER (p < 0.05), CPR (p < 0.001), LBR (p < 0.001) and LBW (p < 0.05) after the completion of IVF treatment. The value of HER could derived from the paternal effects of frozen sperm, while the other three values (CPR, LBR and LBW) could derived from the maternal effects of frozen embryo transfer. This complementary effect may be explained by the physiological orientation of sperm and embryos. The sperm initiates the process of fertilization while the embryo plays key role in the pregnancy. Furthermore, the freezing of sperm acts as an artificial selection, eliminating the damage sperm (probably including the defect of acrosomes, sperm membranes or sperm DNA et al.,) [29–32]; and the cryopreservation of embryo is to keep the transplantation the same pace with the growth of the endometrium.\nOver the last 70 years, the cryobiology of reproductive cells (sperm and oocytes), embryos, blastomeres, and ovarian and testicular tissue has made rapid progress and has been widely used in human reproductive medicine [22]. Unfortunately, people are only concerned about the survival and viability of cells following the freezing and thawing processes, which could result in a live birth baby. However, little is known about the long-term development of newborns developed from paternal (or maternal) frozen gametes or even the genomic integrity of such frozen cells and tissues [23, 33]. More basic research on the mechanism of the cellular response adaptations to cryopreservation is needed in the future. Eventually, we may uncover some of the cellular defense mechanisms that make cryopreserved sperm/embryos more able to survive.", "This study demonstrates that using frozen sperm or frozen embryo transfer have distinguished effects on the different IVF stages. Especially, the use of frozen embryos and frozen sperm have complementary IVF outcomes. Basic research on the mechanism of the cellular response adaptations to cryopreservation are needed to support our findings." ]
[ "introduction", null, null, null, "results", null, null, null, null, null, "discussion", "conclusion" ]
[ "Cryopreservation", "IVF outcome", "Sperm", "Embryo", "Accumulative effect" ]
Do coagulation or fibrinolysis reflect the disease condition in patients with soft tissue sarcoma?
36258189
Coagulation and fibrinolysis are distinct processes that are highly correlated. Cells control coagulation and fibrinolysis by expression of tissue factor and urokinase-type plasminogen activator receptor on their surface. Tumor cells express these proteins, adjust their microenvironment and induce tumor exacerbation. We hypothesized that the expression of plasma markers for coagulation and fibrinolysis in patients with soft tissue sarcomas (STSs) was dependent on the level of tumor malignancy. To elucidate which markers are predictive of recurrence, metastasis and prognosis, coagulation or fibrinolysis, we analyzed the correlation between plasma levels of thrombin-antithrombin III complex (TAT), soluble fibrin (SF), plasmin-α2 plasmin inhibitor complex (PIC), D-dimer (DD) and clinical parameters in patients with STSs.
BACKGROUND
TAT, SF, PIC or DD were measured in pre-treatment blood samples from 64 patients with primary STSs and analyzed with clinicopathological parameters, and 5-year recurrence free survival (RFS), 5-year metastasis free survival (MFS) and 5-year overall survival (OS) were evaluated.
METHODS
The metastasis group had significantly higher DD (p = 0.0394), PIC (p = 0.00532) and SF (p = 0.00249) concentrations than the group without metastasis. The group that died of disease showed significantly higher DD (p = 0.00105), PIC (p = 0.000542), SF (p = 0.000126) and TAT (p = 0.0373) than surviving patients. By dividing the patients into low and high groups, the group with high DD, PIC, SF and TAT showed significantly lower 5-year MFS and 5-year OS than the corresponding low group. Furthermore, in multivariate COX proportional hazard analysis of continuous variables for 5-year MFS, only PIC was found to be a significant factor (HR: 2.14).
RESULTS
Fibrinolysis was better than coagulation at reflecting the disease condition of patients with STS. Notably, PIC levels ≥ 1.1 can not only predict the risk of metastasis and poor prognosis, but also increasing PIC levels correspond to further increases in risks of metastasis and poor prognosis.
CONCLUSION
[ "Humans", "Fibrinolysis", "Fibrinolysin", "Receptors, Urokinase Plasminogen Activator", "Antifibrinolytic Agents", "Thromboplastin", "Peptide Hydrolases", "Biomarkers", "Sarcoma", "Tumor Microenvironment" ]
9580209
null
null
null
null
Results
[SUBTITLE] Patient and tumor characteristics [SUBSECTION] A total of 64 patients with primary STSs visiting Mie University Hospital from 2012 to 2014 were included in this study. The average age of the patients was 63 years (range: 17–89 years), and the average tumor size was 12.8 cm (range: 3.5–31 cm). Histopathological diagnosis was as follows: 28 liposarcomas (17 well-differentiated liposarcomas, 6 dedifferentiated liposarcomas, and 5 myxoid liposarcomas), 10 undifferentiated pleomorphic sarcomas, 9 myxofibrosarcomas, 4 leiomyosarcomas, 3 synovial sarcomas, 3 malignant peripheral nerve sheath tumors, and 7 others in STS. Patients with STS received wide resection (44 patients), marginal resection (17 patients) and ion beam radiotherapy (3 patients). The average follow-up on patients with STS was 43.4 months (range: 0.6–75.6 months). A total of 64 patients with primary STSs visiting Mie University Hospital from 2012 to 2014 were included in this study. The average age of the patients was 63 years (range: 17–89 years), and the average tumor size was 12.8 cm (range: 3.5–31 cm). Histopathological diagnosis was as follows: 28 liposarcomas (17 well-differentiated liposarcomas, 6 dedifferentiated liposarcomas, and 5 myxoid liposarcomas), 10 undifferentiated pleomorphic sarcomas, 9 myxofibrosarcomas, 4 leiomyosarcomas, 3 synovial sarcomas, 3 malignant peripheral nerve sheath tumors, and 7 others in STS. Patients with STS received wide resection (44 patients), marginal resection (17 patients) and ion beam radiotherapy (3 patients). The average follow-up on patients with STS was 43.4 months (range: 0.6–75.6 months).
Conclusion
Serum levels of DD ≥ 0.74, PIC ≥ 1.1, SF ≥ 1.6 and TAT ≥ 1.71 were useful as makers for predicting future metastasis and poor prognosis. Moreover, fibrinolysis was better than coagulation at reflecting the disease condition of patients with STS. Notably, PIC levels ≥ 1.1 can not only predict the risk of metastasis and poor prognosis, but also increasing PIC levels correspond to further increases in risks of metastasis and poor prognosis. Our study had the following limitations. This study was retrospective and the number of patients was small. Statistical analysis could not be performed by each disease subtype, because STSs are rare entity and including many subtypes. Many studies have needed to analyze STS as a group, rather than by each histological classification. The information of concomitant pharmacological treatments were not included in the statistical analysis.
[ "Background", "Methods", "Statistical analysis", "Patient and tumor characteristics", "Characteristics of soft tissue sarcoma", "DD, PIC, SF and TAT levels in recurrence, metastasis and DOD in STS", "COX proportional hazard analysis in STS using continuous variables", "Determination of threshold by ROC analysis for identifying 5-year DOD", "Kaplan-Meier analysis in STS", "COX proportional hazard analysis of STS with binary DD, PIC, SF and TAT variables", "" ]
[ "Coagulation and fibrinolysis are distinct processes that are highly correlated. Fibrin production and degradation are important phenomena in vascular disease and injuries. The expression of tissue factor (TF), the primary initiator of extrinsic coagulation cascade, leads to the generation of thrombin and fibrin. Similarly, expression of urokinase-type plasminogen activator receptor (uPAR) on the cell surface results in the formation of a complex with urokinase-type plasminogen activator (uPA), converts plasminogen into plasmin, and leads to fibrin degradation. Cells can control their microenvironment by adjusting the expression of TF and uPAR. In particular, tumor cells overexpress these factors and alter their microenvironment, a process that is deeply involved in tumor exacerbation.\nTF expression is typically observed in endothelium, leukocytes and monocytes. Many tumor cells express TF, including lung cancer, pancreatic cancer, prostate cancer, laryngeal carcinoma, glioma, ovarian cancer, breast cancer and osteosarcoma cells [1–8]. Additionally, circulating TF-positive extracellular vesicles have been observed in lung cancer, breast cancer, pancreatic cancer and leukemia. [9–12]. These various sources of TF expression are powerful triggers for thrombin generation. TF expression is associated with poor prognosis in several malignant tumors [7, 13, 14]. TF is a potential molecular target for cancer therapy and TF inhibitors have been shown to suppress tumor exacerbation in vivo [15]. Following TF expression, the generated thrombin stimulates tumor cell adhesion to platelets, endothelial cells, extracellular matrix proteins, as well as tumor cell mitogenesis and invasion [16–19]. Furthermore, thrombin can induce secretion of various cytokines and proteases. Notably, thrombin stimulates VEGF expression in tumor and endothelial cells, and is involved in angiogenesis [20–23]. The biological activity of thrombin is mediated by the thrombin receptor protease-activated receptor-1 (PAR-1) [24], and PAR-1 is a promising molecular target for cancer treatment [25]. Thus, activation of coagulation leads to exacerbation of malignant tumors.\nIn contrast, uPAR is usually expressed in endothelial cells [26], fibroblasts [27, 28], neutrophils and monocytes [29]. Moreover, a variety of malignant cells expresses uPAR, such as breast cancer, gastric cancer, lung cancer and sarcoma cells [30–35]. uPAR-expressing tumor cells can activate oncogenic pathways such as MAPK, RTK, ERK2 and FAK [36–38]. HIF-1 increase in cancer cells lead to upregulation of uPA/uPAR [39]. Plasmin generated by uPA causes a variety of proteolytic events, including fibrin degradation. Plasmin activates pro-MMPs and cleaves extracellular matrix components [40]. Overexpression of uPAR in breast cancer enhances tumor invasion, growth and metastasis [41]. Furthermore, uPAR expression is related to prognosis in oral cancer [42]. Given this, activation of fibrinolysis leads to exacerbation of malignant tumors.\nCoagulation and fibrinolytic state can be assessed using several markers, including thrombin-antithrombin III complex (TAT), soluble fibrin (SF), prothrombin fragment 1 + 2, plasmin-α2 plasmin inhibitor complex (PIC), D-dimer (DD) and fibrin degradation products (FDP). Of these markers, TAT, SF, PIC and DD were evaluated in this study. After thrombin generation by activated coagulation cascade, a proportion of thrombin forms a complex with antithrombin III (TAT) [43]. Fibrin monomer generated by thrombin forms a complex with fibrinogen (SF) [44]. On the other hand, Fibrinolytic state can be assessed by fibrinolysis markers, such as PIC or DD. After plasmin is generated from plasminogen, a proportion of the plasmin forms a complex with α2 plasmin inhibitor (PIC) [45]. DD is a degradation product of cross-linked D fragments of fibrin, generated from proteolysis by plasmin [46]. DD levels are indicative of plasmin-induced fibrinolysis after fibrin formation [47].\nSTSs are categorized as rare cancers, and the relationship between STS and coagulation and fibrinolysis is sparsely reported. We previously reported increased PIC levels were the most suitable for the detection of STS from soft tissue tumors [48]. Based on the findings described above, we hypothesized that plasma markers of coagulation and fibrinolysis in patients with STS is affected by sarcoma tissues and is dependent on its malignancy. We analyzed the correlation between plasma levels of TAT, SF, PIC or DD and clinical parameters in patients with STS to elucidate which markers predict the recurrence, metastasis and prognosis of patients with STS.", "Patients.\nA total of 64 patients with primary STSs who visited Mie University Hospital from 2012 to 2014 were enrolled in this study. Patients who had local recurrence or who were referred for additional resection after inadequate resection in a previous hospital, or had trauma, surgical treatment, thrombosis and disseminated intravascular coagulation (DIC) were excluded from this study. Histopathological diagnosis and the histological grade were verified based on the French Federation of Cancer Centers Sarcoma group system (FNCLCC) by independent pathologists. Patients were classified from stage I to IV according to the 7th edition of the American Joint Committee on Cancer (AJCC) classification of soft tissue sarcomas. Blood samples from all patients were collected in sodium citrate before biopsy or treatment.\nThe levels of TAT, SF, PIC or DD in plasma were quantitatively measured using a chemiluminescent enzyme immunoassay or latex photometric immunoassay. Written, informed consent was obtained from each patient. For patients below 19 years of age, informed consent was obtained from their parents or legal guardian. This study was approved by the Ethics Committee of the Mie University Graduate School of Medicine (approval number: 1310). All procedures performed in studies involving human participants were in accordance with the ethical standards of the Ethics Committee of Mie University and with the Helsinki declaration of 1975.\n[SUBTITLE] Statistical analysis [SUBSECTION] Clinicopathological analysis was performed to compare the plasma levels of TAT, SF, PIC or DD to various clinical parameters using the Mann-Whitney test or Kruskal Wallis test for quantitative data and Fisher’s exact test for qualitative data. To evaluate the threshold for detecting recurrence, metastasis or mortality due to disease, receiver operating characteristic (ROC) curves were generated. ROC curves were created by plotting the sensitivity on the y-axis and the false positive rate (1-specificity) on the x-axis. To measure the effectiveness of TAT, SF, PIC or DD levels as a marker for recurrence, metastasis or mortality due to disease, the area under the curve (AUC) was assessed. Local recurrence-free survival (RFS) was defined as the time from initial treatment to the date of clinically documented local recurrence. Metastasis-free survival (MFS) was defined as the time from initial treatment to the date of clinically documented distant metastasis. Overall survival (OS) was defined as the time from initial treatment to the date of mortality attributed to the neoplasm. Kaplan-Meier survival plots and log-rank tests were used to assess the differences in time to local recurrence, distant metastasis or overall survival. To adjust the imbalance in prognostic factors among patients, Cox proportional hazard analysis was used. p < 0.05 was considered statistically significant. The EZR software program was used for statistical analyses [49].\nClinicopathological analysis was performed to compare the plasma levels of TAT, SF, PIC or DD to various clinical parameters using the Mann-Whitney test or Kruskal Wallis test for quantitative data and Fisher’s exact test for qualitative data. To evaluate the threshold for detecting recurrence, metastasis or mortality due to disease, receiver operating characteristic (ROC) curves were generated. ROC curves were created by plotting the sensitivity on the y-axis and the false positive rate (1-specificity) on the x-axis. To measure the effectiveness of TAT, SF, PIC or DD levels as a marker for recurrence, metastasis or mortality due to disease, the area under the curve (AUC) was assessed. Local recurrence-free survival (RFS) was defined as the time from initial treatment to the date of clinically documented local recurrence. Metastasis-free survival (MFS) was defined as the time from initial treatment to the date of clinically documented distant metastasis. Overall survival (OS) was defined as the time from initial treatment to the date of mortality attributed to the neoplasm. Kaplan-Meier survival plots and log-rank tests were used to assess the differences in time to local recurrence, distant metastasis or overall survival. To adjust the imbalance in prognostic factors among patients, Cox proportional hazard analysis was used. p < 0.05 was considered statistically significant. The EZR software program was used for statistical analyses [49].", "Clinicopathological analysis was performed to compare the plasma levels of TAT, SF, PIC or DD to various clinical parameters using the Mann-Whitney test or Kruskal Wallis test for quantitative data and Fisher’s exact test for qualitative data. To evaluate the threshold for detecting recurrence, metastasis or mortality due to disease, receiver operating characteristic (ROC) curves were generated. ROC curves were created by plotting the sensitivity on the y-axis and the false positive rate (1-specificity) on the x-axis. To measure the effectiveness of TAT, SF, PIC or DD levels as a marker for recurrence, metastasis or mortality due to disease, the area under the curve (AUC) was assessed. Local recurrence-free survival (RFS) was defined as the time from initial treatment to the date of clinically documented local recurrence. Metastasis-free survival (MFS) was defined as the time from initial treatment to the date of clinically documented distant metastasis. Overall survival (OS) was defined as the time from initial treatment to the date of mortality attributed to the neoplasm. Kaplan-Meier survival plots and log-rank tests were used to assess the differences in time to local recurrence, distant metastasis or overall survival. To adjust the imbalance in prognostic factors among patients, Cox proportional hazard analysis was used. p < 0.05 was considered statistically significant. The EZR software program was used for statistical analyses [49].", "A total of 64 patients with primary STSs visiting Mie University Hospital from 2012 to 2014 were included in this study. The average age of the patients was 63 years (range: 17–89 years), and the average tumor size was 12.8 cm (range: 3.5–31 cm). Histopathological diagnosis was as follows: 28 liposarcomas (17 well-differentiated liposarcomas, 6 dedifferentiated liposarcomas, and 5 myxoid liposarcomas), 10 undifferentiated pleomorphic sarcomas, 9 myxofibrosarcomas, 4 leiomyosarcomas, 3 synovial sarcomas, 3 malignant peripheral nerve sheath tumors, and 7 others in STS. Patients with STS received wide resection (44 patients), marginal resection (17 patients) and ion beam radiotherapy (3 patients). The average follow-up on patients with STS was 43.4 months (range: 0.6–75.6 months).", "Plasma STS, DD, PIC, SF and TAT values were compared according to sex, age, tumor size, location, tumor depth, FNCLCC grade, AJCC stage and treatment. Patients ≥ 60 years of age showed significantly higher DD, PIC, SF and TAT than those under 60. Patients with tumors ≥ 10 cm in size showed significantly higher DD than those with tumors < 10 cm. In evaluating FNCLCC grade, DD and SF were significantly different depending on FNCLCC grade. However, the average DD, SF and TAT levels of grade 1 were higher than in grade 2 (Table 1; Fig. 1). DD and SF were found to significantly vary according to AJCC stage. However, the lowest levels of DD and SF were observed in stage 2. PIC levels were observed to increase according to stage, despite a lack of statistical significance (Table 1; Fig. 2).\n\nTable 1Plasma levels of DD, PIC, SF and TAT according to various characteristics of patients with STS.Characteristics of STSN (64)DD AVE (SD)PIC AVE (SD)SF AVE (SD)TAT AVE (SD)SexMale331.086 (1.639)1.000 (0.910)2.990 (6.804)1.090 (0.813)Female310.904 (1.579)1.093 (0.689)5.783 (13.11)2.988 (6.432)Age (years old)< 6027\n*0.616 (1.403)\n\n*0.781 (0.802)\n\n*1.500 (2.281)\n\n*1.002 (0.789)\n≥ 6037\n*1.276 (1.694)\n\n*1.237 (0.762)\n\n*6.418 (13.18)\n\n*2.744 (5.908)\nTumor size< 1029\n*0.792 (1.434)\n1.062 (1.063)2.217 (3.053)1.112 (0.859)(cm)≥ 1035\n*1.169 (1.728)\n1.031 (0.521)6.105 (13.57)2.753 (6.079)LocationExtremity350.946 (1.617)0.934 (0.600)4.065 (9.540)1.446 (1.989)Trunk291.060 (1.605)1.179 (0.994)4.679 (11.44)2.689 (6.439)Tumor depthSuperficial200.907 (1.654)1.235 (1.066)7.455 (16.04)2.409 (4.192)Deep441.039 (1.593)0.959 (0.652)2.929 (6.096)1.827 (4.777)FNCLCCGrade 119\n#\n0.673 (1.124)\n0.784 (0.365)\n#\n3.957 (14.00)\n1.631 (3.683)Grade 222\n#\n0.534 (0.587)\n0.940 (0.681)\n#\n1.940 (2.683)\n1.124 (0.816)Grade 323\n#\n1.710 (2.266)\n1.360 (1.065)\n#\n6.960 (11.28)\n3.168 (6.779)AJCC stageI19\n#\n0.673 (1.124)\n0.784 (0.365)\n#\n3.957 (14.00)\n1.631 (3.683)II22\n#\n0.534 (0.587)\n0.940 (0.681)\n#\n1.940 (2.683)\n1.124 (0.816)III18\n#\n1.571 (2.124)\n1.233 (0.845)\n#\n7.805 (12.56)\n3.744 (7.598)IV5\n#\n2.212 (2.943)\n1.820 (1.693)\n#\n3.920 (3.784)\n1.096 (0.713)TreatmentWide resection441.184 (1.767)1.181 (0.915)\n#\n4.581 (8.657)\n2.181 (4.991)Marginal resection170.650 (1.165)0.776 (0.366)\n#\n4.347 (14.80)\n1.717 (3.895)Ion beam radiotherapy:30.243 (0.198)0.566 (0.152)\n#\n0.833 (0.611)\n1.140 (0.680)Chemotherapy-481.115 (1.759)1.162 (0.894)4.466 (10.81)2.286 (5.250)+160.648 (0.942)0.693 (0.211)3.975 (9.206)1.178 (0.807)Radiotherapy-520.775 (0.955)0.998 (0.694)3.990 (10.34)2.134 (5.050)+121.961 (3.032)1.250 (1.194)5.875 (10.76)1.468 (1.162)Average (AVE) values and standard deviation (SD) of DD, PIC, SF and TAT are shown according to each listed parameter. * and # indicate significant differences using the Mann-Whitney test (p < 0.05) and the Kruskal-Wallis test (P < 0.05), respectively\n\nPlasma levels of DD, PIC, SF and TAT according to various characteristics of patients with STS.\nAverage (AVE) values and standard deviation (SD) of DD, PIC, SF and TAT are shown according to each listed parameter. * and # indicate significant differences using the Mann-Whitney test (p < 0.05) and the Kruskal-Wallis test (P < 0.05), respectively\n\nFig. 1BOX plot of plasma DD, PIC, SF and TAT levels according to FNCLCC grade. G1, G2 and G3 correspond to FNCLCC grades 1, 2 and 3. Statistical analysis was performed by the Kruskal-Wallis test\n\nBOX plot of plasma DD, PIC, SF and TAT levels according to FNCLCC grade. G1, G2 and G3 correspond to FNCLCC grades 1, 2 and 3. Statistical analysis was performed by the Kruskal-Wallis test\n\nFig. 2BOX plot of plasma DD, PIC, SF and TAT levels according to AJCC stage. I, II, III and IV correspond to AJCC stages I, II, III and IV. Statistical analysis was performed by the Kruskal-Wallis test\n\nBOX plot of plasma DD, PIC, SF and TAT levels according to AJCC stage. I, II, III and IV correspond to AJCC stages I, II, III and IV. Statistical analysis was performed by the Kruskal-Wallis test", "During the period of this study, 7 patients developed recurrence, 21 patients developed metastasis (metastasis group), and 17 patients died of disease (DOD group). No significant differences were observed in DD, PIC, SF and TAT between patients with and without recurrence. The metastasis group had significantly higher plasma DD (p = 0.0394), PIC (p = 0.00532) and SF (p = 0.00249) levels than the group without metastasis. The DOD group showed significantly higher DD (p = 0.00105), PIC (p = 0.000542), SF (p = 0.000126) and TAT (p = 0.0373) compared to the surviving patients (Fig. 3).\n\nFig. 3BOX plot of plasma DD, PIC, SF and TAT levels according to clinical outcome. Rec: recurrence, meta: metastasis, DOD: died of disease. Statistical analysis was performed by the Mann-Whitney test (p < 0.05) \n\nBOX plot of plasma DD, PIC, SF and TAT levels according to clinical outcome. Rec: recurrence, meta: metastasis, DOD: died of disease. Statistical analysis was performed by the Mann-Whitney test (p < 0.05) ", "Univariate COX proportional hazard analysis was performed using DD, PIC, SF and TAT as continuous variable. For RFS, only stage III/IV was significantly different (HR: 6.20, 95%CI: 1.19–32.1, p = 0.0298). For MFS, DD (HR: 1.26, 95%CI: 1.07–1.50, p = 0.00612), PIC (HR: 2.39, 95%CI: 1.59–3.58, p = 0.000022) and stage III/IV (HR: 3.34, 95%CI: 1.40–7.96, p = 0.006494) showed significant differences. For OS, DD (HR: 1.33, 95%CI: 1.11–1.58, p = 0.001362), PIC (HR: 1.99, 95%CI: 1.42–2.79, p = 0.000059), SF (HR: 1.02, 95%CI: 1.01–1.05, p = 0.026) and stage III/IV (HR: 5.66, 95%CI: 1.98–16.1, p = 0.001164) demonstrated significant differences (Table 2).\n\nTable 2Univariate COX proportional hazard analysisRFSMFSOSHR95%CIp-valueHR95%CIp-valueHR95%CIp-valueMale4.9 × 108/0.9980.780.33–1.840.5730.670.24–1.860.447Age ≥ 600.590.13–2.650.4951.450.58–3.610.4233.320.09–11.70.063Size ≥ 10 cm0.840.18–3.780.8280.980.41–2.310.9651.660.59–4.670.335Superficial0.250.03–2.110.2040.880.35–2.180.7871.110.39–3.150.830Trunk2.490.55–11.20.2331.710.72–4.050.2182.090.75–5.810.154DD1.110.77–1.590.5621.261.07–1.50\n0.0061\n1.331.11–1.58\n0.0014\nPIC1.260.60–2.650.5342.391.59–3.58\n0.00003\n1.991.42–2.79\n0.00006\nSF1.020.98–1.070.2191.020.99–1.040.0961.021.01–1.05\n0.026\nTAT0.950.71–1.270.7360.980.89–1.090.8291.010.92–1.100.855High grade0.0250.0-16.20.26439.30.79–19570.06567.20.76–51.30.087Stage I, II vs. III, IV6.201.19-32.1-\n0.029\n3.341.40–7.96\n0.007\n5.661.98–16.1\n0.0011\nPlasma DD, PIC, SF and TAT levels were analyzed as continuous variables\n\nUnivariate COX proportional hazard analysis\nPlasma DD, PIC, SF and TAT levels were analyzed as continuous variables\nMultivariate COX proportional hazard analysis was used to adjust for the imbalance in prognostic factors among patients. Since only stage III/IV showed a significant difference in RFS, multivariate analysis was not performed. DD, PIC and stage III/IV were adopted for multivariate analysis of MFS, with only PIC exhibiting a significant difference (HR: 2.14, 95% CI: 1.26–3.62, p = 0.00477). DD, PIC, SF and stage III/IV were adopted for multivariate analysis of OS, with only stage III, IV exhibiting a significant difference (HR: 4.54, 95% CI: 1.46-14.0, p = 0.00872). PIC showed a risk of poor prognosis, but there was no significant difference (HR: 1.75, 95%CI: 0.96–3.19, p = 0.0655) (Table 3).\n\nTable 3Multivariate COX proportional hazard analysisMFSOSHR95%CIp-valueHR95%CIp-valueDD0.990.77–1.270.961DD0.930.65–1.310.692PIC2.141.26–3.62\n0.0048\nPIC1.750.96–3.190.065Stage III, IV2.380.94–6.060.067SF1.0280.99–1.060.108Stage I, II vs. III, IV4.541.46-14.0\n0.0087\nPlasma DD, PIC and SF levels were analyzed as continuous variables\n\nMultivariate COX proportional hazard analysis\nPlasma DD, PIC and SF levels were analyzed as continuous variables\nFor further prognostic analysis, patients with STS of FNCLCC grade 1 or with distant metastasis at initial diagnosis (stage IV) were excluded. Univariate COX proportional hazard analysis indicated that only SF was significantly different in RFS (HR: 1.08, 95%CI: 1.01–1.16, p = 0.013). For MFS, significant differences were detected in DD (HR: 1.22, 95%CI: 1.00-1.49, p = 0.049), PIC (HR: 2.65, 95%CI: 1.53–4.59, p = 0.00051) and SF (HR: 1.07, 95%CI: 1.03–1.11, p = 0.00054); whereas for OS, significant differences were detected in DD (HR: 1.35, 95%CI: 1.10–1.67, p = 0.0043), PIC (HR: 2.45, 95%CI: 1.47–4.08, p = 0.00057) and SF (HR: 1.07, 95%CI: 1.03–1.11, p = 0.00028) (Supplementary Table 1). DD, PIC and SF were adopted for multivariate analysis. PIC (HR: 2.37, 95%CI: 1.31–4.29, p = 0.004) and SF (HR: 1.06, 95%CI: 1.01–1.12, p = 0.013) exhibited significant differences in MFS, while PIC (HR: 2.13, 95%CI: 1.21–3.72, p = 0.0079) and SF (HR: 1.05, 95%CI: 1.00-1.11, p = 0.025) were significantly different in OS (Supplementary Table 2).", "ROC analysis was performed and UAC was evaluated to determine the diagnostic accuracy for identifying DOD within 5-years in STS. ROC analysis of DD produced an AUC of 0.77 (95% confidence interval (CI): 0.635–0.906). With a threshold of 0.74, the sensitivity and specificity for identifying 5-year DOD were 70.6% and 80.9%, respectively. ROC analysis of PIC produced an AUC of 0.784 (95% CI: 0.651–0.917). With a threshold of 1.1, the sensitivity and specificity for identifying 5-year DOD were 64.7% and 83.0%, respectively. ROC analysis of SF produced an AUC of 0.815 (95% CI: 0.694–0.935). With a threshold of 1.6, the sensitivity and specificity for identifying 5-year DOD were 76.5% and 78.7%, respectively. ROC analysis of TAT produced an AUC of 0.672 (95% CI: 0.509–0.835). With a threshold of 1.71, the sensitivity and specificity for identifying 5-year DOD were 47.1% and 89.4%, respectively. To divide patients into two groups for further analysis, thresholds of 0.74 (DD), 1.1 (PIC), 1.6 (SF) and 1.71 (TAT) were adopted as the Youden index, and the low and high groups for each threshold were analyzed. (Fig. 4).\n\nFig. 4ROC analysis. ROC analysis of DD, PIC, SF and TAT to determine diagnostic accuracy of detecting DOD in sarcoma patients. AUC: area under the curve. Thresholds were determined by the Youden index\n\nROC analysis. ROC analysis of DD, PIC, SF and TAT to determine diagnostic accuracy of detecting DOD in sarcoma patients. AUC: area under the curve. Thresholds were determined by the Youden index", "DD (low < 0.74 ≤ high), PIC (low < 1.1 ≤ high), SF (low < 1.6 ≤ high) and TAT (low < 1.71 ≤ high) were divided into two groups. Five-year recurrence-free survival (RFS), metastasis-free survival (MFS), and over-all survival (OS) between the low- and high-DD, PIC, SF and TAT groups were compared using Kaplan-Meier analysis and the log-rank tests. RFS showed no significant difference in DD (low DD 88.1%, high DD 87.4%, p = 0.809), PIC (low PIC 87.9%, high PIC 87.7%, p = 0.855), SF (low SF 89.8%, high SF 82.6%, p = 0.456) or TAT (low TAT 89.4%, high TAT 77.9%, p = 0.267). MFS showed significant differences in DD (low DD 75.4%, high DD 42.9%, p = 0.000628), PIC (low PIC 73.6%, high PIC 42.1%, p = 0.00164), SF (low SF 73.5%, high SF 47.8%, p = 0.0047) and TAT (low TAT 70.4%, high TAT 38.5%, p = 0.00556). OS and MFS showed significant differences in DD (low DD 87.7%, high DD 41.6%, p = 0.00000601), PIC (low PIC 86.1%, high PIC 38.3%, p = 0.0000516), SF (low SF 89.8%, high TM 41.0%, p = 0.00000925) and TAT (low TM 81.6%, high TM 35.9%, p = 0.000143) (Fig. 5). Thus, high levels of DD, PIC, SF and TAT appear to be important factors involved in metastatic potential and lead to poor prognosis.\n\nFig. 5Kaplan-Meier analysis. Kaplan-Meier analysis was performed according to the low and high expression sub-groups for each marker (DD, PIC, SF, and TAT). RFS indicates recurrence-free survival. MFS indicates metastasis-free survival. OS indicates over-all survival. Significance was analyzed using the log-rank test\n\nKaplan-Meier analysis. Kaplan-Meier analysis was performed according to the low and high expression sub-groups for each marker (DD, PIC, SF, and TAT). RFS indicates recurrence-free survival. MFS indicates metastasis-free survival. OS indicates over-all survival. Significance was analyzed using the log-rank test", "The low and high DD, PIC, SF and TAT groups were analyzed using univariate COX proportional analysis. For RFS, no significant differences were observed between the low and high groups for DD, PIC, SF and TAT. In contrast, analysis of MFS and OS revealed significant differences between the low and high groups for DD, PIC, SF and TAT (Table 4). Significance was mutually nullified by inclusion of high DD, high PIC, high SF and high TAT in multivariate analysis, thus separate analyses were performed with stage III/IV. High DD, high PIC and high SF showed significant differences in MFS and all high groups showed significant difference in OS (Table 5).\n\nTable 4Univariate COX proportional hazard analysisRFSMFSOSHR95%CIp-valueHR95%CIp-value95%CIp-valueHigh DD0.960.18–4.990.9643.581.48–8.66\n0.0045\n11.43.16-41.0\n0.0002\nHigh PIC1.030.19–5.330.9693.161.38–7.93\n0.0071\n8.192.57–25.7\n0.0004\nHigh SF0.730.14–3.800.7132.591.08–6.20\n0.032\n8.412.35–30.1\n0.0011\nHigh TAT1.750.33–9.140.5052.711.11–6.64\n0.028\n5.801.53–12.4\n0.0056\nPlasma DD, PIC, SF and TAT levels were divided into low and high groups by their respective thresholds (Youden index). COX analysis was performed using binary variables\n\nUnivariate COX proportional hazard analysis\nPlasma DD, PIC, SF and TAT levels were divided into low and high groups by their respective thresholds (Youden index). COX analysis was performed using binary variables\n\nTable 5Multivariate COX proportional hazard analysisMFSOSHR95%CIp-value95%CIp-valueHigh DD3.051.2–7.7\n0.019\n11.33.16-41.0\n0.0002\nStage I, II vs. III, IV2.310.91–5.830.0765.661.98–16.1\n0.0012\nHigh PIC2.901.19–7.03\n0.019\n8.192.57–25.7\n0.0004\nStage I, II vs. III, IV2.661.08–6.50\n0.032\n5.661.98–16.1\n0.0012\nHigh SF2.211.08–6.20\n0.032\n8.412.35–30.1\n0.0011\nStage I, II vs. III, IV2.330.87–6.190.0885.661.98–16.1\n0.0012\nHigh TAT2.711.11–6.640.1115.801.53–12.40\n0.0056\nStage I, II vs. III, IV3.341.40–7.96\n0.0065\n5.661.98–16.1\n0.0012\nPlasma DD, PIC, SF and TAT levels were analyzed as continuous variables. From Table 3, stage III/IV had significant difference and was adopted this analysis\n\nMultivariate COX proportional hazard analysis\nPlasma DD, PIC, SF and TAT levels were analyzed as continuous variables. From Table 3, stage III/IV had significant difference and was adopted this analysis", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Statistical analysis", "Results", "Patient and tumor characteristics", "Characteristics of soft tissue sarcoma", "DD, PIC, SF and TAT levels in recurrence, metastasis and DOD in STS", "COX proportional hazard analysis in STS using continuous variables", "Determination of threshold by ROC analysis for identifying 5-year DOD", "Kaplan-Meier analysis in STS", "COX proportional hazard analysis of STS with binary DD, PIC, SF and TAT variables", "Discussion", "Conclusion", "Electronic supplementary material", "" ]
[ "Coagulation and fibrinolysis are distinct processes that are highly correlated. Fibrin production and degradation are important phenomena in vascular disease and injuries. The expression of tissue factor (TF), the primary initiator of extrinsic coagulation cascade, leads to the generation of thrombin and fibrin. Similarly, expression of urokinase-type plasminogen activator receptor (uPAR) on the cell surface results in the formation of a complex with urokinase-type plasminogen activator (uPA), converts plasminogen into plasmin, and leads to fibrin degradation. Cells can control their microenvironment by adjusting the expression of TF and uPAR. In particular, tumor cells overexpress these factors and alter their microenvironment, a process that is deeply involved in tumor exacerbation.\nTF expression is typically observed in endothelium, leukocytes and monocytes. Many tumor cells express TF, including lung cancer, pancreatic cancer, prostate cancer, laryngeal carcinoma, glioma, ovarian cancer, breast cancer and osteosarcoma cells [1–8]. Additionally, circulating TF-positive extracellular vesicles have been observed in lung cancer, breast cancer, pancreatic cancer and leukemia. [9–12]. These various sources of TF expression are powerful triggers for thrombin generation. TF expression is associated with poor prognosis in several malignant tumors [7, 13, 14]. TF is a potential molecular target for cancer therapy and TF inhibitors have been shown to suppress tumor exacerbation in vivo [15]. Following TF expression, the generated thrombin stimulates tumor cell adhesion to platelets, endothelial cells, extracellular matrix proteins, as well as tumor cell mitogenesis and invasion [16–19]. Furthermore, thrombin can induce secretion of various cytokines and proteases. Notably, thrombin stimulates VEGF expression in tumor and endothelial cells, and is involved in angiogenesis [20–23]. The biological activity of thrombin is mediated by the thrombin receptor protease-activated receptor-1 (PAR-1) [24], and PAR-1 is a promising molecular target for cancer treatment [25]. Thus, activation of coagulation leads to exacerbation of malignant tumors.\nIn contrast, uPAR is usually expressed in endothelial cells [26], fibroblasts [27, 28], neutrophils and monocytes [29]. Moreover, a variety of malignant cells expresses uPAR, such as breast cancer, gastric cancer, lung cancer and sarcoma cells [30–35]. uPAR-expressing tumor cells can activate oncogenic pathways such as MAPK, RTK, ERK2 and FAK [36–38]. HIF-1 increase in cancer cells lead to upregulation of uPA/uPAR [39]. Plasmin generated by uPA causes a variety of proteolytic events, including fibrin degradation. Plasmin activates pro-MMPs and cleaves extracellular matrix components [40]. Overexpression of uPAR in breast cancer enhances tumor invasion, growth and metastasis [41]. Furthermore, uPAR expression is related to prognosis in oral cancer [42]. Given this, activation of fibrinolysis leads to exacerbation of malignant tumors.\nCoagulation and fibrinolytic state can be assessed using several markers, including thrombin-antithrombin III complex (TAT), soluble fibrin (SF), prothrombin fragment 1 + 2, plasmin-α2 plasmin inhibitor complex (PIC), D-dimer (DD) and fibrin degradation products (FDP). Of these markers, TAT, SF, PIC and DD were evaluated in this study. After thrombin generation by activated coagulation cascade, a proportion of thrombin forms a complex with antithrombin III (TAT) [43]. Fibrin monomer generated by thrombin forms a complex with fibrinogen (SF) [44]. On the other hand, Fibrinolytic state can be assessed by fibrinolysis markers, such as PIC or DD. After plasmin is generated from plasminogen, a proportion of the plasmin forms a complex with α2 plasmin inhibitor (PIC) [45]. DD is a degradation product of cross-linked D fragments of fibrin, generated from proteolysis by plasmin [46]. DD levels are indicative of plasmin-induced fibrinolysis after fibrin formation [47].\nSTSs are categorized as rare cancers, and the relationship between STS and coagulation and fibrinolysis is sparsely reported. We previously reported increased PIC levels were the most suitable for the detection of STS from soft tissue tumors [48]. Based on the findings described above, we hypothesized that plasma markers of coagulation and fibrinolysis in patients with STS is affected by sarcoma tissues and is dependent on its malignancy. We analyzed the correlation between plasma levels of TAT, SF, PIC or DD and clinical parameters in patients with STS to elucidate which markers predict the recurrence, metastasis and prognosis of patients with STS.", "Patients.\nA total of 64 patients with primary STSs who visited Mie University Hospital from 2012 to 2014 were enrolled in this study. Patients who had local recurrence or who were referred for additional resection after inadequate resection in a previous hospital, or had trauma, surgical treatment, thrombosis and disseminated intravascular coagulation (DIC) were excluded from this study. Histopathological diagnosis and the histological grade were verified based on the French Federation of Cancer Centers Sarcoma group system (FNCLCC) by independent pathologists. Patients were classified from stage I to IV according to the 7th edition of the American Joint Committee on Cancer (AJCC) classification of soft tissue sarcomas. Blood samples from all patients were collected in sodium citrate before biopsy or treatment.\nThe levels of TAT, SF, PIC or DD in plasma were quantitatively measured using a chemiluminescent enzyme immunoassay or latex photometric immunoassay. Written, informed consent was obtained from each patient. For patients below 19 years of age, informed consent was obtained from their parents or legal guardian. This study was approved by the Ethics Committee of the Mie University Graduate School of Medicine (approval number: 1310). All procedures performed in studies involving human participants were in accordance with the ethical standards of the Ethics Committee of Mie University and with the Helsinki declaration of 1975.\n[SUBTITLE] Statistical analysis [SUBSECTION] Clinicopathological analysis was performed to compare the plasma levels of TAT, SF, PIC or DD to various clinical parameters using the Mann-Whitney test or Kruskal Wallis test for quantitative data and Fisher’s exact test for qualitative data. To evaluate the threshold for detecting recurrence, metastasis or mortality due to disease, receiver operating characteristic (ROC) curves were generated. ROC curves were created by plotting the sensitivity on the y-axis and the false positive rate (1-specificity) on the x-axis. To measure the effectiveness of TAT, SF, PIC or DD levels as a marker for recurrence, metastasis or mortality due to disease, the area under the curve (AUC) was assessed. Local recurrence-free survival (RFS) was defined as the time from initial treatment to the date of clinically documented local recurrence. Metastasis-free survival (MFS) was defined as the time from initial treatment to the date of clinically documented distant metastasis. Overall survival (OS) was defined as the time from initial treatment to the date of mortality attributed to the neoplasm. Kaplan-Meier survival plots and log-rank tests were used to assess the differences in time to local recurrence, distant metastasis or overall survival. To adjust the imbalance in prognostic factors among patients, Cox proportional hazard analysis was used. p < 0.05 was considered statistically significant. The EZR software program was used for statistical analyses [49].\nClinicopathological analysis was performed to compare the plasma levels of TAT, SF, PIC or DD to various clinical parameters using the Mann-Whitney test or Kruskal Wallis test for quantitative data and Fisher’s exact test for qualitative data. To evaluate the threshold for detecting recurrence, metastasis or mortality due to disease, receiver operating characteristic (ROC) curves were generated. ROC curves were created by plotting the sensitivity on the y-axis and the false positive rate (1-specificity) on the x-axis. To measure the effectiveness of TAT, SF, PIC or DD levels as a marker for recurrence, metastasis or mortality due to disease, the area under the curve (AUC) was assessed. Local recurrence-free survival (RFS) was defined as the time from initial treatment to the date of clinically documented local recurrence. Metastasis-free survival (MFS) was defined as the time from initial treatment to the date of clinically documented distant metastasis. Overall survival (OS) was defined as the time from initial treatment to the date of mortality attributed to the neoplasm. Kaplan-Meier survival plots and log-rank tests were used to assess the differences in time to local recurrence, distant metastasis or overall survival. To adjust the imbalance in prognostic factors among patients, Cox proportional hazard analysis was used. p < 0.05 was considered statistically significant. The EZR software program was used for statistical analyses [49].", "Clinicopathological analysis was performed to compare the plasma levels of TAT, SF, PIC or DD to various clinical parameters using the Mann-Whitney test or Kruskal Wallis test for quantitative data and Fisher’s exact test for qualitative data. To evaluate the threshold for detecting recurrence, metastasis or mortality due to disease, receiver operating characteristic (ROC) curves were generated. ROC curves were created by plotting the sensitivity on the y-axis and the false positive rate (1-specificity) on the x-axis. To measure the effectiveness of TAT, SF, PIC or DD levels as a marker for recurrence, metastasis or mortality due to disease, the area under the curve (AUC) was assessed. Local recurrence-free survival (RFS) was defined as the time from initial treatment to the date of clinically documented local recurrence. Metastasis-free survival (MFS) was defined as the time from initial treatment to the date of clinically documented distant metastasis. Overall survival (OS) was defined as the time from initial treatment to the date of mortality attributed to the neoplasm. Kaplan-Meier survival plots and log-rank tests were used to assess the differences in time to local recurrence, distant metastasis or overall survival. To adjust the imbalance in prognostic factors among patients, Cox proportional hazard analysis was used. p < 0.05 was considered statistically significant. The EZR software program was used for statistical analyses [49].", "[SUBTITLE] Patient and tumor characteristics [SUBSECTION] A total of 64 patients with primary STSs visiting Mie University Hospital from 2012 to 2014 were included in this study. The average age of the patients was 63 years (range: 17–89 years), and the average tumor size was 12.8 cm (range: 3.5–31 cm). Histopathological diagnosis was as follows: 28 liposarcomas (17 well-differentiated liposarcomas, 6 dedifferentiated liposarcomas, and 5 myxoid liposarcomas), 10 undifferentiated pleomorphic sarcomas, 9 myxofibrosarcomas, 4 leiomyosarcomas, 3 synovial sarcomas, 3 malignant peripheral nerve sheath tumors, and 7 others in STS. Patients with STS received wide resection (44 patients), marginal resection (17 patients) and ion beam radiotherapy (3 patients). The average follow-up on patients with STS was 43.4 months (range: 0.6–75.6 months).\nA total of 64 patients with primary STSs visiting Mie University Hospital from 2012 to 2014 were included in this study. The average age of the patients was 63 years (range: 17–89 years), and the average tumor size was 12.8 cm (range: 3.5–31 cm). Histopathological diagnosis was as follows: 28 liposarcomas (17 well-differentiated liposarcomas, 6 dedifferentiated liposarcomas, and 5 myxoid liposarcomas), 10 undifferentiated pleomorphic sarcomas, 9 myxofibrosarcomas, 4 leiomyosarcomas, 3 synovial sarcomas, 3 malignant peripheral nerve sheath tumors, and 7 others in STS. Patients with STS received wide resection (44 patients), marginal resection (17 patients) and ion beam radiotherapy (3 patients). The average follow-up on patients with STS was 43.4 months (range: 0.6–75.6 months).", "A total of 64 patients with primary STSs visiting Mie University Hospital from 2012 to 2014 were included in this study. The average age of the patients was 63 years (range: 17–89 years), and the average tumor size was 12.8 cm (range: 3.5–31 cm). Histopathological diagnosis was as follows: 28 liposarcomas (17 well-differentiated liposarcomas, 6 dedifferentiated liposarcomas, and 5 myxoid liposarcomas), 10 undifferentiated pleomorphic sarcomas, 9 myxofibrosarcomas, 4 leiomyosarcomas, 3 synovial sarcomas, 3 malignant peripheral nerve sheath tumors, and 7 others in STS. Patients with STS received wide resection (44 patients), marginal resection (17 patients) and ion beam radiotherapy (3 patients). The average follow-up on patients with STS was 43.4 months (range: 0.6–75.6 months).", "Plasma STS, DD, PIC, SF and TAT values were compared according to sex, age, tumor size, location, tumor depth, FNCLCC grade, AJCC stage and treatment. Patients ≥ 60 years of age showed significantly higher DD, PIC, SF and TAT than those under 60. Patients with tumors ≥ 10 cm in size showed significantly higher DD than those with tumors < 10 cm. In evaluating FNCLCC grade, DD and SF were significantly different depending on FNCLCC grade. However, the average DD, SF and TAT levels of grade 1 were higher than in grade 2 (Table 1; Fig. 1). DD and SF were found to significantly vary according to AJCC stage. However, the lowest levels of DD and SF were observed in stage 2. PIC levels were observed to increase according to stage, despite a lack of statistical significance (Table 1; Fig. 2).\n\nTable 1Plasma levels of DD, PIC, SF and TAT according to various characteristics of patients with STS.Characteristics of STSN (64)DD AVE (SD)PIC AVE (SD)SF AVE (SD)TAT AVE (SD)SexMale331.086 (1.639)1.000 (0.910)2.990 (6.804)1.090 (0.813)Female310.904 (1.579)1.093 (0.689)5.783 (13.11)2.988 (6.432)Age (years old)< 6027\n*0.616 (1.403)\n\n*0.781 (0.802)\n\n*1.500 (2.281)\n\n*1.002 (0.789)\n≥ 6037\n*1.276 (1.694)\n\n*1.237 (0.762)\n\n*6.418 (13.18)\n\n*2.744 (5.908)\nTumor size< 1029\n*0.792 (1.434)\n1.062 (1.063)2.217 (3.053)1.112 (0.859)(cm)≥ 1035\n*1.169 (1.728)\n1.031 (0.521)6.105 (13.57)2.753 (6.079)LocationExtremity350.946 (1.617)0.934 (0.600)4.065 (9.540)1.446 (1.989)Trunk291.060 (1.605)1.179 (0.994)4.679 (11.44)2.689 (6.439)Tumor depthSuperficial200.907 (1.654)1.235 (1.066)7.455 (16.04)2.409 (4.192)Deep441.039 (1.593)0.959 (0.652)2.929 (6.096)1.827 (4.777)FNCLCCGrade 119\n#\n0.673 (1.124)\n0.784 (0.365)\n#\n3.957 (14.00)\n1.631 (3.683)Grade 222\n#\n0.534 (0.587)\n0.940 (0.681)\n#\n1.940 (2.683)\n1.124 (0.816)Grade 323\n#\n1.710 (2.266)\n1.360 (1.065)\n#\n6.960 (11.28)\n3.168 (6.779)AJCC stageI19\n#\n0.673 (1.124)\n0.784 (0.365)\n#\n3.957 (14.00)\n1.631 (3.683)II22\n#\n0.534 (0.587)\n0.940 (0.681)\n#\n1.940 (2.683)\n1.124 (0.816)III18\n#\n1.571 (2.124)\n1.233 (0.845)\n#\n7.805 (12.56)\n3.744 (7.598)IV5\n#\n2.212 (2.943)\n1.820 (1.693)\n#\n3.920 (3.784)\n1.096 (0.713)TreatmentWide resection441.184 (1.767)1.181 (0.915)\n#\n4.581 (8.657)\n2.181 (4.991)Marginal resection170.650 (1.165)0.776 (0.366)\n#\n4.347 (14.80)\n1.717 (3.895)Ion beam radiotherapy:30.243 (0.198)0.566 (0.152)\n#\n0.833 (0.611)\n1.140 (0.680)Chemotherapy-481.115 (1.759)1.162 (0.894)4.466 (10.81)2.286 (5.250)+160.648 (0.942)0.693 (0.211)3.975 (9.206)1.178 (0.807)Radiotherapy-520.775 (0.955)0.998 (0.694)3.990 (10.34)2.134 (5.050)+121.961 (3.032)1.250 (1.194)5.875 (10.76)1.468 (1.162)Average (AVE) values and standard deviation (SD) of DD, PIC, SF and TAT are shown according to each listed parameter. * and # indicate significant differences using the Mann-Whitney test (p < 0.05) and the Kruskal-Wallis test (P < 0.05), respectively\n\nPlasma levels of DD, PIC, SF and TAT according to various characteristics of patients with STS.\nAverage (AVE) values and standard deviation (SD) of DD, PIC, SF and TAT are shown according to each listed parameter. * and # indicate significant differences using the Mann-Whitney test (p < 0.05) and the Kruskal-Wallis test (P < 0.05), respectively\n\nFig. 1BOX plot of plasma DD, PIC, SF and TAT levels according to FNCLCC grade. G1, G2 and G3 correspond to FNCLCC grades 1, 2 and 3. Statistical analysis was performed by the Kruskal-Wallis test\n\nBOX plot of plasma DD, PIC, SF and TAT levels according to FNCLCC grade. G1, G2 and G3 correspond to FNCLCC grades 1, 2 and 3. Statistical analysis was performed by the Kruskal-Wallis test\n\nFig. 2BOX plot of plasma DD, PIC, SF and TAT levels according to AJCC stage. I, II, III and IV correspond to AJCC stages I, II, III and IV. Statistical analysis was performed by the Kruskal-Wallis test\n\nBOX plot of plasma DD, PIC, SF and TAT levels according to AJCC stage. I, II, III and IV correspond to AJCC stages I, II, III and IV. Statistical analysis was performed by the Kruskal-Wallis test", "During the period of this study, 7 patients developed recurrence, 21 patients developed metastasis (metastasis group), and 17 patients died of disease (DOD group). No significant differences were observed in DD, PIC, SF and TAT between patients with and without recurrence. The metastasis group had significantly higher plasma DD (p = 0.0394), PIC (p = 0.00532) and SF (p = 0.00249) levels than the group without metastasis. The DOD group showed significantly higher DD (p = 0.00105), PIC (p = 0.000542), SF (p = 0.000126) and TAT (p = 0.0373) compared to the surviving patients (Fig. 3).\n\nFig. 3BOX plot of plasma DD, PIC, SF and TAT levels according to clinical outcome. Rec: recurrence, meta: metastasis, DOD: died of disease. Statistical analysis was performed by the Mann-Whitney test (p < 0.05) \n\nBOX plot of plasma DD, PIC, SF and TAT levels according to clinical outcome. Rec: recurrence, meta: metastasis, DOD: died of disease. Statistical analysis was performed by the Mann-Whitney test (p < 0.05) ", "Univariate COX proportional hazard analysis was performed using DD, PIC, SF and TAT as continuous variable. For RFS, only stage III/IV was significantly different (HR: 6.20, 95%CI: 1.19–32.1, p = 0.0298). For MFS, DD (HR: 1.26, 95%CI: 1.07–1.50, p = 0.00612), PIC (HR: 2.39, 95%CI: 1.59–3.58, p = 0.000022) and stage III/IV (HR: 3.34, 95%CI: 1.40–7.96, p = 0.006494) showed significant differences. For OS, DD (HR: 1.33, 95%CI: 1.11–1.58, p = 0.001362), PIC (HR: 1.99, 95%CI: 1.42–2.79, p = 0.000059), SF (HR: 1.02, 95%CI: 1.01–1.05, p = 0.026) and stage III/IV (HR: 5.66, 95%CI: 1.98–16.1, p = 0.001164) demonstrated significant differences (Table 2).\n\nTable 2Univariate COX proportional hazard analysisRFSMFSOSHR95%CIp-valueHR95%CIp-valueHR95%CIp-valueMale4.9 × 108/0.9980.780.33–1.840.5730.670.24–1.860.447Age ≥ 600.590.13–2.650.4951.450.58–3.610.4233.320.09–11.70.063Size ≥ 10 cm0.840.18–3.780.8280.980.41–2.310.9651.660.59–4.670.335Superficial0.250.03–2.110.2040.880.35–2.180.7871.110.39–3.150.830Trunk2.490.55–11.20.2331.710.72–4.050.2182.090.75–5.810.154DD1.110.77–1.590.5621.261.07–1.50\n0.0061\n1.331.11–1.58\n0.0014\nPIC1.260.60–2.650.5342.391.59–3.58\n0.00003\n1.991.42–2.79\n0.00006\nSF1.020.98–1.070.2191.020.99–1.040.0961.021.01–1.05\n0.026\nTAT0.950.71–1.270.7360.980.89–1.090.8291.010.92–1.100.855High grade0.0250.0-16.20.26439.30.79–19570.06567.20.76–51.30.087Stage I, II vs. III, IV6.201.19-32.1-\n0.029\n3.341.40–7.96\n0.007\n5.661.98–16.1\n0.0011\nPlasma DD, PIC, SF and TAT levels were analyzed as continuous variables\n\nUnivariate COX proportional hazard analysis\nPlasma DD, PIC, SF and TAT levels were analyzed as continuous variables\nMultivariate COX proportional hazard analysis was used to adjust for the imbalance in prognostic factors among patients. Since only stage III/IV showed a significant difference in RFS, multivariate analysis was not performed. DD, PIC and stage III/IV were adopted for multivariate analysis of MFS, with only PIC exhibiting a significant difference (HR: 2.14, 95% CI: 1.26–3.62, p = 0.00477). DD, PIC, SF and stage III/IV were adopted for multivariate analysis of OS, with only stage III, IV exhibiting a significant difference (HR: 4.54, 95% CI: 1.46-14.0, p = 0.00872). PIC showed a risk of poor prognosis, but there was no significant difference (HR: 1.75, 95%CI: 0.96–3.19, p = 0.0655) (Table 3).\n\nTable 3Multivariate COX proportional hazard analysisMFSOSHR95%CIp-valueHR95%CIp-valueDD0.990.77–1.270.961DD0.930.65–1.310.692PIC2.141.26–3.62\n0.0048\nPIC1.750.96–3.190.065Stage III, IV2.380.94–6.060.067SF1.0280.99–1.060.108Stage I, II vs. III, IV4.541.46-14.0\n0.0087\nPlasma DD, PIC and SF levels were analyzed as continuous variables\n\nMultivariate COX proportional hazard analysis\nPlasma DD, PIC and SF levels were analyzed as continuous variables\nFor further prognostic analysis, patients with STS of FNCLCC grade 1 or with distant metastasis at initial diagnosis (stage IV) were excluded. Univariate COX proportional hazard analysis indicated that only SF was significantly different in RFS (HR: 1.08, 95%CI: 1.01–1.16, p = 0.013). For MFS, significant differences were detected in DD (HR: 1.22, 95%CI: 1.00-1.49, p = 0.049), PIC (HR: 2.65, 95%CI: 1.53–4.59, p = 0.00051) and SF (HR: 1.07, 95%CI: 1.03–1.11, p = 0.00054); whereas for OS, significant differences were detected in DD (HR: 1.35, 95%CI: 1.10–1.67, p = 0.0043), PIC (HR: 2.45, 95%CI: 1.47–4.08, p = 0.00057) and SF (HR: 1.07, 95%CI: 1.03–1.11, p = 0.00028) (Supplementary Table 1). DD, PIC and SF were adopted for multivariate analysis. PIC (HR: 2.37, 95%CI: 1.31–4.29, p = 0.004) and SF (HR: 1.06, 95%CI: 1.01–1.12, p = 0.013) exhibited significant differences in MFS, while PIC (HR: 2.13, 95%CI: 1.21–3.72, p = 0.0079) and SF (HR: 1.05, 95%CI: 1.00-1.11, p = 0.025) were significantly different in OS (Supplementary Table 2).", "ROC analysis was performed and UAC was evaluated to determine the diagnostic accuracy for identifying DOD within 5-years in STS. ROC analysis of DD produced an AUC of 0.77 (95% confidence interval (CI): 0.635–0.906). With a threshold of 0.74, the sensitivity and specificity for identifying 5-year DOD were 70.6% and 80.9%, respectively. ROC analysis of PIC produced an AUC of 0.784 (95% CI: 0.651–0.917). With a threshold of 1.1, the sensitivity and specificity for identifying 5-year DOD were 64.7% and 83.0%, respectively. ROC analysis of SF produced an AUC of 0.815 (95% CI: 0.694–0.935). With a threshold of 1.6, the sensitivity and specificity for identifying 5-year DOD were 76.5% and 78.7%, respectively. ROC analysis of TAT produced an AUC of 0.672 (95% CI: 0.509–0.835). With a threshold of 1.71, the sensitivity and specificity for identifying 5-year DOD were 47.1% and 89.4%, respectively. To divide patients into two groups for further analysis, thresholds of 0.74 (DD), 1.1 (PIC), 1.6 (SF) and 1.71 (TAT) were adopted as the Youden index, and the low and high groups for each threshold were analyzed. (Fig. 4).\n\nFig. 4ROC analysis. ROC analysis of DD, PIC, SF and TAT to determine diagnostic accuracy of detecting DOD in sarcoma patients. AUC: area under the curve. Thresholds were determined by the Youden index\n\nROC analysis. ROC analysis of DD, PIC, SF and TAT to determine diagnostic accuracy of detecting DOD in sarcoma patients. AUC: area under the curve. Thresholds were determined by the Youden index", "DD (low < 0.74 ≤ high), PIC (low < 1.1 ≤ high), SF (low < 1.6 ≤ high) and TAT (low < 1.71 ≤ high) were divided into two groups. Five-year recurrence-free survival (RFS), metastasis-free survival (MFS), and over-all survival (OS) between the low- and high-DD, PIC, SF and TAT groups were compared using Kaplan-Meier analysis and the log-rank tests. RFS showed no significant difference in DD (low DD 88.1%, high DD 87.4%, p = 0.809), PIC (low PIC 87.9%, high PIC 87.7%, p = 0.855), SF (low SF 89.8%, high SF 82.6%, p = 0.456) or TAT (low TAT 89.4%, high TAT 77.9%, p = 0.267). MFS showed significant differences in DD (low DD 75.4%, high DD 42.9%, p = 0.000628), PIC (low PIC 73.6%, high PIC 42.1%, p = 0.00164), SF (low SF 73.5%, high SF 47.8%, p = 0.0047) and TAT (low TAT 70.4%, high TAT 38.5%, p = 0.00556). OS and MFS showed significant differences in DD (low DD 87.7%, high DD 41.6%, p = 0.00000601), PIC (low PIC 86.1%, high PIC 38.3%, p = 0.0000516), SF (low SF 89.8%, high TM 41.0%, p = 0.00000925) and TAT (low TM 81.6%, high TM 35.9%, p = 0.000143) (Fig. 5). Thus, high levels of DD, PIC, SF and TAT appear to be important factors involved in metastatic potential and lead to poor prognosis.\n\nFig. 5Kaplan-Meier analysis. Kaplan-Meier analysis was performed according to the low and high expression sub-groups for each marker (DD, PIC, SF, and TAT). RFS indicates recurrence-free survival. MFS indicates metastasis-free survival. OS indicates over-all survival. Significance was analyzed using the log-rank test\n\nKaplan-Meier analysis. Kaplan-Meier analysis was performed according to the low and high expression sub-groups for each marker (DD, PIC, SF, and TAT). RFS indicates recurrence-free survival. MFS indicates metastasis-free survival. OS indicates over-all survival. Significance was analyzed using the log-rank test", "The low and high DD, PIC, SF and TAT groups were analyzed using univariate COX proportional analysis. For RFS, no significant differences were observed between the low and high groups for DD, PIC, SF and TAT. In contrast, analysis of MFS and OS revealed significant differences between the low and high groups for DD, PIC, SF and TAT (Table 4). Significance was mutually nullified by inclusion of high DD, high PIC, high SF and high TAT in multivariate analysis, thus separate analyses were performed with stage III/IV. High DD, high PIC and high SF showed significant differences in MFS and all high groups showed significant difference in OS (Table 5).\n\nTable 4Univariate COX proportional hazard analysisRFSMFSOSHR95%CIp-valueHR95%CIp-value95%CIp-valueHigh DD0.960.18–4.990.9643.581.48–8.66\n0.0045\n11.43.16-41.0\n0.0002\nHigh PIC1.030.19–5.330.9693.161.38–7.93\n0.0071\n8.192.57–25.7\n0.0004\nHigh SF0.730.14–3.800.7132.591.08–6.20\n0.032\n8.412.35–30.1\n0.0011\nHigh TAT1.750.33–9.140.5052.711.11–6.64\n0.028\n5.801.53–12.4\n0.0056\nPlasma DD, PIC, SF and TAT levels were divided into low and high groups by their respective thresholds (Youden index). COX analysis was performed using binary variables\n\nUnivariate COX proportional hazard analysis\nPlasma DD, PIC, SF and TAT levels were divided into low and high groups by their respective thresholds (Youden index). COX analysis was performed using binary variables\n\nTable 5Multivariate COX proportional hazard analysisMFSOSHR95%CIp-value95%CIp-valueHigh DD3.051.2–7.7\n0.019\n11.33.16-41.0\n0.0002\nStage I, II vs. III, IV2.310.91–5.830.0765.661.98–16.1\n0.0012\nHigh PIC2.901.19–7.03\n0.019\n8.192.57–25.7\n0.0004\nStage I, II vs. III, IV2.661.08–6.50\n0.032\n5.661.98–16.1\n0.0012\nHigh SF2.211.08–6.20\n0.032\n8.412.35–30.1\n0.0011\nStage I, II vs. III, IV2.330.87–6.190.0885.661.98–16.1\n0.0012\nHigh TAT2.711.11–6.640.1115.801.53–12.40\n0.0056\nStage I, II vs. III, IV3.341.40–7.96\n0.0065\n5.661.98–16.1\n0.0012\nPlasma DD, PIC, SF and TAT levels were analyzed as continuous variables. From Table 3, stage III/IV had significant difference and was adopted this analysis\n\nMultivariate COX proportional hazard analysis\nPlasma DD, PIC, SF and TAT levels were analyzed as continuous variables. From Table 3, stage III/IV had significant difference and was adopted this analysis", "The intimate relationship between coagulation and fibrinolysis in malignant tumors is a common and well-known phenomena. This study is the first to report on the association between STSs and coagulation and fibrinolysis factors. In this study, plasma levels of DD, PIC, SF and TAT were evaluated to assess the state of coagulation and fibrinolysis in patients with STS. Enhanced coagulation activity or activation of the coagulation cascade originating from TF on normal cells, tumor cells or micro vesicles can be monitored by assessing SF and TAT. In a study examining the relationship between SF and tumors, SF was found to bind lymphocytes and tumor cells, thereby inhibiting both cell adherence and promoting tumor cell resistance to cytotoxicity [50]. High levels of SF in tumor tissues of lung cancer were found to be related to poor prognosis [51]. TAT has proven to be a useful marker for discriminating patients with benign and malignant ovarian tumors [52]. Furthermore, increases in plasma TAT levels appear to be related to tumor spread in lung cancer [53]. Although many tumor cells are known to be highly correlated with coagulation, reports regarding the association between SF, TAT and prognosis are not common.\nIn contrast, many studies on the relationship between fibrinolysis makers and malignant tumors have been reported. DD is correlated with prognosis in patients with various malignant tumors. High DD levels are associated with poor prognosis in patients with breast cancer, renal cell carcinoma, gastric cancer, lung cancer, bladder cancers, colorectal cancers, gynecological tumors and lymphoma and sarcoma [47, 54–61]. High levels of PIC have been reported to be correlated with poor prognosis in lung cancer [62]. For sarcomas, studies have mainly analyzed D-dimer, reporting that high DD levels are associated with poor prognosis [63–66]. However, there have been no reports evaluating coagulation and fibrinolysis states in patients with STS. Here, we elucidated this relationship by comparing the severity of coagulation and fibrinolysis with the disease condition in sarcoma patients.\nIn clinical situations, higher stage diagnoses are highly correlated with poor prognosis in patients with STS [67]. Histological grade [68] and stage indicate tumor aggressiveness and spread, information that is indispensable to making decisions about tumor treatment strategies. In considering FNCLCC grades, the levels of DD and SF were found to be significantly different (Fig. 1). Similarly, the levels of DD and SF differed significantly according to AJCC stage (Fig. 2). However, only PIC levels were increased in both higher grade and higher stage, with DD showing a similar, if not significant, pattern. This indicates that PIC, and probably DD, is correlated with malignancy and tumor progression.\nIn the subsequent analysis, thresholds were adopted to determine the utility of DD, PIC, SF and TAT as predictive markers of prognosis. Thresholds of ≥ 0.74 DD, ≥ 1.1 PIC, ≥ 1.6 SF successfully demonstrated risk of metastasis and poor prognosis. Specifically, DD levels ≥ 0.74 (HR: 3.05) and SF levels ≥ 1.6 (HR: 2.21) can be used as independent predictors of future metastasis (Tables 4 and 5). DD and SF were useful as diagnostic tools only when evaluated using a threshold. In contrast, univariate COX analysis of continuous variables indicated that SF and TAT show weak or no association with metastasis (HR: 1.02, 0.98) and prognosis (HR: 1.02, 1.01) (Table 2). Increases in coagulation markers were not accurately associated with the disease condition. Univariate analysis of fibrinolysis markers indicated that the HR of metastasis (DD: 1.26, PIC: 2.39) and prognosis (DD: 1.33, PIC: 1.99) was higher than with coagulation markers (Table 2). Furthermore, multivariate COX analysis indicted that only PIC exhibited a significant difference in metastasis (HR: 2.14) (Table 3). Additionally, in patients with STS (excluding FNCLCC grade 1 and distant metastasis at initial diagnosis), PIC (MFS, HR: 2.37, p = 0.004, OS, HR: 2.13, p = 0.0079) more effectively and significantly predicted metastasis and poor prognosis than SF (MFS, HR: 1.06, p = 0.013, OS, HR: 1.05, p = 0.025) (Supplementary Tables 1, 2). Thus, fibrinolysis markers were more effective than coagulation markers in reflecting sarcoma malignancy and spread in patients with STS. Especially, PIC levels ≥ 1.1 can be used to predict metastasis risk, and further increases in PIC levels correspond to further increases in metastasis risk.\nAccording to previous sarcoma studies, conversion of plasminogen to plasmin was observed on the sarcoma cell surface [69]. High expression of uPA, uPAR and PAI-1 in tumor tissue, and high levels of serum uPAR were associated with poor prognosis [70]. It was also reported that elevated levels of uPA were observed in leiomyosarcoma, malignant fibrous histiocytoma, higher stage malignancies, sarcomas with necrosis, metastasis or local recurrence. High levels of uPA in tumor tissues were reported to be associated with malignant phenotype [71]. Additionally, synthetic ligands targeting EGFR and uPAR effectively induced sarcoma cell death in vitro and suppressed tumor growth in vivo [72, 73]. These previous reports indicate that the uPA/uPAR system is highly associated with sarcoma exacerbation. Furthermore, inhibitors of PAI-I and uPA have been reported to reduce lung metastasis of osteosarcoma cells [74],[34]. Thus, the uPAR/uPA system appears to be involved in mediating metastatic potential, which is consistent with the findings of the present study. PIC was thought to be a sensitive tool for detecting activation of the uPAR/uPA system in STS and led to successful prediction of metastasis and poor prognosis in patients with STS.", "Serum levels of DD ≥ 0.74, PIC ≥ 1.1, SF ≥ 1.6 and TAT ≥ 1.71 were useful as makers for predicting future metastasis and poor prognosis. Moreover, fibrinolysis was better than coagulation at reflecting the disease condition of patients with STS. Notably, PIC levels ≥ 1.1 can not only predict the risk of metastasis and poor prognosis, but also increasing PIC levels correspond to further increases in risks of metastasis and poor prognosis.\nOur study had the following limitations. This study was retrospective and the number of patients was small. Statistical analysis could not be performed by each disease subtype, because STSs are rare entity and including many subtypes. Many studies have needed to analyze STS as a group, rather than by each histological classification. The information of concomitant pharmacological treatments were not included in the statistical analysis.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, null, null, "results", null, null, null, null, null, null, null, "discussion", "conclusion", "supplementary-material", null ]
[ "Soft tissue sarcoma", "Metastasis", "Prognosis", "Coagulation", "Fibrinolysis", "D-dimer", "plasmin-α2 plasmin inhibitor complex", "Soluble fibrin", "And thrombin-antithrombin III complex" ]
Role of community pharmacy professionals in child health service provision in Ethiopia: a cross-sectional survey in six cities of Amhara regional state.
36258191
Community pharmacy professionals have great potential to deliver various public health services aimed at improving service access, particularly in countries with a shortage of health professionals. However, little is known about their involvement in child health service provision in Ethiopia.
BACKGROUND
A multi-center cross-sectional survey was conducted among 238 community pharmacy professionals from March to July 2020 in Amhara regional state of Ethiopia. Independent samples t-test and one way Analysis of Variance (ANOVA) was used to test the mean difference.
METHODS
Most community pharmacy professionals were 'involved' in providing child health services related to 'advice about vitamins/supplements' (46.6%), 'advice about infant milk/formulas' (47.1%) and 'responding to minor symptoms' (50.8%) for children. The survey revealed that, community pharmacy professionals were less frequently involved in providing childhood 'vaccination' services. Further, level of involvement of community pharmacy professionals differed according to participants' licensure level, setting type, responsibility in the facility and previous training experience in child health services.
RESULTS
Community pharmacy professionals have been delivering various levels of child health services, demonstrating ability and capacity in improving access to child health services in Ethiopia. However, there is a need for training and government support to optimize pharmacist engagement and contribution to service delivery.
CONCLUSION
[ "Child", "Humans", "Pharmacies", "Cross-Sectional Studies", "Ethiopia", "Cities", "Pharmacists", "Child Health Services", "Vitamins", "Community Pharmacy Services" ]
9578271
null
null
null
null
Results
[SUBTITLE] Characteristics of the study participants [SUBSECTION] From the total of 264 community pharmacy professionals approached, 238 participants completed the questionnaires: a response rate of 90%. Characteristics of the study participants are presented in Table 1. Table 1Characteristics of the community pharmacy professionals included in the studyCharacteristicsTotal (n = 238)Characteristics of the community pharmacy professionalsN (%) Sex Male117(49.2)Female121(50.8) Educational qualification in Pharmacy Diploma in Pharmacy130(54.6)Bachelor of Pharmacy (BPharm)91(38.2)Master of Pharmacy (MSc)17(7.1) Work experience in years (Mean, SD) 6.05 ± 5.49Less than 5 years130(54.6)5–10 years40(16.8)Greater than 10 years68(28.6) Licensure by regulatory Authority Druggist/Pharmacy technician96(40.3)Junior Pharmacist48(20.2)Senior Pharmacist/Druggist45(18.9)Chief Pharmacist19(8.0)Expert Pharmacist30(12.6) Facility (CDRO)type Drug store106(44.5)Pharmacy132(55.5) Responsibility in the CDRO Owner96(40.3)Employed142(59.7) Have you received any in -services training regarding maternal and child health services delivery in CDROs? Yes42(17.6)No196(82.4) Characteristics of the community pharmacy professionals included in the study From the total of 264 community pharmacy professionals approached, 238 participants completed the questionnaires: a response rate of 90%. Characteristics of the study participants are presented in Table 1. Table 1Characteristics of the community pharmacy professionals included in the studyCharacteristicsTotal (n = 238)Characteristics of the community pharmacy professionalsN (%) Sex Male117(49.2)Female121(50.8) Educational qualification in Pharmacy Diploma in Pharmacy130(54.6)Bachelor of Pharmacy (BPharm)91(38.2)Master of Pharmacy (MSc)17(7.1) Work experience in years (Mean, SD) 6.05 ± 5.49Less than 5 years130(54.6)5–10 years40(16.8)Greater than 10 years68(28.6) Licensure by regulatory Authority Druggist/Pharmacy technician96(40.3)Junior Pharmacist48(20.2)Senior Pharmacist/Druggist45(18.9)Chief Pharmacist19(8.0)Expert Pharmacist30(12.6) Facility (CDRO)type Drug store106(44.5)Pharmacy132(55.5) Responsibility in the CDRO Owner96(40.3)Employed142(59.7) Have you received any in -services training regarding maternal and child health services delivery in CDROs? Yes42(17.6)No196(82.4) Characteristics of the community pharmacy professionals included in the study [SUBTITLE] Involvement of community pharmacy professionals in child health services provision [SUBSECTION] As presented in Table 2, community pharmacy professionals had the highest mean involvement score in ‘advising about vitamins/ supplements’ to children (mean = 3.24, SD = 0.717). The percentage distribution also showed that most community pharmacy professionals reported that they have been either ‘very involved’ (39.5%) or ‘involved’ (46.6%) in ‘advising about vitamins/ supplements’ to children. The second highest mean involvement score of community pharmacy professionals was in providing child health services related to ‘advising about infant formulas /milk’, with 37% ‘very involved’ and 47.1% ‘involved’. Most community pharmacy professionals also reported involvement in child health service provision related to ‘responding to minor symptoms’, where more than 50% of the participants reported being ‘involved’. The lowest mean involvement score was provision of child health service related to ‘vaccination’ (mean = 2.65, SD = 0.923). Compared to the other child health services, a higher proportion of participants were a ‘little involved’ (30.7%) and ‘not at all involved’ (11.8%) in providing services related to vaccinations. Additional details are provided in Table 2. Table 2Percentage distribution and mean levels of involvement of community pharmacy professionals in child health servicesChild health serviceVery involved, n (%)Involved, n (%)Little involved n (%)Not at all involved, n (%)Mean involvement scoreStd. DeviationAdvising about vitamins/ supplements94(39.5)111(46.6)30(12.6)3(1.3)3.240.717Provision of vaccination/ Checking the child’s vaccination status and refer for vaccination/ provide child vaccination advice to the parent46(19.3)91(38.2)73(30.7)28(11.8)2.650.923Responding to minor symptoms (fever, acute diarrhea, respiratory symptoms)79(33.2)121(50.8)32(13.4)6(2.5)3.150.740Advising about infant formulas /milk88(37.0)112(47.1)30(12.6)8(3.4)3.180.776Overall mean involvement score12.222.452 Percentage distribution and mean levels of involvement of community pharmacy professionals in child health services As presented in Table 2, community pharmacy professionals had the highest mean involvement score in ‘advising about vitamins/ supplements’ to children (mean = 3.24, SD = 0.717). The percentage distribution also showed that most community pharmacy professionals reported that they have been either ‘very involved’ (39.5%) or ‘involved’ (46.6%) in ‘advising about vitamins/ supplements’ to children. The second highest mean involvement score of community pharmacy professionals was in providing child health services related to ‘advising about infant formulas /milk’, with 37% ‘very involved’ and 47.1% ‘involved’. Most community pharmacy professionals also reported involvement in child health service provision related to ‘responding to minor symptoms’, where more than 50% of the participants reported being ‘involved’. The lowest mean involvement score was provision of child health service related to ‘vaccination’ (mean = 2.65, SD = 0.923). Compared to the other child health services, a higher proportion of participants were a ‘little involved’ (30.7%) and ‘not at all involved’ (11.8%) in providing services related to vaccinations. Additional details are provided in Table 2. Table 2Percentage distribution and mean levels of involvement of community pharmacy professionals in child health servicesChild health serviceVery involved, n (%)Involved, n (%)Little involved n (%)Not at all involved, n (%)Mean involvement scoreStd. DeviationAdvising about vitamins/ supplements94(39.5)111(46.6)30(12.6)3(1.3)3.240.717Provision of vaccination/ Checking the child’s vaccination status and refer for vaccination/ provide child vaccination advice to the parent46(19.3)91(38.2)73(30.7)28(11.8)2.650.923Responding to minor symptoms (fever, acute diarrhea, respiratory symptoms)79(33.2)121(50.8)32(13.4)6(2.5)3.150.740Advising about infant formulas /milk88(37.0)112(47.1)30(12.6)8(3.4)3.180.776Overall mean involvement score12.222.452 Percentage distribution and mean levels of involvement of community pharmacy professionals in child health services [SUBTITLE] Difference in the involvement of community pharmacy professionals in child health provision based on sub-groups of respondents’ characteristics [SUBSECTION] As depicted in Table 3, significant mean difference of involvement in child health service provision was observed among different sub-groups of pharmacists. To be more specific, significant differences in mean involvement was observed based on educational qualifications in Pharmacy, professional licensure levels, Facility (CDRO)types, responsibilities in the CDROs and in services training status. A significantly higher level of pharmacy professionals’ involvement in child health service provision was observed among those working in ‘pharmacy’ settings compared to those working in ‘drug store’ settings. Involvement of community pharmacy professionals in child health service provision was also significantly higher among CDRO ‘owners’ in comparison to their counterparts (‘employees’). Additionally, a significant difference in involvement was also observed based on ‘in-service training status’, with less involvement in child health service provision observed among those who did not report having received training about child health service provision compared to those who had received training. The post-hoc analyses (see Table 4) showed that significant differences in mean involvement score was observed among sub-groups of the study participants based on professional ‘licensure level’. Involvement scores in child health services provision were higher among community pharmacy professionals with licensure level of ‘senior pharmacist’ and ‘expert pharmacist’ compared to pharmacy professionals with a licensure level of ‘druggist/ pharmacy technicians’. Table 3Total involvement of community pharmacy professionals in provision of child health service among different subgroups of respondents’: Independent sample t-test and one way ANOVADemographic and related characteristicsCategoriesInvolvement in child health service Mean ± SD P-value- mean difference P-value- Levene’s Test for Equality of Variances Sex Male12.38 ± 2.4800.3310.557Female12.07 ± 2.425 Educational qualification in Pharmacy Diploma in Pharmacy11.85 ± 2.5940.034*0.450Bachelor of Pharmacy (BPharm)12.59 ± 2.196Master of Pharmacy (MSc)13.00 ± 2.264 Work experience in years Less than 5 years11.89 ± 2.4280.0740.7015–10 years12.67 ± 2.506Greater than 10 years12.50 ± 2.311 Licensure by regulatory Authority Druggist/Pharmacy technician11.39 ± 2.114< 0.001*0.124Junior Pharmacist12.38 ± 2.878Senior Pharmacist/Druggist13.16 ± 2.256Chief Pharmacist12.79 ± 2.440Expert Pharmacist12.87 ± 2.300 Facility (CDRO)type Drug store11.72 ± 2.5440.004*0.973Pharmacy12.62 ± 2.307 Responsibility in the CDROs Owner12.79 ± 2.1900.003*0.158Employed11.83 ± 2.549 In -services training received Yes13.38 ± 2.3370.001*0.868No11.97 ± 2.409*The mean difference is significant at the 0.05 level Total involvement of community pharmacy professionals in provision of child health service among different subgroups of respondents’: Independent sample t-test and one way ANOVA *The mean difference is significant at the 0.05 level Table 4Post hoc analyses of factors influencing community pharmacy pharmacists’ involvement in child health servicesVariablesFactorsGroupMean differenceP-value95% Confidence Interval Lower Bound Lower Bound Educational qualification in PharmacyDiploma in pharmacyBachelor of Pharmacy (BPharm)− 0.7400.080-1.540.06Master of Pharmacy (MSc)-1.1460.205-2.660.36Bachelor of Pharmacy (BPharm)Diploma in pharmacy0.7400.080− 0.061.54Master of Pharmacy (MSc)− 0.4071.000-1.951.14Master of Pharmacy (MSc)Diploma in pharmacy1.1460.205− 0.362.66Bachelor of Pharmacy (BPharm)0.4071.000-1.141.95Licensure by regulatory AuthorityDruggist/Pharmacy technicianJunior Pharmacist− 0.9900.185-2.170.19Senior Pharmacist-1.770*< 0.001-2.98− 0.56Chief Pharmacist-1.4040.187-3.080.28Expert Pharmacist-1.481* 0.030 -2.88− 0.08Junior PharmacistDruggist/Pharmacy technician0.9900.185− 0.192.17Senior Pharmacist− 0.7811.000-2.170.61Chief Pharmacist− 0.4141.000-2.231.40Expert Pharmacist− 0.4921.000-2.051.07Senior PharmacistDruggist/Pharmacy technician1.770*< 0.0010.562.98Junior Pharmacist0.7811.000− 0.612.17Chief Pharmacist0.3661.000-1.462.20Expert Pharmacist0.2891.000-1.291.87Chief PharmacistDruggist/Pharmacy technician1.4040.187− 0.283.08Junior Pharmacist0.4141.000-1.402.23Senior Pharmacist− 0.3661.000-2.201.46Expert Pharmacist− 0.0771.000-2.041.88Expert PharmacistDruggist/Pharmacy technician1.481*0.0300.082.88Junior Pharmacist0.4921.000-1.072.05Senior Pharmacist− 0.2891.000-1.871.29Chief Pharmacist0.0771.000-1.882.04* The mean difference is significant at the 0.05 level Post hoc analyses of factors influencing community pharmacy pharmacists’ involvement in child health services * The mean difference is significant at the 0.05 level As depicted in Table 3, significant mean difference of involvement in child health service provision was observed among different sub-groups of pharmacists. To be more specific, significant differences in mean involvement was observed based on educational qualifications in Pharmacy, professional licensure levels, Facility (CDRO)types, responsibilities in the CDROs and in services training status. A significantly higher level of pharmacy professionals’ involvement in child health service provision was observed among those working in ‘pharmacy’ settings compared to those working in ‘drug store’ settings. Involvement of community pharmacy professionals in child health service provision was also significantly higher among CDRO ‘owners’ in comparison to their counterparts (‘employees’). Additionally, a significant difference in involvement was also observed based on ‘in-service training status’, with less involvement in child health service provision observed among those who did not report having received training about child health service provision compared to those who had received training. The post-hoc analyses (see Table 4) showed that significant differences in mean involvement score was observed among sub-groups of the study participants based on professional ‘licensure level’. Involvement scores in child health services provision were higher among community pharmacy professionals with licensure level of ‘senior pharmacist’ and ‘expert pharmacist’ compared to pharmacy professionals with a licensure level of ‘druggist/ pharmacy technicians’. Table 3Total involvement of community pharmacy professionals in provision of child health service among different subgroups of respondents’: Independent sample t-test and one way ANOVADemographic and related characteristicsCategoriesInvolvement in child health service Mean ± SD P-value- mean difference P-value- Levene’s Test for Equality of Variances Sex Male12.38 ± 2.4800.3310.557Female12.07 ± 2.425 Educational qualification in Pharmacy Diploma in Pharmacy11.85 ± 2.5940.034*0.450Bachelor of Pharmacy (BPharm)12.59 ± 2.196Master of Pharmacy (MSc)13.00 ± 2.264 Work experience in years Less than 5 years11.89 ± 2.4280.0740.7015–10 years12.67 ± 2.506Greater than 10 years12.50 ± 2.311 Licensure by regulatory Authority Druggist/Pharmacy technician11.39 ± 2.114< 0.001*0.124Junior Pharmacist12.38 ± 2.878Senior Pharmacist/Druggist13.16 ± 2.256Chief Pharmacist12.79 ± 2.440Expert Pharmacist12.87 ± 2.300 Facility (CDRO)type Drug store11.72 ± 2.5440.004*0.973Pharmacy12.62 ± 2.307 Responsibility in the CDROs Owner12.79 ± 2.1900.003*0.158Employed11.83 ± 2.549 In -services training received Yes13.38 ± 2.3370.001*0.868No11.97 ± 2.409*The mean difference is significant at the 0.05 level Total involvement of community pharmacy professionals in provision of child health service among different subgroups of respondents’: Independent sample t-test and one way ANOVA *The mean difference is significant at the 0.05 level Table 4Post hoc analyses of factors influencing community pharmacy pharmacists’ involvement in child health servicesVariablesFactorsGroupMean differenceP-value95% Confidence Interval Lower Bound Lower Bound Educational qualification in PharmacyDiploma in pharmacyBachelor of Pharmacy (BPharm)− 0.7400.080-1.540.06Master of Pharmacy (MSc)-1.1460.205-2.660.36Bachelor of Pharmacy (BPharm)Diploma in pharmacy0.7400.080− 0.061.54Master of Pharmacy (MSc)− 0.4071.000-1.951.14Master of Pharmacy (MSc)Diploma in pharmacy1.1460.205− 0.362.66Bachelor of Pharmacy (BPharm)0.4071.000-1.141.95Licensure by regulatory AuthorityDruggist/Pharmacy technicianJunior Pharmacist− 0.9900.185-2.170.19Senior Pharmacist-1.770*< 0.001-2.98− 0.56Chief Pharmacist-1.4040.187-3.080.28Expert Pharmacist-1.481* 0.030 -2.88− 0.08Junior PharmacistDruggist/Pharmacy technician0.9900.185− 0.192.17Senior Pharmacist− 0.7811.000-2.170.61Chief Pharmacist− 0.4141.000-2.231.40Expert Pharmacist− 0.4921.000-2.051.07Senior PharmacistDruggist/Pharmacy technician1.770*< 0.0010.562.98Junior Pharmacist0.7811.000− 0.612.17Chief Pharmacist0.3661.000-1.462.20Expert Pharmacist0.2891.000-1.291.87Chief PharmacistDruggist/Pharmacy technician1.4040.187− 0.283.08Junior Pharmacist0.4141.000-1.402.23Senior Pharmacist− 0.3661.000-2.201.46Expert Pharmacist− 0.0771.000-2.041.88Expert PharmacistDruggist/Pharmacy technician1.481*0.0300.082.88Junior Pharmacist0.4921.000-1.072.05Senior Pharmacist− 0.2891.000-1.871.29Chief Pharmacist0.0771.000-1.882.04* The mean difference is significant at the 0.05 level Post hoc analyses of factors influencing community pharmacy pharmacists’ involvement in child health services * The mean difference is significant at the 0.05 level
Conclusion
Community pharmacy professionals have been involved in providing child health services such as advice about vitamins/supplements, advice about infant milk/formulas and responding to minor symptoms for children. The increasing role of pharmacy professionals beyond merely dispensing of medications has the potential to improve access to child health services. Integrating and enhancing the role of community pharmacists in child health care service delivery could assist countries with their efforts in reducing child mortality and, subsequently, in meeting SDG targets. Empowering community pharmacy professionals through training focusing on child health service provision would also be needed to improve their involvement in the service delivery.
[ "Background", "Methods", "Study design, study area and sampling", "Inclusion and exclusion criteria", "Data collection tools and procedures", "Data analysis", "Characteristics of the study participants", "Involvement of community pharmacy professionals in child health services provision", "Difference in the involvement of community pharmacy professionals in child health provision based on sub-groups of respondents’ characteristics", "Strengths and limitations of the study", "Implications for policy, future practice, and research" ]
[ "Lowering child morality is a global priority with the United Nations (UN) Sustainable Development Goal (SDG) 3.2 calling for a reduction in under-five mortalities to at least 25/1000 live births and neonatal mortality to at least 12 per 1000 live births by 2030 [1, 2]. Despite remarkable progress having been achieved in the reduction of global under five mortalities in the past three decades, the rate of death is still high in sub-Saharan Africa and south Asian countries (4.2 million deaths as of 2019) [3]. While the two regions (sub-Saharan Africa and south Asia) alone contributed to 80% of global under five moralities in 2019, about half of these global deaths occurred in just five countries: Nigeria, India, Pakistan, Democratic Republic of the Congo, and Ethiopia [3]. The 2019 global child mortality estimation result shows that Ethiopia’s progress towards reducing newborns and under five mortality lies at 28 and 50.7 per 1000 live births, compared with 59 and 200 per 1000 live births in 1990 [3]. Although trends in declining child mortality in Ethiopia have been recorded from 1990 to 2019, the rate of both under five and infant mortality is unacceptably high and far from the SDG targets of reducing neonatal mortality to 12 deaths per 1,000 live births and under five mortalities to 25 deaths per 1,000 live births by 2030 [4].\nAccess to child health services remains an important instrument in lowering child mortality rates in order to achieve SDG targets. However, the significant shortage of skilled health professionals poses challenges and inequities in access to basic child health services in low-income countries [5]. The World Health Organization (WHO) suggests the integration of various health professionals in the primary healthcare system, especially in developing countries, to alleviate the shortage of skilled health professionals and to increase access to universal health coverage is needed [6]. Optimizing the roles of community pharmacy professionals in different public health settings, including child health provision could improve both access to services and minimize costs associated with hospital visit [7, 8].\nCommunity pharmacy professionals’ role in primary health care has been increasing globally for the past two decades from being limited to dispensing medications, to involvement in direct patient care for different age groups [8–13]. When it comes to child health, community pharmacy professionals can play prominent roles in service provision, including routine immunization, advice about vitamins, management of minor ailments, counseling about infant milk formulas and chronic disease management [14–17]. Currently, in Europe community pharmacists are an important source of primary care services for children [18]. Further, some developed countries such as the United Kingdom have integrated community pharmacy into primary health care to improve access to various public health services including child health services [19, 20].\nIn developing countries including Ethiopia, evidence regarding the involvement of community pharmacists in child health service provision is limited. Our previously published systematic review conducted in this area identifies that there are few studies from the context of developing countries [21]. Currently, there is paucity in the literature from Ethiopia that focused on the involvement of community pharmacists in public health services in general and the evidence that is available does not include information about their engagement in child health service provisions [22]. The few studies that reported child health services were specific to management of acute diarrhea with no details related to the objectives of our study [23, 24]. Therefore, the aim of this study was to provide evidence on the extent of involvement of community pharmacy professionals in various aspects of child health service provision in Ethiopia. The findings of this study could be used as input for policy development and new initiatives concerning the role of pharmacists in child health service in community pharmacy settings in developing countries in general, and the Amhara region of Ethiopia in particular.", "[SUBTITLE] Study design, study area and sampling [SUBSECTION] This study employed a multicenter cross-sectional survey to assess the level of involvement of community pharmacy professionals in providing child health services in one regional state of Ethiopia. The study was conducted with community pharmacy professionals practicing in Community Drug Retail outlets (CDROs) of six randomly selected zonal cities (out of 11) in the Amhara regional state, Ethiopia. Selected cities were (1) Debre Markos, (2) Gondar, (3) Dessie, (4) Bahir Dar, (5) Debre Tabor and (6) Debre Birhan. The region is the second most populous region among the 11 regional states in Ethiopia with a total projected population of 22,877,365(2022) and an area of 154,709 km2 [25]. As of November 2019, there were 852 active CDROs in Amhara regional state. The total sample needed for the study was calculated using single proportion formula with the assumptions of 5% margin of error, 95% confidence level, and 50% esponse distribution. The sample calculation further employed correction formula given that the total number of sampling frames found in the study region were 852 active CDROs, which is less than 10,000. Following the correction formula, the study sample was 264 community pharmacy professionals practicing in the selected cities.\nThis study employed a multicenter cross-sectional survey to assess the level of involvement of community pharmacy professionals in providing child health services in one regional state of Ethiopia. The study was conducted with community pharmacy professionals practicing in Community Drug Retail outlets (CDROs) of six randomly selected zonal cities (out of 11) in the Amhara regional state, Ethiopia. Selected cities were (1) Debre Markos, (2) Gondar, (3) Dessie, (4) Bahir Dar, (5) Debre Tabor and (6) Debre Birhan. The region is the second most populous region among the 11 regional states in Ethiopia with a total projected population of 22,877,365(2022) and an area of 154,709 km2 [25]. As of November 2019, there were 852 active CDROs in Amhara regional state. The total sample needed for the study was calculated using single proportion formula with the assumptions of 5% margin of error, 95% confidence level, and 50% esponse distribution. The sample calculation further employed correction formula given that the total number of sampling frames found in the study region were 852 active CDROs, which is less than 10,000. Following the correction formula, the study sample was 264 community pharmacy professionals practicing in the selected cities.\n[SUBTITLE] Inclusion and exclusion criteria [SUBSECTION] Qualified and registered pharmacy professionals who had been working for at least three months in the selected CDROs were included in this study. Pharmacy professionals who had less than three months of work experience in CDROs were excluded to reduce under exposure bias, assuming that they may not have had adequate exposure to child health services. Intern pharmacy students were also excluded due to their limited practice.\nQualified and registered pharmacy professionals who had been working for at least three months in the selected CDROs were included in this study. Pharmacy professionals who had less than three months of work experience in CDROs were excluded to reduce under exposure bias, assuming that they may not have had adequate exposure to child health services. Intern pharmacy students were also excluded due to their limited practice.\n[SUBTITLE] Data collection tools and procedures [SUBSECTION] A self-administered questionnaire was developed by the research team from the published literature relevant to the potential role of pharmacists in child health service provision [26–28]. The questionnaire was further reviewed by three experts in pharmacy practice and public health to ensure content validity. Content of the questionnaire was modified according to feedback from the experts. A pilot study was conducted with 10 community pharmacy professionals recruited from two study sites prior to data collection to check the clarity of the survey. Following the pilot study, minor changes in wordings and phrasing were incorporated. There were two parts of the questionnaire: the first part assessed information related to the study participants’ characteristics such as gender, educational achievements, professional licensing level, experience in the CDROs, responsibility in the CDROs, and training experience regarding child health services. The second part of the questionnaire was designed to assess involvement of community pharmacy professionals in child health service provision. The second part of the questionnaire had four items and responses to these questions were recorded on four-point Likert-type scales as ‘very involved’, ‘involved’, ‘little involved’ and ‘not all involved’ with scores of 4,3,2 and 1 respectively. By summing responses to the four items, the maximum score is 16 and the minimum score is four. Higher total scores indicate higher involvement in the provision of child health services. The items pertaining to involvement in service provision demonstrated adequate internal consistency α = 0.775.\nRandom sampling technique was employed to recruit all study participants. Lists of currently functional CDROs in the study sites (selected zonal cities) were requested and accessed from zonal administration health departments. Then the proportions of CDROs needed in each city were calculated considering the total sample size. Equal proportion was not given because the number of CDROs were not equal in each city. From the accessed lists, samples of CDROs were randomly selected to recruit study participants (one pharmacy professional/CDRO). Trained data collectors (pharmacists) recruited study participants randomly from the list of selected CDROs at each study site. Once the data collectors identified the licensed pharmacy professionals in the CDRO, they provided participant information and consent forms. The self-reported survey questionnaires were provided to participants who signed the consent form. Data collection was undertaken between March and July 2020.\nA self-administered questionnaire was developed by the research team from the published literature relevant to the potential role of pharmacists in child health service provision [26–28]. The questionnaire was further reviewed by three experts in pharmacy practice and public health to ensure content validity. Content of the questionnaire was modified according to feedback from the experts. A pilot study was conducted with 10 community pharmacy professionals recruited from two study sites prior to data collection to check the clarity of the survey. Following the pilot study, minor changes in wordings and phrasing were incorporated. There were two parts of the questionnaire: the first part assessed information related to the study participants’ characteristics such as gender, educational achievements, professional licensing level, experience in the CDROs, responsibility in the CDROs, and training experience regarding child health services. The second part of the questionnaire was designed to assess involvement of community pharmacy professionals in child health service provision. The second part of the questionnaire had four items and responses to these questions were recorded on four-point Likert-type scales as ‘very involved’, ‘involved’, ‘little involved’ and ‘not all involved’ with scores of 4,3,2 and 1 respectively. By summing responses to the four items, the maximum score is 16 and the minimum score is four. Higher total scores indicate higher involvement in the provision of child health services. The items pertaining to involvement in service provision demonstrated adequate internal consistency α = 0.775.\nRandom sampling technique was employed to recruit all study participants. Lists of currently functional CDROs in the study sites (selected zonal cities) were requested and accessed from zonal administration health departments. Then the proportions of CDROs needed in each city were calculated considering the total sample size. Equal proportion was not given because the number of CDROs were not equal in each city. From the accessed lists, samples of CDROs were randomly selected to recruit study participants (one pharmacy professional/CDRO). Trained data collectors (pharmacists) recruited study participants randomly from the list of selected CDROs at each study site. Once the data collectors identified the licensed pharmacy professionals in the CDRO, they provided participant information and consent forms. The self-reported survey questionnaires were provided to participants who signed the consent form. Data collection was undertaken between March and July 2020.\n[SUBTITLE] Data analysis [SUBSECTION] The data were analyzed using the Statistical Package for the Social Sciences (SPSS), version 26 (SPSS, Inc., Chicago, IL, USA). While frequencies and percentages were used to summarize categorical variables, means with standard deviations were used to summarize continuous variables. The total involvement score was computed as a sum of the responses scored in each item to construct an approximately continuous variable. Sum of responses of all the ordinal Likert scales in each item is much higher than the scores of the individual items which could result in an approximately continuous variable. Independent sample t-test (for two group variables) and one way Analysis of Variance (ANOVA) was employed to test the mean difference of involvement scores of community pharmacy professionals in child health service provision among subgroups of participant characteristics. Where ANOVAs were significant, Bonferroni post analyses were used to identify a further mean difference. Level of statistical significance was determined at a two-sided p-value < 0.05.\nThe data were analyzed using the Statistical Package for the Social Sciences (SPSS), version 26 (SPSS, Inc., Chicago, IL, USA). While frequencies and percentages were used to summarize categorical variables, means with standard deviations were used to summarize continuous variables. The total involvement score was computed as a sum of the responses scored in each item to construct an approximately continuous variable. Sum of responses of all the ordinal Likert scales in each item is much higher than the scores of the individual items which could result in an approximately continuous variable. Independent sample t-test (for two group variables) and one way Analysis of Variance (ANOVA) was employed to test the mean difference of involvement scores of community pharmacy professionals in child health service provision among subgroups of participant characteristics. Where ANOVAs were significant, Bonferroni post analyses were used to identify a further mean difference. Level of statistical significance was determined at a two-sided p-value < 0.05.", "This study employed a multicenter cross-sectional survey to assess the level of involvement of community pharmacy professionals in providing child health services in one regional state of Ethiopia. The study was conducted with community pharmacy professionals practicing in Community Drug Retail outlets (CDROs) of six randomly selected zonal cities (out of 11) in the Amhara regional state, Ethiopia. Selected cities were (1) Debre Markos, (2) Gondar, (3) Dessie, (4) Bahir Dar, (5) Debre Tabor and (6) Debre Birhan. The region is the second most populous region among the 11 regional states in Ethiopia with a total projected population of 22,877,365(2022) and an area of 154,709 km2 [25]. As of November 2019, there were 852 active CDROs in Amhara regional state. The total sample needed for the study was calculated using single proportion formula with the assumptions of 5% margin of error, 95% confidence level, and 50% esponse distribution. The sample calculation further employed correction formula given that the total number of sampling frames found in the study region were 852 active CDROs, which is less than 10,000. Following the correction formula, the study sample was 264 community pharmacy professionals practicing in the selected cities.", "Qualified and registered pharmacy professionals who had been working for at least three months in the selected CDROs were included in this study. Pharmacy professionals who had less than three months of work experience in CDROs were excluded to reduce under exposure bias, assuming that they may not have had adequate exposure to child health services. Intern pharmacy students were also excluded due to their limited practice.", "A self-administered questionnaire was developed by the research team from the published literature relevant to the potential role of pharmacists in child health service provision [26–28]. The questionnaire was further reviewed by three experts in pharmacy practice and public health to ensure content validity. Content of the questionnaire was modified according to feedback from the experts. A pilot study was conducted with 10 community pharmacy professionals recruited from two study sites prior to data collection to check the clarity of the survey. Following the pilot study, minor changes in wordings and phrasing were incorporated. There were two parts of the questionnaire: the first part assessed information related to the study participants’ characteristics such as gender, educational achievements, professional licensing level, experience in the CDROs, responsibility in the CDROs, and training experience regarding child health services. The second part of the questionnaire was designed to assess involvement of community pharmacy professionals in child health service provision. The second part of the questionnaire had four items and responses to these questions were recorded on four-point Likert-type scales as ‘very involved’, ‘involved’, ‘little involved’ and ‘not all involved’ with scores of 4,3,2 and 1 respectively. By summing responses to the four items, the maximum score is 16 and the minimum score is four. Higher total scores indicate higher involvement in the provision of child health services. The items pertaining to involvement in service provision demonstrated adequate internal consistency α = 0.775.\nRandom sampling technique was employed to recruit all study participants. Lists of currently functional CDROs in the study sites (selected zonal cities) were requested and accessed from zonal administration health departments. Then the proportions of CDROs needed in each city were calculated considering the total sample size. Equal proportion was not given because the number of CDROs were not equal in each city. From the accessed lists, samples of CDROs were randomly selected to recruit study participants (one pharmacy professional/CDRO). Trained data collectors (pharmacists) recruited study participants randomly from the list of selected CDROs at each study site. Once the data collectors identified the licensed pharmacy professionals in the CDRO, they provided participant information and consent forms. The self-reported survey questionnaires were provided to participants who signed the consent form. Data collection was undertaken between March and July 2020.", "The data were analyzed using the Statistical Package for the Social Sciences (SPSS), version 26 (SPSS, Inc., Chicago, IL, USA). While frequencies and percentages were used to summarize categorical variables, means with standard deviations were used to summarize continuous variables. The total involvement score was computed as a sum of the responses scored in each item to construct an approximately continuous variable. Sum of responses of all the ordinal Likert scales in each item is much higher than the scores of the individual items which could result in an approximately continuous variable. Independent sample t-test (for two group variables) and one way Analysis of Variance (ANOVA) was employed to test the mean difference of involvement scores of community pharmacy professionals in child health service provision among subgroups of participant characteristics. Where ANOVAs were significant, Bonferroni post analyses were used to identify a further mean difference. Level of statistical significance was determined at a two-sided p-value < 0.05.", "From the total of 264 community pharmacy professionals approached, 238 participants completed the questionnaires: a response rate of 90%. Characteristics of the study participants are presented in Table 1.\n\nTable 1Characteristics of the community pharmacy professionals included in the studyCharacteristicsTotal (n = 238)Characteristics of the community pharmacy professionalsN (%)\nSex\nMale117(49.2)Female121(50.8)\nEducational qualification in Pharmacy\nDiploma in Pharmacy130(54.6)Bachelor of Pharmacy (BPharm)91(38.2)Master of Pharmacy (MSc)17(7.1)\nWork experience in years (Mean, SD)\n6.05 ± 5.49Less than 5 years130(54.6)5–10 years40(16.8)Greater than 10 years68(28.6)\nLicensure by regulatory Authority\nDruggist/Pharmacy technician96(40.3)Junior Pharmacist48(20.2)Senior Pharmacist/Druggist45(18.9)Chief Pharmacist19(8.0)Expert Pharmacist30(12.6)\nFacility (CDRO)type\nDrug store106(44.5)Pharmacy132(55.5)\nResponsibility in the CDRO\nOwner96(40.3)Employed142(59.7)\nHave you received any in -services training regarding maternal and child health services delivery in CDROs?\nYes42(17.6)No196(82.4)\n\nCharacteristics of the community pharmacy professionals included in the study", "As presented in Table 2, community pharmacy professionals had the highest mean involvement score in ‘advising about vitamins/ supplements’ to children (mean = 3.24, SD = 0.717). The percentage distribution also showed that most community pharmacy professionals reported that they have been either ‘very involved’ (39.5%) or ‘involved’ (46.6%) in ‘advising about vitamins/ supplements’ to children. The second highest mean involvement score of community pharmacy professionals was in providing child health services related to ‘advising about infant formulas /milk’, with 37% ‘very involved’ and 47.1% ‘involved’. Most community pharmacy professionals also reported involvement in child health service provision related to ‘responding to minor symptoms’, where more than 50% of the participants reported being ‘involved’. The lowest mean involvement score was provision of child health service related to ‘vaccination’ (mean = 2.65, SD = 0.923). Compared to the other child health services, a higher proportion of participants were a ‘little involved’ (30.7%) and ‘not at all involved’ (11.8%) in providing services related to vaccinations. Additional details are provided in Table 2.\n\nTable 2Percentage distribution and mean levels of involvement of community pharmacy professionals in child health servicesChild health serviceVery involved, n (%)Involved, n (%)Little involved n (%)Not at all involved, n (%)Mean involvement scoreStd. DeviationAdvising about vitamins/ supplements94(39.5)111(46.6)30(12.6)3(1.3)3.240.717Provision of vaccination/ Checking the child’s vaccination status and refer for vaccination/ provide child vaccination advice to the parent46(19.3)91(38.2)73(30.7)28(11.8)2.650.923Responding to minor symptoms (fever, acute diarrhea, respiratory symptoms)79(33.2)121(50.8)32(13.4)6(2.5)3.150.740Advising about infant formulas /milk88(37.0)112(47.1)30(12.6)8(3.4)3.180.776Overall mean involvement score12.222.452\n\nPercentage distribution and mean levels of involvement of community pharmacy professionals in child health services", "As depicted in Table 3, significant mean difference of involvement in child health service provision was observed among different sub-groups of pharmacists. To be more specific, significant differences in mean involvement was observed based on educational qualifications in Pharmacy, professional licensure levels, Facility (CDRO)types, responsibilities in the CDROs and in services training status. A significantly higher level of pharmacy professionals’ involvement in child health service provision was observed among those working in ‘pharmacy’ settings compared to those working in ‘drug store’ settings. Involvement of community pharmacy professionals in child health service provision was also significantly higher among CDRO ‘owners’ in comparison to their counterparts (‘employees’). Additionally, a significant difference in involvement was also observed based on ‘in-service training status’, with less involvement in child health service provision observed among those who did not report having received training about child health service provision compared to those who had received training. The post-hoc analyses (see Table 4) showed that significant differences in mean involvement score was observed among sub-groups of the study participants based on professional ‘licensure level’. Involvement scores in child health services provision were higher among community pharmacy professionals with licensure level of ‘senior pharmacist’ and ‘expert pharmacist’ compared to pharmacy professionals with a licensure level of ‘druggist/ pharmacy technicians’.\n\nTable 3Total involvement of community pharmacy professionals in provision of child health service among different subgroups of respondents’: Independent sample t-test and one way ANOVADemographic and related characteristicsCategoriesInvolvement in child health service\nMean ± SD\n\nP-value- mean difference\n\nP-value- Levene’s Test for Equality of Variances\n\nSex\nMale12.38 ± 2.4800.3310.557Female12.07 ± 2.425\nEducational qualification in Pharmacy\nDiploma in Pharmacy11.85 ± 2.5940.034*0.450Bachelor of Pharmacy (BPharm)12.59 ± 2.196Master of Pharmacy (MSc)13.00 ± 2.264\nWork experience in years\nLess than 5 years11.89 ± 2.4280.0740.7015–10 years12.67 ± 2.506Greater than 10 years12.50 ± 2.311\nLicensure by regulatory Authority\nDruggist/Pharmacy technician11.39 ± 2.114< 0.001*0.124Junior Pharmacist12.38 ± 2.878Senior Pharmacist/Druggist13.16 ± 2.256Chief Pharmacist12.79 ± 2.440Expert Pharmacist12.87 ± 2.300\nFacility (CDRO)type\nDrug store11.72 ± 2.5440.004*0.973Pharmacy12.62 ± 2.307\nResponsibility in the CDROs\nOwner12.79 ± 2.1900.003*0.158Employed11.83 ± 2.549\nIn -services training received\nYes13.38 ± 2.3370.001*0.868No11.97 ± 2.409*The mean difference is significant at the 0.05 level\n\nTotal involvement of community pharmacy professionals in provision of child health service among different subgroups of respondents’: Independent sample t-test and one way ANOVA\n*The mean difference is significant at the 0.05 level\n\nTable 4Post hoc analyses of factors influencing community pharmacy pharmacists’ involvement in child health servicesVariablesFactorsGroupMean differenceP-value95% Confidence Interval\nLower Bound\n\nLower Bound\nEducational qualification in PharmacyDiploma in pharmacyBachelor of Pharmacy (BPharm)− 0.7400.080-1.540.06Master of Pharmacy (MSc)-1.1460.205-2.660.36Bachelor of Pharmacy (BPharm)Diploma in pharmacy0.7400.080− 0.061.54Master of Pharmacy (MSc)− 0.4071.000-1.951.14Master of Pharmacy (MSc)Diploma in pharmacy1.1460.205− 0.362.66Bachelor of Pharmacy (BPharm)0.4071.000-1.141.95Licensure by regulatory AuthorityDruggist/Pharmacy technicianJunior Pharmacist− 0.9900.185-2.170.19Senior Pharmacist-1.770*< 0.001-2.98− 0.56Chief Pharmacist-1.4040.187-3.080.28Expert Pharmacist-1.481*\n0.030\n-2.88− 0.08Junior PharmacistDruggist/Pharmacy technician0.9900.185− 0.192.17Senior Pharmacist− 0.7811.000-2.170.61Chief Pharmacist− 0.4141.000-2.231.40Expert Pharmacist− 0.4921.000-2.051.07Senior PharmacistDruggist/Pharmacy technician1.770*< 0.0010.562.98Junior Pharmacist0.7811.000− 0.612.17Chief Pharmacist0.3661.000-1.462.20Expert Pharmacist0.2891.000-1.291.87Chief PharmacistDruggist/Pharmacy technician1.4040.187− 0.283.08Junior Pharmacist0.4141.000-1.402.23Senior Pharmacist− 0.3661.000-2.201.46Expert Pharmacist− 0.0771.000-2.041.88Expert PharmacistDruggist/Pharmacy technician1.481*0.0300.082.88Junior Pharmacist0.4921.000-1.072.05Senior Pharmacist− 0.2891.000-1.871.29Chief Pharmacist0.0771.000-1.882.04* The mean difference is significant at the 0.05 level\n\nPost hoc analyses of factors influencing community pharmacy pharmacists’ involvement in child health services\n* The mean difference is significant at the 0.05 level", "We acknowledge that this study has limitations to be considered while interpreting the results. Firstly, the cross-sectional nature of the study design cannot show trends of changes over time among community pharmacy professionals’ involvement in child health service delivery. Secondly, the findings are based on self-reported responses which is susceptible to social desirability bias. However, despite these limitations, the data in this study were generated from multiple centers that employed appropriate sampling techniques in order to fill evidence gaps in the area. Although this study was conducted in one region (Amhara regional state), the findings could be representative to other regions of the country considering the similarity in pharmacy professionals and CDROs regulations. This is because issues related with health professionals and health facilities including pharmacists and CDROs in Ethiopia are mainly regulated centrally by Ethiopian food and drug authority.", "There are no clear directives or policy that supports the expanded role of community pharmacy professionals in providing different public health services including child health services in Ethiopia. Therefore, taking the potential role of pharmacy professionals in child health delivery into consideration, development of national policy that clearly defines how pharmacy professionals could engage in providing various types of child health services in community pharmacy settings in Ethiopia is needed. In addition, to maximize the benefits of community pharmacy-based child health service delivery, providing capacity building training and empowerment is needed to address the potential gaps in practice. The findings of this study have provided some insights about the potential role of community pharmacy professionals in child health service delivery in Ethiopia and developing countries in general. Exploring the barriers and facilitators to community pharmacy professional in providing child health services could be an area of future research." ]
[ null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Study design, study area and sampling", "Inclusion and exclusion criteria", "Data collection tools and procedures", "Data analysis", "Results", "Characteristics of the study participants", "Involvement of community pharmacy professionals in child health services provision", "Difference in the involvement of community pharmacy professionals in child health provision based on sub-groups of respondents’ characteristics", "Discussion", "Strengths and limitations of the study", "Implications for policy, future practice, and research", "Conclusion" ]
[ "Lowering child morality is a global priority with the United Nations (UN) Sustainable Development Goal (SDG) 3.2 calling for a reduction in under-five mortalities to at least 25/1000 live births and neonatal mortality to at least 12 per 1000 live births by 2030 [1, 2]. Despite remarkable progress having been achieved in the reduction of global under five mortalities in the past three decades, the rate of death is still high in sub-Saharan Africa and south Asian countries (4.2 million deaths as of 2019) [3]. While the two regions (sub-Saharan Africa and south Asia) alone contributed to 80% of global under five moralities in 2019, about half of these global deaths occurred in just five countries: Nigeria, India, Pakistan, Democratic Republic of the Congo, and Ethiopia [3]. The 2019 global child mortality estimation result shows that Ethiopia’s progress towards reducing newborns and under five mortality lies at 28 and 50.7 per 1000 live births, compared with 59 and 200 per 1000 live births in 1990 [3]. Although trends in declining child mortality in Ethiopia have been recorded from 1990 to 2019, the rate of both under five and infant mortality is unacceptably high and far from the SDG targets of reducing neonatal mortality to 12 deaths per 1,000 live births and under five mortalities to 25 deaths per 1,000 live births by 2030 [4].\nAccess to child health services remains an important instrument in lowering child mortality rates in order to achieve SDG targets. However, the significant shortage of skilled health professionals poses challenges and inequities in access to basic child health services in low-income countries [5]. The World Health Organization (WHO) suggests the integration of various health professionals in the primary healthcare system, especially in developing countries, to alleviate the shortage of skilled health professionals and to increase access to universal health coverage is needed [6]. Optimizing the roles of community pharmacy professionals in different public health settings, including child health provision could improve both access to services and minimize costs associated with hospital visit [7, 8].\nCommunity pharmacy professionals’ role in primary health care has been increasing globally for the past two decades from being limited to dispensing medications, to involvement in direct patient care for different age groups [8–13]. When it comes to child health, community pharmacy professionals can play prominent roles in service provision, including routine immunization, advice about vitamins, management of minor ailments, counseling about infant milk formulas and chronic disease management [14–17]. Currently, in Europe community pharmacists are an important source of primary care services for children [18]. Further, some developed countries such as the United Kingdom have integrated community pharmacy into primary health care to improve access to various public health services including child health services [19, 20].\nIn developing countries including Ethiopia, evidence regarding the involvement of community pharmacists in child health service provision is limited. Our previously published systematic review conducted in this area identifies that there are few studies from the context of developing countries [21]. Currently, there is paucity in the literature from Ethiopia that focused on the involvement of community pharmacists in public health services in general and the evidence that is available does not include information about their engagement in child health service provisions [22]. The few studies that reported child health services were specific to management of acute diarrhea with no details related to the objectives of our study [23, 24]. Therefore, the aim of this study was to provide evidence on the extent of involvement of community pharmacy professionals in various aspects of child health service provision in Ethiopia. The findings of this study could be used as input for policy development and new initiatives concerning the role of pharmacists in child health service in community pharmacy settings in developing countries in general, and the Amhara region of Ethiopia in particular.", "[SUBTITLE] Study design, study area and sampling [SUBSECTION] This study employed a multicenter cross-sectional survey to assess the level of involvement of community pharmacy professionals in providing child health services in one regional state of Ethiopia. The study was conducted with community pharmacy professionals practicing in Community Drug Retail outlets (CDROs) of six randomly selected zonal cities (out of 11) in the Amhara regional state, Ethiopia. Selected cities were (1) Debre Markos, (2) Gondar, (3) Dessie, (4) Bahir Dar, (5) Debre Tabor and (6) Debre Birhan. The region is the second most populous region among the 11 regional states in Ethiopia with a total projected population of 22,877,365(2022) and an area of 154,709 km2 [25]. As of November 2019, there were 852 active CDROs in Amhara regional state. The total sample needed for the study was calculated using single proportion formula with the assumptions of 5% margin of error, 95% confidence level, and 50% esponse distribution. The sample calculation further employed correction formula given that the total number of sampling frames found in the study region were 852 active CDROs, which is less than 10,000. Following the correction formula, the study sample was 264 community pharmacy professionals practicing in the selected cities.\nThis study employed a multicenter cross-sectional survey to assess the level of involvement of community pharmacy professionals in providing child health services in one regional state of Ethiopia. The study was conducted with community pharmacy professionals practicing in Community Drug Retail outlets (CDROs) of six randomly selected zonal cities (out of 11) in the Amhara regional state, Ethiopia. Selected cities were (1) Debre Markos, (2) Gondar, (3) Dessie, (4) Bahir Dar, (5) Debre Tabor and (6) Debre Birhan. The region is the second most populous region among the 11 regional states in Ethiopia with a total projected population of 22,877,365(2022) and an area of 154,709 km2 [25]. As of November 2019, there were 852 active CDROs in Amhara regional state. The total sample needed for the study was calculated using single proportion formula with the assumptions of 5% margin of error, 95% confidence level, and 50% esponse distribution. The sample calculation further employed correction formula given that the total number of sampling frames found in the study region were 852 active CDROs, which is less than 10,000. Following the correction formula, the study sample was 264 community pharmacy professionals practicing in the selected cities.\n[SUBTITLE] Inclusion and exclusion criteria [SUBSECTION] Qualified and registered pharmacy professionals who had been working for at least three months in the selected CDROs were included in this study. Pharmacy professionals who had less than three months of work experience in CDROs were excluded to reduce under exposure bias, assuming that they may not have had adequate exposure to child health services. Intern pharmacy students were also excluded due to their limited practice.\nQualified and registered pharmacy professionals who had been working for at least three months in the selected CDROs were included in this study. Pharmacy professionals who had less than three months of work experience in CDROs were excluded to reduce under exposure bias, assuming that they may not have had adequate exposure to child health services. Intern pharmacy students were also excluded due to their limited practice.\n[SUBTITLE] Data collection tools and procedures [SUBSECTION] A self-administered questionnaire was developed by the research team from the published literature relevant to the potential role of pharmacists in child health service provision [26–28]. The questionnaire was further reviewed by three experts in pharmacy practice and public health to ensure content validity. Content of the questionnaire was modified according to feedback from the experts. A pilot study was conducted with 10 community pharmacy professionals recruited from two study sites prior to data collection to check the clarity of the survey. Following the pilot study, minor changes in wordings and phrasing were incorporated. There were two parts of the questionnaire: the first part assessed information related to the study participants’ characteristics such as gender, educational achievements, professional licensing level, experience in the CDROs, responsibility in the CDROs, and training experience regarding child health services. The second part of the questionnaire was designed to assess involvement of community pharmacy professionals in child health service provision. The second part of the questionnaire had four items and responses to these questions were recorded on four-point Likert-type scales as ‘very involved’, ‘involved’, ‘little involved’ and ‘not all involved’ with scores of 4,3,2 and 1 respectively. By summing responses to the four items, the maximum score is 16 and the minimum score is four. Higher total scores indicate higher involvement in the provision of child health services. The items pertaining to involvement in service provision demonstrated adequate internal consistency α = 0.775.\nRandom sampling technique was employed to recruit all study participants. Lists of currently functional CDROs in the study sites (selected zonal cities) were requested and accessed from zonal administration health departments. Then the proportions of CDROs needed in each city were calculated considering the total sample size. Equal proportion was not given because the number of CDROs were not equal in each city. From the accessed lists, samples of CDROs were randomly selected to recruit study participants (one pharmacy professional/CDRO). Trained data collectors (pharmacists) recruited study participants randomly from the list of selected CDROs at each study site. Once the data collectors identified the licensed pharmacy professionals in the CDRO, they provided participant information and consent forms. The self-reported survey questionnaires were provided to participants who signed the consent form. Data collection was undertaken between March and July 2020.\nA self-administered questionnaire was developed by the research team from the published literature relevant to the potential role of pharmacists in child health service provision [26–28]. The questionnaire was further reviewed by three experts in pharmacy practice and public health to ensure content validity. Content of the questionnaire was modified according to feedback from the experts. A pilot study was conducted with 10 community pharmacy professionals recruited from two study sites prior to data collection to check the clarity of the survey. Following the pilot study, minor changes in wordings and phrasing were incorporated. There were two parts of the questionnaire: the first part assessed information related to the study participants’ characteristics such as gender, educational achievements, professional licensing level, experience in the CDROs, responsibility in the CDROs, and training experience regarding child health services. The second part of the questionnaire was designed to assess involvement of community pharmacy professionals in child health service provision. The second part of the questionnaire had four items and responses to these questions were recorded on four-point Likert-type scales as ‘very involved’, ‘involved’, ‘little involved’ and ‘not all involved’ with scores of 4,3,2 and 1 respectively. By summing responses to the four items, the maximum score is 16 and the minimum score is four. Higher total scores indicate higher involvement in the provision of child health services. The items pertaining to involvement in service provision demonstrated adequate internal consistency α = 0.775.\nRandom sampling technique was employed to recruit all study participants. Lists of currently functional CDROs in the study sites (selected zonal cities) were requested and accessed from zonal administration health departments. Then the proportions of CDROs needed in each city were calculated considering the total sample size. Equal proportion was not given because the number of CDROs were not equal in each city. From the accessed lists, samples of CDROs were randomly selected to recruit study participants (one pharmacy professional/CDRO). Trained data collectors (pharmacists) recruited study participants randomly from the list of selected CDROs at each study site. Once the data collectors identified the licensed pharmacy professionals in the CDRO, they provided participant information and consent forms. The self-reported survey questionnaires were provided to participants who signed the consent form. Data collection was undertaken between March and July 2020.\n[SUBTITLE] Data analysis [SUBSECTION] The data were analyzed using the Statistical Package for the Social Sciences (SPSS), version 26 (SPSS, Inc., Chicago, IL, USA). While frequencies and percentages were used to summarize categorical variables, means with standard deviations were used to summarize continuous variables. The total involvement score was computed as a sum of the responses scored in each item to construct an approximately continuous variable. Sum of responses of all the ordinal Likert scales in each item is much higher than the scores of the individual items which could result in an approximately continuous variable. Independent sample t-test (for two group variables) and one way Analysis of Variance (ANOVA) was employed to test the mean difference of involvement scores of community pharmacy professionals in child health service provision among subgroups of participant characteristics. Where ANOVAs were significant, Bonferroni post analyses were used to identify a further mean difference. Level of statistical significance was determined at a two-sided p-value < 0.05.\nThe data were analyzed using the Statistical Package for the Social Sciences (SPSS), version 26 (SPSS, Inc., Chicago, IL, USA). While frequencies and percentages were used to summarize categorical variables, means with standard deviations were used to summarize continuous variables. The total involvement score was computed as a sum of the responses scored in each item to construct an approximately continuous variable. Sum of responses of all the ordinal Likert scales in each item is much higher than the scores of the individual items which could result in an approximately continuous variable. Independent sample t-test (for two group variables) and one way Analysis of Variance (ANOVA) was employed to test the mean difference of involvement scores of community pharmacy professionals in child health service provision among subgroups of participant characteristics. Where ANOVAs were significant, Bonferroni post analyses were used to identify a further mean difference. Level of statistical significance was determined at a two-sided p-value < 0.05.", "This study employed a multicenter cross-sectional survey to assess the level of involvement of community pharmacy professionals in providing child health services in one regional state of Ethiopia. The study was conducted with community pharmacy professionals practicing in Community Drug Retail outlets (CDROs) of six randomly selected zonal cities (out of 11) in the Amhara regional state, Ethiopia. Selected cities were (1) Debre Markos, (2) Gondar, (3) Dessie, (4) Bahir Dar, (5) Debre Tabor and (6) Debre Birhan. The region is the second most populous region among the 11 regional states in Ethiopia with a total projected population of 22,877,365(2022) and an area of 154,709 km2 [25]. As of November 2019, there were 852 active CDROs in Amhara regional state. The total sample needed for the study was calculated using single proportion formula with the assumptions of 5% margin of error, 95% confidence level, and 50% esponse distribution. The sample calculation further employed correction formula given that the total number of sampling frames found in the study region were 852 active CDROs, which is less than 10,000. Following the correction formula, the study sample was 264 community pharmacy professionals practicing in the selected cities.", "Qualified and registered pharmacy professionals who had been working for at least three months in the selected CDROs were included in this study. Pharmacy professionals who had less than three months of work experience in CDROs were excluded to reduce under exposure bias, assuming that they may not have had adequate exposure to child health services. Intern pharmacy students were also excluded due to their limited practice.", "A self-administered questionnaire was developed by the research team from the published literature relevant to the potential role of pharmacists in child health service provision [26–28]. The questionnaire was further reviewed by three experts in pharmacy practice and public health to ensure content validity. Content of the questionnaire was modified according to feedback from the experts. A pilot study was conducted with 10 community pharmacy professionals recruited from two study sites prior to data collection to check the clarity of the survey. Following the pilot study, minor changes in wordings and phrasing were incorporated. There were two parts of the questionnaire: the first part assessed information related to the study participants’ characteristics such as gender, educational achievements, professional licensing level, experience in the CDROs, responsibility in the CDROs, and training experience regarding child health services. The second part of the questionnaire was designed to assess involvement of community pharmacy professionals in child health service provision. The second part of the questionnaire had four items and responses to these questions were recorded on four-point Likert-type scales as ‘very involved’, ‘involved’, ‘little involved’ and ‘not all involved’ with scores of 4,3,2 and 1 respectively. By summing responses to the four items, the maximum score is 16 and the minimum score is four. Higher total scores indicate higher involvement in the provision of child health services. The items pertaining to involvement in service provision demonstrated adequate internal consistency α = 0.775.\nRandom sampling technique was employed to recruit all study participants. Lists of currently functional CDROs in the study sites (selected zonal cities) were requested and accessed from zonal administration health departments. Then the proportions of CDROs needed in each city were calculated considering the total sample size. Equal proportion was not given because the number of CDROs were not equal in each city. From the accessed lists, samples of CDROs were randomly selected to recruit study participants (one pharmacy professional/CDRO). Trained data collectors (pharmacists) recruited study participants randomly from the list of selected CDROs at each study site. Once the data collectors identified the licensed pharmacy professionals in the CDRO, they provided participant information and consent forms. The self-reported survey questionnaires were provided to participants who signed the consent form. Data collection was undertaken between March and July 2020.", "The data were analyzed using the Statistical Package for the Social Sciences (SPSS), version 26 (SPSS, Inc., Chicago, IL, USA). While frequencies and percentages were used to summarize categorical variables, means with standard deviations were used to summarize continuous variables. The total involvement score was computed as a sum of the responses scored in each item to construct an approximately continuous variable. Sum of responses of all the ordinal Likert scales in each item is much higher than the scores of the individual items which could result in an approximately continuous variable. Independent sample t-test (for two group variables) and one way Analysis of Variance (ANOVA) was employed to test the mean difference of involvement scores of community pharmacy professionals in child health service provision among subgroups of participant characteristics. Where ANOVAs were significant, Bonferroni post analyses were used to identify a further mean difference. Level of statistical significance was determined at a two-sided p-value < 0.05.", "[SUBTITLE] Characteristics of the study participants [SUBSECTION] From the total of 264 community pharmacy professionals approached, 238 participants completed the questionnaires: a response rate of 90%. Characteristics of the study participants are presented in Table 1.\n\nTable 1Characteristics of the community pharmacy professionals included in the studyCharacteristicsTotal (n = 238)Characteristics of the community pharmacy professionalsN (%)\nSex\nMale117(49.2)Female121(50.8)\nEducational qualification in Pharmacy\nDiploma in Pharmacy130(54.6)Bachelor of Pharmacy (BPharm)91(38.2)Master of Pharmacy (MSc)17(7.1)\nWork experience in years (Mean, SD)\n6.05 ± 5.49Less than 5 years130(54.6)5–10 years40(16.8)Greater than 10 years68(28.6)\nLicensure by regulatory Authority\nDruggist/Pharmacy technician96(40.3)Junior Pharmacist48(20.2)Senior Pharmacist/Druggist45(18.9)Chief Pharmacist19(8.0)Expert Pharmacist30(12.6)\nFacility (CDRO)type\nDrug store106(44.5)Pharmacy132(55.5)\nResponsibility in the CDRO\nOwner96(40.3)Employed142(59.7)\nHave you received any in -services training regarding maternal and child health services delivery in CDROs?\nYes42(17.6)No196(82.4)\n\nCharacteristics of the community pharmacy professionals included in the study\nFrom the total of 264 community pharmacy professionals approached, 238 participants completed the questionnaires: a response rate of 90%. Characteristics of the study participants are presented in Table 1.\n\nTable 1Characteristics of the community pharmacy professionals included in the studyCharacteristicsTotal (n = 238)Characteristics of the community pharmacy professionalsN (%)\nSex\nMale117(49.2)Female121(50.8)\nEducational qualification in Pharmacy\nDiploma in Pharmacy130(54.6)Bachelor of Pharmacy (BPharm)91(38.2)Master of Pharmacy (MSc)17(7.1)\nWork experience in years (Mean, SD)\n6.05 ± 5.49Less than 5 years130(54.6)5–10 years40(16.8)Greater than 10 years68(28.6)\nLicensure by regulatory Authority\nDruggist/Pharmacy technician96(40.3)Junior Pharmacist48(20.2)Senior Pharmacist/Druggist45(18.9)Chief Pharmacist19(8.0)Expert Pharmacist30(12.6)\nFacility (CDRO)type\nDrug store106(44.5)Pharmacy132(55.5)\nResponsibility in the CDRO\nOwner96(40.3)Employed142(59.7)\nHave you received any in -services training regarding maternal and child health services delivery in CDROs?\nYes42(17.6)No196(82.4)\n\nCharacteristics of the community pharmacy professionals included in the study\n[SUBTITLE] Involvement of community pharmacy professionals in child health services provision [SUBSECTION] As presented in Table 2, community pharmacy professionals had the highest mean involvement score in ‘advising about vitamins/ supplements’ to children (mean = 3.24, SD = 0.717). The percentage distribution also showed that most community pharmacy professionals reported that they have been either ‘very involved’ (39.5%) or ‘involved’ (46.6%) in ‘advising about vitamins/ supplements’ to children. The second highest mean involvement score of community pharmacy professionals was in providing child health services related to ‘advising about infant formulas /milk’, with 37% ‘very involved’ and 47.1% ‘involved’. Most community pharmacy professionals also reported involvement in child health service provision related to ‘responding to minor symptoms’, where more than 50% of the participants reported being ‘involved’. The lowest mean involvement score was provision of child health service related to ‘vaccination’ (mean = 2.65, SD = 0.923). Compared to the other child health services, a higher proportion of participants were a ‘little involved’ (30.7%) and ‘not at all involved’ (11.8%) in providing services related to vaccinations. Additional details are provided in Table 2.\n\nTable 2Percentage distribution and mean levels of involvement of community pharmacy professionals in child health servicesChild health serviceVery involved, n (%)Involved, n (%)Little involved n (%)Not at all involved, n (%)Mean involvement scoreStd. DeviationAdvising about vitamins/ supplements94(39.5)111(46.6)30(12.6)3(1.3)3.240.717Provision of vaccination/ Checking the child’s vaccination status and refer for vaccination/ provide child vaccination advice to the parent46(19.3)91(38.2)73(30.7)28(11.8)2.650.923Responding to minor symptoms (fever, acute diarrhea, respiratory symptoms)79(33.2)121(50.8)32(13.4)6(2.5)3.150.740Advising about infant formulas /milk88(37.0)112(47.1)30(12.6)8(3.4)3.180.776Overall mean involvement score12.222.452\n\nPercentage distribution and mean levels of involvement of community pharmacy professionals in child health services\nAs presented in Table 2, community pharmacy professionals had the highest mean involvement score in ‘advising about vitamins/ supplements’ to children (mean = 3.24, SD = 0.717). The percentage distribution also showed that most community pharmacy professionals reported that they have been either ‘very involved’ (39.5%) or ‘involved’ (46.6%) in ‘advising about vitamins/ supplements’ to children. The second highest mean involvement score of community pharmacy professionals was in providing child health services related to ‘advising about infant formulas /milk’, with 37% ‘very involved’ and 47.1% ‘involved’. Most community pharmacy professionals also reported involvement in child health service provision related to ‘responding to minor symptoms’, where more than 50% of the participants reported being ‘involved’. The lowest mean involvement score was provision of child health service related to ‘vaccination’ (mean = 2.65, SD = 0.923). Compared to the other child health services, a higher proportion of participants were a ‘little involved’ (30.7%) and ‘not at all involved’ (11.8%) in providing services related to vaccinations. Additional details are provided in Table 2.\n\nTable 2Percentage distribution and mean levels of involvement of community pharmacy professionals in child health servicesChild health serviceVery involved, n (%)Involved, n (%)Little involved n (%)Not at all involved, n (%)Mean involvement scoreStd. DeviationAdvising about vitamins/ supplements94(39.5)111(46.6)30(12.6)3(1.3)3.240.717Provision of vaccination/ Checking the child’s vaccination status and refer for vaccination/ provide child vaccination advice to the parent46(19.3)91(38.2)73(30.7)28(11.8)2.650.923Responding to minor symptoms (fever, acute diarrhea, respiratory symptoms)79(33.2)121(50.8)32(13.4)6(2.5)3.150.740Advising about infant formulas /milk88(37.0)112(47.1)30(12.6)8(3.4)3.180.776Overall mean involvement score12.222.452\n\nPercentage distribution and mean levels of involvement of community pharmacy professionals in child health services\n[SUBTITLE] Difference in the involvement of community pharmacy professionals in child health provision based on sub-groups of respondents’ characteristics [SUBSECTION] As depicted in Table 3, significant mean difference of involvement in child health service provision was observed among different sub-groups of pharmacists. To be more specific, significant differences in mean involvement was observed based on educational qualifications in Pharmacy, professional licensure levels, Facility (CDRO)types, responsibilities in the CDROs and in services training status. A significantly higher level of pharmacy professionals’ involvement in child health service provision was observed among those working in ‘pharmacy’ settings compared to those working in ‘drug store’ settings. Involvement of community pharmacy professionals in child health service provision was also significantly higher among CDRO ‘owners’ in comparison to their counterparts (‘employees’). Additionally, a significant difference in involvement was also observed based on ‘in-service training status’, with less involvement in child health service provision observed among those who did not report having received training about child health service provision compared to those who had received training. The post-hoc analyses (see Table 4) showed that significant differences in mean involvement score was observed among sub-groups of the study participants based on professional ‘licensure level’. Involvement scores in child health services provision were higher among community pharmacy professionals with licensure level of ‘senior pharmacist’ and ‘expert pharmacist’ compared to pharmacy professionals with a licensure level of ‘druggist/ pharmacy technicians’.\n\nTable 3Total involvement of community pharmacy professionals in provision of child health service among different subgroups of respondents’: Independent sample t-test and one way ANOVADemographic and related characteristicsCategoriesInvolvement in child health service\nMean ± SD\n\nP-value- mean difference\n\nP-value- Levene’s Test for Equality of Variances\n\nSex\nMale12.38 ± 2.4800.3310.557Female12.07 ± 2.425\nEducational qualification in Pharmacy\nDiploma in Pharmacy11.85 ± 2.5940.034*0.450Bachelor of Pharmacy (BPharm)12.59 ± 2.196Master of Pharmacy (MSc)13.00 ± 2.264\nWork experience in years\nLess than 5 years11.89 ± 2.4280.0740.7015–10 years12.67 ± 2.506Greater than 10 years12.50 ± 2.311\nLicensure by regulatory Authority\nDruggist/Pharmacy technician11.39 ± 2.114< 0.001*0.124Junior Pharmacist12.38 ± 2.878Senior Pharmacist/Druggist13.16 ± 2.256Chief Pharmacist12.79 ± 2.440Expert Pharmacist12.87 ± 2.300\nFacility (CDRO)type\nDrug store11.72 ± 2.5440.004*0.973Pharmacy12.62 ± 2.307\nResponsibility in the CDROs\nOwner12.79 ± 2.1900.003*0.158Employed11.83 ± 2.549\nIn -services training received\nYes13.38 ± 2.3370.001*0.868No11.97 ± 2.409*The mean difference is significant at the 0.05 level\n\nTotal involvement of community pharmacy professionals in provision of child health service among different subgroups of respondents’: Independent sample t-test and one way ANOVA\n*The mean difference is significant at the 0.05 level\n\nTable 4Post hoc analyses of factors influencing community pharmacy pharmacists’ involvement in child health servicesVariablesFactorsGroupMean differenceP-value95% Confidence Interval\nLower Bound\n\nLower Bound\nEducational qualification in PharmacyDiploma in pharmacyBachelor of Pharmacy (BPharm)− 0.7400.080-1.540.06Master of Pharmacy (MSc)-1.1460.205-2.660.36Bachelor of Pharmacy (BPharm)Diploma in pharmacy0.7400.080− 0.061.54Master of Pharmacy (MSc)− 0.4071.000-1.951.14Master of Pharmacy (MSc)Diploma in pharmacy1.1460.205− 0.362.66Bachelor of Pharmacy (BPharm)0.4071.000-1.141.95Licensure by regulatory AuthorityDruggist/Pharmacy technicianJunior Pharmacist− 0.9900.185-2.170.19Senior Pharmacist-1.770*< 0.001-2.98− 0.56Chief Pharmacist-1.4040.187-3.080.28Expert Pharmacist-1.481*\n0.030\n-2.88− 0.08Junior PharmacistDruggist/Pharmacy technician0.9900.185− 0.192.17Senior Pharmacist− 0.7811.000-2.170.61Chief Pharmacist− 0.4141.000-2.231.40Expert Pharmacist− 0.4921.000-2.051.07Senior PharmacistDruggist/Pharmacy technician1.770*< 0.0010.562.98Junior Pharmacist0.7811.000− 0.612.17Chief Pharmacist0.3661.000-1.462.20Expert Pharmacist0.2891.000-1.291.87Chief PharmacistDruggist/Pharmacy technician1.4040.187− 0.283.08Junior Pharmacist0.4141.000-1.402.23Senior Pharmacist− 0.3661.000-2.201.46Expert Pharmacist− 0.0771.000-2.041.88Expert PharmacistDruggist/Pharmacy technician1.481*0.0300.082.88Junior Pharmacist0.4921.000-1.072.05Senior Pharmacist− 0.2891.000-1.871.29Chief Pharmacist0.0771.000-1.882.04* The mean difference is significant at the 0.05 level\n\nPost hoc analyses of factors influencing community pharmacy pharmacists’ involvement in child health services\n* The mean difference is significant at the 0.05 level\nAs depicted in Table 3, significant mean difference of involvement in child health service provision was observed among different sub-groups of pharmacists. To be more specific, significant differences in mean involvement was observed based on educational qualifications in Pharmacy, professional licensure levels, Facility (CDRO)types, responsibilities in the CDROs and in services training status. A significantly higher level of pharmacy professionals’ involvement in child health service provision was observed among those working in ‘pharmacy’ settings compared to those working in ‘drug store’ settings. Involvement of community pharmacy professionals in child health service provision was also significantly higher among CDRO ‘owners’ in comparison to their counterparts (‘employees’). Additionally, a significant difference in involvement was also observed based on ‘in-service training status’, with less involvement in child health service provision observed among those who did not report having received training about child health service provision compared to those who had received training. The post-hoc analyses (see Table 4) showed that significant differences in mean involvement score was observed among sub-groups of the study participants based on professional ‘licensure level’. Involvement scores in child health services provision were higher among community pharmacy professionals with licensure level of ‘senior pharmacist’ and ‘expert pharmacist’ compared to pharmacy professionals with a licensure level of ‘druggist/ pharmacy technicians’.\n\nTable 3Total involvement of community pharmacy professionals in provision of child health service among different subgroups of respondents’: Independent sample t-test and one way ANOVADemographic and related characteristicsCategoriesInvolvement in child health service\nMean ± SD\n\nP-value- mean difference\n\nP-value- Levene’s Test for Equality of Variances\n\nSex\nMale12.38 ± 2.4800.3310.557Female12.07 ± 2.425\nEducational qualification in Pharmacy\nDiploma in Pharmacy11.85 ± 2.5940.034*0.450Bachelor of Pharmacy (BPharm)12.59 ± 2.196Master of Pharmacy (MSc)13.00 ± 2.264\nWork experience in years\nLess than 5 years11.89 ± 2.4280.0740.7015–10 years12.67 ± 2.506Greater than 10 years12.50 ± 2.311\nLicensure by regulatory Authority\nDruggist/Pharmacy technician11.39 ± 2.114< 0.001*0.124Junior Pharmacist12.38 ± 2.878Senior Pharmacist/Druggist13.16 ± 2.256Chief Pharmacist12.79 ± 2.440Expert Pharmacist12.87 ± 2.300\nFacility (CDRO)type\nDrug store11.72 ± 2.5440.004*0.973Pharmacy12.62 ± 2.307\nResponsibility in the CDROs\nOwner12.79 ± 2.1900.003*0.158Employed11.83 ± 2.549\nIn -services training received\nYes13.38 ± 2.3370.001*0.868No11.97 ± 2.409*The mean difference is significant at the 0.05 level\n\nTotal involvement of community pharmacy professionals in provision of child health service among different subgroups of respondents’: Independent sample t-test and one way ANOVA\n*The mean difference is significant at the 0.05 level\n\nTable 4Post hoc analyses of factors influencing community pharmacy pharmacists’ involvement in child health servicesVariablesFactorsGroupMean differenceP-value95% Confidence Interval\nLower Bound\n\nLower Bound\nEducational qualification in PharmacyDiploma in pharmacyBachelor of Pharmacy (BPharm)− 0.7400.080-1.540.06Master of Pharmacy (MSc)-1.1460.205-2.660.36Bachelor of Pharmacy (BPharm)Diploma in pharmacy0.7400.080− 0.061.54Master of Pharmacy (MSc)− 0.4071.000-1.951.14Master of Pharmacy (MSc)Diploma in pharmacy1.1460.205− 0.362.66Bachelor of Pharmacy (BPharm)0.4071.000-1.141.95Licensure by regulatory AuthorityDruggist/Pharmacy technicianJunior Pharmacist− 0.9900.185-2.170.19Senior Pharmacist-1.770*< 0.001-2.98− 0.56Chief Pharmacist-1.4040.187-3.080.28Expert Pharmacist-1.481*\n0.030\n-2.88− 0.08Junior PharmacistDruggist/Pharmacy technician0.9900.185− 0.192.17Senior Pharmacist− 0.7811.000-2.170.61Chief Pharmacist− 0.4141.000-2.231.40Expert Pharmacist− 0.4921.000-2.051.07Senior PharmacistDruggist/Pharmacy technician1.770*< 0.0010.562.98Junior Pharmacist0.7811.000− 0.612.17Chief Pharmacist0.3661.000-1.462.20Expert Pharmacist0.2891.000-1.291.87Chief PharmacistDruggist/Pharmacy technician1.4040.187− 0.283.08Junior Pharmacist0.4141.000-1.402.23Senior Pharmacist− 0.3661.000-2.201.46Expert Pharmacist− 0.0771.000-2.041.88Expert PharmacistDruggist/Pharmacy technician1.481*0.0300.082.88Junior Pharmacist0.4921.000-1.072.05Senior Pharmacist− 0.2891.000-1.871.29Chief Pharmacist0.0771.000-1.882.04* The mean difference is significant at the 0.05 level\n\nPost hoc analyses of factors influencing community pharmacy pharmacists’ involvement in child health services\n* The mean difference is significant at the 0.05 level", "From the total of 264 community pharmacy professionals approached, 238 participants completed the questionnaires: a response rate of 90%. Characteristics of the study participants are presented in Table 1.\n\nTable 1Characteristics of the community pharmacy professionals included in the studyCharacteristicsTotal (n = 238)Characteristics of the community pharmacy professionalsN (%)\nSex\nMale117(49.2)Female121(50.8)\nEducational qualification in Pharmacy\nDiploma in Pharmacy130(54.6)Bachelor of Pharmacy (BPharm)91(38.2)Master of Pharmacy (MSc)17(7.1)\nWork experience in years (Mean, SD)\n6.05 ± 5.49Less than 5 years130(54.6)5–10 years40(16.8)Greater than 10 years68(28.6)\nLicensure by regulatory Authority\nDruggist/Pharmacy technician96(40.3)Junior Pharmacist48(20.2)Senior Pharmacist/Druggist45(18.9)Chief Pharmacist19(8.0)Expert Pharmacist30(12.6)\nFacility (CDRO)type\nDrug store106(44.5)Pharmacy132(55.5)\nResponsibility in the CDRO\nOwner96(40.3)Employed142(59.7)\nHave you received any in -services training regarding maternal and child health services delivery in CDROs?\nYes42(17.6)No196(82.4)\n\nCharacteristics of the community pharmacy professionals included in the study", "As presented in Table 2, community pharmacy professionals had the highest mean involvement score in ‘advising about vitamins/ supplements’ to children (mean = 3.24, SD = 0.717). The percentage distribution also showed that most community pharmacy professionals reported that they have been either ‘very involved’ (39.5%) or ‘involved’ (46.6%) in ‘advising about vitamins/ supplements’ to children. The second highest mean involvement score of community pharmacy professionals was in providing child health services related to ‘advising about infant formulas /milk’, with 37% ‘very involved’ and 47.1% ‘involved’. Most community pharmacy professionals also reported involvement in child health service provision related to ‘responding to minor symptoms’, where more than 50% of the participants reported being ‘involved’. The lowest mean involvement score was provision of child health service related to ‘vaccination’ (mean = 2.65, SD = 0.923). Compared to the other child health services, a higher proportion of participants were a ‘little involved’ (30.7%) and ‘not at all involved’ (11.8%) in providing services related to vaccinations. Additional details are provided in Table 2.\n\nTable 2Percentage distribution and mean levels of involvement of community pharmacy professionals in child health servicesChild health serviceVery involved, n (%)Involved, n (%)Little involved n (%)Not at all involved, n (%)Mean involvement scoreStd. DeviationAdvising about vitamins/ supplements94(39.5)111(46.6)30(12.6)3(1.3)3.240.717Provision of vaccination/ Checking the child’s vaccination status and refer for vaccination/ provide child vaccination advice to the parent46(19.3)91(38.2)73(30.7)28(11.8)2.650.923Responding to minor symptoms (fever, acute diarrhea, respiratory symptoms)79(33.2)121(50.8)32(13.4)6(2.5)3.150.740Advising about infant formulas /milk88(37.0)112(47.1)30(12.6)8(3.4)3.180.776Overall mean involvement score12.222.452\n\nPercentage distribution and mean levels of involvement of community pharmacy professionals in child health services", "As depicted in Table 3, significant mean difference of involvement in child health service provision was observed among different sub-groups of pharmacists. To be more specific, significant differences in mean involvement was observed based on educational qualifications in Pharmacy, professional licensure levels, Facility (CDRO)types, responsibilities in the CDROs and in services training status. A significantly higher level of pharmacy professionals’ involvement in child health service provision was observed among those working in ‘pharmacy’ settings compared to those working in ‘drug store’ settings. Involvement of community pharmacy professionals in child health service provision was also significantly higher among CDRO ‘owners’ in comparison to their counterparts (‘employees’). Additionally, a significant difference in involvement was also observed based on ‘in-service training status’, with less involvement in child health service provision observed among those who did not report having received training about child health service provision compared to those who had received training. The post-hoc analyses (see Table 4) showed that significant differences in mean involvement score was observed among sub-groups of the study participants based on professional ‘licensure level’. Involvement scores in child health services provision were higher among community pharmacy professionals with licensure level of ‘senior pharmacist’ and ‘expert pharmacist’ compared to pharmacy professionals with a licensure level of ‘druggist/ pharmacy technicians’.\n\nTable 3Total involvement of community pharmacy professionals in provision of child health service among different subgroups of respondents’: Independent sample t-test and one way ANOVADemographic and related characteristicsCategoriesInvolvement in child health service\nMean ± SD\n\nP-value- mean difference\n\nP-value- Levene’s Test for Equality of Variances\n\nSex\nMale12.38 ± 2.4800.3310.557Female12.07 ± 2.425\nEducational qualification in Pharmacy\nDiploma in Pharmacy11.85 ± 2.5940.034*0.450Bachelor of Pharmacy (BPharm)12.59 ± 2.196Master of Pharmacy (MSc)13.00 ± 2.264\nWork experience in years\nLess than 5 years11.89 ± 2.4280.0740.7015–10 years12.67 ± 2.506Greater than 10 years12.50 ± 2.311\nLicensure by regulatory Authority\nDruggist/Pharmacy technician11.39 ± 2.114< 0.001*0.124Junior Pharmacist12.38 ± 2.878Senior Pharmacist/Druggist13.16 ± 2.256Chief Pharmacist12.79 ± 2.440Expert Pharmacist12.87 ± 2.300\nFacility (CDRO)type\nDrug store11.72 ± 2.5440.004*0.973Pharmacy12.62 ± 2.307\nResponsibility in the CDROs\nOwner12.79 ± 2.1900.003*0.158Employed11.83 ± 2.549\nIn -services training received\nYes13.38 ± 2.3370.001*0.868No11.97 ± 2.409*The mean difference is significant at the 0.05 level\n\nTotal involvement of community pharmacy professionals in provision of child health service among different subgroups of respondents’: Independent sample t-test and one way ANOVA\n*The mean difference is significant at the 0.05 level\n\nTable 4Post hoc analyses of factors influencing community pharmacy pharmacists’ involvement in child health servicesVariablesFactorsGroupMean differenceP-value95% Confidence Interval\nLower Bound\n\nLower Bound\nEducational qualification in PharmacyDiploma in pharmacyBachelor of Pharmacy (BPharm)− 0.7400.080-1.540.06Master of Pharmacy (MSc)-1.1460.205-2.660.36Bachelor of Pharmacy (BPharm)Diploma in pharmacy0.7400.080− 0.061.54Master of Pharmacy (MSc)− 0.4071.000-1.951.14Master of Pharmacy (MSc)Diploma in pharmacy1.1460.205− 0.362.66Bachelor of Pharmacy (BPharm)0.4071.000-1.141.95Licensure by regulatory AuthorityDruggist/Pharmacy technicianJunior Pharmacist− 0.9900.185-2.170.19Senior Pharmacist-1.770*< 0.001-2.98− 0.56Chief Pharmacist-1.4040.187-3.080.28Expert Pharmacist-1.481*\n0.030\n-2.88− 0.08Junior PharmacistDruggist/Pharmacy technician0.9900.185− 0.192.17Senior Pharmacist− 0.7811.000-2.170.61Chief Pharmacist− 0.4141.000-2.231.40Expert Pharmacist− 0.4921.000-2.051.07Senior PharmacistDruggist/Pharmacy technician1.770*< 0.0010.562.98Junior Pharmacist0.7811.000− 0.612.17Chief Pharmacist0.3661.000-1.462.20Expert Pharmacist0.2891.000-1.291.87Chief PharmacistDruggist/Pharmacy technician1.4040.187− 0.283.08Junior Pharmacist0.4141.000-1.402.23Senior Pharmacist− 0.3661.000-2.201.46Expert Pharmacist− 0.0771.000-2.041.88Expert PharmacistDruggist/Pharmacy technician1.481*0.0300.082.88Junior Pharmacist0.4921.000-1.072.05Senior Pharmacist− 0.2891.000-1.871.29Chief Pharmacist0.0771.000-1.882.04* The mean difference is significant at the 0.05 level\n\nPost hoc analyses of factors influencing community pharmacy pharmacists’ involvement in child health services\n* The mean difference is significant at the 0.05 level", "This study highlights the involvement of community pharmacy professionals in providing child health services in Ethiopia. Overall, this study showed that, community pharmacy professionals in Ethiopia have been involved in providing different types of child health services. To be more specific, advice about ‘vitamins/ supplements’, advice about ‘infant formula/milk’ and ‘responding to minor symptoms’ were the child health services most frequently provided by the community pharmacy professionals surveyed. This indicates the growth of community pharmacy professionals’ role and involvement in providing various child health services in general in Ethiopia. Published evidence has also reported the increase in utilization of community pharmacy-based child health service delivery particularly in relation to immunization services, responding to minor ailments and management of chronic disease [14, 16, 18, 29]. Engaging community pharmacy professionals in child health service provision could reduce service demands in clinical settings, particularly in resource limited countries including Ethiopia. For example, managing minor symptoms in children in community pharmacy settings could improve access to the service and reduce costs related to unnecessary hospital visits [7]. In addition, the high involvement of community pharmacy professionals in providing advice about ‘vitamins/ supplements’ and ‘infant formula/milk’ have the potential to increase awareness about fraudulent products among clients. Evidence suggests that fraudulent food products are highly prevalent in developing countries [30]. In tackling these problems, enhancing services of community pharmacy professionals in providing advice about ‘vitamins/ supplements’ and ‘infant formula/milk’ could be an imperative strategy considering their knowledge and professional skills.\nAlthough community pharmacy professionals reported that they have been involved in child health service provision, their reported involvement in certain services such as vaccinations was limited. The low level of involvement in providing childhood vaccination services might be due to lack of policy support by the ministry of health of Ethiopia. This is because in most cases, childhood vaccination services have been delivered in public health facilities. In developing countries including Ethiopia, childhood vaccination uptake is challenging and low in coverage [31]. In such cases, considering their wider availability and long opening hours, enabling community pharmacy professionals to provide vaccination services might be helpful to improve uptake and access [32]. Unlike Ethiopia, there are countries who have authorized community pharmacy professionals as providers of various types of vaccinations for different age groups including children [33–36].\nThe extent of involvement in providing child health services was found to be varied based on community pharmacy professionals’ professional licensure levels, facility types, responsibilities in the CDROs and training status. For example, higher level of involvement was observed among pharmacy professionals who had been working in ‘pharmacy’ settings compared with in a ‘drug store’. The low level of involvement in the drug stores could be associated with regulatory authorizations. In Ethiopia, drug stores are authorized to sell limited types of medications which might limit their future involvement in providing child health services, particularly medication related advice. Similarly, licensure level in pharmacy was found to be linked with variation in level of involvement in overall child health service provision. The low level of involvement in child health service provision among pharmacy professionals with licensure level ‘druggist/ pharmacy technician’ could be linked with the limited exposure to courses relevant to child health during their formal pharmacy education. This is because both the courses and duration of training for pharmacy technicians are less than a bachelor’s degree in pharmacy. However, through additional training and capacity building, pharmacy professionals with lower educational attainment could be empowered to engage in providing child health services. The effectiveness of educational interventions and professional training to improve knowledge and practice skills of pharmacy professionals have been evidenced in the published literature [37, 38].\nMoreover, involvement of community pharmacy professionals in child health service provision was also significantly higher among CDRO ‘owners’ compared with ‘employees’. This can be seen from two perspectives: business motivated involvement and the freedom of choices for action. From the first perspective, the pharmacy owner might not hesitate to provide services thinking of the business he/she can make. The second perspective is related with freedom of practice, where employees might not have full freedom of providing what services.", "We acknowledge that this study has limitations to be considered while interpreting the results. Firstly, the cross-sectional nature of the study design cannot show trends of changes over time among community pharmacy professionals’ involvement in child health service delivery. Secondly, the findings are based on self-reported responses which is susceptible to social desirability bias. However, despite these limitations, the data in this study were generated from multiple centers that employed appropriate sampling techniques in order to fill evidence gaps in the area. Although this study was conducted in one region (Amhara regional state), the findings could be representative to other regions of the country considering the similarity in pharmacy professionals and CDROs regulations. This is because issues related with health professionals and health facilities including pharmacists and CDROs in Ethiopia are mainly regulated centrally by Ethiopian food and drug authority.", "There are no clear directives or policy that supports the expanded role of community pharmacy professionals in providing different public health services including child health services in Ethiopia. Therefore, taking the potential role of pharmacy professionals in child health delivery into consideration, development of national policy that clearly defines how pharmacy professionals could engage in providing various types of child health services in community pharmacy settings in Ethiopia is needed. In addition, to maximize the benefits of community pharmacy-based child health service delivery, providing capacity building training and empowerment is needed to address the potential gaps in practice. The findings of this study have provided some insights about the potential role of community pharmacy professionals in child health service delivery in Ethiopia and developing countries in general. Exploring the barriers and facilitators to community pharmacy professional in providing child health services could be an area of future research.", "Community pharmacy professionals have been involved in providing child health services such as advice about vitamins/supplements, advice about infant milk/formulas and responding to minor symptoms for children. The increasing role of pharmacy professionals beyond merely dispensing of medications has the potential to improve access to child health services. Integrating and enhancing the role of community pharmacists in child health care service delivery could assist countries with their efforts in reducing child mortality and, subsequently, in meeting SDG targets. Empowering community pharmacy professionals through training focusing on child health service provision would also be needed to improve their involvement in the service delivery." ]
[ null, null, null, null, null, null, "results", null, null, null, "discussion", null, null, "conclusion" ]
[ "Child health", "Community drug retails outlet", "Community pharmacy", "Community pharmacy professionals", "Health service" ]
A systematic review of strategies used for controlling consumer moral hazard in health systems.
36258192
Consumer moral hazard refers to an increase in demand for health services or a decrease in preventive care due to insurance coverage. This phenomenon as one of the most evident forms of moral hazard must be reduced and prevented because of its important role in increasing health costs. This study aimed to determine and analyze the strategies used to control consumer moral hazards in health systems.
BACKGROUND
In this systematic review. Web of Sciences, PubMed, Scopus, Embase, ProQuest, Iranian databases(Magiran and SID), and Google Scholar engine were searched using search terms related to moral hazard and healthcare utilization without time limitation. Eligible English and Persian studies on consumer moral hazard in health were included, and papers outside the health and in other languages were excluded. Thematic content analysis was used for data analysis.
METHODS
Content analysis of 68 studies included in the study was presented in the form of two group, six themes, and 11 categories. Two group included "changing behavior at the time of receiving health services" and "changing behavior before needing health services." The first group included four themes: demand-side cost sharing, health savings accounts, drug price regulation, and rationing of health services. The second approach consisted of two themes Development of incentive insurance programs and community empowerment.
RESULTS
Strategies to control consumer moral hazards focus on changing consumer consumptive and health-related behaviors, which are designed according to the structure of health and financing systems. Since "changing consumptive behavior" strategies are the most commonly used strategies; therefore, it is necessary to strengthen strategies to control health-related behaviors and develop new strategies in future studies. In addition, in the application of existing strategies, the adaptation to the structure of the health and financing system, and the pattern of consumption of health services in society should be considered.
CONCLUSION
[ "Humans", "Iran", "Insurance Coverage", "Cost Sharing", "Medical Assistance", "Morals" ]
9580205
null
null
null
null
Results
In the search of databases and other sources, 7488 articles were retrieved, and after removing duplicate sources and applying inclusion and exclusion criteria, 68 eligible articles were selected (Fig. 1). General description of the selected studies is shown in Table 1. As Table 1 shows, out of 68 included papers, the majority of studies were conducted in a period from 2016 to 2021(n = 29), in a quantitative approach (n = 46). And, health service demand and utilization was the most common outcome investigated in these studies. (n = 38). Table 1General description of selected studiesClassification categorySub categoryNumresourcesYear of publication1990–20001[18]2001–200510[19–28]2006–201011[6, 29–38]2011–201517[5, 9, 39–53]2016–202129[10, 13, 15, 54–79]Study designQuantitative46[5,6,9, 13, 20, 22–25, 28, 32, 33, 35–38, 40–46, 48, 51–55, 57, 58, 61–67, 69, 71–76, 78]Qualitative5[26, 27, 39, 49, 70]Review5[10, 15, 47, 56, 60]Theatrical approach(model – based)12[18, 19, 21, 29–31, 34, 50, 59, 68, 77, 79]Outcomes variableDemand & utilization38[5, 15, 20, 22, 23, 28, 30–33, 35, 36, 39–41, 43–48, 52, 54–56, 59, 61, 63–66, 69, 71–74, 77, 78]Cost & optimality26[5, 15, 18, 19, 29–31, 34, 35, 37–40, 50, 51, 53, 57, 58, 60, 68, 71, 73–76, 79]Health related behavior2[13, 46, 50, 77]Opinion & acceptance4[24, 27, 49, 70]Choice of plan2[42, 69] General description of selected studies The content analysis of 68 studies included in the study are presented in the form of two group, six themes, and 11 categories. Since, based on the moral hazard theory, this phenomenon is defined as a change in consumer behavior because of insurance coverage, focusing on changing behavior and modifying it is the main goal of the controlling strategies. Therefore, the results of this study were summarized in the two groups: “Changing behavior at the time of receiving health services” (Table 2) and “Changing behavior before needing health services” (Table 3). The first group includes four themes: demand-side cost-sharing or consumer cost-sharing, health savings accounts, drug price regulation, and rationing for health services. The second group includes two themes: development of incentive insurance programs and community empowerment. The relationship between these two groups is drawn in the form of a diagram (Fig. 2.) In reviewing the findings of the review studies, the results of four primary studies [18–21] were repeated in one review study [22] and the results of one primary study [23] were repeated in another review study [24]. Since in the qualitative analysis, the criteria for analyzing the findings are different from the quantitative results, repeated findings were not excluded in the qualitative analysis, but in the narrative report of the findings of quantitative studies, only the findings of the primary studies were presented. Fig. 2Relationship of strategies of “Changing behavior before needing health services” with strategies of “Changing behavior at the time of receiving health services” Relationship of strategies of “Changing behavior before needing health services” with strategies of “Changing behavior at the time of receiving health services” [SUBTITLE] Strategies for changing behavior at the time of receiving health services [SUBSECTION] The themes (strategies) in this group, which are used when consumers refer to the health system and receive health services by increasing awareness of services costs and other indirect financial consequences (waiting), attempt to increase their responsibility to reduce the consumption of unnecessary health services. The themes of this group consider the changing consumptive behavior of health service consumers. This group includes four themes: demand-side cost-sharing or consumer cost-sharing, health savings accounts, drug price regulation, and rationing for health services. [SUBTITLE] Demand side cost sharing [SUBSECTION] Demand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29]. The results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71]. In our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance. Demand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29]. The results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71]. In our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance. [SUBTITLE] Uniform cost-sharing [SUBSECTION] The uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers. The uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers. [SUBTITLE] Differential cost-sharing [SUBSECTION] In differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies, Strategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount. The studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54]. The reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68]. Despite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50]. In differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies, Strategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount. The studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54]. The reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68]. Despite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50]. The themes (strategies) in this group, which are used when consumers refer to the health system and receive health services by increasing awareness of services costs and other indirect financial consequences (waiting), attempt to increase their responsibility to reduce the consumption of unnecessary health services. The themes of this group consider the changing consumptive behavior of health service consumers. This group includes four themes: demand-side cost-sharing or consumer cost-sharing, health savings accounts, drug price regulation, and rationing for health services. [SUBTITLE] Demand side cost sharing [SUBSECTION] Demand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29]. The results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71]. In our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance. Demand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29]. The results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71]. In our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance. [SUBTITLE] Uniform cost-sharing [SUBSECTION] The uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers. The uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers. [SUBTITLE] Differential cost-sharing [SUBSECTION] In differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies, Strategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount. The studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54]. The reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68]. Despite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50]. In differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies, Strategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount. The studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54]. The reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68]. Despite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50]. [SUBTITLE] Health savings accounts (HSAs) [SUBSECTION] HSAs are considered as alternative tools for financing and dealing with future demographic challenges [72]. This financing theme under the title of health savings accounts [73, 74] or medical savings accounts [72, 75, 76], are currently used to decrease moral hazard and cost in four countries around the world [72, 75]. In this mechanism, owners of savings accounts should save a certain percentage of their income in these accounts for future health expenses. Funds of these accounts are used to pay for health expenses. Saving accounts increase people’s motivation to take responsibility by providing tax benefits and informed participation in health care decisions based on cost awareness and monitoring of physicians’ decisions [73]. In the United States and South Africa, they are used in combination with private insurance and is voluntarily. In Singapore and China, health saving accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system, respectively [72, 75]. This strategy in private health insurance and combination with high deductible health plans (HDHPs) is known as consumer directed health plans or consumer-driven health plans (CDHPs) [59]. In our study, health savings accounts were divided into two categories: voluntary health savings accounts (HSAs) and compulsory health savings accounts (HSAs), which were classified according to the mandatory and optional nature of the plan and the type of financing system in each country. [SUBTITLE] Voluntary health savings accounts [SUBSECTION] In this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75]. In this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75]. [SUBTITLE] Compulsory health savings accounts [SUBSECTION] Compulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75]. Findings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76]. Some experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74]. Compulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75]. Findings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76]. Some experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74]. HSAs are considered as alternative tools for financing and dealing with future demographic challenges [72]. This financing theme under the title of health savings accounts [73, 74] or medical savings accounts [72, 75, 76], are currently used to decrease moral hazard and cost in four countries around the world [72, 75]. In this mechanism, owners of savings accounts should save a certain percentage of their income in these accounts for future health expenses. Funds of these accounts are used to pay for health expenses. Saving accounts increase people’s motivation to take responsibility by providing tax benefits and informed participation in health care decisions based on cost awareness and monitoring of physicians’ decisions [73]. In the United States and South Africa, they are used in combination with private insurance and is voluntarily. In Singapore and China, health saving accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system, respectively [72, 75]. This strategy in private health insurance and combination with high deductible health plans (HDHPs) is known as consumer directed health plans or consumer-driven health plans (CDHPs) [59]. In our study, health savings accounts were divided into two categories: voluntary health savings accounts (HSAs) and compulsory health savings accounts (HSAs), which were classified according to the mandatory and optional nature of the plan and the type of financing system in each country. [SUBTITLE] Voluntary health savings accounts [SUBSECTION] In this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75]. In this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75]. [SUBTITLE] Compulsory health savings accounts [SUBSECTION] Compulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75]. Findings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76]. Some experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74]. Compulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75]. Findings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76]. Some experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74]. [SUBTITLE] Drug price regulation [SUBSECTION] Drug pricing is an influential component of drug access and rational use of drugs. In addition to improving access, consumption management should be considered [77]. Drug price regulation is the third theme of this group, with three categories, uniform pricing, discriminatory pricing and two part pricing, which are based on drug pricing policies and the fix or different prices for each drug unit. As the findings of this theme were extracted from only one study, the categories related to this theme included one code that could not be combined and summarized further due to dissimilarity. [SUBTITLE] Uniform pricing [SUBSECTION] Uniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81]. Uniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81]. [SUBTITLE] Discriminatory pricing [SUBSECTION] Discriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81]. Discriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81]. [SUBTITLE] Two part pricing [SUBSECTION] Two-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79]. Results related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81]. Two-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79]. Results related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81]. Drug pricing is an influential component of drug access and rational use of drugs. In addition to improving access, consumption management should be considered [77]. Drug price regulation is the third theme of this group, with three categories, uniform pricing, discriminatory pricing and two part pricing, which are based on drug pricing policies and the fix or different prices for each drug unit. As the findings of this theme were extracted from only one study, the categories related to this theme included one code that could not be combined and summarized further due to dissimilarity. [SUBTITLE] Uniform pricing [SUBSECTION] Uniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81]. Uniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81]. [SUBTITLE] Discriminatory pricing [SUBSECTION] Discriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81]. Discriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81]. [SUBTITLE] Two part pricing [SUBSECTION] Two-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79]. Results related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81]. Two-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79]. Results related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81]. [SUBTITLE] Rationing of health services [SUBSECTION] Rationing of health services based on the waiting list (number) and waiting time (period) is one of the demand management strategies for non-emergency and elective health services [82]. Rationing of health services is the last theme that has one category named “rationing by waiting” which refers to strategies that control the consumer moral hazard by considering the cost of lost time. This strategy is one of the ways to reduce health costs that replaces user payments in countries without this system (national health system) to control costs and reduces unnecessary demand by imposing costs through the queue [30]. Findings related to the “Rationing of health services” were extracted from three studies [10, 30, 83]. These three studies investigated this mechanism using review and model-based theoretical approaches. The results of the theoretical analysis of these strategies regarding optimality [30] and well-being [83] outcomes were not associated with positive results.These strategies are not very popular and people tend to pay instead of waiting [83]. Table 2Strategies for changing behavior at the time of receiving health servicesTheme/sourceCategoryCodeOutcome variablesDemand side cost sharing[5, 6, 9, 10, 13, 15, 18–20, 22–25, 27–33, 35–49, 51–57, 59–67, 70–73, 78]Uniformcost sharing- Fixed rate of deductible: traditional deductible, mandatory deductible, First euro deductible/ a first-dollar deductible, doughnut hole deductible- Fixed rate of co-payments or coinsurance/co-insurance: /copayment/ user charges / user-fee, lump sum co-payments, fixed payment, Mandatory co-payments- Fixed-rate insurance coverage limit /caps on insurance /stop loss. / payment ceiling limit on coverage, limit on out-of-pocket expenses- Health services utilization- Health costs- Health related behavior- Choice of insurance plan- The opinion and acceptance of consumersDifferentialcost sharing- Income base cost sharing: income base deductible: income-related copayment /coinsurance/ different copayment according to the socioeconomic status- Varied deducible: Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles- Tier cost sharing: :multitier copayments / tiered copayment/ price-related co-payment tier, tier coinsurance- Value-based cost sharing: Value Based Insurance Design (VBID) / value-based cost sharing (lower coinsurance for services with higher costs benefits / value-based coinsurance target / variable co-insurance based on demand elasticity/ treatment-specific copayments disease-specific cost sharing/ differential cost sharing based on disease status- Cost sharing with discount: co-payment exemption, co-payment with rebate, / waiving copaysHealth savings accounts(HSAs)[26, 50, 54, 58, 60, 74, 75]Voluntary Health Savings Accounts (HSAs)- Health savings accounts (HSAs) / Health Reimbursement Accounts (HRAs) / medical savings accounts (MSAs) in combination with private insurance- Consumer-Directed Health Plans (CDHPS) /Consumer-Driven Health Plans (CDHPS) / Three-Tier Payment System(HDHPS Coupled With Personal Savings Account)- Health services utilization- Health costs- Saving for futureCompulsory Health Savings Accounts (HSAs)- Medisave (Medical savings accounts) (MSAs) in combination with social health insurance- Tongdao (MSA )in combination social risk-pooling (SRP)/ medical savings accounts (MSAs) in combination with social insurance pool (SIP)/ the three-tiered design (MSA-deductible-SIP)- Bankuai (MSA) separately to finance outpatient servicesDrug price regulation[21]Uniform pricingdiscriminatory pricing- Uniform pricing/uniform monopoly pricing- Third degree price discrimination / (different prices to different markets or groups) of consumers- Drug utilizationTwo PartPricing- Two-part tariffs.) combines a uniform price with market-specific lump-sum paymentsRationing for health services [10, 31, 34]Rationing by waiting- Waiting lists / queuing /expectancy queue/the cost of the lost time- Waiting time- Optimality- Well-being Strategies for changing behavior at the time of receiving health services Demand side cost sharing [5, 6, 9, 10, 13, 15, 18–20, 22–25, 27–33, 35–49, 51–57, 59–67, 70–73, 78] Uniform cost sharing - Fixed rate of deductible: traditional deductible, mandatory deductible, First euro deductible/ a first-dollar deductible, doughnut hole deductible - Fixed rate of co-payments or coinsurance/co-insurance: /copayment/ user charges / user-fee, lump sum co-payments, fixed payment, Mandatory co-payments - Fixed-rate insurance coverage limit /caps on insurance /stop loss. / payment ceiling limit on coverage, limit on out-of-pocket expenses - Health services utilization - Health costs - Health related behavior - Choice of insurance plan - The opinion and acceptance of consumers Differential cost sharing - Income base cost sharing: income base deductible: income-related copayment /coinsurance/ different copayment according to the socioeconomic status - Varied deducible: Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles - Tier cost sharing: :multitier copayments / tiered copayment/ price-related co-payment tier, tier coinsurance - Value-based cost sharing: Value Based Insurance Design (VBID) / value-based cost sharing (lower coinsurance for services with higher costs benefits / value-based coinsurance target / variable co-insurance based on demand elasticity/ treatment-specific copayments disease-specific cost sharing/ differential cost sharing based on disease status - Cost sharing with discount: co-payment exemption, co-payment with rebate, / waiving copays Health savings accounts (HSAs) [26, 50, 54, 58, 60, 74, 75] - Health savings accounts (HSAs) / Health Reimbursement Accounts (HRAs) / medical savings accounts (MSAs) in combination with private insurance - Consumer-Directed Health Plans (CDHPS) /Consumer-Driven Health Plans (CDHPS) / Three-Tier Payment System(HDHPS Coupled With Personal Savings Account) - Health services utilization - Health costs - Saving for future - Medisave (Medical savings accounts) (MSAs) in combination with social health insurance - Tongdao (MSA )in combination social risk-pooling (SRP)/ medical savings accounts (MSAs) in combination with social insurance pool (SIP)/ the three-tiered design (MSA-deductible-SIP) - Bankuai (MSA) separately to finance outpatient services Drug price regulation [21] Uniform pricing discriminatory pricing - Uniform pricing/uniform monopoly pricing - Third degree price discrimination / (different prices to different markets or groups) of consumers - Waiting lists / queuing /expectancy queue/the cost of the lost time - Waiting time - Optimality - Well-being Rationing of health services based on the waiting list (number) and waiting time (period) is one of the demand management strategies for non-emergency and elective health services [82]. Rationing of health services is the last theme that has one category named “rationing by waiting” which refers to strategies that control the consumer moral hazard by considering the cost of lost time. This strategy is one of the ways to reduce health costs that replaces user payments in countries without this system (national health system) to control costs and reduces unnecessary demand by imposing costs through the queue [30]. Findings related to the “Rationing of health services” were extracted from three studies [10, 30, 83]. These three studies investigated this mechanism using review and model-based theoretical approaches. The results of the theoretical analysis of these strategies regarding optimality [30] and well-being [83] outcomes were not associated with positive results.These strategies are not very popular and people tend to pay instead of waiting [83]. Table 2Strategies for changing behavior at the time of receiving health servicesTheme/sourceCategoryCodeOutcome variablesDemand side cost sharing[5, 6, 9, 10, 13, 15, 18–20, 22–25, 27–33, 35–49, 51–57, 59–67, 70–73, 78]Uniformcost sharing- Fixed rate of deductible: traditional deductible, mandatory deductible, First euro deductible/ a first-dollar deductible, doughnut hole deductible- Fixed rate of co-payments or coinsurance/co-insurance: /copayment/ user charges / user-fee, lump sum co-payments, fixed payment, Mandatory co-payments- Fixed-rate insurance coverage limit /caps on insurance /stop loss. / payment ceiling limit on coverage, limit on out-of-pocket expenses- Health services utilization- Health costs- Health related behavior- Choice of insurance plan- The opinion and acceptance of consumersDifferentialcost sharing- Income base cost sharing: income base deductible: income-related copayment /coinsurance/ different copayment according to the socioeconomic status- Varied deducible: Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles- Tier cost sharing: :multitier copayments / tiered copayment/ price-related co-payment tier, tier coinsurance- Value-based cost sharing: Value Based Insurance Design (VBID) / value-based cost sharing (lower coinsurance for services with higher costs benefits / value-based coinsurance target / variable co-insurance based on demand elasticity/ treatment-specific copayments disease-specific cost sharing/ differential cost sharing based on disease status- Cost sharing with discount: co-payment exemption, co-payment with rebate, / waiving copaysHealth savings accounts(HSAs)[26, 50, 54, 58, 60, 74, 75]Voluntary Health Savings Accounts (HSAs)- Health savings accounts (HSAs) / Health Reimbursement Accounts (HRAs) / medical savings accounts (MSAs) in combination with private insurance- Consumer-Directed Health Plans (CDHPS) /Consumer-Driven Health Plans (CDHPS) / Three-Tier Payment System(HDHPS Coupled With Personal Savings Account)- Health services utilization- Health costs- Saving for futureCompulsory Health Savings Accounts (HSAs)- Medisave (Medical savings accounts) (MSAs) in combination with social health insurance- Tongdao (MSA )in combination social risk-pooling (SRP)/ medical savings accounts (MSAs) in combination with social insurance pool (SIP)/ the three-tiered design (MSA-deductible-SIP)- Bankuai (MSA) separately to finance outpatient servicesDrug price regulation[21]Uniform pricingdiscriminatory pricing- Uniform pricing/uniform monopoly pricing- Third degree price discrimination / (different prices to different markets or groups) of consumers- Drug utilizationTwo PartPricing- Two-part tariffs.) combines a uniform price with market-specific lump-sum paymentsRationing for health services [10, 31, 34]Rationing by waiting- Waiting lists / queuing /expectancy queue/the cost of the lost time- Waiting time- Optimality- Well-being Strategies for changing behavior at the time of receiving health services Demand side cost sharing [5, 6, 9, 10, 13, 15, 18–20, 22–25, 27–33, 35–49, 51–57, 59–67, 70–73, 78] Uniform cost sharing - Fixed rate of deductible: traditional deductible, mandatory deductible, First euro deductible/ a first-dollar deductible, doughnut hole deductible - Fixed rate of co-payments or coinsurance/co-insurance: /copayment/ user charges / user-fee, lump sum co-payments, fixed payment, Mandatory co-payments - Fixed-rate insurance coverage limit /caps on insurance /stop loss. / payment ceiling limit on coverage, limit on out-of-pocket expenses - Health services utilization - Health costs - Health related behavior - Choice of insurance plan - The opinion and acceptance of consumers Differential cost sharing - Income base cost sharing: income base deductible: income-related copayment /coinsurance/ different copayment according to the socioeconomic status - Varied deducible: Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles - Tier cost sharing: :multitier copayments / tiered copayment/ price-related co-payment tier, tier coinsurance - Value-based cost sharing: Value Based Insurance Design (VBID) / value-based cost sharing (lower coinsurance for services with higher costs benefits / value-based coinsurance target / variable co-insurance based on demand elasticity/ treatment-specific copayments disease-specific cost sharing/ differential cost sharing based on disease status - Cost sharing with discount: co-payment exemption, co-payment with rebate, / waiving copays Health savings accounts (HSAs) [26, 50, 54, 58, 60, 74, 75] - Health savings accounts (HSAs) / Health Reimbursement Accounts (HRAs) / medical savings accounts (MSAs) in combination with private insurance - Consumer-Directed Health Plans (CDHPS) /Consumer-Driven Health Plans (CDHPS) / Three-Tier Payment System(HDHPS Coupled With Personal Savings Account) - Health services utilization - Health costs - Saving for future - Medisave (Medical savings accounts) (MSAs) in combination with social health insurance - Tongdao (MSA )in combination social risk-pooling (SRP)/ medical savings accounts (MSAs) in combination with social insurance pool (SIP)/ the three-tiered design (MSA-deductible-SIP) - Bankuai (MSA) separately to finance outpatient services Drug price regulation [21] Uniform pricing discriminatory pricing - Uniform pricing/uniform monopoly pricing - Third degree price discrimination / (different prices to different markets or groups) of consumers - Waiting lists / queuing /expectancy queue/the cost of the lost time - Waiting time - Optimality - Well-being [SUBTITLE] Strategies for changing behavior before needing health services [SUBSECTION] The themes in this group deal with the strategies that are applied outside the health system, before the need for healthcare services, and through the consciousness of health and positive financial incentives, increase healthy behavior or prevent unhealthy behavior. Changing individual behaviors to reduce high-risk behaviors and improve health-promoting behaviors is the approach of this group. This group includes two themes: development of incentive insurance programs and community empowerment. The themes in this group deal with the strategies that are applied outside the health system, before the need for healthcare services, and through the consciousness of health and positive financial incentives, increase healthy behavior or prevent unhealthy behavior. Changing individual behaviors to reduce high-risk behaviors and improve health-promoting behaviors is the approach of this group. This group includes two themes: development of incentive insurance programs and community empowerment. [SUBTITLE] Development of incentive insurance programs [SUBSECTION] The themes of this group focus on the measures of insurance companies and purchasers. This theme refers to strategies aimed at reducing the risk of disease and the need for health services or unhealthy behaviors and consists of two categories: extending preventive care insurance and developing bonus-oriented insurance. Findings related to this theme were extracted from11 studies [10, 15, 25, 37, 43, 46, 58, 84–87]. [SUBTITLE] Extending preventive care insurance [SUBSECTION] Expanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84]. Expanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84]. [SUBTITLE] Developing bonus-oriented insurance [SUBSECTION] These strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58]. Health service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85]. These strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58]. Health service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85]. The themes of this group focus on the measures of insurance companies and purchasers. This theme refers to strategies aimed at reducing the risk of disease and the need for health services or unhealthy behaviors and consists of two categories: extending preventive care insurance and developing bonus-oriented insurance. Findings related to this theme were extracted from11 studies [10, 15, 25, 37, 43, 46, 58, 84–87]. [SUBTITLE] Extending preventive care insurance [SUBSECTION] Expanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84]. Expanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84]. [SUBTITLE] Developing bonus-oriented insurance [SUBSECTION] These strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58]. Health service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85]. These strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58]. Health service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85]. [SUBTITLE] Community empowerment [SUBSECTION] Community empowerment is the second and last theme in this group with one category called Community education.The theme of ”community empowerment”, mentioned by only one study [33]. [SUBTITLE] Community education [SUBSECTION] This category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme. The sources from which each code is extracted are provided in Additional file 3. This category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme. The sources from which each code is extracted are provided in Additional file 3. Community empowerment is the second and last theme in this group with one category called Community education.The theme of ”community empowerment”, mentioned by only one study [33]. [SUBTITLE] Community education [SUBSECTION] This category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme. The sources from which each code is extracted are provided in Additional file 3. This category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme. The sources from which each code is extracted are provided in Additional file 3. [SUBTITLE] Risk of bias consideration [SUBSECTION] The risk of bias assessment in this study consisted of the following: To reduce publication bias, unpublished papers were searched in the Dissertations database of ProQuest for grey literature, but no related papers were found. In this regard, there is a possibility of language bias due to the limitation of non-English articles in publishing or indexing the results and the focus of this study on Persian and English articles, which is mentioned as a limitation in the limitations section. Table 3Strategies for changing behavior before needing health servicesTheme/sourceCategoryCodeOutcome variablesDevelopment of incentive insurance programs[10, 15, 27, 30, 39, 42, 59, 69, 76, 77, 79]Extending preventive care insurance- Proposing insurance coverage for preventive care- Separating insurance coverage for prevention and treatment- To encourage insureds to use more secondary preventive care- Improving perception of health status through secondary preventive care- Health services utilization- Health costs- Risk-reducing behaviors- Choice of expensive health services healthDevelopment of bonus oriented insurance- Adjusted premiums according level of preventive effort- Premium reduction, bonus payments/ rebate insurance /to encourage non-use or limited use in return for next premium reduction, risk adjustment .no-claims bonus, risk rating premium- No-claim Bonus and Coverage Upper BoundCommunity empowerment [23]Community education- Health promotion education- Civic education about the consequences of unnecessary use of health services- Not evaluated Strategies for changing behavior before needing health services Development of incentive insurance programs [10, 15, 27, 30, 39, 42, 59, 69, 76, 77, 79] - Proposing insurance coverage for preventive care - Separating insurance coverage for prevention and treatment - To encourage insureds to use more secondary preventive care - Improving perception of health status through secondary preventive care - Health services utilization - Health costs - Risk-reducing behaviors - Choice of expensive health services health - Adjusted premiums according level of preventive effort - Premium reduction, bonus payments/ rebate insurance /to encourage non-use or limited use in return for next premium reduction, risk adjustment .no-claims bonus, risk rating premium - No-claim Bonus and Coverage Upper Bound - Health promotion education - Civic education about the consequences of unnecessary use of health services The risk of bias assessment in this study consisted of the following: To reduce publication bias, unpublished papers were searched in the Dissertations database of ProQuest for grey literature, but no related papers were found. In this regard, there is a possibility of language bias due to the limitation of non-English articles in publishing or indexing the results and the focus of this study on Persian and English articles, which is mentioned as a limitation in the limitations section. Table 3Strategies for changing behavior before needing health servicesTheme/sourceCategoryCodeOutcome variablesDevelopment of incentive insurance programs[10, 15, 27, 30, 39, 42, 59, 69, 76, 77, 79]Extending preventive care insurance- Proposing insurance coverage for preventive care- Separating insurance coverage for prevention and treatment- To encourage insureds to use more secondary preventive care- Improving perception of health status through secondary preventive care- Health services utilization- Health costs- Risk-reducing behaviors- Choice of expensive health services healthDevelopment of bonus oriented insurance- Adjusted premiums according level of preventive effort- Premium reduction, bonus payments/ rebate insurance /to encourage non-use or limited use in return for next premium reduction, risk adjustment .no-claims bonus, risk rating premium- No-claim Bonus and Coverage Upper BoundCommunity empowerment [23]Community education- Health promotion education- Civic education about the consequences of unnecessary use of health services- Not evaluated Strategies for changing behavior before needing health services Development of incentive insurance programs [10, 15, 27, 30, 39, 42, 59, 69, 76, 77, 79] - Proposing insurance coverage for preventive care - Separating insurance coverage for prevention and treatment - To encourage insureds to use more secondary preventive care - Improving perception of health status through secondary preventive care - Health services utilization - Health costs - Risk-reducing behaviors - Choice of expensive health services health - Adjusted premiums according level of preventive effort - Premium reduction, bonus payments/ rebate insurance /to encourage non-use or limited use in return for next premium reduction, risk adjustment .no-claims bonus, risk rating premium - No-claim Bonus and Coverage Upper Bound - Health promotion education - Civic education about the consequences of unnecessary use of health services
Conclusion
Strategies to control consumer moral hazards focus on changing consumer consumptive and health-related behaviors, which are designed according to the structure of health and financing systems. Since “changing consumptive behavior” strategies are the most commonly used strategies; therefore, it is necessary to strengthen strategies to control health-related behaviors and develop new strategies in future studies. In addition, in the application of existing strategies, the adaptation to the structure of the health and financing system, and the pattern of consumption of health services in society should be considered.
[ "Background", "Methods", "Databases and search strategies", "Inclusion and exclusion criteria", "Methods of screening and selection criteria", "Quality appraisal", "Data analysis", "Strategies for changing behavior at the time of receiving health services", "Demand side cost sharing", "Uniform cost-sharing", "Differential cost-sharing", "Health savings accounts (HSAs)", "Voluntary health savings accounts", "Compulsory health savings accounts", "Drug price regulation", "Uniform pricing", "Discriminatory pricing", "Two part pricing", "Rationing of health services", "Strategies for changing behavior before needing health services", "Development of incentive insurance programs", "Extending preventive care insurance", "Developing bonus-oriented insurance", "Community empowerment", "Community education", "Risk of bias consideration", "Limitations", "" ]
[ "Maximizing the health of people and populations is one of the main goals of any health system which leads to improving personal, social, and economic well-being [1]. The efficient function of the health system depends on providing improved health services at a minimum cost [2]. Evidence shows that health spending is growing faster than economic growth [1, 3]. On average, health spending in OECD countries was equivalent to 9% of GDP in 2018 [1]. The United States spent 18% of its GDP on health care in 2015 [4]. Moral hazard is one of the most important reasons for increasing health costs [5, 6]. Moral hazard is the change in health behavior and consumption of health services because of insurance coverage [7]. According to the theory of moral hazard, health insurance and third-party payers, by lowering the price of care, encourage the consumer to consume more care than when they consume at the market price [8]. Insurance coverage leads to the consumption of health services above an efficient level [9].\nThis phenomenon is classified in different ways: ex-ante moral hazard and ex-post moral hazard, hidden information and hidden action moral hazard, provider moral hazard, and consumer moral hazard [10, 11]. Ex-ante moral hazard occurs before illness and increases a person’s unhealthy behavior; in contrast, ex-post moral hazard occurs after the onset of illness and will increase costs by increasing demand and consuming unnecessary services. In the hidden information, the insurer cannot observe the real condition and severity of the disease to pay the cost according to the real need. In hidden action individuals’ precautionary measures are not visible, and the insured person does not take the necessary precautions to prevent the disease. Provider moral hazard occurs when a provider provides more services to increase its revenue. The provider moral hazard is also known as the provider’s induced demand. Consumer moral hazard means insured people demand more care than uninsured people [11, 12]. Additionally, consumer moral hazard deals with the reduction in preventive healthcare behaviors resulting from insurance coverage[13].\nMoral hazard is known as one of the main causes of market failure [14] that has many adverse consequences, such as the impact on demand elasticity, reduction of welfare, inefficiency in using resources, reduction in technical and allocation efficiency, reduction of benefits of risk pooling, and price increase [11, 15]. Moral hazard, as a concern in the health insurance market, requires the application of appropriate policies and interventions to be controlled. In this regard, consumer moral hazard as the most obvious form of moral hazard [16] has been one of the topics of concern for policymakers and economic experts in recent years [8]. The aim of this study was to determine and analyze strategies used to control consumer moral hazards in health systems. The results of this study can be used for health insurance planning, health system financing, and health cost reduction.", "This study was written as part of a Ph.D thesis entitled “Developing a model to control consumer moral hazard in Iran’s health system” which was designed and performed based on its proposal and was approved by the local ethics committee of the Iran University of Medical Sciences (code: IR.IUMS.REC.1399.1103).\nResearch questions.\n\nWhat strategies or interventions are used to control consumers’ moral hazards?What is the approach of identifying strategies to control moral hazards?\n\nWhat strategies or interventions are used to control consumers’ moral hazards?\nWhat is the approach of identifying strategies to control moral hazards?\n[SUBTITLE] Databases and search strategies [SUBSECTION] For this systematic review, Web of Sciences, PubMed, Scopus, Embase, ProQuest (Dissertations database), and Iranian databases Magiran (the largest Iranian database in various scientific and specialized fields) and SID (open access database to Iranian Persian and English studies) were searched without time limitation, until the seventh of February 2021. In addition, to complete the search and ensure access to all related articles, the Google Scholar search engine was also searched. On July 21, 2022, the mentioned databases were researched to Identify new publications between February 2021and July 2022. During the new search five studies were added.\nSearch terms were used for the titles or abstracts of the records included “moral hazard”, “unnecessary use”, “unnecessary utilization”, “non-essential use”, “non-essential utilization”, “overutilization”, “health”, “health system”, “health insurance”, “health care”, “health service”, “health services”, “healthcare”, “medical care”, and “medical service”. In Web of Sciences, Scopus, and ProQuest due to their defined search strategy, in addition to the titles and abstracts, keywords were also searched. Search in any of the databases was performed using the defined search strategy of each database. The complete search strategy is shown in Additional file 1.\nFor this systematic review, Web of Sciences, PubMed, Scopus, Embase, ProQuest (Dissertations database), and Iranian databases Magiran (the largest Iranian database in various scientific and specialized fields) and SID (open access database to Iranian Persian and English studies) were searched without time limitation, until the seventh of February 2021. In addition, to complete the search and ensure access to all related articles, the Google Scholar search engine was also searched. On July 21, 2022, the mentioned databases were researched to Identify new publications between February 2021and July 2022. During the new search five studies were added.\nSearch terms were used for the titles or abstracts of the records included “moral hazard”, “unnecessary use”, “unnecessary utilization”, “non-essential use”, “non-essential utilization”, “overutilization”, “health”, “health system”, “health insurance”, “health care”, “health service”, “health services”, “healthcare”, “medical care”, and “medical service”. In Web of Sciences, Scopus, and ProQuest due to their defined search strategy, in addition to the titles and abstracts, keywords were also searched. Search in any of the databases was performed using the defined search strategy of each database. The complete search strategy is shown in Additional file 1.\n[SUBTITLE] Inclusion and exclusion criteria [SUBSECTION] Papers in English and Persian languages in the field of reducing and controlling consumer moral hazard in the health system, conducted in a quantitative, qualitative, and mixed methods design with theoretical and empirical approaches, that were of moderate and high quality based on Dixon Wood et al. ‘s checklist [17] were included. Abstracts, letters to the editors, conference, and seminar presentations were excluded.\nPapers in English and Persian languages in the field of reducing and controlling consumer moral hazard in the health system, conducted in a quantitative, qualitative, and mixed methods design with theoretical and empirical approaches, that were of moderate and high quality based on Dixon Wood et al. ‘s checklist [17] were included. Abstracts, letters to the editors, conference, and seminar presentations were excluded.\n[SUBTITLE] Methods of screening and selection criteria [SUBSECTION] All found articles were imported into Endnote software (version X9 (and duplicate articles were removed. Two researchers who were experts in the research topic and systematic review process independently screened the titles and abstracts of the articles (ZKR and MJ). In the final screening step, the full texts of the remaining articles were independently assessed by two researchers. Disagreements between the two researchers were resolved based on the opinion of a third researcher. Finally, the references of the retrieved articles were reviewed to find related articles that were not found in the first search. The screening process of retrieved papers is presented in Figure 1. Data extraction was conducted based on author’s name, title, year of publication, country, study design, strategies used to control consumer moral hazards, outcome variables, main results and quality assessment status. The main characteristics of the included study is shown in Additional file 2.\n\nFig. 1Literature selection and retrieval flow diagram.\n\nLiterature selection and retrieval flow diagram.\nAll found articles were imported into Endnote software (version X9 (and duplicate articles were removed. Two researchers who were experts in the research topic and systematic review process independently screened the titles and abstracts of the articles (ZKR and MJ). In the final screening step, the full texts of the remaining articles were independently assessed by two researchers. Disagreements between the two researchers were resolved based on the opinion of a third researcher. Finally, the references of the retrieved articles were reviewed to find related articles that were not found in the first search. The screening process of retrieved papers is presented in Figure 1. Data extraction was conducted based on author’s name, title, year of publication, country, study design, strategies used to control consumer moral hazards, outcome variables, main results and quality assessment status. The main characteristics of the included study is shown in Additional file 2.\n\nFig. 1Literature selection and retrieval flow diagram.\n\nLiterature selection and retrieval flow diagram.\n[SUBTITLE] Quality appraisal [SUBSECTION] In the first stage, the quality of the articles was assessed by two members of the research team who were familiar with the issue of moral hazard (ZKR and MJ), and the consensus was reached regarding the quality of the selected articles. The most important criteria for selecting high-quality articles were their relevance and role in the development of the study concept. Disagreements were resolved by a third author. The next step in the quality appraisal was based on Dixon-Woods et al. ‘s checklist [17], which included five questions regarding the clarity of the study objective(s), suitability of the study design to the objective(s), presentation of a clear report of the process of generating findings, use of sufficient data to support the interpretations, and use of appropriate analysis methods. The quality of the articles was determined based on 10 scores: 9–10 (high quality), 6–8 (moderate quality), and ≤ 5 (low quality). Articles with a score ≥ 6 were included in the study.\nIn the first stage, the quality of the articles was assessed by two members of the research team who were familiar with the issue of moral hazard (ZKR and MJ), and the consensus was reached regarding the quality of the selected articles. The most important criteria for selecting high-quality articles were their relevance and role in the development of the study concept. Disagreements were resolved by a third author. The next step in the quality appraisal was based on Dixon-Woods et al. ‘s checklist [17], which included five questions regarding the clarity of the study objective(s), suitability of the study design to the objective(s), presentation of a clear report of the process of generating findings, use of sufficient data to support the interpretations, and use of appropriate analysis methods. The quality of the articles was determined based on 10 scores: 9–10 (high quality), 6–8 (moderate quality), and ≤ 5 (low quality). Articles with a score ≥ 6 were included in the study.\n[SUBTITLE] Data analysis [SUBSECTION] A content analysis approach was used to summarize the findings of this qualitative systematic reviews. In this way, to achieve a general understanding, each article was read and re-read, and then each text was broken into small units called code; then, the codes were classified into categories based on their similarities and differences. After interpreting the categories, based on the purpose of the study, the main themes were identified. The process of coding and classifying the codes were done by two coders) ZKR and MJ).\nA content analysis approach was used to summarize the findings of this qualitative systematic reviews. In this way, to achieve a general understanding, each article was read and re-read, and then each text was broken into small units called code; then, the codes were classified into categories based on their similarities and differences. After interpreting the categories, based on the purpose of the study, the main themes were identified. The process of coding and classifying the codes were done by two coders) ZKR and MJ).", "For this systematic review, Web of Sciences, PubMed, Scopus, Embase, ProQuest (Dissertations database), and Iranian databases Magiran (the largest Iranian database in various scientific and specialized fields) and SID (open access database to Iranian Persian and English studies) were searched without time limitation, until the seventh of February 2021. In addition, to complete the search and ensure access to all related articles, the Google Scholar search engine was also searched. On July 21, 2022, the mentioned databases were researched to Identify new publications between February 2021and July 2022. During the new search five studies were added.\nSearch terms were used for the titles or abstracts of the records included “moral hazard”, “unnecessary use”, “unnecessary utilization”, “non-essential use”, “non-essential utilization”, “overutilization”, “health”, “health system”, “health insurance”, “health care”, “health service”, “health services”, “healthcare”, “medical care”, and “medical service”. In Web of Sciences, Scopus, and ProQuest due to their defined search strategy, in addition to the titles and abstracts, keywords were also searched. Search in any of the databases was performed using the defined search strategy of each database. The complete search strategy is shown in Additional file 1.", "Papers in English and Persian languages in the field of reducing and controlling consumer moral hazard in the health system, conducted in a quantitative, qualitative, and mixed methods design with theoretical and empirical approaches, that were of moderate and high quality based on Dixon Wood et al. ‘s checklist [17] were included. Abstracts, letters to the editors, conference, and seminar presentations were excluded.", "All found articles were imported into Endnote software (version X9 (and duplicate articles were removed. Two researchers who were experts in the research topic and systematic review process independently screened the titles and abstracts of the articles (ZKR and MJ). In the final screening step, the full texts of the remaining articles were independently assessed by two researchers. Disagreements between the two researchers were resolved based on the opinion of a third researcher. Finally, the references of the retrieved articles were reviewed to find related articles that were not found in the first search. The screening process of retrieved papers is presented in Figure 1. Data extraction was conducted based on author’s name, title, year of publication, country, study design, strategies used to control consumer moral hazards, outcome variables, main results and quality assessment status. The main characteristics of the included study is shown in Additional file 2.\n\nFig. 1Literature selection and retrieval flow diagram.\n\nLiterature selection and retrieval flow diagram.", "In the first stage, the quality of the articles was assessed by two members of the research team who were familiar with the issue of moral hazard (ZKR and MJ), and the consensus was reached regarding the quality of the selected articles. The most important criteria for selecting high-quality articles were their relevance and role in the development of the study concept. Disagreements were resolved by a third author. The next step in the quality appraisal was based on Dixon-Woods et al. ‘s checklist [17], which included five questions regarding the clarity of the study objective(s), suitability of the study design to the objective(s), presentation of a clear report of the process of generating findings, use of sufficient data to support the interpretations, and use of appropriate analysis methods. The quality of the articles was determined based on 10 scores: 9–10 (high quality), 6–8 (moderate quality), and ≤ 5 (low quality). Articles with a score ≥ 6 were included in the study.", "A content analysis approach was used to summarize the findings of this qualitative systematic reviews. In this way, to achieve a general understanding, each article was read and re-read, and then each text was broken into small units called code; then, the codes were classified into categories based on their similarities and differences. After interpreting the categories, based on the purpose of the study, the main themes were identified. The process of coding and classifying the codes were done by two coders) ZKR and MJ).", "The themes (strategies) in this group, which are used when consumers refer to the health system and receive health services by increasing awareness of services costs and other indirect financial consequences (waiting), attempt to increase their responsibility to reduce the consumption of unnecessary health services. The themes of this group consider the changing consumptive behavior of health service consumers. This group includes four themes: demand-side cost-sharing or consumer cost-sharing, health savings accounts, drug price regulation, and rationing for health services.\n[SUBTITLE] Demand side cost sharing [SUBSECTION] Demand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29].\nThe results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71].\nIn our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance.\nDemand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29].\nThe results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71].\nIn our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance.\n[SUBTITLE] Uniform cost-sharing [SUBSECTION] The uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers.\nThe uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers.\n[SUBTITLE] Differential cost-sharing [SUBSECTION] In differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies,\nStrategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount.\nThe studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54].\nThe reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68].\nDespite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50].\nIn differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies,\nStrategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount.\nThe studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54].\nThe reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68].\nDespite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50].", "Demand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29].\nThe results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71].\nIn our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance.", "The uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers.", "In differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies,\nStrategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount.\nThe studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54].\nThe reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68].\nDespite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50].", "HSAs are considered as alternative tools for financing and dealing with future demographic challenges [72]. This financing theme under the title of health savings accounts [73, 74] or medical savings accounts [72, 75, 76], are currently used to decrease moral hazard and cost in four countries around the world [72, 75]. In this mechanism, owners of savings accounts should save a certain percentage of their income in these accounts for future health expenses. Funds of these accounts are used to pay for health expenses. Saving accounts increase people’s motivation to take responsibility by providing tax benefits and informed participation in health care decisions based on cost awareness and monitoring of physicians’ decisions [73].\nIn the United States and South Africa, they are used in combination with private insurance and is voluntarily. In Singapore and China, health saving accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system, respectively [72, 75]. This strategy in private health insurance and combination with high deductible health plans (HDHPs) is known as consumer directed health plans or consumer-driven health plans (CDHPs) [59].\nIn our study, health savings accounts were divided into two categories: voluntary health savings accounts (HSAs) and compulsory health savings accounts (HSAs), which were classified according to the mandatory and optional nature of the plan and the type of financing system in each country.\n[SUBTITLE] Voluntary health savings accounts [SUBSECTION] In this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75].\nIn this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75].\n[SUBTITLE] Compulsory health savings accounts [SUBSECTION] Compulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75].\nFindings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76].\nSome experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74].\nCompulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75].\nFindings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76].\nSome experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74].", "In this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75].", "Compulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75].\nFindings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76].\nSome experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74].", "Drug pricing is an influential component of drug access and rational use of drugs. In addition to improving access, consumption management should be considered [77]. Drug price regulation is the third theme of this group, with three categories, uniform pricing, discriminatory pricing and two part pricing, which are based on drug pricing policies and the fix or different prices for each drug unit. As the findings of this theme were extracted from only one study, the categories related to this theme included one code that could not be combined and summarized further due to dissimilarity.\n[SUBTITLE] Uniform pricing [SUBSECTION] Uniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81].\nUniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81].\n[SUBTITLE] Discriminatory pricing [SUBSECTION] Discriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81].\nDiscriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81].\n[SUBTITLE] Two part pricing [SUBSECTION] Two-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79].\nResults related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81].\nTwo-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79].\nResults related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81].", "Uniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81].", "Discriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81].", "Two-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79].\nResults related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81].", "Rationing of health services based on the waiting list (number) and waiting time (period) is one of the demand management strategies for non-emergency and elective health services [82]. Rationing of health services is the last theme that has one category named “rationing by waiting” which refers to strategies that control the consumer moral hazard by considering the cost of lost time. This strategy is one of the ways to reduce health costs that replaces user payments in countries without this system (national health system) to control costs and reduces unnecessary demand by imposing costs through the queue [30]. Findings related to the “Rationing of health services” were extracted from three studies [10, 30, 83]. These three studies investigated this mechanism using review and model-based theoretical approaches. The results of the theoretical analysis of these strategies regarding optimality [30] and well-being [83] outcomes were not associated with positive results.These strategies are not very popular and people tend to pay instead of waiting [83].\n\nTable 2Strategies for changing behavior at the time of receiving health servicesTheme/sourceCategoryCodeOutcome variablesDemand side cost sharing[5, 6, 9, 10, 13, 15, 18–20, 22–25, 27–33, 35–49, 51–57, 59–67, 70–73, 78]Uniformcost sharing- Fixed rate of deductible: traditional deductible, mandatory deductible, First euro deductible/ a first-dollar deductible, doughnut hole deductible- Fixed rate of co-payments or coinsurance/co-insurance: /copayment/ user charges / user-fee, lump sum co-payments, fixed payment, Mandatory co-payments- Fixed-rate insurance coverage limit /caps on insurance /stop loss. / payment ceiling limit on coverage, limit on out-of-pocket expenses- Health services utilization- Health costs- Health related behavior- Choice of insurance plan- The opinion and acceptance of consumersDifferentialcost sharing- Income base cost sharing: income base deductible: income-related copayment /coinsurance/ different copayment according to the socioeconomic status- Varied deducible: Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles- Tier cost sharing: :multitier copayments / tiered copayment/ price-related co-payment tier, tier coinsurance- Value-based cost sharing: Value Based Insurance Design (VBID) / value-based cost sharing (lower coinsurance for services with higher costs benefits / value-based coinsurance target / variable co-insurance based on demand elasticity/ treatment-specific copayments disease-specific cost sharing/ differential cost sharing based on disease status- Cost sharing with discount: co-payment exemption, co-payment with rebate, / waiving copaysHealth savings accounts(HSAs)[26, 50, 54, 58, 60, 74, 75]Voluntary Health Savings Accounts (HSAs)- Health savings accounts (HSAs) / Health Reimbursement Accounts (HRAs) / medical savings accounts (MSAs) in combination with private insurance- Consumer-Directed Health Plans (CDHPS) /Consumer-Driven Health Plans (CDHPS) / Three-Tier Payment System(HDHPS Coupled With Personal Savings Account)- Health services utilization- Health costs- Saving for futureCompulsory Health Savings Accounts (HSAs)- Medisave (Medical savings accounts) (MSAs) in combination with social health insurance- Tongdao (MSA )in combination social risk-pooling (SRP)/ medical savings accounts (MSAs) in combination with social insurance pool (SIP)/ the three-tiered design (MSA-deductible-SIP)- Bankuai (MSA) separately to finance outpatient servicesDrug price regulation[21]Uniform pricingdiscriminatory pricing- Uniform pricing/uniform monopoly pricing- Third degree price discrimination / (different prices to different markets or groups) of consumers- Drug utilizationTwo PartPricing- Two-part tariffs.) combines a uniform price with market-specific lump-sum paymentsRationing for health services [10, 31, 34]Rationing by waiting- Waiting lists / queuing /expectancy queue/the cost of the lost time- Waiting time- Optimality- Well-being\n\nStrategies for changing behavior at the time of receiving health services\nDemand side cost sharing\n[5, 6, 9, 10, 13, 15, 18–20, 22–25, 27–33, 35–49, 51–57, 59–67, 70–73, 78]\nUniform\ncost sharing\n- Fixed rate of deductible: traditional deductible, mandatory deductible, First euro deductible/ a first-dollar deductible, doughnut hole deductible\n- Fixed rate of co-payments or coinsurance/co-insurance: /copayment/ user charges / user-fee, lump sum co-payments, fixed payment, Mandatory co-payments\n- Fixed-rate insurance coverage limit /caps on insurance /stop loss. / payment ceiling limit on coverage, limit on out-of-pocket expenses\n- Health services utilization\n- Health costs\n- Health related behavior\n- Choice of insurance plan\n- The opinion and acceptance of consumers\nDifferential\ncost sharing\n- Income base cost sharing: income base deductible: income-related copayment /coinsurance/ different copayment according to the socioeconomic status\n- Varied deducible: Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles\n- Tier cost sharing: :multitier copayments / tiered copayment/ price-related co-payment tier, tier coinsurance\n- Value-based cost sharing: Value Based Insurance Design (VBID) / value-based cost sharing (lower coinsurance for services with higher costs benefits / value-based coinsurance target / variable co-insurance based on demand elasticity/ treatment-specific copayments disease-specific cost sharing/ differential cost sharing based on disease status\n- Cost sharing with discount: co-payment exemption, co-payment with rebate, / waiving copays\nHealth savings accounts\n(HSAs)\n[26, 50, 54, 58, 60, 74, 75]\n- Health savings accounts (HSAs) / Health Reimbursement Accounts (HRAs) / medical savings accounts (MSAs) in combination with private insurance\n- Consumer-Directed Health Plans (CDHPS) /Consumer-Driven Health Plans (CDHPS) / Three-Tier Payment System(HDHPS Coupled With Personal Savings Account)\n- Health services utilization\n- Health costs\n- Saving for future\n- Medisave (Medical savings accounts) (MSAs) in combination with social health insurance\n- Tongdao (MSA )in combination social risk-pooling (SRP)/ medical savings accounts (MSAs) in combination with social insurance pool (SIP)/ the three-tiered design (MSA-deductible-SIP)\n- Bankuai (MSA) separately to finance outpatient services\nDrug price regulation\n[21]\nUniform pricing\ndiscriminatory pricing\n- Uniform pricing/uniform monopoly pricing\n- Third degree price discrimination / (different prices to different markets or groups) of consumers\n- Waiting lists / queuing /expectancy queue/the cost of the lost time\n- Waiting time\n- Optimality\n- Well-being", "The themes in this group deal with the strategies that are applied outside the health system, before the need for healthcare services, and through the consciousness of health and positive financial incentives, increase healthy behavior or prevent unhealthy behavior. Changing individual behaviors to reduce high-risk behaviors and improve health-promoting behaviors is the approach of this group. This group includes two themes: development of incentive insurance programs and community empowerment.", "The themes of this group focus on the measures of insurance companies and purchasers. This theme refers to strategies aimed at reducing the risk of disease and the need for health services or unhealthy behaviors and consists of two categories: extending preventive care insurance and developing bonus-oriented insurance. Findings related to this theme were extracted from11 studies [10, 15, 25, 37, 43, 46, 58, 84–87].\n[SUBTITLE] Extending preventive care insurance [SUBSECTION] Expanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84].\nExpanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84].\n[SUBTITLE] Developing bonus-oriented insurance [SUBSECTION] These strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58].\nHealth service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85].\nThese strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58].\nHealth service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85].", "Expanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84].", "These strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58].\nHealth service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85].", "Community empowerment is the second and last theme in this group with one category called Community education.The theme of ”community empowerment”, mentioned by only one study [33].\n[SUBTITLE] Community education [SUBSECTION] This category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme.\nThe sources from which each code is extracted are provided in Additional file 3.\nThis category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme.\nThe sources from which each code is extracted are provided in Additional file 3.", "This category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme.\nThe sources from which each code is extracted are provided in Additional file 3.", "The risk of bias assessment in this study consisted of the following: To reduce publication bias, unpublished papers were searched in the Dissertations database of ProQuest for grey literature, but no related papers were found. In this regard, there is a possibility of language bias due to the limitation of non-English articles in publishing or indexing the results and the focus of this study on Persian and English articles, which is mentioned as a limitation in the limitations section.\n\nTable 3Strategies for changing behavior before needing health servicesTheme/sourceCategoryCodeOutcome variablesDevelopment of incentive insurance programs[10, 15, 27, 30, 39, 42, 59, 69, 76, 77, 79]Extending preventive care insurance- Proposing insurance coverage for preventive care- Separating insurance coverage for prevention and treatment- To encourage insureds to use more secondary preventive care- Improving perception of health status through secondary preventive care- Health services utilization- Health costs- Risk-reducing behaviors- Choice of expensive health services healthDevelopment of bonus oriented insurance- Adjusted premiums according level of preventive effort- Premium reduction, bonus payments/ rebate insurance /to encourage non-use or limited use in return for next premium reduction, risk adjustment .no-claims bonus, risk rating premium- No-claim Bonus and Coverage Upper BoundCommunity empowerment [23]Community education- Health promotion education- Civic education about the consequences of unnecessary use of health services- Not evaluated\n\nStrategies for changing behavior before needing health services\nDevelopment of incentive insurance programs\n[10, 15, 27, 30, 39, 42, 59, 69, 76, 77, 79]\n- Proposing insurance coverage for preventive care\n- Separating insurance coverage for prevention and treatment\n- To encourage insureds to use more secondary preventive care\n- Improving perception of health status through secondary preventive care\n- Health services utilization\n- Health costs\n- Risk-reducing behaviors\n- Choice of expensive health services health\n- Adjusted premiums according level of preventive effort\n- Premium reduction, bonus payments/ rebate insurance /to encourage non-use or limited use in return for next premium reduction, risk adjustment .no-claims bonus, risk rating premium\n- No-claim Bonus and Coverage Upper Bound\n- Health promotion education\n- Civic education about the consequences of unnecessary use of health services", "This study has several limitations. The first, is the restriction of studies in Persian and English languages. No clear boundary between the consumer and provider moral hazard in some articles is another limitation. The researchers separated these two issues by studying the full text of the articles, focusing on the type and setting of service delivery and the role of physicians in providing services. The last limitation was the methodological diversity and heterogeneity of the quantitative studies, which did not allow for quantitative analysis and reporting the effectiveness of the strategies.", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2\n\nSupplementary Material 3\n\nSupplementary Material 3" ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Databases and search strategies", "Inclusion and exclusion criteria", "Methods of screening and selection criteria", "Quality appraisal", "Data analysis", "Results", "Strategies for changing behavior at the time of receiving health services", "Demand side cost sharing", "Uniform cost-sharing", "Differential cost-sharing", "Health savings accounts (HSAs)", "Voluntary health savings accounts", "Compulsory health savings accounts", "Drug price regulation", "Uniform pricing", "Discriminatory pricing", "Two part pricing", "Rationing of health services", "Strategies for changing behavior before needing health services", "Development of incentive insurance programs", "Extending preventive care insurance", "Developing bonus-oriented insurance", "Community empowerment", "Community education", "Risk of bias consideration", "Discussion", "Limitations", "Conclusion", "Electronic supplementary material", "" ]
[ "Maximizing the health of people and populations is one of the main goals of any health system which leads to improving personal, social, and economic well-being [1]. The efficient function of the health system depends on providing improved health services at a minimum cost [2]. Evidence shows that health spending is growing faster than economic growth [1, 3]. On average, health spending in OECD countries was equivalent to 9% of GDP in 2018 [1]. The United States spent 18% of its GDP on health care in 2015 [4]. Moral hazard is one of the most important reasons for increasing health costs [5, 6]. Moral hazard is the change in health behavior and consumption of health services because of insurance coverage [7]. According to the theory of moral hazard, health insurance and third-party payers, by lowering the price of care, encourage the consumer to consume more care than when they consume at the market price [8]. Insurance coverage leads to the consumption of health services above an efficient level [9].\nThis phenomenon is classified in different ways: ex-ante moral hazard and ex-post moral hazard, hidden information and hidden action moral hazard, provider moral hazard, and consumer moral hazard [10, 11]. Ex-ante moral hazard occurs before illness and increases a person’s unhealthy behavior; in contrast, ex-post moral hazard occurs after the onset of illness and will increase costs by increasing demand and consuming unnecessary services. In the hidden information, the insurer cannot observe the real condition and severity of the disease to pay the cost according to the real need. In hidden action individuals’ precautionary measures are not visible, and the insured person does not take the necessary precautions to prevent the disease. Provider moral hazard occurs when a provider provides more services to increase its revenue. The provider moral hazard is also known as the provider’s induced demand. Consumer moral hazard means insured people demand more care than uninsured people [11, 12]. Additionally, consumer moral hazard deals with the reduction in preventive healthcare behaviors resulting from insurance coverage[13].\nMoral hazard is known as one of the main causes of market failure [14] that has many adverse consequences, such as the impact on demand elasticity, reduction of welfare, inefficiency in using resources, reduction in technical and allocation efficiency, reduction of benefits of risk pooling, and price increase [11, 15]. Moral hazard, as a concern in the health insurance market, requires the application of appropriate policies and interventions to be controlled. In this regard, consumer moral hazard as the most obvious form of moral hazard [16] has been one of the topics of concern for policymakers and economic experts in recent years [8]. The aim of this study was to determine and analyze strategies used to control consumer moral hazards in health systems. The results of this study can be used for health insurance planning, health system financing, and health cost reduction.", "This study was written as part of a Ph.D thesis entitled “Developing a model to control consumer moral hazard in Iran’s health system” which was designed and performed based on its proposal and was approved by the local ethics committee of the Iran University of Medical Sciences (code: IR.IUMS.REC.1399.1103).\nResearch questions.\n\nWhat strategies or interventions are used to control consumers’ moral hazards?What is the approach of identifying strategies to control moral hazards?\n\nWhat strategies or interventions are used to control consumers’ moral hazards?\nWhat is the approach of identifying strategies to control moral hazards?\n[SUBTITLE] Databases and search strategies [SUBSECTION] For this systematic review, Web of Sciences, PubMed, Scopus, Embase, ProQuest (Dissertations database), and Iranian databases Magiran (the largest Iranian database in various scientific and specialized fields) and SID (open access database to Iranian Persian and English studies) were searched without time limitation, until the seventh of February 2021. In addition, to complete the search and ensure access to all related articles, the Google Scholar search engine was also searched. On July 21, 2022, the mentioned databases were researched to Identify new publications between February 2021and July 2022. During the new search five studies were added.\nSearch terms were used for the titles or abstracts of the records included “moral hazard”, “unnecessary use”, “unnecessary utilization”, “non-essential use”, “non-essential utilization”, “overutilization”, “health”, “health system”, “health insurance”, “health care”, “health service”, “health services”, “healthcare”, “medical care”, and “medical service”. In Web of Sciences, Scopus, and ProQuest due to their defined search strategy, in addition to the titles and abstracts, keywords were also searched. Search in any of the databases was performed using the defined search strategy of each database. The complete search strategy is shown in Additional file 1.\nFor this systematic review, Web of Sciences, PubMed, Scopus, Embase, ProQuest (Dissertations database), and Iranian databases Magiran (the largest Iranian database in various scientific and specialized fields) and SID (open access database to Iranian Persian and English studies) were searched without time limitation, until the seventh of February 2021. In addition, to complete the search and ensure access to all related articles, the Google Scholar search engine was also searched. On July 21, 2022, the mentioned databases were researched to Identify new publications between February 2021and July 2022. During the new search five studies were added.\nSearch terms were used for the titles or abstracts of the records included “moral hazard”, “unnecessary use”, “unnecessary utilization”, “non-essential use”, “non-essential utilization”, “overutilization”, “health”, “health system”, “health insurance”, “health care”, “health service”, “health services”, “healthcare”, “medical care”, and “medical service”. In Web of Sciences, Scopus, and ProQuest due to their defined search strategy, in addition to the titles and abstracts, keywords were also searched. Search in any of the databases was performed using the defined search strategy of each database. The complete search strategy is shown in Additional file 1.\n[SUBTITLE] Inclusion and exclusion criteria [SUBSECTION] Papers in English and Persian languages in the field of reducing and controlling consumer moral hazard in the health system, conducted in a quantitative, qualitative, and mixed methods design with theoretical and empirical approaches, that were of moderate and high quality based on Dixon Wood et al. ‘s checklist [17] were included. Abstracts, letters to the editors, conference, and seminar presentations were excluded.\nPapers in English and Persian languages in the field of reducing and controlling consumer moral hazard in the health system, conducted in a quantitative, qualitative, and mixed methods design with theoretical and empirical approaches, that were of moderate and high quality based on Dixon Wood et al. ‘s checklist [17] were included. Abstracts, letters to the editors, conference, and seminar presentations were excluded.\n[SUBTITLE] Methods of screening and selection criteria [SUBSECTION] All found articles were imported into Endnote software (version X9 (and duplicate articles were removed. Two researchers who were experts in the research topic and systematic review process independently screened the titles and abstracts of the articles (ZKR and MJ). In the final screening step, the full texts of the remaining articles were independently assessed by two researchers. Disagreements between the two researchers were resolved based on the opinion of a third researcher. Finally, the references of the retrieved articles were reviewed to find related articles that were not found in the first search. The screening process of retrieved papers is presented in Figure 1. Data extraction was conducted based on author’s name, title, year of publication, country, study design, strategies used to control consumer moral hazards, outcome variables, main results and quality assessment status. The main characteristics of the included study is shown in Additional file 2.\n\nFig. 1Literature selection and retrieval flow diagram.\n\nLiterature selection and retrieval flow diagram.\nAll found articles were imported into Endnote software (version X9 (and duplicate articles were removed. Two researchers who were experts in the research topic and systematic review process independently screened the titles and abstracts of the articles (ZKR and MJ). In the final screening step, the full texts of the remaining articles were independently assessed by two researchers. Disagreements between the two researchers were resolved based on the opinion of a third researcher. Finally, the references of the retrieved articles were reviewed to find related articles that were not found in the first search. The screening process of retrieved papers is presented in Figure 1. Data extraction was conducted based on author’s name, title, year of publication, country, study design, strategies used to control consumer moral hazards, outcome variables, main results and quality assessment status. The main characteristics of the included study is shown in Additional file 2.\n\nFig. 1Literature selection and retrieval flow diagram.\n\nLiterature selection and retrieval flow diagram.\n[SUBTITLE] Quality appraisal [SUBSECTION] In the first stage, the quality of the articles was assessed by two members of the research team who were familiar with the issue of moral hazard (ZKR and MJ), and the consensus was reached regarding the quality of the selected articles. The most important criteria for selecting high-quality articles were their relevance and role in the development of the study concept. Disagreements were resolved by a third author. The next step in the quality appraisal was based on Dixon-Woods et al. ‘s checklist [17], which included five questions regarding the clarity of the study objective(s), suitability of the study design to the objective(s), presentation of a clear report of the process of generating findings, use of sufficient data to support the interpretations, and use of appropriate analysis methods. The quality of the articles was determined based on 10 scores: 9–10 (high quality), 6–8 (moderate quality), and ≤ 5 (low quality). Articles with a score ≥ 6 were included in the study.\nIn the first stage, the quality of the articles was assessed by two members of the research team who were familiar with the issue of moral hazard (ZKR and MJ), and the consensus was reached regarding the quality of the selected articles. The most important criteria for selecting high-quality articles were their relevance and role in the development of the study concept. Disagreements were resolved by a third author. The next step in the quality appraisal was based on Dixon-Woods et al. ‘s checklist [17], which included five questions regarding the clarity of the study objective(s), suitability of the study design to the objective(s), presentation of a clear report of the process of generating findings, use of sufficient data to support the interpretations, and use of appropriate analysis methods. The quality of the articles was determined based on 10 scores: 9–10 (high quality), 6–8 (moderate quality), and ≤ 5 (low quality). Articles with a score ≥ 6 were included in the study.\n[SUBTITLE] Data analysis [SUBSECTION] A content analysis approach was used to summarize the findings of this qualitative systematic reviews. In this way, to achieve a general understanding, each article was read and re-read, and then each text was broken into small units called code; then, the codes were classified into categories based on their similarities and differences. After interpreting the categories, based on the purpose of the study, the main themes were identified. The process of coding and classifying the codes were done by two coders) ZKR and MJ).\nA content analysis approach was used to summarize the findings of this qualitative systematic reviews. In this way, to achieve a general understanding, each article was read and re-read, and then each text was broken into small units called code; then, the codes were classified into categories based on their similarities and differences. After interpreting the categories, based on the purpose of the study, the main themes were identified. The process of coding and classifying the codes were done by two coders) ZKR and MJ).", "For this systematic review, Web of Sciences, PubMed, Scopus, Embase, ProQuest (Dissertations database), and Iranian databases Magiran (the largest Iranian database in various scientific and specialized fields) and SID (open access database to Iranian Persian and English studies) were searched without time limitation, until the seventh of February 2021. In addition, to complete the search and ensure access to all related articles, the Google Scholar search engine was also searched. On July 21, 2022, the mentioned databases were researched to Identify new publications between February 2021and July 2022. During the new search five studies were added.\nSearch terms were used for the titles or abstracts of the records included “moral hazard”, “unnecessary use”, “unnecessary utilization”, “non-essential use”, “non-essential utilization”, “overutilization”, “health”, “health system”, “health insurance”, “health care”, “health service”, “health services”, “healthcare”, “medical care”, and “medical service”. In Web of Sciences, Scopus, and ProQuest due to their defined search strategy, in addition to the titles and abstracts, keywords were also searched. Search in any of the databases was performed using the defined search strategy of each database. The complete search strategy is shown in Additional file 1.", "Papers in English and Persian languages in the field of reducing and controlling consumer moral hazard in the health system, conducted in a quantitative, qualitative, and mixed methods design with theoretical and empirical approaches, that were of moderate and high quality based on Dixon Wood et al. ‘s checklist [17] were included. Abstracts, letters to the editors, conference, and seminar presentations were excluded.", "All found articles were imported into Endnote software (version X9 (and duplicate articles were removed. Two researchers who were experts in the research topic and systematic review process independently screened the titles and abstracts of the articles (ZKR and MJ). In the final screening step, the full texts of the remaining articles were independently assessed by two researchers. Disagreements between the two researchers were resolved based on the opinion of a third researcher. Finally, the references of the retrieved articles were reviewed to find related articles that were not found in the first search. The screening process of retrieved papers is presented in Figure 1. Data extraction was conducted based on author’s name, title, year of publication, country, study design, strategies used to control consumer moral hazards, outcome variables, main results and quality assessment status. The main characteristics of the included study is shown in Additional file 2.\n\nFig. 1Literature selection and retrieval flow diagram.\n\nLiterature selection and retrieval flow diagram.", "In the first stage, the quality of the articles was assessed by two members of the research team who were familiar with the issue of moral hazard (ZKR and MJ), and the consensus was reached regarding the quality of the selected articles. The most important criteria for selecting high-quality articles were their relevance and role in the development of the study concept. Disagreements were resolved by a third author. The next step in the quality appraisal was based on Dixon-Woods et al. ‘s checklist [17], which included five questions regarding the clarity of the study objective(s), suitability of the study design to the objective(s), presentation of a clear report of the process of generating findings, use of sufficient data to support the interpretations, and use of appropriate analysis methods. The quality of the articles was determined based on 10 scores: 9–10 (high quality), 6–8 (moderate quality), and ≤ 5 (low quality). Articles with a score ≥ 6 were included in the study.", "A content analysis approach was used to summarize the findings of this qualitative systematic reviews. In this way, to achieve a general understanding, each article was read and re-read, and then each text was broken into small units called code; then, the codes were classified into categories based on their similarities and differences. After interpreting the categories, based on the purpose of the study, the main themes were identified. The process of coding and classifying the codes were done by two coders) ZKR and MJ).", "In the search of databases and other sources, 7488 articles were retrieved, and after removing duplicate sources and applying inclusion and exclusion criteria, 68 eligible articles were selected (Fig. 1). General description of the selected studies is shown in Table 1. As Table 1 shows, out of 68 included papers, the majority of studies were conducted in a period from 2016 to 2021(n = 29), in a quantitative approach (n = 46). And, health service demand and utilization was the most common outcome investigated in these studies. (n = 38).\n\nTable 1General description of selected studiesClassification categorySub categoryNumresourcesYear of publication1990–20001[18]2001–200510[19–28]2006–201011[6, 29–38]2011–201517[5, 9, 39–53]2016–202129[10, 13, 15, 54–79]Study designQuantitative46[5,6,9, 13, 20, 22–25, 28, 32, 33, 35–38, 40–46, 48, 51–55, 57, 58, 61–67, 69, 71–76, 78]Qualitative5[26, 27, 39, 49, 70]Review5[10, 15, 47, 56, 60]Theatrical approach(model – based)12[18, 19, 21, 29–31, 34, 50, 59, 68, 77, 79]Outcomes variableDemand & utilization38[5, 15, 20, 22, 23, 28, 30–33, 35, 36, 39–41, 43–48, 52, 54–56, 59, 61, 63–66, 69, 71–74, 77, 78]Cost & optimality26[5, 15, 18, 19, 29–31, 34, 35, 37–40, 50, 51, 53, 57, 58, 60, 68, 71, 73–76, 79]Health related behavior2[13, 46, 50, 77]Opinion & acceptance4[24, 27, 49, 70]Choice of plan2[42, 69]\n\nGeneral description of selected studies\nThe content analysis of 68 studies included in the study are presented in the form of two group, six themes, and 11 categories.\nSince, based on the moral hazard theory, this phenomenon is defined as a change in consumer behavior because of insurance coverage, focusing on changing behavior and modifying it is the main goal of the controlling strategies. Therefore, the results of this study were summarized in the two groups: “Changing behavior at the time of receiving health services” (Table 2) and “Changing behavior before needing health services” (Table 3). The first group includes four themes: demand-side cost-sharing or consumer cost-sharing, health savings accounts, drug price regulation, and rationing for health services. The second group includes two themes: development of incentive insurance programs and community empowerment.\nThe relationship between these two groups is drawn in the form of a diagram (Fig. 2.)\nIn reviewing the findings of the review studies, the results of four primary studies [18–21] were repeated in one review study [22] and the results of one primary study [23] were repeated in another review study [24]. Since in the qualitative analysis, the criteria for analyzing the findings are different from the quantitative results, repeated findings were not excluded in the qualitative analysis, but in the narrative report of the findings of quantitative studies, only the findings of the primary studies were presented.\n\nFig. 2Relationship of strategies of “Changing behavior before needing health services” with strategies of “Changing behavior at the time of receiving health services”\n\nRelationship of strategies of “Changing behavior before needing health services” with strategies of “Changing behavior at the time of receiving health services”\n[SUBTITLE] Strategies for changing behavior at the time of receiving health services [SUBSECTION] The themes (strategies) in this group, which are used when consumers refer to the health system and receive health services by increasing awareness of services costs and other indirect financial consequences (waiting), attempt to increase their responsibility to reduce the consumption of unnecessary health services. The themes of this group consider the changing consumptive behavior of health service consumers. This group includes four themes: demand-side cost-sharing or consumer cost-sharing, health savings accounts, drug price regulation, and rationing for health services.\n[SUBTITLE] Demand side cost sharing [SUBSECTION] Demand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29].\nThe results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71].\nIn our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance.\nDemand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29].\nThe results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71].\nIn our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance.\n[SUBTITLE] Uniform cost-sharing [SUBSECTION] The uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers.\nThe uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers.\n[SUBTITLE] Differential cost-sharing [SUBSECTION] In differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies,\nStrategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount.\nThe studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54].\nThe reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68].\nDespite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50].\nIn differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies,\nStrategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount.\nThe studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54].\nThe reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68].\nDespite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50].\nThe themes (strategies) in this group, which are used when consumers refer to the health system and receive health services by increasing awareness of services costs and other indirect financial consequences (waiting), attempt to increase their responsibility to reduce the consumption of unnecessary health services. The themes of this group consider the changing consumptive behavior of health service consumers. This group includes four themes: demand-side cost-sharing or consumer cost-sharing, health savings accounts, drug price regulation, and rationing for health services.\n[SUBTITLE] Demand side cost sharing [SUBSECTION] Demand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29].\nThe results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71].\nIn our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance.\nDemand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29].\nThe results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71].\nIn our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance.\n[SUBTITLE] Uniform cost-sharing [SUBSECTION] The uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers.\nThe uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers.\n[SUBTITLE] Differential cost-sharing [SUBSECTION] In differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies,\nStrategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount.\nThe studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54].\nThe reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68].\nDespite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50].\nIn differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies,\nStrategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount.\nThe studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54].\nThe reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68].\nDespite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50].\n[SUBTITLE] Health savings accounts (HSAs) [SUBSECTION] HSAs are considered as alternative tools for financing and dealing with future demographic challenges [72]. This financing theme under the title of health savings accounts [73, 74] or medical savings accounts [72, 75, 76], are currently used to decrease moral hazard and cost in four countries around the world [72, 75]. In this mechanism, owners of savings accounts should save a certain percentage of their income in these accounts for future health expenses. Funds of these accounts are used to pay for health expenses. Saving accounts increase people’s motivation to take responsibility by providing tax benefits and informed participation in health care decisions based on cost awareness and monitoring of physicians’ decisions [73].\nIn the United States and South Africa, they are used in combination with private insurance and is voluntarily. In Singapore and China, health saving accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system, respectively [72, 75]. This strategy in private health insurance and combination with high deductible health plans (HDHPs) is known as consumer directed health plans or consumer-driven health plans (CDHPs) [59].\nIn our study, health savings accounts were divided into two categories: voluntary health savings accounts (HSAs) and compulsory health savings accounts (HSAs), which were classified according to the mandatory and optional nature of the plan and the type of financing system in each country.\n[SUBTITLE] Voluntary health savings accounts [SUBSECTION] In this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75].\nIn this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75].\n[SUBTITLE] Compulsory health savings accounts [SUBSECTION] Compulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75].\nFindings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76].\nSome experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74].\nCompulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75].\nFindings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76].\nSome experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74].\nHSAs are considered as alternative tools for financing and dealing with future demographic challenges [72]. This financing theme under the title of health savings accounts [73, 74] or medical savings accounts [72, 75, 76], are currently used to decrease moral hazard and cost in four countries around the world [72, 75]. In this mechanism, owners of savings accounts should save a certain percentage of their income in these accounts for future health expenses. Funds of these accounts are used to pay for health expenses. Saving accounts increase people’s motivation to take responsibility by providing tax benefits and informed participation in health care decisions based on cost awareness and monitoring of physicians’ decisions [73].\nIn the United States and South Africa, they are used in combination with private insurance and is voluntarily. In Singapore and China, health saving accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system, respectively [72, 75]. This strategy in private health insurance and combination with high deductible health plans (HDHPs) is known as consumer directed health plans or consumer-driven health plans (CDHPs) [59].\nIn our study, health savings accounts were divided into two categories: voluntary health savings accounts (HSAs) and compulsory health savings accounts (HSAs), which were classified according to the mandatory and optional nature of the plan and the type of financing system in each country.\n[SUBTITLE] Voluntary health savings accounts [SUBSECTION] In this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75].\nIn this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75].\n[SUBTITLE] Compulsory health savings accounts [SUBSECTION] Compulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75].\nFindings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76].\nSome experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74].\nCompulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75].\nFindings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76].\nSome experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74].\n[SUBTITLE] Drug price regulation [SUBSECTION] Drug pricing is an influential component of drug access and rational use of drugs. In addition to improving access, consumption management should be considered [77]. Drug price regulation is the third theme of this group, with three categories, uniform pricing, discriminatory pricing and two part pricing, which are based on drug pricing policies and the fix or different prices for each drug unit. As the findings of this theme were extracted from only one study, the categories related to this theme included one code that could not be combined and summarized further due to dissimilarity.\n[SUBTITLE] Uniform pricing [SUBSECTION] Uniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81].\nUniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81].\n[SUBTITLE] Discriminatory pricing [SUBSECTION] Discriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81].\nDiscriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81].\n[SUBTITLE] Two part pricing [SUBSECTION] Two-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79].\nResults related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81].\nTwo-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79].\nResults related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81].\nDrug pricing is an influential component of drug access and rational use of drugs. In addition to improving access, consumption management should be considered [77]. Drug price regulation is the third theme of this group, with three categories, uniform pricing, discriminatory pricing and two part pricing, which are based on drug pricing policies and the fix or different prices for each drug unit. As the findings of this theme were extracted from only one study, the categories related to this theme included one code that could not be combined and summarized further due to dissimilarity.\n[SUBTITLE] Uniform pricing [SUBSECTION] Uniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81].\nUniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81].\n[SUBTITLE] Discriminatory pricing [SUBSECTION] Discriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81].\nDiscriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81].\n[SUBTITLE] Two part pricing [SUBSECTION] Two-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79].\nResults related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81].\nTwo-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79].\nResults related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81].\n[SUBTITLE] Rationing of health services [SUBSECTION] Rationing of health services based on the waiting list (number) and waiting time (period) is one of the demand management strategies for non-emergency and elective health services [82]. Rationing of health services is the last theme that has one category named “rationing by waiting” which refers to strategies that control the consumer moral hazard by considering the cost of lost time. This strategy is one of the ways to reduce health costs that replaces user payments in countries without this system (national health system) to control costs and reduces unnecessary demand by imposing costs through the queue [30]. Findings related to the “Rationing of health services” were extracted from three studies [10, 30, 83]. These three studies investigated this mechanism using review and model-based theoretical approaches. The results of the theoretical analysis of these strategies regarding optimality [30] and well-being [83] outcomes were not associated with positive results.These strategies are not very popular and people tend to pay instead of waiting [83].\n\nTable 2Strategies for changing behavior at the time of receiving health servicesTheme/sourceCategoryCodeOutcome variablesDemand side cost sharing[5, 6, 9, 10, 13, 15, 18–20, 22–25, 27–33, 35–49, 51–57, 59–67, 70–73, 78]Uniformcost sharing- Fixed rate of deductible: traditional deductible, mandatory deductible, First euro deductible/ a first-dollar deductible, doughnut hole deductible- Fixed rate of co-payments or coinsurance/co-insurance: /copayment/ user charges / user-fee, lump sum co-payments, fixed payment, Mandatory co-payments- Fixed-rate insurance coverage limit /caps on insurance /stop loss. / payment ceiling limit on coverage, limit on out-of-pocket expenses- Health services utilization- Health costs- Health related behavior- Choice of insurance plan- The opinion and acceptance of consumersDifferentialcost sharing- Income base cost sharing: income base deductible: income-related copayment /coinsurance/ different copayment according to the socioeconomic status- Varied deducible: Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles- Tier cost sharing: :multitier copayments / tiered copayment/ price-related co-payment tier, tier coinsurance- Value-based cost sharing: Value Based Insurance Design (VBID) / value-based cost sharing (lower coinsurance for services with higher costs benefits / value-based coinsurance target / variable co-insurance based on demand elasticity/ treatment-specific copayments disease-specific cost sharing/ differential cost sharing based on disease status- Cost sharing with discount: co-payment exemption, co-payment with rebate, / waiving copaysHealth savings accounts(HSAs)[26, 50, 54, 58, 60, 74, 75]Voluntary Health Savings Accounts (HSAs)- Health savings accounts (HSAs) / Health Reimbursement Accounts (HRAs) / medical savings accounts (MSAs) in combination with private insurance- Consumer-Directed Health Plans (CDHPS) /Consumer-Driven Health Plans (CDHPS) / Three-Tier Payment System(HDHPS Coupled With Personal Savings Account)- Health services utilization- Health costs- Saving for futureCompulsory Health Savings Accounts (HSAs)- Medisave (Medical savings accounts) (MSAs) in combination with social health insurance- Tongdao (MSA )in combination social risk-pooling (SRP)/ medical savings accounts (MSAs) in combination with social insurance pool (SIP)/ the three-tiered design (MSA-deductible-SIP)- Bankuai (MSA) separately to finance outpatient servicesDrug price regulation[21]Uniform pricingdiscriminatory pricing- Uniform pricing/uniform monopoly pricing- Third degree price discrimination / (different prices to different markets or groups) of consumers- Drug utilizationTwo PartPricing- Two-part tariffs.) combines a uniform price with market-specific lump-sum paymentsRationing for health services [10, 31, 34]Rationing by waiting- Waiting lists / queuing /expectancy queue/the cost of the lost time- Waiting time- Optimality- Well-being\n\nStrategies for changing behavior at the time of receiving health services\nDemand side cost sharing\n[5, 6, 9, 10, 13, 15, 18–20, 22–25, 27–33, 35–49, 51–57, 59–67, 70–73, 78]\nUniform\ncost sharing\n- Fixed rate of deductible: traditional deductible, mandatory deductible, First euro deductible/ a first-dollar deductible, doughnut hole deductible\n- Fixed rate of co-payments or coinsurance/co-insurance: /copayment/ user charges / user-fee, lump sum co-payments, fixed payment, Mandatory co-payments\n- Fixed-rate insurance coverage limit /caps on insurance /stop loss. / payment ceiling limit on coverage, limit on out-of-pocket expenses\n- Health services utilization\n- Health costs\n- Health related behavior\n- Choice of insurance plan\n- The opinion and acceptance of consumers\nDifferential\ncost sharing\n- Income base cost sharing: income base deductible: income-related copayment /coinsurance/ different copayment according to the socioeconomic status\n- Varied deducible: Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles\n- Tier cost sharing: :multitier copayments / tiered copayment/ price-related co-payment tier, tier coinsurance\n- Value-based cost sharing: Value Based Insurance Design (VBID) / value-based cost sharing (lower coinsurance for services with higher costs benefits / value-based coinsurance target / variable co-insurance based on demand elasticity/ treatment-specific copayments disease-specific cost sharing/ differential cost sharing based on disease status\n- Cost sharing with discount: co-payment exemption, co-payment with rebate, / waiving copays\nHealth savings accounts\n(HSAs)\n[26, 50, 54, 58, 60, 74, 75]\n- Health savings accounts (HSAs) / Health Reimbursement Accounts (HRAs) / medical savings accounts (MSAs) in combination with private insurance\n- Consumer-Directed Health Plans (CDHPS) /Consumer-Driven Health Plans (CDHPS) / Three-Tier Payment System(HDHPS Coupled With Personal Savings Account)\n- Health services utilization\n- Health costs\n- Saving for future\n- Medisave (Medical savings accounts) (MSAs) in combination with social health insurance\n- Tongdao (MSA )in combination social risk-pooling (SRP)/ medical savings accounts (MSAs) in combination with social insurance pool (SIP)/ the three-tiered design (MSA-deductible-SIP)\n- Bankuai (MSA) separately to finance outpatient services\nDrug price regulation\n[21]\nUniform pricing\ndiscriminatory pricing\n- Uniform pricing/uniform monopoly pricing\n- Third degree price discrimination / (different prices to different markets or groups) of consumers\n- Waiting lists / queuing /expectancy queue/the cost of the lost time\n- Waiting time\n- Optimality\n- Well-being\nRationing of health services based on the waiting list (number) and waiting time (period) is one of the demand management strategies for non-emergency and elective health services [82]. Rationing of health services is the last theme that has one category named “rationing by waiting” which refers to strategies that control the consumer moral hazard by considering the cost of lost time. This strategy is one of the ways to reduce health costs that replaces user payments in countries without this system (national health system) to control costs and reduces unnecessary demand by imposing costs through the queue [30]. Findings related to the “Rationing of health services” were extracted from three studies [10, 30, 83]. These three studies investigated this mechanism using review and model-based theoretical approaches. The results of the theoretical analysis of these strategies regarding optimality [30] and well-being [83] outcomes were not associated with positive results.These strategies are not very popular and people tend to pay instead of waiting [83].\n\nTable 2Strategies for changing behavior at the time of receiving health servicesTheme/sourceCategoryCodeOutcome variablesDemand side cost sharing[5, 6, 9, 10, 13, 15, 18–20, 22–25, 27–33, 35–49, 51–57, 59–67, 70–73, 78]Uniformcost sharing- Fixed rate of deductible: traditional deductible, mandatory deductible, First euro deductible/ a first-dollar deductible, doughnut hole deductible- Fixed rate of co-payments or coinsurance/co-insurance: /copayment/ user charges / user-fee, lump sum co-payments, fixed payment, Mandatory co-payments- Fixed-rate insurance coverage limit /caps on insurance /stop loss. / payment ceiling limit on coverage, limit on out-of-pocket expenses- Health services utilization- Health costs- Health related behavior- Choice of insurance plan- The opinion and acceptance of consumersDifferentialcost sharing- Income base cost sharing: income base deductible: income-related copayment /coinsurance/ different copayment according to the socioeconomic status- Varied deducible: Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles- Tier cost sharing: :multitier copayments / tiered copayment/ price-related co-payment tier, tier coinsurance- Value-based cost sharing: Value Based Insurance Design (VBID) / value-based cost sharing (lower coinsurance for services with higher costs benefits / value-based coinsurance target / variable co-insurance based on demand elasticity/ treatment-specific copayments disease-specific cost sharing/ differential cost sharing based on disease status- Cost sharing with discount: co-payment exemption, co-payment with rebate, / waiving copaysHealth savings accounts(HSAs)[26, 50, 54, 58, 60, 74, 75]Voluntary Health Savings Accounts (HSAs)- Health savings accounts (HSAs) / Health Reimbursement Accounts (HRAs) / medical savings accounts (MSAs) in combination with private insurance- Consumer-Directed Health Plans (CDHPS) /Consumer-Driven Health Plans (CDHPS) / Three-Tier Payment System(HDHPS Coupled With Personal Savings Account)- Health services utilization- Health costs- Saving for futureCompulsory Health Savings Accounts (HSAs)- Medisave (Medical savings accounts) (MSAs) in combination with social health insurance- Tongdao (MSA )in combination social risk-pooling (SRP)/ medical savings accounts (MSAs) in combination with social insurance pool (SIP)/ the three-tiered design (MSA-deductible-SIP)- Bankuai (MSA) separately to finance outpatient servicesDrug price regulation[21]Uniform pricingdiscriminatory pricing- Uniform pricing/uniform monopoly pricing- Third degree price discrimination / (different prices to different markets or groups) of consumers- Drug utilizationTwo PartPricing- Two-part tariffs.) combines a uniform price with market-specific lump-sum paymentsRationing for health services [10, 31, 34]Rationing by waiting- Waiting lists / queuing /expectancy queue/the cost of the lost time- Waiting time- Optimality- Well-being\n\nStrategies for changing behavior at the time of receiving health services\nDemand side cost sharing\n[5, 6, 9, 10, 13, 15, 18–20, 22–25, 27–33, 35–49, 51–57, 59–67, 70–73, 78]\nUniform\ncost sharing\n- Fixed rate of deductible: traditional deductible, mandatory deductible, First euro deductible/ a first-dollar deductible, doughnut hole deductible\n- Fixed rate of co-payments or coinsurance/co-insurance: /copayment/ user charges / user-fee, lump sum co-payments, fixed payment, Mandatory co-payments\n- Fixed-rate insurance coverage limit /caps on insurance /stop loss. / payment ceiling limit on coverage, limit on out-of-pocket expenses\n- Health services utilization\n- Health costs\n- Health related behavior\n- Choice of insurance plan\n- The opinion and acceptance of consumers\nDifferential\ncost sharing\n- Income base cost sharing: income base deductible: income-related copayment /coinsurance/ different copayment according to the socioeconomic status\n- Varied deducible: Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles\n- Tier cost sharing: :multitier copayments / tiered copayment/ price-related co-payment tier, tier coinsurance\n- Value-based cost sharing: Value Based Insurance Design (VBID) / value-based cost sharing (lower coinsurance for services with higher costs benefits / value-based coinsurance target / variable co-insurance based on demand elasticity/ treatment-specific copayments disease-specific cost sharing/ differential cost sharing based on disease status\n- Cost sharing with discount: co-payment exemption, co-payment with rebate, / waiving copays\nHealth savings accounts\n(HSAs)\n[26, 50, 54, 58, 60, 74, 75]\n- Health savings accounts (HSAs) / Health Reimbursement Accounts (HRAs) / medical savings accounts (MSAs) in combination with private insurance\n- Consumer-Directed Health Plans (CDHPS) /Consumer-Driven Health Plans (CDHPS) / Three-Tier Payment System(HDHPS Coupled With Personal Savings Account)\n- Health services utilization\n- Health costs\n- Saving for future\n- Medisave (Medical savings accounts) (MSAs) in combination with social health insurance\n- Tongdao (MSA )in combination social risk-pooling (SRP)/ medical savings accounts (MSAs) in combination with social insurance pool (SIP)/ the three-tiered design (MSA-deductible-SIP)\n- Bankuai (MSA) separately to finance outpatient services\nDrug price regulation\n[21]\nUniform pricing\ndiscriminatory pricing\n- Uniform pricing/uniform monopoly pricing\n- Third degree price discrimination / (different prices to different markets or groups) of consumers\n- Waiting lists / queuing /expectancy queue/the cost of the lost time\n- Waiting time\n- Optimality\n- Well-being\n[SUBTITLE] Strategies for changing behavior before needing health services [SUBSECTION] The themes in this group deal with the strategies that are applied outside the health system, before the need for healthcare services, and through the consciousness of health and positive financial incentives, increase healthy behavior or prevent unhealthy behavior. Changing individual behaviors to reduce high-risk behaviors and improve health-promoting behaviors is the approach of this group. This group includes two themes: development of incentive insurance programs and community empowerment.\nThe themes in this group deal with the strategies that are applied outside the health system, before the need for healthcare services, and through the consciousness of health and positive financial incentives, increase healthy behavior or prevent unhealthy behavior. Changing individual behaviors to reduce high-risk behaviors and improve health-promoting behaviors is the approach of this group. This group includes two themes: development of incentive insurance programs and community empowerment.\n[SUBTITLE] Development of incentive insurance programs [SUBSECTION] The themes of this group focus on the measures of insurance companies and purchasers. This theme refers to strategies aimed at reducing the risk of disease and the need for health services or unhealthy behaviors and consists of two categories: extending preventive care insurance and developing bonus-oriented insurance. Findings related to this theme were extracted from11 studies [10, 15, 25, 37, 43, 46, 58, 84–87].\n[SUBTITLE] Extending preventive care insurance [SUBSECTION] Expanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84].\nExpanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84].\n[SUBTITLE] Developing bonus-oriented insurance [SUBSECTION] These strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58].\nHealth service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85].\nThese strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58].\nHealth service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85].\nThe themes of this group focus on the measures of insurance companies and purchasers. This theme refers to strategies aimed at reducing the risk of disease and the need for health services or unhealthy behaviors and consists of two categories: extending preventive care insurance and developing bonus-oriented insurance. Findings related to this theme were extracted from11 studies [10, 15, 25, 37, 43, 46, 58, 84–87].\n[SUBTITLE] Extending preventive care insurance [SUBSECTION] Expanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84].\nExpanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84].\n[SUBTITLE] Developing bonus-oriented insurance [SUBSECTION] These strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58].\nHealth service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85].\nThese strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58].\nHealth service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85].\n[SUBTITLE] Community empowerment [SUBSECTION] Community empowerment is the second and last theme in this group with one category called Community education.The theme of ”community empowerment”, mentioned by only one study [33].\n[SUBTITLE] Community education [SUBSECTION] This category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme.\nThe sources from which each code is extracted are provided in Additional file 3.\nThis category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme.\nThe sources from which each code is extracted are provided in Additional file 3.\nCommunity empowerment is the second and last theme in this group with one category called Community education.The theme of ”community empowerment”, mentioned by only one study [33].\n[SUBTITLE] Community education [SUBSECTION] This category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme.\nThe sources from which each code is extracted are provided in Additional file 3.\nThis category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme.\nThe sources from which each code is extracted are provided in Additional file 3.\n[SUBTITLE] Risk of bias consideration [SUBSECTION] The risk of bias assessment in this study consisted of the following: To reduce publication bias, unpublished papers were searched in the Dissertations database of ProQuest for grey literature, but no related papers were found. In this regard, there is a possibility of language bias due to the limitation of non-English articles in publishing or indexing the results and the focus of this study on Persian and English articles, which is mentioned as a limitation in the limitations section.\n\nTable 3Strategies for changing behavior before needing health servicesTheme/sourceCategoryCodeOutcome variablesDevelopment of incentive insurance programs[10, 15, 27, 30, 39, 42, 59, 69, 76, 77, 79]Extending preventive care insurance- Proposing insurance coverage for preventive care- Separating insurance coverage for prevention and treatment- To encourage insureds to use more secondary preventive care- Improving perception of health status through secondary preventive care- Health services utilization- Health costs- Risk-reducing behaviors- Choice of expensive health services healthDevelopment of bonus oriented insurance- Adjusted premiums according level of preventive effort- Premium reduction, bonus payments/ rebate insurance /to encourage non-use or limited use in return for next premium reduction, risk adjustment .no-claims bonus, risk rating premium- No-claim Bonus and Coverage Upper BoundCommunity empowerment [23]Community education- Health promotion education- Civic education about the consequences of unnecessary use of health services- Not evaluated\n\nStrategies for changing behavior before needing health services\nDevelopment of incentive insurance programs\n[10, 15, 27, 30, 39, 42, 59, 69, 76, 77, 79]\n- Proposing insurance coverage for preventive care\n- Separating insurance coverage for prevention and treatment\n- To encourage insureds to use more secondary preventive care\n- Improving perception of health status through secondary preventive care\n- Health services utilization\n- Health costs\n- Risk-reducing behaviors\n- Choice of expensive health services health\n- Adjusted premiums according level of preventive effort\n- Premium reduction, bonus payments/ rebate insurance /to encourage non-use or limited use in return for next premium reduction, risk adjustment .no-claims bonus, risk rating premium\n- No-claim Bonus and Coverage Upper Bound\n- Health promotion education\n- Civic education about the consequences of unnecessary use of health services\nThe risk of bias assessment in this study consisted of the following: To reduce publication bias, unpublished papers were searched in the Dissertations database of ProQuest for grey literature, but no related papers were found. In this regard, there is a possibility of language bias due to the limitation of non-English articles in publishing or indexing the results and the focus of this study on Persian and English articles, which is mentioned as a limitation in the limitations section.\n\nTable 3Strategies for changing behavior before needing health servicesTheme/sourceCategoryCodeOutcome variablesDevelopment of incentive insurance programs[10, 15, 27, 30, 39, 42, 59, 69, 76, 77, 79]Extending preventive care insurance- Proposing insurance coverage for preventive care- Separating insurance coverage for prevention and treatment- To encourage insureds to use more secondary preventive care- Improving perception of health status through secondary preventive care- Health services utilization- Health costs- Risk-reducing behaviors- Choice of expensive health services healthDevelopment of bonus oriented insurance- Adjusted premiums according level of preventive effort- Premium reduction, bonus payments/ rebate insurance /to encourage non-use or limited use in return for next premium reduction, risk adjustment .no-claims bonus, risk rating premium- No-claim Bonus and Coverage Upper BoundCommunity empowerment [23]Community education- Health promotion education- Civic education about the consequences of unnecessary use of health services- Not evaluated\n\nStrategies for changing behavior before needing health services\nDevelopment of incentive insurance programs\n[10, 15, 27, 30, 39, 42, 59, 69, 76, 77, 79]\n- Proposing insurance coverage for preventive care\n- Separating insurance coverage for prevention and treatment\n- To encourage insureds to use more secondary preventive care\n- Improving perception of health status through secondary preventive care\n- Health services utilization\n- Health costs\n- Risk-reducing behaviors\n- Choice of expensive health services health\n- Adjusted premiums according level of preventive effort\n- Premium reduction, bonus payments/ rebate insurance /to encourage non-use or limited use in return for next premium reduction, risk adjustment .no-claims bonus, risk rating premium\n- No-claim Bonus and Coverage Upper Bound\n- Health promotion education\n- Civic education about the consequences of unnecessary use of health services", "The themes (strategies) in this group, which are used when consumers refer to the health system and receive health services by increasing awareness of services costs and other indirect financial consequences (waiting), attempt to increase their responsibility to reduce the consumption of unnecessary health services. The themes of this group consider the changing consumptive behavior of health service consumers. This group includes four themes: demand-side cost-sharing or consumer cost-sharing, health savings accounts, drug price regulation, and rationing for health services.\n[SUBTITLE] Demand side cost sharing [SUBSECTION] Demand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29].\nThe results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71].\nIn our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance.\nDemand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29].\nThe results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71].\nIn our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance.\n[SUBTITLE] Uniform cost-sharing [SUBSECTION] The uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers.\nThe uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers.\n[SUBTITLE] Differential cost-sharing [SUBSECTION] In differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies,\nStrategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount.\nThe studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54].\nThe reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68].\nDespite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50].\nIn differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies,\nStrategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount.\nThe studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54].\nThe reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68].\nDespite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50].", "Demand side cost sharing is a financial tool [25] and a kind of out-of-pocket payment [26] that is used to decrease the demand for health services or improve the utility of useful services [27], these strategies provide incentives to reduce unnecessary demands by paying part of the service cost by consumers [11]. Deductibles (The amount of health costs that a person must pay before the insurance begins to pay), copayment (paying a fixed amount of the cost of each health service unit), coinsurance (paying a percentage of the cost of each health service unit) [28], and capping (applying a cap on out-of-pocket payments or insurance claims) are different forms of cost sharing [15] that their design methods are diverse in insurance systems [29]. Cost sharing is often used in countries with social health insurance systems [30]. In the United States, it is also used in private insurance and Medicare and Medicaid systems [29].\nThe results of the study showed that “demand side cost sharing“ or “consumer cost sharing” were the main strategies used for controlling consumer moral hazard as addressed by 56 studies [5, 6, 9, 10, 13, 15, 18–25, 30–71].\nIn our analysis, demand-side cost-sharing was divided into two categories: uniform cost-sharing and differential cost-sharing, which were classified based on the fixed or different rates of out-of-pocket payments in the form of deductible, copayment, and coinsurance.", "The uniform cost-sharing strategy are also known as traditional cost sharing methods includes strategies (codes) in which the rate of patients’ out-of-pocket payments (deductible, copayment, and coinsurance) is fixed or flat to use each unit of health services for different consumers.", "In differential cost-sharing, deductibles, copayment, and coinsurance are adjusted based on criteria such as income level; health status; consumer choice, and the type, value, price, and elasticity of the health product or service. Differential cost - sharing have been proposed in response to the high sensitivity of low-income people [45, 49, 60, 64, 69] and inefficiency [64] of fixed and low cost - sharing strategies,\nStrategies related to this category include income-based cost sharing, variable deductible plans (Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles), tier cost sharing (higher copay or coinsurance for services with higher price of the product or service(, value-based cost sharing (lower copay or coinsurance for services with higher costs benefits or lower elasticity), differential cost sharing based on disease status and Cost sharing with discount.\nThe studies included in this theme investigated consequences such as the utilization of health services, health costs, health related behavior, the choice of insurance plan, and the opinion and acceptance of consumers using quantitative, qualitative, review, and theoretical approaches (model-based), the results of quantitative studies are reported in narrative form as follows: modest to high reduction in health services and medications utilization [9, 20, 21, 23, 33, 38, 44, 45, 47, 49, 51, 55, 56, 64, 69, 71], reduction in health cost [21, 39, 41, 42, 44, 53, 65, 68], low or no significant effect on health services utilization [6, 34, 35, 40, 62, 63, 65], low or no significant effect on health costs [5], increasing cost contaminate incentive (CCI) [57], significant correlation with higher preventive behavior [13, 50] modest efficiency gain [18], increasing medication adherence [48], increasing demand for low price drug [61], optimal insurance [54].\nThe reported negative consequences are as follows: Decreasing the utilization of both necessary and unnecessary care [44, 55, 71], substitution effect from cares with cost-sharing to free care or with lower cost sharing [18, 70], and vulnerability of low-income groups [49, 60, 68].\nDespite the focus of differential strategies on eliminating the shortcomings of uniform strategies, some studies indicate the ineffectiveness of these strategies in response to the problem of reduced consumption of non-essential services [44], delayed treatment, and medical debt in the vulnerable group [68]. Also, the low level of unhealthy behavior in members of HDHP may be due to the individual characteristics of the people who chose the plan not the impact of the plan [50].", "HSAs are considered as alternative tools for financing and dealing with future demographic challenges [72]. This financing theme under the title of health savings accounts [73, 74] or medical savings accounts [72, 75, 76], are currently used to decrease moral hazard and cost in four countries around the world [72, 75]. In this mechanism, owners of savings accounts should save a certain percentage of their income in these accounts for future health expenses. Funds of these accounts are used to pay for health expenses. Saving accounts increase people’s motivation to take responsibility by providing tax benefits and informed participation in health care decisions based on cost awareness and monitoring of physicians’ decisions [73].\nIn the United States and South Africa, they are used in combination with private insurance and is voluntarily. In Singapore and China, health saving accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system, respectively [72, 75]. This strategy in private health insurance and combination with high deductible health plans (HDHPs) is known as consumer directed health plans or consumer-driven health plans (CDHPs) [59].\nIn our study, health savings accounts were divided into two categories: voluntary health savings accounts (HSAs) and compulsory health savings accounts (HSAs), which were classified according to the mandatory and optional nature of the plan and the type of financing system in each country.\n[SUBTITLE] Voluntary health savings accounts [SUBSECTION] In this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75].\nIn this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75].\n[SUBTITLE] Compulsory health savings accounts [SUBSECTION] Compulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75].\nFindings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76].\nSome experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74].\nCompulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75].\nFindings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76].\nSome experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74].", "In this category, health savings accounts are optional, and in combination with private insurance and high deductible health plans (HDHPs)that is known as consumer directed health plans or consumer-driven health plans (CDHPs)[59]. In the United States and South Africa, this strategy are used [75].", "Compulsory health savings accounts are governmental and compulsory which are implemented in combination with social health insurance and social risk-pooling system. Medisave (Medical savings accounts) (MSAs) in combination with social health insurance, Tongdao (MSA) in combination social risk-pooling (SRP) and Bankuai: (MSA) separately to finance outpatient services, are related to this category [75].\nFindings related to health savings accounts were extracted from seven studies [55, 59, 72–76]. In studies related to this theme, the outcome such as health services utilization, health costs, and health promotion behaviors was assess using quantitative, review, and model-based theoretical approaches. The narrative reports of these outcomes in the quantitative studies are as follows: reduction in health services utilization and cost [75], useful for future savings [75] No or less effective in controlling healthcare costs [74, 76].\nSome experts regard the usefulness of precautionary savings as positive point of these strategies [72]. In a study that assessed the effect of these strategies for prevention efforts and precautionary savings, it is stated that consumers do not take these two measures at the same time; in case of precautionary savings, preventive action is reduced, and vice versa [73]. Adverse selection, consequences of inflation, reduction of equity, and restraint of essential consumption are other negative consequences of this strategy [74].", "Drug pricing is an influential component of drug access and rational use of drugs. In addition to improving access, consumption management should be considered [77]. Drug price regulation is the third theme of this group, with three categories, uniform pricing, discriminatory pricing and two part pricing, which are based on drug pricing policies and the fix or different prices for each drug unit. As the findings of this theme were extracted from only one study, the categories related to this theme included one code that could not be combined and summarized further due to dissimilarity.\n[SUBTITLE] Uniform pricing [SUBSECTION] Uniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81].\nUniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81].\n[SUBTITLE] Discriminatory pricing [SUBSECTION] Discriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81].\nDiscriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81].\n[SUBTITLE] Two part pricing [SUBSECTION] Two-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79].\nResults related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81].\nTwo-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79].\nResults related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81].", "Uniform pricing refers to strategies in which a product is offered at the same price for all market segment regardless of the characteristics of each segments and its ability to pay [78]. It is one of the traditional methods of pricing in the pharmaceutical industry [79]. This pricing method, despite the ease of administration, is not able to satisfy all market segments. From the perspective of high-level customers, the suggested price may be low and indicate low desirability, whereas low-income customers may consider the price high and avoid buying it [80]. Results related to this category were extracted from a study [81].", "Discriminatory pricing offers different prices for the same drug in different markets or groups [78]. Price discrimination is caused by the inability of developing countries to provide the medicines they need. Discriminatory pricing involves a segmented market that charges different prices based on each country’s ability to pay [77]. This category includes different types; however, in this study, only third degree price discrimination was introduced as an intervention to control moral hazard [81].", "Two-part pricing, another name for two-part tariffs, determines the price of medicine from the combination of uniform price and lump-sum payments [81] which has recently been proposed instead of uniform pricing for drugs [79].\nResults related to the “drug pricing” theme were extracted from a study [81]. In this study, three types of pricing mechanisms, including uniform pricing, two-part tariffs and third degree price discrimination were compared in order to control the consumer’s moral hazard, the results showed two-part tariffs were considered a better strategy to address consumer moral hazard [81].", "Rationing of health services based on the waiting list (number) and waiting time (period) is one of the demand management strategies for non-emergency and elective health services [82]. Rationing of health services is the last theme that has one category named “rationing by waiting” which refers to strategies that control the consumer moral hazard by considering the cost of lost time. This strategy is one of the ways to reduce health costs that replaces user payments in countries without this system (national health system) to control costs and reduces unnecessary demand by imposing costs through the queue [30]. Findings related to the “Rationing of health services” were extracted from three studies [10, 30, 83]. These three studies investigated this mechanism using review and model-based theoretical approaches. The results of the theoretical analysis of these strategies regarding optimality [30] and well-being [83] outcomes were not associated with positive results.These strategies are not very popular and people tend to pay instead of waiting [83].\n\nTable 2Strategies for changing behavior at the time of receiving health servicesTheme/sourceCategoryCodeOutcome variablesDemand side cost sharing[5, 6, 9, 10, 13, 15, 18–20, 22–25, 27–33, 35–49, 51–57, 59–67, 70–73, 78]Uniformcost sharing- Fixed rate of deductible: traditional deductible, mandatory deductible, First euro deductible/ a first-dollar deductible, doughnut hole deductible- Fixed rate of co-payments or coinsurance/co-insurance: /copayment/ user charges / user-fee, lump sum co-payments, fixed payment, Mandatory co-payments- Fixed-rate insurance coverage limit /caps on insurance /stop loss. / payment ceiling limit on coverage, limit on out-of-pocket expenses- Health services utilization- Health costs- Health related behavior- Choice of insurance plan- The opinion and acceptance of consumersDifferentialcost sharing- Income base cost sharing: income base deductible: income-related copayment /coinsurance/ different copayment according to the socioeconomic status- Varied deducible: Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles- Tier cost sharing: :multitier copayments / tiered copayment/ price-related co-payment tier, tier coinsurance- Value-based cost sharing: Value Based Insurance Design (VBID) / value-based cost sharing (lower coinsurance for services with higher costs benefits / value-based coinsurance target / variable co-insurance based on demand elasticity/ treatment-specific copayments disease-specific cost sharing/ differential cost sharing based on disease status- Cost sharing with discount: co-payment exemption, co-payment with rebate, / waiving copaysHealth savings accounts(HSAs)[26, 50, 54, 58, 60, 74, 75]Voluntary Health Savings Accounts (HSAs)- Health savings accounts (HSAs) / Health Reimbursement Accounts (HRAs) / medical savings accounts (MSAs) in combination with private insurance- Consumer-Directed Health Plans (CDHPS) /Consumer-Driven Health Plans (CDHPS) / Three-Tier Payment System(HDHPS Coupled With Personal Savings Account)- Health services utilization- Health costs- Saving for futureCompulsory Health Savings Accounts (HSAs)- Medisave (Medical savings accounts) (MSAs) in combination with social health insurance- Tongdao (MSA )in combination social risk-pooling (SRP)/ medical savings accounts (MSAs) in combination with social insurance pool (SIP)/ the three-tiered design (MSA-deductible-SIP)- Bankuai (MSA) separately to finance outpatient servicesDrug price regulation[21]Uniform pricingdiscriminatory pricing- Uniform pricing/uniform monopoly pricing- Third degree price discrimination / (different prices to different markets or groups) of consumers- Drug utilizationTwo PartPricing- Two-part tariffs.) combines a uniform price with market-specific lump-sum paymentsRationing for health services [10, 31, 34]Rationing by waiting- Waiting lists / queuing /expectancy queue/the cost of the lost time- Waiting time- Optimality- Well-being\n\nStrategies for changing behavior at the time of receiving health services\nDemand side cost sharing\n[5, 6, 9, 10, 13, 15, 18–20, 22–25, 27–33, 35–49, 51–57, 59–67, 70–73, 78]\nUniform\ncost sharing\n- Fixed rate of deductible: traditional deductible, mandatory deductible, First euro deductible/ a first-dollar deductible, doughnut hole deductible\n- Fixed rate of co-payments or coinsurance/co-insurance: /copayment/ user charges / user-fee, lump sum co-payments, fixed payment, Mandatory co-payments\n- Fixed-rate insurance coverage limit /caps on insurance /stop loss. / payment ceiling limit on coverage, limit on out-of-pocket expenses\n- Health services utilization\n- Health costs\n- Health related behavior\n- Choice of insurance plan\n- The opinion and acceptance of consumers\nDifferential\ncost sharing\n- Income base cost sharing: income base deductible: income-related copayment /coinsurance/ different copayment according to the socioeconomic status\n- Varied deducible: Shift deductible, variable deductible, different size of deductible. optional deductible, voluntary deductible (VD), high-deductible health plans (HDHPs), higher insurance deductibles\n- Tier cost sharing: :multitier copayments / tiered copayment/ price-related co-payment tier, tier coinsurance\n- Value-based cost sharing: Value Based Insurance Design (VBID) / value-based cost sharing (lower coinsurance for services with higher costs benefits / value-based coinsurance target / variable co-insurance based on demand elasticity/ treatment-specific copayments disease-specific cost sharing/ differential cost sharing based on disease status\n- Cost sharing with discount: co-payment exemption, co-payment with rebate, / waiving copays\nHealth savings accounts\n(HSAs)\n[26, 50, 54, 58, 60, 74, 75]\n- Health savings accounts (HSAs) / Health Reimbursement Accounts (HRAs) / medical savings accounts (MSAs) in combination with private insurance\n- Consumer-Directed Health Plans (CDHPS) /Consumer-Driven Health Plans (CDHPS) / Three-Tier Payment System(HDHPS Coupled With Personal Savings Account)\n- Health services utilization\n- Health costs\n- Saving for future\n- Medisave (Medical savings accounts) (MSAs) in combination with social health insurance\n- Tongdao (MSA )in combination social risk-pooling (SRP)/ medical savings accounts (MSAs) in combination with social insurance pool (SIP)/ the three-tiered design (MSA-deductible-SIP)\n- Bankuai (MSA) separately to finance outpatient services\nDrug price regulation\n[21]\nUniform pricing\ndiscriminatory pricing\n- Uniform pricing/uniform monopoly pricing\n- Third degree price discrimination / (different prices to different markets or groups) of consumers\n- Waiting lists / queuing /expectancy queue/the cost of the lost time\n- Waiting time\n- Optimality\n- Well-being", "The themes in this group deal with the strategies that are applied outside the health system, before the need for healthcare services, and through the consciousness of health and positive financial incentives, increase healthy behavior or prevent unhealthy behavior. Changing individual behaviors to reduce high-risk behaviors and improve health-promoting behaviors is the approach of this group. This group includes two themes: development of incentive insurance programs and community empowerment.", "The themes of this group focus on the measures of insurance companies and purchasers. This theme refers to strategies aimed at reducing the risk of disease and the need for health services or unhealthy behaviors and consists of two categories: extending preventive care insurance and developing bonus-oriented insurance. Findings related to this theme were extracted from11 studies [10, 15, 25, 37, 43, 46, 58, 84–87].\n[SUBTITLE] Extending preventive care insurance [SUBSECTION] Expanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84].\nExpanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84].\n[SUBTITLE] Developing bonus-oriented insurance [SUBSECTION] These strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58].\nHealth service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85].\nThese strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58].\nHealth service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85].", "Expanding preventive care insurance refers to strategies that, by developing various types of preventive insurance, sensitize consumers to their health and reduce the need for more health services in the future by preventing the deterioration of their health status. Additionally, these strategies prevent the demand for specialized and expensive services by providing medium insurance plans [37, 84]. This category includes the following strategies (code): proposing insurance coverage for preventive care [37], separating insurance coverage for prevention and treatment [37], to encourage insureds to use more secondary preventive care [84] and improving perception of health status through secondary preventive care [84].", "These strategies reduce unnecessary consumption by providing incentives to avoid inefficient service. In these strategies, insurance attempts to control consumers’ moral hazard by applying positive financial incentives in the form of premium discounts [10], or more coverage [86] in the following year’s contract, in the case of less service consumption or applying preventive effort Bonuses for non-consumption or limited consumption are often used in risk adjustment schemes [58].\nHealth service utilization, health costs, risk-reducing behaviors, and choice of expensive health services were among the variables investigated in studies of this them in quantitative, qualitative, and theoretical approaches. The results of these studies can be summarized as follows: more feasibility to incentivize consumers to purchase more secondary preventive care [84] higher reduction in moral hazard in the copayment with a premium reduction frame than copayment reduction frame [46] moral hazard reduction in voluntary deductible is expected to be larger in a system with risk-rated premiums than in a system with community-rated premiums [85].", "Community empowerment is the second and last theme in this group with one category called Community education.The theme of ”community empowerment”, mentioned by only one study [33].\n[SUBTITLE] Community education [SUBSECTION] This category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme.\nThe sources from which each code is extracted are provided in Additional file 3.\nThis category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme.\nThe sources from which each code is extracted are provided in Additional file 3.", "This category refers to the development of health-promoting behaviors through community education and increasing people’s awareness of the function of insurance and the consequences of the unnecessary use of health services by using the capacity of civil society [33]. Despite the fact that only one study had dealt with this issue marginally, due to the importance of the subject, the research team decided to set this code as an independent theme.\nThe sources from which each code is extracted are provided in Additional file 3.", "The risk of bias assessment in this study consisted of the following: To reduce publication bias, unpublished papers were searched in the Dissertations database of ProQuest for grey literature, but no related papers were found. In this regard, there is a possibility of language bias due to the limitation of non-English articles in publishing or indexing the results and the focus of this study on Persian and English articles, which is mentioned as a limitation in the limitations section.\n\nTable 3Strategies for changing behavior before needing health servicesTheme/sourceCategoryCodeOutcome variablesDevelopment of incentive insurance programs[10, 15, 27, 30, 39, 42, 59, 69, 76, 77, 79]Extending preventive care insurance- Proposing insurance coverage for preventive care- Separating insurance coverage for prevention and treatment- To encourage insureds to use more secondary preventive care- Improving perception of health status through secondary preventive care- Health services utilization- Health costs- Risk-reducing behaviors- Choice of expensive health services healthDevelopment of bonus oriented insurance- Adjusted premiums according level of preventive effort- Premium reduction, bonus payments/ rebate insurance /to encourage non-use or limited use in return for next premium reduction, risk adjustment .no-claims bonus, risk rating premium- No-claim Bonus and Coverage Upper BoundCommunity empowerment [23]Community education- Health promotion education- Civic education about the consequences of unnecessary use of health services- Not evaluated\n\nStrategies for changing behavior before needing health services\nDevelopment of incentive insurance programs\n[10, 15, 27, 30, 39, 42, 59, 69, 76, 77, 79]\n- Proposing insurance coverage for preventive care\n- Separating insurance coverage for prevention and treatment\n- To encourage insureds to use more secondary preventive care\n- Improving perception of health status through secondary preventive care\n- Health services utilization\n- Health costs\n- Risk-reducing behaviors\n- Choice of expensive health services health\n- Adjusted premiums according level of preventive effort\n- Premium reduction, bonus payments/ rebate insurance /to encourage non-use or limited use in return for next premium reduction, risk adjustment .no-claims bonus, risk rating premium\n- No-claim Bonus and Coverage Upper Bound\n- Health promotion education\n- Civic education about the consequences of unnecessary use of health services", "The aim of this study was to determine and analyze strategies used to control consumer moral hazards in health systems. A wide range of goals, approaches, and various research designs have been investigated and reported.\nControlling strategies for consumer moral hazard are known as demand-side strategies whose goal are to motivate consumers to reduce unnecessary demand or consumption. In this study, the strategies to control the consumer moral hazard were divided into two groups.The first group aims to control consumer consumptive behavior when receiving health services. The second group focuses on reducing the need for health services by controlling health-related behaviors before needing health services. This classification of controlling strategies was taken from the approach of dividing moral hazards into ex-post and ex-ante moral hazard. Ex-post moral hazard means an increase in demand for health services due to price reduction, which indicates consumer price sensitivity [85]. Ex-ante moral hazard refers to a reduction in preventive behaviors and an increase in risky behaviors due to insurance coverage [88, 89].\nThe results of this study show a greater frequency of studies related to the strategies of the first group and control of consumptive behavior, In contrast, strategies used for changing health-related behavior are limited which indicates that researchers pay more attention to ex-post moral hazard. Ex-post moral hazard has been widely studied, but evidence of ex-ante moral hazard is very limited [88, 90]. The reason for less attention paid to the ex-ante moral hazard modeled by Ehrlich and Becker in 1972 may be criticized as follows: cost is not the only consequence of illness that, if paid by someone else, makes people indifferent to their healthcare [90, 91].\nAnother noteworthy point of this study’s findings is the financial nature of most strategies, including demand-side cost sharing as one of the most effective methods, health savings accounts, and drug pricing. Imposing a cost through the waiting list can also be considered a financial tool. Access restrictions caused by the negative financial incentives is one of the adverse effects of these strategies. A recent study on cost sharing showed a significant relationship between cost sharing and adult mortality in poor countries. The authors believe that this issue should be considered when analyzing the social welfare consequences of cost-sharing [92]. Also, during the analysis, Michaela et al. stated that medical savings accounts cause inequality and provide little financial protection [93].\nAlthough bonus insurance is a positive incentive financial tool because the individual receives a reward in the form of a premium discount or more coverage in exchange for reduced or non-consumption, the results of a qualitative study in insured individuals proved these strategies to be less optimistic and justified compared to demand-side cost sharing strategies [25]. In addition, the community empowerment strategy, despite being a non-financial tool, needs further investigation in future studies due to the limitations of effective studies.\nThe outcomes analyzed in the included studies are other points of debate in this review. The majority of the outcomes analyzed included demand and utilization of health services [5, 15, 18–24, 30, 33, 37–40, 43–45, 47–51, 55, 56, 58, 60, 62–65, 68–71, 75, 84, 86] and health services costs and expenditures, optimality and efficiency of strategies [5, 15, 30–32, 36, 37, 39, 41–44, 53, 54, 57, 59, 68, 70, 73–76, 83, 85, 87, 94]. Limited studies have addressed other important aspects such as access to low-income people [49, 60, 68], reduction in the consumption of both essential (such as preventive and diagnostic services) and non-essential services [24, 44, 58], and people’s attitudes and acceptance [25, 34, 52, 67].\nThe impact of strategies on outcomes such as the utilization of health services and health costs has been different, which seems normal due to the different implementation and management methods and whether the programs are mandatory or optional. However, in this context, the point to consider is to pay attention to the negative consequences, including the higher sensitivity of low-income groups and the shifting financial burden to insureds and an increase in total costs of health costs due to the substitution effect especially in cost-sharing methods. Thesee consequences challenge the achievement of equity in the access and efficiency of the health system. Since moral hazard is one of the factors of the inefficiency of the health system [95] therefore, in its control, improving efficiency should be the most important goal.\nIn this regard, the results of this research showed that controlling strategies need further investigation in future study. Due to the focus of most studies on the controlling strategies of consumptive behavior at the point of receiving the service, therefore, the suggestions are as follows. Reviewing existing strategies, especially strategies with negative financial incentives to minimize adverse consequences, paying more attention to current strategies from the perspective of preventing the need for health services and the introduction of new strategies with preferably non-financial approaches that do not limit access. Obviously, in the design of new interventions and revision of existing interventions, important consequences such as access, financial protection, equality, and quality of services provided along with service utilization and service costs should be taken into consideration.\nOn the other hand, considering that each of these strategies is used in different health systems with different financing mechanisms, so managers in each health system need to adjust strategy to the characteristics of their health system. In addition, considering the nature of behavioral change of strategies, knowing the characteristics of consumers, the pattern and culture of health service consumption and their health-related behaviors is the first step to choosing the most appropriate strategy and adapting it to each Society’s conditions.", "This study has several limitations. The first, is the restriction of studies in Persian and English languages. No clear boundary between the consumer and provider moral hazard in some articles is another limitation. The researchers separated these two issues by studying the full text of the articles, focusing on the type and setting of service delivery and the role of physicians in providing services. The last limitation was the methodological diversity and heterogeneity of the quantitative studies, which did not allow for quantitative analysis and reporting the effectiveness of the strategies.", "Strategies to control consumer moral hazards focus on changing consumer consumptive and health-related behaviors, which are designed according to the structure of health and financing systems. Since “changing consumptive behavior” strategies are the most commonly used strategies; therefore, it is necessary to strengthen strategies to control health-related behaviors and develop new strategies in future studies. In addition, in the application of existing strategies, the adaptation to the structure of the health and financing system, and the pattern of consumption of health services in society should be considered.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2\n\nSupplementary Material 3\n\nSupplementary Material 3\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2\n\nSupplementary Material 3\n\nSupplementary Material 3", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2\n\nSupplementary Material 3\n\nSupplementary Material 3" ]
[ null, null, null, null, null, null, null, "results", null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, "discussion", null, "conclusion", "supplementary-material", null ]
[ "Consumer Moral hazard", "Health system", "Systematic review" ]
Association between exercise and risk of cardiovascular diseases in patients with non-cystic fibrosis bronchiectasis.
36258193
Although cardiovascular comorbidities negatively impact survival in patients with bronchiectasis, there is limited evidence to recommend exercise in this population. We aimed to evaluate whether exercise habit changes are related to reduced cardiovascular disease risk and explore an optimal exercise amount.
BACKGROUND
This study identified 165,842 patients with newly diagnosed bronchiectasis during 2010-2016 who underwent two health examinations and were followed up until December 2020. The exposure was the change in weekly habits of moderate- or vigorous-intensity physical activity between the two examinations, classified into non-exercisers and exercisers (further classified into new exercisers, exercise dropouts, and exercise maintainers). The amount of exercise was measured using metabolic equivalents of task (MET). The outcome was the incidence of myocardial infarction (MI) or stroke.
METHODS
During a mean of 6.2 ± 2.1 follow-up years, 4,233 (2.6%) and 3,745 (2.3%) of patients with bronchiectasis had MI or stroke, respectively. Compared to non-exercisers, exercisers had a significantly lower risk of MI or stroke by 9-28% (p < 0.001 for both). Among exercisers, exercise maintainers showed the lowest risk of MI (adjusted hazard ratio [aHR], 0.72; 95% confidence interval [CI], 0.64-0.81) and stroke (aHR, 0.72; 95% CI, 0.64-0.82) compared to non-exercisers. Regarding exercise amount, a significant risk reduction was observed only in patients with bronchiectasis who exercised for ≥ 500 MET-min/wk.
RESULTS
Exercise was associated with a reduced risk of cardiovascular diseases in patients with bronchiectasis. In particular, the risk was lowest in exercise maintainers, and cardiovascular risk reduction was significant when exercising more than 500 MET-min/wk.
CONCLUSION
[ "Humans", "Cardiovascular Diseases", "Bronchiectasis", "Incidence", "Stroke", "Fibrosis", "Risk Factors" ]
9580142
Introduction
The prevalence and disease burden of non-cystic fibrosis bronchiectasis (hereafter referred to as bronchiectasis) have been substantially increasing worldwide [1–3]. The prevalence is as high as 464–566 cases per 100,000 population [1, 2, 4, 5]. Furthermore, patients with bronchiectasis have a higher mortality risk than the general population (comparative mortality figures of 2.26 in women and 2.14 in men) [1] or than the age-, sex-, and comorbidity-matched patients without bronchiectasis (hazard ratio, 1.15) [6]. According to the literature, comorbid cardiovascular diseases largely explain higher mortality in patients with bronchiectasis [7, 8]. Hence, alleviating the risk of cardiovascular diseases can significantly reduce the long-term mortality of patients with bronchiectasis. Despite the importance of preventing cardiovascular diseases, there is a limited evidence to recommend appropriate prevention strategies for cardiovascular comorbidities in patients with bronchiectasis. Regular exercise and physical activity are known to be effective interventions for preventing cardiovascular diseases in various populations [9]. However, there have been no studies evaluating its protective effects against cardiovascular diseases in patients with bronchiectasis. Although the current international bronchiectasis guidelines recommend regular exercise and participation in a pulmonary rehabilitation program [10, 11], the main reasons for these recommendations are to improve exercise capacity and respiratory symptoms in patients with bronchiectasis [12, 13] and not to prevent their cardiovascular comorbidities. Additionally, there is limited evidence regarding the effects of exercise and physical activity on the long-term outcomes of bronchiectasis, particularly cardiovascular comorbidities, and the optimal amount of physical activity beneficial for cardiovascular outcomes in patients with bronchiectasis. From this view, evidence-based recommendations to guide optimal exercise methods would be invaluable in preventing cardiovascular diseases and possibly improving survival in patients with bronchiectasis. This large population-based study aimed to investigate the effects of changing the amount of physical activity on cardiovascular outcomes in patients with bronchiectasis.
null
null
Results
[SUBTITLE] Baseline characteristics [SUBSECTION] In a total of 165,842 patients with bronchiectasis, the mean age was 59.8 years, and 48.4% were men. The study population comprised 66.6% non-exercisers and 33.4% exercisers. Moreover, exercisers were composed of new exercisers (n = 21,056, 12.7%), exercise dropouts (n = 19,377, 11.7%), and exercise maintainers (n = 14,910, 9.0%) according to changes in exercise habits (Fig. 1). Exercisers were more likely to be older, male, non-smokers, and from low-income groups than non-exercisers (all p < 0.001). Regarding measurements in health screening exams, exercisers had lower total cholesterol levels, eGFR, and proportion of central obesity than non-exercisers (all p < 0.001); however, exercisers showed higher BMI, fasting blood sugar, systolic blood pressure, and longer waist circumference than non-exercisers (all p < 0.001). Of comorbidities, exercisers showed higher rates of hypertension, dyslipidemia, diabetes mellitus, and asthma or COPD compared with non-exercisers, and the differences between the four categories were statistically significant. Meanwhile, non-exercisers showed the lowest energy expenditure (32.5% for ≥ 500 MET-min/wk), followed by exercise dropouts (46.5%), new exercisers (97.8%), and exercise maintainers (98.1%) (p < 0.001) (Table 1). Table 1Baseline characteristics of the study populationVariablesTotal(N = 165,842)Non-exercisers(n = 110,499, 66.6%)Exercisers (n = 55,343, 33.4%)New exercisers(n = 21,056, 12.7%)Exercise dropouts(n = 19,377, 11.7%)Exercise maintainers(n = 14,910, 9.0%)P-value Sociodemographics Age, years59.8 ± 11.959.5 ± 12.360.1 ± 11.160.8 ± 11.360.4 ± 10.6< 0.001  < 40 years9,621 (5.8)7,215 (6.5)950 (4.5)917 (4.7)539 (3.6)< 0.001  40–64 years97,408 (58.7)64,907 (58.8)12,625 (60.0)10,952 (56.5)8,924 (59.9)  ≥ 65 years58,813 (35.5)38,377 (34.7)7,481 (35.5)7,508 (38.8)5,447 (36.5)Sex< 0.001  Male80,336 (48.4)50,714 (45.9)10,856 (51.6)9,935 (51.3)8,831 (59.2)  Female85,506 (51.6)59,785 (54.1)10,200 (48.4)9,442 (48.7)6,079 (40.8)Low income*31,365 (18.9)21,210 (19.2)3,923 (18.6)3,674 (19.0)2,558 (17.2)< 0.001Smoking status  Current or past smoker24,294 (14.7)17,239 (15.6)2,739 (13.0)2,526 (13.0)1,790 (12.0)< 0.001Alcohol consumption  Heavy drinker8,402 (5.1)5,579 (5.1)1,093 (5.2)865 (4.5)865 (5.8)< 0.001 Measurements Body mass index, kg/m223.5 ± 3.323.5 ± 3.323.6 ± 3.123.7 ± 3.223.7 ± 3.0< 0.001Waist circumference, cm80.9 ± 9.180.8 ± 9.380.9 ± 8.981.3 ± 8.981.2 ± 8.6< 0.001Central obesity38,903 (23.5)26,626 (24.1)4,653 (22.1)4,514 (23.3)3,110 (20.9)< 0.001Fasting glucose, mg/dL100.3 ± 23.0100.0 ± 23.1100.4 ± 22.7101.0 ± 23.3101.1 ± 22.8< 0.001Systolic BP, mmHg123.3 ± 14.6123.1 ± 14.8123.4 ± 14.4123.8 ± 14.5123.7 ± 14.1< 0.001Diastolic BP, mmHg75.7 ± 9.575.7 ± 9.675.7 ± 9.475.9 ± 9.475.8 ± 9.40.057Total cholesterol, mg/dL193.7 ± 37.8194.2 ± 38.0192.7 ± 37.5193.1 ± 37.9192.1 ± 36.5< 0.001eGFR, ml/min/1.73m289.1 ± 40.789.5 ± 41.089.0 ± 44.787.9 ± 33.688.1 ± 41.5< 0.001 Comorbidities Hypertension64,250 (38.7)42,287 (38.3)8,204 (39.0)7,884 (40.7)5,875 (39.4)< 0.001Dyslipidemia50,422 (30.4)32,984 (29.9)6,496 (30.9)6,326 (32.7)4,616 (31.0)< 0.001Diabetes mellitus22,762 (13.7)14,623 (13.2)2,985 (14.2)2,981 (15.4)2,173 (14.6)< 0.001Chronic kidney disease10,948 (6.6)7,333 (6.6)1,355 (6.4)1,355 (7.0)905 (6.1)0.005Asthma or COPD7,885 (4.8)4,871 (4.4)1,204 (5.7)975 (5.0)835 (5.6)< 0.001Cancer57,243 (34.5)38,630 (35.0)7,120 (33.8)6,810 (35.1)4,683 (31.4)< 0.001 Amounts of exercise Energy expenditure,MET-min/wk624.1 ± 635.2347.0 ± 330.71,552.3 ± 556.7463.0 ± 356.31,618.9 ± 585.2< 0.001≥ 500 MET-min/wk80,102 (48.3)35,871 (32.5)20,599 (97.8)9,000 (46.5)14,632 (98.1)< 0.001Data are presented as number (percentage) or mean ± standard deviation*Low income denotes income in the lowest 20% of the entire Korean populationBP blood pressure, eGFR estimated glomerular filtration rate, COPD chronic obstructive pulmonary disease, MET metabolic equivalent of task Baseline characteristics of the study population Energy expenditure, MET-min/wk Data are presented as number (percentage) or mean ± standard deviation *Low income denotes income in the lowest 20% of the entire Korean population BP blood pressure, eGFR estimated glomerular filtration rate, COPD chronic obstructive pulmonary disease, MET metabolic equivalent of task In a total of 165,842 patients with bronchiectasis, the mean age was 59.8 years, and 48.4% were men. The study population comprised 66.6% non-exercisers and 33.4% exercisers. Moreover, exercisers were composed of new exercisers (n = 21,056, 12.7%), exercise dropouts (n = 19,377, 11.7%), and exercise maintainers (n = 14,910, 9.0%) according to changes in exercise habits (Fig. 1). Exercisers were more likely to be older, male, non-smokers, and from low-income groups than non-exercisers (all p < 0.001). Regarding measurements in health screening exams, exercisers had lower total cholesterol levels, eGFR, and proportion of central obesity than non-exercisers (all p < 0.001); however, exercisers showed higher BMI, fasting blood sugar, systolic blood pressure, and longer waist circumference than non-exercisers (all p < 0.001). Of comorbidities, exercisers showed higher rates of hypertension, dyslipidemia, diabetes mellitus, and asthma or COPD compared with non-exercisers, and the differences between the four categories were statistically significant. Meanwhile, non-exercisers showed the lowest energy expenditure (32.5% for ≥ 500 MET-min/wk), followed by exercise dropouts (46.5%), new exercisers (97.8%), and exercise maintainers (98.1%) (p < 0.001) (Table 1). Table 1Baseline characteristics of the study populationVariablesTotal(N = 165,842)Non-exercisers(n = 110,499, 66.6%)Exercisers (n = 55,343, 33.4%)New exercisers(n = 21,056, 12.7%)Exercise dropouts(n = 19,377, 11.7%)Exercise maintainers(n = 14,910, 9.0%)P-value Sociodemographics Age, years59.8 ± 11.959.5 ± 12.360.1 ± 11.160.8 ± 11.360.4 ± 10.6< 0.001  < 40 years9,621 (5.8)7,215 (6.5)950 (4.5)917 (4.7)539 (3.6)< 0.001  40–64 years97,408 (58.7)64,907 (58.8)12,625 (60.0)10,952 (56.5)8,924 (59.9)  ≥ 65 years58,813 (35.5)38,377 (34.7)7,481 (35.5)7,508 (38.8)5,447 (36.5)Sex< 0.001  Male80,336 (48.4)50,714 (45.9)10,856 (51.6)9,935 (51.3)8,831 (59.2)  Female85,506 (51.6)59,785 (54.1)10,200 (48.4)9,442 (48.7)6,079 (40.8)Low income*31,365 (18.9)21,210 (19.2)3,923 (18.6)3,674 (19.0)2,558 (17.2)< 0.001Smoking status  Current or past smoker24,294 (14.7)17,239 (15.6)2,739 (13.0)2,526 (13.0)1,790 (12.0)< 0.001Alcohol consumption  Heavy drinker8,402 (5.1)5,579 (5.1)1,093 (5.2)865 (4.5)865 (5.8)< 0.001 Measurements Body mass index, kg/m223.5 ± 3.323.5 ± 3.323.6 ± 3.123.7 ± 3.223.7 ± 3.0< 0.001Waist circumference, cm80.9 ± 9.180.8 ± 9.380.9 ± 8.981.3 ± 8.981.2 ± 8.6< 0.001Central obesity38,903 (23.5)26,626 (24.1)4,653 (22.1)4,514 (23.3)3,110 (20.9)< 0.001Fasting glucose, mg/dL100.3 ± 23.0100.0 ± 23.1100.4 ± 22.7101.0 ± 23.3101.1 ± 22.8< 0.001Systolic BP, mmHg123.3 ± 14.6123.1 ± 14.8123.4 ± 14.4123.8 ± 14.5123.7 ± 14.1< 0.001Diastolic BP, mmHg75.7 ± 9.575.7 ± 9.675.7 ± 9.475.9 ± 9.475.8 ± 9.40.057Total cholesterol, mg/dL193.7 ± 37.8194.2 ± 38.0192.7 ± 37.5193.1 ± 37.9192.1 ± 36.5< 0.001eGFR, ml/min/1.73m289.1 ± 40.789.5 ± 41.089.0 ± 44.787.9 ± 33.688.1 ± 41.5< 0.001 Comorbidities Hypertension64,250 (38.7)42,287 (38.3)8,204 (39.0)7,884 (40.7)5,875 (39.4)< 0.001Dyslipidemia50,422 (30.4)32,984 (29.9)6,496 (30.9)6,326 (32.7)4,616 (31.0)< 0.001Diabetes mellitus22,762 (13.7)14,623 (13.2)2,985 (14.2)2,981 (15.4)2,173 (14.6)< 0.001Chronic kidney disease10,948 (6.6)7,333 (6.6)1,355 (6.4)1,355 (7.0)905 (6.1)0.005Asthma or COPD7,885 (4.8)4,871 (4.4)1,204 (5.7)975 (5.0)835 (5.6)< 0.001Cancer57,243 (34.5)38,630 (35.0)7,120 (33.8)6,810 (35.1)4,683 (31.4)< 0.001 Amounts of exercise Energy expenditure,MET-min/wk624.1 ± 635.2347.0 ± 330.71,552.3 ± 556.7463.0 ± 356.31,618.9 ± 585.2< 0.001≥ 500 MET-min/wk80,102 (48.3)35,871 (32.5)20,599 (97.8)9,000 (46.5)14,632 (98.1)< 0.001Data are presented as number (percentage) or mean ± standard deviation*Low income denotes income in the lowest 20% of the entire Korean populationBP blood pressure, eGFR estimated glomerular filtration rate, COPD chronic obstructive pulmonary disease, MET metabolic equivalent of task Baseline characteristics of the study population Energy expenditure, MET-min/wk Data are presented as number (percentage) or mean ± standard deviation *Low income denotes income in the lowest 20% of the entire Korean population BP blood pressure, eGFR estimated glomerular filtration rate, COPD chronic obstructive pulmonary disease, MET metabolic equivalent of task [SUBTITLE] Effect of exercise on the risk of MI or stroke [SUBSECTION] During a mean follow-up duration of 6.2 ± 2.1 years, 4,233 (2.6%) and 3,745 (2.3%) patients with bronchiectasis experienced MI and stroke, respectively. The Kaplan-Meier curves showed a significantly lower incidence probability for MI in exercisers than in non-exercisers, regardless of changes in exercise habits. In contrast, the incidence probability for stroke was reduced in new exercisers and exercise maintainers, but not in exercise dropouts, compared with non-exercisers (Fig. 2). Fig. 2Kaplan-Meier curves of incidence probability for myocardial infarction or stroke according to change in exercise habits Kaplan-Meier curves of incidence probability for myocardial infarction or stroke according to change in exercise habits Table 2 reveals the effect of exercise on the risk of incident MI or stroke in patients with bronchiectasis. Performing an exercise was associated with a reduced risk of 18–28% in MI or of 9–28% in stroke (p < 0.001 for both). Compared to non-exercisers, exercise maintainers showed the lowest risk of MI (adjusted hazard ratio [aHR], 0.72; 95% confidence interval [CI], 0.64–0.81), followed by exercise dropouts (aHR, 0.79; 95% CI, 0.72–0.87) and new exercisers (aHR, 0.82; 95% CI, 0.75–0.91) (p < 0.001). Regarding stroke, exercise maintainers also showed the lowest risk (aHR, 0.72; 95% CI, 0.64–0.82), followed by new exercisers (aHR, 0.79; 95% CI, 0.71–0.88) and exercise dropouts (aHR, 0.91; 95% CI, 0.82–1.00) compared with non-exercisers. Table 2Effect of exercise on risk of myocardial infarction or strokeNo. of patientsMyocardial infarctionStrokeNo. ofeventsIR (/1,000 PY)aHR* (95% CI)No. ofeventsIR (/1,000 PY)aHR* (95% CI)OverallNon-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)ExercisersNew exercisers21,0564823.70.82 (0.75–0.91)4033.10.79 (0.71–0.88)Exercise dropouts19,3774443.70.79 (0.72–0.87)4473.70.91 (0.82–1.00)Exercise maintainers14,9102963.20.72 (0.64–0.81)2612.80.72 (0.64–0.82)P-value< 0.001< 0.001Age, years  < 40 yearsNon-exercisers7,215350.71 (Reference)140.31 (Reference)ExercisersNew exercisers95010.20.22 (0.03–1.62)10.20.56 (0.07–4.23)Exercise dropouts91771.11.53 (0.68–3.45)40.62.21 (0.73–6.71)Exercise maintainers53920.60.78 (0.19–3.26)20.61.94 (0.44–8.55)P-value0.2280.479  40–64 yearsNon-exercisers64,9071,3013.11 (Reference)8112.01 (Reference)ExercisersNew exercisers12,6252292.80.87 (0.76–1.00)1471.80.88 (0.74–1.05)Exercise dropouts10,9521832.60.79 (0.67–0.92)1372.00.94 (0.78–1.12)Exercise maintainers8,9241382.40.76 (0.64–0.90)681.20.58 (0.45–0.74)P-value< 0.001< 0.001  ≥ 65 yearsNon-exercisers38,3771,6757.51 (Reference)1,8098.11 (Reference)ExercisersNew exercisers7,4812525.70.79 (0.69–0.90)2555.80.75 (0.65–0.85)Exercise dropouts7,5082545.80.78 (0.69–0.90)3067.00.88 (0.78–1.00)Exercise maintainers5,4471564.90.69 (0.59–0.82)1916.00.79 (0.68–0.91)P-value< 0.001< 0.001P for interaction†0.4340.074Sex  MaleNon-exercisers50,7141,5504.91 (Reference)1,4304.61 (Reference)ExercisersNew exercisers10,8562954.40.84 (0.74–0.95)2553.80.78 (0.68–0.89)Exercise dropouts9,9352624.30.79 (0.69–0.90)2804.60.89 (0.79–1.02)Exercise maintainers8,8312033.70.73 (0.63–0.84)1973.60.75 (0.65–0.87)P-value< 0.001< 0.001  FemaleNon-exercisers59,7851,4613.91 (Reference)1,2043.21 (Reference)ExercisersNew exercisers10,2001872.90.79 (0.68–0.93)1482.30.82 (0.69–0.97)Exercise dropouts9,4421823.10.79 (0.68–0.92)1672.80.93 (0.79–1.09)Exercise maintainers6,079932.50.71 (0.58–0.88)641.70.65 (0.51–0.84)P-value< 0.0010.002P for interaction†0.9460.714*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included†The P for interaction was calculated by adding an interaction term to the multivariable modelIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, COPD chronic obstructive pulmonary disease Effect of exercise on risk of myocardial infarction or stroke *In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included †The P for interaction was calculated by adding an interaction term to the multivariable model IR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, COPD chronic obstructive pulmonary disease In stratified analysis, age or sex did not have a significant interaction with the association between exercise and cardiovascular outcomes. The risk-reducing effect of exercise for MI or stroke was only significant in those ≥ 40 years of age (p < 0.001 for both 40–64 years and ≥ 65 years). The effect of exercise was not significantly different between men and women, and exercisers of both genders showed a significant reduction in the risk of MI (p < 0.001 for both) or stroke (p < 0.001 for men; p = 0.002 for women) (Table 2). During a mean follow-up duration of 6.2 ± 2.1 years, 4,233 (2.6%) and 3,745 (2.3%) patients with bronchiectasis experienced MI and stroke, respectively. The Kaplan-Meier curves showed a significantly lower incidence probability for MI in exercisers than in non-exercisers, regardless of changes in exercise habits. In contrast, the incidence probability for stroke was reduced in new exercisers and exercise maintainers, but not in exercise dropouts, compared with non-exercisers (Fig. 2). Fig. 2Kaplan-Meier curves of incidence probability for myocardial infarction or stroke according to change in exercise habits Kaplan-Meier curves of incidence probability for myocardial infarction or stroke according to change in exercise habits Table 2 reveals the effect of exercise on the risk of incident MI or stroke in patients with bronchiectasis. Performing an exercise was associated with a reduced risk of 18–28% in MI or of 9–28% in stroke (p < 0.001 for both). Compared to non-exercisers, exercise maintainers showed the lowest risk of MI (adjusted hazard ratio [aHR], 0.72; 95% confidence interval [CI], 0.64–0.81), followed by exercise dropouts (aHR, 0.79; 95% CI, 0.72–0.87) and new exercisers (aHR, 0.82; 95% CI, 0.75–0.91) (p < 0.001). Regarding stroke, exercise maintainers also showed the lowest risk (aHR, 0.72; 95% CI, 0.64–0.82), followed by new exercisers (aHR, 0.79; 95% CI, 0.71–0.88) and exercise dropouts (aHR, 0.91; 95% CI, 0.82–1.00) compared with non-exercisers. Table 2Effect of exercise on risk of myocardial infarction or strokeNo. of patientsMyocardial infarctionStrokeNo. ofeventsIR (/1,000 PY)aHR* (95% CI)No. ofeventsIR (/1,000 PY)aHR* (95% CI)OverallNon-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)ExercisersNew exercisers21,0564823.70.82 (0.75–0.91)4033.10.79 (0.71–0.88)Exercise dropouts19,3774443.70.79 (0.72–0.87)4473.70.91 (0.82–1.00)Exercise maintainers14,9102963.20.72 (0.64–0.81)2612.80.72 (0.64–0.82)P-value< 0.001< 0.001Age, years  < 40 yearsNon-exercisers7,215350.71 (Reference)140.31 (Reference)ExercisersNew exercisers95010.20.22 (0.03–1.62)10.20.56 (0.07–4.23)Exercise dropouts91771.11.53 (0.68–3.45)40.62.21 (0.73–6.71)Exercise maintainers53920.60.78 (0.19–3.26)20.61.94 (0.44–8.55)P-value0.2280.479  40–64 yearsNon-exercisers64,9071,3013.11 (Reference)8112.01 (Reference)ExercisersNew exercisers12,6252292.80.87 (0.76–1.00)1471.80.88 (0.74–1.05)Exercise dropouts10,9521832.60.79 (0.67–0.92)1372.00.94 (0.78–1.12)Exercise maintainers8,9241382.40.76 (0.64–0.90)681.20.58 (0.45–0.74)P-value< 0.001< 0.001  ≥ 65 yearsNon-exercisers38,3771,6757.51 (Reference)1,8098.11 (Reference)ExercisersNew exercisers7,4812525.70.79 (0.69–0.90)2555.80.75 (0.65–0.85)Exercise dropouts7,5082545.80.78 (0.69–0.90)3067.00.88 (0.78–1.00)Exercise maintainers5,4471564.90.69 (0.59–0.82)1916.00.79 (0.68–0.91)P-value< 0.001< 0.001P for interaction†0.4340.074Sex  MaleNon-exercisers50,7141,5504.91 (Reference)1,4304.61 (Reference)ExercisersNew exercisers10,8562954.40.84 (0.74–0.95)2553.80.78 (0.68–0.89)Exercise dropouts9,9352624.30.79 (0.69–0.90)2804.60.89 (0.79–1.02)Exercise maintainers8,8312033.70.73 (0.63–0.84)1973.60.75 (0.65–0.87)P-value< 0.001< 0.001  FemaleNon-exercisers59,7851,4613.91 (Reference)1,2043.21 (Reference)ExercisersNew exercisers10,2001872.90.79 (0.68–0.93)1482.30.82 (0.69–0.97)Exercise dropouts9,4421823.10.79 (0.68–0.92)1672.80.93 (0.79–1.09)Exercise maintainers6,079932.50.71 (0.58–0.88)641.70.65 (0.51–0.84)P-value< 0.0010.002P for interaction†0.9460.714*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included†The P for interaction was calculated by adding an interaction term to the multivariable modelIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, COPD chronic obstructive pulmonary disease Effect of exercise on risk of myocardial infarction or stroke *In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included †The P for interaction was calculated by adding an interaction term to the multivariable model IR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, COPD chronic obstructive pulmonary disease In stratified analysis, age or sex did not have a significant interaction with the association between exercise and cardiovascular outcomes. The risk-reducing effect of exercise for MI or stroke was only significant in those ≥ 40 years of age (p < 0.001 for both 40–64 years and ≥ 65 years). The effect of exercise was not significantly different between men and women, and exercisers of both genders showed a significant reduction in the risk of MI (p < 0.001 for both) or stroke (p < 0.001 for men; p = 0.002 for women) (Table 2). [SUBTITLE] Amount of exercise and risk of MI or stroke [SUBSECTION] The associations between amount of exercise, measured by energy expenditure using METs, and risk of MI or stroke in patients with bronchiectasis are described in Table 3. Exercisers showed a lower risk for MI or stroke than non-exercisers regardless of the amount of exercise (p < 0.001 for both). However, a significant reduction was observed only in those with ≥ 500 MET-min/wk. Table 3The relationship between amount of exercise and risk of myocardial infarction or strokeNo. of patientsMyocardial infarctionStrokeNo. of eventsIR (/1,000 PY)aHR* (95% CI)No. of eventsIR (/1,000 PY)aHR* (95% CI)Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Overall exercisers  < 500 MET-min/wk11,1123024.30.91(0.81–1.03)2874.10.99(0.88–1.12)  500–999 MET-min/wk11,4072283.20.71(0.62–0.82)2223.10.80(0.69–0.91)  ≥ 1000 MET-min/wk32,8246923.40.76(0.70–0.83)6023.00.76(0.69–0.83)P-value< 0.001< 0.001Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)New exercisers  < 500 MET-min/wk457113.80.89(0.49–1.61)144.81.38(0.82–2.33)  500–999 MET-min/wk2,384583.80.92(0.71–1.19)352.30.66(0.48–0.93)  ≥ 1000 MET-min/wk18,2154133.60.81(0.73–0.90)3543.10.79(0.71–0.89)P-value< 0.001< 0.001Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Exercise dropouts  < 500 MET-min/wk10,3772844.40.91(0.81–1.03)2694.10.99(0.87–1.12)  500–999 MET-min/wk7,5201423.10.66(0.56–0.78)1583.40.83(0.71–0.98)  ≥ 1000 MET-min/wk1,480182.00.51(0.32–0.81)202.20.67(0.43–1.04)P-value< 0.0010.046Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Exercise maintainers  < 500 MET-min/wk27873.80.93(0.44–1.94)42.20.64(0.24–1.71)  500–999 MET-min/wk1,503282.90.68(0.47–0.98)293.10.82(0.57–1.18)  ≥ 1000 MET-min/wk13,1292613.20.72(0.64–0.82)2282.80.72(0.62–0.82)P-value< 0.001< 0.001*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were includedIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, MET metabolic equivalent of task, COPD chronic obstructive pulmonary disease The relationship between amount of exercise and risk of myocardial infarction or stroke 0.91 (0.81–1.03) 0.99 (0.88–1.12) 0.71 (0.62–0.82) 0.80 (0.69–0.91) 0.76 (0.70–0.83) 0.76 (0.69–0.83) 0.89 (0.49–1.61) 1.38 (0.82–2.33) 0.92 (0.71–1.19) 0.66 (0.48–0.93) 0.81 (0.73–0.90) 0.79 (0.71–0.89) 0.91 (0.81–1.03) 0.99 (0.87–1.12) 0.66 (0.56–0.78) 0.83 (0.71–0.98) 0.51 (0.32–0.81) 0.67 (0.43–1.04) 0.93 (0.44–1.94) 0.64 (0.24–1.71) 0.68 (0.47–0.98) 0.82 (0.57–1.18) 0.72 (0.64–0.82) 0.72 (0.62–0.82) *In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included IR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, MET metabolic equivalent of task, COPD chronic obstructive pulmonary disease Regarding MI, compared to non-exercisers, exercisers with 500–999 MET-min/wk showed the lowest risk (aHR, 0.71; 95% CI, 0.62–0.82), followed by exercisers with ≥ 1000 MET-min/wk (aHR, 0.76; 95% CI, 0.70–0.83) and exercisers with < 500 MET-min/wk (aHR, 0.91; 95% CI, 0.81–1.03). Subgroup analysis showed a significantly reduced risk for MI in new exercisers with ≥ 1000 MET-min/wk and in exercise dropouts and exercise maintainers with ≥ 500 MET-min/wk (all p < 0.001). The greatest risk reduction for MI was 49%, observed in exercise dropouts with initial ≥ 1000 MET-min/wk (Table 3). For stroke, compared to non-exercisers, exercisers with ≥ 1000 MET-min/wk showed the lowest risk (aHR, 0.76; 95% CI, 0.69–0.83), followed by exercisers with 500–999 MET-min/wk (aHR, 0.80; 95% CI, 0.69–0.91) and exercisers with < 500 MET-min/wk (aHR, 0.99; 95% CI, 0.88–1.12). In subgroup analysis, a significantly reduced risk for stroke was observed in new exercisers with ≥ 500 MET-min/wk (p < 0.001), in exercise dropouts with 500–999 MET-min/wk (p = 0.046), and in exercise maintainers with ≥ 1000 MET-min/wk (p < 0.001). The greatest risk reduction for stroke was 44%, observed in new exercisers with 500–999 MET-min/wk (Table 3). The associations between amount of exercise, measured by energy expenditure using METs, and risk of MI or stroke in patients with bronchiectasis are described in Table 3. Exercisers showed a lower risk for MI or stroke than non-exercisers regardless of the amount of exercise (p < 0.001 for both). However, a significant reduction was observed only in those with ≥ 500 MET-min/wk. Table 3The relationship between amount of exercise and risk of myocardial infarction or strokeNo. of patientsMyocardial infarctionStrokeNo. of eventsIR (/1,000 PY)aHR* (95% CI)No. of eventsIR (/1,000 PY)aHR* (95% CI)Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Overall exercisers  < 500 MET-min/wk11,1123024.30.91(0.81–1.03)2874.10.99(0.88–1.12)  500–999 MET-min/wk11,4072283.20.71(0.62–0.82)2223.10.80(0.69–0.91)  ≥ 1000 MET-min/wk32,8246923.40.76(0.70–0.83)6023.00.76(0.69–0.83)P-value< 0.001< 0.001Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)New exercisers  < 500 MET-min/wk457113.80.89(0.49–1.61)144.81.38(0.82–2.33)  500–999 MET-min/wk2,384583.80.92(0.71–1.19)352.30.66(0.48–0.93)  ≥ 1000 MET-min/wk18,2154133.60.81(0.73–0.90)3543.10.79(0.71–0.89)P-value< 0.001< 0.001Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Exercise dropouts  < 500 MET-min/wk10,3772844.40.91(0.81–1.03)2694.10.99(0.87–1.12)  500–999 MET-min/wk7,5201423.10.66(0.56–0.78)1583.40.83(0.71–0.98)  ≥ 1000 MET-min/wk1,480182.00.51(0.32–0.81)202.20.67(0.43–1.04)P-value< 0.0010.046Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Exercise maintainers  < 500 MET-min/wk27873.80.93(0.44–1.94)42.20.64(0.24–1.71)  500–999 MET-min/wk1,503282.90.68(0.47–0.98)293.10.82(0.57–1.18)  ≥ 1000 MET-min/wk13,1292613.20.72(0.64–0.82)2282.80.72(0.62–0.82)P-value< 0.001< 0.001*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were includedIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, MET metabolic equivalent of task, COPD chronic obstructive pulmonary disease The relationship between amount of exercise and risk of myocardial infarction or stroke 0.91 (0.81–1.03) 0.99 (0.88–1.12) 0.71 (0.62–0.82) 0.80 (0.69–0.91) 0.76 (0.70–0.83) 0.76 (0.69–0.83) 0.89 (0.49–1.61) 1.38 (0.82–2.33) 0.92 (0.71–1.19) 0.66 (0.48–0.93) 0.81 (0.73–0.90) 0.79 (0.71–0.89) 0.91 (0.81–1.03) 0.99 (0.87–1.12) 0.66 (0.56–0.78) 0.83 (0.71–0.98) 0.51 (0.32–0.81) 0.67 (0.43–1.04) 0.93 (0.44–1.94) 0.64 (0.24–1.71) 0.68 (0.47–0.98) 0.82 (0.57–1.18) 0.72 (0.64–0.82) 0.72 (0.62–0.82) *In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included IR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, MET metabolic equivalent of task, COPD chronic obstructive pulmonary disease Regarding MI, compared to non-exercisers, exercisers with 500–999 MET-min/wk showed the lowest risk (aHR, 0.71; 95% CI, 0.62–0.82), followed by exercisers with ≥ 1000 MET-min/wk (aHR, 0.76; 95% CI, 0.70–0.83) and exercisers with < 500 MET-min/wk (aHR, 0.91; 95% CI, 0.81–1.03). Subgroup analysis showed a significantly reduced risk for MI in new exercisers with ≥ 1000 MET-min/wk and in exercise dropouts and exercise maintainers with ≥ 500 MET-min/wk (all p < 0.001). The greatest risk reduction for MI was 49%, observed in exercise dropouts with initial ≥ 1000 MET-min/wk (Table 3). For stroke, compared to non-exercisers, exercisers with ≥ 1000 MET-min/wk showed the lowest risk (aHR, 0.76; 95% CI, 0.69–0.83), followed by exercisers with 500–999 MET-min/wk (aHR, 0.80; 95% CI, 0.69–0.91) and exercisers with < 500 MET-min/wk (aHR, 0.99; 95% CI, 0.88–1.12). In subgroup analysis, a significantly reduced risk for stroke was observed in new exercisers with ≥ 500 MET-min/wk (p < 0.001), in exercise dropouts with 500–999 MET-min/wk (p = 0.046), and in exercise maintainers with ≥ 1000 MET-min/wk (p < 0.001). The greatest risk reduction for stroke was 44%, observed in new exercisers with 500–999 MET-min/wk (Table 3).
Conclusion
Exercise was associated with reduced risk of cardiovascular diseases in patients with bronchiectasis. In particular, risk was lowest in exercise maintainers, and cardiovascular risk reduction was significant in those who exercised more than 500 MET-min/wk. Our study provides evidence of exercise prescription for patients with bronchiectasis to prevent cardiovascular diseases.
[ "Methods", "Data source and study population", "Exposure: change in exercise habit", "Outcomes: incidence of cardiovascular diseases", "Covariates", "Statistical analysis", "Baseline characteristics", "Effect of exercise on the risk of MI or stroke", "Amount of exercise and risk of MI or stroke", "" ]
[ "[SUBTITLE] Data source and study population [SUBSECTION] We used the National Health Insurance Service (NHIS) database of Korea, a nationwide population-based cohort [14]. The NHIS is a single-payer universal health system covering approximately 97% of the entire Korean population. In addition, the NHIS provides biennial health screening exams and examination reports on sociodemographic data, a self-questionnaire survey, clinical laboratory findings, inpatient and outpatient usage, prescription records, and recorded diagnosis based on the International Classification of Diseases 10th Revision (ICD-10) codes. More detailed information about the NHIS cohort was provided in previous studies [15, 16].\nAmong the 552,510 patients who were newly diagnosed with bronchiectasis (ICD-10 code J47) between 1 January 2010 and 31 December 2016, we included 204,235 who underwent two consecutive health examinations within two years before and after bronchiectasis diagnosis. We included those who underwent two consecutive health examinations to measure the changes in exercise habits between the two time points. Among the remaining 204,235, we excluded 4,584 with missing data, seven younger than 20 years, and 60 diagnosed with cystic fibrosis (ICD-10 code E84). Furthermore, 33,742 patients with a prior history of myocardial infarction (MI) or stroke before bronchiectasis diagnosis were excluded because this study focused on the de novo incidence of MI or stroke after bronchiectasis diagnosis. Finally, 165,842 patients with bronchiectasis were included in the final analytic cohort (Fig. 1).\n\nFig. 1Flow chart of the study population\n\nFlow chart of the study population\nWe used the National Health Insurance Service (NHIS) database of Korea, a nationwide population-based cohort [14]. The NHIS is a single-payer universal health system covering approximately 97% of the entire Korean population. In addition, the NHIS provides biennial health screening exams and examination reports on sociodemographic data, a self-questionnaire survey, clinical laboratory findings, inpatient and outpatient usage, prescription records, and recorded diagnosis based on the International Classification of Diseases 10th Revision (ICD-10) codes. More detailed information about the NHIS cohort was provided in previous studies [15, 16].\nAmong the 552,510 patients who were newly diagnosed with bronchiectasis (ICD-10 code J47) between 1 January 2010 and 31 December 2016, we included 204,235 who underwent two consecutive health examinations within two years before and after bronchiectasis diagnosis. We included those who underwent two consecutive health examinations to measure the changes in exercise habits between the two time points. Among the remaining 204,235, we excluded 4,584 with missing data, seven younger than 20 years, and 60 diagnosed with cystic fibrosis (ICD-10 code E84). Furthermore, 33,742 patients with a prior history of myocardial infarction (MI) or stroke before bronchiectasis diagnosis were excluded because this study focused on the de novo incidence of MI or stroke after bronchiectasis diagnosis. Finally, 165,842 patients with bronchiectasis were included in the final analytic cohort (Fig. 1).\n\nFig. 1Flow chart of the study population\n\nFlow chart of the study population\n[SUBTITLE] Exposure: change in exercise habit [SUBSECTION] To evaluate exercise habits, the intensity and frequency of physical activity were assessed using a self-report questionnaire, the International Physical Activity Questionnaire [17], during two health screening examinations performed before and after bronchiectasis diagnosis. This questionnaire has been used in several high-quality studies and contained the frequencies of weekly physical activity of varying intensities of light, moderate, or vigorous [18, 19]. Light-intensity physical activity was defined as to perform activities, such as walking slowly or vacuuming for more than 30 min. Moderate-intensity physical activity was defined as performing activities causing mild shortness of breath, such as brisk-pace walking, tennis doubles, or bicycling leisurely for more than 30 min. Vigorous-intensity physical activity was defined as performing activities causing greater shortness of breath than moderate-intensity physical activities, such as running, climbing, fast cycling, or aerobics for more than 20 min [20].\nExercise was defined as moderate- or vigorous-intensity physical activity at least once a week. To evaluate the effect of changes in exercise habits on the risk of cardiovascular diseases, the study population was subdivided into four groups according to changes in exercise performance: non-exercisers (never performed exercise in both examinations), new exercisers (non-exercisers in the first examination but exercisers in the second examination), exercise dropouts (exercisers in the first examination changed to non-exercisers in the second examination), and exercise maintainers (exercisers in both examinations). To further evaluate the overall effect of exercising habits on the risk of cardiovascular diseases compared to non-exercising habits, we grouped new exercisers, exercise dropouts, and exercise maintainers into exercisers and compared the risk of cardiovascular diseases between non-exercisers and exercisers.\nThe amount of exercise was also measured in this study. The energy expenditure, a minimum amount of energy consumption, was used to define the amount of exercise using metabolic equivalent of task (MET). The total MET-hours per week were calculated as the sum of the conventionally accepted intensity levels, (i.e., 2.9 METs for light-intensity exercise, 4.0 METs for moderate-intensity exercise, and 7.0 METs for vigorous-intensity exercise) [21]. For analysis, the amount of exercise was classified into < 500 MET-min/wk, 500–999 MET-min/wk, and ≥ 1000 MET-min/wk [21].\nTo evaluate exercise habits, the intensity and frequency of physical activity were assessed using a self-report questionnaire, the International Physical Activity Questionnaire [17], during two health screening examinations performed before and after bronchiectasis diagnosis. This questionnaire has been used in several high-quality studies and contained the frequencies of weekly physical activity of varying intensities of light, moderate, or vigorous [18, 19]. Light-intensity physical activity was defined as to perform activities, such as walking slowly or vacuuming for more than 30 min. Moderate-intensity physical activity was defined as performing activities causing mild shortness of breath, such as brisk-pace walking, tennis doubles, or bicycling leisurely for more than 30 min. Vigorous-intensity physical activity was defined as performing activities causing greater shortness of breath than moderate-intensity physical activities, such as running, climbing, fast cycling, or aerobics for more than 20 min [20].\nExercise was defined as moderate- or vigorous-intensity physical activity at least once a week. To evaluate the effect of changes in exercise habits on the risk of cardiovascular diseases, the study population was subdivided into four groups according to changes in exercise performance: non-exercisers (never performed exercise in both examinations), new exercisers (non-exercisers in the first examination but exercisers in the second examination), exercise dropouts (exercisers in the first examination changed to non-exercisers in the second examination), and exercise maintainers (exercisers in both examinations). To further evaluate the overall effect of exercising habits on the risk of cardiovascular diseases compared to non-exercising habits, we grouped new exercisers, exercise dropouts, and exercise maintainers into exercisers and compared the risk of cardiovascular diseases between non-exercisers and exercisers.\nThe amount of exercise was also measured in this study. The energy expenditure, a minimum amount of energy consumption, was used to define the amount of exercise using metabolic equivalent of task (MET). The total MET-hours per week were calculated as the sum of the conventionally accepted intensity levels, (i.e., 2.9 METs for light-intensity exercise, 4.0 METs for moderate-intensity exercise, and 7.0 METs for vigorous-intensity exercise) [21]. For analysis, the amount of exercise was classified into < 500 MET-min/wk, 500–999 MET-min/wk, and ≥ 1000 MET-min/wk [21].\n[SUBTITLE] Outcomes: incidence of cardiovascular diseases [SUBSECTION] The primary outcome was to compare the risk of incidence of cardiovascular diseases between non-exercisers and exercisers, including new exercisers, exercise dropouts, and exercise maintainers in patients with bronchiectasis. The secondary outcome was to evaluate the relationship between amount of exercise and risk of cardiovascular diseases.\nAs cardiovascular diseases, MI and stroke were defined using ICD-10 codes I21–I22 and I63–I64, respectively. The study population was followed until the date of the first occurrence of a cardiovascular event or the last follow-up date (31 December 2020), whichever came first (Additional file 1: Fig. S1).\nThe primary outcome was to compare the risk of incidence of cardiovascular diseases between non-exercisers and exercisers, including new exercisers, exercise dropouts, and exercise maintainers in patients with bronchiectasis. The secondary outcome was to evaluate the relationship between amount of exercise and risk of cardiovascular diseases.\nAs cardiovascular diseases, MI and stroke were defined using ICD-10 codes I21–I22 and I63–I64, respectively. The study population was followed until the date of the first occurrence of a cardiovascular event or the last follow-up date (31 December 2020), whichever came first (Additional file 1: Fig. S1).\n[SUBTITLE] Covariates [SUBSECTION] Data for age, sex, and income were collected from the Korean NHIS database. Income level was dichotomized at the lowest 20%; the medical aid program supports individuals with a low income. Smoking status and alcohol consumption were determined by self-questionnaire. Heavy alcohol consumption was defined as more than 30 g per day. Body mass index (BMI) was calculated by dividing the weight by the square of height [22]. Blood pressure was measured using a standard mercury sphygmomanometer. Waist circumference was measured at the narrowest point between the lower rib and the iliac crest (measured to the nearest 0.1 cm). Central obesity was defined as waist circumference ≥ 90 cm in men and ≥ 85 cm in women [23]. Blood samples were collected after fasting for at least eight hours, and serum levels of glucose, total cholesterol, and creatinine were measured. The estimated glomerular filtration rate (eGFR) was calculated using the chronic kidney disease (CKD)-epidemiology collaboration equation [24].\nComorbidities that can affect the incidence of cardiovascular diseases were defined using the following ICD-10 codes: hypertension (I10–I13, I15), dyslipidemia (E78), diabetes mellitus (E10–E14), CKD (N18.1–N18.5 and N18.9), asthma (J45–J46), chronic obstructive pulmonary disease (COPD) (J42–J44, except J43.0 [unilateral emphysema]), and cancer (C00–C99 and V193) [2, 6, 25, 26].\nData for age, sex, and income were collected from the Korean NHIS database. Income level was dichotomized at the lowest 20%; the medical aid program supports individuals with a low income. Smoking status and alcohol consumption were determined by self-questionnaire. Heavy alcohol consumption was defined as more than 30 g per day. Body mass index (BMI) was calculated by dividing the weight by the square of height [22]. Blood pressure was measured using a standard mercury sphygmomanometer. Waist circumference was measured at the narrowest point between the lower rib and the iliac crest (measured to the nearest 0.1 cm). Central obesity was defined as waist circumference ≥ 90 cm in men and ≥ 85 cm in women [23]. Blood samples were collected after fasting for at least eight hours, and serum levels of glucose, total cholesterol, and creatinine were measured. The estimated glomerular filtration rate (eGFR) was calculated using the chronic kidney disease (CKD)-epidemiology collaboration equation [24].\nComorbidities that can affect the incidence of cardiovascular diseases were defined using the following ICD-10 codes: hypertension (I10–I13, I15), dyslipidemia (E78), diabetes mellitus (E10–E14), CKD (N18.1–N18.5 and N18.9), asthma (J45–J46), chronic obstructive pulmonary disease (COPD) (J42–J44, except J43.0 [unilateral emphysema]), and cancer (C00–C99 and V193) [2, 6, 25, 26].\n[SUBTITLE] Statistical analysis [SUBSECTION] Data are expressed as mean ± standard deviation (SD) for continuous variables and as number (percentage) for categorical variables. Among the four groups, differences of baseline characteristics were confirmed using analysis of variance (ANOVA) for continuous variables and χ2 test for categorical variables. The incidence rate of cardiovascular diseases was calculated by dividing the number of incident cases by the total follow-up duration (1,000 person-years). Cox proportional hazards regression analyses were used to evaluate the associations of change and amount of exercise with incidence of MI and stroke. The multivariable model was fully adjusted for age, sex, low income, smoking status, alcohol consumption, BMI, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer. The proportional hazards assumptions were tested using graphical methods. Kaplan-Meier curves of incidence probability for incident MI or stroke were plotted according to changes in exercise habits. A two-sided p value < 0.05 was considered statistically significant. All analyses were conducted using SAS 9.3 (SAS Institute, Cary, NC, USA). \nData are expressed as mean ± standard deviation (SD) for continuous variables and as number (percentage) for categorical variables. Among the four groups, differences of baseline characteristics were confirmed using analysis of variance (ANOVA) for continuous variables and χ2 test for categorical variables. The incidence rate of cardiovascular diseases was calculated by dividing the number of incident cases by the total follow-up duration (1,000 person-years). Cox proportional hazards regression analyses were used to evaluate the associations of change and amount of exercise with incidence of MI and stroke. The multivariable model was fully adjusted for age, sex, low income, smoking status, alcohol consumption, BMI, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer. The proportional hazards assumptions were tested using graphical methods. Kaplan-Meier curves of incidence probability for incident MI or stroke were plotted according to changes in exercise habits. A two-sided p value < 0.05 was considered statistically significant. All analyses were conducted using SAS 9.3 (SAS Institute, Cary, NC, USA). ", "We used the National Health Insurance Service (NHIS) database of Korea, a nationwide population-based cohort [14]. The NHIS is a single-payer universal health system covering approximately 97% of the entire Korean population. In addition, the NHIS provides biennial health screening exams and examination reports on sociodemographic data, a self-questionnaire survey, clinical laboratory findings, inpatient and outpatient usage, prescription records, and recorded diagnosis based on the International Classification of Diseases 10th Revision (ICD-10) codes. More detailed information about the NHIS cohort was provided in previous studies [15, 16].\nAmong the 552,510 patients who were newly diagnosed with bronchiectasis (ICD-10 code J47) between 1 January 2010 and 31 December 2016, we included 204,235 who underwent two consecutive health examinations within two years before and after bronchiectasis diagnosis. We included those who underwent two consecutive health examinations to measure the changes in exercise habits between the two time points. Among the remaining 204,235, we excluded 4,584 with missing data, seven younger than 20 years, and 60 diagnosed with cystic fibrosis (ICD-10 code E84). Furthermore, 33,742 patients with a prior history of myocardial infarction (MI) or stroke before bronchiectasis diagnosis were excluded because this study focused on the de novo incidence of MI or stroke after bronchiectasis diagnosis. Finally, 165,842 patients with bronchiectasis were included in the final analytic cohort (Fig. 1).\n\nFig. 1Flow chart of the study population\n\nFlow chart of the study population", "To evaluate exercise habits, the intensity and frequency of physical activity were assessed using a self-report questionnaire, the International Physical Activity Questionnaire [17], during two health screening examinations performed before and after bronchiectasis diagnosis. This questionnaire has been used in several high-quality studies and contained the frequencies of weekly physical activity of varying intensities of light, moderate, or vigorous [18, 19]. Light-intensity physical activity was defined as to perform activities, such as walking slowly or vacuuming for more than 30 min. Moderate-intensity physical activity was defined as performing activities causing mild shortness of breath, such as brisk-pace walking, tennis doubles, or bicycling leisurely for more than 30 min. Vigorous-intensity physical activity was defined as performing activities causing greater shortness of breath than moderate-intensity physical activities, such as running, climbing, fast cycling, or aerobics for more than 20 min [20].\nExercise was defined as moderate- or vigorous-intensity physical activity at least once a week. To evaluate the effect of changes in exercise habits on the risk of cardiovascular diseases, the study population was subdivided into four groups according to changes in exercise performance: non-exercisers (never performed exercise in both examinations), new exercisers (non-exercisers in the first examination but exercisers in the second examination), exercise dropouts (exercisers in the first examination changed to non-exercisers in the second examination), and exercise maintainers (exercisers in both examinations). To further evaluate the overall effect of exercising habits on the risk of cardiovascular diseases compared to non-exercising habits, we grouped new exercisers, exercise dropouts, and exercise maintainers into exercisers and compared the risk of cardiovascular diseases between non-exercisers and exercisers.\nThe amount of exercise was also measured in this study. The energy expenditure, a minimum amount of energy consumption, was used to define the amount of exercise using metabolic equivalent of task (MET). The total MET-hours per week were calculated as the sum of the conventionally accepted intensity levels, (i.e., 2.9 METs for light-intensity exercise, 4.0 METs for moderate-intensity exercise, and 7.0 METs for vigorous-intensity exercise) [21]. For analysis, the amount of exercise was classified into < 500 MET-min/wk, 500–999 MET-min/wk, and ≥ 1000 MET-min/wk [21].", "The primary outcome was to compare the risk of incidence of cardiovascular diseases between non-exercisers and exercisers, including new exercisers, exercise dropouts, and exercise maintainers in patients with bronchiectasis. The secondary outcome was to evaluate the relationship between amount of exercise and risk of cardiovascular diseases.\nAs cardiovascular diseases, MI and stroke were defined using ICD-10 codes I21–I22 and I63–I64, respectively. The study population was followed until the date of the first occurrence of a cardiovascular event or the last follow-up date (31 December 2020), whichever came first (Additional file 1: Fig. S1).", "Data for age, sex, and income were collected from the Korean NHIS database. Income level was dichotomized at the lowest 20%; the medical aid program supports individuals with a low income. Smoking status and alcohol consumption were determined by self-questionnaire. Heavy alcohol consumption was defined as more than 30 g per day. Body mass index (BMI) was calculated by dividing the weight by the square of height [22]. Blood pressure was measured using a standard mercury sphygmomanometer. Waist circumference was measured at the narrowest point between the lower rib and the iliac crest (measured to the nearest 0.1 cm). Central obesity was defined as waist circumference ≥ 90 cm in men and ≥ 85 cm in women [23]. Blood samples were collected after fasting for at least eight hours, and serum levels of glucose, total cholesterol, and creatinine were measured. The estimated glomerular filtration rate (eGFR) was calculated using the chronic kidney disease (CKD)-epidemiology collaboration equation [24].\nComorbidities that can affect the incidence of cardiovascular diseases were defined using the following ICD-10 codes: hypertension (I10–I13, I15), dyslipidemia (E78), diabetes mellitus (E10–E14), CKD (N18.1–N18.5 and N18.9), asthma (J45–J46), chronic obstructive pulmonary disease (COPD) (J42–J44, except J43.0 [unilateral emphysema]), and cancer (C00–C99 and V193) [2, 6, 25, 26].", "Data are expressed as mean ± standard deviation (SD) for continuous variables and as number (percentage) for categorical variables. Among the four groups, differences of baseline characteristics were confirmed using analysis of variance (ANOVA) for continuous variables and χ2 test for categorical variables. The incidence rate of cardiovascular diseases was calculated by dividing the number of incident cases by the total follow-up duration (1,000 person-years). Cox proportional hazards regression analyses were used to evaluate the associations of change and amount of exercise with incidence of MI and stroke. The multivariable model was fully adjusted for age, sex, low income, smoking status, alcohol consumption, BMI, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer. The proportional hazards assumptions were tested using graphical methods. Kaplan-Meier curves of incidence probability for incident MI or stroke were plotted according to changes in exercise habits. A two-sided p value < 0.05 was considered statistically significant. All analyses were conducted using SAS 9.3 (SAS Institute, Cary, NC, USA). ", "In a total of 165,842 patients with bronchiectasis, the mean age was 59.8 years, and 48.4% were men. The study population comprised 66.6% non-exercisers and 33.4% exercisers. Moreover, exercisers were composed of new exercisers (n = 21,056, 12.7%), exercise dropouts (n = 19,377, 11.7%), and exercise maintainers (n = 14,910, 9.0%) according to changes in exercise habits (Fig. 1).\nExercisers were more likely to be older, male, non-smokers, and from low-income groups than non-exercisers (all p < 0.001). Regarding measurements in health screening exams, exercisers had lower total cholesterol levels, eGFR, and proportion of central obesity than non-exercisers (all p < 0.001); however, exercisers showed higher BMI, fasting blood sugar, systolic blood pressure, and longer waist circumference than non-exercisers (all p < 0.001). Of comorbidities, exercisers showed higher rates of hypertension, dyslipidemia, diabetes mellitus, and asthma or COPD compared with non-exercisers, and the differences between the four categories were statistically significant. Meanwhile, non-exercisers showed the lowest energy expenditure (32.5% for ≥ 500 MET-min/wk), followed by exercise dropouts (46.5%), new exercisers (97.8%), and exercise maintainers (98.1%) (p < 0.001) (Table 1).\n\nTable 1Baseline characteristics of the study populationVariablesTotal(N = 165,842)Non-exercisers(n = 110,499, 66.6%)Exercisers (n = 55,343, 33.4%)New exercisers(n = 21,056, 12.7%)Exercise dropouts(n = 19,377, 11.7%)Exercise maintainers(n = 14,910, 9.0%)P-value\nSociodemographics\nAge, years59.8 ± 11.959.5 ± 12.360.1 ± 11.160.8 ± 11.360.4 ± 10.6< 0.001  < 40 years9,621 (5.8)7,215 (6.5)950 (4.5)917 (4.7)539 (3.6)< 0.001  40–64 years97,408 (58.7)64,907 (58.8)12,625 (60.0)10,952 (56.5)8,924 (59.9)  ≥ 65 years58,813 (35.5)38,377 (34.7)7,481 (35.5)7,508 (38.8)5,447 (36.5)Sex< 0.001  Male80,336 (48.4)50,714 (45.9)10,856 (51.6)9,935 (51.3)8,831 (59.2)  Female85,506 (51.6)59,785 (54.1)10,200 (48.4)9,442 (48.7)6,079 (40.8)Low income*31,365 (18.9)21,210 (19.2)3,923 (18.6)3,674 (19.0)2,558 (17.2)< 0.001Smoking status  Current or past smoker24,294 (14.7)17,239 (15.6)2,739 (13.0)2,526 (13.0)1,790 (12.0)< 0.001Alcohol consumption  Heavy drinker8,402 (5.1)5,579 (5.1)1,093 (5.2)865 (4.5)865 (5.8)< 0.001\nMeasurements\nBody mass index, kg/m223.5 ± 3.323.5 ± 3.323.6 ± 3.123.7 ± 3.223.7 ± 3.0< 0.001Waist circumference, cm80.9 ± 9.180.8 ± 9.380.9 ± 8.981.3 ± 8.981.2 ± 8.6< 0.001Central obesity38,903 (23.5)26,626 (24.1)4,653 (22.1)4,514 (23.3)3,110 (20.9)< 0.001Fasting glucose, mg/dL100.3 ± 23.0100.0 ± 23.1100.4 ± 22.7101.0 ± 23.3101.1 ± 22.8< 0.001Systolic BP, mmHg123.3 ± 14.6123.1 ± 14.8123.4 ± 14.4123.8 ± 14.5123.7 ± 14.1< 0.001Diastolic BP, mmHg75.7 ± 9.575.7 ± 9.675.7 ± 9.475.9 ± 9.475.8 ± 9.40.057Total cholesterol, mg/dL193.7 ± 37.8194.2 ± 38.0192.7 ± 37.5193.1 ± 37.9192.1 ± 36.5< 0.001eGFR, ml/min/1.73m289.1 ± 40.789.5 ± 41.089.0 ± 44.787.9 ± 33.688.1 ± 41.5< 0.001\nComorbidities\nHypertension64,250 (38.7)42,287 (38.3)8,204 (39.0)7,884 (40.7)5,875 (39.4)< 0.001Dyslipidemia50,422 (30.4)32,984 (29.9)6,496 (30.9)6,326 (32.7)4,616 (31.0)< 0.001Diabetes mellitus22,762 (13.7)14,623 (13.2)2,985 (14.2)2,981 (15.4)2,173 (14.6)< 0.001Chronic kidney disease10,948 (6.6)7,333 (6.6)1,355 (6.4)1,355 (7.0)905 (6.1)0.005Asthma or COPD7,885 (4.8)4,871 (4.4)1,204 (5.7)975 (5.0)835 (5.6)< 0.001Cancer57,243 (34.5)38,630 (35.0)7,120 (33.8)6,810 (35.1)4,683 (31.4)< 0.001\nAmounts of exercise\nEnergy expenditure,MET-min/wk624.1 ± 635.2347.0 ± 330.71,552.3 ± 556.7463.0 ± 356.31,618.9 ± 585.2< 0.001≥ 500 MET-min/wk80,102 (48.3)35,871 (32.5)20,599 (97.8)9,000 (46.5)14,632 (98.1)< 0.001Data are presented as number (percentage) or mean ± standard deviation*Low income denotes income in the lowest 20% of the entire Korean populationBP blood pressure, eGFR estimated glomerular filtration rate, COPD chronic obstructive pulmonary disease, MET metabolic equivalent of task\n\nBaseline characteristics of the study population\nEnergy expenditure,\nMET-min/wk\nData are presented as number (percentage) or mean ± standard deviation\n*Low income denotes income in the lowest 20% of the entire Korean population\nBP blood pressure, eGFR estimated glomerular filtration rate, COPD chronic obstructive pulmonary disease, MET metabolic equivalent of task", "During a mean follow-up duration of 6.2 ± 2.1 years, 4,233 (2.6%) and 3,745 (2.3%) patients with bronchiectasis experienced MI and stroke, respectively. The Kaplan-Meier curves showed a significantly lower incidence probability for MI in exercisers than in non-exercisers, regardless of changes in exercise habits. In contrast, the incidence probability for stroke was reduced in new exercisers and exercise maintainers, but not in exercise dropouts, compared with non-exercisers (Fig. 2).\n\nFig. 2Kaplan-Meier curves of incidence probability for myocardial infarction or stroke according to change in exercise habits\n\nKaplan-Meier curves of incidence probability for myocardial infarction or stroke according to change in exercise habits\nTable 2 reveals the effect of exercise on the risk of incident MI or stroke in patients with bronchiectasis. Performing an exercise was associated with a reduced risk of 18–28% in MI or of 9–28% in stroke (p < 0.001 for both). Compared to non-exercisers, exercise maintainers showed the lowest risk of MI (adjusted hazard ratio [aHR], 0.72; 95% confidence interval [CI], 0.64–0.81), followed by exercise dropouts (aHR, 0.79; 95% CI, 0.72–0.87) and new exercisers (aHR, 0.82; 95% CI, 0.75–0.91) (p < 0.001). Regarding stroke, exercise maintainers also showed the lowest risk (aHR, 0.72; 95% CI, 0.64–0.82), followed by new exercisers (aHR, 0.79; 95% CI, 0.71–0.88) and exercise dropouts (aHR, 0.91; 95% CI, 0.82–1.00) compared with non-exercisers.\n\nTable 2Effect of exercise on risk of myocardial infarction or strokeNo. of patientsMyocardial infarctionStrokeNo. ofeventsIR (/1,000 PY)aHR* (95% CI)No. ofeventsIR (/1,000 PY)aHR* (95% CI)OverallNon-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)ExercisersNew exercisers21,0564823.70.82 (0.75–0.91)4033.10.79 (0.71–0.88)Exercise dropouts19,3774443.70.79 (0.72–0.87)4473.70.91 (0.82–1.00)Exercise maintainers14,9102963.20.72 (0.64–0.81)2612.80.72 (0.64–0.82)P-value< 0.001< 0.001Age, years  < 40 yearsNon-exercisers7,215350.71 (Reference)140.31 (Reference)ExercisersNew exercisers95010.20.22 (0.03–1.62)10.20.56 (0.07–4.23)Exercise dropouts91771.11.53 (0.68–3.45)40.62.21 (0.73–6.71)Exercise maintainers53920.60.78 (0.19–3.26)20.61.94 (0.44–8.55)P-value0.2280.479  40–64 yearsNon-exercisers64,9071,3013.11 (Reference)8112.01 (Reference)ExercisersNew exercisers12,6252292.80.87 (0.76–1.00)1471.80.88 (0.74–1.05)Exercise dropouts10,9521832.60.79 (0.67–0.92)1372.00.94 (0.78–1.12)Exercise maintainers8,9241382.40.76 (0.64–0.90)681.20.58 (0.45–0.74)P-value< 0.001< 0.001  ≥ 65 yearsNon-exercisers38,3771,6757.51 (Reference)1,8098.11 (Reference)ExercisersNew exercisers7,4812525.70.79 (0.69–0.90)2555.80.75 (0.65–0.85)Exercise dropouts7,5082545.80.78 (0.69–0.90)3067.00.88 (0.78–1.00)Exercise maintainers5,4471564.90.69 (0.59–0.82)1916.00.79 (0.68–0.91)P-value< 0.001< 0.001P for interaction†0.4340.074Sex  MaleNon-exercisers50,7141,5504.91 (Reference)1,4304.61 (Reference)ExercisersNew exercisers10,8562954.40.84 (0.74–0.95)2553.80.78 (0.68–0.89)Exercise dropouts9,9352624.30.79 (0.69–0.90)2804.60.89 (0.79–1.02)Exercise maintainers8,8312033.70.73 (0.63–0.84)1973.60.75 (0.65–0.87)P-value< 0.001< 0.001  FemaleNon-exercisers59,7851,4613.91 (Reference)1,2043.21 (Reference)ExercisersNew exercisers10,2001872.90.79 (0.68–0.93)1482.30.82 (0.69–0.97)Exercise dropouts9,4421823.10.79 (0.68–0.92)1672.80.93 (0.79–1.09)Exercise maintainers6,079932.50.71 (0.58–0.88)641.70.65 (0.51–0.84)P-value< 0.0010.002P for interaction†0.9460.714*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included†The P for interaction was calculated by adding an interaction term to the multivariable modelIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, COPD chronic obstructive pulmonary disease\n\nEffect of exercise on risk of myocardial infarction or stroke\n*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included\n†The P for interaction was calculated by adding an interaction term to the multivariable model\nIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, COPD chronic obstructive pulmonary disease\nIn stratified analysis, age or sex did not have a significant interaction with the association between exercise and cardiovascular outcomes. The risk-reducing effect of exercise for MI or stroke was only significant in those ≥ 40 years of age (p < 0.001 for both 40–64 years and ≥ 65 years). The effect of exercise was not significantly different between men and women, and exercisers of both genders showed a significant reduction in the risk of MI (p < 0.001 for both) or stroke (p < 0.001 for men; p = 0.002 for women) (Table 2).", "The associations between amount of exercise, measured by energy expenditure using METs, and risk of MI or stroke in patients with bronchiectasis are described in Table 3. Exercisers showed a lower risk for MI or stroke than non-exercisers regardless of the amount of exercise (p < 0.001 for both). However, a significant reduction was observed only in those with ≥ 500 MET-min/wk.\n\nTable 3The relationship between amount of exercise and risk of myocardial infarction or strokeNo. of patientsMyocardial infarctionStrokeNo. of eventsIR (/1,000 PY)aHR* (95% CI)No. of eventsIR (/1,000 PY)aHR* (95% CI)Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Overall exercisers  < 500 MET-min/wk11,1123024.30.91(0.81–1.03)2874.10.99(0.88–1.12)  500–999 MET-min/wk11,4072283.20.71(0.62–0.82)2223.10.80(0.69–0.91)  ≥ 1000 MET-min/wk32,8246923.40.76(0.70–0.83)6023.00.76(0.69–0.83)P-value< 0.001< 0.001Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)New exercisers  < 500 MET-min/wk457113.80.89(0.49–1.61)144.81.38(0.82–2.33)  500–999 MET-min/wk2,384583.80.92(0.71–1.19)352.30.66(0.48–0.93)  ≥ 1000 MET-min/wk18,2154133.60.81(0.73–0.90)3543.10.79(0.71–0.89)P-value< 0.001< 0.001Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Exercise dropouts  < 500 MET-min/wk10,3772844.40.91(0.81–1.03)2694.10.99(0.87–1.12)  500–999 MET-min/wk7,5201423.10.66(0.56–0.78)1583.40.83(0.71–0.98)  ≥ 1000 MET-min/wk1,480182.00.51(0.32–0.81)202.20.67(0.43–1.04)P-value< 0.0010.046Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Exercise maintainers  < 500 MET-min/wk27873.80.93(0.44–1.94)42.20.64(0.24–1.71)  500–999 MET-min/wk1,503282.90.68(0.47–0.98)293.10.82(0.57–1.18)  ≥ 1000 MET-min/wk13,1292613.20.72(0.64–0.82)2282.80.72(0.62–0.82)P-value< 0.001< 0.001*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were includedIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, MET metabolic equivalent of task, COPD chronic obstructive pulmonary disease\n\nThe relationship between amount of exercise and risk of myocardial infarction or stroke\n0.91\n(0.81–1.03)\n0.99\n(0.88–1.12)\n0.71\n(0.62–0.82)\n0.80\n(0.69–0.91)\n0.76\n(0.70–0.83)\n0.76\n(0.69–0.83)\n0.89\n(0.49–1.61)\n1.38\n(0.82–2.33)\n0.92\n(0.71–1.19)\n0.66\n(0.48–0.93)\n0.81\n(0.73–0.90)\n0.79\n(0.71–0.89)\n0.91\n(0.81–1.03)\n0.99\n(0.87–1.12)\n0.66\n(0.56–0.78)\n0.83\n(0.71–0.98)\n0.51\n(0.32–0.81)\n0.67\n(0.43–1.04)\n0.93\n(0.44–1.94)\n0.64\n(0.24–1.71)\n0.68\n(0.47–0.98)\n0.82\n(0.57–1.18)\n0.72\n(0.64–0.82)\n0.72\n(0.62–0.82)\n*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included\nIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, MET metabolic equivalent of task, COPD chronic obstructive pulmonary disease\nRegarding MI, compared to non-exercisers, exercisers with 500–999 MET-min/wk showed the lowest risk (aHR, 0.71; 95% CI, 0.62–0.82), followed by exercisers with ≥ 1000 MET-min/wk (aHR, 0.76; 95% CI, 0.70–0.83) and exercisers with < 500 MET-min/wk (aHR, 0.91; 95% CI, 0.81–1.03). Subgroup analysis showed a significantly reduced risk for MI in new exercisers with ≥ 1000 MET-min/wk and in exercise dropouts and exercise maintainers with ≥ 500 MET-min/wk (all p < 0.001). The greatest risk reduction for MI was 49%, observed in exercise dropouts with initial ≥ 1000 MET-min/wk (Table 3).\nFor stroke, compared to non-exercisers, exercisers with ≥ 1000 MET-min/wk showed the lowest risk (aHR, 0.76; 95% CI, 0.69–0.83), followed by exercisers with 500–999 MET-min/wk (aHR, 0.80; 95% CI, 0.69–0.91) and exercisers with < 500 MET-min/wk (aHR, 0.99; 95% CI, 0.88–1.12). In subgroup analysis, a significantly reduced risk for stroke was observed in new exercisers with ≥ 500 MET-min/wk (p < 0.001), in exercise dropouts with 500–999 MET-min/wk (p = 0.046), and in exercise maintainers with ≥ 1000 MET-min/wk (p < 0.001). The greatest risk reduction for stroke was 44%, observed in new exercisers with 500–999 MET-min/wk (Table 3).", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, null, null, null, null, null, null, null, null, null ]
[ "Introduction", "Methods", "Data source and study population", "Exposure: change in exercise habit", "Outcomes: incidence of cardiovascular diseases", "Covariates", "Statistical analysis", "Results", "Baseline characteristics", "Effect of exercise on the risk of MI or stroke", "Amount of exercise and risk of MI or stroke", "Discussion", "Conclusion", "Electronic supplementary material", "" ]
[ "The prevalence and disease burden of non-cystic fibrosis bronchiectasis (hereafter referred to as bronchiectasis) have been substantially increasing worldwide [1–3]. The prevalence is as high as 464–566 cases per 100,000 population [1, 2, 4, 5]. Furthermore, patients with bronchiectasis have a higher mortality risk than the general population (comparative mortality figures of 2.26 in women and 2.14 in men) [1] or than the age-, sex-, and comorbidity-matched patients without bronchiectasis (hazard ratio, 1.15) [6]. According to the literature, comorbid cardiovascular diseases largely explain higher mortality in patients with bronchiectasis [7, 8]. Hence, alleviating the risk of cardiovascular diseases can significantly reduce the long-term mortality of patients with bronchiectasis.\nDespite the importance of preventing cardiovascular diseases, there is a limited evidence to recommend appropriate prevention strategies for cardiovascular comorbidities in patients with bronchiectasis. Regular exercise and physical activity are known to be effective interventions for preventing cardiovascular diseases in various populations [9]. However, there have been no studies evaluating its protective effects against cardiovascular diseases in patients with bronchiectasis. Although the current international bronchiectasis guidelines recommend regular exercise and participation in a pulmonary rehabilitation program [10, 11], the main reasons for these recommendations are to improve exercise capacity and respiratory symptoms in patients with bronchiectasis [12, 13] and not to prevent their cardiovascular comorbidities. Additionally, there is limited evidence regarding the effects of exercise and physical activity on the long-term outcomes of bronchiectasis, particularly cardiovascular comorbidities, and the optimal amount of physical activity beneficial for cardiovascular outcomes in patients with bronchiectasis. From this view, evidence-based recommendations to guide optimal exercise methods would be invaluable in preventing cardiovascular diseases and possibly improving survival in patients with bronchiectasis.\nThis large population-based study aimed to investigate the effects of changing the amount of physical activity on cardiovascular outcomes in patients with bronchiectasis.", "[SUBTITLE] Data source and study population [SUBSECTION] We used the National Health Insurance Service (NHIS) database of Korea, a nationwide population-based cohort [14]. The NHIS is a single-payer universal health system covering approximately 97% of the entire Korean population. In addition, the NHIS provides biennial health screening exams and examination reports on sociodemographic data, a self-questionnaire survey, clinical laboratory findings, inpatient and outpatient usage, prescription records, and recorded diagnosis based on the International Classification of Diseases 10th Revision (ICD-10) codes. More detailed information about the NHIS cohort was provided in previous studies [15, 16].\nAmong the 552,510 patients who were newly diagnosed with bronchiectasis (ICD-10 code J47) between 1 January 2010 and 31 December 2016, we included 204,235 who underwent two consecutive health examinations within two years before and after bronchiectasis diagnosis. We included those who underwent two consecutive health examinations to measure the changes in exercise habits between the two time points. Among the remaining 204,235, we excluded 4,584 with missing data, seven younger than 20 years, and 60 diagnosed with cystic fibrosis (ICD-10 code E84). Furthermore, 33,742 patients with a prior history of myocardial infarction (MI) or stroke before bronchiectasis diagnosis were excluded because this study focused on the de novo incidence of MI or stroke after bronchiectasis diagnosis. Finally, 165,842 patients with bronchiectasis were included in the final analytic cohort (Fig. 1).\n\nFig. 1Flow chart of the study population\n\nFlow chart of the study population\nWe used the National Health Insurance Service (NHIS) database of Korea, a nationwide population-based cohort [14]. The NHIS is a single-payer universal health system covering approximately 97% of the entire Korean population. In addition, the NHIS provides biennial health screening exams and examination reports on sociodemographic data, a self-questionnaire survey, clinical laboratory findings, inpatient and outpatient usage, prescription records, and recorded diagnosis based on the International Classification of Diseases 10th Revision (ICD-10) codes. More detailed information about the NHIS cohort was provided in previous studies [15, 16].\nAmong the 552,510 patients who were newly diagnosed with bronchiectasis (ICD-10 code J47) between 1 January 2010 and 31 December 2016, we included 204,235 who underwent two consecutive health examinations within two years before and after bronchiectasis diagnosis. We included those who underwent two consecutive health examinations to measure the changes in exercise habits between the two time points. Among the remaining 204,235, we excluded 4,584 with missing data, seven younger than 20 years, and 60 diagnosed with cystic fibrosis (ICD-10 code E84). Furthermore, 33,742 patients with a prior history of myocardial infarction (MI) or stroke before bronchiectasis diagnosis were excluded because this study focused on the de novo incidence of MI or stroke after bronchiectasis diagnosis. Finally, 165,842 patients with bronchiectasis were included in the final analytic cohort (Fig. 1).\n\nFig. 1Flow chart of the study population\n\nFlow chart of the study population\n[SUBTITLE] Exposure: change in exercise habit [SUBSECTION] To evaluate exercise habits, the intensity and frequency of physical activity were assessed using a self-report questionnaire, the International Physical Activity Questionnaire [17], during two health screening examinations performed before and after bronchiectasis diagnosis. This questionnaire has been used in several high-quality studies and contained the frequencies of weekly physical activity of varying intensities of light, moderate, or vigorous [18, 19]. Light-intensity physical activity was defined as to perform activities, such as walking slowly or vacuuming for more than 30 min. Moderate-intensity physical activity was defined as performing activities causing mild shortness of breath, such as brisk-pace walking, tennis doubles, or bicycling leisurely for more than 30 min. Vigorous-intensity physical activity was defined as performing activities causing greater shortness of breath than moderate-intensity physical activities, such as running, climbing, fast cycling, or aerobics for more than 20 min [20].\nExercise was defined as moderate- or vigorous-intensity physical activity at least once a week. To evaluate the effect of changes in exercise habits on the risk of cardiovascular diseases, the study population was subdivided into four groups according to changes in exercise performance: non-exercisers (never performed exercise in both examinations), new exercisers (non-exercisers in the first examination but exercisers in the second examination), exercise dropouts (exercisers in the first examination changed to non-exercisers in the second examination), and exercise maintainers (exercisers in both examinations). To further evaluate the overall effect of exercising habits on the risk of cardiovascular diseases compared to non-exercising habits, we grouped new exercisers, exercise dropouts, and exercise maintainers into exercisers and compared the risk of cardiovascular diseases between non-exercisers and exercisers.\nThe amount of exercise was also measured in this study. The energy expenditure, a minimum amount of energy consumption, was used to define the amount of exercise using metabolic equivalent of task (MET). The total MET-hours per week were calculated as the sum of the conventionally accepted intensity levels, (i.e., 2.9 METs for light-intensity exercise, 4.0 METs for moderate-intensity exercise, and 7.0 METs for vigorous-intensity exercise) [21]. For analysis, the amount of exercise was classified into < 500 MET-min/wk, 500–999 MET-min/wk, and ≥ 1000 MET-min/wk [21].\nTo evaluate exercise habits, the intensity and frequency of physical activity were assessed using a self-report questionnaire, the International Physical Activity Questionnaire [17], during two health screening examinations performed before and after bronchiectasis diagnosis. This questionnaire has been used in several high-quality studies and contained the frequencies of weekly physical activity of varying intensities of light, moderate, or vigorous [18, 19]. Light-intensity physical activity was defined as to perform activities, such as walking slowly or vacuuming for more than 30 min. Moderate-intensity physical activity was defined as performing activities causing mild shortness of breath, such as brisk-pace walking, tennis doubles, or bicycling leisurely for more than 30 min. Vigorous-intensity physical activity was defined as performing activities causing greater shortness of breath than moderate-intensity physical activities, such as running, climbing, fast cycling, or aerobics for more than 20 min [20].\nExercise was defined as moderate- or vigorous-intensity physical activity at least once a week. To evaluate the effect of changes in exercise habits on the risk of cardiovascular diseases, the study population was subdivided into four groups according to changes in exercise performance: non-exercisers (never performed exercise in both examinations), new exercisers (non-exercisers in the first examination but exercisers in the second examination), exercise dropouts (exercisers in the first examination changed to non-exercisers in the second examination), and exercise maintainers (exercisers in both examinations). To further evaluate the overall effect of exercising habits on the risk of cardiovascular diseases compared to non-exercising habits, we grouped new exercisers, exercise dropouts, and exercise maintainers into exercisers and compared the risk of cardiovascular diseases between non-exercisers and exercisers.\nThe amount of exercise was also measured in this study. The energy expenditure, a minimum amount of energy consumption, was used to define the amount of exercise using metabolic equivalent of task (MET). The total MET-hours per week were calculated as the sum of the conventionally accepted intensity levels, (i.e., 2.9 METs for light-intensity exercise, 4.0 METs for moderate-intensity exercise, and 7.0 METs for vigorous-intensity exercise) [21]. For analysis, the amount of exercise was classified into < 500 MET-min/wk, 500–999 MET-min/wk, and ≥ 1000 MET-min/wk [21].\n[SUBTITLE] Outcomes: incidence of cardiovascular diseases [SUBSECTION] The primary outcome was to compare the risk of incidence of cardiovascular diseases between non-exercisers and exercisers, including new exercisers, exercise dropouts, and exercise maintainers in patients with bronchiectasis. The secondary outcome was to evaluate the relationship between amount of exercise and risk of cardiovascular diseases.\nAs cardiovascular diseases, MI and stroke were defined using ICD-10 codes I21–I22 and I63–I64, respectively. The study population was followed until the date of the first occurrence of a cardiovascular event or the last follow-up date (31 December 2020), whichever came first (Additional file 1: Fig. S1).\nThe primary outcome was to compare the risk of incidence of cardiovascular diseases between non-exercisers and exercisers, including new exercisers, exercise dropouts, and exercise maintainers in patients with bronchiectasis. The secondary outcome was to evaluate the relationship between amount of exercise and risk of cardiovascular diseases.\nAs cardiovascular diseases, MI and stroke were defined using ICD-10 codes I21–I22 and I63–I64, respectively. The study population was followed until the date of the first occurrence of a cardiovascular event or the last follow-up date (31 December 2020), whichever came first (Additional file 1: Fig. S1).\n[SUBTITLE] Covariates [SUBSECTION] Data for age, sex, and income were collected from the Korean NHIS database. Income level was dichotomized at the lowest 20%; the medical aid program supports individuals with a low income. Smoking status and alcohol consumption were determined by self-questionnaire. Heavy alcohol consumption was defined as more than 30 g per day. Body mass index (BMI) was calculated by dividing the weight by the square of height [22]. Blood pressure was measured using a standard mercury sphygmomanometer. Waist circumference was measured at the narrowest point between the lower rib and the iliac crest (measured to the nearest 0.1 cm). Central obesity was defined as waist circumference ≥ 90 cm in men and ≥ 85 cm in women [23]. Blood samples were collected after fasting for at least eight hours, and serum levels of glucose, total cholesterol, and creatinine were measured. The estimated glomerular filtration rate (eGFR) was calculated using the chronic kidney disease (CKD)-epidemiology collaboration equation [24].\nComorbidities that can affect the incidence of cardiovascular diseases were defined using the following ICD-10 codes: hypertension (I10–I13, I15), dyslipidemia (E78), diabetes mellitus (E10–E14), CKD (N18.1–N18.5 and N18.9), asthma (J45–J46), chronic obstructive pulmonary disease (COPD) (J42–J44, except J43.0 [unilateral emphysema]), and cancer (C00–C99 and V193) [2, 6, 25, 26].\nData for age, sex, and income were collected from the Korean NHIS database. Income level was dichotomized at the lowest 20%; the medical aid program supports individuals with a low income. Smoking status and alcohol consumption were determined by self-questionnaire. Heavy alcohol consumption was defined as more than 30 g per day. Body mass index (BMI) was calculated by dividing the weight by the square of height [22]. Blood pressure was measured using a standard mercury sphygmomanometer. Waist circumference was measured at the narrowest point between the lower rib and the iliac crest (measured to the nearest 0.1 cm). Central obesity was defined as waist circumference ≥ 90 cm in men and ≥ 85 cm in women [23]. Blood samples were collected after fasting for at least eight hours, and serum levels of glucose, total cholesterol, and creatinine were measured. The estimated glomerular filtration rate (eGFR) was calculated using the chronic kidney disease (CKD)-epidemiology collaboration equation [24].\nComorbidities that can affect the incidence of cardiovascular diseases were defined using the following ICD-10 codes: hypertension (I10–I13, I15), dyslipidemia (E78), diabetes mellitus (E10–E14), CKD (N18.1–N18.5 and N18.9), asthma (J45–J46), chronic obstructive pulmonary disease (COPD) (J42–J44, except J43.0 [unilateral emphysema]), and cancer (C00–C99 and V193) [2, 6, 25, 26].\n[SUBTITLE] Statistical analysis [SUBSECTION] Data are expressed as mean ± standard deviation (SD) for continuous variables and as number (percentage) for categorical variables. Among the four groups, differences of baseline characteristics were confirmed using analysis of variance (ANOVA) for continuous variables and χ2 test for categorical variables. The incidence rate of cardiovascular diseases was calculated by dividing the number of incident cases by the total follow-up duration (1,000 person-years). Cox proportional hazards regression analyses were used to evaluate the associations of change and amount of exercise with incidence of MI and stroke. The multivariable model was fully adjusted for age, sex, low income, smoking status, alcohol consumption, BMI, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer. The proportional hazards assumptions were tested using graphical methods. Kaplan-Meier curves of incidence probability for incident MI or stroke were plotted according to changes in exercise habits. A two-sided p value < 0.05 was considered statistically significant. All analyses were conducted using SAS 9.3 (SAS Institute, Cary, NC, USA). \nData are expressed as mean ± standard deviation (SD) for continuous variables and as number (percentage) for categorical variables. Among the four groups, differences of baseline characteristics were confirmed using analysis of variance (ANOVA) for continuous variables and χ2 test for categorical variables. The incidence rate of cardiovascular diseases was calculated by dividing the number of incident cases by the total follow-up duration (1,000 person-years). Cox proportional hazards regression analyses were used to evaluate the associations of change and amount of exercise with incidence of MI and stroke. The multivariable model was fully adjusted for age, sex, low income, smoking status, alcohol consumption, BMI, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer. The proportional hazards assumptions were tested using graphical methods. Kaplan-Meier curves of incidence probability for incident MI or stroke were plotted according to changes in exercise habits. A two-sided p value < 0.05 was considered statistically significant. All analyses were conducted using SAS 9.3 (SAS Institute, Cary, NC, USA). ", "We used the National Health Insurance Service (NHIS) database of Korea, a nationwide population-based cohort [14]. The NHIS is a single-payer universal health system covering approximately 97% of the entire Korean population. In addition, the NHIS provides biennial health screening exams and examination reports on sociodemographic data, a self-questionnaire survey, clinical laboratory findings, inpatient and outpatient usage, prescription records, and recorded diagnosis based on the International Classification of Diseases 10th Revision (ICD-10) codes. More detailed information about the NHIS cohort was provided in previous studies [15, 16].\nAmong the 552,510 patients who were newly diagnosed with bronchiectasis (ICD-10 code J47) between 1 January 2010 and 31 December 2016, we included 204,235 who underwent two consecutive health examinations within two years before and after bronchiectasis diagnosis. We included those who underwent two consecutive health examinations to measure the changes in exercise habits between the two time points. Among the remaining 204,235, we excluded 4,584 with missing data, seven younger than 20 years, and 60 diagnosed with cystic fibrosis (ICD-10 code E84). Furthermore, 33,742 patients with a prior history of myocardial infarction (MI) or stroke before bronchiectasis diagnosis were excluded because this study focused on the de novo incidence of MI or stroke after bronchiectasis diagnosis. Finally, 165,842 patients with bronchiectasis were included in the final analytic cohort (Fig. 1).\n\nFig. 1Flow chart of the study population\n\nFlow chart of the study population", "To evaluate exercise habits, the intensity and frequency of physical activity were assessed using a self-report questionnaire, the International Physical Activity Questionnaire [17], during two health screening examinations performed before and after bronchiectasis diagnosis. This questionnaire has been used in several high-quality studies and contained the frequencies of weekly physical activity of varying intensities of light, moderate, or vigorous [18, 19]. Light-intensity physical activity was defined as to perform activities, such as walking slowly or vacuuming for more than 30 min. Moderate-intensity physical activity was defined as performing activities causing mild shortness of breath, such as brisk-pace walking, tennis doubles, or bicycling leisurely for more than 30 min. Vigorous-intensity physical activity was defined as performing activities causing greater shortness of breath than moderate-intensity physical activities, such as running, climbing, fast cycling, or aerobics for more than 20 min [20].\nExercise was defined as moderate- or vigorous-intensity physical activity at least once a week. To evaluate the effect of changes in exercise habits on the risk of cardiovascular diseases, the study population was subdivided into four groups according to changes in exercise performance: non-exercisers (never performed exercise in both examinations), new exercisers (non-exercisers in the first examination but exercisers in the second examination), exercise dropouts (exercisers in the first examination changed to non-exercisers in the second examination), and exercise maintainers (exercisers in both examinations). To further evaluate the overall effect of exercising habits on the risk of cardiovascular diseases compared to non-exercising habits, we grouped new exercisers, exercise dropouts, and exercise maintainers into exercisers and compared the risk of cardiovascular diseases between non-exercisers and exercisers.\nThe amount of exercise was also measured in this study. The energy expenditure, a minimum amount of energy consumption, was used to define the amount of exercise using metabolic equivalent of task (MET). The total MET-hours per week were calculated as the sum of the conventionally accepted intensity levels, (i.e., 2.9 METs for light-intensity exercise, 4.0 METs for moderate-intensity exercise, and 7.0 METs for vigorous-intensity exercise) [21]. For analysis, the amount of exercise was classified into < 500 MET-min/wk, 500–999 MET-min/wk, and ≥ 1000 MET-min/wk [21].", "The primary outcome was to compare the risk of incidence of cardiovascular diseases between non-exercisers and exercisers, including new exercisers, exercise dropouts, and exercise maintainers in patients with bronchiectasis. The secondary outcome was to evaluate the relationship between amount of exercise and risk of cardiovascular diseases.\nAs cardiovascular diseases, MI and stroke were defined using ICD-10 codes I21–I22 and I63–I64, respectively. The study population was followed until the date of the first occurrence of a cardiovascular event or the last follow-up date (31 December 2020), whichever came first (Additional file 1: Fig. S1).", "Data for age, sex, and income were collected from the Korean NHIS database. Income level was dichotomized at the lowest 20%; the medical aid program supports individuals with a low income. Smoking status and alcohol consumption were determined by self-questionnaire. Heavy alcohol consumption was defined as more than 30 g per day. Body mass index (BMI) was calculated by dividing the weight by the square of height [22]. Blood pressure was measured using a standard mercury sphygmomanometer. Waist circumference was measured at the narrowest point between the lower rib and the iliac crest (measured to the nearest 0.1 cm). Central obesity was defined as waist circumference ≥ 90 cm in men and ≥ 85 cm in women [23]. Blood samples were collected after fasting for at least eight hours, and serum levels of glucose, total cholesterol, and creatinine were measured. The estimated glomerular filtration rate (eGFR) was calculated using the chronic kidney disease (CKD)-epidemiology collaboration equation [24].\nComorbidities that can affect the incidence of cardiovascular diseases were defined using the following ICD-10 codes: hypertension (I10–I13, I15), dyslipidemia (E78), diabetes mellitus (E10–E14), CKD (N18.1–N18.5 and N18.9), asthma (J45–J46), chronic obstructive pulmonary disease (COPD) (J42–J44, except J43.0 [unilateral emphysema]), and cancer (C00–C99 and V193) [2, 6, 25, 26].", "Data are expressed as mean ± standard deviation (SD) for continuous variables and as number (percentage) for categorical variables. Among the four groups, differences of baseline characteristics were confirmed using analysis of variance (ANOVA) for continuous variables and χ2 test for categorical variables. The incidence rate of cardiovascular diseases was calculated by dividing the number of incident cases by the total follow-up duration (1,000 person-years). Cox proportional hazards regression analyses were used to evaluate the associations of change and amount of exercise with incidence of MI and stroke. The multivariable model was fully adjusted for age, sex, low income, smoking status, alcohol consumption, BMI, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer. The proportional hazards assumptions were tested using graphical methods. Kaplan-Meier curves of incidence probability for incident MI or stroke were plotted according to changes in exercise habits. A two-sided p value < 0.05 was considered statistically significant. All analyses were conducted using SAS 9.3 (SAS Institute, Cary, NC, USA). ", "[SUBTITLE] Baseline characteristics [SUBSECTION] In a total of 165,842 patients with bronchiectasis, the mean age was 59.8 years, and 48.4% were men. The study population comprised 66.6% non-exercisers and 33.4% exercisers. Moreover, exercisers were composed of new exercisers (n = 21,056, 12.7%), exercise dropouts (n = 19,377, 11.7%), and exercise maintainers (n = 14,910, 9.0%) according to changes in exercise habits (Fig. 1).\nExercisers were more likely to be older, male, non-smokers, and from low-income groups than non-exercisers (all p < 0.001). Regarding measurements in health screening exams, exercisers had lower total cholesterol levels, eGFR, and proportion of central obesity than non-exercisers (all p < 0.001); however, exercisers showed higher BMI, fasting blood sugar, systolic blood pressure, and longer waist circumference than non-exercisers (all p < 0.001). Of comorbidities, exercisers showed higher rates of hypertension, dyslipidemia, diabetes mellitus, and asthma or COPD compared with non-exercisers, and the differences between the four categories were statistically significant. Meanwhile, non-exercisers showed the lowest energy expenditure (32.5% for ≥ 500 MET-min/wk), followed by exercise dropouts (46.5%), new exercisers (97.8%), and exercise maintainers (98.1%) (p < 0.001) (Table 1).\n\nTable 1Baseline characteristics of the study populationVariablesTotal(N = 165,842)Non-exercisers(n = 110,499, 66.6%)Exercisers (n = 55,343, 33.4%)New exercisers(n = 21,056, 12.7%)Exercise dropouts(n = 19,377, 11.7%)Exercise maintainers(n = 14,910, 9.0%)P-value\nSociodemographics\nAge, years59.8 ± 11.959.5 ± 12.360.1 ± 11.160.8 ± 11.360.4 ± 10.6< 0.001  < 40 years9,621 (5.8)7,215 (6.5)950 (4.5)917 (4.7)539 (3.6)< 0.001  40–64 years97,408 (58.7)64,907 (58.8)12,625 (60.0)10,952 (56.5)8,924 (59.9)  ≥ 65 years58,813 (35.5)38,377 (34.7)7,481 (35.5)7,508 (38.8)5,447 (36.5)Sex< 0.001  Male80,336 (48.4)50,714 (45.9)10,856 (51.6)9,935 (51.3)8,831 (59.2)  Female85,506 (51.6)59,785 (54.1)10,200 (48.4)9,442 (48.7)6,079 (40.8)Low income*31,365 (18.9)21,210 (19.2)3,923 (18.6)3,674 (19.0)2,558 (17.2)< 0.001Smoking status  Current or past smoker24,294 (14.7)17,239 (15.6)2,739 (13.0)2,526 (13.0)1,790 (12.0)< 0.001Alcohol consumption  Heavy drinker8,402 (5.1)5,579 (5.1)1,093 (5.2)865 (4.5)865 (5.8)< 0.001\nMeasurements\nBody mass index, kg/m223.5 ± 3.323.5 ± 3.323.6 ± 3.123.7 ± 3.223.7 ± 3.0< 0.001Waist circumference, cm80.9 ± 9.180.8 ± 9.380.9 ± 8.981.3 ± 8.981.2 ± 8.6< 0.001Central obesity38,903 (23.5)26,626 (24.1)4,653 (22.1)4,514 (23.3)3,110 (20.9)< 0.001Fasting glucose, mg/dL100.3 ± 23.0100.0 ± 23.1100.4 ± 22.7101.0 ± 23.3101.1 ± 22.8< 0.001Systolic BP, mmHg123.3 ± 14.6123.1 ± 14.8123.4 ± 14.4123.8 ± 14.5123.7 ± 14.1< 0.001Diastolic BP, mmHg75.7 ± 9.575.7 ± 9.675.7 ± 9.475.9 ± 9.475.8 ± 9.40.057Total cholesterol, mg/dL193.7 ± 37.8194.2 ± 38.0192.7 ± 37.5193.1 ± 37.9192.1 ± 36.5< 0.001eGFR, ml/min/1.73m289.1 ± 40.789.5 ± 41.089.0 ± 44.787.9 ± 33.688.1 ± 41.5< 0.001\nComorbidities\nHypertension64,250 (38.7)42,287 (38.3)8,204 (39.0)7,884 (40.7)5,875 (39.4)< 0.001Dyslipidemia50,422 (30.4)32,984 (29.9)6,496 (30.9)6,326 (32.7)4,616 (31.0)< 0.001Diabetes mellitus22,762 (13.7)14,623 (13.2)2,985 (14.2)2,981 (15.4)2,173 (14.6)< 0.001Chronic kidney disease10,948 (6.6)7,333 (6.6)1,355 (6.4)1,355 (7.0)905 (6.1)0.005Asthma or COPD7,885 (4.8)4,871 (4.4)1,204 (5.7)975 (5.0)835 (5.6)< 0.001Cancer57,243 (34.5)38,630 (35.0)7,120 (33.8)6,810 (35.1)4,683 (31.4)< 0.001\nAmounts of exercise\nEnergy expenditure,MET-min/wk624.1 ± 635.2347.0 ± 330.71,552.3 ± 556.7463.0 ± 356.31,618.9 ± 585.2< 0.001≥ 500 MET-min/wk80,102 (48.3)35,871 (32.5)20,599 (97.8)9,000 (46.5)14,632 (98.1)< 0.001Data are presented as number (percentage) or mean ± standard deviation*Low income denotes income in the lowest 20% of the entire Korean populationBP blood pressure, eGFR estimated glomerular filtration rate, COPD chronic obstructive pulmonary disease, MET metabolic equivalent of task\n\nBaseline characteristics of the study population\nEnergy expenditure,\nMET-min/wk\nData are presented as number (percentage) or mean ± standard deviation\n*Low income denotes income in the lowest 20% of the entire Korean population\nBP blood pressure, eGFR estimated glomerular filtration rate, COPD chronic obstructive pulmonary disease, MET metabolic equivalent of task\nIn a total of 165,842 patients with bronchiectasis, the mean age was 59.8 years, and 48.4% were men. The study population comprised 66.6% non-exercisers and 33.4% exercisers. Moreover, exercisers were composed of new exercisers (n = 21,056, 12.7%), exercise dropouts (n = 19,377, 11.7%), and exercise maintainers (n = 14,910, 9.0%) according to changes in exercise habits (Fig. 1).\nExercisers were more likely to be older, male, non-smokers, and from low-income groups than non-exercisers (all p < 0.001). Regarding measurements in health screening exams, exercisers had lower total cholesterol levels, eGFR, and proportion of central obesity than non-exercisers (all p < 0.001); however, exercisers showed higher BMI, fasting blood sugar, systolic blood pressure, and longer waist circumference than non-exercisers (all p < 0.001). Of comorbidities, exercisers showed higher rates of hypertension, dyslipidemia, diabetes mellitus, and asthma or COPD compared with non-exercisers, and the differences between the four categories were statistically significant. Meanwhile, non-exercisers showed the lowest energy expenditure (32.5% for ≥ 500 MET-min/wk), followed by exercise dropouts (46.5%), new exercisers (97.8%), and exercise maintainers (98.1%) (p < 0.001) (Table 1).\n\nTable 1Baseline characteristics of the study populationVariablesTotal(N = 165,842)Non-exercisers(n = 110,499, 66.6%)Exercisers (n = 55,343, 33.4%)New exercisers(n = 21,056, 12.7%)Exercise dropouts(n = 19,377, 11.7%)Exercise maintainers(n = 14,910, 9.0%)P-value\nSociodemographics\nAge, years59.8 ± 11.959.5 ± 12.360.1 ± 11.160.8 ± 11.360.4 ± 10.6< 0.001  < 40 years9,621 (5.8)7,215 (6.5)950 (4.5)917 (4.7)539 (3.6)< 0.001  40–64 years97,408 (58.7)64,907 (58.8)12,625 (60.0)10,952 (56.5)8,924 (59.9)  ≥ 65 years58,813 (35.5)38,377 (34.7)7,481 (35.5)7,508 (38.8)5,447 (36.5)Sex< 0.001  Male80,336 (48.4)50,714 (45.9)10,856 (51.6)9,935 (51.3)8,831 (59.2)  Female85,506 (51.6)59,785 (54.1)10,200 (48.4)9,442 (48.7)6,079 (40.8)Low income*31,365 (18.9)21,210 (19.2)3,923 (18.6)3,674 (19.0)2,558 (17.2)< 0.001Smoking status  Current or past smoker24,294 (14.7)17,239 (15.6)2,739 (13.0)2,526 (13.0)1,790 (12.0)< 0.001Alcohol consumption  Heavy drinker8,402 (5.1)5,579 (5.1)1,093 (5.2)865 (4.5)865 (5.8)< 0.001\nMeasurements\nBody mass index, kg/m223.5 ± 3.323.5 ± 3.323.6 ± 3.123.7 ± 3.223.7 ± 3.0< 0.001Waist circumference, cm80.9 ± 9.180.8 ± 9.380.9 ± 8.981.3 ± 8.981.2 ± 8.6< 0.001Central obesity38,903 (23.5)26,626 (24.1)4,653 (22.1)4,514 (23.3)3,110 (20.9)< 0.001Fasting glucose, mg/dL100.3 ± 23.0100.0 ± 23.1100.4 ± 22.7101.0 ± 23.3101.1 ± 22.8< 0.001Systolic BP, mmHg123.3 ± 14.6123.1 ± 14.8123.4 ± 14.4123.8 ± 14.5123.7 ± 14.1< 0.001Diastolic BP, mmHg75.7 ± 9.575.7 ± 9.675.7 ± 9.475.9 ± 9.475.8 ± 9.40.057Total cholesterol, mg/dL193.7 ± 37.8194.2 ± 38.0192.7 ± 37.5193.1 ± 37.9192.1 ± 36.5< 0.001eGFR, ml/min/1.73m289.1 ± 40.789.5 ± 41.089.0 ± 44.787.9 ± 33.688.1 ± 41.5< 0.001\nComorbidities\nHypertension64,250 (38.7)42,287 (38.3)8,204 (39.0)7,884 (40.7)5,875 (39.4)< 0.001Dyslipidemia50,422 (30.4)32,984 (29.9)6,496 (30.9)6,326 (32.7)4,616 (31.0)< 0.001Diabetes mellitus22,762 (13.7)14,623 (13.2)2,985 (14.2)2,981 (15.4)2,173 (14.6)< 0.001Chronic kidney disease10,948 (6.6)7,333 (6.6)1,355 (6.4)1,355 (7.0)905 (6.1)0.005Asthma or COPD7,885 (4.8)4,871 (4.4)1,204 (5.7)975 (5.0)835 (5.6)< 0.001Cancer57,243 (34.5)38,630 (35.0)7,120 (33.8)6,810 (35.1)4,683 (31.4)< 0.001\nAmounts of exercise\nEnergy expenditure,MET-min/wk624.1 ± 635.2347.0 ± 330.71,552.3 ± 556.7463.0 ± 356.31,618.9 ± 585.2< 0.001≥ 500 MET-min/wk80,102 (48.3)35,871 (32.5)20,599 (97.8)9,000 (46.5)14,632 (98.1)< 0.001Data are presented as number (percentage) or mean ± standard deviation*Low income denotes income in the lowest 20% of the entire Korean populationBP blood pressure, eGFR estimated glomerular filtration rate, COPD chronic obstructive pulmonary disease, MET metabolic equivalent of task\n\nBaseline characteristics of the study population\nEnergy expenditure,\nMET-min/wk\nData are presented as number (percentage) or mean ± standard deviation\n*Low income denotes income in the lowest 20% of the entire Korean population\nBP blood pressure, eGFR estimated glomerular filtration rate, COPD chronic obstructive pulmonary disease, MET metabolic equivalent of task\n[SUBTITLE] Effect of exercise on the risk of MI or stroke [SUBSECTION] During a mean follow-up duration of 6.2 ± 2.1 years, 4,233 (2.6%) and 3,745 (2.3%) patients with bronchiectasis experienced MI and stroke, respectively. The Kaplan-Meier curves showed a significantly lower incidence probability for MI in exercisers than in non-exercisers, regardless of changes in exercise habits. In contrast, the incidence probability for stroke was reduced in new exercisers and exercise maintainers, but not in exercise dropouts, compared with non-exercisers (Fig. 2).\n\nFig. 2Kaplan-Meier curves of incidence probability for myocardial infarction or stroke according to change in exercise habits\n\nKaplan-Meier curves of incidence probability for myocardial infarction or stroke according to change in exercise habits\nTable 2 reveals the effect of exercise on the risk of incident MI or stroke in patients with bronchiectasis. Performing an exercise was associated with a reduced risk of 18–28% in MI or of 9–28% in stroke (p < 0.001 for both). Compared to non-exercisers, exercise maintainers showed the lowest risk of MI (adjusted hazard ratio [aHR], 0.72; 95% confidence interval [CI], 0.64–0.81), followed by exercise dropouts (aHR, 0.79; 95% CI, 0.72–0.87) and new exercisers (aHR, 0.82; 95% CI, 0.75–0.91) (p < 0.001). Regarding stroke, exercise maintainers also showed the lowest risk (aHR, 0.72; 95% CI, 0.64–0.82), followed by new exercisers (aHR, 0.79; 95% CI, 0.71–0.88) and exercise dropouts (aHR, 0.91; 95% CI, 0.82–1.00) compared with non-exercisers.\n\nTable 2Effect of exercise on risk of myocardial infarction or strokeNo. of patientsMyocardial infarctionStrokeNo. ofeventsIR (/1,000 PY)aHR* (95% CI)No. ofeventsIR (/1,000 PY)aHR* (95% CI)OverallNon-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)ExercisersNew exercisers21,0564823.70.82 (0.75–0.91)4033.10.79 (0.71–0.88)Exercise dropouts19,3774443.70.79 (0.72–0.87)4473.70.91 (0.82–1.00)Exercise maintainers14,9102963.20.72 (0.64–0.81)2612.80.72 (0.64–0.82)P-value< 0.001< 0.001Age, years  < 40 yearsNon-exercisers7,215350.71 (Reference)140.31 (Reference)ExercisersNew exercisers95010.20.22 (0.03–1.62)10.20.56 (0.07–4.23)Exercise dropouts91771.11.53 (0.68–3.45)40.62.21 (0.73–6.71)Exercise maintainers53920.60.78 (0.19–3.26)20.61.94 (0.44–8.55)P-value0.2280.479  40–64 yearsNon-exercisers64,9071,3013.11 (Reference)8112.01 (Reference)ExercisersNew exercisers12,6252292.80.87 (0.76–1.00)1471.80.88 (0.74–1.05)Exercise dropouts10,9521832.60.79 (0.67–0.92)1372.00.94 (0.78–1.12)Exercise maintainers8,9241382.40.76 (0.64–0.90)681.20.58 (0.45–0.74)P-value< 0.001< 0.001  ≥ 65 yearsNon-exercisers38,3771,6757.51 (Reference)1,8098.11 (Reference)ExercisersNew exercisers7,4812525.70.79 (0.69–0.90)2555.80.75 (0.65–0.85)Exercise dropouts7,5082545.80.78 (0.69–0.90)3067.00.88 (0.78–1.00)Exercise maintainers5,4471564.90.69 (0.59–0.82)1916.00.79 (0.68–0.91)P-value< 0.001< 0.001P for interaction†0.4340.074Sex  MaleNon-exercisers50,7141,5504.91 (Reference)1,4304.61 (Reference)ExercisersNew exercisers10,8562954.40.84 (0.74–0.95)2553.80.78 (0.68–0.89)Exercise dropouts9,9352624.30.79 (0.69–0.90)2804.60.89 (0.79–1.02)Exercise maintainers8,8312033.70.73 (0.63–0.84)1973.60.75 (0.65–0.87)P-value< 0.001< 0.001  FemaleNon-exercisers59,7851,4613.91 (Reference)1,2043.21 (Reference)ExercisersNew exercisers10,2001872.90.79 (0.68–0.93)1482.30.82 (0.69–0.97)Exercise dropouts9,4421823.10.79 (0.68–0.92)1672.80.93 (0.79–1.09)Exercise maintainers6,079932.50.71 (0.58–0.88)641.70.65 (0.51–0.84)P-value< 0.0010.002P for interaction†0.9460.714*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included†The P for interaction was calculated by adding an interaction term to the multivariable modelIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, COPD chronic obstructive pulmonary disease\n\nEffect of exercise on risk of myocardial infarction or stroke\n*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included\n†The P for interaction was calculated by adding an interaction term to the multivariable model\nIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, COPD chronic obstructive pulmonary disease\nIn stratified analysis, age or sex did not have a significant interaction with the association between exercise and cardiovascular outcomes. The risk-reducing effect of exercise for MI or stroke was only significant in those ≥ 40 years of age (p < 0.001 for both 40–64 years and ≥ 65 years). The effect of exercise was not significantly different between men and women, and exercisers of both genders showed a significant reduction in the risk of MI (p < 0.001 for both) or stroke (p < 0.001 for men; p = 0.002 for women) (Table 2).\nDuring a mean follow-up duration of 6.2 ± 2.1 years, 4,233 (2.6%) and 3,745 (2.3%) patients with bronchiectasis experienced MI and stroke, respectively. The Kaplan-Meier curves showed a significantly lower incidence probability for MI in exercisers than in non-exercisers, regardless of changes in exercise habits. In contrast, the incidence probability for stroke was reduced in new exercisers and exercise maintainers, but not in exercise dropouts, compared with non-exercisers (Fig. 2).\n\nFig. 2Kaplan-Meier curves of incidence probability for myocardial infarction or stroke according to change in exercise habits\n\nKaplan-Meier curves of incidence probability for myocardial infarction or stroke according to change in exercise habits\nTable 2 reveals the effect of exercise on the risk of incident MI or stroke in patients with bronchiectasis. Performing an exercise was associated with a reduced risk of 18–28% in MI or of 9–28% in stroke (p < 0.001 for both). Compared to non-exercisers, exercise maintainers showed the lowest risk of MI (adjusted hazard ratio [aHR], 0.72; 95% confidence interval [CI], 0.64–0.81), followed by exercise dropouts (aHR, 0.79; 95% CI, 0.72–0.87) and new exercisers (aHR, 0.82; 95% CI, 0.75–0.91) (p < 0.001). Regarding stroke, exercise maintainers also showed the lowest risk (aHR, 0.72; 95% CI, 0.64–0.82), followed by new exercisers (aHR, 0.79; 95% CI, 0.71–0.88) and exercise dropouts (aHR, 0.91; 95% CI, 0.82–1.00) compared with non-exercisers.\n\nTable 2Effect of exercise on risk of myocardial infarction or strokeNo. of patientsMyocardial infarctionStrokeNo. ofeventsIR (/1,000 PY)aHR* (95% CI)No. ofeventsIR (/1,000 PY)aHR* (95% CI)OverallNon-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)ExercisersNew exercisers21,0564823.70.82 (0.75–0.91)4033.10.79 (0.71–0.88)Exercise dropouts19,3774443.70.79 (0.72–0.87)4473.70.91 (0.82–1.00)Exercise maintainers14,9102963.20.72 (0.64–0.81)2612.80.72 (0.64–0.82)P-value< 0.001< 0.001Age, years  < 40 yearsNon-exercisers7,215350.71 (Reference)140.31 (Reference)ExercisersNew exercisers95010.20.22 (0.03–1.62)10.20.56 (0.07–4.23)Exercise dropouts91771.11.53 (0.68–3.45)40.62.21 (0.73–6.71)Exercise maintainers53920.60.78 (0.19–3.26)20.61.94 (0.44–8.55)P-value0.2280.479  40–64 yearsNon-exercisers64,9071,3013.11 (Reference)8112.01 (Reference)ExercisersNew exercisers12,6252292.80.87 (0.76–1.00)1471.80.88 (0.74–1.05)Exercise dropouts10,9521832.60.79 (0.67–0.92)1372.00.94 (0.78–1.12)Exercise maintainers8,9241382.40.76 (0.64–0.90)681.20.58 (0.45–0.74)P-value< 0.001< 0.001  ≥ 65 yearsNon-exercisers38,3771,6757.51 (Reference)1,8098.11 (Reference)ExercisersNew exercisers7,4812525.70.79 (0.69–0.90)2555.80.75 (0.65–0.85)Exercise dropouts7,5082545.80.78 (0.69–0.90)3067.00.88 (0.78–1.00)Exercise maintainers5,4471564.90.69 (0.59–0.82)1916.00.79 (0.68–0.91)P-value< 0.001< 0.001P for interaction†0.4340.074Sex  MaleNon-exercisers50,7141,5504.91 (Reference)1,4304.61 (Reference)ExercisersNew exercisers10,8562954.40.84 (0.74–0.95)2553.80.78 (0.68–0.89)Exercise dropouts9,9352624.30.79 (0.69–0.90)2804.60.89 (0.79–1.02)Exercise maintainers8,8312033.70.73 (0.63–0.84)1973.60.75 (0.65–0.87)P-value< 0.001< 0.001  FemaleNon-exercisers59,7851,4613.91 (Reference)1,2043.21 (Reference)ExercisersNew exercisers10,2001872.90.79 (0.68–0.93)1482.30.82 (0.69–0.97)Exercise dropouts9,4421823.10.79 (0.68–0.92)1672.80.93 (0.79–1.09)Exercise maintainers6,079932.50.71 (0.58–0.88)641.70.65 (0.51–0.84)P-value< 0.0010.002P for interaction†0.9460.714*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included†The P for interaction was calculated by adding an interaction term to the multivariable modelIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, COPD chronic obstructive pulmonary disease\n\nEffect of exercise on risk of myocardial infarction or stroke\n*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included\n†The P for interaction was calculated by adding an interaction term to the multivariable model\nIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, COPD chronic obstructive pulmonary disease\nIn stratified analysis, age or sex did not have a significant interaction with the association between exercise and cardiovascular outcomes. The risk-reducing effect of exercise for MI or stroke was only significant in those ≥ 40 years of age (p < 0.001 for both 40–64 years and ≥ 65 years). The effect of exercise was not significantly different between men and women, and exercisers of both genders showed a significant reduction in the risk of MI (p < 0.001 for both) or stroke (p < 0.001 for men; p = 0.002 for women) (Table 2).\n[SUBTITLE] Amount of exercise and risk of MI or stroke [SUBSECTION] The associations between amount of exercise, measured by energy expenditure using METs, and risk of MI or stroke in patients with bronchiectasis are described in Table 3. Exercisers showed a lower risk for MI or stroke than non-exercisers regardless of the amount of exercise (p < 0.001 for both). However, a significant reduction was observed only in those with ≥ 500 MET-min/wk.\n\nTable 3The relationship between amount of exercise and risk of myocardial infarction or strokeNo. of patientsMyocardial infarctionStrokeNo. of eventsIR (/1,000 PY)aHR* (95% CI)No. of eventsIR (/1,000 PY)aHR* (95% CI)Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Overall exercisers  < 500 MET-min/wk11,1123024.30.91(0.81–1.03)2874.10.99(0.88–1.12)  500–999 MET-min/wk11,4072283.20.71(0.62–0.82)2223.10.80(0.69–0.91)  ≥ 1000 MET-min/wk32,8246923.40.76(0.70–0.83)6023.00.76(0.69–0.83)P-value< 0.001< 0.001Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)New exercisers  < 500 MET-min/wk457113.80.89(0.49–1.61)144.81.38(0.82–2.33)  500–999 MET-min/wk2,384583.80.92(0.71–1.19)352.30.66(0.48–0.93)  ≥ 1000 MET-min/wk18,2154133.60.81(0.73–0.90)3543.10.79(0.71–0.89)P-value< 0.001< 0.001Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Exercise dropouts  < 500 MET-min/wk10,3772844.40.91(0.81–1.03)2694.10.99(0.87–1.12)  500–999 MET-min/wk7,5201423.10.66(0.56–0.78)1583.40.83(0.71–0.98)  ≥ 1000 MET-min/wk1,480182.00.51(0.32–0.81)202.20.67(0.43–1.04)P-value< 0.0010.046Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Exercise maintainers  < 500 MET-min/wk27873.80.93(0.44–1.94)42.20.64(0.24–1.71)  500–999 MET-min/wk1,503282.90.68(0.47–0.98)293.10.82(0.57–1.18)  ≥ 1000 MET-min/wk13,1292613.20.72(0.64–0.82)2282.80.72(0.62–0.82)P-value< 0.001< 0.001*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were includedIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, MET metabolic equivalent of task, COPD chronic obstructive pulmonary disease\n\nThe relationship between amount of exercise and risk of myocardial infarction or stroke\n0.91\n(0.81–1.03)\n0.99\n(0.88–1.12)\n0.71\n(0.62–0.82)\n0.80\n(0.69–0.91)\n0.76\n(0.70–0.83)\n0.76\n(0.69–0.83)\n0.89\n(0.49–1.61)\n1.38\n(0.82–2.33)\n0.92\n(0.71–1.19)\n0.66\n(0.48–0.93)\n0.81\n(0.73–0.90)\n0.79\n(0.71–0.89)\n0.91\n(0.81–1.03)\n0.99\n(0.87–1.12)\n0.66\n(0.56–0.78)\n0.83\n(0.71–0.98)\n0.51\n(0.32–0.81)\n0.67\n(0.43–1.04)\n0.93\n(0.44–1.94)\n0.64\n(0.24–1.71)\n0.68\n(0.47–0.98)\n0.82\n(0.57–1.18)\n0.72\n(0.64–0.82)\n0.72\n(0.62–0.82)\n*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included\nIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, MET metabolic equivalent of task, COPD chronic obstructive pulmonary disease\nRegarding MI, compared to non-exercisers, exercisers with 500–999 MET-min/wk showed the lowest risk (aHR, 0.71; 95% CI, 0.62–0.82), followed by exercisers with ≥ 1000 MET-min/wk (aHR, 0.76; 95% CI, 0.70–0.83) and exercisers with < 500 MET-min/wk (aHR, 0.91; 95% CI, 0.81–1.03). Subgroup analysis showed a significantly reduced risk for MI in new exercisers with ≥ 1000 MET-min/wk and in exercise dropouts and exercise maintainers with ≥ 500 MET-min/wk (all p < 0.001). The greatest risk reduction for MI was 49%, observed in exercise dropouts with initial ≥ 1000 MET-min/wk (Table 3).\nFor stroke, compared to non-exercisers, exercisers with ≥ 1000 MET-min/wk showed the lowest risk (aHR, 0.76; 95% CI, 0.69–0.83), followed by exercisers with 500–999 MET-min/wk (aHR, 0.80; 95% CI, 0.69–0.91) and exercisers with < 500 MET-min/wk (aHR, 0.99; 95% CI, 0.88–1.12). In subgroup analysis, a significantly reduced risk for stroke was observed in new exercisers with ≥ 500 MET-min/wk (p < 0.001), in exercise dropouts with 500–999 MET-min/wk (p = 0.046), and in exercise maintainers with ≥ 1000 MET-min/wk (p < 0.001). The greatest risk reduction for stroke was 44%, observed in new exercisers with 500–999 MET-min/wk (Table 3).\nThe associations between amount of exercise, measured by energy expenditure using METs, and risk of MI or stroke in patients with bronchiectasis are described in Table 3. Exercisers showed a lower risk for MI or stroke than non-exercisers regardless of the amount of exercise (p < 0.001 for both). However, a significant reduction was observed only in those with ≥ 500 MET-min/wk.\n\nTable 3The relationship between amount of exercise and risk of myocardial infarction or strokeNo. of patientsMyocardial infarctionStrokeNo. of eventsIR (/1,000 PY)aHR* (95% CI)No. of eventsIR (/1,000 PY)aHR* (95% CI)Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Overall exercisers  < 500 MET-min/wk11,1123024.30.91(0.81–1.03)2874.10.99(0.88–1.12)  500–999 MET-min/wk11,4072283.20.71(0.62–0.82)2223.10.80(0.69–0.91)  ≥ 1000 MET-min/wk32,8246923.40.76(0.70–0.83)6023.00.76(0.69–0.83)P-value< 0.001< 0.001Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)New exercisers  < 500 MET-min/wk457113.80.89(0.49–1.61)144.81.38(0.82–2.33)  500–999 MET-min/wk2,384583.80.92(0.71–1.19)352.30.66(0.48–0.93)  ≥ 1000 MET-min/wk18,2154133.60.81(0.73–0.90)3543.10.79(0.71–0.89)P-value< 0.001< 0.001Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Exercise dropouts  < 500 MET-min/wk10,3772844.40.91(0.81–1.03)2694.10.99(0.87–1.12)  500–999 MET-min/wk7,5201423.10.66(0.56–0.78)1583.40.83(0.71–0.98)  ≥ 1000 MET-min/wk1,480182.00.51(0.32–0.81)202.20.67(0.43–1.04)P-value< 0.0010.046Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Exercise maintainers  < 500 MET-min/wk27873.80.93(0.44–1.94)42.20.64(0.24–1.71)  500–999 MET-min/wk1,503282.90.68(0.47–0.98)293.10.82(0.57–1.18)  ≥ 1000 MET-min/wk13,1292613.20.72(0.64–0.82)2282.80.72(0.62–0.82)P-value< 0.001< 0.001*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were includedIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, MET metabolic equivalent of task, COPD chronic obstructive pulmonary disease\n\nThe relationship between amount of exercise and risk of myocardial infarction or stroke\n0.91\n(0.81–1.03)\n0.99\n(0.88–1.12)\n0.71\n(0.62–0.82)\n0.80\n(0.69–0.91)\n0.76\n(0.70–0.83)\n0.76\n(0.69–0.83)\n0.89\n(0.49–1.61)\n1.38\n(0.82–2.33)\n0.92\n(0.71–1.19)\n0.66\n(0.48–0.93)\n0.81\n(0.73–0.90)\n0.79\n(0.71–0.89)\n0.91\n(0.81–1.03)\n0.99\n(0.87–1.12)\n0.66\n(0.56–0.78)\n0.83\n(0.71–0.98)\n0.51\n(0.32–0.81)\n0.67\n(0.43–1.04)\n0.93\n(0.44–1.94)\n0.64\n(0.24–1.71)\n0.68\n(0.47–0.98)\n0.82\n(0.57–1.18)\n0.72\n(0.64–0.82)\n0.72\n(0.62–0.82)\n*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included\nIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, MET metabolic equivalent of task, COPD chronic obstructive pulmonary disease\nRegarding MI, compared to non-exercisers, exercisers with 500–999 MET-min/wk showed the lowest risk (aHR, 0.71; 95% CI, 0.62–0.82), followed by exercisers with ≥ 1000 MET-min/wk (aHR, 0.76; 95% CI, 0.70–0.83) and exercisers with < 500 MET-min/wk (aHR, 0.91; 95% CI, 0.81–1.03). Subgroup analysis showed a significantly reduced risk for MI in new exercisers with ≥ 1000 MET-min/wk and in exercise dropouts and exercise maintainers with ≥ 500 MET-min/wk (all p < 0.001). The greatest risk reduction for MI was 49%, observed in exercise dropouts with initial ≥ 1000 MET-min/wk (Table 3).\nFor stroke, compared to non-exercisers, exercisers with ≥ 1000 MET-min/wk showed the lowest risk (aHR, 0.76; 95% CI, 0.69–0.83), followed by exercisers with 500–999 MET-min/wk (aHR, 0.80; 95% CI, 0.69–0.91) and exercisers with < 500 MET-min/wk (aHR, 0.99; 95% CI, 0.88–1.12). In subgroup analysis, a significantly reduced risk for stroke was observed in new exercisers with ≥ 500 MET-min/wk (p < 0.001), in exercise dropouts with 500–999 MET-min/wk (p = 0.046), and in exercise maintainers with ≥ 1000 MET-min/wk (p < 0.001). The greatest risk reduction for stroke was 44%, observed in new exercisers with 500–999 MET-min/wk (Table 3).", "In a total of 165,842 patients with bronchiectasis, the mean age was 59.8 years, and 48.4% were men. The study population comprised 66.6% non-exercisers and 33.4% exercisers. Moreover, exercisers were composed of new exercisers (n = 21,056, 12.7%), exercise dropouts (n = 19,377, 11.7%), and exercise maintainers (n = 14,910, 9.0%) according to changes in exercise habits (Fig. 1).\nExercisers were more likely to be older, male, non-smokers, and from low-income groups than non-exercisers (all p < 0.001). Regarding measurements in health screening exams, exercisers had lower total cholesterol levels, eGFR, and proportion of central obesity than non-exercisers (all p < 0.001); however, exercisers showed higher BMI, fasting blood sugar, systolic blood pressure, and longer waist circumference than non-exercisers (all p < 0.001). Of comorbidities, exercisers showed higher rates of hypertension, dyslipidemia, diabetes mellitus, and asthma or COPD compared with non-exercisers, and the differences between the four categories were statistically significant. Meanwhile, non-exercisers showed the lowest energy expenditure (32.5% for ≥ 500 MET-min/wk), followed by exercise dropouts (46.5%), new exercisers (97.8%), and exercise maintainers (98.1%) (p < 0.001) (Table 1).\n\nTable 1Baseline characteristics of the study populationVariablesTotal(N = 165,842)Non-exercisers(n = 110,499, 66.6%)Exercisers (n = 55,343, 33.4%)New exercisers(n = 21,056, 12.7%)Exercise dropouts(n = 19,377, 11.7%)Exercise maintainers(n = 14,910, 9.0%)P-value\nSociodemographics\nAge, years59.8 ± 11.959.5 ± 12.360.1 ± 11.160.8 ± 11.360.4 ± 10.6< 0.001  < 40 years9,621 (5.8)7,215 (6.5)950 (4.5)917 (4.7)539 (3.6)< 0.001  40–64 years97,408 (58.7)64,907 (58.8)12,625 (60.0)10,952 (56.5)8,924 (59.9)  ≥ 65 years58,813 (35.5)38,377 (34.7)7,481 (35.5)7,508 (38.8)5,447 (36.5)Sex< 0.001  Male80,336 (48.4)50,714 (45.9)10,856 (51.6)9,935 (51.3)8,831 (59.2)  Female85,506 (51.6)59,785 (54.1)10,200 (48.4)9,442 (48.7)6,079 (40.8)Low income*31,365 (18.9)21,210 (19.2)3,923 (18.6)3,674 (19.0)2,558 (17.2)< 0.001Smoking status  Current or past smoker24,294 (14.7)17,239 (15.6)2,739 (13.0)2,526 (13.0)1,790 (12.0)< 0.001Alcohol consumption  Heavy drinker8,402 (5.1)5,579 (5.1)1,093 (5.2)865 (4.5)865 (5.8)< 0.001\nMeasurements\nBody mass index, kg/m223.5 ± 3.323.5 ± 3.323.6 ± 3.123.7 ± 3.223.7 ± 3.0< 0.001Waist circumference, cm80.9 ± 9.180.8 ± 9.380.9 ± 8.981.3 ± 8.981.2 ± 8.6< 0.001Central obesity38,903 (23.5)26,626 (24.1)4,653 (22.1)4,514 (23.3)3,110 (20.9)< 0.001Fasting glucose, mg/dL100.3 ± 23.0100.0 ± 23.1100.4 ± 22.7101.0 ± 23.3101.1 ± 22.8< 0.001Systolic BP, mmHg123.3 ± 14.6123.1 ± 14.8123.4 ± 14.4123.8 ± 14.5123.7 ± 14.1< 0.001Diastolic BP, mmHg75.7 ± 9.575.7 ± 9.675.7 ± 9.475.9 ± 9.475.8 ± 9.40.057Total cholesterol, mg/dL193.7 ± 37.8194.2 ± 38.0192.7 ± 37.5193.1 ± 37.9192.1 ± 36.5< 0.001eGFR, ml/min/1.73m289.1 ± 40.789.5 ± 41.089.0 ± 44.787.9 ± 33.688.1 ± 41.5< 0.001\nComorbidities\nHypertension64,250 (38.7)42,287 (38.3)8,204 (39.0)7,884 (40.7)5,875 (39.4)< 0.001Dyslipidemia50,422 (30.4)32,984 (29.9)6,496 (30.9)6,326 (32.7)4,616 (31.0)< 0.001Diabetes mellitus22,762 (13.7)14,623 (13.2)2,985 (14.2)2,981 (15.4)2,173 (14.6)< 0.001Chronic kidney disease10,948 (6.6)7,333 (6.6)1,355 (6.4)1,355 (7.0)905 (6.1)0.005Asthma or COPD7,885 (4.8)4,871 (4.4)1,204 (5.7)975 (5.0)835 (5.6)< 0.001Cancer57,243 (34.5)38,630 (35.0)7,120 (33.8)6,810 (35.1)4,683 (31.4)< 0.001\nAmounts of exercise\nEnergy expenditure,MET-min/wk624.1 ± 635.2347.0 ± 330.71,552.3 ± 556.7463.0 ± 356.31,618.9 ± 585.2< 0.001≥ 500 MET-min/wk80,102 (48.3)35,871 (32.5)20,599 (97.8)9,000 (46.5)14,632 (98.1)< 0.001Data are presented as number (percentage) or mean ± standard deviation*Low income denotes income in the lowest 20% of the entire Korean populationBP blood pressure, eGFR estimated glomerular filtration rate, COPD chronic obstructive pulmonary disease, MET metabolic equivalent of task\n\nBaseline characteristics of the study population\nEnergy expenditure,\nMET-min/wk\nData are presented as number (percentage) or mean ± standard deviation\n*Low income denotes income in the lowest 20% of the entire Korean population\nBP blood pressure, eGFR estimated glomerular filtration rate, COPD chronic obstructive pulmonary disease, MET metabolic equivalent of task", "During a mean follow-up duration of 6.2 ± 2.1 years, 4,233 (2.6%) and 3,745 (2.3%) patients with bronchiectasis experienced MI and stroke, respectively. The Kaplan-Meier curves showed a significantly lower incidence probability for MI in exercisers than in non-exercisers, regardless of changes in exercise habits. In contrast, the incidence probability for stroke was reduced in new exercisers and exercise maintainers, but not in exercise dropouts, compared with non-exercisers (Fig. 2).\n\nFig. 2Kaplan-Meier curves of incidence probability for myocardial infarction or stroke according to change in exercise habits\n\nKaplan-Meier curves of incidence probability for myocardial infarction or stroke according to change in exercise habits\nTable 2 reveals the effect of exercise on the risk of incident MI or stroke in patients with bronchiectasis. Performing an exercise was associated with a reduced risk of 18–28% in MI or of 9–28% in stroke (p < 0.001 for both). Compared to non-exercisers, exercise maintainers showed the lowest risk of MI (adjusted hazard ratio [aHR], 0.72; 95% confidence interval [CI], 0.64–0.81), followed by exercise dropouts (aHR, 0.79; 95% CI, 0.72–0.87) and new exercisers (aHR, 0.82; 95% CI, 0.75–0.91) (p < 0.001). Regarding stroke, exercise maintainers also showed the lowest risk (aHR, 0.72; 95% CI, 0.64–0.82), followed by new exercisers (aHR, 0.79; 95% CI, 0.71–0.88) and exercise dropouts (aHR, 0.91; 95% CI, 0.82–1.00) compared with non-exercisers.\n\nTable 2Effect of exercise on risk of myocardial infarction or strokeNo. of patientsMyocardial infarctionStrokeNo. ofeventsIR (/1,000 PY)aHR* (95% CI)No. ofeventsIR (/1,000 PY)aHR* (95% CI)OverallNon-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)ExercisersNew exercisers21,0564823.70.82 (0.75–0.91)4033.10.79 (0.71–0.88)Exercise dropouts19,3774443.70.79 (0.72–0.87)4473.70.91 (0.82–1.00)Exercise maintainers14,9102963.20.72 (0.64–0.81)2612.80.72 (0.64–0.82)P-value< 0.001< 0.001Age, years  < 40 yearsNon-exercisers7,215350.71 (Reference)140.31 (Reference)ExercisersNew exercisers95010.20.22 (0.03–1.62)10.20.56 (0.07–4.23)Exercise dropouts91771.11.53 (0.68–3.45)40.62.21 (0.73–6.71)Exercise maintainers53920.60.78 (0.19–3.26)20.61.94 (0.44–8.55)P-value0.2280.479  40–64 yearsNon-exercisers64,9071,3013.11 (Reference)8112.01 (Reference)ExercisersNew exercisers12,6252292.80.87 (0.76–1.00)1471.80.88 (0.74–1.05)Exercise dropouts10,9521832.60.79 (0.67–0.92)1372.00.94 (0.78–1.12)Exercise maintainers8,9241382.40.76 (0.64–0.90)681.20.58 (0.45–0.74)P-value< 0.001< 0.001  ≥ 65 yearsNon-exercisers38,3771,6757.51 (Reference)1,8098.11 (Reference)ExercisersNew exercisers7,4812525.70.79 (0.69–0.90)2555.80.75 (0.65–0.85)Exercise dropouts7,5082545.80.78 (0.69–0.90)3067.00.88 (0.78–1.00)Exercise maintainers5,4471564.90.69 (0.59–0.82)1916.00.79 (0.68–0.91)P-value< 0.001< 0.001P for interaction†0.4340.074Sex  MaleNon-exercisers50,7141,5504.91 (Reference)1,4304.61 (Reference)ExercisersNew exercisers10,8562954.40.84 (0.74–0.95)2553.80.78 (0.68–0.89)Exercise dropouts9,9352624.30.79 (0.69–0.90)2804.60.89 (0.79–1.02)Exercise maintainers8,8312033.70.73 (0.63–0.84)1973.60.75 (0.65–0.87)P-value< 0.001< 0.001  FemaleNon-exercisers59,7851,4613.91 (Reference)1,2043.21 (Reference)ExercisersNew exercisers10,2001872.90.79 (0.68–0.93)1482.30.82 (0.69–0.97)Exercise dropouts9,4421823.10.79 (0.68–0.92)1672.80.93 (0.79–1.09)Exercise maintainers6,079932.50.71 (0.58–0.88)641.70.65 (0.51–0.84)P-value< 0.0010.002P for interaction†0.9460.714*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included†The P for interaction was calculated by adding an interaction term to the multivariable modelIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, COPD chronic obstructive pulmonary disease\n\nEffect of exercise on risk of myocardial infarction or stroke\n*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included\n†The P for interaction was calculated by adding an interaction term to the multivariable model\nIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, COPD chronic obstructive pulmonary disease\nIn stratified analysis, age or sex did not have a significant interaction with the association between exercise and cardiovascular outcomes. The risk-reducing effect of exercise for MI or stroke was only significant in those ≥ 40 years of age (p < 0.001 for both 40–64 years and ≥ 65 years). The effect of exercise was not significantly different between men and women, and exercisers of both genders showed a significant reduction in the risk of MI (p < 0.001 for both) or stroke (p < 0.001 for men; p = 0.002 for women) (Table 2).", "The associations between amount of exercise, measured by energy expenditure using METs, and risk of MI or stroke in patients with bronchiectasis are described in Table 3. Exercisers showed a lower risk for MI or stroke than non-exercisers regardless of the amount of exercise (p < 0.001 for both). However, a significant reduction was observed only in those with ≥ 500 MET-min/wk.\n\nTable 3The relationship between amount of exercise and risk of myocardial infarction or strokeNo. of patientsMyocardial infarctionStrokeNo. of eventsIR (/1,000 PY)aHR* (95% CI)No. of eventsIR (/1,000 PY)aHR* (95% CI)Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Overall exercisers  < 500 MET-min/wk11,1123024.30.91(0.81–1.03)2874.10.99(0.88–1.12)  500–999 MET-min/wk11,4072283.20.71(0.62–0.82)2223.10.80(0.69–0.91)  ≥ 1000 MET-min/wk32,8246923.40.76(0.70–0.83)6023.00.76(0.69–0.83)P-value< 0.001< 0.001Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)New exercisers  < 500 MET-min/wk457113.80.89(0.49–1.61)144.81.38(0.82–2.33)  500–999 MET-min/wk2,384583.80.92(0.71–1.19)352.30.66(0.48–0.93)  ≥ 1000 MET-min/wk18,2154133.60.81(0.73–0.90)3543.10.79(0.71–0.89)P-value< 0.001< 0.001Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Exercise dropouts  < 500 MET-min/wk10,3772844.40.91(0.81–1.03)2694.10.99(0.87–1.12)  500–999 MET-min/wk7,5201423.10.66(0.56–0.78)1583.40.83(0.71–0.98)  ≥ 1000 MET-min/wk1,480182.00.51(0.32–0.81)202.20.67(0.43–1.04)P-value< 0.0010.046Non-exercisers110,4993,0114.41 (Reference)2,6343.81 (Reference)Exercise maintainers  < 500 MET-min/wk27873.80.93(0.44–1.94)42.20.64(0.24–1.71)  500–999 MET-min/wk1,503282.90.68(0.47–0.98)293.10.82(0.57–1.18)  ≥ 1000 MET-min/wk13,1292613.20.72(0.64–0.82)2282.80.72(0.62–0.82)P-value< 0.001< 0.001*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were includedIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, MET metabolic equivalent of task, COPD chronic obstructive pulmonary disease\n\nThe relationship between amount of exercise and risk of myocardial infarction or stroke\n0.91\n(0.81–1.03)\n0.99\n(0.88–1.12)\n0.71\n(0.62–0.82)\n0.80\n(0.69–0.91)\n0.76\n(0.70–0.83)\n0.76\n(0.69–0.83)\n0.89\n(0.49–1.61)\n1.38\n(0.82–2.33)\n0.92\n(0.71–1.19)\n0.66\n(0.48–0.93)\n0.81\n(0.73–0.90)\n0.79\n(0.71–0.89)\n0.91\n(0.81–1.03)\n0.99\n(0.87–1.12)\n0.66\n(0.56–0.78)\n0.83\n(0.71–0.98)\n0.51\n(0.32–0.81)\n0.67\n(0.43–1.04)\n0.93\n(0.44–1.94)\n0.64\n(0.24–1.71)\n0.68\n(0.47–0.98)\n0.82\n(0.57–1.18)\n0.72\n(0.64–0.82)\n0.72\n(0.62–0.82)\n*In the adjustment, age, sex, low income, smoking status, alcohol consumption, body mass index, hypertension, diabetes mellitus, dyslipidemia, asthma or COPD, and cancer were included\nIR incidence rate, PY person-years, aHR adjusted hazard ratio, CI confidence interval, MET metabolic equivalent of task, COPD chronic obstructive pulmonary disease\nRegarding MI, compared to non-exercisers, exercisers with 500–999 MET-min/wk showed the lowest risk (aHR, 0.71; 95% CI, 0.62–0.82), followed by exercisers with ≥ 1000 MET-min/wk (aHR, 0.76; 95% CI, 0.70–0.83) and exercisers with < 500 MET-min/wk (aHR, 0.91; 95% CI, 0.81–1.03). Subgroup analysis showed a significantly reduced risk for MI in new exercisers with ≥ 1000 MET-min/wk and in exercise dropouts and exercise maintainers with ≥ 500 MET-min/wk (all p < 0.001). The greatest risk reduction for MI was 49%, observed in exercise dropouts with initial ≥ 1000 MET-min/wk (Table 3).\nFor stroke, compared to non-exercisers, exercisers with ≥ 1000 MET-min/wk showed the lowest risk (aHR, 0.76; 95% CI, 0.69–0.83), followed by exercisers with 500–999 MET-min/wk (aHR, 0.80; 95% CI, 0.69–0.91) and exercisers with < 500 MET-min/wk (aHR, 0.99; 95% CI, 0.88–1.12). In subgroup analysis, a significantly reduced risk for stroke was observed in new exercisers with ≥ 500 MET-min/wk (p < 0.001), in exercise dropouts with 500–999 MET-min/wk (p = 0.046), and in exercise maintainers with ≥ 1000 MET-min/wk (p < 0.001). The greatest risk reduction for stroke was 44%, observed in new exercisers with 500–999 MET-min/wk (Table 3).", "In this large-scale population-based cohort study, notable findings are as follows. First, more than 2% of patients with bronchiectasis developed MI or stroke during a mean of 6.2 follow-up years. Second, approximately two-thirds of patients with bronchiectasis have remained non-exercisers after their diagnosis of bronchiectasis. Third, exercise in patients with bronchiectasis was associated with a lower risk of MI or stroke. The effect of exercise on cardiovascular risk reduction was highest in exercise maintainers. This effect was also significant in those who initiated exercise after being diagnosed with bronchiectasis. Finally, exercising more than 500 MET-min/wk reduced the risk of cardiovascular diseases. Our findings suggest that a proper amount of regular exercise could prevent cardiovascular diseases in patients with bronchiectasis.\nEvidence supports increased cardiovascular risk in patients with bronchiectasis [6, 7, 27]. Although the reasons for increased cardiovascular risk in patients with bronchiectasis remain unclear, endothelial dysfunction is one of the proposed mechanisms [28]. Endothelial damage can be directly induced by increased platelet activation and systemic inflammation in patients with bronchiectasis [29]. Moreover, the increased cardiovascular risk was more pronounced after a respiratory tract infection in patients with bronchiectasis compared with their baseline (1.56 of incident rate ratio in the 91 days) [7]. Consequently, we need interventions to modify cardiovascular risk in patients with bronchiectasis during their stable state. Some studies demonstrated that exercise could alter catecholamine secretion, signaling protein expression and endothelial cell metabolism, leading to endothelial repair [30, 31]. In the same vein, the current study also revealed that exercise was related to reduced cardiovascular risk in patients with bronchiectasis. Therefore, future bronchiectasis guidelines should include a recommendation for exercise to reduce the risk of cardiovascular diseases.\nIn this study, exercise was associated with reduced cardiovascular disease risk in patients with bronchiectasis; risk of MI and stroke was reduced by up to 28% in exercise maintainers. Additionally, the effect of exercise is significant for patients who initiated an exercise program after being diagnosed with bronchiectasis. Also, a previous study revealed that low levels of physical activity and high sedentary time were associated with a 5.9-fold higher risk of hospitalization due to bronchiectasis exacerbation [32], which is a well-known risk factor for long-term mortality [33]. Taken together, these findings suggest that exercise has positive effects on long-term survival in patients with bronchiectasis via two mechanisms: reduced cardiovascular disease risk and reduced bronchiectasis exacerbation. In the near future, clinicians might have to evaluate the exercise status of patients with bronchiectasis and prescribe exercise to improve the long-term survival of their patients.\nNotably, exercisers were older, had a higher proportion of males, higher BMI, higher fasting blood glucose, and higher systolic blood pressure than non-exercisers. The aforementioned factors, such as older age, male sex, BMI, diabetes mellitus, and hypertension, are well-established determinants of cardiovascular disease risk [34–37]. Despite having many risk factors for cardiovascular diseases, patients with bronchiectasis who exercise revealed a significantly lower cardiovascular risk than those who did not exercise even before adjusting for those factors (see incidence rate of MI or stroke in Table 2), which might be an important finding for clinicians. When patients are diagnosed with bronchiectasis, respiratory physicians cannot modify some cardiovascular risk determinants, such as sex and comorbidities; however, they may be able to modify exercise habits and sedentary behavior to improve long-term prognosis in patients with bronchiectasis.\nRegarding the amount of exercise, more than 500 MET-min/wk was associated with a lower risk of cardiovascular diseases in patients with bronchiectasis. These results were consistent with the World Health Organization Guidelines on Physical Activity and Sedentary Behaviour, suggesting regular exercise more than 500 MET-min/wk for adults to achieve health benefits [38]. Regardless of the change in exercise habits, the risk of MI and stroke was reduced in those who exercised more than the recommended amount (500 MET-min/wk). In line with those results, a previous study of 64 patients with bronchiectasis revealed that reduced physical activity (< 6,290 steps/day) or high sedentary behavior (≥ 7.8 h/day) were related to a higher risk of hospitalization due to bronchiectasis exacerbation [32]. However, the optimal amount of exercise in patients with bronchiectasis is not known, and future studies are warranted to reveal this issue.\nTo the best of our knowledge, this is the first study evaluating whether exercise reduces the risk of cardiovascular diseases in patients with bronchiectasis and exploring the optimal physical activity in the population. Another strength of this study was the inclusion of various covariates such as BMI, smoking status, alcohol consumption, and comorbidities. However, this study has several limitations that should be acknowledged. First, the intensity and frequency of physical activity were evaluated by self-reported questionnaires that could be influenced by recall bias. Second, physiologic parameters such as peak heart rate and oxygen uptake could not be included to evaluate exercise intensity. Future research using more objective parameters on exercise intensity is required. Third, assessment for bronchiectasis severity was not available. Bronchiectasis is a chronic respiratory disease that affects workout abilities, and the effect of exercise can vary depending on the severity of bronchiectasis. When prescribing exercise in real practice, the exercise capacity of the patient with bronchiectasis should be considered. Fourth, this study was conducted in a single Asian country with a limited ethnic diversity. Therefore, generalization of our findings to other ethnic groups should be done with caution. Fifth, because our dataset lacked data on energy consumption, we could not consider the effect of energy consumption, an important factor affecting cardiovascular risks [39], on the relationship between physical activity and cardiovascular disease.", "Exercise was associated with reduced risk of cardiovascular diseases in patients with bronchiectasis. In particular, risk was lowest in exercise maintainers, and cardiovascular risk reduction was significant in those who exercised more than 500 MET-min/wk. Our study provides evidence of exercise prescription for patients with bronchiectasis to prevent cardiovascular diseases.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ "introduction", null, null, null, null, null, null, "results", null, null, null, "discussion", "conclusion", "supplementary-material", null ]
[ "Bronchiectasis", "Exercise", "Cardiovascular diseases", "Disease prevention, sedentary behavior" ]
Identification of castration-dependent and -independent driver genes and pathways in castration-resistant prostate cancer (CRPC).
36258196
Prostate cancer (PCa) is one of the most diagnosed cancers in the world. PCa inevitably progresses to castration-resistant prostate cancer (CRPC) after androgen deprivation therapy treatment, and castration-resistant state means a shorter survival time than other causes. Here we aimed to define castration-dependent and -independent diver genes and molecular pathways in CRPC which are responsible for such lethal metastatic events.
BACKGROUND
By employing digital gene expression (DGE) profiling, the alterations of the epididymal gene expression profile in the mature and bilateral castrated rat were explored. Then we detect and characterize the castration-dependent and -independent genes and pathways with two data set of CPRC-associated gene expression profiles publicly available on the NCBI.
METHODS
We identified 1,632 up-regulated and 816 down-regulated genes in rat's epididymis after bilateral castration. Differential expression analysis of CRPC samples compared with the primary PCa samples was also done. In contrast to castration, we identified 97 up-regulated genes and 128 down-regulated genes that changed in both GEO dataset and DGE profile, and 120 up-regulated genes and 136 down-regulated genes changed only in CRPC, considered as CRPC-specific genes independent of castration. CRPC-specific DEGs were mainly enriched in cell proliferation, while CRPC-castration genes were associated with prostate gland development. NUSAP1 and NCAPG were identified as key genes, which might be promising biomarkers of the diagnosis and prognosis of CRPC.
RESULTS
Our study will provide insights into gene regulation of CRPC dependent or independent of castration and will improve understandings of CRPC development and progression.
CONCLUSION
[ "Humans", "Male", "Rats", "Animals", "Prostatic Neoplasms, Castration-Resistant", "Androgen Antagonists", "Androgens", "Gene Expression Regulation, Neoplastic", "Orchiectomy", "Cell Line, Tumor", "Receptors, Androgen" ]
9580185
null
null
null
null
Results
[SUBTITLE] Dramatic transcriptional changes in rats’ epididymis after bilateral castration [SUBSECTION] To explore the transcriptional differences in the epididymis between Con-EP and Cas-EP rats, we obtained more than 4 million clean tags for each library. As demonstrated in Additional file 1, the distribution of total clean tags and distinct clean tags showed similar patterns in both DGE libraries, showing no bias between the library construction of Con-EP and Cas-EP. Around 3 million tags were mapped to genes for both Con-EP and Cas-EP, accounting for 62.89% and 70.32%, respectively. A total of 2,448 genes changed significantly between the two libraries. Among these genes, 1,632 were up-regulated while 816 were down-regulated after bilateral castration. More than 90% of the genes (2,274) were up- or down-regulated between 1.0 and 5.0 fold (data not shown). The Pearson correlation coefficient of the two libraries was only 0.658, reflecting the tremendous influence of castration on the gene expression profile of rat epididymis. We then performed qPCR of six selective genes which were up-regulated (Nfkbie, Plscr4 and Apoc1) or down-regulated (Lcn8, Prdx1 and Prdx3) to confirm the validity of the differentially expressed genes identified by DGE (n = 10). As expected, we obtained results that coincided with our sequencing data, confirming the authenticity of our sequencing results (Fig. 2). Fig. 2Real-time fluorescent quantitative PCR analysis of differentially expressed genes before and after castration. a: down-regulated genes after bilateral castration, b: genes up-regulated after bilateral castration Real-time fluorescent quantitative PCR analysis of differentially expressed genes before and after castration. a: down-regulated genes after bilateral castration, b: genes up-regulated after bilateral castration To explore the transcriptional differences in the epididymis between Con-EP and Cas-EP rats, we obtained more than 4 million clean tags for each library. As demonstrated in Additional file 1, the distribution of total clean tags and distinct clean tags showed similar patterns in both DGE libraries, showing no bias between the library construction of Con-EP and Cas-EP. Around 3 million tags were mapped to genes for both Con-EP and Cas-EP, accounting for 62.89% and 70.32%, respectively. A total of 2,448 genes changed significantly between the two libraries. Among these genes, 1,632 were up-regulated while 816 were down-regulated after bilateral castration. More than 90% of the genes (2,274) were up- or down-regulated between 1.0 and 5.0 fold (data not shown). The Pearson correlation coefficient of the two libraries was only 0.658, reflecting the tremendous influence of castration on the gene expression profile of rat epididymis. We then performed qPCR of six selective genes which were up-regulated (Nfkbie, Plscr4 and Apoc1) or down-regulated (Lcn8, Prdx1 and Prdx3) to confirm the validity of the differentially expressed genes identified by DGE (n = 10). As expected, we obtained results that coincided with our sequencing data, confirming the authenticity of our sequencing results (Fig. 2). Fig. 2Real-time fluorescent quantitative PCR analysis of differentially expressed genes before and after castration. a: down-regulated genes after bilateral castration, b: genes up-regulated after bilateral castration Real-time fluorescent quantitative PCR analysis of differentially expressed genes before and after castration. a: down-regulated genes after bilateral castration, b: genes up-regulated after bilateral castration [SUBTITLE] CRPC-castration genes were enriched in cell proliferation and prostate gland development [SUBSECTION] To obtain potential CRPC driver genes dependent or independent of castration, we identified DEGs in the CRPC samples compared with the primary PCa samples. We obtained 1,904 up-regulated genes and 3,391 down-regulated genes (fold change > 1, P < 0.001) from GSE35988 (Fig. 3a), and 512 increased genes and 244 decreased genes (fold change > 1, P < 0.001) from GSE32269 (Fig. 3b). A total of 97 up-regulated genes (Fig. 3d) and 128 down-regulated genes (Fig. 3c) were shared by CRPC (GSE35988) and testis castration (data not shown), considered as CRPC DEGs related to castration (CRPC-castration). In contrast to testis castration, 120 up-regulated genes and 136 down-regulated genes changed only in CRPC (both GSE35988 and GSE32269) (Fig. 3e, data not shown), considered as CRPC-specific DEGs independent of castration. Fig. 3Database analysis. a and b: Volcano plots of all genes in GSE35988 and GSE32269 Red dots represent genes with fold change ≥ 2 and P < 0.001, blue dots represent genes with fold change ≤ − 2 and P < 0.001, and the other dots represent the rest of genes with no statistically significant change in expression. FC, fold change. c, d and e: Venn diagrams showing the number of genes expressed as common and unique between the identified DEGs in DGE profiles of castrated rat and genes in the NCBI-gene database Database analysis. a and b: Volcano plots of all genes in GSE35988 and GSE32269 Red dots represent genes with fold change ≥ 2 and P < 0.001, blue dots represent genes with fold change ≤ − 2 and P < 0.001, and the other dots represent the rest of genes with no statistically significant change in expression. FC, fold change. c, d and e: Venn diagrams showing the number of genes expressed as common and unique between the identified DEGs in DGE profiles of castrated rat and genes in the NCBI-gene database Next, we explored the functional enrichment of these two lists of DEGs by using GO and KEGG analysis. The CRPC-specific DEGs significantly enriched cell division or mitotic processes (Fig. 4a), consistent with their oncogenic role in tumor cell proliferation. Concordantly, pathway analysis identified enrichment in the cell cycle pathway and focal adhesion pathway (Fig. 4b) (Additional file 2). On the other hand, GO enrichment of CRPC-castration DEGs (data not shown) showed that gland development, epithelial cell proliferation, and prostate gland development were substantially affected by castration (Fig. 5a). Pathway analysis showed that the pyruvate metabolism pathway was significantly enriched (Additional file 3). Fig. 4Go and KEGG analysis. a: The top 10 results of GO functional analysis in CRPC-specific DEGs. b: KEGG functional analysis in CRPC-specific DEGs. Go and KEGG analysis. a: The top 10 results of GO functional analysis in CRPC-specific DEGs. b: KEGG functional analysis in CRPC-specific DEGs. Fig. 5Go and PPI analysis. a: The top 10 results of GO functional analysis in CRPC-castration DEGs. b: Construction of PPI network of CRPC-castration DEGs. Go and PPI analysis. a: The top 10 results of GO functional analysis in CRPC-castration DEGs. b: Construction of PPI network of CRPC-castration DEGs. Furthermore, we used STRING to explore the protein-protein interaction (PPI) of the DEGs. CRPC-specific DEGs formed a PPI network with 208 nodes and 1353 edges. Among the 41 hub genes (node degree > 30) were chromatin remodeling factors EZH2 and PRC1, and cell cycle regulators CDCA3, CCNA2, CCNB2, CDKN3, CDK1, CCNB1, CDC20, MCM4, SMC4, CENPF, BUB1, BUB1B, NUSAP1, and NCAPG (Fig. 6). In contrast, CRPC-castration DEGs formed PPI networks with 171 nodes and sparse connections. The most connected sub-network with the hub gene ACTR10 was involved in innate immunity and cell polarity (Fig. 5b). Fig. 6Top 41 of PPI network of CRPC-specific DEGs. Top 41 of PPI network of CRPC-specific DEGs. Finally, based on network and pathway analyses, we identified two key genes (NUSAP1 and NCAPG) among the top 41 nodes in the CRPC-specific PPI network, which participate in various pathways including chromosome segregation and nuclear division. Further analysis showed that, for both genes, higher expression was associated with shorter disease-free survival of patients (Fig. 7c and d). Interestingly, the expression of NUSAP1 and NCAPG differed between primary PCa and normal prostate tissues (Fig. 7a and b), which led us to hypothesize that they may have a role both in the initiation and in the progression of PCa. Fig. 7Key genes analysis. a and b: Expression of the key genes NUSAP1 and NCAPG in TCGA with GEPIA. c and d: Disease-free survival in relation to the expression of the key genes (NUSAP1 and NCAPG) in TCGA. Key genes analysis. a and b: Expression of the key genes NUSAP1 and NCAPG in TCGA with GEPIA. c and d: Disease-free survival in relation to the expression of the key genes (NUSAP1 and NCAPG) in TCGA. To obtain potential CRPC driver genes dependent or independent of castration, we identified DEGs in the CRPC samples compared with the primary PCa samples. We obtained 1,904 up-regulated genes and 3,391 down-regulated genes (fold change > 1, P < 0.001) from GSE35988 (Fig. 3a), and 512 increased genes and 244 decreased genes (fold change > 1, P < 0.001) from GSE32269 (Fig. 3b). A total of 97 up-regulated genes (Fig. 3d) and 128 down-regulated genes (Fig. 3c) were shared by CRPC (GSE35988) and testis castration (data not shown), considered as CRPC DEGs related to castration (CRPC-castration). In contrast to testis castration, 120 up-regulated genes and 136 down-regulated genes changed only in CRPC (both GSE35988 and GSE32269) (Fig. 3e, data not shown), considered as CRPC-specific DEGs independent of castration. Fig. 3Database analysis. a and b: Volcano plots of all genes in GSE35988 and GSE32269 Red dots represent genes with fold change ≥ 2 and P < 0.001, blue dots represent genes with fold change ≤ − 2 and P < 0.001, and the other dots represent the rest of genes with no statistically significant change in expression. FC, fold change. c, d and e: Venn diagrams showing the number of genes expressed as common and unique between the identified DEGs in DGE profiles of castrated rat and genes in the NCBI-gene database Database analysis. a and b: Volcano plots of all genes in GSE35988 and GSE32269 Red dots represent genes with fold change ≥ 2 and P < 0.001, blue dots represent genes with fold change ≤ − 2 and P < 0.001, and the other dots represent the rest of genes with no statistically significant change in expression. FC, fold change. c, d and e: Venn diagrams showing the number of genes expressed as common and unique between the identified DEGs in DGE profiles of castrated rat and genes in the NCBI-gene database Next, we explored the functional enrichment of these two lists of DEGs by using GO and KEGG analysis. The CRPC-specific DEGs significantly enriched cell division or mitotic processes (Fig. 4a), consistent with their oncogenic role in tumor cell proliferation. Concordantly, pathway analysis identified enrichment in the cell cycle pathway and focal adhesion pathway (Fig. 4b) (Additional file 2). On the other hand, GO enrichment of CRPC-castration DEGs (data not shown) showed that gland development, epithelial cell proliferation, and prostate gland development were substantially affected by castration (Fig. 5a). Pathway analysis showed that the pyruvate metabolism pathway was significantly enriched (Additional file 3). Fig. 4Go and KEGG analysis. a: The top 10 results of GO functional analysis in CRPC-specific DEGs. b: KEGG functional analysis in CRPC-specific DEGs. Go and KEGG analysis. a: The top 10 results of GO functional analysis in CRPC-specific DEGs. b: KEGG functional analysis in CRPC-specific DEGs. Fig. 5Go and PPI analysis. a: The top 10 results of GO functional analysis in CRPC-castration DEGs. b: Construction of PPI network of CRPC-castration DEGs. Go and PPI analysis. a: The top 10 results of GO functional analysis in CRPC-castration DEGs. b: Construction of PPI network of CRPC-castration DEGs. Furthermore, we used STRING to explore the protein-protein interaction (PPI) of the DEGs. CRPC-specific DEGs formed a PPI network with 208 nodes and 1353 edges. Among the 41 hub genes (node degree > 30) were chromatin remodeling factors EZH2 and PRC1, and cell cycle regulators CDCA3, CCNA2, CCNB2, CDKN3, CDK1, CCNB1, CDC20, MCM4, SMC4, CENPF, BUB1, BUB1B, NUSAP1, and NCAPG (Fig. 6). In contrast, CRPC-castration DEGs formed PPI networks with 171 nodes and sparse connections. The most connected sub-network with the hub gene ACTR10 was involved in innate immunity and cell polarity (Fig. 5b). Fig. 6Top 41 of PPI network of CRPC-specific DEGs. Top 41 of PPI network of CRPC-specific DEGs. Finally, based on network and pathway analyses, we identified two key genes (NUSAP1 and NCAPG) among the top 41 nodes in the CRPC-specific PPI network, which participate in various pathways including chromosome segregation and nuclear division. Further analysis showed that, for both genes, higher expression was associated with shorter disease-free survival of patients (Fig. 7c and d). Interestingly, the expression of NUSAP1 and NCAPG differed between primary PCa and normal prostate tissues (Fig. 7a and b), which led us to hypothesize that they may have a role both in the initiation and in the progression of PCa. Fig. 7Key genes analysis. a and b: Expression of the key genes NUSAP1 and NCAPG in TCGA with GEPIA. c and d: Disease-free survival in relation to the expression of the key genes (NUSAP1 and NCAPG) in TCGA. Key genes analysis. a and b: Expression of the key genes NUSAP1 and NCAPG in TCGA with GEPIA. c and d: Disease-free survival in relation to the expression of the key genes (NUSAP1 and NCAPG) in TCGA.
Conclusion
Our study provided an insight into the regulation of epididymal gene expression after bilateral castration. These results contribute to our understanding of testis-dependent epididymal functions. Still, we preformed insights into gene regulation of CRPC dependent or independent of castration and improved our understanding of CRPC development and progression.
[ "Background", "Methods", "Microarray data and DGE profiling", "Animals and castration procedure", "Library preparation and sequencing", "Tag annotation and gene expression level quantification", "Real-time quantitative PCR analysis of genes", "Identification of DEGs of digital profiling data", "Identification of DEGs of microarray data", "Functional enrichment and PPI network analysis of DEGs", "Dramatic transcriptional changes in rats’ epididymis after bilateral castration", "CRPC-castration genes were enriched in cell proliferation and prostate gland development", "" ]
[ "One of the most common malignancies in man is prostate cancer (PCa), which also causes the second highest deaths in man. Absolutely, PCa arises as an androgen driven disease [1]. Therefore androgen deprivation therapy (ADT) was adopted as the major treatment for metastatic hormone-sensitive PCa, by suppressing circulating testosterone to decrease its level to “castrate levels” ( < = 50 ng/dL) [2] and induce of apoptosis [3]. However, although the initial treatment effect is significant, almost all PCa patients ultimately progress and castration resistant prostate cancer (CRPC) ensue, which is associated with high mortality rates [4]. CRPC represents a particular stage in the continuum of the disease and is defined by continuous rise of prostate specific antigen levels in serum or/and progression of metastatic spread in the setting of castrate levels of testosterone [5–7].\nNowadays, suppression of androgen receptor (AR) signaling through ADT has remained the first-line treatment for men with advanced PCa. ADT includes surgical or medical castration by using luteinizing hormone-releasing hormone (LHRH) agonists or antagonists with or without anti-androgen drugs [8]. Exposure to LHRH agonists downregulates the LHRH receptor, decreasing LH release and inhibiting testosterone production, while LHRH antagonists, such as cetrorelix and abarelix, directly inhibit the LHRH receptor, resulting in the decreased production of LH and testosterone. Surgical bilaterial castration also decreases testosterone levels by removing the testes, the source of its production organ, although surgical castration has been practically set aside for some time, given the spread of ADT [9]. However, by using LHRH agonists or antagonists, for nearly all patients, after 1–2 years remission, cancer cells become resistant with the emergence of metastatic CRPC [10]. Today’s standard of care for advanced PCa includes gonadotropin-releasing hormone agonists (leuprolide), second-generation nonsteroidal AR antagonists (apalutamide, enzalutamide and darolutamide) and the androgen biosynthesis inhibitor abiraterone [11]. Currently, with minimal additional toxicity, abiraterone acetate has been proved to be a well-tolerated, convenient and effective treatment option [12].\nBesides the above mentioned drugs for the treatment of CRPC, AR-V7, the androgen receptor isoform encoded by splice variant 7, has become an attractive biomarker predicting the effect of androgen signaling inhibitor [10, 13]. AR-V7 lacks the ligand binding domain and is the target of enzalutamide and abiraterone. It is supposed that detection of AR-V7 in circulating tumor cells from patients with CPRC might be closely related to resistance to enzalutamide and abiraterone [14].\nMany mechanisms of CRPC have been proposed, including reactivation of androgen synthesis, up-regulation of genes related to androgen metabolism, and reprogramming of the tumor microenvironment [13]. However, CRPC treatment is challenging [15]. Thus, identifying effective molecular biomarkers and therapeutic targets of CRPC is of ultra-importance for PCa prediction and treatment. Previously, Microarray techniques have been used to identify biomarkers and targets in CRPC. For example, D’Antonio et al. [16] identified pathways of androgen independence of CRPC by comparing PCa cells (androgen-independent) with LNCaP cells (androgen-dependent). Terada et al. [17] found that prostaglandin E2 receptor subtype 4 (EP4) is a central gene in CRPC by comparing a mouse xenograft model of PCa with later-derived CRPC.\nThe biological functions of the mammalian epididymis, an important male accessory gland, are substantially affected by androgen [18]. The mammalian epididymis is an important male accessory gland with many biological functions, including maturation and concentration of spermatozoa produced by testis, secretion and resorption of different molecules and proteins, and storage of spermatozoa. The development and normal functions of the epididymis are regulated mainly by androgens and testicular factors [19]. Bilateral castration led to systematic changes of epididymal gene expression profile [20]. It would be useful to identify critical androgen-related pathways in the epididymis to help understand CRPC and identify biomarkers or targets, thus providing opportunities to combat the disease.\nCompared with microarray, digital gene expression profiling provides absolute gene expression measurements and an unbiased view of the whole transcriptome with greater precision and sensitivity. Here we identified differentially expressed genes in the epididymides of sham-operated control and bilateral castrated male mature rats on the 7th day after surgery by using the DGE system on the whole-genome scale, as previous studies have demonstrated that gene expression profile is most likely to reflects transcriptional changes in surviving epididymal cells on the 7th day of post castration because apoptotic cell death is no longer detected at this moment [20]. Then we combined our data with two published datasets to identify differentially expressed genes (DEGs) between CRPC and primary PCa. We obtained DEGs of either dependent (CRPC-castration) or independent (CRPC-specific) of castration to investigate the role of castration in CRPC. Functional enrichment analysis, protein-protein interaction (PPI) network, and survival analysis of these DEGs showed distinct functional roles of CRPC-castration and CRPC-specific genes (Fig. 1). CRPC-specific DEGs were mainly enriched in cell proliferation, while CRPC-castration genes were associated with prostate gland development. In summary, our study provided insights into a deeper understanding of CRPC pathogenesis.\n\nFig. 1Flow diagram showing an overview of the study\n\nFlow diagram showing an overview of the study", "[SUBTITLE] Microarray data and DGE profiling [SUBSECTION] Two gene expression profiles (GSE35988, GSE32269) were obtained from Gene Expression Omnibus (GEO, https://www.ncbi.nlm.nih.gov/geo). GSE35988 contained 27 metastatic CPRC samples and 47 localized PCa samples, and GSE32269 was composed of 29 metastatic CPRC samples and 22 primary PCa samples.\nTwo gene expression profiles (GSE35988, GSE32269) were obtained from Gene Expression Omnibus (GEO, https://www.ncbi.nlm.nih.gov/geo). GSE35988 contained 27 metastatic CPRC samples and 47 localized PCa samples, and GSE32269 was composed of 29 metastatic CPRC samples and 22 primary PCa samples.\n[SUBTITLE] Animals and castration procedure [SUBSECTION] Twenty healthy and mature male Sprague-Dawley (SD) rats (12 weeks old and the body weight reached 350 ~ 400 g) were obtained from the Animal Research Centre of the Lv Ye Pharmaceutical Company (Yantai, China) and were housed at an environmental temperature of 20 ± 2 °C, receiving 12 h of light and 12 h of dark, with food and water provided ad libitum. Rats were randomly divided into two groups by using a random number Table (10 animals of each group), namely sham-operated control group (Con-EP) and castrated group (Cas-EP). Before castration treatment, rats were anesthetized by intraperitoneally injection of sodium pentobarbital (30 mg/kg body weight). Thereafter, the efferent ducts and the testicular arteries and veins were ligated, then the testes were removed from the ligation point, leaving the intact epididymides in the scrotal area. In the meantime, the sham operation was also taken for the control group. Analgesia was provided by administration of meloxicam Q 24 h (1 mg/kg PO or SC) (Metacam, Boehringer Ingelheim, St Joseph, MO) the day prior, the day of and the day following surgery. No obvious behavioral changes were observed in both control and the operation group. At the conclusion of the experiment, all the surviving rats were intraperitoneally anesthetized with sodium pentobarbital (30 mg/kg body weight) and their necks were dislocated for euthanasia. The whole epididymides were taken out from the castrated and sham-operated control rats after 7 days’ recovery and were immediately frozen in liquid nitrogen and stored at -80 ℃ before use. Protocols for the use of animals in these experiments were approved by the Research Animal Care and Use Committee of Yantai University (the approval reference number: YD.No20190916S0350210[315]) and were carried out in strict accordance with Guidelines for Ethical Review of Laboratory Animal Welfare of China. Ethical approval was obtained before the initiation of the research work and all efforts were made to minimize suffering.\nTwenty healthy and mature male Sprague-Dawley (SD) rats (12 weeks old and the body weight reached 350 ~ 400 g) were obtained from the Animal Research Centre of the Lv Ye Pharmaceutical Company (Yantai, China) and were housed at an environmental temperature of 20 ± 2 °C, receiving 12 h of light and 12 h of dark, with food and water provided ad libitum. Rats were randomly divided into two groups by using a random number Table (10 animals of each group), namely sham-operated control group (Con-EP) and castrated group (Cas-EP). Before castration treatment, rats were anesthetized by intraperitoneally injection of sodium pentobarbital (30 mg/kg body weight). Thereafter, the efferent ducts and the testicular arteries and veins were ligated, then the testes were removed from the ligation point, leaving the intact epididymides in the scrotal area. In the meantime, the sham operation was also taken for the control group. Analgesia was provided by administration of meloxicam Q 24 h (1 mg/kg PO or SC) (Metacam, Boehringer Ingelheim, St Joseph, MO) the day prior, the day of and the day following surgery. No obvious behavioral changes were observed in both control and the operation group. At the conclusion of the experiment, all the surviving rats were intraperitoneally anesthetized with sodium pentobarbital (30 mg/kg body weight) and their necks were dislocated for euthanasia. The whole epididymides were taken out from the castrated and sham-operated control rats after 7 days’ recovery and were immediately frozen in liquid nitrogen and stored at -80 ℃ before use. Protocols for the use of animals in these experiments were approved by the Research Animal Care and Use Committee of Yantai University (the approval reference number: YD.No20190916S0350210[315]) and were carried out in strict accordance with Guidelines for Ethical Review of Laboratory Animal Welfare of China. Ethical approval was obtained before the initiation of the research work and all efforts were made to minimize suffering.\n[SUBTITLE] Library preparation and sequencing [SUBSECTION] Total RNA was isolated from the epididymides from the control group and castrated group, respectively, by using RNAiso Plus reagent (TaKaRa, Dalian, China). The purity and quality of total RNA were evaluated by Agilent 2000 and were considered sufficient for subsequent library construction and sequencing. Tag libraries for the samples were prepared using the Illumina gene expression sample, Prep Kit. Each sample of 6 µg of the total RNA was extracted and purified by oligo (dT) magnetic bead adsorption. Primed by oligo (dT), mRNA bound to the bead was used as the template to synthesize a double-stranded cDNA. The 5’ ends of tags were digested with restriction enzyme NlaIII at CATG sites. The cDNA fragments with 3’ ends connected to Oligo(dT) beads were purified and the Illumina adaptor 1 was ligated to the 5’ ends. The junction of Illumina adaptor 1 and CATG site is the recognition site of MmeI, which is a type of endonuclease with separated recognition sites and digestion sites. It cuts at 17 bp downstream of the CATG site, producing tags with adaptor 1. After removing 3’ fragments with magnetic beads precipitation, Illumina adaptor 2 is ligated to the 3’ ends of tags, acquiring tags with different adaptors of both ends to form a tag library. After 15 cycles of linear PCR amplification, 105 bp fragments were purified by 6% TBE PAGE Gel electrophoresis. After denaturation, the single-chain molecules were fixed onto the Illumina Sequencing Chip (flowcell). Each molecule was grown into a single-molecule cluster sequencing template through Situ amplification. Then, they were sequenced with the method of sequencing by synthesis (SBS). Each tunnel generated millions of raw reads with a sequencing length of 49 bp.\nTotal RNA was isolated from the epididymides from the control group and castrated group, respectively, by using RNAiso Plus reagent (TaKaRa, Dalian, China). The purity and quality of total RNA were evaluated by Agilent 2000 and were considered sufficient for subsequent library construction and sequencing. Tag libraries for the samples were prepared using the Illumina gene expression sample, Prep Kit. Each sample of 6 µg of the total RNA was extracted and purified by oligo (dT) magnetic bead adsorption. Primed by oligo (dT), mRNA bound to the bead was used as the template to synthesize a double-stranded cDNA. The 5’ ends of tags were digested with restriction enzyme NlaIII at CATG sites. The cDNA fragments with 3’ ends connected to Oligo(dT) beads were purified and the Illumina adaptor 1 was ligated to the 5’ ends. The junction of Illumina adaptor 1 and CATG site is the recognition site of MmeI, which is a type of endonuclease with separated recognition sites and digestion sites. It cuts at 17 bp downstream of the CATG site, producing tags with adaptor 1. After removing 3’ fragments with magnetic beads precipitation, Illumina adaptor 2 is ligated to the 3’ ends of tags, acquiring tags with different adaptors of both ends to form a tag library. After 15 cycles of linear PCR amplification, 105 bp fragments were purified by 6% TBE PAGE Gel electrophoresis. After denaturation, the single-chain molecules were fixed onto the Illumina Sequencing Chip (flowcell). Each molecule was grown into a single-molecule cluster sequencing template through Situ amplification. Then, they were sequenced with the method of sequencing by synthesis (SBS). Each tunnel generated millions of raw reads with a sequencing length of 49 bp.\n[SUBTITLE] Tag annotation and gene expression level quantification [SUBSECTION] Five steps were needed to transform raw tags into clean tags: (1) removal of the 3’ adaptor to preserve the 21 nt long sequences; (2) removal of the empty reads (reads with only adaptor sequences but no tags); (3) removal of the low-quality tags (tags with unknown sequences); (4) removal of too long or too short tags, keeping the 21 nt tags; (5) removal of tags with only one copy number (probably because of sequencing error). To convert digital profiles to gene expression, the tag sequences were mapped to the reference genes of rat from NCBI, with no more than 1 nucleotide mismatch. Clean tags mapped to multiple loci were filtered, only unambiguous mapped tags were kept. To identify gene expression, the number of unambiguous tags for each gene was calculated and then normalized to the number of transcripts per million tags (TPM). Gene ontology was assigned to all mapped genes to annotate their possible functions.\nFive steps were needed to transform raw tags into clean tags: (1) removal of the 3’ adaptor to preserve the 21 nt long sequences; (2) removal of the empty reads (reads with only adaptor sequences but no tags); (3) removal of the low-quality tags (tags with unknown sequences); (4) removal of too long or too short tags, keeping the 21 nt tags; (5) removal of tags with only one copy number (probably because of sequencing error). To convert digital profiles to gene expression, the tag sequences were mapped to the reference genes of rat from NCBI, with no more than 1 nucleotide mismatch. Clean tags mapped to multiple loci were filtered, only unambiguous mapped tags were kept. To identify gene expression, the number of unambiguous tags for each gene was calculated and then normalized to the number of transcripts per million tags (TPM). Gene ontology was assigned to all mapped genes to annotate their possible functions.\n[SUBTITLE] Real-time quantitative PCR analysis of genes [SUBSECTION] Real-time quantitative PCR (qPCR) was performed to examine the relative gene expression levels. Briefly, by using ReverTra Ace reverse transcriptase (Toyobo Co., Osaka, Japan), 1 ng ~ 1 µg total RNA was reverse-transcribed into cDNA with oligo(dT)18, and then qPCR was performed on the QIAGEN’s Roter-Gene Q instrument by using Platinum SYBR Green qPCR SuperMix-UDG (Life Technologies, Cat. no.: 11733-038, CA, USA). In each reaction, 20 µL reaction mixtures containing 1 µL cDNA were incubated at 95℃ for 5 min, followed by 40 cycles of 95℃ for 10 s and 60℃ for 45 s. GAPDH was taken as an endogenous reference. 2−△△Ct method was used to calculate the differences of the expression level of genes in samples examined [21]. All experiments were run in triplicate and results were shown as the mean ± SD (n = 10). The primer sequences were available in Table 1.\n\nTable 1Primer sequences of qPCRGenePrimer sequence (5’-3’)\nNfkbie\nF: GTGGACTGGATGGAGATTCTTGR: TTTCCTGGTGGCTGGTAATG\nPlscr4\nF: CAGCTTGGGACACTAGGTTATTR: GGGAACTAAGGGCGTCATTT\nApoc1\nF: ACAAGGACAGGGTAGAGAAGAR: ACAGGAAGTGCGATGAAGAG\nLcn8\nF: GGGTAGAAGGCTTGTTCCTTACR: CTCTTTCTGAACCCACTGATCTT\nPrdx1\nF: TGTGTCCCACGGAGATCATTGCTTR: TGTTCATGGGTCCCAATCCTCCTT\nPrdx3\nF: TGGTGTCATCAAGCACCTGAGTGTR: AAGCTGTTGGACTTGGCTTGATCG\nGAPDH\nF: TGGGTGTGAACCACGAGAAR: GGCATGGACTGTGGTCATGA\n\nPrimer sequences of qPCR\nF: GTGGACTGGATGGAGATTCTTG\nR: TTTCCTGGTGGCTGGTAATG\nF: CAGCTTGGGACACTAGGTTATT\nR: GGGAACTAAGGGCGTCATTT\nF: ACAAGGACAGGGTAGAGAAGA\nR: ACAGGAAGTGCGATGAAGAG\nF: GGGTAGAAGGCTTGTTCCTTAC\nR: CTCTTTCTGAACCCACTGATCTT\nF: TGTGTCCCACGGAGATCATTGCTT\nR: TGTTCATGGGTCCCAATCCTCCTT\nF: TGGTGTCATCAAGCACCTGAGTGT\nR: AAGCTGTTGGACTTGGCTTGATCG\nF: TGGGTGTGAACCACGAGAA\nR: GGCATGGACTGTGGTCATGA\nReal-time quantitative PCR (qPCR) was performed to examine the relative gene expression levels. Briefly, by using ReverTra Ace reverse transcriptase (Toyobo Co., Osaka, Japan), 1 ng ~ 1 µg total RNA was reverse-transcribed into cDNA with oligo(dT)18, and then qPCR was performed on the QIAGEN’s Roter-Gene Q instrument by using Platinum SYBR Green qPCR SuperMix-UDG (Life Technologies, Cat. no.: 11733-038, CA, USA). In each reaction, 20 µL reaction mixtures containing 1 µL cDNA were incubated at 95℃ for 5 min, followed by 40 cycles of 95℃ for 10 s and 60℃ for 45 s. GAPDH was taken as an endogenous reference. 2−△△Ct method was used to calculate the differences of the expression level of genes in samples examined [21]. All experiments were run in triplicate and results were shown as the mean ± SD (n = 10). The primer sequences were available in Table 1.\n\nTable 1Primer sequences of qPCRGenePrimer sequence (5’-3’)\nNfkbie\nF: GTGGACTGGATGGAGATTCTTGR: TTTCCTGGTGGCTGGTAATG\nPlscr4\nF: CAGCTTGGGACACTAGGTTATTR: GGGAACTAAGGGCGTCATTT\nApoc1\nF: ACAAGGACAGGGTAGAGAAGAR: ACAGGAAGTGCGATGAAGAG\nLcn8\nF: GGGTAGAAGGCTTGTTCCTTACR: CTCTTTCTGAACCCACTGATCTT\nPrdx1\nF: TGTGTCCCACGGAGATCATTGCTTR: TGTTCATGGGTCCCAATCCTCCTT\nPrdx3\nF: TGGTGTCATCAAGCACCTGAGTGTR: AAGCTGTTGGACTTGGCTTGATCG\nGAPDH\nF: TGGGTGTGAACCACGAGAAR: GGCATGGACTGTGGTCATGA\n\nPrimer sequences of qPCR\nF: GTGGACTGGATGGAGATTCTTG\nR: TTTCCTGGTGGCTGGTAATG\nF: CAGCTTGGGACACTAGGTTATT\nR: GGGAACTAAGGGCGTCATTT\nF: ACAAGGACAGGGTAGAGAAGA\nR: ACAGGAAGTGCGATGAAGAG\nF: GGGTAGAAGGCTTGTTCCTTAC\nR: CTCTTTCTGAACCCACTGATCTT\nF: TGTGTCCCACGGAGATCATTGCTT\nR: TGTTCATGGGTCCCAATCCTCCTT\nF: TGGTGTCATCAAGCACCTGAGTGT\nR: AAGCTGTTGGACTTGGCTTGATCG\nF: TGGGTGTGAACCACGAGAA\nR: GGCATGGACTGTGGTCATGA\n[SUBTITLE] Identification of DEGs of digital profiling data [SUBSECTION] To identify the differentially expressed genes in the epididymis of rats after bilateral castration, a modified algorithm according to Audic S, et al. was used [22]. Suppose the number of clean tags corresponding to gene A was x, the expression level of each gene was only a small fraction of all genes expression level, therefore, p(x) follows the Poisson distribution. Furthermore, suppose the number of clean tags of sample 1 and 2 was N1 and N2, respectively, while the clean tags of gene A in sample 1 and 2 were x and y, respectively. The probability of gene A expression was equal in two samples could be calculated as the formula:\nP(y|x)=(\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\frac{{N}_{2}}{{N}_{1}}$$\\end{document})y\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\frac{\\left(x+y\\right)!}{{x!y!(1+{N}_{2}/{N}_{1})}^{(x+y+1)}}$$\\end{document}\nThe acquired p-value was controlled with false discovery rate (FDR) using Benjamini-Hochberg method [23]. Differentially expressed genes with FDR ≤ 0.001 and log2 fold-change ≥ 1 were extracted. The correlation between the two libraries was measured by the Pearson correlation coefficient.\nTo identify the differentially expressed genes in the epididymis of rats after bilateral castration, a modified algorithm according to Audic S, et al. was used [22]. Suppose the number of clean tags corresponding to gene A was x, the expression level of each gene was only a small fraction of all genes expression level, therefore, p(x) follows the Poisson distribution. Furthermore, suppose the number of clean tags of sample 1 and 2 was N1 and N2, respectively, while the clean tags of gene A in sample 1 and 2 were x and y, respectively. The probability of gene A expression was equal in two samples could be calculated as the formula:\nP(y|x)=(\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\frac{{N}_{2}}{{N}_{1}}$$\\end{document})y\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\frac{\\left(x+y\\right)!}{{x!y!(1+{N}_{2}/{N}_{1})}^{(x+y+1)}}$$\\end{document}\nThe acquired p-value was controlled with false discovery rate (FDR) using Benjamini-Hochberg method [23]. Differentially expressed genes with FDR ≤ 0.001 and log2 fold-change ≥ 1 were extracted. The correlation between the two libraries was measured by the Pearson correlation coefficient.\n[SUBTITLE] Identification of DEGs of microarray data [SUBSECTION] Online analysis tool GEO2R (https://www.ncbi.nlm.nih.gov/geo/geo2r) and R software (v4.0.5; https://www.r-project.org) were used for the identification of DEGs. The raw data from GSE35988 on GPL6480 and GSE32269 on GPL96 were normalized, transformed into expression values with GEO2R. Then empirical Bayes methods were applied to identify DEGs between CRPC and primary PCa with the limma package [24]. DEGs were defined with false discovery rate (FDR) < 0.001 and |log2(fold change)|>1 [25].\nOnline analysis tool GEO2R (https://www.ncbi.nlm.nih.gov/geo/geo2r) and R software (v4.0.5; https://www.r-project.org) were used for the identification of DEGs. The raw data from GSE35988 on GPL6480 and GSE32269 on GPL96 were normalized, transformed into expression values with GEO2R. Then empirical Bayes methods were applied to identify DEGs between CRPC and primary PCa with the limma package [24]. DEGs were defined with false discovery rate (FDR) < 0.001 and |log2(fold change)|>1 [25].\n[SUBTITLE] Functional enrichment and PPI network analysis of DEGs [SUBSECTION] The Database for Annotation, Visualization, and Integrated Discovery (DAVID; http://www.david.niaid.nih.gov) [26] and the R package clusterProfiler were used to perform GO and KEGG pathway analyses separately on CRPC-castration and CRPC-specific DEGs. Search Tool for the Retrieval of Interacting Genes/ Proteins (STRING; https://string-db.org) was used to construct PPI networks [27, 28]. Protein interactions with combined scores of > 0.15 were selected for the PPI network construction. Further, the Cytoscape software (v3.8.2) was utilized to calculate the node degree by MCODE and CytoHubba apps [29].\nThe Database for Annotation, Visualization, and Integrated Discovery (DAVID; http://www.david.niaid.nih.gov) [26] and the R package clusterProfiler were used to perform GO and KEGG pathway analyses separately on CRPC-castration and CRPC-specific DEGs. Search Tool for the Retrieval of Interacting Genes/ Proteins (STRING; https://string-db.org) was used to construct PPI networks [27, 28]. Protein interactions with combined scores of > 0.15 were selected for the PPI network construction. Further, the Cytoscape software (v3.8.2) was utilized to calculate the node degree by MCODE and CytoHubba apps [29].", "Two gene expression profiles (GSE35988, GSE32269) were obtained from Gene Expression Omnibus (GEO, https://www.ncbi.nlm.nih.gov/geo). GSE35988 contained 27 metastatic CPRC samples and 47 localized PCa samples, and GSE32269 was composed of 29 metastatic CPRC samples and 22 primary PCa samples.", "Twenty healthy and mature male Sprague-Dawley (SD) rats (12 weeks old and the body weight reached 350 ~ 400 g) were obtained from the Animal Research Centre of the Lv Ye Pharmaceutical Company (Yantai, China) and were housed at an environmental temperature of 20 ± 2 °C, receiving 12 h of light and 12 h of dark, with food and water provided ad libitum. Rats were randomly divided into two groups by using a random number Table (10 animals of each group), namely sham-operated control group (Con-EP) and castrated group (Cas-EP). Before castration treatment, rats were anesthetized by intraperitoneally injection of sodium pentobarbital (30 mg/kg body weight). Thereafter, the efferent ducts and the testicular arteries and veins were ligated, then the testes were removed from the ligation point, leaving the intact epididymides in the scrotal area. In the meantime, the sham operation was also taken for the control group. Analgesia was provided by administration of meloxicam Q 24 h (1 mg/kg PO or SC) (Metacam, Boehringer Ingelheim, St Joseph, MO) the day prior, the day of and the day following surgery. No obvious behavioral changes were observed in both control and the operation group. At the conclusion of the experiment, all the surviving rats were intraperitoneally anesthetized with sodium pentobarbital (30 mg/kg body weight) and their necks were dislocated for euthanasia. The whole epididymides were taken out from the castrated and sham-operated control rats after 7 days’ recovery and were immediately frozen in liquid nitrogen and stored at -80 ℃ before use. Protocols for the use of animals in these experiments were approved by the Research Animal Care and Use Committee of Yantai University (the approval reference number: YD.No20190916S0350210[315]) and were carried out in strict accordance with Guidelines for Ethical Review of Laboratory Animal Welfare of China. Ethical approval was obtained before the initiation of the research work and all efforts were made to minimize suffering.", "Total RNA was isolated from the epididymides from the control group and castrated group, respectively, by using RNAiso Plus reagent (TaKaRa, Dalian, China). The purity and quality of total RNA were evaluated by Agilent 2000 and were considered sufficient for subsequent library construction and sequencing. Tag libraries for the samples were prepared using the Illumina gene expression sample, Prep Kit. Each sample of 6 µg of the total RNA was extracted and purified by oligo (dT) magnetic bead adsorption. Primed by oligo (dT), mRNA bound to the bead was used as the template to synthesize a double-stranded cDNA. The 5’ ends of tags were digested with restriction enzyme NlaIII at CATG sites. The cDNA fragments with 3’ ends connected to Oligo(dT) beads were purified and the Illumina adaptor 1 was ligated to the 5’ ends. The junction of Illumina adaptor 1 and CATG site is the recognition site of MmeI, which is a type of endonuclease with separated recognition sites and digestion sites. It cuts at 17 bp downstream of the CATG site, producing tags with adaptor 1. After removing 3’ fragments with magnetic beads precipitation, Illumina adaptor 2 is ligated to the 3’ ends of tags, acquiring tags with different adaptors of both ends to form a tag library. After 15 cycles of linear PCR amplification, 105 bp fragments were purified by 6% TBE PAGE Gel electrophoresis. After denaturation, the single-chain molecules were fixed onto the Illumina Sequencing Chip (flowcell). Each molecule was grown into a single-molecule cluster sequencing template through Situ amplification. Then, they were sequenced with the method of sequencing by synthesis (SBS). Each tunnel generated millions of raw reads with a sequencing length of 49 bp.", "Five steps were needed to transform raw tags into clean tags: (1) removal of the 3’ adaptor to preserve the 21 nt long sequences; (2) removal of the empty reads (reads with only adaptor sequences but no tags); (3) removal of the low-quality tags (tags with unknown sequences); (4) removal of too long or too short tags, keeping the 21 nt tags; (5) removal of tags with only one copy number (probably because of sequencing error). To convert digital profiles to gene expression, the tag sequences were mapped to the reference genes of rat from NCBI, with no more than 1 nucleotide mismatch. Clean tags mapped to multiple loci were filtered, only unambiguous mapped tags were kept. To identify gene expression, the number of unambiguous tags for each gene was calculated and then normalized to the number of transcripts per million tags (TPM). Gene ontology was assigned to all mapped genes to annotate their possible functions.", "Real-time quantitative PCR (qPCR) was performed to examine the relative gene expression levels. Briefly, by using ReverTra Ace reverse transcriptase (Toyobo Co., Osaka, Japan), 1 ng ~ 1 µg total RNA was reverse-transcribed into cDNA with oligo(dT)18, and then qPCR was performed on the QIAGEN’s Roter-Gene Q instrument by using Platinum SYBR Green qPCR SuperMix-UDG (Life Technologies, Cat. no.: 11733-038, CA, USA). In each reaction, 20 µL reaction mixtures containing 1 µL cDNA were incubated at 95℃ for 5 min, followed by 40 cycles of 95℃ for 10 s and 60℃ for 45 s. GAPDH was taken as an endogenous reference. 2−△△Ct method was used to calculate the differences of the expression level of genes in samples examined [21]. All experiments were run in triplicate and results were shown as the mean ± SD (n = 10). The primer sequences were available in Table 1.\n\nTable 1Primer sequences of qPCRGenePrimer sequence (5’-3’)\nNfkbie\nF: GTGGACTGGATGGAGATTCTTGR: TTTCCTGGTGGCTGGTAATG\nPlscr4\nF: CAGCTTGGGACACTAGGTTATTR: GGGAACTAAGGGCGTCATTT\nApoc1\nF: ACAAGGACAGGGTAGAGAAGAR: ACAGGAAGTGCGATGAAGAG\nLcn8\nF: GGGTAGAAGGCTTGTTCCTTACR: CTCTTTCTGAACCCACTGATCTT\nPrdx1\nF: TGTGTCCCACGGAGATCATTGCTTR: TGTTCATGGGTCCCAATCCTCCTT\nPrdx3\nF: TGGTGTCATCAAGCACCTGAGTGTR: AAGCTGTTGGACTTGGCTTGATCG\nGAPDH\nF: TGGGTGTGAACCACGAGAAR: GGCATGGACTGTGGTCATGA\n\nPrimer sequences of qPCR\nF: GTGGACTGGATGGAGATTCTTG\nR: TTTCCTGGTGGCTGGTAATG\nF: CAGCTTGGGACACTAGGTTATT\nR: GGGAACTAAGGGCGTCATTT\nF: ACAAGGACAGGGTAGAGAAGA\nR: ACAGGAAGTGCGATGAAGAG\nF: GGGTAGAAGGCTTGTTCCTTAC\nR: CTCTTTCTGAACCCACTGATCTT\nF: TGTGTCCCACGGAGATCATTGCTT\nR: TGTTCATGGGTCCCAATCCTCCTT\nF: TGGTGTCATCAAGCACCTGAGTGT\nR: AAGCTGTTGGACTTGGCTTGATCG\nF: TGGGTGTGAACCACGAGAA\nR: GGCATGGACTGTGGTCATGA", "To identify the differentially expressed genes in the epididymis of rats after bilateral castration, a modified algorithm according to Audic S, et al. was used [22]. Suppose the number of clean tags corresponding to gene A was x, the expression level of each gene was only a small fraction of all genes expression level, therefore, p(x) follows the Poisson distribution. Furthermore, suppose the number of clean tags of sample 1 and 2 was N1 and N2, respectively, while the clean tags of gene A in sample 1 and 2 were x and y, respectively. The probability of gene A expression was equal in two samples could be calculated as the formula:\nP(y|x)=(\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\frac{{N}_{2}}{{N}_{1}}$$\\end{document})y\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\frac{\\left(x+y\\right)!}{{x!y!(1+{N}_{2}/{N}_{1})}^{(x+y+1)}}$$\\end{document}\nThe acquired p-value was controlled with false discovery rate (FDR) using Benjamini-Hochberg method [23]. Differentially expressed genes with FDR ≤ 0.001 and log2 fold-change ≥ 1 were extracted. The correlation between the two libraries was measured by the Pearson correlation coefficient.", "Online analysis tool GEO2R (https://www.ncbi.nlm.nih.gov/geo/geo2r) and R software (v4.0.5; https://www.r-project.org) were used for the identification of DEGs. The raw data from GSE35988 on GPL6480 and GSE32269 on GPL96 were normalized, transformed into expression values with GEO2R. Then empirical Bayes methods were applied to identify DEGs between CRPC and primary PCa with the limma package [24]. DEGs were defined with false discovery rate (FDR) < 0.001 and |log2(fold change)|>1 [25].", "The Database for Annotation, Visualization, and Integrated Discovery (DAVID; http://www.david.niaid.nih.gov) [26] and the R package clusterProfiler were used to perform GO and KEGG pathway analyses separately on CRPC-castration and CRPC-specific DEGs. Search Tool for the Retrieval of Interacting Genes/ Proteins (STRING; https://string-db.org) was used to construct PPI networks [27, 28]. Protein interactions with combined scores of > 0.15 were selected for the PPI network construction. Further, the Cytoscape software (v3.8.2) was utilized to calculate the node degree by MCODE and CytoHubba apps [29].", "To explore the transcriptional differences in the epididymis between Con-EP and Cas-EP rats, we obtained more than 4 million clean tags for each library. As demonstrated in Additional file 1, the distribution of total clean tags and distinct clean tags showed similar patterns in both DGE libraries, showing no bias between the library construction of Con-EP and Cas-EP. Around 3 million tags were mapped to genes for both Con-EP and Cas-EP, accounting for 62.89% and 70.32%, respectively.\nA total of 2,448 genes changed significantly between the two libraries. Among these genes, 1,632 were up-regulated while 816 were down-regulated after bilateral castration. More than 90% of the genes (2,274) were up- or down-regulated between 1.0 and 5.0 fold (data not shown). The Pearson correlation coefficient of the two libraries was only 0.658, reflecting the tremendous influence of castration on the gene expression profile of rat epididymis. We then performed qPCR of six selective genes which were up-regulated (Nfkbie, Plscr4 and Apoc1) or down-regulated (Lcn8, Prdx1 and Prdx3) to confirm the validity of the differentially expressed genes identified by DGE (n = 10). As expected, we obtained results that coincided with our sequencing data, confirming the authenticity of our sequencing results (Fig. 2).\n\nFig. 2Real-time fluorescent quantitative PCR analysis of differentially expressed genes before and after castration. a: down-regulated genes after bilateral castration, b: genes up-regulated after bilateral castration\n\nReal-time fluorescent quantitative PCR analysis of differentially expressed genes before and after castration. a: down-regulated genes after bilateral castration, b: genes up-regulated after bilateral castration", "To obtain potential CRPC driver genes dependent or independent of castration, we identified DEGs in the CRPC samples compared with the primary PCa samples. We obtained 1,904 up-regulated genes and 3,391 down-regulated genes (fold change > 1, P < 0.001) from GSE35988 (Fig. 3a), and 512 increased genes and 244 decreased genes (fold change > 1, P < 0.001) from GSE32269 (Fig. 3b). A total of 97 up-regulated genes (Fig. 3d) and 128 down-regulated genes (Fig. 3c) were shared by CRPC (GSE35988) and testis castration (data not shown), considered as CRPC DEGs related to castration (CRPC-castration). In contrast to testis castration, 120 up-regulated genes and 136 down-regulated genes changed only in CRPC (both GSE35988 and GSE32269) (Fig. 3e, data not shown), considered as CRPC-specific DEGs independent of castration.\n\nFig. 3Database analysis. a and b: Volcano plots of all genes in GSE35988 and GSE32269 Red dots represent genes with fold change ≥ 2 and P < 0.001, blue dots represent genes with fold change ≤ − 2 and P < 0.001, and the other dots represent the rest of genes with no statistically significant change in expression. FC, fold change. c, d and e: Venn diagrams showing the number of genes expressed as common and unique between the identified DEGs in DGE profiles of castrated rat and genes in the NCBI-gene database\n\nDatabase analysis. a and b: Volcano plots of all genes in GSE35988 and GSE32269 Red dots represent genes with fold change ≥ 2 and P < 0.001, blue dots represent genes with fold change ≤ − 2 and P < 0.001, and the other dots represent the rest of genes with no statistically significant change in expression. FC, fold change. c, d and e: Venn diagrams showing the number of genes expressed as common and unique between the identified DEGs in DGE profiles of castrated rat and genes in the NCBI-gene database\nNext, we explored the functional enrichment of these two lists of DEGs by using GO and KEGG analysis. The CRPC-specific DEGs significantly enriched cell division or mitotic processes (Fig. 4a), consistent with their oncogenic role in tumor cell proliferation. Concordantly, pathway analysis identified enrichment in the cell cycle pathway and focal adhesion pathway (Fig. 4b) (Additional file 2). On the other hand, GO enrichment of CRPC-castration DEGs (data not shown) showed that gland development, epithelial cell proliferation, and prostate gland development were substantially affected by castration (Fig. 5a). Pathway analysis showed that the pyruvate metabolism pathway was significantly enriched (Additional file 3).\n\nFig. 4Go and KEGG analysis. a: The top 10 results of GO functional analysis in CRPC-specific DEGs. b: KEGG functional analysis in CRPC-specific DEGs.\n\nGo and KEGG analysis. a: The top 10 results of GO functional analysis in CRPC-specific DEGs. b: KEGG functional analysis in CRPC-specific DEGs.\n\nFig. 5Go and PPI analysis. a: The top 10 results of GO functional analysis in CRPC-castration DEGs. b: Construction of PPI network of CRPC-castration DEGs.\n\nGo and PPI analysis. a: The top 10 results of GO functional analysis in CRPC-castration DEGs. b: Construction of PPI network of CRPC-castration DEGs.\nFurthermore, we used STRING to explore the protein-protein interaction (PPI) of the DEGs. CRPC-specific DEGs formed a PPI network with 208 nodes and 1353 edges. Among the 41 hub genes (node degree > 30) were chromatin remodeling factors EZH2 and PRC1, and cell cycle regulators CDCA3, CCNA2, CCNB2, CDKN3, CDK1, CCNB1, CDC20, MCM4, SMC4, CENPF, BUB1, BUB1B, NUSAP1, and NCAPG (Fig. 6). In contrast, CRPC-castration DEGs formed PPI networks with 171 nodes and sparse connections. The most connected sub-network with the hub gene ACTR10 was involved in innate immunity and cell polarity (Fig. 5b).\n\nFig. 6Top 41 of PPI network of CRPC-specific DEGs.\n\nTop 41 of PPI network of CRPC-specific DEGs.\nFinally, based on network and pathway analyses, we identified two key genes (NUSAP1 and NCAPG) among the top 41 nodes in the CRPC-specific PPI network, which participate in various pathways including chromosome segregation and nuclear division. Further analysis showed that, for both genes, higher expression was associated with shorter disease-free survival of patients (Fig. 7c and d). Interestingly, the expression of NUSAP1 and NCAPG differed between primary PCa and normal prostate tissues (Fig. 7a and b), which led us to hypothesize that they may have a role both in the initiation and in the progression of PCa.\n\nFig. 7Key genes analysis. a and b: Expression of the key genes NUSAP1 and NCAPG in TCGA with GEPIA. c and d: Disease-free survival in relation to the expression of the key genes (NUSAP1 and NCAPG) in TCGA.\n\nKey genes analysis. a and b: Expression of the key genes NUSAP1 and NCAPG in TCGA with GEPIA. c and d: Disease-free survival in relation to the expression of the key genes (NUSAP1 and NCAPG) in TCGA.", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Microarray data and DGE profiling", "Animals and castration procedure", "Library preparation and sequencing", "Tag annotation and gene expression level quantification", "Real-time quantitative PCR analysis of genes", "Identification of DEGs of digital profiling data", "Identification of DEGs of microarray data", "Functional enrichment and PPI network analysis of DEGs", "Results", "Dramatic transcriptional changes in rats’ epididymis after bilateral castration", "CRPC-castration genes were enriched in cell proliferation and prostate gland development", "Discussion", "Conclusion", "Electronic supplementary material", "" ]
[ "One of the most common malignancies in man is prostate cancer (PCa), which also causes the second highest deaths in man. Absolutely, PCa arises as an androgen driven disease [1]. Therefore androgen deprivation therapy (ADT) was adopted as the major treatment for metastatic hormone-sensitive PCa, by suppressing circulating testosterone to decrease its level to “castrate levels” ( < = 50 ng/dL) [2] and induce of apoptosis [3]. However, although the initial treatment effect is significant, almost all PCa patients ultimately progress and castration resistant prostate cancer (CRPC) ensue, which is associated with high mortality rates [4]. CRPC represents a particular stage in the continuum of the disease and is defined by continuous rise of prostate specific antigen levels in serum or/and progression of metastatic spread in the setting of castrate levels of testosterone [5–7].\nNowadays, suppression of androgen receptor (AR) signaling through ADT has remained the first-line treatment for men with advanced PCa. ADT includes surgical or medical castration by using luteinizing hormone-releasing hormone (LHRH) agonists or antagonists with or without anti-androgen drugs [8]. Exposure to LHRH agonists downregulates the LHRH receptor, decreasing LH release and inhibiting testosterone production, while LHRH antagonists, such as cetrorelix and abarelix, directly inhibit the LHRH receptor, resulting in the decreased production of LH and testosterone. Surgical bilaterial castration also decreases testosterone levels by removing the testes, the source of its production organ, although surgical castration has been practically set aside for some time, given the spread of ADT [9]. However, by using LHRH agonists or antagonists, for nearly all patients, after 1–2 years remission, cancer cells become resistant with the emergence of metastatic CRPC [10]. Today’s standard of care for advanced PCa includes gonadotropin-releasing hormone agonists (leuprolide), second-generation nonsteroidal AR antagonists (apalutamide, enzalutamide and darolutamide) and the androgen biosynthesis inhibitor abiraterone [11]. Currently, with minimal additional toxicity, abiraterone acetate has been proved to be a well-tolerated, convenient and effective treatment option [12].\nBesides the above mentioned drugs for the treatment of CRPC, AR-V7, the androgen receptor isoform encoded by splice variant 7, has become an attractive biomarker predicting the effect of androgen signaling inhibitor [10, 13]. AR-V7 lacks the ligand binding domain and is the target of enzalutamide and abiraterone. It is supposed that detection of AR-V7 in circulating tumor cells from patients with CPRC might be closely related to resistance to enzalutamide and abiraterone [14].\nMany mechanisms of CRPC have been proposed, including reactivation of androgen synthesis, up-regulation of genes related to androgen metabolism, and reprogramming of the tumor microenvironment [13]. However, CRPC treatment is challenging [15]. Thus, identifying effective molecular biomarkers and therapeutic targets of CRPC is of ultra-importance for PCa prediction and treatment. Previously, Microarray techniques have been used to identify biomarkers and targets in CRPC. For example, D’Antonio et al. [16] identified pathways of androgen independence of CRPC by comparing PCa cells (androgen-independent) with LNCaP cells (androgen-dependent). Terada et al. [17] found that prostaglandin E2 receptor subtype 4 (EP4) is a central gene in CRPC by comparing a mouse xenograft model of PCa with later-derived CRPC.\nThe biological functions of the mammalian epididymis, an important male accessory gland, are substantially affected by androgen [18]. The mammalian epididymis is an important male accessory gland with many biological functions, including maturation and concentration of spermatozoa produced by testis, secretion and resorption of different molecules and proteins, and storage of spermatozoa. The development and normal functions of the epididymis are regulated mainly by androgens and testicular factors [19]. Bilateral castration led to systematic changes of epididymal gene expression profile [20]. It would be useful to identify critical androgen-related pathways in the epididymis to help understand CRPC and identify biomarkers or targets, thus providing opportunities to combat the disease.\nCompared with microarray, digital gene expression profiling provides absolute gene expression measurements and an unbiased view of the whole transcriptome with greater precision and sensitivity. Here we identified differentially expressed genes in the epididymides of sham-operated control and bilateral castrated male mature rats on the 7th day after surgery by using the DGE system on the whole-genome scale, as previous studies have demonstrated that gene expression profile is most likely to reflects transcriptional changes in surviving epididymal cells on the 7th day of post castration because apoptotic cell death is no longer detected at this moment [20]. Then we combined our data with two published datasets to identify differentially expressed genes (DEGs) between CRPC and primary PCa. We obtained DEGs of either dependent (CRPC-castration) or independent (CRPC-specific) of castration to investigate the role of castration in CRPC. Functional enrichment analysis, protein-protein interaction (PPI) network, and survival analysis of these DEGs showed distinct functional roles of CRPC-castration and CRPC-specific genes (Fig. 1). CRPC-specific DEGs were mainly enriched in cell proliferation, while CRPC-castration genes were associated with prostate gland development. In summary, our study provided insights into a deeper understanding of CRPC pathogenesis.\n\nFig. 1Flow diagram showing an overview of the study\n\nFlow diagram showing an overview of the study", "[SUBTITLE] Microarray data and DGE profiling [SUBSECTION] Two gene expression profiles (GSE35988, GSE32269) were obtained from Gene Expression Omnibus (GEO, https://www.ncbi.nlm.nih.gov/geo). GSE35988 contained 27 metastatic CPRC samples and 47 localized PCa samples, and GSE32269 was composed of 29 metastatic CPRC samples and 22 primary PCa samples.\nTwo gene expression profiles (GSE35988, GSE32269) were obtained from Gene Expression Omnibus (GEO, https://www.ncbi.nlm.nih.gov/geo). GSE35988 contained 27 metastatic CPRC samples and 47 localized PCa samples, and GSE32269 was composed of 29 metastatic CPRC samples and 22 primary PCa samples.\n[SUBTITLE] Animals and castration procedure [SUBSECTION] Twenty healthy and mature male Sprague-Dawley (SD) rats (12 weeks old and the body weight reached 350 ~ 400 g) were obtained from the Animal Research Centre of the Lv Ye Pharmaceutical Company (Yantai, China) and were housed at an environmental temperature of 20 ± 2 °C, receiving 12 h of light and 12 h of dark, with food and water provided ad libitum. Rats were randomly divided into two groups by using a random number Table (10 animals of each group), namely sham-operated control group (Con-EP) and castrated group (Cas-EP). Before castration treatment, rats were anesthetized by intraperitoneally injection of sodium pentobarbital (30 mg/kg body weight). Thereafter, the efferent ducts and the testicular arteries and veins were ligated, then the testes were removed from the ligation point, leaving the intact epididymides in the scrotal area. In the meantime, the sham operation was also taken for the control group. Analgesia was provided by administration of meloxicam Q 24 h (1 mg/kg PO or SC) (Metacam, Boehringer Ingelheim, St Joseph, MO) the day prior, the day of and the day following surgery. No obvious behavioral changes were observed in both control and the operation group. At the conclusion of the experiment, all the surviving rats were intraperitoneally anesthetized with sodium pentobarbital (30 mg/kg body weight) and their necks were dislocated for euthanasia. The whole epididymides were taken out from the castrated and sham-operated control rats after 7 days’ recovery and were immediately frozen in liquid nitrogen and stored at -80 ℃ before use. Protocols for the use of animals in these experiments were approved by the Research Animal Care and Use Committee of Yantai University (the approval reference number: YD.No20190916S0350210[315]) and were carried out in strict accordance with Guidelines for Ethical Review of Laboratory Animal Welfare of China. Ethical approval was obtained before the initiation of the research work and all efforts were made to minimize suffering.\nTwenty healthy and mature male Sprague-Dawley (SD) rats (12 weeks old and the body weight reached 350 ~ 400 g) were obtained from the Animal Research Centre of the Lv Ye Pharmaceutical Company (Yantai, China) and were housed at an environmental temperature of 20 ± 2 °C, receiving 12 h of light and 12 h of dark, with food and water provided ad libitum. Rats were randomly divided into two groups by using a random number Table (10 animals of each group), namely sham-operated control group (Con-EP) and castrated group (Cas-EP). Before castration treatment, rats were anesthetized by intraperitoneally injection of sodium pentobarbital (30 mg/kg body weight). Thereafter, the efferent ducts and the testicular arteries and veins were ligated, then the testes were removed from the ligation point, leaving the intact epididymides in the scrotal area. In the meantime, the sham operation was also taken for the control group. Analgesia was provided by administration of meloxicam Q 24 h (1 mg/kg PO or SC) (Metacam, Boehringer Ingelheim, St Joseph, MO) the day prior, the day of and the day following surgery. No obvious behavioral changes were observed in both control and the operation group. At the conclusion of the experiment, all the surviving rats were intraperitoneally anesthetized with sodium pentobarbital (30 mg/kg body weight) and their necks were dislocated for euthanasia. The whole epididymides were taken out from the castrated and sham-operated control rats after 7 days’ recovery and were immediately frozen in liquid nitrogen and stored at -80 ℃ before use. Protocols for the use of animals in these experiments were approved by the Research Animal Care and Use Committee of Yantai University (the approval reference number: YD.No20190916S0350210[315]) and were carried out in strict accordance with Guidelines for Ethical Review of Laboratory Animal Welfare of China. Ethical approval was obtained before the initiation of the research work and all efforts were made to minimize suffering.\n[SUBTITLE] Library preparation and sequencing [SUBSECTION] Total RNA was isolated from the epididymides from the control group and castrated group, respectively, by using RNAiso Plus reagent (TaKaRa, Dalian, China). The purity and quality of total RNA were evaluated by Agilent 2000 and were considered sufficient for subsequent library construction and sequencing. Tag libraries for the samples were prepared using the Illumina gene expression sample, Prep Kit. Each sample of 6 µg of the total RNA was extracted and purified by oligo (dT) magnetic bead adsorption. Primed by oligo (dT), mRNA bound to the bead was used as the template to synthesize a double-stranded cDNA. The 5’ ends of tags were digested with restriction enzyme NlaIII at CATG sites. The cDNA fragments with 3’ ends connected to Oligo(dT) beads were purified and the Illumina adaptor 1 was ligated to the 5’ ends. The junction of Illumina adaptor 1 and CATG site is the recognition site of MmeI, which is a type of endonuclease with separated recognition sites and digestion sites. It cuts at 17 bp downstream of the CATG site, producing tags with adaptor 1. After removing 3’ fragments with magnetic beads precipitation, Illumina adaptor 2 is ligated to the 3’ ends of tags, acquiring tags with different adaptors of both ends to form a tag library. After 15 cycles of linear PCR amplification, 105 bp fragments were purified by 6% TBE PAGE Gel electrophoresis. After denaturation, the single-chain molecules were fixed onto the Illumina Sequencing Chip (flowcell). Each molecule was grown into a single-molecule cluster sequencing template through Situ amplification. Then, they were sequenced with the method of sequencing by synthesis (SBS). Each tunnel generated millions of raw reads with a sequencing length of 49 bp.\nTotal RNA was isolated from the epididymides from the control group and castrated group, respectively, by using RNAiso Plus reagent (TaKaRa, Dalian, China). The purity and quality of total RNA were evaluated by Agilent 2000 and were considered sufficient for subsequent library construction and sequencing. Tag libraries for the samples were prepared using the Illumina gene expression sample, Prep Kit. Each sample of 6 µg of the total RNA was extracted and purified by oligo (dT) magnetic bead adsorption. Primed by oligo (dT), mRNA bound to the bead was used as the template to synthesize a double-stranded cDNA. The 5’ ends of tags were digested with restriction enzyme NlaIII at CATG sites. The cDNA fragments with 3’ ends connected to Oligo(dT) beads were purified and the Illumina adaptor 1 was ligated to the 5’ ends. The junction of Illumina adaptor 1 and CATG site is the recognition site of MmeI, which is a type of endonuclease with separated recognition sites and digestion sites. It cuts at 17 bp downstream of the CATG site, producing tags with adaptor 1. After removing 3’ fragments with magnetic beads precipitation, Illumina adaptor 2 is ligated to the 3’ ends of tags, acquiring tags with different adaptors of both ends to form a tag library. After 15 cycles of linear PCR amplification, 105 bp fragments were purified by 6% TBE PAGE Gel electrophoresis. After denaturation, the single-chain molecules were fixed onto the Illumina Sequencing Chip (flowcell). Each molecule was grown into a single-molecule cluster sequencing template through Situ amplification. Then, they were sequenced with the method of sequencing by synthesis (SBS). Each tunnel generated millions of raw reads with a sequencing length of 49 bp.\n[SUBTITLE] Tag annotation and gene expression level quantification [SUBSECTION] Five steps were needed to transform raw tags into clean tags: (1) removal of the 3’ adaptor to preserve the 21 nt long sequences; (2) removal of the empty reads (reads with only adaptor sequences but no tags); (3) removal of the low-quality tags (tags with unknown sequences); (4) removal of too long or too short tags, keeping the 21 nt tags; (5) removal of tags with only one copy number (probably because of sequencing error). To convert digital profiles to gene expression, the tag sequences were mapped to the reference genes of rat from NCBI, with no more than 1 nucleotide mismatch. Clean tags mapped to multiple loci were filtered, only unambiguous mapped tags were kept. To identify gene expression, the number of unambiguous tags for each gene was calculated and then normalized to the number of transcripts per million tags (TPM). Gene ontology was assigned to all mapped genes to annotate their possible functions.\nFive steps were needed to transform raw tags into clean tags: (1) removal of the 3’ adaptor to preserve the 21 nt long sequences; (2) removal of the empty reads (reads with only adaptor sequences but no tags); (3) removal of the low-quality tags (tags with unknown sequences); (4) removal of too long or too short tags, keeping the 21 nt tags; (5) removal of tags with only one copy number (probably because of sequencing error). To convert digital profiles to gene expression, the tag sequences were mapped to the reference genes of rat from NCBI, with no more than 1 nucleotide mismatch. Clean tags mapped to multiple loci were filtered, only unambiguous mapped tags were kept. To identify gene expression, the number of unambiguous tags for each gene was calculated and then normalized to the number of transcripts per million tags (TPM). Gene ontology was assigned to all mapped genes to annotate their possible functions.\n[SUBTITLE] Real-time quantitative PCR analysis of genes [SUBSECTION] Real-time quantitative PCR (qPCR) was performed to examine the relative gene expression levels. Briefly, by using ReverTra Ace reverse transcriptase (Toyobo Co., Osaka, Japan), 1 ng ~ 1 µg total RNA was reverse-transcribed into cDNA with oligo(dT)18, and then qPCR was performed on the QIAGEN’s Roter-Gene Q instrument by using Platinum SYBR Green qPCR SuperMix-UDG (Life Technologies, Cat. no.: 11733-038, CA, USA). In each reaction, 20 µL reaction mixtures containing 1 µL cDNA were incubated at 95℃ for 5 min, followed by 40 cycles of 95℃ for 10 s and 60℃ for 45 s. GAPDH was taken as an endogenous reference. 2−△△Ct method was used to calculate the differences of the expression level of genes in samples examined [21]. All experiments were run in triplicate and results were shown as the mean ± SD (n = 10). The primer sequences were available in Table 1.\n\nTable 1Primer sequences of qPCRGenePrimer sequence (5’-3’)\nNfkbie\nF: GTGGACTGGATGGAGATTCTTGR: TTTCCTGGTGGCTGGTAATG\nPlscr4\nF: CAGCTTGGGACACTAGGTTATTR: GGGAACTAAGGGCGTCATTT\nApoc1\nF: ACAAGGACAGGGTAGAGAAGAR: ACAGGAAGTGCGATGAAGAG\nLcn8\nF: GGGTAGAAGGCTTGTTCCTTACR: CTCTTTCTGAACCCACTGATCTT\nPrdx1\nF: TGTGTCCCACGGAGATCATTGCTTR: TGTTCATGGGTCCCAATCCTCCTT\nPrdx3\nF: TGGTGTCATCAAGCACCTGAGTGTR: AAGCTGTTGGACTTGGCTTGATCG\nGAPDH\nF: TGGGTGTGAACCACGAGAAR: GGCATGGACTGTGGTCATGA\n\nPrimer sequences of qPCR\nF: GTGGACTGGATGGAGATTCTTG\nR: TTTCCTGGTGGCTGGTAATG\nF: CAGCTTGGGACACTAGGTTATT\nR: GGGAACTAAGGGCGTCATTT\nF: ACAAGGACAGGGTAGAGAAGA\nR: ACAGGAAGTGCGATGAAGAG\nF: GGGTAGAAGGCTTGTTCCTTAC\nR: CTCTTTCTGAACCCACTGATCTT\nF: TGTGTCCCACGGAGATCATTGCTT\nR: TGTTCATGGGTCCCAATCCTCCTT\nF: TGGTGTCATCAAGCACCTGAGTGT\nR: AAGCTGTTGGACTTGGCTTGATCG\nF: TGGGTGTGAACCACGAGAA\nR: GGCATGGACTGTGGTCATGA\nReal-time quantitative PCR (qPCR) was performed to examine the relative gene expression levels. Briefly, by using ReverTra Ace reverse transcriptase (Toyobo Co., Osaka, Japan), 1 ng ~ 1 µg total RNA was reverse-transcribed into cDNA with oligo(dT)18, and then qPCR was performed on the QIAGEN’s Roter-Gene Q instrument by using Platinum SYBR Green qPCR SuperMix-UDG (Life Technologies, Cat. no.: 11733-038, CA, USA). In each reaction, 20 µL reaction mixtures containing 1 µL cDNA were incubated at 95℃ for 5 min, followed by 40 cycles of 95℃ for 10 s and 60℃ for 45 s. GAPDH was taken as an endogenous reference. 2−△△Ct method was used to calculate the differences of the expression level of genes in samples examined [21]. All experiments were run in triplicate and results were shown as the mean ± SD (n = 10). The primer sequences were available in Table 1.\n\nTable 1Primer sequences of qPCRGenePrimer sequence (5’-3’)\nNfkbie\nF: GTGGACTGGATGGAGATTCTTGR: TTTCCTGGTGGCTGGTAATG\nPlscr4\nF: CAGCTTGGGACACTAGGTTATTR: GGGAACTAAGGGCGTCATTT\nApoc1\nF: ACAAGGACAGGGTAGAGAAGAR: ACAGGAAGTGCGATGAAGAG\nLcn8\nF: GGGTAGAAGGCTTGTTCCTTACR: CTCTTTCTGAACCCACTGATCTT\nPrdx1\nF: TGTGTCCCACGGAGATCATTGCTTR: TGTTCATGGGTCCCAATCCTCCTT\nPrdx3\nF: TGGTGTCATCAAGCACCTGAGTGTR: AAGCTGTTGGACTTGGCTTGATCG\nGAPDH\nF: TGGGTGTGAACCACGAGAAR: GGCATGGACTGTGGTCATGA\n\nPrimer sequences of qPCR\nF: GTGGACTGGATGGAGATTCTTG\nR: TTTCCTGGTGGCTGGTAATG\nF: CAGCTTGGGACACTAGGTTATT\nR: GGGAACTAAGGGCGTCATTT\nF: ACAAGGACAGGGTAGAGAAGA\nR: ACAGGAAGTGCGATGAAGAG\nF: GGGTAGAAGGCTTGTTCCTTAC\nR: CTCTTTCTGAACCCACTGATCTT\nF: TGTGTCCCACGGAGATCATTGCTT\nR: TGTTCATGGGTCCCAATCCTCCTT\nF: TGGTGTCATCAAGCACCTGAGTGT\nR: AAGCTGTTGGACTTGGCTTGATCG\nF: TGGGTGTGAACCACGAGAA\nR: GGCATGGACTGTGGTCATGA\n[SUBTITLE] Identification of DEGs of digital profiling data [SUBSECTION] To identify the differentially expressed genes in the epididymis of rats after bilateral castration, a modified algorithm according to Audic S, et al. was used [22]. Suppose the number of clean tags corresponding to gene A was x, the expression level of each gene was only a small fraction of all genes expression level, therefore, p(x) follows the Poisson distribution. Furthermore, suppose the number of clean tags of sample 1 and 2 was N1 and N2, respectively, while the clean tags of gene A in sample 1 and 2 were x and y, respectively. The probability of gene A expression was equal in two samples could be calculated as the formula:\nP(y|x)=(\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\frac{{N}_{2}}{{N}_{1}}$$\\end{document})y\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\frac{\\left(x+y\\right)!}{{x!y!(1+{N}_{2}/{N}_{1})}^{(x+y+1)}}$$\\end{document}\nThe acquired p-value was controlled with false discovery rate (FDR) using Benjamini-Hochberg method [23]. Differentially expressed genes with FDR ≤ 0.001 and log2 fold-change ≥ 1 were extracted. The correlation between the two libraries was measured by the Pearson correlation coefficient.\nTo identify the differentially expressed genes in the epididymis of rats after bilateral castration, a modified algorithm according to Audic S, et al. was used [22]. Suppose the number of clean tags corresponding to gene A was x, the expression level of each gene was only a small fraction of all genes expression level, therefore, p(x) follows the Poisson distribution. Furthermore, suppose the number of clean tags of sample 1 and 2 was N1 and N2, respectively, while the clean tags of gene A in sample 1 and 2 were x and y, respectively. The probability of gene A expression was equal in two samples could be calculated as the formula:\nP(y|x)=(\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\frac{{N}_{2}}{{N}_{1}}$$\\end{document})y\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\frac{\\left(x+y\\right)!}{{x!y!(1+{N}_{2}/{N}_{1})}^{(x+y+1)}}$$\\end{document}\nThe acquired p-value was controlled with false discovery rate (FDR) using Benjamini-Hochberg method [23]. Differentially expressed genes with FDR ≤ 0.001 and log2 fold-change ≥ 1 were extracted. The correlation between the two libraries was measured by the Pearson correlation coefficient.\n[SUBTITLE] Identification of DEGs of microarray data [SUBSECTION] Online analysis tool GEO2R (https://www.ncbi.nlm.nih.gov/geo/geo2r) and R software (v4.0.5; https://www.r-project.org) were used for the identification of DEGs. The raw data from GSE35988 on GPL6480 and GSE32269 on GPL96 were normalized, transformed into expression values with GEO2R. Then empirical Bayes methods were applied to identify DEGs between CRPC and primary PCa with the limma package [24]. DEGs were defined with false discovery rate (FDR) < 0.001 and |log2(fold change)|>1 [25].\nOnline analysis tool GEO2R (https://www.ncbi.nlm.nih.gov/geo/geo2r) and R software (v4.0.5; https://www.r-project.org) were used for the identification of DEGs. The raw data from GSE35988 on GPL6480 and GSE32269 on GPL96 were normalized, transformed into expression values with GEO2R. Then empirical Bayes methods were applied to identify DEGs between CRPC and primary PCa with the limma package [24]. DEGs were defined with false discovery rate (FDR) < 0.001 and |log2(fold change)|>1 [25].\n[SUBTITLE] Functional enrichment and PPI network analysis of DEGs [SUBSECTION] The Database for Annotation, Visualization, and Integrated Discovery (DAVID; http://www.david.niaid.nih.gov) [26] and the R package clusterProfiler were used to perform GO and KEGG pathway analyses separately on CRPC-castration and CRPC-specific DEGs. Search Tool for the Retrieval of Interacting Genes/ Proteins (STRING; https://string-db.org) was used to construct PPI networks [27, 28]. Protein interactions with combined scores of > 0.15 were selected for the PPI network construction. Further, the Cytoscape software (v3.8.2) was utilized to calculate the node degree by MCODE and CytoHubba apps [29].\nThe Database for Annotation, Visualization, and Integrated Discovery (DAVID; http://www.david.niaid.nih.gov) [26] and the R package clusterProfiler were used to perform GO and KEGG pathway analyses separately on CRPC-castration and CRPC-specific DEGs. Search Tool for the Retrieval of Interacting Genes/ Proteins (STRING; https://string-db.org) was used to construct PPI networks [27, 28]. Protein interactions with combined scores of > 0.15 were selected for the PPI network construction. Further, the Cytoscape software (v3.8.2) was utilized to calculate the node degree by MCODE and CytoHubba apps [29].", "Two gene expression profiles (GSE35988, GSE32269) were obtained from Gene Expression Omnibus (GEO, https://www.ncbi.nlm.nih.gov/geo). GSE35988 contained 27 metastatic CPRC samples and 47 localized PCa samples, and GSE32269 was composed of 29 metastatic CPRC samples and 22 primary PCa samples.", "Twenty healthy and mature male Sprague-Dawley (SD) rats (12 weeks old and the body weight reached 350 ~ 400 g) were obtained from the Animal Research Centre of the Lv Ye Pharmaceutical Company (Yantai, China) and were housed at an environmental temperature of 20 ± 2 °C, receiving 12 h of light and 12 h of dark, with food and water provided ad libitum. Rats were randomly divided into two groups by using a random number Table (10 animals of each group), namely sham-operated control group (Con-EP) and castrated group (Cas-EP). Before castration treatment, rats were anesthetized by intraperitoneally injection of sodium pentobarbital (30 mg/kg body weight). Thereafter, the efferent ducts and the testicular arteries and veins were ligated, then the testes were removed from the ligation point, leaving the intact epididymides in the scrotal area. In the meantime, the sham operation was also taken for the control group. Analgesia was provided by administration of meloxicam Q 24 h (1 mg/kg PO or SC) (Metacam, Boehringer Ingelheim, St Joseph, MO) the day prior, the day of and the day following surgery. No obvious behavioral changes were observed in both control and the operation group. At the conclusion of the experiment, all the surviving rats were intraperitoneally anesthetized with sodium pentobarbital (30 mg/kg body weight) and their necks were dislocated for euthanasia. The whole epididymides were taken out from the castrated and sham-operated control rats after 7 days’ recovery and were immediately frozen in liquid nitrogen and stored at -80 ℃ before use. Protocols for the use of animals in these experiments were approved by the Research Animal Care and Use Committee of Yantai University (the approval reference number: YD.No20190916S0350210[315]) and were carried out in strict accordance with Guidelines for Ethical Review of Laboratory Animal Welfare of China. Ethical approval was obtained before the initiation of the research work and all efforts were made to minimize suffering.", "Total RNA was isolated from the epididymides from the control group and castrated group, respectively, by using RNAiso Plus reagent (TaKaRa, Dalian, China). The purity and quality of total RNA were evaluated by Agilent 2000 and were considered sufficient for subsequent library construction and sequencing. Tag libraries for the samples were prepared using the Illumina gene expression sample, Prep Kit. Each sample of 6 µg of the total RNA was extracted and purified by oligo (dT) magnetic bead adsorption. Primed by oligo (dT), mRNA bound to the bead was used as the template to synthesize a double-stranded cDNA. The 5’ ends of tags were digested with restriction enzyme NlaIII at CATG sites. The cDNA fragments with 3’ ends connected to Oligo(dT) beads were purified and the Illumina adaptor 1 was ligated to the 5’ ends. The junction of Illumina adaptor 1 and CATG site is the recognition site of MmeI, which is a type of endonuclease with separated recognition sites and digestion sites. It cuts at 17 bp downstream of the CATG site, producing tags with adaptor 1. After removing 3’ fragments with magnetic beads precipitation, Illumina adaptor 2 is ligated to the 3’ ends of tags, acquiring tags with different adaptors of both ends to form a tag library. After 15 cycles of linear PCR amplification, 105 bp fragments were purified by 6% TBE PAGE Gel electrophoresis. After denaturation, the single-chain molecules were fixed onto the Illumina Sequencing Chip (flowcell). Each molecule was grown into a single-molecule cluster sequencing template through Situ amplification. Then, they were sequenced with the method of sequencing by synthesis (SBS). Each tunnel generated millions of raw reads with a sequencing length of 49 bp.", "Five steps were needed to transform raw tags into clean tags: (1) removal of the 3’ adaptor to preserve the 21 nt long sequences; (2) removal of the empty reads (reads with only adaptor sequences but no tags); (3) removal of the low-quality tags (tags with unknown sequences); (4) removal of too long or too short tags, keeping the 21 nt tags; (5) removal of tags with only one copy number (probably because of sequencing error). To convert digital profiles to gene expression, the tag sequences were mapped to the reference genes of rat from NCBI, with no more than 1 nucleotide mismatch. Clean tags mapped to multiple loci were filtered, only unambiguous mapped tags were kept. To identify gene expression, the number of unambiguous tags for each gene was calculated and then normalized to the number of transcripts per million tags (TPM). Gene ontology was assigned to all mapped genes to annotate their possible functions.", "Real-time quantitative PCR (qPCR) was performed to examine the relative gene expression levels. Briefly, by using ReverTra Ace reverse transcriptase (Toyobo Co., Osaka, Japan), 1 ng ~ 1 µg total RNA was reverse-transcribed into cDNA with oligo(dT)18, and then qPCR was performed on the QIAGEN’s Roter-Gene Q instrument by using Platinum SYBR Green qPCR SuperMix-UDG (Life Technologies, Cat. no.: 11733-038, CA, USA). In each reaction, 20 µL reaction mixtures containing 1 µL cDNA were incubated at 95℃ for 5 min, followed by 40 cycles of 95℃ for 10 s and 60℃ for 45 s. GAPDH was taken as an endogenous reference. 2−△△Ct method was used to calculate the differences of the expression level of genes in samples examined [21]. All experiments were run in triplicate and results were shown as the mean ± SD (n = 10). The primer sequences were available in Table 1.\n\nTable 1Primer sequences of qPCRGenePrimer sequence (5’-3’)\nNfkbie\nF: GTGGACTGGATGGAGATTCTTGR: TTTCCTGGTGGCTGGTAATG\nPlscr4\nF: CAGCTTGGGACACTAGGTTATTR: GGGAACTAAGGGCGTCATTT\nApoc1\nF: ACAAGGACAGGGTAGAGAAGAR: ACAGGAAGTGCGATGAAGAG\nLcn8\nF: GGGTAGAAGGCTTGTTCCTTACR: CTCTTTCTGAACCCACTGATCTT\nPrdx1\nF: TGTGTCCCACGGAGATCATTGCTTR: TGTTCATGGGTCCCAATCCTCCTT\nPrdx3\nF: TGGTGTCATCAAGCACCTGAGTGTR: AAGCTGTTGGACTTGGCTTGATCG\nGAPDH\nF: TGGGTGTGAACCACGAGAAR: GGCATGGACTGTGGTCATGA\n\nPrimer sequences of qPCR\nF: GTGGACTGGATGGAGATTCTTG\nR: TTTCCTGGTGGCTGGTAATG\nF: CAGCTTGGGACACTAGGTTATT\nR: GGGAACTAAGGGCGTCATTT\nF: ACAAGGACAGGGTAGAGAAGA\nR: ACAGGAAGTGCGATGAAGAG\nF: GGGTAGAAGGCTTGTTCCTTAC\nR: CTCTTTCTGAACCCACTGATCTT\nF: TGTGTCCCACGGAGATCATTGCTT\nR: TGTTCATGGGTCCCAATCCTCCTT\nF: TGGTGTCATCAAGCACCTGAGTGT\nR: AAGCTGTTGGACTTGGCTTGATCG\nF: TGGGTGTGAACCACGAGAA\nR: GGCATGGACTGTGGTCATGA", "To identify the differentially expressed genes in the epididymis of rats after bilateral castration, a modified algorithm according to Audic S, et al. was used [22]. Suppose the number of clean tags corresponding to gene A was x, the expression level of each gene was only a small fraction of all genes expression level, therefore, p(x) follows the Poisson distribution. Furthermore, suppose the number of clean tags of sample 1 and 2 was N1 and N2, respectively, while the clean tags of gene A in sample 1 and 2 were x and y, respectively. The probability of gene A expression was equal in two samples could be calculated as the formula:\nP(y|x)=(\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\frac{{N}_{2}}{{N}_{1}}$$\\end{document})y\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\frac{\\left(x+y\\right)!}{{x!y!(1+{N}_{2}/{N}_{1})}^{(x+y+1)}}$$\\end{document}\nThe acquired p-value was controlled with false discovery rate (FDR) using Benjamini-Hochberg method [23]. Differentially expressed genes with FDR ≤ 0.001 and log2 fold-change ≥ 1 were extracted. The correlation between the two libraries was measured by the Pearson correlation coefficient.", "Online analysis tool GEO2R (https://www.ncbi.nlm.nih.gov/geo/geo2r) and R software (v4.0.5; https://www.r-project.org) were used for the identification of DEGs. The raw data from GSE35988 on GPL6480 and GSE32269 on GPL96 were normalized, transformed into expression values with GEO2R. Then empirical Bayes methods were applied to identify DEGs between CRPC and primary PCa with the limma package [24]. DEGs were defined with false discovery rate (FDR) < 0.001 and |log2(fold change)|>1 [25].", "The Database for Annotation, Visualization, and Integrated Discovery (DAVID; http://www.david.niaid.nih.gov) [26] and the R package clusterProfiler were used to perform GO and KEGG pathway analyses separately on CRPC-castration and CRPC-specific DEGs. Search Tool for the Retrieval of Interacting Genes/ Proteins (STRING; https://string-db.org) was used to construct PPI networks [27, 28]. Protein interactions with combined scores of > 0.15 were selected for the PPI network construction. Further, the Cytoscape software (v3.8.2) was utilized to calculate the node degree by MCODE and CytoHubba apps [29].", "[SUBTITLE] Dramatic transcriptional changes in rats’ epididymis after bilateral castration [SUBSECTION] To explore the transcriptional differences in the epididymis between Con-EP and Cas-EP rats, we obtained more than 4 million clean tags for each library. As demonstrated in Additional file 1, the distribution of total clean tags and distinct clean tags showed similar patterns in both DGE libraries, showing no bias between the library construction of Con-EP and Cas-EP. Around 3 million tags were mapped to genes for both Con-EP and Cas-EP, accounting for 62.89% and 70.32%, respectively.\nA total of 2,448 genes changed significantly between the two libraries. Among these genes, 1,632 were up-regulated while 816 were down-regulated after bilateral castration. More than 90% of the genes (2,274) were up- or down-regulated between 1.0 and 5.0 fold (data not shown). The Pearson correlation coefficient of the two libraries was only 0.658, reflecting the tremendous influence of castration on the gene expression profile of rat epididymis. We then performed qPCR of six selective genes which were up-regulated (Nfkbie, Plscr4 and Apoc1) or down-regulated (Lcn8, Prdx1 and Prdx3) to confirm the validity of the differentially expressed genes identified by DGE (n = 10). As expected, we obtained results that coincided with our sequencing data, confirming the authenticity of our sequencing results (Fig. 2).\n\nFig. 2Real-time fluorescent quantitative PCR analysis of differentially expressed genes before and after castration. a: down-regulated genes after bilateral castration, b: genes up-regulated after bilateral castration\n\nReal-time fluorescent quantitative PCR analysis of differentially expressed genes before and after castration. a: down-regulated genes after bilateral castration, b: genes up-regulated after bilateral castration\nTo explore the transcriptional differences in the epididymis between Con-EP and Cas-EP rats, we obtained more than 4 million clean tags for each library. As demonstrated in Additional file 1, the distribution of total clean tags and distinct clean tags showed similar patterns in both DGE libraries, showing no bias between the library construction of Con-EP and Cas-EP. Around 3 million tags were mapped to genes for both Con-EP and Cas-EP, accounting for 62.89% and 70.32%, respectively.\nA total of 2,448 genes changed significantly between the two libraries. Among these genes, 1,632 were up-regulated while 816 were down-regulated after bilateral castration. More than 90% of the genes (2,274) were up- or down-regulated between 1.0 and 5.0 fold (data not shown). The Pearson correlation coefficient of the two libraries was only 0.658, reflecting the tremendous influence of castration on the gene expression profile of rat epididymis. We then performed qPCR of six selective genes which were up-regulated (Nfkbie, Plscr4 and Apoc1) or down-regulated (Lcn8, Prdx1 and Prdx3) to confirm the validity of the differentially expressed genes identified by DGE (n = 10). As expected, we obtained results that coincided with our sequencing data, confirming the authenticity of our sequencing results (Fig. 2).\n\nFig. 2Real-time fluorescent quantitative PCR analysis of differentially expressed genes before and after castration. a: down-regulated genes after bilateral castration, b: genes up-regulated after bilateral castration\n\nReal-time fluorescent quantitative PCR analysis of differentially expressed genes before and after castration. a: down-regulated genes after bilateral castration, b: genes up-regulated after bilateral castration\n[SUBTITLE] CRPC-castration genes were enriched in cell proliferation and prostate gland development [SUBSECTION] To obtain potential CRPC driver genes dependent or independent of castration, we identified DEGs in the CRPC samples compared with the primary PCa samples. We obtained 1,904 up-regulated genes and 3,391 down-regulated genes (fold change > 1, P < 0.001) from GSE35988 (Fig. 3a), and 512 increased genes and 244 decreased genes (fold change > 1, P < 0.001) from GSE32269 (Fig. 3b). A total of 97 up-regulated genes (Fig. 3d) and 128 down-regulated genes (Fig. 3c) were shared by CRPC (GSE35988) and testis castration (data not shown), considered as CRPC DEGs related to castration (CRPC-castration). In contrast to testis castration, 120 up-regulated genes and 136 down-regulated genes changed only in CRPC (both GSE35988 and GSE32269) (Fig. 3e, data not shown), considered as CRPC-specific DEGs independent of castration.\n\nFig. 3Database analysis. a and b: Volcano plots of all genes in GSE35988 and GSE32269 Red dots represent genes with fold change ≥ 2 and P < 0.001, blue dots represent genes with fold change ≤ − 2 and P < 0.001, and the other dots represent the rest of genes with no statistically significant change in expression. FC, fold change. c, d and e: Venn diagrams showing the number of genes expressed as common and unique between the identified DEGs in DGE profiles of castrated rat and genes in the NCBI-gene database\n\nDatabase analysis. a and b: Volcano plots of all genes in GSE35988 and GSE32269 Red dots represent genes with fold change ≥ 2 and P < 0.001, blue dots represent genes with fold change ≤ − 2 and P < 0.001, and the other dots represent the rest of genes with no statistically significant change in expression. FC, fold change. c, d and e: Venn diagrams showing the number of genes expressed as common and unique between the identified DEGs in DGE profiles of castrated rat and genes in the NCBI-gene database\nNext, we explored the functional enrichment of these two lists of DEGs by using GO and KEGG analysis. The CRPC-specific DEGs significantly enriched cell division or mitotic processes (Fig. 4a), consistent with their oncogenic role in tumor cell proliferation. Concordantly, pathway analysis identified enrichment in the cell cycle pathway and focal adhesion pathway (Fig. 4b) (Additional file 2). On the other hand, GO enrichment of CRPC-castration DEGs (data not shown) showed that gland development, epithelial cell proliferation, and prostate gland development were substantially affected by castration (Fig. 5a). Pathway analysis showed that the pyruvate metabolism pathway was significantly enriched (Additional file 3).\n\nFig. 4Go and KEGG analysis. a: The top 10 results of GO functional analysis in CRPC-specific DEGs. b: KEGG functional analysis in CRPC-specific DEGs.\n\nGo and KEGG analysis. a: The top 10 results of GO functional analysis in CRPC-specific DEGs. b: KEGG functional analysis in CRPC-specific DEGs.\n\nFig. 5Go and PPI analysis. a: The top 10 results of GO functional analysis in CRPC-castration DEGs. b: Construction of PPI network of CRPC-castration DEGs.\n\nGo and PPI analysis. a: The top 10 results of GO functional analysis in CRPC-castration DEGs. b: Construction of PPI network of CRPC-castration DEGs.\nFurthermore, we used STRING to explore the protein-protein interaction (PPI) of the DEGs. CRPC-specific DEGs formed a PPI network with 208 nodes and 1353 edges. Among the 41 hub genes (node degree > 30) were chromatin remodeling factors EZH2 and PRC1, and cell cycle regulators CDCA3, CCNA2, CCNB2, CDKN3, CDK1, CCNB1, CDC20, MCM4, SMC4, CENPF, BUB1, BUB1B, NUSAP1, and NCAPG (Fig. 6). In contrast, CRPC-castration DEGs formed PPI networks with 171 nodes and sparse connections. The most connected sub-network with the hub gene ACTR10 was involved in innate immunity and cell polarity (Fig. 5b).\n\nFig. 6Top 41 of PPI network of CRPC-specific DEGs.\n\nTop 41 of PPI network of CRPC-specific DEGs.\nFinally, based on network and pathway analyses, we identified two key genes (NUSAP1 and NCAPG) among the top 41 nodes in the CRPC-specific PPI network, which participate in various pathways including chromosome segregation and nuclear division. Further analysis showed that, for both genes, higher expression was associated with shorter disease-free survival of patients (Fig. 7c and d). Interestingly, the expression of NUSAP1 and NCAPG differed between primary PCa and normal prostate tissues (Fig. 7a and b), which led us to hypothesize that they may have a role both in the initiation and in the progression of PCa.\n\nFig. 7Key genes analysis. a and b: Expression of the key genes NUSAP1 and NCAPG in TCGA with GEPIA. c and d: Disease-free survival in relation to the expression of the key genes (NUSAP1 and NCAPG) in TCGA.\n\nKey genes analysis. a and b: Expression of the key genes NUSAP1 and NCAPG in TCGA with GEPIA. c and d: Disease-free survival in relation to the expression of the key genes (NUSAP1 and NCAPG) in TCGA.\nTo obtain potential CRPC driver genes dependent or independent of castration, we identified DEGs in the CRPC samples compared with the primary PCa samples. We obtained 1,904 up-regulated genes and 3,391 down-regulated genes (fold change > 1, P < 0.001) from GSE35988 (Fig. 3a), and 512 increased genes and 244 decreased genes (fold change > 1, P < 0.001) from GSE32269 (Fig. 3b). A total of 97 up-regulated genes (Fig. 3d) and 128 down-regulated genes (Fig. 3c) were shared by CRPC (GSE35988) and testis castration (data not shown), considered as CRPC DEGs related to castration (CRPC-castration). In contrast to testis castration, 120 up-regulated genes and 136 down-regulated genes changed only in CRPC (both GSE35988 and GSE32269) (Fig. 3e, data not shown), considered as CRPC-specific DEGs independent of castration.\n\nFig. 3Database analysis. a and b: Volcano plots of all genes in GSE35988 and GSE32269 Red dots represent genes with fold change ≥ 2 and P < 0.001, blue dots represent genes with fold change ≤ − 2 and P < 0.001, and the other dots represent the rest of genes with no statistically significant change in expression. FC, fold change. c, d and e: Venn diagrams showing the number of genes expressed as common and unique between the identified DEGs in DGE profiles of castrated rat and genes in the NCBI-gene database\n\nDatabase analysis. a and b: Volcano plots of all genes in GSE35988 and GSE32269 Red dots represent genes with fold change ≥ 2 and P < 0.001, blue dots represent genes with fold change ≤ − 2 and P < 0.001, and the other dots represent the rest of genes with no statistically significant change in expression. FC, fold change. c, d and e: Venn diagrams showing the number of genes expressed as common and unique between the identified DEGs in DGE profiles of castrated rat and genes in the NCBI-gene database\nNext, we explored the functional enrichment of these two lists of DEGs by using GO and KEGG analysis. The CRPC-specific DEGs significantly enriched cell division or mitotic processes (Fig. 4a), consistent with their oncogenic role in tumor cell proliferation. Concordantly, pathway analysis identified enrichment in the cell cycle pathway and focal adhesion pathway (Fig. 4b) (Additional file 2). On the other hand, GO enrichment of CRPC-castration DEGs (data not shown) showed that gland development, epithelial cell proliferation, and prostate gland development were substantially affected by castration (Fig. 5a). Pathway analysis showed that the pyruvate metabolism pathway was significantly enriched (Additional file 3).\n\nFig. 4Go and KEGG analysis. a: The top 10 results of GO functional analysis in CRPC-specific DEGs. b: KEGG functional analysis in CRPC-specific DEGs.\n\nGo and KEGG analysis. a: The top 10 results of GO functional analysis in CRPC-specific DEGs. b: KEGG functional analysis in CRPC-specific DEGs.\n\nFig. 5Go and PPI analysis. a: The top 10 results of GO functional analysis in CRPC-castration DEGs. b: Construction of PPI network of CRPC-castration DEGs.\n\nGo and PPI analysis. a: The top 10 results of GO functional analysis in CRPC-castration DEGs. b: Construction of PPI network of CRPC-castration DEGs.\nFurthermore, we used STRING to explore the protein-protein interaction (PPI) of the DEGs. CRPC-specific DEGs formed a PPI network with 208 nodes and 1353 edges. Among the 41 hub genes (node degree > 30) were chromatin remodeling factors EZH2 and PRC1, and cell cycle regulators CDCA3, CCNA2, CCNB2, CDKN3, CDK1, CCNB1, CDC20, MCM4, SMC4, CENPF, BUB1, BUB1B, NUSAP1, and NCAPG (Fig. 6). In contrast, CRPC-castration DEGs formed PPI networks with 171 nodes and sparse connections. The most connected sub-network with the hub gene ACTR10 was involved in innate immunity and cell polarity (Fig. 5b).\n\nFig. 6Top 41 of PPI network of CRPC-specific DEGs.\n\nTop 41 of PPI network of CRPC-specific DEGs.\nFinally, based on network and pathway analyses, we identified two key genes (NUSAP1 and NCAPG) among the top 41 nodes in the CRPC-specific PPI network, which participate in various pathways including chromosome segregation and nuclear division. Further analysis showed that, for both genes, higher expression was associated with shorter disease-free survival of patients (Fig. 7c and d). Interestingly, the expression of NUSAP1 and NCAPG differed between primary PCa and normal prostate tissues (Fig. 7a and b), which led us to hypothesize that they may have a role both in the initiation and in the progression of PCa.\n\nFig. 7Key genes analysis. a and b: Expression of the key genes NUSAP1 and NCAPG in TCGA with GEPIA. c and d: Disease-free survival in relation to the expression of the key genes (NUSAP1 and NCAPG) in TCGA.\n\nKey genes analysis. a and b: Expression of the key genes NUSAP1 and NCAPG in TCGA with GEPIA. c and d: Disease-free survival in relation to the expression of the key genes (NUSAP1 and NCAPG) in TCGA.", "To explore the transcriptional differences in the epididymis between Con-EP and Cas-EP rats, we obtained more than 4 million clean tags for each library. As demonstrated in Additional file 1, the distribution of total clean tags and distinct clean tags showed similar patterns in both DGE libraries, showing no bias between the library construction of Con-EP and Cas-EP. Around 3 million tags were mapped to genes for both Con-EP and Cas-EP, accounting for 62.89% and 70.32%, respectively.\nA total of 2,448 genes changed significantly between the two libraries. Among these genes, 1,632 were up-regulated while 816 were down-regulated after bilateral castration. More than 90% of the genes (2,274) were up- or down-regulated between 1.0 and 5.0 fold (data not shown). The Pearson correlation coefficient of the two libraries was only 0.658, reflecting the tremendous influence of castration on the gene expression profile of rat epididymis. We then performed qPCR of six selective genes which were up-regulated (Nfkbie, Plscr4 and Apoc1) or down-regulated (Lcn8, Prdx1 and Prdx3) to confirm the validity of the differentially expressed genes identified by DGE (n = 10). As expected, we obtained results that coincided with our sequencing data, confirming the authenticity of our sequencing results (Fig. 2).\n\nFig. 2Real-time fluorescent quantitative PCR analysis of differentially expressed genes before and after castration. a: down-regulated genes after bilateral castration, b: genes up-regulated after bilateral castration\n\nReal-time fluorescent quantitative PCR analysis of differentially expressed genes before and after castration. a: down-regulated genes after bilateral castration, b: genes up-regulated after bilateral castration", "To obtain potential CRPC driver genes dependent or independent of castration, we identified DEGs in the CRPC samples compared with the primary PCa samples. We obtained 1,904 up-regulated genes and 3,391 down-regulated genes (fold change > 1, P < 0.001) from GSE35988 (Fig. 3a), and 512 increased genes and 244 decreased genes (fold change > 1, P < 0.001) from GSE32269 (Fig. 3b). A total of 97 up-regulated genes (Fig. 3d) and 128 down-regulated genes (Fig. 3c) were shared by CRPC (GSE35988) and testis castration (data not shown), considered as CRPC DEGs related to castration (CRPC-castration). In contrast to testis castration, 120 up-regulated genes and 136 down-regulated genes changed only in CRPC (both GSE35988 and GSE32269) (Fig. 3e, data not shown), considered as CRPC-specific DEGs independent of castration.\n\nFig. 3Database analysis. a and b: Volcano plots of all genes in GSE35988 and GSE32269 Red dots represent genes with fold change ≥ 2 and P < 0.001, blue dots represent genes with fold change ≤ − 2 and P < 0.001, and the other dots represent the rest of genes with no statistically significant change in expression. FC, fold change. c, d and e: Venn diagrams showing the number of genes expressed as common and unique between the identified DEGs in DGE profiles of castrated rat and genes in the NCBI-gene database\n\nDatabase analysis. a and b: Volcano plots of all genes in GSE35988 and GSE32269 Red dots represent genes with fold change ≥ 2 and P < 0.001, blue dots represent genes with fold change ≤ − 2 and P < 0.001, and the other dots represent the rest of genes with no statistically significant change in expression. FC, fold change. c, d and e: Venn diagrams showing the number of genes expressed as common and unique between the identified DEGs in DGE profiles of castrated rat and genes in the NCBI-gene database\nNext, we explored the functional enrichment of these two lists of DEGs by using GO and KEGG analysis. The CRPC-specific DEGs significantly enriched cell division or mitotic processes (Fig. 4a), consistent with their oncogenic role in tumor cell proliferation. Concordantly, pathway analysis identified enrichment in the cell cycle pathway and focal adhesion pathway (Fig. 4b) (Additional file 2). On the other hand, GO enrichment of CRPC-castration DEGs (data not shown) showed that gland development, epithelial cell proliferation, and prostate gland development were substantially affected by castration (Fig. 5a). Pathway analysis showed that the pyruvate metabolism pathway was significantly enriched (Additional file 3).\n\nFig. 4Go and KEGG analysis. a: The top 10 results of GO functional analysis in CRPC-specific DEGs. b: KEGG functional analysis in CRPC-specific DEGs.\n\nGo and KEGG analysis. a: The top 10 results of GO functional analysis in CRPC-specific DEGs. b: KEGG functional analysis in CRPC-specific DEGs.\n\nFig. 5Go and PPI analysis. a: The top 10 results of GO functional analysis in CRPC-castration DEGs. b: Construction of PPI network of CRPC-castration DEGs.\n\nGo and PPI analysis. a: The top 10 results of GO functional analysis in CRPC-castration DEGs. b: Construction of PPI network of CRPC-castration DEGs.\nFurthermore, we used STRING to explore the protein-protein interaction (PPI) of the DEGs. CRPC-specific DEGs formed a PPI network with 208 nodes and 1353 edges. Among the 41 hub genes (node degree > 30) were chromatin remodeling factors EZH2 and PRC1, and cell cycle regulators CDCA3, CCNA2, CCNB2, CDKN3, CDK1, CCNB1, CDC20, MCM4, SMC4, CENPF, BUB1, BUB1B, NUSAP1, and NCAPG (Fig. 6). In contrast, CRPC-castration DEGs formed PPI networks with 171 nodes and sparse connections. The most connected sub-network with the hub gene ACTR10 was involved in innate immunity and cell polarity (Fig. 5b).\n\nFig. 6Top 41 of PPI network of CRPC-specific DEGs.\n\nTop 41 of PPI network of CRPC-specific DEGs.\nFinally, based on network and pathway analyses, we identified two key genes (NUSAP1 and NCAPG) among the top 41 nodes in the CRPC-specific PPI network, which participate in various pathways including chromosome segregation and nuclear division. Further analysis showed that, for both genes, higher expression was associated with shorter disease-free survival of patients (Fig. 7c and d). Interestingly, the expression of NUSAP1 and NCAPG differed between primary PCa and normal prostate tissues (Fig. 7a and b), which led us to hypothesize that they may have a role both in the initiation and in the progression of PCa.\n\nFig. 7Key genes analysis. a and b: Expression of the key genes NUSAP1 and NCAPG in TCGA with GEPIA. c and d: Disease-free survival in relation to the expression of the key genes (NUSAP1 and NCAPG) in TCGA.\n\nKey genes analysis. a and b: Expression of the key genes NUSAP1 and NCAPG in TCGA with GEPIA. c and d: Disease-free survival in relation to the expression of the key genes (NUSAP1 and NCAPG) in TCGA.", "In this research, we focused on the role of bilateral castration-related genes in tumors and genes that are associated with tumors after blocking out these bilateral castration-related genes. We performed in silico analysis with DEG data in this study and publicly available CPRC-associated gene expression profiles datasets, and identified two significant genes, namely NUSAP1 and NCAPG in CRPC. NUSAP1 and NCAPG participated in various pathways including chromosome segregation, sister chromatid segregation, nuclear division. In consideration of the important role of these pathways in the initiation, progression and metastasis in PCa, it’s reasonable to assume that NUSAP1 and NCAPG play a critical role in the initiation and progression of PCa. In accordance with our hypothesis, a number of authoritative papers have reported that NUSAP1 plays a key role in cancers including PCa [30, 31]. Current understanding of NUSAP1’s function in specific mechanisms of cancer is limited, although some data indicates it could be a potential marker of cell proliferation [32]. On the molecular and cellular level, NUSAP is an essential microtubule-stabilizing and bundling protein that crosslinks microtubules at the central part of the spindle during mitosis [33]. Li’s group further reveals that NUSAP functions as a modulator of the dynamics of kinetochore microtubules and plays a pivotal role in chromosome oscillation [34]. In PCa cell lines, NUSAP1 is proved to be regulated by retinoblastoma-associated protein 1 (RB1), whose knockdown upregulated the expression of NUSAP1 via the RB1/E2F1 axis [35]. Furthermore, elevated expression level of NUSAP1 may increase proliferation and invasion of PCa cells, positively affecting prostate cancer progression. Despite limited effects on cell proliferation, NUSAP1 participates in development of metastatic disease, possibly by regulation of the expression of family with sequence similarity 101 member B (FAM101B). FAM101B is associated with the organization of perinuclear actin networks as well as the regulation of nuclear shape during epithelial-mesenchymal transition (EMT), a crucial event involved in the invasion and spread of cancer cells [36]. Besides, FAM101B functions as a signaling effector of TGF-β1, which has been widely demonstrated to promote invasion and metastatic spread during the progression of prostate tumor [37]. In other types of cancer, gastric cancer for example, low NUSAP1 expression is proven to inhibit mTORC1 pathway, hence suppressing proliferation, migration, and invasion of cancer cells [38].\nNCAPG has not been well studied in CRPC. NCAPG is a subunit of condensing | complex and is involved in proper segregation of sister chromatids during the cell cycle. There are several miRNA published involved in CRPC. Current research showed that NCAPG is targeted by miR-145-3p, a passenger strand downregulated in CRPC. High expression of NCAPG in CRPC compared with hormone-sensitive prostate cancer suggested that NCAPG is closely associated with the pathogenesis of CRPC [39]. Another CRPC-related passenger strand miR-99a-3p was also found to downregulate NCAPG significantly, indicating potent antitumor effects [40]. A recent study showed that overexpression of NCAPG promoted cell proliferation and decreased cell apoptosis in hepatocellular carcinoma via activating PI3K-AKT signaling pathway [41]. Further studies about the concrete mechanism of NCAPG regulation in CRPC are in progress.", "Our study provided an insight into the regulation of epididymal gene expression after bilateral castration. These results contribute to our understanding of testis-dependent epididymal functions. Still, we preformed insights into gene regulation of CRPC dependent or independent of castration and improved our understanding of CRPC development and progression.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, null, null, null, null, null, null, null, null, null, "results", null, null, "discussion", "conclusion", "supplementary-material", null ]
[ "Differentially expressed genes (DEGs)", "Castration-resistant prostate cancer (CRPC)", "Cell cycle", "Cell proliferation", "Prostate gland development" ]
Superoxide-producing thermostable associate from the small intestines of control and alloxan-induced diabetic rats: quantitative and qualitative changes.
36258207
NADPH oxidase 1 (Nox1), which is highly expressed in the colon, is thought to play a potential role in host defense as a physical and innate immune barrier against commensal or pathogenic microbes in the gastrointestinal epithelium. Diabetes can be caused by several biological factors, including insulin resistance is one of them. Alloxan is widely used to induce insulin-dependent diabetes in experimental animals. Alloxan increases the generation of reactive oxygen species as a result of metabolic reactions in the body, along with a massive increase in cytosolic calcium concentration.
BACKGROUND
Using a universal method, a superoxide radical (О2-)-thermostable associate between NADPH-containing lipoprotein (NLP) and NADPH oxidase (Nox)- NLP-Nox was isolated and purified from the small intestine (SI) of control (C) and alloxan-induced diabetic (AD) albino rats.
METHODS
In comparison to the C indices, in AD in the SI, an increase in the specific content of NLP-Nox associate and a decrease in the stationary concentration of produced О2- in liquid phase (in solution) and gas phase (during blowing by oxygen of the NLP-Nox solution) were observed. The NLP-Nox of SI associate in C and AD rats produced О2- by an immediate mechanism, using NLP as a substrate. The phenomenon of the hiding of the optical absorption maxima of the Nox in oxidized states at pH10,5 was observed in the composition of these SI associates of the C and AD rat groups. The characteristic absorption maxima of the «hidden» Nox were observed under these conditions after reduction by potassium dithionite.
RESULTS
Thus, at AD, the decrease in the stationary concentration of produced О2- in the solution and gas phase was compensated for by an increase in the specific amount of associate. In addition,  the decrease in the stationary concentration of produced О2- by NLP-Nox associates at AD can be linked to a decrease in the level of NADPH in NLP-Nox composition. This could be used as a new mechanism of AD pathogenesis.
CONCLUSION
[ "Animals", "Superoxides", "Alloxan", "NADPH Oxidase 1", "Reactive Oxygen Species", "NADP", "Diabetes Mellitus, Experimental", "Calcium", "Dithionite", "NADPH Oxidases", "Intestine, Small", "Lipoproteins", "Oxygen", "Potassium", "Insulins" ]
9580182
null
null
null
null
Results
These О2−-producing associates NLP-Nox from SI of C and AD rats did not undergo PAAG electrophoresis and remained aggregated on the gel tube entry. Indirectly, the purity of these associates is evidenced by the absence of strips of accompanying water-soluble proteins of acidic and basic nature during electrophoresis of these associates’ opalescent solutions on 10% PAAG tubes. The symmetry of the elution diagrams of the Nox-NLP associates after gel filtration through Sephadex G-100 and the unchanged optical spectral index (A280/A400) show the purity of these associates. The specific content of NLP-Nox from SI of C and AD rats was 10,1 ± 0,12 mg/g and 16,6 ± 0,10 mg/g (p < 0,001, n = 6), correspondingly. After heating in boiling water for 10–12 min, the C and AD small intestine associates practically did not reduce nativity and О2−-producing activity. The characteristic absorption maxima for Nox were observed in the optical absorption spectra of NLP-Nox from SI, C, and AD rats in the oxidized and reduced states at pH 9,5 (Fig. 1). Fig. 1Optical absorption spectra of opalescent NLP-Nox associate solutions from SI of C (1) and AD rats (2) at pH 9.5. The spectra (3) were obtained after reduction of (2) by sodium dithionite Optical absorption spectra of opalescent NLP-Nox associate solutions from SI of C (1) and AD rats (2) at pH 9.5. The spectra (3) were obtained after reduction of (2) by sodium dithionite In comparison to Nox, in the associate of NLP-Nox the “free” Nox from SI in the associate of NLP-Nox had low oxidative/reductive properties, as shown in Fig. 2. Fig. 2The optical absorption spectra of NOx from SI of C (a) and AD (b) rats in oxidized (1) and after reduction by sodium dithionite (2) The optical absorption spectra of NOx from SI of C (a) and AD (b) rats in oxidized (1) and after reduction by sodium dithionite (2) These results showed that the Nox in NLP-Nox associate was more easily reduced by sodium dithionite than free Nox from the SI of the C and AD rats. The slow opalescence of NLP-Nox associates from the SI of C and AD rats at pH 10,5 in oxidized and reduced states by sodium dithionite, which differed significantly, as shown in Fig. 3. Fig. 3a – Optical absorption spectra of NLP-Nox associate from the SI of C rats in an oxidized state at pH 10,5 (1) and after reduction by sodium dithionite (2). b - Optical absorption spectra of the NLP-Nox associate from the SI of AD rats in an oxidized state (1) at pH10,5 and after reduction by sodium dithionite (2). a – Optical absorption spectra of NLP-Nox associate from the SI of C rats in an oxidized state at pH 10,5 (1) and after reduction by sodium dithionite (2). b - Optical absorption spectra of the NLP-Nox associate from the SI of AD rats in an oxidized state (1) at pH10,5 and after reduction by sodium dithionite (2). As shown in Fig. 3, the characteristic optical absorption spectra of Nox in the composition of the NLP-Nox associate did not exist in oxidized states at pH 10.5. However, the characteristic optical spectra of Nox in the composition of NLP-Nox were observed at pH 10.5 after the reduction of NLP-Nox associate with potassium dithionite. This phenomenon was observed for the first time in the present study. Reversible changes in the optical absorption spectra of NLP-Nox associates from the SI of the C and AD rats were observed. The effect of NADPH on Nox activation in the gastrointestinal tract has already been revealed [11]. This NADPH, on the other hand, is a component of the NLP in the composition of the О2−-producing NLP-Nox associate from SI. In comparison to similar indicators in C rats, the fluorescence intensity «F» (in relative units) of NADPH in the composition of the associate of NLP-Nox from the SI of AD rats was observed up to 27,5 ± 1,6% (p < 0,005, n = 6) (Fig. 4). Fig. 4Spectrofluorescence spectra of NLP-Nox solution from the SI of C (1) and AD (2) rats (p < 0,005, n = 6) Spectrofluorescence spectra of NLP-Nox solution from the SI of C (1) and AD (2) rats (p < 0,005, n = 6) The content of the lipid component (malondialdehyde) in NLP-Nox associate from the SI of AD rats is higher to 22,4%, than that in C rats. The stationary concentrations of produced О2− by NLP-Nox associates (1,4 mg/ml) in homogeneous phase (in solution) from C and AD rats were 5,6 ± 0,2 mM and 3,8 ± 0,2 mM (p < 0,05, n = 6).
Conclusion
Our results suggest that the NLP-Nox associate from the rats’ small intestine is a new О2−-producing thermostable associate in the tissue. As a new AD diagnostic test, it immediately produces О2− with the corresponding quantitative and qualitative changes in C and AD rats.
[ "Background", "Method", "Isolation and purification of О2−-producing associate NLP- Nox from rat`s SI", "Determination of NADPH in the composition of SI NLP-Nox", "Determination of the lipid component in the SI NLP-Nox composition", "Determination of the Nox in the NLP-Nox composition of SI", "Determination of the stationary concentration of О2−produced by SI NLP-Nox associate" ]
[ "Insulin resistance and deficiency are associated with hyperglycemia and hyperlipidemia, leading to the development of many chronic diseases, such as neuropathy, nephropathy, and various vascular diseases related to the heart, kidneys, brain, peripheral vessels, and retinopathy. Diabetes can be caused by a number of biological factors including obesity, stress, excessive alcohol consumption, and improper exercise [1, 2].\nAlloxan is an organic compound, urea derivative, and cytotoxic glucose analog. Alloxan is widely used to induce insulin-dependent diabetes (type 1) in experimental animals. It acts by increasing the generation of reactive oxygen species as a result of metabolic reactions in the body, along with a massive increase in cytosolic calcium concentration, and can rapidly cause pancreatic beta cell destruction [3–6].\nNADPH oxidase 1 (Nox1), which is highly expressed in the colon, is thought to play a potential role in protecting the body as a physical and innate immune barrier against commensal or pathogenic microbes in the gastrointestinal epithelium. Vitamin E has been shown to improve SI changes in diabetic rats. These effects may be partially mediated by an increase in plasma antioxidant capacity and a decrease in lipid peroxidation [8–10]. It is well known that intravital NADPH fluorescence lifetime imaging in the SI of fluorescent reporter mice can be used to monitor NADPH-dependent metabolism of epithelial and myeloid cells. Diabetes causes numerous morphological and functional changes in the small intestine. After 6 weeks of diabetes, there was a fourfold increase in lipid peroxidation, as measured by thiobarbituric acid reactive substances, and a 38% increase in protein oxidation, as measured by protein carbonyl content. There was a significant increase in the activities of catalase (123.9%) and superoxide dismutase (71.9%) and a decrease in the activity of glutathione peroxidase (67.7%) [11–13]. We have shown previously that natural non-discontinuous permanent and stable О2−-producing system in the spinal cord injured rats with experimental type II diabetes are significantly destabilized. This destabilization was considerably intensified in diabetic rats subjected to additional spinal cord injury. Correspondingly, improvement of the antioxidant status may be an important component in the strategy of treatment of both diabetes and consequences of spinal cord injury [14]. Thus, these results suggest that oxidative stress occurs in the SI during diabetes and may be involved in some of the associated functional changes. The gastrointestinal epithelium serves as a physical and innate immune barrier against commensal and pathogenic microbes. NADPH oxidase 1 (Nox1) and dual oxidase 2 (Duox2), which are highly expressed in the colon, may play a role in the host defense. ROS derived from Nox1 have been linked to the pathogenesis of inflammation-related tumor development. The human stomach does not express Nox1 [10, 11,15]. The thermostable associate from the SI of C and alloxan diabetic rats can be obtained using a universal method [7] for preparing isoforms of thermostable associates from biomembranes of animal origin.\nThe goal of this study was to isolate and purify NLP-Nox isoforms from the SI of C and AD rats by determining the specific content of associates and the stationary concentration of produced О2− in the solution and gas phase.", "Diabetes was induced in rats by intraperitoneal administration of alloxan. Rats were divided into two groups (eight rats each): the control group (C) and the alloxan-induced diabetic group. Prior to the experiments, rats (220–240 g) were fed standard rodent food for one week for acclimation to the laboratory conditions. The acclimated rats were fasted for 12 h with free access to water, and then injected with 0.5% alloxan dissolved in 10 mM sodium citrate (pH 4.5) at a dose of 150 mg/kg. Under the same conditions, C rats were injected with a saline solution. After 5 days, blood glucose was measured using a «Saterlit» glucometer. After 5 days, the animals were fasted, anesthetized with halothane, and dissected [16].\nThe experimental protocol corresponded to the conditions of the European Communities Council Directive (2010/63/ UE) and was approved by the Ethics Committee of Yerevan State Medical University after Mkhitar Heratsi (IRB Approval N4, November 15, 2018).\n[SUBTITLE] Isolation and purification of О2−-producing associate NLP- Nox from rat`s SI [SUBSECTION] NLP – Nox associates from rat SI were isolated and purified using a universal method [7], which uses human ferrihemoglobin (Hb) for the release of Nox and NLP-Nox from SI [17]. In particular, a water homogenate of SI (up to 6 g) was incubated for 1.5 h at pH 9,5 and 37 °C in the presence of 50 mM human Hb. The pH of the supernatant was adjusted to 4 after centrifugation at 5000 g for 10 min. The precipitate of the NLP-Nox fraction was soluble in water at pH9.5. After centrifugation, the supernatant was subjected to ion-exchange chromatography using cellulose DE-52 at pH 9.5. The Nox-NLP associate was eluted free because it was not absorbed by the column. The Nox fraction was eluted with 0.2 M potassium phosphate buffer (PPB) at pH 7.4. Following the concentration of the Nox and NLP-Nox fractions, gel filtration was performed on a separate Sephadex G-100 column at pH 9.5. The fractions of NLP-Nox and Nox eluted with a symmetrical elution diagram were collected and vacuum lyophilized after deionization of the Nox and NLP-Nox associates. They were weighed and stored in closed containers under a nitrogen atmosphere at -10oC.\nElectrophoresis of the Nox-NLP associates was performed on a 10% PAAG (polyacrylamide gel) for proteins of acidic or basic characteristics.\nNLP – Nox associates from rat SI were isolated and purified using a universal method [7], which uses human ferrihemoglobin (Hb) for the release of Nox and NLP-Nox from SI [17]. In particular, a water homogenate of SI (up to 6 g) was incubated for 1.5 h at pH 9,5 and 37 °C in the presence of 50 mM human Hb. The pH of the supernatant was adjusted to 4 after centrifugation at 5000 g for 10 min. The precipitate of the NLP-Nox fraction was soluble in water at pH9.5. After centrifugation, the supernatant was subjected to ion-exchange chromatography using cellulose DE-52 at pH 9.5. The Nox-NLP associate was eluted free because it was not absorbed by the column. The Nox fraction was eluted with 0.2 M potassium phosphate buffer (PPB) at pH 7.4. Following the concentration of the Nox and NLP-Nox fractions, gel filtration was performed on a separate Sephadex G-100 column at pH 9.5. The fractions of NLP-Nox and Nox eluted with a symmetrical elution diagram were collected and vacuum lyophilized after deionization of the Nox and NLP-Nox associates. They were weighed and stored in closed containers under a nitrogen atmosphere at -10oC.\nElectrophoresis of the Nox-NLP associates was performed on a 10% PAAG (polyacrylamide gel) for proteins of acidic or basic characteristics.\n[SUBTITLE] Determination of NADPH in the composition of SI NLP-Nox [SUBSECTION] The presence of NADPH in the composition of SI NLP-Nox was determined using a spectrofluorimetric method that measured the fluorescence intensities in comparative units (F) at 430 nm with excitation at 370 nm [18].\nThe presence of NADPH in the composition of SI NLP-Nox was determined using a spectrofluorimetric method that measured the fluorescence intensities in comparative units (F) at 430 nm with excitation at 370 nm [18].\n[SUBTITLE] Determination of the lipid component in the SI NLP-Nox composition [SUBSECTION] The lipid component of SI NLP-Nox was determined by measuring the lipid peroxidation product, malondialdehyde (MDA) [19].\nThe lipid component of SI NLP-Nox was determined by measuring the lipid peroxidation product, malondialdehyde (MDA) [19].\n[SUBTITLE] Determination of the Nox in the NLP-Nox composition of SI [SUBSECTION] The Nox in the composition of NLP-Nox associate from SI was determined using the optical absorbance characteristic for Nox at 558, 525, and 418 nm in the reduced states by sodium dithionite.\nThe Nox in the composition of NLP-Nox associate from SI was determined using the optical absorbance characteristic for Nox at 558, 525, and 418 nm in the reduced states by sodium dithionite.\n[SUBTITLE] Determination of the stationary concentration of О2−produced by SI NLP-Nox associate [SUBSECTION] The stationary concentration of О2− produced by NLP-Nox associate from SI was determined using the adrenaline method. The maximal optical absorbance of adrenochrome (at 500 nm), which is formed during the oxidation of adrenaline by produced О2− [20], was determined. At the same, the stationary concentration (M) of produced О2− is equal to the concentration of formed adrenochrome, with a molar extinction (E) of up to 750 M-1 cm-1. The stationary concentration (M) of О2− produced by this associate NLP-Nox was determined in the homogeneous phase (in solution) and gas phase by determining the value of A500/E. The optical absorbance of adrenochrome, which is formed during the oxidation of adrenaline by air oxygen, was used as a control.\nThe content of the lipid component (malondialdehyde) in NLP-Nox associate from SI in AD rats was 22.4% higher than that in C rats.\nThe specific content of NLP-Nox was determined by weighing it after deionization and vacuum lyophilization and was expressed in mg per 1 g SI (mg/g).\nThe cellulose of DE-52 («Whatman», England), Sephadex G-100 («Pharmacia», Sweden), adrenaline («Sigma», USA), spectrophotometer «Cary 60» (USA), spectrofluorometer «Perkin-Elmer», (USA), centrifuge K-70D and K-24 «Janetzki» (Germany) were used during the investigation.\nStatistical analysis of the results using the variational statistics method of Student-Fisher was performed to determine the validation criterion (M ± m).\nThe stationary concentration of О2− produced by NLP-Nox associate from SI was determined using the adrenaline method. The maximal optical absorbance of adrenochrome (at 500 nm), which is formed during the oxidation of adrenaline by produced О2− [20], was determined. At the same, the stationary concentration (M) of produced О2− is equal to the concentration of formed adrenochrome, with a molar extinction (E) of up to 750 M-1 cm-1. The stationary concentration (M) of О2− produced by this associate NLP-Nox was determined in the homogeneous phase (in solution) and gas phase by determining the value of A500/E. The optical absorbance of adrenochrome, which is formed during the oxidation of adrenaline by air oxygen, was used as a control.\nThe content of the lipid component (malondialdehyde) in NLP-Nox associate from SI in AD rats was 22.4% higher than that in C rats.\nThe specific content of NLP-Nox was determined by weighing it after deionization and vacuum lyophilization and was expressed in mg per 1 g SI (mg/g).\nThe cellulose of DE-52 («Whatman», England), Sephadex G-100 («Pharmacia», Sweden), adrenaline («Sigma», USA), spectrophotometer «Cary 60» (USA), spectrofluorometer «Perkin-Elmer», (USA), centrifuge K-70D and K-24 «Janetzki» (Germany) were used during the investigation.\nStatistical analysis of the results using the variational statistics method of Student-Fisher was performed to determine the validation criterion (M ± m).", "NLP – Nox associates from rat SI were isolated and purified using a universal method [7], which uses human ferrihemoglobin (Hb) for the release of Nox and NLP-Nox from SI [17]. In particular, a water homogenate of SI (up to 6 g) was incubated for 1.5 h at pH 9,5 and 37 °C in the presence of 50 mM human Hb. The pH of the supernatant was adjusted to 4 after centrifugation at 5000 g for 10 min. The precipitate of the NLP-Nox fraction was soluble in water at pH9.5. After centrifugation, the supernatant was subjected to ion-exchange chromatography using cellulose DE-52 at pH 9.5. The Nox-NLP associate was eluted free because it was not absorbed by the column. The Nox fraction was eluted with 0.2 M potassium phosphate buffer (PPB) at pH 7.4. Following the concentration of the Nox and NLP-Nox fractions, gel filtration was performed on a separate Sephadex G-100 column at pH 9.5. The fractions of NLP-Nox and Nox eluted with a symmetrical elution diagram were collected and vacuum lyophilized after deionization of the Nox and NLP-Nox associates. They were weighed and stored in closed containers under a nitrogen atmosphere at -10oC.\nElectrophoresis of the Nox-NLP associates was performed on a 10% PAAG (polyacrylamide gel) for proteins of acidic or basic characteristics.", "The presence of NADPH in the composition of SI NLP-Nox was determined using a spectrofluorimetric method that measured the fluorescence intensities in comparative units (F) at 430 nm with excitation at 370 nm [18].", "The lipid component of SI NLP-Nox was determined by measuring the lipid peroxidation product, malondialdehyde (MDA) [19].", "The Nox in the composition of NLP-Nox associate from SI was determined using the optical absorbance characteristic for Nox at 558, 525, and 418 nm in the reduced states by sodium dithionite.", "The stationary concentration of О2− produced by NLP-Nox associate from SI was determined using the adrenaline method. The maximal optical absorbance of adrenochrome (at 500 nm), which is formed during the oxidation of adrenaline by produced О2− [20], was determined. At the same, the stationary concentration (M) of produced О2− is equal to the concentration of formed adrenochrome, with a molar extinction (E) of up to 750 M-1 cm-1. The stationary concentration (M) of О2− produced by this associate NLP-Nox was determined in the homogeneous phase (in solution) and gas phase by determining the value of A500/E. The optical absorbance of adrenochrome, which is formed during the oxidation of adrenaline by air oxygen, was used as a control.\nThe content of the lipid component (malondialdehyde) in NLP-Nox associate from SI in AD rats was 22.4% higher than that in C rats.\nThe specific content of NLP-Nox was determined by weighing it after deionization and vacuum lyophilization and was expressed in mg per 1 g SI (mg/g).\nThe cellulose of DE-52 («Whatman», England), Sephadex G-100 («Pharmacia», Sweden), adrenaline («Sigma», USA), spectrophotometer «Cary 60» (USA), spectrofluorometer «Perkin-Elmer», (USA), centrifuge K-70D and K-24 «Janetzki» (Germany) were used during the investigation.\nStatistical analysis of the results using the variational statistics method of Student-Fisher was performed to determine the validation criterion (M ± m)." ]
[ null, null, null, null, null, null, null ]
[ "Background", "Method", "Isolation and purification of О2−-producing associate NLP- Nox from rat`s SI", "Determination of NADPH in the composition of SI NLP-Nox", "Determination of the lipid component in the SI NLP-Nox composition", "Determination of the Nox in the NLP-Nox composition of SI", "Determination of the stationary concentration of О2−produced by SI NLP-Nox associate", "Results", "Discussion", "Conclusion" ]
[ "Insulin resistance and deficiency are associated with hyperglycemia and hyperlipidemia, leading to the development of many chronic diseases, such as neuropathy, nephropathy, and various vascular diseases related to the heart, kidneys, brain, peripheral vessels, and retinopathy. Diabetes can be caused by a number of biological factors including obesity, stress, excessive alcohol consumption, and improper exercise [1, 2].\nAlloxan is an organic compound, urea derivative, and cytotoxic glucose analog. Alloxan is widely used to induce insulin-dependent diabetes (type 1) in experimental animals. It acts by increasing the generation of reactive oxygen species as a result of metabolic reactions in the body, along with a massive increase in cytosolic calcium concentration, and can rapidly cause pancreatic beta cell destruction [3–6].\nNADPH oxidase 1 (Nox1), which is highly expressed in the colon, is thought to play a potential role in protecting the body as a physical and innate immune barrier against commensal or pathogenic microbes in the gastrointestinal epithelium. Vitamin E has been shown to improve SI changes in diabetic rats. These effects may be partially mediated by an increase in plasma antioxidant capacity and a decrease in lipid peroxidation [8–10]. It is well known that intravital NADPH fluorescence lifetime imaging in the SI of fluorescent reporter mice can be used to monitor NADPH-dependent metabolism of epithelial and myeloid cells. Diabetes causes numerous morphological and functional changes in the small intestine. After 6 weeks of diabetes, there was a fourfold increase in lipid peroxidation, as measured by thiobarbituric acid reactive substances, and a 38% increase in protein oxidation, as measured by protein carbonyl content. There was a significant increase in the activities of catalase (123.9%) and superoxide dismutase (71.9%) and a decrease in the activity of glutathione peroxidase (67.7%) [11–13]. We have shown previously that natural non-discontinuous permanent and stable О2−-producing system in the spinal cord injured rats with experimental type II diabetes are significantly destabilized. This destabilization was considerably intensified in diabetic rats subjected to additional spinal cord injury. Correspondingly, improvement of the antioxidant status may be an important component in the strategy of treatment of both diabetes and consequences of spinal cord injury [14]. Thus, these results suggest that oxidative stress occurs in the SI during diabetes and may be involved in some of the associated functional changes. The gastrointestinal epithelium serves as a physical and innate immune barrier against commensal and pathogenic microbes. NADPH oxidase 1 (Nox1) and dual oxidase 2 (Duox2), which are highly expressed in the colon, may play a role in the host defense. ROS derived from Nox1 have been linked to the pathogenesis of inflammation-related tumor development. The human stomach does not express Nox1 [10, 11,15]. The thermostable associate from the SI of C and alloxan diabetic rats can be obtained using a universal method [7] for preparing isoforms of thermostable associates from biomembranes of animal origin.\nThe goal of this study was to isolate and purify NLP-Nox isoforms from the SI of C and AD rats by determining the specific content of associates and the stationary concentration of produced О2− in the solution and gas phase.", "Diabetes was induced in rats by intraperitoneal administration of alloxan. Rats were divided into two groups (eight rats each): the control group (C) and the alloxan-induced diabetic group. Prior to the experiments, rats (220–240 g) were fed standard rodent food for one week for acclimation to the laboratory conditions. The acclimated rats were fasted for 12 h with free access to water, and then injected with 0.5% alloxan dissolved in 10 mM sodium citrate (pH 4.5) at a dose of 150 mg/kg. Under the same conditions, C rats were injected with a saline solution. After 5 days, blood glucose was measured using a «Saterlit» glucometer. After 5 days, the animals were fasted, anesthetized with halothane, and dissected [16].\nThe experimental protocol corresponded to the conditions of the European Communities Council Directive (2010/63/ UE) and was approved by the Ethics Committee of Yerevan State Medical University after Mkhitar Heratsi (IRB Approval N4, November 15, 2018).\n[SUBTITLE] Isolation and purification of О2−-producing associate NLP- Nox from rat`s SI [SUBSECTION] NLP – Nox associates from rat SI were isolated and purified using a universal method [7], which uses human ferrihemoglobin (Hb) for the release of Nox and NLP-Nox from SI [17]. In particular, a water homogenate of SI (up to 6 g) was incubated for 1.5 h at pH 9,5 and 37 °C in the presence of 50 mM human Hb. The pH of the supernatant was adjusted to 4 after centrifugation at 5000 g for 10 min. The precipitate of the NLP-Nox fraction was soluble in water at pH9.5. After centrifugation, the supernatant was subjected to ion-exchange chromatography using cellulose DE-52 at pH 9.5. The Nox-NLP associate was eluted free because it was not absorbed by the column. The Nox fraction was eluted with 0.2 M potassium phosphate buffer (PPB) at pH 7.4. Following the concentration of the Nox and NLP-Nox fractions, gel filtration was performed on a separate Sephadex G-100 column at pH 9.5. The fractions of NLP-Nox and Nox eluted with a symmetrical elution diagram were collected and vacuum lyophilized after deionization of the Nox and NLP-Nox associates. They were weighed and stored in closed containers under a nitrogen atmosphere at -10oC.\nElectrophoresis of the Nox-NLP associates was performed on a 10% PAAG (polyacrylamide gel) for proteins of acidic or basic characteristics.\nNLP – Nox associates from rat SI were isolated and purified using a universal method [7], which uses human ferrihemoglobin (Hb) for the release of Nox and NLP-Nox from SI [17]. In particular, a water homogenate of SI (up to 6 g) was incubated for 1.5 h at pH 9,5 and 37 °C in the presence of 50 mM human Hb. The pH of the supernatant was adjusted to 4 after centrifugation at 5000 g for 10 min. The precipitate of the NLP-Nox fraction was soluble in water at pH9.5. After centrifugation, the supernatant was subjected to ion-exchange chromatography using cellulose DE-52 at pH 9.5. The Nox-NLP associate was eluted free because it was not absorbed by the column. The Nox fraction was eluted with 0.2 M potassium phosphate buffer (PPB) at pH 7.4. Following the concentration of the Nox and NLP-Nox fractions, gel filtration was performed on a separate Sephadex G-100 column at pH 9.5. The fractions of NLP-Nox and Nox eluted with a symmetrical elution diagram were collected and vacuum lyophilized after deionization of the Nox and NLP-Nox associates. They were weighed and stored in closed containers under a nitrogen atmosphere at -10oC.\nElectrophoresis of the Nox-NLP associates was performed on a 10% PAAG (polyacrylamide gel) for proteins of acidic or basic characteristics.\n[SUBTITLE] Determination of NADPH in the composition of SI NLP-Nox [SUBSECTION] The presence of NADPH in the composition of SI NLP-Nox was determined using a spectrofluorimetric method that measured the fluorescence intensities in comparative units (F) at 430 nm with excitation at 370 nm [18].\nThe presence of NADPH in the composition of SI NLP-Nox was determined using a spectrofluorimetric method that measured the fluorescence intensities in comparative units (F) at 430 nm with excitation at 370 nm [18].\n[SUBTITLE] Determination of the lipid component in the SI NLP-Nox composition [SUBSECTION] The lipid component of SI NLP-Nox was determined by measuring the lipid peroxidation product, malondialdehyde (MDA) [19].\nThe lipid component of SI NLP-Nox was determined by measuring the lipid peroxidation product, malondialdehyde (MDA) [19].\n[SUBTITLE] Determination of the Nox in the NLP-Nox composition of SI [SUBSECTION] The Nox in the composition of NLP-Nox associate from SI was determined using the optical absorbance characteristic for Nox at 558, 525, and 418 nm in the reduced states by sodium dithionite.\nThe Nox in the composition of NLP-Nox associate from SI was determined using the optical absorbance characteristic for Nox at 558, 525, and 418 nm in the reduced states by sodium dithionite.\n[SUBTITLE] Determination of the stationary concentration of О2−produced by SI NLP-Nox associate [SUBSECTION] The stationary concentration of О2− produced by NLP-Nox associate from SI was determined using the adrenaline method. The maximal optical absorbance of adrenochrome (at 500 nm), which is formed during the oxidation of adrenaline by produced О2− [20], was determined. At the same, the stationary concentration (M) of produced О2− is equal to the concentration of formed adrenochrome, with a molar extinction (E) of up to 750 M-1 cm-1. The stationary concentration (M) of О2− produced by this associate NLP-Nox was determined in the homogeneous phase (in solution) and gas phase by determining the value of A500/E. The optical absorbance of adrenochrome, which is formed during the oxidation of adrenaline by air oxygen, was used as a control.\nThe content of the lipid component (malondialdehyde) in NLP-Nox associate from SI in AD rats was 22.4% higher than that in C rats.\nThe specific content of NLP-Nox was determined by weighing it after deionization and vacuum lyophilization and was expressed in mg per 1 g SI (mg/g).\nThe cellulose of DE-52 («Whatman», England), Sephadex G-100 («Pharmacia», Sweden), adrenaline («Sigma», USA), spectrophotometer «Cary 60» (USA), spectrofluorometer «Perkin-Elmer», (USA), centrifuge K-70D and K-24 «Janetzki» (Germany) were used during the investigation.\nStatistical analysis of the results using the variational statistics method of Student-Fisher was performed to determine the validation criterion (M ± m).\nThe stationary concentration of О2− produced by NLP-Nox associate from SI was determined using the adrenaline method. The maximal optical absorbance of adrenochrome (at 500 nm), which is formed during the oxidation of adrenaline by produced О2− [20], was determined. At the same, the stationary concentration (M) of produced О2− is equal to the concentration of formed adrenochrome, with a molar extinction (E) of up to 750 M-1 cm-1. The stationary concentration (M) of О2− produced by this associate NLP-Nox was determined in the homogeneous phase (in solution) and gas phase by determining the value of A500/E. The optical absorbance of adrenochrome, which is formed during the oxidation of adrenaline by air oxygen, was used as a control.\nThe content of the lipid component (malondialdehyde) in NLP-Nox associate from SI in AD rats was 22.4% higher than that in C rats.\nThe specific content of NLP-Nox was determined by weighing it after deionization and vacuum lyophilization and was expressed in mg per 1 g SI (mg/g).\nThe cellulose of DE-52 («Whatman», England), Sephadex G-100 («Pharmacia», Sweden), adrenaline («Sigma», USA), spectrophotometer «Cary 60» (USA), spectrofluorometer «Perkin-Elmer», (USA), centrifuge K-70D and K-24 «Janetzki» (Germany) were used during the investigation.\nStatistical analysis of the results using the variational statistics method of Student-Fisher was performed to determine the validation criterion (M ± m).", "NLP – Nox associates from rat SI were isolated and purified using a universal method [7], which uses human ferrihemoglobin (Hb) for the release of Nox and NLP-Nox from SI [17]. In particular, a water homogenate of SI (up to 6 g) was incubated for 1.5 h at pH 9,5 and 37 °C in the presence of 50 mM human Hb. The pH of the supernatant was adjusted to 4 after centrifugation at 5000 g for 10 min. The precipitate of the NLP-Nox fraction was soluble in water at pH9.5. After centrifugation, the supernatant was subjected to ion-exchange chromatography using cellulose DE-52 at pH 9.5. The Nox-NLP associate was eluted free because it was not absorbed by the column. The Nox fraction was eluted with 0.2 M potassium phosphate buffer (PPB) at pH 7.4. Following the concentration of the Nox and NLP-Nox fractions, gel filtration was performed on a separate Sephadex G-100 column at pH 9.5. The fractions of NLP-Nox and Nox eluted with a symmetrical elution diagram were collected and vacuum lyophilized after deionization of the Nox and NLP-Nox associates. They were weighed and stored in closed containers under a nitrogen atmosphere at -10oC.\nElectrophoresis of the Nox-NLP associates was performed on a 10% PAAG (polyacrylamide gel) for proteins of acidic or basic characteristics.", "The presence of NADPH in the composition of SI NLP-Nox was determined using a spectrofluorimetric method that measured the fluorescence intensities in comparative units (F) at 430 nm with excitation at 370 nm [18].", "The lipid component of SI NLP-Nox was determined by measuring the lipid peroxidation product, malondialdehyde (MDA) [19].", "The Nox in the composition of NLP-Nox associate from SI was determined using the optical absorbance characteristic for Nox at 558, 525, and 418 nm in the reduced states by sodium dithionite.", "The stationary concentration of О2− produced by NLP-Nox associate from SI was determined using the adrenaline method. The maximal optical absorbance of adrenochrome (at 500 nm), which is formed during the oxidation of adrenaline by produced О2− [20], was determined. At the same, the stationary concentration (M) of produced О2− is equal to the concentration of formed adrenochrome, with a molar extinction (E) of up to 750 M-1 cm-1. The stationary concentration (M) of О2− produced by this associate NLP-Nox was determined in the homogeneous phase (in solution) and gas phase by determining the value of A500/E. The optical absorbance of adrenochrome, which is formed during the oxidation of adrenaline by air oxygen, was used as a control.\nThe content of the lipid component (malondialdehyde) in NLP-Nox associate from SI in AD rats was 22.4% higher than that in C rats.\nThe specific content of NLP-Nox was determined by weighing it after deionization and vacuum lyophilization and was expressed in mg per 1 g SI (mg/g).\nThe cellulose of DE-52 («Whatman», England), Sephadex G-100 («Pharmacia», Sweden), adrenaline («Sigma», USA), spectrophotometer «Cary 60» (USA), spectrofluorometer «Perkin-Elmer», (USA), centrifuge K-70D and K-24 «Janetzki» (Germany) were used during the investigation.\nStatistical analysis of the results using the variational statistics method of Student-Fisher was performed to determine the validation criterion (M ± m).", "These О2−-producing associates NLP-Nox from SI of C and AD rats did not undergo PAAG electrophoresis and remained aggregated on the gel tube entry. Indirectly, the purity of these associates is evidenced by the absence of strips of accompanying water-soluble proteins of acidic and basic nature during electrophoresis of these associates’ opalescent solutions on 10% PAAG tubes. The symmetry of the elution diagrams of the Nox-NLP associates after gel filtration through Sephadex G-100 and the unchanged optical spectral index (A280/A400) show the purity of these associates.\nThe specific content of NLP-Nox from SI of C and AD rats was 10,1 ± 0,12 mg/g and 16,6 ± 0,10 mg/g (p < 0,001, n = 6), correspondingly. After heating in boiling water for 10–12 min, the C and AD small intestine associates practically did not reduce nativity and О2−-producing activity. The characteristic absorption maxima for Nox were observed in the optical absorption spectra of NLP-Nox from SI, C, and AD rats in the oxidized and reduced states at pH 9,5 (Fig. 1).\n\nFig. 1Optical absorption spectra of opalescent NLP-Nox associate solutions from SI of C (1) and AD rats (2) at pH 9.5. The spectra (3) were obtained after reduction of (2) by sodium dithionite\n\nOptical absorption spectra of opalescent NLP-Nox associate solutions from SI of C (1) and AD rats (2) at pH 9.5. The spectra (3) were obtained after reduction of (2) by sodium dithionite\nIn comparison to Nox, in the associate of NLP-Nox the “free” Nox from SI in the associate of NLP-Nox had low oxidative/reductive properties, as shown in Fig. 2.\n\nFig. 2The optical absorption spectra of NOx from SI of C (a) and AD (b) rats in oxidized (1) and after reduction by sodium dithionite (2)\n\nThe optical absorption spectra of NOx from SI of C (a) and AD (b) rats in oxidized (1) and after reduction by sodium dithionite (2)\nThese results showed that the Nox in NLP-Nox associate was more easily reduced by sodium dithionite than free Nox from the SI of the C and AD rats. The slow opalescence of NLP-Nox associates from the SI of C and AD rats at pH 10,5 in oxidized and reduced states by sodium dithionite, which differed significantly, as shown in Fig. 3.\n\nFig. 3a – Optical absorption spectra of NLP-Nox associate from the SI of C rats in an oxidized state at pH 10,5 (1) and after reduction by sodium dithionite (2). b - Optical absorption spectra of the NLP-Nox associate from the SI of AD rats in an oxidized state (1) at pH10,5 and after reduction by sodium dithionite (2).\n\na – Optical absorption spectra of NLP-Nox associate from the SI of C rats in an oxidized state at pH 10,5 (1) and after reduction by sodium dithionite (2). b - Optical absorption spectra of the NLP-Nox associate from the SI of AD rats in an oxidized state (1) at pH10,5 and after reduction by sodium dithionite (2).\nAs shown in Fig. 3, the characteristic optical absorption spectra of Nox in the composition of the NLP-Nox associate did not exist in oxidized states at pH 10.5. However, the characteristic optical spectra of Nox in the composition of NLP-Nox were observed at pH 10.5 after the reduction of NLP-Nox associate with potassium dithionite. This phenomenon was observed for the first time in the present study. Reversible changes in the optical absorption spectra of NLP-Nox associates from the SI of the C and AD rats were observed.\nThe effect of NADPH on Nox activation in the gastrointestinal tract has already been revealed [11]. This NADPH, on the other hand, is a component of the NLP in the composition of the О2−-producing NLP-Nox associate from SI. In comparison to similar indicators in C rats, the fluorescence intensity «F» (in relative units) of NADPH in the composition of the associate of NLP-Nox from the SI of AD rats was observed up to 27,5 ± 1,6% (p < 0,005, n = 6) (Fig. 4).\n\nFig. 4Spectrofluorescence spectra of NLP-Nox solution from the SI of C (1) and AD (2) rats (p < 0,005, n = 6)\n\nSpectrofluorescence spectra of NLP-Nox solution from the SI of C (1) and AD (2) rats (p < 0,005, n = 6)\nThe content of the lipid component (malondialdehyde) in NLP-Nox associate from the SI of AD rats is higher to 22,4%, than that in C rats.\nThe stationary concentrations of produced О2− by NLP-Nox associates (1,4 mg/ml) in homogeneous phase (in solution) from C and AD rats were 5,6 ± 0,2 mM and 3,8 ± 0,2 mM (p < 0,05, n = 6).", "Alloxan-induced diabetes animal models have been commonly used to study diabetes in vivo. Studies have shown that alloxan-diabetic rats are hyperglycemic due to damage to the insulin-secreting cells of the pancreas [21]. Many studies have indicated that alloxan alters the gut microbiota community in alloxan-diabetic rats [22]. For the first time, after blowing the NLP-Nox solutions with oxygen (0,2 atm, for 10 min at room temperature), the stationary concentration of produced and transferred into small silicone or glass tube gas phase О2− by the NLP-Nox associate (1,4 mg/ml) from SI of C and AD rats is 4,8 ± 0,3 mM/ml (p < 0,001) and 3,2 ± 0,3 mM/ml (p < 0.001) were observed, correspondingly.\nThe decrease in the stationary concentration of produced О2− by the NLP-Nox associate from SI of the AD rats can be linked to the decrease in the amount of NADPH in the composition of the NLP-Nox associate.\nIn fact, the oxygen is stabilized in the gas phase О2− by forming a coordination band between О2 and О2−. On the other hand, it is known that the gas phase О2− is formed: (a) in air by reducing molecular oxygen with traces of negative metal ions; (b) by an electrochemical method known as a “gas-phase superoxide generator”; (c) under the influence of earth crust radioactivity; and (d) during plant photosynthesis [23].\nThe enzymatically generated monocomponent gas phase О2− from NLP-Nox associate from SI of the rats with regulated and effective stationary concentration of О2− can be used by oxygen mask in experiments and, in the future, in clinics to treat lung infections. However, the monocomponent and regulated stationary concentration of enzymatically produced О2− is preferred. NLP was used as a substrate in the composition of these associates.\nDenaturation and decrease in superoxide production were not observed after boiling the associates in boiling water for 10–12 min. The higher thermostability of the О2−−producing NLP-Nox associates from SI of C and AD rats can be linked to the pulsed increase in temperatures up to 280-300oC during nanoseconds for the transmission of redox metabolic processes [24]. The following are the fundamental significance of the obtained results: (1) the immediate mechanism of the production of О2− by the NLP-Nox associate from rats SI; (2) the formation of the monocomponent and regulated stationary concentration of gas phase О2− form solutions of these NLP-Nox; and (3) the highly reductive properties of Nox in the composition of NLP-Nox associate from rats SI at pH 10.5. A family of NADPH oxidases is especially important for redox signaling and may be amenable to specific therapeutic targets. Nox isoforms are expressed in a cell- and tissue-specific manner, are subject to independent activation and regulation, and may have distinct functions. The complexity of regulation of NADPH oxidases may provide the possibility of targeted therapeutic manipulation in a tissue- and pathway-specific manner at appropriate points in the disease process [25]. The available data suggest that there is a perspective for the using of NCL from donor blood serum, as a potential Nox activator on the surface of immune cells at immunodeficiency in experiment (in which the О2−- producing activity of immune cells decreases [26]. Cells of immune system employ RNS/ROS to kill invading pathogens. A reduction of the redox state of immune cells may render immune cells less effective against invading organisms, resulting in an increased severity of the infection [27] and reduced form of NADPH for various cellular reactions including glutathione (GSH) recycling, superoxide anion production via NADPH oxidase, nitric oxide (NO) synthesis, and cholesterol synthesis [28], therefore quantitative and qualitative changes in NCL from blood serum can be used as new and sensitive diagnostic markers for various pathological conditions in experiments and clinics, in which a decrease in immune activity is observed.\nThe following are the implications of the obtained results:1) the qualitative and quantitative changes of these associates as new sensitive diagnostic tests in various pathological states and diseases, including gastrointestinal disorders and neurodegenerative diseases; 2) the use of gas phase О2− as an antimicrobial and anticancer agent, stimulator of bone marrow stem cell proliferation, and so on.", "Our results suggest that the NLP-Nox associate from the rats’ small intestine is a new О2−-producing thermostable associate in the tissue. As a new AD diagnostic test, it immediately produces О2− with the corresponding quantitative and qualitative changes in C and AD rats." ]
[ null, null, null, null, null, null, null, "results", "discussion", "conclusion" ]
[ "Superoxide", "Associate", "Small intestine", "Alloxan diabetes" ]
Impact of cage position on biomechanical performance of stand-alone lateral lumbar interbody fusion: a finite element analysis.
36258213
This study aimed to compare the biomechanical performance of various cage positions in stand-alone lateral lumbar interbody fusion(SA LLIF).
BACKGROUND
An intact finite element model of the L3-L5 was reconstructed. The model was verified and analyzed. Through changing the position of the cage, SA LLIF was established in four directions: anterior placement(AP), middle placement(MP), posterior placement(PP), oblique placement(OP). A 400 N vertical axial pre-load was imposed on the superior surface of L3 and a 10 N/m moment was applied on the L3 superior surface along the radial direction to simulate movements of flexion, extension, lateral bending, and axial rotation. Various biomechanical parameters were evaluated for intact and implanted models in all loading conditions, including the range of motion (ROM) and maximum stress.
METHODS
In the SA LLIF models, the ROM of L4-5 was reduced by 84.21-89.03% in flexion, 72.64-82.26% in extension, 92.5-95.85% in right and left lateral bending, and 87.22-92.77% in right and left axial rotation, respectively. Meanwhile, ROM of L3-4 was mildly increased by an average of 9.6% in all motion directions. Almost all stress peaks were increased after SA LLIF, including adjacent disc, facet joints, and endplates. MP had lower stress peaks of cage and endplates in most motion modes. In terms of the stress on facet joints and disc of the cephalad segment, MP had the smallest increment.
RESULTS
In our study, SA LLIF risked accelerating the adjacent segment degeneration. The cage position had an influence on the distribution of endplate stress and the magnitude of facet joint stress. Compared with other positions, MP had the slightest effect on the stress in the adjacent facet joints. Meanwhile, MP seems to play an important role in reducing the risk of cage subsidence.
CONCLUSION
[ "Humans", "Finite Element Analysis", "Lumbar Vertebrae", "Biomechanical Phenomena", "Spinal Fusion", "Range of Motion, Articular" ]
9578219
null
null
null
null
Result
[SUBTITLE] Model validity and mesh convergence verification [SUBSECTION] Figure 3 indicates the result of model verification. The convergence of the model was verified by analyzing the maximum deformation and von-Mises stress in three element sizes (1 mm, 1.5 mm, 2 mm) models. The resolution difference between the predicted results of the 1.5 mm mesh size and 1 mm mesh size was < 5%, the 1.5 mm mesh was considered convergent (Fig. 4). Owing to modeling inconsistencies, some individual values were inconsistent with previous finite element results, however, all values in different conditions were within the vitro range (Yamamoto [28]). Fig. 3Comparison of ROM between the current intact finite element model and previous studies:(a) L3-4, (b) L4-5; L, left;R, right Comparison of ROM between the current intact finite element model and previous studies:(a) L3-4, (b) L4-5; L, left;R, right Fig. 4Result of meshing sensitivity study Result of meshing sensitivity study Figure 3 indicates the result of model verification. The convergence of the model was verified by analyzing the maximum deformation and von-Mises stress in three element sizes (1 mm, 1.5 mm, 2 mm) models. The resolution difference between the predicted results of the 1.5 mm mesh size and 1 mm mesh size was < 5%, the 1.5 mm mesh was considered convergent (Fig. 4). Owing to modeling inconsistencies, some individual values were inconsistent with previous finite element results, however, all values in different conditions were within the vitro range (Yamamoto [28]). Fig. 3Comparison of ROM between the current intact finite element model and previous studies:(a) L3-4, (b) L4-5; L, left;R, right Comparison of ROM between the current intact finite element model and previous studies:(a) L3-4, (b) L4-5; L, left;R, right Fig. 4Result of meshing sensitivity study Result of meshing sensitivity study [SUBTITLE] ROM of L4-5 and L3-4 [SUBSECTION] Figure 5 shows ROM of surgical and adjacent segment under various engineering conditions. When LLIF cage was used, a dramatic decline loss of motion at surgical level L4-5 occurred regardless the placement direction of intervertebral cage. The ROM of L4-5 was reduced by 84.21–89.03% in flexion, 72.64–82.26% in extension, 92.5-95.85% in right and left lateral bending, and 87.22–92.77% in right and left axial rotation, respectively. Meanwhile, ROM of L3-4 after LLIF was mildly increased by an average of 9.6% in all motion directions. Fig. 5Changes in ROM before and after surgery: (a) L3-4, (b) L4-5; intact model (IM). Changes in ROM before and after surgery: (a) L3-4, (b) L4-5; intact model (IM). Figure 5 shows ROM of surgical and adjacent segment under various engineering conditions. When LLIF cage was used, a dramatic decline loss of motion at surgical level L4-5 occurred regardless the placement direction of intervertebral cage. The ROM of L4-5 was reduced by 84.21–89.03% in flexion, 72.64–82.26% in extension, 92.5-95.85% in right and left lateral bending, and 87.22–92.77% in right and left axial rotation, respectively. Meanwhile, ROM of L3-4 after LLIF was mildly increased by an average of 9.6% in all motion directions. Fig. 5Changes in ROM before and after surgery: (a) L3-4, (b) L4-5; intact model (IM). Changes in ROM before and after surgery: (a) L3-4, (b) L4-5; intact model (IM). [SUBTITLE] Equivalent (von Mises) stress of endplate and cage [SUBSECTION] Interbody fusion cage subsidence and displacement often occur in the upper endplate postoperatively. Figure 6 shows the stress nephogram of the L5 superior endplate. The distribution of stress was consistent with the contact surface between cage and endplate. Figure 7(a-d) represents the maximum stress variation of cage and endplate. MP had the smallest stress peaks in most motion modes. The cage stress of MP was 43.95Mpa in flexion, 38.05Mpa in extension, 25.68Mpa in left bending, 43.53Mpa in right bending, 19.74Mpa in left rotation, and 22.44Mpa in right rotation. Fig. 6Stress nephogram of the L5 superior endplate Stress nephogram of the L5 superior endplate Fig. 7Details of maximum stress:(a) cage, (b) L5 superior endplate, (c) L4 superior endplate, (d) L4 inferior endplate, (e) Intervertebral disc, (f) facet joints Details of maximum stress:(a) cage, (b) L5 superior endplate, (c) L4 superior endplate, (d) L4 inferior endplate, (e) Intervertebral disc, (f) facet joints Interbody fusion cage subsidence and displacement often occur in the upper endplate postoperatively. Figure 6 shows the stress nephogram of the L5 superior endplate. The distribution of stress was consistent with the contact surface between cage and endplate. Figure 7(a-d) represents the maximum stress variation of cage and endplate. MP had the smallest stress peaks in most motion modes. The cage stress of MP was 43.95Mpa in flexion, 38.05Mpa in extension, 25.68Mpa in left bending, 43.53Mpa in right bending, 19.74Mpa in left rotation, and 22.44Mpa in right rotation. Fig. 6Stress nephogram of the L5 superior endplate Stress nephogram of the L5 superior endplate Fig. 7Details of maximum stress:(a) cage, (b) L5 superior endplate, (c) L4 superior endplate, (d) L4 inferior endplate, (e) Intervertebral disc, (f) facet joints Details of maximum stress:(a) cage, (b) L5 superior endplate, (c) L4 superior endplate, (d) L4 inferior endplate, (e) Intervertebral disc, (f) facet joints [SUBTITLE] Stress of facet joints and proximal intervertebral disc [SUBSECTION] The maximum stress of facet joints and intervertebral disc are displayed in Fig. 7(e, f). Almost all stress peaks were increased after SA LLIF, including adjacent disc, facet joints, and endplates. As shown in our result, MP had the slightest tendency to raise the stress. The facet joints stress of MP was 7.93Mpa in flexion, 9.11Mpa in extension, 10.09Mpa in left bending, 12.13Mpa in right bending, 3.87Mpa in left rotation, and 4.43Mpa in right rotation. The L3/4 intervertebral disc stress of MP was 1.72 Mpa in flexion, 1.88Mpa in extension, 2.79Mpa in left bending, 2.59Mpa in right bending, 1.31Mpa in left rotation, and 1. 42Mpa in right rotation. The maximum stress of facet joints and intervertebral disc are displayed in Fig. 7(e, f). Almost all stress peaks were increased after SA LLIF, including adjacent disc, facet joints, and endplates. As shown in our result, MP had the slightest tendency to raise the stress. The facet joints stress of MP was 7.93Mpa in flexion, 9.11Mpa in extension, 10.09Mpa in left bending, 12.13Mpa in right bending, 3.87Mpa in left rotation, and 4.43Mpa in right rotation. The L3/4 intervertebral disc stress of MP was 1.72 Mpa in flexion, 1.88Mpa in extension, 2.79Mpa in left bending, 2.59Mpa in right bending, 1.31Mpa in left rotation, and 1. 42Mpa in right rotation.
Conclusion
In our study, SA LLIF risked accelerating the adjacent segment degeneration. The cage position had an influence on the distribution of endplate stress and the magnitude of facet joint stress. Compared with other positions, MP had the slightest effect on the stress in the adjacent facet joints. Meanwhile, MP seems to play an important role in reducing the risk of cage subsidence.
[ "Background", "Building an intact model", "Development of SA LLIF model", "Parameter setting and meshing", "Boundary conditions and loads", "Evaluation criteria", "Model validity and mesh convergence verification", "ROM of L4-5 and L3-4", "Equivalent (von Mises) stress of endplate and cage", "Stress of facet joints and proximal intervertebral disc" ]
[ "In light of the popularity of minimally invasive Intervertebral fusion surgery, lateral lumbar interbody fusion (LLIF) is increasingly being chosen by surgeons. This technology has been progressively matured and optimized since it was systematically reported in 2006 [1, 2]. LLIF was designed as a retroperitoneal approach to reduce the risk of vascular injury seen with anterior lumbar interbody fusion(ALIF), avoid the disruption of spinal stability seen with transforaminal lumbar interbody fusion (TLIF), and prevent the muscular trauma seen with posterior lumbar interbody fusion (PLIF) [3–5]. Meanwhile, a larger size cage is permitted during LLIF, which can improve the fusion rate [6]. Additionally, this operation has particular advantages in therapy for adjacent segment disease (ASD) and revision surgery, when a conventional posterior decompression has a relatively high dural injury rate, whereas epidural damage during a lateral fusion operation is extremely uncommon [7, 8]. Another minimally invasive superiority of LLIF is that its anatomic approach is away from previous surgical incision and granulation tissue to reduce intraoperative bleeding risk. Louie PK et al. [9–11] demonstrated the availability of the lateral approach procedure for ASD through biomechanical experiments.\nThe current literature is divided on the rationalization of auxiliary internal fixation. A few experiments [12, 13] have indicated assisted internal fixation exhibits more range of motion (ROM) reduction than stand-alone approach under the same mechanical conditions, especially cage plus bilateral pedicle screw fixation (PSF). Though additional fixation could enhance the mechanical stability to some extent, it also increased the risk of adjacent segment degeneration simultaneously. In practice, since stabilizing structures remain intact, it is suggested that stand-alone lateral lumbar interbody fusion (SA LLIF) can be used for appropriate indications [14]. Marchi et al. [15] found segmental lordosis could be restored by the stand-alone cage. Considering subjective factors such as intraoperative endplate damage, SA LLIF was associated with fusion failure, and higher rate of cage displacement, and high possibility of revision operation due to recurrence of symptoms [16]. To reduce the detrimental impact, placement and size of cage are the uppermost consideration during SA LLIF. Yuan et al. [17, 18] demonstrated that cage size correlated strongly with bone fusion and cage subsidence. And yet in respect to clinical outcomes, the position of interbody fusion cage in LLIF is controversial [19, 20], Qiao et al. [21, 22] reported that disc height and segmental angle could be influenced by cage position. However, little experiment has been designed to investigate the cage position in SA LLIF from a biomechanical point of view.\nAccordingly, three-dimensional digital models of lumbar were created to simulate several surgical conditions. The objective of this study was to compare biomechanical performance of various cage positions in SA LLIF by finite element analysis (FEA). The von Mises stress and displacement were used as risk parameters associated with subsidence and adjacent segment degeneration.", "The volunteer we recruited (40-year-old, female, 60Kg, 160 cm) did not have any spine or systemic disease. All procedures were in accordance with the ethical standards of the institutional and national research committee.Written informed consent was acquired before the examination. The normal L3-5 segments of the volunteer were scanned using a computed tomography scanner (Philips, Netherlands, 128 spiral, 120kv, 81mAs, 279mA, Pitch 1. 38). A series of images (Slice Thk 0. 5 mm ) were exported and saved as Digital Imaging and Communications in Medicine (DICOM) format. These images were imported Mimics20. 0 (Materialise Inc, Leuven, Belgium). After predefined threshold, region growing, mask modifying and preliminary borders smoothing, the project was outputted in STL format. This model was imported into Geomagic-Warp2017 (3DSystems Inc, Rock Hill, SC, USA) software for advanced smoothing, adjusting polygon mesh and fitting surface and also integrated into the Solidworks2017 (Dassault Systemes, Vélizy-Villacoublay, France).\nCancellous bone and cortical bone were combined by Boolean logic operations;the thickness of cortical bone and bony endplates were set to 1 mm [23]. Facet joints and intervertebral discs were created in part pattern; articular cartilage filled the joint space with a thickness of 0. 2 mm to withstand rotational stress; the volume ratio of annulus fibrosus and nucleus pulposus was approximately 1:1 (Fig. 1).\n\nFig. 1Details of models: (a) intact model, (b) intervertebral disc, (c) facet joint and articular cartilage, (d) uneven meshes of vertebral body\n\nDetails of models: (a) intact model, (b) intervertebral disc, (c) facet joint and articular cartilage, (d) uneven meshes of vertebral body", "Due to a higher incidence of degeneration, L4-L5 segment, this segment was selected to simulate intervertebral fusion surgery. The model of interbody fusion cage was reconstructed with the prototype provided by Zimmer (8-10 mm height, 45 mm length, 17 mm width). The LLIF cage was placed into L4/5 disc space in four directions:anterior placement (AP), middle placement (MP), posterior placement (PP), oblique placement (OP) (Fig. 2). The cage of AP was close to the anterior edge of the endplate and parallel to the coronal midline. The cage of MP was close to the coronal midline of the endplate and perpendicular to the sagittal midline. The cage of PP was close to the posterior edge of the endplate and parallel to the coronal midline. The cage of OP was close to the centre of endplate and at 45 degree to the coronal midline. The cartilaginous endplate in L4/5 was removed, anterior longitudinal ligament and posterior structure were preserved in the meantime. The gap between the cage and the bony endplate was filled with cancellous bone. Some simplification of details were applicable in the modeling process, considering fault tolerance and reliability issues. In the end, all of the parts were detected by interference to evaluate their process quality, then were imported into Ansys 17. 0 (Ansys, Canonsburg, PA, USA).\n\nFig. 2Different positions of cage: (a) anterior placement (AP), (b) middle placement(MP), (c) posterior placement (PP), (d) oblique placement (OP).\n\nDifferent positions of cage: (a) anterior placement (AP), (b) middle placement(MP), (c) posterior placement (PP), (d) oblique placement (OP).", "We created a simplified model in which the various materials were recognized to be homogeneous. According to the previously set material property parameters [24, 25], the model including cortical bone, cancellous bone, cartilage endplates and interbody fusion cage were assigned to different elastic modulus and Poisson’s ratio. The details are presented in Table 1. The start and end points of the ligament were set according to anatomical characteristics, and their material properties were represented using cable elements such as spring patterns [26]. To improve the calculation accuracy, mixed mesh method was adopted to build meshes of the lumbar model, and different mesh types were established. This method allowed for more nodes and elements, which resulted in higher accuracy of biomechanical analysis (Fig. 1). The minimum effective size of four-node tetrahedron body element was 1.0 mm, which was used for facet joints.\n\nTable 1Mechanical parameters of spinal modelsMaterialYoung’s modulus (MPa)Poisson’s ratioCross-sectional area (mm2)Cancellous bone1000.2-Cortical bone12,0000.3-Posterior bone35000.25-Articular cartilages350.4-Endplate250.25-Annulus fibrosus4.20.45-Nucleus pulposus10.499-Spinal cage36000.25-Capsular ligament7.50.3-posterior longitudinal ligament10 (< 11%) 20 (> 11%)-20Ligament flavum15 (< 6.2%) 19 (> 6.2%)-40Interspinous ligament10 (< 14%) 12 (> 14%)-40Superspinous ligament8 (< 20%) 15 (> 20%)-30Intertansverse ligament10 (< 18%) 59 (> 18%)-1.8Anterior longitudinal ligament7.8 (< 12%) 20 (> 12%)-63.7\n\nMechanical parameters of spinal models", "L5 bottom was set as a fixed support surface, and six degrees of freedom were constrained. A vertical load of 400 N and the bending moment of 10 Nm were applied to the upper surface of the L3 vertebra to simulate movements of flexion, extension, lateral bending, and axial rotation. Contact types were defined as ‘Bonded’, except for the contact type between facet joint and cartilage, which was defined as ‘Frictionless’ [27].", "Firstly, the range of motion (ROM) of all functional spinal units (FSU) were recorded, including surgical segment and adjacent segment. Secondly, the equivalent(von Mises)stress of endplate, facet joint and intervertebral fusion cage were observed.", "Figure 3 indicates the result of model verification. The convergence of the model was verified by analyzing the maximum deformation and von-Mises stress in three element sizes (1 mm, 1.5 mm, 2 mm) models. The resolution difference between the predicted results of the 1.5 mm mesh size and 1 mm mesh size was < 5%, the 1.5 mm mesh was considered convergent (Fig. 4). Owing to modeling inconsistencies, some individual values were inconsistent with previous finite element results, however, all values in different conditions were within the vitro range (Yamamoto [28]).\n\nFig. 3Comparison of ROM between the current intact finite element model and previous studies:(a) L3-4, (b) L4-5; L, left;R, right\n\nComparison of ROM between the current intact finite element model and previous studies:(a) L3-4, (b) L4-5; L, left;R, right\n\nFig. 4Result of meshing sensitivity study\n\nResult of meshing sensitivity study", "Figure 5 shows ROM of surgical and adjacent segment under various engineering conditions. When LLIF cage was used, a dramatic decline loss of motion at surgical level L4-5 occurred regardless the placement direction of intervertebral cage. The ROM of L4-5 was reduced by 84.21–89.03% in flexion, 72.64–82.26% in extension, 92.5-95.85% in right and left lateral bending, and 87.22–92.77% in right and left axial rotation, respectively. Meanwhile, ROM of L3-4 after LLIF was mildly increased by an average of 9.6% in all motion directions.\n\nFig. 5Changes in ROM before and after surgery: (a) L3-4, (b) L4-5; intact model (IM).\n\nChanges in ROM before and after surgery: (a) L3-4, (b) L4-5; intact model (IM).", "Interbody fusion cage subsidence and displacement often occur in the upper endplate postoperatively. Figure 6 shows the stress nephogram of the L5 superior endplate. The distribution of stress was consistent with the contact surface between cage and endplate. Figure 7(a-d) represents the maximum stress variation of cage and endplate. MP had the smallest stress peaks in most motion modes. The cage stress of MP was 43.95Mpa in flexion, 38.05Mpa in extension, 25.68Mpa in left bending, 43.53Mpa in right bending, 19.74Mpa in left rotation, and 22.44Mpa in right rotation.\n\nFig. 6Stress nephogram of the L5 superior endplate\n\nStress nephogram of the L5 superior endplate\n\nFig. 7Details of maximum stress:(a) cage, (b) L5 superior endplate, (c) L4 superior endplate, (d) L4 inferior endplate, (e) Intervertebral disc, (f) facet joints\n\nDetails of maximum stress:(a) cage, (b) L5 superior endplate, (c) L4 superior endplate, (d) L4 inferior endplate, (e) Intervertebral disc, (f) facet joints", "The maximum stress of facet joints and intervertebral disc are displayed in Fig. 7(e, f). Almost all stress peaks were increased after SA LLIF, including adjacent disc, facet joints, and endplates. As shown in our result, MP had the slightest tendency to raise the stress. The facet joints stress of MP was 7.93Mpa in flexion, 9.11Mpa in extension, 10.09Mpa in left bending, 12.13Mpa in right bending, 3.87Mpa in left rotation, and 4.43Mpa in right rotation. The L3/4 intervertebral disc stress of MP was 1.72 Mpa in flexion, 1.88Mpa in extension, 2.79Mpa in left bending, 2.59Mpa in right bending, 1.31Mpa in left rotation, and 1. 42Mpa in right rotation." ]
[ null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Materials and methods", "Building an intact model", "Development of SA LLIF model", "Parameter setting and meshing", "Boundary conditions and loads", "Evaluation criteria", "Result", "Model validity and mesh convergence verification", "ROM of L4-5 and L3-4", "Equivalent (von Mises) stress of endplate and cage", "Stress of facet joints and proximal intervertebral disc", "Discussion", "Conclusion" ]
[ "In light of the popularity of minimally invasive Intervertebral fusion surgery, lateral lumbar interbody fusion (LLIF) is increasingly being chosen by surgeons. This technology has been progressively matured and optimized since it was systematically reported in 2006 [1, 2]. LLIF was designed as a retroperitoneal approach to reduce the risk of vascular injury seen with anterior lumbar interbody fusion(ALIF), avoid the disruption of spinal stability seen with transforaminal lumbar interbody fusion (TLIF), and prevent the muscular trauma seen with posterior lumbar interbody fusion (PLIF) [3–5]. Meanwhile, a larger size cage is permitted during LLIF, which can improve the fusion rate [6]. Additionally, this operation has particular advantages in therapy for adjacent segment disease (ASD) and revision surgery, when a conventional posterior decompression has a relatively high dural injury rate, whereas epidural damage during a lateral fusion operation is extremely uncommon [7, 8]. Another minimally invasive superiority of LLIF is that its anatomic approach is away from previous surgical incision and granulation tissue to reduce intraoperative bleeding risk. Louie PK et al. [9–11] demonstrated the availability of the lateral approach procedure for ASD through biomechanical experiments.\nThe current literature is divided on the rationalization of auxiliary internal fixation. A few experiments [12, 13] have indicated assisted internal fixation exhibits more range of motion (ROM) reduction than stand-alone approach under the same mechanical conditions, especially cage plus bilateral pedicle screw fixation (PSF). Though additional fixation could enhance the mechanical stability to some extent, it also increased the risk of adjacent segment degeneration simultaneously. In practice, since stabilizing structures remain intact, it is suggested that stand-alone lateral lumbar interbody fusion (SA LLIF) can be used for appropriate indications [14]. Marchi et al. [15] found segmental lordosis could be restored by the stand-alone cage. Considering subjective factors such as intraoperative endplate damage, SA LLIF was associated with fusion failure, and higher rate of cage displacement, and high possibility of revision operation due to recurrence of symptoms [16]. To reduce the detrimental impact, placement and size of cage are the uppermost consideration during SA LLIF. Yuan et al. [17, 18] demonstrated that cage size correlated strongly with bone fusion and cage subsidence. And yet in respect to clinical outcomes, the position of interbody fusion cage in LLIF is controversial [19, 20], Qiao et al. [21, 22] reported that disc height and segmental angle could be influenced by cage position. However, little experiment has been designed to investigate the cage position in SA LLIF from a biomechanical point of view.\nAccordingly, three-dimensional digital models of lumbar were created to simulate several surgical conditions. The objective of this study was to compare biomechanical performance of various cage positions in SA LLIF by finite element analysis (FEA). The von Mises stress and displacement were used as risk parameters associated with subsidence and adjacent segment degeneration.", "[SUBTITLE] Building an intact model [SUBSECTION] The volunteer we recruited (40-year-old, female, 60Kg, 160 cm) did not have any spine or systemic disease. All procedures were in accordance with the ethical standards of the institutional and national research committee.Written informed consent was acquired before the examination. The normal L3-5 segments of the volunteer were scanned using a computed tomography scanner (Philips, Netherlands, 128 spiral, 120kv, 81mAs, 279mA, Pitch 1. 38). A series of images (Slice Thk 0. 5 mm ) were exported and saved as Digital Imaging and Communications in Medicine (DICOM) format. These images were imported Mimics20. 0 (Materialise Inc, Leuven, Belgium). After predefined threshold, region growing, mask modifying and preliminary borders smoothing, the project was outputted in STL format. This model was imported into Geomagic-Warp2017 (3DSystems Inc, Rock Hill, SC, USA) software for advanced smoothing, adjusting polygon mesh and fitting surface and also integrated into the Solidworks2017 (Dassault Systemes, Vélizy-Villacoublay, France).\nCancellous bone and cortical bone were combined by Boolean logic operations;the thickness of cortical bone and bony endplates were set to 1 mm [23]. Facet joints and intervertebral discs were created in part pattern; articular cartilage filled the joint space with a thickness of 0. 2 mm to withstand rotational stress; the volume ratio of annulus fibrosus and nucleus pulposus was approximately 1:1 (Fig. 1).\n\nFig. 1Details of models: (a) intact model, (b) intervertebral disc, (c) facet joint and articular cartilage, (d) uneven meshes of vertebral body\n\nDetails of models: (a) intact model, (b) intervertebral disc, (c) facet joint and articular cartilage, (d) uneven meshes of vertebral body\nThe volunteer we recruited (40-year-old, female, 60Kg, 160 cm) did not have any spine or systemic disease. All procedures were in accordance with the ethical standards of the institutional and national research committee.Written informed consent was acquired before the examination. The normal L3-5 segments of the volunteer were scanned using a computed tomography scanner (Philips, Netherlands, 128 spiral, 120kv, 81mAs, 279mA, Pitch 1. 38). A series of images (Slice Thk 0. 5 mm ) were exported and saved as Digital Imaging and Communications in Medicine (DICOM) format. These images were imported Mimics20. 0 (Materialise Inc, Leuven, Belgium). After predefined threshold, region growing, mask modifying and preliminary borders smoothing, the project was outputted in STL format. This model was imported into Geomagic-Warp2017 (3DSystems Inc, Rock Hill, SC, USA) software for advanced smoothing, adjusting polygon mesh and fitting surface and also integrated into the Solidworks2017 (Dassault Systemes, Vélizy-Villacoublay, France).\nCancellous bone and cortical bone were combined by Boolean logic operations;the thickness of cortical bone and bony endplates were set to 1 mm [23]. Facet joints and intervertebral discs were created in part pattern; articular cartilage filled the joint space with a thickness of 0. 2 mm to withstand rotational stress; the volume ratio of annulus fibrosus and nucleus pulposus was approximately 1:1 (Fig. 1).\n\nFig. 1Details of models: (a) intact model, (b) intervertebral disc, (c) facet joint and articular cartilage, (d) uneven meshes of vertebral body\n\nDetails of models: (a) intact model, (b) intervertebral disc, (c) facet joint and articular cartilage, (d) uneven meshes of vertebral body\n[SUBTITLE] Development of SA LLIF model [SUBSECTION] Due to a higher incidence of degeneration, L4-L5 segment, this segment was selected to simulate intervertebral fusion surgery. The model of interbody fusion cage was reconstructed with the prototype provided by Zimmer (8-10 mm height, 45 mm length, 17 mm width). The LLIF cage was placed into L4/5 disc space in four directions:anterior placement (AP), middle placement (MP), posterior placement (PP), oblique placement (OP) (Fig. 2). The cage of AP was close to the anterior edge of the endplate and parallel to the coronal midline. The cage of MP was close to the coronal midline of the endplate and perpendicular to the sagittal midline. The cage of PP was close to the posterior edge of the endplate and parallel to the coronal midline. The cage of OP was close to the centre of endplate and at 45 degree to the coronal midline. The cartilaginous endplate in L4/5 was removed, anterior longitudinal ligament and posterior structure were preserved in the meantime. The gap between the cage and the bony endplate was filled with cancellous bone. Some simplification of details were applicable in the modeling process, considering fault tolerance and reliability issues. In the end, all of the parts were detected by interference to evaluate their process quality, then were imported into Ansys 17. 0 (Ansys, Canonsburg, PA, USA).\n\nFig. 2Different positions of cage: (a) anterior placement (AP), (b) middle placement(MP), (c) posterior placement (PP), (d) oblique placement (OP).\n\nDifferent positions of cage: (a) anterior placement (AP), (b) middle placement(MP), (c) posterior placement (PP), (d) oblique placement (OP).\nDue to a higher incidence of degeneration, L4-L5 segment, this segment was selected to simulate intervertebral fusion surgery. The model of interbody fusion cage was reconstructed with the prototype provided by Zimmer (8-10 mm height, 45 mm length, 17 mm width). The LLIF cage was placed into L4/5 disc space in four directions:anterior placement (AP), middle placement (MP), posterior placement (PP), oblique placement (OP) (Fig. 2). The cage of AP was close to the anterior edge of the endplate and parallel to the coronal midline. The cage of MP was close to the coronal midline of the endplate and perpendicular to the sagittal midline. The cage of PP was close to the posterior edge of the endplate and parallel to the coronal midline. The cage of OP was close to the centre of endplate and at 45 degree to the coronal midline. The cartilaginous endplate in L4/5 was removed, anterior longitudinal ligament and posterior structure were preserved in the meantime. The gap between the cage and the bony endplate was filled with cancellous bone. Some simplification of details were applicable in the modeling process, considering fault tolerance and reliability issues. In the end, all of the parts were detected by interference to evaluate their process quality, then were imported into Ansys 17. 0 (Ansys, Canonsburg, PA, USA).\n\nFig. 2Different positions of cage: (a) anterior placement (AP), (b) middle placement(MP), (c) posterior placement (PP), (d) oblique placement (OP).\n\nDifferent positions of cage: (a) anterior placement (AP), (b) middle placement(MP), (c) posterior placement (PP), (d) oblique placement (OP).\n[SUBTITLE] Parameter setting and meshing [SUBSECTION] We created a simplified model in which the various materials were recognized to be homogeneous. According to the previously set material property parameters [24, 25], the model including cortical bone, cancellous bone, cartilage endplates and interbody fusion cage were assigned to different elastic modulus and Poisson’s ratio. The details are presented in Table 1. The start and end points of the ligament were set according to anatomical characteristics, and their material properties were represented using cable elements such as spring patterns [26]. To improve the calculation accuracy, mixed mesh method was adopted to build meshes of the lumbar model, and different mesh types were established. This method allowed for more nodes and elements, which resulted in higher accuracy of biomechanical analysis (Fig. 1). The minimum effective size of four-node tetrahedron body element was 1.0 mm, which was used for facet joints.\n\nTable 1Mechanical parameters of spinal modelsMaterialYoung’s modulus (MPa)Poisson’s ratioCross-sectional area (mm2)Cancellous bone1000.2-Cortical bone12,0000.3-Posterior bone35000.25-Articular cartilages350.4-Endplate250.25-Annulus fibrosus4.20.45-Nucleus pulposus10.499-Spinal cage36000.25-Capsular ligament7.50.3-posterior longitudinal ligament10 (< 11%) 20 (> 11%)-20Ligament flavum15 (< 6.2%) 19 (> 6.2%)-40Interspinous ligament10 (< 14%) 12 (> 14%)-40Superspinous ligament8 (< 20%) 15 (> 20%)-30Intertansverse ligament10 (< 18%) 59 (> 18%)-1.8Anterior longitudinal ligament7.8 (< 12%) 20 (> 12%)-63.7\n\nMechanical parameters of spinal models\nWe created a simplified model in which the various materials were recognized to be homogeneous. According to the previously set material property parameters [24, 25], the model including cortical bone, cancellous bone, cartilage endplates and interbody fusion cage were assigned to different elastic modulus and Poisson’s ratio. The details are presented in Table 1. The start and end points of the ligament were set according to anatomical characteristics, and their material properties were represented using cable elements such as spring patterns [26]. To improve the calculation accuracy, mixed mesh method was adopted to build meshes of the lumbar model, and different mesh types were established. This method allowed for more nodes and elements, which resulted in higher accuracy of biomechanical analysis (Fig. 1). The minimum effective size of four-node tetrahedron body element was 1.0 mm, which was used for facet joints.\n\nTable 1Mechanical parameters of spinal modelsMaterialYoung’s modulus (MPa)Poisson’s ratioCross-sectional area (mm2)Cancellous bone1000.2-Cortical bone12,0000.3-Posterior bone35000.25-Articular cartilages350.4-Endplate250.25-Annulus fibrosus4.20.45-Nucleus pulposus10.499-Spinal cage36000.25-Capsular ligament7.50.3-posterior longitudinal ligament10 (< 11%) 20 (> 11%)-20Ligament flavum15 (< 6.2%) 19 (> 6.2%)-40Interspinous ligament10 (< 14%) 12 (> 14%)-40Superspinous ligament8 (< 20%) 15 (> 20%)-30Intertansverse ligament10 (< 18%) 59 (> 18%)-1.8Anterior longitudinal ligament7.8 (< 12%) 20 (> 12%)-63.7\n\nMechanical parameters of spinal models\n[SUBTITLE] Boundary conditions and loads [SUBSECTION] L5 bottom was set as a fixed support surface, and six degrees of freedom were constrained. A vertical load of 400 N and the bending moment of 10 Nm were applied to the upper surface of the L3 vertebra to simulate movements of flexion, extension, lateral bending, and axial rotation. Contact types were defined as ‘Bonded’, except for the contact type between facet joint and cartilage, which was defined as ‘Frictionless’ [27].\nL5 bottom was set as a fixed support surface, and six degrees of freedom were constrained. A vertical load of 400 N and the bending moment of 10 Nm were applied to the upper surface of the L3 vertebra to simulate movements of flexion, extension, lateral bending, and axial rotation. Contact types were defined as ‘Bonded’, except for the contact type between facet joint and cartilage, which was defined as ‘Frictionless’ [27].\n[SUBTITLE] Evaluation criteria [SUBSECTION] Firstly, the range of motion (ROM) of all functional spinal units (FSU) were recorded, including surgical segment and adjacent segment. Secondly, the equivalent(von Mises)stress of endplate, facet joint and intervertebral fusion cage were observed.\nFirstly, the range of motion (ROM) of all functional spinal units (FSU) were recorded, including surgical segment and adjacent segment. Secondly, the equivalent(von Mises)stress of endplate, facet joint and intervertebral fusion cage were observed.", "The volunteer we recruited (40-year-old, female, 60Kg, 160 cm) did not have any spine or systemic disease. All procedures were in accordance with the ethical standards of the institutional and national research committee.Written informed consent was acquired before the examination. The normal L3-5 segments of the volunteer were scanned using a computed tomography scanner (Philips, Netherlands, 128 spiral, 120kv, 81mAs, 279mA, Pitch 1. 38). A series of images (Slice Thk 0. 5 mm ) were exported and saved as Digital Imaging and Communications in Medicine (DICOM) format. These images were imported Mimics20. 0 (Materialise Inc, Leuven, Belgium). After predefined threshold, region growing, mask modifying and preliminary borders smoothing, the project was outputted in STL format. This model was imported into Geomagic-Warp2017 (3DSystems Inc, Rock Hill, SC, USA) software for advanced smoothing, adjusting polygon mesh and fitting surface and also integrated into the Solidworks2017 (Dassault Systemes, Vélizy-Villacoublay, France).\nCancellous bone and cortical bone were combined by Boolean logic operations;the thickness of cortical bone and bony endplates were set to 1 mm [23]. Facet joints and intervertebral discs were created in part pattern; articular cartilage filled the joint space with a thickness of 0. 2 mm to withstand rotational stress; the volume ratio of annulus fibrosus and nucleus pulposus was approximately 1:1 (Fig. 1).\n\nFig. 1Details of models: (a) intact model, (b) intervertebral disc, (c) facet joint and articular cartilage, (d) uneven meshes of vertebral body\n\nDetails of models: (a) intact model, (b) intervertebral disc, (c) facet joint and articular cartilage, (d) uneven meshes of vertebral body", "Due to a higher incidence of degeneration, L4-L5 segment, this segment was selected to simulate intervertebral fusion surgery. The model of interbody fusion cage was reconstructed with the prototype provided by Zimmer (8-10 mm height, 45 mm length, 17 mm width). The LLIF cage was placed into L4/5 disc space in four directions:anterior placement (AP), middle placement (MP), posterior placement (PP), oblique placement (OP) (Fig. 2). The cage of AP was close to the anterior edge of the endplate and parallel to the coronal midline. The cage of MP was close to the coronal midline of the endplate and perpendicular to the sagittal midline. The cage of PP was close to the posterior edge of the endplate and parallel to the coronal midline. The cage of OP was close to the centre of endplate and at 45 degree to the coronal midline. The cartilaginous endplate in L4/5 was removed, anterior longitudinal ligament and posterior structure were preserved in the meantime. The gap between the cage and the bony endplate was filled with cancellous bone. Some simplification of details were applicable in the modeling process, considering fault tolerance and reliability issues. In the end, all of the parts were detected by interference to evaluate their process quality, then were imported into Ansys 17. 0 (Ansys, Canonsburg, PA, USA).\n\nFig. 2Different positions of cage: (a) anterior placement (AP), (b) middle placement(MP), (c) posterior placement (PP), (d) oblique placement (OP).\n\nDifferent positions of cage: (a) anterior placement (AP), (b) middle placement(MP), (c) posterior placement (PP), (d) oblique placement (OP).", "We created a simplified model in which the various materials were recognized to be homogeneous. According to the previously set material property parameters [24, 25], the model including cortical bone, cancellous bone, cartilage endplates and interbody fusion cage were assigned to different elastic modulus and Poisson’s ratio. The details are presented in Table 1. The start and end points of the ligament were set according to anatomical characteristics, and their material properties were represented using cable elements such as spring patterns [26]. To improve the calculation accuracy, mixed mesh method was adopted to build meshes of the lumbar model, and different mesh types were established. This method allowed for more nodes and elements, which resulted in higher accuracy of biomechanical analysis (Fig. 1). The minimum effective size of four-node tetrahedron body element was 1.0 mm, which was used for facet joints.\n\nTable 1Mechanical parameters of spinal modelsMaterialYoung’s modulus (MPa)Poisson’s ratioCross-sectional area (mm2)Cancellous bone1000.2-Cortical bone12,0000.3-Posterior bone35000.25-Articular cartilages350.4-Endplate250.25-Annulus fibrosus4.20.45-Nucleus pulposus10.499-Spinal cage36000.25-Capsular ligament7.50.3-posterior longitudinal ligament10 (< 11%) 20 (> 11%)-20Ligament flavum15 (< 6.2%) 19 (> 6.2%)-40Interspinous ligament10 (< 14%) 12 (> 14%)-40Superspinous ligament8 (< 20%) 15 (> 20%)-30Intertansverse ligament10 (< 18%) 59 (> 18%)-1.8Anterior longitudinal ligament7.8 (< 12%) 20 (> 12%)-63.7\n\nMechanical parameters of spinal models", "L5 bottom was set as a fixed support surface, and six degrees of freedom were constrained. A vertical load of 400 N and the bending moment of 10 Nm were applied to the upper surface of the L3 vertebra to simulate movements of flexion, extension, lateral bending, and axial rotation. Contact types were defined as ‘Bonded’, except for the contact type between facet joint and cartilage, which was defined as ‘Frictionless’ [27].", "Firstly, the range of motion (ROM) of all functional spinal units (FSU) were recorded, including surgical segment and adjacent segment. Secondly, the equivalent(von Mises)stress of endplate, facet joint and intervertebral fusion cage were observed.", "[SUBTITLE] Model validity and mesh convergence verification [SUBSECTION] Figure 3 indicates the result of model verification. The convergence of the model was verified by analyzing the maximum deformation and von-Mises stress in three element sizes (1 mm, 1.5 mm, 2 mm) models. The resolution difference between the predicted results of the 1.5 mm mesh size and 1 mm mesh size was < 5%, the 1.5 mm mesh was considered convergent (Fig. 4). Owing to modeling inconsistencies, some individual values were inconsistent with previous finite element results, however, all values in different conditions were within the vitro range (Yamamoto [28]).\n\nFig. 3Comparison of ROM between the current intact finite element model and previous studies:(a) L3-4, (b) L4-5; L, left;R, right\n\nComparison of ROM between the current intact finite element model and previous studies:(a) L3-4, (b) L4-5; L, left;R, right\n\nFig. 4Result of meshing sensitivity study\n\nResult of meshing sensitivity study\nFigure 3 indicates the result of model verification. The convergence of the model was verified by analyzing the maximum deformation and von-Mises stress in three element sizes (1 mm, 1.5 mm, 2 mm) models. The resolution difference between the predicted results of the 1.5 mm mesh size and 1 mm mesh size was < 5%, the 1.5 mm mesh was considered convergent (Fig. 4). Owing to modeling inconsistencies, some individual values were inconsistent with previous finite element results, however, all values in different conditions were within the vitro range (Yamamoto [28]).\n\nFig. 3Comparison of ROM between the current intact finite element model and previous studies:(a) L3-4, (b) L4-5; L, left;R, right\n\nComparison of ROM between the current intact finite element model and previous studies:(a) L3-4, (b) L4-5; L, left;R, right\n\nFig. 4Result of meshing sensitivity study\n\nResult of meshing sensitivity study\n[SUBTITLE] ROM of L4-5 and L3-4 [SUBSECTION] Figure 5 shows ROM of surgical and adjacent segment under various engineering conditions. When LLIF cage was used, a dramatic decline loss of motion at surgical level L4-5 occurred regardless the placement direction of intervertebral cage. The ROM of L4-5 was reduced by 84.21–89.03% in flexion, 72.64–82.26% in extension, 92.5-95.85% in right and left lateral bending, and 87.22–92.77% in right and left axial rotation, respectively. Meanwhile, ROM of L3-4 after LLIF was mildly increased by an average of 9.6% in all motion directions.\n\nFig. 5Changes in ROM before and after surgery: (a) L3-4, (b) L4-5; intact model (IM).\n\nChanges in ROM before and after surgery: (a) L3-4, (b) L4-5; intact model (IM).\nFigure 5 shows ROM of surgical and adjacent segment under various engineering conditions. When LLIF cage was used, a dramatic decline loss of motion at surgical level L4-5 occurred regardless the placement direction of intervertebral cage. The ROM of L4-5 was reduced by 84.21–89.03% in flexion, 72.64–82.26% in extension, 92.5-95.85% in right and left lateral bending, and 87.22–92.77% in right and left axial rotation, respectively. Meanwhile, ROM of L3-4 after LLIF was mildly increased by an average of 9.6% in all motion directions.\n\nFig. 5Changes in ROM before and after surgery: (a) L3-4, (b) L4-5; intact model (IM).\n\nChanges in ROM before and after surgery: (a) L3-4, (b) L4-5; intact model (IM).\n[SUBTITLE] Equivalent (von Mises) stress of endplate and cage [SUBSECTION] Interbody fusion cage subsidence and displacement often occur in the upper endplate postoperatively. Figure 6 shows the stress nephogram of the L5 superior endplate. The distribution of stress was consistent with the contact surface between cage and endplate. Figure 7(a-d) represents the maximum stress variation of cage and endplate. MP had the smallest stress peaks in most motion modes. The cage stress of MP was 43.95Mpa in flexion, 38.05Mpa in extension, 25.68Mpa in left bending, 43.53Mpa in right bending, 19.74Mpa in left rotation, and 22.44Mpa in right rotation.\n\nFig. 6Stress nephogram of the L5 superior endplate\n\nStress nephogram of the L5 superior endplate\n\nFig. 7Details of maximum stress:(a) cage, (b) L5 superior endplate, (c) L4 superior endplate, (d) L4 inferior endplate, (e) Intervertebral disc, (f) facet joints\n\nDetails of maximum stress:(a) cage, (b) L5 superior endplate, (c) L4 superior endplate, (d) L4 inferior endplate, (e) Intervertebral disc, (f) facet joints\nInterbody fusion cage subsidence and displacement often occur in the upper endplate postoperatively. Figure 6 shows the stress nephogram of the L5 superior endplate. The distribution of stress was consistent with the contact surface between cage and endplate. Figure 7(a-d) represents the maximum stress variation of cage and endplate. MP had the smallest stress peaks in most motion modes. The cage stress of MP was 43.95Mpa in flexion, 38.05Mpa in extension, 25.68Mpa in left bending, 43.53Mpa in right bending, 19.74Mpa in left rotation, and 22.44Mpa in right rotation.\n\nFig. 6Stress nephogram of the L5 superior endplate\n\nStress nephogram of the L5 superior endplate\n\nFig. 7Details of maximum stress:(a) cage, (b) L5 superior endplate, (c) L4 superior endplate, (d) L4 inferior endplate, (e) Intervertebral disc, (f) facet joints\n\nDetails of maximum stress:(a) cage, (b) L5 superior endplate, (c) L4 superior endplate, (d) L4 inferior endplate, (e) Intervertebral disc, (f) facet joints\n[SUBTITLE] Stress of facet joints and proximal intervertebral disc [SUBSECTION] The maximum stress of facet joints and intervertebral disc are displayed in Fig. 7(e, f). Almost all stress peaks were increased after SA LLIF, including adjacent disc, facet joints, and endplates. As shown in our result, MP had the slightest tendency to raise the stress. The facet joints stress of MP was 7.93Mpa in flexion, 9.11Mpa in extension, 10.09Mpa in left bending, 12.13Mpa in right bending, 3.87Mpa in left rotation, and 4.43Mpa in right rotation. The L3/4 intervertebral disc stress of MP was 1.72 Mpa in flexion, 1.88Mpa in extension, 2.79Mpa in left bending, 2.59Mpa in right bending, 1.31Mpa in left rotation, and 1. 42Mpa in right rotation.\nThe maximum stress of facet joints and intervertebral disc are displayed in Fig. 7(e, f). Almost all stress peaks were increased after SA LLIF, including adjacent disc, facet joints, and endplates. As shown in our result, MP had the slightest tendency to raise the stress. The facet joints stress of MP was 7.93Mpa in flexion, 9.11Mpa in extension, 10.09Mpa in left bending, 12.13Mpa in right bending, 3.87Mpa in left rotation, and 4.43Mpa in right rotation. The L3/4 intervertebral disc stress of MP was 1.72 Mpa in flexion, 1.88Mpa in extension, 2.79Mpa in left bending, 2.59Mpa in right bending, 1.31Mpa in left rotation, and 1. 42Mpa in right rotation.", "Figure 3 indicates the result of model verification. The convergence of the model was verified by analyzing the maximum deformation and von-Mises stress in three element sizes (1 mm, 1.5 mm, 2 mm) models. The resolution difference between the predicted results of the 1.5 mm mesh size and 1 mm mesh size was < 5%, the 1.5 mm mesh was considered convergent (Fig. 4). Owing to modeling inconsistencies, some individual values were inconsistent with previous finite element results, however, all values in different conditions were within the vitro range (Yamamoto [28]).\n\nFig. 3Comparison of ROM between the current intact finite element model and previous studies:(a) L3-4, (b) L4-5; L, left;R, right\n\nComparison of ROM between the current intact finite element model and previous studies:(a) L3-4, (b) L4-5; L, left;R, right\n\nFig. 4Result of meshing sensitivity study\n\nResult of meshing sensitivity study", "Figure 5 shows ROM of surgical and adjacent segment under various engineering conditions. When LLIF cage was used, a dramatic decline loss of motion at surgical level L4-5 occurred regardless the placement direction of intervertebral cage. The ROM of L4-5 was reduced by 84.21–89.03% in flexion, 72.64–82.26% in extension, 92.5-95.85% in right and left lateral bending, and 87.22–92.77% in right and left axial rotation, respectively. Meanwhile, ROM of L3-4 after LLIF was mildly increased by an average of 9.6% in all motion directions.\n\nFig. 5Changes in ROM before and after surgery: (a) L3-4, (b) L4-5; intact model (IM).\n\nChanges in ROM before and after surgery: (a) L3-4, (b) L4-5; intact model (IM).", "Interbody fusion cage subsidence and displacement often occur in the upper endplate postoperatively. Figure 6 shows the stress nephogram of the L5 superior endplate. The distribution of stress was consistent with the contact surface between cage and endplate. Figure 7(a-d) represents the maximum stress variation of cage and endplate. MP had the smallest stress peaks in most motion modes. The cage stress of MP was 43.95Mpa in flexion, 38.05Mpa in extension, 25.68Mpa in left bending, 43.53Mpa in right bending, 19.74Mpa in left rotation, and 22.44Mpa in right rotation.\n\nFig. 6Stress nephogram of the L5 superior endplate\n\nStress nephogram of the L5 superior endplate\n\nFig. 7Details of maximum stress:(a) cage, (b) L5 superior endplate, (c) L4 superior endplate, (d) L4 inferior endplate, (e) Intervertebral disc, (f) facet joints\n\nDetails of maximum stress:(a) cage, (b) L5 superior endplate, (c) L4 superior endplate, (d) L4 inferior endplate, (e) Intervertebral disc, (f) facet joints", "The maximum stress of facet joints and intervertebral disc are displayed in Fig. 7(e, f). Almost all stress peaks were increased after SA LLIF, including adjacent disc, facet joints, and endplates. As shown in our result, MP had the slightest tendency to raise the stress. The facet joints stress of MP was 7.93Mpa in flexion, 9.11Mpa in extension, 10.09Mpa in left bending, 12.13Mpa in right bending, 3.87Mpa in left rotation, and 4.43Mpa in right rotation. The L3/4 intervertebral disc stress of MP was 1.72 Mpa in flexion, 1.88Mpa in extension, 2.79Mpa in left bending, 2.59Mpa in right bending, 1.31Mpa in left rotation, and 1. 42Mpa in right rotation.", "Undeniably, SA LLIF has clear advantages in appropriate cases in terms of clinical outcome and complications. Regarding the location of the cage in lumbar fusion, investigators have also conducted clinical and mechanics studies. A previous study [29] showed a lower cage center was more liable to cause adjacent facet joint degeneration (AFD) following the transforaminal lumbar interbody fusion (TLIF). Yan Liang [30] advised the cage should be placed in traverse orientation in TLIF. Unlike TLIF, SA LLIF does not have an auxiliary internal fixation and its stability is more dependent on intervertebral fusion. The anteriorly placed cage can contribute to restoration of segmental angle, whereas the posteriorly located cage might maintain the indirect decompression effect [31]. Although there are many perspectives on the position of the cage, the optimal position is a combination of low stress and sufficient stiffness. Therefore, we are dedicated to optimizing the cage position in SA LLIF. The aim is to explore the effect of less on the adjacent segments while maintaining stability through mechanical experiments.\nIn our finite element research, intact model and four commonly used clinical placements were reconstructed. From a mechanical point of view, excessive division of angles is meaningless, and simplified models can make trends in complex variables easier to understand and intuitive. In terms of model verification, many researchers have compared model simulations with vitro experiments. However, as an indirect validation method, segments motion does not fully represent the in vivo situation. Due to complex coupled loading patterns in the model, more detailed sensitivity analysis is necessary. Both indirect validation and sensitivity analysis of the grid were applied in this experiment. Of particular note is the number of mesh. A large amount of calculations can easily lead to computational failure. In addition, attention should also be paid to the application of tetrahedral and hexahedral meshes. For simple geometry, it relies more on an 8-node hexahedral mesh. While for curved boundary models, such as the posterior part of the vertebral body, more nodes and elements are needed to obtain more accurate solutions, or a quadratic tetrahedron is used. Excessive use of hexahedra will make the meshing process too long, prone to errors or even failure. Meanwhile, the inhomogeneous meshing method was used in this study. Under the condition of convergence, mainly 2 mm meshes were used, and smaller meshes were allowed for local contact points, such as facet joints. Thus, the accuracy of the operation could be improved without affecting the overall operation time.\nThe priority to be analyzed is the effect of cage position on the stability of the surgical segment. The ROM of the FE model can directly reflect the stability of the postoperative model. Sufficient stability of the postoperative FSU is beneficial for intervertebral bone fusion. In our study, compared with the IM, the ROM of four placements was significantly reduced. The variation in the magnitude of reduction of all placements was not very obvious. The elasticity modulus of cage is much greater than that of the disc, which in turn leads to increased segmental stiffness of the FUS. In terms of movement patterns, the most influential is the posterior extension. The ROM of the L4-5 segment in AP, MP, PP, OP mode under extension was significantly reduced by 73.01%, 82.07%, 82. 22%, 72.64%, respectively. It means that SA LLIF can effectively reduce the activity of the surgical segment (> 70%) at the index level.\nOur results also indicated that the cage location was informative for cage subsidence, which was associated with segmental instability. The mechanical manifestation is the excessive stress on the contact surface. The stress distribution cloud showed that the stress was concentrated at the contact surface of the cage and the endplate. In terms of the stress peak in cage, MP had the lowest stress in the other motions, except for OP in right bending. More peak stress occurred in AP, which not indicated its stability. It is commonly believed that the smaller the internal fixation stress, the more stable it is. However, values in all tests are below the yield stress of cage, so stability should be determined comprehensively [32]. In addition, the size of deformation can also reflect the stability of segments to a certain extent. Our research proved that the minimum deformation occurred in MP under flexion and extension. The deformation in flexion, extension, left bending, right bending was 0. 4356 mm, 0. 4351 mm, 0. 0614 mm, 0. 0667 mm, respectively. The average displacement of AP was the largest, which was similar to the stress distribution of cage.\nCompared with intact model, the maximum stress on the superior and inferior endplates of the surgical segment also increased significantly, the upper endplate stress of L5 was higher than the lower endplate stress of L4. The maximum stress of MP under flexion and extension conditions was similar to that of OP, and the peak stress of MP under bending and rotation conditions was smaller. The oblique position leads to uneven force during the movement, and the stress is too concentrated, resulting in a large force on the endplate. The stiffness of the cage is much greater than that of the endplate, so the stress trends in the cage and the endplate are not the same. In the view of stress variation, L5 upper endplate stress had more influence on settlement. Overall, middle placement (MP) showed lower stress in the endplate and cage.\nThe previous study [33–36] has indicated biomechanical changes following lumbar fusion may cause degeneration in adjacent segments. The reason is mainly the increased activity and stress in the adjacent segment. Our research showed that the ROM of adjacent segment (L3-4) increased slightly for all postures following SA LLIF. The increased activity of L3-4 was compensatory because of the enhanced stiffness of L4-5. Due to the greater stiffness, its activity increased even more. As the ROM increases, the discs and facet joints are exposed to greater stress, accelerating the degeneration of the discs and facet joints. In the upright situation, 70–90% of the static load is borne by the vertebral body and 10–20% of the axial load is borne by the facet joints [37]. In the case of body flexion, the facet joint bears more shear stress. As shown in our results, under flexion and extension, the stress on the small joint increased after SA LLIF. In comparison, the stress increase in the facet joints of MP was less than in other positions. Similarly, the disc pressure in the adjacent segment also changed in the SA LLIF models. As shown in our results, the interbody disc stress of L3/4 had increased in flexion, extension and bending. MP had the smallest increment, increasing by 47% in flexion, 38%in extension, 14% in left lateral bending, 11% in right bending. Excessive loading can lead to mechanical failure of the intervertebral disc. Further, these loads may lead to the rupture of central zone in the end plate. Common structural degeneration includes reduced nucleus pulposus volume, loss of disc height, and tearing of the annulus fibrosus. Structural degeneration of the intervertebral disc is often accompanied by functional degeneration, which can cause clinical symptoms. In essence, the change of intervertebral disc force of the adjacent segment is also an indicator for judging the adjacent segment degeneration (ASD), consistent with the conclusions of Du [38]. Therefore, we believe that the MP has the least effect on the cephalad adjacent segment among all positions.\nOur research was based on finite element analysis and has some inadequacies. First, modeling of geometry varied slightly due to individual differences in volunteers and differences in accuracy during CT scanning. Second, models were simplified to analyze the trend of force changes. For example, the effects of skeletal muscle and other soft tissues were ignored, and the variation in bone density in different regions of the endplate was not considered. Full spine modeling was not performed, which could lead to some deviations in the specific values, but would not affect the overall trend. Third, the effect of the front convex angle and size of the cage on the mechanical performance were neglected. Our next research will focus on topology optimization of the cage in SA LLIF.", "In our study, SA LLIF risked accelerating the adjacent segment degeneration. The cage position had an influence on the distribution of endplate stress and the magnitude of facet joint stress. Compared with other positions, MP had the slightest effect on the stress in the adjacent facet joints. Meanwhile, MP seems to play an important role in reducing the risk of cage subsidence." ]
[ null, "materials|methods", null, null, null, null, null, "results", null, null, null, null, "discussion", "conclusion" ]
[ "Stand-alone lateral lumbar interbody fusion", "Cage position", "Finite element analysis", "Adjacent segment degeneration", "Cage subsidence" ]
Perceptions of dental health professionals (DHPs) on job satisfaction in Fiji: a qualitative study.
36258214
Reviewing job satisfaction is crucial as it has an impact on a person's physical and mental wellbeing, as well as leading to a better organizational commitment of employees that enhances the organizations succession and progress as well as better staff retention. This study aimed to explore the perceptions of job satisfaction amongst Dental Health Professionals (DHPs) in Fiji and associated factors.
BACKGROUND
This study used a phenomenological qualitative method approach commencing from August to November, 2021. The target group for this study were the DHPs who provide prosthetic services. This study was conducted among DHPs from 4 purposively selected clinics in Fiji. A semi- structured open-ended questionnaire was used to collect data. Thematic analysis was used to transcribe and analyze the audio qualitative data collected from the interviews.
METHODS
Twenty-nine DHPs took part in the in-depth interview and the responses were grouped into three themes. The findings from the study indicate that DHPs are most satisfied with their teamwork and the relationship they have with their colleagues and co-workers, followed by the nature of the work and the supervision they received. The participants indicated that they were less satisfied with professional development opportunities and least satisfied with their pay and organizational support they receive.
RESULTS
The results of this study have identified gaps and areas for improvement of job satisfaction for DHPs who provide prosthetic services in Fiji such as need for more career and professional development pathways, improved infrastructure to support prosthetic service delivery in Fiji and improve remuneration for DHPs. Understanding the factors that affect satisfaction levels and being able to act accordingly are likely to lead to positive outcomes both for DHPs and their organization.
CONCLUSION
[ "Humans", "Job Satisfaction", "Fiji", "Health Personnel", "Qualitative Research", "Attitude of Health Personnel" ]
9579612
null
null
null
null
Results
Twenty-nine DHPs participated in the online one on one interview via zoom. Females made up majority of the participants (76%, n = 22). Most of the participants were in the 30–40 age range (48%, n = 14). Participants were made up of two ethnicities, ITaukei (14%, n = 4) and Indo Fijians (86%, n = 25). Looking at educational levels majority of the staff just have a baseline graduate qualification (66%, n = 19), post graduate qualification (31%, n = 9) and just 1 staff who progressed to become a specialist (3%). MoHMS is the largest employer of the participants (72%, n = 21) from Nakasi dental clinic (28%, n = 8), Lautoka dental clinic (24%, n = 7) and Labasa dental clinic (21%, n = 6) followed by FNU (28%, n = 8). In terms of years of experience, 24%(n = 7) had more than 20 years of experience. DHPs who are involved in prosthetic services were dentists (38%, n = 11) and dental technicians (62%, n = 18) (Table 1). Table 1General information of participants (n = 29)CharacteristicsFrequencyPercentage Gender Male724Female2276 Ethnicity Indo Fijian2586ITaukei414 Age (years) 20–3062131–40144841–5051751–60414 Qualification Graduate qualification1966Post graduate931Specialist13 Work experience (years) Less than 20yrs2276More than 20yrs724 Location of practice Nakasi dental clinic828Lautoka dental clinic724Labasa dental clinic621FNU828 Dental Specialty Dentist1138Dental technician1862 General information of participants (n = 29) [SUBTITLE] Themes related to job satisfaction [SUBSECTION] Three major themes had emerged from the thematic analysis which included perception on work-related factors, barriers of job satisfaction and recommendation to promote job satisfaction. Eight sub-themes identified under three main themes including nature of work, work perspectives, work environment, limitations, Covid 19, income, professional relationships, and professional development. Under sub-themes, codes were identified as summarized in Table 2. Participants quotations were referenced as P1 to P 29, DHPs –D (dentist) or DHPs –DT (dental technician). Table 2Themes and sub-themes identifiedThemesSub-themesCodesPerception on work related factorsNature of workUnique job, feel satisfied, highly recommend, no job opportunitiesWork perspectivesVery hectic job, very tiring, multitaskingWork environmentIdeal work environment, Not fully utilize skills, Outdated facilitiesBarriers of job satisfactionLimitationsFrustration due to limitations, lack of support, lack of equipment’s and materialCovid 19Work becomes risky, not practicing full range, reduced workload, work environment becomes frustratingIncomeDissatisfied with salary, return on investment, are in debtRecommendations to promote job satisfactionProfessional relationshipsGood relationship, good teamwork, supportive supervisorsProfessional developmentProfession is bottle necked, take up non-clinical courses, lack of post graduate courses Themes and sub-themes identified Three major themes had emerged from the thematic analysis which included perception on work-related factors, barriers of job satisfaction and recommendation to promote job satisfaction. Eight sub-themes identified under three main themes including nature of work, work perspectives, work environment, limitations, Covid 19, income, professional relationships, and professional development. Under sub-themes, codes were identified as summarized in Table 2. Participants quotations were referenced as P1 to P 29, DHPs –D (dentist) or DHPs –DT (dental technician). Table 2Themes and sub-themes identifiedThemesSub-themesCodesPerception on work related factorsNature of workUnique job, feel satisfied, highly recommend, no job opportunitiesWork perspectivesVery hectic job, very tiring, multitaskingWork environmentIdeal work environment, Not fully utilize skills, Outdated facilitiesBarriers of job satisfactionLimitationsFrustration due to limitations, lack of support, lack of equipment’s and materialCovid 19Work becomes risky, not practicing full range, reduced workload, work environment becomes frustratingIncomeDissatisfied with salary, return on investment, are in debtRecommendations to promote job satisfactionProfessional relationshipsGood relationship, good teamwork, supportive supervisorsProfessional developmentProfession is bottle necked, take up non-clinical courses, lack of post graduate courses Themes and sub-themes identified [SUBTITLE] Theme 1: Perception on work-related factors [SUBSECTION] Three factors were discussed under this theme including the nature of work, work perspectives and work environment. [SUBTITLE] Nature of work [SUBSECTION] The first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession. Some considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D) “Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D) When asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT) “I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT) In contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT) “Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT) The first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession. Some considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D) “Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D) When asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT) “I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT) In contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT) “Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT) [SUBTITLE] Work perspectives [SUBSECTION] Participants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT) “Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT) Most of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D) “I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D) Participants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT) “Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT) Most of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D) “I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D) [SUBTITLE] Work environment [SUBSECTION] Here participants describe the type of work environment and conditions they are working in. Some participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT) “My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT) Some find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT) “We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT) Here participants describe the type of work environment and conditions they are working in. Some participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT) “My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT) Some find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT) “We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT) Three factors were discussed under this theme including the nature of work, work perspectives and work environment. [SUBTITLE] Nature of work [SUBSECTION] The first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession. Some considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D) “Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D) When asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT) “I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT) In contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT) “Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT) The first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession. Some considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D) “Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D) When asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT) “I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT) In contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT) “Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT) [SUBTITLE] Work perspectives [SUBSECTION] Participants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT) “Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT) Most of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D) “I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D) Participants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT) “Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT) Most of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D) “I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D) [SUBTITLE] Work environment [SUBSECTION] Here participants describe the type of work environment and conditions they are working in. Some participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT) “My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT) Some find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT) “We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT) Here participants describe the type of work environment and conditions they are working in. Some participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT) “My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT) Some find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT) “We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT) [SUBTITLE] Theme 2: barriers of job satisfaction [SUBSECTION] Different limitations, challenges faced due to Covid-19 and inadequate income were the main challenges the study participants highlighted. [SUBTITLE] Limitations [SUBSECTION] Most participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT) “Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT) A few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT) “As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT) Most participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT) “Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT) A few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT) “As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT) [SUBTITLE] Covid 19 [SUBSECTION] COVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D) “The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D) A few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D) “AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D) Some expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D) “We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D) COVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D) “The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D) A few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D) “AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D) Some expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D) “We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D) [SUBTITLE] Income [SUBSECTION] Majority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D) “Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D) Perceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT) “I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT) Majority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D) “Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D) Perceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT) “I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT) Different limitations, challenges faced due to Covid-19 and inadequate income were the main challenges the study participants highlighted. [SUBTITLE] Limitations [SUBSECTION] Most participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT) “Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT) A few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT) “As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT) Most participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT) “Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT) A few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT) “As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT) [SUBTITLE] Covid 19 [SUBSECTION] COVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D) “The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D) A few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D) “AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D) Some expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D) “We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D) COVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D) “The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D) A few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D) “AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D) Some expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D) “We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D) [SUBTITLE] Income [SUBSECTION] Majority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D) “Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D) Perceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT) “I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT) Majority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D) “Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D) Perceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT) “I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT) [SUBTITLE] Theme 3: recommendation to promote job satisfaction [SUBSECTION] Participants advised the needs for changing in professional relationship as well as professional development to increase the level of job satisfaction. [SUBTITLE] Professional relationships [SUBSECTION] Here we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement. Majority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT) “For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT) Most participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D) “Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D) Some mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D) “I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D) Here we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement. Majority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT) “For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT) Most participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D) “Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D) Some mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D) “I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D) [SUBTITLE] Professional development [SUBSECTION] This sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT) “As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT) They also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D) “I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D) Most participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D) “At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D) This sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT) “As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT) They also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D) “I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D) Most participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D) “At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D) Participants advised the needs for changing in professional relationship as well as professional development to increase the level of job satisfaction. [SUBTITLE] Professional relationships [SUBSECTION] Here we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement. Majority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT) “For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT) Most participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D) “Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D) Some mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D) “I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D) Here we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement. Majority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT) “For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT) Most participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D) “Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D) Some mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D) “I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D) [SUBTITLE] Professional development [SUBSECTION] This sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT) “As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT) They also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D) “I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D) Most participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D) “At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D) This sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT) “As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT) They also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D) “I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D) Most participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D) “At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)
Conclusion
To date, this has been the only study conducted exploring the perceptions of job satisfaction amongst DHPs in Fiji using qualitative analysis. The results of this study can be used to inform DHPs, their employers and academic institutes about what factors affect job satisfaction of individuals working within this profession. Factors such as nature of work, work perspectives of participants, work environment, limitations, Covid 19, income, professional relationships, and professional development were found to contribute to job satisfaction. The empirical findings from the study indicate that DHPs employed by MoHMS and FNU, are most satisfied with their teamwork and the relationship they have with their colleagues and co-workers, followed by the nature of the work and the supervision they received. They however, indicated that they are less satisfied with professional development opportunities and least satisfied with the pay they receive and organizational support they receive. The entire structure of education and career progression in dentistry and dental technology should be addressed as a matter of urgency to achieve SDG 3, in order to encourage individuals both to join and remain within the profession.
[ "Background", "Conceptual framework", "Methods", "Study design and setting", "Sampling", "Data collection tool", "Data collection", "Trustworthiness and reflexivity", "Research team", "Data management and analysis", "Themes related to job satisfaction", "Theme 1: Perception on work-related factors", "Nature of work", "Work perspectives", "Work environment", "Theme 2: barriers of job satisfaction", "Limitations", "Covid 19", "Income", "Theme 3: recommendation to promote job satisfaction", "Professional relationships", "Professional development", "Limitations", "" ]
[ "In any institution, the most valuable asset is human resources making job satisfaction as one of the most important factors for efficiency and productivity of a sustainable service delivery [1]. Job satisfaction can be defined as having a “positive emotional state resulting from the appraisal of one’s job or job experience and is related to many aspects of patient care and outcomes of health system as well as to contentment in general life and job related performance” [2]. Reviewing job satisfaction is crucial as it has an impact on a person’s physical and mental wellbeing. It also leads to improved organizational commitment of employees that enhances the organizations success and progress as well as increased staff retention. Job satisfaction is the essence of any successful dental practice [3].\nDentistry can be a very stressful profession thus to encourage dental professionals to provide high quality service, assessing job satisfaction becomes important for understanding how work environmental factors impact it [4]. According to Gilmour et al. [5] dentistry has frequently been described as a stressful occupation, and is associated with greater incidence of ill health, alcoholism, and suicide than other medical professions. Different levels of job satisfaction have been observed in different counties such a more than 80% of Australian and Lithuanians dental professional reported being highly satisfied, while the Koreans and Egyptians had and an overall average job satisfaction of 51% and the Iranians being the least satisfied having a mean score of 1.5 out of 5 indicating they may have a very stressful work environment [6–10]. Nikolovska et al. [11] found that dental professionals in Macedonia showed overall low levels of job satisfaction among the public and private dental practitioners due to high stress levels and that private practitioners were the most satisfied compared to public practitioners. A similar result was seen in study in the United Arab Emirates (UAE) where private sector dental professionals had a higher level of satisfaction compared with the public sector [12]. Job satisfaction may be generalized as a person’s attitude towards their profession. People who experience high levels of satisfaction are likely to have a more positive outlook towards their profession in comparison to those who experience low levels of satisfaction [13].\nTuisuva et al. [14] characterized dentistry in Fiji and the region as ‘Pacific setup” which is a community-based approach where daily functions are limited by malfunctioning equipment’s, lack of quality resources and inadequate remuneration for staff, poor working environments which are indicators for low job satisfaction level. Although there is extensive literature on job satisfaction, there is little to none research on job satisfaction amongst dental professionals in Fiji, in particular dentists and dental technicians. Therefore, this study aimed to explore the perceptions and factors affecting job satisfaction among Dental Health Professionals (DHPs) in Fiji. In any institution, the most valuable asset is human resources making job satisfaction as one of the most important factors for efficiency and productivity of a sustainable service delivery. To promote the quality of life among DHPs it is important to consider factors affecting their job satisfaction where they are working and providing service to patients. This study is trying to address the Sustainable Development Goal (SDG) 3 to “ensure healthy lives and promote well-being for all at all ages”. The findings of this study will be helpful to policymakers to design plans in order to increase the level of job satisfaction of DHPs by highlighting areas where there is a need for improvement in services. This will also be helpful in identifying the need for program development for dental academic institutes.\n[SUBTITLE] Conceptual framework [SUBSECTION] A coherent conceptual framework is constructed to elaborate the theoretical relationship between job satisfaction factors and employee commitment to organizational culture as shown in Fig. 1. This conceptual framework summarizes the variables that affect job satisfaction among dental professionals. This relationship will be tested and verified based on this research.\nFactors categorized as personal and organizational determinants are associated with job satisfaction and can influence a person’s level of satisfaction and organizational commitment [1].\nPersonal determinants include age, gender, and level of education. Gender based differences is a determinant of job satisfaction, female practitioners ascribe more importance to social factors, while males place greater value on income, progression and other extrinsic aspects [15]. Older practitioners are generally happier with their jobs than younger employees, while people who are more experienced in their jobs are more highly satisfied than those who are less experienced.\nOrganizational factors include nature of work, income, interactions and career development which includes autonomy, working conditions, recognition of work, professional development, availability of resources, and organization structure. Autonomy given to employees allow them to feel confident and competent thus allowing then to make a positive impact on the organization. If employees feel that the work done by them is meaningful, has significance and is recognized, they will be satisfied. Any person would want a pay system that they perceive as just, unequivocal, and in line with their expectations. Satisfaction is likely to result when one’s income is seen as fair based on job demands, individual skill level, and in accordance with community pay standards. In any organization, employees seek fair promotion policies and practices that provide opportunities for personal growth, additional responsibilities, and improved social status. Individuals who recognize that promotional decisions are made in a fair and just manner, experience satisfaction with their jobs. Work environments that support personal comfort, growth and good job performance are highly desirable. Most employees also prefer working closer to home, in a clean and modern facility with adequate staff and resources may result in satisfaction or dissatisfaction if work environment is not suitable. Practices with more-satisfied employees tend to be more productive than those with less-satisfied employees [16].\nProfessional relationships, location of practice and quality of interaction indicating a need for healthy work interaction and professional networking which is a major cause of satisfaction. Studies have found that employees feel satisfaction when superior staff understands them, is friendly and approachable, compliments good performance, listens to employees’ opinions, and shows a personal interest in them. On the contrary, practices with toxic work culture where staff feel undermined, disrespected, undervalued and threated, are generally dissatisfied [17].\nFinally, professional factors include job identity, work status, job security and qualification. Employees with renewed contracts/ permanent placement have a sense of job security, feel more satisfied. This gives them job identity and status. Career development and qualification upgrade incentives provide opportunities for staff enhance their skills and knowledge which in turn benefits both the organization and staff and are satisfying aspects of workplaces [18].\nJob satisfaction can be an important indicator of how DHPs feel about their jobs add value to their practice such as retentions of skilled staff, increase productivity, reduce turnover, enhanced patient satisfaction and loyalty, improve teamwork, high quality service delivery and care. On the other hand, dissatisfaction can cause employees to have frequent absenteeism, high turnover, thoughts of changing profession, low motivation and moral, high stress levels and poor performance. All these factors are interrelated and has an implication on the other which determines satisfaction or dissatisfaction.\n\nFig. 1Conceptual framework\n\nConceptual framework\nA coherent conceptual framework is constructed to elaborate the theoretical relationship between job satisfaction factors and employee commitment to organizational culture as shown in Fig. 1. This conceptual framework summarizes the variables that affect job satisfaction among dental professionals. This relationship will be tested and verified based on this research.\nFactors categorized as personal and organizational determinants are associated with job satisfaction and can influence a person’s level of satisfaction and organizational commitment [1].\nPersonal determinants include age, gender, and level of education. Gender based differences is a determinant of job satisfaction, female practitioners ascribe more importance to social factors, while males place greater value on income, progression and other extrinsic aspects [15]. Older practitioners are generally happier with their jobs than younger employees, while people who are more experienced in their jobs are more highly satisfied than those who are less experienced.\nOrganizational factors include nature of work, income, interactions and career development which includes autonomy, working conditions, recognition of work, professional development, availability of resources, and organization structure. Autonomy given to employees allow them to feel confident and competent thus allowing then to make a positive impact on the organization. If employees feel that the work done by them is meaningful, has significance and is recognized, they will be satisfied. Any person would want a pay system that they perceive as just, unequivocal, and in line with their expectations. Satisfaction is likely to result when one’s income is seen as fair based on job demands, individual skill level, and in accordance with community pay standards. In any organization, employees seek fair promotion policies and practices that provide opportunities for personal growth, additional responsibilities, and improved social status. Individuals who recognize that promotional decisions are made in a fair and just manner, experience satisfaction with their jobs. Work environments that support personal comfort, growth and good job performance are highly desirable. Most employees also prefer working closer to home, in a clean and modern facility with adequate staff and resources may result in satisfaction or dissatisfaction if work environment is not suitable. Practices with more-satisfied employees tend to be more productive than those with less-satisfied employees [16].\nProfessional relationships, location of practice and quality of interaction indicating a need for healthy work interaction and professional networking which is a major cause of satisfaction. Studies have found that employees feel satisfaction when superior staff understands them, is friendly and approachable, compliments good performance, listens to employees’ opinions, and shows a personal interest in them. On the contrary, practices with toxic work culture where staff feel undermined, disrespected, undervalued and threated, are generally dissatisfied [17].\nFinally, professional factors include job identity, work status, job security and qualification. Employees with renewed contracts/ permanent placement have a sense of job security, feel more satisfied. This gives them job identity and status. Career development and qualification upgrade incentives provide opportunities for staff enhance their skills and knowledge which in turn benefits both the organization and staff and are satisfying aspects of workplaces [18].\nJob satisfaction can be an important indicator of how DHPs feel about their jobs add value to their practice such as retentions of skilled staff, increase productivity, reduce turnover, enhanced patient satisfaction and loyalty, improve teamwork, high quality service delivery and care. On the other hand, dissatisfaction can cause employees to have frequent absenteeism, high turnover, thoughts of changing profession, low motivation and moral, high stress levels and poor performance. All these factors are interrelated and has an implication on the other which determines satisfaction or dissatisfaction.\n\nFig. 1Conceptual framework\n\nConceptual framework", "A coherent conceptual framework is constructed to elaborate the theoretical relationship between job satisfaction factors and employee commitment to organizational culture as shown in Fig. 1. This conceptual framework summarizes the variables that affect job satisfaction among dental professionals. This relationship will be tested and verified based on this research.\nFactors categorized as personal and organizational determinants are associated with job satisfaction and can influence a person’s level of satisfaction and organizational commitment [1].\nPersonal determinants include age, gender, and level of education. Gender based differences is a determinant of job satisfaction, female practitioners ascribe more importance to social factors, while males place greater value on income, progression and other extrinsic aspects [15]. Older practitioners are generally happier with their jobs than younger employees, while people who are more experienced in their jobs are more highly satisfied than those who are less experienced.\nOrganizational factors include nature of work, income, interactions and career development which includes autonomy, working conditions, recognition of work, professional development, availability of resources, and organization structure. Autonomy given to employees allow them to feel confident and competent thus allowing then to make a positive impact on the organization. If employees feel that the work done by them is meaningful, has significance and is recognized, they will be satisfied. Any person would want a pay system that they perceive as just, unequivocal, and in line with their expectations. Satisfaction is likely to result when one’s income is seen as fair based on job demands, individual skill level, and in accordance with community pay standards. In any organization, employees seek fair promotion policies and practices that provide opportunities for personal growth, additional responsibilities, and improved social status. Individuals who recognize that promotional decisions are made in a fair and just manner, experience satisfaction with their jobs. Work environments that support personal comfort, growth and good job performance are highly desirable. Most employees also prefer working closer to home, in a clean and modern facility with adequate staff and resources may result in satisfaction or dissatisfaction if work environment is not suitable. Practices with more-satisfied employees tend to be more productive than those with less-satisfied employees [16].\nProfessional relationships, location of practice and quality of interaction indicating a need for healthy work interaction and professional networking which is a major cause of satisfaction. Studies have found that employees feel satisfaction when superior staff understands them, is friendly and approachable, compliments good performance, listens to employees’ opinions, and shows a personal interest in them. On the contrary, practices with toxic work culture where staff feel undermined, disrespected, undervalued and threated, are generally dissatisfied [17].\nFinally, professional factors include job identity, work status, job security and qualification. Employees with renewed contracts/ permanent placement have a sense of job security, feel more satisfied. This gives them job identity and status. Career development and qualification upgrade incentives provide opportunities for staff enhance their skills and knowledge which in turn benefits both the organization and staff and are satisfying aspects of workplaces [18].\nJob satisfaction can be an important indicator of how DHPs feel about their jobs add value to their practice such as retentions of skilled staff, increase productivity, reduce turnover, enhanced patient satisfaction and loyalty, improve teamwork, high quality service delivery and care. On the other hand, dissatisfaction can cause employees to have frequent absenteeism, high turnover, thoughts of changing profession, low motivation and moral, high stress levels and poor performance. All these factors are interrelated and has an implication on the other which determines satisfaction or dissatisfaction.\n\nFig. 1Conceptual framework\n\nConceptual framework", "[SUBTITLE] Study design and setting [SUBSECTION] This study applied a descriptive qualitative study design among DHPs of Fiji which allowed for much richer and detailed responses from subjects being studied [19]. This study was conducted among the DHPs based at the dental clinics of the Ministry of Health and Medical Services (MoHMS) that provide prosthetic services located in Fiji namely Nakasi Dental Clinic, Lautoka Hospital, Labasa Hospital, and at the FNU Dental Clinic in Suva. These clinics had been purposively selected as they had an attached established laboratory support with registered staff under the Fiji Dental Council (FDC). In this study, the Consolidated Criteria for Reporting Qualitative Studies (COREQ) guidelines were followed to ensure rigor (Appendix 1).\nThis study applied a descriptive qualitative study design among DHPs of Fiji which allowed for much richer and detailed responses from subjects being studied [19]. This study was conducted among the DHPs based at the dental clinics of the Ministry of Health and Medical Services (MoHMS) that provide prosthetic services located in Fiji namely Nakasi Dental Clinic, Lautoka Hospital, Labasa Hospital, and at the FNU Dental Clinic in Suva. These clinics had been purposively selected as they had an attached established laboratory support with registered staff under the Fiji Dental Council (FDC). In this study, the Consolidated Criteria for Reporting Qualitative Studies (COREQ) guidelines were followed to ensure rigor (Appendix 1).\n[SUBTITLE] Sampling [SUBSECTION] A non-probability sampling design [20] in the form of a homogeneous purposive sampling method was used to gather data as target population information. This study included all the DHPs in Fiji. Those DHPs who met the study criteria were included in this study as study sample. They were Dentists and dental technicians registered under the FDC, Minimum work experience of 6 months, working at the selected clinics in the time of data collection, must be attached to the dental prosthetic clinic/ lab, and DHPs of both genders or any ethnicity. Those who worked in private dental clinics or were not willing to participate in this study were excluded.\nA total of 29 DHPs were approached for the in-depth interviews. The sample participants included 18 dental technicians and 11 dentists. The final sample size was based on the data saturation.\nA non-probability sampling design [20] in the form of a homogeneous purposive sampling method was used to gather data as target population information. This study included all the DHPs in Fiji. Those DHPs who met the study criteria were included in this study as study sample. They were Dentists and dental technicians registered under the FDC, Minimum work experience of 6 months, working at the selected clinics in the time of data collection, must be attached to the dental prosthetic clinic/ lab, and DHPs of both genders or any ethnicity. Those who worked in private dental clinics or were not willing to participate in this study were excluded.\nA total of 29 DHPs were approached for the in-depth interviews. The sample participants included 18 dental technicians and 11 dentists. The final sample size was based on the data saturation.\n[SUBTITLE] Data collection tool [SUBSECTION] The data had been collected through a semi-structured open-ended questionnaire in the form of an interview via zoom with participants. The questions were developed based on the literatures and study research questions which comprised of two sections including (Appendix 2):\n1) Demographic variables containing five questions including: age, race, gender, qualification, years of work experience and dental specialty.\n2) Seven open ended questions related to job satisfaction in the English language as all participants were literate in English which eliminated the need for translation in other languages.\nThe data had been collected through a semi-structured open-ended questionnaire in the form of an interview via zoom with participants. The questions were developed based on the literatures and study research questions which comprised of two sections including (Appendix 2):\n1) Demographic variables containing five questions including: age, race, gender, qualification, years of work experience and dental specialty.\n2) Seven open ended questions related to job satisfaction in the English language as all participants were literate in English which eliminated the need for translation in other languages.\n[SUBTITLE] Data collection [SUBSECTION] Due to the global pandemic crisis of COVID 19, the study procedure had to be amended to accommodate the movement restrictions put in place by the Fijian government. Thus, the use of a virtual platform had been employed to aid in data collection. Ethical approval was obtained from the College Health Research Ethics Committee (CHREC) with the ID#032.21 before commencement of the study. A list of suitable participants was made by contacting the dental clinics and those that met the inclusion/ exclusion criteria were contacted via phone/email requesting their participation and date and time of interview was confirmed. An information sheet together with a consent form was emailed to them. On the day of interview, a guideline was explained, and a verbal consent was recorded before commencing the interview question on Zoom. It was made clear to the participants that their identity would not be disclosed, confidentiality will be maintained and were requested to give their frank opinions. Each interview took about 20–30 min. During the interview, the conversation was recorded and note taking was done by the principal researcher. After each interview, the resulting audio was transcribed, and notes from the interviews were reviewed to identify all references and content that related to job satisfaction. Transcripts were discussed with participants for comment and/or correction.\nDue to the global pandemic crisis of COVID 19, the study procedure had to be amended to accommodate the movement restrictions put in place by the Fijian government. Thus, the use of a virtual platform had been employed to aid in data collection. Ethical approval was obtained from the College Health Research Ethics Committee (CHREC) with the ID#032.21 before commencement of the study. A list of suitable participants was made by contacting the dental clinics and those that met the inclusion/ exclusion criteria were contacted via phone/email requesting their participation and date and time of interview was confirmed. An information sheet together with a consent form was emailed to them. On the day of interview, a guideline was explained, and a verbal consent was recorded before commencing the interview question on Zoom. It was made clear to the participants that their identity would not be disclosed, confidentiality will be maintained and were requested to give their frank opinions. Each interview took about 20–30 min. During the interview, the conversation was recorded and note taking was done by the principal researcher. After each interview, the resulting audio was transcribed, and notes from the interviews were reviewed to identify all references and content that related to job satisfaction. Transcripts were discussed with participants for comment and/or correction.\n[SUBTITLE] Trustworthiness and reflexivity [SUBSECTION] Trustworthiness is considered a more appropriate criterion for evaluating qualitative studies. In order to ensure the process is trustworthy, four criterions had been adopted. They are credibility, transferability, dependability, and confirmability [21]. There were many strategies to address credibility that include “prolonged engagement” and member checks. In-depth interviews were conducted over the period of 1 month and each interview lasted for at least 20–30 min. All in depth interviews were recorded and transcribed by the researcher on the same day. Every step of the thesis and results was discussed with the principal supervisor.\nTransferability related to the ability of the findings to be transferred to other contexts or settings. Because qualitative research is specific to a particular context, it is important a “thick description” of the particular research context is provided to allow the reader to assess whether it is transferable to their situation or not. Purposive sampling technique was used in this research and in-depth interviews were carried out until data saturation was achieved. Dependability ensured that the process is described in sufficient detail to facilitate another researcher to repeat the work. This study was developed from the early stages through a systematic search of the existing literature. Same in-depth interview questions were asked to all participants of the study and review of transcriptions were done to correct errors. All data was coded and the coding was checked and rechecked thorough. Confirmability was comparable to objectivity in quantitative studies. Here, the goal was to minimize investigator bias by acknowledging researcher predispositions. Adherence to this framework by adopting strategies, such as those outlined, to address the individual criteria supports a rigorous research process. Data was checked by the principal supervisor-MM. During the interview process, note taking of the participants comments was done by principle researcher SK.\nTrustworthiness is considered a more appropriate criterion for evaluating qualitative studies. In order to ensure the process is trustworthy, four criterions had been adopted. They are credibility, transferability, dependability, and confirmability [21]. There were many strategies to address credibility that include “prolonged engagement” and member checks. In-depth interviews were conducted over the period of 1 month and each interview lasted for at least 20–30 min. All in depth interviews were recorded and transcribed by the researcher on the same day. Every step of the thesis and results was discussed with the principal supervisor.\nTransferability related to the ability of the findings to be transferred to other contexts or settings. Because qualitative research is specific to a particular context, it is important a “thick description” of the particular research context is provided to allow the reader to assess whether it is transferable to their situation or not. Purposive sampling technique was used in this research and in-depth interviews were carried out until data saturation was achieved. Dependability ensured that the process is described in sufficient detail to facilitate another researcher to repeat the work. This study was developed from the early stages through a systematic search of the existing literature. Same in-depth interview questions were asked to all participants of the study and review of transcriptions were done to correct errors. All data was coded and the coding was checked and rechecked thorough. Confirmability was comparable to objectivity in quantitative studies. Here, the goal was to minimize investigator bias by acknowledging researcher predispositions. Adherence to this framework by adopting strategies, such as those outlined, to address the individual criteria supports a rigorous research process. Data was checked by the principal supervisor-MM. During the interview process, note taking of the participants comments was done by principle researcher SK.\n[SUBTITLE] Research team [SUBSECTION] The research team consisted of the principle female researcher who was a master student in health services management and attended a 2-hour training for conducting interviews (SK) while being supervised and guided by principal supervisor (MM).\nThe research team consisted of the principle female researcher who was a master student in health services management and attended a 2-hour training for conducting interviews (SK) while being supervised and guided by principal supervisor (MM).\n[SUBTITLE] Data management and analysis [SUBSECTION] The content of interview was transcribed by the main researcher. The transcribed data was then checked for sufficient quality and accuracy by main researcher before major analysis is conducted. Backup files had been created considering data management system and were updated as data preparation and analysis proceeded. The collected data were analyzed using manual thematic analysis [22]. Field notes were arranged according to codes and themes which were combined into major categories and files (word documents) will be created and labeled. The principal researcher SK read and re-read all interview transcripts and identified similar phrases and words for which numbers were assigned. The coded data that had similar characteristics was grouped together. Once grouping of similar data was completed, descriptive themes and sub themes were identified to reflect the perceptions of participants. The themes and sub-themes were checked by the principal supervisor as well MM.\nThe content of interview was transcribed by the main researcher. The transcribed data was then checked for sufficient quality and accuracy by main researcher before major analysis is conducted. Backup files had been created considering data management system and were updated as data preparation and analysis proceeded. The collected data were analyzed using manual thematic analysis [22]. Field notes were arranged according to codes and themes which were combined into major categories and files (word documents) will be created and labeled. The principal researcher SK read and re-read all interview transcripts and identified similar phrases and words for which numbers were assigned. The coded data that had similar characteristics was grouped together. Once grouping of similar data was completed, descriptive themes and sub themes were identified to reflect the perceptions of participants. The themes and sub-themes were checked by the principal supervisor as well MM.", "This study applied a descriptive qualitative study design among DHPs of Fiji which allowed for much richer and detailed responses from subjects being studied [19]. This study was conducted among the DHPs based at the dental clinics of the Ministry of Health and Medical Services (MoHMS) that provide prosthetic services located in Fiji namely Nakasi Dental Clinic, Lautoka Hospital, Labasa Hospital, and at the FNU Dental Clinic in Suva. These clinics had been purposively selected as they had an attached established laboratory support with registered staff under the Fiji Dental Council (FDC). In this study, the Consolidated Criteria for Reporting Qualitative Studies (COREQ) guidelines were followed to ensure rigor (Appendix 1).", "A non-probability sampling design [20] in the form of a homogeneous purposive sampling method was used to gather data as target population information. This study included all the DHPs in Fiji. Those DHPs who met the study criteria were included in this study as study sample. They were Dentists and dental technicians registered under the FDC, Minimum work experience of 6 months, working at the selected clinics in the time of data collection, must be attached to the dental prosthetic clinic/ lab, and DHPs of both genders or any ethnicity. Those who worked in private dental clinics or were not willing to participate in this study were excluded.\nA total of 29 DHPs were approached for the in-depth interviews. The sample participants included 18 dental technicians and 11 dentists. The final sample size was based on the data saturation.", "The data had been collected through a semi-structured open-ended questionnaire in the form of an interview via zoom with participants. The questions were developed based on the literatures and study research questions which comprised of two sections including (Appendix 2):\n1) Demographic variables containing five questions including: age, race, gender, qualification, years of work experience and dental specialty.\n2) Seven open ended questions related to job satisfaction in the English language as all participants were literate in English which eliminated the need for translation in other languages.", "Due to the global pandemic crisis of COVID 19, the study procedure had to be amended to accommodate the movement restrictions put in place by the Fijian government. Thus, the use of a virtual platform had been employed to aid in data collection. Ethical approval was obtained from the College Health Research Ethics Committee (CHREC) with the ID#032.21 before commencement of the study. A list of suitable participants was made by contacting the dental clinics and those that met the inclusion/ exclusion criteria were contacted via phone/email requesting their participation and date and time of interview was confirmed. An information sheet together with a consent form was emailed to them. On the day of interview, a guideline was explained, and a verbal consent was recorded before commencing the interview question on Zoom. It was made clear to the participants that their identity would not be disclosed, confidentiality will be maintained and were requested to give their frank opinions. Each interview took about 20–30 min. During the interview, the conversation was recorded and note taking was done by the principal researcher. After each interview, the resulting audio was transcribed, and notes from the interviews were reviewed to identify all references and content that related to job satisfaction. Transcripts were discussed with participants for comment and/or correction.", "Trustworthiness is considered a more appropriate criterion for evaluating qualitative studies. In order to ensure the process is trustworthy, four criterions had been adopted. They are credibility, transferability, dependability, and confirmability [21]. There were many strategies to address credibility that include “prolonged engagement” and member checks. In-depth interviews were conducted over the period of 1 month and each interview lasted for at least 20–30 min. All in depth interviews were recorded and transcribed by the researcher on the same day. Every step of the thesis and results was discussed with the principal supervisor.\nTransferability related to the ability of the findings to be transferred to other contexts or settings. Because qualitative research is specific to a particular context, it is important a “thick description” of the particular research context is provided to allow the reader to assess whether it is transferable to their situation or not. Purposive sampling technique was used in this research and in-depth interviews were carried out until data saturation was achieved. Dependability ensured that the process is described in sufficient detail to facilitate another researcher to repeat the work. This study was developed from the early stages through a systematic search of the existing literature. Same in-depth interview questions were asked to all participants of the study and review of transcriptions were done to correct errors. All data was coded and the coding was checked and rechecked thorough. Confirmability was comparable to objectivity in quantitative studies. Here, the goal was to minimize investigator bias by acknowledging researcher predispositions. Adherence to this framework by adopting strategies, such as those outlined, to address the individual criteria supports a rigorous research process. Data was checked by the principal supervisor-MM. During the interview process, note taking of the participants comments was done by principle researcher SK.", "The research team consisted of the principle female researcher who was a master student in health services management and attended a 2-hour training for conducting interviews (SK) while being supervised and guided by principal supervisor (MM).", "The content of interview was transcribed by the main researcher. The transcribed data was then checked for sufficient quality and accuracy by main researcher before major analysis is conducted. Backup files had been created considering data management system and were updated as data preparation and analysis proceeded. The collected data were analyzed using manual thematic analysis [22]. Field notes were arranged according to codes and themes which were combined into major categories and files (word documents) will be created and labeled. The principal researcher SK read and re-read all interview transcripts and identified similar phrases and words for which numbers were assigned. The coded data that had similar characteristics was grouped together. Once grouping of similar data was completed, descriptive themes and sub themes were identified to reflect the perceptions of participants. The themes and sub-themes were checked by the principal supervisor as well MM.", "Three major themes had emerged from the thematic analysis which included perception on work-related factors, barriers of job satisfaction and recommendation to promote job satisfaction. Eight sub-themes identified under three main themes including nature of work, work perspectives, work environment, limitations, Covid 19, income, professional relationships, and professional development.\nUnder sub-themes, codes were identified as summarized in Table 2. Participants quotations were referenced as P1 to P 29, DHPs –D (dentist) or DHPs –DT (dental technician).\n\nTable 2Themes and sub-themes identifiedThemesSub-themesCodesPerception on work related factorsNature of workUnique job, feel satisfied, highly recommend, no job opportunitiesWork perspectivesVery hectic job, very tiring, multitaskingWork environmentIdeal work environment, Not fully utilize skills, Outdated facilitiesBarriers of job satisfactionLimitationsFrustration due to limitations, lack of support, lack of equipment’s and materialCovid 19Work becomes risky, not practicing full range, reduced workload, work environment becomes frustratingIncomeDissatisfied with salary, return on investment, are in debtRecommendations to promote job satisfactionProfessional relationshipsGood relationship, good teamwork, supportive supervisorsProfessional developmentProfession is bottle necked, take up non-clinical courses, lack of post graduate courses\n\nThemes and sub-themes identified", "Three factors were discussed under this theme including the nature of work, work perspectives and work environment.\n[SUBTITLE] Nature of work [SUBSECTION] The first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession.\nSome considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\n“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\nWhen asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\n“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\nIn contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\nThe first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession.\nSome considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\n“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\nWhen asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\n“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\nIn contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n[SUBTITLE] Work perspectives [SUBSECTION] Participants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\n“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\nMost of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\nParticipants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\n“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\nMost of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n[SUBTITLE] Work environment [SUBSECTION] Here participants describe the type of work environment and conditions they are working in.\nSome participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\n“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\nSome find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\n“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\nHere participants describe the type of work environment and conditions they are working in.\nSome participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\n“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\nSome find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\n“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)", "The first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession.\nSome considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\n“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\nWhen asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\n“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\nIn contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)", "Participants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\n“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\nMost of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)", "Here participants describe the type of work environment and conditions they are working in.\nSome participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\n“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\nSome find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\n“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)", "Different limitations, challenges faced due to Covid-19 and inadequate income were the main challenges the study participants highlighted.\n[SUBTITLE] Limitations [SUBSECTION] Most participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\n“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\nA few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\nMost participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\n“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\nA few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n[SUBTITLE] Covid 19 [SUBSECTION] COVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\n“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\nA few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\n“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\nSome expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\nCOVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\n“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\nA few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\n“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\nSome expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n[SUBTITLE] Income [SUBSECTION] Majority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\n“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\nPerceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\n“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\nMajority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\n“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\nPerceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\n“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)", "Most participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\n“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\nA few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)", "COVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\n“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\nA few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\n“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\nSome expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)", "Majority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\n“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\nPerceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\n“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)", "Participants advised the needs for changing in professional relationship as well as professional development to increase the level of job satisfaction.\n[SUBTITLE] Professional relationships [SUBSECTION] Here we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement.\nMajority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\n“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\nMost participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\n“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\nSome mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\nHere we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement.\nMajority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\n“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\nMost participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\n“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\nSome mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n[SUBTITLE] Professional development [SUBSECTION] This sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\n“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\nThey also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\n“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\nMost participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\n“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\nThis sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\n“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\nThey also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\n“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\nMost participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\n“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)", "Here we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement.\nMajority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\n“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\nMost participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\n“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\nSome mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)", "This sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\n“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\nThey also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\n“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\nMost participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\n“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)", "Findings of this research must be interpreted within the context of its limitations due to qualitative design; this study is limited to DHPs providing prosthetic services in Fiji, study findings included only DHPs working at MoHMS and FNU. It would be ideal for future researchers to include DHPs who offer prosthetic services in the private sector. Due to the setting of the study and the nature of the study participants, timing of data collection was limited. Data was collected via a virtual platform called Zoom based at a time convenient for the participants.", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2" ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Conceptual framework", "Methods", "Study design and setting", "Sampling", "Data collection tool", "Data collection", "Trustworthiness and reflexivity", "Research team", "Data management and analysis", "Results", "Themes related to job satisfaction", "Theme 1: Perception on work-related factors", "Nature of work", "Work perspectives", "Work environment", "Theme 2: barriers of job satisfaction", "Limitations", "Covid 19", "Income", "Theme 3: recommendation to promote job satisfaction", "Professional relationships", "Professional development", "Discussion", "Limitations", "Conclusion", "Electronic supplementary material", "" ]
[ "In any institution, the most valuable asset is human resources making job satisfaction as one of the most important factors for efficiency and productivity of a sustainable service delivery [1]. Job satisfaction can be defined as having a “positive emotional state resulting from the appraisal of one’s job or job experience and is related to many aspects of patient care and outcomes of health system as well as to contentment in general life and job related performance” [2]. Reviewing job satisfaction is crucial as it has an impact on a person’s physical and mental wellbeing. It also leads to improved organizational commitment of employees that enhances the organizations success and progress as well as increased staff retention. Job satisfaction is the essence of any successful dental practice [3].\nDentistry can be a very stressful profession thus to encourage dental professionals to provide high quality service, assessing job satisfaction becomes important for understanding how work environmental factors impact it [4]. According to Gilmour et al. [5] dentistry has frequently been described as a stressful occupation, and is associated with greater incidence of ill health, alcoholism, and suicide than other medical professions. Different levels of job satisfaction have been observed in different counties such a more than 80% of Australian and Lithuanians dental professional reported being highly satisfied, while the Koreans and Egyptians had and an overall average job satisfaction of 51% and the Iranians being the least satisfied having a mean score of 1.5 out of 5 indicating they may have a very stressful work environment [6–10]. Nikolovska et al. [11] found that dental professionals in Macedonia showed overall low levels of job satisfaction among the public and private dental practitioners due to high stress levels and that private practitioners were the most satisfied compared to public practitioners. A similar result was seen in study in the United Arab Emirates (UAE) where private sector dental professionals had a higher level of satisfaction compared with the public sector [12]. Job satisfaction may be generalized as a person’s attitude towards their profession. People who experience high levels of satisfaction are likely to have a more positive outlook towards their profession in comparison to those who experience low levels of satisfaction [13].\nTuisuva et al. [14] characterized dentistry in Fiji and the region as ‘Pacific setup” which is a community-based approach where daily functions are limited by malfunctioning equipment’s, lack of quality resources and inadequate remuneration for staff, poor working environments which are indicators for low job satisfaction level. Although there is extensive literature on job satisfaction, there is little to none research on job satisfaction amongst dental professionals in Fiji, in particular dentists and dental technicians. Therefore, this study aimed to explore the perceptions and factors affecting job satisfaction among Dental Health Professionals (DHPs) in Fiji. In any institution, the most valuable asset is human resources making job satisfaction as one of the most important factors for efficiency and productivity of a sustainable service delivery. To promote the quality of life among DHPs it is important to consider factors affecting their job satisfaction where they are working and providing service to patients. This study is trying to address the Sustainable Development Goal (SDG) 3 to “ensure healthy lives and promote well-being for all at all ages”. The findings of this study will be helpful to policymakers to design plans in order to increase the level of job satisfaction of DHPs by highlighting areas where there is a need for improvement in services. This will also be helpful in identifying the need for program development for dental academic institutes.\n[SUBTITLE] Conceptual framework [SUBSECTION] A coherent conceptual framework is constructed to elaborate the theoretical relationship between job satisfaction factors and employee commitment to organizational culture as shown in Fig. 1. This conceptual framework summarizes the variables that affect job satisfaction among dental professionals. This relationship will be tested and verified based on this research.\nFactors categorized as personal and organizational determinants are associated with job satisfaction and can influence a person’s level of satisfaction and organizational commitment [1].\nPersonal determinants include age, gender, and level of education. Gender based differences is a determinant of job satisfaction, female practitioners ascribe more importance to social factors, while males place greater value on income, progression and other extrinsic aspects [15]. Older practitioners are generally happier with their jobs than younger employees, while people who are more experienced in their jobs are more highly satisfied than those who are less experienced.\nOrganizational factors include nature of work, income, interactions and career development which includes autonomy, working conditions, recognition of work, professional development, availability of resources, and organization structure. Autonomy given to employees allow them to feel confident and competent thus allowing then to make a positive impact on the organization. If employees feel that the work done by them is meaningful, has significance and is recognized, they will be satisfied. Any person would want a pay system that they perceive as just, unequivocal, and in line with their expectations. Satisfaction is likely to result when one’s income is seen as fair based on job demands, individual skill level, and in accordance with community pay standards. In any organization, employees seek fair promotion policies and practices that provide opportunities for personal growth, additional responsibilities, and improved social status. Individuals who recognize that promotional decisions are made in a fair and just manner, experience satisfaction with their jobs. Work environments that support personal comfort, growth and good job performance are highly desirable. Most employees also prefer working closer to home, in a clean and modern facility with adequate staff and resources may result in satisfaction or dissatisfaction if work environment is not suitable. Practices with more-satisfied employees tend to be more productive than those with less-satisfied employees [16].\nProfessional relationships, location of practice and quality of interaction indicating a need for healthy work interaction and professional networking which is a major cause of satisfaction. Studies have found that employees feel satisfaction when superior staff understands them, is friendly and approachable, compliments good performance, listens to employees’ opinions, and shows a personal interest in them. On the contrary, practices with toxic work culture where staff feel undermined, disrespected, undervalued and threated, are generally dissatisfied [17].\nFinally, professional factors include job identity, work status, job security and qualification. Employees with renewed contracts/ permanent placement have a sense of job security, feel more satisfied. This gives them job identity and status. Career development and qualification upgrade incentives provide opportunities for staff enhance their skills and knowledge which in turn benefits both the organization and staff and are satisfying aspects of workplaces [18].\nJob satisfaction can be an important indicator of how DHPs feel about their jobs add value to their practice such as retentions of skilled staff, increase productivity, reduce turnover, enhanced patient satisfaction and loyalty, improve teamwork, high quality service delivery and care. On the other hand, dissatisfaction can cause employees to have frequent absenteeism, high turnover, thoughts of changing profession, low motivation and moral, high stress levels and poor performance. All these factors are interrelated and has an implication on the other which determines satisfaction or dissatisfaction.\n\nFig. 1Conceptual framework\n\nConceptual framework\nA coherent conceptual framework is constructed to elaborate the theoretical relationship between job satisfaction factors and employee commitment to organizational culture as shown in Fig. 1. This conceptual framework summarizes the variables that affect job satisfaction among dental professionals. This relationship will be tested and verified based on this research.\nFactors categorized as personal and organizational determinants are associated with job satisfaction and can influence a person’s level of satisfaction and organizational commitment [1].\nPersonal determinants include age, gender, and level of education. Gender based differences is a determinant of job satisfaction, female practitioners ascribe more importance to social factors, while males place greater value on income, progression and other extrinsic aspects [15]. Older practitioners are generally happier with their jobs than younger employees, while people who are more experienced in their jobs are more highly satisfied than those who are less experienced.\nOrganizational factors include nature of work, income, interactions and career development which includes autonomy, working conditions, recognition of work, professional development, availability of resources, and organization structure. Autonomy given to employees allow them to feel confident and competent thus allowing then to make a positive impact on the organization. If employees feel that the work done by them is meaningful, has significance and is recognized, they will be satisfied. Any person would want a pay system that they perceive as just, unequivocal, and in line with their expectations. Satisfaction is likely to result when one’s income is seen as fair based on job demands, individual skill level, and in accordance with community pay standards. In any organization, employees seek fair promotion policies and practices that provide opportunities for personal growth, additional responsibilities, and improved social status. Individuals who recognize that promotional decisions are made in a fair and just manner, experience satisfaction with their jobs. Work environments that support personal comfort, growth and good job performance are highly desirable. Most employees also prefer working closer to home, in a clean and modern facility with adequate staff and resources may result in satisfaction or dissatisfaction if work environment is not suitable. Practices with more-satisfied employees tend to be more productive than those with less-satisfied employees [16].\nProfessional relationships, location of practice and quality of interaction indicating a need for healthy work interaction and professional networking which is a major cause of satisfaction. Studies have found that employees feel satisfaction when superior staff understands them, is friendly and approachable, compliments good performance, listens to employees’ opinions, and shows a personal interest in them. On the contrary, practices with toxic work culture where staff feel undermined, disrespected, undervalued and threated, are generally dissatisfied [17].\nFinally, professional factors include job identity, work status, job security and qualification. Employees with renewed contracts/ permanent placement have a sense of job security, feel more satisfied. This gives them job identity and status. Career development and qualification upgrade incentives provide opportunities for staff enhance their skills and knowledge which in turn benefits both the organization and staff and are satisfying aspects of workplaces [18].\nJob satisfaction can be an important indicator of how DHPs feel about their jobs add value to their practice such as retentions of skilled staff, increase productivity, reduce turnover, enhanced patient satisfaction and loyalty, improve teamwork, high quality service delivery and care. On the other hand, dissatisfaction can cause employees to have frequent absenteeism, high turnover, thoughts of changing profession, low motivation and moral, high stress levels and poor performance. All these factors are interrelated and has an implication on the other which determines satisfaction or dissatisfaction.\n\nFig. 1Conceptual framework\n\nConceptual framework", "A coherent conceptual framework is constructed to elaborate the theoretical relationship between job satisfaction factors and employee commitment to organizational culture as shown in Fig. 1. This conceptual framework summarizes the variables that affect job satisfaction among dental professionals. This relationship will be tested and verified based on this research.\nFactors categorized as personal and organizational determinants are associated with job satisfaction and can influence a person’s level of satisfaction and organizational commitment [1].\nPersonal determinants include age, gender, and level of education. Gender based differences is a determinant of job satisfaction, female practitioners ascribe more importance to social factors, while males place greater value on income, progression and other extrinsic aspects [15]. Older practitioners are generally happier with their jobs than younger employees, while people who are more experienced in their jobs are more highly satisfied than those who are less experienced.\nOrganizational factors include nature of work, income, interactions and career development which includes autonomy, working conditions, recognition of work, professional development, availability of resources, and organization structure. Autonomy given to employees allow them to feel confident and competent thus allowing then to make a positive impact on the organization. If employees feel that the work done by them is meaningful, has significance and is recognized, they will be satisfied. Any person would want a pay system that they perceive as just, unequivocal, and in line with their expectations. Satisfaction is likely to result when one’s income is seen as fair based on job demands, individual skill level, and in accordance with community pay standards. In any organization, employees seek fair promotion policies and practices that provide opportunities for personal growth, additional responsibilities, and improved social status. Individuals who recognize that promotional decisions are made in a fair and just manner, experience satisfaction with their jobs. Work environments that support personal comfort, growth and good job performance are highly desirable. Most employees also prefer working closer to home, in a clean and modern facility with adequate staff and resources may result in satisfaction or dissatisfaction if work environment is not suitable. Practices with more-satisfied employees tend to be more productive than those with less-satisfied employees [16].\nProfessional relationships, location of practice and quality of interaction indicating a need for healthy work interaction and professional networking which is a major cause of satisfaction. Studies have found that employees feel satisfaction when superior staff understands them, is friendly and approachable, compliments good performance, listens to employees’ opinions, and shows a personal interest in them. On the contrary, practices with toxic work culture where staff feel undermined, disrespected, undervalued and threated, are generally dissatisfied [17].\nFinally, professional factors include job identity, work status, job security and qualification. Employees with renewed contracts/ permanent placement have a sense of job security, feel more satisfied. This gives them job identity and status. Career development and qualification upgrade incentives provide opportunities for staff enhance their skills and knowledge which in turn benefits both the organization and staff and are satisfying aspects of workplaces [18].\nJob satisfaction can be an important indicator of how DHPs feel about their jobs add value to their practice such as retentions of skilled staff, increase productivity, reduce turnover, enhanced patient satisfaction and loyalty, improve teamwork, high quality service delivery and care. On the other hand, dissatisfaction can cause employees to have frequent absenteeism, high turnover, thoughts of changing profession, low motivation and moral, high stress levels and poor performance. All these factors are interrelated and has an implication on the other which determines satisfaction or dissatisfaction.\n\nFig. 1Conceptual framework\n\nConceptual framework", "[SUBTITLE] Study design and setting [SUBSECTION] This study applied a descriptive qualitative study design among DHPs of Fiji which allowed for much richer and detailed responses from subjects being studied [19]. This study was conducted among the DHPs based at the dental clinics of the Ministry of Health and Medical Services (MoHMS) that provide prosthetic services located in Fiji namely Nakasi Dental Clinic, Lautoka Hospital, Labasa Hospital, and at the FNU Dental Clinic in Suva. These clinics had been purposively selected as they had an attached established laboratory support with registered staff under the Fiji Dental Council (FDC). In this study, the Consolidated Criteria for Reporting Qualitative Studies (COREQ) guidelines were followed to ensure rigor (Appendix 1).\nThis study applied a descriptive qualitative study design among DHPs of Fiji which allowed for much richer and detailed responses from subjects being studied [19]. This study was conducted among the DHPs based at the dental clinics of the Ministry of Health and Medical Services (MoHMS) that provide prosthetic services located in Fiji namely Nakasi Dental Clinic, Lautoka Hospital, Labasa Hospital, and at the FNU Dental Clinic in Suva. These clinics had been purposively selected as they had an attached established laboratory support with registered staff under the Fiji Dental Council (FDC). In this study, the Consolidated Criteria for Reporting Qualitative Studies (COREQ) guidelines were followed to ensure rigor (Appendix 1).\n[SUBTITLE] Sampling [SUBSECTION] A non-probability sampling design [20] in the form of a homogeneous purposive sampling method was used to gather data as target population information. This study included all the DHPs in Fiji. Those DHPs who met the study criteria were included in this study as study sample. They were Dentists and dental technicians registered under the FDC, Minimum work experience of 6 months, working at the selected clinics in the time of data collection, must be attached to the dental prosthetic clinic/ lab, and DHPs of both genders or any ethnicity. Those who worked in private dental clinics or were not willing to participate in this study were excluded.\nA total of 29 DHPs were approached for the in-depth interviews. The sample participants included 18 dental technicians and 11 dentists. The final sample size was based on the data saturation.\nA non-probability sampling design [20] in the form of a homogeneous purposive sampling method was used to gather data as target population information. This study included all the DHPs in Fiji. Those DHPs who met the study criteria were included in this study as study sample. They were Dentists and dental technicians registered under the FDC, Minimum work experience of 6 months, working at the selected clinics in the time of data collection, must be attached to the dental prosthetic clinic/ lab, and DHPs of both genders or any ethnicity. Those who worked in private dental clinics or were not willing to participate in this study were excluded.\nA total of 29 DHPs were approached for the in-depth interviews. The sample participants included 18 dental technicians and 11 dentists. The final sample size was based on the data saturation.\n[SUBTITLE] Data collection tool [SUBSECTION] The data had been collected through a semi-structured open-ended questionnaire in the form of an interview via zoom with participants. The questions were developed based on the literatures and study research questions which comprised of two sections including (Appendix 2):\n1) Demographic variables containing five questions including: age, race, gender, qualification, years of work experience and dental specialty.\n2) Seven open ended questions related to job satisfaction in the English language as all participants were literate in English which eliminated the need for translation in other languages.\nThe data had been collected through a semi-structured open-ended questionnaire in the form of an interview via zoom with participants. The questions were developed based on the literatures and study research questions which comprised of two sections including (Appendix 2):\n1) Demographic variables containing five questions including: age, race, gender, qualification, years of work experience and dental specialty.\n2) Seven open ended questions related to job satisfaction in the English language as all participants were literate in English which eliminated the need for translation in other languages.\n[SUBTITLE] Data collection [SUBSECTION] Due to the global pandemic crisis of COVID 19, the study procedure had to be amended to accommodate the movement restrictions put in place by the Fijian government. Thus, the use of a virtual platform had been employed to aid in data collection. Ethical approval was obtained from the College Health Research Ethics Committee (CHREC) with the ID#032.21 before commencement of the study. A list of suitable participants was made by contacting the dental clinics and those that met the inclusion/ exclusion criteria were contacted via phone/email requesting their participation and date and time of interview was confirmed. An information sheet together with a consent form was emailed to them. On the day of interview, a guideline was explained, and a verbal consent was recorded before commencing the interview question on Zoom. It was made clear to the participants that their identity would not be disclosed, confidentiality will be maintained and were requested to give their frank opinions. Each interview took about 20–30 min. During the interview, the conversation was recorded and note taking was done by the principal researcher. After each interview, the resulting audio was transcribed, and notes from the interviews were reviewed to identify all references and content that related to job satisfaction. Transcripts were discussed with participants for comment and/or correction.\nDue to the global pandemic crisis of COVID 19, the study procedure had to be amended to accommodate the movement restrictions put in place by the Fijian government. Thus, the use of a virtual platform had been employed to aid in data collection. Ethical approval was obtained from the College Health Research Ethics Committee (CHREC) with the ID#032.21 before commencement of the study. A list of suitable participants was made by contacting the dental clinics and those that met the inclusion/ exclusion criteria were contacted via phone/email requesting their participation and date and time of interview was confirmed. An information sheet together with a consent form was emailed to them. On the day of interview, a guideline was explained, and a verbal consent was recorded before commencing the interview question on Zoom. It was made clear to the participants that their identity would not be disclosed, confidentiality will be maintained and were requested to give their frank opinions. Each interview took about 20–30 min. During the interview, the conversation was recorded and note taking was done by the principal researcher. After each interview, the resulting audio was transcribed, and notes from the interviews were reviewed to identify all references and content that related to job satisfaction. Transcripts were discussed with participants for comment and/or correction.\n[SUBTITLE] Trustworthiness and reflexivity [SUBSECTION] Trustworthiness is considered a more appropriate criterion for evaluating qualitative studies. In order to ensure the process is trustworthy, four criterions had been adopted. They are credibility, transferability, dependability, and confirmability [21]. There were many strategies to address credibility that include “prolonged engagement” and member checks. In-depth interviews were conducted over the period of 1 month and each interview lasted for at least 20–30 min. All in depth interviews were recorded and transcribed by the researcher on the same day. Every step of the thesis and results was discussed with the principal supervisor.\nTransferability related to the ability of the findings to be transferred to other contexts or settings. Because qualitative research is specific to a particular context, it is important a “thick description” of the particular research context is provided to allow the reader to assess whether it is transferable to their situation or not. Purposive sampling technique was used in this research and in-depth interviews were carried out until data saturation was achieved. Dependability ensured that the process is described in sufficient detail to facilitate another researcher to repeat the work. This study was developed from the early stages through a systematic search of the existing literature. Same in-depth interview questions were asked to all participants of the study and review of transcriptions were done to correct errors. All data was coded and the coding was checked and rechecked thorough. Confirmability was comparable to objectivity in quantitative studies. Here, the goal was to minimize investigator bias by acknowledging researcher predispositions. Adherence to this framework by adopting strategies, such as those outlined, to address the individual criteria supports a rigorous research process. Data was checked by the principal supervisor-MM. During the interview process, note taking of the participants comments was done by principle researcher SK.\nTrustworthiness is considered a more appropriate criterion for evaluating qualitative studies. In order to ensure the process is trustworthy, four criterions had been adopted. They are credibility, transferability, dependability, and confirmability [21]. There were many strategies to address credibility that include “prolonged engagement” and member checks. In-depth interviews were conducted over the period of 1 month and each interview lasted for at least 20–30 min. All in depth interviews were recorded and transcribed by the researcher on the same day. Every step of the thesis and results was discussed with the principal supervisor.\nTransferability related to the ability of the findings to be transferred to other contexts or settings. Because qualitative research is specific to a particular context, it is important a “thick description” of the particular research context is provided to allow the reader to assess whether it is transferable to their situation or not. Purposive sampling technique was used in this research and in-depth interviews were carried out until data saturation was achieved. Dependability ensured that the process is described in sufficient detail to facilitate another researcher to repeat the work. This study was developed from the early stages through a systematic search of the existing literature. Same in-depth interview questions were asked to all participants of the study and review of transcriptions were done to correct errors. All data was coded and the coding was checked and rechecked thorough. Confirmability was comparable to objectivity in quantitative studies. Here, the goal was to minimize investigator bias by acknowledging researcher predispositions. Adherence to this framework by adopting strategies, such as those outlined, to address the individual criteria supports a rigorous research process. Data was checked by the principal supervisor-MM. During the interview process, note taking of the participants comments was done by principle researcher SK.\n[SUBTITLE] Research team [SUBSECTION] The research team consisted of the principle female researcher who was a master student in health services management and attended a 2-hour training for conducting interviews (SK) while being supervised and guided by principal supervisor (MM).\nThe research team consisted of the principle female researcher who was a master student in health services management and attended a 2-hour training for conducting interviews (SK) while being supervised and guided by principal supervisor (MM).\n[SUBTITLE] Data management and analysis [SUBSECTION] The content of interview was transcribed by the main researcher. The transcribed data was then checked for sufficient quality and accuracy by main researcher before major analysis is conducted. Backup files had been created considering data management system and were updated as data preparation and analysis proceeded. The collected data were analyzed using manual thematic analysis [22]. Field notes were arranged according to codes and themes which were combined into major categories and files (word documents) will be created and labeled. The principal researcher SK read and re-read all interview transcripts and identified similar phrases and words for which numbers were assigned. The coded data that had similar characteristics was grouped together. Once grouping of similar data was completed, descriptive themes and sub themes were identified to reflect the perceptions of participants. The themes and sub-themes were checked by the principal supervisor as well MM.\nThe content of interview was transcribed by the main researcher. The transcribed data was then checked for sufficient quality and accuracy by main researcher before major analysis is conducted. Backup files had been created considering data management system and were updated as data preparation and analysis proceeded. The collected data were analyzed using manual thematic analysis [22]. Field notes were arranged according to codes and themes which were combined into major categories and files (word documents) will be created and labeled. The principal researcher SK read and re-read all interview transcripts and identified similar phrases and words for which numbers were assigned. The coded data that had similar characteristics was grouped together. Once grouping of similar data was completed, descriptive themes and sub themes were identified to reflect the perceptions of participants. The themes and sub-themes were checked by the principal supervisor as well MM.", "This study applied a descriptive qualitative study design among DHPs of Fiji which allowed for much richer and detailed responses from subjects being studied [19]. This study was conducted among the DHPs based at the dental clinics of the Ministry of Health and Medical Services (MoHMS) that provide prosthetic services located in Fiji namely Nakasi Dental Clinic, Lautoka Hospital, Labasa Hospital, and at the FNU Dental Clinic in Suva. These clinics had been purposively selected as they had an attached established laboratory support with registered staff under the Fiji Dental Council (FDC). In this study, the Consolidated Criteria for Reporting Qualitative Studies (COREQ) guidelines were followed to ensure rigor (Appendix 1).", "A non-probability sampling design [20] in the form of a homogeneous purposive sampling method was used to gather data as target population information. This study included all the DHPs in Fiji. Those DHPs who met the study criteria were included in this study as study sample. They were Dentists and dental technicians registered under the FDC, Minimum work experience of 6 months, working at the selected clinics in the time of data collection, must be attached to the dental prosthetic clinic/ lab, and DHPs of both genders or any ethnicity. Those who worked in private dental clinics or were not willing to participate in this study were excluded.\nA total of 29 DHPs were approached for the in-depth interviews. The sample participants included 18 dental technicians and 11 dentists. The final sample size was based on the data saturation.", "The data had been collected through a semi-structured open-ended questionnaire in the form of an interview via zoom with participants. The questions were developed based on the literatures and study research questions which comprised of two sections including (Appendix 2):\n1) Demographic variables containing five questions including: age, race, gender, qualification, years of work experience and dental specialty.\n2) Seven open ended questions related to job satisfaction in the English language as all participants were literate in English which eliminated the need for translation in other languages.", "Due to the global pandemic crisis of COVID 19, the study procedure had to be amended to accommodate the movement restrictions put in place by the Fijian government. Thus, the use of a virtual platform had been employed to aid in data collection. Ethical approval was obtained from the College Health Research Ethics Committee (CHREC) with the ID#032.21 before commencement of the study. A list of suitable participants was made by contacting the dental clinics and those that met the inclusion/ exclusion criteria were contacted via phone/email requesting their participation and date and time of interview was confirmed. An information sheet together with a consent form was emailed to them. On the day of interview, a guideline was explained, and a verbal consent was recorded before commencing the interview question on Zoom. It was made clear to the participants that their identity would not be disclosed, confidentiality will be maintained and were requested to give their frank opinions. Each interview took about 20–30 min. During the interview, the conversation was recorded and note taking was done by the principal researcher. After each interview, the resulting audio was transcribed, and notes from the interviews were reviewed to identify all references and content that related to job satisfaction. Transcripts were discussed with participants for comment and/or correction.", "Trustworthiness is considered a more appropriate criterion for evaluating qualitative studies. In order to ensure the process is trustworthy, four criterions had been adopted. They are credibility, transferability, dependability, and confirmability [21]. There were many strategies to address credibility that include “prolonged engagement” and member checks. In-depth interviews were conducted over the period of 1 month and each interview lasted for at least 20–30 min. All in depth interviews were recorded and transcribed by the researcher on the same day. Every step of the thesis and results was discussed with the principal supervisor.\nTransferability related to the ability of the findings to be transferred to other contexts or settings. Because qualitative research is specific to a particular context, it is important a “thick description” of the particular research context is provided to allow the reader to assess whether it is transferable to their situation or not. Purposive sampling technique was used in this research and in-depth interviews were carried out until data saturation was achieved. Dependability ensured that the process is described in sufficient detail to facilitate another researcher to repeat the work. This study was developed from the early stages through a systematic search of the existing literature. Same in-depth interview questions were asked to all participants of the study and review of transcriptions were done to correct errors. All data was coded and the coding was checked and rechecked thorough. Confirmability was comparable to objectivity in quantitative studies. Here, the goal was to minimize investigator bias by acknowledging researcher predispositions. Adherence to this framework by adopting strategies, such as those outlined, to address the individual criteria supports a rigorous research process. Data was checked by the principal supervisor-MM. During the interview process, note taking of the participants comments was done by principle researcher SK.", "The research team consisted of the principle female researcher who was a master student in health services management and attended a 2-hour training for conducting interviews (SK) while being supervised and guided by principal supervisor (MM).", "The content of interview was transcribed by the main researcher. The transcribed data was then checked for sufficient quality and accuracy by main researcher before major analysis is conducted. Backup files had been created considering data management system and were updated as data preparation and analysis proceeded. The collected data were analyzed using manual thematic analysis [22]. Field notes were arranged according to codes and themes which were combined into major categories and files (word documents) will be created and labeled. The principal researcher SK read and re-read all interview transcripts and identified similar phrases and words for which numbers were assigned. The coded data that had similar characteristics was grouped together. Once grouping of similar data was completed, descriptive themes and sub themes were identified to reflect the perceptions of participants. The themes and sub-themes were checked by the principal supervisor as well MM.", "Twenty-nine DHPs participated in the online one on one interview via zoom. Females made up majority of the participants (76%, n = 22). Most of the participants were in the 30–40 age range (48%, n = 14). Participants were made up of two ethnicities, ITaukei (14%, n = 4) and Indo Fijians (86%, n = 25). Looking at educational levels majority of the staff just have a baseline graduate qualification (66%, n = 19), post graduate qualification (31%, n = 9) and just 1 staff who progressed to become a specialist (3%). MoHMS is the largest employer of the participants (72%, n = 21) from Nakasi dental clinic (28%, n = 8), Lautoka dental clinic (24%, n = 7) and Labasa dental clinic (21%, n = 6) followed by FNU (28%, n = 8). In terms of years of experience, 24%(n = 7) had more than 20 years of experience. DHPs who are involved in prosthetic services were dentists (38%, n = 11) and dental technicians (62%, n = 18) (Table 1).\n\nTable 1General information of participants (n = 29)CharacteristicsFrequencyPercentage\nGender\nMale724Female2276\nEthnicity\nIndo Fijian2586ITaukei414\nAge (years)\n20–3062131–40144841–5051751–60414\nQualification\nGraduate qualification1966Post graduate931Specialist13\nWork experience (years)\nLess than 20yrs2276More than 20yrs724\nLocation of practice\nNakasi dental clinic828Lautoka dental clinic724Labasa dental clinic621FNU828\nDental Specialty\nDentist1138Dental technician1862\n\nGeneral information of participants (n = 29)\n[SUBTITLE] Themes related to job satisfaction [SUBSECTION] Three major themes had emerged from the thematic analysis which included perception on work-related factors, barriers of job satisfaction and recommendation to promote job satisfaction. Eight sub-themes identified under three main themes including nature of work, work perspectives, work environment, limitations, Covid 19, income, professional relationships, and professional development.\nUnder sub-themes, codes were identified as summarized in Table 2. Participants quotations were referenced as P1 to P 29, DHPs –D (dentist) or DHPs –DT (dental technician).\n\nTable 2Themes and sub-themes identifiedThemesSub-themesCodesPerception on work related factorsNature of workUnique job, feel satisfied, highly recommend, no job opportunitiesWork perspectivesVery hectic job, very tiring, multitaskingWork environmentIdeal work environment, Not fully utilize skills, Outdated facilitiesBarriers of job satisfactionLimitationsFrustration due to limitations, lack of support, lack of equipment’s and materialCovid 19Work becomes risky, not practicing full range, reduced workload, work environment becomes frustratingIncomeDissatisfied with salary, return on investment, are in debtRecommendations to promote job satisfactionProfessional relationshipsGood relationship, good teamwork, supportive supervisorsProfessional developmentProfession is bottle necked, take up non-clinical courses, lack of post graduate courses\n\nThemes and sub-themes identified\nThree major themes had emerged from the thematic analysis which included perception on work-related factors, barriers of job satisfaction and recommendation to promote job satisfaction. Eight sub-themes identified under three main themes including nature of work, work perspectives, work environment, limitations, Covid 19, income, professional relationships, and professional development.\nUnder sub-themes, codes were identified as summarized in Table 2. Participants quotations were referenced as P1 to P 29, DHPs –D (dentist) or DHPs –DT (dental technician).\n\nTable 2Themes and sub-themes identifiedThemesSub-themesCodesPerception on work related factorsNature of workUnique job, feel satisfied, highly recommend, no job opportunitiesWork perspectivesVery hectic job, very tiring, multitaskingWork environmentIdeal work environment, Not fully utilize skills, Outdated facilitiesBarriers of job satisfactionLimitationsFrustration due to limitations, lack of support, lack of equipment’s and materialCovid 19Work becomes risky, not practicing full range, reduced workload, work environment becomes frustratingIncomeDissatisfied with salary, return on investment, are in debtRecommendations to promote job satisfactionProfessional relationshipsGood relationship, good teamwork, supportive supervisorsProfessional developmentProfession is bottle necked, take up non-clinical courses, lack of post graduate courses\n\nThemes and sub-themes identified\n[SUBTITLE] Theme 1: Perception on work-related factors [SUBSECTION] Three factors were discussed under this theme including the nature of work, work perspectives and work environment.\n[SUBTITLE] Nature of work [SUBSECTION] The first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession.\nSome considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\n“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\nWhen asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\n“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\nIn contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\nThe first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession.\nSome considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\n“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\nWhen asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\n“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\nIn contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n[SUBTITLE] Work perspectives [SUBSECTION] Participants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\n“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\nMost of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\nParticipants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\n“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\nMost of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n[SUBTITLE] Work environment [SUBSECTION] Here participants describe the type of work environment and conditions they are working in.\nSome participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\n“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\nSome find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\n“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\nHere participants describe the type of work environment and conditions they are working in.\nSome participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\n“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\nSome find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\n“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\nThree factors were discussed under this theme including the nature of work, work perspectives and work environment.\n[SUBTITLE] Nature of work [SUBSECTION] The first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession.\nSome considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\n“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\nWhen asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\n“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\nIn contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\nThe first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession.\nSome considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\n“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\nWhen asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\n“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\nIn contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n[SUBTITLE] Work perspectives [SUBSECTION] Participants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\n“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\nMost of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\nParticipants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\n“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\nMost of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n[SUBTITLE] Work environment [SUBSECTION] Here participants describe the type of work environment and conditions they are working in.\nSome participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\n“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\nSome find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\n“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\nHere participants describe the type of work environment and conditions they are working in.\nSome participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\n“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\nSome find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\n“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\n[SUBTITLE] Theme 2: barriers of job satisfaction [SUBSECTION] Different limitations, challenges faced due to Covid-19 and inadequate income were the main challenges the study participants highlighted.\n[SUBTITLE] Limitations [SUBSECTION] Most participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\n“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\nA few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\nMost participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\n“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\nA few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n[SUBTITLE] Covid 19 [SUBSECTION] COVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\n“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\nA few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\n“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\nSome expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\nCOVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\n“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\nA few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\n“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\nSome expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n[SUBTITLE] Income [SUBSECTION] Majority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\n“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\nPerceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\n“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\nMajority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\n“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\nPerceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\n“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\nDifferent limitations, challenges faced due to Covid-19 and inadequate income were the main challenges the study participants highlighted.\n[SUBTITLE] Limitations [SUBSECTION] Most participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\n“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\nA few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\nMost participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\n“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\nA few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n[SUBTITLE] Covid 19 [SUBSECTION] COVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\n“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\nA few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\n“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\nSome expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\nCOVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\n“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\nA few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\n“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\nSome expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n[SUBTITLE] Income [SUBSECTION] Majority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\n“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\nPerceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\n“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\nMajority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\n“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\nPerceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\n“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\n[SUBTITLE] Theme 3: recommendation to promote job satisfaction [SUBSECTION] Participants advised the needs for changing in professional relationship as well as professional development to increase the level of job satisfaction.\n[SUBTITLE] Professional relationships [SUBSECTION] Here we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement.\nMajority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\n“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\nMost participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\n“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\nSome mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\nHere we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement.\nMajority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\n“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\nMost participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\n“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\nSome mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n[SUBTITLE] Professional development [SUBSECTION] This sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\n“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\nThey also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\n“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\nMost participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\n“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\nThis sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\n“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\nThey also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\n“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\nMost participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\n“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\nParticipants advised the needs for changing in professional relationship as well as professional development to increase the level of job satisfaction.\n[SUBTITLE] Professional relationships [SUBSECTION] Here we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement.\nMajority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\n“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\nMost participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\n“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\nSome mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\nHere we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement.\nMajority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\n“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\nMost participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\n“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\nSome mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n[SUBTITLE] Professional development [SUBSECTION] This sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\n“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\nThey also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\n“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\nMost participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\n“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\nThis sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\n“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\nThey also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\n“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\nMost participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\n“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)", "Three major themes had emerged from the thematic analysis which included perception on work-related factors, barriers of job satisfaction and recommendation to promote job satisfaction. Eight sub-themes identified under three main themes including nature of work, work perspectives, work environment, limitations, Covid 19, income, professional relationships, and professional development.\nUnder sub-themes, codes were identified as summarized in Table 2. Participants quotations were referenced as P1 to P 29, DHPs –D (dentist) or DHPs –DT (dental technician).\n\nTable 2Themes and sub-themes identifiedThemesSub-themesCodesPerception on work related factorsNature of workUnique job, feel satisfied, highly recommend, no job opportunitiesWork perspectivesVery hectic job, very tiring, multitaskingWork environmentIdeal work environment, Not fully utilize skills, Outdated facilitiesBarriers of job satisfactionLimitationsFrustration due to limitations, lack of support, lack of equipment’s and materialCovid 19Work becomes risky, not practicing full range, reduced workload, work environment becomes frustratingIncomeDissatisfied with salary, return on investment, are in debtRecommendations to promote job satisfactionProfessional relationshipsGood relationship, good teamwork, supportive supervisorsProfessional developmentProfession is bottle necked, take up non-clinical courses, lack of post graduate courses\n\nThemes and sub-themes identified", "Three factors were discussed under this theme including the nature of work, work perspectives and work environment.\n[SUBTITLE] Nature of work [SUBSECTION] The first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession.\nSome considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\n“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\nWhen asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\n“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\nIn contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\nThe first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession.\nSome considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\n“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\nWhen asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\n“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\nIn contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n[SUBTITLE] Work perspectives [SUBSECTION] Participants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\n“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\nMost of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\nParticipants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\n“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\nMost of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n[SUBTITLE] Work environment [SUBSECTION] Here participants describe the type of work environment and conditions they are working in.\nSome participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\n“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\nSome find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\n“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\nHere participants describe the type of work environment and conditions they are working in.\nSome participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\n“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\nSome find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\n“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)", "The first few questions asked to DHPs were based on their profession and the perceptions they have of their work. These professionals share their experience of being in this profession. Here participants described their experience of being in the profession.\nSome considered their profession as a unique job due to consequence of the services they provide. It makes a good memory for the patients when they receive a service that makes them happy.“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\n“Being a dentist, I think it is a unique job, it is a good service that we provide to people as we maintain peoples smile, make peoples smile, so it is very satisfying to see patients happy which encourages me to provide more and better services.” (P 24, DHPs - D)\nWhen asked if they would recommend this career to a high school student who is deciding which career path to take, some said they would recommend based on satisfaction they get from being in the profession:“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\n“I highly recommend this career as it is very satisfying. You feel great when you see your patient satisfied.” (P 10, DHPs -DT)\nIn contrast, majority participants said they would not recommend this career based the investment they make towards the career and lack of job opportunity.“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)\n“Well looking at the current situation, my answer would be not as there are no job opportunities at the moment. It is not worth the time and investment put into the study.” (P 05, DHPs -DT)", "Participants were asked to describe what their job is like most of the time. Some said that despite being a very hectic job, due to services they provide that serve people they still enjoy doing it.“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\n“Its quiet hectic and messy, but if you have passion for this job, it is very enjoyable as we serve the older generation in the community. I really like working in the prosthetic service.” (P 09, DHPs -DT)\nMost of the participant’s mentioned multitasking at their workplace that limit them to take care of themselves.“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)\n“I would say it’s quiet multitasking working at the school. It starts off with teaching and clinical practice as well as research and you don’t get bored with what you are doing, you keep learning new things and a lot of problem solving.” (P 07 DHPs –D)", "Here participants describe the type of work environment and conditions they are working in.\nSome participants mentioned that they do not have an ideal work environment that could make a better situation for them to enjoy.“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\n“My work environment is not ideal; space is constrained, and we are overcrowded. Equipment’s are really old, and materials are not available readily. I am not satisfied with the type of work that we do. We are not able to apply our skills that we graduated with at MoHMS. Services are very limited.” (P 04 DHPs - DT)\nSome find their work disappointing as they are not able to fully utilize and practicing all their skills.“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)\n“We do removable prosthesis at MoHMS. I’m not satisfied with my work but its ok for the time being. At the moment we only focus on removable prosthetics only. So, we are kind of confined to one part of prosthetics whereas the broader part we are not doing so it’s a bit disappointing not being able to practice all our skills that we graduate with.” (P 14, DHPs -DT)", "Different limitations, challenges faced due to Covid-19 and inadequate income were the main challenges the study participants highlighted.\n[SUBTITLE] Limitations [SUBSECTION] Most participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\n“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\nA few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\nMost participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\n“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\nA few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n[SUBTITLE] Covid 19 [SUBSECTION] COVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\n“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\nA few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\n“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\nSome expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\nCOVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\n“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\nA few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\n“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\nSome expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n[SUBTITLE] Income [SUBSECTION] Majority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\n“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\nPerceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\n“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\nMajority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\n“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\nPerceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\n“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)", "Most participants expressed their frustrations due to the limiting situations they face at their workplace. They don’t provide all services the y expect as well as the equipment that helps them to provide better services.“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\n“Currently at MOHMS we are only doing complete and partial dentures. The things we have learnt from FNU like crown and bridge and orthodontics, we are unable to practice that here because it is not being offered by the ministry. Our machines are really old and in poor conditions and we have to share equipment’s among staff which is quiet frustrating. Location and size of our lab is really small and is difficult for older patients to excess.” (P 19, DHPs -DT)\nA few participants voiced that due to lack of support from organization, there is poor advancement in service delivery:“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)\n“As a teaching institute, we should incorporate the latest technology into our teaching but the university is quiet hesitant to assist in that and stagnates the advancements in service and delivery. There needs to be back up support for the lab and equipment breakdowns.” (P 06, DHPs -DT)", "COVID 19 also posed many work challenges. Some participant mentioned that works becomes risky due high number of patients and short of staff that directly affected the quality of service as well as their satisfaction.“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\n“The type of work that we do is quiet risky especially now during COVID it becomes very risky and sometimes there are increased number of patients we have to see and we are short staffed and we do work beyond our JD. So, it becomes very loaded.” (P 15, DHPs –D)\nA few participants mentioned not being able to practice a full range of dental services during Covid-19 pandemic.“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\n“AT the moment due to COVID, we limited to the range of services we provide. we are only just doing emergency extractions and preventive dentistry.” (P23, DHPs – D)\nSome expressed that COVID has reduced their workload due to restriction of number of patients they visit daily.“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)\n“We see so many patients in a day that we get burnout. It is hard to continuously see 15 patients a day. now thankfully to COVID, they have really restricted the number of patients we see in a day as they also have to be swabbed. So that has given us a bit of a breather.” (P 16, DHPs – D)", "Majority of the participants expressed their dissatisfaction with the type of salary they are getting for the amount of work they do.“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\n“Actually, the salary does not justify our profession. We tend to work more, and if we start thinking of that, we are not satisfied as other professions like the medical staffs get paid more.” (P 24, DHPs -D)\nPerceptions based on investments made for the career. Majority participants mentioned that their returns on their investments made for their career are not worthwhile.“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)\n“I personally feel that the pay Dental technicians get is not enough as we invest so much into our studies, and we expect to get some returns when we start working doesn’t seem to be a good investment. Secondly, dental technology is a specialized field and not everyone can do it, I feel that we should be paid more if that is to be considered.” (P 04, DHPs -DT)", "Participants advised the needs for changing in professional relationship as well as professional development to increase the level of job satisfaction.\n[SUBTITLE] Professional relationships [SUBSECTION] Here we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement.\nMajority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\n“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\nMost participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\n“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\nSome mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\nHere we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement.\nMajority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\n“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\nMost participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\n“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\nSome mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n[SUBTITLE] Professional development [SUBSECTION] This sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\n“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\nThey also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\n“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\nMost participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\n“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\nThis sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\n“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\nThey also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\n“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\nMost participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\n“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)", "Here we look at participants expressing their perceptions on interaction they have with their supervisors and managers, colleagues and coworkers, the type of teamwork they have going on and employee involvement.\nMajority of the participants expressed that they have a good relationship with their coworker which helps them to make a better situation to promote their job satisfaction.“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\n“For me the working environment is good, it’s very friendly, my colleagues are very good.” (P 05, DHPs -DT)\nMost participants mentioned they have a very good team with great teamwork and feedback.“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\n“Work environment varies day to day. Someday are good and some are not so. We have a very good team and teamwork, good feedback and communication. With teamwork, come responsibilities which should be in par with communication. Each staff have their own workload and has to be mindful that the workload we have is completed in a timely manner. We talk to each other as feedbacks are very important so that each of the staffs can improve as in any organization, feedbacks are quiet important. Another thing is that without teamwork, any school or organization will not be able to function. So, the team I’m working with, they are very good.” (P 08, DHPs -D)\nSome mentioned that their supervisors are supportive and engaged.“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)\n“I think my current supervisor is ummm quiet supportive and engaged, empowering and makes it possible for you to progress in whatever you want to do in terms of work. He is the kind of person who takes on ideas quiet easily.” (P 11, DHPs -D)", "This sub-theme describes staff development as experienced by each participant. Majority of the participants feel that there is no growth, and the profession has limited progression.“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\n“As technicians, I feel this profession is bottle necked. To progress we need to have more courses included in the dental technology area.” (P 01, DHPs -DT)\nThey also added that due to dentistry being an expensive field, they had to take up non – clinical courses.“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\n“I think dentistry is such a field that is quiet expensive to begin with. So, when it comes to clinical training or post grad clinical training, there is a lot of money that is required, so financially this organization might not be very supportive and this is one thing that has hindered my development. I had to take up a non-clinical masters just because of this very reason.” (P 07, DHPs -D)\nMost participants feel there are not many professional development opportunities such as postgraduate courses available which limits staff from progressing in their career.“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)\n“At MoHMS it is quiet limited because each specialty of dentistry has not really developed except for oral surgery in which there is a master’s program available locally. Areas like endodontics and prosthodontics there are no training available and there are no specific dental officers, so due to this there is always rotations happening so no one is really able to specialize in a particular area that they like.” (P 12, DHPs -D)", "Majority of the participants were satisfied with the nature of their work. They reported feeling satisfaction with providing dental services and seeing their patients satisfied. Overall, dentists have a high level of professional satisfaction and the level of satisfaction is influenced by various socio demographic and psycho-behavioral factors [14]. Participants in this study described their profession as a very hands-on work, fulfilling and unique job. According to Kaipa et al. [23], job satisfaction is a pleasurable or positive emotional state as a result of one’s job experience, that one’s job fulfils or allows for fulfilment of one’s own job values. Furthermore, he describes dentistry as rewarding profession that combines art and science, personal communication skills, with high ethical standards. This profession is a social interaction between helper and recipient in their limited job setting and with personal characteristics. This was evident in the findings of this study where participants mentioned dentistry as being a rewarding profession. Rahmi et al. [24] concluded that dentistry is a caring and prestigious profession which were important motivators for job satisfaction. Some said that they would recommend this career based on the satisfaction they get from the profession. Findings from a study conducted in Romania by Murariu et al. [25] indicates that 52% of DHPs said they would choose dentistry as a career again as they found it to be a satisfying career. On the contrary, the findings of an Indian study suggest that majority of the DHPs included in the study were partly satisfied with the dental profession as a career. This is similar to the findings of the current study where due to market saturation and poor return of investments were hindering factors that led to dissatisfaction of the career.\nTaking into consideration the amount of responsibility DHPs have, dentistry is one of the most stressful of the health professions [3]. DHPs tend to address a large number of challenges faced on a daily basis: healthcare sector crisis, unsatisfied patients, lack of professionalism among the staff, unsatisfactory working conditions, stress, and pressure from the environment regarding high social and economic standards related to dental professionals [4]. All of these contribute to a strong dissatisfaction of DHPs, which is reflected in their everyday practice [8]. Additionally, some least satisfying aspects were related to the lack of motivation for continuing professional education and ongoing professional training [25]. DHPs in different work settings reported differences in satisfaction level. Those working in MoHMS were mostly dissatisfied with the work they are doing in their profession. Majority of the participants felt they are limited to only a few basic procedures they can perform they found to be disappointing. Some participant said that their job required them to multitask, play various roles in their workplace and take up administrative responsibilities which they find to be burdening. This can be related to the finding of another study stating that despite having the same education ⁄training levels, dentists in the public and private system should provide similar type of care and services [6]. However, public dentists are constrained by what the public system can afford to provide and which limits their ability to practice a full range of dentistry [26].\nWork environment plays a huge role in job satisfaction and its importance cannot be underestimated [27, 28]. The working environment is one of the most crucial factors which influence the level of satisfaction as well as motivation of its employees. A study conducted by Agbozo et al. [27] found that there is direct correlation between work environment and job satisfaction. His findings concluded that work environment has a significant effect on employees’ satisfaction and emphasized the need for management to improve the work environment of employees to boost productivity. Majority participants in the study were not happy in the work environment they were in. They found it was not an ideal environment due to many reasons such as over crowdedness due to space constrains, working with poor quality materials and outdated machines and equipment’s, lack of resources available to them that inhibits good quality service delivery. A study in the United States of America (USA), reinforces this finding based on staff morale having a linear and positive relationship with organizational productivity [28].\nMajority of the participants expressed their frustrations due to the limitedness of their profession in terms of range of services they can offer, lack of specialization limits the profession from progressing. These were identified as some of the major dissatisfying aspects [13]. The frustration was a result of not being able to fully utilize their skills and knowledge they graduated with at their workplace due to lack of availability of resources to cater for advanced services. This had direct implications on the range of services that could be offered at their workplace [27]. Due to only having 4 dental labs in country that serves the prosthetic needs of the general population, the centralized location of practice becomes a limiting factor not only in terms of service delivery, but it limits the opportunities for more job establishments. This leads to a higher volume of patients being tended to which in turn causes overcrowding, overworked and burnout staff [25].\nCOVID 19 presented many challenges to the dental profession in Fiji. Many participants in this study experienced feelings of frustrations among team member and were concerned about the risks they were exposed to while seeing patients. This was a cause of dissatisfaction for them. According to Mishra et al. [29] concern relating to being infected with the COVID-19 contagion was one of the most frequent sources of stress among DHPs in Chhattisgarh, India and further elaborates that dentistry is such a profession that involves working in close proximity to the oral cavity and its secretions, and includes procedures that are aerosol producing is unavoidable in most of the dental procedures due to which the risk of getting infected is heightened thus leading to anxiety and frustrations. Similar findings were made by Gohil et al. [30] where fear of contagion, subjective overload, and perceived job insecurity and loss of income are causing distress among the DHPs. Some participant’s expressed that COVID has reduced their workload which allowed for some feeling of satisfaction. Similar findings were made by Prasetyo et al. [31] where Indonesian DHPs were found to have significant association with job stress reduction leading to a higher satisfaction level. However, Celik et al. [32] concluded that DHPs in the public sector examined reduced number of patients as compared to private practitioners during the pandemic. This led to DHPs being dissatisfied with the management of the pandemic. A few participants mentioned not being able to practice a full range of dental services and were only doing emergency extractions only. Ahamadi et al. [33] reported similar strategies implemented in Iran to decrease the risk of spread of contagion, the majority of non-emergency procedures we halted.\nAccording to many studies, income is a major determinant for job satisfaction [7, 9, 29]. Income has been identified as one of the major dissatisfying aspects of this profession. Since dentistry is an expensive field to study, it is only fair to get a return on your investment made in terms of the type of pay you’d get when one starts working. In this study, majority participants felt dissatisfied as they are earning less than they invested. Some respondents reported being in debt as they are unable to repay their loans based on the salary they get. Similar findings were made by Puryer et al. [34] where 43% of DHPs were concerned with personal debt after paying for tuition fees.\nFeelings of satisfaction with supervisors were reported by majority of the participants. Some of the satisfying aspects they found were their supervisors to be very helpful, understanding, good team leader, professional, supportive and engaged. Supervisor support plays a substantial role in increasing employee job satisfaction [35]. Similar finding were made by Luzzi et al. [36] that relationships with patients, colleagues and staff contribute to job satisfaction and further pointed out that there is compounding benefits to having an effective and stable dental team.21 Thus, in terms of the job satisfaction dimensions explored within this study, working in an effective and stable team may contribute to increased satisfaction on the relationships with colleagues, patients and staff dimensions as practices become more productive environment. Healthy relationships between superiors and their staff significantly contributes to the quality of workplace performance and has social impact which positively moderates the association between healthy workplace interactions and job stress.\nProfessional development is an essential aspect of an employee’s career [28]. It is critical for maintaining a stable employment and ensure growth is not stagnated [4]. For any proficient employee, the need to upskill and grow is a major priority [28]. Majority of the participants feel that there is no growth, and the profession is bottle necked. They also added that due to dentistry being an expensive field, they had to take up non – clinical course as there are not many post graduate courses available locally which limits staff from progressing in their career which led to dissatisfaction. This indicates that there is a passion among DHPs to acquire new skills and knowledge in the field of dentistry, but due to the lack of postgraduate and specialists training locally most are opting for modules and courses outside their field of work [6]. This also indicates poor design of the dental curriculum and also that the output product is short of necessary standards and competencies desired by these professionals [27]. There is a need to broaden the dental profession spectrum from being merely a graduate program [8]. Those that have upskilled, are dissatisfied as well as are limited to the types of services they can provide at their organization. This result is similar to the findings of a Romanian study where majority of the DHPSs who had post-graduate training (which is a key factor for career development) were dissatisfied [22]. Further to this Kumar et al. [1] study showed that the majority of the Pakistani DHPs who worked in the public sector were dissatisfied with the professional and career development opportunities they received during their professional life. Literature indicates human resource in health sector needs continuous training and refresher courses. Training is known to increase the self-confidence and self-esteem of employees and improves the quality of service that would significantly elevate the morale of employees in the organization [37]. Contrary to these findings, other studies suggest that those with postgraduate and specialist trainings are more satisfied than those with a degree or diploma [3, 6, 8, 19].\n[SUBTITLE] Limitations [SUBSECTION] Findings of this research must be interpreted within the context of its limitations due to qualitative design; this study is limited to DHPs providing prosthetic services in Fiji, study findings included only DHPs working at MoHMS and FNU. It would be ideal for future researchers to include DHPs who offer prosthetic services in the private sector. Due to the setting of the study and the nature of the study participants, timing of data collection was limited. Data was collected via a virtual platform called Zoom based at a time convenient for the participants.\nFindings of this research must be interpreted within the context of its limitations due to qualitative design; this study is limited to DHPs providing prosthetic services in Fiji, study findings included only DHPs working at MoHMS and FNU. It would be ideal for future researchers to include DHPs who offer prosthetic services in the private sector. Due to the setting of the study and the nature of the study participants, timing of data collection was limited. Data was collected via a virtual platform called Zoom based at a time convenient for the participants.", "Findings of this research must be interpreted within the context of its limitations due to qualitative design; this study is limited to DHPs providing prosthetic services in Fiji, study findings included only DHPs working at MoHMS and FNU. It would be ideal for future researchers to include DHPs who offer prosthetic services in the private sector. Due to the setting of the study and the nature of the study participants, timing of data collection was limited. Data was collected via a virtual platform called Zoom based at a time convenient for the participants.", "To date, this has been the only study conducted exploring the perceptions of job satisfaction amongst DHPs in Fiji using qualitative analysis. The results of this study can be used to inform DHPs, their employers and academic institutes about what factors affect job satisfaction of individuals working within this profession. Factors such as nature of work, work perspectives of participants, work environment, limitations, Covid 19, income, professional relationships, and professional development were found to contribute to job satisfaction.\nThe empirical findings from the study indicate that DHPs employed by MoHMS and FNU, are most satisfied with their teamwork and the relationship they have with their colleagues and co-workers, followed by the nature of the work and the supervision they received. They however, indicated that they are less satisfied with professional development opportunities and least satisfied with the pay they receive and organizational support they receive. The entire structure of education and career progression in dentistry and dental technology should be addressed as a matter of urgency to achieve SDG 3, in order to encourage individuals both to join and remain within the profession.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2" ]
[ null, null, null, null, null, null, null, null, null, null, "results", null, null, null, null, null, null, null, null, null, null, null, null, "discussion", null, "conclusion", "supplementary-material", null ]
[ "Job satisfaction", "Determinants", "Dental Health Professionals", "Perceptions", "Fiji" ]
Cost-effectiveness of topical pharmacological, oral pharmacological, physical and combined treatments for acne vulgaris.
36258288
Acne vulgaris is a common skin condition that may cause psychosocial distress. There is evidence that topical treatment combinations, chemical peels and photochemical therapy (combined blue/red light) are effective for mild-to-moderate acne, while topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and photodynamic therapy are most effective for moderate-to-severe acne. Effective treatments have varying costs. The National Institute for Health and Care Excellence (NICE) in England considers cost-effectiveness when producing national clinical, public health and social care guidance.
BACKGROUND
A decision-analytical model compared costs and quality-adjusted life-years (QALYs) of effective topical pharmacological, oral pharmacological, physical and combined treatments for mild-to-moderate and moderate-to-severe acne, from the perspective of the National Health Service in England. Effectiveness data were derived from a network meta-analysis. Other model input parameters were based on published sources, supplemented by expert opinion.
METHODS
All of the assessed treatments were more cost-effective than treatment with placebo (general practitioner visits without active treatment). For mild-to-moderate acne, topical treatment combinations and photochemical therapy (combined blue/red light) were most cost-effective. For moderate-to-severe acne, topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin were the most cost-effective. Results showed uncertainty, as reflected in the wide confidence intervals around mean treatment rankings.
RESULTS
A range of treatments are cost-effective for the management of acne. Well-conducted studies are needed to examine the long-term clinical efficacy and cost-effectiveness of the full range of acne treatments.
CONCLUSION
[ "Humans", "Acne Vulgaris", "Anti-Bacterial Agents", "Cost-Benefit Analysis", "Isotretinoin", "State Medicine" ]
10091701
null
null
Methods
[SUBTITLE] Population [SUBSECTION] The study population comprised people with mild‐to‐moderate and moderate‐to‐severe acne presenting to primary care services. Separate analyses were conducted for males and females with moderate‐to‐severe acne because (i) the intervention cost of oral isotretinoin is higher for females due to pregnancy tests and increased monitoring visits, and (ii) sex‐specific discontinuation data were available for moderate‐to‐severe acne. The study population comprised people with mild‐to‐moderate and moderate‐to‐severe acne presenting to primary care services. Separate analyses were conducted for males and females with moderate‐to‐severe acne because (i) the intervention cost of oral isotretinoin is higher for females due to pregnancy tests and increased monitoring visits, and (ii) sex‐specific discontinuation data were available for moderate‐to‐severe acne. [SUBTITLE] Interventions [SUBSECTION] We included treatment classes showing evidence of efficacy vs. placebo [indicated by 95% credible intervals (CrI) around the effects not crossing the ‘no effect’ line] in the NMA that informed the NICE guideline (the source NMA). 5 We considered only treatment classes with ≥ 50 observations across randomized controlled trials (RCTs) included in the efficacy NMA, as this was deemed the minimum adequate evidence base to enable robust conclusions on clinical and cost‐effectiveness. One intervention from each treatment class was selected as representative for costing purposes. All interventions were assumed to be delivered within the NHS. Selection of interventions assessed in the economic analysis (Table 1) was based on their availability and usage in the UK, practicalities of use (e.g. fixed topical treatment combinations were preferred to nonfixed formulations), the size of their evidence base and their risk of side effects relative to other interventions within the class. Types of treatment, classes and interventions assessed in the economic analysis for each level of acne severity. BPO, benzoyl peroxide. aApplied and monitored by health professionals; bmodelled as general practitioner visits without active treatment; cform of phototherapy that involves a source of light (e.g. red light, blue light, broad‐spectrum light, daylight) and a photosensitizing chemical (e.g. 5‐aminolaevulinic acid or methyl aminolaevulinate). We included treatment classes showing evidence of efficacy vs. placebo [indicated by 95% credible intervals (CrI) around the effects not crossing the ‘no effect’ line] in the NMA that informed the NICE guideline (the source NMA). 5 We considered only treatment classes with ≥ 50 observations across randomized controlled trials (RCTs) included in the efficacy NMA, as this was deemed the minimum adequate evidence base to enable robust conclusions on clinical and cost‐effectiveness. One intervention from each treatment class was selected as representative for costing purposes. All interventions were assumed to be delivered within the NHS. Selection of interventions assessed in the economic analysis (Table 1) was based on their availability and usage in the UK, practicalities of use (e.g. fixed topical treatment combinations were preferred to nonfixed formulations), the size of their evidence base and their risk of side effects relative to other interventions within the class. Types of treatment, classes and interventions assessed in the economic analysis for each level of acne severity. BPO, benzoyl peroxide. aApplied and monitored by health professionals; bmodelled as general practitioner visits without active treatment; cform of phototherapy that involves a source of light (e.g. red light, blue light, broad‐spectrum light, daylight) and a photosensitizing chemical (e.g. 5‐aminolaevulinic acid or methyl aminolaevulinate). [SUBTITLE] Economic model structure [SUBSECTION] A decision tree was constructed to estimate total NHS costs and quality‐adjusted life years (QALYs) for each treatment over 1 year (Fig. 1). This time horizon, determined by the available follow‐up data, was deemed adequate to capture longer‐term outcomes and costs of acne treatment without significant extrapolation. Patients were modelled as having excellent, good, moderate or no perceived improvement following treatment (Supplementary Data S1 provides full details of the model). Schematic diagram of the economic model structure for people with mild‐to‐moderate and people with moderate‐to‐severe acne vulgaris. Chemical peels were assessed only in people with mild‐to‐moderate acne. Oral isotretinoin was assessed only in people with moderate‐to‐severe acne. A decision tree was constructed to estimate total NHS costs and quality‐adjusted life years (QALYs) for each treatment over 1 year (Fig. 1). This time horizon, determined by the available follow‐up data, was deemed adequate to capture longer‐term outcomes and costs of acne treatment without significant extrapolation. Patients were modelled as having excellent, good, moderate or no perceived improvement following treatment (Supplementary Data S1 provides full details of the model). Schematic diagram of the economic model structure for people with mild‐to‐moderate and people with moderate‐to‐severe acne vulgaris. Chemical peels were assessed only in people with mild‐to‐moderate acne. Oral isotretinoin was assessed only in people with moderate‐to‐severe acne. [SUBTITLE] Clinical model inputs [SUBSECTION] Relative effects on efficacy [percentage change in total acne lesion count from baseline (%CFB)], discontinuation due to any reason and discontinuation due to side effects were obtained from the source NMA 5 (Supplementary Data S2). For mild‐to‐moderate acne, the economic analysis considered only treatments with evidence of efficacy in a bias‐adjusted NMA, owing to indication of small study bias. 5 To obtain absolute effects for each treatment and outcome, we combined NMA relative effects with absolute effects of a reference treatment. We selected adapalene (topical retinoid) as the reference treatment based on data availability. Absolute effects for adapalene were estimated from large RCTs included in the source NMA, 5 and discontinuation data from a small non‐UK observational study 7 (Supplementary Data S3). Patients' perception of their acne improvement may differ from the clinical measurement of improvement as expressed by %CFB. The relationship between the two (Table 2) was determined using published trial evidence. 8 The same evidence 8 was used to determine the distribution around the mean %CFB at treatment endpoint, in order to estimate the proportion of people with excellent, good, moderate and no improvement for each treatment (Supplementary Data S4). Owing to limited and heterogeneous evidence, the risk of relapse following excellent, good and moderate improvement was based on expert opinion. Relationship between percentage change in total acne lesion count from baseline, perceived acne improvement and utility value. %CFB, percentage change in total lesion count from baseline; NA, not applicable. aSupplementary Data S5. Relative effects on efficacy [percentage change in total acne lesion count from baseline (%CFB)], discontinuation due to any reason and discontinuation due to side effects were obtained from the source NMA 5 (Supplementary Data S2). For mild‐to‐moderate acne, the economic analysis considered only treatments with evidence of efficacy in a bias‐adjusted NMA, owing to indication of small study bias. 5 To obtain absolute effects for each treatment and outcome, we combined NMA relative effects with absolute effects of a reference treatment. We selected adapalene (topical retinoid) as the reference treatment based on data availability. Absolute effects for adapalene were estimated from large RCTs included in the source NMA, 5 and discontinuation data from a small non‐UK observational study 7 (Supplementary Data S3). Patients' perception of their acne improvement may differ from the clinical measurement of improvement as expressed by %CFB. The relationship between the two (Table 2) was determined using published trial evidence. 8 The same evidence 8 was used to determine the distribution around the mean %CFB at treatment endpoint, in order to estimate the proportion of people with excellent, good, moderate and no improvement for each treatment (Supplementary Data S4). Owing to limited and heterogeneous evidence, the risk of relapse following excellent, good and moderate improvement was based on expert opinion. Relationship between percentage change in total acne lesion count from baseline, perceived acne improvement and utility value. %CFB, percentage change in total lesion count from baseline; NA, not applicable. aSupplementary Data S5. [SUBTITLE] Utility data [SUBSECTION] Utility values express people's preferences regarding health‐related quality of life (HRQoL) on a scale from 0 (death) to 1 (perfect health) and are necessary for estimating QALYs. The utility values used in the economic model (Table 2) were determined using utility data from two studies 9 , 10 identified from a systematic search, a published mapping algorithm, 11 utility values of the UK general young adult population 12 and further assumptions (Supplementary Data S5). Utility values express people's preferences regarding health‐related quality of life (HRQoL) on a scale from 0 (death) to 1 (perfect health) and are necessary for estimating QALYs. The utility values used in the economic model (Table 2) were determined using utility data from two studies 9 , 10 identified from a systematic search, a published mapping algorithm, 11 utility values of the UK general young adult population 12 and further assumptions (Supplementary Data S5). [SUBTITLE] Resource use and cost data [SUBSECTION] The analysis included intervention costs for people who completed a course of treatment and those who discontinued early and the costs of average acne care (AAC), which comprises a mixture of care currently received by people with acne in the NHS, which may include GP consultations, specialist dermatologist care, drug treatment or no treatment. AAC represented NHS care provided to people with acne following early treatment discontinuation, completion of a course of physical treatment, inadequate improvement or relapse. Costs were estimated by combining resource use with respective unit costs. Intervention resource use was based on relevant descriptions from RCTs included in the source NMA, 5 modified to reflect optimal routine practice in the UK. This incorporated (as relevant) the drug dosage and optimal duration, time of the HCP (GP and/or specialist care), laboratory testing for people receiving oral isotretinoin and any equipment used. Resource use related to AAC was obtained from UK primary care consultation and prescription data, 13 supplemented with expert opinion and further assumptions, particularly regarding specialist care received by a proportion of people with acne. Unit costs were obtained from national sources 14 , 15 , 16 , 17 and other published literature. 18 Intervention costs are summarized in Table 3, with full details on methods used for their estimation reported in Supplementary Data S6. Details of the estimation of the AAC cost are shown in Supplementary Data S7. Intervention costs of treatments for mild‐to‐moderate and moderate‐to‐severe acne following a full course (acute and maintenance treatment) and early discontinuation (2019 prices). BPO, benzoyl peroxide; F, costs for females; M, costs for males; Maint, maintenance; NA, not applicable. aCosts included drug acquisition, healthcare professional time, laboratory testing for oral isotretinoin, procedure costs for photochemical and photodynamic therapies; no costs of contraception included for oral or topical retinoids. bIncluding topical antibiotic monotherapy; cpotentially including topical antibiotic component; dmodelled as general practitioner visits without active treatment. Costs were expressed in 2019 prices. Discounting of costs and benefits was not needed as the time horizon of the analysis was 1 year. The analysis included intervention costs for people who completed a course of treatment and those who discontinued early and the costs of average acne care (AAC), which comprises a mixture of care currently received by people with acne in the NHS, which may include GP consultations, specialist dermatologist care, drug treatment or no treatment. AAC represented NHS care provided to people with acne following early treatment discontinuation, completion of a course of physical treatment, inadequate improvement or relapse. Costs were estimated by combining resource use with respective unit costs. Intervention resource use was based on relevant descriptions from RCTs included in the source NMA, 5 modified to reflect optimal routine practice in the UK. This incorporated (as relevant) the drug dosage and optimal duration, time of the HCP (GP and/or specialist care), laboratory testing for people receiving oral isotretinoin and any equipment used. Resource use related to AAC was obtained from UK primary care consultation and prescription data, 13 supplemented with expert opinion and further assumptions, particularly regarding specialist care received by a proportion of people with acne. Unit costs were obtained from national sources 14 , 15 , 16 , 17 and other published literature. 18 Intervention costs are summarized in Table 3, with full details on methods used for their estimation reported in Supplementary Data S6. Details of the estimation of the AAC cost are shown in Supplementary Data S7. Intervention costs of treatments for mild‐to‐moderate and moderate‐to‐severe acne following a full course (acute and maintenance treatment) and early discontinuation (2019 prices). BPO, benzoyl peroxide; F, costs for females; M, costs for males; Maint, maintenance; NA, not applicable. aCosts included drug acquisition, healthcare professional time, laboratory testing for oral isotretinoin, procedure costs for photochemical and photodynamic therapies; no costs of contraception included for oral or topical retinoids. bIncluding topical antibiotic monotherapy; cpotentially including topical antibiotic component; dmodelled as general practitioner visits without active treatment. Costs were expressed in 2019 prices. Discounting of costs and benefits was not needed as the time horizon of the analysis was 1 year. [SUBTITLE] Statistical analysis [SUBSECTION] To account for the uncertainty around input parameter point estimates, a probabilistic analysis was undertaken, with input parameters being assigned probability distributions. 19 Subsequently, 10 000 model iterations were performed, each drawing random values out of the distributions fitted onto the model input parameters. The mean costs and QALYs for each treatment were calculated by averaging across the 10 000 iterations. The net monetary benefit (NMB) for each intervention was estimated for each iteration and averaged across the 10 000 iterations, determined by the formula: NMB=E×λ–C, where E and C are the effects (QALYs) and costs of each intervention respectively, and λ represents the willingness to pay (WTP) per QALY, set at the NICE lower cost‐effectiveness threshold of £20 000 per QALY. 20 The intervention with the highest NMB is the most cost‐effective. 21 The mean ranking by cost‐effectiveness (out of 10 000 iterations) is also reported for each intervention, with a rank of 1 indicating highest cost‐effectiveness. Supplementary Data S8 reports the model input values with probability distributions and additional sensitivity analyses conducted to test the robustness of the results. To account for the uncertainty around input parameter point estimates, a probabilistic analysis was undertaken, with input parameters being assigned probability distributions. 19 Subsequently, 10 000 model iterations were performed, each drawing random values out of the distributions fitted onto the model input parameters. The mean costs and QALYs for each treatment were calculated by averaging across the 10 000 iterations. The net monetary benefit (NMB) for each intervention was estimated for each iteration and averaged across the 10 000 iterations, determined by the formula: NMB=E×λ–C, where E and C are the effects (QALYs) and costs of each intervention respectively, and λ represents the willingness to pay (WTP) per QALY, set at the NICE lower cost‐effectiveness threshold of £20 000 per QALY. 20 The intervention with the highest NMB is the most cost‐effective. 21 The mean ranking by cost‐effectiveness (out of 10 000 iterations) is also reported for each intervention, with a rank of 1 indicating highest cost‐effectiveness. Supplementary Data S8 reports the model input values with probability distributions and additional sensitivity analyses conducted to test the robustness of the results. [SUBTITLE] Model validation [SUBSECTION] The economic model was constructed following the guideline committee's expert advice. All inputs and model formulae were systematically checked. The model was tested for logical consistency. The results were discussed with the committee to confirm plausibility. The economic model was constructed following the guideline committee's expert advice. All inputs and model formulae were systematically checked. The model was tested for logical consistency. The results were discussed with the committee to confirm plausibility.
Results
Table 4 shows the results of the economic analysis for treatments for mild‐to‐moderate acne (same for both sexes) and for moderate‐to‐severe acne in females. Results for treatments for moderate‐to‐severe acne in males (Supplementary Table S1) differed from those for females only in the ranking of oral isotretinoin, which was higher for males and is attributable to its lower intervention cost due to less intensive monitoring compared with females. The results are characterized by uncertainty, reflected in the wide 95% CIs around mean rankings. Cost‐effectiveness results for treatments for mild‐to‐moderate and moderate‐to‐severe acne.a,b BPO, benzoyl peroxide; CI, confidence interval; Int, intervention; NMB, net monetary benefit; QALY, quality‐adjusted life year. aClasses are ordered by NMB (highest NMB indicates highest cost‐effectiveness); bNMB and ranking estimated using a cost‐effectiveness threshold (willingness to pay) of £20 000/QALY; c n, number of observations across randomized controlled trials included in the network meta‐analysis of efficacy that informed the economic analysis. The cost‐effectiveness plane (Fig. 2) depicts the mean incremental costs and QALYs of all treatments vs. treatment with placebo (placed at the origin) in each analysis, ordered by magnitude of clinical benefits (QALYs). The cost‐effectiveness acceptability frontier 21 (Fig. 3) shows the most cost‐effective treatments for a range of values of WTP for a QALY (between £0 and £40 000 per QALY) and the probability of each treatment being cost‐effective. Cost‐effectiveness planes. Results for 1000 people with acne vulgaris. In each graph, the points for each treatment show its incremental quality‐adjusted life years (QALYs) (horizontal axis) and costs (vertical axis) vs. treatment with placebo, which is placed at the origin. The slope of the dotted line indicates the National Institute for Health and Care Excellence lower cost‐effectiveness threshold of £20 000/QALY. Moving towards the right of the horizontal axis, treatments result in more QALYs. For both acne severity levels, all treatments produce more QALYs compared with treatment with placebo. Moving towards the top of the vertical axis, treatments become more costly. For both acne severity levels, all treatments are more costly than treatment with placebo, with the exception of BPO in mild‐to‐moderate acne, and with the exception of BPO, topical clindamycin, combined topical tretinoin with clindamycin, oral lymecycline, and azelaic acid combined with oral lymecycline in moderate‐to‐severe acne. In all three graphs, treatments lie on the right side of the dotted line, suggesting that in all three analyses all assessed treatments are cost‐effective compared with treatment with placebo. Cost‐effectiveness acceptability frontier. Each graph shows the most cost‐effective treatment of each analysis, over a range of values of willingness to pay for a quality‐adjusted life year (QALY), which was varied between £0 and £40 000 per QALY (horizontal axis) and the probability that this treatment is the most cost‐effective of those assessed, reflecting the uncertainty in the results (vertical axis). The results were overall robust to the scenarios explored through deterministic sensitivity analysis (Supplementary Data S9). The relative cost‐effectiveness of physical therapies (chemical peels, photochemical therapies, PDT) and oral isotretinoin was reduced when the efficacy of the reference treatment (topical retinoid) was reduced or when the spread around the mean %CFB was increased. The latter was caused by ceiling effects, as people reached 100% improvement and could not improve further.
Conclusion
This economic analysis allowed estimation of the relative cost‐effectiveness of a range of topical pharmacological, oral pharmacological, physical and combined treatments for acne stratified by severity level. The results informed the NICE national guidance on the management of acne vulgaris. There remains a need for well‐conducted studies that examine the long‐term clinical efficacy and cost‐effectiveness of the full range of acne treatments.
[ "Introduction", "Population", "Interventions", "Economic model structure", "Clinical model inputs", "Utility data", "Resource use and cost data", "Statistical analysis", "Model validation", "What's already known about this topic?", "What's already known about this topic?", "What does this study add?", "Funding", "Ethics statement" ]
[ "Acne vulgaris is the eighth most common disease globally\n1\n and a common presentation to dermatologists.\n2\n Acne may have a detrimental physical, psychological and social impact.\n2\n, \n3\n, \n4\n A network meta‐analysis (NMA), assessing topical pharmacological, oral pharmacological, physical and combined treatments, found that topical treatment combinations, chemical peels and photochemical therapy (combined blue/red light) are effective treatments for mild‐to‐moderate acne, while topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and photodynamic therapy (PDT) are most effective for moderate‐to‐severe acne.\n5\n Effective treatments have varying treatment costs, including drug acquisition, time of the healthcare professional [HCP; general practitioner (GP) or specialist dermatologist] and special equipment. Evidence on the cost‐effectiveness of acne treatments is currently lacking. Thus, our study objective was to examine the cost‐effectiveness of effective treatments for mild‐to‐moderate and moderate‐to‐severe acne from the perspective of the National Health Service (NHS) in England, using decision–analytical economic modelling.\nThis economic analysis informed the development of National Institute for Health and Care Excellence (NICE) guidance for the management of acne in England.\n6\n NICE considers cost‐effectiveness when producing national guidance for health and social care services. The guideline was developed by a committee of clinical academics, HCPs and service users with expertise and experience in acne. The committee contributed to the development of the economic model by providing advice on the natural history of acne and its treatment patterns in England, and on model assumptions where evidence was lacking.", "The study population comprised people with mild‐to‐moderate and moderate‐to‐severe acne presenting to primary care services. Separate analyses were conducted for males and females with moderate‐to‐severe acne because (i) the intervention cost of oral isotretinoin is higher for females due to pregnancy tests and increased monitoring visits, and (ii) sex‐specific discontinuation data were available for moderate‐to‐severe acne.", "We included treatment classes showing evidence of efficacy vs. placebo [indicated by 95% credible intervals (CrI) around the effects not crossing the ‘no effect’ line] in the NMA that informed the NICE guideline (the source NMA).\n5\n We considered only treatment classes with ≥ 50 observations across randomized controlled trials (RCTs) included in the efficacy NMA, as this was deemed the minimum adequate evidence base to enable robust conclusions on clinical and cost‐effectiveness.\nOne intervention from each treatment class was selected as representative for costing purposes. All interventions were assumed to be delivered within the NHS. Selection of interventions assessed in the economic analysis (Table 1) was based on their availability and usage in the UK, practicalities of use (e.g. fixed topical treatment combinations were preferred to nonfixed formulations), the size of their evidence base and their risk of side effects relative to other interventions within the class.\nTypes of treatment, classes and interventions assessed in the economic analysis for each level of acne severity.\nBPO, benzoyl peroxide. aApplied and monitored by health professionals; bmodelled as general practitioner visits without active treatment; cform of phototherapy that involves a source of light (e.g. red light, blue light, broad‐spectrum light, daylight) and a photosensitizing chemical (e.g. 5‐aminolaevulinic acid or methyl aminolaevulinate).", "A decision tree was constructed to estimate total NHS costs and quality‐adjusted life years (QALYs) for each treatment over 1 year (Fig. 1). This time horizon, determined by the available follow‐up data, was deemed adequate to capture longer‐term outcomes and costs of acne treatment without significant extrapolation. Patients were modelled as having excellent, good, moderate or no perceived improvement following treatment (Supplementary Data S1 provides full details of the model).\nSchematic diagram of the economic model structure for people with mild‐to‐moderate and people with moderate‐to‐severe acne vulgaris. Chemical peels were assessed only in people with mild‐to‐moderate acne. Oral isotretinoin was assessed only in people with moderate‐to‐severe acne.", "Relative effects on efficacy [percentage change in total acne lesion count from baseline (%CFB)], discontinuation due to any reason and discontinuation due to side effects were obtained from the source NMA\n5\n (Supplementary Data S2). For mild‐to‐moderate acne, the economic analysis considered only treatments with evidence of efficacy in a bias‐adjusted NMA, owing to indication of small study bias.\n5\n\n\nTo obtain absolute effects for each treatment and outcome, we combined NMA relative effects with absolute effects of a reference treatment. We selected adapalene (topical retinoid) as the reference treatment based on data availability. Absolute effects for adapalene were estimated from large RCTs included in the source NMA,\n5\n and discontinuation data from a small non‐UK observational study\n7\n (Supplementary Data S3).\nPatients' perception of their acne improvement may differ from the clinical measurement of improvement as expressed by %CFB. The relationship between the two (Table 2) was determined using published trial evidence.\n8\n The same evidence\n8\n was used to determine the distribution around the mean %CFB at treatment endpoint, in order to estimate the proportion of people with excellent, good, moderate and no improvement for each treatment (Supplementary Data S4). Owing to limited and heterogeneous evidence, the risk of relapse following excellent, good and moderate improvement was based on expert opinion.\nRelationship between percentage change in total acne lesion count from baseline, perceived acne improvement and utility value.\n%CFB, percentage change in total lesion count from baseline; NA, not applicable. aSupplementary Data S5.", "Utility values express people's preferences regarding health‐related quality of life (HRQoL) on a scale from 0 (death) to 1 (perfect health) and are necessary for estimating QALYs. The utility values used in the economic model (Table 2) were determined using utility data from two studies\n9\n, \n10\n identified from a systematic search, a published mapping algorithm,\n11\n utility values of the UK general young adult population\n12\n and further assumptions (Supplementary Data S5).", "The analysis included intervention costs for people who completed a course of treatment and those who discontinued early and the costs of average acne care (AAC), which comprises a mixture of care currently received by people with acne in the NHS, which may include GP consultations, specialist dermatologist care, drug treatment or no treatment. AAC represented NHS care provided to people with acne following early treatment discontinuation, completion of a course of physical treatment, inadequate improvement or relapse. Costs were estimated by combining resource use with respective unit costs.\nIntervention resource use was based on relevant descriptions from RCTs included in the source NMA,\n5\n modified to reflect optimal routine practice in the UK. This incorporated (as relevant) the drug dosage and optimal duration, time of the HCP (GP and/or specialist care), laboratory testing for people receiving oral isotretinoin and any equipment used. Resource use related to AAC was obtained from UK primary care consultation and prescription data,\n13\n supplemented with expert opinion and further assumptions, particularly regarding specialist care received by a proportion of people with acne. Unit costs were obtained from national sources\n14\n, \n15\n, \n16\n, \n17\n and other published literature.\n18\n Intervention costs are summarized in Table 3, with full details on methods used for their estimation reported in Supplementary Data S6. Details of the estimation of the AAC cost are shown in Supplementary Data S7.\nIntervention costs of treatments for mild‐to‐moderate and moderate‐to‐severe acne following a full course (acute and maintenance treatment) and early discontinuation (2019 prices).\nBPO, benzoyl peroxide; F, costs for females; M, costs for males; Maint, maintenance; NA, not applicable.\n\naCosts included drug acquisition, healthcare professional time, laboratory testing for oral isotretinoin, procedure costs for photochemical and photodynamic therapies; no costs of contraception included for oral or topical retinoids. bIncluding topical antibiotic monotherapy; cpotentially including topical antibiotic component; dmodelled as general practitioner visits without active treatment.\nCosts were expressed in 2019 prices. Discounting of costs and benefits was not needed as the time horizon of the analysis was 1 year.", "To account for the uncertainty around input parameter point estimates, a probabilistic analysis was undertaken, with input parameters being assigned probability distributions.\n19\n Subsequently, 10 000 model iterations were performed, each drawing random values out of the distributions fitted onto the model input parameters. The mean costs and QALYs for each treatment were calculated by averaging across the 10 000 iterations. The net monetary benefit (NMB) for each intervention was estimated for each iteration and averaged across the 10 000 iterations, determined by the formula:\nNMB=E×λ–C,\nwhere E and C are the effects (QALYs) and costs of each intervention respectively, and λ represents the willingness to pay (WTP) per QALY, set at the NICE lower cost‐effectiveness threshold of £20 000 per QALY.\n20\n The intervention with the highest NMB is the most cost‐effective.\n21\n The mean ranking by cost‐effectiveness (out of 10 000 iterations) is also reported for each intervention, with a rank of 1 indicating highest cost‐effectiveness.\nSupplementary Data S8 reports the model input values with probability distributions and additional sensitivity analyses conducted to test the robustness of the results.", "The economic model was constructed following the guideline committee's expert advice. All inputs and model formulae were systematically checked. The model was tested for logical consistency. The results were discussed with the committee to confirm plausibility.", "\nWhat's already known about this topic?\nAcne vulgaris is the eighth most common disease globally.In a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.Evidence on the cost‐effectiveness of acne vulgaris treatments is lacking.Identifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.\nWhat does this study add?\nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.For mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).For moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.These findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.\n\n\n[SUBTITLE] What's already known about this topic? [SUBSECTION] \nAcne vulgaris is the eighth most common disease globally.In a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.Evidence on the cost‐effectiveness of acne vulgaris treatments is lacking.Identifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.\n\nAcne vulgaris is the eighth most common disease globally.\nIn a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.\nEvidence on the cost‐effectiveness of acne vulgaris treatments is lacking.\nIdentifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.\n\nAcne vulgaris is the eighth most common disease globally.In a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.Evidence on the cost‐effectiveness of acne vulgaris treatments is lacking.Identifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.\n\nAcne vulgaris is the eighth most common disease globally.\nIn a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.\nEvidence on the cost‐effectiveness of acne vulgaris treatments is lacking.\nIdentifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.\n[SUBTITLE] What does this study add? [SUBSECTION] \nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.For mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).For moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.These findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.\n\nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.\nFor mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).\nFor moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.\nThese findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.\n\nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.For mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).For moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.These findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.\n\nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.\nFor mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).\nFor moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.\nThese findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.", "\nAcne vulgaris is the eighth most common disease globally.In a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.Evidence on the cost‐effectiveness of acne vulgaris treatments is lacking.Identifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.\n\nAcne vulgaris is the eighth most common disease globally.\nIn a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.\nEvidence on the cost‐effectiveness of acne vulgaris treatments is lacking.\nIdentifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.", "\nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.For mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).For moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.These findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.\n\nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.\nFor mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).\nFor moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.\nThese findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.", "This work was undertaken by the National Guideline Alliance (NGA) at the Royal College of Obstetricians and Gynaecologists (RCOG), with support from the National Institute for Health and Care Excellence (NICE) Guidelines Technical Support Unit (TSU), University of Bristol, which is funded by the NICE Centre for Guidelines. NGA has received funding from NICE to develop clinical, public health and social care guidelines. For the development of this guideline, NICE worked with the British Association of Dermatologists. The views expressed in this publication are those of the authors and not necessarily those of RCOG, NGA or NICE. The funder of the study had no further role in the study design, data collection, data analysis, data interpretation, or writing of the report. All authors had full access to all the data in the study and had final responsibility for the decision to submit for publication. National Institute for Health and Care Excellence (2021) Acne Vulgaris: management. Available from: https://nice.org.uk/guidance/ng198.", "Ethics approval and informed consent not applicable." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Introduction", "Methods", "Population", "Interventions", "Economic model structure", "Clinical model inputs", "Utility data", "Resource use and cost data", "Statistical analysis", "Model validation", "Results", "Discussion", "Conclusion", "What's already known about this topic?", "What's already known about this topic?", "What does this study add?", "Conflict of interest", "Funding", "Ethics statement", "Supporting information" ]
[ "Acne vulgaris is the eighth most common disease globally\n1\n and a common presentation to dermatologists.\n2\n Acne may have a detrimental physical, psychological and social impact.\n2\n, \n3\n, \n4\n A network meta‐analysis (NMA), assessing topical pharmacological, oral pharmacological, physical and combined treatments, found that topical treatment combinations, chemical peels and photochemical therapy (combined blue/red light) are effective treatments for mild‐to‐moderate acne, while topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and photodynamic therapy (PDT) are most effective for moderate‐to‐severe acne.\n5\n Effective treatments have varying treatment costs, including drug acquisition, time of the healthcare professional [HCP; general practitioner (GP) or specialist dermatologist] and special equipment. Evidence on the cost‐effectiveness of acne treatments is currently lacking. Thus, our study objective was to examine the cost‐effectiveness of effective treatments for mild‐to‐moderate and moderate‐to‐severe acne from the perspective of the National Health Service (NHS) in England, using decision–analytical economic modelling.\nThis economic analysis informed the development of National Institute for Health and Care Excellence (NICE) guidance for the management of acne in England.\n6\n NICE considers cost‐effectiveness when producing national guidance for health and social care services. The guideline was developed by a committee of clinical academics, HCPs and service users with expertise and experience in acne. The committee contributed to the development of the economic model by providing advice on the natural history of acne and its treatment patterns in England, and on model assumptions where evidence was lacking.", "[SUBTITLE] Population [SUBSECTION] The study population comprised people with mild‐to‐moderate and moderate‐to‐severe acne presenting to primary care services. Separate analyses were conducted for males and females with moderate‐to‐severe acne because (i) the intervention cost of oral isotretinoin is higher for females due to pregnancy tests and increased monitoring visits, and (ii) sex‐specific discontinuation data were available for moderate‐to‐severe acne.\nThe study population comprised people with mild‐to‐moderate and moderate‐to‐severe acne presenting to primary care services. Separate analyses were conducted for males and females with moderate‐to‐severe acne because (i) the intervention cost of oral isotretinoin is higher for females due to pregnancy tests and increased monitoring visits, and (ii) sex‐specific discontinuation data were available for moderate‐to‐severe acne.\n[SUBTITLE] Interventions [SUBSECTION] We included treatment classes showing evidence of efficacy vs. placebo [indicated by 95% credible intervals (CrI) around the effects not crossing the ‘no effect’ line] in the NMA that informed the NICE guideline (the source NMA).\n5\n We considered only treatment classes with ≥ 50 observations across randomized controlled trials (RCTs) included in the efficacy NMA, as this was deemed the minimum adequate evidence base to enable robust conclusions on clinical and cost‐effectiveness.\nOne intervention from each treatment class was selected as representative for costing purposes. All interventions were assumed to be delivered within the NHS. Selection of interventions assessed in the economic analysis (Table 1) was based on their availability and usage in the UK, practicalities of use (e.g. fixed topical treatment combinations were preferred to nonfixed formulations), the size of their evidence base and their risk of side effects relative to other interventions within the class.\nTypes of treatment, classes and interventions assessed in the economic analysis for each level of acne severity.\nBPO, benzoyl peroxide. aApplied and monitored by health professionals; bmodelled as general practitioner visits without active treatment; cform of phototherapy that involves a source of light (e.g. red light, blue light, broad‐spectrum light, daylight) and a photosensitizing chemical (e.g. 5‐aminolaevulinic acid or methyl aminolaevulinate).\nWe included treatment classes showing evidence of efficacy vs. placebo [indicated by 95% credible intervals (CrI) around the effects not crossing the ‘no effect’ line] in the NMA that informed the NICE guideline (the source NMA).\n5\n We considered only treatment classes with ≥ 50 observations across randomized controlled trials (RCTs) included in the efficacy NMA, as this was deemed the minimum adequate evidence base to enable robust conclusions on clinical and cost‐effectiveness.\nOne intervention from each treatment class was selected as representative for costing purposes. All interventions were assumed to be delivered within the NHS. Selection of interventions assessed in the economic analysis (Table 1) was based on their availability and usage in the UK, practicalities of use (e.g. fixed topical treatment combinations were preferred to nonfixed formulations), the size of their evidence base and their risk of side effects relative to other interventions within the class.\nTypes of treatment, classes and interventions assessed in the economic analysis for each level of acne severity.\nBPO, benzoyl peroxide. aApplied and monitored by health professionals; bmodelled as general practitioner visits without active treatment; cform of phototherapy that involves a source of light (e.g. red light, blue light, broad‐spectrum light, daylight) and a photosensitizing chemical (e.g. 5‐aminolaevulinic acid or methyl aminolaevulinate).\n[SUBTITLE] Economic model structure [SUBSECTION] A decision tree was constructed to estimate total NHS costs and quality‐adjusted life years (QALYs) for each treatment over 1 year (Fig. 1). This time horizon, determined by the available follow‐up data, was deemed adequate to capture longer‐term outcomes and costs of acne treatment without significant extrapolation. Patients were modelled as having excellent, good, moderate or no perceived improvement following treatment (Supplementary Data S1 provides full details of the model).\nSchematic diagram of the economic model structure for people with mild‐to‐moderate and people with moderate‐to‐severe acne vulgaris. Chemical peels were assessed only in people with mild‐to‐moderate acne. Oral isotretinoin was assessed only in people with moderate‐to‐severe acne.\nA decision tree was constructed to estimate total NHS costs and quality‐adjusted life years (QALYs) for each treatment over 1 year (Fig. 1). This time horizon, determined by the available follow‐up data, was deemed adequate to capture longer‐term outcomes and costs of acne treatment without significant extrapolation. Patients were modelled as having excellent, good, moderate or no perceived improvement following treatment (Supplementary Data S1 provides full details of the model).\nSchematic diagram of the economic model structure for people with mild‐to‐moderate and people with moderate‐to‐severe acne vulgaris. Chemical peels were assessed only in people with mild‐to‐moderate acne. Oral isotretinoin was assessed only in people with moderate‐to‐severe acne.\n[SUBTITLE] Clinical model inputs [SUBSECTION] Relative effects on efficacy [percentage change in total acne lesion count from baseline (%CFB)], discontinuation due to any reason and discontinuation due to side effects were obtained from the source NMA\n5\n (Supplementary Data S2). For mild‐to‐moderate acne, the economic analysis considered only treatments with evidence of efficacy in a bias‐adjusted NMA, owing to indication of small study bias.\n5\n\n\nTo obtain absolute effects for each treatment and outcome, we combined NMA relative effects with absolute effects of a reference treatment. We selected adapalene (topical retinoid) as the reference treatment based on data availability. Absolute effects for adapalene were estimated from large RCTs included in the source NMA,\n5\n and discontinuation data from a small non‐UK observational study\n7\n (Supplementary Data S3).\nPatients' perception of their acne improvement may differ from the clinical measurement of improvement as expressed by %CFB. The relationship between the two (Table 2) was determined using published trial evidence.\n8\n The same evidence\n8\n was used to determine the distribution around the mean %CFB at treatment endpoint, in order to estimate the proportion of people with excellent, good, moderate and no improvement for each treatment (Supplementary Data S4). Owing to limited and heterogeneous evidence, the risk of relapse following excellent, good and moderate improvement was based on expert opinion.\nRelationship between percentage change in total acne lesion count from baseline, perceived acne improvement and utility value.\n%CFB, percentage change in total lesion count from baseline; NA, not applicable. aSupplementary Data S5.\nRelative effects on efficacy [percentage change in total acne lesion count from baseline (%CFB)], discontinuation due to any reason and discontinuation due to side effects were obtained from the source NMA\n5\n (Supplementary Data S2). For mild‐to‐moderate acne, the economic analysis considered only treatments with evidence of efficacy in a bias‐adjusted NMA, owing to indication of small study bias.\n5\n\n\nTo obtain absolute effects for each treatment and outcome, we combined NMA relative effects with absolute effects of a reference treatment. We selected adapalene (topical retinoid) as the reference treatment based on data availability. Absolute effects for adapalene were estimated from large RCTs included in the source NMA,\n5\n and discontinuation data from a small non‐UK observational study\n7\n (Supplementary Data S3).\nPatients' perception of their acne improvement may differ from the clinical measurement of improvement as expressed by %CFB. The relationship between the two (Table 2) was determined using published trial evidence.\n8\n The same evidence\n8\n was used to determine the distribution around the mean %CFB at treatment endpoint, in order to estimate the proportion of people with excellent, good, moderate and no improvement for each treatment (Supplementary Data S4). Owing to limited and heterogeneous evidence, the risk of relapse following excellent, good and moderate improvement was based on expert opinion.\nRelationship between percentage change in total acne lesion count from baseline, perceived acne improvement and utility value.\n%CFB, percentage change in total lesion count from baseline; NA, not applicable. aSupplementary Data S5.\n[SUBTITLE] Utility data [SUBSECTION] Utility values express people's preferences regarding health‐related quality of life (HRQoL) on a scale from 0 (death) to 1 (perfect health) and are necessary for estimating QALYs. The utility values used in the economic model (Table 2) were determined using utility data from two studies\n9\n, \n10\n identified from a systematic search, a published mapping algorithm,\n11\n utility values of the UK general young adult population\n12\n and further assumptions (Supplementary Data S5).\nUtility values express people's preferences regarding health‐related quality of life (HRQoL) on a scale from 0 (death) to 1 (perfect health) and are necessary for estimating QALYs. The utility values used in the economic model (Table 2) were determined using utility data from two studies\n9\n, \n10\n identified from a systematic search, a published mapping algorithm,\n11\n utility values of the UK general young adult population\n12\n and further assumptions (Supplementary Data S5).\n[SUBTITLE] Resource use and cost data [SUBSECTION] The analysis included intervention costs for people who completed a course of treatment and those who discontinued early and the costs of average acne care (AAC), which comprises a mixture of care currently received by people with acne in the NHS, which may include GP consultations, specialist dermatologist care, drug treatment or no treatment. AAC represented NHS care provided to people with acne following early treatment discontinuation, completion of a course of physical treatment, inadequate improvement or relapse. Costs were estimated by combining resource use with respective unit costs.\nIntervention resource use was based on relevant descriptions from RCTs included in the source NMA,\n5\n modified to reflect optimal routine practice in the UK. This incorporated (as relevant) the drug dosage and optimal duration, time of the HCP (GP and/or specialist care), laboratory testing for people receiving oral isotretinoin and any equipment used. Resource use related to AAC was obtained from UK primary care consultation and prescription data,\n13\n supplemented with expert opinion and further assumptions, particularly regarding specialist care received by a proportion of people with acne. Unit costs were obtained from national sources\n14\n, \n15\n, \n16\n, \n17\n and other published literature.\n18\n Intervention costs are summarized in Table 3, with full details on methods used for their estimation reported in Supplementary Data S6. Details of the estimation of the AAC cost are shown in Supplementary Data S7.\nIntervention costs of treatments for mild‐to‐moderate and moderate‐to‐severe acne following a full course (acute and maintenance treatment) and early discontinuation (2019 prices).\nBPO, benzoyl peroxide; F, costs for females; M, costs for males; Maint, maintenance; NA, not applicable.\n\naCosts included drug acquisition, healthcare professional time, laboratory testing for oral isotretinoin, procedure costs for photochemical and photodynamic therapies; no costs of contraception included for oral or topical retinoids. bIncluding topical antibiotic monotherapy; cpotentially including topical antibiotic component; dmodelled as general practitioner visits without active treatment.\nCosts were expressed in 2019 prices. Discounting of costs and benefits was not needed as the time horizon of the analysis was 1 year.\nThe analysis included intervention costs for people who completed a course of treatment and those who discontinued early and the costs of average acne care (AAC), which comprises a mixture of care currently received by people with acne in the NHS, which may include GP consultations, specialist dermatologist care, drug treatment or no treatment. AAC represented NHS care provided to people with acne following early treatment discontinuation, completion of a course of physical treatment, inadequate improvement or relapse. Costs were estimated by combining resource use with respective unit costs.\nIntervention resource use was based on relevant descriptions from RCTs included in the source NMA,\n5\n modified to reflect optimal routine practice in the UK. This incorporated (as relevant) the drug dosage and optimal duration, time of the HCP (GP and/or specialist care), laboratory testing for people receiving oral isotretinoin and any equipment used. Resource use related to AAC was obtained from UK primary care consultation and prescription data,\n13\n supplemented with expert opinion and further assumptions, particularly regarding specialist care received by a proportion of people with acne. Unit costs were obtained from national sources\n14\n, \n15\n, \n16\n, \n17\n and other published literature.\n18\n Intervention costs are summarized in Table 3, with full details on methods used for their estimation reported in Supplementary Data S6. Details of the estimation of the AAC cost are shown in Supplementary Data S7.\nIntervention costs of treatments for mild‐to‐moderate and moderate‐to‐severe acne following a full course (acute and maintenance treatment) and early discontinuation (2019 prices).\nBPO, benzoyl peroxide; F, costs for females; M, costs for males; Maint, maintenance; NA, not applicable.\n\naCosts included drug acquisition, healthcare professional time, laboratory testing for oral isotretinoin, procedure costs for photochemical and photodynamic therapies; no costs of contraception included for oral or topical retinoids. bIncluding topical antibiotic monotherapy; cpotentially including topical antibiotic component; dmodelled as general practitioner visits without active treatment.\nCosts were expressed in 2019 prices. Discounting of costs and benefits was not needed as the time horizon of the analysis was 1 year.\n[SUBTITLE] Statistical analysis [SUBSECTION] To account for the uncertainty around input parameter point estimates, a probabilistic analysis was undertaken, with input parameters being assigned probability distributions.\n19\n Subsequently, 10 000 model iterations were performed, each drawing random values out of the distributions fitted onto the model input parameters. The mean costs and QALYs for each treatment were calculated by averaging across the 10 000 iterations. The net monetary benefit (NMB) for each intervention was estimated for each iteration and averaged across the 10 000 iterations, determined by the formula:\nNMB=E×λ–C,\nwhere E and C are the effects (QALYs) and costs of each intervention respectively, and λ represents the willingness to pay (WTP) per QALY, set at the NICE lower cost‐effectiveness threshold of £20 000 per QALY.\n20\n The intervention with the highest NMB is the most cost‐effective.\n21\n The mean ranking by cost‐effectiveness (out of 10 000 iterations) is also reported for each intervention, with a rank of 1 indicating highest cost‐effectiveness.\nSupplementary Data S8 reports the model input values with probability distributions and additional sensitivity analyses conducted to test the robustness of the results.\nTo account for the uncertainty around input parameter point estimates, a probabilistic analysis was undertaken, with input parameters being assigned probability distributions.\n19\n Subsequently, 10 000 model iterations were performed, each drawing random values out of the distributions fitted onto the model input parameters. The mean costs and QALYs for each treatment were calculated by averaging across the 10 000 iterations. The net monetary benefit (NMB) for each intervention was estimated for each iteration and averaged across the 10 000 iterations, determined by the formula:\nNMB=E×λ–C,\nwhere E and C are the effects (QALYs) and costs of each intervention respectively, and λ represents the willingness to pay (WTP) per QALY, set at the NICE lower cost‐effectiveness threshold of £20 000 per QALY.\n20\n The intervention with the highest NMB is the most cost‐effective.\n21\n The mean ranking by cost‐effectiveness (out of 10 000 iterations) is also reported for each intervention, with a rank of 1 indicating highest cost‐effectiveness.\nSupplementary Data S8 reports the model input values with probability distributions and additional sensitivity analyses conducted to test the robustness of the results.\n[SUBTITLE] Model validation [SUBSECTION] The economic model was constructed following the guideline committee's expert advice. All inputs and model formulae were systematically checked. The model was tested for logical consistency. The results were discussed with the committee to confirm plausibility.\nThe economic model was constructed following the guideline committee's expert advice. All inputs and model formulae were systematically checked. The model was tested for logical consistency. The results were discussed with the committee to confirm plausibility.", "The study population comprised people with mild‐to‐moderate and moderate‐to‐severe acne presenting to primary care services. Separate analyses were conducted for males and females with moderate‐to‐severe acne because (i) the intervention cost of oral isotretinoin is higher for females due to pregnancy tests and increased monitoring visits, and (ii) sex‐specific discontinuation data were available for moderate‐to‐severe acne.", "We included treatment classes showing evidence of efficacy vs. placebo [indicated by 95% credible intervals (CrI) around the effects not crossing the ‘no effect’ line] in the NMA that informed the NICE guideline (the source NMA).\n5\n We considered only treatment classes with ≥ 50 observations across randomized controlled trials (RCTs) included in the efficacy NMA, as this was deemed the minimum adequate evidence base to enable robust conclusions on clinical and cost‐effectiveness.\nOne intervention from each treatment class was selected as representative for costing purposes. All interventions were assumed to be delivered within the NHS. Selection of interventions assessed in the economic analysis (Table 1) was based on their availability and usage in the UK, practicalities of use (e.g. fixed topical treatment combinations were preferred to nonfixed formulations), the size of their evidence base and their risk of side effects relative to other interventions within the class.\nTypes of treatment, classes and interventions assessed in the economic analysis for each level of acne severity.\nBPO, benzoyl peroxide. aApplied and monitored by health professionals; bmodelled as general practitioner visits without active treatment; cform of phototherapy that involves a source of light (e.g. red light, blue light, broad‐spectrum light, daylight) and a photosensitizing chemical (e.g. 5‐aminolaevulinic acid or methyl aminolaevulinate).", "A decision tree was constructed to estimate total NHS costs and quality‐adjusted life years (QALYs) for each treatment over 1 year (Fig. 1). This time horizon, determined by the available follow‐up data, was deemed adequate to capture longer‐term outcomes and costs of acne treatment without significant extrapolation. Patients were modelled as having excellent, good, moderate or no perceived improvement following treatment (Supplementary Data S1 provides full details of the model).\nSchematic diagram of the economic model structure for people with mild‐to‐moderate and people with moderate‐to‐severe acne vulgaris. Chemical peels were assessed only in people with mild‐to‐moderate acne. Oral isotretinoin was assessed only in people with moderate‐to‐severe acne.", "Relative effects on efficacy [percentage change in total acne lesion count from baseline (%CFB)], discontinuation due to any reason and discontinuation due to side effects were obtained from the source NMA\n5\n (Supplementary Data S2). For mild‐to‐moderate acne, the economic analysis considered only treatments with evidence of efficacy in a bias‐adjusted NMA, owing to indication of small study bias.\n5\n\n\nTo obtain absolute effects for each treatment and outcome, we combined NMA relative effects with absolute effects of a reference treatment. We selected adapalene (topical retinoid) as the reference treatment based on data availability. Absolute effects for adapalene were estimated from large RCTs included in the source NMA,\n5\n and discontinuation data from a small non‐UK observational study\n7\n (Supplementary Data S3).\nPatients' perception of their acne improvement may differ from the clinical measurement of improvement as expressed by %CFB. The relationship between the two (Table 2) was determined using published trial evidence.\n8\n The same evidence\n8\n was used to determine the distribution around the mean %CFB at treatment endpoint, in order to estimate the proportion of people with excellent, good, moderate and no improvement for each treatment (Supplementary Data S4). Owing to limited and heterogeneous evidence, the risk of relapse following excellent, good and moderate improvement was based on expert opinion.\nRelationship between percentage change in total acne lesion count from baseline, perceived acne improvement and utility value.\n%CFB, percentage change in total lesion count from baseline; NA, not applicable. aSupplementary Data S5.", "Utility values express people's preferences regarding health‐related quality of life (HRQoL) on a scale from 0 (death) to 1 (perfect health) and are necessary for estimating QALYs. The utility values used in the economic model (Table 2) were determined using utility data from two studies\n9\n, \n10\n identified from a systematic search, a published mapping algorithm,\n11\n utility values of the UK general young adult population\n12\n and further assumptions (Supplementary Data S5).", "The analysis included intervention costs for people who completed a course of treatment and those who discontinued early and the costs of average acne care (AAC), which comprises a mixture of care currently received by people with acne in the NHS, which may include GP consultations, specialist dermatologist care, drug treatment or no treatment. AAC represented NHS care provided to people with acne following early treatment discontinuation, completion of a course of physical treatment, inadequate improvement or relapse. Costs were estimated by combining resource use with respective unit costs.\nIntervention resource use was based on relevant descriptions from RCTs included in the source NMA,\n5\n modified to reflect optimal routine practice in the UK. This incorporated (as relevant) the drug dosage and optimal duration, time of the HCP (GP and/or specialist care), laboratory testing for people receiving oral isotretinoin and any equipment used. Resource use related to AAC was obtained from UK primary care consultation and prescription data,\n13\n supplemented with expert opinion and further assumptions, particularly regarding specialist care received by a proportion of people with acne. Unit costs were obtained from national sources\n14\n, \n15\n, \n16\n, \n17\n and other published literature.\n18\n Intervention costs are summarized in Table 3, with full details on methods used for their estimation reported in Supplementary Data S6. Details of the estimation of the AAC cost are shown in Supplementary Data S7.\nIntervention costs of treatments for mild‐to‐moderate and moderate‐to‐severe acne following a full course (acute and maintenance treatment) and early discontinuation (2019 prices).\nBPO, benzoyl peroxide; F, costs for females; M, costs for males; Maint, maintenance; NA, not applicable.\n\naCosts included drug acquisition, healthcare professional time, laboratory testing for oral isotretinoin, procedure costs for photochemical and photodynamic therapies; no costs of contraception included for oral or topical retinoids. bIncluding topical antibiotic monotherapy; cpotentially including topical antibiotic component; dmodelled as general practitioner visits without active treatment.\nCosts were expressed in 2019 prices. Discounting of costs and benefits was not needed as the time horizon of the analysis was 1 year.", "To account for the uncertainty around input parameter point estimates, a probabilistic analysis was undertaken, with input parameters being assigned probability distributions.\n19\n Subsequently, 10 000 model iterations were performed, each drawing random values out of the distributions fitted onto the model input parameters. The mean costs and QALYs for each treatment were calculated by averaging across the 10 000 iterations. The net monetary benefit (NMB) for each intervention was estimated for each iteration and averaged across the 10 000 iterations, determined by the formula:\nNMB=E×λ–C,\nwhere E and C are the effects (QALYs) and costs of each intervention respectively, and λ represents the willingness to pay (WTP) per QALY, set at the NICE lower cost‐effectiveness threshold of £20 000 per QALY.\n20\n The intervention with the highest NMB is the most cost‐effective.\n21\n The mean ranking by cost‐effectiveness (out of 10 000 iterations) is also reported for each intervention, with a rank of 1 indicating highest cost‐effectiveness.\nSupplementary Data S8 reports the model input values with probability distributions and additional sensitivity analyses conducted to test the robustness of the results.", "The economic model was constructed following the guideline committee's expert advice. All inputs and model formulae were systematically checked. The model was tested for logical consistency. The results were discussed with the committee to confirm plausibility.", "Table 4 shows the results of the economic analysis for treatments for mild‐to‐moderate acne (same for both sexes) and for moderate‐to‐severe acne in females. Results for treatments for moderate‐to‐severe acne in males (Supplementary Table S1) differed from those for females only in the ranking of oral isotretinoin, which was higher for males and is attributable to its lower intervention cost due to less intensive monitoring compared with females. The results are characterized by uncertainty, reflected in the wide 95% CIs around mean rankings.\nCost‐effectiveness results for treatments for mild‐to‐moderate and moderate‐to‐severe acne.a,b\n\nBPO, benzoyl peroxide; CI, confidence interval; Int, intervention; NMB, net monetary benefit; QALY, quality‐adjusted life year. aClasses are ordered by NMB (highest NMB indicates highest cost‐effectiveness); bNMB and ranking estimated using a cost‐effectiveness threshold (willingness to pay) of £20 000/QALY; c\nn, number of observations across randomized controlled trials included in the network meta‐analysis of efficacy that informed the economic analysis.\nThe cost‐effectiveness plane (Fig. 2) depicts the mean incremental costs and QALYs of all treatments vs. treatment with placebo (placed at the origin) in each analysis, ordered by magnitude of clinical benefits (QALYs). The cost‐effectiveness acceptability frontier\n21\n (Fig. 3) shows the most cost‐effective treatments for a range of values of WTP for a QALY (between £0 and £40 000 per QALY) and the probability of each treatment being cost‐effective.\nCost‐effectiveness planes. Results for 1000 people with acne vulgaris. In each graph, the points for each treatment show its incremental quality‐adjusted life years (QALYs) (horizontal axis) and costs (vertical axis) vs. treatment with placebo, which is placed at the origin. The slope of the dotted line indicates the National Institute for Health and Care Excellence lower cost‐effectiveness threshold of £20 000/QALY. Moving towards the right of the horizontal axis, treatments result in more QALYs. For both acne severity levels, all treatments produce more QALYs compared with treatment with placebo. Moving towards the top of the vertical axis, treatments become more costly. For both acne severity levels, all treatments are more costly than treatment with placebo, with the exception of BPO in mild‐to‐moderate acne, and with the exception of BPO, topical clindamycin, combined topical tretinoin with clindamycin, oral lymecycline, and azelaic acid combined with oral lymecycline in moderate‐to‐severe acne. In all three graphs, treatments lie on the right side of the dotted line, suggesting that in all three analyses all assessed treatments are cost‐effective compared with treatment with placebo.\nCost‐effectiveness acceptability frontier. Each graph shows the most cost‐effective treatment of each analysis, over a range of values of willingness to pay for a quality‐adjusted life year (QALY), which was varied between £0 and £40 000 per QALY (horizontal axis) and the probability that this treatment is the most cost‐effective of those assessed, reflecting the uncertainty in the results (vertical axis).\nThe results were overall robust to the scenarios explored through deterministic sensitivity analysis (Supplementary Data S9). The relative cost‐effectiveness of physical therapies (chemical peels, photochemical therapies, PDT) and oral isotretinoin was reduced when the efficacy of the reference treatment (topical retinoid) was reduced or when the spread around the mean %CFB was increased. The latter was caused by ceiling effects, as people reached 100% improvement and could not improve further.", "Our analysis explored the relative cost‐effectiveness of a wide range of topical pharmacological, oral pharmacological, physical and combined treatments for acne stratified by severity level. All treatments were more cost‐effective than treatment with placebo (modelled as GP visits without active treatment). For mild‐to‐moderate acne, the most cost‐effective treatments included topical treatment combinations and photochemical therapy (combined blue/red light). For moderate‐to‐severe acne, the most cost‐effective treatments included topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.\nTo our knowledge, this is the first analysis exploring the relative cost‐effectiveness of a wide range of treatment options for acne from a healthcare perspective, considering, in addition to drug acquisition costs, HCP resource use, including costs associated with inadequate response to treatment or relapse. Previous economic studies (identified through a systematic search conducted to inform the NICE guideline)\n6\n made limited comparisons of acne treatments, and the majority considered exclusively drug acquisition costs. A number of studies were simple cost analyses and most of them were characterized by important methodological limitations (Supplementary Data S10). Therefore, no robust conclusions can be drawn from the existing economic literature.\nIn our analysis, we used efficacy and discontinuation data derived from a large systematic review and NMA.\n5\n This approach combines direct (i.e. head‐to‐head comparisons) and indirect evidence (e.g. comparisons through a common comparator) and allows simultaneous comparisons across all options while preserving randomization.\n22\n, \n23\n There was some inconsistency between the direct and indirect evidence synthesized in the NMA, possibly reflecting heterogeneity in populations, treatments or study design across the included RCTs. However, NMA conclusions were robust to potential bias in the evidence. The RCTs included in the NMAs were of moderate to very low quality. The strengths and limitations of the source NMA and included RCTs should be considered when interpreting the cost‐effectiveness results.\nThe size of the evidence differed considerably across the examined treatments. Topical treatments had the largest evidence base on efficacy (several topical treatments had > 500 observations each), followed by oral treatments alone or combined with topical treatments, whereas the evidence for physical therapies (chemical peels, photochemical therapies, PDT) was based on < 300 observations for each treatment class.\nModel input parameters were obtained from sources of varying quality (ranging from RCTs through to large retrospective analyses to expert opinion), depending on data availability. The time horizon of the analysis (1 year) was considered adequate to capture longer‐term outcomes and costs associated with a course of acne treatment, but some long‐term effects, such as potential subsequent scarring and associated management costs were not captured because of insufficient evidence. Costs and utility decrements associated with side effects of treatment were not considered; however, we did incorporate the impact of intolerable side effects on HRQoL and costs. Antimicrobial resistance associated with antibiotic use was not considered. These omissions constitute limitations of our analysis.\nWe carried out probabilistic analyses to handle uncertainty around model parameters and deterministic sensitivity analyses to address gaps in the evidence. The results were characterized by some uncertainty; however, they were overall robust to scenarios tested through deterministic sensitivity analysis.\nOur analysis was conducted from the perspective of the NHS in England. The results may be generalizable to other settings with similar funding and structure of healthcare services and comparable care pathways for people with acne. Conclusions on cost‐effectiveness ultimately rely on the cost‐effectiveness threshold adopted, and this depends on the policymakers' willingness to pay for treatment benefits, which may vary across countries and health systems.\nThe results of this economic analysis, along with clinical evidence from the NMA\n5\n and other considerations, informed the NICE national guidance on the management of acne vulgaris.\n6\n These considerations included concerns about antibiotic resistance, current regulations regarding oral isotretinoin,\n24\n, \n25\n long‐term harms of chemical peel use outside specialist settings (e.g. risk for significant skin damage from inappropriate strength or type of peel) and limited availability and use of some treatments in NHS routine practice (e.g. chemical peels, photochemical therapies, PDT, topical antifungals). Doxycycline was considered a suitable alternative to lymecycline, as they have similar efficacy, AE profile and acquisition cost.\nFixed topical treatment combinations [adapalene with benzoyl peroxide (BPO); clindamycin with BPO; tretinoin with clindamycin] were recommended as first‐line treatments of mild‐to‐moderate acne. Fixed topical treatment combinations (adapalene with BPO; tretinoin with clindamycin), or oral tetracyclines (doxycycline or lymecycline) combined with topical treatments (azelaic acid, or a fixed combination of adapalene with BPO) were recommended as first‐line treatments of moderate‐to‐severe acne. Where oral lymecycline or doxycycline are contraindicated or not tolerated, alternative oral antibiotics (trimethoprim or an oral macrolide such as erythromycin) might be considered. BPO might be considered across both severity levels if other recommended first‐line treatments are contraindicated (e.g. during pregnancy) or if there is a patient preference against their use. PDT might be considered for adults with moderate‐to‐severe acne if other treatments are ineffective, not tolerated or contraindicated.", "This economic analysis allowed estimation of the relative cost‐effectiveness of a range of topical pharmacological, oral pharmacological, physical and combined treatments for acne stratified by severity level. The results informed the NICE national guidance on the management of acne vulgaris. There remains a need for well‐conducted studies that examine the long‐term clinical efficacy and cost‐effectiveness of the full range of acne treatments.", "\nWhat's already known about this topic?\nAcne vulgaris is the eighth most common disease globally.In a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.Evidence on the cost‐effectiveness of acne vulgaris treatments is lacking.Identifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.\nWhat does this study add?\nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.For mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).For moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.These findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.\n\n\n[SUBTITLE] What's already known about this topic? [SUBSECTION] \nAcne vulgaris is the eighth most common disease globally.In a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.Evidence on the cost‐effectiveness of acne vulgaris treatments is lacking.Identifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.\n\nAcne vulgaris is the eighth most common disease globally.\nIn a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.\nEvidence on the cost‐effectiveness of acne vulgaris treatments is lacking.\nIdentifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.\n\nAcne vulgaris is the eighth most common disease globally.In a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.Evidence on the cost‐effectiveness of acne vulgaris treatments is lacking.Identifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.\n\nAcne vulgaris is the eighth most common disease globally.\nIn a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.\nEvidence on the cost‐effectiveness of acne vulgaris treatments is lacking.\nIdentifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.\n[SUBTITLE] What does this study add? [SUBSECTION] \nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.For mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).For moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.These findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.\n\nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.\nFor mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).\nFor moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.\nThese findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.\n\nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.For mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).For moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.These findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.\n\nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.\nFor mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).\nFor moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.\nThese findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.", "\nAcne vulgaris is the eighth most common disease globally.In a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.Evidence on the cost‐effectiveness of acne vulgaris treatments is lacking.Identifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.\n\nAcne vulgaris is the eighth most common disease globally.\nIn a previous NMA, topical treatment combinations, chemical peels and photochemical therapies were most effective for mild‐to‐moderate acne; and topical treatment combinations, oral antibiotics combined with topical treatments, oral isotretinoin and PDT were most effective for moderate‐to‐severe acne.\nEvidence on the cost‐effectiveness of acne vulgaris treatments is lacking.\nIdentifying acne vulgaris treatments that ensure efficient healthcare resource use is needed.", "\nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.For mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).For moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.These findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.\n\nAll treatments are more cost‐effective than treatment with placebo, modelled as GP visits without active treatment.\nFor mild‐to‐moderate acne, the most cost‐effective treatments include topical treatment combinations and photochemical therapy (combined blue/red light).\nFor moderate‐to‐severe acne, the most cost‐effective treatments include topical treatment combinations, oral antibiotics combined with topical treatments, and oral isotretinoin.\nThese findings, combined with NMA findings and other clinical considerations, informed NICE guidance on the management of acne vulgaris.", "IM, NB, LB and KD received support from the NGA for the submitted work. CHD and NJW received support from the NICE Centre for Guidelines for the submitted work. JW, JCR, DW and EH declared the following interests based on the NICE policy on conflicts of interests: https://www.nice.org.uk/guidance/ng198/documents/register‐of‐interests. The authors report no other relationships or activities that could appear to have influenced the submitted work.", "This work was undertaken by the National Guideline Alliance (NGA) at the Royal College of Obstetricians and Gynaecologists (RCOG), with support from the National Institute for Health and Care Excellence (NICE) Guidelines Technical Support Unit (TSU), University of Bristol, which is funded by the NICE Centre for Guidelines. NGA has received funding from NICE to develop clinical, public health and social care guidelines. For the development of this guideline, NICE worked with the British Association of Dermatologists. The views expressed in this publication are those of the authors and not necessarily those of RCOG, NGA or NICE. The funder of the study had no further role in the study design, data collection, data analysis, data interpretation, or writing of the report. All authors had full access to all the data in the study and had final responsibility for the decision to submit for publication. National Institute for Health and Care Excellence (2021) Acne Vulgaris: management. Available from: https://nice.org.uk/guidance/ng198.", "Ethics approval and informed consent not applicable.", "\nSupplementary Data S1. Description of economic model structure and assumptions.\nClick here for additional data file.\n\nSupplementary Data S2. Relative effects on efficacy and discontinuation used in the economic analysis.\nClick here for additional data file.\n\nSupplementary Data S3. Methods of estimation of absolute effects for the reference treatment.\nClick here for additional data file.\n\nSupplementary Data S4. Distribution around the mean percentage change in total acne lesion count from baseline (%CFB) in the economic model.\nClick here for additional data file.\n\nSupplementary Data S5. Methods of estimation of utility values used in the economic analysis.\nClick here for additional data file.\n\nSupplementary Data S6. Intervention costs of treatments for acne vulgaris used in the economic analysis.\nClick here for additional data file.\n\nSupplementary Data S7. Estimation of the average acne care cost.\nClick here for additional data file.\n\nSupplementary Data S8. Model input values, probability distributions and scenarios tested in deterministic sensitivity analysis.\nClick here for additional data file.\n\nSupplementary Data S9. Results of deterministic sensitivity analysis.\nClick here for additional data file.\n\nSupplementary Data S10. Published economic assessments of acne treatments.\nClick here for additional data file.\n\nSupplementary Table S1. Cost‐effectiveness results for treatments for moderate‐to‐severe acne in males.\nClick here for additional data file." ]
[ null, "methods", null, null, null, null, null, null, null, null, "results", "discussion", "conclusions", null, null, null, "COI-statement", null, null, "supplementary-material" ]
[]
Diagnostic value of total testosterone and free androgen index measured by LC-MS/MS for PCOS and insulin resistance.
36258308
The objective of the study was to explore the clinical significance of steroid hormones in the diagnosis of PCOS and PCOS-related insulin resistance through liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS) and chemiluminescent immunoassay (CLIA).
BACKGROUND
The study included 114 patients with PCOS and 100 controls. Steroid hormone levels in serum were measured using LC-MS/MS and CLIA. The Bland-Altman method was used to check the consistency between the two methods. The diagnostic value of the LC-MS/MS method for female hyperandrogenemia and PCOS was evaluated.
METHODS
Women with PCOS were younger than controls on average (p < 0.001). PCOS patients had higher luteal hormone (LH, p < 0.001), insulin (p = 0.002), estradiol (E2, p < 0.001), total testosterone (TT, p < 0.001), free androgen index (FAI, p = 0.021), dehydroepiandrosterone sulfate (DHEA, p = 0.021), insulin resistance index (HOMA-IR) (p = 0.034), and fasting glucose (p = 0.017) levels than controls as measured by CLIA. The diagnostic value of TT was the best, and the area under the AUC curve was 0.766. Women with PCOS had higher androstenedione (A2, p < 0.001), FAI (p < 0.001), TT (p < 0.001), and 17-hydroxyprogesterone (17-OHP, p < 0.001) levels than controls as measured by LC-MS/MS. The ROC curve showed that the diagnostic efficacy of A2, TT, and 17-OHP was 0.830, 0.851, and 0.714, respectively. The consistency of TT detected by LC-MS/MS and CLIA was poor according to the Bland-Altman method. Detected TT by LC-MS/MS had the highest diagnostic efficiency for PCOS. The diagnostic power of the LC-MS/MS results for PCOS-related insulin resistance was analyzed. The results showed that the FAI had the highest diagnostic power, with an ROC curve of 0.798.
RESULTS
LC-MS/MS is more sensitive and accurate than CLIA in the determination of serum TT and FAI. TT is more effective for the diagnosis of PCOS, whereas FAI is more valuable in the diagnosis of insulin resistance.
CONCLUSION
[ "Female", "Humans", "Testosterone", "Chromatography, Liquid", "Androgens", "Insulin Resistance", "Polycystic Ovary Syndrome", "Tandem Mass Spectrometry", "Insulin", "Steroids" ]
9701831
null
null
null
null
RESULTS
[SUBTITLE] Baseline characteristics and hormone levels between the control and PCOS groups [SUBSECTION] There was a significant difference in age between the groups (control vs. PCOS, 26.53 ± 3.30 vs. 24.18 ± 4.98, p < 0.001). However, there was no difference in height (control vs. PCOS, 1.62 ± 0.05 vs. 1.63 ± 0.05, p = 0.276), weight (control vs. PCOS, 63.15 ± 10.23 vs. 65.94 ± 13.54, p = 0.088) or BMI (control vs. PCOS, 24.01 ± 3.55 vs. 24.84 ± 4.57, p = 0.140). The main baseline characteristics of the subjects are given in Table 1. As given in Table 1, there were differences between the control group and the PCOS group in luteal hormone (LH) (4.90 ± 4.21 vs. 13.86 ± 6.99, p < 0.001), insulin (8.58 ± 7.52 vs. 13.53 ± 14.97, p = 0.002), E2 (44.00 ± 15.54 vs. 75.19 ± 49.61, p < 0.001), HOMA‐IR (1.18 ± 2.20 vs. 3.15 ± 4.00, p = 0.034), TT (0.56 ± 0.27 vs. 0.75 ± 0.23, p < 0.001), FAI (0.019 ± 0.027 vs. 0.032 ± 0.027, p = 0.021), DHEA (6.89 ± 0.26 vs. 7.91 ± 3.16, p = 0.021), uric acid (223.19 ± 149.87 vs. 283.64 ± 151.93, p = 0.004), total protein (53.55 ± 34.52 vs. 62.91 ± 28.56, p = 0.033), total cholesterol (3.29 ± 2.14 vs. 4.13 ± 1.81, p = 0.002), GPT (17.02 ± 21.06 vs. 28.10 ± 29.84, p = 0.002), GOT (16.92 ± 15.04 vs. 22.28 ± 13.99, p = 0.008), and fasting glucose (4.04 ± 2.34 vs. 4.71 ± 1.64, p = 0.017) levels as measured by CLIA (Table 1). Since the LC–MS/MS method has higher accuracy and lower uncertainty, LC–MS/MS was used to detect steroid hormones in both groups. As given in Table 2, women with PCOS had higher androstenedione (5.04 ± 1.68 vs. 3.08 ± 1.28, p < 0.001), FAI (2.17 ± 1.64 vs. 0.94 ± 1.19, p < 0.001), TT (0.53 ± 0.21 vs. 0.30 ± 0.14, p < 0.001), and 17‐OHP (1.08 ± 0.71 vs. 0.69 ± 0.44, p < 0.001) levels than those in the controls, whereas there were no differences in serum concentrations of the steroids DHEA‐S, cortisol, cortisone, corticosterone, and 11‐deoxycortisol. There was a significant difference in age between the groups (control vs. PCOS, 26.53 ± 3.30 vs. 24.18 ± 4.98, p < 0.001). However, there was no difference in height (control vs. PCOS, 1.62 ± 0.05 vs. 1.63 ± 0.05, p = 0.276), weight (control vs. PCOS, 63.15 ± 10.23 vs. 65.94 ± 13.54, p = 0.088) or BMI (control vs. PCOS, 24.01 ± 3.55 vs. 24.84 ± 4.57, p = 0.140). The main baseline characteristics of the subjects are given in Table 1. As given in Table 1, there were differences between the control group and the PCOS group in luteal hormone (LH) (4.90 ± 4.21 vs. 13.86 ± 6.99, p < 0.001), insulin (8.58 ± 7.52 vs. 13.53 ± 14.97, p = 0.002), E2 (44.00 ± 15.54 vs. 75.19 ± 49.61, p < 0.001), HOMA‐IR (1.18 ± 2.20 vs. 3.15 ± 4.00, p = 0.034), TT (0.56 ± 0.27 vs. 0.75 ± 0.23, p < 0.001), FAI (0.019 ± 0.027 vs. 0.032 ± 0.027, p = 0.021), DHEA (6.89 ± 0.26 vs. 7.91 ± 3.16, p = 0.021), uric acid (223.19 ± 149.87 vs. 283.64 ± 151.93, p = 0.004), total protein (53.55 ± 34.52 vs. 62.91 ± 28.56, p = 0.033), total cholesterol (3.29 ± 2.14 vs. 4.13 ± 1.81, p = 0.002), GPT (17.02 ± 21.06 vs. 28.10 ± 29.84, p = 0.002), GOT (16.92 ± 15.04 vs. 22.28 ± 13.99, p = 0.008), and fasting glucose (4.04 ± 2.34 vs. 4.71 ± 1.64, p = 0.017) levels as measured by CLIA (Table 1). Since the LC–MS/MS method has higher accuracy and lower uncertainty, LC–MS/MS was used to detect steroid hormones in both groups. As given in Table 2, women with PCOS had higher androstenedione (5.04 ± 1.68 vs. 3.08 ± 1.28, p < 0.001), FAI (2.17 ± 1.64 vs. 0.94 ± 1.19, p < 0.001), TT (0.53 ± 0.21 vs. 0.30 ± 0.14, p < 0.001), and 17‐OHP (1.08 ± 0.71 vs. 0.69 ± 0.44, p < 0.001) levels than those in the controls, whereas there were no differences in serum concentrations of the steroids DHEA‐S, cortisol, cortisone, corticosterone, and 11‐deoxycortisol. [SUBTITLE] Diagnostic value of TT detected by LC–MS/MS for PCOS [SUBSECTION] The diagnostic values of hormones with significant differences between the control and PCOS groups were evaluated. As shown in Figure 1A, E2, TT, and DHES had good diagnostic value for PCOS detected by CLIA. Moreover, TT had the highest diagnostic potency for PCOS in the CLIA group, with an area under the curve (AUC) of 0.766. A2, TT and 17‐OHP tested by LC–MS/MS were further analyzed for their diagnostic value for PCOS. The AUCs of A2, TT, and 17‐OHP were 0.83, 0.851, and 0.714, respectively (Figure 1B). These results supported that TT had a better diagnostic value for PCOS not only detected by CLIA but also by LC–MS/MS. Moreover, TT detected by LC–MS/MS had a higher diagnostic value than TT detected by CLIA. ROC curve for detecting the different hormones which were difference between NC and women with PCOS by CLIA and by LC–MS/MS. (A). AUC of E2, TT, and DHES by CLIA were 0.741, 0.766, and 0.569. (B). AUC of A2, TT, and 17‐OHP by LC–MS/MS were 0.831, 0.851, and 0.714. Since TT detected by LC–MS/MS had a higher diagnostic value than TT detected by CLIA, the agreement between the two methods was analyzed. The results suggested that the consistency between the results of the two methods was poor, and the stability of TT detected by LC–MS/MS was higher than that of TT detected by CLIA (Figure 2). Comparison of TT concentration between CLIA and LC–MS/MS. (A). The concentration of TT between control women and PCOS women by CLIA or LC–MS/MS. The concentration of TT in LC–MS/MS was lower and more stable than the CLIA. ***indicated p < 0.001. (B). The Bland–Altman method was used to detected the consistency of TT between two methods. The showed poor consistency between CLIA and LC–MS/MS. The diagnostic values of hormones with significant differences between the control and PCOS groups were evaluated. As shown in Figure 1A, E2, TT, and DHES had good diagnostic value for PCOS detected by CLIA. Moreover, TT had the highest diagnostic potency for PCOS in the CLIA group, with an area under the curve (AUC) of 0.766. A2, TT and 17‐OHP tested by LC–MS/MS were further analyzed for their diagnostic value for PCOS. The AUCs of A2, TT, and 17‐OHP were 0.83, 0.851, and 0.714, respectively (Figure 1B). These results supported that TT had a better diagnostic value for PCOS not only detected by CLIA but also by LC–MS/MS. Moreover, TT detected by LC–MS/MS had a higher diagnostic value than TT detected by CLIA. ROC curve for detecting the different hormones which were difference between NC and women with PCOS by CLIA and by LC–MS/MS. (A). AUC of E2, TT, and DHES by CLIA were 0.741, 0.766, and 0.569. (B). AUC of A2, TT, and 17‐OHP by LC–MS/MS were 0.831, 0.851, and 0.714. Since TT detected by LC–MS/MS had a higher diagnostic value than TT detected by CLIA, the agreement between the two methods was analyzed. The results suggested that the consistency between the results of the two methods was poor, and the stability of TT detected by LC–MS/MS was higher than that of TT detected by CLIA (Figure 2). Comparison of TT concentration between CLIA and LC–MS/MS. (A). The concentration of TT between control women and PCOS women by CLIA or LC–MS/MS. The concentration of TT in LC–MS/MS was lower and more stable than the CLIA. ***indicated p < 0.001. (B). The Bland–Altman method was used to detected the consistency of TT between two methods. The showed poor consistency between CLIA and LC–MS/MS. [SUBTITLE] Diagnostic value of FAI detected by LC–MS/MS for insulin resistance of PCOS [SUBSECTION] Insulin resistance is the most serious complication of PCOS, and the diagnostic value of steroid hormones detected by CLIA and LC–MS/MS for insulin resistance was tested. First, Pearson's correlation was used to test the steroid hormones associated with insulin resistance. A2 (R = 0.201, p = 0.003) and 17‐OHP (R = 0.232, p = 0.001) had better positive relationships with HOMA‐IR than TT, detected by both CLIA (R = 0.149, p = 0.03) and by LC–MS/MS (R = 0.149, p = 0.029) (Figure 3). The diagnostic efficacy of all differential indicators was analyzed for insulin resistance among PCOS patients. The AUCs of TT (CLIA), A2, TT (LC–MS/MS), and 17‐OHP were 0.587, 0.611, 0.602, and 0.572, respectively (Figure 4). TT in the body is either combined with SHBG or free in serum. It is generally thought that only free testosterone in serum, called FAI, could play an important role in PCOS. 15 The diagnostic efficacy of FAI by CLIA and LC–MS/MS was analyzed. According to the results, the AUCs of FAI detected by CLIA and LC–MS/MS were 0.788 and 0.798, respectively (Figure 4). Therefore, FAI detected by LC–MS/MS had the best diagnostic value for insulin resistance among PCOS patients. Correlation between the different hormones which were difference between NC and women with PCOS and HOMA‐IR. (A). Correlation between TT detected by CLIA and HOMA‐IR in women with PCOS (R = 0.149, p = 0.03); (B) Correlation between testosterone detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.149, p = 0.029); (C). Correlation between androstenedione detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.201, p = 0.003); (D). Correlation between 17α‐hydroxyprogesterone detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.232, p = 0.001). Assessment of the diagnostic potency of androgens for insulin resistance in women with PCOS by CLIA and LC–MS/MS. AUC of TT by CLIA, TT by LC–MS/MS, A2 by LC–MS/MS, 17‐OHP by LC–MS/MS, FAI by CLIA and FAI by LC–MS/MS were 0.587, 0.602, 0.611, 0.572, 0.788, and 0.798. Insulin resistance is the most serious complication of PCOS, and the diagnostic value of steroid hormones detected by CLIA and LC–MS/MS for insulin resistance was tested. First, Pearson's correlation was used to test the steroid hormones associated with insulin resistance. A2 (R = 0.201, p = 0.003) and 17‐OHP (R = 0.232, p = 0.001) had better positive relationships with HOMA‐IR than TT, detected by both CLIA (R = 0.149, p = 0.03) and by LC–MS/MS (R = 0.149, p = 0.029) (Figure 3). The diagnostic efficacy of all differential indicators was analyzed for insulin resistance among PCOS patients. The AUCs of TT (CLIA), A2, TT (LC–MS/MS), and 17‐OHP were 0.587, 0.611, 0.602, and 0.572, respectively (Figure 4). TT in the body is either combined with SHBG or free in serum. It is generally thought that only free testosterone in serum, called FAI, could play an important role in PCOS. 15 The diagnostic efficacy of FAI by CLIA and LC–MS/MS was analyzed. According to the results, the AUCs of FAI detected by CLIA and LC–MS/MS were 0.788 and 0.798, respectively (Figure 4). Therefore, FAI detected by LC–MS/MS had the best diagnostic value for insulin resistance among PCOS patients. Correlation between the different hormones which were difference between NC and women with PCOS and HOMA‐IR. (A). Correlation between TT detected by CLIA and HOMA‐IR in women with PCOS (R = 0.149, p = 0.03); (B) Correlation between testosterone detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.149, p = 0.029); (C). Correlation between androstenedione detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.201, p = 0.003); (D). Correlation between 17α‐hydroxyprogesterone detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.232, p = 0.001). Assessment of the diagnostic potency of androgens for insulin resistance in women with PCOS by CLIA and LC–MS/MS. AUC of TT by CLIA, TT by LC–MS/MS, A2 by LC–MS/MS, 17‐OHP by LC–MS/MS, FAI by CLIA and FAI by LC–MS/MS were 0.587, 0.602, 0.611, 0.572, 0.788, and 0.798.
null
null
[ "INTRODUCTION", "Study subjects", "Hormone measurement by CLIA\n", "Steroid hormone measurement by LC–MS/MS\n", "Statistical analysis", "Baseline characteristics and hormone levels between the control and PCOS groups", "Diagnostic value of TT detected by LC–MS/MS for PCOS\n", "Diagnostic value of FAI detected by LC–MS/MS for insulin resistance of PCOS\n", "FUNDING INFORMATION", "ETHICS STATEMENT" ]
[ "Polycystic ovarian syndrome (PCOS) is a common endocrine system disorder affecting 5–15% of fertile women worldwide.\n1\n The symptoms of PCOS are often hyperandrogenism, hyperinsulinemia, menstrual dysfunction, hirsutism, and infertility.\n2\n PCOS patients need early detection and early treatment; if not treated in time, PCOS may lead to diabetes, hyperlipidemia, cardiovascular diseases, and even endometrial cancer, which have a substantial influence on women's physical and mental health.\n3\n\n\nAlthough the etiology of PCOS is not clear, many views believe that hyperandrogenism plays a key role in the pathogenesis and progression of PCOS. On the one hand, hyperandrogenism inhibits the maturation of follicles and causes multiple follicular cysts; on the other hand, the increase in androgen causes dysfunction of the hypothalamic–pituitary–gonadal axis; thus, a vicious cycle of excessive androgen and continuous anovulation is formed.\n4\n Research shows that approximately 60%–80% of PCOS patients have hyperandrogenemia.\n5\n At present, clinical antiandrogen therapy has not achieved satisfactory results, which is caused by the complicated mechanisms of androgen production.\n6\n The diagnosis of hyperandrogenemia plays a positive role in the early diagnosis, treatment, and long‐term prevention of PCOS.\nInsulin resistance is the most serious complication in PCOS patients and can lead to lipid metabolism disorder, vascular endothelial damage, and diabetes. Studies show that 75% of obese PCOS patients have insulin resistance; even if their weight is normal, they may also have hyperinsulinemia and abnormal blood glucose.\n7\n Therefore, the diagnosis of insulin resistance in PCOS is a key step in the treatment of the long‐term risks of PCOS.\nAt present, the level of steroid hormones is mainly detected by chemiluminescence immunoassay (CLIA).\n8\n, \n9\n, \n10\n CLIA is relatively safe (no radiation hazard), stable, rapid, and easily automated, and the detection level can reach ng/ml.\n11\n However, CLIA is mainly based on antigen antibody immune reactions, and nonspecific binding is inevitable; CLIA may overestimate androgen levels, resulting in poor accuracy and sensitivity.\n12\n Increasing evidence has shown that liquid chromatography–mass spectrometry/mass spectrometry (LC–MS/MS) is more precise than CLIA.\n13\n, \n14\n LC–MS/MS methods have higher accuracy and lower uncertainty than immunoassays, especially when the concentration of testosterone is relatively low.\n14\n In recent years, with the continuous maturity of technology, an increasing number of institutions have used LC–MS/MS to detect testosterone. It is believed that precise measurement of total testosterone is very important for the diagnosis, treatment, and prevention of PCOS.\nIn this study, we analyzed and compared the total testosterone (TT) and free testosterone index (FAI) levels of PCOS patients and control women detected by LC–MS/MS and CLIA and evaluated the diagnostic value of the LC/MS method for female hyperandrogenemia and PCOS.", "A total of 114 patients with PCOS and 100 controls were recruited at Nanjing Maternal and Child Health Care Hospital. All procedures were performed with written informed consent. PCOS is diagnosed when at least two of the following criteria are met: (i) oligomenorrhoea and/or anovulation, (ii) clinical and/or biochemical hyperandrogenism and (iii) polycystic ovaries according to the Rotterdam 2003 criteria. None of the women took hormonal drugs for at least 3 months before our study.", "Hormone levels were tested in the Clinical Laboratory of Nanjing Maternal and Child Health Care Hospital. As given in Table 1, luteal hormone (LH), follitropin (FSH), prolaction (PRL), prpgesterone (PROG), cortisol, insulin, estradiol (E2), TT, dehydroepiandrosterone sulfate (DHEA) were measured by a Beckman automated chemiluminescence immunoanalyzer 7500 (Beckman). Sex hormone‐binding globulin (SHBG) were tested by Cobas 6000 (Roche). Uric acid, total protein, total cholesterol, glutamic‐pyruvic transaminase (GPT), glutamic oxiracetam transaminase (GPT), and fasting glucose were test by Beckman coulter AU5800 (Beckman). All the reagents required for the test were matching reagents, which were used according to the specified requirements. The remainng serum was stored at −80°C for LC–MS/MS analysis.\nBaseline characteristics and hormone levels by CLIA in control and PCOS patients\nAbbreviations: FSH, follicle‐stimulating hormone; GOT, glutamic oxiracetam transaminase; GPT, glutamic‐pyruvic transaminase; LH, luteal hormone; SHBG, sex hormone‐binding globulin.a\nP values were determined by two tailed student's t‐test.\nHomeostatic model assessment of insulin resistance (HOMA‐IR) is an indicator used to evaluate insulin resistance. HOMA‐IR is equal to fasting blood glucose (FBG) × fasting insulin (fin)/22.5. The FAI is equal to 100 × total testosterone (TT)/sex hormone‐binding globulin (SHBG). Insulin resistance (IR) was defined as HOMA‐IR≥2.14 and fin≥12.6 μIU/ml.", "As given in Table 2, aldosterone (ALD), 18‐hydroxycorticosterone(18‐OH‐B), cortisone, cortisol/hydrocortisone, 21‐deoxycortisol(21‐DOC), corticosterone, 11‐deoxycortisol, androstenedione (A2), 11‐deoxycorticosterone, total testosterone 17‐hydroxyprogesterone (17‐OHP), 17‐hydroxy pregnenolone, DHT, PROG, androsterone, pregnenolone, and HHEA‐S were assayed by LC–MS/MS.\nSteroid hormone level by LC–MS in control and PCOS patients\nAbbreviations: 17‐OHP, 17‐hydroxyprogesterone; 18‐OH‐B, 18‐hydroxycorticosterone; 21‐DOC, 21‐deoxycortisol; ALD, aldosterone; DEHA‐S, dehydroepiandrosterone sulfate; FAI, free androgen index.a\nP values were determined by two tailed student's t‐test.\nFirstly, 10 mg of each pure calibrator was dissolved in 10 ml methanol (Merck Millipore) to obtain a 1 mg/ml solution, which was stored at −80°C. The stock solutions were diluted with methanol and mixed to obtain preworking solutions containing the 18 calibrators (Cambridge Isotope Laboratories). Then, the samples were serially diluted to obtain a standard curve intermediate. The calibrator working solution was prepared with 5% methanol/water and stored in the refrigerator at −20°C. Secondly, each internal standard (IS) was weighed and dissolved in methanol to obtain solutions of different concentrations. Each internal standard mother solution was diluted and mixed with acetonitrile (Merck Millipore, USA) to obtain the internal standard working solution and stored at −20°C. Then, steroid hormones in serum were treated by protein precipitation combined with solid phase extraction (SPE). The specific steps are as follows: A 200 μl serum sample was mixed with 200 μl internal standard solution. The mixture was rotated for 3 min. Then, 500 μl of deionized water was added, followed by 1 min of shaking. After centrifugation at 15,000 × g for 10 min, the mixture was transferred to a well on a Waters Oasis Prime HLB μElution column plate for SPE. A 700 μl extraction supernatant was added to the 96‐well Oasis Prime HLB μElution column plate, the balance was activated with 200 μl methanol and 200 μl ddH2O, and the supernatant slowly flowed through the SPE plate and into the waste container by positive pressure. Then, 200 ml methanol/ddH2O (15:85, v/v) was added to the SPE plate in sequence. Then, 30 ml acetonitrile was added to the SPE plate twice, and the filtrate was collected. The filtrate was diluted with 60 μl methanol/ddH2O (5:95, v/v) and vortexed. The dilution filtrates were collected for LC–MS/MS analysis.\nTen microliters of filtrate from SPE was added to the Waters® ACQUITY UPLC® System – XevoTM TQS MS for LC–MS/MS analysis. The auto sampler temperature was 10°C. The binary mobile phase consisted of 2 mM ammonium acetate (Sigma‐Aldrich, USA) + 0.1% formic acid (Sigma‐Aldrich) + ddH2O (A) and 2 mM ammonium acetate+0.1% formic acid +methanol (B). The column temperature was kept at 45°C. The optimized parameters for MS/MS were as follows: capillary voltage, 3.2 kV; cone voltage, 50 V; source temperature, 150°C; desolvation temperature, 650°C; cone gas flow, 150 L/h; desolvation gas flow, 800 L/h; and collision gas flow, 0.15 ml/min. The steroid profile was based on electrospray ionization‐mass spectrometry (ESI‐MS) data combined with tandem mass spectrometry in the positive ion mode or negative ion mode. Quantitation by multiple reaction monitoring (MRM) analysis was performed in the positive ion mode for analytes and in the negative ion mode for DHEAs.", "SPSS 20.0 was used for analysis. The variables are presented as the mean ± standard deviation. Independent sample t tests were used for intergroup comparisons. The differences detected by LC–MS/MS and CLIA between the PCOS group and the control group were compared by interaction analysis. p < 0.05 was considered statistically significant. Spearman rank correlation analysis was used to analyze the correlation between total testosterone as detected by the LC–MS/MS method and by the CLIA method. The chi square test was used to compare the rates between groups. The Bland–Altman method was used to measure the consistency of the two methods. ROC curves were used to analyze the PCOS diagnostic efficiency.", "There was a significant difference in age between the groups (control vs. PCOS, 26.53 ± 3.30 vs. 24.18 ± 4.98, p < 0.001). However, there was no difference in height (control vs. PCOS, 1.62 ± 0.05 vs. 1.63 ± 0.05, p = 0.276), weight (control vs. PCOS, 63.15 ± 10.23 vs. 65.94 ± 13.54, p = 0.088) or BMI (control vs. PCOS, 24.01 ± 3.55 vs. 24.84 ± 4.57, p = 0.140). The main baseline characteristics of the subjects are given in Table 1.\nAs given in Table 1, there were differences between the control group and the PCOS group in luteal hormone (LH) (4.90 ± 4.21 vs. 13.86 ± 6.99, p < 0.001), insulin (8.58 ± 7.52 vs. 13.53 ± 14.97, p = 0.002), E2 (44.00 ± 15.54 vs. 75.19 ± 49.61, p < 0.001), HOMA‐IR (1.18 ± 2.20 vs. 3.15 ± 4.00, p = 0.034), TT (0.56 ± 0.27 vs. 0.75 ± 0.23, p < 0.001), FAI (0.019 ± 0.027 vs. 0.032 ± 0.027, p = 0.021), DHEA (6.89 ± 0.26 vs. 7.91 ± 3.16, p = 0.021), uric acid (223.19 ± 149.87 vs. 283.64 ± 151.93, p = 0.004), total protein (53.55 ± 34.52 vs. 62.91 ± 28.56, p = 0.033), total cholesterol (3.29 ± 2.14 vs. 4.13 ± 1.81, p = 0.002), GPT (17.02 ± 21.06 vs. 28.10 ± 29.84, p = 0.002), GOT (16.92 ± 15.04 vs. 22.28 ± 13.99, p = 0.008), and fasting glucose (4.04 ± 2.34 vs. 4.71 ± 1.64, p = 0.017) levels as measured by CLIA (Table 1).\nSince the LC–MS/MS method has higher accuracy and lower uncertainty, LC–MS/MS was used to detect steroid hormones in both groups. As given in Table 2, women with PCOS had higher androstenedione (5.04 ± 1.68 vs. 3.08 ± 1.28, p < 0.001), FAI (2.17 ± 1.64 vs. 0.94 ± 1.19, p < 0.001), TT (0.53 ± 0.21 vs. 0.30 ± 0.14, p < 0.001), and 17‐OHP (1.08 ± 0.71 vs. 0.69 ± 0.44, p < 0.001) levels than those in the controls, whereas there were no differences in serum concentrations of the steroids DHEA‐S, cortisol, cortisone, corticosterone, and 11‐deoxycortisol.", "The diagnostic values of hormones with significant differences between the control and PCOS groups were evaluated. As shown in Figure 1A, E2, TT, and DHES had good diagnostic value for PCOS detected by CLIA. Moreover, TT had the highest diagnostic potency for PCOS in the CLIA group, with an area under the curve (AUC) of 0.766. A2, TT and 17‐OHP tested by LC–MS/MS were further analyzed for their diagnostic value for PCOS. The AUCs of A2, TT, and 17‐OHP were 0.83, 0.851, and 0.714, respectively (Figure 1B). These results supported that TT had a better diagnostic value for PCOS not only detected by CLIA but also by LC–MS/MS. Moreover, TT detected by LC–MS/MS had a higher diagnostic value than TT detected by CLIA.\nROC curve for detecting the different hormones which were difference between NC and women with PCOS by CLIA and by LC–MS/MS. (A). AUC of E2, TT, and DHES by CLIA were 0.741, 0.766, and 0.569. (B). AUC of A2, TT, and 17‐OHP by LC–MS/MS were 0.831, 0.851, and 0.714.\nSince TT detected by LC–MS/MS had a higher diagnostic value than TT detected by CLIA, the agreement between the two methods was analyzed. The results suggested that the consistency between the results of the two methods was poor, and the stability of TT detected by LC–MS/MS was higher than that of TT detected by CLIA (Figure 2).\nComparison of TT concentration between CLIA and LC–MS/MS. (A). The concentration of TT between control women and PCOS women by CLIA or LC–MS/MS. The concentration of TT in LC–MS/MS was lower and more stable than the CLIA. ***indicated p < 0.001. (B). The Bland–Altman method was used to detected the consistency of TT between two methods. The showed poor consistency between CLIA and LC–MS/MS.", "Insulin resistance is the most serious complication of PCOS, and the diagnostic value of steroid hormones detected by CLIA and LC–MS/MS for insulin resistance was tested. First, Pearson's correlation was used to test the steroid hormones associated with insulin resistance. A2 (R = 0.201, p = 0.003) and 17‐OHP (R = 0.232, p = 0.001) had better positive relationships with HOMA‐IR than TT, detected by both CLIA (R = 0.149, p = 0.03) and by LC–MS/MS (R = 0.149, p = 0.029) (Figure 3). The diagnostic efficacy of all differential indicators was analyzed for insulin resistance among PCOS patients. The AUCs of TT (CLIA), A2, TT (LC–MS/MS), and 17‐OHP were 0.587, 0.611, 0.602, and 0.572, respectively (Figure 4).\nTT in the body is either combined with SHBG or free in serum. It is generally thought that only free testosterone in serum, called FAI, could play an important role in PCOS.\n15\n The diagnostic efficacy of FAI by CLIA and LC–MS/MS was analyzed. According to the results, the AUCs of FAI detected by CLIA and LC–MS/MS were 0.788 and 0.798, respectively (Figure 4). Therefore, FAI detected by LC–MS/MS had the best diagnostic value for insulin resistance among PCOS patients.\nCorrelation between the different hormones which were difference between NC and women with PCOS and HOMA‐IR. (A). Correlation between TT detected by CLIA and HOMA‐IR in women with PCOS (R = 0.149, p = 0.03); (B) Correlation between testosterone detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.149, p = 0.029); (C). Correlation between androstenedione detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.201, p = 0.003); (D). Correlation between 17α‐hydroxyprogesterone detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.232, p = 0.001).\nAssessment of the diagnostic potency of androgens for insulin resistance in women with PCOS by CLIA and LC–MS/MS. AUC of TT by CLIA, TT by LC–MS/MS, A2 by LC–MS/MS, 17‐OHP by LC–MS/MS, FAI by CLIA and FAI by LC–MS/MS were 0.587, 0.602, 0.611, 0.572, 0.788, and 0.798.", "This project was supported by the National Natural Science Foundation of China (Grant Nos. 81901446, 81901555, and 81971351) and the Fund of Nanjing Health and Health Committee (YKK18161).", "The experimental protocols were approved by the Ethics Committee of the Nanjing Maternity and Child Health Care Hospital. The methods were performed in accordance with the approved guidelines by the Ethics Committee of the Nanjing Maternity and Child Health Care Hospital." ]
[ null, null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "MATERIALS AND METHODS", "Study subjects", "Hormone measurement by CLIA\n", "Steroid hormone measurement by LC–MS/MS\n", "Statistical analysis", "RESULTS", "Baseline characteristics and hormone levels between the control and PCOS groups", "Diagnostic value of TT detected by LC–MS/MS for PCOS\n", "Diagnostic value of FAI detected by LC–MS/MS for insulin resistance of PCOS\n", "DISCUSSION", "FUNDING INFORMATION", "CONFLICT OF INTEREST", "ETHICS STATEMENT" ]
[ "Polycystic ovarian syndrome (PCOS) is a common endocrine system disorder affecting 5–15% of fertile women worldwide.\n1\n The symptoms of PCOS are often hyperandrogenism, hyperinsulinemia, menstrual dysfunction, hirsutism, and infertility.\n2\n PCOS patients need early detection and early treatment; if not treated in time, PCOS may lead to diabetes, hyperlipidemia, cardiovascular diseases, and even endometrial cancer, which have a substantial influence on women's physical and mental health.\n3\n\n\nAlthough the etiology of PCOS is not clear, many views believe that hyperandrogenism plays a key role in the pathogenesis and progression of PCOS. On the one hand, hyperandrogenism inhibits the maturation of follicles and causes multiple follicular cysts; on the other hand, the increase in androgen causes dysfunction of the hypothalamic–pituitary–gonadal axis; thus, a vicious cycle of excessive androgen and continuous anovulation is formed.\n4\n Research shows that approximately 60%–80% of PCOS patients have hyperandrogenemia.\n5\n At present, clinical antiandrogen therapy has not achieved satisfactory results, which is caused by the complicated mechanisms of androgen production.\n6\n The diagnosis of hyperandrogenemia plays a positive role in the early diagnosis, treatment, and long‐term prevention of PCOS.\nInsulin resistance is the most serious complication in PCOS patients and can lead to lipid metabolism disorder, vascular endothelial damage, and diabetes. Studies show that 75% of obese PCOS patients have insulin resistance; even if their weight is normal, they may also have hyperinsulinemia and abnormal blood glucose.\n7\n Therefore, the diagnosis of insulin resistance in PCOS is a key step in the treatment of the long‐term risks of PCOS.\nAt present, the level of steroid hormones is mainly detected by chemiluminescence immunoassay (CLIA).\n8\n, \n9\n, \n10\n CLIA is relatively safe (no radiation hazard), stable, rapid, and easily automated, and the detection level can reach ng/ml.\n11\n However, CLIA is mainly based on antigen antibody immune reactions, and nonspecific binding is inevitable; CLIA may overestimate androgen levels, resulting in poor accuracy and sensitivity.\n12\n Increasing evidence has shown that liquid chromatography–mass spectrometry/mass spectrometry (LC–MS/MS) is more precise than CLIA.\n13\n, \n14\n LC–MS/MS methods have higher accuracy and lower uncertainty than immunoassays, especially when the concentration of testosterone is relatively low.\n14\n In recent years, with the continuous maturity of technology, an increasing number of institutions have used LC–MS/MS to detect testosterone. It is believed that precise measurement of total testosterone is very important for the diagnosis, treatment, and prevention of PCOS.\nIn this study, we analyzed and compared the total testosterone (TT) and free testosterone index (FAI) levels of PCOS patients and control women detected by LC–MS/MS and CLIA and evaluated the diagnostic value of the LC/MS method for female hyperandrogenemia and PCOS.", "[SUBTITLE] Study subjects [SUBSECTION] A total of 114 patients with PCOS and 100 controls were recruited at Nanjing Maternal and Child Health Care Hospital. All procedures were performed with written informed consent. PCOS is diagnosed when at least two of the following criteria are met: (i) oligomenorrhoea and/or anovulation, (ii) clinical and/or biochemical hyperandrogenism and (iii) polycystic ovaries according to the Rotterdam 2003 criteria. None of the women took hormonal drugs for at least 3 months before our study.\nA total of 114 patients with PCOS and 100 controls were recruited at Nanjing Maternal and Child Health Care Hospital. All procedures were performed with written informed consent. PCOS is diagnosed when at least two of the following criteria are met: (i) oligomenorrhoea and/or anovulation, (ii) clinical and/or biochemical hyperandrogenism and (iii) polycystic ovaries according to the Rotterdam 2003 criteria. None of the women took hormonal drugs for at least 3 months before our study.\n[SUBTITLE] Hormone measurement by CLIA\n [SUBSECTION] Hormone levels were tested in the Clinical Laboratory of Nanjing Maternal and Child Health Care Hospital. As given in Table 1, luteal hormone (LH), follitropin (FSH), prolaction (PRL), prpgesterone (PROG), cortisol, insulin, estradiol (E2), TT, dehydroepiandrosterone sulfate (DHEA) were measured by a Beckman automated chemiluminescence immunoanalyzer 7500 (Beckman). Sex hormone‐binding globulin (SHBG) were tested by Cobas 6000 (Roche). Uric acid, total protein, total cholesterol, glutamic‐pyruvic transaminase (GPT), glutamic oxiracetam transaminase (GPT), and fasting glucose were test by Beckman coulter AU5800 (Beckman). All the reagents required for the test were matching reagents, which were used according to the specified requirements. The remainng serum was stored at −80°C for LC–MS/MS analysis.\nBaseline characteristics and hormone levels by CLIA in control and PCOS patients\nAbbreviations: FSH, follicle‐stimulating hormone; GOT, glutamic oxiracetam transaminase; GPT, glutamic‐pyruvic transaminase; LH, luteal hormone; SHBG, sex hormone‐binding globulin.a\nP values were determined by two tailed student's t‐test.\nHomeostatic model assessment of insulin resistance (HOMA‐IR) is an indicator used to evaluate insulin resistance. HOMA‐IR is equal to fasting blood glucose (FBG) × fasting insulin (fin)/22.5. The FAI is equal to 100 × total testosterone (TT)/sex hormone‐binding globulin (SHBG). Insulin resistance (IR) was defined as HOMA‐IR≥2.14 and fin≥12.6 μIU/ml.\nHormone levels were tested in the Clinical Laboratory of Nanjing Maternal and Child Health Care Hospital. As given in Table 1, luteal hormone (LH), follitropin (FSH), prolaction (PRL), prpgesterone (PROG), cortisol, insulin, estradiol (E2), TT, dehydroepiandrosterone sulfate (DHEA) were measured by a Beckman automated chemiluminescence immunoanalyzer 7500 (Beckman). Sex hormone‐binding globulin (SHBG) were tested by Cobas 6000 (Roche). Uric acid, total protein, total cholesterol, glutamic‐pyruvic transaminase (GPT), glutamic oxiracetam transaminase (GPT), and fasting glucose were test by Beckman coulter AU5800 (Beckman). All the reagents required for the test were matching reagents, which were used according to the specified requirements. The remainng serum was stored at −80°C for LC–MS/MS analysis.\nBaseline characteristics and hormone levels by CLIA in control and PCOS patients\nAbbreviations: FSH, follicle‐stimulating hormone; GOT, glutamic oxiracetam transaminase; GPT, glutamic‐pyruvic transaminase; LH, luteal hormone; SHBG, sex hormone‐binding globulin.a\nP values were determined by two tailed student's t‐test.\nHomeostatic model assessment of insulin resistance (HOMA‐IR) is an indicator used to evaluate insulin resistance. HOMA‐IR is equal to fasting blood glucose (FBG) × fasting insulin (fin)/22.5. The FAI is equal to 100 × total testosterone (TT)/sex hormone‐binding globulin (SHBG). Insulin resistance (IR) was defined as HOMA‐IR≥2.14 and fin≥12.6 μIU/ml.\n[SUBTITLE] Steroid hormone measurement by LC–MS/MS\n [SUBSECTION] As given in Table 2, aldosterone (ALD), 18‐hydroxycorticosterone(18‐OH‐B), cortisone, cortisol/hydrocortisone, 21‐deoxycortisol(21‐DOC), corticosterone, 11‐deoxycortisol, androstenedione (A2), 11‐deoxycorticosterone, total testosterone 17‐hydroxyprogesterone (17‐OHP), 17‐hydroxy pregnenolone, DHT, PROG, androsterone, pregnenolone, and HHEA‐S were assayed by LC–MS/MS.\nSteroid hormone level by LC–MS in control and PCOS patients\nAbbreviations: 17‐OHP, 17‐hydroxyprogesterone; 18‐OH‐B, 18‐hydroxycorticosterone; 21‐DOC, 21‐deoxycortisol; ALD, aldosterone; DEHA‐S, dehydroepiandrosterone sulfate; FAI, free androgen index.a\nP values were determined by two tailed student's t‐test.\nFirstly, 10 mg of each pure calibrator was dissolved in 10 ml methanol (Merck Millipore) to obtain a 1 mg/ml solution, which was stored at −80°C. The stock solutions were diluted with methanol and mixed to obtain preworking solutions containing the 18 calibrators (Cambridge Isotope Laboratories). Then, the samples were serially diluted to obtain a standard curve intermediate. The calibrator working solution was prepared with 5% methanol/water and stored in the refrigerator at −20°C. Secondly, each internal standard (IS) was weighed and dissolved in methanol to obtain solutions of different concentrations. Each internal standard mother solution was diluted and mixed with acetonitrile (Merck Millipore, USA) to obtain the internal standard working solution and stored at −20°C. Then, steroid hormones in serum were treated by protein precipitation combined with solid phase extraction (SPE). The specific steps are as follows: A 200 μl serum sample was mixed with 200 μl internal standard solution. The mixture was rotated for 3 min. Then, 500 μl of deionized water was added, followed by 1 min of shaking. After centrifugation at 15,000 × g for 10 min, the mixture was transferred to a well on a Waters Oasis Prime HLB μElution column plate for SPE. A 700 μl extraction supernatant was added to the 96‐well Oasis Prime HLB μElution column plate, the balance was activated with 200 μl methanol and 200 μl ddH2O, and the supernatant slowly flowed through the SPE plate and into the waste container by positive pressure. Then, 200 ml methanol/ddH2O (15:85, v/v) was added to the SPE plate in sequence. Then, 30 ml acetonitrile was added to the SPE plate twice, and the filtrate was collected. The filtrate was diluted with 60 μl methanol/ddH2O (5:95, v/v) and vortexed. The dilution filtrates were collected for LC–MS/MS analysis.\nTen microliters of filtrate from SPE was added to the Waters® ACQUITY UPLC® System – XevoTM TQS MS for LC–MS/MS analysis. The auto sampler temperature was 10°C. The binary mobile phase consisted of 2 mM ammonium acetate (Sigma‐Aldrich, USA) + 0.1% formic acid (Sigma‐Aldrich) + ddH2O (A) and 2 mM ammonium acetate+0.1% formic acid +methanol (B). The column temperature was kept at 45°C. The optimized parameters for MS/MS were as follows: capillary voltage, 3.2 kV; cone voltage, 50 V; source temperature, 150°C; desolvation temperature, 650°C; cone gas flow, 150 L/h; desolvation gas flow, 800 L/h; and collision gas flow, 0.15 ml/min. The steroid profile was based on electrospray ionization‐mass spectrometry (ESI‐MS) data combined with tandem mass spectrometry in the positive ion mode or negative ion mode. Quantitation by multiple reaction monitoring (MRM) analysis was performed in the positive ion mode for analytes and in the negative ion mode for DHEAs.\nAs given in Table 2, aldosterone (ALD), 18‐hydroxycorticosterone(18‐OH‐B), cortisone, cortisol/hydrocortisone, 21‐deoxycortisol(21‐DOC), corticosterone, 11‐deoxycortisol, androstenedione (A2), 11‐deoxycorticosterone, total testosterone 17‐hydroxyprogesterone (17‐OHP), 17‐hydroxy pregnenolone, DHT, PROG, androsterone, pregnenolone, and HHEA‐S were assayed by LC–MS/MS.\nSteroid hormone level by LC–MS in control and PCOS patients\nAbbreviations: 17‐OHP, 17‐hydroxyprogesterone; 18‐OH‐B, 18‐hydroxycorticosterone; 21‐DOC, 21‐deoxycortisol; ALD, aldosterone; DEHA‐S, dehydroepiandrosterone sulfate; FAI, free androgen index.a\nP values were determined by two tailed student's t‐test.\nFirstly, 10 mg of each pure calibrator was dissolved in 10 ml methanol (Merck Millipore) to obtain a 1 mg/ml solution, which was stored at −80°C. The stock solutions were diluted with methanol and mixed to obtain preworking solutions containing the 18 calibrators (Cambridge Isotope Laboratories). Then, the samples were serially diluted to obtain a standard curve intermediate. The calibrator working solution was prepared with 5% methanol/water and stored in the refrigerator at −20°C. Secondly, each internal standard (IS) was weighed and dissolved in methanol to obtain solutions of different concentrations. Each internal standard mother solution was diluted and mixed with acetonitrile (Merck Millipore, USA) to obtain the internal standard working solution and stored at −20°C. Then, steroid hormones in serum were treated by protein precipitation combined with solid phase extraction (SPE). The specific steps are as follows: A 200 μl serum sample was mixed with 200 μl internal standard solution. The mixture was rotated for 3 min. Then, 500 μl of deionized water was added, followed by 1 min of shaking. After centrifugation at 15,000 × g for 10 min, the mixture was transferred to a well on a Waters Oasis Prime HLB μElution column plate for SPE. A 700 μl extraction supernatant was added to the 96‐well Oasis Prime HLB μElution column plate, the balance was activated with 200 μl methanol and 200 μl ddH2O, and the supernatant slowly flowed through the SPE plate and into the waste container by positive pressure. Then, 200 ml methanol/ddH2O (15:85, v/v) was added to the SPE plate in sequence. Then, 30 ml acetonitrile was added to the SPE plate twice, and the filtrate was collected. The filtrate was diluted with 60 μl methanol/ddH2O (5:95, v/v) and vortexed. The dilution filtrates were collected for LC–MS/MS analysis.\nTen microliters of filtrate from SPE was added to the Waters® ACQUITY UPLC® System – XevoTM TQS MS for LC–MS/MS analysis. The auto sampler temperature was 10°C. The binary mobile phase consisted of 2 mM ammonium acetate (Sigma‐Aldrich, USA) + 0.1% formic acid (Sigma‐Aldrich) + ddH2O (A) and 2 mM ammonium acetate+0.1% formic acid +methanol (B). The column temperature was kept at 45°C. The optimized parameters for MS/MS were as follows: capillary voltage, 3.2 kV; cone voltage, 50 V; source temperature, 150°C; desolvation temperature, 650°C; cone gas flow, 150 L/h; desolvation gas flow, 800 L/h; and collision gas flow, 0.15 ml/min. The steroid profile was based on electrospray ionization‐mass spectrometry (ESI‐MS) data combined with tandem mass spectrometry in the positive ion mode or negative ion mode. Quantitation by multiple reaction monitoring (MRM) analysis was performed in the positive ion mode for analytes and in the negative ion mode for DHEAs.\n[SUBTITLE] Statistical analysis [SUBSECTION] SPSS 20.0 was used for analysis. The variables are presented as the mean ± standard deviation. Independent sample t tests were used for intergroup comparisons. The differences detected by LC–MS/MS and CLIA between the PCOS group and the control group were compared by interaction analysis. p < 0.05 was considered statistically significant. Spearman rank correlation analysis was used to analyze the correlation between total testosterone as detected by the LC–MS/MS method and by the CLIA method. The chi square test was used to compare the rates between groups. The Bland–Altman method was used to measure the consistency of the two methods. ROC curves were used to analyze the PCOS diagnostic efficiency.\nSPSS 20.0 was used for analysis. The variables are presented as the mean ± standard deviation. Independent sample t tests were used for intergroup comparisons. The differences detected by LC–MS/MS and CLIA between the PCOS group and the control group were compared by interaction analysis. p < 0.05 was considered statistically significant. Spearman rank correlation analysis was used to analyze the correlation between total testosterone as detected by the LC–MS/MS method and by the CLIA method. The chi square test was used to compare the rates between groups. The Bland–Altman method was used to measure the consistency of the two methods. ROC curves were used to analyze the PCOS diagnostic efficiency.", "A total of 114 patients with PCOS and 100 controls were recruited at Nanjing Maternal and Child Health Care Hospital. All procedures were performed with written informed consent. PCOS is diagnosed when at least two of the following criteria are met: (i) oligomenorrhoea and/or anovulation, (ii) clinical and/or biochemical hyperandrogenism and (iii) polycystic ovaries according to the Rotterdam 2003 criteria. None of the women took hormonal drugs for at least 3 months before our study.", "Hormone levels were tested in the Clinical Laboratory of Nanjing Maternal and Child Health Care Hospital. As given in Table 1, luteal hormone (LH), follitropin (FSH), prolaction (PRL), prpgesterone (PROG), cortisol, insulin, estradiol (E2), TT, dehydroepiandrosterone sulfate (DHEA) were measured by a Beckman automated chemiluminescence immunoanalyzer 7500 (Beckman). Sex hormone‐binding globulin (SHBG) were tested by Cobas 6000 (Roche). Uric acid, total protein, total cholesterol, glutamic‐pyruvic transaminase (GPT), glutamic oxiracetam transaminase (GPT), and fasting glucose were test by Beckman coulter AU5800 (Beckman). All the reagents required for the test were matching reagents, which were used according to the specified requirements. The remainng serum was stored at −80°C for LC–MS/MS analysis.\nBaseline characteristics and hormone levels by CLIA in control and PCOS patients\nAbbreviations: FSH, follicle‐stimulating hormone; GOT, glutamic oxiracetam transaminase; GPT, glutamic‐pyruvic transaminase; LH, luteal hormone; SHBG, sex hormone‐binding globulin.a\nP values were determined by two tailed student's t‐test.\nHomeostatic model assessment of insulin resistance (HOMA‐IR) is an indicator used to evaluate insulin resistance. HOMA‐IR is equal to fasting blood glucose (FBG) × fasting insulin (fin)/22.5. The FAI is equal to 100 × total testosterone (TT)/sex hormone‐binding globulin (SHBG). Insulin resistance (IR) was defined as HOMA‐IR≥2.14 and fin≥12.6 μIU/ml.", "As given in Table 2, aldosterone (ALD), 18‐hydroxycorticosterone(18‐OH‐B), cortisone, cortisol/hydrocortisone, 21‐deoxycortisol(21‐DOC), corticosterone, 11‐deoxycortisol, androstenedione (A2), 11‐deoxycorticosterone, total testosterone 17‐hydroxyprogesterone (17‐OHP), 17‐hydroxy pregnenolone, DHT, PROG, androsterone, pregnenolone, and HHEA‐S were assayed by LC–MS/MS.\nSteroid hormone level by LC–MS in control and PCOS patients\nAbbreviations: 17‐OHP, 17‐hydroxyprogesterone; 18‐OH‐B, 18‐hydroxycorticosterone; 21‐DOC, 21‐deoxycortisol; ALD, aldosterone; DEHA‐S, dehydroepiandrosterone sulfate; FAI, free androgen index.a\nP values were determined by two tailed student's t‐test.\nFirstly, 10 mg of each pure calibrator was dissolved in 10 ml methanol (Merck Millipore) to obtain a 1 mg/ml solution, which was stored at −80°C. The stock solutions were diluted with methanol and mixed to obtain preworking solutions containing the 18 calibrators (Cambridge Isotope Laboratories). Then, the samples were serially diluted to obtain a standard curve intermediate. The calibrator working solution was prepared with 5% methanol/water and stored in the refrigerator at −20°C. Secondly, each internal standard (IS) was weighed and dissolved in methanol to obtain solutions of different concentrations. Each internal standard mother solution was diluted and mixed with acetonitrile (Merck Millipore, USA) to obtain the internal standard working solution and stored at −20°C. Then, steroid hormones in serum were treated by protein precipitation combined with solid phase extraction (SPE). The specific steps are as follows: A 200 μl serum sample was mixed with 200 μl internal standard solution. The mixture was rotated for 3 min. Then, 500 μl of deionized water was added, followed by 1 min of shaking. After centrifugation at 15,000 × g for 10 min, the mixture was transferred to a well on a Waters Oasis Prime HLB μElution column plate for SPE. A 700 μl extraction supernatant was added to the 96‐well Oasis Prime HLB μElution column plate, the balance was activated with 200 μl methanol and 200 μl ddH2O, and the supernatant slowly flowed through the SPE plate and into the waste container by positive pressure. Then, 200 ml methanol/ddH2O (15:85, v/v) was added to the SPE plate in sequence. Then, 30 ml acetonitrile was added to the SPE plate twice, and the filtrate was collected. The filtrate was diluted with 60 μl methanol/ddH2O (5:95, v/v) and vortexed. The dilution filtrates were collected for LC–MS/MS analysis.\nTen microliters of filtrate from SPE was added to the Waters® ACQUITY UPLC® System – XevoTM TQS MS for LC–MS/MS analysis. The auto sampler temperature was 10°C. The binary mobile phase consisted of 2 mM ammonium acetate (Sigma‐Aldrich, USA) + 0.1% formic acid (Sigma‐Aldrich) + ddH2O (A) and 2 mM ammonium acetate+0.1% formic acid +methanol (B). The column temperature was kept at 45°C. The optimized parameters for MS/MS were as follows: capillary voltage, 3.2 kV; cone voltage, 50 V; source temperature, 150°C; desolvation temperature, 650°C; cone gas flow, 150 L/h; desolvation gas flow, 800 L/h; and collision gas flow, 0.15 ml/min. The steroid profile was based on electrospray ionization‐mass spectrometry (ESI‐MS) data combined with tandem mass spectrometry in the positive ion mode or negative ion mode. Quantitation by multiple reaction monitoring (MRM) analysis was performed in the positive ion mode for analytes and in the negative ion mode for DHEAs.", "SPSS 20.0 was used for analysis. The variables are presented as the mean ± standard deviation. Independent sample t tests were used for intergroup comparisons. The differences detected by LC–MS/MS and CLIA between the PCOS group and the control group were compared by interaction analysis. p < 0.05 was considered statistically significant. Spearman rank correlation analysis was used to analyze the correlation between total testosterone as detected by the LC–MS/MS method and by the CLIA method. The chi square test was used to compare the rates between groups. The Bland–Altman method was used to measure the consistency of the two methods. ROC curves were used to analyze the PCOS diagnostic efficiency.", "[SUBTITLE] Baseline characteristics and hormone levels between the control and PCOS groups [SUBSECTION] There was a significant difference in age between the groups (control vs. PCOS, 26.53 ± 3.30 vs. 24.18 ± 4.98, p < 0.001). However, there was no difference in height (control vs. PCOS, 1.62 ± 0.05 vs. 1.63 ± 0.05, p = 0.276), weight (control vs. PCOS, 63.15 ± 10.23 vs. 65.94 ± 13.54, p = 0.088) or BMI (control vs. PCOS, 24.01 ± 3.55 vs. 24.84 ± 4.57, p = 0.140). The main baseline characteristics of the subjects are given in Table 1.\nAs given in Table 1, there were differences between the control group and the PCOS group in luteal hormone (LH) (4.90 ± 4.21 vs. 13.86 ± 6.99, p < 0.001), insulin (8.58 ± 7.52 vs. 13.53 ± 14.97, p = 0.002), E2 (44.00 ± 15.54 vs. 75.19 ± 49.61, p < 0.001), HOMA‐IR (1.18 ± 2.20 vs. 3.15 ± 4.00, p = 0.034), TT (0.56 ± 0.27 vs. 0.75 ± 0.23, p < 0.001), FAI (0.019 ± 0.027 vs. 0.032 ± 0.027, p = 0.021), DHEA (6.89 ± 0.26 vs. 7.91 ± 3.16, p = 0.021), uric acid (223.19 ± 149.87 vs. 283.64 ± 151.93, p = 0.004), total protein (53.55 ± 34.52 vs. 62.91 ± 28.56, p = 0.033), total cholesterol (3.29 ± 2.14 vs. 4.13 ± 1.81, p = 0.002), GPT (17.02 ± 21.06 vs. 28.10 ± 29.84, p = 0.002), GOT (16.92 ± 15.04 vs. 22.28 ± 13.99, p = 0.008), and fasting glucose (4.04 ± 2.34 vs. 4.71 ± 1.64, p = 0.017) levels as measured by CLIA (Table 1).\nSince the LC–MS/MS method has higher accuracy and lower uncertainty, LC–MS/MS was used to detect steroid hormones in both groups. As given in Table 2, women with PCOS had higher androstenedione (5.04 ± 1.68 vs. 3.08 ± 1.28, p < 0.001), FAI (2.17 ± 1.64 vs. 0.94 ± 1.19, p < 0.001), TT (0.53 ± 0.21 vs. 0.30 ± 0.14, p < 0.001), and 17‐OHP (1.08 ± 0.71 vs. 0.69 ± 0.44, p < 0.001) levels than those in the controls, whereas there were no differences in serum concentrations of the steroids DHEA‐S, cortisol, cortisone, corticosterone, and 11‐deoxycortisol.\nThere was a significant difference in age between the groups (control vs. PCOS, 26.53 ± 3.30 vs. 24.18 ± 4.98, p < 0.001). However, there was no difference in height (control vs. PCOS, 1.62 ± 0.05 vs. 1.63 ± 0.05, p = 0.276), weight (control vs. PCOS, 63.15 ± 10.23 vs. 65.94 ± 13.54, p = 0.088) or BMI (control vs. PCOS, 24.01 ± 3.55 vs. 24.84 ± 4.57, p = 0.140). The main baseline characteristics of the subjects are given in Table 1.\nAs given in Table 1, there were differences between the control group and the PCOS group in luteal hormone (LH) (4.90 ± 4.21 vs. 13.86 ± 6.99, p < 0.001), insulin (8.58 ± 7.52 vs. 13.53 ± 14.97, p = 0.002), E2 (44.00 ± 15.54 vs. 75.19 ± 49.61, p < 0.001), HOMA‐IR (1.18 ± 2.20 vs. 3.15 ± 4.00, p = 0.034), TT (0.56 ± 0.27 vs. 0.75 ± 0.23, p < 0.001), FAI (0.019 ± 0.027 vs. 0.032 ± 0.027, p = 0.021), DHEA (6.89 ± 0.26 vs. 7.91 ± 3.16, p = 0.021), uric acid (223.19 ± 149.87 vs. 283.64 ± 151.93, p = 0.004), total protein (53.55 ± 34.52 vs. 62.91 ± 28.56, p = 0.033), total cholesterol (3.29 ± 2.14 vs. 4.13 ± 1.81, p = 0.002), GPT (17.02 ± 21.06 vs. 28.10 ± 29.84, p = 0.002), GOT (16.92 ± 15.04 vs. 22.28 ± 13.99, p = 0.008), and fasting glucose (4.04 ± 2.34 vs. 4.71 ± 1.64, p = 0.017) levels as measured by CLIA (Table 1).\nSince the LC–MS/MS method has higher accuracy and lower uncertainty, LC–MS/MS was used to detect steroid hormones in both groups. As given in Table 2, women with PCOS had higher androstenedione (5.04 ± 1.68 vs. 3.08 ± 1.28, p < 0.001), FAI (2.17 ± 1.64 vs. 0.94 ± 1.19, p < 0.001), TT (0.53 ± 0.21 vs. 0.30 ± 0.14, p < 0.001), and 17‐OHP (1.08 ± 0.71 vs. 0.69 ± 0.44, p < 0.001) levels than those in the controls, whereas there were no differences in serum concentrations of the steroids DHEA‐S, cortisol, cortisone, corticosterone, and 11‐deoxycortisol.\n[SUBTITLE] Diagnostic value of TT detected by LC–MS/MS for PCOS\n [SUBSECTION] The diagnostic values of hormones with significant differences between the control and PCOS groups were evaluated. As shown in Figure 1A, E2, TT, and DHES had good diagnostic value for PCOS detected by CLIA. Moreover, TT had the highest diagnostic potency for PCOS in the CLIA group, with an area under the curve (AUC) of 0.766. A2, TT and 17‐OHP tested by LC–MS/MS were further analyzed for their diagnostic value for PCOS. The AUCs of A2, TT, and 17‐OHP were 0.83, 0.851, and 0.714, respectively (Figure 1B). These results supported that TT had a better diagnostic value for PCOS not only detected by CLIA but also by LC–MS/MS. Moreover, TT detected by LC–MS/MS had a higher diagnostic value than TT detected by CLIA.\nROC curve for detecting the different hormones which were difference between NC and women with PCOS by CLIA and by LC–MS/MS. (A). AUC of E2, TT, and DHES by CLIA were 0.741, 0.766, and 0.569. (B). AUC of A2, TT, and 17‐OHP by LC–MS/MS were 0.831, 0.851, and 0.714.\nSince TT detected by LC–MS/MS had a higher diagnostic value than TT detected by CLIA, the agreement between the two methods was analyzed. The results suggested that the consistency between the results of the two methods was poor, and the stability of TT detected by LC–MS/MS was higher than that of TT detected by CLIA (Figure 2).\nComparison of TT concentration between CLIA and LC–MS/MS. (A). The concentration of TT between control women and PCOS women by CLIA or LC–MS/MS. The concentration of TT in LC–MS/MS was lower and more stable than the CLIA. ***indicated p < 0.001. (B). The Bland–Altman method was used to detected the consistency of TT between two methods. The showed poor consistency between CLIA and LC–MS/MS.\nThe diagnostic values of hormones with significant differences between the control and PCOS groups were evaluated. As shown in Figure 1A, E2, TT, and DHES had good diagnostic value for PCOS detected by CLIA. Moreover, TT had the highest diagnostic potency for PCOS in the CLIA group, with an area under the curve (AUC) of 0.766. A2, TT and 17‐OHP tested by LC–MS/MS were further analyzed for their diagnostic value for PCOS. The AUCs of A2, TT, and 17‐OHP were 0.83, 0.851, and 0.714, respectively (Figure 1B). These results supported that TT had a better diagnostic value for PCOS not only detected by CLIA but also by LC–MS/MS. Moreover, TT detected by LC–MS/MS had a higher diagnostic value than TT detected by CLIA.\nROC curve for detecting the different hormones which were difference between NC and women with PCOS by CLIA and by LC–MS/MS. (A). AUC of E2, TT, and DHES by CLIA were 0.741, 0.766, and 0.569. (B). AUC of A2, TT, and 17‐OHP by LC–MS/MS were 0.831, 0.851, and 0.714.\nSince TT detected by LC–MS/MS had a higher diagnostic value than TT detected by CLIA, the agreement between the two methods was analyzed. The results suggested that the consistency between the results of the two methods was poor, and the stability of TT detected by LC–MS/MS was higher than that of TT detected by CLIA (Figure 2).\nComparison of TT concentration between CLIA and LC–MS/MS. (A). The concentration of TT between control women and PCOS women by CLIA or LC–MS/MS. The concentration of TT in LC–MS/MS was lower and more stable than the CLIA. ***indicated p < 0.001. (B). The Bland–Altman method was used to detected the consistency of TT between two methods. The showed poor consistency between CLIA and LC–MS/MS.\n[SUBTITLE] Diagnostic value of FAI detected by LC–MS/MS for insulin resistance of PCOS\n [SUBSECTION] Insulin resistance is the most serious complication of PCOS, and the diagnostic value of steroid hormones detected by CLIA and LC–MS/MS for insulin resistance was tested. First, Pearson's correlation was used to test the steroid hormones associated with insulin resistance. A2 (R = 0.201, p = 0.003) and 17‐OHP (R = 0.232, p = 0.001) had better positive relationships with HOMA‐IR than TT, detected by both CLIA (R = 0.149, p = 0.03) and by LC–MS/MS (R = 0.149, p = 0.029) (Figure 3). The diagnostic efficacy of all differential indicators was analyzed for insulin resistance among PCOS patients. The AUCs of TT (CLIA), A2, TT (LC–MS/MS), and 17‐OHP were 0.587, 0.611, 0.602, and 0.572, respectively (Figure 4).\nTT in the body is either combined with SHBG or free in serum. It is generally thought that only free testosterone in serum, called FAI, could play an important role in PCOS.\n15\n The diagnostic efficacy of FAI by CLIA and LC–MS/MS was analyzed. According to the results, the AUCs of FAI detected by CLIA and LC–MS/MS were 0.788 and 0.798, respectively (Figure 4). Therefore, FAI detected by LC–MS/MS had the best diagnostic value for insulin resistance among PCOS patients.\nCorrelation between the different hormones which were difference between NC and women with PCOS and HOMA‐IR. (A). Correlation between TT detected by CLIA and HOMA‐IR in women with PCOS (R = 0.149, p = 0.03); (B) Correlation between testosterone detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.149, p = 0.029); (C). Correlation between androstenedione detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.201, p = 0.003); (D). Correlation between 17α‐hydroxyprogesterone detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.232, p = 0.001).\nAssessment of the diagnostic potency of androgens for insulin resistance in women with PCOS by CLIA and LC–MS/MS. AUC of TT by CLIA, TT by LC–MS/MS, A2 by LC–MS/MS, 17‐OHP by LC–MS/MS, FAI by CLIA and FAI by LC–MS/MS were 0.587, 0.602, 0.611, 0.572, 0.788, and 0.798.\nInsulin resistance is the most serious complication of PCOS, and the diagnostic value of steroid hormones detected by CLIA and LC–MS/MS for insulin resistance was tested. First, Pearson's correlation was used to test the steroid hormones associated with insulin resistance. A2 (R = 0.201, p = 0.003) and 17‐OHP (R = 0.232, p = 0.001) had better positive relationships with HOMA‐IR than TT, detected by both CLIA (R = 0.149, p = 0.03) and by LC–MS/MS (R = 0.149, p = 0.029) (Figure 3). The diagnostic efficacy of all differential indicators was analyzed for insulin resistance among PCOS patients. The AUCs of TT (CLIA), A2, TT (LC–MS/MS), and 17‐OHP were 0.587, 0.611, 0.602, and 0.572, respectively (Figure 4).\nTT in the body is either combined with SHBG or free in serum. It is generally thought that only free testosterone in serum, called FAI, could play an important role in PCOS.\n15\n The diagnostic efficacy of FAI by CLIA and LC–MS/MS was analyzed. According to the results, the AUCs of FAI detected by CLIA and LC–MS/MS were 0.788 and 0.798, respectively (Figure 4). Therefore, FAI detected by LC–MS/MS had the best diagnostic value for insulin resistance among PCOS patients.\nCorrelation between the different hormones which were difference between NC and women with PCOS and HOMA‐IR. (A). Correlation between TT detected by CLIA and HOMA‐IR in women with PCOS (R = 0.149, p = 0.03); (B) Correlation between testosterone detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.149, p = 0.029); (C). Correlation between androstenedione detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.201, p = 0.003); (D). Correlation between 17α‐hydroxyprogesterone detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.232, p = 0.001).\nAssessment of the diagnostic potency of androgens for insulin resistance in women with PCOS by CLIA and LC–MS/MS. AUC of TT by CLIA, TT by LC–MS/MS, A2 by LC–MS/MS, 17‐OHP by LC–MS/MS, FAI by CLIA and FAI by LC–MS/MS were 0.587, 0.602, 0.611, 0.572, 0.788, and 0.798.", "There was a significant difference in age between the groups (control vs. PCOS, 26.53 ± 3.30 vs. 24.18 ± 4.98, p < 0.001). However, there was no difference in height (control vs. PCOS, 1.62 ± 0.05 vs. 1.63 ± 0.05, p = 0.276), weight (control vs. PCOS, 63.15 ± 10.23 vs. 65.94 ± 13.54, p = 0.088) or BMI (control vs. PCOS, 24.01 ± 3.55 vs. 24.84 ± 4.57, p = 0.140). The main baseline characteristics of the subjects are given in Table 1.\nAs given in Table 1, there were differences between the control group and the PCOS group in luteal hormone (LH) (4.90 ± 4.21 vs. 13.86 ± 6.99, p < 0.001), insulin (8.58 ± 7.52 vs. 13.53 ± 14.97, p = 0.002), E2 (44.00 ± 15.54 vs. 75.19 ± 49.61, p < 0.001), HOMA‐IR (1.18 ± 2.20 vs. 3.15 ± 4.00, p = 0.034), TT (0.56 ± 0.27 vs. 0.75 ± 0.23, p < 0.001), FAI (0.019 ± 0.027 vs. 0.032 ± 0.027, p = 0.021), DHEA (6.89 ± 0.26 vs. 7.91 ± 3.16, p = 0.021), uric acid (223.19 ± 149.87 vs. 283.64 ± 151.93, p = 0.004), total protein (53.55 ± 34.52 vs. 62.91 ± 28.56, p = 0.033), total cholesterol (3.29 ± 2.14 vs. 4.13 ± 1.81, p = 0.002), GPT (17.02 ± 21.06 vs. 28.10 ± 29.84, p = 0.002), GOT (16.92 ± 15.04 vs. 22.28 ± 13.99, p = 0.008), and fasting glucose (4.04 ± 2.34 vs. 4.71 ± 1.64, p = 0.017) levels as measured by CLIA (Table 1).\nSince the LC–MS/MS method has higher accuracy and lower uncertainty, LC–MS/MS was used to detect steroid hormones in both groups. As given in Table 2, women with PCOS had higher androstenedione (5.04 ± 1.68 vs. 3.08 ± 1.28, p < 0.001), FAI (2.17 ± 1.64 vs. 0.94 ± 1.19, p < 0.001), TT (0.53 ± 0.21 vs. 0.30 ± 0.14, p < 0.001), and 17‐OHP (1.08 ± 0.71 vs. 0.69 ± 0.44, p < 0.001) levels than those in the controls, whereas there were no differences in serum concentrations of the steroids DHEA‐S, cortisol, cortisone, corticosterone, and 11‐deoxycortisol.", "The diagnostic values of hormones with significant differences between the control and PCOS groups were evaluated. As shown in Figure 1A, E2, TT, and DHES had good diagnostic value for PCOS detected by CLIA. Moreover, TT had the highest diagnostic potency for PCOS in the CLIA group, with an area under the curve (AUC) of 0.766. A2, TT and 17‐OHP tested by LC–MS/MS were further analyzed for their diagnostic value for PCOS. The AUCs of A2, TT, and 17‐OHP were 0.83, 0.851, and 0.714, respectively (Figure 1B). These results supported that TT had a better diagnostic value for PCOS not only detected by CLIA but also by LC–MS/MS. Moreover, TT detected by LC–MS/MS had a higher diagnostic value than TT detected by CLIA.\nROC curve for detecting the different hormones which were difference between NC and women with PCOS by CLIA and by LC–MS/MS. (A). AUC of E2, TT, and DHES by CLIA were 0.741, 0.766, and 0.569. (B). AUC of A2, TT, and 17‐OHP by LC–MS/MS were 0.831, 0.851, and 0.714.\nSince TT detected by LC–MS/MS had a higher diagnostic value than TT detected by CLIA, the agreement between the two methods was analyzed. The results suggested that the consistency between the results of the two methods was poor, and the stability of TT detected by LC–MS/MS was higher than that of TT detected by CLIA (Figure 2).\nComparison of TT concentration between CLIA and LC–MS/MS. (A). The concentration of TT between control women and PCOS women by CLIA or LC–MS/MS. The concentration of TT in LC–MS/MS was lower and more stable than the CLIA. ***indicated p < 0.001. (B). The Bland–Altman method was used to detected the consistency of TT between two methods. The showed poor consistency between CLIA and LC–MS/MS.", "Insulin resistance is the most serious complication of PCOS, and the diagnostic value of steroid hormones detected by CLIA and LC–MS/MS for insulin resistance was tested. First, Pearson's correlation was used to test the steroid hormones associated with insulin resistance. A2 (R = 0.201, p = 0.003) and 17‐OHP (R = 0.232, p = 0.001) had better positive relationships with HOMA‐IR than TT, detected by both CLIA (R = 0.149, p = 0.03) and by LC–MS/MS (R = 0.149, p = 0.029) (Figure 3). The diagnostic efficacy of all differential indicators was analyzed for insulin resistance among PCOS patients. The AUCs of TT (CLIA), A2, TT (LC–MS/MS), and 17‐OHP were 0.587, 0.611, 0.602, and 0.572, respectively (Figure 4).\nTT in the body is either combined with SHBG or free in serum. It is generally thought that only free testosterone in serum, called FAI, could play an important role in PCOS.\n15\n The diagnostic efficacy of FAI by CLIA and LC–MS/MS was analyzed. According to the results, the AUCs of FAI detected by CLIA and LC–MS/MS were 0.788 and 0.798, respectively (Figure 4). Therefore, FAI detected by LC–MS/MS had the best diagnostic value for insulin resistance among PCOS patients.\nCorrelation between the different hormones which were difference between NC and women with PCOS and HOMA‐IR. (A). Correlation between TT detected by CLIA and HOMA‐IR in women with PCOS (R = 0.149, p = 0.03); (B) Correlation between testosterone detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.149, p = 0.029); (C). Correlation between androstenedione detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.201, p = 0.003); (D). Correlation between 17α‐hydroxyprogesterone detected by LC–MS/MS and HOMA‐IR in women with PCOS (R = 0.232, p = 0.001).\nAssessment of the diagnostic potency of androgens for insulin resistance in women with PCOS by CLIA and LC–MS/MS. AUC of TT by CLIA, TT by LC–MS/MS, A2 by LC–MS/MS, 17‐OHP by LC–MS/MS, FAI by CLIA and FAI by LC–MS/MS were 0.587, 0.602, 0.611, 0.572, 0.788, and 0.798.", "At present, most clinical labs use the chemiluminescence method based on antigen antibody reaction (CLIA) or enzyme‐linked immunosorbent assay to detect androgens.\n8\n, \n9\n, \n10\n Due to the large amount of impurities in serum, the specificity of the test results is not high. This study compared the application of LC–MS/MS and CLIA to detect TT in PCOS patients and healthy women control, which provided new clinical data for the study of female hyperandrogenemia.\nAs expected, the mean TT level, FAI, measured by both LC–MS/MS and CLIA, was significantly higher in the PCOS group than in the control group. In women with PCOS, DHEA detected by CLIA was elevated compared with controls. Interestingly, the levels of androstenedione and 17‐OHP detected by LC–MS/MS were elevated in women with PCOS compared to control women. It is well known that TT is not the only androgen elevated in women with PCOS. There is a view that androgens are not equal in women with PCOS, and testosterone has a much greater affinity with the androgen receptor than other androgens.\n16\n A study carried out by Keefe C, in which LC–MS/MS was used to measure 13 steroids, found that women with PCOS had higher testosterone, androstenedione, and 17‐OH progesterone levels than controls, but no differences in DHEAS, cortisol, corticosterone or their 11‐deoxy precursors.\n17\n In Cao, Z et al.'s study, the authors developed a simultaneous quantitation LC–MS/MS method for testosterone (T), androstenedione (A4), dehydroepiandrosterone sulfate (DHEAS), dihydrotestosterone (DHT), and 17‐hydroxyprogesterone (17‐OHP) in female serum, which was validated with good performance. The results provided better diagnostic power for PCOS with the combination of T, A4, DHEAS, and DHT measurements.\n18\n Another study also confirmed that T, A4, and DHT together might contribute to a more accurate diagnosis of PCOS.\n19\n\n\nAmong women with PCOS, increased HOMA‐IR was also observed. These results were consistent with two other studies reported by Stepto NK and Kogure GS.\n20\n, \n21\n Stepto NK reported that the IR of women with PCOS was higher than that of the BMI‐matched control group. IR was present in 62% of the overweight control group and 95% of the overweight PCOS group. Kogure GS found that women with PCOS had higher serum levels of total testosterone and androstenedione and higher HOMA‐IR scores than the control group.\nInsulin resistance is the pathophysiological basis of PCOS. HOMA‐IR is one of the indices used to evaluate insulin resistance. It has been reported that approximately 70% of women with PCOS are insulin resistant.\n7\n In our study, testosterone detected by CLIA and androstenedione, testosterone and 17 α‐hydroxyprogesterone detected by LC–MS/MS were positively correlated with HOMA‐IR. FAI detected by LC–MS/MS had the highest diagnostic value, with an ROC curve of 0.798. Sezai Sahmay found that BMI and free testosterone were positively correlated with HOMA‐IR.\n22\n Soulmaz Shorakae reported that testosterone and FAI were both correlated with insulin resistance in women with PCOS.\n6\n A study reported that insulin resistance was highly related to serum free testosterone but weakly related to total testosterone or other androgens.\n23\n A recent study has shown that insulin clearance was significantly damaged in women with PCOS compared with controls.\n24\n Insulin can stimulate ovarian theca cells to secrete androgens.\n25\n IR stimulates the production of androgens in the ovaries and reduces SHBG levels in the liver, which increases free testosterone.\n26\n It is believed that although the diagnostic criteria of PCOS do not distinguish between different androgens in detail, androgens are not equal in action. Testosterone and DHT have a much greater affinity for the androgen receptor than androstenedione and DHEA.\n16\n\n\nIn this study, TT detected by LC–MS/MS had the highest diagnostic efficacy, with an AUC of 0.851, which was better than the AUC of 0.766 for CLIA. The consistency between the two methods was poor, which suggested that there were differences between them. Joëlle Taieb found that the immunoassay method was unreliable for the measurement of serum from children and women, especially when their testosterone concentration was low (<1.7 nmoL/L).\n21\n Urszula Ambroziak concluded that LC–MS/MS was more reliable in the measurement of serum 17‐OHP and androgens than immunoassays in women with hyperandrogenism.\n22\n Željko Debeljak assessed the analytical bias of six serum steroid hormones using LC–MS/MS, and the results showed that there was a large difference between all immunoassays and the LC–MS/MS assay.\n27\n In a longitudinal study of healthy children, the author demonstrated the importance of testing serum testosterone by LC–MS/MS, especially at low levels in children.\n28\n Due to its high specificity and high sensitivity, LC–MS/MS can produce more accurate and reliable detection results, which are more suitable for complex biological samples, and its sensitivity can reach the pg/ml\n10\n, \n11\n, \n12\n level,\n29\n which is especially useful for low‐concentration hormone detection, so it can provide better services for clinical diagnosis and is increasingly favored by clinical detection.\nAlthough the LC–MS/MS method requires an expensive apparatus and long operation procedures, it has been used as a reference method for hormone measurements by professional societies. In conclusion, our study revealed a close relationship between HA and IR by LC–MS/MS measurement of TT.", "This project was supported by the National Natural Science Foundation of China (Grant Nos. 81901446, 81901555, and 81971351) and the Fund of Nanjing Health and Health Committee (YKK18161).", "The authors have declared that no competing interests exist.", "The experimental protocols were approved by the Ethics Committee of the Nanjing Maternity and Child Health Care Hospital. The methods were performed in accordance with the approved guidelines by the Ethics Committee of the Nanjing Maternity and Child Health Care Hospital." ]
[ null, "materials-and-methods", null, null, null, null, "results", null, null, null, "discussion", null, "COI-statement", null ]
[ "diagnostic power", "FAI", "insulin resistance", "PCOS", "TT" ]
Clinical features and management of snake bites in 70 dogs in Korea.
36259100
Snakebites remain a devastating and life-threatening environmental hazard. While the management of snakebites has been well described in humans, few clinical data and guidelines exist for dogs, especially in Korea.
BACKGROUND
The medical records of 72 dogs that presented to three animal hospitals from June 2008 to July 2021 were reviewed; among these, 70 dogs that met the inclusion criteria were enrolled. Their signalment, history, clinical signs, physical examination, blood analysis, treatment, and prognosis were also evaluated.
METHODS
Of 70 dog owners, 35 (50%) witnessed the bite, with a mean time between bite and hospital presentation of 9.7 ± 4.1 h in 58 dogs. Blood smears were evaluated in 45 dogs, of which 28 (62%) showed echinocytosis. Anemia and acute kidney injury were found in 21 (29%) and 2 dogs (3%), respectively. A total of 37 dogs (53%) were hospitalized, 5 (7%) of which died.
RESULTS
The most significant finding was the high prevalence of echinocytosis. The data from this retrospective study could inform the management of dogs bitten by snakes in Korea.
CONCLUSIONS
[ "Humans", "Dogs", "Animals", "Snake Bites", "Retrospective Studies", "Snakes", "Republic of Korea", "Prevalence", "Antivenins", "Dog Diseases" ]
9715381
INTRODUCTION
Snakes are carnivorous reptiles with elongated bodies and no legs. Of the approximately 3,000 snake species worldwide, 15% are considered venomous [1]. Korea has 10 types of non-venomous snakes and 4 types of venomous snakes. The venomous snakes include Gloydius brevicaudus, G. saxatilis, G. ussuriensis, and Rhabdophis tigrinus [2]. According to the World Health Organization (WHO), 5.4 million snakebites occur annually in people, of which 2.7 million are venomous, with 81,000–138,000 deaths [1]. An estimated 150,000 animals, primarily dogs and cats, are bitten by snakes in the United States annually [3]. Although mortality in humans after snakebites in the United States is low (0.06%), the reported canine mortality rates range from 1% to 30% [45]. In Korea, 200–600 cases of snake bites occur in humans annually, with 60% of snakes belonging to the genus Gloydius [67]. However, retrospective studies on snake bites in dogs in Korea are scarce. Mamushi is the most common venomous snake in Korea and is also found in Japan and China [8]. This pit viper is found in a variety of colors and is small (approximately 60 cm); thus, its attack range is only approximately 30 cm. The canine teeth (fangs) are approximately 5 mm long and have very thin tips. Snakes of this genus tend to live near rivers, ponds, and rice fields and are active during the day in spring and autumn and during the night in summer [9]. In Korea, mamushi are found nationwide, except on Jeju Island, and are reportedly more frequently encountered at low altitudes [10]. Owing to these characteristics, the risk of snakebite increases in dogs that frequently walk outdoors. R. tigrinus is also common in Korea, growing to 1–1.5 meters and sharing the same habitats as mamushi. While this species has long been considered non-venomous, recent studies have revealed that the venom has high toxicity and contains the same venom components as those in mamushi [29]. Snake envenomation causes a combination of local and systemic clinical signs with considerable variations, depending on the snake species. Many of these human-specific signs are similar to those observed in dogs [11]. Local manifestations such as edema at the bite site, infection, and tissue necrosis are the most common. The systemic manifestations include vomiting, nausea, dizziness, rhabdomyolysis due to muscle necrosis, renal function damage, and various other clinical conditions that lead to death. [1112]. Mamushi venom is characterized by hemorrhagic activity and induces widespread bleeding in humans [12]. Only one case report has described the effects of mamushi envenomation in dog [13]. In this study, the bitten dog presented with extensive edema and bleeding but no other systemic disorders [13]. Few studies with small sample sizes have described the clinical features and management of snakebites in dogs in Korea. Therefore, the purpose of this study was to describe the signalment, history, clinical presentation, laboratory values, treatment, and prognosis of dogs bitten by snakes in Korea.
null
null
RESULTS
The records of 72 dogs assumed to have been bitten by a snake were reviewed at the three hospitals from June 2008 to July 2021; two dogs were excluded due to the absence of fang marks. Thus, the analysis included a total of 70 animals. The dogs represented 23 breeds including mixed (11), Dachshund (7), Jindo (7), Maltese (7), Poodle (6), Yorkshire Terrier (6), French Bulldog (3), Cocker Spaniel (2), German Shepherd (2), Golden Retriever (2), Labrador Retriever (2), Pungsan dog (2), Shiba Inu (2), Border Collie (1), Beagle (1), Cane Corso (1), Chihuahua (1), Miniature Schnauzer (1), Pomeranian (1), Shih-tzu (1), Siberian Husky (1), Spitz (1), and Wolf dog (1). Overall, 20% (14) of dogs were intact males, 33% (23) were castrated males, 20% (14) were intact females, and 27% (19) were spayed females. The sex of the dogs did not differ significantly between the survivor and non-survivor groups (p = 0.149). The median age was 48 mon (range, 2–156 mon) and median weight was 12.6 kg (range, 2–50.4 kg). Bites were observed by 50% (35) of the owners, with a median time from observation of the clinical sign onset to hospital presentation of 9.7 h (range, 1–96 h). The bite wounds were localized to the face (74%, 48/65), body (3%, 2/65), and legs (23%, 15/65). The bite location was not significantly associated with survival (p = 0.072). Four clinical signs were observed on presentation: edema, erythema, continuous bleeding, and cyanosis. Of the 65 dogs with clinical signs, 64 (98%) had edema, 54 (83%) had erythema, 6 (9%) had continuous bleeding, and 3 (5%) had cyanosis. On admission, the mean temperature was 39.1°C ± 0.6°C (64 dogs; range, 37.4°C–40.1°C) and mean heart rate was 148.5 ± 37.9 bpm (58 dogs; range, 84–240 bpm). Panting was observed in 22 dogs (40%) during physical examination, and the mean respiratory rate was 46.5 ± 23.8 breaths/min in 33 dogs. Normal skin turgor was observed in 28 dogs, whereas a loss of turgor was observed in 8 dogs. Doppler blood pressure recorded in 54 dogs showed a median arterial blood pressure of 155 mmHg (range, 70–230 mmHg). The results of complete blood count and serum chemistry tests are summarized in Table 1. A complete blood count was obtained for 64 dogs (91%), 21 of which (29%) had anemia (median hematocrit of the 64 dogs, 52%; range, 22.2%–78%). Hemoglobin levels and platelet counts were within the reference ranges. The median white blood cell count was 14.3 K/µL (range, 6.8–43.5 K/µL), which was close to the upper limit of the reference range (reference range, 5.1–16.8 K/µL). The plasma lactate concentration on admission was available for 51 dogs, with a median of 2.7 mmol/L (range, 0.9–6.9 mmol/L) (reference range, 0.5–2.5 mmol/L). Serum biochemistry panels were performed in 64 dogs (91%), 2 of which were diagnosed with acute kidney injury. C-reactive protein concentrations were measured in 39 dogs, with increased levels noted in 25 dogs (median, 24 mg/L; range, 10–186 mg/L; reference range, 0–20 mg/L). Blood smear evaluation was performed in 45 dogs, 28 of which (62%) had echinocytosis. The coagulation parameters PT and aPTT were tested in 62 dogs; 10 (16%) had prolonged PT and 8 (13%) had prolonged aPTT. D-dimer levels were tested in 39 dogs, with a median concentration of 0.7 mg/L (range, 0.1–5.9 mg/L); 26 (67%) had increased levels of D-dimer. CBC, complete blood count; WBC, white blood cell; BUN, blood urea nitrogen; ALT, alanine aminotransferase; ALP, alkaline phosphatase; CRP, C-reactive protein. The types of treatment received by the dogs are summarized in Table 2. Out of 70 dogs, 53 (76%) received more than one vial of antivenin and 17 (24%) did not receive antivenom treatment. A total of 50 dogs received analgesics and intravenous fluid support. The most frequently used analgesics were tramadol (41%) and hydromorphone (14%). Regarding fluid therapy, normal saline was administered to 41% of dogs, followed by Plasma-Lyte (17%). The most frequent administration rate was 2.5–5 mL/kg/h. Additionally, 51 of 70 dogs (73%) received chlorpheniramine, 32 (46%) received glucocorticoids, and 62 (89%) received antibiotics. The administration of antivenin, glucocorticoids, and antibiotics was associated with increased survival (p = 0.008, p = 0.030, and p = 0.007, respectively). Survival and antihistamine use were not significantly related (p = 0.172). TLK, tramadol lidocaine ketamine; CRI, constant rate infusion; N/S, normal saline; H/S: Hartmann’s solution; DW, dextrose water. The median hospitalization time was 2.5 days in 68 dogs. Of the 70 dogs, 37 (53%) survived until discharge, 5 (7%) died, and 28 were lost to follow-up. Among 42 hospitalized dogs with follow-up data, 37 (88%) survived to discharge and 5 (12%) died. The prognosis of the dogs was assessed and the dogs were divided into two groups depending on the administration of antivenin. Of 53 dogs treated with antivenin, 31 (58%) survived to discharge, 3 (6%) died, and 19 (36%) were limited to follow-up. Of 17 dogs not treated with antivenin, 6 (35%) survived to discharge, 2 (12%) died, and 9 (53%) were limited to follow-up.
null
null
[ "Statistical analysis" ]
[ "Descriptive data are presented as mean ± SD for normally distributed data and median (range) for skewed data. The χ2 test was performed to compare categorical data between survivors and non-survivors. Sex, bite location, and treatment were also compared between the groups. Statistical significance was set at a p value of < 0.05. Analyses were performed using IBM SPSS Statistics for Windows, version 25.0 (IBM Corp., USA)." ]
[ null ]
[ "INTRODUCTION", "MATERIALS AND METHODS", "Statistical analysis", "RESULTS", "DISCUSSION" ]
[ "Snakes are carnivorous reptiles with elongated bodies and no legs. Of the approximately 3,000 snake species worldwide, 15% are considered venomous [1]. Korea has 10 types of non-venomous snakes and 4 types of venomous snakes. The venomous snakes include Gloydius brevicaudus, G. saxatilis, G. ussuriensis, and Rhabdophis tigrinus [2]. According to the World Health Organization (WHO), 5.4 million snakebites occur annually in people, of which 2.7 million are venomous, with 81,000–138,000 deaths [1]. An estimated 150,000 animals, primarily dogs and cats, are bitten by snakes in the United States annually [3]. Although mortality in humans after snakebites in the United States is low (0.06%), the reported canine mortality rates range from 1% to 30% [45]. In Korea, 200–600 cases of snake bites occur in humans annually, with 60% of snakes belonging to the genus Gloydius [67]. However, retrospective studies on snake bites in dogs in Korea are scarce.\nMamushi is the most common venomous snake in Korea and is also found in Japan and China [8]. This pit viper is found in a variety of colors and is small (approximately 60 cm); thus, its attack range is only approximately 30 cm. The canine teeth (fangs) are approximately 5 mm long and have very thin tips. Snakes of this genus tend to live near rivers, ponds, and rice fields and are active during the day in spring and autumn and during the night in summer [9]. In Korea, mamushi are found nationwide, except on Jeju Island, and are reportedly more frequently encountered at low altitudes [10]. Owing to these characteristics, the risk of snakebite increases in dogs that frequently walk outdoors. R. tigrinus is also common in Korea, growing to 1–1.5 meters and sharing the same habitats as mamushi. While this species has long been considered non-venomous, recent studies have revealed that the venom has high toxicity and contains the same venom components as those in mamushi [29].\nSnake envenomation causes a combination of local and systemic clinical signs with considerable variations, depending on the snake species. Many of these human-specific signs are similar to those observed in dogs [11]. Local manifestations such as edema at the bite site, infection, and tissue necrosis are the most common. The systemic manifestations include vomiting, nausea, dizziness, rhabdomyolysis due to muscle necrosis, renal function damage, and various other clinical conditions that lead to death. [1112]. Mamushi venom is characterized by hemorrhagic activity and induces widespread bleeding in humans [12]. Only one case report has described the effects of mamushi envenomation in dog [13]. In this study, the bitten dog presented with extensive edema and bleeding but no other systemic disorders [13].\nFew studies with small sample sizes have described the clinical features and management of snakebites in dogs in Korea. Therefore, the purpose of this study was to describe the signalment, history, clinical presentation, laboratory values, treatment, and prognosis of dogs bitten by snakes in Korea.", "We retrospectively reviewed the medical records of 72 dogs admitted with snakebites to the Kyungpook National Veterinary Medical Teaching Hospital, Haemaru Animal Referral Hospital, and Daegu Animal Medical Center from June 2008 to July 2021. This study included dogs in which two fang marks were observed at the wound site. Two dogs with bite wounds of unclear origin were excluded from the analysis.\nThe data collected from each record included signalment, history including witnessed snakebite and time since the snakebite, clinical signs, and physical examination records on presentation. Routine test results, including complete blood count, serum chemical profile, coagulation profile, and blood smear examination, were also recorded. The numbers of vials of antivenom as well as the types of analgesics, fluids, and additional medications were recorded. We also collected data on the length of hospitalization and outcomes for all animals. Dogs were diagnosed with anemia if the hematocrit value was < 35% [14]; acute kidney injury was defined as an increase in the serum creatinine concentration of 0.3 mg/dL from baseline within 48 h [15]. Increases in prothrombin time (PT) and activated partial thromboplastin time (aPTT) were defined as 150% of the upper limit of the reference range [16]. Dogs discharged alive or referred to another hospital were considered survivors, whereas those that died during hospitalization were defined as non-survivors.\n[SUBTITLE] Statistical analysis [SUBSECTION] Descriptive data are presented as mean ± SD for normally distributed data and median (range) for skewed data. The χ2 test was performed to compare categorical data between survivors and non-survivors. Sex, bite location, and treatment were also compared between the groups. Statistical significance was set at a p value of < 0.05. Analyses were performed using IBM SPSS Statistics for Windows, version 25.0 (IBM Corp., USA).\nDescriptive data are presented as mean ± SD for normally distributed data and median (range) for skewed data. The χ2 test was performed to compare categorical data between survivors and non-survivors. Sex, bite location, and treatment were also compared between the groups. Statistical significance was set at a p value of < 0.05. Analyses were performed using IBM SPSS Statistics for Windows, version 25.0 (IBM Corp., USA).", "Descriptive data are presented as mean ± SD for normally distributed data and median (range) for skewed data. The χ2 test was performed to compare categorical data between survivors and non-survivors. Sex, bite location, and treatment were also compared between the groups. Statistical significance was set at a p value of < 0.05. Analyses were performed using IBM SPSS Statistics for Windows, version 25.0 (IBM Corp., USA).", "The records of 72 dogs assumed to have been bitten by a snake were reviewed at the three hospitals from June 2008 to July 2021; two dogs were excluded due to the absence of fang marks. Thus, the analysis included a total of 70 animals. The dogs represented 23 breeds including mixed (11), Dachshund (7), Jindo (7), Maltese (7), Poodle (6), Yorkshire Terrier (6), French Bulldog (3), Cocker Spaniel (2), German Shepherd (2), Golden Retriever (2), Labrador Retriever (2), Pungsan dog (2), Shiba Inu (2), Border Collie (1), Beagle (1), Cane Corso (1), Chihuahua (1), Miniature Schnauzer (1), Pomeranian (1), Shih-tzu (1), Siberian Husky (1), Spitz (1), and Wolf dog (1). Overall, 20% (14) of dogs were intact males, 33% (23) were castrated males, 20% (14) were intact females, and 27% (19) were spayed females. The sex of the dogs did not differ significantly between the survivor and non-survivor groups (p = 0.149). The median age was 48 mon (range, 2–156 mon) and median weight was 12.6 kg (range, 2–50.4 kg). Bites were observed by 50% (35) of the owners, with a median time from observation of the clinical sign onset to hospital presentation of 9.7 h (range, 1–96 h).\nThe bite wounds were localized to the face (74%, 48/65), body (3%, 2/65), and legs (23%, 15/65). The bite location was not significantly associated with survival (p = 0.072). Four clinical signs were observed on presentation: edema, erythema, continuous bleeding, and cyanosis. Of the 65 dogs with clinical signs, 64 (98%) had edema, 54 (83%) had erythema, 6 (9%) had continuous bleeding, and 3 (5%) had cyanosis. On admission, the mean temperature was 39.1°C ± 0.6°C (64 dogs; range, 37.4°C–40.1°C) and mean heart rate was 148.5 ± 37.9 bpm (58 dogs; range, 84–240 bpm). Panting was observed in 22 dogs (40%) during physical examination, and the mean respiratory rate was 46.5 ± 23.8 breaths/min in 33 dogs. Normal skin turgor was observed in 28 dogs, whereas a loss of turgor was observed in 8 dogs. Doppler blood pressure recorded in 54 dogs showed a median arterial blood pressure of 155 mmHg (range, 70–230 mmHg).\nThe results of complete blood count and serum chemistry tests are summarized in Table 1. A complete blood count was obtained for 64 dogs (91%), 21 of which (29%) had anemia (median hematocrit of the 64 dogs, 52%; range, 22.2%–78%). Hemoglobin levels and platelet counts were within the reference ranges. The median white blood cell count was 14.3 K/µL (range, 6.8–43.5 K/µL), which was close to the upper limit of the reference range (reference range, 5.1–16.8 K/µL). The plasma lactate concentration on admission was available for 51 dogs, with a median of 2.7 mmol/L (range, 0.9–6.9 mmol/L) (reference range, 0.5–2.5 mmol/L). Serum biochemistry panels were performed in 64 dogs (91%), 2 of which were diagnosed with acute kidney injury. C-reactive protein concentrations were measured in 39 dogs, with increased levels noted in 25 dogs (median, 24 mg/L; range, 10–186 mg/L; reference range, 0–20 mg/L). Blood smear evaluation was performed in 45 dogs, 28 of which (62%) had echinocytosis. The coagulation parameters PT and aPTT were tested in 62 dogs; 10 (16%) had prolonged PT and 8 (13%) had prolonged aPTT. D-dimer levels were tested in 39 dogs, with a median concentration of 0.7 mg/L (range, 0.1–5.9 mg/L); 26 (67%) had increased levels of D-dimer.\nCBC, complete blood count; WBC, white blood cell; BUN, blood urea nitrogen; ALT, alanine aminotransferase; ALP, alkaline phosphatase; CRP, C-reactive protein.\nThe types of treatment received by the dogs are summarized in Table 2. Out of 70 dogs, 53 (76%) received more than one vial of antivenin and 17 (24%) did not receive antivenom treatment. A total of 50 dogs received analgesics and intravenous fluid support. The most frequently used analgesics were tramadol (41%) and hydromorphone (14%). Regarding fluid therapy, normal saline was administered to 41% of dogs, followed by Plasma-Lyte (17%). The most frequent administration rate was 2.5–5 mL/kg/h. Additionally, 51 of 70 dogs (73%) received chlorpheniramine, 32 (46%) received glucocorticoids, and 62 (89%) received antibiotics. The administration of antivenin, glucocorticoids, and antibiotics was associated with increased survival (p = 0.008, p = 0.030, and p = 0.007, respectively). Survival and antihistamine use were not significantly related (p = 0.172).\nTLK, tramadol lidocaine ketamine; CRI, constant rate infusion; N/S, normal saline; H/S: Hartmann’s solution; DW, dextrose water.\nThe median hospitalization time was 2.5 days in 68 dogs. Of the 70 dogs, 37 (53%) survived until discharge, 5 (7%) died, and 28 were lost to follow-up. Among 42 hospitalized dogs with follow-up data, 37 (88%) survived to discharge and 5 (12%) died. The prognosis of the dogs was assessed and the dogs were divided into two groups depending on the administration of antivenin. Of 53 dogs treated with antivenin, 31 (58%) survived to discharge, 3 (6%) died, and 19 (36%) were limited to follow-up. Of 17 dogs not treated with antivenin, 6 (35%) survived to discharge, 2 (12%) died, and 9 (53%) were limited to follow-up.", "To the best of our knowledge, this is the first large-scale study conducted in Korea to assess the clinical features of dogs bitten by snakes. Our results showed that there was no predominance of sex. In humans, men are approximately 50% more likely to be bitten than women due to their occupational characteristics (farmers, gardeners, and security guards) [17]. These differences can be attributed to varying species-specific behaviors between dogs and humans. Furthermore, one study reported that young, mature dogs of medium to large breeds are most commonly bitten by snakes [18]. This finding might be explained by the fact that some of the dogs enrolled in this study were guard dogs and tended to attack and provoke snakes to bite [18]. However, contrasting results were observed in this study. We found that small breeds were more frequently bitten than large breeds, possibly because small and toy breeds are among the most prevalent breeds in South Korea [19].\nIn the present study, snakebites were mostly observed in the head region or on the limbs, findings consistent with those of other reports [182021]. A previous study reported that, of 96 bites, 78 (81.2%) occurred in the head region and 21 (21.9%) on the limbs [20], similar to our results observed in 70 dogs (74% and 23%, respectively). These results might indicate that dogs are prone to intentional contact with vipers, as opposed to humans, who are almost exclusively bitten accidentally [121]. Moreover, due to these behavioral characteristics in dogs, most snakebites occur in the distal parts of the limbs or head region.\nMamushi venom contains phospholipase A2, hyaluronidases, proteases, and other enzymes. In humans, these enzymes induce erythema, local pain, and swelling at the bite site. Mamushi venom also contains hemorrhage factor I or II, which cause a significant increase in platelet aggregation, resulting in extensive ecchymosis and gastrointestinal bleeding. Hypotension can occur due to severe swelling. In these cases, increased serum levels of creatinine phosphokinase and blood levels of myoglobin due to rhabdomyolysis can cause acute renal failure [21222]. In the current study, 98% of dogs had edema, 83% had erythema, and only a small number of dogs presented persistent bleeding, cyanosis, and acute kidney injury. These results may be of non-venomous snakebites or “dry” bites, which induce insufficient envenomation. While approximately 25% of bites by pit vipers in humans are dry, there are no studies on the incidence of dry bites in dogs [23]. In the present study, the number of dry bites could not be fully estimated because we did not record whether a patient was envenomated since envenomation is generally determined by witnessing snakebites, snake species, clinical signs, and typical fang marks.\nThe hematologic findings in the current study suggest an inflammatory reaction caused by the bites, characterized by median white blood cell counts close to the upper limit of the reference range and C-reactive protein exceeding the reference range. These results are similar to those of a previous study that suggested stress as a contributing factor [24]. Echinocytosis was another hematological abnormality observed in 28 dogs (62%). Echinocytosis is the most common hematologic abnormality in dogs envenomed by snakes from the subfamily Crotalinae [2025]. Previous studies showed that echinocytosis is caused by phospholipase A2, a component of snake venom with a dose-dependent effect on the red blood cell membrane [2627]. Although no relationship between echinocytosis and mamushi envenomation has been reported, echinocytosis appears to be a reasonably good marker in dogs because the venom of this snake also contains phospholipase A2. Thrombocytopenia and acute kidney injury are common conditions following hemorrhagic envenomation in both humans and dogs [23521232829]. However, in our study, the mean platelet counts were within the reference ranges, and only two dogs had acute kidney injury. Moreover, blood analysis was performed only once at presentation, which might have contributed to these results. Further research is needed to evaluate the complications of snakebites in Korea, and serial blood sampling after hospitalization is recommended.\nA standard treatment protocol for snake envenomation is lacking for dogs; however, the only accepted treatment is the administration of antivenom along with supportive care consisting of intravenous crystalloid fluid therapy and pain control [2330]. Antivenom limits clinical signs and reverses coagulopathy [233132]. In the current study, 70 dogs (76%) were administered at least one vial of antivenom; additional antivenin was administered to dogs with persistent clinical signs as there is no evidence-based protocol available for antivenom administration or dosage in dogs [23]. Regarding pain control, opioid agonists, especially fentanyl, are usually recommended because they can provide excellent short-term analgesia. And morphine should be used with caution owing to the risk of histamine release [323]. However, tramadol was used most frequently in the present study, possibly due to its non-narcotic properties and the fact that it is legally available and widely used in the Korean veterinary community. Nonetheless, there are some controversies regarding the efficacy of tramadol in dogs [33]. As snakebites are usually associated with pain [31125], warranted analgesics should be used. Lastly, while the recommended intravenous fluids include isotonic crystalloid solutions such as lactated Ringer’s solution, 0.9% sodium chloride, or Plasma-Lyte solution [23], most of our patients which administered IV fluids treated with isotonic crystalloid solutions.\nAlthough there remain controversies regarding the use of glucocorticoids and antihistamines for snake envenomation [2325], they were administered to 46% and 73% of dogs, respectively. Some authors favor the use of glucocorticoids in snake envenomation due to their anti-inflammatory effects, which might reduce pain and swelling, whereas others suggest that glucocorticoids might slow and reduce antivenom activity, increase the risk of infection, and interfere with the normal healing process [2334]. Most of the literature on envenomation in humans does not recommend the routine use of antihistamines because of their potential hypotensive adverse effects; moreover, they are only indicated for the treatment of anaphylaxis [2335]. However, two retrospective studies on dogs reported increased survival rates with diphenhydramine administration after snakebites [525]. All dogs that died in the present study had received both glucocorticoids and antihistamines; therefore, these drugs may worsen the clinical status due to the complications described above. However, in our study, glucocorticoid administration was positively associated with survival. Currently, there is no evidence for the effectiveness of glucocorticoids and antihistamines in snake envenomation; thus, further research is warranted.\nThe administration of systemic antimicrobials to patients with snake envenomation remains controversial in both human and veterinary medicine [2336]. In human medicine, prophylactic antibiotics are not indicated as initial therapy because the incidence of infection is low following pit viper bites, likely due to the bactericidal effects of the venom itself [3637]. Hence, antibiotics are only recommended when there is clinical and microbiological evidence of wound infection [36]. In the past veterinary studies, prophylactic antibiotics are often indicated after snake envenomation because of the variety of bacteria that can be found in dog hair and snake mouth [32138]. However, according to recent veterinary study, transmission of infection through the snakebite is rare because of dilutional effects associated with the hemorrhagic lymphedema and proteolytic effects of snake venom [23]. And one prospective study performed in dogs in rattlesnake envenomation discourage antibiotic use as there was low incidences of wound infection after snake bite [39]. While use of antibiotics in treatment of snake bites is not currently promoted in veterinary medicine, our study found common usage by Korean veterinarians. These results are not supported by most recent literature and that our study could not reveal any benefit or detriment of their use. Moreover, our study revealed an association of antibiotic use with survival but could not demonstrate that antibiotic use affected survival.\nThe overall mortality rate in the present study was 7%. Previous studies reported mortality rates for bites of pit vipers, rattlesnakes, and tiger snakes of 4%, 1%, and 15%, respectively; however, mamushi bites have not been reported in dogs [182021]. Discrepancies in mortality rates may be associated with differences in venom constituents between snake species and the amount of venom inoculated [24]. Mortality rates after antivenom administration also vary between groups [244041]. In pit viper envenomation, the administration of antivenom did not increase the survival rate [21]; however, in tiger snake envenomation, the mortality rate with antivenom was 17% compared with 77% in dogs that were not treated with antivenom [4243]. In the present study, the mortality rate of dogs that received antivenom was 6%, compared with 12% in those that did not receive this treatment. Although antivenom seemed to have a positive effect on mortality, due to the nature of a veterinary referral hospital, follow-up information on patients was limited; hence, the effectiveness of antivenom in our study cannot be estimated.\nAccording to the WHO, snakebites remain a devastating and life-threatening environmental hazard for humans not only in tropical developing countries but also in developed nations with high population densities [144]. Envenoming resulting from snakebites is an important public health problem because it can lead to death and requires antivenom, the only specific treatment [1]. Despite increasing knowledge of the management of snake bites in humans, satisfactory data and guidelines remain lacking in veterinary medicine, especially in Korea. Our study is of great importance for veterinary public health as it provides baseline information for veterinary clinics to better control patients bitten by snakes.\nThe present study has several limitations owing to its retrospective design. First, the medical records were occasionally incomplete because data from three different animal hospitals were collected for analysis due to the low incidence of snakebites in dogs in Korea. Second, several species of snakes are implicated in snakebites and our study was not confined to mamushi only. Thus, it was impossible to evaluate whether envenomation occurred in the dogs included in this study. Third, many of the dogs in our study presented to the hospital only for antivenom treatment; thus, the possibility of follow-up and assessment of prognosis was limited, and complications could not be evaluated properly because serial laboratory examinations were performed only in a small number of dogs.\nWhile snakebite in dogs in Korea is an uncommon medical problem, it should be considered as it can eventually lead to death. To the best of our knowledge, this is the first study to retrospectively analyze the clinical features of snakebites in dogs in Korea. The most significant finding of our study is that the most prevalent clinical signs were edema, erythema, and echinocytosis in blood smears. The mortality rate in this study was 7%, and antivenom seemed to have a positive effect on mortality; however, further research is essential to determine the effectiveness of antivenom. In our opinion, the presented data could help in the management of dogs with snakebites in Korean veterinary medicine." ]
[ "intro", "materials|methods", null, "results", "discussion" ]
[ "Snakebites", "mamushi", "echinocytosis", "dogs", "antivenin" ]
Chlorogenic acid alleviates the reduction of Akt and Bad phosphorylation and of phospho-Bad and 14-3-3 binding in an animal model of stroke.
36259103
Stroke is caused by disruption of blood supply and results in permanent disabilities as well as death. Chlorogenic acid is a phenolic compound found in various fruits and coffee and exerts antioxidant, anti-inflammatory, and anti-apoptotic effects.
BACKGROUND
Chlorogenic acid (30 mg/kg) or vehicle was administered peritoneally to adult male rats 2 h after MCAO surgery, and animals were sacrificed 24 h after MCAO surgery. Neurobehavioral tests were performed, and brain tissues were isolated. The cerebral cortex was collected for Western blot and immunoprecipitation analyses.
METHODS
MCAO damage caused severe neurobehavioral disorders and chlorogenic acid improved the neurological disorders. Chlorogenic acid alleviated the MCAO-induced histopathological changes and decreased the number of terminal deoxynucleotidyl transferase dUTP nick end labeling-positive cells. Furthermore, MCAO-induced damage reduced the expression of phospho-PDK1, phospho-Akt, and phospho-Bad, which was alleviated with administration of chlorogenic acid. The interaction between phospho-Bad and 14-3-3 levels was reduced in MCAO animals, which was attenuated by chlorogenic acid treatment. In addition, chlorogenic acid alleviated the increase of cytochrome c and caspase-3 expression caused by MCAO damage.
RESULTS
The results of the present study showed that chlorogenic acid activates phospho-Akt and phospho-Bad and promotes the interaction between phospho-Bad and 14-3-3 during MCAO damage. In conclusion, chlorogenic acid exerts neuroprotective effects by activating the Akt-Bad signaling pathway and maintaining the interaction between phospho-Bad and 14-3-3 in ischemic stroke model.
CONCLUSIONS
[ "Animals", "Male", "Rats", "Apoptosis", "bcl-Associated Death Protein", "Brain Ischemia", "Chlorogenic Acid", "Disease Models, Animal", "Infarction, Middle Cerebral Artery", "Phosphatidylinositol 3-Kinases", "Phosphorylation", "Proto-Oncogene Proteins c-akt", "Rats, Sprague-Dawley", "Stroke", "14-3-3 Proteins" ]
9715392
INTRODUCTION
Stroke is a serious neurological disorder and the second leading cause of death [1]. The two main categories of stroke are ischemic and hemorrhagic. Ischemic stroke is caused by a blockage of blood supply, and hemorrhagic stroke is due to a vascular rupture [2]. A stroke blocks glucose and oxygen supply to the nerve cells, leading to cell death. Ischemic stroke generates reactive oxygen species and causes neuronal damage and apoptosis [3]. Cerebral ischemia prevents the survival signaling pathway and activates the apoptotic signaling pathway, resulting in neuronal cell death [45]. Phosphatidylinositol 3 kinase (PI3K) plays a key role in various cellular processes, including metabolism, inflammation, and cell survival [6]. PI3K is responsible for the initiation of a signaling cascade by activating 3-phosphoinositide-dependent kinase 1 (PDK1) and continuously phosphorylating Akt in the activation loop [7]. Akt is a key mediator of signal transduction pathways that regulate cell proliferation and survival. Akt phosphorylates pro-apoptotic proteins, including Bad, glycogen synthase kinase 3β, and forkhead transcription factor, and prevents apoptotic functions of these proteins [8]. Thus, Akt activation suppresses the activity of caspases and protects cells from apoptosis [9]. Bad is a representative pro-apoptotic protein of the Bcl-2 family and promotes cell death by heterodimerization with Bcl-2 or Bcl-xL [10]. However, the phosphorylated form of Bad (phospho-Bad) has lower capability to interact with Bcl-xL and binds to 14-3-3 proteins [11]. The binding of phospho-Bad and 14-3-3 proteins suppresses cell death by preventing interactions with Bad/Bcl-2 or Bad/Bcl-xL. However, dephosphorylated Bad binds to Bcl-2 or Bcl-xL and continuously releases cytochrome c from mitochondria into the cytosol and initiates the caspase cascade, resulting in apoptosis [1213]. Chlorogenic acid is a phenolic compound found in various foods such as coffee, cocoa, and citrus fruits [14]. Chlorogenic acid exerts anti-oxidative and anti-inflammatory effects and protects brain tissues from ischemic damage by controlling inflammatory and nerve growth factors [1516]. We recently reported the antioxidant, anti-inflammatory, and neuroprotective effects of chlorogenic acid on focal cerebral ischemia [1718]. Furthermore, chlorogenic acid exerts cytoprotective effects against oxidative stress through the PI3K/Akt pathway [19]. The results of the above-mentioned studies provide sufficient evidence regarding the numerous effects of chlorogenic acid. However, the exact neuroprotective mechanism of chlorogenic acid is not fully known. Whether chlorogenic acid regulates the interaction between phospho-Bad and 14-3-3 proteins has not been reported to date. It was hypothesized here that chlorogenic acid exerts neuroprotective effects by regulating the phosphorylation of Akt and Bad and the interaction between phospho-Bad and 14-3-3 proteins. Thus, the changes of phospho-Akt and phospho-Bad expression and of phospho-Bad and 14-3-3 protein binding were investigated in an animal model of focal cerebral ischemia treated with chlorogenic acid.
null
null
RESULTS
Neurological tests were performed to assess behavioral deficits and investigate the protective effect of chlorogenic acid on MCAO damage. MCAO animals showed severe neurological deficits such as paralysis, loss of balance, and seizures. However, chlorogenic acid treatment improved these deficits. The neurological deficit scores were 4.09 ± 0.46 and 2.01 ± 0.28 in vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 1A). Neurological deficits were not found in sham animals regardless of vehicle or chlorogenic acid treatment. The results of the corner test showed a significant increase in the number of right turns in MCAO animals treated with vehicle. However, chlorogenic acid treatment alleviated this increase. The numbers of right turns and left turns were the same in sham animals regardless of vehicle or chlorogenic acid treatment. The number of right turns was 9.16 ± 0.27 and 6.13 ± 0.17 in vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 1B). MCAO damage caused sensory motor impairment. The removal time of red adhesive dots was significantly increased in MCAO animals treated with vehicle; chlorogenic acid treatment alleviated these changes. The time required to remove the adhesive dots was 175.2 ± 8.5 sec and 80.2 ± 6.8 sec in the vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 1C). In addition, the grip strength test was performed. A decrease in grip strength of the contralateral forelimb was observed in animals with MCAO damage and was alleviated by chlorogenic acid. The grip strength in the left forelimb was 0.14 ± 0.02 and 0.39 ± 0.02 in the vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 1D). The grip strength of the forelimb was nearly identical in vehicle + sham and chlorogenic acid + sham animals. MCAO, middle cerebral artery occlusion; CGA, chlorogenic acid. ap < 0.05. In addition, severe structural and histopathological changes were observed in the right cerebral cortex of MCAO animals. Neuronal and cytoplasmic shrinkage, vacuole formation, and shrunken dendrites were observed in MCAO animals (Fig. 2A-D). However, chlorogenic acid treatment alleviated the histopathological changes. Sham animals had normal neuronal structure with round nuclei and well-developed dendrites. The number of damaged cells was 84 ± 5.77 and 43 ± 5.77 in vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 2I). TUNEL histochemical staining was performed to detect apoptotic cells. An increase in the number of TUNEL-positive cells was observed in MCAO animals, and chlorogenic acid treatment attenuated the increase (Fig. 2E-H). The number of TUNEL-positive cells was 94 ± 6.03 and 39 ± 5.98 in vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 2I). TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling; MCAO, middle cerebral artery occlusion; CGA, chlorogenic acid. ap < 0.05. The expression of phospho-PDK1, phospho-Akt, and phospho-Bad in the cerebral cortex was analyzed. Western blot analysis showed that MCAO damage decreased phospho-PDK1, phospho-Akt, and phospho-Bad expression, and chlorogenic acid treatment prevented the decrease (Fig. 3A). Phospho-PDK1 level was 0.42 ± 0.03 and 1.06 ± 0.05 in vehicle + MCAO animals and chlorogenic acid + MCAO animals, respectively (Fig. 3B). Phospho-Akt level was 0.32 ± 0.04 in vehicle + MCAO animals and 0.80 ± 0.03 in chlorogenic acid + MCAO animals. Phospho-Bad level was 0.36 ± 0.05 and 0.90 ± 0.03 in vehicle + MCAO and chlorogenic acid + MCAO animals, respectively. The protein expression levels were nearly identical in sham animals regardless of vehicle or chlorogenic acid treatment. Furthermore, PDK1 and Akt expression was maintained at similar levels in vehicle + MCAO animals and chlorogenic acid + MCAO animals. The results of immunofluorescence staining confirmed the change of phospho-Akt and phospho-Bad levels in MCAO animals (Fig. 4). DAPI staining was performed to confirm the nucleus, and phospho-Akt and phospho-Bad were located in cytoplasm. These proteins were significantly decreased in the cerebral cortex of MCAO animals with vehicle treatment, but these decreases were alleviated by chlorogenic acid treatment. The percentage of phospho-Akt-positive cells was 12.17 ± 2.25 and 36.00 ± 4.05 in vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 4B). The number of phospho-Bad-positive cells was 21.30 ± 3.55 and 59.24 ± 4.02 in vehicle + MCAO animals and chlorogenic acid + MCAO animals, respectively (Fig. 4C). The number of positive cells was similar between vehicle + sham and chlorogenic acid + sham animals. PDK1, phosphoinositide-dependent kinase 1; MCAO, middle cerebral artery occlusion; CGA, chlorogenic acid. ap < 0.05. MCAO, middle cerebral artery occlusion; CGA, chlorogenic acid. ap < 0.05. Immunoprecipitation analysis was performed to investigate changes in the interaction between phospho-Bad and 14-3-3 proteins in animals with MCAO damage. The interaction level decreased in MCAO animals treated with vehicle but chlorogenic acid treatment alleviated the reduced interaction (Fig. 5A). Phospho-Bad and 14-3-3 interaction level was 0.62 ± 0.04 and 1.08 ± 0.05 in vehicle + MCAO animals and chlorogenic acid + MCAO animals, respectively (Fig. 5B). Changes in cytochrome c expression were also observed in animals with MCAO damage. Cytochrome c expression was increased in MCAO animals treated with vehicle, and chlorogenic acid treatment alleviated this increase (Fig. 5C). Cytochrome c level was 0.91 ± 0.03 in vehicle + MCAO animals and 0.34 ± 0.01 in chlorogenic acid + MCAO animals (Fig. 5D). In addition, the expressions of caspase-3 and cleaved caspase-3 were increased, and these increases were alleviated by chlorogenic acid treatment (Fig. 6A). Caspase-3 level was 1.32 ± 0.04 and 0.64 ± 0.04 in vehicle + MCAO animals and chlorogenic acid + MCAO animals, respectively (Fig. 6B). Cleaved caspase-3 level was 1.06 ± 0.03 in vehicle + MCAO animals and 0.58 ± 0.05 in chlorogenic acid + MCAO animals (Fig. 6B). The ratio between cleaved caspase-3 and caspase-3 level was 0.96 ± 0.05 and 0.87 ± 0.07 in vehicle + MCAO animals and chlorogenic acid + MCAO animals, respectively (Fig. 6C). Significant difference was not observed between vehicle + sham and chlorogenic acid + sham animals. MCAO, middle cerebral artery occlusion; CGA, chlorogenic acid. ap < 0.05. MCAO, middle cerebral artery occlusion; CGA, chlorogenic acid. ap < 0.05.
null
null
[ "Experimental animals and drug treatment", "MCAO", "Neurological deficit scoring test", "Corner test", "Adhesive-removal test", "Grip strength test", "Hematoxylin and eosin staining", "Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay", "Western blot analysis", "Immunofluorescence staining", "Immunoprecipitation assay", "Statistical analysis" ]
[ "Male Sprague Dawley rats (200–230 g, n = 60) were purchased from Samtako Co. (Animal Breeding Centre, Osan, Korea). All experimental procedures were carried out according to the guideline of the Institutional Animal Care and Use Committee of Gyeongsang National University (GNU-220222-R0021). Rats were maintained under a controlled environment (25ºC, 12 h light/12 h dark cycle) and were provided free access to feed and water. Animals were randomly divided into four groups: vehicle + sham, chlorogenic acid + sham, vehicle + middle cerebral artery occlusion (MCAO), and chlorogenic acid + MCAO group. Chlorogenic acid (Sigma-Aldrich, USA) was dissolved in phosphate buffer saline (PBS) and was intraperitoneally injected 2 h after the MCAO surgery [1620]. PBS was used as solvent agent and vehicle-treated animals were injected with PBS without chlorogenic acid. Fifteen rats per group were used for the following experiments: histopathological studies (n = 5 for each group), Western blot (n = 5 for each group), and immunoprecipitation analysis (n = 5 for each group). Neurobehavioral tests were performed in all animals (n = 60).", "Animals were anesthetized with 50 mg of Zoletil (Virbac, France) before MCAO surgery. They were placed on a heating pad to prevent hypothermia during the surgical procedure. We performed MCAO surgery as previously described mannuals [21]. Animals were kept in a supine position and a midline incision was given to the neck. The right common carotid artery (CCA) was exposed by separation from the adjacent muscles, tissues, and nerves. The right external carotid artery (ECA) and the right internal carotid artery (ICA) were continuously exposed and the right CCA was temporarily ligated with microvascular clamp. The proximal end of the right ECA was ligated and cut. A 4/0 nylon suture with rounded tip by heating was carefully inserted into the right ECA and moved forward to the right ICA. It was inserted until resistance was felt to block the origin of the middle cerebral artery. The length of the inserted nylon suture is almost 22–24 mm. The inserted nylon and the ECA were ligated with black silk to fix the nylon suture. The skin of neck was sutured with black silk. Animals were kept on heating pads until they were fully conscious and transferred to animal cage. They were performed neurological behavioral tests 24 h after MCAO and euthanized by cervical dislocation immediately after neurological behavioral tests. The whole brains were carefully isolated from skull and fixed for morphological study. The cerebral cortex tissues were separated from the whole brain and collected further experiments.", "A neurological deficit scoring test was carried out for the evaluation of neurological behavior deficits 24 h after MCAO surgery. It was based on a five-point scale system [22]. It was given to animals according to their neurological responses: normal posture and no sign of neurological abnormality (no neurological deficit, 0), lack of the ability to completely extend the contralateral forelimb (mild neurological deficit, 1), circling to the contralateral side (moderate neurological deficit, 2), inability to walk and falling to the contralateral side with signs of seizures and sensitivity to stimulus (severe neurological deficit, 3), and no movement or no sign of consciousness (very severe neurological deficit, 4).", "The corner test was performed for the examination of sensory-motor asymmetry [23]. Two whiteboards (30 × 20 × 1 cm3) were kept perpendicular at a 30º angle to each other. Small spaces in between the boards were kept for animals to move forward to the corner. Animals were kept at the wide side of the whiteboards and allowed to move freely toward the corner. When the animals reach the corner, their vibrissae was touched to the side of the boards and animals turned to the right or left side. The number of left and right turns for each animal was recorded and the test was repeated ten times. Animals were trained for seven days before MCAO surgery and animals with the same rate of right and left turns were selected for this study.", "Adhesive-removal test was performed using red adhesive dots with approximately 12 mm in diameter for the evaluation of somatosensory sensation [24]. Animals were removed from their cages and kept on a table. They were carefully catched from the neck and red dots were attached to both the forelimbs. They were kept back in their cage and time for the removal of these dots from both the forelimbs was recorded with a stopwatch. The same procedure was repeated five times for each animal. Animals were trained for three days before performing the MCAO surgery and were selected that successfully removed the dots within 10 sec.", "The grip strength test was performed for the evaluation of strength in the left and right forelimbs using a grip strength meter (Jeung Do Bio & Plant Co., Ltd., Korea) [25]. The grip strength meter was set to zero and the right or left forelimb was placed on the metal mesh of the gripper. When the animals grabbed the metal mesh with their right or left paw, we pulled them back from their tails to evaluate maximum force from each forelimb. The test was repeated five times for the left and right paw of each animal.", "Whole brains were carefully removed from the skull and immediately fixed in a 4% paraformaldehyde solution. They were sliced with a brain matrix (Ted Pella, USA) and washed with tap water for overnight. Tissue slices were dehydrated with graded ethyl alcohol series (70% to 100%) and cleaned in xylene. They were kept in the vacuum chamber of the paraffin embedding center (Leica, Germany) for 1 h and embedded. Paraffin blocks were cut into 4 μm thick sections using a rotary microtome (Leica). Paraffin ribbons were placed on glass slides and dried on slide warmer (Thermo Fischer Scientific, USA). Section were deparaffinized with xylene, rehydrated in graded ethyl alcohol series (100% to 70%), and kept in tap water. Sections were stained with Harris’ hematoxylin solution (Sigma-Aldrich) for 10 min and washed with running tap water for 10 min. They were dipped in a 1% hydrochloric acid solution with 70% ethyl alcohol for differentiation and washed with tap water. They were neutralized by dipping in a 1% ammonia solution and washed with tap water. They were stained with eosin Y solution (Sigma-Aldrich) for 1 min, dehydrated with graded ethyl alcohol series (70% to 100%), and cleaned with xylene. The permount mounting medium (Thermo Fischer Scientific) was dropped and the tissues were covered with cover glass. The sections were observed and photographed using an Olympus microscope (Olympus, Japan). The images of the right cerebral cortex were presented in the results. Five regions of the cerebral cortex were selected and damaged cells were counted in each region. Cells with shrunken dendrite, vacuoles formation, nucleus condensation were consider as damaged cells. Damaged cells were expressed as a percentage of the number of the damaged cells to the number of total cells.", "We performed TUNEL assay to detect apoptotic cell death and TUNEL assay was performed with an ApopTag Peroxidase In Situ Apoptosis Detection Kit (Merck, USA) according to the manufacturer’s manual. Paraffin sections were deparaffinized with xylene and rehydrated with graded ethyl alcohol series (100% to 70%). Sections were incubated with proteinase K (20 µg/mL) for 1 min and washed three times with PBS for 5 min. They were dipped in methanol of 3% hydrogen peroxide for 5 min at room temperature, washed three times with PBS for 5 min, and incubated with equilibration buffer for 1 h at 4°C. They were reacted with working strength terminal deoxynucleotidyl transferase (TdT) enzyme in a humidified chamber for 90 min at 37°C and applied with stop/wash buffer for 10 min to terminate TdT enzyme reaction. They were washed twice with PBS for 5 min and incubated with anti-digoxigenin conjugate in a humidified chamber for 1 h at room temperature. Sections were washed with PBS for three times for 5 min, stained with 3,3'-diaminobenzidine (Sigma-Aldrich), and washed three times with PBS for 5 min. They were counterstained with hematoxylin solution, washed with tap water, dehydrated in graded ethyl alcohol series (70% to 100%), and cleaned with xylene. They were coverslipped with permount mounting medium (Thermo Fisher Scientific, USA) and observed under an Olympus microscope (Olympus). Cells stained with dark brown were considered TUNEL-positive cells. We randomly selected five regions of the cerebral cortex and TUNEL-positive cells were counted in each region. Apoptotic index was expressed as a percentage of the number of the TUNEL-positive cells to the number of total cells.", "Right cerebral cortex tissues were homogenized in lysis buffer (1% Triton X-100, 1 mM ethylenediaminetetraacetic acid in 1 × PBS [pH 7.4]) containing 200 μM phenylmethylsulfonyl fluoride. Homogenized samples were sonicated for 3 min and centrifuged at 15,000 g for 20 min. The supernatants were collected and the pellets were discarded. Bicinchoninic acid protein assay kit (Pierce, USA) was used to determine the concentration of proteins. Protein assay was performed according to the manufacturer’s instructions. Total proteins of 30 μg were kept in ice and loaded into 10% sodium dodecyl sulfate-polyacrylamide (SDS-PAGE) gels for electrophoresis. Samples were electrophorized until the dye went down to the bottom of the gels using mini trans-blot cell electrophoresis (Bio-Rad Laboratories, USA). The gels were removed from glass plate and proteins were transferred into polyvinylidene difluoride (PVDF) membranes in transfer tank for Western blot (Bio-Rad Laboratories). PVDF membranes were reacted with a 5% skim milk solution in tris-buffered saline solution with 0.1% Tween 20 (TBST) for 1 h at room temperature to block non-specific bindings and washed three times with TBST for 10 min. They were incubated overnight at 4°C with following primary antibodies: anti-PDK1, anti-phospho-PDK1, anti-Akt, anti-phospho-Akt, anti-Bad, anti-phospho-Bad, anti-cytochrome c, anti-caspase-3, and anti-β-actin (diluted 1:1,000, Cell Signaling Technology, USA, Santa Cruz Biotechnology, USA). Membranes were washed three times with TBST for 10 min and incubated with horseradish peroxidase-conjugated anti-mouse IgG or anti-rabbit IgG (diluted 1:5,000, Cell Signaling Technology) for 2 h at room temperature. They were washed three times with TBST for 10 min and reacted with chemiluminescence detection reagents (GE Healthcare, UK) for 1 min. Membranes were exposed on X-ray film (Fuji Film, Japan) for 1 min, developed in developer solution (Poohung Photo Chemical, Korea), washed with tap water, and fixed in fixation solution (Poohung Photo Chemical). The detected protein bands were scanned and band intensities were calculated with Image J (Media Cybernetics, USA). The relative integrated density of proteins was expressed as a ratio of the density of proteins to that of β-actin.", "Paraffin sections were deparaffinized with xylene and rehydrated in graded ethyl alcohol series (100% to 70%). Sections were washed three times with PBS for 10 min and reacted with 1% normal goat serum for 1 h at room temperature for blocking of non-specific antibody bindings. They were washed three times with PBS for 10 min and incubated with anti-phospho-Akt or anti-phospho-Bad (diluted 1:100, Santa Cruz Biotechnology) overnight at 4ºC. They were washed three times with PBS and incubated with fluorescein isothiocyanate-conjugated secondary antibody (diluted 1:100, Santa Cruz Biotechnology) for 90 min at room temperature. Sections were washed three times with PBS for 10 min, reacted with 4′,6-diamidino-2-phenylindole (DAPI, Sigma-Aldrich) for 10 min, and cover slipped with fluorescent mounting medium (Agilent Technologies, USA). Stained tissues were observed under a confocal microscope (FV-1000, Olympus) and images were taken from the cortical region. Relative integrated densities were analyzed with Image J (Media Cybernetics) and expressed as a ratio of the integrated density of each animal to that of a vehicle + sham animals.", "We performed an immunoprecipitation assay to assess the level of interaction between phospho-Bad and 14-3-3 proteins. Proteins were extracted from the cerebral cortex in the same method that were performed in Western blot analysis. Total protein (200 μg) were reacted with protein A/G agarose beads (Santa Cruz Biotechnology) for blockage of nonspecific binding of other proteins. The mixture was centrifuged at 5,000 g for 1 min and the supernatant was removed. The rest were mixed with anti-14-3-3 antibody and the mixture was incubated for overnight at 4°C on a rocker (FINEPCR CR100, Korea). Protein A/G agarose beads were added in mixture and reacted for 2 h at 4°C. The mixture was washed with radioimmunoprecipitation assay buffer (Sigma-Aldrich) containing PMSF and centrifuged at 10,000 g for 1 min. The supernatant was discarded and sample buffer was added to the rest. Samples were heated at 100°C for 5 min and centrifuged at 10,000 g for 10 min. The supernatant was collected and loaded into a 10% SDS-PAGE gel. They were electrophoresed until the blue dye went down to the bottom of the gel and transferred to PVDF membrane. Membrane was incubated with 5% skim milk solution for 1 h at room temperature and incubated with anti-phospho-Bad antibody (1:1,000, diluted with TBST, Cell Signaling Technology) overnight at 4°C. They were washed three times with TBST for 10 min and reacted with secondary antibody (1:5,000, diluted with TBST, Cell Signaling Technology) for 2 h at room temperature. They were washed three times with TBST for 10 min and reacted with chemiluminescence detection reagents (GE Healthcare). They were exposed on X-ray film (Fuji Film) for 1 min, developed in developer (Poohung Photo Chemical), and continuously fixed in fixer (Poohung Photo Chemical). The detected protein bands were scanned and band intensities were calculated with Image J (Media Cybernetics).", "All data is represented as mean ± standard error of means. Two-way analysis of variance followed by post hoc Scheffe’s test was used to analyze the differences among groups. The p < 0.05 was considered to be statistically significant." ]
[ null, null, null, null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "MATERIALS AND METHODS", "Experimental animals and drug treatment", "MCAO", "Neurological deficit scoring test", "Corner test", "Adhesive-removal test", "Grip strength test", "Hematoxylin and eosin staining", "Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay", "Western blot analysis", "Immunofluorescence staining", "Immunoprecipitation assay", "Statistical analysis", "RESULTS", "DISCUSSION" ]
[ "Stroke is a serious neurological disorder and the second leading cause of death [1]. The two main categories of stroke are ischemic and hemorrhagic. Ischemic stroke is caused by a blockage of blood supply, and hemorrhagic stroke is due to a vascular rupture [2]. A stroke blocks glucose and oxygen supply to the nerve cells, leading to cell death. Ischemic stroke generates reactive oxygen species and causes neuronal damage and apoptosis [3]. Cerebral ischemia prevents the survival signaling pathway and activates the apoptotic signaling pathway, resulting in neuronal cell death [45]. Phosphatidylinositol 3 kinase (PI3K) plays a key role in various cellular processes, including metabolism, inflammation, and cell survival [6]. PI3K is responsible for the initiation of a signaling cascade by activating 3-phosphoinositide-dependent kinase 1 (PDK1) and continuously phosphorylating Akt in the activation loop [7]. Akt is a key mediator of signal transduction pathways that regulate cell proliferation and survival. Akt phosphorylates pro-apoptotic proteins, including Bad, glycogen synthase kinase 3β, and forkhead transcription factor, and prevents apoptotic functions of these proteins [8]. Thus, Akt activation suppresses the activity of caspases and protects cells from apoptosis [9]. Bad is a representative pro-apoptotic protein of the Bcl-2 family and promotes cell death by heterodimerization with Bcl-2 or Bcl-xL [10]. However, the phosphorylated form of Bad (phospho-Bad) has lower capability to interact with Bcl-xL and binds to 14-3-3 proteins [11]. The binding of phospho-Bad and 14-3-3 proteins suppresses cell death by preventing interactions with Bad/Bcl-2 or Bad/Bcl-xL. However, dephosphorylated Bad binds to Bcl-2 or Bcl-xL and continuously releases cytochrome c from mitochondria into the cytosol and initiates the caspase cascade, resulting in apoptosis [1213].\nChlorogenic acid is a phenolic compound found in various foods such as coffee, cocoa, and citrus fruits [14]. Chlorogenic acid exerts anti-oxidative and anti-inflammatory effects and protects brain tissues from ischemic damage by controlling inflammatory and nerve growth factors [1516]. We recently reported the antioxidant, anti-inflammatory, and neuroprotective effects of chlorogenic acid on focal cerebral ischemia [1718]. Furthermore, chlorogenic acid exerts cytoprotective effects against oxidative stress through the PI3K/Akt pathway [19]. The results of the above-mentioned studies provide sufficient evidence regarding the numerous effects of chlorogenic acid. However, the exact neuroprotective mechanism of chlorogenic acid is not fully known. Whether chlorogenic acid regulates the interaction between phospho-Bad and 14-3-3 proteins has not been reported to date. It was hypothesized here that chlorogenic acid exerts neuroprotective effects by regulating the phosphorylation of Akt and Bad and the interaction between phospho-Bad and 14-3-3 proteins. Thus, the changes of phospho-Akt and phospho-Bad expression and of phospho-Bad and 14-3-3 protein binding were investigated in an animal model of focal cerebral ischemia treated with chlorogenic acid.", "[SUBTITLE] Experimental animals and drug treatment [SUBSECTION] Male Sprague Dawley rats (200–230 g, n = 60) were purchased from Samtako Co. (Animal Breeding Centre, Osan, Korea). All experimental procedures were carried out according to the guideline of the Institutional Animal Care and Use Committee of Gyeongsang National University (GNU-220222-R0021). Rats were maintained under a controlled environment (25ºC, 12 h light/12 h dark cycle) and were provided free access to feed and water. Animals were randomly divided into four groups: vehicle + sham, chlorogenic acid + sham, vehicle + middle cerebral artery occlusion (MCAO), and chlorogenic acid + MCAO group. Chlorogenic acid (Sigma-Aldrich, USA) was dissolved in phosphate buffer saline (PBS) and was intraperitoneally injected 2 h after the MCAO surgery [1620]. PBS was used as solvent agent and vehicle-treated animals were injected with PBS without chlorogenic acid. Fifteen rats per group were used for the following experiments: histopathological studies (n = 5 for each group), Western blot (n = 5 for each group), and immunoprecipitation analysis (n = 5 for each group). Neurobehavioral tests were performed in all animals (n = 60).\nMale Sprague Dawley rats (200–230 g, n = 60) were purchased from Samtako Co. (Animal Breeding Centre, Osan, Korea). All experimental procedures were carried out according to the guideline of the Institutional Animal Care and Use Committee of Gyeongsang National University (GNU-220222-R0021). Rats were maintained under a controlled environment (25ºC, 12 h light/12 h dark cycle) and were provided free access to feed and water. Animals were randomly divided into four groups: vehicle + sham, chlorogenic acid + sham, vehicle + middle cerebral artery occlusion (MCAO), and chlorogenic acid + MCAO group. Chlorogenic acid (Sigma-Aldrich, USA) was dissolved in phosphate buffer saline (PBS) and was intraperitoneally injected 2 h after the MCAO surgery [1620]. PBS was used as solvent agent and vehicle-treated animals were injected with PBS without chlorogenic acid. Fifteen rats per group were used for the following experiments: histopathological studies (n = 5 for each group), Western blot (n = 5 for each group), and immunoprecipitation analysis (n = 5 for each group). Neurobehavioral tests were performed in all animals (n = 60).\n[SUBTITLE] MCAO [SUBSECTION] Animals were anesthetized with 50 mg of Zoletil (Virbac, France) before MCAO surgery. They were placed on a heating pad to prevent hypothermia during the surgical procedure. We performed MCAO surgery as previously described mannuals [21]. Animals were kept in a supine position and a midline incision was given to the neck. The right common carotid artery (CCA) was exposed by separation from the adjacent muscles, tissues, and nerves. The right external carotid artery (ECA) and the right internal carotid artery (ICA) were continuously exposed and the right CCA was temporarily ligated with microvascular clamp. The proximal end of the right ECA was ligated and cut. A 4/0 nylon suture with rounded tip by heating was carefully inserted into the right ECA and moved forward to the right ICA. It was inserted until resistance was felt to block the origin of the middle cerebral artery. The length of the inserted nylon suture is almost 22–24 mm. The inserted nylon and the ECA were ligated with black silk to fix the nylon suture. The skin of neck was sutured with black silk. Animals were kept on heating pads until they were fully conscious and transferred to animal cage. They were performed neurological behavioral tests 24 h after MCAO and euthanized by cervical dislocation immediately after neurological behavioral tests. The whole brains were carefully isolated from skull and fixed for morphological study. The cerebral cortex tissues were separated from the whole brain and collected further experiments.\nAnimals were anesthetized with 50 mg of Zoletil (Virbac, France) before MCAO surgery. They were placed on a heating pad to prevent hypothermia during the surgical procedure. We performed MCAO surgery as previously described mannuals [21]. Animals were kept in a supine position and a midline incision was given to the neck. The right common carotid artery (CCA) was exposed by separation from the adjacent muscles, tissues, and nerves. The right external carotid artery (ECA) and the right internal carotid artery (ICA) were continuously exposed and the right CCA was temporarily ligated with microvascular clamp. The proximal end of the right ECA was ligated and cut. A 4/0 nylon suture with rounded tip by heating was carefully inserted into the right ECA and moved forward to the right ICA. It was inserted until resistance was felt to block the origin of the middle cerebral artery. The length of the inserted nylon suture is almost 22–24 mm. The inserted nylon and the ECA were ligated with black silk to fix the nylon suture. The skin of neck was sutured with black silk. Animals were kept on heating pads until they were fully conscious and transferred to animal cage. They were performed neurological behavioral tests 24 h after MCAO and euthanized by cervical dislocation immediately after neurological behavioral tests. The whole brains were carefully isolated from skull and fixed for morphological study. The cerebral cortex tissues were separated from the whole brain and collected further experiments.\n[SUBTITLE] Neurological deficit scoring test [SUBSECTION] A neurological deficit scoring test was carried out for the evaluation of neurological behavior deficits 24 h after MCAO surgery. It was based on a five-point scale system [22]. It was given to animals according to their neurological responses: normal posture and no sign of neurological abnormality (no neurological deficit, 0), lack of the ability to completely extend the contralateral forelimb (mild neurological deficit, 1), circling to the contralateral side (moderate neurological deficit, 2), inability to walk and falling to the contralateral side with signs of seizures and sensitivity to stimulus (severe neurological deficit, 3), and no movement or no sign of consciousness (very severe neurological deficit, 4).\nA neurological deficit scoring test was carried out for the evaluation of neurological behavior deficits 24 h after MCAO surgery. It was based on a five-point scale system [22]. It was given to animals according to their neurological responses: normal posture and no sign of neurological abnormality (no neurological deficit, 0), lack of the ability to completely extend the contralateral forelimb (mild neurological deficit, 1), circling to the contralateral side (moderate neurological deficit, 2), inability to walk and falling to the contralateral side with signs of seizures and sensitivity to stimulus (severe neurological deficit, 3), and no movement or no sign of consciousness (very severe neurological deficit, 4).\n[SUBTITLE] Corner test [SUBSECTION] The corner test was performed for the examination of sensory-motor asymmetry [23]. Two whiteboards (30 × 20 × 1 cm3) were kept perpendicular at a 30º angle to each other. Small spaces in between the boards were kept for animals to move forward to the corner. Animals were kept at the wide side of the whiteboards and allowed to move freely toward the corner. When the animals reach the corner, their vibrissae was touched to the side of the boards and animals turned to the right or left side. The number of left and right turns for each animal was recorded and the test was repeated ten times. Animals were trained for seven days before MCAO surgery and animals with the same rate of right and left turns were selected for this study.\nThe corner test was performed for the examination of sensory-motor asymmetry [23]. Two whiteboards (30 × 20 × 1 cm3) were kept perpendicular at a 30º angle to each other. Small spaces in between the boards were kept for animals to move forward to the corner. Animals were kept at the wide side of the whiteboards and allowed to move freely toward the corner. When the animals reach the corner, their vibrissae was touched to the side of the boards and animals turned to the right or left side. The number of left and right turns for each animal was recorded and the test was repeated ten times. Animals were trained for seven days before MCAO surgery and animals with the same rate of right and left turns were selected for this study.\n[SUBTITLE] Adhesive-removal test [SUBSECTION] Adhesive-removal test was performed using red adhesive dots with approximately 12 mm in diameter for the evaluation of somatosensory sensation [24]. Animals were removed from their cages and kept on a table. They were carefully catched from the neck and red dots were attached to both the forelimbs. They were kept back in their cage and time for the removal of these dots from both the forelimbs was recorded with a stopwatch. The same procedure was repeated five times for each animal. Animals were trained for three days before performing the MCAO surgery and were selected that successfully removed the dots within 10 sec.\nAdhesive-removal test was performed using red adhesive dots with approximately 12 mm in diameter for the evaluation of somatosensory sensation [24]. Animals were removed from their cages and kept on a table. They were carefully catched from the neck and red dots were attached to both the forelimbs. They were kept back in their cage and time for the removal of these dots from both the forelimbs was recorded with a stopwatch. The same procedure was repeated five times for each animal. Animals were trained for three days before performing the MCAO surgery and were selected that successfully removed the dots within 10 sec.\n[SUBTITLE] Grip strength test [SUBSECTION] The grip strength test was performed for the evaluation of strength in the left and right forelimbs using a grip strength meter (Jeung Do Bio & Plant Co., Ltd., Korea) [25]. The grip strength meter was set to zero and the right or left forelimb was placed on the metal mesh of the gripper. When the animals grabbed the metal mesh with their right or left paw, we pulled them back from their tails to evaluate maximum force from each forelimb. The test was repeated five times for the left and right paw of each animal.\nThe grip strength test was performed for the evaluation of strength in the left and right forelimbs using a grip strength meter (Jeung Do Bio & Plant Co., Ltd., Korea) [25]. The grip strength meter was set to zero and the right or left forelimb was placed on the metal mesh of the gripper. When the animals grabbed the metal mesh with their right or left paw, we pulled them back from their tails to evaluate maximum force from each forelimb. The test was repeated five times for the left and right paw of each animal.\n[SUBTITLE] Hematoxylin and eosin staining [SUBSECTION] Whole brains were carefully removed from the skull and immediately fixed in a 4% paraformaldehyde solution. They were sliced with a brain matrix (Ted Pella, USA) and washed with tap water for overnight. Tissue slices were dehydrated with graded ethyl alcohol series (70% to 100%) and cleaned in xylene. They were kept in the vacuum chamber of the paraffin embedding center (Leica, Germany) for 1 h and embedded. Paraffin blocks were cut into 4 μm thick sections using a rotary microtome (Leica). Paraffin ribbons were placed on glass slides and dried on slide warmer (Thermo Fischer Scientific, USA). Section were deparaffinized with xylene, rehydrated in graded ethyl alcohol series (100% to 70%), and kept in tap water. Sections were stained with Harris’ hematoxylin solution (Sigma-Aldrich) for 10 min and washed with running tap water for 10 min. They were dipped in a 1% hydrochloric acid solution with 70% ethyl alcohol for differentiation and washed with tap water. They were neutralized by dipping in a 1% ammonia solution and washed with tap water. They were stained with eosin Y solution (Sigma-Aldrich) for 1 min, dehydrated with graded ethyl alcohol series (70% to 100%), and cleaned with xylene. The permount mounting medium (Thermo Fischer Scientific) was dropped and the tissues were covered with cover glass. The sections were observed and photographed using an Olympus microscope (Olympus, Japan). The images of the right cerebral cortex were presented in the results. Five regions of the cerebral cortex were selected and damaged cells were counted in each region. Cells with shrunken dendrite, vacuoles formation, nucleus condensation were consider as damaged cells. Damaged cells were expressed as a percentage of the number of the damaged cells to the number of total cells.\nWhole brains were carefully removed from the skull and immediately fixed in a 4% paraformaldehyde solution. They were sliced with a brain matrix (Ted Pella, USA) and washed with tap water for overnight. Tissue slices were dehydrated with graded ethyl alcohol series (70% to 100%) and cleaned in xylene. They were kept in the vacuum chamber of the paraffin embedding center (Leica, Germany) for 1 h and embedded. Paraffin blocks were cut into 4 μm thick sections using a rotary microtome (Leica). Paraffin ribbons were placed on glass slides and dried on slide warmer (Thermo Fischer Scientific, USA). Section were deparaffinized with xylene, rehydrated in graded ethyl alcohol series (100% to 70%), and kept in tap water. Sections were stained with Harris’ hematoxylin solution (Sigma-Aldrich) for 10 min and washed with running tap water for 10 min. They were dipped in a 1% hydrochloric acid solution with 70% ethyl alcohol for differentiation and washed with tap water. They were neutralized by dipping in a 1% ammonia solution and washed with tap water. They were stained with eosin Y solution (Sigma-Aldrich) for 1 min, dehydrated with graded ethyl alcohol series (70% to 100%), and cleaned with xylene. The permount mounting medium (Thermo Fischer Scientific) was dropped and the tissues were covered with cover glass. The sections were observed and photographed using an Olympus microscope (Olympus, Japan). The images of the right cerebral cortex were presented in the results. Five regions of the cerebral cortex were selected and damaged cells were counted in each region. Cells with shrunken dendrite, vacuoles formation, nucleus condensation were consider as damaged cells. Damaged cells were expressed as a percentage of the number of the damaged cells to the number of total cells.\n[SUBTITLE] Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay [SUBSECTION] We performed TUNEL assay to detect apoptotic cell death and TUNEL assay was performed with an ApopTag Peroxidase In Situ Apoptosis Detection Kit (Merck, USA) according to the manufacturer’s manual. Paraffin sections were deparaffinized with xylene and rehydrated with graded ethyl alcohol series (100% to 70%). Sections were incubated with proteinase K (20 µg/mL) for 1 min and washed three times with PBS for 5 min. They were dipped in methanol of 3% hydrogen peroxide for 5 min at room temperature, washed three times with PBS for 5 min, and incubated with equilibration buffer for 1 h at 4°C. They were reacted with working strength terminal deoxynucleotidyl transferase (TdT) enzyme in a humidified chamber for 90 min at 37°C and applied with stop/wash buffer for 10 min to terminate TdT enzyme reaction. They were washed twice with PBS for 5 min and incubated with anti-digoxigenin conjugate in a humidified chamber for 1 h at room temperature. Sections were washed with PBS for three times for 5 min, stained with 3,3'-diaminobenzidine (Sigma-Aldrich), and washed three times with PBS for 5 min. They were counterstained with hematoxylin solution, washed with tap water, dehydrated in graded ethyl alcohol series (70% to 100%), and cleaned with xylene. They were coverslipped with permount mounting medium (Thermo Fisher Scientific, USA) and observed under an Olympus microscope (Olympus). Cells stained with dark brown were considered TUNEL-positive cells. We randomly selected five regions of the cerebral cortex and TUNEL-positive cells were counted in each region. Apoptotic index was expressed as a percentage of the number of the TUNEL-positive cells to the number of total cells.\nWe performed TUNEL assay to detect apoptotic cell death and TUNEL assay was performed with an ApopTag Peroxidase In Situ Apoptosis Detection Kit (Merck, USA) according to the manufacturer’s manual. Paraffin sections were deparaffinized with xylene and rehydrated with graded ethyl alcohol series (100% to 70%). Sections were incubated with proteinase K (20 µg/mL) for 1 min and washed three times with PBS for 5 min. They were dipped in methanol of 3% hydrogen peroxide for 5 min at room temperature, washed three times with PBS for 5 min, and incubated with equilibration buffer for 1 h at 4°C. They were reacted with working strength terminal deoxynucleotidyl transferase (TdT) enzyme in a humidified chamber for 90 min at 37°C and applied with stop/wash buffer for 10 min to terminate TdT enzyme reaction. They were washed twice with PBS for 5 min and incubated with anti-digoxigenin conjugate in a humidified chamber for 1 h at room temperature. Sections were washed with PBS for three times for 5 min, stained with 3,3'-diaminobenzidine (Sigma-Aldrich), and washed three times with PBS for 5 min. They were counterstained with hematoxylin solution, washed with tap water, dehydrated in graded ethyl alcohol series (70% to 100%), and cleaned with xylene. They were coverslipped with permount mounting medium (Thermo Fisher Scientific, USA) and observed under an Olympus microscope (Olympus). Cells stained with dark brown were considered TUNEL-positive cells. We randomly selected five regions of the cerebral cortex and TUNEL-positive cells were counted in each region. Apoptotic index was expressed as a percentage of the number of the TUNEL-positive cells to the number of total cells.\n[SUBTITLE] Western blot analysis [SUBSECTION] Right cerebral cortex tissues were homogenized in lysis buffer (1% Triton X-100, 1 mM ethylenediaminetetraacetic acid in 1 × PBS [pH 7.4]) containing 200 μM phenylmethylsulfonyl fluoride. Homogenized samples were sonicated for 3 min and centrifuged at 15,000 g for 20 min. The supernatants were collected and the pellets were discarded. Bicinchoninic acid protein assay kit (Pierce, USA) was used to determine the concentration of proteins. Protein assay was performed according to the manufacturer’s instructions. Total proteins of 30 μg were kept in ice and loaded into 10% sodium dodecyl sulfate-polyacrylamide (SDS-PAGE) gels for electrophoresis. Samples were electrophorized until the dye went down to the bottom of the gels using mini trans-blot cell electrophoresis (Bio-Rad Laboratories, USA). The gels were removed from glass plate and proteins were transferred into polyvinylidene difluoride (PVDF) membranes in transfer tank for Western blot (Bio-Rad Laboratories). PVDF membranes were reacted with a 5% skim milk solution in tris-buffered saline solution with 0.1% Tween 20 (TBST) for 1 h at room temperature to block non-specific bindings and washed three times with TBST for 10 min. They were incubated overnight at 4°C with following primary antibodies: anti-PDK1, anti-phospho-PDK1, anti-Akt, anti-phospho-Akt, anti-Bad, anti-phospho-Bad, anti-cytochrome c, anti-caspase-3, and anti-β-actin (diluted 1:1,000, Cell Signaling Technology, USA, Santa Cruz Biotechnology, USA). Membranes were washed three times with TBST for 10 min and incubated with horseradish peroxidase-conjugated anti-mouse IgG or anti-rabbit IgG (diluted 1:5,000, Cell Signaling Technology) for 2 h at room temperature. They were washed three times with TBST for 10 min and reacted with chemiluminescence detection reagents (GE Healthcare, UK) for 1 min. Membranes were exposed on X-ray film (Fuji Film, Japan) for 1 min, developed in developer solution (Poohung Photo Chemical, Korea), washed with tap water, and fixed in fixation solution (Poohung Photo Chemical). The detected protein bands were scanned and band intensities were calculated with Image J (Media Cybernetics, USA). The relative integrated density of proteins was expressed as a ratio of the density of proteins to that of β-actin.\nRight cerebral cortex tissues were homogenized in lysis buffer (1% Triton X-100, 1 mM ethylenediaminetetraacetic acid in 1 × PBS [pH 7.4]) containing 200 μM phenylmethylsulfonyl fluoride. Homogenized samples were sonicated for 3 min and centrifuged at 15,000 g for 20 min. The supernatants were collected and the pellets were discarded. Bicinchoninic acid protein assay kit (Pierce, USA) was used to determine the concentration of proteins. Protein assay was performed according to the manufacturer’s instructions. Total proteins of 30 μg were kept in ice and loaded into 10% sodium dodecyl sulfate-polyacrylamide (SDS-PAGE) gels for electrophoresis. Samples were electrophorized until the dye went down to the bottom of the gels using mini trans-blot cell electrophoresis (Bio-Rad Laboratories, USA). The gels were removed from glass plate and proteins were transferred into polyvinylidene difluoride (PVDF) membranes in transfer tank for Western blot (Bio-Rad Laboratories). PVDF membranes were reacted with a 5% skim milk solution in tris-buffered saline solution with 0.1% Tween 20 (TBST) for 1 h at room temperature to block non-specific bindings and washed three times with TBST for 10 min. They were incubated overnight at 4°C with following primary antibodies: anti-PDK1, anti-phospho-PDK1, anti-Akt, anti-phospho-Akt, anti-Bad, anti-phospho-Bad, anti-cytochrome c, anti-caspase-3, and anti-β-actin (diluted 1:1,000, Cell Signaling Technology, USA, Santa Cruz Biotechnology, USA). Membranes were washed three times with TBST for 10 min and incubated with horseradish peroxidase-conjugated anti-mouse IgG or anti-rabbit IgG (diluted 1:5,000, Cell Signaling Technology) for 2 h at room temperature. They were washed three times with TBST for 10 min and reacted with chemiluminescence detection reagents (GE Healthcare, UK) for 1 min. Membranes were exposed on X-ray film (Fuji Film, Japan) for 1 min, developed in developer solution (Poohung Photo Chemical, Korea), washed with tap water, and fixed in fixation solution (Poohung Photo Chemical). The detected protein bands were scanned and band intensities were calculated with Image J (Media Cybernetics, USA). The relative integrated density of proteins was expressed as a ratio of the density of proteins to that of β-actin.\n[SUBTITLE] Immunofluorescence staining [SUBSECTION] Paraffin sections were deparaffinized with xylene and rehydrated in graded ethyl alcohol series (100% to 70%). Sections were washed three times with PBS for 10 min and reacted with 1% normal goat serum for 1 h at room temperature for blocking of non-specific antibody bindings. They were washed three times with PBS for 10 min and incubated with anti-phospho-Akt or anti-phospho-Bad (diluted 1:100, Santa Cruz Biotechnology) overnight at 4ºC. They were washed three times with PBS and incubated with fluorescein isothiocyanate-conjugated secondary antibody (diluted 1:100, Santa Cruz Biotechnology) for 90 min at room temperature. Sections were washed three times with PBS for 10 min, reacted with 4′,6-diamidino-2-phenylindole (DAPI, Sigma-Aldrich) for 10 min, and cover slipped with fluorescent mounting medium (Agilent Technologies, USA). Stained tissues were observed under a confocal microscope (FV-1000, Olympus) and images were taken from the cortical region. Relative integrated densities were analyzed with Image J (Media Cybernetics) and expressed as a ratio of the integrated density of each animal to that of a vehicle + sham animals.\nParaffin sections were deparaffinized with xylene and rehydrated in graded ethyl alcohol series (100% to 70%). Sections were washed three times with PBS for 10 min and reacted with 1% normal goat serum for 1 h at room temperature for blocking of non-specific antibody bindings. They were washed three times with PBS for 10 min and incubated with anti-phospho-Akt or anti-phospho-Bad (diluted 1:100, Santa Cruz Biotechnology) overnight at 4ºC. They were washed three times with PBS and incubated with fluorescein isothiocyanate-conjugated secondary antibody (diluted 1:100, Santa Cruz Biotechnology) for 90 min at room temperature. Sections were washed three times with PBS for 10 min, reacted with 4′,6-diamidino-2-phenylindole (DAPI, Sigma-Aldrich) for 10 min, and cover slipped with fluorescent mounting medium (Agilent Technologies, USA). Stained tissues were observed under a confocal microscope (FV-1000, Olympus) and images were taken from the cortical region. Relative integrated densities were analyzed with Image J (Media Cybernetics) and expressed as a ratio of the integrated density of each animal to that of a vehicle + sham animals.\n[SUBTITLE] Immunoprecipitation assay [SUBSECTION] We performed an immunoprecipitation assay to assess the level of interaction between phospho-Bad and 14-3-3 proteins. Proteins were extracted from the cerebral cortex in the same method that were performed in Western blot analysis. Total protein (200 μg) were reacted with protein A/G agarose beads (Santa Cruz Biotechnology) for blockage of nonspecific binding of other proteins. The mixture was centrifuged at 5,000 g for 1 min and the supernatant was removed. The rest were mixed with anti-14-3-3 antibody and the mixture was incubated for overnight at 4°C on a rocker (FINEPCR CR100, Korea). Protein A/G agarose beads were added in mixture and reacted for 2 h at 4°C. The mixture was washed with radioimmunoprecipitation assay buffer (Sigma-Aldrich) containing PMSF and centrifuged at 10,000 g for 1 min. The supernatant was discarded and sample buffer was added to the rest. Samples were heated at 100°C for 5 min and centrifuged at 10,000 g for 10 min. The supernatant was collected and loaded into a 10% SDS-PAGE gel. They were electrophoresed until the blue dye went down to the bottom of the gel and transferred to PVDF membrane. Membrane was incubated with 5% skim milk solution for 1 h at room temperature and incubated with anti-phospho-Bad antibody (1:1,000, diluted with TBST, Cell Signaling Technology) overnight at 4°C. They were washed three times with TBST for 10 min and reacted with secondary antibody (1:5,000, diluted with TBST, Cell Signaling Technology) for 2 h at room temperature. They were washed three times with TBST for 10 min and reacted with chemiluminescence detection reagents (GE Healthcare). They were exposed on X-ray film (Fuji Film) for 1 min, developed in developer (Poohung Photo Chemical), and continuously fixed in fixer (Poohung Photo Chemical). The detected protein bands were scanned and band intensities were calculated with Image J (Media Cybernetics).\nWe performed an immunoprecipitation assay to assess the level of interaction between phospho-Bad and 14-3-3 proteins. Proteins were extracted from the cerebral cortex in the same method that were performed in Western blot analysis. Total protein (200 μg) were reacted with protein A/G agarose beads (Santa Cruz Biotechnology) for blockage of nonspecific binding of other proteins. The mixture was centrifuged at 5,000 g for 1 min and the supernatant was removed. The rest were mixed with anti-14-3-3 antibody and the mixture was incubated for overnight at 4°C on a rocker (FINEPCR CR100, Korea). Protein A/G agarose beads were added in mixture and reacted for 2 h at 4°C. The mixture was washed with radioimmunoprecipitation assay buffer (Sigma-Aldrich) containing PMSF and centrifuged at 10,000 g for 1 min. The supernatant was discarded and sample buffer was added to the rest. Samples were heated at 100°C for 5 min and centrifuged at 10,000 g for 10 min. The supernatant was collected and loaded into a 10% SDS-PAGE gel. They were electrophoresed until the blue dye went down to the bottom of the gel and transferred to PVDF membrane. Membrane was incubated with 5% skim milk solution for 1 h at room temperature and incubated with anti-phospho-Bad antibody (1:1,000, diluted with TBST, Cell Signaling Technology) overnight at 4°C. They were washed three times with TBST for 10 min and reacted with secondary antibody (1:5,000, diluted with TBST, Cell Signaling Technology) for 2 h at room temperature. They were washed three times with TBST for 10 min and reacted with chemiluminescence detection reagents (GE Healthcare). They were exposed on X-ray film (Fuji Film) for 1 min, developed in developer (Poohung Photo Chemical), and continuously fixed in fixer (Poohung Photo Chemical). The detected protein bands were scanned and band intensities were calculated with Image J (Media Cybernetics).\n[SUBTITLE] Statistical analysis [SUBSECTION] All data is represented as mean ± standard error of means. Two-way analysis of variance followed by post hoc Scheffe’s test was used to analyze the differences among groups. The p < 0.05 was considered to be statistically significant.\nAll data is represented as mean ± standard error of means. Two-way analysis of variance followed by post hoc Scheffe’s test was used to analyze the differences among groups. The p < 0.05 was considered to be statistically significant.", "Male Sprague Dawley rats (200–230 g, n = 60) were purchased from Samtako Co. (Animal Breeding Centre, Osan, Korea). All experimental procedures were carried out according to the guideline of the Institutional Animal Care and Use Committee of Gyeongsang National University (GNU-220222-R0021). Rats were maintained under a controlled environment (25ºC, 12 h light/12 h dark cycle) and were provided free access to feed and water. Animals were randomly divided into four groups: vehicle + sham, chlorogenic acid + sham, vehicle + middle cerebral artery occlusion (MCAO), and chlorogenic acid + MCAO group. Chlorogenic acid (Sigma-Aldrich, USA) was dissolved in phosphate buffer saline (PBS) and was intraperitoneally injected 2 h after the MCAO surgery [1620]. PBS was used as solvent agent and vehicle-treated animals were injected with PBS without chlorogenic acid. Fifteen rats per group were used for the following experiments: histopathological studies (n = 5 for each group), Western blot (n = 5 for each group), and immunoprecipitation analysis (n = 5 for each group). Neurobehavioral tests were performed in all animals (n = 60).", "Animals were anesthetized with 50 mg of Zoletil (Virbac, France) before MCAO surgery. They were placed on a heating pad to prevent hypothermia during the surgical procedure. We performed MCAO surgery as previously described mannuals [21]. Animals were kept in a supine position and a midline incision was given to the neck. The right common carotid artery (CCA) was exposed by separation from the adjacent muscles, tissues, and nerves. The right external carotid artery (ECA) and the right internal carotid artery (ICA) were continuously exposed and the right CCA was temporarily ligated with microvascular clamp. The proximal end of the right ECA was ligated and cut. A 4/0 nylon suture with rounded tip by heating was carefully inserted into the right ECA and moved forward to the right ICA. It was inserted until resistance was felt to block the origin of the middle cerebral artery. The length of the inserted nylon suture is almost 22–24 mm. The inserted nylon and the ECA were ligated with black silk to fix the nylon suture. The skin of neck was sutured with black silk. Animals were kept on heating pads until they were fully conscious and transferred to animal cage. They were performed neurological behavioral tests 24 h after MCAO and euthanized by cervical dislocation immediately after neurological behavioral tests. The whole brains were carefully isolated from skull and fixed for morphological study. The cerebral cortex tissues were separated from the whole brain and collected further experiments.", "A neurological deficit scoring test was carried out for the evaluation of neurological behavior deficits 24 h after MCAO surgery. It was based on a five-point scale system [22]. It was given to animals according to their neurological responses: normal posture and no sign of neurological abnormality (no neurological deficit, 0), lack of the ability to completely extend the contralateral forelimb (mild neurological deficit, 1), circling to the contralateral side (moderate neurological deficit, 2), inability to walk and falling to the contralateral side with signs of seizures and sensitivity to stimulus (severe neurological deficit, 3), and no movement or no sign of consciousness (very severe neurological deficit, 4).", "The corner test was performed for the examination of sensory-motor asymmetry [23]. Two whiteboards (30 × 20 × 1 cm3) were kept perpendicular at a 30º angle to each other. Small spaces in between the boards were kept for animals to move forward to the corner. Animals were kept at the wide side of the whiteboards and allowed to move freely toward the corner. When the animals reach the corner, their vibrissae was touched to the side of the boards and animals turned to the right or left side. The number of left and right turns for each animal was recorded and the test was repeated ten times. Animals were trained for seven days before MCAO surgery and animals with the same rate of right and left turns were selected for this study.", "Adhesive-removal test was performed using red adhesive dots with approximately 12 mm in diameter for the evaluation of somatosensory sensation [24]. Animals were removed from their cages and kept on a table. They were carefully catched from the neck and red dots were attached to both the forelimbs. They were kept back in their cage and time for the removal of these dots from both the forelimbs was recorded with a stopwatch. The same procedure was repeated five times for each animal. Animals were trained for three days before performing the MCAO surgery and were selected that successfully removed the dots within 10 sec.", "The grip strength test was performed for the evaluation of strength in the left and right forelimbs using a grip strength meter (Jeung Do Bio & Plant Co., Ltd., Korea) [25]. The grip strength meter was set to zero and the right or left forelimb was placed on the metal mesh of the gripper. When the animals grabbed the metal mesh with their right or left paw, we pulled them back from their tails to evaluate maximum force from each forelimb. The test was repeated five times for the left and right paw of each animal.", "Whole brains were carefully removed from the skull and immediately fixed in a 4% paraformaldehyde solution. They were sliced with a brain matrix (Ted Pella, USA) and washed with tap water for overnight. Tissue slices were dehydrated with graded ethyl alcohol series (70% to 100%) and cleaned in xylene. They were kept in the vacuum chamber of the paraffin embedding center (Leica, Germany) for 1 h and embedded. Paraffin blocks were cut into 4 μm thick sections using a rotary microtome (Leica). Paraffin ribbons were placed on glass slides and dried on slide warmer (Thermo Fischer Scientific, USA). Section were deparaffinized with xylene, rehydrated in graded ethyl alcohol series (100% to 70%), and kept in tap water. Sections were stained with Harris’ hematoxylin solution (Sigma-Aldrich) for 10 min and washed with running tap water for 10 min. They were dipped in a 1% hydrochloric acid solution with 70% ethyl alcohol for differentiation and washed with tap water. They were neutralized by dipping in a 1% ammonia solution and washed with tap water. They were stained with eosin Y solution (Sigma-Aldrich) for 1 min, dehydrated with graded ethyl alcohol series (70% to 100%), and cleaned with xylene. The permount mounting medium (Thermo Fischer Scientific) was dropped and the tissues were covered with cover glass. The sections were observed and photographed using an Olympus microscope (Olympus, Japan). The images of the right cerebral cortex were presented in the results. Five regions of the cerebral cortex were selected and damaged cells were counted in each region. Cells with shrunken dendrite, vacuoles formation, nucleus condensation were consider as damaged cells. Damaged cells were expressed as a percentage of the number of the damaged cells to the number of total cells.", "We performed TUNEL assay to detect apoptotic cell death and TUNEL assay was performed with an ApopTag Peroxidase In Situ Apoptosis Detection Kit (Merck, USA) according to the manufacturer’s manual. Paraffin sections were deparaffinized with xylene and rehydrated with graded ethyl alcohol series (100% to 70%). Sections were incubated with proteinase K (20 µg/mL) for 1 min and washed three times with PBS for 5 min. They were dipped in methanol of 3% hydrogen peroxide for 5 min at room temperature, washed three times with PBS for 5 min, and incubated with equilibration buffer for 1 h at 4°C. They were reacted with working strength terminal deoxynucleotidyl transferase (TdT) enzyme in a humidified chamber for 90 min at 37°C and applied with stop/wash buffer for 10 min to terminate TdT enzyme reaction. They were washed twice with PBS for 5 min and incubated with anti-digoxigenin conjugate in a humidified chamber for 1 h at room temperature. Sections were washed with PBS for three times for 5 min, stained with 3,3'-diaminobenzidine (Sigma-Aldrich), and washed three times with PBS for 5 min. They were counterstained with hematoxylin solution, washed with tap water, dehydrated in graded ethyl alcohol series (70% to 100%), and cleaned with xylene. They were coverslipped with permount mounting medium (Thermo Fisher Scientific, USA) and observed under an Olympus microscope (Olympus). Cells stained with dark brown were considered TUNEL-positive cells. We randomly selected five regions of the cerebral cortex and TUNEL-positive cells were counted in each region. Apoptotic index was expressed as a percentage of the number of the TUNEL-positive cells to the number of total cells.", "Right cerebral cortex tissues were homogenized in lysis buffer (1% Triton X-100, 1 mM ethylenediaminetetraacetic acid in 1 × PBS [pH 7.4]) containing 200 μM phenylmethylsulfonyl fluoride. Homogenized samples were sonicated for 3 min and centrifuged at 15,000 g for 20 min. The supernatants were collected and the pellets were discarded. Bicinchoninic acid protein assay kit (Pierce, USA) was used to determine the concentration of proteins. Protein assay was performed according to the manufacturer’s instructions. Total proteins of 30 μg were kept in ice and loaded into 10% sodium dodecyl sulfate-polyacrylamide (SDS-PAGE) gels for electrophoresis. Samples were electrophorized until the dye went down to the bottom of the gels using mini trans-blot cell electrophoresis (Bio-Rad Laboratories, USA). The gels were removed from glass plate and proteins were transferred into polyvinylidene difluoride (PVDF) membranes in transfer tank for Western blot (Bio-Rad Laboratories). PVDF membranes were reacted with a 5% skim milk solution in tris-buffered saline solution with 0.1% Tween 20 (TBST) for 1 h at room temperature to block non-specific bindings and washed three times with TBST for 10 min. They were incubated overnight at 4°C with following primary antibodies: anti-PDK1, anti-phospho-PDK1, anti-Akt, anti-phospho-Akt, anti-Bad, anti-phospho-Bad, anti-cytochrome c, anti-caspase-3, and anti-β-actin (diluted 1:1,000, Cell Signaling Technology, USA, Santa Cruz Biotechnology, USA). Membranes were washed three times with TBST for 10 min and incubated with horseradish peroxidase-conjugated anti-mouse IgG or anti-rabbit IgG (diluted 1:5,000, Cell Signaling Technology) for 2 h at room temperature. They were washed three times with TBST for 10 min and reacted with chemiluminescence detection reagents (GE Healthcare, UK) for 1 min. Membranes were exposed on X-ray film (Fuji Film, Japan) for 1 min, developed in developer solution (Poohung Photo Chemical, Korea), washed with tap water, and fixed in fixation solution (Poohung Photo Chemical). The detected protein bands were scanned and band intensities were calculated with Image J (Media Cybernetics, USA). The relative integrated density of proteins was expressed as a ratio of the density of proteins to that of β-actin.", "Paraffin sections were deparaffinized with xylene and rehydrated in graded ethyl alcohol series (100% to 70%). Sections were washed three times with PBS for 10 min and reacted with 1% normal goat serum for 1 h at room temperature for blocking of non-specific antibody bindings. They were washed three times with PBS for 10 min and incubated with anti-phospho-Akt or anti-phospho-Bad (diluted 1:100, Santa Cruz Biotechnology) overnight at 4ºC. They were washed three times with PBS and incubated with fluorescein isothiocyanate-conjugated secondary antibody (diluted 1:100, Santa Cruz Biotechnology) for 90 min at room temperature. Sections were washed three times with PBS for 10 min, reacted with 4′,6-diamidino-2-phenylindole (DAPI, Sigma-Aldrich) for 10 min, and cover slipped with fluorescent mounting medium (Agilent Technologies, USA). Stained tissues were observed under a confocal microscope (FV-1000, Olympus) and images were taken from the cortical region. Relative integrated densities were analyzed with Image J (Media Cybernetics) and expressed as a ratio of the integrated density of each animal to that of a vehicle + sham animals.", "We performed an immunoprecipitation assay to assess the level of interaction between phospho-Bad and 14-3-3 proteins. Proteins were extracted from the cerebral cortex in the same method that were performed in Western blot analysis. Total protein (200 μg) were reacted with protein A/G agarose beads (Santa Cruz Biotechnology) for blockage of nonspecific binding of other proteins. The mixture was centrifuged at 5,000 g for 1 min and the supernatant was removed. The rest were mixed with anti-14-3-3 antibody and the mixture was incubated for overnight at 4°C on a rocker (FINEPCR CR100, Korea). Protein A/G agarose beads were added in mixture and reacted for 2 h at 4°C. The mixture was washed with radioimmunoprecipitation assay buffer (Sigma-Aldrich) containing PMSF and centrifuged at 10,000 g for 1 min. The supernatant was discarded and sample buffer was added to the rest. Samples were heated at 100°C for 5 min and centrifuged at 10,000 g for 10 min. The supernatant was collected and loaded into a 10% SDS-PAGE gel. They were electrophoresed until the blue dye went down to the bottom of the gel and transferred to PVDF membrane. Membrane was incubated with 5% skim milk solution for 1 h at room temperature and incubated with anti-phospho-Bad antibody (1:1,000, diluted with TBST, Cell Signaling Technology) overnight at 4°C. They were washed three times with TBST for 10 min and reacted with secondary antibody (1:5,000, diluted with TBST, Cell Signaling Technology) for 2 h at room temperature. They were washed three times with TBST for 10 min and reacted with chemiluminescence detection reagents (GE Healthcare). They were exposed on X-ray film (Fuji Film) for 1 min, developed in developer (Poohung Photo Chemical), and continuously fixed in fixer (Poohung Photo Chemical). The detected protein bands were scanned and band intensities were calculated with Image J (Media Cybernetics).", "All data is represented as mean ± standard error of means. Two-way analysis of variance followed by post hoc Scheffe’s test was used to analyze the differences among groups. The p < 0.05 was considered to be statistically significant.", "Neurological tests were performed to assess behavioral deficits and investigate the protective effect of chlorogenic acid on MCAO damage. MCAO animals showed severe neurological deficits such as paralysis, loss of balance, and seizures. However, chlorogenic acid treatment improved these deficits. The neurological deficit scores were 4.09 ± 0.46 and 2.01 ± 0.28 in vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 1A). Neurological deficits were not found in sham animals regardless of vehicle or chlorogenic acid treatment. The results of the corner test showed a significant increase in the number of right turns in MCAO animals treated with vehicle. However, chlorogenic acid treatment alleviated this increase. The numbers of right turns and left turns were the same in sham animals regardless of vehicle or chlorogenic acid treatment. The number of right turns was 9.16 ± 0.27 and 6.13 ± 0.17 in vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 1B). MCAO damage caused sensory motor impairment. The removal time of red adhesive dots was significantly increased in MCAO animals treated with vehicle; chlorogenic acid treatment alleviated these changes. The time required to remove the adhesive dots was 175.2 ± 8.5 sec and 80.2 ± 6.8 sec in the vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 1C). In addition, the grip strength test was performed. A decrease in grip strength of the contralateral forelimb was observed in animals with MCAO damage and was alleviated by chlorogenic acid. The grip strength in the left forelimb was 0.14 ± 0.02 and 0.39 ± 0.02 in the vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 1D). The grip strength of the forelimb was nearly identical in vehicle + sham and chlorogenic acid + sham animals.\nMCAO, middle cerebral artery occlusion; CGA, chlorogenic acid.\nap < 0.05.\nIn addition, severe structural and histopathological changes were observed in the right cerebral cortex of MCAO animals. Neuronal and cytoplasmic shrinkage, vacuole formation, and shrunken dendrites were observed in MCAO animals (Fig. 2A-D). However, chlorogenic acid treatment alleviated the histopathological changes. Sham animals had normal neuronal structure with round nuclei and well-developed dendrites. The number of damaged cells was 84 ± 5.77 and 43 ± 5.77 in vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 2I). TUNEL histochemical staining was performed to detect apoptotic cells. An increase in the number of TUNEL-positive cells was observed in MCAO animals, and chlorogenic acid treatment attenuated the increase (Fig. 2E-H). The number of TUNEL-positive cells was 94 ± 6.03 and 39 ± 5.98 in vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 2I).\nTUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling; MCAO, middle cerebral artery occlusion; CGA, chlorogenic acid.\nap < 0.05.\nThe expression of phospho-PDK1, phospho-Akt, and phospho-Bad in the cerebral cortex was analyzed. Western blot analysis showed that MCAO damage decreased phospho-PDK1, phospho-Akt, and phospho-Bad expression, and chlorogenic acid treatment prevented the decrease (Fig. 3A). Phospho-PDK1 level was 0.42 ± 0.03 and 1.06 ± 0.05 in vehicle + MCAO animals and chlorogenic acid + MCAO animals, respectively (Fig. 3B). Phospho-Akt level was 0.32 ± 0.04 in vehicle + MCAO animals and 0.80 ± 0.03 in chlorogenic acid + MCAO animals. Phospho-Bad level was 0.36 ± 0.05 and 0.90 ± 0.03 in vehicle + MCAO and chlorogenic acid + MCAO animals, respectively. The protein expression levels were nearly identical in sham animals regardless of vehicle or chlorogenic acid treatment. Furthermore, PDK1 and Akt expression was maintained at similar levels in vehicle + MCAO animals and chlorogenic acid + MCAO animals. The results of immunofluorescence staining confirmed the change of phospho-Akt and phospho-Bad levels in MCAO animals (Fig. 4). DAPI staining was performed to confirm the nucleus, and phospho-Akt and phospho-Bad were located in cytoplasm. These proteins were significantly decreased in the cerebral cortex of MCAO animals with vehicle treatment, but these decreases were alleviated by chlorogenic acid treatment. The percentage of phospho-Akt-positive cells was 12.17 ± 2.25 and 36.00 ± 4.05 in vehicle + MCAO and chlorogenic acid + MCAO animals, respectively (Fig. 4B). The number of phospho-Bad-positive cells was 21.30 ± 3.55 and 59.24 ± 4.02 in vehicle + MCAO animals and chlorogenic acid + MCAO animals, respectively (Fig. 4C). The number of positive cells was similar between vehicle + sham and chlorogenic acid + sham animals.\nPDK1, phosphoinositide-dependent kinase 1; MCAO, middle cerebral artery occlusion; CGA, chlorogenic acid.\nap < 0.05.\nMCAO, middle cerebral artery occlusion; CGA, chlorogenic acid.\nap < 0.05.\nImmunoprecipitation analysis was performed to investigate changes in the interaction between phospho-Bad and 14-3-3 proteins in animals with MCAO damage. The interaction level decreased in MCAO animals treated with vehicle but chlorogenic acid treatment alleviated the reduced interaction (Fig. 5A). Phospho-Bad and 14-3-3 interaction level was 0.62 ± 0.04 and 1.08 ± 0.05 in vehicle + MCAO animals and chlorogenic acid + MCAO animals, respectively (Fig. 5B). Changes in cytochrome c expression were also observed in animals with MCAO damage. Cytochrome c expression was increased in MCAO animals treated with vehicle, and chlorogenic acid treatment alleviated this increase (Fig. 5C). Cytochrome c level was 0.91 ± 0.03 in vehicle + MCAO animals and 0.34 ± 0.01 in chlorogenic acid + MCAO animals (Fig. 5D). In addition, the expressions of caspase-3 and cleaved caspase-3 were increased, and these increases were alleviated by chlorogenic acid treatment (Fig. 6A). Caspase-3 level was 1.32 ± 0.04 and 0.64 ± 0.04 in vehicle + MCAO animals and chlorogenic acid + MCAO animals, respectively (Fig. 6B). Cleaved caspase-3 level was 1.06 ± 0.03 in vehicle + MCAO animals and 0.58 ± 0.05 in chlorogenic acid + MCAO animals (Fig. 6B). The ratio between cleaved caspase-3 and caspase-3 level was 0.96 ± 0.05 and 0.87 ± 0.07 in vehicle + MCAO animals and chlorogenic acid + MCAO animals, respectively (Fig. 6C). Significant difference was not observed between vehicle + sham and chlorogenic acid + sham animals.\nMCAO, middle cerebral artery occlusion; CGA, chlorogenic acid.\nap < 0.05.\nMCAO, middle cerebral artery occlusion; CGA, chlorogenic acid.\nap < 0.05.", "The neuroprotective effect of chlorogenic acid on cerebral ischemia was confirmed in the present study. Neurological behavior tests were performed to elucidate the neuroprotective function of chlorogenic acid. MCAO damage induced severe neurobehavioral disorders, and chlorogenic acid alleviated these disorders. Chlorogenic acid also prevented histopathological changes and attenuated the number of TUNEL-positive cells in MCAO animals. Chlorogenic acid was confirmed to attenuate the apoptosis process caused by MCAO damage. Activation of the Akt signaling pathway by chlorogenic acid in an animal model of cerebral ischemia was further investigated in the present study.\nPI3K plays an important role in cell survival and activates PDK1 [67]. The activated PDK1 consecutively activates Akt, which regulates cell proliferation and survival and prevents neuronal cell death. We previously reported that the PI3K/Akt signaling pathway contributes to the neuroprotective effects of various neuroprotective agents in cerebral ischemia [262728]. Activation of the Akt pathway is an important neuroprotective mechanism in cerebral ischemia. Chlorogenic acid activates the PI3/Akt survival pathway in hydrogen peroxide-induced oxidative stress [19]. Chlorogenic acid represents anti-inflammatory and anti-apoptotic effects against transient cerebral ischemia [29]. Furthermore, chlorogenic acid binds to Akt and regulates downstream proteins including GSK-3β and FOXO1, and protect glucose metabolism [30]. In the present study, cerebral ischemia significantly reduced phospho-PDK1 expression and continuously reduced phospho-Akt and phospho-Bad expression. These decreases were mitigated by chlorogenic acid treatment. However, total protein levels did not significantly change in animals with MCAO damage. The results indicate the phosphorylation of these proteins is important for activation of the Akt signaling pathway and of a neuroprotective mechanism during cerebral ischemia. Chlorogenic acid regulates the phosphorylation of these proteins in cerebral ischemia. In addition, the attenuation of decreased phospho-Akt and phospho-Bad levels induced by chlorogenic acid was confirmed using immunofluorescence staining. The number of positive cells was decreased in animals with MCAO damage, and chlorogenic acid prevented the decrease. Activation of phospho-Akt and phospho-Bad is important for cell survival. The results of the present study demonstrate that chlorogenic acid activates the PI3K/Akt signaling pathway and contributes to a neuroprotective mechanism in cerebral ischemia. Bad is a pro-apoptotic protein and a representative downstream target of Akt. Growth or survival factors phosphorylate Bad and attenuate the pro-apoptotic function of Bad. Thus, the attenuation of reduced phospho-Akt and phospho-Bad levels induced by chlorogenic acid in animals with MCAO damage shows the neuroprotective mechanism of chlorogenic acid to be associated with the Akt and Bad signaling pathway.\nThe phosphorylation of Bad dissociates Bad from the Bcl-xL and Bad complex. The phospho-Bad binds to 14-3-3 proteins and attenuates the pro-apoptotic function of Bad [12]. 14-3-3 interacts with pro-apoptotic proteins such as Bax and Bad [31]. The binding of phospho-Bad and 14-3-3 continuously inhibits cytochrome c release from mitochondria into the cytoplasm, preventing the apoptotic cascade [32]. Thus, the interaction between phospho-Bad and 14-3-3 proteins is important for cell survival and prevents cell death [33]. Results of immunoprecipitation showed a decrease in the interaction between phospho-Bad and 14-3-3 proteins in cerebral ischemia. Chlorogenic acid alleviated the decrease of phospho-Bad and 14-3-3 binding. The results showed that chlorogenic acid regulates binding of phospho-Bad and 14-3-3 proteins in animals with MCAO damage. Maintenance of phospho-Bad and 14-3-3 binding is important for preventing the apoptotic function of Bad and attenuating cell death [32]. However, information regarding the change in phospho-Bad and 14-3-3 binding in the presence of chlorogenic acid during cerebral ischemia is limited. Chlorogenic acid was shown to modulate phospho-Bad and 14-3-3 binding in an animal model of stroke. In addition, chlorogenic acid alleviated MCAO damage-induced increase in cytochrome c and caspase-3. These findings demonstrated the anti-apoptotic effect of chlorogenic acid on cerebral ischemia.\nIn the present study, chlorogenic acid activated the Akt survival pathway and promoted the interaction between phospho-Bad and 14-3-3 proteins, indicating anti-apoptotic properties. These findings suggest that chlorogenic acid exerts a neuroprotective effect in cerebral ischemia by activating the Akt-Bad signal pathway and maintaining phospho-Bad and 14-3-3 binding." ]
[ "intro", "materials|methods", null, null, null, null, null, null, null, null, null, null, null, null, "results", "discussion" ]
[ "Chlorogenic acid", "ischemic stroke", "neuroprotection" ]
Effectiveness of mRNA Booster Vaccination Against Mild, Moderate, and Severe COVID-19 Caused by the Omicron Variant in a Large, Population-Based, Norwegian Cohort.
36259543
Understanding how booster vaccination can prevent moderate and severe illness without hospitalization is crucial to evaluate the full advantage of mRNA boosters.
BACKGROUND
We followed 85 801 participants (aged 31-81 years) in 2 large population-based cohorts during the Omicron BA.1/2 wave. Information on home testing, PCR testing, and symptoms of coronavirus disease 2019 (COVID-19) was extracted from biweekly questionnaires covering the period 12 January 2022 to 7 April 2022. Vaccination status and data on previous SARS-CoV-2 infection were obtained from national registries. Cox regression was used to estimate the effectiveness of booster vaccination compared to receipt of 2-dose primary series >130 days previously.
METHODS
The effectiveness of booster vaccination increased with increasing severity of COVID-19 and decreased with time since booster vaccination. The effectiveness against severe COVID-19 was reduced from 80.9% shortly after booster vaccination to 63.4% in the period >90 days after vaccination. There was hardly any effect against mild COVID-19. The effectiveness tended to be lower among subjects aged ≥60 years than those aged <50 years.
RESULTS
This is the first population-based study to evaluate booster effectiveness against self-reported mild, moderate, and severe COVID-19. Our findings contribute valuable information on duration of protection and thus timing of additional booster vaccinations.
CONCLUSIONS
[ "Humans", "COVID-19", "RNA, Messenger", "SARS-CoV-2", "Vaccination" ]
9620770
null
null
METHODS
In Norway, the national immunization program against SARS-CoV-2 started on 27 December 2020 [11]. In the primary 2-dose vaccine course, most subjects received the mRNA vaccines Comirnaty (Pfizer/BioNTech; BNT162b2), or Spikevax (Moderna; mRNA-1273), or a combination of the 2. The adenovector-based vaccine Vaxzevria (AstraZeneca; ChAdOx nCoV-19; AZD1222) was only offered through the program until March 2021 [19]. Booster vaccination was recommended from 5 October 2021 to risk groups and people 65 years and older, then to all adults 45 years and older from 26 November 2021. Although not issued as a recommendation, adults aged 18–45 years were also eligible for booster vaccination. Only mRNA vaccines were offered as booster. In December, the minimum interval between dose 2 and the booster dose was reduced from 6 months to 20 weeks [20]. By January 2022, 88% of subjects 18 years and older had received at least 2 vaccine doses, while 51.2% were vaccinated with 3 doses [5]. Among adults 45 years and older, the main target group for booster vaccination, the coverage of 3 doses was 73.3%. Surveillance of SARS-CoV-2 infections in Norway was initially based on mandatory reporting of laboratory-confirmed (polymerase chain reaction [PCR]) infections to the Norwegian Surveillance System for Communicable Disease (MSIS). The high transmission rate of the Omicron variant, limited laboratory capacity, and increased availability of SARS-CoV-2 antigen self-testing kits led the government to recommend self-testing from 12 February 2022 [21]. As a consequence, there has been an increased underreporting of SARS-CoV-2 cases to MSIS. MSIS can therefore no longer be used to assess vaccine effectiveness, as previously done in Norway [11, 12, 22]. Thus, population-based cohorts with frequent follow-up of self-testing, PCR testing, and symptoms are needed for estimates of vaccine effectiveness. [SUBTITLE] The Norwegian Mother, Father, and Child Cohort Study and the Senior Cohort [SUBSECTION] The Norwegian Mother, Father, and Child Cohort Study (MoBa) is a population-based pregnancy cohort study conducted by the Norwegian Institute of Public Health. Participants were recruited from all over Norway during 1999–2008 [23]. The women consented to participation in 41% of the pregnancies. Since March 2020, adult participants have been invited every 14 days to answer electronic questionnaires on the SARS-CoV-2 pandemic. We used data from 5 consecutive questionnaires (Q1–Q5) sent out to 101 765 participants in January-March, 2022. The response rates were 67%–72%. To cover older age groups, a senior cohort was established in December 2020. About 13 000 randomly selected adults, aged 65–80 years living in Oslo, were invited, and 36% consented to participation. Four questionnaires (Q1–Q4), distributed to 4804 participants in January-March 2022, have been included. The response rates were 88%–95%. The participants in the 2 cohorts were asked identical questions about testing for SARS-CoV-2, symptoms of COVID-19, and severity of symptomatic COVID-19. The participants were not given instructions on test frequency or indication. Each questionnaire was open for response for 14 days. The study was approved by The Regional Committee for Medical and Health Research Ethics, Southeast Norway. Written informed consent was obtained from all participants. The Norwegian Mother, Father, and Child Cohort Study (MoBa) is a population-based pregnancy cohort study conducted by the Norwegian Institute of Public Health. Participants were recruited from all over Norway during 1999–2008 [23]. The women consented to participation in 41% of the pregnancies. Since March 2020, adult participants have been invited every 14 days to answer electronic questionnaires on the SARS-CoV-2 pandemic. We used data from 5 consecutive questionnaires (Q1–Q5) sent out to 101 765 participants in January-March, 2022. The response rates were 67%–72%. To cover older age groups, a senior cohort was established in December 2020. About 13 000 randomly selected adults, aged 65–80 years living in Oslo, were invited, and 36% consented to participation. Four questionnaires (Q1–Q4), distributed to 4804 participants in January-March 2022, have been included. The response rates were 88%–95%. The participants in the 2 cohorts were asked identical questions about testing for SARS-CoV-2, symptoms of COVID-19, and severity of symptomatic COVID-19. The participants were not given instructions on test frequency or indication. Each questionnaire was open for response for 14 days. The study was approved by The Regional Committee for Medical and Health Research Ethics, Southeast Norway. Written informed consent was obtained from all participants. [SUBTITLE] Exposure [SUBSECTION] Information on vaccinations was obtained from the Norwegian Immunization Registry. Notification of vaccination against COVID-19 is mandatory. Booster vaccination was defined as a third vaccine dose received at least 130 days after dose 2. According to national recommendations, the minimum interval between the second and third doses should be 20 weeks (140 days). However, we used 130 days as a cutoff because many received the third dose a few days early. In the study population, the vaccination coverage of at least 1 dose was very high (98%), thus the unvaccinated was not a suitable reference group. We therefore compared booster vaccination to the primary series of 2 doses, estimating relative vaccine effectiveness (rVE) [24]. Information on vaccinations was obtained from the Norwegian Immunization Registry. Notification of vaccination against COVID-19 is mandatory. Booster vaccination was defined as a third vaccine dose received at least 130 days after dose 2. According to national recommendations, the minimum interval between the second and third doses should be 20 weeks (140 days). However, we used 130 days as a cutoff because many received the third dose a few days early. In the study population, the vaccination coverage of at least 1 dose was very high (98%), thus the unvaccinated was not a suitable reference group. We therefore compared booster vaccination to the primary series of 2 doses, estimating relative vaccine effectiveness (rVE) [24]. [SUBTITLE] Outcome [SUBSECTION] SARS-CoV-2 infection was defined as a self-reported positive test for SARS-CoV-2. The test could be either a self-sample rapid antigen test or a laboratory test (PCR). COVID-19 was defined as symptomatic SARS-CoV-2 infection. Participants with a SARS-CoV-2 infection were defined as symptomatic if they answered “yes” to the question “have you been feeling ill, had respiratory symptoms, or fever during the last 14 days” in the same questionnaire where they reported the positive test. The severity of COVID-19 was based on the question “how ill did you feel?” and classified as mild illness (“barely ill”), moderate illness (“moderately ill, bedridden for several days”), or severe illness (“very ill”). The infected participants who did not answer this question or who reported no symptoms were only included among all SARS-CoV-2 infections. In Q4 and Q5, participants were asked specifically about the date of the positive SARS-CoV-2 test. In Q1–Q3, participants could only indicate whether they had tested positive at any time during the last 14 days. The participants were also asked how many days prior to filling out the questionnaire potential COVID-19 symptoms occurred (0–1, 2–3, 4–5, 6–7, 8–9, or 10–14 days). We estimated the date of infection by sampling randomly among the candidate dates of symptom onset. For participants who reported a positive test but no symptoms, the date of infection was estimated by sampling randomly from all the 14 dates covered in that questionnaire period. SARS-CoV-2 infection was defined as a self-reported positive test for SARS-CoV-2. The test could be either a self-sample rapid antigen test or a laboratory test (PCR). COVID-19 was defined as symptomatic SARS-CoV-2 infection. Participants with a SARS-CoV-2 infection were defined as symptomatic if they answered “yes” to the question “have you been feeling ill, had respiratory symptoms, or fever during the last 14 days” in the same questionnaire where they reported the positive test. The severity of COVID-19 was based on the question “how ill did you feel?” and classified as mild illness (“barely ill”), moderate illness (“moderately ill, bedridden for several days”), or severe illness (“very ill”). The infected participants who did not answer this question or who reported no symptoms were only included among all SARS-CoV-2 infections. In Q4 and Q5, participants were asked specifically about the date of the positive SARS-CoV-2 test. In Q1–Q3, participants could only indicate whether they had tested positive at any time during the last 14 days. The participants were also asked how many days prior to filling out the questionnaire potential COVID-19 symptoms occurred (0–1, 2–3, 4–5, 6–7, 8–9, or 10–14 days). We estimated the date of infection by sampling randomly among the candidate dates of symptom onset. For participants who reported a positive test but no symptoms, the date of infection was estimated by sampling randomly from all the 14 dates covered in that questionnaire period. [SUBTITLE] Covariates [SUBSECTION] Information on sex, age, and county of residence was obtained from the existing MoBa and senior cohort databases. Information from MSIS was used to identify participants with a previous SARS-CoV-2 infection, defined as a registered infection with a date more than 14 days prior to the participant's start of follow-up (defined below). Information on sex, age, and county of residence was obtained from the existing MoBa and senior cohort databases. Information from MSIS was used to identify participants with a previous SARS-CoV-2 infection, defined as a registered infection with a date more than 14 days prior to the participant's start of follow-up (defined below). [SUBTITLE] Follow-Up and Study Sample [SUBSECTION] In each questionnaire, the participants reported test activity and symptoms during the period from 14 days prior to and up to the fill-in date. Thus, for each questionnaire they returned, the participants could contribute up to 15 days of follow-up time. Participants may have answered the questionnaires at uneven time intervals or not answered all questionnaires and may therefore have questionnaire periods that partly overlap or have gaps between them. Each participant's start of follow-up was defined as the start date of their first questionnaire period or the date corresponding to 130 days after their second dose, whichever occurred last. The participants were followed until the last day of their last questionnaire period, receipt of a fourth vaccine dose, or the date of the first reported SARS-CoV-2 infection, whichever occurred first. The follow-up fell within the periods 12 January to 7 April 2022, for MoBa participants, and 13 January to 24 March 2022 for the senior cohort participants. Only participants answering at least 1 of the questionnaires were eligible for inclusion in the analysis (Figure 1). We excluded 642 participants who were registered with a SARS-CoV-2 infection in MSIS during the study period but did not report a positive test for SARS-CoV-2 on any of the questionnaires. We also excluded 1459 participants registered with an infection in MSIS 14 days or fewer prior to start of follow-up, because it was difficult to differentiate between previous infections and reported outcomes in this period. We furthermore excluded 1335 participants who received dose 3 fewer than 130 days after dose 2, 6 participants who received booster vaccines other than Comirnaty or Spikevax, and 23 participants with missing information on county of residence. Finally, we excluded 4985 participants who did not contribute any follow-up time. In total, 85 801 participants were included in the final study sample (Figure 1). Flow chart of the study population. Abbreviations: MoBa, Norwegian Mother, Father, and Child Cohort Study; MSIS, Norwegian Surveillance System for Communicable Disease; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2. In each questionnaire, the participants reported test activity and symptoms during the period from 14 days prior to and up to the fill-in date. Thus, for each questionnaire they returned, the participants could contribute up to 15 days of follow-up time. Participants may have answered the questionnaires at uneven time intervals or not answered all questionnaires and may therefore have questionnaire periods that partly overlap or have gaps between them. Each participant's start of follow-up was defined as the start date of their first questionnaire period or the date corresponding to 130 days after their second dose, whichever occurred last. The participants were followed until the last day of their last questionnaire period, receipt of a fourth vaccine dose, or the date of the first reported SARS-CoV-2 infection, whichever occurred first. The follow-up fell within the periods 12 January to 7 April 2022, for MoBa participants, and 13 January to 24 March 2022 for the senior cohort participants. Only participants answering at least 1 of the questionnaires were eligible for inclusion in the analysis (Figure 1). We excluded 642 participants who were registered with a SARS-CoV-2 infection in MSIS during the study period but did not report a positive test for SARS-CoV-2 on any of the questionnaires. We also excluded 1459 participants registered with an infection in MSIS 14 days or fewer prior to start of follow-up, because it was difficult to differentiate between previous infections and reported outcomes in this period. We furthermore excluded 1335 participants who received dose 3 fewer than 130 days after dose 2, 6 participants who received booster vaccines other than Comirnaty or Spikevax, and 23 participants with missing information on county of residence. Finally, we excluded 4985 participants who did not contribute any follow-up time. In total, 85 801 participants were included in the final study sample (Figure 1). Flow chart of the study population. Abbreviations: MoBa, Norwegian Mother, Father, and Child Cohort Study; MSIS, Norwegian Surveillance System for Communicable Disease; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2. [SUBTITLE] Statistical Analysis [SUBSECTION] We defined the test frequency for each questionnaire as the number of participants reporting in the questionnaire that they had been tested for SARS-CoV-2 during the last 14 days divided by the number of participants responding to the questionnaire. We calculated test frequency with any test, antigen test, and PCR test (PCR test alone or PCR test plus antigen test). Furthermore, we assessed test frequency by vaccination status at the fill-in date of the questionnaire, sex, and age. We also assessed reasons for testing by vaccination status. Hazard ratios (HRs) and 95% confidence intervals (CIs) for the association between vaccination and risk of SARS-CoV-2 infection were estimated with Cox regression. We used a stratified Cox model with county as strata and calendar time as the underlying time scale. Vaccination was treated as a time-dependent covariate. The reference category consisted of person-time more than 130 days after receipt of the second vaccine dose up until booster vaccination, that is, person-time during which the participants were eligible for, but had not yet received, booster vaccination. Booster vaccination was categorized based on time since vaccination (0–6, 7–30, 31–60, 61–90, 91–120, and >120 days). Full effect of the booster vaccination is not expected until 7 days after vaccination. The model was adjusted for previous SARS-CoV-2 infection, sex, and age group (<40, 40–49, 50–59, and ≥ 60 years). The rVE of booster vaccination compared to 2 doses was calculated as 100% multiplied by (1 − HR). In addition to SARS CoV-2 infection, we also used the categories of COVID-19 severity as outcomes: mild, moderate, and severe. In these analyses, participants were censored at the time of infection if their infection was not included in the severity outcome in question. We performed additional analyses limited to participants with no previous SARS-CoV-2 infection. We also examined the effectiveness of each type of booster vaccine. In this analysis, booster vaccination was categorized both according to time since vaccination and type of vaccine (Comirnaty or Spikevax). We also performed analyses by sex and age (<50, 50–59, and ≥60 years). Here, the categories moderate and severe COVID-19 were combined due to few severe cases. Nonresponse to a questionnaire may be related to the outcome. To assess whether the missingness was informative, we performed a sensitivity analysis including only participants who returned all questionnaires. We defined the test frequency for each questionnaire as the number of participants reporting in the questionnaire that they had been tested for SARS-CoV-2 during the last 14 days divided by the number of participants responding to the questionnaire. We calculated test frequency with any test, antigen test, and PCR test (PCR test alone or PCR test plus antigen test). Furthermore, we assessed test frequency by vaccination status at the fill-in date of the questionnaire, sex, and age. We also assessed reasons for testing by vaccination status. Hazard ratios (HRs) and 95% confidence intervals (CIs) for the association between vaccination and risk of SARS-CoV-2 infection were estimated with Cox regression. We used a stratified Cox model with county as strata and calendar time as the underlying time scale. Vaccination was treated as a time-dependent covariate. The reference category consisted of person-time more than 130 days after receipt of the second vaccine dose up until booster vaccination, that is, person-time during which the participants were eligible for, but had not yet received, booster vaccination. Booster vaccination was categorized based on time since vaccination (0–6, 7–30, 31–60, 61–90, 91–120, and >120 days). Full effect of the booster vaccination is not expected until 7 days after vaccination. The model was adjusted for previous SARS-CoV-2 infection, sex, and age group (<40, 40–49, 50–59, and ≥ 60 years). The rVE of booster vaccination compared to 2 doses was calculated as 100% multiplied by (1 − HR). In addition to SARS CoV-2 infection, we also used the categories of COVID-19 severity as outcomes: mild, moderate, and severe. In these analyses, participants were censored at the time of infection if their infection was not included in the severity outcome in question. We performed additional analyses limited to participants with no previous SARS-CoV-2 infection. We also examined the effectiveness of each type of booster vaccine. In this analysis, booster vaccination was categorized both according to time since vaccination and type of vaccine (Comirnaty or Spikevax). We also performed analyses by sex and age (<50, 50–59, and ≥60 years). Here, the categories moderate and severe COVID-19 were combined due to few severe cases. Nonresponse to a questionnaire may be related to the outcome. To assess whether the missingness was informative, we performed a sensitivity analysis including only participants who returned all questionnaires.
RESULTS
The majority of the 85 801 participants were included in MoBa (94.7%), and 61.3% were women (Table 1). The median age was 49 years (range, 30–81 years); most participants (89.6%) were aged 40–59 years. About 5% had been infected with SARS-CoV-2 prior to the start of follow-up. At the start of follow-up, 23 297 participants (27.2%) had only received 2 vaccine doses, but more than half of these (13 813 participants, 59.3%), received the booster during follow-up. The median follow-up time was 40 days (range, 1–74 days). Characteristics at Start of Follow-up of the Participants in the Norwegian Mother, Father, and Child Cohort Study (MoBa) and Senior Cohort Included in Vaccine Effectiveness Analyses, N = 85 801 Data are No. (%) except where indicated. Participants in MoBa received 5 questionnaires and participants in the senior cohort received 4 questionnaires. Defined as an infection registered in the Norwegian Surveillance System for Communicable Disease with date more than 14 days prior to the participant's start of follow-up. All 85 801 participants included in the analyses had received at least 2 vaccine doses at start of follow-up. Of the participants, 75 911 (88.5%) reported to have been tested for SARS-CoV-2 during the last 14 days on at least 1 questionnaire. Test frequency was highest in Q2 (65.2%) and lowest in Q5 (25.3%) (Supplementary Figure 1). The antigen test frequency was much higher than the PCR test frequency, which decreased from 12.5% (Q1) to 2.2% (Q5) (Supplementary Figure 2). Test frequency was higher among booster-vaccinated participants than among participants with 2 doses, that is, those who had not yet received the booster (Supplementary Figure 3). However, participants with 2 doses were more likely to have taken a PCR test (Supplementary Figure 4). Test frequency was higher among women than among men and decreased with age (Supplementary Figures 5 and 6). Having symptoms was a more common reason for testing among those without a booster (Supplementary Figure 7). In total, 41 462 participants (48.3%) were infected with SARS-CoV-2 during follow-up. Of those infected, 18 519 (44.7%), 20 440 (49.3%), and 1247 (3.0%) reported mild, moderate, and severe COVID-19, respectively. The remaining 1256 cases (3.0%) were either asymptomatic (n = 1102) or not possible to classify (n = 154) because they had not answered the relevant questions. Only 3 of the severe COVID-19 cases reported to have been hospitalized during the same period. The rVE of the booster vaccination increased with increasing severity of symptoms, while it decreased with time since the booster (Table 2 and Figure 2). For SARS-CoV-2 infections, regardless of symptoms, rVE was 41.4% (95% CI, 39.2%–43.5%) 7–30 days after booster vaccination and decreased to 12.2% (95% CI, −2.6% to 24.8%) > 120 days after vaccination. The rVE against mild illness was only 7.9% (95% CI, 2.1%–13.4%) shortly after vaccination and negative >120 days after vaccination. For moderate illness, rVE was 56.0% (95% CI, 53.7%–58.1%) and 27.1% (95% CI, 8.6%–41.8%), respectively, in the 2 time periods. For severe disease, rVE decreased from 80.9% (95% CI, 76.9%–84.2%) 7–30 days after vaccination to 63.4% (95% CI, 50.5%–72.9%) >90 days after vaccination. Relative effectiveness of booster vaccination (third dose) with mRNA vaccine against SARS-CoV-2 infection caused by the Omicron variant compared to a reference group vaccinated with 2 doses. Bars represent 95% confidence intervals of the relative vaccine effectiveness. Effectiveness of Booster Vaccination with mRNA Vaccine Against SARS-CoV-2 Infection and COVID-19 Caused by the Omicron Variant Among Participants in the Norwegian Mother, Father, and Child Cohort Study and the Senior Cohort, N = 85 801 Abbreviations: CI, confidence interval; COVID-19, coronavirus disease 2019; HR, hazard ratio; Ref, reference; rVE, relative vaccine effectiveness; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2. Includes asymptomatic SARS-CoV-2 infections and SARS-CoV-2 infection not possible to classify according to severity. Thus, the number of cases exceeds the sum of mild, moderate, and severe COVID-19 cases. rVE = 100% × (1 − HR). HR was estimated using a stratified Cox model with county as strata and calendar time as the underlying time scale. The model was adjusted for previous SARS-CoV-2 infection, age, and sex. The reference category consisted of person-time more than 130 days after the second vaccine dose up until booster vaccination. Booster vaccination was defined as a third vaccine dose received at least 130 days after the second dose. For severe COVID-19, the upper category was receipt of booster vaccination more than 90 days previously. When we excluded participants with a previous SARS-CoV-2 infection, results were basically unchanged for moderate and severe COVID-19. For mild COVID-19, the negative effect observed in the period >120 days after vaccination was somewhat attenuated (Supplementary Table 1). The effectiveness tended to be higher among men than among women (Table 3). In the period 7–30 days after booster vaccination, the effectiveness against moderate or severe COVID-19 was 62.6% (95% CI, 59.1%–65.8%) among men, as compared to 56.5% (95% CI, 53.9%–59.0%) among women. However, >90 days after vaccination, rVE was slightly higher among women. Effectiveness of Booster Vaccination With mRNA Vaccine Against SARS-CoV-2 Infection and COVID-19 Caused by the Omicron Variant Among Participants in the Norwegian Mother, Father, and Child Cohort Study and the Senior Cohort, by Sex Abbreviations: CI, confidence interval; COVID-19, coronavirus disease 2019; HR, hazard ratio; Ref, reference; rVE, relative vaccine effectiveness; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2. Includes asymptomatic SARS-CoV-2 infections and SARS-CoV-2 infection not possible to classify according to severity. Thus, the number of cases exceeds the sum of mild, moderate, and severe COVID-19 cases. rVE = 100% × (1 − HR). HR was estimated using a stratified Cox model with county as strata and calendar time as the underlying time scale. The model was adjusted for prior SARS-CoV-2 infection and age. The reference category consisted of person-time more than 130 days after the second vaccine dose up until booster vaccination. Booster vaccination was defined as a third vaccine dose received at least 130 days after the second dose. During the first 90 days after booster vaccination, the effectiveness was similar in the 2 youngest age groups (<50 years and 50–59 years; Table 4). However, >90 days after vaccination the effectiveness was lower among the 50–59 year olds. The effectiveness tended to be lower among those ≥60 years as compared to those <50 years, but confidence intervals in the ≥60 years category were wide. Effectiveness of Booster Vaccination With mRNA Vaccine Against SARS-CoV-2 Infection and COVID-19 Caused by the Omicron Variant Among Participants in the Norwegian Mother, Father, and Child Cohort Study and the Senior Cohort, by Age Abbreviations: CI, confidence interval; COVID-19, coronavirus disease 2019; HR, hazard ratio; Ref, reference; rVE, relative vaccine effectiveness; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2. Includes asymptomatic SARS-CoV-2 infections and SARS-CoV-2 infection not possible to classify according to severity. Thus, the number of cases exceeds the sum of mild, moderate, and severe COVID-19 cases. rVE = 100% × (1 − HR). HR was estimated using a stratified Cox model with county as strata and calendar time as the underlying time scale. The model was adjusted for prior SARS-CoV-2 infection, age, and sex. The reference category consisted of person-time more than 130 days after the second vaccine dose up until booster vaccination. Booster vaccination was defined as a third vaccine dose received at least 130 days after the second dose. There was no difference in effectiveness between the 2 mRNA vaccine types used for booster vaccination (Supplementary Table 2). Results from analyses limited to the 48 379 participants who returned all questionnaires were similar to the results for the full study sample (Supplementary Table 3).
null
null
[ "The Norwegian Mother, Father, and Child Cohort Study and the Senior Cohort", "Exposure", "Outcome", "Covariates", "Follow-Up and Study Sample", "Statistical Analysis", "Supplementary Data", "Notes" ]
[ "The Norwegian Mother, Father, and Child Cohort Study (MoBa) is a population-based pregnancy cohort study conducted by the Norwegian Institute of Public Health. Participants were recruited from all over Norway during 1999–2008 [23]. The women consented to participation in 41% of the pregnancies. Since March 2020, adult participants have been invited every 14 days to answer electronic questionnaires on the SARS-CoV-2 pandemic. We used data from 5 consecutive questionnaires (Q1–Q5) sent out to 101 765 participants in January-March, 2022. The response rates were 67%–72%.\nTo cover older age groups, a senior cohort was established in December 2020. About 13 000 randomly selected adults, aged 65–80 years living in Oslo, were invited, and 36% consented to participation. Four questionnaires (Q1–Q4), distributed to 4804 participants in January-March 2022, have been included. The response rates were 88%–95%.\nThe participants in the 2 cohorts were asked identical questions about testing for SARS-CoV-2, symptoms of COVID-19, and severity of symptomatic COVID-19. The participants were not given instructions on test frequency or indication. Each questionnaire was open for response for 14 days.\nThe study was approved by The Regional Committee for Medical and Health Research Ethics, Southeast Norway. Written informed consent was obtained from all participants.", "Information on vaccinations was obtained from the Norwegian Immunization Registry. Notification of vaccination against COVID-19 is mandatory. Booster vaccination was defined as a third vaccine dose received at least 130 days after dose 2. According to national recommendations, the minimum interval between the second and third doses should be 20 weeks (140 days). However, we used 130 days as a cutoff because many received the third dose a few days early. In the study population, the vaccination coverage of at least 1 dose was very high (98%), thus the unvaccinated was not a suitable reference group. We therefore compared booster vaccination to the primary series of 2 doses, estimating relative vaccine effectiveness (rVE) [24].", "SARS-CoV-2 infection was defined as a self-reported positive test for SARS-CoV-2. The test could be either a self-sample rapid antigen test or a laboratory test (PCR).\nCOVID-19 was defined as symptomatic SARS-CoV-2 infection. Participants with a SARS-CoV-2 infection were defined as symptomatic if they answered “yes” to the question “have you been feeling ill, had respiratory symptoms, or fever during the last 14 days” in the same questionnaire where they reported the positive test.\nThe severity of COVID-19 was based on the question “how ill did you feel?” and classified as mild illness (“barely ill”), moderate illness (“moderately ill, bedridden for several days”), or severe illness (“very ill”). The infected participants who did not answer this question or who reported no symptoms were only included among all SARS-CoV-2 infections.\nIn Q4 and Q5, participants were asked specifically about the date of the positive SARS-CoV-2 test. In Q1–Q3, participants could only indicate whether they had tested positive at any time during the last 14 days. The participants were also asked how many days prior to filling out the questionnaire potential COVID-19 symptoms occurred (0–1, 2–3, 4–5, 6–7, 8–9, or 10–14 days). We estimated the date of infection by sampling randomly among the candidate dates of symptom onset. For participants who reported a positive test but no symptoms, the date of infection was estimated by sampling randomly from all the 14 dates covered in that questionnaire period.", "Information on sex, age, and county of residence was obtained from the existing MoBa and senior cohort databases. Information from MSIS was used to identify participants with a previous SARS-CoV-2 infection, defined as a registered infection with a date more than 14 days prior to the participant's start of follow-up (defined below).", "In each questionnaire, the participants reported test activity and symptoms during the period from 14 days prior to and up to the fill-in date. Thus, for each questionnaire they returned, the participants could contribute up to 15 days of follow-up time. Participants may have answered the questionnaires at uneven time intervals or not answered all questionnaires and may therefore have questionnaire periods that partly overlap or have gaps between them.\nEach participant's start of follow-up was defined as the start date of their first questionnaire period or the date corresponding to 130 days after their second dose, whichever occurred last. The participants were followed until the last day of their last questionnaire period, receipt of a fourth vaccine dose, or the date of the first reported SARS-CoV-2 infection, whichever occurred first. The follow-up fell within the periods 12 January to 7 April 2022, for MoBa participants, and 13 January to 24 March 2022 for the senior cohort participants.\nOnly participants answering at least 1 of the questionnaires were eligible for inclusion in the analysis (Figure 1). We excluded 642 participants who were registered with a SARS-CoV-2 infection in MSIS during the study period but did not report a positive test for SARS-CoV-2 on any of the questionnaires. We also excluded 1459 participants registered with an infection in MSIS 14 days or fewer prior to start of follow-up, because it was difficult to differentiate between previous infections and reported outcomes in this period. We furthermore excluded 1335 participants who received dose 3 fewer than 130 days after dose 2, 6 participants who received booster vaccines other than Comirnaty or Spikevax, and 23 participants with missing information on county of residence. Finally, we excluded 4985 participants who did not contribute any follow-up time. In total, 85 801 participants were included in the final study sample (Figure 1).\nFlow chart of the study population. Abbreviations: MoBa, Norwegian Mother, Father, and Child Cohort Study; MSIS, Norwegian Surveillance System for Communicable Disease; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.", "We defined the test frequency for each questionnaire as the number of participants reporting in the questionnaire that they had been tested for SARS-CoV-2 during the last 14 days divided by the number of participants responding to the questionnaire. We calculated test frequency with any test, antigen test, and PCR test (PCR test alone or PCR test plus antigen test). Furthermore, we assessed test frequency by vaccination status at the fill-in date of the questionnaire, sex, and age. We also assessed reasons for testing by vaccination status.\nHazard ratios (HRs) and 95% confidence intervals (CIs) for the association between vaccination and risk of SARS-CoV-2 infection were estimated with Cox regression. We used a stratified Cox model with county as strata and calendar time as the underlying time scale. Vaccination was treated as a time-dependent covariate. The reference category consisted of person-time more than 130 days after receipt of the second vaccine dose up until booster vaccination, that is, person-time during which the participants were eligible for, but had not yet received, booster vaccination. Booster vaccination was categorized based on time since vaccination (0–6, 7–30, 31–60, 61–90, 91–120, and >120 days). Full effect of the booster vaccination is not expected until 7 days after vaccination. The model was adjusted for previous SARS-CoV-2 infection, sex, and age group (<40, 40–49, 50–59, and ≥ 60 years). The rVE of booster vaccination compared to 2 doses was calculated as 100% multiplied by (1 − HR). In addition to SARS CoV-2 infection, we also used the categories of COVID-19 severity as outcomes: mild, moderate, and severe. In these analyses, participants were censored at the time of infection if their infection was not included in the severity outcome in question.\nWe performed additional analyses limited to participants with no previous SARS-CoV-2 infection. We also examined the effectiveness of each type of booster vaccine. In this analysis, booster vaccination was categorized both according to time since vaccination and type of vaccine (Comirnaty or Spikevax). We also performed analyses by sex and age (<50, 50–59, and ≥60 years). Here, the categories moderate and severe COVID-19 were combined due to few severe cases.\nNonresponse to a questionnaire may be related to the outcome. To assess whether the missingness was informative, we performed a sensitivity analysis including only participants who returned all questionnaires.", "\nSupplementary materials are available at The Journal of Infectious Diseases online. Consisting of data provided by the authors to benefit the reader, the posted materials are not copyedited and are the sole responsibility of the authors, so questions or comments should be addressed to the corresponding author.", "\n\nAcknowledgments. We are grateful to all the participants in the Norwegian Mother, Father and Child Cohort Study (MoBa) and the senior cohort. We thank the staff at the Norwegian Institute of Public Health involved in collection and preparation of data and follow-up of cohort participants.\n\n\nFinancial support. This work was partly supported by the Research Council of Norway through its Centers of Excellence funding scheme (project number 262700). MoBa is supported by the Norwegian Ministry of Health and Care Services and the Ministry of Education and Research." ]
[ null, null, null, null, null, null, null, null ]
[ "METHODS", "The Norwegian Mother, Father, and Child Cohort Study and the Senior Cohort", "Exposure", "Outcome", "Covariates", "Follow-Up and Study Sample", "Statistical Analysis", "RESULTS", "DISCUSSION", "Supplementary Data", "Notes", "Supplementary Material" ]
[ "In Norway, the national immunization program against SARS-CoV-2 started on 27 December 2020 [11]. In the primary 2-dose vaccine course, most subjects received the mRNA vaccines Comirnaty (Pfizer/BioNTech; BNT162b2), or Spikevax (Moderna; mRNA-1273), or a combination of the 2. The adenovector-based vaccine Vaxzevria (AstraZeneca; ChAdOx nCoV-19; AZD1222) was only offered through the program until March 2021 [19].\nBooster vaccination was recommended from 5 October 2021 to risk groups and people 65 years and older, then to all adults 45 years and older from 26 November 2021. Although not issued as a recommendation, adults aged 18–45 years were also eligible for booster vaccination. Only mRNA vaccines were offered as booster. In December, the minimum interval between dose 2 and the booster dose was reduced from 6 months to 20 weeks [20]. By January 2022, 88% of subjects 18 years and older had received at least 2 vaccine doses, while 51.2% were vaccinated with 3 doses [5]. Among adults 45 years and older, the main target group for booster vaccination, the coverage of 3 doses was 73.3%.\nSurveillance of SARS-CoV-2 infections in Norway was initially based on mandatory reporting of laboratory-confirmed (polymerase chain reaction [PCR]) infections to the Norwegian Surveillance System for Communicable Disease (MSIS). The high transmission rate of the Omicron variant, limited laboratory capacity, and increased availability of SARS-CoV-2 antigen self-testing kits led the government to recommend self-testing from 12 February 2022 [21]. As a consequence, there has been an increased underreporting of SARS-CoV-2 cases to MSIS. MSIS can therefore no longer be used to assess vaccine effectiveness, as previously done in Norway [11, 12, 22]. Thus, population-based cohorts with frequent follow-up of self-testing, PCR testing, and symptoms are needed for estimates of vaccine effectiveness.\n[SUBTITLE] The Norwegian Mother, Father, and Child Cohort Study and the Senior Cohort [SUBSECTION] The Norwegian Mother, Father, and Child Cohort Study (MoBa) is a population-based pregnancy cohort study conducted by the Norwegian Institute of Public Health. Participants were recruited from all over Norway during 1999–2008 [23]. The women consented to participation in 41% of the pregnancies. Since March 2020, adult participants have been invited every 14 days to answer electronic questionnaires on the SARS-CoV-2 pandemic. We used data from 5 consecutive questionnaires (Q1–Q5) sent out to 101 765 participants in January-March, 2022. The response rates were 67%–72%.\nTo cover older age groups, a senior cohort was established in December 2020. About 13 000 randomly selected adults, aged 65–80 years living in Oslo, were invited, and 36% consented to participation. Four questionnaires (Q1–Q4), distributed to 4804 participants in January-March 2022, have been included. The response rates were 88%–95%.\nThe participants in the 2 cohorts were asked identical questions about testing for SARS-CoV-2, symptoms of COVID-19, and severity of symptomatic COVID-19. The participants were not given instructions on test frequency or indication. Each questionnaire was open for response for 14 days.\nThe study was approved by The Regional Committee for Medical and Health Research Ethics, Southeast Norway. Written informed consent was obtained from all participants.\nThe Norwegian Mother, Father, and Child Cohort Study (MoBa) is a population-based pregnancy cohort study conducted by the Norwegian Institute of Public Health. Participants were recruited from all over Norway during 1999–2008 [23]. The women consented to participation in 41% of the pregnancies. Since March 2020, adult participants have been invited every 14 days to answer electronic questionnaires on the SARS-CoV-2 pandemic. We used data from 5 consecutive questionnaires (Q1–Q5) sent out to 101 765 participants in January-March, 2022. The response rates were 67%–72%.\nTo cover older age groups, a senior cohort was established in December 2020. About 13 000 randomly selected adults, aged 65–80 years living in Oslo, were invited, and 36% consented to participation. Four questionnaires (Q1–Q4), distributed to 4804 participants in January-March 2022, have been included. The response rates were 88%–95%.\nThe participants in the 2 cohorts were asked identical questions about testing for SARS-CoV-2, symptoms of COVID-19, and severity of symptomatic COVID-19. The participants were not given instructions on test frequency or indication. Each questionnaire was open for response for 14 days.\nThe study was approved by The Regional Committee for Medical and Health Research Ethics, Southeast Norway. Written informed consent was obtained from all participants.\n[SUBTITLE] Exposure [SUBSECTION] Information on vaccinations was obtained from the Norwegian Immunization Registry. Notification of vaccination against COVID-19 is mandatory. Booster vaccination was defined as a third vaccine dose received at least 130 days after dose 2. According to national recommendations, the minimum interval between the second and third doses should be 20 weeks (140 days). However, we used 130 days as a cutoff because many received the third dose a few days early. In the study population, the vaccination coverage of at least 1 dose was very high (98%), thus the unvaccinated was not a suitable reference group. We therefore compared booster vaccination to the primary series of 2 doses, estimating relative vaccine effectiveness (rVE) [24].\nInformation on vaccinations was obtained from the Norwegian Immunization Registry. Notification of vaccination against COVID-19 is mandatory. Booster vaccination was defined as a third vaccine dose received at least 130 days after dose 2. According to national recommendations, the minimum interval between the second and third doses should be 20 weeks (140 days). However, we used 130 days as a cutoff because many received the third dose a few days early. In the study population, the vaccination coverage of at least 1 dose was very high (98%), thus the unvaccinated was not a suitable reference group. We therefore compared booster vaccination to the primary series of 2 doses, estimating relative vaccine effectiveness (rVE) [24].\n[SUBTITLE] Outcome [SUBSECTION] SARS-CoV-2 infection was defined as a self-reported positive test for SARS-CoV-2. The test could be either a self-sample rapid antigen test or a laboratory test (PCR).\nCOVID-19 was defined as symptomatic SARS-CoV-2 infection. Participants with a SARS-CoV-2 infection were defined as symptomatic if they answered “yes” to the question “have you been feeling ill, had respiratory symptoms, or fever during the last 14 days” in the same questionnaire where they reported the positive test.\nThe severity of COVID-19 was based on the question “how ill did you feel?” and classified as mild illness (“barely ill”), moderate illness (“moderately ill, bedridden for several days”), or severe illness (“very ill”). The infected participants who did not answer this question or who reported no symptoms were only included among all SARS-CoV-2 infections.\nIn Q4 and Q5, participants were asked specifically about the date of the positive SARS-CoV-2 test. In Q1–Q3, participants could only indicate whether they had tested positive at any time during the last 14 days. The participants were also asked how many days prior to filling out the questionnaire potential COVID-19 symptoms occurred (0–1, 2–3, 4–5, 6–7, 8–9, or 10–14 days). We estimated the date of infection by sampling randomly among the candidate dates of symptom onset. For participants who reported a positive test but no symptoms, the date of infection was estimated by sampling randomly from all the 14 dates covered in that questionnaire period.\nSARS-CoV-2 infection was defined as a self-reported positive test for SARS-CoV-2. The test could be either a self-sample rapid antigen test or a laboratory test (PCR).\nCOVID-19 was defined as symptomatic SARS-CoV-2 infection. Participants with a SARS-CoV-2 infection were defined as symptomatic if they answered “yes” to the question “have you been feeling ill, had respiratory symptoms, or fever during the last 14 days” in the same questionnaire where they reported the positive test.\nThe severity of COVID-19 was based on the question “how ill did you feel?” and classified as mild illness (“barely ill”), moderate illness (“moderately ill, bedridden for several days”), or severe illness (“very ill”). The infected participants who did not answer this question or who reported no symptoms were only included among all SARS-CoV-2 infections.\nIn Q4 and Q5, participants were asked specifically about the date of the positive SARS-CoV-2 test. In Q1–Q3, participants could only indicate whether they had tested positive at any time during the last 14 days. The participants were also asked how many days prior to filling out the questionnaire potential COVID-19 symptoms occurred (0–1, 2–3, 4–5, 6–7, 8–9, or 10–14 days). We estimated the date of infection by sampling randomly among the candidate dates of symptom onset. For participants who reported a positive test but no symptoms, the date of infection was estimated by sampling randomly from all the 14 dates covered in that questionnaire period.\n[SUBTITLE] Covariates [SUBSECTION] Information on sex, age, and county of residence was obtained from the existing MoBa and senior cohort databases. Information from MSIS was used to identify participants with a previous SARS-CoV-2 infection, defined as a registered infection with a date more than 14 days prior to the participant's start of follow-up (defined below).\nInformation on sex, age, and county of residence was obtained from the existing MoBa and senior cohort databases. Information from MSIS was used to identify participants with a previous SARS-CoV-2 infection, defined as a registered infection with a date more than 14 days prior to the participant's start of follow-up (defined below).\n[SUBTITLE] Follow-Up and Study Sample [SUBSECTION] In each questionnaire, the participants reported test activity and symptoms during the period from 14 days prior to and up to the fill-in date. Thus, for each questionnaire they returned, the participants could contribute up to 15 days of follow-up time. Participants may have answered the questionnaires at uneven time intervals or not answered all questionnaires and may therefore have questionnaire periods that partly overlap or have gaps between them.\nEach participant's start of follow-up was defined as the start date of their first questionnaire period or the date corresponding to 130 days after their second dose, whichever occurred last. The participants were followed until the last day of their last questionnaire period, receipt of a fourth vaccine dose, or the date of the first reported SARS-CoV-2 infection, whichever occurred first. The follow-up fell within the periods 12 January to 7 April 2022, for MoBa participants, and 13 January to 24 March 2022 for the senior cohort participants.\nOnly participants answering at least 1 of the questionnaires were eligible for inclusion in the analysis (Figure 1). We excluded 642 participants who were registered with a SARS-CoV-2 infection in MSIS during the study period but did not report a positive test for SARS-CoV-2 on any of the questionnaires. We also excluded 1459 participants registered with an infection in MSIS 14 days or fewer prior to start of follow-up, because it was difficult to differentiate between previous infections and reported outcomes in this period. We furthermore excluded 1335 participants who received dose 3 fewer than 130 days after dose 2, 6 participants who received booster vaccines other than Comirnaty or Spikevax, and 23 participants with missing information on county of residence. Finally, we excluded 4985 participants who did not contribute any follow-up time. In total, 85 801 participants were included in the final study sample (Figure 1).\nFlow chart of the study population. Abbreviations: MoBa, Norwegian Mother, Father, and Child Cohort Study; MSIS, Norwegian Surveillance System for Communicable Disease; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.\nIn each questionnaire, the participants reported test activity and symptoms during the period from 14 days prior to and up to the fill-in date. Thus, for each questionnaire they returned, the participants could contribute up to 15 days of follow-up time. Participants may have answered the questionnaires at uneven time intervals or not answered all questionnaires and may therefore have questionnaire periods that partly overlap or have gaps between them.\nEach participant's start of follow-up was defined as the start date of their first questionnaire period or the date corresponding to 130 days after their second dose, whichever occurred last. The participants were followed until the last day of their last questionnaire period, receipt of a fourth vaccine dose, or the date of the first reported SARS-CoV-2 infection, whichever occurred first. The follow-up fell within the periods 12 January to 7 April 2022, for MoBa participants, and 13 January to 24 March 2022 for the senior cohort participants.\nOnly participants answering at least 1 of the questionnaires were eligible for inclusion in the analysis (Figure 1). We excluded 642 participants who were registered with a SARS-CoV-2 infection in MSIS during the study period but did not report a positive test for SARS-CoV-2 on any of the questionnaires. We also excluded 1459 participants registered with an infection in MSIS 14 days or fewer prior to start of follow-up, because it was difficult to differentiate between previous infections and reported outcomes in this period. We furthermore excluded 1335 participants who received dose 3 fewer than 130 days after dose 2, 6 participants who received booster vaccines other than Comirnaty or Spikevax, and 23 participants with missing information on county of residence. Finally, we excluded 4985 participants who did not contribute any follow-up time. In total, 85 801 participants were included in the final study sample (Figure 1).\nFlow chart of the study population. Abbreviations: MoBa, Norwegian Mother, Father, and Child Cohort Study; MSIS, Norwegian Surveillance System for Communicable Disease; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.\n[SUBTITLE] Statistical Analysis [SUBSECTION] We defined the test frequency for each questionnaire as the number of participants reporting in the questionnaire that they had been tested for SARS-CoV-2 during the last 14 days divided by the number of participants responding to the questionnaire. We calculated test frequency with any test, antigen test, and PCR test (PCR test alone or PCR test plus antigen test). Furthermore, we assessed test frequency by vaccination status at the fill-in date of the questionnaire, sex, and age. We also assessed reasons for testing by vaccination status.\nHazard ratios (HRs) and 95% confidence intervals (CIs) for the association between vaccination and risk of SARS-CoV-2 infection were estimated with Cox regression. We used a stratified Cox model with county as strata and calendar time as the underlying time scale. Vaccination was treated as a time-dependent covariate. The reference category consisted of person-time more than 130 days after receipt of the second vaccine dose up until booster vaccination, that is, person-time during which the participants were eligible for, but had not yet received, booster vaccination. Booster vaccination was categorized based on time since vaccination (0–6, 7–30, 31–60, 61–90, 91–120, and >120 days). Full effect of the booster vaccination is not expected until 7 days after vaccination. The model was adjusted for previous SARS-CoV-2 infection, sex, and age group (<40, 40–49, 50–59, and ≥ 60 years). The rVE of booster vaccination compared to 2 doses was calculated as 100% multiplied by (1 − HR). In addition to SARS CoV-2 infection, we also used the categories of COVID-19 severity as outcomes: mild, moderate, and severe. In these analyses, participants were censored at the time of infection if their infection was not included in the severity outcome in question.\nWe performed additional analyses limited to participants with no previous SARS-CoV-2 infection. We also examined the effectiveness of each type of booster vaccine. In this analysis, booster vaccination was categorized both according to time since vaccination and type of vaccine (Comirnaty or Spikevax). We also performed analyses by sex and age (<50, 50–59, and ≥60 years). Here, the categories moderate and severe COVID-19 were combined due to few severe cases.\nNonresponse to a questionnaire may be related to the outcome. To assess whether the missingness was informative, we performed a sensitivity analysis including only participants who returned all questionnaires.\nWe defined the test frequency for each questionnaire as the number of participants reporting in the questionnaire that they had been tested for SARS-CoV-2 during the last 14 days divided by the number of participants responding to the questionnaire. We calculated test frequency with any test, antigen test, and PCR test (PCR test alone or PCR test plus antigen test). Furthermore, we assessed test frequency by vaccination status at the fill-in date of the questionnaire, sex, and age. We also assessed reasons for testing by vaccination status.\nHazard ratios (HRs) and 95% confidence intervals (CIs) for the association between vaccination and risk of SARS-CoV-2 infection were estimated with Cox regression. We used a stratified Cox model with county as strata and calendar time as the underlying time scale. Vaccination was treated as a time-dependent covariate. The reference category consisted of person-time more than 130 days after receipt of the second vaccine dose up until booster vaccination, that is, person-time during which the participants were eligible for, but had not yet received, booster vaccination. Booster vaccination was categorized based on time since vaccination (0–6, 7–30, 31–60, 61–90, 91–120, and >120 days). Full effect of the booster vaccination is not expected until 7 days after vaccination. The model was adjusted for previous SARS-CoV-2 infection, sex, and age group (<40, 40–49, 50–59, and ≥ 60 years). The rVE of booster vaccination compared to 2 doses was calculated as 100% multiplied by (1 − HR). In addition to SARS CoV-2 infection, we also used the categories of COVID-19 severity as outcomes: mild, moderate, and severe. In these analyses, participants were censored at the time of infection if their infection was not included in the severity outcome in question.\nWe performed additional analyses limited to participants with no previous SARS-CoV-2 infection. We also examined the effectiveness of each type of booster vaccine. In this analysis, booster vaccination was categorized both according to time since vaccination and type of vaccine (Comirnaty or Spikevax). We also performed analyses by sex and age (<50, 50–59, and ≥60 years). Here, the categories moderate and severe COVID-19 were combined due to few severe cases.\nNonresponse to a questionnaire may be related to the outcome. To assess whether the missingness was informative, we performed a sensitivity analysis including only participants who returned all questionnaires.", "The Norwegian Mother, Father, and Child Cohort Study (MoBa) is a population-based pregnancy cohort study conducted by the Norwegian Institute of Public Health. Participants were recruited from all over Norway during 1999–2008 [23]. The women consented to participation in 41% of the pregnancies. Since March 2020, adult participants have been invited every 14 days to answer electronic questionnaires on the SARS-CoV-2 pandemic. We used data from 5 consecutive questionnaires (Q1–Q5) sent out to 101 765 participants in January-March, 2022. The response rates were 67%–72%.\nTo cover older age groups, a senior cohort was established in December 2020. About 13 000 randomly selected adults, aged 65–80 years living in Oslo, were invited, and 36% consented to participation. Four questionnaires (Q1–Q4), distributed to 4804 participants in January-March 2022, have been included. The response rates were 88%–95%.\nThe participants in the 2 cohorts were asked identical questions about testing for SARS-CoV-2, symptoms of COVID-19, and severity of symptomatic COVID-19. The participants were not given instructions on test frequency or indication. Each questionnaire was open for response for 14 days.\nThe study was approved by The Regional Committee for Medical and Health Research Ethics, Southeast Norway. Written informed consent was obtained from all participants.", "Information on vaccinations was obtained from the Norwegian Immunization Registry. Notification of vaccination against COVID-19 is mandatory. Booster vaccination was defined as a third vaccine dose received at least 130 days after dose 2. According to national recommendations, the minimum interval between the second and third doses should be 20 weeks (140 days). However, we used 130 days as a cutoff because many received the third dose a few days early. In the study population, the vaccination coverage of at least 1 dose was very high (98%), thus the unvaccinated was not a suitable reference group. We therefore compared booster vaccination to the primary series of 2 doses, estimating relative vaccine effectiveness (rVE) [24].", "SARS-CoV-2 infection was defined as a self-reported positive test for SARS-CoV-2. The test could be either a self-sample rapid antigen test or a laboratory test (PCR).\nCOVID-19 was defined as symptomatic SARS-CoV-2 infection. Participants with a SARS-CoV-2 infection were defined as symptomatic if they answered “yes” to the question “have you been feeling ill, had respiratory symptoms, or fever during the last 14 days” in the same questionnaire where they reported the positive test.\nThe severity of COVID-19 was based on the question “how ill did you feel?” and classified as mild illness (“barely ill”), moderate illness (“moderately ill, bedridden for several days”), or severe illness (“very ill”). The infected participants who did not answer this question or who reported no symptoms were only included among all SARS-CoV-2 infections.\nIn Q4 and Q5, participants were asked specifically about the date of the positive SARS-CoV-2 test. In Q1–Q3, participants could only indicate whether they had tested positive at any time during the last 14 days. The participants were also asked how many days prior to filling out the questionnaire potential COVID-19 symptoms occurred (0–1, 2–3, 4–5, 6–7, 8–9, or 10–14 days). We estimated the date of infection by sampling randomly among the candidate dates of symptom onset. For participants who reported a positive test but no symptoms, the date of infection was estimated by sampling randomly from all the 14 dates covered in that questionnaire period.", "Information on sex, age, and county of residence was obtained from the existing MoBa and senior cohort databases. Information from MSIS was used to identify participants with a previous SARS-CoV-2 infection, defined as a registered infection with a date more than 14 days prior to the participant's start of follow-up (defined below).", "In each questionnaire, the participants reported test activity and symptoms during the period from 14 days prior to and up to the fill-in date. Thus, for each questionnaire they returned, the participants could contribute up to 15 days of follow-up time. Participants may have answered the questionnaires at uneven time intervals or not answered all questionnaires and may therefore have questionnaire periods that partly overlap or have gaps between them.\nEach participant's start of follow-up was defined as the start date of their first questionnaire period or the date corresponding to 130 days after their second dose, whichever occurred last. The participants were followed until the last day of their last questionnaire period, receipt of a fourth vaccine dose, or the date of the first reported SARS-CoV-2 infection, whichever occurred first. The follow-up fell within the periods 12 January to 7 April 2022, for MoBa participants, and 13 January to 24 March 2022 for the senior cohort participants.\nOnly participants answering at least 1 of the questionnaires were eligible for inclusion in the analysis (Figure 1). We excluded 642 participants who were registered with a SARS-CoV-2 infection in MSIS during the study period but did not report a positive test for SARS-CoV-2 on any of the questionnaires. We also excluded 1459 participants registered with an infection in MSIS 14 days or fewer prior to start of follow-up, because it was difficult to differentiate between previous infections and reported outcomes in this period. We furthermore excluded 1335 participants who received dose 3 fewer than 130 days after dose 2, 6 participants who received booster vaccines other than Comirnaty or Spikevax, and 23 participants with missing information on county of residence. Finally, we excluded 4985 participants who did not contribute any follow-up time. In total, 85 801 participants were included in the final study sample (Figure 1).\nFlow chart of the study population. Abbreviations: MoBa, Norwegian Mother, Father, and Child Cohort Study; MSIS, Norwegian Surveillance System for Communicable Disease; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.", "We defined the test frequency for each questionnaire as the number of participants reporting in the questionnaire that they had been tested for SARS-CoV-2 during the last 14 days divided by the number of participants responding to the questionnaire. We calculated test frequency with any test, antigen test, and PCR test (PCR test alone or PCR test plus antigen test). Furthermore, we assessed test frequency by vaccination status at the fill-in date of the questionnaire, sex, and age. We also assessed reasons for testing by vaccination status.\nHazard ratios (HRs) and 95% confidence intervals (CIs) for the association between vaccination and risk of SARS-CoV-2 infection were estimated with Cox regression. We used a stratified Cox model with county as strata and calendar time as the underlying time scale. Vaccination was treated as a time-dependent covariate. The reference category consisted of person-time more than 130 days after receipt of the second vaccine dose up until booster vaccination, that is, person-time during which the participants were eligible for, but had not yet received, booster vaccination. Booster vaccination was categorized based on time since vaccination (0–6, 7–30, 31–60, 61–90, 91–120, and >120 days). Full effect of the booster vaccination is not expected until 7 days after vaccination. The model was adjusted for previous SARS-CoV-2 infection, sex, and age group (<40, 40–49, 50–59, and ≥ 60 years). The rVE of booster vaccination compared to 2 doses was calculated as 100% multiplied by (1 − HR). In addition to SARS CoV-2 infection, we also used the categories of COVID-19 severity as outcomes: mild, moderate, and severe. In these analyses, participants were censored at the time of infection if their infection was not included in the severity outcome in question.\nWe performed additional analyses limited to participants with no previous SARS-CoV-2 infection. We also examined the effectiveness of each type of booster vaccine. In this analysis, booster vaccination was categorized both according to time since vaccination and type of vaccine (Comirnaty or Spikevax). We also performed analyses by sex and age (<50, 50–59, and ≥60 years). Here, the categories moderate and severe COVID-19 were combined due to few severe cases.\nNonresponse to a questionnaire may be related to the outcome. To assess whether the missingness was informative, we performed a sensitivity analysis including only participants who returned all questionnaires.", "The majority of the 85 801 participants were included in MoBa (94.7%), and 61.3% were women (Table 1). The median age was 49 years (range, 30–81 years); most participants (89.6%) were aged 40–59 years. About 5% had been infected with SARS-CoV-2 prior to the start of follow-up. At the start of follow-up, 23 297 participants (27.2%) had only received 2 vaccine doses, but more than half of these (13 813 participants, 59.3%), received the booster during follow-up. The median follow-up time was 40 days (range, 1–74 days).\nCharacteristics at Start of Follow-up of the Participants in the Norwegian Mother, Father, and Child Cohort Study (MoBa) and Senior Cohort Included in Vaccine Effectiveness Analyses, N = 85 801\nData are No. (%) except where indicated.\nParticipants in MoBa received 5 questionnaires and participants in the senior cohort received 4 questionnaires.\nDefined as an infection registered in the Norwegian Surveillance System for Communicable Disease with date more than 14 days prior to the participant's start of follow-up.\nAll 85 801 participants included in the analyses had received at least 2 vaccine doses at start of follow-up.\nOf the participants, 75 911 (88.5%) reported to have been tested for SARS-CoV-2 during the last 14 days on at least 1 questionnaire. Test frequency was highest in Q2 (65.2%) and lowest in Q5 (25.3%) (Supplementary Figure 1). The antigen test frequency was much higher than the PCR test frequency, which decreased from 12.5% (Q1) to 2.2% (Q5) (Supplementary Figure 2). Test frequency was higher among booster-vaccinated participants than among participants with 2 doses, that is, those who had not yet received the booster (Supplementary Figure 3). However, participants with 2 doses were more likely to have taken a PCR test (Supplementary Figure 4). Test frequency was higher among women than among men and decreased with age (Supplementary Figures 5 and 6). Having symptoms was a more common reason for testing among those without a booster (Supplementary Figure 7).\nIn total, 41 462 participants (48.3%) were infected with SARS-CoV-2 during follow-up. Of those infected, 18 519 (44.7%), 20 440 (49.3%), and 1247 (3.0%) reported mild, moderate, and severe COVID-19, respectively. The remaining 1256 cases (3.0%) were either asymptomatic (n = 1102) or not possible to classify (n = 154) because they had not answered the relevant questions. Only 3 of the severe COVID-19 cases reported to have been hospitalized during the same period.\nThe rVE of the booster vaccination increased with increasing severity of symptoms, while it decreased with time since the booster (Table 2 and Figure 2). For SARS-CoV-2 infections, regardless of symptoms, rVE was 41.4% (95% CI, 39.2%–43.5%) 7–30 days after booster vaccination and decreased to 12.2% (95% CI, −2.6% to 24.8%) > 120 days after vaccination. The rVE against mild illness was only 7.9% (95% CI, 2.1%–13.4%) shortly after vaccination and negative >120 days after vaccination. For moderate illness, rVE was 56.0% (95% CI, 53.7%–58.1%) and 27.1% (95% CI, 8.6%–41.8%), respectively, in the 2 time periods. For severe disease, rVE decreased from 80.9% (95% CI, 76.9%–84.2%) 7–30 days after vaccination to 63.4% (95% CI, 50.5%–72.9%) >90 days after vaccination.\nRelative effectiveness of booster vaccination (third dose) with mRNA vaccine against SARS-CoV-2 infection caused by the Omicron variant compared to a reference group vaccinated with 2 doses. Bars represent 95% confidence intervals of the relative vaccine effectiveness.\nEffectiveness of Booster Vaccination with mRNA Vaccine Against SARS-CoV-2 Infection and COVID-19 Caused by the Omicron Variant Among Participants in the Norwegian Mother, Father, and Child Cohort Study and the Senior Cohort, N = 85 801\nAbbreviations: CI, confidence interval; COVID-19, coronavirus disease 2019; HR, hazard ratio; Ref, reference; rVE, relative vaccine effectiveness; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.\nIncludes asymptomatic SARS-CoV-2 infections and SARS-CoV-2 infection not possible to classify according to severity. Thus, the number of cases exceeds the sum of mild, moderate, and severe COVID-19 cases.\nrVE = 100% × (1 − HR). HR was estimated using a stratified Cox model with county as strata and calendar time as the underlying time scale. The model was adjusted for previous SARS-CoV-2 infection, age, and sex.\nThe reference category consisted of person-time more than 130 days after the second vaccine dose up until booster vaccination.\nBooster vaccination was defined as a third vaccine dose received at least 130 days after the second dose.\nFor severe COVID-19, the upper category was receipt of booster vaccination more than 90 days previously.\nWhen we excluded participants with a previous SARS-CoV-2 infection, results were basically unchanged for moderate and severe COVID-19. For mild COVID-19, the negative effect observed in the period >120 days after vaccination was somewhat attenuated (Supplementary Table 1).\nThe effectiveness tended to be higher among men than among women (Table 3). In the period 7–30 days after booster vaccination, the effectiveness against moderate or severe COVID-19 was 62.6% (95% CI, 59.1%–65.8%) among men, as compared to 56.5% (95% CI, 53.9%–59.0%) among women. However, >90 days after vaccination, rVE was slightly higher among women.\nEffectiveness of Booster Vaccination With mRNA Vaccine Against SARS-CoV-2 Infection and COVID-19 Caused by the Omicron Variant Among Participants in the Norwegian Mother, Father, and Child Cohort Study and the Senior Cohort, by Sex\nAbbreviations: CI, confidence interval; COVID-19, coronavirus disease 2019; HR, hazard ratio; Ref, reference; rVE, relative vaccine effectiveness; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.\nIncludes asymptomatic SARS-CoV-2 infections and SARS-CoV-2 infection not possible to classify according to severity. Thus, the number of cases exceeds the sum of mild, moderate, and severe COVID-19 cases.\nrVE = 100% × (1 − HR). HR was estimated using a stratified Cox model with county as strata and calendar time as the underlying time scale. The model was adjusted for prior SARS-CoV-2 infection and age.\nThe reference category consisted of person-time more than 130 days after the second vaccine dose up until booster vaccination.\nBooster vaccination was defined as a third vaccine dose received at least 130 days after the second dose.\nDuring the first 90 days after booster vaccination, the effectiveness was similar in the 2 youngest age groups (<50 years and 50–59 years; Table 4). However, >90 days after vaccination the effectiveness was lower among the 50–59 year olds. The effectiveness tended to be lower among those ≥60 years as compared to those <50 years, but confidence intervals in the ≥60 years category were wide.\nEffectiveness of Booster Vaccination With mRNA Vaccine Against SARS-CoV-2 Infection and COVID-19 Caused by the Omicron Variant Among Participants in the Norwegian Mother, Father, and Child Cohort Study and the Senior Cohort, by Age\nAbbreviations: CI, confidence interval; COVID-19, coronavirus disease 2019; HR, hazard ratio; Ref, reference; rVE, relative vaccine effectiveness; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2.\nIncludes asymptomatic SARS-CoV-2 infections and SARS-CoV-2 infection not possible to classify according to severity. Thus, the number of cases exceeds the sum of mild, moderate, and severe COVID-19 cases.\nrVE = 100% × (1 − HR). HR was estimated using a stratified Cox model with county as strata and calendar time as the underlying time scale. The model was adjusted for prior SARS-CoV-2 infection, age, and sex.\nThe reference category consisted of person-time more than 130 days after the second vaccine dose up until booster vaccination.\nBooster vaccination was defined as a third vaccine dose received at least 130 days after the second dose.\nThere was no difference in effectiveness between the 2 mRNA vaccine types used for booster vaccination (Supplementary Table 2). Results from analyses limited to the 48 379 participants who returned all questionnaires were similar to the results for the full study sample (Supplementary Table 3).", "In this large, population-based cohort study with more than 85 000 participants, we found that booster vaccination gave substantially better protection against severe and moderate than mild COVID-19, and that the protection decreased with time since vaccination. Furthermore, the effectiveness tended to be lower among those ≥60 years as compared to the younger age groups. We found no difference in effectiveness between the 2 mRNA vaccines used for booster vaccination in Norway.\nIn agreement with recent publications [15, 25, 26], we found that mRNA boosters provide protection against COVID-19 caused by Omicron. Previous studies of booster vaccination have demonstrated better effectiveness against hospitalization/death than against Omicron infection of any severity [26–29].\nWhile some studies report booster effectiveness against Omicron infection compared to unvaccinated subjects [25], it is increasingly common to show VE relative to 2 doses [14, 24, 28, 30–33]. Estimates of rVE with 2 doses as reference will be somewhat lower due to better baseline protection. In the current epidemiological setting, where only a small proportion remain unvaccinated, those who have completed the primary series may be a more representative comparison group. Furthermore, subjects with a completed primary course are the target population for future booster vaccination.\nIn agreement with previous studies [25, 30, 33], we found substantial and rapid waning of the effectiveness against Omicron infection. In a recent surveillance report from the United Kingdom, VE estimates for mRNA booster vaccination against Omicron infection compared to unvaccinated individuals ranged from 60% to 75% in weeks 2–4 after the booster, declining to almost no effect 20 weeks after vaccination [16]. We found that the booster provided substantially better and longer-lasting protection against severe than mild disease. This gradient across self-reported severity within a generally nonhospitalized population has not been demonstrated before. Few of the severe COVID-19 cases in our study were hospitalized. This is not surprising because most participants were younger than 60 years.\nDespite the clear age dependency in COVID-19 vaccination programs, the effectiveness of an mRNA booster by age is not well studied. Some studies have found a similar effect across age categories [28, 34]. In contrast, Kirsebom et al reported slightly greater waning in adults aged ≥65 years compared to 40–64 year olds [35]. Our findings also suggest a lower effectiveness in the oldest age group. However, this group was small and clear conclusions could not be drawn.\nAlmost 90% of the participants reported to have been tested for SARS-CoV2. The participants were much more likely to have taken an antigen test than a PCR test. The specificity of antigen tests is high, but the sensitivity is lower compared to PCR tests [36, 37]. Since January 2022, people with 3 vaccine doses are not recommended a confirmatory PCR test after a positive self-sample test [38]. As expected, booster-vaccinated participants were less likely to have had a PCR test than those with 2 doses. Consequently, the false-negative rate may be higher among booster-vaccinated participants. We cannot rule out that this may have resulted in an overestimation of the vaccine effectiveness. However, the higher rate of false negatives could have been outweighed by the higher test frequency among booster-vaccinated participants. In addition, booster-vaccinated participants were somewhat less likely to get tested due to symptoms. Thus, the detection rate for mild COVID-19 may have been higher among the booster-vaccinated participants, possibly explaining the negative booster effectiveness against mild illness observed >120 days after vaccination. We also found that women were more likely to be tested than men. Higher test frequency and selective testing may explain why the negative booster effectiveness against mild illness was limited to women.\nSome studies have shown different VE by type of mRNA booster [25, 29]. In the present study, different combinations of vaccine types in the primary course were not considered, which probably limits the interpretation of the rVE according to booster type.\nNearly 95% of study participants belong to MoBa, a large, nationwide, population-based cohort that was established many years prior to the SARS-CoV-2 pandemic. In addition, we included older subjects from the newly established senior cohort. The use of electronic questionnaires made it possible to collect data on symptoms and mild infections not requiring medical care and thus not captured by patient registries. Our study covered most of the Omicron BA.1/BA.2 surge in Norway. During this period, antigen test kits were easily available and to a large extent offered for free. Another strength is the linkage to the national immunization registry, which gave detailed and complete information about vaccination dates and brand. In addition, MSIS provided reliable information about the participants’ history of positive laboratory-confirmed PCR tests for SARS-CoV-2 until February 2022. Previous SARS-CoV-2 infection influences both risk of reinfection and likelihood of receiving booster vaccination. Although we adjusted for SARS-CoV-2 infections registered in MSIS prior to start of follow-up, we have not been able to capture all infections. Among participants with no previous SARS-CoV-2 infection, the negative booster effectiveness against mild COVID-19 was attenuated but did not disappear. This suggests that some residual confounding due to previous infection remains.\nInformation on infections after booster vaccination, the outcome in our study, was based on self-report. Although questionnaires were sent out frequently, SARS-CoV-2 infections and COVID-19 cases are still likely to be underreported to some degree.\nBooster vaccination with mRNA vaccine partly prevents moderate and severe COVID-19, but not infection with mild symptoms. Hospitalization with COVID-19 was rare in this study of subjects mainly aged 40–59 years. A better understanding of how booster vaccination can prevent moderate and severe illness without hospitalization is crucial to evaluate the full advantage of an mRNA booster. Ours is the first study to evaluate booster effectiveness against self-reported mild, moderate, and severe COVID-19 in a population-based study, giving a better assessment of the burden of disease due to Omicron infection. The results contribute valuable information to policy makers on duration of protection and thus timing of additional booster vaccinations.", "\nSupplementary materials are available at The Journal of Infectious Diseases online. Consisting of data provided by the authors to benefit the reader, the posted materials are not copyedited and are the sole responsibility of the authors, so questions or comments should be addressed to the corresponding author.", "\n\nAcknowledgments. We are grateful to all the participants in the Norwegian Mother, Father and Child Cohort Study (MoBa) and the senior cohort. We thank the staff at the Norwegian Institute of Public Health involved in collection and preparation of data and follow-up of cohort participants.\n\n\nFinancial support. This work was partly supported by the Research Council of Norway through its Centers of Excellence funding scheme (project number 262700). MoBa is supported by the Norwegian Ministry of Health and Care Services and the Ministry of Education and Research.", "Click here for additional data file." ]
[ "methods", null, null, null, null, null, null, "results", "discussion", null, null, "supplementary-material" ]
[ "SARS-CoV-2", "COVID-19", "booster vaccination", "disease severity", "mRNA vaccine", "MoBa", "vaccine effectiveness" ]
Heterogeneous persistence of Mycobacterium leprae in oral and nasal mucosa of multibacillary patients during multidrug therapy.
36259791
Leprosy is curable by multidrug therapy (MDT) treatment regimen ranging from six to 12 months. The variable levels of tolerance and adherence among patients can, however, result in treatment failure and the emergence of drug-resistant strains.
BACKGROUND
Mycobacterium leprae viability was monitored by quantitative polymerase chain reaction (qPCR) quantification of 16S rRNA in lateral and contralateral scrapings of oral and nasal mucosa of 10 multibacillary patients along the initial five months of treatment.
METHODS
The results demonstrated high heterogenicity of M. leprae viability among patients and between nasal and oral samples. Of six patients who presented good adherence and tolerance to the treatment, only four displayed absence of M. leprae viability in both samples three months after the first MDT dose, while for the other two, the absence of M. leprae viability in the oral and nasal cavities was only detected five months after the first dose.
FINDINGS
We concluded that qPCR of 16S rRNA for the determination of M. leprae viability in nasal and oral scraping samples could represent an interesting approach to monitor treatment efficacy.
MAIN CONCLUSIONS
[ "Humans", "Mycobacterium leprae", "RNA, Ribosomal, 16S", "Leprostatic Agents", "Drug Therapy, Combination", "Nasal Mucosa", "DNA, Bacterial" ]
9575966
null
null
null
null
RESULTS
Information on the clinical and bacteriological characteristics, as well as the treatment schemes, of the patients enrolled in the present study are given in Table. M. leprae viability curves were generated by the RNA/DNA ratio for the M. leprae 16S rRNA gene, as determined by qPCR, from the oral and nasal scrapings of the patients (Figure). Curiously, we observed that, throughout the treatment period, there were peaks of M. leprae viability in both samples. Some of these viability peaks could be explained by the interruption of treatment, represented as a broken line, and indicated in the x-axis of the graphs of patients 5, 8, and 9. This, however, cannot explain the increases seen in the samples of patients 1, 7, and 10, as these patients presented good tolerance to the treatment and received a monthly assisted dose of rifampicin. Mycobacterium leprae molecular viability analysis in oral and nasal scrapings from multibacillary patients. M. leprae molecular viability over the first five months of the multidrug therapy (MDT) regime. For each patient’s temporal series (oral or nasal), the highest viability observed was arbitrary determined as 100% of viable M. leprae load, while 0% indicates the absence of 16S rRNA or absence of viable M. leprae. The black line represents conventional MDT (rifampicin, clofazimine, and dapsone); the red line represents alternative MDT (rifampicin, clofazimine, and ofloxacin); the broken line represents interruption of treatment. The red asterisk indicates erythema nodosum leprosum occurrence; the triangle indicates pneumonia; the square indicates dapsone-associated hemolytic anaemia; the circle indicates treatment-related fever and asthenia. The non-parametric Friedman test was performed by comparing each time point with the next one, where * means p value < 0.05, ** means p < 0.005, and *** means p < 0.001. All patients presented an absence of the 16S rRNA signal in at least one of the samples in the time interval between one and five months after the first treatment dose. It is important to note that even though patients showed a drastic reduction in the viability of the bacillus in their oral mucosa after the first dose of MDT (patients 2, 3, 4, 5, and 6), some demonstrated signs of viable M. leprae in their nasal mucosa up to the second month after the first dose (patients 5 and 6). Considering all the patients who did not have their treatments interrupted, the time to achieve absence of M. leprae viability in both oral and nasal cavities ranged from two to five months after the first dose, except for patient 4, who already displayed an absence of M. leprae viability at 30 days after the first dose of MDT. This time range is extended as patients 7 and 10 curiously showed a viability peak of the bacillus in the nasal cavity samples three and four months, respectively, after the administration of the first dose. The reinfection of these patients’ nasal cavities along treatment by infected household contacts could not be discarded, since two multibacillary leprosy patients were identified in the household contacts of patient 7, one related with patient 9 and one related with patient 10 (Table).The considerable differences observed between the M. leprae viability profiles of the patients spurred the investigation into the potential drug resistance of the M. leprae in the samples by sequencing specific regions of the folP1, gyrA, and rpoB genes. We could not detect any antibiotic resistance polymorphisms (data not shown), which was corroborated by the clinical aspects of these patients at the end of the treatment, as they were all considered cured. This cohort captured the current situation related to the treatment of leprosy. Among the 10 patients followed up in this study, three had complications related to MDT adverse effects: two suffered from dapsone-associated hemolytic anaemia (patients 5 and 9) and another presented treatment-related fever and asthenia (patient 8). The conventional MDT had to be discontinued in these three patients and we observed that this led to an increase in the M. leprae viability in the oral sample of patient 9 and nasal samples of all three patients. Upon resuming treatment with the replacement of dapsone by ofloxacin, a drastic reduction in the viability of M. leprae was observed after the first dose (patient 8) or after the third dose (patients 5 and 9). It is interesting to note that viability increments in the oral scaping, related or not to interruption of treatment, were accompanied by similar viability increments in the nasal scraping, generally with a delay of two months, as observed in patients 7, 8, and 9, or one month in the case of patients 6 and 10. We could also observe the occurrence of two cases of erythema nodosum leprosum during the first months of treatment (patients 4 and 10), apparently without interference in M. leprae viability.
null
null
[]
[]
[]
[ "SUBJECTS AND METHODS", "RESULTS", "DISCUSSION" ]
[ "\nPatients and samples - Ten multibacillary leprosy outpatients (four females and six males; median age: 40 years; age range: 12-62 years) with BI above 3.5 were recruited from the Fiocruz Leprosy Clinic (Rio de Janeiro, Brazil) (Table). Leprosy was diagnosed by clinical, bacteriological, and histological criteria. After signing the Informed Consent Form approved by the Human Research Ethics Committee (No. 1950.165), monthly scrapings (just before and during the first five months of MDT) of both nasal cavities and cheeks using a sterile plastic scraper were collected. All individuals were monitored annually at the Fiocruz Leprosy Clinic after clinical cure and all individuals were considered healthy after five years. All patients presented reactional episodes after cure except for patient 6.\n\nTABLEClinical profile of leprosy patientsPatientSexAgeBI at admissionLeprosy cases / investigated household contacts\n*\n\nTreatment (months)IntercurrenceBI after 12 months of treatmentReactional episode after cure1M3850/4R/C/D (0-12)No1.75Yes2M4450/3R/C/D (0-12)No3.75Yes3F565.50/11R/C/D (0-12)No4.75Yes4F405.751/2R/C/D (0-12)No4.75Yes5M6240/3NT (0-4) R/C/O (4-12)Pneumonia / Hemolytic anaemia4Yes6M4140/4R/C/D (0-12)No2.5No7F1252/4R/C/D (0-12)No4.75Yes8M544.750/11R/C/O (0-12)No2.3Yes9F2051/20R/C/D (0-2) NT (2-4) R/C/O (4-12)Dapsone sensitivity / Hemolytic anaemia4.85Yes10M335.251/11R/C/D (0-12)No3.7YesM: male; F: female; BI: bacilloscopic index; R/C/D: rifampicin, clofazimine, and dapsone (conventional treatment); R/C/O: rifampicin, clofazimine, and ofloxacine (alternative treatment); NT: not treated; *all household contacts leprosy cases were diagnosed as multibacillary.\n\nM: male; F: female; BI: bacilloscopic index; R/C/D: rifampicin, clofazimine, and dapsone (conventional treatment); R/C/O: rifampicin, clofazimine, and ofloxacine (alternative treatment); NT: not treated; *all household contacts leprosy cases were diagnosed as multibacillary.\n\nMycobacterium leprae viability quantification in oral and nasal scrapings - The viability of M. leprae was determined after simultaneously extraction of RNA and DNA from each sample by TRIzolTM reagent (Invitrogen) as described elsewere.\n16\n Briefly, 500 µl of TRIzol™ Reagent (Invitrogen) was immediately added to the collected samples, which were then homogenised by vortexing, incubated for 30 min at room temperature cooled for 5 minutes and then 100 µL of chloroform was added. After rapid mixing by inversion, the tubes were centrifuged at 1200 x g at 4ºC for 15 min. The supernatant aqueous layer containing RNA was transferred to a new tube and precipitated with isoamyl alcohol. The RNA pellet was centrifuged for 30 min at 12,000 x g and washed once with ethanol 70%. The pellet was dissolved in 30 µL of sterile distilled water and stored at -70ºC until use. DNA was purified from the organic phase. 50 µL of 10mM Tris-EDTA solution (pH 8.0) and 75 µL of chloroform-isoamyl alcohol solution (24:1) were added and homogenised by vortexing. After centrifugation at 12,000 x g for 10 min at 4ºC, the aqueous phase was transferred to another tube and the DNA was precipitated with isoamyl alcohol. The DNA pellet was washed in 70% ethanol, centrifuged for 10 min at 10,000 x g, dissolved in 20 µL of sterile distilled water, and stored at -70ºC until use.\nFor the removal of DNA to obtain RNA, samples were treated using the TURBO DNA-free kit (Thermo Fisher Scientific), following the manufacturer’s recommendations. The quantification of RNA and DNA samples was performed using the NanoDrop™ 1000 spectrophotometer (Thermo Fisher Scientific). RNA (500 ng) was then used for cDNA synthesis using the GoScript™ Reverse Transcription System kit (Promega), following the manufacturer’s recommendations. Levels of M. leprae 16S rRNA were determined by real-time PCR (qPCR) using 50 ng of DNA and cDNA. Reactions were performed using TaqMan® PCR Universal Master Mix (Thermo Fisher Scientific), TaqMan® probe and 16S rRNA Primer Mix (Supplementary data - Table), using a StepOne Plus® time PCR system (Applied Biosystems). As a control for DNA contamination of the cDNA, the DNase-treated RNA samples were used in the qPCR reactions in the absence of reverse transcriptase. Additionally, negative (water) and positive controls (purified M. leprae DNA) were added to each experiment. The M. leprae viability was expressed by a twofold Cts value curve (2-ΔΔCT), measuring the levels of 16S rRNA cDNA, and normalising against the bacillus DNA.\nAnalysis was performed in duplicate using the lateral and contralateral of the oral and nasal samples and normalised in the GraphPad Prism 5 program by assigning 100% of viable M. leprae load to the highest value of each temporal analysis. Absence of 16S rRNA transcript detection was determined as 0%. The non-parametric Friedman test was performed by comparing each time point with the next one.\n\nAmplification and sequencing analysis of part of rpoB, folP1, and gyrA - About 200 ng of nasal DNA samples collected at each time point were submitted to PCR using primers targeting three main M. leprae genes related to resistance: folP1, gyrA, and rpoB (Supplementary data - Table) according to Avanzi and collaborators.\n18\n Briefly, DNA was denatured at 94ºC for 15 min, and then amplified by PCR in a Veriti™ 96-Well Thermal Cycler (Applied Biosystems) by submitting samples to six cycles consisting of 94ºC for 45 s, 68ºC for 45 s, 63ºC for 45 s, and 72ºC for 90 s followed by 35 cycles of 94ºC for 45 s, 62ºC for 45 s, and 72ºC for 90 s, with a final extension at 72ºC for 10 min. For evaluation of PCR yield, gel electrophoresis was performed using the PCR product, and amplicons were purified using the QIAquick PCR Purification kit (QIAGEN), followed by sequencing using the ABI PRISM BigDye™ Terminator v 3.0 Sequencing kit (Applied Biosystems) using 30 cycles of 96ºC for 45 s, 50ºC for 5 s, and 60ºC for 10 min. Analysis was performed using the ABI Prism 3730 Automated DNA Sequencer (Applied Biosystems) and the Molecular Evolutionary Genetics Analysis (MEGA) version 7.0 program.", "Information on the clinical and bacteriological characteristics, as well as the treatment schemes, of the patients enrolled in the present study are given in Table. M. leprae viability curves were generated by the RNA/DNA ratio for the M. leprae 16S rRNA gene, as determined by qPCR, from the oral and nasal scrapings of the patients (Figure). Curiously, we observed that, throughout the treatment period, there were peaks of M. leprae viability in both samples. Some of these viability peaks could be explained by the interruption of treatment, represented as a broken line, and indicated in the x-axis of the graphs of patients 5, 8, and 9. This, however, cannot explain the increases seen in the samples of patients 1, 7, and 10, as these patients presented good tolerance to the treatment and received a monthly assisted dose of rifampicin.\n\n\nMycobacterium leprae molecular viability analysis in oral and nasal scrapings from multibacillary patients. M. leprae molecular viability over the first five months of the multidrug therapy (MDT) regime. For each patient’s temporal series (oral or nasal), the highest viability observed was arbitrary determined as 100% of viable M. leprae load, while 0% indicates the absence of 16S rRNA or absence of viable M. leprae. The black line represents conventional MDT (rifampicin, clofazimine, and dapsone); the red line represents alternative MDT (rifampicin, clofazimine, and ofloxacin); the broken line represents interruption of treatment. The red asterisk indicates erythema nodosum leprosum occurrence; the triangle indicates pneumonia; the square indicates dapsone-associated hemolytic anaemia; the circle indicates treatment-related fever and asthenia. The non-parametric Friedman test was performed by comparing each time point with the next one, where * means p value < 0.05, ** means p < 0.005, and *** means p < 0.001.\n\nAll patients presented an absence of the 16S rRNA signal in at least one of the samples in the time interval between one and five months after the first treatment dose. It is important to note that even though patients showed a drastic reduction in the viability of the bacillus in their oral mucosa after the first dose of MDT (patients 2, 3, 4, 5, and 6), some demonstrated signs of viable M. leprae in their nasal mucosa up to the second month after the first dose (patients 5 and 6). Considering all the patients who did not have their treatments interrupted, the time to achieve absence of M. leprae viability in both oral and nasal cavities ranged from two to five months after the first dose, except for patient 4, who already displayed an absence of M. leprae viability at 30 days after the first dose of MDT. This time range is extended as patients 7 and 10 curiously showed a viability peak of the bacillus in the nasal cavity samples three and four months, respectively, after the administration of the first dose. The reinfection of these patients’ nasal cavities along treatment by infected household contacts could not be discarded, since two multibacillary leprosy patients were identified in the household contacts of patient 7, one related with patient 9 and one related with patient 10 (Table).The considerable differences observed between the M. leprae viability profiles of the patients spurred the investigation into the potential drug resistance of the M. leprae in the samples by sequencing specific regions of the folP1, gyrA, and rpoB genes. We could not detect any antibiotic resistance polymorphisms (data not shown), which was corroborated by the clinical aspects of these patients at the end of the treatment, as they were all considered cured.\nThis cohort captured the current situation related to the treatment of leprosy. Among the 10 patients followed up in this study, three had complications related to MDT adverse effects: two suffered from dapsone-associated hemolytic anaemia (patients 5 and 9) and another presented treatment-related fever and asthenia (patient 8). The conventional MDT had to be discontinued in these three patients and we observed that this led to an increase in the M. leprae viability in the oral sample of patient 9 and nasal samples of all three patients. Upon resuming treatment with the replacement of dapsone by ofloxacin, a drastic reduction in the viability of M. leprae was observed after the first dose (patient 8) or after the third dose (patients 5 and 9).\nIt is interesting to note that viability increments in the oral scaping, related or not to interruption of treatment, were accompanied by similar viability increments in the nasal scraping, generally with a delay of two months, as observed in patients 7, 8, and 9, or one month in the case of patients 6 and 10. We could also observe the occurrence of two cases of erythema nodosum leprosum during the first months of treatment (patients 4 and 10), apparently without interference in M. leprae viability.", "Despite the general belief by physicians and health professionals that multibacillary patients stop being a relevant source of infective M. leprae after the administration of the first dose of MDT,\n19\n there is a lack of studies on the viability of M. leprae in the oral and nasal cavities of these patients throughout the treatment.\nThe classic Shepard’s method to monitor M. leprae viability, efficacy of treatment, and drug resistance is based on bacterial growth in the mouse footpad and is a labor-intensive and time-consuming procedure.\n20\n In the present work, we apply qPCR of the 16S rRNA gene to monitor M. leprae viability in clinical samples from 10 patients during the first five months of treatment.\nWe observed that, on the contrary to the common belief, even patients that took the rifampicin dose with the correct periodicity still presented viable M. leprae in their nose and/or mouth from two to four months after the first dose. This data is in accordance with those of Davis and colleagues, who observed a reduction in M. leprae viability only after the administration of five doses of rifampicin in athymic nu/nu mice.\n21\n\n\nNone of the M. leprae strains from this cohort of patients presented a high confidence mutation associated with antibiotic resistance in the folP1, gyrA, or rpoB genes. In accordance with that, none of the patients presented a relapse after five years of follow-up. For this reason, we assume that these variations in M. leprae viability throughout treatment are related to factors other than these known resistance mechanisms of the bacillus to MDT. Such mechanisms involved could be host immunity, the patient’s drug metabolisation profile, genetic background, and/or adherence to treatment, since only the administration of rifampicin was supervised.\nSeveral studies that were based on nucleic acid amplification have demonstrated that nasal cavities are an important reservoir of M. leprae in humans.\n22\n\n,\n\n23\n\n,\n\n24\n\n,\n\n25\n\n,\n\n26\n Using a similar approach, researchers detected M. leprae viability from the nasal swabs of 50% (10 out of 20) of non-treated multibacillary patients.26 In our cohort of patients, we observed that 30% (3 out of 10) of the patient nasal scrapings were negative before treatment (patients 6, 8, and 10). Curiously, all three patients became positive during the treatment period before returning to a negative state again. This indicates that M. leprae viability assessment of the nasal mucosa is a dynamic event, and for that reason PCR assessment of a nasal swab sample taken at a single time point might not be a useful tool in M. leprae diagnosis and/or control.\nAnother limitation of this tool is the persistence of M. leprae viability in the oral or nasal mucosa due to continuous exposition during treatment to infected household contacts, as observed in patients 7, 9 and 10. On the other hand patient 4, which presented a strong and persistent drop in M. leprae viability, in both oral and nasal mucosa, also presented one multibacillary new case among household contacts.\nOur findings also demonstrate a certain correlation of viability profiles in the nose and mouth cavities. Although four patients presented a similar M. leprae viability level in the mouth and nose environment (patients 1, 2, 3, and 4), another four presented a higher persistence of M. leprae viability in the nose. Since the method used to determined M. leprae viability is based on a relation between RNA and DNA, and therefore not strictly related to the number of M. leprae bacilli present, this difference observed between the oral and nasal samples cannot be explained by differences in bacterial load or presence of PCR inhibitors in oral samples. However, earlier studies have demonstrated that many other environmental factors could impact the detection on M. leprae DNA in nasal swabs, such as air humidity.\n27\n\n\nWe observed considerable heterogeneity in the quantity of M. leprae RNA in the mouth and the nose of this cohort of patients ranging from 30 days to five months after the first dose of MDT. We also observed a substantial increase in M. leprae viability after interruption of treatment, followed by a significant drop after implementation of and adherence to an alternative treatment.\nIn conclusion, monitoring of M. leprae viability by 16S rRNA qPCR in nasal and oral scraping samples throughout treatment seems to be a promising tool to monitor treatment efficacy and could be useful for transmission surveillance among patient contacts and in the general population in epidemiological studies." ]
[ "materials|methods", "results", "discussion" ]
[ "leprosy", "nasal scrapings", "oral scrapings", "molecular viability", "qPCR", "treatment" ]
An updated gene regulatory network reconstruction of multidrug-resistant Pseudomonas aeruginosa CCBH4851.
36259790
Healthcare-associated infections due to multidrug-resistant (MDR) bacteria such as Pseudomonas aeruginosa are significant public health issues worldwide. A system biology approach can help understand bacterial behaviour and provide novel ways to identify potential therapeutic targets and develop new drugs. Gene regulatory networks (GRN) are examples of in silico representation of interaction between regulatory genes and their targets.
BACKGROUND
We based this study on the gene orthology inference methodology using the reciprocal best hit method. The P. aeruginosa CCBH4851 genome and GRN, published in 2019, and the P. aeruginosa PAO1 GRN, published in 2020, were used for this update reconstruction process.
METHODS
Our result is a GRN with a greater number of regulatory genes, target genes, and interactions compared to the previous networks, and its structural properties are consistent with the complexity of biological networks and the biological features of P. aeruginosa.
FINDINGS
Here, we present the largest and most complete version of P. aeruginosa GRN published to this date, to the best of our knowledge.
MAIN CONCLUSIONS
[ "Humans", "Pseudomonas aeruginosa", "Gene Regulatory Networks", "Drug Resistance, Multiple, Bacterial", "Cross Infection", "Pseudomonas Infections", "Anti-Bacterial Agents" ]
9565603
null
null
null
null
RESULTS
CCBH-2022 consists of 5452 regulatory interactions among 3186 gene products, of which 218 were identified as regulatory genes and 2968 as target genes. Of the 218 regulatory proteins, 87 are TFs, 19 are sigma factors (SF), and 13 are RNAs. Of these 13 RNAs, 11 are SF as well. The tables containing their relations are presented in the Supplementary data. Given the 6577 predicted protein-coding genes of P. aeruginosa CCBH4851, the model organism in this study, the current network represents roughly 50% of the genome, against 16.52% from CCBH-2019. Specific regulatory genes and their interactions were kept as described in CCBH-2019, such as the ones resulting from the P. aeruginosa PA7 and P. aeruginosa PA14 orthology, and in dedicated biological databases and scientific literature, e.g., IHF (integration host factor). This bacterial DNA-bending protein, essential in gene expression regulation, is absent in the CCBH4851 genome. However, Delic-Attree et al. 69 demonstrated that P. aeruginosa contains the IHF protein composed of the products of the himA and himD genes. These genes act in combination as a TF for several TGs, 2 and all were listed as regulatory genes in CCBH-2019. Consequently, equivalent annotations to the previous CCBH4851 GRN were maintained. CCBH-2022 has 5452 edges, and these interactions were classified into activation (“+”), repression (“-”), dual (“d”, when, depending on the conditions, the regulatory gene act as an activator or a repressor), and unknown (“?”), as described in biological databases and scientific literature. An illustration of CCBH-2022 is presented in Fig. 1. Fig.1:visualisation of CCBH-2022. Yellow circles indicate regulatory genes, light blue circles indicate target genes (TGs), black lines indicate an unknown mode of regulation, green lines indicate activation, and red lines indicate repression. Purple lines indicate a dual-mode of regulation. A: the gene regulatory networks (GRNs) large highly connected network component; B: all regulatory and TGs with no connections with A. Regarding the structural measurements of the updated network, the summarised statistical results are presented in Table I. It contains the standard measures (the number of nodes and edges, number of autoregulatory motifs, network diameter, and average path length), the number of feed-forward motifs, and clustering coefficients. Also, Table I presents a comparison with data from PAO1-2011, CCBH-2019, PAO1-2020, and CCBH-2022. TABLE IComparison of structural statistic measures between PAO1-2011, CCBH-2019, PAO1-2020, CCBH-2022 PAO1-2011CCBH-2019PAO1-2020CCBH-2022Vertices690104630093186Edges1020157650405452Regulatory genes76138173218Target genes59390827092968Positive regulation77977238513829Negative regulation218454390649Dual regulation11131019Unknown regulation12337789955Autoregulation (total)29725091Positive autoregulation16212429Negative autoregulation13391546Unknown autoregulation-121117Feed-forward loop motifs (total) a 137208702968Coherent type I feed-forward loop motifs a 8279226239Incoherent type II feed-forward loop motifs a 34810Density2.12e-031.44e-036.07e-045.99e-04Diameter9121212Average shortest path length04.084.8004.014.67Global clustering coefficient2.28e-023.2e-023.03e-034.42e-03Local clustering coefficient2.5e-011.92e-011.63e-011.87e-01 a: number of feed-forward loop motifs determined using the igraph package. a: number of feed-forward loop motifs determined using the igraph package. Since both CCBH-2022 and PAO1-2020 contain significant updates from their previous counterparts, the comparison between CCBH-2022 and PAO1-2020 is most relevant. CCBH-2022 had a density of 5.99e-04, slightly lower than the density of PAO1-2020 (6.07e-04) but showed the same order of magnitude. The diameter was 12, the same as CCBH-2019 and PAO1-2020 and higher than PAO1-2011, which was 9. The average shortest path distance was 4.67, higher than PAO1-2020 (4.01) but slightly lower than CCBH-2019 (4.80). Similar to the previous GRN, CCBH-2022 was disconnected, showing one large connected component (3102 genes) and 20 small connected components. The degree distributions of the four networks can be seen in Fig. 2A-D, with A and B being the incoming and C and D the outcoming degree distribution. Fig. 2B, D is on a log-log axis, and the straight line is consistent with a power-law distribution. For k-in, the estimated value for γ was 2.79, within the range 2<γ<3, consistent with a power law distribution. For PAO1-2020, the corresponding value was 2.67, 2.89 for CCBH-2019 and 2.71 for PAO1-2011. Fig. 2:graphical representation of structural measurements of CCBH-2022 (red) compared to the previously published networks: PAO1-2011 (purple), CCBH-2019 (orange), and PAO1-2020 (green). (A-B) incoming degree distribution of the four gene regulatory networks (GRNs); (C-D) outgoing distribution of the four GRNs. The distributions are plotted on a linear (A, C) and on a logarithmic scale (B, D); (E) local clustering coefficient distribution; (F) clustering coefficient by degree. The distribution of local clustering coefficients can be seen in Fig. 2E. CCBH-2022 had a global clustering coefficient equal to 4.42e-03, higher than PAO1-2020 (3.03e-03). The scatter plot in Fig. 2F shows the correlation between the local clustering coefficient C(i) and the degree k(i). The most frequent mode of regulation in CCBH-2022 is activation, occurring in 70,2% of the total interactions in the network, followed by roughly 12% of repression mode and 17.8% of dual or unknown mode. Autoregulation occurs when a gene regulates its expression, and the prevalence in CCBH-2022 is of negative autoregulatory motifs. The most abundant motif in all four networks was the coherent type I FFL, with 239 in CCBH-2022 (PAO1-2011: 82; CCBH-2019: 79; PAO1-2020: 226). In addition, there were 10 incoherent type II FFL motifs in CCBH-2022 (PAO1-2011: 3; CCBH-2019: 4; PAO1-2020: 8). Table II shows the 30 most influential hubs in CCBH-2022 and PAO1-2020. An analysis was performed to determine whether the hubs are interconnected through direct interactions (Fig. 3). TABLE IIThe 30 most influential hubs of CCBH-2022 and PAO1-2020CCBH-2020PAO1-2020GeneTotal number of connections (k-out)FunctionGeneTotal number of connections (k-out) rpoD 740Control of expression of housekeeping genes 70 rpoD 749 rpoN 650Nitrogen metabolism, adhesion, quorum sensing (QS), biofilm formation 71 rpoN 658 algU 353Positive regulation of response to oxidative stress 72 algU 357 sigX 298Positive regulation of cell growth 73 sigX 319 rpoS 278QS, Biofilm, virulence, antibiotic resistance 74 fliA 281 fliA 270Adhesion, flagellin biosynthesis 75 rpoS 271 rpoH 184Heat-shock response 76 rpoH 194 gacA 121Monolayer and biofilm formation 77 gacA 128 algR 119Cell motility, biofilm formation 78 algR 122 amrZ 109Cell motility, biofilm formation 79 amrZ 115 lasR 106QS, regulation of elastin catabolic process 80 lasR 95 fleQ 92Regulation of mucin adhesion and flagellar expression 81 pvdS 91 fur 88Control of expression of siderophores and exotoxin A 82 sphR 90 pvdS 87Iron metabolism, pyoverdine, virulence 83 , 84 fleQ 85 sphR 74Sphingosine catabolic process 85 fur 69 mvfR 65QS, regulation of lyase activity, control production of virulence factors 86 vqsM 65 vqsM 61QS, control production of virulence factors 87 mvfR 62 anr 58Regulation of oxidoreductase activity 88 pchR 57 rhlR 56QS, regulation of lipid biosynthetic and proteolysis 89 , 90 anr 53 mexT 53Antibiotic efflux pump 91 mexT 51 pchR 47Regulation of pyochelin siderophore, ferripyochelin receptor synthesis 92 argR 46 argR 46Controls arginine uptake and metabolism 93 fecI 44 gbdR 44Regulation of cellular amino acid metabolic process 94 gbdR 42 pmrA 43Antibiotic efflux pump 95 , 96 rhlR 40 fecI 41Regulation of iron ion transport 97 phoB 40 soxR 40Antibiotic efflux pump 98 pmrA 40 phoB 40Cell motility, regulation of cellular response to phosphate starvation 99 , 100 soxR 39 vfr 37QS, exotoxin A regulator, cell motility 101 dnr 34 dnr 34Regulation of nitrogen compound metabolic process 102 himA 30 rsaL 34QS, biofilm formation, regulation of virulence factors 103 himD 30 Fig.3:connectivity relationships among the 30 most influential hubs of CCBH-2022. Yellow circles indicate regulatory genes considered hubs, light blue circles indicate target genes, black lines indicate an unknown mode of regulation, green lines indicate activation, and red lines indicate repression. Purple lines indicate a dual-mode of regulation.
null
null
[]
[]
[]
[ "MATERIALS AND METHODS", "RESULTS", "DISCUSSION" ]
[ "In this work we study the P. aeruginosa CCBH4851 strain, which is deposited at the Culture Collection of Hospital-Acquired Bacteria (CCBH) located at the Laboratório de Pesquisa em Infecção Hospitalar, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz (Fiocruz) (WDCM947; 39 CGEN022/2010). The genome sequence is available in the GenBank database (Accession CP021380.2).\n14\n\n\nCCBH-2019 and PAO1-2020 models were the bases for the reconstruction of this GRN. CCBH-2022 GRN results from the orthology analysis between the P. aeruginosa PAO1 and CCBH4851 gene sequences. CCBH-2022 model also inherits the orthologs between CCBH4851 and P. aeruginosa PA7\n21\n and P. aeruginosa PA14\n22\n strains, which were already present in CCBH-2019 GRN. The evolutionary histories of genes and species reconstruction are based critically on the accurate identification of orthologs.\n23\n Orthology refers to a specific relationship between homologous characters that arose by speciation at their most recent point of origin,\n24\n\n,\n\n25\n a common ancestor. One of the most common approaches to determining orthology in comparative genomics is the Reciprocal Best Hits (RBH), which relies on BLAST.\n26\n An RBH occurs when two genes from different genomes find themselves the best scoring match in the opposite genome.\n27\n\n,\n\n28\n Regulatory interactions between TFs and TGs in the PAO1 GRN were propagated to CCBH-2022 GRN if the TF and the TG formed an RBH. Medeiros et al.\n2\n designed and implemented an algorithm using the Python programming language to automate and generate a list of RBHs in a tabular format (available as Supplementary data). All the protein sequences from P. aeruginosa CCBH4851 (P1) and P. aeruginosa PAO1 (P2) were considered. BLAST+\n29\n was used to query the proteins from P1 against those from P2 (forward results) and P2 against P1 (reverse results). Each P1 query sequence was considered in turns, and its best match from P2 was identified from forwarding results (x). Likewise, each P2’s query sequence was considered from the reverse results, with its best match in P1 (x’). If x = x’, then they are RBH. Local BLASTP searches of each protein set against the other were executed, with the following cut-off parameters: identity ≥ 90%, coverage ≥ 90%, and E-value ≤ 1 e-5, showing the results in tabular format. If the search returned no hits, the gene was considered to have no ortholog within the opposite genome. Manual BLASTP was used to prevent false negatives, aligning these gene sequences with the opposite genome, considering the above parameters. If they still returned no hits but were present in either PAO1-2020 or CCBH-2019 models, the results were evaluated with a literature search to determine if they were accurate and whether they should be part of CCBH-2022.\nThe final GRN table is available as Supplementary data and is organised into six columns: Regulatory gene, Ortholog of the regulatory gene, Target gene, Ortholog of the target gene, Mode of regulation, and Reference. The first column lists the regulatory genes of P. aeruginosa CCBH4851, while the second column contains orthologs of regulatory genes in the reference strain (PAO1 and PA7 or PA14 from the exclusive interactions in CCBH-2019; the same applies to TG’s orthologs). The third column refers to the target gene in P. aeruginosa CCBH4851, while the fourth column lists orthologs of TGs in the reference strain. Finally, the fifth column describes the mode of regulation, and the sixth column indicates the corresponding data source.\nThe interactions between transcription factor proteins and the genes they regulate in an organism define a directed graph. For the computational analysis, the structure of GRN can be represented as a directed graph, formed by a set of vertices (or nodes) connected by a set of directed edges (or links). Basic network measurements are related to vertex connectivity, the occurrence of cycles, and the distances between pairs of nodes, among other possibilities.\n30\n\n\nThe degree of vertices is the most elementary characterisation of node i, and k(i) is defined as its number of edges. In directed networks, there are incoming (k-in degree) edges and outgoing (k-out degree) edges.\n31\n The degree distribution can follow a functional form P(k) = Ak -γ, called power-law distribution, where P(k) is the likelihood that a randomly chosen node from the network has k direct interactions, A is a constant that ensures that the P(k) values add up to one, and γ is the degree exponent.\n32\n\n,\n\n33\n\n,\n\n34\n\n,\n\n35\n\n,\n\n36\n According to Albert,\n37\n this function indicates high diversity of node degrees, with the P(k) value decaying as a power law that is free of a characteristic scale, resulting in the absence of a typical node in the network that could be used to characterise the rest of the nodes. Most real networks with structural information available exhibit this scale-free behaviour, deviating from a Poisson distribution expected in a classical random network.\n38\n\n,\n\n39\n\n\nStudies have shown a scale-free structure in cellular metabolic networks,\n32\n\n,\n\n40\n protein interaction networks, including in cancer,\n41\n\n,\n\n42\n transcription regulatory networks, and GRN.\n20\n\n,\n\n43\n\n,\n\n44\n\n,\n\n45\n Following the literature,\n36\n\n,\n\n37\n\n,\n\n46\n\n,\n\n47\n\n,\n\n48\n there are some qualitative and quantitative characteristics to ensure that a network is scale-free: the power-law distribution appears as a straight line on a log-log plot; the γ value usually is in the range 2<γ<3; and the presence of high-degree nodes, called hubs, the most highly connected nodes,\n47\n with most nodes clustered around them. The hubs demonstrate the absence of a uniform connectivity distribution in the network, presenting the 80-20 rule (also referred to as the Pareto principle), with small-degree nodes being the most abundant. However, the frequency of high-degree nodes decreases slowly.\n37\n Hubs are fundamental for determining therapeutic targets against an infectious agent.\n2\n Scale-free networks are heterogeneous,\n49\n so random node disruptions generally do not lead to a significant loss of connectivity. However, the loss of the hubs causes the breakdown of the network into isolated clusters.\n50\n Some studies validate these general conclusions for cellular networks.\n51\n\n,\n\n52\n\n,\n\n53\n\n\nIn the GRN, determining the vertices with the highest k-out degrees is a method for identifying a hub,\n2\n The degree threshold is the exact number of interactions that characterise a hub, and this criterion differs from one study to another.\n54\n The degree threshold adopted in this work was the average number of connections of all nodes having at least two edges, resulting in a cut-off value of 16 connections.\nMotifs are connectivity patterns, a small set of recurring regulation patterns from which the networks are built\n55\n\n,\n\n56\n that are associated with specific functions.\n57\n A triangle, i.e., three fully connected vertices, is the simplest type of motif.\n47\n These genes are a regulator, X, which regulates Y, and gene Z, which is regulated by both X and Y.\n58\n Triangles can be closed (three connections within the set) or open (two edges).\n38\n This 3-genes motif is the feedforward loop (FFL) and the most common in GRN, appearing in gene systems in bacteria and other organisms,\n59\n\n,\n\n60\n with the possibility of either activation or repression in each of the three regulatory interactions.\n61\n The coherent type-1 FFL and the incoherent type-2 FFL occur more frequently in transcriptional networks.\n58\n\n\nThe clustering coefficient is the probability that two genes with a common neighbour in a graph are also interconnected.\n19\n This measure has two popular definitions: the local and global clustering coefficient. The local clustering coefficient of vertex i, Ci, is defined as Ci = 2ei / ki (ki - 1), where ei is the number of edges connecting node i with degree ki, and ki (ki - 1) / 2 is the maximum number of edges in the neighbourhood of node i.\n36\n In GRNs, the local clustering coefficient C(i) is interpreted as the interaction between genes forming the regulatory groups.\n2\n The clustering coefficient of a network, C, is calculated by the average of Ci over all vertices.\n19\n\n,\n\n62\n Not considering the directionality of the edges, the global clustering coefficient is the ratio between the number of closed triangles and the total number of triangles (open or closed) in the network.\n2\n C(k) represents the mean clustering coefficient over the vertices with degree k.\n36\n Some biological networks tend to present high clustering coefficient values, e.g., in the protein-protein interaction network of S. cerevisiae, <C> ≈ 0.18.\n30\n\n,\n\n47\n\n\nThe network density measure is the number of edges of the network over the maximum possible number of edges, measuring the interconnectivity between vertices, and is strongly correlated to the potential to generate gene expression heterogeneity.\n63\n The network diameter is the path length between the two most distant nodes.\n36\n The average path length is the measure that indicates the distances between pairs of vertices (the average of the shortest path length over all pairs of nodes in the network).\n46\n\n\nSeveral genes are connected in the GRN. When the nodes interact through a direct or an indirect link (intermediate connections), they are considered part of a connected component. These associations are the concept of network connectivity, and for this analysis in the present work, network interactions were considered undirected.\n2\n\n\nAnalysing the structural characteristics (connected components, hubs, and motifs) can help determine the best approach to disturb a network to promote a desired phenotype in the cell.\n64\n\n\nFor CCBH-2022 structural analyses, the R programming language and RStudio were used.\n65\n Scales, dplyr, tibble, readr and igraph packages were used for data manipulation and plotting the structural analyses.\n2\n\n,\n\n20\n\n,\n\n66\n The igraph library was used to compute most properties described above: the in and out degrees, centrality, clustering coefficients, feed-forward loop motifs, connectivity, cycles, paths, and hierarchical levels analyses.\n67\n\n\nThe illustrations of the GRN, the hub’s network, and the connectivity analysis were made in Cytoscape.\n68\n All figures are presented with higher resolution in the Supplementary data.\nThe codes for the structural analysis in R and for finding RBH in python, implemented by Medeiros et al.,\n2\n and the CCBH-2022 file in CSV format are available as Supplementary data in our Github repository (https://github.com/FioSysBio/CCBH2022).", "CCBH-2022 consists of 5452 regulatory interactions among 3186 gene products, of which 218 were identified as regulatory genes and 2968 as target genes. Of the 218 regulatory proteins, 87 are TFs, 19 are sigma factors (SF), and 13 are RNAs. Of these 13 RNAs, 11 are SF as well. The tables containing their relations are presented in the Supplementary data.\nGiven the 6577 predicted protein-coding genes of P. aeruginosa CCBH4851, the model organism in this study, the current network represents roughly 50% of the genome, against 16.52% from CCBH-2019.\nSpecific regulatory genes and their interactions were kept as described in CCBH-2019, such as the ones resulting from the P. aeruginosa PA7 and P. aeruginosa PA14 orthology, and in dedicated biological databases and scientific literature, e.g., IHF (integration host factor). This bacterial DNA-bending protein, essential in gene expression regulation, is absent in the CCBH4851 genome. However, Delic-Attree et al.\n69\n demonstrated that P. aeruginosa contains the IHF protein composed of the products of the himA and himD genes. These genes act in combination as a TF for several TGs,\n2\n and all were listed as regulatory genes in CCBH-2019. Consequently, equivalent annotations to the previous CCBH4851 GRN were maintained.\nCCBH-2022 has 5452 edges, and these interactions were classified into activation (“+”), repression (“-”), dual (“d”, when, depending on the conditions, the regulatory gene act as an activator or a repressor), and unknown (“?”), as described in biological databases and scientific literature. An illustration of CCBH-2022 is presented in Fig. 1.\n\nFig.1:visualisation of CCBH-2022. Yellow circles indicate regulatory genes, light blue circles indicate target genes (TGs), black lines indicate an unknown mode of regulation, green lines indicate activation, and red lines indicate repression. Purple lines indicate a dual-mode of regulation. A: the gene regulatory networks (GRNs) large highly connected network component; B: all regulatory and TGs with no connections with A.\n\nRegarding the structural measurements of the updated network, the summarised statistical results are presented in Table I. It contains the standard measures (the number of nodes and edges, number of autoregulatory motifs, network diameter, and average path length), the number of feed-forward motifs, and clustering coefficients. Also, Table I presents a comparison with data from PAO1-2011, CCBH-2019, PAO1-2020, and CCBH-2022.\n\nTABLE IComparison of structural statistic measures between PAO1-2011, CCBH-2019, PAO1-2020, CCBH-2022\nPAO1-2011CCBH-2019PAO1-2020CCBH-2022Vertices690104630093186Edges1020157650405452Regulatory genes76138173218Target genes59390827092968Positive regulation77977238513829Negative regulation218454390649Dual regulation11131019Unknown regulation12337789955Autoregulation (total)29725091Positive autoregulation16212429Negative autoregulation13391546Unknown autoregulation-121117Feed-forward loop motifs (total)\na\n\n137208702968Coherent type I feed-forward loop motifs\na\n\n8279226239Incoherent type II feed-forward loop motifs\na\n\n34810Density2.12e-031.44e-036.07e-045.99e-04Diameter9121212Average shortest path length04.084.8004.014.67Global clustering coefficient2.28e-023.2e-023.03e-034.42e-03Local clustering coefficient2.5e-011.92e-011.63e-011.87e-01\na: number of feed-forward loop motifs determined using the igraph package.\n\n\na: number of feed-forward loop motifs determined using the igraph package.\nSince both CCBH-2022 and PAO1-2020 contain significant updates from their previous counterparts, the comparison between CCBH-2022 and PAO1-2020 is most relevant. CCBH-2022 had a density of 5.99e-04, slightly lower than the density of PAO1-2020 (6.07e-04) but showed the same order of magnitude. The diameter was 12, the same as CCBH-2019 and PAO1-2020 and higher than PAO1-2011, which was 9. The average shortest path distance was 4.67, higher than PAO1-2020 (4.01) but slightly lower than CCBH-2019 (4.80). Similar to the previous GRN, CCBH-2022 was disconnected, showing one large connected component (3102 genes) and 20 small connected components.\nThe degree distributions of the four networks can be seen in Fig. 2A-D, with A and B being the incoming and C and D the outcoming degree distribution. Fig. 2B, D is on a log-log axis, and the straight line is consistent with a power-law distribution. For k-in, the estimated value for γ was 2.79, within the range 2<γ<3, consistent with a power law distribution. For PAO1-2020, the corresponding value was 2.67, 2.89 for CCBH-2019 and 2.71 for PAO1-2011.\n\nFig. 2:graphical representation of structural measurements of CCBH-2022 (red) compared to the previously published networks: PAO1-2011 (purple), CCBH-2019 (orange), and PAO1-2020 (green). (A-B) incoming degree distribution of the four gene regulatory networks (GRNs); (C-D) outgoing distribution of the four GRNs. The distributions are plotted on a linear (A, C) and on a logarithmic scale (B, D); (E) local clustering coefficient distribution; (F) clustering coefficient by degree.\n\nThe distribution of local clustering coefficients can be seen in Fig. 2E. CCBH-2022 had a global clustering coefficient equal to 4.42e-03, higher than PAO1-2020 (3.03e-03). The scatter plot in Fig. 2F shows the correlation between the local clustering coefficient C(i) and the degree k(i).\nThe most frequent mode of regulation in CCBH-2022 is activation, occurring in 70,2% of the total interactions in the network, followed by roughly 12% of repression mode and 17.8% of dual or unknown mode. Autoregulation occurs when a gene regulates its expression, and the prevalence in CCBH-2022 is of negative autoregulatory motifs.\nThe most abundant motif in all four networks was the coherent type I FFL, with 239 in CCBH-2022 (PAO1-2011: 82; CCBH-2019: 79; PAO1-2020: 226). In addition, there were 10 incoherent type II FFL motifs in CCBH-2022 (PAO1-2011: 3; CCBH-2019: 4; PAO1-2020: 8).\n\nTable II shows the 30 most influential hubs in CCBH-2022 and PAO1-2020.\nAn analysis was performed to determine whether the hubs are interconnected through direct interactions (Fig. 3).\n\nTABLE IIThe 30 most influential hubs of CCBH-2022 and PAO1-2020CCBH-2020PAO1-2020GeneTotal number of connections (k-out)FunctionGeneTotal number of connections (k-out)\nrpoD\n740Control of expression of housekeeping genes\n70\n\n\nrpoD\n749\nrpoN\n650Nitrogen metabolism, adhesion, quorum sensing (QS), biofilm formation\n71\n\n\nrpoN\n658\nalgU\n353Positive regulation of response to oxidative stress\n72\n\n\nalgU\n357\nsigX\n298Positive regulation of cell growth\n73\n\n\nsigX\n319\nrpoS\n278QS, Biofilm, virulence, antibiotic resistance\n74\n\n\nfliA\n281\nfliA\n270Adhesion, flagellin biosynthesis\n75\n\n\nrpoS\n271\nrpoH\n184Heat-shock response\n76\n\n\nrpoH\n194\ngacA\n121Monolayer and biofilm formation\n77\n\n\ngacA\n128\nalgR\n119Cell motility, biofilm formation\n78\n\n\nalgR\n122\namrZ\n109Cell motility, biofilm formation\n79\n\n\namrZ\n115\nlasR\n106QS, regulation of elastin catabolic process\n80\n\n\nlasR\n95\nfleQ\n92Regulation of mucin adhesion and flagellar expression\n81\n\n\npvdS\n91\nfur\n88Control of expression of siderophores and exotoxin A\n82\n\n\nsphR\n90\npvdS\n87Iron metabolism, pyoverdine, virulence\n83\n\n,\n\n84\n\n\nfleQ\n85\nsphR\n74Sphingosine catabolic process\n85\n\n\nfur\n69\nmvfR\n65QS, regulation of lyase activity, control production of virulence factors\n86\n\n\nvqsM\n65\nvqsM\n61QS, control production of virulence factors\n87\n\n\nmvfR\n62\nanr\n58Regulation of oxidoreductase activity\n88\n\n\npchR\n57\nrhlR\n56QS, regulation of lipid biosynthetic and proteolysis\n89\n\n,\n\n90\n\n\nanr\n53\nmexT\n53Antibiotic efflux pump\n91\n\n\nmexT\n51\npchR\n47Regulation of pyochelin siderophore, ferripyochelin receptor synthesis\n92\n\n\nargR\n46\nargR\n46Controls arginine uptake and metabolism\n93\n\n\nfecI\n44\ngbdR\n44Regulation of cellular amino acid metabolic process\n94\n\n\ngbdR\n42\npmrA\n43Antibiotic efflux pump\n95\n\n,\n\n96\n\n\nrhlR\n40\nfecI\n41Regulation of iron ion transport\n97\n\n\nphoB\n40\nsoxR\n40Antibiotic efflux pump\n98\n\n\npmrA\n40\nphoB\n40Cell motility, regulation of cellular response to phosphate starvation\n99\n\n,\n\n100\n\n\nsoxR\n39\nvfr\n37QS, exotoxin A regulator, cell motility\n101\n\n\ndnr\n34\ndnr\n34Regulation of nitrogen compound metabolic process\n102\n\n\nhimA\n30\nrsaL\n34QS, biofilm formation, regulation of virulence factors\n103\n\n\nhimD\n30\n\n\nFig.3:connectivity relationships among the 30 most influential hubs of CCBH-2022. Yellow circles indicate regulatory genes considered hubs, light blue circles indicate target genes, black lines indicate an unknown mode of regulation, green lines indicate activation, and red lines indicate repression. Purple lines indicate a dual-mode of regulation.\n", "The reconstruction and analysis of the P. aeruginosa GRN contribute to a better understanding of its antibiotic resistance mechanisms. It also contributes to a greater knowledge of related cellular behaviours, such as adaptation and pathogenicity, mainly based on an MDR strain such as CCBH4851.\nIn this work, we have good coverage of roughly 50% of the genome on this updated network. The genome of reference strain PAO1 has 6.2Mbp, and PAO1-2020 has a coverage of 50% as well, with 5040 interactions and 3006 genes.\n20\n However, considering that the CCBH4851 genome has 6.8Mbp and has 5452 edges, and 3186 nodes, we can affirm that, to the best of our knowledge, this study presents the largest GRN of P. aeruginosa that has been assembled to date.\nOn the structural aspects, the charts in Fig. 2 and data in Table I make clear that CCBH-2022 represents a substantial improvement in terms of network completeness and complexity when compared with the previous P. aeruginosa GRNs since it includes more nodes, edges, and network motifs, and when comparing clustering coefficients (Fig. 2E-F). For the in silico approach, the network structural analysis is essential to understand the network architecture and performance.\nThe structural differences between CCBH-2022 and PA01-2020 results from additional information available due to the new version of PAO1 and recent experimental work on characterising the complete closed genome of P. aeruginosa CCBH4851.\n104\n\n\nThe structural measures of CCBH-2022, such as node degree distribution and clustering coefficient, are consistent with a qualitative description of a scale-free network type. Indeed, the degree distribution followed the power-law distribution (Fig. 2B, D): a small number of nodes had many connections (the hubs) and many nodes had few connections.\nThe local clustering coefficient and node degree correlation (Fig. 2F) showed that nodes with lower degrees had greater local clustering coefficients than nodes with higher degrees. These characteristics are representative of several biological processes, e.g., RNA binding.\n105\n\n,\n\n106\n\n\nCCBH-2022 showed a lower density value than PAO1-2020. The density of both GRNs was low due to the dynamic and structural flexibility of the networks, a characteristic typical of natural phenomena-based networks,\n107\n and because the nodes were not all interconnected.\n2\n However, CCBH-2022 density was lower probably because it has 20 small connected components disconnected from the larger one (Fig. 1), while PAO1-2020 had 12 separated components. The variation in the number of connected components is plausible due to their size difference and the biological information about interactions available for the reconstruction.\nAll the previous P. aeruginosa GRNs are disconnected graphs, showing one large connected component and a separated few small connected components, and there may be several reasons for this disconnection in specific points. According to Medeiros et al.,\n2\n interactions among all genes are not expected since some genes in an organism are independent of each other, compartmentalised or global, constitutive or growth phase-dependent, and are triggered in different growth phases, thus resulting in a disconnected network, which corroborates with the observed low density. The reason can also be from loss of existing interactions or a gain of interactions still not fully described from additional strain-specific blocks of genes acquired by horizontal gene transfer.\n108\n The large number of connected components found in CCBH-2022 results from connectivity parameters and the global clustering coefficient. Both structural measures are affected by the same biological behaviours.\n107\n\n\nCCBH-2019 presented more negative regulations than PAO1-2011, a trend that continued between CCBH-2022 and PAO1-2020. Also, the most frequent regulatory activity in CCBH-2022 is activation, but ~50% of the autoregulation was negative, which may be a consequence of the increase in negative autoregulation in the overall network interactions compared to the previous ones. Negative auto-regulation in biological systems is commonly observed.\n109\n The Escherichia coli GRN exhibited the same pattern, with negative autoregulation prevailing concurrently with the positive regulation in the overall network.\n110\n The continuity of biological processes is ensured by positive autoregulation.\n111\n For example, quorum sensing, biofilm formation, secretion of toxins, virulence, and resistance factors production, once initiated, must reach a final stage to have the expected effect.\n2\n In CCBH-2022, genes involved in these processes, such as lasR,\n80\n rhlR,\n90\n\npvdS,\n83\n\nalgU,\n72\n\ndnr\n\n102\n and anr,\n88\n have positive autoregulation (and are amongst the 30 principal hubs).\nNegative cycles are also crucial for life-sustaining cyclic processes such as metabolic processes\n112\n and cellular homeostasis.\n113\n In CCBH-2022, genes involved in arginine metabolism (iscR, desT, lexA, hutC, and mvat)\n110\n showed a predominance of negative mode of autoregulation. Negative autoregulation is associated with cellular stability.\n114\n It rapidly responds to variations in concentrations of proteins, toxins, and (or) metabolites to avoid undesired effects such as the energy cost of unneeded synthesis.\n115\n In CCBH-2022, algZ (transcriptional activator of AlgD, involved in alginate production),\n116\n\nlexA (involved in the SOS response),\n117\n\nmetR (involved in swarming motility and methionine synthesis),\n118\n\n,\n\n119\n\nptxR (affects exotoxin A production)\n120\n and rsaL (quorum-sensing repressor)\n121\n presented negative autoregulatory interactions. Autoregulation is common among genes positioned upstream in GRN with crucial developmental functions.\n122\n\n,\n\n123\n\n\nThe FFL motifs are essential for the modulation of cellular processes according to environmental conditions.\n124\n CCBH-2022 has 968 FFL motifs, which are patterns of structural structures, while PAO1-2020 has 702. There are 239 coherent type I FFL motifs in CCBH-2022, an abundant presence. According to Mangan and Alon\n61\n these motifs act as sign-sensitive delay elements, i.e., a circuit that responds rapidly to step-like stimuli in one direction (ON to OFF) and as a delay to steps in the opposite direction (OFF to ON); the temporary removal of the stimulus ceases transcription, so the activation of expression requires a persistent signal to carry on. The incoherent type II FFL motif was less represented but also found in all the GRNs, with a total of 10 in CCBH-2022. Contrastingly with the coherent FFL, the type II FFL acts as a sign-sensitive accelerator, i.e., a circuit that responds rapidly to step-like stimuli in one direction but not in the other.\n61\n\n\nOne last characteristic revealed by the structural analysis was the presence of hubs. The hub’s network (Fig. 3) shows the connection among their interactions; they are all interconnected and belong to the largest connected component of the GRN (Fig. 1A). This connectivity reflects the importance of the influential genes. The hubs can be considered the basis of the GRN. They are crucial in searching for potential drug targets for developing new drugs, as in direct interaction with their specific targets or for an indirect interaction with the subsequent process regulation triggered by them. CCBH-2022 hubs are mainly associated with efflux pump mechanisms (mexT, pmrA, soxR),\n91\n\n,\n\n95\n\n,\n\n98\n alginate biosynthesis (algU, algR, rpoN),\n125\n and biofilm formation (rpoN, rpoS, gacA, amrZ).\n126\n\n\n\nTable II shows the 30 hubs of PAO1-2020. They are very similar to CCBH-2022 hubs, with some changes in the k-out connections. However, two CCBH-2022 hubs were not in the 30 most influential hubs of PAO1-2020: vfr, a global virulence factor regulator\n127\n that directly regulates 37 genes, and rsaL, associated with bacterial tolerance to antibiotics, including ciprofloxacin and carbenicillin\n128\n which directly regulates 34 genes. In PAO1-2020, vfr directly regulates only 12 target genes, while rsaL regulates 19, being one of these an exclusive PAO1-2020 interaction. There are 25 exclusive vfr interactions in CCBH-2022 and 16 exclusive rsaL interactions compared with PAO1-2020. These distinctions can be explained by the fact that the P. aeruginosa CCBH4851 strain is more virulent and multidrug-resistant and also because CCBH-2022 is 7.6% larger in the number of regulatory interactions (5452) than PAO1-2020 (5040), and 20,7% larger in the number of regulatory genes (218) than PAO1-2020 (173). The tables containing these exclusive interactions are in the Supplementary data. These facts strongly indicate that the operation of the main network hubs is not identical. The functioning of CCBH4851 is different, probably due to the greater influence of these two critical genes associated with multi-drug resistance and antibiotic tolerance mechanisms.\nThe Vfr gene’s role in regulating virulence factor production is related to the production of exotoxin A, a toxin that modifies specific target proteins within mammalian cells and induces necrosis in different tissues and organs in MDR P. aeruginosa infections.\n129\n\n,\n\n130\n The Vfr gene also regulates the las and consequently the rhl quorum-sensing system, two systems that together control the expression of several genes associated with virulence factor production,\n131\n including alkaline protease, exotoxin A, pyocyanin, and rhamnolipid, as well as critical genes such as rpoS (the 5th most influential gene in CCBH-2022).\n132\n The signal receptor (R gene) is one of the essential components of the las and rhl QS systems. It is necessary for coding the transcriptional activator protein (R protein).\n133\n The lasR and rhlR genes are among the 20 principal hubs. In interactions present only in CCBH-2022, vfr regulates genes associated with virulence and alginate production.\n134\n\n\nThe rsaL gene has an important role in P. aeruginosa as a global regulator of quorum sensing, virulence, and biofilm formation.\n103\n\n,\n\n135\n Fan et al.\n128\n showed that the mutation of rsaL increased bacterial tolerance to ciprofloxacin and carbenicillin. In interactions present only in CCBH-2022, rsaL regulates mostly genes of the phz1 and phz2 clusters, showing the control that rsaL has on phenazine expression in P. aeruginosa, driving the production of phenazine-1-carboxylic acid (PCA) which is further converted in the virulence factor pyocyanin.\n136\n\n,\n\n137\n The pyocyanin production contributes to bacterial tolerance to ciprofloxacin and carbenicillin.\n128\n\n\n\nPseudomonas aeruginosa evades antimicrobial activity during treatment and exerts antimicrobial resistance by mainly intrinsic resistance mechanisms. Examples of resistance mechanisms are multi-drug efflux pumps, biofilm synthesis, enzymatic inactivation/degradation, drug permeability restriction, production of beta-lactamases, acquired resistance by a mutation in drug targets, and acquisition of resistance genes via horizontal gene transfer.\n138\n\n\nThere is a directed regulatory connection from alginate biosynthesis to iron metabolism and some antibiotic resistance mechanisms.\n139\n The algU, algR, rpoN, pvdS, and fecI genes are related to these processes\n140\n\n,\n\n141\n and are among the most influential hubs.\n\nPseudomonas aeruginosa has multiple efflux pump systems that prevent the antimicrobial agents from accumulating in adequate concentration to cause an effect in the cell, extruding the drug out.\n138\n Efflux pump systems are associated with resistance to beta-lactams, fluoroquinolones, tetracycline, chloramphenicol, macrolides, and aminoglycosides.\n142\n Differential expression or mutations of efflux system genes are also contributing factors for carbapenem and aminoglycoside resistance.\n143\n The mexT, pmrA, soxR genes, related to multidrug antibiotic efflux pumps, are also amongst the most influential hubs.\nThe fleQ gene is also among the hubs and affects psl (polysaccharide synthesis locus) genes and regulates the efflux pump genes, mexA, mexE, and oprH, by brlR.\n2\n\n,\n\n144\n The psl cluster comprises 15 exopolysaccharide biosynthesis-related genes organised in tandem that are important for biofilm formation.\n145\n\n\nThe mexT and soxR genes positively regulate an efflux pump system, and several virulence factors,\n146\n\n,\n\n147\n and pmrA regulate efflux pumps and the polymyxin B and colistin resistance.\n95\n\n,\n\n148\n\n,\n\n149\n\n\nEfflux pumps also help biofilm formation.\n150\n Biofilms are also related to protection from the host immune system and antibiotic penetration and tolerance, preventing them from entering the microbial population and inhibiting its action as a first-line defense mechanism.\n123\n\n,\n\n151\n\n,\n\n152\n The rpoN, rpoS, gacA, algR and amrZ hubs participate in the regulation of P. aeruginosa biofilm.\nThis system biology approach to characterise the MDR P. aeruginosa CCBH4851 regulatory network may lead to the development of strategies to disrupt the connectivity of these essential processes, thus, possibly decreasing the pathogenicity and suppressing the resistance of this bacterium.\n\nIn conclusion - This manuscript reports the reconstruction and structural analysis of the largest P. aeruginosa regulatory network available in the literature to date. This work can give new insights into identifying novel candidate antibiotic targets and contributes to an increase in our understanding of the behaviour of this bacterium.\nThis network’s dynamic model construction is one of our future studies, intending to help researchers working on experimental drug design and screening. The goal is to predict the dynamic behaviour better and improve the understanding of P. aeruginosa, allowing the simulation of normal and stress conditions to discover potential therapeutic targets and help develop new drugs against P. aeruginosa bacterial infection." ]
[ "materials|methods", "results", "discussion" ]
[ "Pseudomonas aeruginosa", "gene regulatory network", "multidrug resistance", "system biology" ]
Boroxine benzaldehyde complex for pharmaceutical applications probed by electron interactions.
36261319
2,4,6-Tris(4-formylphenyl)boroxine (TFPB) is a substituted boroxine containing a benzaldehyde molecule bonded to each boron atom. Boroxine cages are an emerging class of functional nanostructures used in host-guest chemistry, and benzaldehyde is a potential radiosensitizer. Reactions initiated by low-energy electrons with such complexes may dictate and bring new fundamental knowledge for biomedical and pharmaceutical applications.
RATIONALE
The electron ionization properties of TFPB are investigated using a gas-phase electron-molecule crossed beam apparatus coupled with a reflectron time-of-flight mass spectrometer in an orthogonal geometry. Ionization and threshold energies are experimentally determined by mass spectra acquisition as a function of the electron energy.
METHODS
The abundance of the molecular precursor cation in the mass spectrum at 70 eV is significantly lower than that of the most abundant fragment C7 H5 O+ . Twenty-nine cationic fragments with relative intensities >2% are detected and identified. The appearance energies of six fragment cations are reported, and the experimental first ionization potential is found at 9.46 ± 0.11  eV. Moreover, eight double cations are identified. The present results are supported by quantum chemical calculations based on bound state techniques, electron ionization models and thermodynamic thresholds.
RESULTS
According to these results, the TPFB properties may combine the potential radiosensitizer effect of benzaldehyde with the stability of the boroxine ring.
CONCLUSIONS
[ "Electrons", "Benzaldehydes", "Cations", "Pharmaceutical Preparations" ]
9787961
null
null
null
null
null
null
CONCLUSIONS
TFPB shows rich fragmentation patterns upon electron interactions at 70 eV, including several double‐ionization cations. The precursor cation's relative intensity is significantly lower compared to the most abundant fragment C7H5O+, which indicates that the presence of benzaldehyde as a substituent on the boron atom induces molecular instability when compared to triphenylboroxine (phenyl groups as substituents). Twenty‐nine cationic fragments with relative intensities >2% were detected and identified. The ionization energy of TFPB was found at 9.46±0.11 eV. In addition, it was observed that three of the most intense cationic fragments do not include boron in their molecular composition, suggesting the boroxine ring's stability. Thus, these results suggest that the TFPB properties may combine the potential radiosensitizer effect of benzaldehyde with the stability of the boroxine ring, which might be important in drug delivery. This comprehensive description of electron‐induced dissociation of TFPB may contribute to the development of novel medical strategies, namely in anticancer therapy, including chemo‐radiotherapy protocols.
[ "INTRODUCTION", "EXPERIMENTAL SETUP", "Positive ion formation", "Minor fragments", "Ionization and appearance energies", "PEER REVIEW", "PEER REVIEW" ]
[ "Alongside the relevance in plasma physics, atmospheric sciences, astrophysics and many other fields, the study of electron–molecule interactions at low energies is also motivated by the sparse track of secondary electrons created by the interaction of high‐energy radiation in a biological medium,\n1\n, \n2\n which is an interesting feature for radiotherapy. The sensitivity of tumors to high‐energy radiation together with the generated secondary species is enhanced by the use of chemoagents known as radiosensitizers. In summary, the effect of secondary electrons in the cell environment is ultimately linked to the radiation damage to DNA and other biomolecules, which is supported by several studies.\n3\n, \n4\n, \n5\n\n\nAmong the outcomes of the interaction between low‐energy electrons (LEEs) and biomolecules, two mechanisms can be highlighted depending on the energy range. At very low collision energies (units of eV), electrons can be attached to neutral targets and form reactive temporary negative ions, or resonances, whose dynamics can be usually viewed as a competition between auto‐detachment and nuclear relaxation on the femtosecond scale.\n6\n, \n7\n In the context of radiosensitizers, the temporary negative ions can be dissociative and may decay via dissociative electron attachment (DEA).\n6\n, \n7\n, \n8\n However, if the energy of the incident electron is above the ionization threshold of the target, electron‐impact ionization becomes prevalent, in comparison to electron attachment, and gradually dominates the total cross‐section for electron–molecule scattering. According to Sanche and coworkers,\n1\n when plasmid DNA is irradiated with electrons above 30 eV, cationic fragments can be formed after the precursor ionization and subsequently damage DNA segments.\nSome recent studies have shown the efficiency of LEEs in inducing dissociation in electrophilic compounds through DEA mechanisms yielding reactive radicals that cause DNA damage.\n4\n, \n5\n In recent gas‐phase studies, benzaldehyde has been investigated as a potential radiosensitizer by analyzing single electron–molecule interactions.\n9\n These studies, supported by quantum chemical calculations, suggested rich fragmentation patterns leading to the formation of a vast number of anions.\n9\n Besides, boroxines, also known as boronic acid anhydrides, are the dehydration products of organoboronic acids.\n10\n Recent studies have been conducted to understand the structural interconversion mechanisms of boroxine cages containing pyridine that will enable the optimization of several properties such as porosity and size‐selective molecular recognition.\n11\n, \n12\n Besides, in previous gas‐phase studies, different substituted boroxines were irradiated with electrons above the ionization potential,\n13\n, \n14\n and one DEA study was also performed on triphenylboroxine.\n15\n In these studies, triphenylboroxine was demonstrated to be stable towards fragmentation upon electron interaction,\n15\n and the stability of boroxines is affected by the substituent on the boron atom.\n13\n\n\n2,4,6‐Tris(4‐formylphenyl)boroxine (TFPB) is a substituted boroxine containing a benzaldehyde molecule bonded to each boron atom. Benzaldehyde is an aromatic aldehyde used in the food, cosmetic and chemical industries.\n16\n In addition, due to its chemotherapeutic potential and low toxicity, it has been investigated in patients with advanced carcinomas.\n17\n, \n18\n Several in vitro\n\n18\n, \n19\n and in vivo\n\n20\n studies have been conducted to assess the anticancer effect of benzaldehyde and its derivatives as well as to understand the mechanism of action at the cellular level. Two antitumor mechanisms of benzaldehyde were proposed, namely inhibition of essential nutrient uptake in cancer cells\n21\n and inhibition of glutathione peroxidase.\n22\n, \n23\n The latter mechanism induces an increase of oxidative stress related to the production of highly reactive oxygen species.\nConsidering the stability of triphenylboroxine and the potential radiosensitizer effect of benzaldehyde, a deeper knowledge of LEE interactions with TPFB may help to develop new chemo‐radiotherapy approaches and protocols. In the present paper, we report the positive ion formation upon electron interactions in the gas phase with TFPB using a crossed electron–molecule beam setup composed of a trochoidal electron monochromator (TEM) coupled with an orthogonal reflectron time‐of‐flight mass spectrometer (OReTOFMS). Ionization efficiency curves for the most abundant fragment cations were obtained for energies between 4 and 40 eV with experimental determination of threshold energies. Additionally, at 70 eV, was recorded a mass spectrum.", "The present study was performed using an experimental setup composed of a trochoidal electron monochromator coupled with an OReTOFMS, described in detail elsewhere.\n24\n The electrons are emitted from a tungsten hairpin filament, and a quasi‐monoenergetic electron beam is generated due to an electric field orthogonal to the magnetic field. The stability of this quasi‐monoenergetic electron beam with a resolution of about 180 meV was constantly monitored by a picoampere meter connected to a Faraday plate (ca 100 nA for electron energy of 70 eV). Upon the interaction electrons and molecules, ions are formed and then extracted using an extraction field (ca 1 V/cm) into the ion guide and further mass‐separated in the OReToFMS (ca 800 m/Δm) and detected by a microchannel plate detector.", "Figure 2 shows the positive mass spectrum of TFPB recorded at an incident electron energy of 70 eV.\nTFPB (C21H15B3O6) electron ionization mass spectrum recorded at 70 eV incident electron energy.\nAt 70 eV the processes are well above the ionization threshold of the TFPB molecule; therefore, several fragmentation pathways are accessible at this energy. All fragment peaks were assigned according to the isotopic distribution of the elements, including the natural abundance of 10B/11B. In addition, all cationic peak intensities were normalized according to the C7H5O+ peak signal, which is the most abundant fragment. As far as the authors are aware, no mass spectra obtained by electron ionization have been reported previously.\nComparing the 70 eV spectra of TFPB and triphenylboroxine,\n15\n the precursor cation's relative intensity is significantly lower for TFPB. In the case of triphenylboroxine the most intense cation is the precursor cation followed by Ph2(C6H4)B2O2\n+ (m/z 207) with relative intensity of 0.16. In the present case of TFPB, the most intense fragment is C7H5O+ (m/z 105), and precursor cation is formed with a relative intensity of 0.06. In the triphenylboroxine electron ionization mass spectrum, the precursor cation is the most intense fragment. On the other hand, in the TFPB spectrum the precursor cation presents a relative peak intensity of \n6%. This indicates that the presence of the formyl group attached to the benzene ring (benzaldehyde) induces molecular instability and, thus, a wide range of fragmentation patterns. The list of 29 cationic fragments with relative intensities >2% is presented in Table 1.\nList of cationic fragments with relative intensities >2% under electron ionization at 70 eV electron energy\nIt is also important to remark that the two most abundant cations, C7H5O+ (m/z 105) and C7H6O+ (m/z 106), do not include boron in their chemical structure. This suggests that the boroxine ring is significantly preserved upon electron ionization. We do not discard the possibility of electronic rearrangements, although C7H5O+ can be formed by simple cleavage of C&bond;B bond and C7H6O+ by the cleavage of C‐B bond followed by proton transfer. In addition, eight double cationic fragments are formed (Table 2). The double cations have been assigned regarding 10B isotopic contribution. Precursor double cation is attributed at m/z 197.5; double cation C13H12BO2\n2+ (m/z 105.5) corresponds to the double ionization of cationic species m/z 211.\nList of the observed double‐ionized cationic fragments\nDouble cation m/z 82.5 is assigned as C6H7B2O4\n2+; m/z 77.5, m/z 53.5 and m/z 52.5 are assigned to the double cationic species C8H5B2O2\n2+, C7H6O2+ and C7H5O2+, respectively. The latter two correspond to the double ionization of C7H6O+ and C7H5O+, m/z 107 and m/z 106 respectively. The only boron‐absent double cation identified appears at m/z 38.5 being attributed to the phenyl double cation C6H5\n2+. The lightest identified double cation is attributed to C5H3O2+ (m/z 37.5).", "Apart from the four cations described in Table 1, there were identified 18 more fragments with relative intensities \n<2%. The fragment cations \nC18H15B3O3+ (m/z 312), \nC13H14B2O3+ (m/z 240), \nC14H13B2O2+ (m/z 235), \nC15H2B2O+ (m/z 220), \nC12H9B2O2+ (m/z 207), \nC12H9O2+ (m/z 185), \nC9H5B2O3+ (m/z 183), \nC11H6BO2+ (m/z 181), \nC6H9B2O4+ (m/z 167), \nC6H7B2O4+ (m/z 165), \nC10H6BO+ (m/z 152), \nC6H4BO3+ (m/z 135), \nC2HB2O4+ (m/z 111), \nCBO3+ (m/z 71), \nCHO2+ (m/z 45), \nC2HO+ (m/z 41) and \nCH3O+ or \nH4BO+ (m/z 31) are reported in this electron ionization study.", "The ionization efficiency curves near the threshold were measured for six abundant cations in the mass spectrum at 70 eV. The experimental data of cations m/z 51, m/z 77, m/z 91, m/z 105, m/z 210 and m/z 396 were fitted by the method based on the Wannier law\n14\n to determine the experimental ionization and appearance energies. The experimental ionization energy of TFPB was determined at 9.46 ± 0.11 eV. Figure 3 depicts the experimental data and the respective Wannier‐type fits for the six most intense cations.\nExperimental thresholds for the six most abundant cations. The blue dots represent the experimental data and the green dashed line represents the Wannier‐type fitting function. The values in parentheses correspond to the experimental threshold along with the respective statistical uncertainties. (A) \nCHB2O+ (m/z 51); (B) \nC6H5+ (m/z 77); (C) \nC4BO+ or \nC5H4BO+ (m/z 91); (D) \nC7H5O+ (m/z 105); (E) precursor cation \nC13H11BO2+ (m/z 210); (F) precursor cation \nC21H15B3O6+ (m/z 396) [Color figure can be viewed at wileyonlinelibrary.com]\nThe appearance energy of \nC7H5O+ (m/z 105), which is assigned to C&bond;B cleavage (bond break between the benzaldehyde and the boroxine ring), was found at 10.33 ± 0.15 eV. The appearance energy of phenyl cation (m/z 77) that is related to the cleavage of the formyl group and the C&bond;B bond was observed at 10.7 ± 0.1 eV. The remaining appearance energies of the other cations \nCHB2O+ (m/z 51), \nC13H11BO2+ (m/z 210) and \nC4BO+ or \nC5H4BO+ (m/z 91) were determined at 17.72 ± 0.32, 9.59 ± 0.11 and 10.76 ± 0.61 eV, respectively.\nTheoretical values for the first ionization potentials (IPs) were determined using the outer‐valence Green's function (OVGF) method, together with the 6‐311G(df,p) basis set. The calculations were performed with the gaussian09 package.\n26\n In this approach, the IPs correspond to the absolute values of the OVGF energies of the molecular orbitals of the neutral molecule. The first IP was obtained at 9.49 eV, which is in very good agreement with the appearance energy of the precursor cation (m/z 396). Besides, we conducted auxiliary investigation of the dissociation thresholds associated with some of the most abundant singly ionized fragments, from which we drew a conclusion that is worth mentioning. Due to the complexity of the molecule, we only focused on reactions involving single bond breaks and/or the simplest possible rearrangements. The thresholds were calculated with the G4(MP2) methodology, as implemented in gaussian09, at \nT=313 K. For the cationic fragment m/z 210, our estimate for the reaction\n\nTFPB+e−→C13H11BO2++C8H4B2O4\nis \nΔEth=10.77 eV, which is more than 1 eV higher than the observed appearance energy for that channel. A similar discrepancy was observed for the cationic fragment m/z 105. The calculated threshold for the reaction\n\nTFPB+e−→C7H5O++C14H10B3O5\n\n\ncorresponding to a single C&bond;B bond break was \nΔEth=11.89 eV, about 1.56 eV above the value observed experimentally. These results indicate that parallel reactions may take place. Along the dissociation paths, secondary rearrangements with more than two products should occur, therefore lowering the reaction threshold. As suggested, a parallel reaction leading to formation of boroxine \nH3B3O3 is exothermic by 1217.54 kJ/mol.\n27\n The energy release in a possible lateral reaction will contribute to the thermodynamics lowering. This fact may explain the difference between experimental and theoretical values.", "[SUBTITLE] PEER REVIEW [SUBSECTION] The peer review history for this article is available at https://publons.com/publon/10.1002/rcm.9418.\nThe peer review history for this article is available at https://publons.com/publon/10.1002/rcm.9418.", "The peer review history for this article is available at https://publons.com/publon/10.1002/rcm.9418." ]
[ null, null, null, null, null, null, null ]
[ "INTRODUCTION", "EXPERIMENTAL SETUP", "MATERIALS AND METHODS", "DISCUSSION", "Positive ion formation", "Minor fragments", "Ionization and appearance energies", "CONCLUSIONS", "PEER REVIEW", "PEER REVIEW" ]
[ "Alongside the relevance in plasma physics, atmospheric sciences, astrophysics and many other fields, the study of electron–molecule interactions at low energies is also motivated by the sparse track of secondary electrons created by the interaction of high‐energy radiation in a biological medium,\n1\n, \n2\n which is an interesting feature for radiotherapy. The sensitivity of tumors to high‐energy radiation together with the generated secondary species is enhanced by the use of chemoagents known as radiosensitizers. In summary, the effect of secondary electrons in the cell environment is ultimately linked to the radiation damage to DNA and other biomolecules, which is supported by several studies.\n3\n, \n4\n, \n5\n\n\nAmong the outcomes of the interaction between low‐energy electrons (LEEs) and biomolecules, two mechanisms can be highlighted depending on the energy range. At very low collision energies (units of eV), electrons can be attached to neutral targets and form reactive temporary negative ions, or resonances, whose dynamics can be usually viewed as a competition between auto‐detachment and nuclear relaxation on the femtosecond scale.\n6\n, \n7\n In the context of radiosensitizers, the temporary negative ions can be dissociative and may decay via dissociative electron attachment (DEA).\n6\n, \n7\n, \n8\n However, if the energy of the incident electron is above the ionization threshold of the target, electron‐impact ionization becomes prevalent, in comparison to electron attachment, and gradually dominates the total cross‐section for electron–molecule scattering. According to Sanche and coworkers,\n1\n when plasmid DNA is irradiated with electrons above 30 eV, cationic fragments can be formed after the precursor ionization and subsequently damage DNA segments.\nSome recent studies have shown the efficiency of LEEs in inducing dissociation in electrophilic compounds through DEA mechanisms yielding reactive radicals that cause DNA damage.\n4\n, \n5\n In recent gas‐phase studies, benzaldehyde has been investigated as a potential radiosensitizer by analyzing single electron–molecule interactions.\n9\n These studies, supported by quantum chemical calculations, suggested rich fragmentation patterns leading to the formation of a vast number of anions.\n9\n Besides, boroxines, also known as boronic acid anhydrides, are the dehydration products of organoboronic acids.\n10\n Recent studies have been conducted to understand the structural interconversion mechanisms of boroxine cages containing pyridine that will enable the optimization of several properties such as porosity and size‐selective molecular recognition.\n11\n, \n12\n Besides, in previous gas‐phase studies, different substituted boroxines were irradiated with electrons above the ionization potential,\n13\n, \n14\n and one DEA study was also performed on triphenylboroxine.\n15\n In these studies, triphenylboroxine was demonstrated to be stable towards fragmentation upon electron interaction,\n15\n and the stability of boroxines is affected by the substituent on the boron atom.\n13\n\n\n2,4,6‐Tris(4‐formylphenyl)boroxine (TFPB) is a substituted boroxine containing a benzaldehyde molecule bonded to each boron atom. Benzaldehyde is an aromatic aldehyde used in the food, cosmetic and chemical industries.\n16\n In addition, due to its chemotherapeutic potential and low toxicity, it has been investigated in patients with advanced carcinomas.\n17\n, \n18\n Several in vitro\n\n18\n, \n19\n and in vivo\n\n20\n studies have been conducted to assess the anticancer effect of benzaldehyde and its derivatives as well as to understand the mechanism of action at the cellular level. Two antitumor mechanisms of benzaldehyde were proposed, namely inhibition of essential nutrient uptake in cancer cells\n21\n and inhibition of glutathione peroxidase.\n22\n, \n23\n The latter mechanism induces an increase of oxidative stress related to the production of highly reactive oxygen species.\nConsidering the stability of triphenylboroxine and the potential radiosensitizer effect of benzaldehyde, a deeper knowledge of LEE interactions with TPFB may help to develop new chemo‐radiotherapy approaches and protocols. In the present paper, we report the positive ion formation upon electron interactions in the gas phase with TFPB using a crossed electron–molecule beam setup composed of a trochoidal electron monochromator (TEM) coupled with an orthogonal reflectron time‐of‐flight mass spectrometer (OReTOFMS). Ionization efficiency curves for the most abundant fragment cations were obtained for energies between 4 and 40 eV with experimental determination of threshold energies. Additionally, at 70 eV, was recorded a mass spectrum.", "The present study was performed using an experimental setup composed of a trochoidal electron monochromator coupled with an OReTOFMS, described in detail elsewhere.\n24\n The electrons are emitted from a tungsten hairpin filament, and a quasi‐monoenergetic electron beam is generated due to an electric field orthogonal to the magnetic field. The stability of this quasi‐monoenergetic electron beam with a resolution of about 180 meV was constantly monitored by a picoampere meter connected to a Faraday plate (ca 100 nA for electron energy of 70 eV). Upon the interaction electrons and molecules, ions are formed and then extracted using an extraction field (ca 1 V/cm) into the ion guide and further mass‐separated in the OReToFMS (ca 800 m/Δm) and detected by a microchannel plate detector.", "4‐Formylphenylboronic acid (FPBA) was purchased from Sigma‐Aldrich with stated purity of 95%. The conversion of FPBA to TFPB is a dehydration reaction driven thermally (Figure 1).\nDehydration reaction of 4‐formylphenylboronic acid to 2,4,6‐tris(4‐formylphenyl)boroxine\nWhen a sample is heated, FPBA undergoes a dehydration process forming the trimeric anhydride. At the typical experimental base pressure of about \n5×10−5 Pa, the sample oven was heated to 40°C in order to achieve sufficient sublimation of TFPB. An effusive beam of molecules was formed by a capillary (1 mm in diameter) assembled onto the oven. The ionization energy and threshold energies were experimentally determined using a fitting method based on the Wannier law, and the energy scale was calibrated using the ionization efficiency curve of Ar+, determined by Weitzel et al.\n25\n\n", "[SUBTITLE] Positive ion formation [SUBSECTION] Figure 2 shows the positive mass spectrum of TFPB recorded at an incident electron energy of 70 eV.\nTFPB (C21H15B3O6) electron ionization mass spectrum recorded at 70 eV incident electron energy.\nAt 70 eV the processes are well above the ionization threshold of the TFPB molecule; therefore, several fragmentation pathways are accessible at this energy. All fragment peaks were assigned according to the isotopic distribution of the elements, including the natural abundance of 10B/11B. In addition, all cationic peak intensities were normalized according to the C7H5O+ peak signal, which is the most abundant fragment. As far as the authors are aware, no mass spectra obtained by electron ionization have been reported previously.\nComparing the 70 eV spectra of TFPB and triphenylboroxine,\n15\n the precursor cation's relative intensity is significantly lower for TFPB. In the case of triphenylboroxine the most intense cation is the precursor cation followed by Ph2(C6H4)B2O2\n+ (m/z 207) with relative intensity of 0.16. In the present case of TFPB, the most intense fragment is C7H5O+ (m/z 105), and precursor cation is formed with a relative intensity of 0.06. In the triphenylboroxine electron ionization mass spectrum, the precursor cation is the most intense fragment. On the other hand, in the TFPB spectrum the precursor cation presents a relative peak intensity of \n6%. This indicates that the presence of the formyl group attached to the benzene ring (benzaldehyde) induces molecular instability and, thus, a wide range of fragmentation patterns. The list of 29 cationic fragments with relative intensities >2% is presented in Table 1.\nList of cationic fragments with relative intensities >2% under electron ionization at 70 eV electron energy\nIt is also important to remark that the two most abundant cations, C7H5O+ (m/z 105) and C7H6O+ (m/z 106), do not include boron in their chemical structure. This suggests that the boroxine ring is significantly preserved upon electron ionization. We do not discard the possibility of electronic rearrangements, although C7H5O+ can be formed by simple cleavage of C&bond;B bond and C7H6O+ by the cleavage of C‐B bond followed by proton transfer. In addition, eight double cationic fragments are formed (Table 2). The double cations have been assigned regarding 10B isotopic contribution. Precursor double cation is attributed at m/z 197.5; double cation C13H12BO2\n2+ (m/z 105.5) corresponds to the double ionization of cationic species m/z 211.\nList of the observed double‐ionized cationic fragments\nDouble cation m/z 82.5 is assigned as C6H7B2O4\n2+; m/z 77.5, m/z 53.5 and m/z 52.5 are assigned to the double cationic species C8H5B2O2\n2+, C7H6O2+ and C7H5O2+, respectively. The latter two correspond to the double ionization of C7H6O+ and C7H5O+, m/z 107 and m/z 106 respectively. The only boron‐absent double cation identified appears at m/z 38.5 being attributed to the phenyl double cation C6H5\n2+. The lightest identified double cation is attributed to C5H3O2+ (m/z 37.5).\nFigure 2 shows the positive mass spectrum of TFPB recorded at an incident electron energy of 70 eV.\nTFPB (C21H15B3O6) electron ionization mass spectrum recorded at 70 eV incident electron energy.\nAt 70 eV the processes are well above the ionization threshold of the TFPB molecule; therefore, several fragmentation pathways are accessible at this energy. All fragment peaks were assigned according to the isotopic distribution of the elements, including the natural abundance of 10B/11B. In addition, all cationic peak intensities were normalized according to the C7H5O+ peak signal, which is the most abundant fragment. As far as the authors are aware, no mass spectra obtained by electron ionization have been reported previously.\nComparing the 70 eV spectra of TFPB and triphenylboroxine,\n15\n the precursor cation's relative intensity is significantly lower for TFPB. In the case of triphenylboroxine the most intense cation is the precursor cation followed by Ph2(C6H4)B2O2\n+ (m/z 207) with relative intensity of 0.16. In the present case of TFPB, the most intense fragment is C7H5O+ (m/z 105), and precursor cation is formed with a relative intensity of 0.06. In the triphenylboroxine electron ionization mass spectrum, the precursor cation is the most intense fragment. On the other hand, in the TFPB spectrum the precursor cation presents a relative peak intensity of \n6%. This indicates that the presence of the formyl group attached to the benzene ring (benzaldehyde) induces molecular instability and, thus, a wide range of fragmentation patterns. The list of 29 cationic fragments with relative intensities >2% is presented in Table 1.\nList of cationic fragments with relative intensities >2% under electron ionization at 70 eV electron energy\nIt is also important to remark that the two most abundant cations, C7H5O+ (m/z 105) and C7H6O+ (m/z 106), do not include boron in their chemical structure. This suggests that the boroxine ring is significantly preserved upon electron ionization. We do not discard the possibility of electronic rearrangements, although C7H5O+ can be formed by simple cleavage of C&bond;B bond and C7H6O+ by the cleavage of C‐B bond followed by proton transfer. In addition, eight double cationic fragments are formed (Table 2). The double cations have been assigned regarding 10B isotopic contribution. Precursor double cation is attributed at m/z 197.5; double cation C13H12BO2\n2+ (m/z 105.5) corresponds to the double ionization of cationic species m/z 211.\nList of the observed double‐ionized cationic fragments\nDouble cation m/z 82.5 is assigned as C6H7B2O4\n2+; m/z 77.5, m/z 53.5 and m/z 52.5 are assigned to the double cationic species C8H5B2O2\n2+, C7H6O2+ and C7H5O2+, respectively. The latter two correspond to the double ionization of C7H6O+ and C7H5O+, m/z 107 and m/z 106 respectively. The only boron‐absent double cation identified appears at m/z 38.5 being attributed to the phenyl double cation C6H5\n2+. The lightest identified double cation is attributed to C5H3O2+ (m/z 37.5).\n[SUBTITLE] Minor fragments [SUBSECTION] Apart from the four cations described in Table 1, there were identified 18 more fragments with relative intensities \n<2%. The fragment cations \nC18H15B3O3+ (m/z 312), \nC13H14B2O3+ (m/z 240), \nC14H13B2O2+ (m/z 235), \nC15H2B2O+ (m/z 220), \nC12H9B2O2+ (m/z 207), \nC12H9O2+ (m/z 185), \nC9H5B2O3+ (m/z 183), \nC11H6BO2+ (m/z 181), \nC6H9B2O4+ (m/z 167), \nC6H7B2O4+ (m/z 165), \nC10H6BO+ (m/z 152), \nC6H4BO3+ (m/z 135), \nC2HB2O4+ (m/z 111), \nCBO3+ (m/z 71), \nCHO2+ (m/z 45), \nC2HO+ (m/z 41) and \nCH3O+ or \nH4BO+ (m/z 31) are reported in this electron ionization study.\nApart from the four cations described in Table 1, there were identified 18 more fragments with relative intensities \n<2%. The fragment cations \nC18H15B3O3+ (m/z 312), \nC13H14B2O3+ (m/z 240), \nC14H13B2O2+ (m/z 235), \nC15H2B2O+ (m/z 220), \nC12H9B2O2+ (m/z 207), \nC12H9O2+ (m/z 185), \nC9H5B2O3+ (m/z 183), \nC11H6BO2+ (m/z 181), \nC6H9B2O4+ (m/z 167), \nC6H7B2O4+ (m/z 165), \nC10H6BO+ (m/z 152), \nC6H4BO3+ (m/z 135), \nC2HB2O4+ (m/z 111), \nCBO3+ (m/z 71), \nCHO2+ (m/z 45), \nC2HO+ (m/z 41) and \nCH3O+ or \nH4BO+ (m/z 31) are reported in this electron ionization study.\n[SUBTITLE] Ionization and appearance energies [SUBSECTION] The ionization efficiency curves near the threshold were measured for six abundant cations in the mass spectrum at 70 eV. The experimental data of cations m/z 51, m/z 77, m/z 91, m/z 105, m/z 210 and m/z 396 were fitted by the method based on the Wannier law\n14\n to determine the experimental ionization and appearance energies. The experimental ionization energy of TFPB was determined at 9.46 ± 0.11 eV. Figure 3 depicts the experimental data and the respective Wannier‐type fits for the six most intense cations.\nExperimental thresholds for the six most abundant cations. The blue dots represent the experimental data and the green dashed line represents the Wannier‐type fitting function. The values in parentheses correspond to the experimental threshold along with the respective statistical uncertainties. (A) \nCHB2O+ (m/z 51); (B) \nC6H5+ (m/z 77); (C) \nC4BO+ or \nC5H4BO+ (m/z 91); (D) \nC7H5O+ (m/z 105); (E) precursor cation \nC13H11BO2+ (m/z 210); (F) precursor cation \nC21H15B3O6+ (m/z 396) [Color figure can be viewed at wileyonlinelibrary.com]\nThe appearance energy of \nC7H5O+ (m/z 105), which is assigned to C&bond;B cleavage (bond break between the benzaldehyde and the boroxine ring), was found at 10.33 ± 0.15 eV. The appearance energy of phenyl cation (m/z 77) that is related to the cleavage of the formyl group and the C&bond;B bond was observed at 10.7 ± 0.1 eV. The remaining appearance energies of the other cations \nCHB2O+ (m/z 51), \nC13H11BO2+ (m/z 210) and \nC4BO+ or \nC5H4BO+ (m/z 91) were determined at 17.72 ± 0.32, 9.59 ± 0.11 and 10.76 ± 0.61 eV, respectively.\nTheoretical values for the first ionization potentials (IPs) were determined using the outer‐valence Green's function (OVGF) method, together with the 6‐311G(df,p) basis set. The calculations were performed with the gaussian09 package.\n26\n In this approach, the IPs correspond to the absolute values of the OVGF energies of the molecular orbitals of the neutral molecule. The first IP was obtained at 9.49 eV, which is in very good agreement with the appearance energy of the precursor cation (m/z 396). Besides, we conducted auxiliary investigation of the dissociation thresholds associated with some of the most abundant singly ionized fragments, from which we drew a conclusion that is worth mentioning. Due to the complexity of the molecule, we only focused on reactions involving single bond breaks and/or the simplest possible rearrangements. The thresholds were calculated with the G4(MP2) methodology, as implemented in gaussian09, at \nT=313 K. For the cationic fragment m/z 210, our estimate for the reaction\n\nTFPB+e−→C13H11BO2++C8H4B2O4\nis \nΔEth=10.77 eV, which is more than 1 eV higher than the observed appearance energy for that channel. A similar discrepancy was observed for the cationic fragment m/z 105. The calculated threshold for the reaction\n\nTFPB+e−→C7H5O++C14H10B3O5\n\n\ncorresponding to a single C&bond;B bond break was \nΔEth=11.89 eV, about 1.56 eV above the value observed experimentally. These results indicate that parallel reactions may take place. Along the dissociation paths, secondary rearrangements with more than two products should occur, therefore lowering the reaction threshold. As suggested, a parallel reaction leading to formation of boroxine \nH3B3O3 is exothermic by 1217.54 kJ/mol.\n27\n The energy release in a possible lateral reaction will contribute to the thermodynamics lowering. This fact may explain the difference between experimental and theoretical values.\nThe ionization efficiency curves near the threshold were measured for six abundant cations in the mass spectrum at 70 eV. The experimental data of cations m/z 51, m/z 77, m/z 91, m/z 105, m/z 210 and m/z 396 were fitted by the method based on the Wannier law\n14\n to determine the experimental ionization and appearance energies. The experimental ionization energy of TFPB was determined at 9.46 ± 0.11 eV. Figure 3 depicts the experimental data and the respective Wannier‐type fits for the six most intense cations.\nExperimental thresholds for the six most abundant cations. The blue dots represent the experimental data and the green dashed line represents the Wannier‐type fitting function. The values in parentheses correspond to the experimental threshold along with the respective statistical uncertainties. (A) \nCHB2O+ (m/z 51); (B) \nC6H5+ (m/z 77); (C) \nC4BO+ or \nC5H4BO+ (m/z 91); (D) \nC7H5O+ (m/z 105); (E) precursor cation \nC13H11BO2+ (m/z 210); (F) precursor cation \nC21H15B3O6+ (m/z 396) [Color figure can be viewed at wileyonlinelibrary.com]\nThe appearance energy of \nC7H5O+ (m/z 105), which is assigned to C&bond;B cleavage (bond break between the benzaldehyde and the boroxine ring), was found at 10.33 ± 0.15 eV. The appearance energy of phenyl cation (m/z 77) that is related to the cleavage of the formyl group and the C&bond;B bond was observed at 10.7 ± 0.1 eV. The remaining appearance energies of the other cations \nCHB2O+ (m/z 51), \nC13H11BO2+ (m/z 210) and \nC4BO+ or \nC5H4BO+ (m/z 91) were determined at 17.72 ± 0.32, 9.59 ± 0.11 and 10.76 ± 0.61 eV, respectively.\nTheoretical values for the first ionization potentials (IPs) were determined using the outer‐valence Green's function (OVGF) method, together with the 6‐311G(df,p) basis set. The calculations were performed with the gaussian09 package.\n26\n In this approach, the IPs correspond to the absolute values of the OVGF energies of the molecular orbitals of the neutral molecule. The first IP was obtained at 9.49 eV, which is in very good agreement with the appearance energy of the precursor cation (m/z 396). Besides, we conducted auxiliary investigation of the dissociation thresholds associated with some of the most abundant singly ionized fragments, from which we drew a conclusion that is worth mentioning. Due to the complexity of the molecule, we only focused on reactions involving single bond breaks and/or the simplest possible rearrangements. The thresholds were calculated with the G4(MP2) methodology, as implemented in gaussian09, at \nT=313 K. For the cationic fragment m/z 210, our estimate for the reaction\n\nTFPB+e−→C13H11BO2++C8H4B2O4\nis \nΔEth=10.77 eV, which is more than 1 eV higher than the observed appearance energy for that channel. A similar discrepancy was observed for the cationic fragment m/z 105. The calculated threshold for the reaction\n\nTFPB+e−→C7H5O++C14H10B3O5\n\n\ncorresponding to a single C&bond;B bond break was \nΔEth=11.89 eV, about 1.56 eV above the value observed experimentally. These results indicate that parallel reactions may take place. Along the dissociation paths, secondary rearrangements with more than two products should occur, therefore lowering the reaction threshold. As suggested, a parallel reaction leading to formation of boroxine \nH3B3O3 is exothermic by 1217.54 kJ/mol.\n27\n The energy release in a possible lateral reaction will contribute to the thermodynamics lowering. This fact may explain the difference between experimental and theoretical values.", "Figure 2 shows the positive mass spectrum of TFPB recorded at an incident electron energy of 70 eV.\nTFPB (C21H15B3O6) electron ionization mass spectrum recorded at 70 eV incident electron energy.\nAt 70 eV the processes are well above the ionization threshold of the TFPB molecule; therefore, several fragmentation pathways are accessible at this energy. All fragment peaks were assigned according to the isotopic distribution of the elements, including the natural abundance of 10B/11B. In addition, all cationic peak intensities were normalized according to the C7H5O+ peak signal, which is the most abundant fragment. As far as the authors are aware, no mass spectra obtained by electron ionization have been reported previously.\nComparing the 70 eV spectra of TFPB and triphenylboroxine,\n15\n the precursor cation's relative intensity is significantly lower for TFPB. In the case of triphenylboroxine the most intense cation is the precursor cation followed by Ph2(C6H4)B2O2\n+ (m/z 207) with relative intensity of 0.16. In the present case of TFPB, the most intense fragment is C7H5O+ (m/z 105), and precursor cation is formed with a relative intensity of 0.06. In the triphenylboroxine electron ionization mass spectrum, the precursor cation is the most intense fragment. On the other hand, in the TFPB spectrum the precursor cation presents a relative peak intensity of \n6%. This indicates that the presence of the formyl group attached to the benzene ring (benzaldehyde) induces molecular instability and, thus, a wide range of fragmentation patterns. The list of 29 cationic fragments with relative intensities >2% is presented in Table 1.\nList of cationic fragments with relative intensities >2% under electron ionization at 70 eV electron energy\nIt is also important to remark that the two most abundant cations, C7H5O+ (m/z 105) and C7H6O+ (m/z 106), do not include boron in their chemical structure. This suggests that the boroxine ring is significantly preserved upon electron ionization. We do not discard the possibility of electronic rearrangements, although C7H5O+ can be formed by simple cleavage of C&bond;B bond and C7H6O+ by the cleavage of C‐B bond followed by proton transfer. In addition, eight double cationic fragments are formed (Table 2). The double cations have been assigned regarding 10B isotopic contribution. Precursor double cation is attributed at m/z 197.5; double cation C13H12BO2\n2+ (m/z 105.5) corresponds to the double ionization of cationic species m/z 211.\nList of the observed double‐ionized cationic fragments\nDouble cation m/z 82.5 is assigned as C6H7B2O4\n2+; m/z 77.5, m/z 53.5 and m/z 52.5 are assigned to the double cationic species C8H5B2O2\n2+, C7H6O2+ and C7H5O2+, respectively. The latter two correspond to the double ionization of C7H6O+ and C7H5O+, m/z 107 and m/z 106 respectively. The only boron‐absent double cation identified appears at m/z 38.5 being attributed to the phenyl double cation C6H5\n2+. The lightest identified double cation is attributed to C5H3O2+ (m/z 37.5).", "Apart from the four cations described in Table 1, there were identified 18 more fragments with relative intensities \n<2%. The fragment cations \nC18H15B3O3+ (m/z 312), \nC13H14B2O3+ (m/z 240), \nC14H13B2O2+ (m/z 235), \nC15H2B2O+ (m/z 220), \nC12H9B2O2+ (m/z 207), \nC12H9O2+ (m/z 185), \nC9H5B2O3+ (m/z 183), \nC11H6BO2+ (m/z 181), \nC6H9B2O4+ (m/z 167), \nC6H7B2O4+ (m/z 165), \nC10H6BO+ (m/z 152), \nC6H4BO3+ (m/z 135), \nC2HB2O4+ (m/z 111), \nCBO3+ (m/z 71), \nCHO2+ (m/z 45), \nC2HO+ (m/z 41) and \nCH3O+ or \nH4BO+ (m/z 31) are reported in this electron ionization study.", "The ionization efficiency curves near the threshold were measured for six abundant cations in the mass spectrum at 70 eV. The experimental data of cations m/z 51, m/z 77, m/z 91, m/z 105, m/z 210 and m/z 396 were fitted by the method based on the Wannier law\n14\n to determine the experimental ionization and appearance energies. The experimental ionization energy of TFPB was determined at 9.46 ± 0.11 eV. Figure 3 depicts the experimental data and the respective Wannier‐type fits for the six most intense cations.\nExperimental thresholds for the six most abundant cations. The blue dots represent the experimental data and the green dashed line represents the Wannier‐type fitting function. The values in parentheses correspond to the experimental threshold along with the respective statistical uncertainties. (A) \nCHB2O+ (m/z 51); (B) \nC6H5+ (m/z 77); (C) \nC4BO+ or \nC5H4BO+ (m/z 91); (D) \nC7H5O+ (m/z 105); (E) precursor cation \nC13H11BO2+ (m/z 210); (F) precursor cation \nC21H15B3O6+ (m/z 396) [Color figure can be viewed at wileyonlinelibrary.com]\nThe appearance energy of \nC7H5O+ (m/z 105), which is assigned to C&bond;B cleavage (bond break between the benzaldehyde and the boroxine ring), was found at 10.33 ± 0.15 eV. The appearance energy of phenyl cation (m/z 77) that is related to the cleavage of the formyl group and the C&bond;B bond was observed at 10.7 ± 0.1 eV. The remaining appearance energies of the other cations \nCHB2O+ (m/z 51), \nC13H11BO2+ (m/z 210) and \nC4BO+ or \nC5H4BO+ (m/z 91) were determined at 17.72 ± 0.32, 9.59 ± 0.11 and 10.76 ± 0.61 eV, respectively.\nTheoretical values for the first ionization potentials (IPs) were determined using the outer‐valence Green's function (OVGF) method, together with the 6‐311G(df,p) basis set. The calculations were performed with the gaussian09 package.\n26\n In this approach, the IPs correspond to the absolute values of the OVGF energies of the molecular orbitals of the neutral molecule. The first IP was obtained at 9.49 eV, which is in very good agreement with the appearance energy of the precursor cation (m/z 396). Besides, we conducted auxiliary investigation of the dissociation thresholds associated with some of the most abundant singly ionized fragments, from which we drew a conclusion that is worth mentioning. Due to the complexity of the molecule, we only focused on reactions involving single bond breaks and/or the simplest possible rearrangements. The thresholds were calculated with the G4(MP2) methodology, as implemented in gaussian09, at \nT=313 K. For the cationic fragment m/z 210, our estimate for the reaction\n\nTFPB+e−→C13H11BO2++C8H4B2O4\nis \nΔEth=10.77 eV, which is more than 1 eV higher than the observed appearance energy for that channel. A similar discrepancy was observed for the cationic fragment m/z 105. The calculated threshold for the reaction\n\nTFPB+e−→C7H5O++C14H10B3O5\n\n\ncorresponding to a single C&bond;B bond break was \nΔEth=11.89 eV, about 1.56 eV above the value observed experimentally. These results indicate that parallel reactions may take place. Along the dissociation paths, secondary rearrangements with more than two products should occur, therefore lowering the reaction threshold. As suggested, a parallel reaction leading to formation of boroxine \nH3B3O3 is exothermic by 1217.54 kJ/mol.\n27\n The energy release in a possible lateral reaction will contribute to the thermodynamics lowering. This fact may explain the difference between experimental and theoretical values.", "TFPB shows rich fragmentation patterns upon electron interactions at 70 eV, including several double‐ionization cations. The precursor cation's relative intensity is significantly lower compared to the most abundant fragment \nC7H5O+, which indicates that the presence of benzaldehyde as a substituent on the boron atom induces molecular instability when compared to triphenylboroxine (phenyl groups as substituents). Twenty‐nine cationic fragments with relative intensities \n>2% were detected and identified. The ionization energy of TFPB was found at \n9.46±0.11 eV. In addition, it was observed that three of the most intense cationic fragments do not include boron in their molecular composition, suggesting the boroxine ring's stability. Thus, these results suggest that the TFPB properties may combine the potential radiosensitizer effect of benzaldehyde with the stability of the boroxine ring, which might be important in drug delivery. This comprehensive description of electron‐induced dissociation of TFPB may contribute to the development of novel medical strategies, namely in anticancer therapy, including chemo‐radiotherapy protocols.", "[SUBTITLE] PEER REVIEW [SUBSECTION] The peer review history for this article is available at https://publons.com/publon/10.1002/rcm.9418.\nThe peer review history for this article is available at https://publons.com/publon/10.1002/rcm.9418.", "The peer review history for this article is available at https://publons.com/publon/10.1002/rcm.9418." ]
[ null, null, "materials-and-methods", "discussion", null, null, null, "conclusions", null, null ]
[]
New Approach to Privacy-Preserving Clinical Decision Support Systems for HIV Treatment.
36261621
HIV treatment prescription is a complex process. Clinical decision support systems (CDSS) are a category of health information technologies that can assist clinicians to choose optimal treatments based on clinical trials and expert knowledge. The usability of some CDSSs for HIV treatment would be significantly improved by using the knowledge obtained by treating other patients. This knowledge, however, is mainly contained in patient records, whose usage is restricted due to privacy and confidentiality constraints.
BACKGROUND
A treatment effectiveness measure, containing valuable information for HIV treatment prescription, was defined and a method to extract this measure from patient records was developed. This method uses an advanced cryptographic technology, known as secure Multiparty Computation (henceforth referred to as MPC), to preserve the privacy of the patient records and the confidentiality of the clinicians' decisions.
METHODS
Our solution enables to compute an effectiveness measure of an HIV treatment, the average time-to-treatment-failure, while preserving privacy. Experimental results show that our solution, although at proof-of-concept stage, has good efficiency and provides a result to a query within 24 min for a dataset of realistic size.
FINDINGS
This paper presents a novel and efficient approach HIV clinical decision support systems, that harnesses the potential and insights acquired from treatment data, while preserving the privacy of patient records and the confidentiality of clinician decisions.
INTERPRETATION
[ "Humans", "Privacy", "Computer Security", "Decision Support Systems, Clinical", "Confidentiality", "HIV Infections" ]
9581834
null
null
null
null
Results
The functionality we have achieved utilizes HIV patient records to gain new insights in the effectiveness of HIV treatments. The MPC protocol ensures privacy of the patients and the confidentiality of the clinicians’ treatment decisions. The proposed solution distinguishes between ‘input’ parties, the clinicians supplying the database records, and ‘computing’ parties running the SPDZ protocol, which can be different medical institutions or IT service providers. The input parties additively secret-share their data records and distribute the shares amongst the computing parties (see Fig. 2). Fig. 2Secret sharing database records Secret sharing database records As a result, the two computing parties each hold a share of all the database records. SPDZ allows the evaluation of queries to this secret-shared database in such a way that only the output of the query (the average time-to-treatment-failure ( \documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\overline{TTF}$$\end{document}) per treatment) is revealed to the clinician, and no additional information is leaked to either the querying clinician, or the computing parties (cf. previous section). In order to protect the private information in the query (the viral genotype), we secret-share the query amongst the computing parties in a similar manner. The computing parties thus take as private inputs their shares of the database records and their share of the query. They do not reconstruct the result of the computation (the average TTF) themselves; instead, each of them sends their share of the result to the querying clinician who, in turn, recombines the shares to reconstruct the output. This way the result is only revealed to the clinician, and not to the computing parties (cf. Figure 3). Fig. 3Query architecture of the privacy-preserving CDSS Query architecture of the privacy-preserving CDSS Our solution allows clinicians to compare their treatment of choice against the outcome of treatments previously chosen by other clinicians for patients with similar genotype, without revealing any private information to the clinicians or the computing parties, who only learn the size and format of the database and the number of queries to the database. This system is secure as long as the two computing parties do not collude. [SUBTITLE] Performance – Online Phase [SUBSECTION] In comparison to implementing the functionality without privacy protection, using MPC inherently introduces computational and communication overhead. The main reason for this unavoidable overhead is that, in an MPC protocol, the computation path has to be oblivious, i.e., independent, of the input values, since it would otherwise leak information. Moreover, as explained in the previous section, some basic operations on the input data are translated by MPC into more complex, interactive processes, which lead to unavoidable overhead. We have evaluated the performance of the online phase of our protocol by deploying the computing parties on two different machines, each using one core of a i7-7567U CPU running at 3.50 GHz and 32 GB of RAM, in a local network with 1 Gbit/s throughput. The system ran on a Fedora operating system and has been developed within the SPDZ-2 software suite discussed in the previous section; the overall orchestration of the scalability experiments has been performed via scripts for the Bash shell. Finally, we have instantiated the SPDZ protocol with 40-bit statistical security, 128-bit computational security and a 128-bit prime field. The experiments that we have run measure the time it takes for the solution to return the average time-to-treatment-failure \documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\overline{TTF}_{tr}\left(v\right)$$\end{document} of a given input treatment tr, where the additional input value v is the genotype of a given patient. The formal definition of \documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\overline{TTF}_{tr}\left(v\right)$$\end{document} is presented in the “Materials and Methods” section; as a reminder, it is given by the average over the times-to-treatment-failure of patients with a similar genotype, for the same treatment tr. The results in Fig. 4 show the computation times that are needed for answering one query, for artificially-generated databases with sizes ranging from 100 to 20 000 records. The maximum 20 000 approximates the number of HIV-positive registered individuals in the Netherlands [62]. The experiment is repeated multiple times, resulting in several data points per database size. Recall that per query we compute the average TTF conditioned on ‘similar’ patients for 100 different treatments. The computational complexity scales linearly in the number of database records. Notice that the threshold value B and the number of patient genotypes that have Hamming distance at most B from the given input do not affect the running time of the computation: this is inherent to MPC solutions, which have a computation time which does not depend on the input values. Fig. 4CDSS computation time CDSS computation time Also notice that these figures refer to the time needed to answer a single query; with the current state of our implementation, the running time would scale linearly in the number of queries. This is an aspect to be kept in mind should, for instance, a practitioner want to query the system for different values of the threshold B. In comparison to implementing the functionality without privacy protection, using MPC inherently introduces computational and communication overhead. The main reason for this unavoidable overhead is that, in an MPC protocol, the computation path has to be oblivious, i.e., independent, of the input values, since it would otherwise leak information. Moreover, as explained in the previous section, some basic operations on the input data are translated by MPC into more complex, interactive processes, which lead to unavoidable overhead. We have evaluated the performance of the online phase of our protocol by deploying the computing parties on two different machines, each using one core of a i7-7567U CPU running at 3.50 GHz and 32 GB of RAM, in a local network with 1 Gbit/s throughput. The system ran on a Fedora operating system and has been developed within the SPDZ-2 software suite discussed in the previous section; the overall orchestration of the scalability experiments has been performed via scripts for the Bash shell. Finally, we have instantiated the SPDZ protocol with 40-bit statistical security, 128-bit computational security and a 128-bit prime field. The experiments that we have run measure the time it takes for the solution to return the average time-to-treatment-failure \documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\overline{TTF}_{tr}\left(v\right)$$\end{document} of a given input treatment tr, where the additional input value v is the genotype of a given patient. The formal definition of \documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\overline{TTF}_{tr}\left(v\right)$$\end{document} is presented in the “Materials and Methods” section; as a reminder, it is given by the average over the times-to-treatment-failure of patients with a similar genotype, for the same treatment tr. The results in Fig. 4 show the computation times that are needed for answering one query, for artificially-generated databases with sizes ranging from 100 to 20 000 records. The maximum 20 000 approximates the number of HIV-positive registered individuals in the Netherlands [62]. The experiment is repeated multiple times, resulting in several data points per database size. Recall that per query we compute the average TTF conditioned on ‘similar’ patients for 100 different treatments. The computational complexity scales linearly in the number of database records. Notice that the threshold value B and the number of patient genotypes that have Hamming distance at most B from the given input do not affect the running time of the computation: this is inherent to MPC solutions, which have a computation time which does not depend on the input values. Fig. 4CDSS computation time CDSS computation time Also notice that these figures refer to the time needed to answer a single query; with the current state of our implementation, the running time would scale linearly in the number of queries. This is an aspect to be kept in mind should, for instance, a practitioner want to query the system for different values of the threshold B. [SUBTITLE] Performance – Offline Phase [SUBSECTION] In the SPDZ protocol certain computational tasks are executed in the offline phase, that is independent of the MPC use case and that can be implemented with existing protocols. For this reason, we have merely estimated the computational costs of it. The offline phase can be run at any time to generate a large database of preprocessed data which, in turn, is consumed during the online phase. The performance of the offline phase can be quantified in the number of the so-called multiplication triples that are generated per second. In [63] various approaches for generating multiplication triples in a setting similar to ours were evaluated, generating 30 000 triples/s. To evaluate a single query on a database with 20 000 records approximately 40 million multiplication triples are required. In this setting these triples can thus be generated in approximately 22 min. In the SPDZ protocol certain computational tasks are executed in the offline phase, that is independent of the MPC use case and that can be implemented with existing protocols. For this reason, we have merely estimated the computational costs of it. The offline phase can be run at any time to generate a large database of preprocessed data which, in turn, is consumed during the online phase. The performance of the offline phase can be quantified in the number of the so-called multiplication triples that are generated per second. In [63] various approaches for generating multiplication triples in a setting similar to ours were evaluated, generating 30 000 triples/s. To evaluate a single query on a database with 20 000 records approximately 40 million multiplication triples are required. In this setting these triples can thus be generated in approximately 22 min.
null
null
[ "Background and Significance", "Related Work", "Outline", "Measuring Treatment Effectiveness from Patient Records", "Secure Multiparty Computation", "The MPC framework of our choice: SPDZ", "Performance – Online Phase", "Performance – Offline Phase", "Discussion and Conclusions" ]
[ "The constantly rising cost of national healthcare [1] associated to an aging population has highlighted the need for a critical change in traditional healthcare [2, 3]. Most stakeholders (clinicians, healthcare providers, policy makers and patients) agree that the solution lies in new approaches in which technology and health information technology (HIT) play a critical role [4, 5]. HIT services aim to automate and optimize healthcare processes with the overall goal of providing a more effective treatment process for patients. One of the main barriers to the adoption of HIT lies in the challenges associated with the need to preserve the privacy of the patients’ data; legislation on the privacy of sensitive data, such as the General Data Protection Regulation (EU) 2016/679 (GDPR), is becoming increasingly more stringent, affecting all parties who handle sensitive data.\nIn this paper, we focus on one specific category of HIT systems: Clinical Decision Support Systems (CDSSs). A CDSS is a system that provides clinicians, patients, and other individuals with intelligently processed disease-specific and patient-specific data. Several different categories of CDSSs can be found in literature, such as diagnostic tools, expert systems, and workflow support. Systematic reviews reported that CDSSs significantly improved clinical practice: a review [6] on one hundred studies reported improvements for more than 62% of the trials on practitioner performance, reminder systems, drug-dosing systems and disease management systems. A review on seventy studies [7] reported a significant improvement of clinical practice in 68% of trials. Recent systematic reviews [8, 9] report an improvement in health care processes in 148 randomized, controlled trials and in 85% of twenty-two studies respectively.\nAs a use case to present our proposed solution to the problem of preserving the privacy of patients’ data, we focus on an expert system for HIV treatment. The prescription of antiretroviral drugs to HIV1 infected patients is a complex process in which clinicians have to take into account several factors in a short amount of time. In particular, clinicians need to choose the most appropriate treatment based on the genotype of each patient’s most prevalent strain of the virus in order to minimize drug resistance. A suboptimal treatment will likely result in a more rapid emergence of drug-resistant strains, and, eventually, in increased morbidity and mortality.\nCDSSs are used in order to minimize or, ideally, prevent the prescription of suboptimal HIV1 treatments. Some examples of relevant CDSSs range from simple quality improvement consultation programs like HIVQUAL-US [10] that monitors clinical performance, to more sophisticated data-driven systems like Euresist [11] and knowledge-based systems like the HIVdb Program [12]. The main advantage of the use of these CDSSs is that they save a considerable amount of the clinician’s time, since it would be impossible for the clinicians to analyze in detail the differences between the HIV genotype that is prevalent in a specific patient in search of critical mutations. However, in this paper we focus on the “comparative Drug Ranking System” (cDRS), a CDSS that helps to minimize the choice of sub-optimal HIV treatments by performing a meta-ranking analysis of three expert systems for HIV-1 genotypic drug resistance interpretation (ANRS, HIVdb, Rega) to resolve possible discordances between them [13–16]. The discordances in drug resistance between the three expert systems are not negligible [17, 18], and are the result of the limited amount of clinical data available for each specific set of mutations and of different methodologies used by the systems. A CDSS able to help clinicians in resolving such discordances is essential to avoid the administration of sub-optimal HIV treatments.\nResearch on the spread of the HIV epidemics has led to the development of tools (such as phylogenetic trees) able to correlate specific viral sequences in different patients and reconstruct with good accuracy the network of infections within a community [19]. In addition, transmission events between patients can be identified by analyzing the viral genotypes, given the uniqueness of specific sets of mutations [20, 21]. Hence, strict privacy regulations prevent the sharing of patient data (e.g., viral genotype) that feed and improve these clinical decision support systems. Moreover, clinicians might not be able, or willing, to openly share their treatment decisions and the resulting outcomes, even though such information might be beneficial for the decision-making process of their colleagues. In conclusion, there is a tremendous amount of valuable information that is unavailable to clinicians because of privacy and confidentiality constraints.\nAn ideal system should allow clinicians to compare their chosen treatment against the outcome of the treatments chosen by other clinicians for similar genotypes, while solving the issue of utilizing patient and clinicians’ data in a secure, privacy-preserving way.\nIn this exploratory work, we present a solution that uses cryptographic techniques, namely a so-called secure Multiparty Computation (MPC) protocol, to achieve this functionality without violating any of the privacy constraints. Informally stated, MPC is a collection of cryptographic techniques that allow several parties, each of which holds some private input, to evaluate a function on those inputs without disclosing any extra information on the input themselves, and without resorting to a trusted external party. Our MPC-based solution would allow clinicians to compare past treatments of ‘similar’ patients to find the optimal treatment for new patients preventing any unauthorized party, including the ones performing the computations, to access the input data.\n[SUBTITLE] Related Work [SUBSECTION] Privacy-preserving CDSSs have been presented in recent years [22–25]. However, this line of work focusses on CDSSs for disease-prediction and enables clinicians to securely query remote machine-learning based systems for a given patient’s health condition, in a privacy-preserving way. As such, it is not directly comparable with our solution, which has a different scope within the paradigm of privacy-preserving CDSSs.\nIn more general terms, proposed applications of MPC to the healthcare sector have flourished in recent years. To the best of our knowledge, there exists no article summarizing the scientific literature on MPC applied to the healthcare sector1; we provide here a list of recent and relevant work on the topic, but we stress the fact that this list cannot be exhaustive, due to the high number of publications on the topic.\nA large sub-domain of the application of MPC (and other related cryptographic techniques) to the medical domain aims to deploy machine-learning techniques on medical datasets held by distinct organizations; examples in this sense include privacy-preserving reinforcement learning [26], Kaplan-Meier survival analysis and genome-wide association studies [27], grid logistic regression for biomedical data [28], training of linear [29] and Lasso [30] regression models on medical data, and computing patient risk-stratification metrics [31].\nOther relevant work include medical record searching [32, 33], the study of general methods such as privacy-preserving data mining for joint data analysis between hospitals [34] and branching programs for privacy-preserving classification of medical ElectroCardioGram signals [35], the presentation of specific use case scenarios such as secure disclosure of patient data for disease surveillance [36], R-based healthcare statistics [37], and privacy-preserving genome-wide association study [38], privacy-preserving genome analysis [39] and search of similar patients in genomic data [40].\nFinally, iDASH [41] is an important public initiative to stimulate the development of techniques for privacy-preserving sharing of medical data.\nPrivacy-preserving CDSSs have been presented in recent years [22–25]. However, this line of work focusses on CDSSs for disease-prediction and enables clinicians to securely query remote machine-learning based systems for a given patient’s health condition, in a privacy-preserving way. As such, it is not directly comparable with our solution, which has a different scope within the paradigm of privacy-preserving CDSSs.\nIn more general terms, proposed applications of MPC to the healthcare sector have flourished in recent years. To the best of our knowledge, there exists no article summarizing the scientific literature on MPC applied to the healthcare sector1; we provide here a list of recent and relevant work on the topic, but we stress the fact that this list cannot be exhaustive, due to the high number of publications on the topic.\nA large sub-domain of the application of MPC (and other related cryptographic techniques) to the medical domain aims to deploy machine-learning techniques on medical datasets held by distinct organizations; examples in this sense include privacy-preserving reinforcement learning [26], Kaplan-Meier survival analysis and genome-wide association studies [27], grid logistic regression for biomedical data [28], training of linear [29] and Lasso [30] regression models on medical data, and computing patient risk-stratification metrics [31].\nOther relevant work include medical record searching [32, 33], the study of general methods such as privacy-preserving data mining for joint data analysis between hospitals [34] and branching programs for privacy-preserving classification of medical ElectroCardioGram signals [35], the presentation of specific use case scenarios such as secure disclosure of patient data for disease surveillance [36], R-based healthcare statistics [37], and privacy-preserving genome-wide association study [38], privacy-preserving genome analysis [39] and search of similar patients in genomic data [40].\nFinally, iDASH [41] is an important public initiative to stimulate the development of techniques for privacy-preserving sharing of medical data.\n[SUBTITLE] Outline [SUBSECTION] The rest of the article is organized as follows. In the “Materials and Methods” section we first discuss how to measure the effectiveness of a treatment from patient records and present the method that we propose (setting aside the privacy-preserving aspect); we then provide a brief overview of MPC and of the framework of our choice, SPDZ. In the “Results” section we then explain how the effectiveness measure is securely implemented within SPDZ and present an evaluation of the efficiency of our solution. Finally, the “Discussion and Conclusions” section summarizes the results of the article and provides an appraisal of the achieved results and on possible future work.\nThe rest of the article is organized as follows. In the “Materials and Methods” section we first discuss how to measure the effectiveness of a treatment from patient records and present the method that we propose (setting aside the privacy-preserving aspect); we then provide a brief overview of MPC and of the framework of our choice, SPDZ. In the “Results” section we then explain how the effectiveness measure is securely implemented within SPDZ and present an evaluation of the efficiency of our solution. Finally, the “Discussion and Conclusions” section summarizes the results of the article and provides an appraisal of the achieved results and on possible future work.", "Privacy-preserving CDSSs have been presented in recent years [22–25]. However, this line of work focusses on CDSSs for disease-prediction and enables clinicians to securely query remote machine-learning based systems for a given patient’s health condition, in a privacy-preserving way. As such, it is not directly comparable with our solution, which has a different scope within the paradigm of privacy-preserving CDSSs.\nIn more general terms, proposed applications of MPC to the healthcare sector have flourished in recent years. To the best of our knowledge, there exists no article summarizing the scientific literature on MPC applied to the healthcare sector1; we provide here a list of recent and relevant work on the topic, but we stress the fact that this list cannot be exhaustive, due to the high number of publications on the topic.\nA large sub-domain of the application of MPC (and other related cryptographic techniques) to the medical domain aims to deploy machine-learning techniques on medical datasets held by distinct organizations; examples in this sense include privacy-preserving reinforcement learning [26], Kaplan-Meier survival analysis and genome-wide association studies [27], grid logistic regression for biomedical data [28], training of linear [29] and Lasso [30] regression models on medical data, and computing patient risk-stratification metrics [31].\nOther relevant work include medical record searching [32, 33], the study of general methods such as privacy-preserving data mining for joint data analysis between hospitals [34] and branching programs for privacy-preserving classification of medical ElectroCardioGram signals [35], the presentation of specific use case scenarios such as secure disclosure of patient data for disease surveillance [36], R-based healthcare statistics [37], and privacy-preserving genome-wide association study [38], privacy-preserving genome analysis [39] and search of similar patients in genomic data [40].\nFinally, iDASH [41] is an important public initiative to stimulate the development of techniques for privacy-preserving sharing of medical data.", "The rest of the article is organized as follows. In the “Materials and Methods” section we first discuss how to measure the effectiveness of a treatment from patient records and present the method that we propose (setting aside the privacy-preserving aspect); we then provide a brief overview of MPC and of the framework of our choice, SPDZ. In the “Results” section we then explain how the effectiveness measure is securely implemented within SPDZ and present an evaluation of the efficiency of our solution. Finally, the “Discussion and Conclusions” section summarizes the results of the article and provides an appraisal of the achieved results and on possible future work.", "The viral genotype of a patient refers to the genetic sequence(s) of the HIV-1 virus strain that is most prevalent at the time of the blood test. The HIV-1 virus RNA genome contains 3 key regions that encode for enzymes critical to the life cycle of the virus: protease (P), integrase (I) and reverse transcriptase (RT). Each region encodes for enzymes with 99, 288 and 560 amino acids, respectively, all of which could in principle mutate. These mutations play an important role in the drug resistance of the virus strains.\nGiven an HIV-1 patient, our goal is to obtain treatment results of ‘similar’ patients, and therefore we need to define a metric or distance function that quantifies the similarity between two patients, or two viral genotypes. Since all expert systems indicate resistance to drugs based on substitutions in the amino acid sequence of the wild-type HIV-1, we need a way to compute the distance in the amino acid sequences of the viral proteins. Metrics of distances between amino acid sequences are fairly complex and often assessed via neural networks [42]. The assignment of a suitable similarity metric is outside the scope of this paper, and for this reason, we have chosen to use a simplified viral genotype representation with a generic metric as a proof of concept. However, our solution is flexible, since it can support other representations and metrics.\nFrom now on we shall represent viral genotypes as bit strings v of a fixed length N, i.e., \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$v\\in {\\left\\{\\text{0,1}\\right\\}}^{N}$$\\end{document}. We can think of each bit in this bit-string as an indicator for the presence or the absence of a specific mutation at a specific position.\nSince there are only 97 relevant positions with commonly 1 or 2 resistance-associated substitutions [64] we can expect \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$N$$\\end{document} to be somewhere between 100 and 200. The distance between two viral genotypes \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$v_{1}$$\\end{document} and \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$v_{2}$$\\end{document} is defined by the Hamming distance between the bit strings:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$H\\left({v}_{1},{v}_{2}\\right)=\\left|\\left\\{i : {v}_{1}\\left(i\\right)\\ne {v}_{2}\\left(i\\right)\\right\\}\\right|,$$\\end{document}\nGiven this metric we can define two viral genotypes v1 and v2 to be similar if their Hamming distance is smaller than a certain threshold B, i.e., \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$H\\left({v}_{1},{v}_{2}\\right)<B.$$\\end{document} Even though this metric is a simplification of the metrics used in practice, it is quite similar to the rule-based metrics used in the CDSSs of [13, 16]. These CDSSs match viral genotypes based on the presence of resistance-associated substitutions in amino acid positions, which can be seen as a Boolean expression. In a clinical setting, these CDSSs compare the two complete viral strings to identify specific insertion, deletions, and substitutions but do not rely on a single threshold value defined as a Hamming distance. In fact, it is well known from specific studies which additions, deletions or substitutions trigger a clinically relevant mutation. In a practical implementation, we would have to look at the difference in specific positions of the sequences of two amino acid strings. The threshold that would be used in that case would be defined by clinicians who set of rules used by the specific CDSS instead of the Hamming distance described in the example. However, the rulesets that would trigger an alert are still Boolean in nature and would fit the proposed secure MPC solution.\nSuboptimal treatments of HIV-1 patients result in faster emergence of resistant strains and this emergence renders the treatment ineffective. Hence, a way to measure the effectiveness of a treatment tr for genotype v is by indicating the time-to-treatment-failure\n\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$TT{F}_{tr}\\left(v\\right)$$\\end{document}. The \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$TT{F}_{tr}\\left(v\\right)$$\\end{document} is defined as the time (in days) between the start of a therapy tr and either a therapy switch, a discontinuation of therapy or death [65, 66], for a patient with genotype v. Hence, given an HIV-1 patient with genotype v we would, for example, like to compute the average \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)$$\\end{document} over all patients with similar genotype vi, as an indication for the unknown true effectiveness measure \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$TT{F}_{tr}\\left(v\\right)$$\\end{document}:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)= \\frac{1}{\\left|\\left\\{i : H\\left(v,{v}_{i}\\right)<B\\right\\}\\right|}\\sum _{i : H\\left(v,{v}_{i}\\right)<B}TT{F}_{tr}\\left({v}_{i}\\right) ,$$\\end{document}\nWhere H denotes, as discussed above, the Hamming distance and B denotes a fixed threshold value.", "MPC has been introduced by Yao in the 1980s [43]. Given n mutually distrusting parties \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$${P}_{1},\\dots , {P}_{n}$$\\end{document}, each holding private inputs x1,…, xn, the goal of MPC is to allow the parties to compute the value \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f({x}_{1},\\dots , {x}_{n})$$\\end{document} of a function \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\text{f}$$\\end{document} on their inputs, without revealing any other information than \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f({x}_{1},\\dots , {x}_{n})$$\\end{document}, and without resorting to an external trusted party.\nEarly research in the 1980s [43–46] established the theoretical feasibility bounds for MPC; informally stated, this line of research proved that any function f with finite domain and finite image can be evaluated securely in an MPC fashion. The precise security properties that can be achieved depend on the behavior of players and on the underlying communication model.\nSince the first market-ready deployment of MPC in 2008 [47], MPC solutions have been used in various practical contexts, e.g., stock market order matching [48], job market inquiries [49], and frequency bands auctions [50]. Moreover, various software suites and implementation frameworks for MPC have been made available [51–55].\nSeveral considerations have to be made when applying MPC to a given problem. For instance, one may assume that parties \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$${P}_{1},\\dots , {P}_{n}$$\\end{document} will behave semi-honestly (meaning that they may try to learn information on the other parties’ inputs, but do follow the protocol), or that it is instead necessary to provide security against fully malicious players that deviate from the protocol instructions. Another important parameter that varies among protocols is the number t of corrupted parties that can be tolerated out of the total number n of parties.\nA remark of notable importance is that many desirable properties of MPC may negatively impact performance, or even be mutually exclusive, which means that the choice of an MPC protocol may be subject to important trade-offs. The reader can refer to [56] for a comprehensive discussion of MPC.", "We base our MPC solution on the SPDZ protocol [57, 58]. The protocol is distinguished for its fast performance, and is implemented in a freely accessible software suite called SPDZ-2 [52, 54] for UNIX-based systems2; SPDZ-2 allow developers to write programs in Python-like syntax, and it then compiles the code to executable format.\nSPDZ follows the so-called share-compute-reveal paradigm: each input xi of the function f to be computed is ‘dispersed’ (or, formally speaking, secret-shared3) into n pieces of data, called shares, each of which is assigned to a party; this process has the property that no information on xi can be extracted from a set of shares, unless such a set contains all shares (in which case xi can be completely recovered). Subsequently, parties execute a ‘computation’ protocol; as a result of this step, each party will have a share of the output \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f\\left({x}_{1},\\dots ,{x}_{n}\\right)$$\\end{document} of the function. Once all shares have been gathered, the output can then be reconstructed.\nA schematic representation of this paradigm for the addition of two values x and y among two parties is provided in Fig. 1. The top row represents the shares held by the first party, while the bottom row represents the shares of the second party. The assumption here is that x1 and x2 are two random values subject to the condition that \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$${x}_{1}+{x}_{2}=x$$\\end{document}, and similarly for y; bearing this fact in mind, it is then seen how the process respects the privacy and reconstruction requirements discussed above.\n\nFig. 1Example of share-compute-reveal paradigm for addition of two values\n\nExample of share-compute-reveal paradigm for addition of two values\nIn more general terms, the core idea behind MPC protocols based on the share-compute-reveal paradigm is that the function \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document} to be evaluated on the input values is “decomposed” into basic operations (such as sum and products); these basic operations are then translated into similar operations on the shares and executed in the same order. An important remark is that, in general, these operations on shares require some form of interaction among the parties (for instance, multiplication of two values cannot simply be performed by multiplying the corresponding shares, and requires a more involved and interactive process). The reader can refer to the literature on MPC and on SPDZ that we have provided for a more formal and complete discussion of this topic.\nOther cryptographic techniques such as homomorphic encryption [59, 60] could potentially be of relevance for private data analysis, but we ruled out these alternatives, because they would induce a huge computational overhead in our setting.\nThe share-compute-reveal approach is particularly well-suited for the client-server model we are interested in: the ‘input’ parties, clinicians (clients) simply need to supply their secret-shared inputs to two or more ‘computing’ parties (servers), who will execute the computation protocol on these inputs, and then communicate the shares of the output to the input parties, which can thus reconstruct the output.\nIt is important to remark that the SPDZ protocol does not, per se, distinguish between input and computing parties. A framework for MPC in a client-server model was presented in [61]; moreover, in [63] the SPDZ protocol was adjusted to the client-server setting.\nThe SPDZ protocol is divided into an ‘offline’ phase and an ‘online’ phase. The offline phase can be executed before the function inputs x1,…,xn are known, and its goal is to produce some secret-shared auxiliary data that will be used in the evaluation of \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document}; producing this data can be a computationally-intensive process, but since secret inputs are not required, this step can be executed during idle time and well before the actual secure computation will take place. Once the auxiliary data has been produced, the evaluation of \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document} can be performed very efficiently: this is of particular relevance for our use case, where input parties (clinicians) need to obtain the output of the function \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document} within a matter of minutes, while preprocessing material can be produced in the background by the computing parties.", "In comparison to implementing the functionality without privacy protection, using MPC inherently introduces computational and communication overhead. The main reason for this unavoidable overhead is that, in an MPC protocol, the computation path has to be oblivious, i.e., independent, of the input values, since it would otherwise leak information. Moreover, as explained in the previous section, some basic operations on the input data are translated by MPC into more complex, interactive processes, which lead to unavoidable overhead.\nWe have evaluated the performance of the online phase of our protocol by deploying the computing parties on two different machines, each using one core of a i7-7567U CPU running at 3.50 GHz and 32 GB of RAM, in a local network with 1 Gbit/s throughput. The system ran on a Fedora operating system and has been developed within the SPDZ-2 software suite discussed in the previous section; the overall orchestration of the scalability experiments has been performed via scripts for the Bash shell. Finally, we have instantiated the SPDZ protocol with 40-bit statistical security, 128-bit computational security and a 128-bit prime field.\nThe experiments that we have run measure the time it takes for the solution to return the average time-to-treatment-failure \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)$$\\end{document} of a given input treatment tr, where the additional input value v is the genotype of a given patient. The formal definition of \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)$$\\end{document} is presented in the “Materials and Methods” section; as a reminder, it is given by the average over the times-to-treatment-failure of patients with a similar genotype, for the same treatment tr.\nThe results in Fig. 4 show the computation times that are needed for answering one query, for artificially-generated databases with sizes ranging from 100 to 20 000 records. The maximum 20 000 approximates the number of HIV-positive registered individuals in the Netherlands [62]. The experiment is repeated multiple times, resulting in several data points per database size. Recall that per query we compute the average TTF conditioned on ‘similar’ patients for 100 different treatments. The computational complexity scales linearly in the number of database records. Notice that the threshold value B and the number of patient genotypes that have Hamming distance at most B from the given input do not affect the running time of the computation: this is inherent to MPC solutions, which have a computation time which does not depend on the input values.\n\nFig. 4CDSS computation time\n\nCDSS computation time\nAlso notice that these figures refer to the time needed to answer a single query; with the current state of our implementation, the running time would scale linearly in the number of queries. This is an aspect to be kept in mind should, for instance, a practitioner want to query the system for different values of the threshold B.", "In the SPDZ protocol certain computational tasks are executed in the offline phase, that is independent of the MPC use case and that can be implemented with existing protocols. For this reason, we have merely estimated the computational costs of it. The offline phase can be run at any time to generate a large database of preprocessed data which, in turn, is consumed during the online phase.\nThe performance of the offline phase can be quantified in the number of the so-called multiplication triples that are generated per second. In [63] various approaches for generating multiplication triples in a setting similar to ours were evaluated, generating 30 000 triples/s. To evaluate a single query on a database with 20 000 records approximately 40 million multiplication triples are required. In this setting these triples can thus be generated in approximately 22 min.", "We presented a novel approach for HIV1 clinical decision support systems, making use of advanced cryptographic techniques to process private information without revealing it. By making use of MPC, we can ensure both the privacy of the clinicians’ treatment choices and the privacy of patients.\nTowards a fully operational deployment some points are yet to be addressed. Notably, the SPDZ software framework is designed for research purposes only, which means that our implementation should be audited and checked for vulnerabilities. For what concerns efficiency and scalability, we stress the fact that any CDSS for HIV treatment should produce a suggestion within minutes, since practitioners would typically query the system right after visiting a patient and would expect an answer before the patient leaves their office. As shown in Fig. 3, our solution answers a query within 24 min, for a database size roughly matching the number of HIV-positive registered individuals in the Netherlands [62]; while we consider this result to be sufficient for the proof-of-concept presented in this paper, some further work would be needed for a full-scale deployment. The running time of the implementation could be improved by several means, e.g., by using a low-level but very fast programming language such as C, by further parallelizing the computation, or by making use of high-performance computing machines instead of consumer-level hardware." ]
[ null, null, null, null, null, null, null, null, null ]
[ "Background and Significance", "Related Work", "Outline", "Materials and Methods", "Measuring Treatment Effectiveness from Patient Records", "Secure Multiparty Computation", "The MPC framework of our choice: SPDZ", "Results", "Performance – Online Phase", "Performance – Offline Phase", "Discussion and Conclusions" ]
[ "The constantly rising cost of national healthcare [1] associated to an aging population has highlighted the need for a critical change in traditional healthcare [2, 3]. Most stakeholders (clinicians, healthcare providers, policy makers and patients) agree that the solution lies in new approaches in which technology and health information technology (HIT) play a critical role [4, 5]. HIT services aim to automate and optimize healthcare processes with the overall goal of providing a more effective treatment process for patients. One of the main barriers to the adoption of HIT lies in the challenges associated with the need to preserve the privacy of the patients’ data; legislation on the privacy of sensitive data, such as the General Data Protection Regulation (EU) 2016/679 (GDPR), is becoming increasingly more stringent, affecting all parties who handle sensitive data.\nIn this paper, we focus on one specific category of HIT systems: Clinical Decision Support Systems (CDSSs). A CDSS is a system that provides clinicians, patients, and other individuals with intelligently processed disease-specific and patient-specific data. Several different categories of CDSSs can be found in literature, such as diagnostic tools, expert systems, and workflow support. Systematic reviews reported that CDSSs significantly improved clinical practice: a review [6] on one hundred studies reported improvements for more than 62% of the trials on practitioner performance, reminder systems, drug-dosing systems and disease management systems. A review on seventy studies [7] reported a significant improvement of clinical practice in 68% of trials. Recent systematic reviews [8, 9] report an improvement in health care processes in 148 randomized, controlled trials and in 85% of twenty-two studies respectively.\nAs a use case to present our proposed solution to the problem of preserving the privacy of patients’ data, we focus on an expert system for HIV treatment. The prescription of antiretroviral drugs to HIV1 infected patients is a complex process in which clinicians have to take into account several factors in a short amount of time. In particular, clinicians need to choose the most appropriate treatment based on the genotype of each patient’s most prevalent strain of the virus in order to minimize drug resistance. A suboptimal treatment will likely result in a more rapid emergence of drug-resistant strains, and, eventually, in increased morbidity and mortality.\nCDSSs are used in order to minimize or, ideally, prevent the prescription of suboptimal HIV1 treatments. Some examples of relevant CDSSs range from simple quality improvement consultation programs like HIVQUAL-US [10] that monitors clinical performance, to more sophisticated data-driven systems like Euresist [11] and knowledge-based systems like the HIVdb Program [12]. The main advantage of the use of these CDSSs is that they save a considerable amount of the clinician’s time, since it would be impossible for the clinicians to analyze in detail the differences between the HIV genotype that is prevalent in a specific patient in search of critical mutations. However, in this paper we focus on the “comparative Drug Ranking System” (cDRS), a CDSS that helps to minimize the choice of sub-optimal HIV treatments by performing a meta-ranking analysis of three expert systems for HIV-1 genotypic drug resistance interpretation (ANRS, HIVdb, Rega) to resolve possible discordances between them [13–16]. The discordances in drug resistance between the three expert systems are not negligible [17, 18], and are the result of the limited amount of clinical data available for each specific set of mutations and of different methodologies used by the systems. A CDSS able to help clinicians in resolving such discordances is essential to avoid the administration of sub-optimal HIV treatments.\nResearch on the spread of the HIV epidemics has led to the development of tools (such as phylogenetic trees) able to correlate specific viral sequences in different patients and reconstruct with good accuracy the network of infections within a community [19]. In addition, transmission events between patients can be identified by analyzing the viral genotypes, given the uniqueness of specific sets of mutations [20, 21]. Hence, strict privacy regulations prevent the sharing of patient data (e.g., viral genotype) that feed and improve these clinical decision support systems. Moreover, clinicians might not be able, or willing, to openly share their treatment decisions and the resulting outcomes, even though such information might be beneficial for the decision-making process of their colleagues. In conclusion, there is a tremendous amount of valuable information that is unavailable to clinicians because of privacy and confidentiality constraints.\nAn ideal system should allow clinicians to compare their chosen treatment against the outcome of the treatments chosen by other clinicians for similar genotypes, while solving the issue of utilizing patient and clinicians’ data in a secure, privacy-preserving way.\nIn this exploratory work, we present a solution that uses cryptographic techniques, namely a so-called secure Multiparty Computation (MPC) protocol, to achieve this functionality without violating any of the privacy constraints. Informally stated, MPC is a collection of cryptographic techniques that allow several parties, each of which holds some private input, to evaluate a function on those inputs without disclosing any extra information on the input themselves, and without resorting to a trusted external party. Our MPC-based solution would allow clinicians to compare past treatments of ‘similar’ patients to find the optimal treatment for new patients preventing any unauthorized party, including the ones performing the computations, to access the input data.\n[SUBTITLE] Related Work [SUBSECTION] Privacy-preserving CDSSs have been presented in recent years [22–25]. However, this line of work focusses on CDSSs for disease-prediction and enables clinicians to securely query remote machine-learning based systems for a given patient’s health condition, in a privacy-preserving way. As such, it is not directly comparable with our solution, which has a different scope within the paradigm of privacy-preserving CDSSs.\nIn more general terms, proposed applications of MPC to the healthcare sector have flourished in recent years. To the best of our knowledge, there exists no article summarizing the scientific literature on MPC applied to the healthcare sector1; we provide here a list of recent and relevant work on the topic, but we stress the fact that this list cannot be exhaustive, due to the high number of publications on the topic.\nA large sub-domain of the application of MPC (and other related cryptographic techniques) to the medical domain aims to deploy machine-learning techniques on medical datasets held by distinct organizations; examples in this sense include privacy-preserving reinforcement learning [26], Kaplan-Meier survival analysis and genome-wide association studies [27], grid logistic regression for biomedical data [28], training of linear [29] and Lasso [30] regression models on medical data, and computing patient risk-stratification metrics [31].\nOther relevant work include medical record searching [32, 33], the study of general methods such as privacy-preserving data mining for joint data analysis between hospitals [34] and branching programs for privacy-preserving classification of medical ElectroCardioGram signals [35], the presentation of specific use case scenarios such as secure disclosure of patient data for disease surveillance [36], R-based healthcare statistics [37], and privacy-preserving genome-wide association study [38], privacy-preserving genome analysis [39] and search of similar patients in genomic data [40].\nFinally, iDASH [41] is an important public initiative to stimulate the development of techniques for privacy-preserving sharing of medical data.\nPrivacy-preserving CDSSs have been presented in recent years [22–25]. However, this line of work focusses on CDSSs for disease-prediction and enables clinicians to securely query remote machine-learning based systems for a given patient’s health condition, in a privacy-preserving way. As such, it is not directly comparable with our solution, which has a different scope within the paradigm of privacy-preserving CDSSs.\nIn more general terms, proposed applications of MPC to the healthcare sector have flourished in recent years. To the best of our knowledge, there exists no article summarizing the scientific literature on MPC applied to the healthcare sector1; we provide here a list of recent and relevant work on the topic, but we stress the fact that this list cannot be exhaustive, due to the high number of publications on the topic.\nA large sub-domain of the application of MPC (and other related cryptographic techniques) to the medical domain aims to deploy machine-learning techniques on medical datasets held by distinct organizations; examples in this sense include privacy-preserving reinforcement learning [26], Kaplan-Meier survival analysis and genome-wide association studies [27], grid logistic regression for biomedical data [28], training of linear [29] and Lasso [30] regression models on medical data, and computing patient risk-stratification metrics [31].\nOther relevant work include medical record searching [32, 33], the study of general methods such as privacy-preserving data mining for joint data analysis between hospitals [34] and branching programs for privacy-preserving classification of medical ElectroCardioGram signals [35], the presentation of specific use case scenarios such as secure disclosure of patient data for disease surveillance [36], R-based healthcare statistics [37], and privacy-preserving genome-wide association study [38], privacy-preserving genome analysis [39] and search of similar patients in genomic data [40].\nFinally, iDASH [41] is an important public initiative to stimulate the development of techniques for privacy-preserving sharing of medical data.\n[SUBTITLE] Outline [SUBSECTION] The rest of the article is organized as follows. In the “Materials and Methods” section we first discuss how to measure the effectiveness of a treatment from patient records and present the method that we propose (setting aside the privacy-preserving aspect); we then provide a brief overview of MPC and of the framework of our choice, SPDZ. In the “Results” section we then explain how the effectiveness measure is securely implemented within SPDZ and present an evaluation of the efficiency of our solution. Finally, the “Discussion and Conclusions” section summarizes the results of the article and provides an appraisal of the achieved results and on possible future work.\nThe rest of the article is organized as follows. In the “Materials and Methods” section we first discuss how to measure the effectiveness of a treatment from patient records and present the method that we propose (setting aside the privacy-preserving aspect); we then provide a brief overview of MPC and of the framework of our choice, SPDZ. In the “Results” section we then explain how the effectiveness measure is securely implemented within SPDZ and present an evaluation of the efficiency of our solution. Finally, the “Discussion and Conclusions” section summarizes the results of the article and provides an appraisal of the achieved results and on possible future work.", "Privacy-preserving CDSSs have been presented in recent years [22–25]. However, this line of work focusses on CDSSs for disease-prediction and enables clinicians to securely query remote machine-learning based systems for a given patient’s health condition, in a privacy-preserving way. As such, it is not directly comparable with our solution, which has a different scope within the paradigm of privacy-preserving CDSSs.\nIn more general terms, proposed applications of MPC to the healthcare sector have flourished in recent years. To the best of our knowledge, there exists no article summarizing the scientific literature on MPC applied to the healthcare sector1; we provide here a list of recent and relevant work on the topic, but we stress the fact that this list cannot be exhaustive, due to the high number of publications on the topic.\nA large sub-domain of the application of MPC (and other related cryptographic techniques) to the medical domain aims to deploy machine-learning techniques on medical datasets held by distinct organizations; examples in this sense include privacy-preserving reinforcement learning [26], Kaplan-Meier survival analysis and genome-wide association studies [27], grid logistic regression for biomedical data [28], training of linear [29] and Lasso [30] regression models on medical data, and computing patient risk-stratification metrics [31].\nOther relevant work include medical record searching [32, 33], the study of general methods such as privacy-preserving data mining for joint data analysis between hospitals [34] and branching programs for privacy-preserving classification of medical ElectroCardioGram signals [35], the presentation of specific use case scenarios such as secure disclosure of patient data for disease surveillance [36], R-based healthcare statistics [37], and privacy-preserving genome-wide association study [38], privacy-preserving genome analysis [39] and search of similar patients in genomic data [40].\nFinally, iDASH [41] is an important public initiative to stimulate the development of techniques for privacy-preserving sharing of medical data.", "The rest of the article is organized as follows. In the “Materials and Methods” section we first discuss how to measure the effectiveness of a treatment from patient records and present the method that we propose (setting aside the privacy-preserving aspect); we then provide a brief overview of MPC and of the framework of our choice, SPDZ. In the “Results” section we then explain how the effectiveness measure is securely implemented within SPDZ and present an evaluation of the efficiency of our solution. Finally, the “Discussion and Conclusions” section summarizes the results of the article and provides an appraisal of the achieved results and on possible future work.", "[SUBTITLE] Measuring Treatment Effectiveness from Patient Records [SUBSECTION] The viral genotype of a patient refers to the genetic sequence(s) of the HIV-1 virus strain that is most prevalent at the time of the blood test. The HIV-1 virus RNA genome contains 3 key regions that encode for enzymes critical to the life cycle of the virus: protease (P), integrase (I) and reverse transcriptase (RT). Each region encodes for enzymes with 99, 288 and 560 amino acids, respectively, all of which could in principle mutate. These mutations play an important role in the drug resistance of the virus strains.\nGiven an HIV-1 patient, our goal is to obtain treatment results of ‘similar’ patients, and therefore we need to define a metric or distance function that quantifies the similarity between two patients, or two viral genotypes. Since all expert systems indicate resistance to drugs based on substitutions in the amino acid sequence of the wild-type HIV-1, we need a way to compute the distance in the amino acid sequences of the viral proteins. Metrics of distances between amino acid sequences are fairly complex and often assessed via neural networks [42]. The assignment of a suitable similarity metric is outside the scope of this paper, and for this reason, we have chosen to use a simplified viral genotype representation with a generic metric as a proof of concept. However, our solution is flexible, since it can support other representations and metrics.\nFrom now on we shall represent viral genotypes as bit strings v of a fixed length N, i.e., \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$v\\in {\\left\\{\\text{0,1}\\right\\}}^{N}$$\\end{document}. We can think of each bit in this bit-string as an indicator for the presence or the absence of a specific mutation at a specific position.\nSince there are only 97 relevant positions with commonly 1 or 2 resistance-associated substitutions [64] we can expect \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$N$$\\end{document} to be somewhere between 100 and 200. The distance between two viral genotypes \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$v_{1}$$\\end{document} and \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$v_{2}$$\\end{document} is defined by the Hamming distance between the bit strings:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$H\\left({v}_{1},{v}_{2}\\right)=\\left|\\left\\{i : {v}_{1}\\left(i\\right)\\ne {v}_{2}\\left(i\\right)\\right\\}\\right|,$$\\end{document}\nGiven this metric we can define two viral genotypes v1 and v2 to be similar if their Hamming distance is smaller than a certain threshold B, i.e., \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$H\\left({v}_{1},{v}_{2}\\right)<B.$$\\end{document} Even though this metric is a simplification of the metrics used in practice, it is quite similar to the rule-based metrics used in the CDSSs of [13, 16]. These CDSSs match viral genotypes based on the presence of resistance-associated substitutions in amino acid positions, which can be seen as a Boolean expression. In a clinical setting, these CDSSs compare the two complete viral strings to identify specific insertion, deletions, and substitutions but do not rely on a single threshold value defined as a Hamming distance. In fact, it is well known from specific studies which additions, deletions or substitutions trigger a clinically relevant mutation. In a practical implementation, we would have to look at the difference in specific positions of the sequences of two amino acid strings. The threshold that would be used in that case would be defined by clinicians who set of rules used by the specific CDSS instead of the Hamming distance described in the example. However, the rulesets that would trigger an alert are still Boolean in nature and would fit the proposed secure MPC solution.\nSuboptimal treatments of HIV-1 patients result in faster emergence of resistant strains and this emergence renders the treatment ineffective. Hence, a way to measure the effectiveness of a treatment tr for genotype v is by indicating the time-to-treatment-failure\n\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$TT{F}_{tr}\\left(v\\right)$$\\end{document}. The \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$TT{F}_{tr}\\left(v\\right)$$\\end{document} is defined as the time (in days) between the start of a therapy tr and either a therapy switch, a discontinuation of therapy or death [65, 66], for a patient with genotype v. Hence, given an HIV-1 patient with genotype v we would, for example, like to compute the average \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)$$\\end{document} over all patients with similar genotype vi, as an indication for the unknown true effectiveness measure \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$TT{F}_{tr}\\left(v\\right)$$\\end{document}:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)= \\frac{1}{\\left|\\left\\{i : H\\left(v,{v}_{i}\\right)<B\\right\\}\\right|}\\sum _{i : H\\left(v,{v}_{i}\\right)<B}TT{F}_{tr}\\left({v}_{i}\\right) ,$$\\end{document}\nWhere H denotes, as discussed above, the Hamming distance and B denotes a fixed threshold value.\nThe viral genotype of a patient refers to the genetic sequence(s) of the HIV-1 virus strain that is most prevalent at the time of the blood test. The HIV-1 virus RNA genome contains 3 key regions that encode for enzymes critical to the life cycle of the virus: protease (P), integrase (I) and reverse transcriptase (RT). Each region encodes for enzymes with 99, 288 and 560 amino acids, respectively, all of which could in principle mutate. These mutations play an important role in the drug resistance of the virus strains.\nGiven an HIV-1 patient, our goal is to obtain treatment results of ‘similar’ patients, and therefore we need to define a metric or distance function that quantifies the similarity between two patients, or two viral genotypes. Since all expert systems indicate resistance to drugs based on substitutions in the amino acid sequence of the wild-type HIV-1, we need a way to compute the distance in the amino acid sequences of the viral proteins. Metrics of distances between amino acid sequences are fairly complex and often assessed via neural networks [42]. The assignment of a suitable similarity metric is outside the scope of this paper, and for this reason, we have chosen to use a simplified viral genotype representation with a generic metric as a proof of concept. However, our solution is flexible, since it can support other representations and metrics.\nFrom now on we shall represent viral genotypes as bit strings v of a fixed length N, i.e., \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$v\\in {\\left\\{\\text{0,1}\\right\\}}^{N}$$\\end{document}. We can think of each bit in this bit-string as an indicator for the presence or the absence of a specific mutation at a specific position.\nSince there are only 97 relevant positions with commonly 1 or 2 resistance-associated substitutions [64] we can expect \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$N$$\\end{document} to be somewhere between 100 and 200. The distance between two viral genotypes \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$v_{1}$$\\end{document} and \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$v_{2}$$\\end{document} is defined by the Hamming distance between the bit strings:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$H\\left({v}_{1},{v}_{2}\\right)=\\left|\\left\\{i : {v}_{1}\\left(i\\right)\\ne {v}_{2}\\left(i\\right)\\right\\}\\right|,$$\\end{document}\nGiven this metric we can define two viral genotypes v1 and v2 to be similar if their Hamming distance is smaller than a certain threshold B, i.e., \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$H\\left({v}_{1},{v}_{2}\\right)<B.$$\\end{document} Even though this metric is a simplification of the metrics used in practice, it is quite similar to the rule-based metrics used in the CDSSs of [13, 16]. These CDSSs match viral genotypes based on the presence of resistance-associated substitutions in amino acid positions, which can be seen as a Boolean expression. In a clinical setting, these CDSSs compare the two complete viral strings to identify specific insertion, deletions, and substitutions but do not rely on a single threshold value defined as a Hamming distance. In fact, it is well known from specific studies which additions, deletions or substitutions trigger a clinically relevant mutation. In a practical implementation, we would have to look at the difference in specific positions of the sequences of two amino acid strings. The threshold that would be used in that case would be defined by clinicians who set of rules used by the specific CDSS instead of the Hamming distance described in the example. However, the rulesets that would trigger an alert are still Boolean in nature and would fit the proposed secure MPC solution.\nSuboptimal treatments of HIV-1 patients result in faster emergence of resistant strains and this emergence renders the treatment ineffective. Hence, a way to measure the effectiveness of a treatment tr for genotype v is by indicating the time-to-treatment-failure\n\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$TT{F}_{tr}\\left(v\\right)$$\\end{document}. The \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$TT{F}_{tr}\\left(v\\right)$$\\end{document} is defined as the time (in days) between the start of a therapy tr and either a therapy switch, a discontinuation of therapy or death [65, 66], for a patient with genotype v. Hence, given an HIV-1 patient with genotype v we would, for example, like to compute the average \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)$$\\end{document} over all patients with similar genotype vi, as an indication for the unknown true effectiveness measure \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$TT{F}_{tr}\\left(v\\right)$$\\end{document}:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)= \\frac{1}{\\left|\\left\\{i : H\\left(v,{v}_{i}\\right)<B\\right\\}\\right|}\\sum _{i : H\\left(v,{v}_{i}\\right)<B}TT{F}_{tr}\\left({v}_{i}\\right) ,$$\\end{document}\nWhere H denotes, as discussed above, the Hamming distance and B denotes a fixed threshold value.\n[SUBTITLE] Secure Multiparty Computation [SUBSECTION] MPC has been introduced by Yao in the 1980s [43]. Given n mutually distrusting parties \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$${P}_{1},\\dots , {P}_{n}$$\\end{document}, each holding private inputs x1,…, xn, the goal of MPC is to allow the parties to compute the value \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f({x}_{1},\\dots , {x}_{n})$$\\end{document} of a function \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\text{f}$$\\end{document} on their inputs, without revealing any other information than \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f({x}_{1},\\dots , {x}_{n})$$\\end{document}, and without resorting to an external trusted party.\nEarly research in the 1980s [43–46] established the theoretical feasibility bounds for MPC; informally stated, this line of research proved that any function f with finite domain and finite image can be evaluated securely in an MPC fashion. The precise security properties that can be achieved depend on the behavior of players and on the underlying communication model.\nSince the first market-ready deployment of MPC in 2008 [47], MPC solutions have been used in various practical contexts, e.g., stock market order matching [48], job market inquiries [49], and frequency bands auctions [50]. Moreover, various software suites and implementation frameworks for MPC have been made available [51–55].\nSeveral considerations have to be made when applying MPC to a given problem. For instance, one may assume that parties \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$${P}_{1},\\dots , {P}_{n}$$\\end{document} will behave semi-honestly (meaning that they may try to learn information on the other parties’ inputs, but do follow the protocol), or that it is instead necessary to provide security against fully malicious players that deviate from the protocol instructions. Another important parameter that varies among protocols is the number t of corrupted parties that can be tolerated out of the total number n of parties.\nA remark of notable importance is that many desirable properties of MPC may negatively impact performance, or even be mutually exclusive, which means that the choice of an MPC protocol may be subject to important trade-offs. The reader can refer to [56] for a comprehensive discussion of MPC.\nMPC has been introduced by Yao in the 1980s [43]. Given n mutually distrusting parties \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$${P}_{1},\\dots , {P}_{n}$$\\end{document}, each holding private inputs x1,…, xn, the goal of MPC is to allow the parties to compute the value \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f({x}_{1},\\dots , {x}_{n})$$\\end{document} of a function \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\text{f}$$\\end{document} on their inputs, without revealing any other information than \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f({x}_{1},\\dots , {x}_{n})$$\\end{document}, and without resorting to an external trusted party.\nEarly research in the 1980s [43–46] established the theoretical feasibility bounds for MPC; informally stated, this line of research proved that any function f with finite domain and finite image can be evaluated securely in an MPC fashion. The precise security properties that can be achieved depend on the behavior of players and on the underlying communication model.\nSince the first market-ready deployment of MPC in 2008 [47], MPC solutions have been used in various practical contexts, e.g., stock market order matching [48], job market inquiries [49], and frequency bands auctions [50]. Moreover, various software suites and implementation frameworks for MPC have been made available [51–55].\nSeveral considerations have to be made when applying MPC to a given problem. For instance, one may assume that parties \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$${P}_{1},\\dots , {P}_{n}$$\\end{document} will behave semi-honestly (meaning that they may try to learn information on the other parties’ inputs, but do follow the protocol), or that it is instead necessary to provide security against fully malicious players that deviate from the protocol instructions. Another important parameter that varies among protocols is the number t of corrupted parties that can be tolerated out of the total number n of parties.\nA remark of notable importance is that many desirable properties of MPC may negatively impact performance, or even be mutually exclusive, which means that the choice of an MPC protocol may be subject to important trade-offs. The reader can refer to [56] for a comprehensive discussion of MPC.\n[SUBTITLE] The MPC framework of our choice: SPDZ [SUBSECTION] We base our MPC solution on the SPDZ protocol [57, 58]. The protocol is distinguished for its fast performance, and is implemented in a freely accessible software suite called SPDZ-2 [52, 54] for UNIX-based systems2; SPDZ-2 allow developers to write programs in Python-like syntax, and it then compiles the code to executable format.\nSPDZ follows the so-called share-compute-reveal paradigm: each input xi of the function f to be computed is ‘dispersed’ (or, formally speaking, secret-shared3) into n pieces of data, called shares, each of which is assigned to a party; this process has the property that no information on xi can be extracted from a set of shares, unless such a set contains all shares (in which case xi can be completely recovered). Subsequently, parties execute a ‘computation’ protocol; as a result of this step, each party will have a share of the output \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f\\left({x}_{1},\\dots ,{x}_{n}\\right)$$\\end{document} of the function. Once all shares have been gathered, the output can then be reconstructed.\nA schematic representation of this paradigm for the addition of two values x and y among two parties is provided in Fig. 1. The top row represents the shares held by the first party, while the bottom row represents the shares of the second party. The assumption here is that x1 and x2 are two random values subject to the condition that \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$${x}_{1}+{x}_{2}=x$$\\end{document}, and similarly for y; bearing this fact in mind, it is then seen how the process respects the privacy and reconstruction requirements discussed above.\n\nFig. 1Example of share-compute-reveal paradigm for addition of two values\n\nExample of share-compute-reveal paradigm for addition of two values\nIn more general terms, the core idea behind MPC protocols based on the share-compute-reveal paradigm is that the function \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document} to be evaluated on the input values is “decomposed” into basic operations (such as sum and products); these basic operations are then translated into similar operations on the shares and executed in the same order. An important remark is that, in general, these operations on shares require some form of interaction among the parties (for instance, multiplication of two values cannot simply be performed by multiplying the corresponding shares, and requires a more involved and interactive process). The reader can refer to the literature on MPC and on SPDZ that we have provided for a more formal and complete discussion of this topic.\nOther cryptographic techniques such as homomorphic encryption [59, 60] could potentially be of relevance for private data analysis, but we ruled out these alternatives, because they would induce a huge computational overhead in our setting.\nThe share-compute-reveal approach is particularly well-suited for the client-server model we are interested in: the ‘input’ parties, clinicians (clients) simply need to supply their secret-shared inputs to two or more ‘computing’ parties (servers), who will execute the computation protocol on these inputs, and then communicate the shares of the output to the input parties, which can thus reconstruct the output.\nIt is important to remark that the SPDZ protocol does not, per se, distinguish between input and computing parties. A framework for MPC in a client-server model was presented in [61]; moreover, in [63] the SPDZ protocol was adjusted to the client-server setting.\nThe SPDZ protocol is divided into an ‘offline’ phase and an ‘online’ phase. The offline phase can be executed before the function inputs x1,…,xn are known, and its goal is to produce some secret-shared auxiliary data that will be used in the evaluation of \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document}; producing this data can be a computationally-intensive process, but since secret inputs are not required, this step can be executed during idle time and well before the actual secure computation will take place. Once the auxiliary data has been produced, the evaluation of \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document} can be performed very efficiently: this is of particular relevance for our use case, where input parties (clinicians) need to obtain the output of the function \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document} within a matter of minutes, while preprocessing material can be produced in the background by the computing parties.\nWe base our MPC solution on the SPDZ protocol [57, 58]. The protocol is distinguished for its fast performance, and is implemented in a freely accessible software suite called SPDZ-2 [52, 54] for UNIX-based systems2; SPDZ-2 allow developers to write programs in Python-like syntax, and it then compiles the code to executable format.\nSPDZ follows the so-called share-compute-reveal paradigm: each input xi of the function f to be computed is ‘dispersed’ (or, formally speaking, secret-shared3) into n pieces of data, called shares, each of which is assigned to a party; this process has the property that no information on xi can be extracted from a set of shares, unless such a set contains all shares (in which case xi can be completely recovered). Subsequently, parties execute a ‘computation’ protocol; as a result of this step, each party will have a share of the output \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f\\left({x}_{1},\\dots ,{x}_{n}\\right)$$\\end{document} of the function. Once all shares have been gathered, the output can then be reconstructed.\nA schematic representation of this paradigm for the addition of two values x and y among two parties is provided in Fig. 1. The top row represents the shares held by the first party, while the bottom row represents the shares of the second party. The assumption here is that x1 and x2 are two random values subject to the condition that \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$${x}_{1}+{x}_{2}=x$$\\end{document}, and similarly for y; bearing this fact in mind, it is then seen how the process respects the privacy and reconstruction requirements discussed above.\n\nFig. 1Example of share-compute-reveal paradigm for addition of two values\n\nExample of share-compute-reveal paradigm for addition of two values\nIn more general terms, the core idea behind MPC protocols based on the share-compute-reveal paradigm is that the function \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document} to be evaluated on the input values is “decomposed” into basic operations (such as sum and products); these basic operations are then translated into similar operations on the shares and executed in the same order. An important remark is that, in general, these operations on shares require some form of interaction among the parties (for instance, multiplication of two values cannot simply be performed by multiplying the corresponding shares, and requires a more involved and interactive process). The reader can refer to the literature on MPC and on SPDZ that we have provided for a more formal and complete discussion of this topic.\nOther cryptographic techniques such as homomorphic encryption [59, 60] could potentially be of relevance for private data analysis, but we ruled out these alternatives, because they would induce a huge computational overhead in our setting.\nThe share-compute-reveal approach is particularly well-suited for the client-server model we are interested in: the ‘input’ parties, clinicians (clients) simply need to supply their secret-shared inputs to two or more ‘computing’ parties (servers), who will execute the computation protocol on these inputs, and then communicate the shares of the output to the input parties, which can thus reconstruct the output.\nIt is important to remark that the SPDZ protocol does not, per se, distinguish between input and computing parties. A framework for MPC in a client-server model was presented in [61]; moreover, in [63] the SPDZ protocol was adjusted to the client-server setting.\nThe SPDZ protocol is divided into an ‘offline’ phase and an ‘online’ phase. The offline phase can be executed before the function inputs x1,…,xn are known, and its goal is to produce some secret-shared auxiliary data that will be used in the evaluation of \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document}; producing this data can be a computationally-intensive process, but since secret inputs are not required, this step can be executed during idle time and well before the actual secure computation will take place. Once the auxiliary data has been produced, the evaluation of \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document} can be performed very efficiently: this is of particular relevance for our use case, where input parties (clinicians) need to obtain the output of the function \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document} within a matter of minutes, while preprocessing material can be produced in the background by the computing parties.", "The viral genotype of a patient refers to the genetic sequence(s) of the HIV-1 virus strain that is most prevalent at the time of the blood test. The HIV-1 virus RNA genome contains 3 key regions that encode for enzymes critical to the life cycle of the virus: protease (P), integrase (I) and reverse transcriptase (RT). Each region encodes for enzymes with 99, 288 and 560 amino acids, respectively, all of which could in principle mutate. These mutations play an important role in the drug resistance of the virus strains.\nGiven an HIV-1 patient, our goal is to obtain treatment results of ‘similar’ patients, and therefore we need to define a metric or distance function that quantifies the similarity between two patients, or two viral genotypes. Since all expert systems indicate resistance to drugs based on substitutions in the amino acid sequence of the wild-type HIV-1, we need a way to compute the distance in the amino acid sequences of the viral proteins. Metrics of distances between amino acid sequences are fairly complex and often assessed via neural networks [42]. The assignment of a suitable similarity metric is outside the scope of this paper, and for this reason, we have chosen to use a simplified viral genotype representation with a generic metric as a proof of concept. However, our solution is flexible, since it can support other representations and metrics.\nFrom now on we shall represent viral genotypes as bit strings v of a fixed length N, i.e., \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$v\\in {\\left\\{\\text{0,1}\\right\\}}^{N}$$\\end{document}. We can think of each bit in this bit-string as an indicator for the presence or the absence of a specific mutation at a specific position.\nSince there are only 97 relevant positions with commonly 1 or 2 resistance-associated substitutions [64] we can expect \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$N$$\\end{document} to be somewhere between 100 and 200. The distance between two viral genotypes \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$v_{1}$$\\end{document} and \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$v_{2}$$\\end{document} is defined by the Hamming distance between the bit strings:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$H\\left({v}_{1},{v}_{2}\\right)=\\left|\\left\\{i : {v}_{1}\\left(i\\right)\\ne {v}_{2}\\left(i\\right)\\right\\}\\right|,$$\\end{document}\nGiven this metric we can define two viral genotypes v1 and v2 to be similar if their Hamming distance is smaller than a certain threshold B, i.e., \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$H\\left({v}_{1},{v}_{2}\\right)<B.$$\\end{document} Even though this metric is a simplification of the metrics used in practice, it is quite similar to the rule-based metrics used in the CDSSs of [13, 16]. These CDSSs match viral genotypes based on the presence of resistance-associated substitutions in amino acid positions, which can be seen as a Boolean expression. In a clinical setting, these CDSSs compare the two complete viral strings to identify specific insertion, deletions, and substitutions but do not rely on a single threshold value defined as a Hamming distance. In fact, it is well known from specific studies which additions, deletions or substitutions trigger a clinically relevant mutation. In a practical implementation, we would have to look at the difference in specific positions of the sequences of two amino acid strings. The threshold that would be used in that case would be defined by clinicians who set of rules used by the specific CDSS instead of the Hamming distance described in the example. However, the rulesets that would trigger an alert are still Boolean in nature and would fit the proposed secure MPC solution.\nSuboptimal treatments of HIV-1 patients result in faster emergence of resistant strains and this emergence renders the treatment ineffective. Hence, a way to measure the effectiveness of a treatment tr for genotype v is by indicating the time-to-treatment-failure\n\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$TT{F}_{tr}\\left(v\\right)$$\\end{document}. The \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$TT{F}_{tr}\\left(v\\right)$$\\end{document} is defined as the time (in days) between the start of a therapy tr and either a therapy switch, a discontinuation of therapy or death [65, 66], for a patient with genotype v. Hence, given an HIV-1 patient with genotype v we would, for example, like to compute the average \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)$$\\end{document} over all patients with similar genotype vi, as an indication for the unknown true effectiveness measure \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$TT{F}_{tr}\\left(v\\right)$$\\end{document}:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)= \\frac{1}{\\left|\\left\\{i : H\\left(v,{v}_{i}\\right)<B\\right\\}\\right|}\\sum _{i : H\\left(v,{v}_{i}\\right)<B}TT{F}_{tr}\\left({v}_{i}\\right) ,$$\\end{document}\nWhere H denotes, as discussed above, the Hamming distance and B denotes a fixed threshold value.", "MPC has been introduced by Yao in the 1980s [43]. Given n mutually distrusting parties \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$${P}_{1},\\dots , {P}_{n}$$\\end{document}, each holding private inputs x1,…, xn, the goal of MPC is to allow the parties to compute the value \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f({x}_{1},\\dots , {x}_{n})$$\\end{document} of a function \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\text{f}$$\\end{document} on their inputs, without revealing any other information than \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f({x}_{1},\\dots , {x}_{n})$$\\end{document}, and without resorting to an external trusted party.\nEarly research in the 1980s [43–46] established the theoretical feasibility bounds for MPC; informally stated, this line of research proved that any function f with finite domain and finite image can be evaluated securely in an MPC fashion. The precise security properties that can be achieved depend on the behavior of players and on the underlying communication model.\nSince the first market-ready deployment of MPC in 2008 [47], MPC solutions have been used in various practical contexts, e.g., stock market order matching [48], job market inquiries [49], and frequency bands auctions [50]. Moreover, various software suites and implementation frameworks for MPC have been made available [51–55].\nSeveral considerations have to be made when applying MPC to a given problem. For instance, one may assume that parties \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$${P}_{1},\\dots , {P}_{n}$$\\end{document} will behave semi-honestly (meaning that they may try to learn information on the other parties’ inputs, but do follow the protocol), or that it is instead necessary to provide security against fully malicious players that deviate from the protocol instructions. Another important parameter that varies among protocols is the number t of corrupted parties that can be tolerated out of the total number n of parties.\nA remark of notable importance is that many desirable properties of MPC may negatively impact performance, or even be mutually exclusive, which means that the choice of an MPC protocol may be subject to important trade-offs. The reader can refer to [56] for a comprehensive discussion of MPC.", "We base our MPC solution on the SPDZ protocol [57, 58]. The protocol is distinguished for its fast performance, and is implemented in a freely accessible software suite called SPDZ-2 [52, 54] for UNIX-based systems2; SPDZ-2 allow developers to write programs in Python-like syntax, and it then compiles the code to executable format.\nSPDZ follows the so-called share-compute-reveal paradigm: each input xi of the function f to be computed is ‘dispersed’ (or, formally speaking, secret-shared3) into n pieces of data, called shares, each of which is assigned to a party; this process has the property that no information on xi can be extracted from a set of shares, unless such a set contains all shares (in which case xi can be completely recovered). Subsequently, parties execute a ‘computation’ protocol; as a result of this step, each party will have a share of the output \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f\\left({x}_{1},\\dots ,{x}_{n}\\right)$$\\end{document} of the function. Once all shares have been gathered, the output can then be reconstructed.\nA schematic representation of this paradigm for the addition of two values x and y among two parties is provided in Fig. 1. The top row represents the shares held by the first party, while the bottom row represents the shares of the second party. The assumption here is that x1 and x2 are two random values subject to the condition that \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$${x}_{1}+{x}_{2}=x$$\\end{document}, and similarly for y; bearing this fact in mind, it is then seen how the process respects the privacy and reconstruction requirements discussed above.\n\nFig. 1Example of share-compute-reveal paradigm for addition of two values\n\nExample of share-compute-reveal paradigm for addition of two values\nIn more general terms, the core idea behind MPC protocols based on the share-compute-reveal paradigm is that the function \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document} to be evaluated on the input values is “decomposed” into basic operations (such as sum and products); these basic operations are then translated into similar operations on the shares and executed in the same order. An important remark is that, in general, these operations on shares require some form of interaction among the parties (for instance, multiplication of two values cannot simply be performed by multiplying the corresponding shares, and requires a more involved and interactive process). The reader can refer to the literature on MPC and on SPDZ that we have provided for a more formal and complete discussion of this topic.\nOther cryptographic techniques such as homomorphic encryption [59, 60] could potentially be of relevance for private data analysis, but we ruled out these alternatives, because they would induce a huge computational overhead in our setting.\nThe share-compute-reveal approach is particularly well-suited for the client-server model we are interested in: the ‘input’ parties, clinicians (clients) simply need to supply their secret-shared inputs to two or more ‘computing’ parties (servers), who will execute the computation protocol on these inputs, and then communicate the shares of the output to the input parties, which can thus reconstruct the output.\nIt is important to remark that the SPDZ protocol does not, per se, distinguish between input and computing parties. A framework for MPC in a client-server model was presented in [61]; moreover, in [63] the SPDZ protocol was adjusted to the client-server setting.\nThe SPDZ protocol is divided into an ‘offline’ phase and an ‘online’ phase. The offline phase can be executed before the function inputs x1,…,xn are known, and its goal is to produce some secret-shared auxiliary data that will be used in the evaluation of \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document}; producing this data can be a computationally-intensive process, but since secret inputs are not required, this step can be executed during idle time and well before the actual secure computation will take place. Once the auxiliary data has been produced, the evaluation of \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document} can be performed very efficiently: this is of particular relevance for our use case, where input parties (clinicians) need to obtain the output of the function \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$f$$\\end{document} within a matter of minutes, while preprocessing material can be produced in the background by the computing parties.", "The functionality we have achieved utilizes HIV patient records to gain new insights in the effectiveness of HIV treatments. The MPC protocol ensures privacy of the patients and the confidentiality of the clinicians’ treatment decisions.\nThe proposed solution distinguishes between ‘input’ parties, the clinicians supplying the database records, and ‘computing’ parties running the SPDZ protocol, which can be different medical institutions or IT service providers. The input parties additively secret-share their data records and distribute the shares amongst the computing parties (see Fig. 2).\n\nFig. 2Secret sharing database records\n\nSecret sharing database records\nAs a result, the two computing parties each hold a share of all the database records. SPDZ allows the evaluation of queries to this secret-shared database in such a way that only the output of the query (the average time-to-treatment-failure ( \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}$$\\end{document}) per treatment) is revealed to the clinician, and no additional information is leaked to either the querying clinician, or the computing parties (cf. previous section). In order to protect the private information in the query (the viral genotype), we secret-share the query amongst the computing parties in a similar manner. The computing parties thus take as private inputs their shares of the database records and their share of the query. They do not reconstruct the result of the computation (the average TTF) themselves; instead, each of them sends their share of the result to the querying clinician who, in turn, recombines the shares to reconstruct the output. This way the result is only revealed to the clinician, and not to the computing parties (cf. Figure 3).\n\nFig. 3Query architecture of the privacy-preserving CDSS\n\nQuery architecture of the privacy-preserving CDSS\nOur solution allows clinicians to compare their treatment of choice against the outcome of treatments previously chosen by other clinicians for patients with similar genotype, without revealing any private information to the clinicians or the computing parties, who only learn the size and format of the database and the number of queries to the database. This system is secure as long as the two computing parties do not collude.\n[SUBTITLE] Performance – Online Phase [SUBSECTION] In comparison to implementing the functionality without privacy protection, using MPC inherently introduces computational and communication overhead. The main reason for this unavoidable overhead is that, in an MPC protocol, the computation path has to be oblivious, i.e., independent, of the input values, since it would otherwise leak information. Moreover, as explained in the previous section, some basic operations on the input data are translated by MPC into more complex, interactive processes, which lead to unavoidable overhead.\nWe have evaluated the performance of the online phase of our protocol by deploying the computing parties on two different machines, each using one core of a i7-7567U CPU running at 3.50 GHz and 32 GB of RAM, in a local network with 1 Gbit/s throughput. The system ran on a Fedora operating system and has been developed within the SPDZ-2 software suite discussed in the previous section; the overall orchestration of the scalability experiments has been performed via scripts for the Bash shell. Finally, we have instantiated the SPDZ protocol with 40-bit statistical security, 128-bit computational security and a 128-bit prime field.\nThe experiments that we have run measure the time it takes for the solution to return the average time-to-treatment-failure \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)$$\\end{document} of a given input treatment tr, where the additional input value v is the genotype of a given patient. The formal definition of \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)$$\\end{document} is presented in the “Materials and Methods” section; as a reminder, it is given by the average over the times-to-treatment-failure of patients with a similar genotype, for the same treatment tr.\nThe results in Fig. 4 show the computation times that are needed for answering one query, for artificially-generated databases with sizes ranging from 100 to 20 000 records. The maximum 20 000 approximates the number of HIV-positive registered individuals in the Netherlands [62]. The experiment is repeated multiple times, resulting in several data points per database size. Recall that per query we compute the average TTF conditioned on ‘similar’ patients for 100 different treatments. The computational complexity scales linearly in the number of database records. Notice that the threshold value B and the number of patient genotypes that have Hamming distance at most B from the given input do not affect the running time of the computation: this is inherent to MPC solutions, which have a computation time which does not depend on the input values.\n\nFig. 4CDSS computation time\n\nCDSS computation time\nAlso notice that these figures refer to the time needed to answer a single query; with the current state of our implementation, the running time would scale linearly in the number of queries. This is an aspect to be kept in mind should, for instance, a practitioner want to query the system for different values of the threshold B.\nIn comparison to implementing the functionality without privacy protection, using MPC inherently introduces computational and communication overhead. The main reason for this unavoidable overhead is that, in an MPC protocol, the computation path has to be oblivious, i.e., independent, of the input values, since it would otherwise leak information. Moreover, as explained in the previous section, some basic operations on the input data are translated by MPC into more complex, interactive processes, which lead to unavoidable overhead.\nWe have evaluated the performance of the online phase of our protocol by deploying the computing parties on two different machines, each using one core of a i7-7567U CPU running at 3.50 GHz and 32 GB of RAM, in a local network with 1 Gbit/s throughput. The system ran on a Fedora operating system and has been developed within the SPDZ-2 software suite discussed in the previous section; the overall orchestration of the scalability experiments has been performed via scripts for the Bash shell. Finally, we have instantiated the SPDZ protocol with 40-bit statistical security, 128-bit computational security and a 128-bit prime field.\nThe experiments that we have run measure the time it takes for the solution to return the average time-to-treatment-failure \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)$$\\end{document} of a given input treatment tr, where the additional input value v is the genotype of a given patient. The formal definition of \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)$$\\end{document} is presented in the “Materials and Methods” section; as a reminder, it is given by the average over the times-to-treatment-failure of patients with a similar genotype, for the same treatment tr.\nThe results in Fig. 4 show the computation times that are needed for answering one query, for artificially-generated databases with sizes ranging from 100 to 20 000 records. The maximum 20 000 approximates the number of HIV-positive registered individuals in the Netherlands [62]. The experiment is repeated multiple times, resulting in several data points per database size. Recall that per query we compute the average TTF conditioned on ‘similar’ patients for 100 different treatments. The computational complexity scales linearly in the number of database records. Notice that the threshold value B and the number of patient genotypes that have Hamming distance at most B from the given input do not affect the running time of the computation: this is inherent to MPC solutions, which have a computation time which does not depend on the input values.\n\nFig. 4CDSS computation time\n\nCDSS computation time\nAlso notice that these figures refer to the time needed to answer a single query; with the current state of our implementation, the running time would scale linearly in the number of queries. This is an aspect to be kept in mind should, for instance, a practitioner want to query the system for different values of the threshold B.\n[SUBTITLE] Performance – Offline Phase [SUBSECTION] In the SPDZ protocol certain computational tasks are executed in the offline phase, that is independent of the MPC use case and that can be implemented with existing protocols. For this reason, we have merely estimated the computational costs of it. The offline phase can be run at any time to generate a large database of preprocessed data which, in turn, is consumed during the online phase.\nThe performance of the offline phase can be quantified in the number of the so-called multiplication triples that are generated per second. In [63] various approaches for generating multiplication triples in a setting similar to ours were evaluated, generating 30 000 triples/s. To evaluate a single query on a database with 20 000 records approximately 40 million multiplication triples are required. In this setting these triples can thus be generated in approximately 22 min.\nIn the SPDZ protocol certain computational tasks are executed in the offline phase, that is independent of the MPC use case and that can be implemented with existing protocols. For this reason, we have merely estimated the computational costs of it. The offline phase can be run at any time to generate a large database of preprocessed data which, in turn, is consumed during the online phase.\nThe performance of the offline phase can be quantified in the number of the so-called multiplication triples that are generated per second. In [63] various approaches for generating multiplication triples in a setting similar to ours were evaluated, generating 30 000 triples/s. To evaluate a single query on a database with 20 000 records approximately 40 million multiplication triples are required. In this setting these triples can thus be generated in approximately 22 min.", "In comparison to implementing the functionality without privacy protection, using MPC inherently introduces computational and communication overhead. The main reason for this unavoidable overhead is that, in an MPC protocol, the computation path has to be oblivious, i.e., independent, of the input values, since it would otherwise leak information. Moreover, as explained in the previous section, some basic operations on the input data are translated by MPC into more complex, interactive processes, which lead to unavoidable overhead.\nWe have evaluated the performance of the online phase of our protocol by deploying the computing parties on two different machines, each using one core of a i7-7567U CPU running at 3.50 GHz and 32 GB of RAM, in a local network with 1 Gbit/s throughput. The system ran on a Fedora operating system and has been developed within the SPDZ-2 software suite discussed in the previous section; the overall orchestration of the scalability experiments has been performed via scripts for the Bash shell. Finally, we have instantiated the SPDZ protocol with 40-bit statistical security, 128-bit computational security and a 128-bit prime field.\nThe experiments that we have run measure the time it takes for the solution to return the average time-to-treatment-failure \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)$$\\end{document} of a given input treatment tr, where the additional input value v is the genotype of a given patient. The formal definition of \\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\overline{TTF}_{tr}\\left(v\\right)$$\\end{document} is presented in the “Materials and Methods” section; as a reminder, it is given by the average over the times-to-treatment-failure of patients with a similar genotype, for the same treatment tr.\nThe results in Fig. 4 show the computation times that are needed for answering one query, for artificially-generated databases with sizes ranging from 100 to 20 000 records. The maximum 20 000 approximates the number of HIV-positive registered individuals in the Netherlands [62]. The experiment is repeated multiple times, resulting in several data points per database size. Recall that per query we compute the average TTF conditioned on ‘similar’ patients for 100 different treatments. The computational complexity scales linearly in the number of database records. Notice that the threshold value B and the number of patient genotypes that have Hamming distance at most B from the given input do not affect the running time of the computation: this is inherent to MPC solutions, which have a computation time which does not depend on the input values.\n\nFig. 4CDSS computation time\n\nCDSS computation time\nAlso notice that these figures refer to the time needed to answer a single query; with the current state of our implementation, the running time would scale linearly in the number of queries. This is an aspect to be kept in mind should, for instance, a practitioner want to query the system for different values of the threshold B.", "In the SPDZ protocol certain computational tasks are executed in the offline phase, that is independent of the MPC use case and that can be implemented with existing protocols. For this reason, we have merely estimated the computational costs of it. The offline phase can be run at any time to generate a large database of preprocessed data which, in turn, is consumed during the online phase.\nThe performance of the offline phase can be quantified in the number of the so-called multiplication triples that are generated per second. In [63] various approaches for generating multiplication triples in a setting similar to ours were evaluated, generating 30 000 triples/s. To evaluate a single query on a database with 20 000 records approximately 40 million multiplication triples are required. In this setting these triples can thus be generated in approximately 22 min.", "We presented a novel approach for HIV1 clinical decision support systems, making use of advanced cryptographic techniques to process private information without revealing it. By making use of MPC, we can ensure both the privacy of the clinicians’ treatment choices and the privacy of patients.\nTowards a fully operational deployment some points are yet to be addressed. Notably, the SPDZ software framework is designed for research purposes only, which means that our implementation should be audited and checked for vulnerabilities. For what concerns efficiency and scalability, we stress the fact that any CDSS for HIV treatment should produce a suggestion within minutes, since practitioners would typically query the system right after visiting a patient and would expect an answer before the patient leaves their office. As shown in Fig. 3, our solution answers a query within 24 min, for a database size roughly matching the number of HIV-positive registered individuals in the Netherlands [62]; while we consider this result to be sufficient for the proof-of-concept presented in this paper, some further work would be needed for a full-scale deployment. The running time of the implementation could be improved by several means, e.g., by using a low-level but very fast programming language such as C, by further parallelizing the computation, or by making use of high-performance computing machines instead of consumer-level hardware." ]
[ null, null, null, "materials|methods", null, null, null, "results", null, null, null ]
[ "Clinical decision support systems", "Anti-HIV agents", "Secure multiparty computation", "Privacy", "Confidentiality" ]
Evaluating continuum of maternal and newborn healthcare in Rwanda: evidence from the 2019-2020 Rwanda demographic health survey.
36261801
Access to a complete continuum of maternal and child health care has been recommended globally for better pregnancy outcomes. Hence this study determined the level (pooled prevalence) and predictors of successfully completing continuum of care (CoC) in Rwanda.
BACKGROUND
We analyzed weighted secondary data from the 2019-2020 Rwanda Demographic and Health Survey (RDHS) that included 6,302 women aged 15 to 49 years who were selected using multistage stratified sampling. We analyzed complete continuum of care as a composite variable of three maternal care services: at least four ANC contacts, SBA, maternal and neonatal post-natal care. We used the SPSS version 25 complex samples package to conduct multivariable logistic regression.
METHODS
Of the 6,302 women, 2,131 (33.8%) (95% CI: 32.8-35.1) had complete continuum of care. The odds of having complete continuum of care were higher among women who had exposure to newspapers (adjusted odds ratio (AOR): 1.30, 95% CI: 1.11-1.52), those belonging to the eastern region (AOR): 1.24, 95% CI: 1.01-1.52), southern region (AOR): 1.26, 95% CI: 1.04-1.53), those with health insurance (AOR): 1.55, 95% CI: 1.30-1.85), those who had been visited by a field health worker (AOR: 1.31, 95% CI: 1.15-1.49), those with no big problems with distance to health facility (AOR): 1.25, 95% CI: 1.07-1.46), those who were married (AOR): 1.35, 95% CI: 1.11-1.64), those with tertiary level of education (AOR): 1.61, 95% CI: 1.05-2.49), those belonging to richer households (AOR): 1.33, 95% CI: 1.07-1.65) and those whose parity was less than 2 (AOR): 1.52, 95% CI: 1.18-1.95).
RESULTS
We have identified modifiable factors (exposure to mass media, having been visited by a field health worker, having health insurance, having no big problems with distance to the nearest health facility, belonging to richer households, being married and educated), that can be targeted to improve utilization of the entire continuum of care. Promoting maternity services through mass media, strengthening the community health programmes, increasing access to health insurance and promoting girl child education to tertiary level may improve the level of utilization of maternity services.
CONCLUSION
[ "Pregnancy", "Child", "Infant, Newborn", "Female", "Humans", "Rwanda", "Health Facilities", "Health Surveys", "Marriage", "Delivery of Health Care" ]
9583497
null
null
null
null
Results
A total of 6,302 women were included in the analysis (Table 2). Of these, 2,131 (33.8%) (95% CI: 32.8–35.1) had complete continuum of care (further details in Table 3 and Supplementary file 1). Only 16 (0.3%) of the women were able to utilize at least eight ANC contacts. Majority of the women had less than four ANC contacts (52.8%), had had skilled birth attendance (94.3%), had a postnatal check (70.7%), Furthermore, majority of the women were aged 20 to 34 years (63.0%), had primary education (64.5%), had no exposure to internet (89.3%), were married (80.8%), working (75.7%) and resided in rural areas (82.2%). The mean age and household size were 31.56 ± 6.74 years and 5.14 ± 1.86 members respectively. Table 2Socio-demographic characteristics of women who gave birth within the last 5 years prior to the 2020 RDHSCharacteristicsN = 6,302% Age 35 to 49220735.020 to 34397063.015 to 191252.0 Household size 6 and above235737.4Less than 6394562.6 Exposure to newspapers/magazines No507080.5Yes123219.5 Exposure to radio No144823.0Yes485477.0 Exposure to TV No375359.5Yes254940.5 Sex of Household head Male478375.9Female151924.1 Internet access No562689.3Yes67610.7 Wealth Index Poorest144823.0Poorer121719.3Middle122419.4Richer123419.6Richest117818.7 Parity 1158725.22–4355056.35 and above116518.5 Marital Not married120819.2Married/cohabiting509480.8 Visited by a fieldworker No399463.4Yes230736.6 Has health insurance No119418.9Yes510881.1 Working status Not working153224.3Working477075.7 Permission to access healthcare Big problem2223.5Not big problem608096.5 Distance to health facility Big problem146123.2Not big problem484176.8 Region North100415.9East170227.0West142522.6South130520.7Kigali86613.7 Residence Rural517982.2Urban112317.8 Education Level No Education69811.1Primary Education407164.5Secondary Education125820.0Tertiary Education2754.4 Year of birth 201490.120155568.8201693214.82017129220.52018158825.32019163325.920202924.6 Socio-demographic characteristics of women who gave birth within the last 5 years prior to the 2020 RDHS Table 3Utilization of the different components of continuum of careServiceFrequencyN = 6,302%95% CI 4 or more ANC contacts 297547.246.1–48.6 Skilled birth attendance 595094.393.9–95.0 Maternal PNC 445670.769.7–72.0 Neonatal PNC 481576.475.5–77.6 Continuum of care 213133.832.8–35.1 Utilization of the different components of continuum of care [SUBTITLE] Factors associated with CoC utilisation [SUBSECTION] Factors associated with CoC utilisation are shown in Table 4. Women who had exposure to newspapers (adjusted odds ratio (AOR): 1.30, 95% CI: 1.11–1.52) those belonging to the eastern region (AOR): 1.24, 95% CI: 1.01–1.52), southern region (AOR): 1.26, 95% CI: 1.04–1.53), those with health insurance (AOR): 1.55, 95% CI: 1.30–1.85), those who had been visited by a field health worker (AOR: 1.31, 95% CI: 1.15–1.49), those with no big problems with distance to health facility (AOR): 1.25, 95% CI: 1.07–1.46), those who were married (AOR): 1.35, 95% CI: 1.11–1.64), those with tertiary level of education (AOR): 1.61, 95% CI: 1.05–2.49), those belonging to richer households (AOR): 1.33, 95% CI: 1.07–1.65) and those whose parity is less than 2 (AOR): 1.52, 95% CI: 1.18–1.95) were more likely to utilize complete continuum of care. Women belonging to the western region (AOR): 0.76, 95% CI: 0.61–0.94) were less likely to utilize complete continuum of care. Table 4Factors associated with CoC utilizationCharacteristicscrude modelcOR (95% CI)P-valueAdjusted modelaOR (95% CI)P-value Age 35 to 491125 to 341.12 (0.98–1.27)0.0970.92 (0.79–1.08)0.30115 to 240.89 (0.59–1.36)0.6000.75 (0.48–1.17)0.203 Household size 6 and above11Less than 61.27 (1.12–1.43)< 0.0011.12 (0.96–1.29)0.150 Exposure to newspapers/magazines No11Yes1.84 (1.59–2.13)< 0.0011.30 (1.11–1.52)0.001 Exposure to radio No11Yes1.62 (1.39–1.89)< 0.0011.15 (0.96–1.38)0.137 Exposure to TV No11Yes1.50 (1.32–1.71)< 0.0011.15 (0.99–1.34)0.075 Sex of Household head Male11Female0.84 (0.73–0.96)0.0110.99 (0.85–1.17)0.967 Internet No11Yes2.06 (1.70–2.49)< 0.0011.30 (0.98–1.73)0.66 Wealth Index Poorest11Poorer1.28 (1.06–1.55)0.0101.18 (0.97–1.44)0.101Middle1.52 (1.26–1.83)< 0.0011.34 (1.09–1.64)0.005Richer1.67 (1.39–2.02)< 0.0011.33 (1.07–1.65)0.009Richest2.01 (1.64–2.47)< 0.0011.21 (0.93–1.57)0.161 Parity 5 and above112–41.57 (1.32–1.86)< 0.0011.41 (1.15–1.74)0.001Less than 21.66 (1.36–2.03)< 0.0011.52 (1.18–1.95)0.001 Married/cohabiting No11Yes1.42 (1.22–1.65)< 0.0011.35 (1.11–1.64)0.002 Visited by a fieldworker No11Yes1.35 (1.19–1.52)< 0.0011.31 (1.15–1.49)< 0.001 Health insurance No11Yes1.99 (1.69–2.34)< 0.0011.55 (1.30–1.85)< 0.001 Working No 1- Yes 0.93 (0.81–1.07)0.308 Permission to access healthcare Big problem11Not big problem1.80 (1.29–2.52)0.0011.33 (0.92–1.91)0.129 Distance to health facility Big problem11Not big problem1.43 (1.24–1.66)< 0.0011.25 (1.07–1.46)0.004 Region North11East1.22 (0.99–1.48)0.0521.24 (1.01–1.52)0.037West0.73 (0.59–0.91)0.0060.76 (0.61–0.94)0.013South1.25 (1.03–1.52)0.0231.26 (1.04–1.53)0.021Kigali1.09 (0.84–1.42)0.5000.85 (0.66–1.11)0.240 Residence Rural1-Urban1.09 (0.92–1.29)0.330 Education level None11Primary1.27 (1.03–1.56)0.0240.95 (0.77–1.18)0.663Secondary1.58 (1.23–2.02)< 0.0010.92 (0.69–1.21)0.528Tertiary3.82 (2.75–5.30)< 0.0011.61 (1.05–2.49)0.031 Year of Birth 2014–20151120161.12 (0.90–1.40)0.3101.11 (0.88–1.41)0.37820170.95 (0.77–1.16)0.5930.95 (0.76–1.18)0.62420181.15 (0.93–1.42)0.2111.16 (0.92–145)0.20820191.19 (0.95–1.49)0.1251.11 (0.88–1.41)0.38320201.16 (0.83–1.61)0.3810.95 (0.67–1.33)0.759 Factors associated with CoC utilization Factors associated with CoC utilisation are shown in Table 4. Women who had exposure to newspapers (adjusted odds ratio (AOR): 1.30, 95% CI: 1.11–1.52) those belonging to the eastern region (AOR): 1.24, 95% CI: 1.01–1.52), southern region (AOR): 1.26, 95% CI: 1.04–1.53), those with health insurance (AOR): 1.55, 95% CI: 1.30–1.85), those who had been visited by a field health worker (AOR: 1.31, 95% CI: 1.15–1.49), those with no big problems with distance to health facility (AOR): 1.25, 95% CI: 1.07–1.46), those who were married (AOR): 1.35, 95% CI: 1.11–1.64), those with tertiary level of education (AOR): 1.61, 95% CI: 1.05–2.49), those belonging to richer households (AOR): 1.33, 95% CI: 1.07–1.65) and those whose parity is less than 2 (AOR): 1.52, 95% CI: 1.18–1.95) were more likely to utilize complete continuum of care. Women belonging to the western region (AOR): 0.76, 95% CI: 0.61–0.94) were less likely to utilize complete continuum of care. Table 4Factors associated with CoC utilizationCharacteristicscrude modelcOR (95% CI)P-valueAdjusted modelaOR (95% CI)P-value Age 35 to 491125 to 341.12 (0.98–1.27)0.0970.92 (0.79–1.08)0.30115 to 240.89 (0.59–1.36)0.6000.75 (0.48–1.17)0.203 Household size 6 and above11Less than 61.27 (1.12–1.43)< 0.0011.12 (0.96–1.29)0.150 Exposure to newspapers/magazines No11Yes1.84 (1.59–2.13)< 0.0011.30 (1.11–1.52)0.001 Exposure to radio No11Yes1.62 (1.39–1.89)< 0.0011.15 (0.96–1.38)0.137 Exposure to TV No11Yes1.50 (1.32–1.71)< 0.0011.15 (0.99–1.34)0.075 Sex of Household head Male11Female0.84 (0.73–0.96)0.0110.99 (0.85–1.17)0.967 Internet No11Yes2.06 (1.70–2.49)< 0.0011.30 (0.98–1.73)0.66 Wealth Index Poorest11Poorer1.28 (1.06–1.55)0.0101.18 (0.97–1.44)0.101Middle1.52 (1.26–1.83)< 0.0011.34 (1.09–1.64)0.005Richer1.67 (1.39–2.02)< 0.0011.33 (1.07–1.65)0.009Richest2.01 (1.64–2.47)< 0.0011.21 (0.93–1.57)0.161 Parity 5 and above112–41.57 (1.32–1.86)< 0.0011.41 (1.15–1.74)0.001Less than 21.66 (1.36–2.03)< 0.0011.52 (1.18–1.95)0.001 Married/cohabiting No11Yes1.42 (1.22–1.65)< 0.0011.35 (1.11–1.64)0.002 Visited by a fieldworker No11Yes1.35 (1.19–1.52)< 0.0011.31 (1.15–1.49)< 0.001 Health insurance No11Yes1.99 (1.69–2.34)< 0.0011.55 (1.30–1.85)< 0.001 Working No 1- Yes 0.93 (0.81–1.07)0.308 Permission to access healthcare Big problem11Not big problem1.80 (1.29–2.52)0.0011.33 (0.92–1.91)0.129 Distance to health facility Big problem11Not big problem1.43 (1.24–1.66)< 0.0011.25 (1.07–1.46)0.004 Region North11East1.22 (0.99–1.48)0.0521.24 (1.01–1.52)0.037West0.73 (0.59–0.91)0.0060.76 (0.61–0.94)0.013South1.25 (1.03–1.52)0.0231.26 (1.04–1.53)0.021Kigali1.09 (0.84–1.42)0.5000.85 (0.66–1.11)0.240 Residence Rural1-Urban1.09 (0.92–1.29)0.330 Education level None11Primary1.27 (1.03–1.56)0.0240.95 (0.77–1.18)0.663Secondary1.58 (1.23–2.02)< 0.0010.92 (0.69–1.21)0.528Tertiary3.82 (2.75–5.30)< 0.0011.61 (1.05–2.49)0.031 Year of Birth 2014–20151120161.12 (0.90–1.40)0.3101.11 (0.88–1.41)0.37820170.95 (0.77–1.16)0.5930.95 (0.76–1.18)0.62420181.15 (0.93–1.42)0.2111.16 (0.92–145)0.20820191.19 (0.95–1.49)0.1251.11 (0.88–1.41)0.38320201.16 (0.83–1.61)0.3810.95 (0.67–1.33)0.759 Factors associated with CoC utilization
Conclusion
Only 3 out of 10 women were able to utilize the entire continuum of maternity care services. Regarding ANC utilization, less than half (47.2%) and only 0.3% of women were able to utilize at least four and eight ANC contacts respectively a finding showing that Rwanda is not on track towards achieving the recommended eight ANC contacts for each pregnant woman in the 2016 WHO ANC guidelines. Belonging to eastern and southern Rwanda, exposure to mass media, having been visited by a field health worker, having health insurance, having no big problems with distance to the nearest health facility, belonging to richer households, having tertiary level of education, being married and having low parity were positively associated with completion of maternal and newborn continuum of care. Thus, we recommend the different stakeholders engaged in maternal health care to focus on women from western region, those without insurance, from poor households, with low levels of education, high parity and unmarried to ensure utilization of complete continuum of maternal and newborn care. Promoting maternity services through mass media, strengthening the community health programmes, increasing access to health insurance, strengthening and promoting girl child education to post-secondary level, increased support to household economic development programmes may improve the level of utilization of maternity services. Utilization of mass media specially to sensitize the population on the benefits of continuum of care should be encouraged with great attention given for early initiation of ANC, increasing the number of ANC contacts, health facility and skilled birth attendance during childbirth and postnatal care. During ANC contacts in health facilities and in the community by CHWs unmarried and high parity women need to be given more time during care, awareness on the importance of having CoC and followed up actively.
[ "Background", "Methods", "Setting", "Study sampling and participants", "Variables", "Outcome variables", "Independent variables", "Statistical analysis", "Factors associated with CoC utilisation", "" ]
[ "Globally, almost all neonatal deaths (75%) occur in the first week of life [1]. Majority of the global maternal and neonatal deaths occur in low and middle income countries (LMICs) with sub-Saharan Africa (SSA) having the largest burden and the least access to the complete continuum of maternal care [2, 3]. Furthermore, 98% of the global stillbirth burden occurs in LMICs with 75% occurring in SSA and south Asia [4].Timely access to antenatal care (ANC), skilled birth attendance (SBA) and postnatal (PNC) care are key evidence based strategies that have been proven to ensure a reduction in stillbirths, maternal and neonatal mortality and morbidity [5–7]. Accordingly, the world health organization (WHO) highly advocates for the implementation of continuum of maternal and newborn care which ensures continual access to care from preconception through to the postpartum period [8, 9].\nSustainable Development Goal (SDG) three targets to reduce maternal and neonatal mortality to less than 70 deaths per 100 000 livebirths and 12 per 1000 livebirths respectively [10]. As much as they sound ambitious, they reaffirmed the global prioritization of maternal child health (MCH). However, achieving this will need a high universal coverage of the components of maternal and newborn care for early detection of complications to enable timely intervention [10–12]. Therefore, a clear understanding of the utilization behavior of the components of maternal and newborn continuum of care is vital not only towards reducing inequity in health but also towards designing and implementing better strategies aimed at improving the unacceptably poor MCH indicators [13].\nAlthough the 1994 genocide devastated Rwanda’s health system, efforts were put in place to ensure that the system is rebuilt which partly contributed to the country’s achievement of the Millennium-um Development Goals (MDG) 4 of reducing the under-five mortality rate by two-thirds and MDG 5 of reducing the maternal mortality ratio by three-quarters [14–16]. However, Rwanda has not had similarly sized reductions in neonatal mortality compared to non-neonatal under-5 mortality [17, 18] with neonatal mortality rate (NMR) currently at 19 deaths per 1,000 live births and contributing over 42% of under 5 mortality [19]. Furthermore, although Rwanda reduced its the maternal mortality ratio (MMR) from 750 deaths per 100,000 live births to 210 deaths per 100,000 live births within a decade (2005–2015), the MMR has stagnated in the last five years from 210 to 203 deaths per 100,000 live births [19, 20].\nThe 2020 Rwanda Demographic and Health Survey (RDHS) reports variation in maternal care utilization with only 47% of the women accessing at least four ANC contacts compared to SBA of 94% [19]. Given that WHO modified the minimum number of ANC visits from four to at least eight contacts in 2016, the current ANC utilization in Rwanda is far behind the global recommendations [21–23]. Therefore, in order for Rwanda to reduce the MMR to less than 70 per 100,000 live births and the NMR to less than 12 per 1000 live births by 2030, up-to-date data is needed to further guide the ongoing maternal and newborn health projects. Studies that have looked at continuum of care (CoC) have combined Rwanda’s DHS data with other countries ending up with results that cannot be specific to Rwanda [13, 24]. Hategeka et al. only looked at single measures (ANC and place of delivery), PNC, SBA were not considered in this study [25]. Therefore, we used the most recent nationally representative survey data to determine level and predictors of successfully completing CoC.", "[SUBTITLE] Setting [SUBSECTION] Rwanda is a central-eastern African nation of about 12 million people [19, 25] whose health system consists of eight national referral hospitals, four hospitals at provincial level, 35 district level hospitals, 495 health centers, 406 health posts and over 45,000 community health workers (CHWs) [20, 26, 27]. Each village has a male-female CHW pair and one female Agent de Sante Maternelle (ASM) and these CHWs are responsible in delivering the first line of health services including maternal and newborn health services [20, 28]. The country has a universal, community-based health insurance program that has a household subscription and co-payments at the time of care and all citizens are eligible to enroll into it [20, 29].\nRwanda is a central-eastern African nation of about 12 million people [19, 25] whose health system consists of eight national referral hospitals, four hospitals at provincial level, 35 district level hospitals, 495 health centers, 406 health posts and over 45,000 community health workers (CHWs) [20, 26, 27]. Each village has a male-female CHW pair and one female Agent de Sante Maternelle (ASM) and these CHWs are responsible in delivering the first line of health services including maternal and newborn health services [20, 28]. The country has a universal, community-based health insurance program that has a household subscription and co-payments at the time of care and all citizens are eligible to enroll into it [20, 29].\n[SUBTITLE] Study sampling and participants [SUBSECTION] The 2019-20 Rwanda Demographic Survey (RDHS) was used for this analysis employed a two-stage sample design with the first stage involving sample points (clusters) selection consisting of enumeration areas (EAs) [19]. Between November 2019 and July 2020, a total of 13,005 households were selected from the first stage selected EAs [19]. The household and the woman’s questionnaires provided the data used in this secondary analysis.\nEligibility for women to participate in the RDHS was being aged between 15 and 49 years and being either permanent residents of the selected households or visitors who stayed in the household the night before the survey [19]. Out of the total 13,005 households originally sampled, 12,951 had occupants and 12,949 were interviewed [19]. We included only women who had given birth within five years preceding the survey in this analysis. Out of the 14,675 women found eligible for the RDHS, the team was able to interview 14,634 women of which only 6,302 women had given birth within the last five years preceding the survey [19].\nThe 2019-20 Rwanda Demographic Survey (RDHS) was used for this analysis employed a two-stage sample design with the first stage involving sample points (clusters) selection consisting of enumeration areas (EAs) [19]. Between November 2019 and July 2020, a total of 13,005 households were selected from the first stage selected EAs [19]. The household and the woman’s questionnaires provided the data used in this secondary analysis.\nEligibility for women to participate in the RDHS was being aged between 15 and 49 years and being either permanent residents of the selected households or visitors who stayed in the household the night before the survey [19]. Out of the total 13,005 households originally sampled, 12,951 had occupants and 12,949 were interviewed [19]. We included only women who had given birth within five years preceding the survey in this analysis. Out of the 14,675 women found eligible for the RDHS, the team was able to interview 14,634 women of which only 6,302 women had given birth within the last five years preceding the survey [19].\n[SUBTITLE] Variables [SUBSECTION] [SUBTITLE] Outcome variables [SUBSECTION] Complete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].\nComplete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].\n[SUBTITLE] Independent variables [SUBSECTION] Nineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)\nNineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)\n[SUBTITLE] Outcome variables [SUBSECTION] Complete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].\nComplete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].\n[SUBTITLE] Independent variables [SUBSECTION] Nineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)\nNineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)\n[SUBTITLE] Statistical analysis [SUBSECTION] We used the complex sample package of SPSS (version 25.0) statistical software which accounted for the multi-stage cluster study design [35]. Individual sample weight, sample strata In order to ensure representativeness of the survey results at the national and regional level and to minimize the effects of unequal probability sampling in different strata, data were weighted [12]. Initially, we did descriptive statistics. Frequencies and proportions/percentages for categorical variables have been presented. We then used bivariable logistic regression to assess the association between each independent variable and the outcome whose crude odds ratio (COR), 95% confidence interval (CI) and p-values have been presented. Independent variables found significant at p-value less than 0.25 at bivariable level with other independent variables were considered for multivariable logistic regression to assess the independent effect of each variable on the CoC utilisation [6]. Before multivariable analysis, multicollinearity was assessed using variance inflation factor (VIF) and no VIF above 2.5 was observed. Model fitness was assessed with the F-test with a p-value of < 0.001. Adjusted odds ratios (aOR), 95% confidence intervals (CI) and p-values were calculated with statistical significance level set at p-value < 0.05.\nWe used the complex sample package of SPSS (version 25.0) statistical software which accounted for the multi-stage cluster study design [35]. Individual sample weight, sample strata In order to ensure representativeness of the survey results at the national and regional level and to minimize the effects of unequal probability sampling in different strata, data were weighted [12]. Initially, we did descriptive statistics. Frequencies and proportions/percentages for categorical variables have been presented. We then used bivariable logistic regression to assess the association between each independent variable and the outcome whose crude odds ratio (COR), 95% confidence interval (CI) and p-values have been presented. Independent variables found significant at p-value less than 0.25 at bivariable level with other independent variables were considered for multivariable logistic regression to assess the independent effect of each variable on the CoC utilisation [6]. Before multivariable analysis, multicollinearity was assessed using variance inflation factor (VIF) and no VIF above 2.5 was observed. Model fitness was assessed with the F-test with a p-value of < 0.001. Adjusted odds ratios (aOR), 95% confidence intervals (CI) and p-values were calculated with statistical significance level set at p-value < 0.05.", "Rwanda is a central-eastern African nation of about 12 million people [19, 25] whose health system consists of eight national referral hospitals, four hospitals at provincial level, 35 district level hospitals, 495 health centers, 406 health posts and over 45,000 community health workers (CHWs) [20, 26, 27]. Each village has a male-female CHW pair and one female Agent de Sante Maternelle (ASM) and these CHWs are responsible in delivering the first line of health services including maternal and newborn health services [20, 28]. The country has a universal, community-based health insurance program that has a household subscription and co-payments at the time of care and all citizens are eligible to enroll into it [20, 29].", "The 2019-20 Rwanda Demographic Survey (RDHS) was used for this analysis employed a two-stage sample design with the first stage involving sample points (clusters) selection consisting of enumeration areas (EAs) [19]. Between November 2019 and July 2020, a total of 13,005 households were selected from the first stage selected EAs [19]. The household and the woman’s questionnaires provided the data used in this secondary analysis.\nEligibility for women to participate in the RDHS was being aged between 15 and 49 years and being either permanent residents of the selected households or visitors who stayed in the household the night before the survey [19]. Out of the total 13,005 households originally sampled, 12,951 had occupants and 12,949 were interviewed [19]. We included only women who had given birth within five years preceding the survey in this analysis. Out of the 14,675 women found eligible for the RDHS, the team was able to interview 14,634 women of which only 6,302 women had given birth within the last five years preceding the survey [19].", "[SUBTITLE] Outcome variables [SUBSECTION] Complete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].\nComplete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].\n[SUBTITLE] Independent variables [SUBSECTION] Nineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)\nNineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)", "Complete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].", "Nineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)", "We used the complex sample package of SPSS (version 25.0) statistical software which accounted for the multi-stage cluster study design [35]. Individual sample weight, sample strata In order to ensure representativeness of the survey results at the national and regional level and to minimize the effects of unequal probability sampling in different strata, data were weighted [12]. Initially, we did descriptive statistics. Frequencies and proportions/percentages for categorical variables have been presented. We then used bivariable logistic regression to assess the association between each independent variable and the outcome whose crude odds ratio (COR), 95% confidence interval (CI) and p-values have been presented. Independent variables found significant at p-value less than 0.25 at bivariable level with other independent variables were considered for multivariable logistic regression to assess the independent effect of each variable on the CoC utilisation [6]. Before multivariable analysis, multicollinearity was assessed using variance inflation factor (VIF) and no VIF above 2.5 was observed. Model fitness was assessed with the F-test with a p-value of < 0.001. Adjusted odds ratios (aOR), 95% confidence intervals (CI) and p-values were calculated with statistical significance level set at p-value < 0.05.", "Factors associated with CoC utilisation are shown in Table 4. Women who had exposure to newspapers (adjusted odds ratio (AOR): 1.30, 95% CI: 1.11–1.52) those belonging to the eastern region (AOR): 1.24, 95% CI: 1.01–1.52), southern region (AOR): 1.26, 95% CI: 1.04–1.53), those with health insurance (AOR): 1.55, 95% CI: 1.30–1.85), those who had been visited by a field health worker (AOR: 1.31, 95% CI: 1.15–1.49), those with no big problems with distance to health facility (AOR): 1.25, 95% CI: 1.07–1.46), those who were married (AOR): 1.35, 95% CI: 1.11–1.64), those with tertiary level of education (AOR): 1.61, 95% CI: 1.05–2.49), those belonging to richer households (AOR): 1.33, 95% CI: 1.07–1.65) and those whose parity is less than 2 (AOR): 1.52, 95% CI: 1.18–1.95) were more likely to utilize complete continuum of care. Women belonging to the western region (AOR): 0.76, 95% CI: 0.61–0.94) were less likely to utilize complete continuum of care.\n\nTable 4Factors associated with CoC utilizationCharacteristicscrude modelcOR (95% CI)P-valueAdjusted modelaOR (95% CI)P-value\nAge\n35 to 491125 to 341.12 (0.98–1.27)0.0970.92 (0.79–1.08)0.30115 to 240.89 (0.59–1.36)0.6000.75 (0.48–1.17)0.203\nHousehold size\n6 and above11Less than 61.27 (1.12–1.43)< 0.0011.12 (0.96–1.29)0.150\nExposure to newspapers/magazines\nNo11Yes1.84 (1.59–2.13)< 0.0011.30 (1.11–1.52)0.001\nExposure to radio\nNo11Yes1.62 (1.39–1.89)< 0.0011.15 (0.96–1.38)0.137\nExposure to TV\nNo11Yes1.50 (1.32–1.71)< 0.0011.15 (0.99–1.34)0.075\nSex of Household head\nMale11Female0.84 (0.73–0.96)0.0110.99 (0.85–1.17)0.967\nInternet\nNo11Yes2.06 (1.70–2.49)< 0.0011.30 (0.98–1.73)0.66\nWealth Index\nPoorest11Poorer1.28 (1.06–1.55)0.0101.18 (0.97–1.44)0.101Middle1.52 (1.26–1.83)< 0.0011.34 (1.09–1.64)0.005Richer1.67 (1.39–2.02)< 0.0011.33 (1.07–1.65)0.009Richest2.01 (1.64–2.47)< 0.0011.21 (0.93–1.57)0.161\nParity\n5 and above112–41.57 (1.32–1.86)< 0.0011.41 (1.15–1.74)0.001Less than 21.66 (1.36–2.03)< 0.0011.52 (1.18–1.95)0.001\nMarried/cohabiting\nNo11Yes1.42 (1.22–1.65)< 0.0011.35 (1.11–1.64)0.002\nVisited by a fieldworker\nNo11Yes1.35 (1.19–1.52)< 0.0011.31 (1.15–1.49)< 0.001\nHealth insurance\nNo11Yes1.99 (1.69–2.34)< 0.0011.55 (1.30–1.85)< 0.001\nWorking\n\nNo\n1-\nYes\n0.93 (0.81–1.07)0.308\nPermission to access healthcare\nBig problem11Not big problem1.80 (1.29–2.52)0.0011.33 (0.92–1.91)0.129\nDistance to health facility\nBig problem11Not big problem1.43 (1.24–1.66)< 0.0011.25 (1.07–1.46)0.004\nRegion\nNorth11East1.22 (0.99–1.48)0.0521.24 (1.01–1.52)0.037West0.73 (0.59–0.91)0.0060.76 (0.61–0.94)0.013South1.25 (1.03–1.52)0.0231.26 (1.04–1.53)0.021Kigali1.09 (0.84–1.42)0.5000.85 (0.66–1.11)0.240\nResidence\nRural1-Urban1.09 (0.92–1.29)0.330\nEducation level\nNone11Primary1.27 (1.03–1.56)0.0240.95 (0.77–1.18)0.663Secondary1.58 (1.23–2.02)< 0.0010.92 (0.69–1.21)0.528Tertiary3.82 (2.75–5.30)< 0.0011.61 (1.05–2.49)0.031\nYear of Birth\n2014–20151120161.12 (0.90–1.40)0.3101.11 (0.88–1.41)0.37820170.95 (0.77–1.16)0.5930.95 (0.76–1.18)0.62420181.15 (0.93–1.42)0.2111.16 (0.92–145)0.20820191.19 (0.95–1.49)0.1251.11 (0.88–1.41)0.38320201.16 (0.83–1.61)0.3810.95 (0.67–1.33)0.759\n\nFactors associated with CoC utilization", "Below is the link to the electronic supplementary material.\n\n\nSupplementary Material 1\n\n\n\nSupplementary Material 1\n" ]
[ null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Setting", "Study sampling and participants", "Variables", "Outcome variables", "Independent variables", "Statistical analysis", "Results", "Factors associated with CoC utilisation", "Discussion", "Conclusion", "Electronic supplementary material", "" ]
[ "Globally, almost all neonatal deaths (75%) occur in the first week of life [1]. Majority of the global maternal and neonatal deaths occur in low and middle income countries (LMICs) with sub-Saharan Africa (SSA) having the largest burden and the least access to the complete continuum of maternal care [2, 3]. Furthermore, 98% of the global stillbirth burden occurs in LMICs with 75% occurring in SSA and south Asia [4].Timely access to antenatal care (ANC), skilled birth attendance (SBA) and postnatal (PNC) care are key evidence based strategies that have been proven to ensure a reduction in stillbirths, maternal and neonatal mortality and morbidity [5–7]. Accordingly, the world health organization (WHO) highly advocates for the implementation of continuum of maternal and newborn care which ensures continual access to care from preconception through to the postpartum period [8, 9].\nSustainable Development Goal (SDG) three targets to reduce maternal and neonatal mortality to less than 70 deaths per 100 000 livebirths and 12 per 1000 livebirths respectively [10]. As much as they sound ambitious, they reaffirmed the global prioritization of maternal child health (MCH). However, achieving this will need a high universal coverage of the components of maternal and newborn care for early detection of complications to enable timely intervention [10–12]. Therefore, a clear understanding of the utilization behavior of the components of maternal and newborn continuum of care is vital not only towards reducing inequity in health but also towards designing and implementing better strategies aimed at improving the unacceptably poor MCH indicators [13].\nAlthough the 1994 genocide devastated Rwanda’s health system, efforts were put in place to ensure that the system is rebuilt which partly contributed to the country’s achievement of the Millennium-um Development Goals (MDG) 4 of reducing the under-five mortality rate by two-thirds and MDG 5 of reducing the maternal mortality ratio by three-quarters [14–16]. However, Rwanda has not had similarly sized reductions in neonatal mortality compared to non-neonatal under-5 mortality [17, 18] with neonatal mortality rate (NMR) currently at 19 deaths per 1,000 live births and contributing over 42% of under 5 mortality [19]. Furthermore, although Rwanda reduced its the maternal mortality ratio (MMR) from 750 deaths per 100,000 live births to 210 deaths per 100,000 live births within a decade (2005–2015), the MMR has stagnated in the last five years from 210 to 203 deaths per 100,000 live births [19, 20].\nThe 2020 Rwanda Demographic and Health Survey (RDHS) reports variation in maternal care utilization with only 47% of the women accessing at least four ANC contacts compared to SBA of 94% [19]. Given that WHO modified the minimum number of ANC visits from four to at least eight contacts in 2016, the current ANC utilization in Rwanda is far behind the global recommendations [21–23]. Therefore, in order for Rwanda to reduce the MMR to less than 70 per 100,000 live births and the NMR to less than 12 per 1000 live births by 2030, up-to-date data is needed to further guide the ongoing maternal and newborn health projects. Studies that have looked at continuum of care (CoC) have combined Rwanda’s DHS data with other countries ending up with results that cannot be specific to Rwanda [13, 24]. Hategeka et al. only looked at single measures (ANC and place of delivery), PNC, SBA were not considered in this study [25]. Therefore, we used the most recent nationally representative survey data to determine level and predictors of successfully completing CoC.", "[SUBTITLE] Setting [SUBSECTION] Rwanda is a central-eastern African nation of about 12 million people [19, 25] whose health system consists of eight national referral hospitals, four hospitals at provincial level, 35 district level hospitals, 495 health centers, 406 health posts and over 45,000 community health workers (CHWs) [20, 26, 27]. Each village has a male-female CHW pair and one female Agent de Sante Maternelle (ASM) and these CHWs are responsible in delivering the first line of health services including maternal and newborn health services [20, 28]. The country has a universal, community-based health insurance program that has a household subscription and co-payments at the time of care and all citizens are eligible to enroll into it [20, 29].\nRwanda is a central-eastern African nation of about 12 million people [19, 25] whose health system consists of eight national referral hospitals, four hospitals at provincial level, 35 district level hospitals, 495 health centers, 406 health posts and over 45,000 community health workers (CHWs) [20, 26, 27]. Each village has a male-female CHW pair and one female Agent de Sante Maternelle (ASM) and these CHWs are responsible in delivering the first line of health services including maternal and newborn health services [20, 28]. The country has a universal, community-based health insurance program that has a household subscription and co-payments at the time of care and all citizens are eligible to enroll into it [20, 29].\n[SUBTITLE] Study sampling and participants [SUBSECTION] The 2019-20 Rwanda Demographic Survey (RDHS) was used for this analysis employed a two-stage sample design with the first stage involving sample points (clusters) selection consisting of enumeration areas (EAs) [19]. Between November 2019 and July 2020, a total of 13,005 households were selected from the first stage selected EAs [19]. The household and the woman’s questionnaires provided the data used in this secondary analysis.\nEligibility for women to participate in the RDHS was being aged between 15 and 49 years and being either permanent residents of the selected households or visitors who stayed in the household the night before the survey [19]. Out of the total 13,005 households originally sampled, 12,951 had occupants and 12,949 were interviewed [19]. We included only women who had given birth within five years preceding the survey in this analysis. Out of the 14,675 women found eligible for the RDHS, the team was able to interview 14,634 women of which only 6,302 women had given birth within the last five years preceding the survey [19].\nThe 2019-20 Rwanda Demographic Survey (RDHS) was used for this analysis employed a two-stage sample design with the first stage involving sample points (clusters) selection consisting of enumeration areas (EAs) [19]. Between November 2019 and July 2020, a total of 13,005 households were selected from the first stage selected EAs [19]. The household and the woman’s questionnaires provided the data used in this secondary analysis.\nEligibility for women to participate in the RDHS was being aged between 15 and 49 years and being either permanent residents of the selected households or visitors who stayed in the household the night before the survey [19]. Out of the total 13,005 households originally sampled, 12,951 had occupants and 12,949 were interviewed [19]. We included only women who had given birth within five years preceding the survey in this analysis. Out of the 14,675 women found eligible for the RDHS, the team was able to interview 14,634 women of which only 6,302 women had given birth within the last five years preceding the survey [19].\n[SUBTITLE] Variables [SUBSECTION] [SUBTITLE] Outcome variables [SUBSECTION] Complete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].\nComplete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].\n[SUBTITLE] Independent variables [SUBSECTION] Nineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)\nNineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)\n[SUBTITLE] Outcome variables [SUBSECTION] Complete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].\nComplete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].\n[SUBTITLE] Independent variables [SUBSECTION] Nineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)\nNineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)\n[SUBTITLE] Statistical analysis [SUBSECTION] We used the complex sample package of SPSS (version 25.0) statistical software which accounted for the multi-stage cluster study design [35]. Individual sample weight, sample strata In order to ensure representativeness of the survey results at the national and regional level and to minimize the effects of unequal probability sampling in different strata, data were weighted [12]. Initially, we did descriptive statistics. Frequencies and proportions/percentages for categorical variables have been presented. We then used bivariable logistic regression to assess the association between each independent variable and the outcome whose crude odds ratio (COR), 95% confidence interval (CI) and p-values have been presented. Independent variables found significant at p-value less than 0.25 at bivariable level with other independent variables were considered for multivariable logistic regression to assess the independent effect of each variable on the CoC utilisation [6]. Before multivariable analysis, multicollinearity was assessed using variance inflation factor (VIF) and no VIF above 2.5 was observed. Model fitness was assessed with the F-test with a p-value of < 0.001. Adjusted odds ratios (aOR), 95% confidence intervals (CI) and p-values were calculated with statistical significance level set at p-value < 0.05.\nWe used the complex sample package of SPSS (version 25.0) statistical software which accounted for the multi-stage cluster study design [35]. Individual sample weight, sample strata In order to ensure representativeness of the survey results at the national and regional level and to minimize the effects of unequal probability sampling in different strata, data were weighted [12]. Initially, we did descriptive statistics. Frequencies and proportions/percentages for categorical variables have been presented. We then used bivariable logistic regression to assess the association between each independent variable and the outcome whose crude odds ratio (COR), 95% confidence interval (CI) and p-values have been presented. Independent variables found significant at p-value less than 0.25 at bivariable level with other independent variables were considered for multivariable logistic regression to assess the independent effect of each variable on the CoC utilisation [6]. Before multivariable analysis, multicollinearity was assessed using variance inflation factor (VIF) and no VIF above 2.5 was observed. Model fitness was assessed with the F-test with a p-value of < 0.001. Adjusted odds ratios (aOR), 95% confidence intervals (CI) and p-values were calculated with statistical significance level set at p-value < 0.05.", "Rwanda is a central-eastern African nation of about 12 million people [19, 25] whose health system consists of eight national referral hospitals, four hospitals at provincial level, 35 district level hospitals, 495 health centers, 406 health posts and over 45,000 community health workers (CHWs) [20, 26, 27]. Each village has a male-female CHW pair and one female Agent de Sante Maternelle (ASM) and these CHWs are responsible in delivering the first line of health services including maternal and newborn health services [20, 28]. The country has a universal, community-based health insurance program that has a household subscription and co-payments at the time of care and all citizens are eligible to enroll into it [20, 29].", "The 2019-20 Rwanda Demographic Survey (RDHS) was used for this analysis employed a two-stage sample design with the first stage involving sample points (clusters) selection consisting of enumeration areas (EAs) [19]. Between November 2019 and July 2020, a total of 13,005 households were selected from the first stage selected EAs [19]. The household and the woman’s questionnaires provided the data used in this secondary analysis.\nEligibility for women to participate in the RDHS was being aged between 15 and 49 years and being either permanent residents of the selected households or visitors who stayed in the household the night before the survey [19]. Out of the total 13,005 households originally sampled, 12,951 had occupants and 12,949 were interviewed [19]. We included only women who had given birth within five years preceding the survey in this analysis. Out of the 14,675 women found eligible for the RDHS, the team was able to interview 14,634 women of which only 6,302 women had given birth within the last five years preceding the survey [19].", "[SUBTITLE] Outcome variables [SUBSECTION] Complete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].\nComplete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].\n[SUBTITLE] Independent variables [SUBSECTION] Nineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)\nNineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)", "Complete continuum of maternal and newborn healthcare was coded 1 and incomplete coded as 0 [6]. Complete continuum was considered as having utilized all the three maternal healthcare services; at least four ANC contacts, SBA, at least one maternal and neonatal PNC checkup within six weeks after childbirth [6, 30, 31].", "Nineteen independent variables were categorized into women and household characteristics, and were chosen basing on previous studies [32–34] and availability in the RDHS database as shown in Table 1.\n\nTable 1Categorization of independent variablesVariableCategorizationExposure to newspapers or magazinesYes, and NoExposure to internetYes and NoExposure to radioYes and NoExposure to television (TV)Yes and NoAccess to internetYes and NoAge15–24 years, 25–34 years and 35–49 yearsResidenceUrban and RuralRegionNorth, East, South, West and KigaliHousehold sizeLess than 6 and 6 and above.(this was based on the average household size of 5)Sex of household headMale and FemaleParity1, 2–4, 5, and aboveLevel of educationNo education, primary, secondary, and tertiary(highest level of education attended)Working statusYes and NoWealth indexRichest, richer, middle, poorer, and poorest quintilesHaving health insuranceYes and NoHaving been visited by a field health worker within the last 12 monthsYes and NoProblems seeking permission to get medical careNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Problems with distance to nearest health facilityNo big problems and big problems(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)Marital statusMarried/cohabiting and Not marriedYear of birth2014–2015, 2016, 2017, 2018, 2019 and 2020(we combined 2014 with 2015 because 2014 only had 9 births)\n\nCategorization of independent variables\nLess than 6 and 6 and above.\n(this was based on the average household size of 5)\nNo education, primary, secondary, and tertiary\n(highest level of education attended)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\nNo big problems and big problems\n(RDHS had three original categories (no problem, no big problem and big problem) however, after data collection, no woman reported no problem)\n2014–2015, 2016, 2017, 2018, 2019 and 2020\n(we combined 2014 with 2015 because 2014 only had 9 births)", "We used the complex sample package of SPSS (version 25.0) statistical software which accounted for the multi-stage cluster study design [35]. Individual sample weight, sample strata In order to ensure representativeness of the survey results at the national and regional level and to minimize the effects of unequal probability sampling in different strata, data were weighted [12]. Initially, we did descriptive statistics. Frequencies and proportions/percentages for categorical variables have been presented. We then used bivariable logistic regression to assess the association between each independent variable and the outcome whose crude odds ratio (COR), 95% confidence interval (CI) and p-values have been presented. Independent variables found significant at p-value less than 0.25 at bivariable level with other independent variables were considered for multivariable logistic regression to assess the independent effect of each variable on the CoC utilisation [6]. Before multivariable analysis, multicollinearity was assessed using variance inflation factor (VIF) and no VIF above 2.5 was observed. Model fitness was assessed with the F-test with a p-value of < 0.001. Adjusted odds ratios (aOR), 95% confidence intervals (CI) and p-values were calculated with statistical significance level set at p-value < 0.05.", "A total of 6,302 women were included in the analysis (Table 2). Of these, 2,131 (33.8%) (95% CI: 32.8–35.1) had complete continuum of care (further details in Table 3 and Supplementary file 1). Only 16 (0.3%) of the women were able to utilize at least eight ANC contacts. Majority of the women had less than four ANC contacts (52.8%), had had skilled birth attendance (94.3%), had a postnatal check (70.7%), Furthermore, majority of the women were aged 20 to 34 years (63.0%), had primary education (64.5%), had no exposure to internet (89.3%), were married (80.8%), working (75.7%) and resided in rural areas (82.2%). The mean age and household size were 31.56 ± 6.74 years and 5.14 ± 1.86 members respectively.\n\nTable 2Socio-demographic characteristics of women who gave birth within the last 5 years prior to the 2020 RDHSCharacteristicsN = 6,302%\nAge\n35 to 49220735.020 to 34397063.015 to 191252.0\nHousehold size\n6 and above235737.4Less than 6394562.6\nExposure to newspapers/magazines\nNo507080.5Yes123219.5\nExposure to radio\nNo144823.0Yes485477.0\nExposure to TV\nNo375359.5Yes254940.5\nSex of Household head\nMale478375.9Female151924.1\nInternet access\nNo562689.3Yes67610.7\nWealth Index\nPoorest144823.0Poorer121719.3Middle122419.4Richer123419.6Richest117818.7\nParity\n1158725.22–4355056.35 and above116518.5\nMarital\nNot married120819.2Married/cohabiting509480.8\nVisited by a fieldworker\nNo399463.4Yes230736.6\nHas health insurance\nNo119418.9Yes510881.1\nWorking status\nNot working153224.3Working477075.7\nPermission to access healthcare\nBig problem2223.5Not big problem608096.5\nDistance to health facility\nBig problem146123.2Not big problem484176.8\nRegion\nNorth100415.9East170227.0West142522.6South130520.7Kigali86613.7\nResidence\nRural517982.2Urban112317.8\nEducation Level\nNo Education69811.1Primary Education407164.5Secondary Education125820.0Tertiary Education2754.4\nYear of birth\n201490.120155568.8201693214.82017129220.52018158825.32019163325.920202924.6\n\nSocio-demographic characteristics of women who gave birth within the last 5 years prior to the 2020 RDHS\n\nTable 3Utilization of the different components of continuum of careServiceFrequencyN = 6,302%95% CI\n4 or more ANC contacts\n297547.246.1–48.6\nSkilled birth attendance\n595094.393.9–95.0\nMaternal PNC\n445670.769.7–72.0\nNeonatal PNC\n481576.475.5–77.6\nContinuum of care\n213133.832.8–35.1\n\nUtilization of the different components of continuum of care\n[SUBTITLE] Factors associated with CoC utilisation [SUBSECTION] Factors associated with CoC utilisation are shown in Table 4. Women who had exposure to newspapers (adjusted odds ratio (AOR): 1.30, 95% CI: 1.11–1.52) those belonging to the eastern region (AOR): 1.24, 95% CI: 1.01–1.52), southern region (AOR): 1.26, 95% CI: 1.04–1.53), those with health insurance (AOR): 1.55, 95% CI: 1.30–1.85), those who had been visited by a field health worker (AOR: 1.31, 95% CI: 1.15–1.49), those with no big problems with distance to health facility (AOR): 1.25, 95% CI: 1.07–1.46), those who were married (AOR): 1.35, 95% CI: 1.11–1.64), those with tertiary level of education (AOR): 1.61, 95% CI: 1.05–2.49), those belonging to richer households (AOR): 1.33, 95% CI: 1.07–1.65) and those whose parity is less than 2 (AOR): 1.52, 95% CI: 1.18–1.95) were more likely to utilize complete continuum of care. Women belonging to the western region (AOR): 0.76, 95% CI: 0.61–0.94) were less likely to utilize complete continuum of care.\n\nTable 4Factors associated with CoC utilizationCharacteristicscrude modelcOR (95% CI)P-valueAdjusted modelaOR (95% CI)P-value\nAge\n35 to 491125 to 341.12 (0.98–1.27)0.0970.92 (0.79–1.08)0.30115 to 240.89 (0.59–1.36)0.6000.75 (0.48–1.17)0.203\nHousehold size\n6 and above11Less than 61.27 (1.12–1.43)< 0.0011.12 (0.96–1.29)0.150\nExposure to newspapers/magazines\nNo11Yes1.84 (1.59–2.13)< 0.0011.30 (1.11–1.52)0.001\nExposure to radio\nNo11Yes1.62 (1.39–1.89)< 0.0011.15 (0.96–1.38)0.137\nExposure to TV\nNo11Yes1.50 (1.32–1.71)< 0.0011.15 (0.99–1.34)0.075\nSex of Household head\nMale11Female0.84 (0.73–0.96)0.0110.99 (0.85–1.17)0.967\nInternet\nNo11Yes2.06 (1.70–2.49)< 0.0011.30 (0.98–1.73)0.66\nWealth Index\nPoorest11Poorer1.28 (1.06–1.55)0.0101.18 (0.97–1.44)0.101Middle1.52 (1.26–1.83)< 0.0011.34 (1.09–1.64)0.005Richer1.67 (1.39–2.02)< 0.0011.33 (1.07–1.65)0.009Richest2.01 (1.64–2.47)< 0.0011.21 (0.93–1.57)0.161\nParity\n5 and above112–41.57 (1.32–1.86)< 0.0011.41 (1.15–1.74)0.001Less than 21.66 (1.36–2.03)< 0.0011.52 (1.18–1.95)0.001\nMarried/cohabiting\nNo11Yes1.42 (1.22–1.65)< 0.0011.35 (1.11–1.64)0.002\nVisited by a fieldworker\nNo11Yes1.35 (1.19–1.52)< 0.0011.31 (1.15–1.49)< 0.001\nHealth insurance\nNo11Yes1.99 (1.69–2.34)< 0.0011.55 (1.30–1.85)< 0.001\nWorking\n\nNo\n1-\nYes\n0.93 (0.81–1.07)0.308\nPermission to access healthcare\nBig problem11Not big problem1.80 (1.29–2.52)0.0011.33 (0.92–1.91)0.129\nDistance to health facility\nBig problem11Not big problem1.43 (1.24–1.66)< 0.0011.25 (1.07–1.46)0.004\nRegion\nNorth11East1.22 (0.99–1.48)0.0521.24 (1.01–1.52)0.037West0.73 (0.59–0.91)0.0060.76 (0.61–0.94)0.013South1.25 (1.03–1.52)0.0231.26 (1.04–1.53)0.021Kigali1.09 (0.84–1.42)0.5000.85 (0.66–1.11)0.240\nResidence\nRural1-Urban1.09 (0.92–1.29)0.330\nEducation level\nNone11Primary1.27 (1.03–1.56)0.0240.95 (0.77–1.18)0.663Secondary1.58 (1.23–2.02)< 0.0010.92 (0.69–1.21)0.528Tertiary3.82 (2.75–5.30)< 0.0011.61 (1.05–2.49)0.031\nYear of Birth\n2014–20151120161.12 (0.90–1.40)0.3101.11 (0.88–1.41)0.37820170.95 (0.77–1.16)0.5930.95 (0.76–1.18)0.62420181.15 (0.93–1.42)0.2111.16 (0.92–145)0.20820191.19 (0.95–1.49)0.1251.11 (0.88–1.41)0.38320201.16 (0.83–1.61)0.3810.95 (0.67–1.33)0.759\n\nFactors associated with CoC utilization\nFactors associated with CoC utilisation are shown in Table 4. Women who had exposure to newspapers (adjusted odds ratio (AOR): 1.30, 95% CI: 1.11–1.52) those belonging to the eastern region (AOR): 1.24, 95% CI: 1.01–1.52), southern region (AOR): 1.26, 95% CI: 1.04–1.53), those with health insurance (AOR): 1.55, 95% CI: 1.30–1.85), those who had been visited by a field health worker (AOR: 1.31, 95% CI: 1.15–1.49), those with no big problems with distance to health facility (AOR): 1.25, 95% CI: 1.07–1.46), those who were married (AOR): 1.35, 95% CI: 1.11–1.64), those with tertiary level of education (AOR): 1.61, 95% CI: 1.05–2.49), those belonging to richer households (AOR): 1.33, 95% CI: 1.07–1.65) and those whose parity is less than 2 (AOR): 1.52, 95% CI: 1.18–1.95) were more likely to utilize complete continuum of care. Women belonging to the western region (AOR): 0.76, 95% CI: 0.61–0.94) were less likely to utilize complete continuum of care.\n\nTable 4Factors associated with CoC utilizationCharacteristicscrude modelcOR (95% CI)P-valueAdjusted modelaOR (95% CI)P-value\nAge\n35 to 491125 to 341.12 (0.98–1.27)0.0970.92 (0.79–1.08)0.30115 to 240.89 (0.59–1.36)0.6000.75 (0.48–1.17)0.203\nHousehold size\n6 and above11Less than 61.27 (1.12–1.43)< 0.0011.12 (0.96–1.29)0.150\nExposure to newspapers/magazines\nNo11Yes1.84 (1.59–2.13)< 0.0011.30 (1.11–1.52)0.001\nExposure to radio\nNo11Yes1.62 (1.39–1.89)< 0.0011.15 (0.96–1.38)0.137\nExposure to TV\nNo11Yes1.50 (1.32–1.71)< 0.0011.15 (0.99–1.34)0.075\nSex of Household head\nMale11Female0.84 (0.73–0.96)0.0110.99 (0.85–1.17)0.967\nInternet\nNo11Yes2.06 (1.70–2.49)< 0.0011.30 (0.98–1.73)0.66\nWealth Index\nPoorest11Poorer1.28 (1.06–1.55)0.0101.18 (0.97–1.44)0.101Middle1.52 (1.26–1.83)< 0.0011.34 (1.09–1.64)0.005Richer1.67 (1.39–2.02)< 0.0011.33 (1.07–1.65)0.009Richest2.01 (1.64–2.47)< 0.0011.21 (0.93–1.57)0.161\nParity\n5 and above112–41.57 (1.32–1.86)< 0.0011.41 (1.15–1.74)0.001Less than 21.66 (1.36–2.03)< 0.0011.52 (1.18–1.95)0.001\nMarried/cohabiting\nNo11Yes1.42 (1.22–1.65)< 0.0011.35 (1.11–1.64)0.002\nVisited by a fieldworker\nNo11Yes1.35 (1.19–1.52)< 0.0011.31 (1.15–1.49)< 0.001\nHealth insurance\nNo11Yes1.99 (1.69–2.34)< 0.0011.55 (1.30–1.85)< 0.001\nWorking\n\nNo\n1-\nYes\n0.93 (0.81–1.07)0.308\nPermission to access healthcare\nBig problem11Not big problem1.80 (1.29–2.52)0.0011.33 (0.92–1.91)0.129\nDistance to health facility\nBig problem11Not big problem1.43 (1.24–1.66)< 0.0011.25 (1.07–1.46)0.004\nRegion\nNorth11East1.22 (0.99–1.48)0.0521.24 (1.01–1.52)0.037West0.73 (0.59–0.91)0.0060.76 (0.61–0.94)0.013South1.25 (1.03–1.52)0.0231.26 (1.04–1.53)0.021Kigali1.09 (0.84–1.42)0.5000.85 (0.66–1.11)0.240\nResidence\nRural1-Urban1.09 (0.92–1.29)0.330\nEducation level\nNone11Primary1.27 (1.03–1.56)0.0240.95 (0.77–1.18)0.663Secondary1.58 (1.23–2.02)< 0.0010.92 (0.69–1.21)0.528Tertiary3.82 (2.75–5.30)< 0.0011.61 (1.05–2.49)0.031\nYear of Birth\n2014–20151120161.12 (0.90–1.40)0.3101.11 (0.88–1.41)0.37820170.95 (0.77–1.16)0.5930.95 (0.76–1.18)0.62420181.15 (0.93–1.42)0.2111.16 (0.92–145)0.20820191.19 (0.95–1.49)0.1251.11 (0.88–1.41)0.38320201.16 (0.83–1.61)0.3810.95 (0.67–1.33)0.759\n\nFactors associated with CoC utilization", "Factors associated with CoC utilisation are shown in Table 4. Women who had exposure to newspapers (adjusted odds ratio (AOR): 1.30, 95% CI: 1.11–1.52) those belonging to the eastern region (AOR): 1.24, 95% CI: 1.01–1.52), southern region (AOR): 1.26, 95% CI: 1.04–1.53), those with health insurance (AOR): 1.55, 95% CI: 1.30–1.85), those who had been visited by a field health worker (AOR: 1.31, 95% CI: 1.15–1.49), those with no big problems with distance to health facility (AOR): 1.25, 95% CI: 1.07–1.46), those who were married (AOR): 1.35, 95% CI: 1.11–1.64), those with tertiary level of education (AOR): 1.61, 95% CI: 1.05–2.49), those belonging to richer households (AOR): 1.33, 95% CI: 1.07–1.65) and those whose parity is less than 2 (AOR): 1.52, 95% CI: 1.18–1.95) were more likely to utilize complete continuum of care. Women belonging to the western region (AOR): 0.76, 95% CI: 0.61–0.94) were less likely to utilize complete continuum of care.\n\nTable 4Factors associated with CoC utilizationCharacteristicscrude modelcOR (95% CI)P-valueAdjusted modelaOR (95% CI)P-value\nAge\n35 to 491125 to 341.12 (0.98–1.27)0.0970.92 (0.79–1.08)0.30115 to 240.89 (0.59–1.36)0.6000.75 (0.48–1.17)0.203\nHousehold size\n6 and above11Less than 61.27 (1.12–1.43)< 0.0011.12 (0.96–1.29)0.150\nExposure to newspapers/magazines\nNo11Yes1.84 (1.59–2.13)< 0.0011.30 (1.11–1.52)0.001\nExposure to radio\nNo11Yes1.62 (1.39–1.89)< 0.0011.15 (0.96–1.38)0.137\nExposure to TV\nNo11Yes1.50 (1.32–1.71)< 0.0011.15 (0.99–1.34)0.075\nSex of Household head\nMale11Female0.84 (0.73–0.96)0.0110.99 (0.85–1.17)0.967\nInternet\nNo11Yes2.06 (1.70–2.49)< 0.0011.30 (0.98–1.73)0.66\nWealth Index\nPoorest11Poorer1.28 (1.06–1.55)0.0101.18 (0.97–1.44)0.101Middle1.52 (1.26–1.83)< 0.0011.34 (1.09–1.64)0.005Richer1.67 (1.39–2.02)< 0.0011.33 (1.07–1.65)0.009Richest2.01 (1.64–2.47)< 0.0011.21 (0.93–1.57)0.161\nParity\n5 and above112–41.57 (1.32–1.86)< 0.0011.41 (1.15–1.74)0.001Less than 21.66 (1.36–2.03)< 0.0011.52 (1.18–1.95)0.001\nMarried/cohabiting\nNo11Yes1.42 (1.22–1.65)< 0.0011.35 (1.11–1.64)0.002\nVisited by a fieldworker\nNo11Yes1.35 (1.19–1.52)< 0.0011.31 (1.15–1.49)< 0.001\nHealth insurance\nNo11Yes1.99 (1.69–2.34)< 0.0011.55 (1.30–1.85)< 0.001\nWorking\n\nNo\n1-\nYes\n0.93 (0.81–1.07)0.308\nPermission to access healthcare\nBig problem11Not big problem1.80 (1.29–2.52)0.0011.33 (0.92–1.91)0.129\nDistance to health facility\nBig problem11Not big problem1.43 (1.24–1.66)< 0.0011.25 (1.07–1.46)0.004\nRegion\nNorth11East1.22 (0.99–1.48)0.0521.24 (1.01–1.52)0.037West0.73 (0.59–0.91)0.0060.76 (0.61–0.94)0.013South1.25 (1.03–1.52)0.0231.26 (1.04–1.53)0.021Kigali1.09 (0.84–1.42)0.5000.85 (0.66–1.11)0.240\nResidence\nRural1-Urban1.09 (0.92–1.29)0.330\nEducation level\nNone11Primary1.27 (1.03–1.56)0.0240.95 (0.77–1.18)0.663Secondary1.58 (1.23–2.02)< 0.0010.92 (0.69–1.21)0.528Tertiary3.82 (2.75–5.30)< 0.0011.61 (1.05–2.49)0.031\nYear of Birth\n2014–20151120161.12 (0.90–1.40)0.3101.11 (0.88–1.41)0.37820170.95 (0.77–1.16)0.5930.95 (0.76–1.18)0.62420181.15 (0.93–1.42)0.2111.16 (0.92–145)0.20820191.19 (0.95–1.49)0.1251.11 (0.88–1.41)0.38320201.16 (0.83–1.61)0.3810.95 (0.67–1.33)0.759\n\nFactors associated with CoC utilization", "The current study reveals that only a third of Rwandan women received all elements of the continuum of care, and these women tended to be those from the Eastern region, those exposed to mass media (newspapers), those who had been visited by a field health worker, those who are from richer households, having tertiary level of education, married, low parity those with insurance and those with no big problems with distance to the nearest health facility.\nThe prevalence of complete continuum of care in this study is lower than findings from Cambodia, Egypt and Zambia [6, 31, 36] and higher than findings from studies conducted in Pakistan, Tanzania, Ethiopia, Ghana and Uganda [30, 37–40]. This low CoC finding is surprising because Rwanda is one of the countries in SSA with comprehensive national health insurance [41]. However, it is evident from this study that ANC utilization is low given that less than half (47.2%) of women are able to utilize at least four ANC contacts and this contributed to less completion of CoC.\nExposure to mass media (newspapers/magazines) showed a significant association with CoC utilization with the exposed women having higher odds of CoC. This finding is not surprising because several similar studies from other countries within in the region have reported this positive association between utilisation of mass media and the utilisation of MCH services [6, 7, 42, 43]. Gugsa et al. while examining newspaper coverage of maternal health in Bangladesh, Rwanda and South Africa showed that unlike South Africa, Rwanda was on track having the second highest maternal health coverage having increased from 15 articles to 158 in four years with 69% of the articles having a ‘human-rights’ or ‘policy-based’ frame [44].The influence of media on ensuring increased utilization of maternal health services including ANC, SBA and PNC could be linked to the role health information broadcasted on mass media play in ensuring reduction of maternal health knowledge shortage by sensitizing women and their partners on the benefits of timely access to care leading to positive attitudes and improves health seeking behavior [6, 45, 46]. In addition, women who are exposed to newspapers and television are more likely to be educated/literate and are in better position to have heath literacy related discussions with others which may further contribute to the challenging negative norms that might affect health seeking and hence lead to positive health seeking behavioral change [6, 47, 48].\nWomen who had been visited by a field health worker had higher odds of CoC utilization. Rwanda’s health system has a strong community health based programme [49]. Each village in Rwanda consists of three community health workers (CHWs) including one female Agent de Sante Maternelle (ASM) and a male–female pair (Binômes) [28]. Maternal care is provided by the ASMs through home and follow-up visits and referrals. CHWs are supported by the health ministry through the nearest health centres and are provided with a kit that has the needed supplies and a prepaid mobile phone to ease communication with the CHW cell coordinators [28]. In addition to the trainings supported by the government, some non-governmental organizations provides additional training and financial support to the CHWs [28].\nThese field based health workers are crucial in ensuring access to maternal care especially in areas with shortage of health facilities and large distances needed to be covered by women. The field health workers help equip mothers with knowledge on the dangers of using unskilled birth attendants and complications of pregnancies in addition to providing care, referring the complicated cases and also encouraging them to seek care within health facilities where possible [50]. Furthermore, these field health workers are being used to help track the progress of women along the continuum of care and follow-up defaulters [24]. Being visited by field health workers has been shown to be associated with maternal healthcare service utilization in several other studies [24, 51–53].\nWomen in the eastern region had higher odds of CoC utilization. This could be partly attributed to the high focus of national and international non-governmental organizations (NGOs) in the eastern region after it experienced some of the worst effects of the genocide that negatively affected the health status [14, 15]. These NGOs focused mainly on maternal and newborn care, worked closely with the ministry of health (MOH) and provided highly effective support that included; direct care provision, ensuring adequate and qualified staff, infrastructure development, robust quality improvement mechanisms and financial and technical assistance to support clinical innovation areas [14, 15]. Likewise, the smoother landscape found in eastern provinces compared to the northern region could be attributed to a higher likelihood of MCH service uptake because it enables for easier access to health facilities, as other studies have shown [54, 55]. The odds of having CoC in the western region were less. Despite the government’s initiatives to strengthen its healthcare system, the western region has lagged behind, with the majority of the population still lacking access to primary health care and more than half of the facilities operating under capacity [56]. Huerta et al. while assessing “geographical accessibility and spatial coverage modeling of the primary health care network in the Western Province” showed that when considering walking as the single mode of transportation, less than a third of the western province population is covered by the catchment area of the existing primary health facility network [56]. Fadelu et al. when assessing access to tertiary cancer care further showed that geographically, the most underserved region was the western one as none (0%) of the population were able to access care within an hour [57]. Hence there is high need for maternal stakeholders to target access to care in the western region.\nWomen who had no big problems with distance to the nearest health facility had higher odds of CoC utilization. Distance to health facilities has been found elsewhere in similar contexts to be an important determinant of maternal healthcare services utilization [11]. Given that most women ( 82.2% in this study) reside in rural areas which tend to have shortage of health facilities hence have to cover longer distances with poor road infrastructure [58]. This leads them to incur indirect transport costs or cover long distances and those that are unable to afford these transport costs or cover the long distances will end up attending less or no ANC contacts, have unskilled birth attendance and have no PNC [54, 58, 59]. Distance doesn’t only affect women but also community health workers who provide care in the community [54]. Thus, Rwanda’s maternal healthcare stakeholders need to explore sustainable solutions to health facility access barriers such as maternity waiting homes and construction of more health facilities.\nWomen with health insurance had higher odds of CoC utilization a finding that has been shown in other studies [60–63]. Rwanda’s community-based health insurance (CBHI) program is locally coordinated at the district level with enrollment being promoted by the village mobilization committees at the village level [61, 64, 65]. The program is funded by donor and development partners, the government, and individual contributions with prenatal and postnatal care being covered in the minimum package [29, 61]. Pregnant women incur several hidden expenses which may arise from the long distances covered when accessing care, buying medicines and supplies when they are out of stock in public facilities leading to high out-of-pocket (OOP) expenditures which tend to have a negative effect on access to maternal health services [63, 66]. Health insurance coverage reduces the rates of out-of-pocket payments associated with the use of MHC services utilization hence can improve health seeking and utilization behaviors [60, 63].\nWomen belonging to richer households had more odds of having CoC compared to those from the poorest households a finding consistent with several SSA studies [43, 67, 68]. Women belonging to households with limited financial resources may be unable to afford the medical and non-medical costs involved in seeking maternal care [43, 69, 70]. This might lead to those women not seeking maternal care, seeking late or limiting the number of ANC contacts which negatively affects completion of CoC [43]. Although Rwanda has a very good national health insurance scheme, not all women have subscribed to it as evidenced by over 18% of women in this analysis having no insurance hence some women still pay out of pocket for direct maternal healthcare costs and even those that have insurance, many still have to pay for the indirect costs such as transportation. This implies that lower levels of wealth can also be a barrier to accessing maternal health services hence multi-sectoral actors and different attentions are needed to eliminate financial barriers to improve the continuity of maternity care in Rwanda.\nConsistent with findings from Ethiopia, Ghana and Uganda, increased levels of education have been shown to be associated with higher odds of completing CoC [40, 71, 72]. Educated women are more likely to have better maternal health literacy, more likely to have better paying jobs hence increased ability to afford direct and indirect costs associated with accessing maternal care [73]. Furthermore, highly educated women have been shown to be more receptive to new maternal health related information, increased knowledge of availability and location of maternal health resources and increased decision-making abilities which factors are crucial to ensure positive health seeking behaviour [71, 74].\nMarried women had higher odds of having CoC compared to their non-married counterparts. Marital status’s positive association with utilization and outcomes of maternal healthcare has been documented in several countries [43, 75–78]. Marriage provides better planning/desirability of care, societal acceptability, more psychosocial and financial support to women from their partners and in laws which support provides women with more time to access care and ability to afford the direct and indirect costs involved in accessing maternal care [43, 75, 79, 80]. Furthermore, the psychosocial support from partners, partner’s friends and in laws means understanding, sympathy and communication, which can positively motivate the women to seek timely and adequate maternal care [80].\nWomen with less parity had higher odds of having CoC compared to those whose parity was five and above. Increased utilization of maternal healthcare services among women with less parity has been shown in several other SSA countries [81–84]. This could be partly attributed to the fact that women with more children believe they are experienced with pregnancy and childbirth hence they are less anxious about the possibility of negative outcomes hence utilize maternal healthcare services late and less compared to those with lower parity [43, 81–83].", "Only 3 out of 10 women were able to utilize the entire continuum of maternity care services. Regarding ANC utilization, less than half (47.2%) and only 0.3% of women were able to utilize at least four and eight ANC contacts respectively a finding showing that Rwanda is not on track towards achieving the recommended eight ANC contacts for each pregnant woman in the 2016 WHO ANC guidelines. Belonging to eastern and southern Rwanda, exposure to mass media, having been visited by a field health worker, having health insurance, having no big problems with distance to the nearest health facility, belonging to richer households, having tertiary level of education, being married and having low parity were positively associated with completion of maternal and newborn continuum of care. Thus, we recommend the different stakeholders engaged in maternal health care to focus on women from western region, those without insurance, from poor households, with low levels of education, high parity and unmarried to ensure utilization of complete continuum of maternal and newborn care. Promoting maternity services through mass media, strengthening the community health programmes, increasing access to health insurance, strengthening and promoting girl child education to post-secondary level, increased support to household economic development programmes may improve the level of utilization of maternity services. Utilization of mass media specially to sensitize the population on the benefits of continuum of care should be encouraged with great attention given for early initiation of ANC, increasing the number of ANC contacts, health facility and skilled birth attendance during childbirth and postnatal care. During ANC contacts in health facilities and in the community by CHWs unmarried and high parity women need to be given more time during care, awareness on the importance of having CoC and followed up actively.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\n\nSupplementary Material 1\n\n\n\nSupplementary Material 1\n\nBelow is the link to the electronic supplementary material.\n\n\nSupplementary Material 1\n\n\n\nSupplementary Material 1\n", "Below is the link to the electronic supplementary material.\n\n\nSupplementary Material 1\n\n\n\nSupplementary Material 1\n" ]
[ null, null, null, null, null, null, null, null, "results", null, "discussion", "conclusion", "supplementary-material", null ]
[ "Continuum of care", "Antenatal care", "Postnatal care", "Skilled birth attendance", "Women and Rwanda" ]
Implementation barriers and enablers of midwifery group practice for vulnerable women: a qualitative study in a tertiary urban Australian health service.
36261823
Maternity services have limited formalised guidance on planning new services such as midwifery group practice for vulnerable women, for example women with a history of substance abuse (alcohol, tobacco and other drugs), mental health challenges, complex social issues or other vulnerability. Continuity of care through midwifery group practice is mostly restricted to women with low-risk pregnancies and is not universally available to vulnerable women, despite evidence supporting benefits of this model of care for all women. The perception that midwifery group practice for vulnerable women is a high-risk model of care lacking in evidence may have in the past, thwarted implementation planning studies that seek to improve care for these women. We therefore aimed to identify the barriers and enablers that might impact the implementation of a midwifery group practice for vulnerable women.
BACKGROUND
A qualitative context analysis using the Consolidated Framework for Implementation Research was conducted at a single-site tertiary health facility in Queensland, Australia. An interdisciplinary group of stakeholders from a purposeful sample of 31 people participated in semi-structured interviews. Data were analysed using manual and then Leximancer computer assisted methods. Themes were compared and mapped to the Framework.
METHODS
Themes identified were the woman's experience, midwifery workforce capabilities, identifying "gold standard care", the interdisciplinary team and costs. Potential enablers of implementation included perceptions that the model facilitates a relationship of trust with vulnerable women, that clinical benefit outweighs cost and universal stakeholder acceptance. Potential barriers were: potential isolation of the interdisciplinary team, costs and the potential for vicarious trauma for midwives.
RESULTS
There was recognition that the proposed model of care is supported by research and a view that clinical benefits will outweigh costs, however supervision and support is required for midwives to manage and limit vicarious trauma. An interdisciplinary team structure is also an essential component of the service design. Attention to these key themes, barriers and enablers will assist with identification of strategies to aid successful implementation. Australian maternity services can use our results to compare how the perceptions of local stakeholders might be similar or different to the results presented in this paper.
CONCLUSION
[ "Female", "Pregnancy", "Humans", "Midwifery", "Maternal Health Services", "Compassion Fatigue", "Australia", "Group Practice" ]
9583548
null
null
null
null
Results
Of the 40 people invited to an interview, 31 consented to participate (77.5%) with no response received from 9 others. No additional people contacted the study team to offer their involvement. Twenty individual or small group interviews were conducted. Data saturation was achieved following 10 interviews however, the research team elected to continue data collection due to many stakeholders wishing to participate. Most participants were female (87%) and between 41 and 50 years of age (35.5%). They were predominantly permanent employees (74.2%) and had more than 10 years’ experience (70.9%) (Table 2). Participant characteristics generally aligned with that of the Australian health workforce, with most participants being nurses and midwives (Australian Institute of Health and Welfare, 2020). However, more people aged 41 and over participated in the research, whereas the comparable Australian workforce is mostly aged 20 to 34 years. Table 2Characteristics of participants [N = 31]Participant characteristicn (%)Age20–3031–4041–5051–6061+2 (6.5)6 (19.4)11 (35.5)9 (29)3 (9.6)GenderMaleFemaleNot stated3 (9.7)27 (87)1 (3.2)Stakeholder roleMidwifeMedical OfficerNurseNurse/Midwife LeaderAllied Health PractitionerOther (Business Representative, Administration Officer, Consumer)13 (41.9)5 (16.1)6 (19.4)3 (9.7)1 (3.2)3 (9.7)Work hoursFull timePart timeNot stated15 (48.4)13 (41.9)3 (9.7)Employment statusTemporary appointmentPermanent appointmentNot stated1 (3.2)23 (74.2)7 (22.6)Professional experience*< 5 years5–10 years10 + yearsNot stated07 (22.6)22 (70.9)1 (6.5)*The consumer representative was not included in this group, thus n = 30 Characteristics of participants [N = 31] Age 20–30 31–40 41–50 51–60 61+ 2 (6.5) 6 (19.4) 11 (35.5) 9 (29) 3 (9.6) Gender Male Female Not stated 3 (9.7) 27 (87) 1 (3.2) Stakeholder role Midwife Medical Officer Nurse Nurse/Midwife Leader Allied Health Practitioner Other (Business Representative, Administration Officer, Consumer) 13 (41.9) 5 (16.1) 6 (19.4) 3 (9.7) 1 (3.2) 3 (9.7) Work hours Full time Part time Not stated 15 (48.4) 13 (41.9) 3 (9.7) Employment status Temporary appointment Permanent appointment Not stated 1 (3.2) 23 (74.2) 7 (22.6) Professional experience* < 5 years 5–10 years 10 + years Not stated 0 7 (22.6) 22 (70.9) 1 (6.5) *The consumer representative was not included in this group, thus n = 30 Potential barriers and enablers were identified from the interview data and grouped into themes: the woman’s experience, midwifery workforce, gold standard care, the interdisciplinary team, and costs. These themes function as ways in which we have organised and expressed the barriers and enablers. A summary of potential barriers and enablers from which these themes emerged is presented in Supplementary File 1. [SUBTITLE] The woman’s experience [SUBSECTION] Discussion around the woman’s experience identified enablers for the proposed model. Many participants expressed empathy for the likely patient cohort and placed the proposed model as a high priority for the health service. Enablers included perceptions of improved clinical outcomes, building of safety and trust, patient satisfaction and the potential to reduce ‘fail to attend’ rates at the specialty clinic. This is supported by comments from participants: … vulnerable or disadvantaged groups would benefit … (Other role, Interview 1). … some with abuse histories don’t want to go over those histories over and over again…some come with diverse cultural situations and specific needs … (Midwife, Interview 2).… they can build up trust, they can have the tough talk with a familiar face … (Nurse, Interview 12). … vulnerable or disadvantaged groups would benefit … (Other role, Interview 1). … some with abuse histories don’t want to go over those histories over and over again…some come with diverse cultural situations and specific needs … (Midwife, Interview 2). … they can build up trust, they can have the tough talk with a familiar face … (Nurse, Interview 12). The general sentiment expressed by many participants is captured by this statement: …the patient can build a rapport and have trust with the clinicians…. It not only makes the patient feel safe and more comfortable with their surroundings, in my opinion, it makes the patient a bit more accountable and builds rapport with that clinician and women would be more engaged to come back… (Other role, Interview 1). …the patient can build a rapport and have trust with the clinicians…. It not only makes the patient feel safe and more comfortable with their surroundings, in my opinion, it makes the patient a bit more accountable and builds rapport with that clinician and women would be more engaged to come back… (Other role, Interview 1). The proposed model was being viewed as positive and with a woman-centred focus. These views become potential enablers when preparing detailed business cases that demonstrate strategic and strong stakeholder support as well as alignment with evidence and policy advocating woman-centred care. However, potential barriers centred around concerns that women may disengage if they did not bond with the known midwife, were socially isolated or feared being reported to child safety services: For the women it’s positive all around, unless they felt they couldn’t engage with the midwife, which could lead to the women disengaging completely. (Midwife, Interview 6)The women can be fearful about opening up…that there may be negative consequences… (Midwife, Interview 10). For the women it’s positive all around, unless they felt they couldn’t engage with the midwife, which could lead to the women disengaging completely. (Midwife, Interview 6) The women can be fearful about opening up…that there may be negative consequences… (Midwife, Interview 10). Participants also suggested ways to address the woman’s disengagement through awareness and actions by the midwives: We have to be open and transparent…so there’s a consistent approach and that we can be strong and recognise it when it (disengagement) happens and maintain the relationship … (Midwife, Interview 10).… the midwife would need to be capable of referring… to those that can help … (Midwife, Interview 16).… if that relationship isn’t working there could be space to swap … (Other role, Interview 17). We have to be open and transparent…so there’s a consistent approach and that we can be strong and recognise it when it (disengagement) happens and maintain the relationship … (Midwife, Interview 10). … the midwife would need to be capable of referring… to those that can help … (Midwife, Interview 16). … if that relationship isn’t working there could be space to swap … (Other role, Interview 17). While women’s disengagement from the proposed model might be identified as a risk, in discussions around this barrier solutions were identified. Such solutions would inform risk mitigation. Discussion around the woman’s experience identified enablers for the proposed model. Many participants expressed empathy for the likely patient cohort and placed the proposed model as a high priority for the health service. Enablers included perceptions of improved clinical outcomes, building of safety and trust, patient satisfaction and the potential to reduce ‘fail to attend’ rates at the specialty clinic. This is supported by comments from participants: … vulnerable or disadvantaged groups would benefit … (Other role, Interview 1). … some with abuse histories don’t want to go over those histories over and over again…some come with diverse cultural situations and specific needs … (Midwife, Interview 2).… they can build up trust, they can have the tough talk with a familiar face … (Nurse, Interview 12). … vulnerable or disadvantaged groups would benefit … (Other role, Interview 1). … some with abuse histories don’t want to go over those histories over and over again…some come with diverse cultural situations and specific needs … (Midwife, Interview 2). … they can build up trust, they can have the tough talk with a familiar face … (Nurse, Interview 12). The general sentiment expressed by many participants is captured by this statement: …the patient can build a rapport and have trust with the clinicians…. It not only makes the patient feel safe and more comfortable with their surroundings, in my opinion, it makes the patient a bit more accountable and builds rapport with that clinician and women would be more engaged to come back… (Other role, Interview 1). …the patient can build a rapport and have trust with the clinicians…. It not only makes the patient feel safe and more comfortable with their surroundings, in my opinion, it makes the patient a bit more accountable and builds rapport with that clinician and women would be more engaged to come back… (Other role, Interview 1). The proposed model was being viewed as positive and with a woman-centred focus. These views become potential enablers when preparing detailed business cases that demonstrate strategic and strong stakeholder support as well as alignment with evidence and policy advocating woman-centred care. However, potential barriers centred around concerns that women may disengage if they did not bond with the known midwife, were socially isolated or feared being reported to child safety services: For the women it’s positive all around, unless they felt they couldn’t engage with the midwife, which could lead to the women disengaging completely. (Midwife, Interview 6)The women can be fearful about opening up…that there may be negative consequences… (Midwife, Interview 10). For the women it’s positive all around, unless they felt they couldn’t engage with the midwife, which could lead to the women disengaging completely. (Midwife, Interview 6) The women can be fearful about opening up…that there may be negative consequences… (Midwife, Interview 10). Participants also suggested ways to address the woman’s disengagement through awareness and actions by the midwives: We have to be open and transparent…so there’s a consistent approach and that we can be strong and recognise it when it (disengagement) happens and maintain the relationship … (Midwife, Interview 10).… the midwife would need to be capable of referring… to those that can help … (Midwife, Interview 16).… if that relationship isn’t working there could be space to swap … (Other role, Interview 17). We have to be open and transparent…so there’s a consistent approach and that we can be strong and recognise it when it (disengagement) happens and maintain the relationship … (Midwife, Interview 10). … the midwife would need to be capable of referring… to those that can help … (Midwife, Interview 16). … if that relationship isn’t working there could be space to swap … (Other role, Interview 17). While women’s disengagement from the proposed model might be identified as a risk, in discussions around this barrier solutions were identified. Such solutions would inform risk mitigation. [SUBTITLE] Midwifery workforce [SUBSECTION] Participants saw the proposed new model as an opportunity for midwives to gain new skills and expand their scope of practice which was identified as an enabler: There are a few midwives out there currently upskilling themselves and are really passionate and interested and already preparing for being part of the team … (Nurse/Midwife Leader, Interview 3).Midwives will actually see this as a positive move and it’s going to be development for them, and it’s going to be opportunistic for them … (Nurse/Midwife Leader, Interview 4). There are a few midwives out there currently upskilling themselves and are really passionate and interested and already preparing for being part of the team … (Nurse/Midwife Leader, Interview 3). Midwives will actually see this as a positive move and it’s going to be development for them, and it’s going to be opportunistic for them … (Nurse/Midwife Leader, Interview 4). A major workforce barrier to implementation success was how midwifery group practice would place high psychological demand on the midwives leading to burnout or vicarious trauma: … very complex women with personality disorders and high psychological needs and that could be quite demanding of one midwife as the primary caregiver … (Nurse/Midwife Leader, Interview 3).… to field all those phone calls and constantly support that person would be really challenging … (Nurse/Midwife Leader, Interview 3).For the midwife dealing with only these women, it could over time be mentally challenging … potentially exhausting and tiring … (Midwife, Interview 6). … very complex women with personality disorders and high psychological needs and that could be quite demanding of one midwife as the primary caregiver … (Nurse/Midwife Leader, Interview 3). … to field all those phone calls and constantly support that person would be really challenging … (Nurse/Midwife Leader, Interview 3). For the midwife dealing with only these women, it could over time be mentally challenging … potentially exhausting and tiring … (Midwife, Interview 6). The difficulty in attracting midwives to the proposed model of care was an identified barrier to implementing a high standard of care: … attracting the midwives that would have an interest in it, we almost need a mother-like figure…would have to be resilient and have had a few more life experiences … (Medical Officer, Interview 13). … attracting the midwives that would have an interest in it, we almost need a mother-like figure…would have to be resilient and have had a few more life experiences … (Medical Officer, Interview 13). While this second barrier is contradictory to the initial workforce enabler identified in this theme, participants became solution focussed in the interviews, which is reflected in the theme reported below ‘The interdisciplinary team’. Participants saw the proposed new model as an opportunity for midwives to gain new skills and expand their scope of practice which was identified as an enabler: There are a few midwives out there currently upskilling themselves and are really passionate and interested and already preparing for being part of the team … (Nurse/Midwife Leader, Interview 3).Midwives will actually see this as a positive move and it’s going to be development for them, and it’s going to be opportunistic for them … (Nurse/Midwife Leader, Interview 4). There are a few midwives out there currently upskilling themselves and are really passionate and interested and already preparing for being part of the team … (Nurse/Midwife Leader, Interview 3). Midwives will actually see this as a positive move and it’s going to be development for them, and it’s going to be opportunistic for them … (Nurse/Midwife Leader, Interview 4). A major workforce barrier to implementation success was how midwifery group practice would place high psychological demand on the midwives leading to burnout or vicarious trauma: … very complex women with personality disorders and high psychological needs and that could be quite demanding of one midwife as the primary caregiver … (Nurse/Midwife Leader, Interview 3).… to field all those phone calls and constantly support that person would be really challenging … (Nurse/Midwife Leader, Interview 3).For the midwife dealing with only these women, it could over time be mentally challenging … potentially exhausting and tiring … (Midwife, Interview 6). … very complex women with personality disorders and high psychological needs and that could be quite demanding of one midwife as the primary caregiver … (Nurse/Midwife Leader, Interview 3). … to field all those phone calls and constantly support that person would be really challenging … (Nurse/Midwife Leader, Interview 3). For the midwife dealing with only these women, it could over time be mentally challenging … potentially exhausting and tiring … (Midwife, Interview 6). The difficulty in attracting midwives to the proposed model of care was an identified barrier to implementing a high standard of care: … attracting the midwives that would have an interest in it, we almost need a mother-like figure…would have to be resilient and have had a few more life experiences … (Medical Officer, Interview 13). … attracting the midwives that would have an interest in it, we almost need a mother-like figure…would have to be resilient and have had a few more life experiences … (Medical Officer, Interview 13). While this second barrier is contradictory to the initial workforce enabler identified in this theme, participants became solution focussed in the interviews, which is reflected in the theme reported below ‘The interdisciplinary team’. [SUBTITLE] Gold standard care [SUBSECTION] The belief amongst participants of strong, quality evidence in favour of the proposed model was identified as an enabler, and there were no barriers highlighted by participants in terms of available evidence. Many participants identified through discussion around evidence that a midwifery group practice for vulnerable women would be a positive step for the women involved. This was particularly evident for participants who had undertaken reading prior to the interview with participants expressing: I have looked online to have a look at the research… shows the best outcome for babies and mothers across the board in terms of continuity of care models … (Other role, Interview 1).MGP is gold standard and… there’s lots of research out there that shows that continuity of care is best for these women to develop a relationship … (Midwife, Interview 2).… the people caring for them are more likely to pick up on deviations from a normal emotional state … (Midwife, Interview 2). I have looked online to have a look at the research… shows the best outcome for babies and mothers across the board in terms of continuity of care models … (Other role, Interview 1). MGP is gold standard and… there’s lots of research out there that shows that continuity of care is best for these women to develop a relationship … (Midwife, Interview 2). … the people caring for them are more likely to pick up on deviations from a normal emotional state … (Midwife, Interview 2). Some participants had undertaken self-directed reading to source additional information regarding midwifery group practice and the needs of vulnerable women. Whilst participants were sent relevant information containing brief background to the proposed study in advance of the interview, some attendees advised they wanted to come well prepared. For example, two participants advised the researchers that they were very grateful to have been invited to interview and were now more aware of the benefits of midwifery group practice for women including that this care was “gold standard”. The integral way in which having a known midwife provides benefit and support for women was repeatedly discussed by participants. For many, the perception from the evidence that a midwifery group practice for vulnerable women is “gold standard” was the sole driver for supporting the proposed model and was seen as a strong enabler which would enhance a business case for the proposed model. The belief amongst participants of strong, quality evidence in favour of the proposed model was identified as an enabler, and there were no barriers highlighted by participants in terms of available evidence. Many participants identified through discussion around evidence that a midwifery group practice for vulnerable women would be a positive step for the women involved. This was particularly evident for participants who had undertaken reading prior to the interview with participants expressing: I have looked online to have a look at the research… shows the best outcome for babies and mothers across the board in terms of continuity of care models … (Other role, Interview 1).MGP is gold standard and… there’s lots of research out there that shows that continuity of care is best for these women to develop a relationship … (Midwife, Interview 2).… the people caring for them are more likely to pick up on deviations from a normal emotional state … (Midwife, Interview 2). I have looked online to have a look at the research… shows the best outcome for babies and mothers across the board in terms of continuity of care models … (Other role, Interview 1). MGP is gold standard and… there’s lots of research out there that shows that continuity of care is best for these women to develop a relationship … (Midwife, Interview 2). … the people caring for them are more likely to pick up on deviations from a normal emotional state … (Midwife, Interview 2). Some participants had undertaken self-directed reading to source additional information regarding midwifery group practice and the needs of vulnerable women. Whilst participants were sent relevant information containing brief background to the proposed study in advance of the interview, some attendees advised they wanted to come well prepared. For example, two participants advised the researchers that they were very grateful to have been invited to interview and were now more aware of the benefits of midwifery group practice for women including that this care was “gold standard”. The integral way in which having a known midwife provides benefit and support for women was repeatedly discussed by participants. For many, the perception from the evidence that a midwifery group practice for vulnerable women is “gold standard” was the sole driver for supporting the proposed model and was seen as a strong enabler which would enhance a business case for the proposed model. [SUBTITLE] The interdisciplinary team [SUBSECTION] Support for the inclusion of an interdisciplinary team in the proposed model of care was identified as an enabler and was particularly supported by non-midwife participants: … I would love it, because we felt [from observation of previous staffing arrangements] that when the midwives were staying in the role longer it felt very organised and that we knew the patients very well … (Allied Health Practitioner, Interview 9).There’s also the opportunity to develop an interdisciplinary trust … (Nurse, Interview 14). … I would love it, because we felt [from observation of previous staffing arrangements] that when the midwives were staying in the role longer it felt very organised and that we knew the patients very well … (Allied Health Practitioner, Interview 9). There’s also the opportunity to develop an interdisciplinary trust … (Nurse, Interview 14). Midwife participants felt similarly: Need a multi-disciplinary team … and.“… still having that multi-disciplinary approach… is fantastic … (Midwife, Interview 11). Need a multi-disciplinary team … and. “… still having that multi-disciplinary approach… is fantastic … (Midwife, Interview 11). Other responses emphasised the importance of midwives working with other disciplines and not practising in isolation: It’s really beneficial having a whole team caring for them (Midwife, Interview 2) …and.… continuing to have the multi-disciplinary input and multi-disciplinary team meeting is going to be beneficial … (Medical Officer, Interview 20). It’s really beneficial having a whole team caring for them (Midwife, Interview 2) …and. … continuing to have the multi-disciplinary input and multi-disciplinary team meeting is going to be beneficial … (Medical Officer, Interview 20). Others commented: … it’s an addition to the multi-disciplinary teams, so it doesn’t take anything away but they’ve got some-one they can trust that follows them through all the way … (Medical Officer, Interview 8).… the midwives … would need to be engaged with the multi-disciplinary team more than … MGP’s … (Other role, Interview 17). … it’s an addition to the multi-disciplinary teams, so it doesn’t take anything away but they’ve got some-one they can trust that follows them through all the way … (Medical Officer, Interview 8). … the midwives … would need to be engaged with the multi-disciplinary team more than … MGP’s … (Other role, Interview 17). The overall sentiment across disciplines was that: Everyone’s ready for a change in the space and a growth in the space and how we can improve for the women and I think it would be highly supported, valued and everyone would be on board … (Nurse/Midwife Leader, Interview 3). Everyone’s ready for a change in the space and a growth in the space and how we can improve for the women and I think it would be highly supported, valued and everyone would be on board … (Nurse/Midwife Leader, Interview 3). An interdisciplinary team was therefore an important and well supported component of the model, and no barriers to including an inter-disciplinary team in the model were identified. Participants were clear that midwives would lead continuity of care, while having expert health professionals involved to provide comprehensive care for the women. They would also provide professional support for midwives who might feel isolated in their role. These design details can be included in a business case to ensure successful and sustained implementation. Support for the inclusion of an interdisciplinary team in the proposed model of care was identified as an enabler and was particularly supported by non-midwife participants: … I would love it, because we felt [from observation of previous staffing arrangements] that when the midwives were staying in the role longer it felt very organised and that we knew the patients very well … (Allied Health Practitioner, Interview 9).There’s also the opportunity to develop an interdisciplinary trust … (Nurse, Interview 14). … I would love it, because we felt [from observation of previous staffing arrangements] that when the midwives were staying in the role longer it felt very organised and that we knew the patients very well … (Allied Health Practitioner, Interview 9). There’s also the opportunity to develop an interdisciplinary trust … (Nurse, Interview 14). Midwife participants felt similarly: Need a multi-disciplinary team … and.“… still having that multi-disciplinary approach… is fantastic … (Midwife, Interview 11). Need a multi-disciplinary team … and. “… still having that multi-disciplinary approach… is fantastic … (Midwife, Interview 11). Other responses emphasised the importance of midwives working with other disciplines and not practising in isolation: It’s really beneficial having a whole team caring for them (Midwife, Interview 2) …and.… continuing to have the multi-disciplinary input and multi-disciplinary team meeting is going to be beneficial … (Medical Officer, Interview 20). It’s really beneficial having a whole team caring for them (Midwife, Interview 2) …and. … continuing to have the multi-disciplinary input and multi-disciplinary team meeting is going to be beneficial … (Medical Officer, Interview 20). Others commented: … it’s an addition to the multi-disciplinary teams, so it doesn’t take anything away but they’ve got some-one they can trust that follows them through all the way … (Medical Officer, Interview 8).… the midwives … would need to be engaged with the multi-disciplinary team more than … MGP’s … (Other role, Interview 17). … it’s an addition to the multi-disciplinary teams, so it doesn’t take anything away but they’ve got some-one they can trust that follows them through all the way … (Medical Officer, Interview 8). … the midwives … would need to be engaged with the multi-disciplinary team more than … MGP’s … (Other role, Interview 17). The overall sentiment across disciplines was that: Everyone’s ready for a change in the space and a growth in the space and how we can improve for the women and I think it would be highly supported, valued and everyone would be on board … (Nurse/Midwife Leader, Interview 3). Everyone’s ready for a change in the space and a growth in the space and how we can improve for the women and I think it would be highly supported, valued and everyone would be on board … (Nurse/Midwife Leader, Interview 3). An interdisciplinary team was therefore an important and well supported component of the model, and no barriers to including an inter-disciplinary team in the model were identified. Participants were clear that midwives would lead continuity of care, while having expert health professionals involved to provide comprehensive care for the women. They would also provide professional support for midwives who might feel isolated in their role. These design details can be included in a business case to ensure successful and sustained implementation. [SUBTITLE] Costs [SUBSECTION] Participants believed that the health benefits of the proposed model of care would outweigh the perception that a midwifery group practice for vulnerable women was a more expensive model of care. This belief was a clear enabler of the proposed model: … the cost would come with great reward … (Nurse/Midwife Leader, Interview 3).… there will be extra cost, but the trade off in terms of good follow up might not save money but it will be money well spent … (Medical Officer, Interview 8). … the cost would come with great reward … (Nurse/Midwife Leader, Interview 3). … there will be extra cost, but the trade off in terms of good follow up might not save money but it will be money well spent … (Medical Officer, Interview 8). However, participants did express concern at the perceived increased need for resources and challenges with attributing costs to a range of clinical areas. The concern focussed around the potential impact on other teams which was seen as a barrier: I think there is resistance…they see this is going to be taking away from their skill mix and FTE (Full Time Equivalent) … (Nurse/Midwife Leader, Interview 4). I think there is resistance…they see this is going to be taking away from their skill mix and FTE (Full Time Equivalent) … (Nurse/Midwife Leader, Interview 4). However, other participants believed that these perceptions and challenges could be overcome and that the proposed model of care should be a priority for the hospital: … yes there are financial implications and barriers, there doesn’t seem to be good evidence to show why not … (Other role, Interview 1).… yes, it is very important that we generate the activity to get something for the work that we’re doing, but at the end of the day we are looking at patient centred care, so if it’s easier and the best outcome for mother and baby… then that’s what we have to do … (Other role, Interview 1).Why shouldn’t they have an MGP, they shouldn’t be excluded just because they’ve had drug and alcohol or mental health issues in the past … (Nurse, Interview 15).I see it as high priority to look at how we can increase activity and treat this vulnerable group … (Nurse/Midwife Leader, Interview 4). … yes there are financial implications and barriers, there doesn’t seem to be good evidence to show why not … (Other role, Interview 1). … yes, it is very important that we generate the activity to get something for the work that we’re doing, but at the end of the day we are looking at patient centred care, so if it’s easier and the best outcome for mother and baby… then that’s what we have to do … (Other role, Interview 1). Why shouldn’t they have an MGP, they shouldn’t be excluded just because they’ve had drug and alcohol or mental health issues in the past … (Nurse, Interview 15). I see it as high priority to look at how we can increase activity and treat this vulnerable group … (Nurse/Midwife Leader, Interview 4). Perceptions around cost that are both potential enablers and barriers to gaining support and successful implementation would need to be clarified as fact in a business case before the proposed model is implemented. Participants had concerns around costs which may be reflected by decision-makers when examining the rigor in which a business case has been prepared. This may also influence future expansion of midwifery group practice as a model of care more generally. In the computer-assisted analysis, five themes were identified from the data: women with complex care: specialty clinic; continuity of care; workforce; and opportunity (Fig. 1). Themes and sub-themes similar to those that emerged from manual analysis were around the woman’s experience, workforce, standards of care and opportunity. Fig. 1Leximancer 4™ generated themes from stakeholder interviews Leximancer 4™ generated themes from stakeholder interviews Table 3 shows the mapped overarching themes and how they relate to the five CFIR domains and constructs. In this table it becomes clear what the requirements for successful and sustained implementation of the proposed model might be. The theme Gold standard care mapped to five constructs indicates that either a perception, or knowledge of evidence supporting midwifery group practice for vulnerable women would be critical to its success. A thorough consideration of potential experiences for the woman, the workforce and costs when preparing a business case, will be a determinant of model implementation success. An interdisciplinary team that is already part of the organisational structure and engaged in planning the model is likely to be essential. Processes that demonstrate evidence of planning and reflecting across all CFIR domains, especially regarding linkages between different health professional disciplines and costs, are also important. Table 3Summary of interview themes mapped to Consolidated Framework for Implementation Research (CFIR) domains and constructsCFIR domains and constructsThemes Intervention characteristics Evidence strength and qualityGold standard careRelative advantageThe woman’s experienceMidwifery workforceCostsCostCostsAdaptabilityMidwifery workforce Outer setting Patient needs and resourcesThe woman’s experiencePeer PressureGold standard care Inner setting Structural characteristicsThe interdisciplinary teamImplementation climateMidwifery workforceRelative prioritiesGold standard careThe woman’s experience Characteristics of individuals Knowledge and beliefs about the interventionGold standard careIndividual stage of changeGold standard careSelf-efficacyMidwifery workforce Process PlanningThe interdisciplinary teamReflecting and evaluatingCosts Summary of interview themes mapped to Consolidated Framework for Implementation Research (CFIR) domains and constructs The woman’s experience Midwifery workforce Costs Gold standard care The woman’s experience Participants believed that the health benefits of the proposed model of care would outweigh the perception that a midwifery group practice for vulnerable women was a more expensive model of care. This belief was a clear enabler of the proposed model: … the cost would come with great reward … (Nurse/Midwife Leader, Interview 3).… there will be extra cost, but the trade off in terms of good follow up might not save money but it will be money well spent … (Medical Officer, Interview 8). … the cost would come with great reward … (Nurse/Midwife Leader, Interview 3). … there will be extra cost, but the trade off in terms of good follow up might not save money but it will be money well spent … (Medical Officer, Interview 8). However, participants did express concern at the perceived increased need for resources and challenges with attributing costs to a range of clinical areas. The concern focussed around the potential impact on other teams which was seen as a barrier: I think there is resistance…they see this is going to be taking away from their skill mix and FTE (Full Time Equivalent) … (Nurse/Midwife Leader, Interview 4). I think there is resistance…they see this is going to be taking away from their skill mix and FTE (Full Time Equivalent) … (Nurse/Midwife Leader, Interview 4). However, other participants believed that these perceptions and challenges could be overcome and that the proposed model of care should be a priority for the hospital: … yes there are financial implications and barriers, there doesn’t seem to be good evidence to show why not … (Other role, Interview 1).… yes, it is very important that we generate the activity to get something for the work that we’re doing, but at the end of the day we are looking at patient centred care, so if it’s easier and the best outcome for mother and baby… then that’s what we have to do … (Other role, Interview 1).Why shouldn’t they have an MGP, they shouldn’t be excluded just because they’ve had drug and alcohol or mental health issues in the past … (Nurse, Interview 15).I see it as high priority to look at how we can increase activity and treat this vulnerable group … (Nurse/Midwife Leader, Interview 4). … yes there are financial implications and barriers, there doesn’t seem to be good evidence to show why not … (Other role, Interview 1). … yes, it is very important that we generate the activity to get something for the work that we’re doing, but at the end of the day we are looking at patient centred care, so if it’s easier and the best outcome for mother and baby… then that’s what we have to do … (Other role, Interview 1). Why shouldn’t they have an MGP, they shouldn’t be excluded just because they’ve had drug and alcohol or mental health issues in the past … (Nurse, Interview 15). I see it as high priority to look at how we can increase activity and treat this vulnerable group … (Nurse/Midwife Leader, Interview 4). Perceptions around cost that are both potential enablers and barriers to gaining support and successful implementation would need to be clarified as fact in a business case before the proposed model is implemented. Participants had concerns around costs which may be reflected by decision-makers when examining the rigor in which a business case has been prepared. This may also influence future expansion of midwifery group practice as a model of care more generally. In the computer-assisted analysis, five themes were identified from the data: women with complex care: specialty clinic; continuity of care; workforce; and opportunity (Fig. 1). Themes and sub-themes similar to those that emerged from manual analysis were around the woman’s experience, workforce, standards of care and opportunity. Fig. 1Leximancer 4™ generated themes from stakeholder interviews Leximancer 4™ generated themes from stakeholder interviews Table 3 shows the mapped overarching themes and how they relate to the five CFIR domains and constructs. In this table it becomes clear what the requirements for successful and sustained implementation of the proposed model might be. The theme Gold standard care mapped to five constructs indicates that either a perception, or knowledge of evidence supporting midwifery group practice for vulnerable women would be critical to its success. A thorough consideration of potential experiences for the woman, the workforce and costs when preparing a business case, will be a determinant of model implementation success. An interdisciplinary team that is already part of the organisational structure and engaged in planning the model is likely to be essential. Processes that demonstrate evidence of planning and reflecting across all CFIR domains, especially regarding linkages between different health professional disciplines and costs, are also important. Table 3Summary of interview themes mapped to Consolidated Framework for Implementation Research (CFIR) domains and constructsCFIR domains and constructsThemes Intervention characteristics Evidence strength and qualityGold standard careRelative advantageThe woman’s experienceMidwifery workforceCostsCostCostsAdaptabilityMidwifery workforce Outer setting Patient needs and resourcesThe woman’s experiencePeer PressureGold standard care Inner setting Structural characteristicsThe interdisciplinary teamImplementation climateMidwifery workforceRelative prioritiesGold standard careThe woman’s experience Characteristics of individuals Knowledge and beliefs about the interventionGold standard careIndividual stage of changeGold standard careSelf-efficacyMidwifery workforce Process PlanningThe interdisciplinary teamReflecting and evaluatingCosts Summary of interview themes mapped to Consolidated Framework for Implementation Research (CFIR) domains and constructs The woman’s experience Midwifery workforce Costs Gold standard care The woman’s experience
Conclusion
In this study, we have used the CFIR to identify potential barriers and enablers to implementing a midwifery group practice for vulnerable women, with both local and national relevance. There was recognition that the proposed model of care is supported by research and a view that clinical benefits will outweigh costs, however supervision and support is required for midwives to manage and limit vicarious trauma. An interdisciplinary team supporting the midwives is also an essential component of the service design. For maternity services seeking to implement a midwifery group practice for vulnerable women, our results can be leveraged to further investigate other local contexts, and quickly identify strategies for effective and sustained implementation of the new model of care [48].
[ "Background", "Methods", "Setting and participants", "Process", "Data Analysis", "The woman’s experience", "Midwifery workforce", "Gold standard care", "The interdisciplinary team", "Costs", "Intervention characteristics", "Outer setting", "Inner setting", "Characteristics of individuals", "Process", "Strengths", "Limitations", "" ]
[ "Pregnant women with a history of substance use, mental health challenges and complex social issues have unique care needs during pregnancy, birth and the postpartum period [1] and are estimated to represent over 5% of the approximately 295,000 women who give birth each year in Australia [2]. There is strong international evidence that these women’s pregnancies are more likely to result in placental abruption, preterm birth, neonates that are small for gestational age, and neonatal admission to an intensive care unit [3, 4]. Vulnerable women may also experience domestic and family violence isolation in addition to poor maternal health, further compromising the fetus and neonate [1].\nExisting maternity services may not meet the needs of vulnerable women during pregnancy, resulting in non-attendance of scheduled antenatal care and raising the risk of poor maternal and neonatal outcomes [5]. Whilst Australian data on non-attendance at antenatal appointments is limited, international evidence suggests antenatal care is not well accessed by vulnerable women. In the United Kingdom, smokers were significantly more likely to have a late booking appointment after 18 weeks gestation (Odds Ratio 1.6) [6]. In Finland, women who smoked and consumed alcohol were significantly more likely to receive insufficient antenatal care defined as between zero and five visits (Odds Ratios 1.87 and 1.48 respectively) [7]. Other studies in New Zealand, Belgium and the Netherlands demonstrated low antenatal care attendance by women with social and other vulnerability [8, 9]. Developing and implementing an antenatal, birth and postnatal service that addresses the challenges experienced by vulnerable women requires consideration of potential barriers to and enablers of successful care at both the health care provider and broader health service levels.\nContinuity of care and carer is known to provide many benefits in terms of the health outcomes of mothers and babies and the levels of satisfaction of care for both consumers and care providers [10]. In Australia, most available and publicly funded models of maternity care are fragmented with limited continuity of carer across the whole pregnancy and post-partum period [11]. These Australian models facilitate monitoring of antenatal clinical indicators but may miss an opportunity to establish trusting relationships through continuity of carers during pregnancy and the postpartum period [12]. Establishment of trusting relationships is likely to improve attendance at care and enable discussion of behaviour changes during pregnancy. An alternative to this is Midwifery Group Practice (MGP), also called continuity of midwifery or caseload midwifery which has been implemented globally and largely focusses on low-risk pregnancies [10, 13]. However, such a model of care is rarely available for Australian women with complex social circumstances or with specific cultural or other needs [11]. The benefits of continuity of care and carer are well documented [10] and are likely to be seen in vulnerable women. Such antenatal care may facilitate equitable access, high quality health care and the best possible health outcomes during pregnancy, birth and the postpartum period [12].\nMidwifery and midwifery group practice is recommended for all vulnerable women [12, 14–16] because of improved health outcomes for both mothers and babies. There are benefits in the areas of child protection interventions [17], mental illness [18, 19], substance-use [15, 20, 21] and infant neurodevelopment problems [22]. An absence of continuity of care has been identified as a barrier to seeking help for mental illness [18, 23]. Relationships between women and carers that are grounded in an interdisciplinary continuity of care and carer model increases women’s access to services and provide safe spaces for disclosure of sensitive information that guides high quality health care delivery [19]. High levels of consumer satisfaction are reported by women who experience midwifery group practice. Midwifery group practice is also safe in terms of maternal and neonatal outcomes, is associated with a reduction in maternal risk-taking behaviours, and is a less-costly model of maternity care [24–26]. While midwifery group practice for vulnerable women is recommended based on evidence from small international studies, it is not known how widespread or effective the model is in Australia.\nIt is also unknown how accepted midwifery group practice is amongst key stakeholders in Australia. Midwifery group practices have been established for Torres Strait Islander women and women carrying Aboriginal and Torres Strait Islander babies in recognition of the importance of continuity of carers for this vulnerable group [27, 28]. These midwifery group practices cater for only a small proportion of vulnerable pregnant women and so many other women continue to have antenatal care that does not meet their needs. One study has reported that health professionals are supportive of an interdisciplinary midwifery group practice model of care for vulnerable women where disciplines provide specialty support with the benefits of a continuity of care model [29]. Furthermore, to ensure sustained success, implementation of a midwifery group practice for vulnerable women requires careful planning using the principles of implementation science [30]. Risk management assessment indicates the need to identify likely barriers and enablers. Effective processes and strategies used to implement a midwifery group practice for vulnerable women depend on full engagement of stakeholders and a clear picture of the health service context [30, 31]. Not all stakeholders may be supportive and there may be less-visible aspects of the health service that may make sustained implementation of a midwifery group practice for vulnerable women difficult.\nTo identify these potential barriers and enablers, a context assessment was undertaken for a proposed midwifery group practice for vulnerable women at a single site tertiary maternity service in Queensland, Australia. Our intent is to identify and share the results of the context assessment which can be applied to other maternity services across Australia and demonstrate implementation science methods as an appropriate approach. Our research may expedite the implementation of such a model of care in other Australian maternity services.", "We aimed to identify the potential barriers and enablers for implementing a midwifery group practice for vulnerable women. To do this, we conducted a qualitative context assessment using the Consolidated Framework for Implementation Research (CFIR) [31–33]. The CFIR was chosen to guide the context assessment because the process required engagement with individuals and groups across multiple levels of the health service and external stakeholders. The framework has five domains that reflect key elements of a health service that need to be investigated before implementing change in an established interdisciplinary service. These are: “intervention characteristics” such as details of the new service being proposed; “outer setting” such as external influencing factors; “inner setting” such as unique aspects of the health service itself; “characteristics of individuals” who are involved in and/or exposed to the new service; and “process” such as ways of implementing and evaluating the new service [31]. The CFIR has demonstrated applicability to data collection, analysis and implementation within maternity settings [34, 35] and provided a practical framework to assess the multiple factors involved in planning for a new midwifery group practice for vulnerable women.\n[SUBTITLE] Setting and participants [SUBSECTION] The 1000-bed health service is located in an inner suburb of an Australian capital city, with a diverse population catchment including large numbers of disadvantaged groups. There are 8000 staff on campus encompassing a multitude of complex health services [34] This study setting was selected as midwifery managers sought to improve accessibility of care delivered to vulnerable women, acutely aware of a high failure-to-attend rate where more than 25% (n = 205) of vulnerable women do not attend their scheduled antenatal care compared to 8% in the general population each year (unpublished health service data available on request). Whilst some MGP’s are already established there is no ability to be engaged concomitantly with the antenatal model of care available to vulnerable women [36]. As a specialty service the number of staff involved with the model of care for vulnerable women is small. In total, 40 internal and external stakeholders were invited to participate including: medical, nursing, midwifery, allied health, business, administration, and consumer representatives.\nThe 1000-bed health service is located in an inner suburb of an Australian capital city, with a diverse population catchment including large numbers of disadvantaged groups. There are 8000 staff on campus encompassing a multitude of complex health services [34] This study setting was selected as midwifery managers sought to improve accessibility of care delivered to vulnerable women, acutely aware of a high failure-to-attend rate where more than 25% (n = 205) of vulnerable women do not attend their scheduled antenatal care compared to 8% in the general population each year (unpublished health service data available on request). Whilst some MGP’s are already established there is no ability to be engaged concomitantly with the antenatal model of care available to vulnerable women [36]. As a specialty service the number of staff involved with the model of care for vulnerable women is small. In total, 40 internal and external stakeholders were invited to participate including: medical, nursing, midwifery, allied health, business, administration, and consumer representatives.\n[SUBTITLE] Process [SUBSECTION] Qualitative semi-structured interviews (individual and small group) were designed using questions from the CFIR toolkit that explored each CFIR domain [32]. Some questions were adapted for local contextualisation and conversation style. Participant knowledge and awareness was assessed through the use of nine open-ended questions (Table 1). Participants (n = 9) in management / leadership roles were invited to respond to an additional question in relation to costs. A modified survey was completed by the business representative officer with clinically based questions removed and others added to retain a business and cost focus. As the interviews were guided by the CFIR, questions were not pilot tested.\n\nTable 1Collated interview questions mapped to Consolidated Framework for Implementation Research (CFIR) domainsCFIR DomainInterview QuestionIntervention CharacteristicsWhat kind of information or evidence are you aware of that shows whether MGP would work in this setting?What advantages and disadvantages does MGP have for the needs for vulnerable women?What kinds of change would need to be made for an MGP to be effective for vulnerable women?What costs will be incurred to implement MGP for vulnerable women?Outer SettingHow well do you think the women who would use the current service will respond to an MGP?Can you tell me what you know about other organisations that have implemented MGP or similar programs for vulnerable women?What kind of local, state or national performance measures, policies, regulations or guidelines influence the decision?Inner SettingWhat is the general level of knowledge and acceptance at the hospital for an MGP or similar program for vulnerable women?To what extent might the implementation take a back seat to other high priority initiatives going on now?Characteristics of IndividualsHow do you feel about an MGP for vulnerable women?ProcessTo what extent has the organisation / unit set goals for implementation of further continuity of care models with a known midwife (through Midwifery Group Practice)?How important do you think it is to implement the intervention compared to the other priorities?How important is it to others, such as your co-workers or leaders, to implement the intervention compared to the other priorities?(Question for the business representative only).\n\nCollated interview questions mapped to Consolidated Framework for Implementation Research (CFIR) domains\nCan you tell me what you know about other organisations that have implemented MGP or similar programs for vulnerable women?\nWhat kind of local, state or national performance measures, policies, regulations or guidelines influence the decision?\nTo what extent has the organisation / unit set goals for implementation of further continuity of care models with a known midwife (through Midwifery Group Practice)?\nHow important do you think it is to implement the intervention compared to the other priorities?\nHow important is it to others, such as your co-workers or leaders, to implement the intervention compared to the other priorities?\n(Question for the business representative only).\nThe participants identified through purposeful sampling [37] were sent invitations including an information sheet providing a brief background to the study, via email, with open invitations also promoted at staff meetings. Participants could choose between attending individual or small group interviews that lasted 20 to 30 min. Thirteen individual interviews and 7 group sessions were held over a month’s period in 2019 on-site at the facility. Group interviews included between two and five members based on availability of attendees. All interviews were conducted in-person except for two where phone interviews were used at the request of the participants. Two female researchers (PS and DR) conducted the interviews with PS facilitating all, and DR co-leading all but three sessions due to her clinical availability. The two novice researchers, both dual registered nurses and midwives at senior and middle management levels were known to all participants. Demographic data were collected from all participants by a written survey at the interview to provide an overview of participants’ characteristics. Interviews were recorded with participant consent and transcribed verbatim by an administration support officer. Notes were also made by the interviewers. All data were de-identified in preparation for data analysis. Following the interviews, participants were notified that their de-identified information would be considered in subsequent planning for a midwifery group practice for vulnerable women. Participants were also invited to provide further information via email at their convenience, but transcripts would not be returned to comments for comment or correction. Information received by email post-interview was collated for de-identification and included in the analysis.\nThe study was deemed by the hospital’s and university’s Human Research Ethics Committees as a quality assurance or quality improvement activity and thus not requiring formal ethics approval (Exemption number: LNR/2019/QRBW/54,360). As part of this ethical review, we were required to provide brief background information on the proposed MGP model of care to participants. Background information included one sentence about the evidence for midwifery group practice being an appropriate solution for vulnerable pregnant women and the purpose of the study and risks and benefits of participating. Participants were also advised their involvement was voluntary and that their responses could be withdrawn at any stage up to two-weeks post-interview. Feedback on study results at completion of the study was provided to participants who requested follow-up.\nQualitative semi-structured interviews (individual and small group) were designed using questions from the CFIR toolkit that explored each CFIR domain [32]. Some questions were adapted for local contextualisation and conversation style. Participant knowledge and awareness was assessed through the use of nine open-ended questions (Table 1). Participants (n = 9) in management / leadership roles were invited to respond to an additional question in relation to costs. A modified survey was completed by the business representative officer with clinically based questions removed and others added to retain a business and cost focus. As the interviews were guided by the CFIR, questions were not pilot tested.\n\nTable 1Collated interview questions mapped to Consolidated Framework for Implementation Research (CFIR) domainsCFIR DomainInterview QuestionIntervention CharacteristicsWhat kind of information or evidence are you aware of that shows whether MGP would work in this setting?What advantages and disadvantages does MGP have for the needs for vulnerable women?What kinds of change would need to be made for an MGP to be effective for vulnerable women?What costs will be incurred to implement MGP for vulnerable women?Outer SettingHow well do you think the women who would use the current service will respond to an MGP?Can you tell me what you know about other organisations that have implemented MGP or similar programs for vulnerable women?What kind of local, state or national performance measures, policies, regulations or guidelines influence the decision?Inner SettingWhat is the general level of knowledge and acceptance at the hospital for an MGP or similar program for vulnerable women?To what extent might the implementation take a back seat to other high priority initiatives going on now?Characteristics of IndividualsHow do you feel about an MGP for vulnerable women?ProcessTo what extent has the organisation / unit set goals for implementation of further continuity of care models with a known midwife (through Midwifery Group Practice)?How important do you think it is to implement the intervention compared to the other priorities?How important is it to others, such as your co-workers or leaders, to implement the intervention compared to the other priorities?(Question for the business representative only).\n\nCollated interview questions mapped to Consolidated Framework for Implementation Research (CFIR) domains\nCan you tell me what you know about other organisations that have implemented MGP or similar programs for vulnerable women?\nWhat kind of local, state or national performance measures, policies, regulations or guidelines influence the decision?\nTo what extent has the organisation / unit set goals for implementation of further continuity of care models with a known midwife (through Midwifery Group Practice)?\nHow important do you think it is to implement the intervention compared to the other priorities?\nHow important is it to others, such as your co-workers or leaders, to implement the intervention compared to the other priorities?\n(Question for the business representative only).\nThe participants identified through purposeful sampling [37] were sent invitations including an information sheet providing a brief background to the study, via email, with open invitations also promoted at staff meetings. Participants could choose between attending individual or small group interviews that lasted 20 to 30 min. Thirteen individual interviews and 7 group sessions were held over a month’s period in 2019 on-site at the facility. Group interviews included between two and five members based on availability of attendees. All interviews were conducted in-person except for two where phone interviews were used at the request of the participants. Two female researchers (PS and DR) conducted the interviews with PS facilitating all, and DR co-leading all but three sessions due to her clinical availability. The two novice researchers, both dual registered nurses and midwives at senior and middle management levels were known to all participants. Demographic data were collected from all participants by a written survey at the interview to provide an overview of participants’ characteristics. Interviews were recorded with participant consent and transcribed verbatim by an administration support officer. Notes were also made by the interviewers. All data were de-identified in preparation for data analysis. Following the interviews, participants were notified that their de-identified information would be considered in subsequent planning for a midwifery group practice for vulnerable women. Participants were also invited to provide further information via email at their convenience, but transcripts would not be returned to comments for comment or correction. Information received by email post-interview was collated for de-identification and included in the analysis.\nThe study was deemed by the hospital’s and university’s Human Research Ethics Committees as a quality assurance or quality improvement activity and thus not requiring formal ethics approval (Exemption number: LNR/2019/QRBW/54,360). As part of this ethical review, we were required to provide brief background information on the proposed MGP model of care to participants. Background information included one sentence about the evidence for midwifery group practice being an appropriate solution for vulnerable pregnant women and the purpose of the study and risks and benefits of participating. Participants were also advised their involvement was voluntary and that their responses could be withdrawn at any stage up to two-weeks post-interview. Feedback on study results at completion of the study was provided to participants who requested follow-up.\n[SUBTITLE] Data Analysis [SUBSECTION] In the initial manual thematic analysis, two midwife researchers used a grounded-theory approach and facilitated emergence of multiple themes from the data [38]. Each researcher independently analysed the data, highlighted key terms and assigned their own codes. The researchers conferred to agree on a joint understanding of the themes which emerged. Key words and phrases which were repeated amongst participants were tabulated. Agreed key words and phrases were organised into barriers and enablers as determined by consensus amongst the researchers and then coded “in vivo” using participants’ own words. As patterns emerged in the reassembling of data and coding, recurrent themes were identified to enable thematic analysis [38]. A coding tree was not created, as software was not used for this component of the analysis.\nReflexivity and reduction of potential researcher bias was identified and considered throughout the interview and analysis processes [38]. The midwife researchers reflected on and acknowledged both the potential bias of being midwives investigating a topic they may have a self-interest in, along with the benefits of improved engagement from participants as they were known colleagues.\nTo promote rigor and dependability in the study findings, a second round of analysis was conducted [39–41]. Peer checking was undertaken independently (by CK) through analysis of the de-identified research transcripts using Leximancer V4. Leximancer is a well-known text mining software used to identify concepts grounded in the study data [39] and has been used in Australian maternity settings [42, 43]. Peer checking using a text mining software reduces inadvertent researcher bias and provides another level of dependability within the study findings. The second analysis was compared with themes from first round analysis thus establishing findings across three researchers using two methods, and substantiating trustworthiness in the study [44]. If there were differences in manual and computer analysis results, the research team planned to reach a consensus on emerging themes through discussion. Themes were then mapped to CFIR domains and constructs within each domain to finalise the analysis. Key phrases and meaning from interview data were used to allocate themes to constructs.\nIn the initial manual thematic analysis, two midwife researchers used a grounded-theory approach and facilitated emergence of multiple themes from the data [38]. Each researcher independently analysed the data, highlighted key terms and assigned their own codes. The researchers conferred to agree on a joint understanding of the themes which emerged. Key words and phrases which were repeated amongst participants were tabulated. Agreed key words and phrases were organised into barriers and enablers as determined by consensus amongst the researchers and then coded “in vivo” using participants’ own words. As patterns emerged in the reassembling of data and coding, recurrent themes were identified to enable thematic analysis [38]. A coding tree was not created, as software was not used for this component of the analysis.\nReflexivity and reduction of potential researcher bias was identified and considered throughout the interview and analysis processes [38]. The midwife researchers reflected on and acknowledged both the potential bias of being midwives investigating a topic they may have a self-interest in, along with the benefits of improved engagement from participants as they were known colleagues.\nTo promote rigor and dependability in the study findings, a second round of analysis was conducted [39–41]. Peer checking was undertaken independently (by CK) through analysis of the de-identified research transcripts using Leximancer V4. Leximancer is a well-known text mining software used to identify concepts grounded in the study data [39] and has been used in Australian maternity settings [42, 43]. Peer checking using a text mining software reduces inadvertent researcher bias and provides another level of dependability within the study findings. The second analysis was compared with themes from first round analysis thus establishing findings across three researchers using two methods, and substantiating trustworthiness in the study [44]. If there were differences in manual and computer analysis results, the research team planned to reach a consensus on emerging themes through discussion. Themes were then mapped to CFIR domains and constructs within each domain to finalise the analysis. Key phrases and meaning from interview data were used to allocate themes to constructs.", "The 1000-bed health service is located in an inner suburb of an Australian capital city, with a diverse population catchment including large numbers of disadvantaged groups. There are 8000 staff on campus encompassing a multitude of complex health services [34] This study setting was selected as midwifery managers sought to improve accessibility of care delivered to vulnerable women, acutely aware of a high failure-to-attend rate where more than 25% (n = 205) of vulnerable women do not attend their scheduled antenatal care compared to 8% in the general population each year (unpublished health service data available on request). Whilst some MGP’s are already established there is no ability to be engaged concomitantly with the antenatal model of care available to vulnerable women [36]. As a specialty service the number of staff involved with the model of care for vulnerable women is small. In total, 40 internal and external stakeholders were invited to participate including: medical, nursing, midwifery, allied health, business, administration, and consumer representatives.", "Qualitative semi-structured interviews (individual and small group) were designed using questions from the CFIR toolkit that explored each CFIR domain [32]. Some questions were adapted for local contextualisation and conversation style. Participant knowledge and awareness was assessed through the use of nine open-ended questions (Table 1). Participants (n = 9) in management / leadership roles were invited to respond to an additional question in relation to costs. A modified survey was completed by the business representative officer with clinically based questions removed and others added to retain a business and cost focus. As the interviews were guided by the CFIR, questions were not pilot tested.\n\nTable 1Collated interview questions mapped to Consolidated Framework for Implementation Research (CFIR) domainsCFIR DomainInterview QuestionIntervention CharacteristicsWhat kind of information or evidence are you aware of that shows whether MGP would work in this setting?What advantages and disadvantages does MGP have for the needs for vulnerable women?What kinds of change would need to be made for an MGP to be effective for vulnerable women?What costs will be incurred to implement MGP for vulnerable women?Outer SettingHow well do you think the women who would use the current service will respond to an MGP?Can you tell me what you know about other organisations that have implemented MGP or similar programs for vulnerable women?What kind of local, state or national performance measures, policies, regulations or guidelines influence the decision?Inner SettingWhat is the general level of knowledge and acceptance at the hospital for an MGP or similar program for vulnerable women?To what extent might the implementation take a back seat to other high priority initiatives going on now?Characteristics of IndividualsHow do you feel about an MGP for vulnerable women?ProcessTo what extent has the organisation / unit set goals for implementation of further continuity of care models with a known midwife (through Midwifery Group Practice)?How important do you think it is to implement the intervention compared to the other priorities?How important is it to others, such as your co-workers or leaders, to implement the intervention compared to the other priorities?(Question for the business representative only).\n\nCollated interview questions mapped to Consolidated Framework for Implementation Research (CFIR) domains\nCan you tell me what you know about other organisations that have implemented MGP or similar programs for vulnerable women?\nWhat kind of local, state or national performance measures, policies, regulations or guidelines influence the decision?\nTo what extent has the organisation / unit set goals for implementation of further continuity of care models with a known midwife (through Midwifery Group Practice)?\nHow important do you think it is to implement the intervention compared to the other priorities?\nHow important is it to others, such as your co-workers or leaders, to implement the intervention compared to the other priorities?\n(Question for the business representative only).\nThe participants identified through purposeful sampling [37] were sent invitations including an information sheet providing a brief background to the study, via email, with open invitations also promoted at staff meetings. Participants could choose between attending individual or small group interviews that lasted 20 to 30 min. Thirteen individual interviews and 7 group sessions were held over a month’s period in 2019 on-site at the facility. Group interviews included between two and five members based on availability of attendees. All interviews were conducted in-person except for two where phone interviews were used at the request of the participants. Two female researchers (PS and DR) conducted the interviews with PS facilitating all, and DR co-leading all but three sessions due to her clinical availability. The two novice researchers, both dual registered nurses and midwives at senior and middle management levels were known to all participants. Demographic data were collected from all participants by a written survey at the interview to provide an overview of participants’ characteristics. Interviews were recorded with participant consent and transcribed verbatim by an administration support officer. Notes were also made by the interviewers. All data were de-identified in preparation for data analysis. Following the interviews, participants were notified that their de-identified information would be considered in subsequent planning for a midwifery group practice for vulnerable women. Participants were also invited to provide further information via email at their convenience, but transcripts would not be returned to comments for comment or correction. Information received by email post-interview was collated for de-identification and included in the analysis.\nThe study was deemed by the hospital’s and university’s Human Research Ethics Committees as a quality assurance or quality improvement activity and thus not requiring formal ethics approval (Exemption number: LNR/2019/QRBW/54,360). As part of this ethical review, we were required to provide brief background information on the proposed MGP model of care to participants. Background information included one sentence about the evidence for midwifery group practice being an appropriate solution for vulnerable pregnant women and the purpose of the study and risks and benefits of participating. Participants were also advised their involvement was voluntary and that their responses could be withdrawn at any stage up to two-weeks post-interview. Feedback on study results at completion of the study was provided to participants who requested follow-up.", "In the initial manual thematic analysis, two midwife researchers used a grounded-theory approach and facilitated emergence of multiple themes from the data [38]. Each researcher independently analysed the data, highlighted key terms and assigned their own codes. The researchers conferred to agree on a joint understanding of the themes which emerged. Key words and phrases which were repeated amongst participants were tabulated. Agreed key words and phrases were organised into barriers and enablers as determined by consensus amongst the researchers and then coded “in vivo” using participants’ own words. As patterns emerged in the reassembling of data and coding, recurrent themes were identified to enable thematic analysis [38]. A coding tree was not created, as software was not used for this component of the analysis.\nReflexivity and reduction of potential researcher bias was identified and considered throughout the interview and analysis processes [38]. The midwife researchers reflected on and acknowledged both the potential bias of being midwives investigating a topic they may have a self-interest in, along with the benefits of improved engagement from participants as they were known colleagues.\nTo promote rigor and dependability in the study findings, a second round of analysis was conducted [39–41]. Peer checking was undertaken independently (by CK) through analysis of the de-identified research transcripts using Leximancer V4. Leximancer is a well-known text mining software used to identify concepts grounded in the study data [39] and has been used in Australian maternity settings [42, 43]. Peer checking using a text mining software reduces inadvertent researcher bias and provides another level of dependability within the study findings. The second analysis was compared with themes from first round analysis thus establishing findings across three researchers using two methods, and substantiating trustworthiness in the study [44]. If there were differences in manual and computer analysis results, the research team planned to reach a consensus on emerging themes through discussion. Themes were then mapped to CFIR domains and constructs within each domain to finalise the analysis. Key phrases and meaning from interview data were used to allocate themes to constructs.", "Discussion around the woman’s experience identified enablers for the proposed model. Many participants expressed empathy for the likely patient cohort and placed the proposed model as a high priority for the health service. Enablers included perceptions of improved clinical outcomes, building of safety and trust, patient satisfaction and the potential to reduce ‘fail to attend’ rates at the specialty clinic. This is supported by comments from participants:\n\n… vulnerable or disadvantaged groups would benefit … (Other role, Interview 1).\n… some with abuse histories don’t want to go over those histories over and over\nagain…some come with diverse cultural situations and specific needs … (Midwife, Interview 2).… they can build up trust, they can have the tough talk with a familiar face … (Nurse, Interview 12).\n\n… vulnerable or disadvantaged groups would benefit … (Other role, Interview 1).\n\n… some with abuse histories don’t want to go over those histories over and over\n\nagain…some come with diverse cultural situations and specific needs … (Midwife, Interview 2).\n… they can build up trust, they can have the tough talk with a familiar face … (Nurse, Interview 12).\nThe general sentiment expressed by many participants is captured by this statement:\n\n…the patient can build a rapport and have trust with the clinicians…. It not only makes the patient feel safe and more comfortable with their surroundings, in my opinion, it makes the patient a bit more accountable and builds rapport with that clinician and women would be more engaged to come back… (Other role, Interview 1).\n\n…the patient can build a rapport and have trust with the clinicians…. It not only makes the patient feel safe and more comfortable with their surroundings, in my opinion, it makes the patient a bit more accountable and builds rapport with that clinician and women would be more engaged to come back… (Other role, Interview 1).\nThe proposed model was being viewed as positive and with a woman-centred focus. These views become potential enablers when preparing detailed business cases that demonstrate strategic and strong stakeholder support as well as alignment with evidence and policy advocating woman-centred care.\nHowever, potential barriers centred around concerns that women may disengage if they did not bond with the known midwife, were socially isolated or feared being reported to child safety services:\n\nFor the women it’s positive all around, unless they felt they couldn’t engage with the midwife, which could lead to the women disengaging completely. (Midwife, Interview 6)The women can be fearful about opening up…that there may be negative consequences… (Midwife, Interview 10).\n\nFor the women it’s positive all around, unless they felt they couldn’t engage with the midwife, which could lead to the women disengaging completely. (Midwife, Interview 6)\nThe women can be fearful about opening up…that there may be negative consequences… (Midwife, Interview 10).\nParticipants also suggested ways to address the woman’s disengagement through awareness and actions by the midwives:\n\nWe have to be open and transparent…so there’s a consistent approach and that we can be strong and recognise it when it (disengagement) happens and maintain the relationship … (Midwife, Interview 10).… the midwife would need to be capable of referring… to those that can help … (Midwife, Interview 16).… if that relationship isn’t working there could be space to swap … (Other role, Interview 17).\n\nWe have to be open and transparent…so there’s a consistent approach and that we can be strong and recognise it when it (disengagement) happens and maintain the relationship … (Midwife, Interview 10).\n… the midwife would need to be capable of referring… to those that can help … (Midwife, Interview 16).\n… if that relationship isn’t working there could be space to swap … (Other role, Interview 17).\nWhile women’s disengagement from the proposed model might be identified as a risk, in discussions around this barrier solutions were identified. Such solutions would inform risk mitigation.", "Participants saw the proposed new model as an opportunity for midwives to gain new skills and expand their scope of practice which was identified as an enabler:\n\nThere are a few midwives out there currently upskilling themselves and are really passionate and interested and already preparing for being part of the team … (Nurse/Midwife Leader, Interview 3).Midwives will actually see this as a positive move and it’s going to be development for them, and it’s going to be opportunistic for them … (Nurse/Midwife Leader, Interview 4).\n\nThere are a few midwives out there currently upskilling themselves and are really passionate and interested and already preparing for being part of the team … (Nurse/Midwife Leader, Interview 3).\nMidwives will actually see this as a positive move and it’s going to be development for them, and it’s going to be opportunistic for them … (Nurse/Midwife Leader, Interview 4).\nA major workforce barrier to implementation success was how midwifery group practice would place high psychological demand on the midwives leading to burnout or vicarious trauma:\n\n… very complex women with personality disorders and high psychological needs and that could be quite demanding of one midwife as the primary caregiver … (Nurse/Midwife Leader, Interview 3).… to field all those phone calls and constantly support that person would be really challenging … (Nurse/Midwife Leader, Interview 3).For the midwife dealing with only these women, it could over time be mentally challenging … potentially exhausting and tiring … (Midwife, Interview 6).\n\n… very complex women with personality disorders and high psychological needs and that could be quite demanding of one midwife as the primary caregiver … (Nurse/Midwife Leader, Interview 3).\n… to field all those phone calls and constantly support that person would be really challenging … (Nurse/Midwife Leader, Interview 3).\nFor the midwife dealing with only these women, it could over time be mentally challenging … potentially exhausting and tiring … (Midwife, Interview 6).\nThe difficulty in attracting midwives to the proposed model of care was an identified barrier to implementing a high standard of care:\n\n… attracting the midwives that would have an interest in it, we almost need a mother-like figure…would have to be resilient and have had a few more life experiences … (Medical Officer, Interview 13).\n\n… attracting the midwives that would have an interest in it, we almost need a mother-like figure…would have to be resilient and have had a few more life experiences … (Medical Officer, Interview 13).\nWhile this second barrier is contradictory to the initial workforce enabler identified in this theme, participants became solution focussed in the interviews, which is reflected in the theme reported below ‘The interdisciplinary team’.", "The belief amongst participants of strong, quality evidence in favour of the proposed model was identified as an enabler, and there were no barriers highlighted by participants in terms of available evidence. Many participants identified through discussion around evidence that a midwifery group practice for vulnerable women would be a positive step for the women involved. This was particularly evident for participants who had undertaken reading prior to the interview with participants expressing:\n\nI have looked online to have a look at the research… shows the best outcome for babies and mothers across the board in terms of continuity of care models … (Other role, Interview 1).MGP is gold standard and… there’s lots of research out there that shows that continuity of care is best for these women to develop a relationship … (Midwife, Interview 2).… the people caring for them are more likely to pick up on deviations from a normal emotional state … (Midwife, Interview 2).\n\nI have looked online to have a look at the research… shows the best outcome for babies and mothers across the board in terms of continuity of care models … (Other role, Interview 1).\nMGP is gold standard and… there’s lots of research out there that shows that continuity of care is best for these women to develop a relationship … (Midwife, Interview 2).\n… the people caring for them are more likely to pick up on deviations from a normal emotional state … (Midwife, Interview 2).\nSome participants had undertaken self-directed reading to source additional information regarding midwifery group practice and the needs of vulnerable women. Whilst participants were sent relevant information containing brief background to the proposed study in advance of the interview, some attendees advised they wanted to come well prepared. For example, two participants advised the researchers that they were very grateful to have been invited to interview and were now more aware of the benefits of midwifery group practice for women including that this care was “gold standard”. The integral way in which having a known midwife provides benefit and support for women was repeatedly discussed by participants. For many, the perception from the evidence that a midwifery group practice for vulnerable women is “gold standard” was the sole driver for supporting the proposed model and was seen as a strong enabler which would enhance a business case for the proposed model.", "Support for the inclusion of an interdisciplinary team in the proposed model of care was identified as an enabler and was particularly supported by non-midwife participants:\n\n… I would love it, because we felt [from observation of previous staffing arrangements] that when the midwives were staying in the role longer it felt very organised and that we knew the patients very well … (Allied Health Practitioner, Interview 9).There’s also the opportunity to develop an interdisciplinary trust … (Nurse, Interview 14).\n\n… I would love it, because we felt [from observation of previous staffing arrangements] that when the midwives were staying in the role longer it felt very organised and that we knew the patients very well … (Allied Health Practitioner, Interview 9).\nThere’s also the opportunity to develop an interdisciplinary trust … (Nurse, Interview 14).\nMidwife participants felt similarly:\n\nNeed a multi-disciplinary team … and.“… still having that multi-disciplinary approach… is fantastic … (Midwife, Interview 11).\n\nNeed a multi-disciplinary team … and.\n“… still having that multi-disciplinary approach… is fantastic … (Midwife, Interview 11).\nOther responses emphasised the importance of midwives working with other disciplines and not practising in isolation:\n\nIt’s really beneficial having a whole team caring for them (Midwife, Interview 2) …and.… continuing to have the multi-disciplinary input and multi-disciplinary team meeting is going to be beneficial … (Medical Officer, Interview 20).\n\nIt’s really beneficial having a whole team caring for them (Midwife, Interview 2) …and.\n… continuing to have the multi-disciplinary input and multi-disciplinary team meeting is going to be beneficial … (Medical Officer, Interview 20).\nOthers commented:\n\n… it’s an addition to the multi-disciplinary teams, so it doesn’t take anything away but they’ve got some-one they can trust that follows them through all the way … (Medical Officer, Interview 8).… the midwives … would need to be engaged with the multi-disciplinary team more than … MGP’s … (Other role, Interview 17).\n\n… it’s an addition to the multi-disciplinary teams, so it doesn’t take anything away but they’ve got some-one they can trust that follows them through all the way … (Medical Officer, Interview 8).\n… the midwives … would need to be engaged with the multi-disciplinary team more than … MGP’s … (Other role, Interview 17).\nThe overall sentiment across disciplines was that:\n\nEveryone’s ready for a change in the space and a growth in the space and how we can improve for the women and I think it would be highly supported, valued and everyone would be on board … (Nurse/Midwife Leader, Interview 3).\n\nEveryone’s ready for a change in the space and a growth in the space and how we can improve for the women and I think it would be highly supported, valued and everyone would be on board … (Nurse/Midwife Leader, Interview 3).\nAn interdisciplinary team was therefore an important and well supported component of the model, and no barriers to including an inter-disciplinary team in the model were identified. Participants were clear that midwives would lead continuity of care, while having expert health professionals involved to provide comprehensive care for the women. They would also provide professional support for midwives who might feel isolated in their role. These design details can be included in a business case to ensure successful and sustained implementation.", "Participants believed that the health benefits of the proposed model of care would outweigh the perception that a midwifery group practice for vulnerable women was a more expensive model of care. This belief was a clear enabler of the proposed model:\n\n… the cost would come with great reward … (Nurse/Midwife Leader, Interview 3).… there will be extra cost, but the trade off in terms of good follow up might not save money but it will be money well spent … (Medical Officer, Interview 8).\n\n… the cost would come with great reward … (Nurse/Midwife Leader, Interview 3).\n… there will be extra cost, but the trade off in terms of good follow up might not save money but it will be money well spent … (Medical Officer, Interview 8).\nHowever, participants did express concern at the perceived increased need for resources and challenges with attributing costs to a range of clinical areas. The concern focussed around the potential impact on other teams which was seen as a barrier:\n\nI think there is resistance…they see this is going to be taking away from their skill mix and FTE (Full Time Equivalent) … (Nurse/Midwife Leader, Interview 4).\n\nI think there is resistance…they see this is going to be taking away from their skill mix and FTE (Full Time Equivalent) … (Nurse/Midwife Leader, Interview 4).\nHowever, other participants believed that these perceptions and challenges could be overcome and that the proposed model of care should be a priority for the hospital:\n\n… yes there are financial implications and barriers, there doesn’t seem to be good evidence to show why not … (Other role, Interview 1).… yes, it is very important that we generate the activity to get something for the work that we’re doing, but at the end of the day we are looking at patient centred care, so if it’s easier and the best outcome for mother and baby… then that’s what we have to do … (Other role, Interview 1).Why shouldn’t they have an MGP, they shouldn’t be excluded just because they’ve had drug and alcohol or mental health issues in the past … (Nurse, Interview 15).I see it as high priority to look at how we can increase activity and treat this vulnerable group … (Nurse/Midwife Leader, Interview 4).\n\n… yes there are financial implications and barriers, there doesn’t seem to be good evidence to show why not … (Other role, Interview 1).\n… yes, it is very important that we generate the activity to get something for the work that we’re doing, but at the end of the day we are looking at patient centred care, so if it’s easier and the best outcome for mother and baby… then that’s what we have to do … (Other role, Interview 1).\nWhy shouldn’t they have an MGP, they shouldn’t be excluded just because they’ve had drug and alcohol or mental health issues in the past … (Nurse, Interview 15).\nI see it as high priority to look at how we can increase activity and treat this vulnerable group … (Nurse/Midwife Leader, Interview 4).\nPerceptions around cost that are both potential enablers and barriers to gaining support and successful implementation would need to be clarified as fact in a business case before the proposed model is implemented. Participants had concerns around costs which may be reflected by decision-makers when examining the rigor in which a business case has been prepared. This may also influence future expansion of midwifery group practice as a model of care more generally.\nIn the computer-assisted analysis, five themes were identified from the data: women with complex care: specialty clinic; continuity of care; workforce; and opportunity (Fig. 1). Themes and sub-themes similar to those that emerged from manual analysis were around the woman’s experience, workforce, standards of care and opportunity.\n\nFig. 1Leximancer 4™ generated themes from stakeholder interviews\n\nLeximancer 4™ generated themes from stakeholder interviews\nTable 3 shows the mapped overarching themes and how they relate to the five CFIR domains and constructs. In this table it becomes clear what the requirements for successful and sustained implementation of the proposed model might be. The theme Gold standard care mapped to five constructs indicates that either a perception, or knowledge of evidence supporting midwifery group practice for vulnerable women would be critical to its success. A thorough consideration of potential experiences for the woman, the workforce and costs when preparing a business case, will be a determinant of model implementation success. An interdisciplinary team that is already part of the organisational structure and engaged in planning the model is likely to be essential. Processes that demonstrate evidence of planning and reflecting across all CFIR domains, especially regarding linkages between different health professional disciplines and costs, are also important.\n\nTable 3Summary of interview themes mapped to Consolidated Framework for Implementation Research (CFIR) domains and constructsCFIR domains and constructsThemes\nIntervention characteristics\nEvidence strength and qualityGold standard careRelative advantageThe woman’s experienceMidwifery workforceCostsCostCostsAdaptabilityMidwifery workforce\nOuter setting\nPatient needs and resourcesThe woman’s experiencePeer PressureGold standard care\nInner setting\nStructural characteristicsThe interdisciplinary teamImplementation climateMidwifery workforceRelative prioritiesGold standard careThe woman’s experience\nCharacteristics of individuals\nKnowledge and beliefs about the interventionGold standard careIndividual stage of changeGold standard careSelf-efficacyMidwifery workforce\nProcess\nPlanningThe interdisciplinary teamReflecting and evaluatingCosts\n\nSummary of interview themes mapped to Consolidated Framework for Implementation Research (CFIR) domains and constructs\nThe woman’s experience\nMidwifery workforce\nCosts\nGold standard care\nThe woman’s experience", "Stakeholders perceived that there was high quality and valid evidence supporting midwifery group practice for vulnerable women as “gold standard care” (Midwife, Interview 2). Quality evidence drawn from Australian studies [10, 13] builds confidence amongst stakeholders, which is a strong enabler. Participants weighed the relative advantages of the proposed model of care over the existing care provided to vulnerable women and believed that the health benefits for the women and infants would outweigh the costs. This belief in the net benefit when costs were also considered was not rooted in available evidence, which contrasts with the desire for evidence for “gold standard” effectiveness. The cost of the proposed model had not been established but was imagined by participants to be higher than both the current model of care and comparative midwifery group practices.\nParticipants were mostly clinicians with more than 10 years’ experience and so were likely able to make accurate assumptions around the number of workforce hours required to build rapport with women, discuss the care being delivered with the women and amongst colleagues, and deliver the volume of care required to optimise health outcomes. However, there is no known evidence for the cost-effectiveness of such a niche model of care, only generalised costs reported for Australian midwifery group practice [10, 45]. For Australian maternity services looking to implement a midwifery group practice for vulnerable women, careful understanding of costs and transparent communication to decision-makers is needed. Our research suggests that stakeholders naturally seek evidence for both costs and effectiveness, and in the absence of evidence, local costs should be examined.", "A midwifery group practice was perceived to meet the needs of vulnerable women because for example, “some with abuse histories don’t want to go over those histories over and over…” (Midwife, Interview 2). The continuity of midwives would ensure the deep needs of each woman were met. However, concerns were expressed regarding when having a known midwife might be a disincentive for women to engage. Primarily this related to women with involvement of child protection services or times where personal factors impacted on building rapport and a therapeutic relationship was not established between the woman and the midwife. A midwifery group practice for vulnerable women should be designed with flexibility in the case where a rapport is not being established between the care givers and the woman [46]. The service design would need to ensure that midwives can be changed across groups.", "The structural characteristics of the setting for this research were that it is a mature and large maternity service, with a range of existing and well-supported midwifery group practices already established. Consequently, there may have been fewer inner setting barriers to establishing the proposed model of care compared to Australian maternity services in which midwifery group practice is new or not yet established. Further, the interdisciplinary team engaged in this research was supportive of midwifery group practice. Although, it was highlighted that any potential changes to how the team works with the midwifery group practice would need to be implemented carefully. The interdisciplinary team expressed views that to continue safe and effective care, it was important the team’s role be maintained. The implementation climate of the inner setting was generally supportive, and there were some important reflections from the interdisciplinary team that would need to be acknowledged when preparing a business case for the proposed model.", "Stakeholders had a positive attitude towards the intervention; they placed a high value on the proposed model of care. Participants even sought evidence in preparation for the interviews and ensured they were familiar with the proposal. This demonstrates that individuals’ knowledge and beliefs about the intervention was a strong enabler for proceeding with the proposal. Further, that demonstrated engagement in the context assessment by stakeholders was an indicator that individuals were at an advanced stage of change in relation to redesigning maternity care for vulnerable women. The identification of this enabler suggests that the initial enthusiasm for the intervention would sustain its implementation over time. The results also demonstrated a moderate level of self-efficacy – there were mixed beliefs amongst individuals in their own capabilities to deliver the model of care, while also identifying that the proposed model of care would provide an opportunity for midwives to build their self-efficacy through gaining new skills and expanding their scope of practice. Modifications would need to be made to traditional midwifery group practice design due to perceived heavy demands from deep engagement with vulnerable women and the potential for vicarious trauma, burnout, and other emotional impacts for the midwives. Participants suggested that to overcome this barrier, caseloads should be reduced, and midwives should be well supervised and mentored as described also by others [46], especially when caring for vulnerable women [47]. To enable such an intervention to be implemented in other Australian maternity services, stakeholders would need to have confidence in their ability to seek and interpret the evidence and have an awareness of the strengths and limitations of the workforce capabilities to execute the proposed model of care.", "Conducting the context assessment instilled confidence in readiness of the maternity service to adopt the proposed change. The context assessment was conducted early in the planning stages of the intervention and the strong engagement suggested interest in the intervention and acceptance of the planning methods. This enabler resulted in positive and open communication and was an unintended consequence of the context assessment, as participation from a large and broad range of disciplines was not expected.", "A strength of this study was the use of the CFIR to guide interviews, along with two independent forms of data analysis and comparison of study findings. The CFIR outlines domains and constructs that are associated with effective implementation of new interventions. Using the CFIR in this study presents results that may guide other Australian maternity services on how to best plan and implement a midwifery group practice for vulnerable women. An additional strength was the alignment of the manual and computer-assisted thematic results. Computer-assisted analysis was undertaken to mitigate the recognised and acknowledged potential inherent bias in qualitative analysis [41]. Independent analysis and consistency of results further enhances the credibility and trustworthiness of the study, along with research reflexivity throughout the study. The midwife researchers (PS and DR) concluded in their reflections that the broad range of disciplines from which participants were drawn resulted in very positive engagement from the team and enhanced marketing of the proposed change in service delivery. The diversity and large relative number of stakeholders involved in the study also ensured the qualitative data were reflective of a comprehensive sample from which data saturation was readily achieved.", "A limitation of the study was that due to local facility arrangements for selection of consumer representatives only one consumer participated in the interviews. The perspectives of consumers, and staff who were less experienced and/or from culturally and socially diverse backgrounds were not given, nor was there discussion in the interviews about the impact of the proposed model on these women. In addition, the homogenous sample, high level of experience and mature age of participants may indicate unintended sampling bias. This may be further exacerbated by the reading and preparation prior to interview done by some participants. Active pursuit of the voices of consumers and staff from a representative range of backgrounds in planning new models of maternity care across Australia is recommended by the researchers. As this study was carried out in a facility with established and supported midwifery group practices, caution should be applied in generalising the specific local results to other services for which midwifery group practice is a new concept. Instead, our interpretation of results for Australian maternity services should prompt services to identify which of our results mapped to the CFIR domains are relevant, and how they might be similar or different to the local context.", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Setting and participants", "Process", "Data Analysis", "Results", "The woman’s experience", "Midwifery workforce", "Gold standard care", "The interdisciplinary team", "Costs", "Discussion", "Intervention characteristics", "Outer setting", "Inner setting", "Characteristics of individuals", "Process", "Strengths", "Limitations", "Conclusion", "Electronic supplementary material", "" ]
[ "Pregnant women with a history of substance use, mental health challenges and complex social issues have unique care needs during pregnancy, birth and the postpartum period [1] and are estimated to represent over 5% of the approximately 295,000 women who give birth each year in Australia [2]. There is strong international evidence that these women’s pregnancies are more likely to result in placental abruption, preterm birth, neonates that are small for gestational age, and neonatal admission to an intensive care unit [3, 4]. Vulnerable women may also experience domestic and family violence isolation in addition to poor maternal health, further compromising the fetus and neonate [1].\nExisting maternity services may not meet the needs of vulnerable women during pregnancy, resulting in non-attendance of scheduled antenatal care and raising the risk of poor maternal and neonatal outcomes [5]. Whilst Australian data on non-attendance at antenatal appointments is limited, international evidence suggests antenatal care is not well accessed by vulnerable women. In the United Kingdom, smokers were significantly more likely to have a late booking appointment after 18 weeks gestation (Odds Ratio 1.6) [6]. In Finland, women who smoked and consumed alcohol were significantly more likely to receive insufficient antenatal care defined as between zero and five visits (Odds Ratios 1.87 and 1.48 respectively) [7]. Other studies in New Zealand, Belgium and the Netherlands demonstrated low antenatal care attendance by women with social and other vulnerability [8, 9]. Developing and implementing an antenatal, birth and postnatal service that addresses the challenges experienced by vulnerable women requires consideration of potential barriers to and enablers of successful care at both the health care provider and broader health service levels.\nContinuity of care and carer is known to provide many benefits in terms of the health outcomes of mothers and babies and the levels of satisfaction of care for both consumers and care providers [10]. In Australia, most available and publicly funded models of maternity care are fragmented with limited continuity of carer across the whole pregnancy and post-partum period [11]. These Australian models facilitate monitoring of antenatal clinical indicators but may miss an opportunity to establish trusting relationships through continuity of carers during pregnancy and the postpartum period [12]. Establishment of trusting relationships is likely to improve attendance at care and enable discussion of behaviour changes during pregnancy. An alternative to this is Midwifery Group Practice (MGP), also called continuity of midwifery or caseload midwifery which has been implemented globally and largely focusses on low-risk pregnancies [10, 13]. However, such a model of care is rarely available for Australian women with complex social circumstances or with specific cultural or other needs [11]. The benefits of continuity of care and carer are well documented [10] and are likely to be seen in vulnerable women. Such antenatal care may facilitate equitable access, high quality health care and the best possible health outcomes during pregnancy, birth and the postpartum period [12].\nMidwifery and midwifery group practice is recommended for all vulnerable women [12, 14–16] because of improved health outcomes for both mothers and babies. There are benefits in the areas of child protection interventions [17], mental illness [18, 19], substance-use [15, 20, 21] and infant neurodevelopment problems [22]. An absence of continuity of care has been identified as a barrier to seeking help for mental illness [18, 23]. Relationships between women and carers that are grounded in an interdisciplinary continuity of care and carer model increases women’s access to services and provide safe spaces for disclosure of sensitive information that guides high quality health care delivery [19]. High levels of consumer satisfaction are reported by women who experience midwifery group practice. Midwifery group practice is also safe in terms of maternal and neonatal outcomes, is associated with a reduction in maternal risk-taking behaviours, and is a less-costly model of maternity care [24–26]. While midwifery group practice for vulnerable women is recommended based on evidence from small international studies, it is not known how widespread or effective the model is in Australia.\nIt is also unknown how accepted midwifery group practice is amongst key stakeholders in Australia. Midwifery group practices have been established for Torres Strait Islander women and women carrying Aboriginal and Torres Strait Islander babies in recognition of the importance of continuity of carers for this vulnerable group [27, 28]. These midwifery group practices cater for only a small proportion of vulnerable pregnant women and so many other women continue to have antenatal care that does not meet their needs. One study has reported that health professionals are supportive of an interdisciplinary midwifery group practice model of care for vulnerable women where disciplines provide specialty support with the benefits of a continuity of care model [29]. Furthermore, to ensure sustained success, implementation of a midwifery group practice for vulnerable women requires careful planning using the principles of implementation science [30]. Risk management assessment indicates the need to identify likely barriers and enablers. Effective processes and strategies used to implement a midwifery group practice for vulnerable women depend on full engagement of stakeholders and a clear picture of the health service context [30, 31]. Not all stakeholders may be supportive and there may be less-visible aspects of the health service that may make sustained implementation of a midwifery group practice for vulnerable women difficult.\nTo identify these potential barriers and enablers, a context assessment was undertaken for a proposed midwifery group practice for vulnerable women at a single site tertiary maternity service in Queensland, Australia. Our intent is to identify and share the results of the context assessment which can be applied to other maternity services across Australia and demonstrate implementation science methods as an appropriate approach. Our research may expedite the implementation of such a model of care in other Australian maternity services.", "We aimed to identify the potential barriers and enablers for implementing a midwifery group practice for vulnerable women. To do this, we conducted a qualitative context assessment using the Consolidated Framework for Implementation Research (CFIR) [31–33]. The CFIR was chosen to guide the context assessment because the process required engagement with individuals and groups across multiple levels of the health service and external stakeholders. The framework has five domains that reflect key elements of a health service that need to be investigated before implementing change in an established interdisciplinary service. These are: “intervention characteristics” such as details of the new service being proposed; “outer setting” such as external influencing factors; “inner setting” such as unique aspects of the health service itself; “characteristics of individuals” who are involved in and/or exposed to the new service; and “process” such as ways of implementing and evaluating the new service [31]. The CFIR has demonstrated applicability to data collection, analysis and implementation within maternity settings [34, 35] and provided a practical framework to assess the multiple factors involved in planning for a new midwifery group practice for vulnerable women.\n[SUBTITLE] Setting and participants [SUBSECTION] The 1000-bed health service is located in an inner suburb of an Australian capital city, with a diverse population catchment including large numbers of disadvantaged groups. There are 8000 staff on campus encompassing a multitude of complex health services [34] This study setting was selected as midwifery managers sought to improve accessibility of care delivered to vulnerable women, acutely aware of a high failure-to-attend rate where more than 25% (n = 205) of vulnerable women do not attend their scheduled antenatal care compared to 8% in the general population each year (unpublished health service data available on request). Whilst some MGP’s are already established there is no ability to be engaged concomitantly with the antenatal model of care available to vulnerable women [36]. As a specialty service the number of staff involved with the model of care for vulnerable women is small. In total, 40 internal and external stakeholders were invited to participate including: medical, nursing, midwifery, allied health, business, administration, and consumer representatives.\nThe 1000-bed health service is located in an inner suburb of an Australian capital city, with a diverse population catchment including large numbers of disadvantaged groups. There are 8000 staff on campus encompassing a multitude of complex health services [34] This study setting was selected as midwifery managers sought to improve accessibility of care delivered to vulnerable women, acutely aware of a high failure-to-attend rate where more than 25% (n = 205) of vulnerable women do not attend their scheduled antenatal care compared to 8% in the general population each year (unpublished health service data available on request). Whilst some MGP’s are already established there is no ability to be engaged concomitantly with the antenatal model of care available to vulnerable women [36]. As a specialty service the number of staff involved with the model of care for vulnerable women is small. In total, 40 internal and external stakeholders were invited to participate including: medical, nursing, midwifery, allied health, business, administration, and consumer representatives.\n[SUBTITLE] Process [SUBSECTION] Qualitative semi-structured interviews (individual and small group) were designed using questions from the CFIR toolkit that explored each CFIR domain [32]. Some questions were adapted for local contextualisation and conversation style. Participant knowledge and awareness was assessed through the use of nine open-ended questions (Table 1). Participants (n = 9) in management / leadership roles were invited to respond to an additional question in relation to costs. A modified survey was completed by the business representative officer with clinically based questions removed and others added to retain a business and cost focus. As the interviews were guided by the CFIR, questions were not pilot tested.\n\nTable 1Collated interview questions mapped to Consolidated Framework for Implementation Research (CFIR) domainsCFIR DomainInterview QuestionIntervention CharacteristicsWhat kind of information or evidence are you aware of that shows whether MGP would work in this setting?What advantages and disadvantages does MGP have for the needs for vulnerable women?What kinds of change would need to be made for an MGP to be effective for vulnerable women?What costs will be incurred to implement MGP for vulnerable women?Outer SettingHow well do you think the women who would use the current service will respond to an MGP?Can you tell me what you know about other organisations that have implemented MGP or similar programs for vulnerable women?What kind of local, state or national performance measures, policies, regulations or guidelines influence the decision?Inner SettingWhat is the general level of knowledge and acceptance at the hospital for an MGP or similar program for vulnerable women?To what extent might the implementation take a back seat to other high priority initiatives going on now?Characteristics of IndividualsHow do you feel about an MGP for vulnerable women?ProcessTo what extent has the organisation / unit set goals for implementation of further continuity of care models with a known midwife (through Midwifery Group Practice)?How important do you think it is to implement the intervention compared to the other priorities?How important is it to others, such as your co-workers or leaders, to implement the intervention compared to the other priorities?(Question for the business representative only).\n\nCollated interview questions mapped to Consolidated Framework for Implementation Research (CFIR) domains\nCan you tell me what you know about other organisations that have implemented MGP or similar programs for vulnerable women?\nWhat kind of local, state or national performance measures, policies, regulations or guidelines influence the decision?\nTo what extent has the organisation / unit set goals for implementation of further continuity of care models with a known midwife (through Midwifery Group Practice)?\nHow important do you think it is to implement the intervention compared to the other priorities?\nHow important is it to others, such as your co-workers or leaders, to implement the intervention compared to the other priorities?\n(Question for the business representative only).\nThe participants identified through purposeful sampling [37] were sent invitations including an information sheet providing a brief background to the study, via email, with open invitations also promoted at staff meetings. Participants could choose between attending individual or small group interviews that lasted 20 to 30 min. Thirteen individual interviews and 7 group sessions were held over a month’s period in 2019 on-site at the facility. Group interviews included between two and five members based on availability of attendees. All interviews were conducted in-person except for two where phone interviews were used at the request of the participants. Two female researchers (PS and DR) conducted the interviews with PS facilitating all, and DR co-leading all but three sessions due to her clinical availability. The two novice researchers, both dual registered nurses and midwives at senior and middle management levels were known to all participants. Demographic data were collected from all participants by a written survey at the interview to provide an overview of participants’ characteristics. Interviews were recorded with participant consent and transcribed verbatim by an administration support officer. Notes were also made by the interviewers. All data were de-identified in preparation for data analysis. Following the interviews, participants were notified that their de-identified information would be considered in subsequent planning for a midwifery group practice for vulnerable women. Participants were also invited to provide further information via email at their convenience, but transcripts would not be returned to comments for comment or correction. Information received by email post-interview was collated for de-identification and included in the analysis.\nThe study was deemed by the hospital’s and university’s Human Research Ethics Committees as a quality assurance or quality improvement activity and thus not requiring formal ethics approval (Exemption number: LNR/2019/QRBW/54,360). As part of this ethical review, we were required to provide brief background information on the proposed MGP model of care to participants. Background information included one sentence about the evidence for midwifery group practice being an appropriate solution for vulnerable pregnant women and the purpose of the study and risks and benefits of participating. Participants were also advised their involvement was voluntary and that their responses could be withdrawn at any stage up to two-weeks post-interview. Feedback on study results at completion of the study was provided to participants who requested follow-up.\nQualitative semi-structured interviews (individual and small group) were designed using questions from the CFIR toolkit that explored each CFIR domain [32]. Some questions were adapted for local contextualisation and conversation style. Participant knowledge and awareness was assessed through the use of nine open-ended questions (Table 1). Participants (n = 9) in management / leadership roles were invited to respond to an additional question in relation to costs. A modified survey was completed by the business representative officer with clinically based questions removed and others added to retain a business and cost focus. As the interviews were guided by the CFIR, questions were not pilot tested.\n\nTable 1Collated interview questions mapped to Consolidated Framework for Implementation Research (CFIR) domainsCFIR DomainInterview QuestionIntervention CharacteristicsWhat kind of information or evidence are you aware of that shows whether MGP would work in this setting?What advantages and disadvantages does MGP have for the needs for vulnerable women?What kinds of change would need to be made for an MGP to be effective for vulnerable women?What costs will be incurred to implement MGP for vulnerable women?Outer SettingHow well do you think the women who would use the current service will respond to an MGP?Can you tell me what you know about other organisations that have implemented MGP or similar programs for vulnerable women?What kind of local, state or national performance measures, policies, regulations or guidelines influence the decision?Inner SettingWhat is the general level of knowledge and acceptance at the hospital for an MGP or similar program for vulnerable women?To what extent might the implementation take a back seat to other high priority initiatives going on now?Characteristics of IndividualsHow do you feel about an MGP for vulnerable women?ProcessTo what extent has the organisation / unit set goals for implementation of further continuity of care models with a known midwife (through Midwifery Group Practice)?How important do you think it is to implement the intervention compared to the other priorities?How important is it to others, such as your co-workers or leaders, to implement the intervention compared to the other priorities?(Question for the business representative only).\n\nCollated interview questions mapped to Consolidated Framework for Implementation Research (CFIR) domains\nCan you tell me what you know about other organisations that have implemented MGP or similar programs for vulnerable women?\nWhat kind of local, state or national performance measures, policies, regulations or guidelines influence the decision?\nTo what extent has the organisation / unit set goals for implementation of further continuity of care models with a known midwife (through Midwifery Group Practice)?\nHow important do you think it is to implement the intervention compared to the other priorities?\nHow important is it to others, such as your co-workers or leaders, to implement the intervention compared to the other priorities?\n(Question for the business representative only).\nThe participants identified through purposeful sampling [37] were sent invitations including an information sheet providing a brief background to the study, via email, with open invitations also promoted at staff meetings. Participants could choose between attending individual or small group interviews that lasted 20 to 30 min. Thirteen individual interviews and 7 group sessions were held over a month’s period in 2019 on-site at the facility. Group interviews included between two and five members based on availability of attendees. All interviews were conducted in-person except for two where phone interviews were used at the request of the participants. Two female researchers (PS and DR) conducted the interviews with PS facilitating all, and DR co-leading all but three sessions due to her clinical availability. The two novice researchers, both dual registered nurses and midwives at senior and middle management levels were known to all participants. Demographic data were collected from all participants by a written survey at the interview to provide an overview of participants’ characteristics. Interviews were recorded with participant consent and transcribed verbatim by an administration support officer. Notes were also made by the interviewers. All data were de-identified in preparation for data analysis. Following the interviews, participants were notified that their de-identified information would be considered in subsequent planning for a midwifery group practice for vulnerable women. Participants were also invited to provide further information via email at their convenience, but transcripts would not be returned to comments for comment or correction. Information received by email post-interview was collated for de-identification and included in the analysis.\nThe study was deemed by the hospital’s and university’s Human Research Ethics Committees as a quality assurance or quality improvement activity and thus not requiring formal ethics approval (Exemption number: LNR/2019/QRBW/54,360). As part of this ethical review, we were required to provide brief background information on the proposed MGP model of care to participants. Background information included one sentence about the evidence for midwifery group practice being an appropriate solution for vulnerable pregnant women and the purpose of the study and risks and benefits of participating. Participants were also advised their involvement was voluntary and that their responses could be withdrawn at any stage up to two-weeks post-interview. Feedback on study results at completion of the study was provided to participants who requested follow-up.\n[SUBTITLE] Data Analysis [SUBSECTION] In the initial manual thematic analysis, two midwife researchers used a grounded-theory approach and facilitated emergence of multiple themes from the data [38]. Each researcher independently analysed the data, highlighted key terms and assigned their own codes. The researchers conferred to agree on a joint understanding of the themes which emerged. Key words and phrases which were repeated amongst participants were tabulated. Agreed key words and phrases were organised into barriers and enablers as determined by consensus amongst the researchers and then coded “in vivo” using participants’ own words. As patterns emerged in the reassembling of data and coding, recurrent themes were identified to enable thematic analysis [38]. A coding tree was not created, as software was not used for this component of the analysis.\nReflexivity and reduction of potential researcher bias was identified and considered throughout the interview and analysis processes [38]. The midwife researchers reflected on and acknowledged both the potential bias of being midwives investigating a topic they may have a self-interest in, along with the benefits of improved engagement from participants as they were known colleagues.\nTo promote rigor and dependability in the study findings, a second round of analysis was conducted [39–41]. Peer checking was undertaken independently (by CK) through analysis of the de-identified research transcripts using Leximancer V4. Leximancer is a well-known text mining software used to identify concepts grounded in the study data [39] and has been used in Australian maternity settings [42, 43]. Peer checking using a text mining software reduces inadvertent researcher bias and provides another level of dependability within the study findings. The second analysis was compared with themes from first round analysis thus establishing findings across three researchers using two methods, and substantiating trustworthiness in the study [44]. If there were differences in manual and computer analysis results, the research team planned to reach a consensus on emerging themes through discussion. Themes were then mapped to CFIR domains and constructs within each domain to finalise the analysis. Key phrases and meaning from interview data were used to allocate themes to constructs.\nIn the initial manual thematic analysis, two midwife researchers used a grounded-theory approach and facilitated emergence of multiple themes from the data [38]. Each researcher independently analysed the data, highlighted key terms and assigned their own codes. The researchers conferred to agree on a joint understanding of the themes which emerged. Key words and phrases which were repeated amongst participants were tabulated. Agreed key words and phrases were organised into barriers and enablers as determined by consensus amongst the researchers and then coded “in vivo” using participants’ own words. As patterns emerged in the reassembling of data and coding, recurrent themes were identified to enable thematic analysis [38]. A coding tree was not created, as software was not used for this component of the analysis.\nReflexivity and reduction of potential researcher bias was identified and considered throughout the interview and analysis processes [38]. The midwife researchers reflected on and acknowledged both the potential bias of being midwives investigating a topic they may have a self-interest in, along with the benefits of improved engagement from participants as they were known colleagues.\nTo promote rigor and dependability in the study findings, a second round of analysis was conducted [39–41]. Peer checking was undertaken independently (by CK) through analysis of the de-identified research transcripts using Leximancer V4. Leximancer is a well-known text mining software used to identify concepts grounded in the study data [39] and has been used in Australian maternity settings [42, 43]. Peer checking using a text mining software reduces inadvertent researcher bias and provides another level of dependability within the study findings. The second analysis was compared with themes from first round analysis thus establishing findings across three researchers using two methods, and substantiating trustworthiness in the study [44]. If there were differences in manual and computer analysis results, the research team planned to reach a consensus on emerging themes through discussion. Themes were then mapped to CFIR domains and constructs within each domain to finalise the analysis. Key phrases and meaning from interview data were used to allocate themes to constructs.", "The 1000-bed health service is located in an inner suburb of an Australian capital city, with a diverse population catchment including large numbers of disadvantaged groups. There are 8000 staff on campus encompassing a multitude of complex health services [34] This study setting was selected as midwifery managers sought to improve accessibility of care delivered to vulnerable women, acutely aware of a high failure-to-attend rate where more than 25% (n = 205) of vulnerable women do not attend their scheduled antenatal care compared to 8% in the general population each year (unpublished health service data available on request). Whilst some MGP’s are already established there is no ability to be engaged concomitantly with the antenatal model of care available to vulnerable women [36]. As a specialty service the number of staff involved with the model of care for vulnerable women is small. In total, 40 internal and external stakeholders were invited to participate including: medical, nursing, midwifery, allied health, business, administration, and consumer representatives.", "Qualitative semi-structured interviews (individual and small group) were designed using questions from the CFIR toolkit that explored each CFIR domain [32]. Some questions were adapted for local contextualisation and conversation style. Participant knowledge and awareness was assessed through the use of nine open-ended questions (Table 1). Participants (n = 9) in management / leadership roles were invited to respond to an additional question in relation to costs. A modified survey was completed by the business representative officer with clinically based questions removed and others added to retain a business and cost focus. As the interviews were guided by the CFIR, questions were not pilot tested.\n\nTable 1Collated interview questions mapped to Consolidated Framework for Implementation Research (CFIR) domainsCFIR DomainInterview QuestionIntervention CharacteristicsWhat kind of information or evidence are you aware of that shows whether MGP would work in this setting?What advantages and disadvantages does MGP have for the needs for vulnerable women?What kinds of change would need to be made for an MGP to be effective for vulnerable women?What costs will be incurred to implement MGP for vulnerable women?Outer SettingHow well do you think the women who would use the current service will respond to an MGP?Can you tell me what you know about other organisations that have implemented MGP or similar programs for vulnerable women?What kind of local, state or national performance measures, policies, regulations or guidelines influence the decision?Inner SettingWhat is the general level of knowledge and acceptance at the hospital for an MGP or similar program for vulnerable women?To what extent might the implementation take a back seat to other high priority initiatives going on now?Characteristics of IndividualsHow do you feel about an MGP for vulnerable women?ProcessTo what extent has the organisation / unit set goals for implementation of further continuity of care models with a known midwife (through Midwifery Group Practice)?How important do you think it is to implement the intervention compared to the other priorities?How important is it to others, such as your co-workers or leaders, to implement the intervention compared to the other priorities?(Question for the business representative only).\n\nCollated interview questions mapped to Consolidated Framework for Implementation Research (CFIR) domains\nCan you tell me what you know about other organisations that have implemented MGP or similar programs for vulnerable women?\nWhat kind of local, state or national performance measures, policies, regulations or guidelines influence the decision?\nTo what extent has the organisation / unit set goals for implementation of further continuity of care models with a known midwife (through Midwifery Group Practice)?\nHow important do you think it is to implement the intervention compared to the other priorities?\nHow important is it to others, such as your co-workers or leaders, to implement the intervention compared to the other priorities?\n(Question for the business representative only).\nThe participants identified through purposeful sampling [37] were sent invitations including an information sheet providing a brief background to the study, via email, with open invitations also promoted at staff meetings. Participants could choose between attending individual or small group interviews that lasted 20 to 30 min. Thirteen individual interviews and 7 group sessions were held over a month’s period in 2019 on-site at the facility. Group interviews included between two and five members based on availability of attendees. All interviews were conducted in-person except for two where phone interviews were used at the request of the participants. Two female researchers (PS and DR) conducted the interviews with PS facilitating all, and DR co-leading all but three sessions due to her clinical availability. The two novice researchers, both dual registered nurses and midwives at senior and middle management levels were known to all participants. Demographic data were collected from all participants by a written survey at the interview to provide an overview of participants’ characteristics. Interviews were recorded with participant consent and transcribed verbatim by an administration support officer. Notes were also made by the interviewers. All data were de-identified in preparation for data analysis. Following the interviews, participants were notified that their de-identified information would be considered in subsequent planning for a midwifery group practice for vulnerable women. Participants were also invited to provide further information via email at their convenience, but transcripts would not be returned to comments for comment or correction. Information received by email post-interview was collated for de-identification and included in the analysis.\nThe study was deemed by the hospital’s and university’s Human Research Ethics Committees as a quality assurance or quality improvement activity and thus not requiring formal ethics approval (Exemption number: LNR/2019/QRBW/54,360). As part of this ethical review, we were required to provide brief background information on the proposed MGP model of care to participants. Background information included one sentence about the evidence for midwifery group practice being an appropriate solution for vulnerable pregnant women and the purpose of the study and risks and benefits of participating. Participants were also advised their involvement was voluntary and that their responses could be withdrawn at any stage up to two-weeks post-interview. Feedback on study results at completion of the study was provided to participants who requested follow-up.", "In the initial manual thematic analysis, two midwife researchers used a grounded-theory approach and facilitated emergence of multiple themes from the data [38]. Each researcher independently analysed the data, highlighted key terms and assigned their own codes. The researchers conferred to agree on a joint understanding of the themes which emerged. Key words and phrases which were repeated amongst participants were tabulated. Agreed key words and phrases were organised into barriers and enablers as determined by consensus amongst the researchers and then coded “in vivo” using participants’ own words. As patterns emerged in the reassembling of data and coding, recurrent themes were identified to enable thematic analysis [38]. A coding tree was not created, as software was not used for this component of the analysis.\nReflexivity and reduction of potential researcher bias was identified and considered throughout the interview and analysis processes [38]. The midwife researchers reflected on and acknowledged both the potential bias of being midwives investigating a topic they may have a self-interest in, along with the benefits of improved engagement from participants as they were known colleagues.\nTo promote rigor and dependability in the study findings, a second round of analysis was conducted [39–41]. Peer checking was undertaken independently (by CK) through analysis of the de-identified research transcripts using Leximancer V4. Leximancer is a well-known text mining software used to identify concepts grounded in the study data [39] and has been used in Australian maternity settings [42, 43]. Peer checking using a text mining software reduces inadvertent researcher bias and provides another level of dependability within the study findings. The second analysis was compared with themes from first round analysis thus establishing findings across three researchers using two methods, and substantiating trustworthiness in the study [44]. If there were differences in manual and computer analysis results, the research team planned to reach a consensus on emerging themes through discussion. Themes were then mapped to CFIR domains and constructs within each domain to finalise the analysis. Key phrases and meaning from interview data were used to allocate themes to constructs.", "Of the 40 people invited to an interview, 31 consented to participate (77.5%) with no response received from 9 others. No additional people contacted the study team to offer their involvement. Twenty individual or small group interviews were conducted. Data saturation was achieved following 10 interviews however, the research team elected to continue data collection due to many stakeholders wishing to participate. Most participants were female (87%) and between 41 and 50 years of age (35.5%). They were predominantly permanent employees (74.2%) and had more than 10 years’ experience (70.9%) (Table 2). Participant characteristics generally aligned with that of the Australian health workforce, with most participants being nurses and midwives (Australian Institute of Health and Welfare, 2020). However, more people aged 41 and over participated in the research, whereas the comparable Australian workforce is mostly aged 20 to 34 years.\n\nTable 2Characteristics of participants [N = 31]Participant characteristicn (%)Age20–3031–4041–5051–6061+2 (6.5)6 (19.4)11 (35.5)9 (29)3 (9.6)GenderMaleFemaleNot stated3 (9.7)27 (87)1 (3.2)Stakeholder roleMidwifeMedical OfficerNurseNurse/Midwife LeaderAllied Health PractitionerOther (Business Representative, Administration Officer, Consumer)13 (41.9)5 (16.1)6 (19.4)3 (9.7)1 (3.2)3 (9.7)Work hoursFull timePart timeNot stated15 (48.4)13 (41.9)3 (9.7)Employment statusTemporary appointmentPermanent appointmentNot stated1 (3.2)23 (74.2)7 (22.6)Professional experience*< 5 years5–10 years10 + yearsNot stated07 (22.6)22 (70.9)1 (6.5)*The consumer representative was not included in this group, thus n = 30\n\nCharacteristics of participants [N = 31]\nAge\n20–30\n31–40\n41–50\n51–60\n61+\n2 (6.5)\n6 (19.4)\n11 (35.5)\n9 (29)\n3 (9.6)\nGender\nMale\nFemale\nNot stated\n3 (9.7)\n27 (87)\n1 (3.2)\nStakeholder role\nMidwife\nMedical Officer\nNurse\nNurse/Midwife Leader\nAllied Health Practitioner\nOther (Business Representative, Administration Officer, Consumer)\n13 (41.9)\n5 (16.1)\n6 (19.4)\n3 (9.7)\n1 (3.2)\n3 (9.7)\nWork hours\nFull time\nPart time\nNot stated\n15 (48.4)\n13 (41.9)\n3 (9.7)\nEmployment status\nTemporary appointment\nPermanent appointment\nNot stated\n1 (3.2)\n23 (74.2)\n7 (22.6)\nProfessional experience*\n< 5 years\n5–10 years\n10 + years\nNot stated\n0\n7 (22.6)\n22 (70.9)\n1 (6.5)\n*The consumer representative was not included in this group, thus n = 30\nPotential barriers and enablers were identified from the interview data and grouped into themes: the woman’s experience, midwifery workforce, gold standard care, the interdisciplinary team, and costs. These themes function as ways in which we have organised and expressed the barriers and enablers. A summary of potential barriers and enablers from which these themes emerged is presented in Supplementary File 1.\n[SUBTITLE] The woman’s experience [SUBSECTION] Discussion around the woman’s experience identified enablers for the proposed model. Many participants expressed empathy for the likely patient cohort and placed the proposed model as a high priority for the health service. Enablers included perceptions of improved clinical outcomes, building of safety and trust, patient satisfaction and the potential to reduce ‘fail to attend’ rates at the specialty clinic. This is supported by comments from participants:\n\n… vulnerable or disadvantaged groups would benefit … (Other role, Interview 1).\n… some with abuse histories don’t want to go over those histories over and over\nagain…some come with diverse cultural situations and specific needs … (Midwife, Interview 2).… they can build up trust, they can have the tough talk with a familiar face … (Nurse, Interview 12).\n\n… vulnerable or disadvantaged groups would benefit … (Other role, Interview 1).\n\n… some with abuse histories don’t want to go over those histories over and over\n\nagain…some come with diverse cultural situations and specific needs … (Midwife, Interview 2).\n… they can build up trust, they can have the tough talk with a familiar face … (Nurse, Interview 12).\nThe general sentiment expressed by many participants is captured by this statement:\n\n…the patient can build a rapport and have trust with the clinicians…. It not only makes the patient feel safe and more comfortable with their surroundings, in my opinion, it makes the patient a bit more accountable and builds rapport with that clinician and women would be more engaged to come back… (Other role, Interview 1).\n\n…the patient can build a rapport and have trust with the clinicians…. It not only makes the patient feel safe and more comfortable with their surroundings, in my opinion, it makes the patient a bit more accountable and builds rapport with that clinician and women would be more engaged to come back… (Other role, Interview 1).\nThe proposed model was being viewed as positive and with a woman-centred focus. These views become potential enablers when preparing detailed business cases that demonstrate strategic and strong stakeholder support as well as alignment with evidence and policy advocating woman-centred care.\nHowever, potential barriers centred around concerns that women may disengage if they did not bond with the known midwife, were socially isolated or feared being reported to child safety services:\n\nFor the women it’s positive all around, unless they felt they couldn’t engage with the midwife, which could lead to the women disengaging completely. (Midwife, Interview 6)The women can be fearful about opening up…that there may be negative consequences… (Midwife, Interview 10).\n\nFor the women it’s positive all around, unless they felt they couldn’t engage with the midwife, which could lead to the women disengaging completely. (Midwife, Interview 6)\nThe women can be fearful about opening up…that there may be negative consequences… (Midwife, Interview 10).\nParticipants also suggested ways to address the woman’s disengagement through awareness and actions by the midwives:\n\nWe have to be open and transparent…so there’s a consistent approach and that we can be strong and recognise it when it (disengagement) happens and maintain the relationship … (Midwife, Interview 10).… the midwife would need to be capable of referring… to those that can help … (Midwife, Interview 16).… if that relationship isn’t working there could be space to swap … (Other role, Interview 17).\n\nWe have to be open and transparent…so there’s a consistent approach and that we can be strong and recognise it when it (disengagement) happens and maintain the relationship … (Midwife, Interview 10).\n… the midwife would need to be capable of referring… to those that can help … (Midwife, Interview 16).\n… if that relationship isn’t working there could be space to swap … (Other role, Interview 17).\nWhile women’s disengagement from the proposed model might be identified as a risk, in discussions around this barrier solutions were identified. Such solutions would inform risk mitigation.\nDiscussion around the woman’s experience identified enablers for the proposed model. Many participants expressed empathy for the likely patient cohort and placed the proposed model as a high priority for the health service. Enablers included perceptions of improved clinical outcomes, building of safety and trust, patient satisfaction and the potential to reduce ‘fail to attend’ rates at the specialty clinic. This is supported by comments from participants:\n\n… vulnerable or disadvantaged groups would benefit … (Other role, Interview 1).\n… some with abuse histories don’t want to go over those histories over and over\nagain…some come with diverse cultural situations and specific needs … (Midwife, Interview 2).… they can build up trust, they can have the tough talk with a familiar face … (Nurse, Interview 12).\n\n… vulnerable or disadvantaged groups would benefit … (Other role, Interview 1).\n\n… some with abuse histories don’t want to go over those histories over and over\n\nagain…some come with diverse cultural situations and specific needs … (Midwife, Interview 2).\n… they can build up trust, they can have the tough talk with a familiar face … (Nurse, Interview 12).\nThe general sentiment expressed by many participants is captured by this statement:\n\n…the patient can build a rapport and have trust with the clinicians…. It not only makes the patient feel safe and more comfortable with their surroundings, in my opinion, it makes the patient a bit more accountable and builds rapport with that clinician and women would be more engaged to come back… (Other role, Interview 1).\n\n…the patient can build a rapport and have trust with the clinicians…. It not only makes the patient feel safe and more comfortable with their surroundings, in my opinion, it makes the patient a bit more accountable and builds rapport with that clinician and women would be more engaged to come back… (Other role, Interview 1).\nThe proposed model was being viewed as positive and with a woman-centred focus. These views become potential enablers when preparing detailed business cases that demonstrate strategic and strong stakeholder support as well as alignment with evidence and policy advocating woman-centred care.\nHowever, potential barriers centred around concerns that women may disengage if they did not bond with the known midwife, were socially isolated or feared being reported to child safety services:\n\nFor the women it’s positive all around, unless they felt they couldn’t engage with the midwife, which could lead to the women disengaging completely. (Midwife, Interview 6)The women can be fearful about opening up…that there may be negative consequences… (Midwife, Interview 10).\n\nFor the women it’s positive all around, unless they felt they couldn’t engage with the midwife, which could lead to the women disengaging completely. (Midwife, Interview 6)\nThe women can be fearful about opening up…that there may be negative consequences… (Midwife, Interview 10).\nParticipants also suggested ways to address the woman’s disengagement through awareness and actions by the midwives:\n\nWe have to be open and transparent…so there’s a consistent approach and that we can be strong and recognise it when it (disengagement) happens and maintain the relationship … (Midwife, Interview 10).… the midwife would need to be capable of referring… to those that can help … (Midwife, Interview 16).… if that relationship isn’t working there could be space to swap … (Other role, Interview 17).\n\nWe have to be open and transparent…so there’s a consistent approach and that we can be strong and recognise it when it (disengagement) happens and maintain the relationship … (Midwife, Interview 10).\n… the midwife would need to be capable of referring… to those that can help … (Midwife, Interview 16).\n… if that relationship isn’t working there could be space to swap … (Other role, Interview 17).\nWhile women’s disengagement from the proposed model might be identified as a risk, in discussions around this barrier solutions were identified. Such solutions would inform risk mitigation.\n[SUBTITLE] Midwifery workforce [SUBSECTION] Participants saw the proposed new model as an opportunity for midwives to gain new skills and expand their scope of practice which was identified as an enabler:\n\nThere are a few midwives out there currently upskilling themselves and are really passionate and interested and already preparing for being part of the team … (Nurse/Midwife Leader, Interview 3).Midwives will actually see this as a positive move and it’s going to be development for them, and it’s going to be opportunistic for them … (Nurse/Midwife Leader, Interview 4).\n\nThere are a few midwives out there currently upskilling themselves and are really passionate and interested and already preparing for being part of the team … (Nurse/Midwife Leader, Interview 3).\nMidwives will actually see this as a positive move and it’s going to be development for them, and it’s going to be opportunistic for them … (Nurse/Midwife Leader, Interview 4).\nA major workforce barrier to implementation success was how midwifery group practice would place high psychological demand on the midwives leading to burnout or vicarious trauma:\n\n… very complex women with personality disorders and high psychological needs and that could be quite demanding of one midwife as the primary caregiver … (Nurse/Midwife Leader, Interview 3).… to field all those phone calls and constantly support that person would be really challenging … (Nurse/Midwife Leader, Interview 3).For the midwife dealing with only these women, it could over time be mentally challenging … potentially exhausting and tiring … (Midwife, Interview 6).\n\n… very complex women with personality disorders and high psychological needs and that could be quite demanding of one midwife as the primary caregiver … (Nurse/Midwife Leader, Interview 3).\n… to field all those phone calls and constantly support that person would be really challenging … (Nurse/Midwife Leader, Interview 3).\nFor the midwife dealing with only these women, it could over time be mentally challenging … potentially exhausting and tiring … (Midwife, Interview 6).\nThe difficulty in attracting midwives to the proposed model of care was an identified barrier to implementing a high standard of care:\n\n… attracting the midwives that would have an interest in it, we almost need a mother-like figure…would have to be resilient and have had a few more life experiences … (Medical Officer, Interview 13).\n\n… attracting the midwives that would have an interest in it, we almost need a mother-like figure…would have to be resilient and have had a few more life experiences … (Medical Officer, Interview 13).\nWhile this second barrier is contradictory to the initial workforce enabler identified in this theme, participants became solution focussed in the interviews, which is reflected in the theme reported below ‘The interdisciplinary team’.\nParticipants saw the proposed new model as an opportunity for midwives to gain new skills and expand their scope of practice which was identified as an enabler:\n\nThere are a few midwives out there currently upskilling themselves and are really passionate and interested and already preparing for being part of the team … (Nurse/Midwife Leader, Interview 3).Midwives will actually see this as a positive move and it’s going to be development for them, and it’s going to be opportunistic for them … (Nurse/Midwife Leader, Interview 4).\n\nThere are a few midwives out there currently upskilling themselves and are really passionate and interested and already preparing for being part of the team … (Nurse/Midwife Leader, Interview 3).\nMidwives will actually see this as a positive move and it’s going to be development for them, and it’s going to be opportunistic for them … (Nurse/Midwife Leader, Interview 4).\nA major workforce barrier to implementation success was how midwifery group practice would place high psychological demand on the midwives leading to burnout or vicarious trauma:\n\n… very complex women with personality disorders and high psychological needs and that could be quite demanding of one midwife as the primary caregiver … (Nurse/Midwife Leader, Interview 3).… to field all those phone calls and constantly support that person would be really challenging … (Nurse/Midwife Leader, Interview 3).For the midwife dealing with only these women, it could over time be mentally challenging … potentially exhausting and tiring … (Midwife, Interview 6).\n\n… very complex women with personality disorders and high psychological needs and that could be quite demanding of one midwife as the primary caregiver … (Nurse/Midwife Leader, Interview 3).\n… to field all those phone calls and constantly support that person would be really challenging … (Nurse/Midwife Leader, Interview 3).\nFor the midwife dealing with only these women, it could over time be mentally challenging … potentially exhausting and tiring … (Midwife, Interview 6).\nThe difficulty in attracting midwives to the proposed model of care was an identified barrier to implementing a high standard of care:\n\n… attracting the midwives that would have an interest in it, we almost need a mother-like figure…would have to be resilient and have had a few more life experiences … (Medical Officer, Interview 13).\n\n… attracting the midwives that would have an interest in it, we almost need a mother-like figure…would have to be resilient and have had a few more life experiences … (Medical Officer, Interview 13).\nWhile this second barrier is contradictory to the initial workforce enabler identified in this theme, participants became solution focussed in the interviews, which is reflected in the theme reported below ‘The interdisciplinary team’.\n[SUBTITLE] Gold standard care [SUBSECTION] The belief amongst participants of strong, quality evidence in favour of the proposed model was identified as an enabler, and there were no barriers highlighted by participants in terms of available evidence. Many participants identified through discussion around evidence that a midwifery group practice for vulnerable women would be a positive step for the women involved. This was particularly evident for participants who had undertaken reading prior to the interview with participants expressing:\n\nI have looked online to have a look at the research… shows the best outcome for babies and mothers across the board in terms of continuity of care models … (Other role, Interview 1).MGP is gold standard and… there’s lots of research out there that shows that continuity of care is best for these women to develop a relationship … (Midwife, Interview 2).… the people caring for them are more likely to pick up on deviations from a normal emotional state … (Midwife, Interview 2).\n\nI have looked online to have a look at the research… shows the best outcome for babies and mothers across the board in terms of continuity of care models … (Other role, Interview 1).\nMGP is gold standard and… there’s lots of research out there that shows that continuity of care is best for these women to develop a relationship … (Midwife, Interview 2).\n… the people caring for them are more likely to pick up on deviations from a normal emotional state … (Midwife, Interview 2).\nSome participants had undertaken self-directed reading to source additional information regarding midwifery group practice and the needs of vulnerable women. Whilst participants were sent relevant information containing brief background to the proposed study in advance of the interview, some attendees advised they wanted to come well prepared. For example, two participants advised the researchers that they were very grateful to have been invited to interview and were now more aware of the benefits of midwifery group practice for women including that this care was “gold standard”. The integral way in which having a known midwife provides benefit and support for women was repeatedly discussed by participants. For many, the perception from the evidence that a midwifery group practice for vulnerable women is “gold standard” was the sole driver for supporting the proposed model and was seen as a strong enabler which would enhance a business case for the proposed model.\nThe belief amongst participants of strong, quality evidence in favour of the proposed model was identified as an enabler, and there were no barriers highlighted by participants in terms of available evidence. Many participants identified through discussion around evidence that a midwifery group practice for vulnerable women would be a positive step for the women involved. This was particularly evident for participants who had undertaken reading prior to the interview with participants expressing:\n\nI have looked online to have a look at the research… shows the best outcome for babies and mothers across the board in terms of continuity of care models … (Other role, Interview 1).MGP is gold standard and… there’s lots of research out there that shows that continuity of care is best for these women to develop a relationship … (Midwife, Interview 2).… the people caring for them are more likely to pick up on deviations from a normal emotional state … (Midwife, Interview 2).\n\nI have looked online to have a look at the research… shows the best outcome for babies and mothers across the board in terms of continuity of care models … (Other role, Interview 1).\nMGP is gold standard and… there’s lots of research out there that shows that continuity of care is best for these women to develop a relationship … (Midwife, Interview 2).\n… the people caring for them are more likely to pick up on deviations from a normal emotional state … (Midwife, Interview 2).\nSome participants had undertaken self-directed reading to source additional information regarding midwifery group practice and the needs of vulnerable women. Whilst participants were sent relevant information containing brief background to the proposed study in advance of the interview, some attendees advised they wanted to come well prepared. For example, two participants advised the researchers that they were very grateful to have been invited to interview and were now more aware of the benefits of midwifery group practice for women including that this care was “gold standard”. The integral way in which having a known midwife provides benefit and support for women was repeatedly discussed by participants. For many, the perception from the evidence that a midwifery group practice for vulnerable women is “gold standard” was the sole driver for supporting the proposed model and was seen as a strong enabler which would enhance a business case for the proposed model.\n[SUBTITLE] The interdisciplinary team [SUBSECTION] Support for the inclusion of an interdisciplinary team in the proposed model of care was identified as an enabler and was particularly supported by non-midwife participants:\n\n… I would love it, because we felt [from observation of previous staffing arrangements] that when the midwives were staying in the role longer it felt very organised and that we knew the patients very well … (Allied Health Practitioner, Interview 9).There’s also the opportunity to develop an interdisciplinary trust … (Nurse, Interview 14).\n\n… I would love it, because we felt [from observation of previous staffing arrangements] that when the midwives were staying in the role longer it felt very organised and that we knew the patients very well … (Allied Health Practitioner, Interview 9).\nThere’s also the opportunity to develop an interdisciplinary trust … (Nurse, Interview 14).\nMidwife participants felt similarly:\n\nNeed a multi-disciplinary team … and.“… still having that multi-disciplinary approach… is fantastic … (Midwife, Interview 11).\n\nNeed a multi-disciplinary team … and.\n“… still having that multi-disciplinary approach… is fantastic … (Midwife, Interview 11).\nOther responses emphasised the importance of midwives working with other disciplines and not practising in isolation:\n\nIt’s really beneficial having a whole team caring for them (Midwife, Interview 2) …and.… continuing to have the multi-disciplinary input and multi-disciplinary team meeting is going to be beneficial … (Medical Officer, Interview 20).\n\nIt’s really beneficial having a whole team caring for them (Midwife, Interview 2) …and.\n… continuing to have the multi-disciplinary input and multi-disciplinary team meeting is going to be beneficial … (Medical Officer, Interview 20).\nOthers commented:\n\n… it’s an addition to the multi-disciplinary teams, so it doesn’t take anything away but they’ve got some-one they can trust that follows them through all the way … (Medical Officer, Interview 8).… the midwives … would need to be engaged with the multi-disciplinary team more than … MGP’s … (Other role, Interview 17).\n\n… it’s an addition to the multi-disciplinary teams, so it doesn’t take anything away but they’ve got some-one they can trust that follows them through all the way … (Medical Officer, Interview 8).\n… the midwives … would need to be engaged with the multi-disciplinary team more than … MGP’s … (Other role, Interview 17).\nThe overall sentiment across disciplines was that:\n\nEveryone’s ready for a change in the space and a growth in the space and how we can improve for the women and I think it would be highly supported, valued and everyone would be on board … (Nurse/Midwife Leader, Interview 3).\n\nEveryone’s ready for a change in the space and a growth in the space and how we can improve for the women and I think it would be highly supported, valued and everyone would be on board … (Nurse/Midwife Leader, Interview 3).\nAn interdisciplinary team was therefore an important and well supported component of the model, and no barriers to including an inter-disciplinary team in the model were identified. Participants were clear that midwives would lead continuity of care, while having expert health professionals involved to provide comprehensive care for the women. They would also provide professional support for midwives who might feel isolated in their role. These design details can be included in a business case to ensure successful and sustained implementation.\nSupport for the inclusion of an interdisciplinary team in the proposed model of care was identified as an enabler and was particularly supported by non-midwife participants:\n\n… I would love it, because we felt [from observation of previous staffing arrangements] that when the midwives were staying in the role longer it felt very organised and that we knew the patients very well … (Allied Health Practitioner, Interview 9).There’s also the opportunity to develop an interdisciplinary trust … (Nurse, Interview 14).\n\n… I would love it, because we felt [from observation of previous staffing arrangements] that when the midwives were staying in the role longer it felt very organised and that we knew the patients very well … (Allied Health Practitioner, Interview 9).\nThere’s also the opportunity to develop an interdisciplinary trust … (Nurse, Interview 14).\nMidwife participants felt similarly:\n\nNeed a multi-disciplinary team … and.“… still having that multi-disciplinary approach… is fantastic … (Midwife, Interview 11).\n\nNeed a multi-disciplinary team … and.\n“… still having that multi-disciplinary approach… is fantastic … (Midwife, Interview 11).\nOther responses emphasised the importance of midwives working with other disciplines and not practising in isolation:\n\nIt’s really beneficial having a whole team caring for them (Midwife, Interview 2) …and.… continuing to have the multi-disciplinary input and multi-disciplinary team meeting is going to be beneficial … (Medical Officer, Interview 20).\n\nIt’s really beneficial having a whole team caring for them (Midwife, Interview 2) …and.\n… continuing to have the multi-disciplinary input and multi-disciplinary team meeting is going to be beneficial … (Medical Officer, Interview 20).\nOthers commented:\n\n… it’s an addition to the multi-disciplinary teams, so it doesn’t take anything away but they’ve got some-one they can trust that follows them through all the way … (Medical Officer, Interview 8).… the midwives … would need to be engaged with the multi-disciplinary team more than … MGP’s … (Other role, Interview 17).\n\n… it’s an addition to the multi-disciplinary teams, so it doesn’t take anything away but they’ve got some-one they can trust that follows them through all the way … (Medical Officer, Interview 8).\n… the midwives … would need to be engaged with the multi-disciplinary team more than … MGP’s … (Other role, Interview 17).\nThe overall sentiment across disciplines was that:\n\nEveryone’s ready for a change in the space and a growth in the space and how we can improve for the women and I think it would be highly supported, valued and everyone would be on board … (Nurse/Midwife Leader, Interview 3).\n\nEveryone’s ready for a change in the space and a growth in the space and how we can improve for the women and I think it would be highly supported, valued and everyone would be on board … (Nurse/Midwife Leader, Interview 3).\nAn interdisciplinary team was therefore an important and well supported component of the model, and no barriers to including an inter-disciplinary team in the model were identified. Participants were clear that midwives would lead continuity of care, while having expert health professionals involved to provide comprehensive care for the women. They would also provide professional support for midwives who might feel isolated in their role. These design details can be included in a business case to ensure successful and sustained implementation.\n[SUBTITLE] Costs [SUBSECTION] Participants believed that the health benefits of the proposed model of care would outweigh the perception that a midwifery group practice for vulnerable women was a more expensive model of care. This belief was a clear enabler of the proposed model:\n\n… the cost would come with great reward … (Nurse/Midwife Leader, Interview 3).… there will be extra cost, but the trade off in terms of good follow up might not save money but it will be money well spent … (Medical Officer, Interview 8).\n\n… the cost would come with great reward … (Nurse/Midwife Leader, Interview 3).\n… there will be extra cost, but the trade off in terms of good follow up might not save money but it will be money well spent … (Medical Officer, Interview 8).\nHowever, participants did express concern at the perceived increased need for resources and challenges with attributing costs to a range of clinical areas. The concern focussed around the potential impact on other teams which was seen as a barrier:\n\nI think there is resistance…they see this is going to be taking away from their skill mix and FTE (Full Time Equivalent) … (Nurse/Midwife Leader, Interview 4).\n\nI think there is resistance…they see this is going to be taking away from their skill mix and FTE (Full Time Equivalent) … (Nurse/Midwife Leader, Interview 4).\nHowever, other participants believed that these perceptions and challenges could be overcome and that the proposed model of care should be a priority for the hospital:\n\n… yes there are financial implications and barriers, there doesn’t seem to be good evidence to show why not … (Other role, Interview 1).… yes, it is very important that we generate the activity to get something for the work that we’re doing, but at the end of the day we are looking at patient centred care, so if it’s easier and the best outcome for mother and baby… then that’s what we have to do … (Other role, Interview 1).Why shouldn’t they have an MGP, they shouldn’t be excluded just because they’ve had drug and alcohol or mental health issues in the past … (Nurse, Interview 15).I see it as high priority to look at how we can increase activity and treat this vulnerable group … (Nurse/Midwife Leader, Interview 4).\n\n… yes there are financial implications and barriers, there doesn’t seem to be good evidence to show why not … (Other role, Interview 1).\n… yes, it is very important that we generate the activity to get something for the work that we’re doing, but at the end of the day we are looking at patient centred care, so if it’s easier and the best outcome for mother and baby… then that’s what we have to do … (Other role, Interview 1).\nWhy shouldn’t they have an MGP, they shouldn’t be excluded just because they’ve had drug and alcohol or mental health issues in the past … (Nurse, Interview 15).\nI see it as high priority to look at how we can increase activity and treat this vulnerable group … (Nurse/Midwife Leader, Interview 4).\nPerceptions around cost that are both potential enablers and barriers to gaining support and successful implementation would need to be clarified as fact in a business case before the proposed model is implemented. Participants had concerns around costs which may be reflected by decision-makers when examining the rigor in which a business case has been prepared. This may also influence future expansion of midwifery group practice as a model of care more generally.\nIn the computer-assisted analysis, five themes were identified from the data: women with complex care: specialty clinic; continuity of care; workforce; and opportunity (Fig. 1). Themes and sub-themes similar to those that emerged from manual analysis were around the woman’s experience, workforce, standards of care and opportunity.\n\nFig. 1Leximancer 4™ generated themes from stakeholder interviews\n\nLeximancer 4™ generated themes from stakeholder interviews\nTable 3 shows the mapped overarching themes and how they relate to the five CFIR domains and constructs. In this table it becomes clear what the requirements for successful and sustained implementation of the proposed model might be. The theme Gold standard care mapped to five constructs indicates that either a perception, or knowledge of evidence supporting midwifery group practice for vulnerable women would be critical to its success. A thorough consideration of potential experiences for the woman, the workforce and costs when preparing a business case, will be a determinant of model implementation success. An interdisciplinary team that is already part of the organisational structure and engaged in planning the model is likely to be essential. Processes that demonstrate evidence of planning and reflecting across all CFIR domains, especially regarding linkages between different health professional disciplines and costs, are also important.\n\nTable 3Summary of interview themes mapped to Consolidated Framework for Implementation Research (CFIR) domains and constructsCFIR domains and constructsThemes\nIntervention characteristics\nEvidence strength and qualityGold standard careRelative advantageThe woman’s experienceMidwifery workforceCostsCostCostsAdaptabilityMidwifery workforce\nOuter setting\nPatient needs and resourcesThe woman’s experiencePeer PressureGold standard care\nInner setting\nStructural characteristicsThe interdisciplinary teamImplementation climateMidwifery workforceRelative prioritiesGold standard careThe woman’s experience\nCharacteristics of individuals\nKnowledge and beliefs about the interventionGold standard careIndividual stage of changeGold standard careSelf-efficacyMidwifery workforce\nProcess\nPlanningThe interdisciplinary teamReflecting and evaluatingCosts\n\nSummary of interview themes mapped to Consolidated Framework for Implementation Research (CFIR) domains and constructs\nThe woman’s experience\nMidwifery workforce\nCosts\nGold standard care\nThe woman’s experience\nParticipants believed that the health benefits of the proposed model of care would outweigh the perception that a midwifery group practice for vulnerable women was a more expensive model of care. This belief was a clear enabler of the proposed model:\n\n… the cost would come with great reward … (Nurse/Midwife Leader, Interview 3).… there will be extra cost, but the trade off in terms of good follow up might not save money but it will be money well spent … (Medical Officer, Interview 8).\n\n… the cost would come with great reward … (Nurse/Midwife Leader, Interview 3).\n… there will be extra cost, but the trade off in terms of good follow up might not save money but it will be money well spent … (Medical Officer, Interview 8).\nHowever, participants did express concern at the perceived increased need for resources and challenges with attributing costs to a range of clinical areas. The concern focussed around the potential impact on other teams which was seen as a barrier:\n\nI think there is resistance…they see this is going to be taking away from their skill mix and FTE (Full Time Equivalent) … (Nurse/Midwife Leader, Interview 4).\n\nI think there is resistance…they see this is going to be taking away from their skill mix and FTE (Full Time Equivalent) … (Nurse/Midwife Leader, Interview 4).\nHowever, other participants believed that these perceptions and challenges could be overcome and that the proposed model of care should be a priority for the hospital:\n\n… yes there are financial implications and barriers, there doesn’t seem to be good evidence to show why not … (Other role, Interview 1).… yes, it is very important that we generate the activity to get something for the work that we’re doing, but at the end of the day we are looking at patient centred care, so if it’s easier and the best outcome for mother and baby… then that’s what we have to do … (Other role, Interview 1).Why shouldn’t they have an MGP, they shouldn’t be excluded just because they’ve had drug and alcohol or mental health issues in the past … (Nurse, Interview 15).I see it as high priority to look at how we can increase activity and treat this vulnerable group … (Nurse/Midwife Leader, Interview 4).\n\n… yes there are financial implications and barriers, there doesn’t seem to be good evidence to show why not … (Other role, Interview 1).\n… yes, it is very important that we generate the activity to get something for the work that we’re doing, but at the end of the day we are looking at patient centred care, so if it’s easier and the best outcome for mother and baby… then that’s what we have to do … (Other role, Interview 1).\nWhy shouldn’t they have an MGP, they shouldn’t be excluded just because they’ve had drug and alcohol or mental health issues in the past … (Nurse, Interview 15).\nI see it as high priority to look at how we can increase activity and treat this vulnerable group … (Nurse/Midwife Leader, Interview 4).\nPerceptions around cost that are both potential enablers and barriers to gaining support and successful implementation would need to be clarified as fact in a business case before the proposed model is implemented. Participants had concerns around costs which may be reflected by decision-makers when examining the rigor in which a business case has been prepared. This may also influence future expansion of midwifery group practice as a model of care more generally.\nIn the computer-assisted analysis, five themes were identified from the data: women with complex care: specialty clinic; continuity of care; workforce; and opportunity (Fig. 1). Themes and sub-themes similar to those that emerged from manual analysis were around the woman’s experience, workforce, standards of care and opportunity.\n\nFig. 1Leximancer 4™ generated themes from stakeholder interviews\n\nLeximancer 4™ generated themes from stakeholder interviews\nTable 3 shows the mapped overarching themes and how they relate to the five CFIR domains and constructs. In this table it becomes clear what the requirements for successful and sustained implementation of the proposed model might be. The theme Gold standard care mapped to five constructs indicates that either a perception, or knowledge of evidence supporting midwifery group practice for vulnerable women would be critical to its success. A thorough consideration of potential experiences for the woman, the workforce and costs when preparing a business case, will be a determinant of model implementation success. An interdisciplinary team that is already part of the organisational structure and engaged in planning the model is likely to be essential. Processes that demonstrate evidence of planning and reflecting across all CFIR domains, especially regarding linkages between different health professional disciplines and costs, are also important.\n\nTable 3Summary of interview themes mapped to Consolidated Framework for Implementation Research (CFIR) domains and constructsCFIR domains and constructsThemes\nIntervention characteristics\nEvidence strength and qualityGold standard careRelative advantageThe woman’s experienceMidwifery workforceCostsCostCostsAdaptabilityMidwifery workforce\nOuter setting\nPatient needs and resourcesThe woman’s experiencePeer PressureGold standard care\nInner setting\nStructural characteristicsThe interdisciplinary teamImplementation climateMidwifery workforceRelative prioritiesGold standard careThe woman’s experience\nCharacteristics of individuals\nKnowledge and beliefs about the interventionGold standard careIndividual stage of changeGold standard careSelf-efficacyMidwifery workforce\nProcess\nPlanningThe interdisciplinary teamReflecting and evaluatingCosts\n\nSummary of interview themes mapped to Consolidated Framework for Implementation Research (CFIR) domains and constructs\nThe woman’s experience\nMidwifery workforce\nCosts\nGold standard care\nThe woman’s experience", "Discussion around the woman’s experience identified enablers for the proposed model. Many participants expressed empathy for the likely patient cohort and placed the proposed model as a high priority for the health service. Enablers included perceptions of improved clinical outcomes, building of safety and trust, patient satisfaction and the potential to reduce ‘fail to attend’ rates at the specialty clinic. This is supported by comments from participants:\n\n… vulnerable or disadvantaged groups would benefit … (Other role, Interview 1).\n… some with abuse histories don’t want to go over those histories over and over\nagain…some come with diverse cultural situations and specific needs … (Midwife, Interview 2).… they can build up trust, they can have the tough talk with a familiar face … (Nurse, Interview 12).\n\n… vulnerable or disadvantaged groups would benefit … (Other role, Interview 1).\n\n… some with abuse histories don’t want to go over those histories over and over\n\nagain…some come with diverse cultural situations and specific needs … (Midwife, Interview 2).\n… they can build up trust, they can have the tough talk with a familiar face … (Nurse, Interview 12).\nThe general sentiment expressed by many participants is captured by this statement:\n\n…the patient can build a rapport and have trust with the clinicians…. It not only makes the patient feel safe and more comfortable with their surroundings, in my opinion, it makes the patient a bit more accountable and builds rapport with that clinician and women would be more engaged to come back… (Other role, Interview 1).\n\n…the patient can build a rapport and have trust with the clinicians…. It not only makes the patient feel safe and more comfortable with their surroundings, in my opinion, it makes the patient a bit more accountable and builds rapport with that clinician and women would be more engaged to come back… (Other role, Interview 1).\nThe proposed model was being viewed as positive and with a woman-centred focus. These views become potential enablers when preparing detailed business cases that demonstrate strategic and strong stakeholder support as well as alignment with evidence and policy advocating woman-centred care.\nHowever, potential barriers centred around concerns that women may disengage if they did not bond with the known midwife, were socially isolated or feared being reported to child safety services:\n\nFor the women it’s positive all around, unless they felt they couldn’t engage with the midwife, which could lead to the women disengaging completely. (Midwife, Interview 6)The women can be fearful about opening up…that there may be negative consequences… (Midwife, Interview 10).\n\nFor the women it’s positive all around, unless they felt they couldn’t engage with the midwife, which could lead to the women disengaging completely. (Midwife, Interview 6)\nThe women can be fearful about opening up…that there may be negative consequences… (Midwife, Interview 10).\nParticipants also suggested ways to address the woman’s disengagement through awareness and actions by the midwives:\n\nWe have to be open and transparent…so there’s a consistent approach and that we can be strong and recognise it when it (disengagement) happens and maintain the relationship … (Midwife, Interview 10).… the midwife would need to be capable of referring… to those that can help … (Midwife, Interview 16).… if that relationship isn’t working there could be space to swap … (Other role, Interview 17).\n\nWe have to be open and transparent…so there’s a consistent approach and that we can be strong and recognise it when it (disengagement) happens and maintain the relationship … (Midwife, Interview 10).\n… the midwife would need to be capable of referring… to those that can help … (Midwife, Interview 16).\n… if that relationship isn’t working there could be space to swap … (Other role, Interview 17).\nWhile women’s disengagement from the proposed model might be identified as a risk, in discussions around this barrier solutions were identified. Such solutions would inform risk mitigation.", "Participants saw the proposed new model as an opportunity for midwives to gain new skills and expand their scope of practice which was identified as an enabler:\n\nThere are a few midwives out there currently upskilling themselves and are really passionate and interested and already preparing for being part of the team … (Nurse/Midwife Leader, Interview 3).Midwives will actually see this as a positive move and it’s going to be development for them, and it’s going to be opportunistic for them … (Nurse/Midwife Leader, Interview 4).\n\nThere are a few midwives out there currently upskilling themselves and are really passionate and interested and already preparing for being part of the team … (Nurse/Midwife Leader, Interview 3).\nMidwives will actually see this as a positive move and it’s going to be development for them, and it’s going to be opportunistic for them … (Nurse/Midwife Leader, Interview 4).\nA major workforce barrier to implementation success was how midwifery group practice would place high psychological demand on the midwives leading to burnout or vicarious trauma:\n\n… very complex women with personality disorders and high psychological needs and that could be quite demanding of one midwife as the primary caregiver … (Nurse/Midwife Leader, Interview 3).… to field all those phone calls and constantly support that person would be really challenging … (Nurse/Midwife Leader, Interview 3).For the midwife dealing with only these women, it could over time be mentally challenging … potentially exhausting and tiring … (Midwife, Interview 6).\n\n… very complex women with personality disorders and high psychological needs and that could be quite demanding of one midwife as the primary caregiver … (Nurse/Midwife Leader, Interview 3).\n… to field all those phone calls and constantly support that person would be really challenging … (Nurse/Midwife Leader, Interview 3).\nFor the midwife dealing with only these women, it could over time be mentally challenging … potentially exhausting and tiring … (Midwife, Interview 6).\nThe difficulty in attracting midwives to the proposed model of care was an identified barrier to implementing a high standard of care:\n\n… attracting the midwives that would have an interest in it, we almost need a mother-like figure…would have to be resilient and have had a few more life experiences … (Medical Officer, Interview 13).\n\n… attracting the midwives that would have an interest in it, we almost need a mother-like figure…would have to be resilient and have had a few more life experiences … (Medical Officer, Interview 13).\nWhile this second barrier is contradictory to the initial workforce enabler identified in this theme, participants became solution focussed in the interviews, which is reflected in the theme reported below ‘The interdisciplinary team’.", "The belief amongst participants of strong, quality evidence in favour of the proposed model was identified as an enabler, and there were no barriers highlighted by participants in terms of available evidence. Many participants identified through discussion around evidence that a midwifery group practice for vulnerable women would be a positive step for the women involved. This was particularly evident for participants who had undertaken reading prior to the interview with participants expressing:\n\nI have looked online to have a look at the research… shows the best outcome for babies and mothers across the board in terms of continuity of care models … (Other role, Interview 1).MGP is gold standard and… there’s lots of research out there that shows that continuity of care is best for these women to develop a relationship … (Midwife, Interview 2).… the people caring for them are more likely to pick up on deviations from a normal emotional state … (Midwife, Interview 2).\n\nI have looked online to have a look at the research… shows the best outcome for babies and mothers across the board in terms of continuity of care models … (Other role, Interview 1).\nMGP is gold standard and… there’s lots of research out there that shows that continuity of care is best for these women to develop a relationship … (Midwife, Interview 2).\n… the people caring for them are more likely to pick up on deviations from a normal emotional state … (Midwife, Interview 2).\nSome participants had undertaken self-directed reading to source additional information regarding midwifery group practice and the needs of vulnerable women. Whilst participants were sent relevant information containing brief background to the proposed study in advance of the interview, some attendees advised they wanted to come well prepared. For example, two participants advised the researchers that they were very grateful to have been invited to interview and were now more aware of the benefits of midwifery group practice for women including that this care was “gold standard”. The integral way in which having a known midwife provides benefit and support for women was repeatedly discussed by participants. For many, the perception from the evidence that a midwifery group practice for vulnerable women is “gold standard” was the sole driver for supporting the proposed model and was seen as a strong enabler which would enhance a business case for the proposed model.", "Support for the inclusion of an interdisciplinary team in the proposed model of care was identified as an enabler and was particularly supported by non-midwife participants:\n\n… I would love it, because we felt [from observation of previous staffing arrangements] that when the midwives were staying in the role longer it felt very organised and that we knew the patients very well … (Allied Health Practitioner, Interview 9).There’s also the opportunity to develop an interdisciplinary trust … (Nurse, Interview 14).\n\n… I would love it, because we felt [from observation of previous staffing arrangements] that when the midwives were staying in the role longer it felt very organised and that we knew the patients very well … (Allied Health Practitioner, Interview 9).\nThere’s also the opportunity to develop an interdisciplinary trust … (Nurse, Interview 14).\nMidwife participants felt similarly:\n\nNeed a multi-disciplinary team … and.“… still having that multi-disciplinary approach… is fantastic … (Midwife, Interview 11).\n\nNeed a multi-disciplinary team … and.\n“… still having that multi-disciplinary approach… is fantastic … (Midwife, Interview 11).\nOther responses emphasised the importance of midwives working with other disciplines and not practising in isolation:\n\nIt’s really beneficial having a whole team caring for them (Midwife, Interview 2) …and.… continuing to have the multi-disciplinary input and multi-disciplinary team meeting is going to be beneficial … (Medical Officer, Interview 20).\n\nIt’s really beneficial having a whole team caring for them (Midwife, Interview 2) …and.\n… continuing to have the multi-disciplinary input and multi-disciplinary team meeting is going to be beneficial … (Medical Officer, Interview 20).\nOthers commented:\n\n… it’s an addition to the multi-disciplinary teams, so it doesn’t take anything away but they’ve got some-one they can trust that follows them through all the way … (Medical Officer, Interview 8).… the midwives … would need to be engaged with the multi-disciplinary team more than … MGP’s … (Other role, Interview 17).\n\n… it’s an addition to the multi-disciplinary teams, so it doesn’t take anything away but they’ve got some-one they can trust that follows them through all the way … (Medical Officer, Interview 8).\n… the midwives … would need to be engaged with the multi-disciplinary team more than … MGP’s … (Other role, Interview 17).\nThe overall sentiment across disciplines was that:\n\nEveryone’s ready for a change in the space and a growth in the space and how we can improve for the women and I think it would be highly supported, valued and everyone would be on board … (Nurse/Midwife Leader, Interview 3).\n\nEveryone’s ready for a change in the space and a growth in the space and how we can improve for the women and I think it would be highly supported, valued and everyone would be on board … (Nurse/Midwife Leader, Interview 3).\nAn interdisciplinary team was therefore an important and well supported component of the model, and no barriers to including an inter-disciplinary team in the model were identified. Participants were clear that midwives would lead continuity of care, while having expert health professionals involved to provide comprehensive care for the women. They would also provide professional support for midwives who might feel isolated in their role. These design details can be included in a business case to ensure successful and sustained implementation.", "Participants believed that the health benefits of the proposed model of care would outweigh the perception that a midwifery group practice for vulnerable women was a more expensive model of care. This belief was a clear enabler of the proposed model:\n\n… the cost would come with great reward … (Nurse/Midwife Leader, Interview 3).… there will be extra cost, but the trade off in terms of good follow up might not save money but it will be money well spent … (Medical Officer, Interview 8).\n\n… the cost would come with great reward … (Nurse/Midwife Leader, Interview 3).\n… there will be extra cost, but the trade off in terms of good follow up might not save money but it will be money well spent … (Medical Officer, Interview 8).\nHowever, participants did express concern at the perceived increased need for resources and challenges with attributing costs to a range of clinical areas. The concern focussed around the potential impact on other teams which was seen as a barrier:\n\nI think there is resistance…they see this is going to be taking away from their skill mix and FTE (Full Time Equivalent) … (Nurse/Midwife Leader, Interview 4).\n\nI think there is resistance…they see this is going to be taking away from their skill mix and FTE (Full Time Equivalent) … (Nurse/Midwife Leader, Interview 4).\nHowever, other participants believed that these perceptions and challenges could be overcome and that the proposed model of care should be a priority for the hospital:\n\n… yes there are financial implications and barriers, there doesn’t seem to be good evidence to show why not … (Other role, Interview 1).… yes, it is very important that we generate the activity to get something for the work that we’re doing, but at the end of the day we are looking at patient centred care, so if it’s easier and the best outcome for mother and baby… then that’s what we have to do … (Other role, Interview 1).Why shouldn’t they have an MGP, they shouldn’t be excluded just because they’ve had drug and alcohol or mental health issues in the past … (Nurse, Interview 15).I see it as high priority to look at how we can increase activity and treat this vulnerable group … (Nurse/Midwife Leader, Interview 4).\n\n… yes there are financial implications and barriers, there doesn’t seem to be good evidence to show why not … (Other role, Interview 1).\n… yes, it is very important that we generate the activity to get something for the work that we’re doing, but at the end of the day we are looking at patient centred care, so if it’s easier and the best outcome for mother and baby… then that’s what we have to do … (Other role, Interview 1).\nWhy shouldn’t they have an MGP, they shouldn’t be excluded just because they’ve had drug and alcohol or mental health issues in the past … (Nurse, Interview 15).\nI see it as high priority to look at how we can increase activity and treat this vulnerable group … (Nurse/Midwife Leader, Interview 4).\nPerceptions around cost that are both potential enablers and barriers to gaining support and successful implementation would need to be clarified as fact in a business case before the proposed model is implemented. Participants had concerns around costs which may be reflected by decision-makers when examining the rigor in which a business case has been prepared. This may also influence future expansion of midwifery group practice as a model of care more generally.\nIn the computer-assisted analysis, five themes were identified from the data: women with complex care: specialty clinic; continuity of care; workforce; and opportunity (Fig. 1). Themes and sub-themes similar to those that emerged from manual analysis were around the woman’s experience, workforce, standards of care and opportunity.\n\nFig. 1Leximancer 4™ generated themes from stakeholder interviews\n\nLeximancer 4™ generated themes from stakeholder interviews\nTable 3 shows the mapped overarching themes and how they relate to the five CFIR domains and constructs. In this table it becomes clear what the requirements for successful and sustained implementation of the proposed model might be. The theme Gold standard care mapped to five constructs indicates that either a perception, or knowledge of evidence supporting midwifery group practice for vulnerable women would be critical to its success. A thorough consideration of potential experiences for the woman, the workforce and costs when preparing a business case, will be a determinant of model implementation success. An interdisciplinary team that is already part of the organisational structure and engaged in planning the model is likely to be essential. Processes that demonstrate evidence of planning and reflecting across all CFIR domains, especially regarding linkages between different health professional disciplines and costs, are also important.\n\nTable 3Summary of interview themes mapped to Consolidated Framework for Implementation Research (CFIR) domains and constructsCFIR domains and constructsThemes\nIntervention characteristics\nEvidence strength and qualityGold standard careRelative advantageThe woman’s experienceMidwifery workforceCostsCostCostsAdaptabilityMidwifery workforce\nOuter setting\nPatient needs and resourcesThe woman’s experiencePeer PressureGold standard care\nInner setting\nStructural characteristicsThe interdisciplinary teamImplementation climateMidwifery workforceRelative prioritiesGold standard careThe woman’s experience\nCharacteristics of individuals\nKnowledge and beliefs about the interventionGold standard careIndividual stage of changeGold standard careSelf-efficacyMidwifery workforce\nProcess\nPlanningThe interdisciplinary teamReflecting and evaluatingCosts\n\nSummary of interview themes mapped to Consolidated Framework for Implementation Research (CFIR) domains and constructs\nThe woman’s experience\nMidwifery workforce\nCosts\nGold standard care\nThe woman’s experience", "In this study we have identified, with an interdisciplinary stakeholder perspective, both the potential barriers and enablers that will need to be considered in the next phases of planning and implementing a midwifery group practice for vulnerable women. Participants identified that the proposed model is likely to provide health benefits for women due to the rapport built with a small group of care givers including a supportive interdisciplinary team providing continuity of care. However, participants were conscious of the burden of such a maternity care model on the workforce, both in terms of the emotional challenge due to the women’s complex care requirements and managing the financial cost of the service which would require further evaluation.\nWhen mapping the themes to the CFIR domains, the implications of local results to Australian maternity services became evident. Organising the data in this way also enabled understanding of factors that may lead to successful and sustained implementation of a MGP for vulnerable women internationally as the CFIR has become a universal implementation ‘language’.\n[SUBTITLE] Intervention characteristics [SUBSECTION] Stakeholders perceived that there was high quality and valid evidence supporting midwifery group practice for vulnerable women as “gold standard care” (Midwife, Interview 2). Quality evidence drawn from Australian studies [10, 13] builds confidence amongst stakeholders, which is a strong enabler. Participants weighed the relative advantages of the proposed model of care over the existing care provided to vulnerable women and believed that the health benefits for the women and infants would outweigh the costs. This belief in the net benefit when costs were also considered was not rooted in available evidence, which contrasts with the desire for evidence for “gold standard” effectiveness. The cost of the proposed model had not been established but was imagined by participants to be higher than both the current model of care and comparative midwifery group practices.\nParticipants were mostly clinicians with more than 10 years’ experience and so were likely able to make accurate assumptions around the number of workforce hours required to build rapport with women, discuss the care being delivered with the women and amongst colleagues, and deliver the volume of care required to optimise health outcomes. However, there is no known evidence for the cost-effectiveness of such a niche model of care, only generalised costs reported for Australian midwifery group practice [10, 45]. For Australian maternity services looking to implement a midwifery group practice for vulnerable women, careful understanding of costs and transparent communication to decision-makers is needed. Our research suggests that stakeholders naturally seek evidence for both costs and effectiveness, and in the absence of evidence, local costs should be examined.\nStakeholders perceived that there was high quality and valid evidence supporting midwifery group practice for vulnerable women as “gold standard care” (Midwife, Interview 2). Quality evidence drawn from Australian studies [10, 13] builds confidence amongst stakeholders, which is a strong enabler. Participants weighed the relative advantages of the proposed model of care over the existing care provided to vulnerable women and believed that the health benefits for the women and infants would outweigh the costs. This belief in the net benefit when costs were also considered was not rooted in available evidence, which contrasts with the desire for evidence for “gold standard” effectiveness. The cost of the proposed model had not been established but was imagined by participants to be higher than both the current model of care and comparative midwifery group practices.\nParticipants were mostly clinicians with more than 10 years’ experience and so were likely able to make accurate assumptions around the number of workforce hours required to build rapport with women, discuss the care being delivered with the women and amongst colleagues, and deliver the volume of care required to optimise health outcomes. However, there is no known evidence for the cost-effectiveness of such a niche model of care, only generalised costs reported for Australian midwifery group practice [10, 45]. For Australian maternity services looking to implement a midwifery group practice for vulnerable women, careful understanding of costs and transparent communication to decision-makers is needed. Our research suggests that stakeholders naturally seek evidence for both costs and effectiveness, and in the absence of evidence, local costs should be examined.\n[SUBTITLE] Outer setting [SUBSECTION] A midwifery group practice was perceived to meet the needs of vulnerable women because for example, “some with abuse histories don’t want to go over those histories over and over…” (Midwife, Interview 2). The continuity of midwives would ensure the deep needs of each woman were met. However, concerns were expressed regarding when having a known midwife might be a disincentive for women to engage. Primarily this related to women with involvement of child protection services or times where personal factors impacted on building rapport and a therapeutic relationship was not established between the woman and the midwife. A midwifery group practice for vulnerable women should be designed with flexibility in the case where a rapport is not being established between the care givers and the woman [46]. The service design would need to ensure that midwives can be changed across groups.\nA midwifery group practice was perceived to meet the needs of vulnerable women because for example, “some with abuse histories don’t want to go over those histories over and over…” (Midwife, Interview 2). The continuity of midwives would ensure the deep needs of each woman were met. However, concerns were expressed regarding when having a known midwife might be a disincentive for women to engage. Primarily this related to women with involvement of child protection services or times where personal factors impacted on building rapport and a therapeutic relationship was not established between the woman and the midwife. A midwifery group practice for vulnerable women should be designed with flexibility in the case where a rapport is not being established between the care givers and the woman [46]. The service design would need to ensure that midwives can be changed across groups.\n[SUBTITLE] Inner setting [SUBSECTION] The structural characteristics of the setting for this research were that it is a mature and large maternity service, with a range of existing and well-supported midwifery group practices already established. Consequently, there may have been fewer inner setting barriers to establishing the proposed model of care compared to Australian maternity services in which midwifery group practice is new or not yet established. Further, the interdisciplinary team engaged in this research was supportive of midwifery group practice. Although, it was highlighted that any potential changes to how the team works with the midwifery group practice would need to be implemented carefully. The interdisciplinary team expressed views that to continue safe and effective care, it was important the team’s role be maintained. The implementation climate of the inner setting was generally supportive, and there were some important reflections from the interdisciplinary team that would need to be acknowledged when preparing a business case for the proposed model.\nThe structural characteristics of the setting for this research were that it is a mature and large maternity service, with a range of existing and well-supported midwifery group practices already established. Consequently, there may have been fewer inner setting barriers to establishing the proposed model of care compared to Australian maternity services in which midwifery group practice is new or not yet established. Further, the interdisciplinary team engaged in this research was supportive of midwifery group practice. Although, it was highlighted that any potential changes to how the team works with the midwifery group practice would need to be implemented carefully. The interdisciplinary team expressed views that to continue safe and effective care, it was important the team’s role be maintained. The implementation climate of the inner setting was generally supportive, and there were some important reflections from the interdisciplinary team that would need to be acknowledged when preparing a business case for the proposed model.\n[SUBTITLE] Characteristics of individuals [SUBSECTION] Stakeholders had a positive attitude towards the intervention; they placed a high value on the proposed model of care. Participants even sought evidence in preparation for the interviews and ensured they were familiar with the proposal. This demonstrates that individuals’ knowledge and beliefs about the intervention was a strong enabler for proceeding with the proposal. Further, that demonstrated engagement in the context assessment by stakeholders was an indicator that individuals were at an advanced stage of change in relation to redesigning maternity care for vulnerable women. The identification of this enabler suggests that the initial enthusiasm for the intervention would sustain its implementation over time. The results also demonstrated a moderate level of self-efficacy – there were mixed beliefs amongst individuals in their own capabilities to deliver the model of care, while also identifying that the proposed model of care would provide an opportunity for midwives to build their self-efficacy through gaining new skills and expanding their scope of practice. Modifications would need to be made to traditional midwifery group practice design due to perceived heavy demands from deep engagement with vulnerable women and the potential for vicarious trauma, burnout, and other emotional impacts for the midwives. Participants suggested that to overcome this barrier, caseloads should be reduced, and midwives should be well supervised and mentored as described also by others [46], especially when caring for vulnerable women [47]. To enable such an intervention to be implemented in other Australian maternity services, stakeholders would need to have confidence in their ability to seek and interpret the evidence and have an awareness of the strengths and limitations of the workforce capabilities to execute the proposed model of care.\nStakeholders had a positive attitude towards the intervention; they placed a high value on the proposed model of care. Participants even sought evidence in preparation for the interviews and ensured they were familiar with the proposal. This demonstrates that individuals’ knowledge and beliefs about the intervention was a strong enabler for proceeding with the proposal. Further, that demonstrated engagement in the context assessment by stakeholders was an indicator that individuals were at an advanced stage of change in relation to redesigning maternity care for vulnerable women. The identification of this enabler suggests that the initial enthusiasm for the intervention would sustain its implementation over time. The results also demonstrated a moderate level of self-efficacy – there were mixed beliefs amongst individuals in their own capabilities to deliver the model of care, while also identifying that the proposed model of care would provide an opportunity for midwives to build their self-efficacy through gaining new skills and expanding their scope of practice. Modifications would need to be made to traditional midwifery group practice design due to perceived heavy demands from deep engagement with vulnerable women and the potential for vicarious trauma, burnout, and other emotional impacts for the midwives. Participants suggested that to overcome this barrier, caseloads should be reduced, and midwives should be well supervised and mentored as described also by others [46], especially when caring for vulnerable women [47]. To enable such an intervention to be implemented in other Australian maternity services, stakeholders would need to have confidence in their ability to seek and interpret the evidence and have an awareness of the strengths and limitations of the workforce capabilities to execute the proposed model of care.\n[SUBTITLE] Process [SUBSECTION] Conducting the context assessment instilled confidence in readiness of the maternity service to adopt the proposed change. The context assessment was conducted early in the planning stages of the intervention and the strong engagement suggested interest in the intervention and acceptance of the planning methods. This enabler resulted in positive and open communication and was an unintended consequence of the context assessment, as participation from a large and broad range of disciplines was not expected.\nConducting the context assessment instilled confidence in readiness of the maternity service to adopt the proposed change. The context assessment was conducted early in the planning stages of the intervention and the strong engagement suggested interest in the intervention and acceptance of the planning methods. This enabler resulted in positive and open communication and was an unintended consequence of the context assessment, as participation from a large and broad range of disciplines was not expected.\n[SUBTITLE] Strengths [SUBSECTION] A strength of this study was the use of the CFIR to guide interviews, along with two independent forms of data analysis and comparison of study findings. The CFIR outlines domains and constructs that are associated with effective implementation of new interventions. Using the CFIR in this study presents results that may guide other Australian maternity services on how to best plan and implement a midwifery group practice for vulnerable women. An additional strength was the alignment of the manual and computer-assisted thematic results. Computer-assisted analysis was undertaken to mitigate the recognised and acknowledged potential inherent bias in qualitative analysis [41]. Independent analysis and consistency of results further enhances the credibility and trustworthiness of the study, along with research reflexivity throughout the study. The midwife researchers (PS and DR) concluded in their reflections that the broad range of disciplines from which participants were drawn resulted in very positive engagement from the team and enhanced marketing of the proposed change in service delivery. The diversity and large relative number of stakeholders involved in the study also ensured the qualitative data were reflective of a comprehensive sample from which data saturation was readily achieved.\nA strength of this study was the use of the CFIR to guide interviews, along with two independent forms of data analysis and comparison of study findings. The CFIR outlines domains and constructs that are associated with effective implementation of new interventions. Using the CFIR in this study presents results that may guide other Australian maternity services on how to best plan and implement a midwifery group practice for vulnerable women. An additional strength was the alignment of the manual and computer-assisted thematic results. Computer-assisted analysis was undertaken to mitigate the recognised and acknowledged potential inherent bias in qualitative analysis [41]. Independent analysis and consistency of results further enhances the credibility and trustworthiness of the study, along with research reflexivity throughout the study. The midwife researchers (PS and DR) concluded in their reflections that the broad range of disciplines from which participants were drawn resulted in very positive engagement from the team and enhanced marketing of the proposed change in service delivery. The diversity and large relative number of stakeholders involved in the study also ensured the qualitative data were reflective of a comprehensive sample from which data saturation was readily achieved.\n[SUBTITLE] Limitations [SUBSECTION] A limitation of the study was that due to local facility arrangements for selection of consumer representatives only one consumer participated in the interviews. The perspectives of consumers, and staff who were less experienced and/or from culturally and socially diverse backgrounds were not given, nor was there discussion in the interviews about the impact of the proposed model on these women. In addition, the homogenous sample, high level of experience and mature age of participants may indicate unintended sampling bias. This may be further exacerbated by the reading and preparation prior to interview done by some participants. Active pursuit of the voices of consumers and staff from a representative range of backgrounds in planning new models of maternity care across Australia is recommended by the researchers. As this study was carried out in a facility with established and supported midwifery group practices, caution should be applied in generalising the specific local results to other services for which midwifery group practice is a new concept. Instead, our interpretation of results for Australian maternity services should prompt services to identify which of our results mapped to the CFIR domains are relevant, and how they might be similar or different to the local context.\nA limitation of the study was that due to local facility arrangements for selection of consumer representatives only one consumer participated in the interviews. The perspectives of consumers, and staff who were less experienced and/or from culturally and socially diverse backgrounds were not given, nor was there discussion in the interviews about the impact of the proposed model on these women. In addition, the homogenous sample, high level of experience and mature age of participants may indicate unintended sampling bias. This may be further exacerbated by the reading and preparation prior to interview done by some participants. Active pursuit of the voices of consumers and staff from a representative range of backgrounds in planning new models of maternity care across Australia is recommended by the researchers. As this study was carried out in a facility with established and supported midwifery group practices, caution should be applied in generalising the specific local results to other services for which midwifery group practice is a new concept. Instead, our interpretation of results for Australian maternity services should prompt services to identify which of our results mapped to the CFIR domains are relevant, and how they might be similar or different to the local context.", "Stakeholders perceived that there was high quality and valid evidence supporting midwifery group practice for vulnerable women as “gold standard care” (Midwife, Interview 2). Quality evidence drawn from Australian studies [10, 13] builds confidence amongst stakeholders, which is a strong enabler. Participants weighed the relative advantages of the proposed model of care over the existing care provided to vulnerable women and believed that the health benefits for the women and infants would outweigh the costs. This belief in the net benefit when costs were also considered was not rooted in available evidence, which contrasts with the desire for evidence for “gold standard” effectiveness. The cost of the proposed model had not been established but was imagined by participants to be higher than both the current model of care and comparative midwifery group practices.\nParticipants were mostly clinicians with more than 10 years’ experience and so were likely able to make accurate assumptions around the number of workforce hours required to build rapport with women, discuss the care being delivered with the women and amongst colleagues, and deliver the volume of care required to optimise health outcomes. However, there is no known evidence for the cost-effectiveness of such a niche model of care, only generalised costs reported for Australian midwifery group practice [10, 45]. For Australian maternity services looking to implement a midwifery group practice for vulnerable women, careful understanding of costs and transparent communication to decision-makers is needed. Our research suggests that stakeholders naturally seek evidence for both costs and effectiveness, and in the absence of evidence, local costs should be examined.", "A midwifery group practice was perceived to meet the needs of vulnerable women because for example, “some with abuse histories don’t want to go over those histories over and over…” (Midwife, Interview 2). The continuity of midwives would ensure the deep needs of each woman were met. However, concerns were expressed regarding when having a known midwife might be a disincentive for women to engage. Primarily this related to women with involvement of child protection services or times where personal factors impacted on building rapport and a therapeutic relationship was not established between the woman and the midwife. A midwifery group practice for vulnerable women should be designed with flexibility in the case where a rapport is not being established between the care givers and the woman [46]. The service design would need to ensure that midwives can be changed across groups.", "The structural characteristics of the setting for this research were that it is a mature and large maternity service, with a range of existing and well-supported midwifery group practices already established. Consequently, there may have been fewer inner setting barriers to establishing the proposed model of care compared to Australian maternity services in which midwifery group practice is new or not yet established. Further, the interdisciplinary team engaged in this research was supportive of midwifery group practice. Although, it was highlighted that any potential changes to how the team works with the midwifery group practice would need to be implemented carefully. The interdisciplinary team expressed views that to continue safe and effective care, it was important the team’s role be maintained. The implementation climate of the inner setting was generally supportive, and there were some important reflections from the interdisciplinary team that would need to be acknowledged when preparing a business case for the proposed model.", "Stakeholders had a positive attitude towards the intervention; they placed a high value on the proposed model of care. Participants even sought evidence in preparation for the interviews and ensured they were familiar with the proposal. This demonstrates that individuals’ knowledge and beliefs about the intervention was a strong enabler for proceeding with the proposal. Further, that demonstrated engagement in the context assessment by stakeholders was an indicator that individuals were at an advanced stage of change in relation to redesigning maternity care for vulnerable women. The identification of this enabler suggests that the initial enthusiasm for the intervention would sustain its implementation over time. The results also demonstrated a moderate level of self-efficacy – there were mixed beliefs amongst individuals in their own capabilities to deliver the model of care, while also identifying that the proposed model of care would provide an opportunity for midwives to build their self-efficacy through gaining new skills and expanding their scope of practice. Modifications would need to be made to traditional midwifery group practice design due to perceived heavy demands from deep engagement with vulnerable women and the potential for vicarious trauma, burnout, and other emotional impacts for the midwives. Participants suggested that to overcome this barrier, caseloads should be reduced, and midwives should be well supervised and mentored as described also by others [46], especially when caring for vulnerable women [47]. To enable such an intervention to be implemented in other Australian maternity services, stakeholders would need to have confidence in their ability to seek and interpret the evidence and have an awareness of the strengths and limitations of the workforce capabilities to execute the proposed model of care.", "Conducting the context assessment instilled confidence in readiness of the maternity service to adopt the proposed change. The context assessment was conducted early in the planning stages of the intervention and the strong engagement suggested interest in the intervention and acceptance of the planning methods. This enabler resulted in positive and open communication and was an unintended consequence of the context assessment, as participation from a large and broad range of disciplines was not expected.", "A strength of this study was the use of the CFIR to guide interviews, along with two independent forms of data analysis and comparison of study findings. The CFIR outlines domains and constructs that are associated with effective implementation of new interventions. Using the CFIR in this study presents results that may guide other Australian maternity services on how to best plan and implement a midwifery group practice for vulnerable women. An additional strength was the alignment of the manual and computer-assisted thematic results. Computer-assisted analysis was undertaken to mitigate the recognised and acknowledged potential inherent bias in qualitative analysis [41]. Independent analysis and consistency of results further enhances the credibility and trustworthiness of the study, along with research reflexivity throughout the study. The midwife researchers (PS and DR) concluded in their reflections that the broad range of disciplines from which participants were drawn resulted in very positive engagement from the team and enhanced marketing of the proposed change in service delivery. The diversity and large relative number of stakeholders involved in the study also ensured the qualitative data were reflective of a comprehensive sample from which data saturation was readily achieved.", "A limitation of the study was that due to local facility arrangements for selection of consumer representatives only one consumer participated in the interviews. The perspectives of consumers, and staff who were less experienced and/or from culturally and socially diverse backgrounds were not given, nor was there discussion in the interviews about the impact of the proposed model on these women. In addition, the homogenous sample, high level of experience and mature age of participants may indicate unintended sampling bias. This may be further exacerbated by the reading and preparation prior to interview done by some participants. Active pursuit of the voices of consumers and staff from a representative range of backgrounds in planning new models of maternity care across Australia is recommended by the researchers. As this study was carried out in a facility with established and supported midwifery group practices, caution should be applied in generalising the specific local results to other services for which midwifery group practice is a new concept. Instead, our interpretation of results for Australian maternity services should prompt services to identify which of our results mapped to the CFIR domains are relevant, and how they might be similar or different to the local context.", "In this study, we have used the CFIR to identify potential barriers and enablers to implementing a midwifery group practice for vulnerable women, with both local and national relevance. There was recognition that the proposed model of care is supported by research and a view that clinical benefits will outweigh costs, however supervision and support is required for midwives to manage and limit vicarious trauma. An interdisciplinary team supporting the midwives is also an essential component of the service design. For maternity services seeking to implement a midwifery group practice for vulnerable women, our results can be leveraged to further investigate other local contexts, and quickly identify strategies for effective and sustained implementation of the new model of care [48].", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, null, null, null, null, "results", null, null, null, null, null, "discussion", null, null, null, null, null, null, null, "conclusion", "supplementary-material", null ]
[ "Midwifery", "Midwifery Group Practice", "Continuity", "Pregnancy", "Vulnerable Women", "Implementation science" ]
Components of stigma and its impact on maternal and child health service and outcomes: perspective of Akha hill tribe women in Thailand.
36261838
Maternal and child health (MCH) is crucial to the well-being of mothers and children. Stigma regarding access to MCH services is a major challenge, especially for hill tribe people in Thailand. The study aimed to understand the components of stigma and its impact on MCH service and outcomes including experiences and expectations to address the stigma in perspective of Akha hill tribe women in Thailand.
BACKGROUND
A phenomenological qualitative approach was used to gather information from Akha women who had attended MCH service one year prior and had an experience with stigma. A validated question guide was used in the study. The interview was conducted in private and confidential rooms in the Akha hill tribe villages between June and September 2021. A thematic analysis was used to extract the major and minor themes and develop the findings.
METHODS
A total of 61 Akha postdelivery participants were recruited to provide information; the average age was 28.9 years, 32.8% had no Thai ID card, and 93.4% were married. Language, traditional clothing, poverty, and name were identified as drivers of stigma, while health care providers' background, gender differences between clients and health care providers, and knowledge gaps facilitated the stigma. Being a member of a hill tribe acted as the stigma marker. Stigma manifestation was presented in the forms of verbal or physical abuse, refusal to provide treatment, and intentional disclosure of personal information to the public. Accepting the situation with no better option, defending oneself to receive better care and services, and using a private care service were experiences in addressing the stigma. Gender matching, active MCH service, mobile emergency clinics, and appropriate, permanent medical equipment in health care facilities located in their villages were the expectations.
RESULTS
Akha women face a variety of stigmas in access to MCH services, with substantial impacts on health outcomes, especially the rate of services in women and child health. Creating laws to prevent the occurrence of any forms of stigma and implementing gender matching in MCH services should be considered.
CONCLUSION
[ "Child", "Humans", "Female", "Adult", "Thailand", "Population Groups", "Child Health Services", "Family", "Surveys and Questionnaires" ]
9583464
null
null
null
null
Results
[SUBTITLE] General characteristics of the study population [SUBSECTION] A total of 61 postdelivery women were recruited into the study; the average age was 28.9 years, 32.8% had no Thai ID cards, and 93.4% were married. More than half were Buddhist (55.7%), 67.2% had completed either primary or high school, 59.0% were unemployed, and 44.3% had no regular income. Less than half had been screened for cervical cancer (31.1%) and breast cancer (21.3%) in the previous year (Table 1). Table 1General characteristics of the participantsn% Total 61100.0Age mean age = 28.9 years, min = 18, max = 41Identification card (ID card)No2032.8Yes4167.2 Marital status Married5793.4Ever married46.6 Religion Buddhist3455.7Christian2744.3 Education No education1524.6Primary school1626.2High school2541.0Diploma34.9Bachelor’s degree23.3 Occupation Unemployed3659.0Daily wage job1321.3Farmer914.8Merchant34.9Income (bath)No income2744.3≤1,00011.61,001–5,0001524.65,001–10,0001219.7≥10,00169.8 Number of children mean = 2.3, min = 1, max = 9 Number of pregnancies mean = 2.4, min = 1, max = 12 Had been screened for cervical cancer one year prior No4268.9Yes1931.1 Had been screened for breast cancer one year prior No4878.7Yes1321.3 General characteristics of the participants A total of 61 postdelivery women were recruited into the study; the average age was 28.9 years, 32.8% had no Thai ID cards, and 93.4% were married. More than half were Buddhist (55.7%), 67.2% had completed either primary or high school, 59.0% were unemployed, and 44.3% had no regular income. Less than half had been screened for cervical cancer (31.1%) and breast cancer (21.3%) in the previous year (Table 1). Table 1General characteristics of the participantsn% Total 61100.0Age mean age = 28.9 years, min = 18, max = 41Identification card (ID card)No2032.8Yes4167.2 Marital status Married5793.4Ever married46.6 Religion Buddhist3455.7Christian2744.3 Education No education1524.6Primary school1626.2High school2541.0Diploma34.9Bachelor’s degree23.3 Occupation Unemployed3659.0Daily wage job1321.3Farmer914.8Merchant34.9Income (bath)No income2744.3≤1,00011.61,001–5,0001524.65,001–10,0001219.7≥10,00169.8 Number of children mean = 2.3, min = 1, max = 9 Number of pregnancies mean = 2.4, min = 1, max = 12 Had been screened for cervical cancer one year prior No4268.9Yes1931.1 Had been screened for breast cancer one year prior No4878.7Yes1321.3 General characteristics of the participants [SUBTITLE] Components of stigma [SUBSECTION] The component of stigma consisted of the stigma driver, stigma facilitator, stigma markers, and stigma manifestations. Several factors were clearly identified as drivers, facilitators, markers, and manifestations of stigma for Akha women seeking MCH services. Different levels of the health outcome impact were reported (Fig. 1). [SUBTITLE] A) drivers of stigma for akha women seeking MCH services [SUBSECTION] Being members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services. A 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us. When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us. A 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai. If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai. The Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience. A 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them. I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them. A large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services. A 28-year-old woman said the following [P#25]: Due to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable. Due to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable. Even though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women. A 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down. At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down. Being members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services. A 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us. When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us. A 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai. If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai. The Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience. A 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them. I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them. A large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services. A 28-year-old woman said the following [P#25]: Due to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable. Due to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable. Even though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women. A 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down. At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down. [SUBTITLE] B) facilitators of stigma for akha women seeking MCH services [SUBSECTION] Several factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps. Most health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services. A 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now. When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now. A 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently. I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently. One additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services. A 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this. When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this. A 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave. I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave. The knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator. A 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that? Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that? Several factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps. Most health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services. A 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now. When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now. A 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently. I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently. One additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services. A 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this. When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this. A 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave. I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave. The knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator. A 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that? Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that? [SUBTITLE] C) stigma markers for akha women seeking MCH services [SUBSECTION] Being a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma. A 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people. I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people. Being a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma. A 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people. I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people. [SUBTITLE] D) manifestations of stigma for akha women seeking MCH services [SUBSECTION] Several manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public. Verbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers. A 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member). My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member). A 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses. Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses. Some Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery. A 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me. During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me. Refusing to give treatment was also reported among the Akha hill tribe women while attending MCH services. A 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people? When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people? Some Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment. A 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this. The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this. Several manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public. Verbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers. A 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member). My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member). A 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses. Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses. Some Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery. A 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me. During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me. Refusing to give treatment was also reported among the Akha hill tribe women while attending MCH services. A 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people? When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people? Some Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment. A 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this. The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this. [SUBTITLE] E) health outcomes [SUBSECTION] Due to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%). A 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage. When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage. A 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me? I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me? Fig. 1Stigma frame among hill tribe women who access MCH services Stigma frame among hill tribe women who access MCH services Due to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%). A 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage. When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage. A 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me? I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me? Fig. 1Stigma frame among hill tribe women who access MCH services Stigma frame among hill tribe women who access MCH services The component of stigma consisted of the stigma driver, stigma facilitator, stigma markers, and stigma manifestations. Several factors were clearly identified as drivers, facilitators, markers, and manifestations of stigma for Akha women seeking MCH services. Different levels of the health outcome impact were reported (Fig. 1). [SUBTITLE] A) drivers of stigma for akha women seeking MCH services [SUBSECTION] Being members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services. A 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us. When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us. A 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai. If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai. The Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience. A 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them. I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them. A large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services. A 28-year-old woman said the following [P#25]: Due to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable. Due to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable. Even though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women. A 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down. At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down. Being members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services. A 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us. When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us. A 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai. If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai. The Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience. A 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them. I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them. A large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services. A 28-year-old woman said the following [P#25]: Due to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable. Due to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable. Even though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women. A 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down. At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down. [SUBTITLE] B) facilitators of stigma for akha women seeking MCH services [SUBSECTION] Several factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps. Most health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services. A 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now. When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now. A 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently. I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently. One additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services. A 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this. When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this. A 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave. I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave. The knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator. A 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that? Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that? Several factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps. Most health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services. A 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now. When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now. A 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently. I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently. One additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services. A 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this. When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this. A 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave. I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave. The knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator. A 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that? Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that? [SUBTITLE] C) stigma markers for akha women seeking MCH services [SUBSECTION] Being a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma. A 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people. I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people. Being a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma. A 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people. I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people. [SUBTITLE] D) manifestations of stigma for akha women seeking MCH services [SUBSECTION] Several manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public. Verbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers. A 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member). My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member). A 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses. Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses. Some Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery. A 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me. During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me. Refusing to give treatment was also reported among the Akha hill tribe women while attending MCH services. A 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people? When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people? Some Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment. A 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this. The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this. Several manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public. Verbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers. A 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member). My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member). A 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses. Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses. Some Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery. A 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me. During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me. Refusing to give treatment was also reported among the Akha hill tribe women while attending MCH services. A 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people? When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people? Some Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment. A 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this. The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this. [SUBTITLE] E) health outcomes [SUBSECTION] Due to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%). A 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage. When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage. A 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me? I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me? Fig. 1Stigma frame among hill tribe women who access MCH services Stigma frame among hill tribe women who access MCH services Due to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%). A 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage. When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage. A 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me? I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me? Fig. 1Stigma frame among hill tribe women who access MCH services Stigma frame among hill tribe women who access MCH services [SUBTITLE] Experience in addressing the stigma [SUBSECTION] After discussing Akha hill tibe women experiences of stigma in many forms, they reported their adaptations in addressing the problem in different ways. [SUBTITLE] Accepting the situation with no better option [SUBSECTION] The first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time. A 34-year-old woman said the following [P#20]: “I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.” “I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.” The first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time. A 34-year-old woman said the following [P#20]: “I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.” “I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.” [SUBTITLE] Defending oneself to receive better care and services [SUBSECTION] Some people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs. A 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital. When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital. A 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely. A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely. Some people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs. A 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital. When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital. A 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely. A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely. [SUBTITLE] Using a private health care service [SUBSECTION] Some Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages. A 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me. My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me. A 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma. Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma. Some Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages. A 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me. My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me. A 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma. Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma. After discussing Akha hill tibe women experiences of stigma in many forms, they reported their adaptations in addressing the problem in different ways. [SUBTITLE] Accepting the situation with no better option [SUBSECTION] The first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time. A 34-year-old woman said the following [P#20]: “I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.” “I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.” The first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time. A 34-year-old woman said the following [P#20]: “I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.” “I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.” [SUBTITLE] Defending oneself to receive better care and services [SUBSECTION] Some people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs. A 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital. When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital. A 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely. A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely. Some people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs. A 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital. When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital. A 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely. A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely. [SUBTITLE] Using a private health care service [SUBSECTION] Some Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages. A 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me. My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me. A 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma. Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma. Some Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages. A 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me. My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me. A 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma. Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma. [SUBTITLE] Expectations [SUBSECTION] Regarding their expectations of MCH services, the Akha hill tribe women requested gender-matched providers in MCH services, especially for Papanicolaou test (PAP) smears, breast cancer screening, and postpartum care. Regularly offered services at the village level were an expectation because the distance from the village to the health care setting posed a major barrier to accessing care, particularly in the rainy season. Emergency mobile clinics were another need of Akha pregnant hill tribe women because when close to delivery, they needed to be able to secure timely access to a hospital, and using their everyday motorcycle was not comfortable and safe for them during late pregnancy. Moreover, the Akha hill tribe women expressed the necessity of basic standard medical equipment at small health-promoting hospitals located at the village level. The Akha women often needed to obtain services at a district hospital for many medical procedures during pregnancy. A 39-year-old woman said the following [P#53]:As I am a female, I prefer female staff at the MCH clinic. At least a female understands (another) female, and I can ask her whatever I want to know. As I am a female, I prefer female staff at the MCH clinic. At least a female understands (another) female, and I can ask her whatever I want to know. A 28-year-old woman said the following [P#10]:I want at-home postpartum visit service from health staff. Therefore, I would not need to travel to a city. I cannot drive, and my husband has to work at the farm. I want at-home postpartum visit service from health staff. Therefore, I would not need to travel to a city. I cannot drive, and my husband has to work at the farm. Regarding their expectations of MCH services, the Akha hill tribe women requested gender-matched providers in MCH services, especially for Papanicolaou test (PAP) smears, breast cancer screening, and postpartum care. Regularly offered services at the village level were an expectation because the distance from the village to the health care setting posed a major barrier to accessing care, particularly in the rainy season. Emergency mobile clinics were another need of Akha pregnant hill tribe women because when close to delivery, they needed to be able to secure timely access to a hospital, and using their everyday motorcycle was not comfortable and safe for them during late pregnancy. Moreover, the Akha hill tribe women expressed the necessity of basic standard medical equipment at small health-promoting hospitals located at the village level. The Akha women often needed to obtain services at a district hospital for many medical procedures during pregnancy. A 39-year-old woman said the following [P#53]:As I am a female, I prefer female staff at the MCH clinic. At least a female understands (another) female, and I can ask her whatever I want to know. As I am a female, I prefer female staff at the MCH clinic. At least a female understands (another) female, and I can ask her whatever I want to know. A 28-year-old woman said the following [P#10]:I want at-home postpartum visit service from health staff. Therefore, I would not need to travel to a city. I cannot drive, and my husband has to work at the farm. I want at-home postpartum visit service from health staff. Therefore, I would not need to travel to a city. I cannot drive, and my husband has to work at the farm.
Conclusion
Akha women who attend a public MCH clinic in Thailand suffer stigma driven by their specific characteristics, such as being unable to speak Thai (or lacking fluency), being poor, wearing their traditional dress and using traditional naming conventions. Health care providers’ background, gender mismatches and knowledge gaps between health care providers and clients were identified as facilitators of stigma for women seeking MCH services. Akha women face many forms of stigma when receiving clinical services, including verbal and physical abuse, a refusal to provide treatment, and the intentional disclosure of their personal information to the public. As a result, such mistreatment affects multiple health and health service outcomes, such as poor rates of attending antenatal care and low rates of cervical and breast cancer screening. Akha hill tribe women use many approaches to adjust to the stigma that they encounter, such as accepting the situation with no better option, defending themselves to obtain better care and services, and using a private clinic instead. Akha women expect to have active services and gender-matched health care providers at MCH services and mobile emergency clinics and that the appropriate equipment be provided to equip standard MCH services at hospitals located in or near the hill tribe villages. Serious consideration of the problems posed by the stigmatization for Akha hill tribe women is needed to improve their access to health care services, particularly those attending MCH services. Policies with standard protocols to ensure the provision of equal care for everyone should be implemented. The improvement of health facilities located in hill tribe villages should also be considered to familiarize hill tribe people with health care providers in their home setting. Moreover, encouraging hill tribe people to be trained as nurses or medical doctors and then sending them back to work at a hospital in their village could eventually reduce stigma.
[ "Background", "Methods", "Study design and setting", "Research tool and its development", "Sampling and recruitment", "Data collection", "Data analysis", "Rigor and trustworthiness", "General characteristics of the study population", "Components of stigma", "A) drivers of stigma for akha women seeking MCH services", "B) facilitators of stigma for akha women seeking MCH services", "C) stigma markers for akha women seeking MCH services", "D) manifestations of stigma for akha women seeking MCH services", "E) health outcomes", "Experience in addressing the stigma", "Accepting the situation with no better option", "Defending oneself to receive better care and services", "Using a private health care service", "Expectations", "" ]
[ "Stigma is defined as one of the major barriers to accessing health care services [1–4]. Moreover, its impact is greater among vulnerable populations [5, 6]. Maternal health and child care (MCH) is a significant service to people globally to ensure that both mothers and children have good health, including promoting survival during the pregnancy, childbirth and postnatal periods [7]. In 2017, the World Health Organization (WHO) reported that 295,000 women died during and following pregnancy and childbirth [8]. The WHO also recommends that in all stages of MCH, care should minimize negative experiences to ensure that women and their infants reach their full potential of health and wellbeing, especially among vulnerable populations [7]. The major negative experience among pregnant women is stigma [9]. The stigma that women encounter during their visits to MCH services could minimize the rate of access to MCH [10, 11]. Several impacts have been reported from the stigma in access to MCH services, such as poor child health [9] and human immunodeficiency virus (HIV) infection [12]. Many illnesses related to unable to access MCH are required a large amount of money for the treatment and care, especially in developing countries [13, 14], including Thailand [15]. Furthermore, a number of illnesses related to women’s reproductive health can lead to premature death and reduce quality of life [16–18].\nIn Thailand, MCH services are provided to all reproductive groups and children, including those who do not hold Thai identification (ID) cards, which are used for access to all public services [19]. Commonly, all health institutes provide an MCH service every Tuesday, including a small, so-called health-promoting hospital located in the hill tribe villages [20]. At the health-promoting hospital, pregnant women are cared for under the guidelines of the Thai Ministry of Public Health, including an assessment of general health and detection of potential risks [21]. However, one study in Chiang Rai, Thailand, reported that only 7.1% received three doses of tetanus toxoid during pregnancy, and less than 50% of pregnant hill tribe women accessed MCH services properly [22].Moreover, 64.3% of Akha pregnant women gave birth at home by untrained midwives, and only 30% of Akha children received vaccines based on Thailand expanded program on immunization (EPI) program [23].\nThe presence of stigma when attending a clinic exerts the greatest impact on certain populations with specific characteristics [24–26]. The hill tribe people in Thailand have moved down from South China over several centuries [27]. There are six main tribes: Akah, Lahu, Hmong, Yao, Karen, and Lisu [27]. Akha people comprise the largest group, with their own culture, lifestyle, and language [27] which is different from Thai people [28]. Most Akha in Thailand live under the national poverty line [29] and have poor education [24]. Inaddition, Akha people are very limited in their use of the Thai language [30]. While all health caregivers are Thai then it is difficult to Akha people to access the services [31].\nThe stigma present for minority groups when attending health care institutions is well recognized [32]. In the current study, a health stigma and discrimination framework was used as a guideline for understanding the enacted stigma that exists, which is the form of the stigma perceived by individuals [33] while accessing MCH services among Akha women [5]. According to Stangl et al. [5] concept of stigma, a stigma driver is the factor that drives stigma presentation in a phenomenon, and some factors work to facilitate stigma presentation, while the stigma marker is the original marker for the existing stigma. Understanding the component of stigma present for hill tribe women when access MCH services can be applied for health policy formulation and public health implementation to reduce the stigma. Minimizing the stigma encountered by hill tribe women seeking MCH services could improve their access to all clinical services related to women’s health, including screening for cervical and breast cancer. Reducing the stigma encountered during health care services, especially in an MCH service, will improve both the quality and quantity of the services.\nThe study aimed to understand the components of stigma and its impact on MCH service and outcomes including experience and expectation to address the stigma in perspective of Akha hill tribe women in Thailand.", "[SUBTITLE] Study design and setting [SUBSECTION] A phenomenological qualitative approach [34] was used to elicit information from participants who were Akha hill tribe women living in seven hill tribe villages located along the Thailand-Myanmar border who experienced stigma while accessing MCH services. Akha women who were pregnant or had delivered their child one year prior to data collection and had accessed an MCH service at least once were invited to participate in the study.\nA phenomenological qualitative approach [34] was used to elicit information from participants who were Akha hill tribe women living in seven hill tribe villages located along the Thailand-Myanmar border who experienced stigma while accessing MCH services. Akha women who were pregnant or had delivered their child one year prior to data collection and had accessed an MCH service at least once were invited to participate in the study.\n[SUBTITLE] Research tool and its development [SUBSECTION] The questions were developed from a review of the literature, information obtained from health care providers who worked in the hill tribe villages, and from some pregnant women who had experienced stigma while attending an MCH service. The validity and reliability of the questions were tested before use in the field. Three external experts who were public health professional, medical anthropologist, and nurse working at MCH services were invited to validate the question information and the research context and content. The objective of the validity test was to confirm that the contents of the questions covered the context required in the study. The questions were piloted among six postdelivery women who lived in two hill tribe villages in Mae Chan District, Chiang Rai Province, Thailand. The main objective of the pilot test was to ensure that both the researchers and participants understood the same meaning and sense of the questions provided. Ultimately, seven questions were finalized for use in the study: (1) Which hospital did you attend for MCH services? (2) Did you experience any discomfort or stigma when attending MCH services? (3) Can you provide information in terms of frequency, who displayed stigmatizing behaviors, and in what form? (4) How did you feel about this experience? (5) How did you respond to these behaviors? (6) What is your expectation about accessing MCH services? (7) Did you experience other barriers to accessing MCH services?\nThe questions were developed from a review of the literature, information obtained from health care providers who worked in the hill tribe villages, and from some pregnant women who had experienced stigma while attending an MCH service. The validity and reliability of the questions were tested before use in the field. Three external experts who were public health professional, medical anthropologist, and nurse working at MCH services were invited to validate the question information and the research context and content. The objective of the validity test was to confirm that the contents of the questions covered the context required in the study. The questions were piloted among six postdelivery women who lived in two hill tribe villages in Mae Chan District, Chiang Rai Province, Thailand. The main objective of the pilot test was to ensure that both the researchers and participants understood the same meaning and sense of the questions provided. Ultimately, seven questions were finalized for use in the study: (1) Which hospital did you attend for MCH services? (2) Did you experience any discomfort or stigma when attending MCH services? (3) Can you provide information in terms of frequency, who displayed stigmatizing behaviors, and in what form? (4) How did you feel about this experience? (5) How did you respond to these behaviors? (6) What is your expectation about accessing MCH services? (7) Did you experience other barriers to accessing MCH services?\n[SUBTITLE] Sampling and recruitment [SUBSECTION] Village headmen were informed about the study and asked to select participants five days in advance according to the inclusion criteria. The participants were purposively selected from seven hill tribe villages. Hill tribe women who were postdelivery one year prior who had experienced stigma when attending an MCH clinic and who able to use Thai met the inclusion criteria. Women who met the criteria were informed by the village headman and asked to participate in the study. At the date of the interview, women who met the criteria and intended to provide information to the researcher were screened again to determine whether they had evidence according to the criteria. Only those who had a strong experience with stigma were invited to an interview. All participants were provided with information about the study and signed written consent forms that stated the voluntary nature of participation. Three researchers who were trained in qualitative methods (one female medical anthropologist (Ph.D.), one female health behavioral scientist (Ph.D.), and one male public health expert (Ph.D.)) and working as university faculty were the interviewers. All interviewers were women who were familiar from previous projects with the hill tribe people living in these areas.\nVillage headmen were informed about the study and asked to select participants five days in advance according to the inclusion criteria. The participants were purposively selected from seven hill tribe villages. Hill tribe women who were postdelivery one year prior who had experienced stigma when attending an MCH clinic and who able to use Thai met the inclusion criteria. Women who met the criteria were informed by the village headman and asked to participate in the study. At the date of the interview, women who met the criteria and intended to provide information to the researcher were screened again to determine whether they had evidence according to the criteria. Only those who had a strong experience with stigma were invited to an interview. All participants were provided with information about the study and signed written consent forms that stated the voluntary nature of participation. Three researchers who were trained in qualitative methods (one female medical anthropologist (Ph.D.), one female health behavioral scientist (Ph.D.), and one male public health expert (Ph.D.)) and working as university faculty were the interviewers. All interviewers were women who were familiar from previous projects with the hill tribe people living in these areas.\n[SUBTITLE] Data collection [SUBSECTION] Face-to-face interviews were conducted in a private and confidential room at the community hall in each village between June and September 2021. A question guide was used in the interview. Before the interview, the participants were asked for permission to record it and take field notes. The interviews started with the objective of research and general questions about maternal and child health. The specific questions on asking about the experience of stigma while attending MCH services were followed. Each interview lasted for 45 min. All methods were carried out in accordance with the Declaration of Helsinki [35] in the ethical principles for medical research that involve human subjects.\nFace-to-face interviews were conducted in a private and confidential room at the community hall in each village between June and September 2021. A question guide was used in the interview. Before the interview, the participants were asked for permission to record it and take field notes. The interviews started with the objective of research and general questions about maternal and child health. The specific questions on asking about the experience of stigma while attending MCH services were followed. Each interview lasted for 45 min. All methods were carried out in accordance with the Declaration of Helsinki [35] in the ethical principles for medical research that involve human subjects.\n[SUBTITLE] Data analysis [SUBSECTION] All records were transcribed and checked before further analysis. The transcript was sent to all participants who were the information owner to check its accuracy before further analysis. The information in the transcripts was coded, and coding trees were developed. The codes were transferred into the NVivo program (NVivo, qualitative data analysis software; QSR International Pty Ltd., version 11, 2015) for theme extraction. A content analysis was used to extract major and minor themes with the inductive method, which usually uses the keywords presented from interviews to construct the themes. The major theme was used to present the form or pattern of the stigma while attending MCH services. The minor theme was focused on the other significant surrounding information, including the experiences in addressing the stigma, and expectations of the participants to further solve the problem. All themes identified were constructed and formed. Significant quotations were presented to support the findings.\nAll records were transcribed and checked before further analysis. The transcript was sent to all participants who were the information owner to check its accuracy before further analysis. The information in the transcripts was coded, and coding trees were developed. The codes were transferred into the NVivo program (NVivo, qualitative data analysis software; QSR International Pty Ltd., version 11, 2015) for theme extraction. A content analysis was used to extract major and minor themes with the inductive method, which usually uses the keywords presented from interviews to construct the themes. The major theme was used to present the form or pattern of the stigma while attending MCH services. The minor theme was focused on the other significant surrounding information, including the experiences in addressing the stigma, and expectations of the participants to further solve the problem. All themes identified were constructed and formed. Significant quotations were presented to support the findings.\n[SUBTITLE] Rigor and trustworthiness [SUBSECTION] Before deciding on final interpretations, the researchers once again sent the information back to the participant who was the information owner to ensure the accuracy of the final findings. Two qualitative research experts in the field were asked to validate the final findings and framework (Fig. 1). The final framework was discussed and validated again with eight local Akha people (five women and three community leaders).\nBefore deciding on final interpretations, the researchers once again sent the information back to the participant who was the information owner to ensure the accuracy of the final findings. Two qualitative research experts in the field were asked to validate the final findings and framework (Fig. 1). The final framework was discussed and validated again with eight local Akha people (five women and three community leaders).", "A phenomenological qualitative approach [34] was used to elicit information from participants who were Akha hill tribe women living in seven hill tribe villages located along the Thailand-Myanmar border who experienced stigma while accessing MCH services. Akha women who were pregnant or had delivered their child one year prior to data collection and had accessed an MCH service at least once were invited to participate in the study.", "The questions were developed from a review of the literature, information obtained from health care providers who worked in the hill tribe villages, and from some pregnant women who had experienced stigma while attending an MCH service. The validity and reliability of the questions were tested before use in the field. Three external experts who were public health professional, medical anthropologist, and nurse working at MCH services were invited to validate the question information and the research context and content. The objective of the validity test was to confirm that the contents of the questions covered the context required in the study. The questions were piloted among six postdelivery women who lived in two hill tribe villages in Mae Chan District, Chiang Rai Province, Thailand. The main objective of the pilot test was to ensure that both the researchers and participants understood the same meaning and sense of the questions provided. Ultimately, seven questions were finalized for use in the study: (1) Which hospital did you attend for MCH services? (2) Did you experience any discomfort or stigma when attending MCH services? (3) Can you provide information in terms of frequency, who displayed stigmatizing behaviors, and in what form? (4) How did you feel about this experience? (5) How did you respond to these behaviors? (6) What is your expectation about accessing MCH services? (7) Did you experience other barriers to accessing MCH services?", "Village headmen were informed about the study and asked to select participants five days in advance according to the inclusion criteria. The participants were purposively selected from seven hill tribe villages. Hill tribe women who were postdelivery one year prior who had experienced stigma when attending an MCH clinic and who able to use Thai met the inclusion criteria. Women who met the criteria were informed by the village headman and asked to participate in the study. At the date of the interview, women who met the criteria and intended to provide information to the researcher were screened again to determine whether they had evidence according to the criteria. Only those who had a strong experience with stigma were invited to an interview. All participants were provided with information about the study and signed written consent forms that stated the voluntary nature of participation. Three researchers who were trained in qualitative methods (one female medical anthropologist (Ph.D.), one female health behavioral scientist (Ph.D.), and one male public health expert (Ph.D.)) and working as university faculty were the interviewers. All interviewers were women who were familiar from previous projects with the hill tribe people living in these areas.", "Face-to-face interviews were conducted in a private and confidential room at the community hall in each village between June and September 2021. A question guide was used in the interview. Before the interview, the participants were asked for permission to record it and take field notes. The interviews started with the objective of research and general questions about maternal and child health. The specific questions on asking about the experience of stigma while attending MCH services were followed. Each interview lasted for 45 min. All methods were carried out in accordance with the Declaration of Helsinki [35] in the ethical principles for medical research that involve human subjects.", "All records were transcribed and checked before further analysis. The transcript was sent to all participants who were the information owner to check its accuracy before further analysis. The information in the transcripts was coded, and coding trees were developed. The codes were transferred into the NVivo program (NVivo, qualitative data analysis software; QSR International Pty Ltd., version 11, 2015) for theme extraction. A content analysis was used to extract major and minor themes with the inductive method, which usually uses the keywords presented from interviews to construct the themes. The major theme was used to present the form or pattern of the stigma while attending MCH services. The minor theme was focused on the other significant surrounding information, including the experiences in addressing the stigma, and expectations of the participants to further solve the problem. All themes identified were constructed and formed. Significant quotations were presented to support the findings.", "Before deciding on final interpretations, the researchers once again sent the information back to the participant who was the information owner to ensure the accuracy of the final findings. Two qualitative research experts in the field were asked to validate the final findings and framework (Fig. 1). The final framework was discussed and validated again with eight local Akha people (five women and three community leaders).", "A total of 61 postdelivery women were recruited into the study; the average age was 28.9 years, 32.8% had no Thai ID cards, and 93.4% were married. More than half were Buddhist (55.7%), 67.2% had completed either primary or high school, 59.0% were unemployed, and 44.3% had no regular income. Less than half had been screened for cervical cancer (31.1%) and breast cancer (21.3%) in the previous year (Table 1).\n\nTable 1General characteristics of the participantsn%\nTotal\n61100.0Age mean age = 28.9 years, min = 18, max = 41Identification card (ID card)No2032.8Yes4167.2\nMarital status\nMarried5793.4Ever married46.6\nReligion\nBuddhist3455.7Christian2744.3\nEducation\nNo education1524.6Primary school1626.2High school2541.0Diploma34.9Bachelor’s degree23.3\nOccupation\nUnemployed3659.0Daily wage job1321.3Farmer914.8Merchant34.9Income (bath)No income2744.3≤1,00011.61,001–5,0001524.65,001–10,0001219.7≥10,00169.8\nNumber of children\nmean = 2.3, min = 1, max = 9\nNumber of pregnancies\nmean = 2.4, min = 1, max = 12\nHad been screened for cervical cancer one year prior\nNo4268.9Yes1931.1\nHad been screened for breast cancer one year prior\nNo4878.7Yes1321.3\n\nGeneral characteristics of the participants", "The component of stigma consisted of the stigma driver, stigma facilitator, stigma markers, and stigma manifestations. Several factors were clearly identified as drivers, facilitators, markers, and manifestations of stigma for Akha women seeking MCH services. Different levels of the health outcome impact were reported (Fig. 1).\n[SUBTITLE] A) drivers of stigma for akha women seeking MCH services [SUBSECTION] Being members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services.\nA 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nWhen I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nA 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nIf I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nThe Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience.\nA 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nI am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nA large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services.\nA 28-year-old woman said the following [P#25]:\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\nEven though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women.\nA 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nAt first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nBeing members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services.\nA 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nWhen I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nA 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nIf I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nThe Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience.\nA 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nI am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nA large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services.\nA 28-year-old woman said the following [P#25]:\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\nEven though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women.\nA 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nAt first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\n[SUBTITLE] B) facilitators of stigma for akha women seeking MCH services [SUBSECTION] Several factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps.\nMost health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services.\nA 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nWhen I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nA 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nI feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nOne additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services.\nA 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nWhen I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nA 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nI had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nThe knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator.\nA 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nAnytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nSeveral factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps.\nMost health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services.\nA 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nWhen I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nA 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nI feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nOne additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services.\nA 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nWhen I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nA 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nI had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nThe knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator.\nA 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nAnytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\n[SUBTITLE] C) stigma markers for akha women seeking MCH services [SUBSECTION] Being a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma.\nA 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nI have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nBeing a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma.\nA 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nI have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\n[SUBTITLE] D) manifestations of stigma for akha women seeking MCH services [SUBSECTION] Several manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public.\nVerbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers.\nA 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nMy bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nA 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nLast year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nSome Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery.\nA 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nDuring my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nRefusing to give treatment was also reported among the Akha hill tribe women while attending MCH services.\nA 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nWhen I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nSome Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment.\nA 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nThe staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nSeveral manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public.\nVerbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers.\nA 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nMy bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nA 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nLast year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nSome Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery.\nA 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nDuring my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nRefusing to give treatment was also reported among the Akha hill tribe women while attending MCH services.\nA 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nWhen I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nSome Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment.\nA 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nThe staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\n[SUBTITLE] E) health outcomes [SUBSECTION] Due to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%).\nA 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nWhen I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nA 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\nI had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\n\nFig. 1Stigma frame among hill tribe women who access MCH services\n\nStigma frame among hill tribe women who access MCH services\nDue to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%).\nA 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nWhen I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nA 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\nI had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\n\nFig. 1Stigma frame among hill tribe women who access MCH services\n\nStigma frame among hill tribe women who access MCH services", "Being members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services.\nA 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nWhen I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nA 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nIf I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nThe Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience.\nA 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nI am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nA large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services.\nA 28-year-old woman said the following [P#25]:\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\nEven though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women.\nA 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nAt first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.", "Several factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps.\nMost health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services.\nA 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nWhen I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nA 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nI feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nOne additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services.\nA 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nWhen I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nA 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nI had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nThe knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator.\nA 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nAnytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?", "Being a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma.\nA 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nI have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.", "Several manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public.\nVerbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers.\nA 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nMy bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nA 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nLast year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nSome Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery.\nA 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nDuring my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nRefusing to give treatment was also reported among the Akha hill tribe women while attending MCH services.\nA 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nWhen I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nSome Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment.\nA 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nThe staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.", "Due to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%).\nA 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nWhen I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nA 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\nI had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\n\nFig. 1Stigma frame among hill tribe women who access MCH services\n\nStigma frame among hill tribe women who access MCH services", "After discussing Akha hill tibe women experiences of stigma in many forms, they reported their adaptations in addressing the problem in different ways.\n[SUBTITLE] Accepting the situation with no better option [SUBSECTION] The first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time.\nA 34-year-old woman said the following [P#20]:\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\nThe first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time.\nA 34-year-old woman said the following [P#20]:\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n[SUBTITLE] Defending oneself to receive better care and services [SUBSECTION] Some people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs.\nA 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nWhen my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nA 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nA staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nSome people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs.\nA 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nWhen my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nA 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nA staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\n[SUBTITLE] Using a private health care service [SUBSECTION] Some Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages.\nA 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nMy personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nA 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nMoney can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nSome Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages.\nA 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nMy personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nA 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nMoney can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.", "The first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time.\nA 34-year-old woman said the following [P#20]:\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”", "Some people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs.\nA 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nWhen my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nA 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nA staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.", "Some Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages.\nA 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nMy personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nA 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nMoney can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.", "Regarding their expectations of MCH services, the Akha hill tribe women requested gender-matched providers in MCH services, especially for Papanicolaou test (PAP) smears, breast cancer screening, and postpartum care. Regularly offered services at the village level were an expectation because the distance from the village to the health care setting posed a major barrier to accessing care, particularly in the rainy season. Emergency mobile clinics were another need of Akha pregnant hill tribe women because when close to delivery, they needed to be able to secure timely access to a hospital, and using their everyday motorcycle was not comfortable and safe for them during late pregnancy. Moreover, the Akha hill tribe women expressed the necessity of basic standard medical equipment at small health-promoting hospitals located at the village level. The Akha women often needed to obtain services at a district hospital for many medical procedures during pregnancy.\nA 39-year-old woman said the following [P#53]:As I am a female, I prefer female staff at the MCH clinic. At least a female understands (another) female, and I can ask her whatever I want to know.\nAs I am a female, I prefer female staff at the MCH clinic. At least a female understands (another) female, and I can ask her whatever I want to know.\nA 28-year-old woman said the following [P#10]:I want at-home postpartum visit service from health staff. Therefore, I would not need to travel to a city. I cannot drive, and my husband has to work at the farm.\nI want at-home postpartum visit service from health staff. Therefore, I would not need to travel to a city. I cannot drive, and my husband has to work at the farm.", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2" ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Study design and setting", "Research tool and its development", "Sampling and recruitment", "Data collection", "Data analysis", "Rigor and trustworthiness", "Results", "General characteristics of the study population", "Components of stigma", "A) drivers of stigma for akha women seeking MCH services", "B) facilitators of stigma for akha women seeking MCH services", "C) stigma markers for akha women seeking MCH services", "D) manifestations of stigma for akha women seeking MCH services", "E) health outcomes", "Experience in addressing the stigma", "Accepting the situation with no better option", "Defending oneself to receive better care and services", "Using a private health care service", "Expectations", "Discussion", "Conclusion", "Electronic supplementary material", "" ]
[ "Stigma is defined as one of the major barriers to accessing health care services [1–4]. Moreover, its impact is greater among vulnerable populations [5, 6]. Maternal health and child care (MCH) is a significant service to people globally to ensure that both mothers and children have good health, including promoting survival during the pregnancy, childbirth and postnatal periods [7]. In 2017, the World Health Organization (WHO) reported that 295,000 women died during and following pregnancy and childbirth [8]. The WHO also recommends that in all stages of MCH, care should minimize negative experiences to ensure that women and their infants reach their full potential of health and wellbeing, especially among vulnerable populations [7]. The major negative experience among pregnant women is stigma [9]. The stigma that women encounter during their visits to MCH services could minimize the rate of access to MCH [10, 11]. Several impacts have been reported from the stigma in access to MCH services, such as poor child health [9] and human immunodeficiency virus (HIV) infection [12]. Many illnesses related to unable to access MCH are required a large amount of money for the treatment and care, especially in developing countries [13, 14], including Thailand [15]. Furthermore, a number of illnesses related to women’s reproductive health can lead to premature death and reduce quality of life [16–18].\nIn Thailand, MCH services are provided to all reproductive groups and children, including those who do not hold Thai identification (ID) cards, which are used for access to all public services [19]. Commonly, all health institutes provide an MCH service every Tuesday, including a small, so-called health-promoting hospital located in the hill tribe villages [20]. At the health-promoting hospital, pregnant women are cared for under the guidelines of the Thai Ministry of Public Health, including an assessment of general health and detection of potential risks [21]. However, one study in Chiang Rai, Thailand, reported that only 7.1% received three doses of tetanus toxoid during pregnancy, and less than 50% of pregnant hill tribe women accessed MCH services properly [22].Moreover, 64.3% of Akha pregnant women gave birth at home by untrained midwives, and only 30% of Akha children received vaccines based on Thailand expanded program on immunization (EPI) program [23].\nThe presence of stigma when attending a clinic exerts the greatest impact on certain populations with specific characteristics [24–26]. The hill tribe people in Thailand have moved down from South China over several centuries [27]. There are six main tribes: Akah, Lahu, Hmong, Yao, Karen, and Lisu [27]. Akha people comprise the largest group, with their own culture, lifestyle, and language [27] which is different from Thai people [28]. Most Akha in Thailand live under the national poverty line [29] and have poor education [24]. Inaddition, Akha people are very limited in their use of the Thai language [30]. While all health caregivers are Thai then it is difficult to Akha people to access the services [31].\nThe stigma present for minority groups when attending health care institutions is well recognized [32]. In the current study, a health stigma and discrimination framework was used as a guideline for understanding the enacted stigma that exists, which is the form of the stigma perceived by individuals [33] while accessing MCH services among Akha women [5]. According to Stangl et al. [5] concept of stigma, a stigma driver is the factor that drives stigma presentation in a phenomenon, and some factors work to facilitate stigma presentation, while the stigma marker is the original marker for the existing stigma. Understanding the component of stigma present for hill tribe women when access MCH services can be applied for health policy formulation and public health implementation to reduce the stigma. Minimizing the stigma encountered by hill tribe women seeking MCH services could improve their access to all clinical services related to women’s health, including screening for cervical and breast cancer. Reducing the stigma encountered during health care services, especially in an MCH service, will improve both the quality and quantity of the services.\nThe study aimed to understand the components of stigma and its impact on MCH service and outcomes including experience and expectation to address the stigma in perspective of Akha hill tribe women in Thailand.", "[SUBTITLE] Study design and setting [SUBSECTION] A phenomenological qualitative approach [34] was used to elicit information from participants who were Akha hill tribe women living in seven hill tribe villages located along the Thailand-Myanmar border who experienced stigma while accessing MCH services. Akha women who were pregnant or had delivered their child one year prior to data collection and had accessed an MCH service at least once were invited to participate in the study.\nA phenomenological qualitative approach [34] was used to elicit information from participants who were Akha hill tribe women living in seven hill tribe villages located along the Thailand-Myanmar border who experienced stigma while accessing MCH services. Akha women who were pregnant or had delivered their child one year prior to data collection and had accessed an MCH service at least once were invited to participate in the study.\n[SUBTITLE] Research tool and its development [SUBSECTION] The questions were developed from a review of the literature, information obtained from health care providers who worked in the hill tribe villages, and from some pregnant women who had experienced stigma while attending an MCH service. The validity and reliability of the questions were tested before use in the field. Three external experts who were public health professional, medical anthropologist, and nurse working at MCH services were invited to validate the question information and the research context and content. The objective of the validity test was to confirm that the contents of the questions covered the context required in the study. The questions were piloted among six postdelivery women who lived in two hill tribe villages in Mae Chan District, Chiang Rai Province, Thailand. The main objective of the pilot test was to ensure that both the researchers and participants understood the same meaning and sense of the questions provided. Ultimately, seven questions were finalized for use in the study: (1) Which hospital did you attend for MCH services? (2) Did you experience any discomfort or stigma when attending MCH services? (3) Can you provide information in terms of frequency, who displayed stigmatizing behaviors, and in what form? (4) How did you feel about this experience? (5) How did you respond to these behaviors? (6) What is your expectation about accessing MCH services? (7) Did you experience other barriers to accessing MCH services?\nThe questions were developed from a review of the literature, information obtained from health care providers who worked in the hill tribe villages, and from some pregnant women who had experienced stigma while attending an MCH service. The validity and reliability of the questions were tested before use in the field. Three external experts who were public health professional, medical anthropologist, and nurse working at MCH services were invited to validate the question information and the research context and content. The objective of the validity test was to confirm that the contents of the questions covered the context required in the study. The questions were piloted among six postdelivery women who lived in two hill tribe villages in Mae Chan District, Chiang Rai Province, Thailand. The main objective of the pilot test was to ensure that both the researchers and participants understood the same meaning and sense of the questions provided. Ultimately, seven questions were finalized for use in the study: (1) Which hospital did you attend for MCH services? (2) Did you experience any discomfort or stigma when attending MCH services? (3) Can you provide information in terms of frequency, who displayed stigmatizing behaviors, and in what form? (4) How did you feel about this experience? (5) How did you respond to these behaviors? (6) What is your expectation about accessing MCH services? (7) Did you experience other barriers to accessing MCH services?\n[SUBTITLE] Sampling and recruitment [SUBSECTION] Village headmen were informed about the study and asked to select participants five days in advance according to the inclusion criteria. The participants were purposively selected from seven hill tribe villages. Hill tribe women who were postdelivery one year prior who had experienced stigma when attending an MCH clinic and who able to use Thai met the inclusion criteria. Women who met the criteria were informed by the village headman and asked to participate in the study. At the date of the interview, women who met the criteria and intended to provide information to the researcher were screened again to determine whether they had evidence according to the criteria. Only those who had a strong experience with stigma were invited to an interview. All participants were provided with information about the study and signed written consent forms that stated the voluntary nature of participation. Three researchers who were trained in qualitative methods (one female medical anthropologist (Ph.D.), one female health behavioral scientist (Ph.D.), and one male public health expert (Ph.D.)) and working as university faculty were the interviewers. All interviewers were women who were familiar from previous projects with the hill tribe people living in these areas.\nVillage headmen were informed about the study and asked to select participants five days in advance according to the inclusion criteria. The participants were purposively selected from seven hill tribe villages. Hill tribe women who were postdelivery one year prior who had experienced stigma when attending an MCH clinic and who able to use Thai met the inclusion criteria. Women who met the criteria were informed by the village headman and asked to participate in the study. At the date of the interview, women who met the criteria and intended to provide information to the researcher were screened again to determine whether they had evidence according to the criteria. Only those who had a strong experience with stigma were invited to an interview. All participants were provided with information about the study and signed written consent forms that stated the voluntary nature of participation. Three researchers who were trained in qualitative methods (one female medical anthropologist (Ph.D.), one female health behavioral scientist (Ph.D.), and one male public health expert (Ph.D.)) and working as university faculty were the interviewers. All interviewers were women who were familiar from previous projects with the hill tribe people living in these areas.\n[SUBTITLE] Data collection [SUBSECTION] Face-to-face interviews were conducted in a private and confidential room at the community hall in each village between June and September 2021. A question guide was used in the interview. Before the interview, the participants were asked for permission to record it and take field notes. The interviews started with the objective of research and general questions about maternal and child health. The specific questions on asking about the experience of stigma while attending MCH services were followed. Each interview lasted for 45 min. All methods were carried out in accordance with the Declaration of Helsinki [35] in the ethical principles for medical research that involve human subjects.\nFace-to-face interviews were conducted in a private and confidential room at the community hall in each village between June and September 2021. A question guide was used in the interview. Before the interview, the participants were asked for permission to record it and take field notes. The interviews started with the objective of research and general questions about maternal and child health. The specific questions on asking about the experience of stigma while attending MCH services were followed. Each interview lasted for 45 min. All methods were carried out in accordance with the Declaration of Helsinki [35] in the ethical principles for medical research that involve human subjects.\n[SUBTITLE] Data analysis [SUBSECTION] All records were transcribed and checked before further analysis. The transcript was sent to all participants who were the information owner to check its accuracy before further analysis. The information in the transcripts was coded, and coding trees were developed. The codes were transferred into the NVivo program (NVivo, qualitative data analysis software; QSR International Pty Ltd., version 11, 2015) for theme extraction. A content analysis was used to extract major and minor themes with the inductive method, which usually uses the keywords presented from interviews to construct the themes. The major theme was used to present the form or pattern of the stigma while attending MCH services. The minor theme was focused on the other significant surrounding information, including the experiences in addressing the stigma, and expectations of the participants to further solve the problem. All themes identified were constructed and formed. Significant quotations were presented to support the findings.\nAll records were transcribed and checked before further analysis. The transcript was sent to all participants who were the information owner to check its accuracy before further analysis. The information in the transcripts was coded, and coding trees were developed. The codes were transferred into the NVivo program (NVivo, qualitative data analysis software; QSR International Pty Ltd., version 11, 2015) for theme extraction. A content analysis was used to extract major and minor themes with the inductive method, which usually uses the keywords presented from interviews to construct the themes. The major theme was used to present the form or pattern of the stigma while attending MCH services. The minor theme was focused on the other significant surrounding information, including the experiences in addressing the stigma, and expectations of the participants to further solve the problem. All themes identified were constructed and formed. Significant quotations were presented to support the findings.\n[SUBTITLE] Rigor and trustworthiness [SUBSECTION] Before deciding on final interpretations, the researchers once again sent the information back to the participant who was the information owner to ensure the accuracy of the final findings. Two qualitative research experts in the field were asked to validate the final findings and framework (Fig. 1). The final framework was discussed and validated again with eight local Akha people (five women and three community leaders).\nBefore deciding on final interpretations, the researchers once again sent the information back to the participant who was the information owner to ensure the accuracy of the final findings. Two qualitative research experts in the field were asked to validate the final findings and framework (Fig. 1). The final framework was discussed and validated again with eight local Akha people (five women and three community leaders).", "A phenomenological qualitative approach [34] was used to elicit information from participants who were Akha hill tribe women living in seven hill tribe villages located along the Thailand-Myanmar border who experienced stigma while accessing MCH services. Akha women who were pregnant or had delivered their child one year prior to data collection and had accessed an MCH service at least once were invited to participate in the study.", "The questions were developed from a review of the literature, information obtained from health care providers who worked in the hill tribe villages, and from some pregnant women who had experienced stigma while attending an MCH service. The validity and reliability of the questions were tested before use in the field. Three external experts who were public health professional, medical anthropologist, and nurse working at MCH services were invited to validate the question information and the research context and content. The objective of the validity test was to confirm that the contents of the questions covered the context required in the study. The questions were piloted among six postdelivery women who lived in two hill tribe villages in Mae Chan District, Chiang Rai Province, Thailand. The main objective of the pilot test was to ensure that both the researchers and participants understood the same meaning and sense of the questions provided. Ultimately, seven questions were finalized for use in the study: (1) Which hospital did you attend for MCH services? (2) Did you experience any discomfort or stigma when attending MCH services? (3) Can you provide information in terms of frequency, who displayed stigmatizing behaviors, and in what form? (4) How did you feel about this experience? (5) How did you respond to these behaviors? (6) What is your expectation about accessing MCH services? (7) Did you experience other barriers to accessing MCH services?", "Village headmen were informed about the study and asked to select participants five days in advance according to the inclusion criteria. The participants were purposively selected from seven hill tribe villages. Hill tribe women who were postdelivery one year prior who had experienced stigma when attending an MCH clinic and who able to use Thai met the inclusion criteria. Women who met the criteria were informed by the village headman and asked to participate in the study. At the date of the interview, women who met the criteria and intended to provide information to the researcher were screened again to determine whether they had evidence according to the criteria. Only those who had a strong experience with stigma were invited to an interview. All participants were provided with information about the study and signed written consent forms that stated the voluntary nature of participation. Three researchers who were trained in qualitative methods (one female medical anthropologist (Ph.D.), one female health behavioral scientist (Ph.D.), and one male public health expert (Ph.D.)) and working as university faculty were the interviewers. All interviewers were women who were familiar from previous projects with the hill tribe people living in these areas.", "Face-to-face interviews were conducted in a private and confidential room at the community hall in each village between June and September 2021. A question guide was used in the interview. Before the interview, the participants were asked for permission to record it and take field notes. The interviews started with the objective of research and general questions about maternal and child health. The specific questions on asking about the experience of stigma while attending MCH services were followed. Each interview lasted for 45 min. All methods were carried out in accordance with the Declaration of Helsinki [35] in the ethical principles for medical research that involve human subjects.", "All records were transcribed and checked before further analysis. The transcript was sent to all participants who were the information owner to check its accuracy before further analysis. The information in the transcripts was coded, and coding trees were developed. The codes were transferred into the NVivo program (NVivo, qualitative data analysis software; QSR International Pty Ltd., version 11, 2015) for theme extraction. A content analysis was used to extract major and minor themes with the inductive method, which usually uses the keywords presented from interviews to construct the themes. The major theme was used to present the form or pattern of the stigma while attending MCH services. The minor theme was focused on the other significant surrounding information, including the experiences in addressing the stigma, and expectations of the participants to further solve the problem. All themes identified were constructed and formed. Significant quotations were presented to support the findings.", "Before deciding on final interpretations, the researchers once again sent the information back to the participant who was the information owner to ensure the accuracy of the final findings. Two qualitative research experts in the field were asked to validate the final findings and framework (Fig. 1). The final framework was discussed and validated again with eight local Akha people (five women and three community leaders).", "[SUBTITLE] General characteristics of the study population [SUBSECTION] A total of 61 postdelivery women were recruited into the study; the average age was 28.9 years, 32.8% had no Thai ID cards, and 93.4% were married. More than half were Buddhist (55.7%), 67.2% had completed either primary or high school, 59.0% were unemployed, and 44.3% had no regular income. Less than half had been screened for cervical cancer (31.1%) and breast cancer (21.3%) in the previous year (Table 1).\n\nTable 1General characteristics of the participantsn%\nTotal\n61100.0Age mean age = 28.9 years, min = 18, max = 41Identification card (ID card)No2032.8Yes4167.2\nMarital status\nMarried5793.4Ever married46.6\nReligion\nBuddhist3455.7Christian2744.3\nEducation\nNo education1524.6Primary school1626.2High school2541.0Diploma34.9Bachelor’s degree23.3\nOccupation\nUnemployed3659.0Daily wage job1321.3Farmer914.8Merchant34.9Income (bath)No income2744.3≤1,00011.61,001–5,0001524.65,001–10,0001219.7≥10,00169.8\nNumber of children\nmean = 2.3, min = 1, max = 9\nNumber of pregnancies\nmean = 2.4, min = 1, max = 12\nHad been screened for cervical cancer one year prior\nNo4268.9Yes1931.1\nHad been screened for breast cancer one year prior\nNo4878.7Yes1321.3\n\nGeneral characteristics of the participants\nA total of 61 postdelivery women were recruited into the study; the average age was 28.9 years, 32.8% had no Thai ID cards, and 93.4% were married. More than half were Buddhist (55.7%), 67.2% had completed either primary or high school, 59.0% were unemployed, and 44.3% had no regular income. Less than half had been screened for cervical cancer (31.1%) and breast cancer (21.3%) in the previous year (Table 1).\n\nTable 1General characteristics of the participantsn%\nTotal\n61100.0Age mean age = 28.9 years, min = 18, max = 41Identification card (ID card)No2032.8Yes4167.2\nMarital status\nMarried5793.4Ever married46.6\nReligion\nBuddhist3455.7Christian2744.3\nEducation\nNo education1524.6Primary school1626.2High school2541.0Diploma34.9Bachelor’s degree23.3\nOccupation\nUnemployed3659.0Daily wage job1321.3Farmer914.8Merchant34.9Income (bath)No income2744.3≤1,00011.61,001–5,0001524.65,001–10,0001219.7≥10,00169.8\nNumber of children\nmean = 2.3, min = 1, max = 9\nNumber of pregnancies\nmean = 2.4, min = 1, max = 12\nHad been screened for cervical cancer one year prior\nNo4268.9Yes1931.1\nHad been screened for breast cancer one year prior\nNo4878.7Yes1321.3\n\nGeneral characteristics of the participants\n[SUBTITLE] Components of stigma [SUBSECTION] The component of stigma consisted of the stigma driver, stigma facilitator, stigma markers, and stigma manifestations. Several factors were clearly identified as drivers, facilitators, markers, and manifestations of stigma for Akha women seeking MCH services. Different levels of the health outcome impact were reported (Fig. 1).\n[SUBTITLE] A) drivers of stigma for akha women seeking MCH services [SUBSECTION] Being members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services.\nA 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nWhen I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nA 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nIf I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nThe Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience.\nA 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nI am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nA large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services.\nA 28-year-old woman said the following [P#25]:\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\nEven though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women.\nA 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nAt first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nBeing members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services.\nA 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nWhen I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nA 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nIf I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nThe Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience.\nA 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nI am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nA large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services.\nA 28-year-old woman said the following [P#25]:\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\nEven though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women.\nA 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nAt first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\n[SUBTITLE] B) facilitators of stigma for akha women seeking MCH services [SUBSECTION] Several factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps.\nMost health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services.\nA 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nWhen I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nA 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nI feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nOne additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services.\nA 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nWhen I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nA 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nI had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nThe knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator.\nA 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nAnytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nSeveral factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps.\nMost health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services.\nA 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nWhen I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nA 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nI feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nOne additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services.\nA 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nWhen I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nA 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nI had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nThe knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator.\nA 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nAnytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\n[SUBTITLE] C) stigma markers for akha women seeking MCH services [SUBSECTION] Being a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma.\nA 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nI have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nBeing a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma.\nA 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nI have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\n[SUBTITLE] D) manifestations of stigma for akha women seeking MCH services [SUBSECTION] Several manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public.\nVerbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers.\nA 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nMy bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nA 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nLast year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nSome Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery.\nA 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nDuring my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nRefusing to give treatment was also reported among the Akha hill tribe women while attending MCH services.\nA 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nWhen I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nSome Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment.\nA 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nThe staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nSeveral manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public.\nVerbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers.\nA 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nMy bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nA 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nLast year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nSome Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery.\nA 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nDuring my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nRefusing to give treatment was also reported among the Akha hill tribe women while attending MCH services.\nA 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nWhen I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nSome Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment.\nA 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nThe staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\n[SUBTITLE] E) health outcomes [SUBSECTION] Due to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%).\nA 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nWhen I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nA 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\nI had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\n\nFig. 1Stigma frame among hill tribe women who access MCH services\n\nStigma frame among hill tribe women who access MCH services\nDue to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%).\nA 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nWhen I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nA 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\nI had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\n\nFig. 1Stigma frame among hill tribe women who access MCH services\n\nStigma frame among hill tribe women who access MCH services\nThe component of stigma consisted of the stigma driver, stigma facilitator, stigma markers, and stigma manifestations. Several factors were clearly identified as drivers, facilitators, markers, and manifestations of stigma for Akha women seeking MCH services. Different levels of the health outcome impact were reported (Fig. 1).\n[SUBTITLE] A) drivers of stigma for akha women seeking MCH services [SUBSECTION] Being members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services.\nA 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nWhen I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nA 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nIf I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nThe Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience.\nA 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nI am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nA large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services.\nA 28-year-old woman said the following [P#25]:\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\nEven though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women.\nA 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nAt first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nBeing members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services.\nA 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nWhen I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nA 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nIf I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nThe Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience.\nA 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nI am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nA large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services.\nA 28-year-old woman said the following [P#25]:\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\nEven though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women.\nA 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nAt first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\n[SUBTITLE] B) facilitators of stigma for akha women seeking MCH services [SUBSECTION] Several factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps.\nMost health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services.\nA 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nWhen I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nA 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nI feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nOne additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services.\nA 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nWhen I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nA 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nI had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nThe knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator.\nA 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nAnytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nSeveral factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps.\nMost health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services.\nA 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nWhen I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nA 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nI feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nOne additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services.\nA 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nWhen I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nA 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nI had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nThe knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator.\nA 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nAnytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\n[SUBTITLE] C) stigma markers for akha women seeking MCH services [SUBSECTION] Being a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma.\nA 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nI have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nBeing a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma.\nA 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nI have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\n[SUBTITLE] D) manifestations of stigma for akha women seeking MCH services [SUBSECTION] Several manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public.\nVerbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers.\nA 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nMy bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nA 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nLast year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nSome Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery.\nA 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nDuring my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nRefusing to give treatment was also reported among the Akha hill tribe women while attending MCH services.\nA 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nWhen I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nSome Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment.\nA 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nThe staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nSeveral manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public.\nVerbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers.\nA 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nMy bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nA 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nLast year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nSome Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery.\nA 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nDuring my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nRefusing to give treatment was also reported among the Akha hill tribe women while attending MCH services.\nA 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nWhen I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nSome Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment.\nA 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nThe staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\n[SUBTITLE] E) health outcomes [SUBSECTION] Due to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%).\nA 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nWhen I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nA 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\nI had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\n\nFig. 1Stigma frame among hill tribe women who access MCH services\n\nStigma frame among hill tribe women who access MCH services\nDue to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%).\nA 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nWhen I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nA 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\nI had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\n\nFig. 1Stigma frame among hill tribe women who access MCH services\n\nStigma frame among hill tribe women who access MCH services\n[SUBTITLE] Experience in addressing the stigma [SUBSECTION] After discussing Akha hill tibe women experiences of stigma in many forms, they reported their adaptations in addressing the problem in different ways.\n[SUBTITLE] Accepting the situation with no better option [SUBSECTION] The first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time.\nA 34-year-old woman said the following [P#20]:\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\nThe first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time.\nA 34-year-old woman said the following [P#20]:\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n[SUBTITLE] Defending oneself to receive better care and services [SUBSECTION] Some people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs.\nA 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nWhen my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nA 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nA staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nSome people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs.\nA 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nWhen my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nA 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nA staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\n[SUBTITLE] Using a private health care service [SUBSECTION] Some Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages.\nA 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nMy personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nA 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nMoney can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nSome Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages.\nA 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nMy personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nA 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nMoney can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nAfter discussing Akha hill tibe women experiences of stigma in many forms, they reported their adaptations in addressing the problem in different ways.\n[SUBTITLE] Accepting the situation with no better option [SUBSECTION] The first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time.\nA 34-year-old woman said the following [P#20]:\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\nThe first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time.\nA 34-year-old woman said the following [P#20]:\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n[SUBTITLE] Defending oneself to receive better care and services [SUBSECTION] Some people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs.\nA 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nWhen my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nA 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nA staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nSome people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs.\nA 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nWhen my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nA 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nA staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\n[SUBTITLE] Using a private health care service [SUBSECTION] Some Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages.\nA 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nMy personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nA 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nMoney can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nSome Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages.\nA 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nMy personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nA 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nMoney can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\n[SUBTITLE] Expectations [SUBSECTION] Regarding their expectations of MCH services, the Akha hill tribe women requested gender-matched providers in MCH services, especially for Papanicolaou test (PAP) smears, breast cancer screening, and postpartum care. Regularly offered services at the village level were an expectation because the distance from the village to the health care setting posed a major barrier to accessing care, particularly in the rainy season. Emergency mobile clinics were another need of Akha pregnant hill tribe women because when close to delivery, they needed to be able to secure timely access to a hospital, and using their everyday motorcycle was not comfortable and safe for them during late pregnancy. Moreover, the Akha hill tribe women expressed the necessity of basic standard medical equipment at small health-promoting hospitals located at the village level. The Akha women often needed to obtain services at a district hospital for many medical procedures during pregnancy.\nA 39-year-old woman said the following [P#53]:As I am a female, I prefer female staff at the MCH clinic. At least a female understands (another) female, and I can ask her whatever I want to know.\nAs I am a female, I prefer female staff at the MCH clinic. At least a female understands (another) female, and I can ask her whatever I want to know.\nA 28-year-old woman said the following [P#10]:I want at-home postpartum visit service from health staff. Therefore, I would not need to travel to a city. I cannot drive, and my husband has to work at the farm.\nI want at-home postpartum visit service from health staff. Therefore, I would not need to travel to a city. I cannot drive, and my husband has to work at the farm.\nRegarding their expectations of MCH services, the Akha hill tribe women requested gender-matched providers in MCH services, especially for Papanicolaou test (PAP) smears, breast cancer screening, and postpartum care. Regularly offered services at the village level were an expectation because the distance from the village to the health care setting posed a major barrier to accessing care, particularly in the rainy season. Emergency mobile clinics were another need of Akha pregnant hill tribe women because when close to delivery, they needed to be able to secure timely access to a hospital, and using their everyday motorcycle was not comfortable and safe for them during late pregnancy. Moreover, the Akha hill tribe women expressed the necessity of basic standard medical equipment at small health-promoting hospitals located at the village level. The Akha women often needed to obtain services at a district hospital for many medical procedures during pregnancy.\nA 39-year-old woman said the following [P#53]:As I am a female, I prefer female staff at the MCH clinic. At least a female understands (another) female, and I can ask her whatever I want to know.\nAs I am a female, I prefer female staff at the MCH clinic. At least a female understands (another) female, and I can ask her whatever I want to know.\nA 28-year-old woman said the following [P#10]:I want at-home postpartum visit service from health staff. Therefore, I would not need to travel to a city. I cannot drive, and my husband has to work at the farm.\nI want at-home postpartum visit service from health staff. Therefore, I would not need to travel to a city. I cannot drive, and my husband has to work at the farm.", "A total of 61 postdelivery women were recruited into the study; the average age was 28.9 years, 32.8% had no Thai ID cards, and 93.4% were married. More than half were Buddhist (55.7%), 67.2% had completed either primary or high school, 59.0% were unemployed, and 44.3% had no regular income. Less than half had been screened for cervical cancer (31.1%) and breast cancer (21.3%) in the previous year (Table 1).\n\nTable 1General characteristics of the participantsn%\nTotal\n61100.0Age mean age = 28.9 years, min = 18, max = 41Identification card (ID card)No2032.8Yes4167.2\nMarital status\nMarried5793.4Ever married46.6\nReligion\nBuddhist3455.7Christian2744.3\nEducation\nNo education1524.6Primary school1626.2High school2541.0Diploma34.9Bachelor’s degree23.3\nOccupation\nUnemployed3659.0Daily wage job1321.3Farmer914.8Merchant34.9Income (bath)No income2744.3≤1,00011.61,001–5,0001524.65,001–10,0001219.7≥10,00169.8\nNumber of children\nmean = 2.3, min = 1, max = 9\nNumber of pregnancies\nmean = 2.4, min = 1, max = 12\nHad been screened for cervical cancer one year prior\nNo4268.9Yes1931.1\nHad been screened for breast cancer one year prior\nNo4878.7Yes1321.3\n\nGeneral characteristics of the participants", "The component of stigma consisted of the stigma driver, stigma facilitator, stigma markers, and stigma manifestations. Several factors were clearly identified as drivers, facilitators, markers, and manifestations of stigma for Akha women seeking MCH services. Different levels of the health outcome impact were reported (Fig. 1).\n[SUBTITLE] A) drivers of stigma for akha women seeking MCH services [SUBSECTION] Being members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services.\nA 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nWhen I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nA 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nIf I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nThe Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience.\nA 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nI am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nA large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services.\nA 28-year-old woman said the following [P#25]:\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\nEven though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women.\nA 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nAt first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nBeing members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services.\nA 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nWhen I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nA 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nIf I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nThe Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience.\nA 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nI am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nA large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services.\nA 28-year-old woman said the following [P#25]:\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\nEven though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women.\nA 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nAt first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\n[SUBTITLE] B) facilitators of stigma for akha women seeking MCH services [SUBSECTION] Several factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps.\nMost health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services.\nA 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nWhen I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nA 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nI feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nOne additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services.\nA 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nWhen I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nA 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nI had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nThe knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator.\nA 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nAnytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nSeveral factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps.\nMost health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services.\nA 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nWhen I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nA 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nI feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nOne additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services.\nA 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nWhen I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nA 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nI had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nThe knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator.\nA 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nAnytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\n[SUBTITLE] C) stigma markers for akha women seeking MCH services [SUBSECTION] Being a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma.\nA 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nI have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nBeing a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma.\nA 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nI have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\n[SUBTITLE] D) manifestations of stigma for akha women seeking MCH services [SUBSECTION] Several manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public.\nVerbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers.\nA 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nMy bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nA 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nLast year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nSome Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery.\nA 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nDuring my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nRefusing to give treatment was also reported among the Akha hill tribe women while attending MCH services.\nA 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nWhen I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nSome Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment.\nA 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nThe staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nSeveral manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public.\nVerbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers.\nA 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nMy bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nA 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nLast year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nSome Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery.\nA 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nDuring my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nRefusing to give treatment was also reported among the Akha hill tribe women while attending MCH services.\nA 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nWhen I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nSome Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment.\nA 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nThe staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\n[SUBTITLE] E) health outcomes [SUBSECTION] Due to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%).\nA 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nWhen I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nA 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\nI had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\n\nFig. 1Stigma frame among hill tribe women who access MCH services\n\nStigma frame among hill tribe women who access MCH services\nDue to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%).\nA 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nWhen I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nA 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\nI had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\n\nFig. 1Stigma frame among hill tribe women who access MCH services\n\nStigma frame among hill tribe women who access MCH services", "Being members of Akha hill tribe with specific culture, as presented through their language, clothing, poverty, and name, was identified as a driver of stigma for Akha women attending MCH services. Most Akha women spoke Thai as a second language, which is completely different from their native language, and only a few Akha women could speak Thai fluently. Most Akha hill tribe women were not supported in school during their youth compared to men, and limited Thai fluency was very common. Therefore, language issues were one of the drivers of the stigma that the Akha women encountered when attending MCH services.\nA 30-year-old woman said the following [P#08]:When I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nWhen I was waiting for delivery, I saw a hill tribe woman next to me being scolded by a nurse after she screamed because she had great pain from the contractions. I truly felt disappointed with the situation. I understood that being a tribe member could cause trouble in communication with Thai people (nurse). I thought, why did a nurse blame her? The nurse should empathize with us.\nA 34-year-old woman said the following [P#47]:If I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nIf I did not respond to a doctor, it was not because I did not understand what he said, but I could not speak in Thai.\nThe Akha hill tribe women still favored their traditional cultural clothing, which is different from that of Thai people, and Thai people worked as the main health care providers in the hospital. The Akha hill tribe women’s style of dress differentiated them from other people who attended the clinic and was viewed as non-Thai by others, which was identified as being a driver of the stigmatizing experience.\nA 38-year-old woman said the following [P#30]:I am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nI am Akha, and I always wear my traditional costume, including at the time to see the doctor. In addition, I have been looked down on by hospital staff. It might be different from the other (Thai people), but I love to dress in my own traditional style. I feel I am lower than them.\nA large proportion of Akha hill tribe people live in poor economic situations, which is a major consequence of being poorly educated and having scant access to skilled jobs. The Akha hill tribe women identified not being able to speak Thai, dressing in their traditional clothing, and living in a poor family as major drivers of stigma when attending MCH services. Poverty was a strong image for health care providers and was identified as one of the greatest drivers of stigma among Akha hill tribe women attending MCH services.\nA 28-year-old woman said the following [P#25]:\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\n\nDue to being the hill tribes, we were blamed by a nurse, and the blaming statements that I always hear are ‘I don’t understand you, what you said. Why do you have more and more children? I told you many times to get sterilization, but you won’t.’ All of these (statements) make me feel truly uncomfortable.\nEven though some Akha hill tribe women had been educated in Thai schools, were able to speak Thai fluently, and dressed in a modern style, some of them still used their local tribal name, which easily identified them as hill tribe people. Thus, the name provided to health care providers when attending MCH acted as one of the drivers of the stigma encountered by the Akha hill tribe women.\nA 26-year-old woman said the following [P#17]:At first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.\nAt first, the staff did not say anything. She treated me like other people until she saw my surname. She knew automatically that I’m a hill tribe (member). She changed her actions, including speaking with a negative tone. She ordered me to stand away from her and her eyes looked me up and down.", "Several factors were identified as facilitators of the stigma encountered by Akha hill tribe women attending MCH services, specifically, the health care providers’ background, gender differences between clients and health care providers, and knowledge gaps.\nMost health caregivers were Thai and had different cultures and lifestyles with the Akha hill tribe people. Thus, when the Akha hill tribe women attended MCH services, they were treated based on the health care provider’s background, and some practices hurt or embarrassed the Akha hill tribe women. Moreover, the differences in culture, lifestyle and background between health care providers and clients made it easy to misunderstand communications when receiving services.\nA 30-year-old woman said the following [P#31]:When I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nWhen I went to give birth to my first child, I didn’t know what things I had to prepare for the situation. The hospital staff asked me, ‘You do know you are giving birth, so why do you not prepare all the things you need? Are you going to use hospital supplies?’ The way she spoke to me made me feel guilty and ashamed. I didn’t know how to respond to her at that time. I felt so bad, and it hurts me even now.\nA 34-year-old woman said the following [P#47]:I feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nI feel that sometimes, the MCH staff were not happy to serve me. I knew from the words spoken by a nurse. Additionally, a doctor said to me that there were many patients, he didn’t have time to serve only me. I truly understand that the staff have to spend lots of time with us, who don’t know Thai. I wish I could understand and speak Thai fluently.\nOne additional factor that acted as a facilitator of the stigma encountered by Akha hill tribe women attending MCH services was the gender difference between health care providers and clients. Such differences reduced Akha hill tribe women’s comfort and willingness to attend MCH services.\nA 39-year-old woman said the following [P#53]:When I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nWhen I went to give birth to my second child at a hospital a year ago, a male medical student came to see me while giving birth. It made me feel tense, embarrassed and lacking privacy. In my culture, a man should not get involved in a woman’s delivery, even her husband. It’s not an easy thing to deal with this.\nA 31-year-old woman said the following [P#55]:I had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nI had a cervical cancer screening a few months ago. Unexpectedly, the doctor was a male, so I did not want to go to check things anymore. I felt embarrassed. I prefer a female doctor. If I see him again, I won’t know how to behave.\nThe knowledge gap between clients and health care providers was identified as a significant facilitator of stigma for the Akha hill tribe women attending MCH services. Different knowledge related to care and practice during pregnancy and postdelivery was found to facilitate stigma. In general, the Akha hill tribe women engaged in practices based on their own traditional methods to care for themselves during pregnancy and postdelivery, which might run counter to the knowledge and desirable practices proposed by health care providers. Such discrepancies facilitated the experience of stigmatization. The Akha hill tribe women’s poor knowledge also led to a lack of understanding about how to use their rights to access MCH services under the national universal coverage scheme, which was also a stigma facilitator.\nA 32-year-old woman said the following [P#07]:Anytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?\nAnytime I go to see a doctor, I have been treated like I know nothing. Of course, I do not know what he said. For instance, ‘cervical,’ what does it look like? and how’s it important to me? We know nothing, especially medical terms. Additionally, we hill tribe women never attended a school, so then how can we know about that?", "Being a member of a hill tribe, with or without holding a Thai ID card, was viewed as a source of stigma in the Akha hill tribe women’s access to MCH services. Being part of a hill tribe was clearly viewed by health care providers as marking these women as “other.” Moreover, being an individual without a Thai ID card was reported as generating stigma.\nA 31-year-old woman said the following [P#03]:I have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.\nI have been viewed as ‘the other’ for all my life. This is because I am a hill tribe (member). Regardless of whether you have a Thai ID card, you are still a hill tribe (member). I have been facing the inequity of health care services since I was born. They speak to me not ‘to inform’ but ‘to order,’ unlike other Thai people.", "Several manifestations of stigma were identified by the Akha hill tribe women attending MCH services such as verbal abuse, physical abuse, a refusal to provide treatment, and the intentional disclosure of personal information to the public.\nVerbal abuse was very commonly reported among the Akha hill tribe women when attending MCH services. The Akha hill tribe women faced improper, unhealthy, and uncaring speech from health care providers.\nA 22-year-old woman said the following [P#05]:My bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nMy bad experience happened when I went to a hospital with my one-month-old daughter. I was waiting for (the doctor), and I sat in the wrong chair. Then, a nurse asked me to move to another place. She used an inappropriate voice with me. I was thinking about why she did not speak to me with many polite words. I realized that it was because I am a hill tribe (member).\nA 38-year-old woman said the following [P#49]:Last year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nLast year, I went to an MCH clinic at a hospital. The staff asked me to explain my problem. I responded to them in my way, but it might not be clear to them. The staff shouted at me with angry responses.\nSome Akha hill tribe women experienced physical abuse when attending MCH services, such as being hitting on the leg and nurses not caring appropriately for mothers at the moment of delivery.\nA 40-year-old woman said the following [P#16]:During my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nDuring my first childbirth, a nurse forced me to deliver by telling me to push...push and push. I didn’t understand what push is. Therefore, I did not follow the instructions. A few seconds later, a nurse slapped my legs. I cried and still wonder why they did this to me.\nRefusing to give treatment was also reported among the Akha hill tribe women while attending MCH services.\nA 26-year-old woman said the following [P#21]:When I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nWhen I went to work in another province, I chose to go to a health care center near my workplace. When I was there, the staff told me that I did not have the right to receive the services at that center and blamed me for why I was so careless. I felt very sad and asked myself, am I a human? Why cannot the hill tribe people have rights? Why can’t the hill tribe people get health services at other places like the Thai people?\nSome Akha women reported that their private information was disclosed, especially to hired staff from the same village as the clients. Once individual information is released to the public, this violation reduces women’s trust in the service and makes them less likely to pursue further care and treatment.\nA 25-year-old woman said the following [P#45]:The staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.\nThe staff did not care about me. She did not care about my personal information. I had a cervical cancer screening once, and the staff wanted more women in the village to get this screening, so she took me as an example. She told my personal information to others. I am not OK with this.", "Due to the experience of stigma faced by the Akha hill tribe women when attending MCH services, many poor health outcomes were reported, such as poor rates of antenatal and postpartum care, breast cancer screening (21.3%), and cervical cancer screening (47.5%).\nA 41-year-old woman said the following [P#26]:When I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nWhen I had my third child, I had a very bad experience during delivery at a hospital. With ineffective communication between me and a nurse in the process of antenatal care (ANC), I was ignored in careful care and delayed care. Resulting, I had a large blood bleeding or postpartum hemorrhage.\nA 27-year-old woman said the following [P#14]:I had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\nI had a serious experience with access and care during my first pregnancy. I was asked to wait for the delivery for a long time, and they (nurses) even carefully cared for me during the waiting. Finally, I found that my baby was in the breech position, and I was transferred to a tertiary hospital for operation. I have a big question to the nurse and doctor that if I was not attending at a hospital close to a tertiary hospital, what would have happened to me?\n\nFig. 1Stigma frame among hill tribe women who access MCH services\n\nStigma frame among hill tribe women who access MCH services", "After discussing Akha hill tibe women experiences of stigma in many forms, they reported their adaptations in addressing the problem in different ways.\n[SUBTITLE] Accepting the situation with no better option [SUBSECTION] The first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time.\nA 34-year-old woman said the following [P#20]:\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\nThe first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time.\nA 34-year-old woman said the following [P#20]:\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n[SUBTITLE] Defending oneself to receive better care and services [SUBSECTION] Some people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs.\nA 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nWhen my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nA 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nA staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nSome people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs.\nA 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nWhen my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nA 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nA staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\n[SUBTITLE] Using a private health care service [SUBSECTION] Some Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages.\nA 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nMy personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nA 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nMoney can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nSome Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages.\nA 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nMy personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nA 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nMoney can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.", "The first group of Akha hill tribe women reported that they did not have a better option to access MCH services. Some people reported being provided with poor service but being able to accept the suffering caused by mistreatment. Most impacted Akha hill tribe women accepted their suffering due to the fear of being treated even more poorly the next time.\nA 34-year-old woman said the following [P#20]:\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”\n\n“I did not have any response when the staff made me feel uncomfortable or feeling angry by shouting at me or looking down on me. I accepted everything, as I knew I am a hill tribe (woman) who needs help from them. In addition, only they can help me. If I respond to them in bad way, I am afraid that they will not care well for me and my child.”", "Some people preferred to stand up to mistreatment and voice their needs. Their responses to health care providers took different forms, such as reminding the health care providers of their responsibilities, asking the reasons why their case was being ignored, and asking for further information regarding their needs.\nA 25-year-old woman said the following [P#06]:When my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nWhen my first child was born, I remembered that a staff member expressed their negative emotion to me. She tried to talk to me many times, and I felt like I received bad service from this hospital. I asked them to treat me better in their practice. I promise myself that if I have my next child, I won’t come to this hospital.\nA 33-year-old woman said the following [P#23]:A staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.\nA staff member spoke to me impolitely. She told me how to feed my baby. I asked her many times to make sure that I understood the right things. However, at the end, she shouted at me, ‘Don’t you understand?’ I responded immediately, ‘You have your responsibilities to me, or do you want me to complain to your director?’ Then, she changed her emotion and spoke to me nicely.", "Some Akha hill tribe women preferred to obtain services from a private hospital. Generally, at a private hospital, most services are provided based on need. The service mindset of health care providers at private hospitals was perceived as much better than that at public hospitals, which could be due to the impacts of policy and organizational advantages.\nA 31-year-old woman said the following [P#40]:My personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nMy personal opinion, if I have a lot of money, I prefer to get health care services from a private hospital. The staff will not look down on me.\nA 34-year-old woman said the following [P#47]:Money can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.\nMoney can help everything. If I had enough money, I would not go to a public hospital but rather to a private hospital without facing stigma.", "Regarding their expectations of MCH services, the Akha hill tribe women requested gender-matched providers in MCH services, especially for Papanicolaou test (PAP) smears, breast cancer screening, and postpartum care. Regularly offered services at the village level were an expectation because the distance from the village to the health care setting posed a major barrier to accessing care, particularly in the rainy season. Emergency mobile clinics were another need of Akha pregnant hill tribe women because when close to delivery, they needed to be able to secure timely access to a hospital, and using their everyday motorcycle was not comfortable and safe for them during late pregnancy. Moreover, the Akha hill tribe women expressed the necessity of basic standard medical equipment at small health-promoting hospitals located at the village level. The Akha women often needed to obtain services at a district hospital for many medical procedures during pregnancy.\nA 39-year-old woman said the following [P#53]:As I am a female, I prefer female staff at the MCH clinic. At least a female understands (another) female, and I can ask her whatever I want to know.\nAs I am a female, I prefer female staff at the MCH clinic. At least a female understands (another) female, and I can ask her whatever I want to know.\nA 28-year-old woman said the following [P#10]:I want at-home postpartum visit service from health staff. Therefore, I would not need to travel to a city. I cannot drive, and my husband has to work at the farm.\nI want at-home postpartum visit service from health staff. Therefore, I would not need to travel to a city. I cannot drive, and my husband has to work at the farm.", "The Akha women who were pregnant or had delivered their infant one year prior and had attended MCH services generally had a poor socioeconomic status. They faced many forms of stigma when attending MCH services in the previous year, such as being spoken to impolitely or with hostility, being physically abused, being refused treatment, and having their personal information intentionally disclosed to the public. Several factors were extracted as the drivers and facilitators of such stigma. Being a member of the Akha people was a stigma marker. Poor rates of accessing MCH services and breast and cervical cancer screening were found among the Akha hill tribe women who needed to access these services. Some people accepted the suffering caused by the stigmatizing experience, while others preferred to ask for better care and service, including seeking care at a private health care provider. Most Akha women expected to have gender-matched health care providers, active and mobile emergency clinics, and full, standard medical equipment at hospitals located in their village. They also wanted an active health care service.\nIn our study, speaking Akha, wearing traditional dress, being poor, and having tribal names were found to be the drivers of stigma for Akha women when accessing MCH services, especially in public hospitals in Thailand. These factors were very specific stigma scenarios among the Akha hill tribe women when accessing public hospitals in Thailand. These drivers are very difficult to address fully because it is often impossible for Akha people aged 25 years and older to be fluent Thai speakers because they have never been to school and are unable to speak Thai [23]. Changing their traditional dress is also unlikely to be undertaken, since Akha hill tribe women strongly prefer to dress in their own style, which relates to their culture and religion [36]. Moreover, it is very difficult to improve Akha hill tribe family income in the near future given tribe members’ current education status and their nearly universal lack of training in professional skills [30].\nAll health care providers being Thai, knowledge gaps between health care providers and clients, and gender mismatch with the client were found to be facilitators of stigma. These findings are supported by a study by Nyblade et al. [32], which reported that stigma greatly impacts access to health care services, especially stigma related to different characteristics or backgrounds and knowledge gaps between health caregivers and clients, which eventually lead to poor health outcomes for the population. Many personal characteristics, including the attitudes and behaviors of health care providers, have been reported as facilitators of stigma, especially when directed at vulnerable populations [1, 36, 37].\nWhen attending MCH services in the public hospitals in Thailand, the majority of Akha hill tribes reported experiencing stigma through verbal and physical abuse, a refusal to provide proper treatment and the release of individual information to the public. A high frequency of verbal abuse from health care providers was reported. This point was the strongest and resulted in immense harm to the Akha hill tribe women who attended MCH services in public hospitals. Many questions arose that expressed how this abuse could happen to these people, especially questions regarding why people become nurses or doctors if they will not provide appropriate service or care to people suffering from health problems. The Akha hill tribe women commented that if individuals did not have a health problem, they would not go to a hospital. Verbal abuse from health care providers toward the Akha women made them feel confused about these professionals. Physical abuse, a refusal of treatment and the public sharing of client information were reported in some cases, but none of these should occur in health care settings. Such behavior is contrary to the ethics of health professionals according to the WHO guidelines [38] and the principles of health care ethics [39]. People should receive standard care regardless of their race, tribe, or sociodemographic status.\nDue to the stigma encountered by the Akha women when attending an MCH clinic in a public hospital, poor access to the services and poor rates of cervical and breast cancer screening were reported. These findings were supported by a study in two district public hospitals located in the hill tribe villages in northern Thailand, which reported a very poor rate of antenatal care (ANC), cervical and breast cancer screening, and other activities related to women’s reproductive health [40]. In 2018, the Thai Ministry of Public Health [41] reported 8,622 new annual cases of cervical cancer and 5,015 cervical cancer-related deaths among women. In our study, the cervical cancer screening rate among the Akha women was 31.1% compared to 45.6% for Thai people [42].\nMost activities at MCH services do not require serious medical attention. Being screened for breast and cervical cancer, given the low rate of these diseases, was not viewed as a serious problem by either clients or health care providers. The lack of regular visits to MCH services during pregnancy also did not have serious consequences, which made the Akha women less concerned and consequently, less likely to engage in activities related to reproductive health and MCH. In addition, given the stigma experienced when accessing MCH services, the patients did not expect to visit a large hospital. The Akha women favored visiting a small hospital in their village. At such hospitals, having standard medical equipment and care at the village level were their main expectations. Moreover, access to MCH services at small hospitals located in their village made accessing them less time-consuming and eliminated the need to travel and other barriers posed by the more complicated process of accessing services in a large, urban hospital. Another expectation was being attended by a gender-matched health care provider when receiving MCH services, which could reduce stigma and improve women’s personal perception of services.\nSeveral limitations were detected in the study. First, although no participants refused to participate in the study or provide information, given the familiarity between the Akha women and the health care workers at the small hospital in their village, many women preferred not to voice too many of their negative experiences with these health care providers. Thus, most of the negative experiences discussed occurred with health care providers who were working in large, urban hospitals. Second, the participants in the study were recruited from seven hill tribe villages, thus, the study primarily presents information and experiences from the Akha people in northern Thailand. In addition, some key informants experienced stigma some time ago, which could have impacted their recall of certain points during the interview and might affect the interpretation. Finally, because the participant selection was primarily executed by the village headmen, it is possible that some people who had experienced serious and direct stigma that is relevant to the research question might not have been selected. Having a clear and careful selection of participants for the study is one of the critical processes in the qualitative method.", "Akha women who attend a public MCH clinic in Thailand suffer stigma driven by their specific characteristics, such as being unable to speak Thai (or lacking fluency), being poor, wearing their traditional dress and using traditional naming conventions. Health care providers’ background, gender mismatches and knowledge gaps between health care providers and clients were identified as facilitators of stigma for women seeking MCH services. Akha women face many forms of stigma when receiving clinical services, including verbal and physical abuse, a refusal to provide treatment, and the intentional disclosure of their personal information to the public. As a result, such mistreatment affects multiple health and health service outcomes, such as poor rates of attending antenatal care and low rates of cervical and breast cancer screening. Akha hill tribe women use many approaches to adjust to the stigma that they encounter, such as accepting the situation with no better option, defending themselves to obtain better care and services, and using a private clinic instead. Akha women expect to have active services and gender-matched health care providers at MCH services and mobile emergency clinics and that the appropriate equipment be provided to equip standard MCH services at hospitals located in or near the hill tribe villages.\nSerious consideration of the problems posed by the stigmatization for Akha hill tribe women is needed to improve their access to health care services, particularly those attending MCH services. Policies with standard protocols to ensure the provision of equal care for everyone should be implemented. The improvement of health facilities located in hill tribe villages should also be considered to familiarize hill tribe people with health care providers in their home setting. Moreover, encouraging hill tribe people to be trained as nurses or medical doctors and then sending them back to work at a hospital in their village could eventually reduce stigma.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2" ]
[ null, null, null, null, null, null, null, null, "results", null, null, null, null, null, null, null, null, null, null, null, null, "discussion", "conclusion", "supplementary-material", null ]
[ "Akha", "Hill tribe", "Stigma", "Experience", "Maternal and Child Health Care" ]
Parental satisfaction and its associated factors towards neonatal intensive care unit service: a cross-sectional study.
36261864
Parental satisfaction is a well-established outcome indicator and tool for assessing a healthcare system's quality, as well as input for developing strategies for providing acceptable patient care. This study aimed to assess parental satisfaction with neonatal intensive care unit service and its associated factors.
BACKGROUND
A cross-sectional study design was conducted on parents whose neonates were admitted to the neonatal intensive care unit at Debre Tabor Comprehensive Specialized Hospital, in North Central Ethiopia. Data were collected by adopting an EMPATHIC-N instrument during the day of neonatal discharge, after translating the English version of the instrument to the local language (Amharic). Both Bivariable and multivariable logistic analyses were done to identify factors associated with parental satisfaction with neonatal intensive care unit service. P < 0.05 with 95% CI was considered statistically significant.
METHOD
The data analysis was done on 385 parents with a response rate of 95.06%. The overall average satisfaction of parents with neonatal intensive care unit service was 47.8% [95% CI= (43.1-52.5)]. The average parental satisfaction of neonatal intensive care unit service in the information dimension was 50.40%; in the care and treatment dimension was 36.9%, in the parental participation dimension was 50.1%, in the organization dimension was 59.0% and the professional attitude dimension was 48.6%. Gender of parents, residency, parental hospital stay, birth weight, and gestational age were factors associated with parental satisfaction.
RESULTS
There was a low level of parental satisfaction with neonatal intensive care unit service. Among the dimensions of EMPATHIC-N, the lowest parental satisfaction score was in the care and treatment while the highest parental satisfaction score was in the organization dimension.
CONCLUSION
[ "Infant, Newborn", "Humans", "Intensive Care Units, Neonatal", "Personal Satisfaction", "Cross-Sectional Studies", "Parents", "Quality of Health Care" ]
9583552
Introduction
Neonatal intensive care units (NICUs) are areas that require careful risk management with a wide range of neonatal care services [1–3]. It necessitates high-cost and efficient critical care delivery with a multidisciplinary team approach that focuses on preventive strategies for improved outcomes [4–6]. Parental tension and emotions are high when their child is admitted to a neonatal intensive care unit (NICU) due to serious illnesses [7, 8]. Satisfaction is a belief and attitude about a specific service provision of an institution. Parental and patient satisfaction has become a well-established outcome indicator and tool for assessing a healthcare system’s quality, as well as input for developing strategies and providing accessible, sustainable, economical, as well as acceptable patient care [7, 9, 10]. Parental satisfaction reflects the balance between their expectations of ideal care and their perception of real and available care [3, 11, 12]. It is also one of the objectives and missions of every health care center that gives NICU care service [10, 11]. Parent and patient satisfaction has become an important aspect of hospital management initiatives for quality assurance and accreditation. Also, parental involvement has an important role in the delivery of high-quality care, ranging from assisting with activities of daily living to being directly involved in important health care decisions [13, 14]. However, parental participation may not always be possible, but, effective communication will reduce the effects of crises and improve parental satisfaction [11, 15]. Ethiopia has been working to enhance its healthcare delivery systems by focusing on quality healthcare service giving special attention to mothers and children. The Ethiopian Health Service Alliance for Quality has agreed that the initial priority area would be self-motivated and transparent partnerships that stimulate innovation in health care quality management and learning across all hospitals [16]. Given the scarcity of studies on parental satisfaction with NICU care in Ethiopia, as well as clinical observations of parents complaining about NICU care, it is critical to determine the level of parental satisfaction. So, this study aimed to identify the level of parental satisfaction and associated factors with NICU care service in Debre Tabor Comprehensive Specialized Hospital (DTCSH). Also, this study helps the administrators to understand the deficiencies of the hospital’s NICU services, and then propose corresponding improvement strategies.
null
null
Results
This study was conducted on a total of 385 parents, with a 95.06% response rate. The majority of the parents were mothers 224 (58.2%), whereas 322 (83.6%) were married. Full-term newborns 325(84.4%) and those with respiratory issues 115 (29.9%) had the highest number of NICU admissions (Table 1). Table 1Socio-demographic characteristics of study participants (n = 385)VariablesFrequency (n)Percentage (%) Parental gender  Mother22458.2 Father16141.8 Age  18–2413234.3 25–3916643.1 40 and above8722.6 Marital status  Married32283.6 Not married6316.4 Residency  Urban18748.6 Rural19851.4 Educational level  No formal education11930.9 Elementary school9223.9 Secondary school8321.6 Collage and above9123.6 Profession  Housewife7018.2 Farmer10627.5 Student5915.3 Gov’t employ7218.7 Private employ7820.3 Parental hospital stay  ≤ 15 days24463.4 > 15 days14136.6 Neonatal gender  Male18848.8 Female19751.2 Birth weight  High birth weight8321.5 Normal birth weight16242.1 Low birth weight14036.4 Gestational age  Premature6015.6 Full term32584.4 Neonatal hospital stay  ≤ 15 days23561 > 15 days15039 Admission diagnosis  Infection6516.9 Respiratory problems11529.9 Prematurity5915.3 Gastrointestinal problems389.9 Jaundice4712.2 Neurological problems215.5 Cardiological problems266.8 Others143.5 Socio-demographic characteristics of study participants (n = 385) [SUBTITLE] Explanatory factor analysis for subscales of parental satisfaction with NICU-care service [SUBSECTION] Before assessing parental satisfaction levels, confirmatory factor analysis was used to ensure that the measured variables accurately represented the constructs. The factor correlation matrix was used in the analysis. KMO and Bartlett’s tests of sphericity were used to check the correlation of measurement variables, with a KMO value of 0.875 and Bartlett’s tests of sphericity (p = 0.00) respectively. The correlation matrix was checked for commonality extraction, and all item values were larger than 0.3. The parallel analysis determined the number of item components (factors), and two new components for parental participation, three new components for information and organization, and four new components for Care & Treatment and professional attitude of eigenvalues met the criteria, and the Varimax component correlation matrix was an appropriate model (Table 2). Table 2Factor loadings and identified components’ of EMPATHIC-N toolItemFactors 1 2 3 4 Information  What level of doctor’s information do you have regarding the child’s expected health outcomes?0.752 How satisfied are you with the physicians’ and nurses’ information similarity?0.704 How are you satisfied with doctors’ and nurses’ honesty in providing information?0.59 How satisfied are you with daily discussions with doctors and nurses about your child’s care and treatment?0.58 How understandable was the information provided by the doctors and nurses?0.763 How satisfied were you with the correct information when the child’s physical condition deteriorated?0.4280.657 How satisfied were you with the clear answers to your questions?0.653 How clear is the doctor’s information about the consequences of the child’s treatment?0.533 To what extent do you receive clear information about the examinations and tests?0.713 To what extent the information brochure received was complete and clear?0.641 How much clear information is given regarding a child’s illness?0.626 Level of received understandable information about the effects of the drugs?0.606 Care & Treatment  Level of child’s comfort taken into account by the doctors and nurses?0.826 The extent of satisfaction During acute situations on availability of nurses to support?0.822 Level of team alertness to the prevention and treatment of pain of neonate?0.797 Level of care taken to nurses while in the incubator/bed?0.6340.501 Level of correct medication always administered on time?0.6310.402 level of emotional support that has been provided?0.84 The doctors and nurses responded well to our own needs0.809 Transferals of care from the neonatal intensive care unit staff to colleagues in the high-care unit or pediatric ward had gone well?0.782 Every day we knew who of the doctors and nurses was responsible for our child.0.695 How closely did doctors and nurses collaborate during work?0.5780.466 Level of a common goal: to provide the finest care and treatment for our child and ourselves.0.774 Level of physicians and nurses paid close attention to our child’s development?0.756 The team as a whole was concerned for our child and you.0.698 Our child’s requirements were met promptly0.455 The extent of doctors’ and nurses’ professional knowledge of what they are doing?0.847 How satisfied are you with the doctors’ and nurses’ understanding of the child’s medical history at the time of admission?0.765 How satisfied are you with the rapid actions taken by doctors and nurses when a child’s condition deteriorated?0.723 Parental Participation  How involved are you in making decisions about our child’s care and treatment?0.803 The nurses had trained us on the specific aspects of newborn care.0.796 We were encouraged to stay close to our children.0.714 Before discharge, the care for our child was once more discussed with us.0.617 Even during intensive procedures, we could always stay close to our child.0.841 The nurses stimulated us to help in the care of our child0.837 The nurses helped us in the bonding with our child0.5320.575 We had confidence in the team0.561 Organization  The Neonatology unit made us feel safe0.885 There was a warm atmosphere in the Neonatology unit without hostility0.805 The Neonatology unit was clean0.7330.404 The unit could easily be reached by telephone0.809 Our child’s incubator or bed was clean0.8 The team worked efficiently0.661 There was enough space around our child’s incubator/bed0.9 Noise in the unit was muffled as good as possible0.788 Professional Attitude  Our child’s health always came first for the doctors and nurses0.799 The team worked hygienically0.747 Our cultural background was taken into account0.728 The doctors and nurses always took time to listen to us0.876 We felt welcome by the team0.804 Despite the workload, sufficient attention was paid to our child and us by the team0.5840.508 Nurses and doctors always introduced themselves by name and function0.794 We received sympathy from the doctors and nurses0.786 At our bedside, the discussion between the doctors and nurses was only about our child.0.4570.582 The team respected the privacy of our children and us.0.874 There was a pleasant atmosphere among the staff0.743 The team showed respect for our child and us0.5190.609 Factor loadings and identified components’ of EMPATHIC-N tool Before assessing parental satisfaction levels, confirmatory factor analysis was used to ensure that the measured variables accurately represented the constructs. The factor correlation matrix was used in the analysis. KMO and Bartlett’s tests of sphericity were used to check the correlation of measurement variables, with a KMO value of 0.875 and Bartlett’s tests of sphericity (p = 0.00) respectively. The correlation matrix was checked for commonality extraction, and all item values were larger than 0.3. The parallel analysis determined the number of item components (factors), and two new components for parental participation, three new components for information and organization, and four new components for Care & Treatment and professional attitude of eigenvalues met the criteria, and the Varimax component correlation matrix was an appropriate model (Table 2). Table 2Factor loadings and identified components’ of EMPATHIC-N toolItemFactors 1 2 3 4 Information  What level of doctor’s information do you have regarding the child’s expected health outcomes?0.752 How satisfied are you with the physicians’ and nurses’ information similarity?0.704 How are you satisfied with doctors’ and nurses’ honesty in providing information?0.59 How satisfied are you with daily discussions with doctors and nurses about your child’s care and treatment?0.58 How understandable was the information provided by the doctors and nurses?0.763 How satisfied were you with the correct information when the child’s physical condition deteriorated?0.4280.657 How satisfied were you with the clear answers to your questions?0.653 How clear is the doctor’s information about the consequences of the child’s treatment?0.533 To what extent do you receive clear information about the examinations and tests?0.713 To what extent the information brochure received was complete and clear?0.641 How much clear information is given regarding a child’s illness?0.626 Level of received understandable information about the effects of the drugs?0.606 Care & Treatment  Level of child’s comfort taken into account by the doctors and nurses?0.826 The extent of satisfaction During acute situations on availability of nurses to support?0.822 Level of team alertness to the prevention and treatment of pain of neonate?0.797 Level of care taken to nurses while in the incubator/bed?0.6340.501 Level of correct medication always administered on time?0.6310.402 level of emotional support that has been provided?0.84 The doctors and nurses responded well to our own needs0.809 Transferals of care from the neonatal intensive care unit staff to colleagues in the high-care unit or pediatric ward had gone well?0.782 Every day we knew who of the doctors and nurses was responsible for our child.0.695 How closely did doctors and nurses collaborate during work?0.5780.466 Level of a common goal: to provide the finest care and treatment for our child and ourselves.0.774 Level of physicians and nurses paid close attention to our child’s development?0.756 The team as a whole was concerned for our child and you.0.698 Our child’s requirements were met promptly0.455 The extent of doctors’ and nurses’ professional knowledge of what they are doing?0.847 How satisfied are you with the doctors’ and nurses’ understanding of the child’s medical history at the time of admission?0.765 How satisfied are you with the rapid actions taken by doctors and nurses when a child’s condition deteriorated?0.723 Parental Participation  How involved are you in making decisions about our child’s care and treatment?0.803 The nurses had trained us on the specific aspects of newborn care.0.796 We were encouraged to stay close to our children.0.714 Before discharge, the care for our child was once more discussed with us.0.617 Even during intensive procedures, we could always stay close to our child.0.841 The nurses stimulated us to help in the care of our child0.837 The nurses helped us in the bonding with our child0.5320.575 We had confidence in the team0.561 Organization  The Neonatology unit made us feel safe0.885 There was a warm atmosphere in the Neonatology unit without hostility0.805 The Neonatology unit was clean0.7330.404 The unit could easily be reached by telephone0.809 Our child’s incubator or bed was clean0.8 The team worked efficiently0.661 There was enough space around our child’s incubator/bed0.9 Noise in the unit was muffled as good as possible0.788 Professional Attitude  Our child’s health always came first for the doctors and nurses0.799 The team worked hygienically0.747 Our cultural background was taken into account0.728 The doctors and nurses always took time to listen to us0.876 We felt welcome by the team0.804 Despite the workload, sufficient attention was paid to our child and us by the team0.5840.508 Nurses and doctors always introduced themselves by name and function0.794 We received sympathy from the doctors and nurses0.786 At our bedside, the discussion between the doctors and nurses was only about our child.0.4570.582 The team respected the privacy of our children and us.0.874 There was a pleasant atmosphere among the staff0.743 The team showed respect for our child and us0.5190.609 Factor loadings and identified components’ of EMPATHIC-N tool [SUBTITLE] Reliability of items of EMPATHIC-N tool [SUBSECTION] The total EMPATHIC-N values and the five domains showed a good level of internal consistency. The five domains’ Cronbach’s values range from 0.639 to 0.791, whereas the total EMPATHIC-N Cronbach’s value was 0.904. The inter-item correlation (IIC) of the five domains had significant internal consistency (Table 3). Table 3Reliability of items of parental satisfaction with NICU-care serviceDimensionNumber of ItemsΧηρονβαχη αMean dimension score (SD)Maximum possible dimension scoreInter-item correlation (IIC)Item-discriminant validity (IDV)Information120.63939.55(8.42)720.22–0.48*0.20–0.57Care & Treatment170.79181.70(20.49)1700.14–0.64*0.25–0.38Parental Participation80.75524.86(7.79)480.01–0.71*0.37–0.54Organization80.72925.22(7.39)480.11–0.64*0.34–0.69Professional Attitude120.69438.106 (9.07)720.13–0.56*0.22–0.45EMPATHIC-N tool570.904209.44(42.82)410cc*Significant value (p < 0.001), c = not computable Reliability of items of parental satisfaction with NICU-care service *Significant value (p < 0.001), c = not computable The total EMPATHIC-N values and the five domains showed a good level of internal consistency. The five domains’ Cronbach’s values range from 0.639 to 0.791, whereas the total EMPATHIC-N Cronbach’s value was 0.904. The inter-item correlation (IIC) of the five domains had significant internal consistency (Table 3). Table 3Reliability of items of parental satisfaction with NICU-care serviceDimensionNumber of ItemsΧηρονβαχη αMean dimension score (SD)Maximum possible dimension scoreInter-item correlation (IIC)Item-discriminant validity (IDV)Information120.63939.55(8.42)720.22–0.48*0.20–0.57Care & Treatment170.79181.70(20.49)1700.14–0.64*0.25–0.38Parental Participation80.75524.86(7.79)480.01–0.71*0.37–0.54Organization80.72925.22(7.39)480.11–0.64*0.34–0.69Professional Attitude120.69438.106 (9.07)720.13–0.56*0.22–0.45EMPATHIC-N tool570.904209.44(42.82)410cc*Significant value (p < 0.001), c = not computable Reliability of items of parental satisfaction with NICU-care service *Significant value (p < 0.001), c = not computable [SUBTITLE] Parental satisfaction level with NICU-care service [SUBSECTION] Overall mean satisfaction level of parents in NICU-care service was 47.8% [95% CI= (43.1–52.5)]. The domains of parental satisfaction scores were compared with each other and, the lowest parental satisfaction score was for the domain of care and treatment 36.9% while the organizational domain showed the highest parental satisfaction level 59.0%. The domains of information, parental participation, and professional attitude showed comparable parental satisfaction scores (Fig. 1). Fig. 1Parents’ overall and dimensional satisfaction with NICU-care services Parents’ overall and dimensional satisfaction with NICU-care services Overall mean satisfaction level of parents in NICU-care service was 47.8% [95% CI= (43.1–52.5)]. The domains of parental satisfaction scores were compared with each other and, the lowest parental satisfaction score was for the domain of care and treatment 36.9% while the organizational domain showed the highest parental satisfaction level 59.0%. The domains of information, parental participation, and professional attitude showed comparable parental satisfaction scores (Fig. 1). Fig. 1Parents’ overall and dimensional satisfaction with NICU-care services Parents’ overall and dimensional satisfaction with NICU-care services [SUBTITLE] Factors associated with the overall parental satisfaction of NICU care service [SUBSECTION] In this study, the bivariable logistic regression showed that parental gender, residency, parental hospital stay, neonatal hospital stay, birth weight, and gestational age were factors with a p-value of less than 0.2 and were fitted with a multivariable logistic regression model. However, neonatal hospital stay was not associated with multivariable logistic regression with a p-value greater than 0.05. The multivariable logistic analyses showed that mothers were 2.16 (AOR = 2.16; 95%CI: 1.28–3.63) times more satisfied than fathers. Also, parents who are from the rural area were 2.94 (AOR = 2.94; 95%CI: 1.42–6.06) more satisfied than urban. Parents who say less than 15 days were 2.18 (AOR = 2.18; 95%CI: 1.13–4.20) times more satisfied than parents who stay 15 or more days in the hospital. Also, parents of a neonate with a normal birth weight of 2.14 (AOR = 2.14; 95%CI: 1.16–3.94) and full-term neonate 2.53(AOR = 2.53; 95%CI: 1.29–4.97) times more satisfied than their counterparts (Table 4). Table 4Factors associated with satisfaction of parents in NICU-care service (n = 385)VariablesSatisfaction level on NICU- serviceCrude odds ratioAdjusted odds ratiop-valueSatisfiedNot Satisfied(95% CI)(95% CI) Gender  Mother117(52.2%)107(47.8%)1.53(1.02,2.31)2.16(1.28,3.63)0.004* Father67(41.6%)94(58.4%)1 Residency  Urban68(36.4%)119(63.6%)1 Rural116(58.6%)82(41.4%)2.48(1.64,3.73)2.94(1.42,6.06)0.004* Parental hospital stay  ≤ 15 days140(57.4%)104(42.6%)2.97(1.92,4.59)2.18(1.13,4.20)0.019* > 15days44(31.2%)97(68.8%)1 Birth weight  High birth weight34(41.0%)49(59.0%)1 Normal birth weight96(59.3%)66(40.7%)2.09(1.22,3.59)2.14(1.16,3.94)0.015* Low birth weight54(38.6%)86(61.4%)0.91(0.52,1.58)0.76(0.41,1.41)0.38 Gestational age  Premature18(30.0%)42(70.0%)1 Full term166(51.1%)159(48.9%)2.44(1.35,4.41)2.53(1.29,4.97)0.007**= p-value < 0.05 1 = reference BWt-birth weightNote: p-values were extracted from the multivariate logistic regression model Factors associated with satisfaction of parents in NICU-care service (n = 385) *= p-value < 0.05 1 = reference BWt-birth weight Note: p-values were extracted from the multivariate logistic regression model In this study, the bivariable logistic regression showed that parental gender, residency, parental hospital stay, neonatal hospital stay, birth weight, and gestational age were factors with a p-value of less than 0.2 and were fitted with a multivariable logistic regression model. However, neonatal hospital stay was not associated with multivariable logistic regression with a p-value greater than 0.05. The multivariable logistic analyses showed that mothers were 2.16 (AOR = 2.16; 95%CI: 1.28–3.63) times more satisfied than fathers. Also, parents who are from the rural area were 2.94 (AOR = 2.94; 95%CI: 1.42–6.06) more satisfied than urban. Parents who say less than 15 days were 2.18 (AOR = 2.18; 95%CI: 1.13–4.20) times more satisfied than parents who stay 15 or more days in the hospital. Also, parents of a neonate with a normal birth weight of 2.14 (AOR = 2.14; 95%CI: 1.16–3.94) and full-term neonate 2.53(AOR = 2.53; 95%CI: 1.29–4.97) times more satisfied than their counterparts (Table 4). Table 4Factors associated with satisfaction of parents in NICU-care service (n = 385)VariablesSatisfaction level on NICU- serviceCrude odds ratioAdjusted odds ratiop-valueSatisfiedNot Satisfied(95% CI)(95% CI) Gender  Mother117(52.2%)107(47.8%)1.53(1.02,2.31)2.16(1.28,3.63)0.004* Father67(41.6%)94(58.4%)1 Residency  Urban68(36.4%)119(63.6%)1 Rural116(58.6%)82(41.4%)2.48(1.64,3.73)2.94(1.42,6.06)0.004* Parental hospital stay  ≤ 15 days140(57.4%)104(42.6%)2.97(1.92,4.59)2.18(1.13,4.20)0.019* > 15days44(31.2%)97(68.8%)1 Birth weight  High birth weight34(41.0%)49(59.0%)1 Normal birth weight96(59.3%)66(40.7%)2.09(1.22,3.59)2.14(1.16,3.94)0.015* Low birth weight54(38.6%)86(61.4%)0.91(0.52,1.58)0.76(0.41,1.41)0.38 Gestational age  Premature18(30.0%)42(70.0%)1 Full term166(51.1%)159(48.9%)2.44(1.35,4.41)2.53(1.29,4.97)0.007**= p-value < 0.05 1 = reference BWt-birth weightNote: p-values were extracted from the multivariate logistic regression model Factors associated with satisfaction of parents in NICU-care service (n = 385) *= p-value < 0.05 1 = reference BWt-birth weight Note: p-values were extracted from the multivariate logistic regression model
Conclusion
There was a low level of parental satisfaction with neonatal intensive care unit service. Among the dimensions of EMPATHIC-N, the lowest parental satisfaction score was in the care and treatment while the highest parental satisfaction score was in the organization dimension. As a result, health professionals and hospital administrators should collaborate to improve NICU services to provide high-quality service and satisfy parents.
[ "Methods and materials", "Study area and period", "Inclusion and exclusion criteria", "Sample size and sampling technique", "Data collection instrument and procedures", "Data quality assurance", "Ethical consideration", "Data entry and analysis", "Operational definitions", "Explanatory factor analysis for subscales of parental satisfaction with NICU-care service", "Reliability of items of EMPATHIC-N tool", "Parental satisfaction level with NICU-care service", "Factors associated with the overall parental satisfaction of NICU care service", "Limitations of the study" ]
[ "[SUBTITLE] Study area and period [SUBSECTION] A cross-sectional study was conducted in DTCSH which is a public hospital established in 1934 and located in the South Gondar Zone of Amara Regional State of Ethiopia. It is 97 km to the southwest of Bahir Dar, the capital city of Amara Regional State. According to the 2007 census, the total population of Debre tabor town was 155,596. It has a latitude and longitude of 11051N3801’E11.8500 N 38.0170E with an elevation of 2,706 m (8878ft) above sea level. The hospital provides neonatal intensive care unit service with five separate NICU rooms. According to the hospital’s 2017 report, 1159 neonates were admitted to the NICU, but according to evidence from a chart review in 2020, 1489 neonates were admitted [17]. This study was conducted on parents of neonates who were admitted to NICU at DTCSH from November 05, 2021, to April 30, 2022.\nA cross-sectional study was conducted in DTCSH which is a public hospital established in 1934 and located in the South Gondar Zone of Amara Regional State of Ethiopia. It is 97 km to the southwest of Bahir Dar, the capital city of Amara Regional State. According to the 2007 census, the total population of Debre tabor town was 155,596. It has a latitude and longitude of 11051N3801’E11.8500 N 38.0170E with an elevation of 2,706 m (8878ft) above sea level. The hospital provides neonatal intensive care unit service with five separate NICU rooms. According to the hospital’s 2017 report, 1159 neonates were admitted to the NICU, but according to evidence from a chart review in 2020, 1489 neonates were admitted [17]. This study was conducted on parents of neonates who were admitted to NICU at DTCSH from November 05, 2021, to April 30, 2022.\n[SUBTITLE] Inclusion and exclusion criteria [SUBSECTION] Parents whose neonate was discharged from NICU or transferred to high dependency neonatal ward, parents who can read and write or understand the Amharic language, and neonatal stay in NICU of less than three months were included in the study. Whereas, parents with neonatal death in NICU and parents with neonatal admissions of less than 24 h were excluded from the study.\nParents whose neonate was discharged from NICU or transferred to high dependency neonatal ward, parents who can read and write or understand the Amharic language, and neonatal stay in NICU of less than three months were included in the study. Whereas, parents with neonatal death in NICU and parents with neonatal admissions of less than 24 h were excluded from the study.\n[SUBTITLE] Sample size and sampling technique [SUBSECTION] The sample size was determined by using single proportion population formula taking 50% (P) of the proportion using a 95% confidence interval and 5% margin of error (d). The sample size was determined using the following formula.\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$n={\\left({Z}_{\\frac{a}{2}}\\right)}^{2}P{(1-P)/d}^{2}$$\\end{document}\nn= [(1.96)2 0.5(1-0.5)2] / (0.05)2.\nN = 385.\nBy considering a 5% non-respondent rate the final sample size was 405.\nThe data were collected from all consecutive parents who met the inclusion criteria until the intended sample size was achieved.\nThe sample size was determined by using single proportion population formula taking 50% (P) of the proportion using a 95% confidence interval and 5% margin of error (d). The sample size was determined using the following formula.\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$n={\\left({Z}_{\\frac{a}{2}}\\right)}^{2}P{(1-P)/d}^{2}$$\\end{document}\nn= [(1.96)2 0.5(1-0.5)2] / (0.05)2.\nN = 385.\nBy considering a 5% non-respondent rate the final sample size was 405.\nThe data were collected from all consecutive parents who met the inclusion criteria until the intended sample size was achieved.\n[SUBTITLE] Data collection instrument and procedures [SUBSECTION] The Amharic version anonymous questionnaire was used for data collection after translating the English version of the EMPATHIC-N tool by three language experts and then back to English by the other three experts to ensure that the translation was correct. The tool’s content validity was also examined and guaranteed by members of the Anesthesia department’s research committee.\nThe tool has been widely used to assess parental satisfaction with NICU care services, and it has strong reliability and validity, with reliability (Cronbach’s) values of the domains ranging from 0.82 to 0.95 [18–20]. There are five domains in the EMPATHIC-N tool: Information (12 questions with a six-point Likert scale); Care & Treatment (17 questions with a ten-point Likert scale); Parental Participation (8 questions with a six-point Likert scale); Organization (8 questions with a six-point Likert scale); and Professional Attitude (12 questions with a six-point Likert scale) [18, 21, 22].\nThe data were collected by three anesthetists on the day of neonatal discharge using an adopted Dutch instrument of Empowerment of Parents in The Intensive Care-Neonatology (EMPATHIC-N).\nThe Amharic version anonymous questionnaire was used for data collection after translating the English version of the EMPATHIC-N tool by three language experts and then back to English by the other three experts to ensure that the translation was correct. The tool’s content validity was also examined and guaranteed by members of the Anesthesia department’s research committee.\nThe tool has been widely used to assess parental satisfaction with NICU care services, and it has strong reliability and validity, with reliability (Cronbach’s) values of the domains ranging from 0.82 to 0.95 [18–20]. There are five domains in the EMPATHIC-N tool: Information (12 questions with a six-point Likert scale); Care & Treatment (17 questions with a ten-point Likert scale); Parental Participation (8 questions with a six-point Likert scale); Organization (8 questions with a six-point Likert scale); and Professional Attitude (12 questions with a six-point Likert scale) [18, 21, 22].\nThe data were collected by three anesthetists on the day of neonatal discharge using an adopted Dutch instrument of Empowerment of Parents in The Intensive Care-Neonatology (EMPATHIC-N).\n[SUBTITLE] Data quality assurance [SUBSECTION] To ensure the quality of data, pre-testing of the data collection tool (the questionnaire) was done on 5% of study parents from Felege Hiwot Comprehensive Specialized Hospital who were not included in the main study. The training was given to data collectors; data were collected and properly filled in the prepared format. Supervision was made throughout the data collection period to make sure the accuracy, clarity, and consistency of the collected data.\nTo ensure the quality of data, pre-testing of the data collection tool (the questionnaire) was done on 5% of study parents from Felege Hiwot Comprehensive Specialized Hospital who were not included in the main study. The training was given to data collectors; data were collected and properly filled in the prepared format. Supervision was made throughout the data collection period to make sure the accuracy, clarity, and consistency of the collected data.\n[SUBTITLE] Ethical consideration [SUBSECTION] Debre Tabor University provided ethical clearance, and each parent was given written informed consent after being briefed about the purpose study.\nDebre Tabor University provided ethical clearance, and each parent was given written informed consent after being briefed about the purpose study.\n[SUBTITLE] Data entry and analysis [SUBSECTION] Data were cleaned, coded, and entered into Epidata version 4.2 and exported to SPSS version 23 for statistical analysis. The adopted EMPATHIC-N instrument was validated using analysis (validity, reliability, standard factor loadings, and factor analysis). Cronbach’s alpha was used to determine the reliability and validity of the tool. Explanatory factor analysis was done to test how well the measured variables represent the number of constructs and to identify relationships between the measured items. The inter-item correlation was used to assess the relationship between items on the same scale, whereas item-discriminant validity was used to assess the relationship between scales. After categorizing the overall mean parental satisfaction score, independent variables were analyzed using binary logistic regression with parental satisfaction with NICU care service. Variables from the bivariable logistic regression with a p-value of 0.2 were fitted to a multivariable logistic regression, and certain variables were included in the model with their clinical importance. Both crude odds ratio (COR) in bivariable logistic regression and adjusted odds ratio (AOR) in multivariable logistic regression with the corresponding 95% Confidence interval were calculated to show the strength of association. In multivariable logistic regression analysis, variables with a p-value of < 0.05 were considered statistically significant. The Mann–Whitney test was used to determine the influencing factors of the parental satisfaction domains, while the Hosmer–Lemeshow goodness of fit test was performed to ensure that the analysis model was appropriate.\nData were cleaned, coded, and entered into Epidata version 4.2 and exported to SPSS version 23 for statistical analysis. The adopted EMPATHIC-N instrument was validated using analysis (validity, reliability, standard factor loadings, and factor analysis). Cronbach’s alpha was used to determine the reliability and validity of the tool. Explanatory factor analysis was done to test how well the measured variables represent the number of constructs and to identify relationships between the measured items. The inter-item correlation was used to assess the relationship between items on the same scale, whereas item-discriminant validity was used to assess the relationship between scales. After categorizing the overall mean parental satisfaction score, independent variables were analyzed using binary logistic regression with parental satisfaction with NICU care service. Variables from the bivariable logistic regression with a p-value of 0.2 were fitted to a multivariable logistic regression, and certain variables were included in the model with their clinical importance. Both crude odds ratio (COR) in bivariable logistic regression and adjusted odds ratio (AOR) in multivariable logistic regression with the corresponding 95% Confidence interval were calculated to show the strength of association. In multivariable logistic regression analysis, variables with a p-value of < 0.05 were considered statistically significant. The Mann–Whitney test was used to determine the influencing factors of the parental satisfaction domains, while the Hosmer–Lemeshow goodness of fit test was performed to ensure that the analysis model was appropriate.\n[SUBTITLE] Operational definitions [SUBSECTION] Satisfied: parents who scored greater than or equal to the overall mean EMPATHIC-N values were considered satisfied.\nDissatisfied: parents who scored less than the overall mean EMPATHIC-N values were considered dissatisfied.\nHigh birth weight: neonates with a birth weight of more than 400 g [23].\nNormal birth weight: neonates with a birth weight range from 250 to 400 g [24].\nLow birth weight: neonates with a birth weight of lower than 250 g [25].\nSatisfied: parents who scored greater than or equal to the overall mean EMPATHIC-N values were considered satisfied.\nDissatisfied: parents who scored less than the overall mean EMPATHIC-N values were considered dissatisfied.\nHigh birth weight: neonates with a birth weight of more than 400 g [23].\nNormal birth weight: neonates with a birth weight range from 250 to 400 g [24].\nLow birth weight: neonates with a birth weight of lower than 250 g [25].", "A cross-sectional study was conducted in DTCSH which is a public hospital established in 1934 and located in the South Gondar Zone of Amara Regional State of Ethiopia. It is 97 km to the southwest of Bahir Dar, the capital city of Amara Regional State. According to the 2007 census, the total population of Debre tabor town was 155,596. It has a latitude and longitude of 11051N3801’E11.8500 N 38.0170E with an elevation of 2,706 m (8878ft) above sea level. The hospital provides neonatal intensive care unit service with five separate NICU rooms. According to the hospital’s 2017 report, 1159 neonates were admitted to the NICU, but according to evidence from a chart review in 2020, 1489 neonates were admitted [17]. This study was conducted on parents of neonates who were admitted to NICU at DTCSH from November 05, 2021, to April 30, 2022.", "Parents whose neonate was discharged from NICU or transferred to high dependency neonatal ward, parents who can read and write or understand the Amharic language, and neonatal stay in NICU of less than three months were included in the study. Whereas, parents with neonatal death in NICU and parents with neonatal admissions of less than 24 h were excluded from the study.", "The sample size was determined by using single proportion population formula taking 50% (P) of the proportion using a 95% confidence interval and 5% margin of error (d). The sample size was determined using the following formula.\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$n={\\left({Z}_{\\frac{a}{2}}\\right)}^{2}P{(1-P)/d}^{2}$$\\end{document}\nn= [(1.96)2 0.5(1-0.5)2] / (0.05)2.\nN = 385.\nBy considering a 5% non-respondent rate the final sample size was 405.\nThe data were collected from all consecutive parents who met the inclusion criteria until the intended sample size was achieved.", "The Amharic version anonymous questionnaire was used for data collection after translating the English version of the EMPATHIC-N tool by three language experts and then back to English by the other three experts to ensure that the translation was correct. The tool’s content validity was also examined and guaranteed by members of the Anesthesia department’s research committee.\nThe tool has been widely used to assess parental satisfaction with NICU care services, and it has strong reliability and validity, with reliability (Cronbach’s) values of the domains ranging from 0.82 to 0.95 [18–20]. There are five domains in the EMPATHIC-N tool: Information (12 questions with a six-point Likert scale); Care & Treatment (17 questions with a ten-point Likert scale); Parental Participation (8 questions with a six-point Likert scale); Organization (8 questions with a six-point Likert scale); and Professional Attitude (12 questions with a six-point Likert scale) [18, 21, 22].\nThe data were collected by three anesthetists on the day of neonatal discharge using an adopted Dutch instrument of Empowerment of Parents in The Intensive Care-Neonatology (EMPATHIC-N).", "To ensure the quality of data, pre-testing of the data collection tool (the questionnaire) was done on 5% of study parents from Felege Hiwot Comprehensive Specialized Hospital who were not included in the main study. The training was given to data collectors; data were collected and properly filled in the prepared format. Supervision was made throughout the data collection period to make sure the accuracy, clarity, and consistency of the collected data.", "Debre Tabor University provided ethical clearance, and each parent was given written informed consent after being briefed about the purpose study.", "Data were cleaned, coded, and entered into Epidata version 4.2 and exported to SPSS version 23 for statistical analysis. The adopted EMPATHIC-N instrument was validated using analysis (validity, reliability, standard factor loadings, and factor analysis). Cronbach’s alpha was used to determine the reliability and validity of the tool. Explanatory factor analysis was done to test how well the measured variables represent the number of constructs and to identify relationships between the measured items. The inter-item correlation was used to assess the relationship between items on the same scale, whereas item-discriminant validity was used to assess the relationship between scales. After categorizing the overall mean parental satisfaction score, independent variables were analyzed using binary logistic regression with parental satisfaction with NICU care service. Variables from the bivariable logistic regression with a p-value of 0.2 were fitted to a multivariable logistic regression, and certain variables were included in the model with their clinical importance. Both crude odds ratio (COR) in bivariable logistic regression and adjusted odds ratio (AOR) in multivariable logistic regression with the corresponding 95% Confidence interval were calculated to show the strength of association. In multivariable logistic regression analysis, variables with a p-value of < 0.05 were considered statistically significant. The Mann–Whitney test was used to determine the influencing factors of the parental satisfaction domains, while the Hosmer–Lemeshow goodness of fit test was performed to ensure that the analysis model was appropriate.", "Satisfied: parents who scored greater than or equal to the overall mean EMPATHIC-N values were considered satisfied.\nDissatisfied: parents who scored less than the overall mean EMPATHIC-N values were considered dissatisfied.\nHigh birth weight: neonates with a birth weight of more than 400 g [23].\nNormal birth weight: neonates with a birth weight range from 250 to 400 g [24].\nLow birth weight: neonates with a birth weight of lower than 250 g [25].", "Before assessing parental satisfaction levels, confirmatory factor analysis was used to ensure that the measured variables accurately represented the constructs. The factor correlation matrix was used in the analysis. KMO and Bartlett’s tests of sphericity were used to check the correlation of measurement variables, with a KMO value of 0.875 and Bartlett’s tests of sphericity (p = 0.00) respectively. The correlation matrix was checked for commonality extraction, and all item values were larger than 0.3. The parallel analysis determined the number of item components (factors), and two new components for parental participation, three new components for information and organization, and four new components for Care & Treatment and professional attitude of eigenvalues met the criteria, and the Varimax component correlation matrix was an appropriate model (Table 2).\n\nTable 2Factor loadings and identified components’ of EMPATHIC-N toolItemFactors\n1\n\n2\n\n3\n\n4\n\nInformation\n What level of doctor’s information do you have regarding the child’s expected health outcomes?0.752 How satisfied are you with the physicians’ and nurses’ information similarity?0.704 How are you satisfied with doctors’ and nurses’ honesty in providing information?0.59 How satisfied are you with daily discussions with doctors and nurses about your child’s care and treatment?0.58 How understandable was the information provided by the doctors and nurses?0.763 How satisfied were you with the correct information when the child’s physical condition deteriorated?0.4280.657 How satisfied were you with the clear answers to your questions?0.653 How clear is the doctor’s information about the consequences of the child’s treatment?0.533 To what extent do you receive clear information about the examinations and tests?0.713 To what extent the information brochure received was complete and clear?0.641 How much clear information is given regarding a child’s illness?0.626 Level of received understandable information about the effects of the drugs?0.606\nCare & Treatment\n Level of child’s comfort taken into account by the doctors and nurses?0.826 The extent of satisfaction During acute situations on availability of nurses to support?0.822 Level of team alertness to the prevention and treatment of pain of neonate?0.797 Level of care taken to nurses while in the incubator/bed?0.6340.501 Level of correct medication always administered on time?0.6310.402 level of emotional support that has been provided?0.84 The doctors and nurses responded well to our own needs0.809 Transferals of care from the neonatal intensive care unit staff to colleagues in the high-care unit or pediatric ward had gone well?0.782 Every day we knew who of the doctors and nurses was responsible for our child.0.695 How closely did doctors and nurses collaborate during work?0.5780.466 Level of a common goal: to provide the finest care and treatment for our child and ourselves.0.774 Level of physicians and nurses paid close attention to our child’s development?0.756 The team as a whole was concerned for our child and you.0.698 Our child’s requirements were met promptly0.455 The extent of doctors’ and nurses’ professional knowledge of what they are doing?0.847 How satisfied are you with the doctors’ and nurses’ understanding of the child’s medical history at the time of admission?0.765 How satisfied are you with the rapid actions taken by doctors and nurses when a child’s condition deteriorated?0.723\nParental Participation\n How involved are you in making decisions about our child’s care and treatment?0.803 The nurses had trained us on the specific aspects of newborn care.0.796 We were encouraged to stay close to our children.0.714 Before discharge, the care for our child was once more discussed with us.0.617 Even during intensive procedures, we could always stay close to our child.0.841 The nurses stimulated us to help in the care of our child0.837 The nurses helped us in the bonding with our child0.5320.575 We had confidence in the team0.561\nOrganization\n The Neonatology unit made us feel safe0.885 There was a warm atmosphere in the Neonatology unit without hostility0.805 The Neonatology unit was clean0.7330.404 The unit could easily be reached by telephone0.809 Our child’s incubator or bed was clean0.8 The team worked efficiently0.661 There was enough space around our child’s incubator/bed0.9 Noise in the unit was muffled as good as possible0.788\nProfessional Attitude\n Our child’s health always came first for the doctors and nurses0.799 The team worked hygienically0.747 Our cultural background was taken into account0.728 The doctors and nurses always took time to listen to us0.876 We felt welcome by the team0.804 Despite the workload, sufficient attention was paid to our child and us by the team0.5840.508 Nurses and doctors always introduced themselves by name and function0.794 We received sympathy from the doctors and nurses0.786 At our bedside, the discussion between the doctors and nurses was only about our child.0.4570.582 The team respected the privacy of our children and us.0.874 There was a pleasant atmosphere among the staff0.743 The team showed respect for our child and us0.5190.609\n\nFactor loadings and identified components’ of EMPATHIC-N tool", "The total EMPATHIC-N values and the five domains showed a good level of internal consistency. The five domains’ Cronbach’s values range from 0.639 to 0.791, whereas the total EMPATHIC-N Cronbach’s value was 0.904. The inter-item correlation (IIC) of the five domains had significant internal consistency (Table 3).\n\nTable 3Reliability of items of parental satisfaction with NICU-care serviceDimensionNumber of ItemsΧηρονβαχη αMean dimension score (SD)Maximum possible dimension scoreInter-item correlation (IIC)Item-discriminant validity (IDV)Information120.63939.55(8.42)720.22–0.48*0.20–0.57Care & Treatment170.79181.70(20.49)1700.14–0.64*0.25–0.38Parental Participation80.75524.86(7.79)480.01–0.71*0.37–0.54Organization80.72925.22(7.39)480.11–0.64*0.34–0.69Professional Attitude120.69438.106 (9.07)720.13–0.56*0.22–0.45EMPATHIC-N tool570.904209.44(42.82)410cc*Significant value (p < 0.001), c = not computable\n\nReliability of items of parental satisfaction with NICU-care service\n*Significant value (p < 0.001), c = not computable", "Overall mean satisfaction level of parents in NICU-care service was 47.8% [95% CI= (43.1–52.5)]. The domains of parental satisfaction scores were compared with each other and, the lowest parental satisfaction score was for the domain of care and treatment 36.9% while the organizational domain showed the highest parental satisfaction level 59.0%. The domains of information, parental participation, and professional attitude showed comparable parental satisfaction scores (Fig. 1).\n\nFig. 1Parents’ overall and dimensional satisfaction with NICU-care services\n\nParents’ overall and dimensional satisfaction with NICU-care services", "In this study, the bivariable logistic regression showed that parental gender, residency, parental hospital stay, neonatal hospital stay, birth weight, and gestational age were factors with a p-value of less than 0.2 and were fitted with a multivariable logistic regression model. However, neonatal hospital stay was not associated with multivariable logistic regression with a p-value greater than 0.05. The multivariable logistic analyses showed that mothers were 2.16 (AOR = 2.16; 95%CI: 1.28–3.63) times more satisfied than fathers. Also, parents who are from the rural area were 2.94 (AOR = 2.94; 95%CI: 1.42–6.06) more satisfied than urban. Parents who say less than 15 days were 2.18 (AOR = 2.18; 95%CI: 1.13–4.20) times more satisfied than parents who stay 15 or more days in the hospital. Also, parents of a neonate with a normal birth weight of 2.14 (AOR = 2.14; 95%CI: 1.16–3.94) and full-term neonate 2.53(AOR = 2.53; 95%CI: 1.29–4.97) times more satisfied than their counterparts (Table 4).\n\nTable 4Factors associated with satisfaction of parents in NICU-care service (n = 385)VariablesSatisfaction level on NICU- serviceCrude odds ratioAdjusted odds ratiop-valueSatisfiedNot Satisfied(95% CI)(95% CI)\nGender\n Mother117(52.2%)107(47.8%)1.53(1.02,2.31)2.16(1.28,3.63)0.004* Father67(41.6%)94(58.4%)1\nResidency\n Urban68(36.4%)119(63.6%)1 Rural116(58.6%)82(41.4%)2.48(1.64,3.73)2.94(1.42,6.06)0.004*\nParental hospital stay\n ≤ 15 days140(57.4%)104(42.6%)2.97(1.92,4.59)2.18(1.13,4.20)0.019* > 15days44(31.2%)97(68.8%)1\nBirth weight\n High birth weight34(41.0%)49(59.0%)1 Normal birth weight96(59.3%)66(40.7%)2.09(1.22,3.59)2.14(1.16,3.94)0.015* Low birth weight54(38.6%)86(61.4%)0.91(0.52,1.58)0.76(0.41,1.41)0.38\nGestational age\n Premature18(30.0%)42(70.0%)1 Full term166(51.1%)159(48.9%)2.44(1.35,4.41)2.53(1.29,4.97)0.007**= p-value < 0.05 1 = reference BWt-birth weightNote: p-values were extracted from the multivariate logistic regression model\n\nFactors associated with satisfaction of parents in NICU-care service (n = 385)\n*= p-value < 0.05 1 = reference BWt-birth weight\nNote: p-values were extracted from the multivariate logistic regression model", "The study’s limitations include being a single-center nature and the lack of analysis of various alternatives to logistic regression that might be more applicable for this investigation." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Introduction", "Methods and materials", "Study area and period", "Inclusion and exclusion criteria", "Sample size and sampling technique", "Data collection instrument and procedures", "Data quality assurance", "Ethical consideration", "Data entry and analysis", "Operational definitions", "Results", "Explanatory factor analysis for subscales of parental satisfaction with NICU-care service", "Reliability of items of EMPATHIC-N tool", "Parental satisfaction level with NICU-care service", "Factors associated with the overall parental satisfaction of NICU care service", "Discussion", "Limitations of the study", "Conclusion" ]
[ "Neonatal intensive care units (NICUs) are areas that require careful risk management with a wide range of neonatal care services [1–3]. It necessitates high-cost and efficient critical care delivery with a multidisciplinary team approach that focuses on preventive strategies for improved outcomes [4–6]. Parental tension and emotions are high when their child is admitted to a neonatal intensive care unit (NICU) due to serious illnesses [7, 8].\nSatisfaction is a belief and attitude about a specific service provision of an institution. Parental and patient satisfaction has become a well-established outcome indicator and tool for assessing a healthcare system’s quality, as well as input for developing strategies and providing accessible, sustainable, economical, as well as acceptable patient care [7, 9, 10]. Parental satisfaction reflects the balance between their expectations of ideal care and their perception of real and available care [3, 11, 12]. It is also one of the objectives and missions of every health care center that gives NICU care service [10, 11].\nParent and patient satisfaction has become an important aspect of hospital management initiatives for quality assurance and accreditation. Also, parental involvement has an important role in the delivery of high-quality care, ranging from assisting with activities of daily living to being directly involved in important health care decisions [13, 14]. However, parental participation may not always be possible, but, effective communication will reduce the effects of crises and improve parental satisfaction [11, 15].\nEthiopia has been working to enhance its healthcare delivery systems by focusing on quality healthcare service giving special attention to mothers and children. The Ethiopian Health Service Alliance for Quality has agreed that the initial priority area would be self-motivated and transparent partnerships that stimulate innovation in health care quality management and learning across all hospitals [16].\nGiven the scarcity of studies on parental satisfaction with NICU care in Ethiopia, as well as clinical observations of parents complaining about NICU care, it is critical to determine the level of parental satisfaction. So, this study aimed to identify the level of parental satisfaction and associated factors with NICU care service in Debre Tabor Comprehensive Specialized Hospital (DTCSH). Also, this study helps the administrators to understand the deficiencies of the hospital’s NICU services, and then propose corresponding improvement strategies.", "[SUBTITLE] Study area and period [SUBSECTION] A cross-sectional study was conducted in DTCSH which is a public hospital established in 1934 and located in the South Gondar Zone of Amara Regional State of Ethiopia. It is 97 km to the southwest of Bahir Dar, the capital city of Amara Regional State. According to the 2007 census, the total population of Debre tabor town was 155,596. It has a latitude and longitude of 11051N3801’E11.8500 N 38.0170E with an elevation of 2,706 m (8878ft) above sea level. The hospital provides neonatal intensive care unit service with five separate NICU rooms. According to the hospital’s 2017 report, 1159 neonates were admitted to the NICU, but according to evidence from a chart review in 2020, 1489 neonates were admitted [17]. This study was conducted on parents of neonates who were admitted to NICU at DTCSH from November 05, 2021, to April 30, 2022.\nA cross-sectional study was conducted in DTCSH which is a public hospital established in 1934 and located in the South Gondar Zone of Amara Regional State of Ethiopia. It is 97 km to the southwest of Bahir Dar, the capital city of Amara Regional State. According to the 2007 census, the total population of Debre tabor town was 155,596. It has a latitude and longitude of 11051N3801’E11.8500 N 38.0170E with an elevation of 2,706 m (8878ft) above sea level. The hospital provides neonatal intensive care unit service with five separate NICU rooms. According to the hospital’s 2017 report, 1159 neonates were admitted to the NICU, but according to evidence from a chart review in 2020, 1489 neonates were admitted [17]. This study was conducted on parents of neonates who were admitted to NICU at DTCSH from November 05, 2021, to April 30, 2022.\n[SUBTITLE] Inclusion and exclusion criteria [SUBSECTION] Parents whose neonate was discharged from NICU or transferred to high dependency neonatal ward, parents who can read and write or understand the Amharic language, and neonatal stay in NICU of less than three months were included in the study. Whereas, parents with neonatal death in NICU and parents with neonatal admissions of less than 24 h were excluded from the study.\nParents whose neonate was discharged from NICU or transferred to high dependency neonatal ward, parents who can read and write or understand the Amharic language, and neonatal stay in NICU of less than three months were included in the study. Whereas, parents with neonatal death in NICU and parents with neonatal admissions of less than 24 h were excluded from the study.\n[SUBTITLE] Sample size and sampling technique [SUBSECTION] The sample size was determined by using single proportion population formula taking 50% (P) of the proportion using a 95% confidence interval and 5% margin of error (d). The sample size was determined using the following formula.\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$n={\\left({Z}_{\\frac{a}{2}}\\right)}^{2}P{(1-P)/d}^{2}$$\\end{document}\nn= [(1.96)2 0.5(1-0.5)2] / (0.05)2.\nN = 385.\nBy considering a 5% non-respondent rate the final sample size was 405.\nThe data were collected from all consecutive parents who met the inclusion criteria until the intended sample size was achieved.\nThe sample size was determined by using single proportion population formula taking 50% (P) of the proportion using a 95% confidence interval and 5% margin of error (d). The sample size was determined using the following formula.\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$n={\\left({Z}_{\\frac{a}{2}}\\right)}^{2}P{(1-P)/d}^{2}$$\\end{document}\nn= [(1.96)2 0.5(1-0.5)2] / (0.05)2.\nN = 385.\nBy considering a 5% non-respondent rate the final sample size was 405.\nThe data were collected from all consecutive parents who met the inclusion criteria until the intended sample size was achieved.\n[SUBTITLE] Data collection instrument and procedures [SUBSECTION] The Amharic version anonymous questionnaire was used for data collection after translating the English version of the EMPATHIC-N tool by three language experts and then back to English by the other three experts to ensure that the translation was correct. The tool’s content validity was also examined and guaranteed by members of the Anesthesia department’s research committee.\nThe tool has been widely used to assess parental satisfaction with NICU care services, and it has strong reliability and validity, with reliability (Cronbach’s) values of the domains ranging from 0.82 to 0.95 [18–20]. There are five domains in the EMPATHIC-N tool: Information (12 questions with a six-point Likert scale); Care & Treatment (17 questions with a ten-point Likert scale); Parental Participation (8 questions with a six-point Likert scale); Organization (8 questions with a six-point Likert scale); and Professional Attitude (12 questions with a six-point Likert scale) [18, 21, 22].\nThe data were collected by three anesthetists on the day of neonatal discharge using an adopted Dutch instrument of Empowerment of Parents in The Intensive Care-Neonatology (EMPATHIC-N).\nThe Amharic version anonymous questionnaire was used for data collection after translating the English version of the EMPATHIC-N tool by three language experts and then back to English by the other three experts to ensure that the translation was correct. The tool’s content validity was also examined and guaranteed by members of the Anesthesia department’s research committee.\nThe tool has been widely used to assess parental satisfaction with NICU care services, and it has strong reliability and validity, with reliability (Cronbach’s) values of the domains ranging from 0.82 to 0.95 [18–20]. There are five domains in the EMPATHIC-N tool: Information (12 questions with a six-point Likert scale); Care & Treatment (17 questions with a ten-point Likert scale); Parental Participation (8 questions with a six-point Likert scale); Organization (8 questions with a six-point Likert scale); and Professional Attitude (12 questions with a six-point Likert scale) [18, 21, 22].\nThe data were collected by three anesthetists on the day of neonatal discharge using an adopted Dutch instrument of Empowerment of Parents in The Intensive Care-Neonatology (EMPATHIC-N).\n[SUBTITLE] Data quality assurance [SUBSECTION] To ensure the quality of data, pre-testing of the data collection tool (the questionnaire) was done on 5% of study parents from Felege Hiwot Comprehensive Specialized Hospital who were not included in the main study. The training was given to data collectors; data were collected and properly filled in the prepared format. Supervision was made throughout the data collection period to make sure the accuracy, clarity, and consistency of the collected data.\nTo ensure the quality of data, pre-testing of the data collection tool (the questionnaire) was done on 5% of study parents from Felege Hiwot Comprehensive Specialized Hospital who were not included in the main study. The training was given to data collectors; data were collected and properly filled in the prepared format. Supervision was made throughout the data collection period to make sure the accuracy, clarity, and consistency of the collected data.\n[SUBTITLE] Ethical consideration [SUBSECTION] Debre Tabor University provided ethical clearance, and each parent was given written informed consent after being briefed about the purpose study.\nDebre Tabor University provided ethical clearance, and each parent was given written informed consent after being briefed about the purpose study.\n[SUBTITLE] Data entry and analysis [SUBSECTION] Data were cleaned, coded, and entered into Epidata version 4.2 and exported to SPSS version 23 for statistical analysis. The adopted EMPATHIC-N instrument was validated using analysis (validity, reliability, standard factor loadings, and factor analysis). Cronbach’s alpha was used to determine the reliability and validity of the tool. Explanatory factor analysis was done to test how well the measured variables represent the number of constructs and to identify relationships between the measured items. The inter-item correlation was used to assess the relationship between items on the same scale, whereas item-discriminant validity was used to assess the relationship between scales. After categorizing the overall mean parental satisfaction score, independent variables were analyzed using binary logistic regression with parental satisfaction with NICU care service. Variables from the bivariable logistic regression with a p-value of 0.2 were fitted to a multivariable logistic regression, and certain variables were included in the model with their clinical importance. Both crude odds ratio (COR) in bivariable logistic regression and adjusted odds ratio (AOR) in multivariable logistic regression with the corresponding 95% Confidence interval were calculated to show the strength of association. In multivariable logistic regression analysis, variables with a p-value of < 0.05 were considered statistically significant. The Mann–Whitney test was used to determine the influencing factors of the parental satisfaction domains, while the Hosmer–Lemeshow goodness of fit test was performed to ensure that the analysis model was appropriate.\nData were cleaned, coded, and entered into Epidata version 4.2 and exported to SPSS version 23 for statistical analysis. The adopted EMPATHIC-N instrument was validated using analysis (validity, reliability, standard factor loadings, and factor analysis). Cronbach’s alpha was used to determine the reliability and validity of the tool. Explanatory factor analysis was done to test how well the measured variables represent the number of constructs and to identify relationships between the measured items. The inter-item correlation was used to assess the relationship between items on the same scale, whereas item-discriminant validity was used to assess the relationship between scales. After categorizing the overall mean parental satisfaction score, independent variables were analyzed using binary logistic regression with parental satisfaction with NICU care service. Variables from the bivariable logistic regression with a p-value of 0.2 were fitted to a multivariable logistic regression, and certain variables were included in the model with their clinical importance. Both crude odds ratio (COR) in bivariable logistic regression and adjusted odds ratio (AOR) in multivariable logistic regression with the corresponding 95% Confidence interval were calculated to show the strength of association. In multivariable logistic regression analysis, variables with a p-value of < 0.05 were considered statistically significant. The Mann–Whitney test was used to determine the influencing factors of the parental satisfaction domains, while the Hosmer–Lemeshow goodness of fit test was performed to ensure that the analysis model was appropriate.\n[SUBTITLE] Operational definitions [SUBSECTION] Satisfied: parents who scored greater than or equal to the overall mean EMPATHIC-N values were considered satisfied.\nDissatisfied: parents who scored less than the overall mean EMPATHIC-N values were considered dissatisfied.\nHigh birth weight: neonates with a birth weight of more than 400 g [23].\nNormal birth weight: neonates with a birth weight range from 250 to 400 g [24].\nLow birth weight: neonates with a birth weight of lower than 250 g [25].\nSatisfied: parents who scored greater than or equal to the overall mean EMPATHIC-N values were considered satisfied.\nDissatisfied: parents who scored less than the overall mean EMPATHIC-N values were considered dissatisfied.\nHigh birth weight: neonates with a birth weight of more than 400 g [23].\nNormal birth weight: neonates with a birth weight range from 250 to 400 g [24].\nLow birth weight: neonates with a birth weight of lower than 250 g [25].", "A cross-sectional study was conducted in DTCSH which is a public hospital established in 1934 and located in the South Gondar Zone of Amara Regional State of Ethiopia. It is 97 km to the southwest of Bahir Dar, the capital city of Amara Regional State. According to the 2007 census, the total population of Debre tabor town was 155,596. It has a latitude and longitude of 11051N3801’E11.8500 N 38.0170E with an elevation of 2,706 m (8878ft) above sea level. The hospital provides neonatal intensive care unit service with five separate NICU rooms. According to the hospital’s 2017 report, 1159 neonates were admitted to the NICU, but according to evidence from a chart review in 2020, 1489 neonates were admitted [17]. This study was conducted on parents of neonates who were admitted to NICU at DTCSH from November 05, 2021, to April 30, 2022.", "Parents whose neonate was discharged from NICU or transferred to high dependency neonatal ward, parents who can read and write or understand the Amharic language, and neonatal stay in NICU of less than three months were included in the study. Whereas, parents with neonatal death in NICU and parents with neonatal admissions of less than 24 h were excluded from the study.", "The sample size was determined by using single proportion population formula taking 50% (P) of the proportion using a 95% confidence interval and 5% margin of error (d). The sample size was determined using the following formula.\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$n={\\left({Z}_{\\frac{a}{2}}\\right)}^{2}P{(1-P)/d}^{2}$$\\end{document}\nn= [(1.96)2 0.5(1-0.5)2] / (0.05)2.\nN = 385.\nBy considering a 5% non-respondent rate the final sample size was 405.\nThe data were collected from all consecutive parents who met the inclusion criteria until the intended sample size was achieved.", "The Amharic version anonymous questionnaire was used for data collection after translating the English version of the EMPATHIC-N tool by three language experts and then back to English by the other three experts to ensure that the translation was correct. The tool’s content validity was also examined and guaranteed by members of the Anesthesia department’s research committee.\nThe tool has been widely used to assess parental satisfaction with NICU care services, and it has strong reliability and validity, with reliability (Cronbach’s) values of the domains ranging from 0.82 to 0.95 [18–20]. There are five domains in the EMPATHIC-N tool: Information (12 questions with a six-point Likert scale); Care & Treatment (17 questions with a ten-point Likert scale); Parental Participation (8 questions with a six-point Likert scale); Organization (8 questions with a six-point Likert scale); and Professional Attitude (12 questions with a six-point Likert scale) [18, 21, 22].\nThe data were collected by three anesthetists on the day of neonatal discharge using an adopted Dutch instrument of Empowerment of Parents in The Intensive Care-Neonatology (EMPATHIC-N).", "To ensure the quality of data, pre-testing of the data collection tool (the questionnaire) was done on 5% of study parents from Felege Hiwot Comprehensive Specialized Hospital who were not included in the main study. The training was given to data collectors; data were collected and properly filled in the prepared format. Supervision was made throughout the data collection period to make sure the accuracy, clarity, and consistency of the collected data.", "Debre Tabor University provided ethical clearance, and each parent was given written informed consent after being briefed about the purpose study.", "Data were cleaned, coded, and entered into Epidata version 4.2 and exported to SPSS version 23 for statistical analysis. The adopted EMPATHIC-N instrument was validated using analysis (validity, reliability, standard factor loadings, and factor analysis). Cronbach’s alpha was used to determine the reliability and validity of the tool. Explanatory factor analysis was done to test how well the measured variables represent the number of constructs and to identify relationships between the measured items. The inter-item correlation was used to assess the relationship between items on the same scale, whereas item-discriminant validity was used to assess the relationship between scales. After categorizing the overall mean parental satisfaction score, independent variables were analyzed using binary logistic regression with parental satisfaction with NICU care service. Variables from the bivariable logistic regression with a p-value of 0.2 were fitted to a multivariable logistic regression, and certain variables were included in the model with their clinical importance. Both crude odds ratio (COR) in bivariable logistic regression and adjusted odds ratio (AOR) in multivariable logistic regression with the corresponding 95% Confidence interval were calculated to show the strength of association. In multivariable logistic regression analysis, variables with a p-value of < 0.05 were considered statistically significant. The Mann–Whitney test was used to determine the influencing factors of the parental satisfaction domains, while the Hosmer–Lemeshow goodness of fit test was performed to ensure that the analysis model was appropriate.", "Satisfied: parents who scored greater than or equal to the overall mean EMPATHIC-N values were considered satisfied.\nDissatisfied: parents who scored less than the overall mean EMPATHIC-N values were considered dissatisfied.\nHigh birth weight: neonates with a birth weight of more than 400 g [23].\nNormal birth weight: neonates with a birth weight range from 250 to 400 g [24].\nLow birth weight: neonates with a birth weight of lower than 250 g [25].", "This study was conducted on a total of 385 parents, with a 95.06% response rate. The majority of the parents were mothers 224 (58.2%), whereas 322 (83.6%) were married. Full-term newborns 325(84.4%) and those with respiratory issues 115 (29.9%) had the highest number of NICU admissions (Table 1).\n\nTable 1Socio-demographic characteristics of study participants (n = 385)VariablesFrequency (n)Percentage (%)\nParental gender\n Mother22458.2 Father16141.8\nAge\n 18–2413234.3 25–3916643.1 40 and above8722.6\nMarital status\n Married32283.6 Not married6316.4\nResidency\n Urban18748.6 Rural19851.4\nEducational level\n No formal education11930.9 Elementary school9223.9 Secondary school8321.6 Collage and above9123.6\nProfession\n Housewife7018.2 Farmer10627.5 Student5915.3 Gov’t employ7218.7 Private employ7820.3\nParental hospital stay\n ≤ 15 days24463.4 > 15 days14136.6\nNeonatal gender\n Male18848.8 Female19751.2\nBirth weight\n High birth weight8321.5 Normal birth weight16242.1 Low birth weight14036.4\nGestational age\n Premature6015.6 Full term32584.4\nNeonatal hospital stay\n ≤ 15 days23561 > 15 days15039\nAdmission diagnosis\n Infection6516.9 Respiratory problems11529.9 Prematurity5915.3 Gastrointestinal problems389.9 Jaundice4712.2 Neurological problems215.5 Cardiological problems266.8 Others143.5\n\nSocio-demographic characteristics of study participants (n = 385)\n[SUBTITLE] Explanatory factor analysis for subscales of parental satisfaction with NICU-care service [SUBSECTION] Before assessing parental satisfaction levels, confirmatory factor analysis was used to ensure that the measured variables accurately represented the constructs. The factor correlation matrix was used in the analysis. KMO and Bartlett’s tests of sphericity were used to check the correlation of measurement variables, with a KMO value of 0.875 and Bartlett’s tests of sphericity (p = 0.00) respectively. The correlation matrix was checked for commonality extraction, and all item values were larger than 0.3. The parallel analysis determined the number of item components (factors), and two new components for parental participation, three new components for information and organization, and four new components for Care & Treatment and professional attitude of eigenvalues met the criteria, and the Varimax component correlation matrix was an appropriate model (Table 2).\n\nTable 2Factor loadings and identified components’ of EMPATHIC-N toolItemFactors\n1\n\n2\n\n3\n\n4\n\nInformation\n What level of doctor’s information do you have regarding the child’s expected health outcomes?0.752 How satisfied are you with the physicians’ and nurses’ information similarity?0.704 How are you satisfied with doctors’ and nurses’ honesty in providing information?0.59 How satisfied are you with daily discussions with doctors and nurses about your child’s care and treatment?0.58 How understandable was the information provided by the doctors and nurses?0.763 How satisfied were you with the correct information when the child’s physical condition deteriorated?0.4280.657 How satisfied were you with the clear answers to your questions?0.653 How clear is the doctor’s information about the consequences of the child’s treatment?0.533 To what extent do you receive clear information about the examinations and tests?0.713 To what extent the information brochure received was complete and clear?0.641 How much clear information is given regarding a child’s illness?0.626 Level of received understandable information about the effects of the drugs?0.606\nCare & Treatment\n Level of child’s comfort taken into account by the doctors and nurses?0.826 The extent of satisfaction During acute situations on availability of nurses to support?0.822 Level of team alertness to the prevention and treatment of pain of neonate?0.797 Level of care taken to nurses while in the incubator/bed?0.6340.501 Level of correct medication always administered on time?0.6310.402 level of emotional support that has been provided?0.84 The doctors and nurses responded well to our own needs0.809 Transferals of care from the neonatal intensive care unit staff to colleagues in the high-care unit or pediatric ward had gone well?0.782 Every day we knew who of the doctors and nurses was responsible for our child.0.695 How closely did doctors and nurses collaborate during work?0.5780.466 Level of a common goal: to provide the finest care and treatment for our child and ourselves.0.774 Level of physicians and nurses paid close attention to our child’s development?0.756 The team as a whole was concerned for our child and you.0.698 Our child’s requirements were met promptly0.455 The extent of doctors’ and nurses’ professional knowledge of what they are doing?0.847 How satisfied are you with the doctors’ and nurses’ understanding of the child’s medical history at the time of admission?0.765 How satisfied are you with the rapid actions taken by doctors and nurses when a child’s condition deteriorated?0.723\nParental Participation\n How involved are you in making decisions about our child’s care and treatment?0.803 The nurses had trained us on the specific aspects of newborn care.0.796 We were encouraged to stay close to our children.0.714 Before discharge, the care for our child was once more discussed with us.0.617 Even during intensive procedures, we could always stay close to our child.0.841 The nurses stimulated us to help in the care of our child0.837 The nurses helped us in the bonding with our child0.5320.575 We had confidence in the team0.561\nOrganization\n The Neonatology unit made us feel safe0.885 There was a warm atmosphere in the Neonatology unit without hostility0.805 The Neonatology unit was clean0.7330.404 The unit could easily be reached by telephone0.809 Our child’s incubator or bed was clean0.8 The team worked efficiently0.661 There was enough space around our child’s incubator/bed0.9 Noise in the unit was muffled as good as possible0.788\nProfessional Attitude\n Our child’s health always came first for the doctors and nurses0.799 The team worked hygienically0.747 Our cultural background was taken into account0.728 The doctors and nurses always took time to listen to us0.876 We felt welcome by the team0.804 Despite the workload, sufficient attention was paid to our child and us by the team0.5840.508 Nurses and doctors always introduced themselves by name and function0.794 We received sympathy from the doctors and nurses0.786 At our bedside, the discussion between the doctors and nurses was only about our child.0.4570.582 The team respected the privacy of our children and us.0.874 There was a pleasant atmosphere among the staff0.743 The team showed respect for our child and us0.5190.609\n\nFactor loadings and identified components’ of EMPATHIC-N tool\nBefore assessing parental satisfaction levels, confirmatory factor analysis was used to ensure that the measured variables accurately represented the constructs. The factor correlation matrix was used in the analysis. KMO and Bartlett’s tests of sphericity were used to check the correlation of measurement variables, with a KMO value of 0.875 and Bartlett’s tests of sphericity (p = 0.00) respectively. The correlation matrix was checked for commonality extraction, and all item values were larger than 0.3. The parallel analysis determined the number of item components (factors), and two new components for parental participation, three new components for information and organization, and four new components for Care & Treatment and professional attitude of eigenvalues met the criteria, and the Varimax component correlation matrix was an appropriate model (Table 2).\n\nTable 2Factor loadings and identified components’ of EMPATHIC-N toolItemFactors\n1\n\n2\n\n3\n\n4\n\nInformation\n What level of doctor’s information do you have regarding the child’s expected health outcomes?0.752 How satisfied are you with the physicians’ and nurses’ information similarity?0.704 How are you satisfied with doctors’ and nurses’ honesty in providing information?0.59 How satisfied are you with daily discussions with doctors and nurses about your child’s care and treatment?0.58 How understandable was the information provided by the doctors and nurses?0.763 How satisfied were you with the correct information when the child’s physical condition deteriorated?0.4280.657 How satisfied were you with the clear answers to your questions?0.653 How clear is the doctor’s information about the consequences of the child’s treatment?0.533 To what extent do you receive clear information about the examinations and tests?0.713 To what extent the information brochure received was complete and clear?0.641 How much clear information is given regarding a child’s illness?0.626 Level of received understandable information about the effects of the drugs?0.606\nCare & Treatment\n Level of child’s comfort taken into account by the doctors and nurses?0.826 The extent of satisfaction During acute situations on availability of nurses to support?0.822 Level of team alertness to the prevention and treatment of pain of neonate?0.797 Level of care taken to nurses while in the incubator/bed?0.6340.501 Level of correct medication always administered on time?0.6310.402 level of emotional support that has been provided?0.84 The doctors and nurses responded well to our own needs0.809 Transferals of care from the neonatal intensive care unit staff to colleagues in the high-care unit or pediatric ward had gone well?0.782 Every day we knew who of the doctors and nurses was responsible for our child.0.695 How closely did doctors and nurses collaborate during work?0.5780.466 Level of a common goal: to provide the finest care and treatment for our child and ourselves.0.774 Level of physicians and nurses paid close attention to our child’s development?0.756 The team as a whole was concerned for our child and you.0.698 Our child’s requirements were met promptly0.455 The extent of doctors’ and nurses’ professional knowledge of what they are doing?0.847 How satisfied are you with the doctors’ and nurses’ understanding of the child’s medical history at the time of admission?0.765 How satisfied are you with the rapid actions taken by doctors and nurses when a child’s condition deteriorated?0.723\nParental Participation\n How involved are you in making decisions about our child’s care and treatment?0.803 The nurses had trained us on the specific aspects of newborn care.0.796 We were encouraged to stay close to our children.0.714 Before discharge, the care for our child was once more discussed with us.0.617 Even during intensive procedures, we could always stay close to our child.0.841 The nurses stimulated us to help in the care of our child0.837 The nurses helped us in the bonding with our child0.5320.575 We had confidence in the team0.561\nOrganization\n The Neonatology unit made us feel safe0.885 There was a warm atmosphere in the Neonatology unit without hostility0.805 The Neonatology unit was clean0.7330.404 The unit could easily be reached by telephone0.809 Our child’s incubator or bed was clean0.8 The team worked efficiently0.661 There was enough space around our child’s incubator/bed0.9 Noise in the unit was muffled as good as possible0.788\nProfessional Attitude\n Our child’s health always came first for the doctors and nurses0.799 The team worked hygienically0.747 Our cultural background was taken into account0.728 The doctors and nurses always took time to listen to us0.876 We felt welcome by the team0.804 Despite the workload, sufficient attention was paid to our child and us by the team0.5840.508 Nurses and doctors always introduced themselves by name and function0.794 We received sympathy from the doctors and nurses0.786 At our bedside, the discussion between the doctors and nurses was only about our child.0.4570.582 The team respected the privacy of our children and us.0.874 There was a pleasant atmosphere among the staff0.743 The team showed respect for our child and us0.5190.609\n\nFactor loadings and identified components’ of EMPATHIC-N tool\n[SUBTITLE] Reliability of items of EMPATHIC-N tool [SUBSECTION] The total EMPATHIC-N values and the five domains showed a good level of internal consistency. The five domains’ Cronbach’s values range from 0.639 to 0.791, whereas the total EMPATHIC-N Cronbach’s value was 0.904. The inter-item correlation (IIC) of the five domains had significant internal consistency (Table 3).\n\nTable 3Reliability of items of parental satisfaction with NICU-care serviceDimensionNumber of ItemsΧηρονβαχη αMean dimension score (SD)Maximum possible dimension scoreInter-item correlation (IIC)Item-discriminant validity (IDV)Information120.63939.55(8.42)720.22–0.48*0.20–0.57Care & Treatment170.79181.70(20.49)1700.14–0.64*0.25–0.38Parental Participation80.75524.86(7.79)480.01–0.71*0.37–0.54Organization80.72925.22(7.39)480.11–0.64*0.34–0.69Professional Attitude120.69438.106 (9.07)720.13–0.56*0.22–0.45EMPATHIC-N tool570.904209.44(42.82)410cc*Significant value (p < 0.001), c = not computable\n\nReliability of items of parental satisfaction with NICU-care service\n*Significant value (p < 0.001), c = not computable\nThe total EMPATHIC-N values and the five domains showed a good level of internal consistency. The five domains’ Cronbach’s values range from 0.639 to 0.791, whereas the total EMPATHIC-N Cronbach’s value was 0.904. The inter-item correlation (IIC) of the five domains had significant internal consistency (Table 3).\n\nTable 3Reliability of items of parental satisfaction with NICU-care serviceDimensionNumber of ItemsΧηρονβαχη αMean dimension score (SD)Maximum possible dimension scoreInter-item correlation (IIC)Item-discriminant validity (IDV)Information120.63939.55(8.42)720.22–0.48*0.20–0.57Care & Treatment170.79181.70(20.49)1700.14–0.64*0.25–0.38Parental Participation80.75524.86(7.79)480.01–0.71*0.37–0.54Organization80.72925.22(7.39)480.11–0.64*0.34–0.69Professional Attitude120.69438.106 (9.07)720.13–0.56*0.22–0.45EMPATHIC-N tool570.904209.44(42.82)410cc*Significant value (p < 0.001), c = not computable\n\nReliability of items of parental satisfaction with NICU-care service\n*Significant value (p < 0.001), c = not computable\n[SUBTITLE] Parental satisfaction level with NICU-care service [SUBSECTION] Overall mean satisfaction level of parents in NICU-care service was 47.8% [95% CI= (43.1–52.5)]. The domains of parental satisfaction scores were compared with each other and, the lowest parental satisfaction score was for the domain of care and treatment 36.9% while the organizational domain showed the highest parental satisfaction level 59.0%. The domains of information, parental participation, and professional attitude showed comparable parental satisfaction scores (Fig. 1).\n\nFig. 1Parents’ overall and dimensional satisfaction with NICU-care services\n\nParents’ overall and dimensional satisfaction with NICU-care services\nOverall mean satisfaction level of parents in NICU-care service was 47.8% [95% CI= (43.1–52.5)]. The domains of parental satisfaction scores were compared with each other and, the lowest parental satisfaction score was for the domain of care and treatment 36.9% while the organizational domain showed the highest parental satisfaction level 59.0%. The domains of information, parental participation, and professional attitude showed comparable parental satisfaction scores (Fig. 1).\n\nFig. 1Parents’ overall and dimensional satisfaction with NICU-care services\n\nParents’ overall and dimensional satisfaction with NICU-care services\n[SUBTITLE] Factors associated with the overall parental satisfaction of NICU care service [SUBSECTION] In this study, the bivariable logistic regression showed that parental gender, residency, parental hospital stay, neonatal hospital stay, birth weight, and gestational age were factors with a p-value of less than 0.2 and were fitted with a multivariable logistic regression model. However, neonatal hospital stay was not associated with multivariable logistic regression with a p-value greater than 0.05. The multivariable logistic analyses showed that mothers were 2.16 (AOR = 2.16; 95%CI: 1.28–3.63) times more satisfied than fathers. Also, parents who are from the rural area were 2.94 (AOR = 2.94; 95%CI: 1.42–6.06) more satisfied than urban. Parents who say less than 15 days were 2.18 (AOR = 2.18; 95%CI: 1.13–4.20) times more satisfied than parents who stay 15 or more days in the hospital. Also, parents of a neonate with a normal birth weight of 2.14 (AOR = 2.14; 95%CI: 1.16–3.94) and full-term neonate 2.53(AOR = 2.53; 95%CI: 1.29–4.97) times more satisfied than their counterparts (Table 4).\n\nTable 4Factors associated with satisfaction of parents in NICU-care service (n = 385)VariablesSatisfaction level on NICU- serviceCrude odds ratioAdjusted odds ratiop-valueSatisfiedNot Satisfied(95% CI)(95% CI)\nGender\n Mother117(52.2%)107(47.8%)1.53(1.02,2.31)2.16(1.28,3.63)0.004* Father67(41.6%)94(58.4%)1\nResidency\n Urban68(36.4%)119(63.6%)1 Rural116(58.6%)82(41.4%)2.48(1.64,3.73)2.94(1.42,6.06)0.004*\nParental hospital stay\n ≤ 15 days140(57.4%)104(42.6%)2.97(1.92,4.59)2.18(1.13,4.20)0.019* > 15days44(31.2%)97(68.8%)1\nBirth weight\n High birth weight34(41.0%)49(59.0%)1 Normal birth weight96(59.3%)66(40.7%)2.09(1.22,3.59)2.14(1.16,3.94)0.015* Low birth weight54(38.6%)86(61.4%)0.91(0.52,1.58)0.76(0.41,1.41)0.38\nGestational age\n Premature18(30.0%)42(70.0%)1 Full term166(51.1%)159(48.9%)2.44(1.35,4.41)2.53(1.29,4.97)0.007**= p-value < 0.05 1 = reference BWt-birth weightNote: p-values were extracted from the multivariate logistic regression model\n\nFactors associated with satisfaction of parents in NICU-care service (n = 385)\n*= p-value < 0.05 1 = reference BWt-birth weight\nNote: p-values were extracted from the multivariate logistic regression model\nIn this study, the bivariable logistic regression showed that parental gender, residency, parental hospital stay, neonatal hospital stay, birth weight, and gestational age were factors with a p-value of less than 0.2 and were fitted with a multivariable logistic regression model. However, neonatal hospital stay was not associated with multivariable logistic regression with a p-value greater than 0.05. The multivariable logistic analyses showed that mothers were 2.16 (AOR = 2.16; 95%CI: 1.28–3.63) times more satisfied than fathers. Also, parents who are from the rural area were 2.94 (AOR = 2.94; 95%CI: 1.42–6.06) more satisfied than urban. Parents who say less than 15 days were 2.18 (AOR = 2.18; 95%CI: 1.13–4.20) times more satisfied than parents who stay 15 or more days in the hospital. Also, parents of a neonate with a normal birth weight of 2.14 (AOR = 2.14; 95%CI: 1.16–3.94) and full-term neonate 2.53(AOR = 2.53; 95%CI: 1.29–4.97) times more satisfied than their counterparts (Table 4).\n\nTable 4Factors associated with satisfaction of parents in NICU-care service (n = 385)VariablesSatisfaction level on NICU- serviceCrude odds ratioAdjusted odds ratiop-valueSatisfiedNot Satisfied(95% CI)(95% CI)\nGender\n Mother117(52.2%)107(47.8%)1.53(1.02,2.31)2.16(1.28,3.63)0.004* Father67(41.6%)94(58.4%)1\nResidency\n Urban68(36.4%)119(63.6%)1 Rural116(58.6%)82(41.4%)2.48(1.64,3.73)2.94(1.42,6.06)0.004*\nParental hospital stay\n ≤ 15 days140(57.4%)104(42.6%)2.97(1.92,4.59)2.18(1.13,4.20)0.019* > 15days44(31.2%)97(68.8%)1\nBirth weight\n High birth weight34(41.0%)49(59.0%)1 Normal birth weight96(59.3%)66(40.7%)2.09(1.22,3.59)2.14(1.16,3.94)0.015* Low birth weight54(38.6%)86(61.4%)0.91(0.52,1.58)0.76(0.41,1.41)0.38\nGestational age\n Premature18(30.0%)42(70.0%)1 Full term166(51.1%)159(48.9%)2.44(1.35,4.41)2.53(1.29,4.97)0.007**= p-value < 0.05 1 = reference BWt-birth weightNote: p-values were extracted from the multivariate logistic regression model\n\nFactors associated with satisfaction of parents in NICU-care service (n = 385)\n*= p-value < 0.05 1 = reference BWt-birth weight\nNote: p-values were extracted from the multivariate logistic regression model", "Before assessing parental satisfaction levels, confirmatory factor analysis was used to ensure that the measured variables accurately represented the constructs. The factor correlation matrix was used in the analysis. KMO and Bartlett’s tests of sphericity were used to check the correlation of measurement variables, with a KMO value of 0.875 and Bartlett’s tests of sphericity (p = 0.00) respectively. The correlation matrix was checked for commonality extraction, and all item values were larger than 0.3. The parallel analysis determined the number of item components (factors), and two new components for parental participation, three new components for information and organization, and four new components for Care & Treatment and professional attitude of eigenvalues met the criteria, and the Varimax component correlation matrix was an appropriate model (Table 2).\n\nTable 2Factor loadings and identified components’ of EMPATHIC-N toolItemFactors\n1\n\n2\n\n3\n\n4\n\nInformation\n What level of doctor’s information do you have regarding the child’s expected health outcomes?0.752 How satisfied are you with the physicians’ and nurses’ information similarity?0.704 How are you satisfied with doctors’ and nurses’ honesty in providing information?0.59 How satisfied are you with daily discussions with doctors and nurses about your child’s care and treatment?0.58 How understandable was the information provided by the doctors and nurses?0.763 How satisfied were you with the correct information when the child’s physical condition deteriorated?0.4280.657 How satisfied were you with the clear answers to your questions?0.653 How clear is the doctor’s information about the consequences of the child’s treatment?0.533 To what extent do you receive clear information about the examinations and tests?0.713 To what extent the information brochure received was complete and clear?0.641 How much clear information is given regarding a child’s illness?0.626 Level of received understandable information about the effects of the drugs?0.606\nCare & Treatment\n Level of child’s comfort taken into account by the doctors and nurses?0.826 The extent of satisfaction During acute situations on availability of nurses to support?0.822 Level of team alertness to the prevention and treatment of pain of neonate?0.797 Level of care taken to nurses while in the incubator/bed?0.6340.501 Level of correct medication always administered on time?0.6310.402 level of emotional support that has been provided?0.84 The doctors and nurses responded well to our own needs0.809 Transferals of care from the neonatal intensive care unit staff to colleagues in the high-care unit or pediatric ward had gone well?0.782 Every day we knew who of the doctors and nurses was responsible for our child.0.695 How closely did doctors and nurses collaborate during work?0.5780.466 Level of a common goal: to provide the finest care and treatment for our child and ourselves.0.774 Level of physicians and nurses paid close attention to our child’s development?0.756 The team as a whole was concerned for our child and you.0.698 Our child’s requirements were met promptly0.455 The extent of doctors’ and nurses’ professional knowledge of what they are doing?0.847 How satisfied are you with the doctors’ and nurses’ understanding of the child’s medical history at the time of admission?0.765 How satisfied are you with the rapid actions taken by doctors and nurses when a child’s condition deteriorated?0.723\nParental Participation\n How involved are you in making decisions about our child’s care and treatment?0.803 The nurses had trained us on the specific aspects of newborn care.0.796 We were encouraged to stay close to our children.0.714 Before discharge, the care for our child was once more discussed with us.0.617 Even during intensive procedures, we could always stay close to our child.0.841 The nurses stimulated us to help in the care of our child0.837 The nurses helped us in the bonding with our child0.5320.575 We had confidence in the team0.561\nOrganization\n The Neonatology unit made us feel safe0.885 There was a warm atmosphere in the Neonatology unit without hostility0.805 The Neonatology unit was clean0.7330.404 The unit could easily be reached by telephone0.809 Our child’s incubator or bed was clean0.8 The team worked efficiently0.661 There was enough space around our child’s incubator/bed0.9 Noise in the unit was muffled as good as possible0.788\nProfessional Attitude\n Our child’s health always came first for the doctors and nurses0.799 The team worked hygienically0.747 Our cultural background was taken into account0.728 The doctors and nurses always took time to listen to us0.876 We felt welcome by the team0.804 Despite the workload, sufficient attention was paid to our child and us by the team0.5840.508 Nurses and doctors always introduced themselves by name and function0.794 We received sympathy from the doctors and nurses0.786 At our bedside, the discussion between the doctors and nurses was only about our child.0.4570.582 The team respected the privacy of our children and us.0.874 There was a pleasant atmosphere among the staff0.743 The team showed respect for our child and us0.5190.609\n\nFactor loadings and identified components’ of EMPATHIC-N tool", "The total EMPATHIC-N values and the five domains showed a good level of internal consistency. The five domains’ Cronbach’s values range from 0.639 to 0.791, whereas the total EMPATHIC-N Cronbach’s value was 0.904. The inter-item correlation (IIC) of the five domains had significant internal consistency (Table 3).\n\nTable 3Reliability of items of parental satisfaction with NICU-care serviceDimensionNumber of ItemsΧηρονβαχη αMean dimension score (SD)Maximum possible dimension scoreInter-item correlation (IIC)Item-discriminant validity (IDV)Information120.63939.55(8.42)720.22–0.48*0.20–0.57Care & Treatment170.79181.70(20.49)1700.14–0.64*0.25–0.38Parental Participation80.75524.86(7.79)480.01–0.71*0.37–0.54Organization80.72925.22(7.39)480.11–0.64*0.34–0.69Professional Attitude120.69438.106 (9.07)720.13–0.56*0.22–0.45EMPATHIC-N tool570.904209.44(42.82)410cc*Significant value (p < 0.001), c = not computable\n\nReliability of items of parental satisfaction with NICU-care service\n*Significant value (p < 0.001), c = not computable", "Overall mean satisfaction level of parents in NICU-care service was 47.8% [95% CI= (43.1–52.5)]. The domains of parental satisfaction scores were compared with each other and, the lowest parental satisfaction score was for the domain of care and treatment 36.9% while the organizational domain showed the highest parental satisfaction level 59.0%. The domains of information, parental participation, and professional attitude showed comparable parental satisfaction scores (Fig. 1).\n\nFig. 1Parents’ overall and dimensional satisfaction with NICU-care services\n\nParents’ overall and dimensional satisfaction with NICU-care services", "In this study, the bivariable logistic regression showed that parental gender, residency, parental hospital stay, neonatal hospital stay, birth weight, and gestational age were factors with a p-value of less than 0.2 and were fitted with a multivariable logistic regression model. However, neonatal hospital stay was not associated with multivariable logistic regression with a p-value greater than 0.05. The multivariable logistic analyses showed that mothers were 2.16 (AOR = 2.16; 95%CI: 1.28–3.63) times more satisfied than fathers. Also, parents who are from the rural area were 2.94 (AOR = 2.94; 95%CI: 1.42–6.06) more satisfied than urban. Parents who say less than 15 days were 2.18 (AOR = 2.18; 95%CI: 1.13–4.20) times more satisfied than parents who stay 15 or more days in the hospital. Also, parents of a neonate with a normal birth weight of 2.14 (AOR = 2.14; 95%CI: 1.16–3.94) and full-term neonate 2.53(AOR = 2.53; 95%CI: 1.29–4.97) times more satisfied than their counterparts (Table 4).\n\nTable 4Factors associated with satisfaction of parents in NICU-care service (n = 385)VariablesSatisfaction level on NICU- serviceCrude odds ratioAdjusted odds ratiop-valueSatisfiedNot Satisfied(95% CI)(95% CI)\nGender\n Mother117(52.2%)107(47.8%)1.53(1.02,2.31)2.16(1.28,3.63)0.004* Father67(41.6%)94(58.4%)1\nResidency\n Urban68(36.4%)119(63.6%)1 Rural116(58.6%)82(41.4%)2.48(1.64,3.73)2.94(1.42,6.06)0.004*\nParental hospital stay\n ≤ 15 days140(57.4%)104(42.6%)2.97(1.92,4.59)2.18(1.13,4.20)0.019* > 15days44(31.2%)97(68.8%)1\nBirth weight\n High birth weight34(41.0%)49(59.0%)1 Normal birth weight96(59.3%)66(40.7%)2.09(1.22,3.59)2.14(1.16,3.94)0.015* Low birth weight54(38.6%)86(61.4%)0.91(0.52,1.58)0.76(0.41,1.41)0.38\nGestational age\n Premature18(30.0%)42(70.0%)1 Full term166(51.1%)159(48.9%)2.44(1.35,4.41)2.53(1.29,4.97)0.007**= p-value < 0.05 1 = reference BWt-birth weightNote: p-values were extracted from the multivariate logistic regression model\n\nFactors associated with satisfaction of parents in NICU-care service (n = 385)\n*= p-value < 0.05 1 = reference BWt-birth weight\nNote: p-values were extracted from the multivariate logistic regression model", "In this study, the total average parental satisfaction score with NICU service was 47.8%. This finding is nearly similar to a study done in Ethiopia (50%) [26]. Contrary to this finding, studies done in Norway (76%) [7] and Greece (99%) [27] had a higher parental satisfaction score with NICU service.\nThe study found that parental satisfaction with NICU service was 50.4% in the information subscale, 36.9% in the care and treatment subscale, 50.1% in the parental participation subscale, 59.0% in the organization subscale, and 48.6% in the professional attitude subscale. Parental satisfaction was lowest in the care and treatment subscale. A study in Italy [20] and South Africa [28] on the other hand, discovered the lowest parental satisfaction score in the professional attitude and parental participation subscales respectively. This disparity could be attributed to the NICU’s lack of professional and medical resources to treat and care for neonates in this situation.\nAccording to this study, mothers, parents from rural regions, parents of neonates who stayed less than 15 days in the hospital, parents of neonates with normal birth weight, and parents of full-term neonates were all more satisfied with NICU services than their counterparts.\nThis study showed that mothers were more satisfied with NICU service than fathers. Similar studies conducted in Italy [20], Israel [11], and Greece [29] also showed that mothers were more satisfied than fathers in NICU service. The possible explanation might be women are allowed to spend more time in the NICU and participate in the care of their newborns and cultivate more relationships with medical caregivers than fathers.\nAlso in this study parents who came from rural areas were more satisfied than parents from urban areas. This founding was also similar to a study done in Greece [30] found that parents from rural areas were more satisfied. The probable reason for this might be parents from rural areas may have a low-level awareness of the hospital, expectations, and demand for NICU service as compared with actual practice.\nParents of normal birth weight and full-term neonates were more satisfied with NICU service than parents of low birth weight and preterm neonates in this study. In addition, a Norwegian study [7] found that parents of newborns with normal birth weights were more satisfied than those with low birth weights. Reasonably, parents of full-term and normal-weight neonates are likely to face unexpected concerns, which may lead to greater satisfaction with positive outcomes.\nIn this study, parents of neonates who stayed less than 15 days in the hospital were more satisfied with NICU service than parents who stayed 15 or more days. Similarly, studies done in Ethiopia [26] and Iran [10] found parents who stayed for a short period in the NICU were more satisfied. The possible reason for this might be that parents with short-stay are less likely to see their neonatal serious conditions that make emotional and care mismanagements.\n[SUBTITLE] Limitations of the study [SUBSECTION] The study’s limitations include being a single-center nature and the lack of analysis of various alternatives to logistic regression that might be more applicable for this investigation.\nThe study’s limitations include being a single-center nature and the lack of analysis of various alternatives to logistic regression that might be more applicable for this investigation.", "The study’s limitations include being a single-center nature and the lack of analysis of various alternatives to logistic regression that might be more applicable for this investigation.", "There was a low level of parental satisfaction with neonatal intensive care unit service. Among the dimensions of EMPATHIC-N, the lowest parental satisfaction score was in the care and treatment while the highest parental satisfaction score was in the organization dimension. As a result, health professionals and hospital administrators should collaborate to improve NICU services to provide high-quality service and satisfy parents." ]
[ "introduction", null, null, null, null, null, null, null, null, null, "results", null, null, null, null, "discussion", null, "conclusion" ]
[ "NICU", "Parents", "Satisfaction", "Service" ]
'Corona is coming': COVID-19 vaccination perspectives and experiences amongst Culturally and Linguistically Diverse West Australians.
36262050
Culturally and Linguistically Diverse (CALD) groups within high-income countries are at risk of being left behind by the COVID-19 vaccination rollout. They face both access and attitudinal barriers, including low trust in government and health authorities.
BACKGROUND
Perth, WA's capital, was chosen as the state's study site because most of the state's CALD population lives there. Eleven semistructured in-depth interviews and three focus groups (with 37 participants) were conducted with CALD residents between August and October 2021. Thematic analysis was conducted, informed by the 'Capability', 'Opportunity', 'Motivation', 'Behaviour' model.
DESIGN AND PARTICIPANTS
CALD participants faced barriers including a lack of knowledge about COVID-19 and the vaccines, low self-rated English proficiency and education levels, misinformation, passive government communication strategies and limited access to vaccine clinics/providers. They were, however, motivated to vaccinate by the imminent opening of state and international borders, trust in government and healthcare authorities, travel intentions and the desire to protect themselves and others.
RESULTS
Despite high levels of trust and significant desire for vaccines among CALD communities in Perth, current strategies were not meeting their needs and the community remains at risk from COVID-19. Tailored intervention strategies are required to provide knowledge, address misinformation and facilitate access to ensure uptake of COVID-19 vaccines-including for additional doses-amongst CALD communities. Governments should work with trusted CALD community members to disseminate tailored COVID-19 vaccine information and adequately translated resources.
CONCLUSIONS
[ "Humans", "Female", "COVID-19 Vaccines", "Australia", "COVID-19", "Cultural Diversity", "Vaccination" ]
9700143
null
null
METHODS
[SUBTITLE] Study design [SUBSECTION] This CALD study was conducted as part of the larger mixed methods research project, Coronavax. Coronavax was initiated in April 2020 with the aim of ascertaining community attitudes towards vaccination and vaccine access in WA through a series of qualitative interviews with segmented population groups. A key aspect of the project involved translating findings to the state and federal governments in real‐time via Functional Dialogues, a novel formal research exchange mechanism in which data on community attitudes are presented to government officials, who then contribute their own reflections and experiences as additional research data (not reported here). 12 Coronavax received ethics approval from the Child and Adolescent Services (CAHS) Human Research Ethics Committee (RGS0000004457). This CALD study was conducted as part of the larger mixed methods research project, Coronavax. Coronavax was initiated in April 2020 with the aim of ascertaining community attitudes towards vaccination and vaccine access in WA through a series of qualitative interviews with segmented population groups. A key aspect of the project involved translating findings to the state and federal governments in real‐time via Functional Dialogues, a novel formal research exchange mechanism in which data on community attitudes are presented to government officials, who then contribute their own reflections and experiences as additional research data (not reported here). 12 Coronavax received ethics approval from the Child and Adolescent Services (CAHS) Human Research Ethics Committee (RGS0000004457). [SUBTITLE] Consumer consultation process [SUBSECTION] Consumer reference for Coronavax is led by Ms. Catherine Hughes who also chairs the Wesfarmers Centre of Vaccines and Infectious Diseases Community Reference Group at Telethon Kids Institute. This Group consulted on Coronavax in September 2020. Consumer involvement in Coronavax has sought to ensure the appropriateness and sensitivity of research questions and recruitment and to ensure that the research priorities and design reflect the needs of the community. Ishar Multicultural Women's Health Services, one of the largest multicultural health providers in Western Australia, provided additional consultation for this CALD study, as well as aiding with the recruitment of their clients, providing facilities, and hosting the focus groups. Consumer reference for Coronavax is led by Ms. Catherine Hughes who also chairs the Wesfarmers Centre of Vaccines and Infectious Diseases Community Reference Group at Telethon Kids Institute. This Group consulted on Coronavax in September 2020. Consumer involvement in Coronavax has sought to ensure the appropriateness and sensitivity of research questions and recruitment and to ensure that the research priorities and design reflect the needs of the community. Ishar Multicultural Women's Health Services, one of the largest multicultural health providers in Western Australia, provided additional consultation for this CALD study, as well as aiding with the recruitment of their clients, providing facilities, and hosting the focus groups. [SUBTITLE] Study setting [SUBSECTION] In the first 2 years of the COVID‐19 pandemic, WA experienced just over 1100 cases in a population of approximately 2.6 million people, 13 with transmission rapidly increasing only from February 2022. 13 As of March 2022, over 95% of people aged 12 years and older in WA had received at least two COVID‐19 vaccine doses, 14 and over 85.5% of those eligible for their third dose in WA having received it. 15 However, coverage rates at the time of our study were lower, as Australia had faced significant supply problems early in the rollout. 16 Responsibility for vaccine operations sat with both State and Federal Governments. WA residents access COVID‐19 vaccines through general practice clinics, federal respiratory clinics, pharmacies, community health services and state‐led mass vaccination clinics. As governments sought to rapidly increase vaccine uptake across the entire population, populations requiring more complex and bespoke interventions, such as CALD communities, remained under‐served. For example, Local Government Areas (LGAs) in Perth with the highest proportion of those born in nonmain English‐speaking countries had some of the lowest uptake in Perth 8 months into the rollout. 17 A more tailored approach to CALD communities commenced in late 2021 after our study, including pop‐up vaccination clinics at major religious events (e.g., the Indian festival of Diwali, November 2021), or at multicultural community centres. The state‐wide campaign ‘Roll Up For WA’ translated resources into over 50 non‐English languages, 18 although at the time of our study only approximately 20 were in place. CALD individuals comprise almost one‐fifth of the WA population. 19 In WA, 90% of individuals born in what the Australian Bureau of Statistics calls ‘non‐main English‐speaking countries’ reside in the Perth metropolitan region, 19 so we focused on recruiting CALD adults living in Perth. In the first 2 years of the COVID‐19 pandemic, WA experienced just over 1100 cases in a population of approximately 2.6 million people, 13 with transmission rapidly increasing only from February 2022. 13 As of March 2022, over 95% of people aged 12 years and older in WA had received at least two COVID‐19 vaccine doses, 14 and over 85.5% of those eligible for their third dose in WA having received it. 15 However, coverage rates at the time of our study were lower, as Australia had faced significant supply problems early in the rollout. 16 Responsibility for vaccine operations sat with both State and Federal Governments. WA residents access COVID‐19 vaccines through general practice clinics, federal respiratory clinics, pharmacies, community health services and state‐led mass vaccination clinics. As governments sought to rapidly increase vaccine uptake across the entire population, populations requiring more complex and bespoke interventions, such as CALD communities, remained under‐served. For example, Local Government Areas (LGAs) in Perth with the highest proportion of those born in nonmain English‐speaking countries had some of the lowest uptake in Perth 8 months into the rollout. 17 A more tailored approach to CALD communities commenced in late 2021 after our study, including pop‐up vaccination clinics at major religious events (e.g., the Indian festival of Diwali, November 2021), or at multicultural community centres. The state‐wide campaign ‘Roll Up For WA’ translated resources into over 50 non‐English languages, 18 although at the time of our study only approximately 20 were in place. CALD individuals comprise almost one‐fifth of the WA population. 19 In WA, 90% of individuals born in what the Australian Bureau of Statistics calls ‘non‐main English‐speaking countries’ reside in the Perth metropolitan region, 19 so we focused on recruiting CALD adults living in Perth. [SUBTITLE] Sampling, recruitment and data collection [SUBSECTION] Interviewees were recruited through CALD organizations, social media (including a specific campaign directed towards CALD men), and promotional campaigns (in English) that ran from August to October 2021. Interviewees voluntarily signed up via the secure web‐based survey platform REDCap hosted by Telethon Kids Institute. 20 , 21 Here, they provided demographic and contact information. They were selected if they met the CALD definition and then contacted up to three times each via phone or email to organize the interview. The CALD definition was based on the Australian Bureau of Statistics' definition, 22 and included individuals born overseas, having one or both parents born overseas from non‐English speaking countries, or whose first language is a Language Other Than English (LOTE). Interviews were conducted in English by SC over the phone or via teleconferencing software—interviewees who signed up all had sufficient English proficiency as the self‐sign‐up process was only in English. Due to the high levels of English proficiency required for interview sign‐ups, we approached Ishar to partner on this research so that we could engage with CALD individuals who had lower English proficiency. Accordingly, focus group participants were recruited separately through their CALD community centre for women based in Stirling, the LGA with the highest proportion of individuals born in nonmain English‐speaking countries. 19 Women attending regular English classes were invited face‐to‐face by researchers and their teachers to participate in a focus group in lieu of their class. Focus groups were run face‐to‐face by S. J. C., G. E. and K. A. in the CALD community centre; they were conducted in English with the use of peer translators for participants with low English proficiency. Data were collected between August and October 2021. Of the 23 CALD individuals who voluntarily signed up online to participate in the individual interviews, we interviewed 11, with the remaining 12 being lost to follow‐up. We ran three focus groups at the Ishar multicultural centre with their female clients, comprised of 6–16 participants per group (total N = 37). All women who were present consented and participated in a focus group—there were no refusals. Interviews and focus groups lasted approximately 60 min. Of the total 48 participants, 90% were female and 29% were aged 30–39 years. Approximately, one third (35%) had been to university, 54% self‐reported high English proficiency and 27% reported having received at least their first COVID‐19 vaccine (Table 1). Further, 46% had migrated to Australia since 2010, 35% were born in Southern Asia, and 75% identified with a religion (most common being Islam and Christianity). Two of the 11 interviewees self‐identified as community leaders. Interviewees gave voluntary, informed, written consent; focus group participants gave voluntary, informed, verbal consent. All have been assigned pseudonyms. Sociodemographic characteristics of the 48 CALD participants, Perth, WA Abbreviations: CALD, Culturally and Linguistically Diverse; TAFE, Technical and Further Education; WA, Western Australia. Some participants spoke ≥1 language. Other languages include Acholi, Azeri, Bomwali, Hazaragi, Hindi, Ilocano, Indonesian, Italian, Kikuyu (Bantu), Korean, Macedonian, Malay, Oromo, Somali, Tedim, Urdu and Vietnamese. Interviewees were recruited through CALD organizations, social media (including a specific campaign directed towards CALD men), and promotional campaigns (in English) that ran from August to October 2021. Interviewees voluntarily signed up via the secure web‐based survey platform REDCap hosted by Telethon Kids Institute. 20 , 21 Here, they provided demographic and contact information. They were selected if they met the CALD definition and then contacted up to three times each via phone or email to organize the interview. The CALD definition was based on the Australian Bureau of Statistics' definition, 22 and included individuals born overseas, having one or both parents born overseas from non‐English speaking countries, or whose first language is a Language Other Than English (LOTE). Interviews were conducted in English by SC over the phone or via teleconferencing software—interviewees who signed up all had sufficient English proficiency as the self‐sign‐up process was only in English. Due to the high levels of English proficiency required for interview sign‐ups, we approached Ishar to partner on this research so that we could engage with CALD individuals who had lower English proficiency. Accordingly, focus group participants were recruited separately through their CALD community centre for women based in Stirling, the LGA with the highest proportion of individuals born in nonmain English‐speaking countries. 19 Women attending regular English classes were invited face‐to‐face by researchers and their teachers to participate in a focus group in lieu of their class. Focus groups were run face‐to‐face by S. J. C., G. E. and K. A. in the CALD community centre; they were conducted in English with the use of peer translators for participants with low English proficiency. Data were collected between August and October 2021. Of the 23 CALD individuals who voluntarily signed up online to participate in the individual interviews, we interviewed 11, with the remaining 12 being lost to follow‐up. We ran three focus groups at the Ishar multicultural centre with their female clients, comprised of 6–16 participants per group (total N = 37). All women who were present consented and participated in a focus group—there were no refusals. Interviews and focus groups lasted approximately 60 min. Of the total 48 participants, 90% were female and 29% were aged 30–39 years. Approximately, one third (35%) had been to university, 54% self‐reported high English proficiency and 27% reported having received at least their first COVID‐19 vaccine (Table 1). Further, 46% had migrated to Australia since 2010, 35% were born in Southern Asia, and 75% identified with a religion (most common being Islam and Christianity). Two of the 11 interviewees self‐identified as community leaders. Interviewees gave voluntary, informed, written consent; focus group participants gave voluntary, informed, verbal consent. All have been assigned pseudonyms. Sociodemographic characteristics of the 48 CALD participants, Perth, WA Abbreviations: CALD, Culturally and Linguistically Diverse; TAFE, Technical and Further Education; WA, Western Australia. Some participants spoke ≥1 language. Other languages include Acholi, Azeri, Bomwali, Hazaragi, Hindi, Ilocano, Indonesian, Italian, Kikuyu (Bantu), Korean, Macedonian, Malay, Oromo, Somali, Tedim, Urdu and Vietnamese. [SUBTITLE] Interview guide [SUBSECTION] Interview guides were developed by the multidisciplinary (project) team comprised of vaccination, medical, policy, social science and government scholars, and an indicative guide is published open‐access with our protocol. 12 The questions were simplified for the focus groups. Interview guides were developed by the multidisciplinary (project) team comprised of vaccination, medical, policy, social science and government scholars, and an indicative guide is published open‐access with our protocol. 12 The questions were simplified for the focus groups. [SUBTITLE] Analysis [SUBSECTION] All interviews and focus groups were audio‐recorded and professionally transcribed verbatim. Thematic analysis followed the Braun and Clarke method, 23 with deductive analysis in NVivo 12 using the Capability, Opportunity, Motivation, Behaviour (COM‐B) model. 24 We selected COM‐B as it is used extensively by the World Health Organization to understand reasons for (non)vaccination among specific populations. 25 G. E. generated initial codes, S. J. C. and B. N. each coded a manuscript to provide additional perspectives, including a CALD lived experience lens, and the three coders revised the matrix via a collaborative meeting before GC applied it to the full data set. All interviews and focus groups were audio‐recorded and professionally transcribed verbatim. Thematic analysis followed the Braun and Clarke method, 23 with deductive analysis in NVivo 12 using the Capability, Opportunity, Motivation, Behaviour (COM‐B) model. 24 We selected COM‐B as it is used extensively by the World Health Organization to understand reasons for (non)vaccination among specific populations. 25 G. E. generated initial codes, S. J. C. and B. N. each coded a manuscript to provide additional perspectives, including a CALD lived experience lens, and the three coders revised the matrix via a collaborative meeting before GC applied it to the full data set.
null
null
CONCLUSION
This study provides in‐depth insight into the perceptions and attitudes of CALD communities in Perth, WA, regarding the COVID‐19 vaccine, identifying facilitators and barriers for engagement and uptake. CALD participants faced barriers including a lack of knowledge about COVID‐19 and the vaccines, low English proficiency and education levels, misinformation, passive government communication strategies and limited access to vaccine clinics/providers. They were, however, motivated to vaccinate by the imminent opening of state and international borders, trust in government and healthcare authorities, travel intentions and the desire to protect themselves and others. Despite high levels of trust and significant desire for vaccines, strategies at the time of our study were not meeting the CALD community's needs, and thus those within it remain at risk from the disease. Tailored intervention strategies are required to provide knowledge, address misinformation, and facilitate access to ensure uptake of COVID‐19 vaccines—including for additional doses—amongst CALD communities. Governments must work with trusted CALD community members and immunization providers such as GPs to disseminate tailored COVID‐19 vaccine information and adequately translated resources so that community members themselves can become stronger vaccine ambassadors to each other.
[ "INTRODUCTION", "Study design", "Consumer consultation process", "Study setting", "Sampling, recruitment and data collection", "Interview guide", "Analysis", "FINDINGS", "Capability", "Knowledge about COVID‐19 and COVID‐19 vaccination", "Knowledge about how and where to get vaccinated", "Opportunity", "Relationships with general practitioners (GPs)", "Access to information/misinformation", "Access to vaccine clinics", "Community influence", "Motivation", "IMPLICATIONS FOR POLICY AND PRACTICE", "Limitations", "AUTHOR CONTRIBUTIONS" ]
[ "Scholars and commentators recognize that even within high‐income countries, pockets of populations are at risk of being left behind in the globally inequitable COVID‐19 vaccine rollout.\n1\n Culturally and Linguistically Diverse (CALD) populations, which include refugees and migrants, are at particular risk due to inaccessible or suboptimal healthcare experiences, which may compound attitudinal factors, including vaccine hesitancy.\n2\n, \n3\n Unique local factors can also create barriers: in the case of Western Australia (WA), almost nonexistent levels of community transmission of COVID‐19  led some locals to ‘wait awhile’ before being vaccinated as state borders remained closed throughout 2021.\n4\n Meanwhile, earlier large outbreaks in other Australian states disproportionately affected CALD communities, with significantly higher morbidity and mortality.\n5\n, \n6\n\n\nIn Australia, COVID‐19 vaccination is recorded on the Australian Immunisation Register, however, country of birth and language spoken at home are not captured on this register. Therefore, there is no publicly available data on uptake among CALD communities in Australia. However, an online survey of refugees in Australia in mid‐2021 identified that 88% had not yet received a COVID‐19 vaccine.\n7\n Emerging evidence suggests that COVID‐19 vaccine hesitancy is prevalent among Australian CALD populations.\n8\n, \n9\n Focus groups conducted in mid‐2021 in New South Wales with CALD people indicated that 42% of participants were not planning to be vaccinated, and 29% were unsure or hesitant.\n8\n Many had safety concerns, were uncertain how vaccines work or lacked trust in government.\n8\n These uncertainties are unsurprising, given little of the public health information on COVID‐19 in Australia was tailored towards those with low English proficiency or health literacy.\n10\n Lockyer et al.'s\n11\n qualitative study in multicultural Bradford, UK, also found that participants' trust in government and the National Health Service dropped during the pandemic and that vaccine hesitancy was expressed by ethnic or national minorities.\nIn this study, we sought to understand the perceptions and attitudes to COVID‐19 vaccines amongst WA's CALD communities, to identify barriers preventing uptake, and to consider strategies to promote uptake in this population. Findings can support the development of strategies to enhance uptake among the CALD communities locally, nationwide and in comparable high‐income English‐speaking countries. They are particularly pertinent in the context of needing to enhance third and fourth‐dose rates for adults and protecting CALD children with paediatric COVID‐19 vaccinations.", "This CALD study was conducted as part of the larger mixed methods research project, Coronavax. Coronavax was initiated in April 2020 with the aim of ascertaining community attitudes towards vaccination and vaccine access in WA through a series of qualitative interviews with segmented population groups. A key aspect of the project involved translating findings to the state and federal governments in real‐time via Functional Dialogues, a novel formal research exchange mechanism in which data on community attitudes are presented to government officials, who then contribute their own reflections and experiences as additional research data (not reported here).\n12\n Coronavax received ethics approval from the Child and Adolescent Services (CAHS) Human Research Ethics Committee (RGS0000004457).", "Consumer reference for Coronavax is led by Ms. Catherine Hughes who also chairs the Wesfarmers Centre of Vaccines and Infectious Diseases Community Reference Group at Telethon Kids Institute. This Group consulted on Coronavax in September 2020. Consumer involvement in Coronavax has sought to ensure the appropriateness and sensitivity of research questions and recruitment and to ensure that the research priorities and design reflect the needs of the community. Ishar Multicultural Women's Health Services, one of the largest multicultural health providers in Western Australia, provided additional consultation for this CALD study, as well as aiding with the recruitment of their clients, providing facilities, and hosting the focus groups.", "In the first 2 years of the COVID‐19 pandemic, WA experienced just over 1100 cases in a population of approximately 2.6 million people,\n13\n with transmission rapidly increasing only from February 2022.\n13\n As of March 2022, over 95% of people aged 12 years and older in WA had received at least two COVID‐19 vaccine doses,\n14\n and over 85.5% of those eligible for their third dose in WA having received it.\n15\n However, coverage rates at the time of our study were lower, as Australia had faced significant supply problems early in the rollout.\n16\n Responsibility for vaccine operations sat with both State and Federal Governments. WA residents access COVID‐19 vaccines through general practice clinics, federal respiratory clinics, pharmacies, community health services and state‐led mass vaccination clinics.\nAs governments sought to rapidly increase vaccine uptake across the entire population, populations requiring more complex and bespoke interventions, such as CALD communities, remained under‐served. For example, Local Government Areas (LGAs) in Perth with the highest proportion of those born in nonmain English‐speaking countries had some of the lowest uptake in Perth 8 months into the rollout.\n17\n A more tailored approach to CALD communities commenced in late 2021 after our study, including pop‐up vaccination clinics at major religious events (e.g., the Indian festival of Diwali, November 2021), or at multicultural community centres. The state‐wide campaign ‘Roll Up For WA’ translated resources into over 50 non‐English languages,\n18\n although at the time of our study only approximately 20 were in place.\nCALD individuals comprise almost one‐fifth of the WA population.\n19\n In WA, 90% of individuals born in what the Australian Bureau of Statistics calls ‘non‐main English‐speaking countries’ reside in the Perth metropolitan region,\n19\n so we focused on recruiting CALD adults living in Perth.", "Interviewees were recruited through CALD organizations, social media (including a specific campaign directed towards CALD men), and promotional campaigns (in English) that ran from August to October 2021. Interviewees voluntarily signed up via the secure web‐based survey platform REDCap hosted by Telethon Kids Institute.\n20\n, \n21\n Here, they provided demographic and contact information. They were selected if they met the CALD definition and then contacted up to three times each via phone or email to organize the interview. The CALD definition was based on the Australian Bureau of Statistics' definition,\n22\n and included individuals born overseas, having one or both parents born overseas from non‐English speaking countries, or whose first language is a Language Other Than English (LOTE). Interviews were conducted in English by SC over the phone or via teleconferencing software—interviewees who signed up all had sufficient English proficiency as the self‐sign‐up process was only in English.\nDue to the high levels of English proficiency required for interview sign‐ups, we approached Ishar to partner on this research so that we could engage with CALD individuals who had lower English proficiency. Accordingly, focus group participants were recruited separately through their CALD community centre for women based in Stirling, the LGA with the highest proportion of individuals born in nonmain English‐speaking countries.\n19\n Women attending regular English classes were invited face‐to‐face by researchers and their teachers to participate in a focus group in lieu of their class. Focus groups were run face‐to‐face by S. J. C., G. E. and K. A. in the CALD community centre; they were conducted in English with the use of peer translators for participants with low English proficiency.\nData were collected between August and October 2021. Of the 23 CALD individuals who voluntarily signed up online to participate in the individual interviews, we interviewed 11, with the remaining 12 being lost to follow‐up. We ran three focus groups at the Ishar multicultural centre with their female clients, comprised of 6–16 participants per group (total N = 37). All women who were present consented and participated in a focus group—there were no refusals. Interviews and focus groups lasted approximately 60 min. Of the total 48 participants, 90% were female and 29% were aged 30–39 years. Approximately, one third (35%) had been to university, 54% self‐reported high English proficiency and 27% reported having received at least their first COVID‐19 vaccine (Table 1). Further, 46% had migrated to Australia since 2010, 35% were born in Southern Asia, and 75% identified with a religion (most common being Islam and Christianity). Two of the 11 interviewees self‐identified as community leaders. Interviewees gave voluntary, informed, written consent; focus group participants gave voluntary, informed, verbal consent. All have been assigned pseudonyms.\nSociodemographic characteristics of the 48 CALD participants, Perth, WA\nAbbreviations: CALD, Culturally and Linguistically Diverse; TAFE, Technical and Further Education; WA, Western Australia.\nSome participants spoke ≥1 language.\nOther languages include Acholi, Azeri, Bomwali, Hazaragi, Hindi, Ilocano, Indonesian, Italian, Kikuyu (Bantu), Korean, Macedonian, Malay, Oromo, Somali, Tedim, Urdu and Vietnamese.", "Interview guides were developed by the multidisciplinary (project) team comprised of vaccination, medical, policy, social science and government scholars, and an indicative guide is published open‐access with our protocol.\n12\n The questions were simplified for the focus groups.", "All interviews and focus groups were audio‐recorded and professionally transcribed verbatim. Thematic analysis followed the Braun and Clarke method,\n23\n with deductive analysis in NVivo 12 using the Capability, Opportunity, Motivation, Behaviour (COM‐B) model.\n24\n We selected COM‐B as it is used extensively by the World Health Organization to understand reasons for (non)vaccination among specific populations.\n25\n G. E. generated initial codes, S. J. C. and B. N. each coded a manuscript to provide additional perspectives, including a CALD lived experience lens, and the three coders revised the matrix via a collaborative meeting before GC applied it to the full data set.", "Drawing on data from our interviews and focus groups, we created 10 subthemes encompassed within the three superordinate themes of Capability, Opportunity and Motivation (Figure 1).\nThe Capability, Opportunity and Motivation influencing COVID‐19 vaccine uptake among Culturally and Linguistically Diverse individuals in Perth, WA. Figure modified from COM‐B model\n24\n; arrows indicate influence of factor on other factors.\n[SUBTITLE] Capability [SUBSECTION] [SUBTITLE] Knowledge about COVID‐19 and COVID‐19 vaccination [SUBSECTION] Most participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nMost participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n[SUBTITLE] Knowledge about how and where to get vaccinated [SUBSECTION] Though many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\nThough many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n[SUBTITLE] Knowledge about COVID‐19 and COVID‐19 vaccination [SUBSECTION] Most participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nMost participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n[SUBTITLE] Knowledge about how and where to get vaccinated [SUBSECTION] Though many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\nThough many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n[SUBTITLE] Opportunity [SUBSECTION] [SUBTITLE] Relationships with general practitioners (GPs) [SUBSECTION] Many participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\nMany participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n[SUBTITLE] Access to information/misinformation [SUBSECTION] Participants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.\nParticipants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.\n[SUBTITLE] Access to vaccine clinics [SUBSECTION] Locations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.\nLocations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.\n[SUBTITLE] Community influence [SUBSECTION] Participants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n[SUBTITLE] Relationships with general practitioners (GPs) [SUBSECTION] Many participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\nMany participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n[SUBTITLE] Access to information/misinformation [SUBSECTION] Participants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.\nParticipants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.\n[SUBTITLE] Access to vaccine clinics [SUBSECTION] Locations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.\nLocations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.\n[SUBTITLE] Community influence [SUBSECTION] Participants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n[SUBTITLE] Motivation [SUBSECTION] Most participants were “scared”, “confused” or “unsure” about COVID‐19 vaccines, particularly because, as Umme said in a focus group “this medicine only come in 2 years, it come quickly, why so quickly, maybe something wrong”. Yet despite these concerns, many were nevertheless willing to be vaccinated to protect themselves, their family and their community.I'm getting this so that others around me don't get sick. I'm getting this so that I can protect my mum, I can protect my dad, I can protect my friends—(Angelo [Male, 37 years]; Interview)\n\nI'm getting this so that others around me don't get sick. I'm getting this so that I can protect my mum, I can protect my dad, I can protect my friends—(Angelo [Male, 37 years]; Interview)\nParticipants, aware that “Corona is coming”, felt they would be at greater risk once international and interstate borders opened, declaring that they would get vaccinated once WA borders opened (but likely not before). Others said the border opening would mean they could visit their friends and family overseas; they believed COVID‐19 vaccination would be a requirement for this:I want to go visit my family [in Ethiopia] … But [also] I'm happy that we are closed here. Because then we … stay safe—(Mahsa [Female, 34 years]; Focus Group)\n\nI want to go visit my family [in Ethiopia] … But [also] I'm happy that we are closed here. Because then we … stay safe—(Mahsa [Female, 34 years]; Focus Group)\nMost participants were “scared”, “confused” or “unsure” about COVID‐19 vaccines, particularly because, as Umme said in a focus group “this medicine only come in 2 years, it come quickly, why so quickly, maybe something wrong”. Yet despite these concerns, many were nevertheless willing to be vaccinated to protect themselves, their family and their community.I'm getting this so that others around me don't get sick. I'm getting this so that I can protect my mum, I can protect my dad, I can protect my friends—(Angelo [Male, 37 years]; Interview)\n\nI'm getting this so that others around me don't get sick. I'm getting this so that I can protect my mum, I can protect my dad, I can protect my friends—(Angelo [Male, 37 years]; Interview)\nParticipants, aware that “Corona is coming”, felt they would be at greater risk once international and interstate borders opened, declaring that they would get vaccinated once WA borders opened (but likely not before). Others said the border opening would mean they could visit their friends and family overseas; they believed COVID‐19 vaccination would be a requirement for this:I want to go visit my family [in Ethiopia] … But [also] I'm happy that we are closed here. Because then we … stay safe—(Mahsa [Female, 34 years]; Focus Group)\n\nI want to go visit my family [in Ethiopia] … But [also] I'm happy that we are closed here. Because then we … stay safe—(Mahsa [Female, 34 years]; Focus Group)", "[SUBTITLE] Knowledge about COVID‐19 and COVID‐19 vaccination [SUBSECTION] Most participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nMost participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n[SUBTITLE] Knowledge about how and where to get vaccinated [SUBSECTION] Though many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\nThough many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)", "Most participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])", "Though many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)", "[SUBTITLE] Relationships with general practitioners (GPs) [SUBSECTION] Many participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\nMany participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n[SUBTITLE] Access to information/misinformation [SUBSECTION] Participants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.\nParticipants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.\n[SUBTITLE] Access to vaccine clinics [SUBSECTION] Locations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.\nLocations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.\n[SUBTITLE] Community influence [SUBSECTION] Participants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)", "Many participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)", "Participants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.", "Locations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.", "Participants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)", "Most participants were “scared”, “confused” or “unsure” about COVID‐19 vaccines, particularly because, as Umme said in a focus group “this medicine only come in 2 years, it come quickly, why so quickly, maybe something wrong”. Yet despite these concerns, many were nevertheless willing to be vaccinated to protect themselves, their family and their community.I'm getting this so that others around me don't get sick. I'm getting this so that I can protect my mum, I can protect my dad, I can protect my friends—(Angelo [Male, 37 years]; Interview)\n\nI'm getting this so that others around me don't get sick. I'm getting this so that I can protect my mum, I can protect my dad, I can protect my friends—(Angelo [Male, 37 years]; Interview)\nParticipants, aware that “Corona is coming”, felt they would be at greater risk once international and interstate borders opened, declaring that they would get vaccinated once WA borders opened (but likely not before). Others said the border opening would mean they could visit their friends and family overseas; they believed COVID‐19 vaccination would be a requirement for this:I want to go visit my family [in Ethiopia] … But [also] I'm happy that we are closed here. Because then we … stay safe—(Mahsa [Female, 34 years]; Focus Group)\n\nI want to go visit my family [in Ethiopia] … But [also] I'm happy that we are closed here. Because then we … stay safe—(Mahsa [Female, 34 years]; Focus Group)", "A strong motivation for this research was to assist in strategic design, informing ongoing and future implementation of public health initiatives among CALD populations. Our experiences engaging with our CALD participants and their partners, including helping them to book appointments after the focus groups, emphasized how important it is that governments promptly address vaccine access issues, as well as campaigning and working directly with CALD groups, and featuring them visibly in messaging. The barriers regarding lack of access to resources and the circulation of misinformation must be addressed as soon as possible to reduce the COVID‐19 burden amongst the CALD population in WA. These lessons can then be utilized for future vaccine doses, for reaching the children of CALD populations, and for wider public health initiatives.\nAt the time of our study, CALD participants and their communities were not publicly sharing their vaccinated status and proudly promoting COVID‐19 vaccinations. However, it is unreasonable to demand more social courage from CALD individuals when what they need is more support. Given that governments are responsible for making vaccination accessible and acceptable to improve and sustain uptake,\n34\n they must recognize, work with, and train influential community leaders on the importance of and how to have meaningful vaccine conversations with their peers.\n32\n A recent review identified the who, the what and the how for effective community engagement during a pandemic: essentially, it is important to engage with the community from the very beginning for a better chance of behaviour change within the community.\n35\n In eastern Australia, Mahimbo et al.\n9\n identified that CALD communities are eager for community engagement, and for their leaders to champion the vaccine. Seale et al.\n29\n identified that while some CALD organizations in Australia were updating local CALD leaders on relevant COVID‐19 information to share with their community, there was a lack of training in how to do so. Training influential community leaders can occur over a relatively short period of time: a COVID‐19 information and support programme was offered in the United States to CALD community leaders which involved 1‐hour conference calls twice a week (we acknowledge that this is then reliant on the leaders having a decent level of computer literacy.)\n29\n Further, given the trust and respect our participants held for their health practitioners, it is imperative that governments support GPs to effectively communicate about COVID‐19 vaccination with CALD patients; this requires resources, training and remuneration.\n36\n Since our data collection, the Australian National Centre for Immunisation Research and Surveillance have published a COVID‐19 decision aid in Greek, Vietnamese, Arabic, Simplified Chinese and Traditional Chinese.\n37\n Immunisation providers could use or share this tool with their CALD patients (though its effectiveness in increasing vaccine uptake is yet to be evaluated).\nOur participants' high levels of trust in governing authorities is an untapped opportunity. WA participants knew that ‘Corona is coming’. Now that it has arrived, the government and healthcare providers must maintain the trust of CALD by reaching them promptly with vaccine doses and culturally appropriate information to reduce the burden of COVID‐19. If these measures fail, it would be entirely reasonable for CALD individuals to not blame their own choices, and instead look to the authorities they believed would protect them and ask why they were not offered greater support and access.\n[SUBTITLE] Limitations [SUBSECTION] Qualitative data is not widely generalizable, as its function is to generate deep insights when little is known about an issue, or when data cannot be captured appropriately via quantitative means, including across language barriers. Different Australian states had different pandemic experiences and our data may not be applicable to other states or countries, although our insights and reflections remain pertinent. Our interviews and focus groups took place well into the COVID‐19 pandemic and vaccine rollout, but before community transmission in WA—the subsequent ‘first wave’ from February 2022 may have changed participants' views. Our recruitment method for interviews skewed demographics towards those with high English proficiency, females (despite a social media campaign specifically recruiting men), high levels of education and high COVID‐19 vaccine uptake. However, we mitigated this by collaborating with a community centre for recent migrants/refugees to undertake focus groups with women possessing low levels of English proficiency, education, and COVID‐19 vaccine uptake. Participants' language constraints were mitigated by strong peer relationships that facilitated peer translation. Despite our efforts to mitigate these limitations, our study does not represent the entire CALD community in Perth.\nQualitative data is not widely generalizable, as its function is to generate deep insights when little is known about an issue, or when data cannot be captured appropriately via quantitative means, including across language barriers. Different Australian states had different pandemic experiences and our data may not be applicable to other states or countries, although our insights and reflections remain pertinent. Our interviews and focus groups took place well into the COVID‐19 pandemic and vaccine rollout, but before community transmission in WA—the subsequent ‘first wave’ from February 2022 may have changed participants' views. Our recruitment method for interviews skewed demographics towards those with high English proficiency, females (despite a social media campaign specifically recruiting men), high levels of education and high COVID‐19 vaccine uptake. However, we mitigated this by collaborating with a community centre for recent migrants/refugees to undertake focus groups with women possessing low levels of English proficiency, education, and COVID‐19 vaccine uptake. Participants' language constraints were mitigated by strong peer relationships that facilitated peer translation. Despite our efforts to mitigate these limitations, our study does not represent the entire CALD community in Perth.", "Qualitative data is not widely generalizable, as its function is to generate deep insights when little is known about an issue, or when data cannot be captured appropriately via quantitative means, including across language barriers. Different Australian states had different pandemic experiences and our data may not be applicable to other states or countries, although our insights and reflections remain pertinent. Our interviews and focus groups took place well into the COVID‐19 pandemic and vaccine rollout, but before community transmission in WA—the subsequent ‘first wave’ from February 2022 may have changed participants' views. Our recruitment method for interviews skewed demographics towards those with high English proficiency, females (despite a social media campaign specifically recruiting men), high levels of education and high COVID‐19 vaccine uptake. However, we mitigated this by collaborating with a community centre for recent migrants/refugees to undertake focus groups with women possessing low levels of English proficiency, education, and COVID‐19 vaccine uptake. Participants' language constraints were mitigated by strong peer relationships that facilitated peer translation. Despite our efforts to mitigate these limitations, our study does not represent the entire CALD community in Perth.", "Samantha J. Carlson supervised the data collection and analysis, led the initial manuscript drafting, contributed to the study design, contributed to project administration and coordinated the ethics and stakeholder engagement. Gracie Edwards collected the data and conducted the analysis as well as drafted the literature review and contributed to the overall manuscript drafting. Christopher C. Blyth contributed to the conceptualization of the project and provided oversight and input into the study and funding attainment, and reviewed and edited the manuscript. Barbara Nattabi supervised the data collection and analysis, contributed to the coding framework, provided CALD expertise, contributed to project administration and reviewed and edited the manuscript. Katie Attwell conceptualized the project, co‐developed the broader study methodology with Christopher C. Blyth, led the funding attainment, and supervised the conduct and administration of the broader research project. She contributed to the literature review and the drafting, reviewing and editing of the manuscript. All authors have approved the final manuscript." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "METHODS", "Study design", "Consumer consultation process", "Study setting", "Sampling, recruitment and data collection", "Interview guide", "Analysis", "FINDINGS", "Capability", "Knowledge about COVID‐19 and COVID‐19 vaccination", "Knowledge about how and where to get vaccinated", "Opportunity", "Relationships with general practitioners (GPs)", "Access to information/misinformation", "Access to vaccine clinics", "Community influence", "Motivation", "DISCUSSION", "IMPLICATIONS FOR POLICY AND PRACTICE", "Limitations", "CONCLUSION", "AUTHOR CONTRIBUTIONS", "CONFLICT OF INTEREST" ]
[ "Scholars and commentators recognize that even within high‐income countries, pockets of populations are at risk of being left behind in the globally inequitable COVID‐19 vaccine rollout.\n1\n Culturally and Linguistically Diverse (CALD) populations, which include refugees and migrants, are at particular risk due to inaccessible or suboptimal healthcare experiences, which may compound attitudinal factors, including vaccine hesitancy.\n2\n, \n3\n Unique local factors can also create barriers: in the case of Western Australia (WA), almost nonexistent levels of community transmission of COVID‐19  led some locals to ‘wait awhile’ before being vaccinated as state borders remained closed throughout 2021.\n4\n Meanwhile, earlier large outbreaks in other Australian states disproportionately affected CALD communities, with significantly higher morbidity and mortality.\n5\n, \n6\n\n\nIn Australia, COVID‐19 vaccination is recorded on the Australian Immunisation Register, however, country of birth and language spoken at home are not captured on this register. Therefore, there is no publicly available data on uptake among CALD communities in Australia. However, an online survey of refugees in Australia in mid‐2021 identified that 88% had not yet received a COVID‐19 vaccine.\n7\n Emerging evidence suggests that COVID‐19 vaccine hesitancy is prevalent among Australian CALD populations.\n8\n, \n9\n Focus groups conducted in mid‐2021 in New South Wales with CALD people indicated that 42% of participants were not planning to be vaccinated, and 29% were unsure or hesitant.\n8\n Many had safety concerns, were uncertain how vaccines work or lacked trust in government.\n8\n These uncertainties are unsurprising, given little of the public health information on COVID‐19 in Australia was tailored towards those with low English proficiency or health literacy.\n10\n Lockyer et al.'s\n11\n qualitative study in multicultural Bradford, UK, also found that participants' trust in government and the National Health Service dropped during the pandemic and that vaccine hesitancy was expressed by ethnic or national minorities.\nIn this study, we sought to understand the perceptions and attitudes to COVID‐19 vaccines amongst WA's CALD communities, to identify barriers preventing uptake, and to consider strategies to promote uptake in this population. Findings can support the development of strategies to enhance uptake among the CALD communities locally, nationwide and in comparable high‐income English‐speaking countries. They are particularly pertinent in the context of needing to enhance third and fourth‐dose rates for adults and protecting CALD children with paediatric COVID‐19 vaccinations.", "[SUBTITLE] Study design [SUBSECTION] This CALD study was conducted as part of the larger mixed methods research project, Coronavax. Coronavax was initiated in April 2020 with the aim of ascertaining community attitudes towards vaccination and vaccine access in WA through a series of qualitative interviews with segmented population groups. A key aspect of the project involved translating findings to the state and federal governments in real‐time via Functional Dialogues, a novel formal research exchange mechanism in which data on community attitudes are presented to government officials, who then contribute their own reflections and experiences as additional research data (not reported here).\n12\n Coronavax received ethics approval from the Child and Adolescent Services (CAHS) Human Research Ethics Committee (RGS0000004457).\nThis CALD study was conducted as part of the larger mixed methods research project, Coronavax. Coronavax was initiated in April 2020 with the aim of ascertaining community attitudes towards vaccination and vaccine access in WA through a series of qualitative interviews with segmented population groups. A key aspect of the project involved translating findings to the state and federal governments in real‐time via Functional Dialogues, a novel formal research exchange mechanism in which data on community attitudes are presented to government officials, who then contribute their own reflections and experiences as additional research data (not reported here).\n12\n Coronavax received ethics approval from the Child and Adolescent Services (CAHS) Human Research Ethics Committee (RGS0000004457).\n[SUBTITLE] Consumer consultation process [SUBSECTION] Consumer reference for Coronavax is led by Ms. Catherine Hughes who also chairs the Wesfarmers Centre of Vaccines and Infectious Diseases Community Reference Group at Telethon Kids Institute. This Group consulted on Coronavax in September 2020. Consumer involvement in Coronavax has sought to ensure the appropriateness and sensitivity of research questions and recruitment and to ensure that the research priorities and design reflect the needs of the community. Ishar Multicultural Women's Health Services, one of the largest multicultural health providers in Western Australia, provided additional consultation for this CALD study, as well as aiding with the recruitment of their clients, providing facilities, and hosting the focus groups.\nConsumer reference for Coronavax is led by Ms. Catherine Hughes who also chairs the Wesfarmers Centre of Vaccines and Infectious Diseases Community Reference Group at Telethon Kids Institute. This Group consulted on Coronavax in September 2020. Consumer involvement in Coronavax has sought to ensure the appropriateness and sensitivity of research questions and recruitment and to ensure that the research priorities and design reflect the needs of the community. Ishar Multicultural Women's Health Services, one of the largest multicultural health providers in Western Australia, provided additional consultation for this CALD study, as well as aiding with the recruitment of their clients, providing facilities, and hosting the focus groups.\n[SUBTITLE] Study setting [SUBSECTION] In the first 2 years of the COVID‐19 pandemic, WA experienced just over 1100 cases in a population of approximately 2.6 million people,\n13\n with transmission rapidly increasing only from February 2022.\n13\n As of March 2022, over 95% of people aged 12 years and older in WA had received at least two COVID‐19 vaccine doses,\n14\n and over 85.5% of those eligible for their third dose in WA having received it.\n15\n However, coverage rates at the time of our study were lower, as Australia had faced significant supply problems early in the rollout.\n16\n Responsibility for vaccine operations sat with both State and Federal Governments. WA residents access COVID‐19 vaccines through general practice clinics, federal respiratory clinics, pharmacies, community health services and state‐led mass vaccination clinics.\nAs governments sought to rapidly increase vaccine uptake across the entire population, populations requiring more complex and bespoke interventions, such as CALD communities, remained under‐served. For example, Local Government Areas (LGAs) in Perth with the highest proportion of those born in nonmain English‐speaking countries had some of the lowest uptake in Perth 8 months into the rollout.\n17\n A more tailored approach to CALD communities commenced in late 2021 after our study, including pop‐up vaccination clinics at major religious events (e.g., the Indian festival of Diwali, November 2021), or at multicultural community centres. The state‐wide campaign ‘Roll Up For WA’ translated resources into over 50 non‐English languages,\n18\n although at the time of our study only approximately 20 were in place.\nCALD individuals comprise almost one‐fifth of the WA population.\n19\n In WA, 90% of individuals born in what the Australian Bureau of Statistics calls ‘non‐main English‐speaking countries’ reside in the Perth metropolitan region,\n19\n so we focused on recruiting CALD adults living in Perth.\nIn the first 2 years of the COVID‐19 pandemic, WA experienced just over 1100 cases in a population of approximately 2.6 million people,\n13\n with transmission rapidly increasing only from February 2022.\n13\n As of March 2022, over 95% of people aged 12 years and older in WA had received at least two COVID‐19 vaccine doses,\n14\n and over 85.5% of those eligible for their third dose in WA having received it.\n15\n However, coverage rates at the time of our study were lower, as Australia had faced significant supply problems early in the rollout.\n16\n Responsibility for vaccine operations sat with both State and Federal Governments. WA residents access COVID‐19 vaccines through general practice clinics, federal respiratory clinics, pharmacies, community health services and state‐led mass vaccination clinics.\nAs governments sought to rapidly increase vaccine uptake across the entire population, populations requiring more complex and bespoke interventions, such as CALD communities, remained under‐served. For example, Local Government Areas (LGAs) in Perth with the highest proportion of those born in nonmain English‐speaking countries had some of the lowest uptake in Perth 8 months into the rollout.\n17\n A more tailored approach to CALD communities commenced in late 2021 after our study, including pop‐up vaccination clinics at major religious events (e.g., the Indian festival of Diwali, November 2021), or at multicultural community centres. The state‐wide campaign ‘Roll Up For WA’ translated resources into over 50 non‐English languages,\n18\n although at the time of our study only approximately 20 were in place.\nCALD individuals comprise almost one‐fifth of the WA population.\n19\n In WA, 90% of individuals born in what the Australian Bureau of Statistics calls ‘non‐main English‐speaking countries’ reside in the Perth metropolitan region,\n19\n so we focused on recruiting CALD adults living in Perth.\n[SUBTITLE] Sampling, recruitment and data collection [SUBSECTION] Interviewees were recruited through CALD organizations, social media (including a specific campaign directed towards CALD men), and promotional campaigns (in English) that ran from August to October 2021. Interviewees voluntarily signed up via the secure web‐based survey platform REDCap hosted by Telethon Kids Institute.\n20\n, \n21\n Here, they provided demographic and contact information. They were selected if they met the CALD definition and then contacted up to three times each via phone or email to organize the interview. The CALD definition was based on the Australian Bureau of Statistics' definition,\n22\n and included individuals born overseas, having one or both parents born overseas from non‐English speaking countries, or whose first language is a Language Other Than English (LOTE). Interviews were conducted in English by SC over the phone or via teleconferencing software—interviewees who signed up all had sufficient English proficiency as the self‐sign‐up process was only in English.\nDue to the high levels of English proficiency required for interview sign‐ups, we approached Ishar to partner on this research so that we could engage with CALD individuals who had lower English proficiency. Accordingly, focus group participants were recruited separately through their CALD community centre for women based in Stirling, the LGA with the highest proportion of individuals born in nonmain English‐speaking countries.\n19\n Women attending regular English classes were invited face‐to‐face by researchers and their teachers to participate in a focus group in lieu of their class. Focus groups were run face‐to‐face by S. J. C., G. E. and K. A. in the CALD community centre; they were conducted in English with the use of peer translators for participants with low English proficiency.\nData were collected between August and October 2021. Of the 23 CALD individuals who voluntarily signed up online to participate in the individual interviews, we interviewed 11, with the remaining 12 being lost to follow‐up. We ran three focus groups at the Ishar multicultural centre with their female clients, comprised of 6–16 participants per group (total N = 37). All women who were present consented and participated in a focus group—there were no refusals. Interviews and focus groups lasted approximately 60 min. Of the total 48 participants, 90% were female and 29% were aged 30–39 years. Approximately, one third (35%) had been to university, 54% self‐reported high English proficiency and 27% reported having received at least their first COVID‐19 vaccine (Table 1). Further, 46% had migrated to Australia since 2010, 35% were born in Southern Asia, and 75% identified with a religion (most common being Islam and Christianity). Two of the 11 interviewees self‐identified as community leaders. Interviewees gave voluntary, informed, written consent; focus group participants gave voluntary, informed, verbal consent. All have been assigned pseudonyms.\nSociodemographic characteristics of the 48 CALD participants, Perth, WA\nAbbreviations: CALD, Culturally and Linguistically Diverse; TAFE, Technical and Further Education; WA, Western Australia.\nSome participants spoke ≥1 language.\nOther languages include Acholi, Azeri, Bomwali, Hazaragi, Hindi, Ilocano, Indonesian, Italian, Kikuyu (Bantu), Korean, Macedonian, Malay, Oromo, Somali, Tedim, Urdu and Vietnamese.\nInterviewees were recruited through CALD organizations, social media (including a specific campaign directed towards CALD men), and promotional campaigns (in English) that ran from August to October 2021. Interviewees voluntarily signed up via the secure web‐based survey platform REDCap hosted by Telethon Kids Institute.\n20\n, \n21\n Here, they provided demographic and contact information. They were selected if they met the CALD definition and then contacted up to three times each via phone or email to organize the interview. The CALD definition was based on the Australian Bureau of Statistics' definition,\n22\n and included individuals born overseas, having one or both parents born overseas from non‐English speaking countries, or whose first language is a Language Other Than English (LOTE). Interviews were conducted in English by SC over the phone or via teleconferencing software—interviewees who signed up all had sufficient English proficiency as the self‐sign‐up process was only in English.\nDue to the high levels of English proficiency required for interview sign‐ups, we approached Ishar to partner on this research so that we could engage with CALD individuals who had lower English proficiency. Accordingly, focus group participants were recruited separately through their CALD community centre for women based in Stirling, the LGA with the highest proportion of individuals born in nonmain English‐speaking countries.\n19\n Women attending regular English classes were invited face‐to‐face by researchers and their teachers to participate in a focus group in lieu of their class. Focus groups were run face‐to‐face by S. J. C., G. E. and K. A. in the CALD community centre; they were conducted in English with the use of peer translators for participants with low English proficiency.\nData were collected between August and October 2021. Of the 23 CALD individuals who voluntarily signed up online to participate in the individual interviews, we interviewed 11, with the remaining 12 being lost to follow‐up. We ran three focus groups at the Ishar multicultural centre with their female clients, comprised of 6–16 participants per group (total N = 37). All women who were present consented and participated in a focus group—there were no refusals. Interviews and focus groups lasted approximately 60 min. Of the total 48 participants, 90% were female and 29% were aged 30–39 years. Approximately, one third (35%) had been to university, 54% self‐reported high English proficiency and 27% reported having received at least their first COVID‐19 vaccine (Table 1). Further, 46% had migrated to Australia since 2010, 35% were born in Southern Asia, and 75% identified with a religion (most common being Islam and Christianity). Two of the 11 interviewees self‐identified as community leaders. Interviewees gave voluntary, informed, written consent; focus group participants gave voluntary, informed, verbal consent. All have been assigned pseudonyms.\nSociodemographic characteristics of the 48 CALD participants, Perth, WA\nAbbreviations: CALD, Culturally and Linguistically Diverse; TAFE, Technical and Further Education; WA, Western Australia.\nSome participants spoke ≥1 language.\nOther languages include Acholi, Azeri, Bomwali, Hazaragi, Hindi, Ilocano, Indonesian, Italian, Kikuyu (Bantu), Korean, Macedonian, Malay, Oromo, Somali, Tedim, Urdu and Vietnamese.\n[SUBTITLE] Interview guide [SUBSECTION] Interview guides were developed by the multidisciplinary (project) team comprised of vaccination, medical, policy, social science and government scholars, and an indicative guide is published open‐access with our protocol.\n12\n The questions were simplified for the focus groups.\nInterview guides were developed by the multidisciplinary (project) team comprised of vaccination, medical, policy, social science and government scholars, and an indicative guide is published open‐access with our protocol.\n12\n The questions were simplified for the focus groups.\n[SUBTITLE] Analysis [SUBSECTION] All interviews and focus groups were audio‐recorded and professionally transcribed verbatim. Thematic analysis followed the Braun and Clarke method,\n23\n with deductive analysis in NVivo 12 using the Capability, Opportunity, Motivation, Behaviour (COM‐B) model.\n24\n We selected COM‐B as it is used extensively by the World Health Organization to understand reasons for (non)vaccination among specific populations.\n25\n G. E. generated initial codes, S. J. C. and B. N. each coded a manuscript to provide additional perspectives, including a CALD lived experience lens, and the three coders revised the matrix via a collaborative meeting before GC applied it to the full data set.\nAll interviews and focus groups were audio‐recorded and professionally transcribed verbatim. Thematic analysis followed the Braun and Clarke method,\n23\n with deductive analysis in NVivo 12 using the Capability, Opportunity, Motivation, Behaviour (COM‐B) model.\n24\n We selected COM‐B as it is used extensively by the World Health Organization to understand reasons for (non)vaccination among specific populations.\n25\n G. E. generated initial codes, S. J. C. and B. N. each coded a manuscript to provide additional perspectives, including a CALD lived experience lens, and the three coders revised the matrix via a collaborative meeting before GC applied it to the full data set.", "This CALD study was conducted as part of the larger mixed methods research project, Coronavax. Coronavax was initiated in April 2020 with the aim of ascertaining community attitudes towards vaccination and vaccine access in WA through a series of qualitative interviews with segmented population groups. A key aspect of the project involved translating findings to the state and federal governments in real‐time via Functional Dialogues, a novel formal research exchange mechanism in which data on community attitudes are presented to government officials, who then contribute their own reflections and experiences as additional research data (not reported here).\n12\n Coronavax received ethics approval from the Child and Adolescent Services (CAHS) Human Research Ethics Committee (RGS0000004457).", "Consumer reference for Coronavax is led by Ms. Catherine Hughes who also chairs the Wesfarmers Centre of Vaccines and Infectious Diseases Community Reference Group at Telethon Kids Institute. This Group consulted on Coronavax in September 2020. Consumer involvement in Coronavax has sought to ensure the appropriateness and sensitivity of research questions and recruitment and to ensure that the research priorities and design reflect the needs of the community. Ishar Multicultural Women's Health Services, one of the largest multicultural health providers in Western Australia, provided additional consultation for this CALD study, as well as aiding with the recruitment of their clients, providing facilities, and hosting the focus groups.", "In the first 2 years of the COVID‐19 pandemic, WA experienced just over 1100 cases in a population of approximately 2.6 million people,\n13\n with transmission rapidly increasing only from February 2022.\n13\n As of March 2022, over 95% of people aged 12 years and older in WA had received at least two COVID‐19 vaccine doses,\n14\n and over 85.5% of those eligible for their third dose in WA having received it.\n15\n However, coverage rates at the time of our study were lower, as Australia had faced significant supply problems early in the rollout.\n16\n Responsibility for vaccine operations sat with both State and Federal Governments. WA residents access COVID‐19 vaccines through general practice clinics, federal respiratory clinics, pharmacies, community health services and state‐led mass vaccination clinics.\nAs governments sought to rapidly increase vaccine uptake across the entire population, populations requiring more complex and bespoke interventions, such as CALD communities, remained under‐served. For example, Local Government Areas (LGAs) in Perth with the highest proportion of those born in nonmain English‐speaking countries had some of the lowest uptake in Perth 8 months into the rollout.\n17\n A more tailored approach to CALD communities commenced in late 2021 after our study, including pop‐up vaccination clinics at major religious events (e.g., the Indian festival of Diwali, November 2021), or at multicultural community centres. The state‐wide campaign ‘Roll Up For WA’ translated resources into over 50 non‐English languages,\n18\n although at the time of our study only approximately 20 were in place.\nCALD individuals comprise almost one‐fifth of the WA population.\n19\n In WA, 90% of individuals born in what the Australian Bureau of Statistics calls ‘non‐main English‐speaking countries’ reside in the Perth metropolitan region,\n19\n so we focused on recruiting CALD adults living in Perth.", "Interviewees were recruited through CALD organizations, social media (including a specific campaign directed towards CALD men), and promotional campaigns (in English) that ran from August to October 2021. Interviewees voluntarily signed up via the secure web‐based survey platform REDCap hosted by Telethon Kids Institute.\n20\n, \n21\n Here, they provided demographic and contact information. They were selected if they met the CALD definition and then contacted up to three times each via phone or email to organize the interview. The CALD definition was based on the Australian Bureau of Statistics' definition,\n22\n and included individuals born overseas, having one or both parents born overseas from non‐English speaking countries, or whose first language is a Language Other Than English (LOTE). Interviews were conducted in English by SC over the phone or via teleconferencing software—interviewees who signed up all had sufficient English proficiency as the self‐sign‐up process was only in English.\nDue to the high levels of English proficiency required for interview sign‐ups, we approached Ishar to partner on this research so that we could engage with CALD individuals who had lower English proficiency. Accordingly, focus group participants were recruited separately through their CALD community centre for women based in Stirling, the LGA with the highest proportion of individuals born in nonmain English‐speaking countries.\n19\n Women attending regular English classes were invited face‐to‐face by researchers and their teachers to participate in a focus group in lieu of their class. Focus groups were run face‐to‐face by S. J. C., G. E. and K. A. in the CALD community centre; they were conducted in English with the use of peer translators for participants with low English proficiency.\nData were collected between August and October 2021. Of the 23 CALD individuals who voluntarily signed up online to participate in the individual interviews, we interviewed 11, with the remaining 12 being lost to follow‐up. We ran three focus groups at the Ishar multicultural centre with their female clients, comprised of 6–16 participants per group (total N = 37). All women who were present consented and participated in a focus group—there were no refusals. Interviews and focus groups lasted approximately 60 min. Of the total 48 participants, 90% were female and 29% were aged 30–39 years. Approximately, one third (35%) had been to university, 54% self‐reported high English proficiency and 27% reported having received at least their first COVID‐19 vaccine (Table 1). Further, 46% had migrated to Australia since 2010, 35% were born in Southern Asia, and 75% identified with a religion (most common being Islam and Christianity). Two of the 11 interviewees self‐identified as community leaders. Interviewees gave voluntary, informed, written consent; focus group participants gave voluntary, informed, verbal consent. All have been assigned pseudonyms.\nSociodemographic characteristics of the 48 CALD participants, Perth, WA\nAbbreviations: CALD, Culturally and Linguistically Diverse; TAFE, Technical and Further Education; WA, Western Australia.\nSome participants spoke ≥1 language.\nOther languages include Acholi, Azeri, Bomwali, Hazaragi, Hindi, Ilocano, Indonesian, Italian, Kikuyu (Bantu), Korean, Macedonian, Malay, Oromo, Somali, Tedim, Urdu and Vietnamese.", "Interview guides were developed by the multidisciplinary (project) team comprised of vaccination, medical, policy, social science and government scholars, and an indicative guide is published open‐access with our protocol.\n12\n The questions were simplified for the focus groups.", "All interviews and focus groups were audio‐recorded and professionally transcribed verbatim. Thematic analysis followed the Braun and Clarke method,\n23\n with deductive analysis in NVivo 12 using the Capability, Opportunity, Motivation, Behaviour (COM‐B) model.\n24\n We selected COM‐B as it is used extensively by the World Health Organization to understand reasons for (non)vaccination among specific populations.\n25\n G. E. generated initial codes, S. J. C. and B. N. each coded a manuscript to provide additional perspectives, including a CALD lived experience lens, and the three coders revised the matrix via a collaborative meeting before GC applied it to the full data set.", "Drawing on data from our interviews and focus groups, we created 10 subthemes encompassed within the three superordinate themes of Capability, Opportunity and Motivation (Figure 1).\nThe Capability, Opportunity and Motivation influencing COVID‐19 vaccine uptake among Culturally and Linguistically Diverse individuals in Perth, WA. Figure modified from COM‐B model\n24\n; arrows indicate influence of factor on other factors.\n[SUBTITLE] Capability [SUBSECTION] [SUBTITLE] Knowledge about COVID‐19 and COVID‐19 vaccination [SUBSECTION] Most participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nMost participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n[SUBTITLE] Knowledge about how and where to get vaccinated [SUBSECTION] Though many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\nThough many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n[SUBTITLE] Knowledge about COVID‐19 and COVID‐19 vaccination [SUBSECTION] Most participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nMost participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n[SUBTITLE] Knowledge about how and where to get vaccinated [SUBSECTION] Though many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\nThough many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n[SUBTITLE] Opportunity [SUBSECTION] [SUBTITLE] Relationships with general practitioners (GPs) [SUBSECTION] Many participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\nMany participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n[SUBTITLE] Access to information/misinformation [SUBSECTION] Participants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.\nParticipants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.\n[SUBTITLE] Access to vaccine clinics [SUBSECTION] Locations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.\nLocations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.\n[SUBTITLE] Community influence [SUBSECTION] Participants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n[SUBTITLE] Relationships with general practitioners (GPs) [SUBSECTION] Many participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\nMany participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n[SUBTITLE] Access to information/misinformation [SUBSECTION] Participants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.\nParticipants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.\n[SUBTITLE] Access to vaccine clinics [SUBSECTION] Locations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.\nLocations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.\n[SUBTITLE] Community influence [SUBSECTION] Participants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n[SUBTITLE] Motivation [SUBSECTION] Most participants were “scared”, “confused” or “unsure” about COVID‐19 vaccines, particularly because, as Umme said in a focus group “this medicine only come in 2 years, it come quickly, why so quickly, maybe something wrong”. Yet despite these concerns, many were nevertheless willing to be vaccinated to protect themselves, their family and their community.I'm getting this so that others around me don't get sick. I'm getting this so that I can protect my mum, I can protect my dad, I can protect my friends—(Angelo [Male, 37 years]; Interview)\n\nI'm getting this so that others around me don't get sick. I'm getting this so that I can protect my mum, I can protect my dad, I can protect my friends—(Angelo [Male, 37 years]; Interview)\nParticipants, aware that “Corona is coming”, felt they would be at greater risk once international and interstate borders opened, declaring that they would get vaccinated once WA borders opened (but likely not before). Others said the border opening would mean they could visit their friends and family overseas; they believed COVID‐19 vaccination would be a requirement for this:I want to go visit my family [in Ethiopia] … But [also] I'm happy that we are closed here. Because then we … stay safe—(Mahsa [Female, 34 years]; Focus Group)\n\nI want to go visit my family [in Ethiopia] … But [also] I'm happy that we are closed here. Because then we … stay safe—(Mahsa [Female, 34 years]; Focus Group)\nMost participants were “scared”, “confused” or “unsure” about COVID‐19 vaccines, particularly because, as Umme said in a focus group “this medicine only come in 2 years, it come quickly, why so quickly, maybe something wrong”. Yet despite these concerns, many were nevertheless willing to be vaccinated to protect themselves, their family and their community.I'm getting this so that others around me don't get sick. I'm getting this so that I can protect my mum, I can protect my dad, I can protect my friends—(Angelo [Male, 37 years]; Interview)\n\nI'm getting this so that others around me don't get sick. I'm getting this so that I can protect my mum, I can protect my dad, I can protect my friends—(Angelo [Male, 37 years]; Interview)\nParticipants, aware that “Corona is coming”, felt they would be at greater risk once international and interstate borders opened, declaring that they would get vaccinated once WA borders opened (but likely not before). Others said the border opening would mean they could visit their friends and family overseas; they believed COVID‐19 vaccination would be a requirement for this:I want to go visit my family [in Ethiopia] … But [also] I'm happy that we are closed here. Because then we … stay safe—(Mahsa [Female, 34 years]; Focus Group)\n\nI want to go visit my family [in Ethiopia] … But [also] I'm happy that we are closed here. Because then we … stay safe—(Mahsa [Female, 34 years]; Focus Group)", "[SUBTITLE] Knowledge about COVID‐19 and COVID‐19 vaccination [SUBSECTION] Most participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nMost participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n[SUBTITLE] Knowledge about how and where to get vaccinated [SUBSECTION] Though many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\nThough many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)", "Most participants lacked an understanding of the potential severity of COVID‐19 infection, transmission routes, and vaccine ingredients or efficacy. This was particularly expressed by female focus group participants who had low English proficiency and education levels. Zi (Female, age unknown) said:I want to know, if you have your vaccines, how you know that you're protected from the virus?\n\nI want to know, if you have your vaccines, how you know that you're protected from the virus?\nSome were also unsure whether the COVID‐19 vaccine brands being used in Australia would be accepted by their home country. Further, many with pre‐existing health conditions feared severe side effects from the vaccines:She has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\nShe has her two [adult] boys back in Sudan. She applied for them to come here 7–8 years ago, but they [the Australian government] didn't accept them in … and now she has so many health problem … she's scared if she take [the vaccine] … maybe something will happen to her [like death] and she will not see her [adult] kids … she doesn't know what is right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])", "Though many participants knew they could receive a vaccine at a mass vaccination clinic, few were aware they could be vaccinated at a general practice clinic. Many also did not know vaccination was free. Finally, some did not know how to navigate the online booking system; a few had tried, but with no success:Yes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)\n\nYes, I try yesterday to do online … doesn't work with me—(Ezma [Female, 40 years]; Focus Group)", "[SUBTITLE] Relationships with general practitioners (GPs) [SUBSECTION] Many participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\nMany participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n[SUBTITLE] Access to information/misinformation [SUBSECTION] Participants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.\nParticipants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.\n[SUBTITLE] Access to vaccine clinics [SUBSECTION] Locations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.\nLocations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.\n[SUBTITLE] Community influence [SUBSECTION] Participants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)", "Many participants had GPs who spoke their language and/or were CALD, leading to strong trust in their recommendations:If my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\n\nIf my doctor said [COVID‐19 vaccination] was good, I would get it—(Hana [Female, age unknown]; Focus Group)\nAlthough many participants were unvaccinated, the appeal of convenient, trustworthy, and familiar healthcare providers meant they would choose to be vaccinated at their general practice clinics rather than travelling to mass clinics. Participants also felt more comfortable asking their CALD GP questions about the vaccine, compared to unfamiliar providers who don't speak their language. They also trusted that their GPs “don't want to kill us”. However, not all participants had established comfortable relationships with GPs, and some were concerned that GPs lack the cultural awareness skills or resources to communicate with participants.…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n…some of our [CALD] community members have got their own GPs that they trust. Some [general practice clinics are] just like going to a factory—(Reyna [Female, 53 years]; Community Leader, Interview)", "Participants cited significant concerns about COVID‐19 vaccine safety, often informed by misinformation. They depicted their immersion within tight‐knit communities, sharing information at gatherings, informal community events and social interactions. Friends and contacts in countries of origin or in other destination countries also spread (mis)information, including through social media.…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she doesn't have any ideas about the vaccine. And she hearing so many, you know, gossip around … she doesn't know what is the right—(Sita; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n‘Gossip’ was centred on safety: people were discussing contracting COVID‐19 from the vaccine, or extreme vaccine side effects including infertility, memory, cognition issues and death occurring specifically 2 years after vaccine receipt:…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\n\n…almost everyone, they talking about, like, you know, the injection for the COVID, if they take it or not. And everyone said, ‘Don't take it! You're going to die! You're going to die!’—(Ezma [Female, 40 years]; Focus Group)\nCALD individuals wanted to protect themselves against COVID‐19, but not always through vaccination, given these safety fears. Alternatives to vaccination—seen as being risk‐free—were actively shared among community members:My mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\n\nMy mum's side [of the family] started sending … WhatsApp messages … ‘Oh, if you take an onion and you cut it in half … leave it in a corner of your house, all the coronavirus is going to go to the onion and you'll be safe’—(Inda [Female, 23 years]; Interview)\nSome participants relied on WA government online resources and mainstream media (predominantly broadcast in English only) such as Channels Seven, Nine and the Australian Broadcasting Corporation (ABC). Few were aware of or consumed media in their language distributed by the Special Broadcasting Service (SBS), the main multilingual broadcaster in Australia. Most participants with no/low English proficiency relied heavily on social media (mostly Facebook or WhatsApp) and sourced information in their preferred language, shared by internationally residing relatives or their local CALD community.Even though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\n\nEven though my mum can read Chinese … she wouldn't be actively going to websites to check, and she would be relying on her daughters or friends to tell her what's happening—(Liling [Female, 50 years]; Interview)\nSome participants spoke multiple languages but still expressed a preference to consume vaccination information in only one of them—that being their first language.\nSome participants sought accurate information from their doctors; however, their vaccine‐related fears were not always allayed.…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\n\n…she went to the doctor but … the doctor just want to give her the vaccine. But for herself, she's not sure, you know, she confused. She want the doctor to check everything before she takes the vaccine—(Sita [Female, 29 years]; focus group attendee providing real‐time peer translation for Nell [Female, 70 years old])\nSeveral multicultural organizations and community leaders seeking to provide accurate information attempted to collaborate with government agencies or coordinate seminars engaging the community in their own environments.…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\n\n…she [CALD public health academic] came to the centre and gave talks about the vaccination … that's a good way of doing it—(Susi [Female, 50 years]; Interview)\nHowever, some community leaders were themselves sharing COVID‐19 vaccine misinformation on social media:On Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\n\nOn Facebook … even the priests and bishops are actually announcing to their parishioners the [the] vaccine is not good. (Reyna [Female, 53 years]; Community Leader, Interview)\nParticipants were disappointed in government attempts to access and work with their communities. There were no facilitation programmes in CALD communities, who lacked resources and noted little lingual or visual diversity in promotional campaigns.[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\n\n[Create] ‘Roll Up Your Sleeve’ [resources] in Arabic or ‘Roll Up Your Sleeve’ [resources] in Chinese. Not just English—(Kasih [Female, 50 years]; Interview)\nWhy not approach [CALD communities] instead of asking them to come in? Because it means we give them … practical plan … the [CALD] community people … [the WA government] cannot wait for the ball, [they] need to also try to get the ball—(Audrey [Female, 43 years]; Interview)\nParticipants believed that governments needed to be more active in reaching out to them, and said they would welcome efforts made by government and health representatives to come to the community to talk about COVID‐19 vaccination.", "Locations of mass COVID‐19 vaccination clinics were inconvenient for participants, given a lack of transport options, particularly as some could not drive. In an interview, Susi said:Even [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\n\nEven [the mass vaccination centre] in the city [Perth convention centre] … [it's] too hard, how will people go!?\nOther participants feared leaving their perceived safe and COVID‐19‐free community to travel into an unfamiliar part of Perth.My wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\n\nMy wife she was really, really panicked [to leave the house] … she was kind of anxious. Same for my mum—(Tai [Male, 29 years]; Interview)\nFor participants who did get vaccinated, the most convenient locations were clinics at or close to their workplaces or educational institutions. Some did attend mass vaccination clinics and found walk‐in processes highly convenient and well‐facilitated.", "Participants had high levels of trust in their own communities; opinions and experiences shared by peers were greatly valued and reassuring.In the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\n\nIn the community, we were very, very scared. But after that, when I saw friend and family [had been vaccinated], a couple of people they do fine, they are perfectly fine. My friend, she's a doctor. She done vaccine last month … her husband and other colleague did it, but they got a fever … they took Panadol after … is fine—(Momo [Female, 37 years]; Focus Group)\nHowever, the sharing of misinformation and negative opinions created situations in which some participants were torn between what their community was saying and the professional medical advice:When I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\n\nWhen I saw, yesterday, the doctor, everybody comes to me … say don't do this … But in my heart, I say, ‘No, I'm going to take it!’—(Ezma [Female, 40 years]; Focus Group)\nSome participants felt shame around being vaccinated, and sought to avoid undue attention or stigma from hesitant friends/family who were concerned that vaccinated people would die in 2 years:People … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\n\nPeople … said ‘be nice with them, they going to live only two years … After two years they will die’. (Momo [Female, 37 years]; Focus Group)\nOf the two community leaders we interviewed, one was completely against the idea of any medical intervention, including vaccination:I've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\n\nI've never taken any flu vaccines, I never take any painkillers, I avoid doctors like the plague … I want my body to learn how to cope and how to fight those diseases … And if I can't cope, I will simply go to the next world—(Ubel [Male, 78 years]; Interview)\nUbel volunteers at a community organization for refugees and migrants in Perth. His role is to share information with those in the database. When asked whether he shares information on COVID‐19 vaccination, he said:We get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\n\nWe get a quite a number [of emails from various official and unofficial sources] and I've passed on. Except yesterday for the first time, I did not send one on because this [particular] group sent me an email and they are totally convinced about COVID and it was a bit too strong for my liking. So I did not send it, but that's the first one.\nSome participants also discussed the role of community leaders, saying these leaders did not promote that they were vaccinated, fearing negative feedback or loss of power:[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\n\n[If community leaders] had a vaccine, they don't tell people, their own group … because other people … look up to them and … most of us don't believe in [the vaccine]—(Reyna [Female, 53 years]; Community Leader, Interview)\nReyna was a pro‐vaccination community leader, and she expressed frustration in feeling solely responsible for vaccine uptake in her wider CALD community. She felt the government had not come to her organization to provide support; community leaders instead had to organize seminars themselves with health representatives to answer community questions.We're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)\n\nWe're not the only not‐for‐profit community centre that are not well informed of the COVID and now [the WA government is] asking us to ‘roll up’ … how could you have a vaccine if you weren't knowledgeable, you have no education about it at all?—(Reyna [Female, 53 years]; Community Leader, Interview)", "Most participants were “scared”, “confused” or “unsure” about COVID‐19 vaccines, particularly because, as Umme said in a focus group “this medicine only come in 2 years, it come quickly, why so quickly, maybe something wrong”. Yet despite these concerns, many were nevertheless willing to be vaccinated to protect themselves, their family and their community.I'm getting this so that others around me don't get sick. I'm getting this so that I can protect my mum, I can protect my dad, I can protect my friends—(Angelo [Male, 37 years]; Interview)\n\nI'm getting this so that others around me don't get sick. I'm getting this so that I can protect my mum, I can protect my dad, I can protect my friends—(Angelo [Male, 37 years]; Interview)\nParticipants, aware that “Corona is coming”, felt they would be at greater risk once international and interstate borders opened, declaring that they would get vaccinated once WA borders opened (but likely not before). Others said the border opening would mean they could visit their friends and family overseas; they believed COVID‐19 vaccination would be a requirement for this:I want to go visit my family [in Ethiopia] … But [also] I'm happy that we are closed here. Because then we … stay safe—(Mahsa [Female, 34 years]; Focus Group)\n\nI want to go visit my family [in Ethiopia] … But [also] I'm happy that we are closed here. Because then we … stay safe—(Mahsa [Female, 34 years]; Focus Group)", "This is the first study to explore perceptions and attitudes to the COVID‐19 vaccine, barriers to preventing and strategies promoting vaccine uptake among CALD populations in Perth, WA. Similar to studies from the United Kingdom\n11\n, \n26\n and New South Wales,\n8\n we found high levels of COVID‐19 vaccine hesitancy among CALD individuals due to safety concerns, lack of understanding and high levels of misinformation perpetuated by CALD peers or social media platforms. Hesitancy and low uptake were particularly apparent among participants with low levels of education and low English proficiency. These findings have since been confirmed in a large regression analysis of over 12 million Australians, which identified that those with low levels of education and English proficiency were less likely to have received their first COVID‐19 vaccine dose.\n27\n\n\nDespite their concerns, participants were not completely opposed to vaccination, and we were struck by how they did not see the COVID‐19 vaccination programme as being for people like them. They were exposed to misinformation through their social networks, as previous studies have also found,\n11\n and the hesitancy this generated is a wider problem. However, participants also felt that governments were not forthcoming with resources to address their concerns. Participants were ostensibly included in cohorts that were eligible for vaccination at the time of data collection, and some of them had been eligible many months prior, due to their age. However, although they were ostensibly being asked to ‘roll up’, they received few or no personal cues to avail themselves of vaccinations.\nPractical barriers were also evident. As previously identified by Abdi et al.,\n28\n refugees and migrants tend to have quite demanding competing priorities, such as seeking employment and accommodation and thus need prompting for vaccination. In some cases, our participants' efforts to be vaccinated were thwarted by the online booking system. In fact, some of our research team assisted participants to book vaccination appointments on their smartphones after the focus groups or provide further information to partners who waited in the car park to speak with them. It is not just our participants who struggled with the online booking system: Seale et al.\n29\n describe a ‘digital divide’ in which those without access to the internet and have low English proficiency are disadvantaged in their ability to access COVID‐19 vaccines, and Biddle et al.\n27\n identified that Australians who completed the 2016 census on paper rather than online had a lower probability of vaccination, suggesting that individuals with lower digital literacy may experience more barriers to vaccination. Booking systems must be accessible to varying levels of digital literacy and English proficiency.\nSocial norms are a strong influence on vaccine uptake.\n16\n, \n30\n For CALD people in Perth, social norms were blocking COVID‐19 vaccination. Vaccinated CALD individuals were not sharing their experiences or reasons for vaccinating, facilitating ongoing shame and stigma, and thereby perpetuating continued silence for those who did vaccinate; tendencies that have also been found in local ‘alternative’ communities with regard to childhood vaccination.\n31\n Some CALD leaders were also not actively encouraging vaccination due to their own concerns or opposition, an issue identified early on by the Australian multicultural sector as a potential barrier.\n32\n\n\nAnother well‐established influence on vaccine uptake is receiving a recommendation from a healthcare provider.\n30\n Our participants showed great trust in their GPs and reported positive experiences of consultation. Accordingly, they believed that GPs would have more opportunities to address COVID‐19 vaccine concerns than mass vaccination clinic staff. However, GPs may not always be easily accessible within an acceptable timeframe to the patient,\n33\n meaning some CALD individuals are turning to or relying on social media for their health/COVID‐19 information.\n9\n\n\nOur participants' high levels of trust in governing authorities was a novel finding, mostly related to feeling safe from COVID‐19 in WA. Focus groups with CALD individuals in New South Wales instead found participants were concerned that the government was using COVID‐19 vaccination to control people.\n8\n We were struck by how our participants' trust in government emanated from people whose experiences in their countries of origin had often been traumatic, culminating in their refugee status.", "A strong motivation for this research was to assist in strategic design, informing ongoing and future implementation of public health initiatives among CALD populations. Our experiences engaging with our CALD participants and their partners, including helping them to book appointments after the focus groups, emphasized how important it is that governments promptly address vaccine access issues, as well as campaigning and working directly with CALD groups, and featuring them visibly in messaging. The barriers regarding lack of access to resources and the circulation of misinformation must be addressed as soon as possible to reduce the COVID‐19 burden amongst the CALD population in WA. These lessons can then be utilized for future vaccine doses, for reaching the children of CALD populations, and for wider public health initiatives.\nAt the time of our study, CALD participants and their communities were not publicly sharing their vaccinated status and proudly promoting COVID‐19 vaccinations. However, it is unreasonable to demand more social courage from CALD individuals when what they need is more support. Given that governments are responsible for making vaccination accessible and acceptable to improve and sustain uptake,\n34\n they must recognize, work with, and train influential community leaders on the importance of and how to have meaningful vaccine conversations with their peers.\n32\n A recent review identified the who, the what and the how for effective community engagement during a pandemic: essentially, it is important to engage with the community from the very beginning for a better chance of behaviour change within the community.\n35\n In eastern Australia, Mahimbo et al.\n9\n identified that CALD communities are eager for community engagement, and for their leaders to champion the vaccine. Seale et al.\n29\n identified that while some CALD organizations in Australia were updating local CALD leaders on relevant COVID‐19 information to share with their community, there was a lack of training in how to do so. Training influential community leaders can occur over a relatively short period of time: a COVID‐19 information and support programme was offered in the United States to CALD community leaders which involved 1‐hour conference calls twice a week (we acknowledge that this is then reliant on the leaders having a decent level of computer literacy.)\n29\n Further, given the trust and respect our participants held for their health practitioners, it is imperative that governments support GPs to effectively communicate about COVID‐19 vaccination with CALD patients; this requires resources, training and remuneration.\n36\n Since our data collection, the Australian National Centre for Immunisation Research and Surveillance have published a COVID‐19 decision aid in Greek, Vietnamese, Arabic, Simplified Chinese and Traditional Chinese.\n37\n Immunisation providers could use or share this tool with their CALD patients (though its effectiveness in increasing vaccine uptake is yet to be evaluated).\nOur participants' high levels of trust in governing authorities is an untapped opportunity. WA participants knew that ‘Corona is coming’. Now that it has arrived, the government and healthcare providers must maintain the trust of CALD by reaching them promptly with vaccine doses and culturally appropriate information to reduce the burden of COVID‐19. If these measures fail, it would be entirely reasonable for CALD individuals to not blame their own choices, and instead look to the authorities they believed would protect them and ask why they were not offered greater support and access.\n[SUBTITLE] Limitations [SUBSECTION] Qualitative data is not widely generalizable, as its function is to generate deep insights when little is known about an issue, or when data cannot be captured appropriately via quantitative means, including across language barriers. Different Australian states had different pandemic experiences and our data may not be applicable to other states or countries, although our insights and reflections remain pertinent. Our interviews and focus groups took place well into the COVID‐19 pandemic and vaccine rollout, but before community transmission in WA—the subsequent ‘first wave’ from February 2022 may have changed participants' views. Our recruitment method for interviews skewed demographics towards those with high English proficiency, females (despite a social media campaign specifically recruiting men), high levels of education and high COVID‐19 vaccine uptake. However, we mitigated this by collaborating with a community centre for recent migrants/refugees to undertake focus groups with women possessing low levels of English proficiency, education, and COVID‐19 vaccine uptake. Participants' language constraints were mitigated by strong peer relationships that facilitated peer translation. Despite our efforts to mitigate these limitations, our study does not represent the entire CALD community in Perth.\nQualitative data is not widely generalizable, as its function is to generate deep insights when little is known about an issue, or when data cannot be captured appropriately via quantitative means, including across language barriers. Different Australian states had different pandemic experiences and our data may not be applicable to other states or countries, although our insights and reflections remain pertinent. Our interviews and focus groups took place well into the COVID‐19 pandemic and vaccine rollout, but before community transmission in WA—the subsequent ‘first wave’ from February 2022 may have changed participants' views. Our recruitment method for interviews skewed demographics towards those with high English proficiency, females (despite a social media campaign specifically recruiting men), high levels of education and high COVID‐19 vaccine uptake. However, we mitigated this by collaborating with a community centre for recent migrants/refugees to undertake focus groups with women possessing low levels of English proficiency, education, and COVID‐19 vaccine uptake. Participants' language constraints were mitigated by strong peer relationships that facilitated peer translation. Despite our efforts to mitigate these limitations, our study does not represent the entire CALD community in Perth.", "Qualitative data is not widely generalizable, as its function is to generate deep insights when little is known about an issue, or when data cannot be captured appropriately via quantitative means, including across language barriers. Different Australian states had different pandemic experiences and our data may not be applicable to other states or countries, although our insights and reflections remain pertinent. Our interviews and focus groups took place well into the COVID‐19 pandemic and vaccine rollout, but before community transmission in WA—the subsequent ‘first wave’ from February 2022 may have changed participants' views. Our recruitment method for interviews skewed demographics towards those with high English proficiency, females (despite a social media campaign specifically recruiting men), high levels of education and high COVID‐19 vaccine uptake. However, we mitigated this by collaborating with a community centre for recent migrants/refugees to undertake focus groups with women possessing low levels of English proficiency, education, and COVID‐19 vaccine uptake. Participants' language constraints were mitigated by strong peer relationships that facilitated peer translation. Despite our efforts to mitigate these limitations, our study does not represent the entire CALD community in Perth.", "This study provides in‐depth insight into the perceptions and attitudes of CALD communities in Perth, WA, regarding the COVID‐19 vaccine, identifying facilitators and barriers for engagement and uptake. CALD participants faced barriers including a lack of knowledge about COVID‐19 and the vaccines, low English proficiency and education levels, misinformation, passive government communication strategies and limited access to vaccine clinics/providers. They were, however, motivated to vaccinate by the imminent opening of state and international borders, trust in government and healthcare authorities, travel intentions and the desire to protect themselves and others. Despite high levels of trust and significant desire for vaccines, strategies at the time of our study were not meeting the CALD community's needs, and thus those within it remain at risk from the disease. Tailored intervention strategies are required to provide knowledge, address misinformation, and facilitate access to ensure uptake of COVID‐19 vaccines—including for additional doses—amongst CALD communities. Governments must work with trusted CALD community members and immunization providers such as GPs to disseminate tailored COVID‐19 vaccine information and adequately translated resources so that community members themselves can become stronger vaccine ambassadors to each other.", "Samantha J. Carlson supervised the data collection and analysis, led the initial manuscript drafting, contributed to the study design, contributed to project administration and coordinated the ethics and stakeholder engagement. Gracie Edwards collected the data and conducted the analysis as well as drafted the literature review and contributed to the overall manuscript drafting. Christopher C. Blyth contributed to the conceptualization of the project and provided oversight and input into the study and funding attainment, and reviewed and edited the manuscript. Barbara Nattabi supervised the data collection and analysis, contributed to the coding framework, provided CALD expertise, contributed to project administration and reviewed and edited the manuscript. Katie Attwell conceptualized the project, co‐developed the broader study methodology with Christopher C. Blyth, led the funding attainment, and supervised the conduct and administration of the broader research project. She contributed to the literature review and the drafting, reviewing and editing of the manuscript. All authors have approved the final manuscript.", "K. A. and C. C. B. are specialist advisors to the Australian Technical Advisory Group on Immunisation. The remaining authors declare no conflict of interest." ]
[ null, "methods", null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, "discussion", null, null, "conclusions", null, "COI-statement" ]
[ "attitudes", "COVID‐19", "ethnic and racial minorities", "health knowledge", "immunization", "misinformation", "vaccination" ]
Impact of COVID-19 on HIV late diagnosis in a specialized German centre.
36263724
The ongoing COVID-19 pandemic has been impeding HIV diagnosis and treatment worldwide. Data on the impact of COVID-19 on late diagnosis (LD) in Germany are lacking. Here we present novel data of a single-centre German HIV cohort assessing LD during COVID-19.
BACKGROUND
This is a non-interventional, single-centre retrospective cohort assessing the rate of LD comparing HIV diagnoses pre-COVID-19 with those during the COVID-19 pandemic. New diagnoses between 1 January 2019 and 1 February 2020 were classified as pre-COVID-19, and diagnoses between 1 February 2020 and 1 October 2021 were classified as during COVID-19.
METHODS
Between 1 January 2019 and 1 October 2021, 75 patients presented with newly diagnosed HIV infection, 34 pre-COVID-19 and 41 during COVID-19. LD increased to 83% (n = 34/41) during COVID-19 versus 59% (n = 20/34) pre-COVID-19, and CDC stage C3 rose to 44% (n = 18) versus 27%. Hospitalization rate increased to 49% (n = 20) during COVID-19 versus 29% pre-COVID-19, and 12% (n = 5) presented with HIV-associated neurological disease, whereas none were observed in the pre-COVID-19 group. The incidence of LD (p = 0.020), CD4 count < 350 cells/μL (p = 0.037) and < 200 cells/μL (p = 0.022) were statistically significantly associated with the ongoing COVID-pandemic. An association with HIV transmission risk was borderline significant (p = 0.055).
RESULTS
Despite comparable annual rates of new HIV diagnoses, LD has been increasing during the COVID-19 pandemic, resulting in more opportunistic infections and higher hospitalization rates, possibly reflecting pandemic-related shortages in HIV testing and care facilities. Maintaining HIV testing opportunities and access to treatment during a pandemic is crucial so as not to impede WHO elimination goals and so as to prevent an increase in AIDS-related morbidity and mortality.
CONCLUSIONS
[ "Humans", "Delayed Diagnosis", "HIV Infections", "COVID-19", "Retrospective Studies", "Pandemics" ]
9874758
null
null
METHODS
The study was designed as a non‐interventional single‐centre retrospective cohort at the University Hospital Bonn, a maximum healthcare provider with a specialized infectious disease outpatient clinic as well as an infectious disease ward. The aim was to assess the rate of LD, comparing HIV diagnoses pre‐COVID‐19 (PC) with those during the COVID‐19 pandemic (DC). LD was defined as a first CD4 count < 350 cells/μL with or without an AIDS‐defining illness at the time of HIV diagnosis. New diagnoses between 1 January 2019 and 1 February 2020 were classified as PC patients, and diagnoses between 1 February 2020 and 1 October 2021 were classified as DC patients. In addition to assessment of demographic data, mode of HIV transmission, CD4 count, HIV‐RNA and concomitant opportunistic infections (OIs), the hospitalization rate and possible necessary intensive care treatment and mortality were assessed. CD4 count was assessed by flow cytometry using XN‐Series (Sysmex America Inc., Lincolnshire, IL, USA) at the Institute of Clinical Biochemistry, University Hospital Bonn. Through use of the reagent Fluorocell WDF (Sysmex Europe GmbH, Norderstedt, Germany), absolute count (number/μL) and relative count (%) of CD4 and CD8 were assessed. HIV‐RNA was measured with an Abbot m2000 assay, with a lower detection rate of < 40 copies/mL blood. [SUBTITLE] Statistical Analysis [SUBSECTION] Statistical analysis was performed with IBM SPSS 28. Fisher's exact, χ2 and Mann–Whitney U‐test were used for statistical analysis. The study was conducted according to the Declaration of Helsinki. Statistical analysis was performed with IBM SPSS 28. Fisher's exact, χ2 and Mann–Whitney U‐test were used for statistical analysis. The study was conducted according to the Declaration of Helsinki.
RESULTS
Baseline characteristics are shown in Table 1. Between 1 January 2019 and 1 October 2021, 75 patients presented with newly diagnosed HIV infection at the University Hospital Bonn, 34 PC and 41 DC. Median ages were 33 years (range 18–71) for PC presentation and 34 years (18–76) for DC presentation. The main route of transmission was men who have sex with men (MSM), which decreased from 71% (n = 24/34) for PC patients to 51% (n = 21/41) for DC patients. The rate of first diagnosis during hospital stay was found to be highest among DC patients (46%, n = 19/41) and was greater than among PC patients, 30% (n = 10/33) of whom were diagnosed during hospital admission. Diagnosis through testing at specialist care (e.g. haematologist, dermatologist, gynaecologist, infectious disease specialist) due to referral from a general practitioner was the most common diagnostic method among PC patients (39%, n = 13/33). Baseline characteristics Abbreviation: MSM, men who have sex with men. Late presentation increased to 83% (n = 34/41) for DC patients versus 59% (n = 20/34) for PC patients, and CDC stage C3 rose to 44% (n = 18/41) versus 27% (n = 9/34), respectively. Median CD4 count was 291 cells/μL (range 3–930) for PC patients and decreased to 118 cells/μL (10–782) for DC patients. Consistently, median HIV‐RNA almost doubled in DC patients, with a median of 93.759 copies/mL (range 408–9110.000) versus 48.598 copies/mL (range 957–8012.596) for PC patients; 46% (n = 19/41) of DC patients presented with AIDS‐defining illness versus 26% (n = 9/34) of PC patients. Pneumocystis jirovecii pneumonia (PJP) was the most often reported AIDS‐defining illness and was found more frequently among PC (67%, n = 6/9) than among DC patients (37%, n = 7/19). The rate of hospitalization increased to 49% (n = 20/41) in DC patients versus 29% (n = 10/34) in PC patients. Thus, the proportion of patients who required intensive care treatment was higher among the PC cohort (40%, n = 4/10) than among the DC cohort (30%, n = 6/20). Interestingly, 26% (n = 5/19) of DC patients with AIDS‐defining illness presented with AIDS‐associated neurological disease, including cerebral toxoplasmosis and HIV‐associated neurological disorder, whereas none were observed among PC patients. In both groups the death rate was low (3%, n = 1/34 for PC and 2%, n = 1/41 for DC); one patient died due to PJP and one died due to lymphoma. The incidence of LD (p = 0.020), CD4 count < 350 cells/μL (p = 0.037) and < 200 cells /μL (p = 0.022) were statistically significantly associated with the ongoing COVID pandemic. Association with transmission risk was borderline significant (p = 0.055).
null
null
[ "INTRODUCTION", "Statistical Analysis", "AUTHOR CONTRIBUTIONS" ]
[ "Late diagnosis (LD) in HIV remains a challenging problem despite widespread accessibility to diagnostic tests and combined antiretroviral therapy (cART). In Germany, about one‐third of people living with HIV are diagnosed late, as defined by CD4 T‐cell count < 350 cells/μL with/or without manifest AIDS‐defining illness [1]. LD is known to increase morbidity and mortality in affected in‐patients and, moreover, to increase healthcare costs as well as transmission risk as a result of individuals being unaware of their HIV status [2, 3]. The ongoing COVID‐19 pandemic has been impeding HIV diagnosis and treatment worldwide [4].\nAs there has been substantial progress in decreasing the number of new HIV infections through low‐threshold HIV testing, treatment as prevention (TasP) and initiation of HIV pre‐exposure prophylaxis, the ongoing COVID‐19 pandemic has raised major concerns in keeping the high‐quality standard accessible, which is potentially increasing the rate of HIV transmissions and additionally increasing the number of HIV late diagosis.\nCurrently, data on the impact of COVID‐19 on late diagnosis in Germany are lacking. Here we present novel data of a single‐centre German HIV cohort assessing the rate and impact of late diagnosis during COVID‐19.", "Statistical analysis was performed with IBM SPSS 28. Fisher's exact, χ2 and Mann–Whitney U‐test were used for statistical analysis.\nThe study was conducted according to the Declaration of Helsinki.", "KvB designed the study and concept. Sample collection was performed by all co‐authors. Data analysis was performed by KvB. The manuscript was written by KvB and CB with full input from all co‐authors." ]
[ null, null, null ]
[ "INTRODUCTION", "METHODS", "Statistical Analysis", "RESULTS", "DISCUSSION", "AUTHOR CONTRIBUTIONS" ]
[ "Late diagnosis (LD) in HIV remains a challenging problem despite widespread accessibility to diagnostic tests and combined antiretroviral therapy (cART). In Germany, about one‐third of people living with HIV are diagnosed late, as defined by CD4 T‐cell count < 350 cells/μL with/or without manifest AIDS‐defining illness [1]. LD is known to increase morbidity and mortality in affected in‐patients and, moreover, to increase healthcare costs as well as transmission risk as a result of individuals being unaware of their HIV status [2, 3]. The ongoing COVID‐19 pandemic has been impeding HIV diagnosis and treatment worldwide [4].\nAs there has been substantial progress in decreasing the number of new HIV infections through low‐threshold HIV testing, treatment as prevention (TasP) and initiation of HIV pre‐exposure prophylaxis, the ongoing COVID‐19 pandemic has raised major concerns in keeping the high‐quality standard accessible, which is potentially increasing the rate of HIV transmissions and additionally increasing the number of HIV late diagosis.\nCurrently, data on the impact of COVID‐19 on late diagnosis in Germany are lacking. Here we present novel data of a single‐centre German HIV cohort assessing the rate and impact of late diagnosis during COVID‐19.", "The study was designed as a non‐interventional single‐centre retrospective cohort at the University Hospital Bonn, a maximum healthcare provider with a specialized infectious disease outpatient clinic as well as an infectious disease ward. The aim was to assess the rate of LD, comparing HIV diagnoses pre‐COVID‐19 (PC) with those during the COVID‐19 pandemic (DC). LD was defined as a first CD4 count < 350 cells/μL with or without an AIDS‐defining illness at the time of HIV diagnosis. New diagnoses between 1 January 2019 and 1 February 2020 were classified as PC patients, and diagnoses between 1 February 2020 and 1 October 2021 were classified as DC patients. In addition to assessment of demographic data, mode of HIV transmission, CD4 count, HIV‐RNA and concomitant opportunistic infections (OIs), the hospitalization rate and possible necessary intensive care treatment and mortality were assessed. CD4 count was assessed by flow cytometry using XN‐Series (Sysmex America Inc., Lincolnshire, IL, USA) at the Institute of Clinical Biochemistry, University Hospital Bonn. Through use of the reagent Fluorocell WDF (Sysmex Europe GmbH, Norderstedt, Germany), absolute count (number/μL) and relative count (%) of CD4 and CD8 were assessed. HIV‐RNA was measured with an Abbot m2000 assay, with a lower detection rate of < 40 copies/mL blood.\n[SUBTITLE] Statistical Analysis [SUBSECTION] Statistical analysis was performed with IBM SPSS 28. Fisher's exact, χ2 and Mann–Whitney U‐test were used for statistical analysis.\nThe study was conducted according to the Declaration of Helsinki.\nStatistical analysis was performed with IBM SPSS 28. Fisher's exact, χ2 and Mann–Whitney U‐test were used for statistical analysis.\nThe study was conducted according to the Declaration of Helsinki.", "Statistical analysis was performed with IBM SPSS 28. Fisher's exact, χ2 and Mann–Whitney U‐test were used for statistical analysis.\nThe study was conducted according to the Declaration of Helsinki.", "Baseline characteristics are shown in Table 1. Between 1 January 2019 and 1 October 2021, 75 patients presented with newly diagnosed HIV infection at the University Hospital Bonn, 34 PC and 41 DC. Median ages were 33 years (range 18–71) for PC presentation and 34 years (18–76) for DC presentation. The main route of transmission was men who have sex with men (MSM), which decreased from 71% (n = 24/34) for PC patients to 51% (n = 21/41) for DC patients. The rate of first diagnosis during hospital stay was found to be highest among DC patients (46%, n = 19/41) and was greater than among PC patients, 30% (n = 10/33) of whom were diagnosed during hospital admission. Diagnosis through testing at specialist care (e.g. haematologist, dermatologist, gynaecologist, infectious disease specialist) due to referral from a general practitioner was the most common diagnostic method among PC patients (39%, n = 13/33).\nBaseline characteristics\nAbbreviation: MSM, men who have sex with men.\nLate presentation increased to 83% (n = 34/41) for DC patients versus 59% (n = 20/34) for PC patients, and CDC stage C3 rose to 44% (n = 18/41) versus 27% (n = 9/34), respectively. Median CD4 count was 291 cells/μL (range 3–930) for PC patients and decreased to 118 cells/μL (10–782) for DC patients. Consistently, median HIV‐RNA almost doubled in DC patients, with a median of 93.759 copies/mL (range 408–9110.000) versus 48.598 copies/mL (range 957–8012.596) for PC patients; 46% (n = 19/41) of DC patients presented with AIDS‐defining illness versus 26% (n = 9/34) of PC patients. Pneumocystis jirovecii pneumonia (PJP) was the most often reported AIDS‐defining illness and was found more frequently among PC (67%, n = 6/9) than among DC patients (37%, n = 7/19). The rate of hospitalization increased to 49% (n = 20/41) in DC patients versus 29% (n = 10/34) in PC patients. Thus, the proportion of patients who required intensive care treatment was higher among the PC cohort (40%, n = 4/10) than among the DC cohort (30%, n = 6/20). Interestingly, 26% (n = 5/19) of DC patients with AIDS‐defining illness presented with AIDS‐associated neurological disease, including cerebral toxoplasmosis and HIV‐associated neurological disorder, whereas none were observed among PC patients. In both groups the death rate was low (3%, n = 1/34 for PC and 2%, n = 1/41 for DC); one patient died due to PJP and one died due to lymphoma.\nThe incidence of LD (p = 0.020), CD4 count < 350 cells/μL (p = 0.037) and < 200 cells /μL (p = 0.022) were statistically significantly associated with the ongoing COVID pandemic. Association with transmission risk was borderline significant (p = 0.055).", "To our knowledge this is the first real‐life cohort presenting novel data on the impact of COVID‐19 in HIV late diagnosis in Germany.\nEven with easy accessibility to testing facilities and the prompt treatment of newly diagnosed HIV patients, 40–55% of patients are still diagnosed with late diagnosis in Germany [5, 6]. Other European cohorts have shown similar LD rates between 42% and 58% [7, 8, 9, 10]. In the COHERE cohort, being heterosexual, originating from southern Europe or being of African descent were found to be associated with the highest risk of LD [10]. Older age has also been associated with an increased risk of LD [5]. It is an urgent goal to diagnose HIV patients at an early stage as it is a known fact that LD leads to increased morbidity and mortality as well as increased health costs. The UNAIDS 95‐95‐95 goals are at increasing risk of being missed due to the ongoing focus on COVID‐19 and the resulting missed opportunities of treating HIV patients. As reported in the UNAIDS HIV Services Tracking, a large, sustained decrease in HIV testing in the reporting countries was observed [11].\nWith the beginning of the COVID‐19 pandemic in Germany in February 2020, we found the rate of LD to be rising at our infectious disease department. The overall rate of LD during the ongoing pandemic has shown a 40% increase over the pre‐pandemic time‐frame, resulting in a 3.4‐fold higher odds ratio (95% confidence interval: 1.18–9.83). This is most likely due to the numerous impacts of the ongoing COVID‐9 pandemic in Germany. Beginning with increasing numbers of SARS‐CoV‐2 infections in spring 2020, the fear of acquiring SARS‐CoV‐2 infection rose steadily, which led to an overall decreased consultation rate by, and access to, physicians. Therefore, possible symptoms of progressed HIV infection might have been neglected or their investigation postponed by the individual, or been misinterpreted due to the main focus on COVID‐19.\nAccess to low‐threshold testing was impeded by down‐regulation of community service testing at checkpoints and municipal health centres. As a result, two main areas for detecting undiagnosed HIV infection (whether with symptoms or asymptomatic) were disrupted.\nConcerning risk of transmission, we found a decreasing rate of new HIV infections in MSM which was also observed in an Italian cohort during the ongoing COVID‐19 pandemic [12]. This could be due to reduced contacts during lockdown as well as the fewer possibilities for testing mentioned earlier, which is more widespread and implemented in this risk group. Previous cohorts have shown a decrease in late diagnosis in MSM over time, which might be due to the implementation of pre‐exposure prophylaxis [10]. In our study we found an increase in LD among heterosexuals. Moreover, transmission via heterosexual contact has been described earlier as an associated risk factor for LD [11]. Indeed, this group might be more affected by having less information about testing strategies as well as by a lack of awareness regarding their individual risk of infection and could therefore have been even more impeded during the ongoing COVID‐19 pandemic.\nRegarding the institution in which HIV infection was diagnosed, we found a shift towards diagnosis in hospital due to mandatory treatment, whereas diagnosis before COVID‐19 was most often performed in specialist practices. However, due to the high rate of LD in our cohort, this finding has to be interpreted with caution. This reflects the fact that ongoing LD during the COVID‐19 pandemic might be due to the reduced availability of low‐threshold testing and reduced slots at general practitioners. Moreover, LD, with its concomitant laboratory and clinical abnormalities, requires special awareness, underlining the fact that all physicians, regardless of their speciality, need to be aware of HIV‐associated symptoms and disease in order to determine the HIV diagnosis.\nA low CD4 count reflects the impaired immune function and increased risk of developing AIDS‐defining diseases, which all lead to increased morbidity and mortality. In our study, CD4 counts < 350 cells/μL and < 200 cells/μL were statistically significantly associated with the ongoing COVID‐19 pandemic, underlining the fact that we need to put all our efforts towards diagnosing patients at an earlier stage in order to prevent severe immune dysfunction and subsequent AIDS‐defining diseases.\nAt the same time, we found an almost doubled rate of AIDS‐defining diseases during the ongoing COVID‐19 pandemic. PJP, which is the most prevalent AIDS‐defining disease, was found most often in our study. It is important to note that more than half of patients with PJP are so severely ill that they require intensive care treatment; 40% (n = 2/5) of DC patients with PJP who required intensive care treatment had to undergo mechanical ventilation via intubation, as compared with 25% (n = 1/4) of PC patients. The overall increased rate of AIDS‐defining diseases raises morbidity and mortality, which also increases the cost of healthcare. Moreover, persistent organ damage or development of renal insufficiency after recovering from PJP or other AIDS‐defining diseases has to be taken into consideration.\nInterestingly, we diagnosed several patients with AIDS‐related neurological disorders, whereas none had been observed in the comparable pre‐COVID cohort. Since implementation of cART, incidence of HIV‐associated neurological diseases has been decreasing, and thus patients with LD are still at an elevated risk [13]. In particular, AIDS‐related neurological disorders cause significant and, most often, permanent harm to the patient and therefore early diagnosis is of vital importance. Both of our patients with cerebral toxoplasmosis still have tremendous neurological deficits. One patient who presented with hemiplegia due to cerebral toxoplasmosis and severely spread Kaposi sarcoma even went blind due to cytomegalovirus‐associated retinitis.\nOur study has several limitations. First, it is a monocentric study, and second it only reflects data from two time slots which are close together. The overall higher rate of LD might be overestimated due to coincidence and the historical fact of the increased amount of LD in our hospital, as it is well known for its special infectious disease department. Thus, this study reflects a real‐life cohort being affected by the ongoing COVID‐19 pandemic.\nIn conclusion we found an increased rate of LD in our cohort during the persisting COVID‐19 pandemic despite stable annual numbers of new HIV diagnoses, which underlines the barriers to HIV testing as well as general care as a result of the focus on COVID‐19. The outcome is more AIDS‐defining diseases and higher hospitalization rates. Maintaining HIV testing opportunities and access to treatment during a pandemic is crucial if it is not to impede WHO elimination goals and we are to prevent an increase in AIDS‐related morbidity and mortality.", "KvB designed the study and concept. Sample collection was performed by all co‐authors. Data analysis was performed by KvB. The manuscript was written by KvB and CB with full input from all co‐authors." ]
[ null, "methods", null, "results", "discussion", null ]
[ "CD4 count", "COVID‐19", "HIV", "late diagnosis" ]
Clinical evaluation of a novel molecular diagnosis kit for detecting Helicobacter pylori and clarithromycin-resistant using intragastric fluid.
36263754
Although there are many Helicobacter pylori (H. pylori) diagnostic methods, the culture and antibiotic susceptibility test is an important method for selecting the most effective H. pylori eradication regimen. However, this diagnostic method is complicated and takes several days; therefore, the development of a rapid and simple diagnostic method is required. Eradication failure due to clarithromycin (CAM) resistance should also be considered. In this study, we report the clinical evaluation of point-of-care testing (POCT) kit using intragastric fluid, a novel kit for detecting H. pylori and CAM resistance.
BACKGROUND
The study participants were 143 patients suspected of H. pylori infection and had an endoscopic examination. The novel diagnostic kit diagnosed H. pylori infection and CAM resistance-associated mutation using intragastric fluid. To diagnose H. pylori infection, the relationship between the diagnostic kit and conventional diagnostic methods (urea breath test, stool antigen test, culture test, and real-time polymerase chain reaction [PCR]) was evaluated. For CAM resistance-associated mutation detection, the concordance between the diagnostic kit and antibiotic susceptibility test was evaluated.
MATERIALS AND METHODS
The diagnosis of H. pylori infection with the novel molecular diagnostic kit using intragastric fluid showed significant relationship with conventional diagnostic methods. Especially when the culture was control, the sensitivity was 100% (67/67), the specificity was 95.9% (71/74), and the overall concordance was 97.9% (138/141). The detection of CAM resistance-associated mutations had a concordance rate of 97.0% (65/67) when compared with the antibiotic susceptibility test.
RESULTS
The H. pylori molecular POCT kit uses intragastric fluid as a sample and can diagnose H. pylori infection and detect CAM resistance-associated mutations within an hour. This novel kit is expected to prove useful in selecting the most effective eradication regimen for H. pylori.
CONCLUSIONS
[ "Humans", "Helicobacter pylori", "Clarithromycin", "Helicobacter Infections", "Drug Resistance, Bacterial", "Anti-Bacterial Agents", "Microbial Sensitivity Tests" ]
9788249
null
null
null
null
RESULTS
[SUBTITLE] Evaluation of “Helicobacter pylori molecular POCT kit” by Smart Gene™ in Helicobacter pylori infection diagnosis [SUBSECTION] Results from the use of the “H. pylori molecular POCT kit” by Smart Gene™ was strongly correlated with the results from conventional H. pylori diagnosing methods. To evaluate the diagnostic accuracy of “the molecular PCOT kit,” conventional H. pylori diagnostic methods, such as UBT, SAT, culture test, and PCR, were compared (Table 1). The culture test, with a sensitivity of 100% (67/67) and specificity of 95.9% (71/74), showed the highest concordance rate with POCT kit. Comparison of Smart Gene™ assay using intragastric fluid and conventional diagnosis of Helicobacter pylori infection Abbreviations: PCR, polymerase chain reaction; SAT, stool antigen test; UBT, urea breath test. Results from the use of the “H. pylori molecular POCT kit” by Smart Gene™ was strongly correlated with the results from conventional H. pylori diagnosing methods. To evaluate the diagnostic accuracy of “the molecular PCOT kit,” conventional H. pylori diagnostic methods, such as UBT, SAT, culture test, and PCR, were compared (Table 1). The culture test, with a sensitivity of 100% (67/67) and specificity of 95.9% (71/74), showed the highest concordance rate with POCT kit. Comparison of Smart Gene™ assay using intragastric fluid and conventional diagnosis of Helicobacter pylori infection Abbreviations: PCR, polymerase chain reaction; SAT, stool antigen test; UBT, urea breath test. [SUBTITLE] Discrepancy in the diagnosis of Helicobacter pylori by Smart Gene [SUBSECTION] The mismatch of “H. pylori molecular POCT kit” by Smart Gene™ from conventional H. pylori diagnosis methods occurred in 19 cases in this study. (Table 2). In most cases, the results of POCT kit did not match completely. However, in several test results, except Case 6, POCT kit gave different results. In Case 6, only the results of POCT kit showed positive. Three cases (Case 4, 5, 13) tested positive for only PCR but negative for other methods including POCT kit. List of the mismatch cases between Smart Gene™ assay and control methods Abbreviations: N.D., no data; PCR, polymerase chain reaction; SAT, Stool antigen test; UBT, UREA BREATH test. The mismatch of “H. pylori molecular POCT kit” by Smart Gene™ from conventional H. pylori diagnosis methods occurred in 19 cases in this study. (Table 2). In most cases, the results of POCT kit did not match completely. However, in several test results, except Case 6, POCT kit gave different results. In Case 6, only the results of POCT kit showed positive. Three cases (Case 4, 5, 13) tested positive for only PCR but negative for other methods including POCT kit. List of the mismatch cases between Smart Gene™ assay and control methods Abbreviations: N.D., no data; PCR, polymerase chain reaction; SAT, Stool antigen test; UBT, UREA BREATH test. [SUBTITLE] Comparison of CAM resistance and the mutation ratio of Helicobacter pylori 23S rRNA by pyrosequencing [SUBSECTION] Quantitative pyrosequencing analysis of mutation rates at positions 2142 and 2143 of the H. pylori 23S rRNA gene supported the results of “H. pylori molecular POCT kit” by Smart Gene™ in detecting clarithromycin resistance up to a mutation rate of about 20%. The results of POCT kit were compared with quantitative pyrosequencing analysis of the mutation rates of H. pylori 23S rRNA genes at positions 2142 and 2143. Regardless of their locations, all the mutations at position 2142 detected by pyrosequencing analysis were also detected as mutations by POCT kit. On the contrary, although mutations were detected by pyrosequencing analysis at position 2143, POCT kit could only detect the mutations at position 2143 if they were 20% or more (Figure 3). For the mutation at position 2143, the minimum mutation rate determined as a mutation by POCT kit was 15%, while the maximum mutation rate determined as a wild type by POCT kit was 23%. Comparison of CAM resistance determined by Smart Gene™ assay and mutation ratio of Helicobacter pylori 23S rRNA gene by pyrosequencing. CAM, clarithromycin Quantitative pyrosequencing analysis of mutation rates at positions 2142 and 2143 of the H. pylori 23S rRNA gene supported the results of “H. pylori molecular POCT kit” by Smart Gene™ in detecting clarithromycin resistance up to a mutation rate of about 20%. The results of POCT kit were compared with quantitative pyrosequencing analysis of the mutation rates of H. pylori 23S rRNA genes at positions 2142 and 2143. Regardless of their locations, all the mutations at position 2142 detected by pyrosequencing analysis were also detected as mutations by POCT kit. On the contrary, although mutations were detected by pyrosequencing analysis at position 2143, POCT kit could only detect the mutations at position 2143 if they were 20% or more (Figure 3). For the mutation at position 2143, the minimum mutation rate determined as a mutation by POCT kit was 15%, while the maximum mutation rate determined as a wild type by POCT kit was 23%. Comparison of CAM resistance determined by Smart Gene™ assay and mutation ratio of Helicobacter pylori 23S rRNA gene by pyrosequencing. CAM, clarithromycin [SUBTITLE] Evaluation of the detection of clarithromycin resistance using “Helicobacter pylori molecular POCT kit” by Smart Gene™ [SUBSECTION] “Helicobacter pylori molecular POCT kit” by Smart Gene™ was able to detect clarithromycin resistance up to the mutation rate of about 20% by pyrosequencing. In determining clarithromycin resistance, the concordance rate for resistance was 91.7% (22/24) and the concordance rate for susceptibility was 100% (43/43) between Smart Gene™ and the control test, antibiotic susceptibility test (AST). The overall match rate was 97.0% (65/67; Table 3). Two discrepancy results represented 13% and 23% in the mutation rate through quantitative pyrosequencing analysis, which are low mutation rates. Comparison of CAM resistance determined by Smart Gene™ assay and AST Abbreviation: AST, Antibiotic susceptibility test. “Helicobacter pylori molecular POCT kit” by Smart Gene™ was able to detect clarithromycin resistance up to the mutation rate of about 20% by pyrosequencing. In determining clarithromycin resistance, the concordance rate for resistance was 91.7% (22/24) and the concordance rate for susceptibility was 100% (43/43) between Smart Gene™ and the control test, antibiotic susceptibility test (AST). The overall match rate was 97.0% (65/67; Table 3). Two discrepancy results represented 13% and 23% in the mutation rate through quantitative pyrosequencing analysis, which are low mutation rates. Comparison of CAM resistance determined by Smart Gene™ assay and AST Abbreviation: AST, Antibiotic susceptibility test.
null
null
[ "INTRODUCTION", "Patient enrollment and Helicobacter pylori conventional testing", "Sample collection of gastric mucosal biopsy and intragastric fluid", "\nHelicobacter pylori antibiotic susceptibility test", "Smart Gene assay for detection of Helicobacter pylori\nDNA and CAM‐resistant mutation", "Real‐time PCR of Helicobacter pylori\n16S rRNA\n", "Quantitative analysis of Helicobacter pylori\nCAM‐resistant mutation ratio by pyrosequencing", "Statistical analysis", "Evaluation of “Helicobacter pylori molecular POCT kit” by Smart Gene™ in Helicobacter pylori infection diagnosis", "Discrepancy in the diagnosis of Helicobacter pylori by Smart Gene", "Comparison of CAM resistance and the mutation ratio of Helicobacter pylori\n23S rRNA by pyrosequencing", "Evaluation of the detection of clarithromycin resistance using “Helicobacter pylori molecular POCT kit” by Smart Gene™\n", "AUTHOR CONTRIBUTIONS", "FUNDING INFORMATION" ]
[ "\nHelicobacter pylori (H. pylori) infection causes various upper gastrointestinal disorders such as atrophic gastritis, gastric ulcer, duodenal ulcer, gastric cancer, gastric mucosa associated lymphoid tissue (MALT) lymphoma, and gastric hyperplastic polyp associated with chronic inflammation of the gastric mucosa.\n1\n\nH. pylori infection is associated with gastric cancer as an important cause. The International Agency for Research on Cancer, the cancer research agency of the World Health Organization, strongly recommends designing a global preventive method for gastric cancer, including H. pylori eradication therapy.\n2\n, \n3\n\n\nIn Japan, “H. pylori‐infected gastritis” has been included in the list of diseases covered by insurances since 2013. Consequently, eradication therapy for chronic gastritis is compensated by the national health insurance scheme. Interestingly, there has been a decrease in gastric cancer deaths since the start of the insurance coverage.\n4\n\n\nThe Japanese Society for Helicobacter Research guidelines recommend H. pylori eradication therapy as the primary preventive method for gastric cancer after antibiotic susceptibility test. Furthermore, the guidelines recommend the use of the therapy in two or three drug combinations to maximize the bacteria eradication rate because of possible eradication therapy failure.\n1\n The failure of H. pylori eradication therapy is largely due to the resistance to clarithromycin (CAM), which is the primary drug for primary eradication. CAM resistance is caused by a gene mutation at positions 2142 and 2143 of the 23S rRNA gene Domain V region of H. pylori.\n5\n We previously reported the applications of gastric wash sample (intragastric fluid), such as the detection of CAM resistance‐associated gene mutation of H. pylori, for selecting eradication therapy and quantitative analysis of H. pylori genotype via pyrosequencing analysis.\n6\n This detection method using gastric wash sample (intragastric fluid) results in non‐invasive and high safety compared with the collect gastric mucosal biopsy for culture and susceptibility testing.\n6\n, \n7\n In addition, the results of antibiotic susceptibility testing take about 1 week; therefore, the method is too time‐consuming and impractical in clinical practice. Therefore, physicians need a new, time‐saving test to determine antibiotic susceptibility.\nSmart Gene™ (MIZUHO MEDY Co., Ltd.) was developed on the concept of point‐of‐care testing (POCT), which can automatically perform nucleic acid extraction, amplification, and detection. The detection of Mycoplasma pneumoniae and coronavirus by Smart Gene™ with pharyngeal swabs shows high reproducibility and useability and allows quick patient triage.\n8\n, \n9\n, \n10\n, \n11\n In addition, the detection of H. pylori by Smart Gene™ with stool proved to be an effective non‐invasive test.\n12\n The “H. pylori gene detection POCT reagent,” a reagent for the fully automatic Smart Gene™, allows for the automatic detection of H. pylori and CAM resistance‐associated mutations based on the Qprobe method.\n13\n, \n14\n, \n15\n\n\nOur goal was the fast and accurate diagnosis of H. pylori and detection of CAM resistance for the purpose of determining a precise eradication therapy. Therefore, we evaluated the clinical performance of the “H. pylori molecular POCT kit” in diagnosing H. pylori infection and detecting CAM‐resistant mutations. In this paper, we report new fast and accurate clinical technique for the diagnosis H. pylori infection in as fast as about an hour with the kit by Smart Gene™.", "To collect samples through esophagogastroduodenoscopy (EGD) for this study, we assessed the eligibility of 151 patients suspected of H. pylori infection at the National Hospital Organization Hakodate Hospital and the Kawasaki Rinko Hospital from December 2019 to March 2021. However, a total 8 patients were excluded from the study because they withdraw their consent (n = 2) or did not undergo EGD (n = 6); therefore, 143 patients were enrolled in the study (Figure 1). In addition to EGD, selected patients underwent a urea breath test (UBT, Urea breath test, UBIT tablet, 100 mg; Otsuka Pharmaceutical Co., Ltd.) stool antigen test (SAT, Stool antigen test, Testmate rapid pylori antigen; Wakamoto Pharmaceutical Co., Ltd) and H. pylori culture test (BML Co., Ltd.) as part of the conventional tests required for the diagnosis of H. pylori infection. From the result of the tests, 70 patients were confirmed to be H. pylori‐positive and 73 patients were confirmed to be H. pylori‐negative.\nThe flowchart of participants. We finally enrolled 143 patients for whom we performed endoscopic examination to detect Helicobacter pylori infection and clarithromycin‐resistant mutation by conventional testing, pyrosequencing, and H. pylori gene measurement. AST, Antibiotic susceptibility test; EGD, esophagogastroduodenoscopy; PCR, Polymerase chain reaction; SAT, Stool antigen test; UBT, Urea breath test", "To diagnose H. pylori infection and antibiotic susceptibility for CAM, we collected the patients' intragastric fluid and performed gastric mucosal biopsy as part of the endoscopic examination procedure, which has been previously reported.\n6\n Patient drank 100 ml of water (including 80 mg dimethylpolysiloxane, 1 g sodium bicarbonate, and 20,000 units of pronase) 10 min prior to their endoscopy. We collected a sample tissue of approximately 5 mm under endoscopic observation using biopsy forceps. Patients' intragastric fluid was collected directly to a sample container (MD‐33050, SB‐Kawasumi Laboratories, Inc.) through the connected suction base of the endoscope (Figure 2A).\n(A) Collection of intragastric fluid using sample container. (B) Assay workflow for Helicobacter pylori molecular POCT kit with Smart Gene™", "To determine H. pylori susceptibility, we performed antibiotic susceptibility testing of CAM using gastric mucosal biopsy as a conventional diagnosing method. The susceptibility tests were conducted by the clinical testing contractor BML Co., Ltd. In detail, gastric biopsies were cultured on Nissui Plate Helicobacter agar (Nissui Pharmaceutical Co., Ltd.) under microaerophilic conditions (5% O2, 15% CO2, and 80% N2) for 4 days at 37°C. Then, the isolated 3 colonies were enriched on 5% sheep blood agar (Nippon Becton Dickinson Co., Ltd.). The enriched colonies were prepared to a McF1.0 bacterial solution, and 25 μl bacterial solution was added to 6 ml of 10% horse serum‐added Mueller Hinton broth (Nikken Biological Co., Ltd.) to prepare a sample. Antibiotic susceptibility testing of CAM was performed using the dry plate “Eiken” (Eiken Chemical Co., Ltd.). The antibiotic susceptibility of CAM was determined using 1 μg/ml, minimal inhibitory concentration (MIC) breakpoint, recommended by the Japan Society of Chemotherapy and the Clinical & Laboratory Standards Institute (CLSI).\n16\n, \n17\n\n", "To evaluate the effectiveness of the novel “H. pylori molecular POCT kit” in detecting H. pylori and CAM‐resistant mutations using Smart Gene™ (Mizuho Medy Co., Ltd.), we used intragastric fluid as a sample. We simply dropped intragastric fluid suspended into the test cartridge “H. pylori molecular POCT kit.” Smart Gene™ detects the H. pylori DNA and CAM‐resistant mutations at positions 2142 and 2143 of the 23S rRNA gene based on polymerase chain reaction (PCR) and QProbe. The principle of detecting gene mutations by QProbe is based on previous report that demonstrated use of the macrolide‐resistant Mycoplasma pneumoniae (MRMp) diagnostic kit.\n10\n The use of Smart Gene™ allows for the simultaneous diagnosis of H. pylori infection and detection CAM resistance‐associated mutation. In our study, the Smart Gene™ automatically processed the samples and generated the results within an hour. In addition, because we utilized intragastric fluid, there was no additional invasive procedure carried out on study participants at the time of endoscopy (Figure 2B).", "To compare the outcomes of different testing methods, we performed a molecular diagnostic real‐time PCR targeting the H. pylori 16S rRNA gene as our control.\n18\n We extracted DNA from 200 μl intragastric fluid with the QIAamp DNA Mini Kit (QIAGEN GmbH) to obtain 150 μl purified DNA. The forward primer CGC‐TAA‐GAG‐ATC‐AGC‐CTA‐TGT‐CC and the reverse primer CCG‐TGT‐CTC‐AGT‐TCC‐AGT‐GTG‐T were used for real‐time PCR. Real‐time PCR was performed using the Thermal Cycler Dice Real Time System III instrument (Takara Bio) using TB Green Premix Dimer Eraser Perfect Real Time reagent (Takara Bio) under the following PCR conditions: preheating at 95°C for 30 s, and 50 cycles at 95°C for 5 s and at 55°C for 30 s and at 72°C for 30 s. The PCR amplicon was confirmed by melting curve analysis.", "To assess and determine CAM resistance, we quantitatively analyzed mutation rates at positions 2142 and 2143 of the H. pylori 23S rRNA gene via pyrosequencing, adopting previously reported procedures.\n6\n One milliliters intragastric fluid was centrifuged at 1200 g for 15 min and 800 μl supernatant was decanted. 200 μl purified DNA was extracted from residue sample with the QIAamp DNA Mini Kit (QIAGEN GmbH). Thereafter, the H. pylori 23S rRNA gene was amplified and biotinylated by nested PCR. In the first reaction, a 255‐bp fragment was amplified with the forward primer ACG‐AGA‐TGG‐GAG‐CTG‐TCT‐CAA‐CC and the reverse primer AGC‐ATT‐GTC‐CTG‐CCT‐GTG‐GAT‐AAC. The amplified fragments were, thereafter, used as a template in the second reaction to amplify a 90‐bp fragment with the forward primer GAG‐GTG‐AAA‐ATT‐CCT‐CCT‐ACC‐CGC‐G and the reverse primer GCG‐CAT‐GAT‐ATT‐CCC‐ATT‐AGC‐AGT‐GC. Reactions consisted of touchdown PCR with denaturation at 95°C for 30 s, annealing at appropriate temperatures for 30 s, and extension at 72°C for 30 s. Finally, the amplified fragments were analyzed by pyrosequencing on a Pyromark Q24 system (QIAGEN) using primer ACC‐CGC‐GGC‐AAG‐ACG.", "Concordance between Smart Gene assay and conventional diagnosis was evaluated by Cohen's kappa coefficient. All statistical analyses were conducted using the R 4.2.1 software program (www.r‐project.org).", "Results from the use of the “H. pylori molecular POCT kit” by Smart Gene™ was strongly correlated with the results from conventional H. pylori diagnosing methods. To evaluate the diagnostic accuracy of “the molecular PCOT kit,” conventional H. pylori diagnostic methods, such as UBT, SAT, culture test, and PCR, were compared (Table 1). The culture test, with a sensitivity of 100% (67/67) and specificity of 95.9% (71/74), showed the highest concordance rate with POCT kit.\nComparison of Smart Gene™ assay using intragastric fluid and conventional diagnosis of Helicobacter pylori infection\nAbbreviations: PCR, polymerase chain reaction; SAT, stool antigen test; UBT, urea breath test.", "The mismatch of “H. pylori molecular POCT kit” by Smart Gene™ from conventional H. pylori diagnosis methods occurred in 19 cases in this study. (Table 2). In most cases, the results of POCT kit did not match completely. However, in several test results, except Case 6, POCT kit gave different results. In Case 6, only the results of POCT kit showed positive. Three cases (Case 4, 5, 13) tested positive for only PCR but negative for other methods including POCT kit.\nList of the mismatch cases between Smart Gene™ assay and control methods\nAbbreviations: N.D., no data; PCR, polymerase chain reaction; SAT, Stool antigen test; UBT, UREA BREATH test.", "Quantitative pyrosequencing analysis of mutation rates at positions 2142 and 2143 of the H. pylori 23S rRNA gene supported the results of “H. pylori molecular POCT kit” by Smart Gene™ in detecting clarithromycin resistance up to a mutation rate of about 20%. The results of POCT kit were compared with quantitative pyrosequencing analysis of the mutation rates of H. pylori 23S rRNA genes at positions 2142 and 2143. Regardless of their locations, all the mutations at position 2142 detected by pyrosequencing analysis were also detected as mutations by POCT kit. On the contrary, although mutations were detected by pyrosequencing analysis at position 2143, POCT kit could only detect the mutations at position 2143 if they were 20% or more (Figure 3). For the mutation at position 2143, the minimum mutation rate determined as a mutation by POCT kit was 15%, while the maximum mutation rate determined as a wild type by POCT kit was 23%.\nComparison of CAM resistance determined by Smart Gene™ assay and mutation ratio of Helicobacter pylori 23S rRNA gene by pyrosequencing. CAM, clarithromycin", "“Helicobacter pylori molecular POCT kit” by Smart Gene™ was able to detect clarithromycin resistance up to the mutation rate of about 20% by pyrosequencing. In determining clarithromycin resistance, the concordance rate for resistance was 91.7% (22/24) and the concordance rate for susceptibility was 100% (43/43) between Smart Gene™ and the control test, antibiotic susceptibility test (AST). The overall match rate was 97.0% (65/67; Table 3). Two discrepancy results represented 13% and 23% in the mutation rate through quantitative pyrosequencing analysis, which are low mutation rates.\nComparison of CAM resistance determined by Smart Gene™ assay and AST\nAbbreviation: AST, Antibiotic susceptibility test.", "MT performed the experiments and analyses and drafted the manuscript. YW performed the experiments. RO performed pyrosequencing. RW, MH, and KK performed the experiments. HY and FI supervised the experiments. MK supervised the entire project.", "MIZUHO MEDY Co., Ltd provided grants and H. pylori molecular POCT kit for this study, but played no role in the study design, data collection and interpretation, or in the decision to submit the work for publication." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "MATERIAL AND METHODS", "Patient enrollment and Helicobacter pylori conventional testing", "Sample collection of gastric mucosal biopsy and intragastric fluid", "\nHelicobacter pylori antibiotic susceptibility test", "Smart Gene assay for detection of Helicobacter pylori\nDNA and CAM‐resistant mutation", "Real‐time PCR of Helicobacter pylori\n16S rRNA\n", "Quantitative analysis of Helicobacter pylori\nCAM‐resistant mutation ratio by pyrosequencing", "Statistical analysis", "RESULTS", "Evaluation of “Helicobacter pylori molecular POCT kit” by Smart Gene™ in Helicobacter pylori infection diagnosis", "Discrepancy in the diagnosis of Helicobacter pylori by Smart Gene", "Comparison of CAM resistance and the mutation ratio of Helicobacter pylori\n23S rRNA by pyrosequencing", "Evaluation of the detection of clarithromycin resistance using “Helicobacter pylori molecular POCT kit” by Smart Gene™\n", "DISCUSSION", "AUTHOR CONTRIBUTIONS", "FUNDING INFORMATION", "CONFLICT OF INTEREST" ]
[ "\nHelicobacter pylori (H. pylori) infection causes various upper gastrointestinal disorders such as atrophic gastritis, gastric ulcer, duodenal ulcer, gastric cancer, gastric mucosa associated lymphoid tissue (MALT) lymphoma, and gastric hyperplastic polyp associated with chronic inflammation of the gastric mucosa.\n1\n\nH. pylori infection is associated with gastric cancer as an important cause. The International Agency for Research on Cancer, the cancer research agency of the World Health Organization, strongly recommends designing a global preventive method for gastric cancer, including H. pylori eradication therapy.\n2\n, \n3\n\n\nIn Japan, “H. pylori‐infected gastritis” has been included in the list of diseases covered by insurances since 2013. Consequently, eradication therapy for chronic gastritis is compensated by the national health insurance scheme. Interestingly, there has been a decrease in gastric cancer deaths since the start of the insurance coverage.\n4\n\n\nThe Japanese Society for Helicobacter Research guidelines recommend H. pylori eradication therapy as the primary preventive method for gastric cancer after antibiotic susceptibility test. Furthermore, the guidelines recommend the use of the therapy in two or three drug combinations to maximize the bacteria eradication rate because of possible eradication therapy failure.\n1\n The failure of H. pylori eradication therapy is largely due to the resistance to clarithromycin (CAM), which is the primary drug for primary eradication. CAM resistance is caused by a gene mutation at positions 2142 and 2143 of the 23S rRNA gene Domain V region of H. pylori.\n5\n We previously reported the applications of gastric wash sample (intragastric fluid), such as the detection of CAM resistance‐associated gene mutation of H. pylori, for selecting eradication therapy and quantitative analysis of H. pylori genotype via pyrosequencing analysis.\n6\n This detection method using gastric wash sample (intragastric fluid) results in non‐invasive and high safety compared with the collect gastric mucosal biopsy for culture and susceptibility testing.\n6\n, \n7\n In addition, the results of antibiotic susceptibility testing take about 1 week; therefore, the method is too time‐consuming and impractical in clinical practice. Therefore, physicians need a new, time‐saving test to determine antibiotic susceptibility.\nSmart Gene™ (MIZUHO MEDY Co., Ltd.) was developed on the concept of point‐of‐care testing (POCT), which can automatically perform nucleic acid extraction, amplification, and detection. The detection of Mycoplasma pneumoniae and coronavirus by Smart Gene™ with pharyngeal swabs shows high reproducibility and useability and allows quick patient triage.\n8\n, \n9\n, \n10\n, \n11\n In addition, the detection of H. pylori by Smart Gene™ with stool proved to be an effective non‐invasive test.\n12\n The “H. pylori gene detection POCT reagent,” a reagent for the fully automatic Smart Gene™, allows for the automatic detection of H. pylori and CAM resistance‐associated mutations based on the Qprobe method.\n13\n, \n14\n, \n15\n\n\nOur goal was the fast and accurate diagnosis of H. pylori and detection of CAM resistance for the purpose of determining a precise eradication therapy. Therefore, we evaluated the clinical performance of the “H. pylori molecular POCT kit” in diagnosing H. pylori infection and detecting CAM‐resistant mutations. In this paper, we report new fast and accurate clinical technique for the diagnosis H. pylori infection in as fast as about an hour with the kit by Smart Gene™.", "[SUBTITLE] Patient enrollment and Helicobacter pylori conventional testing [SUBSECTION] To collect samples through esophagogastroduodenoscopy (EGD) for this study, we assessed the eligibility of 151 patients suspected of H. pylori infection at the National Hospital Organization Hakodate Hospital and the Kawasaki Rinko Hospital from December 2019 to March 2021. However, a total 8 patients were excluded from the study because they withdraw their consent (n = 2) or did not undergo EGD (n = 6); therefore, 143 patients were enrolled in the study (Figure 1). In addition to EGD, selected patients underwent a urea breath test (UBT, Urea breath test, UBIT tablet, 100 mg; Otsuka Pharmaceutical Co., Ltd.) stool antigen test (SAT, Stool antigen test, Testmate rapid pylori antigen; Wakamoto Pharmaceutical Co., Ltd) and H. pylori culture test (BML Co., Ltd.) as part of the conventional tests required for the diagnosis of H. pylori infection. From the result of the tests, 70 patients were confirmed to be H. pylori‐positive and 73 patients were confirmed to be H. pylori‐negative.\nThe flowchart of participants. We finally enrolled 143 patients for whom we performed endoscopic examination to detect Helicobacter pylori infection and clarithromycin‐resistant mutation by conventional testing, pyrosequencing, and H. pylori gene measurement. AST, Antibiotic susceptibility test; EGD, esophagogastroduodenoscopy; PCR, Polymerase chain reaction; SAT, Stool antigen test; UBT, Urea breath test\nTo collect samples through esophagogastroduodenoscopy (EGD) for this study, we assessed the eligibility of 151 patients suspected of H. pylori infection at the National Hospital Organization Hakodate Hospital and the Kawasaki Rinko Hospital from December 2019 to March 2021. However, a total 8 patients were excluded from the study because they withdraw their consent (n = 2) or did not undergo EGD (n = 6); therefore, 143 patients were enrolled in the study (Figure 1). In addition to EGD, selected patients underwent a urea breath test (UBT, Urea breath test, UBIT tablet, 100 mg; Otsuka Pharmaceutical Co., Ltd.) stool antigen test (SAT, Stool antigen test, Testmate rapid pylori antigen; Wakamoto Pharmaceutical Co., Ltd) and H. pylori culture test (BML Co., Ltd.) as part of the conventional tests required for the diagnosis of H. pylori infection. From the result of the tests, 70 patients were confirmed to be H. pylori‐positive and 73 patients were confirmed to be H. pylori‐negative.\nThe flowchart of participants. We finally enrolled 143 patients for whom we performed endoscopic examination to detect Helicobacter pylori infection and clarithromycin‐resistant mutation by conventional testing, pyrosequencing, and H. pylori gene measurement. AST, Antibiotic susceptibility test; EGD, esophagogastroduodenoscopy; PCR, Polymerase chain reaction; SAT, Stool antigen test; UBT, Urea breath test\n[SUBTITLE] Sample collection of gastric mucosal biopsy and intragastric fluid [SUBSECTION] To diagnose H. pylori infection and antibiotic susceptibility for CAM, we collected the patients' intragastric fluid and performed gastric mucosal biopsy as part of the endoscopic examination procedure, which has been previously reported.\n6\n Patient drank 100 ml of water (including 80 mg dimethylpolysiloxane, 1 g sodium bicarbonate, and 20,000 units of pronase) 10 min prior to their endoscopy. We collected a sample tissue of approximately 5 mm under endoscopic observation using biopsy forceps. Patients' intragastric fluid was collected directly to a sample container (MD‐33050, SB‐Kawasumi Laboratories, Inc.) through the connected suction base of the endoscope (Figure 2A).\n(A) Collection of intragastric fluid using sample container. (B) Assay workflow for Helicobacter pylori molecular POCT kit with Smart Gene™\nTo diagnose H. pylori infection and antibiotic susceptibility for CAM, we collected the patients' intragastric fluid and performed gastric mucosal biopsy as part of the endoscopic examination procedure, which has been previously reported.\n6\n Patient drank 100 ml of water (including 80 mg dimethylpolysiloxane, 1 g sodium bicarbonate, and 20,000 units of pronase) 10 min prior to their endoscopy. We collected a sample tissue of approximately 5 mm under endoscopic observation using biopsy forceps. Patients' intragastric fluid was collected directly to a sample container (MD‐33050, SB‐Kawasumi Laboratories, Inc.) through the connected suction base of the endoscope (Figure 2A).\n(A) Collection of intragastric fluid using sample container. (B) Assay workflow for Helicobacter pylori molecular POCT kit with Smart Gene™\n[SUBTITLE] \nHelicobacter pylori antibiotic susceptibility test [SUBSECTION] To determine H. pylori susceptibility, we performed antibiotic susceptibility testing of CAM using gastric mucosal biopsy as a conventional diagnosing method. The susceptibility tests were conducted by the clinical testing contractor BML Co., Ltd. In detail, gastric biopsies were cultured on Nissui Plate Helicobacter agar (Nissui Pharmaceutical Co., Ltd.) under microaerophilic conditions (5% O2, 15% CO2, and 80% N2) for 4 days at 37°C. Then, the isolated 3 colonies were enriched on 5% sheep blood agar (Nippon Becton Dickinson Co., Ltd.). The enriched colonies were prepared to a McF1.0 bacterial solution, and 25 μl bacterial solution was added to 6 ml of 10% horse serum‐added Mueller Hinton broth (Nikken Biological Co., Ltd.) to prepare a sample. Antibiotic susceptibility testing of CAM was performed using the dry plate “Eiken” (Eiken Chemical Co., Ltd.). The antibiotic susceptibility of CAM was determined using 1 μg/ml, minimal inhibitory concentration (MIC) breakpoint, recommended by the Japan Society of Chemotherapy and the Clinical & Laboratory Standards Institute (CLSI).\n16\n, \n17\n\n\nTo determine H. pylori susceptibility, we performed antibiotic susceptibility testing of CAM using gastric mucosal biopsy as a conventional diagnosing method. The susceptibility tests were conducted by the clinical testing contractor BML Co., Ltd. In detail, gastric biopsies were cultured on Nissui Plate Helicobacter agar (Nissui Pharmaceutical Co., Ltd.) under microaerophilic conditions (5% O2, 15% CO2, and 80% N2) for 4 days at 37°C. Then, the isolated 3 colonies were enriched on 5% sheep blood agar (Nippon Becton Dickinson Co., Ltd.). The enriched colonies were prepared to a McF1.0 bacterial solution, and 25 μl bacterial solution was added to 6 ml of 10% horse serum‐added Mueller Hinton broth (Nikken Biological Co., Ltd.) to prepare a sample. Antibiotic susceptibility testing of CAM was performed using the dry plate “Eiken” (Eiken Chemical Co., Ltd.). The antibiotic susceptibility of CAM was determined using 1 μg/ml, minimal inhibitory concentration (MIC) breakpoint, recommended by the Japan Society of Chemotherapy and the Clinical & Laboratory Standards Institute (CLSI).\n16\n, \n17\n\n\n[SUBTITLE] Smart Gene assay for detection of Helicobacter pylori\nDNA and CAM‐resistant mutation [SUBSECTION] To evaluate the effectiveness of the novel “H. pylori molecular POCT kit” in detecting H. pylori and CAM‐resistant mutations using Smart Gene™ (Mizuho Medy Co., Ltd.), we used intragastric fluid as a sample. We simply dropped intragastric fluid suspended into the test cartridge “H. pylori molecular POCT kit.” Smart Gene™ detects the H. pylori DNA and CAM‐resistant mutations at positions 2142 and 2143 of the 23S rRNA gene based on polymerase chain reaction (PCR) and QProbe. The principle of detecting gene mutations by QProbe is based on previous report that demonstrated use of the macrolide‐resistant Mycoplasma pneumoniae (MRMp) diagnostic kit.\n10\n The use of Smart Gene™ allows for the simultaneous diagnosis of H. pylori infection and detection CAM resistance‐associated mutation. In our study, the Smart Gene™ automatically processed the samples and generated the results within an hour. In addition, because we utilized intragastric fluid, there was no additional invasive procedure carried out on study participants at the time of endoscopy (Figure 2B).\nTo evaluate the effectiveness of the novel “H. pylori molecular POCT kit” in detecting H. pylori and CAM‐resistant mutations using Smart Gene™ (Mizuho Medy Co., Ltd.), we used intragastric fluid as a sample. We simply dropped intragastric fluid suspended into the test cartridge “H. pylori molecular POCT kit.” Smart Gene™ detects the H. pylori DNA and CAM‐resistant mutations at positions 2142 and 2143 of the 23S rRNA gene based on polymerase chain reaction (PCR) and QProbe. The principle of detecting gene mutations by QProbe is based on previous report that demonstrated use of the macrolide‐resistant Mycoplasma pneumoniae (MRMp) diagnostic kit.\n10\n The use of Smart Gene™ allows for the simultaneous diagnosis of H. pylori infection and detection CAM resistance‐associated mutation. In our study, the Smart Gene™ automatically processed the samples and generated the results within an hour. In addition, because we utilized intragastric fluid, there was no additional invasive procedure carried out on study participants at the time of endoscopy (Figure 2B).\n[SUBTITLE] Real‐time PCR of Helicobacter pylori\n16S rRNA\n [SUBSECTION] To compare the outcomes of different testing methods, we performed a molecular diagnostic real‐time PCR targeting the H. pylori 16S rRNA gene as our control.\n18\n We extracted DNA from 200 μl intragastric fluid with the QIAamp DNA Mini Kit (QIAGEN GmbH) to obtain 150 μl purified DNA. The forward primer CGC‐TAA‐GAG‐ATC‐AGC‐CTA‐TGT‐CC and the reverse primer CCG‐TGT‐CTC‐AGT‐TCC‐AGT‐GTG‐T were used for real‐time PCR. Real‐time PCR was performed using the Thermal Cycler Dice Real Time System III instrument (Takara Bio) using TB Green Premix Dimer Eraser Perfect Real Time reagent (Takara Bio) under the following PCR conditions: preheating at 95°C for 30 s, and 50 cycles at 95°C for 5 s and at 55°C for 30 s and at 72°C for 30 s. The PCR amplicon was confirmed by melting curve analysis.\nTo compare the outcomes of different testing methods, we performed a molecular diagnostic real‐time PCR targeting the H. pylori 16S rRNA gene as our control.\n18\n We extracted DNA from 200 μl intragastric fluid with the QIAamp DNA Mini Kit (QIAGEN GmbH) to obtain 150 μl purified DNA. The forward primer CGC‐TAA‐GAG‐ATC‐AGC‐CTA‐TGT‐CC and the reverse primer CCG‐TGT‐CTC‐AGT‐TCC‐AGT‐GTG‐T were used for real‐time PCR. Real‐time PCR was performed using the Thermal Cycler Dice Real Time System III instrument (Takara Bio) using TB Green Premix Dimer Eraser Perfect Real Time reagent (Takara Bio) under the following PCR conditions: preheating at 95°C for 30 s, and 50 cycles at 95°C for 5 s and at 55°C for 30 s and at 72°C for 30 s. The PCR amplicon was confirmed by melting curve analysis.\n[SUBTITLE] Quantitative analysis of Helicobacter pylori\nCAM‐resistant mutation ratio by pyrosequencing [SUBSECTION] To assess and determine CAM resistance, we quantitatively analyzed mutation rates at positions 2142 and 2143 of the H. pylori 23S rRNA gene via pyrosequencing, adopting previously reported procedures.\n6\n One milliliters intragastric fluid was centrifuged at 1200 g for 15 min and 800 μl supernatant was decanted. 200 μl purified DNA was extracted from residue sample with the QIAamp DNA Mini Kit (QIAGEN GmbH). Thereafter, the H. pylori 23S rRNA gene was amplified and biotinylated by nested PCR. In the first reaction, a 255‐bp fragment was amplified with the forward primer ACG‐AGA‐TGG‐GAG‐CTG‐TCT‐CAA‐CC and the reverse primer AGC‐ATT‐GTC‐CTG‐CCT‐GTG‐GAT‐AAC. The amplified fragments were, thereafter, used as a template in the second reaction to amplify a 90‐bp fragment with the forward primer GAG‐GTG‐AAA‐ATT‐CCT‐CCT‐ACC‐CGC‐G and the reverse primer GCG‐CAT‐GAT‐ATT‐CCC‐ATT‐AGC‐AGT‐GC. Reactions consisted of touchdown PCR with denaturation at 95°C for 30 s, annealing at appropriate temperatures for 30 s, and extension at 72°C for 30 s. Finally, the amplified fragments were analyzed by pyrosequencing on a Pyromark Q24 system (QIAGEN) using primer ACC‐CGC‐GGC‐AAG‐ACG.\nTo assess and determine CAM resistance, we quantitatively analyzed mutation rates at positions 2142 and 2143 of the H. pylori 23S rRNA gene via pyrosequencing, adopting previously reported procedures.\n6\n One milliliters intragastric fluid was centrifuged at 1200 g for 15 min and 800 μl supernatant was decanted. 200 μl purified DNA was extracted from residue sample with the QIAamp DNA Mini Kit (QIAGEN GmbH). Thereafter, the H. pylori 23S rRNA gene was amplified and biotinylated by nested PCR. In the first reaction, a 255‐bp fragment was amplified with the forward primer ACG‐AGA‐TGG‐GAG‐CTG‐TCT‐CAA‐CC and the reverse primer AGC‐ATT‐GTC‐CTG‐CCT‐GTG‐GAT‐AAC. The amplified fragments were, thereafter, used as a template in the second reaction to amplify a 90‐bp fragment with the forward primer GAG‐GTG‐AAA‐ATT‐CCT‐CCT‐ACC‐CGC‐G and the reverse primer GCG‐CAT‐GAT‐ATT‐CCC‐ATT‐AGC‐AGT‐GC. Reactions consisted of touchdown PCR with denaturation at 95°C for 30 s, annealing at appropriate temperatures for 30 s, and extension at 72°C for 30 s. Finally, the amplified fragments were analyzed by pyrosequencing on a Pyromark Q24 system (QIAGEN) using primer ACC‐CGC‐GGC‐AAG‐ACG.\n[SUBTITLE] Statistical analysis [SUBSECTION] Concordance between Smart Gene assay and conventional diagnosis was evaluated by Cohen's kappa coefficient. All statistical analyses were conducted using the R 4.2.1 software program (www.r‐project.org).\nConcordance between Smart Gene assay and conventional diagnosis was evaluated by Cohen's kappa coefficient. All statistical analyses were conducted using the R 4.2.1 software program (www.r‐project.org).", "To collect samples through esophagogastroduodenoscopy (EGD) for this study, we assessed the eligibility of 151 patients suspected of H. pylori infection at the National Hospital Organization Hakodate Hospital and the Kawasaki Rinko Hospital from December 2019 to March 2021. However, a total 8 patients were excluded from the study because they withdraw their consent (n = 2) or did not undergo EGD (n = 6); therefore, 143 patients were enrolled in the study (Figure 1). In addition to EGD, selected patients underwent a urea breath test (UBT, Urea breath test, UBIT tablet, 100 mg; Otsuka Pharmaceutical Co., Ltd.) stool antigen test (SAT, Stool antigen test, Testmate rapid pylori antigen; Wakamoto Pharmaceutical Co., Ltd) and H. pylori culture test (BML Co., Ltd.) as part of the conventional tests required for the diagnosis of H. pylori infection. From the result of the tests, 70 patients were confirmed to be H. pylori‐positive and 73 patients were confirmed to be H. pylori‐negative.\nThe flowchart of participants. We finally enrolled 143 patients for whom we performed endoscopic examination to detect Helicobacter pylori infection and clarithromycin‐resistant mutation by conventional testing, pyrosequencing, and H. pylori gene measurement. AST, Antibiotic susceptibility test; EGD, esophagogastroduodenoscopy; PCR, Polymerase chain reaction; SAT, Stool antigen test; UBT, Urea breath test", "To diagnose H. pylori infection and antibiotic susceptibility for CAM, we collected the patients' intragastric fluid and performed gastric mucosal biopsy as part of the endoscopic examination procedure, which has been previously reported.\n6\n Patient drank 100 ml of water (including 80 mg dimethylpolysiloxane, 1 g sodium bicarbonate, and 20,000 units of pronase) 10 min prior to their endoscopy. We collected a sample tissue of approximately 5 mm under endoscopic observation using biopsy forceps. Patients' intragastric fluid was collected directly to a sample container (MD‐33050, SB‐Kawasumi Laboratories, Inc.) through the connected suction base of the endoscope (Figure 2A).\n(A) Collection of intragastric fluid using sample container. (B) Assay workflow for Helicobacter pylori molecular POCT kit with Smart Gene™", "To determine H. pylori susceptibility, we performed antibiotic susceptibility testing of CAM using gastric mucosal biopsy as a conventional diagnosing method. The susceptibility tests were conducted by the clinical testing contractor BML Co., Ltd. In detail, gastric biopsies were cultured on Nissui Plate Helicobacter agar (Nissui Pharmaceutical Co., Ltd.) under microaerophilic conditions (5% O2, 15% CO2, and 80% N2) for 4 days at 37°C. Then, the isolated 3 colonies were enriched on 5% sheep blood agar (Nippon Becton Dickinson Co., Ltd.). The enriched colonies were prepared to a McF1.0 bacterial solution, and 25 μl bacterial solution was added to 6 ml of 10% horse serum‐added Mueller Hinton broth (Nikken Biological Co., Ltd.) to prepare a sample. Antibiotic susceptibility testing of CAM was performed using the dry plate “Eiken” (Eiken Chemical Co., Ltd.). The antibiotic susceptibility of CAM was determined using 1 μg/ml, minimal inhibitory concentration (MIC) breakpoint, recommended by the Japan Society of Chemotherapy and the Clinical & Laboratory Standards Institute (CLSI).\n16\n, \n17\n\n", "To evaluate the effectiveness of the novel “H. pylori molecular POCT kit” in detecting H. pylori and CAM‐resistant mutations using Smart Gene™ (Mizuho Medy Co., Ltd.), we used intragastric fluid as a sample. We simply dropped intragastric fluid suspended into the test cartridge “H. pylori molecular POCT kit.” Smart Gene™ detects the H. pylori DNA and CAM‐resistant mutations at positions 2142 and 2143 of the 23S rRNA gene based on polymerase chain reaction (PCR) and QProbe. The principle of detecting gene mutations by QProbe is based on previous report that demonstrated use of the macrolide‐resistant Mycoplasma pneumoniae (MRMp) diagnostic kit.\n10\n The use of Smart Gene™ allows for the simultaneous diagnosis of H. pylori infection and detection CAM resistance‐associated mutation. In our study, the Smart Gene™ automatically processed the samples and generated the results within an hour. In addition, because we utilized intragastric fluid, there was no additional invasive procedure carried out on study participants at the time of endoscopy (Figure 2B).", "To compare the outcomes of different testing methods, we performed a molecular diagnostic real‐time PCR targeting the H. pylori 16S rRNA gene as our control.\n18\n We extracted DNA from 200 μl intragastric fluid with the QIAamp DNA Mini Kit (QIAGEN GmbH) to obtain 150 μl purified DNA. The forward primer CGC‐TAA‐GAG‐ATC‐AGC‐CTA‐TGT‐CC and the reverse primer CCG‐TGT‐CTC‐AGT‐TCC‐AGT‐GTG‐T were used for real‐time PCR. Real‐time PCR was performed using the Thermal Cycler Dice Real Time System III instrument (Takara Bio) using TB Green Premix Dimer Eraser Perfect Real Time reagent (Takara Bio) under the following PCR conditions: preheating at 95°C for 30 s, and 50 cycles at 95°C for 5 s and at 55°C for 30 s and at 72°C for 30 s. The PCR amplicon was confirmed by melting curve analysis.", "To assess and determine CAM resistance, we quantitatively analyzed mutation rates at positions 2142 and 2143 of the H. pylori 23S rRNA gene via pyrosequencing, adopting previously reported procedures.\n6\n One milliliters intragastric fluid was centrifuged at 1200 g for 15 min and 800 μl supernatant was decanted. 200 μl purified DNA was extracted from residue sample with the QIAamp DNA Mini Kit (QIAGEN GmbH). Thereafter, the H. pylori 23S rRNA gene was amplified and biotinylated by nested PCR. In the first reaction, a 255‐bp fragment was amplified with the forward primer ACG‐AGA‐TGG‐GAG‐CTG‐TCT‐CAA‐CC and the reverse primer AGC‐ATT‐GTC‐CTG‐CCT‐GTG‐GAT‐AAC. The amplified fragments were, thereafter, used as a template in the second reaction to amplify a 90‐bp fragment with the forward primer GAG‐GTG‐AAA‐ATT‐CCT‐CCT‐ACC‐CGC‐G and the reverse primer GCG‐CAT‐GAT‐ATT‐CCC‐ATT‐AGC‐AGT‐GC. Reactions consisted of touchdown PCR with denaturation at 95°C for 30 s, annealing at appropriate temperatures for 30 s, and extension at 72°C for 30 s. Finally, the amplified fragments were analyzed by pyrosequencing on a Pyromark Q24 system (QIAGEN) using primer ACC‐CGC‐GGC‐AAG‐ACG.", "Concordance between Smart Gene assay and conventional diagnosis was evaluated by Cohen's kappa coefficient. All statistical analyses were conducted using the R 4.2.1 software program (www.r‐project.org).", "[SUBTITLE] Evaluation of “Helicobacter pylori molecular POCT kit” by Smart Gene™ in Helicobacter pylori infection diagnosis [SUBSECTION] Results from the use of the “H. pylori molecular POCT kit” by Smart Gene™ was strongly correlated with the results from conventional H. pylori diagnosing methods. To evaluate the diagnostic accuracy of “the molecular PCOT kit,” conventional H. pylori diagnostic methods, such as UBT, SAT, culture test, and PCR, were compared (Table 1). The culture test, with a sensitivity of 100% (67/67) and specificity of 95.9% (71/74), showed the highest concordance rate with POCT kit.\nComparison of Smart Gene™ assay using intragastric fluid and conventional diagnosis of Helicobacter pylori infection\nAbbreviations: PCR, polymerase chain reaction; SAT, stool antigen test; UBT, urea breath test.\nResults from the use of the “H. pylori molecular POCT kit” by Smart Gene™ was strongly correlated with the results from conventional H. pylori diagnosing methods. To evaluate the diagnostic accuracy of “the molecular PCOT kit,” conventional H. pylori diagnostic methods, such as UBT, SAT, culture test, and PCR, were compared (Table 1). The culture test, with a sensitivity of 100% (67/67) and specificity of 95.9% (71/74), showed the highest concordance rate with POCT kit.\nComparison of Smart Gene™ assay using intragastric fluid and conventional diagnosis of Helicobacter pylori infection\nAbbreviations: PCR, polymerase chain reaction; SAT, stool antigen test; UBT, urea breath test.\n[SUBTITLE] Discrepancy in the diagnosis of Helicobacter pylori by Smart Gene [SUBSECTION] The mismatch of “H. pylori molecular POCT kit” by Smart Gene™ from conventional H. pylori diagnosis methods occurred in 19 cases in this study. (Table 2). In most cases, the results of POCT kit did not match completely. However, in several test results, except Case 6, POCT kit gave different results. In Case 6, only the results of POCT kit showed positive. Three cases (Case 4, 5, 13) tested positive for only PCR but negative for other methods including POCT kit.\nList of the mismatch cases between Smart Gene™ assay and control methods\nAbbreviations: N.D., no data; PCR, polymerase chain reaction; SAT, Stool antigen test; UBT, UREA BREATH test.\nThe mismatch of “H. pylori molecular POCT kit” by Smart Gene™ from conventional H. pylori diagnosis methods occurred in 19 cases in this study. (Table 2). In most cases, the results of POCT kit did not match completely. However, in several test results, except Case 6, POCT kit gave different results. In Case 6, only the results of POCT kit showed positive. Three cases (Case 4, 5, 13) tested positive for only PCR but negative for other methods including POCT kit.\nList of the mismatch cases between Smart Gene™ assay and control methods\nAbbreviations: N.D., no data; PCR, polymerase chain reaction; SAT, Stool antigen test; UBT, UREA BREATH test.\n[SUBTITLE] Comparison of CAM resistance and the mutation ratio of Helicobacter pylori\n23S rRNA by pyrosequencing [SUBSECTION] Quantitative pyrosequencing analysis of mutation rates at positions 2142 and 2143 of the H. pylori 23S rRNA gene supported the results of “H. pylori molecular POCT kit” by Smart Gene™ in detecting clarithromycin resistance up to a mutation rate of about 20%. The results of POCT kit were compared with quantitative pyrosequencing analysis of the mutation rates of H. pylori 23S rRNA genes at positions 2142 and 2143. Regardless of their locations, all the mutations at position 2142 detected by pyrosequencing analysis were also detected as mutations by POCT kit. On the contrary, although mutations were detected by pyrosequencing analysis at position 2143, POCT kit could only detect the mutations at position 2143 if they were 20% or more (Figure 3). For the mutation at position 2143, the minimum mutation rate determined as a mutation by POCT kit was 15%, while the maximum mutation rate determined as a wild type by POCT kit was 23%.\nComparison of CAM resistance determined by Smart Gene™ assay and mutation ratio of Helicobacter pylori 23S rRNA gene by pyrosequencing. CAM, clarithromycin\nQuantitative pyrosequencing analysis of mutation rates at positions 2142 and 2143 of the H. pylori 23S rRNA gene supported the results of “H. pylori molecular POCT kit” by Smart Gene™ in detecting clarithromycin resistance up to a mutation rate of about 20%. The results of POCT kit were compared with quantitative pyrosequencing analysis of the mutation rates of H. pylori 23S rRNA genes at positions 2142 and 2143. Regardless of their locations, all the mutations at position 2142 detected by pyrosequencing analysis were also detected as mutations by POCT kit. On the contrary, although mutations were detected by pyrosequencing analysis at position 2143, POCT kit could only detect the mutations at position 2143 if they were 20% or more (Figure 3). For the mutation at position 2143, the minimum mutation rate determined as a mutation by POCT kit was 15%, while the maximum mutation rate determined as a wild type by POCT kit was 23%.\nComparison of CAM resistance determined by Smart Gene™ assay and mutation ratio of Helicobacter pylori 23S rRNA gene by pyrosequencing. CAM, clarithromycin\n[SUBTITLE] Evaluation of the detection of clarithromycin resistance using “Helicobacter pylori molecular POCT kit” by Smart Gene™\n [SUBSECTION] “Helicobacter pylori molecular POCT kit” by Smart Gene™ was able to detect clarithromycin resistance up to the mutation rate of about 20% by pyrosequencing. In determining clarithromycin resistance, the concordance rate for resistance was 91.7% (22/24) and the concordance rate for susceptibility was 100% (43/43) between Smart Gene™ and the control test, antibiotic susceptibility test (AST). The overall match rate was 97.0% (65/67; Table 3). Two discrepancy results represented 13% and 23% in the mutation rate through quantitative pyrosequencing analysis, which are low mutation rates.\nComparison of CAM resistance determined by Smart Gene™ assay and AST\nAbbreviation: AST, Antibiotic susceptibility test.\n“Helicobacter pylori molecular POCT kit” by Smart Gene™ was able to detect clarithromycin resistance up to the mutation rate of about 20% by pyrosequencing. In determining clarithromycin resistance, the concordance rate for resistance was 91.7% (22/24) and the concordance rate for susceptibility was 100% (43/43) between Smart Gene™ and the control test, antibiotic susceptibility test (AST). The overall match rate was 97.0% (65/67; Table 3). Two discrepancy results represented 13% and 23% in the mutation rate through quantitative pyrosequencing analysis, which are low mutation rates.\nComparison of CAM resistance determined by Smart Gene™ assay and AST\nAbbreviation: AST, Antibiotic susceptibility test.", "Results from the use of the “H. pylori molecular POCT kit” by Smart Gene™ was strongly correlated with the results from conventional H. pylori diagnosing methods. To evaluate the diagnostic accuracy of “the molecular PCOT kit,” conventional H. pylori diagnostic methods, such as UBT, SAT, culture test, and PCR, were compared (Table 1). The culture test, with a sensitivity of 100% (67/67) and specificity of 95.9% (71/74), showed the highest concordance rate with POCT kit.\nComparison of Smart Gene™ assay using intragastric fluid and conventional diagnosis of Helicobacter pylori infection\nAbbreviations: PCR, polymerase chain reaction; SAT, stool antigen test; UBT, urea breath test.", "The mismatch of “H. pylori molecular POCT kit” by Smart Gene™ from conventional H. pylori diagnosis methods occurred in 19 cases in this study. (Table 2). In most cases, the results of POCT kit did not match completely. However, in several test results, except Case 6, POCT kit gave different results. In Case 6, only the results of POCT kit showed positive. Three cases (Case 4, 5, 13) tested positive for only PCR but negative for other methods including POCT kit.\nList of the mismatch cases between Smart Gene™ assay and control methods\nAbbreviations: N.D., no data; PCR, polymerase chain reaction; SAT, Stool antigen test; UBT, UREA BREATH test.", "Quantitative pyrosequencing analysis of mutation rates at positions 2142 and 2143 of the H. pylori 23S rRNA gene supported the results of “H. pylori molecular POCT kit” by Smart Gene™ in detecting clarithromycin resistance up to a mutation rate of about 20%. The results of POCT kit were compared with quantitative pyrosequencing analysis of the mutation rates of H. pylori 23S rRNA genes at positions 2142 and 2143. Regardless of their locations, all the mutations at position 2142 detected by pyrosequencing analysis were also detected as mutations by POCT kit. On the contrary, although mutations were detected by pyrosequencing analysis at position 2143, POCT kit could only detect the mutations at position 2143 if they were 20% or more (Figure 3). For the mutation at position 2143, the minimum mutation rate determined as a mutation by POCT kit was 15%, while the maximum mutation rate determined as a wild type by POCT kit was 23%.\nComparison of CAM resistance determined by Smart Gene™ assay and mutation ratio of Helicobacter pylori 23S rRNA gene by pyrosequencing. CAM, clarithromycin", "“Helicobacter pylori molecular POCT kit” by Smart Gene™ was able to detect clarithromycin resistance up to the mutation rate of about 20% by pyrosequencing. In determining clarithromycin resistance, the concordance rate for resistance was 91.7% (22/24) and the concordance rate for susceptibility was 100% (43/43) between Smart Gene™ and the control test, antibiotic susceptibility test (AST). The overall match rate was 97.0% (65/67; Table 3). Two discrepancy results represented 13% and 23% in the mutation rate through quantitative pyrosequencing analysis, which are low mutation rates.\nComparison of CAM resistance determined by Smart Gene™ assay and AST\nAbbreviation: AST, Antibiotic susceptibility test.", "“Helicobacter pylori molecular POCT kit” by Smart Gene™ allows physicians to safely collect specimens and obtain diagnostic results on time. The simplicity of the kit allows for fast and accurate diagnosis of CAM resistance in determining a patient's precise eradication therapy. Consequently, its wider application will lead to more patients being tested for their H. pylori infection status.\nThe effectiveness of the “H. pylori molecular POCT kit” in detecting a CAM resistance‐associated mutation rate at a high concordance rate as an antibiotic susceptibility test proves beneficial in daily clinical practice. This novel kit can detect gene mutation at positions 2142 and 2143 of the 23S rRNA gene domain V region of H. pylori, precisely indicating CAM resistance. Furthermore, the “H. pylori molecular POCT kit” can detect CAM resistance‐associated mutations at a mutation rate of 20% or more when compared to pyrosequencing analysis. Results obtained by “H. pylori molecular POCT kit” in detecting CAM resistance is a great indication of patients' eradication results.\nUsing “H. pylori molecular POCT kit” for detecting CAM‐resistant mutation can significantly improve time‐efficiency in planning a specific treatment plan for patients. Due to its simplicity and speed, compared with other existing antibiotic susceptibility tests, physicians can speed up the process of selecting the appropriate antibiotic for the eradication of H. pylori, allowing patients to receive treatment as early as possible. As a result of the quick results provided by “H. pylori molecular POCT kit,” physicians can provide individualized treatment for patients with CAM resistance, such as skipping CAM‐based therapy and jumping into non‐CAM‐based therapy. Consequently, this will save physicians a significant amount of time they would have wasted on waiting for results and trying out ineffective treatments.\nAdditionally, “H. pylori molecular POCT kit” is safer for patients than conventional tests because it requires no additional invasive procedure during endoscopic examination and offers painless procedures. Patients only need to provide their intragastric fluid as a test specimen and do not have to go through gastric mucosal biopsy, which could cause complications, including excessive bleeding. Furthermore, because collecting intragastric fluid requires very little preparation time, patients do not have to suffer any pain, which they are likely to experience with longer preparation time. This significantly lower the risk of patient's vital transition.\nThe ease‐of‐use and reliability of results with the “H. pylori molecular POCT kit” could lead to the consolidation of all the different H. pylori diagnostic tests currently in use. Although the occurrence of false positives and negatives need to be further investigated, the kit has a high likelihood of replacing existing diagnostic tests. In this study, three cases (Case 4, 5, 13) tested positive for only PCR assumed to be the result of insufficient H. pylori amount.\n“H. pylori molecular POCT kit” is so simple and so capable of accurately and safely diagnosing H. pylori infection that it could be a great alternative diagnostic method in near future. Because the kit has a wide application and allows for the simultaneous testing of H. pylori infection and CAM resistance statuses, physicians can save time on eradication failure caused by CAM resistance.\nOne limitation of our study is the sample size. Although our sample size was sufficient enough to give us acceptable data, to increase the accuracy of the kit, we would like to enroll and study more people diagnosed with H. pylori infection. Because participating the study and eradicating H. pylori only lower participants' risk of developing gastric cancer, this study is also a part of eradicating overall gastric cancer itself.", "MT performed the experiments and analyses and drafted the manuscript. YW performed the experiments. RO performed pyrosequencing. RW, MH, and KK performed the experiments. HY and FI supervised the experiments. MK supervised the entire project.", "MIZUHO MEDY Co., Ltd provided grants and H. pylori molecular POCT kit for this study, but played no role in the study design, data collection and interpretation, or in the decision to submit the work for publication.", "The authors declare no conflict of interests associated with this study." ]
[ null, "materials-and-methods", null, null, null, null, null, null, null, "results", null, null, null, null, "discussion", null, null, "COI-statement" ]
[ "gastric juice", "\nH. pylori diagnosis", "PCR", "point‐of‐care testing", "susceptibility test" ]
Proprotein Convertase Subtilisin/Kexin 9 (PCSK9) Promotes Macrophage Activation via LDL Receptor-Independent Mechanisms.
36263780
Activated macrophages contribute to the pathogenesis of vascular disease. Vein graft failure is a major clinical problem with limited therapeutic options. PCSK9 (proprotein convertase subtilisin/kexin 9) increases low-density lipoprotein (LDL)-cholesterol levels via LDL receptor (LDLR) degradation. The role of PCSK9 in macrophage activation and vein graft failure is largely unknown, especially through LDLR-independent mechanisms. This study aimed to explore a novel mechanism of macrophage activation and vein graft disease induced by circulating PCSK9 in an LDLR-independent fashion.
BACKGROUND
We used Ldlr-/- mice to examine the LDLR-independent roles of circulating PCSK9 in experimental vein grafts. Adeno-associated virus (AAV) vector encoding a gain-of-function mutant of PCSK9 (rAAV8/D377Y-mPCSK9) induced hepatic PCSK9 overproduction. To explore novel inflammatory targets of PCSK9, we used systems biology in Ldlr-/- mouse macrophages.
METHODS
In Ldlr-/- mice, AAV-PCSK9 increased circulating PCSK9, but did not change serum cholesterol and triglyceride levels. AAV-PCSK9 promoted vein graft lesion development when compared with control AAV. In vivo molecular imaging revealed that AAV-PCSK9 increased macrophage accumulation and matrix metalloproteinase activity associated with decreased fibrillar collagen, a molecular determinant of atherosclerotic plaque stability. AAV-PCSK9 induced mRNA expression of the pro-inflammatory mediators IL-1β (interleukin-1 beta), TNFα (tumor necrosis factor alpha), and MCP-1 (monocyte chemoattractant protein-1) in peritoneal macrophages underpinned by an in vitro analysis of Ldlr-/- mouse macrophages stimulated with endotoxin-free recombinant PCSK9. A combination of unbiased global transcriptomics and new network-based hyperedge entanglement prediction analysis identified the NF-κB (nuclear factor-kappa B) signaling molecules, lectin-like oxidized LOX-1 (LDL receptor-1), and SDC4 (syndecan-4) as potential PCSK9 targets mediating pro-inflammatory responses in macrophages.
RESULTS
Circulating PCSK9 induces macrophage activation and vein graft lesion development via LDLR-independent mechanisms. PCSK9 may be a potential target for pharmacologic treatment for this unmet medical need.
CONCLUSIONS
[ "Animals", "Mice", "Cholesterol", "Lipoproteins, LDL", "Macrophage Activation", "NF-kappa B", "Proprotein Convertase 9", "Receptors, LDL", "Serine Endopeptidases", "Subtilisins" ]
9973449
null
null
Animal Procedures
All animal experiments were approved by the Brigham and Women’s Hospital’s Animal Welfare Assurance (protocol 2016N000219). Male Ldlr-/- mice were fed a high-fat diet (1.25% cholesterol, D12108C, Research Diets, Inc., New Brunswick, NJ). To create experimental vein grafts in Ldlr-/- mice, inferior vena cava were harvested from a donor mouse and implanted into the carotid artery of recipient mice using cuff technique, as previously described.8,21 To induce liver-specific gain-of-function mutant PCSK9, we administered adeno-associated virus (AAV) encoding a gain-of-function form of mouse PCSK9 (AAV-PCSK9, 1×1011 vg, i.v.).22 We constructed AAV encoding LacZ (AAV-LacZ) that we used as a control for AAV-PCSK9. Four weeks after implantation, grafts were evaluated. After ultrasonography and in vivo molecular imaging of vein grafts, vein graft tissues were harvested, sectioned, and used for Masson trichrome staining, picrosirius red staining, immunohistochemistry, and in situ hybridization.
Results
[SUBTITLE] AAV-PCSK9 Promotes Vein Graft Lesion Development in an LDLR-Independent Manner [SUBSECTION] In vivo studies have suggested a direct link between PCSK9 and atherosclerosis28,29; however, no evidence has yet implicated PCSK9 in vein graft lesion development. Furthermore, these in vivo studies indicate that the underlying mechanism of atherosclerotic lesion development by PCSK9 may depend mainly on LDLR. Whether PCSK9 can exert pro-inflammatory effects via LDLR-independent mechanisms is unclear. To test the hypothesis that circulating PCSK9 promotes vein graft lesion development in vivo, we used experimental vein grafts in mice.21 A single intravenous injection of AAV-PCSK9 induced liver production of PCSK9. To exclude potential systemic toxicity of AAV, AAV-LacZ was developed and used as a control (Figure 1A; Online Supplemental Methods). All experiments used Ldlr-/- mice to explore LDLR-independent mechanisms unless otherwise noted. The effects of Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) on vein graft lesion development in Ldlr-/- mice. A, An experimental protocol for the treatments in Ldlr-/- mice. B, In situ hybridization of PCSK9 (red) in the liver of Ldlr-/- mice 1 week after intravenous injection with AAV-LacZ or AAV-PCSK9. Nuclei are visualized with DAPI. The data represent 4 mice per group. Scale bars indicate 100 μm. C, PCSK9 mRNA level in the liver of AAV treated Ldlr-/- mice (n=12 and 11 for AAV-LacZ and AAV-PCSK9 groups, respectively), and serum levels of PCSK9, total cholesterol, and triglyceride in AAV treated Ldlr-/- mice (n=11 per group). D, Left, ultrasonography images of vein grafts from Ldlr-/- mice treated with AAV-LacZ or AAV-PCSK9 28 days after implantation. The red, green, and yellow dotted lines indicate lumen, near wall, and far wall, respectively. Right, The quantitative analysis of lumen and vessel wall area in short axis view and 3D reconstructed vessel wall volume of vein grafts by ultrasonography (n=12 and 11 for AAV-LacZ and AAV-PCSK9 groups, respectively). E, Left, Masson’s trichrome staining of vein grafts from Ldlr-/- mice treated with AAV-LacZ or AAV-PCSK9 28 days after implantation. Scale bars indicate 500 μm. Right, The quantitative analysis of intimal area, intimal and media/adventitia thickness, and lumen and vessel diameter of vein grafts (n=12 and 11 for AAV-LacZ and AAV-PCSK9 group, respectively). P value was calculated by Mann-Whitney U-test (triglyceride in C, media/adventitia thickness, lumen diameter, and vessel diameter in E) and unpaired Student t test (A, B, and D). Data are reported as mean ± SEM. Body weight and blood pressure did not differ significantly between the AAV-LacZ and AAV-PCSK9 groups 4 weeks after vein graft implantation (Supplemental Table 1). In situ hybridization showed that AAV-PCSK9 markedly increased PCSK9 mRNA levels in the liver (Figure 1B). AAV-PCSK9 increased PCSK9 mRNA expression in the liver and serum levels of PCSK9 (Figure 1C; Supplemental Table 1). Serum levels of total cholesterol and triglycerides did not differ between the AAV-LacZ and AAV-PCSK9 groups (Figure 1C; Supplemental Table 1). These findings indicate that the effects of AAV-PCSK9 on vein graft lesions described below did not depend on blood cholesterol and triglyceride concentrations (Supplemental Table 1). Noninvasive ultrasonography visualized that AAV-PCSK9 injection caused increased vessel wall area, but not lumen area, in the short axis view and 3-dimensional reconstructed vessel wall volume of vein grafts (Figure 1D). Consistent with the ultrasonography, histological analysis by Masson trichrome staining showed that AAV-PCSK9 increased intimal area and thickness of vein grafts. Lumen diameter, vessel diameter, and media/adventitia thickness did not differ significantly, suggesting compensatory outward remodeling of vein grafts (Figure 1E). In vivo studies have suggested a direct link between PCSK9 and atherosclerosis28,29; however, no evidence has yet implicated PCSK9 in vein graft lesion development. Furthermore, these in vivo studies indicate that the underlying mechanism of atherosclerotic lesion development by PCSK9 may depend mainly on LDLR. Whether PCSK9 can exert pro-inflammatory effects via LDLR-independent mechanisms is unclear. To test the hypothesis that circulating PCSK9 promotes vein graft lesion development in vivo, we used experimental vein grafts in mice.21 A single intravenous injection of AAV-PCSK9 induced liver production of PCSK9. To exclude potential systemic toxicity of AAV, AAV-LacZ was developed and used as a control (Figure 1A; Online Supplemental Methods). All experiments used Ldlr-/- mice to explore LDLR-independent mechanisms unless otherwise noted. The effects of Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) on vein graft lesion development in Ldlr-/- mice. A, An experimental protocol for the treatments in Ldlr-/- mice. B, In situ hybridization of PCSK9 (red) in the liver of Ldlr-/- mice 1 week after intravenous injection with AAV-LacZ or AAV-PCSK9. Nuclei are visualized with DAPI. The data represent 4 mice per group. Scale bars indicate 100 μm. C, PCSK9 mRNA level in the liver of AAV treated Ldlr-/- mice (n=12 and 11 for AAV-LacZ and AAV-PCSK9 groups, respectively), and serum levels of PCSK9, total cholesterol, and triglyceride in AAV treated Ldlr-/- mice (n=11 per group). D, Left, ultrasonography images of vein grafts from Ldlr-/- mice treated with AAV-LacZ or AAV-PCSK9 28 days after implantation. The red, green, and yellow dotted lines indicate lumen, near wall, and far wall, respectively. Right, The quantitative analysis of lumen and vessel wall area in short axis view and 3D reconstructed vessel wall volume of vein grafts by ultrasonography (n=12 and 11 for AAV-LacZ and AAV-PCSK9 groups, respectively). E, Left, Masson’s trichrome staining of vein grafts from Ldlr-/- mice treated with AAV-LacZ or AAV-PCSK9 28 days after implantation. Scale bars indicate 500 μm. Right, The quantitative analysis of intimal area, intimal and media/adventitia thickness, and lumen and vessel diameter of vein grafts (n=12 and 11 for AAV-LacZ and AAV-PCSK9 group, respectively). P value was calculated by Mann-Whitney U-test (triglyceride in C, media/adventitia thickness, lumen diameter, and vessel diameter in E) and unpaired Student t test (A, B, and D). Data are reported as mean ± SEM. Body weight and blood pressure did not differ significantly between the AAV-LacZ and AAV-PCSK9 groups 4 weeks after vein graft implantation (Supplemental Table 1). In situ hybridization showed that AAV-PCSK9 markedly increased PCSK9 mRNA levels in the liver (Figure 1B). AAV-PCSK9 increased PCSK9 mRNA expression in the liver and serum levels of PCSK9 (Figure 1C; Supplemental Table 1). Serum levels of total cholesterol and triglycerides did not differ between the AAV-LacZ and AAV-PCSK9 groups (Figure 1C; Supplemental Table 1). These findings indicate that the effects of AAV-PCSK9 on vein graft lesions described below did not depend on blood cholesterol and triglyceride concentrations (Supplemental Table 1). Noninvasive ultrasonography visualized that AAV-PCSK9 injection caused increased vessel wall area, but not lumen area, in the short axis view and 3-dimensional reconstructed vessel wall volume of vein grafts (Figure 1D). Consistent with the ultrasonography, histological analysis by Masson trichrome staining showed that AAV-PCSK9 increased intimal area and thickness of vein grafts. Lumen diameter, vessel diameter, and media/adventitia thickness did not differ significantly, suggesting compensatory outward remodeling of vein grafts (Figure 1E). [SUBTITLE] AAV-PCSK9 Increases Thin Collagen Fibers and Macrophage Accumulation in Vein Graft Lesions [SUBSECTION] Evidence suggests that vein grafts in humans can develop lesions similar to those of advanced arterial atherosclerosis, and plaque rupture can occur in inflamed vein grafts.30,31 Therefore, exploring vein graft lesion composition in depth is critical to understanding what contributes to this occurrence. We then examined the collagen content of vein grafts by picrosirius red staining. AAV-PCSK9 increased the percentage of thin collagen fibers as a ratio of the total collagen fibers in the vein grafts, suggesting that circulating PCSK9 induced pathological features similar to those in thin-capped atherosclerotic plaques (Figure 2A). In vivo molecular imaging further provided insight into the effects of circulating PCSK9 on matrix metalloproteinase activity and macrophage accumulation in vein grafts. We intravenously co-injected 2 spectrally different imaging agents that elaborate near-infrared signals for visualization of matrix metalloproteinase activity (MMPsense, 680 nm) and macrophage accumulation (AminoSPARK, 750 nm). Following the evidence that activated macrophages express MMPs that degrade interstitial collagen as previously reported by our group,32,33 in vivo molecular imaging demonstrated that increased matrix metalloproteinase activity co-localized with accumulated macrophages (Figure 2B, left). In addition, AAV-PCSK9 treatment promoted matrix metalloproteinase activity and macrophage accumulation in vein grafts (Figure 2B), suggesting a potential mechanism of collagen remodeling as determined by Picrosirius red staining. Immunohistochemical analysis further demonstrated increased macrophage accumulation in vein grafts by AAV-PCSK9 (Mac3; Figure 2C). Macrophage proliferation and migration may contribute to the mechanisms of macrophage accumulation in vascular diseases.34 Supporting this mechanism, AAV-PCSK9 increased the percentage of macrophages positive for Ki-67, an indicator of proliferating cells (Supplemental Figure 1A, B). Consistent with this in vivo finding, recombinant PCSK9 augmented M-CSF-induced cell proliferation in mouse bone marrow-derived macrophages (BMDMs) (Supplemental Figure 2A, B). Monocyte migration assay using the human monocytic cell line THP-1 showed that recombinant PCSK9 tended to promote MCP-1 (monocyte chemoattractant protein-1)-induced chemotaxis, suggesting lipid-independent effects of PCSK9 on monocyte migration (Supplemental Figure 2C). On the other hand, AAV-PCSK9 did not increase apoptosis determined by TUNEL staining in vein grafts of Ldlr-/- mice (Supplemental Figure 3A). Accordingly, AAV-PCSK9 did not change necrotic core area in the vein grafts (Supplemental Figure 3B). The effects of Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) on collagen thinning and macrophage accumulation in vein graft lesions of Ldlr-/- mice. A, Picrosirius red staining of vein grafts without (top) or with (bottom) polarized light (n=12 and 11 for AAV-LacZ and AAV-PCSK9 group, respectively). Scale bars indicate 500 μm. Circle graphs show the percentage of thin (green) and thick (red) collagen fibers compared with total fibers. B, Intravital microscopy images of MMPSense 680 (red) and AminoSPARK750 (green) in vein grafts for visualization of MMP activity and macrophage accumulation, respectively (n=10 and 9 for AAV-LacZ and AAV-PCSK9 group, respectively). C, Mac3 (macrophages) staining in vein grafts. Scale bars indicate 500 μm. P value was calculated by Mann-Whitney U-test (thick collagen in A, macrophage accumulation mean fluorescence intensity in B) and unpaired Student t test (C). Data are reported as mean ± SEM. Evidence suggests that vein grafts in humans can develop lesions similar to those of advanced arterial atherosclerosis, and plaque rupture can occur in inflamed vein grafts.30,31 Therefore, exploring vein graft lesion composition in depth is critical to understanding what contributes to this occurrence. We then examined the collagen content of vein grafts by picrosirius red staining. AAV-PCSK9 increased the percentage of thin collagen fibers as a ratio of the total collagen fibers in the vein grafts, suggesting that circulating PCSK9 induced pathological features similar to those in thin-capped atherosclerotic plaques (Figure 2A). In vivo molecular imaging further provided insight into the effects of circulating PCSK9 on matrix metalloproteinase activity and macrophage accumulation in vein grafts. We intravenously co-injected 2 spectrally different imaging agents that elaborate near-infrared signals for visualization of matrix metalloproteinase activity (MMPsense, 680 nm) and macrophage accumulation (AminoSPARK, 750 nm). Following the evidence that activated macrophages express MMPs that degrade interstitial collagen as previously reported by our group,32,33 in vivo molecular imaging demonstrated that increased matrix metalloproteinase activity co-localized with accumulated macrophages (Figure 2B, left). In addition, AAV-PCSK9 treatment promoted matrix metalloproteinase activity and macrophage accumulation in vein grafts (Figure 2B), suggesting a potential mechanism of collagen remodeling as determined by Picrosirius red staining. Immunohistochemical analysis further demonstrated increased macrophage accumulation in vein grafts by AAV-PCSK9 (Mac3; Figure 2C). Macrophage proliferation and migration may contribute to the mechanisms of macrophage accumulation in vascular diseases.34 Supporting this mechanism, AAV-PCSK9 increased the percentage of macrophages positive for Ki-67, an indicator of proliferating cells (Supplemental Figure 1A, B). Consistent with this in vivo finding, recombinant PCSK9 augmented M-CSF-induced cell proliferation in mouse bone marrow-derived macrophages (BMDMs) (Supplemental Figure 2A, B). Monocyte migration assay using the human monocytic cell line THP-1 showed that recombinant PCSK9 tended to promote MCP-1 (monocyte chemoattractant protein-1)-induced chemotaxis, suggesting lipid-independent effects of PCSK9 on monocyte migration (Supplemental Figure 2C). On the other hand, AAV-PCSK9 did not increase apoptosis determined by TUNEL staining in vein grafts of Ldlr-/- mice (Supplemental Figure 3A). Accordingly, AAV-PCSK9 did not change necrotic core area in the vein grafts (Supplemental Figure 3B). The effects of Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) on collagen thinning and macrophage accumulation in vein graft lesions of Ldlr-/- mice. A, Picrosirius red staining of vein grafts without (top) or with (bottom) polarized light (n=12 and 11 for AAV-LacZ and AAV-PCSK9 group, respectively). Scale bars indicate 500 μm. Circle graphs show the percentage of thin (green) and thick (red) collagen fibers compared with total fibers. B, Intravital microscopy images of MMPSense 680 (red) and AminoSPARK750 (green) in vein grafts for visualization of MMP activity and macrophage accumulation, respectively (n=10 and 9 for AAV-LacZ and AAV-PCSK9 group, respectively). C, Mac3 (macrophages) staining in vein grafts. Scale bars indicate 500 μm. P value was calculated by Mann-Whitney U-test (thick collagen in A, macrophage accumulation mean fluorescence intensity in B) and unpaired Student t test (C). Data are reported as mean ± SEM. [SUBTITLE] AAV-PCSK9 Induces Macrophage Activation via an LDLR-Independent Route In Vivo [SUBSECTION] We previously reported that macrophage activation plays a key role in vein graft lesion development.8 To explore the effects of circulating PCSK9 on macrophage activation, we examined mRNA levels of pro-inflammatory (IL-1β [interleukin-1 beta], IL-6, TNFα, and MCP-1) and antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1 [transforming growth factor-beta 1], and IL-10) in the peritoneal F4/80+ macrophages of Ldlr-/- mice 1 or 4 weeks after intravenous injection of AAVs. AAV-PCSK9 promoted pro-inflammatory IL-1β, IL-6, TNFα, and MCP-1 mRNA levels at 1 week (Figure 3A). AAV-PCSK9 also suppressed Arginase-1 mRNA levels at 4 weeks when the increase in the mRNA levels of pro-inflammatory molecules at 1 week had subsided (Figure 3B). We have recently reported that predominant pathways of the vein grafts change over time during lesion development. In the same study, pathways with immune responses represent the early responsive proteins in proteomics analysis.9 These results indicate that PCSK9 can induce pro-inflammatory responses in macrophages initially and later impair resolution of inflammation in vivo. Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) induced macrophage activation in murine peritoneal macrophages via LDL receptor (LDLR)-independent mechanisms in vivo. mRNA levels of pro-inflammatory molecules (IL-1β, IL-6, TNFα, MCP-1) and antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1, IL-10) were measured in murine peritoneal macrophages of Ldlr-/- mice 1 week (A) or 4 weeks (B) after intravenous injection with AAV-LacZ or AAV-PCSK9 (n=7 to 8 per group). P value was calculated by Mann-Whitney U-test (TNFa in A, IL-1b, TNFa, MCP-1, Arginase-1, Ym1, and IL-10 in B) and unpaired Student t test. Data are reported as mean ± SEM. We previously reported that macrophage activation plays a key role in vein graft lesion development.8 To explore the effects of circulating PCSK9 on macrophage activation, we examined mRNA levels of pro-inflammatory (IL-1β [interleukin-1 beta], IL-6, TNFα, and MCP-1) and antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1 [transforming growth factor-beta 1], and IL-10) in the peritoneal F4/80+ macrophages of Ldlr-/- mice 1 or 4 weeks after intravenous injection of AAVs. AAV-PCSK9 promoted pro-inflammatory IL-1β, IL-6, TNFα, and MCP-1 mRNA levels at 1 week (Figure 3A). AAV-PCSK9 also suppressed Arginase-1 mRNA levels at 4 weeks when the increase in the mRNA levels of pro-inflammatory molecules at 1 week had subsided (Figure 3B). We have recently reported that predominant pathways of the vein grafts change over time during lesion development. In the same study, pathways with immune responses represent the early responsive proteins in proteomics analysis.9 These results indicate that PCSK9 can induce pro-inflammatory responses in macrophages initially and later impair resolution of inflammation in vivo. Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) induced macrophage activation in murine peritoneal macrophages via LDL receptor (LDLR)-independent mechanisms in vivo. mRNA levels of pro-inflammatory molecules (IL-1β, IL-6, TNFα, MCP-1) and antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1, IL-10) were measured in murine peritoneal macrophages of Ldlr-/- mice 1 week (A) or 4 weeks (B) after intravenous injection with AAV-LacZ or AAV-PCSK9 (n=7 to 8 per group). P value was calculated by Mann-Whitney U-test (TNFa in A, IL-1b, TNFa, MCP-1, Arginase-1, Ym1, and IL-10 in B) and unpaired Student t test. Data are reported as mean ± SEM. [SUBTITLE] Endotoxin-Free Recombinant PCSK9 Induced Macrophage Activation in Peritoneal Macrophages of Ldlr -/- Mice In Vitro [SUBSECTION] To support our in vivo evidence, we examined in vitro the effects of physiologically relevant levels of recombinant mouse PCSK9 (0.1–2.5 μg/mL)35 on the mRNA levels of the same pro-inflammatory and antiinflammatory molecules examined in Ldlr-/- mouse macrophages. First, we conducted a time-course study of recombinant PCSK9-induced TNFα and Arginase-1 mRNA expression levels to determine an optimal time point for harvesting Ldlr-/- mouse macrophages. The increase of TNFα mRNA levels peaked 3 hours after stimulation, whereas the decrease of Arginase-1 mRNA levels plateaued 12 hours after stimulation with recombinant PCSK9 (Figure 4A). For all experiments, Ldlr-/- mouse macrophages were therefore harvested 3 and 12 hours after stimulation to measure pro- and antiinflammatory molecules, respectively. Peritoneal macrophages treated with recombinant PCSK9 exhibited increased mRNA levels of IL-1β, IL-6, TNFα, and MCP-1 in a concentration-dependent manner (Figure 4B). In contrast, recombinant PCSK9 did not decrease the mRNA levels of antiinflammatory molecules (Figure 4C). These results support the in vivo evidence of macrophage activation by circulating PCSK9 in an LDLR-independent fashion. Recombinant PCSK9 (proprotein convertase subtilisin/kexin 9) induced macrophage activation in LDL receptor (LDLR)-independent mechanisms in vitro. A, Time course of TNFα and Arginase-1 mRNA levels in peritoneal macrophages of wild-type mice after stimulation with recombinant mouse PCSK9 (MmPCSK9; 2.5 μg/mL) (n=5 per group). P value was calculated by Kruskal-Wallis test, followed by Dunn multiple comparison test. B, mRNA levels of pro-inflammatory molecules (IL-1β, IL-6, TNFα, MCP-1) and (C) antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1, IL-10) were measured in murine peritoneal macrophages from Ldlr-/- mice 3 and 12 hours after stimulation with recombinant mouse PCSK9 for pro-inflammatory and antiinflammatory molecules, respectively (n=9 per group). P value was calculated by 1-way ANOVA, followed by Dunnett multiple comparison test (IL-6, TNFa, MCP-1‚ MRC1) or Kruskal-Wallis test, followed by Dunn multiple comparison test. Data are reported as mean ± SEM. Endotoxin contamination in recombinant proteins can induce pro-inflammatory responses. Therefore, we chose a recombinant protein derived from a mammalian system, mouse myeloma cell line (NS0). The endotoxin level of recombinant mouse PCSK9 used in this study was 0.00441 EU/µg or less, well below the threshold of 0.1 EU/µg required for accurate cell-based assays.19 To exclude further endotoxin contamination in the recombinant PCSK9 protein affecting any measurements, we performed the chromogenic Limulus amebocyte lysate endotoxin assay. The final endotoxin level of the maximum dose of the recombinant protein (2.5 µg/mL) was 0.005 EU/mL or less, which coincides with the endotoxin levels of the commercially available endotoxin-free medium. In addition, mass spectrometry conducted on the solvent derived from reconstituted recombinant mouse PCSK9 protein solution did not produce signals, suggesting that there were no small molecule impurities present (Supplemental Figure 4A). Furthermore, stimulation with heat-inactivated recombinant mouse PCSK9 protein (2.5 μg/mL) caused no changes in the mRNA levels of IL-1β, TNF-α, and MCP-1, demonstrating that endotoxin contamination did not drive the effects observed (Supplemental Figure 4B). To support our in vivo evidence, we examined in vitro the effects of physiologically relevant levels of recombinant mouse PCSK9 (0.1–2.5 μg/mL)35 on the mRNA levels of the same pro-inflammatory and antiinflammatory molecules examined in Ldlr-/- mouse macrophages. First, we conducted a time-course study of recombinant PCSK9-induced TNFα and Arginase-1 mRNA expression levels to determine an optimal time point for harvesting Ldlr-/- mouse macrophages. The increase of TNFα mRNA levels peaked 3 hours after stimulation, whereas the decrease of Arginase-1 mRNA levels plateaued 12 hours after stimulation with recombinant PCSK9 (Figure 4A). For all experiments, Ldlr-/- mouse macrophages were therefore harvested 3 and 12 hours after stimulation to measure pro- and antiinflammatory molecules, respectively. Peritoneal macrophages treated with recombinant PCSK9 exhibited increased mRNA levels of IL-1β, IL-6, TNFα, and MCP-1 in a concentration-dependent manner (Figure 4B). In contrast, recombinant PCSK9 did not decrease the mRNA levels of antiinflammatory molecules (Figure 4C). These results support the in vivo evidence of macrophage activation by circulating PCSK9 in an LDLR-independent fashion. Recombinant PCSK9 (proprotein convertase subtilisin/kexin 9) induced macrophage activation in LDL receptor (LDLR)-independent mechanisms in vitro. A, Time course of TNFα and Arginase-1 mRNA levels in peritoneal macrophages of wild-type mice after stimulation with recombinant mouse PCSK9 (MmPCSK9; 2.5 μg/mL) (n=5 per group). P value was calculated by Kruskal-Wallis test, followed by Dunn multiple comparison test. B, mRNA levels of pro-inflammatory molecules (IL-1β, IL-6, TNFα, MCP-1) and (C) antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1, IL-10) were measured in murine peritoneal macrophages from Ldlr-/- mice 3 and 12 hours after stimulation with recombinant mouse PCSK9 for pro-inflammatory and antiinflammatory molecules, respectively (n=9 per group). P value was calculated by 1-way ANOVA, followed by Dunnett multiple comparison test (IL-6, TNFa, MCP-1‚ MRC1) or Kruskal-Wallis test, followed by Dunn multiple comparison test. Data are reported as mean ± SEM. Endotoxin contamination in recombinant proteins can induce pro-inflammatory responses. Therefore, we chose a recombinant protein derived from a mammalian system, mouse myeloma cell line (NS0). The endotoxin level of recombinant mouse PCSK9 used in this study was 0.00441 EU/µg or less, well below the threshold of 0.1 EU/µg required for accurate cell-based assays.19 To exclude further endotoxin contamination in the recombinant PCSK9 protein affecting any measurements, we performed the chromogenic Limulus amebocyte lysate endotoxin assay. The final endotoxin level of the maximum dose of the recombinant protein (2.5 µg/mL) was 0.005 EU/mL or less, which coincides with the endotoxin levels of the commercially available endotoxin-free medium. In addition, mass spectrometry conducted on the solvent derived from reconstituted recombinant mouse PCSK9 protein solution did not produce signals, suggesting that there were no small molecule impurities present (Supplemental Figure 4A). Furthermore, stimulation with heat-inactivated recombinant mouse PCSK9 protein (2.5 μg/mL) caused no changes in the mRNA levels of IL-1β, TNF-α, and MCP-1, demonstrating that endotoxin contamination did not drive the effects observed (Supplemental Figure 4B). [SUBTITLE] A combination of Unbiased Global Transcriptomics and Network-Based Hyperedge Entanglement Prediction Analysis Revealed Potential Targets of PCSK9 in Macrophages [SUBSECTION] We explored further LDLR-independent pro-inflammatory signaling pathways in an unbiased manner, using RNA-sequencing of Ldlr-/- mouse macrophages stimulated with recombinant PCSK9. When analyzing the differentially expressed transcripts (FC>2.0, q<0.001), we observed 46 transcripts significantly increased by PCSK9 stimulation in Ldlr-/- macrophages compared with controls (Figure 5A). A hierarchical heat map of Ldlr-/- mouse macrophages filtered by differentially expressed transcripts showed a clear separation between control (no stimulus) and PCSK9 group (Figure 5B). We provided statistically significantly overrepresented transcripts ranked according to their fold change abundance (Supplemental Table 3A) to further examine these differentially expressed transcripts. PCSK9 treatment increased many transcripts implicated in pro-inflammatory responses in Ldlr-/- macrophages (eg, IL-1b, TNF, CXCL2, NF-κB [nuclear factor-kappa B]-related genes).19 Transcriptomics, network analysis, and hyperedge entanglement prediction (HEP) of mouse macrophages stimulated with recombinant PCSK9 (proprotein convertase subtilisin/kexin 9). A, Volcano plot for Ldlr-/- mouse macrophages stimulated with recombinant PCSK9. Red markers indicate significantly expressed transcripts with a fold change cutoff of 2.0 and adjusted P value cutoff of 0.001. The enriched transcripts that were previously reported were denoted with their names. B, Heat map of the transcriptomics of Ldlr-/- mouse macrophages (FC>2, q<0.001-filtered data for the difference between macrophages stimulated with and without recombinant PCSK9, n=4 per group). C, The schematic network visualization by HEP prediction. The significantly predicted association of PCSK9 to the 24 mapped inflammation-related transcripts derived from the list of transcripts (FC>2.0, q<0.05) indicates every unknown pairs of interaction (dashed line) that have to be proven yet experimentally. One of them was validated by silencing Sdc4 (syndecan-4) mRNA (blue dashed line). All other links represent known physical interactions (solid gray lines). The node size illustrates the degree of the proteins in the protein-protein interaction network. D, Network analysis of the 24 overrepresented transcripts in Ldlr-/- mouse macrophages using STRING database. Nodes are colored according to k-means clustering (number of clusters=3). Only connected nodes are shown here. To provide additional mechanistic links, we attempted to identify PCSK9-regulated proteins other than LDLR that mediate the aforementioned pro-inflammatory downstream responses. Many cascading effects in the transcriptional regulatory machinery could control the differentially expressed mRNAs. This set of differentially expressed transcripts were identified as the result of a genome-wide transcriptional profiling method, which might not necessarily interact directly with PCSK9. To estimate the likelihood of PCSK9 directly interacting with those differentially expressed transcripts, we tested this hypothesis verification through network computational analysis. In brief, PCSK9 can be seen as a node in a protein-protein interaction network,36 and the set of differentially expressed transcripts can be seen as a hyperedge projected on the protein-protein interaction network (a group of nodes that are associated by a common feature of biological functions). Muscoloni et al recently introduced a novel algorithm for HEP that exploits an ensemble of link predictors and that provides a level of statistical significance for the entanglement between each node and hyperedge that does not have a direct interaction in the network.20 We then performed the HEP analysis to quantify the significance of the entanglement between PCSK9 and the hyperedge of 24 differentially expressed transcripts (FC>2.0, q<0.05) in the dataset (Supplemental Figure 5A and 5B, Supplemental Table 3B). The resulting P values of 7/12 node-node link prediction methods (RA, CH2-L2, CH3-L2, and all 4 L3-based methods) established a significant association between PCSK9 and the hyperedge of the 24 differentially expressed transcripts (Figure 5C; Supplemental Table 2). These data suggest that any of the 24 differentially expressed transcripts have a high likelihood of directly interacting with PCSK9, enabling us to validate a direct interaction between PCSK9 and one of these 24 differentially expressed transcripts. To understand the biological relevance of these transcripts, we explored the network connectivity of these transcripts identified by HEP analysis using the STRING database (confidence interaction score ≥0.7).37 The network analysis showed a highly clustered network (average local clustering coefficient: 0.547) containing 24 nodes with 52 edges (expected number of edges=3), providing significantly more interactions than expected for a random set of genes of similar size (P value ≤1.0e−16). The k-means clustering (number of clusters=3) showed that one of these 3 clusters mainly consisted of the NF-κB signaling pathway based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (IL-1b, CXCL1 [chemokine (C-X-C motif) ligand 1], CXCL2 [chemokine (C-X-C motif) ligand 2], ICAM1, VCAM1 [vascular cell adhesion molecule 1], RELB [RELB Proto-Oncogene, NF-kB subunit], NFKB2 [nuclear factor kappa B subunit 2]), and the other 2 clusters included OLR1 (LOX-1) and SDC4, respectively (Figure 5D). It is already reported that NF-κB signaling pathway is involved in PCSK9-induced atherosclerotic inflammation.38 In the present study, AAV-PCSK9 promoted NF-κB p65 mRNA level in the peritoneal F4/80+ macrophages of Ldlr-/- mice (Supplemental Figure 6A). Inhibition of NF-κB signaling pathway with IκB kinase inhibitor, 2-[(aminocarbonyl)amino]-5-(4-fluorophenyl)-3-thiophenecarboxamide (TPCA1), decreased pro-inflammatory responses to recombinant PCSK9 in Ldlr-/- mouse macrophages (Supplemental Figure 6B). These data indicate that NF-κB may mediate PCSK9-induced pro-inflammatory responses in vein graft lesions. PCSK9 stimulates the expression of LOX-1, which in turn takes up ox-LDL in macrophages.39 AAV-PCSK9 increased foam cell formation determined by Oil red O positive area in vein grafts (Supplemental Figure 7A). We further found that recombinant PCSK9 increased ox-LDL uptake in bone marrow-derived macrophages from Ldlr-/- mice (Supplemental Figure 7B). These results indicate that PCSK9 also induces foam cell formation by increased ox-LDL uptake, contributing not only to atherosclerotic but also to vein graft lesion development. Among the members of this differentially expressed transcript hyperedge, we found that SDC4 included in the last cluster could be a novel target of PCSK9 (Figure 5D). While SDC4 was not a high-ranking molecule in the transcript ranking lists (30th in Supplemental Table 3A and 67th in Supplemental Table 3B), its fold change after stimulation with recombinant PCSK9 in Ldlr-/- mice macrophages was statistically significant (2.65, log2FC>1). AAV-PCSK9 also increased SDC4 mRNA level in the peritoneal F4/80+ macrophages of Ldlr-/- mice (Figure 6A). SDC4 is a heparan sulfate proteoglycan40 expressed on the surface of human macrophages.41 In addition, SDC4 mRNA is increased in M(LPS) but not M(IL-4/IL-13) or M(IL-10) in bone marrow-derived macrophages.42 A recent study reported that heparan sulfate proteoglycans physically bind to PCSK9 on the hepatocyte surface43 However, the investigators did not address the interaction between PCSK9 and any specific individual syndecans. Our in silico protein-protein docking analysis using HPEPDOCK, a web server for protein-protein docking based on a hierarchical algorithm, predicted binding between PCSK9 and SDC4 (Supplemental Figure 8A).44 We entered the pdb files of PCSK9 (PDB ID: 2PMW), LDLR (PDB ID: 1N7D), and SDC4 (PDB ID: 1EJP) into HPEPDOCK server, which in turn presented the top 100 docking models based on docking energy minimized scores. Protein-protein docking analysis showed that the binding efficiency of SDC4 and PCSK9 (−212.23 to −235.6 kJ/mol) was comparable to that of LDLR and PCSK9 (−259.85 to −303.21 kJ/mol) based on the docking score of the top 5 binding predictions (Supplemental Figure 8A). Besides, the ligand root mean square deviation, often used to evaluate the correctness of the docking geometry and optimal superimposition of the receptor-ligand binding, was lower in the binding of SDC4 and PCSK9 (39.12–85.16) compared with that of LDLR and PCSK9 (43.76–156.25) for the top 5 binding predictions, suggesting better binding between SDC4 and PCSK9 than in LDLR and PCSK9. Furthermore, co-immunoprecipitation analysis of mouse liver tissue lysates validated the occurrence of the predicted PCSK9-SDC4 binding in vivo (Supplemental Figure 8B). RNA silencing validated SDC4 (syndecan-4) as a potential target of PCSK9 (proprotein convertase subtilisin/kexin 9)-induced pro-inflammatory response in macrophages, identified via hyperedge entanglement prediction (HEP) prediction. A, SDC4 mRNA level was measured in murine peritoneal macrophages of Ldlr-/- mice 1 week after intravenous injection with AAV-LacZ or AAV-PCSK9 (n=7 per group). B, mRNA levels of SDC4, IL-1b, NLRP3, CD40, and ICAM1 were measured by RT-qPCR in mouse Ldlr-/- peritoneal macrophages after pretreatment with siSDC4 or siControl (Ctrl) for 48 hours and stimulation with 2.5 μg/mL recombinant mouse PCSK9 (MmPCSK9) for 3 hours (n=4–5). P value was calculated by 1-way ANOVA, followed by Bonferroni multiple comparison test. Error bars indicate ± SEM. Data are reported as mean ± SEM. We explored further LDLR-independent pro-inflammatory signaling pathways in an unbiased manner, using RNA-sequencing of Ldlr-/- mouse macrophages stimulated with recombinant PCSK9. When analyzing the differentially expressed transcripts (FC>2.0, q<0.001), we observed 46 transcripts significantly increased by PCSK9 stimulation in Ldlr-/- macrophages compared with controls (Figure 5A). A hierarchical heat map of Ldlr-/- mouse macrophages filtered by differentially expressed transcripts showed a clear separation between control (no stimulus) and PCSK9 group (Figure 5B). We provided statistically significantly overrepresented transcripts ranked according to their fold change abundance (Supplemental Table 3A) to further examine these differentially expressed transcripts. PCSK9 treatment increased many transcripts implicated in pro-inflammatory responses in Ldlr-/- macrophages (eg, IL-1b, TNF, CXCL2, NF-κB [nuclear factor-kappa B]-related genes).19 Transcriptomics, network analysis, and hyperedge entanglement prediction (HEP) of mouse macrophages stimulated with recombinant PCSK9 (proprotein convertase subtilisin/kexin 9). A, Volcano plot for Ldlr-/- mouse macrophages stimulated with recombinant PCSK9. Red markers indicate significantly expressed transcripts with a fold change cutoff of 2.0 and adjusted P value cutoff of 0.001. The enriched transcripts that were previously reported were denoted with their names. B, Heat map of the transcriptomics of Ldlr-/- mouse macrophages (FC>2, q<0.001-filtered data for the difference between macrophages stimulated with and without recombinant PCSK9, n=4 per group). C, The schematic network visualization by HEP prediction. The significantly predicted association of PCSK9 to the 24 mapped inflammation-related transcripts derived from the list of transcripts (FC>2.0, q<0.05) indicates every unknown pairs of interaction (dashed line) that have to be proven yet experimentally. One of them was validated by silencing Sdc4 (syndecan-4) mRNA (blue dashed line). All other links represent known physical interactions (solid gray lines). The node size illustrates the degree of the proteins in the protein-protein interaction network. D, Network analysis of the 24 overrepresented transcripts in Ldlr-/- mouse macrophages using STRING database. Nodes are colored according to k-means clustering (number of clusters=3). Only connected nodes are shown here. To provide additional mechanistic links, we attempted to identify PCSK9-regulated proteins other than LDLR that mediate the aforementioned pro-inflammatory downstream responses. Many cascading effects in the transcriptional regulatory machinery could control the differentially expressed mRNAs. This set of differentially expressed transcripts were identified as the result of a genome-wide transcriptional profiling method, which might not necessarily interact directly with PCSK9. To estimate the likelihood of PCSK9 directly interacting with those differentially expressed transcripts, we tested this hypothesis verification through network computational analysis. In brief, PCSK9 can be seen as a node in a protein-protein interaction network,36 and the set of differentially expressed transcripts can be seen as a hyperedge projected on the protein-protein interaction network (a group of nodes that are associated by a common feature of biological functions). Muscoloni et al recently introduced a novel algorithm for HEP that exploits an ensemble of link predictors and that provides a level of statistical significance for the entanglement between each node and hyperedge that does not have a direct interaction in the network.20 We then performed the HEP analysis to quantify the significance of the entanglement between PCSK9 and the hyperedge of 24 differentially expressed transcripts (FC>2.0, q<0.05) in the dataset (Supplemental Figure 5A and 5B, Supplemental Table 3B). The resulting P values of 7/12 node-node link prediction methods (RA, CH2-L2, CH3-L2, and all 4 L3-based methods) established a significant association between PCSK9 and the hyperedge of the 24 differentially expressed transcripts (Figure 5C; Supplemental Table 2). These data suggest that any of the 24 differentially expressed transcripts have a high likelihood of directly interacting with PCSK9, enabling us to validate a direct interaction between PCSK9 and one of these 24 differentially expressed transcripts. To understand the biological relevance of these transcripts, we explored the network connectivity of these transcripts identified by HEP analysis using the STRING database (confidence interaction score ≥0.7).37 The network analysis showed a highly clustered network (average local clustering coefficient: 0.547) containing 24 nodes with 52 edges (expected number of edges=3), providing significantly more interactions than expected for a random set of genes of similar size (P value ≤1.0e−16). The k-means clustering (number of clusters=3) showed that one of these 3 clusters mainly consisted of the NF-κB signaling pathway based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (IL-1b, CXCL1 [chemokine (C-X-C motif) ligand 1], CXCL2 [chemokine (C-X-C motif) ligand 2], ICAM1, VCAM1 [vascular cell adhesion molecule 1], RELB [RELB Proto-Oncogene, NF-kB subunit], NFKB2 [nuclear factor kappa B subunit 2]), and the other 2 clusters included OLR1 (LOX-1) and SDC4, respectively (Figure 5D). It is already reported that NF-κB signaling pathway is involved in PCSK9-induced atherosclerotic inflammation.38 In the present study, AAV-PCSK9 promoted NF-κB p65 mRNA level in the peritoneal F4/80+ macrophages of Ldlr-/- mice (Supplemental Figure 6A). Inhibition of NF-κB signaling pathway with IκB kinase inhibitor, 2-[(aminocarbonyl)amino]-5-(4-fluorophenyl)-3-thiophenecarboxamide (TPCA1), decreased pro-inflammatory responses to recombinant PCSK9 in Ldlr-/- mouse macrophages (Supplemental Figure 6B). These data indicate that NF-κB may mediate PCSK9-induced pro-inflammatory responses in vein graft lesions. PCSK9 stimulates the expression of LOX-1, which in turn takes up ox-LDL in macrophages.39 AAV-PCSK9 increased foam cell formation determined by Oil red O positive area in vein grafts (Supplemental Figure 7A). We further found that recombinant PCSK9 increased ox-LDL uptake in bone marrow-derived macrophages from Ldlr-/- mice (Supplemental Figure 7B). These results indicate that PCSK9 also induces foam cell formation by increased ox-LDL uptake, contributing not only to atherosclerotic but also to vein graft lesion development. Among the members of this differentially expressed transcript hyperedge, we found that SDC4 included in the last cluster could be a novel target of PCSK9 (Figure 5D). While SDC4 was not a high-ranking molecule in the transcript ranking lists (30th in Supplemental Table 3A and 67th in Supplemental Table 3B), its fold change after stimulation with recombinant PCSK9 in Ldlr-/- mice macrophages was statistically significant (2.65, log2FC>1). AAV-PCSK9 also increased SDC4 mRNA level in the peritoneal F4/80+ macrophages of Ldlr-/- mice (Figure 6A). SDC4 is a heparan sulfate proteoglycan40 expressed on the surface of human macrophages.41 In addition, SDC4 mRNA is increased in M(LPS) but not M(IL-4/IL-13) or M(IL-10) in bone marrow-derived macrophages.42 A recent study reported that heparan sulfate proteoglycans physically bind to PCSK9 on the hepatocyte surface43 However, the investigators did not address the interaction between PCSK9 and any specific individual syndecans. Our in silico protein-protein docking analysis using HPEPDOCK, a web server for protein-protein docking based on a hierarchical algorithm, predicted binding between PCSK9 and SDC4 (Supplemental Figure 8A).44 We entered the pdb files of PCSK9 (PDB ID: 2PMW), LDLR (PDB ID: 1N7D), and SDC4 (PDB ID: 1EJP) into HPEPDOCK server, which in turn presented the top 100 docking models based on docking energy minimized scores. Protein-protein docking analysis showed that the binding efficiency of SDC4 and PCSK9 (−212.23 to −235.6 kJ/mol) was comparable to that of LDLR and PCSK9 (−259.85 to −303.21 kJ/mol) based on the docking score of the top 5 binding predictions (Supplemental Figure 8A). Besides, the ligand root mean square deviation, often used to evaluate the correctness of the docking geometry and optimal superimposition of the receptor-ligand binding, was lower in the binding of SDC4 and PCSK9 (39.12–85.16) compared with that of LDLR and PCSK9 (43.76–156.25) for the top 5 binding predictions, suggesting better binding between SDC4 and PCSK9 than in LDLR and PCSK9. Furthermore, co-immunoprecipitation analysis of mouse liver tissue lysates validated the occurrence of the predicted PCSK9-SDC4 binding in vivo (Supplemental Figure 8B). RNA silencing validated SDC4 (syndecan-4) as a potential target of PCSK9 (proprotein convertase subtilisin/kexin 9)-induced pro-inflammatory response in macrophages, identified via hyperedge entanglement prediction (HEP) prediction. A, SDC4 mRNA level was measured in murine peritoneal macrophages of Ldlr-/- mice 1 week after intravenous injection with AAV-LacZ or AAV-PCSK9 (n=7 per group). B, mRNA levels of SDC4, IL-1b, NLRP3, CD40, and ICAM1 were measured by RT-qPCR in mouse Ldlr-/- peritoneal macrophages after pretreatment with siSDC4 or siControl (Ctrl) for 48 hours and stimulation with 2.5 μg/mL recombinant mouse PCSK9 (MmPCSK9) for 3 hours (n=4–5). P value was calculated by 1-way ANOVA, followed by Bonferroni multiple comparison test. Error bars indicate ± SEM. Data are reported as mean ± SEM. [SUBTITLE] SDC4 Silencing Suppresses PCSK9-Induced Pro-Inflammatory Responses in Macrophages [SUBSECTION] We further validated our prediction that SDC4 is a potential target of PCSK9 using siRNA silencing of mouse Sdc4 in mouse peritoneal macrophages. Silencing Sdc4 with siSDC4 treatment achieved 93% reduction of SDC4 mRNA expression (Figure 6B). siSDC4 was then used to determine whether it could suppress PCSK9-induced pro-inflammatory molecules, including those predicted to be linked with PCSK9 by the HEP prediction (Figure 5C). siSDC4 suppressed mRNAs of the pro-inflammatory molecules IL-1β, NLRP3, CD40, and ICAM1, but not TNFα, MCP-1, TLR2, CXCL1, CXCL2, or CXCL10 after stimulation with recombinant PCSK9 (Figure 6B, Supplemental Figure 9). These data suggest that SDC4 partially regulates a PCSK9-induced pro-inflammatory response. Furthermore, these mRNA silencing results substantiate the validity of the prediction, yielding the integration of PCSK9 in our hyperedge of 24 disconnected differentially expressed transcripts. We further validated our prediction that SDC4 is a potential target of PCSK9 using siRNA silencing of mouse Sdc4 in mouse peritoneal macrophages. Silencing Sdc4 with siSDC4 treatment achieved 93% reduction of SDC4 mRNA expression (Figure 6B). siSDC4 was then used to determine whether it could suppress PCSK9-induced pro-inflammatory molecules, including those predicted to be linked with PCSK9 by the HEP prediction (Figure 5C). siSDC4 suppressed mRNAs of the pro-inflammatory molecules IL-1β, NLRP3, CD40, and ICAM1, but not TNFα, MCP-1, TLR2, CXCL1, CXCL2, or CXCL10 after stimulation with recombinant PCSK9 (Figure 6B, Supplemental Figure 9). These data suggest that SDC4 partially regulates a PCSK9-induced pro-inflammatory response. Furthermore, these mRNA silencing results substantiate the validity of the prediction, yielding the integration of PCSK9 in our hyperedge of 24 disconnected differentially expressed transcripts. [SUBTITLE] Local Production of PCSK9 in the Liver Was Greater Than in Vein Graft Lesions [SUBSECTION] Our study demonstrated that circulating PCSK9 produced various changes in Ldlr-/- mice. We, however, attempted to examine the potential impact of local PCSK9 production in vein grafts. Immunohistochemical analysis using an anti-PCSK9 antibody in previous studies showed that vascular smooth muscle cells in human atherosclerotic plaque and collar-induced neointima of murine carotid artery express PCSK9.45,46 Based on these results, we examined whether locally produced PCSK9 in vein grafts may also contribute to vein graft lesion development. The local expression of PCSK9, as determined by in situ hybridization in the intima of vein grafts, was minimal compared with that in the liver (Figure 1B; Supplemental Figure 10A). RT-qPCR analysis further confirmed numerically significantly less PCSK9 mRNA expression in vein grafts than in the liver, the primary source of circulating PCSK9 (Supplemental Figure 10B). Moreover, we examined which cells within vein grafts can produce PCSK9 in humans by comparing PCSK9 mRNA levels in human saphenous vein endothelial cells, smooth muscle cells, and human primary macrophages derived from peripheral blood mononuclear cells. While we detected substantial PCSK9 expression in human saphenous vein smooth muscle cells, the level was significantly lower than that in HepG2 (liver) cells (Supplemental Figure 10C). Human primary macrophages and human saphenous vein endothelial cells contained minimal PCSK9 mRNA (Supplemental Figure 10C). Recent studies reported a modest increase of PCSK9 mRNA and protein levels in macrophages exposed to pro-inflammatory stimuli.39,47 We, therefore, examined PCSK9 mRNA expression in human primary macrophages treated with pro-inflammatory stimuli. Although LPS and IFNγ induced a pro-inflammatory response in the macrophages, as determined by TNFα mRNA expression (Supplemental Figures 10D and 10E, right graphs), PCSK9 mRNA expression did not increase after LPS or IFNγ (Supplemental Figures 10D and 10E, left graphs). These results indicate that smooth muscle cells (SMC), but not macrophages, may primarily produce PCSK9 in vein grafts, but to a much lower extent than the liver. Our study demonstrated that circulating PCSK9 produced various changes in Ldlr-/- mice. We, however, attempted to examine the potential impact of local PCSK9 production in vein grafts. Immunohistochemical analysis using an anti-PCSK9 antibody in previous studies showed that vascular smooth muscle cells in human atherosclerotic plaque and collar-induced neointima of murine carotid artery express PCSK9.45,46 Based on these results, we examined whether locally produced PCSK9 in vein grafts may also contribute to vein graft lesion development. The local expression of PCSK9, as determined by in situ hybridization in the intima of vein grafts, was minimal compared with that in the liver (Figure 1B; Supplemental Figure 10A). RT-qPCR analysis further confirmed numerically significantly less PCSK9 mRNA expression in vein grafts than in the liver, the primary source of circulating PCSK9 (Supplemental Figure 10B). Moreover, we examined which cells within vein grafts can produce PCSK9 in humans by comparing PCSK9 mRNA levels in human saphenous vein endothelial cells, smooth muscle cells, and human primary macrophages derived from peripheral blood mononuclear cells. While we detected substantial PCSK9 expression in human saphenous vein smooth muscle cells, the level was significantly lower than that in HepG2 (liver) cells (Supplemental Figure 10C). Human primary macrophages and human saphenous vein endothelial cells contained minimal PCSK9 mRNA (Supplemental Figure 10C). Recent studies reported a modest increase of PCSK9 mRNA and protein levels in macrophages exposed to pro-inflammatory stimuli.39,47 We, therefore, examined PCSK9 mRNA expression in human primary macrophages treated with pro-inflammatory stimuli. Although LPS and IFNγ induced a pro-inflammatory response in the macrophages, as determined by TNFα mRNA expression (Supplemental Figures 10D and 10E, right graphs), PCSK9 mRNA expression did not increase after LPS or IFNγ (Supplemental Figures 10D and 10E, left graphs). These results indicate that smooth muscle cells (SMC), but not macrophages, may primarily produce PCSK9 in vein grafts, but to a much lower extent than the liver. [SUBTITLE] Macrophages Rather Than Endothelial Cells and SMCs May Mediate the Effects of PCSK9 on Vein Graft Lesion Development [SUBSECTION] Although our study focuses on macrophages, we examined the role of PCSK9 in the activation of endothelial cells and SMCs because other investigators reported potential interactions between PCSK9 and these cell types.46,48 Endothelial cell activation precedes vein graft lesion development.49 Due to the scarcity of mRNA in CD31-positive endothelial cells in vein grafts, we sorted them from the liver and lungs of Ldlr-/- mice injected with AAV-PCSK9 instead to examine the effects of PCSK9 on endothelial cell activation in an LDLR-independent fashion. AAV-PCSK9 did not increase mRNA levels of adhesion molecules, such as VCAM-1, ICAM-1, and E-selectin, in EC from either organ when compared with AAV-LacZ (Supplemental Figure 11A). We further found in vitro that recombinant PCSK9 did not increase mRNA levels of these adhesion molecules in human saphenous vein endothelial cells (Supplemental Figure 11B). These results indirectly indicate that PCSK9 may not induce endothelial activation in vein grafts. SMC modulation, migration, and proliferation contribute to the development of vascular disorders, including atherosclerosis50 and vein graft.51 A recent study showed that PCSK9 may sustain SMC dedifferentiation, migration, and proliferation in neointimal hyperplasia in response to vascular injury,46 suggesting the involvement of SMCs in PCSK9-induced vein graft lesion development. To explore this possibility, we examined the effects of recombinant PCSK9 on SMC dedifferentiation, migration, and proliferation in human saphenous vein smooth muscle cells. Recombinant PCSK9 slightly decreased SM22α mRNA levels (0.74 and 0.68-fold for 1.0 and 5.0 µg/mL, respectively), but did not affect differentiation molecules, such as αSMA, calponin, SM1, or SM2 (Supplemental Figure 12A). In addition, recombinant PCSK9 did not affect PDGF-induced migration activity of human saphenous vein smooth muscle cells (Supplemental Figure 12B). A BrdU assay using human saphenous vein smooth muscle cells showed that recombinant PCSK9 did not enhance 10% fetal bovine serum-induced proliferation (Supplemental Figure 12C). AAV-PCSK9 did not affect the SMC content in vein grafts (Supplemental Figure 12D). Collectively, in our experimental settings, our data did not provide the mechanistic evidence in vitro and in vivo that activation of endothelial cells or SMCs contribute to PCSK9-induced vein graft lesion formation. These results indicate that circulating PCSK9 promotes vein graft lesion development predominantly through macrophage activation. Although our study focuses on macrophages, we examined the role of PCSK9 in the activation of endothelial cells and SMCs because other investigators reported potential interactions between PCSK9 and these cell types.46,48 Endothelial cell activation precedes vein graft lesion development.49 Due to the scarcity of mRNA in CD31-positive endothelial cells in vein grafts, we sorted them from the liver and lungs of Ldlr-/- mice injected with AAV-PCSK9 instead to examine the effects of PCSK9 on endothelial cell activation in an LDLR-independent fashion. AAV-PCSK9 did not increase mRNA levels of adhesion molecules, such as VCAM-1, ICAM-1, and E-selectin, in EC from either organ when compared with AAV-LacZ (Supplemental Figure 11A). We further found in vitro that recombinant PCSK9 did not increase mRNA levels of these adhesion molecules in human saphenous vein endothelial cells (Supplemental Figure 11B). These results indirectly indicate that PCSK9 may not induce endothelial activation in vein grafts. SMC modulation, migration, and proliferation contribute to the development of vascular disorders, including atherosclerosis50 and vein graft.51 A recent study showed that PCSK9 may sustain SMC dedifferentiation, migration, and proliferation in neointimal hyperplasia in response to vascular injury,46 suggesting the involvement of SMCs in PCSK9-induced vein graft lesion development. To explore this possibility, we examined the effects of recombinant PCSK9 on SMC dedifferentiation, migration, and proliferation in human saphenous vein smooth muscle cells. Recombinant PCSK9 slightly decreased SM22α mRNA levels (0.74 and 0.68-fold for 1.0 and 5.0 µg/mL, respectively), but did not affect differentiation molecules, such as αSMA, calponin, SM1, or SM2 (Supplemental Figure 12A). In addition, recombinant PCSK9 did not affect PDGF-induced migration activity of human saphenous vein smooth muscle cells (Supplemental Figure 12B). A BrdU assay using human saphenous vein smooth muscle cells showed that recombinant PCSK9 did not enhance 10% fetal bovine serum-induced proliferation (Supplemental Figure 12C). AAV-PCSK9 did not affect the SMC content in vein grafts (Supplemental Figure 12D). Collectively, in our experimental settings, our data did not provide the mechanistic evidence in vitro and in vivo that activation of endothelial cells or SMCs contribute to PCSK9-induced vein graft lesion formation. These results indicate that circulating PCSK9 promotes vein graft lesion development predominantly through macrophage activation.
null
null
[ "What Is Known?", "What New Information Does This Article Contribute?", "Methods", "Culture and Stimulation of Mouse Primary Macrophages", "Transcriptomics", "Network-Based Hyperedge Entanglement Prediction Analysis", "Statistics", "AAV-PCSK9 Promotes Vein Graft Lesion Development in an LDLR-Independent Manner", "AAV-PCSK9 Increases Thin Collagen Fibers and Macrophage Accumulation in Vein Graft Lesions", "AAV-PCSK9 Induces Macrophage Activation via an LDLR-Independent Route In Vivo", "Endotoxin-Free Recombinant PCSK9 Induced Macrophage Activation in Peritoneal Macrophages of Ldlr\n\n-/- Mice In Vitro", "A combination of Unbiased Global Transcriptomics and Network-Based Hyperedge Entanglement Prediction Analysis Revealed Potential Targets of PCSK9 in Macrophages", "SDC4 Silencing Suppresses PCSK9-Induced Pro-Inflammatory Responses in Macrophages", "Local Production of PCSK9 in the Liver Was Greater Than in Vein Graft Lesions", "Macrophages Rather Than Endothelial Cells and SMCs May Mediate the Effects of PCSK9 on Vein Graft Lesion Development", "Article Information", "Acknowledgments", "Sources of Funding", "Supplemental Materials" ]
[ "Autologous saphenous vein is a widely used surgical bypass for lower extremity peripheral artery disease and coronary artery disease.\nPCSK9 (proprotein convertase subtilisin/kexin 9) targets receptors other than LDL receptor (LDLR) such as LRP1 (low-density lipoprotein receptor-related protein-1), ApoER2 (apolipoprotein E receptor 2), VLDLR (very low-density lipoprotein receptor), and CD36.\nPCSK9 may induce pro-inflammatory responses in macrophages and arterial atherosclerotic lesions primarily via LDLR-dependent mechanisms.", "PCSK9 promotes vein graft lesion development by the mechanisms unassociated with either LDLR degradation or blood cholesterol levels.\nPCSK9 induces pro-inflammatory macrophage activation: immune responses, proliferation and migration, via LDLR-independent mechanisms.\nPotential PCSK9 targets mediating pro-inflammatory responses in macrophages include NF-κB (nuclear factor-kappa B) signaling molecules, lectin-like oxidized LOX-1 (LDL receptor-1), and SDC4 (syndecan-4).\nThe role of PCSK9 on vascular inflammation remains controversial in the clinical setting because clinical trials with PCSK9 inhibitors did not show any significant changes in high sensitivity-CRP (C-reactive protein) levels in patients with coronary artery disease. However, CRP may not solely capture changes in inflammation. Although there are a few emerging in vivo studies suggesting a direct link between PCSK9 and atherosclerosis primarily via LDLR-dependent mechanisms, the role of PCSK9 in vein graft failure, especially via LDLR-independent mechanisms, remains largely unknown. To test the hypothesis, we developed a systems approach, involving unbiased transcriptomics and network analysis. Our in vivo evidence suggests that PCSK9 promotes vein graft lesion development, macrophage proliferation, monocyte migration, foam cell formation, and collagen remodeling. We further identified NF-κB signaling molecules, LOX-1 and SDC4 as potential targets of PCSK9 mediating pro-inflammatory responses in macrophages. The evidence, including our own, has suggested that circulating PCSK9 may exert pro-inflammatory effects on vascular lesions.", "[SUBTITLE] Data Availability [SUBSECTION] The online-only Supplemental Data provided detailed methods of all procedures. The data, analytic methods, and study materials will be made available to other researchers, upon request for purposes of reproducing the results or replicating the procedure.\nThe online-only Supplemental Data provided detailed methods of all procedures. The data, analytic methods, and study materials will be made available to other researchers, upon request for purposes of reproducing the results or replicating the procedure.\n[SUBTITLE] Animal Procedures [SUBSECTION] All animal experiments were approved by the Brigham and Women’s Hospital’s Animal Welfare Assurance (protocol 2016N000219). Male Ldlr-/- mice were fed a high-fat diet (1.25% cholesterol, D12108C, Research Diets, Inc., New Brunswick, NJ). To create experimental vein grafts in Ldlr-/- mice, inferior vena cava were harvested from a donor mouse and implanted into the carotid artery of recipient mice using cuff technique, as previously described.8,21 To induce liver-specific gain-of-function mutant PCSK9, we administered adeno-associated virus (AAV) encoding a gain-of-function form of mouse PCSK9 (AAV-PCSK9, 1×1011 vg, i.v.).22 We constructed AAV encoding LacZ (AAV-LacZ) that we used as a control for AAV-PCSK9. Four weeks after implantation, grafts were evaluated. After ultrasonography and in vivo molecular imaging of vein grafts, vein graft tissues were harvested, sectioned, and used for Masson trichrome staining, picrosirius red staining, immunohistochemistry, and in situ hybridization.\nAll animal experiments were approved by the Brigham and Women’s Hospital’s Animal Welfare Assurance (protocol 2016N000219). Male Ldlr-/- mice were fed a high-fat diet (1.25% cholesterol, D12108C, Research Diets, Inc., New Brunswick, NJ). To create experimental vein grafts in Ldlr-/- mice, inferior vena cava were harvested from a donor mouse and implanted into the carotid artery of recipient mice using cuff technique, as previously described.8,21 To induce liver-specific gain-of-function mutant PCSK9, we administered adeno-associated virus (AAV) encoding a gain-of-function form of mouse PCSK9 (AAV-PCSK9, 1×1011 vg, i.v.).22 We constructed AAV encoding LacZ (AAV-LacZ) that we used as a control for AAV-PCSK9. Four weeks after implantation, grafts were evaluated. After ultrasonography and in vivo molecular imaging of vein grafts, vein graft tissues were harvested, sectioned, and used for Masson trichrome staining, picrosirius red staining, immunohistochemistry, and in situ hybridization.\n[SUBTITLE] Culture and Stimulation of Mouse Primary Macrophages [SUBSECTION] Mouse peritoneal macrophages cultured with RPMI 1640 containing 10% fetal bovine serum were prepared as previously described.8 Briefly, 4% Brewer thioglycolate medium (BD Diagnostic Systems) was injected into the peritoneal cavity of mice 4 days before macrophage collection. After euthanasia, 10 mL of ice-cold phosphate-buffered saline was injected into the peritoneal cavity, and cells were harvested. The cells were then washed with phosphate-buffered saline once and plated for further experiments. Overnight starvation was performed with 0.5% fetal bovine serum before each experiment. Endotoxin-free recombinant mouse PCSK9 derived from mouse myeloma cell line NS0 was used to stimulate mouse primary macrophages (R&D Systems).\nMouse peritoneal macrophages cultured with RPMI 1640 containing 10% fetal bovine serum were prepared as previously described.8 Briefly, 4% Brewer thioglycolate medium (BD Diagnostic Systems) was injected into the peritoneal cavity of mice 4 days before macrophage collection. After euthanasia, 10 mL of ice-cold phosphate-buffered saline was injected into the peritoneal cavity, and cells were harvested. The cells were then washed with phosphate-buffered saline once and plated for further experiments. Overnight starvation was performed with 0.5% fetal bovine serum before each experiment. Endotoxin-free recombinant mouse PCSK9 derived from mouse myeloma cell line NS0 was used to stimulate mouse primary macrophages (R&D Systems).\n[SUBTITLE] Transcriptomics [SUBSECTION] RNA-sequencing—mRNAseq (polyA enriched) library prep, single-end 75bp sequencing on NextSeq of 12-16 pooled barcoded samples, and VIPER analysis23—was performed at the Molecular Biology Core Facilities at Dana-Farber Cancer Institute. The data were then analyzed using DESeq2 and Qlucore (http://www.qlucore.com/) to perform a 2-group comparison and identify differentially expressed transcripts (increase/decrease by 2-fold) between control and PCSK9 groups. STRING (version 11.5) was used to evaluate known and predicted transcript interconnectivity. The differentially expressed transcripts (increase/decrease by 2-fold, q<0.05), which were compared with the “inflammatome”24 dataset, were used for the following network analysis.\nRNA-sequencing—mRNAseq (polyA enriched) library prep, single-end 75bp sequencing on NextSeq of 12-16 pooled barcoded samples, and VIPER analysis23—was performed at the Molecular Biology Core Facilities at Dana-Farber Cancer Institute. The data were then analyzed using DESeq2 and Qlucore (http://www.qlucore.com/) to perform a 2-group comparison and identify differentially expressed transcripts (increase/decrease by 2-fold) between control and PCSK9 groups. STRING (version 11.5) was used to evaluate known and predicted transcript interconnectivity. The differentially expressed transcripts (increase/decrease by 2-fold, q<0.05), which were compared with the “inflammatome”24 dataset, were used for the following network analysis.\n[SUBTITLE] Network-Based Hyperedge Entanglement Prediction Analysis [SUBSECTION] Network-Based Hyperedge Entanglement Prediction (HEP) algorithm20 was performed to estimate the likelihood of PCSK9 directly interacting with any of the differentially expressed transcripts in the “inflammatome.” Given only a network topology and its high-order hypergraph organization, the HEP algorithm uses an ensemble of link predictors to calculate a level of statistical significance for each candidate node to hyperedge entanglement that is characterized by not having a direct connection. The link predictor is an operator that, exploiting topological information of the graph, can associate a similarity score to any disconnected node pair, suggesting the likelihood for a link between them to exist. Several link predictors perform under the evidence that 2 nodes are more likely to be linked if their common neighbors are members of a strongly inner-linked cohort, named a local community,25 and recent studies demonstrated that this growth mechanism emerges mainly considering missing links between nonadjacent nodes distanced by paths of length 2 (L2) or 3 (L3)26 following a scheme that can be well described by the Cannistraci-Hebb network automata modeling.27 The HEP analysis provides 12 different estimations of the P value that quantifies the significance of the entanglement (association) between each node and hyperedge (also called node2hyperedge entanglement) using the 12 available node-node link prediction variants.25, 27 In this study, we applied HEP algorithm according to the following specifications: the scaling of the similarity scores was not necessary, and the average operator to estimate the node2hyperedge entanglement scores was the mean.\nNetwork-Based Hyperedge Entanglement Prediction (HEP) algorithm20 was performed to estimate the likelihood of PCSK9 directly interacting with any of the differentially expressed transcripts in the “inflammatome.” Given only a network topology and its high-order hypergraph organization, the HEP algorithm uses an ensemble of link predictors to calculate a level of statistical significance for each candidate node to hyperedge entanglement that is characterized by not having a direct connection. The link predictor is an operator that, exploiting topological information of the graph, can associate a similarity score to any disconnected node pair, suggesting the likelihood for a link between them to exist. Several link predictors perform under the evidence that 2 nodes are more likely to be linked if their common neighbors are members of a strongly inner-linked cohort, named a local community,25 and recent studies demonstrated that this growth mechanism emerges mainly considering missing links between nonadjacent nodes distanced by paths of length 2 (L2) or 3 (L3)26 following a scheme that can be well described by the Cannistraci-Hebb network automata modeling.27 The HEP analysis provides 12 different estimations of the P value that quantifies the significance of the entanglement (association) between each node and hyperedge (also called node2hyperedge entanglement) using the 12 available node-node link prediction variants.25, 27 In this study, we applied HEP algorithm according to the following specifications: the scaling of the similarity scores was not necessary, and the average operator to estimate the node2hyperedge entanglement scores was the mean.\n[SUBTITLE] Statistics [SUBSECTION] Statistical analyses were performed using GraphPad Prism 8.0 (GraphPad Software). For experiments with a small sample size (n<6), P values were determined by nonparametric analysis except for RNA-sequencing analysis. Other data were examined for normality before analysis by Shapiro-Wilk test. For all normally distributed data, an unpaired 2-tailed Student t test was used for comparisons between 2 groups, 1-way ANOVA followed by Bonferroni or Dunnett post-hoc test was used to compare multiple groups. When there were 2 experimental factors, 2-way ANOVA for comparisons between multiple groups was used. If the data were not normally distributed, nonparametric unpaired 2-tailed Mann-Whitney U test was used to compare 2 groups, and Kruskal-Wallis test with Dunn post-hoc test was used to compare multiple groups. Wald Test in DESeq2 was used for bulk RNA-sequencing analysis. Data are expressed as mean ± SEM for continuous variables. P values of <0.05 were considered statistically significant.\nStatistical analyses were performed using GraphPad Prism 8.0 (GraphPad Software). For experiments with a small sample size (n<6), P values were determined by nonparametric analysis except for RNA-sequencing analysis. Other data were examined for normality before analysis by Shapiro-Wilk test. For all normally distributed data, an unpaired 2-tailed Student t test was used for comparisons between 2 groups, 1-way ANOVA followed by Bonferroni or Dunnett post-hoc test was used to compare multiple groups. When there were 2 experimental factors, 2-way ANOVA for comparisons between multiple groups was used. If the data were not normally distributed, nonparametric unpaired 2-tailed Mann-Whitney U test was used to compare 2 groups, and Kruskal-Wallis test with Dunn post-hoc test was used to compare multiple groups. Wald Test in DESeq2 was used for bulk RNA-sequencing analysis. Data are expressed as mean ± SEM for continuous variables. P values of <0.05 were considered statistically significant.", "Mouse peritoneal macrophages cultured with RPMI 1640 containing 10% fetal bovine serum were prepared as previously described.8 Briefly, 4% Brewer thioglycolate medium (BD Diagnostic Systems) was injected into the peritoneal cavity of mice 4 days before macrophage collection. After euthanasia, 10 mL of ice-cold phosphate-buffered saline was injected into the peritoneal cavity, and cells were harvested. The cells were then washed with phosphate-buffered saline once and plated for further experiments. Overnight starvation was performed with 0.5% fetal bovine serum before each experiment. Endotoxin-free recombinant mouse PCSK9 derived from mouse myeloma cell line NS0 was used to stimulate mouse primary macrophages (R&D Systems).", "RNA-sequencing—mRNAseq (polyA enriched) library prep, single-end 75bp sequencing on NextSeq of 12-16 pooled barcoded samples, and VIPER analysis23—was performed at the Molecular Biology Core Facilities at Dana-Farber Cancer Institute. The data were then analyzed using DESeq2 and Qlucore (http://www.qlucore.com/) to perform a 2-group comparison and identify differentially expressed transcripts (increase/decrease by 2-fold) between control and PCSK9 groups. STRING (version 11.5) was used to evaluate known and predicted transcript interconnectivity. The differentially expressed transcripts (increase/decrease by 2-fold, q<0.05), which were compared with the “inflammatome”24 dataset, were used for the following network analysis.", "Network-Based Hyperedge Entanglement Prediction (HEP) algorithm20 was performed to estimate the likelihood of PCSK9 directly interacting with any of the differentially expressed transcripts in the “inflammatome.” Given only a network topology and its high-order hypergraph organization, the HEP algorithm uses an ensemble of link predictors to calculate a level of statistical significance for each candidate node to hyperedge entanglement that is characterized by not having a direct connection. The link predictor is an operator that, exploiting topological information of the graph, can associate a similarity score to any disconnected node pair, suggesting the likelihood for a link between them to exist. Several link predictors perform under the evidence that 2 nodes are more likely to be linked if their common neighbors are members of a strongly inner-linked cohort, named a local community,25 and recent studies demonstrated that this growth mechanism emerges mainly considering missing links between nonadjacent nodes distanced by paths of length 2 (L2) or 3 (L3)26 following a scheme that can be well described by the Cannistraci-Hebb network automata modeling.27 The HEP analysis provides 12 different estimations of the P value that quantifies the significance of the entanglement (association) between each node and hyperedge (also called node2hyperedge entanglement) using the 12 available node-node link prediction variants.25, 27 In this study, we applied HEP algorithm according to the following specifications: the scaling of the similarity scores was not necessary, and the average operator to estimate the node2hyperedge entanglement scores was the mean.", "Statistical analyses were performed using GraphPad Prism 8.0 (GraphPad Software). For experiments with a small sample size (n<6), P values were determined by nonparametric analysis except for RNA-sequencing analysis. Other data were examined for normality before analysis by Shapiro-Wilk test. For all normally distributed data, an unpaired 2-tailed Student t test was used for comparisons between 2 groups, 1-way ANOVA followed by Bonferroni or Dunnett post-hoc test was used to compare multiple groups. When there were 2 experimental factors, 2-way ANOVA for comparisons between multiple groups was used. If the data were not normally distributed, nonparametric unpaired 2-tailed Mann-Whitney U test was used to compare 2 groups, and Kruskal-Wallis test with Dunn post-hoc test was used to compare multiple groups. Wald Test in DESeq2 was used for bulk RNA-sequencing analysis. Data are expressed as mean ± SEM for continuous variables. P values of <0.05 were considered statistically significant.", "In vivo studies have suggested a direct link between PCSK9 and atherosclerosis28,29; however, no evidence has yet implicated PCSK9 in vein graft lesion development. Furthermore, these in vivo studies indicate that the underlying mechanism of atherosclerotic lesion development by PCSK9 may depend mainly on LDLR. Whether PCSK9 can exert pro-inflammatory effects via LDLR-independent mechanisms is unclear. To test the hypothesis that circulating PCSK9 promotes vein graft lesion development in vivo, we used experimental vein grafts in mice.21 A single intravenous injection of AAV-PCSK9 induced liver production of PCSK9. To exclude potential systemic toxicity of AAV, AAV-LacZ was developed and used as a control (Figure 1A; Online Supplemental Methods). All experiments used Ldlr-/- mice to explore LDLR-independent mechanisms unless otherwise noted.\nThe effects of Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) on vein graft lesion development in Ldlr-/- mice. A, An experimental protocol for the treatments in Ldlr-/- mice. B, In situ hybridization of PCSK9 (red) in the liver of Ldlr-/- mice 1 week after intravenous injection with AAV-LacZ or AAV-PCSK9. Nuclei are visualized with DAPI. The data represent 4 mice per group. Scale bars indicate 100 μm. C, PCSK9 mRNA level in the liver of AAV treated Ldlr-/- mice (n=12 and 11 for AAV-LacZ and AAV-PCSK9 groups, respectively), and serum levels of PCSK9, total cholesterol, and triglyceride in AAV treated Ldlr-/- mice (n=11 per group). D, Left, ultrasonography images of vein grafts from Ldlr-/- mice treated with AAV-LacZ or AAV-PCSK9 28 days after implantation. The red, green, and yellow dotted lines indicate lumen, near wall, and far wall, respectively. Right, The quantitative analysis of lumen and vessel wall area in short axis view and 3D reconstructed vessel wall volume of vein grafts by ultrasonography (n=12 and 11 for AAV-LacZ and AAV-PCSK9 groups, respectively). E, Left, Masson’s trichrome staining of vein grafts from Ldlr-/- mice treated with AAV-LacZ or AAV-PCSK9 28 days after implantation. Scale bars indicate 500 μm. Right, The quantitative analysis of intimal area, intimal and media/adventitia thickness, and lumen and vessel diameter of vein grafts (n=12 and 11 for AAV-LacZ and AAV-PCSK9 group, respectively). P value was calculated by Mann-Whitney U-test (triglyceride in C, media/adventitia thickness, lumen diameter, and vessel diameter in E) and unpaired Student t test (A, B, and D). Data are reported as mean ± SEM.\nBody weight and blood pressure did not differ significantly between the AAV-LacZ and AAV-PCSK9 groups 4 weeks after vein graft implantation (Supplemental Table 1). In situ hybridization showed that AAV-PCSK9 markedly increased PCSK9 mRNA levels in the liver (Figure 1B). AAV-PCSK9 increased PCSK9 mRNA expression in the liver and serum levels of PCSK9 (Figure 1C; Supplemental Table 1). Serum levels of total cholesterol and triglycerides did not differ between the AAV-LacZ and AAV-PCSK9 groups (Figure 1C; Supplemental Table 1). These findings indicate that the effects of AAV-PCSK9 on vein graft lesions described below did not depend on blood cholesterol and triglyceride concentrations (Supplemental Table 1).\nNoninvasive ultrasonography visualized that AAV-PCSK9 injection caused increased vessel wall area, but not lumen area, in the short axis view and 3-dimensional reconstructed vessel wall volume of vein grafts (Figure 1D). Consistent with the ultrasonography, histological analysis by Masson trichrome staining showed that AAV-PCSK9 increased intimal area and thickness of vein grafts. Lumen diameter, vessel diameter, and media/adventitia thickness did not differ significantly, suggesting compensatory outward remodeling of vein grafts (Figure 1E).", "Evidence suggests that vein grafts in humans can develop lesions similar to those of advanced arterial atherosclerosis, and plaque rupture can occur in inflamed vein grafts.30,31 Therefore, exploring vein graft lesion composition in depth is critical to understanding what contributes to this occurrence. We then examined the collagen content of vein grafts by picrosirius red staining. AAV-PCSK9 increased the percentage of thin collagen fibers as a ratio of the total collagen fibers in the vein grafts, suggesting that circulating PCSK9 induced pathological features similar to those in thin-capped atherosclerotic plaques (Figure 2A). In vivo molecular imaging further provided insight into the effects of circulating PCSK9 on matrix metalloproteinase activity and macrophage accumulation in vein grafts. We intravenously co-injected 2 spectrally different imaging agents that elaborate near-infrared signals for visualization of matrix metalloproteinase activity (MMPsense, 680 nm) and macrophage accumulation (AminoSPARK, 750 nm). Following the evidence that activated macrophages express MMPs that degrade interstitial collagen as previously reported by our group,32,33 in vivo molecular imaging demonstrated that increased matrix metalloproteinase activity co-localized with accumulated macrophages (Figure 2B, left). In addition, AAV-PCSK9 treatment promoted matrix metalloproteinase activity and macrophage accumulation in vein grafts (Figure 2B), suggesting a potential mechanism of collagen remodeling as determined by Picrosirius red staining. Immunohistochemical analysis further demonstrated increased macrophage accumulation in vein grafts by AAV-PCSK9 (Mac3; Figure 2C). Macrophage proliferation and migration may contribute to the mechanisms of macrophage accumulation in vascular diseases.34 Supporting this mechanism, AAV-PCSK9 increased the percentage of macrophages positive for Ki-67, an indicator of proliferating cells (Supplemental Figure 1A, B). Consistent with this in vivo finding, recombinant PCSK9 augmented M-CSF-induced cell proliferation in mouse bone marrow-derived macrophages (BMDMs) (Supplemental Figure 2A, B). Monocyte migration assay using the human monocytic cell line THP-1 showed that recombinant PCSK9 tended to promote MCP-1 (monocyte chemoattractant protein-1)-induced chemotaxis, suggesting lipid-independent effects of PCSK9 on monocyte migration (Supplemental Figure 2C). On the other hand, AAV-PCSK9 did not increase apoptosis determined by TUNEL staining in vein grafts of Ldlr-/- mice (Supplemental Figure 3A). Accordingly, AAV-PCSK9 did not change necrotic core area in the vein grafts (Supplemental Figure 3B).\nThe effects of Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) on collagen thinning and macrophage accumulation in vein graft lesions of Ldlr-/- mice. A, Picrosirius red staining of vein grafts without (top) or with (bottom) polarized light (n=12 and 11 for AAV-LacZ and AAV-PCSK9 group, respectively). Scale bars indicate 500 μm. Circle graphs show the percentage of thin (green) and thick (red) collagen fibers compared with total fibers. B, Intravital microscopy images of MMPSense 680 (red) and AminoSPARK750 (green) in vein grafts for visualization of MMP activity and macrophage accumulation, respectively (n=10 and 9 for AAV-LacZ and AAV-PCSK9 group, respectively). C, Mac3 (macrophages) staining in vein grafts. Scale bars indicate 500 μm. P value was calculated by Mann-Whitney U-test (thick collagen in A, macrophage accumulation mean fluorescence intensity in B) and unpaired Student t test (C). Data are reported as mean ± SEM.", "We previously reported that macrophage activation plays a key role in vein graft lesion development.8 To explore the effects of circulating PCSK9 on macrophage activation, we examined mRNA levels of pro-inflammatory (IL-1β [interleukin-1 beta], IL-6, TNFα, and MCP-1) and antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1 [transforming growth factor-beta 1], and IL-10) in the peritoneal F4/80+ macrophages of Ldlr-/- mice 1 or 4 weeks after intravenous injection of AAVs. AAV-PCSK9 promoted pro-inflammatory IL-1β, IL-6, TNFα, and MCP-1 mRNA levels at 1 week (Figure 3A). AAV-PCSK9 also suppressed Arginase-1 mRNA levels at 4 weeks when the increase in the mRNA levels of pro-inflammatory molecules at 1 week had subsided (Figure 3B). We have recently reported that predominant pathways of the vein grafts change over time during lesion development. In the same study, pathways with immune responses represent the early responsive proteins in proteomics analysis.9 These results indicate that PCSK9 can induce pro-inflammatory responses in macrophages initially and later impair resolution of inflammation in vivo.\nAdeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) induced macrophage activation in murine peritoneal macrophages via LDL receptor (LDLR)-independent mechanisms in vivo. mRNA levels of pro-inflammatory molecules (IL-1β, IL-6, TNFα, MCP-1) and antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1, IL-10) were measured in murine peritoneal macrophages of Ldlr-/- mice 1 week (A) or 4 weeks (B) after intravenous injection with AAV-LacZ or AAV-PCSK9 (n=7 to 8 per group). P value was calculated by Mann-Whitney U-test (TNFa in A, IL-1b, TNFa, MCP-1, Arginase-1, Ym1, and IL-10 in B) and unpaired Student t test. Data are reported as mean ± SEM.", "To support our in vivo evidence, we examined in vitro the effects of physiologically relevant levels of recombinant mouse PCSK9 (0.1–2.5 μg/mL)35 on the mRNA levels of the same pro-inflammatory and antiinflammatory molecules examined in Ldlr-/- mouse macrophages. First, we conducted a time-course study of recombinant PCSK9-induced TNFα and Arginase-1 mRNA expression levels to determine an optimal time point for harvesting Ldlr-/- mouse macrophages. The increase of TNFα mRNA levels peaked 3 hours after stimulation, whereas the decrease of Arginase-1 mRNA levels plateaued 12 hours after stimulation with recombinant PCSK9 (Figure 4A). For all experiments, Ldlr-/- mouse macrophages were therefore harvested 3 and 12 hours after stimulation to measure pro- and antiinflammatory molecules, respectively. Peritoneal macrophages treated with recombinant PCSK9 exhibited increased mRNA levels of IL-1β, IL-6, TNFα, and MCP-1 in a concentration-dependent manner (Figure 4B). In contrast, recombinant PCSK9 did not decrease the mRNA levels of antiinflammatory molecules (Figure 4C). These results support the in vivo evidence of macrophage activation by circulating PCSK9 in an LDLR-independent fashion.\nRecombinant PCSK9 (proprotein convertase subtilisin/kexin 9) induced macrophage activation in LDL receptor (LDLR)-independent mechanisms in vitro. A, Time course of TNFα and Arginase-1 mRNA levels in peritoneal macrophages of wild-type mice after stimulation with recombinant mouse PCSK9 (MmPCSK9; 2.5 μg/mL) (n=5 per group). P value was calculated by Kruskal-Wallis test, followed by Dunn multiple comparison test. B, mRNA levels of pro-inflammatory molecules (IL-1β, IL-6, TNFα, MCP-1) and (C) antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1, IL-10) were measured in murine peritoneal macrophages from Ldlr-/- mice 3 and 12 hours after stimulation with recombinant mouse PCSK9 for pro-inflammatory and antiinflammatory molecules, respectively (n=9 per group). P value was calculated by 1-way ANOVA, followed by Dunnett multiple comparison test (IL-6, TNFa, MCP-1‚ MRC1) or Kruskal-Wallis test, followed by Dunn multiple comparison test. Data are reported as mean ± SEM.\nEndotoxin contamination in recombinant proteins can induce pro-inflammatory responses. Therefore, we chose a recombinant protein derived from a mammalian system, mouse myeloma cell line (NS0). The endotoxin level of recombinant mouse PCSK9 used in this study was 0.00441 EU/µg or less, well below the threshold of 0.1 EU/µg required for accurate cell-based assays.19 To exclude further endotoxin contamination in the recombinant PCSK9 protein affecting any measurements, we performed the chromogenic Limulus amebocyte lysate endotoxin assay. The final endotoxin level of the maximum dose of the recombinant protein (2.5 µg/mL) was 0.005 EU/mL or less, which coincides with the endotoxin levels of the commercially available endotoxin-free medium. In addition, mass spectrometry conducted on the solvent derived from reconstituted recombinant mouse PCSK9 protein solution did not produce signals, suggesting that there were no small molecule impurities present (Supplemental Figure 4A). Furthermore, stimulation with heat-inactivated recombinant mouse PCSK9 protein (2.5 μg/mL) caused no changes in the mRNA levels of IL-1β, TNF-α, and MCP-1, demonstrating that endotoxin contamination did not drive the effects observed (Supplemental Figure 4B).", "We explored further LDLR-independent pro-inflammatory signaling pathways in an unbiased manner, using RNA-sequencing of Ldlr-/- mouse macrophages stimulated with recombinant PCSK9. When analyzing the differentially expressed transcripts (FC>2.0, q<0.001), we observed 46 transcripts significantly increased by PCSK9 stimulation in Ldlr-/- macrophages compared with controls (Figure 5A). A hierarchical heat map of Ldlr-/- mouse macrophages filtered by differentially expressed transcripts showed a clear separation between control (no stimulus) and PCSK9 group (Figure 5B). We provided statistically significantly overrepresented transcripts ranked according to their fold change abundance (Supplemental Table 3A) to further examine these differentially expressed transcripts. PCSK9 treatment increased many transcripts implicated in pro-inflammatory responses in Ldlr-/- macrophages (eg, IL-1b, TNF, CXCL2, NF-κB [nuclear factor-kappa B]-related genes).19\nTranscriptomics, network analysis, and hyperedge entanglement prediction (HEP) of mouse macrophages stimulated with recombinant PCSK9 (proprotein convertase subtilisin/kexin 9). A, Volcano plot for Ldlr-/- mouse macrophages stimulated with recombinant PCSK9. Red markers indicate significantly expressed transcripts with a fold change cutoff of 2.0 and adjusted P value cutoff of 0.001. The enriched transcripts that were previously reported were denoted with their names. B, Heat map of the transcriptomics of Ldlr-/- mouse macrophages (FC>2, q<0.001-filtered data for the difference between macrophages stimulated with and without recombinant PCSK9, n=4 per group). C, The schematic network visualization by HEP prediction. The significantly predicted association of PCSK9 to the 24 mapped inflammation-related transcripts derived from the list of transcripts (FC>2.0, q<0.05) indicates every unknown pairs of interaction (dashed line) that have to be proven yet experimentally. One of them was validated by silencing Sdc4 (syndecan-4) mRNA (blue dashed line). All other links represent known physical interactions (solid gray lines). The node size illustrates the degree of the proteins in the protein-protein interaction network. D, Network analysis of the 24 overrepresented transcripts in Ldlr-/- mouse macrophages using STRING database. Nodes are colored according to k-means clustering (number of clusters=3). Only connected nodes are shown here.\nTo provide additional mechanistic links, we attempted to identify PCSK9-regulated proteins other than LDLR that mediate the aforementioned pro-inflammatory downstream responses. Many cascading effects in the transcriptional regulatory machinery could control the differentially expressed mRNAs. This set of differentially expressed transcripts were identified as the result of a genome-wide transcriptional profiling method, which might not necessarily interact directly with PCSK9. To estimate the likelihood of PCSK9 directly interacting with those differentially expressed transcripts, we tested this hypothesis verification through network computational analysis. In brief, PCSK9 can be seen as a node in a protein-protein interaction network,36 and the set of differentially expressed transcripts can be seen as a hyperedge projected on the protein-protein interaction network (a group of nodes that are associated by a common feature of biological functions). Muscoloni et al recently introduced a novel algorithm for HEP that exploits an ensemble of link predictors and that provides a level of statistical significance for the entanglement between each node and hyperedge that does not have a direct interaction in the network.20 We then performed the HEP analysis to quantify the significance of the entanglement between PCSK9 and the hyperedge of 24 differentially expressed transcripts (FC>2.0, q<0.05) in the dataset (Supplemental Figure 5A and 5B, Supplemental Table 3B). The resulting P values of 7/12 node-node link prediction methods (RA, CH2-L2, CH3-L2, and all 4 L3-based methods) established a significant association between PCSK9 and the hyperedge of the 24 differentially expressed transcripts (Figure 5C; Supplemental Table 2). These data suggest that any of the 24 differentially expressed transcripts have a high likelihood of directly interacting with PCSK9, enabling us to validate a direct interaction between PCSK9 and one of these 24 differentially expressed transcripts. To understand the biological relevance of these transcripts, we explored the network connectivity of these transcripts identified by HEP analysis using the STRING database (confidence interaction score ≥0.7).37 The network analysis showed a highly clustered network (average local clustering coefficient: 0.547) containing 24 nodes with 52 edges (expected number of edges=3), providing significantly more interactions than expected for a random set of genes of similar size (P value ≤1.0e−16). The k-means clustering (number of clusters=3) showed that one of these 3 clusters mainly consisted of the NF-κB signaling pathway based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (IL-1b, CXCL1 [chemokine (C-X-C motif) ligand 1], CXCL2 [chemokine (C-X-C motif) ligand 2], ICAM1, VCAM1 [vascular cell adhesion molecule 1], RELB [RELB Proto-Oncogene, NF-kB subunit], NFKB2 [nuclear factor kappa B subunit 2]), and the other 2 clusters included OLR1 (LOX-1) and SDC4, respectively (Figure 5D).\nIt is already reported that NF-κB signaling pathway is involved in PCSK9-induced atherosclerotic inflammation.38 In the present study, AAV-PCSK9 promoted NF-κB p65 mRNA level in the peritoneal F4/80+ macrophages of Ldlr-/- mice (Supplemental Figure 6A). Inhibition of NF-κB signaling pathway with IκB kinase inhibitor, 2-[(aminocarbonyl)amino]-5-(4-fluorophenyl)-3-thiophenecarboxamide (TPCA1), decreased pro-inflammatory responses to recombinant PCSK9 in Ldlr-/- mouse macrophages (Supplemental Figure 6B). These data indicate that NF-κB may mediate PCSK9-induced pro-inflammatory responses in vein graft lesions.\nPCSK9 stimulates the expression of LOX-1, which in turn takes up ox-LDL in macrophages.39 AAV-PCSK9 increased foam cell formation determined by Oil red O positive area in vein grafts (Supplemental Figure 7A). We further found that recombinant PCSK9 increased ox-LDL uptake in bone marrow-derived macrophages from Ldlr-/- mice (Supplemental Figure 7B). These results indicate that PCSK9 also induces foam cell formation by increased ox-LDL uptake, contributing not only to atherosclerotic but also to vein graft lesion development.\nAmong the members of this differentially expressed transcript hyperedge, we found that SDC4 included in the last cluster could be a novel target of PCSK9 (Figure 5D). While SDC4 was not a high-ranking molecule in the transcript ranking lists (30th in Supplemental Table 3A and 67th in Supplemental Table 3B), its fold change after stimulation with recombinant PCSK9 in Ldlr-/- mice macrophages was statistically significant (2.65, log2FC>1). AAV-PCSK9 also increased SDC4 mRNA level in the peritoneal F4/80+ macrophages of Ldlr-/- mice (Figure 6A). SDC4 is a heparan sulfate proteoglycan40 expressed on the surface of human macrophages.41 In addition, SDC4 mRNA is increased in M(LPS) but not M(IL-4/IL-13) or M(IL-10) in bone marrow-derived macrophages.42 A recent study reported that heparan sulfate proteoglycans physically bind to PCSK9 on the hepatocyte surface43 However, the investigators did not address the interaction between PCSK9 and any specific individual syndecans. Our in silico protein-protein docking analysis using HPEPDOCK, a web server for protein-protein docking based on a hierarchical algorithm, predicted binding between PCSK9 and SDC4 (Supplemental Figure 8A).44 We entered the pdb files of PCSK9 (PDB ID: 2PMW), LDLR (PDB ID: 1N7D), and SDC4 (PDB ID: 1EJP) into HPEPDOCK server, which in turn presented the top 100 docking models based on docking energy minimized scores. Protein-protein docking analysis showed that the binding efficiency of SDC4 and PCSK9 (−212.23 to −235.6 kJ/mol) was comparable to that of LDLR and PCSK9 (−259.85 to −303.21 kJ/mol) based on the docking score of the top 5 binding predictions (Supplemental Figure 8A). Besides, the ligand root mean square deviation, often used to evaluate the correctness of the docking geometry and optimal superimposition of the receptor-ligand binding, was lower in the binding of SDC4 and PCSK9 (39.12–85.16) compared with that of LDLR and PCSK9 (43.76–156.25) for the top 5 binding predictions, suggesting better binding between SDC4 and PCSK9 than in LDLR and PCSK9. Furthermore, co-immunoprecipitation analysis of mouse liver tissue lysates validated the occurrence of the predicted PCSK9-SDC4 binding in vivo (Supplemental Figure 8B).\nRNA silencing validated SDC4 (syndecan-4) as a potential target of PCSK9 (proprotein convertase subtilisin/kexin 9)-induced pro-inflammatory response in macrophages, identified via hyperedge entanglement prediction (HEP) prediction. A, SDC4 mRNA level was measured in murine peritoneal macrophages of Ldlr-/- mice 1 week after intravenous injection with AAV-LacZ or AAV-PCSK9 (n=7 per group). B, mRNA levels of SDC4, IL-1b, NLRP3, CD40, and ICAM1 were measured by RT-qPCR in mouse Ldlr-/- peritoneal macrophages after pretreatment with siSDC4 or siControl (Ctrl) for 48 hours and stimulation with 2.5 μg/mL recombinant mouse PCSK9 (MmPCSK9) for 3 hours (n=4–5). P value was calculated by 1-way ANOVA, followed by Bonferroni multiple comparison test. Error bars indicate ± SEM. Data are reported as mean ± SEM.", "We further validated our prediction that SDC4 is a potential target of PCSK9 using siRNA silencing of mouse Sdc4 in mouse peritoneal macrophages. Silencing Sdc4 with siSDC4 treatment achieved 93% reduction of SDC4 mRNA expression (Figure 6B). siSDC4 was then used to determine whether it could suppress PCSK9-induced pro-inflammatory molecules, including those predicted to be linked with PCSK9 by the HEP prediction (Figure 5C). siSDC4 suppressed mRNAs of the pro-inflammatory molecules IL-1β, NLRP3, CD40, and ICAM1, but not TNFα, MCP-1, TLR2, CXCL1, CXCL2, or CXCL10 after stimulation with recombinant PCSK9 (Figure 6B, Supplemental Figure 9). These data suggest that SDC4 partially regulates a PCSK9-induced pro-inflammatory response. Furthermore, these mRNA silencing results substantiate the validity of the prediction, yielding the integration of PCSK9 in our hyperedge of 24 disconnected differentially expressed transcripts.", "Our study demonstrated that circulating PCSK9 produced various changes in Ldlr-/- mice. We, however, attempted to examine the potential impact of local PCSK9 production in vein grafts. Immunohistochemical analysis using an anti-PCSK9 antibody in previous studies showed that vascular smooth muscle cells in human atherosclerotic plaque and collar-induced neointima of murine carotid artery express PCSK9.45,46 Based on these results, we examined whether locally produced PCSK9 in vein grafts may also contribute to vein graft lesion development. The local expression of PCSK9, as determined by in situ hybridization in the intima of vein grafts, was minimal compared with that in the liver (Figure 1B; Supplemental Figure 10A). RT-qPCR analysis further confirmed numerically significantly less PCSK9 mRNA expression in vein grafts than in the liver, the primary source of circulating PCSK9 (Supplemental Figure 10B). Moreover, we examined which cells within vein grafts can produce PCSK9 in humans by comparing PCSK9 mRNA levels in human saphenous vein endothelial cells, smooth muscle cells, and human primary macrophages derived from peripheral blood mononuclear cells. While we detected substantial PCSK9 expression in human saphenous vein smooth muscle cells, the level was significantly lower than that in HepG2 (liver) cells (Supplemental Figure 10C). Human primary macrophages and human saphenous vein endothelial cells contained minimal PCSK9 mRNA (Supplemental Figure 10C). Recent studies reported a modest increase of PCSK9 mRNA and protein levels in macrophages exposed to pro-inflammatory stimuli.39,47 We, therefore, examined PCSK9 mRNA expression in human primary macrophages treated with pro-inflammatory stimuli. Although LPS and IFNγ induced a pro-inflammatory response in the macrophages, as determined by TNFα mRNA expression (Supplemental Figures 10D and 10E, right graphs), PCSK9 mRNA expression did not increase after LPS or IFNγ (Supplemental Figures 10D and 10E, left graphs). These results indicate that smooth muscle cells (SMC), but not macrophages, may primarily produce PCSK9 in vein grafts, but to a much lower extent than the liver.", "Although our study focuses on macrophages, we examined the role of PCSK9 in the activation of endothelial cells and SMCs because other investigators reported potential interactions between PCSK9 and these cell types.46,48 Endothelial cell activation precedes vein graft lesion development.49 Due to the scarcity of mRNA in CD31-positive endothelial cells in vein grafts, we sorted them from the liver and lungs of Ldlr-/- mice injected with AAV-PCSK9 instead to examine the effects of PCSK9 on endothelial cell activation in an LDLR-independent fashion. AAV-PCSK9 did not increase mRNA levels of adhesion molecules, such as VCAM-1, ICAM-1, and E-selectin, in EC from either organ when compared with AAV-LacZ (Supplemental Figure 11A). We further found in vitro that recombinant PCSK9 did not increase mRNA levels of these adhesion molecules in human saphenous vein endothelial cells (Supplemental Figure 11B). These results indirectly indicate that PCSK9 may not induce endothelial activation in vein grafts.\nSMC modulation, migration, and proliferation contribute to the development of vascular disorders, including atherosclerosis50 and vein graft.51 A recent study showed that PCSK9 may sustain SMC dedifferentiation, migration, and proliferation in neointimal hyperplasia in response to vascular injury,46 suggesting the involvement of SMCs in PCSK9-induced vein graft lesion development. To explore this possibility, we examined the effects of recombinant PCSK9 on SMC dedifferentiation, migration, and proliferation in human saphenous vein smooth muscle cells. Recombinant PCSK9 slightly decreased SM22α mRNA levels (0.74 and 0.68-fold for 1.0 and 5.0 µg/mL, respectively), but did not affect differentiation molecules, such as αSMA, calponin, SM1, or SM2 (Supplemental Figure 12A). In addition, recombinant PCSK9 did not affect PDGF-induced migration activity of human saphenous vein smooth muscle cells (Supplemental Figure 12B). A BrdU assay using human saphenous vein smooth muscle cells showed that recombinant PCSK9 did not enhance 10% fetal bovine serum-induced proliferation (Supplemental Figure 12C). AAV-PCSK9 did not affect the SMC content in vein grafts (Supplemental Figure 12D). Collectively, in our experimental settings, our data did not provide the mechanistic evidence in vitro and in vivo that activation of endothelial cells or SMCs contribute to PCSK9-induced vein graft lesion formation. These results indicate that circulating PCSK9 promotes vein graft lesion development predominantly through macrophage activation.", "[SUBTITLE] Acknowledgments [SUBSECTION] We thank Ryo Kawakami, Yuki Tsukano, Dayanna C. Romero, Whitney S. Golden, Keishi Nihira, Andrew K. Mlynarchik, Edward Guzman, and Daniel G. Anderson for their technical assistance. The graphical figure was created with Biorender.com. Additional Information: Coauthor Amitabh Sharma died on November 24, 2019.\nWe thank Ryo Kawakami, Yuki Tsukano, Dayanna C. Romero, Whitney S. Golden, Keishi Nihira, Andrew K. Mlynarchik, Edward Guzman, and Daniel G. Anderson for their technical assistance. The graphical figure was created with Biorender.com. Additional Information: Coauthor Amitabh Sharma died on November 24, 2019.\n[SUBTITLE] Sources of Funding [SUBSECTION] This work was supported by the National Institute of Health grants R01HL126901 and R01HL149302, and Pfizer ASPIRE Award to MA; R01HL136431, R01HL147095, and R01HL141917 to EA; and the MSD Scholarships for Overseas Study, and Japan Society for the Promotion of Science Fellowships for Overseas Researchers to SK.\nThis work was supported by the National Institute of Health grants R01HL126901 and R01HL149302, and Pfizer ASPIRE Award to MA; R01HL136431, R01HL147095, and R01HL141917 to EA; and the MSD Scholarships for Overseas Study, and Japan Society for the Promotion of Science Fellowships for Overseas Researchers to SK.\n[SUBTITLE] Disclosures [SUBSECTION] Pfizer was not involved in the study other than funding. MA also received research grants from Kowa Company and Sanofi.\nPfizer was not involved in the study other than funding. MA also received research grants from Kowa Company and Sanofi.\n[SUBTITLE] Supplemental Materials [SUBSECTION] Online Supplemental Materials and Methods\nSupplemental Figures 1–14\nSupplemental Tables 1–4\nReferences 69–75\nOnline Supplemental Materials and Methods\nSupplemental Figures 1–14\nSupplemental Tables 1–4\nReferences 69–75", "We thank Ryo Kawakami, Yuki Tsukano, Dayanna C. Romero, Whitney S. Golden, Keishi Nihira, Andrew K. Mlynarchik, Edward Guzman, and Daniel G. Anderson for their technical assistance. The graphical figure was created with Biorender.com. Additional Information: Coauthor Amitabh Sharma died on November 24, 2019.", "This work was supported by the National Institute of Health grants R01HL126901 and R01HL149302, and Pfizer ASPIRE Award to MA; R01HL136431, R01HL147095, and R01HL141917 to EA; and the MSD Scholarships for Overseas Study, and Japan Society for the Promotion of Science Fellowships for Overseas Researchers to SK.", "Online Supplemental Materials and Methods\nSupplemental Figures 1–14\nSupplemental Tables 1–4\nReferences 69–75" ]
[ null, null, "methods", null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "What Is Known?", "What New Information Does This Article Contribute?", "Methods", "Data Availability", "Animal Procedures", "Culture and Stimulation of Mouse Primary Macrophages", "Transcriptomics", "Network-Based Hyperedge Entanglement Prediction Analysis", "Statistics", "Results", "AAV-PCSK9 Promotes Vein Graft Lesion Development in an LDLR-Independent Manner", "AAV-PCSK9 Increases Thin Collagen Fibers and Macrophage Accumulation in Vein Graft Lesions", "AAV-PCSK9 Induces Macrophage Activation via an LDLR-Independent Route In Vivo", "Endotoxin-Free Recombinant PCSK9 Induced Macrophage Activation in Peritoneal Macrophages of Ldlr\n\n-/- Mice In Vitro", "A combination of Unbiased Global Transcriptomics and Network-Based Hyperedge Entanglement Prediction Analysis Revealed Potential Targets of PCSK9 in Macrophages", "SDC4 Silencing Suppresses PCSK9-Induced Pro-Inflammatory Responses in Macrophages", "Local Production of PCSK9 in the Liver Was Greater Than in Vein Graft Lesions", "Macrophages Rather Than Endothelial Cells and SMCs May Mediate the Effects of PCSK9 on Vein Graft Lesion Development", "Discussion", "Article Information", "Acknowledgments", "Sources of Funding", "Disclosures", "Supplemental Materials", "Supplementary Material" ]
[ "Autologous saphenous vein is a widely used surgical bypass for lower extremity peripheral artery disease and coronary artery disease.\nPCSK9 (proprotein convertase subtilisin/kexin 9) targets receptors other than LDL receptor (LDLR) such as LRP1 (low-density lipoprotein receptor-related protein-1), ApoER2 (apolipoprotein E receptor 2), VLDLR (very low-density lipoprotein receptor), and CD36.\nPCSK9 may induce pro-inflammatory responses in macrophages and arterial atherosclerotic lesions primarily via LDLR-dependent mechanisms.", "PCSK9 promotes vein graft lesion development by the mechanisms unassociated with either LDLR degradation or blood cholesterol levels.\nPCSK9 induces pro-inflammatory macrophage activation: immune responses, proliferation and migration, via LDLR-independent mechanisms.\nPotential PCSK9 targets mediating pro-inflammatory responses in macrophages include NF-κB (nuclear factor-kappa B) signaling molecules, lectin-like oxidized LOX-1 (LDL receptor-1), and SDC4 (syndecan-4).\nThe role of PCSK9 on vascular inflammation remains controversial in the clinical setting because clinical trials with PCSK9 inhibitors did not show any significant changes in high sensitivity-CRP (C-reactive protein) levels in patients with coronary artery disease. However, CRP may not solely capture changes in inflammation. Although there are a few emerging in vivo studies suggesting a direct link between PCSK9 and atherosclerosis primarily via LDLR-dependent mechanisms, the role of PCSK9 in vein graft failure, especially via LDLR-independent mechanisms, remains largely unknown. To test the hypothesis, we developed a systems approach, involving unbiased transcriptomics and network analysis. Our in vivo evidence suggests that PCSK9 promotes vein graft lesion development, macrophage proliferation, monocyte migration, foam cell formation, and collagen remodeling. We further identified NF-κB signaling molecules, LOX-1 and SDC4 as potential targets of PCSK9 mediating pro-inflammatory responses in macrophages. The evidence, including our own, has suggested that circulating PCSK9 may exert pro-inflammatory effects on vascular lesions.", "[SUBTITLE] Data Availability [SUBSECTION] The online-only Supplemental Data provided detailed methods of all procedures. The data, analytic methods, and study materials will be made available to other researchers, upon request for purposes of reproducing the results or replicating the procedure.\nThe online-only Supplemental Data provided detailed methods of all procedures. The data, analytic methods, and study materials will be made available to other researchers, upon request for purposes of reproducing the results or replicating the procedure.\n[SUBTITLE] Animal Procedures [SUBSECTION] All animal experiments were approved by the Brigham and Women’s Hospital’s Animal Welfare Assurance (protocol 2016N000219). Male Ldlr-/- mice were fed a high-fat diet (1.25% cholesterol, D12108C, Research Diets, Inc., New Brunswick, NJ). To create experimental vein grafts in Ldlr-/- mice, inferior vena cava were harvested from a donor mouse and implanted into the carotid artery of recipient mice using cuff technique, as previously described.8,21 To induce liver-specific gain-of-function mutant PCSK9, we administered adeno-associated virus (AAV) encoding a gain-of-function form of mouse PCSK9 (AAV-PCSK9, 1×1011 vg, i.v.).22 We constructed AAV encoding LacZ (AAV-LacZ) that we used as a control for AAV-PCSK9. Four weeks after implantation, grafts were evaluated. After ultrasonography and in vivo molecular imaging of vein grafts, vein graft tissues were harvested, sectioned, and used for Masson trichrome staining, picrosirius red staining, immunohistochemistry, and in situ hybridization.\nAll animal experiments were approved by the Brigham and Women’s Hospital’s Animal Welfare Assurance (protocol 2016N000219). Male Ldlr-/- mice were fed a high-fat diet (1.25% cholesterol, D12108C, Research Diets, Inc., New Brunswick, NJ). To create experimental vein grafts in Ldlr-/- mice, inferior vena cava were harvested from a donor mouse and implanted into the carotid artery of recipient mice using cuff technique, as previously described.8,21 To induce liver-specific gain-of-function mutant PCSK9, we administered adeno-associated virus (AAV) encoding a gain-of-function form of mouse PCSK9 (AAV-PCSK9, 1×1011 vg, i.v.).22 We constructed AAV encoding LacZ (AAV-LacZ) that we used as a control for AAV-PCSK9. Four weeks after implantation, grafts were evaluated. After ultrasonography and in vivo molecular imaging of vein grafts, vein graft tissues were harvested, sectioned, and used for Masson trichrome staining, picrosirius red staining, immunohistochemistry, and in situ hybridization.\n[SUBTITLE] Culture and Stimulation of Mouse Primary Macrophages [SUBSECTION] Mouse peritoneal macrophages cultured with RPMI 1640 containing 10% fetal bovine serum were prepared as previously described.8 Briefly, 4% Brewer thioglycolate medium (BD Diagnostic Systems) was injected into the peritoneal cavity of mice 4 days before macrophage collection. After euthanasia, 10 mL of ice-cold phosphate-buffered saline was injected into the peritoneal cavity, and cells were harvested. The cells were then washed with phosphate-buffered saline once and plated for further experiments. Overnight starvation was performed with 0.5% fetal bovine serum before each experiment. Endotoxin-free recombinant mouse PCSK9 derived from mouse myeloma cell line NS0 was used to stimulate mouse primary macrophages (R&D Systems).\nMouse peritoneal macrophages cultured with RPMI 1640 containing 10% fetal bovine serum were prepared as previously described.8 Briefly, 4% Brewer thioglycolate medium (BD Diagnostic Systems) was injected into the peritoneal cavity of mice 4 days before macrophage collection. After euthanasia, 10 mL of ice-cold phosphate-buffered saline was injected into the peritoneal cavity, and cells were harvested. The cells were then washed with phosphate-buffered saline once and plated for further experiments. Overnight starvation was performed with 0.5% fetal bovine serum before each experiment. Endotoxin-free recombinant mouse PCSK9 derived from mouse myeloma cell line NS0 was used to stimulate mouse primary macrophages (R&D Systems).\n[SUBTITLE] Transcriptomics [SUBSECTION] RNA-sequencing—mRNAseq (polyA enriched) library prep, single-end 75bp sequencing on NextSeq of 12-16 pooled barcoded samples, and VIPER analysis23—was performed at the Molecular Biology Core Facilities at Dana-Farber Cancer Institute. The data were then analyzed using DESeq2 and Qlucore (http://www.qlucore.com/) to perform a 2-group comparison and identify differentially expressed transcripts (increase/decrease by 2-fold) between control and PCSK9 groups. STRING (version 11.5) was used to evaluate known and predicted transcript interconnectivity. The differentially expressed transcripts (increase/decrease by 2-fold, q<0.05), which were compared with the “inflammatome”24 dataset, were used for the following network analysis.\nRNA-sequencing—mRNAseq (polyA enriched) library prep, single-end 75bp sequencing on NextSeq of 12-16 pooled barcoded samples, and VIPER analysis23—was performed at the Molecular Biology Core Facilities at Dana-Farber Cancer Institute. The data were then analyzed using DESeq2 and Qlucore (http://www.qlucore.com/) to perform a 2-group comparison and identify differentially expressed transcripts (increase/decrease by 2-fold) between control and PCSK9 groups. STRING (version 11.5) was used to evaluate known and predicted transcript interconnectivity. The differentially expressed transcripts (increase/decrease by 2-fold, q<0.05), which were compared with the “inflammatome”24 dataset, were used for the following network analysis.\n[SUBTITLE] Network-Based Hyperedge Entanglement Prediction Analysis [SUBSECTION] Network-Based Hyperedge Entanglement Prediction (HEP) algorithm20 was performed to estimate the likelihood of PCSK9 directly interacting with any of the differentially expressed transcripts in the “inflammatome.” Given only a network topology and its high-order hypergraph organization, the HEP algorithm uses an ensemble of link predictors to calculate a level of statistical significance for each candidate node to hyperedge entanglement that is characterized by not having a direct connection. The link predictor is an operator that, exploiting topological information of the graph, can associate a similarity score to any disconnected node pair, suggesting the likelihood for a link between them to exist. Several link predictors perform under the evidence that 2 nodes are more likely to be linked if their common neighbors are members of a strongly inner-linked cohort, named a local community,25 and recent studies demonstrated that this growth mechanism emerges mainly considering missing links between nonadjacent nodes distanced by paths of length 2 (L2) or 3 (L3)26 following a scheme that can be well described by the Cannistraci-Hebb network automata modeling.27 The HEP analysis provides 12 different estimations of the P value that quantifies the significance of the entanglement (association) between each node and hyperedge (also called node2hyperedge entanglement) using the 12 available node-node link prediction variants.25, 27 In this study, we applied HEP algorithm according to the following specifications: the scaling of the similarity scores was not necessary, and the average operator to estimate the node2hyperedge entanglement scores was the mean.\nNetwork-Based Hyperedge Entanglement Prediction (HEP) algorithm20 was performed to estimate the likelihood of PCSK9 directly interacting with any of the differentially expressed transcripts in the “inflammatome.” Given only a network topology and its high-order hypergraph organization, the HEP algorithm uses an ensemble of link predictors to calculate a level of statistical significance for each candidate node to hyperedge entanglement that is characterized by not having a direct connection. The link predictor is an operator that, exploiting topological information of the graph, can associate a similarity score to any disconnected node pair, suggesting the likelihood for a link between them to exist. Several link predictors perform under the evidence that 2 nodes are more likely to be linked if their common neighbors are members of a strongly inner-linked cohort, named a local community,25 and recent studies demonstrated that this growth mechanism emerges mainly considering missing links between nonadjacent nodes distanced by paths of length 2 (L2) or 3 (L3)26 following a scheme that can be well described by the Cannistraci-Hebb network automata modeling.27 The HEP analysis provides 12 different estimations of the P value that quantifies the significance of the entanglement (association) between each node and hyperedge (also called node2hyperedge entanglement) using the 12 available node-node link prediction variants.25, 27 In this study, we applied HEP algorithm according to the following specifications: the scaling of the similarity scores was not necessary, and the average operator to estimate the node2hyperedge entanglement scores was the mean.\n[SUBTITLE] Statistics [SUBSECTION] Statistical analyses were performed using GraphPad Prism 8.0 (GraphPad Software). For experiments with a small sample size (n<6), P values were determined by nonparametric analysis except for RNA-sequencing analysis. Other data were examined for normality before analysis by Shapiro-Wilk test. For all normally distributed data, an unpaired 2-tailed Student t test was used for comparisons between 2 groups, 1-way ANOVA followed by Bonferroni or Dunnett post-hoc test was used to compare multiple groups. When there were 2 experimental factors, 2-way ANOVA for comparisons between multiple groups was used. If the data were not normally distributed, nonparametric unpaired 2-tailed Mann-Whitney U test was used to compare 2 groups, and Kruskal-Wallis test with Dunn post-hoc test was used to compare multiple groups. Wald Test in DESeq2 was used for bulk RNA-sequencing analysis. Data are expressed as mean ± SEM for continuous variables. P values of <0.05 were considered statistically significant.\nStatistical analyses were performed using GraphPad Prism 8.0 (GraphPad Software). For experiments with a small sample size (n<6), P values were determined by nonparametric analysis except for RNA-sequencing analysis. Other data were examined for normality before analysis by Shapiro-Wilk test. For all normally distributed data, an unpaired 2-tailed Student t test was used for comparisons between 2 groups, 1-way ANOVA followed by Bonferroni or Dunnett post-hoc test was used to compare multiple groups. When there were 2 experimental factors, 2-way ANOVA for comparisons between multiple groups was used. If the data were not normally distributed, nonparametric unpaired 2-tailed Mann-Whitney U test was used to compare 2 groups, and Kruskal-Wallis test with Dunn post-hoc test was used to compare multiple groups. Wald Test in DESeq2 was used for bulk RNA-sequencing analysis. Data are expressed as mean ± SEM for continuous variables. P values of <0.05 were considered statistically significant.", "The online-only Supplemental Data provided detailed methods of all procedures. The data, analytic methods, and study materials will be made available to other researchers, upon request for purposes of reproducing the results or replicating the procedure.", "All animal experiments were approved by the Brigham and Women’s Hospital’s Animal Welfare Assurance (protocol 2016N000219). Male Ldlr-/- mice were fed a high-fat diet (1.25% cholesterol, D12108C, Research Diets, Inc., New Brunswick, NJ). To create experimental vein grafts in Ldlr-/- mice, inferior vena cava were harvested from a donor mouse and implanted into the carotid artery of recipient mice using cuff technique, as previously described.8,21 To induce liver-specific gain-of-function mutant PCSK9, we administered adeno-associated virus (AAV) encoding a gain-of-function form of mouse PCSK9 (AAV-PCSK9, 1×1011 vg, i.v.).22 We constructed AAV encoding LacZ (AAV-LacZ) that we used as a control for AAV-PCSK9. Four weeks after implantation, grafts were evaluated. After ultrasonography and in vivo molecular imaging of vein grafts, vein graft tissues were harvested, sectioned, and used for Masson trichrome staining, picrosirius red staining, immunohistochemistry, and in situ hybridization.", "Mouse peritoneal macrophages cultured with RPMI 1640 containing 10% fetal bovine serum were prepared as previously described.8 Briefly, 4% Brewer thioglycolate medium (BD Diagnostic Systems) was injected into the peritoneal cavity of mice 4 days before macrophage collection. After euthanasia, 10 mL of ice-cold phosphate-buffered saline was injected into the peritoneal cavity, and cells were harvested. The cells were then washed with phosphate-buffered saline once and plated for further experiments. Overnight starvation was performed with 0.5% fetal bovine serum before each experiment. Endotoxin-free recombinant mouse PCSK9 derived from mouse myeloma cell line NS0 was used to stimulate mouse primary macrophages (R&D Systems).", "RNA-sequencing—mRNAseq (polyA enriched) library prep, single-end 75bp sequencing on NextSeq of 12-16 pooled barcoded samples, and VIPER analysis23—was performed at the Molecular Biology Core Facilities at Dana-Farber Cancer Institute. The data were then analyzed using DESeq2 and Qlucore (http://www.qlucore.com/) to perform a 2-group comparison and identify differentially expressed transcripts (increase/decrease by 2-fold) between control and PCSK9 groups. STRING (version 11.5) was used to evaluate known and predicted transcript interconnectivity. The differentially expressed transcripts (increase/decrease by 2-fold, q<0.05), which were compared with the “inflammatome”24 dataset, were used for the following network analysis.", "Network-Based Hyperedge Entanglement Prediction (HEP) algorithm20 was performed to estimate the likelihood of PCSK9 directly interacting with any of the differentially expressed transcripts in the “inflammatome.” Given only a network topology and its high-order hypergraph organization, the HEP algorithm uses an ensemble of link predictors to calculate a level of statistical significance for each candidate node to hyperedge entanglement that is characterized by not having a direct connection. The link predictor is an operator that, exploiting topological information of the graph, can associate a similarity score to any disconnected node pair, suggesting the likelihood for a link between them to exist. Several link predictors perform under the evidence that 2 nodes are more likely to be linked if their common neighbors are members of a strongly inner-linked cohort, named a local community,25 and recent studies demonstrated that this growth mechanism emerges mainly considering missing links between nonadjacent nodes distanced by paths of length 2 (L2) or 3 (L3)26 following a scheme that can be well described by the Cannistraci-Hebb network automata modeling.27 The HEP analysis provides 12 different estimations of the P value that quantifies the significance of the entanglement (association) between each node and hyperedge (also called node2hyperedge entanglement) using the 12 available node-node link prediction variants.25, 27 In this study, we applied HEP algorithm according to the following specifications: the scaling of the similarity scores was not necessary, and the average operator to estimate the node2hyperedge entanglement scores was the mean.", "Statistical analyses were performed using GraphPad Prism 8.0 (GraphPad Software). For experiments with a small sample size (n<6), P values were determined by nonparametric analysis except for RNA-sequencing analysis. Other data were examined for normality before analysis by Shapiro-Wilk test. For all normally distributed data, an unpaired 2-tailed Student t test was used for comparisons between 2 groups, 1-way ANOVA followed by Bonferroni or Dunnett post-hoc test was used to compare multiple groups. When there were 2 experimental factors, 2-way ANOVA for comparisons between multiple groups was used. If the data were not normally distributed, nonparametric unpaired 2-tailed Mann-Whitney U test was used to compare 2 groups, and Kruskal-Wallis test with Dunn post-hoc test was used to compare multiple groups. Wald Test in DESeq2 was used for bulk RNA-sequencing analysis. Data are expressed as mean ± SEM for continuous variables. P values of <0.05 were considered statistically significant.", "[SUBTITLE] AAV-PCSK9 Promotes Vein Graft Lesion Development in an LDLR-Independent Manner [SUBSECTION] In vivo studies have suggested a direct link between PCSK9 and atherosclerosis28,29; however, no evidence has yet implicated PCSK9 in vein graft lesion development. Furthermore, these in vivo studies indicate that the underlying mechanism of atherosclerotic lesion development by PCSK9 may depend mainly on LDLR. Whether PCSK9 can exert pro-inflammatory effects via LDLR-independent mechanisms is unclear. To test the hypothesis that circulating PCSK9 promotes vein graft lesion development in vivo, we used experimental vein grafts in mice.21 A single intravenous injection of AAV-PCSK9 induced liver production of PCSK9. To exclude potential systemic toxicity of AAV, AAV-LacZ was developed and used as a control (Figure 1A; Online Supplemental Methods). All experiments used Ldlr-/- mice to explore LDLR-independent mechanisms unless otherwise noted.\nThe effects of Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) on vein graft lesion development in Ldlr-/- mice. A, An experimental protocol for the treatments in Ldlr-/- mice. B, In situ hybridization of PCSK9 (red) in the liver of Ldlr-/- mice 1 week after intravenous injection with AAV-LacZ or AAV-PCSK9. Nuclei are visualized with DAPI. The data represent 4 mice per group. Scale bars indicate 100 μm. C, PCSK9 mRNA level in the liver of AAV treated Ldlr-/- mice (n=12 and 11 for AAV-LacZ and AAV-PCSK9 groups, respectively), and serum levels of PCSK9, total cholesterol, and triglyceride in AAV treated Ldlr-/- mice (n=11 per group). D, Left, ultrasonography images of vein grafts from Ldlr-/- mice treated with AAV-LacZ or AAV-PCSK9 28 days after implantation. The red, green, and yellow dotted lines indicate lumen, near wall, and far wall, respectively. Right, The quantitative analysis of lumen and vessel wall area in short axis view and 3D reconstructed vessel wall volume of vein grafts by ultrasonography (n=12 and 11 for AAV-LacZ and AAV-PCSK9 groups, respectively). E, Left, Masson’s trichrome staining of vein grafts from Ldlr-/- mice treated with AAV-LacZ or AAV-PCSK9 28 days after implantation. Scale bars indicate 500 μm. Right, The quantitative analysis of intimal area, intimal and media/adventitia thickness, and lumen and vessel diameter of vein grafts (n=12 and 11 for AAV-LacZ and AAV-PCSK9 group, respectively). P value was calculated by Mann-Whitney U-test (triglyceride in C, media/adventitia thickness, lumen diameter, and vessel diameter in E) and unpaired Student t test (A, B, and D). Data are reported as mean ± SEM.\nBody weight and blood pressure did not differ significantly between the AAV-LacZ and AAV-PCSK9 groups 4 weeks after vein graft implantation (Supplemental Table 1). In situ hybridization showed that AAV-PCSK9 markedly increased PCSK9 mRNA levels in the liver (Figure 1B). AAV-PCSK9 increased PCSK9 mRNA expression in the liver and serum levels of PCSK9 (Figure 1C; Supplemental Table 1). Serum levels of total cholesterol and triglycerides did not differ between the AAV-LacZ and AAV-PCSK9 groups (Figure 1C; Supplemental Table 1). These findings indicate that the effects of AAV-PCSK9 on vein graft lesions described below did not depend on blood cholesterol and triglyceride concentrations (Supplemental Table 1).\nNoninvasive ultrasonography visualized that AAV-PCSK9 injection caused increased vessel wall area, but not lumen area, in the short axis view and 3-dimensional reconstructed vessel wall volume of vein grafts (Figure 1D). Consistent with the ultrasonography, histological analysis by Masson trichrome staining showed that AAV-PCSK9 increased intimal area and thickness of vein grafts. Lumen diameter, vessel diameter, and media/adventitia thickness did not differ significantly, suggesting compensatory outward remodeling of vein grafts (Figure 1E).\nIn vivo studies have suggested a direct link between PCSK9 and atherosclerosis28,29; however, no evidence has yet implicated PCSK9 in vein graft lesion development. Furthermore, these in vivo studies indicate that the underlying mechanism of atherosclerotic lesion development by PCSK9 may depend mainly on LDLR. Whether PCSK9 can exert pro-inflammatory effects via LDLR-independent mechanisms is unclear. To test the hypothesis that circulating PCSK9 promotes vein graft lesion development in vivo, we used experimental vein grafts in mice.21 A single intravenous injection of AAV-PCSK9 induced liver production of PCSK9. To exclude potential systemic toxicity of AAV, AAV-LacZ was developed and used as a control (Figure 1A; Online Supplemental Methods). All experiments used Ldlr-/- mice to explore LDLR-independent mechanisms unless otherwise noted.\nThe effects of Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) on vein graft lesion development in Ldlr-/- mice. A, An experimental protocol for the treatments in Ldlr-/- mice. B, In situ hybridization of PCSK9 (red) in the liver of Ldlr-/- mice 1 week after intravenous injection with AAV-LacZ or AAV-PCSK9. Nuclei are visualized with DAPI. The data represent 4 mice per group. Scale bars indicate 100 μm. C, PCSK9 mRNA level in the liver of AAV treated Ldlr-/- mice (n=12 and 11 for AAV-LacZ and AAV-PCSK9 groups, respectively), and serum levels of PCSK9, total cholesterol, and triglyceride in AAV treated Ldlr-/- mice (n=11 per group). D, Left, ultrasonography images of vein grafts from Ldlr-/- mice treated with AAV-LacZ or AAV-PCSK9 28 days after implantation. The red, green, and yellow dotted lines indicate lumen, near wall, and far wall, respectively. Right, The quantitative analysis of lumen and vessel wall area in short axis view and 3D reconstructed vessel wall volume of vein grafts by ultrasonography (n=12 and 11 for AAV-LacZ and AAV-PCSK9 groups, respectively). E, Left, Masson’s trichrome staining of vein grafts from Ldlr-/- mice treated with AAV-LacZ or AAV-PCSK9 28 days after implantation. Scale bars indicate 500 μm. Right, The quantitative analysis of intimal area, intimal and media/adventitia thickness, and lumen and vessel diameter of vein grafts (n=12 and 11 for AAV-LacZ and AAV-PCSK9 group, respectively). P value was calculated by Mann-Whitney U-test (triglyceride in C, media/adventitia thickness, lumen diameter, and vessel diameter in E) and unpaired Student t test (A, B, and D). Data are reported as mean ± SEM.\nBody weight and blood pressure did not differ significantly between the AAV-LacZ and AAV-PCSK9 groups 4 weeks after vein graft implantation (Supplemental Table 1). In situ hybridization showed that AAV-PCSK9 markedly increased PCSK9 mRNA levels in the liver (Figure 1B). AAV-PCSK9 increased PCSK9 mRNA expression in the liver and serum levels of PCSK9 (Figure 1C; Supplemental Table 1). Serum levels of total cholesterol and triglycerides did not differ between the AAV-LacZ and AAV-PCSK9 groups (Figure 1C; Supplemental Table 1). These findings indicate that the effects of AAV-PCSK9 on vein graft lesions described below did not depend on blood cholesterol and triglyceride concentrations (Supplemental Table 1).\nNoninvasive ultrasonography visualized that AAV-PCSK9 injection caused increased vessel wall area, but not lumen area, in the short axis view and 3-dimensional reconstructed vessel wall volume of vein grafts (Figure 1D). Consistent with the ultrasonography, histological analysis by Masson trichrome staining showed that AAV-PCSK9 increased intimal area and thickness of vein grafts. Lumen diameter, vessel diameter, and media/adventitia thickness did not differ significantly, suggesting compensatory outward remodeling of vein grafts (Figure 1E).\n[SUBTITLE] AAV-PCSK9 Increases Thin Collagen Fibers and Macrophage Accumulation in Vein Graft Lesions [SUBSECTION] Evidence suggests that vein grafts in humans can develop lesions similar to those of advanced arterial atherosclerosis, and plaque rupture can occur in inflamed vein grafts.30,31 Therefore, exploring vein graft lesion composition in depth is critical to understanding what contributes to this occurrence. We then examined the collagen content of vein grafts by picrosirius red staining. AAV-PCSK9 increased the percentage of thin collagen fibers as a ratio of the total collagen fibers in the vein grafts, suggesting that circulating PCSK9 induced pathological features similar to those in thin-capped atherosclerotic plaques (Figure 2A). In vivo molecular imaging further provided insight into the effects of circulating PCSK9 on matrix metalloproteinase activity and macrophage accumulation in vein grafts. We intravenously co-injected 2 spectrally different imaging agents that elaborate near-infrared signals for visualization of matrix metalloproteinase activity (MMPsense, 680 nm) and macrophage accumulation (AminoSPARK, 750 nm). Following the evidence that activated macrophages express MMPs that degrade interstitial collagen as previously reported by our group,32,33 in vivo molecular imaging demonstrated that increased matrix metalloproteinase activity co-localized with accumulated macrophages (Figure 2B, left). In addition, AAV-PCSK9 treatment promoted matrix metalloproteinase activity and macrophage accumulation in vein grafts (Figure 2B), suggesting a potential mechanism of collagen remodeling as determined by Picrosirius red staining. Immunohistochemical analysis further demonstrated increased macrophage accumulation in vein grafts by AAV-PCSK9 (Mac3; Figure 2C). Macrophage proliferation and migration may contribute to the mechanisms of macrophage accumulation in vascular diseases.34 Supporting this mechanism, AAV-PCSK9 increased the percentage of macrophages positive for Ki-67, an indicator of proliferating cells (Supplemental Figure 1A, B). Consistent with this in vivo finding, recombinant PCSK9 augmented M-CSF-induced cell proliferation in mouse bone marrow-derived macrophages (BMDMs) (Supplemental Figure 2A, B). Monocyte migration assay using the human monocytic cell line THP-1 showed that recombinant PCSK9 tended to promote MCP-1 (monocyte chemoattractant protein-1)-induced chemotaxis, suggesting lipid-independent effects of PCSK9 on monocyte migration (Supplemental Figure 2C). On the other hand, AAV-PCSK9 did not increase apoptosis determined by TUNEL staining in vein grafts of Ldlr-/- mice (Supplemental Figure 3A). Accordingly, AAV-PCSK9 did not change necrotic core area in the vein grafts (Supplemental Figure 3B).\nThe effects of Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) on collagen thinning and macrophage accumulation in vein graft lesions of Ldlr-/- mice. A, Picrosirius red staining of vein grafts without (top) or with (bottom) polarized light (n=12 and 11 for AAV-LacZ and AAV-PCSK9 group, respectively). Scale bars indicate 500 μm. Circle graphs show the percentage of thin (green) and thick (red) collagen fibers compared with total fibers. B, Intravital microscopy images of MMPSense 680 (red) and AminoSPARK750 (green) in vein grafts for visualization of MMP activity and macrophage accumulation, respectively (n=10 and 9 for AAV-LacZ and AAV-PCSK9 group, respectively). C, Mac3 (macrophages) staining in vein grafts. Scale bars indicate 500 μm. P value was calculated by Mann-Whitney U-test (thick collagen in A, macrophage accumulation mean fluorescence intensity in B) and unpaired Student t test (C). Data are reported as mean ± SEM.\nEvidence suggests that vein grafts in humans can develop lesions similar to those of advanced arterial atherosclerosis, and plaque rupture can occur in inflamed vein grafts.30,31 Therefore, exploring vein graft lesion composition in depth is critical to understanding what contributes to this occurrence. We then examined the collagen content of vein grafts by picrosirius red staining. AAV-PCSK9 increased the percentage of thin collagen fibers as a ratio of the total collagen fibers in the vein grafts, suggesting that circulating PCSK9 induced pathological features similar to those in thin-capped atherosclerotic plaques (Figure 2A). In vivo molecular imaging further provided insight into the effects of circulating PCSK9 on matrix metalloproteinase activity and macrophage accumulation in vein grafts. We intravenously co-injected 2 spectrally different imaging agents that elaborate near-infrared signals for visualization of matrix metalloproteinase activity (MMPsense, 680 nm) and macrophage accumulation (AminoSPARK, 750 nm). Following the evidence that activated macrophages express MMPs that degrade interstitial collagen as previously reported by our group,32,33 in vivo molecular imaging demonstrated that increased matrix metalloproteinase activity co-localized with accumulated macrophages (Figure 2B, left). In addition, AAV-PCSK9 treatment promoted matrix metalloproteinase activity and macrophage accumulation in vein grafts (Figure 2B), suggesting a potential mechanism of collagen remodeling as determined by Picrosirius red staining. Immunohistochemical analysis further demonstrated increased macrophage accumulation in vein grafts by AAV-PCSK9 (Mac3; Figure 2C). Macrophage proliferation and migration may contribute to the mechanisms of macrophage accumulation in vascular diseases.34 Supporting this mechanism, AAV-PCSK9 increased the percentage of macrophages positive for Ki-67, an indicator of proliferating cells (Supplemental Figure 1A, B). Consistent with this in vivo finding, recombinant PCSK9 augmented M-CSF-induced cell proliferation in mouse bone marrow-derived macrophages (BMDMs) (Supplemental Figure 2A, B). Monocyte migration assay using the human monocytic cell line THP-1 showed that recombinant PCSK9 tended to promote MCP-1 (monocyte chemoattractant protein-1)-induced chemotaxis, suggesting lipid-independent effects of PCSK9 on monocyte migration (Supplemental Figure 2C). On the other hand, AAV-PCSK9 did not increase apoptosis determined by TUNEL staining in vein grafts of Ldlr-/- mice (Supplemental Figure 3A). Accordingly, AAV-PCSK9 did not change necrotic core area in the vein grafts (Supplemental Figure 3B).\nThe effects of Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) on collagen thinning and macrophage accumulation in vein graft lesions of Ldlr-/- mice. A, Picrosirius red staining of vein grafts without (top) or with (bottom) polarized light (n=12 and 11 for AAV-LacZ and AAV-PCSK9 group, respectively). Scale bars indicate 500 μm. Circle graphs show the percentage of thin (green) and thick (red) collagen fibers compared with total fibers. B, Intravital microscopy images of MMPSense 680 (red) and AminoSPARK750 (green) in vein grafts for visualization of MMP activity and macrophage accumulation, respectively (n=10 and 9 for AAV-LacZ and AAV-PCSK9 group, respectively). C, Mac3 (macrophages) staining in vein grafts. Scale bars indicate 500 μm. P value was calculated by Mann-Whitney U-test (thick collagen in A, macrophage accumulation mean fluorescence intensity in B) and unpaired Student t test (C). Data are reported as mean ± SEM.\n[SUBTITLE] AAV-PCSK9 Induces Macrophage Activation via an LDLR-Independent Route In Vivo [SUBSECTION] We previously reported that macrophage activation plays a key role in vein graft lesion development.8 To explore the effects of circulating PCSK9 on macrophage activation, we examined mRNA levels of pro-inflammatory (IL-1β [interleukin-1 beta], IL-6, TNFα, and MCP-1) and antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1 [transforming growth factor-beta 1], and IL-10) in the peritoneal F4/80+ macrophages of Ldlr-/- mice 1 or 4 weeks after intravenous injection of AAVs. AAV-PCSK9 promoted pro-inflammatory IL-1β, IL-6, TNFα, and MCP-1 mRNA levels at 1 week (Figure 3A). AAV-PCSK9 also suppressed Arginase-1 mRNA levels at 4 weeks when the increase in the mRNA levels of pro-inflammatory molecules at 1 week had subsided (Figure 3B). We have recently reported that predominant pathways of the vein grafts change over time during lesion development. In the same study, pathways with immune responses represent the early responsive proteins in proteomics analysis.9 These results indicate that PCSK9 can induce pro-inflammatory responses in macrophages initially and later impair resolution of inflammation in vivo.\nAdeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) induced macrophage activation in murine peritoneal macrophages via LDL receptor (LDLR)-independent mechanisms in vivo. mRNA levels of pro-inflammatory molecules (IL-1β, IL-6, TNFα, MCP-1) and antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1, IL-10) were measured in murine peritoneal macrophages of Ldlr-/- mice 1 week (A) or 4 weeks (B) after intravenous injection with AAV-LacZ or AAV-PCSK9 (n=7 to 8 per group). P value was calculated by Mann-Whitney U-test (TNFa in A, IL-1b, TNFa, MCP-1, Arginase-1, Ym1, and IL-10 in B) and unpaired Student t test. Data are reported as mean ± SEM.\nWe previously reported that macrophage activation plays a key role in vein graft lesion development.8 To explore the effects of circulating PCSK9 on macrophage activation, we examined mRNA levels of pro-inflammatory (IL-1β [interleukin-1 beta], IL-6, TNFα, and MCP-1) and antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1 [transforming growth factor-beta 1], and IL-10) in the peritoneal F4/80+ macrophages of Ldlr-/- mice 1 or 4 weeks after intravenous injection of AAVs. AAV-PCSK9 promoted pro-inflammatory IL-1β, IL-6, TNFα, and MCP-1 mRNA levels at 1 week (Figure 3A). AAV-PCSK9 also suppressed Arginase-1 mRNA levels at 4 weeks when the increase in the mRNA levels of pro-inflammatory molecules at 1 week had subsided (Figure 3B). We have recently reported that predominant pathways of the vein grafts change over time during lesion development. In the same study, pathways with immune responses represent the early responsive proteins in proteomics analysis.9 These results indicate that PCSK9 can induce pro-inflammatory responses in macrophages initially and later impair resolution of inflammation in vivo.\nAdeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) induced macrophage activation in murine peritoneal macrophages via LDL receptor (LDLR)-independent mechanisms in vivo. mRNA levels of pro-inflammatory molecules (IL-1β, IL-6, TNFα, MCP-1) and antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1, IL-10) were measured in murine peritoneal macrophages of Ldlr-/- mice 1 week (A) or 4 weeks (B) after intravenous injection with AAV-LacZ or AAV-PCSK9 (n=7 to 8 per group). P value was calculated by Mann-Whitney U-test (TNFa in A, IL-1b, TNFa, MCP-1, Arginase-1, Ym1, and IL-10 in B) and unpaired Student t test. Data are reported as mean ± SEM.\n[SUBTITLE] Endotoxin-Free Recombinant PCSK9 Induced Macrophage Activation in Peritoneal Macrophages of Ldlr\n\n-/- Mice In Vitro [SUBSECTION] To support our in vivo evidence, we examined in vitro the effects of physiologically relevant levels of recombinant mouse PCSK9 (0.1–2.5 μg/mL)35 on the mRNA levels of the same pro-inflammatory and antiinflammatory molecules examined in Ldlr-/- mouse macrophages. First, we conducted a time-course study of recombinant PCSK9-induced TNFα and Arginase-1 mRNA expression levels to determine an optimal time point for harvesting Ldlr-/- mouse macrophages. The increase of TNFα mRNA levels peaked 3 hours after stimulation, whereas the decrease of Arginase-1 mRNA levels plateaued 12 hours after stimulation with recombinant PCSK9 (Figure 4A). For all experiments, Ldlr-/- mouse macrophages were therefore harvested 3 and 12 hours after stimulation to measure pro- and antiinflammatory molecules, respectively. Peritoneal macrophages treated with recombinant PCSK9 exhibited increased mRNA levels of IL-1β, IL-6, TNFα, and MCP-1 in a concentration-dependent manner (Figure 4B). In contrast, recombinant PCSK9 did not decrease the mRNA levels of antiinflammatory molecules (Figure 4C). These results support the in vivo evidence of macrophage activation by circulating PCSK9 in an LDLR-independent fashion.\nRecombinant PCSK9 (proprotein convertase subtilisin/kexin 9) induced macrophage activation in LDL receptor (LDLR)-independent mechanisms in vitro. A, Time course of TNFα and Arginase-1 mRNA levels in peritoneal macrophages of wild-type mice after stimulation with recombinant mouse PCSK9 (MmPCSK9; 2.5 μg/mL) (n=5 per group). P value was calculated by Kruskal-Wallis test, followed by Dunn multiple comparison test. B, mRNA levels of pro-inflammatory molecules (IL-1β, IL-6, TNFα, MCP-1) and (C) antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1, IL-10) were measured in murine peritoneal macrophages from Ldlr-/- mice 3 and 12 hours after stimulation with recombinant mouse PCSK9 for pro-inflammatory and antiinflammatory molecules, respectively (n=9 per group). P value was calculated by 1-way ANOVA, followed by Dunnett multiple comparison test (IL-6, TNFa, MCP-1‚ MRC1) or Kruskal-Wallis test, followed by Dunn multiple comparison test. Data are reported as mean ± SEM.\nEndotoxin contamination in recombinant proteins can induce pro-inflammatory responses. Therefore, we chose a recombinant protein derived from a mammalian system, mouse myeloma cell line (NS0). The endotoxin level of recombinant mouse PCSK9 used in this study was 0.00441 EU/µg or less, well below the threshold of 0.1 EU/µg required for accurate cell-based assays.19 To exclude further endotoxin contamination in the recombinant PCSK9 protein affecting any measurements, we performed the chromogenic Limulus amebocyte lysate endotoxin assay. The final endotoxin level of the maximum dose of the recombinant protein (2.5 µg/mL) was 0.005 EU/mL or less, which coincides with the endotoxin levels of the commercially available endotoxin-free medium. In addition, mass spectrometry conducted on the solvent derived from reconstituted recombinant mouse PCSK9 protein solution did not produce signals, suggesting that there were no small molecule impurities present (Supplemental Figure 4A). Furthermore, stimulation with heat-inactivated recombinant mouse PCSK9 protein (2.5 μg/mL) caused no changes in the mRNA levels of IL-1β, TNF-α, and MCP-1, demonstrating that endotoxin contamination did not drive the effects observed (Supplemental Figure 4B).\nTo support our in vivo evidence, we examined in vitro the effects of physiologically relevant levels of recombinant mouse PCSK9 (0.1–2.5 μg/mL)35 on the mRNA levels of the same pro-inflammatory and antiinflammatory molecules examined in Ldlr-/- mouse macrophages. First, we conducted a time-course study of recombinant PCSK9-induced TNFα and Arginase-1 mRNA expression levels to determine an optimal time point for harvesting Ldlr-/- mouse macrophages. The increase of TNFα mRNA levels peaked 3 hours after stimulation, whereas the decrease of Arginase-1 mRNA levels plateaued 12 hours after stimulation with recombinant PCSK9 (Figure 4A). For all experiments, Ldlr-/- mouse macrophages were therefore harvested 3 and 12 hours after stimulation to measure pro- and antiinflammatory molecules, respectively. Peritoneal macrophages treated with recombinant PCSK9 exhibited increased mRNA levels of IL-1β, IL-6, TNFα, and MCP-1 in a concentration-dependent manner (Figure 4B). In contrast, recombinant PCSK9 did not decrease the mRNA levels of antiinflammatory molecules (Figure 4C). These results support the in vivo evidence of macrophage activation by circulating PCSK9 in an LDLR-independent fashion.\nRecombinant PCSK9 (proprotein convertase subtilisin/kexin 9) induced macrophage activation in LDL receptor (LDLR)-independent mechanisms in vitro. A, Time course of TNFα and Arginase-1 mRNA levels in peritoneal macrophages of wild-type mice after stimulation with recombinant mouse PCSK9 (MmPCSK9; 2.5 μg/mL) (n=5 per group). P value was calculated by Kruskal-Wallis test, followed by Dunn multiple comparison test. B, mRNA levels of pro-inflammatory molecules (IL-1β, IL-6, TNFα, MCP-1) and (C) antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1, IL-10) were measured in murine peritoneal macrophages from Ldlr-/- mice 3 and 12 hours after stimulation with recombinant mouse PCSK9 for pro-inflammatory and antiinflammatory molecules, respectively (n=9 per group). P value was calculated by 1-way ANOVA, followed by Dunnett multiple comparison test (IL-6, TNFa, MCP-1‚ MRC1) or Kruskal-Wallis test, followed by Dunn multiple comparison test. Data are reported as mean ± SEM.\nEndotoxin contamination in recombinant proteins can induce pro-inflammatory responses. Therefore, we chose a recombinant protein derived from a mammalian system, mouse myeloma cell line (NS0). The endotoxin level of recombinant mouse PCSK9 used in this study was 0.00441 EU/µg or less, well below the threshold of 0.1 EU/µg required for accurate cell-based assays.19 To exclude further endotoxin contamination in the recombinant PCSK9 protein affecting any measurements, we performed the chromogenic Limulus amebocyte lysate endotoxin assay. The final endotoxin level of the maximum dose of the recombinant protein (2.5 µg/mL) was 0.005 EU/mL or less, which coincides with the endotoxin levels of the commercially available endotoxin-free medium. In addition, mass spectrometry conducted on the solvent derived from reconstituted recombinant mouse PCSK9 protein solution did not produce signals, suggesting that there were no small molecule impurities present (Supplemental Figure 4A). Furthermore, stimulation with heat-inactivated recombinant mouse PCSK9 protein (2.5 μg/mL) caused no changes in the mRNA levels of IL-1β, TNF-α, and MCP-1, demonstrating that endotoxin contamination did not drive the effects observed (Supplemental Figure 4B).\n[SUBTITLE] A combination of Unbiased Global Transcriptomics and Network-Based Hyperedge Entanglement Prediction Analysis Revealed Potential Targets of PCSK9 in Macrophages [SUBSECTION] We explored further LDLR-independent pro-inflammatory signaling pathways in an unbiased manner, using RNA-sequencing of Ldlr-/- mouse macrophages stimulated with recombinant PCSK9. When analyzing the differentially expressed transcripts (FC>2.0, q<0.001), we observed 46 transcripts significantly increased by PCSK9 stimulation in Ldlr-/- macrophages compared with controls (Figure 5A). A hierarchical heat map of Ldlr-/- mouse macrophages filtered by differentially expressed transcripts showed a clear separation between control (no stimulus) and PCSK9 group (Figure 5B). We provided statistically significantly overrepresented transcripts ranked according to their fold change abundance (Supplemental Table 3A) to further examine these differentially expressed transcripts. PCSK9 treatment increased many transcripts implicated in pro-inflammatory responses in Ldlr-/- macrophages (eg, IL-1b, TNF, CXCL2, NF-κB [nuclear factor-kappa B]-related genes).19\nTranscriptomics, network analysis, and hyperedge entanglement prediction (HEP) of mouse macrophages stimulated with recombinant PCSK9 (proprotein convertase subtilisin/kexin 9). A, Volcano plot for Ldlr-/- mouse macrophages stimulated with recombinant PCSK9. Red markers indicate significantly expressed transcripts with a fold change cutoff of 2.0 and adjusted P value cutoff of 0.001. The enriched transcripts that were previously reported were denoted with their names. B, Heat map of the transcriptomics of Ldlr-/- mouse macrophages (FC>2, q<0.001-filtered data for the difference between macrophages stimulated with and without recombinant PCSK9, n=4 per group). C, The schematic network visualization by HEP prediction. The significantly predicted association of PCSK9 to the 24 mapped inflammation-related transcripts derived from the list of transcripts (FC>2.0, q<0.05) indicates every unknown pairs of interaction (dashed line) that have to be proven yet experimentally. One of them was validated by silencing Sdc4 (syndecan-4) mRNA (blue dashed line). All other links represent known physical interactions (solid gray lines). The node size illustrates the degree of the proteins in the protein-protein interaction network. D, Network analysis of the 24 overrepresented transcripts in Ldlr-/- mouse macrophages using STRING database. Nodes are colored according to k-means clustering (number of clusters=3). Only connected nodes are shown here.\nTo provide additional mechanistic links, we attempted to identify PCSK9-regulated proteins other than LDLR that mediate the aforementioned pro-inflammatory downstream responses. Many cascading effects in the transcriptional regulatory machinery could control the differentially expressed mRNAs. This set of differentially expressed transcripts were identified as the result of a genome-wide transcriptional profiling method, which might not necessarily interact directly with PCSK9. To estimate the likelihood of PCSK9 directly interacting with those differentially expressed transcripts, we tested this hypothesis verification through network computational analysis. In brief, PCSK9 can be seen as a node in a protein-protein interaction network,36 and the set of differentially expressed transcripts can be seen as a hyperedge projected on the protein-protein interaction network (a group of nodes that are associated by a common feature of biological functions). Muscoloni et al recently introduced a novel algorithm for HEP that exploits an ensemble of link predictors and that provides a level of statistical significance for the entanglement between each node and hyperedge that does not have a direct interaction in the network.20 We then performed the HEP analysis to quantify the significance of the entanglement between PCSK9 and the hyperedge of 24 differentially expressed transcripts (FC>2.0, q<0.05) in the dataset (Supplemental Figure 5A and 5B, Supplemental Table 3B). The resulting P values of 7/12 node-node link prediction methods (RA, CH2-L2, CH3-L2, and all 4 L3-based methods) established a significant association between PCSK9 and the hyperedge of the 24 differentially expressed transcripts (Figure 5C; Supplemental Table 2). These data suggest that any of the 24 differentially expressed transcripts have a high likelihood of directly interacting with PCSK9, enabling us to validate a direct interaction between PCSK9 and one of these 24 differentially expressed transcripts. To understand the biological relevance of these transcripts, we explored the network connectivity of these transcripts identified by HEP analysis using the STRING database (confidence interaction score ≥0.7).37 The network analysis showed a highly clustered network (average local clustering coefficient: 0.547) containing 24 nodes with 52 edges (expected number of edges=3), providing significantly more interactions than expected for a random set of genes of similar size (P value ≤1.0e−16). The k-means clustering (number of clusters=3) showed that one of these 3 clusters mainly consisted of the NF-κB signaling pathway based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (IL-1b, CXCL1 [chemokine (C-X-C motif) ligand 1], CXCL2 [chemokine (C-X-C motif) ligand 2], ICAM1, VCAM1 [vascular cell adhesion molecule 1], RELB [RELB Proto-Oncogene, NF-kB subunit], NFKB2 [nuclear factor kappa B subunit 2]), and the other 2 clusters included OLR1 (LOX-1) and SDC4, respectively (Figure 5D).\nIt is already reported that NF-κB signaling pathway is involved in PCSK9-induced atherosclerotic inflammation.38 In the present study, AAV-PCSK9 promoted NF-κB p65 mRNA level in the peritoneal F4/80+ macrophages of Ldlr-/- mice (Supplemental Figure 6A). Inhibition of NF-κB signaling pathway with IκB kinase inhibitor, 2-[(aminocarbonyl)amino]-5-(4-fluorophenyl)-3-thiophenecarboxamide (TPCA1), decreased pro-inflammatory responses to recombinant PCSK9 in Ldlr-/- mouse macrophages (Supplemental Figure 6B). These data indicate that NF-κB may mediate PCSK9-induced pro-inflammatory responses in vein graft lesions.\nPCSK9 stimulates the expression of LOX-1, which in turn takes up ox-LDL in macrophages.39 AAV-PCSK9 increased foam cell formation determined by Oil red O positive area in vein grafts (Supplemental Figure 7A). We further found that recombinant PCSK9 increased ox-LDL uptake in bone marrow-derived macrophages from Ldlr-/- mice (Supplemental Figure 7B). These results indicate that PCSK9 also induces foam cell formation by increased ox-LDL uptake, contributing not only to atherosclerotic but also to vein graft lesion development.\nAmong the members of this differentially expressed transcript hyperedge, we found that SDC4 included in the last cluster could be a novel target of PCSK9 (Figure 5D). While SDC4 was not a high-ranking molecule in the transcript ranking lists (30th in Supplemental Table 3A and 67th in Supplemental Table 3B), its fold change after stimulation with recombinant PCSK9 in Ldlr-/- mice macrophages was statistically significant (2.65, log2FC>1). AAV-PCSK9 also increased SDC4 mRNA level in the peritoneal F4/80+ macrophages of Ldlr-/- mice (Figure 6A). SDC4 is a heparan sulfate proteoglycan40 expressed on the surface of human macrophages.41 In addition, SDC4 mRNA is increased in M(LPS) but not M(IL-4/IL-13) or M(IL-10) in bone marrow-derived macrophages.42 A recent study reported that heparan sulfate proteoglycans physically bind to PCSK9 on the hepatocyte surface43 However, the investigators did not address the interaction between PCSK9 and any specific individual syndecans. Our in silico protein-protein docking analysis using HPEPDOCK, a web server for protein-protein docking based on a hierarchical algorithm, predicted binding between PCSK9 and SDC4 (Supplemental Figure 8A).44 We entered the pdb files of PCSK9 (PDB ID: 2PMW), LDLR (PDB ID: 1N7D), and SDC4 (PDB ID: 1EJP) into HPEPDOCK server, which in turn presented the top 100 docking models based on docking energy minimized scores. Protein-protein docking analysis showed that the binding efficiency of SDC4 and PCSK9 (−212.23 to −235.6 kJ/mol) was comparable to that of LDLR and PCSK9 (−259.85 to −303.21 kJ/mol) based on the docking score of the top 5 binding predictions (Supplemental Figure 8A). Besides, the ligand root mean square deviation, often used to evaluate the correctness of the docking geometry and optimal superimposition of the receptor-ligand binding, was lower in the binding of SDC4 and PCSK9 (39.12–85.16) compared with that of LDLR and PCSK9 (43.76–156.25) for the top 5 binding predictions, suggesting better binding between SDC4 and PCSK9 than in LDLR and PCSK9. Furthermore, co-immunoprecipitation analysis of mouse liver tissue lysates validated the occurrence of the predicted PCSK9-SDC4 binding in vivo (Supplemental Figure 8B).\nRNA silencing validated SDC4 (syndecan-4) as a potential target of PCSK9 (proprotein convertase subtilisin/kexin 9)-induced pro-inflammatory response in macrophages, identified via hyperedge entanglement prediction (HEP) prediction. A, SDC4 mRNA level was measured in murine peritoneal macrophages of Ldlr-/- mice 1 week after intravenous injection with AAV-LacZ or AAV-PCSK9 (n=7 per group). B, mRNA levels of SDC4, IL-1b, NLRP3, CD40, and ICAM1 were measured by RT-qPCR in mouse Ldlr-/- peritoneal macrophages after pretreatment with siSDC4 or siControl (Ctrl) for 48 hours and stimulation with 2.5 μg/mL recombinant mouse PCSK9 (MmPCSK9) for 3 hours (n=4–5). P value was calculated by 1-way ANOVA, followed by Bonferroni multiple comparison test. Error bars indicate ± SEM. Data are reported as mean ± SEM.\nWe explored further LDLR-independent pro-inflammatory signaling pathways in an unbiased manner, using RNA-sequencing of Ldlr-/- mouse macrophages stimulated with recombinant PCSK9. When analyzing the differentially expressed transcripts (FC>2.0, q<0.001), we observed 46 transcripts significantly increased by PCSK9 stimulation in Ldlr-/- macrophages compared with controls (Figure 5A). A hierarchical heat map of Ldlr-/- mouse macrophages filtered by differentially expressed transcripts showed a clear separation between control (no stimulus) and PCSK9 group (Figure 5B). We provided statistically significantly overrepresented transcripts ranked according to their fold change abundance (Supplemental Table 3A) to further examine these differentially expressed transcripts. PCSK9 treatment increased many transcripts implicated in pro-inflammatory responses in Ldlr-/- macrophages (eg, IL-1b, TNF, CXCL2, NF-κB [nuclear factor-kappa B]-related genes).19\nTranscriptomics, network analysis, and hyperedge entanglement prediction (HEP) of mouse macrophages stimulated with recombinant PCSK9 (proprotein convertase subtilisin/kexin 9). A, Volcano plot for Ldlr-/- mouse macrophages stimulated with recombinant PCSK9. Red markers indicate significantly expressed transcripts with a fold change cutoff of 2.0 and adjusted P value cutoff of 0.001. The enriched transcripts that were previously reported were denoted with their names. B, Heat map of the transcriptomics of Ldlr-/- mouse macrophages (FC>2, q<0.001-filtered data for the difference between macrophages stimulated with and without recombinant PCSK9, n=4 per group). C, The schematic network visualization by HEP prediction. The significantly predicted association of PCSK9 to the 24 mapped inflammation-related transcripts derived from the list of transcripts (FC>2.0, q<0.05) indicates every unknown pairs of interaction (dashed line) that have to be proven yet experimentally. One of them was validated by silencing Sdc4 (syndecan-4) mRNA (blue dashed line). All other links represent known physical interactions (solid gray lines). The node size illustrates the degree of the proteins in the protein-protein interaction network. D, Network analysis of the 24 overrepresented transcripts in Ldlr-/- mouse macrophages using STRING database. Nodes are colored according to k-means clustering (number of clusters=3). Only connected nodes are shown here.\nTo provide additional mechanistic links, we attempted to identify PCSK9-regulated proteins other than LDLR that mediate the aforementioned pro-inflammatory downstream responses. Many cascading effects in the transcriptional regulatory machinery could control the differentially expressed mRNAs. This set of differentially expressed transcripts were identified as the result of a genome-wide transcriptional profiling method, which might not necessarily interact directly with PCSK9. To estimate the likelihood of PCSK9 directly interacting with those differentially expressed transcripts, we tested this hypothesis verification through network computational analysis. In brief, PCSK9 can be seen as a node in a protein-protein interaction network,36 and the set of differentially expressed transcripts can be seen as a hyperedge projected on the protein-protein interaction network (a group of nodes that are associated by a common feature of biological functions). Muscoloni et al recently introduced a novel algorithm for HEP that exploits an ensemble of link predictors and that provides a level of statistical significance for the entanglement between each node and hyperedge that does not have a direct interaction in the network.20 We then performed the HEP analysis to quantify the significance of the entanglement between PCSK9 and the hyperedge of 24 differentially expressed transcripts (FC>2.0, q<0.05) in the dataset (Supplemental Figure 5A and 5B, Supplemental Table 3B). The resulting P values of 7/12 node-node link prediction methods (RA, CH2-L2, CH3-L2, and all 4 L3-based methods) established a significant association between PCSK9 and the hyperedge of the 24 differentially expressed transcripts (Figure 5C; Supplemental Table 2). These data suggest that any of the 24 differentially expressed transcripts have a high likelihood of directly interacting with PCSK9, enabling us to validate a direct interaction between PCSK9 and one of these 24 differentially expressed transcripts. To understand the biological relevance of these transcripts, we explored the network connectivity of these transcripts identified by HEP analysis using the STRING database (confidence interaction score ≥0.7).37 The network analysis showed a highly clustered network (average local clustering coefficient: 0.547) containing 24 nodes with 52 edges (expected number of edges=3), providing significantly more interactions than expected for a random set of genes of similar size (P value ≤1.0e−16). The k-means clustering (number of clusters=3) showed that one of these 3 clusters mainly consisted of the NF-κB signaling pathway based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (IL-1b, CXCL1 [chemokine (C-X-C motif) ligand 1], CXCL2 [chemokine (C-X-C motif) ligand 2], ICAM1, VCAM1 [vascular cell adhesion molecule 1], RELB [RELB Proto-Oncogene, NF-kB subunit], NFKB2 [nuclear factor kappa B subunit 2]), and the other 2 clusters included OLR1 (LOX-1) and SDC4, respectively (Figure 5D).\nIt is already reported that NF-κB signaling pathway is involved in PCSK9-induced atherosclerotic inflammation.38 In the present study, AAV-PCSK9 promoted NF-κB p65 mRNA level in the peritoneal F4/80+ macrophages of Ldlr-/- mice (Supplemental Figure 6A). Inhibition of NF-κB signaling pathway with IκB kinase inhibitor, 2-[(aminocarbonyl)amino]-5-(4-fluorophenyl)-3-thiophenecarboxamide (TPCA1), decreased pro-inflammatory responses to recombinant PCSK9 in Ldlr-/- mouse macrophages (Supplemental Figure 6B). These data indicate that NF-κB may mediate PCSK9-induced pro-inflammatory responses in vein graft lesions.\nPCSK9 stimulates the expression of LOX-1, which in turn takes up ox-LDL in macrophages.39 AAV-PCSK9 increased foam cell formation determined by Oil red O positive area in vein grafts (Supplemental Figure 7A). We further found that recombinant PCSK9 increased ox-LDL uptake in bone marrow-derived macrophages from Ldlr-/- mice (Supplemental Figure 7B). These results indicate that PCSK9 also induces foam cell formation by increased ox-LDL uptake, contributing not only to atherosclerotic but also to vein graft lesion development.\nAmong the members of this differentially expressed transcript hyperedge, we found that SDC4 included in the last cluster could be a novel target of PCSK9 (Figure 5D). While SDC4 was not a high-ranking molecule in the transcript ranking lists (30th in Supplemental Table 3A and 67th in Supplemental Table 3B), its fold change after stimulation with recombinant PCSK9 in Ldlr-/- mice macrophages was statistically significant (2.65, log2FC>1). AAV-PCSK9 also increased SDC4 mRNA level in the peritoneal F4/80+ macrophages of Ldlr-/- mice (Figure 6A). SDC4 is a heparan sulfate proteoglycan40 expressed on the surface of human macrophages.41 In addition, SDC4 mRNA is increased in M(LPS) but not M(IL-4/IL-13) or M(IL-10) in bone marrow-derived macrophages.42 A recent study reported that heparan sulfate proteoglycans physically bind to PCSK9 on the hepatocyte surface43 However, the investigators did not address the interaction between PCSK9 and any specific individual syndecans. Our in silico protein-protein docking analysis using HPEPDOCK, a web server for protein-protein docking based on a hierarchical algorithm, predicted binding between PCSK9 and SDC4 (Supplemental Figure 8A).44 We entered the pdb files of PCSK9 (PDB ID: 2PMW), LDLR (PDB ID: 1N7D), and SDC4 (PDB ID: 1EJP) into HPEPDOCK server, which in turn presented the top 100 docking models based on docking energy minimized scores. Protein-protein docking analysis showed that the binding efficiency of SDC4 and PCSK9 (−212.23 to −235.6 kJ/mol) was comparable to that of LDLR and PCSK9 (−259.85 to −303.21 kJ/mol) based on the docking score of the top 5 binding predictions (Supplemental Figure 8A). Besides, the ligand root mean square deviation, often used to evaluate the correctness of the docking geometry and optimal superimposition of the receptor-ligand binding, was lower in the binding of SDC4 and PCSK9 (39.12–85.16) compared with that of LDLR and PCSK9 (43.76–156.25) for the top 5 binding predictions, suggesting better binding between SDC4 and PCSK9 than in LDLR and PCSK9. Furthermore, co-immunoprecipitation analysis of mouse liver tissue lysates validated the occurrence of the predicted PCSK9-SDC4 binding in vivo (Supplemental Figure 8B).\nRNA silencing validated SDC4 (syndecan-4) as a potential target of PCSK9 (proprotein convertase subtilisin/kexin 9)-induced pro-inflammatory response in macrophages, identified via hyperedge entanglement prediction (HEP) prediction. A, SDC4 mRNA level was measured in murine peritoneal macrophages of Ldlr-/- mice 1 week after intravenous injection with AAV-LacZ or AAV-PCSK9 (n=7 per group). B, mRNA levels of SDC4, IL-1b, NLRP3, CD40, and ICAM1 were measured by RT-qPCR in mouse Ldlr-/- peritoneal macrophages after pretreatment with siSDC4 or siControl (Ctrl) for 48 hours and stimulation with 2.5 μg/mL recombinant mouse PCSK9 (MmPCSK9) for 3 hours (n=4–5). P value was calculated by 1-way ANOVA, followed by Bonferroni multiple comparison test. Error bars indicate ± SEM. Data are reported as mean ± SEM.\n[SUBTITLE] SDC4 Silencing Suppresses PCSK9-Induced Pro-Inflammatory Responses in Macrophages [SUBSECTION] We further validated our prediction that SDC4 is a potential target of PCSK9 using siRNA silencing of mouse Sdc4 in mouse peritoneal macrophages. Silencing Sdc4 with siSDC4 treatment achieved 93% reduction of SDC4 mRNA expression (Figure 6B). siSDC4 was then used to determine whether it could suppress PCSK9-induced pro-inflammatory molecules, including those predicted to be linked with PCSK9 by the HEP prediction (Figure 5C). siSDC4 suppressed mRNAs of the pro-inflammatory molecules IL-1β, NLRP3, CD40, and ICAM1, but not TNFα, MCP-1, TLR2, CXCL1, CXCL2, or CXCL10 after stimulation with recombinant PCSK9 (Figure 6B, Supplemental Figure 9). These data suggest that SDC4 partially regulates a PCSK9-induced pro-inflammatory response. Furthermore, these mRNA silencing results substantiate the validity of the prediction, yielding the integration of PCSK9 in our hyperedge of 24 disconnected differentially expressed transcripts.\nWe further validated our prediction that SDC4 is a potential target of PCSK9 using siRNA silencing of mouse Sdc4 in mouse peritoneal macrophages. Silencing Sdc4 with siSDC4 treatment achieved 93% reduction of SDC4 mRNA expression (Figure 6B). siSDC4 was then used to determine whether it could suppress PCSK9-induced pro-inflammatory molecules, including those predicted to be linked with PCSK9 by the HEP prediction (Figure 5C). siSDC4 suppressed mRNAs of the pro-inflammatory molecules IL-1β, NLRP3, CD40, and ICAM1, but not TNFα, MCP-1, TLR2, CXCL1, CXCL2, or CXCL10 after stimulation with recombinant PCSK9 (Figure 6B, Supplemental Figure 9). These data suggest that SDC4 partially regulates a PCSK9-induced pro-inflammatory response. Furthermore, these mRNA silencing results substantiate the validity of the prediction, yielding the integration of PCSK9 in our hyperedge of 24 disconnected differentially expressed transcripts.\n[SUBTITLE] Local Production of PCSK9 in the Liver Was Greater Than in Vein Graft Lesions [SUBSECTION] Our study demonstrated that circulating PCSK9 produced various changes in Ldlr-/- mice. We, however, attempted to examine the potential impact of local PCSK9 production in vein grafts. Immunohistochemical analysis using an anti-PCSK9 antibody in previous studies showed that vascular smooth muscle cells in human atherosclerotic plaque and collar-induced neointima of murine carotid artery express PCSK9.45,46 Based on these results, we examined whether locally produced PCSK9 in vein grafts may also contribute to vein graft lesion development. The local expression of PCSK9, as determined by in situ hybridization in the intima of vein grafts, was minimal compared with that in the liver (Figure 1B; Supplemental Figure 10A). RT-qPCR analysis further confirmed numerically significantly less PCSK9 mRNA expression in vein grafts than in the liver, the primary source of circulating PCSK9 (Supplemental Figure 10B). Moreover, we examined which cells within vein grafts can produce PCSK9 in humans by comparing PCSK9 mRNA levels in human saphenous vein endothelial cells, smooth muscle cells, and human primary macrophages derived from peripheral blood mononuclear cells. While we detected substantial PCSK9 expression in human saphenous vein smooth muscle cells, the level was significantly lower than that in HepG2 (liver) cells (Supplemental Figure 10C). Human primary macrophages and human saphenous vein endothelial cells contained minimal PCSK9 mRNA (Supplemental Figure 10C). Recent studies reported a modest increase of PCSK9 mRNA and protein levels in macrophages exposed to pro-inflammatory stimuli.39,47 We, therefore, examined PCSK9 mRNA expression in human primary macrophages treated with pro-inflammatory stimuli. Although LPS and IFNγ induced a pro-inflammatory response in the macrophages, as determined by TNFα mRNA expression (Supplemental Figures 10D and 10E, right graphs), PCSK9 mRNA expression did not increase after LPS or IFNγ (Supplemental Figures 10D and 10E, left graphs). These results indicate that smooth muscle cells (SMC), but not macrophages, may primarily produce PCSK9 in vein grafts, but to a much lower extent than the liver.\nOur study demonstrated that circulating PCSK9 produced various changes in Ldlr-/- mice. We, however, attempted to examine the potential impact of local PCSK9 production in vein grafts. Immunohistochemical analysis using an anti-PCSK9 antibody in previous studies showed that vascular smooth muscle cells in human atherosclerotic plaque and collar-induced neointima of murine carotid artery express PCSK9.45,46 Based on these results, we examined whether locally produced PCSK9 in vein grafts may also contribute to vein graft lesion development. The local expression of PCSK9, as determined by in situ hybridization in the intima of vein grafts, was minimal compared with that in the liver (Figure 1B; Supplemental Figure 10A). RT-qPCR analysis further confirmed numerically significantly less PCSK9 mRNA expression in vein grafts than in the liver, the primary source of circulating PCSK9 (Supplemental Figure 10B). Moreover, we examined which cells within vein grafts can produce PCSK9 in humans by comparing PCSK9 mRNA levels in human saphenous vein endothelial cells, smooth muscle cells, and human primary macrophages derived from peripheral blood mononuclear cells. While we detected substantial PCSK9 expression in human saphenous vein smooth muscle cells, the level was significantly lower than that in HepG2 (liver) cells (Supplemental Figure 10C). Human primary macrophages and human saphenous vein endothelial cells contained minimal PCSK9 mRNA (Supplemental Figure 10C). Recent studies reported a modest increase of PCSK9 mRNA and protein levels in macrophages exposed to pro-inflammatory stimuli.39,47 We, therefore, examined PCSK9 mRNA expression in human primary macrophages treated with pro-inflammatory stimuli. Although LPS and IFNγ induced a pro-inflammatory response in the macrophages, as determined by TNFα mRNA expression (Supplemental Figures 10D and 10E, right graphs), PCSK9 mRNA expression did not increase after LPS or IFNγ (Supplemental Figures 10D and 10E, left graphs). These results indicate that smooth muscle cells (SMC), but not macrophages, may primarily produce PCSK9 in vein grafts, but to a much lower extent than the liver.\n[SUBTITLE] Macrophages Rather Than Endothelial Cells and SMCs May Mediate the Effects of PCSK9 on Vein Graft Lesion Development [SUBSECTION] Although our study focuses on macrophages, we examined the role of PCSK9 in the activation of endothelial cells and SMCs because other investigators reported potential interactions between PCSK9 and these cell types.46,48 Endothelial cell activation precedes vein graft lesion development.49 Due to the scarcity of mRNA in CD31-positive endothelial cells in vein grafts, we sorted them from the liver and lungs of Ldlr-/- mice injected with AAV-PCSK9 instead to examine the effects of PCSK9 on endothelial cell activation in an LDLR-independent fashion. AAV-PCSK9 did not increase mRNA levels of adhesion molecules, such as VCAM-1, ICAM-1, and E-selectin, in EC from either organ when compared with AAV-LacZ (Supplemental Figure 11A). We further found in vitro that recombinant PCSK9 did not increase mRNA levels of these adhesion molecules in human saphenous vein endothelial cells (Supplemental Figure 11B). These results indirectly indicate that PCSK9 may not induce endothelial activation in vein grafts.\nSMC modulation, migration, and proliferation contribute to the development of vascular disorders, including atherosclerosis50 and vein graft.51 A recent study showed that PCSK9 may sustain SMC dedifferentiation, migration, and proliferation in neointimal hyperplasia in response to vascular injury,46 suggesting the involvement of SMCs in PCSK9-induced vein graft lesion development. To explore this possibility, we examined the effects of recombinant PCSK9 on SMC dedifferentiation, migration, and proliferation in human saphenous vein smooth muscle cells. Recombinant PCSK9 slightly decreased SM22α mRNA levels (0.74 and 0.68-fold for 1.0 and 5.0 µg/mL, respectively), but did not affect differentiation molecules, such as αSMA, calponin, SM1, or SM2 (Supplemental Figure 12A). In addition, recombinant PCSK9 did not affect PDGF-induced migration activity of human saphenous vein smooth muscle cells (Supplemental Figure 12B). A BrdU assay using human saphenous vein smooth muscle cells showed that recombinant PCSK9 did not enhance 10% fetal bovine serum-induced proliferation (Supplemental Figure 12C). AAV-PCSK9 did not affect the SMC content in vein grafts (Supplemental Figure 12D). Collectively, in our experimental settings, our data did not provide the mechanistic evidence in vitro and in vivo that activation of endothelial cells or SMCs contribute to PCSK9-induced vein graft lesion formation. These results indicate that circulating PCSK9 promotes vein graft lesion development predominantly through macrophage activation.\nAlthough our study focuses on macrophages, we examined the role of PCSK9 in the activation of endothelial cells and SMCs because other investigators reported potential interactions between PCSK9 and these cell types.46,48 Endothelial cell activation precedes vein graft lesion development.49 Due to the scarcity of mRNA in CD31-positive endothelial cells in vein grafts, we sorted them from the liver and lungs of Ldlr-/- mice injected with AAV-PCSK9 instead to examine the effects of PCSK9 on endothelial cell activation in an LDLR-independent fashion. AAV-PCSK9 did not increase mRNA levels of adhesion molecules, such as VCAM-1, ICAM-1, and E-selectin, in EC from either organ when compared with AAV-LacZ (Supplemental Figure 11A). We further found in vitro that recombinant PCSK9 did not increase mRNA levels of these adhesion molecules in human saphenous vein endothelial cells (Supplemental Figure 11B). These results indirectly indicate that PCSK9 may not induce endothelial activation in vein grafts.\nSMC modulation, migration, and proliferation contribute to the development of vascular disorders, including atherosclerosis50 and vein graft.51 A recent study showed that PCSK9 may sustain SMC dedifferentiation, migration, and proliferation in neointimal hyperplasia in response to vascular injury,46 suggesting the involvement of SMCs in PCSK9-induced vein graft lesion development. To explore this possibility, we examined the effects of recombinant PCSK9 on SMC dedifferentiation, migration, and proliferation in human saphenous vein smooth muscle cells. Recombinant PCSK9 slightly decreased SM22α mRNA levels (0.74 and 0.68-fold for 1.0 and 5.0 µg/mL, respectively), but did not affect differentiation molecules, such as αSMA, calponin, SM1, or SM2 (Supplemental Figure 12A). In addition, recombinant PCSK9 did not affect PDGF-induced migration activity of human saphenous vein smooth muscle cells (Supplemental Figure 12B). A BrdU assay using human saphenous vein smooth muscle cells showed that recombinant PCSK9 did not enhance 10% fetal bovine serum-induced proliferation (Supplemental Figure 12C). AAV-PCSK9 did not affect the SMC content in vein grafts (Supplemental Figure 12D). Collectively, in our experimental settings, our data did not provide the mechanistic evidence in vitro and in vivo that activation of endothelial cells or SMCs contribute to PCSK9-induced vein graft lesion formation. These results indicate that circulating PCSK9 promotes vein graft lesion development predominantly through macrophage activation.", "In vivo studies have suggested a direct link between PCSK9 and atherosclerosis28,29; however, no evidence has yet implicated PCSK9 in vein graft lesion development. Furthermore, these in vivo studies indicate that the underlying mechanism of atherosclerotic lesion development by PCSK9 may depend mainly on LDLR. Whether PCSK9 can exert pro-inflammatory effects via LDLR-independent mechanisms is unclear. To test the hypothesis that circulating PCSK9 promotes vein graft lesion development in vivo, we used experimental vein grafts in mice.21 A single intravenous injection of AAV-PCSK9 induced liver production of PCSK9. To exclude potential systemic toxicity of AAV, AAV-LacZ was developed and used as a control (Figure 1A; Online Supplemental Methods). All experiments used Ldlr-/- mice to explore LDLR-independent mechanisms unless otherwise noted.\nThe effects of Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) on vein graft lesion development in Ldlr-/- mice. A, An experimental protocol for the treatments in Ldlr-/- mice. B, In situ hybridization of PCSK9 (red) in the liver of Ldlr-/- mice 1 week after intravenous injection with AAV-LacZ or AAV-PCSK9. Nuclei are visualized with DAPI. The data represent 4 mice per group. Scale bars indicate 100 μm. C, PCSK9 mRNA level in the liver of AAV treated Ldlr-/- mice (n=12 and 11 for AAV-LacZ and AAV-PCSK9 groups, respectively), and serum levels of PCSK9, total cholesterol, and triglyceride in AAV treated Ldlr-/- mice (n=11 per group). D, Left, ultrasonography images of vein grafts from Ldlr-/- mice treated with AAV-LacZ or AAV-PCSK9 28 days after implantation. The red, green, and yellow dotted lines indicate lumen, near wall, and far wall, respectively. Right, The quantitative analysis of lumen and vessel wall area in short axis view and 3D reconstructed vessel wall volume of vein grafts by ultrasonography (n=12 and 11 for AAV-LacZ and AAV-PCSK9 groups, respectively). E, Left, Masson’s trichrome staining of vein grafts from Ldlr-/- mice treated with AAV-LacZ or AAV-PCSK9 28 days after implantation. Scale bars indicate 500 μm. Right, The quantitative analysis of intimal area, intimal and media/adventitia thickness, and lumen and vessel diameter of vein grafts (n=12 and 11 for AAV-LacZ and AAV-PCSK9 group, respectively). P value was calculated by Mann-Whitney U-test (triglyceride in C, media/adventitia thickness, lumen diameter, and vessel diameter in E) and unpaired Student t test (A, B, and D). Data are reported as mean ± SEM.\nBody weight and blood pressure did not differ significantly between the AAV-LacZ and AAV-PCSK9 groups 4 weeks after vein graft implantation (Supplemental Table 1). In situ hybridization showed that AAV-PCSK9 markedly increased PCSK9 mRNA levels in the liver (Figure 1B). AAV-PCSK9 increased PCSK9 mRNA expression in the liver and serum levels of PCSK9 (Figure 1C; Supplemental Table 1). Serum levels of total cholesterol and triglycerides did not differ between the AAV-LacZ and AAV-PCSK9 groups (Figure 1C; Supplemental Table 1). These findings indicate that the effects of AAV-PCSK9 on vein graft lesions described below did not depend on blood cholesterol and triglyceride concentrations (Supplemental Table 1).\nNoninvasive ultrasonography visualized that AAV-PCSK9 injection caused increased vessel wall area, but not lumen area, in the short axis view and 3-dimensional reconstructed vessel wall volume of vein grafts (Figure 1D). Consistent with the ultrasonography, histological analysis by Masson trichrome staining showed that AAV-PCSK9 increased intimal area and thickness of vein grafts. Lumen diameter, vessel diameter, and media/adventitia thickness did not differ significantly, suggesting compensatory outward remodeling of vein grafts (Figure 1E).", "Evidence suggests that vein grafts in humans can develop lesions similar to those of advanced arterial atherosclerosis, and plaque rupture can occur in inflamed vein grafts.30,31 Therefore, exploring vein graft lesion composition in depth is critical to understanding what contributes to this occurrence. We then examined the collagen content of vein grafts by picrosirius red staining. AAV-PCSK9 increased the percentage of thin collagen fibers as a ratio of the total collagen fibers in the vein grafts, suggesting that circulating PCSK9 induced pathological features similar to those in thin-capped atherosclerotic plaques (Figure 2A). In vivo molecular imaging further provided insight into the effects of circulating PCSK9 on matrix metalloproteinase activity and macrophage accumulation in vein grafts. We intravenously co-injected 2 spectrally different imaging agents that elaborate near-infrared signals for visualization of matrix metalloproteinase activity (MMPsense, 680 nm) and macrophage accumulation (AminoSPARK, 750 nm). Following the evidence that activated macrophages express MMPs that degrade interstitial collagen as previously reported by our group,32,33 in vivo molecular imaging demonstrated that increased matrix metalloproteinase activity co-localized with accumulated macrophages (Figure 2B, left). In addition, AAV-PCSK9 treatment promoted matrix metalloproteinase activity and macrophage accumulation in vein grafts (Figure 2B), suggesting a potential mechanism of collagen remodeling as determined by Picrosirius red staining. Immunohistochemical analysis further demonstrated increased macrophage accumulation in vein grafts by AAV-PCSK9 (Mac3; Figure 2C). Macrophage proliferation and migration may contribute to the mechanisms of macrophage accumulation in vascular diseases.34 Supporting this mechanism, AAV-PCSK9 increased the percentage of macrophages positive for Ki-67, an indicator of proliferating cells (Supplemental Figure 1A, B). Consistent with this in vivo finding, recombinant PCSK9 augmented M-CSF-induced cell proliferation in mouse bone marrow-derived macrophages (BMDMs) (Supplemental Figure 2A, B). Monocyte migration assay using the human monocytic cell line THP-1 showed that recombinant PCSK9 tended to promote MCP-1 (monocyte chemoattractant protein-1)-induced chemotaxis, suggesting lipid-independent effects of PCSK9 on monocyte migration (Supplemental Figure 2C). On the other hand, AAV-PCSK9 did not increase apoptosis determined by TUNEL staining in vein grafts of Ldlr-/- mice (Supplemental Figure 3A). Accordingly, AAV-PCSK9 did not change necrotic core area in the vein grafts (Supplemental Figure 3B).\nThe effects of Adeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) on collagen thinning and macrophage accumulation in vein graft lesions of Ldlr-/- mice. A, Picrosirius red staining of vein grafts without (top) or with (bottom) polarized light (n=12 and 11 for AAV-LacZ and AAV-PCSK9 group, respectively). Scale bars indicate 500 μm. Circle graphs show the percentage of thin (green) and thick (red) collagen fibers compared with total fibers. B, Intravital microscopy images of MMPSense 680 (red) and AminoSPARK750 (green) in vein grafts for visualization of MMP activity and macrophage accumulation, respectively (n=10 and 9 for AAV-LacZ and AAV-PCSK9 group, respectively). C, Mac3 (macrophages) staining in vein grafts. Scale bars indicate 500 μm. P value was calculated by Mann-Whitney U-test (thick collagen in A, macrophage accumulation mean fluorescence intensity in B) and unpaired Student t test (C). Data are reported as mean ± SEM.", "We previously reported that macrophage activation plays a key role in vein graft lesion development.8 To explore the effects of circulating PCSK9 on macrophage activation, we examined mRNA levels of pro-inflammatory (IL-1β [interleukin-1 beta], IL-6, TNFα, and MCP-1) and antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1 [transforming growth factor-beta 1], and IL-10) in the peritoneal F4/80+ macrophages of Ldlr-/- mice 1 or 4 weeks after intravenous injection of AAVs. AAV-PCSK9 promoted pro-inflammatory IL-1β, IL-6, TNFα, and MCP-1 mRNA levels at 1 week (Figure 3A). AAV-PCSK9 also suppressed Arginase-1 mRNA levels at 4 weeks when the increase in the mRNA levels of pro-inflammatory molecules at 1 week had subsided (Figure 3B). We have recently reported that predominant pathways of the vein grafts change over time during lesion development. In the same study, pathways with immune responses represent the early responsive proteins in proteomics analysis.9 These results indicate that PCSK9 can induce pro-inflammatory responses in macrophages initially and later impair resolution of inflammation in vivo.\nAdeno-associated virus (AAV)-PCSK9 (proprotein convertase subtilisin/kexin 9) induced macrophage activation in murine peritoneal macrophages via LDL receptor (LDLR)-independent mechanisms in vivo. mRNA levels of pro-inflammatory molecules (IL-1β, IL-6, TNFα, MCP-1) and antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1, IL-10) were measured in murine peritoneal macrophages of Ldlr-/- mice 1 week (A) or 4 weeks (B) after intravenous injection with AAV-LacZ or AAV-PCSK9 (n=7 to 8 per group). P value was calculated by Mann-Whitney U-test (TNFa in A, IL-1b, TNFa, MCP-1, Arginase-1, Ym1, and IL-10 in B) and unpaired Student t test. Data are reported as mean ± SEM.", "To support our in vivo evidence, we examined in vitro the effects of physiologically relevant levels of recombinant mouse PCSK9 (0.1–2.5 μg/mL)35 on the mRNA levels of the same pro-inflammatory and antiinflammatory molecules examined in Ldlr-/- mouse macrophages. First, we conducted a time-course study of recombinant PCSK9-induced TNFα and Arginase-1 mRNA expression levels to determine an optimal time point for harvesting Ldlr-/- mouse macrophages. The increase of TNFα mRNA levels peaked 3 hours after stimulation, whereas the decrease of Arginase-1 mRNA levels plateaued 12 hours after stimulation with recombinant PCSK9 (Figure 4A). For all experiments, Ldlr-/- mouse macrophages were therefore harvested 3 and 12 hours after stimulation to measure pro- and antiinflammatory molecules, respectively. Peritoneal macrophages treated with recombinant PCSK9 exhibited increased mRNA levels of IL-1β, IL-6, TNFα, and MCP-1 in a concentration-dependent manner (Figure 4B). In contrast, recombinant PCSK9 did not decrease the mRNA levels of antiinflammatory molecules (Figure 4C). These results support the in vivo evidence of macrophage activation by circulating PCSK9 in an LDLR-independent fashion.\nRecombinant PCSK9 (proprotein convertase subtilisin/kexin 9) induced macrophage activation in LDL receptor (LDLR)-independent mechanisms in vitro. A, Time course of TNFα and Arginase-1 mRNA levels in peritoneal macrophages of wild-type mice after stimulation with recombinant mouse PCSK9 (MmPCSK9; 2.5 μg/mL) (n=5 per group). P value was calculated by Kruskal-Wallis test, followed by Dunn multiple comparison test. B, mRNA levels of pro-inflammatory molecules (IL-1β, IL-6, TNFα, MCP-1) and (C) antiinflammatory molecules (Arginase-1, Ym1, MRC1, TGF-β1, IL-10) were measured in murine peritoneal macrophages from Ldlr-/- mice 3 and 12 hours after stimulation with recombinant mouse PCSK9 for pro-inflammatory and antiinflammatory molecules, respectively (n=9 per group). P value was calculated by 1-way ANOVA, followed by Dunnett multiple comparison test (IL-6, TNFa, MCP-1‚ MRC1) or Kruskal-Wallis test, followed by Dunn multiple comparison test. Data are reported as mean ± SEM.\nEndotoxin contamination in recombinant proteins can induce pro-inflammatory responses. Therefore, we chose a recombinant protein derived from a mammalian system, mouse myeloma cell line (NS0). The endotoxin level of recombinant mouse PCSK9 used in this study was 0.00441 EU/µg or less, well below the threshold of 0.1 EU/µg required for accurate cell-based assays.19 To exclude further endotoxin contamination in the recombinant PCSK9 protein affecting any measurements, we performed the chromogenic Limulus amebocyte lysate endotoxin assay. The final endotoxin level of the maximum dose of the recombinant protein (2.5 µg/mL) was 0.005 EU/mL or less, which coincides with the endotoxin levels of the commercially available endotoxin-free medium. In addition, mass spectrometry conducted on the solvent derived from reconstituted recombinant mouse PCSK9 protein solution did not produce signals, suggesting that there were no small molecule impurities present (Supplemental Figure 4A). Furthermore, stimulation with heat-inactivated recombinant mouse PCSK9 protein (2.5 μg/mL) caused no changes in the mRNA levels of IL-1β, TNF-α, and MCP-1, demonstrating that endotoxin contamination did not drive the effects observed (Supplemental Figure 4B).", "We explored further LDLR-independent pro-inflammatory signaling pathways in an unbiased manner, using RNA-sequencing of Ldlr-/- mouse macrophages stimulated with recombinant PCSK9. When analyzing the differentially expressed transcripts (FC>2.0, q<0.001), we observed 46 transcripts significantly increased by PCSK9 stimulation in Ldlr-/- macrophages compared with controls (Figure 5A). A hierarchical heat map of Ldlr-/- mouse macrophages filtered by differentially expressed transcripts showed a clear separation between control (no stimulus) and PCSK9 group (Figure 5B). We provided statistically significantly overrepresented transcripts ranked according to their fold change abundance (Supplemental Table 3A) to further examine these differentially expressed transcripts. PCSK9 treatment increased many transcripts implicated in pro-inflammatory responses in Ldlr-/- macrophages (eg, IL-1b, TNF, CXCL2, NF-κB [nuclear factor-kappa B]-related genes).19\nTranscriptomics, network analysis, and hyperedge entanglement prediction (HEP) of mouse macrophages stimulated with recombinant PCSK9 (proprotein convertase subtilisin/kexin 9). A, Volcano plot for Ldlr-/- mouse macrophages stimulated with recombinant PCSK9. Red markers indicate significantly expressed transcripts with a fold change cutoff of 2.0 and adjusted P value cutoff of 0.001. The enriched transcripts that were previously reported were denoted with their names. B, Heat map of the transcriptomics of Ldlr-/- mouse macrophages (FC>2, q<0.001-filtered data for the difference between macrophages stimulated with and without recombinant PCSK9, n=4 per group). C, The schematic network visualization by HEP prediction. The significantly predicted association of PCSK9 to the 24 mapped inflammation-related transcripts derived from the list of transcripts (FC>2.0, q<0.05) indicates every unknown pairs of interaction (dashed line) that have to be proven yet experimentally. One of them was validated by silencing Sdc4 (syndecan-4) mRNA (blue dashed line). All other links represent known physical interactions (solid gray lines). The node size illustrates the degree of the proteins in the protein-protein interaction network. D, Network analysis of the 24 overrepresented transcripts in Ldlr-/- mouse macrophages using STRING database. Nodes are colored according to k-means clustering (number of clusters=3). Only connected nodes are shown here.\nTo provide additional mechanistic links, we attempted to identify PCSK9-regulated proteins other than LDLR that mediate the aforementioned pro-inflammatory downstream responses. Many cascading effects in the transcriptional regulatory machinery could control the differentially expressed mRNAs. This set of differentially expressed transcripts were identified as the result of a genome-wide transcriptional profiling method, which might not necessarily interact directly with PCSK9. To estimate the likelihood of PCSK9 directly interacting with those differentially expressed transcripts, we tested this hypothesis verification through network computational analysis. In brief, PCSK9 can be seen as a node in a protein-protein interaction network,36 and the set of differentially expressed transcripts can be seen as a hyperedge projected on the protein-protein interaction network (a group of nodes that are associated by a common feature of biological functions). Muscoloni et al recently introduced a novel algorithm for HEP that exploits an ensemble of link predictors and that provides a level of statistical significance for the entanglement between each node and hyperedge that does not have a direct interaction in the network.20 We then performed the HEP analysis to quantify the significance of the entanglement between PCSK9 and the hyperedge of 24 differentially expressed transcripts (FC>2.0, q<0.05) in the dataset (Supplemental Figure 5A and 5B, Supplemental Table 3B). The resulting P values of 7/12 node-node link prediction methods (RA, CH2-L2, CH3-L2, and all 4 L3-based methods) established a significant association between PCSK9 and the hyperedge of the 24 differentially expressed transcripts (Figure 5C; Supplemental Table 2). These data suggest that any of the 24 differentially expressed transcripts have a high likelihood of directly interacting with PCSK9, enabling us to validate a direct interaction between PCSK9 and one of these 24 differentially expressed transcripts. To understand the biological relevance of these transcripts, we explored the network connectivity of these transcripts identified by HEP analysis using the STRING database (confidence interaction score ≥0.7).37 The network analysis showed a highly clustered network (average local clustering coefficient: 0.547) containing 24 nodes with 52 edges (expected number of edges=3), providing significantly more interactions than expected for a random set of genes of similar size (P value ≤1.0e−16). The k-means clustering (number of clusters=3) showed that one of these 3 clusters mainly consisted of the NF-κB signaling pathway based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (IL-1b, CXCL1 [chemokine (C-X-C motif) ligand 1], CXCL2 [chemokine (C-X-C motif) ligand 2], ICAM1, VCAM1 [vascular cell adhesion molecule 1], RELB [RELB Proto-Oncogene, NF-kB subunit], NFKB2 [nuclear factor kappa B subunit 2]), and the other 2 clusters included OLR1 (LOX-1) and SDC4, respectively (Figure 5D).\nIt is already reported that NF-κB signaling pathway is involved in PCSK9-induced atherosclerotic inflammation.38 In the present study, AAV-PCSK9 promoted NF-κB p65 mRNA level in the peritoneal F4/80+ macrophages of Ldlr-/- mice (Supplemental Figure 6A). Inhibition of NF-κB signaling pathway with IκB kinase inhibitor, 2-[(aminocarbonyl)amino]-5-(4-fluorophenyl)-3-thiophenecarboxamide (TPCA1), decreased pro-inflammatory responses to recombinant PCSK9 in Ldlr-/- mouse macrophages (Supplemental Figure 6B). These data indicate that NF-κB may mediate PCSK9-induced pro-inflammatory responses in vein graft lesions.\nPCSK9 stimulates the expression of LOX-1, which in turn takes up ox-LDL in macrophages.39 AAV-PCSK9 increased foam cell formation determined by Oil red O positive area in vein grafts (Supplemental Figure 7A). We further found that recombinant PCSK9 increased ox-LDL uptake in bone marrow-derived macrophages from Ldlr-/- mice (Supplemental Figure 7B). These results indicate that PCSK9 also induces foam cell formation by increased ox-LDL uptake, contributing not only to atherosclerotic but also to vein graft lesion development.\nAmong the members of this differentially expressed transcript hyperedge, we found that SDC4 included in the last cluster could be a novel target of PCSK9 (Figure 5D). While SDC4 was not a high-ranking molecule in the transcript ranking lists (30th in Supplemental Table 3A and 67th in Supplemental Table 3B), its fold change after stimulation with recombinant PCSK9 in Ldlr-/- mice macrophages was statistically significant (2.65, log2FC>1). AAV-PCSK9 also increased SDC4 mRNA level in the peritoneal F4/80+ macrophages of Ldlr-/- mice (Figure 6A). SDC4 is a heparan sulfate proteoglycan40 expressed on the surface of human macrophages.41 In addition, SDC4 mRNA is increased in M(LPS) but not M(IL-4/IL-13) or M(IL-10) in bone marrow-derived macrophages.42 A recent study reported that heparan sulfate proteoglycans physically bind to PCSK9 on the hepatocyte surface43 However, the investigators did not address the interaction between PCSK9 and any specific individual syndecans. Our in silico protein-protein docking analysis using HPEPDOCK, a web server for protein-protein docking based on a hierarchical algorithm, predicted binding between PCSK9 and SDC4 (Supplemental Figure 8A).44 We entered the pdb files of PCSK9 (PDB ID: 2PMW), LDLR (PDB ID: 1N7D), and SDC4 (PDB ID: 1EJP) into HPEPDOCK server, which in turn presented the top 100 docking models based on docking energy minimized scores. Protein-protein docking analysis showed that the binding efficiency of SDC4 and PCSK9 (−212.23 to −235.6 kJ/mol) was comparable to that of LDLR and PCSK9 (−259.85 to −303.21 kJ/mol) based on the docking score of the top 5 binding predictions (Supplemental Figure 8A). Besides, the ligand root mean square deviation, often used to evaluate the correctness of the docking geometry and optimal superimposition of the receptor-ligand binding, was lower in the binding of SDC4 and PCSK9 (39.12–85.16) compared with that of LDLR and PCSK9 (43.76–156.25) for the top 5 binding predictions, suggesting better binding between SDC4 and PCSK9 than in LDLR and PCSK9. Furthermore, co-immunoprecipitation analysis of mouse liver tissue lysates validated the occurrence of the predicted PCSK9-SDC4 binding in vivo (Supplemental Figure 8B).\nRNA silencing validated SDC4 (syndecan-4) as a potential target of PCSK9 (proprotein convertase subtilisin/kexin 9)-induced pro-inflammatory response in macrophages, identified via hyperedge entanglement prediction (HEP) prediction. A, SDC4 mRNA level was measured in murine peritoneal macrophages of Ldlr-/- mice 1 week after intravenous injection with AAV-LacZ or AAV-PCSK9 (n=7 per group). B, mRNA levels of SDC4, IL-1b, NLRP3, CD40, and ICAM1 were measured by RT-qPCR in mouse Ldlr-/- peritoneal macrophages after pretreatment with siSDC4 or siControl (Ctrl) for 48 hours and stimulation with 2.5 μg/mL recombinant mouse PCSK9 (MmPCSK9) for 3 hours (n=4–5). P value was calculated by 1-way ANOVA, followed by Bonferroni multiple comparison test. Error bars indicate ± SEM. Data are reported as mean ± SEM.", "We further validated our prediction that SDC4 is a potential target of PCSK9 using siRNA silencing of mouse Sdc4 in mouse peritoneal macrophages. Silencing Sdc4 with siSDC4 treatment achieved 93% reduction of SDC4 mRNA expression (Figure 6B). siSDC4 was then used to determine whether it could suppress PCSK9-induced pro-inflammatory molecules, including those predicted to be linked with PCSK9 by the HEP prediction (Figure 5C). siSDC4 suppressed mRNAs of the pro-inflammatory molecules IL-1β, NLRP3, CD40, and ICAM1, but not TNFα, MCP-1, TLR2, CXCL1, CXCL2, or CXCL10 after stimulation with recombinant PCSK9 (Figure 6B, Supplemental Figure 9). These data suggest that SDC4 partially regulates a PCSK9-induced pro-inflammatory response. Furthermore, these mRNA silencing results substantiate the validity of the prediction, yielding the integration of PCSK9 in our hyperedge of 24 disconnected differentially expressed transcripts.", "Our study demonstrated that circulating PCSK9 produced various changes in Ldlr-/- mice. We, however, attempted to examine the potential impact of local PCSK9 production in vein grafts. Immunohistochemical analysis using an anti-PCSK9 antibody in previous studies showed that vascular smooth muscle cells in human atherosclerotic plaque and collar-induced neointima of murine carotid artery express PCSK9.45,46 Based on these results, we examined whether locally produced PCSK9 in vein grafts may also contribute to vein graft lesion development. The local expression of PCSK9, as determined by in situ hybridization in the intima of vein grafts, was minimal compared with that in the liver (Figure 1B; Supplemental Figure 10A). RT-qPCR analysis further confirmed numerically significantly less PCSK9 mRNA expression in vein grafts than in the liver, the primary source of circulating PCSK9 (Supplemental Figure 10B). Moreover, we examined which cells within vein grafts can produce PCSK9 in humans by comparing PCSK9 mRNA levels in human saphenous vein endothelial cells, smooth muscle cells, and human primary macrophages derived from peripheral blood mononuclear cells. While we detected substantial PCSK9 expression in human saphenous vein smooth muscle cells, the level was significantly lower than that in HepG2 (liver) cells (Supplemental Figure 10C). Human primary macrophages and human saphenous vein endothelial cells contained minimal PCSK9 mRNA (Supplemental Figure 10C). Recent studies reported a modest increase of PCSK9 mRNA and protein levels in macrophages exposed to pro-inflammatory stimuli.39,47 We, therefore, examined PCSK9 mRNA expression in human primary macrophages treated with pro-inflammatory stimuli. Although LPS and IFNγ induced a pro-inflammatory response in the macrophages, as determined by TNFα mRNA expression (Supplemental Figures 10D and 10E, right graphs), PCSK9 mRNA expression did not increase after LPS or IFNγ (Supplemental Figures 10D and 10E, left graphs). These results indicate that smooth muscle cells (SMC), but not macrophages, may primarily produce PCSK9 in vein grafts, but to a much lower extent than the liver.", "Although our study focuses on macrophages, we examined the role of PCSK9 in the activation of endothelial cells and SMCs because other investigators reported potential interactions between PCSK9 and these cell types.46,48 Endothelial cell activation precedes vein graft lesion development.49 Due to the scarcity of mRNA in CD31-positive endothelial cells in vein grafts, we sorted them from the liver and lungs of Ldlr-/- mice injected with AAV-PCSK9 instead to examine the effects of PCSK9 on endothelial cell activation in an LDLR-independent fashion. AAV-PCSK9 did not increase mRNA levels of adhesion molecules, such as VCAM-1, ICAM-1, and E-selectin, in EC from either organ when compared with AAV-LacZ (Supplemental Figure 11A). We further found in vitro that recombinant PCSK9 did not increase mRNA levels of these adhesion molecules in human saphenous vein endothelial cells (Supplemental Figure 11B). These results indirectly indicate that PCSK9 may not induce endothelial activation in vein grafts.\nSMC modulation, migration, and proliferation contribute to the development of vascular disorders, including atherosclerosis50 and vein graft.51 A recent study showed that PCSK9 may sustain SMC dedifferentiation, migration, and proliferation in neointimal hyperplasia in response to vascular injury,46 suggesting the involvement of SMCs in PCSK9-induced vein graft lesion development. To explore this possibility, we examined the effects of recombinant PCSK9 on SMC dedifferentiation, migration, and proliferation in human saphenous vein smooth muscle cells. Recombinant PCSK9 slightly decreased SM22α mRNA levels (0.74 and 0.68-fold for 1.0 and 5.0 µg/mL, respectively), but did not affect differentiation molecules, such as αSMA, calponin, SM1, or SM2 (Supplemental Figure 12A). In addition, recombinant PCSK9 did not affect PDGF-induced migration activity of human saphenous vein smooth muscle cells (Supplemental Figure 12B). A BrdU assay using human saphenous vein smooth muscle cells showed that recombinant PCSK9 did not enhance 10% fetal bovine serum-induced proliferation (Supplemental Figure 12C). AAV-PCSK9 did not affect the SMC content in vein grafts (Supplemental Figure 12D). Collectively, in our experimental settings, our data did not provide the mechanistic evidence in vitro and in vivo that activation of endothelial cells or SMCs contribute to PCSK9-induced vein graft lesion formation. These results indicate that circulating PCSK9 promotes vein graft lesion development predominantly through macrophage activation.", "This study investigated LDLR-independent mechanisms by which PCSK9 induces pro-inflammatory activation of macrophages and accelerates vein graft lesion development. The key findings include: (1) AAV-PCSK9 did not change lipid profile but promoted vein graft lesion development in Ldlr-/- mice, suggesting LDLR-independent mechanisms; (2) AAV-PCSK9 increased macrophage accumulation and decreased fibrillar collagen in vein graft lesions; (3) PCSK9 induced macrophage activation in Ldlr-/- mice; (4) unbiased global transcriptomics of Ldlr-/- mouse macrophages stimulated with recombinant PCSK9 revealed activation of several pro-inflammatory pathways; (5) a combination of the global transcriptomics and novel network-based hyperedge entanglement prediction analysis showed NF-κB signaling pathway and OLR1(LOX-1) could play a key role in vein graft lesion development; (6) SDC4, a heparan sulfate proteoglycan, could be a novel target of PCSK9 that mediates pro-inflammatory responses in macrophages.\nReversed autologous saphenous veins are widely used for surgical bypass for treatment of lower extremity peripheral artery disease and coronary artery disease. Peripheral artery disease affects approximately 8.5 million people in the United States aged ≥40 years52 and a total of 202 million worldwide, as of 2010.53 The incidence of peripheral artery disease has been rising by 23.5% globally from 2000 to 2010 largely due to aging populations.53 Of note, 40% of vein grafts for peripheral artery disease are failing or fail within a year.1 The volume of CABG procedures for severe coronary artery disease has declined since 1998; however, 371 000 bypass procedures were still performed in the United States in 2014,52 and approximately 30% of these costly procedures are failing or fail 12 to 18 months after surgery.2\nEvidence suggests that pro-inflammatory responses to PCSK9 in macrophages and arterial atherosclerotic lesions may primarily depend on LDLR. Recombinant PCSK9 induced TNFα mRNA in bone marrow-derived macrophages mainly in an LDLR-dependent fashion.19 Furthermore, overexpression of human PCSK9 in macrophages promoted atherosclerotic lesions.28 Deletion of the PCSK9 gene in the liver reduced atherosclerotic lesions, primarily via mechanisms dependent on LDLR.29 To the best of our knowledge, no in vivo studies have demonstrated LDLR-independent pro-inflammatory effects of PCSK9 on vascular lesion development. The present study demonstrated that circulating PCSK9 induces macrophage activation and vein graft lesion development in LDLR-independent mechanisms. The relative contributions of the LDLR-dependent and independent mechanisms, however, remain unknown.\nClinical reports demonstrated that cholesterol-lowering with PCSK9 inhibitors reverses the pro-inflammatory profile of monocytes in patients with hypercholesterolemia,54 indicating the lipid-dependent pro-inflammatory effects of PCSK9 in humans. Clinical trials also demonstrated that PCSK9 inhibitors lower not only serum levels of LDL cholesterol but also lipoprotein (a) (Lp(a)).55 Mice lack apolipoprotein (a) or Lp(a),56 which enabled us to rule out the role of Lp(a) in the effects of PCSK9 in our in vivo study. Although genetic evidence suggests LDL-dependent effects of PCSK9 in gain-of-function and loss-of-function carriers57 and the clinical benefits of antiPCSK9 strategies derive largely from LDL lowering, nonLDL-dependent effects might also contribute.58 As serum total cholesterol and triglyceride levels did not differ in AAV-LacZ and AAV-PCSK9 groups, our findings in Ldlr-/- mice do not seem to depend on PCSK9’s effects on the profile of circulating lipids.\nHigh sensitivity C-reactive protein is the most established marker of inflammation to predict cardiovascular events independently in primary and secondary prevention.59 Clinical trials with PCSK9 inhibitors did not show any significant changes in high sensitivity C-reactive protein levels in patients with coronary artery disease.60,61 Antiinflammatory changes in monocyte phenotype with PCSK9 antibodies were not accompanied by a CRP (C-reactive protein) reduction in patients with familial hypercholesterolemia, suggesting that CRP may not solely capture changes in local inflammation produced by PCSK9 inhibition.54 In addition, the ATHEROREMO-IVUS study (The European Collaborative Project on Inflammation and Vascular Wall Remodeling in Atherosclerosis - Intravascular Ultrasound) demonstrated a linear relationship between serum levels of PCSK9 and the fraction or amount of the necrotic core in the nonculprit lesions of patients with acute coronary syndrome, independently of serum levels of LDL cholesterol. Our study provides support for the notion that circulating PCSK9 may exert pro-inflammatory effects on vascular lesions. Further studies are needed to explore whether PCSK9 inhibition ameliorates local inflammation in vascular lesions in patients.\nIn general, multi-omics technologies, such as proteomics and transcriptomics, identify many differentially expressed molecules. It is challenging to assess direct interactions between these molecules or their causal roles in a biological or disease context by examining each molecule or interaction among many. This challenge has driven efforts in computational network biomedicine to develop unbiased prediction methods. By exploiting the mere topology of a protein-protein interaction network, one can quantify the likelihood of direct interactions between a molecule of interest (eg, PCSK9 in the present study) and a hyperedge of differentially expressed molecules (which represents a network functional module). We used a novel node2hyperedge entanglement method20 and showed any of the 24 differentially expressed transcripts, including NF-κB signaling molecules, OLR1(LOX-1), and SDC4, as a potential target of PCSK9 associated with inflammation. In the present study, we identified SDC4 as a potential novel target of PCSK9. Although Sdc4 was not a high-ranking differential expressed transcript, its fold change after stimulation with recombinant PCSK9 in the RNA-sequencing was statistically significant. We further performed in vitro experiments involving siRNA silencing in primary macrophages to substantiate our computational prediction platform and demonstrated that SDC4 indeed binds to PCSK9 in vivo and mediates PCSK9-induced pro-inflammatory responses. The contribution of macrophage SDC4 to PCSK9-induced pro-inflammatory responses in vivo remains to be elucidated. This novel prediction method may offer a powerful and necessary tool for the completion of the human interactome and for generating new hypotheses.\nSDC4 regulates several robust pro-inflammatory molecules, such as IL-1β, NLRP3 (NLR family pyrin domain containing 3), CD40 (cluster of differentiation 40) and ICAM1. Various endogenous signals abundantly present in atherosclerotic lesions activates NLRP3, a key mediator of IL-1β maturation.62,63 T cell-mediated macrophage activation involves CD40 to produce interstitial collagens and tissue-factor pro-coagulant.64 The role of macrophage ICAM1 in atherosclerosis remains obscure, but a recent study reported that macrophage ICAM1 suppresses “M2-like” macrophages during tumor progression. SDC4 silencing inhibited these pro-inflammatory molecules. The magnitude of inhibition, however, was modest, suggesting that while we propose SDC4 is a novel target of PCSK9, but PCSK9 may also target other molecules (eg, NF-κB signaling molecules). The mechanisms mediated via SDC4 we report here appears to be one of multiple pathways affected by PCSK9. Heparan sulfate proteoglycans modulate macrophage activation by maintaining cells in a quiescent state through the capture and sequestration of IFNβ.65,66 We speculate that SDC4 serves as a mediator of PCSK9-induced pro-inflammatory responses through the capture of PCSK9 on the surface of macrophages as seen in the hepatocyte.43 Our data demonstrated that NF-κB may mediate pro-inflammatory responses downstream of the PCSK9 and SDC4 binding. Understanding more detailed mechanisms by which SDC4 mediates the pro-inflammatory responses and their downstream signaling induced by PCSK9 requires future investigations.\nOur study does not demonstrate definitively that local PCSK9 plays an important role in vein graft lesion development. In situ hybridization and qPCR data suggest that SMCs may be the primary source of PCSK9 in these lesions. The expression level of PCSK9 in SMCs was much lower than in the liver, although the relative gene expression level may not be a valid determinant of its biological role or importance. Whether and how circulating PCSK9 affects lesional macrophages through the contact with endothelial cells (which in turn affects macrophage phenotype or the entry into the subendothelial space, which results in direct contact with macrophages) remains to be elucidated.67 Future work needs to define detailed molecular mechanisms.\nOur data do not demonstrate that human primary macrophages can express PCSK9, even when stimulated with pro-inflammatory stimuli, which was consistent with previous studies.45,68 Evidence suggests, however, the participation of endogenous PCSK9 in macrophages during pro-inflammatory responses. In vitro experiments showed that overexpression of PCSK9 in macrophage-like THP-1 cells enhanced the pro-inflammatory response induced by oxidized LDL via NF-κB activation.38 In contrast, PCSK9 gene silencing inhibited the response.38,47 In vivo evidence also showed that overexpression of human PCSK9 in macrophages increased atherosclerotic lesion formation in Apoe-/- mice.28 These results suggest that not only circulating PCSK9 produced from the liver but also local PCSK9 produced from SMCs and macrophages may induce a pro-inflammatory response in macrophages and contribute to vascular lesion development. What cell types other than SMCs contribute to monocyte/macrophage infiltration, and activation remains to be elucidated.\nThe present study demonstrates several lines of novel evidence that circulating PCSK9 promotes macrophage activation and vein graft lesion development via mechanisms independent of blood cholesterol and LDLR degradation. Despite current therapeutics and research advancements, effective medical solutions are limited to prevent vein graft failure. Our study provides molecular bases that circulating PCSK9 deserves investigations as a potential therapeutic target for vein graft failure.", "[SUBTITLE] Acknowledgments [SUBSECTION] We thank Ryo Kawakami, Yuki Tsukano, Dayanna C. Romero, Whitney S. Golden, Keishi Nihira, Andrew K. Mlynarchik, Edward Guzman, and Daniel G. Anderson for their technical assistance. The graphical figure was created with Biorender.com. Additional Information: Coauthor Amitabh Sharma died on November 24, 2019.\nWe thank Ryo Kawakami, Yuki Tsukano, Dayanna C. Romero, Whitney S. Golden, Keishi Nihira, Andrew K. Mlynarchik, Edward Guzman, and Daniel G. Anderson for their technical assistance. The graphical figure was created with Biorender.com. Additional Information: Coauthor Amitabh Sharma died on November 24, 2019.\n[SUBTITLE] Sources of Funding [SUBSECTION] This work was supported by the National Institute of Health grants R01HL126901 and R01HL149302, and Pfizer ASPIRE Award to MA; R01HL136431, R01HL147095, and R01HL141917 to EA; and the MSD Scholarships for Overseas Study, and Japan Society for the Promotion of Science Fellowships for Overseas Researchers to SK.\nThis work was supported by the National Institute of Health grants R01HL126901 and R01HL149302, and Pfizer ASPIRE Award to MA; R01HL136431, R01HL147095, and R01HL141917 to EA; and the MSD Scholarships for Overseas Study, and Japan Society for the Promotion of Science Fellowships for Overseas Researchers to SK.\n[SUBTITLE] Disclosures [SUBSECTION] Pfizer was not involved in the study other than funding. MA also received research grants from Kowa Company and Sanofi.\nPfizer was not involved in the study other than funding. MA also received research grants from Kowa Company and Sanofi.\n[SUBTITLE] Supplemental Materials [SUBSECTION] Online Supplemental Materials and Methods\nSupplemental Figures 1–14\nSupplemental Tables 1–4\nReferences 69–75\nOnline Supplemental Materials and Methods\nSupplemental Figures 1–14\nSupplemental Tables 1–4\nReferences 69–75", "We thank Ryo Kawakami, Yuki Tsukano, Dayanna C. Romero, Whitney S. Golden, Keishi Nihira, Andrew K. Mlynarchik, Edward Guzman, and Daniel G. Anderson for their technical assistance. The graphical figure was created with Biorender.com. Additional Information: Coauthor Amitabh Sharma died on November 24, 2019.", "This work was supported by the National Institute of Health grants R01HL126901 and R01HL149302, and Pfizer ASPIRE Award to MA; R01HL136431, R01HL147095, and R01HL141917 to EA; and the MSD Scholarships for Overseas Study, and Japan Society for the Promotion of Science Fellowships for Overseas Researchers to SK.", "Pfizer was not involved in the study other than funding. MA also received research grants from Kowa Company and Sanofi.", "Online Supplemental Materials and Methods\nSupplemental Figures 1–14\nSupplemental Tables 1–4\nReferences 69–75", "" ]
[ null, null, "methods", "data-availability", "methods", null, null, null, null, "results", null, null, null, null, null, null, null, null, "discussion", null, null, null, "COI-Statement", null, "supplementary-material" ]
[ "graft occlusion, vascular", "inflammation", "macrophage activation", "receptors, LDL", "systems biology" ]
Optimizing the implementation of lung cancer screening in Scotland: Focus group participant perspectives in the LUNGSCOT study.
36263948
Targeted lung cancer screening is effective in reducing lung cancer and all-cause mortality according to major trials in the United Kingdom and Europe. However, the best ways of implementing screening in local communities requires an understanding of the population the programme will serve. We undertook a study to explore the views of those potentially eligible for, and to identify potential barriers and facilitators to taking part in, lung screening, to inform the development of a feasibility study.
INTRODUCTION
Men and women aged 45-70, living in urban and rural Scotland, and either self-reported people who smoke or who recently quit, were invited to take part in the study via research agency Taylor McKenzie. Eleven men and 14 women took part in three virtual focus groups exploring their views on lung screening. Focus group transcripts were transcribed and analysed using thematic analysis, assisted by QSR NVivo.
METHODS
Three overarching themes were identified: (1) Knowledge, awareness and acceptability of lung screening, (2) Barriers and facilitators to screening and (3) Promoting screening and implementation ideas. Participants were largely supportive of lung screening in principle and described the importance of the early detection of cancer. Emotional and psychological concerns as well as system-level and practical issues were discussed as posing barriers and facilitators to lung screening.
FINDINGS
Understanding the views of people potentially eligible for a lung health check can usefully inform the development of a further study to test the feasibility and acceptability of lung screening in Scotland.
CONCLUSIONS
[ "Male", "Humans", "Female", "Early Detection of Cancer", "Focus Groups", "Lung Neoplasms", "Mass Screening", "Scotland", "Qualitative Research" ]
9700133
null
null
METHODS
We conducted three focus groups with self‐reported ‘heavy smokers’ living in rural, urban and deprived areas of Scotland to ascertain their views on barriers to lung screening. Focus groups were considered the most appropriate method for engaging with those eligible for screening, as they enabled an understanding of how participants perceive the prospect of lung screening through discussion and they enable the sharing and development of ideas. 19 Findings from the focus groups will feed into the development and design of the lung screening pilot. We employed a range of strategies and reflexive practices to ensure that focus groups were participant‐led and data‐driven. 20 [SUBTITLE] Identifying participants [SUBSECTION] Participants were recruited through Taylor McKenzie (TM), a Scottish‐based company that specializes in qualitative research, to identify members of the public eligible to take part in the focus groups (https://www.taylormckenzie.co.uk). TM developed a study‐specific screening questionnaire (further details below) to allow the purposive sampling of eligible participants from their extensive database of people willing to take part in health, social or marketing research. Participants were recruited through Taylor McKenzie (TM), a Scottish‐based company that specializes in qualitative research, to identify members of the public eligible to take part in the focus groups (https://www.taylormckenzie.co.uk). TM developed a study‐specific screening questionnaire (further details below) to allow the purposive sampling of eligible participants from their extensive database of people willing to take part in health, social or marketing research. [SUBTITLE] Recruitment and sampling [SUBSECTION] We aimed to recruit up to 24 people from across Scotland to take part in three separate focus groups of 8 people, considered to be an appropriate number to identify a range of views. 21 Interested people responded to recruitment notices posted on TM's mailing list and social media pages. We used the inclusion and exclusion criteria listed In Box 1. Inclusion criteria Men and womenAge 45–70 (inclusive)Current residence in ScotlandSelf‐identify as smoker or recent quitter (within the last 2 years)Able to undertake focus group interview in EnglishWilling to discuss their views on lung screening Men and women Age 45–70 (inclusive) Current residence in Scotland Self‐identify as smoker or recent quitter (within the last 2 years) Able to undertake focus group interview in English Willing to discuss their views on lung screening Exclusion criteria Lacking capacity to give informed consentNever smokerSmoker who quit more than 2 years agoNon‐English speakers, preventing them from comfortably taking part in a discussion Lacking capacity to give informed consent Never smoker Smoker who quit more than 2 years ago Non‐English speakers, preventing them from comfortably taking part in a discussion Those who responded were provided with a study information sheet by TM and given 7–14 days to consider taking part. TM drew up a list of eligible participants (based on information on their database, e.g., socioeconomic grade [SEG], occupation, and from speaking to potential participants directly, e.g., smoking status) and added it to a secure portal for the researchers to access and review. Eligible participants were allocated to a focus group at a preset date and time. Participants were offered a financial reimbursement for their time, paid via TM. We aimed to recruit up to 24 people from across Scotland to take part in three separate focus groups of 8 people, considered to be an appropriate number to identify a range of views. 21 Interested people responded to recruitment notices posted on TM's mailing list and social media pages. We used the inclusion and exclusion criteria listed In Box 1. Inclusion criteria Men and womenAge 45–70 (inclusive)Current residence in ScotlandSelf‐identify as smoker or recent quitter (within the last 2 years)Able to undertake focus group interview in EnglishWilling to discuss their views on lung screening Men and women Age 45–70 (inclusive) Current residence in Scotland Self‐identify as smoker or recent quitter (within the last 2 years) Able to undertake focus group interview in English Willing to discuss their views on lung screening Exclusion criteria Lacking capacity to give informed consentNever smokerSmoker who quit more than 2 years agoNon‐English speakers, preventing them from comfortably taking part in a discussion Lacking capacity to give informed consent Never smoker Smoker who quit more than 2 years ago Non‐English speakers, preventing them from comfortably taking part in a discussion Those who responded were provided with a study information sheet by TM and given 7–14 days to consider taking part. TM drew up a list of eligible participants (based on information on their database, e.g., socioeconomic grade [SEG], occupation, and from speaking to potential participants directly, e.g., smoking status) and added it to a secure portal for the researchers to access and review. Eligible participants were allocated to a focus group at a preset date and time. Participants were offered a financial reimbursement for their time, paid via TM. [SUBTITLE] Focus groups and consent [SUBSECTION] Three focus groups, lasting approximately 75 min, were run virtually using the online video conferencing platform Zoom, selected due to likely participant familiarity with it, and to comply with the prevailing government restrictions on face‐to‐face meetings at the time due to the Covid‐19 pandemic. Participants were asked to sign a digital consent form via TM ahead of the focus group and verbal consent was agreed upon at the beginning of each focus group. Focus groups were led and facilitated by two researchers (D. C. and M. N.—health services researchers), with one group comprising those living in rural areas and the other two urban groups. Participants did not know one another before the focus group. The format and content of the focus groups were developed by a subgroup of the research team with expertise in behavioural aspects of cancer screening, drawing on relevant literature. The lung screening process was explained to participants: eligible people would be offered a LDCT scan to detect any lung conditions, one of which is lung cancer. The topic guide (see Supporting Information: 1) covered views on lung screening, with a particular focus on perceived barriers to taking part, personal resources to facilitate screening, understanding of the process, and input on what a good screening programme would look like. With consent, focus groups were recorded. Three focus groups, lasting approximately 75 min, were run virtually using the online video conferencing platform Zoom, selected due to likely participant familiarity with it, and to comply with the prevailing government restrictions on face‐to‐face meetings at the time due to the Covid‐19 pandemic. Participants were asked to sign a digital consent form via TM ahead of the focus group and verbal consent was agreed upon at the beginning of each focus group. Focus groups were led and facilitated by two researchers (D. C. and M. N.—health services researchers), with one group comprising those living in rural areas and the other two urban groups. Participants did not know one another before the focus group. The format and content of the focus groups were developed by a subgroup of the research team with expertise in behavioural aspects of cancer screening, drawing on relevant literature. The lung screening process was explained to participants: eligible people would be offered a LDCT scan to detect any lung conditions, one of which is lung cancer. The topic guide (see Supporting Information: 1) covered views on lung screening, with a particular focus on perceived barriers to taking part, personal resources to facilitate screening, understanding of the process, and input on what a good screening programme would look like. With consent, focus groups were recorded. [SUBTITLE] Analysis [SUBSECTION] Focus group recordings were transcribed verbatim and subject to Braun and Clarke's 22 thematic analysis, chosen as it is the most commonly used approach considered appropriate to derive key themes and ideas from the group discussions, taking context into account 19 and consistent with social constructionist underpinnings. Using thematic analysis allowed us to incorporate guidance specific to focus group interviewing and its impact on analysis, for example, considering dynamics, social comparison and power imbalances within groups. 23 , 24 Transcripts were read repeatedly and compared and contrasted to develop a set of common codes by D. C. These codes were applied back across the data and assigned to excerpts from the focus groups using QSR NVivo version 12 Pro (www.qsrinternational.com) by D. C. and J. R. (a research intern). Codes were further refined and a set of overarching themes and subthemes were inductively derived to interpret and explain the data, in discussion with the wider research team and patient advisory group. The LUNGSCOT team comprises health services researchers, health psychologists, clinicians, a health economist and patient and carer representatives. Themes were placed in the context of existing literature and theory to incorporate our findings into the wider evidence base. Focus group recordings were transcribed verbatim and subject to Braun and Clarke's 22 thematic analysis, chosen as it is the most commonly used approach considered appropriate to derive key themes and ideas from the group discussions, taking context into account 19 and consistent with social constructionist underpinnings. Using thematic analysis allowed us to incorporate guidance specific to focus group interviewing and its impact on analysis, for example, considering dynamics, social comparison and power imbalances within groups. 23 , 24 Transcripts were read repeatedly and compared and contrasted to develop a set of common codes by D. C. These codes were applied back across the data and assigned to excerpts from the focus groups using QSR NVivo version 12 Pro (www.qsrinternational.com) by D. C. and J. R. (a research intern). Codes were further refined and a set of overarching themes and subthemes were inductively derived to interpret and explain the data, in discussion with the wider research team and patient advisory group. The LUNGSCOT team comprises health services researchers, health psychologists, clinicians, a health economist and patient and carer representatives. Themes were placed in the context of existing literature and theory to incorporate our findings into the wider evidence base. [SUBTITLE] Patient advisory group [SUBSECTION] The patient advisory group was convened for the purpose of the wider LUNGSCOT study. The group comprises three patients and one carer with experience in lung cancer and two patients with other cancers. The group has been involved in the study design and commented on study documentation as well as two advisors reading transcripts and sharing their views on the analysis. The group meets quarterly to discuss study progress and opportunities to get involved in study tasks. The patient advisory group was convened for the purpose of the wider LUNGSCOT study. The group comprises three patients and one carer with experience in lung cancer and two patients with other cancers. The group has been involved in the study design and commented on study documentation as well as two advisors reading transcripts and sharing their views on the analysis. The group meets quarterly to discuss study progress and opportunities to get involved in study tasks.
null
null
CONCLUSIONS
This focus group study has already identified several perceived individual, practical and system barriers and facilitators to participation in a pilot lung screening programme in Scotland using LDCT. While our results resonate with existing literature in this field, they will be helpful in addressing factors which are especially important in Scotland if it is to embrace lung cancer screening—reducing health inequalities, engaging deprived populations and ensuring access in remote and rural areas. The findings will inform the design and implementation of a Scottish pilot lung screening study.
[ "INTRODUCTION", "Identifying participants", "Recruitment and sampling", "Focus groups and consent", "Analysis", "Patient advisory group", "FINDINGS", "Knowledge, awareness and acceptability of lung screening", "Barriers and facilitators to screening", "Individual level influences on screening intentions", "Psychological and emotional concerns", "Perceived risk and fatalism", "Stigma and judgement as a barrier", "Mistrust of healthcare professionals and services", "Practical and system barriers", "Promoting screening and implementation ideas", "Managing fears and expectations", "Improving accessibility", "Summary", "Comparison with existing literature and theory", "Strengths and limitations", "Implications and future work", "AUTHOR CONTRIBUTIONS", "ETHICS STATEMENT" ]
[ "Lung cancer remains one of the major causes of cancer mortality globally, and Scotland has high incidence and low survival rates compared with the rest of Europe.\n1\n, \n2\n, \n3\n, \n4\n Despite many initiatives to raise awareness and encourage early symptomatic presentation, most lung cancers are diagnosed at a late stage, and overall survival is poor.\n5\n A recent Public Health Scotland report revealed that the Covid‐19 pandemic has had a further negative impact on the clinical presentation of lung cancer, with a reduction in diagnoses during the lockdown periods, and an anticipated wave of late diagnoses from this backlog.\n6\n\n\nOver the last 5 years, there has been a growing body of evidence from trials in the United States and continental Europe, and pilot studies in the United Kingdom, demonstrating survival gains from low‐dose computed tomography (LDCT) screening for lung cancer in high‐risk populations.\n7\n, \n8\n, \n9\n, \n10\n A recent meta‐analysis of this evidence provides a strong case for the implementation of targeted lung screening, supported by expert clinical opinion,\n11\n and the UK National Screening Committee has recently recommended in favour of targeted lung screening. However, there are still many lung screening implementation challenges to be addressed.\n12\n, \n13\n Importantly, lung screening is most effective if targeted at high‐risk groups—ideally using validated risk prediction tools to assess who is at the highest risk of developing lung cancer and would most benefit from screening, namely smokers or recent quitters, aged 55–74 years.\nTo date, UK pilots and trials, including the Early Detection of Cancer of the Lung Scotland (ECLS) trial in Scotland using blood biomarkers to detect early signs of lung cancer, have shown variable and socially patterned uptake of screening,\n8\n, \n14\n, \n15\n, \n16\n and work has been done to understand barriers to participation. Uptake remains lower in more marginalized groups—that is, heavy smokers living in more deprived areas.\n17\n Further research to understand the views of Scotland's population on the acceptability of lung screening, and barriers and facilitators (related to issues of geography, rural and urban deprivation and a high burden of multimorbidity), will help shape future pilots and programmes in Scotland.\nThis focus group study explores the views of people potentially eligible for lung screening, identifies perceived barriers and facilitators to participation, and examines strategies to optimize the implementation of lung screening in Scotland. It forms part of the LUNGSCOT study, which is examining the feasibility of introducing lung screening in Scotland.\n18\n\n", "Participants were recruited through Taylor McKenzie (TM), a Scottish‐based company that specializes in qualitative research, to identify members of the public eligible to take part in the focus groups (https://www.taylormckenzie.co.uk). TM developed a study‐specific screening questionnaire (further details below) to allow the purposive sampling of eligible participants from their extensive database of people willing to take part in health, social or marketing research.", "We aimed to recruit up to 24 people from across Scotland to take part in three separate focus groups of 8 people, considered to be an appropriate number to identify a range of views.\n21\n Interested people responded to recruitment notices posted on TM's mailing list and social media pages. We used the inclusion and exclusion criteria listed In Box 1.\nInclusion criteria\n\nMen and womenAge 45–70 (inclusive)Current residence in ScotlandSelf‐identify as smoker or recent quitter (within the last 2 years)Able to undertake focus group interview in EnglishWilling to discuss their views on lung screening\n\nMen and women\nAge 45–70 (inclusive)\nCurrent residence in Scotland\nSelf‐identify as smoker or recent quitter (within the last 2 years)\nAble to undertake focus group interview in English\nWilling to discuss their views on lung screening\nExclusion criteria\n\nLacking capacity to give informed consentNever smokerSmoker who quit more than 2 years agoNon‐English speakers, preventing them from comfortably taking part in a discussion\n\nLacking capacity to give informed consent\nNever smoker\nSmoker who quit more than 2 years ago\nNon‐English speakers, preventing them from comfortably taking part in a discussion\nThose who responded were provided with a study information sheet by TM and given 7–14 days to consider taking part. TM drew up a list of eligible participants (based on information on their database, e.g., socioeconomic grade [SEG], occupation, and from speaking to potential participants directly, e.g., smoking status) and added it to a secure portal for the researchers to access and review. Eligible participants were allocated to a focus group at a preset date and time. Participants were offered a financial reimbursement for their time, paid via TM.", "Three focus groups, lasting approximately 75 min, were run virtually using the online video conferencing platform Zoom, selected due to likely participant familiarity with it, and to comply with the prevailing government restrictions on face‐to‐face meetings at the time due to the Covid‐19 pandemic. Participants were asked to sign a digital consent form via TM ahead of the focus group and verbal consent was agreed upon at the beginning of each focus group. Focus groups were led and facilitated by two researchers (D. C. and M. N.—health services researchers), with one group comprising those living in rural areas and the other two urban groups. Participants did not know one another before the focus group. The format and content of the focus groups were developed by a subgroup of the research team with expertise in behavioural aspects of cancer screening, drawing on relevant literature. The lung screening process was explained to participants: eligible people would be offered a LDCT scan to detect any lung conditions, one of which is lung cancer. The topic guide (see Supporting Information: 1) covered views on lung screening, with a particular focus on perceived barriers to taking part, personal resources to facilitate screening, understanding of the process, and input on what a good screening programme would look like. With consent, focus groups were recorded.", "Focus group recordings were transcribed verbatim and subject to Braun and Clarke's\n22\n thematic analysis, chosen as it is the most commonly used approach considered appropriate to derive key themes and ideas from the group discussions, taking context into account\n19\n and consistent with social constructionist underpinnings. Using thematic analysis allowed us to incorporate guidance specific to focus group interviewing and its impact on analysis, for example, considering dynamics, social comparison and power imbalances within groups.\n23\n, \n24\n Transcripts were read repeatedly and compared and contrasted to develop a set of common codes by D. C. These codes were applied back across the data and assigned to excerpts from the focus groups using QSR NVivo version 12 Pro (www.qsrinternational.com) by D. C. and J. R. (a research intern). Codes were further refined and a set of overarching themes and subthemes were inductively derived to interpret and explain the data, in discussion with the wider research team and patient advisory group. The LUNGSCOT team comprises health services researchers, health psychologists, clinicians, a health economist and patient and carer representatives. Themes were placed in the context of existing literature and theory to incorporate our findings into the wider evidence base.", "The patient advisory group was convened for the purpose of the wider LUNGSCOT study. The group comprises three patients and one carer with experience in lung cancer and two patients with other cancers. The group has been involved in the study design and commented on study documentation as well as two advisors reading transcripts and sharing their views on the analysis. The group meets quarterly to discuss study progress and opportunities to get involved in study tasks.", "Eleven females and 14 males aged 45–70 years living in a mix of urban and rural areas in Scotland took part across the three focus groups. Eleven participants were current smokers, and 14 had quit within the previous 2 years. All participants were from the lower socio‐economic grades (SEG): C2 (skilled manual workers), D (semiskilled and unskilled manual workers) and E (nonworking). Twenty‐one participants were White British/Scottish, one person was Black British, one British Asian and one South Asian. All participants had school‐level or vocational qualifications but no one had a higher education degree. See Tables 1, 2, 3 for participant characteristics.\nFocus Group 1 participant characteristics\n\nNote: Four males, four females, age 46–71 years, from six different health board regions, living in urban areas. Four current smokers and four of whom had quit within the last 2 years.\nFocus Group 2 participant characteristics\n\nNote: Four females, three males, age range 45–62 years, from three different health board regions, living in rural areas, three smokers and four of whom have quit within the last 2 years.\nFocus Group 3 participant characteristics\n\nNote: Three females, seven males, age range 45–68 years, from three different health board regions, living in urban areas, four smokers and six of whom have quit within the last 2 years.\nOur analysis identified three overarching themes in the data: (1) Knowledge, awareness and acceptability of lung screening, (2) Barriers and facilitators to screening and (3) Promoting screening and implementation ideas.\n[SUBTITLE] Knowledge, awareness and acceptability of lung screening [SUBSECTION] There is currently no national lung screening programme in the United Kingdom, although parts of NHS England are offering lung screening with LDCT.\n25\n Participants were largely unaware of the concept of targeted lung screening:No, I always thought that was something that happened if you develop, you know, or if they suspect you develop then you would have a check, otherwise nothing pre‐emptive…. (FG1, R5)\n\nNo, I always thought that was something that happened if you develop, you know, or if they suspect you develop then you would have a check, otherwise nothing pre‐emptive…. (FG1, R5)\nMost participants had heard of and participated in other forms of screening, including for breast, cervical and bowel cancer, and a few had been referred for lung checks for other reasons. They also described family members having had cancers picked up in this way, as well as their own experience of cancer:For me, it really is, it's very important to be screened, especially breast, bowel, anything. I suffered myself from throat cancer ten years ago and I've been in remission for the past four years, so it's urgently important that people get this done, yeah. (FG3, R4)\n\nFor me, it really is, it's very important to be screened, especially breast, bowel, anything. I suffered myself from throat cancer ten years ago and I've been in remission for the past four years, so it's urgently important that people get this done, yeah. (FG3, R4)\nWhen we described what lung screening would entail, participants were very supportive of this form of screening being available and welcomed the chance to have their lungs screened, with multiple participants saying it was ‘a great idea’. Participants were aware of the benefits of early (asymptomatic) detection and treatment:I think the screening is a good idea to catch things earlier or to see if somebody's got the disease or whatever that they didn't know they had. (FG1, R7)\n\nI think the screening is a good idea to catch things earlier or to see if somebody's got the disease or whatever that they didn't know they had. (FG1, R7)\nParticipants also talked about the importance of screening early and not waiting until the ‘damage was done’, with discussions around age and smoking. It was suggested that raising awareness about lung cancer and lung screening should be introduced to school children, embedding knowledge of screening from an early age. Participants were also largely accepting of the fact that lung screening could pick up other issues,R3: Anything that shows up as a side‐line to it is a benefit and I think most people would welcome it.R1: Yeah, I 100 per cent agree with that.R4: Yeah, totally agree with that one as well, an added bonus. (FG3, R1, 3 and 4)\n\nR3: Anything that shows up as a side‐line to it is a benefit and I think most people would welcome it.\nR1: Yeah, I 100 per cent agree with that.\nR4: Yeah, totally agree with that one as well, an added bonus. (FG3, R1, 3 and 4)\nHowever, there were certain caveats and conditions to participating in screening. The concept of targeted screening for smokers was problematic for participants, as they discussed other risk factors for lung cancer apart from smoking and that some people who never smoke go on to develop lung cancer:There's other causes of lung cancer, it's not just smoking. (FG1, R7)\n\nThere's other causes of lung cancer, it's not just smoking. (FG1, R7)\nParticipants introduced the potentially judgemental and stigmatizing nature of risk‐related eligibility to discussions. The importance of personal informed decision‐making and lack of coercion were also voiced by participants.\nFacilitators asked participants about the role of smoking cessation advice in the lung screening process. Participants suggested that this would not put them off participating, although they reported a dislike of being pushed into stopping smoking or judged for their smoking, such that language and tone were important,I think you have to have that balance … for people to [not] think, ‘oh, we're going there and we're going to have that shoved down our throat [i.e., “forced on us”]’. It has to be choice, but I think always giving people the appropriate choices if somebody is ready to stop, […] and giving them the information being there available, but I don't think making it part of something, because what would then happen is people would then think they're getting this shoved down our throat, we're getting judged for smoking, […] So I think, yes, it's good to have all the information, but it's how it's given. (FG1, R6)\n\nI think you have to have that balance … for people to [not] think, ‘oh, we're going there and we're going to have that shoved down our throat [i.e., “forced on us”]’. It has to be choice, but I think always giving people the appropriate choices if somebody is ready to stop, […] and giving them the information being there available, but I don't think making it part of something, because what would then happen is people would then think they're getting this shoved down our throat, we're getting judged for smoking, […] So I think, yes, it's good to have all the information, but it's how it's given. (FG1, R6)\nParticipants said that most people had received smoking cessation advice before, they already knew that smoking was harmful, and would not be offended by a health professional asking them about smoking. There was a strong sense among participants that the desire to quit smoking and action to quit was self‐motivated.\nThere is currently no national lung screening programme in the United Kingdom, although parts of NHS England are offering lung screening with LDCT.\n25\n Participants were largely unaware of the concept of targeted lung screening:No, I always thought that was something that happened if you develop, you know, or if they suspect you develop then you would have a check, otherwise nothing pre‐emptive…. (FG1, R5)\n\nNo, I always thought that was something that happened if you develop, you know, or if they suspect you develop then you would have a check, otherwise nothing pre‐emptive…. (FG1, R5)\nMost participants had heard of and participated in other forms of screening, including for breast, cervical and bowel cancer, and a few had been referred for lung checks for other reasons. They also described family members having had cancers picked up in this way, as well as their own experience of cancer:For me, it really is, it's very important to be screened, especially breast, bowel, anything. I suffered myself from throat cancer ten years ago and I've been in remission for the past four years, so it's urgently important that people get this done, yeah. (FG3, R4)\n\nFor me, it really is, it's very important to be screened, especially breast, bowel, anything. I suffered myself from throat cancer ten years ago and I've been in remission for the past four years, so it's urgently important that people get this done, yeah. (FG3, R4)\nWhen we described what lung screening would entail, participants were very supportive of this form of screening being available and welcomed the chance to have their lungs screened, with multiple participants saying it was ‘a great idea’. Participants were aware of the benefits of early (asymptomatic) detection and treatment:I think the screening is a good idea to catch things earlier or to see if somebody's got the disease or whatever that they didn't know they had. (FG1, R7)\n\nI think the screening is a good idea to catch things earlier or to see if somebody's got the disease or whatever that they didn't know they had. (FG1, R7)\nParticipants also talked about the importance of screening early and not waiting until the ‘damage was done’, with discussions around age and smoking. It was suggested that raising awareness about lung cancer and lung screening should be introduced to school children, embedding knowledge of screening from an early age. Participants were also largely accepting of the fact that lung screening could pick up other issues,R3: Anything that shows up as a side‐line to it is a benefit and I think most people would welcome it.R1: Yeah, I 100 per cent agree with that.R4: Yeah, totally agree with that one as well, an added bonus. (FG3, R1, 3 and 4)\n\nR3: Anything that shows up as a side‐line to it is a benefit and I think most people would welcome it.\nR1: Yeah, I 100 per cent agree with that.\nR4: Yeah, totally agree with that one as well, an added bonus. (FG3, R1, 3 and 4)\nHowever, there were certain caveats and conditions to participating in screening. The concept of targeted screening for smokers was problematic for participants, as they discussed other risk factors for lung cancer apart from smoking and that some people who never smoke go on to develop lung cancer:There's other causes of lung cancer, it's not just smoking. (FG1, R7)\n\nThere's other causes of lung cancer, it's not just smoking. (FG1, R7)\nParticipants introduced the potentially judgemental and stigmatizing nature of risk‐related eligibility to discussions. The importance of personal informed decision‐making and lack of coercion were also voiced by participants.\nFacilitators asked participants about the role of smoking cessation advice in the lung screening process. Participants suggested that this would not put them off participating, although they reported a dislike of being pushed into stopping smoking or judged for their smoking, such that language and tone were important,I think you have to have that balance … for people to [not] think, ‘oh, we're going there and we're going to have that shoved down our throat [i.e., “forced on us”]’. It has to be choice, but I think always giving people the appropriate choices if somebody is ready to stop, […] and giving them the information being there available, but I don't think making it part of something, because what would then happen is people would then think they're getting this shoved down our throat, we're getting judged for smoking, […] So I think, yes, it's good to have all the information, but it's how it's given. (FG1, R6)\n\nI think you have to have that balance … for people to [not] think, ‘oh, we're going there and we're going to have that shoved down our throat [i.e., “forced on us”]’. It has to be choice, but I think always giving people the appropriate choices if somebody is ready to stop, […] and giving them the information being there available, but I don't think making it part of something, because what would then happen is people would then think they're getting this shoved down our throat, we're getting judged for smoking, […] So I think, yes, it's good to have all the information, but it's how it's given. (FG1, R6)\nParticipants said that most people had received smoking cessation advice before, they already knew that smoking was harmful, and would not be offended by a health professional asking them about smoking. There was a strong sense among participants that the desire to quit smoking and action to quit was self‐motivated.\n[SUBTITLE] Barriers and facilitators to screening [SUBSECTION] It was clear that while there was strong support for the concept of lung screening with participants overwhelmingly in favour of it, a number of factors were raised which qualified their response—with the prospect of mediating the gap between reported intentions and performed screening behaviours in a real‐world scenario. Some of these same issues were potential motivators to participate in lung screening. Broadly, these can be represented under individual level and practical and system level factors.\n[SUBTITLE] Individual level influences on screening intentions [SUBSECTION] On an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nOn an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\n[SUBTITLE] Practical and system barriers [SUBSECTION] A number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.\nA number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.\nIt was clear that while there was strong support for the concept of lung screening with participants overwhelmingly in favour of it, a number of factors were raised which qualified their response—with the prospect of mediating the gap between reported intentions and performed screening behaviours in a real‐world scenario. Some of these same issues were potential motivators to participate in lung screening. Broadly, these can be represented under individual level and practical and system level factors.\n[SUBTITLE] Individual level influences on screening intentions [SUBSECTION] On an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nOn an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\n[SUBTITLE] Practical and system barriers [SUBSECTION] A number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.\nA number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.\n[SUBTITLE] Promoting screening and implementation ideas [SUBSECTION] There was considerable discussion in each focus group on the acceptability and accessibility of screening, leading to suggestions of ways of promoting and implementing screening to increase participation. Participants' views on accessibility were of particular interest due to their socioeconomic status and geographical diversity. Suggestions related to the cognitive and psychological barriers to screening as well as practical issues.\n[SUBTITLE] Managing fears and expectations [SUBSECTION] To address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\nTo address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n[SUBTITLE] Improving accessibility [SUBSECTION] In addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\nIn addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\nThere was considerable discussion in each focus group on the acceptability and accessibility of screening, leading to suggestions of ways of promoting and implementing screening to increase participation. Participants' views on accessibility were of particular interest due to their socioeconomic status and geographical diversity. Suggestions related to the cognitive and psychological barriers to screening as well as practical issues.\n[SUBTITLE] Managing fears and expectations [SUBSECTION] To address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\nTo address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n[SUBTITLE] Improving accessibility [SUBSECTION] In addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\nIn addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)", "There is currently no national lung screening programme in the United Kingdom, although parts of NHS England are offering lung screening with LDCT.\n25\n Participants were largely unaware of the concept of targeted lung screening:No, I always thought that was something that happened if you develop, you know, or if they suspect you develop then you would have a check, otherwise nothing pre‐emptive…. (FG1, R5)\n\nNo, I always thought that was something that happened if you develop, you know, or if they suspect you develop then you would have a check, otherwise nothing pre‐emptive…. (FG1, R5)\nMost participants had heard of and participated in other forms of screening, including for breast, cervical and bowel cancer, and a few had been referred for lung checks for other reasons. They also described family members having had cancers picked up in this way, as well as their own experience of cancer:For me, it really is, it's very important to be screened, especially breast, bowel, anything. I suffered myself from throat cancer ten years ago and I've been in remission for the past four years, so it's urgently important that people get this done, yeah. (FG3, R4)\n\nFor me, it really is, it's very important to be screened, especially breast, bowel, anything. I suffered myself from throat cancer ten years ago and I've been in remission for the past four years, so it's urgently important that people get this done, yeah. (FG3, R4)\nWhen we described what lung screening would entail, participants were very supportive of this form of screening being available and welcomed the chance to have their lungs screened, with multiple participants saying it was ‘a great idea’. Participants were aware of the benefits of early (asymptomatic) detection and treatment:I think the screening is a good idea to catch things earlier or to see if somebody's got the disease or whatever that they didn't know they had. (FG1, R7)\n\nI think the screening is a good idea to catch things earlier or to see if somebody's got the disease or whatever that they didn't know they had. (FG1, R7)\nParticipants also talked about the importance of screening early and not waiting until the ‘damage was done’, with discussions around age and smoking. It was suggested that raising awareness about lung cancer and lung screening should be introduced to school children, embedding knowledge of screening from an early age. Participants were also largely accepting of the fact that lung screening could pick up other issues,R3: Anything that shows up as a side‐line to it is a benefit and I think most people would welcome it.R1: Yeah, I 100 per cent agree with that.R4: Yeah, totally agree with that one as well, an added bonus. (FG3, R1, 3 and 4)\n\nR3: Anything that shows up as a side‐line to it is a benefit and I think most people would welcome it.\nR1: Yeah, I 100 per cent agree with that.\nR4: Yeah, totally agree with that one as well, an added bonus. (FG3, R1, 3 and 4)\nHowever, there were certain caveats and conditions to participating in screening. The concept of targeted screening for smokers was problematic for participants, as they discussed other risk factors for lung cancer apart from smoking and that some people who never smoke go on to develop lung cancer:There's other causes of lung cancer, it's not just smoking. (FG1, R7)\n\nThere's other causes of lung cancer, it's not just smoking. (FG1, R7)\nParticipants introduced the potentially judgemental and stigmatizing nature of risk‐related eligibility to discussions. The importance of personal informed decision‐making and lack of coercion were also voiced by participants.\nFacilitators asked participants about the role of smoking cessation advice in the lung screening process. Participants suggested that this would not put them off participating, although they reported a dislike of being pushed into stopping smoking or judged for their smoking, such that language and tone were important,I think you have to have that balance … for people to [not] think, ‘oh, we're going there and we're going to have that shoved down our throat [i.e., “forced on us”]’. It has to be choice, but I think always giving people the appropriate choices if somebody is ready to stop, […] and giving them the information being there available, but I don't think making it part of something, because what would then happen is people would then think they're getting this shoved down our throat, we're getting judged for smoking, […] So I think, yes, it's good to have all the information, but it's how it's given. (FG1, R6)\n\nI think you have to have that balance … for people to [not] think, ‘oh, we're going there and we're going to have that shoved down our throat [i.e., “forced on us”]’. It has to be choice, but I think always giving people the appropriate choices if somebody is ready to stop, […] and giving them the information being there available, but I don't think making it part of something, because what would then happen is people would then think they're getting this shoved down our throat, we're getting judged for smoking, […] So I think, yes, it's good to have all the information, but it's how it's given. (FG1, R6)\nParticipants said that most people had received smoking cessation advice before, they already knew that smoking was harmful, and would not be offended by a health professional asking them about smoking. There was a strong sense among participants that the desire to quit smoking and action to quit was self‐motivated.", "It was clear that while there was strong support for the concept of lung screening with participants overwhelmingly in favour of it, a number of factors were raised which qualified their response—with the prospect of mediating the gap between reported intentions and performed screening behaviours in a real‐world scenario. Some of these same issues were potential motivators to participate in lung screening. Broadly, these can be represented under individual level and practical and system level factors.\n[SUBTITLE] Individual level influences on screening intentions [SUBSECTION] On an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nOn an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\n[SUBTITLE] Practical and system barriers [SUBSECTION] A number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.\nA number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.", "On an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).", "The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)", "Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)", "A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)", "A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).", "A number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.", "There was considerable discussion in each focus group on the acceptability and accessibility of screening, leading to suggestions of ways of promoting and implementing screening to increase participation. Participants' views on accessibility were of particular interest due to their socioeconomic status and geographical diversity. Suggestions related to the cognitive and psychological barriers to screening as well as practical issues.\n[SUBTITLE] Managing fears and expectations [SUBSECTION] To address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\nTo address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n[SUBTITLE] Improving accessibility [SUBSECTION] In addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\nIn addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)", "To address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)", "In addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)", "This focus group study is the first of its kind to ascertain the views of Scottish residents on a potential LDCT lung screening programme and identify likely barriers and facilitators in this context. Knowledge of lung screening was low among the focus group participants, although they showed awareness and personal experience of other screening programmes. Participants were very supportive of the idea of lung screening and harnessed the early detection narrative to discuss the importance of taking part. Participants reported the process of lung screening via a LDCT scan was acceptable. Two key barrier types were identified: individual level influences on screening intentions, and practical and system barriers. Within the individual level factors, emotional and psychological concerns related to fear of a cancer diagnosis, mistrust, fatalism and perceived stigma were dominant in focus group discussion. For some, screening was part of being a health conscious citizen and prioritizing health matters, while others based their decision‐making on their perceived risk. A number of potential practical barriers to lung screening participation were mooted that were particularly relevant for people living in deprived and rural communities. These included travel, cost, time and competing priorities. Maximizing accessibility was also key in the discussion and a distinct recommendation for future programmes.", "There is a strong consistency in our findings with the growing body of evidence looking at attitudes to screening and screening behaviour, across a range of screening programmes and for lung screening in particular, in the United Kingdom and beyond.\n26\n, \n27\n, \n28\n LDCT screening for lung cancer is largely unheard of in Scotland but has a high degree of acceptability more broadly, or among those who have participated in screening.\n27\n, \n29\n, \n30\n There was an evident awareness among participants of the benefits of early detection and thus support for lung screening, in line with other research.\n17\n, \n31\n\n\nFear was identified as one of the most common psychological barriers to lung screening, which is reflected in the literature in both survey and qualitative explorations of attitudes to screening.\n28\n, \n32\n, \n33\n Linked to this was a sense of fatalism or predicted fatalism among older generations, also mirrored in comparable studies of lung screening.\n29\n, \n34\n While there is evidence to suggest that the fear and anxiety associated with lung screening participation is transient\n35\n and can be a motivating factor to be screened, quit smoking and even other cancer‐preventing health behaviour change,\n36\n, \n37\n, \n38\n it is still vital to minimize this emotional response by addressing and managing it. Positive messaging in the promotion of screening, such as sharing the treatment successes and mortality gains from early detection, is one potential step in approaching this.\nNotions of risk were a key component of our group's considerations of whether or not someone would take part in screening, often related to not experiencing symptoms and having stopped smoking. Perceived risk has been identified widely in the literature to explain decision‐making in relation to screening and help‐seeking for symptomatic illness.\n28\n, \n33\n, \n39\n, \n40\n Risk and decision‐making are discussed further below.\nPerceived judgement and stigma related to smoking featured in focus group discussions. This is widely evident in the literature, along with self‐blame.\n31\n, \n38\n, \n41\n Stigma has been identified in the literature as a barrier to help‐seeking for signs of lung cancer and it seems this also applies to screening participation.\n42\n Related to this was a sense of fatalism—participants in our study did not consider they would blame themselves if they developed lung cancer, but some did feel that the damage was already done and screening could not change that.\n17\n, \n29\n, \n38\n However, this was not a clear barrier. For some, it was a good reason to detect any inevitable lung cancer at an early stage, suggesting that issues such as fear, blame and fatalism are complex and operate on a pendulum when prompting action or inaction.\nDiscussions of stigma inevitably moved onto smoking cessation. Smoking cessation advice was broadly acceptable to our participants, but only if delivered in a noncoercive way; again, other studies have had similar findings.\n17\n, \n43\n, \n44\n, \n45\n In our focus group study, it appeared that participants may have become desensitized to smoking cessation advice. This has implications when considering brief interventions for smoking, and would need further exploration to understand whether small prompts may be enough to stimulate action among people who have intentions to quit smoking. There is evidence to suggest that brief interventions can be effective.\n46\n, \n47\n There is also emerging evidence that incorporating smoking cessation advice into lung screening is effective—seeing images of lungs has been a strong motivator to quit smoking\n36\n as well as being central to the long‐term cost‐effectiveness of screening.\n13\n\n\nSome participants in our focus groups described a difficult relationship and a level of mistrust in interacting with health professionals and services, often related to poor past experiences or a sociocultural divide. Such perceptions are often ingrained in more deprived communities and are often reported in the literature—relating not only to lung screening,\n17\n, \n32\n but beyond to studies of help‐seeking behaviour and doctor–patient relationships.\n48\n Again, this is a complex feature of health service engagement and can be linked to other barriers such as low self‐efficacy, low health literacy and power dynamics, which can be particularly divisive in terms of equity in access to health care for people in disadvantaged groups.\n48\n, \n49\n, \n50\n Development of interventions to repair broken relationships with the health service, a perceived authoritative institution, and other methods to improve accessibility such as communication training for professionals, targeted awareness campaigns and community engagement strategies can address inequities in access to screening services.\n51\n Primary care has an important role to play; current workforce shortages need to be addressed, and solutions identified which do not generate significant extra burden for primary care staff—one example is streamlining procedures to identify high‐risk patients from practice data.\n7\n, \n8\n It also seems logical to harness the successes of implementation strategies for other cancer screening (e.g., the UK's bowel screening programmes).\n52\n, \n53\n\n\nIn addition to the psychosocial issues discussed by focus group participants, practical barriers to participation in lung screening including time, cost, travel and competing work or other commitments, were also mooted. Practical barriers are commonly reported throughout the evidence base related to engaging with screening, with a suggestion that these barriers are heightened in more deprived groups.\n27\n, \n32\n, \n46\n\n\nVon Wagner et al.\n50\n have developed a model of screening behaviour, accounting for a range of factors identified in relation to wider health behaviours, such as health identity and self‐efficacy, that can be usefully mapped onto the findings of this study. Similarly, Robb\n54\n has developed the I‐SAM model to understand screening participation. Application of these models to follow‐up interviews as part of the planned pilot lung screening study in Scotland will be enlightening to confirm the salience of these to screening participation and nonparticipation.\nThere is an abundance of early cancer detection research exploring how people appraise bodily changes, evaluate risk and decide to seek medical help, as well as conceptual models to understand these processes.\n55\n, \n56\n, \n57\n There is also some utility in applying these to screening behaviour, often in the absence of symptoms, to understand nonparticipation in screening. For example, Kummer et al.'s\n37\n, \n39\n cognitive heuristics for help‐seeking for cancer symptoms may act as prompts or inhibitors to participate in screening. Understanding these factors in the context of deprived populations adds further considerations that may compound behaviour in terms of available resources (cognitive, psychological and practical), competing demands and permeability of services.\n48\n, \n49\n\n\nIn terms of ideas to overcome barriers, participants focused primarily on positive messaging in information materials and advertising, and accessibility through the provision of mobile screening vans similar to those used in breast screening or for Covid‐19 vaccination. Positive and nonjudgemental messaging focusing on the gains on offer from screening and early detection has also been found elsewhere and incorporated into the pilot lung screening provision.\n58\n, \n59\n Travel to access screening services as well as fear of hospital environments have also been identified in the literature, and evidence evaluating the use of mobile screening vans to address these issues is beginning to accumulate.\n12\n, \n60\n\n", "This set of focus groups provides a rich data set with an in‐depth discussion of the concept of lung cancer screening and anticipated barriers and facilitators. Focus group participants were a self‐selecting group who are accustomed to taking part in research studies, and who received a financial incentive. However, this form of recruitment strategy does allow access to people from a range of different backgrounds, including both deprived and rural areas of Scotland that may otherwise have been challenging to recruit. Three of the 24 participants were from ethnic minority groups, reflecting the general demographic in the Scottish population. Focus groups were conducted online. Video conferencing software facilitates easy interaction with people from around the country but can pose challenges. These include failing technology, building rapport and ensuring balanced participation within the group simply by being in a room together and acknowledging social cues. It was also necessary to consider that participants were talking about a hypothetical scenario and their intended behaviour, something we know does not simply translate into action.\n61\n Speaking to people who have taken part in screening or chosen not to take part is also essential to further understand the relationship between intention and behaviour.\nWe also reflected on the group dynamic together with the nature of our role in conducting the focus groups, and whether this was likely to influence people in agreeing with and supporting the concept of lung screening. However, the open nature of questioning, reminding participants that we genuinely wanted to hear their views, and the self‐selecting group of individuals who were quite assured in their own responses, suggested that we did not shape this narrative.", "This study informs the development of strategies to improve uptake and informed choice in lung screening. It is essential to understand people's health beliefs and behaviours and to target the barriers to implement a patient‐centred service using a theory‐driven approach.\n62\n This work adds to a growing evidence base shedding light on the behavioural aspects of screening participation and will inform the design and implementation of a new lung screening pilot in Scotland\n40\n, \n45\n, \n46\n (see Box 2 for implementation ideas generated from this work). Minimizing practical barriers is also likely to be instrumental in improving participation and addressing inequity in access to screening. As such, information materials, methods of communication and the design of the process involved in screening (e.g., minimizing the steps, time commitment and waiting intervals), and sensitive, supportive messaging that addresses stigma and fear are all important components of a pilot to break down some of the known barriers. Drawing on the similarities with research based on other UK pilot studies, we are modelling optimized study and participant materials.\n59\n, \n63\n, \n64\n While there is consistency in the findings compared with existing work, it was important to explore the Scottish context with a diverse sample of participants to consider how rurality and deprivation presented any unique issues. Understanding ethnic variations in views on lung screening participation will also be important to ensure equitable provision and uptake of screening. A high burden of multimorbidity is also characteristic of the Scottish population and should be examined in future studies.\n\nMinimize steps in the screening process to lower opportunities for delays and associated distressAvoiding unnecessary travel to scanning facilities with the chance to discuss screening concerns, address fears and perceived stigma, and facilitate informed decision‐makingUse of mobile screening vans as a ‘one stop shop’ to address resource constraints and travel issues for people living in deprived and rural areasConsider covering the cost of travel expenses to screening facilities to ensure equitable accessIncorporating discussion of fears associated with screening into information materialsEnsuring positive messaging with nonjudgemental language around smoking behaviourEnsuring a timely and sensitive approach to smoking cessation advice\n\nMinimize steps in the screening process to lower opportunities for delays and associated distress\nAvoiding unnecessary travel to scanning facilities with the chance to discuss screening concerns, address fears and perceived stigma, and facilitate informed decision‐making\nUse of mobile screening vans as a ‘one stop shop’ to address resource constraints and travel issues for people living in deprived and rural areas\nConsider covering the cost of travel expenses to screening facilities to ensure equitable access\nIncorporating discussion of fears associated with screening into information materials\nEnsuring positive messaging with nonjudgemental language around smoking behaviour\nEnsuring a timely and sensitive approach to smoking cessation advice", "All authors apart from Jasmin Rostron and Lynsey R. Brown contributed toward the design of the focus groups and helped develop study documents. Debbie Cavers and Mia Nelson conducted the focus groups. Debbie Cavers, Mia Nelson and Jasmin Rostron coded and analysed the transcripts. Lynsey R. Brown read and commented on the analysis. Debbie Cavers drafted the manuscript and all authors edited and refined subsequent drafts.", "The LUNGSCOT focus group study was approved by the University of Edinburgh Medical School Ethics Committee on 19 March 2021, reference 21‐EMREC‐002. All participants gave consent before taking part in the focus groups." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "METHODS", "Identifying participants", "Recruitment and sampling", "Focus groups and consent", "Analysis", "Patient advisory group", "FINDINGS", "Knowledge, awareness and acceptability of lung screening", "Barriers and facilitators to screening", "Individual level influences on screening intentions", "Psychological and emotional concerns", "Perceived risk and fatalism", "Stigma and judgement as a barrier", "Mistrust of healthcare professionals and services", "Practical and system barriers", "Promoting screening and implementation ideas", "Managing fears and expectations", "Improving accessibility", "DISCUSSION", "Summary", "Comparison with existing literature and theory", "Strengths and limitations", "Implications and future work", "CONCLUSIONS", "AUTHOR CONTRIBUTIONS", "CONFLICT OF INTEREST", "ETHICS STATEMENT", "Supporting information" ]
[ "Lung cancer remains one of the major causes of cancer mortality globally, and Scotland has high incidence and low survival rates compared with the rest of Europe.\n1\n, \n2\n, \n3\n, \n4\n Despite many initiatives to raise awareness and encourage early symptomatic presentation, most lung cancers are diagnosed at a late stage, and overall survival is poor.\n5\n A recent Public Health Scotland report revealed that the Covid‐19 pandemic has had a further negative impact on the clinical presentation of lung cancer, with a reduction in diagnoses during the lockdown periods, and an anticipated wave of late diagnoses from this backlog.\n6\n\n\nOver the last 5 years, there has been a growing body of evidence from trials in the United States and continental Europe, and pilot studies in the United Kingdom, demonstrating survival gains from low‐dose computed tomography (LDCT) screening for lung cancer in high‐risk populations.\n7\n, \n8\n, \n9\n, \n10\n A recent meta‐analysis of this evidence provides a strong case for the implementation of targeted lung screening, supported by expert clinical opinion,\n11\n and the UK National Screening Committee has recently recommended in favour of targeted lung screening. However, there are still many lung screening implementation challenges to be addressed.\n12\n, \n13\n Importantly, lung screening is most effective if targeted at high‐risk groups—ideally using validated risk prediction tools to assess who is at the highest risk of developing lung cancer and would most benefit from screening, namely smokers or recent quitters, aged 55–74 years.\nTo date, UK pilots and trials, including the Early Detection of Cancer of the Lung Scotland (ECLS) trial in Scotland using blood biomarkers to detect early signs of lung cancer, have shown variable and socially patterned uptake of screening,\n8\n, \n14\n, \n15\n, \n16\n and work has been done to understand barriers to participation. Uptake remains lower in more marginalized groups—that is, heavy smokers living in more deprived areas.\n17\n Further research to understand the views of Scotland's population on the acceptability of lung screening, and barriers and facilitators (related to issues of geography, rural and urban deprivation and a high burden of multimorbidity), will help shape future pilots and programmes in Scotland.\nThis focus group study explores the views of people potentially eligible for lung screening, identifies perceived barriers and facilitators to participation, and examines strategies to optimize the implementation of lung screening in Scotland. It forms part of the LUNGSCOT study, which is examining the feasibility of introducing lung screening in Scotland.\n18\n\n", "We conducted three focus groups with self‐reported ‘heavy smokers’ living in rural, urban and deprived areas of Scotland to ascertain their views on barriers to lung screening. Focus groups were considered the most appropriate method for engaging with those eligible for screening, as they enabled an understanding of how participants perceive the prospect of lung screening through discussion and they enable the sharing and development of ideas.\n19\n Findings from the focus groups will feed into the development and design of the lung screening pilot. We employed a range of strategies and reflexive practices to ensure that focus groups were participant‐led and data‐driven.\n20\n\n\n[SUBTITLE] Identifying participants [SUBSECTION] Participants were recruited through Taylor McKenzie (TM), a Scottish‐based company that specializes in qualitative research, to identify members of the public eligible to take part in the focus groups (https://www.taylormckenzie.co.uk). TM developed a study‐specific screening questionnaire (further details below) to allow the purposive sampling of eligible participants from their extensive database of people willing to take part in health, social or marketing research.\nParticipants were recruited through Taylor McKenzie (TM), a Scottish‐based company that specializes in qualitative research, to identify members of the public eligible to take part in the focus groups (https://www.taylormckenzie.co.uk). TM developed a study‐specific screening questionnaire (further details below) to allow the purposive sampling of eligible participants from their extensive database of people willing to take part in health, social or marketing research.\n[SUBTITLE] Recruitment and sampling [SUBSECTION] We aimed to recruit up to 24 people from across Scotland to take part in three separate focus groups of 8 people, considered to be an appropriate number to identify a range of views.\n21\n Interested people responded to recruitment notices posted on TM's mailing list and social media pages. We used the inclusion and exclusion criteria listed In Box 1.\nInclusion criteria\n\nMen and womenAge 45–70 (inclusive)Current residence in ScotlandSelf‐identify as smoker or recent quitter (within the last 2 years)Able to undertake focus group interview in EnglishWilling to discuss their views on lung screening\n\nMen and women\nAge 45–70 (inclusive)\nCurrent residence in Scotland\nSelf‐identify as smoker or recent quitter (within the last 2 years)\nAble to undertake focus group interview in English\nWilling to discuss their views on lung screening\nExclusion criteria\n\nLacking capacity to give informed consentNever smokerSmoker who quit more than 2 years agoNon‐English speakers, preventing them from comfortably taking part in a discussion\n\nLacking capacity to give informed consent\nNever smoker\nSmoker who quit more than 2 years ago\nNon‐English speakers, preventing them from comfortably taking part in a discussion\nThose who responded were provided with a study information sheet by TM and given 7–14 days to consider taking part. TM drew up a list of eligible participants (based on information on their database, e.g., socioeconomic grade [SEG], occupation, and from speaking to potential participants directly, e.g., smoking status) and added it to a secure portal for the researchers to access and review. Eligible participants were allocated to a focus group at a preset date and time. Participants were offered a financial reimbursement for their time, paid via TM.\nWe aimed to recruit up to 24 people from across Scotland to take part in three separate focus groups of 8 people, considered to be an appropriate number to identify a range of views.\n21\n Interested people responded to recruitment notices posted on TM's mailing list and social media pages. We used the inclusion and exclusion criteria listed In Box 1.\nInclusion criteria\n\nMen and womenAge 45–70 (inclusive)Current residence in ScotlandSelf‐identify as smoker or recent quitter (within the last 2 years)Able to undertake focus group interview in EnglishWilling to discuss their views on lung screening\n\nMen and women\nAge 45–70 (inclusive)\nCurrent residence in Scotland\nSelf‐identify as smoker or recent quitter (within the last 2 years)\nAble to undertake focus group interview in English\nWilling to discuss their views on lung screening\nExclusion criteria\n\nLacking capacity to give informed consentNever smokerSmoker who quit more than 2 years agoNon‐English speakers, preventing them from comfortably taking part in a discussion\n\nLacking capacity to give informed consent\nNever smoker\nSmoker who quit more than 2 years ago\nNon‐English speakers, preventing them from comfortably taking part in a discussion\nThose who responded were provided with a study information sheet by TM and given 7–14 days to consider taking part. TM drew up a list of eligible participants (based on information on their database, e.g., socioeconomic grade [SEG], occupation, and from speaking to potential participants directly, e.g., smoking status) and added it to a secure portal for the researchers to access and review. Eligible participants were allocated to a focus group at a preset date and time. Participants were offered a financial reimbursement for their time, paid via TM.\n[SUBTITLE] Focus groups and consent [SUBSECTION] Three focus groups, lasting approximately 75 min, were run virtually using the online video conferencing platform Zoom, selected due to likely participant familiarity with it, and to comply with the prevailing government restrictions on face‐to‐face meetings at the time due to the Covid‐19 pandemic. Participants were asked to sign a digital consent form via TM ahead of the focus group and verbal consent was agreed upon at the beginning of each focus group. Focus groups were led and facilitated by two researchers (D. C. and M. N.—health services researchers), with one group comprising those living in rural areas and the other two urban groups. Participants did not know one another before the focus group. The format and content of the focus groups were developed by a subgroup of the research team with expertise in behavioural aspects of cancer screening, drawing on relevant literature. The lung screening process was explained to participants: eligible people would be offered a LDCT scan to detect any lung conditions, one of which is lung cancer. The topic guide (see Supporting Information: 1) covered views on lung screening, with a particular focus on perceived barriers to taking part, personal resources to facilitate screening, understanding of the process, and input on what a good screening programme would look like. With consent, focus groups were recorded.\nThree focus groups, lasting approximately 75 min, were run virtually using the online video conferencing platform Zoom, selected due to likely participant familiarity with it, and to comply with the prevailing government restrictions on face‐to‐face meetings at the time due to the Covid‐19 pandemic. Participants were asked to sign a digital consent form via TM ahead of the focus group and verbal consent was agreed upon at the beginning of each focus group. Focus groups were led and facilitated by two researchers (D. C. and M. N.—health services researchers), with one group comprising those living in rural areas and the other two urban groups. Participants did not know one another before the focus group. The format and content of the focus groups were developed by a subgroup of the research team with expertise in behavioural aspects of cancer screening, drawing on relevant literature. The lung screening process was explained to participants: eligible people would be offered a LDCT scan to detect any lung conditions, one of which is lung cancer. The topic guide (see Supporting Information: 1) covered views on lung screening, with a particular focus on perceived barriers to taking part, personal resources to facilitate screening, understanding of the process, and input on what a good screening programme would look like. With consent, focus groups were recorded.\n[SUBTITLE] Analysis [SUBSECTION] Focus group recordings were transcribed verbatim and subject to Braun and Clarke's\n22\n thematic analysis, chosen as it is the most commonly used approach considered appropriate to derive key themes and ideas from the group discussions, taking context into account\n19\n and consistent with social constructionist underpinnings. Using thematic analysis allowed us to incorporate guidance specific to focus group interviewing and its impact on analysis, for example, considering dynamics, social comparison and power imbalances within groups.\n23\n, \n24\n Transcripts were read repeatedly and compared and contrasted to develop a set of common codes by D. C. These codes were applied back across the data and assigned to excerpts from the focus groups using QSR NVivo version 12 Pro (www.qsrinternational.com) by D. C. and J. R. (a research intern). Codes were further refined and a set of overarching themes and subthemes were inductively derived to interpret and explain the data, in discussion with the wider research team and patient advisory group. The LUNGSCOT team comprises health services researchers, health psychologists, clinicians, a health economist and patient and carer representatives. Themes were placed in the context of existing literature and theory to incorporate our findings into the wider evidence base.\nFocus group recordings were transcribed verbatim and subject to Braun and Clarke's\n22\n thematic analysis, chosen as it is the most commonly used approach considered appropriate to derive key themes and ideas from the group discussions, taking context into account\n19\n and consistent with social constructionist underpinnings. Using thematic analysis allowed us to incorporate guidance specific to focus group interviewing and its impact on analysis, for example, considering dynamics, social comparison and power imbalances within groups.\n23\n, \n24\n Transcripts were read repeatedly and compared and contrasted to develop a set of common codes by D. C. These codes were applied back across the data and assigned to excerpts from the focus groups using QSR NVivo version 12 Pro (www.qsrinternational.com) by D. C. and J. R. (a research intern). Codes were further refined and a set of overarching themes and subthemes were inductively derived to interpret and explain the data, in discussion with the wider research team and patient advisory group. The LUNGSCOT team comprises health services researchers, health psychologists, clinicians, a health economist and patient and carer representatives. Themes were placed in the context of existing literature and theory to incorporate our findings into the wider evidence base.\n[SUBTITLE] Patient advisory group [SUBSECTION] The patient advisory group was convened for the purpose of the wider LUNGSCOT study. The group comprises three patients and one carer with experience in lung cancer and two patients with other cancers. The group has been involved in the study design and commented on study documentation as well as two advisors reading transcripts and sharing their views on the analysis. The group meets quarterly to discuss study progress and opportunities to get involved in study tasks.\nThe patient advisory group was convened for the purpose of the wider LUNGSCOT study. The group comprises three patients and one carer with experience in lung cancer and two patients with other cancers. The group has been involved in the study design and commented on study documentation as well as two advisors reading transcripts and sharing their views on the analysis. The group meets quarterly to discuss study progress and opportunities to get involved in study tasks.", "Participants were recruited through Taylor McKenzie (TM), a Scottish‐based company that specializes in qualitative research, to identify members of the public eligible to take part in the focus groups (https://www.taylormckenzie.co.uk). TM developed a study‐specific screening questionnaire (further details below) to allow the purposive sampling of eligible participants from their extensive database of people willing to take part in health, social or marketing research.", "We aimed to recruit up to 24 people from across Scotland to take part in three separate focus groups of 8 people, considered to be an appropriate number to identify a range of views.\n21\n Interested people responded to recruitment notices posted on TM's mailing list and social media pages. We used the inclusion and exclusion criteria listed In Box 1.\nInclusion criteria\n\nMen and womenAge 45–70 (inclusive)Current residence in ScotlandSelf‐identify as smoker or recent quitter (within the last 2 years)Able to undertake focus group interview in EnglishWilling to discuss their views on lung screening\n\nMen and women\nAge 45–70 (inclusive)\nCurrent residence in Scotland\nSelf‐identify as smoker or recent quitter (within the last 2 years)\nAble to undertake focus group interview in English\nWilling to discuss their views on lung screening\nExclusion criteria\n\nLacking capacity to give informed consentNever smokerSmoker who quit more than 2 years agoNon‐English speakers, preventing them from comfortably taking part in a discussion\n\nLacking capacity to give informed consent\nNever smoker\nSmoker who quit more than 2 years ago\nNon‐English speakers, preventing them from comfortably taking part in a discussion\nThose who responded were provided with a study information sheet by TM and given 7–14 days to consider taking part. TM drew up a list of eligible participants (based on information on their database, e.g., socioeconomic grade [SEG], occupation, and from speaking to potential participants directly, e.g., smoking status) and added it to a secure portal for the researchers to access and review. Eligible participants were allocated to a focus group at a preset date and time. Participants were offered a financial reimbursement for their time, paid via TM.", "Three focus groups, lasting approximately 75 min, were run virtually using the online video conferencing platform Zoom, selected due to likely participant familiarity with it, and to comply with the prevailing government restrictions on face‐to‐face meetings at the time due to the Covid‐19 pandemic. Participants were asked to sign a digital consent form via TM ahead of the focus group and verbal consent was agreed upon at the beginning of each focus group. Focus groups were led and facilitated by two researchers (D. C. and M. N.—health services researchers), with one group comprising those living in rural areas and the other two urban groups. Participants did not know one another before the focus group. The format and content of the focus groups were developed by a subgroup of the research team with expertise in behavioural aspects of cancer screening, drawing on relevant literature. The lung screening process was explained to participants: eligible people would be offered a LDCT scan to detect any lung conditions, one of which is lung cancer. The topic guide (see Supporting Information: 1) covered views on lung screening, with a particular focus on perceived barriers to taking part, personal resources to facilitate screening, understanding of the process, and input on what a good screening programme would look like. With consent, focus groups were recorded.", "Focus group recordings were transcribed verbatim and subject to Braun and Clarke's\n22\n thematic analysis, chosen as it is the most commonly used approach considered appropriate to derive key themes and ideas from the group discussions, taking context into account\n19\n and consistent with social constructionist underpinnings. Using thematic analysis allowed us to incorporate guidance specific to focus group interviewing and its impact on analysis, for example, considering dynamics, social comparison and power imbalances within groups.\n23\n, \n24\n Transcripts were read repeatedly and compared and contrasted to develop a set of common codes by D. C. These codes were applied back across the data and assigned to excerpts from the focus groups using QSR NVivo version 12 Pro (www.qsrinternational.com) by D. C. and J. R. (a research intern). Codes were further refined and a set of overarching themes and subthemes were inductively derived to interpret and explain the data, in discussion with the wider research team and patient advisory group. The LUNGSCOT team comprises health services researchers, health psychologists, clinicians, a health economist and patient and carer representatives. Themes were placed in the context of existing literature and theory to incorporate our findings into the wider evidence base.", "The patient advisory group was convened for the purpose of the wider LUNGSCOT study. The group comprises three patients and one carer with experience in lung cancer and two patients with other cancers. The group has been involved in the study design and commented on study documentation as well as two advisors reading transcripts and sharing their views on the analysis. The group meets quarterly to discuss study progress and opportunities to get involved in study tasks.", "Eleven females and 14 males aged 45–70 years living in a mix of urban and rural areas in Scotland took part across the three focus groups. Eleven participants were current smokers, and 14 had quit within the previous 2 years. All participants were from the lower socio‐economic grades (SEG): C2 (skilled manual workers), D (semiskilled and unskilled manual workers) and E (nonworking). Twenty‐one participants were White British/Scottish, one person was Black British, one British Asian and one South Asian. All participants had school‐level or vocational qualifications but no one had a higher education degree. See Tables 1, 2, 3 for participant characteristics.\nFocus Group 1 participant characteristics\n\nNote: Four males, four females, age 46–71 years, from six different health board regions, living in urban areas. Four current smokers and four of whom had quit within the last 2 years.\nFocus Group 2 participant characteristics\n\nNote: Four females, three males, age range 45–62 years, from three different health board regions, living in rural areas, three smokers and four of whom have quit within the last 2 years.\nFocus Group 3 participant characteristics\n\nNote: Three females, seven males, age range 45–68 years, from three different health board regions, living in urban areas, four smokers and six of whom have quit within the last 2 years.\nOur analysis identified three overarching themes in the data: (1) Knowledge, awareness and acceptability of lung screening, (2) Barriers and facilitators to screening and (3) Promoting screening and implementation ideas.\n[SUBTITLE] Knowledge, awareness and acceptability of lung screening [SUBSECTION] There is currently no national lung screening programme in the United Kingdom, although parts of NHS England are offering lung screening with LDCT.\n25\n Participants were largely unaware of the concept of targeted lung screening:No, I always thought that was something that happened if you develop, you know, or if they suspect you develop then you would have a check, otherwise nothing pre‐emptive…. (FG1, R5)\n\nNo, I always thought that was something that happened if you develop, you know, or if they suspect you develop then you would have a check, otherwise nothing pre‐emptive…. (FG1, R5)\nMost participants had heard of and participated in other forms of screening, including for breast, cervical and bowel cancer, and a few had been referred for lung checks for other reasons. They also described family members having had cancers picked up in this way, as well as their own experience of cancer:For me, it really is, it's very important to be screened, especially breast, bowel, anything. I suffered myself from throat cancer ten years ago and I've been in remission for the past four years, so it's urgently important that people get this done, yeah. (FG3, R4)\n\nFor me, it really is, it's very important to be screened, especially breast, bowel, anything. I suffered myself from throat cancer ten years ago and I've been in remission for the past four years, so it's urgently important that people get this done, yeah. (FG3, R4)\nWhen we described what lung screening would entail, participants were very supportive of this form of screening being available and welcomed the chance to have their lungs screened, with multiple participants saying it was ‘a great idea’. Participants were aware of the benefits of early (asymptomatic) detection and treatment:I think the screening is a good idea to catch things earlier or to see if somebody's got the disease or whatever that they didn't know they had. (FG1, R7)\n\nI think the screening is a good idea to catch things earlier or to see if somebody's got the disease or whatever that they didn't know they had. (FG1, R7)\nParticipants also talked about the importance of screening early and not waiting until the ‘damage was done’, with discussions around age and smoking. It was suggested that raising awareness about lung cancer and lung screening should be introduced to school children, embedding knowledge of screening from an early age. Participants were also largely accepting of the fact that lung screening could pick up other issues,R3: Anything that shows up as a side‐line to it is a benefit and I think most people would welcome it.R1: Yeah, I 100 per cent agree with that.R4: Yeah, totally agree with that one as well, an added bonus. (FG3, R1, 3 and 4)\n\nR3: Anything that shows up as a side‐line to it is a benefit and I think most people would welcome it.\nR1: Yeah, I 100 per cent agree with that.\nR4: Yeah, totally agree with that one as well, an added bonus. (FG3, R1, 3 and 4)\nHowever, there were certain caveats and conditions to participating in screening. The concept of targeted screening for smokers was problematic for participants, as they discussed other risk factors for lung cancer apart from smoking and that some people who never smoke go on to develop lung cancer:There's other causes of lung cancer, it's not just smoking. (FG1, R7)\n\nThere's other causes of lung cancer, it's not just smoking. (FG1, R7)\nParticipants introduced the potentially judgemental and stigmatizing nature of risk‐related eligibility to discussions. The importance of personal informed decision‐making and lack of coercion were also voiced by participants.\nFacilitators asked participants about the role of smoking cessation advice in the lung screening process. Participants suggested that this would not put them off participating, although they reported a dislike of being pushed into stopping smoking or judged for their smoking, such that language and tone were important,I think you have to have that balance … for people to [not] think, ‘oh, we're going there and we're going to have that shoved down our throat [i.e., “forced on us”]’. It has to be choice, but I think always giving people the appropriate choices if somebody is ready to stop, […] and giving them the information being there available, but I don't think making it part of something, because what would then happen is people would then think they're getting this shoved down our throat, we're getting judged for smoking, […] So I think, yes, it's good to have all the information, but it's how it's given. (FG1, R6)\n\nI think you have to have that balance … for people to [not] think, ‘oh, we're going there and we're going to have that shoved down our throat [i.e., “forced on us”]’. It has to be choice, but I think always giving people the appropriate choices if somebody is ready to stop, […] and giving them the information being there available, but I don't think making it part of something, because what would then happen is people would then think they're getting this shoved down our throat, we're getting judged for smoking, […] So I think, yes, it's good to have all the information, but it's how it's given. (FG1, R6)\nParticipants said that most people had received smoking cessation advice before, they already knew that smoking was harmful, and would not be offended by a health professional asking them about smoking. There was a strong sense among participants that the desire to quit smoking and action to quit was self‐motivated.\nThere is currently no national lung screening programme in the United Kingdom, although parts of NHS England are offering lung screening with LDCT.\n25\n Participants were largely unaware of the concept of targeted lung screening:No, I always thought that was something that happened if you develop, you know, or if they suspect you develop then you would have a check, otherwise nothing pre‐emptive…. (FG1, R5)\n\nNo, I always thought that was something that happened if you develop, you know, or if they suspect you develop then you would have a check, otherwise nothing pre‐emptive…. (FG1, R5)\nMost participants had heard of and participated in other forms of screening, including for breast, cervical and bowel cancer, and a few had been referred for lung checks for other reasons. They also described family members having had cancers picked up in this way, as well as their own experience of cancer:For me, it really is, it's very important to be screened, especially breast, bowel, anything. I suffered myself from throat cancer ten years ago and I've been in remission for the past four years, so it's urgently important that people get this done, yeah. (FG3, R4)\n\nFor me, it really is, it's very important to be screened, especially breast, bowel, anything. I suffered myself from throat cancer ten years ago and I've been in remission for the past four years, so it's urgently important that people get this done, yeah. (FG3, R4)\nWhen we described what lung screening would entail, participants were very supportive of this form of screening being available and welcomed the chance to have their lungs screened, with multiple participants saying it was ‘a great idea’. Participants were aware of the benefits of early (asymptomatic) detection and treatment:I think the screening is a good idea to catch things earlier or to see if somebody's got the disease or whatever that they didn't know they had. (FG1, R7)\n\nI think the screening is a good idea to catch things earlier or to see if somebody's got the disease or whatever that they didn't know they had. (FG1, R7)\nParticipants also talked about the importance of screening early and not waiting until the ‘damage was done’, with discussions around age and smoking. It was suggested that raising awareness about lung cancer and lung screening should be introduced to school children, embedding knowledge of screening from an early age. Participants were also largely accepting of the fact that lung screening could pick up other issues,R3: Anything that shows up as a side‐line to it is a benefit and I think most people would welcome it.R1: Yeah, I 100 per cent agree with that.R4: Yeah, totally agree with that one as well, an added bonus. (FG3, R1, 3 and 4)\n\nR3: Anything that shows up as a side‐line to it is a benefit and I think most people would welcome it.\nR1: Yeah, I 100 per cent agree with that.\nR4: Yeah, totally agree with that one as well, an added bonus. (FG3, R1, 3 and 4)\nHowever, there were certain caveats and conditions to participating in screening. The concept of targeted screening for smokers was problematic for participants, as they discussed other risk factors for lung cancer apart from smoking and that some people who never smoke go on to develop lung cancer:There's other causes of lung cancer, it's not just smoking. (FG1, R7)\n\nThere's other causes of lung cancer, it's not just smoking. (FG1, R7)\nParticipants introduced the potentially judgemental and stigmatizing nature of risk‐related eligibility to discussions. The importance of personal informed decision‐making and lack of coercion were also voiced by participants.\nFacilitators asked participants about the role of smoking cessation advice in the lung screening process. Participants suggested that this would not put them off participating, although they reported a dislike of being pushed into stopping smoking or judged for their smoking, such that language and tone were important,I think you have to have that balance … for people to [not] think, ‘oh, we're going there and we're going to have that shoved down our throat [i.e., “forced on us”]’. It has to be choice, but I think always giving people the appropriate choices if somebody is ready to stop, […] and giving them the information being there available, but I don't think making it part of something, because what would then happen is people would then think they're getting this shoved down our throat, we're getting judged for smoking, […] So I think, yes, it's good to have all the information, but it's how it's given. (FG1, R6)\n\nI think you have to have that balance … for people to [not] think, ‘oh, we're going there and we're going to have that shoved down our throat [i.e., “forced on us”]’. It has to be choice, but I think always giving people the appropriate choices if somebody is ready to stop, […] and giving them the information being there available, but I don't think making it part of something, because what would then happen is people would then think they're getting this shoved down our throat, we're getting judged for smoking, […] So I think, yes, it's good to have all the information, but it's how it's given. (FG1, R6)\nParticipants said that most people had received smoking cessation advice before, they already knew that smoking was harmful, and would not be offended by a health professional asking them about smoking. There was a strong sense among participants that the desire to quit smoking and action to quit was self‐motivated.\n[SUBTITLE] Barriers and facilitators to screening [SUBSECTION] It was clear that while there was strong support for the concept of lung screening with participants overwhelmingly in favour of it, a number of factors were raised which qualified their response—with the prospect of mediating the gap between reported intentions and performed screening behaviours in a real‐world scenario. Some of these same issues were potential motivators to participate in lung screening. Broadly, these can be represented under individual level and practical and system level factors.\n[SUBTITLE] Individual level influences on screening intentions [SUBSECTION] On an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nOn an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\n[SUBTITLE] Practical and system barriers [SUBSECTION] A number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.\nA number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.\nIt was clear that while there was strong support for the concept of lung screening with participants overwhelmingly in favour of it, a number of factors were raised which qualified their response—with the prospect of mediating the gap between reported intentions and performed screening behaviours in a real‐world scenario. Some of these same issues were potential motivators to participate in lung screening. Broadly, these can be represented under individual level and practical and system level factors.\n[SUBTITLE] Individual level influences on screening intentions [SUBSECTION] On an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nOn an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\n[SUBTITLE] Practical and system barriers [SUBSECTION] A number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.\nA number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.\n[SUBTITLE] Promoting screening and implementation ideas [SUBSECTION] There was considerable discussion in each focus group on the acceptability and accessibility of screening, leading to suggestions of ways of promoting and implementing screening to increase participation. Participants' views on accessibility were of particular interest due to their socioeconomic status and geographical diversity. Suggestions related to the cognitive and psychological barriers to screening as well as practical issues.\n[SUBTITLE] Managing fears and expectations [SUBSECTION] To address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\nTo address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n[SUBTITLE] Improving accessibility [SUBSECTION] In addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\nIn addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\nThere was considerable discussion in each focus group on the acceptability and accessibility of screening, leading to suggestions of ways of promoting and implementing screening to increase participation. Participants' views on accessibility were of particular interest due to their socioeconomic status and geographical diversity. Suggestions related to the cognitive and psychological barriers to screening as well as practical issues.\n[SUBTITLE] Managing fears and expectations [SUBSECTION] To address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\nTo address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n[SUBTITLE] Improving accessibility [SUBSECTION] In addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\nIn addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)", "There is currently no national lung screening programme in the United Kingdom, although parts of NHS England are offering lung screening with LDCT.\n25\n Participants were largely unaware of the concept of targeted lung screening:No, I always thought that was something that happened if you develop, you know, or if they suspect you develop then you would have a check, otherwise nothing pre‐emptive…. (FG1, R5)\n\nNo, I always thought that was something that happened if you develop, you know, or if they suspect you develop then you would have a check, otherwise nothing pre‐emptive…. (FG1, R5)\nMost participants had heard of and participated in other forms of screening, including for breast, cervical and bowel cancer, and a few had been referred for lung checks for other reasons. They also described family members having had cancers picked up in this way, as well as their own experience of cancer:For me, it really is, it's very important to be screened, especially breast, bowel, anything. I suffered myself from throat cancer ten years ago and I've been in remission for the past four years, so it's urgently important that people get this done, yeah. (FG3, R4)\n\nFor me, it really is, it's very important to be screened, especially breast, bowel, anything. I suffered myself from throat cancer ten years ago and I've been in remission for the past four years, so it's urgently important that people get this done, yeah. (FG3, R4)\nWhen we described what lung screening would entail, participants were very supportive of this form of screening being available and welcomed the chance to have their lungs screened, with multiple participants saying it was ‘a great idea’. Participants were aware of the benefits of early (asymptomatic) detection and treatment:I think the screening is a good idea to catch things earlier or to see if somebody's got the disease or whatever that they didn't know they had. (FG1, R7)\n\nI think the screening is a good idea to catch things earlier or to see if somebody's got the disease or whatever that they didn't know they had. (FG1, R7)\nParticipants also talked about the importance of screening early and not waiting until the ‘damage was done’, with discussions around age and smoking. It was suggested that raising awareness about lung cancer and lung screening should be introduced to school children, embedding knowledge of screening from an early age. Participants were also largely accepting of the fact that lung screening could pick up other issues,R3: Anything that shows up as a side‐line to it is a benefit and I think most people would welcome it.R1: Yeah, I 100 per cent agree with that.R4: Yeah, totally agree with that one as well, an added bonus. (FG3, R1, 3 and 4)\n\nR3: Anything that shows up as a side‐line to it is a benefit and I think most people would welcome it.\nR1: Yeah, I 100 per cent agree with that.\nR4: Yeah, totally agree with that one as well, an added bonus. (FG3, R1, 3 and 4)\nHowever, there were certain caveats and conditions to participating in screening. The concept of targeted screening for smokers was problematic for participants, as they discussed other risk factors for lung cancer apart from smoking and that some people who never smoke go on to develop lung cancer:There's other causes of lung cancer, it's not just smoking. (FG1, R7)\n\nThere's other causes of lung cancer, it's not just smoking. (FG1, R7)\nParticipants introduced the potentially judgemental and stigmatizing nature of risk‐related eligibility to discussions. The importance of personal informed decision‐making and lack of coercion were also voiced by participants.\nFacilitators asked participants about the role of smoking cessation advice in the lung screening process. Participants suggested that this would not put them off participating, although they reported a dislike of being pushed into stopping smoking or judged for their smoking, such that language and tone were important,I think you have to have that balance … for people to [not] think, ‘oh, we're going there and we're going to have that shoved down our throat [i.e., “forced on us”]’. It has to be choice, but I think always giving people the appropriate choices if somebody is ready to stop, […] and giving them the information being there available, but I don't think making it part of something, because what would then happen is people would then think they're getting this shoved down our throat, we're getting judged for smoking, […] So I think, yes, it's good to have all the information, but it's how it's given. (FG1, R6)\n\nI think you have to have that balance … for people to [not] think, ‘oh, we're going there and we're going to have that shoved down our throat [i.e., “forced on us”]’. It has to be choice, but I think always giving people the appropriate choices if somebody is ready to stop, […] and giving them the information being there available, but I don't think making it part of something, because what would then happen is people would then think they're getting this shoved down our throat, we're getting judged for smoking, […] So I think, yes, it's good to have all the information, but it's how it's given. (FG1, R6)\nParticipants said that most people had received smoking cessation advice before, they already knew that smoking was harmful, and would not be offended by a health professional asking them about smoking. There was a strong sense among participants that the desire to quit smoking and action to quit was self‐motivated.", "It was clear that while there was strong support for the concept of lung screening with participants overwhelmingly in favour of it, a number of factors were raised which qualified their response—with the prospect of mediating the gap between reported intentions and performed screening behaviours in a real‐world scenario. Some of these same issues were potential motivators to participate in lung screening. Broadly, these can be represented under individual level and practical and system level factors.\n[SUBTITLE] Individual level influences on screening intentions [SUBSECTION] On an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nOn an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\n[SUBTITLE] Practical and system barriers [SUBSECTION] A number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.\nA number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.", "On an individual level, there were a number of cognitive, psychological and emotional factors influencing screening intentions.\n[SUBTITLE] Psychological and emotional concerns [SUBSECTION] The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\nThe most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n[SUBTITLE] Perceived risk and fatalism [SUBSECTION] Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\nPerceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n[SUBTITLE] Stigma and judgement as a barrier [SUBSECTION] A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\nA common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n[SUBTITLE] Mistrust of healthcare professionals and services [SUBSECTION] A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).\nA number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).", "The most pronounced psychological and emotional concerns reported were fear and worry about cancer and interactions with health services. Fear of invasive procedures, disruption of their lives, waiting for results and the challenges of facing a cancer diagnosis were off‐putting for a number of people:Maybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)It's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)A lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\n\nMaybe you don't want to know, maybe you don't want to have cancer so it's better, you know, just to kind of blunder on and not find out, so not even to go and to be scared of going. Also if you find out you have something wrong then you're going to have to change your lifestyle to make things better. (FG1, R5)\nIt's not knowing if you have the underlying issue or not, and then having to wait and then find out. I think that's maybe what puts a lot of people off not actually doing it. I think they're just maybe prepared, until they get a scare themselves and then go through a test, they're willing to just bypass it. (FG2, R5)\nA lot of people are scared to come forward in case that the results of a test are positive. A lot of people don't want to know. And while they feel okay and there are no symptoms, that's fine. And then for someone to say to them, oh, by the way, you have this or that, it's quite scary for some people. (FG3, R3)\nWhile participants (such as respondent 3 above) spoke of fear of the unknown, they were motivated to see what damage had been done to their lungs, showing the complexity of these thoughts in influencing behaviour,I wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\n\nI wonder what stage my lungs are really at? And I'm sure other people think like that as well. […] It's like they know but we don't know and I think we should know. (FG3, R3)\nOther issues raised in the focus groups related to problems engaging the older generation and males in particular, who they suggested were often stoical in their approach to health and illness and reluctant to burden health services—or they may see no point in screening when they are able to continue functioning. For some participants, the intention to be screened was related to being conscientious citizens and prioritizing one's health above other competing demands:You know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)\n\nYou know, most people are concerned for their own health, they want to be healthy, they don't want to be a burden to the doctor or the NHS. But if you've got something wrong, early intervention is the answer. (FG3, R2)", "Perceived risk of lung cancer also appeared to influence people's screening intentions. Many people felt that their relatively young age and lack of symptoms meant that they were unlikely to have lung cancer and so screening was not relevant to them, though this was tied up with fear and a sense of fatalism in a complex way:I'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)I feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\n\nI'm fit and I'm healthy. If I go and get it and it comes back it's chronic or it's terminal, fine, it's only terminal for as long as I'm going to last. (FG3, R4)\nI feel myself slightly younger than everyone here that I see screening as a slightly older person's thing or a female thing, and you don't have to worry about it until you're like a certain age or something like that. (FG1, R5)\nPerceived risk was also associated with family history and advancing age. Bad experiences of cancer in the family were discussed as both a source of avoidance of screening and a motivator to take part to avoid late detection and a poor prognosis:Yeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\n\nYeah, I've been for a cervical one because unfortunately on my mum's side of the family it seems to run in the family. So, my mum and my older sister have all had hysterectomies at a young age … You have to go for these screenings, especially if you find it runs in your family. Even if it doesn't run in your family, the older we get, we are getting older and it's all inside as well so it's good to know what's going on. (FG2, R5)\nAs above, fatalism was also evident in discussions among those who had smoked heavily and felt that lung cancer was unavoidable:At the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)\n\nAt the end of the day, I've smoked since I was 15, so what damage is done [, is done]. (FG3, R4)", "A common theme across all focus groups was the role of judgement of smoking behaviour and perceived stigma as a barrier to presenting for lung screening:The stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\n\nThe stigma of people who smoke is very real and they're made to feel uncomfortable. Although for years and years we were encouraged to do it, it was modern, it was yuppy, it was everything you wanted. It was cool, sophisticated, and yet now smokers feel … Well, I don't know, I'm only speaking for myself, I feel as if, oh well, it's your own fault, you caused it. I did cause it, but I was encouraged to cause it. (FG3, R4)\nParticipants were also conscious of the cost of screening and did not want to be seen to take advantage of the system:No, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)\n\nNo, but you could get members of the public being quite judgemental because I smoke so I know the health concerns, so if I'm choosing to do a risk‐taking behaviour, shall we say, I'm presuming this is something that the taxpayer is going to pay, so I'm thinking somebody might think why should I get tested for something that I'm putting myself at risk for. That might put people off. (FG1, R6)", "A number of participants reported good experiences with health services and a proactive approach to their health, driven by an awareness of the benefits of early detection. However, poor experiences with health services and healthcare professionals compromized trust and limited faith in services for some participants, and therefore an avoidance of any kind of interaction with them:I wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\n\nI wouldn't trust my GP in that case to recommend, you know? Maybe he's biased, he's thinking, okay, that guy's smoking so he's just wasting time and money anyway so I won't recommend him. (FG1, R5)\nParticipants in two of the groups discussed the fact that GPs are often overworked and a perception that patients' smoking results in GPs not investigating issues adequately or treating them fairly. By‐passing GPs and attending screening through an independent screening programme was seen as a positive thing. On the other hand, some participants reported good experiences and welcomed an endorsement from their GP, with one person commenting, ‘It's formal coming from your doctor’ (FG1, R5).", "A number of practical barriers to attending for screening were raised by participants. These included time off from work leading to loss of money, distance of travel to an appointment and access issues:If you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\n\nIf you got a letter in saying, oh, you've got to go at ten o'clock in the morning and it's hardly worth going to work before that because you have to travel or whatever, so you might end up losing four hours' pay, you know what I mean? So if there's an incentive to encourage people for them not to lose money, I think you would get over 90 per cent of people would do it. (FG1, R3)\nLiving in remote areas of Scotland, while having local access to primary care, was also voiced as an issue in terms of accessing secondary care, which could be a problem for a hospital‐based lung screening:We're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\n\nWe're lucky in [place] that we've got a hospital and it covers the whole of the north but there are lots of places that … and there are a lot of old people that can't get to it. (FG1, R2)\nParticipants also had some concerns about the ability of health services to meet the demands of screening in terms of providing an accessible and timely service. Delays and waiting for test results were a source of worry for people and there was a common reported perception of the NHS as an under‐resourced and over‐stretched service.\nHaving outlined what the screening process would entail, participants did not voluntarily raise any concerns about radiation exposure, over‐diagnosis or invasiveness of the LDCT procedure. Participants were supportive of a ‘proper’ check of the lungs, including identification of nodules to be monitored or other incidental findings, particularly when they had existing lung problems.", "There was considerable discussion in each focus group on the acceptability and accessibility of screening, leading to suggestions of ways of promoting and implementing screening to increase participation. Participants' views on accessibility were of particular interest due to their socioeconomic status and geographical diversity. Suggestions related to the cognitive and psychological barriers to screening as well as practical issues.\n[SUBTITLE] Managing fears and expectations [SUBSECTION] To address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\nTo address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n[SUBTITLE] Improving accessibility [SUBSECTION] In addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\nIn addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)", "To address fears around lung cancer, participants suggest that there is an emphasis in any information materials or advertising campaigns on positive messaging. Examples were given of being able to live to play with grandchildren and harnessing the successes of other screening programmes:I think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\n\nI think to get people to go to screening, you need to publicise the success of other screening programmes, whether it's bowel, breast, cervical, whatever it is. Publicise how successful these are and really go for it and say, right, this is the next step in the screening and it's going to be a lung screening. (FG3, R1)\nParticipants said that an invitation to screening should be encouraging without being coercive, should not mention the word ‘cancer’ too much and should not imply judgement about smoking behaviour:There's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)\n\nThere's always going to be a worry anyway. But seeing the word ‘health check’ you would get more people to go. The word cancer and people just say, well I don't want to know. (FG2, R3)", "In addition to the implementation ideas given above, there was a very clear message from the focus groups when it came to addressing any access barriers to screening, relevant to both deprived and remote and rural communities—the use of mobile units. This, it was typically felt, would remove screening from a clinical environment, and bring it closer to local communities.So, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)This is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\n\nSo, if there was screening available as a mobile unit or some sort of drop in people probably would be more willing to go and fit it in. It's just the same as these COVID jabs we've all had to do; if they were more available, a lot more people would do it. (FG2, R3)\nThis is where the mobile vans come in, you know? They can drive to these remote areas, especially in Scotland, when they go further north. We've got two people from [place], […] some of the places are very remote so…. (FG1, R1)\nWhile those in the rural‐dwelling focus group were unanimously in favour of mobile units, support for them was not confined to the rural group. Mobile units were supported as a way of engaging with marginalized groups, such as those who sought to avoid hospitals.\nSupporting people who would lose pay if they attend a screening appointment was suggested as a statutory right, with employer support:I think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\n\nI think your employer should get help if people need time off to go to these things, they should be encouraging and get paid for going to them, you know, get paid for so many hours, if you need so many hours off to go and get these tests done, you shouldn't lose your pay, because if you lose your pay, a lot of people will not go. (FG1, R3)\nAnother suggestion for increasing participation was to ‘flood’ the public with information about screening through all media outlets about screening and engage with local communities, particularly the harder to reach groups, through ‘men's sheds’, workplaces, community hubs for older people as well as speaking to school pupils to normalize screening:And taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)\n\nAnd taking the fear out of it by having all different age groups talking about it … It's trying to get the message out there, people need to start screening themselves from an age where they're invited to do so, and not be fearful of it. It's trying to get around that age group and succeeding with it, and hoping that future generations do go for their screenings when they're invited to do so. (FG2, R6)", "[SUBTITLE] Summary [SUBSECTION] This focus group study is the first of its kind to ascertain the views of Scottish residents on a potential LDCT lung screening programme and identify likely barriers and facilitators in this context. Knowledge of lung screening was low among the focus group participants, although they showed awareness and personal experience of other screening programmes. Participants were very supportive of the idea of lung screening and harnessed the early detection narrative to discuss the importance of taking part. Participants reported the process of lung screening via a LDCT scan was acceptable. Two key barrier types were identified: individual level influences on screening intentions, and practical and system barriers. Within the individual level factors, emotional and psychological concerns related to fear of a cancer diagnosis, mistrust, fatalism and perceived stigma were dominant in focus group discussion. For some, screening was part of being a health conscious citizen and prioritizing health matters, while others based their decision‐making on their perceived risk. A number of potential practical barriers to lung screening participation were mooted that were particularly relevant for people living in deprived and rural communities. These included travel, cost, time and competing priorities. Maximizing accessibility was also key in the discussion and a distinct recommendation for future programmes.\nThis focus group study is the first of its kind to ascertain the views of Scottish residents on a potential LDCT lung screening programme and identify likely barriers and facilitators in this context. Knowledge of lung screening was low among the focus group participants, although they showed awareness and personal experience of other screening programmes. Participants were very supportive of the idea of lung screening and harnessed the early detection narrative to discuss the importance of taking part. Participants reported the process of lung screening via a LDCT scan was acceptable. Two key barrier types were identified: individual level influences on screening intentions, and practical and system barriers. Within the individual level factors, emotional and psychological concerns related to fear of a cancer diagnosis, mistrust, fatalism and perceived stigma were dominant in focus group discussion. For some, screening was part of being a health conscious citizen and prioritizing health matters, while others based their decision‐making on their perceived risk. A number of potential practical barriers to lung screening participation were mooted that were particularly relevant for people living in deprived and rural communities. These included travel, cost, time and competing priorities. Maximizing accessibility was also key in the discussion and a distinct recommendation for future programmes.\n[SUBTITLE] Comparison with existing literature and theory [SUBSECTION] There is a strong consistency in our findings with the growing body of evidence looking at attitudes to screening and screening behaviour, across a range of screening programmes and for lung screening in particular, in the United Kingdom and beyond.\n26\n, \n27\n, \n28\n LDCT screening for lung cancer is largely unheard of in Scotland but has a high degree of acceptability more broadly, or among those who have participated in screening.\n27\n, \n29\n, \n30\n There was an evident awareness among participants of the benefits of early detection and thus support for lung screening, in line with other research.\n17\n, \n31\n\n\nFear was identified as one of the most common psychological barriers to lung screening, which is reflected in the literature in both survey and qualitative explorations of attitudes to screening.\n28\n, \n32\n, \n33\n Linked to this was a sense of fatalism or predicted fatalism among older generations, also mirrored in comparable studies of lung screening.\n29\n, \n34\n While there is evidence to suggest that the fear and anxiety associated with lung screening participation is transient\n35\n and can be a motivating factor to be screened, quit smoking and even other cancer‐preventing health behaviour change,\n36\n, \n37\n, \n38\n it is still vital to minimize this emotional response by addressing and managing it. Positive messaging in the promotion of screening, such as sharing the treatment successes and mortality gains from early detection, is one potential step in approaching this.\nNotions of risk were a key component of our group's considerations of whether or not someone would take part in screening, often related to not experiencing symptoms and having stopped smoking. Perceived risk has been identified widely in the literature to explain decision‐making in relation to screening and help‐seeking for symptomatic illness.\n28\n, \n33\n, \n39\n, \n40\n Risk and decision‐making are discussed further below.\nPerceived judgement and stigma related to smoking featured in focus group discussions. This is widely evident in the literature, along with self‐blame.\n31\n, \n38\n, \n41\n Stigma has been identified in the literature as a barrier to help‐seeking for signs of lung cancer and it seems this also applies to screening participation.\n42\n Related to this was a sense of fatalism—participants in our study did not consider they would blame themselves if they developed lung cancer, but some did feel that the damage was already done and screening could not change that.\n17\n, \n29\n, \n38\n However, this was not a clear barrier. For some, it was a good reason to detect any inevitable lung cancer at an early stage, suggesting that issues such as fear, blame and fatalism are complex and operate on a pendulum when prompting action or inaction.\nDiscussions of stigma inevitably moved onto smoking cessation. Smoking cessation advice was broadly acceptable to our participants, but only if delivered in a noncoercive way; again, other studies have had similar findings.\n17\n, \n43\n, \n44\n, \n45\n In our focus group study, it appeared that participants may have become desensitized to smoking cessation advice. This has implications when considering brief interventions for smoking, and would need further exploration to understand whether small prompts may be enough to stimulate action among people who have intentions to quit smoking. There is evidence to suggest that brief interventions can be effective.\n46\n, \n47\n There is also emerging evidence that incorporating smoking cessation advice into lung screening is effective—seeing images of lungs has been a strong motivator to quit smoking\n36\n as well as being central to the long‐term cost‐effectiveness of screening.\n13\n\n\nSome participants in our focus groups described a difficult relationship and a level of mistrust in interacting with health professionals and services, often related to poor past experiences or a sociocultural divide. Such perceptions are often ingrained in more deprived communities and are often reported in the literature—relating not only to lung screening,\n17\n, \n32\n but beyond to studies of help‐seeking behaviour and doctor–patient relationships.\n48\n Again, this is a complex feature of health service engagement and can be linked to other barriers such as low self‐efficacy, low health literacy and power dynamics, which can be particularly divisive in terms of equity in access to health care for people in disadvantaged groups.\n48\n, \n49\n, \n50\n Development of interventions to repair broken relationships with the health service, a perceived authoritative institution, and other methods to improve accessibility such as communication training for professionals, targeted awareness campaigns and community engagement strategies can address inequities in access to screening services.\n51\n Primary care has an important role to play; current workforce shortages need to be addressed, and solutions identified which do not generate significant extra burden for primary care staff—one example is streamlining procedures to identify high‐risk patients from practice data.\n7\n, \n8\n It also seems logical to harness the successes of implementation strategies for other cancer screening (e.g., the UK's bowel screening programmes).\n52\n, \n53\n\n\nIn addition to the psychosocial issues discussed by focus group participants, practical barriers to participation in lung screening including time, cost, travel and competing work or other commitments, were also mooted. Practical barriers are commonly reported throughout the evidence base related to engaging with screening, with a suggestion that these barriers are heightened in more deprived groups.\n27\n, \n32\n, \n46\n\n\nVon Wagner et al.\n50\n have developed a model of screening behaviour, accounting for a range of factors identified in relation to wider health behaviours, such as health identity and self‐efficacy, that can be usefully mapped onto the findings of this study. Similarly, Robb\n54\n has developed the I‐SAM model to understand screening participation. Application of these models to follow‐up interviews as part of the planned pilot lung screening study in Scotland will be enlightening to confirm the salience of these to screening participation and nonparticipation.\nThere is an abundance of early cancer detection research exploring how people appraise bodily changes, evaluate risk and decide to seek medical help, as well as conceptual models to understand these processes.\n55\n, \n56\n, \n57\n There is also some utility in applying these to screening behaviour, often in the absence of symptoms, to understand nonparticipation in screening. For example, Kummer et al.'s\n37\n, \n39\n cognitive heuristics for help‐seeking for cancer symptoms may act as prompts or inhibitors to participate in screening. Understanding these factors in the context of deprived populations adds further considerations that may compound behaviour in terms of available resources (cognitive, psychological and practical), competing demands and permeability of services.\n48\n, \n49\n\n\nIn terms of ideas to overcome barriers, participants focused primarily on positive messaging in information materials and advertising, and accessibility through the provision of mobile screening vans similar to those used in breast screening or for Covid‐19 vaccination. Positive and nonjudgemental messaging focusing on the gains on offer from screening and early detection has also been found elsewhere and incorporated into the pilot lung screening provision.\n58\n, \n59\n Travel to access screening services as well as fear of hospital environments have also been identified in the literature, and evidence evaluating the use of mobile screening vans to address these issues is beginning to accumulate.\n12\n, \n60\n\n\nThere is a strong consistency in our findings with the growing body of evidence looking at attitudes to screening and screening behaviour, across a range of screening programmes and for lung screening in particular, in the United Kingdom and beyond.\n26\n, \n27\n, \n28\n LDCT screening for lung cancer is largely unheard of in Scotland but has a high degree of acceptability more broadly, or among those who have participated in screening.\n27\n, \n29\n, \n30\n There was an evident awareness among participants of the benefits of early detection and thus support for lung screening, in line with other research.\n17\n, \n31\n\n\nFear was identified as one of the most common psychological barriers to lung screening, which is reflected in the literature in both survey and qualitative explorations of attitudes to screening.\n28\n, \n32\n, \n33\n Linked to this was a sense of fatalism or predicted fatalism among older generations, also mirrored in comparable studies of lung screening.\n29\n, \n34\n While there is evidence to suggest that the fear and anxiety associated with lung screening participation is transient\n35\n and can be a motivating factor to be screened, quit smoking and even other cancer‐preventing health behaviour change,\n36\n, \n37\n, \n38\n it is still vital to minimize this emotional response by addressing and managing it. Positive messaging in the promotion of screening, such as sharing the treatment successes and mortality gains from early detection, is one potential step in approaching this.\nNotions of risk were a key component of our group's considerations of whether or not someone would take part in screening, often related to not experiencing symptoms and having stopped smoking. Perceived risk has been identified widely in the literature to explain decision‐making in relation to screening and help‐seeking for symptomatic illness.\n28\n, \n33\n, \n39\n, \n40\n Risk and decision‐making are discussed further below.\nPerceived judgement and stigma related to smoking featured in focus group discussions. This is widely evident in the literature, along with self‐blame.\n31\n, \n38\n, \n41\n Stigma has been identified in the literature as a barrier to help‐seeking for signs of lung cancer and it seems this also applies to screening participation.\n42\n Related to this was a sense of fatalism—participants in our study did not consider they would blame themselves if they developed lung cancer, but some did feel that the damage was already done and screening could not change that.\n17\n, \n29\n, \n38\n However, this was not a clear barrier. For some, it was a good reason to detect any inevitable lung cancer at an early stage, suggesting that issues such as fear, blame and fatalism are complex and operate on a pendulum when prompting action or inaction.\nDiscussions of stigma inevitably moved onto smoking cessation. Smoking cessation advice was broadly acceptable to our participants, but only if delivered in a noncoercive way; again, other studies have had similar findings.\n17\n, \n43\n, \n44\n, \n45\n In our focus group study, it appeared that participants may have become desensitized to smoking cessation advice. This has implications when considering brief interventions for smoking, and would need further exploration to understand whether small prompts may be enough to stimulate action among people who have intentions to quit smoking. There is evidence to suggest that brief interventions can be effective.\n46\n, \n47\n There is also emerging evidence that incorporating smoking cessation advice into lung screening is effective—seeing images of lungs has been a strong motivator to quit smoking\n36\n as well as being central to the long‐term cost‐effectiveness of screening.\n13\n\n\nSome participants in our focus groups described a difficult relationship and a level of mistrust in interacting with health professionals and services, often related to poor past experiences or a sociocultural divide. Such perceptions are often ingrained in more deprived communities and are often reported in the literature—relating not only to lung screening,\n17\n, \n32\n but beyond to studies of help‐seeking behaviour and doctor–patient relationships.\n48\n Again, this is a complex feature of health service engagement and can be linked to other barriers such as low self‐efficacy, low health literacy and power dynamics, which can be particularly divisive in terms of equity in access to health care for people in disadvantaged groups.\n48\n, \n49\n, \n50\n Development of interventions to repair broken relationships with the health service, a perceived authoritative institution, and other methods to improve accessibility such as communication training for professionals, targeted awareness campaigns and community engagement strategies can address inequities in access to screening services.\n51\n Primary care has an important role to play; current workforce shortages need to be addressed, and solutions identified which do not generate significant extra burden for primary care staff—one example is streamlining procedures to identify high‐risk patients from practice data.\n7\n, \n8\n It also seems logical to harness the successes of implementation strategies for other cancer screening (e.g., the UK's bowel screening programmes).\n52\n, \n53\n\n\nIn addition to the psychosocial issues discussed by focus group participants, practical barriers to participation in lung screening including time, cost, travel and competing work or other commitments, were also mooted. Practical barriers are commonly reported throughout the evidence base related to engaging with screening, with a suggestion that these barriers are heightened in more deprived groups.\n27\n, \n32\n, \n46\n\n\nVon Wagner et al.\n50\n have developed a model of screening behaviour, accounting for a range of factors identified in relation to wider health behaviours, such as health identity and self‐efficacy, that can be usefully mapped onto the findings of this study. Similarly, Robb\n54\n has developed the I‐SAM model to understand screening participation. Application of these models to follow‐up interviews as part of the planned pilot lung screening study in Scotland will be enlightening to confirm the salience of these to screening participation and nonparticipation.\nThere is an abundance of early cancer detection research exploring how people appraise bodily changes, evaluate risk and decide to seek medical help, as well as conceptual models to understand these processes.\n55\n, \n56\n, \n57\n There is also some utility in applying these to screening behaviour, often in the absence of symptoms, to understand nonparticipation in screening. For example, Kummer et al.'s\n37\n, \n39\n cognitive heuristics for help‐seeking for cancer symptoms may act as prompts or inhibitors to participate in screening. Understanding these factors in the context of deprived populations adds further considerations that may compound behaviour in terms of available resources (cognitive, psychological and practical), competing demands and permeability of services.\n48\n, \n49\n\n\nIn terms of ideas to overcome barriers, participants focused primarily on positive messaging in information materials and advertising, and accessibility through the provision of mobile screening vans similar to those used in breast screening or for Covid‐19 vaccination. Positive and nonjudgemental messaging focusing on the gains on offer from screening and early detection has also been found elsewhere and incorporated into the pilot lung screening provision.\n58\n, \n59\n Travel to access screening services as well as fear of hospital environments have also been identified in the literature, and evidence evaluating the use of mobile screening vans to address these issues is beginning to accumulate.\n12\n, \n60\n\n\n[SUBTITLE] Strengths and limitations [SUBSECTION] This set of focus groups provides a rich data set with an in‐depth discussion of the concept of lung cancer screening and anticipated barriers and facilitators. Focus group participants were a self‐selecting group who are accustomed to taking part in research studies, and who received a financial incentive. However, this form of recruitment strategy does allow access to people from a range of different backgrounds, including both deprived and rural areas of Scotland that may otherwise have been challenging to recruit. Three of the 24 participants were from ethnic minority groups, reflecting the general demographic in the Scottish population. Focus groups were conducted online. Video conferencing software facilitates easy interaction with people from around the country but can pose challenges. These include failing technology, building rapport and ensuring balanced participation within the group simply by being in a room together and acknowledging social cues. It was also necessary to consider that participants were talking about a hypothetical scenario and their intended behaviour, something we know does not simply translate into action.\n61\n Speaking to people who have taken part in screening or chosen not to take part is also essential to further understand the relationship between intention and behaviour.\nWe also reflected on the group dynamic together with the nature of our role in conducting the focus groups, and whether this was likely to influence people in agreeing with and supporting the concept of lung screening. However, the open nature of questioning, reminding participants that we genuinely wanted to hear their views, and the self‐selecting group of individuals who were quite assured in their own responses, suggested that we did not shape this narrative.\nThis set of focus groups provides a rich data set with an in‐depth discussion of the concept of lung cancer screening and anticipated barriers and facilitators. Focus group participants were a self‐selecting group who are accustomed to taking part in research studies, and who received a financial incentive. However, this form of recruitment strategy does allow access to people from a range of different backgrounds, including both deprived and rural areas of Scotland that may otherwise have been challenging to recruit. Three of the 24 participants were from ethnic minority groups, reflecting the general demographic in the Scottish population. Focus groups were conducted online. Video conferencing software facilitates easy interaction with people from around the country but can pose challenges. These include failing technology, building rapport and ensuring balanced participation within the group simply by being in a room together and acknowledging social cues. It was also necessary to consider that participants were talking about a hypothetical scenario and their intended behaviour, something we know does not simply translate into action.\n61\n Speaking to people who have taken part in screening or chosen not to take part is also essential to further understand the relationship between intention and behaviour.\nWe also reflected on the group dynamic together with the nature of our role in conducting the focus groups, and whether this was likely to influence people in agreeing with and supporting the concept of lung screening. However, the open nature of questioning, reminding participants that we genuinely wanted to hear their views, and the self‐selecting group of individuals who were quite assured in their own responses, suggested that we did not shape this narrative.\n[SUBTITLE] Implications and future work [SUBSECTION] This study informs the development of strategies to improve uptake and informed choice in lung screening. It is essential to understand people's health beliefs and behaviours and to target the barriers to implement a patient‐centred service using a theory‐driven approach.\n62\n This work adds to a growing evidence base shedding light on the behavioural aspects of screening participation and will inform the design and implementation of a new lung screening pilot in Scotland\n40\n, \n45\n, \n46\n (see Box 2 for implementation ideas generated from this work). Minimizing practical barriers is also likely to be instrumental in improving participation and addressing inequity in access to screening. As such, information materials, methods of communication and the design of the process involved in screening (e.g., minimizing the steps, time commitment and waiting intervals), and sensitive, supportive messaging that addresses stigma and fear are all important components of a pilot to break down some of the known barriers. Drawing on the similarities with research based on other UK pilot studies, we are modelling optimized study and participant materials.\n59\n, \n63\n, \n64\n While there is consistency in the findings compared with existing work, it was important to explore the Scottish context with a diverse sample of participants to consider how rurality and deprivation presented any unique issues. Understanding ethnic variations in views on lung screening participation will also be important to ensure equitable provision and uptake of screening. A high burden of multimorbidity is also characteristic of the Scottish population and should be examined in future studies.\n\nMinimize steps in the screening process to lower opportunities for delays and associated distressAvoiding unnecessary travel to scanning facilities with the chance to discuss screening concerns, address fears and perceived stigma, and facilitate informed decision‐makingUse of mobile screening vans as a ‘one stop shop’ to address resource constraints and travel issues for people living in deprived and rural areasConsider covering the cost of travel expenses to screening facilities to ensure equitable accessIncorporating discussion of fears associated with screening into information materialsEnsuring positive messaging with nonjudgemental language around smoking behaviourEnsuring a timely and sensitive approach to smoking cessation advice\n\nMinimize steps in the screening process to lower opportunities for delays and associated distress\nAvoiding unnecessary travel to scanning facilities with the chance to discuss screening concerns, address fears and perceived stigma, and facilitate informed decision‐making\nUse of mobile screening vans as a ‘one stop shop’ to address resource constraints and travel issues for people living in deprived and rural areas\nConsider covering the cost of travel expenses to screening facilities to ensure equitable access\nIncorporating discussion of fears associated with screening into information materials\nEnsuring positive messaging with nonjudgemental language around smoking behaviour\nEnsuring a timely and sensitive approach to smoking cessation advice\nThis study informs the development of strategies to improve uptake and informed choice in lung screening. It is essential to understand people's health beliefs and behaviours and to target the barriers to implement a patient‐centred service using a theory‐driven approach.\n62\n This work adds to a growing evidence base shedding light on the behavioural aspects of screening participation and will inform the design and implementation of a new lung screening pilot in Scotland\n40\n, \n45\n, \n46\n (see Box 2 for implementation ideas generated from this work). Minimizing practical barriers is also likely to be instrumental in improving participation and addressing inequity in access to screening. As such, information materials, methods of communication and the design of the process involved in screening (e.g., minimizing the steps, time commitment and waiting intervals), and sensitive, supportive messaging that addresses stigma and fear are all important components of a pilot to break down some of the known barriers. Drawing on the similarities with research based on other UK pilot studies, we are modelling optimized study and participant materials.\n59\n, \n63\n, \n64\n While there is consistency in the findings compared with existing work, it was important to explore the Scottish context with a diverse sample of participants to consider how rurality and deprivation presented any unique issues. Understanding ethnic variations in views on lung screening participation will also be important to ensure equitable provision and uptake of screening. A high burden of multimorbidity is also characteristic of the Scottish population and should be examined in future studies.\n\nMinimize steps in the screening process to lower opportunities for delays and associated distressAvoiding unnecessary travel to scanning facilities with the chance to discuss screening concerns, address fears and perceived stigma, and facilitate informed decision‐makingUse of mobile screening vans as a ‘one stop shop’ to address resource constraints and travel issues for people living in deprived and rural areasConsider covering the cost of travel expenses to screening facilities to ensure equitable accessIncorporating discussion of fears associated with screening into information materialsEnsuring positive messaging with nonjudgemental language around smoking behaviourEnsuring a timely and sensitive approach to smoking cessation advice\n\nMinimize steps in the screening process to lower opportunities for delays and associated distress\nAvoiding unnecessary travel to scanning facilities with the chance to discuss screening concerns, address fears and perceived stigma, and facilitate informed decision‐making\nUse of mobile screening vans as a ‘one stop shop’ to address resource constraints and travel issues for people living in deprived and rural areas\nConsider covering the cost of travel expenses to screening facilities to ensure equitable access\nIncorporating discussion of fears associated with screening into information materials\nEnsuring positive messaging with nonjudgemental language around smoking behaviour\nEnsuring a timely and sensitive approach to smoking cessation advice", "This focus group study is the first of its kind to ascertain the views of Scottish residents on a potential LDCT lung screening programme and identify likely barriers and facilitators in this context. Knowledge of lung screening was low among the focus group participants, although they showed awareness and personal experience of other screening programmes. Participants were very supportive of the idea of lung screening and harnessed the early detection narrative to discuss the importance of taking part. Participants reported the process of lung screening via a LDCT scan was acceptable. Two key barrier types were identified: individual level influences on screening intentions, and practical and system barriers. Within the individual level factors, emotional and psychological concerns related to fear of a cancer diagnosis, mistrust, fatalism and perceived stigma were dominant in focus group discussion. For some, screening was part of being a health conscious citizen and prioritizing health matters, while others based their decision‐making on their perceived risk. A number of potential practical barriers to lung screening participation were mooted that were particularly relevant for people living in deprived and rural communities. These included travel, cost, time and competing priorities. Maximizing accessibility was also key in the discussion and a distinct recommendation for future programmes.", "There is a strong consistency in our findings with the growing body of evidence looking at attitudes to screening and screening behaviour, across a range of screening programmes and for lung screening in particular, in the United Kingdom and beyond.\n26\n, \n27\n, \n28\n LDCT screening for lung cancer is largely unheard of in Scotland but has a high degree of acceptability more broadly, or among those who have participated in screening.\n27\n, \n29\n, \n30\n There was an evident awareness among participants of the benefits of early detection and thus support for lung screening, in line with other research.\n17\n, \n31\n\n\nFear was identified as one of the most common psychological barriers to lung screening, which is reflected in the literature in both survey and qualitative explorations of attitudes to screening.\n28\n, \n32\n, \n33\n Linked to this was a sense of fatalism or predicted fatalism among older generations, also mirrored in comparable studies of lung screening.\n29\n, \n34\n While there is evidence to suggest that the fear and anxiety associated with lung screening participation is transient\n35\n and can be a motivating factor to be screened, quit smoking and even other cancer‐preventing health behaviour change,\n36\n, \n37\n, \n38\n it is still vital to minimize this emotional response by addressing and managing it. Positive messaging in the promotion of screening, such as sharing the treatment successes and mortality gains from early detection, is one potential step in approaching this.\nNotions of risk were a key component of our group's considerations of whether or not someone would take part in screening, often related to not experiencing symptoms and having stopped smoking. Perceived risk has been identified widely in the literature to explain decision‐making in relation to screening and help‐seeking for symptomatic illness.\n28\n, \n33\n, \n39\n, \n40\n Risk and decision‐making are discussed further below.\nPerceived judgement and stigma related to smoking featured in focus group discussions. This is widely evident in the literature, along with self‐blame.\n31\n, \n38\n, \n41\n Stigma has been identified in the literature as a barrier to help‐seeking for signs of lung cancer and it seems this also applies to screening participation.\n42\n Related to this was a sense of fatalism—participants in our study did not consider they would blame themselves if they developed lung cancer, but some did feel that the damage was already done and screening could not change that.\n17\n, \n29\n, \n38\n However, this was not a clear barrier. For some, it was a good reason to detect any inevitable lung cancer at an early stage, suggesting that issues such as fear, blame and fatalism are complex and operate on a pendulum when prompting action or inaction.\nDiscussions of stigma inevitably moved onto smoking cessation. Smoking cessation advice was broadly acceptable to our participants, but only if delivered in a noncoercive way; again, other studies have had similar findings.\n17\n, \n43\n, \n44\n, \n45\n In our focus group study, it appeared that participants may have become desensitized to smoking cessation advice. This has implications when considering brief interventions for smoking, and would need further exploration to understand whether small prompts may be enough to stimulate action among people who have intentions to quit smoking. There is evidence to suggest that brief interventions can be effective.\n46\n, \n47\n There is also emerging evidence that incorporating smoking cessation advice into lung screening is effective—seeing images of lungs has been a strong motivator to quit smoking\n36\n as well as being central to the long‐term cost‐effectiveness of screening.\n13\n\n\nSome participants in our focus groups described a difficult relationship and a level of mistrust in interacting with health professionals and services, often related to poor past experiences or a sociocultural divide. Such perceptions are often ingrained in more deprived communities and are often reported in the literature—relating not only to lung screening,\n17\n, \n32\n but beyond to studies of help‐seeking behaviour and doctor–patient relationships.\n48\n Again, this is a complex feature of health service engagement and can be linked to other barriers such as low self‐efficacy, low health literacy and power dynamics, which can be particularly divisive in terms of equity in access to health care for people in disadvantaged groups.\n48\n, \n49\n, \n50\n Development of interventions to repair broken relationships with the health service, a perceived authoritative institution, and other methods to improve accessibility such as communication training for professionals, targeted awareness campaigns and community engagement strategies can address inequities in access to screening services.\n51\n Primary care has an important role to play; current workforce shortages need to be addressed, and solutions identified which do not generate significant extra burden for primary care staff—one example is streamlining procedures to identify high‐risk patients from practice data.\n7\n, \n8\n It also seems logical to harness the successes of implementation strategies for other cancer screening (e.g., the UK's bowel screening programmes).\n52\n, \n53\n\n\nIn addition to the psychosocial issues discussed by focus group participants, practical barriers to participation in lung screening including time, cost, travel and competing work or other commitments, were also mooted. Practical barriers are commonly reported throughout the evidence base related to engaging with screening, with a suggestion that these barriers are heightened in more deprived groups.\n27\n, \n32\n, \n46\n\n\nVon Wagner et al.\n50\n have developed a model of screening behaviour, accounting for a range of factors identified in relation to wider health behaviours, such as health identity and self‐efficacy, that can be usefully mapped onto the findings of this study. Similarly, Robb\n54\n has developed the I‐SAM model to understand screening participation. Application of these models to follow‐up interviews as part of the planned pilot lung screening study in Scotland will be enlightening to confirm the salience of these to screening participation and nonparticipation.\nThere is an abundance of early cancer detection research exploring how people appraise bodily changes, evaluate risk and decide to seek medical help, as well as conceptual models to understand these processes.\n55\n, \n56\n, \n57\n There is also some utility in applying these to screening behaviour, often in the absence of symptoms, to understand nonparticipation in screening. For example, Kummer et al.'s\n37\n, \n39\n cognitive heuristics for help‐seeking for cancer symptoms may act as prompts or inhibitors to participate in screening. Understanding these factors in the context of deprived populations adds further considerations that may compound behaviour in terms of available resources (cognitive, psychological and practical), competing demands and permeability of services.\n48\n, \n49\n\n\nIn terms of ideas to overcome barriers, participants focused primarily on positive messaging in information materials and advertising, and accessibility through the provision of mobile screening vans similar to those used in breast screening or for Covid‐19 vaccination. Positive and nonjudgemental messaging focusing on the gains on offer from screening and early detection has also been found elsewhere and incorporated into the pilot lung screening provision.\n58\n, \n59\n Travel to access screening services as well as fear of hospital environments have also been identified in the literature, and evidence evaluating the use of mobile screening vans to address these issues is beginning to accumulate.\n12\n, \n60\n\n", "This set of focus groups provides a rich data set with an in‐depth discussion of the concept of lung cancer screening and anticipated barriers and facilitators. Focus group participants were a self‐selecting group who are accustomed to taking part in research studies, and who received a financial incentive. However, this form of recruitment strategy does allow access to people from a range of different backgrounds, including both deprived and rural areas of Scotland that may otherwise have been challenging to recruit. Three of the 24 participants were from ethnic minority groups, reflecting the general demographic in the Scottish population. Focus groups were conducted online. Video conferencing software facilitates easy interaction with people from around the country but can pose challenges. These include failing technology, building rapport and ensuring balanced participation within the group simply by being in a room together and acknowledging social cues. It was also necessary to consider that participants were talking about a hypothetical scenario and their intended behaviour, something we know does not simply translate into action.\n61\n Speaking to people who have taken part in screening or chosen not to take part is also essential to further understand the relationship between intention and behaviour.\nWe also reflected on the group dynamic together with the nature of our role in conducting the focus groups, and whether this was likely to influence people in agreeing with and supporting the concept of lung screening. However, the open nature of questioning, reminding participants that we genuinely wanted to hear their views, and the self‐selecting group of individuals who were quite assured in their own responses, suggested that we did not shape this narrative.", "This study informs the development of strategies to improve uptake and informed choice in lung screening. It is essential to understand people's health beliefs and behaviours and to target the barriers to implement a patient‐centred service using a theory‐driven approach.\n62\n This work adds to a growing evidence base shedding light on the behavioural aspects of screening participation and will inform the design and implementation of a new lung screening pilot in Scotland\n40\n, \n45\n, \n46\n (see Box 2 for implementation ideas generated from this work). Minimizing practical barriers is also likely to be instrumental in improving participation and addressing inequity in access to screening. As such, information materials, methods of communication and the design of the process involved in screening (e.g., minimizing the steps, time commitment and waiting intervals), and sensitive, supportive messaging that addresses stigma and fear are all important components of a pilot to break down some of the known barriers. Drawing on the similarities with research based on other UK pilot studies, we are modelling optimized study and participant materials.\n59\n, \n63\n, \n64\n While there is consistency in the findings compared with existing work, it was important to explore the Scottish context with a diverse sample of participants to consider how rurality and deprivation presented any unique issues. Understanding ethnic variations in views on lung screening participation will also be important to ensure equitable provision and uptake of screening. A high burden of multimorbidity is also characteristic of the Scottish population and should be examined in future studies.\n\nMinimize steps in the screening process to lower opportunities for delays and associated distressAvoiding unnecessary travel to scanning facilities with the chance to discuss screening concerns, address fears and perceived stigma, and facilitate informed decision‐makingUse of mobile screening vans as a ‘one stop shop’ to address resource constraints and travel issues for people living in deprived and rural areasConsider covering the cost of travel expenses to screening facilities to ensure equitable accessIncorporating discussion of fears associated with screening into information materialsEnsuring positive messaging with nonjudgemental language around smoking behaviourEnsuring a timely and sensitive approach to smoking cessation advice\n\nMinimize steps in the screening process to lower opportunities for delays and associated distress\nAvoiding unnecessary travel to scanning facilities with the chance to discuss screening concerns, address fears and perceived stigma, and facilitate informed decision‐making\nUse of mobile screening vans as a ‘one stop shop’ to address resource constraints and travel issues for people living in deprived and rural areas\nConsider covering the cost of travel expenses to screening facilities to ensure equitable access\nIncorporating discussion of fears associated with screening into information materials\nEnsuring positive messaging with nonjudgemental language around smoking behaviour\nEnsuring a timely and sensitive approach to smoking cessation advice", "This focus group study has already identified several perceived individual, practical and system barriers and facilitators to participation in a pilot lung screening programme in Scotland using LDCT. While our results resonate with existing literature in this field, they will be helpful in addressing factors which are especially important in Scotland if it is to embrace lung cancer screening—reducing health inequalities, engaging deprived populations and ensuring access in remote and rural areas. The findings will inform the design and implementation of a Scottish pilot lung screening study.", "All authors apart from Jasmin Rostron and Lynsey R. Brown contributed toward the design of the focus groups and helped develop study documents. Debbie Cavers and Mia Nelson conducted the focus groups. Debbie Cavers, Mia Nelson and Jasmin Rostron coded and analysed the transcripts. Lynsey R. Brown read and commented on the analysis. Debbie Cavers drafted the manuscript and all authors edited and refined subsequent drafts.", "F. S. declares that the Universities of Dundee and St. Andrew's received funding from Oncimmune for the Early Detection of Cancer of the Lung Scotland (ECLS) trial from 2013 to 2021. The remaining authors declare no conflict of interest.", "The LUNGSCOT focus group study was approved by the University of Edinburgh Medical School Ethics Committee on 19 March 2021, reference 21‐EMREC‐002. All participants gave consent before taking part in the focus groups.", "Supporting information.\nClick here for additional data file." ]
[ null, "methods", null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, "discussion", null, null, null, null, "conclusions", null, "COI-statement", null, "supplementary-material" ]
[ "early detection", "focus group", "lung cancer", "lung screening", "qualitative", "screening" ]
The burden of chronic diseases and patients' preference for healthcare services among adult patients suffering from chronic diseases in Bangladesh.
36263949
Low- and middle-income countries (LMICs) have a disproportionately high burden of chronic diseases, with inequalities in health care access and quality services. This study aimed to assess patients' preferences for healthcare services for chronic disease management among adult patients in Bangladesh.
BACKGROUND
The present analysis was conducted among 10,385 patients suffering from chronic diseases, drawn from the latest Household Income and Expenditure Survey 2016-2017. We used the multinomial logistic regression to investigate the association of chronic comorbid conditions and healthcare service-related factors with patients' preferences for healthcare services.
METHODS
The top four dimensions of patient preference for healthcare services in order of magnitude were quality of treatment (30.3%), short distance to health facility (27.6%), affordability of health care (21.7%) and availability of doctors (11.0%). Patients with heart disease had a 29% significantly lower preference for healthcare affordability than the quality of healthcare services (relative risk ratio [RRR] = 0.71; 0.56-0.90). Patients who received healthcare services from pharmacies or dispensaries were more likely to prefer a short distance to a health facility (RRR = 6.99; 4.80-9.86) or affordability of healthcare services (RRR = 3.13; 2.25-4.36). Patients with comorbid conditions were more likely to prefer healthcare affordability (RRR = 1.39; 1.15-1.68). In addition, patients who received health care from a public facility had 2.93 times higher preference for the availability of medical doctors (RRR = 2.93; 1.70-5.04) than the quality of treatment in the health facility, when compared with private service providers.
RESULTS
Patient preferences for healthcare services in chronic disease management were significantly associated with the type of disease and its magnitude and characteristics of healthcare providers. Therefore, to enhance service provision and equitable distribution and uptake of health services, policymakers and public health practitioners should consider patient preferences in designing national strategic frameworks for chronic disease management.
CONCLUSIONS
[ "Adult", "Humans", "Patient Preference", "Bangladesh", "Health Services", "Chronic Disease", "Health Services Accessibility", "Logistic Models" ]
9700186
BACKGROUND
Chronic diseases have become a global challenge, imposing an enormous economic and health burden on society. 1 Chronic diseases are defined as health conditions lasting 12 months or more, which require ongoing medical intervention and may result in the limitation of activities of daily living. 1 The epidemiological burden of chronic diseases 2 and exposure to their risk factors are increasing worldwide, 3 particularly in low‐ and middle‐income countries (LMICs), such as Bangladesh. 4 , 5 , 6 Chronic diseases account for around 41 million deaths each year, representing about 71% of all deaths globally. 7 The most common chronic diseases include cardiovascular diseases (e.g., coronary heart diseases, stroke and peripheral vascular diseases), diabetes, cancers, chronic obstructive pulmonary disease, mental illness and arthritis 7 ; approximately 77% of all yearly deaths are related to chronic diseases occur in LMICs, of which 85% occur in the most productive age groups (30–69 years). 7 It has been estimated that chronic diseases will account for an accumulated global economic loss of 47 trillion US dollars by 2030, approximately 75% of the global gross domestic product. 8 In Bangladesh, an LMIC with a substantial social and economic burden, about 886,000 deaths (i.e., 59% of total deaths) occur due to chronic diseases each year. 3 , 9 The burden due to chronic diseases has increased in Bangladesh from 43.4% in 2000 to 66.9% in 2015. 3 It is anticipated that chronic diseases will exceed the combined burden of communicable, maternal, perinatal and nutrition‐related diseases by 2030 globally, including in Bangladesh. 10 , 11 Despite the high burden of chronic diseases, Bangladesh, like many LMIC countries, does not have a national integrated chronic diseases management policy, strategy or action plan. 9 , 11 For instance, the prevalence of undiagnosed chronic diseases is high, and the proportion of unmanaged chronic diseases is even higher in many LMICs, 10 including Bangladesh. 12 This highlights the frequent inadequacies in the diagnosis, prevention and management of chronic diseases among the healthcare systems of LMICs. 9 , 10 Efforts in chronic disease management in Bangladesh continue to be inadequate. Little attention has been given to addressing the contributing behaviours associated with chronic diseases, including unhealthy dietary patterns, lack of physical activity and exposure to factors potentially detrimental to health such as alcohol, drug and tobacco use. 6 However, it is possible to counteract the rising prevalence of the chronic disease by implementing effective prevention strategies, population‐based screening, reduction of risk factors, early detection and appropriate treatments. 10 , 11 If such actions are not taken, the burden of chronic diseases, which is referred to as an emerging prevalence of chronic diseases globally imposing an enormous economic and health burden will likely continue to rise, 10 , 11 which is alarming especially among the vulnerable and marginalized populations of Bangladesh with limited affordability for health services. 11 In addition, the emerging prevalence of chronic disease may also lead to a health system burden in terms of increasing healthcare utilization, treatment costs and chronic disease management. It is well established that chronic diseases are increasingly associated with the over‐utilization of healthcare services and a higher financial burden. 13 For example, a previous study documented that a higher number of chronic diseases was linked to an increased number of outpatient visits. 14 Therefore, adequate preventative services must be in place to reduce the social‐economic burden of chronic disease, thereby ensuring optimal use of health resources. However, the major challenges to ensure effective prevention and management of chronic diseases include social status, power gradients, racial/ethnic differences, poor accessibility and affordability of healthcare services. 2 , 6 , 15 , 16 , 17 Previous research has identified several factors associated with healthcare utilization, such as demographic and socioeconomic characteristics, type of healthcare providers and presence of chronic illnesses. 18 In addition, patients' preference for healthcare services depends on several factors, including personal preference, disease severity, economic capacity, the reputation of healthcare providers 18 as well as affordable costs associated with treatment. 19 Furthermore, short travel time to healthcare facilities, effective interactions with healthcare providers, 19 , 20 respectful service provider attitudes 21 and short waiting time 22 were positively associated with the patient preference for healthcare services. A recent discrete choice experiment study found that the availability of medicine and transport to the health facility were significant attributes of patient preference for healthcare services. 23 Notably, optimum healthcare utilization among chronically ill patients has a significant role in preventing and managing chronic diseases. In Bangladesh, a family spends an average of 11% of their total household budget on health care and half of the population spends 7% of their monthly per capita consumption expenditure on illness. 24 Understanding patients' preferences could help medical professionals and healthcare providers restructure the healthcare delivery model and ensure the quality of services. In addition to clinical guidelines, patients' preferences may also provide guidelines for the selection of treatment options. Patient preferences also help to inform clinical decisions where science has not yet been able to provide effective solutions to healthcare problems. 25 Therefore, information on patients' preferences for healthcare utilization in terms of healthcare expenditure is critical; without this knowledge, an effective national healthcare policy cannot be formulated. 26 Moreover, there is a current lack of evidence on the patients' preferences for healthcare services in chronic disease management in Bangladesh. Since no such study exists, the present study aimed to investigate patients' preference for healthcare services among adult patients in Bangladesh. Understanding patients' preferences are important in designing interventions aimed at reducing the burden of chronic diseases, which undermine economic growth and development and are increasingly becoming the leading cause of death.
METHODS
[SUBTITLE] Study design and data sources [SUBSECTION] This study followed an observational, cross‐sectional design. The data was extracted from the most recent nationally representative Household Income and Expenditure Survey (HIES), conducted during 2016–2017 by the Bangladesh Bureau of Statistics (BBS) (Supporting Information: Document 1). The HIES is a cross‐sectional survey conducted every 5 years since 1973 in Bangladesh with the objective of providing national estimates on income, expenditure and consumption, poverty, standard of living, health status and education. 27 In the HIES survey, a semistructured questionnaire (Supporting Information: Appendix Table A1: HIES 2016–2017 Questionnaire) was used to collect information from the survey participants (adults aged 18 years or above) under nine modules: (1) household information, (2) education, (3) health‐illnesses and injuries, (4) economic activities and wage employment, (5) nonagricultural enterprises, (6) housing, (7) agriculture, (8) other assets and income and (9) consumption (Supporting Information: Appendix Table A1: HIES 2016–2017 Questionnaire). However, our analysis is based on the chronic disease‐related questions included in Module‐3: Health (Illnesses and Injuries) of the HIES (Supporting Information: Appendix Table A2). Therefore, we only used the indicators pertaining to chronic disease and health service utilization along with the sociodemographic characterizes of the participants (Supporting Information: Appendix Table A2). All health‐related information was self‐reported in the HIES. Respondents were asked to prioritize the chronic diseases they were suffering from in order of their importance. We selected the primary disease (i.e., principal diagnosis) based on the patients' experience of diseases in order of their importance. For instance, if one patient was diagnosed with three chronic conditions, they reported three diseases in order of importance (i.e., first, second and third importance). Therefore, a patient's first importance of disease was considered as the primary disease in the present study. This study followed an observational, cross‐sectional design. The data was extracted from the most recent nationally representative Household Income and Expenditure Survey (HIES), conducted during 2016–2017 by the Bangladesh Bureau of Statistics (BBS) (Supporting Information: Document 1). The HIES is a cross‐sectional survey conducted every 5 years since 1973 in Bangladesh with the objective of providing national estimates on income, expenditure and consumption, poverty, standard of living, health status and education. 27 In the HIES survey, a semistructured questionnaire (Supporting Information: Appendix Table A1: HIES 2016–2017 Questionnaire) was used to collect information from the survey participants (adults aged 18 years or above) under nine modules: (1) household information, (2) education, (3) health‐illnesses and injuries, (4) economic activities and wage employment, (5) nonagricultural enterprises, (6) housing, (7) agriculture, (8) other assets and income and (9) consumption (Supporting Information: Appendix Table A1: HIES 2016–2017 Questionnaire). However, our analysis is based on the chronic disease‐related questions included in Module‐3: Health (Illnesses and Injuries) of the HIES (Supporting Information: Appendix Table A2). Therefore, we only used the indicators pertaining to chronic disease and health service utilization along with the sociodemographic characterizes of the participants (Supporting Information: Appendix Table A2). All health‐related information was self‐reported in the HIES. Respondents were asked to prioritize the chronic diseases they were suffering from in order of their importance. We selected the primary disease (i.e., principal diagnosis) based on the patients' experience of diseases in order of their importance. For instance, if one patient was diagnosed with three chronic conditions, they reported three diseases in order of importance (i.e., first, second and third importance). Therefore, a patient's first importance of disease was considered as the primary disease in the present study. [SUBTITLE] HIES survey sampling and sample size calculation [SUBSECTION] The sample frame used in the selection of Primary Sample Units (PSUs) for the HIES 2016–2017 was based on the Census of Population and Housing 2011. 27 In this survey, eight administrative divisions (Barisal, Chittagong, Dhaka, Khulna, Mymensingh, Rajshahi, Rangpur and Sylhet) were included and stratified by three basic localities, being rural, urban and metropolitan areas (city corporations). Thus, these should be 8 divisions × 3 localities totalling 24 strata (8 × 3 = 24). However, the sampling frame (Census of Population and Housing 2011) did not contain three administrative divisions (i.e., Rangpur, Barisal and Sylhet). Additionally, the BBS included only four main city corporations (Dhaka, Chittagong, Khulna and Rajshahi) in the city corporation locality. PSUs were randomly selected from 20 strata (eight rural divisions, eight urban divisions and four metropolitan areas) for national representation. As the PSUs of HIES 2016–2017 were allocated at the district level, the sample design includes a total of 132 substrata: 64 rural, 64 urban and 4 metropolitan areas. 27 Sample size was calculated using the prevalence rate of the main indicator (poverty rate) or the coefficient of variation of per capita consumption or household consumption, which are the core indicators of the HIES 2016–2017. Each one was treated as a target variable for determining the sample size (Supporting Information: Document 1). The required sample size was calculated for each district and explained elsewhere, 27 using the following formula (1) n=Zα2×CVSRS(y¯)r(Y¯)2×DEFF, where, n was the required sample for allocation to each district to achieve a certain level in the accuracy statistic (r(Y®) = 10% relative standard error desired for the mean total household expenditure estimated at the district level associated with the targeted variable (y®);CVSRS(y®) was the coefficient of variation of the targeted variable [i.e., total household expenditure estimated at the national level] estimated under the assumption of simple random sampling; DEFF was the design effect of the target variable [i.e., the average design effect of the target variable across all districts]; and Zα2 [=1.96] was the critical value of a standard normal distribution with an α% (5%) significance level). Substituting all values in Equation (1), the required sample was 715 households for each district, nonetheless, 720 households were allocated to each district for practical consideration and to facilitate fieldwork and survey implementation management. A stratified, two‐stage cluster sampling technique was used in this survey. In the first stage, a total of 36 PSUs were drawn from each district by applying the probability proportional to size systematic sampling technique, using the number of households in each PSU as the measure of size. The 36 PSUs were randomly selected from rural, urban and city corporation substratum. The total number of PSUs included in the analysis was 2304 (64 districts × 36 PSU per district). In the second stage, 20 households were selected per PSU. Using this sampling technique, 46,076 households (2304 PUSs × 20 households per PSU) were included in the study analysis of HIES 2016–2017 data. Among the selected households, a total of 186,076 individuals were included. The sample frame used in the selection of Primary Sample Units (PSUs) for the HIES 2016–2017 was based on the Census of Population and Housing 2011. 27 In this survey, eight administrative divisions (Barisal, Chittagong, Dhaka, Khulna, Mymensingh, Rajshahi, Rangpur and Sylhet) were included and stratified by three basic localities, being rural, urban and metropolitan areas (city corporations). Thus, these should be 8 divisions × 3 localities totalling 24 strata (8 × 3 = 24). However, the sampling frame (Census of Population and Housing 2011) did not contain three administrative divisions (i.e., Rangpur, Barisal and Sylhet). Additionally, the BBS included only four main city corporations (Dhaka, Chittagong, Khulna and Rajshahi) in the city corporation locality. PSUs were randomly selected from 20 strata (eight rural divisions, eight urban divisions and four metropolitan areas) for national representation. As the PSUs of HIES 2016–2017 were allocated at the district level, the sample design includes a total of 132 substrata: 64 rural, 64 urban and 4 metropolitan areas. 27 Sample size was calculated using the prevalence rate of the main indicator (poverty rate) or the coefficient of variation of per capita consumption or household consumption, which are the core indicators of the HIES 2016–2017. Each one was treated as a target variable for determining the sample size (Supporting Information: Document 1). The required sample size was calculated for each district and explained elsewhere, 27 using the following formula (1) n=Zα2×CVSRS(y¯)r(Y¯)2×DEFF, where, n was the required sample for allocation to each district to achieve a certain level in the accuracy statistic (r(Y®) = 10% relative standard error desired for the mean total household expenditure estimated at the district level associated with the targeted variable (y®);CVSRS(y®) was the coefficient of variation of the targeted variable [i.e., total household expenditure estimated at the national level] estimated under the assumption of simple random sampling; DEFF was the design effect of the target variable [i.e., the average design effect of the target variable across all districts]; and Zα2 [=1.96] was the critical value of a standard normal distribution with an α% (5%) significance level). Substituting all values in Equation (1), the required sample was 715 households for each district, nonetheless, 720 households were allocated to each district for practical consideration and to facilitate fieldwork and survey implementation management. A stratified, two‐stage cluster sampling technique was used in this survey. In the first stage, a total of 36 PSUs were drawn from each district by applying the probability proportional to size systematic sampling technique, using the number of households in each PSU as the measure of size. The 36 PSUs were randomly selected from rural, urban and city corporation substratum. The total number of PSUs included in the analysis was 2304 (64 districts × 36 PSU per district). In the second stage, 20 households were selected per PSU. Using this sampling technique, 46,076 households (2304 PUSs × 20 households per PSU) were included in the study analysis of HIES 2016–2017 data. Among the selected households, a total of 186,076 individuals were included.
RESULTS
Table 1 shows the participant's characteristics. Of the 10,385 patients with chronic diseases, approximately 50% were female, 51% were married and 70% were unemployed. The majority of the patients were aged between 18 and 45 (81%) years, and around one‐third (37%) had no formal education. Distribution of participant's characteristics Note: p value was derived using χ 2 test or one‐way analysis of variance where appropriate. Abbreviations: BDT, Bangladeshi Taka; CI, confidence interval; SD, standard deviation. [SUBTITLE] The distribution of chronic disease and utilization of healthcare services [SUBSECTION] The most prevalent chronic diseases reported by the patients included gastric/ulcer (16%) followed by arthritis/rheumatism (14%) (Table 1). Most of the patients (65%) reported one diagnosed chronic condition, while approximately 24% of the patients had two, and 11% had three or more chronic conditions. Most of the patients (91%) reported utilizing outpatient healthcare services in the past 30 days. A high proportion of the patients visited general practitioner clinics (~40%), followed by pharmacy/dispensary (24%) and public hospitals (20%) for healthcare services. Furthermore, approximately 53% of the patients received services at rural health facilities. The average out‐of‐pocket healthcare expenditure for chronic illness in the last 30 days was 3848 BDT (~47.40 USD; 2017 price year). The most prevalent chronic diseases reported by the patients included gastric/ulcer (16%) followed by arthritis/rheumatism (14%) (Table 1). Most of the patients (65%) reported one diagnosed chronic condition, while approximately 24% of the patients had two, and 11% had three or more chronic conditions. Most of the patients (91%) reported utilizing outpatient healthcare services in the past 30 days. A high proportion of the patients visited general practitioner clinics (~40%), followed by pharmacy/dispensary (24%) and public hospitals (20%) for healthcare services. Furthermore, approximately 53% of the patients received services at rural health facilities. The average out‐of‐pocket healthcare expenditure for chronic illness in the last 30 days was 3848 BDT (~47.40 USD; 2017 price year). [SUBTITLE] Preferences for healthcare services for chronic illnesses [SUBSECTION] Approximately 30% of patients reported that they preferred quality healthcare services, whereas 28% preferred a short distance to the health facility. Furthermore, 22% of the patients preferred affordable healthcare costs as the main driver, and 11% expressed a preference for the doctor's availability. However, the scenario varied among different chronic illnesses and healthcare services (Table 1). For example, among the patients who received healthcare services from a pharmacy or dispensary, 49% preferred short distances to health facilities, and 32% reported their preference for healthcare affordability. Approximately 30% of patients reported that they preferred quality healthcare services, whereas 28% preferred a short distance to the health facility. Furthermore, 22% of the patients preferred affordable healthcare costs as the main driver, and 11% expressed a preference for the doctor's availability. However, the scenario varied among different chronic illnesses and healthcare services (Table 1). For example, among the patients who received healthcare services from a pharmacy or dispensary, 49% preferred short distances to health facilities, and 32% reported their preference for healthcare affordability. [SUBTITLE] Correlates of patient preference for chronic disease‐related healthcare services [SUBSECTION] Table 2 highlights the factors that influence a patient's choice of healthcare services. Among patients with heart disease, quality of healthcare services was 29% more preferred than affordability (RRR = 0.71; 95% confidence interval [CI]: 0.56–0.90; p < .001). Influencing factors on patient's preference of healthcare services Note: ***, ** and * denoted significance level at 0.1%, 1% and 5%, respectively. Abbreviations: CI, confidence interval; ref, reference group; RRR, relative risk ratio. Similarly, diabetic patients exhibited a 38% higher preference for quality rather than affordability of healthcare services (RRR = 0.62; 0.47–0.81; p < .001). However, patients with three or more chronic comorbid conditions were 1.39 times more likely to prefer affordability over the quality of healthcare services (RRR = 1.39; 1.15–1.68; p < .001). Patients who received healthcare services from a public facility reported a higher preference for the availability of medical doctors or consultants (RRR = 2.93; 1.70–5.04; p < .001) than those receiving health care from private service providers. In addition, patients who received healthcare services from pharmacies or dispensaries were significantly more likely to prefer a short distance to a healthcare facility (RRR = 6.99; 4.80–9.86; p < .001) or affordability of healthcare services (RRR = 3.13; 2.25–4.36; p < .001), rather than the quality of the healthcare services and availability of doctors. Table 2 highlights the factors that influence a patient's choice of healthcare services. Among patients with heart disease, quality of healthcare services was 29% more preferred than affordability (RRR = 0.71; 95% confidence interval [CI]: 0.56–0.90; p < .001). Influencing factors on patient's preference of healthcare services Note: ***, ** and * denoted significance level at 0.1%, 1% and 5%, respectively. Abbreviations: CI, confidence interval; ref, reference group; RRR, relative risk ratio. Similarly, diabetic patients exhibited a 38% higher preference for quality rather than affordability of healthcare services (RRR = 0.62; 0.47–0.81; p < .001). However, patients with three or more chronic comorbid conditions were 1.39 times more likely to prefer affordability over the quality of healthcare services (RRR = 1.39; 1.15–1.68; p < .001). Patients who received healthcare services from a public facility reported a higher preference for the availability of medical doctors or consultants (RRR = 2.93; 1.70–5.04; p < .001) than those receiving health care from private service providers. In addition, patients who received healthcare services from pharmacies or dispensaries were significantly more likely to prefer a short distance to a healthcare facility (RRR = 6.99; 4.80–9.86; p < .001) or affordability of healthcare services (RRR = 3.13; 2.25–4.36; p < .001), rather than the quality of the healthcare services and availability of doctors.
CONCLUSION
Our study findings highlighted that patient preferences for healthcare services in chronic disease management were significantly associated with disease severity and healthcare providers' attributes. Therefore, policymakers and public health practitioners should consider patient preferences for managing chronic conditions within the national strategic frameworks to improve service provision, equitable distribution and uptake of the services.
[ "Study design and data sources", "HIES survey sampling and sample size calculation", "DATA COLLECTION", "Study population", "DEFINITIONS OF STUDY VARIABLES", "Outcome measures", "Chronic diseases and comorbid conditions", "Covariates", "STATISTICAL ANALYSIS", "The distribution of chronic disease and utilization of healthcare services", "Preferences for healthcare services for chronic illnesses", "Correlates of patient preference for chronic disease‐related healthcare services", "Implications for policy and practice", "AUTHOR CONTRIBUTIONS", "ETHICS STATEMENT" ]
[ "This study followed an observational, cross‐sectional design. The data was extracted from the most recent nationally representative Household Income and Expenditure Survey (HIES), conducted during 2016–2017 by the Bangladesh Bureau of Statistics (BBS) (Supporting Information: Document 1). The HIES is a cross‐sectional survey conducted every 5 years since 1973 in Bangladesh with the objective of providing national estimates on income, expenditure and consumption, poverty, standard of living, health status and education.\n27\n\n\nIn the HIES survey, a semistructured questionnaire (Supporting Information: Appendix Table A1: HIES 2016–2017 Questionnaire) was used to collect information from the survey participants (adults aged 18 years or above) under nine modules: (1) household information, (2) education, (3) health‐illnesses and injuries, (4) economic activities and wage employment, (5) nonagricultural enterprises, (6) housing, (7) agriculture, (8) other assets and income and (9) consumption (Supporting Information: Appendix Table A1: HIES 2016–2017 Questionnaire). However, our analysis is based on the chronic disease‐related questions included in Module‐3: Health (Illnesses and Injuries) of the HIES (Supporting Information: Appendix Table A2). Therefore, we only used the indicators pertaining to chronic disease and health service utilization along with the sociodemographic characterizes of the participants (Supporting Information: Appendix Table A2). All health‐related information was self‐reported in the HIES. Respondents were asked to prioritize the chronic diseases they were suffering from in order of their importance. We selected the primary disease (i.e., principal diagnosis) based on the patients' experience of diseases in order of their importance. For instance, if one patient was diagnosed with three chronic conditions, they reported three diseases in order of importance (i.e., first, second and third importance). Therefore, a patient's first importance of disease was considered as the primary disease in the present study.", "The sample frame used in the selection of Primary Sample Units (PSUs) for the HIES 2016–2017 was based on the Census of Population and Housing 2011.\n27\n In this survey, eight administrative divisions (Barisal, Chittagong, Dhaka, Khulna, Mymensingh, Rajshahi, Rangpur and Sylhet) were included and stratified by three basic localities, being rural, urban and metropolitan areas (city corporations). Thus, these should be 8 divisions × 3 localities totalling 24 strata (8 × 3 = 24). However, the sampling frame (Census of Population and Housing 2011) did not contain three administrative divisions (i.e., Rangpur, Barisal and Sylhet). Additionally, the BBS included only four main city corporations (Dhaka, Chittagong, Khulna and Rajshahi) in the city corporation locality. PSUs were randomly selected from 20 strata (eight rural divisions, eight urban divisions and four metropolitan areas) for national representation. As the PSUs of HIES 2016–2017 were allocated at the district level, the sample design includes a total of 132 substrata: 64 rural, 64 urban and 4 metropolitan areas.\n27\n Sample size was calculated using the prevalence rate of the main indicator (poverty rate) or the coefficient of variation of per capita consumption or household consumption, which are the core indicators of the HIES 2016–2017. Each one was treated as a target variable for determining the sample size (Supporting Information: Document 1). The required sample size was calculated for each district and explained elsewhere,\n27\n using the following formula\n\n(1)\nn=Zα2×CVSRS(y¯)r(Y¯)2×DEFF,\nwhere, n was the required sample for allocation to each district to achieve a certain level in the accuracy statistic (r(Y®) = 10% relative standard error desired for the mean total household expenditure estimated at the district level associated with the targeted variable (y®);CVSRS(y®) was the coefficient of variation of the targeted variable [i.e., total household expenditure estimated at the national level] estimated under the assumption of simple random sampling; DEFF was the design effect of the target variable [i.e., the average design effect of the target variable across all districts]; and Zα2 [=1.96] was the critical value of a standard normal distribution with an α% (5%) significance level). Substituting all values in Equation (1), the required sample was 715 households for each district, nonetheless, 720 households were allocated to each district for practical consideration and to facilitate fieldwork and survey implementation management. A stratified, two‐stage cluster sampling technique was used in this survey. In the first stage, a total of 36 PSUs were drawn from each district by applying the probability proportional to size systematic sampling technique, using the number of households in each PSU as the measure of size. The 36 PSUs were randomly selected from rural, urban and city corporation substratum. The total number of PSUs included in the analysis was 2304 (64 districts × 36 PSU per district). In the second stage, 20 households were selected per PSU. Using this sampling technique, 46,076 households (2304 PUSs × 20 households per PSU) were included in the study analysis of HIES 2016–2017 data. Among the selected households, a total of 186,076 individuals were included.", "Data collection was accomplished between early April 2016 and late March 2017 through face‐to‐face interviews with the participants. A total of 128 enumeration teams, each comprised of five members (one supervising officer, two interviewers and two female facilitators) who were thoroughly trained, completed the data collection. A multilayered quality control measure was employed to ensure the quality of the collected data. The supervising officer of each team was responsible to verify all the questionnaires completed by the field staff before they were sent to the headquarter. Moreover, Deputy Directors of the District Statistical Offices as well as senior officials from the headquarter, frequently visited the sampled areas to ensure data quality (Supporting Information: Document 1).\nThe survey team formulated an operational definition of each variable used in the questionnaire. For example, while collecting chronic disease‐related information, the enumerators were instructed to ask the respondents, ‘Have you suffered from any chronic illness/disability in the last 12 months or more?’ (if yes); then, participants were asked a second question: ‘What chronic illness/disability are you suffering from?’ with response options: (1) chronic fever, (2) Injuries/disability, (3) chronic heart disease, (4) respiratory diseases/asthma/bronchitis, (5) diarrhoea/dysentery, (6) gastric/ulcer, (7) blood pressure, (8) arthritis/rheumatism, (9) skin problem, (10) diabetes, (11) cancer, (12) kidney diseases, (13) liver diseases, (14) mental health, (15) paralysis, (16) ear/ENT problem, (17) eye problem or (18) other (specify) (Supporting Information: Appendix Table A1: HIES 2016–2017 questionnaire). Participants responded based on their disease diagnosis, experiences, symptoms of illness and course of treatment. To get valid information, the enumerators also probed where necessary, asked the respondents to show any relevant documents such as prescriptions and test reports, or explained to the respondents about chronic diseases using various case scenarios as outlined in the survey guidelines and instructions.\n[SUBTITLE] Study population [SUBSECTION] All selected household members were included in the survey. The participants were selected based on the HIES 2016–2017 survey protocol and following the same inclusion criteria: (1) an individual who had suffered from any chronic disease for the last 12 months or more and (2) patients who received any treatment due to chronic disease in the last 30 days. Based on these inclusion criteria, a total of 10,385 patients were selected for the present analysis (Figure 1).\nDistribution of study participants\nAll selected household members were included in the survey. The participants were selected based on the HIES 2016–2017 survey protocol and following the same inclusion criteria: (1) an individual who had suffered from any chronic disease for the last 12 months or more and (2) patients who received any treatment due to chronic disease in the last 30 days. Based on these inclusion criteria, a total of 10,385 patients were selected for the present analysis (Figure 1).\nDistribution of study participants", "All selected household members were included in the survey. The participants were selected based on the HIES 2016–2017 survey protocol and following the same inclusion criteria: (1) an individual who had suffered from any chronic disease for the last 12 months or more and (2) patients who received any treatment due to chronic disease in the last 30 days. Based on these inclusion criteria, a total of 10,385 patients were selected for the present analysis (Figure 1).\nDistribution of study participants", "[SUBTITLE] Outcome measures [SUBSECTION] Patients' preference for chronic disease healthcare utilization was the primary outcome measure for this study. Participants responded to questions that asked them about their preference for selecting a healthcare provider: ‘why did you choose this provider?’ Response options were recoded as the quality of healthcare services, availability of medical doctors (e.g., availability of male or female doctors in a health facility), affordable healthcare services (i.e., acceptable costs), short distance to health facility (i.e., nearby) and others (e.g., referred by other providers).\nPatients' preference for chronic disease healthcare utilization was the primary outcome measure for this study. Participants responded to questions that asked them about their preference for selecting a healthcare provider: ‘why did you choose this provider?’ Response options were recoded as the quality of healthcare services, availability of medical doctors (e.g., availability of male or female doctors in a health facility), affordable healthcare services (i.e., acceptable costs), short distance to health facility (i.e., nearby) and others (e.g., referred by other providers).\n[SUBTITLE] Chronic diseases and comorbid conditions [SUBSECTION] Participants were asked about chronic illness, ‘have you suffered from any chronic illness/disability in the last 12 months or more?’ The prevalence of chronic diseases was assessed based on this question. Individuals who suffered from chronic disease for the last 12 months (or more) and received any treatment due to chronic diseases were the only population considered in this study. The study population was diagnosed with chronic illnesses and conditions such as chronic heart disease, respiratory diseases, chronic gastric or ulcers, high blood pressure, arthritis or rheumatism, diabetes, chronic fever and other diseases. There is no gold‐standard method for measuring comorbidity status among patients with chronic diseases.\n28\n A previous review study identified that 21 separate approaches were executed to measure comorbidity status.\n28\n The selection of the approach depends on the study research questions, study design, data availability and population studied. The most straightforward approach to measuring comorbidity status is to investigate the distribution of individual comorbid conditions and to treat them independently and/or to combine them by summing the total number of conditions.\n29\n, \n30\n A single condition count approach was performed to measure comorbidity status in this study. The count of chronic health condition(s) was measured for each patient based on the number of disease exposures and who had been prescribed medication for their illness. It was counted as multiple responses if the patients had multiple chronic conditions. In addition, the principal diagnosis was defined based on patients diagnosed based on their reported first importance. Chronic comorbid conditions (principal diagnosis plus one, two or three or more comorbid conditions) were assessed in this study.\nParticipants were asked about chronic illness, ‘have you suffered from any chronic illness/disability in the last 12 months or more?’ The prevalence of chronic diseases was assessed based on this question. Individuals who suffered from chronic disease for the last 12 months (or more) and received any treatment due to chronic diseases were the only population considered in this study. The study population was diagnosed with chronic illnesses and conditions such as chronic heart disease, respiratory diseases, chronic gastric or ulcers, high blood pressure, arthritis or rheumatism, diabetes, chronic fever and other diseases. There is no gold‐standard method for measuring comorbidity status among patients with chronic diseases.\n28\n A previous review study identified that 21 separate approaches were executed to measure comorbidity status.\n28\n The selection of the approach depends on the study research questions, study design, data availability and population studied. The most straightforward approach to measuring comorbidity status is to investigate the distribution of individual comorbid conditions and to treat them independently and/or to combine them by summing the total number of conditions.\n29\n, \n30\n A single condition count approach was performed to measure comorbidity status in this study. The count of chronic health condition(s) was measured for each patient based on the number of disease exposures and who had been prescribed medication for their illness. It was counted as multiple responses if the patients had multiple chronic conditions. In addition, the principal diagnosis was defined based on patients diagnosed based on their reported first importance. Chronic comorbid conditions (principal diagnosis plus one, two or three or more comorbid conditions) were assessed in this study.\n[SUBTITLE] Covariates [SUBSECTION] Based on the ongoing literature and the authors' own expertise, we selected variables to address the study objective, including healthcare service‐related factors (e.g., type of healthcare service‐inpatient or outpatient, type of health facilities, waiting time for receiving healthcare services, out‐of‐pocket payment and location of consulted healthcare provider) and patients' sociodemographics factors (e.g., gender, age, marital status, education and employment) (Supporting Information: Appendix Table A2).\nBased on the ongoing literature and the authors' own expertise, we selected variables to address the study objective, including healthcare service‐related factors (e.g., type of healthcare service‐inpatient or outpatient, type of health facilities, waiting time for receiving healthcare services, out‐of‐pocket payment and location of consulted healthcare provider) and patients' sociodemographics factors (e.g., gender, age, marital status, education and employment) (Supporting Information: Appendix Table A2).", "Patients' preference for chronic disease healthcare utilization was the primary outcome measure for this study. Participants responded to questions that asked them about their preference for selecting a healthcare provider: ‘why did you choose this provider?’ Response options were recoded as the quality of healthcare services, availability of medical doctors (e.g., availability of male or female doctors in a health facility), affordable healthcare services (i.e., acceptable costs), short distance to health facility (i.e., nearby) and others (e.g., referred by other providers).", "Participants were asked about chronic illness, ‘have you suffered from any chronic illness/disability in the last 12 months or more?’ The prevalence of chronic diseases was assessed based on this question. Individuals who suffered from chronic disease for the last 12 months (or more) and received any treatment due to chronic diseases were the only population considered in this study. The study population was diagnosed with chronic illnesses and conditions such as chronic heart disease, respiratory diseases, chronic gastric or ulcers, high blood pressure, arthritis or rheumatism, diabetes, chronic fever and other diseases. There is no gold‐standard method for measuring comorbidity status among patients with chronic diseases.\n28\n A previous review study identified that 21 separate approaches were executed to measure comorbidity status.\n28\n The selection of the approach depends on the study research questions, study design, data availability and population studied. The most straightforward approach to measuring comorbidity status is to investigate the distribution of individual comorbid conditions and to treat them independently and/or to combine them by summing the total number of conditions.\n29\n, \n30\n A single condition count approach was performed to measure comorbidity status in this study. The count of chronic health condition(s) was measured for each patient based on the number of disease exposures and who had been prescribed medication for their illness. It was counted as multiple responses if the patients had multiple chronic conditions. In addition, the principal diagnosis was defined based on patients diagnosed based on their reported first importance. Chronic comorbid conditions (principal diagnosis plus one, two or three or more comorbid conditions) were assessed in this study.", "Based on the ongoing literature and the authors' own expertise, we selected variables to address the study objective, including healthcare service‐related factors (e.g., type of healthcare service‐inpatient or outpatient, type of health facilities, waiting time for receiving healthcare services, out‐of‐pocket payment and location of consulted healthcare provider) and patients' sociodemographics factors (e.g., gender, age, marital status, education and employment) (Supporting Information: Appendix Table A2).", "The adjusted multinomial logistic regression model was used to identify the potential factors that had an association with the patient's preference for healthcare service. The dependent variable (chronic illness patient's preference for healthcare services) was characterized as a categorical measure in the regression model. An unadjusted analysis was performed using each of the explanatory variables for the following reasons: (1) primary screening of the selection of qualified variables, which were added in the adjusted model, (2) the χ\n2 tests (or one‐way analysis where appropriate) were used to find the association between outcome and explanatory variables. However, the majority of the explanatory variables were categorical with two or more labels; therefore, an unadjusted analysis was performed to find the association between the outcome variable and different categories of explanatory variables. The explanatory variables were included in the adjusted model only if any label of the predictor was significant at a 5% or less risk level in the unadjusted model, which in turn was used to adjust for the associations of other potential confounders. For the explanatory variables, the category found to be least at risk of having patients' preferences for healthcare services related to chronic illness in the analysis was considered the reference category for constructing the relative risk ratio (RRR). Statistical significance was considered at a 5% risk level. All data analyses were undertaken using the statistical software Stata/SE 14 (StataCorp).", "The most prevalent chronic diseases reported by the patients included gastric/ulcer (16%) followed by arthritis/rheumatism (14%) (Table 1). Most of the patients (65%) reported one diagnosed chronic condition, while approximately 24% of the patients had two, and 11% had three or more chronic conditions. Most of the patients (91%) reported utilizing outpatient healthcare services in the past 30 days. A high proportion of the patients visited general practitioner clinics (~40%), followed by pharmacy/dispensary (24%) and public hospitals (20%) for healthcare services. Furthermore, approximately 53% of the patients received services at rural health facilities. The average out‐of‐pocket healthcare expenditure for chronic illness in the last 30 days was 3848 BDT (~47.40 USD; 2017 price year).", "Approximately 30% of patients reported that they preferred quality healthcare services, whereas 28% preferred a short distance to the health facility. Furthermore, 22% of the patients preferred affordable healthcare costs as the main driver, and 11% expressed a preference for the doctor's availability. However, the scenario varied among different chronic illnesses and healthcare services (Table 1). For example, among the patients who received healthcare services from a pharmacy or dispensary, 49% preferred short distances to health facilities, and 32% reported their preference for healthcare affordability.", "Table 2 highlights the factors that influence a patient's choice of healthcare services. Among patients with heart disease, quality of healthcare services was 29% more preferred than affordability (RRR = 0.71; 95% confidence interval [CI]: 0.56–0.90; p < .001).\nInfluencing factors on patient's preference of healthcare services\n\nNote: ***, ** and * denoted significance level at 0.1%, 1% and 5%, respectively.\nAbbreviations: CI, confidence interval; ref, reference group; RRR, relative risk ratio.\nSimilarly, diabetic patients exhibited a 38% higher preference for quality rather than affordability of healthcare services (RRR = 0.62; 0.47–0.81; p < .001). However, patients with three or more chronic comorbid conditions were 1.39 times more likely to prefer affordability over the quality of healthcare services (RRR = 1.39; 1.15–1.68; p < .001). Patients who received healthcare services from a public facility reported a higher preference for the availability of medical doctors or consultants (RRR = 2.93; 1.70–5.04; p < .001) than those receiving health care from private service providers. In addition, patients who received healthcare services from pharmacies or dispensaries were significantly more likely to prefer a short distance to a healthcare facility (RRR = 6.99; 4.80–9.86; p < .001) or affordability of healthcare services (RRR = 3.13; 2.25–4.36; p < .001), rather than the quality of the healthcare services and availability of doctors.", "Our study highlights the significance of patient preferences for healthcare utilization. This study provides timely findings to address health inequities linked to sociocultural and economic factors in Bangladesh. Understanding patients' preferences in chronic disease management is critical to achieving the Sustainable Development Goal (SDG) target 3.4, which focuses solely on reducing premature mortality from noncommunicable diseases by a third by 2030 relative when compared to 2015 as a baseline. Bangladesh is a signatory to the SDGs and the Colombo declaration (strengthening health systems to accelerate delivery of NCD‐related services at the primary healthcare level),\n48\n providing evidence that informs culturally competent NCD prevention and treatment approaches through tailored and responsive health financing, and expenditure policies will contribute significantly to achieving the SDG target 3.4. However, progress in implementing strategies to meet international targets related to NCDs remains slow.\n9\n, \n11\n, \n49\n\n\nAdditionally, Bangladesh lacks a national surveillance programme focused on chronic diseases and does not have any integrated community public health programme that regularly monitors chronic diseases.\n11\n An established disease management plan should consider the adequacy, accessibility, affordability and quality of services.\n48\n Most importantly, the factors influencing patient preferences for healthcare services may not have been considered when developing the chronic disease management policy.\n11\n Therefore, policymakers and public health practitioners should consider patient preferences regarding healthcare utilization in managing chronic diseases.", "Rashidul A. Mahumud conceptualized the study, directed the data analysis and wrote most of the manuscript. Rashidul A. Mahumud performed the statistical analysis under the supervision of Andre M. N. Renzaho, Khorshed Alam and Asaduzzaman Khan. Marufa Sultana, Satyajit Kundu, Md. A. Rahman and Mostafa Kamal wrote portions of the manuscript. Rashidul A. Mahumud, Khorshed Alam, Andre M. N. Renzaho, Sabuj K. Mistry, Joseph K. Kamara, Md. G. Hossain, Mohammad A. Ali and Asaduzzaman Khan contributed to the study design, data interpretation and edited the manuscript. All named authors contributed to critically reviewing the revised initial draft of the manuscript. All authors read and approved the final draft.", "This study has been prepared using a secondary de‐identified and fully anonymized cross‐sectional data set from the HIES conducted during 2016–2017 by the Bangladesh Bureau of Statistics. Therefore, no ethical approval was not required for this study." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "BACKGROUND", "METHODS", "Study design and data sources", "HIES survey sampling and sample size calculation", "DATA COLLECTION", "Study population", "DEFINITIONS OF STUDY VARIABLES", "Outcome measures", "Chronic diseases and comorbid conditions", "Covariates", "STATISTICAL ANALYSIS", "RESULTS", "The distribution of chronic disease and utilization of healthcare services", "Preferences for healthcare services for chronic illnesses", "Correlates of patient preference for chronic disease‐related healthcare services", "DISCUSSION", "Implications for policy and practice", "CONCLUSION", "AUTHOR CONTRIBUTIONS", "CONFLICT OF INTEREST", "ETHICS STATEMENT", "Supporting information" ]
[ "Chronic diseases have become a global challenge, imposing an enormous economic and health burden on society.\n1\n Chronic diseases are defined as health conditions lasting 12 months or more, which require ongoing medical intervention and may result in the limitation of activities of daily living.\n1\n The epidemiological burden of chronic diseases\n2\n and exposure to their risk factors are increasing worldwide,\n3\n particularly in low‐ and middle‐income countries (LMICs), such as Bangladesh.\n4\n, \n5\n, \n6\n Chronic diseases account for around 41 million deaths each year, representing about 71% of all deaths globally.\n7\n The most common chronic diseases include cardiovascular diseases (e.g., coronary heart diseases, stroke and peripheral vascular diseases), diabetes, cancers, chronic obstructive pulmonary disease, mental illness and arthritis\n7\n; approximately 77% of all yearly deaths are related to chronic diseases occur in LMICs, of which 85% occur in the most productive age groups (30–69 years).\n7\n It has been estimated that chronic diseases will account for an accumulated global economic loss of 47 trillion US dollars by 2030, approximately 75% of the global gross domestic product.\n8\n In Bangladesh, an LMIC with a substantial social and economic burden, about 886,000 deaths (i.e., 59% of total deaths) occur due to chronic diseases each year.\n3\n, \n9\n The burden due to chronic diseases has increased in Bangladesh from 43.4% in 2000 to 66.9% in 2015.\n3\n It is anticipated that chronic diseases will exceed the combined burden of communicable, maternal, perinatal and nutrition‐related diseases by 2030 globally, including in Bangladesh.\n10\n, \n11\n\n\nDespite the high burden of chronic diseases, Bangladesh, like many LMIC countries, does not have a national integrated chronic diseases management policy, strategy or action plan.\n9\n, \n11\n For instance, the prevalence of undiagnosed chronic diseases is high, and the proportion of unmanaged chronic diseases is even higher in many LMICs,\n10\n including Bangladesh.\n12\n This highlights the frequent inadequacies in the diagnosis, prevention and management of chronic diseases among the healthcare systems of LMICs.\n9\n, \n10\n Efforts in chronic disease management in Bangladesh continue to be inadequate. Little attention has been given to addressing the contributing behaviours associated with chronic diseases, including unhealthy dietary patterns, lack of physical activity and exposure to factors potentially detrimental to health such as alcohol, drug and tobacco use.\n6\n However, it is possible to counteract the rising prevalence of the chronic disease by implementing effective prevention strategies, population‐based screening, reduction of risk factors, early detection and appropriate treatments.\n10\n, \n11\n If such actions are not taken, the burden of chronic diseases, which is referred to as an emerging prevalence of chronic diseases globally imposing an enormous economic and health burden will likely continue to rise,\n10\n, \n11\n which is alarming especially among the vulnerable and marginalized populations of Bangladesh with limited affordability for health services.\n11\n In addition, the emerging prevalence of chronic disease may also lead to a health system burden in terms of increasing healthcare utilization, treatment costs and chronic disease management.\nIt is well established that chronic diseases are increasingly associated with the over‐utilization of healthcare services and a higher financial burden.\n13\n For example, a previous study documented that a higher number of chronic diseases was linked to an increased number of outpatient visits.\n14\n Therefore, adequate preventative services must be in place to reduce the social‐economic burden of chronic disease, thereby ensuring optimal use of health resources. However, the major challenges to ensure effective prevention and management of chronic diseases include social status, power gradients, racial/ethnic differences, poor accessibility and affordability of healthcare services.\n2\n, \n6\n, \n15\n, \n16\n, \n17\n Previous research has identified several factors associated with healthcare utilization, such as demographic and socioeconomic characteristics, type of healthcare providers and presence of chronic illnesses.\n18\n In addition, patients' preference for healthcare services depends on several factors, including personal preference, disease severity, economic capacity, the reputation of healthcare providers\n18\n as well as affordable costs associated with treatment.\n19\n Furthermore, short travel time to healthcare facilities, effective interactions with healthcare providers,\n19\n, \n20\n respectful service provider attitudes\n21\n and short waiting time\n22\n were positively associated with the patient preference for healthcare services. A recent discrete choice experiment study found that the availability of medicine and transport to the health facility were significant attributes of patient preference for healthcare services.\n23\n Notably, optimum healthcare utilization among chronically ill patients has a significant role in preventing and managing chronic diseases.\nIn Bangladesh, a family spends an average of 11% of their total household budget on health care and half of the population spends 7% of their monthly per capita consumption expenditure on illness.\n24\n Understanding patients' preferences could help medical professionals and healthcare providers restructure the healthcare delivery model and ensure the quality of services. In addition to clinical guidelines, patients' preferences may also provide guidelines for the selection of treatment options. Patient preferences also help to inform clinical decisions where science has not yet been able to provide effective solutions to healthcare problems.\n25\n Therefore, information on patients' preferences for healthcare utilization in terms of healthcare expenditure is critical; without this knowledge, an effective national healthcare policy cannot be formulated.\n26\n Moreover, there is a current lack of evidence on the patients' preferences for healthcare services in chronic disease management in Bangladesh. Since no such study exists, the present study aimed to investigate patients' preference for healthcare services among adult patients in Bangladesh. Understanding patients' preferences are important in designing interventions aimed at reducing the burden of chronic diseases, which undermine economic growth and development and are increasingly becoming the leading cause of death.", "[SUBTITLE] Study design and data sources [SUBSECTION] This study followed an observational, cross‐sectional design. The data was extracted from the most recent nationally representative Household Income and Expenditure Survey (HIES), conducted during 2016–2017 by the Bangladesh Bureau of Statistics (BBS) (Supporting Information: Document 1). The HIES is a cross‐sectional survey conducted every 5 years since 1973 in Bangladesh with the objective of providing national estimates on income, expenditure and consumption, poverty, standard of living, health status and education.\n27\n\n\nIn the HIES survey, a semistructured questionnaire (Supporting Information: Appendix Table A1: HIES 2016–2017 Questionnaire) was used to collect information from the survey participants (adults aged 18 years or above) under nine modules: (1) household information, (2) education, (3) health‐illnesses and injuries, (4) economic activities and wage employment, (5) nonagricultural enterprises, (6) housing, (7) agriculture, (8) other assets and income and (9) consumption (Supporting Information: Appendix Table A1: HIES 2016–2017 Questionnaire). However, our analysis is based on the chronic disease‐related questions included in Module‐3: Health (Illnesses and Injuries) of the HIES (Supporting Information: Appendix Table A2). Therefore, we only used the indicators pertaining to chronic disease and health service utilization along with the sociodemographic characterizes of the participants (Supporting Information: Appendix Table A2). All health‐related information was self‐reported in the HIES. Respondents were asked to prioritize the chronic diseases they were suffering from in order of their importance. We selected the primary disease (i.e., principal diagnosis) based on the patients' experience of diseases in order of their importance. For instance, if one patient was diagnosed with three chronic conditions, they reported three diseases in order of importance (i.e., first, second and third importance). Therefore, a patient's first importance of disease was considered as the primary disease in the present study.\nThis study followed an observational, cross‐sectional design. The data was extracted from the most recent nationally representative Household Income and Expenditure Survey (HIES), conducted during 2016–2017 by the Bangladesh Bureau of Statistics (BBS) (Supporting Information: Document 1). The HIES is a cross‐sectional survey conducted every 5 years since 1973 in Bangladesh with the objective of providing national estimates on income, expenditure and consumption, poverty, standard of living, health status and education.\n27\n\n\nIn the HIES survey, a semistructured questionnaire (Supporting Information: Appendix Table A1: HIES 2016–2017 Questionnaire) was used to collect information from the survey participants (adults aged 18 years or above) under nine modules: (1) household information, (2) education, (3) health‐illnesses and injuries, (4) economic activities and wage employment, (5) nonagricultural enterprises, (6) housing, (7) agriculture, (8) other assets and income and (9) consumption (Supporting Information: Appendix Table A1: HIES 2016–2017 Questionnaire). However, our analysis is based on the chronic disease‐related questions included in Module‐3: Health (Illnesses and Injuries) of the HIES (Supporting Information: Appendix Table A2). Therefore, we only used the indicators pertaining to chronic disease and health service utilization along with the sociodemographic characterizes of the participants (Supporting Information: Appendix Table A2). All health‐related information was self‐reported in the HIES. Respondents were asked to prioritize the chronic diseases they were suffering from in order of their importance. We selected the primary disease (i.e., principal diagnosis) based on the patients' experience of diseases in order of their importance. For instance, if one patient was diagnosed with three chronic conditions, they reported three diseases in order of importance (i.e., first, second and third importance). Therefore, a patient's first importance of disease was considered as the primary disease in the present study.\n[SUBTITLE] HIES survey sampling and sample size calculation [SUBSECTION] The sample frame used in the selection of Primary Sample Units (PSUs) for the HIES 2016–2017 was based on the Census of Population and Housing 2011.\n27\n In this survey, eight administrative divisions (Barisal, Chittagong, Dhaka, Khulna, Mymensingh, Rajshahi, Rangpur and Sylhet) were included and stratified by three basic localities, being rural, urban and metropolitan areas (city corporations). Thus, these should be 8 divisions × 3 localities totalling 24 strata (8 × 3 = 24). However, the sampling frame (Census of Population and Housing 2011) did not contain three administrative divisions (i.e., Rangpur, Barisal and Sylhet). Additionally, the BBS included only four main city corporations (Dhaka, Chittagong, Khulna and Rajshahi) in the city corporation locality. PSUs were randomly selected from 20 strata (eight rural divisions, eight urban divisions and four metropolitan areas) for national representation. As the PSUs of HIES 2016–2017 were allocated at the district level, the sample design includes a total of 132 substrata: 64 rural, 64 urban and 4 metropolitan areas.\n27\n Sample size was calculated using the prevalence rate of the main indicator (poverty rate) or the coefficient of variation of per capita consumption or household consumption, which are the core indicators of the HIES 2016–2017. Each one was treated as a target variable for determining the sample size (Supporting Information: Document 1). The required sample size was calculated for each district and explained elsewhere,\n27\n using the following formula\n\n(1)\nn=Zα2×CVSRS(y¯)r(Y¯)2×DEFF,\nwhere, n was the required sample for allocation to each district to achieve a certain level in the accuracy statistic (r(Y®) = 10% relative standard error desired for the mean total household expenditure estimated at the district level associated with the targeted variable (y®);CVSRS(y®) was the coefficient of variation of the targeted variable [i.e., total household expenditure estimated at the national level] estimated under the assumption of simple random sampling; DEFF was the design effect of the target variable [i.e., the average design effect of the target variable across all districts]; and Zα2 [=1.96] was the critical value of a standard normal distribution with an α% (5%) significance level). Substituting all values in Equation (1), the required sample was 715 households for each district, nonetheless, 720 households were allocated to each district for practical consideration and to facilitate fieldwork and survey implementation management. A stratified, two‐stage cluster sampling technique was used in this survey. In the first stage, a total of 36 PSUs were drawn from each district by applying the probability proportional to size systematic sampling technique, using the number of households in each PSU as the measure of size. The 36 PSUs were randomly selected from rural, urban and city corporation substratum. The total number of PSUs included in the analysis was 2304 (64 districts × 36 PSU per district). In the second stage, 20 households were selected per PSU. Using this sampling technique, 46,076 households (2304 PUSs × 20 households per PSU) were included in the study analysis of HIES 2016–2017 data. Among the selected households, a total of 186,076 individuals were included.\nThe sample frame used in the selection of Primary Sample Units (PSUs) for the HIES 2016–2017 was based on the Census of Population and Housing 2011.\n27\n In this survey, eight administrative divisions (Barisal, Chittagong, Dhaka, Khulna, Mymensingh, Rajshahi, Rangpur and Sylhet) were included and stratified by three basic localities, being rural, urban and metropolitan areas (city corporations). Thus, these should be 8 divisions × 3 localities totalling 24 strata (8 × 3 = 24). However, the sampling frame (Census of Population and Housing 2011) did not contain three administrative divisions (i.e., Rangpur, Barisal and Sylhet). Additionally, the BBS included only four main city corporations (Dhaka, Chittagong, Khulna and Rajshahi) in the city corporation locality. PSUs were randomly selected from 20 strata (eight rural divisions, eight urban divisions and four metropolitan areas) for national representation. As the PSUs of HIES 2016–2017 were allocated at the district level, the sample design includes a total of 132 substrata: 64 rural, 64 urban and 4 metropolitan areas.\n27\n Sample size was calculated using the prevalence rate of the main indicator (poverty rate) or the coefficient of variation of per capita consumption or household consumption, which are the core indicators of the HIES 2016–2017. Each one was treated as a target variable for determining the sample size (Supporting Information: Document 1). The required sample size was calculated for each district and explained elsewhere,\n27\n using the following formula\n\n(1)\nn=Zα2×CVSRS(y¯)r(Y¯)2×DEFF,\nwhere, n was the required sample for allocation to each district to achieve a certain level in the accuracy statistic (r(Y®) = 10% relative standard error desired for the mean total household expenditure estimated at the district level associated with the targeted variable (y®);CVSRS(y®) was the coefficient of variation of the targeted variable [i.e., total household expenditure estimated at the national level] estimated under the assumption of simple random sampling; DEFF was the design effect of the target variable [i.e., the average design effect of the target variable across all districts]; and Zα2 [=1.96] was the critical value of a standard normal distribution with an α% (5%) significance level). Substituting all values in Equation (1), the required sample was 715 households for each district, nonetheless, 720 households were allocated to each district for practical consideration and to facilitate fieldwork and survey implementation management. A stratified, two‐stage cluster sampling technique was used in this survey. In the first stage, a total of 36 PSUs were drawn from each district by applying the probability proportional to size systematic sampling technique, using the number of households in each PSU as the measure of size. The 36 PSUs were randomly selected from rural, urban and city corporation substratum. The total number of PSUs included in the analysis was 2304 (64 districts × 36 PSU per district). In the second stage, 20 households were selected per PSU. Using this sampling technique, 46,076 households (2304 PUSs × 20 households per PSU) were included in the study analysis of HIES 2016–2017 data. Among the selected households, a total of 186,076 individuals were included.", "This study followed an observational, cross‐sectional design. The data was extracted from the most recent nationally representative Household Income and Expenditure Survey (HIES), conducted during 2016–2017 by the Bangladesh Bureau of Statistics (BBS) (Supporting Information: Document 1). The HIES is a cross‐sectional survey conducted every 5 years since 1973 in Bangladesh with the objective of providing national estimates on income, expenditure and consumption, poverty, standard of living, health status and education.\n27\n\n\nIn the HIES survey, a semistructured questionnaire (Supporting Information: Appendix Table A1: HIES 2016–2017 Questionnaire) was used to collect information from the survey participants (adults aged 18 years or above) under nine modules: (1) household information, (2) education, (3) health‐illnesses and injuries, (4) economic activities and wage employment, (5) nonagricultural enterprises, (6) housing, (7) agriculture, (8) other assets and income and (9) consumption (Supporting Information: Appendix Table A1: HIES 2016–2017 Questionnaire). However, our analysis is based on the chronic disease‐related questions included in Module‐3: Health (Illnesses and Injuries) of the HIES (Supporting Information: Appendix Table A2). Therefore, we only used the indicators pertaining to chronic disease and health service utilization along with the sociodemographic characterizes of the participants (Supporting Information: Appendix Table A2). All health‐related information was self‐reported in the HIES. Respondents were asked to prioritize the chronic diseases they were suffering from in order of their importance. We selected the primary disease (i.e., principal diagnosis) based on the patients' experience of diseases in order of their importance. For instance, if one patient was diagnosed with three chronic conditions, they reported three diseases in order of importance (i.e., first, second and third importance). Therefore, a patient's first importance of disease was considered as the primary disease in the present study.", "The sample frame used in the selection of Primary Sample Units (PSUs) for the HIES 2016–2017 was based on the Census of Population and Housing 2011.\n27\n In this survey, eight administrative divisions (Barisal, Chittagong, Dhaka, Khulna, Mymensingh, Rajshahi, Rangpur and Sylhet) were included and stratified by three basic localities, being rural, urban and metropolitan areas (city corporations). Thus, these should be 8 divisions × 3 localities totalling 24 strata (8 × 3 = 24). However, the sampling frame (Census of Population and Housing 2011) did not contain three administrative divisions (i.e., Rangpur, Barisal and Sylhet). Additionally, the BBS included only four main city corporations (Dhaka, Chittagong, Khulna and Rajshahi) in the city corporation locality. PSUs were randomly selected from 20 strata (eight rural divisions, eight urban divisions and four metropolitan areas) for national representation. As the PSUs of HIES 2016–2017 were allocated at the district level, the sample design includes a total of 132 substrata: 64 rural, 64 urban and 4 metropolitan areas.\n27\n Sample size was calculated using the prevalence rate of the main indicator (poverty rate) or the coefficient of variation of per capita consumption or household consumption, which are the core indicators of the HIES 2016–2017. Each one was treated as a target variable for determining the sample size (Supporting Information: Document 1). The required sample size was calculated for each district and explained elsewhere,\n27\n using the following formula\n\n(1)\nn=Zα2×CVSRS(y¯)r(Y¯)2×DEFF,\nwhere, n was the required sample for allocation to each district to achieve a certain level in the accuracy statistic (r(Y®) = 10% relative standard error desired for the mean total household expenditure estimated at the district level associated with the targeted variable (y®);CVSRS(y®) was the coefficient of variation of the targeted variable [i.e., total household expenditure estimated at the national level] estimated under the assumption of simple random sampling; DEFF was the design effect of the target variable [i.e., the average design effect of the target variable across all districts]; and Zα2 [=1.96] was the critical value of a standard normal distribution with an α% (5%) significance level). Substituting all values in Equation (1), the required sample was 715 households for each district, nonetheless, 720 households were allocated to each district for practical consideration and to facilitate fieldwork and survey implementation management. A stratified, two‐stage cluster sampling technique was used in this survey. In the first stage, a total of 36 PSUs were drawn from each district by applying the probability proportional to size systematic sampling technique, using the number of households in each PSU as the measure of size. The 36 PSUs were randomly selected from rural, urban and city corporation substratum. The total number of PSUs included in the analysis was 2304 (64 districts × 36 PSU per district). In the second stage, 20 households were selected per PSU. Using this sampling technique, 46,076 households (2304 PUSs × 20 households per PSU) were included in the study analysis of HIES 2016–2017 data. Among the selected households, a total of 186,076 individuals were included.", "Data collection was accomplished between early April 2016 and late March 2017 through face‐to‐face interviews with the participants. A total of 128 enumeration teams, each comprised of five members (one supervising officer, two interviewers and two female facilitators) who were thoroughly trained, completed the data collection. A multilayered quality control measure was employed to ensure the quality of the collected data. The supervising officer of each team was responsible to verify all the questionnaires completed by the field staff before they were sent to the headquarter. Moreover, Deputy Directors of the District Statistical Offices as well as senior officials from the headquarter, frequently visited the sampled areas to ensure data quality (Supporting Information: Document 1).\nThe survey team formulated an operational definition of each variable used in the questionnaire. For example, while collecting chronic disease‐related information, the enumerators were instructed to ask the respondents, ‘Have you suffered from any chronic illness/disability in the last 12 months or more?’ (if yes); then, participants were asked a second question: ‘What chronic illness/disability are you suffering from?’ with response options: (1) chronic fever, (2) Injuries/disability, (3) chronic heart disease, (4) respiratory diseases/asthma/bronchitis, (5) diarrhoea/dysentery, (6) gastric/ulcer, (7) blood pressure, (8) arthritis/rheumatism, (9) skin problem, (10) diabetes, (11) cancer, (12) kidney diseases, (13) liver diseases, (14) mental health, (15) paralysis, (16) ear/ENT problem, (17) eye problem or (18) other (specify) (Supporting Information: Appendix Table A1: HIES 2016–2017 questionnaire). Participants responded based on their disease diagnosis, experiences, symptoms of illness and course of treatment. To get valid information, the enumerators also probed where necessary, asked the respondents to show any relevant documents such as prescriptions and test reports, or explained to the respondents about chronic diseases using various case scenarios as outlined in the survey guidelines and instructions.\n[SUBTITLE] Study population [SUBSECTION] All selected household members were included in the survey. The participants were selected based on the HIES 2016–2017 survey protocol and following the same inclusion criteria: (1) an individual who had suffered from any chronic disease for the last 12 months or more and (2) patients who received any treatment due to chronic disease in the last 30 days. Based on these inclusion criteria, a total of 10,385 patients were selected for the present analysis (Figure 1).\nDistribution of study participants\nAll selected household members were included in the survey. The participants were selected based on the HIES 2016–2017 survey protocol and following the same inclusion criteria: (1) an individual who had suffered from any chronic disease for the last 12 months or more and (2) patients who received any treatment due to chronic disease in the last 30 days. Based on these inclusion criteria, a total of 10,385 patients were selected for the present analysis (Figure 1).\nDistribution of study participants", "All selected household members were included in the survey. The participants were selected based on the HIES 2016–2017 survey protocol and following the same inclusion criteria: (1) an individual who had suffered from any chronic disease for the last 12 months or more and (2) patients who received any treatment due to chronic disease in the last 30 days. Based on these inclusion criteria, a total of 10,385 patients were selected for the present analysis (Figure 1).\nDistribution of study participants", "[SUBTITLE] Outcome measures [SUBSECTION] Patients' preference for chronic disease healthcare utilization was the primary outcome measure for this study. Participants responded to questions that asked them about their preference for selecting a healthcare provider: ‘why did you choose this provider?’ Response options were recoded as the quality of healthcare services, availability of medical doctors (e.g., availability of male or female doctors in a health facility), affordable healthcare services (i.e., acceptable costs), short distance to health facility (i.e., nearby) and others (e.g., referred by other providers).\nPatients' preference for chronic disease healthcare utilization was the primary outcome measure for this study. Participants responded to questions that asked them about their preference for selecting a healthcare provider: ‘why did you choose this provider?’ Response options were recoded as the quality of healthcare services, availability of medical doctors (e.g., availability of male or female doctors in a health facility), affordable healthcare services (i.e., acceptable costs), short distance to health facility (i.e., nearby) and others (e.g., referred by other providers).\n[SUBTITLE] Chronic diseases and comorbid conditions [SUBSECTION] Participants were asked about chronic illness, ‘have you suffered from any chronic illness/disability in the last 12 months or more?’ The prevalence of chronic diseases was assessed based on this question. Individuals who suffered from chronic disease for the last 12 months (or more) and received any treatment due to chronic diseases were the only population considered in this study. The study population was diagnosed with chronic illnesses and conditions such as chronic heart disease, respiratory diseases, chronic gastric or ulcers, high blood pressure, arthritis or rheumatism, diabetes, chronic fever and other diseases. There is no gold‐standard method for measuring comorbidity status among patients with chronic diseases.\n28\n A previous review study identified that 21 separate approaches were executed to measure comorbidity status.\n28\n The selection of the approach depends on the study research questions, study design, data availability and population studied. The most straightforward approach to measuring comorbidity status is to investigate the distribution of individual comorbid conditions and to treat them independently and/or to combine them by summing the total number of conditions.\n29\n, \n30\n A single condition count approach was performed to measure comorbidity status in this study. The count of chronic health condition(s) was measured for each patient based on the number of disease exposures and who had been prescribed medication for their illness. It was counted as multiple responses if the patients had multiple chronic conditions. In addition, the principal diagnosis was defined based on patients diagnosed based on their reported first importance. Chronic comorbid conditions (principal diagnosis plus one, two or three or more comorbid conditions) were assessed in this study.\nParticipants were asked about chronic illness, ‘have you suffered from any chronic illness/disability in the last 12 months or more?’ The prevalence of chronic diseases was assessed based on this question. Individuals who suffered from chronic disease for the last 12 months (or more) and received any treatment due to chronic diseases were the only population considered in this study. The study population was diagnosed with chronic illnesses and conditions such as chronic heart disease, respiratory diseases, chronic gastric or ulcers, high blood pressure, arthritis or rheumatism, diabetes, chronic fever and other diseases. There is no gold‐standard method for measuring comorbidity status among patients with chronic diseases.\n28\n A previous review study identified that 21 separate approaches were executed to measure comorbidity status.\n28\n The selection of the approach depends on the study research questions, study design, data availability and population studied. The most straightforward approach to measuring comorbidity status is to investigate the distribution of individual comorbid conditions and to treat them independently and/or to combine them by summing the total number of conditions.\n29\n, \n30\n A single condition count approach was performed to measure comorbidity status in this study. The count of chronic health condition(s) was measured for each patient based on the number of disease exposures and who had been prescribed medication for their illness. It was counted as multiple responses if the patients had multiple chronic conditions. In addition, the principal diagnosis was defined based on patients diagnosed based on their reported first importance. Chronic comorbid conditions (principal diagnosis plus one, two or three or more comorbid conditions) were assessed in this study.\n[SUBTITLE] Covariates [SUBSECTION] Based on the ongoing literature and the authors' own expertise, we selected variables to address the study objective, including healthcare service‐related factors (e.g., type of healthcare service‐inpatient or outpatient, type of health facilities, waiting time for receiving healthcare services, out‐of‐pocket payment and location of consulted healthcare provider) and patients' sociodemographics factors (e.g., gender, age, marital status, education and employment) (Supporting Information: Appendix Table A2).\nBased on the ongoing literature and the authors' own expertise, we selected variables to address the study objective, including healthcare service‐related factors (e.g., type of healthcare service‐inpatient or outpatient, type of health facilities, waiting time for receiving healthcare services, out‐of‐pocket payment and location of consulted healthcare provider) and patients' sociodemographics factors (e.g., gender, age, marital status, education and employment) (Supporting Information: Appendix Table A2).", "Patients' preference for chronic disease healthcare utilization was the primary outcome measure for this study. Participants responded to questions that asked them about their preference for selecting a healthcare provider: ‘why did you choose this provider?’ Response options were recoded as the quality of healthcare services, availability of medical doctors (e.g., availability of male or female doctors in a health facility), affordable healthcare services (i.e., acceptable costs), short distance to health facility (i.e., nearby) and others (e.g., referred by other providers).", "Participants were asked about chronic illness, ‘have you suffered from any chronic illness/disability in the last 12 months or more?’ The prevalence of chronic diseases was assessed based on this question. Individuals who suffered from chronic disease for the last 12 months (or more) and received any treatment due to chronic diseases were the only population considered in this study. The study population was diagnosed with chronic illnesses and conditions such as chronic heart disease, respiratory diseases, chronic gastric or ulcers, high blood pressure, arthritis or rheumatism, diabetes, chronic fever and other diseases. There is no gold‐standard method for measuring comorbidity status among patients with chronic diseases.\n28\n A previous review study identified that 21 separate approaches were executed to measure comorbidity status.\n28\n The selection of the approach depends on the study research questions, study design, data availability and population studied. The most straightforward approach to measuring comorbidity status is to investigate the distribution of individual comorbid conditions and to treat them independently and/or to combine them by summing the total number of conditions.\n29\n, \n30\n A single condition count approach was performed to measure comorbidity status in this study. The count of chronic health condition(s) was measured for each patient based on the number of disease exposures and who had been prescribed medication for their illness. It was counted as multiple responses if the patients had multiple chronic conditions. In addition, the principal diagnosis was defined based on patients diagnosed based on their reported first importance. Chronic comorbid conditions (principal diagnosis plus one, two or three or more comorbid conditions) were assessed in this study.", "Based on the ongoing literature and the authors' own expertise, we selected variables to address the study objective, including healthcare service‐related factors (e.g., type of healthcare service‐inpatient or outpatient, type of health facilities, waiting time for receiving healthcare services, out‐of‐pocket payment and location of consulted healthcare provider) and patients' sociodemographics factors (e.g., gender, age, marital status, education and employment) (Supporting Information: Appendix Table A2).", "The adjusted multinomial logistic regression model was used to identify the potential factors that had an association with the patient's preference for healthcare service. The dependent variable (chronic illness patient's preference for healthcare services) was characterized as a categorical measure in the regression model. An unadjusted analysis was performed using each of the explanatory variables for the following reasons: (1) primary screening of the selection of qualified variables, which were added in the adjusted model, (2) the χ\n2 tests (or one‐way analysis where appropriate) were used to find the association between outcome and explanatory variables. However, the majority of the explanatory variables were categorical with two or more labels; therefore, an unadjusted analysis was performed to find the association between the outcome variable and different categories of explanatory variables. The explanatory variables were included in the adjusted model only if any label of the predictor was significant at a 5% or less risk level in the unadjusted model, which in turn was used to adjust for the associations of other potential confounders. For the explanatory variables, the category found to be least at risk of having patients' preferences for healthcare services related to chronic illness in the analysis was considered the reference category for constructing the relative risk ratio (RRR). Statistical significance was considered at a 5% risk level. All data analyses were undertaken using the statistical software Stata/SE 14 (StataCorp).", "Table 1 shows the participant's characteristics. Of the 10,385 patients with chronic diseases, approximately 50% were female, 51% were married and 70% were unemployed. The majority of the patients were aged between 18 and 45 (81%) years, and around one‐third (37%) had no formal education.\nDistribution of participant's characteristics\n\nNote: p value was derived using χ\n2 test or one‐way analysis of variance where appropriate.\nAbbreviations: BDT, Bangladeshi Taka; CI, confidence interval; SD, standard deviation.\n[SUBTITLE] The distribution of chronic disease and utilization of healthcare services [SUBSECTION] The most prevalent chronic diseases reported by the patients included gastric/ulcer (16%) followed by arthritis/rheumatism (14%) (Table 1). Most of the patients (65%) reported one diagnosed chronic condition, while approximately 24% of the patients had two, and 11% had three or more chronic conditions. Most of the patients (91%) reported utilizing outpatient healthcare services in the past 30 days. A high proportion of the patients visited general practitioner clinics (~40%), followed by pharmacy/dispensary (24%) and public hospitals (20%) for healthcare services. Furthermore, approximately 53% of the patients received services at rural health facilities. The average out‐of‐pocket healthcare expenditure for chronic illness in the last 30 days was 3848 BDT (~47.40 USD; 2017 price year).\nThe most prevalent chronic diseases reported by the patients included gastric/ulcer (16%) followed by arthritis/rheumatism (14%) (Table 1). Most of the patients (65%) reported one diagnosed chronic condition, while approximately 24% of the patients had two, and 11% had three or more chronic conditions. Most of the patients (91%) reported utilizing outpatient healthcare services in the past 30 days. A high proportion of the patients visited general practitioner clinics (~40%), followed by pharmacy/dispensary (24%) and public hospitals (20%) for healthcare services. Furthermore, approximately 53% of the patients received services at rural health facilities. The average out‐of‐pocket healthcare expenditure for chronic illness in the last 30 days was 3848 BDT (~47.40 USD; 2017 price year).\n[SUBTITLE] Preferences for healthcare services for chronic illnesses [SUBSECTION] Approximately 30% of patients reported that they preferred quality healthcare services, whereas 28% preferred a short distance to the health facility. Furthermore, 22% of the patients preferred affordable healthcare costs as the main driver, and 11% expressed a preference for the doctor's availability. However, the scenario varied among different chronic illnesses and healthcare services (Table 1). For example, among the patients who received healthcare services from a pharmacy or dispensary, 49% preferred short distances to health facilities, and 32% reported their preference for healthcare affordability.\nApproximately 30% of patients reported that they preferred quality healthcare services, whereas 28% preferred a short distance to the health facility. Furthermore, 22% of the patients preferred affordable healthcare costs as the main driver, and 11% expressed a preference for the doctor's availability. However, the scenario varied among different chronic illnesses and healthcare services (Table 1). For example, among the patients who received healthcare services from a pharmacy or dispensary, 49% preferred short distances to health facilities, and 32% reported their preference for healthcare affordability.\n[SUBTITLE] Correlates of patient preference for chronic disease‐related healthcare services [SUBSECTION] Table 2 highlights the factors that influence a patient's choice of healthcare services. Among patients with heart disease, quality of healthcare services was 29% more preferred than affordability (RRR = 0.71; 95% confidence interval [CI]: 0.56–0.90; p < .001).\nInfluencing factors on patient's preference of healthcare services\n\nNote: ***, ** and * denoted significance level at 0.1%, 1% and 5%, respectively.\nAbbreviations: CI, confidence interval; ref, reference group; RRR, relative risk ratio.\nSimilarly, diabetic patients exhibited a 38% higher preference for quality rather than affordability of healthcare services (RRR = 0.62; 0.47–0.81; p < .001). However, patients with three or more chronic comorbid conditions were 1.39 times more likely to prefer affordability over the quality of healthcare services (RRR = 1.39; 1.15–1.68; p < .001). Patients who received healthcare services from a public facility reported a higher preference for the availability of medical doctors or consultants (RRR = 2.93; 1.70–5.04; p < .001) than those receiving health care from private service providers. In addition, patients who received healthcare services from pharmacies or dispensaries were significantly more likely to prefer a short distance to a healthcare facility (RRR = 6.99; 4.80–9.86; p < .001) or affordability of healthcare services (RRR = 3.13; 2.25–4.36; p < .001), rather than the quality of the healthcare services and availability of doctors.\nTable 2 highlights the factors that influence a patient's choice of healthcare services. Among patients with heart disease, quality of healthcare services was 29% more preferred than affordability (RRR = 0.71; 95% confidence interval [CI]: 0.56–0.90; p < .001).\nInfluencing factors on patient's preference of healthcare services\n\nNote: ***, ** and * denoted significance level at 0.1%, 1% and 5%, respectively.\nAbbreviations: CI, confidence interval; ref, reference group; RRR, relative risk ratio.\nSimilarly, diabetic patients exhibited a 38% higher preference for quality rather than affordability of healthcare services (RRR = 0.62; 0.47–0.81; p < .001). However, patients with three or more chronic comorbid conditions were 1.39 times more likely to prefer affordability over the quality of healthcare services (RRR = 1.39; 1.15–1.68; p < .001). Patients who received healthcare services from a public facility reported a higher preference for the availability of medical doctors or consultants (RRR = 2.93; 1.70–5.04; p < .001) than those receiving health care from private service providers. In addition, patients who received healthcare services from pharmacies or dispensaries were significantly more likely to prefer a short distance to a healthcare facility (RRR = 6.99; 4.80–9.86; p < .001) or affordability of healthcare services (RRR = 3.13; 2.25–4.36; p < .001), rather than the quality of the healthcare services and availability of doctors.", "The most prevalent chronic diseases reported by the patients included gastric/ulcer (16%) followed by arthritis/rheumatism (14%) (Table 1). Most of the patients (65%) reported one diagnosed chronic condition, while approximately 24% of the patients had two, and 11% had three or more chronic conditions. Most of the patients (91%) reported utilizing outpatient healthcare services in the past 30 days. A high proportion of the patients visited general practitioner clinics (~40%), followed by pharmacy/dispensary (24%) and public hospitals (20%) for healthcare services. Furthermore, approximately 53% of the patients received services at rural health facilities. The average out‐of‐pocket healthcare expenditure for chronic illness in the last 30 days was 3848 BDT (~47.40 USD; 2017 price year).", "Approximately 30% of patients reported that they preferred quality healthcare services, whereas 28% preferred a short distance to the health facility. Furthermore, 22% of the patients preferred affordable healthcare costs as the main driver, and 11% expressed a preference for the doctor's availability. However, the scenario varied among different chronic illnesses and healthcare services (Table 1). For example, among the patients who received healthcare services from a pharmacy or dispensary, 49% preferred short distances to health facilities, and 32% reported their preference for healthcare affordability.", "Table 2 highlights the factors that influence a patient's choice of healthcare services. Among patients with heart disease, quality of healthcare services was 29% more preferred than affordability (RRR = 0.71; 95% confidence interval [CI]: 0.56–0.90; p < .001).\nInfluencing factors on patient's preference of healthcare services\n\nNote: ***, ** and * denoted significance level at 0.1%, 1% and 5%, respectively.\nAbbreviations: CI, confidence interval; ref, reference group; RRR, relative risk ratio.\nSimilarly, diabetic patients exhibited a 38% higher preference for quality rather than affordability of healthcare services (RRR = 0.62; 0.47–0.81; p < .001). However, patients with three or more chronic comorbid conditions were 1.39 times more likely to prefer affordability over the quality of healthcare services (RRR = 1.39; 1.15–1.68; p < .001). Patients who received healthcare services from a public facility reported a higher preference for the availability of medical doctors or consultants (RRR = 2.93; 1.70–5.04; p < .001) than those receiving health care from private service providers. In addition, patients who received healthcare services from pharmacies or dispensaries were significantly more likely to prefer a short distance to a healthcare facility (RRR = 6.99; 4.80–9.86; p < .001) or affordability of healthcare services (RRR = 3.13; 2.25–4.36; p < .001), rather than the quality of the healthcare services and availability of doctors.", "Chronic diseases among adults are becoming a significant health concern in many LMICs, including Bangladesh. This is the first study to focus on patient preferences for healthcare services for chronic disease management, using a recent nationally representative HIES. This study provided evidence of patient preference for the utilization of healthcare services in chronic disease management. The top four dimensions of patient preference for health care in order of preference were quality of treatment (30.3%), short distance to health facilities (27.6%), affordability of health care (21.7%) and availability of doctors (11.0%). The severity of the disease and the characteristics of healthcare providers were the most important contributing factors in patients' preferences and decisions to seek healthcare services.\nThis study showed that patients with heart disease were more likely to prefer quality health care than healthcare affordability or a short distance to a health facility. This is in agreement with previous studies, which have also documented that chronic heart disease patients are more likely to prefer quality healthcare services.\n31\n, \n32\n, \n33\n Several attributes might influence a patient's preference for quality healthcare services. For instance, heart disease may be associated with both acute episodes and high levels of long‐term adverse events (e.g., mortality and disability).\n34\n, \n35\n, \n36\n Such patients may require curative care as a component of a continuous, coordinated care model, covering both primary and postacute hospital care.\n34\n Moreover, due to the high disease burden and prolonged treatment, continuous use of healthcare resources (e.g., specialist consultation, diagnostic and medicine) may lead to a higher economic burden for households, individuals and society.\n35\n, \n36\n, \n37\n From a healthcare service providers' perspective, examining patients' experience and satisfaction with healthcare services could identify whether services are of an acceptable standard or highlight areas for potential quality improvement.\n38\n Existing research has revealed that a positive experience with healthcare professionals or other medical staff is directly related to patient preference for healthcare quality during hospitalization or course of treatment.\n39\n Other studies have indicated the importance of having confidence in the expertise and attentiveness of doctors or nurses,\n40\n, \n41\n healthcare provider's interpersonal communication skills and behaviours,\n42\n although shorter waiting times have also been mentioned/raised.\n43\n\n\nIn the present study, one of the most influential aspects of a patient's healthcare‐seeking behaviour was the healthcare provider's location; the shorter the distance to the healthcare facility the more likely the patient uptake of the healthcare services. It is plausible that patients may be unwilling or unable to travel long distances to access medical expertise or treatment, particularly if the nature of the chronic illness requires frequent appointments. This finding is supported by previous studies which reported that distance to healthcare facilities was a potential barrier to accessing healthcare services in LMICs.\n44\n, \n45\n Patients' educational level, employment status and chronic comorbid conditions were also identified as significant determinants influencing their healthcare‐seeking preferences. This may be due to patients with chronic illnesses being more likely to prioritize treatment, management and quality of life over other life choices they may face.\n46\n In addition, patients with multiple chronic conditions may require medical care from several healthcare providers across various locations throughout the year. This may be attributed to seeking quality care from experienced or specialist medical professionals, and superior diagnosis and treatment facilities, which has also been identified elsewhere.\n47\n\n\nAlthough this is the first study to investigate patient preference for healthcare services to manage chronic diseases among adults in Bangladesh, the extension and transferability of our findings to contexts beyond the study population should be handled with caution because of our study limitations. For instance, due to the cross‐sectional nature of the study, causality cannot be inferred. Furthermore, these findings may be subjected to some level of bias as data on the main variables of interest were self‐reported (i.e., illness, utilization and expenditure), thereby risking recall bias. However, the relatively short recall period (i.e., the last 30 days) of the household income and expenditure survey strives to reduce this potential bias.\n[SUBTITLE] Implications for policy and practice [SUBSECTION] Our study highlights the significance of patient preferences for healthcare utilization. This study provides timely findings to address health inequities linked to sociocultural and economic factors in Bangladesh. Understanding patients' preferences in chronic disease management is critical to achieving the Sustainable Development Goal (SDG) target 3.4, which focuses solely on reducing premature mortality from noncommunicable diseases by a third by 2030 relative when compared to 2015 as a baseline. Bangladesh is a signatory to the SDGs and the Colombo declaration (strengthening health systems to accelerate delivery of NCD‐related services at the primary healthcare level),\n48\n providing evidence that informs culturally competent NCD prevention and treatment approaches through tailored and responsive health financing, and expenditure policies will contribute significantly to achieving the SDG target 3.4. However, progress in implementing strategies to meet international targets related to NCDs remains slow.\n9\n, \n11\n, \n49\n\n\nAdditionally, Bangladesh lacks a national surveillance programme focused on chronic diseases and does not have any integrated community public health programme that regularly monitors chronic diseases.\n11\n An established disease management plan should consider the adequacy, accessibility, affordability and quality of services.\n48\n Most importantly, the factors influencing patient preferences for healthcare services may not have been considered when developing the chronic disease management policy.\n11\n Therefore, policymakers and public health practitioners should consider patient preferences regarding healthcare utilization in managing chronic diseases.\nOur study highlights the significance of patient preferences for healthcare utilization. This study provides timely findings to address health inequities linked to sociocultural and economic factors in Bangladesh. Understanding patients' preferences in chronic disease management is critical to achieving the Sustainable Development Goal (SDG) target 3.4, which focuses solely on reducing premature mortality from noncommunicable diseases by a third by 2030 relative when compared to 2015 as a baseline. Bangladesh is a signatory to the SDGs and the Colombo declaration (strengthening health systems to accelerate delivery of NCD‐related services at the primary healthcare level),\n48\n providing evidence that informs culturally competent NCD prevention and treatment approaches through tailored and responsive health financing, and expenditure policies will contribute significantly to achieving the SDG target 3.4. However, progress in implementing strategies to meet international targets related to NCDs remains slow.\n9\n, \n11\n, \n49\n\n\nAdditionally, Bangladesh lacks a national surveillance programme focused on chronic diseases and does not have any integrated community public health programme that regularly monitors chronic diseases.\n11\n An established disease management plan should consider the adequacy, accessibility, affordability and quality of services.\n48\n Most importantly, the factors influencing patient preferences for healthcare services may not have been considered when developing the chronic disease management policy.\n11\n Therefore, policymakers and public health practitioners should consider patient preferences regarding healthcare utilization in managing chronic diseases.", "Our study highlights the significance of patient preferences for healthcare utilization. This study provides timely findings to address health inequities linked to sociocultural and economic factors in Bangladesh. Understanding patients' preferences in chronic disease management is critical to achieving the Sustainable Development Goal (SDG) target 3.4, which focuses solely on reducing premature mortality from noncommunicable diseases by a third by 2030 relative when compared to 2015 as a baseline. Bangladesh is a signatory to the SDGs and the Colombo declaration (strengthening health systems to accelerate delivery of NCD‐related services at the primary healthcare level),\n48\n providing evidence that informs culturally competent NCD prevention and treatment approaches through tailored and responsive health financing, and expenditure policies will contribute significantly to achieving the SDG target 3.4. However, progress in implementing strategies to meet international targets related to NCDs remains slow.\n9\n, \n11\n, \n49\n\n\nAdditionally, Bangladesh lacks a national surveillance programme focused on chronic diseases and does not have any integrated community public health programme that regularly monitors chronic diseases.\n11\n An established disease management plan should consider the adequacy, accessibility, affordability and quality of services.\n48\n Most importantly, the factors influencing patient preferences for healthcare services may not have been considered when developing the chronic disease management policy.\n11\n Therefore, policymakers and public health practitioners should consider patient preferences regarding healthcare utilization in managing chronic diseases.", "Our study findings highlighted that patient preferences for healthcare services in chronic disease management were significantly associated with disease severity and healthcare providers' attributes. Therefore, policymakers and public health practitioners should consider patient preferences for managing chronic conditions within the national strategic frameworks to improve service provision, equitable distribution and uptake of the services.", "Rashidul A. Mahumud conceptualized the study, directed the data analysis and wrote most of the manuscript. Rashidul A. Mahumud performed the statistical analysis under the supervision of Andre M. N. Renzaho, Khorshed Alam and Asaduzzaman Khan. Marufa Sultana, Satyajit Kundu, Md. A. Rahman and Mostafa Kamal wrote portions of the manuscript. Rashidul A. Mahumud, Khorshed Alam, Andre M. N. Renzaho, Sabuj K. Mistry, Joseph K. Kamara, Md. G. Hossain, Mohammad A. Ali and Asaduzzaman Khan contributed to the study design, data interpretation and edited the manuscript. All named authors contributed to critically reviewing the revised initial draft of the manuscript. All authors read and approved the final draft.", "The authors declare no conflict of interest.", "This study has been prepared using a secondary de‐identified and fully anonymized cross‐sectional data set from the HIES conducted during 2016–2017 by the Bangladesh Bureau of Statistics. Therefore, no ethical approval was not required for this study.", "Supporting information.\nClick here for additional data file.\nSupporting information.\nClick here for additional data file.\nSupporting information.\nClick here for additional data file." ]
[ "background", "methods", null, null, null, null, null, null, null, null, null, "results", null, null, null, "discussion", null, "conclusions", null, "COI-statement", null, "supplementary-material" ]
[ "Bangladesh", "chronic diseases", "healthcare services", "patient preferences" ]
Loss of tetraspanin-7 expression reduces pancreatic β-cell exocytosis Ca
36264270
Tetraspanin-7 (Tspan7) is an islet autoantigen involved in autoimmune type 1 diabetes and known to regulate β-cell L-type Ca2+ channel activity. However, the role of Tspan7 in pancreatic β-cell function is not yet fully understood.
BACKGROUND
Histological analyses were conducted using immunostaining. Whole-body metabolism was tested using glucose tolerance test. Islet hormone secretion was quantified using static batch incubation or dynamic perifusion. β-cell transmembrane currents, electrical activity and exocytosis were measured using whole-cell patch-clamping and capacitance measurements. Gene expression was studied using mRNA-sequencing and quantitative PCR.
METHODS
Tspan7 is expressed in insulin-containing granules of pancreatic β-cells and glucagon-producing α-cells. Tspan7 knockout mice (Tspan7y/- mouse) exhibit reduced body weight and ad libitum plasma glucose but normal glucose tolerance. Tspan7y/- islets have normal insulin content and glucose- or tolbutamide-stimulated insulin secretion. Depolarisation-triggered Ca2+ current was enhanced in Tspan7y/- β-cells, but β-cell electrical activity and depolarisation-evoked exocytosis were unchanged suggesting that exocytosis was less sensitive to Ca2+ . TSPAN7 knockdown (KD) in human pseudo-islets led to a significant reduction in insulin secretion stimulated by 20 mM K+ . Transcriptomic analyses show that TSPAN7 KD in human pseudo-islets correlated with changes in genes involved in hormone secretion, apoptosis and ER stress. Consistent with rodent β-cells, exocytotic Ca2+ sensitivity was reduced in a human β-cell line (EndoC-βH1) following Tspan7 KD.
RESULTS
Tspan7 is involved in the regulation of Ca2+ -dependent exocytosis in β-cells. Its function is more significant in human β-cells than their rodent counterparts.
CONCLUSION
[ "Animals", "Humans", "Mice", "Exocytosis", "Glucose", "Insulin", "Insulin-Secreting Cells", "Islets of Langerhans", "Tetraspanins" ]
9828109
null
null
METHODS
[SUBTITLE] Animals and isolation of pancreatic islets [SUBSECTION] All animal experiments were conducted in accordance with the UK Animals Scientific Procedures Act (1986) and the University of Oxford ethical guidelines. Mouse islets were isolated as previously described. 14 , 15 Tspan7 knockout mice (Tspan7y/−) 9 were gifts from Dr Luca Murru (CNR Institute of Neuroscience, Milano, Italy). Hormone secretion studies and the electrophysiology experiments were performed on islets isolated from Tspan7y/− mice and their wildtype littermates (control). All animal experiments were conducted in accordance with the UK Animals Scientific Procedures Act (1986) and the University of Oxford ethical guidelines. Mouse islets were isolated as previously described. 14 , 15 Tspan7 knockout mice (Tspan7y/−) 9 were gifts from Dr Luca Murru (CNR Institute of Neuroscience, Milano, Italy). Hormone secretion studies and the electrophysiology experiments were performed on islets isolated from Tspan7y/− mice and their wildtype littermates (control). [SUBTITLE] shRNA [SUBSECTION] Adenoviruses expressing shRNA for silencing of human TSPAN7 (Ad‐ShTSPAN7, SKU# shADV‐226,651) or scrambled shRNA control (Ad‐Scramble, SKU# 1122) together with eGFP under a CMV promoter were purchased from Vector Biolabs. Adenoviruses expressing shRNA for silencing of human TSPAN7 (Ad‐ShTSPAN7, SKU# shADV‐226,651) or scrambled shRNA control (Ad‐Scramble, SKU# 1122) together with eGFP under a CMV promoter were purchased from Vector Biolabs. [SUBTITLE] Human pseudo‐islets and EndoC‐βH1 cell line [SUBSECTION] Donor organs were obtained with written consent and research ethics approval at the University of Alberta, and human islets were isolated as previous described 16 at the University of Alberta. All human islet information is listed in Table S1. Islets were dispersed into a single‐cell suspension using trypsin (TryLE Express, Gibco) and transduced with Ad‐ShTSPAN7 or Ad‐Scramble before forming human pseudo‐islets using centrifugal‐forced aggregation as previously described. 17 EndoC‐βH1 was provided by Endocell and cultured as previously described. 18 Donor organs were obtained with written consent and research ethics approval at the University of Alberta, and human islets were isolated as previous described 16 at the University of Alberta. All human islet information is listed in Table S1. Islets were dispersed into a single‐cell suspension using trypsin (TryLE Express, Gibco) and transduced with Ad‐ShTSPAN7 or Ad‐Scramble before forming human pseudo‐islets using centrifugal‐forced aggregation as previously described. 17 EndoC‐βH1 was provided by Endocell and cultured as previously described. 18 [SUBTITLE] Insulin secretion [SUBSECTION] Insulin secretion was measured in static incubations as previously described. 15 Briefly, groups of 10–20 isolated islets (number depended on islet availability) were pre‐incubated for 30 min at 37°C in Krebs‐Ringer buffer (KRB) consisting of (mM): 140 NaCl, 3.6 KCl, 0.5 MgSO4, 2.6 CaCl2, 0.5 NaH2PO4, 2 NaHCO3, 1 glucose and 5 HEPES (pH = 7.4 with NaOH), before being incubated for 60 min at 37°C in 1 ml of KRB supplemented as indicated in the legends. Immediately after incubation, an aliquot of the medium was removed for determination of insulin concentration using an ELISA kit (Mercodia, Sweden). For dynamic insulin secretion measurements, groups of 50 pseudo‐islets were perifused with KRB containing 1 or 16.7 mM glucose with or without 20 mM K+ (as indicated) at the rate of 100 μl/min, using a perifusion system (Biorep, FL, USA). Perfusate was collected every 2 min and insulin concentration was determined using the Alpco stellux human ELISA kits (Salem, NH). The perifusion experiments were conducted at the University of Alberta and the mRNAs of pseudo‐islets were then extracted using TRIzol (ThermoFisher) before shipped to Oxford for transcriptomic study. EndoC‐βH1 cells were sequentially exposed for 20 min to 1 and 20 mM glucose containing KRB. Supernatants were collected at the end of each incubation and insulin cellular contents were extracted using acid ethanol. Insulin concentrations were determined using an ELISA kit (Mercodia, Sweden). Insulin secretion was measured in static incubations as previously described. 15 Briefly, groups of 10–20 isolated islets (number depended on islet availability) were pre‐incubated for 30 min at 37°C in Krebs‐Ringer buffer (KRB) consisting of (mM): 140 NaCl, 3.6 KCl, 0.5 MgSO4, 2.6 CaCl2, 0.5 NaH2PO4, 2 NaHCO3, 1 glucose and 5 HEPES (pH = 7.4 with NaOH), before being incubated for 60 min at 37°C in 1 ml of KRB supplemented as indicated in the legends. Immediately after incubation, an aliquot of the medium was removed for determination of insulin concentration using an ELISA kit (Mercodia, Sweden). For dynamic insulin secretion measurements, groups of 50 pseudo‐islets were perifused with KRB containing 1 or 16.7 mM glucose with or without 20 mM K+ (as indicated) at the rate of 100 μl/min, using a perifusion system (Biorep, FL, USA). Perfusate was collected every 2 min and insulin concentration was determined using the Alpco stellux human ELISA kits (Salem, NH). The perifusion experiments were conducted at the University of Alberta and the mRNAs of pseudo‐islets were then extracted using TRIzol (ThermoFisher) before shipped to Oxford for transcriptomic study. EndoC‐βH1 cells were sequentially exposed for 20 min to 1 and 20 mM glucose containing KRB. Supernatants were collected at the end of each incubation and insulin cellular contents were extracted using acid ethanol. Insulin concentrations were determined using an ELISA kit (Mercodia, Sweden). [SUBTITLE] Electrophysiology [SUBSECTION] Electrophysiological measurements were performed using an EPC‐10 patch‐clamp amplifier and Pulse software (version 8.80, HEKA Electronics, Germany). Electrical activity and KATP‐channel conductance were measured using perforated patch‐clamping technique as previously described 19 ; membrane currents and changes in cell capacitance were recorded using voltage clamp in combination with capacitance measurements, as described in. 14 β‐cells were identified by electrophysiological fingerprinting. 20 Electrophysiological measurements were performed using an EPC‐10 patch‐clamp amplifier and Pulse software (version 8.80, HEKA Electronics, Germany). Electrical activity and KATP‐channel conductance were measured using perforated patch‐clamping technique as previously described 19 ; membrane currents and changes in cell capacitance were recorded using voltage clamp in combination with capacitance measurements, as described in. 14 β‐cells were identified by electrophysiological fingerprinting. 20 [SUBTITLE] Histology [SUBSECTION] Sections of formalin‐fixed paraffin‐embedded tissue were de‐waxed and subjected to epitope retrieval in a microwave pressure cooker in 10 mM citric acid pH 6.0, 0.05% Tween 20. After blocking with 2.5% normal horse serum, sections were incubated overnight (4°C) with rabbit anti‐Tspan7 antibody (Anti‐TM4SF2, 1:50, Sigma‐Aldrich). Antibody labelling was detected with the VectaFluor™ Excel Amplified Anti‐Rabbit IgG, DyLight™ 488 Antibody Kit (Vector Laboratories). Slides were counterstained with guinea pig anti‐insulin (1:500, Dako) and mouse anti‐glucagon (1:500, Sigma) antibodies followed by labelling with goat anti‐guinea pig‐Alexa 633 (1:100) and goat anti‐mouse Alexa 555 (1:100) secondary antibodies (ThermoFisher). Slides were visualised by confocal microscopy. For electron microscopy, isolated human islets were fixed in 2.5% glutaraldehyde in PBS, postfixed in 1% OsO4, dehydrated and embedded in Spurr's resin. Sections were cut onto nickel grids before being immunolabelled using anti‐TM4SF2 (1:20) and protein A gold particles (15 nm, Biocell), guinea pig anti‐insulin antibody (DAKO, 1:500) followed by anti‐guinea pig gold 10 nm (British Biocell International), and mouse anti‐glucagon (1:500, Sigma) followed by anti‐mouse pig gold 10 nm (British Biocell International). Sections were viewed with a Joel 1010 microscope (accelerating voltage 80 kV). To establish the specificity of Tspan7 antibody, immunostaining was conducted using a stable Tspan7 KD INS‐1 cell line (cell line generation is described in Data S1). Tspan7 KD INS‐1 or control cells were fixed in 4% PFA, permeabilised using 0.1% Triton X‐100 and blocked with 5% normal swine serum. Tspan7 was detected using rabbit anti‐Tspan7 primary antibody (anti‐TM4SF2, 1:50, Sigma‐Aldrich; overnight incubation, 4°C), and with goat anti‐rabbit IgG Alexa Fluor 568 secondary antibody (Life Technologies). Nucleus were stained with RedDot2 (BioTium; 1:200 dilution). Slides were visualised by confocal microscopy (BioRad). Sections of formalin‐fixed paraffin‐embedded tissue were de‐waxed and subjected to epitope retrieval in a microwave pressure cooker in 10 mM citric acid pH 6.0, 0.05% Tween 20. After blocking with 2.5% normal horse serum, sections were incubated overnight (4°C) with rabbit anti‐Tspan7 antibody (Anti‐TM4SF2, 1:50, Sigma‐Aldrich). Antibody labelling was detected with the VectaFluor™ Excel Amplified Anti‐Rabbit IgG, DyLight™ 488 Antibody Kit (Vector Laboratories). Slides were counterstained with guinea pig anti‐insulin (1:500, Dako) and mouse anti‐glucagon (1:500, Sigma) antibodies followed by labelling with goat anti‐guinea pig‐Alexa 633 (1:100) and goat anti‐mouse Alexa 555 (1:100) secondary antibodies (ThermoFisher). Slides were visualised by confocal microscopy. For electron microscopy, isolated human islets were fixed in 2.5% glutaraldehyde in PBS, postfixed in 1% OsO4, dehydrated and embedded in Spurr's resin. Sections were cut onto nickel grids before being immunolabelled using anti‐TM4SF2 (1:20) and protein A gold particles (15 nm, Biocell), guinea pig anti‐insulin antibody (DAKO, 1:500) followed by anti‐guinea pig gold 10 nm (British Biocell International), and mouse anti‐glucagon (1:500, Sigma) followed by anti‐mouse pig gold 10 nm (British Biocell International). Sections were viewed with a Joel 1010 microscope (accelerating voltage 80 kV). To establish the specificity of Tspan7 antibody, immunostaining was conducted using a stable Tspan7 KD INS‐1 cell line (cell line generation is described in Data S1). Tspan7 KD INS‐1 or control cells were fixed in 4% PFA, permeabilised using 0.1% Triton X‐100 and blocked with 5% normal swine serum. Tspan7 was detected using rabbit anti‐Tspan7 primary antibody (anti‐TM4SF2, 1:50, Sigma‐Aldrich; overnight incubation, 4°C), and with goat anti‐rabbit IgG Alexa Fluor 568 secondary antibody (Life Technologies). Nucleus were stained with RedDot2 (BioTium; 1:200 dilution). Slides were visualised by confocal microscopy (BioRad). [SUBTITLE] Glucose tolerance test [SUBSECTION] Intraperitoneal (i.p.) glucose tolerance test was performed as previously described. 15 Briefly, the animals were individually fasted for 6 h before i.p. injected with a bolus of glucose (2 g/kg body weight). Blood glucose were monitored for 90 min using a glucometer by tail vein sampling. Intraperitoneal (i.p.) glucose tolerance test was performed as previously described. 15 Briefly, the animals were individually fasted for 6 h before i.p. injected with a bolus of glucose (2 g/kg body weight). Blood glucose were monitored for 90 min using a glucometer by tail vein sampling. [SUBTITLE] quantitative PCR [SUBSECTION] RNA was extracted from human pseudo‐islets using TRIzol Reagent (Life Technologies, 15,596,026) according to the manufacturer's instructions. cDNA was generated using the GoScript Reverse Transcription Kit (Promega, A5000). qPCR was performed using 20 ng of cDNA, and TaqMan Gene Expression Master Mix (Life Technologies Ltd, 4,369,016) or SYBR™ Green PCR Master Mix (Applied Biosystems, 4,309,155) with the gene expression assays or primers as detailed in Table 1. Gene expression was determined using the ΔΔCT method by normalising to HouseKeeping Genes (HKG) and to the level of expression of each target detected in Ad‐Scramble transduced cells. Data are presented as percentage of control. Primers and types of assays used for quantitative PCR analyses RNA was extracted from human pseudo‐islets using TRIzol Reagent (Life Technologies, 15,596,026) according to the manufacturer's instructions. cDNA was generated using the GoScript Reverse Transcription Kit (Promega, A5000). qPCR was performed using 20 ng of cDNA, and TaqMan Gene Expression Master Mix (Life Technologies Ltd, 4,369,016) or SYBR™ Green PCR Master Mix (Applied Biosystems, 4,309,155) with the gene expression assays or primers as detailed in Table 1. Gene expression was determined using the ΔΔCT method by normalising to HouseKeeping Genes (HKG) and to the level of expression of each target detected in Ad‐Scramble transduced cells. Data are presented as percentage of control. Primers and types of assays used for quantitative PCR analyses [SUBTITLE] RNA sequencing [SUBSECTION] Human pseudo‐islets were preserved in Trizol before their total RNA was extracted using Quick‐RNA MicroPrep kit (R1050 Zymo Research). RNA libraries for RNA‐seq were prepared using NEBNext Ultra II Directional RNA Library Prep Kit for Illumina following manufacturer's protocols. Sequencing was performed on NextSeq 550. Single‐end reads were splice‐aligned to a human genome (GRCh38) using GSNAP. FeatureCounts was used to assign reads to exons thus eventually getting counts per gene. EdgeR package of R 21 was used to perform differential analysis between the conditions (shRNA vs control), while controlling for the confounding effect from the donor. Across‐samples normalisation was performed using the TMM normalisation method. Human pseudo‐islets were preserved in Trizol before their total RNA was extracted using Quick‐RNA MicroPrep kit (R1050 Zymo Research). RNA libraries for RNA‐seq were prepared using NEBNext Ultra II Directional RNA Library Prep Kit for Illumina following manufacturer's protocols. Sequencing was performed on NextSeq 550. Single‐end reads were splice‐aligned to a human genome (GRCh38) using GSNAP. FeatureCounts was used to assign reads to exons thus eventually getting counts per gene. EdgeR package of R 21 was used to perform differential analysis between the conditions (shRNA vs control), while controlling for the confounding effect from the donor. Across‐samples normalisation was performed using the TMM normalisation method. [SUBTITLE] Data analyses and statistics [SUBSECTION] Data are expressed as mean value ± SEM. p‐values less than 0.05 were considered significant. Statistical analyses were performed using OriginPro 2017 either by Student's t‐test, paired comparison or two‐way ANOVA and Tukey post hoc test. Ns represent the number of cells for electrophysiological analyses, and of independent mice or cell passages for hormone assays and expression data. Data are expressed as mean value ± SEM. p‐values less than 0.05 were considered significant. Statistical analyses were performed using OriginPro 2017 either by Student's t‐test, paired comparison or two‐way ANOVA and Tukey post hoc test. Ns represent the number of cells for electrophysiological analyses, and of independent mice or cell passages for hormone assays and expression data.
RESULTS
[SUBTITLE] Tspan7 is expressed in pancreatic islets [SUBSECTION] Tspan7 expression in pancreatic islets was evaluated using immunohistochemistry. In both human and mouse pancreatic islets, Tspan7 was found to colocalise with insulin and glucagon, confirming its expression in β‐ and α‐cells 13 (Figure 1a). We observed enriched immunogold labelling of Tspan7 in large dense‐core vesicle of human α‐ and β‐cells (Figure 1b,c), consistent with a previous report that Tspan7 was present in the cytoplasm of islet cells with a distribution pattern similar to islet hormones. 22 Tetraspanin‐7 is expressed in pancreatic islets of Langerhans. (a) Immunofluorescent staining of human (upper) and mouse (lower) pancreas sections. Tetraspanin‐7 (TSPAN7 or Tspan7, red) is detected in β (Insulin, green)‐ and α (Glucagon, grey)‐cells. Scale bar: 30 μm. (b, c) Immunogold labelling of TSPAN7 (15 nm gold particles, black arrows), in glucagon (b) and insulin (c) containing vesicles in human islets. Insulin was labelled with 10 nm gold particles (white arrows). Scale bar: 500 nm. Tspan7 expression in pancreatic islets was evaluated using immunohistochemistry. In both human and mouse pancreatic islets, Tspan7 was found to colocalise with insulin and glucagon, confirming its expression in β‐ and α‐cells 13 (Figure 1a). We observed enriched immunogold labelling of Tspan7 in large dense‐core vesicle of human α‐ and β‐cells (Figure 1b,c), consistent with a previous report that Tspan7 was present in the cytoplasm of islet cells with a distribution pattern similar to islet hormones. 22 Tetraspanin‐7 is expressed in pancreatic islets of Langerhans. (a) Immunofluorescent staining of human (upper) and mouse (lower) pancreas sections. Tetraspanin‐7 (TSPAN7 or Tspan7, red) is detected in β (Insulin, green)‐ and α (Glucagon, grey)‐cells. Scale bar: 30 μm. (b, c) Immunogold labelling of TSPAN7 (15 nm gold particles, black arrows), in glucagon (b) and insulin (c) containing vesicles in human islets. Insulin was labelled with 10 nm gold particles (white arrows). Scale bar: 500 nm. [SUBTITLE] Ablation of Tspan7 has limited impact on glucose tolerance [SUBSECTION] To investigate the role of Tspan7 in islet function and systemic metabolism, a Tspan7 knockout mouse model (Tspan7y/−) 9 was used. Reduced body weight and ad libitum plasma glucose was observed in Tspan7y/− mice (Figure 2a,b), but this was not linked to changes in plasma insulin (measured as insulin and C‐peptide, Figure 2c,d). These effects may instead be attributable to the neurological manifestations of Tspan7 ablation, including depressive behaviour. 23 We observed no change in circulating proinsulin levels (Figure 2e), suggesting normal insulin processing in Tspan7y/− β‐cells. Furthermore, when subjected to intra‐peritoneal glucose tolerance tests (IPGTTs), no difference in glucose tolerance (Figure 2f) or glucose‐induced insulin secretion (GSIS, Figure S1) was observed between Tspan7y/− and control mice. In vivo metabolic phenotyping of Tspan7y/− mouse model. (a–e) Ad libitum bodyweight (a), and plasma concentrations of glucose (b), insulin (c), C‐peptide (d) and proinsulin measured in Tspan7y/− mice (red) and age‐matched (11.3 ± 0.3 wks) litter mate control (black). *p < 0.05 versus control and **p < 0.01 versus control. (f) Plasma glucose concentrations of Tspan7y/− mice (red) and age‐matched litter mate control (black) measured during intraperitoneal glucose tolerance tests. Glucose bolus was injected at 0 min. N = 5 for control and 6 for Tspan7y/−. Data are presented as mean value ± SEM. To investigate the role of Tspan7 in islet function and systemic metabolism, a Tspan7 knockout mouse model (Tspan7y/−) 9 was used. Reduced body weight and ad libitum plasma glucose was observed in Tspan7y/− mice (Figure 2a,b), but this was not linked to changes in plasma insulin (measured as insulin and C‐peptide, Figure 2c,d). These effects may instead be attributable to the neurological manifestations of Tspan7 ablation, including depressive behaviour. 23 We observed no change in circulating proinsulin levels (Figure 2e), suggesting normal insulin processing in Tspan7y/− β‐cells. Furthermore, when subjected to intra‐peritoneal glucose tolerance tests (IPGTTs), no difference in glucose tolerance (Figure 2f) or glucose‐induced insulin secretion (GSIS, Figure S1) was observed between Tspan7y/− and control mice. In vivo metabolic phenotyping of Tspan7y/− mouse model. (a–e) Ad libitum bodyweight (a), and plasma concentrations of glucose (b), insulin (c), C‐peptide (d) and proinsulin measured in Tspan7y/− mice (red) and age‐matched (11.3 ± 0.3 wks) litter mate control (black). *p < 0.05 versus control and **p < 0.01 versus control. (f) Plasma glucose concentrations of Tspan7y/− mice (red) and age‐matched litter mate control (black) measured during intraperitoneal glucose tolerance tests. Glucose bolus was injected at 0 min. N = 5 for control and 6 for Tspan7y/−. Data are presented as mean value ± SEM. [SUBTITLE] Exocytosis sensitivity to Ca2+ was reduced in Tspan7y/− β‐cells [SUBSECTION] In β‐cells, glucose metabolism increases the intracellular ATP/ADP ratio, resulting in closure of ATP‐sensitive K+ channels (KATP‐channels) inducing membrane depolarisation and action potential (AP) firing. APs open voltage‐gated Ca2+ channels (VGCCs), enabling influx of Ca2+ to trigger Ca2+‐dependent exocytosis, culminating in insulin secretion. To interrogate the impact of Tspan7 ablation on β‐cell function at a single‐cell level, we phenotyped Tspan7y/− β‐cell excitability, transmembrane Ca2+ currents and exocytosis using electrophysiological techniques. In the presence of 1 mM glucose, Tspan7y/− and control β‐cells were both repolarised (Tspan7y/− vs. control: −80 ± 1 mV vs. −80 ± 2 mV; p = 0.4) and electrically silent. Elevating extracellular glucose or blocking the KATP channels (using tolbutamide, 200 μM) induced membrane depolarisation and firing of APs in β‐cells of both genotypes (Figure 3a). In the presence of 10 mM glucose, although the Tspan7y/− β‐cells were less depolarised (Tspan7y/− vs. control: −57 ± 4 mV vs. −46 ± 5 mV; p < 0.05), no difference in AP peak or firing frequency was observed across the two genotypes (Figure 3b–d). There was no apparent difference in AP halfwidth or spike duration, either (Figure S2). Further increasing extracellular glucose to 20 mM or inhibiting KATP channels with tolbutamide exerted similar effects on the electrical activity of both Tspan7y/− and control β‐cells (Figure 3b–d). However, Tspan7y/− β cells responded to 20 mM glucose stimulation faster than the control (Tspan7y/− vs. control: 2.4 ± 0.3 min vs. 3.85 ± 0.3 min, p < 0.05). These marginal changes in Tspan7y/− β‐cell electrical activity are not due to any apparent changes in KATP‐channel conductivity or glucose sensitivity (Figure 3e,f). Electrical activity and KATP‐channel conductance measured in Tspan7y/− β‐cells. (a) Membrane potential recordings of control (upper trace, black) and Tspan7y/− (lower trace, red) β‐cells within intact islets in response to changes in extracellular glucose (1 mM, 10 mM and 20 mM) or application of 200 μM tolbutamide, as indicated). (b) Summary of control (black) and Tspan7y/− (red) β‐cell membrane potential measured at conditions indicated. N = 7 for control and 14 for Tspan7y/− β‐cells at 1 mM glucose; N = 7 for control and 11 for Tspan7y/− β‐cells at 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 6 for Tspan7y/− β‐cells when 200 μM tolbutamide was applied. *p < 0.05 versus control. (c, d), as in (b) but summarise the peak voltage (c) and firing frequency (d) of action potentials measured in control (black) and Tspan7y/− (red) β‐cells at conditions indicated. N = 4 for control and 7 for Tspan7y/− β‐cells at 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 3 for Tspan7y/− β‐cells when tolbutamide was applied. (e) Summary of KATP‐channel conductance measured in control (black) and Tspan7y/− (red) β‐cells under the conditions indicated. N = 4 for control and 7 for Tspan7y/− β‐cells at 1 mM and 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 5 for Tspan7y/− β‐cells when tolbutamide was applied. (f) Examples of KATP currents measured in control (black, middle trace) and Tspan7y/− (red, bottom trace) β‐cells. KATP currents were triggered by ±10 mV excursions from −70 mV (200 ms, top) in voltage‐clamped β‐cells under the conditions indicated. All data are presented as mean value ± SEM. Transient knockdown of Tspan7 in β‐cells was recently shown to increase Ca2+ influx through L‐type VGCC. 13 We assessed whether a similar phenotype can be observed in β‐cells of mice with a constitutive Tspan7 knockout. As shown in Figure 4a,b, depolarisation triggered a significantly larger Ca2+ current in Tspan7y/− β‐cells than in the control (p < 0.01, ~160% at 0 and 10 mV), consistent with Dickerson et al. 13 As Ca2+ triggers exocytosis, we tested whether enhanced VGCC activity led to higher exocytosis in Tspan7y/− β‐cells using capacitance measurements. Exocytosis (measured as increases in cell capacitance) was trigged by a series of depolarising pulses from −70 mV to 0 mV with progressively longer durations (from 10 ms to 800 ms). Unexpectedly, exocytosis was comparable between Tspan7y/− and control β‐cells at all pulse durations (Figure 4c,d). We correlated the Ca2+ charges (total influx Ca2+ ions, QCa2+) with the corresponding exocytosis elicited by depolarisation (Figure 4e). Whereas exocytosis could be triggered by 0.15pC/pF QCa2+ in control β‐cells, it required > ~ 0.2pC/pF in Tspan7y/− β‐cells, suggesting a reduced exocytotic Ca2+sensitivity. Ca2+ current and exocytosis in Tspan7y/− β‐cells. Tspan7 loss of function significantly increases Ca2+ current density in β cells (a, b). (a) Representative traces of control (black) and Tspan7y/− (red) β‐cell Ca2+ current elicited by a 100‐ms depolarisation from −70 mV to 0 mV. (b) Summary of control (black) and Tspan7y/− (red) β‐cell Ca2+ current density in relationship to membrane voltages. N = 7 for control and 5 for Tspan7y/− β‐cells, **p < 0.01 versus control, paired comparison and Tukey. (c) Examples of exocytosis induced by membrane depolarisations from −70 mV to 0 mV (at the duration of 100 ms, 200 ms, 300 ms, 500 ms and 800 ms as indicated above the traces) of control (black) and Tspan7y/− (red) β‐cells. (d) Summary of control (black) and Tspan7y/− (red) β‐cell exocytosis (normalised to cell size, fF/pF) in relationship to pulse durations. N = 11 cells for control and 14 cells for Tspan7y/− β‐cell. (e) Control (black) and Tspan7y/− (red) β‐cell charges‐exocytosis relationship for pulses up to 100 ms. Corresponding durations of the pulses are as labelled. N = 7 cells for control and 5 cells for Tspan7y/− β‐cells. (f) Insulin secretion measured from batch‐incubated control (black) and Tspan7y/− (red) islets in response to glucose or tolbutamide (concentrations as indicated). Islets were isolated from mice aged 20 weeks and N ≥ 3 for control and Tspan7y/− islets. **p < 0.01 and ***p < 0.001 versus 1 mM glucose alone of the same genotype. All data are presented as mean value ± SEM. Given the limited changes in electrical activity and exocytosis in Tspan7y/− β‐cells, we reasoned that knocking‐out Tspan7 would have little effect on glucose/tolbutamide‐stimulated insulin secretion. Indeed, no significant difference was observed in insulin secretion, stimulated by 10 mM glucose or 0.2 mM tolbutamide, from islets of control and Tspan7y/− mice (Figure 4f). We reasoned that enhanced VGCC activity augments the concentration of cytosolic Ca2+, and thus may induce apoptosis, 24 affecting islet β cell mass in aged animals. However, no significant difference in GSIS, insulin secretion induced by 70 mM K+ or islet insulin content was observed in control and Tspan7y/− islets isolated from 1‐year‐old mice (Figure S4). In β‐cells, glucose metabolism increases the intracellular ATP/ADP ratio, resulting in closure of ATP‐sensitive K+ channels (KATP‐channels) inducing membrane depolarisation and action potential (AP) firing. APs open voltage‐gated Ca2+ channels (VGCCs), enabling influx of Ca2+ to trigger Ca2+‐dependent exocytosis, culminating in insulin secretion. To interrogate the impact of Tspan7 ablation on β‐cell function at a single‐cell level, we phenotyped Tspan7y/− β‐cell excitability, transmembrane Ca2+ currents and exocytosis using electrophysiological techniques. In the presence of 1 mM glucose, Tspan7y/− and control β‐cells were both repolarised (Tspan7y/− vs. control: −80 ± 1 mV vs. −80 ± 2 mV; p = 0.4) and electrically silent. Elevating extracellular glucose or blocking the KATP channels (using tolbutamide, 200 μM) induced membrane depolarisation and firing of APs in β‐cells of both genotypes (Figure 3a). In the presence of 10 mM glucose, although the Tspan7y/− β‐cells were less depolarised (Tspan7y/− vs. control: −57 ± 4 mV vs. −46 ± 5 mV; p < 0.05), no difference in AP peak or firing frequency was observed across the two genotypes (Figure 3b–d). There was no apparent difference in AP halfwidth or spike duration, either (Figure S2). Further increasing extracellular glucose to 20 mM or inhibiting KATP channels with tolbutamide exerted similar effects on the electrical activity of both Tspan7y/− and control β‐cells (Figure 3b–d). However, Tspan7y/− β cells responded to 20 mM glucose stimulation faster than the control (Tspan7y/− vs. control: 2.4 ± 0.3 min vs. 3.85 ± 0.3 min, p < 0.05). These marginal changes in Tspan7y/− β‐cell electrical activity are not due to any apparent changes in KATP‐channel conductivity or glucose sensitivity (Figure 3e,f). Electrical activity and KATP‐channel conductance measured in Tspan7y/− β‐cells. (a) Membrane potential recordings of control (upper trace, black) and Tspan7y/− (lower trace, red) β‐cells within intact islets in response to changes in extracellular glucose (1 mM, 10 mM and 20 mM) or application of 200 μM tolbutamide, as indicated). (b) Summary of control (black) and Tspan7y/− (red) β‐cell membrane potential measured at conditions indicated. N = 7 for control and 14 for Tspan7y/− β‐cells at 1 mM glucose; N = 7 for control and 11 for Tspan7y/− β‐cells at 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 6 for Tspan7y/− β‐cells when 200 μM tolbutamide was applied. *p < 0.05 versus control. (c, d), as in (b) but summarise the peak voltage (c) and firing frequency (d) of action potentials measured in control (black) and Tspan7y/− (red) β‐cells at conditions indicated. N = 4 for control and 7 for Tspan7y/− β‐cells at 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 3 for Tspan7y/− β‐cells when tolbutamide was applied. (e) Summary of KATP‐channel conductance measured in control (black) and Tspan7y/− (red) β‐cells under the conditions indicated. N = 4 for control and 7 for Tspan7y/− β‐cells at 1 mM and 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 5 for Tspan7y/− β‐cells when tolbutamide was applied. (f) Examples of KATP currents measured in control (black, middle trace) and Tspan7y/− (red, bottom trace) β‐cells. KATP currents were triggered by ±10 mV excursions from −70 mV (200 ms, top) in voltage‐clamped β‐cells under the conditions indicated. All data are presented as mean value ± SEM. Transient knockdown of Tspan7 in β‐cells was recently shown to increase Ca2+ influx through L‐type VGCC. 13 We assessed whether a similar phenotype can be observed in β‐cells of mice with a constitutive Tspan7 knockout. As shown in Figure 4a,b, depolarisation triggered a significantly larger Ca2+ current in Tspan7y/− β‐cells than in the control (p < 0.01, ~160% at 0 and 10 mV), consistent with Dickerson et al. 13 As Ca2+ triggers exocytosis, we tested whether enhanced VGCC activity led to higher exocytosis in Tspan7y/− β‐cells using capacitance measurements. Exocytosis (measured as increases in cell capacitance) was trigged by a series of depolarising pulses from −70 mV to 0 mV with progressively longer durations (from 10 ms to 800 ms). Unexpectedly, exocytosis was comparable between Tspan7y/− and control β‐cells at all pulse durations (Figure 4c,d). We correlated the Ca2+ charges (total influx Ca2+ ions, QCa2+) with the corresponding exocytosis elicited by depolarisation (Figure 4e). Whereas exocytosis could be triggered by 0.15pC/pF QCa2+ in control β‐cells, it required > ~ 0.2pC/pF in Tspan7y/− β‐cells, suggesting a reduced exocytotic Ca2+sensitivity. Ca2+ current and exocytosis in Tspan7y/− β‐cells. Tspan7 loss of function significantly increases Ca2+ current density in β cells (a, b). (a) Representative traces of control (black) and Tspan7y/− (red) β‐cell Ca2+ current elicited by a 100‐ms depolarisation from −70 mV to 0 mV. (b) Summary of control (black) and Tspan7y/− (red) β‐cell Ca2+ current density in relationship to membrane voltages. N = 7 for control and 5 for Tspan7y/− β‐cells, **p < 0.01 versus control, paired comparison and Tukey. (c) Examples of exocytosis induced by membrane depolarisations from −70 mV to 0 mV (at the duration of 100 ms, 200 ms, 300 ms, 500 ms and 800 ms as indicated above the traces) of control (black) and Tspan7y/− (red) β‐cells. (d) Summary of control (black) and Tspan7y/− (red) β‐cell exocytosis (normalised to cell size, fF/pF) in relationship to pulse durations. N = 11 cells for control and 14 cells for Tspan7y/− β‐cell. (e) Control (black) and Tspan7y/− (red) β‐cell charges‐exocytosis relationship for pulses up to 100 ms. Corresponding durations of the pulses are as labelled. N = 7 cells for control and 5 cells for Tspan7y/− β‐cells. (f) Insulin secretion measured from batch‐incubated control (black) and Tspan7y/− (red) islets in response to glucose or tolbutamide (concentrations as indicated). Islets were isolated from mice aged 20 weeks and N ≥ 3 for control and Tspan7y/− islets. **p < 0.01 and ***p < 0.001 versus 1 mM glucose alone of the same genotype. All data are presented as mean value ± SEM. Given the limited changes in electrical activity and exocytosis in Tspan7y/− β‐cells, we reasoned that knocking‐out Tspan7 would have little effect on glucose/tolbutamide‐stimulated insulin secretion. Indeed, no significant difference was observed in insulin secretion, stimulated by 10 mM glucose or 0.2 mM tolbutamide, from islets of control and Tspan7y/− mice (Figure 4f). We reasoned that enhanced VGCC activity augments the concentration of cytosolic Ca2+, and thus may induce apoptosis, 24 affecting islet β cell mass in aged animals. However, no significant difference in GSIS, insulin secretion induced by 70 mM K+ or islet insulin content was observed in control and Tspan7y/− islets isolated from 1‐year‐old mice (Figure S4). [SUBTITLE] Reducing TSPAN7 expression in human islets changes gene expression of islet exocytosis Ca2+ sensors [SUBSECTION] To assess the role of tetraspanin‐7 in human β‐cell function, we utilised an adenovirus encoding shRNATSPAN7 (Ad‐ShTSPAN7) to knockdown (KD) human TSPAN7 expression. Islet cells transduced with Ad‐ShTSPAN7, or the control virus (with scrambled RNA), were used to form pseudo‐islets. RNA sequencing showed a marked reduction in TSPAN7 expression (92.65%) in the TSPAN7‐KD pseudo‐islets (log2[Fold‐change] = −3.765, p adj = 6.42 E‐13) (Figure 5a), confirming successful genetic manipulation. In total, 1030 genes (1000 are protein‐coding genes) were down‐regulated and 395 genes (372 are protein‐coding genes) were up‐regulated in TSPAN7‐KD pseudo‐islets. Pathway analyses showed up‐regulation in apoptosis gene ontology pathway (p adj = 4.08 E−7); and down‐regulation of genes involved in the secretory pathway and transport regulation (p adj = 5.62 E−9 and 4.22 E−08 respectively). Interestingly, β‐cell expression of the exocytosis Ca2+ sensor SYT7 was reduced in TSPAN7‐KD pseudo‐islets (log2[Fold‐change] = −0.39, p adj = 3.89 E−3). GSIS of the pseudo‐islets were tested using dynamic perifusion experiments (Figure 5b). Whereas insulin secretion at basal or 16.7 mM glucose was comparable between the TSPAN7‐KD and control pseudo‐islets, 20 mM K+‐stimulated insulin secretion was significantly lower in TSPAN7‐KD pseudo‐islets. TSPAN7 KD affects exocytosis from human pancreatic β‐cells. (a) Gene expressions that changed significantly in human TSPAN7 KD pseudo‐islet. Volcano plot shows that genes involved in the pathways of secretion (yellow area) and regulation of transport (green area) were down‐regulated; markers of the apoptotic pathway (blue) were up‐regulated. TSPAN7 KD was demonstrated by the marked reduction in TSPAN7 expression (red). (b) Dynamic insulin secretion measured from perifused human pseudo‐islets of TSPAN7 KD (red) or control (Scramble, black) in response to different glucose concentrations and 20 mM K+ as indicated. Inset: area of the curve (AUC) of insulin secretion stimulated by 20 mM K+. N = 3 for control and TSPAN7 KD, *p < 0.05 versus insulin secretion at 1 mM glucose in the same group of pseudo‐islets; † p < 0.05 versus control (Scramble), Student's t‐test. (c) Examples of exocytosis of TSPAN7 KD EndoC‐βH1 (red) or control EndoC‐βH1 (Scramble, black) triggered by depolarisation from −70 to 0 mV with progressively increasing duration (as indicated). (d) Summary of control (black) and exocytosis (normalised to cell size, fF/pF) in relationship to pulse durations. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. (e) The relationship between depolarisation‐triggered charge influx and pulse durations of control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. *p < 0.05 versus control, student's t‐test. (f) The relationship between charges and exocytosis of control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. Corresponding durations of the pulses are as labelled. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. (g) Insulin secretion measured from batch‐incubated control (black) and TSPAN7 KD (red) EndoC‐βH1 cells in response to 1 and 20 mM glucose. Data are the summary of three independent biological repeats in technical triplicate. *p < 0.05 and ***p < 0.001 versus control insulin secretion at 1 mM glucose and †† p < 0.01 versus control insulin secretion at 20 mM glucose; two‐way ANOVA and Tuckey post hoc test. (h) Insulin content measured from control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. Data are the summary of three independent biological repeats in technical triplicate. All data are presented as mean value ± SEM. The insulin secretion experiments suggested that TSPAN7 may play a role in human β‐cell exocytosis. To test this, we performed capacitance measurements using EndoC‐βH1, a human β‐cell line recapturing functional and genetic features of primary human β‐cells, 18 transduced with Ad‐ShTSPAN7 or Ad‐Scramble virus. Similar to that observed in mouse Tspan7y/− β‐cells, exocytosis triggered by depolarisation was comparable between TSPAN7‐KD EndoC‐βH1 and control cells (Figure 5c,d). However, depolarisation triggered larger influx of charges (p < 0.05) in TSPAN7‐KD EndoC‐βH1 cells than that of the control (Ad‐Scramble) (Figure 5e). Importantly, the exocytotic Ca2+‐sensitivity of TSPAN7‐KD EndoC‐βH1 was reduced (same QCa2+ could only evoke exocytosis that was ~50% of the control, Figure 5f). This correlated with a significantly reduced GSIS in TSPAN7‐KD EndoC‐βH1 (p < 0.01, Figure 5g) without affecting insulin content (Figure 5h). We measured the changes in gene expression of VGCC and synaptotagmins in TSPAN7‐KD EndoC‐βH1. Concomitantly to TSPAN7 KD (p = 5.51 E−4), CACNA1A was up‐regulated and SYT5 and SYT7 were down‐regulated (−20%, Figure 6). Changes in VGCC‐ and synaptotagmin‐encoding genes in EndoC‐βH1 following TSPAN7 KD. The expression of genes encoding TSPAN7 (TSPAN7), VGCCs (CACNA1A, CACNA1C and CACNA1D) and synaptotagmins (SYT4, SYT5 and SYT7) measured from control (transduced with Ad‐Scramble, Scramble, black) and TSPAN7 KD (red) EndoC‐βH1 cells. The levels of expression are normalised to that in control cells (as 100%). Error bars of the control cells condition reflect the variation of the control across the biological repeats. Data are summary of measurements made in two to four independent biological replicates. ***p < 0.0001 versus control, Student's t‐test. To assess the role of tetraspanin‐7 in human β‐cell function, we utilised an adenovirus encoding shRNATSPAN7 (Ad‐ShTSPAN7) to knockdown (KD) human TSPAN7 expression. Islet cells transduced with Ad‐ShTSPAN7, or the control virus (with scrambled RNA), were used to form pseudo‐islets. RNA sequencing showed a marked reduction in TSPAN7 expression (92.65%) in the TSPAN7‐KD pseudo‐islets (log2[Fold‐change] = −3.765, p adj = 6.42 E‐13) (Figure 5a), confirming successful genetic manipulation. In total, 1030 genes (1000 are protein‐coding genes) were down‐regulated and 395 genes (372 are protein‐coding genes) were up‐regulated in TSPAN7‐KD pseudo‐islets. Pathway analyses showed up‐regulation in apoptosis gene ontology pathway (p adj = 4.08 E−7); and down‐regulation of genes involved in the secretory pathway and transport regulation (p adj = 5.62 E−9 and 4.22 E−08 respectively). Interestingly, β‐cell expression of the exocytosis Ca2+ sensor SYT7 was reduced in TSPAN7‐KD pseudo‐islets (log2[Fold‐change] = −0.39, p adj = 3.89 E−3). GSIS of the pseudo‐islets were tested using dynamic perifusion experiments (Figure 5b). Whereas insulin secretion at basal or 16.7 mM glucose was comparable between the TSPAN7‐KD and control pseudo‐islets, 20 mM K+‐stimulated insulin secretion was significantly lower in TSPAN7‐KD pseudo‐islets. TSPAN7 KD affects exocytosis from human pancreatic β‐cells. (a) Gene expressions that changed significantly in human TSPAN7 KD pseudo‐islet. Volcano plot shows that genes involved in the pathways of secretion (yellow area) and regulation of transport (green area) were down‐regulated; markers of the apoptotic pathway (blue) were up‐regulated. TSPAN7 KD was demonstrated by the marked reduction in TSPAN7 expression (red). (b) Dynamic insulin secretion measured from perifused human pseudo‐islets of TSPAN7 KD (red) or control (Scramble, black) in response to different glucose concentrations and 20 mM K+ as indicated. Inset: area of the curve (AUC) of insulin secretion stimulated by 20 mM K+. N = 3 for control and TSPAN7 KD, *p < 0.05 versus insulin secretion at 1 mM glucose in the same group of pseudo‐islets; † p < 0.05 versus control (Scramble), Student's t‐test. (c) Examples of exocytosis of TSPAN7 KD EndoC‐βH1 (red) or control EndoC‐βH1 (Scramble, black) triggered by depolarisation from −70 to 0 mV with progressively increasing duration (as indicated). (d) Summary of control (black) and exocytosis (normalised to cell size, fF/pF) in relationship to pulse durations. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. (e) The relationship between depolarisation‐triggered charge influx and pulse durations of control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. *p < 0.05 versus control, student's t‐test. (f) The relationship between charges and exocytosis of control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. Corresponding durations of the pulses are as labelled. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. (g) Insulin secretion measured from batch‐incubated control (black) and TSPAN7 KD (red) EndoC‐βH1 cells in response to 1 and 20 mM glucose. Data are the summary of three independent biological repeats in technical triplicate. *p < 0.05 and ***p < 0.001 versus control insulin secretion at 1 mM glucose and †† p < 0.01 versus control insulin secretion at 20 mM glucose; two‐way ANOVA and Tuckey post hoc test. (h) Insulin content measured from control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. Data are the summary of three independent biological repeats in technical triplicate. All data are presented as mean value ± SEM. The insulin secretion experiments suggested that TSPAN7 may play a role in human β‐cell exocytosis. To test this, we performed capacitance measurements using EndoC‐βH1, a human β‐cell line recapturing functional and genetic features of primary human β‐cells, 18 transduced with Ad‐ShTSPAN7 or Ad‐Scramble virus. Similar to that observed in mouse Tspan7y/− β‐cells, exocytosis triggered by depolarisation was comparable between TSPAN7‐KD EndoC‐βH1 and control cells (Figure 5c,d). However, depolarisation triggered larger influx of charges (p < 0.05) in TSPAN7‐KD EndoC‐βH1 cells than that of the control (Ad‐Scramble) (Figure 5e). Importantly, the exocytotic Ca2+‐sensitivity of TSPAN7‐KD EndoC‐βH1 was reduced (same QCa2+ could only evoke exocytosis that was ~50% of the control, Figure 5f). This correlated with a significantly reduced GSIS in TSPAN7‐KD EndoC‐βH1 (p < 0.01, Figure 5g) without affecting insulin content (Figure 5h). We measured the changes in gene expression of VGCC and synaptotagmins in TSPAN7‐KD EndoC‐βH1. Concomitantly to TSPAN7 KD (p = 5.51 E−4), CACNA1A was up‐regulated and SYT5 and SYT7 were down‐regulated (−20%, Figure 6). Changes in VGCC‐ and synaptotagmin‐encoding genes in EndoC‐βH1 following TSPAN7 KD. The expression of genes encoding TSPAN7 (TSPAN7), VGCCs (CACNA1A, CACNA1C and CACNA1D) and synaptotagmins (SYT4, SYT5 and SYT7) measured from control (transduced with Ad‐Scramble, Scramble, black) and TSPAN7 KD (red) EndoC‐βH1 cells. The levels of expression are normalised to that in control cells (as 100%). Error bars of the control cells condition reflect the variation of the control across the biological repeats. Data are summary of measurements made in two to four independent biological replicates. ***p < 0.0001 versus control, Student's t‐test.
null
null
[ "What is already known?", "What did this study find?", "What are the implications of the study?", "INTRODUCTION", "Animals and isolation of pancreatic islets", "\nshRNA\n", "Human pseudo‐islets and EndoC‐βH1 cell line", "Insulin secretion", "Electrophysiology", "Histology", "Glucose tolerance test", "quantitative PCR\n", "\nRNA sequencing", "Data analyses and statistics", "\nTspan7 is expressed in pancreatic islets", "Ablation of Tspan7 has limited impact on glucose tolerance", "Exocytosis sensitivity to Ca2+ was reduced in Tspan7y/− β‐cells", "Reducing \nTSPAN7\n expression in human islets changes gene expression of islet exocytosis Ca2+ sensors" ]
[ "Tetraspanin‐7, a transmembrane protein widely expressed in pancreatic islet cells, was previously identified as an autoantigen in type 1 diabetes. Tetraspanin‐7 has been shown to regulate β‐cell L‐type Ca2+ channel activity.", "Genetic ablation of tetraspanin‐7 in mice led to a significant reduction in the Ca2+ dependence of β‐cell exocytosis. This phenotype was also observed in human islet cells following ex vivo tetraspanin‐7 knockdown, alongside down‐regulation of the β‐cell exocytosis Ca2+ sensor, synaptotagmin‐7.", "Tetraspanin‐7 expression is required for normal exocytotic machinery and appropriate exocytosis Ca2+ sensitivity in pancreatic β‐cells.", "Tetraspanin‐7 (Tspan7) was recently established as an autoantigen in type 1 diabetes mellitus (T1D),\n1\n an autoimmune disease in which loss of pancreatic β‐cells leads to chronic hyperglycaemia.\n2\n Proteins of the tetraspanin superfamily, of which Tspan7 is a member, are characterised by the presence of four transmembrane domains: one short and one long extracellular loop, a short intracellular loop, and N‐terminal and C‐terminal cytoplasmic tails.\n3\n, \n4\n Tetraspanin proteins are typically organised in tetraspanin‐enriched microdomains on biological membranes\n5\n where they act as molecular facilitators, engaging both transmembrane and intracellular proteins to regulate diverse processes such as cell proliferation, differentiation, activation, adhesion, motility and signalling.\n5\n, \n6\n, \n7\n\n\nTspan7 is highly expressed in neuroendocrine tissue,\n8\n and loss of Tspan7 can lead to defects in neuronal morphogenesis and synaptic transmission,\n9\n similar to those described in people expressing a mutated form.\n10\n Additional functions in cell morphology have been reported in dendritic cells\n11\n and osteoclasts,\n12\n attributed to its role in actin cytoskeleton rearrangement. Within the pancreas, Tspan7 is localised to the pancreatic islets and was recently shown to function as an auxiliary protein of L‐type voltage‐gated Ca2+ channels (VGCCs), modulating β‐cell electrical excitability and exocytosis. Knocking‐down of Tspan7 in β‐cells led to higher Ca2+ influx (through L‐type VGCCs), cellular excitability and glucose‐stimulated insulin secretion.\n13\n\n\nIn this study, we characterised Tspan7y/− mouse metabolic phenotype, β‐cell excitability, glucose‐stimulated insulin secretion and cell exocytosis. These data provide further insights of the role of Tspan7 in regulating β‐cell intracellular Ca2+ dynamics and insulin secretion, and thus to determine the importance of Tspan7 in glucose tolerance.", "All animal experiments were conducted in accordance with the UK Animals Scientific Procedures Act (1986) and the University of Oxford ethical guidelines. Mouse islets were isolated as previously described.\n14\n, \n15\n\nTspan7 knockout mice (Tspan7y/−)\n9\n were gifts from Dr Luca Murru (CNR Institute of Neuroscience, Milano, Italy). Hormone secretion studies and the electrophysiology experiments were performed on islets isolated from Tspan7y/− mice and their wildtype littermates (control).", "Adenoviruses expressing shRNA for silencing of human TSPAN7 (Ad‐ShTSPAN7, SKU# shADV‐226,651) or scrambled shRNA control (Ad‐Scramble, SKU# 1122) together with eGFP under a CMV promoter were purchased from Vector Biolabs.", "Donor organs were obtained with written consent and research ethics approval at the University of Alberta, and human islets were isolated as previous described\n16\n at the University of Alberta. All human islet information is listed in Table S1. Islets were dispersed into a single‐cell suspension using trypsin (TryLE Express, Gibco) and transduced with Ad‐ShTSPAN7 or Ad‐Scramble before forming human pseudo‐islets using centrifugal‐forced aggregation as previously described.\n17\n EndoC‐βH1 was provided by Endocell and cultured as previously described.\n18\n\n", "Insulin secretion was measured in static incubations as previously described.\n15\n Briefly, groups of 10–20 isolated islets (number depended on islet availability) were pre‐incubated for 30 min at 37°C in Krebs‐Ringer buffer (KRB) consisting of (mM): 140 NaCl, 3.6 KCl, 0.5 MgSO4, 2.6 CaCl2, 0.5 NaH2PO4, 2 NaHCO3, 1 glucose and 5 HEPES (pH = 7.4 with NaOH), before being incubated for 60 min at 37°C in 1 ml of KRB supplemented as indicated in the legends. Immediately after incubation, an aliquot of the medium was removed for determination of insulin concentration using an ELISA kit (Mercodia, Sweden).\nFor dynamic insulin secretion measurements, groups of 50 pseudo‐islets were perifused with KRB containing 1 or 16.7 mM glucose with or without 20 mM K+ (as indicated) at the rate of 100 μl/min, using a perifusion system (Biorep, FL, USA). Perfusate was collected every 2 min and insulin concentration was determined using the Alpco stellux human ELISA kits (Salem, NH).\nThe perifusion experiments were conducted at the University of Alberta and the mRNAs of pseudo‐islets were then extracted using TRIzol (ThermoFisher) before shipped to Oxford for transcriptomic study.\nEndoC‐βH1 cells were sequentially exposed for 20 min to 1 and 20 mM glucose containing KRB. Supernatants were collected at the end of each incubation and insulin cellular contents were extracted using acid ethanol. Insulin concentrations were determined using an ELISA kit (Mercodia, Sweden).", "Electrophysiological measurements were performed using an EPC‐10 patch‐clamp amplifier and Pulse software (version 8.80, HEKA Electronics, Germany). Electrical activity and KATP‐channel conductance were measured using perforated patch‐clamping technique as previously described\n19\n; membrane currents and changes in cell capacitance were recorded using voltage clamp in combination with capacitance measurements, as described in.\n14\n β‐cells were identified by electrophysiological fingerprinting.\n20\n\n", "Sections of formalin‐fixed paraffin‐embedded tissue were de‐waxed and subjected to epitope retrieval in a microwave pressure cooker in 10 mM citric acid pH 6.0, 0.05% Tween 20. After blocking with 2.5% normal horse serum, sections were incubated overnight (4°C) with rabbit anti‐Tspan7 antibody (Anti‐TM4SF2, 1:50, Sigma‐Aldrich). Antibody labelling was detected with the VectaFluor™ Excel Amplified Anti‐Rabbit IgG, DyLight™ 488 Antibody Kit (Vector Laboratories). Slides were counterstained with guinea pig anti‐insulin (1:500, Dako) and mouse anti‐glucagon (1:500, Sigma) antibodies followed by labelling with goat anti‐guinea pig‐Alexa 633 (1:100) and goat anti‐mouse Alexa 555 (1:100) secondary antibodies (ThermoFisher). Slides were visualised by confocal microscopy.\nFor electron microscopy, isolated human islets were fixed in 2.5% glutaraldehyde in PBS, postfixed in 1% OsO4, dehydrated and embedded in Spurr's resin. Sections were cut onto nickel grids before being immunolabelled using anti‐TM4SF2 (1:20) and protein A gold particles (15 nm, Biocell), guinea pig anti‐insulin antibody (DAKO, 1:500) followed by anti‐guinea pig gold 10 nm (British Biocell International), and mouse anti‐glucagon (1:500, Sigma) followed by anti‐mouse pig gold 10 nm (British Biocell International). Sections were viewed with a Joel 1010 microscope (accelerating voltage 80 kV).\nTo establish the specificity of Tspan7 antibody, immunostaining was conducted using a stable Tspan7 KD INS‐1 cell line (cell line generation is described in Data S1). Tspan7 KD INS‐1 or control cells were fixed in 4% PFA, permeabilised using 0.1% Triton X‐100 and blocked with 5% normal swine serum. Tspan7 was detected using rabbit anti‐Tspan7 primary antibody (anti‐TM4SF2, 1:50, Sigma‐Aldrich; overnight incubation, 4°C), and with goat anti‐rabbit IgG Alexa Fluor 568 secondary antibody (Life Technologies). Nucleus were stained with RedDot2 (BioTium; 1:200 dilution). Slides were visualised by confocal microscopy (BioRad).", "Intraperitoneal (i.p.) glucose tolerance test was performed as previously described.\n15\n Briefly, the animals were individually fasted for 6 h before i.p. injected with a bolus of glucose (2 g/kg body weight). Blood glucose were monitored for 90 min using a glucometer by tail vein sampling.", "RNA was extracted from human pseudo‐islets using TRIzol Reagent (Life Technologies, 15,596,026) according to the manufacturer's instructions. cDNA was generated using the GoScript Reverse Transcription Kit (Promega, A5000). qPCR was performed using 20 ng of cDNA, and TaqMan Gene Expression Master Mix (Life Technologies Ltd, 4,369,016) or SYBR™ Green PCR Master Mix (Applied Biosystems, 4,309,155) with the gene expression assays or primers as detailed in Table 1. Gene expression was determined using the ΔΔCT method by normalising to HouseKeeping Genes (HKG) and to the level of expression of each target detected in Ad‐Scramble transduced cells. Data are presented as percentage of control.\nPrimers and types of assays used for quantitative PCR analyses", "Human pseudo‐islets were preserved in Trizol before their total RNA was extracted using Quick‐RNA MicroPrep kit (R1050 Zymo Research). RNA libraries for RNA‐seq were prepared using NEBNext Ultra II Directional RNA Library Prep Kit for Illumina following manufacturer's protocols. Sequencing was performed on NextSeq 550. Single‐end reads were splice‐aligned to a human genome (GRCh38) using GSNAP. FeatureCounts was used to assign reads to exons thus eventually getting counts per gene. EdgeR package of R\n21\n was used to perform differential analysis between the conditions (shRNA vs control), while controlling for the confounding effect from the donor. Across‐samples normalisation was performed using the TMM normalisation method.", "Data are expressed as mean value ± SEM. p‐values less than 0.05 were considered significant. Statistical analyses were performed using OriginPro 2017 either by Student's t‐test, paired comparison or two‐way ANOVA and Tukey post hoc test. Ns represent the number of cells for electrophysiological analyses, and of independent mice or cell passages for hormone assays and expression data.", "Tspan7 expression in pancreatic islets was evaluated using immunohistochemistry. In both human and mouse pancreatic islets, Tspan7 was found to colocalise with insulin and glucagon, confirming its expression in β‐ and α‐cells\n13\n (Figure 1a). We observed enriched immunogold labelling of Tspan7 in large dense‐core vesicle of human α‐ and β‐cells (Figure 1b,c), consistent with a previous report that Tspan7 was present in the cytoplasm of islet cells with a distribution pattern similar to islet hormones.\n22\n\n\nTetraspanin‐7 is expressed in pancreatic islets of Langerhans. (a) Immunofluorescent staining of human (upper) and mouse (lower) pancreas sections. Tetraspanin‐7 (TSPAN7 or Tspan7, red) is detected in β (Insulin, green)‐ and α (Glucagon, grey)‐cells. Scale bar: 30 μm. (b, c) Immunogold labelling of TSPAN7 (15 nm gold particles, black arrows), in glucagon (b) and insulin (c) containing vesicles in human islets. Insulin was labelled with 10 nm gold particles (white arrows). Scale bar: 500 nm.", "To investigate the role of Tspan7 in islet function and systemic metabolism, a Tspan7 knockout mouse model (Tspan7y/−)\n9\n was used. Reduced body weight and ad libitum plasma glucose was observed in Tspan7y/− mice (Figure 2a,b), but this was not linked to changes in plasma insulin (measured as insulin and C‐peptide, Figure 2c,d). These effects may instead be attributable to the neurological manifestations of Tspan7 ablation, including depressive behaviour.\n23\n We observed no change in circulating proinsulin levels (Figure 2e), suggesting normal insulin processing in Tspan7y/− β‐cells. Furthermore, when subjected to intra‐peritoneal glucose tolerance tests (IPGTTs), no difference in glucose tolerance (Figure 2f) or glucose‐induced insulin secretion (GSIS, Figure S1) was observed between Tspan7y/− and control mice.\nIn vivo metabolic phenotyping of Tspan7y/− mouse model. (a–e) Ad libitum bodyweight (a), and plasma concentrations of glucose (b), insulin (c), C‐peptide (d) and proinsulin measured in Tspan7y/− mice (red) and age‐matched (11.3 ± 0.3 wks) litter mate control (black). *p < 0.05 versus control and **p < 0.01 versus control. (f) Plasma glucose concentrations of Tspan7y/− mice (red) and age‐matched litter mate control (black) measured during intraperitoneal glucose tolerance tests. Glucose bolus was injected at 0 min. N = 5 for control and 6 for Tspan7y/−. Data are presented as mean value ± SEM.", "In β‐cells, glucose metabolism increases the intracellular ATP/ADP ratio, resulting in closure of ATP‐sensitive K+ channels (KATP‐channels) inducing membrane depolarisation and action potential (AP) firing. APs open voltage‐gated Ca2+ channels (VGCCs), enabling influx of Ca2+ to trigger Ca2+‐dependent exocytosis, culminating in insulin secretion. To interrogate the impact of Tspan7 ablation on β‐cell function at a single‐cell level, we phenotyped Tspan7y/− β‐cell excitability, transmembrane Ca2+ currents and exocytosis using electrophysiological techniques. In the presence of 1 mM glucose, Tspan7y/− and control β‐cells were both repolarised (Tspan7y/− vs. control: −80 ± 1 mV vs. −80 ± 2 mV; p = 0.4) and electrically silent. Elevating extracellular glucose or blocking the KATP channels (using tolbutamide, 200 μM) induced membrane depolarisation and firing of APs in β‐cells of both genotypes (Figure 3a). In the presence of 10 mM glucose, although the Tspan7y/− β‐cells were less depolarised (Tspan7y/− vs. control: −57 ± 4 mV vs. −46 ± 5 mV; p < 0.05), no difference in AP peak or firing frequency was observed across the two genotypes (Figure 3b–d). There was no apparent difference in AP halfwidth or spike duration, either (Figure S2). Further increasing extracellular glucose to 20 mM or inhibiting KATP channels with tolbutamide exerted similar effects on the electrical activity of both Tspan7y/− and control β‐cells (Figure 3b–d). However, Tspan7y/− β cells responded to 20 mM glucose stimulation faster than the control (Tspan7y/− vs. control: 2.4 ± 0.3 min vs. 3.85 ± 0.3 min, p < 0.05). These marginal changes in Tspan7y/− β‐cell electrical activity are not due to any apparent changes in KATP‐channel conductivity or glucose sensitivity (Figure 3e,f).\nElectrical activity and KATP‐channel conductance measured in Tspan7y/− β‐cells. (a) Membrane potential recordings of control (upper trace, black) and Tspan7y/− (lower trace, red) β‐cells within intact islets in response to changes in extracellular glucose (1 mM, 10 mM and 20 mM) or application of 200 μM tolbutamide, as indicated). (b) Summary of control (black) and Tspan7y/− (red) β‐cell membrane potential measured at conditions indicated. N = 7 for control and 14 for Tspan7y/− β‐cells at 1 mM glucose; N = 7 for control and 11 for Tspan7y/− β‐cells at 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 6 for Tspan7y/− β‐cells when 200 μM tolbutamide was applied. *p < 0.05 versus control. (c, d), as in (b) but summarise the peak voltage (c) and firing frequency (d) of action potentials measured in control (black) and Tspan7y/− (red) β‐cells at conditions indicated. N = 4 for control and 7 for Tspan7y/− β‐cells at 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 3 for Tspan7y/− β‐cells when tolbutamide was applied. (e) Summary of KATP‐channel conductance measured in control (black) and Tspan7y/− (red) β‐cells under the conditions indicated. N = 4 for control and 7 for Tspan7y/− β‐cells at 1 mM and 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 5 for Tspan7y/− β‐cells when tolbutamide was applied. (f) Examples of KATP currents measured in control (black, middle trace) and Tspan7y/− (red, bottom trace) β‐cells. KATP currents were triggered by ±10 mV excursions from −70 mV (200 ms, top) in voltage‐clamped β‐cells under the conditions indicated. All data are presented as mean value ± SEM.\nTransient knockdown of Tspan7 in β‐cells was recently shown to increase Ca2+ influx through L‐type VGCC.\n13\n We assessed whether a similar phenotype can be observed in β‐cells of mice with a constitutive Tspan7 knockout. As shown in Figure 4a,b, depolarisation triggered a significantly larger Ca2+ current in Tspan7y/− β‐cells than in the control (p < 0.01, ~160% at 0 and 10 mV), consistent with Dickerson et al.\n13\n As Ca2+ triggers exocytosis, we tested whether enhanced VGCC activity led to higher exocytosis in Tspan7y/− β‐cells using capacitance measurements. Exocytosis (measured as increases in cell capacitance) was trigged by a series of depolarising pulses from −70 mV to 0 mV with progressively longer durations (from 10 ms to 800 ms). Unexpectedly, exocytosis was comparable between Tspan7y/− and control β‐cells at all pulse durations (Figure 4c,d). We correlated the Ca2+ charges (total influx Ca2+ ions, QCa2+) with the corresponding exocytosis elicited by depolarisation (Figure 4e). Whereas exocytosis could be triggered by 0.15pC/pF QCa2+ in control β‐cells, it required > ~ 0.2pC/pF in Tspan7y/− β‐cells, suggesting a reduced exocytotic Ca2+sensitivity.\nCa2+ current and exocytosis in Tspan7y/− β‐cells. Tspan7 loss of function significantly increases Ca2+ current density in β cells (a, b). (a) Representative traces of control (black) and Tspan7y/− (red) β‐cell Ca2+ current elicited by a 100‐ms depolarisation from −70 mV to 0 mV. (b) Summary of control (black) and Tspan7y/− (red) β‐cell Ca2+ current density in relationship to membrane voltages. N = 7 for control and 5 for Tspan7y/− β‐cells, **p < 0.01 versus control, paired comparison and Tukey. (c) Examples of exocytosis induced by membrane depolarisations from −70 mV to 0 mV (at the duration of 100 ms, 200 ms, 300 ms, 500 ms and 800 ms as indicated above the traces) of control (black) and Tspan7y/− (red) β‐cells. (d) Summary of control (black) and Tspan7y/− (red) β‐cell exocytosis (normalised to cell size, fF/pF) in relationship to pulse durations. N = 11 cells for control and 14 cells for Tspan7y/− β‐cell. (e) Control (black) and Tspan7y/− (red) β‐cell charges‐exocytosis relationship for pulses up to 100 ms. Corresponding durations of the pulses are as labelled. N = 7 cells for control and 5 cells for Tspan7y/− β‐cells. (f) Insulin secretion measured from batch‐incubated control (black) and Tspan7y/− (red) islets in response to glucose or tolbutamide (concentrations as indicated). Islets were isolated from mice aged 20 weeks and N ≥ 3 for control and Tspan7y/− islets. **p < 0.01 and ***p < 0.001 versus 1 mM glucose alone of the same genotype. All data are presented as mean value ± SEM.\nGiven the limited changes in electrical activity and exocytosis in Tspan7y/− β‐cells, we reasoned that knocking‐out Tspan7 would have little effect on glucose/tolbutamide‐stimulated insulin secretion. Indeed, no significant difference was observed in insulin secretion, stimulated by 10 mM glucose or 0.2 mM tolbutamide, from islets of control and Tspan7y/− mice (Figure 4f). We reasoned that enhanced VGCC activity augments the concentration of cytosolic Ca2+, and thus may induce apoptosis,\n24\n affecting islet β cell mass in aged animals. However, no significant difference in GSIS, insulin secretion induced by 70 mM K+ or islet insulin content was observed in control and Tspan7y/− islets isolated from 1‐year‐old mice (Figure S4).", "To assess the role of tetraspanin‐7 in human β‐cell function, we utilised an adenovirus encoding shRNATSPAN7 (Ad‐ShTSPAN7) to knockdown (KD) human TSPAN7 expression. Islet cells transduced with Ad‐ShTSPAN7, or the control virus (with scrambled RNA), were used to form pseudo‐islets. RNA sequencing showed a marked reduction in TSPAN7 expression (92.65%) in the TSPAN7‐KD pseudo‐islets (log2[Fold‐change] = −3.765, p\nadj = 6.42 E‐13) (Figure 5a), confirming successful genetic manipulation. In total, 1030 genes (1000 are protein‐coding genes) were down‐regulated and 395 genes (372 are protein‐coding genes) were up‐regulated in TSPAN7‐KD pseudo‐islets. Pathway analyses showed up‐regulation in apoptosis gene ontology pathway (p\nadj = 4.08 E−7); and down‐regulation of genes involved in the secretory pathway and transport regulation (p\nadj = 5.62 E−9 and 4.22 E−08 respectively). Interestingly, β‐cell expression of the exocytosis Ca2+ sensor SYT7 was reduced in TSPAN7‐KD pseudo‐islets (log2[Fold‐change] = −0.39, p\nadj = 3.89 E−3). GSIS of the pseudo‐islets were tested using dynamic perifusion experiments (Figure 5b). Whereas insulin secretion at basal or 16.7 mM glucose was comparable between the TSPAN7‐KD and control pseudo‐islets, 20 mM K+‐stimulated insulin secretion was significantly lower in TSPAN7‐KD pseudo‐islets.\n\nTSPAN7 KD affects exocytosis from human pancreatic β‐cells. (a) Gene expressions that changed significantly in human TSPAN7 KD pseudo‐islet. Volcano plot shows that genes involved in the pathways of secretion (yellow area) and regulation of transport (green area) were down‐regulated; markers of the apoptotic pathway (blue) were up‐regulated. TSPAN7 KD was demonstrated by the marked reduction in TSPAN7 expression (red). (b) Dynamic insulin secretion measured from perifused human pseudo‐islets of TSPAN7 KD (red) or control (Scramble, black) in response to different glucose concentrations and 20 mM K+ as indicated. Inset: area of the curve (AUC) of insulin secretion stimulated by 20 mM K+. N = 3 for control and TSPAN7 KD, *p < 0.05 versus insulin secretion at 1 mM glucose in the same group of pseudo‐islets; †\np < 0.05 versus control (Scramble), Student's t‐test. (c) Examples of exocytosis of TSPAN7 KD EndoC‐βH1 (red) or control EndoC‐βH1 (Scramble, black) triggered by depolarisation from −70 to 0 mV with progressively increasing duration (as indicated). (d) Summary of control (black) and exocytosis (normalised to cell size, fF/pF) in relationship to pulse durations. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. (e) The relationship between depolarisation‐triggered charge influx and pulse durations of control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. *p < 0.05 versus control, student's t‐test. (f) The relationship between charges and exocytosis of control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. Corresponding durations of the pulses are as labelled. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. (g) Insulin secretion measured from batch‐incubated control (black) and TSPAN7 KD (red) EndoC‐βH1 cells in response to 1 and 20 mM glucose. Data are the summary of three independent biological repeats in technical triplicate. *p < 0.05 and ***p < 0.001 versus control insulin secretion at 1 mM glucose and ††\np < 0.01 versus control insulin secretion at 20 mM glucose; two‐way ANOVA and Tuckey post hoc test. (h) Insulin content measured from control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. Data are the summary of three independent biological repeats in technical triplicate. All data are presented as mean value ± SEM.\nThe insulin secretion experiments suggested that TSPAN7 may play a role in human β‐cell exocytosis. To test this, we performed capacitance measurements using EndoC‐βH1, a human β‐cell line recapturing functional and genetic features of primary human β‐cells,\n18\n transduced with Ad‐ShTSPAN7 or Ad‐Scramble virus. Similar to that observed in mouse Tspan7y/− β‐cells, exocytosis triggered by depolarisation was comparable between TSPAN7‐KD EndoC‐βH1 and control cells (Figure 5c,d). However, depolarisation triggered larger influx of charges (p < 0.05) in TSPAN7‐KD EndoC‐βH1 cells than that of the control (Ad‐Scramble) (Figure 5e). Importantly, the exocytotic Ca2+‐sensitivity of TSPAN7‐KD EndoC‐βH1 was reduced (same QCa2+ could only evoke exocytosis that was ~50% of the control, Figure 5f). This correlated with a significantly reduced GSIS in TSPAN7‐KD EndoC‐βH1 (p < 0.01, Figure 5g) without affecting insulin content (Figure 5h). We measured the changes in gene expression of VGCC and synaptotagmins in TSPAN7‐KD EndoC‐βH1. Concomitantly to TSPAN7 KD (p = 5.51 E−4), CACNA1A was up‐regulated and SYT5 and SYT7 were down‐regulated (−20%, Figure 6).\nChanges in VGCC‐ and synaptotagmin‐encoding genes in EndoC‐βH1 following TSPAN7 KD. The expression of genes encoding TSPAN7 (TSPAN7), VGCCs (CACNA1A, CACNA1C and CACNA1D) and synaptotagmins (SYT4, SYT5 and SYT7) measured from control (transduced with Ad‐Scramble, Scramble, black) and TSPAN7 KD (red) EndoC‐βH1 cells. The levels of expression are normalised to that in control cells (as 100%). Error bars of the control cells condition reflect the variation of the control across the biological repeats. Data are summary of measurements made in two to four independent biological replicates. ***p < 0.0001 versus control, Student's t‐test." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "What is already known?", "What did this study find?", "What are the implications of the study?", "INTRODUCTION", "METHODS", "Animals and isolation of pancreatic islets", "\nshRNA\n", "Human pseudo‐islets and EndoC‐βH1 cell line", "Insulin secretion", "Electrophysiology", "Histology", "Glucose tolerance test", "quantitative PCR\n", "\nRNA sequencing", "Data analyses and statistics", "RESULTS", "\nTspan7 is expressed in pancreatic islets", "Ablation of Tspan7 has limited impact on glucose tolerance", "Exocytosis sensitivity to Ca2+ was reduced in Tspan7y/− β‐cells", "Reducing \nTSPAN7\n expression in human islets changes gene expression of islet exocytosis Ca2+ sensors", "DISCUSSION", "CONFLICT OF INTEREST", "Supporting information" ]
[ "Tetraspanin‐7, a transmembrane protein widely expressed in pancreatic islet cells, was previously identified as an autoantigen in type 1 diabetes. Tetraspanin‐7 has been shown to regulate β‐cell L‐type Ca2+ channel activity.", "Genetic ablation of tetraspanin‐7 in mice led to a significant reduction in the Ca2+ dependence of β‐cell exocytosis. This phenotype was also observed in human islet cells following ex vivo tetraspanin‐7 knockdown, alongside down‐regulation of the β‐cell exocytosis Ca2+ sensor, synaptotagmin‐7.", "Tetraspanin‐7 expression is required for normal exocytotic machinery and appropriate exocytosis Ca2+ sensitivity in pancreatic β‐cells.", "Tetraspanin‐7 (Tspan7) was recently established as an autoantigen in type 1 diabetes mellitus (T1D),\n1\n an autoimmune disease in which loss of pancreatic β‐cells leads to chronic hyperglycaemia.\n2\n Proteins of the tetraspanin superfamily, of which Tspan7 is a member, are characterised by the presence of four transmembrane domains: one short and one long extracellular loop, a short intracellular loop, and N‐terminal and C‐terminal cytoplasmic tails.\n3\n, \n4\n Tetraspanin proteins are typically organised in tetraspanin‐enriched microdomains on biological membranes\n5\n where they act as molecular facilitators, engaging both transmembrane and intracellular proteins to regulate diverse processes such as cell proliferation, differentiation, activation, adhesion, motility and signalling.\n5\n, \n6\n, \n7\n\n\nTspan7 is highly expressed in neuroendocrine tissue,\n8\n and loss of Tspan7 can lead to defects in neuronal morphogenesis and synaptic transmission,\n9\n similar to those described in people expressing a mutated form.\n10\n Additional functions in cell morphology have been reported in dendritic cells\n11\n and osteoclasts,\n12\n attributed to its role in actin cytoskeleton rearrangement. Within the pancreas, Tspan7 is localised to the pancreatic islets and was recently shown to function as an auxiliary protein of L‐type voltage‐gated Ca2+ channels (VGCCs), modulating β‐cell electrical excitability and exocytosis. Knocking‐down of Tspan7 in β‐cells led to higher Ca2+ influx (through L‐type VGCCs), cellular excitability and glucose‐stimulated insulin secretion.\n13\n\n\nIn this study, we characterised Tspan7y/− mouse metabolic phenotype, β‐cell excitability, glucose‐stimulated insulin secretion and cell exocytosis. These data provide further insights of the role of Tspan7 in regulating β‐cell intracellular Ca2+ dynamics and insulin secretion, and thus to determine the importance of Tspan7 in glucose tolerance.", "[SUBTITLE] Animals and isolation of pancreatic islets [SUBSECTION] All animal experiments were conducted in accordance with the UK Animals Scientific Procedures Act (1986) and the University of Oxford ethical guidelines. Mouse islets were isolated as previously described.\n14\n, \n15\n\nTspan7 knockout mice (Tspan7y/−)\n9\n were gifts from Dr Luca Murru (CNR Institute of Neuroscience, Milano, Italy). Hormone secretion studies and the electrophysiology experiments were performed on islets isolated from Tspan7y/− mice and their wildtype littermates (control).\nAll animal experiments were conducted in accordance with the UK Animals Scientific Procedures Act (1986) and the University of Oxford ethical guidelines. Mouse islets were isolated as previously described.\n14\n, \n15\n\nTspan7 knockout mice (Tspan7y/−)\n9\n were gifts from Dr Luca Murru (CNR Institute of Neuroscience, Milano, Italy). Hormone secretion studies and the electrophysiology experiments were performed on islets isolated from Tspan7y/− mice and their wildtype littermates (control).\n[SUBTITLE] \nshRNA\n [SUBSECTION] Adenoviruses expressing shRNA for silencing of human TSPAN7 (Ad‐ShTSPAN7, SKU# shADV‐226,651) or scrambled shRNA control (Ad‐Scramble, SKU# 1122) together with eGFP under a CMV promoter were purchased from Vector Biolabs.\nAdenoviruses expressing shRNA for silencing of human TSPAN7 (Ad‐ShTSPAN7, SKU# shADV‐226,651) or scrambled shRNA control (Ad‐Scramble, SKU# 1122) together with eGFP under a CMV promoter were purchased from Vector Biolabs.\n[SUBTITLE] Human pseudo‐islets and EndoC‐βH1 cell line [SUBSECTION] Donor organs were obtained with written consent and research ethics approval at the University of Alberta, and human islets were isolated as previous described\n16\n at the University of Alberta. All human islet information is listed in Table S1. Islets were dispersed into a single‐cell suspension using trypsin (TryLE Express, Gibco) and transduced with Ad‐ShTSPAN7 or Ad‐Scramble before forming human pseudo‐islets using centrifugal‐forced aggregation as previously described.\n17\n EndoC‐βH1 was provided by Endocell and cultured as previously described.\n18\n\n\nDonor organs were obtained with written consent and research ethics approval at the University of Alberta, and human islets were isolated as previous described\n16\n at the University of Alberta. All human islet information is listed in Table S1. Islets were dispersed into a single‐cell suspension using trypsin (TryLE Express, Gibco) and transduced with Ad‐ShTSPAN7 or Ad‐Scramble before forming human pseudo‐islets using centrifugal‐forced aggregation as previously described.\n17\n EndoC‐βH1 was provided by Endocell and cultured as previously described.\n18\n\n\n[SUBTITLE] Insulin secretion [SUBSECTION] Insulin secretion was measured in static incubations as previously described.\n15\n Briefly, groups of 10–20 isolated islets (number depended on islet availability) were pre‐incubated for 30 min at 37°C in Krebs‐Ringer buffer (KRB) consisting of (mM): 140 NaCl, 3.6 KCl, 0.5 MgSO4, 2.6 CaCl2, 0.5 NaH2PO4, 2 NaHCO3, 1 glucose and 5 HEPES (pH = 7.4 with NaOH), before being incubated for 60 min at 37°C in 1 ml of KRB supplemented as indicated in the legends. Immediately after incubation, an aliquot of the medium was removed for determination of insulin concentration using an ELISA kit (Mercodia, Sweden).\nFor dynamic insulin secretion measurements, groups of 50 pseudo‐islets were perifused with KRB containing 1 or 16.7 mM glucose with or without 20 mM K+ (as indicated) at the rate of 100 μl/min, using a perifusion system (Biorep, FL, USA). Perfusate was collected every 2 min and insulin concentration was determined using the Alpco stellux human ELISA kits (Salem, NH).\nThe perifusion experiments were conducted at the University of Alberta and the mRNAs of pseudo‐islets were then extracted using TRIzol (ThermoFisher) before shipped to Oxford for transcriptomic study.\nEndoC‐βH1 cells were sequentially exposed for 20 min to 1 and 20 mM glucose containing KRB. Supernatants were collected at the end of each incubation and insulin cellular contents were extracted using acid ethanol. Insulin concentrations were determined using an ELISA kit (Mercodia, Sweden).\nInsulin secretion was measured in static incubations as previously described.\n15\n Briefly, groups of 10–20 isolated islets (number depended on islet availability) were pre‐incubated for 30 min at 37°C in Krebs‐Ringer buffer (KRB) consisting of (mM): 140 NaCl, 3.6 KCl, 0.5 MgSO4, 2.6 CaCl2, 0.5 NaH2PO4, 2 NaHCO3, 1 glucose and 5 HEPES (pH = 7.4 with NaOH), before being incubated for 60 min at 37°C in 1 ml of KRB supplemented as indicated in the legends. Immediately after incubation, an aliquot of the medium was removed for determination of insulin concentration using an ELISA kit (Mercodia, Sweden).\nFor dynamic insulin secretion measurements, groups of 50 pseudo‐islets were perifused with KRB containing 1 or 16.7 mM glucose with or without 20 mM K+ (as indicated) at the rate of 100 μl/min, using a perifusion system (Biorep, FL, USA). Perfusate was collected every 2 min and insulin concentration was determined using the Alpco stellux human ELISA kits (Salem, NH).\nThe perifusion experiments were conducted at the University of Alberta and the mRNAs of pseudo‐islets were then extracted using TRIzol (ThermoFisher) before shipped to Oxford for transcriptomic study.\nEndoC‐βH1 cells were sequentially exposed for 20 min to 1 and 20 mM glucose containing KRB. Supernatants were collected at the end of each incubation and insulin cellular contents were extracted using acid ethanol. Insulin concentrations were determined using an ELISA kit (Mercodia, Sweden).\n[SUBTITLE] Electrophysiology [SUBSECTION] Electrophysiological measurements were performed using an EPC‐10 patch‐clamp amplifier and Pulse software (version 8.80, HEKA Electronics, Germany). Electrical activity and KATP‐channel conductance were measured using perforated patch‐clamping technique as previously described\n19\n; membrane currents and changes in cell capacitance were recorded using voltage clamp in combination with capacitance measurements, as described in.\n14\n β‐cells were identified by electrophysiological fingerprinting.\n20\n\n\nElectrophysiological measurements were performed using an EPC‐10 patch‐clamp amplifier and Pulse software (version 8.80, HEKA Electronics, Germany). Electrical activity and KATP‐channel conductance were measured using perforated patch‐clamping technique as previously described\n19\n; membrane currents and changes in cell capacitance were recorded using voltage clamp in combination with capacitance measurements, as described in.\n14\n β‐cells were identified by electrophysiological fingerprinting.\n20\n\n\n[SUBTITLE] Histology [SUBSECTION] Sections of formalin‐fixed paraffin‐embedded tissue were de‐waxed and subjected to epitope retrieval in a microwave pressure cooker in 10 mM citric acid pH 6.0, 0.05% Tween 20. After blocking with 2.5% normal horse serum, sections were incubated overnight (4°C) with rabbit anti‐Tspan7 antibody (Anti‐TM4SF2, 1:50, Sigma‐Aldrich). Antibody labelling was detected with the VectaFluor™ Excel Amplified Anti‐Rabbit IgG, DyLight™ 488 Antibody Kit (Vector Laboratories). Slides were counterstained with guinea pig anti‐insulin (1:500, Dako) and mouse anti‐glucagon (1:500, Sigma) antibodies followed by labelling with goat anti‐guinea pig‐Alexa 633 (1:100) and goat anti‐mouse Alexa 555 (1:100) secondary antibodies (ThermoFisher). Slides were visualised by confocal microscopy.\nFor electron microscopy, isolated human islets were fixed in 2.5% glutaraldehyde in PBS, postfixed in 1% OsO4, dehydrated and embedded in Spurr's resin. Sections were cut onto nickel grids before being immunolabelled using anti‐TM4SF2 (1:20) and protein A gold particles (15 nm, Biocell), guinea pig anti‐insulin antibody (DAKO, 1:500) followed by anti‐guinea pig gold 10 nm (British Biocell International), and mouse anti‐glucagon (1:500, Sigma) followed by anti‐mouse pig gold 10 nm (British Biocell International). Sections were viewed with a Joel 1010 microscope (accelerating voltage 80 kV).\nTo establish the specificity of Tspan7 antibody, immunostaining was conducted using a stable Tspan7 KD INS‐1 cell line (cell line generation is described in Data S1). Tspan7 KD INS‐1 or control cells were fixed in 4% PFA, permeabilised using 0.1% Triton X‐100 and blocked with 5% normal swine serum. Tspan7 was detected using rabbit anti‐Tspan7 primary antibody (anti‐TM4SF2, 1:50, Sigma‐Aldrich; overnight incubation, 4°C), and with goat anti‐rabbit IgG Alexa Fluor 568 secondary antibody (Life Technologies). Nucleus were stained with RedDot2 (BioTium; 1:200 dilution). Slides were visualised by confocal microscopy (BioRad).\nSections of formalin‐fixed paraffin‐embedded tissue were de‐waxed and subjected to epitope retrieval in a microwave pressure cooker in 10 mM citric acid pH 6.0, 0.05% Tween 20. After blocking with 2.5% normal horse serum, sections were incubated overnight (4°C) with rabbit anti‐Tspan7 antibody (Anti‐TM4SF2, 1:50, Sigma‐Aldrich). Antibody labelling was detected with the VectaFluor™ Excel Amplified Anti‐Rabbit IgG, DyLight™ 488 Antibody Kit (Vector Laboratories). Slides were counterstained with guinea pig anti‐insulin (1:500, Dako) and mouse anti‐glucagon (1:500, Sigma) antibodies followed by labelling with goat anti‐guinea pig‐Alexa 633 (1:100) and goat anti‐mouse Alexa 555 (1:100) secondary antibodies (ThermoFisher). Slides were visualised by confocal microscopy.\nFor electron microscopy, isolated human islets were fixed in 2.5% glutaraldehyde in PBS, postfixed in 1% OsO4, dehydrated and embedded in Spurr's resin. Sections were cut onto nickel grids before being immunolabelled using anti‐TM4SF2 (1:20) and protein A gold particles (15 nm, Biocell), guinea pig anti‐insulin antibody (DAKO, 1:500) followed by anti‐guinea pig gold 10 nm (British Biocell International), and mouse anti‐glucagon (1:500, Sigma) followed by anti‐mouse pig gold 10 nm (British Biocell International). Sections were viewed with a Joel 1010 microscope (accelerating voltage 80 kV).\nTo establish the specificity of Tspan7 antibody, immunostaining was conducted using a stable Tspan7 KD INS‐1 cell line (cell line generation is described in Data S1). Tspan7 KD INS‐1 or control cells were fixed in 4% PFA, permeabilised using 0.1% Triton X‐100 and blocked with 5% normal swine serum. Tspan7 was detected using rabbit anti‐Tspan7 primary antibody (anti‐TM4SF2, 1:50, Sigma‐Aldrich; overnight incubation, 4°C), and with goat anti‐rabbit IgG Alexa Fluor 568 secondary antibody (Life Technologies). Nucleus were stained with RedDot2 (BioTium; 1:200 dilution). Slides were visualised by confocal microscopy (BioRad).\n[SUBTITLE] Glucose tolerance test [SUBSECTION] Intraperitoneal (i.p.) glucose tolerance test was performed as previously described.\n15\n Briefly, the animals were individually fasted for 6 h before i.p. injected with a bolus of glucose (2 g/kg body weight). Blood glucose were monitored for 90 min using a glucometer by tail vein sampling.\nIntraperitoneal (i.p.) glucose tolerance test was performed as previously described.\n15\n Briefly, the animals were individually fasted for 6 h before i.p. injected with a bolus of glucose (2 g/kg body weight). Blood glucose were monitored for 90 min using a glucometer by tail vein sampling.\n[SUBTITLE] quantitative PCR\n [SUBSECTION] RNA was extracted from human pseudo‐islets using TRIzol Reagent (Life Technologies, 15,596,026) according to the manufacturer's instructions. cDNA was generated using the GoScript Reverse Transcription Kit (Promega, A5000). qPCR was performed using 20 ng of cDNA, and TaqMan Gene Expression Master Mix (Life Technologies Ltd, 4,369,016) or SYBR™ Green PCR Master Mix (Applied Biosystems, 4,309,155) with the gene expression assays or primers as detailed in Table 1. Gene expression was determined using the ΔΔCT method by normalising to HouseKeeping Genes (HKG) and to the level of expression of each target detected in Ad‐Scramble transduced cells. Data are presented as percentage of control.\nPrimers and types of assays used for quantitative PCR analyses\nRNA was extracted from human pseudo‐islets using TRIzol Reagent (Life Technologies, 15,596,026) according to the manufacturer's instructions. cDNA was generated using the GoScript Reverse Transcription Kit (Promega, A5000). qPCR was performed using 20 ng of cDNA, and TaqMan Gene Expression Master Mix (Life Technologies Ltd, 4,369,016) or SYBR™ Green PCR Master Mix (Applied Biosystems, 4,309,155) with the gene expression assays or primers as detailed in Table 1. Gene expression was determined using the ΔΔCT method by normalising to HouseKeeping Genes (HKG) and to the level of expression of each target detected in Ad‐Scramble transduced cells. Data are presented as percentage of control.\nPrimers and types of assays used for quantitative PCR analyses\n[SUBTITLE] \nRNA sequencing [SUBSECTION] Human pseudo‐islets were preserved in Trizol before their total RNA was extracted using Quick‐RNA MicroPrep kit (R1050 Zymo Research). RNA libraries for RNA‐seq were prepared using NEBNext Ultra II Directional RNA Library Prep Kit for Illumina following manufacturer's protocols. Sequencing was performed on NextSeq 550. Single‐end reads were splice‐aligned to a human genome (GRCh38) using GSNAP. FeatureCounts was used to assign reads to exons thus eventually getting counts per gene. EdgeR package of R\n21\n was used to perform differential analysis between the conditions (shRNA vs control), while controlling for the confounding effect from the donor. Across‐samples normalisation was performed using the TMM normalisation method.\nHuman pseudo‐islets were preserved in Trizol before their total RNA was extracted using Quick‐RNA MicroPrep kit (R1050 Zymo Research). RNA libraries for RNA‐seq were prepared using NEBNext Ultra II Directional RNA Library Prep Kit for Illumina following manufacturer's protocols. Sequencing was performed on NextSeq 550. Single‐end reads were splice‐aligned to a human genome (GRCh38) using GSNAP. FeatureCounts was used to assign reads to exons thus eventually getting counts per gene. EdgeR package of R\n21\n was used to perform differential analysis between the conditions (shRNA vs control), while controlling for the confounding effect from the donor. Across‐samples normalisation was performed using the TMM normalisation method.\n[SUBTITLE] Data analyses and statistics [SUBSECTION] Data are expressed as mean value ± SEM. p‐values less than 0.05 were considered significant. Statistical analyses were performed using OriginPro 2017 either by Student's t‐test, paired comparison or two‐way ANOVA and Tukey post hoc test. Ns represent the number of cells for electrophysiological analyses, and of independent mice or cell passages for hormone assays and expression data.\nData are expressed as mean value ± SEM. p‐values less than 0.05 were considered significant. Statistical analyses were performed using OriginPro 2017 either by Student's t‐test, paired comparison or two‐way ANOVA and Tukey post hoc test. Ns represent the number of cells for electrophysiological analyses, and of independent mice or cell passages for hormone assays and expression data.", "All animal experiments were conducted in accordance with the UK Animals Scientific Procedures Act (1986) and the University of Oxford ethical guidelines. Mouse islets were isolated as previously described.\n14\n, \n15\n\nTspan7 knockout mice (Tspan7y/−)\n9\n were gifts from Dr Luca Murru (CNR Institute of Neuroscience, Milano, Italy). Hormone secretion studies and the electrophysiology experiments were performed on islets isolated from Tspan7y/− mice and their wildtype littermates (control).", "Adenoviruses expressing shRNA for silencing of human TSPAN7 (Ad‐ShTSPAN7, SKU# shADV‐226,651) or scrambled shRNA control (Ad‐Scramble, SKU# 1122) together with eGFP under a CMV promoter were purchased from Vector Biolabs.", "Donor organs were obtained with written consent and research ethics approval at the University of Alberta, and human islets were isolated as previous described\n16\n at the University of Alberta. All human islet information is listed in Table S1. Islets were dispersed into a single‐cell suspension using trypsin (TryLE Express, Gibco) and transduced with Ad‐ShTSPAN7 or Ad‐Scramble before forming human pseudo‐islets using centrifugal‐forced aggregation as previously described.\n17\n EndoC‐βH1 was provided by Endocell and cultured as previously described.\n18\n\n", "Insulin secretion was measured in static incubations as previously described.\n15\n Briefly, groups of 10–20 isolated islets (number depended on islet availability) were pre‐incubated for 30 min at 37°C in Krebs‐Ringer buffer (KRB) consisting of (mM): 140 NaCl, 3.6 KCl, 0.5 MgSO4, 2.6 CaCl2, 0.5 NaH2PO4, 2 NaHCO3, 1 glucose and 5 HEPES (pH = 7.4 with NaOH), before being incubated for 60 min at 37°C in 1 ml of KRB supplemented as indicated in the legends. Immediately after incubation, an aliquot of the medium was removed for determination of insulin concentration using an ELISA kit (Mercodia, Sweden).\nFor dynamic insulin secretion measurements, groups of 50 pseudo‐islets were perifused with KRB containing 1 or 16.7 mM glucose with or without 20 mM K+ (as indicated) at the rate of 100 μl/min, using a perifusion system (Biorep, FL, USA). Perfusate was collected every 2 min and insulin concentration was determined using the Alpco stellux human ELISA kits (Salem, NH).\nThe perifusion experiments were conducted at the University of Alberta and the mRNAs of pseudo‐islets were then extracted using TRIzol (ThermoFisher) before shipped to Oxford for transcriptomic study.\nEndoC‐βH1 cells were sequentially exposed for 20 min to 1 and 20 mM glucose containing KRB. Supernatants were collected at the end of each incubation and insulin cellular contents were extracted using acid ethanol. Insulin concentrations were determined using an ELISA kit (Mercodia, Sweden).", "Electrophysiological measurements were performed using an EPC‐10 patch‐clamp amplifier and Pulse software (version 8.80, HEKA Electronics, Germany). Electrical activity and KATP‐channel conductance were measured using perforated patch‐clamping technique as previously described\n19\n; membrane currents and changes in cell capacitance were recorded using voltage clamp in combination with capacitance measurements, as described in.\n14\n β‐cells were identified by electrophysiological fingerprinting.\n20\n\n", "Sections of formalin‐fixed paraffin‐embedded tissue were de‐waxed and subjected to epitope retrieval in a microwave pressure cooker in 10 mM citric acid pH 6.0, 0.05% Tween 20. After blocking with 2.5% normal horse serum, sections were incubated overnight (4°C) with rabbit anti‐Tspan7 antibody (Anti‐TM4SF2, 1:50, Sigma‐Aldrich). Antibody labelling was detected with the VectaFluor™ Excel Amplified Anti‐Rabbit IgG, DyLight™ 488 Antibody Kit (Vector Laboratories). Slides were counterstained with guinea pig anti‐insulin (1:500, Dako) and mouse anti‐glucagon (1:500, Sigma) antibodies followed by labelling with goat anti‐guinea pig‐Alexa 633 (1:100) and goat anti‐mouse Alexa 555 (1:100) secondary antibodies (ThermoFisher). Slides were visualised by confocal microscopy.\nFor electron microscopy, isolated human islets were fixed in 2.5% glutaraldehyde in PBS, postfixed in 1% OsO4, dehydrated and embedded in Spurr's resin. Sections were cut onto nickel grids before being immunolabelled using anti‐TM4SF2 (1:20) and protein A gold particles (15 nm, Biocell), guinea pig anti‐insulin antibody (DAKO, 1:500) followed by anti‐guinea pig gold 10 nm (British Biocell International), and mouse anti‐glucagon (1:500, Sigma) followed by anti‐mouse pig gold 10 nm (British Biocell International). Sections were viewed with a Joel 1010 microscope (accelerating voltage 80 kV).\nTo establish the specificity of Tspan7 antibody, immunostaining was conducted using a stable Tspan7 KD INS‐1 cell line (cell line generation is described in Data S1). Tspan7 KD INS‐1 or control cells were fixed in 4% PFA, permeabilised using 0.1% Triton X‐100 and blocked with 5% normal swine serum. Tspan7 was detected using rabbit anti‐Tspan7 primary antibody (anti‐TM4SF2, 1:50, Sigma‐Aldrich; overnight incubation, 4°C), and with goat anti‐rabbit IgG Alexa Fluor 568 secondary antibody (Life Technologies). Nucleus were stained with RedDot2 (BioTium; 1:200 dilution). Slides were visualised by confocal microscopy (BioRad).", "Intraperitoneal (i.p.) glucose tolerance test was performed as previously described.\n15\n Briefly, the animals were individually fasted for 6 h before i.p. injected with a bolus of glucose (2 g/kg body weight). Blood glucose were monitored for 90 min using a glucometer by tail vein sampling.", "RNA was extracted from human pseudo‐islets using TRIzol Reagent (Life Technologies, 15,596,026) according to the manufacturer's instructions. cDNA was generated using the GoScript Reverse Transcription Kit (Promega, A5000). qPCR was performed using 20 ng of cDNA, and TaqMan Gene Expression Master Mix (Life Technologies Ltd, 4,369,016) or SYBR™ Green PCR Master Mix (Applied Biosystems, 4,309,155) with the gene expression assays or primers as detailed in Table 1. Gene expression was determined using the ΔΔCT method by normalising to HouseKeeping Genes (HKG) and to the level of expression of each target detected in Ad‐Scramble transduced cells. Data are presented as percentage of control.\nPrimers and types of assays used for quantitative PCR analyses", "Human pseudo‐islets were preserved in Trizol before their total RNA was extracted using Quick‐RNA MicroPrep kit (R1050 Zymo Research). RNA libraries for RNA‐seq were prepared using NEBNext Ultra II Directional RNA Library Prep Kit for Illumina following manufacturer's protocols. Sequencing was performed on NextSeq 550. Single‐end reads were splice‐aligned to a human genome (GRCh38) using GSNAP. FeatureCounts was used to assign reads to exons thus eventually getting counts per gene. EdgeR package of R\n21\n was used to perform differential analysis between the conditions (shRNA vs control), while controlling for the confounding effect from the donor. Across‐samples normalisation was performed using the TMM normalisation method.", "Data are expressed as mean value ± SEM. p‐values less than 0.05 were considered significant. Statistical analyses were performed using OriginPro 2017 either by Student's t‐test, paired comparison or two‐way ANOVA and Tukey post hoc test. Ns represent the number of cells for electrophysiological analyses, and of independent mice or cell passages for hormone assays and expression data.", "[SUBTITLE] \nTspan7 is expressed in pancreatic islets [SUBSECTION] Tspan7 expression in pancreatic islets was evaluated using immunohistochemistry. In both human and mouse pancreatic islets, Tspan7 was found to colocalise with insulin and glucagon, confirming its expression in β‐ and α‐cells\n13\n (Figure 1a). We observed enriched immunogold labelling of Tspan7 in large dense‐core vesicle of human α‐ and β‐cells (Figure 1b,c), consistent with a previous report that Tspan7 was present in the cytoplasm of islet cells with a distribution pattern similar to islet hormones.\n22\n\n\nTetraspanin‐7 is expressed in pancreatic islets of Langerhans. (a) Immunofluorescent staining of human (upper) and mouse (lower) pancreas sections. Tetraspanin‐7 (TSPAN7 or Tspan7, red) is detected in β (Insulin, green)‐ and α (Glucagon, grey)‐cells. Scale bar: 30 μm. (b, c) Immunogold labelling of TSPAN7 (15 nm gold particles, black arrows), in glucagon (b) and insulin (c) containing vesicles in human islets. Insulin was labelled with 10 nm gold particles (white arrows). Scale bar: 500 nm.\nTspan7 expression in pancreatic islets was evaluated using immunohistochemistry. In both human and mouse pancreatic islets, Tspan7 was found to colocalise with insulin and glucagon, confirming its expression in β‐ and α‐cells\n13\n (Figure 1a). We observed enriched immunogold labelling of Tspan7 in large dense‐core vesicle of human α‐ and β‐cells (Figure 1b,c), consistent with a previous report that Tspan7 was present in the cytoplasm of islet cells with a distribution pattern similar to islet hormones.\n22\n\n\nTetraspanin‐7 is expressed in pancreatic islets of Langerhans. (a) Immunofluorescent staining of human (upper) and mouse (lower) pancreas sections. Tetraspanin‐7 (TSPAN7 or Tspan7, red) is detected in β (Insulin, green)‐ and α (Glucagon, grey)‐cells. Scale bar: 30 μm. (b, c) Immunogold labelling of TSPAN7 (15 nm gold particles, black arrows), in glucagon (b) and insulin (c) containing vesicles in human islets. Insulin was labelled with 10 nm gold particles (white arrows). Scale bar: 500 nm.\n[SUBTITLE] Ablation of Tspan7 has limited impact on glucose tolerance [SUBSECTION] To investigate the role of Tspan7 in islet function and systemic metabolism, a Tspan7 knockout mouse model (Tspan7y/−)\n9\n was used. Reduced body weight and ad libitum plasma glucose was observed in Tspan7y/− mice (Figure 2a,b), but this was not linked to changes in plasma insulin (measured as insulin and C‐peptide, Figure 2c,d). These effects may instead be attributable to the neurological manifestations of Tspan7 ablation, including depressive behaviour.\n23\n We observed no change in circulating proinsulin levels (Figure 2e), suggesting normal insulin processing in Tspan7y/− β‐cells. Furthermore, when subjected to intra‐peritoneal glucose tolerance tests (IPGTTs), no difference in glucose tolerance (Figure 2f) or glucose‐induced insulin secretion (GSIS, Figure S1) was observed between Tspan7y/− and control mice.\nIn vivo metabolic phenotyping of Tspan7y/− mouse model. (a–e) Ad libitum bodyweight (a), and plasma concentrations of glucose (b), insulin (c), C‐peptide (d) and proinsulin measured in Tspan7y/− mice (red) and age‐matched (11.3 ± 0.3 wks) litter mate control (black). *p < 0.05 versus control and **p < 0.01 versus control. (f) Plasma glucose concentrations of Tspan7y/− mice (red) and age‐matched litter mate control (black) measured during intraperitoneal glucose tolerance tests. Glucose bolus was injected at 0 min. N = 5 for control and 6 for Tspan7y/−. Data are presented as mean value ± SEM.\nTo investigate the role of Tspan7 in islet function and systemic metabolism, a Tspan7 knockout mouse model (Tspan7y/−)\n9\n was used. Reduced body weight and ad libitum plasma glucose was observed in Tspan7y/− mice (Figure 2a,b), but this was not linked to changes in plasma insulin (measured as insulin and C‐peptide, Figure 2c,d). These effects may instead be attributable to the neurological manifestations of Tspan7 ablation, including depressive behaviour.\n23\n We observed no change in circulating proinsulin levels (Figure 2e), suggesting normal insulin processing in Tspan7y/− β‐cells. Furthermore, when subjected to intra‐peritoneal glucose tolerance tests (IPGTTs), no difference in glucose tolerance (Figure 2f) or glucose‐induced insulin secretion (GSIS, Figure S1) was observed between Tspan7y/− and control mice.\nIn vivo metabolic phenotyping of Tspan7y/− mouse model. (a–e) Ad libitum bodyweight (a), and plasma concentrations of glucose (b), insulin (c), C‐peptide (d) and proinsulin measured in Tspan7y/− mice (red) and age‐matched (11.3 ± 0.3 wks) litter mate control (black). *p < 0.05 versus control and **p < 0.01 versus control. (f) Plasma glucose concentrations of Tspan7y/− mice (red) and age‐matched litter mate control (black) measured during intraperitoneal glucose tolerance tests. Glucose bolus was injected at 0 min. N = 5 for control and 6 for Tspan7y/−. Data are presented as mean value ± SEM.\n[SUBTITLE] Exocytosis sensitivity to Ca2+ was reduced in Tspan7y/− β‐cells [SUBSECTION] In β‐cells, glucose metabolism increases the intracellular ATP/ADP ratio, resulting in closure of ATP‐sensitive K+ channels (KATP‐channels) inducing membrane depolarisation and action potential (AP) firing. APs open voltage‐gated Ca2+ channels (VGCCs), enabling influx of Ca2+ to trigger Ca2+‐dependent exocytosis, culminating in insulin secretion. To interrogate the impact of Tspan7 ablation on β‐cell function at a single‐cell level, we phenotyped Tspan7y/− β‐cell excitability, transmembrane Ca2+ currents and exocytosis using electrophysiological techniques. In the presence of 1 mM glucose, Tspan7y/− and control β‐cells were both repolarised (Tspan7y/− vs. control: −80 ± 1 mV vs. −80 ± 2 mV; p = 0.4) and electrically silent. Elevating extracellular glucose or blocking the KATP channels (using tolbutamide, 200 μM) induced membrane depolarisation and firing of APs in β‐cells of both genotypes (Figure 3a). In the presence of 10 mM glucose, although the Tspan7y/− β‐cells were less depolarised (Tspan7y/− vs. control: −57 ± 4 mV vs. −46 ± 5 mV; p < 0.05), no difference in AP peak or firing frequency was observed across the two genotypes (Figure 3b–d). There was no apparent difference in AP halfwidth or spike duration, either (Figure S2). Further increasing extracellular glucose to 20 mM or inhibiting KATP channels with tolbutamide exerted similar effects on the electrical activity of both Tspan7y/− and control β‐cells (Figure 3b–d). However, Tspan7y/− β cells responded to 20 mM glucose stimulation faster than the control (Tspan7y/− vs. control: 2.4 ± 0.3 min vs. 3.85 ± 0.3 min, p < 0.05). These marginal changes in Tspan7y/− β‐cell electrical activity are not due to any apparent changes in KATP‐channel conductivity or glucose sensitivity (Figure 3e,f).\nElectrical activity and KATP‐channel conductance measured in Tspan7y/− β‐cells. (a) Membrane potential recordings of control (upper trace, black) and Tspan7y/− (lower trace, red) β‐cells within intact islets in response to changes in extracellular glucose (1 mM, 10 mM and 20 mM) or application of 200 μM tolbutamide, as indicated). (b) Summary of control (black) and Tspan7y/− (red) β‐cell membrane potential measured at conditions indicated. N = 7 for control and 14 for Tspan7y/− β‐cells at 1 mM glucose; N = 7 for control and 11 for Tspan7y/− β‐cells at 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 6 for Tspan7y/− β‐cells when 200 μM tolbutamide was applied. *p < 0.05 versus control. (c, d), as in (b) but summarise the peak voltage (c) and firing frequency (d) of action potentials measured in control (black) and Tspan7y/− (red) β‐cells at conditions indicated. N = 4 for control and 7 for Tspan7y/− β‐cells at 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 3 for Tspan7y/− β‐cells when tolbutamide was applied. (e) Summary of KATP‐channel conductance measured in control (black) and Tspan7y/− (red) β‐cells under the conditions indicated. N = 4 for control and 7 for Tspan7y/− β‐cells at 1 mM and 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 5 for Tspan7y/− β‐cells when tolbutamide was applied. (f) Examples of KATP currents measured in control (black, middle trace) and Tspan7y/− (red, bottom trace) β‐cells. KATP currents were triggered by ±10 mV excursions from −70 mV (200 ms, top) in voltage‐clamped β‐cells under the conditions indicated. All data are presented as mean value ± SEM.\nTransient knockdown of Tspan7 in β‐cells was recently shown to increase Ca2+ influx through L‐type VGCC.\n13\n We assessed whether a similar phenotype can be observed in β‐cells of mice with a constitutive Tspan7 knockout. As shown in Figure 4a,b, depolarisation triggered a significantly larger Ca2+ current in Tspan7y/− β‐cells than in the control (p < 0.01, ~160% at 0 and 10 mV), consistent with Dickerson et al.\n13\n As Ca2+ triggers exocytosis, we tested whether enhanced VGCC activity led to higher exocytosis in Tspan7y/− β‐cells using capacitance measurements. Exocytosis (measured as increases in cell capacitance) was trigged by a series of depolarising pulses from −70 mV to 0 mV with progressively longer durations (from 10 ms to 800 ms). Unexpectedly, exocytosis was comparable between Tspan7y/− and control β‐cells at all pulse durations (Figure 4c,d). We correlated the Ca2+ charges (total influx Ca2+ ions, QCa2+) with the corresponding exocytosis elicited by depolarisation (Figure 4e). Whereas exocytosis could be triggered by 0.15pC/pF QCa2+ in control β‐cells, it required > ~ 0.2pC/pF in Tspan7y/− β‐cells, suggesting a reduced exocytotic Ca2+sensitivity.\nCa2+ current and exocytosis in Tspan7y/− β‐cells. Tspan7 loss of function significantly increases Ca2+ current density in β cells (a, b). (a) Representative traces of control (black) and Tspan7y/− (red) β‐cell Ca2+ current elicited by a 100‐ms depolarisation from −70 mV to 0 mV. (b) Summary of control (black) and Tspan7y/− (red) β‐cell Ca2+ current density in relationship to membrane voltages. N = 7 for control and 5 for Tspan7y/− β‐cells, **p < 0.01 versus control, paired comparison and Tukey. (c) Examples of exocytosis induced by membrane depolarisations from −70 mV to 0 mV (at the duration of 100 ms, 200 ms, 300 ms, 500 ms and 800 ms as indicated above the traces) of control (black) and Tspan7y/− (red) β‐cells. (d) Summary of control (black) and Tspan7y/− (red) β‐cell exocytosis (normalised to cell size, fF/pF) in relationship to pulse durations. N = 11 cells for control and 14 cells for Tspan7y/− β‐cell. (e) Control (black) and Tspan7y/− (red) β‐cell charges‐exocytosis relationship for pulses up to 100 ms. Corresponding durations of the pulses are as labelled. N = 7 cells for control and 5 cells for Tspan7y/− β‐cells. (f) Insulin secretion measured from batch‐incubated control (black) and Tspan7y/− (red) islets in response to glucose or tolbutamide (concentrations as indicated). Islets were isolated from mice aged 20 weeks and N ≥ 3 for control and Tspan7y/− islets. **p < 0.01 and ***p < 0.001 versus 1 mM glucose alone of the same genotype. All data are presented as mean value ± SEM.\nGiven the limited changes in electrical activity and exocytosis in Tspan7y/− β‐cells, we reasoned that knocking‐out Tspan7 would have little effect on glucose/tolbutamide‐stimulated insulin secretion. Indeed, no significant difference was observed in insulin secretion, stimulated by 10 mM glucose or 0.2 mM tolbutamide, from islets of control and Tspan7y/− mice (Figure 4f). We reasoned that enhanced VGCC activity augments the concentration of cytosolic Ca2+, and thus may induce apoptosis,\n24\n affecting islet β cell mass in aged animals. However, no significant difference in GSIS, insulin secretion induced by 70 mM K+ or islet insulin content was observed in control and Tspan7y/− islets isolated from 1‐year‐old mice (Figure S4).\nIn β‐cells, glucose metabolism increases the intracellular ATP/ADP ratio, resulting in closure of ATP‐sensitive K+ channels (KATP‐channels) inducing membrane depolarisation and action potential (AP) firing. APs open voltage‐gated Ca2+ channels (VGCCs), enabling influx of Ca2+ to trigger Ca2+‐dependent exocytosis, culminating in insulin secretion. To interrogate the impact of Tspan7 ablation on β‐cell function at a single‐cell level, we phenotyped Tspan7y/− β‐cell excitability, transmembrane Ca2+ currents and exocytosis using electrophysiological techniques. In the presence of 1 mM glucose, Tspan7y/− and control β‐cells were both repolarised (Tspan7y/− vs. control: −80 ± 1 mV vs. −80 ± 2 mV; p = 0.4) and electrically silent. Elevating extracellular glucose or blocking the KATP channels (using tolbutamide, 200 μM) induced membrane depolarisation and firing of APs in β‐cells of both genotypes (Figure 3a). In the presence of 10 mM glucose, although the Tspan7y/− β‐cells were less depolarised (Tspan7y/− vs. control: −57 ± 4 mV vs. −46 ± 5 mV; p < 0.05), no difference in AP peak or firing frequency was observed across the two genotypes (Figure 3b–d). There was no apparent difference in AP halfwidth or spike duration, either (Figure S2). Further increasing extracellular glucose to 20 mM or inhibiting KATP channels with tolbutamide exerted similar effects on the electrical activity of both Tspan7y/− and control β‐cells (Figure 3b–d). However, Tspan7y/− β cells responded to 20 mM glucose stimulation faster than the control (Tspan7y/− vs. control: 2.4 ± 0.3 min vs. 3.85 ± 0.3 min, p < 0.05). These marginal changes in Tspan7y/− β‐cell electrical activity are not due to any apparent changes in KATP‐channel conductivity or glucose sensitivity (Figure 3e,f).\nElectrical activity and KATP‐channel conductance measured in Tspan7y/− β‐cells. (a) Membrane potential recordings of control (upper trace, black) and Tspan7y/− (lower trace, red) β‐cells within intact islets in response to changes in extracellular glucose (1 mM, 10 mM and 20 mM) or application of 200 μM tolbutamide, as indicated). (b) Summary of control (black) and Tspan7y/− (red) β‐cell membrane potential measured at conditions indicated. N = 7 for control and 14 for Tspan7y/− β‐cells at 1 mM glucose; N = 7 for control and 11 for Tspan7y/− β‐cells at 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 6 for Tspan7y/− β‐cells when 200 μM tolbutamide was applied. *p < 0.05 versus control. (c, d), as in (b) but summarise the peak voltage (c) and firing frequency (d) of action potentials measured in control (black) and Tspan7y/− (red) β‐cells at conditions indicated. N = 4 for control and 7 for Tspan7y/− β‐cells at 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 3 for Tspan7y/− β‐cells when tolbutamide was applied. (e) Summary of KATP‐channel conductance measured in control (black) and Tspan7y/− (red) β‐cells under the conditions indicated. N = 4 for control and 7 for Tspan7y/− β‐cells at 1 mM and 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 5 for Tspan7y/− β‐cells when tolbutamide was applied. (f) Examples of KATP currents measured in control (black, middle trace) and Tspan7y/− (red, bottom trace) β‐cells. KATP currents were triggered by ±10 mV excursions from −70 mV (200 ms, top) in voltage‐clamped β‐cells under the conditions indicated. All data are presented as mean value ± SEM.\nTransient knockdown of Tspan7 in β‐cells was recently shown to increase Ca2+ influx through L‐type VGCC.\n13\n We assessed whether a similar phenotype can be observed in β‐cells of mice with a constitutive Tspan7 knockout. As shown in Figure 4a,b, depolarisation triggered a significantly larger Ca2+ current in Tspan7y/− β‐cells than in the control (p < 0.01, ~160% at 0 and 10 mV), consistent with Dickerson et al.\n13\n As Ca2+ triggers exocytosis, we tested whether enhanced VGCC activity led to higher exocytosis in Tspan7y/− β‐cells using capacitance measurements. Exocytosis (measured as increases in cell capacitance) was trigged by a series of depolarising pulses from −70 mV to 0 mV with progressively longer durations (from 10 ms to 800 ms). Unexpectedly, exocytosis was comparable between Tspan7y/− and control β‐cells at all pulse durations (Figure 4c,d). We correlated the Ca2+ charges (total influx Ca2+ ions, QCa2+) with the corresponding exocytosis elicited by depolarisation (Figure 4e). Whereas exocytosis could be triggered by 0.15pC/pF QCa2+ in control β‐cells, it required > ~ 0.2pC/pF in Tspan7y/− β‐cells, suggesting a reduced exocytotic Ca2+sensitivity.\nCa2+ current and exocytosis in Tspan7y/− β‐cells. Tspan7 loss of function significantly increases Ca2+ current density in β cells (a, b). (a) Representative traces of control (black) and Tspan7y/− (red) β‐cell Ca2+ current elicited by a 100‐ms depolarisation from −70 mV to 0 mV. (b) Summary of control (black) and Tspan7y/− (red) β‐cell Ca2+ current density in relationship to membrane voltages. N = 7 for control and 5 for Tspan7y/− β‐cells, **p < 0.01 versus control, paired comparison and Tukey. (c) Examples of exocytosis induced by membrane depolarisations from −70 mV to 0 mV (at the duration of 100 ms, 200 ms, 300 ms, 500 ms and 800 ms as indicated above the traces) of control (black) and Tspan7y/− (red) β‐cells. (d) Summary of control (black) and Tspan7y/− (red) β‐cell exocytosis (normalised to cell size, fF/pF) in relationship to pulse durations. N = 11 cells for control and 14 cells for Tspan7y/− β‐cell. (e) Control (black) and Tspan7y/− (red) β‐cell charges‐exocytosis relationship for pulses up to 100 ms. Corresponding durations of the pulses are as labelled. N = 7 cells for control and 5 cells for Tspan7y/− β‐cells. (f) Insulin secretion measured from batch‐incubated control (black) and Tspan7y/− (red) islets in response to glucose or tolbutamide (concentrations as indicated). Islets were isolated from mice aged 20 weeks and N ≥ 3 for control and Tspan7y/− islets. **p < 0.01 and ***p < 0.001 versus 1 mM glucose alone of the same genotype. All data are presented as mean value ± SEM.\nGiven the limited changes in electrical activity and exocytosis in Tspan7y/− β‐cells, we reasoned that knocking‐out Tspan7 would have little effect on glucose/tolbutamide‐stimulated insulin secretion. Indeed, no significant difference was observed in insulin secretion, stimulated by 10 mM glucose or 0.2 mM tolbutamide, from islets of control and Tspan7y/− mice (Figure 4f). We reasoned that enhanced VGCC activity augments the concentration of cytosolic Ca2+, and thus may induce apoptosis,\n24\n affecting islet β cell mass in aged animals. However, no significant difference in GSIS, insulin secretion induced by 70 mM K+ or islet insulin content was observed in control and Tspan7y/− islets isolated from 1‐year‐old mice (Figure S4).\n[SUBTITLE] Reducing \nTSPAN7\n expression in human islets changes gene expression of islet exocytosis Ca2+ sensors [SUBSECTION] To assess the role of tetraspanin‐7 in human β‐cell function, we utilised an adenovirus encoding shRNATSPAN7 (Ad‐ShTSPAN7) to knockdown (KD) human TSPAN7 expression. Islet cells transduced with Ad‐ShTSPAN7, or the control virus (with scrambled RNA), were used to form pseudo‐islets. RNA sequencing showed a marked reduction in TSPAN7 expression (92.65%) in the TSPAN7‐KD pseudo‐islets (log2[Fold‐change] = −3.765, p\nadj = 6.42 E‐13) (Figure 5a), confirming successful genetic manipulation. In total, 1030 genes (1000 are protein‐coding genes) were down‐regulated and 395 genes (372 are protein‐coding genes) were up‐regulated in TSPAN7‐KD pseudo‐islets. Pathway analyses showed up‐regulation in apoptosis gene ontology pathway (p\nadj = 4.08 E−7); and down‐regulation of genes involved in the secretory pathway and transport regulation (p\nadj = 5.62 E−9 and 4.22 E−08 respectively). Interestingly, β‐cell expression of the exocytosis Ca2+ sensor SYT7 was reduced in TSPAN7‐KD pseudo‐islets (log2[Fold‐change] = −0.39, p\nadj = 3.89 E−3). GSIS of the pseudo‐islets were tested using dynamic perifusion experiments (Figure 5b). Whereas insulin secretion at basal or 16.7 mM glucose was comparable between the TSPAN7‐KD and control pseudo‐islets, 20 mM K+‐stimulated insulin secretion was significantly lower in TSPAN7‐KD pseudo‐islets.\n\nTSPAN7 KD affects exocytosis from human pancreatic β‐cells. (a) Gene expressions that changed significantly in human TSPAN7 KD pseudo‐islet. Volcano plot shows that genes involved in the pathways of secretion (yellow area) and regulation of transport (green area) were down‐regulated; markers of the apoptotic pathway (blue) were up‐regulated. TSPAN7 KD was demonstrated by the marked reduction in TSPAN7 expression (red). (b) Dynamic insulin secretion measured from perifused human pseudo‐islets of TSPAN7 KD (red) or control (Scramble, black) in response to different glucose concentrations and 20 mM K+ as indicated. Inset: area of the curve (AUC) of insulin secretion stimulated by 20 mM K+. N = 3 for control and TSPAN7 KD, *p < 0.05 versus insulin secretion at 1 mM glucose in the same group of pseudo‐islets; †\np < 0.05 versus control (Scramble), Student's t‐test. (c) Examples of exocytosis of TSPAN7 KD EndoC‐βH1 (red) or control EndoC‐βH1 (Scramble, black) triggered by depolarisation from −70 to 0 mV with progressively increasing duration (as indicated). (d) Summary of control (black) and exocytosis (normalised to cell size, fF/pF) in relationship to pulse durations. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. (e) The relationship between depolarisation‐triggered charge influx and pulse durations of control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. *p < 0.05 versus control, student's t‐test. (f) The relationship between charges and exocytosis of control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. Corresponding durations of the pulses are as labelled. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. (g) Insulin secretion measured from batch‐incubated control (black) and TSPAN7 KD (red) EndoC‐βH1 cells in response to 1 and 20 mM glucose. Data are the summary of three independent biological repeats in technical triplicate. *p < 0.05 and ***p < 0.001 versus control insulin secretion at 1 mM glucose and ††\np < 0.01 versus control insulin secretion at 20 mM glucose; two‐way ANOVA and Tuckey post hoc test. (h) Insulin content measured from control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. Data are the summary of three independent biological repeats in technical triplicate. All data are presented as mean value ± SEM.\nThe insulin secretion experiments suggested that TSPAN7 may play a role in human β‐cell exocytosis. To test this, we performed capacitance measurements using EndoC‐βH1, a human β‐cell line recapturing functional and genetic features of primary human β‐cells,\n18\n transduced with Ad‐ShTSPAN7 or Ad‐Scramble virus. Similar to that observed in mouse Tspan7y/− β‐cells, exocytosis triggered by depolarisation was comparable between TSPAN7‐KD EndoC‐βH1 and control cells (Figure 5c,d). However, depolarisation triggered larger influx of charges (p < 0.05) in TSPAN7‐KD EndoC‐βH1 cells than that of the control (Ad‐Scramble) (Figure 5e). Importantly, the exocytotic Ca2+‐sensitivity of TSPAN7‐KD EndoC‐βH1 was reduced (same QCa2+ could only evoke exocytosis that was ~50% of the control, Figure 5f). This correlated with a significantly reduced GSIS in TSPAN7‐KD EndoC‐βH1 (p < 0.01, Figure 5g) without affecting insulin content (Figure 5h). We measured the changes in gene expression of VGCC and synaptotagmins in TSPAN7‐KD EndoC‐βH1. Concomitantly to TSPAN7 KD (p = 5.51 E−4), CACNA1A was up‐regulated and SYT5 and SYT7 were down‐regulated (−20%, Figure 6).\nChanges in VGCC‐ and synaptotagmin‐encoding genes in EndoC‐βH1 following TSPAN7 KD. The expression of genes encoding TSPAN7 (TSPAN7), VGCCs (CACNA1A, CACNA1C and CACNA1D) and synaptotagmins (SYT4, SYT5 and SYT7) measured from control (transduced with Ad‐Scramble, Scramble, black) and TSPAN7 KD (red) EndoC‐βH1 cells. The levels of expression are normalised to that in control cells (as 100%). Error bars of the control cells condition reflect the variation of the control across the biological repeats. Data are summary of measurements made in two to four independent biological replicates. ***p < 0.0001 versus control, Student's t‐test.\nTo assess the role of tetraspanin‐7 in human β‐cell function, we utilised an adenovirus encoding shRNATSPAN7 (Ad‐ShTSPAN7) to knockdown (KD) human TSPAN7 expression. Islet cells transduced with Ad‐ShTSPAN7, or the control virus (with scrambled RNA), were used to form pseudo‐islets. RNA sequencing showed a marked reduction in TSPAN7 expression (92.65%) in the TSPAN7‐KD pseudo‐islets (log2[Fold‐change] = −3.765, p\nadj = 6.42 E‐13) (Figure 5a), confirming successful genetic manipulation. In total, 1030 genes (1000 are protein‐coding genes) were down‐regulated and 395 genes (372 are protein‐coding genes) were up‐regulated in TSPAN7‐KD pseudo‐islets. Pathway analyses showed up‐regulation in apoptosis gene ontology pathway (p\nadj = 4.08 E−7); and down‐regulation of genes involved in the secretory pathway and transport regulation (p\nadj = 5.62 E−9 and 4.22 E−08 respectively). Interestingly, β‐cell expression of the exocytosis Ca2+ sensor SYT7 was reduced in TSPAN7‐KD pseudo‐islets (log2[Fold‐change] = −0.39, p\nadj = 3.89 E−3). GSIS of the pseudo‐islets were tested using dynamic perifusion experiments (Figure 5b). Whereas insulin secretion at basal or 16.7 mM glucose was comparable between the TSPAN7‐KD and control pseudo‐islets, 20 mM K+‐stimulated insulin secretion was significantly lower in TSPAN7‐KD pseudo‐islets.\n\nTSPAN7 KD affects exocytosis from human pancreatic β‐cells. (a) Gene expressions that changed significantly in human TSPAN7 KD pseudo‐islet. Volcano plot shows that genes involved in the pathways of secretion (yellow area) and regulation of transport (green area) were down‐regulated; markers of the apoptotic pathway (blue) were up‐regulated. TSPAN7 KD was demonstrated by the marked reduction in TSPAN7 expression (red). (b) Dynamic insulin secretion measured from perifused human pseudo‐islets of TSPAN7 KD (red) or control (Scramble, black) in response to different glucose concentrations and 20 mM K+ as indicated. Inset: area of the curve (AUC) of insulin secretion stimulated by 20 mM K+. N = 3 for control and TSPAN7 KD, *p < 0.05 versus insulin secretion at 1 mM glucose in the same group of pseudo‐islets; †\np < 0.05 versus control (Scramble), Student's t‐test. (c) Examples of exocytosis of TSPAN7 KD EndoC‐βH1 (red) or control EndoC‐βH1 (Scramble, black) triggered by depolarisation from −70 to 0 mV with progressively increasing duration (as indicated). (d) Summary of control (black) and exocytosis (normalised to cell size, fF/pF) in relationship to pulse durations. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. (e) The relationship between depolarisation‐triggered charge influx and pulse durations of control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. *p < 0.05 versus control, student's t‐test. (f) The relationship between charges and exocytosis of control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. Corresponding durations of the pulses are as labelled. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. (g) Insulin secretion measured from batch‐incubated control (black) and TSPAN7 KD (red) EndoC‐βH1 cells in response to 1 and 20 mM glucose. Data are the summary of three independent biological repeats in technical triplicate. *p < 0.05 and ***p < 0.001 versus control insulin secretion at 1 mM glucose and ††\np < 0.01 versus control insulin secretion at 20 mM glucose; two‐way ANOVA and Tuckey post hoc test. (h) Insulin content measured from control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. Data are the summary of three independent biological repeats in technical triplicate. All data are presented as mean value ± SEM.\nThe insulin secretion experiments suggested that TSPAN7 may play a role in human β‐cell exocytosis. To test this, we performed capacitance measurements using EndoC‐βH1, a human β‐cell line recapturing functional and genetic features of primary human β‐cells,\n18\n transduced with Ad‐ShTSPAN7 or Ad‐Scramble virus. Similar to that observed in mouse Tspan7y/− β‐cells, exocytosis triggered by depolarisation was comparable between TSPAN7‐KD EndoC‐βH1 and control cells (Figure 5c,d). However, depolarisation triggered larger influx of charges (p < 0.05) in TSPAN7‐KD EndoC‐βH1 cells than that of the control (Ad‐Scramble) (Figure 5e). Importantly, the exocytotic Ca2+‐sensitivity of TSPAN7‐KD EndoC‐βH1 was reduced (same QCa2+ could only evoke exocytosis that was ~50% of the control, Figure 5f). This correlated with a significantly reduced GSIS in TSPAN7‐KD EndoC‐βH1 (p < 0.01, Figure 5g) without affecting insulin content (Figure 5h). We measured the changes in gene expression of VGCC and synaptotagmins in TSPAN7‐KD EndoC‐βH1. Concomitantly to TSPAN7 KD (p = 5.51 E−4), CACNA1A was up‐regulated and SYT5 and SYT7 were down‐regulated (−20%, Figure 6).\nChanges in VGCC‐ and synaptotagmin‐encoding genes in EndoC‐βH1 following TSPAN7 KD. The expression of genes encoding TSPAN7 (TSPAN7), VGCCs (CACNA1A, CACNA1C and CACNA1D) and synaptotagmins (SYT4, SYT5 and SYT7) measured from control (transduced with Ad‐Scramble, Scramble, black) and TSPAN7 KD (red) EndoC‐βH1 cells. The levels of expression are normalised to that in control cells (as 100%). Error bars of the control cells condition reflect the variation of the control across the biological repeats. Data are summary of measurements made in two to four independent biological replicates. ***p < 0.0001 versus control, Student's t‐test.", "Tspan7 expression in pancreatic islets was evaluated using immunohistochemistry. In both human and mouse pancreatic islets, Tspan7 was found to colocalise with insulin and glucagon, confirming its expression in β‐ and α‐cells\n13\n (Figure 1a). We observed enriched immunogold labelling of Tspan7 in large dense‐core vesicle of human α‐ and β‐cells (Figure 1b,c), consistent with a previous report that Tspan7 was present in the cytoplasm of islet cells with a distribution pattern similar to islet hormones.\n22\n\n\nTetraspanin‐7 is expressed in pancreatic islets of Langerhans. (a) Immunofluorescent staining of human (upper) and mouse (lower) pancreas sections. Tetraspanin‐7 (TSPAN7 or Tspan7, red) is detected in β (Insulin, green)‐ and α (Glucagon, grey)‐cells. Scale bar: 30 μm. (b, c) Immunogold labelling of TSPAN7 (15 nm gold particles, black arrows), in glucagon (b) and insulin (c) containing vesicles in human islets. Insulin was labelled with 10 nm gold particles (white arrows). Scale bar: 500 nm.", "To investigate the role of Tspan7 in islet function and systemic metabolism, a Tspan7 knockout mouse model (Tspan7y/−)\n9\n was used. Reduced body weight and ad libitum plasma glucose was observed in Tspan7y/− mice (Figure 2a,b), but this was not linked to changes in plasma insulin (measured as insulin and C‐peptide, Figure 2c,d). These effects may instead be attributable to the neurological manifestations of Tspan7 ablation, including depressive behaviour.\n23\n We observed no change in circulating proinsulin levels (Figure 2e), suggesting normal insulin processing in Tspan7y/− β‐cells. Furthermore, when subjected to intra‐peritoneal glucose tolerance tests (IPGTTs), no difference in glucose tolerance (Figure 2f) or glucose‐induced insulin secretion (GSIS, Figure S1) was observed between Tspan7y/− and control mice.\nIn vivo metabolic phenotyping of Tspan7y/− mouse model. (a–e) Ad libitum bodyweight (a), and plasma concentrations of glucose (b), insulin (c), C‐peptide (d) and proinsulin measured in Tspan7y/− mice (red) and age‐matched (11.3 ± 0.3 wks) litter mate control (black). *p < 0.05 versus control and **p < 0.01 versus control. (f) Plasma glucose concentrations of Tspan7y/− mice (red) and age‐matched litter mate control (black) measured during intraperitoneal glucose tolerance tests. Glucose bolus was injected at 0 min. N = 5 for control and 6 for Tspan7y/−. Data are presented as mean value ± SEM.", "In β‐cells, glucose metabolism increases the intracellular ATP/ADP ratio, resulting in closure of ATP‐sensitive K+ channels (KATP‐channels) inducing membrane depolarisation and action potential (AP) firing. APs open voltage‐gated Ca2+ channels (VGCCs), enabling influx of Ca2+ to trigger Ca2+‐dependent exocytosis, culminating in insulin secretion. To interrogate the impact of Tspan7 ablation on β‐cell function at a single‐cell level, we phenotyped Tspan7y/− β‐cell excitability, transmembrane Ca2+ currents and exocytosis using electrophysiological techniques. In the presence of 1 mM glucose, Tspan7y/− and control β‐cells were both repolarised (Tspan7y/− vs. control: −80 ± 1 mV vs. −80 ± 2 mV; p = 0.4) and electrically silent. Elevating extracellular glucose or blocking the KATP channels (using tolbutamide, 200 μM) induced membrane depolarisation and firing of APs in β‐cells of both genotypes (Figure 3a). In the presence of 10 mM glucose, although the Tspan7y/− β‐cells were less depolarised (Tspan7y/− vs. control: −57 ± 4 mV vs. −46 ± 5 mV; p < 0.05), no difference in AP peak or firing frequency was observed across the two genotypes (Figure 3b–d). There was no apparent difference in AP halfwidth or spike duration, either (Figure S2). Further increasing extracellular glucose to 20 mM or inhibiting KATP channels with tolbutamide exerted similar effects on the electrical activity of both Tspan7y/− and control β‐cells (Figure 3b–d). However, Tspan7y/− β cells responded to 20 mM glucose stimulation faster than the control (Tspan7y/− vs. control: 2.4 ± 0.3 min vs. 3.85 ± 0.3 min, p < 0.05). These marginal changes in Tspan7y/− β‐cell electrical activity are not due to any apparent changes in KATP‐channel conductivity or glucose sensitivity (Figure 3e,f).\nElectrical activity and KATP‐channel conductance measured in Tspan7y/− β‐cells. (a) Membrane potential recordings of control (upper trace, black) and Tspan7y/− (lower trace, red) β‐cells within intact islets in response to changes in extracellular glucose (1 mM, 10 mM and 20 mM) or application of 200 μM tolbutamide, as indicated). (b) Summary of control (black) and Tspan7y/− (red) β‐cell membrane potential measured at conditions indicated. N = 7 for control and 14 for Tspan7y/− β‐cells at 1 mM glucose; N = 7 for control and 11 for Tspan7y/− β‐cells at 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 6 for Tspan7y/− β‐cells when 200 μM tolbutamide was applied. *p < 0.05 versus control. (c, d), as in (b) but summarise the peak voltage (c) and firing frequency (d) of action potentials measured in control (black) and Tspan7y/− (red) β‐cells at conditions indicated. N = 4 for control and 7 for Tspan7y/− β‐cells at 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 3 for Tspan7y/− β‐cells when tolbutamide was applied. (e) Summary of KATP‐channel conductance measured in control (black) and Tspan7y/− (red) β‐cells under the conditions indicated. N = 4 for control and 7 for Tspan7y/− β‐cells at 1 mM and 10 mM glucose; N = 4 for control and 6 for Tspan7y/− β‐cells at 20 mM glucose and N = 3 for control and 5 for Tspan7y/− β‐cells when tolbutamide was applied. (f) Examples of KATP currents measured in control (black, middle trace) and Tspan7y/− (red, bottom trace) β‐cells. KATP currents were triggered by ±10 mV excursions from −70 mV (200 ms, top) in voltage‐clamped β‐cells under the conditions indicated. All data are presented as mean value ± SEM.\nTransient knockdown of Tspan7 in β‐cells was recently shown to increase Ca2+ influx through L‐type VGCC.\n13\n We assessed whether a similar phenotype can be observed in β‐cells of mice with a constitutive Tspan7 knockout. As shown in Figure 4a,b, depolarisation triggered a significantly larger Ca2+ current in Tspan7y/− β‐cells than in the control (p < 0.01, ~160% at 0 and 10 mV), consistent with Dickerson et al.\n13\n As Ca2+ triggers exocytosis, we tested whether enhanced VGCC activity led to higher exocytosis in Tspan7y/− β‐cells using capacitance measurements. Exocytosis (measured as increases in cell capacitance) was trigged by a series of depolarising pulses from −70 mV to 0 mV with progressively longer durations (from 10 ms to 800 ms). Unexpectedly, exocytosis was comparable between Tspan7y/− and control β‐cells at all pulse durations (Figure 4c,d). We correlated the Ca2+ charges (total influx Ca2+ ions, QCa2+) with the corresponding exocytosis elicited by depolarisation (Figure 4e). Whereas exocytosis could be triggered by 0.15pC/pF QCa2+ in control β‐cells, it required > ~ 0.2pC/pF in Tspan7y/− β‐cells, suggesting a reduced exocytotic Ca2+sensitivity.\nCa2+ current and exocytosis in Tspan7y/− β‐cells. Tspan7 loss of function significantly increases Ca2+ current density in β cells (a, b). (a) Representative traces of control (black) and Tspan7y/− (red) β‐cell Ca2+ current elicited by a 100‐ms depolarisation from −70 mV to 0 mV. (b) Summary of control (black) and Tspan7y/− (red) β‐cell Ca2+ current density in relationship to membrane voltages. N = 7 for control and 5 for Tspan7y/− β‐cells, **p < 0.01 versus control, paired comparison and Tukey. (c) Examples of exocytosis induced by membrane depolarisations from −70 mV to 0 mV (at the duration of 100 ms, 200 ms, 300 ms, 500 ms and 800 ms as indicated above the traces) of control (black) and Tspan7y/− (red) β‐cells. (d) Summary of control (black) and Tspan7y/− (red) β‐cell exocytosis (normalised to cell size, fF/pF) in relationship to pulse durations. N = 11 cells for control and 14 cells for Tspan7y/− β‐cell. (e) Control (black) and Tspan7y/− (red) β‐cell charges‐exocytosis relationship for pulses up to 100 ms. Corresponding durations of the pulses are as labelled. N = 7 cells for control and 5 cells for Tspan7y/− β‐cells. (f) Insulin secretion measured from batch‐incubated control (black) and Tspan7y/− (red) islets in response to glucose or tolbutamide (concentrations as indicated). Islets were isolated from mice aged 20 weeks and N ≥ 3 for control and Tspan7y/− islets. **p < 0.01 and ***p < 0.001 versus 1 mM glucose alone of the same genotype. All data are presented as mean value ± SEM.\nGiven the limited changes in electrical activity and exocytosis in Tspan7y/− β‐cells, we reasoned that knocking‐out Tspan7 would have little effect on glucose/tolbutamide‐stimulated insulin secretion. Indeed, no significant difference was observed in insulin secretion, stimulated by 10 mM glucose or 0.2 mM tolbutamide, from islets of control and Tspan7y/− mice (Figure 4f). We reasoned that enhanced VGCC activity augments the concentration of cytosolic Ca2+, and thus may induce apoptosis,\n24\n affecting islet β cell mass in aged animals. However, no significant difference in GSIS, insulin secretion induced by 70 mM K+ or islet insulin content was observed in control and Tspan7y/− islets isolated from 1‐year‐old mice (Figure S4).", "To assess the role of tetraspanin‐7 in human β‐cell function, we utilised an adenovirus encoding shRNATSPAN7 (Ad‐ShTSPAN7) to knockdown (KD) human TSPAN7 expression. Islet cells transduced with Ad‐ShTSPAN7, or the control virus (with scrambled RNA), were used to form pseudo‐islets. RNA sequencing showed a marked reduction in TSPAN7 expression (92.65%) in the TSPAN7‐KD pseudo‐islets (log2[Fold‐change] = −3.765, p\nadj = 6.42 E‐13) (Figure 5a), confirming successful genetic manipulation. In total, 1030 genes (1000 are protein‐coding genes) were down‐regulated and 395 genes (372 are protein‐coding genes) were up‐regulated in TSPAN7‐KD pseudo‐islets. Pathway analyses showed up‐regulation in apoptosis gene ontology pathway (p\nadj = 4.08 E−7); and down‐regulation of genes involved in the secretory pathway and transport regulation (p\nadj = 5.62 E−9 and 4.22 E−08 respectively). Interestingly, β‐cell expression of the exocytosis Ca2+ sensor SYT7 was reduced in TSPAN7‐KD pseudo‐islets (log2[Fold‐change] = −0.39, p\nadj = 3.89 E−3). GSIS of the pseudo‐islets were tested using dynamic perifusion experiments (Figure 5b). Whereas insulin secretion at basal or 16.7 mM glucose was comparable between the TSPAN7‐KD and control pseudo‐islets, 20 mM K+‐stimulated insulin secretion was significantly lower in TSPAN7‐KD pseudo‐islets.\n\nTSPAN7 KD affects exocytosis from human pancreatic β‐cells. (a) Gene expressions that changed significantly in human TSPAN7 KD pseudo‐islet. Volcano plot shows that genes involved in the pathways of secretion (yellow area) and regulation of transport (green area) were down‐regulated; markers of the apoptotic pathway (blue) were up‐regulated. TSPAN7 KD was demonstrated by the marked reduction in TSPAN7 expression (red). (b) Dynamic insulin secretion measured from perifused human pseudo‐islets of TSPAN7 KD (red) or control (Scramble, black) in response to different glucose concentrations and 20 mM K+ as indicated. Inset: area of the curve (AUC) of insulin secretion stimulated by 20 mM K+. N = 3 for control and TSPAN7 KD, *p < 0.05 versus insulin secretion at 1 mM glucose in the same group of pseudo‐islets; †\np < 0.05 versus control (Scramble), Student's t‐test. (c) Examples of exocytosis of TSPAN7 KD EndoC‐βH1 (red) or control EndoC‐βH1 (Scramble, black) triggered by depolarisation from −70 to 0 mV with progressively increasing duration (as indicated). (d) Summary of control (black) and exocytosis (normalised to cell size, fF/pF) in relationship to pulse durations. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. (e) The relationship between depolarisation‐triggered charge influx and pulse durations of control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. *p < 0.05 versus control, student's t‐test. (f) The relationship between charges and exocytosis of control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. Corresponding durations of the pulses are as labelled. N = 10 cells for control and 11 cells for TSPAN7 KD EndoC‐βH1 cells. (g) Insulin secretion measured from batch‐incubated control (black) and TSPAN7 KD (red) EndoC‐βH1 cells in response to 1 and 20 mM glucose. Data are the summary of three independent biological repeats in technical triplicate. *p < 0.05 and ***p < 0.001 versus control insulin secretion at 1 mM glucose and ††\np < 0.01 versus control insulin secretion at 20 mM glucose; two‐way ANOVA and Tuckey post hoc test. (h) Insulin content measured from control (black) and TSPAN7 KD (red) EndoC‐βH1 cells. Data are the summary of three independent biological repeats in technical triplicate. All data are presented as mean value ± SEM.\nThe insulin secretion experiments suggested that TSPAN7 may play a role in human β‐cell exocytosis. To test this, we performed capacitance measurements using EndoC‐βH1, a human β‐cell line recapturing functional and genetic features of primary human β‐cells,\n18\n transduced with Ad‐ShTSPAN7 or Ad‐Scramble virus. Similar to that observed in mouse Tspan7y/− β‐cells, exocytosis triggered by depolarisation was comparable between TSPAN7‐KD EndoC‐βH1 and control cells (Figure 5c,d). However, depolarisation triggered larger influx of charges (p < 0.05) in TSPAN7‐KD EndoC‐βH1 cells than that of the control (Ad‐Scramble) (Figure 5e). Importantly, the exocytotic Ca2+‐sensitivity of TSPAN7‐KD EndoC‐βH1 was reduced (same QCa2+ could only evoke exocytosis that was ~50% of the control, Figure 5f). This correlated with a significantly reduced GSIS in TSPAN7‐KD EndoC‐βH1 (p < 0.01, Figure 5g) without affecting insulin content (Figure 5h). We measured the changes in gene expression of VGCC and synaptotagmins in TSPAN7‐KD EndoC‐βH1. Concomitantly to TSPAN7 KD (p = 5.51 E−4), CACNA1A was up‐regulated and SYT5 and SYT7 were down‐regulated (−20%, Figure 6).\nChanges in VGCC‐ and synaptotagmin‐encoding genes in EndoC‐βH1 following TSPAN7 KD. The expression of genes encoding TSPAN7 (TSPAN7), VGCCs (CACNA1A, CACNA1C and CACNA1D) and synaptotagmins (SYT4, SYT5 and SYT7) measured from control (transduced with Ad‐Scramble, Scramble, black) and TSPAN7 KD (red) EndoC‐βH1 cells. The levels of expression are normalised to that in control cells (as 100%). Error bars of the control cells condition reflect the variation of the control across the biological repeats. Data are summary of measurements made in two to four independent biological replicates. ***p < 0.0001 versus control, Student's t‐test.", "Autoantigens implicated in type 1 diabetes have been shown to play a role in the regulation of insulin secretion.\n25\n Tspan7, a recently identified T1D autoantigen, can interact with both intracellular and membrane proteins, regulating diverse cellular processes, in a variety of cell types.\n5\n While Tspan7 expression in β‐cells is well established,\n8\n, \n13\n, \n22\n its role in insulin secretion is only partially understood. Here, we show that, in addition to its role in regulating VGCCs,\n13\n adequate expression of Tspan7 is required to maintain Ca2+ sensitivity of β‐cell exocytosis, which is associated with appropriate expression of synaptotagmin 7. This correlation is found in both mouse and human β‐cells but exerts a more significant role in human β‐cell function.\nThe role of Tspan7 in regulating cellular functions has been studied in many different cell types\n5\n, \n6\n, \n9\n, \n10\n, \n12\n, \n23\n and this has been recently extended into the study of pancreatic β‐cells,\n13\n where it was found to reduce L‐type VGCC activity. Therefore, reducing Tspan7 expression in β‐cells would be expected to lead to higher Ca2+ influx, improving glucose tolerance through enhanced GSIS (a Ca2+‐dependent process). Indeed, in Tspan7y/− β‐cells, depolarisation‐triggered Ca2+ current was augmented (Figure 4a; Figure S3a) and this may contribute to the trend of increased action potential peak, decreased inter‐spike plateau potential and lower firing frequency at high glucose (Figure 3b–d), similar to the effect of an L‐type VGCC agonist BAYK 8644.\n26\n However, this increment (~ + 3pA/pF at 0 mV) did not significantly change β‐cell action potential (Figure S1) and thus, may not be sufficient to impact on insulin secretion. This is confirmed by the normal GSIS observed in Tspan7y/− mice, both in vivo (Figure S1) and in vitro (Figure 4f). Similarly, GSIS was not significantly altered in human pseudo‐islets transduced with TSPAN7 shRNA. This differs from the previous report by Dickerson et al.\n13\n and may result from the different experimental paradigms used across the two studies (i.e., 2D single cell culture or 3D pseudo‐islets). Pseudo‐islets, formed by re‐aggregating dispersed islet cells, is an approximation of the primary intact islet, preserving a more physiological environment that includes intra‐islet paracrine signalling. Pseudo‐islet function is comparable with primary human islets\n17\n and allows efficient genetic manipulation using transduction, as highlighted in the present study (Figure 5a).\nThe stimulus‐secretion coupling of β‐cells involves exocytosis that depends on Ca2+ influx through the L‐type VGCC.\n14\n However, the observation that depolarisation‐triggered insulin secretion was unaffected in Tspan7y/− islets (Figure 4f; Figure S4) and slightly reduced in TSPAN7 KD human pseudo‐islets (Figure 5b) suggests reduced Tspan7/TSPAN7 may also affect exocytotic machinery. By correlating the transmembrane Ca2+ influx with high‐resolution exocytosis measurements, it is clear that exocytosis Ca2+ sensitivity is impaired when the level of tetraspanin‐7 in β‐cells is low. This effect could be in part attributed to reduced expression of Syt7, the major β‐cell exocytosis Ca2+ sensor.\n27\n Down‐regulation of the Syt7 following tetraspanin‐7 ablation/knockdown may be due to: (1) a coordinated expressions of these genes, or (2) Ca2+‐dependent changes in gene expression (secondary to augment VGCC activity, reviewed in\n28\n). Either possibility suggests that expression of tetraspanin‐7 may be required for appropriate assembly of exocytotic machinery in β‐cells. It is therefore likely that TSPAN7 plays a more significant role in insulin secretion in human β‐cells, in which exocytosis is less dependent on L‐type VGCC (more P/Q‐type VGCC dependent\n29\n). This notion is consistent with the observation that insulin secretion induced by 20 mM K+ and GSIS was reduced in TSPAN7 KD pseudo‐islets and EndoC‐βH1 cells respectively (Figure 5b,g). It is interesting to note that the impact of TSPAN7 KD on pseudo‐islets GSIS is less pronounced. This highlights the difference between the primary islets or reconstituted organoids and human cell lines. Whereas the primary human islets (and the pseudo‐islets) are comprised of different type of endocrine cells, EndoC‐βH1 cells are a single β‐cell population, lacking of the local paracrine cross‐talks that are important for appropriate GSIS. For example, it has been reported recently that α‐cells facilitate GSIS through glucagon/GLP‐1‐dependent signalling pathways.\n30\n Therefore, it is possible that defects in exocytosis (as the consequence of TSPAN7 KD) could exert a stronger effect in EndoC‐βH1 cells. It will be interesting to test whether a significantly reduced GSIS can be also observed in TSPAN7 KD pseudo‐islet composed by pure human primary β‐cells.\n\nTSPAN7 knockdown related transcription change is not restricted to exocytotic pathways. There is a small but significant up‐regulation of genes involved in apoptosis. It is possible this is due to augmented VGCC activity and secondary to elevated cytosolic Ca2+, which is pro‐apoptotic (reviewed in24). However, the degree of up‐regulation in apoptosis‐related genes may not be sufficient to reduce β‐cell mass in either acute or chronic TSPAN7 loss of function, as evidenced by the normal islet insulin content in shTSPAN7 transduced EndoC‐βH1 cells and in aged Tspan7y/− mice.\nIn summary, in addition to its inhibitory role on β‐cell VGCCs, we found that Tspan7 is required for appropriate expression of genes involved in cell survival and exocytosis. The non‐apparent metabolic phenotype and normal GSIS found in Tspan7y/− mice may be the result of a combined effect of enhanced VGCC activity and reduced sensitivity of exocytosis to Ca2+ in β‐cells.", "The authors do not have any relevant conflict of interest to disclose.", "\nFigure S1.\n\nClick here for additional data file.\n\nFigure S2.\n\nClick here for additional data file.\n\nFigure S3.\n\nClick here for additional data file.\n\nFigure S4.\n\nClick here for additional data file.\n\nFigure S5.\n\nClick here for additional data file.\n\nData S1.\n\nClick here for additional data file." ]
[ null, null, null, null, "methods", null, null, null, null, null, null, null, null, null, null, "results", null, null, null, null, "discussion", "COI-statement", "supplementary-material" ]
[ "β‐cell", "exocytosis", "insulin", "synaptotagmin", "tetraspanin‐7" ]
Colorectal Cancer Survival Trends in the United States From 1992 to 2018 Differ Among Persons From Five Racial and Ethnic Groups According to Stage at Diagnosis: A SEER-Based Study.
36264283
Survival following colorectal cancer (CRC) has improved in the US since 1975, but there is limited information on stage-specific survival trends among racial and ethnic subgroups.
INTRODUCTION
We performed a retrospective cohort study of individuals diagnosed with CRC using the 1992-2018 Surveillance, Epidemiology and End Results (SEER) database. We estimated and compared time trends in 1- and 5-year survival for CRC stage by race/ethnicity.
METHODS
Data from 399 220 individuals diagnosed with CRC were available. There were significant differences in stage-specific 1-year survival trends by race and ethnicity. Differences were most notable for distant stage CRC: survival probabilities increased most consistently for non-Hispanic American Indian/Alaska Native (AIAN) and Black (NHB) persons, but their trend lines were lower than those of Hispanic, and non-Hispanic Asian/Pacific Islander (API) and White (NHW) persons, whose initially greater gains appear to be slowing. Although the data do not support significant racial/ethnic differences in 5-year CRC survival trends by stage, AIAN and NHB persons have the lowest average survival probabilities for multiple CRC stages, and no racial/ethnic group has 5-year survival probabilities above 20% for distant-stage CRC.
RESULTS
Although there has been an overall improvement in adjusted CRC-specific survival probabilities since 1992, AIAN and NHB persons continue to experience worse prognosis than those of other races/ethnicities. This highlights the importance of reinvigorating efforts to understand the causes of mortality in CRC, including those which may differ according to an individual's race or ethnicity.
CONCLUSION
[ "United States", "Humans", "Ethnicity", "Retrospective Studies", "Racial Groups", "Hispanic or Latino", "Colorectal Neoplasms" ]
9597478
Introduction
Colorectal cancer (CRC) is the third most commonly diagnosed cancer and the second leading cause of cancer deaths in the United States.1,2 It has been estimated that 151 030 people will be newly diagnosed with CRC and 52 580 people with die from CRC in 2022.2,3 Although CRC mortality rates have decreased significantly since 1975,1-5 available reports suggest that these declines vary substantially by the stage of the disease, and that disparities in mortality rates for racial and ethnically minoritized groups persist.4-15 For example, according to a report by the American Cancer Society, the 2-year relative survival rate for distant-stage CRC increased from 21% in mid-1990’s to 37% for those diagnosed during 2009-2015, with commensurate improvements for regional- and localized-stage CRC.2 This same report asserts that CRC mortality rates are highest for persons who are non-Hispanic Black, followed by those who are American Indian or Alaska Native, and lowest for those who are Asian or Pacific Islander.2 Several studies have compared trends in CRC mortality by the stage of the disease and race but have done so under broad classifications as follows: White and Black;4,7-10 White, Black, and Hispanic;11,16,17 White, Black, Asian, Hispanic, and other;18 or White, Black, and other.19 In particular, limited information is available on CRC survival trends in persons who are American Indian or Alaska Native.20-22 A 2010 study by Edwards et al,5 compared CRC mortality trends in persons belonging to five racial and ethnic groups, but more than 10 years have passed since that report. To the best of our knowledge, no study has simultaneously compared recent trends in CRC-specific mortality by stage of diagnosis for individuals that are non-Hispanic White, non-Hispanic Black, Hispanic, non-Hispanic Asian or Pacific Islander, and non-Hispanic American Indian or Alaska Native. To address this gap, we undertook an analysis of population-based CRC cause-specific survival data to understand the trends in 1- and 5-year CRC cause-specific survival probabilities by stage of diagnosis within five racial and ethnic groups for individuals who received a CRC diagnosis between 1992 and 2018. Understanding differences in cause-specific survival, both over time and across CRC stages, is critical for further investigations into the social, structural, and political determinants that contribute to the disparities noted in CRC outcomes among individuals from distinct racial and ethnic groups. Through our efforts, we aim to provide new impetus to refocus efforts on improving CRC detection and treatment among racial and ethnically minoritized populations.
Methods
[SUBTITLE] Study Design and Data Source [SUBSECTION] This study was based on a retrospective cohort of CRC patients, ascertained on a population level, with data captured as part of the National Cancer Institute’s Surveillance, Epidemiology and End Results (SEER) program. The SEER program collects cancer incidence and survival data from population-based cancer registries representing approximately 35% of the U.S. population.23 Data from the 12 SEER registries for the period 1992 to 2018 were used in this analysis. The 12 SEER registries cover Alaska, Connecticut, Atlanta, rural Georgia, San Francisco-Oakland, San Jose-Monterey, Hawaii, Iowa, Los Angeles, New Mexico, Seattle-Puget Sound and Utah.24 This project, Study ID: 21-102, was reviewed by the University of New Mexico Health Sciences Institutional Review Board, and was granted exemption on 31 March 2021 according to Category 4: Secondary research on data or specimens (no consent required). This report conforms to RECORD guidelines for SEER-based studies.25 This study was supported by funding from the National Cancer Institute; the funders played no other role in this work. Researchers desiring additional details about the data and programs used to carry out this work may obtain programming code from the corresponding author. This study was based on a retrospective cohort of CRC patients, ascertained on a population level, with data captured as part of the National Cancer Institute’s Surveillance, Epidemiology and End Results (SEER) program. The SEER program collects cancer incidence and survival data from population-based cancer registries representing approximately 35% of the U.S. population.23 Data from the 12 SEER registries for the period 1992 to 2018 were used in this analysis. The 12 SEER registries cover Alaska, Connecticut, Atlanta, rural Georgia, San Francisco-Oakland, San Jose-Monterey, Hawaii, Iowa, Los Angeles, New Mexico, Seattle-Puget Sound and Utah.24 This project, Study ID: 21-102, was reviewed by the University of New Mexico Health Sciences Institutional Review Board, and was granted exemption on 31 March 2021 according to Category 4: Secondary research on data or specimens (no consent required). This report conforms to RECORD guidelines for SEER-based studies.25 This study was supported by funding from the National Cancer Institute; the funders played no other role in this work. Researchers desiring additional details about the data and programs used to carry out this work may obtain programming code from the corresponding author. [SUBTITLE] Study Population and Variables [SUBSECTION] We used SEER*Stat software (version 8.4.0)26 as the data source for this study. We used the “Incidence––SEER Research Plus Data, 12 Registries, Nov 2021 Sub (1992-2019)” database. We included a consecutive series of all individuals who received their first primary CRC diagnosis with malignant behavior from 1992 through 2018. We excluded “All Death Certificate Only and Autopsy Only” and “Alive with No Survival Time” via checkboxes in SEER*Stat. We also excluded CRC cases without a record for summary stage at diagnosis, as well as those with unknown age at diagnosis. We included all CRC cases that originated from the cecum, ascending colon, hepatic flexure of colon, transverse colon, splenic flexure of colon, descending colon, sigmoid colon, overlapping lesion of colon, colon not otherwise specified, rectosigmoid junction, and rectum. We classified CRC cases as localized, regional and distant stage using SEER’s “Combined Summary Stage (2004+)” classifications, supplemented with values from the “Historic Stage A (1973-2015)” variable when necessary. In-situ cancers and cases that were un-staged/unknown were excluded. These rules, as applied in SEER*Stat 8.0.4, defined the number of individuals included in this study. The SEER program works closely with providers of cancer care in their population-based catchment areas to collect patient- and cancer-specific information. Because of its population-based nature, it is typically necessary to rely on medical records as the source of the demographic and other data. SEER-defined race and ethnic categories were utilized and labeled as follows: Non-Hispanic White (NHW), Non-Hispanic Black (NHB), Hispanic, Non-Hispanic American Indian/Alaska Native (AIAN), and Non-Hispanic Asian or Pacific Islander (API). Additional variables were extracted from SEER, and estimates of CRC-specific survival probabilities were obtained for combinations of these variables in the primary analyses: sex as assigned at birth (male or female), age at diagnosis (categorized into decades), year of diagnosis, grade (grades I-IV, or unknown), and Rural-Urban Continuum Code in 2003 (RUCC, coded as: Metro Counties, Non-Metro Counties [Metro-Adjacent], Non-Metro Counties [not Metro-Adjacent], and Alaska or Hawaii, or unknown). The outcome of interest in this work was cause-specific survival for CRC, estimated at 1- and 5-years post diagnosis. We used SEER*Stat software (version 8.4.0)26 to calculate 1- and 5-year cause-specific survival probabilities of CRC using the “Incidence – SEER Research Plus Data, 12 Registries, Nov 2021 (1992-2019)” database while relying on the rules implemented in SEER*Stat for loss to follow-up. We obtained cause-specific survival probabilities and their standard errors within combinations of the characteristics of interest, ie, year of diagnosis, race and ethnicity, stage, sex, grade, and RUCC. No person-level data were used, nor were the SEER data linked to any other data sources. Using SEER*Stat 8.4.0, we formed tables of cause-specific survival probabilities and their standard errors according to all combinations of the factors of interest, with separate “Pages” defined for combinations of Year of diagnosis, age at diagnosis (in decades), and RUCC groups, and with rows defined by combinations of race/ethnicity, summary stage, grade, and sex. We used text processing approaches to form an analysis-ready data set which enumerated the numbers of individuals with a primary CRC diagnosis, and estimates of cause-specific survival probabilities and standard errors, within categories defined by all of the factors of interest in this study. We used SEER*Stat software (version 8.4.0)26 as the data source for this study. We used the “Incidence––SEER Research Plus Data, 12 Registries, Nov 2021 Sub (1992-2019)” database. We included a consecutive series of all individuals who received their first primary CRC diagnosis with malignant behavior from 1992 through 2018. We excluded “All Death Certificate Only and Autopsy Only” and “Alive with No Survival Time” via checkboxes in SEER*Stat. We also excluded CRC cases without a record for summary stage at diagnosis, as well as those with unknown age at diagnosis. We included all CRC cases that originated from the cecum, ascending colon, hepatic flexure of colon, transverse colon, splenic flexure of colon, descending colon, sigmoid colon, overlapping lesion of colon, colon not otherwise specified, rectosigmoid junction, and rectum. We classified CRC cases as localized, regional and distant stage using SEER’s “Combined Summary Stage (2004+)” classifications, supplemented with values from the “Historic Stage A (1973-2015)” variable when necessary. In-situ cancers and cases that were un-staged/unknown were excluded. These rules, as applied in SEER*Stat 8.0.4, defined the number of individuals included in this study. The SEER program works closely with providers of cancer care in their population-based catchment areas to collect patient- and cancer-specific information. Because of its population-based nature, it is typically necessary to rely on medical records as the source of the demographic and other data. SEER-defined race and ethnic categories were utilized and labeled as follows: Non-Hispanic White (NHW), Non-Hispanic Black (NHB), Hispanic, Non-Hispanic American Indian/Alaska Native (AIAN), and Non-Hispanic Asian or Pacific Islander (API). Additional variables were extracted from SEER, and estimates of CRC-specific survival probabilities were obtained for combinations of these variables in the primary analyses: sex as assigned at birth (male or female), age at diagnosis (categorized into decades), year of diagnosis, grade (grades I-IV, or unknown), and Rural-Urban Continuum Code in 2003 (RUCC, coded as: Metro Counties, Non-Metro Counties [Metro-Adjacent], Non-Metro Counties [not Metro-Adjacent], and Alaska or Hawaii, or unknown). The outcome of interest in this work was cause-specific survival for CRC, estimated at 1- and 5-years post diagnosis. We used SEER*Stat software (version 8.4.0)26 to calculate 1- and 5-year cause-specific survival probabilities of CRC using the “Incidence – SEER Research Plus Data, 12 Registries, Nov 2021 (1992-2019)” database while relying on the rules implemented in SEER*Stat for loss to follow-up. We obtained cause-specific survival probabilities and their standard errors within combinations of the characteristics of interest, ie, year of diagnosis, race and ethnicity, stage, sex, grade, and RUCC. No person-level data were used, nor were the SEER data linked to any other data sources. Using SEER*Stat 8.4.0, we formed tables of cause-specific survival probabilities and their standard errors according to all combinations of the factors of interest, with separate “Pages” defined for combinations of Year of diagnosis, age at diagnosis (in decades), and RUCC groups, and with rows defined by combinations of race/ethnicity, summary stage, grade, and sex. We used text processing approaches to form an analysis-ready data set which enumerated the numbers of individuals with a primary CRC diagnosis, and estimates of cause-specific survival probabilities and standard errors, within categories defined by all of the factors of interest in this study. [SUBTITLE] Statistical Analysis [SUBSECTION] We summarized the numbers and percentages of individuals with CRC diagnoses within the levels of the variables of interest. We used linear models to perform meta-regressions, weighting by the inverse of the squared standard errors, to estimate the degree to which the combinations of the variables of interest explained differences in the estimated cause-specific survival probabilities. When the SEER-estimated standard errors of survival probabilities were equal to zero, we approximated them with the square root of the variance of the binomial distribution, calculated after adding a value of .5 to both the numerator and denominator counts reported by SEER for the relevant group. We modelled the survival probabilities obtained at 1 year and 5 years post-diagnosis separately. The initial models included all possible interactions among race/ethnicity, stage, and year of diagnosis, while controlling for potential confounding by age at diagnosis (categorized into decades), sex, grade, and RUCC. We simplified these initial models subsequently modelling year-at-diagnosis trends with natural cubic spline basis functions, and selected the degree of smoothing that best explained the race by stage by year of diagnosis trends by minimizing Akaike’s Information Criterion (AIC) while adjusting for the other factors. We obtained smoothed trend estimates within groups defined by race and ethnicity, and by CRC stage at diagnosis, from a separate model for each of the two follow-up time periods. These final models were obtained by removing non-significant interactions in a hierarchical fashion. Analyses were performed using the tools available in SEER*Stat26 and SAS 9.4 (SAS Institute Inc, Cary, NC). Two-sided type I error was set at .05 for tests of significance. We summarized the numbers and percentages of individuals with CRC diagnoses within the levels of the variables of interest. We used linear models to perform meta-regressions, weighting by the inverse of the squared standard errors, to estimate the degree to which the combinations of the variables of interest explained differences in the estimated cause-specific survival probabilities. When the SEER-estimated standard errors of survival probabilities were equal to zero, we approximated them with the square root of the variance of the binomial distribution, calculated after adding a value of .5 to both the numerator and denominator counts reported by SEER for the relevant group. We modelled the survival probabilities obtained at 1 year and 5 years post-diagnosis separately. The initial models included all possible interactions among race/ethnicity, stage, and year of diagnosis, while controlling for potential confounding by age at diagnosis (categorized into decades), sex, grade, and RUCC. We simplified these initial models subsequently modelling year-at-diagnosis trends with natural cubic spline basis functions, and selected the degree of smoothing that best explained the race by stage by year of diagnosis trends by minimizing Akaike’s Information Criterion (AIC) while adjusting for the other factors. We obtained smoothed trend estimates within groups defined by race and ethnicity, and by CRC stage at diagnosis, from a separate model for each of the two follow-up time periods. These final models were obtained by removing non-significant interactions in a hierarchical fashion. Analyses were performed using the tools available in SEER*Stat26 and SAS 9.4 (SAS Institute Inc, Cary, NC). Two-sided type I error was set at .05 for tests of significance.
Results
This study was based on the 1975-2019 SEER Research Plus Data (November 2021 Submission), which contains information on 425 520 CRC diagnoses from 1992 through 2018. Excluding those individuals without a declared race or ethnicity, and with unknown stage at diagnosis, removed 1372 (.3%) and 24 928 (5.9%) individuals, respectively. This report is based on analysed data from 399 220 individual CRC diagnoses with known race/ethnicity and stage at diagnosis from 1992 through 2018, or 93.8% of all CRC diagnoses from this time period. Table 1 contains tabulations of the numbers of individuals according to levels of the variables of interest, overall and within race and ethnic groups. There was at least 1 year of follow-up for 318 443 (79.8%) persons with CRC eligible at baseline.Table 1.Characteristics of Patients With Colorectal Cancer From 1992 to 2018, by Racial and Ethnic Groups.AIANAPIHispanicNHBNHWTotalN%n%n%n%n%n%Total39311.045 86911.541 94710.534 1648.6273 30968.5399 220100.0SexMale197950.324 40053.222 21653.016 67448.8139 89751.2205 16651.4Female195249.721 46946.819 73147.017 49051.2133 41248.8194 05448.6Age<40 years2145.415723.424155.812173.659992.211 4172.940-49 years45911.740708.9455310.9340110.016 4666.028 9497.350-59 years91823.4924120.1930822.2781322.941 01815.068 29817.160-69 years104226.511 40424.910 65625.4916126.860 83922.393 10223.370-79 years86522.011 40724.9913021.8777222.778 10828.6107 28226.980+ years43311.0817517.8588514.0480014.070 87925.990 17222.6Summary stageLocalized152038.719 18241.816 57539.513 49139.5115 25442.2166 02241.6Regional149137.917 83338.916 15938.512 00135.1103 89638.0151 38037.9Distant92023.4885419.3921322.0867225.454 15919.881 81820.5Year of diagnosis1992-953488.9458310.036238.6407411.943 15115.855 77914.01996-9945411.6558312.2451410.8449113.244 92516.559 96715.02000-0348112.2671014.6527712.6495914.544 34316.261 77015.42004-0758714.9711615.5636815.2543915.940 99215.060 50215.22008-1165716.7781117.0722017.2560716.438 19213.959 48714.92012-1577819.8789717.2799719.0540615.835 53613.057 61414.42016-1862615.9616913.4694816.6418812.326 1709.544 10111.0GradeI3759.534977.6433510.331539.223 9368.835 2968.8II226057.527 75760.523 47756.019 37556.7155 56256.9228 43157.2III44811.4626913.7575313.7434412.746 96117.263 77516.0IV561.44451.05471.34151.243941.658571.5Unknown79220.1790117.2783518.7687720.142 45615.565 86116.5Rural-urban continuum codeMetro counties150038.240 55788.438 81492.533 18697.1228 17983.5342 23685.7Non-metro counties (metro-adjacent)3839.71120.213013.15401.623 5848.625 9206.5Non-metro counties (not metro-adjacent)39610.116543.616914.04101.220 4267.524 5776.2Alaska or Hawaii164441.800.050.000.000.016490.4Unknown80.235467.71360.3280.111200.448381.2 Characteristics of Patients With Colorectal Cancer From 1992 to 2018, by Racial and Ethnic Groups. [SUBTITLE] One- and Five-Year Survival: All Persons With CRC [SUBSECTION] Adjusted 1- and 5-year cause-specific survival probabilities following CRC diagnosis by stage and year of diagnosis are shown in Figure 1. For those diagnosed with local stage CRC, the estimate of linear trend in 1-year survival probabilities was negligible, at −.01% ([95% Confidence Interval (CI)] −.03%-.01%) per year, with an average 1-year survival probability of 96.2% (93.8%-98.6%). Those diagnosed with regional stage CRC displayed a small improvement over time in 1-year survival probabilities, .06% (.001%-.12%) per year. Their average 1-year survival probability was 93.0% (90.5%-95.5%). For those diagnosed with distant stage CRC, there was a significant improvement in 1-year survival probabilities over time (P < .001), but there was evidence of departure from a linear trend (P = .04). The slope of the trend line suggested an improvement in survival probability of 1.0% (.8%-1.2%) per year in 2005, with instantaneous slopes suggesting greater gains, by .05% (.01%-.09%), for each year prior to 2005 and slower gains by that same amount for each year following 2005. Their average 1-year survival probability was 52.8% (48.1%-57.5%).Figure 1.Trends in 1- and 5-year adjusted cause-specific survival probabilities for individuals diagnosed at different stages of colorectal cancer. Shaded bands reflect 95% prediction intervals for the year-specific survival probabilities. Trends in 1- and 5-year adjusted cause-specific survival probabilities for individuals diagnosed at different stages of colorectal cancer. Shaded bands reflect 95% prediction intervals for the year-specific survival probabilities. Those diagnosed with local stage CRC experienced improvements in 5-year cause-specific survival, with an average improvement of .16% (.10%-.21%) per year. Their average 5-year survival probability was 90.9% (87.9%-93.8%). Those diagnosed with regional stage CRC displayed significant improvement in 5-year survival probabilities over time (P < .001), but there was evidence of significant departure from a linear trend (P = .004), such that the slope of the trend line showed an improvement of .55% (.41%-.69%) per year in 2003, with instantaneous slopes suggested greater gains, by .08% (.04-.12%), for each year prior to 2003 and slower gains by that same amount for each year following 2003. Their average 5-year survival probability was 70.3% (66.5%-74.0%). For those diagnosed with distant stage CRC, there was a significant but small improvement over time in 5-year survival probabilities, .15% (.5%-.25%) per year. Their average 5-year survival probability was 8.1% (4.4%-11.8%). Adjusted 1- and 5-year cause-specific survival probabilities following CRC diagnosis by stage and year of diagnosis are shown in Figure 1. For those diagnosed with local stage CRC, the estimate of linear trend in 1-year survival probabilities was negligible, at −.01% ([95% Confidence Interval (CI)] −.03%-.01%) per year, with an average 1-year survival probability of 96.2% (93.8%-98.6%). Those diagnosed with regional stage CRC displayed a small improvement over time in 1-year survival probabilities, .06% (.001%-.12%) per year. Their average 1-year survival probability was 93.0% (90.5%-95.5%). For those diagnosed with distant stage CRC, there was a significant improvement in 1-year survival probabilities over time (P < .001), but there was evidence of departure from a linear trend (P = .04). The slope of the trend line suggested an improvement in survival probability of 1.0% (.8%-1.2%) per year in 2005, with instantaneous slopes suggesting greater gains, by .05% (.01%-.09%), for each year prior to 2005 and slower gains by that same amount for each year following 2005. Their average 1-year survival probability was 52.8% (48.1%-57.5%).Figure 1.Trends in 1- and 5-year adjusted cause-specific survival probabilities for individuals diagnosed at different stages of colorectal cancer. Shaded bands reflect 95% prediction intervals for the year-specific survival probabilities. Trends in 1- and 5-year adjusted cause-specific survival probabilities for individuals diagnosed at different stages of colorectal cancer. Shaded bands reflect 95% prediction intervals for the year-specific survival probabilities. Those diagnosed with local stage CRC experienced improvements in 5-year cause-specific survival, with an average improvement of .16% (.10%-.21%) per year. Their average 5-year survival probability was 90.9% (87.9%-93.8%). Those diagnosed with regional stage CRC displayed significant improvement in 5-year survival probabilities over time (P < .001), but there was evidence of significant departure from a linear trend (P = .004), such that the slope of the trend line showed an improvement of .55% (.41%-.69%) per year in 2003, with instantaneous slopes suggested greater gains, by .08% (.04-.12%), for each year prior to 2003 and slower gains by that same amount for each year following 2003. Their average 5-year survival probability was 70.3% (66.5%-74.0%). For those diagnosed with distant stage CRC, there was a significant but small improvement over time in 5-year survival probabilities, .15% (.5%-.25%) per year. Their average 5-year survival probability was 8.1% (4.4%-11.8%). [SUBTITLE] One-Year Survival by Race and Ethnicity [SUBSECTION] Estimates of adjusted 1-year cause-specific survival probabilities following diagnosis are shown for all race/ethnicity by stage combinations in Tables 2 and 3. Trends in 1-year survival for those of different races/ethnicities who were diagnosed at different stages of CRC differed significantly (P < .001, see Figure 2). For those diagnosed with localized stage CRC, 1-year survival probabilities were consistently high for individuals of all race and ethnic groups. AIAN persons experienced lower 1-year survival probabilities than those from other race and ethnic groups over the study period. Although due to lack of precision in the estimates, the difference was only significantly different when compared to API persons; average 1-year survival probabilities were 1.5 (.5-2.5) percentage points lower for AIAN than for API persons.Table 2.Unadjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN1992100 (84.6-100)91.6 (75.4-100)27.0 (5.6-48.4)72.9 (62.6-83.2)AIAN199696.9 (81.8-100)95.7 (77.9-100)41.8 (23.5-60.1)78.1 (68.2-88.0)AIAN200098.7 (83.6-100)83.2 (65.9-100)43.0 (20.0-66.0)75.0 (64.1-85.8)AIAN200496.7 (85.6-100)96.4 (84.8-100)51.8 (35.8-67.7)81.6 (74.1-89.2)AIAN2008100 (86.4-100)95.5 (83.0-100)53.5 (35.5-71.5)83.0 (74.4-91.6)AIAN2012100 (88.9-100)91.6 (79.1-100)52.7 (38.0-67.4)81.4 (74.0-88.8)AIAN201696.0 (85.5-100)95.9 (85.6-100)46.5 (33.4-59.6)79.5 (72.9-86.0)AIAN2018100 (90.7-100)99.3 (88.9-100)83.5 (67.2-99.8)94.3 (87.1-100)API199299.5 (96.2-100)94.6 (90.8-98.3)46.5 (38.4-54.6)80.2 (77.0-83.3)API199699.4 (97.1-100)95.8 (92.6-98.9)56.5 (48.7-64.3)83.9 (81.0-86.8)API200099.2 (97.0-100)97.9 (95.8-99.9)52.8 (45.0-60.6)83.3 (80.5-86.1)API200499.6 (97.9-100)97.3 (95.1-99.5)68.6 (62.7-74.4)88.5 (86.3-90.6)API200899.7 (98.3-100)98.4 (96.5-100)59.5 (53.9-65.0)85.9 (83.9-87.9)API201299.4 (97.9-100)96.9 (94.7-99.2)55.8 (49.4-62.3)84.1 (81.7-86.4)API201699.1 (97.4-100)98.4 (96.7-100)57.6 (51.8-63.3)85.0 (82.9-87.1)API201899.6 (98.5-100)97.8 (96.0-99.6)69.1 (62.7-75.5)88.8 (86.6-91.1)Hispanic199297.0 (93.0-100)96.7 (93.3-100)55.6 (46.2-64.9)83.1 (79.5-86.7)Hispanic199698.8 (95.8-100)96.1 (92.7-99.5)43.4 (35.7-51.1)79.4 (76.4-82.4)Hispanic200099.2 (96.6-100)96.3 (93.3-99.4)47.0 (39.8-54.2)80.8 (78.1-83.6)Hispanic200498.9 (96.5-100)95.8 (92.9-98.7)57.8 (50.8-64.8)84.2 (81.5-86.8)Hispanic200899.1 (97.3-100)96.2 (93.5-98.8)60.7 (54.4-67.1)85.3 (83.0-87.7)Hispanic201299.2 (97.4-100)95.8 (93.3-98.2)71.6 (66.6-76.7)88.9 (86.9-90.8)Hispanic201698.9 (97.1-100)98.2 (96.6-99.7)69.0 (64.0-74.1)88.7 (86.9-90.6)Hispanic201899.6 (98.6-100)97.8 (96.1-99.5)58.8 (52.5-65.0)85.4 (83.2-87.6)NHB199298.0 (94.7-100)93.7 (89.6-97.8)38.5 (30.0-47.0)76.7 (73.4-80.1)NHB199698.2 (94.7-100)96.3 (93-99.6)30.1 (23.1-37.2)74.9 (72.0-77.7)NHB200097.3 (93.8-100)93.2 (89.4-97.0)34.5 (27.3-41.7)75.0 (72.0-77.9)NHB200498.4 (95.6-100)97.0 (94.3-99.7)37.0 (30.7-43.3)77.5 (75.0-79.9)NHB200898.6 (96.2-100)93.3 (89.4-97.2)51.4 (44.7-58.1)81.1 (78.4-83.8)NHB201299.5 (97.2-100)93.6 (90.0-97.1)68.5 (62.1-74.8)87.2 (84.7-89.7)NHB201699.5 (97.2-100)96.0 (93.0-99.0)53.1 (47.2-58.9)82.9 (80.5-85.2)NHB201899.3 (97.5-100)97.0 (94.2-99.8)57.1 (49.6-64.6)84.5 (81.7-87.2)NHW199298.6 (97.9-99.2)94.6 (93.5-95.7)38.0 (35.0-40.9)77.0 (76.0-78.1)NHW199698.9 (98.3-99.5)96.1 (95.1-97.0)36.7 (33.8-39.5)77.2 (76.2-78.2)NHW200099.0 (98.5-99.6)94.3 (93.2-95.4)40.9 (37.9-43.9)78.1 (77.0-79.1)NHW200498.9 (98.3-99.5)96.1 (95.1-97.1)52.6 (49.7-55.4)82.5 (81.5-83.6)NHW200899.2 (98.6-99.7)97.0 (96.1-98.0)58.6 (55.6-61.7)85.0 (83.9-86.0)NHW201298.9 (98.2-99.6)97.2 (96.2-98.2)55.2 (52.3-58.0)83.8 (82.7-84.8)NHW201699.1 (98.5-99.8)96.9 (95.9-97.9)60.7 (57.9-63.5)85.6 (84.6-86.6)NHW201898.7 (98.0-99.4)97.5 (96.7-98.4)59.0 (55.7-62.3)85.1 (83.9-86.3)Table 3.Adjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199297.5 (83.7-100)89.8 (75.3-100)25.4 (6.2-44.6)70.9 (61.6-80.2)AIAN199695.6 (82.0-100)93.2 (77.3-100)41.9 (25.5-58.3)76.9 (68.0-85.8)AIAN200097.0 (83.5-100)82.5 (67.0-97.9)42.0 (21.4-62.6)73.8 (64.1-83.5)AIAN200495.3 (85.3-100)94.2 (83.8-100)50.4 (36.1-64.7)80.0 (73.2-86.8)AIAN200897.5 (85.3-100)92.4 (81.2-100)51.6 (35.5-67.7)80.5 (72.8-88.2)AIAN201297.5 (87.5-100)89.9 (78.7-100)50.2 (37.0-63.3)79.2 (72.5-85.9)AIAN201693.4 (84.0-100)94.1 (84.8-100)45.0 (33.3-56.8)77.5 (71.6-83.4)AIAN201896.7 (87.8-100)96.9 (87.6-100)64.2 (51.1-77.5)85.9 (79.7-91.7)API199297.5 (94.5-100)92.3 (88.9-95.7)45.8 (38.6-53.1)78.6 (75.7-81.4)API199697.4 (95.2-99.6)93.4 (90.5-96.3)55.6 (48.5-62.6)82.1 (79.4-84.8)API200097.4 (95.3-99.5)95.9 (93.9-97.9)53.1 (46.1-60.1)82.1 (79.6-84.7)API200497.8 (96.2-99.4)94.6 (92.5-96.7)67.1 (61.8-72.4)86.5 (84.5-88.5)API200897.5 (96.0-98.9)95.6 (93.7-97.4)59.4 (54.4-64.4)84.2 (82.2-86.1)API201296.8 (95.3-98.3)94.6 (92.5-96.7)56.1 (50.3-61.9)82.5 (80.3-84.7)API201696.4 (94.8-98.0)95.0 (93.4-96.6)57.8 (52.6-63.0)83.1 (81.1-85.0)API201897.9 (96.5-99.0)96.1 (94.5-97.9)63.8 (58.3-69.3)86.0 (83.9-88.0)Hispanic199294.7 (91.1-98.4)94.4 (91.3-97.5)54.3 (45.9-62.7)81.2 (77.9-84.4)Hispanic199696.8 (93.9-99.6)94.2 (91.0-97.3)42.9 (36.0-49.9)77.9 (75.2-80.7)Hispanic200097.0 (94.6-99.4)93.4 (90.6-96.2)46.6 (40.1-53.0)79.0 (76.4-81.5)Hispanic200497.2 (95.0-99.4)93.3 (90.6-96.0)57.1 (50.8-63.4)82.5 (80.1-85.0)Hispanic200896.2 (94.5-97.9)92.8 (90.4-95.3)59.8 (54.1-65.5)83.0 (80.8-85.2)Hispanic201296.0 (94.3-97.8)92.9 (90.6-95.2)69.3 (64.8-73.9)86.1 (84.2-88.0)Hispanic201694.8 (93.0-96.5)94.1 (92.5-95.6)67.3 (62.8-71.9)85.4 (83.6-87.2)Hispanic201896.8 (95.3-98.3)95.3 (93.7-96.9)63.4 (58.3-68.6)85.2 (83.2-87.2)NHB199295.7 (92.6-98.7)91.1 (87.3-94.8)37.8 (30.2-45.4)74.8 (71.8-77.9)NHB199696.6 (93.4-99.9)93.8 (90.7-96.8)30.8 (24.4-37.1)73.7 (71.1-76.3)NHB200095.6 (92.5-98.8)90.5 (87.1-94.0)35.7 (29.2-42.2)74.0 (71.2-76.7)NHB200495.9 (93.3-98.5)94.0 (91.4-96.5)38.9 (33.2-44.6)76.3 (73.9-78.6)NHB200896.1 (93.9-98.3)90.3 (86.8-93.9)51.2 (45.1-57.2)79.2 (76.7-81.7)NHB201296.6 (94.4-98.7)90.6 (87.4-93.8)66.7 (60.9-72.4)84.6 (82.2-87.0)NHB201696.0 (93.9-98.2)92.3 (89.6-95.1)53.0 (47.7-58.3)80.5 (78.3-82.6)NHB201897.5 (95.6-99.1)94.3 (91.7-97.1)55.2 (49.2-61.3)82.4 (80.0-84.8)NHW199296.8 (95.9-97.7)92.8 (91.6-94.0)39.2 (36.5-42.0)76.3 (75.1-77.4)NHW199696.7 (95.9-97.6)93.1 (92.1-94.2)38.4 (35.8-41.1)76.1 (75.0-77.2)NHW200096.3 (95.5-97.2)92.4 (91.2-93.6)42.4 (39.6-45.1)77.0 (75.9-78.2)NHW200496.5 (95.6-97.3)93.5 (92.4-94.7)53.5 (50.8-56.1)81.2 (80.0-82.3)NHW200896.4 (95.6-97.3)93.8 (92.7-94.9)58.8 (56.0-61.6)83.0 (81.8-84.2)NHW201295.9 (95.0-96.8)93.9 (92.8-95.0)55.7 (53.0-58.3)81.8 (80.7-83.0)NHW201695.2 (94.3-96.0)93.5 (92.4-94.6)60.4 (57.8-63.0)83.0 (81.9-84.2)NHW201896.7 (95.8-97.6)95.0 (93.95-96.1)60.4 (57.6-63.3)84.0 (82.9-85.3)Figure 2.Trends in 1-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2018. Unadjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis. Adjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis. Trends in 1-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2018. For those diagnosed with regional stage CRC, trends in adjusted 1-year cause-specific survival probabilities similarly reflect little change over time, with the possible exception of an imprecisely-estimated positive trend over time among AIAN persons. Although there was little evidence of significant trends over time, there was a clear gradient in the average survival probabilities among those from different race and ethnic groups. AIAN persons had the lowest average survival probability (90.2%, 88.2%-92.2), followed by NHB, NHW, Hispanic, and API persons (93.2%, 92.6%-93.8%; 93.3%, 93.1%-93.5%; 94.2%, 93.8%-94.6%; and 94.8%, 94.4%-95.2%, respectively). All of these estimates were significantly different (all P < .01), except for the comparison between NHB and NHW persons (P = .83). Those diagnosed with distant stage CRC experienced the biggest improvements in 1-year cause-specific survival probabilities over time. An increasing trend in 1-year survival probabilities is apparent for persons of all race and ethnic groups. For AIAN and NHB persons, the two groups with lowest initial 1-year survival probabilities, improvements continued at a consistent rate over time. For API, Hispanic, and NHW persons, there appears to be a slowing of the rate of improvement in the second half of the study period. The adjusted time-averaged survival probabilities were low: 46.6% (42.3%-50.9%) for NHB, 48.3% (37.9%-58.7%) for AIAN, 50.5% (48.5%-52.5%) for NHW, 58.1% (54.2%-62.0%) for Hispanic, and 58.3% (54.2%-62.4%) for API persons. All pairwise comparisons among these groups were statistically significant (all P < .01), except for differences between the two groups with the lowest (AIAN vs NHB, P = .87), and between the two groups with the highest (API vs Hispanic, P = .69), average adjusted 1-year cause-specific survival probabilities. Estimates of adjusted 1-year cause-specific survival probabilities following diagnosis are shown for all race/ethnicity by stage combinations in Tables 2 and 3. Trends in 1-year survival for those of different races/ethnicities who were diagnosed at different stages of CRC differed significantly (P < .001, see Figure 2). For those diagnosed with localized stage CRC, 1-year survival probabilities were consistently high for individuals of all race and ethnic groups. AIAN persons experienced lower 1-year survival probabilities than those from other race and ethnic groups over the study period. Although due to lack of precision in the estimates, the difference was only significantly different when compared to API persons; average 1-year survival probabilities were 1.5 (.5-2.5) percentage points lower for AIAN than for API persons.Table 2.Unadjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN1992100 (84.6-100)91.6 (75.4-100)27.0 (5.6-48.4)72.9 (62.6-83.2)AIAN199696.9 (81.8-100)95.7 (77.9-100)41.8 (23.5-60.1)78.1 (68.2-88.0)AIAN200098.7 (83.6-100)83.2 (65.9-100)43.0 (20.0-66.0)75.0 (64.1-85.8)AIAN200496.7 (85.6-100)96.4 (84.8-100)51.8 (35.8-67.7)81.6 (74.1-89.2)AIAN2008100 (86.4-100)95.5 (83.0-100)53.5 (35.5-71.5)83.0 (74.4-91.6)AIAN2012100 (88.9-100)91.6 (79.1-100)52.7 (38.0-67.4)81.4 (74.0-88.8)AIAN201696.0 (85.5-100)95.9 (85.6-100)46.5 (33.4-59.6)79.5 (72.9-86.0)AIAN2018100 (90.7-100)99.3 (88.9-100)83.5 (67.2-99.8)94.3 (87.1-100)API199299.5 (96.2-100)94.6 (90.8-98.3)46.5 (38.4-54.6)80.2 (77.0-83.3)API199699.4 (97.1-100)95.8 (92.6-98.9)56.5 (48.7-64.3)83.9 (81.0-86.8)API200099.2 (97.0-100)97.9 (95.8-99.9)52.8 (45.0-60.6)83.3 (80.5-86.1)API200499.6 (97.9-100)97.3 (95.1-99.5)68.6 (62.7-74.4)88.5 (86.3-90.6)API200899.7 (98.3-100)98.4 (96.5-100)59.5 (53.9-65.0)85.9 (83.9-87.9)API201299.4 (97.9-100)96.9 (94.7-99.2)55.8 (49.4-62.3)84.1 (81.7-86.4)API201699.1 (97.4-100)98.4 (96.7-100)57.6 (51.8-63.3)85.0 (82.9-87.1)API201899.6 (98.5-100)97.8 (96.0-99.6)69.1 (62.7-75.5)88.8 (86.6-91.1)Hispanic199297.0 (93.0-100)96.7 (93.3-100)55.6 (46.2-64.9)83.1 (79.5-86.7)Hispanic199698.8 (95.8-100)96.1 (92.7-99.5)43.4 (35.7-51.1)79.4 (76.4-82.4)Hispanic200099.2 (96.6-100)96.3 (93.3-99.4)47.0 (39.8-54.2)80.8 (78.1-83.6)Hispanic200498.9 (96.5-100)95.8 (92.9-98.7)57.8 (50.8-64.8)84.2 (81.5-86.8)Hispanic200899.1 (97.3-100)96.2 (93.5-98.8)60.7 (54.4-67.1)85.3 (83.0-87.7)Hispanic201299.2 (97.4-100)95.8 (93.3-98.2)71.6 (66.6-76.7)88.9 (86.9-90.8)Hispanic201698.9 (97.1-100)98.2 (96.6-99.7)69.0 (64.0-74.1)88.7 (86.9-90.6)Hispanic201899.6 (98.6-100)97.8 (96.1-99.5)58.8 (52.5-65.0)85.4 (83.2-87.6)NHB199298.0 (94.7-100)93.7 (89.6-97.8)38.5 (30.0-47.0)76.7 (73.4-80.1)NHB199698.2 (94.7-100)96.3 (93-99.6)30.1 (23.1-37.2)74.9 (72.0-77.7)NHB200097.3 (93.8-100)93.2 (89.4-97.0)34.5 (27.3-41.7)75.0 (72.0-77.9)NHB200498.4 (95.6-100)97.0 (94.3-99.7)37.0 (30.7-43.3)77.5 (75.0-79.9)NHB200898.6 (96.2-100)93.3 (89.4-97.2)51.4 (44.7-58.1)81.1 (78.4-83.8)NHB201299.5 (97.2-100)93.6 (90.0-97.1)68.5 (62.1-74.8)87.2 (84.7-89.7)NHB201699.5 (97.2-100)96.0 (93.0-99.0)53.1 (47.2-58.9)82.9 (80.5-85.2)NHB201899.3 (97.5-100)97.0 (94.2-99.8)57.1 (49.6-64.6)84.5 (81.7-87.2)NHW199298.6 (97.9-99.2)94.6 (93.5-95.7)38.0 (35.0-40.9)77.0 (76.0-78.1)NHW199698.9 (98.3-99.5)96.1 (95.1-97.0)36.7 (33.8-39.5)77.2 (76.2-78.2)NHW200099.0 (98.5-99.6)94.3 (93.2-95.4)40.9 (37.9-43.9)78.1 (77.0-79.1)NHW200498.9 (98.3-99.5)96.1 (95.1-97.1)52.6 (49.7-55.4)82.5 (81.5-83.6)NHW200899.2 (98.6-99.7)97.0 (96.1-98.0)58.6 (55.6-61.7)85.0 (83.9-86.0)NHW201298.9 (98.2-99.6)97.2 (96.2-98.2)55.2 (52.3-58.0)83.8 (82.7-84.8)NHW201699.1 (98.5-99.8)96.9 (95.9-97.9)60.7 (57.9-63.5)85.6 (84.6-86.6)NHW201898.7 (98.0-99.4)97.5 (96.7-98.4)59.0 (55.7-62.3)85.1 (83.9-86.3)Table 3.Adjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199297.5 (83.7-100)89.8 (75.3-100)25.4 (6.2-44.6)70.9 (61.6-80.2)AIAN199695.6 (82.0-100)93.2 (77.3-100)41.9 (25.5-58.3)76.9 (68.0-85.8)AIAN200097.0 (83.5-100)82.5 (67.0-97.9)42.0 (21.4-62.6)73.8 (64.1-83.5)AIAN200495.3 (85.3-100)94.2 (83.8-100)50.4 (36.1-64.7)80.0 (73.2-86.8)AIAN200897.5 (85.3-100)92.4 (81.2-100)51.6 (35.5-67.7)80.5 (72.8-88.2)AIAN201297.5 (87.5-100)89.9 (78.7-100)50.2 (37.0-63.3)79.2 (72.5-85.9)AIAN201693.4 (84.0-100)94.1 (84.8-100)45.0 (33.3-56.8)77.5 (71.6-83.4)AIAN201896.7 (87.8-100)96.9 (87.6-100)64.2 (51.1-77.5)85.9 (79.7-91.7)API199297.5 (94.5-100)92.3 (88.9-95.7)45.8 (38.6-53.1)78.6 (75.7-81.4)API199697.4 (95.2-99.6)93.4 (90.5-96.3)55.6 (48.5-62.6)82.1 (79.4-84.8)API200097.4 (95.3-99.5)95.9 (93.9-97.9)53.1 (46.1-60.1)82.1 (79.6-84.7)API200497.8 (96.2-99.4)94.6 (92.5-96.7)67.1 (61.8-72.4)86.5 (84.5-88.5)API200897.5 (96.0-98.9)95.6 (93.7-97.4)59.4 (54.4-64.4)84.2 (82.2-86.1)API201296.8 (95.3-98.3)94.6 (92.5-96.7)56.1 (50.3-61.9)82.5 (80.3-84.7)API201696.4 (94.8-98.0)95.0 (93.4-96.6)57.8 (52.6-63.0)83.1 (81.1-85.0)API201897.9 (96.5-99.0)96.1 (94.5-97.9)63.8 (58.3-69.3)86.0 (83.9-88.0)Hispanic199294.7 (91.1-98.4)94.4 (91.3-97.5)54.3 (45.9-62.7)81.2 (77.9-84.4)Hispanic199696.8 (93.9-99.6)94.2 (91.0-97.3)42.9 (36.0-49.9)77.9 (75.2-80.7)Hispanic200097.0 (94.6-99.4)93.4 (90.6-96.2)46.6 (40.1-53.0)79.0 (76.4-81.5)Hispanic200497.2 (95.0-99.4)93.3 (90.6-96.0)57.1 (50.8-63.4)82.5 (80.1-85.0)Hispanic200896.2 (94.5-97.9)92.8 (90.4-95.3)59.8 (54.1-65.5)83.0 (80.8-85.2)Hispanic201296.0 (94.3-97.8)92.9 (90.6-95.2)69.3 (64.8-73.9)86.1 (84.2-88.0)Hispanic201694.8 (93.0-96.5)94.1 (92.5-95.6)67.3 (62.8-71.9)85.4 (83.6-87.2)Hispanic201896.8 (95.3-98.3)95.3 (93.7-96.9)63.4 (58.3-68.6)85.2 (83.2-87.2)NHB199295.7 (92.6-98.7)91.1 (87.3-94.8)37.8 (30.2-45.4)74.8 (71.8-77.9)NHB199696.6 (93.4-99.9)93.8 (90.7-96.8)30.8 (24.4-37.1)73.7 (71.1-76.3)NHB200095.6 (92.5-98.8)90.5 (87.1-94.0)35.7 (29.2-42.2)74.0 (71.2-76.7)NHB200495.9 (93.3-98.5)94.0 (91.4-96.5)38.9 (33.2-44.6)76.3 (73.9-78.6)NHB200896.1 (93.9-98.3)90.3 (86.8-93.9)51.2 (45.1-57.2)79.2 (76.7-81.7)NHB201296.6 (94.4-98.7)90.6 (87.4-93.8)66.7 (60.9-72.4)84.6 (82.2-87.0)NHB201696.0 (93.9-98.2)92.3 (89.6-95.1)53.0 (47.7-58.3)80.5 (78.3-82.6)NHB201897.5 (95.6-99.1)94.3 (91.7-97.1)55.2 (49.2-61.3)82.4 (80.0-84.8)NHW199296.8 (95.9-97.7)92.8 (91.6-94.0)39.2 (36.5-42.0)76.3 (75.1-77.4)NHW199696.7 (95.9-97.6)93.1 (92.1-94.2)38.4 (35.8-41.1)76.1 (75.0-77.2)NHW200096.3 (95.5-97.2)92.4 (91.2-93.6)42.4 (39.6-45.1)77.0 (75.9-78.2)NHW200496.5 (95.6-97.3)93.5 (92.4-94.7)53.5 (50.8-56.1)81.2 (80.0-82.3)NHW200896.4 (95.6-97.3)93.8 (92.7-94.9)58.8 (56.0-61.6)83.0 (81.8-84.2)NHW201295.9 (95.0-96.8)93.9 (92.8-95.0)55.7 (53.0-58.3)81.8 (80.7-83.0)NHW201695.2 (94.3-96.0)93.5 (92.4-94.6)60.4 (57.8-63.0)83.0 (81.9-84.2)NHW201896.7 (95.8-97.6)95.0 (93.95-96.1)60.4 (57.6-63.3)84.0 (82.9-85.3)Figure 2.Trends in 1-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2018. Unadjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis. Adjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis. Trends in 1-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2018. For those diagnosed with regional stage CRC, trends in adjusted 1-year cause-specific survival probabilities similarly reflect little change over time, with the possible exception of an imprecisely-estimated positive trend over time among AIAN persons. Although there was little evidence of significant trends over time, there was a clear gradient in the average survival probabilities among those from different race and ethnic groups. AIAN persons had the lowest average survival probability (90.2%, 88.2%-92.2), followed by NHB, NHW, Hispanic, and API persons (93.2%, 92.6%-93.8%; 93.3%, 93.1%-93.5%; 94.2%, 93.8%-94.6%; and 94.8%, 94.4%-95.2%, respectively). All of these estimates were significantly different (all P < .01), except for the comparison between NHB and NHW persons (P = .83). Those diagnosed with distant stage CRC experienced the biggest improvements in 1-year cause-specific survival probabilities over time. An increasing trend in 1-year survival probabilities is apparent for persons of all race and ethnic groups. For AIAN and NHB persons, the two groups with lowest initial 1-year survival probabilities, improvements continued at a consistent rate over time. For API, Hispanic, and NHW persons, there appears to be a slowing of the rate of improvement in the second half of the study period. The adjusted time-averaged survival probabilities were low: 46.6% (42.3%-50.9%) for NHB, 48.3% (37.9%-58.7%) for AIAN, 50.5% (48.5%-52.5%) for NHW, 58.1% (54.2%-62.0%) for Hispanic, and 58.3% (54.2%-62.4%) for API persons. All pairwise comparisons among these groups were statistically significant (all P < .01), except for differences between the two groups with the lowest (AIAN vs NHB, P = .87), and between the two groups with the highest (API vs Hispanic, P = .69), average adjusted 1-year cause-specific survival probabilities. [SUBTITLE] Five-Year Survival by Race and Ethnicity [SUBSECTION] Estimates of adjusted 5-year cause-specific survival probabilities are shown for all race/ethnicity by stage combinations in Tables 4 and 5. Neither the race/ethnicity by stage by year of diagnosis three-way interaction (P = .90) nor the two-way interaction between the year of diagnosis and race/ethnicity (P = .59) were statistically significant; there is not sufficient evidence to conclude that there are race- or ethnic-specific differences in 5-year survival trends either within or across stages of CRC at diagnosis. There were statistically significant interactions between race/ethnicity and stage (P < .001), and between stage and the year of diagnosis trends (P < .001) with respect to differences in 5-year survival (see Figure 3).Table 4.Unadjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199283.2 (67.0-99.3)58.1 (40.1-76.0)8.5 (0-29.1)49.9 (39.3-60.5)AIAN199587.5 (70.6-100)64.7 (47.7-81.7)13.4 (0-33.0)55.2 (44.9-65.5)AIAN199885.4 (69.9-100)73.1 (57.2-89.0)1.6 (0-18.5)53.3 (44.0-62.6)AIAN200291.8 (79.0-100)70.4 (56.5-84.3)10.8 (0-33.9)57.6 (47.7-67.6)AIAN200597.1 (85.6-100)65.6 (52.5-78.6)23.0 (6.8-39.2)61.9 (54.0-69.8)AIAN200897.5 (84.4-100)83.4 (70.7-96.1)5.9 (0-22.7)62.3 (54.0-70.6)AIAN201189.3 (77.0-100)67.5 (55.3-79.6)9.5 (0.0-22.7)55.4 (48.2-62.7)AIAN201496.5 (86.4-100)71.4 (60.1-82.6)15.1 (2.0-28.2)61.0 (54.3-67.6)API199297.1 (93.4-100)63.9 (57.9-70.0)7.1 (.9-13.2)56.0 (52.9-59.1)API199594.8 (91.1-98.4)67.5 (62.0-73.0)4.9 (0-10.7)55.7 (52.8-58.6)API199896.1 (93.0-99.2)76.8 (71.8-81.8)7.9 (2.5-13.4)60.3 (57.6-62.9)API200296.7 (94.3-99.1)79.8 (75.4-84.1)7.1 (2.5-11.8)61.2 (58.9-63.5)API200596.5 (94.1-98.9)80.0 (75.9-84.1)9.8 (4.7-14.8)62.1 (59.8-64.4)API200897.7 (95.7-99.7)80.6 (76.3-85)9.0 (4.1-13.9)62.4 (60.1-64.7)API201196.7 (94.4-99.0)76.7 (72.5-81.0)8.4 (3.6-13.1)60.6 (58.3-62.9)API201497.2 (95.0-99.4)81.5 (77.6-85.5)8.0 (3.8-12.1)62.2 (60.2-64.3)Hispanic199292.6 (87.7-97.5)58.1 (51.7-64.4)6.7 (0-13.4)52.4 (48.9-55.9)Hispanic199590.7 (85.5-95.8)61.0 (54.6-67.4)5.4 (0-11.4)52.4 (49.0-55.8)Hispanic199894.3 (90.6-98.0)72.7 (67.0-78.3)5.5 (0-11.5)57.5 (54.4-60.5)Hispanic200293.8 (90.1-97.4)75.5 (70.3-80.8)6.0 (.8-11.2)58.4 (55.7-61.2)Hispanic200594.6 (91.6-97.6)74.1 (69.3-78.9)9.3 (4.1-14.5)59.3 (56.8-61.9)Hispanic200896.0 (93.5-98.6)74.0 (69.1-78.9)6.3 (1.6-10.9)58.8 (56.4-61.2)Hispanic201197.6 (95.5-99.6)79 (74.6-83.3)8.9 (4.6-13.1)61.8 (59.6-63.9)Hispanic201496.9 (94.8-99)75.2 (70.9-79.4)9.4 (5.1-13.7)60.5 (58.4-62.6)NHB199290.6 (85.7-95.4)60.9 (54.1-67.7)2.2 (0-6.8)51.2 (48.1-54.4)NHB199591.3 (86.7-95.9)63.8 (57.0-70.6)4.3 (0-9.6)53.1 (49.8-56.4)NHB199896.0 (92.5-99.4)66.4 (60.0-72.9)4.8 (0-9.8)55.7 (52.8-58.7)NHB200289.9 (85.2-94.6)71.6 (65.9-77.3)4.5 (0-9.0)55.3 (52.4-58.2)NHB200591.8 (87.9-95.7)73.5 (67.7-79.3)4.3 (.1-8.4)56.5 (53.8-59.2)NHB200893.8 (90.4-97.1)69.3 (63.3-75.3)5.7 (1.4-10.1)56.3 (53.6-59.0)NHB201193.7 (90.2-97.2)68.7 (62.4-75.0)5.4 (1.2-9.5)55.9 (53.2-58.7)NHB201494.7 (91.4-98.0)69.2 (63.1-75.4)5.9 (1.7-10.1)56.6 (53.9-59.3)NHW199292.4 (91.1-93.7)64.4 (62.2-66.5)4.8 (3.2-6.5)53.9 (52.9-54.9)NHW199591.8 (90.5-93.2)65.6 (63.3-67.8)5.9 (4.2-7.6)54.4 (53.4-55.5)NHW199893.2 (92-94.4)67.8 (65.7-69.9)4.2 (2.7-5.7)55.1 (54.1-56)NHW200293.7 (92.6-94.9)73.4 (71.3-75.5)4.1 (2.7-5.5)57.1 (56.2-58)NHW200595.3 (94.2-96.3)77.7 (75.7-79.7)5.1 (3.5-6.7)59.4 (58.4-60.3)NHW200895.4 (94.4-96.4)77.6 (75.6-79.7)9.0 (7.0-11.1)60.7 (59.7-61.7)NHW201196.1 (95.1-97.2)78.8 (76.7-80.9)4.8 (3.3-6.4)59.9 (59.0-60.9)NHW201496.2 (95.1-97.3)79.3 (77.2-81.4)8.1 (6.2-10.1)61.2 (60.2-62.2)Table 5.Adjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199278.8 (64.3-93.4)56.6 (40.4-72.7)7.5 (.0-26.0)47.6 (38.1-57.1)AIAN199585.5 (70.3-100)62.1 (46.8-77.4)13.3 (.0-31.0)53.7 (44.4-63.0)AIAN199881.1 (67.1-95.0)70.0 (55.6-84.3)2.8 (.0-18.0)51.3 (42.8-59.7)AIAN200289.4 (77.9-100)68.8 (56.3-81.3)8.2 (.0-29.1)55.5 (46.5-64.5)AIAN200595.6 (85.2-100)63.8 (52.0-75.6)20.4 (5.8-35.0)59.9 (52.7-67.1)AIAN200893.6 (81.8-100)79.8 (68.3-91.2)4.3 (.0-19.5)59.2 (51.8-66.7)AIAN201184.9 (73.8-96.0)65.4 (54.4-76.3)7.3 (.0-19.2)52.5 (46.0-59.1)AIAN201493.0 (84.0-100)68.6 (58.4-78.7)13.3 (1.5-25.1)58.3 (52.3-64.3)API199295.4 (92.1-98.8)63.1 (57.6-68.5)6.5 (.9-12.0)55.0 (52.1-57.9)API199592.3 (88.9-95.7)67.1 (62.1-72.1)6.4 (1.1-11.6)55.3 (52.5-58.0)API199895.2 (92.4-98.1)75.7 (71.2-80.2)8.8 (3.8-13.7)59.9 (57.4-62.4)API200294.5 (92.2-96.7)78.1 (74.2-82.1)7.9 (3.7-12.1)60.2 (58.0-62.3)API200593.7 (91.4-96.0)78.3 (74.6-82.1)10.6 (6.0-15.2)60.9 (58.6-63.1)API200895.3 (93.3-97.2)78.9 (74.9-82.9)10.7 (6.2-15.2)61.6 (59.4-63.8)API201193.2 (90.9-95.4)75.4 (71.5-79.3)9.3 (5.0-13.7)59.3 (57.1-61.5)API201492.6 (90.5-94.7)78.9 (75.2-82.5)11.6 (7.8-15.4)61.0 (59.0-63.0)Hispanic199289.7 (85.3-94.2)57.3 (51.5-63.0)8.6 (2.5-14.8)51.9 (48.6-55.1)Hispanic199588.8 (84.1-93.5)60.2 (54.4-66.0)5.6 (.2-11.0)51.5 (48.4-54.7)Hispanic199891.2 (87.8-94.6)71.2 (66.1-76.4)6.5 (1.1-12.0)56.3 (53.5-59.1)Hispanic200291.0 (87.7-94.4)73.6 (68.8-78.4)5.8 (1.0-10.5)56.8 (54.2-59.4)Hispanic200591.5 (88.7-94.4)72.7 (68.3-77.0)8.8 (4.0-13.5)57.7 (55.2-60.1)Hispanic200892.6 (90.2-95.0)71.6 (67.2-76.1)6.6 (2.3-10.8)57.0 (54.7-59.2)Hispanic201194.6 (92.6-96.6)76.7 (72.7-80.7)8.4 (4.5-12.3)59.9 (57.8-62.0)Hispanic201492.5 (90.4-94.5)72.8 (68.9-76.7)9.6 (5.7-13.6)58.3 (56.2-60.4)NHB199287.5 (83.0-91.9)59.6 (53.4-65.8)1.3 (.0-5.5)49.4 (46.5-52.4)NHB199589.5 (85.2-93.7)62.3 (56.2-68.5)3.4 (.0-8.3)51.7 (48.7-54.8)NHB199892.7 (89.5-95.9)64.6 (58.7-70.4)5.4 (.9-10.0)54.2 (51.5-57.0)NHB200286.8 (82.5-91.0)69.7 (64.5-74.9)4.2 (.0-8.4)53.6 (50.8-56.3)NHB200589.0 (85.4-92.6)71.5 (66.2-76.8)3.7 (.0-7.5)54.7 (52.2-57.3)NHB200890.0 (86.9-93.2)67.7 (62.2-73.1)6.2 (2.1-10.2)54.6 (52.1-57.2)NHB201190.0 (86.7-93.2)66.8 (61.1-72.6)4.0 (.1-7.8)53.6 (51.0-56.2)NHB201490.9 (87.8-94.0)68.0 (62.4-73.6)6.1 (2.2-10.0)55.0 (52.4-57.6)NHW199290.7 (89.3-92.1)65.1 (63.0-67.2)7.3 (5.7-9.0)54.4 (53.2-55.6)NHW199590.5 (89.1-92.0)66.4 (64.3-68.6)8.0 (6.3-9.8)55.0 (53.8-56.2)NHW199891.1 (89.8-92.4)68.6 (66.6-70.7)9.4 (7.8-10.9)56.4 (55.2-57.5)NHW200292.1 (90.8-93.4)73.5 (71.5-75.6)8.7 (7.2-10.2)58.1 (57.0-59.3)NHW200592.3 (91.0-93.5)77.1 (75.1-79.0)10.7 (9.0-12.3)60.0 (58.9-61.1)NHW200892.7 (91.5-93.9)77.1 (75.0-79.1)12.3 (10.3-14.3)60.7 (59.5-61.9)NHW201193.5 (92.2-94.7)78.0 (75.9-80.0)10.7 (9.1-12.3)60.7 (59.6-61.9)NHW201492.3 (91.1-93.6)77.7 (75.7-79.8)13.2 (11.3-15.1)61.1 (59.9-62.3)Figure 3.Trends in 5-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2014. Unadjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis. Adjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis. Trends in 5-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2014. The trends reflect improvements in 5-year cause-specific survival probabilities over time for all CRC stages. The rate of improvement in 5-year survival probabilities over time was greatest for those with regional stage CRC, although the rate of improvement appears to be slowing in the second half of the study period. As there are no significantly different trends over time in adjusted 5-year survival probabilities by race or ethnicity, key differences among race and ethnic groups are best summarized by differences in their adjusted time-averaged survival probabilities. For those diagnosed at localized stage CRC, estimates of average adjusted 5-year survival probabilities were 86.9% (84.2%-89.6%) for AIAN, 90.2% (89.4%-91.0%) for NHB, 92.0% (91.8%-92.2%) for NHW, 92.1% (91.5%-92.7%) for Hispanic, and 94.1% (93.5%-94.7%) for API. These differed significantly between all pairs of groups (all P < .04), except for the Hispanic vs NHW comparison (P = .56). For those diagnosed at regional stage CRC, estimates of average adjusted 5-year survival probabilities were 65.1% (62.2%-68.0%) for NHB, 67.5% (61.0%-74.0%) for AIAN, 70.5% (68.1%-72.9%) for Hispanic, 72.5% (71.5%-73.5%) for NHW, and 75.1% (72.9%-77.3%) for API persons. These differed significantly between all pairs of groups (all P < .05), except for the AIAN vs NHB comparison (P = .22). For those diagnosed at distant stage CRC, estimates of average adjusted 5-year survival probabilities were 4.6% (3.6%-5.6%) for NHB, 7.8% (6.6%-9.0%) for Hispanic, 8.5% (7.3%-9.7%) for API, 9.0% (5.5%-12.5%) for AIAN, and 9.6% (9.2%-10.0%) for NHW. The AIAN average did not differ significantly from those of the API, Hispanic, or NHW groups, nor did the API vs Hispanic averages. All others differed significantly (all P < .01). Estimates of adjusted 5-year cause-specific survival probabilities are shown for all race/ethnicity by stage combinations in Tables 4 and 5. Neither the race/ethnicity by stage by year of diagnosis three-way interaction (P = .90) nor the two-way interaction between the year of diagnosis and race/ethnicity (P = .59) were statistically significant; there is not sufficient evidence to conclude that there are race- or ethnic-specific differences in 5-year survival trends either within or across stages of CRC at diagnosis. There were statistically significant interactions between race/ethnicity and stage (P < .001), and between stage and the year of diagnosis trends (P < .001) with respect to differences in 5-year survival (see Figure 3).Table 4.Unadjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199283.2 (67.0-99.3)58.1 (40.1-76.0)8.5 (0-29.1)49.9 (39.3-60.5)AIAN199587.5 (70.6-100)64.7 (47.7-81.7)13.4 (0-33.0)55.2 (44.9-65.5)AIAN199885.4 (69.9-100)73.1 (57.2-89.0)1.6 (0-18.5)53.3 (44.0-62.6)AIAN200291.8 (79.0-100)70.4 (56.5-84.3)10.8 (0-33.9)57.6 (47.7-67.6)AIAN200597.1 (85.6-100)65.6 (52.5-78.6)23.0 (6.8-39.2)61.9 (54.0-69.8)AIAN200897.5 (84.4-100)83.4 (70.7-96.1)5.9 (0-22.7)62.3 (54.0-70.6)AIAN201189.3 (77.0-100)67.5 (55.3-79.6)9.5 (0.0-22.7)55.4 (48.2-62.7)AIAN201496.5 (86.4-100)71.4 (60.1-82.6)15.1 (2.0-28.2)61.0 (54.3-67.6)API199297.1 (93.4-100)63.9 (57.9-70.0)7.1 (.9-13.2)56.0 (52.9-59.1)API199594.8 (91.1-98.4)67.5 (62.0-73.0)4.9 (0-10.7)55.7 (52.8-58.6)API199896.1 (93.0-99.2)76.8 (71.8-81.8)7.9 (2.5-13.4)60.3 (57.6-62.9)API200296.7 (94.3-99.1)79.8 (75.4-84.1)7.1 (2.5-11.8)61.2 (58.9-63.5)API200596.5 (94.1-98.9)80.0 (75.9-84.1)9.8 (4.7-14.8)62.1 (59.8-64.4)API200897.7 (95.7-99.7)80.6 (76.3-85)9.0 (4.1-13.9)62.4 (60.1-64.7)API201196.7 (94.4-99.0)76.7 (72.5-81.0)8.4 (3.6-13.1)60.6 (58.3-62.9)API201497.2 (95.0-99.4)81.5 (77.6-85.5)8.0 (3.8-12.1)62.2 (60.2-64.3)Hispanic199292.6 (87.7-97.5)58.1 (51.7-64.4)6.7 (0-13.4)52.4 (48.9-55.9)Hispanic199590.7 (85.5-95.8)61.0 (54.6-67.4)5.4 (0-11.4)52.4 (49.0-55.8)Hispanic199894.3 (90.6-98.0)72.7 (67.0-78.3)5.5 (0-11.5)57.5 (54.4-60.5)Hispanic200293.8 (90.1-97.4)75.5 (70.3-80.8)6.0 (.8-11.2)58.4 (55.7-61.2)Hispanic200594.6 (91.6-97.6)74.1 (69.3-78.9)9.3 (4.1-14.5)59.3 (56.8-61.9)Hispanic200896.0 (93.5-98.6)74.0 (69.1-78.9)6.3 (1.6-10.9)58.8 (56.4-61.2)Hispanic201197.6 (95.5-99.6)79 (74.6-83.3)8.9 (4.6-13.1)61.8 (59.6-63.9)Hispanic201496.9 (94.8-99)75.2 (70.9-79.4)9.4 (5.1-13.7)60.5 (58.4-62.6)NHB199290.6 (85.7-95.4)60.9 (54.1-67.7)2.2 (0-6.8)51.2 (48.1-54.4)NHB199591.3 (86.7-95.9)63.8 (57.0-70.6)4.3 (0-9.6)53.1 (49.8-56.4)NHB199896.0 (92.5-99.4)66.4 (60.0-72.9)4.8 (0-9.8)55.7 (52.8-58.7)NHB200289.9 (85.2-94.6)71.6 (65.9-77.3)4.5 (0-9.0)55.3 (52.4-58.2)NHB200591.8 (87.9-95.7)73.5 (67.7-79.3)4.3 (.1-8.4)56.5 (53.8-59.2)NHB200893.8 (90.4-97.1)69.3 (63.3-75.3)5.7 (1.4-10.1)56.3 (53.6-59.0)NHB201193.7 (90.2-97.2)68.7 (62.4-75.0)5.4 (1.2-9.5)55.9 (53.2-58.7)NHB201494.7 (91.4-98.0)69.2 (63.1-75.4)5.9 (1.7-10.1)56.6 (53.9-59.3)NHW199292.4 (91.1-93.7)64.4 (62.2-66.5)4.8 (3.2-6.5)53.9 (52.9-54.9)NHW199591.8 (90.5-93.2)65.6 (63.3-67.8)5.9 (4.2-7.6)54.4 (53.4-55.5)NHW199893.2 (92-94.4)67.8 (65.7-69.9)4.2 (2.7-5.7)55.1 (54.1-56)NHW200293.7 (92.6-94.9)73.4 (71.3-75.5)4.1 (2.7-5.5)57.1 (56.2-58)NHW200595.3 (94.2-96.3)77.7 (75.7-79.7)5.1 (3.5-6.7)59.4 (58.4-60.3)NHW200895.4 (94.4-96.4)77.6 (75.6-79.7)9.0 (7.0-11.1)60.7 (59.7-61.7)NHW201196.1 (95.1-97.2)78.8 (76.7-80.9)4.8 (3.3-6.4)59.9 (59.0-60.9)NHW201496.2 (95.1-97.3)79.3 (77.2-81.4)8.1 (6.2-10.1)61.2 (60.2-62.2)Table 5.Adjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199278.8 (64.3-93.4)56.6 (40.4-72.7)7.5 (.0-26.0)47.6 (38.1-57.1)AIAN199585.5 (70.3-100)62.1 (46.8-77.4)13.3 (.0-31.0)53.7 (44.4-63.0)AIAN199881.1 (67.1-95.0)70.0 (55.6-84.3)2.8 (.0-18.0)51.3 (42.8-59.7)AIAN200289.4 (77.9-100)68.8 (56.3-81.3)8.2 (.0-29.1)55.5 (46.5-64.5)AIAN200595.6 (85.2-100)63.8 (52.0-75.6)20.4 (5.8-35.0)59.9 (52.7-67.1)AIAN200893.6 (81.8-100)79.8 (68.3-91.2)4.3 (.0-19.5)59.2 (51.8-66.7)AIAN201184.9 (73.8-96.0)65.4 (54.4-76.3)7.3 (.0-19.2)52.5 (46.0-59.1)AIAN201493.0 (84.0-100)68.6 (58.4-78.7)13.3 (1.5-25.1)58.3 (52.3-64.3)API199295.4 (92.1-98.8)63.1 (57.6-68.5)6.5 (.9-12.0)55.0 (52.1-57.9)API199592.3 (88.9-95.7)67.1 (62.1-72.1)6.4 (1.1-11.6)55.3 (52.5-58.0)API199895.2 (92.4-98.1)75.7 (71.2-80.2)8.8 (3.8-13.7)59.9 (57.4-62.4)API200294.5 (92.2-96.7)78.1 (74.2-82.1)7.9 (3.7-12.1)60.2 (58.0-62.3)API200593.7 (91.4-96.0)78.3 (74.6-82.1)10.6 (6.0-15.2)60.9 (58.6-63.1)API200895.3 (93.3-97.2)78.9 (74.9-82.9)10.7 (6.2-15.2)61.6 (59.4-63.8)API201193.2 (90.9-95.4)75.4 (71.5-79.3)9.3 (5.0-13.7)59.3 (57.1-61.5)API201492.6 (90.5-94.7)78.9 (75.2-82.5)11.6 (7.8-15.4)61.0 (59.0-63.0)Hispanic199289.7 (85.3-94.2)57.3 (51.5-63.0)8.6 (2.5-14.8)51.9 (48.6-55.1)Hispanic199588.8 (84.1-93.5)60.2 (54.4-66.0)5.6 (.2-11.0)51.5 (48.4-54.7)Hispanic199891.2 (87.8-94.6)71.2 (66.1-76.4)6.5 (1.1-12.0)56.3 (53.5-59.1)Hispanic200291.0 (87.7-94.4)73.6 (68.8-78.4)5.8 (1.0-10.5)56.8 (54.2-59.4)Hispanic200591.5 (88.7-94.4)72.7 (68.3-77.0)8.8 (4.0-13.5)57.7 (55.2-60.1)Hispanic200892.6 (90.2-95.0)71.6 (67.2-76.1)6.6 (2.3-10.8)57.0 (54.7-59.2)Hispanic201194.6 (92.6-96.6)76.7 (72.7-80.7)8.4 (4.5-12.3)59.9 (57.8-62.0)Hispanic201492.5 (90.4-94.5)72.8 (68.9-76.7)9.6 (5.7-13.6)58.3 (56.2-60.4)NHB199287.5 (83.0-91.9)59.6 (53.4-65.8)1.3 (.0-5.5)49.4 (46.5-52.4)NHB199589.5 (85.2-93.7)62.3 (56.2-68.5)3.4 (.0-8.3)51.7 (48.7-54.8)NHB199892.7 (89.5-95.9)64.6 (58.7-70.4)5.4 (.9-10.0)54.2 (51.5-57.0)NHB200286.8 (82.5-91.0)69.7 (64.5-74.9)4.2 (.0-8.4)53.6 (50.8-56.3)NHB200589.0 (85.4-92.6)71.5 (66.2-76.8)3.7 (.0-7.5)54.7 (52.2-57.3)NHB200890.0 (86.9-93.2)67.7 (62.2-73.1)6.2 (2.1-10.2)54.6 (52.1-57.2)NHB201190.0 (86.7-93.2)66.8 (61.1-72.6)4.0 (.1-7.8)53.6 (51.0-56.2)NHB201490.9 (87.8-94.0)68.0 (62.4-73.6)6.1 (2.2-10.0)55.0 (52.4-57.6)NHW199290.7 (89.3-92.1)65.1 (63.0-67.2)7.3 (5.7-9.0)54.4 (53.2-55.6)NHW199590.5 (89.1-92.0)66.4 (64.3-68.6)8.0 (6.3-9.8)55.0 (53.8-56.2)NHW199891.1 (89.8-92.4)68.6 (66.6-70.7)9.4 (7.8-10.9)56.4 (55.2-57.5)NHW200292.1 (90.8-93.4)73.5 (71.5-75.6)8.7 (7.2-10.2)58.1 (57.0-59.3)NHW200592.3 (91.0-93.5)77.1 (75.1-79.0)10.7 (9.0-12.3)60.0 (58.9-61.1)NHW200892.7 (91.5-93.9)77.1 (75.0-79.1)12.3 (10.3-14.3)60.7 (59.5-61.9)NHW201193.5 (92.2-94.7)78.0 (75.9-80.0)10.7 (9.1-12.3)60.7 (59.6-61.9)NHW201492.3 (91.1-93.6)77.7 (75.7-79.8)13.2 (11.3-15.1)61.1 (59.9-62.3)Figure 3.Trends in 5-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2014. Unadjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis. Adjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis. Trends in 5-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2014. The trends reflect improvements in 5-year cause-specific survival probabilities over time for all CRC stages. The rate of improvement in 5-year survival probabilities over time was greatest for those with regional stage CRC, although the rate of improvement appears to be slowing in the second half of the study period. As there are no significantly different trends over time in adjusted 5-year survival probabilities by race or ethnicity, key differences among race and ethnic groups are best summarized by differences in their adjusted time-averaged survival probabilities. For those diagnosed at localized stage CRC, estimates of average adjusted 5-year survival probabilities were 86.9% (84.2%-89.6%) for AIAN, 90.2% (89.4%-91.0%) for NHB, 92.0% (91.8%-92.2%) for NHW, 92.1% (91.5%-92.7%) for Hispanic, and 94.1% (93.5%-94.7%) for API. These differed significantly between all pairs of groups (all P < .04), except for the Hispanic vs NHW comparison (P = .56). For those diagnosed at regional stage CRC, estimates of average adjusted 5-year survival probabilities were 65.1% (62.2%-68.0%) for NHB, 67.5% (61.0%-74.0%) for AIAN, 70.5% (68.1%-72.9%) for Hispanic, 72.5% (71.5%-73.5%) for NHW, and 75.1% (72.9%-77.3%) for API persons. These differed significantly between all pairs of groups (all P < .05), except for the AIAN vs NHB comparison (P = .22). For those diagnosed at distant stage CRC, estimates of average adjusted 5-year survival probabilities were 4.6% (3.6%-5.6%) for NHB, 7.8% (6.6%-9.0%) for Hispanic, 8.5% (7.3%-9.7%) for API, 9.0% (5.5%-12.5%) for AIAN, and 9.6% (9.2%-10.0%) for NHW. The AIAN average did not differ significantly from those of the API, Hispanic, or NHW groups, nor did the API vs Hispanic averages. All others differed significantly (all P < .01).
null
null
[ "Study Design and Data Source", "Study Population and Variables", "Statistical Analysis", "One- and Five-Year Survival: All Persons With CRC", "One-Year Survival by Race and Ethnicity", "Five-Year Survival by Race and Ethnicity" ]
[ "This study was based on a retrospective cohort of CRC patients, ascertained on a population level, with data captured as part of the National Cancer Institute’s Surveillance, Epidemiology and End Results (SEER) program. The SEER program collects cancer incidence and survival data from population-based cancer registries representing approximately 35% of the U.S. population.23 Data from the 12 SEER registries for the period 1992 to 2018 were used in this analysis. The 12 SEER registries cover Alaska, Connecticut, Atlanta, rural Georgia, San Francisco-Oakland, San Jose-Monterey, Hawaii, Iowa, Los Angeles, New Mexico, Seattle-Puget Sound and Utah.24 This project, Study ID: 21-102, was reviewed by the University of New Mexico Health Sciences Institutional Review Board, and was granted exemption on 31 March 2021 according to Category 4: Secondary research on data or specimens (no consent required). This report conforms to RECORD guidelines for SEER-based studies.25 This study was supported by funding from the National Cancer Institute; the funders played no other role in this work. Researchers desiring additional details about the data and programs used to carry out this work may obtain programming code from the corresponding author.", "We used SEER*Stat software (version 8.4.0)26 as the data source for this study. We used the “Incidence––SEER Research Plus Data, 12 Registries, Nov 2021 Sub (1992-2019)” database. We included a consecutive series of all individuals who received their first primary CRC diagnosis with malignant behavior from 1992 through 2018. We excluded “All Death Certificate Only and Autopsy Only” and “Alive with No Survival Time” via checkboxes in SEER*Stat. We also excluded CRC cases without a record for summary stage at diagnosis, as well as those with unknown age at diagnosis. We included all CRC cases that originated from the cecum, ascending colon, hepatic flexure of colon, transverse colon, splenic flexure of colon, descending colon, sigmoid colon, overlapping lesion of colon, colon not otherwise specified, rectosigmoid junction, and rectum. We classified CRC cases as localized, regional and distant stage using SEER’s “Combined Summary Stage (2004+)” classifications, supplemented with values from the “Historic Stage A (1973-2015)” variable when necessary. In-situ cancers and cases that were un-staged/unknown were excluded. These rules, as applied in SEER*Stat 8.0.4, defined the number of individuals included in this study.\nThe SEER program works closely with providers of cancer care in their population-based catchment areas to collect patient- and cancer-specific information. Because of its population-based nature, it is typically necessary to rely on medical records as the source of the demographic and other data. SEER-defined race and ethnic categories were utilized and labeled as follows: Non-Hispanic White (NHW), Non-Hispanic Black (NHB), Hispanic, Non-Hispanic American Indian/Alaska Native (AIAN), and Non-Hispanic Asian or Pacific Islander (API). Additional variables were extracted from SEER, and estimates of CRC-specific survival probabilities were obtained for combinations of these variables in the primary analyses: sex as assigned at birth (male or female), age at diagnosis (categorized into decades), year of diagnosis, grade (grades I-IV, or unknown), and Rural-Urban Continuum Code in 2003 (RUCC, coded as: Metro Counties, Non-Metro Counties [Metro-Adjacent], Non-Metro Counties [not Metro-Adjacent], and Alaska or Hawaii, or unknown).\nThe outcome of interest in this work was cause-specific survival for CRC, estimated at 1- and 5-years post diagnosis. We used SEER*Stat software (version 8.4.0)26 to calculate 1- and 5-year cause-specific survival probabilities of CRC using the “Incidence – SEER Research Plus Data, 12 Registries, Nov 2021 (1992-2019)” database while relying on the rules implemented in SEER*Stat for loss to follow-up. We obtained cause-specific survival probabilities and their standard errors within combinations of the characteristics of interest, ie, year of diagnosis, race and ethnicity, stage, sex, grade, and RUCC. No person-level data were used, nor were the SEER data linked to any other data sources. Using SEER*Stat 8.4.0, we formed tables of cause-specific survival probabilities and their standard errors according to all combinations of the factors of interest, with separate “Pages” defined for combinations of Year of diagnosis, age at diagnosis (in decades), and RUCC groups, and with rows defined by combinations of race/ethnicity, summary stage, grade, and sex. We used text processing approaches to form an analysis-ready data set which enumerated the numbers of individuals with a primary CRC diagnosis, and estimates of cause-specific survival probabilities and standard errors, within categories defined by all of the factors of interest in this study.", "We summarized the numbers and percentages of individuals with CRC diagnoses within the levels of the variables of interest. We used linear models to perform meta-regressions, weighting by the inverse of the squared standard errors, to estimate the degree to which the combinations of the variables of interest explained differences in the estimated cause-specific survival probabilities. When the SEER-estimated standard errors of survival probabilities were equal to zero, we approximated them with the square root of the variance of the binomial distribution, calculated after adding a value of .5 to both the numerator and denominator counts reported by SEER for the relevant group. We modelled the survival probabilities obtained at 1 year and 5 years post-diagnosis separately. The initial models included all possible interactions among race/ethnicity, stage, and year of diagnosis, while controlling for potential confounding by age at diagnosis (categorized into decades), sex, grade, and RUCC. We simplified these initial models subsequently modelling year-at-diagnosis trends with natural cubic spline basis functions, and selected the degree of smoothing that best explained the race by stage by year of diagnosis trends by minimizing Akaike’s Information Criterion (AIC) while adjusting for the other factors. We obtained smoothed trend estimates within groups defined by race and ethnicity, and by CRC stage at diagnosis, from a separate model for each of the two follow-up time periods. These final models were obtained by removing non-significant interactions in a hierarchical fashion. Analyses were performed using the tools available in SEER*Stat26 and SAS 9.4 (SAS Institute Inc, Cary, NC). Two-sided type I error was set at .05 for tests of significance.", "Adjusted 1- and 5-year cause-specific survival probabilities following CRC diagnosis by stage and year of diagnosis are shown in Figure 1. For those diagnosed with local stage CRC, the estimate of linear trend in 1-year survival probabilities was negligible, at −.01% ([95% Confidence Interval (CI)] −.03%-.01%) per year, with an average 1-year survival probability of 96.2% (93.8%-98.6%). Those diagnosed with regional stage CRC displayed a small improvement over time in 1-year survival probabilities, .06% (.001%-.12%) per year. Their average 1-year survival probability was 93.0% (90.5%-95.5%). For those diagnosed with distant stage CRC, there was a significant improvement in 1-year survival probabilities over time (P < .001), but there was evidence of departure from a linear trend (P = .04). The slope of the trend line suggested an improvement in survival probability of 1.0% (.8%-1.2%) per year in 2005, with instantaneous slopes suggesting greater gains, by .05% (.01%-.09%), for each year prior to 2005 and slower gains by that same amount for each year following 2005. Their average 1-year survival probability was 52.8% (48.1%-57.5%).Figure 1.Trends in 1- and 5-year adjusted cause-specific survival probabilities for individuals diagnosed at different stages of colorectal cancer. Shaded bands reflect 95% prediction intervals for the year-specific survival probabilities.\nTrends in 1- and 5-year adjusted cause-specific survival probabilities for individuals diagnosed at different stages of colorectal cancer. Shaded bands reflect 95% prediction intervals for the year-specific survival probabilities.\nThose diagnosed with local stage CRC experienced improvements in 5-year cause-specific survival, with an average improvement of .16% (.10%-.21%) per year. Their average 5-year survival probability was 90.9% (87.9%-93.8%). Those diagnosed with regional stage CRC displayed significant improvement in 5-year survival probabilities over time (P < .001), but there was evidence of significant departure from a linear trend (P = .004), such that the slope of the trend line showed an improvement of .55% (.41%-.69%) per year in 2003, with instantaneous slopes suggested greater gains, by .08% (.04-.12%), for each year prior to 2003 and slower gains by that same amount for each year following 2003. Their average 5-year survival probability was 70.3% (66.5%-74.0%). For those diagnosed with distant stage CRC, there was a significant but small improvement over time in 5-year survival probabilities, .15% (.5%-.25%) per year. Their average 5-year survival probability was 8.1% (4.4%-11.8%).", "Estimates of adjusted 1-year cause-specific survival probabilities following diagnosis are shown for all race/ethnicity by stage combinations in Tables 2 and 3. Trends in 1-year survival for those of different races/ethnicities who were diagnosed at different stages of CRC differed significantly (P < .001, see Figure 2). For those diagnosed with localized stage CRC, 1-year survival probabilities were consistently high for individuals of all race and ethnic groups. AIAN persons experienced lower 1-year survival probabilities than those from other race and ethnic groups over the study period. Although due to lack of precision in the estimates, the difference was only significantly different when compared to API persons; average 1-year survival probabilities were 1.5 (.5-2.5) percentage points lower for AIAN than for API persons.Table 2.Unadjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN1992100 (84.6-100)91.6 (75.4-100)27.0 (5.6-48.4)72.9 (62.6-83.2)AIAN199696.9 (81.8-100)95.7 (77.9-100)41.8 (23.5-60.1)78.1 (68.2-88.0)AIAN200098.7 (83.6-100)83.2 (65.9-100)43.0 (20.0-66.0)75.0 (64.1-85.8)AIAN200496.7 (85.6-100)96.4 (84.8-100)51.8 (35.8-67.7)81.6 (74.1-89.2)AIAN2008100 (86.4-100)95.5 (83.0-100)53.5 (35.5-71.5)83.0 (74.4-91.6)AIAN2012100 (88.9-100)91.6 (79.1-100)52.7 (38.0-67.4)81.4 (74.0-88.8)AIAN201696.0 (85.5-100)95.9 (85.6-100)46.5 (33.4-59.6)79.5 (72.9-86.0)AIAN2018100 (90.7-100)99.3 (88.9-100)83.5 (67.2-99.8)94.3 (87.1-100)API199299.5 (96.2-100)94.6 (90.8-98.3)46.5 (38.4-54.6)80.2 (77.0-83.3)API199699.4 (97.1-100)95.8 (92.6-98.9)56.5 (48.7-64.3)83.9 (81.0-86.8)API200099.2 (97.0-100)97.9 (95.8-99.9)52.8 (45.0-60.6)83.3 (80.5-86.1)API200499.6 (97.9-100)97.3 (95.1-99.5)68.6 (62.7-74.4)88.5 (86.3-90.6)API200899.7 (98.3-100)98.4 (96.5-100)59.5 (53.9-65.0)85.9 (83.9-87.9)API201299.4 (97.9-100)96.9 (94.7-99.2)55.8 (49.4-62.3)84.1 (81.7-86.4)API201699.1 (97.4-100)98.4 (96.7-100)57.6 (51.8-63.3)85.0 (82.9-87.1)API201899.6 (98.5-100)97.8 (96.0-99.6)69.1 (62.7-75.5)88.8 (86.6-91.1)Hispanic199297.0 (93.0-100)96.7 (93.3-100)55.6 (46.2-64.9)83.1 (79.5-86.7)Hispanic199698.8 (95.8-100)96.1 (92.7-99.5)43.4 (35.7-51.1)79.4 (76.4-82.4)Hispanic200099.2 (96.6-100)96.3 (93.3-99.4)47.0 (39.8-54.2)80.8 (78.1-83.6)Hispanic200498.9 (96.5-100)95.8 (92.9-98.7)57.8 (50.8-64.8)84.2 (81.5-86.8)Hispanic200899.1 (97.3-100)96.2 (93.5-98.8)60.7 (54.4-67.1)85.3 (83.0-87.7)Hispanic201299.2 (97.4-100)95.8 (93.3-98.2)71.6 (66.6-76.7)88.9 (86.9-90.8)Hispanic201698.9 (97.1-100)98.2 (96.6-99.7)69.0 (64.0-74.1)88.7 (86.9-90.6)Hispanic201899.6 (98.6-100)97.8 (96.1-99.5)58.8 (52.5-65.0)85.4 (83.2-87.6)NHB199298.0 (94.7-100)93.7 (89.6-97.8)38.5 (30.0-47.0)76.7 (73.4-80.1)NHB199698.2 (94.7-100)96.3 (93-99.6)30.1 (23.1-37.2)74.9 (72.0-77.7)NHB200097.3 (93.8-100)93.2 (89.4-97.0)34.5 (27.3-41.7)75.0 (72.0-77.9)NHB200498.4 (95.6-100)97.0 (94.3-99.7)37.0 (30.7-43.3)77.5 (75.0-79.9)NHB200898.6 (96.2-100)93.3 (89.4-97.2)51.4 (44.7-58.1)81.1 (78.4-83.8)NHB201299.5 (97.2-100)93.6 (90.0-97.1)68.5 (62.1-74.8)87.2 (84.7-89.7)NHB201699.5 (97.2-100)96.0 (93.0-99.0)53.1 (47.2-58.9)82.9 (80.5-85.2)NHB201899.3 (97.5-100)97.0 (94.2-99.8)57.1 (49.6-64.6)84.5 (81.7-87.2)NHW199298.6 (97.9-99.2)94.6 (93.5-95.7)38.0 (35.0-40.9)77.0 (76.0-78.1)NHW199698.9 (98.3-99.5)96.1 (95.1-97.0)36.7 (33.8-39.5)77.2 (76.2-78.2)NHW200099.0 (98.5-99.6)94.3 (93.2-95.4)40.9 (37.9-43.9)78.1 (77.0-79.1)NHW200498.9 (98.3-99.5)96.1 (95.1-97.1)52.6 (49.7-55.4)82.5 (81.5-83.6)NHW200899.2 (98.6-99.7)97.0 (96.1-98.0)58.6 (55.6-61.7)85.0 (83.9-86.0)NHW201298.9 (98.2-99.6)97.2 (96.2-98.2)55.2 (52.3-58.0)83.8 (82.7-84.8)NHW201699.1 (98.5-99.8)96.9 (95.9-97.9)60.7 (57.9-63.5)85.6 (84.6-86.6)NHW201898.7 (98.0-99.4)97.5 (96.7-98.4)59.0 (55.7-62.3)85.1 (83.9-86.3)Table 3.Adjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199297.5 (83.7-100)89.8 (75.3-100)25.4 (6.2-44.6)70.9 (61.6-80.2)AIAN199695.6 (82.0-100)93.2 (77.3-100)41.9 (25.5-58.3)76.9 (68.0-85.8)AIAN200097.0 (83.5-100)82.5 (67.0-97.9)42.0 (21.4-62.6)73.8 (64.1-83.5)AIAN200495.3 (85.3-100)94.2 (83.8-100)50.4 (36.1-64.7)80.0 (73.2-86.8)AIAN200897.5 (85.3-100)92.4 (81.2-100)51.6 (35.5-67.7)80.5 (72.8-88.2)AIAN201297.5 (87.5-100)89.9 (78.7-100)50.2 (37.0-63.3)79.2 (72.5-85.9)AIAN201693.4 (84.0-100)94.1 (84.8-100)45.0 (33.3-56.8)77.5 (71.6-83.4)AIAN201896.7 (87.8-100)96.9 (87.6-100)64.2 (51.1-77.5)85.9 (79.7-91.7)API199297.5 (94.5-100)92.3 (88.9-95.7)45.8 (38.6-53.1)78.6 (75.7-81.4)API199697.4 (95.2-99.6)93.4 (90.5-96.3)55.6 (48.5-62.6)82.1 (79.4-84.8)API200097.4 (95.3-99.5)95.9 (93.9-97.9)53.1 (46.1-60.1)82.1 (79.6-84.7)API200497.8 (96.2-99.4)94.6 (92.5-96.7)67.1 (61.8-72.4)86.5 (84.5-88.5)API200897.5 (96.0-98.9)95.6 (93.7-97.4)59.4 (54.4-64.4)84.2 (82.2-86.1)API201296.8 (95.3-98.3)94.6 (92.5-96.7)56.1 (50.3-61.9)82.5 (80.3-84.7)API201696.4 (94.8-98.0)95.0 (93.4-96.6)57.8 (52.6-63.0)83.1 (81.1-85.0)API201897.9 (96.5-99.0)96.1 (94.5-97.9)63.8 (58.3-69.3)86.0 (83.9-88.0)Hispanic199294.7 (91.1-98.4)94.4 (91.3-97.5)54.3 (45.9-62.7)81.2 (77.9-84.4)Hispanic199696.8 (93.9-99.6)94.2 (91.0-97.3)42.9 (36.0-49.9)77.9 (75.2-80.7)Hispanic200097.0 (94.6-99.4)93.4 (90.6-96.2)46.6 (40.1-53.0)79.0 (76.4-81.5)Hispanic200497.2 (95.0-99.4)93.3 (90.6-96.0)57.1 (50.8-63.4)82.5 (80.1-85.0)Hispanic200896.2 (94.5-97.9)92.8 (90.4-95.3)59.8 (54.1-65.5)83.0 (80.8-85.2)Hispanic201296.0 (94.3-97.8)92.9 (90.6-95.2)69.3 (64.8-73.9)86.1 (84.2-88.0)Hispanic201694.8 (93.0-96.5)94.1 (92.5-95.6)67.3 (62.8-71.9)85.4 (83.6-87.2)Hispanic201896.8 (95.3-98.3)95.3 (93.7-96.9)63.4 (58.3-68.6)85.2 (83.2-87.2)NHB199295.7 (92.6-98.7)91.1 (87.3-94.8)37.8 (30.2-45.4)74.8 (71.8-77.9)NHB199696.6 (93.4-99.9)93.8 (90.7-96.8)30.8 (24.4-37.1)73.7 (71.1-76.3)NHB200095.6 (92.5-98.8)90.5 (87.1-94.0)35.7 (29.2-42.2)74.0 (71.2-76.7)NHB200495.9 (93.3-98.5)94.0 (91.4-96.5)38.9 (33.2-44.6)76.3 (73.9-78.6)NHB200896.1 (93.9-98.3)90.3 (86.8-93.9)51.2 (45.1-57.2)79.2 (76.7-81.7)NHB201296.6 (94.4-98.7)90.6 (87.4-93.8)66.7 (60.9-72.4)84.6 (82.2-87.0)NHB201696.0 (93.9-98.2)92.3 (89.6-95.1)53.0 (47.7-58.3)80.5 (78.3-82.6)NHB201897.5 (95.6-99.1)94.3 (91.7-97.1)55.2 (49.2-61.3)82.4 (80.0-84.8)NHW199296.8 (95.9-97.7)92.8 (91.6-94.0)39.2 (36.5-42.0)76.3 (75.1-77.4)NHW199696.7 (95.9-97.6)93.1 (92.1-94.2)38.4 (35.8-41.1)76.1 (75.0-77.2)NHW200096.3 (95.5-97.2)92.4 (91.2-93.6)42.4 (39.6-45.1)77.0 (75.9-78.2)NHW200496.5 (95.6-97.3)93.5 (92.4-94.7)53.5 (50.8-56.1)81.2 (80.0-82.3)NHW200896.4 (95.6-97.3)93.8 (92.7-94.9)58.8 (56.0-61.6)83.0 (81.8-84.2)NHW201295.9 (95.0-96.8)93.9 (92.8-95.0)55.7 (53.0-58.3)81.8 (80.7-83.0)NHW201695.2 (94.3-96.0)93.5 (92.4-94.6)60.4 (57.8-63.0)83.0 (81.9-84.2)NHW201896.7 (95.8-97.6)95.0 (93.95-96.1)60.4 (57.6-63.3)84.0 (82.9-85.3)Figure 2.Trends in 1-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2018.\nUnadjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nAdjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nTrends in 1-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2018.\nFor those diagnosed with regional stage CRC, trends in adjusted 1-year cause-specific survival probabilities similarly reflect little change over time, with the possible exception of an imprecisely-estimated positive trend over time among AIAN persons. Although there was little evidence of significant trends over time, there was a clear gradient in the average survival probabilities among those from different race and ethnic groups. AIAN persons had the lowest average survival probability (90.2%, 88.2%-92.2), followed by NHB, NHW, Hispanic, and API persons (93.2%, 92.6%-93.8%; 93.3%, 93.1%-93.5%; 94.2%, 93.8%-94.6%; and 94.8%, 94.4%-95.2%, respectively). All of these estimates were significantly different (all P < .01), except for the comparison between NHB and NHW persons (P = .83).\nThose diagnosed with distant stage CRC experienced the biggest improvements in 1-year cause-specific survival probabilities over time. An increasing trend in 1-year survival probabilities is apparent for persons of all race and ethnic groups. For AIAN and NHB persons, the two groups with lowest initial 1-year survival probabilities, improvements continued at a consistent rate over time. For API, Hispanic, and NHW persons, there appears to be a slowing of the rate of improvement in the second half of the study period. The adjusted time-averaged survival probabilities were low: 46.6% (42.3%-50.9%) for NHB, 48.3% (37.9%-58.7%) for AIAN, 50.5% (48.5%-52.5%) for NHW, 58.1% (54.2%-62.0%) for Hispanic, and 58.3% (54.2%-62.4%) for API persons. All pairwise comparisons among these groups were statistically significant (all P < .01), except for differences between the two groups with the lowest (AIAN vs NHB, P = .87), and between the two groups with the highest (API vs Hispanic, P = .69), average adjusted 1-year cause-specific survival probabilities.", "Estimates of adjusted 5-year cause-specific survival probabilities are shown for all race/ethnicity by stage combinations in Tables 4 and 5. Neither the race/ethnicity by stage by year of diagnosis three-way interaction (P = .90) nor the two-way interaction between the year of diagnosis and race/ethnicity (P = .59) were statistically significant; there is not sufficient evidence to conclude that there are race- or ethnic-specific differences in 5-year survival trends either within or across stages of CRC at diagnosis. There were statistically significant interactions between race/ethnicity and stage (P < .001), and between stage and the year of diagnosis trends (P < .001) with respect to differences in 5-year survival (see Figure 3).Table 4.Unadjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199283.2 (67.0-99.3)58.1 (40.1-76.0)8.5 (0-29.1)49.9 (39.3-60.5)AIAN199587.5 (70.6-100)64.7 (47.7-81.7)13.4 (0-33.0)55.2 (44.9-65.5)AIAN199885.4 (69.9-100)73.1 (57.2-89.0)1.6 (0-18.5)53.3 (44.0-62.6)AIAN200291.8 (79.0-100)70.4 (56.5-84.3)10.8 (0-33.9)57.6 (47.7-67.6)AIAN200597.1 (85.6-100)65.6 (52.5-78.6)23.0 (6.8-39.2)61.9 (54.0-69.8)AIAN200897.5 (84.4-100)83.4 (70.7-96.1)5.9 (0-22.7)62.3 (54.0-70.6)AIAN201189.3 (77.0-100)67.5 (55.3-79.6)9.5 (0.0-22.7)55.4 (48.2-62.7)AIAN201496.5 (86.4-100)71.4 (60.1-82.6)15.1 (2.0-28.2)61.0 (54.3-67.6)API199297.1 (93.4-100)63.9 (57.9-70.0)7.1 (.9-13.2)56.0 (52.9-59.1)API199594.8 (91.1-98.4)67.5 (62.0-73.0)4.9 (0-10.7)55.7 (52.8-58.6)API199896.1 (93.0-99.2)76.8 (71.8-81.8)7.9 (2.5-13.4)60.3 (57.6-62.9)API200296.7 (94.3-99.1)79.8 (75.4-84.1)7.1 (2.5-11.8)61.2 (58.9-63.5)API200596.5 (94.1-98.9)80.0 (75.9-84.1)9.8 (4.7-14.8)62.1 (59.8-64.4)API200897.7 (95.7-99.7)80.6 (76.3-85)9.0 (4.1-13.9)62.4 (60.1-64.7)API201196.7 (94.4-99.0)76.7 (72.5-81.0)8.4 (3.6-13.1)60.6 (58.3-62.9)API201497.2 (95.0-99.4)81.5 (77.6-85.5)8.0 (3.8-12.1)62.2 (60.2-64.3)Hispanic199292.6 (87.7-97.5)58.1 (51.7-64.4)6.7 (0-13.4)52.4 (48.9-55.9)Hispanic199590.7 (85.5-95.8)61.0 (54.6-67.4)5.4 (0-11.4)52.4 (49.0-55.8)Hispanic199894.3 (90.6-98.0)72.7 (67.0-78.3)5.5 (0-11.5)57.5 (54.4-60.5)Hispanic200293.8 (90.1-97.4)75.5 (70.3-80.8)6.0 (.8-11.2)58.4 (55.7-61.2)Hispanic200594.6 (91.6-97.6)74.1 (69.3-78.9)9.3 (4.1-14.5)59.3 (56.8-61.9)Hispanic200896.0 (93.5-98.6)74.0 (69.1-78.9)6.3 (1.6-10.9)58.8 (56.4-61.2)Hispanic201197.6 (95.5-99.6)79 (74.6-83.3)8.9 (4.6-13.1)61.8 (59.6-63.9)Hispanic201496.9 (94.8-99)75.2 (70.9-79.4)9.4 (5.1-13.7)60.5 (58.4-62.6)NHB199290.6 (85.7-95.4)60.9 (54.1-67.7)2.2 (0-6.8)51.2 (48.1-54.4)NHB199591.3 (86.7-95.9)63.8 (57.0-70.6)4.3 (0-9.6)53.1 (49.8-56.4)NHB199896.0 (92.5-99.4)66.4 (60.0-72.9)4.8 (0-9.8)55.7 (52.8-58.7)NHB200289.9 (85.2-94.6)71.6 (65.9-77.3)4.5 (0-9.0)55.3 (52.4-58.2)NHB200591.8 (87.9-95.7)73.5 (67.7-79.3)4.3 (.1-8.4)56.5 (53.8-59.2)NHB200893.8 (90.4-97.1)69.3 (63.3-75.3)5.7 (1.4-10.1)56.3 (53.6-59.0)NHB201193.7 (90.2-97.2)68.7 (62.4-75.0)5.4 (1.2-9.5)55.9 (53.2-58.7)NHB201494.7 (91.4-98.0)69.2 (63.1-75.4)5.9 (1.7-10.1)56.6 (53.9-59.3)NHW199292.4 (91.1-93.7)64.4 (62.2-66.5)4.8 (3.2-6.5)53.9 (52.9-54.9)NHW199591.8 (90.5-93.2)65.6 (63.3-67.8)5.9 (4.2-7.6)54.4 (53.4-55.5)NHW199893.2 (92-94.4)67.8 (65.7-69.9)4.2 (2.7-5.7)55.1 (54.1-56)NHW200293.7 (92.6-94.9)73.4 (71.3-75.5)4.1 (2.7-5.5)57.1 (56.2-58)NHW200595.3 (94.2-96.3)77.7 (75.7-79.7)5.1 (3.5-6.7)59.4 (58.4-60.3)NHW200895.4 (94.4-96.4)77.6 (75.6-79.7)9.0 (7.0-11.1)60.7 (59.7-61.7)NHW201196.1 (95.1-97.2)78.8 (76.7-80.9)4.8 (3.3-6.4)59.9 (59.0-60.9)NHW201496.2 (95.1-97.3)79.3 (77.2-81.4)8.1 (6.2-10.1)61.2 (60.2-62.2)Table 5.Adjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199278.8 (64.3-93.4)56.6 (40.4-72.7)7.5 (.0-26.0)47.6 (38.1-57.1)AIAN199585.5 (70.3-100)62.1 (46.8-77.4)13.3 (.0-31.0)53.7 (44.4-63.0)AIAN199881.1 (67.1-95.0)70.0 (55.6-84.3)2.8 (.0-18.0)51.3 (42.8-59.7)AIAN200289.4 (77.9-100)68.8 (56.3-81.3)8.2 (.0-29.1)55.5 (46.5-64.5)AIAN200595.6 (85.2-100)63.8 (52.0-75.6)20.4 (5.8-35.0)59.9 (52.7-67.1)AIAN200893.6 (81.8-100)79.8 (68.3-91.2)4.3 (.0-19.5)59.2 (51.8-66.7)AIAN201184.9 (73.8-96.0)65.4 (54.4-76.3)7.3 (.0-19.2)52.5 (46.0-59.1)AIAN201493.0 (84.0-100)68.6 (58.4-78.7)13.3 (1.5-25.1)58.3 (52.3-64.3)API199295.4 (92.1-98.8)63.1 (57.6-68.5)6.5 (.9-12.0)55.0 (52.1-57.9)API199592.3 (88.9-95.7)67.1 (62.1-72.1)6.4 (1.1-11.6)55.3 (52.5-58.0)API199895.2 (92.4-98.1)75.7 (71.2-80.2)8.8 (3.8-13.7)59.9 (57.4-62.4)API200294.5 (92.2-96.7)78.1 (74.2-82.1)7.9 (3.7-12.1)60.2 (58.0-62.3)API200593.7 (91.4-96.0)78.3 (74.6-82.1)10.6 (6.0-15.2)60.9 (58.6-63.1)API200895.3 (93.3-97.2)78.9 (74.9-82.9)10.7 (6.2-15.2)61.6 (59.4-63.8)API201193.2 (90.9-95.4)75.4 (71.5-79.3)9.3 (5.0-13.7)59.3 (57.1-61.5)API201492.6 (90.5-94.7)78.9 (75.2-82.5)11.6 (7.8-15.4)61.0 (59.0-63.0)Hispanic199289.7 (85.3-94.2)57.3 (51.5-63.0)8.6 (2.5-14.8)51.9 (48.6-55.1)Hispanic199588.8 (84.1-93.5)60.2 (54.4-66.0)5.6 (.2-11.0)51.5 (48.4-54.7)Hispanic199891.2 (87.8-94.6)71.2 (66.1-76.4)6.5 (1.1-12.0)56.3 (53.5-59.1)Hispanic200291.0 (87.7-94.4)73.6 (68.8-78.4)5.8 (1.0-10.5)56.8 (54.2-59.4)Hispanic200591.5 (88.7-94.4)72.7 (68.3-77.0)8.8 (4.0-13.5)57.7 (55.2-60.1)Hispanic200892.6 (90.2-95.0)71.6 (67.2-76.1)6.6 (2.3-10.8)57.0 (54.7-59.2)Hispanic201194.6 (92.6-96.6)76.7 (72.7-80.7)8.4 (4.5-12.3)59.9 (57.8-62.0)Hispanic201492.5 (90.4-94.5)72.8 (68.9-76.7)9.6 (5.7-13.6)58.3 (56.2-60.4)NHB199287.5 (83.0-91.9)59.6 (53.4-65.8)1.3 (.0-5.5)49.4 (46.5-52.4)NHB199589.5 (85.2-93.7)62.3 (56.2-68.5)3.4 (.0-8.3)51.7 (48.7-54.8)NHB199892.7 (89.5-95.9)64.6 (58.7-70.4)5.4 (.9-10.0)54.2 (51.5-57.0)NHB200286.8 (82.5-91.0)69.7 (64.5-74.9)4.2 (.0-8.4)53.6 (50.8-56.3)NHB200589.0 (85.4-92.6)71.5 (66.2-76.8)3.7 (.0-7.5)54.7 (52.2-57.3)NHB200890.0 (86.9-93.2)67.7 (62.2-73.1)6.2 (2.1-10.2)54.6 (52.1-57.2)NHB201190.0 (86.7-93.2)66.8 (61.1-72.6)4.0 (.1-7.8)53.6 (51.0-56.2)NHB201490.9 (87.8-94.0)68.0 (62.4-73.6)6.1 (2.2-10.0)55.0 (52.4-57.6)NHW199290.7 (89.3-92.1)65.1 (63.0-67.2)7.3 (5.7-9.0)54.4 (53.2-55.6)NHW199590.5 (89.1-92.0)66.4 (64.3-68.6)8.0 (6.3-9.8)55.0 (53.8-56.2)NHW199891.1 (89.8-92.4)68.6 (66.6-70.7)9.4 (7.8-10.9)56.4 (55.2-57.5)NHW200292.1 (90.8-93.4)73.5 (71.5-75.6)8.7 (7.2-10.2)58.1 (57.0-59.3)NHW200592.3 (91.0-93.5)77.1 (75.1-79.0)10.7 (9.0-12.3)60.0 (58.9-61.1)NHW200892.7 (91.5-93.9)77.1 (75.0-79.1)12.3 (10.3-14.3)60.7 (59.5-61.9)NHW201193.5 (92.2-94.7)78.0 (75.9-80.0)10.7 (9.1-12.3)60.7 (59.6-61.9)NHW201492.3 (91.1-93.6)77.7 (75.7-79.8)13.2 (11.3-15.1)61.1 (59.9-62.3)Figure 3.Trends in 5-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2014.\nUnadjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nAdjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nTrends in 5-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2014.\nThe trends reflect improvements in 5-year cause-specific survival probabilities over time for all CRC stages. The rate of improvement in 5-year survival probabilities over time was greatest for those with regional stage CRC, although the rate of improvement appears to be slowing in the second half of the study period. As there are no significantly different trends over time in adjusted 5-year survival probabilities by race or ethnicity, key differences among race and ethnic groups are best summarized by differences in their adjusted time-averaged survival probabilities. For those diagnosed at localized stage CRC, estimates of average adjusted 5-year survival probabilities were 86.9% (84.2%-89.6%) for AIAN, 90.2% (89.4%-91.0%) for NHB, 92.0% (91.8%-92.2%) for NHW, 92.1% (91.5%-92.7%) for Hispanic, and 94.1% (93.5%-94.7%) for API. These differed significantly between all pairs of groups (all P < .04), except for the Hispanic vs NHW comparison (P = .56). For those diagnosed at regional stage CRC, estimates of average adjusted 5-year survival probabilities were 65.1% (62.2%-68.0%) for NHB, 67.5% (61.0%-74.0%) for AIAN, 70.5% (68.1%-72.9%) for Hispanic, 72.5% (71.5%-73.5%) for NHW, and 75.1% (72.9%-77.3%) for API persons. These differed significantly between all pairs of groups (all P < .05), except for the AIAN vs NHB comparison (P = .22). For those diagnosed at distant stage CRC, estimates of average adjusted 5-year survival probabilities were 4.6% (3.6%-5.6%) for NHB, 7.8% (6.6%-9.0%) for Hispanic, 8.5% (7.3%-9.7%) for API, 9.0% (5.5%-12.5%) for AIAN, and 9.6% (9.2%-10.0%) for NHW. The AIAN average did not differ significantly from those of the API, Hispanic, or NHW groups, nor did the API vs Hispanic averages. All others differed significantly (all P < .01)." ]
[ null, null, null, null, null, null ]
[ "Introduction", "Methods", "Study Design and Data Source", "Study Population and Variables", "Statistical Analysis", "Results", "One- and Five-Year Survival: All Persons With CRC", "One-Year Survival by Race and Ethnicity", "Five-Year Survival by Race and Ethnicity", "Discussion" ]
[ "Colorectal cancer (CRC) is the third most commonly diagnosed cancer and the second leading cause of cancer deaths in the United States.1,2 It has been estimated that 151 030 people will be newly diagnosed with CRC and 52 580 people with die from CRC in 2022.2,3 Although CRC mortality rates have decreased significantly since 1975,1-5 available reports suggest that these declines vary substantially by the stage of the disease, and that disparities in mortality rates for racial and ethnically minoritized groups persist.4-15 For example, according to a report by the American Cancer Society, the 2-year relative survival rate for distant-stage CRC increased from 21% in mid-1990’s to 37% for those diagnosed during 2009-2015, with commensurate improvements for regional- and localized-stage CRC.2 This same report asserts that CRC mortality rates are highest for persons who are non-Hispanic Black, followed by those who are American Indian or Alaska Native, and lowest for those who are Asian or Pacific Islander.2\nSeveral studies have compared trends in CRC mortality by the stage of the disease and race but have done so under broad classifications as follows: White and Black;4,7-10 White, Black, and Hispanic;11,16,17 White, Black, Asian, Hispanic, and other;18 or White, Black, and other.19 In particular, limited information is available on CRC survival trends in persons who are American Indian or Alaska Native.20-22 A 2010 study by Edwards et al,5 compared CRC mortality trends in persons belonging to five racial and ethnic groups, but more than 10 years have passed since that report.\nTo the best of our knowledge, no study has simultaneously compared recent trends in CRC-specific mortality by stage of diagnosis for individuals that are non-Hispanic White, non-Hispanic Black, Hispanic, non-Hispanic Asian or Pacific Islander, and non-Hispanic American Indian or Alaska Native. To address this gap, we undertook an analysis of population-based CRC cause-specific survival data to understand the trends in 1- and 5-year CRC cause-specific survival probabilities by stage of diagnosis within five racial and ethnic groups for individuals who received a CRC diagnosis between 1992 and 2018. Understanding differences in cause-specific survival, both over time and across CRC stages, is critical for further investigations into the social, structural, and political determinants that contribute to the disparities noted in CRC outcomes among individuals from distinct racial and ethnic groups. Through our efforts, we aim to provide new impetus to refocus efforts on improving CRC detection and treatment among racial and ethnically minoritized populations.", "[SUBTITLE] Study Design and Data Source [SUBSECTION] This study was based on a retrospective cohort of CRC patients, ascertained on a population level, with data captured as part of the National Cancer Institute’s Surveillance, Epidemiology and End Results (SEER) program. The SEER program collects cancer incidence and survival data from population-based cancer registries representing approximately 35% of the U.S. population.23 Data from the 12 SEER registries for the period 1992 to 2018 were used in this analysis. The 12 SEER registries cover Alaska, Connecticut, Atlanta, rural Georgia, San Francisco-Oakland, San Jose-Monterey, Hawaii, Iowa, Los Angeles, New Mexico, Seattle-Puget Sound and Utah.24 This project, Study ID: 21-102, was reviewed by the University of New Mexico Health Sciences Institutional Review Board, and was granted exemption on 31 March 2021 according to Category 4: Secondary research on data or specimens (no consent required). This report conforms to RECORD guidelines for SEER-based studies.25 This study was supported by funding from the National Cancer Institute; the funders played no other role in this work. Researchers desiring additional details about the data and programs used to carry out this work may obtain programming code from the corresponding author.\nThis study was based on a retrospective cohort of CRC patients, ascertained on a population level, with data captured as part of the National Cancer Institute’s Surveillance, Epidemiology and End Results (SEER) program. The SEER program collects cancer incidence and survival data from population-based cancer registries representing approximately 35% of the U.S. population.23 Data from the 12 SEER registries for the period 1992 to 2018 were used in this analysis. The 12 SEER registries cover Alaska, Connecticut, Atlanta, rural Georgia, San Francisco-Oakland, San Jose-Monterey, Hawaii, Iowa, Los Angeles, New Mexico, Seattle-Puget Sound and Utah.24 This project, Study ID: 21-102, was reviewed by the University of New Mexico Health Sciences Institutional Review Board, and was granted exemption on 31 March 2021 according to Category 4: Secondary research on data or specimens (no consent required). This report conforms to RECORD guidelines for SEER-based studies.25 This study was supported by funding from the National Cancer Institute; the funders played no other role in this work. Researchers desiring additional details about the data and programs used to carry out this work may obtain programming code from the corresponding author.\n[SUBTITLE] Study Population and Variables [SUBSECTION] We used SEER*Stat software (version 8.4.0)26 as the data source for this study. We used the “Incidence––SEER Research Plus Data, 12 Registries, Nov 2021 Sub (1992-2019)” database. We included a consecutive series of all individuals who received their first primary CRC diagnosis with malignant behavior from 1992 through 2018. We excluded “All Death Certificate Only and Autopsy Only” and “Alive with No Survival Time” via checkboxes in SEER*Stat. We also excluded CRC cases without a record for summary stage at diagnosis, as well as those with unknown age at diagnosis. We included all CRC cases that originated from the cecum, ascending colon, hepatic flexure of colon, transverse colon, splenic flexure of colon, descending colon, sigmoid colon, overlapping lesion of colon, colon not otherwise specified, rectosigmoid junction, and rectum. We classified CRC cases as localized, regional and distant stage using SEER’s “Combined Summary Stage (2004+)” classifications, supplemented with values from the “Historic Stage A (1973-2015)” variable when necessary. In-situ cancers and cases that were un-staged/unknown were excluded. These rules, as applied in SEER*Stat 8.0.4, defined the number of individuals included in this study.\nThe SEER program works closely with providers of cancer care in their population-based catchment areas to collect patient- and cancer-specific information. Because of its population-based nature, it is typically necessary to rely on medical records as the source of the demographic and other data. SEER-defined race and ethnic categories were utilized and labeled as follows: Non-Hispanic White (NHW), Non-Hispanic Black (NHB), Hispanic, Non-Hispanic American Indian/Alaska Native (AIAN), and Non-Hispanic Asian or Pacific Islander (API). Additional variables were extracted from SEER, and estimates of CRC-specific survival probabilities were obtained for combinations of these variables in the primary analyses: sex as assigned at birth (male or female), age at diagnosis (categorized into decades), year of diagnosis, grade (grades I-IV, or unknown), and Rural-Urban Continuum Code in 2003 (RUCC, coded as: Metro Counties, Non-Metro Counties [Metro-Adjacent], Non-Metro Counties [not Metro-Adjacent], and Alaska or Hawaii, or unknown).\nThe outcome of interest in this work was cause-specific survival for CRC, estimated at 1- and 5-years post diagnosis. We used SEER*Stat software (version 8.4.0)26 to calculate 1- and 5-year cause-specific survival probabilities of CRC using the “Incidence – SEER Research Plus Data, 12 Registries, Nov 2021 (1992-2019)” database while relying on the rules implemented in SEER*Stat for loss to follow-up. We obtained cause-specific survival probabilities and their standard errors within combinations of the characteristics of interest, ie, year of diagnosis, race and ethnicity, stage, sex, grade, and RUCC. No person-level data were used, nor were the SEER data linked to any other data sources. Using SEER*Stat 8.4.0, we formed tables of cause-specific survival probabilities and their standard errors according to all combinations of the factors of interest, with separate “Pages” defined for combinations of Year of diagnosis, age at diagnosis (in decades), and RUCC groups, and with rows defined by combinations of race/ethnicity, summary stage, grade, and sex. We used text processing approaches to form an analysis-ready data set which enumerated the numbers of individuals with a primary CRC diagnosis, and estimates of cause-specific survival probabilities and standard errors, within categories defined by all of the factors of interest in this study.\nWe used SEER*Stat software (version 8.4.0)26 as the data source for this study. We used the “Incidence––SEER Research Plus Data, 12 Registries, Nov 2021 Sub (1992-2019)” database. We included a consecutive series of all individuals who received their first primary CRC diagnosis with malignant behavior from 1992 through 2018. We excluded “All Death Certificate Only and Autopsy Only” and “Alive with No Survival Time” via checkboxes in SEER*Stat. We also excluded CRC cases without a record for summary stage at diagnosis, as well as those with unknown age at diagnosis. We included all CRC cases that originated from the cecum, ascending colon, hepatic flexure of colon, transverse colon, splenic flexure of colon, descending colon, sigmoid colon, overlapping lesion of colon, colon not otherwise specified, rectosigmoid junction, and rectum. We classified CRC cases as localized, regional and distant stage using SEER’s “Combined Summary Stage (2004+)” classifications, supplemented with values from the “Historic Stage A (1973-2015)” variable when necessary. In-situ cancers and cases that were un-staged/unknown were excluded. These rules, as applied in SEER*Stat 8.0.4, defined the number of individuals included in this study.\nThe SEER program works closely with providers of cancer care in their population-based catchment areas to collect patient- and cancer-specific information. Because of its population-based nature, it is typically necessary to rely on medical records as the source of the demographic and other data. SEER-defined race and ethnic categories were utilized and labeled as follows: Non-Hispanic White (NHW), Non-Hispanic Black (NHB), Hispanic, Non-Hispanic American Indian/Alaska Native (AIAN), and Non-Hispanic Asian or Pacific Islander (API). Additional variables were extracted from SEER, and estimates of CRC-specific survival probabilities were obtained for combinations of these variables in the primary analyses: sex as assigned at birth (male or female), age at diagnosis (categorized into decades), year of diagnosis, grade (grades I-IV, or unknown), and Rural-Urban Continuum Code in 2003 (RUCC, coded as: Metro Counties, Non-Metro Counties [Metro-Adjacent], Non-Metro Counties [not Metro-Adjacent], and Alaska or Hawaii, or unknown).\nThe outcome of interest in this work was cause-specific survival for CRC, estimated at 1- and 5-years post diagnosis. We used SEER*Stat software (version 8.4.0)26 to calculate 1- and 5-year cause-specific survival probabilities of CRC using the “Incidence – SEER Research Plus Data, 12 Registries, Nov 2021 (1992-2019)” database while relying on the rules implemented in SEER*Stat for loss to follow-up. We obtained cause-specific survival probabilities and their standard errors within combinations of the characteristics of interest, ie, year of diagnosis, race and ethnicity, stage, sex, grade, and RUCC. No person-level data were used, nor were the SEER data linked to any other data sources. Using SEER*Stat 8.4.0, we formed tables of cause-specific survival probabilities and their standard errors according to all combinations of the factors of interest, with separate “Pages” defined for combinations of Year of diagnosis, age at diagnosis (in decades), and RUCC groups, and with rows defined by combinations of race/ethnicity, summary stage, grade, and sex. We used text processing approaches to form an analysis-ready data set which enumerated the numbers of individuals with a primary CRC diagnosis, and estimates of cause-specific survival probabilities and standard errors, within categories defined by all of the factors of interest in this study.\n[SUBTITLE] Statistical Analysis [SUBSECTION] We summarized the numbers and percentages of individuals with CRC diagnoses within the levels of the variables of interest. We used linear models to perform meta-regressions, weighting by the inverse of the squared standard errors, to estimate the degree to which the combinations of the variables of interest explained differences in the estimated cause-specific survival probabilities. When the SEER-estimated standard errors of survival probabilities were equal to zero, we approximated them with the square root of the variance of the binomial distribution, calculated after adding a value of .5 to both the numerator and denominator counts reported by SEER for the relevant group. We modelled the survival probabilities obtained at 1 year and 5 years post-diagnosis separately. The initial models included all possible interactions among race/ethnicity, stage, and year of diagnosis, while controlling for potential confounding by age at diagnosis (categorized into decades), sex, grade, and RUCC. We simplified these initial models subsequently modelling year-at-diagnosis trends with natural cubic spline basis functions, and selected the degree of smoothing that best explained the race by stage by year of diagnosis trends by minimizing Akaike’s Information Criterion (AIC) while adjusting for the other factors. We obtained smoothed trend estimates within groups defined by race and ethnicity, and by CRC stage at diagnosis, from a separate model for each of the two follow-up time periods. These final models were obtained by removing non-significant interactions in a hierarchical fashion. Analyses were performed using the tools available in SEER*Stat26 and SAS 9.4 (SAS Institute Inc, Cary, NC). Two-sided type I error was set at .05 for tests of significance.\nWe summarized the numbers and percentages of individuals with CRC diagnoses within the levels of the variables of interest. We used linear models to perform meta-regressions, weighting by the inverse of the squared standard errors, to estimate the degree to which the combinations of the variables of interest explained differences in the estimated cause-specific survival probabilities. When the SEER-estimated standard errors of survival probabilities were equal to zero, we approximated them with the square root of the variance of the binomial distribution, calculated after adding a value of .5 to both the numerator and denominator counts reported by SEER for the relevant group. We modelled the survival probabilities obtained at 1 year and 5 years post-diagnosis separately. The initial models included all possible interactions among race/ethnicity, stage, and year of diagnosis, while controlling for potential confounding by age at diagnosis (categorized into decades), sex, grade, and RUCC. We simplified these initial models subsequently modelling year-at-diagnosis trends with natural cubic spline basis functions, and selected the degree of smoothing that best explained the race by stage by year of diagnosis trends by minimizing Akaike’s Information Criterion (AIC) while adjusting for the other factors. We obtained smoothed trend estimates within groups defined by race and ethnicity, and by CRC stage at diagnosis, from a separate model for each of the two follow-up time periods. These final models were obtained by removing non-significant interactions in a hierarchical fashion. Analyses were performed using the tools available in SEER*Stat26 and SAS 9.4 (SAS Institute Inc, Cary, NC). Two-sided type I error was set at .05 for tests of significance.", "This study was based on a retrospective cohort of CRC patients, ascertained on a population level, with data captured as part of the National Cancer Institute’s Surveillance, Epidemiology and End Results (SEER) program. The SEER program collects cancer incidence and survival data from population-based cancer registries representing approximately 35% of the U.S. population.23 Data from the 12 SEER registries for the period 1992 to 2018 were used in this analysis. The 12 SEER registries cover Alaska, Connecticut, Atlanta, rural Georgia, San Francisco-Oakland, San Jose-Monterey, Hawaii, Iowa, Los Angeles, New Mexico, Seattle-Puget Sound and Utah.24 This project, Study ID: 21-102, was reviewed by the University of New Mexico Health Sciences Institutional Review Board, and was granted exemption on 31 March 2021 according to Category 4: Secondary research on data or specimens (no consent required). This report conforms to RECORD guidelines for SEER-based studies.25 This study was supported by funding from the National Cancer Institute; the funders played no other role in this work. Researchers desiring additional details about the data and programs used to carry out this work may obtain programming code from the corresponding author.", "We used SEER*Stat software (version 8.4.0)26 as the data source for this study. We used the “Incidence––SEER Research Plus Data, 12 Registries, Nov 2021 Sub (1992-2019)” database. We included a consecutive series of all individuals who received their first primary CRC diagnosis with malignant behavior from 1992 through 2018. We excluded “All Death Certificate Only and Autopsy Only” and “Alive with No Survival Time” via checkboxes in SEER*Stat. We also excluded CRC cases without a record for summary stage at diagnosis, as well as those with unknown age at diagnosis. We included all CRC cases that originated from the cecum, ascending colon, hepatic flexure of colon, transverse colon, splenic flexure of colon, descending colon, sigmoid colon, overlapping lesion of colon, colon not otherwise specified, rectosigmoid junction, and rectum. We classified CRC cases as localized, regional and distant stage using SEER’s “Combined Summary Stage (2004+)” classifications, supplemented with values from the “Historic Stage A (1973-2015)” variable when necessary. In-situ cancers and cases that were un-staged/unknown were excluded. These rules, as applied in SEER*Stat 8.0.4, defined the number of individuals included in this study.\nThe SEER program works closely with providers of cancer care in their population-based catchment areas to collect patient- and cancer-specific information. Because of its population-based nature, it is typically necessary to rely on medical records as the source of the demographic and other data. SEER-defined race and ethnic categories were utilized and labeled as follows: Non-Hispanic White (NHW), Non-Hispanic Black (NHB), Hispanic, Non-Hispanic American Indian/Alaska Native (AIAN), and Non-Hispanic Asian or Pacific Islander (API). Additional variables were extracted from SEER, and estimates of CRC-specific survival probabilities were obtained for combinations of these variables in the primary analyses: sex as assigned at birth (male or female), age at diagnosis (categorized into decades), year of diagnosis, grade (grades I-IV, or unknown), and Rural-Urban Continuum Code in 2003 (RUCC, coded as: Metro Counties, Non-Metro Counties [Metro-Adjacent], Non-Metro Counties [not Metro-Adjacent], and Alaska or Hawaii, or unknown).\nThe outcome of interest in this work was cause-specific survival for CRC, estimated at 1- and 5-years post diagnosis. We used SEER*Stat software (version 8.4.0)26 to calculate 1- and 5-year cause-specific survival probabilities of CRC using the “Incidence – SEER Research Plus Data, 12 Registries, Nov 2021 (1992-2019)” database while relying on the rules implemented in SEER*Stat for loss to follow-up. We obtained cause-specific survival probabilities and their standard errors within combinations of the characteristics of interest, ie, year of diagnosis, race and ethnicity, stage, sex, grade, and RUCC. No person-level data were used, nor were the SEER data linked to any other data sources. Using SEER*Stat 8.4.0, we formed tables of cause-specific survival probabilities and their standard errors according to all combinations of the factors of interest, with separate “Pages” defined for combinations of Year of diagnosis, age at diagnosis (in decades), and RUCC groups, and with rows defined by combinations of race/ethnicity, summary stage, grade, and sex. We used text processing approaches to form an analysis-ready data set which enumerated the numbers of individuals with a primary CRC diagnosis, and estimates of cause-specific survival probabilities and standard errors, within categories defined by all of the factors of interest in this study.", "We summarized the numbers and percentages of individuals with CRC diagnoses within the levels of the variables of interest. We used linear models to perform meta-regressions, weighting by the inverse of the squared standard errors, to estimate the degree to which the combinations of the variables of interest explained differences in the estimated cause-specific survival probabilities. When the SEER-estimated standard errors of survival probabilities were equal to zero, we approximated them with the square root of the variance of the binomial distribution, calculated after adding a value of .5 to both the numerator and denominator counts reported by SEER for the relevant group. We modelled the survival probabilities obtained at 1 year and 5 years post-diagnosis separately. The initial models included all possible interactions among race/ethnicity, stage, and year of diagnosis, while controlling for potential confounding by age at diagnosis (categorized into decades), sex, grade, and RUCC. We simplified these initial models subsequently modelling year-at-diagnosis trends with natural cubic spline basis functions, and selected the degree of smoothing that best explained the race by stage by year of diagnosis trends by minimizing Akaike’s Information Criterion (AIC) while adjusting for the other factors. We obtained smoothed trend estimates within groups defined by race and ethnicity, and by CRC stage at diagnosis, from a separate model for each of the two follow-up time periods. These final models were obtained by removing non-significant interactions in a hierarchical fashion. Analyses were performed using the tools available in SEER*Stat26 and SAS 9.4 (SAS Institute Inc, Cary, NC). Two-sided type I error was set at .05 for tests of significance.", "This study was based on the 1975-2019 SEER Research Plus Data (November 2021 Submission), which contains information on 425 520 CRC diagnoses from 1992 through 2018. Excluding those individuals without a declared race or ethnicity, and with unknown stage at diagnosis, removed 1372 (.3%) and 24 928 (5.9%) individuals, respectively. This report is based on analysed data from 399 220 individual CRC diagnoses with known race/ethnicity and stage at diagnosis from 1992 through 2018, or 93.8% of all CRC diagnoses from this time period. Table 1 contains tabulations of the numbers of individuals according to levels of the variables of interest, overall and within race and ethnic groups. There was at least 1 year of follow-up for 318 443 (79.8%) persons with CRC eligible at baseline.Table 1.Characteristics of Patients With Colorectal Cancer From 1992 to 2018, by Racial and Ethnic Groups.AIANAPIHispanicNHBNHWTotalN%n%n%n%n%n%Total39311.045 86911.541 94710.534 1648.6273 30968.5399 220100.0SexMale197950.324 40053.222 21653.016 67448.8139 89751.2205 16651.4Female195249.721 46946.819 73147.017 49051.2133 41248.8194 05448.6Age<40 years2145.415723.424155.812173.659992.211 4172.940-49 years45911.740708.9455310.9340110.016 4666.028 9497.350-59 years91823.4924120.1930822.2781322.941 01815.068 29817.160-69 years104226.511 40424.910 65625.4916126.860 83922.393 10223.370-79 years86522.011 40724.9913021.8777222.778 10828.6107 28226.980+ years43311.0817517.8588514.0480014.070 87925.990 17222.6Summary stageLocalized152038.719 18241.816 57539.513 49139.5115 25442.2166 02241.6Regional149137.917 83338.916 15938.512 00135.1103 89638.0151 38037.9Distant92023.4885419.3921322.0867225.454 15919.881 81820.5Year of diagnosis1992-953488.9458310.036238.6407411.943 15115.855 77914.01996-9945411.6558312.2451410.8449113.244 92516.559 96715.02000-0348112.2671014.6527712.6495914.544 34316.261 77015.42004-0758714.9711615.5636815.2543915.940 99215.060 50215.22008-1165716.7781117.0722017.2560716.438 19213.959 48714.92012-1577819.8789717.2799719.0540615.835 53613.057 61414.42016-1862615.9616913.4694816.6418812.326 1709.544 10111.0GradeI3759.534977.6433510.331539.223 9368.835 2968.8II226057.527 75760.523 47756.019 37556.7155 56256.9228 43157.2III44811.4626913.7575313.7434412.746 96117.263 77516.0IV561.44451.05471.34151.243941.658571.5Unknown79220.1790117.2783518.7687720.142 45615.565 86116.5Rural-urban continuum codeMetro counties150038.240 55788.438 81492.533 18697.1228 17983.5342 23685.7Non-metro counties (metro-adjacent)3839.71120.213013.15401.623 5848.625 9206.5Non-metro counties (not metro-adjacent)39610.116543.616914.04101.220 4267.524 5776.2Alaska or Hawaii164441.800.050.000.000.016490.4Unknown80.235467.71360.3280.111200.448381.2\nCharacteristics of Patients With Colorectal Cancer From 1992 to 2018, by Racial and Ethnic Groups.\n[SUBTITLE] One- and Five-Year Survival: All Persons With CRC [SUBSECTION] Adjusted 1- and 5-year cause-specific survival probabilities following CRC diagnosis by stage and year of diagnosis are shown in Figure 1. For those diagnosed with local stage CRC, the estimate of linear trend in 1-year survival probabilities was negligible, at −.01% ([95% Confidence Interval (CI)] −.03%-.01%) per year, with an average 1-year survival probability of 96.2% (93.8%-98.6%). Those diagnosed with regional stage CRC displayed a small improvement over time in 1-year survival probabilities, .06% (.001%-.12%) per year. Their average 1-year survival probability was 93.0% (90.5%-95.5%). For those diagnosed with distant stage CRC, there was a significant improvement in 1-year survival probabilities over time (P < .001), but there was evidence of departure from a linear trend (P = .04). The slope of the trend line suggested an improvement in survival probability of 1.0% (.8%-1.2%) per year in 2005, with instantaneous slopes suggesting greater gains, by .05% (.01%-.09%), for each year prior to 2005 and slower gains by that same amount for each year following 2005. Their average 1-year survival probability was 52.8% (48.1%-57.5%).Figure 1.Trends in 1- and 5-year adjusted cause-specific survival probabilities for individuals diagnosed at different stages of colorectal cancer. Shaded bands reflect 95% prediction intervals for the year-specific survival probabilities.\nTrends in 1- and 5-year adjusted cause-specific survival probabilities for individuals diagnosed at different stages of colorectal cancer. Shaded bands reflect 95% prediction intervals for the year-specific survival probabilities.\nThose diagnosed with local stage CRC experienced improvements in 5-year cause-specific survival, with an average improvement of .16% (.10%-.21%) per year. Their average 5-year survival probability was 90.9% (87.9%-93.8%). Those diagnosed with regional stage CRC displayed significant improvement in 5-year survival probabilities over time (P < .001), but there was evidence of significant departure from a linear trend (P = .004), such that the slope of the trend line showed an improvement of .55% (.41%-.69%) per year in 2003, with instantaneous slopes suggested greater gains, by .08% (.04-.12%), for each year prior to 2003 and slower gains by that same amount for each year following 2003. Their average 5-year survival probability was 70.3% (66.5%-74.0%). For those diagnosed with distant stage CRC, there was a significant but small improvement over time in 5-year survival probabilities, .15% (.5%-.25%) per year. Their average 5-year survival probability was 8.1% (4.4%-11.8%).\nAdjusted 1- and 5-year cause-specific survival probabilities following CRC diagnosis by stage and year of diagnosis are shown in Figure 1. For those diagnosed with local stage CRC, the estimate of linear trend in 1-year survival probabilities was negligible, at −.01% ([95% Confidence Interval (CI)] −.03%-.01%) per year, with an average 1-year survival probability of 96.2% (93.8%-98.6%). Those diagnosed with regional stage CRC displayed a small improvement over time in 1-year survival probabilities, .06% (.001%-.12%) per year. Their average 1-year survival probability was 93.0% (90.5%-95.5%). For those diagnosed with distant stage CRC, there was a significant improvement in 1-year survival probabilities over time (P < .001), but there was evidence of departure from a linear trend (P = .04). The slope of the trend line suggested an improvement in survival probability of 1.0% (.8%-1.2%) per year in 2005, with instantaneous slopes suggesting greater gains, by .05% (.01%-.09%), for each year prior to 2005 and slower gains by that same amount for each year following 2005. Their average 1-year survival probability was 52.8% (48.1%-57.5%).Figure 1.Trends in 1- and 5-year adjusted cause-specific survival probabilities for individuals diagnosed at different stages of colorectal cancer. Shaded bands reflect 95% prediction intervals for the year-specific survival probabilities.\nTrends in 1- and 5-year adjusted cause-specific survival probabilities for individuals diagnosed at different stages of colorectal cancer. Shaded bands reflect 95% prediction intervals for the year-specific survival probabilities.\nThose diagnosed with local stage CRC experienced improvements in 5-year cause-specific survival, with an average improvement of .16% (.10%-.21%) per year. Their average 5-year survival probability was 90.9% (87.9%-93.8%). Those diagnosed with regional stage CRC displayed significant improvement in 5-year survival probabilities over time (P < .001), but there was evidence of significant departure from a linear trend (P = .004), such that the slope of the trend line showed an improvement of .55% (.41%-.69%) per year in 2003, with instantaneous slopes suggested greater gains, by .08% (.04-.12%), for each year prior to 2003 and slower gains by that same amount for each year following 2003. Their average 5-year survival probability was 70.3% (66.5%-74.0%). For those diagnosed with distant stage CRC, there was a significant but small improvement over time in 5-year survival probabilities, .15% (.5%-.25%) per year. Their average 5-year survival probability was 8.1% (4.4%-11.8%).\n[SUBTITLE] One-Year Survival by Race and Ethnicity [SUBSECTION] Estimates of adjusted 1-year cause-specific survival probabilities following diagnosis are shown for all race/ethnicity by stage combinations in Tables 2 and 3. Trends in 1-year survival for those of different races/ethnicities who were diagnosed at different stages of CRC differed significantly (P < .001, see Figure 2). For those diagnosed with localized stage CRC, 1-year survival probabilities were consistently high for individuals of all race and ethnic groups. AIAN persons experienced lower 1-year survival probabilities than those from other race and ethnic groups over the study period. Although due to lack of precision in the estimates, the difference was only significantly different when compared to API persons; average 1-year survival probabilities were 1.5 (.5-2.5) percentage points lower for AIAN than for API persons.Table 2.Unadjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN1992100 (84.6-100)91.6 (75.4-100)27.0 (5.6-48.4)72.9 (62.6-83.2)AIAN199696.9 (81.8-100)95.7 (77.9-100)41.8 (23.5-60.1)78.1 (68.2-88.0)AIAN200098.7 (83.6-100)83.2 (65.9-100)43.0 (20.0-66.0)75.0 (64.1-85.8)AIAN200496.7 (85.6-100)96.4 (84.8-100)51.8 (35.8-67.7)81.6 (74.1-89.2)AIAN2008100 (86.4-100)95.5 (83.0-100)53.5 (35.5-71.5)83.0 (74.4-91.6)AIAN2012100 (88.9-100)91.6 (79.1-100)52.7 (38.0-67.4)81.4 (74.0-88.8)AIAN201696.0 (85.5-100)95.9 (85.6-100)46.5 (33.4-59.6)79.5 (72.9-86.0)AIAN2018100 (90.7-100)99.3 (88.9-100)83.5 (67.2-99.8)94.3 (87.1-100)API199299.5 (96.2-100)94.6 (90.8-98.3)46.5 (38.4-54.6)80.2 (77.0-83.3)API199699.4 (97.1-100)95.8 (92.6-98.9)56.5 (48.7-64.3)83.9 (81.0-86.8)API200099.2 (97.0-100)97.9 (95.8-99.9)52.8 (45.0-60.6)83.3 (80.5-86.1)API200499.6 (97.9-100)97.3 (95.1-99.5)68.6 (62.7-74.4)88.5 (86.3-90.6)API200899.7 (98.3-100)98.4 (96.5-100)59.5 (53.9-65.0)85.9 (83.9-87.9)API201299.4 (97.9-100)96.9 (94.7-99.2)55.8 (49.4-62.3)84.1 (81.7-86.4)API201699.1 (97.4-100)98.4 (96.7-100)57.6 (51.8-63.3)85.0 (82.9-87.1)API201899.6 (98.5-100)97.8 (96.0-99.6)69.1 (62.7-75.5)88.8 (86.6-91.1)Hispanic199297.0 (93.0-100)96.7 (93.3-100)55.6 (46.2-64.9)83.1 (79.5-86.7)Hispanic199698.8 (95.8-100)96.1 (92.7-99.5)43.4 (35.7-51.1)79.4 (76.4-82.4)Hispanic200099.2 (96.6-100)96.3 (93.3-99.4)47.0 (39.8-54.2)80.8 (78.1-83.6)Hispanic200498.9 (96.5-100)95.8 (92.9-98.7)57.8 (50.8-64.8)84.2 (81.5-86.8)Hispanic200899.1 (97.3-100)96.2 (93.5-98.8)60.7 (54.4-67.1)85.3 (83.0-87.7)Hispanic201299.2 (97.4-100)95.8 (93.3-98.2)71.6 (66.6-76.7)88.9 (86.9-90.8)Hispanic201698.9 (97.1-100)98.2 (96.6-99.7)69.0 (64.0-74.1)88.7 (86.9-90.6)Hispanic201899.6 (98.6-100)97.8 (96.1-99.5)58.8 (52.5-65.0)85.4 (83.2-87.6)NHB199298.0 (94.7-100)93.7 (89.6-97.8)38.5 (30.0-47.0)76.7 (73.4-80.1)NHB199698.2 (94.7-100)96.3 (93-99.6)30.1 (23.1-37.2)74.9 (72.0-77.7)NHB200097.3 (93.8-100)93.2 (89.4-97.0)34.5 (27.3-41.7)75.0 (72.0-77.9)NHB200498.4 (95.6-100)97.0 (94.3-99.7)37.0 (30.7-43.3)77.5 (75.0-79.9)NHB200898.6 (96.2-100)93.3 (89.4-97.2)51.4 (44.7-58.1)81.1 (78.4-83.8)NHB201299.5 (97.2-100)93.6 (90.0-97.1)68.5 (62.1-74.8)87.2 (84.7-89.7)NHB201699.5 (97.2-100)96.0 (93.0-99.0)53.1 (47.2-58.9)82.9 (80.5-85.2)NHB201899.3 (97.5-100)97.0 (94.2-99.8)57.1 (49.6-64.6)84.5 (81.7-87.2)NHW199298.6 (97.9-99.2)94.6 (93.5-95.7)38.0 (35.0-40.9)77.0 (76.0-78.1)NHW199698.9 (98.3-99.5)96.1 (95.1-97.0)36.7 (33.8-39.5)77.2 (76.2-78.2)NHW200099.0 (98.5-99.6)94.3 (93.2-95.4)40.9 (37.9-43.9)78.1 (77.0-79.1)NHW200498.9 (98.3-99.5)96.1 (95.1-97.1)52.6 (49.7-55.4)82.5 (81.5-83.6)NHW200899.2 (98.6-99.7)97.0 (96.1-98.0)58.6 (55.6-61.7)85.0 (83.9-86.0)NHW201298.9 (98.2-99.6)97.2 (96.2-98.2)55.2 (52.3-58.0)83.8 (82.7-84.8)NHW201699.1 (98.5-99.8)96.9 (95.9-97.9)60.7 (57.9-63.5)85.6 (84.6-86.6)NHW201898.7 (98.0-99.4)97.5 (96.7-98.4)59.0 (55.7-62.3)85.1 (83.9-86.3)Table 3.Adjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199297.5 (83.7-100)89.8 (75.3-100)25.4 (6.2-44.6)70.9 (61.6-80.2)AIAN199695.6 (82.0-100)93.2 (77.3-100)41.9 (25.5-58.3)76.9 (68.0-85.8)AIAN200097.0 (83.5-100)82.5 (67.0-97.9)42.0 (21.4-62.6)73.8 (64.1-83.5)AIAN200495.3 (85.3-100)94.2 (83.8-100)50.4 (36.1-64.7)80.0 (73.2-86.8)AIAN200897.5 (85.3-100)92.4 (81.2-100)51.6 (35.5-67.7)80.5 (72.8-88.2)AIAN201297.5 (87.5-100)89.9 (78.7-100)50.2 (37.0-63.3)79.2 (72.5-85.9)AIAN201693.4 (84.0-100)94.1 (84.8-100)45.0 (33.3-56.8)77.5 (71.6-83.4)AIAN201896.7 (87.8-100)96.9 (87.6-100)64.2 (51.1-77.5)85.9 (79.7-91.7)API199297.5 (94.5-100)92.3 (88.9-95.7)45.8 (38.6-53.1)78.6 (75.7-81.4)API199697.4 (95.2-99.6)93.4 (90.5-96.3)55.6 (48.5-62.6)82.1 (79.4-84.8)API200097.4 (95.3-99.5)95.9 (93.9-97.9)53.1 (46.1-60.1)82.1 (79.6-84.7)API200497.8 (96.2-99.4)94.6 (92.5-96.7)67.1 (61.8-72.4)86.5 (84.5-88.5)API200897.5 (96.0-98.9)95.6 (93.7-97.4)59.4 (54.4-64.4)84.2 (82.2-86.1)API201296.8 (95.3-98.3)94.6 (92.5-96.7)56.1 (50.3-61.9)82.5 (80.3-84.7)API201696.4 (94.8-98.0)95.0 (93.4-96.6)57.8 (52.6-63.0)83.1 (81.1-85.0)API201897.9 (96.5-99.0)96.1 (94.5-97.9)63.8 (58.3-69.3)86.0 (83.9-88.0)Hispanic199294.7 (91.1-98.4)94.4 (91.3-97.5)54.3 (45.9-62.7)81.2 (77.9-84.4)Hispanic199696.8 (93.9-99.6)94.2 (91.0-97.3)42.9 (36.0-49.9)77.9 (75.2-80.7)Hispanic200097.0 (94.6-99.4)93.4 (90.6-96.2)46.6 (40.1-53.0)79.0 (76.4-81.5)Hispanic200497.2 (95.0-99.4)93.3 (90.6-96.0)57.1 (50.8-63.4)82.5 (80.1-85.0)Hispanic200896.2 (94.5-97.9)92.8 (90.4-95.3)59.8 (54.1-65.5)83.0 (80.8-85.2)Hispanic201296.0 (94.3-97.8)92.9 (90.6-95.2)69.3 (64.8-73.9)86.1 (84.2-88.0)Hispanic201694.8 (93.0-96.5)94.1 (92.5-95.6)67.3 (62.8-71.9)85.4 (83.6-87.2)Hispanic201896.8 (95.3-98.3)95.3 (93.7-96.9)63.4 (58.3-68.6)85.2 (83.2-87.2)NHB199295.7 (92.6-98.7)91.1 (87.3-94.8)37.8 (30.2-45.4)74.8 (71.8-77.9)NHB199696.6 (93.4-99.9)93.8 (90.7-96.8)30.8 (24.4-37.1)73.7 (71.1-76.3)NHB200095.6 (92.5-98.8)90.5 (87.1-94.0)35.7 (29.2-42.2)74.0 (71.2-76.7)NHB200495.9 (93.3-98.5)94.0 (91.4-96.5)38.9 (33.2-44.6)76.3 (73.9-78.6)NHB200896.1 (93.9-98.3)90.3 (86.8-93.9)51.2 (45.1-57.2)79.2 (76.7-81.7)NHB201296.6 (94.4-98.7)90.6 (87.4-93.8)66.7 (60.9-72.4)84.6 (82.2-87.0)NHB201696.0 (93.9-98.2)92.3 (89.6-95.1)53.0 (47.7-58.3)80.5 (78.3-82.6)NHB201897.5 (95.6-99.1)94.3 (91.7-97.1)55.2 (49.2-61.3)82.4 (80.0-84.8)NHW199296.8 (95.9-97.7)92.8 (91.6-94.0)39.2 (36.5-42.0)76.3 (75.1-77.4)NHW199696.7 (95.9-97.6)93.1 (92.1-94.2)38.4 (35.8-41.1)76.1 (75.0-77.2)NHW200096.3 (95.5-97.2)92.4 (91.2-93.6)42.4 (39.6-45.1)77.0 (75.9-78.2)NHW200496.5 (95.6-97.3)93.5 (92.4-94.7)53.5 (50.8-56.1)81.2 (80.0-82.3)NHW200896.4 (95.6-97.3)93.8 (92.7-94.9)58.8 (56.0-61.6)83.0 (81.8-84.2)NHW201295.9 (95.0-96.8)93.9 (92.8-95.0)55.7 (53.0-58.3)81.8 (80.7-83.0)NHW201695.2 (94.3-96.0)93.5 (92.4-94.6)60.4 (57.8-63.0)83.0 (81.9-84.2)NHW201896.7 (95.8-97.6)95.0 (93.95-96.1)60.4 (57.6-63.3)84.0 (82.9-85.3)Figure 2.Trends in 1-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2018.\nUnadjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nAdjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nTrends in 1-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2018.\nFor those diagnosed with regional stage CRC, trends in adjusted 1-year cause-specific survival probabilities similarly reflect little change over time, with the possible exception of an imprecisely-estimated positive trend over time among AIAN persons. Although there was little evidence of significant trends over time, there was a clear gradient in the average survival probabilities among those from different race and ethnic groups. AIAN persons had the lowest average survival probability (90.2%, 88.2%-92.2), followed by NHB, NHW, Hispanic, and API persons (93.2%, 92.6%-93.8%; 93.3%, 93.1%-93.5%; 94.2%, 93.8%-94.6%; and 94.8%, 94.4%-95.2%, respectively). All of these estimates were significantly different (all P < .01), except for the comparison between NHB and NHW persons (P = .83).\nThose diagnosed with distant stage CRC experienced the biggest improvements in 1-year cause-specific survival probabilities over time. An increasing trend in 1-year survival probabilities is apparent for persons of all race and ethnic groups. For AIAN and NHB persons, the two groups with lowest initial 1-year survival probabilities, improvements continued at a consistent rate over time. For API, Hispanic, and NHW persons, there appears to be a slowing of the rate of improvement in the second half of the study period. The adjusted time-averaged survival probabilities were low: 46.6% (42.3%-50.9%) for NHB, 48.3% (37.9%-58.7%) for AIAN, 50.5% (48.5%-52.5%) for NHW, 58.1% (54.2%-62.0%) for Hispanic, and 58.3% (54.2%-62.4%) for API persons. All pairwise comparisons among these groups were statistically significant (all P < .01), except for differences between the two groups with the lowest (AIAN vs NHB, P = .87), and between the two groups with the highest (API vs Hispanic, P = .69), average adjusted 1-year cause-specific survival probabilities.\nEstimates of adjusted 1-year cause-specific survival probabilities following diagnosis are shown for all race/ethnicity by stage combinations in Tables 2 and 3. Trends in 1-year survival for those of different races/ethnicities who were diagnosed at different stages of CRC differed significantly (P < .001, see Figure 2). For those diagnosed with localized stage CRC, 1-year survival probabilities were consistently high for individuals of all race and ethnic groups. AIAN persons experienced lower 1-year survival probabilities than those from other race and ethnic groups over the study period. Although due to lack of precision in the estimates, the difference was only significantly different when compared to API persons; average 1-year survival probabilities were 1.5 (.5-2.5) percentage points lower for AIAN than for API persons.Table 2.Unadjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN1992100 (84.6-100)91.6 (75.4-100)27.0 (5.6-48.4)72.9 (62.6-83.2)AIAN199696.9 (81.8-100)95.7 (77.9-100)41.8 (23.5-60.1)78.1 (68.2-88.0)AIAN200098.7 (83.6-100)83.2 (65.9-100)43.0 (20.0-66.0)75.0 (64.1-85.8)AIAN200496.7 (85.6-100)96.4 (84.8-100)51.8 (35.8-67.7)81.6 (74.1-89.2)AIAN2008100 (86.4-100)95.5 (83.0-100)53.5 (35.5-71.5)83.0 (74.4-91.6)AIAN2012100 (88.9-100)91.6 (79.1-100)52.7 (38.0-67.4)81.4 (74.0-88.8)AIAN201696.0 (85.5-100)95.9 (85.6-100)46.5 (33.4-59.6)79.5 (72.9-86.0)AIAN2018100 (90.7-100)99.3 (88.9-100)83.5 (67.2-99.8)94.3 (87.1-100)API199299.5 (96.2-100)94.6 (90.8-98.3)46.5 (38.4-54.6)80.2 (77.0-83.3)API199699.4 (97.1-100)95.8 (92.6-98.9)56.5 (48.7-64.3)83.9 (81.0-86.8)API200099.2 (97.0-100)97.9 (95.8-99.9)52.8 (45.0-60.6)83.3 (80.5-86.1)API200499.6 (97.9-100)97.3 (95.1-99.5)68.6 (62.7-74.4)88.5 (86.3-90.6)API200899.7 (98.3-100)98.4 (96.5-100)59.5 (53.9-65.0)85.9 (83.9-87.9)API201299.4 (97.9-100)96.9 (94.7-99.2)55.8 (49.4-62.3)84.1 (81.7-86.4)API201699.1 (97.4-100)98.4 (96.7-100)57.6 (51.8-63.3)85.0 (82.9-87.1)API201899.6 (98.5-100)97.8 (96.0-99.6)69.1 (62.7-75.5)88.8 (86.6-91.1)Hispanic199297.0 (93.0-100)96.7 (93.3-100)55.6 (46.2-64.9)83.1 (79.5-86.7)Hispanic199698.8 (95.8-100)96.1 (92.7-99.5)43.4 (35.7-51.1)79.4 (76.4-82.4)Hispanic200099.2 (96.6-100)96.3 (93.3-99.4)47.0 (39.8-54.2)80.8 (78.1-83.6)Hispanic200498.9 (96.5-100)95.8 (92.9-98.7)57.8 (50.8-64.8)84.2 (81.5-86.8)Hispanic200899.1 (97.3-100)96.2 (93.5-98.8)60.7 (54.4-67.1)85.3 (83.0-87.7)Hispanic201299.2 (97.4-100)95.8 (93.3-98.2)71.6 (66.6-76.7)88.9 (86.9-90.8)Hispanic201698.9 (97.1-100)98.2 (96.6-99.7)69.0 (64.0-74.1)88.7 (86.9-90.6)Hispanic201899.6 (98.6-100)97.8 (96.1-99.5)58.8 (52.5-65.0)85.4 (83.2-87.6)NHB199298.0 (94.7-100)93.7 (89.6-97.8)38.5 (30.0-47.0)76.7 (73.4-80.1)NHB199698.2 (94.7-100)96.3 (93-99.6)30.1 (23.1-37.2)74.9 (72.0-77.7)NHB200097.3 (93.8-100)93.2 (89.4-97.0)34.5 (27.3-41.7)75.0 (72.0-77.9)NHB200498.4 (95.6-100)97.0 (94.3-99.7)37.0 (30.7-43.3)77.5 (75.0-79.9)NHB200898.6 (96.2-100)93.3 (89.4-97.2)51.4 (44.7-58.1)81.1 (78.4-83.8)NHB201299.5 (97.2-100)93.6 (90.0-97.1)68.5 (62.1-74.8)87.2 (84.7-89.7)NHB201699.5 (97.2-100)96.0 (93.0-99.0)53.1 (47.2-58.9)82.9 (80.5-85.2)NHB201899.3 (97.5-100)97.0 (94.2-99.8)57.1 (49.6-64.6)84.5 (81.7-87.2)NHW199298.6 (97.9-99.2)94.6 (93.5-95.7)38.0 (35.0-40.9)77.0 (76.0-78.1)NHW199698.9 (98.3-99.5)96.1 (95.1-97.0)36.7 (33.8-39.5)77.2 (76.2-78.2)NHW200099.0 (98.5-99.6)94.3 (93.2-95.4)40.9 (37.9-43.9)78.1 (77.0-79.1)NHW200498.9 (98.3-99.5)96.1 (95.1-97.1)52.6 (49.7-55.4)82.5 (81.5-83.6)NHW200899.2 (98.6-99.7)97.0 (96.1-98.0)58.6 (55.6-61.7)85.0 (83.9-86.0)NHW201298.9 (98.2-99.6)97.2 (96.2-98.2)55.2 (52.3-58.0)83.8 (82.7-84.8)NHW201699.1 (98.5-99.8)96.9 (95.9-97.9)60.7 (57.9-63.5)85.6 (84.6-86.6)NHW201898.7 (98.0-99.4)97.5 (96.7-98.4)59.0 (55.7-62.3)85.1 (83.9-86.3)Table 3.Adjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199297.5 (83.7-100)89.8 (75.3-100)25.4 (6.2-44.6)70.9 (61.6-80.2)AIAN199695.6 (82.0-100)93.2 (77.3-100)41.9 (25.5-58.3)76.9 (68.0-85.8)AIAN200097.0 (83.5-100)82.5 (67.0-97.9)42.0 (21.4-62.6)73.8 (64.1-83.5)AIAN200495.3 (85.3-100)94.2 (83.8-100)50.4 (36.1-64.7)80.0 (73.2-86.8)AIAN200897.5 (85.3-100)92.4 (81.2-100)51.6 (35.5-67.7)80.5 (72.8-88.2)AIAN201297.5 (87.5-100)89.9 (78.7-100)50.2 (37.0-63.3)79.2 (72.5-85.9)AIAN201693.4 (84.0-100)94.1 (84.8-100)45.0 (33.3-56.8)77.5 (71.6-83.4)AIAN201896.7 (87.8-100)96.9 (87.6-100)64.2 (51.1-77.5)85.9 (79.7-91.7)API199297.5 (94.5-100)92.3 (88.9-95.7)45.8 (38.6-53.1)78.6 (75.7-81.4)API199697.4 (95.2-99.6)93.4 (90.5-96.3)55.6 (48.5-62.6)82.1 (79.4-84.8)API200097.4 (95.3-99.5)95.9 (93.9-97.9)53.1 (46.1-60.1)82.1 (79.6-84.7)API200497.8 (96.2-99.4)94.6 (92.5-96.7)67.1 (61.8-72.4)86.5 (84.5-88.5)API200897.5 (96.0-98.9)95.6 (93.7-97.4)59.4 (54.4-64.4)84.2 (82.2-86.1)API201296.8 (95.3-98.3)94.6 (92.5-96.7)56.1 (50.3-61.9)82.5 (80.3-84.7)API201696.4 (94.8-98.0)95.0 (93.4-96.6)57.8 (52.6-63.0)83.1 (81.1-85.0)API201897.9 (96.5-99.0)96.1 (94.5-97.9)63.8 (58.3-69.3)86.0 (83.9-88.0)Hispanic199294.7 (91.1-98.4)94.4 (91.3-97.5)54.3 (45.9-62.7)81.2 (77.9-84.4)Hispanic199696.8 (93.9-99.6)94.2 (91.0-97.3)42.9 (36.0-49.9)77.9 (75.2-80.7)Hispanic200097.0 (94.6-99.4)93.4 (90.6-96.2)46.6 (40.1-53.0)79.0 (76.4-81.5)Hispanic200497.2 (95.0-99.4)93.3 (90.6-96.0)57.1 (50.8-63.4)82.5 (80.1-85.0)Hispanic200896.2 (94.5-97.9)92.8 (90.4-95.3)59.8 (54.1-65.5)83.0 (80.8-85.2)Hispanic201296.0 (94.3-97.8)92.9 (90.6-95.2)69.3 (64.8-73.9)86.1 (84.2-88.0)Hispanic201694.8 (93.0-96.5)94.1 (92.5-95.6)67.3 (62.8-71.9)85.4 (83.6-87.2)Hispanic201896.8 (95.3-98.3)95.3 (93.7-96.9)63.4 (58.3-68.6)85.2 (83.2-87.2)NHB199295.7 (92.6-98.7)91.1 (87.3-94.8)37.8 (30.2-45.4)74.8 (71.8-77.9)NHB199696.6 (93.4-99.9)93.8 (90.7-96.8)30.8 (24.4-37.1)73.7 (71.1-76.3)NHB200095.6 (92.5-98.8)90.5 (87.1-94.0)35.7 (29.2-42.2)74.0 (71.2-76.7)NHB200495.9 (93.3-98.5)94.0 (91.4-96.5)38.9 (33.2-44.6)76.3 (73.9-78.6)NHB200896.1 (93.9-98.3)90.3 (86.8-93.9)51.2 (45.1-57.2)79.2 (76.7-81.7)NHB201296.6 (94.4-98.7)90.6 (87.4-93.8)66.7 (60.9-72.4)84.6 (82.2-87.0)NHB201696.0 (93.9-98.2)92.3 (89.6-95.1)53.0 (47.7-58.3)80.5 (78.3-82.6)NHB201897.5 (95.6-99.1)94.3 (91.7-97.1)55.2 (49.2-61.3)82.4 (80.0-84.8)NHW199296.8 (95.9-97.7)92.8 (91.6-94.0)39.2 (36.5-42.0)76.3 (75.1-77.4)NHW199696.7 (95.9-97.6)93.1 (92.1-94.2)38.4 (35.8-41.1)76.1 (75.0-77.2)NHW200096.3 (95.5-97.2)92.4 (91.2-93.6)42.4 (39.6-45.1)77.0 (75.9-78.2)NHW200496.5 (95.6-97.3)93.5 (92.4-94.7)53.5 (50.8-56.1)81.2 (80.0-82.3)NHW200896.4 (95.6-97.3)93.8 (92.7-94.9)58.8 (56.0-61.6)83.0 (81.8-84.2)NHW201295.9 (95.0-96.8)93.9 (92.8-95.0)55.7 (53.0-58.3)81.8 (80.7-83.0)NHW201695.2 (94.3-96.0)93.5 (92.4-94.6)60.4 (57.8-63.0)83.0 (81.9-84.2)NHW201896.7 (95.8-97.6)95.0 (93.95-96.1)60.4 (57.6-63.3)84.0 (82.9-85.3)Figure 2.Trends in 1-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2018.\nUnadjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nAdjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nTrends in 1-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2018.\nFor those diagnosed with regional stage CRC, trends in adjusted 1-year cause-specific survival probabilities similarly reflect little change over time, with the possible exception of an imprecisely-estimated positive trend over time among AIAN persons. Although there was little evidence of significant trends over time, there was a clear gradient in the average survival probabilities among those from different race and ethnic groups. AIAN persons had the lowest average survival probability (90.2%, 88.2%-92.2), followed by NHB, NHW, Hispanic, and API persons (93.2%, 92.6%-93.8%; 93.3%, 93.1%-93.5%; 94.2%, 93.8%-94.6%; and 94.8%, 94.4%-95.2%, respectively). All of these estimates were significantly different (all P < .01), except for the comparison between NHB and NHW persons (P = .83).\nThose diagnosed with distant stage CRC experienced the biggest improvements in 1-year cause-specific survival probabilities over time. An increasing trend in 1-year survival probabilities is apparent for persons of all race and ethnic groups. For AIAN and NHB persons, the two groups with lowest initial 1-year survival probabilities, improvements continued at a consistent rate over time. For API, Hispanic, and NHW persons, there appears to be a slowing of the rate of improvement in the second half of the study period. The adjusted time-averaged survival probabilities were low: 46.6% (42.3%-50.9%) for NHB, 48.3% (37.9%-58.7%) for AIAN, 50.5% (48.5%-52.5%) for NHW, 58.1% (54.2%-62.0%) for Hispanic, and 58.3% (54.2%-62.4%) for API persons. All pairwise comparisons among these groups were statistically significant (all P < .01), except for differences between the two groups with the lowest (AIAN vs NHB, P = .87), and between the two groups with the highest (API vs Hispanic, P = .69), average adjusted 1-year cause-specific survival probabilities.\n[SUBTITLE] Five-Year Survival by Race and Ethnicity [SUBSECTION] Estimates of adjusted 5-year cause-specific survival probabilities are shown for all race/ethnicity by stage combinations in Tables 4 and 5. Neither the race/ethnicity by stage by year of diagnosis three-way interaction (P = .90) nor the two-way interaction between the year of diagnosis and race/ethnicity (P = .59) were statistically significant; there is not sufficient evidence to conclude that there are race- or ethnic-specific differences in 5-year survival trends either within or across stages of CRC at diagnosis. There were statistically significant interactions between race/ethnicity and stage (P < .001), and between stage and the year of diagnosis trends (P < .001) with respect to differences in 5-year survival (see Figure 3).Table 4.Unadjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199283.2 (67.0-99.3)58.1 (40.1-76.0)8.5 (0-29.1)49.9 (39.3-60.5)AIAN199587.5 (70.6-100)64.7 (47.7-81.7)13.4 (0-33.0)55.2 (44.9-65.5)AIAN199885.4 (69.9-100)73.1 (57.2-89.0)1.6 (0-18.5)53.3 (44.0-62.6)AIAN200291.8 (79.0-100)70.4 (56.5-84.3)10.8 (0-33.9)57.6 (47.7-67.6)AIAN200597.1 (85.6-100)65.6 (52.5-78.6)23.0 (6.8-39.2)61.9 (54.0-69.8)AIAN200897.5 (84.4-100)83.4 (70.7-96.1)5.9 (0-22.7)62.3 (54.0-70.6)AIAN201189.3 (77.0-100)67.5 (55.3-79.6)9.5 (0.0-22.7)55.4 (48.2-62.7)AIAN201496.5 (86.4-100)71.4 (60.1-82.6)15.1 (2.0-28.2)61.0 (54.3-67.6)API199297.1 (93.4-100)63.9 (57.9-70.0)7.1 (.9-13.2)56.0 (52.9-59.1)API199594.8 (91.1-98.4)67.5 (62.0-73.0)4.9 (0-10.7)55.7 (52.8-58.6)API199896.1 (93.0-99.2)76.8 (71.8-81.8)7.9 (2.5-13.4)60.3 (57.6-62.9)API200296.7 (94.3-99.1)79.8 (75.4-84.1)7.1 (2.5-11.8)61.2 (58.9-63.5)API200596.5 (94.1-98.9)80.0 (75.9-84.1)9.8 (4.7-14.8)62.1 (59.8-64.4)API200897.7 (95.7-99.7)80.6 (76.3-85)9.0 (4.1-13.9)62.4 (60.1-64.7)API201196.7 (94.4-99.0)76.7 (72.5-81.0)8.4 (3.6-13.1)60.6 (58.3-62.9)API201497.2 (95.0-99.4)81.5 (77.6-85.5)8.0 (3.8-12.1)62.2 (60.2-64.3)Hispanic199292.6 (87.7-97.5)58.1 (51.7-64.4)6.7 (0-13.4)52.4 (48.9-55.9)Hispanic199590.7 (85.5-95.8)61.0 (54.6-67.4)5.4 (0-11.4)52.4 (49.0-55.8)Hispanic199894.3 (90.6-98.0)72.7 (67.0-78.3)5.5 (0-11.5)57.5 (54.4-60.5)Hispanic200293.8 (90.1-97.4)75.5 (70.3-80.8)6.0 (.8-11.2)58.4 (55.7-61.2)Hispanic200594.6 (91.6-97.6)74.1 (69.3-78.9)9.3 (4.1-14.5)59.3 (56.8-61.9)Hispanic200896.0 (93.5-98.6)74.0 (69.1-78.9)6.3 (1.6-10.9)58.8 (56.4-61.2)Hispanic201197.6 (95.5-99.6)79 (74.6-83.3)8.9 (4.6-13.1)61.8 (59.6-63.9)Hispanic201496.9 (94.8-99)75.2 (70.9-79.4)9.4 (5.1-13.7)60.5 (58.4-62.6)NHB199290.6 (85.7-95.4)60.9 (54.1-67.7)2.2 (0-6.8)51.2 (48.1-54.4)NHB199591.3 (86.7-95.9)63.8 (57.0-70.6)4.3 (0-9.6)53.1 (49.8-56.4)NHB199896.0 (92.5-99.4)66.4 (60.0-72.9)4.8 (0-9.8)55.7 (52.8-58.7)NHB200289.9 (85.2-94.6)71.6 (65.9-77.3)4.5 (0-9.0)55.3 (52.4-58.2)NHB200591.8 (87.9-95.7)73.5 (67.7-79.3)4.3 (.1-8.4)56.5 (53.8-59.2)NHB200893.8 (90.4-97.1)69.3 (63.3-75.3)5.7 (1.4-10.1)56.3 (53.6-59.0)NHB201193.7 (90.2-97.2)68.7 (62.4-75.0)5.4 (1.2-9.5)55.9 (53.2-58.7)NHB201494.7 (91.4-98.0)69.2 (63.1-75.4)5.9 (1.7-10.1)56.6 (53.9-59.3)NHW199292.4 (91.1-93.7)64.4 (62.2-66.5)4.8 (3.2-6.5)53.9 (52.9-54.9)NHW199591.8 (90.5-93.2)65.6 (63.3-67.8)5.9 (4.2-7.6)54.4 (53.4-55.5)NHW199893.2 (92-94.4)67.8 (65.7-69.9)4.2 (2.7-5.7)55.1 (54.1-56)NHW200293.7 (92.6-94.9)73.4 (71.3-75.5)4.1 (2.7-5.5)57.1 (56.2-58)NHW200595.3 (94.2-96.3)77.7 (75.7-79.7)5.1 (3.5-6.7)59.4 (58.4-60.3)NHW200895.4 (94.4-96.4)77.6 (75.6-79.7)9.0 (7.0-11.1)60.7 (59.7-61.7)NHW201196.1 (95.1-97.2)78.8 (76.7-80.9)4.8 (3.3-6.4)59.9 (59.0-60.9)NHW201496.2 (95.1-97.3)79.3 (77.2-81.4)8.1 (6.2-10.1)61.2 (60.2-62.2)Table 5.Adjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199278.8 (64.3-93.4)56.6 (40.4-72.7)7.5 (.0-26.0)47.6 (38.1-57.1)AIAN199585.5 (70.3-100)62.1 (46.8-77.4)13.3 (.0-31.0)53.7 (44.4-63.0)AIAN199881.1 (67.1-95.0)70.0 (55.6-84.3)2.8 (.0-18.0)51.3 (42.8-59.7)AIAN200289.4 (77.9-100)68.8 (56.3-81.3)8.2 (.0-29.1)55.5 (46.5-64.5)AIAN200595.6 (85.2-100)63.8 (52.0-75.6)20.4 (5.8-35.0)59.9 (52.7-67.1)AIAN200893.6 (81.8-100)79.8 (68.3-91.2)4.3 (.0-19.5)59.2 (51.8-66.7)AIAN201184.9 (73.8-96.0)65.4 (54.4-76.3)7.3 (.0-19.2)52.5 (46.0-59.1)AIAN201493.0 (84.0-100)68.6 (58.4-78.7)13.3 (1.5-25.1)58.3 (52.3-64.3)API199295.4 (92.1-98.8)63.1 (57.6-68.5)6.5 (.9-12.0)55.0 (52.1-57.9)API199592.3 (88.9-95.7)67.1 (62.1-72.1)6.4 (1.1-11.6)55.3 (52.5-58.0)API199895.2 (92.4-98.1)75.7 (71.2-80.2)8.8 (3.8-13.7)59.9 (57.4-62.4)API200294.5 (92.2-96.7)78.1 (74.2-82.1)7.9 (3.7-12.1)60.2 (58.0-62.3)API200593.7 (91.4-96.0)78.3 (74.6-82.1)10.6 (6.0-15.2)60.9 (58.6-63.1)API200895.3 (93.3-97.2)78.9 (74.9-82.9)10.7 (6.2-15.2)61.6 (59.4-63.8)API201193.2 (90.9-95.4)75.4 (71.5-79.3)9.3 (5.0-13.7)59.3 (57.1-61.5)API201492.6 (90.5-94.7)78.9 (75.2-82.5)11.6 (7.8-15.4)61.0 (59.0-63.0)Hispanic199289.7 (85.3-94.2)57.3 (51.5-63.0)8.6 (2.5-14.8)51.9 (48.6-55.1)Hispanic199588.8 (84.1-93.5)60.2 (54.4-66.0)5.6 (.2-11.0)51.5 (48.4-54.7)Hispanic199891.2 (87.8-94.6)71.2 (66.1-76.4)6.5 (1.1-12.0)56.3 (53.5-59.1)Hispanic200291.0 (87.7-94.4)73.6 (68.8-78.4)5.8 (1.0-10.5)56.8 (54.2-59.4)Hispanic200591.5 (88.7-94.4)72.7 (68.3-77.0)8.8 (4.0-13.5)57.7 (55.2-60.1)Hispanic200892.6 (90.2-95.0)71.6 (67.2-76.1)6.6 (2.3-10.8)57.0 (54.7-59.2)Hispanic201194.6 (92.6-96.6)76.7 (72.7-80.7)8.4 (4.5-12.3)59.9 (57.8-62.0)Hispanic201492.5 (90.4-94.5)72.8 (68.9-76.7)9.6 (5.7-13.6)58.3 (56.2-60.4)NHB199287.5 (83.0-91.9)59.6 (53.4-65.8)1.3 (.0-5.5)49.4 (46.5-52.4)NHB199589.5 (85.2-93.7)62.3 (56.2-68.5)3.4 (.0-8.3)51.7 (48.7-54.8)NHB199892.7 (89.5-95.9)64.6 (58.7-70.4)5.4 (.9-10.0)54.2 (51.5-57.0)NHB200286.8 (82.5-91.0)69.7 (64.5-74.9)4.2 (.0-8.4)53.6 (50.8-56.3)NHB200589.0 (85.4-92.6)71.5 (66.2-76.8)3.7 (.0-7.5)54.7 (52.2-57.3)NHB200890.0 (86.9-93.2)67.7 (62.2-73.1)6.2 (2.1-10.2)54.6 (52.1-57.2)NHB201190.0 (86.7-93.2)66.8 (61.1-72.6)4.0 (.1-7.8)53.6 (51.0-56.2)NHB201490.9 (87.8-94.0)68.0 (62.4-73.6)6.1 (2.2-10.0)55.0 (52.4-57.6)NHW199290.7 (89.3-92.1)65.1 (63.0-67.2)7.3 (5.7-9.0)54.4 (53.2-55.6)NHW199590.5 (89.1-92.0)66.4 (64.3-68.6)8.0 (6.3-9.8)55.0 (53.8-56.2)NHW199891.1 (89.8-92.4)68.6 (66.6-70.7)9.4 (7.8-10.9)56.4 (55.2-57.5)NHW200292.1 (90.8-93.4)73.5 (71.5-75.6)8.7 (7.2-10.2)58.1 (57.0-59.3)NHW200592.3 (91.0-93.5)77.1 (75.1-79.0)10.7 (9.0-12.3)60.0 (58.9-61.1)NHW200892.7 (91.5-93.9)77.1 (75.0-79.1)12.3 (10.3-14.3)60.7 (59.5-61.9)NHW201193.5 (92.2-94.7)78.0 (75.9-80.0)10.7 (9.1-12.3)60.7 (59.6-61.9)NHW201492.3 (91.1-93.6)77.7 (75.7-79.8)13.2 (11.3-15.1)61.1 (59.9-62.3)Figure 3.Trends in 5-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2014.\nUnadjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nAdjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nTrends in 5-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2014.\nThe trends reflect improvements in 5-year cause-specific survival probabilities over time for all CRC stages. The rate of improvement in 5-year survival probabilities over time was greatest for those with regional stage CRC, although the rate of improvement appears to be slowing in the second half of the study period. As there are no significantly different trends over time in adjusted 5-year survival probabilities by race or ethnicity, key differences among race and ethnic groups are best summarized by differences in their adjusted time-averaged survival probabilities. For those diagnosed at localized stage CRC, estimates of average adjusted 5-year survival probabilities were 86.9% (84.2%-89.6%) for AIAN, 90.2% (89.4%-91.0%) for NHB, 92.0% (91.8%-92.2%) for NHW, 92.1% (91.5%-92.7%) for Hispanic, and 94.1% (93.5%-94.7%) for API. These differed significantly between all pairs of groups (all P < .04), except for the Hispanic vs NHW comparison (P = .56). For those diagnosed at regional stage CRC, estimates of average adjusted 5-year survival probabilities were 65.1% (62.2%-68.0%) for NHB, 67.5% (61.0%-74.0%) for AIAN, 70.5% (68.1%-72.9%) for Hispanic, 72.5% (71.5%-73.5%) for NHW, and 75.1% (72.9%-77.3%) for API persons. These differed significantly between all pairs of groups (all P < .05), except for the AIAN vs NHB comparison (P = .22). For those diagnosed at distant stage CRC, estimates of average adjusted 5-year survival probabilities were 4.6% (3.6%-5.6%) for NHB, 7.8% (6.6%-9.0%) for Hispanic, 8.5% (7.3%-9.7%) for API, 9.0% (5.5%-12.5%) for AIAN, and 9.6% (9.2%-10.0%) for NHW. The AIAN average did not differ significantly from those of the API, Hispanic, or NHW groups, nor did the API vs Hispanic averages. All others differed significantly (all P < .01).\nEstimates of adjusted 5-year cause-specific survival probabilities are shown for all race/ethnicity by stage combinations in Tables 4 and 5. Neither the race/ethnicity by stage by year of diagnosis three-way interaction (P = .90) nor the two-way interaction between the year of diagnosis and race/ethnicity (P = .59) were statistically significant; there is not sufficient evidence to conclude that there are race- or ethnic-specific differences in 5-year survival trends either within or across stages of CRC at diagnosis. There were statistically significant interactions between race/ethnicity and stage (P < .001), and between stage and the year of diagnosis trends (P < .001) with respect to differences in 5-year survival (see Figure 3).Table 4.Unadjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199283.2 (67.0-99.3)58.1 (40.1-76.0)8.5 (0-29.1)49.9 (39.3-60.5)AIAN199587.5 (70.6-100)64.7 (47.7-81.7)13.4 (0-33.0)55.2 (44.9-65.5)AIAN199885.4 (69.9-100)73.1 (57.2-89.0)1.6 (0-18.5)53.3 (44.0-62.6)AIAN200291.8 (79.0-100)70.4 (56.5-84.3)10.8 (0-33.9)57.6 (47.7-67.6)AIAN200597.1 (85.6-100)65.6 (52.5-78.6)23.0 (6.8-39.2)61.9 (54.0-69.8)AIAN200897.5 (84.4-100)83.4 (70.7-96.1)5.9 (0-22.7)62.3 (54.0-70.6)AIAN201189.3 (77.0-100)67.5 (55.3-79.6)9.5 (0.0-22.7)55.4 (48.2-62.7)AIAN201496.5 (86.4-100)71.4 (60.1-82.6)15.1 (2.0-28.2)61.0 (54.3-67.6)API199297.1 (93.4-100)63.9 (57.9-70.0)7.1 (.9-13.2)56.0 (52.9-59.1)API199594.8 (91.1-98.4)67.5 (62.0-73.0)4.9 (0-10.7)55.7 (52.8-58.6)API199896.1 (93.0-99.2)76.8 (71.8-81.8)7.9 (2.5-13.4)60.3 (57.6-62.9)API200296.7 (94.3-99.1)79.8 (75.4-84.1)7.1 (2.5-11.8)61.2 (58.9-63.5)API200596.5 (94.1-98.9)80.0 (75.9-84.1)9.8 (4.7-14.8)62.1 (59.8-64.4)API200897.7 (95.7-99.7)80.6 (76.3-85)9.0 (4.1-13.9)62.4 (60.1-64.7)API201196.7 (94.4-99.0)76.7 (72.5-81.0)8.4 (3.6-13.1)60.6 (58.3-62.9)API201497.2 (95.0-99.4)81.5 (77.6-85.5)8.0 (3.8-12.1)62.2 (60.2-64.3)Hispanic199292.6 (87.7-97.5)58.1 (51.7-64.4)6.7 (0-13.4)52.4 (48.9-55.9)Hispanic199590.7 (85.5-95.8)61.0 (54.6-67.4)5.4 (0-11.4)52.4 (49.0-55.8)Hispanic199894.3 (90.6-98.0)72.7 (67.0-78.3)5.5 (0-11.5)57.5 (54.4-60.5)Hispanic200293.8 (90.1-97.4)75.5 (70.3-80.8)6.0 (.8-11.2)58.4 (55.7-61.2)Hispanic200594.6 (91.6-97.6)74.1 (69.3-78.9)9.3 (4.1-14.5)59.3 (56.8-61.9)Hispanic200896.0 (93.5-98.6)74.0 (69.1-78.9)6.3 (1.6-10.9)58.8 (56.4-61.2)Hispanic201197.6 (95.5-99.6)79 (74.6-83.3)8.9 (4.6-13.1)61.8 (59.6-63.9)Hispanic201496.9 (94.8-99)75.2 (70.9-79.4)9.4 (5.1-13.7)60.5 (58.4-62.6)NHB199290.6 (85.7-95.4)60.9 (54.1-67.7)2.2 (0-6.8)51.2 (48.1-54.4)NHB199591.3 (86.7-95.9)63.8 (57.0-70.6)4.3 (0-9.6)53.1 (49.8-56.4)NHB199896.0 (92.5-99.4)66.4 (60.0-72.9)4.8 (0-9.8)55.7 (52.8-58.7)NHB200289.9 (85.2-94.6)71.6 (65.9-77.3)4.5 (0-9.0)55.3 (52.4-58.2)NHB200591.8 (87.9-95.7)73.5 (67.7-79.3)4.3 (.1-8.4)56.5 (53.8-59.2)NHB200893.8 (90.4-97.1)69.3 (63.3-75.3)5.7 (1.4-10.1)56.3 (53.6-59.0)NHB201193.7 (90.2-97.2)68.7 (62.4-75.0)5.4 (1.2-9.5)55.9 (53.2-58.7)NHB201494.7 (91.4-98.0)69.2 (63.1-75.4)5.9 (1.7-10.1)56.6 (53.9-59.3)NHW199292.4 (91.1-93.7)64.4 (62.2-66.5)4.8 (3.2-6.5)53.9 (52.9-54.9)NHW199591.8 (90.5-93.2)65.6 (63.3-67.8)5.9 (4.2-7.6)54.4 (53.4-55.5)NHW199893.2 (92-94.4)67.8 (65.7-69.9)4.2 (2.7-5.7)55.1 (54.1-56)NHW200293.7 (92.6-94.9)73.4 (71.3-75.5)4.1 (2.7-5.5)57.1 (56.2-58)NHW200595.3 (94.2-96.3)77.7 (75.7-79.7)5.1 (3.5-6.7)59.4 (58.4-60.3)NHW200895.4 (94.4-96.4)77.6 (75.6-79.7)9.0 (7.0-11.1)60.7 (59.7-61.7)NHW201196.1 (95.1-97.2)78.8 (76.7-80.9)4.8 (3.3-6.4)59.9 (59.0-60.9)NHW201496.2 (95.1-97.3)79.3 (77.2-81.4)8.1 (6.2-10.1)61.2 (60.2-62.2)Table 5.Adjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199278.8 (64.3-93.4)56.6 (40.4-72.7)7.5 (.0-26.0)47.6 (38.1-57.1)AIAN199585.5 (70.3-100)62.1 (46.8-77.4)13.3 (.0-31.0)53.7 (44.4-63.0)AIAN199881.1 (67.1-95.0)70.0 (55.6-84.3)2.8 (.0-18.0)51.3 (42.8-59.7)AIAN200289.4 (77.9-100)68.8 (56.3-81.3)8.2 (.0-29.1)55.5 (46.5-64.5)AIAN200595.6 (85.2-100)63.8 (52.0-75.6)20.4 (5.8-35.0)59.9 (52.7-67.1)AIAN200893.6 (81.8-100)79.8 (68.3-91.2)4.3 (.0-19.5)59.2 (51.8-66.7)AIAN201184.9 (73.8-96.0)65.4 (54.4-76.3)7.3 (.0-19.2)52.5 (46.0-59.1)AIAN201493.0 (84.0-100)68.6 (58.4-78.7)13.3 (1.5-25.1)58.3 (52.3-64.3)API199295.4 (92.1-98.8)63.1 (57.6-68.5)6.5 (.9-12.0)55.0 (52.1-57.9)API199592.3 (88.9-95.7)67.1 (62.1-72.1)6.4 (1.1-11.6)55.3 (52.5-58.0)API199895.2 (92.4-98.1)75.7 (71.2-80.2)8.8 (3.8-13.7)59.9 (57.4-62.4)API200294.5 (92.2-96.7)78.1 (74.2-82.1)7.9 (3.7-12.1)60.2 (58.0-62.3)API200593.7 (91.4-96.0)78.3 (74.6-82.1)10.6 (6.0-15.2)60.9 (58.6-63.1)API200895.3 (93.3-97.2)78.9 (74.9-82.9)10.7 (6.2-15.2)61.6 (59.4-63.8)API201193.2 (90.9-95.4)75.4 (71.5-79.3)9.3 (5.0-13.7)59.3 (57.1-61.5)API201492.6 (90.5-94.7)78.9 (75.2-82.5)11.6 (7.8-15.4)61.0 (59.0-63.0)Hispanic199289.7 (85.3-94.2)57.3 (51.5-63.0)8.6 (2.5-14.8)51.9 (48.6-55.1)Hispanic199588.8 (84.1-93.5)60.2 (54.4-66.0)5.6 (.2-11.0)51.5 (48.4-54.7)Hispanic199891.2 (87.8-94.6)71.2 (66.1-76.4)6.5 (1.1-12.0)56.3 (53.5-59.1)Hispanic200291.0 (87.7-94.4)73.6 (68.8-78.4)5.8 (1.0-10.5)56.8 (54.2-59.4)Hispanic200591.5 (88.7-94.4)72.7 (68.3-77.0)8.8 (4.0-13.5)57.7 (55.2-60.1)Hispanic200892.6 (90.2-95.0)71.6 (67.2-76.1)6.6 (2.3-10.8)57.0 (54.7-59.2)Hispanic201194.6 (92.6-96.6)76.7 (72.7-80.7)8.4 (4.5-12.3)59.9 (57.8-62.0)Hispanic201492.5 (90.4-94.5)72.8 (68.9-76.7)9.6 (5.7-13.6)58.3 (56.2-60.4)NHB199287.5 (83.0-91.9)59.6 (53.4-65.8)1.3 (.0-5.5)49.4 (46.5-52.4)NHB199589.5 (85.2-93.7)62.3 (56.2-68.5)3.4 (.0-8.3)51.7 (48.7-54.8)NHB199892.7 (89.5-95.9)64.6 (58.7-70.4)5.4 (.9-10.0)54.2 (51.5-57.0)NHB200286.8 (82.5-91.0)69.7 (64.5-74.9)4.2 (.0-8.4)53.6 (50.8-56.3)NHB200589.0 (85.4-92.6)71.5 (66.2-76.8)3.7 (.0-7.5)54.7 (52.2-57.3)NHB200890.0 (86.9-93.2)67.7 (62.2-73.1)6.2 (2.1-10.2)54.6 (52.1-57.2)NHB201190.0 (86.7-93.2)66.8 (61.1-72.6)4.0 (.1-7.8)53.6 (51.0-56.2)NHB201490.9 (87.8-94.0)68.0 (62.4-73.6)6.1 (2.2-10.0)55.0 (52.4-57.6)NHW199290.7 (89.3-92.1)65.1 (63.0-67.2)7.3 (5.7-9.0)54.4 (53.2-55.6)NHW199590.5 (89.1-92.0)66.4 (64.3-68.6)8.0 (6.3-9.8)55.0 (53.8-56.2)NHW199891.1 (89.8-92.4)68.6 (66.6-70.7)9.4 (7.8-10.9)56.4 (55.2-57.5)NHW200292.1 (90.8-93.4)73.5 (71.5-75.6)8.7 (7.2-10.2)58.1 (57.0-59.3)NHW200592.3 (91.0-93.5)77.1 (75.1-79.0)10.7 (9.0-12.3)60.0 (58.9-61.1)NHW200892.7 (91.5-93.9)77.1 (75.0-79.1)12.3 (10.3-14.3)60.7 (59.5-61.9)NHW201193.5 (92.2-94.7)78.0 (75.9-80.0)10.7 (9.1-12.3)60.7 (59.6-61.9)NHW201492.3 (91.1-93.6)77.7 (75.7-79.8)13.2 (11.3-15.1)61.1 (59.9-62.3)Figure 3.Trends in 5-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2014.\nUnadjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nAdjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nTrends in 5-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2014.\nThe trends reflect improvements in 5-year cause-specific survival probabilities over time for all CRC stages. The rate of improvement in 5-year survival probabilities over time was greatest for those with regional stage CRC, although the rate of improvement appears to be slowing in the second half of the study period. As there are no significantly different trends over time in adjusted 5-year survival probabilities by race or ethnicity, key differences among race and ethnic groups are best summarized by differences in their adjusted time-averaged survival probabilities. For those diagnosed at localized stage CRC, estimates of average adjusted 5-year survival probabilities were 86.9% (84.2%-89.6%) for AIAN, 90.2% (89.4%-91.0%) for NHB, 92.0% (91.8%-92.2%) for NHW, 92.1% (91.5%-92.7%) for Hispanic, and 94.1% (93.5%-94.7%) for API. These differed significantly between all pairs of groups (all P < .04), except for the Hispanic vs NHW comparison (P = .56). For those diagnosed at regional stage CRC, estimates of average adjusted 5-year survival probabilities were 65.1% (62.2%-68.0%) for NHB, 67.5% (61.0%-74.0%) for AIAN, 70.5% (68.1%-72.9%) for Hispanic, 72.5% (71.5%-73.5%) for NHW, and 75.1% (72.9%-77.3%) for API persons. These differed significantly between all pairs of groups (all P < .05), except for the AIAN vs NHB comparison (P = .22). For those diagnosed at distant stage CRC, estimates of average adjusted 5-year survival probabilities were 4.6% (3.6%-5.6%) for NHB, 7.8% (6.6%-9.0%) for Hispanic, 8.5% (7.3%-9.7%) for API, 9.0% (5.5%-12.5%) for AIAN, and 9.6% (9.2%-10.0%) for NHW. The AIAN average did not differ significantly from those of the API, Hispanic, or NHW groups, nor did the API vs Hispanic averages. All others differed significantly (all P < .01).", "Adjusted 1- and 5-year cause-specific survival probabilities following CRC diagnosis by stage and year of diagnosis are shown in Figure 1. For those diagnosed with local stage CRC, the estimate of linear trend in 1-year survival probabilities was negligible, at −.01% ([95% Confidence Interval (CI)] −.03%-.01%) per year, with an average 1-year survival probability of 96.2% (93.8%-98.6%). Those diagnosed with regional stage CRC displayed a small improvement over time in 1-year survival probabilities, .06% (.001%-.12%) per year. Their average 1-year survival probability was 93.0% (90.5%-95.5%). For those diagnosed with distant stage CRC, there was a significant improvement in 1-year survival probabilities over time (P < .001), but there was evidence of departure from a linear trend (P = .04). The slope of the trend line suggested an improvement in survival probability of 1.0% (.8%-1.2%) per year in 2005, with instantaneous slopes suggesting greater gains, by .05% (.01%-.09%), for each year prior to 2005 and slower gains by that same amount for each year following 2005. Their average 1-year survival probability was 52.8% (48.1%-57.5%).Figure 1.Trends in 1- and 5-year adjusted cause-specific survival probabilities for individuals diagnosed at different stages of colorectal cancer. Shaded bands reflect 95% prediction intervals for the year-specific survival probabilities.\nTrends in 1- and 5-year adjusted cause-specific survival probabilities for individuals diagnosed at different stages of colorectal cancer. Shaded bands reflect 95% prediction intervals for the year-specific survival probabilities.\nThose diagnosed with local stage CRC experienced improvements in 5-year cause-specific survival, with an average improvement of .16% (.10%-.21%) per year. Their average 5-year survival probability was 90.9% (87.9%-93.8%). Those diagnosed with regional stage CRC displayed significant improvement in 5-year survival probabilities over time (P < .001), but there was evidence of significant departure from a linear trend (P = .004), such that the slope of the trend line showed an improvement of .55% (.41%-.69%) per year in 2003, with instantaneous slopes suggested greater gains, by .08% (.04-.12%), for each year prior to 2003 and slower gains by that same amount for each year following 2003. Their average 5-year survival probability was 70.3% (66.5%-74.0%). For those diagnosed with distant stage CRC, there was a significant but small improvement over time in 5-year survival probabilities, .15% (.5%-.25%) per year. Their average 5-year survival probability was 8.1% (4.4%-11.8%).", "Estimates of adjusted 1-year cause-specific survival probabilities following diagnosis are shown for all race/ethnicity by stage combinations in Tables 2 and 3. Trends in 1-year survival for those of different races/ethnicities who were diagnosed at different stages of CRC differed significantly (P < .001, see Figure 2). For those diagnosed with localized stage CRC, 1-year survival probabilities were consistently high for individuals of all race and ethnic groups. AIAN persons experienced lower 1-year survival probabilities than those from other race and ethnic groups over the study period. Although due to lack of precision in the estimates, the difference was only significantly different when compared to API persons; average 1-year survival probabilities were 1.5 (.5-2.5) percentage points lower for AIAN than for API persons.Table 2.Unadjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN1992100 (84.6-100)91.6 (75.4-100)27.0 (5.6-48.4)72.9 (62.6-83.2)AIAN199696.9 (81.8-100)95.7 (77.9-100)41.8 (23.5-60.1)78.1 (68.2-88.0)AIAN200098.7 (83.6-100)83.2 (65.9-100)43.0 (20.0-66.0)75.0 (64.1-85.8)AIAN200496.7 (85.6-100)96.4 (84.8-100)51.8 (35.8-67.7)81.6 (74.1-89.2)AIAN2008100 (86.4-100)95.5 (83.0-100)53.5 (35.5-71.5)83.0 (74.4-91.6)AIAN2012100 (88.9-100)91.6 (79.1-100)52.7 (38.0-67.4)81.4 (74.0-88.8)AIAN201696.0 (85.5-100)95.9 (85.6-100)46.5 (33.4-59.6)79.5 (72.9-86.0)AIAN2018100 (90.7-100)99.3 (88.9-100)83.5 (67.2-99.8)94.3 (87.1-100)API199299.5 (96.2-100)94.6 (90.8-98.3)46.5 (38.4-54.6)80.2 (77.0-83.3)API199699.4 (97.1-100)95.8 (92.6-98.9)56.5 (48.7-64.3)83.9 (81.0-86.8)API200099.2 (97.0-100)97.9 (95.8-99.9)52.8 (45.0-60.6)83.3 (80.5-86.1)API200499.6 (97.9-100)97.3 (95.1-99.5)68.6 (62.7-74.4)88.5 (86.3-90.6)API200899.7 (98.3-100)98.4 (96.5-100)59.5 (53.9-65.0)85.9 (83.9-87.9)API201299.4 (97.9-100)96.9 (94.7-99.2)55.8 (49.4-62.3)84.1 (81.7-86.4)API201699.1 (97.4-100)98.4 (96.7-100)57.6 (51.8-63.3)85.0 (82.9-87.1)API201899.6 (98.5-100)97.8 (96.0-99.6)69.1 (62.7-75.5)88.8 (86.6-91.1)Hispanic199297.0 (93.0-100)96.7 (93.3-100)55.6 (46.2-64.9)83.1 (79.5-86.7)Hispanic199698.8 (95.8-100)96.1 (92.7-99.5)43.4 (35.7-51.1)79.4 (76.4-82.4)Hispanic200099.2 (96.6-100)96.3 (93.3-99.4)47.0 (39.8-54.2)80.8 (78.1-83.6)Hispanic200498.9 (96.5-100)95.8 (92.9-98.7)57.8 (50.8-64.8)84.2 (81.5-86.8)Hispanic200899.1 (97.3-100)96.2 (93.5-98.8)60.7 (54.4-67.1)85.3 (83.0-87.7)Hispanic201299.2 (97.4-100)95.8 (93.3-98.2)71.6 (66.6-76.7)88.9 (86.9-90.8)Hispanic201698.9 (97.1-100)98.2 (96.6-99.7)69.0 (64.0-74.1)88.7 (86.9-90.6)Hispanic201899.6 (98.6-100)97.8 (96.1-99.5)58.8 (52.5-65.0)85.4 (83.2-87.6)NHB199298.0 (94.7-100)93.7 (89.6-97.8)38.5 (30.0-47.0)76.7 (73.4-80.1)NHB199698.2 (94.7-100)96.3 (93-99.6)30.1 (23.1-37.2)74.9 (72.0-77.7)NHB200097.3 (93.8-100)93.2 (89.4-97.0)34.5 (27.3-41.7)75.0 (72.0-77.9)NHB200498.4 (95.6-100)97.0 (94.3-99.7)37.0 (30.7-43.3)77.5 (75.0-79.9)NHB200898.6 (96.2-100)93.3 (89.4-97.2)51.4 (44.7-58.1)81.1 (78.4-83.8)NHB201299.5 (97.2-100)93.6 (90.0-97.1)68.5 (62.1-74.8)87.2 (84.7-89.7)NHB201699.5 (97.2-100)96.0 (93.0-99.0)53.1 (47.2-58.9)82.9 (80.5-85.2)NHB201899.3 (97.5-100)97.0 (94.2-99.8)57.1 (49.6-64.6)84.5 (81.7-87.2)NHW199298.6 (97.9-99.2)94.6 (93.5-95.7)38.0 (35.0-40.9)77.0 (76.0-78.1)NHW199698.9 (98.3-99.5)96.1 (95.1-97.0)36.7 (33.8-39.5)77.2 (76.2-78.2)NHW200099.0 (98.5-99.6)94.3 (93.2-95.4)40.9 (37.9-43.9)78.1 (77.0-79.1)NHW200498.9 (98.3-99.5)96.1 (95.1-97.1)52.6 (49.7-55.4)82.5 (81.5-83.6)NHW200899.2 (98.6-99.7)97.0 (96.1-98.0)58.6 (55.6-61.7)85.0 (83.9-86.0)NHW201298.9 (98.2-99.6)97.2 (96.2-98.2)55.2 (52.3-58.0)83.8 (82.7-84.8)NHW201699.1 (98.5-99.8)96.9 (95.9-97.9)60.7 (57.9-63.5)85.6 (84.6-86.6)NHW201898.7 (98.0-99.4)97.5 (96.7-98.4)59.0 (55.7-62.3)85.1 (83.9-86.3)Table 3.Adjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199297.5 (83.7-100)89.8 (75.3-100)25.4 (6.2-44.6)70.9 (61.6-80.2)AIAN199695.6 (82.0-100)93.2 (77.3-100)41.9 (25.5-58.3)76.9 (68.0-85.8)AIAN200097.0 (83.5-100)82.5 (67.0-97.9)42.0 (21.4-62.6)73.8 (64.1-83.5)AIAN200495.3 (85.3-100)94.2 (83.8-100)50.4 (36.1-64.7)80.0 (73.2-86.8)AIAN200897.5 (85.3-100)92.4 (81.2-100)51.6 (35.5-67.7)80.5 (72.8-88.2)AIAN201297.5 (87.5-100)89.9 (78.7-100)50.2 (37.0-63.3)79.2 (72.5-85.9)AIAN201693.4 (84.0-100)94.1 (84.8-100)45.0 (33.3-56.8)77.5 (71.6-83.4)AIAN201896.7 (87.8-100)96.9 (87.6-100)64.2 (51.1-77.5)85.9 (79.7-91.7)API199297.5 (94.5-100)92.3 (88.9-95.7)45.8 (38.6-53.1)78.6 (75.7-81.4)API199697.4 (95.2-99.6)93.4 (90.5-96.3)55.6 (48.5-62.6)82.1 (79.4-84.8)API200097.4 (95.3-99.5)95.9 (93.9-97.9)53.1 (46.1-60.1)82.1 (79.6-84.7)API200497.8 (96.2-99.4)94.6 (92.5-96.7)67.1 (61.8-72.4)86.5 (84.5-88.5)API200897.5 (96.0-98.9)95.6 (93.7-97.4)59.4 (54.4-64.4)84.2 (82.2-86.1)API201296.8 (95.3-98.3)94.6 (92.5-96.7)56.1 (50.3-61.9)82.5 (80.3-84.7)API201696.4 (94.8-98.0)95.0 (93.4-96.6)57.8 (52.6-63.0)83.1 (81.1-85.0)API201897.9 (96.5-99.0)96.1 (94.5-97.9)63.8 (58.3-69.3)86.0 (83.9-88.0)Hispanic199294.7 (91.1-98.4)94.4 (91.3-97.5)54.3 (45.9-62.7)81.2 (77.9-84.4)Hispanic199696.8 (93.9-99.6)94.2 (91.0-97.3)42.9 (36.0-49.9)77.9 (75.2-80.7)Hispanic200097.0 (94.6-99.4)93.4 (90.6-96.2)46.6 (40.1-53.0)79.0 (76.4-81.5)Hispanic200497.2 (95.0-99.4)93.3 (90.6-96.0)57.1 (50.8-63.4)82.5 (80.1-85.0)Hispanic200896.2 (94.5-97.9)92.8 (90.4-95.3)59.8 (54.1-65.5)83.0 (80.8-85.2)Hispanic201296.0 (94.3-97.8)92.9 (90.6-95.2)69.3 (64.8-73.9)86.1 (84.2-88.0)Hispanic201694.8 (93.0-96.5)94.1 (92.5-95.6)67.3 (62.8-71.9)85.4 (83.6-87.2)Hispanic201896.8 (95.3-98.3)95.3 (93.7-96.9)63.4 (58.3-68.6)85.2 (83.2-87.2)NHB199295.7 (92.6-98.7)91.1 (87.3-94.8)37.8 (30.2-45.4)74.8 (71.8-77.9)NHB199696.6 (93.4-99.9)93.8 (90.7-96.8)30.8 (24.4-37.1)73.7 (71.1-76.3)NHB200095.6 (92.5-98.8)90.5 (87.1-94.0)35.7 (29.2-42.2)74.0 (71.2-76.7)NHB200495.9 (93.3-98.5)94.0 (91.4-96.5)38.9 (33.2-44.6)76.3 (73.9-78.6)NHB200896.1 (93.9-98.3)90.3 (86.8-93.9)51.2 (45.1-57.2)79.2 (76.7-81.7)NHB201296.6 (94.4-98.7)90.6 (87.4-93.8)66.7 (60.9-72.4)84.6 (82.2-87.0)NHB201696.0 (93.9-98.2)92.3 (89.6-95.1)53.0 (47.7-58.3)80.5 (78.3-82.6)NHB201897.5 (95.6-99.1)94.3 (91.7-97.1)55.2 (49.2-61.3)82.4 (80.0-84.8)NHW199296.8 (95.9-97.7)92.8 (91.6-94.0)39.2 (36.5-42.0)76.3 (75.1-77.4)NHW199696.7 (95.9-97.6)93.1 (92.1-94.2)38.4 (35.8-41.1)76.1 (75.0-77.2)NHW200096.3 (95.5-97.2)92.4 (91.2-93.6)42.4 (39.6-45.1)77.0 (75.9-78.2)NHW200496.5 (95.6-97.3)93.5 (92.4-94.7)53.5 (50.8-56.1)81.2 (80.0-82.3)NHW200896.4 (95.6-97.3)93.8 (92.7-94.9)58.8 (56.0-61.6)83.0 (81.8-84.2)NHW201295.9 (95.0-96.8)93.9 (92.8-95.0)55.7 (53.0-58.3)81.8 (80.7-83.0)NHW201695.2 (94.3-96.0)93.5 (92.4-94.6)60.4 (57.8-63.0)83.0 (81.9-84.2)NHW201896.7 (95.8-97.6)95.0 (93.95-96.1)60.4 (57.6-63.3)84.0 (82.9-85.3)Figure 2.Trends in 1-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2018.\nUnadjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nAdjusted 1-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nTrends in 1-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2018.\nFor those diagnosed with regional stage CRC, trends in adjusted 1-year cause-specific survival probabilities similarly reflect little change over time, with the possible exception of an imprecisely-estimated positive trend over time among AIAN persons. Although there was little evidence of significant trends over time, there was a clear gradient in the average survival probabilities among those from different race and ethnic groups. AIAN persons had the lowest average survival probability (90.2%, 88.2%-92.2), followed by NHB, NHW, Hispanic, and API persons (93.2%, 92.6%-93.8%; 93.3%, 93.1%-93.5%; 94.2%, 93.8%-94.6%; and 94.8%, 94.4%-95.2%, respectively). All of these estimates were significantly different (all P < .01), except for the comparison between NHB and NHW persons (P = .83).\nThose diagnosed with distant stage CRC experienced the biggest improvements in 1-year cause-specific survival probabilities over time. An increasing trend in 1-year survival probabilities is apparent for persons of all race and ethnic groups. For AIAN and NHB persons, the two groups with lowest initial 1-year survival probabilities, improvements continued at a consistent rate over time. For API, Hispanic, and NHW persons, there appears to be a slowing of the rate of improvement in the second half of the study period. The adjusted time-averaged survival probabilities were low: 46.6% (42.3%-50.9%) for NHB, 48.3% (37.9%-58.7%) for AIAN, 50.5% (48.5%-52.5%) for NHW, 58.1% (54.2%-62.0%) for Hispanic, and 58.3% (54.2%-62.4%) for API persons. All pairwise comparisons among these groups were statistically significant (all P < .01), except for differences between the two groups with the lowest (AIAN vs NHB, P = .87), and between the two groups with the highest (API vs Hispanic, P = .69), average adjusted 1-year cause-specific survival probabilities.", "Estimates of adjusted 5-year cause-specific survival probabilities are shown for all race/ethnicity by stage combinations in Tables 4 and 5. Neither the race/ethnicity by stage by year of diagnosis three-way interaction (P = .90) nor the two-way interaction between the year of diagnosis and race/ethnicity (P = .59) were statistically significant; there is not sufficient evidence to conclude that there are race- or ethnic-specific differences in 5-year survival trends either within or across stages of CRC at diagnosis. There were statistically significant interactions between race/ethnicity and stage (P < .001), and between stage and the year of diagnosis trends (P < .001) with respect to differences in 5-year survival (see Figure 3).Table 4.Unadjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199283.2 (67.0-99.3)58.1 (40.1-76.0)8.5 (0-29.1)49.9 (39.3-60.5)AIAN199587.5 (70.6-100)64.7 (47.7-81.7)13.4 (0-33.0)55.2 (44.9-65.5)AIAN199885.4 (69.9-100)73.1 (57.2-89.0)1.6 (0-18.5)53.3 (44.0-62.6)AIAN200291.8 (79.0-100)70.4 (56.5-84.3)10.8 (0-33.9)57.6 (47.7-67.6)AIAN200597.1 (85.6-100)65.6 (52.5-78.6)23.0 (6.8-39.2)61.9 (54.0-69.8)AIAN200897.5 (84.4-100)83.4 (70.7-96.1)5.9 (0-22.7)62.3 (54.0-70.6)AIAN201189.3 (77.0-100)67.5 (55.3-79.6)9.5 (0.0-22.7)55.4 (48.2-62.7)AIAN201496.5 (86.4-100)71.4 (60.1-82.6)15.1 (2.0-28.2)61.0 (54.3-67.6)API199297.1 (93.4-100)63.9 (57.9-70.0)7.1 (.9-13.2)56.0 (52.9-59.1)API199594.8 (91.1-98.4)67.5 (62.0-73.0)4.9 (0-10.7)55.7 (52.8-58.6)API199896.1 (93.0-99.2)76.8 (71.8-81.8)7.9 (2.5-13.4)60.3 (57.6-62.9)API200296.7 (94.3-99.1)79.8 (75.4-84.1)7.1 (2.5-11.8)61.2 (58.9-63.5)API200596.5 (94.1-98.9)80.0 (75.9-84.1)9.8 (4.7-14.8)62.1 (59.8-64.4)API200897.7 (95.7-99.7)80.6 (76.3-85)9.0 (4.1-13.9)62.4 (60.1-64.7)API201196.7 (94.4-99.0)76.7 (72.5-81.0)8.4 (3.6-13.1)60.6 (58.3-62.9)API201497.2 (95.0-99.4)81.5 (77.6-85.5)8.0 (3.8-12.1)62.2 (60.2-64.3)Hispanic199292.6 (87.7-97.5)58.1 (51.7-64.4)6.7 (0-13.4)52.4 (48.9-55.9)Hispanic199590.7 (85.5-95.8)61.0 (54.6-67.4)5.4 (0-11.4)52.4 (49.0-55.8)Hispanic199894.3 (90.6-98.0)72.7 (67.0-78.3)5.5 (0-11.5)57.5 (54.4-60.5)Hispanic200293.8 (90.1-97.4)75.5 (70.3-80.8)6.0 (.8-11.2)58.4 (55.7-61.2)Hispanic200594.6 (91.6-97.6)74.1 (69.3-78.9)9.3 (4.1-14.5)59.3 (56.8-61.9)Hispanic200896.0 (93.5-98.6)74.0 (69.1-78.9)6.3 (1.6-10.9)58.8 (56.4-61.2)Hispanic201197.6 (95.5-99.6)79 (74.6-83.3)8.9 (4.6-13.1)61.8 (59.6-63.9)Hispanic201496.9 (94.8-99)75.2 (70.9-79.4)9.4 (5.1-13.7)60.5 (58.4-62.6)NHB199290.6 (85.7-95.4)60.9 (54.1-67.7)2.2 (0-6.8)51.2 (48.1-54.4)NHB199591.3 (86.7-95.9)63.8 (57.0-70.6)4.3 (0-9.6)53.1 (49.8-56.4)NHB199896.0 (92.5-99.4)66.4 (60.0-72.9)4.8 (0-9.8)55.7 (52.8-58.7)NHB200289.9 (85.2-94.6)71.6 (65.9-77.3)4.5 (0-9.0)55.3 (52.4-58.2)NHB200591.8 (87.9-95.7)73.5 (67.7-79.3)4.3 (.1-8.4)56.5 (53.8-59.2)NHB200893.8 (90.4-97.1)69.3 (63.3-75.3)5.7 (1.4-10.1)56.3 (53.6-59.0)NHB201193.7 (90.2-97.2)68.7 (62.4-75.0)5.4 (1.2-9.5)55.9 (53.2-58.7)NHB201494.7 (91.4-98.0)69.2 (63.1-75.4)5.9 (1.7-10.1)56.6 (53.9-59.3)NHW199292.4 (91.1-93.7)64.4 (62.2-66.5)4.8 (3.2-6.5)53.9 (52.9-54.9)NHW199591.8 (90.5-93.2)65.6 (63.3-67.8)5.9 (4.2-7.6)54.4 (53.4-55.5)NHW199893.2 (92-94.4)67.8 (65.7-69.9)4.2 (2.7-5.7)55.1 (54.1-56)NHW200293.7 (92.6-94.9)73.4 (71.3-75.5)4.1 (2.7-5.5)57.1 (56.2-58)NHW200595.3 (94.2-96.3)77.7 (75.7-79.7)5.1 (3.5-6.7)59.4 (58.4-60.3)NHW200895.4 (94.4-96.4)77.6 (75.6-79.7)9.0 (7.0-11.1)60.7 (59.7-61.7)NHW201196.1 (95.1-97.2)78.8 (76.7-80.9)4.8 (3.3-6.4)59.9 (59.0-60.9)NHW201496.2 (95.1-97.3)79.3 (77.2-81.4)8.1 (6.2-10.1)61.2 (60.2-62.2)Table 5.Adjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.LocalizedRegionalDistantOverallRace/ethnicityYear of DxSurvival (95% CI)Survival (95% CI)Survival (95% CI)Survival (95% CI)AIAN199278.8 (64.3-93.4)56.6 (40.4-72.7)7.5 (.0-26.0)47.6 (38.1-57.1)AIAN199585.5 (70.3-100)62.1 (46.8-77.4)13.3 (.0-31.0)53.7 (44.4-63.0)AIAN199881.1 (67.1-95.0)70.0 (55.6-84.3)2.8 (.0-18.0)51.3 (42.8-59.7)AIAN200289.4 (77.9-100)68.8 (56.3-81.3)8.2 (.0-29.1)55.5 (46.5-64.5)AIAN200595.6 (85.2-100)63.8 (52.0-75.6)20.4 (5.8-35.0)59.9 (52.7-67.1)AIAN200893.6 (81.8-100)79.8 (68.3-91.2)4.3 (.0-19.5)59.2 (51.8-66.7)AIAN201184.9 (73.8-96.0)65.4 (54.4-76.3)7.3 (.0-19.2)52.5 (46.0-59.1)AIAN201493.0 (84.0-100)68.6 (58.4-78.7)13.3 (1.5-25.1)58.3 (52.3-64.3)API199295.4 (92.1-98.8)63.1 (57.6-68.5)6.5 (.9-12.0)55.0 (52.1-57.9)API199592.3 (88.9-95.7)67.1 (62.1-72.1)6.4 (1.1-11.6)55.3 (52.5-58.0)API199895.2 (92.4-98.1)75.7 (71.2-80.2)8.8 (3.8-13.7)59.9 (57.4-62.4)API200294.5 (92.2-96.7)78.1 (74.2-82.1)7.9 (3.7-12.1)60.2 (58.0-62.3)API200593.7 (91.4-96.0)78.3 (74.6-82.1)10.6 (6.0-15.2)60.9 (58.6-63.1)API200895.3 (93.3-97.2)78.9 (74.9-82.9)10.7 (6.2-15.2)61.6 (59.4-63.8)API201193.2 (90.9-95.4)75.4 (71.5-79.3)9.3 (5.0-13.7)59.3 (57.1-61.5)API201492.6 (90.5-94.7)78.9 (75.2-82.5)11.6 (7.8-15.4)61.0 (59.0-63.0)Hispanic199289.7 (85.3-94.2)57.3 (51.5-63.0)8.6 (2.5-14.8)51.9 (48.6-55.1)Hispanic199588.8 (84.1-93.5)60.2 (54.4-66.0)5.6 (.2-11.0)51.5 (48.4-54.7)Hispanic199891.2 (87.8-94.6)71.2 (66.1-76.4)6.5 (1.1-12.0)56.3 (53.5-59.1)Hispanic200291.0 (87.7-94.4)73.6 (68.8-78.4)5.8 (1.0-10.5)56.8 (54.2-59.4)Hispanic200591.5 (88.7-94.4)72.7 (68.3-77.0)8.8 (4.0-13.5)57.7 (55.2-60.1)Hispanic200892.6 (90.2-95.0)71.6 (67.2-76.1)6.6 (2.3-10.8)57.0 (54.7-59.2)Hispanic201194.6 (92.6-96.6)76.7 (72.7-80.7)8.4 (4.5-12.3)59.9 (57.8-62.0)Hispanic201492.5 (90.4-94.5)72.8 (68.9-76.7)9.6 (5.7-13.6)58.3 (56.2-60.4)NHB199287.5 (83.0-91.9)59.6 (53.4-65.8)1.3 (.0-5.5)49.4 (46.5-52.4)NHB199589.5 (85.2-93.7)62.3 (56.2-68.5)3.4 (.0-8.3)51.7 (48.7-54.8)NHB199892.7 (89.5-95.9)64.6 (58.7-70.4)5.4 (.9-10.0)54.2 (51.5-57.0)NHB200286.8 (82.5-91.0)69.7 (64.5-74.9)4.2 (.0-8.4)53.6 (50.8-56.3)NHB200589.0 (85.4-92.6)71.5 (66.2-76.8)3.7 (.0-7.5)54.7 (52.2-57.3)NHB200890.0 (86.9-93.2)67.7 (62.2-73.1)6.2 (2.1-10.2)54.6 (52.1-57.2)NHB201190.0 (86.7-93.2)66.8 (61.1-72.6)4.0 (.1-7.8)53.6 (51.0-56.2)NHB201490.9 (87.8-94.0)68.0 (62.4-73.6)6.1 (2.2-10.0)55.0 (52.4-57.6)NHW199290.7 (89.3-92.1)65.1 (63.0-67.2)7.3 (5.7-9.0)54.4 (53.2-55.6)NHW199590.5 (89.1-92.0)66.4 (64.3-68.6)8.0 (6.3-9.8)55.0 (53.8-56.2)NHW199891.1 (89.8-92.4)68.6 (66.6-70.7)9.4 (7.8-10.9)56.4 (55.2-57.5)NHW200292.1 (90.8-93.4)73.5 (71.5-75.6)8.7 (7.2-10.2)58.1 (57.0-59.3)NHW200592.3 (91.0-93.5)77.1 (75.1-79.0)10.7 (9.0-12.3)60.0 (58.9-61.1)NHW200892.7 (91.5-93.9)77.1 (75.0-79.1)12.3 (10.3-14.3)60.7 (59.5-61.9)NHW201193.5 (92.2-94.7)78.0 (75.9-80.0)10.7 (9.1-12.3)60.7 (59.6-61.9)NHW201492.3 (91.1-93.6)77.7 (75.7-79.8)13.2 (11.3-15.1)61.1 (59.9-62.3)Figure 3.Trends in 5-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2014.\nUnadjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nAdjusted 5-Year Cause-Specific Survival Probabilities (%), by Race and Ethnicity, Stage, and Year of Diagnosis.\nTrends in 5-year adjusted survival probabilities for individuals from racial and ethnic subgroups diagnosed at different stages of colorectal cancer from 1992 through 2014.\nThe trends reflect improvements in 5-year cause-specific survival probabilities over time for all CRC stages. The rate of improvement in 5-year survival probabilities over time was greatest for those with regional stage CRC, although the rate of improvement appears to be slowing in the second half of the study period. As there are no significantly different trends over time in adjusted 5-year survival probabilities by race or ethnicity, key differences among race and ethnic groups are best summarized by differences in their adjusted time-averaged survival probabilities. For those diagnosed at localized stage CRC, estimates of average adjusted 5-year survival probabilities were 86.9% (84.2%-89.6%) for AIAN, 90.2% (89.4%-91.0%) for NHB, 92.0% (91.8%-92.2%) for NHW, 92.1% (91.5%-92.7%) for Hispanic, and 94.1% (93.5%-94.7%) for API. These differed significantly between all pairs of groups (all P < .04), except for the Hispanic vs NHW comparison (P = .56). For those diagnosed at regional stage CRC, estimates of average adjusted 5-year survival probabilities were 65.1% (62.2%-68.0%) for NHB, 67.5% (61.0%-74.0%) for AIAN, 70.5% (68.1%-72.9%) for Hispanic, 72.5% (71.5%-73.5%) for NHW, and 75.1% (72.9%-77.3%) for API persons. These differed significantly between all pairs of groups (all P < .05), except for the AIAN vs NHB comparison (P = .22). For those diagnosed at distant stage CRC, estimates of average adjusted 5-year survival probabilities were 4.6% (3.6%-5.6%) for NHB, 7.8% (6.6%-9.0%) for Hispanic, 8.5% (7.3%-9.7%) for API, 9.0% (5.5%-12.5%) for AIAN, and 9.6% (9.2%-10.0%) for NHW. The AIAN average did not differ significantly from those of the API, Hispanic, or NHW groups, nor did the API vs Hispanic averages. All others differed significantly (all P < .01).", "The purpose of this study was to estimate and compare race- and ethnicity-specific 1- and 5-year CRC cause-specific survival trends, within the context of the stage at which the cancer was first detected. We utilized data from the SEER registries for persons diagnosed with CRC from 1992 to 2018 and estimated 1- and 5-year survival probabilities within groups of persons with CRC by race and ethnicity, stage of disease, and year of diagnosis. Study findings contribute to existing knowledge gaps regarding trends in survival by including estimates for an often-overlooked population: American Indian or Alaska Natives.\nThere are significant differences in the degree to which improvements in 1-year survival probabilities following stage-specific CRC are experienced according to a person’s race and ethnicity. The estimates that we report specifically for AIAN and API subgroups contribute towards the limited literature focused on identifying survival trend differences for these racially minoritized populations. Study findings suggest that AIAN individuals have the lowest 1-year survival probabilities compared to those of other races. Lower 1-year CRC-specific survival probabilities are also seen among NHB persons. In particular, these persons have the lowest average survival probabilities following the diagnosis of distant-stage CRC. Although differences among racial and ethnic groups have moderated somewhat over the study period, these disparities do not appear to have resolved completely.\nThere is established evidence that CRC screening can prevent or detect CRC early.27,28 For screening to be effective in improving outcomes, timely follow up of any abnormal test is necessary. Structural barriers (eg, lack of insurance or social support, racism and discrimination) to obtaining appropriate primary care services may play an important role in reducing the possibility for racially minoritized populations to receive guideline-compliant screening services.5,29-31 The fact that some studies conducted within the Veterans’ Health Administration found no differences between White and Black persons in diagnostic follow up testing,32,33 suggests that access to appropriate structures and services may play an important role in appropriate post-screening follow-up for minoritized populations. As expected, the greatest differences in survival probabilities are apparent among individuals diagnosed at different stages of CRC. Although these patterns are largely similar among persons from different race and ethnic groups disparities in survival persist. Enhanced follow-up of abnormal results, may help overcome at least some of the persistent disparities in CRC survival probabilities among persons from different race and ethnic groups.\nOther differences in access to and utilization of quality health care may also contribute to the observed differences of survival trends by race.34-36 Some studies have suggested NHB persons are less likely to receive surgical treatment and adjuvant chemotherapy.35-38 Social, structural and political determinants also contribute towards the likelihood of NHB and AIAN persons to be diagnosed with advanced stage CRC compared to NHW persons.39,40 This may further contribute to the lower survival probabilities observed in NHB persons with distant stage CRC.41,42 Racial and ethnic minoritized populations also experience disparities in terms of post-treatment surveillance and distinct baseline comorbidities, which further contribute to lower survival rates.34,35 Several studies continue to highlight that transportation barriers, cultural beliefs, fear and stigma about screening, and concerns about privacy issues are contributing factors to survival outcomes43-47 for persons with CRC.\nLifestyle and biological factors may also play a role in CRC risk and outcomes. For instance, lifestyle factors such as obesity, smoking, alcohol consumption, and physical inactivity have a higher prevalence among Black populations.48 Also, genetic mutations and microsatellite instability can differ among racially and ethnically minoritized populations. All of these things can affect CRC development and prognosis,49-54 and may play into the disparities noted in this work.\nThere are several potential limitations in our analysis. First, we must acknowledge that this is a retrospective analysis of data from the SEER registries. However, SEER is a population-based resource which provides information about the most critical factors of interest, and we tried to control for these. Second, we cannot fully exclude the possibility that regional variations may contribute to the observed CRC survival probability estimates that average across SEER registries. Third, we chose to report on cause-specific survival, and this may be influenced by different practices in cause of death ascertainment. However, in sensitivity analyses performed using relative survival estimates we found that overall findings were largely concordant. This is similar to a report of data from Canada where differences in survival probabilities were noted, and that differences between First Nations and non-aboriginal persons were somewhat smaller for cause-specific survival when compared to relative survival estimates.55 Fourth, SEER data have relatively little information on comorbidities, access to care, and insurance status. Fifth, racial and ethnic classifications in medical records may reflect misclassification,56 which may bias our estimates. Finally, SEER registries represent a subset of AIAN persons spanning the United States, but do not capture data from some regions with large AIAN populations, including those in Oklahoma, Arizona, or the Northern Plains and Great Lakes.57 Even with these limitations, we are able to provide new data concerning trends in CRC survival over time for multiple racial and ethnic groups in the United States, according to their stage at diagnosis.\nIn conclusion, we present trends in 1- and 5-year CRC cause-specific survival probabilities for persons of five racial and ethnic groups. These estimates of, and trends in, survival probabilities for groups of minoritized races and ethnicities may enable the development of a more complete picture of CRC prognosis. We have identified significant differences in the race- and ethnic-specific trends in 1-year stage-specific CRC survival probabilities. In particular, AIAN persons have historically experienced poorer CRC prognosis, as have NHB individuals. Although these disparities appear to be lessening somewhat, the current differences in survival probabilities continue to call for further work in order to erase them completely. Trends in 1-year survival probabilities are significantly different among those diagnosed at different stages of CRC. This, coupled with the finding that the distribution of stages at diagnosis appears to differ among persons of different racial and ethnic groups, suggests that prioritizing specific stages of disease at diagnosis may provide 1 avenue that may help us to overcome race- and ethnic-specific disparities in CRC-specific survival probability. This also argues for a potential need to improve screening for CRC, as this may influence the distribution of stages at which it is diagnosed. Future research should strive to capture cancer incidence and survival information from all key racial and ethnic subgroups. Future efforts should also incorporate and evaluate multi-level interventions at the individual, structural, and policy levels to address the persistent disparities in CRC survival." ]
[ "intro", "methods", null, null, null, "results", null, null, null, "discussion" ]
[ "colorectal cancer", "cause-specific survival", "trends", "minority health", "disparities" ]
Spatial Analysis of Breast Cancer Mortality Rates in a Rural State.
36265079
Breast cancer affects 1 in 8 women in the US and is the most frequently diagnosed cancer in women. In South Dakota, 102 women die from breast cancer each year. We assessed which sociodemographic factors contributed to mortality rates in South Dakota and used spatial analysis to investigate how counties' observed age-adjusted mortality rates compared with expected rates.
INTRODUCTION
We computed standardized incidence ratios (SIRs) of all counties in South Dakota by using the age-adjusted mortality rates, the 2000 US standard population, and the South Dakota estimated population. We used a linear regression model to identify sociodemographic factors associated with breast cancer mortality rates and to compute a new SIR value, after controlling for relevant factors.
METHODS
Educational level and breast cancer incidence rates were significantly associated with breast cancer mortality rates at the county level. The SIR values based on age-adjusted counts showed which counties had more deaths due to breast cancer than what might be expected using South Dakota as the reference population. After controlling for sociodemographic factors, the range of SIR values decreased and had lower variability.
RESULTS
The regression model helped identify factors associated with mortality and provided insights into which risk factors are at play in South Dakota. This information, in combination with the spatial distribution of mortality by county, can be used to help allocate resources to the counties in South Dakota that need them most.
CONCLUSION
[ "Humans", "Female", "Breast Neoplasms", "Incidence", "Rural Population", "Risk Factors", "Spatial Analysis" ]
9616131
Introduction
In South Dakota, breast cancer is the most commonly diagnosed cancer and the second leading cause of cancer death among women (1,2). In 2022, an estimated 750 new cases and 110 deaths attributed to female breast cancer will occur in South Dakota. In general, a woman in the US has a 1-in-8 lifetime risk of developing breast cancer (3,4). Since 1989, the US breast cancer mortality rate has decreased 40%, but from 2010 to 2019 the rate slowed to a low of decreasing by 1.3% per year (5). Characteristics such as age and race and ethnicity affect a woman’s chances of being diagnosed with or dying of breast cancer, but new evidence has established that sociodemographic factors, including education level, also play a role (6). Albano et al noted a negative relationship between number of years of education and cancer mortality and found that the level of education and race vary considerably with mortality rates (7). Of the South Dakota population aged 25 years or older, 92.2% are high school graduates (higher than the national average) and 29.3% have a bachelor’s degree or higher (lower than national average) (8). Olson et al acknowledged that communities exist in which geographic disparities are more prominent because of rural isolation and small population size (9). Furthermore, 64 of 66 counties in South Dakota are categorized as rural or frontier, and South Dakota contains 9 American Indian reservations (10). Finally, 61.6% of women receiving breast services are White, and 16.7% are American Indian (11); most of the population in South Dakota is White, and the leading minority is American Indian at 8.8% (8). The study aimed to describe the spatial distribution of female breast cancer mortality at the county level in South Dakota and assess the association between mortality rates and risk factors reported in the literature.
Methods
[SUBTITLE] Data source [SUBSECTION] The 66 counties of South Dakota have boundaries that are defined by the South Dakota Legislature and accepted by the US Census. The counties range in population from 183,439 in Minnehaha County to 917 in Jones County, and the median population per county is 5,413. Most residents of South Dakota were White; the median percentage of non-White residents by county was 6.6% and the maximum was 95.2% The 66 counties of South Dakota have boundaries that are defined by the South Dakota Legislature and accepted by the US Census. The counties range in population from 183,439 in Minnehaha County to 917 in Jones County, and the median population per county is 5,413. Most residents of South Dakota were White; the median percentage of non-White residents by county was 6.6% and the maximum was 95.2% [SUBTITLE] Cancer data [SUBSECTION] Breast cancer incidence and mortality rates from 2001 through 2015 were extracted from the South Dakota State Cancer Registry provided by the South Dakota Department of Health (12). Both rates were per 100,000 people and age-adjusted to the 2000 US standard population and the South Dakota estimated population. The proportion of mammography screening rates in South Dakota was based on the numbers reported by Holzhauser et al for the All Women Count! mammography program (13). The average number of participants for 1997 through 2016 was reported for the program; then, the average number of participants was adjusted for the total number of women older than 40 years in the county to get an estimated screening rate for each county (13,14). Breast cancer incidence and mortality rates from 2001 through 2015 were extracted from the South Dakota State Cancer Registry provided by the South Dakota Department of Health (12). Both rates were per 100,000 people and age-adjusted to the 2000 US standard population and the South Dakota estimated population. The proportion of mammography screening rates in South Dakota was based on the numbers reported by Holzhauser et al for the All Women Count! mammography program (13). The average number of participants for 1997 through 2016 was reported for the program; then, the average number of participants was adjusted for the total number of women older than 40 years in the county to get an estimated screening rate for each county (13,14). [SUBTITLE] Demographics [SUBSECTION] We used 2015 data from the US Census Bureau to obtain information on the 66 South Dakota counties, including the number of providers and the education level, poverty level, percentage of uninsured, median age, and race of residents (8). Data on educational attainment were obtained from the US Census Bureau’s American Community Survey (ACS). These data were count estimates for the population of each county aged 25 years or older. Levels were categorized as less than 9th grade, 9th through 12th grade with no diploma, high school graduate or equivalent, some college but no degree, associate degree, bachelor’s degree, and graduate or professional degree. These values were modified into an educational attainment statistic of the percentage of the population with less than a bachelor’s degree of education. The statistic used in this study was the percentage of the population with less than a bachelor’s degree, by county. We collected data on poverty estimates, by county, from the ACS; these data adhered to the standards specified by the Office of Management and Budget in Statistical Policy Directive 14 (8). Poverty was determined by a set of income thresholds that consider the living situation (alone or with nonrelatives), age, and number of people per household. For example, the poverty threshold for 2-person families varies by the age of the primary householder and differs from the poverty threshold for people living alone or with nonrelatives, which also varies by age. Insurance coverage percentages were collected from Small Area Health Insurance Estimates (SAHIE) (15). The uninsured percentage included residents who were not covered by insurance, which excluded those on government assistance such as Medicaid or Medicare. Finally, the data set summarizing racial distributions in a county included estimated population counts for American Indian and Alaska Native, Asian, Black or African American, Native Hawaiian or other Pacific Islander, White, other race, and 2 or more races. Because of South Dakota's predominantly White population, the data were configured into White and non-White, which determined the non-White percentage per county (8) (Table 1). We used 2015 data from the US Census Bureau to obtain information on the 66 South Dakota counties, including the number of providers and the education level, poverty level, percentage of uninsured, median age, and race of residents (8). Data on educational attainment were obtained from the US Census Bureau’s American Community Survey (ACS). These data were count estimates for the population of each county aged 25 years or older. Levels were categorized as less than 9th grade, 9th through 12th grade with no diploma, high school graduate or equivalent, some college but no degree, associate degree, bachelor’s degree, and graduate or professional degree. These values were modified into an educational attainment statistic of the percentage of the population with less than a bachelor’s degree of education. The statistic used in this study was the percentage of the population with less than a bachelor’s degree, by county. We collected data on poverty estimates, by county, from the ACS; these data adhered to the standards specified by the Office of Management and Budget in Statistical Policy Directive 14 (8). Poverty was determined by a set of income thresholds that consider the living situation (alone or with nonrelatives), age, and number of people per household. For example, the poverty threshold for 2-person families varies by the age of the primary householder and differs from the poverty threshold for people living alone or with nonrelatives, which also varies by age. Insurance coverage percentages were collected from Small Area Health Insurance Estimates (SAHIE) (15). The uninsured percentage included residents who were not covered by insurance, which excluded those on government assistance such as Medicaid or Medicare. Finally, the data set summarizing racial distributions in a county included estimated population counts for American Indian and Alaska Native, Asian, Black or African American, Native Hawaiian or other Pacific Islander, White, other race, and 2 or more races. Because of South Dakota's predominantly White population, the data were configured into White and non-White, which determined the non-White percentage per county (8) (Table 1). [SUBTITLE] Statistical analysis [SUBSECTION] [SUBTITLE] Data manipulation and missing value imputation [SUBSECTION] Mortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi with its k nearest neighbors and then approximates xi using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi (16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18). Mortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi with its k nearest neighbors and then approximates xi using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi (16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18). [SUBTITLE] Multiple linear regression [SUBSECTION] We used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20). The resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith county and β^0 is the intercept. The variables X 1 i through Xji represent the values of the factors for the ith county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model. The first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM ). We used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20). The resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith county and β^0 is the intercept. The variables X 1 i through Xji represent the values of the factors for the ith county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model. The first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM ). [SUBTITLE] Data manipulation and missing value imputation [SUBSECTION] Mortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi with its k nearest neighbors and then approximates xi using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi (16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18). Mortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi with its k nearest neighbors and then approximates xi using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi (16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18). [SUBTITLE] Multiple linear regression [SUBSECTION] We used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20). The resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith county and β^0 is the intercept. The variables X 1 i through Xji represent the values of the factors for the ith county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model. The first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM ). We used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20). The resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith county and β^0 is the intercept. The variables X 1 i through Xji represent the values of the factors for the ith county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model. The first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM ). [SUBTITLE] Standardized incidence ratio [SUBSECTION] A SIR was used to compare the spatial distribution of counties in terms of mortality rates due to breast cancer. In general, the SIR compares the expected value of deaths to the observed value of deaths in a county. This is calculated with SIRj=OjEj where Oj is the observed value for county j, and Ej is the expected value for county j. A SIR of greater than 1 means that there were more deaths than expected, whereas a SIR of less than 1 means that there were fewer deaths than expected for that county. Because age-specific mortality rates by county were not available, the observed age-adjusted counts per county were computed from the age-adjusted rates per county as follows: Oj*=Oj100,000yj and Ej*=∑j=166Oj100,000·yj·yj∑j=166yj , where Oj is the jth county’s observed age-adjusted mortality rate and yj is the jth county’s population. The expected count was computed to be on the same scale as the observed count. These manipulations were used only to calculate an age-adjusted count SIR, referred to as SIRCOUNT . All other analysis of the data was completed with the original variable (Oj ), as described in the data source. A SIR was calculated on the mortality in each South Dakota county (N = 66). To account for the age adjustment of the data, 2 different SIRs were found: using the age-adjusted mortality count for South Dakota and using the expected rate obtained from the linear regression model. The first SIR accounted only for the age adjustment, and the second SIR accounted for more factors related to breast cancer. For example, the first SIR used the age-adjusted mortality count for a county for both observed and expected (SIRCOUNT ). The second SIR (SIRLM ) used the mortality rates by county, which were per 100,000 persons and age-adjusted to the 2000 US standard population, and the South Dakota estimated population for observed and predicted mortality rate from the linear regression model for expected, which accounted for incidence rates and educational attainment. All statistical analyses used R version 4.1.2 and RStudio version 2021.09.2 build 382 (R Foundation for Statistical Computing) (23,24). A SIR was used to compare the spatial distribution of counties in terms of mortality rates due to breast cancer. In general, the SIR compares the expected value of deaths to the observed value of deaths in a county. This is calculated with SIRj=OjEj where Oj is the observed value for county j, and Ej is the expected value for county j. A SIR of greater than 1 means that there were more deaths than expected, whereas a SIR of less than 1 means that there were fewer deaths than expected for that county. Because age-specific mortality rates by county were not available, the observed age-adjusted counts per county were computed from the age-adjusted rates per county as follows: Oj*=Oj100,000yj and Ej*=∑j=166Oj100,000·yj·yj∑j=166yj , where Oj is the jth county’s observed age-adjusted mortality rate and yj is the jth county’s population. The expected count was computed to be on the same scale as the observed count. These manipulations were used only to calculate an age-adjusted count SIR, referred to as SIRCOUNT . All other analysis of the data was completed with the original variable (Oj ), as described in the data source. A SIR was calculated on the mortality in each South Dakota county (N = 66). To account for the age adjustment of the data, 2 different SIRs were found: using the age-adjusted mortality count for South Dakota and using the expected rate obtained from the linear regression model. The first SIR accounted only for the age adjustment, and the second SIR accounted for more factors related to breast cancer. For example, the first SIR used the age-adjusted mortality count for a county for both observed and expected (SIRCOUNT ). The second SIR (SIRLM ) used the mortality rates by county, which were per 100,000 persons and age-adjusted to the 2000 US standard population, and the South Dakota estimated population for observed and predicted mortality rate from the linear regression model for expected, which accounted for incidence rates and educational attainment. All statistical analyses used R version 4.1.2 and RStudio version 2021.09.2 build 382 (R Foundation for Statistical Computing) (23,24).
Results
[SUBTITLE] Exploratory data analysis [SUBSECTION] To learn more about the data, we performed an Exploratory Data Analysis (EDA) using several techniques. We explored the geographic distribution of the breast cancer mortality rates by using a choropleth map of South Dakota with age-adjusted mortality and incidence rates (Figure 1). The eastern side of the state had lower and more consistent mortality rates, followed by the far western part of the state. The central west part of the state exhibited higher mortality rates. Map A shows the age-adjusted breast cancer mortality rates and Map B shows the age-adjusted breast cancer incidence rates, by county (N = 66), South Dakota, 2001–2015. Counties whose mortality rates have been imputed are marked with a star. Source: South Dakota State Cancer Registry, South Dakota Department of Health (12). Five counties had higher mortality rates than the rest of the counties: Perkins, Mellette, Aurora, Douglas, and Corson. Two counties had lower mortality rates than the rest of the counties: Ziebach and Jackson. Eight counties (Bennet, Buffalo, Corson, Jackson, Mellette, Oglala, Todd, and Ziebach) had a poverty percentage greater than 30%, which was distinctly higher than the others; each county contains land on an American Indian Reservation. Minnehaha and Pennington counties had high screening rates; these counties are home to the first- and second-largest cities in South Dakota, respectively, so they also had the greatest number of screening providers. All variables, besides the number of providers and screening rate, had a correlation greater than zero with mortality rate. Incidence, educational attainment, and uninsured percentage all had a low positive correlation with mortality rate. The highest correlation with mortality rate was education attainment, at a value of 0.26. The remaining variables had a negligible correlation with mortality rate. To learn more about the data, we performed an Exploratory Data Analysis (EDA) using several techniques. We explored the geographic distribution of the breast cancer mortality rates by using a choropleth map of South Dakota with age-adjusted mortality and incidence rates (Figure 1). The eastern side of the state had lower and more consistent mortality rates, followed by the far western part of the state. The central west part of the state exhibited higher mortality rates. Map A shows the age-adjusted breast cancer mortality rates and Map B shows the age-adjusted breast cancer incidence rates, by county (N = 66), South Dakota, 2001–2015. Counties whose mortality rates have been imputed are marked with a star. Source: South Dakota State Cancer Registry, South Dakota Department of Health (12). Five counties had higher mortality rates than the rest of the counties: Perkins, Mellette, Aurora, Douglas, and Corson. Two counties had lower mortality rates than the rest of the counties: Ziebach and Jackson. Eight counties (Bennet, Buffalo, Corson, Jackson, Mellette, Oglala, Todd, and Ziebach) had a poverty percentage greater than 30%, which was distinctly higher than the others; each county contains land on an American Indian Reservation. Minnehaha and Pennington counties had high screening rates; these counties are home to the first- and second-largest cities in South Dakota, respectively, so they also had the greatest number of screening providers. All variables, besides the number of providers and screening rate, had a correlation greater than zero with mortality rate. Incidence, educational attainment, and uninsured percentage all had a low positive correlation with mortality rate. The highest correlation with mortality rate was education attainment, at a value of 0.26. The remaining variables had a negligible correlation with mortality rate. [SUBTITLE] Regression analysis [SUBSECTION] Model 2 had the highest adjusted R 2 value (0.10) and the lowest AIC (441.79), with 2 significant factors associated with mortality rate; Model 3 had the lowest adjusted R 2 (.004) and the highest AIC (453.32), with no significant predictors of mortality. In Model 2, breast cancer incidence and educational attainment were predictors of breast cancer mortality, indicating that as more people are diagnosed with breast cancer and as the percentage of people with less than a bachelor’s degree increases, breast cancer mortality rate increases (Table 2). Educational attainment was a predictor of mortality in all models, and the educational attainment statistic had a P < .001. Abbreviation: — , not applicable; AIC, Akaike information criterion. P = .01. P = .001. Model 2 had the highest adjusted R 2 value (0.10) and the lowest AIC (441.79), with 2 significant factors associated with mortality rate; Model 3 had the lowest adjusted R 2 (.004) and the highest AIC (453.32), with no significant predictors of mortality. In Model 2, breast cancer incidence and educational attainment were predictors of breast cancer mortality, indicating that as more people are diagnosed with breast cancer and as the percentage of people with less than a bachelor’s degree increases, breast cancer mortality rate increases (Table 2). Educational attainment was a predictor of mortality in all models, and the educational attainment statistic had a P < .001. Abbreviation: — , not applicable; AIC, Akaike information criterion. P = .01. P = .001. [SUBTITLE] Standardized incidence ratio [SUBSECTION] Thirty-five of the 66 counties had a SIRCOUNT greater than 1, meaning that more than half of the counties had more deaths than expected (Figure 2). The 5 counties with the highest SIRCOUNT , in decreasing order, were Perkins, Mellette, Aurora, Douglas, and Corson. Of those, Perkins, Mellette, and Aurora had more than twice the expected number of deaths. Ziebach, Jackson, Davison, Tripp, and Meade counties had the lowest SIRCOUNT , with Ziebach and Jackson both being less than half the expected number of deaths. The highest SIRCOUNT was 2.15 and the lowest was 0.31. Map A shows the predicted breast cancer mortality rate of South Dakota counties, accounting for age-adjustment of the data, and Map B shows the predicted breast cancer mortality rate of South Dakota counties, accounting for age-adjustment, incidence rate, and educational attainment. Abbreviation: SIR, standardized incidence ratio. Map Sources: South Dakota State Cancer Registry, South Dakota Department of Health (12), Holzhauser et al (13), and the US Census Bureau (8). On the other hand, for the SIRLM , most counties had fewer deaths than expected, with 28 of 66 counties having a SIRLM greater than 1. The 5 counties with the highest SIRLM were similar to the SIRCOUNT : Corson, Perkins, Aurora, Jones, and Mellette in decreasing order. No county had more than twice the expected number of deaths. The 5 counties with the lowest SIRLM were Ziebach, Jackson, Tripp, Oglala Lakota, and Davison. Ziebach and Jackson counties had less than half the number of expected deaths. The highest SIRLM was 1.94 and the lowest SIRLM was 0.35. The results of the SIRs (SIRCOUNT and SIRLM ) are presented in Figure 2. The eastern side of the state showed similar SIR values while the western side of the state had more variation. Perkins, Mellette, Aurora, Douglas, and Corson counties had SIRCOUNT values that were much higher than those of the rest of the counties. Ziebach and Jackson counties had the lowest SIRCOUNT values. These counties with the highest SIRCOUNT made up 5 of the 6 counties with the highest SIRLM values, with Jones County replacing Douglas County. The 2 counties with the lowest SIRCOUNT values were the same 2 counties with the lowest SIRLM values. Thirty-five of the 66 counties had a SIRCOUNT greater than 1, meaning that more than half of the counties had more deaths than expected (Figure 2). The 5 counties with the highest SIRCOUNT , in decreasing order, were Perkins, Mellette, Aurora, Douglas, and Corson. Of those, Perkins, Mellette, and Aurora had more than twice the expected number of deaths. Ziebach, Jackson, Davison, Tripp, and Meade counties had the lowest SIRCOUNT , with Ziebach and Jackson both being less than half the expected number of deaths. The highest SIRCOUNT was 2.15 and the lowest was 0.31. Map A shows the predicted breast cancer mortality rate of South Dakota counties, accounting for age-adjustment of the data, and Map B shows the predicted breast cancer mortality rate of South Dakota counties, accounting for age-adjustment, incidence rate, and educational attainment. Abbreviation: SIR, standardized incidence ratio. Map Sources: South Dakota State Cancer Registry, South Dakota Department of Health (12), Holzhauser et al (13), and the US Census Bureau (8). On the other hand, for the SIRLM , most counties had fewer deaths than expected, with 28 of 66 counties having a SIRLM greater than 1. The 5 counties with the highest SIRLM were similar to the SIRCOUNT : Corson, Perkins, Aurora, Jones, and Mellette in decreasing order. No county had more than twice the expected number of deaths. The 5 counties with the lowest SIRLM were Ziebach, Jackson, Tripp, Oglala Lakota, and Davison. Ziebach and Jackson counties had less than half the number of expected deaths. The highest SIRLM was 1.94 and the lowest SIRLM was 0.35. The results of the SIRs (SIRCOUNT and SIRLM ) are presented in Figure 2. The eastern side of the state showed similar SIR values while the western side of the state had more variation. Perkins, Mellette, Aurora, Douglas, and Corson counties had SIRCOUNT values that were much higher than those of the rest of the counties. Ziebach and Jackson counties had the lowest SIRCOUNT values. These counties with the highest SIRCOUNT made up 5 of the 6 counties with the highest SIRLM values, with Jones County replacing Douglas County. The 2 counties with the lowest SIRCOUNT values were the same 2 counties with the lowest SIRLM values.
null
null
[ "Data source", "Cancer data", "Demographics", "Statistical analysis", "Data manipulation and missing value imputation", "Multiple linear regression", "Standardized incidence ratio", "Exploratory data analysis", "Regression analysis", "Standardized incidence ratio" ]
[ "The 66 counties of South Dakota have boundaries that are defined by the South Dakota Legislature and accepted by the US Census. The counties range in population from 183,439 in Minnehaha County to 917 in Jones County, and the median population per county is 5,413. Most residents of South Dakota were White; the median percentage of non-White residents by county was 6.6% and the maximum was 95.2%", "Breast cancer incidence and mortality rates from 2001 through 2015 were extracted from the South Dakota State Cancer Registry provided by the South Dakota Department of Health (12). Both rates were per 100,000 people and age-adjusted to the 2000 US standard population and the South Dakota estimated population. The proportion of mammography screening rates in South Dakota was based on the numbers reported by Holzhauser et al for the All Women Count! mammography program (13). The average number of participants for 1997 through 2016 was reported for the program; then, the average number of participants was adjusted for the total number of women older than 40 years in the county to get an estimated screening rate for each county (13,14).", "We used 2015 data from the US Census Bureau to obtain information on the 66 South Dakota counties, including the number of providers and the education level, poverty level, percentage of uninsured, median age, and race of residents (8).\nData on educational attainment were obtained from the US Census Bureau’s American Community Survey (ACS). These data were count estimates for the population of each county aged 25 years or older. Levels were categorized as less than 9th grade, 9th through 12th grade with no diploma, high school graduate or equivalent, some college but no degree, associate degree, bachelor’s degree, and graduate or professional degree. These values were modified into an educational attainment statistic of the percentage of the population with less than a bachelor’s degree of education. The statistic used in this study was the percentage of the population with less than a bachelor’s degree, by county.\nWe collected data on poverty estimates, by county, from the ACS; these data adhered to the standards specified by the Office of Management and Budget in Statistical Policy Directive 14 (8). Poverty was determined by a set of income thresholds that consider the living situation (alone or with nonrelatives), age, and number of people per household. For example, the poverty threshold for 2-person families varies by the age of the primary householder and differs from the poverty threshold for people living alone or with nonrelatives, which also varies by age. \nInsurance coverage percentages were collected from Small Area Health Insurance Estimates (SAHIE) (15). The uninsured percentage included residents who were not covered by insurance, which excluded those on government assistance such as Medicaid or Medicare. Finally, the data set summarizing racial distributions in a county included estimated population counts for American Indian and Alaska Native, Asian, Black or African American, Native Hawaiian or other Pacific Islander, White, other race, and 2 or more races. Because of South Dakota's predominantly White population, the data were configured into White and non-White, which determined the non-White percentage per county (8) (Table 1).", "[SUBTITLE] Data manipulation and missing value imputation [SUBSECTION] Mortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi\n with its k nearest neighbors and then approximates xi\n using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi \n(16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18).\nMortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi\n with its k nearest neighbors and then approximates xi\n using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi \n(16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18).\n[SUBTITLE] Multiple linear regression [SUBSECTION] We used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20).\nThe resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith\n county and β^0 is the intercept. The variables X\n1\n\ni \nthrough Xji\n represent the values of the factors for the ith\n county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model.\nThe first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM\n).\nWe used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20).\nThe resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith\n county and β^0 is the intercept. The variables X\n1\n\ni \nthrough Xji\n represent the values of the factors for the ith\n county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model.\nThe first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM\n).", "Mortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi\n with its k nearest neighbors and then approximates xi\n using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi \n(16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18).", "We used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20).\nThe resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith\n county and β^0 is the intercept. The variables X\n1\n\ni \nthrough Xji\n represent the values of the factors for the ith\n county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model.\nThe first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM\n).", "A SIR was used to compare the spatial distribution of counties in terms of mortality rates due to breast cancer. In general, the SIR compares the expected value of deaths to the observed value of deaths in a county. This is calculated with SIRj=OjEj where Oj\n is the observed value for county j, and Ej\n is the expected value for county j. A SIR of greater than 1 means that there were more deaths than expected, whereas a SIR of less than 1 means that there were fewer deaths than expected for that county.\nBecause age-specific mortality rates by county were not available, the observed age-adjusted counts per county were computed from the age-adjusted rates per county as follows: Oj*=Oj100,000yj and Ej*=∑j=166Oj100,000·yj·yj∑j=166yj , where Oj\n is the jth\n county’s observed age-adjusted mortality rate and yj\n is the jth\n county’s population. The expected count was computed to be on the same scale as the observed count. These manipulations were used only to calculate an age-adjusted count SIR, referred to as SIRCOUNT\n. All other analysis of the data was completed with the original variable (Oj\n), as described in the data source.\nA SIR was calculated on the mortality in each South Dakota county (N = 66). To account for the age adjustment of the data, 2 different SIRs were found: using the age-adjusted mortality count for South Dakota and using the expected rate obtained from the linear regression model. The first SIR accounted only for the age adjustment, and the second SIR accounted for more factors related to breast cancer. For example, the first SIR used the age-adjusted mortality count for a county for both observed and expected (SIRCOUNT\n). The second SIR (SIRLM\n) used the mortality rates by county, which were per 100,000 persons and age-adjusted to the 2000 US standard population, and the South Dakota estimated population for observed and predicted mortality rate from the linear regression model for expected, which accounted for incidence rates and educational attainment. All statistical analyses used R version 4.1.2 and RStudio version 2021.09.2 build 382 (R Foundation for Statistical Computing) (23,24).", "To learn more about the data, we performed an Exploratory Data Analysis (EDA) using several techniques. We explored the geographic distribution of the breast cancer mortality rates by using a choropleth map of South Dakota with age-adjusted mortality and incidence rates (Figure 1). The eastern side of the state had lower and more consistent mortality rates, followed by the far western part of the state. The central west part of the state exhibited higher mortality rates.\nMap A shows the age-adjusted breast cancer mortality rates and Map B shows the age-adjusted breast cancer incidence rates, by county (N = 66), South Dakota, 2001–2015. Counties whose mortality rates have been imputed are marked with a star. Source: South Dakota State Cancer Registry, South Dakota Department of Health (12).\nFive counties had higher mortality rates than the rest of the counties: Perkins, Mellette, Aurora, Douglas, and Corson. Two counties had lower mortality rates than the rest of the counties: Ziebach and Jackson. Eight counties (Bennet, Buffalo, Corson, Jackson, Mellette, Oglala, Todd, and Ziebach) had a poverty percentage greater than 30%, which was distinctly higher than the others; each county contains land on an American Indian Reservation. Minnehaha and Pennington counties had high screening rates; these counties are home to the first- and second-largest cities in South Dakota, respectively, so they also had the greatest number of screening providers.\nAll variables, besides the number of providers and screening rate, had a correlation greater than zero with mortality rate. Incidence, educational attainment, and uninsured percentage all had a low positive correlation with mortality rate. The highest correlation with mortality rate was education attainment, at a value of 0.26. The remaining variables had a negligible correlation with mortality rate.", "Model 2 had the highest adjusted R\n2 value (0.10) and the lowest AIC (441.79), with 2 significant factors associated with mortality rate; Model 3 had the lowest adjusted R\n2 (.004) and the highest AIC (453.32), with no significant predictors of mortality. In Model 2, breast cancer incidence and educational attainment were predictors of breast cancer mortality, indicating that as more people are diagnosed with breast cancer and as the percentage of people with less than a bachelor’s degree increases, breast cancer mortality rate increases (Table 2). Educational attainment was a predictor of mortality in all models, and the educational attainment statistic had a P < .001. \nAbbreviation: — , not applicable; AIC, Akaike information criterion.\n\nP = .01.\n\nP = .001.", "Thirty-five of the 66 counties had a SIRCOUNT\n greater than 1, meaning that more than half of the counties had more deaths than expected (Figure 2). The 5 counties with the highest SIRCOUNT\n, in decreasing order, were Perkins, Mellette, Aurora, Douglas, and Corson. Of those, Perkins, Mellette, and Aurora had more than twice the expected number of deaths. Ziebach, Jackson, Davison, Tripp, and Meade counties had the lowest SIRCOUNT\n, with Ziebach and Jackson both being less than half the expected number of deaths. The highest SIRCOUNT\n was 2.15 and the lowest was 0.31.\nMap A shows the predicted breast cancer mortality rate of South Dakota counties, accounting for age-adjustment of the data, and Map B shows the predicted breast cancer mortality rate of South Dakota counties, accounting for age-adjustment, incidence rate, and educational attainment. Abbreviation: SIR, standardized incidence ratio. Map Sources: South Dakota State Cancer Registry, South Dakota Department of Health (12), Holzhauser et al (13), and the US Census Bureau (8).\nOn the other hand, for the SIRLM\n, most counties had fewer deaths than expected, with 28 of 66 counties having a SIRLM\n greater than 1. The 5 counties with the highest SIRLM\n were similar to the SIRCOUNT\n: Corson, Perkins, Aurora, Jones, and Mellette in decreasing order. No county had more than twice the expected number of deaths. The 5 counties with the lowest SIRLM\n were Ziebach, Jackson, Tripp, Oglala Lakota, and Davison. Ziebach and Jackson counties had less than half the number of expected deaths. The highest SIRLM\n was 1.94 and the lowest SIRLM\n was 0.35. \nThe results of the SIRs (SIRCOUNT\n and SIRLM\n) are presented in Figure 2. The eastern side of the state showed similar SIR values while the western side of the state had more variation. Perkins, Mellette, Aurora, Douglas, and Corson counties had SIRCOUNT\n values that were much higher than those of the rest of the counties. Ziebach and Jackson counties had the lowest SIRCOUNT\n values. These counties with the highest SIRCOUNT\n made up 5 of the 6 counties with the highest SIRLM\n values, with Jones County replacing Douglas County. The 2 counties with the lowest SIRCOUNT\n values were the same 2 counties with the lowest SIRLM\n values." ]
[ null, null, null, null, null, null, null, null, null, null ]
[ "What is known on this topic?", "What is added by this report?", "What are the implications for public health practice?", "Introduction", "Methods", "Data source", "Cancer data", "Demographics", "Statistical analysis", "Data manipulation and missing value imputation", "Multiple linear regression", "Standardized incidence ratio", "Results", "Exploratory data analysis", "Regression analysis", "Standardized incidence ratio", "Discussion" ]
[ "Breast cancer affects 1 in 8 women in the US and is the most frequently diagnosed cancer in women.", "Because South Dakota is a rural state, sociodemographic factors affect the population differently than in the general US population. We assessed the spatial distribution of breast cancer mortality rates by county, and our findings add insight on educational attainment as a risk factor for breast cancer.", "Our results can be used to help allocate resources to the South Dakota counties that need them most.", "In South Dakota, breast cancer is the most commonly diagnosed cancer and the second leading cause of cancer death among women (1,2). In 2022, an estimated 750 new cases and 110 deaths attributed to female breast cancer will occur in South Dakota. In general, a woman in the US has a 1-in-8 lifetime risk of developing breast cancer (3,4). Since 1989, the US breast cancer mortality rate has decreased 40%, but from 2010 to 2019 the rate slowed to a low of decreasing by 1.3% per year (5).\nCharacteristics such as age and race and ethnicity affect a woman’s chances of being diagnosed with or dying of breast cancer, but new evidence has established that sociodemographic factors, including education level, also play a role (6). Albano et al noted a negative relationship between number of years of education and cancer mortality and found that the level of education and race vary considerably with mortality rates (7). Of the South Dakota population aged 25 years or older, 92.2% are high school graduates (higher than the national average) and 29.3% have a bachelor’s degree or higher (lower than national average) (8). Olson et al acknowledged that communities exist in which geographic disparities are more prominent because of rural isolation and small population size (9). Furthermore, 64 of 66 counties in South Dakota are categorized as rural or frontier, and South Dakota contains 9 American Indian reservations (10). Finally, 61.6% of women receiving breast services are White, and 16.7% are American Indian (11); most of the population in South Dakota is White, and the leading minority is American Indian at 8.8% (8).\nThe study aimed to describe the spatial distribution of female breast cancer mortality at the county level in South Dakota and assess the association between mortality rates and risk factors reported in the literature.", "[SUBTITLE] Data source [SUBSECTION] The 66 counties of South Dakota have boundaries that are defined by the South Dakota Legislature and accepted by the US Census. The counties range in population from 183,439 in Minnehaha County to 917 in Jones County, and the median population per county is 5,413. Most residents of South Dakota were White; the median percentage of non-White residents by county was 6.6% and the maximum was 95.2%\nThe 66 counties of South Dakota have boundaries that are defined by the South Dakota Legislature and accepted by the US Census. The counties range in population from 183,439 in Minnehaha County to 917 in Jones County, and the median population per county is 5,413. Most residents of South Dakota were White; the median percentage of non-White residents by county was 6.6% and the maximum was 95.2%\n[SUBTITLE] Cancer data [SUBSECTION] Breast cancer incidence and mortality rates from 2001 through 2015 were extracted from the South Dakota State Cancer Registry provided by the South Dakota Department of Health (12). Both rates were per 100,000 people and age-adjusted to the 2000 US standard population and the South Dakota estimated population. The proportion of mammography screening rates in South Dakota was based on the numbers reported by Holzhauser et al for the All Women Count! mammography program (13). The average number of participants for 1997 through 2016 was reported for the program; then, the average number of participants was adjusted for the total number of women older than 40 years in the county to get an estimated screening rate for each county (13,14).\nBreast cancer incidence and mortality rates from 2001 through 2015 were extracted from the South Dakota State Cancer Registry provided by the South Dakota Department of Health (12). Both rates were per 100,000 people and age-adjusted to the 2000 US standard population and the South Dakota estimated population. The proportion of mammography screening rates in South Dakota was based on the numbers reported by Holzhauser et al for the All Women Count! mammography program (13). The average number of participants for 1997 through 2016 was reported for the program; then, the average number of participants was adjusted for the total number of women older than 40 years in the county to get an estimated screening rate for each county (13,14).\n[SUBTITLE] Demographics [SUBSECTION] We used 2015 data from the US Census Bureau to obtain information on the 66 South Dakota counties, including the number of providers and the education level, poverty level, percentage of uninsured, median age, and race of residents (8).\nData on educational attainment were obtained from the US Census Bureau’s American Community Survey (ACS). These data were count estimates for the population of each county aged 25 years or older. Levels were categorized as less than 9th grade, 9th through 12th grade with no diploma, high school graduate or equivalent, some college but no degree, associate degree, bachelor’s degree, and graduate or professional degree. These values were modified into an educational attainment statistic of the percentage of the population with less than a bachelor’s degree of education. The statistic used in this study was the percentage of the population with less than a bachelor’s degree, by county.\nWe collected data on poverty estimates, by county, from the ACS; these data adhered to the standards specified by the Office of Management and Budget in Statistical Policy Directive 14 (8). Poverty was determined by a set of income thresholds that consider the living situation (alone or with nonrelatives), age, and number of people per household. For example, the poverty threshold for 2-person families varies by the age of the primary householder and differs from the poverty threshold for people living alone or with nonrelatives, which also varies by age. \nInsurance coverage percentages were collected from Small Area Health Insurance Estimates (SAHIE) (15). The uninsured percentage included residents who were not covered by insurance, which excluded those on government assistance such as Medicaid or Medicare. Finally, the data set summarizing racial distributions in a county included estimated population counts for American Indian and Alaska Native, Asian, Black or African American, Native Hawaiian or other Pacific Islander, White, other race, and 2 or more races. Because of South Dakota's predominantly White population, the data were configured into White and non-White, which determined the non-White percentage per county (8) (Table 1).\nWe used 2015 data from the US Census Bureau to obtain information on the 66 South Dakota counties, including the number of providers and the education level, poverty level, percentage of uninsured, median age, and race of residents (8).\nData on educational attainment were obtained from the US Census Bureau’s American Community Survey (ACS). These data were count estimates for the population of each county aged 25 years or older. Levels were categorized as less than 9th grade, 9th through 12th grade with no diploma, high school graduate or equivalent, some college but no degree, associate degree, bachelor’s degree, and graduate or professional degree. These values were modified into an educational attainment statistic of the percentage of the population with less than a bachelor’s degree of education. The statistic used in this study was the percentage of the population with less than a bachelor’s degree, by county.\nWe collected data on poverty estimates, by county, from the ACS; these data adhered to the standards specified by the Office of Management and Budget in Statistical Policy Directive 14 (8). Poverty was determined by a set of income thresholds that consider the living situation (alone or with nonrelatives), age, and number of people per household. For example, the poverty threshold for 2-person families varies by the age of the primary householder and differs from the poverty threshold for people living alone or with nonrelatives, which also varies by age. \nInsurance coverage percentages were collected from Small Area Health Insurance Estimates (SAHIE) (15). The uninsured percentage included residents who were not covered by insurance, which excluded those on government assistance such as Medicaid or Medicare. Finally, the data set summarizing racial distributions in a county included estimated population counts for American Indian and Alaska Native, Asian, Black or African American, Native Hawaiian or other Pacific Islander, White, other race, and 2 or more races. Because of South Dakota's predominantly White population, the data were configured into White and non-White, which determined the non-White percentage per county (8) (Table 1).\n[SUBTITLE] Statistical analysis [SUBSECTION] [SUBTITLE] Data manipulation and missing value imputation [SUBSECTION] Mortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi\n with its k nearest neighbors and then approximates xi\n using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi \n(16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18).\nMortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi\n with its k nearest neighbors and then approximates xi\n using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi \n(16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18).\n[SUBTITLE] Multiple linear regression [SUBSECTION] We used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20).\nThe resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith\n county and β^0 is the intercept. The variables X\n1\n\ni \nthrough Xji\n represent the values of the factors for the ith\n county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model.\nThe first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM\n).\nWe used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20).\nThe resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith\n county and β^0 is the intercept. The variables X\n1\n\ni \nthrough Xji\n represent the values of the factors for the ith\n county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model.\nThe first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM\n).\n[SUBTITLE] Data manipulation and missing value imputation [SUBSECTION] Mortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi\n with its k nearest neighbors and then approximates xi\n using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi \n(16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18).\nMortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi\n with its k nearest neighbors and then approximates xi\n using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi \n(16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18).\n[SUBTITLE] Multiple linear regression [SUBSECTION] We used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20).\nThe resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith\n county and β^0 is the intercept. The variables X\n1\n\ni \nthrough Xji\n represent the values of the factors for the ith\n county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model.\nThe first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM\n).\nWe used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20).\nThe resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith\n county and β^0 is the intercept. The variables X\n1\n\ni \nthrough Xji\n represent the values of the factors for the ith\n county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model.\nThe first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM\n).\n[SUBTITLE] Standardized incidence ratio [SUBSECTION] A SIR was used to compare the spatial distribution of counties in terms of mortality rates due to breast cancer. In general, the SIR compares the expected value of deaths to the observed value of deaths in a county. This is calculated with SIRj=OjEj where Oj\n is the observed value for county j, and Ej\n is the expected value for county j. A SIR of greater than 1 means that there were more deaths than expected, whereas a SIR of less than 1 means that there were fewer deaths than expected for that county.\nBecause age-specific mortality rates by county were not available, the observed age-adjusted counts per county were computed from the age-adjusted rates per county as follows: Oj*=Oj100,000yj and Ej*=∑j=166Oj100,000·yj·yj∑j=166yj , where Oj\n is the jth\n county’s observed age-adjusted mortality rate and yj\n is the jth\n county’s population. The expected count was computed to be on the same scale as the observed count. These manipulations were used only to calculate an age-adjusted count SIR, referred to as SIRCOUNT\n. All other analysis of the data was completed with the original variable (Oj\n), as described in the data source.\nA SIR was calculated on the mortality in each South Dakota county (N = 66). To account for the age adjustment of the data, 2 different SIRs were found: using the age-adjusted mortality count for South Dakota and using the expected rate obtained from the linear regression model. The first SIR accounted only for the age adjustment, and the second SIR accounted for more factors related to breast cancer. For example, the first SIR used the age-adjusted mortality count for a county for both observed and expected (SIRCOUNT\n). The second SIR (SIRLM\n) used the mortality rates by county, which were per 100,000 persons and age-adjusted to the 2000 US standard population, and the South Dakota estimated population for observed and predicted mortality rate from the linear regression model for expected, which accounted for incidence rates and educational attainment. All statistical analyses used R version 4.1.2 and RStudio version 2021.09.2 build 382 (R Foundation for Statistical Computing) (23,24).\nA SIR was used to compare the spatial distribution of counties in terms of mortality rates due to breast cancer. In general, the SIR compares the expected value of deaths to the observed value of deaths in a county. This is calculated with SIRj=OjEj where Oj\n is the observed value for county j, and Ej\n is the expected value for county j. A SIR of greater than 1 means that there were more deaths than expected, whereas a SIR of less than 1 means that there were fewer deaths than expected for that county.\nBecause age-specific mortality rates by county were not available, the observed age-adjusted counts per county were computed from the age-adjusted rates per county as follows: Oj*=Oj100,000yj and Ej*=∑j=166Oj100,000·yj·yj∑j=166yj , where Oj\n is the jth\n county’s observed age-adjusted mortality rate and yj\n is the jth\n county’s population. The expected count was computed to be on the same scale as the observed count. These manipulations were used only to calculate an age-adjusted count SIR, referred to as SIRCOUNT\n. All other analysis of the data was completed with the original variable (Oj\n), as described in the data source.\nA SIR was calculated on the mortality in each South Dakota county (N = 66). To account for the age adjustment of the data, 2 different SIRs were found: using the age-adjusted mortality count for South Dakota and using the expected rate obtained from the linear regression model. The first SIR accounted only for the age adjustment, and the second SIR accounted for more factors related to breast cancer. For example, the first SIR used the age-adjusted mortality count for a county for both observed and expected (SIRCOUNT\n). The second SIR (SIRLM\n) used the mortality rates by county, which were per 100,000 persons and age-adjusted to the 2000 US standard population, and the South Dakota estimated population for observed and predicted mortality rate from the linear regression model for expected, which accounted for incidence rates and educational attainment. All statistical analyses used R version 4.1.2 and RStudio version 2021.09.2 build 382 (R Foundation for Statistical Computing) (23,24).", "The 66 counties of South Dakota have boundaries that are defined by the South Dakota Legislature and accepted by the US Census. The counties range in population from 183,439 in Minnehaha County to 917 in Jones County, and the median population per county is 5,413. Most residents of South Dakota were White; the median percentage of non-White residents by county was 6.6% and the maximum was 95.2%", "Breast cancer incidence and mortality rates from 2001 through 2015 were extracted from the South Dakota State Cancer Registry provided by the South Dakota Department of Health (12). Both rates were per 100,000 people and age-adjusted to the 2000 US standard population and the South Dakota estimated population. The proportion of mammography screening rates in South Dakota was based on the numbers reported by Holzhauser et al for the All Women Count! mammography program (13). The average number of participants for 1997 through 2016 was reported for the program; then, the average number of participants was adjusted for the total number of women older than 40 years in the county to get an estimated screening rate for each county (13,14).", "We used 2015 data from the US Census Bureau to obtain information on the 66 South Dakota counties, including the number of providers and the education level, poverty level, percentage of uninsured, median age, and race of residents (8).\nData on educational attainment were obtained from the US Census Bureau’s American Community Survey (ACS). These data were count estimates for the population of each county aged 25 years or older. Levels were categorized as less than 9th grade, 9th through 12th grade with no diploma, high school graduate or equivalent, some college but no degree, associate degree, bachelor’s degree, and graduate or professional degree. These values were modified into an educational attainment statistic of the percentage of the population with less than a bachelor’s degree of education. The statistic used in this study was the percentage of the population with less than a bachelor’s degree, by county.\nWe collected data on poverty estimates, by county, from the ACS; these data adhered to the standards specified by the Office of Management and Budget in Statistical Policy Directive 14 (8). Poverty was determined by a set of income thresholds that consider the living situation (alone or with nonrelatives), age, and number of people per household. For example, the poverty threshold for 2-person families varies by the age of the primary householder and differs from the poverty threshold for people living alone or with nonrelatives, which also varies by age. \nInsurance coverage percentages were collected from Small Area Health Insurance Estimates (SAHIE) (15). The uninsured percentage included residents who were not covered by insurance, which excluded those on government assistance such as Medicaid or Medicare. Finally, the data set summarizing racial distributions in a county included estimated population counts for American Indian and Alaska Native, Asian, Black or African American, Native Hawaiian or other Pacific Islander, White, other race, and 2 or more races. Because of South Dakota's predominantly White population, the data were configured into White and non-White, which determined the non-White percentage per county (8) (Table 1).", "[SUBTITLE] Data manipulation and missing value imputation [SUBSECTION] Mortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi\n with its k nearest neighbors and then approximates xi\n using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi \n(16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18).\nMortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi\n with its k nearest neighbors and then approximates xi\n using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi \n(16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18).\n[SUBTITLE] Multiple linear regression [SUBSECTION] We used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20).\nThe resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith\n county and β^0 is the intercept. The variables X\n1\n\ni \nthrough Xji\n represent the values of the factors for the ith\n county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model.\nThe first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM\n).\nWe used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20).\nThe resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith\n county and β^0 is the intercept. The variables X\n1\n\ni \nthrough Xji\n represent the values of the factors for the ith\n county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model.\nThe first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM\n).", "Mortality rates and the various independent variables were combined into 1 data set; 15 of the 66 counties were missing mortality rate data. Mortality rates are often suppressed from public availability when 3 or fewer deaths are reported in a county, to protect patient identity. To remedy the missing data, k-nearest-neighbor (KNN) imputation was used to estimate the missing mortality values. KNN imputation compares a data point xi\n with its k nearest neighbors and then approximates xi\n using the majority vote of these k neighbors in multidimensional space. For the data, k = 9 nearest neighbors were used, and a weighted mean of the k nearest values was placed for each missing xi \n(16,17). This was done with the function “knn()” from the R package VIM version 6.1.1 (R Foundation for Statistical Computing) (18).", "We used multiple linear regression to model the relationship between the factors in this study and breast cancer mortality rates (19). We considered several potential predictor variables with observed correlation, hence a stepwise variable selection technique was used, in both the forward and backward direction, to perform feature selection. As a result, a subset of the factors that were associated with the mortality rates was obtained based on Akaike information criterion (AIC) (19,20).\nThe resulting model is of the form Yi ^= β^0 + β^1X1i+…+β^pXji, for i = 1,2, … ,66 where Yi ^ represents the estimated mortality rate for the ith\n county and β^0 is the intercept. The variables X\n1\n\ni \nthrough Xji\n represent the values of the factors for the ith\n county and β^1 through β^p are the coefficients that were estimated using the least squares regression method (19,21). To explore the data, 4 linear regression models were created, which differ by factors included in the model.\nThe first regression model included all 8 factors as prediction variables resulting in Model 1. The regression model was then fitted by stepwise variable selection in both the forward and backward direction by using AIC as a model selection criterion. AIC rewards goodness of fit and penalizes the model’s complexity (19). This was done by using the R package MASS version 7.3–54 (R Foundation for Statistical Computing) (22). The simplified model resulted in Model 2. At this point, a decision was made to remove the incidence rate from the data to better see how the other sociodemographic factors contributed to breast cancer outcomes, resulting in Model 3. Model 3 was then fitted with stepwise variable selection, yielding Model 4. To best compare the expected mortality to the observed mortality with all predictors available, Model 2 was chosen as the final model. Model 2 was then used to predict the expected mortality rates for the second standardized incidence ratio (SIR) that was computed (SIRLM\n).", "A SIR was used to compare the spatial distribution of counties in terms of mortality rates due to breast cancer. In general, the SIR compares the expected value of deaths to the observed value of deaths in a county. This is calculated with SIRj=OjEj where Oj\n is the observed value for county j, and Ej\n is the expected value for county j. A SIR of greater than 1 means that there were more deaths than expected, whereas a SIR of less than 1 means that there were fewer deaths than expected for that county.\nBecause age-specific mortality rates by county were not available, the observed age-adjusted counts per county were computed from the age-adjusted rates per county as follows: Oj*=Oj100,000yj and Ej*=∑j=166Oj100,000·yj·yj∑j=166yj , where Oj\n is the jth\n county’s observed age-adjusted mortality rate and yj\n is the jth\n county’s population. The expected count was computed to be on the same scale as the observed count. These manipulations were used only to calculate an age-adjusted count SIR, referred to as SIRCOUNT\n. All other analysis of the data was completed with the original variable (Oj\n), as described in the data source.\nA SIR was calculated on the mortality in each South Dakota county (N = 66). To account for the age adjustment of the data, 2 different SIRs were found: using the age-adjusted mortality count for South Dakota and using the expected rate obtained from the linear regression model. The first SIR accounted only for the age adjustment, and the second SIR accounted for more factors related to breast cancer. For example, the first SIR used the age-adjusted mortality count for a county for both observed and expected (SIRCOUNT\n). The second SIR (SIRLM\n) used the mortality rates by county, which were per 100,000 persons and age-adjusted to the 2000 US standard population, and the South Dakota estimated population for observed and predicted mortality rate from the linear regression model for expected, which accounted for incidence rates and educational attainment. All statistical analyses used R version 4.1.2 and RStudio version 2021.09.2 build 382 (R Foundation for Statistical Computing) (23,24).", "[SUBTITLE] Exploratory data analysis [SUBSECTION] To learn more about the data, we performed an Exploratory Data Analysis (EDA) using several techniques. We explored the geographic distribution of the breast cancer mortality rates by using a choropleth map of South Dakota with age-adjusted mortality and incidence rates (Figure 1). The eastern side of the state had lower and more consistent mortality rates, followed by the far western part of the state. The central west part of the state exhibited higher mortality rates.\nMap A shows the age-adjusted breast cancer mortality rates and Map B shows the age-adjusted breast cancer incidence rates, by county (N = 66), South Dakota, 2001–2015. Counties whose mortality rates have been imputed are marked with a star. Source: South Dakota State Cancer Registry, South Dakota Department of Health (12).\nFive counties had higher mortality rates than the rest of the counties: Perkins, Mellette, Aurora, Douglas, and Corson. Two counties had lower mortality rates than the rest of the counties: Ziebach and Jackson. Eight counties (Bennet, Buffalo, Corson, Jackson, Mellette, Oglala, Todd, and Ziebach) had a poverty percentage greater than 30%, which was distinctly higher than the others; each county contains land on an American Indian Reservation. Minnehaha and Pennington counties had high screening rates; these counties are home to the first- and second-largest cities in South Dakota, respectively, so they also had the greatest number of screening providers.\nAll variables, besides the number of providers and screening rate, had a correlation greater than zero with mortality rate. Incidence, educational attainment, and uninsured percentage all had a low positive correlation with mortality rate. The highest correlation with mortality rate was education attainment, at a value of 0.26. The remaining variables had a negligible correlation with mortality rate.\nTo learn more about the data, we performed an Exploratory Data Analysis (EDA) using several techniques. We explored the geographic distribution of the breast cancer mortality rates by using a choropleth map of South Dakota with age-adjusted mortality and incidence rates (Figure 1). The eastern side of the state had lower and more consistent mortality rates, followed by the far western part of the state. The central west part of the state exhibited higher mortality rates.\nMap A shows the age-adjusted breast cancer mortality rates and Map B shows the age-adjusted breast cancer incidence rates, by county (N = 66), South Dakota, 2001–2015. Counties whose mortality rates have been imputed are marked with a star. Source: South Dakota State Cancer Registry, South Dakota Department of Health (12).\nFive counties had higher mortality rates than the rest of the counties: Perkins, Mellette, Aurora, Douglas, and Corson. Two counties had lower mortality rates than the rest of the counties: Ziebach and Jackson. Eight counties (Bennet, Buffalo, Corson, Jackson, Mellette, Oglala, Todd, and Ziebach) had a poverty percentage greater than 30%, which was distinctly higher than the others; each county contains land on an American Indian Reservation. Minnehaha and Pennington counties had high screening rates; these counties are home to the first- and second-largest cities in South Dakota, respectively, so they also had the greatest number of screening providers.\nAll variables, besides the number of providers and screening rate, had a correlation greater than zero with mortality rate. Incidence, educational attainment, and uninsured percentage all had a low positive correlation with mortality rate. The highest correlation with mortality rate was education attainment, at a value of 0.26. The remaining variables had a negligible correlation with mortality rate.\n[SUBTITLE] Regression analysis [SUBSECTION] Model 2 had the highest adjusted R\n2 value (0.10) and the lowest AIC (441.79), with 2 significant factors associated with mortality rate; Model 3 had the lowest adjusted R\n2 (.004) and the highest AIC (453.32), with no significant predictors of mortality. In Model 2, breast cancer incidence and educational attainment were predictors of breast cancer mortality, indicating that as more people are diagnosed with breast cancer and as the percentage of people with less than a bachelor’s degree increases, breast cancer mortality rate increases (Table 2). Educational attainment was a predictor of mortality in all models, and the educational attainment statistic had a P < .001. \nAbbreviation: — , not applicable; AIC, Akaike information criterion.\n\nP = .01.\n\nP = .001.\nModel 2 had the highest adjusted R\n2 value (0.10) and the lowest AIC (441.79), with 2 significant factors associated with mortality rate; Model 3 had the lowest adjusted R\n2 (.004) and the highest AIC (453.32), with no significant predictors of mortality. In Model 2, breast cancer incidence and educational attainment were predictors of breast cancer mortality, indicating that as more people are diagnosed with breast cancer and as the percentage of people with less than a bachelor’s degree increases, breast cancer mortality rate increases (Table 2). Educational attainment was a predictor of mortality in all models, and the educational attainment statistic had a P < .001. \nAbbreviation: — , not applicable; AIC, Akaike information criterion.\n\nP = .01.\n\nP = .001.\n[SUBTITLE] Standardized incidence ratio [SUBSECTION] Thirty-five of the 66 counties had a SIRCOUNT\n greater than 1, meaning that more than half of the counties had more deaths than expected (Figure 2). The 5 counties with the highest SIRCOUNT\n, in decreasing order, were Perkins, Mellette, Aurora, Douglas, and Corson. Of those, Perkins, Mellette, and Aurora had more than twice the expected number of deaths. Ziebach, Jackson, Davison, Tripp, and Meade counties had the lowest SIRCOUNT\n, with Ziebach and Jackson both being less than half the expected number of deaths. The highest SIRCOUNT\n was 2.15 and the lowest was 0.31.\nMap A shows the predicted breast cancer mortality rate of South Dakota counties, accounting for age-adjustment of the data, and Map B shows the predicted breast cancer mortality rate of South Dakota counties, accounting for age-adjustment, incidence rate, and educational attainment. Abbreviation: SIR, standardized incidence ratio. Map Sources: South Dakota State Cancer Registry, South Dakota Department of Health (12), Holzhauser et al (13), and the US Census Bureau (8).\nOn the other hand, for the SIRLM\n, most counties had fewer deaths than expected, with 28 of 66 counties having a SIRLM\n greater than 1. The 5 counties with the highest SIRLM\n were similar to the SIRCOUNT\n: Corson, Perkins, Aurora, Jones, and Mellette in decreasing order. No county had more than twice the expected number of deaths. The 5 counties with the lowest SIRLM\n were Ziebach, Jackson, Tripp, Oglala Lakota, and Davison. Ziebach and Jackson counties had less than half the number of expected deaths. The highest SIRLM\n was 1.94 and the lowest SIRLM\n was 0.35. \nThe results of the SIRs (SIRCOUNT\n and SIRLM\n) are presented in Figure 2. The eastern side of the state showed similar SIR values while the western side of the state had more variation. Perkins, Mellette, Aurora, Douglas, and Corson counties had SIRCOUNT\n values that were much higher than those of the rest of the counties. Ziebach and Jackson counties had the lowest SIRCOUNT\n values. These counties with the highest SIRCOUNT\n made up 5 of the 6 counties with the highest SIRLM\n values, with Jones County replacing Douglas County. The 2 counties with the lowest SIRCOUNT\n values were the same 2 counties with the lowest SIRLM\n values.\nThirty-five of the 66 counties had a SIRCOUNT\n greater than 1, meaning that more than half of the counties had more deaths than expected (Figure 2). The 5 counties with the highest SIRCOUNT\n, in decreasing order, were Perkins, Mellette, Aurora, Douglas, and Corson. Of those, Perkins, Mellette, and Aurora had more than twice the expected number of deaths. Ziebach, Jackson, Davison, Tripp, and Meade counties had the lowest SIRCOUNT\n, with Ziebach and Jackson both being less than half the expected number of deaths. The highest SIRCOUNT\n was 2.15 and the lowest was 0.31.\nMap A shows the predicted breast cancer mortality rate of South Dakota counties, accounting for age-adjustment of the data, and Map B shows the predicted breast cancer mortality rate of South Dakota counties, accounting for age-adjustment, incidence rate, and educational attainment. Abbreviation: SIR, standardized incidence ratio. Map Sources: South Dakota State Cancer Registry, South Dakota Department of Health (12), Holzhauser et al (13), and the US Census Bureau (8).\nOn the other hand, for the SIRLM\n, most counties had fewer deaths than expected, with 28 of 66 counties having a SIRLM\n greater than 1. The 5 counties with the highest SIRLM\n were similar to the SIRCOUNT\n: Corson, Perkins, Aurora, Jones, and Mellette in decreasing order. No county had more than twice the expected number of deaths. The 5 counties with the lowest SIRLM\n were Ziebach, Jackson, Tripp, Oglala Lakota, and Davison. Ziebach and Jackson counties had less than half the number of expected deaths. The highest SIRLM\n was 1.94 and the lowest SIRLM\n was 0.35. \nThe results of the SIRs (SIRCOUNT\n and SIRLM\n) are presented in Figure 2. The eastern side of the state showed similar SIR values while the western side of the state had more variation. Perkins, Mellette, Aurora, Douglas, and Corson counties had SIRCOUNT\n values that were much higher than those of the rest of the counties. Ziebach and Jackson counties had the lowest SIRCOUNT\n values. These counties with the highest SIRCOUNT\n made up 5 of the 6 counties with the highest SIRLM\n values, with Jones County replacing Douglas County. The 2 counties with the lowest SIRCOUNT\n values were the same 2 counties with the lowest SIRLM\n values.", "To learn more about the data, we performed an Exploratory Data Analysis (EDA) using several techniques. We explored the geographic distribution of the breast cancer mortality rates by using a choropleth map of South Dakota with age-adjusted mortality and incidence rates (Figure 1). The eastern side of the state had lower and more consistent mortality rates, followed by the far western part of the state. The central west part of the state exhibited higher mortality rates.\nMap A shows the age-adjusted breast cancer mortality rates and Map B shows the age-adjusted breast cancer incidence rates, by county (N = 66), South Dakota, 2001–2015. Counties whose mortality rates have been imputed are marked with a star. Source: South Dakota State Cancer Registry, South Dakota Department of Health (12).\nFive counties had higher mortality rates than the rest of the counties: Perkins, Mellette, Aurora, Douglas, and Corson. Two counties had lower mortality rates than the rest of the counties: Ziebach and Jackson. Eight counties (Bennet, Buffalo, Corson, Jackson, Mellette, Oglala, Todd, and Ziebach) had a poverty percentage greater than 30%, which was distinctly higher than the others; each county contains land on an American Indian Reservation. Minnehaha and Pennington counties had high screening rates; these counties are home to the first- and second-largest cities in South Dakota, respectively, so they also had the greatest number of screening providers.\nAll variables, besides the number of providers and screening rate, had a correlation greater than zero with mortality rate. Incidence, educational attainment, and uninsured percentage all had a low positive correlation with mortality rate. The highest correlation with mortality rate was education attainment, at a value of 0.26. The remaining variables had a negligible correlation with mortality rate.", "Model 2 had the highest adjusted R\n2 value (0.10) and the lowest AIC (441.79), with 2 significant factors associated with mortality rate; Model 3 had the lowest adjusted R\n2 (.004) and the highest AIC (453.32), with no significant predictors of mortality. In Model 2, breast cancer incidence and educational attainment were predictors of breast cancer mortality, indicating that as more people are diagnosed with breast cancer and as the percentage of people with less than a bachelor’s degree increases, breast cancer mortality rate increases (Table 2). Educational attainment was a predictor of mortality in all models, and the educational attainment statistic had a P < .001. \nAbbreviation: — , not applicable; AIC, Akaike information criterion.\n\nP = .01.\n\nP = .001.", "Thirty-five of the 66 counties had a SIRCOUNT\n greater than 1, meaning that more than half of the counties had more deaths than expected (Figure 2). The 5 counties with the highest SIRCOUNT\n, in decreasing order, were Perkins, Mellette, Aurora, Douglas, and Corson. Of those, Perkins, Mellette, and Aurora had more than twice the expected number of deaths. Ziebach, Jackson, Davison, Tripp, and Meade counties had the lowest SIRCOUNT\n, with Ziebach and Jackson both being less than half the expected number of deaths. The highest SIRCOUNT\n was 2.15 and the lowest was 0.31.\nMap A shows the predicted breast cancer mortality rate of South Dakota counties, accounting for age-adjustment of the data, and Map B shows the predicted breast cancer mortality rate of South Dakota counties, accounting for age-adjustment, incidence rate, and educational attainment. Abbreviation: SIR, standardized incidence ratio. Map Sources: South Dakota State Cancer Registry, South Dakota Department of Health (12), Holzhauser et al (13), and the US Census Bureau (8).\nOn the other hand, for the SIRLM\n, most counties had fewer deaths than expected, with 28 of 66 counties having a SIRLM\n greater than 1. The 5 counties with the highest SIRLM\n were similar to the SIRCOUNT\n: Corson, Perkins, Aurora, Jones, and Mellette in decreasing order. No county had more than twice the expected number of deaths. The 5 counties with the lowest SIRLM\n were Ziebach, Jackson, Tripp, Oglala Lakota, and Davison. Ziebach and Jackson counties had less than half the number of expected deaths. The highest SIRLM\n was 1.94 and the lowest SIRLM\n was 0.35. \nThe results of the SIRs (SIRCOUNT\n and SIRLM\n) are presented in Figure 2. The eastern side of the state showed similar SIR values while the western side of the state had more variation. Perkins, Mellette, Aurora, Douglas, and Corson counties had SIRCOUNT\n values that were much higher than those of the rest of the counties. Ziebach and Jackson counties had the lowest SIRCOUNT\n values. These counties with the highest SIRCOUNT\n made up 5 of the 6 counties with the highest SIRLM\n values, with Jones County replacing Douglas County. The 2 counties with the lowest SIRCOUNT\n values were the same 2 counties with the lowest SIRLM\n values.", "Overall, we found a significant association between incidence rate and educational level with respect to breast cancer mortality rates. Breast cancer incidence was positively associated with mortality rates in South Dakota, which suggests that more breast cancer cases are associated with more breast cancer deaths. In addition, educational attainment was repeatedly identified as a significant factor for mortality. Gadeyne et al found inconclusive results in their study of breast cancer mortality and education; however, Albano et al found a significant association between educational levels and cancer in general (7,25), specifically that lower educational attainment was related to higher cancer mortality rates, reflecting the findings of this study. Race, median age, and number of women screened were not selected in the feature selection during stepwise regression in our study; similarly, race, median age, and number of women screened were not significant in our full model.\nThe 2020–2021 South Dakota Department of Education yearly review stated that American Indians were the largest minority group in school. However, American Indians still have a 63% completion rate for high school graduation and 59% attendance rate, compared with Whites who have a 94% completion rate for high school graduation and 94% attendance rate (26). An interesting point to consider is that South Dakota has no set standards for sex education (27). Thus, students are not taught reproductive health in general, including the importance of breast examinations, Pap smears, or prostate examinations. We advocate that set and scientifically backed health standards in high school would expose students at an early age to the risks of breast cancer and their options for screening.\nThe western half of South Dakota had more variability in SIR values, and the state’s demographics could be a possible explanation. The 4 counties with the highest SIRs for both count and linear model SIRs were Corson, Perkins, Aurora, and Mellette, which are either in an American Indian reservation or neighbor a county within an American Indian reservation. Research on 3 tribes in western South Dakota supported that trust is often a barrier for American Indians (as are remote location and approvals by Indian Health Service programs) (28). Research in New Mexico reported that even after in-depth implementation of screening programs that lowered the barriers of cost, availability, and access to Native American and Hispanic women, the screening rates remained low, under 40% of women annually (26). The high SIRs in or neighboring reservation counties may mean that trust is also an issue, and South Dakota has more to work on than accessibility to Native Americans.\nAfter controlling for incidence rate and educational attainment, the SIRLM\n values became less variable. The SIRs’ decrease in range and mean closer to 1 indicate that the factors did affect mortality rate. This again agrees with findings from Albano et al that educational attainment affects mortality (7).\nWe found that some counties had a higher mortality rate than expected based on the age of the women in the county. Ziebach and Jackson counties had the highest mortality rates, and the counties with the lowest SIRs are not home to major medical centers. Haakon County is vertically between Ziebach and Jackson counties and is one of the counties that does not have a provider; however, Haakon County has a higher mortality rate than the average of counties of South Dakota and both SIRs greater than 1, which means there were more deaths than expected. The areas of the map where there are dark green counties next to dark red counties are either on an American Indian reservation or neighbor an American Indian reservation. The differences between counties do not come from any singular cause, but rather due to variations in race, poverty levels, and population size.\nOur study has limitations, primarily in the absence of portions of mortality data. Because South Dakota is largely a rural state, several counties have small populations and see very few deaths from breast cancer. These numbers are then withheld from the public to protect the privacy of the patients. This suppression resulted in having to impute the mortality rates of 15 counties, possibly introducing errors. The assumptions of the regression model did not account for this error, which may confer bias on the results. The counties with American Indian reservations have another health system that could have resulted in the under-representation or over-representation of breast cancer deaths in those counties (29). In addition, a study found that the misclassification of the race of Native Americans caused an underestimation of mortality rates as well (30). These gaps in databases and their contents highlight research challenges that rural communities will continue to face when few data are collected, populations and incidence are sparse, and data are inconsistently collected by multiple sources. Sociodemographic data are also challenging to consistently collect throughout a state. More detailed data per county would help yield accurate and unbiased results. For example, considering education, Zajacova and Lawrence argue that education is not a single-generation factor (31). Having data on the educational attainment of a patient’s parents or family, in addition to their own educational attainment, would allow us to assess the risk and see the relationship between education and breast cancer incidence or mortality. Thus, more research is needed to understand the effects education level has on financial security, stable employment, social success, and in turn, breast cancer mortality.\nIn conclusion, understanding the risk factors and geographic distribution of breast cancer mortality among women across the state will assist stakeholders with efforts at prevention and resource allocation guided by data." ]
[ "other1", "other2", "other3", "intro", "methods", null, null, null, null, null, null, null, "results", null, null, null, "discussion" ]
[]
Effect of Comorbidities on the Infection Rate and Severity of COVID-19: Nationwide Cohort Study With Propensity Score Matching.
36265125
A vaccine against COVID-19 has been developed; however, COVID-19 transmission continues. Although there have been many studies of comorbidities that have important roles in COVID-19, some studies have reported contradictory results.
BACKGROUND
Data were derived from a nationwide South Korean COVID-19 cohort study with propensity score (PS) matching. We included infected individuals who were COVID-19-positive between January 1, 2020, and May 30, 2020, and PS-matched uninfected controls. PS matching was performed to balance the baseline characteristics of each comorbidity and to adjust for potential confounders, such as age, sex, Charlson Comorbidity Index, medication, and other comorbidities, that were matched with binary variables. The outcomes were the confirmed comorbidities affecting the infection rate and severity of COVID-19. The endpoints were COVID-19 positivity and severe clinical outcomes of COVID-19 (such as tracheostomy, continuous renal replacement therapy, intensive care unit admission, ventilator use, cardiopulmonary resuscitation, and death).
METHODS
The COVID-19 cohort with PS matching included 8070 individuals with positive COVID-19 test results and 8070 matched controls. The proportions of patients in the severe group were higher for individuals 60 years or older (severe clinical outcomes for those 60 years or older, 16.52%; severe clinical outcomes for those of other ages, 2.12%), those insured with Medicaid (Medicaid, 10.81%; other insurance, 5.61%), and those with disabilities (with disabilities, 18.26%; without disabilities, 5.07%). The COVID-19 infection rate was high for patients with pulmonary disease (odds ratio [OR] 1.88; 95% CI 1.70-2.03), dementia (OR 1.75; 95% CI 1.40-2.20), gastrointestinal disease (OR 1.74; 95% CI 1.62-1.88), stroke (OR 1.67; 95% CI 1.23-2.27), hepatobiliary disease (OR 1.31; 95% CI 1.19-1.44), diabetes mellitus (OR 1.28; 95% CI 1.16-1.43), and cardiovascular disease (OR 1.20; 95% CI 1.07-1.35). In contrast, it was lower for individuals with hyperlipidemia (OR 0.73; 95% CI 0.67-0.80), autoimmune disease (OR 0.73; 95% CI 0.60-0.89), and cancer (OR 0.73; 95% CI 0.62-0.86). The severity of COVID-19 was high for individuals with kidney disease (OR 5.59; 95% CI 2.48-12.63), hypertension (OR 2.92; 95% CI 1.91-4.47), dementia (OR 2.92; 95% CI 1.91-4.47), cancer (OR 1.84; 95% CI 1.15-2.94), pulmonary disease (OR 1.72; 95% CI 1.35-2.19), cardiovascular disease (OR 1.54; 95% CI 1.17-2.04), diabetes mellitus (OR 1.43; 95% CI 1.09-1.87), and psychotic disorders (OR 1.29; 95% CI 1.01-6.52). However, it was low for those with hyperlipidemia (OR 0.78; 95% CI 0.60-1.00).
RESULTS
Upon PS matching considering the use of statins, it was concluded that people with hyperlipidemia could have lower infection rates and disease severity of COVID-19.
CONCLUSIONS
[ "United States", "Humans", "Child, Preschool", "COVID-19", "SARS-CoV-2", "Cohort Studies", "Propensity Score", "Cardiovascular Diseases", "COVID-19 Vaccines", "Comorbidity", "Diabetes Mellitus", "Hyperlipidemias", "Dementia" ]
9678330
Introduction
The World Health Organization declared that COVID-19 was a pandemic in March 2020. By August 2022, approximately 600 million individuals had been infected, and more than 6 million had died. Since then, vaccines and therapeutic agents for COVID-19 have been developed. However, the current number of individuals with COVID-19 is still the same as that 1 year ago because it has not yet been eradicated [1]. COVID-19 can result in an asymptomatic presentation or flu-like symptoms. Some patients are admitted to the hospital for conservative treatment, and some require intensive care unit admission. Moreover, some patients may die as a result of COVID-19 [2,3]. As the number of individuals with COVID-19 increases, it is important to identify those who are vulnerable to severe COVID-19 to effectively manage health care resources accordingly and to improve the prognosis [4,5]. Since the COVID-19 outbreak, many studies of the demographic factors that predispose individuals to infection and of the identification of comorbidities of infected individuals have been performed. Most studies have reported similar overall results; however, some results of these studies are contradictory [6-9]. These differences in results may be attributable to the diversity of patients and medical systems in various countries worldwide. Most previous studies on comorbidities analyzed the baseline characteristics of people infected with COVID-19 without considering the bias caused by various factors that influence COVID-19. For instance, to determine whether hyperlipidemia affects the severity of COVID-19, it is necessary to control for statins, which are often used by individuals with hyperlipidemia. Although some studies suggested that statins might have a role in reducing the severity of COVID-19 [10,11], most studies did not confirm the use of statins; they only reported the effect of hyperlipidemia [12-16]. Hence, it is difficult to accurately determine the effect of hyperlipidemia on the severity of COVID-19. We investigated the effects of patient comorbidities on the infection rate and severity of COVID-19. Bias was reduced by propensity score (PS) matching for various variables that may affect COVID-19. We also analyzed the demographic characteristics of patients with COVID-19.
Methods
[SUBTITLE] Study Design and Participants [SUBSECTION] We conducted a large-scale cohort study using a South Korean National Health Insurance claims database [17]. In South Korea, all citizens are registered in the Korean National Health Insurance Service (KNHIS) database. The KNHIS uses a nationwide, large-scale database system including information regarding the diagnostic codes from the International Classification of Diseases (ICD)-10, the names of the procedures performed, prescription drugs, hospital information, direct medical costs of inpatient and outpatient treatments, and medical insurance premiums. Because all Koreans are given unique identification numbers at birth that are used in the KNHIS, the health records of patients are not duplicated nor omitted [18,19]. For COVID-19 studies, KNHIS provides a COVID-19 cohort that includes people infected with COVID-19 and a control group that had never been infected. From January 1, 2020, to May 31, 2020, disease codes B342, B972, U071, U072, MT043, and 3/02 were used to identify patients with confirmed COVID-19. Data from the control group of individuals who were not previously diagnosed with COVID-19 were adjusted for sex, age, and region of residence. Moreover, the number of participants in the control group was 15 times the number of confirmed COVID-19 cases. We conducted a large-scale cohort study using a South Korean National Health Insurance claims database [17]. In South Korea, all citizens are registered in the Korean National Health Insurance Service (KNHIS) database. The KNHIS uses a nationwide, large-scale database system including information regarding the diagnostic codes from the International Classification of Diseases (ICD)-10, the names of the procedures performed, prescription drugs, hospital information, direct medical costs of inpatient and outpatient treatments, and medical insurance premiums. Because all Koreans are given unique identification numbers at birth that are used in the KNHIS, the health records of patients are not duplicated nor omitted [18,19]. For COVID-19 studies, KNHIS provides a COVID-19 cohort that includes people infected with COVID-19 and a control group that had never been infected. From January 1, 2020, to May 31, 2020, disease codes B342, B972, U071, U072, MT043, and 3/02 were used to identify patients with confirmed COVID-19. Data from the control group of individuals who were not previously diagnosed with COVID-19 were adjusted for sex, age, and region of residence. Moreover, the number of participants in the control group was 15 times the number of confirmed COVID-19 cases. [SUBTITLE] Ethical Considerations [SUBSECTION] This study was approved by the relevant institutional review board and research ethics committee (ISPAIK 2020-06-048-001). The need for written consent was formally waived by the ethics committee. This study used the NHIS-2020-1-328 database provided by the KNHIS in 2020. This study was approved by the relevant institutional review board and research ethics committee (ISPAIK 2020-06-048-001). The need for written consent was formally waived by the ethics committee. This study used the NHIS-2020-1-328 database provided by the KNHIS in 2020. [SUBTITLE] Study Population [SUBSECTION] In accordance with the World Health Organization guidelines, laboratory confirmation of COVID-19 was defined as a positive result of a real-time reverse-transcription polymerase chain reaction assay using a sample obtained with nasal and pharyngeal swabs [20]. We combined the claims-based data from the KNHIS between January 1, 2015, and May 31, 2020, and extracted information regarding age, sex, and region of residence from the insurance eligibility data (Figure 1). The Charlson Comorbidity Index (CCI) score was calculated using the ICD-10 codes and previously reported methods [21]. Certain underlying medications and diseases with a high risk of serious illness attributable to SARS-CoV-2, which causes COVID-19, were studied and reported by the Centers for Disease Control and Prevention (CDC) and previous meta-analysis studies [6-9,22]. In these studies, we selected factors to use for PS matching in the analysis (Tables S1 and S2 in Multimedia Appendix 1). Only those (pulmonary disease, cardiovascular disease, hepatobiliary disease, hyperlipidemia, gastrointestinal disease, diabetes mellitus, hypertension, and psychotic disorder) with more than 500 people with COVID-19 were selected because a small number of people with corresponding comorbidities might cause statistical bias (Multimedia Appendix 2). A history of underlying diseases (pulmonary disease, cardiovascular disease, kidney disease, hepatobiliary disease, hyperlipidemia, gastrointestinal disease, diabetes mellitus, hypertension, psychotic disorder, dementia, stroke, neurologic disorder, autoimmune disease, and cancer) was confirmed by the assignment of at least two claims within 1 year using the appropriate ICD-10 code. We used various PS matching methods for factors affecting COVID-19: (1) matching for age, sex, and CCI; (2) additional matching for comorbidities; and (3) additional matching for medications. Finally, the results from (3) were used (Tables S3 and S4 in Multimedia Appendix 1). The financial revenue of the National Health Insurance of Korea consists of contributions from the insured and government subsidies, which can be used to analyze socioeconomic status. The contributions to the National Health Insurance differ according to the family income level. The higher the income, the greater the contribution to the National Health Insurance. Income was divided into 5 categories for the purpose of statistical analyses. The first category is Medicaid, and the successive categories include progressively higher (by 25%) income groups. Disability grades were categorized as mild or severe based on the KNHIS database information for people registered with the Korean government. Disposition of patients in the KNHIS-COVID cohort (South Korea; January 1 to May 31, 2020). CCI: Charlson Comorbidity Index; KNHIS: Korean National Health Insurance Service. In accordance with the World Health Organization guidelines, laboratory confirmation of COVID-19 was defined as a positive result of a real-time reverse-transcription polymerase chain reaction assay using a sample obtained with nasal and pharyngeal swabs [20]. We combined the claims-based data from the KNHIS between January 1, 2015, and May 31, 2020, and extracted information regarding age, sex, and region of residence from the insurance eligibility data (Figure 1). The Charlson Comorbidity Index (CCI) score was calculated using the ICD-10 codes and previously reported methods [21]. Certain underlying medications and diseases with a high risk of serious illness attributable to SARS-CoV-2, which causes COVID-19, were studied and reported by the Centers for Disease Control and Prevention (CDC) and previous meta-analysis studies [6-9,22]. In these studies, we selected factors to use for PS matching in the analysis (Tables S1 and S2 in Multimedia Appendix 1). Only those (pulmonary disease, cardiovascular disease, hepatobiliary disease, hyperlipidemia, gastrointestinal disease, diabetes mellitus, hypertension, and psychotic disorder) with more than 500 people with COVID-19 were selected because a small number of people with corresponding comorbidities might cause statistical bias (Multimedia Appendix 2). A history of underlying diseases (pulmonary disease, cardiovascular disease, kidney disease, hepatobiliary disease, hyperlipidemia, gastrointestinal disease, diabetes mellitus, hypertension, psychotic disorder, dementia, stroke, neurologic disorder, autoimmune disease, and cancer) was confirmed by the assignment of at least two claims within 1 year using the appropriate ICD-10 code. We used various PS matching methods for factors affecting COVID-19: (1) matching for age, sex, and CCI; (2) additional matching for comorbidities; and (3) additional matching for medications. Finally, the results from (3) were used (Tables S3 and S4 in Multimedia Appendix 1). The financial revenue of the National Health Insurance of Korea consists of contributions from the insured and government subsidies, which can be used to analyze socioeconomic status. The contributions to the National Health Insurance differ according to the family income level. The higher the income, the greater the contribution to the National Health Insurance. Income was divided into 5 categories for the purpose of statistical analyses. The first category is Medicaid, and the successive categories include progressively higher (by 25%) income groups. Disability grades were categorized as mild or severe based on the KNHIS database information for people registered with the Korean government. Disposition of patients in the KNHIS-COVID cohort (South Korea; January 1 to May 31, 2020). CCI: Charlson Comorbidity Index; KNHIS: Korean National Health Insurance Service. [SUBTITLE] Outcomes [SUBSECTION] To determine the severity of disease according to the demographic factors of COVID-19–infected patients, the severity scale was divided into the following 4 grades: mild, moderate, severe, and death. In South Korea, patients with asymptomatic or mild symptoms are discharged when a negative COVID-19 test result is confirmed 2 weeks after hospitalization. This time period also corresponds to the period of self-isolation. When we checked the hospitalization period of COVID-19–infected patients, the hospitalization period peaked on day 16 and decreased thereafter. Based on this result, a hospitalization period of ≤16 days was defined as the mild grade corresponding to asymptomatic or mild symptoms. The severe grade was defined as the need for tracheostomy, continuous renal replacement therapy, intensive care unit admission, ventilator use, and cardiopulmonary resuscitation. The moderate grade was defined as a hospitalization period >16 days but not requiring treatment corresponding to the severe grade. The primary aim of this study was to compare the severity grades of the COVID-19–infected and control groups based on demographic factors, comorbidities, and complications. The secondary aim was to perform PS matching for comparisons. We identified the infection rate and severity (severe and death or mild and moderate) of COVID-19 according to the comorbid conditions. To determine the severity of disease according to the demographic factors of COVID-19–infected patients, the severity scale was divided into the following 4 grades: mild, moderate, severe, and death. In South Korea, patients with asymptomatic or mild symptoms are discharged when a negative COVID-19 test result is confirmed 2 weeks after hospitalization. This time period also corresponds to the period of self-isolation. When we checked the hospitalization period of COVID-19–infected patients, the hospitalization period peaked on day 16 and decreased thereafter. Based on this result, a hospitalization period of ≤16 days was defined as the mild grade corresponding to asymptomatic or mild symptoms. The severe grade was defined as the need for tracheostomy, continuous renal replacement therapy, intensive care unit admission, ventilator use, and cardiopulmonary resuscitation. The moderate grade was defined as a hospitalization period >16 days but not requiring treatment corresponding to the severe grade. The primary aim of this study was to compare the severity grades of the COVID-19–infected and control groups based on demographic factors, comorbidities, and complications. The secondary aim was to perform PS matching for comparisons. We identified the infection rate and severity (severe and death or mild and moderate) of COVID-19 according to the comorbid conditions. [SUBTITLE] Statistical Analysis [SUBSECTION] We performed PS matching to balance the baseline characteristics of each comorbidity (existence or nonexistence) and to adjust for potential confounders. Because we focused on each comorbidity, PS matching was performed for each comorbidity. The PS was estimated using a logistic regression model and calculating the predicted probability of covariates. Age and CCI (0, 1, or ≥2) were matched with continuous variables. Sex, medication, and other comorbidities were matched with binary variables. We assessed the PS matching of the comorbidity existence using a 1:1 ratio, the greedy nearest neighbor algorithm, and a scale with a caliper of 0.25 (Multimedia Appendix 2). Data obtained after PS matching were analyzed by calculating the odds ratios (ORs) with 95% CIs for the infection rate and severity (severe and death or mild and moderate) of COVID-19. Statistical analyses were performed using SAS version 9.4 (SAS Institute Inc, Cary, NC). We performed PS matching to balance the baseline characteristics of each comorbidity (existence or nonexistence) and to adjust for potential confounders. Because we focused on each comorbidity, PS matching was performed for each comorbidity. The PS was estimated using a logistic regression model and calculating the predicted probability of covariates. Age and CCI (0, 1, or ≥2) were matched with continuous variables. Sex, medication, and other comorbidities were matched with binary variables. We assessed the PS matching of the comorbidity existence using a 1:1 ratio, the greedy nearest neighbor algorithm, and a scale with a caliper of 0.25 (Multimedia Appendix 2). Data obtained after PS matching were analyzed by calculating the odds ratios (ORs) with 95% CIs for the infection rate and severity (severe and death or mild and moderate) of COVID-19. Statistical analyses were performed using SAS version 9.4 (SAS Institute Inc, Cary, NC). [SUBTITLE] Patient and Public Involvement [SUBSECTION] No patient was directly involved in designing the study question or in conducting the study. No patients were asked for advice regarding the interpretation or writing of the results. There are no plans to involve patients or the relevant patient community in the dissemination of study findings at this time. No patient was directly involved in designing the study question or in conducting the study. No patients were asked for advice regarding the interpretation or writing of the results. There are no plans to involve patients or the relevant patient community in the dissemination of study findings at this time.
Results
[SUBTITLE] Clinical Characteristics of the Study Population [SUBSECTION] A total of 8070 individuals had positive COVID-19 results according to the reverse-transcription polymerase chain reaction assay. We identified 121,050 uninfected individuals as control participants (Multimedia Appendix 2). The demographic characteristics of the entire cohort are displayed in Table 1. The COVID-19 severity grade was mild for 2419 (2419/8070, 29.98%) individuals, moderate for 5160 (5160/8070, 63.94%) individuals, severe for 254 (254/8070, 3.15%) individuals, and death for 237 (237/8070, 2.94%) individuals. Among the total sample of infected individuals, 3236 (3236/8070, 40.10%) were male. Most patients were in their fifth (1567/8070, 19.42%) or sixth (1199/8070, 14.86%) decade of life. In terms of the medical insurance grade, which indicates socioeconomic status, those receiving Medicaid had high rates of severe grade and death. However, there were no obvious trends for the other grades. Individuals with disabilities had more severe infections and a much higher case fatality rate (Table 1). Those with COVID-19 had a medical history of gastrointestinal disease (n=5256), pulmonary disease (n=2539), hyperlipidemia (n=1841), and hypertension (n=1623). The case fatality rate was high for individuals with dementia (74/235, 31.5%), kidney disease (25/86, 29%), and cardiovascular disease (110/675, 16.3%; Table 2). After COVID-19 was confirmed, gastrointestinal disease (n=2912), pulmonary disease (n=2398), and hepatobiliary disease (n=1248) were the most common complications (Table 3). Baseline characteristics of the study population, including those infected (n=8070) and uninfected (n=121,050; controls) with COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020). aParticipants from some specific groups, such as soldiers, were not included. bThe uninfected controls were adjusted for sex, age, and region, resulting in a figure equivalent to 15 times the number of confirmed COVID-19 cases in the KNHIS-COVID cohort. Baseline characteristics of comorbidities of the study population, including those infected with (n=8070) and not infected with (n=121,050; controls) COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020). Baseline characteristics of complications of the study population, including those infected with (n=8070) and not infected with (n=121,050; controls) COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020). A total of 8070 individuals had positive COVID-19 results according to the reverse-transcription polymerase chain reaction assay. We identified 121,050 uninfected individuals as control participants (Multimedia Appendix 2). The demographic characteristics of the entire cohort are displayed in Table 1. The COVID-19 severity grade was mild for 2419 (2419/8070, 29.98%) individuals, moderate for 5160 (5160/8070, 63.94%) individuals, severe for 254 (254/8070, 3.15%) individuals, and death for 237 (237/8070, 2.94%) individuals. Among the total sample of infected individuals, 3236 (3236/8070, 40.10%) were male. Most patients were in their fifth (1567/8070, 19.42%) or sixth (1199/8070, 14.86%) decade of life. In terms of the medical insurance grade, which indicates socioeconomic status, those receiving Medicaid had high rates of severe grade and death. However, there were no obvious trends for the other grades. Individuals with disabilities had more severe infections and a much higher case fatality rate (Table 1). Those with COVID-19 had a medical history of gastrointestinal disease (n=5256), pulmonary disease (n=2539), hyperlipidemia (n=1841), and hypertension (n=1623). The case fatality rate was high for individuals with dementia (74/235, 31.5%), kidney disease (25/86, 29%), and cardiovascular disease (110/675, 16.3%; Table 2). After COVID-19 was confirmed, gastrointestinal disease (n=2912), pulmonary disease (n=2398), and hepatobiliary disease (n=1248) were the most common complications (Table 3). Baseline characteristics of the study population, including those infected (n=8070) and uninfected (n=121,050; controls) with COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020). aParticipants from some specific groups, such as soldiers, were not included. bThe uninfected controls were adjusted for sex, age, and region, resulting in a figure equivalent to 15 times the number of confirmed COVID-19 cases in the KNHIS-COVID cohort. Baseline characteristics of comorbidities of the study population, including those infected with (n=8070) and not infected with (n=121,050; controls) COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020). Baseline characteristics of complications of the study population, including those infected with (n=8070) and not infected with (n=121,050; controls) COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020). [SUBTITLE] Risks of COVID-19 Positivity and Disease Severity According to Comorbidities [SUBSECTION] To identify differences according to comorbidity, predispositions were matched between the COVID-19–infected group and uninfected control group. No significant imbalances in the demographics and clinical characteristics were observed when they were assessed using the standardized mean difference within groups of PS-matched cohorts, which included the standardized mean difference of binary type variables <0.1. PS-matched ORs were checked for age, sex, CCI, medication, and comorbidities. When the control group and COVID-19–infected group were compared, COVID-19 was likely to occur in individuals with a history of the diseases and medical conditions but not for those with a history of hyperlipidemia (OR 0.73; 95% CI 0.67-0.80), autoimmune disease (OR 0.73; 95% CI 0.60-0.89), or cancer (OR 0.73; 95% CI 0.62-0.86; Table 4). The severity grade was high for COVID-19–infected individuals with pulmonary disease (OR 1.72; 95% CI 1.35-2.19), cardiovascular disease (OR 1.54; 95% CI 1.17-2.04), kidney disease (OR 5.59; 95% CI 2.48-12.63), diabetes mellitus (OR 1.43; 95% CI 1.09-1.87), hypertension (OR 1.63; 95% CI 1.23-2.15), psychotic disorder (OR 1.29; 95% CI 1.01-6.52), dementia (OR 2.92; 95% CI 1.91-4.47), or cancer (OR 1.84; 95% CI 1.15-2.94). However, the severity grade was low for COVID-19–infected individuals with hyperlipidemia (OR 0.70; 95% CI 0.55-0.90; Table 5 and Table S5 in Multimedia Appendix 1). Propensity score–matched (age, sex, Charlson Comorbidity Index, medications, and comorbidities) baseline characteristics and COVID-19 infection positivity rates according to comorbidity in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020). aWe assessed each propensity score–matched comorbidity using a 1:1 ratio for those in the COVID-19 and control groups. bComorbidity with more susceptibility to COVID-19. cComorbidity with less susceptibility to COVID-19. Propensity score–matched (age, sex, Charlson Comorbidity Index, medications, and comorbidities) baseline characteristics and clinical outcomes of COVID-19 among patients in the mild or moderate group and those in the severe or death group according to the comorbidity of patients with laboratory-confirmed COVID-19 infection in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020). aWe assessed each propensity score–matched comorbidity using a 1:1 ratio for those in the mild and moderate group and those in the severe and death group. bComorbidity with increasing COVID-19 severity. cComorbidity with decreasing COVID-19 severity. To identify differences according to comorbidity, predispositions were matched between the COVID-19–infected group and uninfected control group. No significant imbalances in the demographics and clinical characteristics were observed when they were assessed using the standardized mean difference within groups of PS-matched cohorts, which included the standardized mean difference of binary type variables <0.1. PS-matched ORs were checked for age, sex, CCI, medication, and comorbidities. When the control group and COVID-19–infected group were compared, COVID-19 was likely to occur in individuals with a history of the diseases and medical conditions but not for those with a history of hyperlipidemia (OR 0.73; 95% CI 0.67-0.80), autoimmune disease (OR 0.73; 95% CI 0.60-0.89), or cancer (OR 0.73; 95% CI 0.62-0.86; Table 4). The severity grade was high for COVID-19–infected individuals with pulmonary disease (OR 1.72; 95% CI 1.35-2.19), cardiovascular disease (OR 1.54; 95% CI 1.17-2.04), kidney disease (OR 5.59; 95% CI 2.48-12.63), diabetes mellitus (OR 1.43; 95% CI 1.09-1.87), hypertension (OR 1.63; 95% CI 1.23-2.15), psychotic disorder (OR 1.29; 95% CI 1.01-6.52), dementia (OR 2.92; 95% CI 1.91-4.47), or cancer (OR 1.84; 95% CI 1.15-2.94). However, the severity grade was low for COVID-19–infected individuals with hyperlipidemia (OR 0.70; 95% CI 0.55-0.90; Table 5 and Table S5 in Multimedia Appendix 1). Propensity score–matched (age, sex, Charlson Comorbidity Index, medications, and comorbidities) baseline characteristics and COVID-19 infection positivity rates according to comorbidity in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020). aWe assessed each propensity score–matched comorbidity using a 1:1 ratio for those in the COVID-19 and control groups. bComorbidity with more susceptibility to COVID-19. cComorbidity with less susceptibility to COVID-19. Propensity score–matched (age, sex, Charlson Comorbidity Index, medications, and comorbidities) baseline characteristics and clinical outcomes of COVID-19 among patients in the mild or moderate group and those in the severe or death group according to the comorbidity of patients with laboratory-confirmed COVID-19 infection in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020). aWe assessed each propensity score–matched comorbidity using a 1:1 ratio for those in the mild and moderate group and those in the severe and death group. bComorbidity with increasing COVID-19 severity. cComorbidity with decreasing COVID-19 severity.
null
null
[ "Study Design and Participants", "Ethical Considerations", "Study Population", "Outcomes", "Statistical Analysis", "Patient and Public Involvement", "Clinical Characteristics of the Study Population", "Risks of COVID-19 Positivity and Disease Severity According to Comorbidities", "Principal Findings", "Limitations", "Conclusions" ]
[ "We conducted a large-scale cohort study using a South Korean National Health Insurance claims database [17]. In South Korea, all citizens are registered in the Korean National Health Insurance Service (KNHIS) database. The KNHIS uses a nationwide, large-scale database system including information regarding the diagnostic codes from the International Classification of Diseases (ICD)-10, the names of the procedures performed, prescription drugs, hospital information, direct medical costs of inpatient and outpatient treatments, and medical insurance premiums. Because all Koreans are given unique identification numbers at birth that are used in the KNHIS, the health records of patients are not duplicated nor omitted [18,19]. For COVID-19 studies, KNHIS provides a COVID-19 cohort that includes people infected with COVID-19 and a control group that had never been infected. From January 1, 2020, to May 31, 2020, disease codes B342, B972, U071, U072, MT043, and 3/02 were used to identify patients with confirmed COVID-19. Data from the control group of individuals who were not previously diagnosed with COVID-19 were adjusted for sex, age, and region of residence. Moreover, the number of participants in the control group was 15 times the number of confirmed COVID-19 cases.", "This study was approved by the relevant institutional review board and research ethics committee (ISPAIK 2020-06-048-001). The need for written consent was formally waived by the ethics committee. This study used the NHIS-2020-1-328 database provided by the KNHIS in 2020.", "In accordance with the World Health Organization guidelines, laboratory confirmation of COVID-19 was defined as a positive result of a real-time reverse-transcription polymerase chain reaction assay using a sample obtained with nasal and pharyngeal swabs [20]. We combined the claims-based data from the KNHIS between January 1, 2015, and May 31, 2020, and extracted information regarding age, sex, and region of residence from the insurance eligibility data (Figure 1). The Charlson Comorbidity Index (CCI) score was calculated using the ICD-10 codes and previously reported methods [21]. Certain underlying medications and diseases with a high risk of serious illness attributable to SARS-CoV-2, which causes COVID-19, were studied and reported by the Centers for Disease Control and Prevention (CDC) and previous meta-analysis studies [6-9,22]. In these studies, we selected factors to use for PS matching in the analysis (Tables S1 and S2 in Multimedia Appendix 1). Only those (pulmonary disease, cardiovascular disease, hepatobiliary disease, hyperlipidemia, gastrointestinal disease, diabetes mellitus, hypertension, and psychotic disorder) with more than 500 people with COVID-19 were selected because a small number of people with corresponding comorbidities might cause statistical bias (Multimedia Appendix 2). A history of underlying diseases (pulmonary disease, cardiovascular disease, kidney disease, hepatobiliary disease, hyperlipidemia, gastrointestinal disease, diabetes mellitus, hypertension, psychotic disorder, dementia, stroke, neurologic disorder, autoimmune disease, and cancer) was confirmed by the assignment of at least two claims within 1 year using the appropriate ICD-10 code.\nWe used various PS matching methods for factors affecting COVID-19: (1) matching for age, sex, and CCI; (2) additional matching for comorbidities; and (3) additional matching for medications. Finally, the results from (3) were used (Tables S3 and S4 in Multimedia Appendix 1). The financial revenue of the National Health Insurance of Korea consists of contributions from the insured and government subsidies, which can be used to analyze socioeconomic status. The contributions to the National Health Insurance differ according to the family income level. The higher the income, the greater the contribution to the National Health Insurance. Income was divided into 5 categories for the purpose of statistical analyses. The first category is Medicaid, and the successive categories include progressively higher (by 25%) income groups. Disability grades were categorized as mild or severe based on the KNHIS database information for people registered with the Korean government.\nDisposition of patients in the KNHIS-COVID cohort (South Korea; January 1 to May 31, 2020). CCI: Charlson Comorbidity Index; KNHIS: Korean National Health Insurance Service.", "To determine the severity of disease according to the demographic factors of COVID-19–infected patients, the severity scale was divided into the following 4 grades: mild, moderate, severe, and death. In South Korea, patients with asymptomatic or mild symptoms are discharged when a negative COVID-19 test result is confirmed 2 weeks after hospitalization. This time period also corresponds to the period of self-isolation. When we checked the hospitalization period of COVID-19–infected patients, the hospitalization period peaked on day 16 and decreased thereafter. Based on this result, a hospitalization period of ≤16 days was defined as the mild grade corresponding to asymptomatic or mild symptoms. The severe grade was defined as the need for tracheostomy, continuous renal replacement therapy, intensive care unit admission, ventilator use, and cardiopulmonary resuscitation. The moderate grade was defined as a hospitalization period >16 days but not requiring treatment corresponding to the severe grade.\nThe primary aim of this study was to compare the severity grades of the COVID-19–infected and control groups based on demographic factors, comorbidities, and complications. The secondary aim was to perform PS matching for comparisons. We identified the infection rate and severity (severe and death or mild and moderate) of COVID-19 according to the comorbid conditions.", "We performed PS matching to balance the baseline characteristics of each comorbidity (existence or nonexistence) and to adjust for potential confounders. Because we focused on each comorbidity, PS matching was performed for each comorbidity. The PS was estimated using a logistic regression model and calculating the predicted probability of covariates. Age and CCI (0, 1, or ≥2) were matched with continuous variables. Sex, medication, and other comorbidities were matched with binary variables. We assessed the PS matching of the comorbidity existence using a 1:1 ratio, the greedy nearest neighbor algorithm, and a scale with a caliper of 0.25 (Multimedia Appendix 2). Data obtained after PS matching were analyzed by calculating the odds ratios (ORs) with 95% CIs for the infection rate and severity (severe and death or mild and moderate) of COVID-19. Statistical analyses were performed using SAS version 9.4 (SAS Institute Inc, Cary, NC).", "No patient was directly involved in designing the study question or in conducting the study. No patients were asked for advice regarding the interpretation or writing of the results. There are no plans to involve patients or the relevant patient community in the dissemination of study findings at this time.", "A total of 8070 individuals had positive COVID-19 results according to the reverse-transcription polymerase chain reaction assay. We identified 121,050 uninfected individuals as control participants (Multimedia Appendix 2). The demographic characteristics of the entire cohort are displayed in Table 1. The COVID-19 severity grade was mild for 2419 (2419/8070, 29.98%) individuals, moderate for 5160 (5160/8070, 63.94%) individuals, severe for 254 (254/8070, 3.15%) individuals, and death for 237 (237/8070, 2.94%) individuals. Among the total sample of infected individuals, 3236 (3236/8070, 40.10%) were male. Most patients were in their fifth (1567/8070, 19.42%) or sixth (1199/8070, 14.86%) decade of life. In terms of the medical insurance grade, which indicates socioeconomic status, those receiving Medicaid had high rates of severe grade and death. However, there were no obvious trends for the other grades. Individuals with disabilities had more severe infections and a much higher case fatality rate (Table 1). Those with COVID-19 had a medical history of gastrointestinal disease (n=5256), pulmonary disease (n=2539), hyperlipidemia (n=1841), and hypertension (n=1623). The case fatality rate was high for individuals with dementia (74/235, 31.5%), kidney disease (25/86, 29%), and cardiovascular disease (110/675, 16.3%; Table 2). After COVID-19 was confirmed, gastrointestinal disease (n=2912), pulmonary disease (n=2398), and hepatobiliary disease (n=1248) were the most common complications (Table 3).\nBaseline characteristics of the study population, including those infected (n=8070) and uninfected (n=121,050; controls) with COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\naParticipants from some specific groups, such as soldiers, were not included.\nbThe uninfected controls were adjusted for sex, age, and region, resulting in a figure equivalent to 15 times the number of confirmed COVID-19 cases in the KNHIS-COVID cohort.\nBaseline characteristics of comorbidities of the study population, including those infected with (n=8070) and not infected with (n=121,050; controls) COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\nBaseline characteristics of complications of the study population, including those infected with (n=8070) and not infected with (n=121,050; controls) COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).", "To identify differences according to comorbidity, predispositions were matched between the COVID-19–infected group and uninfected control group. No significant imbalances in the demographics and clinical characteristics were observed when they were assessed using the standardized mean difference within groups of PS-matched cohorts, which included the standardized mean difference of binary type variables <0.1. PS-matched ORs were checked for age, sex, CCI, medication, and comorbidities. When the control group and COVID-19–infected group were compared, COVID-19 was likely to occur in individuals with a history of the diseases and medical conditions but not for those with a history of hyperlipidemia (OR 0.73; 95% CI 0.67-0.80), autoimmune disease (OR 0.73; 95% CI 0.60-0.89), or cancer (OR 0.73; 95% CI 0.62-0.86; Table 4). The severity grade was high for COVID-19–infected individuals with pulmonary disease (OR 1.72; 95% CI 1.35-2.19), cardiovascular disease (OR 1.54; 95% CI 1.17-2.04), kidney disease (OR 5.59; 95% CI 2.48-12.63), diabetes mellitus (OR 1.43; 95% CI 1.09-1.87), hypertension (OR 1.63; 95% CI 1.23-2.15), psychotic disorder (OR 1.29; 95% CI 1.01-6.52), dementia (OR 2.92; 95% CI 1.91-4.47), or cancer (OR 1.84; 95% CI 1.15-2.94). However, the severity grade was low for COVID-19–infected individuals with hyperlipidemia (OR 0.70; 95% CI 0.55-0.90; Table 5 and Table S5 in Multimedia Appendix 1).\nPropensity score–matched (age, sex, Charlson Comorbidity Index, medications, and comorbidities) baseline characteristics and COVID-19 infection positivity rates according to comorbidity in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\naWe assessed each propensity score–matched comorbidity using a 1:1 ratio for those in the COVID-19 and control groups.\nbComorbidity with more susceptibility to COVID-19.\ncComorbidity with less susceptibility to COVID-19.\nPropensity score–matched (age, sex, Charlson Comorbidity Index, medications, and comorbidities) baseline characteristics and clinical outcomes of COVID-19 among patients in the mild or moderate group and those in the severe or death group according to the comorbidity of patients with laboratory-confirmed COVID-19 infection in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\naWe assessed each propensity score–matched comorbidity using a 1:1 ratio for those in the mild and moderate group and those in the severe and death group.\nbComorbidity with increasing COVID-19 severity.\ncComorbidity with decreasing COVID-19 severity.", "This study was a retrospective cohort study conducted in South Korea from January 2020 to May 2020. It involved confirmed COVID-19 patients with medical insurance. Previous studies of the demographic factors of individuals with COVID-19 showed that male sex, old age, and low income were factors likely associated with COVID-19 with a high severity grade [23,24]. In this study, more women had COVID-19, but the severity grade of COVID-19 was higher for men; this was directly proportional to age, especially for men older than 70 years. All medical expenses for COVID-19 are paid for by the South Korea government; therefore, all patients, including those receiving Medicaid, received the same level of care for COVID-19. Although there was no difference in medical care, those with Medicaid had the lowest income level and a higher severity grade; however, there were no differences between the groups with grades 1 to 4 medical insurance. For individuals with disabilities, the incidence was slightly higher than that of the control group. However, the severity grade was much higher than that of other individuals infected with COVID-19.\nOther studies of COVID-19 reported that SARS-CoV-2 binds to the angiotensin-converting enzyme 2 (ACE2) receptor through the viral structural spike protein at the onset of infection [25]. ACE2 is expressed to varying degrees in almost all human organs. ACE2 is highly expressed in cardiomyocytes, proximal tubule cells of the kidney, and bladder urinary tract cells. Additionally, it is abundantly expressed in intestinal cells of the small intestine, especially in the ileum [25-28]. Therefore, most critically ill patients with COVID-19 experience multiple organ injuries, including acute lung injuries, acute kidney injuries, cardiac injuries, hepatobiliary disease, and pneumothorax [29]. Therefore, to analyze the effect of each comorbidity on the COVID-19 infection severity grade, it is necessary to consider other comorbidities.\nEach demographic factor, comorbidity, and medication may influence each other, resulting in different outcomes in terms of the infection rate and severity of COVID-19. When analyzing comorbidities with hypertension, the effect of hypertension on the infection rate and severity of COVID-19 experienced by an 80-year-old woman with asthma and that of a 30-year-old man without an underlying medical condition may be different. Accurate results can be obtained for sufficiently studied diseases by controlling for only important factors. However, in the case of understudied diseases, such as COVID-19, various factors should be considered. In this study, PS matching was performed for various factors that could affect COVID-19, to minimize bias. When selecting a factor for PS matching, in order to select objective data, data provided by the CDC and meta-analysis studies were used. However, there was a limit, as data may change as research on COVID-19 progresses. Most of the results obtained were similar to those of previously published studies; however, some results were conflicting. For people with cancer and autoimmune disease, infection rates were even lower; these results were possibly affected by reducing social contact because of the risk of COVID-19 infection. Exposure to COVID-19 is an important factor that can affect the infection rate of COVID-19. Individuals with hyperlipidemia had a low COVID-19 infection rate and low severity grade. Previous studies reported that hyperlipidemia should be managed to prevent COVID-19 because high cholesterol levels induce inflammation and increase ACE2 availability [30-32]. Moreover, the use of statins for patients with COVID-19 reduced mortality by interfering with the mevalonate pathway and because of their antiviral effects [10,11,33]. However, some studies have shown that people with low lipid levels are more susceptible to and have more severe COVID-19 infection [34-41]. A meta-analysis published in 2022 indicated that patients with severe COVID-19 had lower total cholesterol levels (pooled mean difference –10.4; 95% CI –18.7 to –2.2), low-density lipoprotein cholesterol levels (pooled mean difference –4.4; 95% CI –8.4 to –0.42), and high-density lipoprotein cholesterol levels (pooled mean difference –4.4; 95% CI –6.9 to –1.8) on admission compared with patients with non-severe disease [42]. This may be similar to the “obesity paradox,” which states that mild obesity is advantageous to improvements after stroke [43,44]. Mild obesity can withstand the systemic catabolic imbalance with impaired metabolic efficiency and body tissue degradation that occur after stroke. Hyperlipidemia may also have a role in minimizing the severity of COVID-19.", "Our study has several limitations. As a limitation of most medical data studies, there is bias caused by confounding factors that may affect our results. When selecting factors for PS matching, information from the CDC and meta-analysis studies were used to select objective data, but these data may change as research on COVID-19 progresses. Further, we defined diseases based on the ICD codes provided in the insurance claims data. There may have been additional unmeasured confounders influencing our results, including genetic polymorphisms, smoking, body mass index, and exposure to the virus. In this study, the infection rate of COVID-19 may have been influenced by the degree of exposure to COVID-19, which may be an important factor in addition to the comorbidity factors. However, the influence of COVID-19 itself could be confirmed because the bias was less than that of previous studies. One race in South Korea comprises more than 95% of the population; hence, there was minimal racial bias compared with previous studies. Because the government funds the treatment for COVID-19 in South Korea and because the medical facilities for COVID-19 treatment are ubiquitous, there was minimal economic bias. The PS matching was performed for sex, age, CCI, comorbidity, and medication, including statins (standardized mean difference <0.1). Hence, selection bias was minimized. Therefore, more accurate information regarding the incidence of COVID-19 and its severity according to comorbidities was provided.\nThis study was based on data from patients who experienced COVID-19 during the early outbreak period; therefore, that strain may differ from the current strain of COVID-19. However, an accurate analysis of recent COVID-19 strains, including Omicron, is difficult because the effects of acquired or natural immunity and vaccination are mixed. Data at the time of its early onset can provide fundamental information, including regarding mutations that may occur in the future.", "Although the severity of COVID-19 has decreased, its hospitalization rate has not decreased significantly, and its burden on medical facilities continues; therefore, an analysis of comorbidities is still important. Therefore, many studies of comorbidities that affect COVID-19 have been published; however, some have reported conflicting results. This may be because various factors such as medication and comorbidities, in addition to demographic factors such as age and sex, affect the infection rate and severity of COVID-19. It is necessary to analyze as many factors as possible to obtain more accurate data regarding COVID-19. Based on the results of previous studies, this study tried to derive objective results by considering various factors affecting COVID-19. In conclusion, certain comorbidities known as risk factors in previous studies increase the infection rate and severity of COVID-19. However, hyperlipidemia decreases the infection rate and severity. These results can be utilized to effectively manage COVID-19." ]
[ null, null, null, null, null, null, null, null, null, null, null ]
[ "Introduction", "Methods", "Study Design and Participants", "Ethical Considerations", "Study Population", "Outcomes", "Statistical Analysis", "Patient and Public Involvement", "Results", "Clinical Characteristics of the Study Population", "Risks of COVID-19 Positivity and Disease Severity According to Comorbidities", "Discussion", "Principal Findings", "Limitations", "Conclusions" ]
[ "The World Health Organization declared that COVID-19 was a pandemic in March 2020. By August 2022, approximately 600 million individuals had been infected, and more than 6 million had died. Since then, vaccines and therapeutic agents for COVID-19 have been developed. However, the current number of individuals with COVID-19 is still the same as that 1 year ago because it has not yet been eradicated [1]. COVID-19 can result in an asymptomatic presentation or flu-like symptoms. Some patients are admitted to the hospital for conservative treatment, and some require intensive care unit admission. Moreover, some patients may die as a result of COVID-19 [2,3]. As the number of individuals with COVID-19 increases, it is important to identify those who are vulnerable to severe COVID-19 to effectively manage health care resources accordingly and to improve the prognosis [4,5].\nSince the COVID-19 outbreak, many studies of the demographic factors that predispose individuals to infection and of the identification of comorbidities of infected individuals have been performed. Most studies have reported similar overall results; however, some results of these studies are contradictory [6-9]. These differences in results may be attributable to the diversity of patients and medical systems in various countries worldwide. Most previous studies on comorbidities analyzed the baseline characteristics of people infected with COVID-19 without considering the bias caused by various factors that influence COVID-19. For instance, to determine whether hyperlipidemia affects the severity of COVID-19, it is necessary to control for statins, which are often used by individuals with hyperlipidemia. Although some studies suggested that statins might have a role in reducing the severity of COVID-19 [10,11], most studies did not confirm the use of statins; they only reported the effect of hyperlipidemia [12-16]. Hence, it is difficult to accurately determine the effect of hyperlipidemia on the severity of COVID-19. We investigated the effects of patient comorbidities on the infection rate and severity of COVID-19. Bias was reduced by propensity score (PS) matching for various variables that may affect COVID-19. We also analyzed the demographic characteristics of patients with COVID-19.", "[SUBTITLE] Study Design and Participants [SUBSECTION] We conducted a large-scale cohort study using a South Korean National Health Insurance claims database [17]. In South Korea, all citizens are registered in the Korean National Health Insurance Service (KNHIS) database. The KNHIS uses a nationwide, large-scale database system including information regarding the diagnostic codes from the International Classification of Diseases (ICD)-10, the names of the procedures performed, prescription drugs, hospital information, direct medical costs of inpatient and outpatient treatments, and medical insurance premiums. Because all Koreans are given unique identification numbers at birth that are used in the KNHIS, the health records of patients are not duplicated nor omitted [18,19]. For COVID-19 studies, KNHIS provides a COVID-19 cohort that includes people infected with COVID-19 and a control group that had never been infected. From January 1, 2020, to May 31, 2020, disease codes B342, B972, U071, U072, MT043, and 3/02 were used to identify patients with confirmed COVID-19. Data from the control group of individuals who were not previously diagnosed with COVID-19 were adjusted for sex, age, and region of residence. Moreover, the number of participants in the control group was 15 times the number of confirmed COVID-19 cases.\nWe conducted a large-scale cohort study using a South Korean National Health Insurance claims database [17]. In South Korea, all citizens are registered in the Korean National Health Insurance Service (KNHIS) database. The KNHIS uses a nationwide, large-scale database system including information regarding the diagnostic codes from the International Classification of Diseases (ICD)-10, the names of the procedures performed, prescription drugs, hospital information, direct medical costs of inpatient and outpatient treatments, and medical insurance premiums. Because all Koreans are given unique identification numbers at birth that are used in the KNHIS, the health records of patients are not duplicated nor omitted [18,19]. For COVID-19 studies, KNHIS provides a COVID-19 cohort that includes people infected with COVID-19 and a control group that had never been infected. From January 1, 2020, to May 31, 2020, disease codes B342, B972, U071, U072, MT043, and 3/02 were used to identify patients with confirmed COVID-19. Data from the control group of individuals who were not previously diagnosed with COVID-19 were adjusted for sex, age, and region of residence. Moreover, the number of participants in the control group was 15 times the number of confirmed COVID-19 cases.\n[SUBTITLE] Ethical Considerations [SUBSECTION] This study was approved by the relevant institutional review board and research ethics committee (ISPAIK 2020-06-048-001). The need for written consent was formally waived by the ethics committee. This study used the NHIS-2020-1-328 database provided by the KNHIS in 2020.\nThis study was approved by the relevant institutional review board and research ethics committee (ISPAIK 2020-06-048-001). The need for written consent was formally waived by the ethics committee. This study used the NHIS-2020-1-328 database provided by the KNHIS in 2020.\n[SUBTITLE] Study Population [SUBSECTION] In accordance with the World Health Organization guidelines, laboratory confirmation of COVID-19 was defined as a positive result of a real-time reverse-transcription polymerase chain reaction assay using a sample obtained with nasal and pharyngeal swabs [20]. We combined the claims-based data from the KNHIS between January 1, 2015, and May 31, 2020, and extracted information regarding age, sex, and region of residence from the insurance eligibility data (Figure 1). The Charlson Comorbidity Index (CCI) score was calculated using the ICD-10 codes and previously reported methods [21]. Certain underlying medications and diseases with a high risk of serious illness attributable to SARS-CoV-2, which causes COVID-19, were studied and reported by the Centers for Disease Control and Prevention (CDC) and previous meta-analysis studies [6-9,22]. In these studies, we selected factors to use for PS matching in the analysis (Tables S1 and S2 in Multimedia Appendix 1). Only those (pulmonary disease, cardiovascular disease, hepatobiliary disease, hyperlipidemia, gastrointestinal disease, diabetes mellitus, hypertension, and psychotic disorder) with more than 500 people with COVID-19 were selected because a small number of people with corresponding comorbidities might cause statistical bias (Multimedia Appendix 2). A history of underlying diseases (pulmonary disease, cardiovascular disease, kidney disease, hepatobiliary disease, hyperlipidemia, gastrointestinal disease, diabetes mellitus, hypertension, psychotic disorder, dementia, stroke, neurologic disorder, autoimmune disease, and cancer) was confirmed by the assignment of at least two claims within 1 year using the appropriate ICD-10 code.\nWe used various PS matching methods for factors affecting COVID-19: (1) matching for age, sex, and CCI; (2) additional matching for comorbidities; and (3) additional matching for medications. Finally, the results from (3) were used (Tables S3 and S4 in Multimedia Appendix 1). The financial revenue of the National Health Insurance of Korea consists of contributions from the insured and government subsidies, which can be used to analyze socioeconomic status. The contributions to the National Health Insurance differ according to the family income level. The higher the income, the greater the contribution to the National Health Insurance. Income was divided into 5 categories for the purpose of statistical analyses. The first category is Medicaid, and the successive categories include progressively higher (by 25%) income groups. Disability grades were categorized as mild or severe based on the KNHIS database information for people registered with the Korean government.\nDisposition of patients in the KNHIS-COVID cohort (South Korea; January 1 to May 31, 2020). CCI: Charlson Comorbidity Index; KNHIS: Korean National Health Insurance Service.\nIn accordance with the World Health Organization guidelines, laboratory confirmation of COVID-19 was defined as a positive result of a real-time reverse-transcription polymerase chain reaction assay using a sample obtained with nasal and pharyngeal swabs [20]. We combined the claims-based data from the KNHIS between January 1, 2015, and May 31, 2020, and extracted information regarding age, sex, and region of residence from the insurance eligibility data (Figure 1). The Charlson Comorbidity Index (CCI) score was calculated using the ICD-10 codes and previously reported methods [21]. Certain underlying medications and diseases with a high risk of serious illness attributable to SARS-CoV-2, which causes COVID-19, were studied and reported by the Centers for Disease Control and Prevention (CDC) and previous meta-analysis studies [6-9,22]. In these studies, we selected factors to use for PS matching in the analysis (Tables S1 and S2 in Multimedia Appendix 1). Only those (pulmonary disease, cardiovascular disease, hepatobiliary disease, hyperlipidemia, gastrointestinal disease, diabetes mellitus, hypertension, and psychotic disorder) with more than 500 people with COVID-19 were selected because a small number of people with corresponding comorbidities might cause statistical bias (Multimedia Appendix 2). A history of underlying diseases (pulmonary disease, cardiovascular disease, kidney disease, hepatobiliary disease, hyperlipidemia, gastrointestinal disease, diabetes mellitus, hypertension, psychotic disorder, dementia, stroke, neurologic disorder, autoimmune disease, and cancer) was confirmed by the assignment of at least two claims within 1 year using the appropriate ICD-10 code.\nWe used various PS matching methods for factors affecting COVID-19: (1) matching for age, sex, and CCI; (2) additional matching for comorbidities; and (3) additional matching for medications. Finally, the results from (3) were used (Tables S3 and S4 in Multimedia Appendix 1). The financial revenue of the National Health Insurance of Korea consists of contributions from the insured and government subsidies, which can be used to analyze socioeconomic status. The contributions to the National Health Insurance differ according to the family income level. The higher the income, the greater the contribution to the National Health Insurance. Income was divided into 5 categories for the purpose of statistical analyses. The first category is Medicaid, and the successive categories include progressively higher (by 25%) income groups. Disability grades were categorized as mild or severe based on the KNHIS database information for people registered with the Korean government.\nDisposition of patients in the KNHIS-COVID cohort (South Korea; January 1 to May 31, 2020). CCI: Charlson Comorbidity Index; KNHIS: Korean National Health Insurance Service.\n[SUBTITLE] Outcomes [SUBSECTION] To determine the severity of disease according to the demographic factors of COVID-19–infected patients, the severity scale was divided into the following 4 grades: mild, moderate, severe, and death. In South Korea, patients with asymptomatic or mild symptoms are discharged when a negative COVID-19 test result is confirmed 2 weeks after hospitalization. This time period also corresponds to the period of self-isolation. When we checked the hospitalization period of COVID-19–infected patients, the hospitalization period peaked on day 16 and decreased thereafter. Based on this result, a hospitalization period of ≤16 days was defined as the mild grade corresponding to asymptomatic or mild symptoms. The severe grade was defined as the need for tracheostomy, continuous renal replacement therapy, intensive care unit admission, ventilator use, and cardiopulmonary resuscitation. The moderate grade was defined as a hospitalization period >16 days but not requiring treatment corresponding to the severe grade.\nThe primary aim of this study was to compare the severity grades of the COVID-19–infected and control groups based on demographic factors, comorbidities, and complications. The secondary aim was to perform PS matching for comparisons. We identified the infection rate and severity (severe and death or mild and moderate) of COVID-19 according to the comorbid conditions.\nTo determine the severity of disease according to the demographic factors of COVID-19–infected patients, the severity scale was divided into the following 4 grades: mild, moderate, severe, and death. In South Korea, patients with asymptomatic or mild symptoms are discharged when a negative COVID-19 test result is confirmed 2 weeks after hospitalization. This time period also corresponds to the period of self-isolation. When we checked the hospitalization period of COVID-19–infected patients, the hospitalization period peaked on day 16 and decreased thereafter. Based on this result, a hospitalization period of ≤16 days was defined as the mild grade corresponding to asymptomatic or mild symptoms. The severe grade was defined as the need for tracheostomy, continuous renal replacement therapy, intensive care unit admission, ventilator use, and cardiopulmonary resuscitation. The moderate grade was defined as a hospitalization period >16 days but not requiring treatment corresponding to the severe grade.\nThe primary aim of this study was to compare the severity grades of the COVID-19–infected and control groups based on demographic factors, comorbidities, and complications. The secondary aim was to perform PS matching for comparisons. We identified the infection rate and severity (severe and death or mild and moderate) of COVID-19 according to the comorbid conditions.\n[SUBTITLE] Statistical Analysis [SUBSECTION] We performed PS matching to balance the baseline characteristics of each comorbidity (existence or nonexistence) and to adjust for potential confounders. Because we focused on each comorbidity, PS matching was performed for each comorbidity. The PS was estimated using a logistic regression model and calculating the predicted probability of covariates. Age and CCI (0, 1, or ≥2) were matched with continuous variables. Sex, medication, and other comorbidities were matched with binary variables. We assessed the PS matching of the comorbidity existence using a 1:1 ratio, the greedy nearest neighbor algorithm, and a scale with a caliper of 0.25 (Multimedia Appendix 2). Data obtained after PS matching were analyzed by calculating the odds ratios (ORs) with 95% CIs for the infection rate and severity (severe and death or mild and moderate) of COVID-19. Statistical analyses were performed using SAS version 9.4 (SAS Institute Inc, Cary, NC).\nWe performed PS matching to balance the baseline characteristics of each comorbidity (existence or nonexistence) and to adjust for potential confounders. Because we focused on each comorbidity, PS matching was performed for each comorbidity. The PS was estimated using a logistic regression model and calculating the predicted probability of covariates. Age and CCI (0, 1, or ≥2) were matched with continuous variables. Sex, medication, and other comorbidities were matched with binary variables. We assessed the PS matching of the comorbidity existence using a 1:1 ratio, the greedy nearest neighbor algorithm, and a scale with a caliper of 0.25 (Multimedia Appendix 2). Data obtained after PS matching were analyzed by calculating the odds ratios (ORs) with 95% CIs for the infection rate and severity (severe and death or mild and moderate) of COVID-19. Statistical analyses were performed using SAS version 9.4 (SAS Institute Inc, Cary, NC).\n[SUBTITLE] Patient and Public Involvement [SUBSECTION] No patient was directly involved in designing the study question or in conducting the study. No patients were asked for advice regarding the interpretation or writing of the results. There are no plans to involve patients or the relevant patient community in the dissemination of study findings at this time.\nNo patient was directly involved in designing the study question or in conducting the study. No patients were asked for advice regarding the interpretation or writing of the results. There are no plans to involve patients or the relevant patient community in the dissemination of study findings at this time.", "We conducted a large-scale cohort study using a South Korean National Health Insurance claims database [17]. In South Korea, all citizens are registered in the Korean National Health Insurance Service (KNHIS) database. The KNHIS uses a nationwide, large-scale database system including information regarding the diagnostic codes from the International Classification of Diseases (ICD)-10, the names of the procedures performed, prescription drugs, hospital information, direct medical costs of inpatient and outpatient treatments, and medical insurance premiums. Because all Koreans are given unique identification numbers at birth that are used in the KNHIS, the health records of patients are not duplicated nor omitted [18,19]. For COVID-19 studies, KNHIS provides a COVID-19 cohort that includes people infected with COVID-19 and a control group that had never been infected. From January 1, 2020, to May 31, 2020, disease codes B342, B972, U071, U072, MT043, and 3/02 were used to identify patients with confirmed COVID-19. Data from the control group of individuals who were not previously diagnosed with COVID-19 were adjusted for sex, age, and region of residence. Moreover, the number of participants in the control group was 15 times the number of confirmed COVID-19 cases.", "This study was approved by the relevant institutional review board and research ethics committee (ISPAIK 2020-06-048-001). The need for written consent was formally waived by the ethics committee. This study used the NHIS-2020-1-328 database provided by the KNHIS in 2020.", "In accordance with the World Health Organization guidelines, laboratory confirmation of COVID-19 was defined as a positive result of a real-time reverse-transcription polymerase chain reaction assay using a sample obtained with nasal and pharyngeal swabs [20]. We combined the claims-based data from the KNHIS between January 1, 2015, and May 31, 2020, and extracted information regarding age, sex, and region of residence from the insurance eligibility data (Figure 1). The Charlson Comorbidity Index (CCI) score was calculated using the ICD-10 codes and previously reported methods [21]. Certain underlying medications and diseases with a high risk of serious illness attributable to SARS-CoV-2, which causes COVID-19, were studied and reported by the Centers for Disease Control and Prevention (CDC) and previous meta-analysis studies [6-9,22]. In these studies, we selected factors to use for PS matching in the analysis (Tables S1 and S2 in Multimedia Appendix 1). Only those (pulmonary disease, cardiovascular disease, hepatobiliary disease, hyperlipidemia, gastrointestinal disease, diabetes mellitus, hypertension, and psychotic disorder) with more than 500 people with COVID-19 were selected because a small number of people with corresponding comorbidities might cause statistical bias (Multimedia Appendix 2). A history of underlying diseases (pulmonary disease, cardiovascular disease, kidney disease, hepatobiliary disease, hyperlipidemia, gastrointestinal disease, diabetes mellitus, hypertension, psychotic disorder, dementia, stroke, neurologic disorder, autoimmune disease, and cancer) was confirmed by the assignment of at least two claims within 1 year using the appropriate ICD-10 code.\nWe used various PS matching methods for factors affecting COVID-19: (1) matching for age, sex, and CCI; (2) additional matching for comorbidities; and (3) additional matching for medications. Finally, the results from (3) were used (Tables S3 and S4 in Multimedia Appendix 1). The financial revenue of the National Health Insurance of Korea consists of contributions from the insured and government subsidies, which can be used to analyze socioeconomic status. The contributions to the National Health Insurance differ according to the family income level. The higher the income, the greater the contribution to the National Health Insurance. Income was divided into 5 categories for the purpose of statistical analyses. The first category is Medicaid, and the successive categories include progressively higher (by 25%) income groups. Disability grades were categorized as mild or severe based on the KNHIS database information for people registered with the Korean government.\nDisposition of patients in the KNHIS-COVID cohort (South Korea; January 1 to May 31, 2020). CCI: Charlson Comorbidity Index; KNHIS: Korean National Health Insurance Service.", "To determine the severity of disease according to the demographic factors of COVID-19–infected patients, the severity scale was divided into the following 4 grades: mild, moderate, severe, and death. In South Korea, patients with asymptomatic or mild symptoms are discharged when a negative COVID-19 test result is confirmed 2 weeks after hospitalization. This time period also corresponds to the period of self-isolation. When we checked the hospitalization period of COVID-19–infected patients, the hospitalization period peaked on day 16 and decreased thereafter. Based on this result, a hospitalization period of ≤16 days was defined as the mild grade corresponding to asymptomatic or mild symptoms. The severe grade was defined as the need for tracheostomy, continuous renal replacement therapy, intensive care unit admission, ventilator use, and cardiopulmonary resuscitation. The moderate grade was defined as a hospitalization period >16 days but not requiring treatment corresponding to the severe grade.\nThe primary aim of this study was to compare the severity grades of the COVID-19–infected and control groups based on demographic factors, comorbidities, and complications. The secondary aim was to perform PS matching for comparisons. We identified the infection rate and severity (severe and death or mild and moderate) of COVID-19 according to the comorbid conditions.", "We performed PS matching to balance the baseline characteristics of each comorbidity (existence or nonexistence) and to adjust for potential confounders. Because we focused on each comorbidity, PS matching was performed for each comorbidity. The PS was estimated using a logistic regression model and calculating the predicted probability of covariates. Age and CCI (0, 1, or ≥2) were matched with continuous variables. Sex, medication, and other comorbidities were matched with binary variables. We assessed the PS matching of the comorbidity existence using a 1:1 ratio, the greedy nearest neighbor algorithm, and a scale with a caliper of 0.25 (Multimedia Appendix 2). Data obtained after PS matching were analyzed by calculating the odds ratios (ORs) with 95% CIs for the infection rate and severity (severe and death or mild and moderate) of COVID-19. Statistical analyses were performed using SAS version 9.4 (SAS Institute Inc, Cary, NC).", "No patient was directly involved in designing the study question or in conducting the study. No patients were asked for advice regarding the interpretation or writing of the results. There are no plans to involve patients or the relevant patient community in the dissemination of study findings at this time.", "[SUBTITLE] Clinical Characteristics of the Study Population [SUBSECTION] A total of 8070 individuals had positive COVID-19 results according to the reverse-transcription polymerase chain reaction assay. We identified 121,050 uninfected individuals as control participants (Multimedia Appendix 2). The demographic characteristics of the entire cohort are displayed in Table 1. The COVID-19 severity grade was mild for 2419 (2419/8070, 29.98%) individuals, moderate for 5160 (5160/8070, 63.94%) individuals, severe for 254 (254/8070, 3.15%) individuals, and death for 237 (237/8070, 2.94%) individuals. Among the total sample of infected individuals, 3236 (3236/8070, 40.10%) were male. Most patients were in their fifth (1567/8070, 19.42%) or sixth (1199/8070, 14.86%) decade of life. In terms of the medical insurance grade, which indicates socioeconomic status, those receiving Medicaid had high rates of severe grade and death. However, there were no obvious trends for the other grades. Individuals with disabilities had more severe infections and a much higher case fatality rate (Table 1). Those with COVID-19 had a medical history of gastrointestinal disease (n=5256), pulmonary disease (n=2539), hyperlipidemia (n=1841), and hypertension (n=1623). The case fatality rate was high for individuals with dementia (74/235, 31.5%), kidney disease (25/86, 29%), and cardiovascular disease (110/675, 16.3%; Table 2). After COVID-19 was confirmed, gastrointestinal disease (n=2912), pulmonary disease (n=2398), and hepatobiliary disease (n=1248) were the most common complications (Table 3).\nBaseline characteristics of the study population, including those infected (n=8070) and uninfected (n=121,050; controls) with COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\naParticipants from some specific groups, such as soldiers, were not included.\nbThe uninfected controls were adjusted for sex, age, and region, resulting in a figure equivalent to 15 times the number of confirmed COVID-19 cases in the KNHIS-COVID cohort.\nBaseline characteristics of comorbidities of the study population, including those infected with (n=8070) and not infected with (n=121,050; controls) COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\nBaseline characteristics of complications of the study population, including those infected with (n=8070) and not infected with (n=121,050; controls) COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\nA total of 8070 individuals had positive COVID-19 results according to the reverse-transcription polymerase chain reaction assay. We identified 121,050 uninfected individuals as control participants (Multimedia Appendix 2). The demographic characteristics of the entire cohort are displayed in Table 1. The COVID-19 severity grade was mild for 2419 (2419/8070, 29.98%) individuals, moderate for 5160 (5160/8070, 63.94%) individuals, severe for 254 (254/8070, 3.15%) individuals, and death for 237 (237/8070, 2.94%) individuals. Among the total sample of infected individuals, 3236 (3236/8070, 40.10%) were male. Most patients were in their fifth (1567/8070, 19.42%) or sixth (1199/8070, 14.86%) decade of life. In terms of the medical insurance grade, which indicates socioeconomic status, those receiving Medicaid had high rates of severe grade and death. However, there were no obvious trends for the other grades. Individuals with disabilities had more severe infections and a much higher case fatality rate (Table 1). Those with COVID-19 had a medical history of gastrointestinal disease (n=5256), pulmonary disease (n=2539), hyperlipidemia (n=1841), and hypertension (n=1623). The case fatality rate was high for individuals with dementia (74/235, 31.5%), kidney disease (25/86, 29%), and cardiovascular disease (110/675, 16.3%; Table 2). After COVID-19 was confirmed, gastrointestinal disease (n=2912), pulmonary disease (n=2398), and hepatobiliary disease (n=1248) were the most common complications (Table 3).\nBaseline characteristics of the study population, including those infected (n=8070) and uninfected (n=121,050; controls) with COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\naParticipants from some specific groups, such as soldiers, were not included.\nbThe uninfected controls were adjusted for sex, age, and region, resulting in a figure equivalent to 15 times the number of confirmed COVID-19 cases in the KNHIS-COVID cohort.\nBaseline characteristics of comorbidities of the study population, including those infected with (n=8070) and not infected with (n=121,050; controls) COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\nBaseline characteristics of complications of the study population, including those infected with (n=8070) and not infected with (n=121,050; controls) COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\n[SUBTITLE] Risks of COVID-19 Positivity and Disease Severity According to Comorbidities [SUBSECTION] To identify differences according to comorbidity, predispositions were matched between the COVID-19–infected group and uninfected control group. No significant imbalances in the demographics and clinical characteristics were observed when they were assessed using the standardized mean difference within groups of PS-matched cohorts, which included the standardized mean difference of binary type variables <0.1. PS-matched ORs were checked for age, sex, CCI, medication, and comorbidities. When the control group and COVID-19–infected group were compared, COVID-19 was likely to occur in individuals with a history of the diseases and medical conditions but not for those with a history of hyperlipidemia (OR 0.73; 95% CI 0.67-0.80), autoimmune disease (OR 0.73; 95% CI 0.60-0.89), or cancer (OR 0.73; 95% CI 0.62-0.86; Table 4). The severity grade was high for COVID-19–infected individuals with pulmonary disease (OR 1.72; 95% CI 1.35-2.19), cardiovascular disease (OR 1.54; 95% CI 1.17-2.04), kidney disease (OR 5.59; 95% CI 2.48-12.63), diabetes mellitus (OR 1.43; 95% CI 1.09-1.87), hypertension (OR 1.63; 95% CI 1.23-2.15), psychotic disorder (OR 1.29; 95% CI 1.01-6.52), dementia (OR 2.92; 95% CI 1.91-4.47), or cancer (OR 1.84; 95% CI 1.15-2.94). However, the severity grade was low for COVID-19–infected individuals with hyperlipidemia (OR 0.70; 95% CI 0.55-0.90; Table 5 and Table S5 in Multimedia Appendix 1).\nPropensity score–matched (age, sex, Charlson Comorbidity Index, medications, and comorbidities) baseline characteristics and COVID-19 infection positivity rates according to comorbidity in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\naWe assessed each propensity score–matched comorbidity using a 1:1 ratio for those in the COVID-19 and control groups.\nbComorbidity with more susceptibility to COVID-19.\ncComorbidity with less susceptibility to COVID-19.\nPropensity score–matched (age, sex, Charlson Comorbidity Index, medications, and comorbidities) baseline characteristics and clinical outcomes of COVID-19 among patients in the mild or moderate group and those in the severe or death group according to the comorbidity of patients with laboratory-confirmed COVID-19 infection in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\naWe assessed each propensity score–matched comorbidity using a 1:1 ratio for those in the mild and moderate group and those in the severe and death group.\nbComorbidity with increasing COVID-19 severity.\ncComorbidity with decreasing COVID-19 severity.\nTo identify differences according to comorbidity, predispositions were matched between the COVID-19–infected group and uninfected control group. No significant imbalances in the demographics and clinical characteristics were observed when they were assessed using the standardized mean difference within groups of PS-matched cohorts, which included the standardized mean difference of binary type variables <0.1. PS-matched ORs were checked for age, sex, CCI, medication, and comorbidities. When the control group and COVID-19–infected group were compared, COVID-19 was likely to occur in individuals with a history of the diseases and medical conditions but not for those with a history of hyperlipidemia (OR 0.73; 95% CI 0.67-0.80), autoimmune disease (OR 0.73; 95% CI 0.60-0.89), or cancer (OR 0.73; 95% CI 0.62-0.86; Table 4). The severity grade was high for COVID-19–infected individuals with pulmonary disease (OR 1.72; 95% CI 1.35-2.19), cardiovascular disease (OR 1.54; 95% CI 1.17-2.04), kidney disease (OR 5.59; 95% CI 2.48-12.63), diabetes mellitus (OR 1.43; 95% CI 1.09-1.87), hypertension (OR 1.63; 95% CI 1.23-2.15), psychotic disorder (OR 1.29; 95% CI 1.01-6.52), dementia (OR 2.92; 95% CI 1.91-4.47), or cancer (OR 1.84; 95% CI 1.15-2.94). However, the severity grade was low for COVID-19–infected individuals with hyperlipidemia (OR 0.70; 95% CI 0.55-0.90; Table 5 and Table S5 in Multimedia Appendix 1).\nPropensity score–matched (age, sex, Charlson Comorbidity Index, medications, and comorbidities) baseline characteristics and COVID-19 infection positivity rates according to comorbidity in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\naWe assessed each propensity score–matched comorbidity using a 1:1 ratio for those in the COVID-19 and control groups.\nbComorbidity with more susceptibility to COVID-19.\ncComorbidity with less susceptibility to COVID-19.\nPropensity score–matched (age, sex, Charlson Comorbidity Index, medications, and comorbidities) baseline characteristics and clinical outcomes of COVID-19 among patients in the mild or moderate group and those in the severe or death group according to the comorbidity of patients with laboratory-confirmed COVID-19 infection in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\naWe assessed each propensity score–matched comorbidity using a 1:1 ratio for those in the mild and moderate group and those in the severe and death group.\nbComorbidity with increasing COVID-19 severity.\ncComorbidity with decreasing COVID-19 severity.", "A total of 8070 individuals had positive COVID-19 results according to the reverse-transcription polymerase chain reaction assay. We identified 121,050 uninfected individuals as control participants (Multimedia Appendix 2). The demographic characteristics of the entire cohort are displayed in Table 1. The COVID-19 severity grade was mild for 2419 (2419/8070, 29.98%) individuals, moderate for 5160 (5160/8070, 63.94%) individuals, severe for 254 (254/8070, 3.15%) individuals, and death for 237 (237/8070, 2.94%) individuals. Among the total sample of infected individuals, 3236 (3236/8070, 40.10%) were male. Most patients were in their fifth (1567/8070, 19.42%) or sixth (1199/8070, 14.86%) decade of life. In terms of the medical insurance grade, which indicates socioeconomic status, those receiving Medicaid had high rates of severe grade and death. However, there were no obvious trends for the other grades. Individuals with disabilities had more severe infections and a much higher case fatality rate (Table 1). Those with COVID-19 had a medical history of gastrointestinal disease (n=5256), pulmonary disease (n=2539), hyperlipidemia (n=1841), and hypertension (n=1623). The case fatality rate was high for individuals with dementia (74/235, 31.5%), kidney disease (25/86, 29%), and cardiovascular disease (110/675, 16.3%; Table 2). After COVID-19 was confirmed, gastrointestinal disease (n=2912), pulmonary disease (n=2398), and hepatobiliary disease (n=1248) were the most common complications (Table 3).\nBaseline characteristics of the study population, including those infected (n=8070) and uninfected (n=121,050; controls) with COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\naParticipants from some specific groups, such as soldiers, were not included.\nbThe uninfected controls were adjusted for sex, age, and region, resulting in a figure equivalent to 15 times the number of confirmed COVID-19 cases in the KNHIS-COVID cohort.\nBaseline characteristics of comorbidities of the study population, including those infected with (n=8070) and not infected with (n=121,050; controls) COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\nBaseline characteristics of complications of the study population, including those infected with (n=8070) and not infected with (n=121,050; controls) COVID-19 in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).", "To identify differences according to comorbidity, predispositions were matched between the COVID-19–infected group and uninfected control group. No significant imbalances in the demographics and clinical characteristics were observed when they were assessed using the standardized mean difference within groups of PS-matched cohorts, which included the standardized mean difference of binary type variables <0.1. PS-matched ORs were checked for age, sex, CCI, medication, and comorbidities. When the control group and COVID-19–infected group were compared, COVID-19 was likely to occur in individuals with a history of the diseases and medical conditions but not for those with a history of hyperlipidemia (OR 0.73; 95% CI 0.67-0.80), autoimmune disease (OR 0.73; 95% CI 0.60-0.89), or cancer (OR 0.73; 95% CI 0.62-0.86; Table 4). The severity grade was high for COVID-19–infected individuals with pulmonary disease (OR 1.72; 95% CI 1.35-2.19), cardiovascular disease (OR 1.54; 95% CI 1.17-2.04), kidney disease (OR 5.59; 95% CI 2.48-12.63), diabetes mellitus (OR 1.43; 95% CI 1.09-1.87), hypertension (OR 1.63; 95% CI 1.23-2.15), psychotic disorder (OR 1.29; 95% CI 1.01-6.52), dementia (OR 2.92; 95% CI 1.91-4.47), or cancer (OR 1.84; 95% CI 1.15-2.94). However, the severity grade was low for COVID-19–infected individuals with hyperlipidemia (OR 0.70; 95% CI 0.55-0.90; Table 5 and Table S5 in Multimedia Appendix 1).\nPropensity score–matched (age, sex, Charlson Comorbidity Index, medications, and comorbidities) baseline characteristics and COVID-19 infection positivity rates according to comorbidity in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\naWe assessed each propensity score–matched comorbidity using a 1:1 ratio for those in the COVID-19 and control groups.\nbComorbidity with more susceptibility to COVID-19.\ncComorbidity with less susceptibility to COVID-19.\nPropensity score–matched (age, sex, Charlson Comorbidity Index, medications, and comorbidities) baseline characteristics and clinical outcomes of COVID-19 among patients in the mild or moderate group and those in the severe or death group according to the comorbidity of patients with laboratory-confirmed COVID-19 infection in the Korean National Health Insurance Service (KNHIS)-COVID cohort (South Korea; January 1, 2020, to May 31, 2020).\naWe assessed each propensity score–matched comorbidity using a 1:1 ratio for those in the mild and moderate group and those in the severe and death group.\nbComorbidity with increasing COVID-19 severity.\ncComorbidity with decreasing COVID-19 severity.", "[SUBTITLE] Principal Findings [SUBSECTION] This study was a retrospective cohort study conducted in South Korea from January 2020 to May 2020. It involved confirmed COVID-19 patients with medical insurance. Previous studies of the demographic factors of individuals with COVID-19 showed that male sex, old age, and low income were factors likely associated with COVID-19 with a high severity grade [23,24]. In this study, more women had COVID-19, but the severity grade of COVID-19 was higher for men; this was directly proportional to age, especially for men older than 70 years. All medical expenses for COVID-19 are paid for by the South Korea government; therefore, all patients, including those receiving Medicaid, received the same level of care for COVID-19. Although there was no difference in medical care, those with Medicaid had the lowest income level and a higher severity grade; however, there were no differences between the groups with grades 1 to 4 medical insurance. For individuals with disabilities, the incidence was slightly higher than that of the control group. However, the severity grade was much higher than that of other individuals infected with COVID-19.\nOther studies of COVID-19 reported that SARS-CoV-2 binds to the angiotensin-converting enzyme 2 (ACE2) receptor through the viral structural spike protein at the onset of infection [25]. ACE2 is expressed to varying degrees in almost all human organs. ACE2 is highly expressed in cardiomyocytes, proximal tubule cells of the kidney, and bladder urinary tract cells. Additionally, it is abundantly expressed in intestinal cells of the small intestine, especially in the ileum [25-28]. Therefore, most critically ill patients with COVID-19 experience multiple organ injuries, including acute lung injuries, acute kidney injuries, cardiac injuries, hepatobiliary disease, and pneumothorax [29]. Therefore, to analyze the effect of each comorbidity on the COVID-19 infection severity grade, it is necessary to consider other comorbidities.\nEach demographic factor, comorbidity, and medication may influence each other, resulting in different outcomes in terms of the infection rate and severity of COVID-19. When analyzing comorbidities with hypertension, the effect of hypertension on the infection rate and severity of COVID-19 experienced by an 80-year-old woman with asthma and that of a 30-year-old man without an underlying medical condition may be different. Accurate results can be obtained for sufficiently studied diseases by controlling for only important factors. However, in the case of understudied diseases, such as COVID-19, various factors should be considered. In this study, PS matching was performed for various factors that could affect COVID-19, to minimize bias. When selecting a factor for PS matching, in order to select objective data, data provided by the CDC and meta-analysis studies were used. However, there was a limit, as data may change as research on COVID-19 progresses. Most of the results obtained were similar to those of previously published studies; however, some results were conflicting. For people with cancer and autoimmune disease, infection rates were even lower; these results were possibly affected by reducing social contact because of the risk of COVID-19 infection. Exposure to COVID-19 is an important factor that can affect the infection rate of COVID-19. Individuals with hyperlipidemia had a low COVID-19 infection rate and low severity grade. Previous studies reported that hyperlipidemia should be managed to prevent COVID-19 because high cholesterol levels induce inflammation and increase ACE2 availability [30-32]. Moreover, the use of statins for patients with COVID-19 reduced mortality by interfering with the mevalonate pathway and because of their antiviral effects [10,11,33]. However, some studies have shown that people with low lipid levels are more susceptible to and have more severe COVID-19 infection [34-41]. A meta-analysis published in 2022 indicated that patients with severe COVID-19 had lower total cholesterol levels (pooled mean difference –10.4; 95% CI –18.7 to –2.2), low-density lipoprotein cholesterol levels (pooled mean difference –4.4; 95% CI –8.4 to –0.42), and high-density lipoprotein cholesterol levels (pooled mean difference –4.4; 95% CI –6.9 to –1.8) on admission compared with patients with non-severe disease [42]. This may be similar to the “obesity paradox,” which states that mild obesity is advantageous to improvements after stroke [43,44]. Mild obesity can withstand the systemic catabolic imbalance with impaired metabolic efficiency and body tissue degradation that occur after stroke. Hyperlipidemia may also have a role in minimizing the severity of COVID-19.\nThis study was a retrospective cohort study conducted in South Korea from January 2020 to May 2020. It involved confirmed COVID-19 patients with medical insurance. Previous studies of the demographic factors of individuals with COVID-19 showed that male sex, old age, and low income were factors likely associated with COVID-19 with a high severity grade [23,24]. In this study, more women had COVID-19, but the severity grade of COVID-19 was higher for men; this was directly proportional to age, especially for men older than 70 years. All medical expenses for COVID-19 are paid for by the South Korea government; therefore, all patients, including those receiving Medicaid, received the same level of care for COVID-19. Although there was no difference in medical care, those with Medicaid had the lowest income level and a higher severity grade; however, there were no differences between the groups with grades 1 to 4 medical insurance. For individuals with disabilities, the incidence was slightly higher than that of the control group. However, the severity grade was much higher than that of other individuals infected with COVID-19.\nOther studies of COVID-19 reported that SARS-CoV-2 binds to the angiotensin-converting enzyme 2 (ACE2) receptor through the viral structural spike protein at the onset of infection [25]. ACE2 is expressed to varying degrees in almost all human organs. ACE2 is highly expressed in cardiomyocytes, proximal tubule cells of the kidney, and bladder urinary tract cells. Additionally, it is abundantly expressed in intestinal cells of the small intestine, especially in the ileum [25-28]. Therefore, most critically ill patients with COVID-19 experience multiple organ injuries, including acute lung injuries, acute kidney injuries, cardiac injuries, hepatobiliary disease, and pneumothorax [29]. Therefore, to analyze the effect of each comorbidity on the COVID-19 infection severity grade, it is necessary to consider other comorbidities.\nEach demographic factor, comorbidity, and medication may influence each other, resulting in different outcomes in terms of the infection rate and severity of COVID-19. When analyzing comorbidities with hypertension, the effect of hypertension on the infection rate and severity of COVID-19 experienced by an 80-year-old woman with asthma and that of a 30-year-old man without an underlying medical condition may be different. Accurate results can be obtained for sufficiently studied diseases by controlling for only important factors. However, in the case of understudied diseases, such as COVID-19, various factors should be considered. In this study, PS matching was performed for various factors that could affect COVID-19, to minimize bias. When selecting a factor for PS matching, in order to select objective data, data provided by the CDC and meta-analysis studies were used. However, there was a limit, as data may change as research on COVID-19 progresses. Most of the results obtained were similar to those of previously published studies; however, some results were conflicting. For people with cancer and autoimmune disease, infection rates were even lower; these results were possibly affected by reducing social contact because of the risk of COVID-19 infection. Exposure to COVID-19 is an important factor that can affect the infection rate of COVID-19. Individuals with hyperlipidemia had a low COVID-19 infection rate and low severity grade. Previous studies reported that hyperlipidemia should be managed to prevent COVID-19 because high cholesterol levels induce inflammation and increase ACE2 availability [30-32]. Moreover, the use of statins for patients with COVID-19 reduced mortality by interfering with the mevalonate pathway and because of their antiviral effects [10,11,33]. However, some studies have shown that people with low lipid levels are more susceptible to and have more severe COVID-19 infection [34-41]. A meta-analysis published in 2022 indicated that patients with severe COVID-19 had lower total cholesterol levels (pooled mean difference –10.4; 95% CI –18.7 to –2.2), low-density lipoprotein cholesterol levels (pooled mean difference –4.4; 95% CI –8.4 to –0.42), and high-density lipoprotein cholesterol levels (pooled mean difference –4.4; 95% CI –6.9 to –1.8) on admission compared with patients with non-severe disease [42]. This may be similar to the “obesity paradox,” which states that mild obesity is advantageous to improvements after stroke [43,44]. Mild obesity can withstand the systemic catabolic imbalance with impaired metabolic efficiency and body tissue degradation that occur after stroke. Hyperlipidemia may also have a role in minimizing the severity of COVID-19.\n[SUBTITLE] Limitations [SUBSECTION] Our study has several limitations. As a limitation of most medical data studies, there is bias caused by confounding factors that may affect our results. When selecting factors for PS matching, information from the CDC and meta-analysis studies were used to select objective data, but these data may change as research on COVID-19 progresses. Further, we defined diseases based on the ICD codes provided in the insurance claims data. There may have been additional unmeasured confounders influencing our results, including genetic polymorphisms, smoking, body mass index, and exposure to the virus. In this study, the infection rate of COVID-19 may have been influenced by the degree of exposure to COVID-19, which may be an important factor in addition to the comorbidity factors. However, the influence of COVID-19 itself could be confirmed because the bias was less than that of previous studies. One race in South Korea comprises more than 95% of the population; hence, there was minimal racial bias compared with previous studies. Because the government funds the treatment for COVID-19 in South Korea and because the medical facilities for COVID-19 treatment are ubiquitous, there was minimal economic bias. The PS matching was performed for sex, age, CCI, comorbidity, and medication, including statins (standardized mean difference <0.1). Hence, selection bias was minimized. Therefore, more accurate information regarding the incidence of COVID-19 and its severity according to comorbidities was provided.\nThis study was based on data from patients who experienced COVID-19 during the early outbreak period; therefore, that strain may differ from the current strain of COVID-19. However, an accurate analysis of recent COVID-19 strains, including Omicron, is difficult because the effects of acquired or natural immunity and vaccination are mixed. Data at the time of its early onset can provide fundamental information, including regarding mutations that may occur in the future.\nOur study has several limitations. As a limitation of most medical data studies, there is bias caused by confounding factors that may affect our results. When selecting factors for PS matching, information from the CDC and meta-analysis studies were used to select objective data, but these data may change as research on COVID-19 progresses. Further, we defined diseases based on the ICD codes provided in the insurance claims data. There may have been additional unmeasured confounders influencing our results, including genetic polymorphisms, smoking, body mass index, and exposure to the virus. In this study, the infection rate of COVID-19 may have been influenced by the degree of exposure to COVID-19, which may be an important factor in addition to the comorbidity factors. However, the influence of COVID-19 itself could be confirmed because the bias was less than that of previous studies. One race in South Korea comprises more than 95% of the population; hence, there was minimal racial bias compared with previous studies. Because the government funds the treatment for COVID-19 in South Korea and because the medical facilities for COVID-19 treatment are ubiquitous, there was minimal economic bias. The PS matching was performed for sex, age, CCI, comorbidity, and medication, including statins (standardized mean difference <0.1). Hence, selection bias was minimized. Therefore, more accurate information regarding the incidence of COVID-19 and its severity according to comorbidities was provided.\nThis study was based on data from patients who experienced COVID-19 during the early outbreak period; therefore, that strain may differ from the current strain of COVID-19. However, an accurate analysis of recent COVID-19 strains, including Omicron, is difficult because the effects of acquired or natural immunity and vaccination are mixed. Data at the time of its early onset can provide fundamental information, including regarding mutations that may occur in the future.\n[SUBTITLE] Conclusions [SUBSECTION] Although the severity of COVID-19 has decreased, its hospitalization rate has not decreased significantly, and its burden on medical facilities continues; therefore, an analysis of comorbidities is still important. Therefore, many studies of comorbidities that affect COVID-19 have been published; however, some have reported conflicting results. This may be because various factors such as medication and comorbidities, in addition to demographic factors such as age and sex, affect the infection rate and severity of COVID-19. It is necessary to analyze as many factors as possible to obtain more accurate data regarding COVID-19. Based on the results of previous studies, this study tried to derive objective results by considering various factors affecting COVID-19. In conclusion, certain comorbidities known as risk factors in previous studies increase the infection rate and severity of COVID-19. However, hyperlipidemia decreases the infection rate and severity. These results can be utilized to effectively manage COVID-19.\nAlthough the severity of COVID-19 has decreased, its hospitalization rate has not decreased significantly, and its burden on medical facilities continues; therefore, an analysis of comorbidities is still important. Therefore, many studies of comorbidities that affect COVID-19 have been published; however, some have reported conflicting results. This may be because various factors such as medication and comorbidities, in addition to demographic factors such as age and sex, affect the infection rate and severity of COVID-19. It is necessary to analyze as many factors as possible to obtain more accurate data regarding COVID-19. Based on the results of previous studies, this study tried to derive objective results by considering various factors affecting COVID-19. In conclusion, certain comorbidities known as risk factors in previous studies increase the infection rate and severity of COVID-19. However, hyperlipidemia decreases the infection rate and severity. These results can be utilized to effectively manage COVID-19.", "This study was a retrospective cohort study conducted in South Korea from January 2020 to May 2020. It involved confirmed COVID-19 patients with medical insurance. Previous studies of the demographic factors of individuals with COVID-19 showed that male sex, old age, and low income were factors likely associated with COVID-19 with a high severity grade [23,24]. In this study, more women had COVID-19, but the severity grade of COVID-19 was higher for men; this was directly proportional to age, especially for men older than 70 years. All medical expenses for COVID-19 are paid for by the South Korea government; therefore, all patients, including those receiving Medicaid, received the same level of care for COVID-19. Although there was no difference in medical care, those with Medicaid had the lowest income level and a higher severity grade; however, there were no differences between the groups with grades 1 to 4 medical insurance. For individuals with disabilities, the incidence was slightly higher than that of the control group. However, the severity grade was much higher than that of other individuals infected with COVID-19.\nOther studies of COVID-19 reported that SARS-CoV-2 binds to the angiotensin-converting enzyme 2 (ACE2) receptor through the viral structural spike protein at the onset of infection [25]. ACE2 is expressed to varying degrees in almost all human organs. ACE2 is highly expressed in cardiomyocytes, proximal tubule cells of the kidney, and bladder urinary tract cells. Additionally, it is abundantly expressed in intestinal cells of the small intestine, especially in the ileum [25-28]. Therefore, most critically ill patients with COVID-19 experience multiple organ injuries, including acute lung injuries, acute kidney injuries, cardiac injuries, hepatobiliary disease, and pneumothorax [29]. Therefore, to analyze the effect of each comorbidity on the COVID-19 infection severity grade, it is necessary to consider other comorbidities.\nEach demographic factor, comorbidity, and medication may influence each other, resulting in different outcomes in terms of the infection rate and severity of COVID-19. When analyzing comorbidities with hypertension, the effect of hypertension on the infection rate and severity of COVID-19 experienced by an 80-year-old woman with asthma and that of a 30-year-old man without an underlying medical condition may be different. Accurate results can be obtained for sufficiently studied diseases by controlling for only important factors. However, in the case of understudied diseases, such as COVID-19, various factors should be considered. In this study, PS matching was performed for various factors that could affect COVID-19, to minimize bias. When selecting a factor for PS matching, in order to select objective data, data provided by the CDC and meta-analysis studies were used. However, there was a limit, as data may change as research on COVID-19 progresses. Most of the results obtained were similar to those of previously published studies; however, some results were conflicting. For people with cancer and autoimmune disease, infection rates were even lower; these results were possibly affected by reducing social contact because of the risk of COVID-19 infection. Exposure to COVID-19 is an important factor that can affect the infection rate of COVID-19. Individuals with hyperlipidemia had a low COVID-19 infection rate and low severity grade. Previous studies reported that hyperlipidemia should be managed to prevent COVID-19 because high cholesterol levels induce inflammation and increase ACE2 availability [30-32]. Moreover, the use of statins for patients with COVID-19 reduced mortality by interfering with the mevalonate pathway and because of their antiviral effects [10,11,33]. However, some studies have shown that people with low lipid levels are more susceptible to and have more severe COVID-19 infection [34-41]. A meta-analysis published in 2022 indicated that patients with severe COVID-19 had lower total cholesterol levels (pooled mean difference –10.4; 95% CI –18.7 to –2.2), low-density lipoprotein cholesterol levels (pooled mean difference –4.4; 95% CI –8.4 to –0.42), and high-density lipoprotein cholesterol levels (pooled mean difference –4.4; 95% CI –6.9 to –1.8) on admission compared with patients with non-severe disease [42]. This may be similar to the “obesity paradox,” which states that mild obesity is advantageous to improvements after stroke [43,44]. Mild obesity can withstand the systemic catabolic imbalance with impaired metabolic efficiency and body tissue degradation that occur after stroke. Hyperlipidemia may also have a role in minimizing the severity of COVID-19.", "Our study has several limitations. As a limitation of most medical data studies, there is bias caused by confounding factors that may affect our results. When selecting factors for PS matching, information from the CDC and meta-analysis studies were used to select objective data, but these data may change as research on COVID-19 progresses. Further, we defined diseases based on the ICD codes provided in the insurance claims data. There may have been additional unmeasured confounders influencing our results, including genetic polymorphisms, smoking, body mass index, and exposure to the virus. In this study, the infection rate of COVID-19 may have been influenced by the degree of exposure to COVID-19, which may be an important factor in addition to the comorbidity factors. However, the influence of COVID-19 itself could be confirmed because the bias was less than that of previous studies. One race in South Korea comprises more than 95% of the population; hence, there was minimal racial bias compared with previous studies. Because the government funds the treatment for COVID-19 in South Korea and because the medical facilities for COVID-19 treatment are ubiquitous, there was minimal economic bias. The PS matching was performed for sex, age, CCI, comorbidity, and medication, including statins (standardized mean difference <0.1). Hence, selection bias was minimized. Therefore, more accurate information regarding the incidence of COVID-19 and its severity according to comorbidities was provided.\nThis study was based on data from patients who experienced COVID-19 during the early outbreak period; therefore, that strain may differ from the current strain of COVID-19. However, an accurate analysis of recent COVID-19 strains, including Omicron, is difficult because the effects of acquired or natural immunity and vaccination are mixed. Data at the time of its early onset can provide fundamental information, including regarding mutations that may occur in the future.", "Although the severity of COVID-19 has decreased, its hospitalization rate has not decreased significantly, and its burden on medical facilities continues; therefore, an analysis of comorbidities is still important. Therefore, many studies of comorbidities that affect COVID-19 have been published; however, some have reported conflicting results. This may be because various factors such as medication and comorbidities, in addition to demographic factors such as age and sex, affect the infection rate and severity of COVID-19. It is necessary to analyze as many factors as possible to obtain more accurate data regarding COVID-19. Based on the results of previous studies, this study tried to derive objective results by considering various factors affecting COVID-19. In conclusion, certain comorbidities known as risk factors in previous studies increase the infection rate and severity of COVID-19. However, hyperlipidemia decreases the infection rate and severity. These results can be utilized to effectively manage COVID-19." ]
[ "introduction", "methods", null, null, null, null, null, null, "results", null, null, "discussion", null, null, null ]
[ "COVID-19", "comorbidity", "infection rate", "severity of illness index", "hyperlipidemia" ]
New Surveillance Metrics for Alerting Community-Acquired Outbreaks of Emerging SARS-CoV-2 Variants Using Imported Case Data: Bayesian Markov Chain Monte Carlo Approach.
36265134
Global transmission from imported cases to domestic cluster infections is often the origin of local community-acquired outbreaks when facing emerging SARS-CoV-2 variants.
BACKGROUND
We used Taiwanese COVID-19 weekly data on imported cases, domestic cluster infections, and community-acquired outbreaks. The study period included the D614G strain in February 2020, the Alpha and Delta variants of concern (VOCs) in 2021, and the Omicron BA.1 and BA.2 VOCs in April 2022. The number of cases arising from domestic cluster infection caused by imported cases (Dci/Imc) per week was used as the SARS-CoV-2 strain-dependent surveillance metric for alerting local community-acquired outbreaks. Its upper 95% credible interval was used as the alert threshold for guiding the rapid preparedness of containment measures, including nonpharmaceutical interventions (NPIs), testing, and vaccination. The 2 metrics were estimated by using the Bayesian Monte Carlo Markov Chain method underpinning the directed acyclic graphic diagram constructed by the extra-Poisson (random-effect) regression model. The proposed model was also used to assess the most likely week lag of imported cases prior to the current week of domestic cluster infections.
METHODS
A 1-week lag of imported cases prior to the current week of domestic cluster infections was considered optimal. Both metrics of Dci/Imc and the alert threshold varied with SARS-CoV-2 variants and available containment measures. The estimates were 9.54% and 12.59%, respectively, for D614G and increased to 14.14% and 25.10%, respectively, for the Alpha VOC when only NPIs and testing were available. The corresponding figures were 10.01% and 13.32% for the Delta VOC, but reduced to 4.29% and 5.19% for the Omicron VOC when NPIs, testing, and vaccination were available. The rapid preparedness of containment measures guided by the estimated metrics accounted for the lack of community-acquired outbreaks during the D614G period, the early Alpha VOC period, the Delta VOC period, and the Omicron VOC period between BA.1 and BA.2. In contrast, community-acquired outbreaks of the Alpha VOC in mid-May 2021, Omicron BA.1 VOC in January 2022, and Omicron BA.2 VOC from April 2022 onwards, were indicative of the failure to prepare containment measures guided by the alert threshold.
RESULTS
We developed new surveillance metrics for estimating the risk of domestic cluster infections with increasing imported cases and its alert threshold for community-acquired infections varying with emerging SARS-CoV-2 strains and the availability of containment measures. The use of new surveillance metrics is important in the rapid preparedness of containment measures for averting large-scale community-acquired outbreaks arising from emerging imported SARS-CoV-2 variants.
CONCLUSIONS
[ "Humans", "SARS-CoV-2", "Markov Chains", "Bayes Theorem", "Benchmarking", "COVID-19", "Disease Outbreaks" ]
9746786
Introduction
During the COVID-19 pandemic lasting for over 2.5 years, countries around the world have experienced cyclical COVID-19 changes alternating between lifting and operating nonpharmaceutical interventions (NPIs) and between the protective and waning effects of vaccines when facing the incessant epidemics of the COVID-19 pandemic [1-4]. The cyclical resurgence of COVID-19 at the country, continental, and global levels is mainly caused by emerging SARS-CoV-2 variants, particularly variants of concern (VOCs). In response to the resurgence of community-acquired outbreaks, 2 containment measures have become important, including the timely adjustment of NPIs (strengthened border control strategies and restricted social activities) combined with testing and the launch of mass primary and booster vaccinations [5-7]. It should be noted that the typical pattern of transmission from an imported case to domestic cluster infection is often the root of local community-acquired outbreaks caused by emerging SARS-CoV-2 variants from any region or country across the globe [2,7-9]. Such an importation-cluster transmission mode has been clearly demonstrated by the resurgence of global epidemic waves following the emergence of dominant strains of Alpha, Beta, Gamma, Delta, and Omicron VOCs. To avert local community-acquired outbreaks of emerging SARS-CoV-2 variants, rapid preparedness of containment measures and effective contact tracing are mandatory when domestic cluster infections are identified after the introduction of emerging imported cases. In addition, the risk of domestic cluster infection on the introduction of imported cases varies with each emerging SARS-CoV-2 strain owing to the evolutionary characteristics of invading VOCs, including an increase in transmissibility and a higher likelihood of escaping immune response after vaccination [4,7-12]. It is therefore important to have new surveillance metrics for monitoring the odds of having domestic cluster infection transmitted from few imported cases and setting up the alert threshold for forestalling community-acquired outbreaks, as traditional surveillance metrics, like effective reproductive number (Rt), are tailored for assessing the spread and control of community-acquired outbreaks at the population level, which may only involve a single country and a specific SARS-CoV-2 strain in a short period rather than the country of the imported case across the world and the full spectrum of SARS-CoV-2 strains with a long period [13-16]. Such a traditional epidemic surveillance model (eg, the SEIR [Susceptible-​Exposed-Infected-Recovery] model) is not only limited to model the relationship of sparse cases of domestic cluster infection and small samples of imported cases from each original country, but also inflexible to make allowance for the heterogeneity of the imported-domestic transmission mode across countries and SARS-CoV-2 strains across time, as well as the variation across local regions in question. To consider these issues of heterogeneity, it is therefore necessary to develop new surveillance models and their corresponding metrics with a new statistical approach, such as a sampling method of machine learning, particularly the Bayesian Markov Chain Monte Carlo (MCMC) method, in conjunction with a sparse event history regression model, such as the extra-Poisson (random-effect) regression model with relevant parameters and random variables parameterized under the directed acyclic graphic (DAG) diagram. Developing these new surveillance metrics for quantifying the effect size of the transmission from importation to domestic cluster infection would not only be helpful for alerting emerging community-acquired outbreaks, but also aid health professionals having rapid preparedness of SARS-CoV-2 strain–dependent containment measures, including effective and efficient contact tracing. Using a series of chronological epidemic data on COVID-19 divided into 2 phases (non-VOC phase [wild type and D614G] and VOC phase) in Taiwan, this study aimed to develop new surveillance metrices across the periods of various SARS-CoV-2 strains for alerting emerging community-acquired outbreaks by monitoring the risk of small domestic cluster infections originating from the transmission of few imported cases of emerging variants in order to forestall community-acquired outbreaks when facing emerging SARS-CoV-2 variants. The Bayesian MCMC sampling method was therefore used to estimate and predict the new surveillance metrics underpinning the DAG diagram of the Poisson or negative binomial random-effect regression model.
Methods
[SUBTITLE] Data Sources [SUBSECTION] Publicly available information on COVID-19, including the daily number of cases, recovered patients, and deaths from January 1, 2020, to April 2, 2022, in Taiwan, was extracted from the report of the Central Epidemic Command Centre and the Taiwan National Infectious Disease Statistics System maintained by the Taiwan Centre for Disease Control [17]. During the period between January 11 and June 20, 2020, tabular data with epidemiologic information on COVID-19 mentioned above by county and origin of cases (domestic versus imported) were obtained. After October 2020, only aggregated numbers of imported and domestic COVID-19 cases without detailed information at the county and city levels were provided. The population sizes of 23 counties and cities in Taiwan were extracted from the official website of the Department of Household Registration [18]. Publicly available information on COVID-19, including the daily number of cases, recovered patients, and deaths from January 1, 2020, to April 2, 2022, in Taiwan, was extracted from the report of the Central Epidemic Command Centre and the Taiwan National Infectious Disease Statistics System maintained by the Taiwan Centre for Disease Control [17]. During the period between January 11 and June 20, 2020, tabular data with epidemiologic information on COVID-19 mentioned above by county and origin of cases (domestic versus imported) were obtained. After October 2020, only aggregated numbers of imported and domestic COVID-19 cases without detailed information at the county and city levels were provided. The population sizes of 23 counties and cities in Taiwan were extracted from the official website of the Department of Household Registration [18]. [SUBTITLE] Containment Measures for the Non-VOC Phase in Taiwan [SUBSECTION] The containment measures for the non-VOC phase in Taiwan centered on 2 strategies, namely border control with quarantine and isolation, and NPIs without various strategies. Multimedia Appendix 1 shows the timelines of the evolution of border control measures for this non-VOC phase. The number of imported and domestic cases of COVID-19 by the date of onset on a weekly basis is presented in Figure 1. COVID-19 epidemics in Taiwan for periods without outbreaks by the origin of cases (imported vs domestic). The COVID-19 cases in Taiwan in 2020 mainly included imported cases (Figure 1). The risk of an outbreak following the transmission of COVID-19 to the community from these imported cases was largely reduced by the very strict border control strategies with quarantine and isolation in conjunction with NPIs, such as wearing masks and social distancing [19]. Multimedia Appendix 2 provides details on the criteria and guidelines for the implementation of 4 COVID-19 alert levels to target outbreaks in Taiwan. The containment measures for the non-VOC phase in Taiwan centered on 2 strategies, namely border control with quarantine and isolation, and NPIs without various strategies. Multimedia Appendix 1 shows the timelines of the evolution of border control measures for this non-VOC phase. The number of imported and domestic cases of COVID-19 by the date of onset on a weekly basis is presented in Figure 1. COVID-19 epidemics in Taiwan for periods without outbreaks by the origin of cases (imported vs domestic). The COVID-19 cases in Taiwan in 2020 mainly included imported cases (Figure 1). The risk of an outbreak following the transmission of COVID-19 to the community from these imported cases was largely reduced by the very strict border control strategies with quarantine and isolation in conjunction with NPIs, such as wearing masks and social distancing [19]. Multimedia Appendix 2 provides details on the criteria and guidelines for the implementation of 4 COVID-19 alert levels to target outbreaks in Taiwan. [SUBTITLE] Containment Measures for the VOC Phase in Taiwan [SUBSECTION] The Alpha VOC became the predominant strain of the global pandemic by the end of 2020. Several cluster infections occurred in hospitals and households since January 2021, but were still under control until mid-May 2021, when a large-scale outbreak of the Alpha VOC occurred. On the top of border control measures with quarantine and isolation implemented since the non-VOC phase in Taiwan, the focus of containment measures for averting community-acquired outbreaks turned to community-based active surveillance with rapid test stations for the hotspots of outbreaks and enhanced NPIs, including strict regulation for wearing masks, restriction of public gathering, setting up of check points for high-risk areas such as public transportation sites and markets, and restriction of nonessential services such as restaurants and pubs. Multimedia Appendix 1 summarizes the timeline of the implementation of a series of containment measures for the VOC phase starting from the enhancement of NPIs from level 1 to level 2 alert until high restriction border control for travelers. The level 3 alert was rapidly extended to a nationwide level 3 alert on May 19, 2021 [20,21]. During the Delta VOC period (from August to December 2021) and Omicron VOC period (December 2021 onward), transmission in the community has been threatened by imported cases. In addition to containment measures, high coverage of vaccination has been an effective prevention strategy during these 2 periods. In response to the rapid spread of the Omicron VOC, inbound passengers have to follow updated regulations with more frequent reverse transcription-polymerase chain reaction (RT-PCR) testing plus rapid testing, and a possible mandatory 14-day quarantine based on the vaccination status. Additionally, inbound passengers have to provide negative COVID-19 RT-PCR test reports within 2 days and have to take a government-funded rapid RT-PCR test on arrival starting January 11, 2022. Owing to waning of the effects of vaccines, booster shots have been allowed for all adults who have received 2 vaccine doses for 12 weeks (84 days), since January 7, 2022. The Alpha VOC became the predominant strain of the global pandemic by the end of 2020. Several cluster infections occurred in hospitals and households since January 2021, but were still under control until mid-May 2021, when a large-scale outbreak of the Alpha VOC occurred. On the top of border control measures with quarantine and isolation implemented since the non-VOC phase in Taiwan, the focus of containment measures for averting community-acquired outbreaks turned to community-based active surveillance with rapid test stations for the hotspots of outbreaks and enhanced NPIs, including strict regulation for wearing masks, restriction of public gathering, setting up of check points for high-risk areas such as public transportation sites and markets, and restriction of nonessential services such as restaurants and pubs. Multimedia Appendix 1 summarizes the timeline of the implementation of a series of containment measures for the VOC phase starting from the enhancement of NPIs from level 1 to level 2 alert until high restriction border control for travelers. The level 3 alert was rapidly extended to a nationwide level 3 alert on May 19, 2021 [20,21]. During the Delta VOC period (from August to December 2021) and Omicron VOC period (December 2021 onward), transmission in the community has been threatened by imported cases. In addition to containment measures, high coverage of vaccination has been an effective prevention strategy during these 2 periods. In response to the rapid spread of the Omicron VOC, inbound passengers have to follow updated regulations with more frequent reverse transcription-polymerase chain reaction (RT-PCR) testing plus rapid testing, and a possible mandatory 14-day quarantine based on the vaccination status. Additionally, inbound passengers have to provide negative COVID-19 RT-PCR test reports within 2 days and have to take a government-funded rapid RT-PCR test on arrival starting January 11, 2022. Owing to waning of the effects of vaccines, booster shots have been allowed for all adults who have received 2 vaccine doses for 12 weeks (84 days), since January 7, 2022. [SUBTITLE] Statistical Analysis [SUBSECTION] [SUBTITLE] New Surveillance Metrics for Quantifying Imported-Domestic Transmission [SUBSECTION] We used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by where offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks. As mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC). Regarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows: where the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k). We used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by where offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks. As mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC). Regarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows: where the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k). [SUBTITLE] Estimation With the Bayesian MCMC Method [SUBSECTION] The Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution, is proportional to the product of the kernel distribution written by In our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα). A block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows: 1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter. 2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β). 3. Compute the acceptance probability 4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3. 5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4. 6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples. Thinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above. We estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak. To validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak. All statistical analyses were performed using SAS 9.4 software (SAS Institute Inc). The Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution, is proportional to the product of the kernel distribution written by In our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα). A block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows: 1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter. 2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β). 3. Compute the acceptance probability 4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3. 5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4. 6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples. Thinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above. We estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak. To validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak. All statistical analyses were performed using SAS 9.4 software (SAS Institute Inc). [SUBTITLE] New Surveillance Metrics for Quantifying Imported-Domestic Transmission [SUBSECTION] We used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by where offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks. As mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC). Regarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows: where the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k). We used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by where offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks. As mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC). Regarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows: where the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k). [SUBTITLE] Estimation With the Bayesian MCMC Method [SUBSECTION] The Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution, is proportional to the product of the kernel distribution written by In our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα). A block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows: 1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter. 2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β). 3. Compute the acceptance probability 4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3. 5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4. 6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples. Thinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above. We estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak. To validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak. All statistical analyses were performed using SAS 9.4 software (SAS Institute Inc). The Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution, is proportional to the product of the kernel distribution written by In our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα). A block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows: 1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter. 2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β). 3. Compute the acceptance probability 4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3. 5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4. 6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples. Thinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above. We estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak. To validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak. All statistical analyses were performed using SAS 9.4 software (SAS Institute Inc).
Results
[SUBTITLE] Evaluation of the Optimal Time Lag Model [SUBSECTION] After the application of the Bayesian MCMC method for the identification of the optimal time (in weeks) lag of imported cases prior to cases arising from domestic cluster infection, the 1-week lag of imported cases yielded a DIC of 255.8, which was smaller than the DICs of the model with concurrent week imported cases (260.3) and the model with a 2-week lag of imported cases (279.5). After the application of the Bayesian MCMC method for the identification of the optimal time (in weeks) lag of imported cases prior to cases arising from domestic cluster infection, the 1-week lag of imported cases yielded a DIC of 255.8, which was smaller than the DICs of the model with concurrent week imported cases (260.3) and the model with a 2-week lag of imported cases (279.5). [SUBTITLE] Surveillance Metrics for the Imported-Domestic Transmission Mode [SUBSECTION] The weekly observed number (red dot) and expected number (green circle) of domestic cases are shown in Figure 2 (wild-type and D614G period, January to September 2020), Figure 3 (Alpha VOC period, October 2020 to May 2021), and Figure 4 (Delta VOC period, mid-August to mid-December 2021; and Omicron VOC period, mid-December 2021 to early-April 2022). Table 1 shows the details of the estimated results of the parameters encoded in the Bayesian extra-Poisson regression model with a 1-week lag of imported cases regarding the 3 periods without outbreaks in Taiwan, namely the wild-type and D614G period, early Alpha VOC period, and Delta VOC period. The upper bound of the 95% CrI of expected cases (dotted line, Figures 2-4) has been plotted to provide the alert threshold of domestic cluster infection in the community caused by transmission from imported cases 1 week before. This 1-week prior alert on the risk of elevated Dci/Imc per week guided the vigilance on NPIs for averting further community-acquired outbreaks. Number of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the wild-type/D614G period. Number of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the Alpha variant of concern period. Number of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the Delta (August 8, 2021, to December 9, 2021) and Omicron (December 12, 2021, to April 2, 2022) variant of concern periods. Estimated results for the risk of imported-domestic transmission of COVID-19 for 3 periods in Taiwan. aVOC: variant of concern. [SUBTITLE] Wild-Type and D614G Period [SUBSECTION] During the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals. Figure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020. During the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals. Figure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020. [SUBTITLE] Alpha VOC Period [SUBSECTION] There had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A. Effective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant. There had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A. Effective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant. [SUBTITLE] Delta and Omicron VOC Period [SUBSECTION] After controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines. The right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5). There was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022. After controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines. The right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5). There was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022. The weekly observed number (red dot) and expected number (green circle) of domestic cases are shown in Figure 2 (wild-type and D614G period, January to September 2020), Figure 3 (Alpha VOC period, October 2020 to May 2021), and Figure 4 (Delta VOC period, mid-August to mid-December 2021; and Omicron VOC period, mid-December 2021 to early-April 2022). Table 1 shows the details of the estimated results of the parameters encoded in the Bayesian extra-Poisson regression model with a 1-week lag of imported cases regarding the 3 periods without outbreaks in Taiwan, namely the wild-type and D614G period, early Alpha VOC period, and Delta VOC period. The upper bound of the 95% CrI of expected cases (dotted line, Figures 2-4) has been plotted to provide the alert threshold of domestic cluster infection in the community caused by transmission from imported cases 1 week before. This 1-week prior alert on the risk of elevated Dci/Imc per week guided the vigilance on NPIs for averting further community-acquired outbreaks. Number of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the wild-type/D614G period. Number of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the Alpha variant of concern period. Number of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the Delta (August 8, 2021, to December 9, 2021) and Omicron (December 12, 2021, to April 2, 2022) variant of concern periods. Estimated results for the risk of imported-domestic transmission of COVID-19 for 3 periods in Taiwan. aVOC: variant of concern. [SUBTITLE] Wild-Type and D614G Period [SUBSECTION] During the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals. Figure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020. During the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals. Figure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020. [SUBTITLE] Alpha VOC Period [SUBSECTION] There had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A. Effective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant. There had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A. Effective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant. [SUBTITLE] Delta and Omicron VOC Period [SUBSECTION] After controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines. The right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5). There was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022. After controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines. The right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5). There was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022. [SUBTITLE] External Validation of the Surveillance Metrics for Domestic Cluster Infections Using Imported Cases in New Zealand [SUBSECTION] To validate the proposed model and extend its application to different periods of SARS-CoV-2 variants, the proposed extra-Poisson regression model was applied to data on the New Zealand COVID-19 outbreak in 2020. Multimedia Appendix 5 shows the estimated results obtained. Notably, in New Zealand, the risk of Dci/Imc per week increased to 9.38% (95% CrI 8.88%-9.86%), which was close to the estimated results based on Taiwan data (9.54%, 95% CrI 6.44%-12.59%) in the same period. Details regarding the spatial temporal distribution of COVID-19 outbreaks by types of cases in New Zealand are provided in Multimedia Appendix 6. Multimedia Appendix 7 shows the predicted number of domestic cases by using the parameters trained from the empirical data of New Zealand (Multimedia Appendix 5). Similar to the application in Taiwan, the risk of an outbreak associated with imported cases could be assessed by comparing the observed cases (red dot in Multimedia Appendix 7) with the alert threshold (dotted line in Multimedia Appendix 7). The detailed interpretation of the results of this external validation is elaborated in Multimedia Appendix 8. To validate the proposed model and extend its application to different periods of SARS-CoV-2 variants, the proposed extra-Poisson regression model was applied to data on the New Zealand COVID-19 outbreak in 2020. Multimedia Appendix 5 shows the estimated results obtained. Notably, in New Zealand, the risk of Dci/Imc per week increased to 9.38% (95% CrI 8.88%-9.86%), which was close to the estimated results based on Taiwan data (9.54%, 95% CrI 6.44%-12.59%) in the same period. Details regarding the spatial temporal distribution of COVID-19 outbreaks by types of cases in New Zealand are provided in Multimedia Appendix 6. Multimedia Appendix 7 shows the predicted number of domestic cases by using the parameters trained from the empirical data of New Zealand (Multimedia Appendix 5). Similar to the application in Taiwan, the risk of an outbreak associated with imported cases could be assessed by comparing the observed cases (red dot in Multimedia Appendix 7) with the alert threshold (dotted line in Multimedia Appendix 7). The detailed interpretation of the results of this external validation is elaborated in Multimedia Appendix 8.
null
null
[ "Data Sources", "Containment Measures for the Non-VOC Phase in Taiwan", "Containment Measures for the VOC Phase in Taiwan", "Statistical Analysis", "New Surveillance Metrics for Quantifying Imported-Domestic Transmission", "Estimation With the Bayesian MCMC Method", "Evaluation of the Optimal Time Lag Model", "Surveillance Metrics for the Imported-Domestic Transmission Mode", "Wild-Type and D614G Period", "Alpha VOC Period", "Delta and Omicron VOC Period", "External Validation of the Surveillance Metrics for Domestic Cluster Infections Using Imported Cases in New Zealand" ]
[ "Publicly available information on COVID-19, including the daily number of cases, recovered patients, and deaths from January 1, 2020, to April 2, 2022, in Taiwan, was extracted from the report of the Central Epidemic Command Centre and the Taiwan National Infectious Disease Statistics System maintained by the Taiwan Centre for Disease Control [17]. During the period between January 11 and June 20, 2020, tabular data with epidemiologic information on COVID-19 mentioned above by county and origin of cases (domestic versus imported) were obtained. After October 2020, only aggregated numbers of imported and domestic COVID-19 cases without detailed information at the county and city levels were provided. The population sizes of 23 counties and cities in Taiwan were extracted from the official website of the Department of Household Registration [18].", "The containment measures for the non-VOC phase in Taiwan centered on 2 strategies, namely border control with quarantine and isolation, and NPIs without various strategies. Multimedia Appendix 1 shows the timelines of the evolution of border control measures for this non-VOC phase. The number of imported and domestic cases of COVID-19 by the date of onset on a weekly basis is presented in Figure 1.\nCOVID-19 epidemics in Taiwan for periods without outbreaks by the origin of cases (imported vs domestic).\nThe COVID-19 cases in Taiwan in 2020 mainly included imported cases (Figure 1). The risk of an outbreak following the transmission of COVID-19 to the community from these imported cases was largely reduced by the very strict border control strategies with quarantine and isolation in conjunction with NPIs, such as wearing masks and social distancing [19]. Multimedia Appendix 2 provides details on the criteria and guidelines for the implementation of 4 COVID-19 alert levels to target outbreaks in Taiwan.", "The Alpha VOC became the predominant strain of the global pandemic by the end of 2020. Several cluster infections occurred in hospitals and households since January 2021, but were still under control until mid-May 2021, when a large-scale outbreak of the Alpha VOC occurred. On the top of border control measures with quarantine and isolation implemented since the non-VOC phase in Taiwan, the focus of containment measures for averting community-acquired outbreaks turned to community-based active surveillance with rapid test stations for the hotspots of outbreaks and enhanced NPIs, including strict regulation for wearing masks, restriction of public gathering, setting up of check points for high-risk areas such as public transportation sites and markets, and restriction of nonessential services such as restaurants and pubs. Multimedia Appendix 1 summarizes the timeline of the implementation of a series of containment measures for the VOC phase starting from the enhancement of NPIs from level 1 to level 2 alert until high restriction border control for travelers. The level 3 alert was rapidly extended to a nationwide level 3 alert on May 19, 2021 [20,21]. During the Delta VOC period (from August to December 2021) and Omicron VOC period (December 2021 onward), transmission in the community has been threatened by imported cases. In addition to containment measures, high coverage of vaccination has been an effective prevention strategy during these 2 periods. In response to the rapid spread of the Omicron VOC, inbound passengers have to follow updated regulations with more frequent reverse transcription-polymerase chain reaction (RT-PCR) testing plus rapid testing, and a possible mandatory 14-day quarantine based on the vaccination status. Additionally, inbound passengers have to provide negative COVID-19 RT-PCR test reports within 2 days and have to take a government-funded rapid RT-PCR test on arrival starting January 11, 2022. Owing to waning of the effects of vaccines, booster shots have been allowed for all adults who have received 2 vaccine doses for 12 weeks (84 days), since January 7, 2022.", "[SUBTITLE] New Surveillance Metrics for Quantifying Imported-Domestic Transmission [SUBSECTION] We used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by\nwhere offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks.\nAs mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC).\nRegarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows:\nwhere the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k).\nWe used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by\nwhere offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks.\nAs mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC).\nRegarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows:\nwhere the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k).\n[SUBTITLE] Estimation With the Bayesian MCMC Method [SUBSECTION] The Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution,\nis proportional to the product of the kernel distribution written by\nIn our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα).\nA block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows:\n1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter.\n2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β).\n3. Compute the acceptance probability\n4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3.\n5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4.\n6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples.\nThinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above.\nWe estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak.\nTo validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak.\nAll statistical analyses were performed using SAS 9.4 software (SAS Institute Inc).\nThe Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution,\nis proportional to the product of the kernel distribution written by\nIn our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα).\nA block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows:\n1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter.\n2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β).\n3. Compute the acceptance probability\n4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3.\n5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4.\n6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples.\nThinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above.\nWe estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak.\nTo validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak.\nAll statistical analyses were performed using SAS 9.4 software (SAS Institute Inc).", "We used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by\nwhere offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks.\nAs mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC).\nRegarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows:\nwhere the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k).", "The Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution,\nis proportional to the product of the kernel distribution written by\nIn our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα).\nA block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows:\n1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter.\n2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β).\n3. Compute the acceptance probability\n4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3.\n5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4.\n6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples.\nThinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above.\nWe estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak.\nTo validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak.\nAll statistical analyses were performed using SAS 9.4 software (SAS Institute Inc).", "After the application of the Bayesian MCMC method for the identification of the optimal time (in weeks) lag of imported cases prior to cases arising from domestic cluster infection, the 1-week lag of imported cases yielded a DIC of 255.8, which was smaller than the DICs of the model with concurrent week imported cases (260.3) and the model with a 2-week lag of imported cases (279.5).", "The weekly observed number (red dot) and expected number (green circle) of domestic cases are shown in Figure 2 (wild-type and D614G period, January to September 2020), Figure 3 (Alpha VOC period, October 2020 to May 2021), and Figure 4 (Delta VOC period, mid-August to mid-December 2021; and Omicron VOC period, mid-December 2021 to early-April 2022). Table 1 shows the details of the estimated results of the parameters encoded in the Bayesian extra-Poisson regression model with a 1-week lag of imported cases regarding the 3 periods without outbreaks in Taiwan, namely the wild-type and D614G period, early Alpha VOC period, and Delta VOC period.\nThe upper bound of the 95% CrI of expected cases (dotted line, Figures 2-4) has been plotted to provide the alert threshold of domestic cluster infection in the community caused by transmission from imported cases 1 week before. This 1-week prior alert on the risk of elevated Dci/Imc per week guided the vigilance on NPIs for averting further community-acquired outbreaks.\nNumber of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the wild-type/D614G period.\nNumber of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the Alpha variant of concern period.\nNumber of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the Delta (August 8, 2021, to December 9, 2021) and Omicron (December 12, 2021, to April 2, 2022) variant of concern periods.\nEstimated results for the risk of imported-domestic transmission of COVID-19 for 3 periods in Taiwan.\naVOC: variant of concern.\n[SUBTITLE] Wild-Type and D614G Period [SUBSECTION] During the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals.\nFigure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020.\nDuring the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals.\nFigure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020.\n[SUBTITLE] Alpha VOC Period [SUBSECTION] There had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A.\nEffective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant.\nThere had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A.\nEffective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant.\n[SUBTITLE] Delta and Omicron VOC Period [SUBSECTION] After controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines.\nThe right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5).\nThere was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022.\nAfter controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines.\nThe right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5).\nThere was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022.", "During the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals.\nFigure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020.", "There had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A.\nEffective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant.", "After controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines.\nThe right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5).\nThere was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022.", "To validate the proposed model and extend its application to different periods of SARS-CoV-2 variants, the proposed extra-Poisson regression model was applied to data on the New Zealand COVID-19 outbreak in 2020. Multimedia Appendix 5 shows the estimated results obtained. Notably, in New Zealand, the risk of Dci/Imc per week increased to 9.38% (95% CrI 8.88%-9.86%), which was close to the estimated results based on Taiwan data (9.54%, 95% CrI 6.44%-12.59%) in the same period. Details regarding the spatial temporal distribution of COVID-19 outbreaks by types of cases in New Zealand are provided in Multimedia Appendix 6. Multimedia Appendix 7 shows the predicted number of domestic cases by using the parameters trained from the empirical data of New Zealand (Multimedia Appendix 5). Similar to the application in Taiwan, the risk of an outbreak associated with imported cases could be assessed by comparing the observed cases (red dot in Multimedia Appendix 7) with the alert threshold (dotted line in Multimedia Appendix 7). The detailed interpretation of the results of this external validation is elaborated in Multimedia Appendix 8." ]
[ null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Introduction", "Methods", "Data Sources", "Containment Measures for the Non-VOC Phase in Taiwan", "Containment Measures for the VOC Phase in Taiwan", "Statistical Analysis", "New Surveillance Metrics for Quantifying Imported-Domestic Transmission", "Estimation With the Bayesian MCMC Method", "Results", "Evaluation of the Optimal Time Lag Model", "Surveillance Metrics for the Imported-Domestic Transmission Mode", "Wild-Type and D614G Period", "Alpha VOC Period", "Delta and Omicron VOC Period", "External Validation of the Surveillance Metrics for Domestic Cluster Infections Using Imported Cases in New Zealand", "Discussion" ]
[ "During the COVID-19 pandemic lasting for over 2.5 years, countries around the world have experienced cyclical COVID-19 changes alternating between lifting and operating nonpharmaceutical interventions (NPIs) and between the protective and waning effects of vaccines when facing the incessant epidemics of the COVID-19 pandemic [1-4]. The cyclical resurgence of COVID-19 at the country, continental, and global levels is mainly caused by emerging SARS-CoV-2 variants, particularly variants of concern (VOCs). In response to the resurgence of community-acquired outbreaks, 2 containment measures have become important, including the timely adjustment of NPIs (strengthened border control strategies and restricted social activities) combined with testing and the launch of mass primary and booster vaccinations [5-7].\nIt should be noted that the typical pattern of transmission from an imported case to domestic cluster infection is often the root of local community-acquired outbreaks caused by emerging SARS-CoV-2 variants from any region or country across the globe [2,7-9]. Such an importation-cluster transmission mode has been clearly demonstrated by the resurgence of global epidemic waves following the emergence of dominant strains of Alpha, Beta, Gamma, Delta, and Omicron VOCs. To avert local community-acquired outbreaks of emerging SARS-CoV-2 variants, rapid preparedness of containment measures and effective contact tracing are mandatory when domestic cluster infections are identified after the introduction of emerging imported cases. In addition, the risk of domestic cluster infection on the introduction of imported cases varies with each emerging SARS-CoV-2 strain owing to the evolutionary characteristics of invading VOCs, including an increase in transmissibility and a higher likelihood of escaping immune response after vaccination [4,7-12].\nIt is therefore important to have new surveillance metrics for monitoring the odds of having domestic cluster infection transmitted from few imported cases and setting up the alert threshold for forestalling community-acquired outbreaks, as traditional surveillance metrics, like effective reproductive number (Rt), are tailored for assessing the spread and control of community-acquired outbreaks at the population level, which may only involve a single country and a specific SARS-CoV-2 strain in a short period rather than the country of the imported case across the world and the full spectrum of SARS-CoV-2 strains with a long period [13-16]. Such a traditional epidemic surveillance model (eg, the SEIR [Susceptible-​Exposed-Infected-Recovery] model) is not only limited to model the relationship of sparse cases of domestic cluster infection and small samples of imported cases from each original country, but also inflexible to make allowance for the heterogeneity of the imported-domestic transmission mode across countries and SARS-CoV-2 strains across time, as well as the variation across local regions in question. To consider these issues of heterogeneity, it is therefore necessary to develop new surveillance models and their corresponding metrics with a new statistical approach, such as a sampling method of machine learning, particularly the Bayesian Markov Chain Monte Carlo (MCMC) method, in conjunction with a sparse event history regression model, such as the extra-Poisson (random-effect) regression model with relevant parameters and random variables parameterized under the directed acyclic graphic (DAG) diagram.\nDeveloping these new surveillance metrics for quantifying the effect size of the transmission from importation to domestic cluster infection would not only be helpful for alerting emerging community-acquired outbreaks, but also aid health professionals having rapid preparedness of SARS-CoV-2 strain–dependent containment measures, including effective and efficient contact tracing. Using a series of chronological epidemic data on COVID-19 divided into 2 phases (non-VOC phase [wild type and D614G] and VOC phase) in Taiwan, this study aimed to develop new surveillance metrices across the periods of various SARS-CoV-2 strains for alerting emerging community-acquired outbreaks by monitoring the risk of small domestic cluster infections originating from the transmission of few imported cases of emerging variants in order to forestall community-acquired outbreaks when facing emerging SARS-CoV-2 variants. The Bayesian MCMC sampling method was therefore used to estimate and predict the new surveillance metrics underpinning the DAG diagram of the Poisson or negative binomial random-effect regression model.", "[SUBTITLE] Data Sources [SUBSECTION] Publicly available information on COVID-19, including the daily number of cases, recovered patients, and deaths from January 1, 2020, to April 2, 2022, in Taiwan, was extracted from the report of the Central Epidemic Command Centre and the Taiwan National Infectious Disease Statistics System maintained by the Taiwan Centre for Disease Control [17]. During the period between January 11 and June 20, 2020, tabular data with epidemiologic information on COVID-19 mentioned above by county and origin of cases (domestic versus imported) were obtained. After October 2020, only aggregated numbers of imported and domestic COVID-19 cases without detailed information at the county and city levels were provided. The population sizes of 23 counties and cities in Taiwan were extracted from the official website of the Department of Household Registration [18].\nPublicly available information on COVID-19, including the daily number of cases, recovered patients, and deaths from January 1, 2020, to April 2, 2022, in Taiwan, was extracted from the report of the Central Epidemic Command Centre and the Taiwan National Infectious Disease Statistics System maintained by the Taiwan Centre for Disease Control [17]. During the period between January 11 and June 20, 2020, tabular data with epidemiologic information on COVID-19 mentioned above by county and origin of cases (domestic versus imported) were obtained. After October 2020, only aggregated numbers of imported and domestic COVID-19 cases without detailed information at the county and city levels were provided. The population sizes of 23 counties and cities in Taiwan were extracted from the official website of the Department of Household Registration [18].\n[SUBTITLE] Containment Measures for the Non-VOC Phase in Taiwan [SUBSECTION] The containment measures for the non-VOC phase in Taiwan centered on 2 strategies, namely border control with quarantine and isolation, and NPIs without various strategies. Multimedia Appendix 1 shows the timelines of the evolution of border control measures for this non-VOC phase. The number of imported and domestic cases of COVID-19 by the date of onset on a weekly basis is presented in Figure 1.\nCOVID-19 epidemics in Taiwan for periods without outbreaks by the origin of cases (imported vs domestic).\nThe COVID-19 cases in Taiwan in 2020 mainly included imported cases (Figure 1). The risk of an outbreak following the transmission of COVID-19 to the community from these imported cases was largely reduced by the very strict border control strategies with quarantine and isolation in conjunction with NPIs, such as wearing masks and social distancing [19]. Multimedia Appendix 2 provides details on the criteria and guidelines for the implementation of 4 COVID-19 alert levels to target outbreaks in Taiwan.\nThe containment measures for the non-VOC phase in Taiwan centered on 2 strategies, namely border control with quarantine and isolation, and NPIs without various strategies. Multimedia Appendix 1 shows the timelines of the evolution of border control measures for this non-VOC phase. The number of imported and domestic cases of COVID-19 by the date of onset on a weekly basis is presented in Figure 1.\nCOVID-19 epidemics in Taiwan for periods without outbreaks by the origin of cases (imported vs domestic).\nThe COVID-19 cases in Taiwan in 2020 mainly included imported cases (Figure 1). The risk of an outbreak following the transmission of COVID-19 to the community from these imported cases was largely reduced by the very strict border control strategies with quarantine and isolation in conjunction with NPIs, such as wearing masks and social distancing [19]. Multimedia Appendix 2 provides details on the criteria and guidelines for the implementation of 4 COVID-19 alert levels to target outbreaks in Taiwan.\n[SUBTITLE] Containment Measures for the VOC Phase in Taiwan [SUBSECTION] The Alpha VOC became the predominant strain of the global pandemic by the end of 2020. Several cluster infections occurred in hospitals and households since January 2021, but were still under control until mid-May 2021, when a large-scale outbreak of the Alpha VOC occurred. On the top of border control measures with quarantine and isolation implemented since the non-VOC phase in Taiwan, the focus of containment measures for averting community-acquired outbreaks turned to community-based active surveillance with rapid test stations for the hotspots of outbreaks and enhanced NPIs, including strict regulation for wearing masks, restriction of public gathering, setting up of check points for high-risk areas such as public transportation sites and markets, and restriction of nonessential services such as restaurants and pubs. Multimedia Appendix 1 summarizes the timeline of the implementation of a series of containment measures for the VOC phase starting from the enhancement of NPIs from level 1 to level 2 alert until high restriction border control for travelers. The level 3 alert was rapidly extended to a nationwide level 3 alert on May 19, 2021 [20,21]. During the Delta VOC period (from August to December 2021) and Omicron VOC period (December 2021 onward), transmission in the community has been threatened by imported cases. In addition to containment measures, high coverage of vaccination has been an effective prevention strategy during these 2 periods. In response to the rapid spread of the Omicron VOC, inbound passengers have to follow updated regulations with more frequent reverse transcription-polymerase chain reaction (RT-PCR) testing plus rapid testing, and a possible mandatory 14-day quarantine based on the vaccination status. Additionally, inbound passengers have to provide negative COVID-19 RT-PCR test reports within 2 days and have to take a government-funded rapid RT-PCR test on arrival starting January 11, 2022. Owing to waning of the effects of vaccines, booster shots have been allowed for all adults who have received 2 vaccine doses for 12 weeks (84 days), since January 7, 2022.\nThe Alpha VOC became the predominant strain of the global pandemic by the end of 2020. Several cluster infections occurred in hospitals and households since January 2021, but were still under control until mid-May 2021, when a large-scale outbreak of the Alpha VOC occurred. On the top of border control measures with quarantine and isolation implemented since the non-VOC phase in Taiwan, the focus of containment measures for averting community-acquired outbreaks turned to community-based active surveillance with rapid test stations for the hotspots of outbreaks and enhanced NPIs, including strict regulation for wearing masks, restriction of public gathering, setting up of check points for high-risk areas such as public transportation sites and markets, and restriction of nonessential services such as restaurants and pubs. Multimedia Appendix 1 summarizes the timeline of the implementation of a series of containment measures for the VOC phase starting from the enhancement of NPIs from level 1 to level 2 alert until high restriction border control for travelers. The level 3 alert was rapidly extended to a nationwide level 3 alert on May 19, 2021 [20,21]. During the Delta VOC period (from August to December 2021) and Omicron VOC period (December 2021 onward), transmission in the community has been threatened by imported cases. In addition to containment measures, high coverage of vaccination has been an effective prevention strategy during these 2 periods. In response to the rapid spread of the Omicron VOC, inbound passengers have to follow updated regulations with more frequent reverse transcription-polymerase chain reaction (RT-PCR) testing plus rapid testing, and a possible mandatory 14-day quarantine based on the vaccination status. Additionally, inbound passengers have to provide negative COVID-19 RT-PCR test reports within 2 days and have to take a government-funded rapid RT-PCR test on arrival starting January 11, 2022. Owing to waning of the effects of vaccines, booster shots have been allowed for all adults who have received 2 vaccine doses for 12 weeks (84 days), since January 7, 2022.\n[SUBTITLE] Statistical Analysis [SUBSECTION] [SUBTITLE] New Surveillance Metrics for Quantifying Imported-Domestic Transmission [SUBSECTION] We used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by\nwhere offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks.\nAs mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC).\nRegarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows:\nwhere the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k).\nWe used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by\nwhere offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks.\nAs mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC).\nRegarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows:\nwhere the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k).\n[SUBTITLE] Estimation With the Bayesian MCMC Method [SUBSECTION] The Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution,\nis proportional to the product of the kernel distribution written by\nIn our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα).\nA block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows:\n1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter.\n2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β).\n3. Compute the acceptance probability\n4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3.\n5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4.\n6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples.\nThinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above.\nWe estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak.\nTo validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak.\nAll statistical analyses were performed using SAS 9.4 software (SAS Institute Inc).\nThe Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution,\nis proportional to the product of the kernel distribution written by\nIn our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα).\nA block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows:\n1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter.\n2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β).\n3. Compute the acceptance probability\n4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3.\n5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4.\n6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples.\nThinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above.\nWe estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak.\nTo validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak.\nAll statistical analyses were performed using SAS 9.4 software (SAS Institute Inc).\n[SUBTITLE] New Surveillance Metrics for Quantifying Imported-Domestic Transmission [SUBSECTION] We used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by\nwhere offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks.\nAs mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC).\nRegarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows:\nwhere the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k).\nWe used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by\nwhere offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks.\nAs mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC).\nRegarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows:\nwhere the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k).\n[SUBTITLE] Estimation With the Bayesian MCMC Method [SUBSECTION] The Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution,\nis proportional to the product of the kernel distribution written by\nIn our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα).\nA block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows:\n1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter.\n2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β).\n3. Compute the acceptance probability\n4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3.\n5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4.\n6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples.\nThinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above.\nWe estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak.\nTo validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak.\nAll statistical analyses were performed using SAS 9.4 software (SAS Institute Inc).\nThe Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution,\nis proportional to the product of the kernel distribution written by\nIn our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα).\nA block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows:\n1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter.\n2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β).\n3. Compute the acceptance probability\n4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3.\n5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4.\n6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples.\nThinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above.\nWe estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak.\nTo validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak.\nAll statistical analyses were performed using SAS 9.4 software (SAS Institute Inc).", "Publicly available information on COVID-19, including the daily number of cases, recovered patients, and deaths from January 1, 2020, to April 2, 2022, in Taiwan, was extracted from the report of the Central Epidemic Command Centre and the Taiwan National Infectious Disease Statistics System maintained by the Taiwan Centre for Disease Control [17]. During the period between January 11 and June 20, 2020, tabular data with epidemiologic information on COVID-19 mentioned above by county and origin of cases (domestic versus imported) were obtained. After October 2020, only aggregated numbers of imported and domestic COVID-19 cases without detailed information at the county and city levels were provided. The population sizes of 23 counties and cities in Taiwan were extracted from the official website of the Department of Household Registration [18].", "The containment measures for the non-VOC phase in Taiwan centered on 2 strategies, namely border control with quarantine and isolation, and NPIs without various strategies. Multimedia Appendix 1 shows the timelines of the evolution of border control measures for this non-VOC phase. The number of imported and domestic cases of COVID-19 by the date of onset on a weekly basis is presented in Figure 1.\nCOVID-19 epidemics in Taiwan for periods without outbreaks by the origin of cases (imported vs domestic).\nThe COVID-19 cases in Taiwan in 2020 mainly included imported cases (Figure 1). The risk of an outbreak following the transmission of COVID-19 to the community from these imported cases was largely reduced by the very strict border control strategies with quarantine and isolation in conjunction with NPIs, such as wearing masks and social distancing [19]. Multimedia Appendix 2 provides details on the criteria and guidelines for the implementation of 4 COVID-19 alert levels to target outbreaks in Taiwan.", "The Alpha VOC became the predominant strain of the global pandemic by the end of 2020. Several cluster infections occurred in hospitals and households since January 2021, but were still under control until mid-May 2021, when a large-scale outbreak of the Alpha VOC occurred. On the top of border control measures with quarantine and isolation implemented since the non-VOC phase in Taiwan, the focus of containment measures for averting community-acquired outbreaks turned to community-based active surveillance with rapid test stations for the hotspots of outbreaks and enhanced NPIs, including strict regulation for wearing masks, restriction of public gathering, setting up of check points for high-risk areas such as public transportation sites and markets, and restriction of nonessential services such as restaurants and pubs. Multimedia Appendix 1 summarizes the timeline of the implementation of a series of containment measures for the VOC phase starting from the enhancement of NPIs from level 1 to level 2 alert until high restriction border control for travelers. The level 3 alert was rapidly extended to a nationwide level 3 alert on May 19, 2021 [20,21]. During the Delta VOC period (from August to December 2021) and Omicron VOC period (December 2021 onward), transmission in the community has been threatened by imported cases. In addition to containment measures, high coverage of vaccination has been an effective prevention strategy during these 2 periods. In response to the rapid spread of the Omicron VOC, inbound passengers have to follow updated regulations with more frequent reverse transcription-polymerase chain reaction (RT-PCR) testing plus rapid testing, and a possible mandatory 14-day quarantine based on the vaccination status. Additionally, inbound passengers have to provide negative COVID-19 RT-PCR test reports within 2 days and have to take a government-funded rapid RT-PCR test on arrival starting January 11, 2022. Owing to waning of the effects of vaccines, booster shots have been allowed for all adults who have received 2 vaccine doses for 12 weeks (84 days), since January 7, 2022.", "[SUBTITLE] New Surveillance Metrics for Quantifying Imported-Domestic Transmission [SUBSECTION] We used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by\nwhere offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks.\nAs mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC).\nRegarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows:\nwhere the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k).\nWe used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by\nwhere offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks.\nAs mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC).\nRegarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows:\nwhere the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k).\n[SUBTITLE] Estimation With the Bayesian MCMC Method [SUBSECTION] The Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution,\nis proportional to the product of the kernel distribution written by\nIn our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα).\nA block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows:\n1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter.\n2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β).\n3. Compute the acceptance probability\n4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3.\n5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4.\n6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples.\nThinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above.\nWe estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak.\nTo validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak.\nAll statistical analyses were performed using SAS 9.4 software (SAS Institute Inc).\nThe Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution,\nis proportional to the product of the kernel distribution written by\nIn our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα).\nA block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows:\n1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter.\n2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β).\n3. Compute the acceptance probability\n4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3.\n5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4.\n6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples.\nThinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above.\nWe estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak.\nTo validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak.\nAll statistical analyses were performed using SAS 9.4 software (SAS Institute Inc).", "We used an extra-Poisson regression model with a Bayesian DAG approach [22] to calculate the expected weekly domestic cluster infections associated with imported cases of COVID-19, as shown in the right panel of Multimedia Appendix 3. For the jth county or city with the Ytj domestic case at week t, the extra-Poisson regression model can be specified by\nwhere offestj is the population of log scale, and the heterogeneity of imported-domestic transmission across counties and cities in Taiwan is captured by a normal distributed random intercept parameter, αj. While the common intercept parameter, α0, represents the common risk of transmission in Taiwan, the heterogeneity is captured by the variance parameter, σ2α. With this framework, the number of cases arising from domestic cluster infection caused by imported cases per week before (Xj,t-1) can be assessed by using the regression coefficient β, which becomes the first surveillance metric and is denoted as Dci/Imc per week for estimating the effect size of domestic cluster infection. The larger the value of this metric estimated, the larger the domestic cluster infection. The extra variation across cities and counties regarding the transmission of COVID-19 associated with imported cases was captured by a random effect (αj) incorporated into the Poisson regression model, which is also called the random-effect Poisson regression model. The predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 3) can be generated by using the number of imported cases in the current week (X[t]; Multimedia Appendix 3) in conjunction with the posterior distribution of the force of transmission (β), standing for the metric of Dci/Imc per week, and the common intercept (α0) taking into account the county-level heterogeneity of COVID-19 transmission (σ2α). The Poisson model has been widely applied to sparse counts of domestic infection, which occur independently if there is a lack of larger cluster infections, with a high potential of developing into a large-scale community-acquired outbreak. If the observed value of our model is beyond the upper limit of the 95% credible interval (CrI), it means that sparse and independent assumptions based on the Poisson distribution are violated and implies a high potential of yielding a large-scale community-acquired outbreak. Accordingly, the second surveillance metric is to build up the alert threshold of emerging community-acquired outbreaks and to provide guidance for the rapid preparedness of containment measures (including effective and efficient contact tracing) for forestalling community-acquired outbreaks.\nAs mentioned above, data were divided into the non-VOC phase and VOC phase. The former period used for estimating the parameters of the following extra-Poisson regression model was based on imported and domestic cases between January 11 and June 20, 2020, covering the wild-type and D614G period in Taiwan. Because imported cases require an incubation time to generate secondary cases, we tested the lag time of imported cases by 0 weeks (concurrent, Xjt), 1 week (Xj,t-1), and 2 weeks (Xj,t-2), and further selected the optimal lag time interval with the smallest deviance information criterion (DIC).\nRegarding the impact of imported cases on the occurrence of domestic cases for the early Alpha (October 11, 2020, to May 12, 2021), Delta (August 8 to December 9, 2021), and Omicron (December 12, 2021, to April 2, 2022) VOC periods without outbreaks in Taiwan, a Bayesian negative binomial regression model was applied to take into account the heterogeneity across counties and cities associated with the imported-domestic transmission of COVID-19 owing to the lack of detailed information on the cases in counties and cities. Multimedia Appendix 4 shows the DAG model for assessing the force of imported-domestic transmission by using a Bayesian negative binomial regression model. Following the approach applied for the wild-type and D614G period, the 1-week lag model was adopted. For week t, the number of cases Yt resulting from imported cases 1 week prior, Xt-1, can be modeled by using the negative binomial regression model as follows:\nwhere the heterogeneity is captured by the dispersion parameter 1/k. Similar to the extra-Poisson regression model as above, the risk of imported-domestic transmission can thus be assessed by using the regression coefficient β for estimating the effect size of Dci/Imc. Following the extra-Poisson approach, the predicted distribution of the number of expected domestic cases in the next week (μ.pred[t+1]; Multimedia Appendix 4) for the Bayesian negative binomial model can be generated from the current number of imported cases (X[t]; Multimedia Appendix 4) by using the posterior distribution of imported-domestic transmission (β) and the dispersion parameter (1/k).", "The Bayesian MCMC method was used to generate the samples derived from the posterior distributions of parameters for estimating 2 surveillance metrics. With the Markov chain underpinning, a stationary distribution for parameters can be reached in the long run under regular conditions. Independent samples can thus be generated from such a stationary posterior distribution on the basis of which inferences can be made [23]. The DAG models depicted in Multimedia Appendix 3 and Multimedia Appendix 4 were applied to facilitate the decomposition of joint distribution into full conditional density distribution by using the relationship between parent and child nodes [24]. Taking the extra-Poisson regression model as an example, the joint distribution,\nis proportional to the product of the kernel distribution written by\nIn our application, noninformative priors were used to derive the samples from the stationary posterior distribution of parameters, including the risk of imported-domestic transmission (β), the common intercept (α0), and the county-specific random effect (σα).\nA block-wise Metropolic-Hasting sampler was applied to generate samples from the stationary posterior distribution. The sampling algorithm is detailed as follows:\n1. Start with an initial value (β(0), α(0), α0(0), σ2α(0)) selected from the support of each parameter.\n2. Draw the candidate value for the first parameter, say β(1), from a normal proposal distribution, q(β).\n3. Compute the acceptance probability\n4. Draw u from uniform (0,1) and update β(0) with β(1) if u < r(β(1), β(0) | α(0), α0(0), σ2α(0)); otherwise, repeat steps 2 and 3.\n5. Draw the candidate value for the next parameter, α(1), to update the parameter sample with (α(0) | α0(0), σ2α(0), β(1)) by using steps 2 to 4.\n6. Repeat steps 2 to 5 for the rest of the parameters, (α0, σ2α), to derive (β(1), α(1), α0(1), σ2α(1)) to complete an iteration of the update for parameter samples.\nThinning intervals of 10 and 100,000 iterations were used to generate the 10,000 posterior samples after 250,000 burn-in iterations by using the Bayesian MCMC methods mentioned above.\nWe estimated the effect size of Dci/Imc per week for each period corresponding to the type of SARS-CoV-2 variant. We built up the alert threshold by using the upper limit of the 95% CrI of the predicted number of domestic cases (μ.pred[t+1]; Multimedia Appendix 3 and Multimedia Appendix 4) generated by the parameters after updating the data on the non-VOC phase in Taiwan for alerting the possibility of yielding a large-scale community-acquired outbreak through imported-domestic transmission in the subsequent epochs. The possibility of a community-acquired outbreak was deemed low if observed domestic cases were not more than the alert threshold, namely the upper limit of the 95% CrI. Otherwise, an outbreak was likely to occur, and therefore, the rapid preparedness of containment measures, including effective and efficient contact tracing, would be flagged to forestall the ensuing community-acquired outbreak.\nTo validate the proposed surveillance model for the transmission from imported to domestic cases during the non-VOC period in Taiwan, the publicly available COVID-19 data provided by the Ministry of Health in New Zealand were used [25]. The chronological order of the incidence of COVID-19 for the hotspots was compared to validate the epidemic surveillance model for an outbreak.\nAll statistical analyses were performed using SAS 9.4 software (SAS Institute Inc).", "[SUBTITLE] Evaluation of the Optimal Time Lag Model [SUBSECTION] After the application of the Bayesian MCMC method for the identification of the optimal time (in weeks) lag of imported cases prior to cases arising from domestic cluster infection, the 1-week lag of imported cases yielded a DIC of 255.8, which was smaller than the DICs of the model with concurrent week imported cases (260.3) and the model with a 2-week lag of imported cases (279.5).\nAfter the application of the Bayesian MCMC method for the identification of the optimal time (in weeks) lag of imported cases prior to cases arising from domestic cluster infection, the 1-week lag of imported cases yielded a DIC of 255.8, which was smaller than the DICs of the model with concurrent week imported cases (260.3) and the model with a 2-week lag of imported cases (279.5).\n[SUBTITLE] Surveillance Metrics for the Imported-Domestic Transmission Mode [SUBSECTION] The weekly observed number (red dot) and expected number (green circle) of domestic cases are shown in Figure 2 (wild-type and D614G period, January to September 2020), Figure 3 (Alpha VOC period, October 2020 to May 2021), and Figure 4 (Delta VOC period, mid-August to mid-December 2021; and Omicron VOC period, mid-December 2021 to early-April 2022). Table 1 shows the details of the estimated results of the parameters encoded in the Bayesian extra-Poisson regression model with a 1-week lag of imported cases regarding the 3 periods without outbreaks in Taiwan, namely the wild-type and D614G period, early Alpha VOC period, and Delta VOC period.\nThe upper bound of the 95% CrI of expected cases (dotted line, Figures 2-4) has been plotted to provide the alert threshold of domestic cluster infection in the community caused by transmission from imported cases 1 week before. This 1-week prior alert on the risk of elevated Dci/Imc per week guided the vigilance on NPIs for averting further community-acquired outbreaks.\nNumber of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the wild-type/D614G period.\nNumber of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the Alpha variant of concern period.\nNumber of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the Delta (August 8, 2021, to December 9, 2021) and Omicron (December 12, 2021, to April 2, 2022) variant of concern periods.\nEstimated results for the risk of imported-domestic transmission of COVID-19 for 3 periods in Taiwan.\naVOC: variant of concern.\n[SUBTITLE] Wild-Type and D614G Period [SUBSECTION] During the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals.\nFigure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020.\nDuring the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals.\nFigure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020.\n[SUBTITLE] Alpha VOC Period [SUBSECTION] There had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A.\nEffective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant.\nThere had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A.\nEffective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant.\n[SUBTITLE] Delta and Omicron VOC Period [SUBSECTION] After controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines.\nThe right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5).\nThere was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022.\nAfter controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines.\nThe right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5).\nThere was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022.\nThe weekly observed number (red dot) and expected number (green circle) of domestic cases are shown in Figure 2 (wild-type and D614G period, January to September 2020), Figure 3 (Alpha VOC period, October 2020 to May 2021), and Figure 4 (Delta VOC period, mid-August to mid-December 2021; and Omicron VOC period, mid-December 2021 to early-April 2022). Table 1 shows the details of the estimated results of the parameters encoded in the Bayesian extra-Poisson regression model with a 1-week lag of imported cases regarding the 3 periods without outbreaks in Taiwan, namely the wild-type and D614G period, early Alpha VOC period, and Delta VOC period.\nThe upper bound of the 95% CrI of expected cases (dotted line, Figures 2-4) has been plotted to provide the alert threshold of domestic cluster infection in the community caused by transmission from imported cases 1 week before. This 1-week prior alert on the risk of elevated Dci/Imc per week guided the vigilance on NPIs for averting further community-acquired outbreaks.\nNumber of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the wild-type/D614G period.\nNumber of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the Alpha variant of concern period.\nNumber of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the Delta (August 8, 2021, to December 9, 2021) and Omicron (December 12, 2021, to April 2, 2022) variant of concern periods.\nEstimated results for the risk of imported-domestic transmission of COVID-19 for 3 periods in Taiwan.\naVOC: variant of concern.\n[SUBTITLE] Wild-Type and D614G Period [SUBSECTION] During the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals.\nFigure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020.\nDuring the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals.\nFigure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020.\n[SUBTITLE] Alpha VOC Period [SUBSECTION] There had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A.\nEffective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant.\nThere had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A.\nEffective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant.\n[SUBTITLE] Delta and Omicron VOC Period [SUBSECTION] After controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines.\nThe right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5).\nThere was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022.\nAfter controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines.\nThe right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5).\nThere was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022.\n[SUBTITLE] External Validation of the Surveillance Metrics for Domestic Cluster Infections Using Imported Cases in New Zealand [SUBSECTION] To validate the proposed model and extend its application to different periods of SARS-CoV-2 variants, the proposed extra-Poisson regression model was applied to data on the New Zealand COVID-19 outbreak in 2020. Multimedia Appendix 5 shows the estimated results obtained. Notably, in New Zealand, the risk of Dci/Imc per week increased to 9.38% (95% CrI 8.88%-9.86%), which was close to the estimated results based on Taiwan data (9.54%, 95% CrI 6.44%-12.59%) in the same period. Details regarding the spatial temporal distribution of COVID-19 outbreaks by types of cases in New Zealand are provided in Multimedia Appendix 6. Multimedia Appendix 7 shows the predicted number of domestic cases by using the parameters trained from the empirical data of New Zealand (Multimedia Appendix 5). Similar to the application in Taiwan, the risk of an outbreak associated with imported cases could be assessed by comparing the observed cases (red dot in Multimedia Appendix 7) with the alert threshold (dotted line in Multimedia Appendix 7). The detailed interpretation of the results of this external validation is elaborated in Multimedia Appendix 8.\nTo validate the proposed model and extend its application to different periods of SARS-CoV-2 variants, the proposed extra-Poisson regression model was applied to data on the New Zealand COVID-19 outbreak in 2020. Multimedia Appendix 5 shows the estimated results obtained. Notably, in New Zealand, the risk of Dci/Imc per week increased to 9.38% (95% CrI 8.88%-9.86%), which was close to the estimated results based on Taiwan data (9.54%, 95% CrI 6.44%-12.59%) in the same period. Details regarding the spatial temporal distribution of COVID-19 outbreaks by types of cases in New Zealand are provided in Multimedia Appendix 6. Multimedia Appendix 7 shows the predicted number of domestic cases by using the parameters trained from the empirical data of New Zealand (Multimedia Appendix 5). Similar to the application in Taiwan, the risk of an outbreak associated with imported cases could be assessed by comparing the observed cases (red dot in Multimedia Appendix 7) with the alert threshold (dotted line in Multimedia Appendix 7). The detailed interpretation of the results of this external validation is elaborated in Multimedia Appendix 8.", "After the application of the Bayesian MCMC method for the identification of the optimal time (in weeks) lag of imported cases prior to cases arising from domestic cluster infection, the 1-week lag of imported cases yielded a DIC of 255.8, which was smaller than the DICs of the model with concurrent week imported cases (260.3) and the model with a 2-week lag of imported cases (279.5).", "The weekly observed number (red dot) and expected number (green circle) of domestic cases are shown in Figure 2 (wild-type and D614G period, January to September 2020), Figure 3 (Alpha VOC period, October 2020 to May 2021), and Figure 4 (Delta VOC period, mid-August to mid-December 2021; and Omicron VOC period, mid-December 2021 to early-April 2022). Table 1 shows the details of the estimated results of the parameters encoded in the Bayesian extra-Poisson regression model with a 1-week lag of imported cases regarding the 3 periods without outbreaks in Taiwan, namely the wild-type and D614G period, early Alpha VOC period, and Delta VOC period.\nThe upper bound of the 95% CrI of expected cases (dotted line, Figures 2-4) has been plotted to provide the alert threshold of domestic cluster infection in the community caused by transmission from imported cases 1 week before. This 1-week prior alert on the risk of elevated Dci/Imc per week guided the vigilance on NPIs for averting further community-acquired outbreaks.\nNumber of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the wild-type/D614G period.\nNumber of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the Alpha variant of concern period.\nNumber of observed (dotted point) and expected (green circle) domestic cases with the upper limit of the 95% credible interval (CrI) (dotted line) by week in the Delta (August 8, 2021, to December 9, 2021) and Omicron (December 12, 2021, to April 2, 2022) variant of concern periods.\nEstimated results for the risk of imported-domestic transmission of COVID-19 for 3 periods in Taiwan.\naVOC: variant of concern.\n[SUBTITLE] Wild-Type and D614G Period [SUBSECTION] During the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals.\nFigure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020.\nDuring the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals.\nFigure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020.\n[SUBTITLE] Alpha VOC Period [SUBSECTION] There had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A.\nEffective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant.\nThere had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A.\nEffective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant.\n[SUBTITLE] Delta and Omicron VOC Period [SUBSECTION] After controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines.\nThe right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5).\nThere was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022.\nAfter controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines.\nThe right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5).\nThere was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022.", "During the wild-type/D614G period, the estimated Dci/Imc per week was 9.54% (95% CrI 6.44%-12.59%; Table 1). Figure 2 shows that there were 5 weeks (January 26 to February 1, February 2 to February 8, February 16 to February 22, February 23 to February 29, and March 15 to March 21, 2020) in which the observed numbers of domestic cases exceeded the alert thresholds. This period yielded 81% of clustered cases (22 of 27 community-acquired cases) in 5 clusters in Taiwan, including 3 household clusters (with 5, 3, and 6 COVID-19 cases, respectively), 1 medical institute cluster (with 9 COVID-19 cases), and 1 academic institute cluster (with 4 COVID-19 cases). Guidance of the alert thresholds from this early period of the wild-type COVID-19 strain provided a strong rationale for being on alert for the ensuing cluster infections from the preceding 1 week when imported cases were introduced. This accounted for why none of these 5 cluster events led to any large-scale community-acquired outbreaks in Taiwan. There was rapid preparedness of containment measures with strict NPIs together with effective and efficient contact tracing of all possible susceptible individuals.\nFigure 2 shows that this surveillance metric was very useful, particularly when there was a substantial surge in imported cases resulting from the large-scale COVID-19 pandemic worldwide. This could be seen in our cases between March and April 2020, as shown in Figure 1. Again, the surveillance metric was used for alerting about possible cluster infections to forestall further community-acquired outbreaks. Alerted by the threshold (20 domestic cases per week), the observed domestic cases were kept lower than the alert threshold to avoid large-scale outbreaks in April. Since then, there had not been any domestic case until December 2020.", "There had been no outbreak during the early Alpha VOC phase of the COVID-19 pandemic from October to December 2020. The second surge of imported cases occurred from January 2021 onwards (Figure 1). Again, there was 1 week (January 17 to January 23, 2021) in which the observed number of domestic cases was beyond the alert threshold, resulting from hospital-based cluster infections and 3 subsequent household clusters (11 family members) (Figure 3). The source of this cluster infection was later identified as an imported case infected with the Alpha VOC. After being alerted by the proposed surveillance metric and following timely contact tracing and containment measures, including quarantine and isolation for all staff members in the hospital and their close contacts for 14 days, there was no further outbreak until early May 2021, when the number of observed domestic cases reached beyond 20, which was higher than the expected surveillance curve (5 cases) and the corresponding upper bound of the 95% CrI (14 cases). The estimated results on the basis of the empirical data during this Alpha VOC period showed that the Dci/Imc increased to 14.14% (95% CrI 5.41%-25.10%; Table 1). The peak of domestic cases far beyond the threshold value in early May 2021 not only presaged the ensuing outbreaks, but also revealed the loose NPIs at that time without an available vaccine in Taiwan. If effective contact tracing and timely containment measures had been deployed in advance on the basis of the increased risk and the alert threshold in the week between May 2 and May 8, the ensuing community-acquired outbreak involving the Alpha VOC could have been prevented (bottom panel of Figure 1). This outbreak lasted for 2.5 months and subsided in July 2021. The corresponding periods of the estimated results for the effectiveness of NPIs and testing implemented in Taiwan during the Alpha VOC phase are shown in Figure 5A.\nEffective reproductive number (Rt) and effectiveness of nonpharmaceutical interventions (NPIs) and testing in Taiwan. (A) Alpha variant. (B) Omicron variant.", "After controlling the outbreak in the Alpha VOC period, there was monitoring of domestic cluster infections for the Delta VOC since August 8, 2021, and the Omicron VOC from December 10, 2021. After mid-August, there had been a cluster infection during the Delta VOC phase of the COVID-19 pandemic. The estimated results (Table 1, Delta VOC period) showed that the Dci/Imc remained at 10.01% (95% CrI 6.85%-13.32%), although the Delta VOC had higher transmissibility and escaped vaccine-induced immunity. The left panel of Figure 4 shows that for 2 weeks (August 22 to August 28, 2021, and September 19 to September 25, 2021) when a surge in the number of domestic cases as a result of Dci/Imc was expected (green circle), the observed number of domestic cases was far below the threshold of outbreak. This can be attributed to the implementation of enhanced containment measures, including the strengthening of border control strategies with multiple tests on arrival and during quarantine, the collective quarantine strategy, and the elevated alerts of NPIs to levels 2 and 3 since the outbreak in May 2021 in Taiwan. Since then, the weekly observed cases were below the alert threshold owing to NPIs with a level 2 alert and the rapid administration of vaccines.\nThe right panel of Figure 4 shows the observed and predicted numbers of cases along with the alert threshold during the Omicron VOC period. Given the increasing coverage rate of vaccination, the risk of domestic cluster infection per imported case for the Omicron BA.1 VOC reduced to 4.59% (95% CrI 3.66%-5.75%), but an upsurge in domestic cases was still observed because the Omicron BA.1 VOC was considered to have a high transmission probability. In the week from January 9 to January 15, 2022, the observed number exceeded the alert threshold, indicating a high potential for a community-acquired outbreak. There was indeed a small-scale community-acquired outbreak of the Omicron BA.1 VOC. After a series of containment measures, including rapid RT-PCR tests for inbound passengers on arrival coupled with stringent quarantine and isolation, rapid booster vaccination, and enhanced NPIs with a level 2 alert in the community, the community-acquired outbreak subsided by the end of February 2022 (Figure 5).\nThere was a return to the imported-domestic transmission model, with the surveillance metric of Dci/Imc estimated at 4.29% (95% CrI 3.52%-5.19%) from February until March 20, 2022. After that, a similar circumstance beyond the alert threshold was noted for the invasion of the imported Omicron BA.2 VOC, and a community-acquired outbreak started from March 20 to 26, 2022, resulting in a large-scale community-acquired outbreak from early April until July 2022.", "To validate the proposed model and extend its application to different periods of SARS-CoV-2 variants, the proposed extra-Poisson regression model was applied to data on the New Zealand COVID-19 outbreak in 2020. Multimedia Appendix 5 shows the estimated results obtained. Notably, in New Zealand, the risk of Dci/Imc per week increased to 9.38% (95% CrI 8.88%-9.86%), which was close to the estimated results based on Taiwan data (9.54%, 95% CrI 6.44%-12.59%) in the same period. Details regarding the spatial temporal distribution of COVID-19 outbreaks by types of cases in New Zealand are provided in Multimedia Appendix 6. Multimedia Appendix 7 shows the predicted number of domestic cases by using the parameters trained from the empirical data of New Zealand (Multimedia Appendix 5). Similar to the application in Taiwan, the risk of an outbreak associated with imported cases could be assessed by comparing the observed cases (red dot in Multimedia Appendix 7) with the alert threshold (dotted line in Multimedia Appendix 7). The detailed interpretation of the results of this external validation is elaborated in Multimedia Appendix 8.", "Many cyclical community-acquired outbreaks in each country or region during the COVID-19 pandemic have been noted from 2020 to 2022, and these epidemics have occurred in parallel with the evolution of various emerging SARS-CoV-2 variants, including the wild-type/D614G strain during the non-VOC phase and the Alpha, Beta, Gamma, Delta, and Omicron strains during the VOC phase. More importantly, when facing emerging variants, there were corresponding chains of containment measures with the following 3 serial steps: (1) border control of imported cases together with quarantine and isolation; (2) contact tracing and epidemic investigation of domestic cluster infection together with testing for detecting the foci of infection earlier (small households to large institutions); and (3) control of large-scale community-acquired infection with population-based approaches, mainly involving NPIs and mass vaccination. Although most countries focus on steps 1 and 2 for averting community-acquired outbreaks in the beginning, they end up having no choice but to adopt step 3 involving population-based approaches. Accordingly, most epidemic surveillance models still follow traditional surveillance metrics like the effective basic reproductive number (Rt) for containing community-acquired outbreaks. However, it is still important to develop a new surveillance model with new surveillance metrics commensurate with steps 1 and 2 for forestalling community-acquired outbreaks when facing emerging SARS-CoV-2 variants like the updated Omicron subvariants BA.4/BA.5. Most importantly, such a new surveillance model with useful metrics can be robustly applied across countries and time, covering various emerging SARS-CoV-2 variants detected from imported cases.\nUsing Taiwan empirical data, the proposed new surveillance model for monitoring cluster infections in the wake of imported cases was assessed in 5 periods (wild-type, D614G, Alpha VOC, Delta VOC, and Omicron VOC). The first metric of Dci/Imc per week was used to estimate the effect size of the risk of infection through imported-domestic transmission. The second metric involving the upper bound of the 95% CrI for predicted domestic cases of cluster infections derived from imported cases 1 week before provided the alert threshold for guiding the preparedness of containment measures for preventing community-acquired outbreaks in each country or region. Such an alert threshold would be affected by the characteristics of each emerging SARS-CoV-2 variant, as well as the underlying coverage rate of vaccination and the extent of NPIs. By using empirical data on imported and domestic COVID-19 cases in Taiwan, the effect size of Dci/Imc and the alert threshold were estimated as 9.54% and 12.59% for the wild-type/D614G strain, 14.14% and 25.10% for the Alpha VOC, 10.05% and 13.32% for the Delta VOC, 4.59% and 5.75% for the Omicron BA.1 VOC, and 4.29% and 5.19% for the Omicron BA.2 VOC, respectively, in 2 periods. It should be noted that the interpretation of the absolute effect size of Dci/Imc across various emerging SARS-CoV-2 variants should be taken with great caution.\nWhen a similar logic is applied to the alert threshold, the threshold value for weekly domestic cases during 3 SARS-CoV-2 variant periods would not go beyond 20 cases under the low coverage rate of vaccination and good performance of NPIs before the Omicron VOC period. After the Omicron VOC period, the alert threshold for weekly domestic cases would be 180 cases under the high vaccination rate and minimal NPIs. Empirical evidence on whether and how community-acquired outbreaks can be averted through different periods of SARS-CoV-2 variants with the proposed new surveillance model has been demonstrated by Taiwan data. Outbreaks were averted during the wild-type and D614G periods. In contrast, large-scale outbreaks could not be averted during the Alpha VOC period when the expected number of domestic cases was far beyond the alert threshold for an outbreak between May 9 and May 12, 2021, because of the increased transmissibility of the Alpha VOC that was supported by the increased risk of imported-domestic transmission in comparison with the wild-type/D614G variant (14.14% vs 9.54%). However, a low level of NPIs might also have contributed to such an outbreak around mid-May 2021 (30%; Figure 5). In the Delta VOC period after excluding the outbreak related to the Alpha VOC in Taiwan, the level of the NPI alert and the strict border control strategies implemented since the outbreak period reduced the risk of imported-domestic transmission to 10.05% (Table 1) and averted a community-acquired outbreak of the Delta VOC.\nGiven the high coverage of full vaccination, lower estimates of Dci/Imc for the Omicron VOC were seen, ranging from 4.3% to 4.6%. As the protective effect of the Oxford/AstraZeneca vaccine in particular started to wane in the community, the observed number of domestic cases was beyond the threshold of outbreak during the early Omicron BA.1 VOC period. Guided by the alert threshold, several containment strategies, including more restricted border control and rapid RT-PCR testing on arrival for travelers, and rapid booster shots for eligible adults, were implemented to avert a large-scale outbreak. However, another large-scale community-acquired outbreak could not be averted in late March 2022 owing to the observed cases going beyond the alert threshold partially due to waning of the protective effect of the mRNA-1273 vaccine (Moderna) or BNT162b2 vaccine (Pfizer–BioNTech).\nAlthough our metrics of the risk for domestic cluster infection and the alert threshold are pivotal in imported cases 1 week before applying the surveillance model for monitoring imported cases 1 week prior to the formation of cluster infections of domestic COVID-19 cases, they are very useful for alerting the surrounding community in proximity to imported cases beyond the threshold of the upper bound of the 95% CrI to enhance NPIs and active rapid testing with effective contact tracing and epidemic investigation for the observed cases. This accounted for the lack of community-acquired outbreaks before mid-May 2021 in Taiwan. Such good control over the COVID-19 epidemic has been reported in previous studies by evaluating NPIs at the individual and population levels [5,19,26], and using the traditional surveillance model for assessing the duration from Rt larger than 1 to Rt smaller than 1 and the case load following the machining learning model [27].\nSeveral previous studies have proposed an early warning model in relation to contact tracing and epidemic investigation before community-acquired outbreaks. However, our study differs from 2 recent previous studies [15,16] that developed an early warning model of COVID-19 outbreaks, in 2 main aspects. First, both studies covered a short period that reflected 1 or 2 SARS-CoV-2 strains and used data based on a single country. They were therefore unable to test the robustness of their models for a series of SARS-CoV-2 variants and samples across countries. Guan et al used human mobility data in Israel over the period from February 1, 2020, to January 7, 2021. Specifically, they trained their model over the period from April 6 to October 24, 2020, and evaluated the model’s predictive ability over 2 very short periods (November 1-30, 2020, and December 1-31, 2021) [15]. Kogan et al employed data in the United States that had been obtained from multiple digital traces over 2 short periods (March 1-May 31, 2020, and June 1-September 30, 2020) [16]. In contrast, the proposed new surveillance model made use of the full chronological empirical data in Taiwan from January 1, 2020, to April 2, 2022, covering various emerging SARS-CoV-2 variants. Our model is robust across a long period involving various variants and across countries covering different geographical and cultural conditions when using imported cases. Second, the data derived from digital traces, for example, Google Trends, used in the studies by Guan et al and Kogan et al may be affected by media activities. Furthermore, it is not easy to obtain accurate real-time data in countries with unavailable technological infrastructure, strong information censoring, and a lack of transparency. Instead of focusing on digital traces, we made use of imported cases that may be less likely to be affected by confounding factors. The use of imported data in our proposed surveillance model can be applied to countries with different political and social conditions and at different technological development stages. Moreover, our study is highly relevant to health regulators and public health policy makers, particularly in countries that have opened their borders and eased/removed NPI measures.\nIn addition to the illustration of the Taiwan experience, the external validation involving New Zealand further adds credibility to the application of this surveillance model in a scenario without an outbreak. This model can also be applied to those countries with controllable community-acquired outbreaks, such as Israel [28] and Qatar [29], after the mass vaccination program since early 2021, to monitor the impact of imported cases on the risk of domestic cluster infection. This is especially important for outbreaks resulting from vaccine breakthrough in countries or regions with high vaccine coverage, such as Singapore [30] and Israel [31], or the rapid waning of booster effectiveness worldwide, possibly affecting the community-level spread of SARS-CoV-2 VOCs [32].\nOne of the major limitations of the current model pertains to the generalization of the proposed new surveillance model. There are 2 major circumstances that require the refinement of the current proposed epidemic surveillance model. The proposed model has not incorporated health care capacity for accommodating the threshold of tolerable COVID-19 cases responsible for each episode of the outbreak. In consideration of resuming prepandemic activity, making allowance for this factor is of paramount importance for the implementation of NPIs and testing given the vaccine coverage rate. Different countries and regions may require different outbreak thresholds based on this global surveillance model. With increasing cases of vaccine breakthrough; the rapid emergence of VOCs with a wide spectrum of immunogenicity, high transmissibility, and resistance to antibodies associated with natural infection or vaccination; and the waning of immunity in the population, there is a high likelihood for the continued spread of SARS-CoV-2 in the population [33]. Given the possibility of a long-term association between SARS-CoV-2 and the human population, the goal of epidemic surveillance may shift from the elimination of this pathogen to a balance among health care capacity, socioeconomic activity, and population immunity. If this occurs, the proposed surveillance model should take this factor into account and should be used as a guide to inform the containment measures required to mitigate large-scale outbreaks according to health care capacity. Moreover, as the border control policy on quarantine and isolation of imported cases gets altered with the advent of high-performance rapid testing and the gradual expansion of vaccine coverage worldwide, the surveillance model for monitoring imported-domestic transmission to avert outbreaks may vary from country to country, depending on the extent of NPIs, administration of tests, coverage rate of vaccines, and administration of vaccine boosters. Such a heterogeneity should be taken into account to refine the surveillance model on imported-domestic transmission when it is applied to avert a large-scale outbreak. More importantly, our new surveillance model and metrics are not meant to replace conventional surveillance and corresponding metrics like Rt for assessing how to eliminate the spread of large-scale community-acquired outbreaks. When a community-acquired outbreak occurs, the conventional surveillance SEIR model is needed to assess the effectiveness of containment measures, as shown in Figure 5. Based on the SEIR model, the Rt decreased from 4.0 to 0.7 from May 18, 2021, to July 31, 2021. The effectiveness of NPIs and testing, which reflects the strategies implemented 2 weeks ago, was over 60% after May 26, 2021, and increased to over 90% after June 14, 2021. A similar finding was noted for a community-acquired outbreak of Omicron BA.2. Again, Rt reduced from 7.7 (value of Rt in early January) to less than 1 (around the end of January; Figure 5B). Our proposed new surveillance model has a supplementary role as a global vigilance method for forestalling large-scale local community-acquired outbreaks of emerging SARS-CoV-2 VOCs in each country and region worldwide.\nIn conclusion, a global new surveillance model and metrics have been proposed for monitoring imported cases of SARS-CoV-2 variants from the non-VOC phase to the VOC phase, using the Taiwan scenario. The new surveillance model and metrics are very useful for forestalling a new large-scale community-acquired outbreak through monitoring of the imported-domestic transmission mode associated with emerging infectious diseases in the future." ]
[ "introduction", "methods", null, null, null, null, null, null, "results", null, null, null, null, null, null, "discussion" ]
[ "COVID-19", "imported case", "surveillance metric", "early detection", "community-acquired outbreak" ]
The Risk of Hospitalization and Mortality After Breakthrough SARS-CoV-2 Infection by Vaccine Type: Observational Study of Medical Claims Data.
36265135
Several risk factors have been identified for severe COVID-19 disease by the scientific community. In this paper, we focus on understanding the risks for severe COVID-19 infections after vaccination (ie, in breakthrough SARS-CoV-2 infections). Studying these risks by vaccine type, age, sex, comorbidities, and any prior SARS-CoV-2 infection is important to policy makers planning further vaccination efforts.
BACKGROUND
We performed Cox regression analysis to calculate the risk factors for developing a severe breakthrough SARS-CoV-2 infection in the study cohort by controlling for vaccine type, age, sex, comorbidities, and a prior SARS-CoV-2 infection.
METHODS
We found lower hazard ratios for those receiving the Moderna vaccine (P<.001) and Pfizer vaccine (P<.001), with the lowest hazard rates being for Moderna, as compared to those who received the Janssen vaccine, independent of age, sex, comorbidities, vaccine type, and prior SARS-CoV-2 infection. Further, individuals who had a SARS-CoV-2 infection prior to vaccination had some increased protection over and above the protection already provided by the vaccines, from hospitalization (P=.001) and death (P=.04), independent of age, sex, comorbidities, and vaccine type. We found that the top statistically significant risk factors for severe breakthrough SARS-CoV-2 infections were age of >50, male gender, moderate and severe renal failure, severe liver disease, leukemia, chronic lung disease, coagulopathy, and alcohol abuse.
RESULTS
Among individuals who were fully vaccinated, the risk of severe breakthrough SARS-CoV-2 infection was lower for recipients of the Moderna or Pfizer vaccines and higher for recipients of the Janssen vaccine. These results from our analysis at a population level will be helpful to public health policy makers. Our result on the influence of a previous SARS-CoV-2 infection necessitates further research into the impact of multiple exposures on the risk of developing severe COVID-19.
CONCLUSIONS
[ "Humans", "Male", "COVID-19", "Viral Vaccines", "SARS-CoV-2", "Vaccination", "Hospitalization" ]
9645422
Introduction
Despite widespread COVID-19 vaccination, high community levels of SARS-CoV-2 circulating throughout the United States have led to many breakthrough SARS-CoV-2 infections [1-3]. Breakthrough infections, where fully vaccinated individuals who are exposed to SARS-CoV-2 get infected, are generally uncommon (0.02% of fully vaccinated individuals reported developing breakthrough infections in a Washington state cohort [4]) and are generally less severe than infections in unvaccinated individuals [5,6]. There now exists a large body of literature studying the risk factors for severe COVID-19 disease, much of which has involved studies in unvaccinated populations [7-10], prior to the large-scale availability of vaccines. Studies on how these risks vary after vaccination are fewer in comparison, mostly focused on vaccine effectiveness in preventing SARS-CoV-2 infections or the influence of specific variants on vaccine effectiveness [11]. The impact of underlying factors on breakthrough infections are quite challenging to understand outside of randomized, placebo-controlled, double-blind field trials due to variation in their severity, distribution in the population, and contribution to transmission [11,12]. Early studies have found that a third dose of vaccine reduces the viral load in breakthrough infections, even for newer variants such as delta and omicron [13,14]. To understand the comparative advantages of the various vaccines [15], it is important to know the rate of severe COVID-19 disease leading to hospitalization or death among individuals who are fully vaccinated [16], as this will help policy makers. While the risk of breakthrough SARS-CoV-2 infection has recently been reported by type of vaccine [17], little information exists regarding the risk of hospitalization or mortality by vaccine type for breakthrough infections [1]. In addition, while a prior SARS-CoV-2 infection is associated with a lower risk of breakthrough infection, it is unknown how large an effect a prior infection has on the severity of breakthrough COVID-19 infections, should one occur [18]. There has been a growing need for retrospective studies on severe breakthrough infections to address the misinformation and vaccine hesitancy in social and public spheres [19]. In this paper, we used de-identified US medical claims records from Change Healthcare to estimate the risk of hospitalization and death, by vaccine type, age, sex, comorbidity factors and previous SARS-CoV2 infection, among SARS-CoV-2 breakthrough infections that occurred between March 10, 2021, and October 14, 2021.
Methods
[SUBTITLE] Ethics Approval and Consent to Participate [SUBSECTION] This study does not constitute as human subjects research due to the use and reporting of only deidentified observational data as determined by the human subjects committee of the University of Washington and thus does not require the review and approval by the institutional review board at the University of Washington. This study does not constitute as human subjects research due to the use and reporting of only deidentified observational data as determined by the human subjects committee of the University of Washington and thus does not require the review and approval by the institutional review board at the University of Washington. [SUBTITLE] Data Source [SUBSECTION] Our study uses de-identified US medical claims records from Change Healthcare collected over a period from March 1, 2020, to October 14, 2021, encompassing over 100 million records from over 8 million patients. Medical claims data contain details about a patient’s interaction with the medical system, which are needed for the accurate billing of the transactions. Each claims record contains patient demographic information, International Classification of Diseases, 10th Revision (ICD-10) codes indicating primary diagnosis and secondary diagnoses, place of diagnosis, ICD-10 codes of procedures performed, patient status at the end of the visit, dates pertaining to the event (where different “from” and “to” dates indicate longer visits whereas the same “from” and “to” dates are for outpatient visits). Our claims data set includes primarily open claims and a subset of closed payer claims that are normalized for analytics purposes. The open claims are derived from broad-based health care sources and consist of all medical claims that Change Healthcare processes and for which it has the usage rights. The closed claims are derived directly from the payer (ie, health insurance provider) and capture nearly all events that occur during the patient’s enrollment period. Roughly 95% of the claims used for this study are commercial, and 5% are Medicare Advantage or other types of plans. Our study uses de-identified US medical claims records from Change Healthcare collected over a period from March 1, 2020, to October 14, 2021, encompassing over 100 million records from over 8 million patients. Medical claims data contain details about a patient’s interaction with the medical system, which are needed for the accurate billing of the transactions. Each claims record contains patient demographic information, International Classification of Diseases, 10th Revision (ICD-10) codes indicating primary diagnosis and secondary diagnoses, place of diagnosis, ICD-10 codes of procedures performed, patient status at the end of the visit, dates pertaining to the event (where different “from” and “to” dates indicate longer visits whereas the same “from” and “to” dates are for outpatient visits). Our claims data set includes primarily open claims and a subset of closed payer claims that are normalized for analytics purposes. The open claims are derived from broad-based health care sources and consist of all medical claims that Change Healthcare processes and for which it has the usage rights. The closed claims are derived directly from the payer (ie, health insurance provider) and capture nearly all events that occur during the patient’s enrollment period. Roughly 95% of the claims used for this study are commercial, and 5% are Medicare Advantage or other types of plans. [SUBTITLE] Study Population [SUBSECTION] Our data set of 8.18 million individuals contains only COVID-19 positive patients, defined as patients with at least one claims record with the ICD-10 diagnosis codes of “U07.1” or “U07.2” in any diagnosis field. We limited our analysis to individuals who had a primary diagnosis of “U07.1” (this is indicated by the principal diagnosis code, which encodes the primary diagnosis rendered by the medical facility or the primary cause of the visit). This ICD-10 diagnosis code indicates a COVID-19 diagnosis where the virus was identified in a lab-confirmed report. We exclude patients for whom the code of U07.1 appears in the “other diagnosis” fields, which contain the list of diagnoses made in addition to the primary cause of visit, which can be any other medical condition such as cancer. We also exclude patients with the code U07.2, which indicates a non-lab–confirmed COVID-19 diagnosis. Subsequently, fully vaccinated individuals are identified by looking for procedure codes encoding the second doses of Pfizer (0002A) and Moderna (0012A) vaccines and the first dose of the Janssen (0031A) vaccine. We do not exclude patients with missing first dose claims records (~5% of the final study cohort), because patients who went to vaccination camps and were not required to provide insurance information would have missing first dose claims records. Some of these ~5% patients with missing first dose claims records may have had mixed vaccines (eg, Pfizer for the first dose and Moderna for the second dose). Since we did not believe that this will be a significant fraction of the vaccinated population, we do not exclude them. Breakthrough patients were defined as those who had a primary, lab-confirmed COVID-19 diagnosis at least 14 days after the date of vaccination. Please see Figure S1 in Multimedia Appendix 1 for a flow diagram showing the criteria used for cohort selection. Our data set of 8.18 million individuals contains only COVID-19 positive patients, defined as patients with at least one claims record with the ICD-10 diagnosis codes of “U07.1” or “U07.2” in any diagnosis field. We limited our analysis to individuals who had a primary diagnosis of “U07.1” (this is indicated by the principal diagnosis code, which encodes the primary diagnosis rendered by the medical facility or the primary cause of the visit). This ICD-10 diagnosis code indicates a COVID-19 diagnosis where the virus was identified in a lab-confirmed report. We exclude patients for whom the code of U07.1 appears in the “other diagnosis” fields, which contain the list of diagnoses made in addition to the primary cause of visit, which can be any other medical condition such as cancer. We also exclude patients with the code U07.2, which indicates a non-lab–confirmed COVID-19 diagnosis. Subsequently, fully vaccinated individuals are identified by looking for procedure codes encoding the second doses of Pfizer (0002A) and Moderna (0012A) vaccines and the first dose of the Janssen (0031A) vaccine. We do not exclude patients with missing first dose claims records (~5% of the final study cohort), because patients who went to vaccination camps and were not required to provide insurance information would have missing first dose claims records. Some of these ~5% patients with missing first dose claims records may have had mixed vaccines (eg, Pfizer for the first dose and Moderna for the second dose). Since we did not believe that this will be a significant fraction of the vaccinated population, we do not exclude them. Breakthrough patients were defined as those who had a primary, lab-confirmed COVID-19 diagnosis at least 14 days after the date of vaccination. Please see Figure S1 in Multimedia Appendix 1 for a flow diagram showing the criteria used for cohort selection. [SUBTITLE] Hospitalization and Mortality [SUBSECTION] We explicitly identify hospitalization by looking for claims where the claim type is “institutional” or “professional” and the bill type indicates an “inpatient” facility. We also look at the dates associated with the hospital stay and only consider patients whose admission duration was at least 2 days (derived from the “admission_from” and “admission_to” date fields). For mortality, we look at the patient status code and consider all codes indicating “expired.” As described already, we only consider cases where the primary diagnosis was COVID-19 for both hospitalization and expiration. The patient status code is available for all hospitalized patients but only for 42% of the outpatients (who went to clinics). Among patients who had the patient status code available, we found only 17 (0.22%) deaths out of a total of 7843 outpatients; therefore, we consider outpatients with missing patient status to be alive. We explicitly identify hospitalization by looking for claims where the claim type is “institutional” or “professional” and the bill type indicates an “inpatient” facility. We also look at the dates associated with the hospital stay and only consider patients whose admission duration was at least 2 days (derived from the “admission_from” and “admission_to” date fields). For mortality, we look at the patient status code and consider all codes indicating “expired.” As described already, we only consider cases where the primary diagnosis was COVID-19 for both hospitalization and expiration. The patient status code is available for all hospitalized patients but only for 42% of the outpatients (who went to clinics). Among patients who had the patient status code available, we found only 17 (0.22%) deaths out of a total of 7843 outpatients; therefore, we consider outpatients with missing patient status to be alive. [SUBTITLE] Study Period [SUBSECTION] COVID-19 vaccinations began in the United States in late December 2020. By late February 2021, the Pfizer-BioNTech (Pfizer), Moderna, and Johnson & Johnson (J&J/Janssen) vaccines were all approved for emergency use authorization. The Pfizer and Moderna vaccination drives started much earlier, in late December (Figures S2 and S3 in Multimedia Appendix 1), as compared to that of the Janssen vaccines, which also saw a stall in vaccine rates in mid-April (Figure S4 in Multimedia Appendix 1). To keep the COVID-19 exposure of the individuals taking any of the 3 vaccines consistent, we use the same study window, though we have data for Pfizer and Moderna from late December. We construct our cohort to consist of individuals who were fully vaccinated between March 10, 2021, and April 27, 2021, the period during which all 3 vaccines were being widely administered. Every individual in this cohort was followed from the date of vaccination of each individual up to the end of the study period, October 14, 2021. The study period over the entire cohort is thus March 10, 2021, to October 14, 2021. In Figure S6 in Multimedia Appendix 1, we show statistics showing the number of days of follow-up after full vaccination by vaccine type in our study cohort. COVID-19 vaccinations began in the United States in late December 2020. By late February 2021, the Pfizer-BioNTech (Pfizer), Moderna, and Johnson & Johnson (J&J/Janssen) vaccines were all approved for emergency use authorization. The Pfizer and Moderna vaccination drives started much earlier, in late December (Figures S2 and S3 in Multimedia Appendix 1), as compared to that of the Janssen vaccines, which also saw a stall in vaccine rates in mid-April (Figure S4 in Multimedia Appendix 1). To keep the COVID-19 exposure of the individuals taking any of the 3 vaccines consistent, we use the same study window, though we have data for Pfizer and Moderna from late December. We construct our cohort to consist of individuals who were fully vaccinated between March 10, 2021, and April 27, 2021, the period during which all 3 vaccines were being widely administered. Every individual in this cohort was followed from the date of vaccination of each individual up to the end of the study period, October 14, 2021. The study period over the entire cohort is thus March 10, 2021, to October 14, 2021. In Figure S6 in Multimedia Appendix 1, we show statistics showing the number of days of follow-up after full vaccination by vaccine type in our study cohort. [SUBTITLE] Comorbidities [SUBSECTION] Preexisting comorbidities were defined based on ICD-10 codes assigned to medical encounters, which contain pointers to previously diagnosed conditions, using claims records during the 6-month period from March 2020 to September 2020. This period does not overlap with the study period, so events during the study period will not also be counted as comorbidities. The Elixhauser comorbidity index [20] was used to define comorbid conditions. This index has a series of codes that define comorbidities with each code mapping to one or several ICD-10 diagnosis codes. For example, the Elixhauser code “BLDLOSS” (blood loss) includes the following four ICD-10 diagnosis codes: D50.0, O90.81, O99.02, and O99.03. We provide the index that we used and the corresponding ICD-10 codes in Multimedia Appendix 2. We also show the relative abundance of comorbidities in our cohort, by vaccine type, in Table S2 and Figure S5 in Multimedia Appendix 1. Preexisting comorbidities were defined based on ICD-10 codes assigned to medical encounters, which contain pointers to previously diagnosed conditions, using claims records during the 6-month period from March 2020 to September 2020. This period does not overlap with the study period, so events during the study period will not also be counted as comorbidities. The Elixhauser comorbidity index [20] was used to define comorbid conditions. This index has a series of codes that define comorbidities with each code mapping to one or several ICD-10 diagnosis codes. For example, the Elixhauser code “BLDLOSS” (blood loss) includes the following four ICD-10 diagnosis codes: D50.0, O90.81, O99.02, and O99.03. We provide the index that we used and the corresponding ICD-10 codes in Multimedia Appendix 2. We also show the relative abundance of comorbidities in our cohort, by vaccine type, in Table S2 and Figure S5 in Multimedia Appendix 1. [SUBTITLE] Previous COVID-19 Infection [SUBSECTION] Since some of the individuals in our cohort may have had a COVID-19 infection during the year 2020, we introduce an additional feature to encode the effect of already being infected with COVID-19. This feature is “yes” if we see a claim involving a COVID-19 diagnosis in any diagnosis field, in the period from March 1, 2020, to the beginning of the study period, March 10, 2021. Since some of the individuals in our cohort may have had a COVID-19 infection during the year 2020, we introduce an additional feature to encode the effect of already being infected with COVID-19. This feature is “yes” if we see a claim involving a COVID-19 diagnosis in any diagnosis field, in the period from March 1, 2020, to the beginning of the study period, March 10, 2021. [SUBTITLE] Statistical Methods [SUBSECTION] Date of full vaccination was defined as 14 days after (1) a single Janssen vaccine, (2) the second Moderna vaccine dose, or (3) the second Pfizer vaccine dose. Cox proportional hazards regression was used to estimate univariate hazard ratios (HRs) and multivariable HRs in a model including the following features: age (categorized), sex (male and female), vaccine type, Elixhauser comorbidities (encoded as independent binary variables), and SARS-CoV-2 infection prior to the first dose of vaccination (yes or no). We remove the negligible number of individuals with sex=unknown. We also model interactions between vaccine type and all other covariates as well as previous infection and all other covariates but find that none were statistically significant. Further, the interaction terms had a negligible impact on the hazard ratios of the other terms and were thus removed for greater clarity in the results. All analyses were performed using the “coxph” function from the R package “survival” (R Foundation for Statistical Computing) [21]. Date of full vaccination was defined as 14 days after (1) a single Janssen vaccine, (2) the second Moderna vaccine dose, or (3) the second Pfizer vaccine dose. Cox proportional hazards regression was used to estimate univariate hazard ratios (HRs) and multivariable HRs in a model including the following features: age (categorized), sex (male and female), vaccine type, Elixhauser comorbidities (encoded as independent binary variables), and SARS-CoV-2 infection prior to the first dose of vaccination (yes or no). We remove the negligible number of individuals with sex=unknown. We also model interactions between vaccine type and all other covariates as well as previous infection and all other covariates but find that none were statistically significant. Further, the interaction terms had a negligible impact on the hazard ratios of the other terms and were thus removed for greater clarity in the results. All analyses were performed using the “coxph” function from the R package “survival” (R Foundation for Statistical Computing) [21].
Results
Our study includes 19,815 fully vaccinated patients with breakthrough SARS-CoV-2 infections between March 10, 2021, and October 14, 2021. Of those patients, 11,339 (57.22%) received the Pfizer vaccine, 5480 (27.66%) received the Moderna vaccine, and 2996 (15.12%) received the Janssen vaccine. Breakthrough cases receiving Janssen were younger than those receiving Pfizer or Moderna and had a slightly greater proportion of male patients (Table 1). Breakthrough cases receiving Moderna had a greater proportion of patients with COVID-19 prior to vaccination. Risk of hospitalization and mortality among breakthrough cases increased with older age and was higher for male patients (Table 2). In multivariable analyses controlling for age, male sex, comorbidities, and prior SARS-CoV-2 infection, the risk of hospitalization was the lowest for breakthrough cases receiving the Moderna vaccine (adjusted hazard ratio [aHR]: 0.42, 95% CI 0.35-0.5; P<.001), comparably low for Pfizer vaccinated individuals (aHR: 0.45, 95% CI 0.39-0.53; P<.001), compared with that for the recipients of the Janssen vaccine. The comorbidities with statistically significant HRs for hospitalization or mortality from a breakthrough SARS-CoV-2 infection include severe liver disease, moderate and severe renal failures, alcohol abuse, chronic lung disease, coagulopathy, cancers, anemia, seizures, and arthritis (Table S1 in Multimedia Appendix 1). Characteristics of SARS-CoV-2 breakthrough infections cohort tracked from March 10, 2021, to October 14, 2021. Prevalence of comorbidities by vaccine type is shown in Figure S5 in Multimedia Appendix 1. Correlates of hospitalization and mortality after breakthrough SARS-CoV-2 infection, estimated from Cox proportional hazards models. We show the adjusted hazard ratio (aHR) and the 95% CI for the significant correlates (P values indicated via superscripts d, e, and f). An aHR of <1.0 indicates a lower risk of hospitalization or mortality as compared to the baseline population for that covariate (analogously, aHR>1.0 indicates a higher risk than the baseline). Hazard ratios (HRs) of comorbidities are shown in Table S1 in Multimedia Appendix 1. aIncidence per 100 person years. bn=19,815; events=1140. cn=19,815; events=159. dP<.001. eP<.05. fP<.01. We see a similar trend with the risk of mortality for breakthrough cases, with the risk being the lowest for those receiving the Moderna vaccines (aHR: 0.38, 95% CI 0.23-0.62; P<.001) and comparably lower for Pfizer recipients (aHR: 0.43, 95% CI 0.28-0.65; P<.001) as compared to that for Janssen recipients. Finally, as expected, the protection offered by vaccines was enhanced for breakthrough cases who already had a previous SARS-CoV-2 infection. These individuals were 40% less likely to be hospitalized due to COVID-19 (aHR: 0.57, 95% CI 0.41-0.80; P=.001) and four times less likely to die of COVID-19 (aHR: 0.22, 95% CI 0.05-0.91; P=.04), when compared to those without a prior SARS-CoV-2 infection independent of age, sex, comorbidities, and vaccine type. We repeat this analysis by excluding the population who had a prior SARS-CoV-2 infection for completeness and show the resulting HRs Table S3 in Multimedia Appendix 1.
null
null
[ "Ethics Approval and Consent to Participate", "Data Source", "Study Population", "Hospitalization and Mortality", "Study Period", "Comorbidities", "Previous COVID-19 Infection", "Statistical Methods", "Principal Findings", "Limitations", "Conclusions" ]
[ "This study does not constitute as human subjects research due to the use and reporting of only deidentified observational data as determined by the human subjects committee of the University of Washington and thus does not require the review and approval by the institutional review board at the University of Washington.", "Our study uses de-identified US medical claims records from Change Healthcare collected over a period from March 1, 2020, to October 14, 2021, encompassing over 100 million records from over 8 million patients. Medical claims data contain details about a patient’s interaction with the medical system, which are needed for the accurate billing of the transactions. Each claims record contains patient demographic information, International Classification of Diseases, 10th Revision (ICD-10) codes indicating primary diagnosis and secondary diagnoses, place of diagnosis, ICD-10 codes of procedures performed, patient status at the end of the visit, dates pertaining to the event (where different “from” and “to” dates indicate longer visits whereas the same “from” and “to” dates are for outpatient visits).\nOur claims data set includes primarily open claims and a subset of closed payer claims that are normalized for analytics purposes. The open claims are derived from broad-based health care sources and consist of all medical claims that Change Healthcare processes and for which it has the usage rights. The closed claims are derived directly from the payer (ie, health insurance provider) and capture nearly all events that occur during the patient’s enrollment period. Roughly 95% of the claims used for this study are commercial, and 5% are Medicare Advantage or other types of plans.", "Our data set of 8.18 million individuals contains only COVID-19 positive patients, defined as patients with at least one claims record with the ICD-10 diagnosis codes of “U07.1” or “U07.2” in any diagnosis field. We limited our analysis to individuals who had a primary diagnosis of “U07.1” (this is indicated by the principal diagnosis code, which encodes the primary diagnosis rendered by the medical facility or the primary cause of the visit). This ICD-10 diagnosis code indicates a COVID-19 diagnosis where the virus was identified in a lab-confirmed report. We exclude patients for whom the code of U07.1 appears in the “other diagnosis” fields, which contain the list of diagnoses made in addition to the primary cause of visit, which can be any other medical condition such as cancer. We also exclude patients with the code U07.2, which indicates a non-lab–confirmed COVID-19 diagnosis.\nSubsequently, fully vaccinated individuals are identified by looking for procedure codes encoding the second doses of Pfizer (0002A) and Moderna (0012A) vaccines and the first dose of the Janssen (0031A) vaccine. We do not exclude patients with missing first dose claims records (~5% of the final study cohort), because patients who went to vaccination camps and were not required to provide insurance information would have missing first dose claims records. Some of these ~5% patients with missing first dose claims records may have had mixed vaccines (eg, Pfizer for the first dose and Moderna for the second dose). Since we did not believe that this will be a significant fraction of the vaccinated population, we do not exclude them. Breakthrough patients were defined as those who had a primary, lab-confirmed COVID-19 diagnosis at least 14 days after the date of vaccination. Please see Figure S1 in Multimedia Appendix 1 for a flow diagram showing the criteria used for cohort selection.", "We explicitly identify hospitalization by looking for claims where the claim type is “institutional” or “professional” and the bill type indicates an “inpatient” facility. We also look at the dates associated with the hospital stay and only consider patients whose admission duration was at least 2 days (derived from the “admission_from” and “admission_to” date fields). For mortality, we look at the patient status code and consider all codes indicating “expired.” As described already, we only consider cases where the primary diagnosis was COVID-19 for both hospitalization and expiration. The patient status code is available for all hospitalized patients but only for 42% of the outpatients (who went to clinics). Among patients who had the patient status code available, we found only 17 (0.22%) deaths out of a total of 7843 outpatients; therefore, we consider outpatients with missing patient status to be alive.", "COVID-19 vaccinations began in the United States in late December 2020. By late February 2021, the Pfizer-BioNTech (Pfizer), Moderna, and Johnson & Johnson (J&J/Janssen) vaccines were all approved for emergency use authorization. The Pfizer and Moderna vaccination drives started much earlier, in late December (Figures S2 and S3 in Multimedia Appendix 1), as compared to that of the Janssen vaccines, which also saw a stall in vaccine rates in mid-April (Figure S4 in Multimedia Appendix 1). To keep the COVID-19 exposure of the individuals taking any of the 3 vaccines consistent, we use the same study window, though we have data for Pfizer and Moderna from late December. We construct our cohort to consist of individuals who were fully vaccinated between March 10, 2021, and April 27, 2021, the period during which all 3 vaccines were being widely administered. Every individual in this cohort was followed from the date of vaccination of each individual up to the end of the study period, October 14, 2021. The study period over the entire cohort is thus March 10, 2021, to October 14, 2021. In Figure S6 in Multimedia Appendix 1, we show statistics showing the number of days of follow-up after full vaccination by vaccine type in our study cohort.", "Preexisting comorbidities were defined based on ICD-10 codes assigned to medical encounters, which contain pointers to previously diagnosed conditions, using claims records during the 6-month period from March 2020 to September 2020. This period does not overlap with the study period, so events during the study period will not also be counted as comorbidities. The Elixhauser comorbidity index [20] was used to define comorbid conditions. This index has a series of codes that define comorbidities with each code mapping to one or several ICD-10 diagnosis codes. For example, the Elixhauser code “BLDLOSS” (blood loss) includes the following four ICD-10 diagnosis codes: D50.0, O90.81, O99.02, and O99.03. We provide the index that we used and the corresponding ICD-10 codes in Multimedia Appendix 2. We also show the relative abundance of comorbidities in our cohort, by vaccine type, in Table S2 and Figure S5 in Multimedia Appendix 1.", "Since some of the individuals in our cohort may have had a COVID-19 infection during the year 2020, we introduce an additional feature to encode the effect of already being infected with COVID-19. This feature is “yes” if we see a claim involving a COVID-19 diagnosis in any diagnosis field, in the period from March 1, 2020, to the beginning of the study period, March 10, 2021.", "Date of full vaccination was defined as 14 days after (1) a single Janssen vaccine, (2) the second Moderna vaccine dose, or (3) the second Pfizer vaccine dose. Cox proportional hazards regression was used to estimate univariate hazard ratios (HRs) and multivariable HRs in a model including the following features: age (categorized), sex (male and female), vaccine type, Elixhauser comorbidities (encoded as independent binary variables), and SARS-CoV-2 infection prior to the first dose of vaccination (yes or no). We remove the negligible number of individuals with sex=unknown. We also model interactions between vaccine type and all other covariates as well as previous infection and all other covariates but find that none were statistically significant. Further, the interaction terms had a negligible impact on the hazard ratios of the other terms and were thus removed for greater clarity in the results. All analyses were performed using the “coxph” function from the R package “survival” (R Foundation for Statistical Computing) [21].", "Using medical claims data, we found that the risk of hospitalization in SARS-CoV-2 breakthrough infections was lower for those receiving the Moderna and Pfizer vaccines compared to those receiving the Janssen vaccine. The risk of mortality was similarly low in breakthrough infections who received Pfizer and Moderna vaccines compared to those receiving the Janssen vaccine. There was no statistically significant difference between the HRs of Pfizer and Moderna for both risks. We also found older age, male sex, and certain comorbidities to be risk factors for hospitalization and mortality in breakthrough infections. Further, we found that risk of hospitalization was 40% less and risk of death was 75% less in SARS-CoV-2 breakthrough infections among individuals who already had a SARS-CoV-2 infection prior to their vaccination compared with fully vaccinated individuals without a previous SARS-CoV-2 infection. While other studies have reported lower risk of breakthrough infection with previous SARS-CoV-2 infection [18], our study analyzes both hospitalization and mortality and shows that the immunity provided by previous infection seems to increase the protection provided by vaccines, against severe COVID-19, independent of vaccine type, age, comorbidities, and sex. Since our cohort only consists of individuals who were all fully vaccinated, this is by no means a comparison of vaccine-induced immunity against acquired immunity from previous infections.\nExcluding patients who had COVID-19 infection prior to vaccination from our Cox regression analysis results in a similar HR for hospitalization risk in patients who received the Pfizer (aHR=0.42) and Moderna (aHR=0.41) vaccines (Table S3 in Multimedia Appendix 1). This might be explained by the fact that 20.7% of patients who received Moderna had a prior COVID-19 infection as compared to ~13% of patients who received Pfizer. Hence, removing all patients with prior COVID-19 infection reduced the influence of the additional immunity that some of Moderna-vaccinated individuals had.\nA number of studies have found that age has a direct effect on the risk of severe COVID-19 disease [22,23]. We find that the proportion of the elderly cohort in our data set who were hospitalized is much higher than the proportion of the younger cohort (Figure S7 in multimedia Appendix 1). In addition, we find a higher HR for the elderly subset of our study cohort (aHR=2.1 for age>50, aHR=3.3 for age>65, and aHR=5.0 for age>80, as compared to the baseline age group of 35-50 years).\nOur findings comparing vaccine types are similar to those reported by the Centers for Disease Control and Prevention for mortality but provide additional information by vaccine type [1,16]. There have been several studies on individual risk factors such as age [22], specific comorbidities [7], focused populations such as Veterans [24], or large-scale projects such as OpenSAFELY, which involved 17 million unvaccinated patients [10]. Our work advances this body of literature by analyzing vaccine type, age, sex, and 39 different comorbidities in a large cohort of breakthrough patients from the general US population. Some of the risk factors that we find for severe breakthrough SARS-CoV-2 infections, such as age, male gender, and certain comorbidities (eg, chronic lung infection, kidney disease, and cancers) are similar to what has been reported in prior studies of SARS-CoV-2 infections among unvaccinated individuals [8,23,25]. However, we find that some risk factors found by initial studies such as hypertension are not a risk factor for breakthrough COVID-19 hospitalization or death (aHRs of 0.75 and 0.59, respectively), neither are diabetes or congestive heart failure. We instead find that both moderate and severe renal failure are significant risk factors, independent of age or other factors, which agrees with other large-scale studies such as OpenSAFELY [10] and the Global Burden of Disease collaboration [26] which identified that worldwide chronic kidney disease is the most prevalent risk factor for severe COVID-19. Even mild impairment of renal function has been found to be an independent risk factor for COVID-19 infection, hospitalization, and mortality [27].\nLastly, to understand the association between outcomes and the time of vaccination, we incorporate a variable indicating the number of days between full vaccination and the onset of the surge in infections caused by the delta variant. However, our population-based data set is inadequate to derive any significant conclusions vis-à-vis the best time for vaccination in anticipation of a surge.", "Limitations of our study include, first, a lack of access to data on unvaccinated individuals or those who had a negative SARS-CoV-2 test result. The former is due to the lack of a medical claims record for vaccinations that were done in vaccination drives and camps; the absence of a vaccination-related claim in our data set therefore does not imply an unvaccinated individual. Second, our medical claims source consists of mostly privately insured individuals and can thus miss people who may be susceptible to the most adverse outcomes. Another caveat of our data set is that most of the claims are open claims, which have the benefit of capturing a patient’s activities over a longer time frame regardless of their insurance provider, but do not necessarily track all medical encounters of patients.", "Our findings add to the growing literature regarding the risk factors for severe breakthrough SARS-CoV-2 infections in fully vaccinated individuals, where we identify the influence of age, sex, and comorbidities that are risk factors; importantly, we found that previous SARS-CoV-2 infections can provide additional protection over that offered by vaccines against severe disease. Our results also necessitate further studies on the optimal number of vaccine doses to protect from the most severe breakthrough SARS-CoV-2 infections. An important strength of our study is that we consider US-wide breakthrough hospitalizations covering a broad demographic and compare all 3 vaccines, whereas most previous studies lack specific data on Janssen." ]
[ null, null, null, null, null, null, null, null, null, null, null ]
[ "Introduction", "Methods", "Ethics Approval and Consent to Participate", "Data Source", "Study Population", "Hospitalization and Mortality", "Study Period", "Comorbidities", "Previous COVID-19 Infection", "Statistical Methods", "Results", "Discussion", "Principal Findings", "Limitations", "Conclusions" ]
[ "Despite widespread COVID-19 vaccination, high community levels of SARS-CoV-2 circulating throughout the United States have led to many breakthrough SARS-CoV-2 infections [1-3]. Breakthrough infections, where fully vaccinated individuals who are exposed to SARS-CoV-2 get infected, are generally uncommon (0.02% of fully vaccinated individuals reported developing breakthrough infections in a Washington state cohort [4]) and are generally less severe than infections in unvaccinated individuals [5,6]. There now exists a large body of literature studying the risk factors for severe COVID-19 disease, much of which has involved studies in unvaccinated populations [7-10], prior to the large-scale availability of vaccines. Studies on how these risks vary after vaccination are fewer in comparison, mostly focused on vaccine effectiveness in preventing SARS-CoV-2 infections or the influence of specific variants on vaccine effectiveness [11].\nThe impact of underlying factors on breakthrough infections are quite challenging to understand outside of randomized, placebo-controlled, double-blind field trials due to variation in their severity, distribution in the population, and contribution to transmission [11,12]. Early studies have found that a third dose of vaccine reduces the viral load in breakthrough infections, even for newer variants such as delta and omicron [13,14]. To understand the comparative advantages of the various vaccines [15], it is important to know the rate of severe COVID-19 disease leading to hospitalization or death among individuals who are fully vaccinated [16], as this will help policy makers.\nWhile the risk of breakthrough SARS-CoV-2 infection has recently been reported by type of vaccine [17], little information exists regarding the risk of hospitalization or mortality by vaccine type for breakthrough infections [1]. In addition, while a prior SARS-CoV-2 infection is associated with a lower risk of breakthrough infection, it is unknown how large an effect a prior infection has on the severity of breakthrough COVID-19 infections, should one occur [18]. There has been a growing need for retrospective studies on severe breakthrough infections to address the misinformation and vaccine hesitancy in social and public spheres [19].\nIn this paper, we used de-identified US medical claims records from Change Healthcare to estimate the risk of hospitalization and death, by vaccine type, age, sex, comorbidity factors and previous SARS-CoV2 infection, among SARS-CoV-2 breakthrough infections that occurred between March 10, 2021, and October 14, 2021.", "[SUBTITLE] Ethics Approval and Consent to Participate [SUBSECTION] This study does not constitute as human subjects research due to the use and reporting of only deidentified observational data as determined by the human subjects committee of the University of Washington and thus does not require the review and approval by the institutional review board at the University of Washington.\nThis study does not constitute as human subjects research due to the use and reporting of only deidentified observational data as determined by the human subjects committee of the University of Washington and thus does not require the review and approval by the institutional review board at the University of Washington.\n[SUBTITLE] Data Source [SUBSECTION] Our study uses de-identified US medical claims records from Change Healthcare collected over a period from March 1, 2020, to October 14, 2021, encompassing over 100 million records from over 8 million patients. Medical claims data contain details about a patient’s interaction with the medical system, which are needed for the accurate billing of the transactions. Each claims record contains patient demographic information, International Classification of Diseases, 10th Revision (ICD-10) codes indicating primary diagnosis and secondary diagnoses, place of diagnosis, ICD-10 codes of procedures performed, patient status at the end of the visit, dates pertaining to the event (where different “from” and “to” dates indicate longer visits whereas the same “from” and “to” dates are for outpatient visits).\nOur claims data set includes primarily open claims and a subset of closed payer claims that are normalized for analytics purposes. The open claims are derived from broad-based health care sources and consist of all medical claims that Change Healthcare processes and for which it has the usage rights. The closed claims are derived directly from the payer (ie, health insurance provider) and capture nearly all events that occur during the patient’s enrollment period. Roughly 95% of the claims used for this study are commercial, and 5% are Medicare Advantage or other types of plans.\nOur study uses de-identified US medical claims records from Change Healthcare collected over a period from March 1, 2020, to October 14, 2021, encompassing over 100 million records from over 8 million patients. Medical claims data contain details about a patient’s interaction with the medical system, which are needed for the accurate billing of the transactions. Each claims record contains patient demographic information, International Classification of Diseases, 10th Revision (ICD-10) codes indicating primary diagnosis and secondary diagnoses, place of diagnosis, ICD-10 codes of procedures performed, patient status at the end of the visit, dates pertaining to the event (where different “from” and “to” dates indicate longer visits whereas the same “from” and “to” dates are for outpatient visits).\nOur claims data set includes primarily open claims and a subset of closed payer claims that are normalized for analytics purposes. The open claims are derived from broad-based health care sources and consist of all medical claims that Change Healthcare processes and for which it has the usage rights. The closed claims are derived directly from the payer (ie, health insurance provider) and capture nearly all events that occur during the patient’s enrollment period. Roughly 95% of the claims used for this study are commercial, and 5% are Medicare Advantage or other types of plans.\n[SUBTITLE] Study Population [SUBSECTION] Our data set of 8.18 million individuals contains only COVID-19 positive patients, defined as patients with at least one claims record with the ICD-10 diagnosis codes of “U07.1” or “U07.2” in any diagnosis field. We limited our analysis to individuals who had a primary diagnosis of “U07.1” (this is indicated by the principal diagnosis code, which encodes the primary diagnosis rendered by the medical facility or the primary cause of the visit). This ICD-10 diagnosis code indicates a COVID-19 diagnosis where the virus was identified in a lab-confirmed report. We exclude patients for whom the code of U07.1 appears in the “other diagnosis” fields, which contain the list of diagnoses made in addition to the primary cause of visit, which can be any other medical condition such as cancer. We also exclude patients with the code U07.2, which indicates a non-lab–confirmed COVID-19 diagnosis.\nSubsequently, fully vaccinated individuals are identified by looking for procedure codes encoding the second doses of Pfizer (0002A) and Moderna (0012A) vaccines and the first dose of the Janssen (0031A) vaccine. We do not exclude patients with missing first dose claims records (~5% of the final study cohort), because patients who went to vaccination camps and were not required to provide insurance information would have missing first dose claims records. Some of these ~5% patients with missing first dose claims records may have had mixed vaccines (eg, Pfizer for the first dose and Moderna for the second dose). Since we did not believe that this will be a significant fraction of the vaccinated population, we do not exclude them. Breakthrough patients were defined as those who had a primary, lab-confirmed COVID-19 diagnosis at least 14 days after the date of vaccination. Please see Figure S1 in Multimedia Appendix 1 for a flow diagram showing the criteria used for cohort selection.\nOur data set of 8.18 million individuals contains only COVID-19 positive patients, defined as patients with at least one claims record with the ICD-10 diagnosis codes of “U07.1” or “U07.2” in any diagnosis field. We limited our analysis to individuals who had a primary diagnosis of “U07.1” (this is indicated by the principal diagnosis code, which encodes the primary diagnosis rendered by the medical facility or the primary cause of the visit). This ICD-10 diagnosis code indicates a COVID-19 diagnosis where the virus was identified in a lab-confirmed report. We exclude patients for whom the code of U07.1 appears in the “other diagnosis” fields, which contain the list of diagnoses made in addition to the primary cause of visit, which can be any other medical condition such as cancer. We also exclude patients with the code U07.2, which indicates a non-lab–confirmed COVID-19 diagnosis.\nSubsequently, fully vaccinated individuals are identified by looking for procedure codes encoding the second doses of Pfizer (0002A) and Moderna (0012A) vaccines and the first dose of the Janssen (0031A) vaccine. We do not exclude patients with missing first dose claims records (~5% of the final study cohort), because patients who went to vaccination camps and were not required to provide insurance information would have missing first dose claims records. Some of these ~5% patients with missing first dose claims records may have had mixed vaccines (eg, Pfizer for the first dose and Moderna for the second dose). Since we did not believe that this will be a significant fraction of the vaccinated population, we do not exclude them. Breakthrough patients were defined as those who had a primary, lab-confirmed COVID-19 diagnosis at least 14 days after the date of vaccination. Please see Figure S1 in Multimedia Appendix 1 for a flow diagram showing the criteria used for cohort selection.\n[SUBTITLE] Hospitalization and Mortality [SUBSECTION] We explicitly identify hospitalization by looking for claims where the claim type is “institutional” or “professional” and the bill type indicates an “inpatient” facility. We also look at the dates associated with the hospital stay and only consider patients whose admission duration was at least 2 days (derived from the “admission_from” and “admission_to” date fields). For mortality, we look at the patient status code and consider all codes indicating “expired.” As described already, we only consider cases where the primary diagnosis was COVID-19 for both hospitalization and expiration. The patient status code is available for all hospitalized patients but only for 42% of the outpatients (who went to clinics). Among patients who had the patient status code available, we found only 17 (0.22%) deaths out of a total of 7843 outpatients; therefore, we consider outpatients with missing patient status to be alive.\nWe explicitly identify hospitalization by looking for claims where the claim type is “institutional” or “professional” and the bill type indicates an “inpatient” facility. We also look at the dates associated with the hospital stay and only consider patients whose admission duration was at least 2 days (derived from the “admission_from” and “admission_to” date fields). For mortality, we look at the patient status code and consider all codes indicating “expired.” As described already, we only consider cases where the primary diagnosis was COVID-19 for both hospitalization and expiration. The patient status code is available for all hospitalized patients but only for 42% of the outpatients (who went to clinics). Among patients who had the patient status code available, we found only 17 (0.22%) deaths out of a total of 7843 outpatients; therefore, we consider outpatients with missing patient status to be alive.\n[SUBTITLE] Study Period [SUBSECTION] COVID-19 vaccinations began in the United States in late December 2020. By late February 2021, the Pfizer-BioNTech (Pfizer), Moderna, and Johnson & Johnson (J&J/Janssen) vaccines were all approved for emergency use authorization. The Pfizer and Moderna vaccination drives started much earlier, in late December (Figures S2 and S3 in Multimedia Appendix 1), as compared to that of the Janssen vaccines, which also saw a stall in vaccine rates in mid-April (Figure S4 in Multimedia Appendix 1). To keep the COVID-19 exposure of the individuals taking any of the 3 vaccines consistent, we use the same study window, though we have data for Pfizer and Moderna from late December. We construct our cohort to consist of individuals who were fully vaccinated between March 10, 2021, and April 27, 2021, the period during which all 3 vaccines were being widely administered. Every individual in this cohort was followed from the date of vaccination of each individual up to the end of the study period, October 14, 2021. The study period over the entire cohort is thus March 10, 2021, to October 14, 2021. In Figure S6 in Multimedia Appendix 1, we show statistics showing the number of days of follow-up after full vaccination by vaccine type in our study cohort.\nCOVID-19 vaccinations began in the United States in late December 2020. By late February 2021, the Pfizer-BioNTech (Pfizer), Moderna, and Johnson & Johnson (J&J/Janssen) vaccines were all approved for emergency use authorization. The Pfizer and Moderna vaccination drives started much earlier, in late December (Figures S2 and S3 in Multimedia Appendix 1), as compared to that of the Janssen vaccines, which also saw a stall in vaccine rates in mid-April (Figure S4 in Multimedia Appendix 1). To keep the COVID-19 exposure of the individuals taking any of the 3 vaccines consistent, we use the same study window, though we have data for Pfizer and Moderna from late December. We construct our cohort to consist of individuals who were fully vaccinated between March 10, 2021, and April 27, 2021, the period during which all 3 vaccines were being widely administered. Every individual in this cohort was followed from the date of vaccination of each individual up to the end of the study period, October 14, 2021. The study period over the entire cohort is thus March 10, 2021, to October 14, 2021. In Figure S6 in Multimedia Appendix 1, we show statistics showing the number of days of follow-up after full vaccination by vaccine type in our study cohort.\n[SUBTITLE] Comorbidities [SUBSECTION] Preexisting comorbidities were defined based on ICD-10 codes assigned to medical encounters, which contain pointers to previously diagnosed conditions, using claims records during the 6-month period from March 2020 to September 2020. This period does not overlap with the study period, so events during the study period will not also be counted as comorbidities. The Elixhauser comorbidity index [20] was used to define comorbid conditions. This index has a series of codes that define comorbidities with each code mapping to one or several ICD-10 diagnosis codes. For example, the Elixhauser code “BLDLOSS” (blood loss) includes the following four ICD-10 diagnosis codes: D50.0, O90.81, O99.02, and O99.03. We provide the index that we used and the corresponding ICD-10 codes in Multimedia Appendix 2. We also show the relative abundance of comorbidities in our cohort, by vaccine type, in Table S2 and Figure S5 in Multimedia Appendix 1.\nPreexisting comorbidities were defined based on ICD-10 codes assigned to medical encounters, which contain pointers to previously diagnosed conditions, using claims records during the 6-month period from March 2020 to September 2020. This period does not overlap with the study period, so events during the study period will not also be counted as comorbidities. The Elixhauser comorbidity index [20] was used to define comorbid conditions. This index has a series of codes that define comorbidities with each code mapping to one or several ICD-10 diagnosis codes. For example, the Elixhauser code “BLDLOSS” (blood loss) includes the following four ICD-10 diagnosis codes: D50.0, O90.81, O99.02, and O99.03. We provide the index that we used and the corresponding ICD-10 codes in Multimedia Appendix 2. We also show the relative abundance of comorbidities in our cohort, by vaccine type, in Table S2 and Figure S5 in Multimedia Appendix 1.\n[SUBTITLE] Previous COVID-19 Infection [SUBSECTION] Since some of the individuals in our cohort may have had a COVID-19 infection during the year 2020, we introduce an additional feature to encode the effect of already being infected with COVID-19. This feature is “yes” if we see a claim involving a COVID-19 diagnosis in any diagnosis field, in the period from March 1, 2020, to the beginning of the study period, March 10, 2021.\nSince some of the individuals in our cohort may have had a COVID-19 infection during the year 2020, we introduce an additional feature to encode the effect of already being infected with COVID-19. This feature is “yes” if we see a claim involving a COVID-19 diagnosis in any diagnosis field, in the period from March 1, 2020, to the beginning of the study period, March 10, 2021.\n[SUBTITLE] Statistical Methods [SUBSECTION] Date of full vaccination was defined as 14 days after (1) a single Janssen vaccine, (2) the second Moderna vaccine dose, or (3) the second Pfizer vaccine dose. Cox proportional hazards regression was used to estimate univariate hazard ratios (HRs) and multivariable HRs in a model including the following features: age (categorized), sex (male and female), vaccine type, Elixhauser comorbidities (encoded as independent binary variables), and SARS-CoV-2 infection prior to the first dose of vaccination (yes or no). We remove the negligible number of individuals with sex=unknown. We also model interactions between vaccine type and all other covariates as well as previous infection and all other covariates but find that none were statistically significant. Further, the interaction terms had a negligible impact on the hazard ratios of the other terms and were thus removed for greater clarity in the results. All analyses were performed using the “coxph” function from the R package “survival” (R Foundation for Statistical Computing) [21].\nDate of full vaccination was defined as 14 days after (1) a single Janssen vaccine, (2) the second Moderna vaccine dose, or (3) the second Pfizer vaccine dose. Cox proportional hazards regression was used to estimate univariate hazard ratios (HRs) and multivariable HRs in a model including the following features: age (categorized), sex (male and female), vaccine type, Elixhauser comorbidities (encoded as independent binary variables), and SARS-CoV-2 infection prior to the first dose of vaccination (yes or no). We remove the negligible number of individuals with sex=unknown. We also model interactions between vaccine type and all other covariates as well as previous infection and all other covariates but find that none were statistically significant. Further, the interaction terms had a negligible impact on the hazard ratios of the other terms and were thus removed for greater clarity in the results. All analyses were performed using the “coxph” function from the R package “survival” (R Foundation for Statistical Computing) [21].", "This study does not constitute as human subjects research due to the use and reporting of only deidentified observational data as determined by the human subjects committee of the University of Washington and thus does not require the review and approval by the institutional review board at the University of Washington.", "Our study uses de-identified US medical claims records from Change Healthcare collected over a period from March 1, 2020, to October 14, 2021, encompassing over 100 million records from over 8 million patients. Medical claims data contain details about a patient’s interaction with the medical system, which are needed for the accurate billing of the transactions. Each claims record contains patient demographic information, International Classification of Diseases, 10th Revision (ICD-10) codes indicating primary diagnosis and secondary diagnoses, place of diagnosis, ICD-10 codes of procedures performed, patient status at the end of the visit, dates pertaining to the event (where different “from” and “to” dates indicate longer visits whereas the same “from” and “to” dates are for outpatient visits).\nOur claims data set includes primarily open claims and a subset of closed payer claims that are normalized for analytics purposes. The open claims are derived from broad-based health care sources and consist of all medical claims that Change Healthcare processes and for which it has the usage rights. The closed claims are derived directly from the payer (ie, health insurance provider) and capture nearly all events that occur during the patient’s enrollment period. Roughly 95% of the claims used for this study are commercial, and 5% are Medicare Advantage or other types of plans.", "Our data set of 8.18 million individuals contains only COVID-19 positive patients, defined as patients with at least one claims record with the ICD-10 diagnosis codes of “U07.1” or “U07.2” in any diagnosis field. We limited our analysis to individuals who had a primary diagnosis of “U07.1” (this is indicated by the principal diagnosis code, which encodes the primary diagnosis rendered by the medical facility or the primary cause of the visit). This ICD-10 diagnosis code indicates a COVID-19 diagnosis where the virus was identified in a lab-confirmed report. We exclude patients for whom the code of U07.1 appears in the “other diagnosis” fields, which contain the list of diagnoses made in addition to the primary cause of visit, which can be any other medical condition such as cancer. We also exclude patients with the code U07.2, which indicates a non-lab–confirmed COVID-19 diagnosis.\nSubsequently, fully vaccinated individuals are identified by looking for procedure codes encoding the second doses of Pfizer (0002A) and Moderna (0012A) vaccines and the first dose of the Janssen (0031A) vaccine. We do not exclude patients with missing first dose claims records (~5% of the final study cohort), because patients who went to vaccination camps and were not required to provide insurance information would have missing first dose claims records. Some of these ~5% patients with missing first dose claims records may have had mixed vaccines (eg, Pfizer for the first dose and Moderna for the second dose). Since we did not believe that this will be a significant fraction of the vaccinated population, we do not exclude them. Breakthrough patients were defined as those who had a primary, lab-confirmed COVID-19 diagnosis at least 14 days after the date of vaccination. Please see Figure S1 in Multimedia Appendix 1 for a flow diagram showing the criteria used for cohort selection.", "We explicitly identify hospitalization by looking for claims where the claim type is “institutional” or “professional” and the bill type indicates an “inpatient” facility. We also look at the dates associated with the hospital stay and only consider patients whose admission duration was at least 2 days (derived from the “admission_from” and “admission_to” date fields). For mortality, we look at the patient status code and consider all codes indicating “expired.” As described already, we only consider cases where the primary diagnosis was COVID-19 for both hospitalization and expiration. The patient status code is available for all hospitalized patients but only for 42% of the outpatients (who went to clinics). Among patients who had the patient status code available, we found only 17 (0.22%) deaths out of a total of 7843 outpatients; therefore, we consider outpatients with missing patient status to be alive.", "COVID-19 vaccinations began in the United States in late December 2020. By late February 2021, the Pfizer-BioNTech (Pfizer), Moderna, and Johnson & Johnson (J&J/Janssen) vaccines were all approved for emergency use authorization. The Pfizer and Moderna vaccination drives started much earlier, in late December (Figures S2 and S3 in Multimedia Appendix 1), as compared to that of the Janssen vaccines, which also saw a stall in vaccine rates in mid-April (Figure S4 in Multimedia Appendix 1). To keep the COVID-19 exposure of the individuals taking any of the 3 vaccines consistent, we use the same study window, though we have data for Pfizer and Moderna from late December. We construct our cohort to consist of individuals who were fully vaccinated between March 10, 2021, and April 27, 2021, the period during which all 3 vaccines were being widely administered. Every individual in this cohort was followed from the date of vaccination of each individual up to the end of the study period, October 14, 2021. The study period over the entire cohort is thus March 10, 2021, to October 14, 2021. In Figure S6 in Multimedia Appendix 1, we show statistics showing the number of days of follow-up after full vaccination by vaccine type in our study cohort.", "Preexisting comorbidities were defined based on ICD-10 codes assigned to medical encounters, which contain pointers to previously diagnosed conditions, using claims records during the 6-month period from March 2020 to September 2020. This period does not overlap with the study period, so events during the study period will not also be counted as comorbidities. The Elixhauser comorbidity index [20] was used to define comorbid conditions. This index has a series of codes that define comorbidities with each code mapping to one or several ICD-10 diagnosis codes. For example, the Elixhauser code “BLDLOSS” (blood loss) includes the following four ICD-10 diagnosis codes: D50.0, O90.81, O99.02, and O99.03. We provide the index that we used and the corresponding ICD-10 codes in Multimedia Appendix 2. We also show the relative abundance of comorbidities in our cohort, by vaccine type, in Table S2 and Figure S5 in Multimedia Appendix 1.", "Since some of the individuals in our cohort may have had a COVID-19 infection during the year 2020, we introduce an additional feature to encode the effect of already being infected with COVID-19. This feature is “yes” if we see a claim involving a COVID-19 diagnosis in any diagnosis field, in the period from March 1, 2020, to the beginning of the study period, March 10, 2021.", "Date of full vaccination was defined as 14 days after (1) a single Janssen vaccine, (2) the second Moderna vaccine dose, or (3) the second Pfizer vaccine dose. Cox proportional hazards regression was used to estimate univariate hazard ratios (HRs) and multivariable HRs in a model including the following features: age (categorized), sex (male and female), vaccine type, Elixhauser comorbidities (encoded as independent binary variables), and SARS-CoV-2 infection prior to the first dose of vaccination (yes or no). We remove the negligible number of individuals with sex=unknown. We also model interactions between vaccine type and all other covariates as well as previous infection and all other covariates but find that none were statistically significant. Further, the interaction terms had a negligible impact on the hazard ratios of the other terms and were thus removed for greater clarity in the results. All analyses were performed using the “coxph” function from the R package “survival” (R Foundation for Statistical Computing) [21].", "Our study includes 19,815 fully vaccinated patients with breakthrough SARS-CoV-2 infections between March 10, 2021, and October 14, 2021. Of those patients, 11,339 (57.22%) received the Pfizer vaccine, 5480 (27.66%) received the Moderna vaccine, and 2996 (15.12%) received the Janssen vaccine. Breakthrough cases receiving Janssen were younger than those receiving Pfizer or Moderna and had a slightly greater proportion of male patients (Table 1). Breakthrough cases receiving Moderna had a greater proportion of patients with COVID-19 prior to vaccination.\nRisk of hospitalization and mortality among breakthrough cases increased with older age and was higher for male patients (Table 2). In multivariable analyses controlling for age, male sex, comorbidities, and prior SARS-CoV-2 infection, the risk of hospitalization was the lowest for breakthrough cases receiving the Moderna vaccine (adjusted hazard ratio [aHR]: 0.42, 95% CI 0.35-0.5; P<.001), comparably low for Pfizer vaccinated individuals (aHR: 0.45, 95% CI 0.39-0.53; P<.001), compared with that for the recipients of the Janssen vaccine. The comorbidities with statistically significant HRs for hospitalization or mortality from a breakthrough SARS-CoV-2 infection include severe liver disease, moderate and severe renal failures, alcohol abuse, chronic lung disease, coagulopathy, cancers, anemia, seizures, and arthritis (Table S1 in Multimedia Appendix 1).\nCharacteristics of SARS-CoV-2 breakthrough infections cohort tracked from March 10, 2021, to October 14, 2021. Prevalence of comorbidities by vaccine type is shown in Figure S5 in Multimedia Appendix 1.\nCorrelates of hospitalization and mortality after breakthrough SARS-CoV-2 infection, estimated from Cox proportional hazards models. We show the adjusted hazard ratio (aHR) and the 95% CI for the significant correlates (P values indicated via superscripts d, e, and f). An aHR of <1.0 indicates a lower risk of hospitalization or mortality as compared to the baseline population for that covariate (analogously, aHR>1.0 indicates a higher risk than the baseline). Hazard ratios (HRs) of comorbidities are shown in Table S1 in Multimedia Appendix 1.\naIncidence per 100 person years.\nbn=19,815; events=1140.\ncn=19,815; events=159.\ndP<.001.\neP<.05.\nfP<.01.\nWe see a similar trend with the risk of mortality for breakthrough cases, with the risk being the lowest for those receiving the Moderna vaccines (aHR: 0.38, 95% CI 0.23-0.62; P<.001) and comparably lower for Pfizer recipients (aHR: 0.43, 95% CI 0.28-0.65; P<.001) as compared to that for Janssen recipients. Finally, as expected, the protection offered by vaccines was enhanced for breakthrough cases who already had a previous SARS-CoV-2 infection. These individuals were 40% less likely to be hospitalized due to COVID-19 (aHR: 0.57, 95% CI 0.41-0.80; P=.001) and four times less likely to die of COVID-19 (aHR: 0.22, 95% CI 0.05-0.91; P=.04), when compared to those without a prior SARS-CoV-2 infection independent of age, sex, comorbidities, and vaccine type.\nWe repeat this analysis by excluding the population who had a prior SARS-CoV-2 infection for completeness and show the resulting HRs Table S3 in Multimedia Appendix 1.", "[SUBTITLE] Principal Findings [SUBSECTION] Using medical claims data, we found that the risk of hospitalization in SARS-CoV-2 breakthrough infections was lower for those receiving the Moderna and Pfizer vaccines compared to those receiving the Janssen vaccine. The risk of mortality was similarly low in breakthrough infections who received Pfizer and Moderna vaccines compared to those receiving the Janssen vaccine. There was no statistically significant difference between the HRs of Pfizer and Moderna for both risks. We also found older age, male sex, and certain comorbidities to be risk factors for hospitalization and mortality in breakthrough infections. Further, we found that risk of hospitalization was 40% less and risk of death was 75% less in SARS-CoV-2 breakthrough infections among individuals who already had a SARS-CoV-2 infection prior to their vaccination compared with fully vaccinated individuals without a previous SARS-CoV-2 infection. While other studies have reported lower risk of breakthrough infection with previous SARS-CoV-2 infection [18], our study analyzes both hospitalization and mortality and shows that the immunity provided by previous infection seems to increase the protection provided by vaccines, against severe COVID-19, independent of vaccine type, age, comorbidities, and sex. Since our cohort only consists of individuals who were all fully vaccinated, this is by no means a comparison of vaccine-induced immunity against acquired immunity from previous infections.\nExcluding patients who had COVID-19 infection prior to vaccination from our Cox regression analysis results in a similar HR for hospitalization risk in patients who received the Pfizer (aHR=0.42) and Moderna (aHR=0.41) vaccines (Table S3 in Multimedia Appendix 1). This might be explained by the fact that 20.7% of patients who received Moderna had a prior COVID-19 infection as compared to ~13% of patients who received Pfizer. Hence, removing all patients with prior COVID-19 infection reduced the influence of the additional immunity that some of Moderna-vaccinated individuals had.\nA number of studies have found that age has a direct effect on the risk of severe COVID-19 disease [22,23]. We find that the proportion of the elderly cohort in our data set who were hospitalized is much higher than the proportion of the younger cohort (Figure S7 in multimedia Appendix 1). In addition, we find a higher HR for the elderly subset of our study cohort (aHR=2.1 for age>50, aHR=3.3 for age>65, and aHR=5.0 for age>80, as compared to the baseline age group of 35-50 years).\nOur findings comparing vaccine types are similar to those reported by the Centers for Disease Control and Prevention for mortality but provide additional information by vaccine type [1,16]. There have been several studies on individual risk factors such as age [22], specific comorbidities [7], focused populations such as Veterans [24], or large-scale projects such as OpenSAFELY, which involved 17 million unvaccinated patients [10]. Our work advances this body of literature by analyzing vaccine type, age, sex, and 39 different comorbidities in a large cohort of breakthrough patients from the general US population. Some of the risk factors that we find for severe breakthrough SARS-CoV-2 infections, such as age, male gender, and certain comorbidities (eg, chronic lung infection, kidney disease, and cancers) are similar to what has been reported in prior studies of SARS-CoV-2 infections among unvaccinated individuals [8,23,25]. However, we find that some risk factors found by initial studies such as hypertension are not a risk factor for breakthrough COVID-19 hospitalization or death (aHRs of 0.75 and 0.59, respectively), neither are diabetes or congestive heart failure. We instead find that both moderate and severe renal failure are significant risk factors, independent of age or other factors, which agrees with other large-scale studies such as OpenSAFELY [10] and the Global Burden of Disease collaboration [26] which identified that worldwide chronic kidney disease is the most prevalent risk factor for severe COVID-19. Even mild impairment of renal function has been found to be an independent risk factor for COVID-19 infection, hospitalization, and mortality [27].\nLastly, to understand the association between outcomes and the time of vaccination, we incorporate a variable indicating the number of days between full vaccination and the onset of the surge in infections caused by the delta variant. However, our population-based data set is inadequate to derive any significant conclusions vis-à-vis the best time for vaccination in anticipation of a surge.\nUsing medical claims data, we found that the risk of hospitalization in SARS-CoV-2 breakthrough infections was lower for those receiving the Moderna and Pfizer vaccines compared to those receiving the Janssen vaccine. The risk of mortality was similarly low in breakthrough infections who received Pfizer and Moderna vaccines compared to those receiving the Janssen vaccine. There was no statistically significant difference between the HRs of Pfizer and Moderna for both risks. We also found older age, male sex, and certain comorbidities to be risk factors for hospitalization and mortality in breakthrough infections. Further, we found that risk of hospitalization was 40% less and risk of death was 75% less in SARS-CoV-2 breakthrough infections among individuals who already had a SARS-CoV-2 infection prior to their vaccination compared with fully vaccinated individuals without a previous SARS-CoV-2 infection. While other studies have reported lower risk of breakthrough infection with previous SARS-CoV-2 infection [18], our study analyzes both hospitalization and mortality and shows that the immunity provided by previous infection seems to increase the protection provided by vaccines, against severe COVID-19, independent of vaccine type, age, comorbidities, and sex. Since our cohort only consists of individuals who were all fully vaccinated, this is by no means a comparison of vaccine-induced immunity against acquired immunity from previous infections.\nExcluding patients who had COVID-19 infection prior to vaccination from our Cox regression analysis results in a similar HR for hospitalization risk in patients who received the Pfizer (aHR=0.42) and Moderna (aHR=0.41) vaccines (Table S3 in Multimedia Appendix 1). This might be explained by the fact that 20.7% of patients who received Moderna had a prior COVID-19 infection as compared to ~13% of patients who received Pfizer. Hence, removing all patients with prior COVID-19 infection reduced the influence of the additional immunity that some of Moderna-vaccinated individuals had.\nA number of studies have found that age has a direct effect on the risk of severe COVID-19 disease [22,23]. We find that the proportion of the elderly cohort in our data set who were hospitalized is much higher than the proportion of the younger cohort (Figure S7 in multimedia Appendix 1). In addition, we find a higher HR for the elderly subset of our study cohort (aHR=2.1 for age>50, aHR=3.3 for age>65, and aHR=5.0 for age>80, as compared to the baseline age group of 35-50 years).\nOur findings comparing vaccine types are similar to those reported by the Centers for Disease Control and Prevention for mortality but provide additional information by vaccine type [1,16]. There have been several studies on individual risk factors such as age [22], specific comorbidities [7], focused populations such as Veterans [24], or large-scale projects such as OpenSAFELY, which involved 17 million unvaccinated patients [10]. Our work advances this body of literature by analyzing vaccine type, age, sex, and 39 different comorbidities in a large cohort of breakthrough patients from the general US population. Some of the risk factors that we find for severe breakthrough SARS-CoV-2 infections, such as age, male gender, and certain comorbidities (eg, chronic lung infection, kidney disease, and cancers) are similar to what has been reported in prior studies of SARS-CoV-2 infections among unvaccinated individuals [8,23,25]. However, we find that some risk factors found by initial studies such as hypertension are not a risk factor for breakthrough COVID-19 hospitalization or death (aHRs of 0.75 and 0.59, respectively), neither are diabetes or congestive heart failure. We instead find that both moderate and severe renal failure are significant risk factors, independent of age or other factors, which agrees with other large-scale studies such as OpenSAFELY [10] and the Global Burden of Disease collaboration [26] which identified that worldwide chronic kidney disease is the most prevalent risk factor for severe COVID-19. Even mild impairment of renal function has been found to be an independent risk factor for COVID-19 infection, hospitalization, and mortality [27].\nLastly, to understand the association between outcomes and the time of vaccination, we incorporate a variable indicating the number of days between full vaccination and the onset of the surge in infections caused by the delta variant. However, our population-based data set is inadequate to derive any significant conclusions vis-à-vis the best time for vaccination in anticipation of a surge.\n[SUBTITLE] Limitations [SUBSECTION] Limitations of our study include, first, a lack of access to data on unvaccinated individuals or those who had a negative SARS-CoV-2 test result. The former is due to the lack of a medical claims record for vaccinations that were done in vaccination drives and camps; the absence of a vaccination-related claim in our data set therefore does not imply an unvaccinated individual. Second, our medical claims source consists of mostly privately insured individuals and can thus miss people who may be susceptible to the most adverse outcomes. Another caveat of our data set is that most of the claims are open claims, which have the benefit of capturing a patient’s activities over a longer time frame regardless of their insurance provider, but do not necessarily track all medical encounters of patients.\nLimitations of our study include, first, a lack of access to data on unvaccinated individuals or those who had a negative SARS-CoV-2 test result. The former is due to the lack of a medical claims record for vaccinations that were done in vaccination drives and camps; the absence of a vaccination-related claim in our data set therefore does not imply an unvaccinated individual. Second, our medical claims source consists of mostly privately insured individuals and can thus miss people who may be susceptible to the most adverse outcomes. Another caveat of our data set is that most of the claims are open claims, which have the benefit of capturing a patient’s activities over a longer time frame regardless of their insurance provider, but do not necessarily track all medical encounters of patients.\n[SUBTITLE] Conclusions [SUBSECTION] Our findings add to the growing literature regarding the risk factors for severe breakthrough SARS-CoV-2 infections in fully vaccinated individuals, where we identify the influence of age, sex, and comorbidities that are risk factors; importantly, we found that previous SARS-CoV-2 infections can provide additional protection over that offered by vaccines against severe disease. Our results also necessitate further studies on the optimal number of vaccine doses to protect from the most severe breakthrough SARS-CoV-2 infections. An important strength of our study is that we consider US-wide breakthrough hospitalizations covering a broad demographic and compare all 3 vaccines, whereas most previous studies lack specific data on Janssen.\nOur findings add to the growing literature regarding the risk factors for severe breakthrough SARS-CoV-2 infections in fully vaccinated individuals, where we identify the influence of age, sex, and comorbidities that are risk factors; importantly, we found that previous SARS-CoV-2 infections can provide additional protection over that offered by vaccines against severe disease. Our results also necessitate further studies on the optimal number of vaccine doses to protect from the most severe breakthrough SARS-CoV-2 infections. An important strength of our study is that we consider US-wide breakthrough hospitalizations covering a broad demographic and compare all 3 vaccines, whereas most previous studies lack specific data on Janssen.", "Using medical claims data, we found that the risk of hospitalization in SARS-CoV-2 breakthrough infections was lower for those receiving the Moderna and Pfizer vaccines compared to those receiving the Janssen vaccine. The risk of mortality was similarly low in breakthrough infections who received Pfizer and Moderna vaccines compared to those receiving the Janssen vaccine. There was no statistically significant difference between the HRs of Pfizer and Moderna for both risks. We also found older age, male sex, and certain comorbidities to be risk factors for hospitalization and mortality in breakthrough infections. Further, we found that risk of hospitalization was 40% less and risk of death was 75% less in SARS-CoV-2 breakthrough infections among individuals who already had a SARS-CoV-2 infection prior to their vaccination compared with fully vaccinated individuals without a previous SARS-CoV-2 infection. While other studies have reported lower risk of breakthrough infection with previous SARS-CoV-2 infection [18], our study analyzes both hospitalization and mortality and shows that the immunity provided by previous infection seems to increase the protection provided by vaccines, against severe COVID-19, independent of vaccine type, age, comorbidities, and sex. Since our cohort only consists of individuals who were all fully vaccinated, this is by no means a comparison of vaccine-induced immunity against acquired immunity from previous infections.\nExcluding patients who had COVID-19 infection prior to vaccination from our Cox regression analysis results in a similar HR for hospitalization risk in patients who received the Pfizer (aHR=0.42) and Moderna (aHR=0.41) vaccines (Table S3 in Multimedia Appendix 1). This might be explained by the fact that 20.7% of patients who received Moderna had a prior COVID-19 infection as compared to ~13% of patients who received Pfizer. Hence, removing all patients with prior COVID-19 infection reduced the influence of the additional immunity that some of Moderna-vaccinated individuals had.\nA number of studies have found that age has a direct effect on the risk of severe COVID-19 disease [22,23]. We find that the proportion of the elderly cohort in our data set who were hospitalized is much higher than the proportion of the younger cohort (Figure S7 in multimedia Appendix 1). In addition, we find a higher HR for the elderly subset of our study cohort (aHR=2.1 for age>50, aHR=3.3 for age>65, and aHR=5.0 for age>80, as compared to the baseline age group of 35-50 years).\nOur findings comparing vaccine types are similar to those reported by the Centers for Disease Control and Prevention for mortality but provide additional information by vaccine type [1,16]. There have been several studies on individual risk factors such as age [22], specific comorbidities [7], focused populations such as Veterans [24], or large-scale projects such as OpenSAFELY, which involved 17 million unvaccinated patients [10]. Our work advances this body of literature by analyzing vaccine type, age, sex, and 39 different comorbidities in a large cohort of breakthrough patients from the general US population. Some of the risk factors that we find for severe breakthrough SARS-CoV-2 infections, such as age, male gender, and certain comorbidities (eg, chronic lung infection, kidney disease, and cancers) are similar to what has been reported in prior studies of SARS-CoV-2 infections among unvaccinated individuals [8,23,25]. However, we find that some risk factors found by initial studies such as hypertension are not a risk factor for breakthrough COVID-19 hospitalization or death (aHRs of 0.75 and 0.59, respectively), neither are diabetes or congestive heart failure. We instead find that both moderate and severe renal failure are significant risk factors, independent of age or other factors, which agrees with other large-scale studies such as OpenSAFELY [10] and the Global Burden of Disease collaboration [26] which identified that worldwide chronic kidney disease is the most prevalent risk factor for severe COVID-19. Even mild impairment of renal function has been found to be an independent risk factor for COVID-19 infection, hospitalization, and mortality [27].\nLastly, to understand the association between outcomes and the time of vaccination, we incorporate a variable indicating the number of days between full vaccination and the onset of the surge in infections caused by the delta variant. However, our population-based data set is inadequate to derive any significant conclusions vis-à-vis the best time for vaccination in anticipation of a surge.", "Limitations of our study include, first, a lack of access to data on unvaccinated individuals or those who had a negative SARS-CoV-2 test result. The former is due to the lack of a medical claims record for vaccinations that were done in vaccination drives and camps; the absence of a vaccination-related claim in our data set therefore does not imply an unvaccinated individual. Second, our medical claims source consists of mostly privately insured individuals and can thus miss people who may be susceptible to the most adverse outcomes. Another caveat of our data set is that most of the claims are open claims, which have the benefit of capturing a patient’s activities over a longer time frame regardless of their insurance provider, but do not necessarily track all medical encounters of patients.", "Our findings add to the growing literature regarding the risk factors for severe breakthrough SARS-CoV-2 infections in fully vaccinated individuals, where we identify the influence of age, sex, and comorbidities that are risk factors; importantly, we found that previous SARS-CoV-2 infections can provide additional protection over that offered by vaccines against severe disease. Our results also necessitate further studies on the optimal number of vaccine doses to protect from the most severe breakthrough SARS-CoV-2 infections. An important strength of our study is that we consider US-wide breakthrough hospitalizations covering a broad demographic and compare all 3 vaccines, whereas most previous studies lack specific data on Janssen." ]
[ "introduction", "methods", null, null, null, null, null, null, null, null, "results", "discussion", null, null, null ]
[ "breakthroughs", "vaccines", "Pfizer", "Moderna", "Janssen", "SARS-CoV-2", "COVID-19", "coronavirus", "infectious disease", "viral infection", "vaccination", "breakthrough infection", "public health", "health policy", "decision making", "booster vaccine", "mortality", "hospitalization", "healthcare system" ]
Outcomes of COVID-19 Infection in People Previously Vaccinated Against Influenza: Population-Based Cohort Study Using Primary Health Care Electronic Records.
36265160
A possible link between influenza immunization and susceptibility to the complications of COVID-19 infection has been previously suggested owing to a boost in the immunity against SARS-CoV-2.
BACKGROUND
We performed a population-based cohort study including all patients with COVID-19 with registered entries in the primary health care (PHC) electronic records during the first wave of the COVID-19 pandemic (March 1 to June 30, 2020) in Catalonia, Spain. We compared individuals who took an influenza vaccine before being infected with COVID-19, with those who had not taken one. Data were obtained from Information System for Research in Primary Care, capturing PHC information of 5.8 million people from Catalonia. The main outcomes assessed during follow-up were a diagnosis of pneumonia, hospital admission, and mortality.
METHODS
We included 309,039 individuals with COVID-19 and compared them on the basis of their influenza immunization status, with 114,181 (36.9%) having been vaccinated at least once and 194,858 (63.1%) having never been vaccinated. In total, 21,721 (19%) vaccinated individuals and 11,000 (5.7%) unvaccinated individuals had at least one of their outcomes assessed. Those vaccinated against influenza at any time (odds ratio [OR] 1.14, 95% CI 1.10-1.19), recently (OR 1.13, 95% CI 1.10-1.18), or recurrently (OR 1.10, 95% CI 1.05-1.15) before being infected with COVID-19 had a higher risk of presenting at least one of the outcomes than did unvaccinated individuals. When we excluded people living in long-term care facilities, the results were similar.
RESULTS
We could not establish a protective role of the immunity conferred by the influenza vaccine on the outcomes of COVID-19 infection, as the risk of COVID-19 complications was higher in vaccinated than in unvaccinated individuals. Our results correspond to the first wave of the COVID-19 pandemic, where more complications and mortalities due to COVID-19 had occurred. Despite that, our study adds more evidence for the analysis of a possible link between the quality of immunity and COVID-19 outcomes, particularly in the PHC setting.
CONCLUSIONS
[ "Humans", "Influenza Vaccines", "Influenza, Human", "COVID-19", "Pandemics", "SARS-CoV-2", "Cohort Studies", "Primary Health Care", "Electronics" ]
9662290
Introduction
COVID-19 is caused by SARS-CoV-2, a novel coronavirus that emerged in China in 2019, which became the primary agent of a new pandemic that rapidly spread worldwide [1], with an average global infection fatality rate of approximately 0.15%, depending on the data analyzed [2]. SARS-CoV-2 mainly affects the respiratory tract and uses surface proteins in order to infect the host [3]. Although new variants of SARS-CoV-2 have emerged since December 2020, the coronavirus’ genome is composed of RNA and depends on the RNA polymerase to generate its proteins, with a mechanism of error correction that results in a lower mutation rate than the influenza virus [4]. This low mutation rate may suggest that the vaccines developed against SARS-CoV-2, as well as the immunity generated in those patients who were infected, could represent a long-lasting immunity [5,6]. COVID-19, similar to influenza A and B, is caused by RNA virus and produce similar symptoms. The influenza virus needs the hemagglutinin and neuraminidase surface proteins to infect cells, whereas SARS-CoV-2 needs the S protein [5]. Previous in vitro and animal studies suggest an induction pathway of indirect etiological immunity between the influenza vaccine and SARS-CoV-2. Animal models suggest that some influenza subtypes might lead to regulation of the angiotensin-converting enzyme-2, with protective properties against SARS [7]. An unspecific effect of infection and vaccination on the immune system and susceptibility to other infections has also been reported, albeit with discordant data [8-10]. Some modeling studies have suggested a possible association between influenza immunization and COVID-19 [11-14]. A study conducted in Australia assessed the cellular and humoral immune responses during and after disease occurrence in a patient with a mild COVID-19 infection. They found that the immune response in different cell types is associated with clinical recovery. These results are coincident with similar findings among patients with influenza reported by the same authors [15,16]. Other studies observed differences in the susceptibility to COVID-19 in children of different ages with a lower infection rate than that in adults and older individuals [17]. Although the mechanism underlying these differences in severity and susceptibility is unclear, a possible explanation might be the difference in the quantity and quality of the immune function determined by the history of infections and the recent vaccines administered [18]. Consequently, a link between the quality of the immunity and recovery from COVID-19 may exist. Thus, we hypothesized that the immunity resulting from the previous influenza vaccination would boost part of the immunity against SARS-CoV-2, and we aimed to investigate whether individuals with COVID-19 could have benefited from vaccination against influenza.
Methods
[SUBTITLE] Study Design [SUBSECTION] We performed a population-based cohort study including all adults with COVID-19 in Catalonia, Spain, who were registered as confirmed cases (through the polymerase chain reaction [PCR]) or as probable cases (not confirmed through PCR but with International Classification of Diseases (ICD)-10 codes registered that are compatible with COVID-19) in the primary health care (PHC) system. All individuals with COVID-19 were diagnosed from the pandemic’s onset (March 2020) to June 30, 2020. Participants were compared on the basis of their influenza vaccination status between those having received the influenza vaccine before having COVID-19 (vaccinated in the previous influenza seasonal campaign in 2019-2020 or before) [19] with those who were not vaccinated. We performed a population-based cohort study including all adults with COVID-19 in Catalonia, Spain, who were registered as confirmed cases (through the polymerase chain reaction [PCR]) or as probable cases (not confirmed through PCR but with International Classification of Diseases (ICD)-10 codes registered that are compatible with COVID-19) in the primary health care (PHC) system. All individuals with COVID-19 were diagnosed from the pandemic’s onset (March 2020) to June 30, 2020. Participants were compared on the basis of their influenza vaccination status between those having received the influenza vaccine before having COVID-19 (vaccinated in the previous influenza seasonal campaign in 2019-2020 or before) [19] with those who were not vaccinated. [SUBTITLE] Data Source [SUBSECTION] Our data source is the Information System for Research in Primary Care [20], which captures clinical information of approximately 5.8 million people from Catalonia, Spain (approximately 80% of the Catalan population). This information is pseudonymized, having originated from different data sources: (1) electronic health records in PHC system of the Catalan Health Institute, including sociodemographic characteristics, residents in nursing homes or long-term care facilities (LTCFs), comorbidities registered as ICD-10 codes [21], specialist referrals, clinical parameters, toxic habits, sickness leave, date of death, laboratory test data, and drug prescriptions issued in the PHC system, registered in accordance with the anatomical therapeutic chemical classification system [22]; (2) pharmacy invoice data corresponding to the PHC drug prescriptions; (3) database of diagnoses upon hospital discharge [23]; and (4) COVID-19 data from the Catalan Agency of Health Quality and Evaluation (AQuAS) [24]. Our data source is the Information System for Research in Primary Care [20], which captures clinical information of approximately 5.8 million people from Catalonia, Spain (approximately 80% of the Catalan population). This information is pseudonymized, having originated from different data sources: (1) electronic health records in PHC system of the Catalan Health Institute, including sociodemographic characteristics, residents in nursing homes or long-term care facilities (LTCFs), comorbidities registered as ICD-10 codes [21], specialist referrals, clinical parameters, toxic habits, sickness leave, date of death, laboratory test data, and drug prescriptions issued in the PHC system, registered in accordance with the anatomical therapeutic chemical classification system [22]; (2) pharmacy invoice data corresponding to the PHC drug prescriptions; (3) database of diagnoses upon hospital discharge [23]; and (4) COVID-19 data from the Catalan Agency of Health Quality and Evaluation (AQuAS) [24]. [SUBTITLE] COVID-19 Classification [SUBSECTION] Participants were classified in accordance with the following criteria: confirmed cases are those with a confirmed COVID-19 diagnosis record, positive PCR outcome, or a positive serology test result. Those with an unconfirmed diagnosis or test (possible or unclear) along with any individual with a record of hospitalization, pneumonia, or death related to COVID-19 were considered probable cases. During the first wave of the COVID-19 pandemic in Catalonia, PCR tests were not routinely conducted for all individuals with compatible symptoms owing to the unavailability of laboratory kits to carry out the tests. Thus, we needed to capture those patients with a possible diagnosis of COVID-19, such as those admitted to hospital with pneumonia or other COVID-19 symptoms, who were not tested. We designed an algorithm to classify patients as “COVID possible” when a test result was unavailable along with registered entries from different databases: PCR tests or serology tests conducted in different settings, discharge diagnoses of pneumonia from Catalan hospitals or from emergency departments, and ICD-10 diagnoses related to COVID-19 coded in PHC. The date of COVID-19 diagnosis was set to be the first of all records used per patient. To guarantee that our algorithm is not far from the Catalan population, the resulting cohort was compared to the official COVID-19 cases reported by the AQuAS during the pandemic [24]. Participants were classified in accordance with the following criteria: confirmed cases are those with a confirmed COVID-19 diagnosis record, positive PCR outcome, or a positive serology test result. Those with an unconfirmed diagnosis or test (possible or unclear) along with any individual with a record of hospitalization, pneumonia, or death related to COVID-19 were considered probable cases. During the first wave of the COVID-19 pandemic in Catalonia, PCR tests were not routinely conducted for all individuals with compatible symptoms owing to the unavailability of laboratory kits to carry out the tests. Thus, we needed to capture those patients with a possible diagnosis of COVID-19, such as those admitted to hospital with pneumonia or other COVID-19 symptoms, who were not tested. We designed an algorithm to classify patients as “COVID possible” when a test result was unavailable along with registered entries from different databases: PCR tests or serology tests conducted in different settings, discharge diagnoses of pneumonia from Catalan hospitals or from emergency departments, and ICD-10 diagnoses related to COVID-19 coded in PHC. The date of COVID-19 diagnosis was set to be the first of all records used per patient. To guarantee that our algorithm is not far from the Catalan population, the resulting cohort was compared to the official COVID-19 cases reported by the AQuAS during the pandemic [24]. [SUBTITLE] Influenza Immunization [SUBSECTION] Patients were classified as having taken the influenza vaccine if they had been vaccinated at any time before having COVID-19, and grouped in accordance with the seasonal vaccination campaign: the immediate previous campaign (2019-2020) or other vaccination campaigns (2018-2019 and before) [19,25]. Patients were classified as having taken the influenza vaccine if they had been vaccinated at any time before having COVID-19, and grouped in accordance with the seasonal vaccination campaign: the immediate previous campaign (2019-2020) or other vaccination campaigns (2018-2019 and before) [19,25]. [SUBTITLE] Variables [SUBSECTION] At baseline, the following variables were captured: sex, age, geographical area, MEDEA (Mortalidad en áreas pequeñas Españolas y Desigualdades Económicas y Ambientales [Mortality in small Spanish areas and economic and environmental inequalities]) socioeconomic index (deprivation index based on 5 indicators of socioeconomic position; it helps analyze health inequity, and higher the MEDEA socioeconomic index, worse the deprivation) [26], BMI, residence in nursing homes, smoking habits, comorbidities, and taking influenza vaccines and pneumococcal and tuberculosis vaccines. The main outcomes assessed during follow-up (up to June 2020) were at least one of the following variables: diagnosis of pneumonia, hospital admission, and mortality. The risk of these events was analyzed in those people who had been vaccinated against influenza at any time before having COVID-19, in those who were recently vaccinated (campaign of 2019-2020), and in those systematically vaccinated (who had been vaccinated at least during 3 different campaigns). We analyzed the same outcomes excluding those of people living in LTCFs, where vaccination is nearly universal in our country [27]. At baseline, the following variables were captured: sex, age, geographical area, MEDEA (Mortalidad en áreas pequeñas Españolas y Desigualdades Económicas y Ambientales [Mortality in small Spanish areas and economic and environmental inequalities]) socioeconomic index (deprivation index based on 5 indicators of socioeconomic position; it helps analyze health inequity, and higher the MEDEA socioeconomic index, worse the deprivation) [26], BMI, residence in nursing homes, smoking habits, comorbidities, and taking influenza vaccines and pneumococcal and tuberculosis vaccines. The main outcomes assessed during follow-up (up to June 2020) were at least one of the following variables: diagnosis of pneumonia, hospital admission, and mortality. The risk of these events was analyzed in those people who had been vaccinated against influenza at any time before having COVID-19, in those who were recently vaccinated (campaign of 2019-2020), and in those systematically vaccinated (who had been vaccinated at least during 3 different campaigns). We analyzed the same outcomes excluding those of people living in LTCFs, where vaccination is nearly universal in our country [27]. [SUBTITLE] Statistical Analysis [SUBSECTION] Quantitative variables were described as mean (SD) values, whereas categorical variables were described as the proportion of vaccinated and unvaccinated individuals. Univariate analyses were based on the Student t test or chi-square test depending on the variable. For each outcome, we fitted a logistic regression model to estimate an odds ratio (OR) comparing the prevalence of each outcome among individuals given the influenza vaccine to that of unvaccinated individuals. The logistic model was fitted along with other covariables such as smoking habits, age, comorbidities (asthma, autoimmune disorders, prior cerebrovascular disease, chronic kidney disease, chronic pulmonary obstructive disease, diabetes, heart failure, hypertension, ischemic heart disease, mental-behavioral disorders, obesity, organ transplant, and other respiratory diseases), concomitant drugs, and previous vaccines (pneumococcal and tuberculosis). As a sensitivity analysis, we conducted the same analysis on a matched population. Individuals vaccinated against influenza and unvaccinated controls were matched 1:2 in accordance with their age and gender at the time of infection or on an index date, and the reported ORs were obtained by fitting a conditional logistic regression model (clogit) accounting for matched pairs and adjusted using the same covariables as in the logistic model. We used the Wald test on the fitted coefficient to determine whether the log-odds were significantly different from 0 at a threshold of .05. All analyses were performed in R (version 4.1.0 or above; The R Foundation). Quantitative variables were described as mean (SD) values, whereas categorical variables were described as the proportion of vaccinated and unvaccinated individuals. Univariate analyses were based on the Student t test or chi-square test depending on the variable. For each outcome, we fitted a logistic regression model to estimate an odds ratio (OR) comparing the prevalence of each outcome among individuals given the influenza vaccine to that of unvaccinated individuals. The logistic model was fitted along with other covariables such as smoking habits, age, comorbidities (asthma, autoimmune disorders, prior cerebrovascular disease, chronic kidney disease, chronic pulmonary obstructive disease, diabetes, heart failure, hypertension, ischemic heart disease, mental-behavioral disorders, obesity, organ transplant, and other respiratory diseases), concomitant drugs, and previous vaccines (pneumococcal and tuberculosis). As a sensitivity analysis, we conducted the same analysis on a matched population. Individuals vaccinated against influenza and unvaccinated controls were matched 1:2 in accordance with their age and gender at the time of infection or on an index date, and the reported ORs were obtained by fitting a conditional logistic regression model (clogit) accounting for matched pairs and adjusted using the same covariables as in the logistic model. We used the Wald test on the fitted coefficient to determine whether the log-odds were significantly different from 0 at a threshold of .05. All analyses were performed in R (version 4.1.0 or above; The R Foundation). [SUBTITLE] Ethical Considerations [SUBSECTION] The study protocol was approved by the Research Ethics Committee of Institut Universitari d’Investigació en Atenció Primària (June 3, 2020). This is a database research study that has been conducted in accordance with the tenets of the Declaration of Helsinki (Fortaleza, Brazil 2013) and does not require consent from the study participants for the purpose of publication. The need for consent was waived by the Research Ethics Committee of Institut Universitari d’Investigació en Atenció Primària as it is deemed unnecessary according to the European legislation (Regulation [EU] 2016/679). The study protocol was approved by the Research Ethics Committee of Institut Universitari d’Investigació en Atenció Primària (June 3, 2020). This is a database research study that has been conducted in accordance with the tenets of the Declaration of Helsinki (Fortaleza, Brazil 2013) and does not require consent from the study participants for the purpose of publication. The need for consent was waived by the Research Ethics Committee of Institut Universitari d’Investigació en Atenció Primària as it is deemed unnecessary according to the European legislation (Regulation [EU] 2016/679).
Results
We included 309,039 individuals with COVID-19 during the first wave of the pandemic in accordance with their influenza immunization status (Table 1, Multimedia Appendix 1); 114,181 (36.9%) participants had received the influenza vaccine at least once before having COVID-19 and 194,858 (63.1%) had not been vaccinated, with more women in both groups, especially in the vaccinated cohort (61.0% women vs 39.0% men). The mean age was higher for vaccinated individuals (64.3 years, with 52.3% of them being older than 65 years). Vaccinated individuals had more comorbidities than unvaccinated individuals. Of those who received the influenza vaccine, 66,611 (58.3%) had been recently vaccinated (2019-2020) and 75,311 (66%) had been systematically vaccinated against influenza at least during 3 different years (Table 2). Of the participants with COVID-19, 11,000 (5.7%) unvaccinated and 21,721 (19%) vaccinated participants presented at least one of the following events: hospital admission, pneumonia, or death. For those who received the influenza vaccine at any time before having COVID-19, the risks of hospitalization (adjusted OR 1.14, 95% CI 1.10-1.19) and death (OR 1.32, 1.23-1.42) were higher than those among unvaccinated participants. For the recently vaccinated participants, the risk was higher for hospitalization (OR 1.16, 95% CI 1.1-1.23), pneumonia (OR 1.12, 95% CI 1.02-1.23), and death (OR 1.14, 95% CI 1.04-1.24). For people with recurrent vaccination, the risk of the 3 outcomes was also higher that among unvaccinated participants (OR 1.07, 1.16, and 1.24, respectively; Table 3). We have also analyzed the results in a matched population of vaccinated versus unvaccinated participants, revealing a higher risk of pneumonia and mortality, with an adjusted OR of 1.11 (95% CI 1.01-1.23) and 1.28 (95% CI 1.07-1.53), respectively (Multimedia Appendix 2). The risks of the outcomes based on influenza vaccination status and excluding those patients living in LTCFs are shown in Figure 1. For non-LTCF residents, the results are similar to those for the whole population, except that there was no significant increase in mortality (OR 0.93, 95% CI 0.85-1.03). Sociodemographic and clinical characteristics of the study population (N=309,039). Taking influenza vaccines before having COVID-19. Logistic regression model of COVID-19 outcomes based on influenza immunization status. aA logistic regression model adjusted with the following relevant covariables was fitted: smoking habits, age, comorbidities (asthma, autoimmune disorders, prior cerebrovascular disease, chronic kidney disease, chronic pulmonary obstructive disease, diabetes, heart failure, hypertension, ischemic heart disease, mental-behavioral disorders, obesity, organ transplant, and other respiratory diseases), co-medication, and previous vaccines (pneumococcal and tuberculosis). Risk of death and of combined COVID-19 complications in all the vaccinated population and excluding people living in long-term care facilities (LTCF).
null
null
[ "Study Design", "Data Source", "COVID-19 Classification", "Influenza Immunization", "Variables", "Statistical Analysis", "Ethical Considerations", "Principal Findings", "Comparison With Prior Work", "Limitations", "Conclusions" ]
[ "We performed a population-based cohort study including all adults with COVID-19 in Catalonia, Spain, who were registered as confirmed cases (through the polymerase chain reaction [PCR]) or as probable cases (not confirmed through PCR but with International Classification of Diseases (ICD)-10 codes registered that are compatible with COVID-19) in the primary health care (PHC) system. All individuals with COVID-19 were diagnosed from the pandemic’s onset (March 2020) to June 30, 2020. Participants were compared on the basis of their influenza vaccination status between those having received the influenza vaccine before having COVID-19 (vaccinated in the previous influenza seasonal campaign in 2019-2020 or before) [19] with those who were not vaccinated.", "Our data source is the Information System for Research in Primary Care [20], which captures clinical information of approximately 5.8 million people from Catalonia, Spain (approximately 80% of the Catalan population). This information is pseudonymized, having originated from different data sources: (1) electronic health records in PHC system of the Catalan Health Institute, including sociodemographic characteristics, residents in nursing homes or long-term care facilities (LTCFs), comorbidities registered as ICD-10 codes [21], specialist referrals, clinical parameters, toxic habits, sickness leave, date of death, laboratory test data, and drug prescriptions issued in the PHC system, registered in accordance with the anatomical therapeutic chemical classification system [22]; (2) pharmacy invoice data corresponding to the PHC drug prescriptions; (3) database of diagnoses upon hospital discharge [23]; and (4) COVID-19 data from the Catalan Agency of Health Quality and Evaluation (AQuAS) [24].", "Participants were classified in accordance with the following criteria: confirmed cases are those with a confirmed COVID-19 diagnosis record, positive PCR outcome, or a positive serology test result. Those with an unconfirmed diagnosis or test (possible or unclear) along with any individual with a record of hospitalization, pneumonia, or death related to COVID-19 were considered probable cases. During the first wave of the COVID-19 pandemic in Catalonia, PCR tests were not routinely conducted for all individuals with compatible symptoms owing to the unavailability of laboratory kits to carry out the tests. Thus, we needed to capture those patients with a possible diagnosis of COVID-19, such as those admitted to hospital with pneumonia or other COVID-19 symptoms, who were not tested. We designed an algorithm to classify patients as “COVID possible” when a test result was unavailable along with registered entries from different databases: PCR tests or serology tests conducted in different settings, discharge diagnoses of pneumonia from Catalan hospitals or from emergency departments, and ICD-10 diagnoses related to COVID-19 coded in PHC. The date of COVID-19 diagnosis was set to be the first of all records used per patient. To guarantee that our algorithm is not far from the Catalan population, the resulting cohort was compared to the official COVID-19 cases reported by the AQuAS during the pandemic [24].", "Patients were classified as having taken the influenza vaccine if they had been vaccinated at any time before having COVID-19, and grouped in accordance with the seasonal vaccination campaign: the immediate previous campaign (2019-2020) or other vaccination campaigns (2018-2019 and before) [19,25].", "At baseline, the following variables were captured: sex, age, geographical area, MEDEA (Mortalidad en áreas pequeñas Españolas y Desigualdades Económicas y Ambientales [Mortality in small Spanish areas and economic and environmental inequalities]) socioeconomic index (deprivation index based on 5 indicators of socioeconomic position; it helps analyze health inequity, and higher the MEDEA socioeconomic index, worse the deprivation) [26], BMI, residence in nursing homes, smoking habits, comorbidities, and taking influenza vaccines and pneumococcal and tuberculosis vaccines.\nThe main outcomes assessed during follow-up (up to June 2020) were at least one of the following variables: diagnosis of pneumonia, hospital admission, and mortality. The risk of these events was analyzed in those people who had been vaccinated against influenza at any time before having COVID-19, in those who were recently vaccinated (campaign of 2019-2020), and in those systematically vaccinated (who had been vaccinated at least during 3 different campaigns). We analyzed the same outcomes excluding those of people living in LTCFs, where vaccination is nearly universal in our country [27].", "Quantitative variables were described as mean (SD) values, whereas categorical variables were described as the proportion of vaccinated and unvaccinated individuals. Univariate analyses were based on the Student t test or chi-square test depending on the variable.\nFor each outcome, we fitted a logistic regression model to estimate an odds ratio (OR) comparing the prevalence of each outcome among individuals given the influenza vaccine to that of unvaccinated individuals. The logistic model was fitted along with other covariables such as smoking habits, age, comorbidities (asthma, autoimmune disorders, prior cerebrovascular disease, chronic kidney disease, chronic pulmonary obstructive disease, diabetes, heart failure, hypertension, ischemic heart disease, mental-behavioral disorders, obesity, organ transplant, and other respiratory diseases), concomitant drugs, and previous vaccines (pneumococcal and tuberculosis). As a sensitivity analysis, we conducted the same analysis on a matched population. Individuals vaccinated against influenza and unvaccinated controls were matched 1:2 in accordance with their age and gender at the time of infection or on an index date, and the reported ORs were obtained by fitting a conditional logistic regression model (clogit) accounting for matched pairs and adjusted using the same covariables as in the logistic model. We used the Wald test on the fitted coefficient to determine whether the log-odds were significantly different from 0 at a threshold of .05. All analyses were performed in R (version 4.1.0 or above; The R Foundation).", "The study protocol was approved by the Research Ethics Committee of Institut Universitari d’Investigació en Atenció Primària (June 3, 2020). This is a database research study that has been conducted in accordance with the tenets of the Declaration of Helsinki (Fortaleza, Brazil 2013) and does not require consent from the study participants for the purpose of publication. The need for consent was waived by the Research Ethics Committee of Institut Universitari d’Investigació en Atenció Primària as it is deemed unnecessary according to the European legislation (Regulation [EU] 2016/679).", "We analyzed the negative outcomes among people with COVID-19 (N=309,039) and compared those who had received the influenza vaccine with those who were never vaccinated. Those who received the vaccine any time before having COVID-19 were at a higher risk of complications than those who were unvaccinated. We obtained similar results for those who were recently vaccinated (2019-2020 campaign) and for those who were systematically vaccinated (at least 3 years), and the same comparisons were carried out after excluding individuals living in LTCFs. We also obtained similar results on matching vaccinated and unvaccinated individuals. Thus, we did not find a possible link between receiving the influenza vaccine and presenting better clinical outcomes after a COVID-19 infection.", "Some researchers have studied this possible association. Massoudi and Mohit [28] conducted a study in a hospital in Iran including health care workers, with 80 of them COVID-19 cases confirmed through PCR or on the basis of their symptoms, and 181 of them were controls. They concluded that individuals who were confirmed cases were less likely to have received the 2019 influenza vaccine (OR 0.04, 95% CI 0.01-0.14), suggesting a protective association between the influenza vaccine and COVID-19. Their study had several limitations, such as the lack of availability of COVID-19 test kits or the samples limited to the workers of a single hospital [28].\nCandelli et al [29] assessed 602 patients with COVID-19 enrolled at the emergency department in a hospital in Italy, of whom 24.9% had been previously vaccinated against influenza. They found that influenza immunization was independently associated with a lower risk of death at 60 days (OR 0.20, 95% CI 0.08-0.51), but not with a reduced need of endotracheal intubation (OR 0.73, 95% CI 0.35-1.56) [29].\nA study conducted in Brazil [30] included 92,664 confirmed cases of COVID-19, of whom 31.1% had been recently vaccinated against influenza. They found that the vaccinated individuals were at a lower risk of needing intensive care for COVID-19 (OR 0.92, 95% CI 0.86-0.99), a lower risk of needing respiratory support (OR 0.81, 95% CI 0.74-0.88), and lower odds of mortality (OR 0.82, 95% CI 0.75-0.89) [30].\nIn a systematic review [31] including 12 studies, the authors examined whether influenza vaccination affects the risk of being infected with SARS-CoV-2 and the risk of complicated illness or poor outcomes among patients with COVID-19, all of whom having been confirmed cases through PCR testing. They concluded that influenza vaccination is unlikely to be associated with an increase in the risk of COVID-19 infection or severity and the risk of associated death [31].\nThere are reports from some countries with high influenza vaccination rates and high incidences of COVID-19 and mortality [32,33]. For instance, Kline et al [33] compared people vaccinated against influenza with unvaccinated individuals admitted to hospital for COVID-19, and they found no differences in the rate of admission to the intensive care unit, intubation, or other complications [33]. Our results follow these same trends in a cohort of the general population attended to in the PHC system and not only hospitalized patients.", "We need to consider that our results correspond to the first wave of the COVID-19 pandemic, when there were more negative outcomes and mortalities due to COVID-19 than in the subsequent waves in our setting; thus, this higher statistical power allowed us to detect differences. Furthermore, in subsequent waves, more confounders might have been present, such as COVID-19 vaccination or effects of the different SARS-CoV-2 variants, making it more difficult to manage their potential effect in the analysis of the outcomes of the infection.\nWe also need to bear in mind that the target population for the influenza vaccine in our country are people older than 60 years, individuals with chronic comorbidities or immunodeficiency, and health care workers among others [34], some of them being at a high risk of COVID-19 complications, which is why confounding variables were used to adjust the logistic regression model [35]. Nevertheless, estimates of the effectiveness of the influenza vaccine have been frequently confounded, indicating that a different approach should be used with alternative study designs, different from the typical methods used to study drug exposure [36-38].\nAmong other limitations of our study is the reliability of the COVID-19 diagnoses; we included individuals without a confirmed result, as during the first wave of the pandemic in our setting, PCR tests were not always performed. This limitation has been described in other studies including those conducted at the beginning of the pandemic when diagnostic tests for COVID-19 were not widely available and clinical algorithms were used to assess COVID-19 diagnoses [39]. We compared our number of COVID-19 cases with the official COVID-19 case numbers provided by the AQuAS during the pandemic [24]. Another limitation is the lack of hospital information: we could not capture ICU admissions, ventilation, or treatments administered upon admission, which clearly have an influence on the prognosis and outcomes of COVID-19. Finally, we have not conducted any subgroup analysis that could have indicated any condition potentially resulting in any benefit or harm from influenza vaccination.", "In conclusion, we were not able to establish a protective role of the immunity conferred by the influenza vaccine on the outcomes of COVID-19 infection. Nonetheless, our study adds more evidence to the analysis of the possible link between the quality of the conferred immunity and outcomes of COVID-19 infection, and it has some strengths, such as the large cohort size, its representativeness with respect to the general population, and the completeness of its sociodemographic data. We have already highlighted that our cohort comprises individuals who received care from the PHC system; hence, we have estimated the risk of complications for a different population from the hospitalized individuals who are usually assessed in multiple studies." ]
[ null, null, null, null, null, null, null, null, null, null, null ]
[ "Introduction", "Methods", "Study Design", "Data Source", "COVID-19 Classification", "Influenza Immunization", "Variables", "Statistical Analysis", "Ethical Considerations", "Results", "Discussion", "Principal Findings", "Comparison With Prior Work", "Limitations", "Conclusions" ]
[ "COVID-19 is caused by SARS-CoV-2, a novel coronavirus that emerged in China in 2019, which became the primary agent of a new pandemic that rapidly spread worldwide [1], with an average global infection fatality rate of approximately 0.15%, depending on the data analyzed [2]. SARS-CoV-2 mainly affects the respiratory tract and uses surface proteins in order to infect the host [3].\nAlthough new variants of SARS-CoV-2 have emerged since December 2020, the coronavirus’ genome is composed of RNA and depends on the RNA polymerase to generate its proteins, with a mechanism of error correction that results in a lower mutation rate than the influenza virus [4]. This low mutation rate may suggest that the vaccines developed against SARS-CoV-2, as well as the immunity generated in those patients who were infected, could represent a long-lasting immunity [5,6].\nCOVID-19, similar to influenza A and B, is caused by RNA virus and produce similar symptoms. The influenza virus needs the hemagglutinin and neuraminidase surface proteins to infect cells, whereas SARS-CoV-2 needs the S protein [5]. Previous in vitro and animal studies suggest an induction pathway of indirect etiological immunity between the influenza vaccine and SARS-CoV-2. Animal models suggest that some influenza subtypes might lead to regulation of the angiotensin-converting enzyme-2, with protective properties against SARS [7]. An unspecific effect of infection and vaccination on the immune system and susceptibility to other infections has also been reported, albeit with discordant data [8-10]. Some modeling studies have suggested a possible association between influenza immunization and COVID-19 [11-14].\nA study conducted in Australia assessed the cellular and humoral immune responses during and after disease occurrence in a patient with a mild COVID-19 infection. They found that the immune response in different cell types is associated with clinical recovery. These results are coincident with similar findings among patients with influenza reported by the same authors [15,16].\nOther studies observed differences in the susceptibility to COVID-19 in children of different ages with a lower infection rate than that in adults and older individuals [17]. Although the mechanism underlying these differences in severity and susceptibility is unclear, a possible explanation might be the difference in the quantity and quality of the immune function determined by the history of infections and the recent vaccines administered [18].\nConsequently, a link between the quality of the immunity and recovery from COVID-19 may exist. Thus, we hypothesized that the immunity resulting from the previous influenza vaccination would boost part of the immunity against SARS-CoV-2, and we aimed to investigate whether individuals with COVID-19 could have benefited from vaccination against influenza.", "[SUBTITLE] Study Design [SUBSECTION] We performed a population-based cohort study including all adults with COVID-19 in Catalonia, Spain, who were registered as confirmed cases (through the polymerase chain reaction [PCR]) or as probable cases (not confirmed through PCR but with International Classification of Diseases (ICD)-10 codes registered that are compatible with COVID-19) in the primary health care (PHC) system. All individuals with COVID-19 were diagnosed from the pandemic’s onset (March 2020) to June 30, 2020. Participants were compared on the basis of their influenza vaccination status between those having received the influenza vaccine before having COVID-19 (vaccinated in the previous influenza seasonal campaign in 2019-2020 or before) [19] with those who were not vaccinated.\nWe performed a population-based cohort study including all adults with COVID-19 in Catalonia, Spain, who were registered as confirmed cases (through the polymerase chain reaction [PCR]) or as probable cases (not confirmed through PCR but with International Classification of Diseases (ICD)-10 codes registered that are compatible with COVID-19) in the primary health care (PHC) system. All individuals with COVID-19 were diagnosed from the pandemic’s onset (March 2020) to June 30, 2020. Participants were compared on the basis of their influenza vaccination status between those having received the influenza vaccine before having COVID-19 (vaccinated in the previous influenza seasonal campaign in 2019-2020 or before) [19] with those who were not vaccinated.\n[SUBTITLE] Data Source [SUBSECTION] Our data source is the Information System for Research in Primary Care [20], which captures clinical information of approximately 5.8 million people from Catalonia, Spain (approximately 80% of the Catalan population). This information is pseudonymized, having originated from different data sources: (1) electronic health records in PHC system of the Catalan Health Institute, including sociodemographic characteristics, residents in nursing homes or long-term care facilities (LTCFs), comorbidities registered as ICD-10 codes [21], specialist referrals, clinical parameters, toxic habits, sickness leave, date of death, laboratory test data, and drug prescriptions issued in the PHC system, registered in accordance with the anatomical therapeutic chemical classification system [22]; (2) pharmacy invoice data corresponding to the PHC drug prescriptions; (3) database of diagnoses upon hospital discharge [23]; and (4) COVID-19 data from the Catalan Agency of Health Quality and Evaluation (AQuAS) [24].\nOur data source is the Information System for Research in Primary Care [20], which captures clinical information of approximately 5.8 million people from Catalonia, Spain (approximately 80% of the Catalan population). This information is pseudonymized, having originated from different data sources: (1) electronic health records in PHC system of the Catalan Health Institute, including sociodemographic characteristics, residents in nursing homes or long-term care facilities (LTCFs), comorbidities registered as ICD-10 codes [21], specialist referrals, clinical parameters, toxic habits, sickness leave, date of death, laboratory test data, and drug prescriptions issued in the PHC system, registered in accordance with the anatomical therapeutic chemical classification system [22]; (2) pharmacy invoice data corresponding to the PHC drug prescriptions; (3) database of diagnoses upon hospital discharge [23]; and (4) COVID-19 data from the Catalan Agency of Health Quality and Evaluation (AQuAS) [24].\n[SUBTITLE] COVID-19 Classification [SUBSECTION] Participants were classified in accordance with the following criteria: confirmed cases are those with a confirmed COVID-19 diagnosis record, positive PCR outcome, or a positive serology test result. Those with an unconfirmed diagnosis or test (possible or unclear) along with any individual with a record of hospitalization, pneumonia, or death related to COVID-19 were considered probable cases. During the first wave of the COVID-19 pandemic in Catalonia, PCR tests were not routinely conducted for all individuals with compatible symptoms owing to the unavailability of laboratory kits to carry out the tests. Thus, we needed to capture those patients with a possible diagnosis of COVID-19, such as those admitted to hospital with pneumonia or other COVID-19 symptoms, who were not tested. We designed an algorithm to classify patients as “COVID possible” when a test result was unavailable along with registered entries from different databases: PCR tests or serology tests conducted in different settings, discharge diagnoses of pneumonia from Catalan hospitals or from emergency departments, and ICD-10 diagnoses related to COVID-19 coded in PHC. The date of COVID-19 diagnosis was set to be the first of all records used per patient. To guarantee that our algorithm is not far from the Catalan population, the resulting cohort was compared to the official COVID-19 cases reported by the AQuAS during the pandemic [24].\nParticipants were classified in accordance with the following criteria: confirmed cases are those with a confirmed COVID-19 diagnosis record, positive PCR outcome, or a positive serology test result. Those with an unconfirmed diagnosis or test (possible or unclear) along with any individual with a record of hospitalization, pneumonia, or death related to COVID-19 were considered probable cases. During the first wave of the COVID-19 pandemic in Catalonia, PCR tests were not routinely conducted for all individuals with compatible symptoms owing to the unavailability of laboratory kits to carry out the tests. Thus, we needed to capture those patients with a possible diagnosis of COVID-19, such as those admitted to hospital with pneumonia or other COVID-19 symptoms, who were not tested. We designed an algorithm to classify patients as “COVID possible” when a test result was unavailable along with registered entries from different databases: PCR tests or serology tests conducted in different settings, discharge diagnoses of pneumonia from Catalan hospitals or from emergency departments, and ICD-10 diagnoses related to COVID-19 coded in PHC. The date of COVID-19 diagnosis was set to be the first of all records used per patient. To guarantee that our algorithm is not far from the Catalan population, the resulting cohort was compared to the official COVID-19 cases reported by the AQuAS during the pandemic [24].\n[SUBTITLE] Influenza Immunization [SUBSECTION] Patients were classified as having taken the influenza vaccine if they had been vaccinated at any time before having COVID-19, and grouped in accordance with the seasonal vaccination campaign: the immediate previous campaign (2019-2020) or other vaccination campaigns (2018-2019 and before) [19,25].\nPatients were classified as having taken the influenza vaccine if they had been vaccinated at any time before having COVID-19, and grouped in accordance with the seasonal vaccination campaign: the immediate previous campaign (2019-2020) or other vaccination campaigns (2018-2019 and before) [19,25].\n[SUBTITLE] Variables [SUBSECTION] At baseline, the following variables were captured: sex, age, geographical area, MEDEA (Mortalidad en áreas pequeñas Españolas y Desigualdades Económicas y Ambientales [Mortality in small Spanish areas and economic and environmental inequalities]) socioeconomic index (deprivation index based on 5 indicators of socioeconomic position; it helps analyze health inequity, and higher the MEDEA socioeconomic index, worse the deprivation) [26], BMI, residence in nursing homes, smoking habits, comorbidities, and taking influenza vaccines and pneumococcal and tuberculosis vaccines.\nThe main outcomes assessed during follow-up (up to June 2020) were at least one of the following variables: diagnosis of pneumonia, hospital admission, and mortality. The risk of these events was analyzed in those people who had been vaccinated against influenza at any time before having COVID-19, in those who were recently vaccinated (campaign of 2019-2020), and in those systematically vaccinated (who had been vaccinated at least during 3 different campaigns). We analyzed the same outcomes excluding those of people living in LTCFs, where vaccination is nearly universal in our country [27].\nAt baseline, the following variables were captured: sex, age, geographical area, MEDEA (Mortalidad en áreas pequeñas Españolas y Desigualdades Económicas y Ambientales [Mortality in small Spanish areas and economic and environmental inequalities]) socioeconomic index (deprivation index based on 5 indicators of socioeconomic position; it helps analyze health inequity, and higher the MEDEA socioeconomic index, worse the deprivation) [26], BMI, residence in nursing homes, smoking habits, comorbidities, and taking influenza vaccines and pneumococcal and tuberculosis vaccines.\nThe main outcomes assessed during follow-up (up to June 2020) were at least one of the following variables: diagnosis of pneumonia, hospital admission, and mortality. The risk of these events was analyzed in those people who had been vaccinated against influenza at any time before having COVID-19, in those who were recently vaccinated (campaign of 2019-2020), and in those systematically vaccinated (who had been vaccinated at least during 3 different campaigns). We analyzed the same outcomes excluding those of people living in LTCFs, where vaccination is nearly universal in our country [27].\n[SUBTITLE] Statistical Analysis [SUBSECTION] Quantitative variables were described as mean (SD) values, whereas categorical variables were described as the proportion of vaccinated and unvaccinated individuals. Univariate analyses were based on the Student t test or chi-square test depending on the variable.\nFor each outcome, we fitted a logistic regression model to estimate an odds ratio (OR) comparing the prevalence of each outcome among individuals given the influenza vaccine to that of unvaccinated individuals. The logistic model was fitted along with other covariables such as smoking habits, age, comorbidities (asthma, autoimmune disorders, prior cerebrovascular disease, chronic kidney disease, chronic pulmonary obstructive disease, diabetes, heart failure, hypertension, ischemic heart disease, mental-behavioral disorders, obesity, organ transplant, and other respiratory diseases), concomitant drugs, and previous vaccines (pneumococcal and tuberculosis). As a sensitivity analysis, we conducted the same analysis on a matched population. Individuals vaccinated against influenza and unvaccinated controls were matched 1:2 in accordance with their age and gender at the time of infection or on an index date, and the reported ORs were obtained by fitting a conditional logistic regression model (clogit) accounting for matched pairs and adjusted using the same covariables as in the logistic model. We used the Wald test on the fitted coefficient to determine whether the log-odds were significantly different from 0 at a threshold of .05. All analyses were performed in R (version 4.1.0 or above; The R Foundation).\nQuantitative variables were described as mean (SD) values, whereas categorical variables were described as the proportion of vaccinated and unvaccinated individuals. Univariate analyses were based on the Student t test or chi-square test depending on the variable.\nFor each outcome, we fitted a logistic regression model to estimate an odds ratio (OR) comparing the prevalence of each outcome among individuals given the influenza vaccine to that of unvaccinated individuals. The logistic model was fitted along with other covariables such as smoking habits, age, comorbidities (asthma, autoimmune disorders, prior cerebrovascular disease, chronic kidney disease, chronic pulmonary obstructive disease, diabetes, heart failure, hypertension, ischemic heart disease, mental-behavioral disorders, obesity, organ transplant, and other respiratory diseases), concomitant drugs, and previous vaccines (pneumococcal and tuberculosis). As a sensitivity analysis, we conducted the same analysis on a matched population. Individuals vaccinated against influenza and unvaccinated controls were matched 1:2 in accordance with their age and gender at the time of infection or on an index date, and the reported ORs were obtained by fitting a conditional logistic regression model (clogit) accounting for matched pairs and adjusted using the same covariables as in the logistic model. We used the Wald test on the fitted coefficient to determine whether the log-odds were significantly different from 0 at a threshold of .05. All analyses were performed in R (version 4.1.0 or above; The R Foundation).\n[SUBTITLE] Ethical Considerations [SUBSECTION] The study protocol was approved by the Research Ethics Committee of Institut Universitari d’Investigació en Atenció Primària (June 3, 2020). This is a database research study that has been conducted in accordance with the tenets of the Declaration of Helsinki (Fortaleza, Brazil 2013) and does not require consent from the study participants for the purpose of publication. The need for consent was waived by the Research Ethics Committee of Institut Universitari d’Investigació en Atenció Primària as it is deemed unnecessary according to the European legislation (Regulation [EU] 2016/679).\nThe study protocol was approved by the Research Ethics Committee of Institut Universitari d’Investigació en Atenció Primària (June 3, 2020). This is a database research study that has been conducted in accordance with the tenets of the Declaration of Helsinki (Fortaleza, Brazil 2013) and does not require consent from the study participants for the purpose of publication. The need for consent was waived by the Research Ethics Committee of Institut Universitari d’Investigació en Atenció Primària as it is deemed unnecessary according to the European legislation (Regulation [EU] 2016/679).", "We performed a population-based cohort study including all adults with COVID-19 in Catalonia, Spain, who were registered as confirmed cases (through the polymerase chain reaction [PCR]) or as probable cases (not confirmed through PCR but with International Classification of Diseases (ICD)-10 codes registered that are compatible with COVID-19) in the primary health care (PHC) system. All individuals with COVID-19 were diagnosed from the pandemic’s onset (March 2020) to June 30, 2020. Participants were compared on the basis of their influenza vaccination status between those having received the influenza vaccine before having COVID-19 (vaccinated in the previous influenza seasonal campaign in 2019-2020 or before) [19] with those who were not vaccinated.", "Our data source is the Information System for Research in Primary Care [20], which captures clinical information of approximately 5.8 million people from Catalonia, Spain (approximately 80% of the Catalan population). This information is pseudonymized, having originated from different data sources: (1) electronic health records in PHC system of the Catalan Health Institute, including sociodemographic characteristics, residents in nursing homes or long-term care facilities (LTCFs), comorbidities registered as ICD-10 codes [21], specialist referrals, clinical parameters, toxic habits, sickness leave, date of death, laboratory test data, and drug prescriptions issued in the PHC system, registered in accordance with the anatomical therapeutic chemical classification system [22]; (2) pharmacy invoice data corresponding to the PHC drug prescriptions; (3) database of diagnoses upon hospital discharge [23]; and (4) COVID-19 data from the Catalan Agency of Health Quality and Evaluation (AQuAS) [24].", "Participants were classified in accordance with the following criteria: confirmed cases are those with a confirmed COVID-19 diagnosis record, positive PCR outcome, or a positive serology test result. Those with an unconfirmed diagnosis or test (possible or unclear) along with any individual with a record of hospitalization, pneumonia, or death related to COVID-19 were considered probable cases. During the first wave of the COVID-19 pandemic in Catalonia, PCR tests were not routinely conducted for all individuals with compatible symptoms owing to the unavailability of laboratory kits to carry out the tests. Thus, we needed to capture those patients with a possible diagnosis of COVID-19, such as those admitted to hospital with pneumonia or other COVID-19 symptoms, who were not tested. We designed an algorithm to classify patients as “COVID possible” when a test result was unavailable along with registered entries from different databases: PCR tests or serology tests conducted in different settings, discharge diagnoses of pneumonia from Catalan hospitals or from emergency departments, and ICD-10 diagnoses related to COVID-19 coded in PHC. The date of COVID-19 diagnosis was set to be the first of all records used per patient. To guarantee that our algorithm is not far from the Catalan population, the resulting cohort was compared to the official COVID-19 cases reported by the AQuAS during the pandemic [24].", "Patients were classified as having taken the influenza vaccine if they had been vaccinated at any time before having COVID-19, and grouped in accordance with the seasonal vaccination campaign: the immediate previous campaign (2019-2020) or other vaccination campaigns (2018-2019 and before) [19,25].", "At baseline, the following variables were captured: sex, age, geographical area, MEDEA (Mortalidad en áreas pequeñas Españolas y Desigualdades Económicas y Ambientales [Mortality in small Spanish areas and economic and environmental inequalities]) socioeconomic index (deprivation index based on 5 indicators of socioeconomic position; it helps analyze health inequity, and higher the MEDEA socioeconomic index, worse the deprivation) [26], BMI, residence in nursing homes, smoking habits, comorbidities, and taking influenza vaccines and pneumococcal and tuberculosis vaccines.\nThe main outcomes assessed during follow-up (up to June 2020) were at least one of the following variables: diagnosis of pneumonia, hospital admission, and mortality. The risk of these events was analyzed in those people who had been vaccinated against influenza at any time before having COVID-19, in those who were recently vaccinated (campaign of 2019-2020), and in those systematically vaccinated (who had been vaccinated at least during 3 different campaigns). We analyzed the same outcomes excluding those of people living in LTCFs, where vaccination is nearly universal in our country [27].", "Quantitative variables were described as mean (SD) values, whereas categorical variables were described as the proportion of vaccinated and unvaccinated individuals. Univariate analyses were based on the Student t test or chi-square test depending on the variable.\nFor each outcome, we fitted a logistic regression model to estimate an odds ratio (OR) comparing the prevalence of each outcome among individuals given the influenza vaccine to that of unvaccinated individuals. The logistic model was fitted along with other covariables such as smoking habits, age, comorbidities (asthma, autoimmune disorders, prior cerebrovascular disease, chronic kidney disease, chronic pulmonary obstructive disease, diabetes, heart failure, hypertension, ischemic heart disease, mental-behavioral disorders, obesity, organ transplant, and other respiratory diseases), concomitant drugs, and previous vaccines (pneumococcal and tuberculosis). As a sensitivity analysis, we conducted the same analysis on a matched population. Individuals vaccinated against influenza and unvaccinated controls were matched 1:2 in accordance with their age and gender at the time of infection or on an index date, and the reported ORs were obtained by fitting a conditional logistic regression model (clogit) accounting for matched pairs and adjusted using the same covariables as in the logistic model. We used the Wald test on the fitted coefficient to determine whether the log-odds were significantly different from 0 at a threshold of .05. All analyses were performed in R (version 4.1.0 or above; The R Foundation).", "The study protocol was approved by the Research Ethics Committee of Institut Universitari d’Investigació en Atenció Primària (June 3, 2020). This is a database research study that has been conducted in accordance with the tenets of the Declaration of Helsinki (Fortaleza, Brazil 2013) and does not require consent from the study participants for the purpose of publication. The need for consent was waived by the Research Ethics Committee of Institut Universitari d’Investigació en Atenció Primària as it is deemed unnecessary according to the European legislation (Regulation [EU] 2016/679).", "We included 309,039 individuals with COVID-19 during the first wave of the pandemic in accordance with their influenza immunization status (Table 1, Multimedia Appendix 1); 114,181 (36.9%) participants had received the influenza vaccine at least once before having COVID-19 and 194,858 (63.1%) had not been vaccinated, with more women in both groups, especially in the vaccinated cohort (61.0% women vs 39.0% men). The mean age was higher for vaccinated individuals (64.3 years, with 52.3% of them being older than 65 years). Vaccinated individuals had more comorbidities than unvaccinated individuals.\nOf those who received the influenza vaccine, 66,611 (58.3%) had been recently vaccinated (2019-2020) and 75,311 (66%) had been systematically vaccinated against influenza at least during 3 different years (Table 2).\nOf the participants with COVID-19, 11,000 (5.7%) unvaccinated and 21,721 (19%) vaccinated participants presented at least one of the following events: hospital admission, pneumonia, or death. For those who received the influenza vaccine at any time before having COVID-19, the risks of hospitalization (adjusted OR 1.14, 95% CI 1.10-1.19) and death (OR 1.32, 1.23-1.42) were higher than those among unvaccinated participants. For the recently vaccinated participants, the risk was higher for hospitalization (OR 1.16, 95% CI 1.1-1.23), pneumonia (OR 1.12, 95% CI 1.02-1.23), and death (OR 1.14, 95% CI 1.04-1.24). For people with recurrent vaccination, the risk of the 3 outcomes was also higher that among unvaccinated participants (OR 1.07, 1.16, and 1.24, respectively; Table 3). We have also analyzed the results in a matched population of vaccinated versus unvaccinated participants, revealing a higher risk of pneumonia and mortality, with an adjusted OR of 1.11 (95% CI 1.01-1.23) and 1.28 (95% CI 1.07-1.53), respectively (Multimedia Appendix 2).\nThe risks of the outcomes based on influenza vaccination status and excluding those patients living in LTCFs are shown in Figure 1. For non-LTCF residents, the results are similar to those for the whole population, except that there was no significant increase in mortality (OR 0.93, 95% CI 0.85-1.03).\nSociodemographic and clinical characteristics of the study population (N=309,039).\nTaking influenza vaccines before having COVID-19.\nLogistic regression model of COVID-19 outcomes based on influenza immunization status.\naA logistic regression model adjusted with the following relevant covariables was fitted: smoking habits, age, comorbidities (asthma, autoimmune disorders, prior cerebrovascular disease, chronic kidney disease, chronic pulmonary obstructive disease, diabetes, heart failure, hypertension, ischemic heart disease, mental-behavioral disorders, obesity, organ transplant, and other respiratory diseases), co-medication, and previous vaccines (pneumococcal and tuberculosis).\nRisk of death and of combined COVID-19 complications in all the vaccinated population and excluding people living in long-term care facilities (LTCF).", "[SUBTITLE] Principal Findings [SUBSECTION] We analyzed the negative outcomes among people with COVID-19 (N=309,039) and compared those who had received the influenza vaccine with those who were never vaccinated. Those who received the vaccine any time before having COVID-19 were at a higher risk of complications than those who were unvaccinated. We obtained similar results for those who were recently vaccinated (2019-2020 campaign) and for those who were systematically vaccinated (at least 3 years), and the same comparisons were carried out after excluding individuals living in LTCFs. We also obtained similar results on matching vaccinated and unvaccinated individuals. Thus, we did not find a possible link between receiving the influenza vaccine and presenting better clinical outcomes after a COVID-19 infection.\nWe analyzed the negative outcomes among people with COVID-19 (N=309,039) and compared those who had received the influenza vaccine with those who were never vaccinated. Those who received the vaccine any time before having COVID-19 were at a higher risk of complications than those who were unvaccinated. We obtained similar results for those who were recently vaccinated (2019-2020 campaign) and for those who were systematically vaccinated (at least 3 years), and the same comparisons were carried out after excluding individuals living in LTCFs. We also obtained similar results on matching vaccinated and unvaccinated individuals. Thus, we did not find a possible link between receiving the influenza vaccine and presenting better clinical outcomes after a COVID-19 infection.\n[SUBTITLE] Comparison With Prior Work [SUBSECTION] Some researchers have studied this possible association. Massoudi and Mohit [28] conducted a study in a hospital in Iran including health care workers, with 80 of them COVID-19 cases confirmed through PCR or on the basis of their symptoms, and 181 of them were controls. They concluded that individuals who were confirmed cases were less likely to have received the 2019 influenza vaccine (OR 0.04, 95% CI 0.01-0.14), suggesting a protective association between the influenza vaccine and COVID-19. Their study had several limitations, such as the lack of availability of COVID-19 test kits or the samples limited to the workers of a single hospital [28].\nCandelli et al [29] assessed 602 patients with COVID-19 enrolled at the emergency department in a hospital in Italy, of whom 24.9% had been previously vaccinated against influenza. They found that influenza immunization was independently associated with a lower risk of death at 60 days (OR 0.20, 95% CI 0.08-0.51), but not with a reduced need of endotracheal intubation (OR 0.73, 95% CI 0.35-1.56) [29].\nA study conducted in Brazil [30] included 92,664 confirmed cases of COVID-19, of whom 31.1% had been recently vaccinated against influenza. They found that the vaccinated individuals were at a lower risk of needing intensive care for COVID-19 (OR 0.92, 95% CI 0.86-0.99), a lower risk of needing respiratory support (OR 0.81, 95% CI 0.74-0.88), and lower odds of mortality (OR 0.82, 95% CI 0.75-0.89) [30].\nIn a systematic review [31] including 12 studies, the authors examined whether influenza vaccination affects the risk of being infected with SARS-CoV-2 and the risk of complicated illness or poor outcomes among patients with COVID-19, all of whom having been confirmed cases through PCR testing. They concluded that influenza vaccination is unlikely to be associated with an increase in the risk of COVID-19 infection or severity and the risk of associated death [31].\nThere are reports from some countries with high influenza vaccination rates and high incidences of COVID-19 and mortality [32,33]. For instance, Kline et al [33] compared people vaccinated against influenza with unvaccinated individuals admitted to hospital for COVID-19, and they found no differences in the rate of admission to the intensive care unit, intubation, or other complications [33]. Our results follow these same trends in a cohort of the general population attended to in the PHC system and not only hospitalized patients.\nSome researchers have studied this possible association. Massoudi and Mohit [28] conducted a study in a hospital in Iran including health care workers, with 80 of them COVID-19 cases confirmed through PCR or on the basis of their symptoms, and 181 of them were controls. They concluded that individuals who were confirmed cases were less likely to have received the 2019 influenza vaccine (OR 0.04, 95% CI 0.01-0.14), suggesting a protective association between the influenza vaccine and COVID-19. Their study had several limitations, such as the lack of availability of COVID-19 test kits or the samples limited to the workers of a single hospital [28].\nCandelli et al [29] assessed 602 patients with COVID-19 enrolled at the emergency department in a hospital in Italy, of whom 24.9% had been previously vaccinated against influenza. They found that influenza immunization was independently associated with a lower risk of death at 60 days (OR 0.20, 95% CI 0.08-0.51), but not with a reduced need of endotracheal intubation (OR 0.73, 95% CI 0.35-1.56) [29].\nA study conducted in Brazil [30] included 92,664 confirmed cases of COVID-19, of whom 31.1% had been recently vaccinated against influenza. They found that the vaccinated individuals were at a lower risk of needing intensive care for COVID-19 (OR 0.92, 95% CI 0.86-0.99), a lower risk of needing respiratory support (OR 0.81, 95% CI 0.74-0.88), and lower odds of mortality (OR 0.82, 95% CI 0.75-0.89) [30].\nIn a systematic review [31] including 12 studies, the authors examined whether influenza vaccination affects the risk of being infected with SARS-CoV-2 and the risk of complicated illness or poor outcomes among patients with COVID-19, all of whom having been confirmed cases through PCR testing. They concluded that influenza vaccination is unlikely to be associated with an increase in the risk of COVID-19 infection or severity and the risk of associated death [31].\nThere are reports from some countries with high influenza vaccination rates and high incidences of COVID-19 and mortality [32,33]. For instance, Kline et al [33] compared people vaccinated against influenza with unvaccinated individuals admitted to hospital for COVID-19, and they found no differences in the rate of admission to the intensive care unit, intubation, or other complications [33]. Our results follow these same trends in a cohort of the general population attended to in the PHC system and not only hospitalized patients.\n[SUBTITLE] Limitations [SUBSECTION] We need to consider that our results correspond to the first wave of the COVID-19 pandemic, when there were more negative outcomes and mortalities due to COVID-19 than in the subsequent waves in our setting; thus, this higher statistical power allowed us to detect differences. Furthermore, in subsequent waves, more confounders might have been present, such as COVID-19 vaccination or effects of the different SARS-CoV-2 variants, making it more difficult to manage their potential effect in the analysis of the outcomes of the infection.\nWe also need to bear in mind that the target population for the influenza vaccine in our country are people older than 60 years, individuals with chronic comorbidities or immunodeficiency, and health care workers among others [34], some of them being at a high risk of COVID-19 complications, which is why confounding variables were used to adjust the logistic regression model [35]. Nevertheless, estimates of the effectiveness of the influenza vaccine have been frequently confounded, indicating that a different approach should be used with alternative study designs, different from the typical methods used to study drug exposure [36-38].\nAmong other limitations of our study is the reliability of the COVID-19 diagnoses; we included individuals without a confirmed result, as during the first wave of the pandemic in our setting, PCR tests were not always performed. This limitation has been described in other studies including those conducted at the beginning of the pandemic when diagnostic tests for COVID-19 were not widely available and clinical algorithms were used to assess COVID-19 diagnoses [39]. We compared our number of COVID-19 cases with the official COVID-19 case numbers provided by the AQuAS during the pandemic [24]. Another limitation is the lack of hospital information: we could not capture ICU admissions, ventilation, or treatments administered upon admission, which clearly have an influence on the prognosis and outcomes of COVID-19. Finally, we have not conducted any subgroup analysis that could have indicated any condition potentially resulting in any benefit or harm from influenza vaccination.\nWe need to consider that our results correspond to the first wave of the COVID-19 pandemic, when there were more negative outcomes and mortalities due to COVID-19 than in the subsequent waves in our setting; thus, this higher statistical power allowed us to detect differences. Furthermore, in subsequent waves, more confounders might have been present, such as COVID-19 vaccination or effects of the different SARS-CoV-2 variants, making it more difficult to manage their potential effect in the analysis of the outcomes of the infection.\nWe also need to bear in mind that the target population for the influenza vaccine in our country are people older than 60 years, individuals with chronic comorbidities or immunodeficiency, and health care workers among others [34], some of them being at a high risk of COVID-19 complications, which is why confounding variables were used to adjust the logistic regression model [35]. Nevertheless, estimates of the effectiveness of the influenza vaccine have been frequently confounded, indicating that a different approach should be used with alternative study designs, different from the typical methods used to study drug exposure [36-38].\nAmong other limitations of our study is the reliability of the COVID-19 diagnoses; we included individuals without a confirmed result, as during the first wave of the pandemic in our setting, PCR tests were not always performed. This limitation has been described in other studies including those conducted at the beginning of the pandemic when diagnostic tests for COVID-19 were not widely available and clinical algorithms were used to assess COVID-19 diagnoses [39]. We compared our number of COVID-19 cases with the official COVID-19 case numbers provided by the AQuAS during the pandemic [24]. Another limitation is the lack of hospital information: we could not capture ICU admissions, ventilation, or treatments administered upon admission, which clearly have an influence on the prognosis and outcomes of COVID-19. Finally, we have not conducted any subgroup analysis that could have indicated any condition potentially resulting in any benefit or harm from influenza vaccination.\n[SUBTITLE] Conclusions [SUBSECTION] In conclusion, we were not able to establish a protective role of the immunity conferred by the influenza vaccine on the outcomes of COVID-19 infection. Nonetheless, our study adds more evidence to the analysis of the possible link between the quality of the conferred immunity and outcomes of COVID-19 infection, and it has some strengths, such as the large cohort size, its representativeness with respect to the general population, and the completeness of its sociodemographic data. We have already highlighted that our cohort comprises individuals who received care from the PHC system; hence, we have estimated the risk of complications for a different population from the hospitalized individuals who are usually assessed in multiple studies.\nIn conclusion, we were not able to establish a protective role of the immunity conferred by the influenza vaccine on the outcomes of COVID-19 infection. Nonetheless, our study adds more evidence to the analysis of the possible link between the quality of the conferred immunity and outcomes of COVID-19 infection, and it has some strengths, such as the large cohort size, its representativeness with respect to the general population, and the completeness of its sociodemographic data. We have already highlighted that our cohort comprises individuals who received care from the PHC system; hence, we have estimated the risk of complications for a different population from the hospitalized individuals who are usually assessed in multiple studies.", "We analyzed the negative outcomes among people with COVID-19 (N=309,039) and compared those who had received the influenza vaccine with those who were never vaccinated. Those who received the vaccine any time before having COVID-19 were at a higher risk of complications than those who were unvaccinated. We obtained similar results for those who were recently vaccinated (2019-2020 campaign) and for those who were systematically vaccinated (at least 3 years), and the same comparisons were carried out after excluding individuals living in LTCFs. We also obtained similar results on matching vaccinated and unvaccinated individuals. Thus, we did not find a possible link between receiving the influenza vaccine and presenting better clinical outcomes after a COVID-19 infection.", "Some researchers have studied this possible association. Massoudi and Mohit [28] conducted a study in a hospital in Iran including health care workers, with 80 of them COVID-19 cases confirmed through PCR or on the basis of their symptoms, and 181 of them were controls. They concluded that individuals who were confirmed cases were less likely to have received the 2019 influenza vaccine (OR 0.04, 95% CI 0.01-0.14), suggesting a protective association between the influenza vaccine and COVID-19. Their study had several limitations, such as the lack of availability of COVID-19 test kits or the samples limited to the workers of a single hospital [28].\nCandelli et al [29] assessed 602 patients with COVID-19 enrolled at the emergency department in a hospital in Italy, of whom 24.9% had been previously vaccinated against influenza. They found that influenza immunization was independently associated with a lower risk of death at 60 days (OR 0.20, 95% CI 0.08-0.51), but not with a reduced need of endotracheal intubation (OR 0.73, 95% CI 0.35-1.56) [29].\nA study conducted in Brazil [30] included 92,664 confirmed cases of COVID-19, of whom 31.1% had been recently vaccinated against influenza. They found that the vaccinated individuals were at a lower risk of needing intensive care for COVID-19 (OR 0.92, 95% CI 0.86-0.99), a lower risk of needing respiratory support (OR 0.81, 95% CI 0.74-0.88), and lower odds of mortality (OR 0.82, 95% CI 0.75-0.89) [30].\nIn a systematic review [31] including 12 studies, the authors examined whether influenza vaccination affects the risk of being infected with SARS-CoV-2 and the risk of complicated illness or poor outcomes among patients with COVID-19, all of whom having been confirmed cases through PCR testing. They concluded that influenza vaccination is unlikely to be associated with an increase in the risk of COVID-19 infection or severity and the risk of associated death [31].\nThere are reports from some countries with high influenza vaccination rates and high incidences of COVID-19 and mortality [32,33]. For instance, Kline et al [33] compared people vaccinated against influenza with unvaccinated individuals admitted to hospital for COVID-19, and they found no differences in the rate of admission to the intensive care unit, intubation, or other complications [33]. Our results follow these same trends in a cohort of the general population attended to in the PHC system and not only hospitalized patients.", "We need to consider that our results correspond to the first wave of the COVID-19 pandemic, when there were more negative outcomes and mortalities due to COVID-19 than in the subsequent waves in our setting; thus, this higher statistical power allowed us to detect differences. Furthermore, in subsequent waves, more confounders might have been present, such as COVID-19 vaccination or effects of the different SARS-CoV-2 variants, making it more difficult to manage their potential effect in the analysis of the outcomes of the infection.\nWe also need to bear in mind that the target population for the influenza vaccine in our country are people older than 60 years, individuals with chronic comorbidities or immunodeficiency, and health care workers among others [34], some of them being at a high risk of COVID-19 complications, which is why confounding variables were used to adjust the logistic regression model [35]. Nevertheless, estimates of the effectiveness of the influenza vaccine have been frequently confounded, indicating that a different approach should be used with alternative study designs, different from the typical methods used to study drug exposure [36-38].\nAmong other limitations of our study is the reliability of the COVID-19 diagnoses; we included individuals without a confirmed result, as during the first wave of the pandemic in our setting, PCR tests were not always performed. This limitation has been described in other studies including those conducted at the beginning of the pandemic when diagnostic tests for COVID-19 were not widely available and clinical algorithms were used to assess COVID-19 diagnoses [39]. We compared our number of COVID-19 cases with the official COVID-19 case numbers provided by the AQuAS during the pandemic [24]. Another limitation is the lack of hospital information: we could not capture ICU admissions, ventilation, or treatments administered upon admission, which clearly have an influence on the prognosis and outcomes of COVID-19. Finally, we have not conducted any subgroup analysis that could have indicated any condition potentially resulting in any benefit or harm from influenza vaccination.", "In conclusion, we were not able to establish a protective role of the immunity conferred by the influenza vaccine on the outcomes of COVID-19 infection. Nonetheless, our study adds more evidence to the analysis of the possible link between the quality of the conferred immunity and outcomes of COVID-19 infection, and it has some strengths, such as the large cohort size, its representativeness with respect to the general population, and the completeness of its sociodemographic data. We have already highlighted that our cohort comprises individuals who received care from the PHC system; hence, we have estimated the risk of complications for a different population from the hospitalized individuals who are usually assessed in multiple studies." ]
[ "introduction", "methods", null, null, null, null, null, null, null, "results", "discussion", null, null, null, null ]
[ "SARS-CoV-2", "COVID-19", "influenza vaccines", "pneumonia", "electronic health records", "primary health care", "vaccination", "public health", "cohort study", "epidemiology", "eHeatlh", "health outcome", "mortality" ]
A RASSF8-AS1 based exosomal lncRNAs panel used for diagnostic and prognostic biomarkers for esophageal squamous cell carcinoma.
36266257
Exosomal long non-coding RNA (lncRNA) has been shown to be potential biomarker for cancer diagnosis and follow up. However, little is known about its application in esophageal squamous cell carcinoma (ESCC) detection. Here, we sought to develop a novel diagnostic model based on serum exosomal lncRNAs to improve ESCC screening efficiency.
BACKGROUND
A multiphase, case-control study was conducted among 140 ESCC patients and 140 healthy controls. Microarray screening was performed to acquire differentially expressed exosomal lncRNAs in the discovery phase. The diagnostic model Index I was constructed based on a panel of three lncRNAs using logistic regression in the training phase, and were confirmed in a subsequent validation phase. A receiver operating characteristic (ROC) curve was generated to calculate the diagnostic value. The effects of the selected lncRNAs level on ESCC mortality were evaluated using a Cox hazard regression model and Kaplan-Meier curve analysis, and the expression level with clinicopathological features was also calculated. Finally, we explored the oncogenic potential of candidate lncRNA RASSF8-AS1 in vitro and by target mRNA sequencing.
METHODS
Index I was able to discriminate ESCC patients from healthy controls, and showed superiority to classic tumor biomarkers. Moreover, serum levels of the exosomal lncRNAs correlated with clinicopathological features and prognosis. The in vitro assays showed that RASSF8-AS1 played an oncogenic role in ESCC. Target mRNA scanning results suggested involvement of RASSF8-AS1 in tumor immunity and metabolism.
RESULTS
The newly identified serum exosomal lncRNAs could be used as new biomarkers for ESCC, and showed oncogenic potential in ESCC.
CONCLUSION
[ "Humans", "Esophageal Squamous Cell Carcinoma", "RNA, Long Noncoding", "Esophageal Neoplasms", "Prognosis", "Case-Control Studies", "Gene Expression Regulation, Neoplastic", "Biomarkers, Tumor", "RNA, Messenger", "Tumor Suppressor Proteins" ]
9715784
null
null
METHODS
[SUBTITLE] Patients and samples [SUBSECTION] From 2017 to 2021, 140 ESCC patients and 140 healthy individuals were randomly recruited from Jinling Hospital of Nanjing Medical University, each of whom provided 5 ml of whole blood at indicated timepoints. Neither patient received preoperative chemo−/radiotherapy, and the overall survival (OS) was measured from the date of diagnosis to the date of death due to any cause or the date of the most recent follow‐up. All patients were followed every 3 months for the first year, every 6 months for the next 2 years, and then annually. The end of the follow‐up was November 31, 2021. The study was approved by the ethics committee of Jinling hospital, and all enrolled participants signed informed consent. All serum samples were stored at −80°C until use for total RNA extraction. The pathological stage was determined according to the eighth edition of the Union for International Cancer Control TNM staging system. From 2017 to 2021, 140 ESCC patients and 140 healthy individuals were randomly recruited from Jinling Hospital of Nanjing Medical University, each of whom provided 5 ml of whole blood at indicated timepoints. Neither patient received preoperative chemo−/radiotherapy, and the overall survival (OS) was measured from the date of diagnosis to the date of death due to any cause or the date of the most recent follow‐up. All patients were followed every 3 months for the first year, every 6 months for the next 2 years, and then annually. The end of the follow‐up was November 31, 2021. The study was approved by the ethics committee of Jinling hospital, and all enrolled participants signed informed consent. All serum samples were stored at −80°C until use for total RNA extraction. The pathological stage was determined according to the eighth edition of the Union for International Cancer Control TNM staging system. [SUBTITLE] Exosome extraction and identification [SUBSECTION] A total of 1.5 ml of serum was centrifuged at 2000 × g, 4°C for 30 min, and the supernatant was then recentrifuged at 10 000 × g, 4°C for 45 min. The supernatant was then collected and filtered through a membrane of 0.22 μm pore size. The filtrate was collected and further subjected to ultracentrifugation at 10 000 × g, 4°C for 70 min. The precipitate was resuspended in 10 ml of precooling 1 × PBS, and ultracentrifugation repeated. Finally, the precipitate was resuspended in 1.5 ml of precooling 1 × PBS. The isolated exosomes were characterized using transmission electron microscopy (TEM). Exosome concentration‐particle size testing (nanoparticle tracking analysis [NTA]) was performed by Zeta View (Particle Metrix Ltd.). A total of 1.5 ml of serum was centrifuged at 2000 × g, 4°C for 30 min, and the supernatant was then recentrifuged at 10 000 × g, 4°C for 45 min. The supernatant was then collected and filtered through a membrane of 0.22 μm pore size. The filtrate was collected and further subjected to ultracentrifugation at 10 000 × g, 4°C for 70 min. The precipitate was resuspended in 10 ml of precooling 1 × PBS, and ultracentrifugation repeated. Finally, the precipitate was resuspended in 1.5 ml of precooling 1 × PBS. The isolated exosomes were characterized using transmission electron microscopy (TEM). Exosome concentration‐particle size testing (nanoparticle tracking analysis [NTA]) was performed by Zeta View (Particle Metrix Ltd.). [SUBTITLE] RNA extraction and quantitative real‐time PCR (qRT‐PCR) [SUBSECTION] RNA was extracted from exosomes or tissues using an exosome RNA isolation kit according to protocols (Invitrogen). The extracted RNA was synthesized into cDNA by the PrimeScript RT Master Mix (Takara). RT‐qPCR was performed to detect the relative RNA levels of target genes. β‐actin was used as an internal reference gene for analysis. The relative expression levels of the lncRNAs were calculated using 2−∆∆Ct. 20 , 21 The sequences of primers used for qRT‐PCR of the lncRNAs are listed in Table S1. RNA was extracted from exosomes or tissues using an exosome RNA isolation kit according to protocols (Invitrogen). The extracted RNA was synthesized into cDNA by the PrimeScript RT Master Mix (Takara). RT‐qPCR was performed to detect the relative RNA levels of target genes. β‐actin was used as an internal reference gene for analysis. The relative expression levels of the lncRNAs were calculated using 2−∆∆Ct. 20 , 21 The sequences of primers used for qRT‐PCR of the lncRNAs are listed in Table S1. [SUBTITLE] Microarray screening and analysis [SUBSECTION] Microarray screening was performed with Arraystar LncRNA_8 × 60 k (Arraystar), which contains both lncRNA and mRNA probes. Sample labeling and array hybridization were performed according to the Agilent One‐Color Microarray‐Based Gene Expression Analysis protocol (Agilent Technology). Agilent Feature Extraction software (version 11.0.1.1) was used to analyze acquired array images. Quantile normalization and subsequent data processing were performed using the GeneSpring GX version 12.1 software package (Agilent Technologies). After quantile normalization of the raw data, LncRNAs and mRNAs that at least four out of eight samples have flags in Present or Marginal (“All Targets Value”) were chosen for further data analysis. Differentially expressed LncRNAs and mRNAs with statistical significance between the two groups were identified through p‐value/FDR filtering. Microarray screening was performed with Arraystar LncRNA_8 × 60 k (Arraystar), which contains both lncRNA and mRNA probes. Sample labeling and array hybridization were performed according to the Agilent One‐Color Microarray‐Based Gene Expression Analysis protocol (Agilent Technology). Agilent Feature Extraction software (version 11.0.1.1) was used to analyze acquired array images. Quantile normalization and subsequent data processing were performed using the GeneSpring GX version 12.1 software package (Agilent Technologies). After quantile normalization of the raw data, LncRNAs and mRNAs that at least four out of eight samples have flags in Present or Marginal (“All Targets Value”) were chosen for further data analysis. Differentially expressed LncRNAs and mRNAs with statistical significance between the two groups were identified through p‐value/FDR filtering. [SUBTITLE] Cell culture [SUBSECTION] Esophageal squamous cell carcinoma cell lines (TE1, TE13, KYSE150, KYSE140) and normal human esophageal epithelial cells (HET‐1A) were obtained from the Shanghai Institutes for Biological Sciences (Shanghai, China). These cells were cultured in 1640 medium (KeyGene) containing 10% fetal bovine serum (FBS) (KeyGene), at 37°C in a 5% CO2 atmosphere. Esophageal squamous cell carcinoma cell lines (TE1, TE13, KYSE150, KYSE140) and normal human esophageal epithelial cells (HET‐1A) were obtained from the Shanghai Institutes for Biological Sciences (Shanghai, China). These cells were cultured in 1640 medium (KeyGene) containing 10% fetal bovine serum (FBS) (KeyGene), at 37°C in a 5% CO2 atmosphere. [SUBTITLE] Stability analysis for exosomal lncRNAs panel [SUBSECTION] Exosome samples (n = 15) were allocated to aliquots for different processing, including storage under different temperature, treated with RNase A, and subjected to freeze–thaw cycles and acidic/alkaline environments, to test to the stability of exosomal lncRNAs, which is essential for clinical use. Exosome samples (n = 15) were allocated to aliquots for different processing, including storage under different temperature, treated with RNase A, and subjected to freeze–thaw cycles and acidic/alkaline environments, to test to the stability of exosomal lncRNAs, which is essential for clinical use. [SUBTITLE] Transfection and RNA interfering [SUBSECTION] Cells were seeded on six‐well plates (2 × 105/well) with 1640 medium supplemented with 10% FBS overnight. Then, siRNA was transfected at a final concentration of 100 nM using lipofectamine 2000 (Invitrogen), and 24 h post transfection cells were harvested and subsequently RNA was extracted by Trizol reagent and the interfering efficiency was determined by RT‐qPCR. Sequences of siRNAs and negative control are provided in Table S2. Cells were seeded on six‐well plates (2 × 105/well) with 1640 medium supplemented with 10% FBS overnight. Then, siRNA was transfected at a final concentration of 100 nM using lipofectamine 2000 (Invitrogen), and 24 h post transfection cells were harvested and subsequently RNA was extracted by Trizol reagent and the interfering efficiency was determined by RT‐qPCR. Sequences of siRNAs and negative control are provided in Table S2. [SUBTITLE] Cell proliferation, migration, and invasion assays [SUBSECTION] Transfected cells were collected and grown in 96‐well plates (2 × 103 cells per well) overnight and cell proliferation was detected using cell counting kit‐8 (CCK‐8). The reaction products were measured at 450 nm according to the manufacturer's instructions. For migration/invasion assays, 5 × 104 cells were incubated using Transwell kits (8 mm PET, 24‐well Millicell) or matrix‐coated inserts (BD Biosciences). After 24 or 48 h of incubation, cells were fixed with 4% paraformaldehyde and stained with crystal violet (Beyotim). Imaging and counting of migrated cells were conducted by microscopy. Transfected cells were collected and grown in 96‐well plates (2 × 103 cells per well) overnight and cell proliferation was detected using cell counting kit‐8 (CCK‐8). The reaction products were measured at 450 nm according to the manufacturer's instructions. For migration/invasion assays, 5 × 104 cells were incubated using Transwell kits (8 mm PET, 24‐well Millicell) or matrix‐coated inserts (BD Biosciences). After 24 or 48 h of incubation, cells were fixed with 4% paraformaldehyde and stained with crystal violet (Beyotim). Imaging and counting of migrated cells were conducted by microscopy. [SUBTITLE] RNA sequencing and enrichment analysis [SUBSECTION] Cell transfection and RNA interfering were performed as described above. Then, 24 h post transfection, cells were harvested and RNA was extracted using Trizol reagent (Invitrogen). RNA‐seq libraries were prepared using the Illumina RNA‐seq preparation kit and sequenced on illumina nova seq 6000 sequencer. The differentially expressed RNAs (fold change >1.5, p < 0.05) were retrieved and subjected to gene ontology and KEGG pathway analysis. Cell transfection and RNA interfering were performed as described above. Then, 24 h post transfection, cells were harvested and RNA was extracted using Trizol reagent (Invitrogen). RNA‐seq libraries were prepared using the Illumina RNA‐seq preparation kit and sequenced on illumina nova seq 6000 sequencer. The differentially expressed RNAs (fold change >1.5, p < 0.05) were retrieved and subjected to gene ontology and KEGG pathway analysis. [SUBTITLE] Statistical analysis [SUBSECTION] All data were statistically analyzed with SPSS 26.0 (Chicago) and visualized with GraphPad Prism version 8.0 (GraphPad Software). Differences in lncRNAs expression between groups were determined using the Mann–Whitney unpaired t‐test or paired t‐test. The χ2 or Fisher's exact test was used to compare the clinical characteristics of the groups. Receiver operating characteristic (ROC) curves were used to assess the diagnostic performance of selected biomarkers. The optimal cutoff point was determined using Youden's index. DeLong's test was used to evaluate the statistical significance when comparing differences of AUCs. Using the binary status of enrolled participants (patients or controls) as the dependent variable, the regression coefficient for each lncRNA was estimated by a univariate logistic regression model, and was used as the weighting to construct the diagnostic index. The diagnostic index developed in the training phase was also directly applied to the validation phase to evaluate the clinical utility of the identified lncRNAs individually or in combination. A forward conditional logistic regression model was used to analyze the association between selected biomarkers and ESCC mortality. The long‐term prognostic values of selected biomarkers were evaluated using Cox's hazard regression model and Kaplan–Meier curve analysis. Univariate analysis was used to explore clinicopathological factors (i.e., tumor depth, stage, size) associated with OS. Factors with p‐value <0.05 were selected for the multivariate analysis with Cox's proportional hazard regression model, hazard ratio and 95% confidence interval (CI) were calculated. All p‐values <0.05 were considered statistically significant. All data were statistically analyzed with SPSS 26.0 (Chicago) and visualized with GraphPad Prism version 8.0 (GraphPad Software). Differences in lncRNAs expression between groups were determined using the Mann–Whitney unpaired t‐test or paired t‐test. The χ2 or Fisher's exact test was used to compare the clinical characteristics of the groups. Receiver operating characteristic (ROC) curves were used to assess the diagnostic performance of selected biomarkers. The optimal cutoff point was determined using Youden's index. DeLong's test was used to evaluate the statistical significance when comparing differences of AUCs. Using the binary status of enrolled participants (patients or controls) as the dependent variable, the regression coefficient for each lncRNA was estimated by a univariate logistic regression model, and was used as the weighting to construct the diagnostic index. The diagnostic index developed in the training phase was also directly applied to the validation phase to evaluate the clinical utility of the identified lncRNAs individually or in combination. A forward conditional logistic regression model was used to analyze the association between selected biomarkers and ESCC mortality. The long‐term prognostic values of selected biomarkers were evaluated using Cox's hazard regression model and Kaplan–Meier curve analysis. Univariate analysis was used to explore clinicopathological factors (i.e., tumor depth, stage, size) associated with OS. Factors with p‐value <0.05 were selected for the multivariate analysis with Cox's proportional hazard regression model, hazard ratio and 95% confidence interval (CI) were calculated. All p‐values <0.05 were considered statistically significant.
RESULTS
[SUBTITLE] Identification of exosomes in serum [SUBSECTION] A total of 280 participants, including 140 ESCC patients and 140 healthy controls were enrolled in the study, as shown in Table S3. To verify whether the exosomes were successfully isolated from serum, we characterized the isolated exosomes using TEM and NTA. The results showed that the isolated exosomes had a bilayer membrane structure (Figure S1 A), and the particle size distribution of exosomes was about 30–150 nm in diameter (Figure S1 B), meeting the criteria of exosome patterns. A total of 280 participants, including 140 ESCC patients and 140 healthy controls were enrolled in the study, as shown in Table S3. To verify whether the exosomes were successfully isolated from serum, we characterized the isolated exosomes using TEM and NTA. The results showed that the isolated exosomes had a bilayer membrane structure (Figure S1 A), and the particle size distribution of exosomes was about 30–150 nm in diameter (Figure S1 B), meeting the criteria of exosome patterns. [SUBTITLE] Discovery phase: Genome‐wide profiling to identify differentially expressed lncRNAs between ESCC and healthy individual controls [SUBSECTION] The overall workflow of this study is illustrated in Figure 1a. In the discovery phase, we performed LncRNA microarray analysis on serum exosome samples from four preoperative ESCC patients and matched healthy controls. The top 100 differential lncRNAs identified in serum exosome samples from four preoperative ESCC patients and healthy control are shown in Figure 1b (FDR <0.005, Fold change >2, GEO accession number: GSE192662). Circos plots globally and genome‐widely displayed kinds of detected lncRNAs in ESCC and control serum samples (Figure 1c). A previous microarray comparing five pairs of tumor and paracancerous tissues (GSE89102) was also included for further analysis. To achieve more reliable aberrantly expressed lncRNAs, we adopted a more restrictive filtering criteria (FDR <0.005, fold change >5) for both datasets, and integrated the two datasets to finally obtain five (AC098818.2, RASSF8‐AS1, LINC00958, GMDS‐DT and AL591721.1) candidate genes with significant overexpression in ESCC serum samples and cancerous tissues (Figure 1d). Microarray screening results of differential lncRNAs and function prediction. (a) Experimental design overview. (b) Heatmap results of microarray analysis for four preoperative esophageal squamous cell carcinoma (ESCC) patients and matched normal healthy serum samples. (c) Circos plots of lncRNAs in the human genome (hg19). The outer tracks represent the cytoband ideogram of chromosome. For the two tracks, the outer one (blue) represents the levels of lncRNAs in normal tissues and the inner one (red) represents the levels in ESCC tissues. (d) Venn diagram showing consistent upregulation of five lncRNAs in both microarray analyses. (e, f) Signal pathway networks of mRNAs involved in lncRNAs‐mRNAs relationships. To investigate the potential role of the candidate lncRNAs in ESCC, we analyzed mRNA results from the same microarray chip derived from serum samples and performed pathway analysis (Figure S2 A–D). By lncRNAs‐mRNA coexpression network analysis, most of the candidate lncRNAs‐related mRNAs could be enriched in pathways such as MAPK, Ras, Wnt, metabolic pathways and HIF‐1α, which have been identified to be involved in ESCC carcinogenesis (Figure 1e,f). To validate the microarray data, we examined the expression of five candidate lncRNAs in a pilot cohort containing serum exosome samples (n = 20 for ESCC and healthy control separately), and 30 paired tissues, respectively. The qRT‐PCR results showed that the expression of AC098818.2, RASSF8‐AS1 and LINC00958 being significantly increased in ESCC patients' serum compared with healthy, while no significant difference in the expression of AL591721.1 and GMDS‐DT (Figure 2a). Compared with normal esophageal tissues, AC098818.2, RASSF8‐AS1 and LINC00958 expression was significantly upregulated in tumor tissues, whereas the expression level of GMDS‐DT was significantly downregulated, and no significant difference was seen in the expression of AL591721.1 (Figure 2b). Moreover, we examined the expression levels of these five lncRNAs in ESCC cell lines (TE1, TE13, KYSE140 and KYSE150) and normal epithelial cell HET‐1A. Compared with HET‐1A, the expression of AC098818.2, RASSF8‐AS1 and LINC00958 was upregulated in the ESCC cell lines, while there was no significant difference in the expression of AL591721.1 and GMDS‐DT (Figure 2c). Meanwhile, by big data mining through ExoRbase, we found that AC098818.2, RASSF8‐AS1 and LINC00958 were significantly overexpressed in ESCC blood exosomes compared with healthy donors and other types of cancers (Figure S3 A‐C), indicating excellent specificity in ESCC screening. By searching in the GEPIA database which obtained expression data from TCGA, a gradually increased trend of RASSF8‐AS1 expression toward TNM stages was found (Figure S3 D). Furthermore, we detected expression level of the three candidates in 12 pairs of serum samples and corresponding cancerous tissues, and found moderate correlation for each lncRNA (Figure 2d‐f). Identification of candidate five lncRNAs expression. (a) The expression levels of the five candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) serum (n = 20) and normal serum samples (n = 20). (b) The expression levels of the five candidate lncRNAs in paired tumors and adjacent normal tissues (n = 30). (c) The expression levels of the five candidate lncRNAs in ESCC cell lines (TE1, TE13, KYSE140 and KYSE150) and the human esophageal epithelial cells line (HET‐1A). (d–f) Spearman's correlation analysis between expression levels of lncRNAs in tumor tissues and paired serum samples of ESCC. *p < 0.05, **p < 0.01 . The overall workflow of this study is illustrated in Figure 1a. In the discovery phase, we performed LncRNA microarray analysis on serum exosome samples from four preoperative ESCC patients and matched healthy controls. The top 100 differential lncRNAs identified in serum exosome samples from four preoperative ESCC patients and healthy control are shown in Figure 1b (FDR <0.005, Fold change >2, GEO accession number: GSE192662). Circos plots globally and genome‐widely displayed kinds of detected lncRNAs in ESCC and control serum samples (Figure 1c). A previous microarray comparing five pairs of tumor and paracancerous tissues (GSE89102) was also included for further analysis. To achieve more reliable aberrantly expressed lncRNAs, we adopted a more restrictive filtering criteria (FDR <0.005, fold change >5) for both datasets, and integrated the two datasets to finally obtain five (AC098818.2, RASSF8‐AS1, LINC00958, GMDS‐DT and AL591721.1) candidate genes with significant overexpression in ESCC serum samples and cancerous tissues (Figure 1d). Microarray screening results of differential lncRNAs and function prediction. (a) Experimental design overview. (b) Heatmap results of microarray analysis for four preoperative esophageal squamous cell carcinoma (ESCC) patients and matched normal healthy serum samples. (c) Circos plots of lncRNAs in the human genome (hg19). The outer tracks represent the cytoband ideogram of chromosome. For the two tracks, the outer one (blue) represents the levels of lncRNAs in normal tissues and the inner one (red) represents the levels in ESCC tissues. (d) Venn diagram showing consistent upregulation of five lncRNAs in both microarray analyses. (e, f) Signal pathway networks of mRNAs involved in lncRNAs‐mRNAs relationships. To investigate the potential role of the candidate lncRNAs in ESCC, we analyzed mRNA results from the same microarray chip derived from serum samples and performed pathway analysis (Figure S2 A–D). By lncRNAs‐mRNA coexpression network analysis, most of the candidate lncRNAs‐related mRNAs could be enriched in pathways such as MAPK, Ras, Wnt, metabolic pathways and HIF‐1α, which have been identified to be involved in ESCC carcinogenesis (Figure 1e,f). To validate the microarray data, we examined the expression of five candidate lncRNAs in a pilot cohort containing serum exosome samples (n = 20 for ESCC and healthy control separately), and 30 paired tissues, respectively. The qRT‐PCR results showed that the expression of AC098818.2, RASSF8‐AS1 and LINC00958 being significantly increased in ESCC patients' serum compared with healthy, while no significant difference in the expression of AL591721.1 and GMDS‐DT (Figure 2a). Compared with normal esophageal tissues, AC098818.2, RASSF8‐AS1 and LINC00958 expression was significantly upregulated in tumor tissues, whereas the expression level of GMDS‐DT was significantly downregulated, and no significant difference was seen in the expression of AL591721.1 (Figure 2b). Moreover, we examined the expression levels of these five lncRNAs in ESCC cell lines (TE1, TE13, KYSE140 and KYSE150) and normal epithelial cell HET‐1A. Compared with HET‐1A, the expression of AC098818.2, RASSF8‐AS1 and LINC00958 was upregulated in the ESCC cell lines, while there was no significant difference in the expression of AL591721.1 and GMDS‐DT (Figure 2c). Meanwhile, by big data mining through ExoRbase, we found that AC098818.2, RASSF8‐AS1 and LINC00958 were significantly overexpressed in ESCC blood exosomes compared with healthy donors and other types of cancers (Figure S3 A‐C), indicating excellent specificity in ESCC screening. By searching in the GEPIA database which obtained expression data from TCGA, a gradually increased trend of RASSF8‐AS1 expression toward TNM stages was found (Figure S3 D). Furthermore, we detected expression level of the three candidates in 12 pairs of serum samples and corresponding cancerous tissues, and found moderate correlation for each lncRNA (Figure 2d‐f). Identification of candidate five lncRNAs expression. (a) The expression levels of the five candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) serum (n = 20) and normal serum samples (n = 20). (b) The expression levels of the five candidate lncRNAs in paired tumors and adjacent normal tissues (n = 30). (c) The expression levels of the five candidate lncRNAs in ESCC cell lines (TE1, TE13, KYSE140 and KYSE150) and the human esophageal epithelial cells line (HET‐1A). (d–f) Spearman's correlation analysis between expression levels of lncRNAs in tumor tissues and paired serum samples of ESCC. *p < 0.05, **p < 0.01 . [SUBTITLE] Evaluation of stability of exosomal lncRNA in serum samples under different conditions [SUBSECTION] Given that stability is a key prerequisite for body fluid tumor biomarkers, we evaluated the stability of exosomal lncRNAs in serum samples under different conditions. Serum samples were subjected to extreme conditions, such as incubation at different temperatures for 3 h, repeated freeze–thaw cycles, RNase A digestion for 3 and 6 h, and exposure to acidic or alkaline environments. Interestingly, these treatments had little or no effect on the level of these serum lncRNAs (Figure 3), indicating the lncRNAs panel was valuable for clinical application. Stability of plasma lncRNAs under extreme conditions. (a) Incubation at different temperature for 3 h. (b) Repeated freeze–thaw cycles. (c) RNase A digestion for 3 and 6 h. (d) Exposure to acidic or alkaline environments. Given that stability is a key prerequisite for body fluid tumor biomarkers, we evaluated the stability of exosomal lncRNAs in serum samples under different conditions. Serum samples were subjected to extreme conditions, such as incubation at different temperatures for 3 h, repeated freeze–thaw cycles, RNase A digestion for 3 and 6 h, and exposure to acidic or alkaline environments. Interestingly, these treatments had little or no effect on the level of these serum lncRNAs (Figure 3), indicating the lncRNAs panel was valuable for clinical application. Stability of plasma lncRNAs under extreme conditions. (a) Incubation at different temperature for 3 h. (b) Repeated freeze–thaw cycles. (c) RNase A digestion for 3 and 6 h. (d) Exposure to acidic or alkaline environments. [SUBTITLE] Training phase: A panel of serum lncRNAs‐based diagnostic indicators Index I [SUBSECTION] To further investigate the diagnostic ability of the three candidate lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) for ESCC, we determined the relative expression levels of these three lncRNAs in serum samples of 40 ESCC patients and 40 healthy individuals and performed ROC curve analysis to evaluate the diagnostic capability for each lncRNA alone. The results showed that the expression levels of these lncRNAs were significantly elevated in ESCC patients (Figure 4a), with AUC values ranging from 0.69 to 0.78 (Figure 4b). Table S4 summarizes in detail the AUC values and 95% CI of each individual lncRNA. The optimal threshold for each lncRNA was determined by the Youden's index. 22 Serum lncRNA RASSF8‐AS1 was the best diagnostic marker, with AUC values of 0.78 and 95% CI: 0.67–0.88, respectively. Training phase: A panel of serum lncRNAs‐based diagnostic indicators Index I. (a) The expression levels of the three candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) serum (n = 40) and normal serum samples (n = 40). (b) Receiver operating characteristic (ROC) curves to evaluate the sensitivity and specificity of three candidate lncRNAs to discriminate ESCC from healthy controls. (c) Paired comparison of the three candidate lncRNAs level before and after surgery (n = 20). (d) ROC curves to evaluate the sensitivity and specificity of traditional tumor markers to discriminate ESCC from healthy control. (e) Comparison of the diagnostic performance of Index I and Index II. *p < 0.05, **p < 0.01. To evaluate whether the expression levels of the lncRNAs panel could be used to monitor tumor dynamics, we collected paired serum samples from 20 pre‐ and postoperative patients. By paired comparison, we found that all three lncRNAs levels descended significantly after surgery (Figure 4c), reflecting potential utility in tumor burden monitoring during the therapy period. Meanwhile, we explored the diagnostic value of several commonly used tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) in ESCC detection. The ROC curves showed that the AUC values of these markers ranged from 0.51 to 0.53 (Figure 4d), and Table S4 showed that CA199 had the best diagnostic ability with AUC values of 0.53 and 95% CI: 0.40–0.66, which is inferior to lncRNAs. Furthermore, we developed logistic regression models for each lncRNA and commonly used tumor markers, and calculated the regression coefficients separately. We subsequently used the regression coefficients as weights to establish the combined index. Interestingly, both combined indices outperformed single lncRNA alone or traditional biomarkers alone in terms of ESCC detection (Table S4). Next, we plotted the ROC curves and compared AUC values by the DeLong's test. 23 Index I had a significantly higher AUC value than Index II (p = 0.0045) with an AUC value of 0.84 (95% CI: 0.74–0.91), showing remarkably better diagnostic performance (Figure 4e and Table S4; p = 0.0045). The above results suggested that the combination of these three lncRNAs exhibited superior discriminatory power than traditional biomarkers. To further investigate the diagnostic ability of the three candidate lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) for ESCC, we determined the relative expression levels of these three lncRNAs in serum samples of 40 ESCC patients and 40 healthy individuals and performed ROC curve analysis to evaluate the diagnostic capability for each lncRNA alone. The results showed that the expression levels of these lncRNAs were significantly elevated in ESCC patients (Figure 4a), with AUC values ranging from 0.69 to 0.78 (Figure 4b). Table S4 summarizes in detail the AUC values and 95% CI of each individual lncRNA. The optimal threshold for each lncRNA was determined by the Youden's index. 22 Serum lncRNA RASSF8‐AS1 was the best diagnostic marker, with AUC values of 0.78 and 95% CI: 0.67–0.88, respectively. Training phase: A panel of serum lncRNAs‐based diagnostic indicators Index I. (a) The expression levels of the three candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) serum (n = 40) and normal serum samples (n = 40). (b) Receiver operating characteristic (ROC) curves to evaluate the sensitivity and specificity of three candidate lncRNAs to discriminate ESCC from healthy controls. (c) Paired comparison of the three candidate lncRNAs level before and after surgery (n = 20). (d) ROC curves to evaluate the sensitivity and specificity of traditional tumor markers to discriminate ESCC from healthy control. (e) Comparison of the diagnostic performance of Index I and Index II. *p < 0.05, **p < 0.01. To evaluate whether the expression levels of the lncRNAs panel could be used to monitor tumor dynamics, we collected paired serum samples from 20 pre‐ and postoperative patients. By paired comparison, we found that all three lncRNAs levels descended significantly after surgery (Figure 4c), reflecting potential utility in tumor burden monitoring during the therapy period. Meanwhile, we explored the diagnostic value of several commonly used tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) in ESCC detection. The ROC curves showed that the AUC values of these markers ranged from 0.51 to 0.53 (Figure 4d), and Table S4 showed that CA199 had the best diagnostic ability with AUC values of 0.53 and 95% CI: 0.40–0.66, which is inferior to lncRNAs. Furthermore, we developed logistic regression models for each lncRNA and commonly used tumor markers, and calculated the regression coefficients separately. We subsequently used the regression coefficients as weights to establish the combined index. Interestingly, both combined indices outperformed single lncRNA alone or traditional biomarkers alone in terms of ESCC detection (Table S4). Next, we plotted the ROC curves and compared AUC values by the DeLong's test. 23 Index I had a significantly higher AUC value than Index II (p = 0.0045) with an AUC value of 0.84 (95% CI: 0.74–0.91), showing remarkably better diagnostic performance (Figure 4e and Table S4; p = 0.0045). The above results suggested that the combination of these three lncRNAs exhibited superior discriminatory power than traditional biomarkers. [SUBTITLE] Validation phase: The lncRNAs based model can distinguish ESCC from healthy control and the levels of lncRNA panel correlated with clinicopathological features [SUBSECTION] We further analyzed the serum lncRNAs levels in 80 ESCC patients and matched healthy samples. Being consistent with the results of the training phase, the expression levels of lncRNAs were significantly higher in ESCC patients than in healthy individuals (Figure 5a). Then, we plotted ROC curves to compare the diagnostic performance between lncRNAs and classic biomarkers, with maximum AUC values of 0.86 and 0.54 for lncRNAs and classic biomarkers, respectively (Figure 5b,c). Table S4 showed in detail the AUC values and 95% CI of lncRNAs and tumor markers during the training phase. Validation phase: A panel of serum lncRNAs‐based diagnostic indicators index I. (a) The expression levels of the three candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) (n = 80) and control's serum samples (n = 80). (b) Receiver operating characteristic (ROC) curves of candidate lncRNAs to discriminate ESCC from healthy control. (c) ROC curves of traditional tumor markers. (d) Comparison of the diagnostic performance of Index I and Index II. (e) Kaplan–Meier survival curves of ESCC patients with low and high Index I.**p < 0.01. Being consistent with the results of the training phase, the lncRNAs‐based Index I obtained the greatest diagnostic performance with an AUC value of 0.86 (95% CI: 0.81–0.92) (Table S4). In addition, the AUC values of lncRNAs‐based Index I were significantly outperformed to that from Index II (Figure 5d and Table S4; p < 0.001). To investigate Index I related clinicopathological factors, we collected ESCC patients from the discovery, training, and validation phases and classified them into high and low levels using the median of the Index I index as the threshold value. As shown in Table 1, higher Index I levels were associated with greater depth of infiltration (p = 0.0267), lymph node metastasis (p = 0.0012), advanced tumor stage (p = 0.0040), and poor prognosis (Figure 5e; p < 0.001). The correlation between the serum Index I expression and clinicopathological factors of ESCC Abbreviations: ESCC, esophageal squamous cell carcinoma; —, no data available. Bold values means p‐value < 0.05. We further analyzed the serum lncRNAs levels in 80 ESCC patients and matched healthy samples. Being consistent with the results of the training phase, the expression levels of lncRNAs were significantly higher in ESCC patients than in healthy individuals (Figure 5a). Then, we plotted ROC curves to compare the diagnostic performance between lncRNAs and classic biomarkers, with maximum AUC values of 0.86 and 0.54 for lncRNAs and classic biomarkers, respectively (Figure 5b,c). Table S4 showed in detail the AUC values and 95% CI of lncRNAs and tumor markers during the training phase. Validation phase: A panel of serum lncRNAs‐based diagnostic indicators index I. (a) The expression levels of the three candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) (n = 80) and control's serum samples (n = 80). (b) Receiver operating characteristic (ROC) curves of candidate lncRNAs to discriminate ESCC from healthy control. (c) ROC curves of traditional tumor markers. (d) Comparison of the diagnostic performance of Index I and Index II. (e) Kaplan–Meier survival curves of ESCC patients with low and high Index I.**p < 0.01. Being consistent with the results of the training phase, the lncRNAs‐based Index I obtained the greatest diagnostic performance with an AUC value of 0.86 (95% CI: 0.81–0.92) (Table S4). In addition, the AUC values of lncRNAs‐based Index I were significantly outperformed to that from Index II (Figure 5d and Table S4; p < 0.001). To investigate Index I related clinicopathological factors, we collected ESCC patients from the discovery, training, and validation phases and classified them into high and low levels using the median of the Index I index as the threshold value. As shown in Table 1, higher Index I levels were associated with greater depth of infiltration (p = 0.0267), lymph node metastasis (p = 0.0012), advanced tumor stage (p = 0.0040), and poor prognosis (Figure 5e; p < 0.001). The correlation between the serum Index I expression and clinicopathological factors of ESCC Abbreviations: ESCC, esophageal squamous cell carcinoma; —, no data available. Bold values means p‐value < 0.05. [SUBTITLE] Prognostic significance of three serum lncRNAs panel [SUBSECTION] We then analyzed the associations between 140 ESCC mortality and both three serum‐derived lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) and classic tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) using a forward conditional logistic regression model. As shown in Table 2, in the univariable analysis, tumor size, RASSF8‐AS1 expression and LINC00958 expression were associated with ESCC mortality. However, in the multivariable analysis, only RASSF8‐AS1 expression and CYFRA21‐1 expression were independently associated with ESCC mortality. The long‐term prognostic values of three serum‐derived lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) and classic tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) were analyzed using a Cox regression model and the results are listed in Table 3. In the univariable analysis, tumor depth, lymph node metastasis and RASSF8‐AS1 expression were significantly associated with long‐term mortality. However, in the multivariable analysis, only RASSF8‐AS1 expression was independently associated with long‐term mortality. In conclusion, RASSF8‐AS1 expression was an independent prognostic factor for both short‐ and long‐term outcomes of ESCC. Associations between three serum‐derived lncRNAs, classic tumor markers and ESCC mortality Abbreviations: CI, confidence interval; ESCC, esophageal squamous cell carcinoma; OR, odds ratio; —, no data available. Univariate and multivariate Cox analysis of overall survival in ESCC patients Abbreviations: CI, confidence interval; ESCC, esophageal squamous cell carcinoma; HR, hazard ration; —, no data available. We then analyzed the associations between 140 ESCC mortality and both three serum‐derived lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) and classic tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) using a forward conditional logistic regression model. As shown in Table 2, in the univariable analysis, tumor size, RASSF8‐AS1 expression and LINC00958 expression were associated with ESCC mortality. However, in the multivariable analysis, only RASSF8‐AS1 expression and CYFRA21‐1 expression were independently associated with ESCC mortality. The long‐term prognostic values of three serum‐derived lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) and classic tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) were analyzed using a Cox regression model and the results are listed in Table 3. In the univariable analysis, tumor depth, lymph node metastasis and RASSF8‐AS1 expression were significantly associated with long‐term mortality. However, in the multivariable analysis, only RASSF8‐AS1 expression was independently associated with long‐term mortality. In conclusion, RASSF8‐AS1 expression was an independent prognostic factor for both short‐ and long‐term outcomes of ESCC. Associations between three serum‐derived lncRNAs, classic tumor markers and ESCC mortality Abbreviations: CI, confidence interval; ESCC, esophageal squamous cell carcinoma; OR, odds ratio; —, no data available. Univariate and multivariate Cox analysis of overall survival in ESCC patients Abbreviations: CI, confidence interval; ESCC, esophageal squamous cell carcinoma; HR, hazard ration; —, no data available. [SUBTITLE] In vitro analysis of the oncogenic potential of RASSF8‐AS1 and target scanning [SUBSECTION] As lncRNA RASSF8‐AS1 showed good performance for ESCC diagnosis and RASSF8‐AS1 expression was an independent prognostic factor for both short‐ and long‐term outcomes of ESCC. We speculated that RASSF8‐AS1 plays an oncogenic role in ESCC. We then determined that higher RASSF8‐AS1 was associated with clinicopathological factors (Table 4), such as larger tumor size (p < 0.0001), greater depth of infiltration (p < 0.0001), lymph node metastasis (p = 0.0278), and advanced tumor stage (p = 0.0110). The expression of RASSF8‐AS1 tended to decrease significantly after tumor resection surgery in ESCC patients (Figure 4c), and RASSF8‐AS1 was found to be significantly higher in ESCC in the mid‐late stage than in stage I through the GEPIA database (Figure S3 D). The above data suggested that RASSF8‐AS1 plays a protumor role for ESCC. The correlation between the serum RASSF8‐AS1 expression and clinicopathological factors of ESCC Abbreviation: ESCC, esophageal squamous cell carcinoma; —, no data available. Bold values means p‐value < 0.05. Subsequently, we explored the oncogenic function of RASSF8‐AS1 by knocking down RASSF8‐AS1 in TE1 and TE13 cells (Figure 6a). In vitro assays showed that the knockdown of RASSF8‐AS1 inhibited cell proliferation (Figure 6b,c), motility and invasiveness (Figure 6d), respectively. RASSF8‐AS1 promotes esophageal squamous cell carcinoma (ESCC) cell proliferation, migration and invasion in vitro. (a) The mRNA expression of RASSF8‐AS1 after transfection by two siRNAs. (b, c) Assessment of knockdown of RASSF8‐AS1 on cell growth of TE1 and TE13 cells. (d) Assessment of knockdown of RASSF8‐AS1 on cell migration and invasion. (e) The mRNA expression of RASSF8‐AS1 in normal tissues and ESCC tissues. (f) Kaplan–Meier survival curves of ESCC patients with low and high RASSF8‐AS1 expression. *p < 0.05, **p < 0.01, ***p < 0.001. In addition, we performed qRT‐PCR on 60 pairs of tumor and adjacent normal tissues, and found that RASSF8‐AS1 was likewise markedly elevated in ESCC tissues (Figure 6e). Next, we divided the 60 ESCC samples into RASSF8‐AS1 low expression group (n = 30) and high expression group (n = 30) according to the median level of RASSF8‐AS1 to explore the prognostic significance of RASSF8‐AS1 in ESCC. Kaplan–Meier analysis showed that patients with high RASSF8‐AS1 expression had a better dismal OS (Figure 6f). Finally, we screened RASSF8‐AS1 targets by RNA‐sequencing and performed gene ontology (GO) and pathway enrichment analysis. The result found 303 positively regulated (Figure S4 C, D) and 430 negatively regulated targets (Figure 4e,f). GO results suggested RASSF8‐AS1 was positively involved, mainly in immune response and cell migration, and KEGG pathway analysis indicated the positively regulated targets mostly enriched in cell metabolism (Figure S4 E, F), all of which have been proven to be essential for tumor progression. As lncRNA RASSF8‐AS1 showed good performance for ESCC diagnosis and RASSF8‐AS1 expression was an independent prognostic factor for both short‐ and long‐term outcomes of ESCC. We speculated that RASSF8‐AS1 plays an oncogenic role in ESCC. We then determined that higher RASSF8‐AS1 was associated with clinicopathological factors (Table 4), such as larger tumor size (p < 0.0001), greater depth of infiltration (p < 0.0001), lymph node metastasis (p = 0.0278), and advanced tumor stage (p = 0.0110). The expression of RASSF8‐AS1 tended to decrease significantly after tumor resection surgery in ESCC patients (Figure 4c), and RASSF8‐AS1 was found to be significantly higher in ESCC in the mid‐late stage than in stage I through the GEPIA database (Figure S3 D). The above data suggested that RASSF8‐AS1 plays a protumor role for ESCC. The correlation between the serum RASSF8‐AS1 expression and clinicopathological factors of ESCC Abbreviation: ESCC, esophageal squamous cell carcinoma; —, no data available. Bold values means p‐value < 0.05. Subsequently, we explored the oncogenic function of RASSF8‐AS1 by knocking down RASSF8‐AS1 in TE1 and TE13 cells (Figure 6a). In vitro assays showed that the knockdown of RASSF8‐AS1 inhibited cell proliferation (Figure 6b,c), motility and invasiveness (Figure 6d), respectively. RASSF8‐AS1 promotes esophageal squamous cell carcinoma (ESCC) cell proliferation, migration and invasion in vitro. (a) The mRNA expression of RASSF8‐AS1 after transfection by two siRNAs. (b, c) Assessment of knockdown of RASSF8‐AS1 on cell growth of TE1 and TE13 cells. (d) Assessment of knockdown of RASSF8‐AS1 on cell migration and invasion. (e) The mRNA expression of RASSF8‐AS1 in normal tissues and ESCC tissues. (f) Kaplan–Meier survival curves of ESCC patients with low and high RASSF8‐AS1 expression. *p < 0.05, **p < 0.01, ***p < 0.001. In addition, we performed qRT‐PCR on 60 pairs of tumor and adjacent normal tissues, and found that RASSF8‐AS1 was likewise markedly elevated in ESCC tissues (Figure 6e). Next, we divided the 60 ESCC samples into RASSF8‐AS1 low expression group (n = 30) and high expression group (n = 30) according to the median level of RASSF8‐AS1 to explore the prognostic significance of RASSF8‐AS1 in ESCC. Kaplan–Meier analysis showed that patients with high RASSF8‐AS1 expression had a better dismal OS (Figure 6f). Finally, we screened RASSF8‐AS1 targets by RNA‐sequencing and performed gene ontology (GO) and pathway enrichment analysis. The result found 303 positively regulated (Figure S4 C, D) and 430 negatively regulated targets (Figure 4e,f). GO results suggested RASSF8‐AS1 was positively involved, mainly in immune response and cell migration, and KEGG pathway analysis indicated the positively regulated targets mostly enriched in cell metabolism (Figure S4 E, F), all of which have been proven to be essential for tumor progression.
null
null
[ "INTRODUCTION", "Patients and samples", "Exosome extraction and identification", "\nRNA extraction and quantitative real‐time PCR (qRT‐PCR)", "Microarray screening and analysis", "Cell culture", "Stability analysis for exosomal lncRNAs panel", "Transfection and RNA interfering", "Cell proliferation, migration, and invasion assays", "\nRNA sequencing and enrichment analysis", "Statistical analysis", "Identification of exosomes in serum", "Discovery phase: Genome‐wide profiling to identify differentially expressed lncRNAs between ESCC and healthy individual controls", "Evaluation of stability of exosomal lncRNA in serum samples under different conditions", "Training phase: A panel of serum lncRNAs‐based diagnostic indicators Index I", "Validation phase: The lncRNAs based model can distinguish ESCC from healthy control and the levels of lncRNA panel correlated with clinicopathological features", "Prognostic significance of three serum lncRNAs panel", "In vitro analysis of the oncogenic potential of RASSF8‐AS1 and target scanning", "FUNDING INFORMATION" ]
[ "Esophageal cancer is the eighth most common cancer and the sixth leading cause of malignancy deaths worldwide.\n1\n Squamous cell carcinoma is the main pathology type in China. The current standard for esophageal squamous cell carcinoma (ESCC) diagnosis depends on endoscopy and pathological biopsy.\n2\n However, the operation is relatively invasive and uncomfortable, resulting in delay or missing the best chance for most ESCC diagnosis and therapy. Liquid biopsy provides an alternative way of ESCC screening, but the traditional biomarkers used in the diagnosis of ESCC, such as serum squamous cell carcinoma antigen (SCC), cancer antigen 125 (CA125), carcinoembryonic antigen (CEA), and cytokeratin‐19‐fragment (CYFRA21‐1),\n3\n, \n4\n lack enough sensitivity and specificity, which limits their clinical application. For example, SCC may be also applied in other types of squamous cell carcinoma, such as lung, head and neck, and cervical squamous cell carcinoma. CEA is mainly used for the detection of colorectal cancer, while CYFRA21‐1 is usually used as a biomarker for diverse subtypes of lung and bladder cancer.\nExosomes are small (50–150 nm) membrane vesicles inside the cell, which may fuse with the membrane and are then released into the circulation.\n5\n, \n6\n They are detected in various body fluids such as serum, plasma, urine and saliva.\n7\n, \n8\n Exosomes execute communication functions intercellularly by transporting various biomolecules (such as DNAs, RNAs and proteins),\n9\n, \n10\n and the contents are protected by a phospholipid bilayer, making them suitable for liquid biopsy. Long noncoding RNAs (LncRNAs) is one of the exosome cargos,\n11\n and previous studies considered that lncRNAs in exosomes may be more stable for exosome protection against RNase.\n12\n, \n13\n LncRNAs detected in body fluids have been shown to be ideal noninvasive biomarkers for cancer diagnosis and prognosis. For example, lncRNA HOTAIR and SBF2‐AS1 in serum exosomes have been implicated for the diagnosis and prognosis assessment of glioblastoma.\n14\n, \n15\n Similarly, Xu et al.\n16\n found in an independent cohort study that ENSG00000258332.1 and LINC00635 in serum had higher sensitivity and specificity for the diagnosis of hepatocellular carcinoma in combination with AFP. Recently, Tao et al.\n17\n found that the expression levels of TBILA and AGAP2‐AS1 in the serum of NSCLC patients were significantly increased, and the combination with serum tumor biomarkers could further improve the diagnostic accuracy of NSCLC patients. Compared with the increasing evidence for exosomal lncRNAs in other types of cancer, little is known about their roles in ESCC. Furthermore, it has previously been reported that a joint model based on the union of multiple biomarkers may improve the diagnostic power, which prompted our study design.\n18\n, \n19\n\n\nHere, we used a multistep study to investigate the possibility that novel serum exosomal lncRNAs act as potential biomarkers for ESCC detection. We selected candidate lncRNAs by integrating microarray results derived from serum and tissue samples separately, as a previous study reported that serum exosomal lncRNAs may be derived from tumor tissues. We deduced that the lncRNAs simultaneously overexpressed in serum and tissue samples may be more likely to meet the criteria. Finally, a diagnostic model based on three lncRNAs panel was established and the association of the novel panel with clinicopathological features or prognosis was also evaluated. Furthermore, we found involvement of candidate lncRNA RASSF8‐AS1 in ESCC malignant phenotypes, and the combined results provide new insights into ESCC diagnosis and target therapy.", "From 2017 to 2021, 140 ESCC patients and 140 healthy individuals were randomly recruited from Jinling Hospital of Nanjing Medical University, each of whom provided 5 ml of whole blood at indicated timepoints. Neither patient received preoperative chemo−/radiotherapy, and the overall survival (OS) was measured from the date of diagnosis to the date of death due to any cause or the date of the most recent follow‐up. All patients were followed every 3 months for the first year, every 6 months for the next 2 years, and then annually. The end of the follow‐up was November 31, 2021. The study was approved by the ethics committee of Jinling hospital, and all enrolled participants signed informed consent. All serum samples were stored at −80°C until use for total RNA extraction. The pathological stage was determined according to the eighth edition of the Union for International Cancer Control TNM staging system.", "A total of 1.5 ml of serum was centrifuged at 2000 × g, 4°C for 30 min, and the supernatant was then recentrifuged at 10 000 × g, 4°C for 45 min. The supernatant was then collected and filtered through a membrane of 0.22 μm pore size. The filtrate was collected and further subjected to ultracentrifugation at 10 000 × g, 4°C for 70 min. The precipitate was resuspended in 10 ml of precooling 1 × PBS, and ultracentrifugation repeated. Finally, the precipitate was resuspended in 1.5 ml of precooling 1 × PBS. The isolated exosomes were characterized using transmission electron microscopy (TEM). Exosome concentration‐particle size testing (nanoparticle tracking analysis [NTA]) was performed by Zeta View (Particle Metrix Ltd.).", "RNA was extracted from exosomes or tissues using an exosome RNA isolation kit according to protocols (Invitrogen). The extracted RNA was synthesized into cDNA by the PrimeScript RT Master Mix (Takara). RT‐qPCR was performed to detect the relative RNA levels of target genes. β‐actin was used as an internal reference gene for analysis. The relative expression levels of the lncRNAs were calculated using 2−∆∆Ct.\n20\n, \n21\n The sequences of primers used for qRT‐PCR of the lncRNAs are listed in Table S1.", "Microarray screening was performed with Arraystar LncRNA_8 × 60 k (Arraystar), which contains both lncRNA and mRNA probes. Sample labeling and array hybridization were performed according to the Agilent One‐Color Microarray‐Based Gene Expression Analysis protocol (Agilent Technology). Agilent Feature Extraction software (version 11.0.1.1) was used to analyze acquired array images. Quantile normalization and subsequent data processing were performed using the GeneSpring GX version 12.1 software package (Agilent Technologies). After quantile normalization of the raw data, LncRNAs and mRNAs that at least four out of eight samples have flags in Present or Marginal (“All Targets Value”) were chosen for further data analysis. Differentially expressed LncRNAs and mRNAs with statistical significance between the two groups were identified through p‐value/FDR filtering.", "Esophageal squamous cell carcinoma cell lines (TE1, TE13, KYSE150, KYSE140) and normal human esophageal epithelial cells (HET‐1A) were obtained from the Shanghai Institutes for Biological Sciences (Shanghai, China). These cells were cultured in 1640 medium (KeyGene) containing 10% fetal bovine serum (FBS) (KeyGene), at 37°C in a 5% CO2 atmosphere.", "Exosome samples (n = 15) were allocated to aliquots for different processing, including storage under different temperature, treated with RNase A, and subjected to freeze–thaw cycles and acidic/alkaline environments, to test to the stability of exosomal lncRNAs, which is essential for clinical use.", "Cells were seeded on six‐well plates (2 × 105/well) with 1640 medium supplemented with 10% FBS overnight. Then, siRNA was transfected at a final concentration of 100 nM using lipofectamine 2000 (Invitrogen), and 24 h post transfection cells were harvested and subsequently RNA was extracted by Trizol reagent and the interfering efficiency was determined by RT‐qPCR. Sequences of siRNAs and negative control are provided in Table S2.", "Transfected cells were collected and grown in 96‐well plates (2 × 103 cells per well) overnight and cell proliferation was detected using cell counting kit‐8 (CCK‐8). The reaction products were measured at 450 nm according to the manufacturer's instructions. For migration/invasion assays, 5 × 104 cells were incubated using Transwell kits (8 mm PET, 24‐well Millicell) or matrix‐coated inserts (BD Biosciences). After 24 or 48 h of incubation, cells were fixed with 4% paraformaldehyde and stained with crystal violet (Beyotim). Imaging and counting of migrated cells were conducted by microscopy.", "Cell transfection and RNA interfering were performed as described above. Then, 24 h post transfection, cells were harvested and RNA was extracted using Trizol reagent (Invitrogen). RNA‐seq libraries were prepared using the Illumina RNA‐seq preparation kit and sequenced on illumina nova seq 6000 sequencer. The differentially expressed RNAs (fold change >1.5, p < 0.05) were retrieved and subjected to gene ontology and KEGG pathway analysis.", "All data were statistically analyzed with SPSS 26.0 (Chicago) and visualized with GraphPad Prism version 8.0 (GraphPad Software). Differences in lncRNAs expression between groups were determined using the Mann–Whitney unpaired t‐test or paired t‐test. The χ2 or Fisher's exact test was used to compare the clinical characteristics of the groups. Receiver operating characteristic (ROC) curves were used to assess the diagnostic performance of selected biomarkers. The optimal cutoff point was determined using Youden's index. DeLong's test was used to evaluate the statistical significance when comparing differences of AUCs. Using the binary status of enrolled participants (patients or controls) as the dependent variable, the regression coefficient for each lncRNA was estimated by a univariate logistic regression model, and was used as the weighting to construct the diagnostic index. The diagnostic index developed in the training phase was also directly applied to the validation phase to evaluate the clinical utility of the identified lncRNAs individually or in combination. A forward conditional logistic regression model was used to analyze the association between selected biomarkers and ESCC mortality. The long‐term prognostic values of selected biomarkers were evaluated using Cox's hazard regression model and Kaplan–Meier curve analysis. Univariate analysis was used to explore clinicopathological factors (i.e., tumor depth, stage, size) associated with OS. Factors with p‐value <0.05 were selected for the multivariate analysis with Cox's proportional hazard regression model, hazard ratio and 95% confidence interval (CI) were calculated. All p‐values <0.05 were considered statistically significant.", "A total of 280 participants, including 140 ESCC patients and 140 healthy controls were enrolled in the study, as shown in Table S3. To verify whether the exosomes were successfully isolated from serum, we characterized the isolated exosomes using TEM and NTA. The results showed that the isolated exosomes had a bilayer membrane structure (Figure S1 A), and the particle size distribution of exosomes was about 30–150 nm in diameter (Figure S1 B), meeting the criteria of exosome patterns.", "The overall workflow of this study is illustrated in Figure 1a. In the discovery phase, we performed LncRNA microarray analysis on serum exosome samples from four preoperative ESCC patients and matched healthy controls. The top 100 differential lncRNAs identified in serum exosome samples from four preoperative ESCC patients and healthy control are shown in Figure 1b (FDR <0.005, Fold change >2, GEO accession number: GSE192662). Circos plots globally and genome‐widely displayed kinds of detected lncRNAs in ESCC and control serum samples (Figure 1c). A previous microarray comparing five pairs of tumor and paracancerous tissues (GSE89102) was also included for further analysis. To achieve more reliable aberrantly expressed lncRNAs, we adopted a more restrictive filtering criteria (FDR <0.005, fold change >5) for both datasets, and integrated the two datasets to finally obtain five (AC098818.2, RASSF8‐AS1, LINC00958, GMDS‐DT and AL591721.1) candidate genes with significant overexpression in ESCC serum samples and cancerous tissues (Figure 1d).\nMicroarray screening results of differential lncRNAs and function prediction. (a) Experimental design overview. (b) Heatmap results of microarray analysis for four preoperative esophageal squamous cell carcinoma (ESCC) patients and matched normal healthy serum samples. (c) Circos plots of lncRNAs in the human genome (hg19). The outer tracks represent the cytoband ideogram of chromosome. For the two tracks, the outer one (blue) represents the levels of lncRNAs in normal tissues and the inner one (red) represents the levels in ESCC tissues. (d) Venn diagram showing consistent upregulation of five lncRNAs in both microarray analyses. (e, f) Signal pathway networks of mRNAs involved in lncRNAs‐mRNAs relationships.\nTo investigate the potential role of the candidate lncRNAs in ESCC, we analyzed mRNA results from the same microarray chip derived from serum samples and performed pathway analysis (Figure S2 A–D). By lncRNAs‐mRNA coexpression network analysis, most of the candidate lncRNAs‐related mRNAs could be enriched in pathways such as MAPK, Ras, Wnt, metabolic pathways and HIF‐1α, which have been identified to be involved in ESCC carcinogenesis (Figure 1e,f).\nTo validate the microarray data, we examined the expression of five candidate lncRNAs in a pilot cohort containing serum exosome samples (n = 20 for ESCC and healthy control separately), and 30 paired tissues, respectively. The qRT‐PCR results showed that the expression of AC098818.2, RASSF8‐AS1 and LINC00958 being significantly increased in ESCC patients' serum compared with healthy, while no significant difference in the expression of AL591721.1 and GMDS‐DT (Figure 2a). Compared with normal esophageal tissues, AC098818.2, RASSF8‐AS1 and LINC00958 expression was significantly upregulated in tumor tissues, whereas the expression level of GMDS‐DT was significantly downregulated, and no significant difference was seen in the expression of AL591721.1 (Figure 2b). Moreover, we examined the expression levels of these five lncRNAs in ESCC cell lines (TE1, TE13, KYSE140 and KYSE150) and normal epithelial cell HET‐1A. Compared with HET‐1A, the expression of AC098818.2, RASSF8‐AS1 and LINC00958 was upregulated in the ESCC cell lines, while there was no significant difference in the expression of AL591721.1 and GMDS‐DT (Figure 2c). Meanwhile, by big data mining through ExoRbase, we found that AC098818.2, RASSF8‐AS1 and LINC00958 were significantly overexpressed in ESCC blood exosomes compared with healthy donors and other types of cancers (Figure S3 A‐C), indicating excellent specificity in ESCC screening. By searching in the GEPIA database which obtained expression data from TCGA, a gradually increased trend of RASSF8‐AS1 expression toward TNM stages was found (Figure S3 D). Furthermore, we detected expression level of the three candidates in 12 pairs of serum samples and corresponding cancerous tissues, and found moderate correlation for each lncRNA (Figure 2d‐f).\nIdentification of candidate five lncRNAs expression. (a) The expression levels of the five candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) serum (n = 20) and normal serum samples (n = 20). (b) The expression levels of the five candidate lncRNAs in paired tumors and adjacent normal tissues (n = 30). (c) The expression levels of the five candidate lncRNAs in ESCC cell lines (TE1, TE13, KYSE140 and KYSE150) and the human esophageal epithelial cells line (HET‐1A). (d–f) Spearman's correlation analysis between expression levels of lncRNAs in tumor tissues and paired serum samples of ESCC. *p < 0.05, **p < 0.01 .", "Given that stability is a key prerequisite for body fluid tumor biomarkers, we evaluated the stability of exosomal lncRNAs in serum samples under different conditions. Serum samples were subjected to extreme conditions, such as incubation at different temperatures for 3 h, repeated freeze–thaw cycles, RNase A digestion for 3 and 6 h, and exposure to acidic or alkaline environments. Interestingly, these treatments had little or no effect on the level of these serum lncRNAs (Figure 3), indicating the lncRNAs panel was valuable for clinical application.\nStability of plasma lncRNAs under extreme conditions. (a) Incubation at different temperature for 3 h. (b) Repeated freeze–thaw cycles. (c) RNase A digestion for 3 and 6 h. (d) Exposure to acidic or alkaline environments.", "To further investigate the diagnostic ability of the three candidate lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) for ESCC, we determined the relative expression levels of these three lncRNAs in serum samples of 40 ESCC patients and 40 healthy individuals and performed ROC curve analysis to evaluate the diagnostic capability for each lncRNA alone. The results showed that the expression levels of these lncRNAs were significantly elevated in ESCC patients (Figure 4a), with AUC values ranging from 0.69 to 0.78 (Figure 4b). Table S4 summarizes in detail the AUC values and 95% CI of each individual lncRNA. The optimal threshold for each lncRNA was determined by the Youden's index.\n22\n Serum lncRNA RASSF8‐AS1 was the best diagnostic marker, with AUC values of 0.78 and 95% CI: 0.67–0.88, respectively.\nTraining phase: A panel of serum lncRNAs‐based diagnostic indicators Index I. (a) The expression levels of the three candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) serum (n = 40) and normal serum samples (n = 40). (b) Receiver operating characteristic (ROC) curves to evaluate the sensitivity and specificity of three candidate lncRNAs to discriminate ESCC from healthy controls. (c) Paired comparison of the three candidate lncRNAs level before and after surgery (n = 20). (d) ROC curves to evaluate the sensitivity and specificity of traditional tumor markers to discriminate ESCC from healthy control. (e) Comparison of the diagnostic performance of Index I and Index II. *p < 0.05, **p < 0.01.\nTo evaluate whether the expression levels of the lncRNAs panel could be used to monitor tumor dynamics, we collected paired serum samples from 20 pre‐ and postoperative patients. By paired comparison, we found that all three lncRNAs levels descended significantly after surgery (Figure 4c), reflecting potential utility in tumor burden monitoring during the therapy period.\nMeanwhile, we explored the diagnostic value of several commonly used tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) in ESCC detection. The ROC curves showed that the AUC values of these markers ranged from 0.51 to 0.53 (Figure 4d), and Table S4 showed that CA199 had the best diagnostic ability with AUC values of 0.53 and 95% CI: 0.40–0.66, which is inferior to lncRNAs.\nFurthermore, we developed logistic regression models for each lncRNA and commonly used tumor markers, and calculated the regression coefficients separately. We subsequently used the regression coefficients as weights to establish the combined index. Interestingly, both combined indices outperformed single lncRNA alone or traditional biomarkers alone in terms of ESCC detection (Table S4). Next, we plotted the ROC curves and compared AUC values by the DeLong's test.\n23\n Index I had a significantly higher AUC value than Index II (p = 0.0045) with an AUC value of 0.84 (95% CI: 0.74–0.91), showing remarkably better diagnostic performance (Figure 4e and Table S4; p = 0.0045). The above results suggested that the combination of these three lncRNAs exhibited superior discriminatory power than traditional biomarkers.", "We further analyzed the serum lncRNAs levels in 80 ESCC patients and matched healthy samples. Being consistent with the results of the training phase, the expression levels of lncRNAs were significantly higher in ESCC patients than in healthy individuals (Figure 5a). Then, we plotted ROC curves to compare the diagnostic performance between lncRNAs and classic biomarkers, with maximum AUC values of 0.86 and 0.54 for lncRNAs and classic biomarkers, respectively (Figure 5b,c). Table S4 showed in detail the AUC values and 95% CI of lncRNAs and tumor markers during the training phase.\nValidation phase: A panel of serum lncRNAs‐based diagnostic indicators index I. (a) The expression levels of the three candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) (n = 80) and control's serum samples (n = 80). (b) Receiver operating characteristic (ROC) curves of candidate lncRNAs to discriminate ESCC from healthy control. (c) ROC curves of traditional tumor markers. (d) Comparison of the diagnostic performance of Index I and Index II. (e) Kaplan–Meier survival curves of ESCC patients with low and high Index I.**p < 0.01.\nBeing consistent with the results of the training phase, the lncRNAs‐based Index I obtained the greatest diagnostic performance with an AUC value of 0.86 (95% CI: 0.81–0.92) (Table S4). In addition, the AUC values of lncRNAs‐based Index I were significantly outperformed to that from Index II (Figure 5d and Table S4; p < 0.001).\nTo investigate Index I related clinicopathological factors, we collected ESCC patients from the discovery, training, and validation phases and classified them into high and low levels using the median of the Index I index as the threshold value. As shown in Table 1, higher Index I levels were associated with greater depth of infiltration (p = 0.0267), lymph node metastasis (p = 0.0012), advanced tumor stage (p = 0.0040), and poor prognosis (Figure 5e; p < 0.001).\nThe correlation between the serum Index I expression and clinicopathological factors of ESCC\nAbbreviations: ESCC, esophageal squamous cell carcinoma; —, no data available.\nBold values means p‐value < 0.05.", "We then analyzed the associations between 140 ESCC mortality and both three serum‐derived lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) and classic tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) using a forward conditional logistic regression model. As shown in Table 2, in the univariable analysis, tumor size, RASSF8‐AS1 expression and LINC00958 expression were associated with ESCC mortality. However, in the multivariable analysis, only RASSF8‐AS1 expression and CYFRA21‐1 expression were independently associated with ESCC mortality. The long‐term prognostic values of three serum‐derived lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) and classic tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) were analyzed using a Cox regression model and the results are listed in Table 3. In the univariable analysis, tumor depth, lymph node metastasis and RASSF8‐AS1 expression were significantly associated with long‐term mortality. However, in the multivariable analysis, only RASSF8‐AS1 expression was independently associated with long‐term mortality. In conclusion, RASSF8‐AS1 expression was an independent prognostic factor for both short‐ and long‐term outcomes of ESCC.\nAssociations between three serum‐derived lncRNAs, classic tumor markers and ESCC mortality\nAbbreviations: CI, confidence interval; ESCC, esophageal squamous cell carcinoma; OR, odds ratio; —, no data available.\nUnivariate and multivariate Cox analysis of overall survival in ESCC patients\nAbbreviations: CI, confidence interval; ESCC, esophageal squamous cell carcinoma; HR, hazard ration; —, no data available.", "As lncRNA RASSF8‐AS1 showed good performance for ESCC diagnosis and RASSF8‐AS1 expression was an independent prognostic factor for both short‐ and long‐term outcomes of ESCC. We speculated that RASSF8‐AS1 plays an oncogenic role in ESCC. We then determined that higher RASSF8‐AS1 was associated with clinicopathological factors (Table 4), such as larger tumor size (p < 0.0001), greater depth of infiltration (p < 0.0001), lymph node metastasis (p = 0.0278), and advanced tumor stage (p = 0.0110). The expression of RASSF8‐AS1 tended to decrease significantly after tumor resection surgery in ESCC patients (Figure 4c), and RASSF8‐AS1 was found to be significantly higher in ESCC in the mid‐late stage than in stage I through the GEPIA database (Figure S3 D). The above data suggested that RASSF8‐AS1 plays a protumor role for ESCC.\nThe correlation between the serum RASSF8‐AS1 expression and clinicopathological factors of ESCC\nAbbreviation: ESCC, esophageal squamous cell carcinoma; —, no data available.\nBold values means p‐value < 0.05.\nSubsequently, we explored the oncogenic function of RASSF8‐AS1 by knocking down RASSF8‐AS1 in TE1 and TE13 cells (Figure 6a). In vitro assays showed that the knockdown of RASSF8‐AS1 inhibited cell proliferation (Figure 6b,c), motility and invasiveness (Figure 6d), respectively.\nRASSF8‐AS1 promotes esophageal squamous cell carcinoma (ESCC) cell proliferation, migration and invasion in vitro. (a) The mRNA expression of RASSF8‐AS1 after transfection by two siRNAs. (b, c) Assessment of knockdown of RASSF8‐AS1 on cell growth of TE1 and TE13 cells. (d) Assessment of knockdown of RASSF8‐AS1 on cell migration and invasion. (e) The mRNA expression of RASSF8‐AS1 in normal tissues and ESCC tissues. (f) Kaplan–Meier survival curves of ESCC patients with low and high RASSF8‐AS1 expression. *p < 0.05, **p < 0.01, ***p < 0.001.\nIn addition, we performed qRT‐PCR on 60 pairs of tumor and adjacent normal tissues, and found that RASSF8‐AS1 was likewise markedly elevated in ESCC tissues (Figure 6e). Next, we divided the 60 ESCC samples into RASSF8‐AS1 low expression group (n = 30) and high expression group (n = 30) according to the median level of RASSF8‐AS1 to explore the prognostic significance of RASSF8‐AS1 in ESCC. Kaplan–Meier analysis showed that patients with high RASSF8‐AS1 expression had a better dismal OS (Figure 6f).\nFinally, we screened RASSF8‐AS1 targets by RNA‐sequencing and performed gene ontology (GO) and pathway enrichment analysis. The result found 303 positively regulated (Figure S4 C, D) and 430 negatively regulated targets (Figure 4e,f). GO results suggested RASSF8‐AS1 was positively involved, mainly in immune response and cell migration, and KEGG pathway analysis indicated the positively regulated targets mostly enriched in cell metabolism (Figure S4 E, F), all of which have been proven to be essential for tumor progression.", "This research was supported by the National Natural Science Foundation for Youth of China (no. 81702444), China Postdoctoral Science Foundation 2018M643884 and 2020T130128ZX." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "METHODS", "Patients and samples", "Exosome extraction and identification", "\nRNA extraction and quantitative real‐time PCR (qRT‐PCR)", "Microarray screening and analysis", "Cell culture", "Stability analysis for exosomal lncRNAs panel", "Transfection and RNA interfering", "Cell proliferation, migration, and invasion assays", "\nRNA sequencing and enrichment analysis", "Statistical analysis", "RESULTS", "Identification of exosomes in serum", "Discovery phase: Genome‐wide profiling to identify differentially expressed lncRNAs between ESCC and healthy individual controls", "Evaluation of stability of exosomal lncRNA in serum samples under different conditions", "Training phase: A panel of serum lncRNAs‐based diagnostic indicators Index I", "Validation phase: The lncRNAs based model can distinguish ESCC from healthy control and the levels of lncRNA panel correlated with clinicopathological features", "Prognostic significance of three serum lncRNAs panel", "In vitro analysis of the oncogenic potential of RASSF8‐AS1 and target scanning", "DISCUSSION", "FUNDING INFORMATION", "CONFLICT OF INTEREST", "Supporting information" ]
[ "Esophageal cancer is the eighth most common cancer and the sixth leading cause of malignancy deaths worldwide.\n1\n Squamous cell carcinoma is the main pathology type in China. The current standard for esophageal squamous cell carcinoma (ESCC) diagnosis depends on endoscopy and pathological biopsy.\n2\n However, the operation is relatively invasive and uncomfortable, resulting in delay or missing the best chance for most ESCC diagnosis and therapy. Liquid biopsy provides an alternative way of ESCC screening, but the traditional biomarkers used in the diagnosis of ESCC, such as serum squamous cell carcinoma antigen (SCC), cancer antigen 125 (CA125), carcinoembryonic antigen (CEA), and cytokeratin‐19‐fragment (CYFRA21‐1),\n3\n, \n4\n lack enough sensitivity and specificity, which limits their clinical application. For example, SCC may be also applied in other types of squamous cell carcinoma, such as lung, head and neck, and cervical squamous cell carcinoma. CEA is mainly used for the detection of colorectal cancer, while CYFRA21‐1 is usually used as a biomarker for diverse subtypes of lung and bladder cancer.\nExosomes are small (50–150 nm) membrane vesicles inside the cell, which may fuse with the membrane and are then released into the circulation.\n5\n, \n6\n They are detected in various body fluids such as serum, plasma, urine and saliva.\n7\n, \n8\n Exosomes execute communication functions intercellularly by transporting various biomolecules (such as DNAs, RNAs and proteins),\n9\n, \n10\n and the contents are protected by a phospholipid bilayer, making them suitable for liquid biopsy. Long noncoding RNAs (LncRNAs) is one of the exosome cargos,\n11\n and previous studies considered that lncRNAs in exosomes may be more stable for exosome protection against RNase.\n12\n, \n13\n LncRNAs detected in body fluids have been shown to be ideal noninvasive biomarkers for cancer diagnosis and prognosis. For example, lncRNA HOTAIR and SBF2‐AS1 in serum exosomes have been implicated for the diagnosis and prognosis assessment of glioblastoma.\n14\n, \n15\n Similarly, Xu et al.\n16\n found in an independent cohort study that ENSG00000258332.1 and LINC00635 in serum had higher sensitivity and specificity for the diagnosis of hepatocellular carcinoma in combination with AFP. Recently, Tao et al.\n17\n found that the expression levels of TBILA and AGAP2‐AS1 in the serum of NSCLC patients were significantly increased, and the combination with serum tumor biomarkers could further improve the diagnostic accuracy of NSCLC patients. Compared with the increasing evidence for exosomal lncRNAs in other types of cancer, little is known about their roles in ESCC. Furthermore, it has previously been reported that a joint model based on the union of multiple biomarkers may improve the diagnostic power, which prompted our study design.\n18\n, \n19\n\n\nHere, we used a multistep study to investigate the possibility that novel serum exosomal lncRNAs act as potential biomarkers for ESCC detection. We selected candidate lncRNAs by integrating microarray results derived from serum and tissue samples separately, as a previous study reported that serum exosomal lncRNAs may be derived from tumor tissues. We deduced that the lncRNAs simultaneously overexpressed in serum and tissue samples may be more likely to meet the criteria. Finally, a diagnostic model based on three lncRNAs panel was established and the association of the novel panel with clinicopathological features or prognosis was also evaluated. Furthermore, we found involvement of candidate lncRNA RASSF8‐AS1 in ESCC malignant phenotypes, and the combined results provide new insights into ESCC diagnosis and target therapy.", "[SUBTITLE] Patients and samples [SUBSECTION] From 2017 to 2021, 140 ESCC patients and 140 healthy individuals were randomly recruited from Jinling Hospital of Nanjing Medical University, each of whom provided 5 ml of whole blood at indicated timepoints. Neither patient received preoperative chemo−/radiotherapy, and the overall survival (OS) was measured from the date of diagnosis to the date of death due to any cause or the date of the most recent follow‐up. All patients were followed every 3 months for the first year, every 6 months for the next 2 years, and then annually. The end of the follow‐up was November 31, 2021. The study was approved by the ethics committee of Jinling hospital, and all enrolled participants signed informed consent. All serum samples were stored at −80°C until use for total RNA extraction. The pathological stage was determined according to the eighth edition of the Union for International Cancer Control TNM staging system.\nFrom 2017 to 2021, 140 ESCC patients and 140 healthy individuals were randomly recruited from Jinling Hospital of Nanjing Medical University, each of whom provided 5 ml of whole blood at indicated timepoints. Neither patient received preoperative chemo−/radiotherapy, and the overall survival (OS) was measured from the date of diagnosis to the date of death due to any cause or the date of the most recent follow‐up. All patients were followed every 3 months for the first year, every 6 months for the next 2 years, and then annually. The end of the follow‐up was November 31, 2021. The study was approved by the ethics committee of Jinling hospital, and all enrolled participants signed informed consent. All serum samples were stored at −80°C until use for total RNA extraction. The pathological stage was determined according to the eighth edition of the Union for International Cancer Control TNM staging system.\n[SUBTITLE] Exosome extraction and identification [SUBSECTION] A total of 1.5 ml of serum was centrifuged at 2000 × g, 4°C for 30 min, and the supernatant was then recentrifuged at 10 000 × g, 4°C for 45 min. The supernatant was then collected and filtered through a membrane of 0.22 μm pore size. The filtrate was collected and further subjected to ultracentrifugation at 10 000 × g, 4°C for 70 min. The precipitate was resuspended in 10 ml of precooling 1 × PBS, and ultracentrifugation repeated. Finally, the precipitate was resuspended in 1.5 ml of precooling 1 × PBS. The isolated exosomes were characterized using transmission electron microscopy (TEM). Exosome concentration‐particle size testing (nanoparticle tracking analysis [NTA]) was performed by Zeta View (Particle Metrix Ltd.).\nA total of 1.5 ml of serum was centrifuged at 2000 × g, 4°C for 30 min, and the supernatant was then recentrifuged at 10 000 × g, 4°C for 45 min. The supernatant was then collected and filtered through a membrane of 0.22 μm pore size. The filtrate was collected and further subjected to ultracentrifugation at 10 000 × g, 4°C for 70 min. The precipitate was resuspended in 10 ml of precooling 1 × PBS, and ultracentrifugation repeated. Finally, the precipitate was resuspended in 1.5 ml of precooling 1 × PBS. The isolated exosomes were characterized using transmission electron microscopy (TEM). Exosome concentration‐particle size testing (nanoparticle tracking analysis [NTA]) was performed by Zeta View (Particle Metrix Ltd.).\n[SUBTITLE] \nRNA extraction and quantitative real‐time PCR (qRT‐PCR) [SUBSECTION] RNA was extracted from exosomes or tissues using an exosome RNA isolation kit according to protocols (Invitrogen). The extracted RNA was synthesized into cDNA by the PrimeScript RT Master Mix (Takara). RT‐qPCR was performed to detect the relative RNA levels of target genes. β‐actin was used as an internal reference gene for analysis. The relative expression levels of the lncRNAs were calculated using 2−∆∆Ct.\n20\n, \n21\n The sequences of primers used for qRT‐PCR of the lncRNAs are listed in Table S1.\nRNA was extracted from exosomes or tissues using an exosome RNA isolation kit according to protocols (Invitrogen). The extracted RNA was synthesized into cDNA by the PrimeScript RT Master Mix (Takara). RT‐qPCR was performed to detect the relative RNA levels of target genes. β‐actin was used as an internal reference gene for analysis. The relative expression levels of the lncRNAs were calculated using 2−∆∆Ct.\n20\n, \n21\n The sequences of primers used for qRT‐PCR of the lncRNAs are listed in Table S1.\n[SUBTITLE] Microarray screening and analysis [SUBSECTION] Microarray screening was performed with Arraystar LncRNA_8 × 60 k (Arraystar), which contains both lncRNA and mRNA probes. Sample labeling and array hybridization were performed according to the Agilent One‐Color Microarray‐Based Gene Expression Analysis protocol (Agilent Technology). Agilent Feature Extraction software (version 11.0.1.1) was used to analyze acquired array images. Quantile normalization and subsequent data processing were performed using the GeneSpring GX version 12.1 software package (Agilent Technologies). After quantile normalization of the raw data, LncRNAs and mRNAs that at least four out of eight samples have flags in Present or Marginal (“All Targets Value”) were chosen for further data analysis. Differentially expressed LncRNAs and mRNAs with statistical significance between the two groups were identified through p‐value/FDR filtering.\nMicroarray screening was performed with Arraystar LncRNA_8 × 60 k (Arraystar), which contains both lncRNA and mRNA probes. Sample labeling and array hybridization were performed according to the Agilent One‐Color Microarray‐Based Gene Expression Analysis protocol (Agilent Technology). Agilent Feature Extraction software (version 11.0.1.1) was used to analyze acquired array images. Quantile normalization and subsequent data processing were performed using the GeneSpring GX version 12.1 software package (Agilent Technologies). After quantile normalization of the raw data, LncRNAs and mRNAs that at least four out of eight samples have flags in Present or Marginal (“All Targets Value”) were chosen for further data analysis. Differentially expressed LncRNAs and mRNAs with statistical significance between the two groups were identified through p‐value/FDR filtering.\n[SUBTITLE] Cell culture [SUBSECTION] Esophageal squamous cell carcinoma cell lines (TE1, TE13, KYSE150, KYSE140) and normal human esophageal epithelial cells (HET‐1A) were obtained from the Shanghai Institutes for Biological Sciences (Shanghai, China). These cells were cultured in 1640 medium (KeyGene) containing 10% fetal bovine serum (FBS) (KeyGene), at 37°C in a 5% CO2 atmosphere.\nEsophageal squamous cell carcinoma cell lines (TE1, TE13, KYSE150, KYSE140) and normal human esophageal epithelial cells (HET‐1A) were obtained from the Shanghai Institutes for Biological Sciences (Shanghai, China). These cells were cultured in 1640 medium (KeyGene) containing 10% fetal bovine serum (FBS) (KeyGene), at 37°C in a 5% CO2 atmosphere.\n[SUBTITLE] Stability analysis for exosomal lncRNAs panel [SUBSECTION] Exosome samples (n = 15) were allocated to aliquots for different processing, including storage under different temperature, treated with RNase A, and subjected to freeze–thaw cycles and acidic/alkaline environments, to test to the stability of exosomal lncRNAs, which is essential for clinical use.\nExosome samples (n = 15) were allocated to aliquots for different processing, including storage under different temperature, treated with RNase A, and subjected to freeze–thaw cycles and acidic/alkaline environments, to test to the stability of exosomal lncRNAs, which is essential for clinical use.\n[SUBTITLE] Transfection and RNA interfering [SUBSECTION] Cells were seeded on six‐well plates (2 × 105/well) with 1640 medium supplemented with 10% FBS overnight. Then, siRNA was transfected at a final concentration of 100 nM using lipofectamine 2000 (Invitrogen), and 24 h post transfection cells were harvested and subsequently RNA was extracted by Trizol reagent and the interfering efficiency was determined by RT‐qPCR. Sequences of siRNAs and negative control are provided in Table S2.\nCells were seeded on six‐well plates (2 × 105/well) with 1640 medium supplemented with 10% FBS overnight. Then, siRNA was transfected at a final concentration of 100 nM using lipofectamine 2000 (Invitrogen), and 24 h post transfection cells were harvested and subsequently RNA was extracted by Trizol reagent and the interfering efficiency was determined by RT‐qPCR. Sequences of siRNAs and negative control are provided in Table S2.\n[SUBTITLE] Cell proliferation, migration, and invasion assays [SUBSECTION] Transfected cells were collected and grown in 96‐well plates (2 × 103 cells per well) overnight and cell proliferation was detected using cell counting kit‐8 (CCK‐8). The reaction products were measured at 450 nm according to the manufacturer's instructions. For migration/invasion assays, 5 × 104 cells were incubated using Transwell kits (8 mm PET, 24‐well Millicell) or matrix‐coated inserts (BD Biosciences). After 24 or 48 h of incubation, cells were fixed with 4% paraformaldehyde and stained with crystal violet (Beyotim). Imaging and counting of migrated cells were conducted by microscopy.\nTransfected cells were collected and grown in 96‐well plates (2 × 103 cells per well) overnight and cell proliferation was detected using cell counting kit‐8 (CCK‐8). The reaction products were measured at 450 nm according to the manufacturer's instructions. For migration/invasion assays, 5 × 104 cells were incubated using Transwell kits (8 mm PET, 24‐well Millicell) or matrix‐coated inserts (BD Biosciences). After 24 or 48 h of incubation, cells were fixed with 4% paraformaldehyde and stained with crystal violet (Beyotim). Imaging and counting of migrated cells were conducted by microscopy.\n[SUBTITLE] \nRNA sequencing and enrichment analysis [SUBSECTION] Cell transfection and RNA interfering were performed as described above. Then, 24 h post transfection, cells were harvested and RNA was extracted using Trizol reagent (Invitrogen). RNA‐seq libraries were prepared using the Illumina RNA‐seq preparation kit and sequenced on illumina nova seq 6000 sequencer. The differentially expressed RNAs (fold change >1.5, p < 0.05) were retrieved and subjected to gene ontology and KEGG pathway analysis.\nCell transfection and RNA interfering were performed as described above. Then, 24 h post transfection, cells were harvested and RNA was extracted using Trizol reagent (Invitrogen). RNA‐seq libraries were prepared using the Illumina RNA‐seq preparation kit and sequenced on illumina nova seq 6000 sequencer. The differentially expressed RNAs (fold change >1.5, p < 0.05) were retrieved and subjected to gene ontology and KEGG pathway analysis.\n[SUBTITLE] Statistical analysis [SUBSECTION] All data were statistically analyzed with SPSS 26.0 (Chicago) and visualized with GraphPad Prism version 8.0 (GraphPad Software). Differences in lncRNAs expression between groups were determined using the Mann–Whitney unpaired t‐test or paired t‐test. The χ2 or Fisher's exact test was used to compare the clinical characteristics of the groups. Receiver operating characteristic (ROC) curves were used to assess the diagnostic performance of selected biomarkers. The optimal cutoff point was determined using Youden's index. DeLong's test was used to evaluate the statistical significance when comparing differences of AUCs. Using the binary status of enrolled participants (patients or controls) as the dependent variable, the regression coefficient for each lncRNA was estimated by a univariate logistic regression model, and was used as the weighting to construct the diagnostic index. The diagnostic index developed in the training phase was also directly applied to the validation phase to evaluate the clinical utility of the identified lncRNAs individually or in combination. A forward conditional logistic regression model was used to analyze the association between selected biomarkers and ESCC mortality. The long‐term prognostic values of selected biomarkers were evaluated using Cox's hazard regression model and Kaplan–Meier curve analysis. Univariate analysis was used to explore clinicopathological factors (i.e., tumor depth, stage, size) associated with OS. Factors with p‐value <0.05 were selected for the multivariate analysis with Cox's proportional hazard regression model, hazard ratio and 95% confidence interval (CI) were calculated. All p‐values <0.05 were considered statistically significant.\nAll data were statistically analyzed with SPSS 26.0 (Chicago) and visualized with GraphPad Prism version 8.0 (GraphPad Software). Differences in lncRNAs expression between groups were determined using the Mann–Whitney unpaired t‐test or paired t‐test. The χ2 or Fisher's exact test was used to compare the clinical characteristics of the groups. Receiver operating characteristic (ROC) curves were used to assess the diagnostic performance of selected biomarkers. The optimal cutoff point was determined using Youden's index. DeLong's test was used to evaluate the statistical significance when comparing differences of AUCs. Using the binary status of enrolled participants (patients or controls) as the dependent variable, the regression coefficient for each lncRNA was estimated by a univariate logistic regression model, and was used as the weighting to construct the diagnostic index. The diagnostic index developed in the training phase was also directly applied to the validation phase to evaluate the clinical utility of the identified lncRNAs individually or in combination. A forward conditional logistic regression model was used to analyze the association between selected biomarkers and ESCC mortality. The long‐term prognostic values of selected biomarkers were evaluated using Cox's hazard regression model and Kaplan–Meier curve analysis. Univariate analysis was used to explore clinicopathological factors (i.e., tumor depth, stage, size) associated with OS. Factors with p‐value <0.05 were selected for the multivariate analysis with Cox's proportional hazard regression model, hazard ratio and 95% confidence interval (CI) were calculated. All p‐values <0.05 were considered statistically significant.", "From 2017 to 2021, 140 ESCC patients and 140 healthy individuals were randomly recruited from Jinling Hospital of Nanjing Medical University, each of whom provided 5 ml of whole blood at indicated timepoints. Neither patient received preoperative chemo−/radiotherapy, and the overall survival (OS) was measured from the date of diagnosis to the date of death due to any cause or the date of the most recent follow‐up. All patients were followed every 3 months for the first year, every 6 months for the next 2 years, and then annually. The end of the follow‐up was November 31, 2021. The study was approved by the ethics committee of Jinling hospital, and all enrolled participants signed informed consent. All serum samples were stored at −80°C until use for total RNA extraction. The pathological stage was determined according to the eighth edition of the Union for International Cancer Control TNM staging system.", "A total of 1.5 ml of serum was centrifuged at 2000 × g, 4°C for 30 min, and the supernatant was then recentrifuged at 10 000 × g, 4°C for 45 min. The supernatant was then collected and filtered through a membrane of 0.22 μm pore size. The filtrate was collected and further subjected to ultracentrifugation at 10 000 × g, 4°C for 70 min. The precipitate was resuspended in 10 ml of precooling 1 × PBS, and ultracentrifugation repeated. Finally, the precipitate was resuspended in 1.5 ml of precooling 1 × PBS. The isolated exosomes were characterized using transmission electron microscopy (TEM). Exosome concentration‐particle size testing (nanoparticle tracking analysis [NTA]) was performed by Zeta View (Particle Metrix Ltd.).", "RNA was extracted from exosomes or tissues using an exosome RNA isolation kit according to protocols (Invitrogen). The extracted RNA was synthesized into cDNA by the PrimeScript RT Master Mix (Takara). RT‐qPCR was performed to detect the relative RNA levels of target genes. β‐actin was used as an internal reference gene for analysis. The relative expression levels of the lncRNAs were calculated using 2−∆∆Ct.\n20\n, \n21\n The sequences of primers used for qRT‐PCR of the lncRNAs are listed in Table S1.", "Microarray screening was performed with Arraystar LncRNA_8 × 60 k (Arraystar), which contains both lncRNA and mRNA probes. Sample labeling and array hybridization were performed according to the Agilent One‐Color Microarray‐Based Gene Expression Analysis protocol (Agilent Technology). Agilent Feature Extraction software (version 11.0.1.1) was used to analyze acquired array images. Quantile normalization and subsequent data processing were performed using the GeneSpring GX version 12.1 software package (Agilent Technologies). After quantile normalization of the raw data, LncRNAs and mRNAs that at least four out of eight samples have flags in Present or Marginal (“All Targets Value”) were chosen for further data analysis. Differentially expressed LncRNAs and mRNAs with statistical significance between the two groups were identified through p‐value/FDR filtering.", "Esophageal squamous cell carcinoma cell lines (TE1, TE13, KYSE150, KYSE140) and normal human esophageal epithelial cells (HET‐1A) were obtained from the Shanghai Institutes for Biological Sciences (Shanghai, China). These cells were cultured in 1640 medium (KeyGene) containing 10% fetal bovine serum (FBS) (KeyGene), at 37°C in a 5% CO2 atmosphere.", "Exosome samples (n = 15) were allocated to aliquots for different processing, including storage under different temperature, treated with RNase A, and subjected to freeze–thaw cycles and acidic/alkaline environments, to test to the stability of exosomal lncRNAs, which is essential for clinical use.", "Cells were seeded on six‐well plates (2 × 105/well) with 1640 medium supplemented with 10% FBS overnight. Then, siRNA was transfected at a final concentration of 100 nM using lipofectamine 2000 (Invitrogen), and 24 h post transfection cells were harvested and subsequently RNA was extracted by Trizol reagent and the interfering efficiency was determined by RT‐qPCR. Sequences of siRNAs and negative control are provided in Table S2.", "Transfected cells were collected and grown in 96‐well plates (2 × 103 cells per well) overnight and cell proliferation was detected using cell counting kit‐8 (CCK‐8). The reaction products were measured at 450 nm according to the manufacturer's instructions. For migration/invasion assays, 5 × 104 cells were incubated using Transwell kits (8 mm PET, 24‐well Millicell) or matrix‐coated inserts (BD Biosciences). After 24 or 48 h of incubation, cells were fixed with 4% paraformaldehyde and stained with crystal violet (Beyotim). Imaging and counting of migrated cells were conducted by microscopy.", "Cell transfection and RNA interfering were performed as described above. Then, 24 h post transfection, cells were harvested and RNA was extracted using Trizol reagent (Invitrogen). RNA‐seq libraries were prepared using the Illumina RNA‐seq preparation kit and sequenced on illumina nova seq 6000 sequencer. The differentially expressed RNAs (fold change >1.5, p < 0.05) were retrieved and subjected to gene ontology and KEGG pathway analysis.", "All data were statistically analyzed with SPSS 26.0 (Chicago) and visualized with GraphPad Prism version 8.0 (GraphPad Software). Differences in lncRNAs expression between groups were determined using the Mann–Whitney unpaired t‐test or paired t‐test. The χ2 or Fisher's exact test was used to compare the clinical characteristics of the groups. Receiver operating characteristic (ROC) curves were used to assess the diagnostic performance of selected biomarkers. The optimal cutoff point was determined using Youden's index. DeLong's test was used to evaluate the statistical significance when comparing differences of AUCs. Using the binary status of enrolled participants (patients or controls) as the dependent variable, the regression coefficient for each lncRNA was estimated by a univariate logistic regression model, and was used as the weighting to construct the diagnostic index. The diagnostic index developed in the training phase was also directly applied to the validation phase to evaluate the clinical utility of the identified lncRNAs individually or in combination. A forward conditional logistic regression model was used to analyze the association between selected biomarkers and ESCC mortality. The long‐term prognostic values of selected biomarkers were evaluated using Cox's hazard regression model and Kaplan–Meier curve analysis. Univariate analysis was used to explore clinicopathological factors (i.e., tumor depth, stage, size) associated with OS. Factors with p‐value <0.05 were selected for the multivariate analysis with Cox's proportional hazard regression model, hazard ratio and 95% confidence interval (CI) were calculated. All p‐values <0.05 were considered statistically significant.", "[SUBTITLE] Identification of exosomes in serum [SUBSECTION] A total of 280 participants, including 140 ESCC patients and 140 healthy controls were enrolled in the study, as shown in Table S3. To verify whether the exosomes were successfully isolated from serum, we characterized the isolated exosomes using TEM and NTA. The results showed that the isolated exosomes had a bilayer membrane structure (Figure S1 A), and the particle size distribution of exosomes was about 30–150 nm in diameter (Figure S1 B), meeting the criteria of exosome patterns.\nA total of 280 participants, including 140 ESCC patients and 140 healthy controls were enrolled in the study, as shown in Table S3. To verify whether the exosomes were successfully isolated from serum, we characterized the isolated exosomes using TEM and NTA. The results showed that the isolated exosomes had a bilayer membrane structure (Figure S1 A), and the particle size distribution of exosomes was about 30–150 nm in diameter (Figure S1 B), meeting the criteria of exosome patterns.\n[SUBTITLE] Discovery phase: Genome‐wide profiling to identify differentially expressed lncRNAs between ESCC and healthy individual controls [SUBSECTION] The overall workflow of this study is illustrated in Figure 1a. In the discovery phase, we performed LncRNA microarray analysis on serum exosome samples from four preoperative ESCC patients and matched healthy controls. The top 100 differential lncRNAs identified in serum exosome samples from four preoperative ESCC patients and healthy control are shown in Figure 1b (FDR <0.005, Fold change >2, GEO accession number: GSE192662). Circos plots globally and genome‐widely displayed kinds of detected lncRNAs in ESCC and control serum samples (Figure 1c). A previous microarray comparing five pairs of tumor and paracancerous tissues (GSE89102) was also included for further analysis. To achieve more reliable aberrantly expressed lncRNAs, we adopted a more restrictive filtering criteria (FDR <0.005, fold change >5) for both datasets, and integrated the two datasets to finally obtain five (AC098818.2, RASSF8‐AS1, LINC00958, GMDS‐DT and AL591721.1) candidate genes with significant overexpression in ESCC serum samples and cancerous tissues (Figure 1d).\nMicroarray screening results of differential lncRNAs and function prediction. (a) Experimental design overview. (b) Heatmap results of microarray analysis for four preoperative esophageal squamous cell carcinoma (ESCC) patients and matched normal healthy serum samples. (c) Circos plots of lncRNAs in the human genome (hg19). The outer tracks represent the cytoband ideogram of chromosome. For the two tracks, the outer one (blue) represents the levels of lncRNAs in normal tissues and the inner one (red) represents the levels in ESCC tissues. (d) Venn diagram showing consistent upregulation of five lncRNAs in both microarray analyses. (e, f) Signal pathway networks of mRNAs involved in lncRNAs‐mRNAs relationships.\nTo investigate the potential role of the candidate lncRNAs in ESCC, we analyzed mRNA results from the same microarray chip derived from serum samples and performed pathway analysis (Figure S2 A–D). By lncRNAs‐mRNA coexpression network analysis, most of the candidate lncRNAs‐related mRNAs could be enriched in pathways such as MAPK, Ras, Wnt, metabolic pathways and HIF‐1α, which have been identified to be involved in ESCC carcinogenesis (Figure 1e,f).\nTo validate the microarray data, we examined the expression of five candidate lncRNAs in a pilot cohort containing serum exosome samples (n = 20 for ESCC and healthy control separately), and 30 paired tissues, respectively. The qRT‐PCR results showed that the expression of AC098818.2, RASSF8‐AS1 and LINC00958 being significantly increased in ESCC patients' serum compared with healthy, while no significant difference in the expression of AL591721.1 and GMDS‐DT (Figure 2a). Compared with normal esophageal tissues, AC098818.2, RASSF8‐AS1 and LINC00958 expression was significantly upregulated in tumor tissues, whereas the expression level of GMDS‐DT was significantly downregulated, and no significant difference was seen in the expression of AL591721.1 (Figure 2b). Moreover, we examined the expression levels of these five lncRNAs in ESCC cell lines (TE1, TE13, KYSE140 and KYSE150) and normal epithelial cell HET‐1A. Compared with HET‐1A, the expression of AC098818.2, RASSF8‐AS1 and LINC00958 was upregulated in the ESCC cell lines, while there was no significant difference in the expression of AL591721.1 and GMDS‐DT (Figure 2c). Meanwhile, by big data mining through ExoRbase, we found that AC098818.2, RASSF8‐AS1 and LINC00958 were significantly overexpressed in ESCC blood exosomes compared with healthy donors and other types of cancers (Figure S3 A‐C), indicating excellent specificity in ESCC screening. By searching in the GEPIA database which obtained expression data from TCGA, a gradually increased trend of RASSF8‐AS1 expression toward TNM stages was found (Figure S3 D). Furthermore, we detected expression level of the three candidates in 12 pairs of serum samples and corresponding cancerous tissues, and found moderate correlation for each lncRNA (Figure 2d‐f).\nIdentification of candidate five lncRNAs expression. (a) The expression levels of the five candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) serum (n = 20) and normal serum samples (n = 20). (b) The expression levels of the five candidate lncRNAs in paired tumors and adjacent normal tissues (n = 30). (c) The expression levels of the five candidate lncRNAs in ESCC cell lines (TE1, TE13, KYSE140 and KYSE150) and the human esophageal epithelial cells line (HET‐1A). (d–f) Spearman's correlation analysis between expression levels of lncRNAs in tumor tissues and paired serum samples of ESCC. *p < 0.05, **p < 0.01 .\nThe overall workflow of this study is illustrated in Figure 1a. In the discovery phase, we performed LncRNA microarray analysis on serum exosome samples from four preoperative ESCC patients and matched healthy controls. The top 100 differential lncRNAs identified in serum exosome samples from four preoperative ESCC patients and healthy control are shown in Figure 1b (FDR <0.005, Fold change >2, GEO accession number: GSE192662). Circos plots globally and genome‐widely displayed kinds of detected lncRNAs in ESCC and control serum samples (Figure 1c). A previous microarray comparing five pairs of tumor and paracancerous tissues (GSE89102) was also included for further analysis. To achieve more reliable aberrantly expressed lncRNAs, we adopted a more restrictive filtering criteria (FDR <0.005, fold change >5) for both datasets, and integrated the two datasets to finally obtain five (AC098818.2, RASSF8‐AS1, LINC00958, GMDS‐DT and AL591721.1) candidate genes with significant overexpression in ESCC serum samples and cancerous tissues (Figure 1d).\nMicroarray screening results of differential lncRNAs and function prediction. (a) Experimental design overview. (b) Heatmap results of microarray analysis for four preoperative esophageal squamous cell carcinoma (ESCC) patients and matched normal healthy serum samples. (c) Circos plots of lncRNAs in the human genome (hg19). The outer tracks represent the cytoband ideogram of chromosome. For the two tracks, the outer one (blue) represents the levels of lncRNAs in normal tissues and the inner one (red) represents the levels in ESCC tissues. (d) Venn diagram showing consistent upregulation of five lncRNAs in both microarray analyses. (e, f) Signal pathway networks of mRNAs involved in lncRNAs‐mRNAs relationships.\nTo investigate the potential role of the candidate lncRNAs in ESCC, we analyzed mRNA results from the same microarray chip derived from serum samples and performed pathway analysis (Figure S2 A–D). By lncRNAs‐mRNA coexpression network analysis, most of the candidate lncRNAs‐related mRNAs could be enriched in pathways such as MAPK, Ras, Wnt, metabolic pathways and HIF‐1α, which have been identified to be involved in ESCC carcinogenesis (Figure 1e,f).\nTo validate the microarray data, we examined the expression of five candidate lncRNAs in a pilot cohort containing serum exosome samples (n = 20 for ESCC and healthy control separately), and 30 paired tissues, respectively. The qRT‐PCR results showed that the expression of AC098818.2, RASSF8‐AS1 and LINC00958 being significantly increased in ESCC patients' serum compared with healthy, while no significant difference in the expression of AL591721.1 and GMDS‐DT (Figure 2a). Compared with normal esophageal tissues, AC098818.2, RASSF8‐AS1 and LINC00958 expression was significantly upregulated in tumor tissues, whereas the expression level of GMDS‐DT was significantly downregulated, and no significant difference was seen in the expression of AL591721.1 (Figure 2b). Moreover, we examined the expression levels of these five lncRNAs in ESCC cell lines (TE1, TE13, KYSE140 and KYSE150) and normal epithelial cell HET‐1A. Compared with HET‐1A, the expression of AC098818.2, RASSF8‐AS1 and LINC00958 was upregulated in the ESCC cell lines, while there was no significant difference in the expression of AL591721.1 and GMDS‐DT (Figure 2c). Meanwhile, by big data mining through ExoRbase, we found that AC098818.2, RASSF8‐AS1 and LINC00958 were significantly overexpressed in ESCC blood exosomes compared with healthy donors and other types of cancers (Figure S3 A‐C), indicating excellent specificity in ESCC screening. By searching in the GEPIA database which obtained expression data from TCGA, a gradually increased trend of RASSF8‐AS1 expression toward TNM stages was found (Figure S3 D). Furthermore, we detected expression level of the three candidates in 12 pairs of serum samples and corresponding cancerous tissues, and found moderate correlation for each lncRNA (Figure 2d‐f).\nIdentification of candidate five lncRNAs expression. (a) The expression levels of the five candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) serum (n = 20) and normal serum samples (n = 20). (b) The expression levels of the five candidate lncRNAs in paired tumors and adjacent normal tissues (n = 30). (c) The expression levels of the five candidate lncRNAs in ESCC cell lines (TE1, TE13, KYSE140 and KYSE150) and the human esophageal epithelial cells line (HET‐1A). (d–f) Spearman's correlation analysis between expression levels of lncRNAs in tumor tissues and paired serum samples of ESCC. *p < 0.05, **p < 0.01 .\n[SUBTITLE] Evaluation of stability of exosomal lncRNA in serum samples under different conditions [SUBSECTION] Given that stability is a key prerequisite for body fluid tumor biomarkers, we evaluated the stability of exosomal lncRNAs in serum samples under different conditions. Serum samples were subjected to extreme conditions, such as incubation at different temperatures for 3 h, repeated freeze–thaw cycles, RNase A digestion for 3 and 6 h, and exposure to acidic or alkaline environments. Interestingly, these treatments had little or no effect on the level of these serum lncRNAs (Figure 3), indicating the lncRNAs panel was valuable for clinical application.\nStability of plasma lncRNAs under extreme conditions. (a) Incubation at different temperature for 3 h. (b) Repeated freeze–thaw cycles. (c) RNase A digestion for 3 and 6 h. (d) Exposure to acidic or alkaline environments.\nGiven that stability is a key prerequisite for body fluid tumor biomarkers, we evaluated the stability of exosomal lncRNAs in serum samples under different conditions. Serum samples were subjected to extreme conditions, such as incubation at different temperatures for 3 h, repeated freeze–thaw cycles, RNase A digestion for 3 and 6 h, and exposure to acidic or alkaline environments. Interestingly, these treatments had little or no effect on the level of these serum lncRNAs (Figure 3), indicating the lncRNAs panel was valuable for clinical application.\nStability of plasma lncRNAs under extreme conditions. (a) Incubation at different temperature for 3 h. (b) Repeated freeze–thaw cycles. (c) RNase A digestion for 3 and 6 h. (d) Exposure to acidic or alkaline environments.\n[SUBTITLE] Training phase: A panel of serum lncRNAs‐based diagnostic indicators Index I [SUBSECTION] To further investigate the diagnostic ability of the three candidate lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) for ESCC, we determined the relative expression levels of these three lncRNAs in serum samples of 40 ESCC patients and 40 healthy individuals and performed ROC curve analysis to evaluate the diagnostic capability for each lncRNA alone. The results showed that the expression levels of these lncRNAs were significantly elevated in ESCC patients (Figure 4a), with AUC values ranging from 0.69 to 0.78 (Figure 4b). Table S4 summarizes in detail the AUC values and 95% CI of each individual lncRNA. The optimal threshold for each lncRNA was determined by the Youden's index.\n22\n Serum lncRNA RASSF8‐AS1 was the best diagnostic marker, with AUC values of 0.78 and 95% CI: 0.67–0.88, respectively.\nTraining phase: A panel of serum lncRNAs‐based diagnostic indicators Index I. (a) The expression levels of the three candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) serum (n = 40) and normal serum samples (n = 40). (b) Receiver operating characteristic (ROC) curves to evaluate the sensitivity and specificity of three candidate lncRNAs to discriminate ESCC from healthy controls. (c) Paired comparison of the three candidate lncRNAs level before and after surgery (n = 20). (d) ROC curves to evaluate the sensitivity and specificity of traditional tumor markers to discriminate ESCC from healthy control. (e) Comparison of the diagnostic performance of Index I and Index II. *p < 0.05, **p < 0.01.\nTo evaluate whether the expression levels of the lncRNAs panel could be used to monitor tumor dynamics, we collected paired serum samples from 20 pre‐ and postoperative patients. By paired comparison, we found that all three lncRNAs levels descended significantly after surgery (Figure 4c), reflecting potential utility in tumor burden monitoring during the therapy period.\nMeanwhile, we explored the diagnostic value of several commonly used tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) in ESCC detection. The ROC curves showed that the AUC values of these markers ranged from 0.51 to 0.53 (Figure 4d), and Table S4 showed that CA199 had the best diagnostic ability with AUC values of 0.53 and 95% CI: 0.40–0.66, which is inferior to lncRNAs.\nFurthermore, we developed logistic regression models for each lncRNA and commonly used tumor markers, and calculated the regression coefficients separately. We subsequently used the regression coefficients as weights to establish the combined index. Interestingly, both combined indices outperformed single lncRNA alone or traditional biomarkers alone in terms of ESCC detection (Table S4). Next, we plotted the ROC curves and compared AUC values by the DeLong's test.\n23\n Index I had a significantly higher AUC value than Index II (p = 0.0045) with an AUC value of 0.84 (95% CI: 0.74–0.91), showing remarkably better diagnostic performance (Figure 4e and Table S4; p = 0.0045). The above results suggested that the combination of these three lncRNAs exhibited superior discriminatory power than traditional biomarkers.\nTo further investigate the diagnostic ability of the three candidate lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) for ESCC, we determined the relative expression levels of these three lncRNAs in serum samples of 40 ESCC patients and 40 healthy individuals and performed ROC curve analysis to evaluate the diagnostic capability for each lncRNA alone. The results showed that the expression levels of these lncRNAs were significantly elevated in ESCC patients (Figure 4a), with AUC values ranging from 0.69 to 0.78 (Figure 4b). Table S4 summarizes in detail the AUC values and 95% CI of each individual lncRNA. The optimal threshold for each lncRNA was determined by the Youden's index.\n22\n Serum lncRNA RASSF8‐AS1 was the best diagnostic marker, with AUC values of 0.78 and 95% CI: 0.67–0.88, respectively.\nTraining phase: A panel of serum lncRNAs‐based diagnostic indicators Index I. (a) The expression levels of the three candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) serum (n = 40) and normal serum samples (n = 40). (b) Receiver operating characteristic (ROC) curves to evaluate the sensitivity and specificity of three candidate lncRNAs to discriminate ESCC from healthy controls. (c) Paired comparison of the three candidate lncRNAs level before and after surgery (n = 20). (d) ROC curves to evaluate the sensitivity and specificity of traditional tumor markers to discriminate ESCC from healthy control. (e) Comparison of the diagnostic performance of Index I and Index II. *p < 0.05, **p < 0.01.\nTo evaluate whether the expression levels of the lncRNAs panel could be used to monitor tumor dynamics, we collected paired serum samples from 20 pre‐ and postoperative patients. By paired comparison, we found that all three lncRNAs levels descended significantly after surgery (Figure 4c), reflecting potential utility in tumor burden monitoring during the therapy period.\nMeanwhile, we explored the diagnostic value of several commonly used tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) in ESCC detection. The ROC curves showed that the AUC values of these markers ranged from 0.51 to 0.53 (Figure 4d), and Table S4 showed that CA199 had the best diagnostic ability with AUC values of 0.53 and 95% CI: 0.40–0.66, which is inferior to lncRNAs.\nFurthermore, we developed logistic regression models for each lncRNA and commonly used tumor markers, and calculated the regression coefficients separately. We subsequently used the regression coefficients as weights to establish the combined index. Interestingly, both combined indices outperformed single lncRNA alone or traditional biomarkers alone in terms of ESCC detection (Table S4). Next, we plotted the ROC curves and compared AUC values by the DeLong's test.\n23\n Index I had a significantly higher AUC value than Index II (p = 0.0045) with an AUC value of 0.84 (95% CI: 0.74–0.91), showing remarkably better diagnostic performance (Figure 4e and Table S4; p = 0.0045). The above results suggested that the combination of these three lncRNAs exhibited superior discriminatory power than traditional biomarkers.\n[SUBTITLE] Validation phase: The lncRNAs based model can distinguish ESCC from healthy control and the levels of lncRNA panel correlated with clinicopathological features [SUBSECTION] We further analyzed the serum lncRNAs levels in 80 ESCC patients and matched healthy samples. Being consistent with the results of the training phase, the expression levels of lncRNAs were significantly higher in ESCC patients than in healthy individuals (Figure 5a). Then, we plotted ROC curves to compare the diagnostic performance between lncRNAs and classic biomarkers, with maximum AUC values of 0.86 and 0.54 for lncRNAs and classic biomarkers, respectively (Figure 5b,c). Table S4 showed in detail the AUC values and 95% CI of lncRNAs and tumor markers during the training phase.\nValidation phase: A panel of serum lncRNAs‐based diagnostic indicators index I. (a) The expression levels of the three candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) (n = 80) and control's serum samples (n = 80). (b) Receiver operating characteristic (ROC) curves of candidate lncRNAs to discriminate ESCC from healthy control. (c) ROC curves of traditional tumor markers. (d) Comparison of the diagnostic performance of Index I and Index II. (e) Kaplan–Meier survival curves of ESCC patients with low and high Index I.**p < 0.01.\nBeing consistent with the results of the training phase, the lncRNAs‐based Index I obtained the greatest diagnostic performance with an AUC value of 0.86 (95% CI: 0.81–0.92) (Table S4). In addition, the AUC values of lncRNAs‐based Index I were significantly outperformed to that from Index II (Figure 5d and Table S4; p < 0.001).\nTo investigate Index I related clinicopathological factors, we collected ESCC patients from the discovery, training, and validation phases and classified them into high and low levels using the median of the Index I index as the threshold value. As shown in Table 1, higher Index I levels were associated with greater depth of infiltration (p = 0.0267), lymph node metastasis (p = 0.0012), advanced tumor stage (p = 0.0040), and poor prognosis (Figure 5e; p < 0.001).\nThe correlation between the serum Index I expression and clinicopathological factors of ESCC\nAbbreviations: ESCC, esophageal squamous cell carcinoma; —, no data available.\nBold values means p‐value < 0.05.\nWe further analyzed the serum lncRNAs levels in 80 ESCC patients and matched healthy samples. Being consistent with the results of the training phase, the expression levels of lncRNAs were significantly higher in ESCC patients than in healthy individuals (Figure 5a). Then, we plotted ROC curves to compare the diagnostic performance between lncRNAs and classic biomarkers, with maximum AUC values of 0.86 and 0.54 for lncRNAs and classic biomarkers, respectively (Figure 5b,c). Table S4 showed in detail the AUC values and 95% CI of lncRNAs and tumor markers during the training phase.\nValidation phase: A panel of serum lncRNAs‐based diagnostic indicators index I. (a) The expression levels of the three candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) (n = 80) and control's serum samples (n = 80). (b) Receiver operating characteristic (ROC) curves of candidate lncRNAs to discriminate ESCC from healthy control. (c) ROC curves of traditional tumor markers. (d) Comparison of the diagnostic performance of Index I and Index II. (e) Kaplan–Meier survival curves of ESCC patients with low and high Index I.**p < 0.01.\nBeing consistent with the results of the training phase, the lncRNAs‐based Index I obtained the greatest diagnostic performance with an AUC value of 0.86 (95% CI: 0.81–0.92) (Table S4). In addition, the AUC values of lncRNAs‐based Index I were significantly outperformed to that from Index II (Figure 5d and Table S4; p < 0.001).\nTo investigate Index I related clinicopathological factors, we collected ESCC patients from the discovery, training, and validation phases and classified them into high and low levels using the median of the Index I index as the threshold value. As shown in Table 1, higher Index I levels were associated with greater depth of infiltration (p = 0.0267), lymph node metastasis (p = 0.0012), advanced tumor stage (p = 0.0040), and poor prognosis (Figure 5e; p < 0.001).\nThe correlation between the serum Index I expression and clinicopathological factors of ESCC\nAbbreviations: ESCC, esophageal squamous cell carcinoma; —, no data available.\nBold values means p‐value < 0.05.\n[SUBTITLE] Prognostic significance of three serum lncRNAs panel [SUBSECTION] We then analyzed the associations between 140 ESCC mortality and both three serum‐derived lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) and classic tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) using a forward conditional logistic regression model. As shown in Table 2, in the univariable analysis, tumor size, RASSF8‐AS1 expression and LINC00958 expression were associated with ESCC mortality. However, in the multivariable analysis, only RASSF8‐AS1 expression and CYFRA21‐1 expression were independently associated with ESCC mortality. The long‐term prognostic values of three serum‐derived lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) and classic tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) were analyzed using a Cox regression model and the results are listed in Table 3. In the univariable analysis, tumor depth, lymph node metastasis and RASSF8‐AS1 expression were significantly associated with long‐term mortality. However, in the multivariable analysis, only RASSF8‐AS1 expression was independently associated with long‐term mortality. In conclusion, RASSF8‐AS1 expression was an independent prognostic factor for both short‐ and long‐term outcomes of ESCC.\nAssociations between three serum‐derived lncRNAs, classic tumor markers and ESCC mortality\nAbbreviations: CI, confidence interval; ESCC, esophageal squamous cell carcinoma; OR, odds ratio; —, no data available.\nUnivariate and multivariate Cox analysis of overall survival in ESCC patients\nAbbreviations: CI, confidence interval; ESCC, esophageal squamous cell carcinoma; HR, hazard ration; —, no data available.\nWe then analyzed the associations between 140 ESCC mortality and both three serum‐derived lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) and classic tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) using a forward conditional logistic regression model. As shown in Table 2, in the univariable analysis, tumor size, RASSF8‐AS1 expression and LINC00958 expression were associated with ESCC mortality. However, in the multivariable analysis, only RASSF8‐AS1 expression and CYFRA21‐1 expression were independently associated with ESCC mortality. The long‐term prognostic values of three serum‐derived lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) and classic tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) were analyzed using a Cox regression model and the results are listed in Table 3. In the univariable analysis, tumor depth, lymph node metastasis and RASSF8‐AS1 expression were significantly associated with long‐term mortality. However, in the multivariable analysis, only RASSF8‐AS1 expression was independently associated with long‐term mortality. In conclusion, RASSF8‐AS1 expression was an independent prognostic factor for both short‐ and long‐term outcomes of ESCC.\nAssociations between three serum‐derived lncRNAs, classic tumor markers and ESCC mortality\nAbbreviations: CI, confidence interval; ESCC, esophageal squamous cell carcinoma; OR, odds ratio; —, no data available.\nUnivariate and multivariate Cox analysis of overall survival in ESCC patients\nAbbreviations: CI, confidence interval; ESCC, esophageal squamous cell carcinoma; HR, hazard ration; —, no data available.\n[SUBTITLE] In vitro analysis of the oncogenic potential of RASSF8‐AS1 and target scanning [SUBSECTION] As lncRNA RASSF8‐AS1 showed good performance for ESCC diagnosis and RASSF8‐AS1 expression was an independent prognostic factor for both short‐ and long‐term outcomes of ESCC. We speculated that RASSF8‐AS1 plays an oncogenic role in ESCC. We then determined that higher RASSF8‐AS1 was associated with clinicopathological factors (Table 4), such as larger tumor size (p < 0.0001), greater depth of infiltration (p < 0.0001), lymph node metastasis (p = 0.0278), and advanced tumor stage (p = 0.0110). The expression of RASSF8‐AS1 tended to decrease significantly after tumor resection surgery in ESCC patients (Figure 4c), and RASSF8‐AS1 was found to be significantly higher in ESCC in the mid‐late stage than in stage I through the GEPIA database (Figure S3 D). The above data suggested that RASSF8‐AS1 plays a protumor role for ESCC.\nThe correlation between the serum RASSF8‐AS1 expression and clinicopathological factors of ESCC\nAbbreviation: ESCC, esophageal squamous cell carcinoma; —, no data available.\nBold values means p‐value < 0.05.\nSubsequently, we explored the oncogenic function of RASSF8‐AS1 by knocking down RASSF8‐AS1 in TE1 and TE13 cells (Figure 6a). In vitro assays showed that the knockdown of RASSF8‐AS1 inhibited cell proliferation (Figure 6b,c), motility and invasiveness (Figure 6d), respectively.\nRASSF8‐AS1 promotes esophageal squamous cell carcinoma (ESCC) cell proliferation, migration and invasion in vitro. (a) The mRNA expression of RASSF8‐AS1 after transfection by two siRNAs. (b, c) Assessment of knockdown of RASSF8‐AS1 on cell growth of TE1 and TE13 cells. (d) Assessment of knockdown of RASSF8‐AS1 on cell migration and invasion. (e) The mRNA expression of RASSF8‐AS1 in normal tissues and ESCC tissues. (f) Kaplan–Meier survival curves of ESCC patients with low and high RASSF8‐AS1 expression. *p < 0.05, **p < 0.01, ***p < 0.001.\nIn addition, we performed qRT‐PCR on 60 pairs of tumor and adjacent normal tissues, and found that RASSF8‐AS1 was likewise markedly elevated in ESCC tissues (Figure 6e). Next, we divided the 60 ESCC samples into RASSF8‐AS1 low expression group (n = 30) and high expression group (n = 30) according to the median level of RASSF8‐AS1 to explore the prognostic significance of RASSF8‐AS1 in ESCC. Kaplan–Meier analysis showed that patients with high RASSF8‐AS1 expression had a better dismal OS (Figure 6f).\nFinally, we screened RASSF8‐AS1 targets by RNA‐sequencing and performed gene ontology (GO) and pathway enrichment analysis. The result found 303 positively regulated (Figure S4 C, D) and 430 negatively regulated targets (Figure 4e,f). GO results suggested RASSF8‐AS1 was positively involved, mainly in immune response and cell migration, and KEGG pathway analysis indicated the positively regulated targets mostly enriched in cell metabolism (Figure S4 E, F), all of which have been proven to be essential for tumor progression.\nAs lncRNA RASSF8‐AS1 showed good performance for ESCC diagnosis and RASSF8‐AS1 expression was an independent prognostic factor for both short‐ and long‐term outcomes of ESCC. We speculated that RASSF8‐AS1 plays an oncogenic role in ESCC. We then determined that higher RASSF8‐AS1 was associated with clinicopathological factors (Table 4), such as larger tumor size (p < 0.0001), greater depth of infiltration (p < 0.0001), lymph node metastasis (p = 0.0278), and advanced tumor stage (p = 0.0110). The expression of RASSF8‐AS1 tended to decrease significantly after tumor resection surgery in ESCC patients (Figure 4c), and RASSF8‐AS1 was found to be significantly higher in ESCC in the mid‐late stage than in stage I through the GEPIA database (Figure S3 D). The above data suggested that RASSF8‐AS1 plays a protumor role for ESCC.\nThe correlation between the serum RASSF8‐AS1 expression and clinicopathological factors of ESCC\nAbbreviation: ESCC, esophageal squamous cell carcinoma; —, no data available.\nBold values means p‐value < 0.05.\nSubsequently, we explored the oncogenic function of RASSF8‐AS1 by knocking down RASSF8‐AS1 in TE1 and TE13 cells (Figure 6a). In vitro assays showed that the knockdown of RASSF8‐AS1 inhibited cell proliferation (Figure 6b,c), motility and invasiveness (Figure 6d), respectively.\nRASSF8‐AS1 promotes esophageal squamous cell carcinoma (ESCC) cell proliferation, migration and invasion in vitro. (a) The mRNA expression of RASSF8‐AS1 after transfection by two siRNAs. (b, c) Assessment of knockdown of RASSF8‐AS1 on cell growth of TE1 and TE13 cells. (d) Assessment of knockdown of RASSF8‐AS1 on cell migration and invasion. (e) The mRNA expression of RASSF8‐AS1 in normal tissues and ESCC tissues. (f) Kaplan–Meier survival curves of ESCC patients with low and high RASSF8‐AS1 expression. *p < 0.05, **p < 0.01, ***p < 0.001.\nIn addition, we performed qRT‐PCR on 60 pairs of tumor and adjacent normal tissues, and found that RASSF8‐AS1 was likewise markedly elevated in ESCC tissues (Figure 6e). Next, we divided the 60 ESCC samples into RASSF8‐AS1 low expression group (n = 30) and high expression group (n = 30) according to the median level of RASSF8‐AS1 to explore the prognostic significance of RASSF8‐AS1 in ESCC. Kaplan–Meier analysis showed that patients with high RASSF8‐AS1 expression had a better dismal OS (Figure 6f).\nFinally, we screened RASSF8‐AS1 targets by RNA‐sequencing and performed gene ontology (GO) and pathway enrichment analysis. The result found 303 positively regulated (Figure S4 C, D) and 430 negatively regulated targets (Figure 4e,f). GO results suggested RASSF8‐AS1 was positively involved, mainly in immune response and cell migration, and KEGG pathway analysis indicated the positively regulated targets mostly enriched in cell metabolism (Figure S4 E, F), all of which have been proven to be essential for tumor progression.", "A total of 280 participants, including 140 ESCC patients and 140 healthy controls were enrolled in the study, as shown in Table S3. To verify whether the exosomes were successfully isolated from serum, we characterized the isolated exosomes using TEM and NTA. The results showed that the isolated exosomes had a bilayer membrane structure (Figure S1 A), and the particle size distribution of exosomes was about 30–150 nm in diameter (Figure S1 B), meeting the criteria of exosome patterns.", "The overall workflow of this study is illustrated in Figure 1a. In the discovery phase, we performed LncRNA microarray analysis on serum exosome samples from four preoperative ESCC patients and matched healthy controls. The top 100 differential lncRNAs identified in serum exosome samples from four preoperative ESCC patients and healthy control are shown in Figure 1b (FDR <0.005, Fold change >2, GEO accession number: GSE192662). Circos plots globally and genome‐widely displayed kinds of detected lncRNAs in ESCC and control serum samples (Figure 1c). A previous microarray comparing five pairs of tumor and paracancerous tissues (GSE89102) was also included for further analysis. To achieve more reliable aberrantly expressed lncRNAs, we adopted a more restrictive filtering criteria (FDR <0.005, fold change >5) for both datasets, and integrated the two datasets to finally obtain five (AC098818.2, RASSF8‐AS1, LINC00958, GMDS‐DT and AL591721.1) candidate genes with significant overexpression in ESCC serum samples and cancerous tissues (Figure 1d).\nMicroarray screening results of differential lncRNAs and function prediction. (a) Experimental design overview. (b) Heatmap results of microarray analysis for four preoperative esophageal squamous cell carcinoma (ESCC) patients and matched normal healthy serum samples. (c) Circos plots of lncRNAs in the human genome (hg19). The outer tracks represent the cytoband ideogram of chromosome. For the two tracks, the outer one (blue) represents the levels of lncRNAs in normal tissues and the inner one (red) represents the levels in ESCC tissues. (d) Venn diagram showing consistent upregulation of five lncRNAs in both microarray analyses. (e, f) Signal pathway networks of mRNAs involved in lncRNAs‐mRNAs relationships.\nTo investigate the potential role of the candidate lncRNAs in ESCC, we analyzed mRNA results from the same microarray chip derived from serum samples and performed pathway analysis (Figure S2 A–D). By lncRNAs‐mRNA coexpression network analysis, most of the candidate lncRNAs‐related mRNAs could be enriched in pathways such as MAPK, Ras, Wnt, metabolic pathways and HIF‐1α, which have been identified to be involved in ESCC carcinogenesis (Figure 1e,f).\nTo validate the microarray data, we examined the expression of five candidate lncRNAs in a pilot cohort containing serum exosome samples (n = 20 for ESCC and healthy control separately), and 30 paired tissues, respectively. The qRT‐PCR results showed that the expression of AC098818.2, RASSF8‐AS1 and LINC00958 being significantly increased in ESCC patients' serum compared with healthy, while no significant difference in the expression of AL591721.1 and GMDS‐DT (Figure 2a). Compared with normal esophageal tissues, AC098818.2, RASSF8‐AS1 and LINC00958 expression was significantly upregulated in tumor tissues, whereas the expression level of GMDS‐DT was significantly downregulated, and no significant difference was seen in the expression of AL591721.1 (Figure 2b). Moreover, we examined the expression levels of these five lncRNAs in ESCC cell lines (TE1, TE13, KYSE140 and KYSE150) and normal epithelial cell HET‐1A. Compared with HET‐1A, the expression of AC098818.2, RASSF8‐AS1 and LINC00958 was upregulated in the ESCC cell lines, while there was no significant difference in the expression of AL591721.1 and GMDS‐DT (Figure 2c). Meanwhile, by big data mining through ExoRbase, we found that AC098818.2, RASSF8‐AS1 and LINC00958 were significantly overexpressed in ESCC blood exosomes compared with healthy donors and other types of cancers (Figure S3 A‐C), indicating excellent specificity in ESCC screening. By searching in the GEPIA database which obtained expression data from TCGA, a gradually increased trend of RASSF8‐AS1 expression toward TNM stages was found (Figure S3 D). Furthermore, we detected expression level of the three candidates in 12 pairs of serum samples and corresponding cancerous tissues, and found moderate correlation for each lncRNA (Figure 2d‐f).\nIdentification of candidate five lncRNAs expression. (a) The expression levels of the five candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) serum (n = 20) and normal serum samples (n = 20). (b) The expression levels of the five candidate lncRNAs in paired tumors and adjacent normal tissues (n = 30). (c) The expression levels of the five candidate lncRNAs in ESCC cell lines (TE1, TE13, KYSE140 and KYSE150) and the human esophageal epithelial cells line (HET‐1A). (d–f) Spearman's correlation analysis between expression levels of lncRNAs in tumor tissues and paired serum samples of ESCC. *p < 0.05, **p < 0.01 .", "Given that stability is a key prerequisite for body fluid tumor biomarkers, we evaluated the stability of exosomal lncRNAs in serum samples under different conditions. Serum samples were subjected to extreme conditions, such as incubation at different temperatures for 3 h, repeated freeze–thaw cycles, RNase A digestion for 3 and 6 h, and exposure to acidic or alkaline environments. Interestingly, these treatments had little or no effect on the level of these serum lncRNAs (Figure 3), indicating the lncRNAs panel was valuable for clinical application.\nStability of plasma lncRNAs under extreme conditions. (a) Incubation at different temperature for 3 h. (b) Repeated freeze–thaw cycles. (c) RNase A digestion for 3 and 6 h. (d) Exposure to acidic or alkaline environments.", "To further investigate the diagnostic ability of the three candidate lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) for ESCC, we determined the relative expression levels of these three lncRNAs in serum samples of 40 ESCC patients and 40 healthy individuals and performed ROC curve analysis to evaluate the diagnostic capability for each lncRNA alone. The results showed that the expression levels of these lncRNAs were significantly elevated in ESCC patients (Figure 4a), with AUC values ranging from 0.69 to 0.78 (Figure 4b). Table S4 summarizes in detail the AUC values and 95% CI of each individual lncRNA. The optimal threshold for each lncRNA was determined by the Youden's index.\n22\n Serum lncRNA RASSF8‐AS1 was the best diagnostic marker, with AUC values of 0.78 and 95% CI: 0.67–0.88, respectively.\nTraining phase: A panel of serum lncRNAs‐based diagnostic indicators Index I. (a) The expression levels of the three candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) serum (n = 40) and normal serum samples (n = 40). (b) Receiver operating characteristic (ROC) curves to evaluate the sensitivity and specificity of three candidate lncRNAs to discriminate ESCC from healthy controls. (c) Paired comparison of the three candidate lncRNAs level before and after surgery (n = 20). (d) ROC curves to evaluate the sensitivity and specificity of traditional tumor markers to discriminate ESCC from healthy control. (e) Comparison of the diagnostic performance of Index I and Index II. *p < 0.05, **p < 0.01.\nTo evaluate whether the expression levels of the lncRNAs panel could be used to monitor tumor dynamics, we collected paired serum samples from 20 pre‐ and postoperative patients. By paired comparison, we found that all three lncRNAs levels descended significantly after surgery (Figure 4c), reflecting potential utility in tumor burden monitoring during the therapy period.\nMeanwhile, we explored the diagnostic value of several commonly used tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) in ESCC detection. The ROC curves showed that the AUC values of these markers ranged from 0.51 to 0.53 (Figure 4d), and Table S4 showed that CA199 had the best diagnostic ability with AUC values of 0.53 and 95% CI: 0.40–0.66, which is inferior to lncRNAs.\nFurthermore, we developed logistic regression models for each lncRNA and commonly used tumor markers, and calculated the regression coefficients separately. We subsequently used the regression coefficients as weights to establish the combined index. Interestingly, both combined indices outperformed single lncRNA alone or traditional biomarkers alone in terms of ESCC detection (Table S4). Next, we plotted the ROC curves and compared AUC values by the DeLong's test.\n23\n Index I had a significantly higher AUC value than Index II (p = 0.0045) with an AUC value of 0.84 (95% CI: 0.74–0.91), showing remarkably better diagnostic performance (Figure 4e and Table S4; p = 0.0045). The above results suggested that the combination of these three lncRNAs exhibited superior discriminatory power than traditional biomarkers.", "We further analyzed the serum lncRNAs levels in 80 ESCC patients and matched healthy samples. Being consistent with the results of the training phase, the expression levels of lncRNAs were significantly higher in ESCC patients than in healthy individuals (Figure 5a). Then, we plotted ROC curves to compare the diagnostic performance between lncRNAs and classic biomarkers, with maximum AUC values of 0.86 and 0.54 for lncRNAs and classic biomarkers, respectively (Figure 5b,c). Table S4 showed in detail the AUC values and 95% CI of lncRNAs and tumor markers during the training phase.\nValidation phase: A panel of serum lncRNAs‐based diagnostic indicators index I. (a) The expression levels of the three candidate lncRNAs in esophageal squamous cell carcinoma (ESCC) (n = 80) and control's serum samples (n = 80). (b) Receiver operating characteristic (ROC) curves of candidate lncRNAs to discriminate ESCC from healthy control. (c) ROC curves of traditional tumor markers. (d) Comparison of the diagnostic performance of Index I and Index II. (e) Kaplan–Meier survival curves of ESCC patients with low and high Index I.**p < 0.01.\nBeing consistent with the results of the training phase, the lncRNAs‐based Index I obtained the greatest diagnostic performance with an AUC value of 0.86 (95% CI: 0.81–0.92) (Table S4). In addition, the AUC values of lncRNAs‐based Index I were significantly outperformed to that from Index II (Figure 5d and Table S4; p < 0.001).\nTo investigate Index I related clinicopathological factors, we collected ESCC patients from the discovery, training, and validation phases and classified them into high and low levels using the median of the Index I index as the threshold value. As shown in Table 1, higher Index I levels were associated with greater depth of infiltration (p = 0.0267), lymph node metastasis (p = 0.0012), advanced tumor stage (p = 0.0040), and poor prognosis (Figure 5e; p < 0.001).\nThe correlation between the serum Index I expression and clinicopathological factors of ESCC\nAbbreviations: ESCC, esophageal squamous cell carcinoma; —, no data available.\nBold values means p‐value < 0.05.", "We then analyzed the associations between 140 ESCC mortality and both three serum‐derived lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) and classic tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) using a forward conditional logistic regression model. As shown in Table 2, in the univariable analysis, tumor size, RASSF8‐AS1 expression and LINC00958 expression were associated with ESCC mortality. However, in the multivariable analysis, only RASSF8‐AS1 expression and CYFRA21‐1 expression were independently associated with ESCC mortality. The long‐term prognostic values of three serum‐derived lncRNAs (AC098818.2, RASSF8‐AS1 and LINC00958) and classic tumor markers (CEA125, CA199, CA153, SCC, NSE, CYFRA21‐1) were analyzed using a Cox regression model and the results are listed in Table 3. In the univariable analysis, tumor depth, lymph node metastasis and RASSF8‐AS1 expression were significantly associated with long‐term mortality. However, in the multivariable analysis, only RASSF8‐AS1 expression was independently associated with long‐term mortality. In conclusion, RASSF8‐AS1 expression was an independent prognostic factor for both short‐ and long‐term outcomes of ESCC.\nAssociations between three serum‐derived lncRNAs, classic tumor markers and ESCC mortality\nAbbreviations: CI, confidence interval; ESCC, esophageal squamous cell carcinoma; OR, odds ratio; —, no data available.\nUnivariate and multivariate Cox analysis of overall survival in ESCC patients\nAbbreviations: CI, confidence interval; ESCC, esophageal squamous cell carcinoma; HR, hazard ration; —, no data available.", "As lncRNA RASSF8‐AS1 showed good performance for ESCC diagnosis and RASSF8‐AS1 expression was an independent prognostic factor for both short‐ and long‐term outcomes of ESCC. We speculated that RASSF8‐AS1 plays an oncogenic role in ESCC. We then determined that higher RASSF8‐AS1 was associated with clinicopathological factors (Table 4), such as larger tumor size (p < 0.0001), greater depth of infiltration (p < 0.0001), lymph node metastasis (p = 0.0278), and advanced tumor stage (p = 0.0110). The expression of RASSF8‐AS1 tended to decrease significantly after tumor resection surgery in ESCC patients (Figure 4c), and RASSF8‐AS1 was found to be significantly higher in ESCC in the mid‐late stage than in stage I through the GEPIA database (Figure S3 D). The above data suggested that RASSF8‐AS1 plays a protumor role for ESCC.\nThe correlation between the serum RASSF8‐AS1 expression and clinicopathological factors of ESCC\nAbbreviation: ESCC, esophageal squamous cell carcinoma; —, no data available.\nBold values means p‐value < 0.05.\nSubsequently, we explored the oncogenic function of RASSF8‐AS1 by knocking down RASSF8‐AS1 in TE1 and TE13 cells (Figure 6a). In vitro assays showed that the knockdown of RASSF8‐AS1 inhibited cell proliferation (Figure 6b,c), motility and invasiveness (Figure 6d), respectively.\nRASSF8‐AS1 promotes esophageal squamous cell carcinoma (ESCC) cell proliferation, migration and invasion in vitro. (a) The mRNA expression of RASSF8‐AS1 after transfection by two siRNAs. (b, c) Assessment of knockdown of RASSF8‐AS1 on cell growth of TE1 and TE13 cells. (d) Assessment of knockdown of RASSF8‐AS1 on cell migration and invasion. (e) The mRNA expression of RASSF8‐AS1 in normal tissues and ESCC tissues. (f) Kaplan–Meier survival curves of ESCC patients with low and high RASSF8‐AS1 expression. *p < 0.05, **p < 0.01, ***p < 0.001.\nIn addition, we performed qRT‐PCR on 60 pairs of tumor and adjacent normal tissues, and found that RASSF8‐AS1 was likewise markedly elevated in ESCC tissues (Figure 6e). Next, we divided the 60 ESCC samples into RASSF8‐AS1 low expression group (n = 30) and high expression group (n = 30) according to the median level of RASSF8‐AS1 to explore the prognostic significance of RASSF8‐AS1 in ESCC. Kaplan–Meier analysis showed that patients with high RASSF8‐AS1 expression had a better dismal OS (Figure 6f).\nFinally, we screened RASSF8‐AS1 targets by RNA‐sequencing and performed gene ontology (GO) and pathway enrichment analysis. The result found 303 positively regulated (Figure S4 C, D) and 430 negatively regulated targets (Figure 4e,f). GO results suggested RASSF8‐AS1 was positively involved, mainly in immune response and cell migration, and KEGG pathway analysis indicated the positively regulated targets mostly enriched in cell metabolism (Figure S4 E, F), all of which have been proven to be essential for tumor progression.", "Delayed diagnosis is the main cause for poor prognosis and rapid progression of ESCC. The five‐year survival rate for patients with early diagnosis of ESCC is >90%, while survival rate with late detection is <20%.\n24\n Endoscopic surveillance and biopsy examination are current applied approaches for ESCC diagnosis and monitoring, but are not popular because of the high cost and level of discomfort, while imaging technology is limited for the low sensitivity, thereby highlighting the urgent need to develop new, relatively noninvasive and reliable tools to improve the early diagnosis of ESCC.\nThere are currently no widely accepted serum biomarkers for ESCC screening, surveillance for treatment response and recurrence. Traditional tumor biomarkers are inadequate in ESCC detection as they have poor sensitivity and specificity.\nIn recent years, the emergence of novel liquid tumor markers has provided a new solution for the early screening of tumors. The discovery of circulating tumor cells (CTC),\n25\n circulating tumor cell DNA (ctDNA),\n26\n circulating cell‐free DNA (cfDNA),\n27\n and exosomes can further improve the efficacy of noninvasive screening. These humoral markers reflect the molecular and genetic information, as well as temporal and spatial heterogeneity of tumors, making the dynamic monitoring of tumor status possible. Among the currently acceptable noninvasive markers, the total amount of CTC is relatively low, while the specificity of ctDNA and cfDNA is insufficient, making the clinical application full of challenges. With the development of exosome isolation and enrichment technologies, exosome diagnosis has become a rising star in the field of liquid biopsy. Benefiting from the protection of the phospholipid bilayer on the surface of exosomes, the exosome contents have good stability, especially the RNAs can be protected by the membrane to avoid being degraded by RNase. The discovery of noncoding RNAs has enriched the understanding of tumorigenesis mechanisms. The role of exosome‐transported noncoding RNAs in tumor metastasis, drug resistance, and radio‐resistance across cells has received increasing attention. Studies have shown that the number of LncRNAs is huge, dozens of times that of currently known mRNAs, with high tissue specificity, which provides ideal conditions for the development of noninvasive screening. It has been reported that serum exosomal biomarkers may be derived from tumor tissues,\n28\n therefore in this study we also incorporated the upregulated lncRNAs from tissue microarray. The linear association between serum and tissue samples for the candidate lncRNAs confirmed the assumption. Finally, we identified a novel panel of exosomal lncRNAs for ESCC liquid biopsy through a multiphase study.\nThe lncRNAs‐based composite Index I showed significantly higher diagnostic power than traditional tumor biomarker based composite Index II in two independent cohorts. Furthermore, the novel lncRNAs signature was stably detectable in serum, supporting the clinically applicable value. We also noted serum lncRNAs declined post‐surgery, indicating that Index I may reflect tumor burden of ESCC patients, which should be used as a follow‐up parameter for ESCC.\nAmong the newly identified lncRNA panel, RASSF8‐AS1 showed the best diagnostic power for all ESCC patients, suggesting its oncogenic role in ESCC. Currently, the function of RASSF8‐AS1 has only been reported in laryngeal squamous cell carcinoma.\n29\n Our results showed its oncogenic role in ESCC for the first time and revealed the underlying mechanisms by target mRNA scanning. Similarly, the role of LIN00958 in ESCC has been recently reported, which also verified the oncogenic role of LIN00958, suggesting our results are reasonable.\n30\n\n\nWe also note the limitations of this study. First, we included only a relatively small number of patients and healthy controls, and the ESCC patients were diagnosed with cancer as they had already been diagnosed pathologically earlier, which should be verified in a much larger sample size. Second, we conducted a retrospective, single‐center, cross‐sectional study, which may have introduced unavoidable selection bias. A prospective multicenter study is needed to further confirm the diagnostic efficiency of Index I, and to check the superiority to traditional biomarkers. Next, how lncRNA enters the exosome, how lncRNA encapsulated exosomes play a role in tumor progress and distant migration call for more attention. In recent years, the role of exosomes in mediating distant tumor metastasis has gained emphasis,\n31\n, \n32\n and our study revealed high expression of serum lncRNA RASSF8‐AS1 was also correlated with metastasis, and our sequencing results suggested its involvement in immune response and cell metabolism. How our newly discovered exosomal lncRNAs play a role in distant metastasis of ESCC deserves further investigation.", "This research was supported by the National Natural Science Foundation for Youth of China (no. 81702444), China Postdoctoral Science Foundation 2018M643884 and 2020T130128ZX.", "All authors declare no conflict of interest.", "\nTable S1 The sequences of qRT‐PCR primers.\n\nTable S2 The sequences of siRNAs.\n\nTable S3 Demographic and clinicopathologic characteristics of the study subjects.\n\nTable S4 Results of ROC curves for individual biomarkers.\nClick here for additional data file.\n\nFigure S1 Characterization of exosomes isolated from serum samples. (A) TEM images of exosomes. HV = 100.0 kV, Direct Mag: 100000× (scale bars 100 nm). Exosomes were winkled oval or spherical in shape under TEM. (B) Size and concentration distribution of exosomes were analyzed by NTA.\nClick here for additional data file.\n\nFigure S2 Bioinformatic analysis of differential mRNAs. (A) Hierarchical clustering of the high‐throughput microarray performed for serum mRNA. (B–D) Gene ontology enrichments of biological process (BP), Cellular component (CC) and molecular function (MF) for differential mRNAs.\nClick here for additional data file.\n\nFigure S3 Big data mining for the candidate lncRNAs. (A–C) Expression of serum exosomal lncRNAs in pan‐cancer compared with healthy donors. (D) Tissues with higher stages exhibit roughly gradually elevated expression of RASSF8‐AS1. *p < 0.05, **p < 0.01, ***p < 0.001.\nClick here for additional data file.\n\nFigure S4 RNA‐sequencing results of RSSF8‐AS1 targets. (A) Volcano plots for the differential mRNAs. (B) Hierarchical Clustering for the differentially expressed RNA. (C, D) Gene Oncology and pathway enrichment analysis for the positively regulated mRNAs of RASSF8‐AS1. (E, F) Oncology and pathway enrichment analysis for the negatively regulated mRNAs of RASSF8‐AS1.\nClick here for additional data file." ]
[ null, "methods", null, null, null, null, null, null, null, null, null, null, "results", null, null, null, null, null, null, null, "discussion", null, "COI-statement", "supplementary-material" ]
[ "circulating lncRNAs", "esophageal squamous cell carcinoma", "exosome; liquid biopsy", "prognosis" ]
Gothenburg Empowerment Scale (GES): psychometric properties and measurement invariance in adults with congenital heart disease from Belgium, Norway and South Korea.
36266608
Patient empowerment is associated with improvements in different patient-reported and clinical outcomes. However, despite being widely researched, high quality and theoretically substantiated disease-generic measures of patient empowerment are lacking. The few good instruments that are available have not reported important psychometric properties, including measurement invariance. The aim of this study was to assess the psychometric properties of the 15-item Gothenburg Empowerment Scale (GES), with a particular focus on measurement invariance of the GES across individuals from three countries.
BACKGROUND
Adults with congenital heart disease from Belgium, Norway and South Korea completed the GES and other patient-reported outcomes as part of an international, cross-sectional, descriptive study called APPROACH-IS II. The scale's content (missing data) and factorial validity (confirmatory factor analyses), measurement invariance (multi-group confirmatory factor analyses), responsiveness (floor and ceiling effects) and reliability (internal consistency) were assessed.
METHODS
Content validity, responsiveness and reliability were confirmed. Nonetheless, metric but not scalar measurement invariance was supported when including the three countries, possibly because the scale performed differently in the sample from South Korea. A second set of analyses supported partial scalar invariance for a sample that was limited to Norway and Belgium.
RESULTS
Our study offers preliminary evidence that GES is a valid and reliable measure of patient empowerment in adults with congenital heart disease. However, cross-country comparisons must be made with caution, given the scale did not perform equivalently across the three countries.
CONCLUSION
[ "Adult", "Humans", "Psychometrics", "Reproducibility of Results", "Surveys and Questionnaires", "Cross-Sectional Studies", "Belgium", "Quality of Life", "Factor Analysis, Statistical", "Republic of Korea", "Heart Defects, Congenital" ]
9583060
null
null
null
null
Results
[SUBTITLE] Content validity [SUBSECTION] The proportion of missing values ranged from 0 to 0.8% (Table 2). “Knowledge and understanding” was the dimension with the highest proportion of missing values. The proportion of missing values ranged from 0 to 0.8% (Table 2). “Knowledge and understanding” was the dimension with the highest proportion of missing values. [SUBTITLE] Factorial validity [SUBSECTION] The five-factor structure as well as the overall factor of patient empowerment of the GES were evaluated through CFA in the entire sample. The five-factor model had an acceptable model fit based on the fit indices (x2(80) = 326.296; CFI = 0.948; RMSEA = 0.060; and SRMR = 0.039). Factor loadings for this model ranged from 0.515 to 0.864 and were all significant with p < 0.001 (Table 2). Item 6 from the “personal control” dimension had the lowest factor loading (i.e., 0.515). A second-order factor model to test the overall construct of patient empowerment also showed an acceptable model fit ( x2(85) = 358.916; CFI = 0.942; RMSEA = 0.062; and SRMR = 0.044). The first-order factor loadings in this model ranged from 0.523 to 0.863 and all were significant (p < 0.001). The second-order factor loadings had the following values: 0.903 (knowledge and understanding), 0.859 (personal control), 0.638 (identity), 0.773 (shared decision-making) and 0.583 (enabling others). Table 2Proportion of missing values and factor loadings for GESItemsMissing valuesn* (%)Factor loadings First-order factor Second-order factor Knowledge and Understanding 1. I know and understand my medical condition7 (0.8)0.7180.7232. I know what to do to stay healthy3 (0.4)0.7750.7653. I know when to contact healthcare providers for my medical condition4 (0.5)0.6320.639 Personal Control 4. I have the skills to manage my medical condition in daily life3 (0.4)0.7790.7735. I have a sense of control over my health6 (0.7)0.6820.6836. I am active in maintaining my health0 (0)0.5150.523 Identity 7. My medical condition is a part of who I am as a person1 (0.1)0.6920.7018. Living with my medical condition makes me stronger as a person3 (0.4)0.6100.5959. I have given my medical condition a place in my life5 (0.6)0.6420.646 Shared decision-making 10. I am capable of expressing to my healthcare providers what is important to me1 (0.1)0.7150.71311. I actively participate in discussions with my health care providers about my health4 (0.5)0.7200.71912. I am capable of making decisions about my health and health care with the healthcare providers2 (0.2)0.7600.762 Enabling Others 13. I have the skills to support other people with a similar medical condition3 (0.4)0.8640.86314. I am able to give helpful advice to people who are struggling with a similar medical condition5 (0.6)0.8530.85315. I can help other people by sharing how I keep myself well1 (0.1)0.7560.757 Proportion of missing values and factor loadings for GES The five-factor structure as well as the overall factor of patient empowerment of the GES were evaluated through CFA in the entire sample. The five-factor model had an acceptable model fit based on the fit indices (x2(80) = 326.296; CFI = 0.948; RMSEA = 0.060; and SRMR = 0.039). Factor loadings for this model ranged from 0.515 to 0.864 and were all significant with p < 0.001 (Table 2). Item 6 from the “personal control” dimension had the lowest factor loading (i.e., 0.515). A second-order factor model to test the overall construct of patient empowerment also showed an acceptable model fit ( x2(85) = 358.916; CFI = 0.942; RMSEA = 0.062; and SRMR = 0.044). The first-order factor loadings in this model ranged from 0.523 to 0.863 and all were significant (p < 0.001). The second-order factor loadings had the following values: 0.903 (knowledge and understanding), 0.859 (personal control), 0.638 (identity), 0.773 (shared decision-making) and 0.583 (enabling others). Table 2Proportion of missing values and factor loadings for GESItemsMissing valuesn* (%)Factor loadings First-order factor Second-order factor Knowledge and Understanding 1. I know and understand my medical condition7 (0.8)0.7180.7232. I know what to do to stay healthy3 (0.4)0.7750.7653. I know when to contact healthcare providers for my medical condition4 (0.5)0.6320.639 Personal Control 4. I have the skills to manage my medical condition in daily life3 (0.4)0.7790.7735. I have a sense of control over my health6 (0.7)0.6820.6836. I am active in maintaining my health0 (0)0.5150.523 Identity 7. My medical condition is a part of who I am as a person1 (0.1)0.6920.7018. Living with my medical condition makes me stronger as a person3 (0.4)0.6100.5959. I have given my medical condition a place in my life5 (0.6)0.6420.646 Shared decision-making 10. I am capable of expressing to my healthcare providers what is important to me1 (0.1)0.7150.71311. I actively participate in discussions with my health care providers about my health4 (0.5)0.7200.71912. I am capable of making decisions about my health and health care with the healthcare providers2 (0.2)0.7600.762 Enabling Others 13. I have the skills to support other people with a similar medical condition3 (0.4)0.8640.86314. I am able to give helpful advice to people who are struggling with a similar medical condition5 (0.6)0.8530.85315. I can help other people by sharing how I keep myself well1 (0.1)0.7560.757 Proportion of missing values and factor loadings for GES [SUBTITLE] Measurement invariance [SUBSECTION] The configural model had a CFI (0.899) and RMSEA (0.085) near the cut-off values for an acceptable model (Table 3). To improve model fit, modification indices were evaluated, and based on this, the residuals of items 6 and 15 were allowed to covary. Even though these items belong to different dimensions (personal control and enabling others, respectively), it is reasonable to expect that persons who are actively involved in their care, also feel more capable of sharing their experiences with others [7]. A second configural model was evaluated with this error correlation, and model fit indices reached the expected threshold (x2(252) = 695.911; CFI = 0.913; RMSEA = 0.079; and SRMR = 0.061). By achieving a well-fitting configural model, we proceeded to test metric and scalar invariance, also including this covariation. The metric model fitted the data well (Table 3). Additionally, changes in model fit indices (∆CFI 0.005; ∆RMSEA 0.002; ∆SRMR 0.006) were within the expected values, indicating metric invariance was supported. An evaluation of scalar invariance came along with slightly worse model fit indices (Table 3). In comparison to the metric model, fit indices were not within the allowed change range (∆CFI 0.034; ∆RMSEA 0.010; ∆SRMR 0.007). Therefore, full scalar invariance could not be established. Given the lack of full scalar invariance, we proceeded to test partial scalar invariance by evaluating modification indices and the equality constraints across groups. Constraints were relaxed sequentially until an acceptable fit was achieved. Fixing the intercept of item 9 (identity) contributed the most to the observed misfit across groups, so this item was estimated freely in a partial scalar invariance model. While model fit improvements were obtained, the indices were below the acceptable threshold. Therefore, a new partial scalar invariance model was tested with the intercepts of items 9 (identity) and 4 (personal control) set free. While improvements were identified (Table 3), model fit indices were still not within an acceptable range. We continued to release items sequentially to achieve acceptable model fit. Models were tested with intercepts of items 9 (identity), 4 (personal control), 12 (shared decision-making) and 2 (knowledge and understanding) unconstrained and while fit indices were almost within the recommended range, it was not possible to achieve acceptable model fit. While models with unconstrained items from the “enabling others” dimension were assessed, none of them led to changes in the model fit indices. Additionally, given that not more than one item per dimension could be estimated freely, models with more than 5 free items where not tested. Therefore, partial scalar invariance was rejected. Since partial scalar invariance was not achieved, an assessment of the three groups independently through CFA was undertaken to understand why this was the case. While fit indices for Norway (x2(85) = 117.694; CFI = 0.964; RMSEA = 0.053; and SRMR = 0.074) and Belgium ( x2(85) = 348.280; CFI = 0.916; RMSEA = 0.079; and SRMR = 0.053) indicated the model fitted these groups well, it appears this was not the case for the data from South Korea (x2(85): 307.753; CFI: 0.788; RMSEA: 0.112; and SRMR: 0.079). Factor loadings for this country ranged between 0.407 and 0.772, with low factor loadings in the “shared decision-making” dimension, though significant (p < 0.001). [SUBTITLE] Measurement invariance: norwegian and belgian samples [SUBSECTION] As the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium. Table 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR Complete sample (Belgium, Norway and South Korea) Configural invariance a 772.074 (255)0.8990.0850.063 Configural invariance b 695.911 (252)0.9130.0790.061 Metric invariance b 750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006 Scalar invariance b 941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007 Partial scalar invariance with intercept of item 9 free b 886.943 (296)0.8850.0840.072 Partial scalar invariance with intercept of items 9 and 4 free b 856.568 (294)0.8900.0820.070 Partial scalar invariance with intercept of items 9, 4 and 12 free b 827.343 (292)0.8960.0800.069 Partial scalar invariance with intercept of items 9, 4, 12 and 2 free b 813.205 (290)0.8980.0800.069 Partial sample (Belgium and Norway) Configural invariance a 463.510 (170)0.9280.0740.057 Metric invariance a 482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004 Scalar invariance a 544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002 Partial scalar invariance with one intercept free c 519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030 Model fit indexes of multi-group confirmatory factor analyses a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030 As the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium. Table 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR Complete sample (Belgium, Norway and South Korea) Configural invariance a 772.074 (255)0.8990.0850.063 Configural invariance b 695.911 (252)0.9130.0790.061 Metric invariance b 750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006 Scalar invariance b 941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007 Partial scalar invariance with intercept of item 9 free b 886.943 (296)0.8850.0840.072 Partial scalar invariance with intercept of items 9 and 4 free b 856.568 (294)0.8900.0820.070 Partial scalar invariance with intercept of items 9, 4 and 12 free b 827.343 (292)0.8960.0800.069 Partial scalar invariance with intercept of items 9, 4, 12 and 2 free b 813.205 (290)0.8980.0800.069 Partial sample (Belgium and Norway) Configural invariance a 463.510 (170)0.9280.0740.057 Metric invariance a 482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004 Scalar invariance a 544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002 Partial scalar invariance with one intercept free c 519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030 Model fit indexes of multi-group confirmatory factor analyses a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030 The configural model had a CFI (0.899) and RMSEA (0.085) near the cut-off values for an acceptable model (Table 3). To improve model fit, modification indices were evaluated, and based on this, the residuals of items 6 and 15 were allowed to covary. Even though these items belong to different dimensions (personal control and enabling others, respectively), it is reasonable to expect that persons who are actively involved in their care, also feel more capable of sharing their experiences with others [7]. A second configural model was evaluated with this error correlation, and model fit indices reached the expected threshold (x2(252) = 695.911; CFI = 0.913; RMSEA = 0.079; and SRMR = 0.061). By achieving a well-fitting configural model, we proceeded to test metric and scalar invariance, also including this covariation. The metric model fitted the data well (Table 3). Additionally, changes in model fit indices (∆CFI 0.005; ∆RMSEA 0.002; ∆SRMR 0.006) were within the expected values, indicating metric invariance was supported. An evaluation of scalar invariance came along with slightly worse model fit indices (Table 3). In comparison to the metric model, fit indices were not within the allowed change range (∆CFI 0.034; ∆RMSEA 0.010; ∆SRMR 0.007). Therefore, full scalar invariance could not be established. Given the lack of full scalar invariance, we proceeded to test partial scalar invariance by evaluating modification indices and the equality constraints across groups. Constraints were relaxed sequentially until an acceptable fit was achieved. Fixing the intercept of item 9 (identity) contributed the most to the observed misfit across groups, so this item was estimated freely in a partial scalar invariance model. While model fit improvements were obtained, the indices were below the acceptable threshold. Therefore, a new partial scalar invariance model was tested with the intercepts of items 9 (identity) and 4 (personal control) set free. While improvements were identified (Table 3), model fit indices were still not within an acceptable range. We continued to release items sequentially to achieve acceptable model fit. Models were tested with intercepts of items 9 (identity), 4 (personal control), 12 (shared decision-making) and 2 (knowledge and understanding) unconstrained and while fit indices were almost within the recommended range, it was not possible to achieve acceptable model fit. While models with unconstrained items from the “enabling others” dimension were assessed, none of them led to changes in the model fit indices. Additionally, given that not more than one item per dimension could be estimated freely, models with more than 5 free items where not tested. Therefore, partial scalar invariance was rejected. Since partial scalar invariance was not achieved, an assessment of the three groups independently through CFA was undertaken to understand why this was the case. While fit indices for Norway (x2(85) = 117.694; CFI = 0.964; RMSEA = 0.053; and SRMR = 0.074) and Belgium ( x2(85) = 348.280; CFI = 0.916; RMSEA = 0.079; and SRMR = 0.053) indicated the model fitted these groups well, it appears this was not the case for the data from South Korea (x2(85): 307.753; CFI: 0.788; RMSEA: 0.112; and SRMR: 0.079). Factor loadings for this country ranged between 0.407 and 0.772, with low factor loadings in the “shared decision-making” dimension, though significant (p < 0.001). [SUBTITLE] Measurement invariance: norwegian and belgian samples [SUBSECTION] As the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium. Table 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR Complete sample (Belgium, Norway and South Korea) Configural invariance a 772.074 (255)0.8990.0850.063 Configural invariance b 695.911 (252)0.9130.0790.061 Metric invariance b 750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006 Scalar invariance b 941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007 Partial scalar invariance with intercept of item 9 free b 886.943 (296)0.8850.0840.072 Partial scalar invariance with intercept of items 9 and 4 free b 856.568 (294)0.8900.0820.070 Partial scalar invariance with intercept of items 9, 4 and 12 free b 827.343 (292)0.8960.0800.069 Partial scalar invariance with intercept of items 9, 4, 12 and 2 free b 813.205 (290)0.8980.0800.069 Partial sample (Belgium and Norway) Configural invariance a 463.510 (170)0.9280.0740.057 Metric invariance a 482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004 Scalar invariance a 544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002 Partial scalar invariance with one intercept free c 519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030 Model fit indexes of multi-group confirmatory factor analyses a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030 As the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium. Table 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR Complete sample (Belgium, Norway and South Korea) Configural invariance a 772.074 (255)0.8990.0850.063 Configural invariance b 695.911 (252)0.9130.0790.061 Metric invariance b 750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006 Scalar invariance b 941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007 Partial scalar invariance with intercept of item 9 free b 886.943 (296)0.8850.0840.072 Partial scalar invariance with intercept of items 9 and 4 free b 856.568 (294)0.8900.0820.070 Partial scalar invariance with intercept of items 9, 4 and 12 free b 827.343 (292)0.8960.0800.069 Partial scalar invariance with intercept of items 9, 4, 12 and 2 free b 813.205 (290)0.8980.0800.069 Partial sample (Belgium and Norway) Configural invariance a 463.510 (170)0.9280.0740.057 Metric invariance a 482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004 Scalar invariance a 544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002 Partial scalar invariance with one intercept free c 519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030 Model fit indexes of multi-group confirmatory factor analyses a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030 [SUBTITLE] Internal consistency [SUBSECTION] The Cronbach’s alpha for the overall scale was 0.873, indicating that it was internally consistent. The alpha values for the subscales were: 0.73 (knowledge and understanding); 0.693 (personal control); 0.680 (identity); 0. 774 (shared decision-making); and 0.863 (enabling others). The Cronbach’s alpha for each country is given in Table 4. An evaluation of the scale’s composite reliability showed that most of the values are above the expected range of 0.7 and those below such threshold were relatively near to this acceptable value. The Cronbach’s alpha for the overall scale was 0.873, indicating that it was internally consistent. The alpha values for the subscales were: 0.73 (knowledge and understanding); 0.693 (personal control); 0.680 (identity); 0. 774 (shared decision-making); and 0.863 (enabling others). The Cronbach’s alpha for each country is given in Table 4. An evaluation of the scale’s composite reliability showed that most of the values are above the expected range of 0.7 and those below such threshold were relatively near to this acceptable value. [SUBTITLE] Responsiveness [SUBSECTION] The mean patient empowerment score for the entire sample was 59.36 ± 8.47. None of the participants had the lowest score (i.e., 15) and only 2.5% had the highest attainable score of 75. Hence, no floor or ceiling effects were identified. Table 4Cronbach’s alpha values, composite reliability and mean empowerment scoresTotal SampleBelgiumNorwaySouth Korea Cronbach’s alpha values Knowledge and understanding 0.7300.7210.7590.728 Personal control 0.6930.6330.6360.751 Identity 0.6800.6820.7510.636 Shared decision-making 0.7740.8330.8040.491 Enabling others 0.8630.8720.9050.782 Overall scale 0.8730.8780.8710.854 Composite reliability Knowledge and understanding 0.7530.7520.7790.732 Personal control 0.7020.6500.6770.753 Identity 0.6840.6820.7700.657 Shared decision-making 0.5590.8350.8050.470 Enabling others 0.8640.8760.9100.782 Overall scale 0.9290.9420.9510.917 Mean patient empowerment scores Knowledge and understanding 12.94 (1.90)13.07 (1.94)12.93 (2.03)12.61 (1.68) Personal control 12.04 (2.10)12.36 (1.97)12.59 (1.85)10.90 (2.12) Identity 11.66 (2.39)11.82 (2.42)11.19 (2.79)11.58 (1.97) Shared decision-making 12.15 (2.26)12.55 (2.22)11.58 (2.75)11.61 (1.74) Enabling others 10.57 (2.83)10.72 (2.90)10.51 (3.12)10.25 (2.40) Overall scale 59.37 (8.47)60.52 (8.53)58.86 (9.07)56.98 (7.33) Cronbach’s alpha values, composite reliability and mean empowerment scores The mean patient empowerment score for the entire sample was 59.36 ± 8.47. None of the participants had the lowest score (i.e., 15) and only 2.5% had the highest attainable score of 75. Hence, no floor or ceiling effects were identified. Table 4Cronbach’s alpha values, composite reliability and mean empowerment scoresTotal SampleBelgiumNorwaySouth Korea Cronbach’s alpha values Knowledge and understanding 0.7300.7210.7590.728 Personal control 0.6930.6330.6360.751 Identity 0.6800.6820.7510.636 Shared decision-making 0.7740.8330.8040.491 Enabling others 0.8630.8720.9050.782 Overall scale 0.8730.8780.8710.854 Composite reliability Knowledge and understanding 0.7530.7520.7790.732 Personal control 0.7020.6500.6770.753 Identity 0.6840.6820.7700.657 Shared decision-making 0.5590.8350.8050.470 Enabling others 0.8640.8760.9100.782 Overall scale 0.9290.9420.9510.917 Mean patient empowerment scores Knowledge and understanding 12.94 (1.90)13.07 (1.94)12.93 (2.03)12.61 (1.68) Personal control 12.04 (2.10)12.36 (1.97)12.59 (1.85)10.90 (2.12) Identity 11.66 (2.39)11.82 (2.42)11.19 (2.79)11.58 (1.97) Shared decision-making 12.15 (2.26)12.55 (2.22)11.58 (2.75)11.61 (1.74) Enabling others 10.57 (2.83)10.72 (2.90)10.51 (3.12)10.25 (2.40) Overall scale 59.37 (8.47)60.52 (8.53)58.86 (9.07)56.98 (7.33) Cronbach’s alpha values, composite reliability and mean empowerment scores
Conclusion
This study provides evidence on the GES’s validity, reliability, and responsiveness in adults with CHD. GES seems to be valid and reliable for the sample of Belgium and Norway. However, this is not the case for South Korea, because shared decision-making seems to have a different meaning in Asian cultures. Hence, cross-cultural comparisons using GES should be made cautiously. It is possible that in countries who are more culturally different from Norway or Belgium, the scale might not perform as well.
[ "Background", "Methods", "Development of GES", "Design", "Sample", "Procedure", "Statistical analyses", "Ethics and informed consent", "Content validity", "Factorial validity", "Measurement invariance", "Measurement invariance: norwegian and belgian samples", "Internal consistency", "Responsiveness", "Methodological considerations" ]
[ "Patient empowerment is a concept associated with increasing peoples’ ability to manage their condition and their lives [1]. It initially emerged in the health promotion field and later demonstrated relevance in the care of persons with chronic conditions (CCs) [2]. It has been suggested that by increasing patient empowerment, patients may eventually become more actively engaged in their care, develop more disease-related knowledge, and improve their quality of life and well-being [3].\nThere is a considerable amount of research on patient empowerment, both theoretically and empirically [4]. However, to date, there is no consensus on how to define patient empowerment or which instrument is best at capturing this construct. There are approximately 30 different definitions available and more than 40 instruments [4]. Moreover, a detailed assessment of current instruments has indicated that a vast majority lack a clear theoretical ground and have poor validity and reliability [5, 6].\nGiven the methodological limitations of existing instruments, a new measure, the Gothenburg Young Persons Empowerment Scale (GYPES), was developed [7]. Scale construction reflected the theoretical work of Small and colleagues who defined patient empowerment as “an enabling process or outcome arising from communication with the healthcare professional and a mutual sharing of resources over information relating to illness, which enhances the patients’ feelings of control, self-efficacy, coping abilities and ability to achieve change over their condition” [8]. From semi-structured interviews with adults with CCs, they concluded that patient empowerment comprises five dimensions [8]. First, knowledge and understanding, related to the level of knowledge patients need to manage their illness and lives. Second, personal control, given that each patient should have the ability to manage their disease. Third, identity, which entails how much the illness influences the patients’ lives and their sense of self. Fourth, shared decision-making, the ability and possibility to make decisions together with the healthcare provider. Fifth, enabling others, referring to the ability to share experiences and coping strategies with other persons who are experiencing a similar situation.\nGYPES was developed for use with adolescents with CCs because at the time there were no existing instruments developed specifically for this age group. The scale’s content and factorial validity, internal consistency, and responsiveness were evaluated in two samples of young persons with congenital heart disease (CHD) and type I diabetes. GYPES was found to have good psychometric properties in these groups [7].\nGiven that the GYPES was found to be valid and reliable for its use with adolescents with CCs, and because it was based on sound conceptual grounds, we developed a slightly modified version to measure empowerment in adults with CCs: the Gothenburg Empowerment Scale (GES). In the evaluation of the scale’s psychometric properties reported herein, we also chose to investigate measurement invariance, given potential use of the GES in international studies. Measurement invariance, also known as measurement equivalence, is often neglected in psychometric evaluation [9], yet it is important to investigate whether an instrument measures the same construct in different contexts. If invariance is not supported, this suggests that the instrument triggers different response mechanisms in different groups, making score comparisons invalid [10]. Invariance is particularly important if cross-cultural comparisons are to be made [11].\nTherefore, the aim of this study was to assess the psychometric properties of the GES, including the level of measurement invariance among adults with CHD from different cultures.", "[SUBTITLE] Development of GES [SUBSECTION] Development of the GES was modeled after the 15-item GYPES, which includes three items each to assess the five dimensions of patient empowerment: (1) personal control; (2) knowledge and understanding; (3) identity; (4) shared decision-making; and (5) enabling others [8]. Scale items are purposefully disease-generic, to facilitate use within different populations of CCs. Originally created in English, the GYPES has currently been translated to other languages including Swedish, Dutch, Mandarin, and Turkish. Additional information on the development and evaluation of GYPES has been published [7].\nThe GES includes the same five dimensions and number of items as GYPES. The only modification was changing the term “young persons” by “persons” throughout the scale. To translate the GES to Dutch, Norwegian and Korean for the present study, the translation process followed the guidelines from the World Health Organization [12]. This process entailed a forward-backward translation, pre-testing the translated questionnaires in a few patients, proofreading the questionnaire and finalizing the translation. To assure consistency, substantial changes from the English version were not permitted.\nDevelopment of the GES was modeled after the 15-item GYPES, which includes three items each to assess the five dimensions of patient empowerment: (1) personal control; (2) knowledge and understanding; (3) identity; (4) shared decision-making; and (5) enabling others [8]. Scale items are purposefully disease-generic, to facilitate use within different populations of CCs. Originally created in English, the GYPES has currently been translated to other languages including Swedish, Dutch, Mandarin, and Turkish. Additional information on the development and evaluation of GYPES has been published [7].\nThe GES includes the same five dimensions and number of items as GYPES. The only modification was changing the term “young persons” by “persons” throughout the scale. To translate the GES to Dutch, Norwegian and Korean for the present study, the translation process followed the guidelines from the World Health Organization [12]. This process entailed a forward-backward translation, pre-testing the translated questionnaires in a few patients, proofreading the questionnaire and finalizing the translation. To assure consistency, substantial changes from the English version were not permitted.\n[SUBTITLE] Design [SUBSECTION] This methodological study is part of a larger international, cross-sectional descriptive study known as APPROACH-IS II, which aims to increase the understanding of patient-reported outcomes in adults with CHD, by enrolling adults with CHD from 32 different countries across the world [13]. APPROACH-IS II is registered at ClinicalTrials.gov: NCT04902768.\nThis methodological study is part of a larger international, cross-sectional descriptive study known as APPROACH-IS II, which aims to increase the understanding of patient-reported outcomes in adults with CHD, by enrolling adults with CHD from 32 different countries across the world [13]. APPROACH-IS II is registered at ClinicalTrials.gov: NCT04902768.\n[SUBTITLE] Sample [SUBSECTION] APPROACH-IS II is enrolling participants from over 50 adult CHD centers worldwide. Data collection is scheduled to be finished by the end of August 2022. For the present study, data from participants of Belgium, Norway and South Korea were included, since the data collection of these three countries was undertaken before the covid-19 pandemic.\nParticipants were eligible for the study if they fulfilled the following criteria: (i) diagnosis of CHD; (ii) aged 18 years or older at the moment of inclusion; (iii) diagnosed before the age of 10 years, (because we wanted participants to have experience living with CHD); (iv) in follow-up at an adult CHD center or included in a national/regional registry; and (v) having the physical, cognitive and language abilities to complete the self-report questionnaires. Patients with prior heart transplantation were ineligible.\nIn the present study, there are a total of 850 people enrolled, 497 were from Belgium, 144 from Norway, and 209 from South Korea. The median age for the total sample was 30 years, and the proportion of men and women was fairly equal. Moderate CHD was found in 56.4% of the participants. Demographic and clinical information of the included participants is detailed in Table 1. There were significant differences in age (p < 0.001, eta square: 0.044) and complexity (p < 0.001, Cramer’s V: 0.227) between the samples, with moderate effect sizes. Indeed, the Norwegian sample was slightly older, and the proportion of severe CHD was larger in South Korea.\n\nTable 1Sociodemographic and clinical aspectsTotal Sample(n = 850)Belgium(n = 497)Norway(n = 144)South Korea(n = 209)\nAge at inclusion (median, (Q1;Q3))\n30.0 (27;43)29.0 (27;38)40.0 (30;53)31.0 (24;45)\nSex (n (%))\nFemaleMale430 (51.0)413 (49.0)240 (48.3)253 (50.9)86 (60.1)57 (39.9)104(50.2)103 (49.8)\nEducation (n (%))\nNo high school educationHigh schoolBachelor’s degreeMaster’s degree or higher447 (5.2)346 (41.2)258 (30.8)190 (22.6)29 (5.9)194 (39.6)160 (32.7)106 (21.6)10 (6.9)60 (41.7)33 (22.9)41 (28.5)5 (2.4)92 (44.9)65 (31.7)43 (21.0)\nCHD complexity (n (%))\nMildModerateSevere116 (13.8)473 (56.4)249 (29.7)98 (19.7)313 (63.0)86 (17.3)8 (6.0)82 (61.7)43 (32.3)10 (4.8)78 (37.5)120 (57.7)\n\nSociodemographic and clinical aspects\n\nSex (n (%))\n\nFemale\nMale\n430 (51.0)\n413 (49.0)\n240 (48.3)\n253 (50.9)\n86 (60.1)\n57 (39.9)\n104(50.2)\n103 (49.8)\n\nEducation (n (%))\n\nNo high school education\nHigh school\nBachelor’s degree\nMaster’s degree or higher\n447 (5.2)\n346 (41.2)\n258 (30.8)\n190 (22.6)\n29 (5.9)\n194 (39.6)\n160 (32.7)\n106 (21.6)\n10 (6.9)\n60 (41.7)\n33 (22.9)\n41 (28.5)\n5 (2.4)\n92 (44.9)\n65 (31.7)\n43 (21.0)\n\nCHD complexity (n (%))\n\nMild\nModerate\nSevere\n116 (13.8)\n473 (56.4)\n249 (29.7)\n98 (19.7)\n313 (63.0)\n86 (17.3)\n8 (6.0)\n82 (61.7)\n43 (32.3)\n10 (4.8)\n78 (37.5)\n120 (57.7)\nAPPROACH-IS II is enrolling participants from over 50 adult CHD centers worldwide. Data collection is scheduled to be finished by the end of August 2022. For the present study, data from participants of Belgium, Norway and South Korea were included, since the data collection of these three countries was undertaken before the covid-19 pandemic.\nParticipants were eligible for the study if they fulfilled the following criteria: (i) diagnosis of CHD; (ii) aged 18 years or older at the moment of inclusion; (iii) diagnosed before the age of 10 years, (because we wanted participants to have experience living with CHD); (iv) in follow-up at an adult CHD center or included in a national/regional registry; and (v) having the physical, cognitive and language abilities to complete the self-report questionnaires. Patients with prior heart transplantation were ineligible.\nIn the present study, there are a total of 850 people enrolled, 497 were from Belgium, 144 from Norway, and 209 from South Korea. The median age for the total sample was 30 years, and the proportion of men and women was fairly equal. Moderate CHD was found in 56.4% of the participants. Demographic and clinical information of the included participants is detailed in Table 1. There were significant differences in age (p < 0.001, eta square: 0.044) and complexity (p < 0.001, Cramer’s V: 0.227) between the samples, with moderate effect sizes. Indeed, the Norwegian sample was slightly older, and the proportion of severe CHD was larger in South Korea.\n\nTable 1Sociodemographic and clinical aspectsTotal Sample(n = 850)Belgium(n = 497)Norway(n = 144)South Korea(n = 209)\nAge at inclusion (median, (Q1;Q3))\n30.0 (27;43)29.0 (27;38)40.0 (30;53)31.0 (24;45)\nSex (n (%))\nFemaleMale430 (51.0)413 (49.0)240 (48.3)253 (50.9)86 (60.1)57 (39.9)104(50.2)103 (49.8)\nEducation (n (%))\nNo high school educationHigh schoolBachelor’s degreeMaster’s degree or higher447 (5.2)346 (41.2)258 (30.8)190 (22.6)29 (5.9)194 (39.6)160 (32.7)106 (21.6)10 (6.9)60 (41.7)33 (22.9)41 (28.5)5 (2.4)92 (44.9)65 (31.7)43 (21.0)\nCHD complexity (n (%))\nMildModerateSevere116 (13.8)473 (56.4)249 (29.7)98 (19.7)313 (63.0)86 (17.3)8 (6.0)82 (61.7)43 (32.3)10 (4.8)78 (37.5)120 (57.7)\n\nSociodemographic and clinical aspects\n\nSex (n (%))\n\nFemale\nMale\n430 (51.0)\n413 (49.0)\n240 (48.3)\n253 (50.9)\n86 (60.1)\n57 (39.9)\n104(50.2)\n103 (49.8)\n\nEducation (n (%))\n\nNo high school education\nHigh school\nBachelor’s degree\nMaster’s degree or higher\n447 (5.2)\n346 (41.2)\n258 (30.8)\n190 (22.6)\n29 (5.9)\n194 (39.6)\n160 (32.7)\n106 (21.6)\n10 (6.9)\n60 (41.7)\n33 (22.9)\n41 (28.5)\n5 (2.4)\n92 (44.9)\n65 (31.7)\n43 (21.0)\n\nCHD complexity (n (%))\n\nMild\nModerate\nSevere\n116 (13.8)\n473 (56.4)\n249 (29.7)\n98 (19.7)\n313 (63.0)\n86 (17.3)\n8 (6.0)\n82 (61.7)\n43 (32.3)\n10 (4.8)\n78 (37.5)\n120 (57.7)\n[SUBTITLE] Procedure [SUBSECTION] Participants completed a set of self-reported questionnaires, including the GES, that were administered during outpatient clinic visits (in Norway, South Korea and Belgium) and/or mailed to their home (in Belgium). Data for this sub-study were collected between August 2019 and February 2020, hence before the COVID-19 pandemic emerged.\nParticipants completed a set of self-reported questionnaires, including the GES, that were administered during outpatient clinic visits (in Norway, South Korea and Belgium) and/or mailed to their home (in Belgium). Data for this sub-study were collected between August 2019 and February 2020, hence before the COVID-19 pandemic emerged.\n[SUBTITLE] Statistical analyses [SUBSECTION] The psychometric evaluation of GES entailed an assessment of the content validity, factorial validity, measurement invariance, internal consistency and responsiveness. Content validity was assessed by the proportion of missing values and invalid scores [7]. While this is not a common approach for evaluating content validity, missing values can be considered an indicator of whether the items in the scale are perceived as relevant by the participants and whether the items are intelligible. Less than 5% missing data was deemed acceptable in this study.\nFactorial validity was evaluated through confirmatory factor analyses (CFA) to assess the hypothesized five-factor structure of the scale and the overall factor of patient empowerment [14]. CFA was performed for the entire sample. A good model fit was obtained if the comparative fit index (CFI) was > 0.90, standardized root mean square residual (SRMR) < 0.08 and root mean square error of approximation (RMSEA) < 0.08 [15]. These fit indices were chosen based on Kline’s suggestion for fit evaluation of CFA [14]. Standardized factor loadings are also reported.\nMeasurement invariance was examined through multigroup CFA (MGCFA) across countries [11]. The MGCFA included three models: (1) configural model (i.e., separates the sample into three subgroups, but no parameter constraints are imposed); (2) metric model (i.e., constrains the factor loadings to be equal across subgroups); and (3) scalar model (i.e., constrains the factor loadings and the item intercepts to be equal across subgroups) [16]. These models are tested sequentially and the process begins with a configural model that is well-fitting. Measurement invariance is based on how well the model fits the data as indicated by the fit indices mentioned before (i.e., CFI, RMSEA, SRMR). Additionally, change fit statistics are used to determine whether measurement invariance is present or not. This change refers to how the fit indices increase or decrease as more constraints are added. As per current recommendations, decreases below 0.01 in CFI (∆CFI) and increases below 0.015 for RMSEA (∆RMSEA) and below 0.03 for SRMR (∆SRMR) are deemed acceptable [17]. If measurement invariance was not achieved at some point, modification indices were assessed to determine whether model fit could be improved or partial invariance could be established [18]. Partial invariance occurs when some items that are different across groups are estimated freely, while keeping at least two indicators per latent construct to be equal across groups [9, 10, 19]. For the GES, this meant that only one item per dimension could be freed to attempt to establish partial invariance. If items were released, this was done following a backward method, based on the items with the highest modification indices [10]. Lastly, if no configural, metric or scalar invariance could be achieved, the factor structure was assessed separately in each group [18].\nInternal consistency was assessed by calculating the Cronbach’s alpha coefficient [20]. Coefficients were calculated for each dimension and for the overall scale. Besides Cronbach’s alpha coefficients, composite reliability values were calculated as an additional method to evaluate the inter-item consistency of the scale. The Cronbach’s alpha value as well as the composite reliability values should be above 0.70 to be considered acceptable [21]. Floor and ceiling effects were calculated as a way to assess issues regarding responsiveness. This is an indirect way to evaluate a scale’s sensitivity to detect change [22]. Floor and ceiling effects were considered present if more than 15% of the participants achieved the lowest (i.e. 15) or highest score (i.e. 75) [22].\nStatistical analyses were performed with the Lavaan package in R [23] and IBM SPSS Statistics for Windows version 27.\nThe psychometric evaluation of GES entailed an assessment of the content validity, factorial validity, measurement invariance, internal consistency and responsiveness. Content validity was assessed by the proportion of missing values and invalid scores [7]. While this is not a common approach for evaluating content validity, missing values can be considered an indicator of whether the items in the scale are perceived as relevant by the participants and whether the items are intelligible. Less than 5% missing data was deemed acceptable in this study.\nFactorial validity was evaluated through confirmatory factor analyses (CFA) to assess the hypothesized five-factor structure of the scale and the overall factor of patient empowerment [14]. CFA was performed for the entire sample. A good model fit was obtained if the comparative fit index (CFI) was > 0.90, standardized root mean square residual (SRMR) < 0.08 and root mean square error of approximation (RMSEA) < 0.08 [15]. These fit indices were chosen based on Kline’s suggestion for fit evaluation of CFA [14]. Standardized factor loadings are also reported.\nMeasurement invariance was examined through multigroup CFA (MGCFA) across countries [11]. The MGCFA included three models: (1) configural model (i.e., separates the sample into three subgroups, but no parameter constraints are imposed); (2) metric model (i.e., constrains the factor loadings to be equal across subgroups); and (3) scalar model (i.e., constrains the factor loadings and the item intercepts to be equal across subgroups) [16]. These models are tested sequentially and the process begins with a configural model that is well-fitting. Measurement invariance is based on how well the model fits the data as indicated by the fit indices mentioned before (i.e., CFI, RMSEA, SRMR). Additionally, change fit statistics are used to determine whether measurement invariance is present or not. This change refers to how the fit indices increase or decrease as more constraints are added. As per current recommendations, decreases below 0.01 in CFI (∆CFI) and increases below 0.015 for RMSEA (∆RMSEA) and below 0.03 for SRMR (∆SRMR) are deemed acceptable [17]. If measurement invariance was not achieved at some point, modification indices were assessed to determine whether model fit could be improved or partial invariance could be established [18]. Partial invariance occurs when some items that are different across groups are estimated freely, while keeping at least two indicators per latent construct to be equal across groups [9, 10, 19]. For the GES, this meant that only one item per dimension could be freed to attempt to establish partial invariance. If items were released, this was done following a backward method, based on the items with the highest modification indices [10]. Lastly, if no configural, metric or scalar invariance could be achieved, the factor structure was assessed separately in each group [18].\nInternal consistency was assessed by calculating the Cronbach’s alpha coefficient [20]. Coefficients were calculated for each dimension and for the overall scale. Besides Cronbach’s alpha coefficients, composite reliability values were calculated as an additional method to evaluate the inter-item consistency of the scale. The Cronbach’s alpha value as well as the composite reliability values should be above 0.70 to be considered acceptable [21]. Floor and ceiling effects were calculated as a way to assess issues regarding responsiveness. This is an indirect way to evaluate a scale’s sensitivity to detect change [22]. Floor and ceiling effects were considered present if more than 15% of the participants achieved the lowest (i.e. 15) or highest score (i.e. 75) [22].\nStatistical analyses were performed with the Lavaan package in R [23] and IBM SPSS Statistics for Windows version 27.\n[SUBTITLE] Ethics and informed consent [SUBSECTION] APPROACH-IS II has its coordinating center at KU Leuven, Belgium. Therefore, ethical approval was granted from the Institutional Review Board of the University Hospitals Leuven/KU Leuven. Additionally, ethics approval was granted by the local ethics committees of the included centers (i.e., Norway and South Korea). All participants included in this study provided verbal and written informed consent.\nAPPROACH-IS II has its coordinating center at KU Leuven, Belgium. Therefore, ethical approval was granted from the Institutional Review Board of the University Hospitals Leuven/KU Leuven. Additionally, ethics approval was granted by the local ethics committees of the included centers (i.e., Norway and South Korea). All participants included in this study provided verbal and written informed consent.", "Development of the GES was modeled after the 15-item GYPES, which includes three items each to assess the five dimensions of patient empowerment: (1) personal control; (2) knowledge and understanding; (3) identity; (4) shared decision-making; and (5) enabling others [8]. Scale items are purposefully disease-generic, to facilitate use within different populations of CCs. Originally created in English, the GYPES has currently been translated to other languages including Swedish, Dutch, Mandarin, and Turkish. Additional information on the development and evaluation of GYPES has been published [7].\nThe GES includes the same five dimensions and number of items as GYPES. The only modification was changing the term “young persons” by “persons” throughout the scale. To translate the GES to Dutch, Norwegian and Korean for the present study, the translation process followed the guidelines from the World Health Organization [12]. This process entailed a forward-backward translation, pre-testing the translated questionnaires in a few patients, proofreading the questionnaire and finalizing the translation. To assure consistency, substantial changes from the English version were not permitted.", "This methodological study is part of a larger international, cross-sectional descriptive study known as APPROACH-IS II, which aims to increase the understanding of patient-reported outcomes in adults with CHD, by enrolling adults with CHD from 32 different countries across the world [13]. APPROACH-IS II is registered at ClinicalTrials.gov: NCT04902768.", "APPROACH-IS II is enrolling participants from over 50 adult CHD centers worldwide. Data collection is scheduled to be finished by the end of August 2022. For the present study, data from participants of Belgium, Norway and South Korea were included, since the data collection of these three countries was undertaken before the covid-19 pandemic.\nParticipants were eligible for the study if they fulfilled the following criteria: (i) diagnosis of CHD; (ii) aged 18 years or older at the moment of inclusion; (iii) diagnosed before the age of 10 years, (because we wanted participants to have experience living with CHD); (iv) in follow-up at an adult CHD center or included in a national/regional registry; and (v) having the physical, cognitive and language abilities to complete the self-report questionnaires. Patients with prior heart transplantation were ineligible.\nIn the present study, there are a total of 850 people enrolled, 497 were from Belgium, 144 from Norway, and 209 from South Korea. The median age for the total sample was 30 years, and the proportion of men and women was fairly equal. Moderate CHD was found in 56.4% of the participants. Demographic and clinical information of the included participants is detailed in Table 1. There were significant differences in age (p < 0.001, eta square: 0.044) and complexity (p < 0.001, Cramer’s V: 0.227) between the samples, with moderate effect sizes. Indeed, the Norwegian sample was slightly older, and the proportion of severe CHD was larger in South Korea.\n\nTable 1Sociodemographic and clinical aspectsTotal Sample(n = 850)Belgium(n = 497)Norway(n = 144)South Korea(n = 209)\nAge at inclusion (median, (Q1;Q3))\n30.0 (27;43)29.0 (27;38)40.0 (30;53)31.0 (24;45)\nSex (n (%))\nFemaleMale430 (51.0)413 (49.0)240 (48.3)253 (50.9)86 (60.1)57 (39.9)104(50.2)103 (49.8)\nEducation (n (%))\nNo high school educationHigh schoolBachelor’s degreeMaster’s degree or higher447 (5.2)346 (41.2)258 (30.8)190 (22.6)29 (5.9)194 (39.6)160 (32.7)106 (21.6)10 (6.9)60 (41.7)33 (22.9)41 (28.5)5 (2.4)92 (44.9)65 (31.7)43 (21.0)\nCHD complexity (n (%))\nMildModerateSevere116 (13.8)473 (56.4)249 (29.7)98 (19.7)313 (63.0)86 (17.3)8 (6.0)82 (61.7)43 (32.3)10 (4.8)78 (37.5)120 (57.7)\n\nSociodemographic and clinical aspects\n\nSex (n (%))\n\nFemale\nMale\n430 (51.0)\n413 (49.0)\n240 (48.3)\n253 (50.9)\n86 (60.1)\n57 (39.9)\n104(50.2)\n103 (49.8)\n\nEducation (n (%))\n\nNo high school education\nHigh school\nBachelor’s degree\nMaster’s degree or higher\n447 (5.2)\n346 (41.2)\n258 (30.8)\n190 (22.6)\n29 (5.9)\n194 (39.6)\n160 (32.7)\n106 (21.6)\n10 (6.9)\n60 (41.7)\n33 (22.9)\n41 (28.5)\n5 (2.4)\n92 (44.9)\n65 (31.7)\n43 (21.0)\n\nCHD complexity (n (%))\n\nMild\nModerate\nSevere\n116 (13.8)\n473 (56.4)\n249 (29.7)\n98 (19.7)\n313 (63.0)\n86 (17.3)\n8 (6.0)\n82 (61.7)\n43 (32.3)\n10 (4.8)\n78 (37.5)\n120 (57.7)", "Participants completed a set of self-reported questionnaires, including the GES, that were administered during outpatient clinic visits (in Norway, South Korea and Belgium) and/or mailed to their home (in Belgium). Data for this sub-study were collected between August 2019 and February 2020, hence before the COVID-19 pandemic emerged.", "The psychometric evaluation of GES entailed an assessment of the content validity, factorial validity, measurement invariance, internal consistency and responsiveness. Content validity was assessed by the proportion of missing values and invalid scores [7]. While this is not a common approach for evaluating content validity, missing values can be considered an indicator of whether the items in the scale are perceived as relevant by the participants and whether the items are intelligible. Less than 5% missing data was deemed acceptable in this study.\nFactorial validity was evaluated through confirmatory factor analyses (CFA) to assess the hypothesized five-factor structure of the scale and the overall factor of patient empowerment [14]. CFA was performed for the entire sample. A good model fit was obtained if the comparative fit index (CFI) was > 0.90, standardized root mean square residual (SRMR) < 0.08 and root mean square error of approximation (RMSEA) < 0.08 [15]. These fit indices were chosen based on Kline’s suggestion for fit evaluation of CFA [14]. Standardized factor loadings are also reported.\nMeasurement invariance was examined through multigroup CFA (MGCFA) across countries [11]. The MGCFA included three models: (1) configural model (i.e., separates the sample into three subgroups, but no parameter constraints are imposed); (2) metric model (i.e., constrains the factor loadings to be equal across subgroups); and (3) scalar model (i.e., constrains the factor loadings and the item intercepts to be equal across subgroups) [16]. These models are tested sequentially and the process begins with a configural model that is well-fitting. Measurement invariance is based on how well the model fits the data as indicated by the fit indices mentioned before (i.e., CFI, RMSEA, SRMR). Additionally, change fit statistics are used to determine whether measurement invariance is present or not. This change refers to how the fit indices increase or decrease as more constraints are added. As per current recommendations, decreases below 0.01 in CFI (∆CFI) and increases below 0.015 for RMSEA (∆RMSEA) and below 0.03 for SRMR (∆SRMR) are deemed acceptable [17]. If measurement invariance was not achieved at some point, modification indices were assessed to determine whether model fit could be improved or partial invariance could be established [18]. Partial invariance occurs when some items that are different across groups are estimated freely, while keeping at least two indicators per latent construct to be equal across groups [9, 10, 19]. For the GES, this meant that only one item per dimension could be freed to attempt to establish partial invariance. If items were released, this was done following a backward method, based on the items with the highest modification indices [10]. Lastly, if no configural, metric or scalar invariance could be achieved, the factor structure was assessed separately in each group [18].\nInternal consistency was assessed by calculating the Cronbach’s alpha coefficient [20]. Coefficients were calculated for each dimension and for the overall scale. Besides Cronbach’s alpha coefficients, composite reliability values were calculated as an additional method to evaluate the inter-item consistency of the scale. The Cronbach’s alpha value as well as the composite reliability values should be above 0.70 to be considered acceptable [21]. Floor and ceiling effects were calculated as a way to assess issues regarding responsiveness. This is an indirect way to evaluate a scale’s sensitivity to detect change [22]. Floor and ceiling effects were considered present if more than 15% of the participants achieved the lowest (i.e. 15) or highest score (i.e. 75) [22].\nStatistical analyses were performed with the Lavaan package in R [23] and IBM SPSS Statistics for Windows version 27.", "APPROACH-IS II has its coordinating center at KU Leuven, Belgium. Therefore, ethical approval was granted from the Institutional Review Board of the University Hospitals Leuven/KU Leuven. Additionally, ethics approval was granted by the local ethics committees of the included centers (i.e., Norway and South Korea). All participants included in this study provided verbal and written informed consent.", "The proportion of missing values ranged from 0 to 0.8% (Table 2). “Knowledge and understanding” was the dimension with the highest proportion of missing values.", "The five-factor structure as well as the overall factor of patient empowerment of the GES were evaluated through CFA in the entire sample. The five-factor model had an acceptable model fit based on the fit indices (x2(80) = 326.296; CFI = 0.948; RMSEA = 0.060; and SRMR = 0.039). Factor loadings for this model ranged from 0.515 to 0.864 and were all significant with p < 0.001 (Table 2). Item 6 from the “personal control” dimension had the lowest factor loading (i.e., 0.515). A second-order factor model to test the overall construct of patient empowerment also showed an acceptable model fit ( x2(85) = 358.916; CFI = 0.942; RMSEA = 0.062; and SRMR = 0.044). The first-order factor loadings in this model ranged from 0.523 to 0.863 and all were significant (p < 0.001). The second-order factor loadings had the following values: 0.903 (knowledge and understanding), 0.859 (personal control), 0.638 (identity), 0.773 (shared decision-making) and 0.583 (enabling others).\n\nTable 2Proportion of missing values and factor loadings for GESItemsMissing valuesn* (%)Factor loadings\nFirst-order factor\n\nSecond-order factor\n\nKnowledge and Understanding\n1. I know and understand my medical condition7 (0.8)0.7180.7232. I know what to do to stay healthy3 (0.4)0.7750.7653. I know when to contact healthcare providers for my medical condition4 (0.5)0.6320.639\nPersonal Control\n4. I have the skills to manage my medical condition in daily life3 (0.4)0.7790.7735. I have a sense of control over my health6 (0.7)0.6820.6836. I am active in maintaining my health0 (0)0.5150.523\nIdentity\n7. My medical condition is a part of who I am as a person1 (0.1)0.6920.7018. Living with my medical condition makes me stronger as a person3 (0.4)0.6100.5959. I have given my medical condition a place in my life5 (0.6)0.6420.646\nShared decision-making\n10. I am capable of expressing to my healthcare providers what is important to me1 (0.1)0.7150.71311. I actively participate in discussions with my health care providers about my health4 (0.5)0.7200.71912. I am capable of making decisions about my health and health care with the healthcare providers2 (0.2)0.7600.762\nEnabling Others\n13. I have the skills to support other people with a similar medical condition3 (0.4)0.8640.86314. I am able to give helpful advice to people who are struggling with a similar medical condition5 (0.6)0.8530.85315. I can help other people by sharing how I keep myself well1 (0.1)0.7560.757\n\nProportion of missing values and factor loadings for GES", "The configural model had a CFI (0.899) and RMSEA (0.085) near the cut-off values for an acceptable model (Table 3). To improve model fit, modification indices were evaluated, and based on this, the residuals of items 6 and 15 were allowed to covary. Even though these items belong to different dimensions (personal control and enabling others, respectively), it is reasonable to expect that persons who are actively involved in their care, also feel more capable of sharing their experiences with others [7]. A second configural model was evaluated with this error correlation, and model fit indices reached the expected threshold (x2(252) = 695.911; CFI = 0.913; RMSEA = 0.079; and SRMR = 0.061). By achieving a well-fitting configural model, we proceeded to test metric and scalar invariance, also including this covariation.\nThe metric model fitted the data well (Table 3). Additionally, changes in model fit indices (∆CFI 0.005; ∆RMSEA 0.002; ∆SRMR 0.006) were within the expected values, indicating metric invariance was supported. An evaluation of scalar invariance came along with slightly worse model fit indices (Table 3). In comparison to the metric model, fit indices were not within the allowed change range (∆CFI 0.034; ∆RMSEA 0.010; ∆SRMR 0.007). Therefore, full scalar invariance could not be established.\nGiven the lack of full scalar invariance, we proceeded to test partial scalar invariance by evaluating modification indices and the equality constraints across groups. Constraints were relaxed sequentially until an acceptable fit was achieved. Fixing the intercept of item 9 (identity) contributed the most to the observed misfit across groups, so this item was estimated freely in a partial scalar invariance model. While model fit improvements were obtained, the indices were below the acceptable threshold. Therefore, a new partial scalar invariance model was tested with the intercepts of items 9 (identity) and 4 (personal control) set free. While improvements were identified (Table 3), model fit indices were still not within an acceptable range. We continued to release items sequentially to achieve acceptable model fit. Models were tested with intercepts of items 9 (identity), 4 (personal control), 12 (shared decision-making) and 2 (knowledge and understanding) unconstrained and while fit indices were almost within the recommended range, it was not possible to achieve acceptable model fit. While models with unconstrained items from the “enabling others” dimension were assessed, none of them led to changes in the model fit indices. Additionally, given that not more than one item per dimension could be estimated freely, models with more than 5 free items where not tested. Therefore, partial scalar invariance was rejected.\nSince partial scalar invariance was not achieved, an assessment of the three groups independently through CFA was undertaken to understand why this was the case. While fit indices for Norway (x2(85) = 117.694; CFI = 0.964; RMSEA = 0.053; and SRMR = 0.074) and Belgium ( x2(85) = 348.280; CFI = 0.916; RMSEA = 0.079; and SRMR = 0.053) indicated the model fitted these groups well, it appears this was not the case for the data from South Korea (x2(85): 307.753; CFI: 0.788; RMSEA: 0.112; and SRMR: 0.079). Factor loadings for this country ranged between 0.407 and 0.772, with low factor loadings in the “shared decision-making” dimension, though significant (p < 0.001).\n[SUBTITLE] Measurement invariance: norwegian and belgian samples [SUBSECTION] As the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium.\n\nTable 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR\nComplete sample (Belgium, Norway and South Korea)\n\nConfigural invariance\na\n772.074 (255)0.8990.0850.063\nConfigural invariance\nb\n695.911 (252)0.9130.0790.061\nMetric invariance\nb\n750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006\nScalar invariance\nb\n941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007\nPartial scalar invariance with intercept of item 9 free\nb\n886.943 (296)0.8850.0840.072\nPartial scalar invariance with intercept of items 9 and 4 free\nb\n856.568 (294)0.8900.0820.070\nPartial scalar invariance with intercept of items 9, 4 and 12 free\nb\n827.343 (292)0.8960.0800.069\nPartial scalar invariance with intercept of items 9, 4, 12 and 2 free\nb\n813.205 (290)0.8980.0800.069\nPartial sample (Belgium and Norway)\n\nConfigural invariance\na\n463.510 (170)0.9280.0740.057\nMetric invariance\na\n482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004\nScalar invariance\na\n544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002\nPartial scalar invariance with one intercept free\nc\n519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\n\nModel fit indexes of multi-group confirmatory factor analyses\na Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\nAs the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium.\n\nTable 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR\nComplete sample (Belgium, Norway and South Korea)\n\nConfigural invariance\na\n772.074 (255)0.8990.0850.063\nConfigural invariance\nb\n695.911 (252)0.9130.0790.061\nMetric invariance\nb\n750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006\nScalar invariance\nb\n941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007\nPartial scalar invariance with intercept of item 9 free\nb\n886.943 (296)0.8850.0840.072\nPartial scalar invariance with intercept of items 9 and 4 free\nb\n856.568 (294)0.8900.0820.070\nPartial scalar invariance with intercept of items 9, 4 and 12 free\nb\n827.343 (292)0.8960.0800.069\nPartial scalar invariance with intercept of items 9, 4, 12 and 2 free\nb\n813.205 (290)0.8980.0800.069\nPartial sample (Belgium and Norway)\n\nConfigural invariance\na\n463.510 (170)0.9280.0740.057\nMetric invariance\na\n482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004\nScalar invariance\na\n544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002\nPartial scalar invariance with one intercept free\nc\n519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\n\nModel fit indexes of multi-group confirmatory factor analyses\na Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030", "As the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium.\n\nTable 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR\nComplete sample (Belgium, Norway and South Korea)\n\nConfigural invariance\na\n772.074 (255)0.8990.0850.063\nConfigural invariance\nb\n695.911 (252)0.9130.0790.061\nMetric invariance\nb\n750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006\nScalar invariance\nb\n941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007\nPartial scalar invariance with intercept of item 9 free\nb\n886.943 (296)0.8850.0840.072\nPartial scalar invariance with intercept of items 9 and 4 free\nb\n856.568 (294)0.8900.0820.070\nPartial scalar invariance with intercept of items 9, 4 and 12 free\nb\n827.343 (292)0.8960.0800.069\nPartial scalar invariance with intercept of items 9, 4, 12 and 2 free\nb\n813.205 (290)0.8980.0800.069\nPartial sample (Belgium and Norway)\n\nConfigural invariance\na\n463.510 (170)0.9280.0740.057\nMetric invariance\na\n482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004\nScalar invariance\na\n544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002\nPartial scalar invariance with one intercept free\nc\n519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\n\nModel fit indexes of multi-group confirmatory factor analyses\na Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030", "The Cronbach’s alpha for the overall scale was 0.873, indicating that it was internally consistent. The alpha values for the subscales were: 0.73 (knowledge and understanding); 0.693 (personal control); 0.680 (identity); 0. 774 (shared decision-making); and 0.863 (enabling others). The Cronbach’s alpha for each country is given in Table 4. An evaluation of the scale’s composite reliability showed that most of the values are above the expected range of 0.7 and those below such threshold were relatively near to this acceptable value.", "The mean patient empowerment score for the entire sample was 59.36 ± 8.47. None of the participants had the lowest score (i.e., 15) and only 2.5% had the highest attainable score of 75. Hence, no floor or ceiling effects were identified.\n\nTable 4Cronbach’s alpha values, composite reliability and mean empowerment scoresTotal SampleBelgiumNorwaySouth Korea\nCronbach’s alpha values\n\nKnowledge and understanding\n0.7300.7210.7590.728\nPersonal control\n0.6930.6330.6360.751\nIdentity\n0.6800.6820.7510.636\nShared decision-making\n0.7740.8330.8040.491\nEnabling others\n0.8630.8720.9050.782\nOverall scale\n0.8730.8780.8710.854\nComposite reliability\n\nKnowledge and understanding\n0.7530.7520.7790.732\nPersonal control\n0.7020.6500.6770.753\nIdentity\n0.6840.6820.7700.657\nShared decision-making\n0.5590.8350.8050.470\nEnabling others\n0.8640.8760.9100.782\nOverall scale\n0.9290.9420.9510.917\nMean patient empowerment scores\n\nKnowledge and understanding\n12.94 (1.90)13.07 (1.94)12.93 (2.03)12.61 (1.68)\nPersonal control\n12.04 (2.10)12.36 (1.97)12.59 (1.85)10.90 (2.12)\nIdentity\n11.66 (2.39)11.82 (2.42)11.19 (2.79)11.58 (1.97)\nShared decision-making\n12.15 (2.26)12.55 (2.22)11.58 (2.75)11.61 (1.74)\nEnabling others\n10.57 (2.83)10.72 (2.90)10.51 (3.12)10.25 (2.40)\nOverall scale\n59.37 (8.47)60.52 (8.53)58.86 (9.07)56.98 (7.33)\n\nCronbach’s alpha values, composite reliability and mean empowerment scores", "There are several strengths of the study that merit mentioning. First, GES development was based on a previously validated questionnaire, which in turn has a strong theoretical foundation and followed a rigorous development process. Therefore, it is plausible to conclude that GES also has a strong theoretical foundation. Second, data were collected as part of an international study, which allows for cross-country comparisons. Third, the data were collected before the COVID-19 pandemic, therefore there was no concern about the potential for survey responses to be impacted by the pandemic [33].\nThere are, however, also some methodological limitations associated with this study. First, some aspects associated with validity and reliability were not assessed. For instance, it was not possible to directly evaluate responsiveness with cross-sectional data. While floor and ceiling effects are an indirect way to measure responsiveness, this is better achieved through longitudinal studies. Future longitudinal studies should evaluate this psychometric property. Second, the study only includes individuals with CHD, who were in current follow-up and who had the abilities to answer the questionnaires. Therefore, the generalizability of the results might be limited." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Development of GES", "Design", "Sample", "Procedure", "Statistical analyses", "Ethics and informed consent", "Results", "Content validity", "Factorial validity", "Measurement invariance", "Measurement invariance: norwegian and belgian samples", "Internal consistency", "Responsiveness", "Discussion", "Methodological considerations", "Conclusion" ]
[ "Patient empowerment is a concept associated with increasing peoples’ ability to manage their condition and their lives [1]. It initially emerged in the health promotion field and later demonstrated relevance in the care of persons with chronic conditions (CCs) [2]. It has been suggested that by increasing patient empowerment, patients may eventually become more actively engaged in their care, develop more disease-related knowledge, and improve their quality of life and well-being [3].\nThere is a considerable amount of research on patient empowerment, both theoretically and empirically [4]. However, to date, there is no consensus on how to define patient empowerment or which instrument is best at capturing this construct. There are approximately 30 different definitions available and more than 40 instruments [4]. Moreover, a detailed assessment of current instruments has indicated that a vast majority lack a clear theoretical ground and have poor validity and reliability [5, 6].\nGiven the methodological limitations of existing instruments, a new measure, the Gothenburg Young Persons Empowerment Scale (GYPES), was developed [7]. Scale construction reflected the theoretical work of Small and colleagues who defined patient empowerment as “an enabling process or outcome arising from communication with the healthcare professional and a mutual sharing of resources over information relating to illness, which enhances the patients’ feelings of control, self-efficacy, coping abilities and ability to achieve change over their condition” [8]. From semi-structured interviews with adults with CCs, they concluded that patient empowerment comprises five dimensions [8]. First, knowledge and understanding, related to the level of knowledge patients need to manage their illness and lives. Second, personal control, given that each patient should have the ability to manage their disease. Third, identity, which entails how much the illness influences the patients’ lives and their sense of self. Fourth, shared decision-making, the ability and possibility to make decisions together with the healthcare provider. Fifth, enabling others, referring to the ability to share experiences and coping strategies with other persons who are experiencing a similar situation.\nGYPES was developed for use with adolescents with CCs because at the time there were no existing instruments developed specifically for this age group. The scale’s content and factorial validity, internal consistency, and responsiveness were evaluated in two samples of young persons with congenital heart disease (CHD) and type I diabetes. GYPES was found to have good psychometric properties in these groups [7].\nGiven that the GYPES was found to be valid and reliable for its use with adolescents with CCs, and because it was based on sound conceptual grounds, we developed a slightly modified version to measure empowerment in adults with CCs: the Gothenburg Empowerment Scale (GES). In the evaluation of the scale’s psychometric properties reported herein, we also chose to investigate measurement invariance, given potential use of the GES in international studies. Measurement invariance, also known as measurement equivalence, is often neglected in psychometric evaluation [9], yet it is important to investigate whether an instrument measures the same construct in different contexts. If invariance is not supported, this suggests that the instrument triggers different response mechanisms in different groups, making score comparisons invalid [10]. Invariance is particularly important if cross-cultural comparisons are to be made [11].\nTherefore, the aim of this study was to assess the psychometric properties of the GES, including the level of measurement invariance among adults with CHD from different cultures.", "[SUBTITLE] Development of GES [SUBSECTION] Development of the GES was modeled after the 15-item GYPES, which includes three items each to assess the five dimensions of patient empowerment: (1) personal control; (2) knowledge and understanding; (3) identity; (4) shared decision-making; and (5) enabling others [8]. Scale items are purposefully disease-generic, to facilitate use within different populations of CCs. Originally created in English, the GYPES has currently been translated to other languages including Swedish, Dutch, Mandarin, and Turkish. Additional information on the development and evaluation of GYPES has been published [7].\nThe GES includes the same five dimensions and number of items as GYPES. The only modification was changing the term “young persons” by “persons” throughout the scale. To translate the GES to Dutch, Norwegian and Korean for the present study, the translation process followed the guidelines from the World Health Organization [12]. This process entailed a forward-backward translation, pre-testing the translated questionnaires in a few patients, proofreading the questionnaire and finalizing the translation. To assure consistency, substantial changes from the English version were not permitted.\nDevelopment of the GES was modeled after the 15-item GYPES, which includes three items each to assess the five dimensions of patient empowerment: (1) personal control; (2) knowledge and understanding; (3) identity; (4) shared decision-making; and (5) enabling others [8]. Scale items are purposefully disease-generic, to facilitate use within different populations of CCs. Originally created in English, the GYPES has currently been translated to other languages including Swedish, Dutch, Mandarin, and Turkish. Additional information on the development and evaluation of GYPES has been published [7].\nThe GES includes the same five dimensions and number of items as GYPES. The only modification was changing the term “young persons” by “persons” throughout the scale. To translate the GES to Dutch, Norwegian and Korean for the present study, the translation process followed the guidelines from the World Health Organization [12]. This process entailed a forward-backward translation, pre-testing the translated questionnaires in a few patients, proofreading the questionnaire and finalizing the translation. To assure consistency, substantial changes from the English version were not permitted.\n[SUBTITLE] Design [SUBSECTION] This methodological study is part of a larger international, cross-sectional descriptive study known as APPROACH-IS II, which aims to increase the understanding of patient-reported outcomes in adults with CHD, by enrolling adults with CHD from 32 different countries across the world [13]. APPROACH-IS II is registered at ClinicalTrials.gov: NCT04902768.\nThis methodological study is part of a larger international, cross-sectional descriptive study known as APPROACH-IS II, which aims to increase the understanding of patient-reported outcomes in adults with CHD, by enrolling adults with CHD from 32 different countries across the world [13]. APPROACH-IS II is registered at ClinicalTrials.gov: NCT04902768.\n[SUBTITLE] Sample [SUBSECTION] APPROACH-IS II is enrolling participants from over 50 adult CHD centers worldwide. Data collection is scheduled to be finished by the end of August 2022. For the present study, data from participants of Belgium, Norway and South Korea were included, since the data collection of these three countries was undertaken before the covid-19 pandemic.\nParticipants were eligible for the study if they fulfilled the following criteria: (i) diagnosis of CHD; (ii) aged 18 years or older at the moment of inclusion; (iii) diagnosed before the age of 10 years, (because we wanted participants to have experience living with CHD); (iv) in follow-up at an adult CHD center or included in a national/regional registry; and (v) having the physical, cognitive and language abilities to complete the self-report questionnaires. Patients with prior heart transplantation were ineligible.\nIn the present study, there are a total of 850 people enrolled, 497 were from Belgium, 144 from Norway, and 209 from South Korea. The median age for the total sample was 30 years, and the proportion of men and women was fairly equal. Moderate CHD was found in 56.4% of the participants. Demographic and clinical information of the included participants is detailed in Table 1. There were significant differences in age (p < 0.001, eta square: 0.044) and complexity (p < 0.001, Cramer’s V: 0.227) between the samples, with moderate effect sizes. Indeed, the Norwegian sample was slightly older, and the proportion of severe CHD was larger in South Korea.\n\nTable 1Sociodemographic and clinical aspectsTotal Sample(n = 850)Belgium(n = 497)Norway(n = 144)South Korea(n = 209)\nAge at inclusion (median, (Q1;Q3))\n30.0 (27;43)29.0 (27;38)40.0 (30;53)31.0 (24;45)\nSex (n (%))\nFemaleMale430 (51.0)413 (49.0)240 (48.3)253 (50.9)86 (60.1)57 (39.9)104(50.2)103 (49.8)\nEducation (n (%))\nNo high school educationHigh schoolBachelor’s degreeMaster’s degree or higher447 (5.2)346 (41.2)258 (30.8)190 (22.6)29 (5.9)194 (39.6)160 (32.7)106 (21.6)10 (6.9)60 (41.7)33 (22.9)41 (28.5)5 (2.4)92 (44.9)65 (31.7)43 (21.0)\nCHD complexity (n (%))\nMildModerateSevere116 (13.8)473 (56.4)249 (29.7)98 (19.7)313 (63.0)86 (17.3)8 (6.0)82 (61.7)43 (32.3)10 (4.8)78 (37.5)120 (57.7)\n\nSociodemographic and clinical aspects\n\nSex (n (%))\n\nFemale\nMale\n430 (51.0)\n413 (49.0)\n240 (48.3)\n253 (50.9)\n86 (60.1)\n57 (39.9)\n104(50.2)\n103 (49.8)\n\nEducation (n (%))\n\nNo high school education\nHigh school\nBachelor’s degree\nMaster’s degree or higher\n447 (5.2)\n346 (41.2)\n258 (30.8)\n190 (22.6)\n29 (5.9)\n194 (39.6)\n160 (32.7)\n106 (21.6)\n10 (6.9)\n60 (41.7)\n33 (22.9)\n41 (28.5)\n5 (2.4)\n92 (44.9)\n65 (31.7)\n43 (21.0)\n\nCHD complexity (n (%))\n\nMild\nModerate\nSevere\n116 (13.8)\n473 (56.4)\n249 (29.7)\n98 (19.7)\n313 (63.0)\n86 (17.3)\n8 (6.0)\n82 (61.7)\n43 (32.3)\n10 (4.8)\n78 (37.5)\n120 (57.7)\nAPPROACH-IS II is enrolling participants from over 50 adult CHD centers worldwide. Data collection is scheduled to be finished by the end of August 2022. For the present study, data from participants of Belgium, Norway and South Korea were included, since the data collection of these three countries was undertaken before the covid-19 pandemic.\nParticipants were eligible for the study if they fulfilled the following criteria: (i) diagnosis of CHD; (ii) aged 18 years or older at the moment of inclusion; (iii) diagnosed before the age of 10 years, (because we wanted participants to have experience living with CHD); (iv) in follow-up at an adult CHD center or included in a national/regional registry; and (v) having the physical, cognitive and language abilities to complete the self-report questionnaires. Patients with prior heart transplantation were ineligible.\nIn the present study, there are a total of 850 people enrolled, 497 were from Belgium, 144 from Norway, and 209 from South Korea. The median age for the total sample was 30 years, and the proportion of men and women was fairly equal. Moderate CHD was found in 56.4% of the participants. Demographic and clinical information of the included participants is detailed in Table 1. There were significant differences in age (p < 0.001, eta square: 0.044) and complexity (p < 0.001, Cramer’s V: 0.227) between the samples, with moderate effect sizes. Indeed, the Norwegian sample was slightly older, and the proportion of severe CHD was larger in South Korea.\n\nTable 1Sociodemographic and clinical aspectsTotal Sample(n = 850)Belgium(n = 497)Norway(n = 144)South Korea(n = 209)\nAge at inclusion (median, (Q1;Q3))\n30.0 (27;43)29.0 (27;38)40.0 (30;53)31.0 (24;45)\nSex (n (%))\nFemaleMale430 (51.0)413 (49.0)240 (48.3)253 (50.9)86 (60.1)57 (39.9)104(50.2)103 (49.8)\nEducation (n (%))\nNo high school educationHigh schoolBachelor’s degreeMaster’s degree or higher447 (5.2)346 (41.2)258 (30.8)190 (22.6)29 (5.9)194 (39.6)160 (32.7)106 (21.6)10 (6.9)60 (41.7)33 (22.9)41 (28.5)5 (2.4)92 (44.9)65 (31.7)43 (21.0)\nCHD complexity (n (%))\nMildModerateSevere116 (13.8)473 (56.4)249 (29.7)98 (19.7)313 (63.0)86 (17.3)8 (6.0)82 (61.7)43 (32.3)10 (4.8)78 (37.5)120 (57.7)\n\nSociodemographic and clinical aspects\n\nSex (n (%))\n\nFemale\nMale\n430 (51.0)\n413 (49.0)\n240 (48.3)\n253 (50.9)\n86 (60.1)\n57 (39.9)\n104(50.2)\n103 (49.8)\n\nEducation (n (%))\n\nNo high school education\nHigh school\nBachelor’s degree\nMaster’s degree or higher\n447 (5.2)\n346 (41.2)\n258 (30.8)\n190 (22.6)\n29 (5.9)\n194 (39.6)\n160 (32.7)\n106 (21.6)\n10 (6.9)\n60 (41.7)\n33 (22.9)\n41 (28.5)\n5 (2.4)\n92 (44.9)\n65 (31.7)\n43 (21.0)\n\nCHD complexity (n (%))\n\nMild\nModerate\nSevere\n116 (13.8)\n473 (56.4)\n249 (29.7)\n98 (19.7)\n313 (63.0)\n86 (17.3)\n8 (6.0)\n82 (61.7)\n43 (32.3)\n10 (4.8)\n78 (37.5)\n120 (57.7)\n[SUBTITLE] Procedure [SUBSECTION] Participants completed a set of self-reported questionnaires, including the GES, that were administered during outpatient clinic visits (in Norway, South Korea and Belgium) and/or mailed to their home (in Belgium). Data for this sub-study were collected between August 2019 and February 2020, hence before the COVID-19 pandemic emerged.\nParticipants completed a set of self-reported questionnaires, including the GES, that were administered during outpatient clinic visits (in Norway, South Korea and Belgium) and/or mailed to their home (in Belgium). Data for this sub-study were collected between August 2019 and February 2020, hence before the COVID-19 pandemic emerged.\n[SUBTITLE] Statistical analyses [SUBSECTION] The psychometric evaluation of GES entailed an assessment of the content validity, factorial validity, measurement invariance, internal consistency and responsiveness. Content validity was assessed by the proportion of missing values and invalid scores [7]. While this is not a common approach for evaluating content validity, missing values can be considered an indicator of whether the items in the scale are perceived as relevant by the participants and whether the items are intelligible. Less than 5% missing data was deemed acceptable in this study.\nFactorial validity was evaluated through confirmatory factor analyses (CFA) to assess the hypothesized five-factor structure of the scale and the overall factor of patient empowerment [14]. CFA was performed for the entire sample. A good model fit was obtained if the comparative fit index (CFI) was > 0.90, standardized root mean square residual (SRMR) < 0.08 and root mean square error of approximation (RMSEA) < 0.08 [15]. These fit indices were chosen based on Kline’s suggestion for fit evaluation of CFA [14]. Standardized factor loadings are also reported.\nMeasurement invariance was examined through multigroup CFA (MGCFA) across countries [11]. The MGCFA included three models: (1) configural model (i.e., separates the sample into three subgroups, but no parameter constraints are imposed); (2) metric model (i.e., constrains the factor loadings to be equal across subgroups); and (3) scalar model (i.e., constrains the factor loadings and the item intercepts to be equal across subgroups) [16]. These models are tested sequentially and the process begins with a configural model that is well-fitting. Measurement invariance is based on how well the model fits the data as indicated by the fit indices mentioned before (i.e., CFI, RMSEA, SRMR). Additionally, change fit statistics are used to determine whether measurement invariance is present or not. This change refers to how the fit indices increase or decrease as more constraints are added. As per current recommendations, decreases below 0.01 in CFI (∆CFI) and increases below 0.015 for RMSEA (∆RMSEA) and below 0.03 for SRMR (∆SRMR) are deemed acceptable [17]. If measurement invariance was not achieved at some point, modification indices were assessed to determine whether model fit could be improved or partial invariance could be established [18]. Partial invariance occurs when some items that are different across groups are estimated freely, while keeping at least two indicators per latent construct to be equal across groups [9, 10, 19]. For the GES, this meant that only one item per dimension could be freed to attempt to establish partial invariance. If items were released, this was done following a backward method, based on the items with the highest modification indices [10]. Lastly, if no configural, metric or scalar invariance could be achieved, the factor structure was assessed separately in each group [18].\nInternal consistency was assessed by calculating the Cronbach’s alpha coefficient [20]. Coefficients were calculated for each dimension and for the overall scale. Besides Cronbach’s alpha coefficients, composite reliability values were calculated as an additional method to evaluate the inter-item consistency of the scale. The Cronbach’s alpha value as well as the composite reliability values should be above 0.70 to be considered acceptable [21]. Floor and ceiling effects were calculated as a way to assess issues regarding responsiveness. This is an indirect way to evaluate a scale’s sensitivity to detect change [22]. Floor and ceiling effects were considered present if more than 15% of the participants achieved the lowest (i.e. 15) or highest score (i.e. 75) [22].\nStatistical analyses were performed with the Lavaan package in R [23] and IBM SPSS Statistics for Windows version 27.\nThe psychometric evaluation of GES entailed an assessment of the content validity, factorial validity, measurement invariance, internal consistency and responsiveness. Content validity was assessed by the proportion of missing values and invalid scores [7]. While this is not a common approach for evaluating content validity, missing values can be considered an indicator of whether the items in the scale are perceived as relevant by the participants and whether the items are intelligible. Less than 5% missing data was deemed acceptable in this study.\nFactorial validity was evaluated through confirmatory factor analyses (CFA) to assess the hypothesized five-factor structure of the scale and the overall factor of patient empowerment [14]. CFA was performed for the entire sample. A good model fit was obtained if the comparative fit index (CFI) was > 0.90, standardized root mean square residual (SRMR) < 0.08 and root mean square error of approximation (RMSEA) < 0.08 [15]. These fit indices were chosen based on Kline’s suggestion for fit evaluation of CFA [14]. Standardized factor loadings are also reported.\nMeasurement invariance was examined through multigroup CFA (MGCFA) across countries [11]. The MGCFA included three models: (1) configural model (i.e., separates the sample into three subgroups, but no parameter constraints are imposed); (2) metric model (i.e., constrains the factor loadings to be equal across subgroups); and (3) scalar model (i.e., constrains the factor loadings and the item intercepts to be equal across subgroups) [16]. These models are tested sequentially and the process begins with a configural model that is well-fitting. Measurement invariance is based on how well the model fits the data as indicated by the fit indices mentioned before (i.e., CFI, RMSEA, SRMR). Additionally, change fit statistics are used to determine whether measurement invariance is present or not. This change refers to how the fit indices increase or decrease as more constraints are added. As per current recommendations, decreases below 0.01 in CFI (∆CFI) and increases below 0.015 for RMSEA (∆RMSEA) and below 0.03 for SRMR (∆SRMR) are deemed acceptable [17]. If measurement invariance was not achieved at some point, modification indices were assessed to determine whether model fit could be improved or partial invariance could be established [18]. Partial invariance occurs when some items that are different across groups are estimated freely, while keeping at least two indicators per latent construct to be equal across groups [9, 10, 19]. For the GES, this meant that only one item per dimension could be freed to attempt to establish partial invariance. If items were released, this was done following a backward method, based on the items with the highest modification indices [10]. Lastly, if no configural, metric or scalar invariance could be achieved, the factor structure was assessed separately in each group [18].\nInternal consistency was assessed by calculating the Cronbach’s alpha coefficient [20]. Coefficients were calculated for each dimension and for the overall scale. Besides Cronbach’s alpha coefficients, composite reliability values were calculated as an additional method to evaluate the inter-item consistency of the scale. The Cronbach’s alpha value as well as the composite reliability values should be above 0.70 to be considered acceptable [21]. Floor and ceiling effects were calculated as a way to assess issues regarding responsiveness. This is an indirect way to evaluate a scale’s sensitivity to detect change [22]. Floor and ceiling effects were considered present if more than 15% of the participants achieved the lowest (i.e. 15) or highest score (i.e. 75) [22].\nStatistical analyses were performed with the Lavaan package in R [23] and IBM SPSS Statistics for Windows version 27.\n[SUBTITLE] Ethics and informed consent [SUBSECTION] APPROACH-IS II has its coordinating center at KU Leuven, Belgium. Therefore, ethical approval was granted from the Institutional Review Board of the University Hospitals Leuven/KU Leuven. Additionally, ethics approval was granted by the local ethics committees of the included centers (i.e., Norway and South Korea). All participants included in this study provided verbal and written informed consent.\nAPPROACH-IS II has its coordinating center at KU Leuven, Belgium. Therefore, ethical approval was granted from the Institutional Review Board of the University Hospitals Leuven/KU Leuven. Additionally, ethics approval was granted by the local ethics committees of the included centers (i.e., Norway and South Korea). All participants included in this study provided verbal and written informed consent.", "Development of the GES was modeled after the 15-item GYPES, which includes three items each to assess the five dimensions of patient empowerment: (1) personal control; (2) knowledge and understanding; (3) identity; (4) shared decision-making; and (5) enabling others [8]. Scale items are purposefully disease-generic, to facilitate use within different populations of CCs. Originally created in English, the GYPES has currently been translated to other languages including Swedish, Dutch, Mandarin, and Turkish. Additional information on the development and evaluation of GYPES has been published [7].\nThe GES includes the same five dimensions and number of items as GYPES. The only modification was changing the term “young persons” by “persons” throughout the scale. To translate the GES to Dutch, Norwegian and Korean for the present study, the translation process followed the guidelines from the World Health Organization [12]. This process entailed a forward-backward translation, pre-testing the translated questionnaires in a few patients, proofreading the questionnaire and finalizing the translation. To assure consistency, substantial changes from the English version were not permitted.", "This methodological study is part of a larger international, cross-sectional descriptive study known as APPROACH-IS II, which aims to increase the understanding of patient-reported outcomes in adults with CHD, by enrolling adults with CHD from 32 different countries across the world [13]. APPROACH-IS II is registered at ClinicalTrials.gov: NCT04902768.", "APPROACH-IS II is enrolling participants from over 50 adult CHD centers worldwide. Data collection is scheduled to be finished by the end of August 2022. For the present study, data from participants of Belgium, Norway and South Korea were included, since the data collection of these three countries was undertaken before the covid-19 pandemic.\nParticipants were eligible for the study if they fulfilled the following criteria: (i) diagnosis of CHD; (ii) aged 18 years or older at the moment of inclusion; (iii) diagnosed before the age of 10 years, (because we wanted participants to have experience living with CHD); (iv) in follow-up at an adult CHD center or included in a national/regional registry; and (v) having the physical, cognitive and language abilities to complete the self-report questionnaires. Patients with prior heart transplantation were ineligible.\nIn the present study, there are a total of 850 people enrolled, 497 were from Belgium, 144 from Norway, and 209 from South Korea. The median age for the total sample was 30 years, and the proportion of men and women was fairly equal. Moderate CHD was found in 56.4% of the participants. Demographic and clinical information of the included participants is detailed in Table 1. There were significant differences in age (p < 0.001, eta square: 0.044) and complexity (p < 0.001, Cramer’s V: 0.227) between the samples, with moderate effect sizes. Indeed, the Norwegian sample was slightly older, and the proportion of severe CHD was larger in South Korea.\n\nTable 1Sociodemographic and clinical aspectsTotal Sample(n = 850)Belgium(n = 497)Norway(n = 144)South Korea(n = 209)\nAge at inclusion (median, (Q1;Q3))\n30.0 (27;43)29.0 (27;38)40.0 (30;53)31.0 (24;45)\nSex (n (%))\nFemaleMale430 (51.0)413 (49.0)240 (48.3)253 (50.9)86 (60.1)57 (39.9)104(50.2)103 (49.8)\nEducation (n (%))\nNo high school educationHigh schoolBachelor’s degreeMaster’s degree or higher447 (5.2)346 (41.2)258 (30.8)190 (22.6)29 (5.9)194 (39.6)160 (32.7)106 (21.6)10 (6.9)60 (41.7)33 (22.9)41 (28.5)5 (2.4)92 (44.9)65 (31.7)43 (21.0)\nCHD complexity (n (%))\nMildModerateSevere116 (13.8)473 (56.4)249 (29.7)98 (19.7)313 (63.0)86 (17.3)8 (6.0)82 (61.7)43 (32.3)10 (4.8)78 (37.5)120 (57.7)\n\nSociodemographic and clinical aspects\n\nSex (n (%))\n\nFemale\nMale\n430 (51.0)\n413 (49.0)\n240 (48.3)\n253 (50.9)\n86 (60.1)\n57 (39.9)\n104(50.2)\n103 (49.8)\n\nEducation (n (%))\n\nNo high school education\nHigh school\nBachelor’s degree\nMaster’s degree or higher\n447 (5.2)\n346 (41.2)\n258 (30.8)\n190 (22.6)\n29 (5.9)\n194 (39.6)\n160 (32.7)\n106 (21.6)\n10 (6.9)\n60 (41.7)\n33 (22.9)\n41 (28.5)\n5 (2.4)\n92 (44.9)\n65 (31.7)\n43 (21.0)\n\nCHD complexity (n (%))\n\nMild\nModerate\nSevere\n116 (13.8)\n473 (56.4)\n249 (29.7)\n98 (19.7)\n313 (63.0)\n86 (17.3)\n8 (6.0)\n82 (61.7)\n43 (32.3)\n10 (4.8)\n78 (37.5)\n120 (57.7)", "Participants completed a set of self-reported questionnaires, including the GES, that were administered during outpatient clinic visits (in Norway, South Korea and Belgium) and/or mailed to their home (in Belgium). Data for this sub-study were collected between August 2019 and February 2020, hence before the COVID-19 pandemic emerged.", "The psychometric evaluation of GES entailed an assessment of the content validity, factorial validity, measurement invariance, internal consistency and responsiveness. Content validity was assessed by the proportion of missing values and invalid scores [7]. While this is not a common approach for evaluating content validity, missing values can be considered an indicator of whether the items in the scale are perceived as relevant by the participants and whether the items are intelligible. Less than 5% missing data was deemed acceptable in this study.\nFactorial validity was evaluated through confirmatory factor analyses (CFA) to assess the hypothesized five-factor structure of the scale and the overall factor of patient empowerment [14]. CFA was performed for the entire sample. A good model fit was obtained if the comparative fit index (CFI) was > 0.90, standardized root mean square residual (SRMR) < 0.08 and root mean square error of approximation (RMSEA) < 0.08 [15]. These fit indices were chosen based on Kline’s suggestion for fit evaluation of CFA [14]. Standardized factor loadings are also reported.\nMeasurement invariance was examined through multigroup CFA (MGCFA) across countries [11]. The MGCFA included three models: (1) configural model (i.e., separates the sample into three subgroups, but no parameter constraints are imposed); (2) metric model (i.e., constrains the factor loadings to be equal across subgroups); and (3) scalar model (i.e., constrains the factor loadings and the item intercepts to be equal across subgroups) [16]. These models are tested sequentially and the process begins with a configural model that is well-fitting. Measurement invariance is based on how well the model fits the data as indicated by the fit indices mentioned before (i.e., CFI, RMSEA, SRMR). Additionally, change fit statistics are used to determine whether measurement invariance is present or not. This change refers to how the fit indices increase or decrease as more constraints are added. As per current recommendations, decreases below 0.01 in CFI (∆CFI) and increases below 0.015 for RMSEA (∆RMSEA) and below 0.03 for SRMR (∆SRMR) are deemed acceptable [17]. If measurement invariance was not achieved at some point, modification indices were assessed to determine whether model fit could be improved or partial invariance could be established [18]. Partial invariance occurs when some items that are different across groups are estimated freely, while keeping at least two indicators per latent construct to be equal across groups [9, 10, 19]. For the GES, this meant that only one item per dimension could be freed to attempt to establish partial invariance. If items were released, this was done following a backward method, based on the items with the highest modification indices [10]. Lastly, if no configural, metric or scalar invariance could be achieved, the factor structure was assessed separately in each group [18].\nInternal consistency was assessed by calculating the Cronbach’s alpha coefficient [20]. Coefficients were calculated for each dimension and for the overall scale. Besides Cronbach’s alpha coefficients, composite reliability values were calculated as an additional method to evaluate the inter-item consistency of the scale. The Cronbach’s alpha value as well as the composite reliability values should be above 0.70 to be considered acceptable [21]. Floor and ceiling effects were calculated as a way to assess issues regarding responsiveness. This is an indirect way to evaluate a scale’s sensitivity to detect change [22]. Floor and ceiling effects were considered present if more than 15% of the participants achieved the lowest (i.e. 15) or highest score (i.e. 75) [22].\nStatistical analyses were performed with the Lavaan package in R [23] and IBM SPSS Statistics for Windows version 27.", "APPROACH-IS II has its coordinating center at KU Leuven, Belgium. Therefore, ethical approval was granted from the Institutional Review Board of the University Hospitals Leuven/KU Leuven. Additionally, ethics approval was granted by the local ethics committees of the included centers (i.e., Norway and South Korea). All participants included in this study provided verbal and written informed consent.", "[SUBTITLE] Content validity [SUBSECTION] The proportion of missing values ranged from 0 to 0.8% (Table 2). “Knowledge and understanding” was the dimension with the highest proportion of missing values.\nThe proportion of missing values ranged from 0 to 0.8% (Table 2). “Knowledge and understanding” was the dimension with the highest proportion of missing values.\n[SUBTITLE] Factorial validity [SUBSECTION] The five-factor structure as well as the overall factor of patient empowerment of the GES were evaluated through CFA in the entire sample. The five-factor model had an acceptable model fit based on the fit indices (x2(80) = 326.296; CFI = 0.948; RMSEA = 0.060; and SRMR = 0.039). Factor loadings for this model ranged from 0.515 to 0.864 and were all significant with p < 0.001 (Table 2). Item 6 from the “personal control” dimension had the lowest factor loading (i.e., 0.515). A second-order factor model to test the overall construct of patient empowerment also showed an acceptable model fit ( x2(85) = 358.916; CFI = 0.942; RMSEA = 0.062; and SRMR = 0.044). The first-order factor loadings in this model ranged from 0.523 to 0.863 and all were significant (p < 0.001). The second-order factor loadings had the following values: 0.903 (knowledge and understanding), 0.859 (personal control), 0.638 (identity), 0.773 (shared decision-making) and 0.583 (enabling others).\n\nTable 2Proportion of missing values and factor loadings for GESItemsMissing valuesn* (%)Factor loadings\nFirst-order factor\n\nSecond-order factor\n\nKnowledge and Understanding\n1. I know and understand my medical condition7 (0.8)0.7180.7232. I know what to do to stay healthy3 (0.4)0.7750.7653. I know when to contact healthcare providers for my medical condition4 (0.5)0.6320.639\nPersonal Control\n4. I have the skills to manage my medical condition in daily life3 (0.4)0.7790.7735. I have a sense of control over my health6 (0.7)0.6820.6836. I am active in maintaining my health0 (0)0.5150.523\nIdentity\n7. My medical condition is a part of who I am as a person1 (0.1)0.6920.7018. Living with my medical condition makes me stronger as a person3 (0.4)0.6100.5959. I have given my medical condition a place in my life5 (0.6)0.6420.646\nShared decision-making\n10. I am capable of expressing to my healthcare providers what is important to me1 (0.1)0.7150.71311. I actively participate in discussions with my health care providers about my health4 (0.5)0.7200.71912. I am capable of making decisions about my health and health care with the healthcare providers2 (0.2)0.7600.762\nEnabling Others\n13. I have the skills to support other people with a similar medical condition3 (0.4)0.8640.86314. I am able to give helpful advice to people who are struggling with a similar medical condition5 (0.6)0.8530.85315. I can help other people by sharing how I keep myself well1 (0.1)0.7560.757\n\nProportion of missing values and factor loadings for GES\nThe five-factor structure as well as the overall factor of patient empowerment of the GES were evaluated through CFA in the entire sample. The five-factor model had an acceptable model fit based on the fit indices (x2(80) = 326.296; CFI = 0.948; RMSEA = 0.060; and SRMR = 0.039). Factor loadings for this model ranged from 0.515 to 0.864 and were all significant with p < 0.001 (Table 2). Item 6 from the “personal control” dimension had the lowest factor loading (i.e., 0.515). A second-order factor model to test the overall construct of patient empowerment also showed an acceptable model fit ( x2(85) = 358.916; CFI = 0.942; RMSEA = 0.062; and SRMR = 0.044). The first-order factor loadings in this model ranged from 0.523 to 0.863 and all were significant (p < 0.001). The second-order factor loadings had the following values: 0.903 (knowledge and understanding), 0.859 (personal control), 0.638 (identity), 0.773 (shared decision-making) and 0.583 (enabling others).\n\nTable 2Proportion of missing values and factor loadings for GESItemsMissing valuesn* (%)Factor loadings\nFirst-order factor\n\nSecond-order factor\n\nKnowledge and Understanding\n1. I know and understand my medical condition7 (0.8)0.7180.7232. I know what to do to stay healthy3 (0.4)0.7750.7653. I know when to contact healthcare providers for my medical condition4 (0.5)0.6320.639\nPersonal Control\n4. I have the skills to manage my medical condition in daily life3 (0.4)0.7790.7735. I have a sense of control over my health6 (0.7)0.6820.6836. I am active in maintaining my health0 (0)0.5150.523\nIdentity\n7. My medical condition is a part of who I am as a person1 (0.1)0.6920.7018. Living with my medical condition makes me stronger as a person3 (0.4)0.6100.5959. I have given my medical condition a place in my life5 (0.6)0.6420.646\nShared decision-making\n10. I am capable of expressing to my healthcare providers what is important to me1 (0.1)0.7150.71311. I actively participate in discussions with my health care providers about my health4 (0.5)0.7200.71912. I am capable of making decisions about my health and health care with the healthcare providers2 (0.2)0.7600.762\nEnabling Others\n13. I have the skills to support other people with a similar medical condition3 (0.4)0.8640.86314. I am able to give helpful advice to people who are struggling with a similar medical condition5 (0.6)0.8530.85315. I can help other people by sharing how I keep myself well1 (0.1)0.7560.757\n\nProportion of missing values and factor loadings for GES\n[SUBTITLE] Measurement invariance [SUBSECTION] The configural model had a CFI (0.899) and RMSEA (0.085) near the cut-off values for an acceptable model (Table 3). To improve model fit, modification indices were evaluated, and based on this, the residuals of items 6 and 15 were allowed to covary. Even though these items belong to different dimensions (personal control and enabling others, respectively), it is reasonable to expect that persons who are actively involved in their care, also feel more capable of sharing their experiences with others [7]. A second configural model was evaluated with this error correlation, and model fit indices reached the expected threshold (x2(252) = 695.911; CFI = 0.913; RMSEA = 0.079; and SRMR = 0.061). By achieving a well-fitting configural model, we proceeded to test metric and scalar invariance, also including this covariation.\nThe metric model fitted the data well (Table 3). Additionally, changes in model fit indices (∆CFI 0.005; ∆RMSEA 0.002; ∆SRMR 0.006) were within the expected values, indicating metric invariance was supported. An evaluation of scalar invariance came along with slightly worse model fit indices (Table 3). In comparison to the metric model, fit indices were not within the allowed change range (∆CFI 0.034; ∆RMSEA 0.010; ∆SRMR 0.007). Therefore, full scalar invariance could not be established.\nGiven the lack of full scalar invariance, we proceeded to test partial scalar invariance by evaluating modification indices and the equality constraints across groups. Constraints were relaxed sequentially until an acceptable fit was achieved. Fixing the intercept of item 9 (identity) contributed the most to the observed misfit across groups, so this item was estimated freely in a partial scalar invariance model. While model fit improvements were obtained, the indices were below the acceptable threshold. Therefore, a new partial scalar invariance model was tested with the intercepts of items 9 (identity) and 4 (personal control) set free. While improvements were identified (Table 3), model fit indices were still not within an acceptable range. We continued to release items sequentially to achieve acceptable model fit. Models were tested with intercepts of items 9 (identity), 4 (personal control), 12 (shared decision-making) and 2 (knowledge and understanding) unconstrained and while fit indices were almost within the recommended range, it was not possible to achieve acceptable model fit. While models with unconstrained items from the “enabling others” dimension were assessed, none of them led to changes in the model fit indices. Additionally, given that not more than one item per dimension could be estimated freely, models with more than 5 free items where not tested. Therefore, partial scalar invariance was rejected.\nSince partial scalar invariance was not achieved, an assessment of the three groups independently through CFA was undertaken to understand why this was the case. While fit indices for Norway (x2(85) = 117.694; CFI = 0.964; RMSEA = 0.053; and SRMR = 0.074) and Belgium ( x2(85) = 348.280; CFI = 0.916; RMSEA = 0.079; and SRMR = 0.053) indicated the model fitted these groups well, it appears this was not the case for the data from South Korea (x2(85): 307.753; CFI: 0.788; RMSEA: 0.112; and SRMR: 0.079). Factor loadings for this country ranged between 0.407 and 0.772, with low factor loadings in the “shared decision-making” dimension, though significant (p < 0.001).\n[SUBTITLE] Measurement invariance: norwegian and belgian samples [SUBSECTION] As the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium.\n\nTable 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR\nComplete sample (Belgium, Norway and South Korea)\n\nConfigural invariance\na\n772.074 (255)0.8990.0850.063\nConfigural invariance\nb\n695.911 (252)0.9130.0790.061\nMetric invariance\nb\n750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006\nScalar invariance\nb\n941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007\nPartial scalar invariance with intercept of item 9 free\nb\n886.943 (296)0.8850.0840.072\nPartial scalar invariance with intercept of items 9 and 4 free\nb\n856.568 (294)0.8900.0820.070\nPartial scalar invariance with intercept of items 9, 4 and 12 free\nb\n827.343 (292)0.8960.0800.069\nPartial scalar invariance with intercept of items 9, 4, 12 and 2 free\nb\n813.205 (290)0.8980.0800.069\nPartial sample (Belgium and Norway)\n\nConfigural invariance\na\n463.510 (170)0.9280.0740.057\nMetric invariance\na\n482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004\nScalar invariance\na\n544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002\nPartial scalar invariance with one intercept free\nc\n519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\n\nModel fit indexes of multi-group confirmatory factor analyses\na Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\nAs the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium.\n\nTable 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR\nComplete sample (Belgium, Norway and South Korea)\n\nConfigural invariance\na\n772.074 (255)0.8990.0850.063\nConfigural invariance\nb\n695.911 (252)0.9130.0790.061\nMetric invariance\nb\n750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006\nScalar invariance\nb\n941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007\nPartial scalar invariance with intercept of item 9 free\nb\n886.943 (296)0.8850.0840.072\nPartial scalar invariance with intercept of items 9 and 4 free\nb\n856.568 (294)0.8900.0820.070\nPartial scalar invariance with intercept of items 9, 4 and 12 free\nb\n827.343 (292)0.8960.0800.069\nPartial scalar invariance with intercept of items 9, 4, 12 and 2 free\nb\n813.205 (290)0.8980.0800.069\nPartial sample (Belgium and Norway)\n\nConfigural invariance\na\n463.510 (170)0.9280.0740.057\nMetric invariance\na\n482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004\nScalar invariance\na\n544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002\nPartial scalar invariance with one intercept free\nc\n519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\n\nModel fit indexes of multi-group confirmatory factor analyses\na Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\nThe configural model had a CFI (0.899) and RMSEA (0.085) near the cut-off values for an acceptable model (Table 3). To improve model fit, modification indices were evaluated, and based on this, the residuals of items 6 and 15 were allowed to covary. Even though these items belong to different dimensions (personal control and enabling others, respectively), it is reasonable to expect that persons who are actively involved in their care, also feel more capable of sharing their experiences with others [7]. A second configural model was evaluated with this error correlation, and model fit indices reached the expected threshold (x2(252) = 695.911; CFI = 0.913; RMSEA = 0.079; and SRMR = 0.061). By achieving a well-fitting configural model, we proceeded to test metric and scalar invariance, also including this covariation.\nThe metric model fitted the data well (Table 3). Additionally, changes in model fit indices (∆CFI 0.005; ∆RMSEA 0.002; ∆SRMR 0.006) were within the expected values, indicating metric invariance was supported. An evaluation of scalar invariance came along with slightly worse model fit indices (Table 3). In comparison to the metric model, fit indices were not within the allowed change range (∆CFI 0.034; ∆RMSEA 0.010; ∆SRMR 0.007). Therefore, full scalar invariance could not be established.\nGiven the lack of full scalar invariance, we proceeded to test partial scalar invariance by evaluating modification indices and the equality constraints across groups. Constraints were relaxed sequentially until an acceptable fit was achieved. Fixing the intercept of item 9 (identity) contributed the most to the observed misfit across groups, so this item was estimated freely in a partial scalar invariance model. While model fit improvements were obtained, the indices were below the acceptable threshold. Therefore, a new partial scalar invariance model was tested with the intercepts of items 9 (identity) and 4 (personal control) set free. While improvements were identified (Table 3), model fit indices were still not within an acceptable range. We continued to release items sequentially to achieve acceptable model fit. Models were tested with intercepts of items 9 (identity), 4 (personal control), 12 (shared decision-making) and 2 (knowledge and understanding) unconstrained and while fit indices were almost within the recommended range, it was not possible to achieve acceptable model fit. While models with unconstrained items from the “enabling others” dimension were assessed, none of them led to changes in the model fit indices. Additionally, given that not more than one item per dimension could be estimated freely, models with more than 5 free items where not tested. Therefore, partial scalar invariance was rejected.\nSince partial scalar invariance was not achieved, an assessment of the three groups independently through CFA was undertaken to understand why this was the case. While fit indices for Norway (x2(85) = 117.694; CFI = 0.964; RMSEA = 0.053; and SRMR = 0.074) and Belgium ( x2(85) = 348.280; CFI = 0.916; RMSEA = 0.079; and SRMR = 0.053) indicated the model fitted these groups well, it appears this was not the case for the data from South Korea (x2(85): 307.753; CFI: 0.788; RMSEA: 0.112; and SRMR: 0.079). Factor loadings for this country ranged between 0.407 and 0.772, with low factor loadings in the “shared decision-making” dimension, though significant (p < 0.001).\n[SUBTITLE] Measurement invariance: norwegian and belgian samples [SUBSECTION] As the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium.\n\nTable 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR\nComplete sample (Belgium, Norway and South Korea)\n\nConfigural invariance\na\n772.074 (255)0.8990.0850.063\nConfigural invariance\nb\n695.911 (252)0.9130.0790.061\nMetric invariance\nb\n750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006\nScalar invariance\nb\n941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007\nPartial scalar invariance with intercept of item 9 free\nb\n886.943 (296)0.8850.0840.072\nPartial scalar invariance with intercept of items 9 and 4 free\nb\n856.568 (294)0.8900.0820.070\nPartial scalar invariance with intercept of items 9, 4 and 12 free\nb\n827.343 (292)0.8960.0800.069\nPartial scalar invariance with intercept of items 9, 4, 12 and 2 free\nb\n813.205 (290)0.8980.0800.069\nPartial sample (Belgium and Norway)\n\nConfigural invariance\na\n463.510 (170)0.9280.0740.057\nMetric invariance\na\n482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004\nScalar invariance\na\n544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002\nPartial scalar invariance with one intercept free\nc\n519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\n\nModel fit indexes of multi-group confirmatory factor analyses\na Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\nAs the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium.\n\nTable 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR\nComplete sample (Belgium, Norway and South Korea)\n\nConfigural invariance\na\n772.074 (255)0.8990.0850.063\nConfigural invariance\nb\n695.911 (252)0.9130.0790.061\nMetric invariance\nb\n750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006\nScalar invariance\nb\n941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007\nPartial scalar invariance with intercept of item 9 free\nb\n886.943 (296)0.8850.0840.072\nPartial scalar invariance with intercept of items 9 and 4 free\nb\n856.568 (294)0.8900.0820.070\nPartial scalar invariance with intercept of items 9, 4 and 12 free\nb\n827.343 (292)0.8960.0800.069\nPartial scalar invariance with intercept of items 9, 4, 12 and 2 free\nb\n813.205 (290)0.8980.0800.069\nPartial sample (Belgium and Norway)\n\nConfigural invariance\na\n463.510 (170)0.9280.0740.057\nMetric invariance\na\n482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004\nScalar invariance\na\n544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002\nPartial scalar invariance with one intercept free\nc\n519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\n\nModel fit indexes of multi-group confirmatory factor analyses\na Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\n[SUBTITLE] Internal consistency [SUBSECTION] The Cronbach’s alpha for the overall scale was 0.873, indicating that it was internally consistent. The alpha values for the subscales were: 0.73 (knowledge and understanding); 0.693 (personal control); 0.680 (identity); 0. 774 (shared decision-making); and 0.863 (enabling others). The Cronbach’s alpha for each country is given in Table 4. An evaluation of the scale’s composite reliability showed that most of the values are above the expected range of 0.7 and those below such threshold were relatively near to this acceptable value.\nThe Cronbach’s alpha for the overall scale was 0.873, indicating that it was internally consistent. The alpha values for the subscales were: 0.73 (knowledge and understanding); 0.693 (personal control); 0.680 (identity); 0. 774 (shared decision-making); and 0.863 (enabling others). The Cronbach’s alpha for each country is given in Table 4. An evaluation of the scale’s composite reliability showed that most of the values are above the expected range of 0.7 and those below such threshold were relatively near to this acceptable value.\n[SUBTITLE] Responsiveness [SUBSECTION] The mean patient empowerment score for the entire sample was 59.36 ± 8.47. None of the participants had the lowest score (i.e., 15) and only 2.5% had the highest attainable score of 75. Hence, no floor or ceiling effects were identified.\n\nTable 4Cronbach’s alpha values, composite reliability and mean empowerment scoresTotal SampleBelgiumNorwaySouth Korea\nCronbach’s alpha values\n\nKnowledge and understanding\n0.7300.7210.7590.728\nPersonal control\n0.6930.6330.6360.751\nIdentity\n0.6800.6820.7510.636\nShared decision-making\n0.7740.8330.8040.491\nEnabling others\n0.8630.8720.9050.782\nOverall scale\n0.8730.8780.8710.854\nComposite reliability\n\nKnowledge and understanding\n0.7530.7520.7790.732\nPersonal control\n0.7020.6500.6770.753\nIdentity\n0.6840.6820.7700.657\nShared decision-making\n0.5590.8350.8050.470\nEnabling others\n0.8640.8760.9100.782\nOverall scale\n0.9290.9420.9510.917\nMean patient empowerment scores\n\nKnowledge and understanding\n12.94 (1.90)13.07 (1.94)12.93 (2.03)12.61 (1.68)\nPersonal control\n12.04 (2.10)12.36 (1.97)12.59 (1.85)10.90 (2.12)\nIdentity\n11.66 (2.39)11.82 (2.42)11.19 (2.79)11.58 (1.97)\nShared decision-making\n12.15 (2.26)12.55 (2.22)11.58 (2.75)11.61 (1.74)\nEnabling others\n10.57 (2.83)10.72 (2.90)10.51 (3.12)10.25 (2.40)\nOverall scale\n59.37 (8.47)60.52 (8.53)58.86 (9.07)56.98 (7.33)\n\nCronbach’s alpha values, composite reliability and mean empowerment scores\nThe mean patient empowerment score for the entire sample was 59.36 ± 8.47. None of the participants had the lowest score (i.e., 15) and only 2.5% had the highest attainable score of 75. Hence, no floor or ceiling effects were identified.\n\nTable 4Cronbach’s alpha values, composite reliability and mean empowerment scoresTotal SampleBelgiumNorwaySouth Korea\nCronbach’s alpha values\n\nKnowledge and understanding\n0.7300.7210.7590.728\nPersonal control\n0.6930.6330.6360.751\nIdentity\n0.6800.6820.7510.636\nShared decision-making\n0.7740.8330.8040.491\nEnabling others\n0.8630.8720.9050.782\nOverall scale\n0.8730.8780.8710.854\nComposite reliability\n\nKnowledge and understanding\n0.7530.7520.7790.732\nPersonal control\n0.7020.6500.6770.753\nIdentity\n0.6840.6820.7700.657\nShared decision-making\n0.5590.8350.8050.470\nEnabling others\n0.8640.8760.9100.782\nOverall scale\n0.9290.9420.9510.917\nMean patient empowerment scores\n\nKnowledge and understanding\n12.94 (1.90)13.07 (1.94)12.93 (2.03)12.61 (1.68)\nPersonal control\n12.04 (2.10)12.36 (1.97)12.59 (1.85)10.90 (2.12)\nIdentity\n11.66 (2.39)11.82 (2.42)11.19 (2.79)11.58 (1.97)\nShared decision-making\n12.15 (2.26)12.55 (2.22)11.58 (2.75)11.61 (1.74)\nEnabling others\n10.57 (2.83)10.72 (2.90)10.51 (3.12)10.25 (2.40)\nOverall scale\n59.37 (8.47)60.52 (8.53)58.86 (9.07)56.98 (7.33)\n\nCronbach’s alpha values, composite reliability and mean empowerment scores", "The proportion of missing values ranged from 0 to 0.8% (Table 2). “Knowledge and understanding” was the dimension with the highest proportion of missing values.", "The five-factor structure as well as the overall factor of patient empowerment of the GES were evaluated through CFA in the entire sample. The five-factor model had an acceptable model fit based on the fit indices (x2(80) = 326.296; CFI = 0.948; RMSEA = 0.060; and SRMR = 0.039). Factor loadings for this model ranged from 0.515 to 0.864 and were all significant with p < 0.001 (Table 2). Item 6 from the “personal control” dimension had the lowest factor loading (i.e., 0.515). A second-order factor model to test the overall construct of patient empowerment also showed an acceptable model fit ( x2(85) = 358.916; CFI = 0.942; RMSEA = 0.062; and SRMR = 0.044). The first-order factor loadings in this model ranged from 0.523 to 0.863 and all were significant (p < 0.001). The second-order factor loadings had the following values: 0.903 (knowledge and understanding), 0.859 (personal control), 0.638 (identity), 0.773 (shared decision-making) and 0.583 (enabling others).\n\nTable 2Proportion of missing values and factor loadings for GESItemsMissing valuesn* (%)Factor loadings\nFirst-order factor\n\nSecond-order factor\n\nKnowledge and Understanding\n1. I know and understand my medical condition7 (0.8)0.7180.7232. I know what to do to stay healthy3 (0.4)0.7750.7653. I know when to contact healthcare providers for my medical condition4 (0.5)0.6320.639\nPersonal Control\n4. I have the skills to manage my medical condition in daily life3 (0.4)0.7790.7735. I have a sense of control over my health6 (0.7)0.6820.6836. I am active in maintaining my health0 (0)0.5150.523\nIdentity\n7. My medical condition is a part of who I am as a person1 (0.1)0.6920.7018. Living with my medical condition makes me stronger as a person3 (0.4)0.6100.5959. I have given my medical condition a place in my life5 (0.6)0.6420.646\nShared decision-making\n10. I am capable of expressing to my healthcare providers what is important to me1 (0.1)0.7150.71311. I actively participate in discussions with my health care providers about my health4 (0.5)0.7200.71912. I am capable of making decisions about my health and health care with the healthcare providers2 (0.2)0.7600.762\nEnabling Others\n13. I have the skills to support other people with a similar medical condition3 (0.4)0.8640.86314. I am able to give helpful advice to people who are struggling with a similar medical condition5 (0.6)0.8530.85315. I can help other people by sharing how I keep myself well1 (0.1)0.7560.757\n\nProportion of missing values and factor loadings for GES", "The configural model had a CFI (0.899) and RMSEA (0.085) near the cut-off values for an acceptable model (Table 3). To improve model fit, modification indices were evaluated, and based on this, the residuals of items 6 and 15 were allowed to covary. Even though these items belong to different dimensions (personal control and enabling others, respectively), it is reasonable to expect that persons who are actively involved in their care, also feel more capable of sharing their experiences with others [7]. A second configural model was evaluated with this error correlation, and model fit indices reached the expected threshold (x2(252) = 695.911; CFI = 0.913; RMSEA = 0.079; and SRMR = 0.061). By achieving a well-fitting configural model, we proceeded to test metric and scalar invariance, also including this covariation.\nThe metric model fitted the data well (Table 3). Additionally, changes in model fit indices (∆CFI 0.005; ∆RMSEA 0.002; ∆SRMR 0.006) were within the expected values, indicating metric invariance was supported. An evaluation of scalar invariance came along with slightly worse model fit indices (Table 3). In comparison to the metric model, fit indices were not within the allowed change range (∆CFI 0.034; ∆RMSEA 0.010; ∆SRMR 0.007). Therefore, full scalar invariance could not be established.\nGiven the lack of full scalar invariance, we proceeded to test partial scalar invariance by evaluating modification indices and the equality constraints across groups. Constraints were relaxed sequentially until an acceptable fit was achieved. Fixing the intercept of item 9 (identity) contributed the most to the observed misfit across groups, so this item was estimated freely in a partial scalar invariance model. While model fit improvements were obtained, the indices were below the acceptable threshold. Therefore, a new partial scalar invariance model was tested with the intercepts of items 9 (identity) and 4 (personal control) set free. While improvements were identified (Table 3), model fit indices were still not within an acceptable range. We continued to release items sequentially to achieve acceptable model fit. Models were tested with intercepts of items 9 (identity), 4 (personal control), 12 (shared decision-making) and 2 (knowledge and understanding) unconstrained and while fit indices were almost within the recommended range, it was not possible to achieve acceptable model fit. While models with unconstrained items from the “enabling others” dimension were assessed, none of them led to changes in the model fit indices. Additionally, given that not more than one item per dimension could be estimated freely, models with more than 5 free items where not tested. Therefore, partial scalar invariance was rejected.\nSince partial scalar invariance was not achieved, an assessment of the three groups independently through CFA was undertaken to understand why this was the case. While fit indices for Norway (x2(85) = 117.694; CFI = 0.964; RMSEA = 0.053; and SRMR = 0.074) and Belgium ( x2(85) = 348.280; CFI = 0.916; RMSEA = 0.079; and SRMR = 0.053) indicated the model fitted these groups well, it appears this was not the case for the data from South Korea (x2(85): 307.753; CFI: 0.788; RMSEA: 0.112; and SRMR: 0.079). Factor loadings for this country ranged between 0.407 and 0.772, with low factor loadings in the “shared decision-making” dimension, though significant (p < 0.001).\n[SUBTITLE] Measurement invariance: norwegian and belgian samples [SUBSECTION] As the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium.\n\nTable 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR\nComplete sample (Belgium, Norway and South Korea)\n\nConfigural invariance\na\n772.074 (255)0.8990.0850.063\nConfigural invariance\nb\n695.911 (252)0.9130.0790.061\nMetric invariance\nb\n750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006\nScalar invariance\nb\n941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007\nPartial scalar invariance with intercept of item 9 free\nb\n886.943 (296)0.8850.0840.072\nPartial scalar invariance with intercept of items 9 and 4 free\nb\n856.568 (294)0.8900.0820.070\nPartial scalar invariance with intercept of items 9, 4 and 12 free\nb\n827.343 (292)0.8960.0800.069\nPartial scalar invariance with intercept of items 9, 4, 12 and 2 free\nb\n813.205 (290)0.8980.0800.069\nPartial sample (Belgium and Norway)\n\nConfigural invariance\na\n463.510 (170)0.9280.0740.057\nMetric invariance\na\n482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004\nScalar invariance\na\n544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002\nPartial scalar invariance with one intercept free\nc\n519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\n\nModel fit indexes of multi-group confirmatory factor analyses\na Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\nAs the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium.\n\nTable 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR\nComplete sample (Belgium, Norway and South Korea)\n\nConfigural invariance\na\n772.074 (255)0.8990.0850.063\nConfigural invariance\nb\n695.911 (252)0.9130.0790.061\nMetric invariance\nb\n750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006\nScalar invariance\nb\n941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007\nPartial scalar invariance with intercept of item 9 free\nb\n886.943 (296)0.8850.0840.072\nPartial scalar invariance with intercept of items 9 and 4 free\nb\n856.568 (294)0.8900.0820.070\nPartial scalar invariance with intercept of items 9, 4 and 12 free\nb\n827.343 (292)0.8960.0800.069\nPartial scalar invariance with intercept of items 9, 4, 12 and 2 free\nb\n813.205 (290)0.8980.0800.069\nPartial sample (Belgium and Norway)\n\nConfigural invariance\na\n463.510 (170)0.9280.0740.057\nMetric invariance\na\n482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004\nScalar invariance\na\n544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002\nPartial scalar invariance with one intercept free\nc\n519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\n\nModel fit indexes of multi-group confirmatory factor analyses\na Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030", "As the CFA models for Norway and Belgium had an acceptable model fit. An evaluation of measurement invariance within these two countries was undertaken. Configural invariance was supported by a well-fitted model (x2(170) = 463.510; CFI = 0.928; RMSEA = 0.074; and SRMR = 0.057). An evaluation of metric invariance also showed acceptable model fit indices (Table 3) and changes in them were also within the acceptable ranges (∆CFI 0.001; ∆RMSEA 0.002; ∆SRMR 0.004). Given that metric invariance was supported, scalar invariance was evaluated next. Fit indexes for this model were within the recommended values (Table 3). However, changes in the CFI were outside of the acceptable range to support measurement invariance (∆CFI 0.013; ∆RMSEA 0.004; ∆SRMR 0.002). Therefore, full scalar invariance was not supported. Modification indices indicated that freeing the intercept of item 11 (shared decision-making) could lead to model improvements. Hence, partial scalar invariance was evaluated with the intercept of item 11 free. This model had acceptable model fit indexes (x2(192) = 519.18; CFI = 0.920; RMSEA = 0.073; and SRMR = 0.063). Additionally, in comparison to the metric model, fit indices suggested worse model fit, but changes were still within acceptable ranges (∆CFI 0.007; ∆RMSEA 0.002; ∆SRMR 0.001). These results support partial scalar invariance for the samples of Norway and Belgium.\n\nTable 3Model fit indexes of multi-group confirmatory factor analysesModelX2 (df)CFIRMSEASRMR∆X2 (∆df)∆CFI∆RMSEA∆SRMR\nComplete sample (Belgium, Norway and South Korea)\n\nConfigural invariance\na\n772.074 (255)0.8990.0850.063\nConfigural invariance\nb\n695.911 (252)0.9130.0790.061\nMetric invariance\nb\n750.492 (280)0.9080.0770.06754.581 (28)0.0050.0020.006\nScalar invariance\nb\n941.720 (298)0.8740.0870.074191.228 (18)0.0340.0100.007\nPartial scalar invariance with intercept of item 9 free\nb\n886.943 (296)0.8850.0840.072\nPartial scalar invariance with intercept of items 9 and 4 free\nb\n856.568 (294)0.8900.0820.070\nPartial scalar invariance with intercept of items 9, 4 and 12 free\nb\n827.343 (292)0.8960.0800.069\nPartial scalar invariance with intercept of items 9, 4, 12 and 2 free\nb\n813.205 (290)0.8980.0800.069\nPartial sample (Belgium and Norway)\n\nConfigural invariance\na\n463.510 (170)0.9280.0740.057\nMetric invariance\na\n482.543 (184)0.9270.0710.06119.032 (14)0.0010.0020.004\nScalar invariance\na\n544.309 (193)0.9140.0750.06461.767 (9)0.0130.0040.002\nPartial scalar invariance with one intercept free\nc\n519.177 (192)0.9200.0730.06336.635 (8)0.0070.0020.001a Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030\n\nModel fit indexes of multi-group confirmatory factor analyses\na Model without error correlation; b Model with error correlation between items 6 and 15. Models were considered to have acceptable fit if CFI > 0.90, RMSEA and SRMR values < 0.08. MGCFA needed also to have it indexes changes within these ranges: ΔCFI < 0.010, ΔRMSEA < 0.015, and ΔSRMR < 0.030", "The Cronbach’s alpha for the overall scale was 0.873, indicating that it was internally consistent. The alpha values for the subscales were: 0.73 (knowledge and understanding); 0.693 (personal control); 0.680 (identity); 0. 774 (shared decision-making); and 0.863 (enabling others). The Cronbach’s alpha for each country is given in Table 4. An evaluation of the scale’s composite reliability showed that most of the values are above the expected range of 0.7 and those below such threshold were relatively near to this acceptable value.", "The mean patient empowerment score for the entire sample was 59.36 ± 8.47. None of the participants had the lowest score (i.e., 15) and only 2.5% had the highest attainable score of 75. Hence, no floor or ceiling effects were identified.\n\nTable 4Cronbach’s alpha values, composite reliability and mean empowerment scoresTotal SampleBelgiumNorwaySouth Korea\nCronbach’s alpha values\n\nKnowledge and understanding\n0.7300.7210.7590.728\nPersonal control\n0.6930.6330.6360.751\nIdentity\n0.6800.6820.7510.636\nShared decision-making\n0.7740.8330.8040.491\nEnabling others\n0.8630.8720.9050.782\nOverall scale\n0.8730.8780.8710.854\nComposite reliability\n\nKnowledge and understanding\n0.7530.7520.7790.732\nPersonal control\n0.7020.6500.6770.753\nIdentity\n0.6840.6820.7700.657\nShared decision-making\n0.5590.8350.8050.470\nEnabling others\n0.8640.8760.9100.782\nOverall scale\n0.9290.9420.9510.917\nMean patient empowerment scores\n\nKnowledge and understanding\n12.94 (1.90)13.07 (1.94)12.93 (2.03)12.61 (1.68)\nPersonal control\n12.04 (2.10)12.36 (1.97)12.59 (1.85)10.90 (2.12)\nIdentity\n11.66 (2.39)11.82 (2.42)11.19 (2.79)11.58 (1.97)\nShared decision-making\n12.15 (2.26)12.55 (2.22)11.58 (2.75)11.61 (1.74)\nEnabling others\n10.57 (2.83)10.72 (2.90)10.51 (3.12)10.25 (2.40)\nOverall scale\n59.37 (8.47)60.52 (8.53)58.86 (9.07)56.98 (7.33)\n\nCronbach’s alpha values, composite reliability and mean empowerment scores", "Recent research highlights the need to improve and evaluate patient empowerment in persons with CCs [4]. However, the availability of instruments with a strong theoretical background and/or acceptable psychometric properties is limited [6]. Therefore, the present study evaluated a slightly modified version of a previously validated patient empowerment in a group of adults with CHD. Results indicate that GES proved reliable, meaning the scale’s items altogether are consistent. Additionally, no floor or ceiling effects were identified, which is indirectly related to the scale’s sensitivity to measure change [22]. Whereas these results are in favor of the GES, a psychometric assessment of the scale’s validity revealed that the scale appears valid in two of the three included countries.\nResults from the CFA of the entire sample indicate the five-factor structure fits the data well. However, due to the lack of measurement invariance, CFAs were performed separately for patients from each country. Analyses revealed that an acceptable model fit was only achieved for the samples from Belgium and Norway. For the South Korean data, the CFI and the RMSEA were above the acceptable ranges although factor loadings from the “shared decision-making” dimension were low. This is an indication that the relations among the latent constructs of GES are not in line with the data, i.e., the data do not support the model [24]. Even though it is common in CFA to conduct an evaluation of modification indexes to improve model fit, changes should be theoretically justified [25]. In this case, we could identify no strong theoretical reasons to allow for model changes that could potentially improve the model fit.\nThe poorly fitted model to the South Korean data might be the reason why invariance was not achieved when comparing the three countries. A lack of invariance suggests that patient empowerment may have a different meaning in different countries. Therefore, making mean comparisons of this construct across countries using the GES should be made with caution. Nonetheless, while cross-country comparisons cannot be made, it is possible to compare how the GES relates to other constructs (i.e., variables) within countries. For example, one could undertake a study to compare how the GES relates to patient functioning in Belgium, vs. how the GES relates to patient functioning in South Korea. Another potential research aim would be to determine whether changes in GES relate to psychological distress and whether these changes have the same magnitude in Belgium, Norway and South Korea.\nThe absence of invariance across the three countries can be an indicator of patient empowerment being interpreted differently across countries. Perhaps individuals in South Korea have different interpretations and values of patient empowerment compared to patients in Belgium and Norway, two European countries that are likely more culturally similar. It has been suggested that patient empowerment is determined by its context and that individuals within a particular environment, organization or country may have a different perception of empowerment [26]. Hence, a measure that fits all persons (or contexts), might be hard to develop. Findings from the present study partially support this notion. It is plausible to consider that different countries (and cultures) have a different understanding of patient empowerment domains, values, and skills.\nEvidence on how patient empowerment may differ between countries is limited. However, authors of a systematic review concluded that although patients do share many perspectives of this construct, there might be variations on how certain aspects of patient empowerment are understood and research has yet to address structural elements of patient empowerment [27].\nConsideration of structural elements is relevant for the current study because the “shared decision-making” dimension had the lowest factor loadings for the South Korean sample, as well as poor reliability. Shared decision-making involves healthcare professionals and patients working together on a care plan [28]. It entails patients who are willing to participate as well as clinicians who want to collaborate with the patient. Hence, it is greatly influenced by the healthcare structure. Within Asian cultures, for example, it is more commonly believed that patients prefer to take a less independent role and that a family-centered (rather than individual-centered) approach towards decision-making is preferred [29–31]. It has been suggested that shared decision-making reflects values associated with western cultures and that this might be different in other cultures [28, 31, 32]. The value placed on shared decision-making and the structure of the healthcare system might therefore potentially impact the value of patient empowerment as perceived by study participants. Therefore, the comparison of this construct between different cultures and regions of the world should be made with caution.\nAlthough the GES appears to be a valid and reliable instrument for the Norwegian and Belgian sample, it is worth noting that future research should evaluate whether the low psychometric properties found in the South Korean sample are indeed applicable to other Asian or non-European countries. Comparatives studies evaluating this construct (and similar constructs) between Western and Asian countries are needed to comprehend this phenomenon, the types of research questions that can be addressed and the conclusions that can be drawn when evaluating outcomes such as patient empowerment. Such studies could be undertaken once the data collection from APPROACH-IS II is finalized.\n[SUBTITLE] Methodological considerations [SUBSECTION] There are several strengths of the study that merit mentioning. First, GES development was based on a previously validated questionnaire, which in turn has a strong theoretical foundation and followed a rigorous development process. Therefore, it is plausible to conclude that GES also has a strong theoretical foundation. Second, data were collected as part of an international study, which allows for cross-country comparisons. Third, the data were collected before the COVID-19 pandemic, therefore there was no concern about the potential for survey responses to be impacted by the pandemic [33].\nThere are, however, also some methodological limitations associated with this study. First, some aspects associated with validity and reliability were not assessed. For instance, it was not possible to directly evaluate responsiveness with cross-sectional data. While floor and ceiling effects are an indirect way to measure responsiveness, this is better achieved through longitudinal studies. Future longitudinal studies should evaluate this psychometric property. Second, the study only includes individuals with CHD, who were in current follow-up and who had the abilities to answer the questionnaires. Therefore, the generalizability of the results might be limited.\nThere are several strengths of the study that merit mentioning. First, GES development was based on a previously validated questionnaire, which in turn has a strong theoretical foundation and followed a rigorous development process. Therefore, it is plausible to conclude that GES also has a strong theoretical foundation. Second, data were collected as part of an international study, which allows for cross-country comparisons. Third, the data were collected before the COVID-19 pandemic, therefore there was no concern about the potential for survey responses to be impacted by the pandemic [33].\nThere are, however, also some methodological limitations associated with this study. First, some aspects associated with validity and reliability were not assessed. For instance, it was not possible to directly evaluate responsiveness with cross-sectional data. While floor and ceiling effects are an indirect way to measure responsiveness, this is better achieved through longitudinal studies. Future longitudinal studies should evaluate this psychometric property. Second, the study only includes individuals with CHD, who were in current follow-up and who had the abilities to answer the questionnaires. Therefore, the generalizability of the results might be limited.", "There are several strengths of the study that merit mentioning. First, GES development was based on a previously validated questionnaire, which in turn has a strong theoretical foundation and followed a rigorous development process. Therefore, it is plausible to conclude that GES also has a strong theoretical foundation. Second, data were collected as part of an international study, which allows for cross-country comparisons. Third, the data were collected before the COVID-19 pandemic, therefore there was no concern about the potential for survey responses to be impacted by the pandemic [33].\nThere are, however, also some methodological limitations associated with this study. First, some aspects associated with validity and reliability were not assessed. For instance, it was not possible to directly evaluate responsiveness with cross-sectional data. While floor and ceiling effects are an indirect way to measure responsiveness, this is better achieved through longitudinal studies. Future longitudinal studies should evaluate this psychometric property. Second, the study only includes individuals with CHD, who were in current follow-up and who had the abilities to answer the questionnaires. Therefore, the generalizability of the results might be limited.", "This study provides evidence on the GES’s validity, reliability, and responsiveness in adults with CHD. GES seems to be valid and reliable for the sample of Belgium and Norway. However, this is not the case for South Korea, because shared decision-making seems to have a different meaning in Asian cultures. Hence, cross-cultural comparisons using GES should be made cautiously. It is possible that in countries who are more culturally different from Norway or Belgium, the scale might not perform as well." ]
[ null, null, null, null, null, null, null, null, "results", null, null, null, null, null, null, "discussion", null, "conclusion" ]
[ "Adults", "Congenital heart defects", "Chronic conditions", "Measurement invariance", "Patient empowerment", "Psychometrics", "Validity", "Reliability" ]
Detection of C8/T1 radiculopathy by measuring the root motor conduction time.
36266617
Root motor conduction time (RMCT) can noninvasively evaluate the status of the proximal root segment. However, its clinical application remains limited, and wider studies regarding its use are scarce. We aimed to investigate the association between C8/T1 level radiculopathy and RMCT.
BACKGROUND
This was a retrospective cross-sectional study. Subjects were extracted from a general hospital's spine clinic database. A total of 48 C8/T1 root lesions from 37 patients were included, and 48 C8/T1 root levels from control subjects were matched for age, sex, and height. RMCT was measured in the abductor pollicis brevis muscle and the assessment of any delays owing to C8/T1 radiculopathy.
METHODS
The RMCT of the C8/T1 radiculopathy group was 1.7 ± 0.6 ms, which was significantly longer than that in the control group (1.2 ± 0.8 ms; p = 0.001). The delayed RMCT was independently associated with radiculopathy (adjusted odds ratio, 1.15; 95% confidence interval, 1.06-1.27; p = 0.011) after adjusting for the peripheral motor conduction time, amplitude of median compound motor nerve action potential, and shortest F-wave latency. The area under the Receiver Operating Characteristic curve for diagnosing C8/T1 radiculopathy using RMCT was 0.72 (0.61-0.82). The RMCT was significantly correlated with symptom duration (coefficient = 0.58; p < 0.001) but was not associated with the degree of arm pain.
RESULTS
Our findings illustrate the clinical applicability of the RMCT by demonstrating its utility in diagnosing radiculopathy at certain spinal levels.
CONCLUSION
[ "Humans", "Radiculopathy", "Retrospective Studies", "Cross-Sectional Studies", "Muscle, Skeletal", "Evoked Potentials", "Neural Conduction" ]
9583482
Introduction
Radiculopathy is a common condition whose symptoms can include pain, sensory change, and motor weakness owing to mechanical and chemical irritation of the spinal nerve root [1, 2]. Imaging studies are essential for diagnosing radiculopathy; magnetic resonance imaging helps identify soft tissues such as discs, ligaments, and nerve roots [3], while computed tomography is primarily used to confirm abnormalities in bony structures [4]. Meanwhile, another key factor in confirming radiculopathy is electrophysiologic diagnosis [5]. In particular, electromyography (EMG) is a classical tool that was used to evaluate radiculopathy before the development of modern imaging instruments [6]; its key advantage is in its ability to detect neurophysiological changes that cannot be confirmed by imaging studies alone [7]. By applying EMG to multiple myotomes, it is possible not only to localize the abnormal spinal root level but also to estimate the temporal aspect of compression by distinguishing motor unit action potential [8]. However, the invasive nature of EMG creates a potential risk of complications such as bleeding or infection at the examination site [9]. Additionally, patient cooperation is essential for observing resting potential and interference patterns [10]. Root motor conduction time (RMCT) is a measure of the state of the proximal root segment that is obtained noninvasively by performing direct cervical stimulation and nerve conduction studies [11, 12]. Notably, RMCT is very localized since it excludes distal neural lesions and measures only the conduction time of the proximal root segment [13–15]. However, it also has the disadvantage of involving a somewhat complicated measurement method wherein well-trained examiners are required [16]. To date, the RMCT has been investigated in only a few disease types, such as demyelinating disease and lumbar spinal stenosis, whereas its clinical applicability in patients with radiculopathy remains unclear [17]. In this study, we hypothesized that the RMCT can be a valuable tool for the diagnosis of radiculopathy at certain spinal root levels. To that end, we investigated whether the RMCT measured in the abductor pollicis brevis (APB) muscle was significantly delayed in the presence of C8/T1 radiculopathy. Moreover, we aimed to determine the association between clinical symptoms and RMCT.
null
null
Results
[SUBTITLE] Baseline characteristics of patients and controls [SUBSECTION] Table 1 summarizes the baseline characteristics of the 37 patients, 11 of whom had bilateral C8/T1 radiculopathy. The mean patient age was 61.1 years and the mean height was 166.5 cm; 75.7% were men. The patients’ median NDI score was 12.0. Among all 48 C8/T1 root levels, 25 (52.1%) were right-sided, and the median duration of symptoms was 4.0 (3.0–12.0) months. The median NRS scores of the neck and arm were 3.0 and 4.5, respectively. Table 1Baseline features of patients with C8/T1 level radiculopathyVariablesValueaTotal patients, n37Age, years61.1 ± 12.2Male, n (%)28 (75.7)Height, cm166.5 ± 8.7Hypertension, n (%)16 (43.2)Diabetes, n (%)5 (13.5)Dyslipidemia, n (%)6 (16.2)NRS, neck3.0 (2.0–4.5)Total C8/T1 root levels, n48Right side, n (%)25 (52.1)Symptom duration, months4.0 (3.0–12.0)NDI12.0 (5.5–19.0)NRS, arm4.5 (3.0–6.0)Abbreviations: NDI, neck disability index; NRS, numerical rating scaleaUnless otherwise indicated, values are means ± standard deviations or medians (interquartile ranges) Baseline features of patients with C8/T1 level radiculopathy Abbreviations: NDI, neck disability index; NRS, numerical rating scale aUnless otherwise indicated, values are means ± standard deviations or medians (interquartile ranges) After performing PSM for comparative analysis, we extracted 48 control C8/T1 root levels and confirmed that there were no significant differences in age, sex, or height between the two groups (Table 2). Table 2Comparison of data from patients with C8/T1 level radiculopathy and matched control subjectsPatients (n = 48)Controls (n = 48)p-valueAge, years61.9 ± 12.458.8 ± 11.10.197Male, n (%)37 (77.1)28 (58.3)0.081Height, cm166.4 ± 8.6164.2 ± 8.80.223APB-RMCT, ms1.7 ± 0.61.2 ± 0.80.001Abbreviations: APB, abductor pollicis brevis; RMCT, root motor conduction time Comparison of data from patients with C8/T1 level radiculopathy and matched control subjects Abbreviations: APB, abductor pollicis brevis; RMCT, root motor conduction time Table 1 summarizes the baseline characteristics of the 37 patients, 11 of whom had bilateral C8/T1 radiculopathy. The mean patient age was 61.1 years and the mean height was 166.5 cm; 75.7% were men. The patients’ median NDI score was 12.0. Among all 48 C8/T1 root levels, 25 (52.1%) were right-sided, and the median duration of symptoms was 4.0 (3.0–12.0) months. The median NRS scores of the neck and arm were 3.0 and 4.5, respectively. Table 1Baseline features of patients with C8/T1 level radiculopathyVariablesValueaTotal patients, n37Age, years61.1 ± 12.2Male, n (%)28 (75.7)Height, cm166.5 ± 8.7Hypertension, n (%)16 (43.2)Diabetes, n (%)5 (13.5)Dyslipidemia, n (%)6 (16.2)NRS, neck3.0 (2.0–4.5)Total C8/T1 root levels, n48Right side, n (%)25 (52.1)Symptom duration, months4.0 (3.0–12.0)NDI12.0 (5.5–19.0)NRS, arm4.5 (3.0–6.0)Abbreviations: NDI, neck disability index; NRS, numerical rating scaleaUnless otherwise indicated, values are means ± standard deviations or medians (interquartile ranges) Baseline features of patients with C8/T1 level radiculopathy Abbreviations: NDI, neck disability index; NRS, numerical rating scale aUnless otherwise indicated, values are means ± standard deviations or medians (interquartile ranges) After performing PSM for comparative analysis, we extracted 48 control C8/T1 root levels and confirmed that there were no significant differences in age, sex, or height between the two groups (Table 2). Table 2Comparison of data from patients with C8/T1 level radiculopathy and matched control subjectsPatients (n = 48)Controls (n = 48)p-valueAge, years61.9 ± 12.458.8 ± 11.10.197Male, n (%)37 (77.1)28 (58.3)0.081Height, cm166.4 ± 8.6164.2 ± 8.80.223APB-RMCT, ms1.7 ± 0.61.2 ± 0.80.001Abbreviations: APB, abductor pollicis brevis; RMCT, root motor conduction time Comparison of data from patients with C8/T1 level radiculopathy and matched control subjects Abbreviations: APB, abductor pollicis brevis; RMCT, root motor conduction time [SUBTITLE] RMCT at the C8/T1 level [SUBSECTION] The mean RMCT among patients with radiculopathy was 1.7 ± 0.6 ms, which was significantly longer than that among control subjects (1.2 ± 0.8 ms; p = 0.001) (Table 2; Fig. 3). Logistic regression models revealed that the delay in RMCT observed in patients with C8/T1 was independently associated with radiculopathy (per Model 3: odds ratio, 1.15; 95% confidence interval, 1.06–1.27; p = 0.011) (Table 3). The area under the Receiver Operating Characteristic curve for diagnosing C8/T1 radiculopathy using RMCT was 0.72 (0.61–0.82) with 0.83 sensitivity and 0.58 specificity (Supplementary Fig. 1). In terms of subjective symptom indices, the RMCT was significantly associated with symptom duration (coefficient = 0.58; p < 0.001) but not with the NRS of the arm (coefficient = 0.22; p = 0.139) All measured values for calculating the RMCT are presented in Supplementary Table 2. Fig. 3Root motor conduction time (RMCT) in each group. Patients with C8/T1 level radiculopathy showed significantly longer RMCTs than did the control group (p = 0.001) Root motor conduction time (RMCT) in each group. Patients with C8/T1 level radiculopathy showed significantly longer RMCTs than did the control group (p = 0.001) Table 3Logistic regression models using data from patients and matched control subjectsOdds ratio (95% CI)p-valueModel 1aRMCT (per 0.1 ms)1.11 (1.04–1.18)0.002Model 2bRMCT (per 0.1 ms)1.16 (1.06–1.27)0.002Model 3cRMCT (per 0.1 ms)1.15 (1.06–1.27)0.011Abbreviations: CI, confidence interval; RMCT, root motor conduction timeaUnivariable analysis; badjusted for stimulated peripheral motor conduction time; cadjusted for stimulated peripheral motor conduction time, amplitude of median compound motor nerve action potential, and F-wave latency Logistic regression models using data from patients and matched control subjects Abbreviations: CI, confidence interval; RMCT, root motor conduction time aUnivariable analysis; badjusted for stimulated peripheral motor conduction time; cadjusted for stimulated peripheral motor conduction time, amplitude of median compound motor nerve action potential, and F-wave latency The mean RMCT among patients with radiculopathy was 1.7 ± 0.6 ms, which was significantly longer than that among control subjects (1.2 ± 0.8 ms; p = 0.001) (Table 2; Fig. 3). Logistic regression models revealed that the delay in RMCT observed in patients with C8/T1 was independently associated with radiculopathy (per Model 3: odds ratio, 1.15; 95% confidence interval, 1.06–1.27; p = 0.011) (Table 3). The area under the Receiver Operating Characteristic curve for diagnosing C8/T1 radiculopathy using RMCT was 0.72 (0.61–0.82) with 0.83 sensitivity and 0.58 specificity (Supplementary Fig. 1). In terms of subjective symptom indices, the RMCT was significantly associated with symptom duration (coefficient = 0.58; p < 0.001) but not with the NRS of the arm (coefficient = 0.22; p = 0.139) All measured values for calculating the RMCT are presented in Supplementary Table 2. Fig. 3Root motor conduction time (RMCT) in each group. Patients with C8/T1 level radiculopathy showed significantly longer RMCTs than did the control group (p = 0.001) Root motor conduction time (RMCT) in each group. Patients with C8/T1 level radiculopathy showed significantly longer RMCTs than did the control group (p = 0.001) Table 3Logistic regression models using data from patients and matched control subjectsOdds ratio (95% CI)p-valueModel 1aRMCT (per 0.1 ms)1.11 (1.04–1.18)0.002Model 2bRMCT (per 0.1 ms)1.16 (1.06–1.27)0.002Model 3cRMCT (per 0.1 ms)1.15 (1.06–1.27)0.011Abbreviations: CI, confidence interval; RMCT, root motor conduction timeaUnivariable analysis; badjusted for stimulated peripheral motor conduction time; cadjusted for stimulated peripheral motor conduction time, amplitude of median compound motor nerve action potential, and F-wave latency Logistic regression models using data from patients and matched control subjects Abbreviations: CI, confidence interval; RMCT, root motor conduction time aUnivariable analysis; badjusted for stimulated peripheral motor conduction time; cadjusted for stimulated peripheral motor conduction time, amplitude of median compound motor nerve action potential, and F-wave latency
Conclusion
We demonstrated that the RMCT is delayed in the root lesions of patients with C8/T1 level radiculopathy. This noninvasive method (compared to EMG) can have an adjuvant role in diagnosing radiculopathy at certain spinal levels. Our study may be a good milestone for future clinical applications of RMCT as related to patients with radiculopathy. However, it is important to validate our results through additional multi-center, multi-ethnic studies.
[ "Methods", "Subjects and clinical evaluations", "Electrodiagnostic assessments", "Statistical analysis", "Baseline characteristics of patients and controls", "RMCT at the C8/T1 level", "" ]
[ "[SUBTITLE] Subjects and clinical evaluations [SUBSECTION] We performed a retrospective cross-sectional study based on medical records of patients treated between June 2007 and December 2021. We extracted patients diagnosed with C8/T1 radiculopathy at our hospital through electrodiagnosis and whose RMCT was measured at the same time. All patients diagnosed with C8/T1 radiculopathy also underwent cervical x-ray and magnetic resonance imaging studies to confirm root compression and exclude other differential diagnoses. Thereafter, the final study group was selected by applying the following exclusion criteria: uncontrolled diabetes, concomitant polyneuropathy or median nerve lesion, previous cervical spine surgery, previous hand injury or surgery, unobtainable electrodiagnostic parameters, and lack of clinical information. Data from 48 C8/T1 root lesions in 37 patients were finally extracted; these included the subjective symptom duration, numerical rating scale (NRS) of the neck and arm pain, and neck disability index (NDI) score at the time of electrodiagnostic examination.\nThe control group consisted of a single-center healthy cohort of 190 prospectively analyzed subjects who were reported previously by the authors [18]. We controlled the potential biases between the two groups by performing propensity score matching (PSM). Consequently, 96 root levels (48 patients and 48 matched controls) were finally investigated (Fig. 1).\n\nFig. 1Flowchart showing the comparative analysis of the patients and control subjects\n\nFlowchart showing the comparative analysis of the patients and control subjects\nThis study was approved by the Institutional Review Board of Pohang Stroke and Spine Hospital (PSSH0475-202207-HR-011-01); informed consent was waived, given its retrospective design. The research complied with the guidelines of the Declaration of Helsinki.\nWe performed a retrospective cross-sectional study based on medical records of patients treated between June 2007 and December 2021. We extracted patients diagnosed with C8/T1 radiculopathy at our hospital through electrodiagnosis and whose RMCT was measured at the same time. All patients diagnosed with C8/T1 radiculopathy also underwent cervical x-ray and magnetic resonance imaging studies to confirm root compression and exclude other differential diagnoses. Thereafter, the final study group was selected by applying the following exclusion criteria: uncontrolled diabetes, concomitant polyneuropathy or median nerve lesion, previous cervical spine surgery, previous hand injury or surgery, unobtainable electrodiagnostic parameters, and lack of clinical information. Data from 48 C8/T1 root lesions in 37 patients were finally extracted; these included the subjective symptom duration, numerical rating scale (NRS) of the neck and arm pain, and neck disability index (NDI) score at the time of electrodiagnostic examination.\nThe control group consisted of a single-center healthy cohort of 190 prospectively analyzed subjects who were reported previously by the authors [18]. We controlled the potential biases between the two groups by performing propensity score matching (PSM). Consequently, 96 root levels (48 patients and 48 matched controls) were finally investigated (Fig. 1).\n\nFig. 1Flowchart showing the comparative analysis of the patients and control subjects\n\nFlowchart showing the comparative analysis of the patients and control subjects\nThis study was approved by the Institutional Review Board of Pohang Stroke and Spine Hospital (PSSH0475-202207-HR-011-01); informed consent was waived, given its retrospective design. The research complied with the guidelines of the Declaration of Helsinki.\n[SUBTITLE] Electrodiagnostic assessments [SUBSECTION] The EMG protocol for radiculopathy comprised of the following [5]: First, we examined muscles innervated by different nerves in the same myotome. Second, we assessed both proximal and distal muscles in the same myotome. Finally, we examined muscles in the myotomes adjacent to the suspected level. EMG-confirmed C8/T1 radiculopathy was defined as the presence of denervation potentials or polyphasic, long-duration, and large-amplitude motor unit action potentials in two or more of the differently innervated following muscles after other conditions were ruled out: flexor pollicis longus, flexor digitorum profundus, flexor carpi radialis, first dorsal interosseous, abductor digiti minimi, and APB [5].\nFor the nerve conduction study, we first recorded the sensory nerve action potentials of the median, ulnar, and superficial radial nerve in the distal arm. Electrical stimulations with 0.1 ms square wave pulses were applied for the sensory nerve conduction studies with a 10–2000 Hz filter setting. Next, we recorded compound motor nerve action potentials from the APB and abductor digiti minimi muscles and F-waves from the APB muscle. To conduct motor nerve conduction studies, each nerve was supramaximally stimulated by 0.2 ms square wave pulses with a 5–5000 Hz filter setting. We recorded nerve action potentials at least 12 times at the same nerve for study reproducibility. Surface electrodes were attached using the belly-tendon method to all recorded muscles. All nerve conduction studies were performed in the supine position. The detailed methods for individual nerve conduction studies are described in Supplementary Table 1 [5, 19].\n\nTo measure the stimulated peripheral motor conduction time (PMCT), we performed magnetic stimulation at the C7 spinous process; the ABP muscle was recorded using the surface electrodes. To provoke median motor evoked potential, we administered supramaximal stimulation (20% above the threshold) with weak isometric contraction in the APB muscle. The simulations were applied biphasic and the active pulse width was 280 µs. Then, we calculated the RMCT using the following equations, which are based on the principle that nerve excitement occurs a few centimeters distal to the anterior horn cell upon magnetic stimulation (Fig. 2) [20].\n\nFig. 2Schematic diagram illustrating the measurement of C8/T1 root motor conduction time\n\nSchematic diagram illustrating the measurement of C8/T1 root motor conduction time\n\n\nRMCT (ms) = calculated PMCT − stimulated PMCTCalculated PMCT (ms) = (compound muscle action potential onset latency + F-wave onset latency − 1) / 2Stimulated PMCT (ms) = spinal motor-evoked potential onset latency\n\n\nRMCT (ms) = calculated PMCT − stimulated PMCT\nCalculated PMCT (ms) = (compound muscle action potential onset latency + F-wave onset latency − 1) / 2\nStimulated PMCT (ms) = spinal motor-evoked potential onset latency\nAll electrodiagnostic evaluations were performed using the Cadwell Sierra Wave EMG system (Cadwell Laboratories Inc., Kennewick, WA, USA). A MagPro Compact with a C-100 circular coil (11 cm outer diameter) (MagVenture Inc, Farum, Denmark) was used for cervical magnetic stimulation. The temperature of the electrodiagnostic laboratory was maintained at approximately 23℃ to 25℃. All electrodiagnostic examinations were interpreted by experienced physiatrists.\nThe EMG protocol for radiculopathy comprised of the following [5]: First, we examined muscles innervated by different nerves in the same myotome. Second, we assessed both proximal and distal muscles in the same myotome. Finally, we examined muscles in the myotomes adjacent to the suspected level. EMG-confirmed C8/T1 radiculopathy was defined as the presence of denervation potentials or polyphasic, long-duration, and large-amplitude motor unit action potentials in two or more of the differently innervated following muscles after other conditions were ruled out: flexor pollicis longus, flexor digitorum profundus, flexor carpi radialis, first dorsal interosseous, abductor digiti minimi, and APB [5].\nFor the nerve conduction study, we first recorded the sensory nerve action potentials of the median, ulnar, and superficial radial nerve in the distal arm. Electrical stimulations with 0.1 ms square wave pulses were applied for the sensory nerve conduction studies with a 10–2000 Hz filter setting. Next, we recorded compound motor nerve action potentials from the APB and abductor digiti minimi muscles and F-waves from the APB muscle. To conduct motor nerve conduction studies, each nerve was supramaximally stimulated by 0.2 ms square wave pulses with a 5–5000 Hz filter setting. We recorded nerve action potentials at least 12 times at the same nerve for study reproducibility. Surface electrodes were attached using the belly-tendon method to all recorded muscles. All nerve conduction studies were performed in the supine position. The detailed methods for individual nerve conduction studies are described in Supplementary Table 1 [5, 19].\n\nTo measure the stimulated peripheral motor conduction time (PMCT), we performed magnetic stimulation at the C7 spinous process; the ABP muscle was recorded using the surface electrodes. To provoke median motor evoked potential, we administered supramaximal stimulation (20% above the threshold) with weak isometric contraction in the APB muscle. The simulations were applied biphasic and the active pulse width was 280 µs. Then, we calculated the RMCT using the following equations, which are based on the principle that nerve excitement occurs a few centimeters distal to the anterior horn cell upon magnetic stimulation (Fig. 2) [20].\n\nFig. 2Schematic diagram illustrating the measurement of C8/T1 root motor conduction time\n\nSchematic diagram illustrating the measurement of C8/T1 root motor conduction time\n\n\nRMCT (ms) = calculated PMCT − stimulated PMCTCalculated PMCT (ms) = (compound muscle action potential onset latency + F-wave onset latency − 1) / 2Stimulated PMCT (ms) = spinal motor-evoked potential onset latency\n\n\nRMCT (ms) = calculated PMCT − stimulated PMCT\nCalculated PMCT (ms) = (compound muscle action potential onset latency + F-wave onset latency − 1) / 2\nStimulated PMCT (ms) = spinal motor-evoked potential onset latency\nAll electrodiagnostic evaluations were performed using the Cadwell Sierra Wave EMG system (Cadwell Laboratories Inc., Kennewick, WA, USA). A MagPro Compact with a C-100 circular coil (11 cm outer diameter) (MagVenture Inc, Farum, Denmark) was used for cervical magnetic stimulation. The temperature of the electrodiagnostic laboratory was maintained at approximately 23℃ to 25℃. All electrodiagnostic examinations were interpreted by experienced physiatrists.\n[SUBTITLE] Statistical analysis [SUBSECTION] The Shapiro-Wilk test was applied to check the normality of continuous variables. These are expressed as means ± standard deviations if normality was satisfied or as medians (interquartile ranges) if not; the independent t-test and Mann-Whitney U-test were applied for comparative analyses of these types of values, respectively. Categorical variables are expressed as frequencies (proportions); groups were compared using the chi-squared test. We established binary logistic regression models for detecting C8/T1 root lesions using data adjusted for age, sex, and height. Model 1 was a predictive model of RMCT alone, Model 2 was adjusted for the stimulated PMCT, and Model 3 was adjusted for the median compound muscle action potential and F-wave in addition to the adjustment of Model 2. The multicollinearity of the model was confirmed, as the variance inflation factor was < 10. We drew the Receiver Operating Characteristic curve and calculated the cutoff value using Youden’s J statistic. We calculated the Spearman coefficient to determine the correlation between RMCT and symptoms. All statistical analyses were performed using SPSS 22.0 (IBM Inc., Armonk, NY, USA).\nFor PSM, the “MatchIt” package of the R software version 4.1.2 (R Core Team, R Foundation for Statistical Computing, Vienna, Austria) was used [21]. The covariates used for matching were age, sex, and height; moreover, 1:1 matching with no replacement was performed. The nearest-neighbor method was applied with the caliper set to 0.2.\nThe Shapiro-Wilk test was applied to check the normality of continuous variables. These are expressed as means ± standard deviations if normality was satisfied or as medians (interquartile ranges) if not; the independent t-test and Mann-Whitney U-test were applied for comparative analyses of these types of values, respectively. Categorical variables are expressed as frequencies (proportions); groups were compared using the chi-squared test. We established binary logistic regression models for detecting C8/T1 root lesions using data adjusted for age, sex, and height. Model 1 was a predictive model of RMCT alone, Model 2 was adjusted for the stimulated PMCT, and Model 3 was adjusted for the median compound muscle action potential and F-wave in addition to the adjustment of Model 2. The multicollinearity of the model was confirmed, as the variance inflation factor was < 10. We drew the Receiver Operating Characteristic curve and calculated the cutoff value using Youden’s J statistic. We calculated the Spearman coefficient to determine the correlation between RMCT and symptoms. All statistical analyses were performed using SPSS 22.0 (IBM Inc., Armonk, NY, USA).\nFor PSM, the “MatchIt” package of the R software version 4.1.2 (R Core Team, R Foundation for Statistical Computing, Vienna, Austria) was used [21]. The covariates used for matching were age, sex, and height; moreover, 1:1 matching with no replacement was performed. The nearest-neighbor method was applied with the caliper set to 0.2.", "We performed a retrospective cross-sectional study based on medical records of patients treated between June 2007 and December 2021. We extracted patients diagnosed with C8/T1 radiculopathy at our hospital through electrodiagnosis and whose RMCT was measured at the same time. All patients diagnosed with C8/T1 radiculopathy also underwent cervical x-ray and magnetic resonance imaging studies to confirm root compression and exclude other differential diagnoses. Thereafter, the final study group was selected by applying the following exclusion criteria: uncontrolled diabetes, concomitant polyneuropathy or median nerve lesion, previous cervical spine surgery, previous hand injury or surgery, unobtainable electrodiagnostic parameters, and lack of clinical information. Data from 48 C8/T1 root lesions in 37 patients were finally extracted; these included the subjective symptom duration, numerical rating scale (NRS) of the neck and arm pain, and neck disability index (NDI) score at the time of electrodiagnostic examination.\nThe control group consisted of a single-center healthy cohort of 190 prospectively analyzed subjects who were reported previously by the authors [18]. We controlled the potential biases between the two groups by performing propensity score matching (PSM). Consequently, 96 root levels (48 patients and 48 matched controls) were finally investigated (Fig. 1).\n\nFig. 1Flowchart showing the comparative analysis of the patients and control subjects\n\nFlowchart showing the comparative analysis of the patients and control subjects\nThis study was approved by the Institutional Review Board of Pohang Stroke and Spine Hospital (PSSH0475-202207-HR-011-01); informed consent was waived, given its retrospective design. The research complied with the guidelines of the Declaration of Helsinki.", "The EMG protocol for radiculopathy comprised of the following [5]: First, we examined muscles innervated by different nerves in the same myotome. Second, we assessed both proximal and distal muscles in the same myotome. Finally, we examined muscles in the myotomes adjacent to the suspected level. EMG-confirmed C8/T1 radiculopathy was defined as the presence of denervation potentials or polyphasic, long-duration, and large-amplitude motor unit action potentials in two or more of the differently innervated following muscles after other conditions were ruled out: flexor pollicis longus, flexor digitorum profundus, flexor carpi radialis, first dorsal interosseous, abductor digiti minimi, and APB [5].\nFor the nerve conduction study, we first recorded the sensory nerve action potentials of the median, ulnar, and superficial radial nerve in the distal arm. Electrical stimulations with 0.1 ms square wave pulses were applied for the sensory nerve conduction studies with a 10–2000 Hz filter setting. Next, we recorded compound motor nerve action potentials from the APB and abductor digiti minimi muscles and F-waves from the APB muscle. To conduct motor nerve conduction studies, each nerve was supramaximally stimulated by 0.2 ms square wave pulses with a 5–5000 Hz filter setting. We recorded nerve action potentials at least 12 times at the same nerve for study reproducibility. Surface electrodes were attached using the belly-tendon method to all recorded muscles. All nerve conduction studies were performed in the supine position. The detailed methods for individual nerve conduction studies are described in Supplementary Table 1 [5, 19].\n\nTo measure the stimulated peripheral motor conduction time (PMCT), we performed magnetic stimulation at the C7 spinous process; the ABP muscle was recorded using the surface electrodes. To provoke median motor evoked potential, we administered supramaximal stimulation (20% above the threshold) with weak isometric contraction in the APB muscle. The simulations were applied biphasic and the active pulse width was 280 µs. Then, we calculated the RMCT using the following equations, which are based on the principle that nerve excitement occurs a few centimeters distal to the anterior horn cell upon magnetic stimulation (Fig. 2) [20].\n\nFig. 2Schematic diagram illustrating the measurement of C8/T1 root motor conduction time\n\nSchematic diagram illustrating the measurement of C8/T1 root motor conduction time\n\n\nRMCT (ms) = calculated PMCT − stimulated PMCTCalculated PMCT (ms) = (compound muscle action potential onset latency + F-wave onset latency − 1) / 2Stimulated PMCT (ms) = spinal motor-evoked potential onset latency\n\n\nRMCT (ms) = calculated PMCT − stimulated PMCT\nCalculated PMCT (ms) = (compound muscle action potential onset latency + F-wave onset latency − 1) / 2\nStimulated PMCT (ms) = spinal motor-evoked potential onset latency\nAll electrodiagnostic evaluations were performed using the Cadwell Sierra Wave EMG system (Cadwell Laboratories Inc., Kennewick, WA, USA). A MagPro Compact with a C-100 circular coil (11 cm outer diameter) (MagVenture Inc, Farum, Denmark) was used for cervical magnetic stimulation. The temperature of the electrodiagnostic laboratory was maintained at approximately 23℃ to 25℃. All electrodiagnostic examinations were interpreted by experienced physiatrists.", "The Shapiro-Wilk test was applied to check the normality of continuous variables. These are expressed as means ± standard deviations if normality was satisfied or as medians (interquartile ranges) if not; the independent t-test and Mann-Whitney U-test were applied for comparative analyses of these types of values, respectively. Categorical variables are expressed as frequencies (proportions); groups were compared using the chi-squared test. We established binary logistic regression models for detecting C8/T1 root lesions using data adjusted for age, sex, and height. Model 1 was a predictive model of RMCT alone, Model 2 was adjusted for the stimulated PMCT, and Model 3 was adjusted for the median compound muscle action potential and F-wave in addition to the adjustment of Model 2. The multicollinearity of the model was confirmed, as the variance inflation factor was < 10. We drew the Receiver Operating Characteristic curve and calculated the cutoff value using Youden’s J statistic. We calculated the Spearman coefficient to determine the correlation between RMCT and symptoms. All statistical analyses were performed using SPSS 22.0 (IBM Inc., Armonk, NY, USA).\nFor PSM, the “MatchIt” package of the R software version 4.1.2 (R Core Team, R Foundation for Statistical Computing, Vienna, Austria) was used [21]. The covariates used for matching were age, sex, and height; moreover, 1:1 matching with no replacement was performed. The nearest-neighbor method was applied with the caliper set to 0.2.", "Table 1 summarizes the baseline characteristics of the 37 patients, 11 of whom had bilateral C8/T1 radiculopathy. The mean patient age was 61.1 years and the mean height was 166.5 cm; 75.7% were men. The patients’ median NDI score was 12.0. Among all 48 C8/T1 root levels, 25 (52.1%) were right-sided, and the median duration of symptoms was 4.0 (3.0–12.0) months. The median NRS scores of the neck and arm were 3.0 and 4.5, respectively.\n\nTable 1Baseline features of patients with C8/T1 level radiculopathyVariablesValueaTotal patients, n37Age, years61.1 ± 12.2Male, n (%)28 (75.7)Height, cm166.5 ± 8.7Hypertension, n (%)16 (43.2)Diabetes, n (%)5 (13.5)Dyslipidemia, n (%)6 (16.2)NRS, neck3.0 (2.0–4.5)Total C8/T1 root levels, n48Right side, n (%)25 (52.1)Symptom duration, months4.0 (3.0–12.0)NDI12.0 (5.5–19.0)NRS, arm4.5 (3.0–6.0)Abbreviations: NDI, neck disability index; NRS, numerical rating scaleaUnless otherwise indicated, values are means ± standard deviations or medians (interquartile ranges)\n\nBaseline features of patients with C8/T1 level radiculopathy\nAbbreviations: NDI, neck disability index; NRS, numerical rating scale\naUnless otherwise indicated, values are means ± standard deviations or medians (interquartile ranges)\nAfter performing PSM for comparative analysis, we extracted 48 control C8/T1 root levels and confirmed that there were no significant differences in age, sex, or height between the two groups (Table 2).\n\nTable 2Comparison of data from patients with C8/T1 level radiculopathy and matched control subjectsPatients (n = 48)Controls (n = 48)p-valueAge, years61.9 ± 12.458.8 ± 11.10.197Male, n (%)37 (77.1)28 (58.3)0.081Height, cm166.4 ± 8.6164.2 ± 8.80.223APB-RMCT, ms1.7 ± 0.61.2 ± 0.80.001Abbreviations: APB, abductor pollicis brevis; RMCT, root motor conduction time\n\nComparison of data from patients with C8/T1 level radiculopathy and matched control subjects\nAbbreviations: APB, abductor pollicis brevis; RMCT, root motor conduction time", "The mean RMCT among patients with radiculopathy was 1.7 ± 0.6 ms, which was significantly longer than that among control subjects (1.2 ± 0.8 ms; p = 0.001) (Table 2; Fig. 3). Logistic regression models revealed that the delay in RMCT observed in patients with C8/T1 was independently associated with radiculopathy (per Model 3: odds ratio, 1.15; 95% confidence interval, 1.06–1.27; p = 0.011) (Table 3). The area under the Receiver Operating Characteristic curve for diagnosing C8/T1 radiculopathy using RMCT was 0.72 (0.61–0.82) with 0.83 sensitivity and 0.58 specificity (Supplementary Fig. 1). In terms of subjective symptom indices, the RMCT was significantly associated with symptom duration (coefficient = 0.58; p < 0.001) but not with the NRS of the arm (coefficient = 0.22; p = 0.139) All measured values for calculating the RMCT are presented in Supplementary Table 2.\n\nFig. 3Root motor conduction time (RMCT) in each group. Patients with C8/T1 level radiculopathy showed significantly longer RMCTs than did the control group (p = 0.001)\n\nRoot motor conduction time (RMCT) in each group. Patients with C8/T1 level radiculopathy showed significantly longer RMCTs than did the control group (p = 0.001)\n\nTable 3Logistic regression models using data from patients and matched control subjectsOdds ratio (95% CI)p-valueModel 1aRMCT (per 0.1 ms)1.11 (1.04–1.18)0.002Model 2bRMCT (per 0.1 ms)1.16 (1.06–1.27)0.002Model 3cRMCT (per 0.1 ms)1.15 (1.06–1.27)0.011Abbreviations: CI, confidence interval; RMCT, root motor conduction timeaUnivariable analysis; badjusted for stimulated peripheral motor conduction time; cadjusted for stimulated peripheral motor conduction time, amplitude of median compound motor nerve action potential, and F-wave latency\n\nLogistic regression models using data from patients and matched control subjects\nAbbreviations: CI, confidence interval; RMCT, root motor conduction time\naUnivariable analysis; badjusted for stimulated peripheral motor conduction time; cadjusted for stimulated peripheral motor conduction time, amplitude of median compound motor nerve action potential, and F-wave latency", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, null, null, null, null, null, null ]
[ "Introduction", "Methods", "Subjects and clinical evaluations", "Electrodiagnostic assessments", "Statistical analysis", "Results", "Baseline characteristics of patients and controls", "RMCT at the C8/T1 level", "Discussion", "Conclusion", "Electronic supplementary material", "" ]
[ "Radiculopathy is a common condition whose symptoms can include pain, sensory change, and motor weakness owing to mechanical and chemical irritation of the spinal nerve root [1, 2]. Imaging studies are essential for diagnosing radiculopathy; magnetic resonance imaging helps identify soft tissues such as discs, ligaments, and nerve roots [3], while computed tomography is primarily used to confirm abnormalities in bony structures [4]. Meanwhile, another key factor in confirming radiculopathy is electrophysiologic diagnosis [5]. In particular, electromyography (EMG) is a classical tool that was used to evaluate radiculopathy before the development of modern imaging instruments [6]; its key advantage is in its ability to detect neurophysiological changes that cannot be confirmed by imaging studies alone [7]. By applying EMG to multiple myotomes, it is possible not only to localize the abnormal spinal root level but also to estimate the temporal aspect of compression by distinguishing motor unit action potential [8]. However, the invasive nature of EMG creates a potential risk of complications such as bleeding or infection at the examination site [9]. Additionally, patient cooperation is essential for observing resting potential and interference patterns [10].\nRoot motor conduction time (RMCT) is a measure of the state of the proximal root segment that is obtained noninvasively by performing direct cervical stimulation and nerve conduction studies [11, 12]. Notably, RMCT is very localized since it excludes distal neural lesions and measures only the conduction time of the proximal root segment [13–15]. However, it also has the disadvantage of involving a somewhat complicated measurement method wherein well-trained examiners are required [16]. To date, the RMCT has been investigated in only a few disease types, such as demyelinating disease and lumbar spinal stenosis, whereas its clinical applicability in patients with radiculopathy remains unclear [17].\nIn this study, we hypothesized that the RMCT can be a valuable tool for the diagnosis of radiculopathy at certain spinal root levels. To that end, we investigated whether the RMCT measured in the abductor pollicis brevis (APB) muscle was significantly delayed in the presence of C8/T1 radiculopathy. Moreover, we aimed to determine the association between clinical symptoms and RMCT.", "[SUBTITLE] Subjects and clinical evaluations [SUBSECTION] We performed a retrospective cross-sectional study based on medical records of patients treated between June 2007 and December 2021. We extracted patients diagnosed with C8/T1 radiculopathy at our hospital through electrodiagnosis and whose RMCT was measured at the same time. All patients diagnosed with C8/T1 radiculopathy also underwent cervical x-ray and magnetic resonance imaging studies to confirm root compression and exclude other differential diagnoses. Thereafter, the final study group was selected by applying the following exclusion criteria: uncontrolled diabetes, concomitant polyneuropathy or median nerve lesion, previous cervical spine surgery, previous hand injury or surgery, unobtainable electrodiagnostic parameters, and lack of clinical information. Data from 48 C8/T1 root lesions in 37 patients were finally extracted; these included the subjective symptom duration, numerical rating scale (NRS) of the neck and arm pain, and neck disability index (NDI) score at the time of electrodiagnostic examination.\nThe control group consisted of a single-center healthy cohort of 190 prospectively analyzed subjects who were reported previously by the authors [18]. We controlled the potential biases between the two groups by performing propensity score matching (PSM). Consequently, 96 root levels (48 patients and 48 matched controls) were finally investigated (Fig. 1).\n\nFig. 1Flowchart showing the comparative analysis of the patients and control subjects\n\nFlowchart showing the comparative analysis of the patients and control subjects\nThis study was approved by the Institutional Review Board of Pohang Stroke and Spine Hospital (PSSH0475-202207-HR-011-01); informed consent was waived, given its retrospective design. The research complied with the guidelines of the Declaration of Helsinki.\nWe performed a retrospective cross-sectional study based on medical records of patients treated between June 2007 and December 2021. We extracted patients diagnosed with C8/T1 radiculopathy at our hospital through electrodiagnosis and whose RMCT was measured at the same time. All patients diagnosed with C8/T1 radiculopathy also underwent cervical x-ray and magnetic resonance imaging studies to confirm root compression and exclude other differential diagnoses. Thereafter, the final study group was selected by applying the following exclusion criteria: uncontrolled diabetes, concomitant polyneuropathy or median nerve lesion, previous cervical spine surgery, previous hand injury or surgery, unobtainable electrodiagnostic parameters, and lack of clinical information. Data from 48 C8/T1 root lesions in 37 patients were finally extracted; these included the subjective symptom duration, numerical rating scale (NRS) of the neck and arm pain, and neck disability index (NDI) score at the time of electrodiagnostic examination.\nThe control group consisted of a single-center healthy cohort of 190 prospectively analyzed subjects who were reported previously by the authors [18]. We controlled the potential biases between the two groups by performing propensity score matching (PSM). Consequently, 96 root levels (48 patients and 48 matched controls) were finally investigated (Fig. 1).\n\nFig. 1Flowchart showing the comparative analysis of the patients and control subjects\n\nFlowchart showing the comparative analysis of the patients and control subjects\nThis study was approved by the Institutional Review Board of Pohang Stroke and Spine Hospital (PSSH0475-202207-HR-011-01); informed consent was waived, given its retrospective design. The research complied with the guidelines of the Declaration of Helsinki.\n[SUBTITLE] Electrodiagnostic assessments [SUBSECTION] The EMG protocol for radiculopathy comprised of the following [5]: First, we examined muscles innervated by different nerves in the same myotome. Second, we assessed both proximal and distal muscles in the same myotome. Finally, we examined muscles in the myotomes adjacent to the suspected level. EMG-confirmed C8/T1 radiculopathy was defined as the presence of denervation potentials or polyphasic, long-duration, and large-amplitude motor unit action potentials in two or more of the differently innervated following muscles after other conditions were ruled out: flexor pollicis longus, flexor digitorum profundus, flexor carpi radialis, first dorsal interosseous, abductor digiti minimi, and APB [5].\nFor the nerve conduction study, we first recorded the sensory nerve action potentials of the median, ulnar, and superficial radial nerve in the distal arm. Electrical stimulations with 0.1 ms square wave pulses were applied for the sensory nerve conduction studies with a 10–2000 Hz filter setting. Next, we recorded compound motor nerve action potentials from the APB and abductor digiti minimi muscles and F-waves from the APB muscle. To conduct motor nerve conduction studies, each nerve was supramaximally stimulated by 0.2 ms square wave pulses with a 5–5000 Hz filter setting. We recorded nerve action potentials at least 12 times at the same nerve for study reproducibility. Surface electrodes were attached using the belly-tendon method to all recorded muscles. All nerve conduction studies were performed in the supine position. The detailed methods for individual nerve conduction studies are described in Supplementary Table 1 [5, 19].\n\nTo measure the stimulated peripheral motor conduction time (PMCT), we performed magnetic stimulation at the C7 spinous process; the ABP muscle was recorded using the surface electrodes. To provoke median motor evoked potential, we administered supramaximal stimulation (20% above the threshold) with weak isometric contraction in the APB muscle. The simulations were applied biphasic and the active pulse width was 280 µs. Then, we calculated the RMCT using the following equations, which are based on the principle that nerve excitement occurs a few centimeters distal to the anterior horn cell upon magnetic stimulation (Fig. 2) [20].\n\nFig. 2Schematic diagram illustrating the measurement of C8/T1 root motor conduction time\n\nSchematic diagram illustrating the measurement of C8/T1 root motor conduction time\n\n\nRMCT (ms) = calculated PMCT − stimulated PMCTCalculated PMCT (ms) = (compound muscle action potential onset latency + F-wave onset latency − 1) / 2Stimulated PMCT (ms) = spinal motor-evoked potential onset latency\n\n\nRMCT (ms) = calculated PMCT − stimulated PMCT\nCalculated PMCT (ms) = (compound muscle action potential onset latency + F-wave onset latency − 1) / 2\nStimulated PMCT (ms) = spinal motor-evoked potential onset latency\nAll electrodiagnostic evaluations were performed using the Cadwell Sierra Wave EMG system (Cadwell Laboratories Inc., Kennewick, WA, USA). A MagPro Compact with a C-100 circular coil (11 cm outer diameter) (MagVenture Inc, Farum, Denmark) was used for cervical magnetic stimulation. The temperature of the electrodiagnostic laboratory was maintained at approximately 23℃ to 25℃. All electrodiagnostic examinations were interpreted by experienced physiatrists.\nThe EMG protocol for radiculopathy comprised of the following [5]: First, we examined muscles innervated by different nerves in the same myotome. Second, we assessed both proximal and distal muscles in the same myotome. Finally, we examined muscles in the myotomes adjacent to the suspected level. EMG-confirmed C8/T1 radiculopathy was defined as the presence of denervation potentials or polyphasic, long-duration, and large-amplitude motor unit action potentials in two or more of the differently innervated following muscles after other conditions were ruled out: flexor pollicis longus, flexor digitorum profundus, flexor carpi radialis, first dorsal interosseous, abductor digiti minimi, and APB [5].\nFor the nerve conduction study, we first recorded the sensory nerve action potentials of the median, ulnar, and superficial radial nerve in the distal arm. Electrical stimulations with 0.1 ms square wave pulses were applied for the sensory nerve conduction studies with a 10–2000 Hz filter setting. Next, we recorded compound motor nerve action potentials from the APB and abductor digiti minimi muscles and F-waves from the APB muscle. To conduct motor nerve conduction studies, each nerve was supramaximally stimulated by 0.2 ms square wave pulses with a 5–5000 Hz filter setting. We recorded nerve action potentials at least 12 times at the same nerve for study reproducibility. Surface electrodes were attached using the belly-tendon method to all recorded muscles. All nerve conduction studies were performed in the supine position. The detailed methods for individual nerve conduction studies are described in Supplementary Table 1 [5, 19].\n\nTo measure the stimulated peripheral motor conduction time (PMCT), we performed magnetic stimulation at the C7 spinous process; the ABP muscle was recorded using the surface electrodes. To provoke median motor evoked potential, we administered supramaximal stimulation (20% above the threshold) with weak isometric contraction in the APB muscle. The simulations were applied biphasic and the active pulse width was 280 µs. Then, we calculated the RMCT using the following equations, which are based on the principle that nerve excitement occurs a few centimeters distal to the anterior horn cell upon magnetic stimulation (Fig. 2) [20].\n\nFig. 2Schematic diagram illustrating the measurement of C8/T1 root motor conduction time\n\nSchematic diagram illustrating the measurement of C8/T1 root motor conduction time\n\n\nRMCT (ms) = calculated PMCT − stimulated PMCTCalculated PMCT (ms) = (compound muscle action potential onset latency + F-wave onset latency − 1) / 2Stimulated PMCT (ms) = spinal motor-evoked potential onset latency\n\n\nRMCT (ms) = calculated PMCT − stimulated PMCT\nCalculated PMCT (ms) = (compound muscle action potential onset latency + F-wave onset latency − 1) / 2\nStimulated PMCT (ms) = spinal motor-evoked potential onset latency\nAll electrodiagnostic evaluations were performed using the Cadwell Sierra Wave EMG system (Cadwell Laboratories Inc., Kennewick, WA, USA). A MagPro Compact with a C-100 circular coil (11 cm outer diameter) (MagVenture Inc, Farum, Denmark) was used for cervical magnetic stimulation. The temperature of the electrodiagnostic laboratory was maintained at approximately 23℃ to 25℃. All electrodiagnostic examinations were interpreted by experienced physiatrists.\n[SUBTITLE] Statistical analysis [SUBSECTION] The Shapiro-Wilk test was applied to check the normality of continuous variables. These are expressed as means ± standard deviations if normality was satisfied or as medians (interquartile ranges) if not; the independent t-test and Mann-Whitney U-test were applied for comparative analyses of these types of values, respectively. Categorical variables are expressed as frequencies (proportions); groups were compared using the chi-squared test. We established binary logistic regression models for detecting C8/T1 root lesions using data adjusted for age, sex, and height. Model 1 was a predictive model of RMCT alone, Model 2 was adjusted for the stimulated PMCT, and Model 3 was adjusted for the median compound muscle action potential and F-wave in addition to the adjustment of Model 2. The multicollinearity of the model was confirmed, as the variance inflation factor was < 10. We drew the Receiver Operating Characteristic curve and calculated the cutoff value using Youden’s J statistic. We calculated the Spearman coefficient to determine the correlation between RMCT and symptoms. All statistical analyses were performed using SPSS 22.0 (IBM Inc., Armonk, NY, USA).\nFor PSM, the “MatchIt” package of the R software version 4.1.2 (R Core Team, R Foundation for Statistical Computing, Vienna, Austria) was used [21]. The covariates used for matching were age, sex, and height; moreover, 1:1 matching with no replacement was performed. The nearest-neighbor method was applied with the caliper set to 0.2.\nThe Shapiro-Wilk test was applied to check the normality of continuous variables. These are expressed as means ± standard deviations if normality was satisfied or as medians (interquartile ranges) if not; the independent t-test and Mann-Whitney U-test were applied for comparative analyses of these types of values, respectively. Categorical variables are expressed as frequencies (proportions); groups were compared using the chi-squared test. We established binary logistic regression models for detecting C8/T1 root lesions using data adjusted for age, sex, and height. Model 1 was a predictive model of RMCT alone, Model 2 was adjusted for the stimulated PMCT, and Model 3 was adjusted for the median compound muscle action potential and F-wave in addition to the adjustment of Model 2. The multicollinearity of the model was confirmed, as the variance inflation factor was < 10. We drew the Receiver Operating Characteristic curve and calculated the cutoff value using Youden’s J statistic. We calculated the Spearman coefficient to determine the correlation between RMCT and symptoms. All statistical analyses were performed using SPSS 22.0 (IBM Inc., Armonk, NY, USA).\nFor PSM, the “MatchIt” package of the R software version 4.1.2 (R Core Team, R Foundation for Statistical Computing, Vienna, Austria) was used [21]. The covariates used for matching were age, sex, and height; moreover, 1:1 matching with no replacement was performed. The nearest-neighbor method was applied with the caliper set to 0.2.", "We performed a retrospective cross-sectional study based on medical records of patients treated between June 2007 and December 2021. We extracted patients diagnosed with C8/T1 radiculopathy at our hospital through electrodiagnosis and whose RMCT was measured at the same time. All patients diagnosed with C8/T1 radiculopathy also underwent cervical x-ray and magnetic resonance imaging studies to confirm root compression and exclude other differential diagnoses. Thereafter, the final study group was selected by applying the following exclusion criteria: uncontrolled diabetes, concomitant polyneuropathy or median nerve lesion, previous cervical spine surgery, previous hand injury or surgery, unobtainable electrodiagnostic parameters, and lack of clinical information. Data from 48 C8/T1 root lesions in 37 patients were finally extracted; these included the subjective symptom duration, numerical rating scale (NRS) of the neck and arm pain, and neck disability index (NDI) score at the time of electrodiagnostic examination.\nThe control group consisted of a single-center healthy cohort of 190 prospectively analyzed subjects who were reported previously by the authors [18]. We controlled the potential biases between the two groups by performing propensity score matching (PSM). Consequently, 96 root levels (48 patients and 48 matched controls) were finally investigated (Fig. 1).\n\nFig. 1Flowchart showing the comparative analysis of the patients and control subjects\n\nFlowchart showing the comparative analysis of the patients and control subjects\nThis study was approved by the Institutional Review Board of Pohang Stroke and Spine Hospital (PSSH0475-202207-HR-011-01); informed consent was waived, given its retrospective design. The research complied with the guidelines of the Declaration of Helsinki.", "The EMG protocol for radiculopathy comprised of the following [5]: First, we examined muscles innervated by different nerves in the same myotome. Second, we assessed both proximal and distal muscles in the same myotome. Finally, we examined muscles in the myotomes adjacent to the suspected level. EMG-confirmed C8/T1 radiculopathy was defined as the presence of denervation potentials or polyphasic, long-duration, and large-amplitude motor unit action potentials in two or more of the differently innervated following muscles after other conditions were ruled out: flexor pollicis longus, flexor digitorum profundus, flexor carpi radialis, first dorsal interosseous, abductor digiti minimi, and APB [5].\nFor the nerve conduction study, we first recorded the sensory nerve action potentials of the median, ulnar, and superficial radial nerve in the distal arm. Electrical stimulations with 0.1 ms square wave pulses were applied for the sensory nerve conduction studies with a 10–2000 Hz filter setting. Next, we recorded compound motor nerve action potentials from the APB and abductor digiti minimi muscles and F-waves from the APB muscle. To conduct motor nerve conduction studies, each nerve was supramaximally stimulated by 0.2 ms square wave pulses with a 5–5000 Hz filter setting. We recorded nerve action potentials at least 12 times at the same nerve for study reproducibility. Surface electrodes were attached using the belly-tendon method to all recorded muscles. All nerve conduction studies were performed in the supine position. The detailed methods for individual nerve conduction studies are described in Supplementary Table 1 [5, 19].\n\nTo measure the stimulated peripheral motor conduction time (PMCT), we performed magnetic stimulation at the C7 spinous process; the ABP muscle was recorded using the surface electrodes. To provoke median motor evoked potential, we administered supramaximal stimulation (20% above the threshold) with weak isometric contraction in the APB muscle. The simulations were applied biphasic and the active pulse width was 280 µs. Then, we calculated the RMCT using the following equations, which are based on the principle that nerve excitement occurs a few centimeters distal to the anterior horn cell upon magnetic stimulation (Fig. 2) [20].\n\nFig. 2Schematic diagram illustrating the measurement of C8/T1 root motor conduction time\n\nSchematic diagram illustrating the measurement of C8/T1 root motor conduction time\n\n\nRMCT (ms) = calculated PMCT − stimulated PMCTCalculated PMCT (ms) = (compound muscle action potential onset latency + F-wave onset latency − 1) / 2Stimulated PMCT (ms) = spinal motor-evoked potential onset latency\n\n\nRMCT (ms) = calculated PMCT − stimulated PMCT\nCalculated PMCT (ms) = (compound muscle action potential onset latency + F-wave onset latency − 1) / 2\nStimulated PMCT (ms) = spinal motor-evoked potential onset latency\nAll electrodiagnostic evaluations were performed using the Cadwell Sierra Wave EMG system (Cadwell Laboratories Inc., Kennewick, WA, USA). A MagPro Compact with a C-100 circular coil (11 cm outer diameter) (MagVenture Inc, Farum, Denmark) was used for cervical magnetic stimulation. The temperature of the electrodiagnostic laboratory was maintained at approximately 23℃ to 25℃. All electrodiagnostic examinations were interpreted by experienced physiatrists.", "The Shapiro-Wilk test was applied to check the normality of continuous variables. These are expressed as means ± standard deviations if normality was satisfied or as medians (interquartile ranges) if not; the independent t-test and Mann-Whitney U-test were applied for comparative analyses of these types of values, respectively. Categorical variables are expressed as frequencies (proportions); groups were compared using the chi-squared test. We established binary logistic regression models for detecting C8/T1 root lesions using data adjusted for age, sex, and height. Model 1 was a predictive model of RMCT alone, Model 2 was adjusted for the stimulated PMCT, and Model 3 was adjusted for the median compound muscle action potential and F-wave in addition to the adjustment of Model 2. The multicollinearity of the model was confirmed, as the variance inflation factor was < 10. We drew the Receiver Operating Characteristic curve and calculated the cutoff value using Youden’s J statistic. We calculated the Spearman coefficient to determine the correlation between RMCT and symptoms. All statistical analyses were performed using SPSS 22.0 (IBM Inc., Armonk, NY, USA).\nFor PSM, the “MatchIt” package of the R software version 4.1.2 (R Core Team, R Foundation for Statistical Computing, Vienna, Austria) was used [21]. The covariates used for matching were age, sex, and height; moreover, 1:1 matching with no replacement was performed. The nearest-neighbor method was applied with the caliper set to 0.2.", "[SUBTITLE] Baseline characteristics of patients and controls [SUBSECTION] Table 1 summarizes the baseline characteristics of the 37 patients, 11 of whom had bilateral C8/T1 radiculopathy. The mean patient age was 61.1 years and the mean height was 166.5 cm; 75.7% were men. The patients’ median NDI score was 12.0. Among all 48 C8/T1 root levels, 25 (52.1%) were right-sided, and the median duration of symptoms was 4.0 (3.0–12.0) months. The median NRS scores of the neck and arm were 3.0 and 4.5, respectively.\n\nTable 1Baseline features of patients with C8/T1 level radiculopathyVariablesValueaTotal patients, n37Age, years61.1 ± 12.2Male, n (%)28 (75.7)Height, cm166.5 ± 8.7Hypertension, n (%)16 (43.2)Diabetes, n (%)5 (13.5)Dyslipidemia, n (%)6 (16.2)NRS, neck3.0 (2.0–4.5)Total C8/T1 root levels, n48Right side, n (%)25 (52.1)Symptom duration, months4.0 (3.0–12.0)NDI12.0 (5.5–19.0)NRS, arm4.5 (3.0–6.0)Abbreviations: NDI, neck disability index; NRS, numerical rating scaleaUnless otherwise indicated, values are means ± standard deviations or medians (interquartile ranges)\n\nBaseline features of patients with C8/T1 level radiculopathy\nAbbreviations: NDI, neck disability index; NRS, numerical rating scale\naUnless otherwise indicated, values are means ± standard deviations or medians (interquartile ranges)\nAfter performing PSM for comparative analysis, we extracted 48 control C8/T1 root levels and confirmed that there were no significant differences in age, sex, or height between the two groups (Table 2).\n\nTable 2Comparison of data from patients with C8/T1 level radiculopathy and matched control subjectsPatients (n = 48)Controls (n = 48)p-valueAge, years61.9 ± 12.458.8 ± 11.10.197Male, n (%)37 (77.1)28 (58.3)0.081Height, cm166.4 ± 8.6164.2 ± 8.80.223APB-RMCT, ms1.7 ± 0.61.2 ± 0.80.001Abbreviations: APB, abductor pollicis brevis; RMCT, root motor conduction time\n\nComparison of data from patients with C8/T1 level radiculopathy and matched control subjects\nAbbreviations: APB, abductor pollicis brevis; RMCT, root motor conduction time\nTable 1 summarizes the baseline characteristics of the 37 patients, 11 of whom had bilateral C8/T1 radiculopathy. The mean patient age was 61.1 years and the mean height was 166.5 cm; 75.7% were men. The patients’ median NDI score was 12.0. Among all 48 C8/T1 root levels, 25 (52.1%) were right-sided, and the median duration of symptoms was 4.0 (3.0–12.0) months. The median NRS scores of the neck and arm were 3.0 and 4.5, respectively.\n\nTable 1Baseline features of patients with C8/T1 level radiculopathyVariablesValueaTotal patients, n37Age, years61.1 ± 12.2Male, n (%)28 (75.7)Height, cm166.5 ± 8.7Hypertension, n (%)16 (43.2)Diabetes, n (%)5 (13.5)Dyslipidemia, n (%)6 (16.2)NRS, neck3.0 (2.0–4.5)Total C8/T1 root levels, n48Right side, n (%)25 (52.1)Symptom duration, months4.0 (3.0–12.0)NDI12.0 (5.5–19.0)NRS, arm4.5 (3.0–6.0)Abbreviations: NDI, neck disability index; NRS, numerical rating scaleaUnless otherwise indicated, values are means ± standard deviations or medians (interquartile ranges)\n\nBaseline features of patients with C8/T1 level radiculopathy\nAbbreviations: NDI, neck disability index; NRS, numerical rating scale\naUnless otherwise indicated, values are means ± standard deviations or medians (interquartile ranges)\nAfter performing PSM for comparative analysis, we extracted 48 control C8/T1 root levels and confirmed that there were no significant differences in age, sex, or height between the two groups (Table 2).\n\nTable 2Comparison of data from patients with C8/T1 level radiculopathy and matched control subjectsPatients (n = 48)Controls (n = 48)p-valueAge, years61.9 ± 12.458.8 ± 11.10.197Male, n (%)37 (77.1)28 (58.3)0.081Height, cm166.4 ± 8.6164.2 ± 8.80.223APB-RMCT, ms1.7 ± 0.61.2 ± 0.80.001Abbreviations: APB, abductor pollicis brevis; RMCT, root motor conduction time\n\nComparison of data from patients with C8/T1 level radiculopathy and matched control subjects\nAbbreviations: APB, abductor pollicis brevis; RMCT, root motor conduction time\n[SUBTITLE] RMCT at the C8/T1 level [SUBSECTION] The mean RMCT among patients with radiculopathy was 1.7 ± 0.6 ms, which was significantly longer than that among control subjects (1.2 ± 0.8 ms; p = 0.001) (Table 2; Fig. 3). Logistic regression models revealed that the delay in RMCT observed in patients with C8/T1 was independently associated with radiculopathy (per Model 3: odds ratio, 1.15; 95% confidence interval, 1.06–1.27; p = 0.011) (Table 3). The area under the Receiver Operating Characteristic curve for diagnosing C8/T1 radiculopathy using RMCT was 0.72 (0.61–0.82) with 0.83 sensitivity and 0.58 specificity (Supplementary Fig. 1). In terms of subjective symptom indices, the RMCT was significantly associated with symptom duration (coefficient = 0.58; p < 0.001) but not with the NRS of the arm (coefficient = 0.22; p = 0.139) All measured values for calculating the RMCT are presented in Supplementary Table 2.\n\nFig. 3Root motor conduction time (RMCT) in each group. Patients with C8/T1 level radiculopathy showed significantly longer RMCTs than did the control group (p = 0.001)\n\nRoot motor conduction time (RMCT) in each group. Patients with C8/T1 level radiculopathy showed significantly longer RMCTs than did the control group (p = 0.001)\n\nTable 3Logistic regression models using data from patients and matched control subjectsOdds ratio (95% CI)p-valueModel 1aRMCT (per 0.1 ms)1.11 (1.04–1.18)0.002Model 2bRMCT (per 0.1 ms)1.16 (1.06–1.27)0.002Model 3cRMCT (per 0.1 ms)1.15 (1.06–1.27)0.011Abbreviations: CI, confidence interval; RMCT, root motor conduction timeaUnivariable analysis; badjusted for stimulated peripheral motor conduction time; cadjusted for stimulated peripheral motor conduction time, amplitude of median compound motor nerve action potential, and F-wave latency\n\nLogistic regression models using data from patients and matched control subjects\nAbbreviations: CI, confidence interval; RMCT, root motor conduction time\naUnivariable analysis; badjusted for stimulated peripheral motor conduction time; cadjusted for stimulated peripheral motor conduction time, amplitude of median compound motor nerve action potential, and F-wave latency\nThe mean RMCT among patients with radiculopathy was 1.7 ± 0.6 ms, which was significantly longer than that among control subjects (1.2 ± 0.8 ms; p = 0.001) (Table 2; Fig. 3). Logistic regression models revealed that the delay in RMCT observed in patients with C8/T1 was independently associated with radiculopathy (per Model 3: odds ratio, 1.15; 95% confidence interval, 1.06–1.27; p = 0.011) (Table 3). The area under the Receiver Operating Characteristic curve for diagnosing C8/T1 radiculopathy using RMCT was 0.72 (0.61–0.82) with 0.83 sensitivity and 0.58 specificity (Supplementary Fig. 1). In terms of subjective symptom indices, the RMCT was significantly associated with symptom duration (coefficient = 0.58; p < 0.001) but not with the NRS of the arm (coefficient = 0.22; p = 0.139) All measured values for calculating the RMCT are presented in Supplementary Table 2.\n\nFig. 3Root motor conduction time (RMCT) in each group. Patients with C8/T1 level radiculopathy showed significantly longer RMCTs than did the control group (p = 0.001)\n\nRoot motor conduction time (RMCT) in each group. Patients with C8/T1 level radiculopathy showed significantly longer RMCTs than did the control group (p = 0.001)\n\nTable 3Logistic regression models using data from patients and matched control subjectsOdds ratio (95% CI)p-valueModel 1aRMCT (per 0.1 ms)1.11 (1.04–1.18)0.002Model 2bRMCT (per 0.1 ms)1.16 (1.06–1.27)0.002Model 3cRMCT (per 0.1 ms)1.15 (1.06–1.27)0.011Abbreviations: CI, confidence interval; RMCT, root motor conduction timeaUnivariable analysis; badjusted for stimulated peripheral motor conduction time; cadjusted for stimulated peripheral motor conduction time, amplitude of median compound motor nerve action potential, and F-wave latency\n\nLogistic regression models using data from patients and matched control subjects\nAbbreviations: CI, confidence interval; RMCT, root motor conduction time\naUnivariable analysis; badjusted for stimulated peripheral motor conduction time; cadjusted for stimulated peripheral motor conduction time, amplitude of median compound motor nerve action potential, and F-wave latency", "Table 1 summarizes the baseline characteristics of the 37 patients, 11 of whom had bilateral C8/T1 radiculopathy. The mean patient age was 61.1 years and the mean height was 166.5 cm; 75.7% were men. The patients’ median NDI score was 12.0. Among all 48 C8/T1 root levels, 25 (52.1%) were right-sided, and the median duration of symptoms was 4.0 (3.0–12.0) months. The median NRS scores of the neck and arm were 3.0 and 4.5, respectively.\n\nTable 1Baseline features of patients with C8/T1 level radiculopathyVariablesValueaTotal patients, n37Age, years61.1 ± 12.2Male, n (%)28 (75.7)Height, cm166.5 ± 8.7Hypertension, n (%)16 (43.2)Diabetes, n (%)5 (13.5)Dyslipidemia, n (%)6 (16.2)NRS, neck3.0 (2.0–4.5)Total C8/T1 root levels, n48Right side, n (%)25 (52.1)Symptom duration, months4.0 (3.0–12.0)NDI12.0 (5.5–19.0)NRS, arm4.5 (3.0–6.0)Abbreviations: NDI, neck disability index; NRS, numerical rating scaleaUnless otherwise indicated, values are means ± standard deviations or medians (interquartile ranges)\n\nBaseline features of patients with C8/T1 level radiculopathy\nAbbreviations: NDI, neck disability index; NRS, numerical rating scale\naUnless otherwise indicated, values are means ± standard deviations or medians (interquartile ranges)\nAfter performing PSM for comparative analysis, we extracted 48 control C8/T1 root levels and confirmed that there were no significant differences in age, sex, or height between the two groups (Table 2).\n\nTable 2Comparison of data from patients with C8/T1 level radiculopathy and matched control subjectsPatients (n = 48)Controls (n = 48)p-valueAge, years61.9 ± 12.458.8 ± 11.10.197Male, n (%)37 (77.1)28 (58.3)0.081Height, cm166.4 ± 8.6164.2 ± 8.80.223APB-RMCT, ms1.7 ± 0.61.2 ± 0.80.001Abbreviations: APB, abductor pollicis brevis; RMCT, root motor conduction time\n\nComparison of data from patients with C8/T1 level radiculopathy and matched control subjects\nAbbreviations: APB, abductor pollicis brevis; RMCT, root motor conduction time", "The mean RMCT among patients with radiculopathy was 1.7 ± 0.6 ms, which was significantly longer than that among control subjects (1.2 ± 0.8 ms; p = 0.001) (Table 2; Fig. 3). Logistic regression models revealed that the delay in RMCT observed in patients with C8/T1 was independently associated with radiculopathy (per Model 3: odds ratio, 1.15; 95% confidence interval, 1.06–1.27; p = 0.011) (Table 3). The area under the Receiver Operating Characteristic curve for diagnosing C8/T1 radiculopathy using RMCT was 0.72 (0.61–0.82) with 0.83 sensitivity and 0.58 specificity (Supplementary Fig. 1). In terms of subjective symptom indices, the RMCT was significantly associated with symptom duration (coefficient = 0.58; p < 0.001) but not with the NRS of the arm (coefficient = 0.22; p = 0.139) All measured values for calculating the RMCT are presented in Supplementary Table 2.\n\nFig. 3Root motor conduction time (RMCT) in each group. Patients with C8/T1 level radiculopathy showed significantly longer RMCTs than did the control group (p = 0.001)\n\nRoot motor conduction time (RMCT) in each group. Patients with C8/T1 level radiculopathy showed significantly longer RMCTs than did the control group (p = 0.001)\n\nTable 3Logistic regression models using data from patients and matched control subjectsOdds ratio (95% CI)p-valueModel 1aRMCT (per 0.1 ms)1.11 (1.04–1.18)0.002Model 2bRMCT (per 0.1 ms)1.16 (1.06–1.27)0.002Model 3cRMCT (per 0.1 ms)1.15 (1.06–1.27)0.011Abbreviations: CI, confidence interval; RMCT, root motor conduction timeaUnivariable analysis; badjusted for stimulated peripheral motor conduction time; cadjusted for stimulated peripheral motor conduction time, amplitude of median compound motor nerve action potential, and F-wave latency\n\nLogistic regression models using data from patients and matched control subjects\nAbbreviations: CI, confidence interval; RMCT, root motor conduction time\naUnivariable analysis; badjusted for stimulated peripheral motor conduction time; cadjusted for stimulated peripheral motor conduction time, amplitude of median compound motor nerve action potential, and F-wave latency", "We observed a significant delay in RMCT among patients with C8/T1 radiculopathy when compared to control subjects, and also demonstrated that this delay was independently associated with the presence of radiculopathy at the C8/T1 level. This finding is notable because it provided a rational basis for the applicability of RMCT in terms of diagnosing radiculopathy at a specific spinal root level.\nThe application of RMCT to assess radiculopathy occurring at particular levels is rare. Most relevant studies were conducted before the 2000s, with only a few published since then. Banerjee et al. [13] described magnetic spinal stimulation as a non-invasive method to evaluate lumbosacral motor radiculopathy in 26 patients and 25 control subjects; they mainly targeted the lower lumbosacral nerve roots (L5, S1, and S2) by recording from the abductor hallucis muscle. According to their results, patients with clinical motor weakness had profoundly prolonged RMCTs; therefore, they suggested that RMCT delay was related to the severity of symptoms. Golez [22] measured motor conduction time via stimulation at the L1 and S1 levels in 25 patients with lumbar spinal stenosis and 36 control subjects. That study was noteworthy because it demonstrated that neurogenic claudication increased the cauda equina motor conduction time, revealing that the clinical symptom and delay of motor conduction time were related. In our study, the RMCT showed a significant correlation with the duration of subjective symptoms, which supports Golez’s results to some extent. However, in our study, the subjective pain level in the lesional-side limb was not related to RMCT as we assumed when we primarily evaluated the motor nerve. Additionally, Seçil et al. [23] measured the cauda equina motor conduction time and showed that it was slower in the lumbar spinal stenosis group than in the control group; however, their study did not identify the relationship between the degree of motor conduction delay and symptom severity.\nOur study is the first to measure RMCT in patients with cervical radiculopathy and has the advantage of being specific to a specific spinal level; moreover, we included a relatively large number of subjects compared to previous related studies. Additionally, variables such as age, sex, and height were analyzed and adjusted for using PSM, rendering the results much more reliable. Since the APB-RMCT was previously found to be affected by height in a linear model [18], for the integrity of our results, it was important that we controlled for this factor when assessing the patient and control groups.\nRMCT has also been studied in patients with demyelinating neuropathy. In a previous study, a significant difference was found in the RMCT of 30 healthy subjects and 12 patients with Guillain-Barre syndrome who were diagnosed within one week [14]; hence, RMCT was deemed to be useful for the early diagnosis of focal segmental demyelinating polyneuropathy, affording this measure a novel clinical utility. In the study of Inaba et al. [24], RMCT was measured in 11 patients with chronic inflammatory demyelinating neuropathy and 10 with Guillain-Barre syndrome, wherein the authors found that the RMCT increasingly normalized as muscle strength recovered. Taken together, such studies illustrate the applicability of the RMCT to various diseases involving the motor root segment. Furthermore, measuring the RMCT is a non-invasive procedure and has the potential to be applied to additional fields given its ability to detect diseases in their early stages and to also assess functional recovery. Therefore, additional research on the use of the RMCT is warranted going forward.\nThis study had several limitations. It was retrospective in nature, requiring additional investigations to validate our results. Additionally, our participants were limited to Koreans; hence, validation among patients of other ethnicities is also required. Our patients comprised a single group diagnosed with C8/T1 radiculopathy that was relatively heterogeneous; future studies that analyze various subgroups of patients (such as those with acute versus chronic lesions or mild versus severe symptoms), would further clarify the relationship between RMCT and radiculopathy. Finally, there were potential limitations in terms of measurement. For example, it is difficult to apply our methods at some spinal root levels because reliable recording of the F-wave is possible only from distal muscles [25]. Moreover, it was challenging to measure the RMCT when reliable motor evoked potentials or compound muscle action potentials could not be obtained owing to severe peripheral nerve lesions in the distal limbs.", "We demonstrated that the RMCT is delayed in the root lesions of patients with C8/T1 level radiculopathy. This noninvasive method (compared to EMG) can have an adjuvant role in diagnosing radiculopathy at certain spinal levels. Our study may be a good milestone for future clinical applications of RMCT as related to patients with radiculopathy. However, it is important to validate our results through additional multi-center, multi-ethnic studies.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ "introduction", null, null, null, null, "results", null, null, "discussion", "conclusion", "supplementary-material", null ]
[ "Radiculopathy", "Electrodiagnosis", "Motor-evoked potential", "Nerve conduction" ]
Overwork among resident physicians: national questionnaire survey results.
36266644
Residents experience the longest working hours among physicians. Thus, it would be beneficial to perform a nationwide survey in Japan on residents' long work hours and the background factors promoting upper limits on working hours of Japanese residents. The aim of this study was to study or assess the state of physicians' excessive work hours and its background factors using a questionnaire survey.
BACKGROUND
The survey was sent to 924 hospitals. The physicians' general attributes, work hours and conditions, and employers' foundational entities were explored. Multiple logistic regression analysis was performed to elucidate the background factors for long work hours.
METHODS
Of the 4306 resident physicians who responded, 67% had ≥ 60 in-hospital hours/week and 27% had ≥ 80 h/week; 51% were on-call ≥ four times/month. Many of them hoped for increased remuneration. Additionally, female (reference: male, OR: 0.65, 95% CI: 0.55-0.76), 35-40 years old (reference: 25-30 years old, OR: 1.83, 95% CI: 1.32-2.54), childlessness (reference: child, OR: 1.41, 95% CI: 1.12-1.75), surgical specialization (reference: internal medicine, OR: 2.51, 95% CI: 1.96-3.23), neurosurgical specialization (reference: internal medicine, OR: 4.38, 95% CI: 2.92-6.59) and hospitals with 200-400 physicians (reference: <100 physicians, OR: 1.82, 95% CI: 1.12-2.96) exhibited significant correlations with ≥ 80 in-hospital hours/week.
RESULTS
Understanding the factors that increase the likelihood of residents working very long hours could aid in making targeted changes to address the specific concerns. Moreover, reducing working hours to a reasonable limit can improve resident physicians' health and the quality of care they provide in their community.
CONCLUSION
[ "Adult", "Female", "Humans", "Male", "Internal Medicine", "Internship and Residency", "Japan", "Physicians", "Surveys and Questionnaires", "Workload" ]
9584270
null
null
null
null
Results
Regarding the respondents’ attributes shown in Tables 1 and 37% were female, 60% were between 30 − 39 years of age, 44% were married, and 21% had children. Additionally, 23% specialized in general internal medicine. The average annual income ranged from ≥ 4 to < 6 million yen. Regarding employer attributes, 54% and 38% worked in large urban areas and national university hospitals, respectively. The number of beds was between 600 and 800, and there were ≥ 600 physicians in most hospitals. Moreover, 70% of the physicians had a part-time job. Table 1Respondent attributes and in-hospital hoursAttributes of Respondentsn%Annual Income (not including part-time work; dollars)n (%)Sex< 16,0001383.2Male271062.9≥ 16,000–<32,000101423.5Female159637.1≥ 32,000–<48,000108325.2Age (years)≥ 48,000–<64,00087920.4≥ 25–<30258760.1≥ 64,000–<80,00067315.6≥ 30–<35127029.5≥ 80,000–<96,0003438.0≥ 35–<402205.1≥ 96,0001764.1≥ 402295.3In-hospital (hours per week)Marital Status< 40 h781.8Married190444.240–60 h130530.3Not married240255.860–80 h178341.4Parental Status80–100 h78318.2Children90921.1≥ 100 h3578.3No children339778.9Foundational Entity of EmployerBasic Area of SpecializationPublic122428.4General Internal Medicine97922.7National University161837.6Surgery43710.1Private University94521.9Orthopedic Surgery3297.6Private51912.1Pediatrics2976.9Employer’s Total No. of BedsOB/GYN2746.4< 200631.5Anesthesiology2726.3≥ 200–<4003207.4Otolaryngology2275.3≥ 400–<60067715.7Psychiatry2124.9≥ 600–<800141232.8Dermatology1874.3≥ 800–<1,000107224.9Ophthalmology1523.5≥ 1,00076217.7Emergency Medicine1463.4Employer’s No. of Full-time PhysiciansUrology1453.4< 1004219.8Radiology1393.2≥ 100–<20076817.8Neurosurgery1252.9≥ 200–<30059213.7Pathology1182.7≥ 300–<40073317.0Plastic Surgery1052.4≥ 400–<50047611.1General Practice892.1≥ 6001,31630.6Rehabilitation Medicine591.4Employer’s Regional ClassificationClinical Testing140.3Urban231553.8Part-time WorkIntermediate191444.4Yes299169.5Rural771.8No131530.5 Respondent attributes and in-hospital hours As for the weekly in-hospital time, 32%, 41%, and 27% of physicians spent < 60, 60–80 h, and greater than 80 h in the hospital per week, respectively. Table 2 shows the results of multiple logistic regression analysis with in-hospital time of ≥ 80 h per week as the explanatory variable in order to elucidate the background factors for long work hours of residents. Table 2Multiple logistic regression analysis with residents’ long work hours as the explanatory variableOdds Ratio95% Confidence IntervalP ValueSexMaleReferenceFemale0.650.55–0.76< 0.01Age (years)≥ 25–<30Reference≥ 30–<351.241.05–1.460.01≥ 35–<401.831.32–2.54< 0.01≥ 400.770.51–1.150.20Marital StatusMarriedReferenceNot married1.110.93–1.320.23Parental StatusChildrenReferenceNo children1.411.12–1.75< 0.01Basic Area of SpecializationGeneral Internal MedicineReferenceSurgery2.511.96–3.23< 0.01Orthopedic Surgery1.290.97–1.740.08Pediatrics1.320.98–1.780.07OB/GYN1.360.99–1.870.06Anesthesiology0.460.31–0.69< 0.01Otolaryngology0.690.47–1.000.05Psychiatry0.570.38–0.85< 0.01Dermatology0.690.45–1.050.08Ophthalmology0.610.39–0.960.03Emergency Medicine1.110.74–1.660.62Urology1.370.93–2.030.11Radiology0.280.15–0.51< 0.01Neurosurgery4.382.92–6.59< 0.01Pathology0.460.26–0.81< 0.01Plastic Surgery1.220.77–1.940.40General Practice0.810.45–1.470.49Rehabilitation Medicine0.500.23–1.080.08Clinical Testing0.240.03–1.950.18Annual Income (dollars)< 16,000Reference≥ 16,000–<32,0000.840.54–1.300.43≥ 32,000–<48,0000.970.62–1.520.90≥ 48,000–<64,0000.790.50–1.250.31≥ 64,000–<80,0000.960.60–1.530.86≥ 80,000–<96,0000.710.42–1.170.18≥ 96,0000.760.42–1.360.36Foundational Entity of EmployerPublicReferenceNational University0.850.62–1.160.30Private University0.960.66–1.400.84Private1.000.76–1.320.99Employer’s Total No. of Beds< 200Reference≥ 200–<4001.450.65–3.210.36≥ 400–<6001.330.58–3.040.50≥ 600–<8001.400.59–3.310.45≥ 800–<1,0001.370.57–3.320.48≥ 1,0001.430.59–3.510.43Employer’s No. of Full-time Physicians< 100Reference≥ 100–<2001.250.86–1.820.25≥ 200–<3001.641.05–2.550.03≥ 300–<4001.821.12–2.960.02≥ 400–<5001.560.93–2.620.10≥ 6001.410.85–2.340.18Employer’s Regional ClassificationUrbanReferenceIntermediate1.000.85–1.180.98Rural0.820.41–1.620.56 Multiple logistic regression analysis with residents’ long work hours as the explanatory variable Sex, age (≥ 30–<40 years), no children, surgical/neurosurgical specialization, and number of physicians (≥ 200–<400) exhibited statistically significant correlation as background factors for physicians being in-hospital for ≥ 80 h per week. However, there were no statistically significant correlations with marital status, income, foundational entity of employer, number of beds, and regional characteristics.
Conclusion
Our national survey shows that many residents work very long hours (more than 80 h/week), with some specialists working even longer hours, while others are required to work part-time jobs in order to be financially stable. Moreover, our results show a variety of background factors that correlate to working very long hours, such as being male, being between 30 and 40 years old, having no children, being in the surgical or neurosurgical specialty, and the number of physicians in the hospital. By understanding the factors that increase the likelihood of residents working very long hours, targeted changes can be made to address the specific concerns. Reducing working hours to a reasonable level can improve the resident physicians’ health and the quality of care they provide in their community.
[ "Background", "Methods", "Survey participants", "Investigation items", "Statistical analysis", "Ethical considerations" ]
[ "The work hours of Japanese workers are indicated to be longer compared to workers in other countries [1]. The long working hours of physicians, approximately more than 60 h a week, account for 42% of full-time physicians working more than 200 days per year, compared to a 14% average for all other professions [2]. Among physicians, residents (physicians undergoing training in order to become specialists) have the longest working hours [3].\nDepression-related suicides and deaths due to ischemic heart disease and cerebrovascular disease caused by overwork are called “karoshi” (death due to overwork) and have become a public health issue specific to East Asia, such as China [4]. Moreover, there is ample evidence regarding the adverse effects of long work hours on health, both in Japan and overseas. According to a systematic review by Bannai et al. [5], long working hours are correlated with depression, anxiety, sleep, and coronary artery disease.\nIn 2003, the US Accreditation Council for Graduate Medical Education (ACGME) limited residents’ work hours to less than 80 h per week due to its adverse effects on their health and the increased risk of medical error [6, 7]. A systematic review that evaluated this measure concluded that the work-hour restrictions of the ACGME are correlated with an improvement in emotional malaise and burnout syndrome symptoms [8].\nMoreover, in Japan, several studies performed at single facilities have indicated the depressive tendencies and burnout of interns and residents [9, 10]. However, unlike those in the US, there have been no restrictions on working hours. Additionally, some physicians not being paid [11] and long working hours [3] have been indicated as issues in the working environment of Japanese residents. However, no nationwide survey studies have been conducted in Japan on resident physicians.\nIn 2016, the Ministry of Health, Labour and Welfare instituted the “Study Meeting on Physician Labor Reform,” which has been examining measures to promote labor reforms for physicians, and it has disclosed a policy wherein from April 2024, the upper limit on annual overtime work hours for residents will be 1860 h (at par with the US ACGME) [12].\nTo administratively promote the setting of upper limits on working hours for Japanese residents, it will be useful to perform a nationwide survey on the condition of long work hours for residents and its background factors, and examine topics relevant to the promotion of physician labor reforms.\nTherefore, a nationwide questionnaire survey was performed on Japanese residents who work particularly long hours, and it discussed the overworked condition of the resident physicians and background factors associated with its effects to suggest policies for the promotion of labor reforms.", "[SUBTITLE] Survey participants [SUBSECTION] Accounting for the 19 basic areas stipulated by the Japanese Medical Specialty Board, the survey was sent to 924 core hospitals nationwide that had received training program approval for each of the basic areas. Effective responses were received from 4306 residents at 416 hospitals (response rate = 49%). The response rate is related to the % of hospitals from which the responses were received. The number of residents and the location and specialty are unknown. The survey was conducted for 14 days, from 10 October to 23 October 2020.\nAccounting for the 19 basic areas stipulated by the Japanese Medical Specialty Board, the survey was sent to 924 core hospitals nationwide that had received training program approval for each of the basic areas. Effective responses were received from 4306 residents at 416 hospitals (response rate = 49%). The response rate is related to the % of hospitals from which the responses were received. The number of residents and the location and specialty are unknown. The survey was conducted for 14 days, from 10 October to 23 October 2020.\n[SUBTITLE] Investigation items [SUBSECTION] A web survey was conducted using a questionnaire. First, the respondents’ attributes (sex, age, basic area of specialization, marital/parental status, income (1 dollar = 125 yen), and foundational entity of employer/no. of beds/no. of full-time doctors/regional classifications) were described. There were four types of foundational entities of the employers: public (except for national universities); national universities; private universities; and private (except for private universities). The regional classifications were divided into Group 1 (urban areas), Group 2 (intermediate areas), and Group 3 (rural areas) based on the combination of the population size and density as of 2019 regarding the 344 secondary care zones nationwide [13].\nNext, with regard to the employment status of the residents, the number of hours spent in the hospital, the number of times on duty, etc. were described.\nA web survey was conducted using a questionnaire. First, the respondents’ attributes (sex, age, basic area of specialization, marital/parental status, income (1 dollar = 125 yen), and foundational entity of employer/no. of beds/no. of full-time doctors/regional classifications) were described. There were four types of foundational entities of the employers: public (except for national universities); national universities; private universities; and private (except for private universities). The regional classifications were divided into Group 1 (urban areas), Group 2 (intermediate areas), and Group 3 (rural areas) based on the combination of the population size and density as of 2019 regarding the 344 secondary care zones nationwide [13].\nNext, with regard to the employment status of the residents, the number of hours spent in the hospital, the number of times on duty, etc. were described.\n[SUBTITLE] Statistical analysis [SUBSECTION] To elucidate the background factors for residents’ long work hours, multiple logistic regression analyses was performed with ≥ 80 in-hospital hours per week as the explanatory variable and sex, age, basic field of specialization, marital/parental status, income, and foundational entity of employer/no. of beds/no. of full-time doctors/regional classifications as response variables. In the statistical analysis, a p value of < 0.05 was considered statistically significant. STATA 15.1 was used in all analyses.\nTo elucidate the background factors for residents’ long work hours, multiple logistic regression analyses was performed with ≥ 80 in-hospital hours per week as the explanatory variable and sex, age, basic field of specialization, marital/parental status, income, and foundational entity of employer/no. of beds/no. of full-time doctors/regional classifications as response variables. In the statistical analysis, a p value of < 0.05 was considered statistically significant. STATA 15.1 was used in all analyses.\n[SUBTITLE] Ethical considerations [SUBSECTION] This study was conducted after receiving approval from the medical ethics board of the University of Tsukuba Faculty of Medicine (no. 1498). All methods were performed in accordance with the relevant guidelines and regulations of the Declaration of Helsinki. On the first page of the questionnaire, the purpose of the study and measures for safe data management were explained. Additionally, participants in this study were informed that their participation was entirely voluntary. Data were collected from participants who provided informed consent to participate. The results of the survey were analyzed separately in order to ensure the anonymization and confidentiality of personal information.\nThis study was conducted after receiving approval from the medical ethics board of the University of Tsukuba Faculty of Medicine (no. 1498). All methods were performed in accordance with the relevant guidelines and regulations of the Declaration of Helsinki. On the first page of the questionnaire, the purpose of the study and measures for safe data management were explained. Additionally, participants in this study were informed that their participation was entirely voluntary. Data were collected from participants who provided informed consent to participate. The results of the survey were analyzed separately in order to ensure the anonymization and confidentiality of personal information.", "Accounting for the 19 basic areas stipulated by the Japanese Medical Specialty Board, the survey was sent to 924 core hospitals nationwide that had received training program approval for each of the basic areas. Effective responses were received from 4306 residents at 416 hospitals (response rate = 49%). The response rate is related to the % of hospitals from which the responses were received. The number of residents and the location and specialty are unknown. The survey was conducted for 14 days, from 10 October to 23 October 2020.", "A web survey was conducted using a questionnaire. First, the respondents’ attributes (sex, age, basic area of specialization, marital/parental status, income (1 dollar = 125 yen), and foundational entity of employer/no. of beds/no. of full-time doctors/regional classifications) were described. There were four types of foundational entities of the employers: public (except for national universities); national universities; private universities; and private (except for private universities). The regional classifications were divided into Group 1 (urban areas), Group 2 (intermediate areas), and Group 3 (rural areas) based on the combination of the population size and density as of 2019 regarding the 344 secondary care zones nationwide [13].\nNext, with regard to the employment status of the residents, the number of hours spent in the hospital, the number of times on duty, etc. were described.", "To elucidate the background factors for residents’ long work hours, multiple logistic regression analyses was performed with ≥ 80 in-hospital hours per week as the explanatory variable and sex, age, basic field of specialization, marital/parental status, income, and foundational entity of employer/no. of beds/no. of full-time doctors/regional classifications as response variables. In the statistical analysis, a p value of < 0.05 was considered statistically significant. STATA 15.1 was used in all analyses.", "This study was conducted after receiving approval from the medical ethics board of the University of Tsukuba Faculty of Medicine (no. 1498). All methods were performed in accordance with the relevant guidelines and regulations of the Declaration of Helsinki. On the first page of the questionnaire, the purpose of the study and measures for safe data management were explained. Additionally, participants in this study were informed that their participation was entirely voluntary. Data were collected from participants who provided informed consent to participate. The results of the survey were analyzed separately in order to ensure the anonymization and confidentiality of personal information." ]
[ null, null, null, null, null, null ]
[ "Background", "Methods", "Survey participants", "Investigation items", "Statistical analysis", "Ethical considerations", "Results", "Discussion", "Conclusion" ]
[ "The work hours of Japanese workers are indicated to be longer compared to workers in other countries [1]. The long working hours of physicians, approximately more than 60 h a week, account for 42% of full-time physicians working more than 200 days per year, compared to a 14% average for all other professions [2]. Among physicians, residents (physicians undergoing training in order to become specialists) have the longest working hours [3].\nDepression-related suicides and deaths due to ischemic heart disease and cerebrovascular disease caused by overwork are called “karoshi” (death due to overwork) and have become a public health issue specific to East Asia, such as China [4]. Moreover, there is ample evidence regarding the adverse effects of long work hours on health, both in Japan and overseas. According to a systematic review by Bannai et al. [5], long working hours are correlated with depression, anxiety, sleep, and coronary artery disease.\nIn 2003, the US Accreditation Council for Graduate Medical Education (ACGME) limited residents’ work hours to less than 80 h per week due to its adverse effects on their health and the increased risk of medical error [6, 7]. A systematic review that evaluated this measure concluded that the work-hour restrictions of the ACGME are correlated with an improvement in emotional malaise and burnout syndrome symptoms [8].\nMoreover, in Japan, several studies performed at single facilities have indicated the depressive tendencies and burnout of interns and residents [9, 10]. However, unlike those in the US, there have been no restrictions on working hours. Additionally, some physicians not being paid [11] and long working hours [3] have been indicated as issues in the working environment of Japanese residents. However, no nationwide survey studies have been conducted in Japan on resident physicians.\nIn 2016, the Ministry of Health, Labour and Welfare instituted the “Study Meeting on Physician Labor Reform,” which has been examining measures to promote labor reforms for physicians, and it has disclosed a policy wherein from April 2024, the upper limit on annual overtime work hours for residents will be 1860 h (at par with the US ACGME) [12].\nTo administratively promote the setting of upper limits on working hours for Japanese residents, it will be useful to perform a nationwide survey on the condition of long work hours for residents and its background factors, and examine topics relevant to the promotion of physician labor reforms.\nTherefore, a nationwide questionnaire survey was performed on Japanese residents who work particularly long hours, and it discussed the overworked condition of the resident physicians and background factors associated with its effects to suggest policies for the promotion of labor reforms.", "[SUBTITLE] Survey participants [SUBSECTION] Accounting for the 19 basic areas stipulated by the Japanese Medical Specialty Board, the survey was sent to 924 core hospitals nationwide that had received training program approval for each of the basic areas. Effective responses were received from 4306 residents at 416 hospitals (response rate = 49%). The response rate is related to the % of hospitals from which the responses were received. The number of residents and the location and specialty are unknown. The survey was conducted for 14 days, from 10 October to 23 October 2020.\nAccounting for the 19 basic areas stipulated by the Japanese Medical Specialty Board, the survey was sent to 924 core hospitals nationwide that had received training program approval for each of the basic areas. Effective responses were received from 4306 residents at 416 hospitals (response rate = 49%). The response rate is related to the % of hospitals from which the responses were received. The number of residents and the location and specialty are unknown. The survey was conducted for 14 days, from 10 October to 23 October 2020.\n[SUBTITLE] Investigation items [SUBSECTION] A web survey was conducted using a questionnaire. First, the respondents’ attributes (sex, age, basic area of specialization, marital/parental status, income (1 dollar = 125 yen), and foundational entity of employer/no. of beds/no. of full-time doctors/regional classifications) were described. There were four types of foundational entities of the employers: public (except for national universities); national universities; private universities; and private (except for private universities). The regional classifications were divided into Group 1 (urban areas), Group 2 (intermediate areas), and Group 3 (rural areas) based on the combination of the population size and density as of 2019 regarding the 344 secondary care zones nationwide [13].\nNext, with regard to the employment status of the residents, the number of hours spent in the hospital, the number of times on duty, etc. were described.\nA web survey was conducted using a questionnaire. First, the respondents’ attributes (sex, age, basic area of specialization, marital/parental status, income (1 dollar = 125 yen), and foundational entity of employer/no. of beds/no. of full-time doctors/regional classifications) were described. There were four types of foundational entities of the employers: public (except for national universities); national universities; private universities; and private (except for private universities). The regional classifications were divided into Group 1 (urban areas), Group 2 (intermediate areas), and Group 3 (rural areas) based on the combination of the population size and density as of 2019 regarding the 344 secondary care zones nationwide [13].\nNext, with regard to the employment status of the residents, the number of hours spent in the hospital, the number of times on duty, etc. were described.\n[SUBTITLE] Statistical analysis [SUBSECTION] To elucidate the background factors for residents’ long work hours, multiple logistic regression analyses was performed with ≥ 80 in-hospital hours per week as the explanatory variable and sex, age, basic field of specialization, marital/parental status, income, and foundational entity of employer/no. of beds/no. of full-time doctors/regional classifications as response variables. In the statistical analysis, a p value of < 0.05 was considered statistically significant. STATA 15.1 was used in all analyses.\nTo elucidate the background factors for residents’ long work hours, multiple logistic regression analyses was performed with ≥ 80 in-hospital hours per week as the explanatory variable and sex, age, basic field of specialization, marital/parental status, income, and foundational entity of employer/no. of beds/no. of full-time doctors/regional classifications as response variables. In the statistical analysis, a p value of < 0.05 was considered statistically significant. STATA 15.1 was used in all analyses.\n[SUBTITLE] Ethical considerations [SUBSECTION] This study was conducted after receiving approval from the medical ethics board of the University of Tsukuba Faculty of Medicine (no. 1498). All methods were performed in accordance with the relevant guidelines and regulations of the Declaration of Helsinki. On the first page of the questionnaire, the purpose of the study and measures for safe data management were explained. Additionally, participants in this study were informed that their participation was entirely voluntary. Data were collected from participants who provided informed consent to participate. The results of the survey were analyzed separately in order to ensure the anonymization and confidentiality of personal information.\nThis study was conducted after receiving approval from the medical ethics board of the University of Tsukuba Faculty of Medicine (no. 1498). All methods were performed in accordance with the relevant guidelines and regulations of the Declaration of Helsinki. On the first page of the questionnaire, the purpose of the study and measures for safe data management were explained. Additionally, participants in this study were informed that their participation was entirely voluntary. Data were collected from participants who provided informed consent to participate. The results of the survey were analyzed separately in order to ensure the anonymization and confidentiality of personal information.", "Accounting for the 19 basic areas stipulated by the Japanese Medical Specialty Board, the survey was sent to 924 core hospitals nationwide that had received training program approval for each of the basic areas. Effective responses were received from 4306 residents at 416 hospitals (response rate = 49%). The response rate is related to the % of hospitals from which the responses were received. The number of residents and the location and specialty are unknown. The survey was conducted for 14 days, from 10 October to 23 October 2020.", "A web survey was conducted using a questionnaire. First, the respondents’ attributes (sex, age, basic area of specialization, marital/parental status, income (1 dollar = 125 yen), and foundational entity of employer/no. of beds/no. of full-time doctors/regional classifications) were described. There were four types of foundational entities of the employers: public (except for national universities); national universities; private universities; and private (except for private universities). The regional classifications were divided into Group 1 (urban areas), Group 2 (intermediate areas), and Group 3 (rural areas) based on the combination of the population size and density as of 2019 regarding the 344 secondary care zones nationwide [13].\nNext, with regard to the employment status of the residents, the number of hours spent in the hospital, the number of times on duty, etc. were described.", "To elucidate the background factors for residents’ long work hours, multiple logistic regression analyses was performed with ≥ 80 in-hospital hours per week as the explanatory variable and sex, age, basic field of specialization, marital/parental status, income, and foundational entity of employer/no. of beds/no. of full-time doctors/regional classifications as response variables. In the statistical analysis, a p value of < 0.05 was considered statistically significant. STATA 15.1 was used in all analyses.", "This study was conducted after receiving approval from the medical ethics board of the University of Tsukuba Faculty of Medicine (no. 1498). All methods were performed in accordance with the relevant guidelines and regulations of the Declaration of Helsinki. On the first page of the questionnaire, the purpose of the study and measures for safe data management were explained. Additionally, participants in this study were informed that their participation was entirely voluntary. Data were collected from participants who provided informed consent to participate. The results of the survey were analyzed separately in order to ensure the anonymization and confidentiality of personal information.", "Regarding the respondents’ attributes shown in Tables 1 and 37% were female, 60% were between 30 − 39 years of age, 44% were married, and 21% had children. Additionally, 23% specialized in general internal medicine. The average annual income ranged from ≥ 4 to < 6 million yen. Regarding employer attributes, 54% and 38% worked in large urban areas and national university hospitals, respectively. The number of beds was between 600 and 800, and there were ≥ 600 physicians in most hospitals. Moreover, 70% of the physicians had a part-time job.\n\nTable 1Respondent attributes and in-hospital hoursAttributes of Respondentsn%Annual Income (not including part-time work; dollars)n (%)Sex< 16,0001383.2Male271062.9≥ 16,000–<32,000101423.5Female159637.1≥ 32,000–<48,000108325.2Age (years)≥ 48,000–<64,00087920.4≥ 25–<30258760.1≥ 64,000–<80,00067315.6≥ 30–<35127029.5≥ 80,000–<96,0003438.0≥ 35–<402205.1≥ 96,0001764.1≥ 402295.3In-hospital (hours per week)Marital Status< 40 h781.8Married190444.240–60 h130530.3Not married240255.860–80 h178341.4Parental Status80–100 h78318.2Children90921.1≥ 100 h3578.3No children339778.9Foundational Entity of EmployerBasic Area of SpecializationPublic122428.4General Internal Medicine97922.7National University161837.6Surgery43710.1Private University94521.9Orthopedic Surgery3297.6Private51912.1Pediatrics2976.9Employer’s Total No. of BedsOB/GYN2746.4< 200631.5Anesthesiology2726.3≥ 200–<4003207.4Otolaryngology2275.3≥ 400–<60067715.7Psychiatry2124.9≥ 600–<800141232.8Dermatology1874.3≥ 800–<1,000107224.9Ophthalmology1523.5≥ 1,00076217.7Emergency Medicine1463.4Employer’s No. of Full-time PhysiciansUrology1453.4< 1004219.8Radiology1393.2≥ 100–<20076817.8Neurosurgery1252.9≥ 200–<30059213.7Pathology1182.7≥ 300–<40073317.0Plastic Surgery1052.4≥ 400–<50047611.1General Practice892.1≥ 6001,31630.6Rehabilitation Medicine591.4Employer’s Regional ClassificationClinical Testing140.3Urban231553.8Part-time WorkIntermediate191444.4Yes299169.5Rural771.8No131530.5\n\nRespondent attributes and in-hospital hours\nAs for the weekly in-hospital time, 32%, 41%, and 27% of physicians spent < 60, 60–80 h, and greater than 80 h in the hospital per week, respectively.\nTable 2 shows the results of multiple logistic regression analysis with in-hospital time of ≥ 80 h per week as the explanatory variable in order to elucidate the background factors for long work hours of residents.\n\nTable 2Multiple logistic regression analysis with residents’ long work hours as the explanatory variableOdds Ratio95% Confidence IntervalP ValueSexMaleReferenceFemale0.650.55–0.76< 0.01Age (years)≥ 25–<30Reference≥ 30–<351.241.05–1.460.01≥ 35–<401.831.32–2.54< 0.01≥ 400.770.51–1.150.20Marital StatusMarriedReferenceNot married1.110.93–1.320.23Parental StatusChildrenReferenceNo children1.411.12–1.75< 0.01Basic Area of SpecializationGeneral Internal MedicineReferenceSurgery2.511.96–3.23< 0.01Orthopedic Surgery1.290.97–1.740.08Pediatrics1.320.98–1.780.07OB/GYN1.360.99–1.870.06Anesthesiology0.460.31–0.69< 0.01Otolaryngology0.690.47–1.000.05Psychiatry0.570.38–0.85< 0.01Dermatology0.690.45–1.050.08Ophthalmology0.610.39–0.960.03Emergency Medicine1.110.74–1.660.62Urology1.370.93–2.030.11Radiology0.280.15–0.51< 0.01Neurosurgery4.382.92–6.59< 0.01Pathology0.460.26–0.81< 0.01Plastic Surgery1.220.77–1.940.40General Practice0.810.45–1.470.49Rehabilitation Medicine0.500.23–1.080.08Clinical Testing0.240.03–1.950.18Annual Income (dollars)< 16,000Reference≥ 16,000–<32,0000.840.54–1.300.43≥ 32,000–<48,0000.970.62–1.520.90≥ 48,000–<64,0000.790.50–1.250.31≥ 64,000–<80,0000.960.60–1.530.86≥ 80,000–<96,0000.710.42–1.170.18≥ 96,0000.760.42–1.360.36Foundational Entity of EmployerPublicReferenceNational University0.850.62–1.160.30Private University0.960.66–1.400.84Private1.000.76–1.320.99Employer’s Total No. of Beds< 200Reference≥ 200–<4001.450.65–3.210.36≥ 400–<6001.330.58–3.040.50≥ 600–<8001.400.59–3.310.45≥ 800–<1,0001.370.57–3.320.48≥ 1,0001.430.59–3.510.43Employer’s No. of Full-time Physicians< 100Reference≥ 100–<2001.250.86–1.820.25≥ 200–<3001.641.05–2.550.03≥ 300–<4001.821.12–2.960.02≥ 400–<5001.560.93–2.620.10≥ 6001.410.85–2.340.18Employer’s Regional ClassificationUrbanReferenceIntermediate1.000.85–1.180.98Rural0.820.41–1.620.56\n\nMultiple logistic regression analysis with residents’ long work hours as the explanatory variable\nSex, age (≥ 30–<40 years), no children, surgical/neurosurgical specialization, and number of physicians (≥ 200–<400) exhibited statistically significant correlation as background factors for physicians being in-hospital for ≥ 80 h per week. However, there were no statistically significant correlations with marital status, income, foundational entity of employer, number of beds, and regional characteristics.", "First, 68% and 27% of the responding physicians exceeded 60 and 80 h of in-hospital time per week, respectively, elucidating the actual state of overwork for Japanese resident physicians. An in-hospital time of 60 h per week converts to four hours of overtime per day for an 8-hours/day 40-hour week. This is the legal work hour stipulated by the Labor Standards Act and generates over 80 h of monthly overtime work.\nLong work hours that continue for several months are called the “death by overwork level” because of the strong correlation with mental disorders and the onset of circulatory organ diseases, and it is a criterion for the recognition of labor accidents [14, 15]. This study’s results suggest that 68% of the respondents work in an environment exceeding the “death by overwork level.”\nThe Japanese Ministry of Health, Labour and Welfare decided to implement “Physician Labor Reform” in April 2024; the principle annual upper limit of overtime work for physicians shall be 960 h, 1860 h in specific cases, and various initiatives to achieve this goal are underway [16]. The plan promotes shifting tasks to other hospital staff and sets a continuous work time limit (28 h) with 9-hour intervals between work shifts and an upper limit for overtime hours [12].\nFor the 1860-hour cases, the average weekly in-hospital time was estimated to be < 80 h. This study’s results suggested that 27% of respondents worked hours that exceeded this criterion. Because such long working hours will become illegal in April 2024, measures must be taken to shorten working hours as soon as possible. However, the background factors for such super-long work hours have not been identified.\nLong work hours of ≥ 80 in-hospital hours per week exhibited a significant correlation with males and no children, whereas no correlation with marital status was observed. The presence of children may require shorter work hours due to the time spent on childcare. Another possibility is that long work hours are not inhibited by the time invested in married life. Regarding sex, a report on American female surgery residents suggests that they have a higher proportion of long work hours and higher rates of experiencing burnout [16]. However, in this study, males had a stronger correlation with > 80 h of work per week. This is affected by the higher proportion of Japanese males in specialized areas that have long work hours, such as surgery and neurosurgery [17]. In this study, when controlling for specialty on multivariate analyses, male gender was still associated with long work hours.\nRegarding age, the odds ratio was higher at ≥ 35–<40 years of age; 5% of participants belonged to this age group. These individuals may have become physicians later, or become residents again after changing their specialization, but the correlation with excessively long work hours is unclear. Further studies, such as additional surveys and interviews, will be necessary to explain why these physicians often work excessively long hours and identify other background factors.\nDifferences in long work hours were based on specialization areas. For example, surgeons often have long work hours. The Japan Surgical Society has stated that promoting initiatives such as drastic task shifting and consolidation of surgeries are necessary to shorten overtime hours. However, deeper discussions on self-improvement are necessary to ensure that reducing work hours does not impair surgeons’ acquisition of knowledge and skills [18]. In several countries, discussions are occurring regarding restricting work hours and whether policies that promote uniform work hour restrictions, regardless of specialization, are not appropriate [19].\nIn Japan, each physician can select their specialization both in medical school and after graduation [20]. Therefore, it is difficult to eliminate the uneven distribution of physician specialties. Physicians may also transfer from their selected, busy specialty to a less busy one [21]. According to the Japanese Ministry of Health, Labour and Welfare, the lowest proportion of physicians is in surgery and neurosurgery, where this study’s results showed significantly longer working hours. In contrast, there is a surplus in ophthalmology and dermatology, where long work hours are significantly scarce.\nNo significant correlation was observed between ≥ 80 in-hospital hours per week and the residents’ income. As stated previously, the annual pay of residents is low, and some physicians are even unpaid [11]. Therefore, because physicians’ long work hours are not necessarily compensated by greater pay, annual income may not be regarded as a background factor.\nThe regulations for residents’ work hours in the “Labor Reforms for Physicians” being advanced by the Japanese Ministry of Health, Labour and Welfare are based on those implemented by the US ACGME [22]. These regulations comprise 80 working hours per week, a continuous working time limit of 16 h, and a 10-hour interval between working hours. Contrastingly, the Japanese reforms comprise an upper limit of 1860 h on annual overtime, a 28-hour limit on continuous working time, and a 9-hour interval between working hours.\nIn the US, various evidence-based reviews have been performed on the ACGME regulations on residents’ work hours. Comparative reports regarding residents in surgery and internal medicine, using an intervention group without the continuous work time and shift interval restrictions and a control group using the ACGME regulated restrictions, indicated no significant difference in patient outcomes or residents’ degrees of satisfaction [23, 24]. The two studies cited were large. However, they had significant limitations, such as power to detect changes in mortality and outcomes metrics indirectly linked to study populations of interest, and the findings generally ran counter to the broader literature on the effects of efforts to reduce work hours on patient safety.\nAnother study in 2005 shows extended-duration work shifts (over 24 h), which were sanctioned by ACGME, pose safety hazards for interns [25]. In 2011, ACGME instituted a continuous working time limit of 16 h for residents. A recent study shows the 2011 ACGME work-hour limit was associated with meaningful improvements in physician safety and health [26]. Implementing such studies and examining evidence-based systemic reviews from the perspectives of patient safety, physician health, physician training, and so forth, is necessary in Japan.\nJapanese resident physicians tend to have a low annual income. In this study, 52% of respondents had an annual income of less than 48,000 dollars (not including part-time work), and 70% were working part-time. Part-time workers are assumed to need to engage in part-time work at medical institutions other than their regular employer to be financially stable or to repay loans. Additionally, some doctors work at university hospitals as “unpaid doctors” [11]. Therefore, the need to improve residents’ remuneration can also be regarded as an issue.\nIf part-time work is treated as one of the causes of residents’ overwork, then the reduction of part-time work would be one method of decreasing residents’ work hours by promoting labor reforms for physicians. However, this may further reduce residents’ incomes. Therefore, there should also be proposals for pay supplementation using the national budget.\nAdditionally, medical institutions that have maintained their treatment systems so far through the part-time employment of residents must remember that treatment systems may become unsustainable with the termination of part-time work. Moreover, consideration should be given at the policy level so that local medical care is not affected, particularly in regions with a shortage of doctors.\nThis study has several limitations. First is the possibility of selection bias due to the individuals’ voluntary participation. Despite this limitation, effective responses were obtained from 4306 participants at 416 hospitals out of the 924 core hospitals nationwide accredited by the Japanese Medical Specialty Board (response rate = 49%). The response rate is related to the % of hospitals from which the responses were received. The number of residents and the location and specialty are unknown. Moreover, both the response rate and the number of respondents ensured a sample that was representative of the population.\nSecond, the survey used self-report responses, which may have introduced various information biases. For example, misreporting was possible because the in-hospital hours per week were not a detailed time study filled out by another person. Therefore, it couldn’t be confirmed whether the in-hospital hours were all work hours. Additionally, because participation in this survey was voluntary, detailed definitions of the specialist jargon used were not provided in order to increase the response rate.\nThird, a statistical correlation was observed between long work hours and background factors. However, the causal relationship was unclear. It is possible that probable confounding factors were not measured in this study. Additionally, the effects of the COVID-19 pandemic were not examined. As shown in previous studies, the COVID-19 pandemic has greatly affected how physicians work [27], and it would be useful to conduct the same survey after the COVID-19 pandemic and compare the results of the studies. Furthermore, because there have been no previous similar nationwide studies on Japanese residents, it is difficult to compare the results of this study with the situation of residents before the COVID-19 pandemic.\nThe harsh working conditions of resident physicians in Japan and the background factors of longer work hours have been elucidated in tandem with discussions of solutions. To ensure the health of resident physicians, the quality of training, and the safety of patients, it is necessary to ascertain the status of resident physicians in greater detail, including vigorously promoting and verifying the effects of labor reforms for physicians.", "Our national survey shows that many residents work very long hours (more than 80 h/week), with some specialists working even longer hours, while others are required to work part-time jobs in order to be financially stable. Moreover, our results show a variety of background factors that correlate to working very long hours, such as being male, being between 30 and 40 years old, having no children, being in the surgical or neurosurgical specialty, and the number of physicians in the hospital. By understanding the factors that increase the likelihood of residents working very long hours, targeted changes can be made to address the specific concerns. Reducing working hours to a reasonable level can improve the resident physicians’ health and the quality of care they provide in their community." ]
[ null, null, null, null, null, null, "results", "discussion", "conclusion" ]
[ "Physicians", "Burnout", "Questionnaire survey", "Japan", "Work hours" ]
The value of combining the simple anthropometric obesity parameters, Body Mass Index (BMI) and a Body Shape Index (ABSI), to assess the risk of non-alcoholic fatty liver disease.
36266655
Body mass index (BMI) and A Body Shape Index (ABSI) are current independent risk factors for non-alcoholic fatty liver disease (NAFLD). The aim of this study was to explore the value of combining these two most common obesity indexes in identifying NAFLD.
BACKGROUND
The subjects in this study were 14,251 individuals from the NAfld in the Gifu Area, Longitudinal Analysis (NAGALA) cohort who underwent routine health examination. We integrated BMI with WC and with ABSI to construct 6 combined obesity indicators-obesity phenotypes, the combined anthropometric risk index (ARI) for BMI and ABSI, optimal proportional combination OBMI+WC and OBMI+ABSI, and multiplicative combination BMI*WC and BMI*ABSI. Several multivariable logistic regression models were established to evaluate the relationship between BMI, WC, ABSI, and the above six combined indicators and NAFLD; receiver operating characteristic (ROC) curves were drawn to compare the ability of each obesity indicator to identify NAFLD.
METHODS
A total of 2,507 (17.59%) subjects were diagnosed with NAFLD. BMI, WC, ABSI, and all other combined obesity indicators were significantly and positively associated with NAFLD in the current study, with BMI*WC having the strongest correlation with NAFLD in female subjects (OR per SD increase: 3.13) and BMI*ABSI having the strongest correlation in male subjects (OR per SD increase: 2.97). ROC analysis showed that ARI and OBMI+ABSI had the best diagnostic performance in both sexes, followed by BMI*WC (area under the curve: female 0.8912; male 0.8270). After further age stratification, it was found that ARI and multiplicative indicators (BMI*WC, BMI*ABSI) and optimal proportional combination indicators (OBMI+WC, OBMI+ABSI) significantly improved the NAFLD risk identification ability of the basic anthropometric parameters in middle-aged females and young and middle-aged males.
RESULTS
In the general population, BMI combined with ABSI best identified obesity-related NAFLD risk and was significantly better than BMI or WC, or ABSI. We find that ARI and the multiplicative combined indicators BMI*WC and BMI*ABSI further improved risk prediction and may be proposed for possible use in clinical practice.
CONCLUSION
[ "Humans", "Male", "Female", "Body Mass Index", "Non-alcoholic Fatty Liver Disease", "Waist Circumference", "Anthropometry", "Obesity", "Obesity, Morbid", "Risk Factors" ]
9585710
null
null
null
null
Results
[SUBTITLE] Demographic and clinical characteristics of subjects [SUBSECTION] The study included 14,251 subjects, of whom 7,411 (52%) were males, with a mean age of 43.82 (8.99) years, and 2,029 (27.37%) were diagnosed with NAFLD; 6,840 were (48%) females, with mean age 43.22 (8.78) years, and 478 (6.99%) were diagnosed with NAFLD. Table 1 is stratified by gender and describes the differences in baseline characteristics between NAFLD patients and non-NAFLD subjects. We found that NAFLD patients consistently had higher age, weight, TC, SBP, DBP, HbA1c, BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, ALT, AST, GGT, FPG, and TG levels in both sexes, but with lower HDL-C levels. Notably, non-NAFLD subjects of both sexes tended to have greater alcohol consumption, which paradoxically appeared to be a protective factor for NAFLD. Additionally, the comparison between the NAFLD group and the non-NAFLD group showed that physical exercise habits were only significantly different in males, height was only significantly different in females, and smoking habits were not significantly different in both sexes. Table 1Characteristics of 14,251 subjects stratified by NAFLD and genderMaleFemaleVariablesNon-NAFLDn = 5382 (72.61%)NAFLDn = 2029 (27.39%)P-valueNon-NAFLDn = 6362 (93.01%)NAFLDn = 478 (6.99%)P-valueAge, years43.7 (9.3)44.1 (8.2)0.00242.9 (8.7)47.6 (8.3)< 0.001Height, cm170.9 (6.0)170.6 (5.9)0.084158.4 (5.4)157.0 (5.3)< 0.001Weight, kg64.6 (8.3)74.3 (10.6)< 0.00151.9 (7.1)63.2 (10.0)< 0.001TC, mmol/L5.1 (0.8)5.4 (0.9)< 0.0015.1 (0.9)5.6 (0.9)< 0.001HDL-C, mmol/L1.3 (0.3)1.1 (0.3)< 0.0011.7 (0.4)1.4 (0.3)< 0.001SBP, mmHg116.0 (13.2)124.0 (14.5)< 0.001108.4 (13.8)120.7 (16.0)< 0.001FPG, mmol/L5.3 (0.4)5.4 (0.3)< 0.0015.0 (0.4)5.3 (0.4)< 0.001DBP, mmHg72.9 (9.3)78.4 (10.1)< 0.00167.0 (9.5)75.1 (10.2)< 0.001HbA1c, %5.1 (0.3)5.3 (0.3)< 0.0015.2 (0.3)5.4 (0.3)< 0.001BMI, kg/m222.1 (2.4)25.5 (3.0)< 0.00120.7 (2.6)25.6 (3.6)< 0.001WC, cm78.0 (6.8)86.6 (7.4)< 0.00170.8 (7.3)83.3 (8.9)< 0.001ABSI75.8 (3.3)76.7 (3.1)< 0.00174.8 (4.6)76.8 (4.5)< 0.001ARI13.1 (0.9)14.3 (1.0)< 0.00112.2 (1.0)14.0 (1.3)< 0.001OBMI+WC34.4 (3.2)38.9 (3.8)< 0.00131.7 (3.4)38.3 (4.5)< 0.001OBMI+ABSI32.0 (2.1)34.9 (2.4)< 0.00128.9 (2.3)33.4 (3.0)< 0.001BMI*WC1714.2 (1507.7-1939.1)2152.9 (1919.2-2457.9)< 0.0011423.8 (1249.1-1645.8)2070.2 (1795.3-2396.7)< 0.001BMI*ABSI1669.3 (1538.1–1806.0)1922.5 (1795.9-2082.3)< 0.0011518.4 (1390.8-1673.5)1917.8 (1756.7-2131.4)< 0.001ALT, U/L18.0 (14.0–23.0)29.0 (22.0–41.0)< 0.00113.0 (11.0–17.0)19.0 (15.0–26.0)< 0.001AST, U/L17.0 (14.0–21.0)21.0 (17.0–26.0)< 0.00116.0 (13.0–19.0)18.0 (15.0–22.0)< 0.001GGT, U/L17.0 (14.0–24.0)24.0 (18.0–35.0)< 0.00112.0 (9.0–14.0)15.0 (12.0–20.0)< 0.001TG, mmol/L0.8 (0.6–1.2)1.3 (0.9–1.9)< 0.0010.5 (0.4–0.8)1.0 (0.7–1.4)< 0.001Habit of exercise, No. (%)< 0.0010.335No4300 (79.9%)1720 (84.8%)5351 (84.1%)410 (85.8%)Yes1082 (20.1%)309 (15.2%)1011 (15.9%)68 (14.2%)Drinking status, No. (%)< 0.0010.004Non or small3731 (69.3%)1623 (80.0%)5986 (94.1%)465 (97.3%)Light1096 (20.4%)273 (13.5%)376 (5.9%)13 (2.7%)Moderate555 (10.3%)133 (6.6%)Smoking status, No. (%)0.0670.664Non1952 (36.3%)758 (37.4%)5609 (88.2%)427 (89.3%)Past1538 (28.6%)615 (30.3%)382 (6.0%)24 (5.0%)Current1892 (35.2%)656 (32.3%)371 (5.8%)27 (5.6%)Values were expressed as mean (standard deviation) or medians (quartile interval) or n (%). Abbreviations: BMI: body mass index; WC: waist circumference; ALT: alanine aminotransferase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FPG: fasting plasma glucose; HbA1c: hemoglobin A1c; SBP: systolic blood pressure; DBP: diastolic blood pressure; NAFLD: non-alcoholic fatty liver disease; ABSI: A body shape index; OBMI+ABSI: Optimal proportional combination of BMI and ABSI; OBMI+WC: Optimal proportional combination of BMI and WC; ARI: Anthropometric risk index Characteristics of 14,251 subjects stratified by NAFLD and gender Values were expressed as mean (standard deviation) or medians (quartile interval) or n (%). Abbreviations: BMI: body mass index; WC: waist circumference; ALT: alanine aminotransferase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FPG: fasting plasma glucose; HbA1c: hemoglobin A1c; SBP: systolic blood pressure; DBP: diastolic blood pressure; NAFLD: non-alcoholic fatty liver disease; ABSI: A body shape index; OBMI+ABSI: Optimal proportional combination of BMI and ABSI; OBMI+WC: Optimal proportional combination of BMI and WC; ARI: Anthropometric risk index Baseline characteristics for all subjects according to the quartile category of BMI*ABSI are shown in Table 2. We found that with the increase of BMI*ABSI quartiles, all baseline metrics of subjects showed a trend of change (all P < 0.001). Of these, a greater proportion of male subjects were in the higher BMI*ABSI quartiles, with a male-to-female ratio of almost 7:3 in Q4, and there were more subjects with smoking and drinking habits and with less physical exercise in Q4. Moreover, subjects’ HDL-C levels decreased with increasing BMI*ABSI quartiles, while age, BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*WC, height, weight, TC, SBP, HbA1c, ALT, AST, DBP, FPG, GGT, TG levels, and the prevalence of NAFLD increased (Q4: 48.5%). Table 2Characteristics of 14,251 subjects stratified according to the BMI*ABSI quartilesBMI*ABSI quartilesvariablesQ1 (961.1-1477.2)Q2 (1477.4-1645.5)Q3 (1645.6-1827.4)Q4 (1827.5-3630.9)P-valueNo. of subjects3563356235633563Male, No. (%)867 (24.3%)1666 (46.8%)2269 (63.7%)2609 (73.2%)Age, years41.0 (8.7)43.1 (8.7)44.5 (8.6)45.6 (8.9)< 0.001BMI, kg/m218.7 (1.3)20.9 (1.2)22.7 (1.2)26.0 (2.5)< 0.001WC, cm65.4 (3.7)72.8 (2.9)78.8 (2.9)87.7 (5.8)< 0.001ABSI73.1 (3.8)75.1 (3.8)76.5 (3.5)77.5 (3.5)< 0.001ARI11.4 (0.4)12.4 (0.3)13.2 (0.3)14.4 (0.8)< 0.001OBMI+WC29.0 (1.5)32.3 (1.0)35.0 (1.1)39.6 (3.0)< 0.001OBMI+ABSI27.4 (1.3)29.9 (1.1)31.9 (1.1)35.0 (2.0)< 0.001BMI*WC1241.2 (1147.1-1318.4)1514.2 (1447.2-1587.4)1782.6 (1703.2-1865.8)2194.4 (2051.7-2436.9)< 0.001Height, cm161.7 (7.7)164.3 (8.3)166.1 (8.5)167.1 (8.4)< 0.001Weight, kg49.0 (5.7)56.4 (6.5)62.7 (7.3)72.9 (10.3)< 0.001TC, mmol/L4.9 (0.8)5.0 (0.9)5.2 (0.9)5.4 (0.9)< 0.001HDL-C, mmol/L1.7 (0.39)1.5 (0.4)1.4 (0.4)1.2 (0.3)< 0.001SBP, mmHg105.6 (12.5)111.1 (12.8)115.9 (13.3)123.1 (14.8)< 0.001FPG, mmol/L4.9 (0.4)5.1 (0.4)5.2 (0.4)5.3 (0.4)< 0.001DBP, mmHg65.5 (8.5)69.0 (9.1)72.5 (9.6)77.6 (10.2)< 0.001HbA1c, %5.1 (0.3)5.1 (0.3)5.2 (0.3)5.3 (0.3)< 0.001ALT, U/L14.0 (11.0–17.0)15.0 (12.0–19.0)18.0 (13.0–23.0)23.0 (17.0–33.0)< 0.001AST, U/L16.0 (13.0–19.0)16.00 (14.0–20.0)17.0 (14.0–21.0)19.0 (16.0–24.0)< 0.001GGT, U/L12.0 (10.0–15.0)13.00 (11.0–18.0)16.0 (12.0–22.0)21.0 (15.0–30.0)< 0.001TG, mmol/L0.51 (0.4–0.7)0.6 (0.5–0.9)0.8 (0.6–1.2)1.1 (0.8–1.6)< 0.001Habit of exercise, No. (%)< 0.001No2963 (83.2%)2858 (80.2%)2920 (82.0%)3040 (85.3%)Yes600 (16.8%)704 (19.8%)643 (18.1%)523 (14.7%)Drinking status, No. (%)< 0.001Non or small3228 (90.6%)2937 (82.5%)2848 (79.9%)2792 (78.4%)Light279 (7.8%)467 (13.1%)507 (14.2%)505 (14.2%)Moderate56 (1.6%)158 (4.4%)208 (5.8%)266 (7.5%)Smoking status, No. (%)< 0.001Non2736 (76.8%)2322 (65.2%)1957 (54.9%)1731 (48.6%)Past337 (9.5%)560 (15.7%)785 (22.0%)877 (24.6%)Current490 (13.8%)680 (19.1%)821 (23.0%)955 (26.8%)NAFLD, No. (%)16 (0.5%)150 (4.2%)611 (17.2%)1730 (48.5%)< 0.001Abbreviations as in Table 1 Characteristics of 14,251 subjects stratified according to the BMI*ABSI quartiles Abbreviations as in Table 1 The study included 14,251 subjects, of whom 7,411 (52%) were males, with a mean age of 43.82 (8.99) years, and 2,029 (27.37%) were diagnosed with NAFLD; 6,840 were (48%) females, with mean age 43.22 (8.78) years, and 478 (6.99%) were diagnosed with NAFLD. Table 1 is stratified by gender and describes the differences in baseline characteristics between NAFLD patients and non-NAFLD subjects. We found that NAFLD patients consistently had higher age, weight, TC, SBP, DBP, HbA1c, BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, ALT, AST, GGT, FPG, and TG levels in both sexes, but with lower HDL-C levels. Notably, non-NAFLD subjects of both sexes tended to have greater alcohol consumption, which paradoxically appeared to be a protective factor for NAFLD. Additionally, the comparison between the NAFLD group and the non-NAFLD group showed that physical exercise habits were only significantly different in males, height was only significantly different in females, and smoking habits were not significantly different in both sexes. Table 1Characteristics of 14,251 subjects stratified by NAFLD and genderMaleFemaleVariablesNon-NAFLDn = 5382 (72.61%)NAFLDn = 2029 (27.39%)P-valueNon-NAFLDn = 6362 (93.01%)NAFLDn = 478 (6.99%)P-valueAge, years43.7 (9.3)44.1 (8.2)0.00242.9 (8.7)47.6 (8.3)< 0.001Height, cm170.9 (6.0)170.6 (5.9)0.084158.4 (5.4)157.0 (5.3)< 0.001Weight, kg64.6 (8.3)74.3 (10.6)< 0.00151.9 (7.1)63.2 (10.0)< 0.001TC, mmol/L5.1 (0.8)5.4 (0.9)< 0.0015.1 (0.9)5.6 (0.9)< 0.001HDL-C, mmol/L1.3 (0.3)1.1 (0.3)< 0.0011.7 (0.4)1.4 (0.3)< 0.001SBP, mmHg116.0 (13.2)124.0 (14.5)< 0.001108.4 (13.8)120.7 (16.0)< 0.001FPG, mmol/L5.3 (0.4)5.4 (0.3)< 0.0015.0 (0.4)5.3 (0.4)< 0.001DBP, mmHg72.9 (9.3)78.4 (10.1)< 0.00167.0 (9.5)75.1 (10.2)< 0.001HbA1c, %5.1 (0.3)5.3 (0.3)< 0.0015.2 (0.3)5.4 (0.3)< 0.001BMI, kg/m222.1 (2.4)25.5 (3.0)< 0.00120.7 (2.6)25.6 (3.6)< 0.001WC, cm78.0 (6.8)86.6 (7.4)< 0.00170.8 (7.3)83.3 (8.9)< 0.001ABSI75.8 (3.3)76.7 (3.1)< 0.00174.8 (4.6)76.8 (4.5)< 0.001ARI13.1 (0.9)14.3 (1.0)< 0.00112.2 (1.0)14.0 (1.3)< 0.001OBMI+WC34.4 (3.2)38.9 (3.8)< 0.00131.7 (3.4)38.3 (4.5)< 0.001OBMI+ABSI32.0 (2.1)34.9 (2.4)< 0.00128.9 (2.3)33.4 (3.0)< 0.001BMI*WC1714.2 (1507.7-1939.1)2152.9 (1919.2-2457.9)< 0.0011423.8 (1249.1-1645.8)2070.2 (1795.3-2396.7)< 0.001BMI*ABSI1669.3 (1538.1–1806.0)1922.5 (1795.9-2082.3)< 0.0011518.4 (1390.8-1673.5)1917.8 (1756.7-2131.4)< 0.001ALT, U/L18.0 (14.0–23.0)29.0 (22.0–41.0)< 0.00113.0 (11.0–17.0)19.0 (15.0–26.0)< 0.001AST, U/L17.0 (14.0–21.0)21.0 (17.0–26.0)< 0.00116.0 (13.0–19.0)18.0 (15.0–22.0)< 0.001GGT, U/L17.0 (14.0–24.0)24.0 (18.0–35.0)< 0.00112.0 (9.0–14.0)15.0 (12.0–20.0)< 0.001TG, mmol/L0.8 (0.6–1.2)1.3 (0.9–1.9)< 0.0010.5 (0.4–0.8)1.0 (0.7–1.4)< 0.001Habit of exercise, No. (%)< 0.0010.335No4300 (79.9%)1720 (84.8%)5351 (84.1%)410 (85.8%)Yes1082 (20.1%)309 (15.2%)1011 (15.9%)68 (14.2%)Drinking status, No. (%)< 0.0010.004Non or small3731 (69.3%)1623 (80.0%)5986 (94.1%)465 (97.3%)Light1096 (20.4%)273 (13.5%)376 (5.9%)13 (2.7%)Moderate555 (10.3%)133 (6.6%)Smoking status, No. (%)0.0670.664Non1952 (36.3%)758 (37.4%)5609 (88.2%)427 (89.3%)Past1538 (28.6%)615 (30.3%)382 (6.0%)24 (5.0%)Current1892 (35.2%)656 (32.3%)371 (5.8%)27 (5.6%)Values were expressed as mean (standard deviation) or medians (quartile interval) or n (%). Abbreviations: BMI: body mass index; WC: waist circumference; ALT: alanine aminotransferase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FPG: fasting plasma glucose; HbA1c: hemoglobin A1c; SBP: systolic blood pressure; DBP: diastolic blood pressure; NAFLD: non-alcoholic fatty liver disease; ABSI: A body shape index; OBMI+ABSI: Optimal proportional combination of BMI and ABSI; OBMI+WC: Optimal proportional combination of BMI and WC; ARI: Anthropometric risk index Characteristics of 14,251 subjects stratified by NAFLD and gender Values were expressed as mean (standard deviation) or medians (quartile interval) or n (%). Abbreviations: BMI: body mass index; WC: waist circumference; ALT: alanine aminotransferase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FPG: fasting plasma glucose; HbA1c: hemoglobin A1c; SBP: systolic blood pressure; DBP: diastolic blood pressure; NAFLD: non-alcoholic fatty liver disease; ABSI: A body shape index; OBMI+ABSI: Optimal proportional combination of BMI and ABSI; OBMI+WC: Optimal proportional combination of BMI and WC; ARI: Anthropometric risk index Baseline characteristics for all subjects according to the quartile category of BMI*ABSI are shown in Table 2. We found that with the increase of BMI*ABSI quartiles, all baseline metrics of subjects showed a trend of change (all P < 0.001). Of these, a greater proportion of male subjects were in the higher BMI*ABSI quartiles, with a male-to-female ratio of almost 7:3 in Q4, and there were more subjects with smoking and drinking habits and with less physical exercise in Q4. Moreover, subjects’ HDL-C levels decreased with increasing BMI*ABSI quartiles, while age, BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*WC, height, weight, TC, SBP, HbA1c, ALT, AST, DBP, FPG, GGT, TG levels, and the prevalence of NAFLD increased (Q4: 48.5%). Table 2Characteristics of 14,251 subjects stratified according to the BMI*ABSI quartilesBMI*ABSI quartilesvariablesQ1 (961.1-1477.2)Q2 (1477.4-1645.5)Q3 (1645.6-1827.4)Q4 (1827.5-3630.9)P-valueNo. of subjects3563356235633563Male, No. (%)867 (24.3%)1666 (46.8%)2269 (63.7%)2609 (73.2%)Age, years41.0 (8.7)43.1 (8.7)44.5 (8.6)45.6 (8.9)< 0.001BMI, kg/m218.7 (1.3)20.9 (1.2)22.7 (1.2)26.0 (2.5)< 0.001WC, cm65.4 (3.7)72.8 (2.9)78.8 (2.9)87.7 (5.8)< 0.001ABSI73.1 (3.8)75.1 (3.8)76.5 (3.5)77.5 (3.5)< 0.001ARI11.4 (0.4)12.4 (0.3)13.2 (0.3)14.4 (0.8)< 0.001OBMI+WC29.0 (1.5)32.3 (1.0)35.0 (1.1)39.6 (3.0)< 0.001OBMI+ABSI27.4 (1.3)29.9 (1.1)31.9 (1.1)35.0 (2.0)< 0.001BMI*WC1241.2 (1147.1-1318.4)1514.2 (1447.2-1587.4)1782.6 (1703.2-1865.8)2194.4 (2051.7-2436.9)< 0.001Height, cm161.7 (7.7)164.3 (8.3)166.1 (8.5)167.1 (8.4)< 0.001Weight, kg49.0 (5.7)56.4 (6.5)62.7 (7.3)72.9 (10.3)< 0.001TC, mmol/L4.9 (0.8)5.0 (0.9)5.2 (0.9)5.4 (0.9)< 0.001HDL-C, mmol/L1.7 (0.39)1.5 (0.4)1.4 (0.4)1.2 (0.3)< 0.001SBP, mmHg105.6 (12.5)111.1 (12.8)115.9 (13.3)123.1 (14.8)< 0.001FPG, mmol/L4.9 (0.4)5.1 (0.4)5.2 (0.4)5.3 (0.4)< 0.001DBP, mmHg65.5 (8.5)69.0 (9.1)72.5 (9.6)77.6 (10.2)< 0.001HbA1c, %5.1 (0.3)5.1 (0.3)5.2 (0.3)5.3 (0.3)< 0.001ALT, U/L14.0 (11.0–17.0)15.0 (12.0–19.0)18.0 (13.0–23.0)23.0 (17.0–33.0)< 0.001AST, U/L16.0 (13.0–19.0)16.00 (14.0–20.0)17.0 (14.0–21.0)19.0 (16.0–24.0)< 0.001GGT, U/L12.0 (10.0–15.0)13.00 (11.0–18.0)16.0 (12.0–22.0)21.0 (15.0–30.0)< 0.001TG, mmol/L0.51 (0.4–0.7)0.6 (0.5–0.9)0.8 (0.6–1.2)1.1 (0.8–1.6)< 0.001Habit of exercise, No. (%)< 0.001No2963 (83.2%)2858 (80.2%)2920 (82.0%)3040 (85.3%)Yes600 (16.8%)704 (19.8%)643 (18.1%)523 (14.7%)Drinking status, No. (%)< 0.001Non or small3228 (90.6%)2937 (82.5%)2848 (79.9%)2792 (78.4%)Light279 (7.8%)467 (13.1%)507 (14.2%)505 (14.2%)Moderate56 (1.6%)158 (4.4%)208 (5.8%)266 (7.5%)Smoking status, No. (%)< 0.001Non2736 (76.8%)2322 (65.2%)1957 (54.9%)1731 (48.6%)Past337 (9.5%)560 (15.7%)785 (22.0%)877 (24.6%)Current490 (13.8%)680 (19.1%)821 (23.0%)955 (26.8%)NAFLD, No. (%)16 (0.5%)150 (4.2%)611 (17.2%)1730 (48.5%)< 0.001Abbreviations as in Table 1 Characteristics of 14,251 subjects stratified according to the BMI*ABSI quartiles Abbreviations as in Table 1 [SUBTITLE] Calculation of the optimal proportional combination of BMI with WC and ABSI [SUBSECTION] In the logistic regression equation constructed to calculate the optimal proportional combination OBMI+WC, the regression coefficient βBMI for BMI was 0.218 (0.154–0.282), and βWC for WC was 0.063 (0.038–0.089) in females; in males, βBMI was 0.177 (0.124–0.299), and βWC was 0.068 (0.048–0.089). Therefore, according to the calculation formula of the optimal proportional coefficients, the values ​​of nBMI and nWC in females were 0.776 and 0.224, respectively, and the values ​​of nBMI and nWC in males were 0.722 and 0.278, respectively. Similarly, in the logistic regression equation constructed to calculate the optimal proportional combination OBMI+ABSI, βBMI and βABSI were 0.356 and 0.064, respectively, in females, resulting in nBMI and nABSI values ​​of 0.848 and 0.152, respectively; in males, βBMI and βABSI were 0.334 and 0.075, respectively, and nBMI and nABSI values ​​were 0.817 and 0.183, respectively. Table 3 shows the calculation formulas of OBMI+WC and OBMI+ABSI. Table 3Optimal combination equations of BMI with WC and ABSI.Logistic regression derived equationsMaleFemaleequations for BMI and WC0.722BMI + 0.278WC0.776BMI + 0.224WCequations for BMI and ABSI0.817BMI + 0.183ABSI0.848BMI + 0.152ABSIAbbreviations as in Table 1 Optimal combination equations of BMI with WC and ABSI. Abbreviations as in Table 1 In the logistic regression equation constructed to calculate the optimal proportional combination OBMI+WC, the regression coefficient βBMI for BMI was 0.218 (0.154–0.282), and βWC for WC was 0.063 (0.038–0.089) in females; in males, βBMI was 0.177 (0.124–0.299), and βWC was 0.068 (0.048–0.089). Therefore, according to the calculation formula of the optimal proportional coefficients, the values ​​of nBMI and nWC in females were 0.776 and 0.224, respectively, and the values ​​of nBMI and nWC in males were 0.722 and 0.278, respectively. Similarly, in the logistic regression equation constructed to calculate the optimal proportional combination OBMI+ABSI, βBMI and βABSI were 0.356 and 0.064, respectively, in females, resulting in nBMI and nABSI values ​​of 0.848 and 0.152, respectively; in males, βBMI and βABSI were 0.334 and 0.075, respectively, and nBMI and nABSI values ​​were 0.817 and 0.183, respectively. Table 3 shows the calculation formulas of OBMI+WC and OBMI+ABSI. Table 3Optimal combination equations of BMI with WC and ABSI.Logistic regression derived equationsMaleFemaleequations for BMI and WC0.722BMI + 0.278WC0.776BMI + 0.224WCequations for BMI and ABSI0.817BMI + 0.183ABSI0.848BMI + 0.152ABSIAbbreviations as in Table 1 Optimal combination equations of BMI with WC and ABSI. Abbreviations as in Table 1 [SUBTITLE] Association of various obesity indicators with NAFLD [SUBSECTION] Correlation analysis of the basic anthropometric measures showed a strong correlation between BMI and WC in both sexes (r: male 0.8796; female 0.8191), while BMI was barely correlated with ABSI (r < 0.05) (Supplementary Table 2). In contrast, the combination of ABSI and BMI may be more appropriate. Table 4 presents the associations between BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC and the risk of NAFLD in both sexes. In multivariate-adjusted logistic regression models, BMI and WC, and ABSI were positively correlated with NAFLD risk. In the current study, the combined indicators ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC were positively correlated with NAFLD risk in all four logistic regression models, and the OR values ​​in the univariate logistic regression model ranged from 1.44 to 5.02; after further adjustment for age, height, GGT, TC, AST, HDL-C, drinking status, ALT, TG, FPG, HbA1c, and SBP (Model 3), the results did not change significantly. The risk of NAFLD increased by 33% (OR 1.33, 95%CI 1.29, 1.37) and 52% (OR 1.52, 95%CI 1.45, 1.60) and 172% (OR 2.72, 95%CI 2.43, 3.04), in females, for each unit increase in OBMI+WC and OBMI+ABSI and ARI respectively. Similarly, in males, NAFLD risk was increased by 30% (OR 1.30, 95%CI 1.27, 1.33) and 50% (OR 1.50, 95%CI 1.45, 1.56) and 172% (OR 2.72, 95%CI 2.48, 2.98), respectively. Each standard deviation increase in BMI*WC and BMI*ABSI in females was associated with a 213% (OR 3.13, 95%CI 2.74,3,56) and 209% (OR 3.09, 95%CI 2.72, 3.52) increased risk for NAFLD respectively, and for males 185% (OR 2.85, 95%CI 2.59, 3.15) and 197% (OR 2.97, 95%CI 2.68, 3.28), respectively. Furthermore, the association between BMI*ABSI and NAFLD remained unchanged after treating BMI*ABSI as a categorical variable; taking the first quartile as the control group, the NAFLD risk increased with the increase of BMI*ABSI quartiles (all P for trend < 0.001). Moreover, we also analyzed the risk of NAFLD for each obesity phenotype in multivariable logistic regression models (Supplementary Table 3), which in model 3 showed that compared with the BMIN/WCN phenotype, male and female BMIO/ WCN, BMIN/WCO, and BMIO/WCO phenotypes all had a significantly higher risk of NAFLD. The risk of NAFLD associated with the BMIO/ WCN, BMIN/WCO, and BMIO/WCO phenotypes had ORs of 3.67, 3.77, and 9.78 in females and 2.76, 2.74, and 4.19 in males, respectively. Clearly, subjects with the BMIO/WCO phenotype consistently had the highest risk of developing NAFLD. Table 4Odds ratios and 95% confidence intervals of BMI, WC, ABSI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and ARI for NAFLD in female and male subjectsOdds ratios (95% confidence interval)Crude modelModel 1Model 2Model 3FemaleBMI1.58 (1.53, 1.64)*1.58 (1.52, 1.63)*1.49 (1.44, 1.55)*1.42 (1.36, 1.47)*WC1.19 (1.17, 1.21)*1.19 (1.17, 1.21)*1.17 (1.15, 1.18)*1.14 (1.12, 1.16)*ABSI1.09 (1.07, 1.12)*1.07 (1.05, 1.09)*1.06 (1.04, 1.08)*1.05 (1.02, 1.07)*ARI3.70 (3.36, 4.07)*3.61 (3.28, 3.98)*3.11 (2.80, 3.44)*2.72 (2.43, 3.04)*OBMI+ABSI1.73 (1.66, 1.80)*1.71 (1.65, 1.79)*1.61 (1.54, 1.68)*1.52 (1.45, 1.60)*OBMI+WC1.44 (1.41, 1.48)*1.44 (1.40, 1.48)*1.38 (1.34, 1.42)*1.33 (1.29, 1.37)*BMI*ABSI (Per SD)4.49 (4.02, 5.02)*4.33 (3.87, 4.84)*3.63 (3.23, 4.09)*3.09 (2.72, 3.52)*BMI*WC (Per SD)4.50 (4.03, 5.03)*4.41 (3.94, 4.93)*3.68 (3.27, 4.15)*3.13 (2.74, 3.56)*BMI*ABSI (quartiles)Quartile 11.01.01.01.0Quartile 210.90 (4.64, 25.59)*10.32 (4.39, 24.24)*8.42 (3.57, 19.86)*7.47 (3.16, 17.65)*Quartile 343.73 (19.20, 99.63)*39.09 (17.13, 89.20)*25.14 (10.94, 57.76)*19.49 (8.44, 44.98)*Quartile 4217.88 (96.72, 490.84)*188.51 (83.50, 425.59)*105.60 (46.43, 240.20)*63.97 (27.87, 146.85)*P for trend< 0.0001< 0.0001< 0.0001< 0.0001MaleBMI1.62 (1.58, 1.67)*1.62 (1.58, 1.67)*1.52 (1.48, 1.57)*1.38 (1.34, 1.42)*WC1.19 (1.18, 1.20)*1.21 (1.20, 1.22)*1.18 (1.17, 1.19)*1.13 (1.12, 1.15)*ABSI1.09 (1.07, 1.10)*1.10 (1.08, 1.12)*1.06 (1.04, 1.08)*1.05 (1.02, 1.07)*ARI4.39 (4.06, 4.75)*4.40 (4.07, 4.77)*3.68 (3.38, 4.00)*2.72 (2.48, 2.98)*OBMI+ABSI1.83 (1.77, 1.89)*1.83 (1.77, 1.89)*1.70 (1.65, 1.76)*1.50 (1.45, 1.56)*OBMI+WC1.47 (1.44, 1.50)*1.47 (1.44, 1.50)*1.41 (1.38, 1.44)*1.30 (1.27, 1.33)*BMI*ABSI (Per SD)5.02 (4.61, 5.48)*5.08 (4.66, 5.55)*4.16 (3.80, 4.56)*2.97 (2.68, 3.28)*BMI*WC (Per SD)4.72 (4.34, 5.13)*4.83 (4.44, 5.26)*3.95 (3.62, 4.32)*2.85 (2.59, 3.15)*BMI*ABSI (quartiles)Quartile 11.01.01.01.0Quartile 25.76 (3.00, 11.09)*5.89 (3.06, 11.33)*4.68 (2.42, 9.02)*3.97 (2.03, 7.78)*Quartile 323.97 (12.75, 45.07)*25.00 (13.29, 47.03)*15.83 (8.38, 29.90)*10.27 (5.35, 19.72)*Quartile 4102.03 (54.44, 191.23)*108.71 (57.95, 203.93)*58.44 (30.99, 110.21)*26.15 (13.61, 50.25)*P for trend< 0.0001< 0.0001< 0.0001< 0.0001*P < 0.0001; Abbreviation: SD: standard deviation; other abbreviations as in Table 1Model 1 adjusted for age and heightModel 2 adjusted for model 1 plus TC, TG, HDL-C, drinking statusModel 3 adjusted for model 2 plus ALT, AST, GGT, FPG, HbA1c, and SBP. Odds ratios and 95% confidence intervals of BMI, WC, ABSI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and ARI for NAFLD in female and male subjects *P < 0.0001; Abbreviation: SD: standard deviation; other abbreviations as in Table 1 Model 1 adjusted for age and height Model 2 adjusted for model 1 plus TC, TG, HDL-C, drinking status Model 3 adjusted for model 2 plus ALT, AST, GGT, FPG, HbA1c, and SBP. Correlation analysis of the basic anthropometric measures showed a strong correlation between BMI and WC in both sexes (r: male 0.8796; female 0.8191), while BMI was barely correlated with ABSI (r < 0.05) (Supplementary Table 2). In contrast, the combination of ABSI and BMI may be more appropriate. Table 4 presents the associations between BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC and the risk of NAFLD in both sexes. In multivariate-adjusted logistic regression models, BMI and WC, and ABSI were positively correlated with NAFLD risk. In the current study, the combined indicators ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC were positively correlated with NAFLD risk in all four logistic regression models, and the OR values ​​in the univariate logistic regression model ranged from 1.44 to 5.02; after further adjustment for age, height, GGT, TC, AST, HDL-C, drinking status, ALT, TG, FPG, HbA1c, and SBP (Model 3), the results did not change significantly. The risk of NAFLD increased by 33% (OR 1.33, 95%CI 1.29, 1.37) and 52% (OR 1.52, 95%CI 1.45, 1.60) and 172% (OR 2.72, 95%CI 2.43, 3.04), in females, for each unit increase in OBMI+WC and OBMI+ABSI and ARI respectively. Similarly, in males, NAFLD risk was increased by 30% (OR 1.30, 95%CI 1.27, 1.33) and 50% (OR 1.50, 95%CI 1.45, 1.56) and 172% (OR 2.72, 95%CI 2.48, 2.98), respectively. Each standard deviation increase in BMI*WC and BMI*ABSI in females was associated with a 213% (OR 3.13, 95%CI 2.74,3,56) and 209% (OR 3.09, 95%CI 2.72, 3.52) increased risk for NAFLD respectively, and for males 185% (OR 2.85, 95%CI 2.59, 3.15) and 197% (OR 2.97, 95%CI 2.68, 3.28), respectively. Furthermore, the association between BMI*ABSI and NAFLD remained unchanged after treating BMI*ABSI as a categorical variable; taking the first quartile as the control group, the NAFLD risk increased with the increase of BMI*ABSI quartiles (all P for trend < 0.001). Moreover, we also analyzed the risk of NAFLD for each obesity phenotype in multivariable logistic regression models (Supplementary Table 3), which in model 3 showed that compared with the BMIN/WCN phenotype, male and female BMIO/ WCN, BMIN/WCO, and BMIO/WCO phenotypes all had a significantly higher risk of NAFLD. The risk of NAFLD associated with the BMIO/ WCN, BMIN/WCO, and BMIO/WCO phenotypes had ORs of 3.67, 3.77, and 9.78 in females and 2.76, 2.74, and 4.19 in males, respectively. Clearly, subjects with the BMIO/WCO phenotype consistently had the highest risk of developing NAFLD. Table 4Odds ratios and 95% confidence intervals of BMI, WC, ABSI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and ARI for NAFLD in female and male subjectsOdds ratios (95% confidence interval)Crude modelModel 1Model 2Model 3FemaleBMI1.58 (1.53, 1.64)*1.58 (1.52, 1.63)*1.49 (1.44, 1.55)*1.42 (1.36, 1.47)*WC1.19 (1.17, 1.21)*1.19 (1.17, 1.21)*1.17 (1.15, 1.18)*1.14 (1.12, 1.16)*ABSI1.09 (1.07, 1.12)*1.07 (1.05, 1.09)*1.06 (1.04, 1.08)*1.05 (1.02, 1.07)*ARI3.70 (3.36, 4.07)*3.61 (3.28, 3.98)*3.11 (2.80, 3.44)*2.72 (2.43, 3.04)*OBMI+ABSI1.73 (1.66, 1.80)*1.71 (1.65, 1.79)*1.61 (1.54, 1.68)*1.52 (1.45, 1.60)*OBMI+WC1.44 (1.41, 1.48)*1.44 (1.40, 1.48)*1.38 (1.34, 1.42)*1.33 (1.29, 1.37)*BMI*ABSI (Per SD)4.49 (4.02, 5.02)*4.33 (3.87, 4.84)*3.63 (3.23, 4.09)*3.09 (2.72, 3.52)*BMI*WC (Per SD)4.50 (4.03, 5.03)*4.41 (3.94, 4.93)*3.68 (3.27, 4.15)*3.13 (2.74, 3.56)*BMI*ABSI (quartiles)Quartile 11.01.01.01.0Quartile 210.90 (4.64, 25.59)*10.32 (4.39, 24.24)*8.42 (3.57, 19.86)*7.47 (3.16, 17.65)*Quartile 343.73 (19.20, 99.63)*39.09 (17.13, 89.20)*25.14 (10.94, 57.76)*19.49 (8.44, 44.98)*Quartile 4217.88 (96.72, 490.84)*188.51 (83.50, 425.59)*105.60 (46.43, 240.20)*63.97 (27.87, 146.85)*P for trend< 0.0001< 0.0001< 0.0001< 0.0001MaleBMI1.62 (1.58, 1.67)*1.62 (1.58, 1.67)*1.52 (1.48, 1.57)*1.38 (1.34, 1.42)*WC1.19 (1.18, 1.20)*1.21 (1.20, 1.22)*1.18 (1.17, 1.19)*1.13 (1.12, 1.15)*ABSI1.09 (1.07, 1.10)*1.10 (1.08, 1.12)*1.06 (1.04, 1.08)*1.05 (1.02, 1.07)*ARI4.39 (4.06, 4.75)*4.40 (4.07, 4.77)*3.68 (3.38, 4.00)*2.72 (2.48, 2.98)*OBMI+ABSI1.83 (1.77, 1.89)*1.83 (1.77, 1.89)*1.70 (1.65, 1.76)*1.50 (1.45, 1.56)*OBMI+WC1.47 (1.44, 1.50)*1.47 (1.44, 1.50)*1.41 (1.38, 1.44)*1.30 (1.27, 1.33)*BMI*ABSI (Per SD)5.02 (4.61, 5.48)*5.08 (4.66, 5.55)*4.16 (3.80, 4.56)*2.97 (2.68, 3.28)*BMI*WC (Per SD)4.72 (4.34, 5.13)*4.83 (4.44, 5.26)*3.95 (3.62, 4.32)*2.85 (2.59, 3.15)*BMI*ABSI (quartiles)Quartile 11.01.01.01.0Quartile 25.76 (3.00, 11.09)*5.89 (3.06, 11.33)*4.68 (2.42, 9.02)*3.97 (2.03, 7.78)*Quartile 323.97 (12.75, 45.07)*25.00 (13.29, 47.03)*15.83 (8.38, 29.90)*10.27 (5.35, 19.72)*Quartile 4102.03 (54.44, 191.23)*108.71 (57.95, 203.93)*58.44 (30.99, 110.21)*26.15 (13.61, 50.25)*P for trend< 0.0001< 0.0001< 0.0001< 0.0001*P < 0.0001; Abbreviation: SD: standard deviation; other abbreviations as in Table 1Model 1 adjusted for age and heightModel 2 adjusted for model 1 plus TC, TG, HDL-C, drinking statusModel 3 adjusted for model 2 plus ALT, AST, GGT, FPG, HbA1c, and SBP. Odds ratios and 95% confidence intervals of BMI, WC, ABSI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and ARI for NAFLD in female and male subjects *P < 0.0001; Abbreviation: SD: standard deviation; other abbreviations as in Table 1 Model 1 adjusted for age and height Model 2 adjusted for model 1 plus TC, TG, HDL-C, drinking status Model 3 adjusted for model 2 plus ALT, AST, GGT, FPG, HbA1c, and SBP. [SUBTITLE] Accuracy of various obesity indicators to identify NAFLD [SUBSECTION] ROC curves were drawn to assess the accuracy of BMI, WC, ABSI, ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC for identifying NAFLD in both sexes (Fig. 2). Table 5 summarizes the AUC and optimal diagnostic thresholds for these obesity indicators. In contrast, ARI and OBMI+ABSI had the same and highest AUC in both female and male subjects, with AUC and optimal diagnostic thresholds of 0.8270, 33.2569 and 0.8912, 30.4143 for ARI in males and females, respectively. The Delong test further showed that both the combined indicators of BMI and WC (OBMI+WC and BMI*WC) and BMI and ABSI (OBMI+ABSI and BMI*ABSI and ARI) were significantly better at identifying the risk of NAFLD than BMI or WC or ABSI alone (Delong test P < 0.05). Fig. 2ROC curve analysis of BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC in females and males ROC curve analysis of BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC in females and males Table 5The best threshold, sensitivities, specificities, Youden’s index, and area under the curve of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI in different gendersAUC95%CI low95%CI upBest thresholdSpecificitySensitivityYouden’s indexFemaleBMI**0.87990.86480.895022.75650.81770.79500.6127WC**0.86950.85450.884474.25000.70500.87450.5804ABSI**ARI**0.61710.89120.59160.87750.64260.904875.792512.78250.59260.77350.59620.85770.18880.6312OBMI+ABSI**0.89120.87750.904830.41430.77330.85770.6310OBMI+WC0.88880.87500.902734.12340.77160.85560.6272BMI*ABSI0.88840.87480.90211704.21000.79220.82850.6207BMI*WC0.88880.87500.90271674.11830.77330.85560.6286MaleBMI**0.81600.80550.826423.55550.73820.73090.4691WC**0.81020.79980.820780.65000.66740.80340.4708ABSI**0.57950.56530.593674.91280.40520.72400.1292ARI**0.82700.81700.836913.60960.74560.74570.4913OBMI+ABSI**0.82700.81700.836933.25690.74580.74570.4915OBMI+WC0.82420.81410.834339.83130.74380.74470.4885BMI*ABSI0.82570.81570.83561758.75900.67610.81170.4878BMI*WC0.82440.81430.83451944.04330.75510.73290.4880Abbreviations as in Table 1**P < 0.05 compared with BMI*WC in each gender The best threshold, sensitivities, specificities, Youden’s index, and area under the curve of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI in different genders ABSI** ARI** 0.6171 0.8912 0.5916 0.8775 0.6426 0.9048 75.7925 12.7825 0.5926 0.7735 0.5962 0.8577 0.1888 0.6312 Abbreviations as in Table 1 **P < 0.05 compared with BMI*WC in each gender To explore the changes in BMI, WC, ABSI, ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC on the risk identification ability of NAFLD in different age subgroups of both sexes, we constructed ROC curves for 20–39, 40–59, and ≥ 60 years in both sexes, respectively; and the corresponding AUC and optimal diagnostic thresholds were summarized in Table 6. We found that ARI and OBMI+ABSI exhibited considerably higher AUCs in all age subgroups for both sexes except in the female ≥ 60-year age group; the AUC of OBMI+ABSI in the female 20–39 age group was 0.9523, the highest value among all age subgroups of both sexes. In the male 20-59-year-old group and the female 40-59-year-old group, the AUC values ​​of ARI and OBMI+ABSI were slightly higher than those of BMI*WC, but all were significantly higher than those of BMI or WC or ABSI alone, while in the female 20–39 years group and the female ≥ 60 years group AUC values for all combined obesity indicators were significantly higher than that of WC and ABSI; in addition, in the male ≥ 60-year-old group, the seven indicators of obesity had similar NAFLD identification abilities, only ABSI underperformed. Therefore, the combined indicators ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC can significantly improve the ability of simple obesity parameters BMI and WC and ABSI to identify NAFLD in middle-aged females and young and middle-aged males and had the highest diagnostic performance in young females. Table 6The area under the curve, best threshold, sensitivities, specificities, and Youden’s index of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI as a predictor of NAFLD in the different age groups of both gendersAUC95%CI low95%CI upBest thresholdSpecificitySensitivityYouden’s indexFemaleAge 20–39 yearsBMI0.95110.93360.968523.12660.89810.87230.7704WC***0.93190.90790.955976.60000.85600.88300.7390ABSI***0.56860.50850.628875.57420.61220.53190.1441ARI0.95220.93370.970812.79220.84980.93620.7860BMI*WC0.95160.93310.97011675.47840.84360.92550.7691BMI*ABSI0.94860.92890.96821701.34860.86140.92550.7869OBMI+ABSI0.95230.93370.970830.43680.84980.93620.7860OBMI+WC0.95160.93300.970134.12960.84240.92550.7679Age 40–59 yearsBMI***0.84840.82820.868622.75270.78410.77440.5585WC***0.84230.82300.861674.25000.66300.86910.5321ABSI***0.61860.58970.647675.79250.58080.60720.1880ARI***0.86220.84380.880513.02830.80770.77440.5821BMI*WC0.86030.84170.87881674.11830.73320.83840.5716BMI*ABSI0.86020.84200.87851704.21000.75460.81340.5680OBMI+ABSI***0.86220.84380.880530.99950.80790.77440.5823OBMI+WC0.86020.84170.878835.03630.80930.76320.5725Age ≥ 60 yearsBMI0.83080.74580.915923.33340.84390.76000.6039WC***0.76960.67780.861379.75000.77070.68000.4507ABSI***0.56600.45510.677078.18360.50240.72000.2224ARI0.81950.73910.899912.92520.70240.84000.5424BMI*WC0.81990.73960.90021686.12800.69760.84000.5376BMI*ABSI0.80140.71860.88411659.09940.56100.96000.5210OBMI+ABSI0.81950.73920.899830.75250.70240.84000.5424OBMI+WC0.82010.73930.900934.33930.70240.84000.5424MaleAge 20–39 yearsBMI***0.84390.82750.860223.47820.74950.76650.5160WC***0.84150.82550.857580.45000.72190.80090.5228ABSI***0.60630.58260.630074.50900.48710.69910.1862ARI***0.85490.83960.870213.50260.76000.78440.5444BMI*WC0.85130.83570.86691862.54150.72190.81140.5333BMI*ABSI***0.85520.83990.87041766.28860.75570.78740.5431OBMI+ABSI***0.85490.83960.870233.00760.76140.78290.5443OBMI+WC0.85140.83580.866939.14530.72900.80840.5374Age 40–59 yearsBMI***0.80070.78670.814723.25050.67790.76390.4418WC***0.79080.77660.805181.25000.65830.77950.4378ABSI***0.56150.54290.580175.42940.38970.70050.0902ARI***0.81040.79690.824013.61420.72320.74510.4683BMI*WC0.80800.79430.82171955.12030.74480.71770.4625BMI*ABSI0.80860.79500.82221816.10160.73340.72870.4621OBMI+ABSI***0.81050.79690.824033.27330.72390.74430.4682OBMI+WC0.80750.79380.821240.11920.74750.71850.4660Age ≥ 60 yearsBMI0.79030.73630.844423.74200.81110.69510.5062WC0.78040.72820.832680.50000.59440.86590.4603ABSI***0.58040.51190.649075.85420.29410.86590.1600ARI0.79490.74370.846213.58190.67800.81710.4951BMI*WC0.80290.75130.85451932.61210.73680.79270.5295BMI*ABSI0.79040.73930.84141800.36700.65940.82930.4887OBMI+ABSI0.79500.74380.846233.18800.67800.81710.4951OBMI+WC0.80230.75090.853639.74760.71210.81710.5292Abbreviations as in Table 1***P < 0.05 compared with BMI*WC in each age group of both genders The area under the curve, best threshold, sensitivities, specificities, and Youden’s index of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI as a predictor of NAFLD in the different age groups of both genders Abbreviations as in Table 1 ***P < 0.05 compared with BMI*WC in each age group of both genders ROC curves were drawn to assess the accuracy of BMI, WC, ABSI, ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC for identifying NAFLD in both sexes (Fig. 2). Table 5 summarizes the AUC and optimal diagnostic thresholds for these obesity indicators. In contrast, ARI and OBMI+ABSI had the same and highest AUC in both female and male subjects, with AUC and optimal diagnostic thresholds of 0.8270, 33.2569 and 0.8912, 30.4143 for ARI in males and females, respectively. The Delong test further showed that both the combined indicators of BMI and WC (OBMI+WC and BMI*WC) and BMI and ABSI (OBMI+ABSI and BMI*ABSI and ARI) were significantly better at identifying the risk of NAFLD than BMI or WC or ABSI alone (Delong test P < 0.05). Fig. 2ROC curve analysis of BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC in females and males ROC curve analysis of BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC in females and males Table 5The best threshold, sensitivities, specificities, Youden’s index, and area under the curve of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI in different gendersAUC95%CI low95%CI upBest thresholdSpecificitySensitivityYouden’s indexFemaleBMI**0.87990.86480.895022.75650.81770.79500.6127WC**0.86950.85450.884474.25000.70500.87450.5804ABSI**ARI**0.61710.89120.59160.87750.64260.904875.792512.78250.59260.77350.59620.85770.18880.6312OBMI+ABSI**0.89120.87750.904830.41430.77330.85770.6310OBMI+WC0.88880.87500.902734.12340.77160.85560.6272BMI*ABSI0.88840.87480.90211704.21000.79220.82850.6207BMI*WC0.88880.87500.90271674.11830.77330.85560.6286MaleBMI**0.81600.80550.826423.55550.73820.73090.4691WC**0.81020.79980.820780.65000.66740.80340.4708ABSI**0.57950.56530.593674.91280.40520.72400.1292ARI**0.82700.81700.836913.60960.74560.74570.4913OBMI+ABSI**0.82700.81700.836933.25690.74580.74570.4915OBMI+WC0.82420.81410.834339.83130.74380.74470.4885BMI*ABSI0.82570.81570.83561758.75900.67610.81170.4878BMI*WC0.82440.81430.83451944.04330.75510.73290.4880Abbreviations as in Table 1**P < 0.05 compared with BMI*WC in each gender The best threshold, sensitivities, specificities, Youden’s index, and area under the curve of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI in different genders ABSI** ARI** 0.6171 0.8912 0.5916 0.8775 0.6426 0.9048 75.7925 12.7825 0.5926 0.7735 0.5962 0.8577 0.1888 0.6312 Abbreviations as in Table 1 **P < 0.05 compared with BMI*WC in each gender To explore the changes in BMI, WC, ABSI, ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC on the risk identification ability of NAFLD in different age subgroups of both sexes, we constructed ROC curves for 20–39, 40–59, and ≥ 60 years in both sexes, respectively; and the corresponding AUC and optimal diagnostic thresholds were summarized in Table 6. We found that ARI and OBMI+ABSI exhibited considerably higher AUCs in all age subgroups for both sexes except in the female ≥ 60-year age group; the AUC of OBMI+ABSI in the female 20–39 age group was 0.9523, the highest value among all age subgroups of both sexes. In the male 20-59-year-old group and the female 40-59-year-old group, the AUC values ​​of ARI and OBMI+ABSI were slightly higher than those of BMI*WC, but all were significantly higher than those of BMI or WC or ABSI alone, while in the female 20–39 years group and the female ≥ 60 years group AUC values for all combined obesity indicators were significantly higher than that of WC and ABSI; in addition, in the male ≥ 60-year-old group, the seven indicators of obesity had similar NAFLD identification abilities, only ABSI underperformed. Therefore, the combined indicators ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC can significantly improve the ability of simple obesity parameters BMI and WC and ABSI to identify NAFLD in middle-aged females and young and middle-aged males and had the highest diagnostic performance in young females. Table 6The area under the curve, best threshold, sensitivities, specificities, and Youden’s index of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI as a predictor of NAFLD in the different age groups of both gendersAUC95%CI low95%CI upBest thresholdSpecificitySensitivityYouden’s indexFemaleAge 20–39 yearsBMI0.95110.93360.968523.12660.89810.87230.7704WC***0.93190.90790.955976.60000.85600.88300.7390ABSI***0.56860.50850.628875.57420.61220.53190.1441ARI0.95220.93370.970812.79220.84980.93620.7860BMI*WC0.95160.93310.97011675.47840.84360.92550.7691BMI*ABSI0.94860.92890.96821701.34860.86140.92550.7869OBMI+ABSI0.95230.93370.970830.43680.84980.93620.7860OBMI+WC0.95160.93300.970134.12960.84240.92550.7679Age 40–59 yearsBMI***0.84840.82820.868622.75270.78410.77440.5585WC***0.84230.82300.861674.25000.66300.86910.5321ABSI***0.61860.58970.647675.79250.58080.60720.1880ARI***0.86220.84380.880513.02830.80770.77440.5821BMI*WC0.86030.84170.87881674.11830.73320.83840.5716BMI*ABSI0.86020.84200.87851704.21000.75460.81340.5680OBMI+ABSI***0.86220.84380.880530.99950.80790.77440.5823OBMI+WC0.86020.84170.878835.03630.80930.76320.5725Age ≥ 60 yearsBMI0.83080.74580.915923.33340.84390.76000.6039WC***0.76960.67780.861379.75000.77070.68000.4507ABSI***0.56600.45510.677078.18360.50240.72000.2224ARI0.81950.73910.899912.92520.70240.84000.5424BMI*WC0.81990.73960.90021686.12800.69760.84000.5376BMI*ABSI0.80140.71860.88411659.09940.56100.96000.5210OBMI+ABSI0.81950.73920.899830.75250.70240.84000.5424OBMI+WC0.82010.73930.900934.33930.70240.84000.5424MaleAge 20–39 yearsBMI***0.84390.82750.860223.47820.74950.76650.5160WC***0.84150.82550.857580.45000.72190.80090.5228ABSI***0.60630.58260.630074.50900.48710.69910.1862ARI***0.85490.83960.870213.50260.76000.78440.5444BMI*WC0.85130.83570.86691862.54150.72190.81140.5333BMI*ABSI***0.85520.83990.87041766.28860.75570.78740.5431OBMI+ABSI***0.85490.83960.870233.00760.76140.78290.5443OBMI+WC0.85140.83580.866939.14530.72900.80840.5374Age 40–59 yearsBMI***0.80070.78670.814723.25050.67790.76390.4418WC***0.79080.77660.805181.25000.65830.77950.4378ABSI***0.56150.54290.580175.42940.38970.70050.0902ARI***0.81040.79690.824013.61420.72320.74510.4683BMI*WC0.80800.79430.82171955.12030.74480.71770.4625BMI*ABSI0.80860.79500.82221816.10160.73340.72870.4621OBMI+ABSI***0.81050.79690.824033.27330.72390.74430.4682OBMI+WC0.80750.79380.821240.11920.74750.71850.4660Age ≥ 60 yearsBMI0.79030.73630.844423.74200.81110.69510.5062WC0.78040.72820.832680.50000.59440.86590.4603ABSI***0.58040.51190.649075.85420.29410.86590.1600ARI0.79490.74370.846213.58190.67800.81710.4951BMI*WC0.80290.75130.85451932.61210.73680.79270.5295BMI*ABSI0.79040.73930.84141800.36700.65940.82930.4887OBMI+ABSI0.79500.74380.846233.18800.67800.81710.4951OBMI+WC0.80230.75090.853639.74760.71210.81710.5292Abbreviations as in Table 1***P < 0.05 compared with BMI*WC in each age group of both genders The area under the curve, best threshold, sensitivities, specificities, and Youden’s index of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI as a predictor of NAFLD in the different age groups of both genders Abbreviations as in Table 1 ***P < 0.05 compared with BMI*WC in each age group of both genders
Conclusion
Taken together, our findings confirmed that BMI combined with ABSI and WC can better explain obesity-related NAFLD risk than a single indicator in the general population, and identified the ARI and the multiplicative combination indicators BMI*ABSI and BMI*WC as the simplest and very effective combination indicators for diagnosing the risk of NAFLD. These new findings underscored the importance of the combined application of BMI and the abdominal obesity index in clinical practice to assess NAFLD risk in the general population and provided a cost-effective tool for precise preventive screening and treatment monitoring for NAFLD.
[ "Background", "Methods", "Data sources and study design", "Definition and derivation of the combined indicators of BMI with WC and ABSI", "Combination using usual cut-offs for BMI and WC", "Optimal proportional combinations OBMI+WC and OBMI+ABSI, multiplicative combination indicators BMI*ABSI and BMI*WC, and ARI", "Diagnosis of NAFLD", "Statistical analysis", "Demographic and clinical characteristics of subjects", "Calculation of the optimal proportional combination of BMI with WC and ABSI", "Association of various obesity indicators with NAFLD", "Accuracy of various obesity indicators to identify NAFLD", "Advantages and limitations of research", "" ]
[ "Non-alcoholic fatty liver disease (NAFLD) is a chronic non-infectious liver disease characterized by oxidative stress, inflammation, and fibrosis in hepatocytes among people without a history of excessive alcohol consumption [1, 2]. NAFLD can present as asymptomatic simple hepatic steatosis in its early stages, and without intervention, oxidative stress, inflammation, and fibrosis in hepatocytes can contribute to the continuous progression of NAFLD to non-alcoholic steatohepatitis and then to cirrhosis and ultimately to hepatocellular carcinoma [2, 3]. Recent epidemiological surveys have shown that about a quarter of the global population suffers from NAFLD, with a 27.4% estimated prevalence in Asia [4]. NAFLD is not only the most common chronic liver disease worldwide but has replaced hepatitis B virus infection as the primary risk factor for advanced liver diseases such as cirrhosis and hepatocellular carcinoma [5]. The high prevalence of NAFLD and its severe complications place a heavy burden on humans and the world’s health care systems, making the early prevention, identification, and intervention of NAFLD of great public health importance.\nObesity is one of the most important risk factors for NAFLD [6]. Waist circumference (WC) and body mass index (BMI) are currently the most widely used basic body measures for the assessment of general and central obesity and also are known risk factors for NAFLD [7–10]. Both BMI and WC alone, however, have some distinct limitations, such as the inability of BMI alone to distinguish between adipose and muscle tissue and the inability of WC to determine the differential contribution of abdominal visceral adipose tissue and abdominal subcutaneous adipose tissue to central obesity [11–13]. It is not clear if combining risk associated with BMI and WC can improve the individual risk association. Several relevant studies have analyzed the performance of the combination of BMI and WC in disease risk assessment, and overall, combining BMI and WC in different ways significantly improved the predictive efficacy for obesity-related diseases such as obesity-related hypertension, type 2 diabetes, cardiovascular disease, and all-cause mortality risk compared to a single indicator [14–17]. In addition, a recent study by Wang et al. also found that high levels of WC and BMI were both associated with NAFLD risk in people with type 2 diabetes and that BMI combined with WC was superior to the single measures in assessing the risk of NAFLD [18]. What remains unclear, however, is whether the combination of BMI and WC best identifies a higher risk of NAFLD in the general population. Furthermore, it is important to note that there is a strong correlation between BMI and WC (r ≈ 0.80), and directly combining BMI with WC has the potential to confound the association between BMI and NAFLD, leading to biased and possibly misleading risk estimates [19–21]. A Body Shape Index (ABSI), an allometric abdominal obesity index calculated from WC, height, and weight, is intended to be independent of BMI and has proven to be useful for identifying individuals at risk for sarcopenic obesity [22]; therefore, we also evaluated combining BMI with the non-correlated abdominal obesity index ABSI. This study sought to explore risk assessment for NAFLD of various combinations of BMI and WC or ABSI in a cohort of general health examinees from NAGALA.", "[SUBTITLE] Data sources and study design [SUBSECTION] In the current study, we used data from 14,251 medical examiners recruited by the NAGALA cohort between 1994 and 2016 [23]. NAGALA is an ongoing longitudinal cohort study initiated by Murakami Memorial Hospital in 1994, using physical examination data from the general population to assess common risk factors for the development of chronic diseases; a detailed description of the NAGALA cohort study design has been previously published and the study data have been uploaded by Professor Okamura to the Dryad database for public access [24]. For the current study, we excluded from the original data set (1) subjects who were diagnosed or self-reported with hepatitis (viral/alcoholic) or diabetes and impaired fasting glucose at baseline (n = 1,547); (2) subjects with alcohol abuse (n = 1,952, males consuming ≥ 210 g of alcohol per week and females consuming ≥ 140 g) [25]; (3) subjects taking any medications at baseline (n = 2,321); and, (4) subjects with missing anthropometric and medical examination data (n = 873) were also excluded. We ultimately included 14,251 eligible subjects, with the inclusion and exclusion criteria shown in Fig. 1. The NAGALA cohort study has been ethically reviewed by the Murakami Memorial Hospital Institutional Ethics Review Committee (IRB2018-09-01) and all subjects signed written informed consent for the use of their data; The current study was a post-hoc analysis of the NAGALA cohort, and the Jiangxi Provincial People’s Hospital Ethics Review Committee reviewed the study design and granted approval (IRB2021-066).\n\nFig. 1Flowchart of the selection process of study subjects\n\nFlowchart of the selection process of study subjects\nIn this study, we extracted the following data from each subject: anthropometric and demographic information including weight, height, sex, age, WC, BMI, ABSI, smoking and drinking status, and exercise habits of the subjects; where drinking status was classified according to the subject’s weekly alcohol consumption in the previous month as moderate drinking, light drinking and never or small drinking; smoking status was defined as never, previous and current smoking; for exercise habits, subjects were considered to have exercise habits if they had at least one physical activity of any form per week. BMI was calculated as weight (kg)/[height (m)]2 and ABSI was calculated as 1,000*WC (m)*[weight (kg)] – 2/3*[height (m)]5/6 [22].\nData on laboratory tests, including blood pressure, triglycerides (TG), aspartate aminotransferase (AST), total cholesterol (TC), fasting blood glucose (FPG), gamma-glutamyl transferase (GGT), high-density lipoprotein cholesterol (HDL-C), alanine aminotransferase (ALT), and glycated hemoglobin (HbA1c). The specific steps for obtaining demographic and anthropometric information and data from laboratory tests have been described in detail elsewhere [19]; all procedures in this study fully followed the Declaration of Helsinki. See STROBE statement (S1 Text).\nIn the current study, we used data from 14,251 medical examiners recruited by the NAGALA cohort between 1994 and 2016 [23]. NAGALA is an ongoing longitudinal cohort study initiated by Murakami Memorial Hospital in 1994, using physical examination data from the general population to assess common risk factors for the development of chronic diseases; a detailed description of the NAGALA cohort study design has been previously published and the study data have been uploaded by Professor Okamura to the Dryad database for public access [24]. For the current study, we excluded from the original data set (1) subjects who were diagnosed or self-reported with hepatitis (viral/alcoholic) or diabetes and impaired fasting glucose at baseline (n = 1,547); (2) subjects with alcohol abuse (n = 1,952, males consuming ≥ 210 g of alcohol per week and females consuming ≥ 140 g) [25]; (3) subjects taking any medications at baseline (n = 2,321); and, (4) subjects with missing anthropometric and medical examination data (n = 873) were also excluded. We ultimately included 14,251 eligible subjects, with the inclusion and exclusion criteria shown in Fig. 1. The NAGALA cohort study has been ethically reviewed by the Murakami Memorial Hospital Institutional Ethics Review Committee (IRB2018-09-01) and all subjects signed written informed consent for the use of their data; The current study was a post-hoc analysis of the NAGALA cohort, and the Jiangxi Provincial People’s Hospital Ethics Review Committee reviewed the study design and granted approval (IRB2021-066).\n\nFig. 1Flowchart of the selection process of study subjects\n\nFlowchart of the selection process of study subjects\nIn this study, we extracted the following data from each subject: anthropometric and demographic information including weight, height, sex, age, WC, BMI, ABSI, smoking and drinking status, and exercise habits of the subjects; where drinking status was classified according to the subject’s weekly alcohol consumption in the previous month as moderate drinking, light drinking and never or small drinking; smoking status was defined as never, previous and current smoking; for exercise habits, subjects were considered to have exercise habits if they had at least one physical activity of any form per week. BMI was calculated as weight (kg)/[height (m)]2 and ABSI was calculated as 1,000*WC (m)*[weight (kg)] – 2/3*[height (m)]5/6 [22].\nData on laboratory tests, including blood pressure, triglycerides (TG), aspartate aminotransferase (AST), total cholesterol (TC), fasting blood glucose (FPG), gamma-glutamyl transferase (GGT), high-density lipoprotein cholesterol (HDL-C), alanine aminotransferase (ALT), and glycated hemoglobin (HbA1c). The specific steps for obtaining demographic and anthropometric information and data from laboratory tests have been described in detail elsewhere [19]; all procedures in this study fully followed the Declaration of Helsinki. See STROBE statement (S1 Text).\n[SUBTITLE] Definition and derivation of the combined indicators of BMI with WC and ABSI [SUBSECTION] Based on previous studies, the present study included four combination types of BMI with WC and ABSI: (1) Using the WHO-recommended clinical cut-offs of WC and BMI combined into four obesity phenotypes; (2) constructing the optimal proportional combination of BMI with WC and ABSI, OBMI+WC and OBMI+ABSI; and (3) using the multiplicative combination of WC and ABSI with BMI, BMI*WC, and BMI*ABSI; (4) construction of anthropometric risk index [ARI (BMI, ABSI)] for combined risk of BMI and ABSI using the method recommended by Krakauer NY et al. [26].\nBased on previous studies, the present study included four combination types of BMI with WC and ABSI: (1) Using the WHO-recommended clinical cut-offs of WC and BMI combined into four obesity phenotypes; (2) constructing the optimal proportional combination of BMI with WC and ABSI, OBMI+WC and OBMI+ABSI; and (3) using the multiplicative combination of WC and ABSI with BMI, BMI*WC, and BMI*ABSI; (4) construction of anthropometric risk index [ARI (BMI, ABSI)] for combined risk of BMI and ABSI using the method recommended by Krakauer NY et al. [26].\n[SUBTITLE] Combination using usual cut-offs for BMI and WC [SUBSECTION] In accordance with the World Health Organization (WHO) expert committee cut-off recommendations for Asian populations, we defined BMI ≥ 25 kg/m2 as overweight/obese and BMI < 25 kg/m2 as normal weight [27]. We defined WC ≥ 85 cm for males or WC ≥ 80 cm for females as central obesity and WC < 85 cm for males or WC < 80 cm for females as normal WC [28]. All subjects were assigned to the following four obesity phenotypes according to the baseline WC and BMI: (1) BMIN/WCN: normal weight + normal WC; (2) BMIO/WCN: overweight/obese + normal WC; (3) BMIN/WCO: normal weight + central obesity; (4) BMIO/WCO: overweight/obese + central obesity.\nIn accordance with the World Health Organization (WHO) expert committee cut-off recommendations for Asian populations, we defined BMI ≥ 25 kg/m2 as overweight/obese and BMI < 25 kg/m2 as normal weight [27]. We defined WC ≥ 85 cm for males or WC ≥ 80 cm for females as central obesity and WC < 85 cm for males or WC < 80 cm for females as normal WC [28]. All subjects were assigned to the following four obesity phenotypes according to the baseline WC and BMI: (1) BMIN/WCN: normal weight + normal WC; (2) BMIO/WCN: overweight/obese + normal WC; (3) BMIN/WCO: normal weight + central obesity; (4) BMIO/WCO: overweight/obese + central obesity.\n[SUBTITLE] Optimal proportional combinations OBMI+WC and OBMI+ABSI, multiplicative combination indicators BMI*ABSI and BMI*WC, and ARI [SUBSECTION] The optimal proportional combination indicator OBMI+WC was calculated as OBMI+WC= nWC*WC + nBMI*BMI. The optimal proportion coefficients for WC (nWC) and BMI (nBMI) were calculated as nWC = βWC / (βBMI + βWC) and nBMI = βBMI / (βBMI + βWC), respectively; where βWC and βBMI were the regression coefficients of WC and BMI in the multivariable logistic regression model, respectively. To acquire regression coefficients for WC and BMI, we included the presence or absence of NAFLD as the outcome event of interest, WC and BMI as variables in a multivariate-adjusted logistic regression model, and age, height, ALT, AST, systolic blood pressure (SBP), TG, GGT, HDL-C, HbA1c, TC, FPG, and drinking status were adjusted as covariates. Similarly, we used the same method to obtain OBMI+ABSI. Since NAFLD risk was positively correlated with BMI, WC, and ABSI, and there was no correlation between BMI and ABSI, which is consistent with the construction of an ARI (BMI, ABSI) for the combined risk of BMI and ABSI, and we calculated ARI (BMI, ABSI) using the method provided by Krakauer NY et al. [26]. In addition, we calculated the products BMI*WC and BMI* ABSI for further analysis.\nThe optimal proportional combination indicator OBMI+WC was calculated as OBMI+WC= nWC*WC + nBMI*BMI. The optimal proportion coefficients for WC (nWC) and BMI (nBMI) were calculated as nWC = βWC / (βBMI + βWC) and nBMI = βBMI / (βBMI + βWC), respectively; where βWC and βBMI were the regression coefficients of WC and BMI in the multivariable logistic regression model, respectively. To acquire regression coefficients for WC and BMI, we included the presence or absence of NAFLD as the outcome event of interest, WC and BMI as variables in a multivariate-adjusted logistic regression model, and age, height, ALT, AST, systolic blood pressure (SBP), TG, GGT, HDL-C, HbA1c, TC, FPG, and drinking status were adjusted as covariates. Similarly, we used the same method to obtain OBMI+ABSI. Since NAFLD risk was positively correlated with BMI, WC, and ABSI, and there was no correlation between BMI and ABSI, which is consistent with the construction of an ARI (BMI, ABSI) for the combined risk of BMI and ABSI, and we calculated ARI (BMI, ABSI) using the method provided by Krakauer NY et al. [26]. In addition, we calculated the products BMI*WC and BMI* ABSI for further analysis.\n[SUBTITLE] Diagnosis of NAFLD [SUBSECTION] Specialized gastroenterologists, blinded to the subjects scored each abdominal ultrasound for NAFLD risk based on four criteria: (1) changes in the intensity of liver blood flow signals (0–4 points); (2) the clarity of liver blood vessels (0–1 points); (3) liver and kidney echo contrast (0–4 points); (4) deep liver echo attenuation (0–2 points). NAFLD was diagnosed if the sum of the above four scores was greater than 2 [29].\nSpecialized gastroenterologists, blinded to the subjects scored each abdominal ultrasound for NAFLD risk based on four criteria: (1) changes in the intensity of liver blood flow signals (0–4 points); (2) the clarity of liver blood vessels (0–1 points); (3) liver and kidney echo contrast (0–4 points); (4) deep liver echo attenuation (0–2 points). NAFLD was diagnosed if the sum of the above four scores was greater than 2 [29].\n[SUBTITLE] Statistical analysis [SUBSECTION] We performed all statistical analysis steps in the current study using Empower (R) version 2.2 and R language version 3.4.3. Significance was defined as P < 0.05 in all comparisons (two-sided). Given the significant differences in body composition, fat deposition patterns, and prevalence of NAFLD between males and females [30], all analyses in the present study were performed separately for female and male subjects and the specific steps were as follows:\nFirst, we calculated the quartile categories of BMI*ABSI using the quartile function and subsequently described baseline information by grouping all subjects based on the quartile categories of BMI*ABSI and whether they had NAFLD. For the measurement data, QQ plots were first used to determine the pattern of data distribution, and the data with a normal and a skewed distribution were described as mean (standard deviation) and median (interquartile range), respectively. T-test or one-way ANOVA was used for group comparisons of normal data, and Mann-Whitney rank-sum test or Kruskal-Wallis rank-sum test was used for group comparisons of skewed data. Count data were described as frequencies (%), using the Pearson chi-square test for comparison between groups.\nSecond, before exploring the association of each obesity indicator with NAFLD, we first analyzed the correlations between several anthropometric measures (height, BMI, WC, ABSI), and it is worth mentioning that a combination of two basic anthropometric indicators of obesity which have smaller correlations is considered more appropriate. Additionally, we also performed a collinearity screening for all covariates [31], where the weight and diastolic blood pressure (DBP) had variance inflation factors (VIF) greater than 5 and were therefore treated as collinear variables and not included in the subsequent model adjustment (Supplementary Table 1). We developed one univariate and three multivariable logistic regression models for assessing the associations between the continuous variables BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and the categorical variable obesity phenotypes and risk of NAFLD, and recorded the corresponding Odds ratio (OR) and 95% confidence interval (CI), respectively. Due to the large numerical span of BMI*WC (689.56-6490.01) and BMI*ABSI (961.13-3630.94), the change in effect value caused by the change of one unit was small, so the BMI*WC and BMI*ABSI were Z-score transformed and included in the regression models. In the model adjustment of multivariable logistic regression, model 1 considered the effect of anthropometric parameters of age and height on NAFLD; model 2 adjusted drinking status and important lipid parameters (TC, TG, HDL-C) on the basis of model 1; model 3 further considered the effects of liver enzyme indicators (ALT, AST, GGT), blood glucose and blood pressure parameters (FPG, HbA1c, SBP) on the basis of model 2. In addition, to further explore the correlation between BMI*ABSI and NAFLD, we also included BMI*ABSI as a categorical variable in the logistic regression models and calculated the trend of the association between the median of each category of BMI*ABSI and NAFLD.\nThird, based on the results of the correlation analysis between each obesity indicator and NAFLD, we also constructed receiver operating characteristic (ROC) curves to evaluate the ability of continuous obesity indicator BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC to identify the risk of NAFLD. The area under the curve (AUC) and Delong test were used to compare the differences in the ability to identify NAFLD risk of each obesity indicator in both sexes and different age subgroups of both sexes, and the optimal diagnostic threshold for each indicator was calculated using Youden’s index.\nWe performed all statistical analysis steps in the current study using Empower (R) version 2.2 and R language version 3.4.3. Significance was defined as P < 0.05 in all comparisons (two-sided). Given the significant differences in body composition, fat deposition patterns, and prevalence of NAFLD between males and females [30], all analyses in the present study were performed separately for female and male subjects and the specific steps were as follows:\nFirst, we calculated the quartile categories of BMI*ABSI using the quartile function and subsequently described baseline information by grouping all subjects based on the quartile categories of BMI*ABSI and whether they had NAFLD. For the measurement data, QQ plots were first used to determine the pattern of data distribution, and the data with a normal and a skewed distribution were described as mean (standard deviation) and median (interquartile range), respectively. T-test or one-way ANOVA was used for group comparisons of normal data, and Mann-Whitney rank-sum test or Kruskal-Wallis rank-sum test was used for group comparisons of skewed data. Count data were described as frequencies (%), using the Pearson chi-square test for comparison between groups.\nSecond, before exploring the association of each obesity indicator with NAFLD, we first analyzed the correlations between several anthropometric measures (height, BMI, WC, ABSI), and it is worth mentioning that a combination of two basic anthropometric indicators of obesity which have smaller correlations is considered more appropriate. Additionally, we also performed a collinearity screening for all covariates [31], where the weight and diastolic blood pressure (DBP) had variance inflation factors (VIF) greater than 5 and were therefore treated as collinear variables and not included in the subsequent model adjustment (Supplementary Table 1). We developed one univariate and three multivariable logistic regression models for assessing the associations between the continuous variables BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and the categorical variable obesity phenotypes and risk of NAFLD, and recorded the corresponding Odds ratio (OR) and 95% confidence interval (CI), respectively. Due to the large numerical span of BMI*WC (689.56-6490.01) and BMI*ABSI (961.13-3630.94), the change in effect value caused by the change of one unit was small, so the BMI*WC and BMI*ABSI were Z-score transformed and included in the regression models. In the model adjustment of multivariable logistic regression, model 1 considered the effect of anthropometric parameters of age and height on NAFLD; model 2 adjusted drinking status and important lipid parameters (TC, TG, HDL-C) on the basis of model 1; model 3 further considered the effects of liver enzyme indicators (ALT, AST, GGT), blood glucose and blood pressure parameters (FPG, HbA1c, SBP) on the basis of model 2. In addition, to further explore the correlation between BMI*ABSI and NAFLD, we also included BMI*ABSI as a categorical variable in the logistic regression models and calculated the trend of the association between the median of each category of BMI*ABSI and NAFLD.\nThird, based on the results of the correlation analysis between each obesity indicator and NAFLD, we also constructed receiver operating characteristic (ROC) curves to evaluate the ability of continuous obesity indicator BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC to identify the risk of NAFLD. The area under the curve (AUC) and Delong test were used to compare the differences in the ability to identify NAFLD risk of each obesity indicator in both sexes and different age subgroups of both sexes, and the optimal diagnostic threshold for each indicator was calculated using Youden’s index.", "In the current study, we used data from 14,251 medical examiners recruited by the NAGALA cohort between 1994 and 2016 [23]. NAGALA is an ongoing longitudinal cohort study initiated by Murakami Memorial Hospital in 1994, using physical examination data from the general population to assess common risk factors for the development of chronic diseases; a detailed description of the NAGALA cohort study design has been previously published and the study data have been uploaded by Professor Okamura to the Dryad database for public access [24]. For the current study, we excluded from the original data set (1) subjects who were diagnosed or self-reported with hepatitis (viral/alcoholic) or diabetes and impaired fasting glucose at baseline (n = 1,547); (2) subjects with alcohol abuse (n = 1,952, males consuming ≥ 210 g of alcohol per week and females consuming ≥ 140 g) [25]; (3) subjects taking any medications at baseline (n = 2,321); and, (4) subjects with missing anthropometric and medical examination data (n = 873) were also excluded. We ultimately included 14,251 eligible subjects, with the inclusion and exclusion criteria shown in Fig. 1. The NAGALA cohort study has been ethically reviewed by the Murakami Memorial Hospital Institutional Ethics Review Committee (IRB2018-09-01) and all subjects signed written informed consent for the use of their data; The current study was a post-hoc analysis of the NAGALA cohort, and the Jiangxi Provincial People’s Hospital Ethics Review Committee reviewed the study design and granted approval (IRB2021-066).\n\nFig. 1Flowchart of the selection process of study subjects\n\nFlowchart of the selection process of study subjects\nIn this study, we extracted the following data from each subject: anthropometric and demographic information including weight, height, sex, age, WC, BMI, ABSI, smoking and drinking status, and exercise habits of the subjects; where drinking status was classified according to the subject’s weekly alcohol consumption in the previous month as moderate drinking, light drinking and never or small drinking; smoking status was defined as never, previous and current smoking; for exercise habits, subjects were considered to have exercise habits if they had at least one physical activity of any form per week. BMI was calculated as weight (kg)/[height (m)]2 and ABSI was calculated as 1,000*WC (m)*[weight (kg)] – 2/3*[height (m)]5/6 [22].\nData on laboratory tests, including blood pressure, triglycerides (TG), aspartate aminotransferase (AST), total cholesterol (TC), fasting blood glucose (FPG), gamma-glutamyl transferase (GGT), high-density lipoprotein cholesterol (HDL-C), alanine aminotransferase (ALT), and glycated hemoglobin (HbA1c). The specific steps for obtaining demographic and anthropometric information and data from laboratory tests have been described in detail elsewhere [19]; all procedures in this study fully followed the Declaration of Helsinki. See STROBE statement (S1 Text).", "Based on previous studies, the present study included four combination types of BMI with WC and ABSI: (1) Using the WHO-recommended clinical cut-offs of WC and BMI combined into four obesity phenotypes; (2) constructing the optimal proportional combination of BMI with WC and ABSI, OBMI+WC and OBMI+ABSI; and (3) using the multiplicative combination of WC and ABSI with BMI, BMI*WC, and BMI*ABSI; (4) construction of anthropometric risk index [ARI (BMI, ABSI)] for combined risk of BMI and ABSI using the method recommended by Krakauer NY et al. [26].", "In accordance with the World Health Organization (WHO) expert committee cut-off recommendations for Asian populations, we defined BMI ≥ 25 kg/m2 as overweight/obese and BMI < 25 kg/m2 as normal weight [27]. We defined WC ≥ 85 cm for males or WC ≥ 80 cm for females as central obesity and WC < 85 cm for males or WC < 80 cm for females as normal WC [28]. All subjects were assigned to the following four obesity phenotypes according to the baseline WC and BMI: (1) BMIN/WCN: normal weight + normal WC; (2) BMIO/WCN: overweight/obese + normal WC; (3) BMIN/WCO: normal weight + central obesity; (4) BMIO/WCO: overweight/obese + central obesity.", "The optimal proportional combination indicator OBMI+WC was calculated as OBMI+WC= nWC*WC + nBMI*BMI. The optimal proportion coefficients for WC (nWC) and BMI (nBMI) were calculated as nWC = βWC / (βBMI + βWC) and nBMI = βBMI / (βBMI + βWC), respectively; where βWC and βBMI were the regression coefficients of WC and BMI in the multivariable logistic regression model, respectively. To acquire regression coefficients for WC and BMI, we included the presence or absence of NAFLD as the outcome event of interest, WC and BMI as variables in a multivariate-adjusted logistic regression model, and age, height, ALT, AST, systolic blood pressure (SBP), TG, GGT, HDL-C, HbA1c, TC, FPG, and drinking status were adjusted as covariates. Similarly, we used the same method to obtain OBMI+ABSI. Since NAFLD risk was positively correlated with BMI, WC, and ABSI, and there was no correlation between BMI and ABSI, which is consistent with the construction of an ARI (BMI, ABSI) for the combined risk of BMI and ABSI, and we calculated ARI (BMI, ABSI) using the method provided by Krakauer NY et al. [26]. In addition, we calculated the products BMI*WC and BMI* ABSI for further analysis.", "Specialized gastroenterologists, blinded to the subjects scored each abdominal ultrasound for NAFLD risk based on four criteria: (1) changes in the intensity of liver blood flow signals (0–4 points); (2) the clarity of liver blood vessels (0–1 points); (3) liver and kidney echo contrast (0–4 points); (4) deep liver echo attenuation (0–2 points). NAFLD was diagnosed if the sum of the above four scores was greater than 2 [29].", "We performed all statistical analysis steps in the current study using Empower (R) version 2.2 and R language version 3.4.3. Significance was defined as P < 0.05 in all comparisons (two-sided). Given the significant differences in body composition, fat deposition patterns, and prevalence of NAFLD between males and females [30], all analyses in the present study were performed separately for female and male subjects and the specific steps were as follows:\nFirst, we calculated the quartile categories of BMI*ABSI using the quartile function and subsequently described baseline information by grouping all subjects based on the quartile categories of BMI*ABSI and whether they had NAFLD. For the measurement data, QQ plots were first used to determine the pattern of data distribution, and the data with a normal and a skewed distribution were described as mean (standard deviation) and median (interquartile range), respectively. T-test or one-way ANOVA was used for group comparisons of normal data, and Mann-Whitney rank-sum test or Kruskal-Wallis rank-sum test was used for group comparisons of skewed data. Count data were described as frequencies (%), using the Pearson chi-square test for comparison between groups.\nSecond, before exploring the association of each obesity indicator with NAFLD, we first analyzed the correlations between several anthropometric measures (height, BMI, WC, ABSI), and it is worth mentioning that a combination of two basic anthropometric indicators of obesity which have smaller correlations is considered more appropriate. Additionally, we also performed a collinearity screening for all covariates [31], where the weight and diastolic blood pressure (DBP) had variance inflation factors (VIF) greater than 5 and were therefore treated as collinear variables and not included in the subsequent model adjustment (Supplementary Table 1). We developed one univariate and three multivariable logistic regression models for assessing the associations between the continuous variables BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and the categorical variable obesity phenotypes and risk of NAFLD, and recorded the corresponding Odds ratio (OR) and 95% confidence interval (CI), respectively. Due to the large numerical span of BMI*WC (689.56-6490.01) and BMI*ABSI (961.13-3630.94), the change in effect value caused by the change of one unit was small, so the BMI*WC and BMI*ABSI were Z-score transformed and included in the regression models. In the model adjustment of multivariable logistic regression, model 1 considered the effect of anthropometric parameters of age and height on NAFLD; model 2 adjusted drinking status and important lipid parameters (TC, TG, HDL-C) on the basis of model 1; model 3 further considered the effects of liver enzyme indicators (ALT, AST, GGT), blood glucose and blood pressure parameters (FPG, HbA1c, SBP) on the basis of model 2. In addition, to further explore the correlation between BMI*ABSI and NAFLD, we also included BMI*ABSI as a categorical variable in the logistic regression models and calculated the trend of the association between the median of each category of BMI*ABSI and NAFLD.\nThird, based on the results of the correlation analysis between each obesity indicator and NAFLD, we also constructed receiver operating characteristic (ROC) curves to evaluate the ability of continuous obesity indicator BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC to identify the risk of NAFLD. The area under the curve (AUC) and Delong test were used to compare the differences in the ability to identify NAFLD risk of each obesity indicator in both sexes and different age subgroups of both sexes, and the optimal diagnostic threshold for each indicator was calculated using Youden’s index.", "The study included 14,251 subjects, of whom 7,411 (52%) were males, with a mean age of 43.82 (8.99) years, and 2,029 (27.37%) were diagnosed with NAFLD; 6,840 were (48%) females, with mean age 43.22 (8.78) years, and 478 (6.99%) were diagnosed with NAFLD. Table 1 is stratified by gender and describes the differences in baseline characteristics between NAFLD patients and non-NAFLD subjects. We found that NAFLD patients consistently had higher age, weight, TC, SBP, DBP, HbA1c, BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, ALT, AST, GGT, FPG, and TG levels in both sexes, but with lower HDL-C levels. Notably, non-NAFLD subjects of both sexes tended to have greater alcohol consumption, which paradoxically appeared to be a protective factor for NAFLD. Additionally, the comparison between the NAFLD group and the non-NAFLD group showed that physical exercise habits were only significantly different in males, height was only significantly different in females, and smoking habits were not significantly different in both sexes.\n\nTable 1Characteristics of 14,251 subjects stratified by NAFLD and genderMaleFemaleVariablesNon-NAFLDn = 5382 (72.61%)NAFLDn = 2029 (27.39%)P-valueNon-NAFLDn = 6362 (93.01%)NAFLDn = 478 (6.99%)P-valueAge, years43.7 (9.3)44.1 (8.2)0.00242.9 (8.7)47.6 (8.3)< 0.001Height, cm170.9 (6.0)170.6 (5.9)0.084158.4 (5.4)157.0 (5.3)< 0.001Weight, kg64.6 (8.3)74.3 (10.6)< 0.00151.9 (7.1)63.2 (10.0)< 0.001TC, mmol/L5.1 (0.8)5.4 (0.9)< 0.0015.1 (0.9)5.6 (0.9)< 0.001HDL-C, mmol/L1.3 (0.3)1.1 (0.3)< 0.0011.7 (0.4)1.4 (0.3)< 0.001SBP, mmHg116.0 (13.2)124.0 (14.5)< 0.001108.4 (13.8)120.7 (16.0)< 0.001FPG, mmol/L5.3 (0.4)5.4 (0.3)< 0.0015.0 (0.4)5.3 (0.4)< 0.001DBP, mmHg72.9 (9.3)78.4 (10.1)< 0.00167.0 (9.5)75.1 (10.2)< 0.001HbA1c, %5.1 (0.3)5.3 (0.3)< 0.0015.2 (0.3)5.4 (0.3)< 0.001BMI, kg/m222.1 (2.4)25.5 (3.0)< 0.00120.7 (2.6)25.6 (3.6)< 0.001WC, cm78.0 (6.8)86.6 (7.4)< 0.00170.8 (7.3)83.3 (8.9)< 0.001ABSI75.8 (3.3)76.7 (3.1)< 0.00174.8 (4.6)76.8 (4.5)< 0.001ARI13.1 (0.9)14.3 (1.0)< 0.00112.2 (1.0)14.0 (1.3)< 0.001OBMI+WC34.4 (3.2)38.9 (3.8)< 0.00131.7 (3.4)38.3 (4.5)< 0.001OBMI+ABSI32.0 (2.1)34.9 (2.4)< 0.00128.9 (2.3)33.4 (3.0)< 0.001BMI*WC1714.2 (1507.7-1939.1)2152.9 (1919.2-2457.9)< 0.0011423.8 (1249.1-1645.8)2070.2 (1795.3-2396.7)< 0.001BMI*ABSI1669.3 (1538.1–1806.0)1922.5 (1795.9-2082.3)< 0.0011518.4 (1390.8-1673.5)1917.8 (1756.7-2131.4)< 0.001ALT, U/L18.0 (14.0–23.0)29.0 (22.0–41.0)< 0.00113.0 (11.0–17.0)19.0 (15.0–26.0)< 0.001AST, U/L17.0 (14.0–21.0)21.0 (17.0–26.0)< 0.00116.0 (13.0–19.0)18.0 (15.0–22.0)< 0.001GGT, U/L17.0 (14.0–24.0)24.0 (18.0–35.0)< 0.00112.0 (9.0–14.0)15.0 (12.0–20.0)< 0.001TG, mmol/L0.8 (0.6–1.2)1.3 (0.9–1.9)< 0.0010.5 (0.4–0.8)1.0 (0.7–1.4)< 0.001Habit of exercise, No. (%)< 0.0010.335No4300 (79.9%)1720 (84.8%)5351 (84.1%)410 (85.8%)Yes1082 (20.1%)309 (15.2%)1011 (15.9%)68 (14.2%)Drinking status, No. (%)< 0.0010.004Non or small3731 (69.3%)1623 (80.0%)5986 (94.1%)465 (97.3%)Light1096 (20.4%)273 (13.5%)376 (5.9%)13 (2.7%)Moderate555 (10.3%)133 (6.6%)Smoking status, No. (%)0.0670.664Non1952 (36.3%)758 (37.4%)5609 (88.2%)427 (89.3%)Past1538 (28.6%)615 (30.3%)382 (6.0%)24 (5.0%)Current1892 (35.2%)656 (32.3%)371 (5.8%)27 (5.6%)Values were expressed as mean (standard deviation) or medians (quartile interval) or n (%). Abbreviations: BMI: body mass index; WC: waist circumference; ALT: alanine aminotransferase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FPG: fasting plasma glucose; HbA1c: hemoglobin A1c; SBP: systolic blood pressure; DBP: diastolic blood pressure; NAFLD: non-alcoholic fatty liver disease; ABSI: A body shape index; OBMI+ABSI: Optimal proportional combination of BMI and ABSI; OBMI+WC: Optimal proportional combination of BMI and WC; ARI: Anthropometric risk index\n\nCharacteristics of 14,251 subjects stratified by NAFLD and gender\nValues were expressed as mean (standard deviation) or medians (quartile interval) or n (%). Abbreviations: BMI: body mass index; WC: waist circumference; ALT: alanine aminotransferase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FPG: fasting plasma glucose; HbA1c: hemoglobin A1c; SBP: systolic blood pressure; DBP: diastolic blood pressure; NAFLD: non-alcoholic fatty liver disease; ABSI: A body shape index; OBMI+ABSI: Optimal proportional combination of BMI and ABSI; OBMI+WC: Optimal proportional combination of BMI and WC; ARI: Anthropometric risk index\nBaseline characteristics for all subjects according to the quartile category of BMI*ABSI are shown in Table 2. We found that with the increase of BMI*ABSI quartiles, all baseline metrics of subjects showed a trend of change (all P < 0.001). Of these, a greater proportion of male subjects were in the higher BMI*ABSI quartiles, with a male-to-female ratio of almost 7:3 in Q4, and there were more subjects with smoking and drinking habits and with less physical exercise in Q4. Moreover, subjects’ HDL-C levels decreased with increasing BMI*ABSI quartiles, while age, BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*WC, height, weight, TC, SBP, HbA1c, ALT, AST, DBP, FPG, GGT, TG levels, and the prevalence of NAFLD increased (Q4: 48.5%).\n\nTable 2Characteristics of 14,251 subjects stratified according to the BMI*ABSI quartilesBMI*ABSI quartilesvariablesQ1 (961.1-1477.2)Q2 (1477.4-1645.5)Q3 (1645.6-1827.4)Q4 (1827.5-3630.9)P-valueNo. of subjects3563356235633563Male, No. (%)867 (24.3%)1666 (46.8%)2269 (63.7%)2609 (73.2%)Age, years41.0 (8.7)43.1 (8.7)44.5 (8.6)45.6 (8.9)< 0.001BMI, kg/m218.7 (1.3)20.9 (1.2)22.7 (1.2)26.0 (2.5)< 0.001WC, cm65.4 (3.7)72.8 (2.9)78.8 (2.9)87.7 (5.8)< 0.001ABSI73.1 (3.8)75.1 (3.8)76.5 (3.5)77.5 (3.5)< 0.001ARI11.4 (0.4)12.4 (0.3)13.2 (0.3)14.4 (0.8)< 0.001OBMI+WC29.0 (1.5)32.3 (1.0)35.0 (1.1)39.6 (3.0)< 0.001OBMI+ABSI27.4 (1.3)29.9 (1.1)31.9 (1.1)35.0 (2.0)< 0.001BMI*WC1241.2 (1147.1-1318.4)1514.2 (1447.2-1587.4)1782.6 (1703.2-1865.8)2194.4 (2051.7-2436.9)< 0.001Height, cm161.7 (7.7)164.3 (8.3)166.1 (8.5)167.1 (8.4)< 0.001Weight, kg49.0 (5.7)56.4 (6.5)62.7 (7.3)72.9 (10.3)< 0.001TC, mmol/L4.9 (0.8)5.0 (0.9)5.2 (0.9)5.4 (0.9)< 0.001HDL-C, mmol/L1.7 (0.39)1.5 (0.4)1.4 (0.4)1.2 (0.3)< 0.001SBP, mmHg105.6 (12.5)111.1 (12.8)115.9 (13.3)123.1 (14.8)< 0.001FPG, mmol/L4.9 (0.4)5.1 (0.4)5.2 (0.4)5.3 (0.4)< 0.001DBP, mmHg65.5 (8.5)69.0 (9.1)72.5 (9.6)77.6 (10.2)< 0.001HbA1c, %5.1 (0.3)5.1 (0.3)5.2 (0.3)5.3 (0.3)< 0.001ALT, U/L14.0 (11.0–17.0)15.0 (12.0–19.0)18.0 (13.0–23.0)23.0 (17.0–33.0)< 0.001AST, U/L16.0 (13.0–19.0)16.00 (14.0–20.0)17.0 (14.0–21.0)19.0 (16.0–24.0)< 0.001GGT, U/L12.0 (10.0–15.0)13.00 (11.0–18.0)16.0 (12.0–22.0)21.0 (15.0–30.0)< 0.001TG, mmol/L0.51 (0.4–0.7)0.6 (0.5–0.9)0.8 (0.6–1.2)1.1 (0.8–1.6)< 0.001Habit of exercise, No. (%)< 0.001No2963 (83.2%)2858 (80.2%)2920 (82.0%)3040 (85.3%)Yes600 (16.8%)704 (19.8%)643 (18.1%)523 (14.7%)Drinking status, No. (%)< 0.001Non or small3228 (90.6%)2937 (82.5%)2848 (79.9%)2792 (78.4%)Light279 (7.8%)467 (13.1%)507 (14.2%)505 (14.2%)Moderate56 (1.6%)158 (4.4%)208 (5.8%)266 (7.5%)Smoking status, No. (%)< 0.001Non2736 (76.8%)2322 (65.2%)1957 (54.9%)1731 (48.6%)Past337 (9.5%)560 (15.7%)785 (22.0%)877 (24.6%)Current490 (13.8%)680 (19.1%)821 (23.0%)955 (26.8%)NAFLD, No. (%)16 (0.5%)150 (4.2%)611 (17.2%)1730 (48.5%)< 0.001Abbreviations as in Table 1\n\nCharacteristics of 14,251 subjects stratified according to the BMI*ABSI quartiles\nAbbreviations as in Table 1", "In the logistic regression equation constructed to calculate the optimal proportional combination OBMI+WC, the regression coefficient βBMI for BMI was 0.218 (0.154–0.282), and βWC for WC was 0.063 (0.038–0.089) in females; in males, βBMI was 0.177 (0.124–0.299), and βWC was 0.068 (0.048–0.089). Therefore, according to the calculation formula of the optimal proportional coefficients, the values ​​of nBMI and nWC in females were 0.776 and 0.224, respectively, and the values ​​of nBMI and nWC in males were 0.722 and 0.278, respectively. Similarly, in the logistic regression equation constructed to calculate the optimal proportional combination OBMI+ABSI, βBMI and βABSI were 0.356 and 0.064, respectively, in females, resulting in nBMI and nABSI values ​​of 0.848 and 0.152, respectively; in males, βBMI and βABSI were 0.334 and 0.075, respectively, and nBMI and nABSI values ​​were 0.817 and 0.183, respectively. Table 3 shows the calculation formulas of OBMI+WC and OBMI+ABSI.\n\nTable 3Optimal combination equations of BMI with WC and ABSI.Logistic regression derived equationsMaleFemaleequations for BMI and WC0.722BMI + 0.278WC0.776BMI + 0.224WCequations for BMI and ABSI0.817BMI + 0.183ABSI0.848BMI + 0.152ABSIAbbreviations as in Table 1\n\nOptimal combination equations of BMI with WC and ABSI.\nAbbreviations as in Table 1", "Correlation analysis of the basic anthropometric measures showed a strong correlation between BMI and WC in both sexes (r: male 0.8796; female 0.8191), while BMI was barely correlated with ABSI (r < 0.05) (Supplementary Table 2). In contrast, the combination of ABSI and BMI may be more appropriate. Table 4 presents the associations between BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC and the risk of NAFLD in both sexes. In multivariate-adjusted logistic regression models, BMI and WC, and ABSI were positively correlated with NAFLD risk. In the current study, the combined indicators ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC were positively correlated with NAFLD risk in all four logistic regression models, and the OR values ​​in the univariate logistic regression model ranged from 1.44 to 5.02; after further adjustment for age, height, GGT, TC, AST, HDL-C, drinking status, ALT, TG, FPG, HbA1c, and SBP (Model 3), the results did not change significantly. The risk of NAFLD increased by 33% (OR 1.33, 95%CI 1.29, 1.37) and 52% (OR 1.52, 95%CI 1.45, 1.60) and 172% (OR 2.72, 95%CI 2.43, 3.04), in females, for each unit increase in OBMI+WC and OBMI+ABSI and ARI respectively. Similarly, in males, NAFLD risk was increased by 30% (OR 1.30, 95%CI 1.27, 1.33) and 50% (OR 1.50, 95%CI 1.45, 1.56) and 172% (OR 2.72, 95%CI 2.48, 2.98), respectively. Each standard deviation increase in BMI*WC and BMI*ABSI in females was associated with a 213% (OR 3.13, 95%CI 2.74,3,56) and 209% (OR 3.09, 95%CI 2.72, 3.52) increased risk for NAFLD respectively, and for males 185% (OR 2.85, 95%CI 2.59, 3.15) and 197% (OR 2.97, 95%CI 2.68, 3.28), respectively. Furthermore, the association between BMI*ABSI and NAFLD remained unchanged after treating BMI*ABSI as a categorical variable; taking the first quartile as the control group, the NAFLD risk increased with the increase of BMI*ABSI quartiles (all P for trend < 0.001). Moreover, we also analyzed the risk of NAFLD for each obesity phenotype in multivariable logistic regression models (Supplementary Table 3), which in model 3 showed that compared with the BMIN/WCN phenotype, male and female BMIO/ WCN, BMIN/WCO, and BMIO/WCO phenotypes all had a significantly higher risk of NAFLD. The risk of NAFLD associated with the BMIO/ WCN, BMIN/WCO, and BMIO/WCO phenotypes had ORs of 3.67, 3.77, and 9.78 in females and 2.76, 2.74, and 4.19 in males, respectively. Clearly, subjects with the BMIO/WCO phenotype consistently had the highest risk of developing NAFLD.\n\nTable 4Odds ratios and 95% confidence intervals of BMI, WC, ABSI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and ARI for NAFLD in female and male subjectsOdds ratios (95% confidence interval)Crude modelModel 1Model 2Model 3FemaleBMI1.58 (1.53, 1.64)*1.58 (1.52, 1.63)*1.49 (1.44, 1.55)*1.42 (1.36, 1.47)*WC1.19 (1.17, 1.21)*1.19 (1.17, 1.21)*1.17 (1.15, 1.18)*1.14 (1.12, 1.16)*ABSI1.09 (1.07, 1.12)*1.07 (1.05, 1.09)*1.06 (1.04, 1.08)*1.05 (1.02, 1.07)*ARI3.70 (3.36, 4.07)*3.61 (3.28, 3.98)*3.11 (2.80, 3.44)*2.72 (2.43, 3.04)*OBMI+ABSI1.73 (1.66, 1.80)*1.71 (1.65, 1.79)*1.61 (1.54, 1.68)*1.52 (1.45, 1.60)*OBMI+WC1.44 (1.41, 1.48)*1.44 (1.40, 1.48)*1.38 (1.34, 1.42)*1.33 (1.29, 1.37)*BMI*ABSI (Per SD)4.49 (4.02, 5.02)*4.33 (3.87, 4.84)*3.63 (3.23, 4.09)*3.09 (2.72, 3.52)*BMI*WC (Per SD)4.50 (4.03, 5.03)*4.41 (3.94, 4.93)*3.68 (3.27, 4.15)*3.13 (2.74, 3.56)*BMI*ABSI (quartiles)Quartile 11.01.01.01.0Quartile 210.90 (4.64, 25.59)*10.32 (4.39, 24.24)*8.42 (3.57, 19.86)*7.47 (3.16, 17.65)*Quartile 343.73 (19.20, 99.63)*39.09 (17.13, 89.20)*25.14 (10.94, 57.76)*19.49 (8.44, 44.98)*Quartile 4217.88 (96.72, 490.84)*188.51 (83.50, 425.59)*105.60 (46.43, 240.20)*63.97 (27.87, 146.85)*P for trend< 0.0001< 0.0001< 0.0001< 0.0001MaleBMI1.62 (1.58, 1.67)*1.62 (1.58, 1.67)*1.52 (1.48, 1.57)*1.38 (1.34, 1.42)*WC1.19 (1.18, 1.20)*1.21 (1.20, 1.22)*1.18 (1.17, 1.19)*1.13 (1.12, 1.15)*ABSI1.09 (1.07, 1.10)*1.10 (1.08, 1.12)*1.06 (1.04, 1.08)*1.05 (1.02, 1.07)*ARI4.39 (4.06, 4.75)*4.40 (4.07, 4.77)*3.68 (3.38, 4.00)*2.72 (2.48, 2.98)*OBMI+ABSI1.83 (1.77, 1.89)*1.83 (1.77, 1.89)*1.70 (1.65, 1.76)*1.50 (1.45, 1.56)*OBMI+WC1.47 (1.44, 1.50)*1.47 (1.44, 1.50)*1.41 (1.38, 1.44)*1.30 (1.27, 1.33)*BMI*ABSI (Per SD)5.02 (4.61, 5.48)*5.08 (4.66, 5.55)*4.16 (3.80, 4.56)*2.97 (2.68, 3.28)*BMI*WC (Per SD)4.72 (4.34, 5.13)*4.83 (4.44, 5.26)*3.95 (3.62, 4.32)*2.85 (2.59, 3.15)*BMI*ABSI (quartiles)Quartile 11.01.01.01.0Quartile 25.76 (3.00, 11.09)*5.89 (3.06, 11.33)*4.68 (2.42, 9.02)*3.97 (2.03, 7.78)*Quartile 323.97 (12.75, 45.07)*25.00 (13.29, 47.03)*15.83 (8.38, 29.90)*10.27 (5.35, 19.72)*Quartile 4102.03 (54.44, 191.23)*108.71 (57.95, 203.93)*58.44 (30.99, 110.21)*26.15 (13.61, 50.25)*P for trend< 0.0001< 0.0001< 0.0001< 0.0001*P < 0.0001; Abbreviation: SD: standard deviation; other abbreviations as in Table 1Model 1 adjusted for age and heightModel 2 adjusted for model 1 plus TC, TG, HDL-C, drinking statusModel 3 adjusted for model 2 plus ALT, AST, GGT, FPG, HbA1c, and SBP.\n\nOdds ratios and 95% confidence intervals of BMI, WC, ABSI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and ARI for NAFLD in female and male subjects\n*P < 0.0001; Abbreviation: SD: standard deviation; other abbreviations as in Table 1\nModel 1 adjusted for age and height\nModel 2 adjusted for model 1 plus TC, TG, HDL-C, drinking status\nModel 3 adjusted for model 2 plus ALT, AST, GGT, FPG, HbA1c, and SBP.", "ROC curves were drawn to assess the accuracy of BMI, WC, ABSI, ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC for identifying NAFLD in both sexes (Fig. 2). Table 5 summarizes the AUC and optimal diagnostic thresholds for these obesity indicators. In contrast, ARI and OBMI+ABSI had the same and highest AUC in both female and male subjects, with AUC and optimal diagnostic thresholds of 0.8270, 33.2569 and 0.8912, 30.4143 for ARI in males and females, respectively. The Delong test further showed that both the combined indicators of BMI and WC (OBMI+WC and BMI*WC) and BMI and ABSI (OBMI+ABSI and BMI*ABSI and ARI) were significantly better at identifying the risk of NAFLD than BMI or WC or ABSI alone (Delong test P < 0.05).\n\nFig. 2ROC curve analysis of BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC in females and males\n\nROC curve analysis of BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC in females and males\n\nTable 5The best threshold, sensitivities, specificities, Youden’s index, and area under the curve of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI in different gendersAUC95%CI low95%CI upBest thresholdSpecificitySensitivityYouden’s indexFemaleBMI**0.87990.86480.895022.75650.81770.79500.6127WC**0.86950.85450.884474.25000.70500.87450.5804ABSI**ARI**0.61710.89120.59160.87750.64260.904875.792512.78250.59260.77350.59620.85770.18880.6312OBMI+ABSI**0.89120.87750.904830.41430.77330.85770.6310OBMI+WC0.88880.87500.902734.12340.77160.85560.6272BMI*ABSI0.88840.87480.90211704.21000.79220.82850.6207BMI*WC0.88880.87500.90271674.11830.77330.85560.6286MaleBMI**0.81600.80550.826423.55550.73820.73090.4691WC**0.81020.79980.820780.65000.66740.80340.4708ABSI**0.57950.56530.593674.91280.40520.72400.1292ARI**0.82700.81700.836913.60960.74560.74570.4913OBMI+ABSI**0.82700.81700.836933.25690.74580.74570.4915OBMI+WC0.82420.81410.834339.83130.74380.74470.4885BMI*ABSI0.82570.81570.83561758.75900.67610.81170.4878BMI*WC0.82440.81430.83451944.04330.75510.73290.4880Abbreviations as in Table 1**P < 0.05 compared with BMI*WC in each gender\n\nThe best threshold, sensitivities, specificities, Youden’s index, and area under the curve of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI in different genders\nABSI**\nARI**\n0.6171\n0.8912\n0.5916\n0.8775\n0.6426\n0.9048\n75.7925\n12.7825\n0.5926\n0.7735\n0.5962\n0.8577\n0.1888\n0.6312\nAbbreviations as in Table 1\n**P < 0.05 compared with BMI*WC in each gender\nTo explore the changes in BMI, WC, ABSI, ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC on the risk identification ability of NAFLD in different age subgroups of both sexes, we constructed ROC curves for 20–39, 40–59, and ≥ 60 years in both sexes, respectively; and the corresponding AUC and optimal diagnostic thresholds were summarized in Table 6. We found that ARI and OBMI+ABSI exhibited considerably higher AUCs in all age subgroups for both sexes except in the female ≥ 60-year age group; the AUC of OBMI+ABSI in the female 20–39 age group was 0.9523, the highest value among all age subgroups of both sexes. In the male 20-59-year-old group and the female 40-59-year-old group, the AUC values ​​of ARI and OBMI+ABSI were slightly higher than those of BMI*WC, but all were significantly higher than those of BMI or WC or ABSI alone, while in the female 20–39 years group and the female ≥ 60 years group AUC values for all combined obesity indicators were significantly higher than that of WC and ABSI; in addition, in the male ≥ 60-year-old group, the seven indicators of obesity had similar NAFLD identification abilities, only ABSI underperformed. Therefore, the combined indicators ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC can significantly improve the ability of simple obesity parameters BMI and WC and ABSI to identify NAFLD in middle-aged females and young and middle-aged males and had the highest diagnostic performance in young females.\n\nTable 6The area under the curve, best threshold, sensitivities, specificities, and Youden’s index of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI as a predictor of NAFLD in the different age groups of both gendersAUC95%CI low95%CI upBest thresholdSpecificitySensitivityYouden’s indexFemaleAge 20–39 yearsBMI0.95110.93360.968523.12660.89810.87230.7704WC***0.93190.90790.955976.60000.85600.88300.7390ABSI***0.56860.50850.628875.57420.61220.53190.1441ARI0.95220.93370.970812.79220.84980.93620.7860BMI*WC0.95160.93310.97011675.47840.84360.92550.7691BMI*ABSI0.94860.92890.96821701.34860.86140.92550.7869OBMI+ABSI0.95230.93370.970830.43680.84980.93620.7860OBMI+WC0.95160.93300.970134.12960.84240.92550.7679Age 40–59 yearsBMI***0.84840.82820.868622.75270.78410.77440.5585WC***0.84230.82300.861674.25000.66300.86910.5321ABSI***0.61860.58970.647675.79250.58080.60720.1880ARI***0.86220.84380.880513.02830.80770.77440.5821BMI*WC0.86030.84170.87881674.11830.73320.83840.5716BMI*ABSI0.86020.84200.87851704.21000.75460.81340.5680OBMI+ABSI***0.86220.84380.880530.99950.80790.77440.5823OBMI+WC0.86020.84170.878835.03630.80930.76320.5725Age ≥ 60 yearsBMI0.83080.74580.915923.33340.84390.76000.6039WC***0.76960.67780.861379.75000.77070.68000.4507ABSI***0.56600.45510.677078.18360.50240.72000.2224ARI0.81950.73910.899912.92520.70240.84000.5424BMI*WC0.81990.73960.90021686.12800.69760.84000.5376BMI*ABSI0.80140.71860.88411659.09940.56100.96000.5210OBMI+ABSI0.81950.73920.899830.75250.70240.84000.5424OBMI+WC0.82010.73930.900934.33930.70240.84000.5424MaleAge 20–39 yearsBMI***0.84390.82750.860223.47820.74950.76650.5160WC***0.84150.82550.857580.45000.72190.80090.5228ABSI***0.60630.58260.630074.50900.48710.69910.1862ARI***0.85490.83960.870213.50260.76000.78440.5444BMI*WC0.85130.83570.86691862.54150.72190.81140.5333BMI*ABSI***0.85520.83990.87041766.28860.75570.78740.5431OBMI+ABSI***0.85490.83960.870233.00760.76140.78290.5443OBMI+WC0.85140.83580.866939.14530.72900.80840.5374Age 40–59 yearsBMI***0.80070.78670.814723.25050.67790.76390.4418WC***0.79080.77660.805181.25000.65830.77950.4378ABSI***0.56150.54290.580175.42940.38970.70050.0902ARI***0.81040.79690.824013.61420.72320.74510.4683BMI*WC0.80800.79430.82171955.12030.74480.71770.4625BMI*ABSI0.80860.79500.82221816.10160.73340.72870.4621OBMI+ABSI***0.81050.79690.824033.27330.72390.74430.4682OBMI+WC0.80750.79380.821240.11920.74750.71850.4660Age ≥ 60 yearsBMI0.79030.73630.844423.74200.81110.69510.5062WC0.78040.72820.832680.50000.59440.86590.4603ABSI***0.58040.51190.649075.85420.29410.86590.1600ARI0.79490.74370.846213.58190.67800.81710.4951BMI*WC0.80290.75130.85451932.61210.73680.79270.5295BMI*ABSI0.79040.73930.84141800.36700.65940.82930.4887OBMI+ABSI0.79500.74380.846233.18800.67800.81710.4951OBMI+WC0.80230.75090.853639.74760.71210.81710.5292Abbreviations as in Table 1***P < 0.05 compared with BMI*WC in each age group of both genders\n\nThe area under the curve, best threshold, sensitivities, specificities, and Youden’s index of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI as a predictor of NAFLD in the different age groups of both genders\nAbbreviations as in Table 1\n***P < 0.05 compared with BMI*WC in each age group of both genders", "Several advantages of the current study need to be mentioned: (1) The current study demonstrated for the first time in a large sample of the general population that BMI combined with WC and ABSI had a stronger ability to identify NAFLD than a basic anthropometric obesity parameter. (2) After combining BMI with WC and ABSI in various ways, we found that the ARI and multiplicative combination indicators (BMI*WC and BMI*ABSI) may be the more appropriate obesity index for clinical screening of NAFLD. These findings provided an important reference for the application of BMI combined with the abdominal obesity index for NAFLD screening in the general population.\nOf course, it is undeniable that the current study has several limitations: (1) The current study was a cross-sectional study, so we can only demonstrate that BMI combined with the abdominal obesity index was stronger than a single indicator for identifying NAFLD in the general population, but whether the predictive performance for NAFLD risk is stronger needs to be validated in a longitudinal cohort study. (2) The diagnosis of NAFLD, the main outcome in this study, was based on ultrasonography, which may have missed some patients with NAFLD [1]. However, in the setting of fewer NAFLD cases, we still demonstrated that BMI combined with the abdominal obesity index had a higher diagnostic performance for NAFLD than a single indicator. (3) Although we systematically adjusted for known NAFLD risk factors, there may be residual confounding due to the limitations of retrospective studies where some NAFLD risk factors could not be measured and obtained.", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2" ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Data sources and study design", "Definition and derivation of the combined indicators of BMI with WC and ABSI", "Combination using usual cut-offs for BMI and WC", "Optimal proportional combinations OBMI+WC and OBMI+ABSI, multiplicative combination indicators BMI*ABSI and BMI*WC, and ARI", "Diagnosis of NAFLD", "Statistical analysis", "Results", "Demographic and clinical characteristics of subjects", "Calculation of the optimal proportional combination of BMI with WC and ABSI", "Association of various obesity indicators with NAFLD", "Accuracy of various obesity indicators to identify NAFLD", "Discussion", "Advantages and limitations of research", "Conclusion", "Electronic supplementary material", "" ]
[ "Non-alcoholic fatty liver disease (NAFLD) is a chronic non-infectious liver disease characterized by oxidative stress, inflammation, and fibrosis in hepatocytes among people without a history of excessive alcohol consumption [1, 2]. NAFLD can present as asymptomatic simple hepatic steatosis in its early stages, and without intervention, oxidative stress, inflammation, and fibrosis in hepatocytes can contribute to the continuous progression of NAFLD to non-alcoholic steatohepatitis and then to cirrhosis and ultimately to hepatocellular carcinoma [2, 3]. Recent epidemiological surveys have shown that about a quarter of the global population suffers from NAFLD, with a 27.4% estimated prevalence in Asia [4]. NAFLD is not only the most common chronic liver disease worldwide but has replaced hepatitis B virus infection as the primary risk factor for advanced liver diseases such as cirrhosis and hepatocellular carcinoma [5]. The high prevalence of NAFLD and its severe complications place a heavy burden on humans and the world’s health care systems, making the early prevention, identification, and intervention of NAFLD of great public health importance.\nObesity is one of the most important risk factors for NAFLD [6]. Waist circumference (WC) and body mass index (BMI) are currently the most widely used basic body measures for the assessment of general and central obesity and also are known risk factors for NAFLD [7–10]. Both BMI and WC alone, however, have some distinct limitations, such as the inability of BMI alone to distinguish between adipose and muscle tissue and the inability of WC to determine the differential contribution of abdominal visceral adipose tissue and abdominal subcutaneous adipose tissue to central obesity [11–13]. It is not clear if combining risk associated with BMI and WC can improve the individual risk association. Several relevant studies have analyzed the performance of the combination of BMI and WC in disease risk assessment, and overall, combining BMI and WC in different ways significantly improved the predictive efficacy for obesity-related diseases such as obesity-related hypertension, type 2 diabetes, cardiovascular disease, and all-cause mortality risk compared to a single indicator [14–17]. In addition, a recent study by Wang et al. also found that high levels of WC and BMI were both associated with NAFLD risk in people with type 2 diabetes and that BMI combined with WC was superior to the single measures in assessing the risk of NAFLD [18]. What remains unclear, however, is whether the combination of BMI and WC best identifies a higher risk of NAFLD in the general population. Furthermore, it is important to note that there is a strong correlation between BMI and WC (r ≈ 0.80), and directly combining BMI with WC has the potential to confound the association between BMI and NAFLD, leading to biased and possibly misleading risk estimates [19–21]. A Body Shape Index (ABSI), an allometric abdominal obesity index calculated from WC, height, and weight, is intended to be independent of BMI and has proven to be useful for identifying individuals at risk for sarcopenic obesity [22]; therefore, we also evaluated combining BMI with the non-correlated abdominal obesity index ABSI. This study sought to explore risk assessment for NAFLD of various combinations of BMI and WC or ABSI in a cohort of general health examinees from NAGALA.", "[SUBTITLE] Data sources and study design [SUBSECTION] In the current study, we used data from 14,251 medical examiners recruited by the NAGALA cohort between 1994 and 2016 [23]. NAGALA is an ongoing longitudinal cohort study initiated by Murakami Memorial Hospital in 1994, using physical examination data from the general population to assess common risk factors for the development of chronic diseases; a detailed description of the NAGALA cohort study design has been previously published and the study data have been uploaded by Professor Okamura to the Dryad database for public access [24]. For the current study, we excluded from the original data set (1) subjects who were diagnosed or self-reported with hepatitis (viral/alcoholic) or diabetes and impaired fasting glucose at baseline (n = 1,547); (2) subjects with alcohol abuse (n = 1,952, males consuming ≥ 210 g of alcohol per week and females consuming ≥ 140 g) [25]; (3) subjects taking any medications at baseline (n = 2,321); and, (4) subjects with missing anthropometric and medical examination data (n = 873) were also excluded. We ultimately included 14,251 eligible subjects, with the inclusion and exclusion criteria shown in Fig. 1. The NAGALA cohort study has been ethically reviewed by the Murakami Memorial Hospital Institutional Ethics Review Committee (IRB2018-09-01) and all subjects signed written informed consent for the use of their data; The current study was a post-hoc analysis of the NAGALA cohort, and the Jiangxi Provincial People’s Hospital Ethics Review Committee reviewed the study design and granted approval (IRB2021-066).\n\nFig. 1Flowchart of the selection process of study subjects\n\nFlowchart of the selection process of study subjects\nIn this study, we extracted the following data from each subject: anthropometric and demographic information including weight, height, sex, age, WC, BMI, ABSI, smoking and drinking status, and exercise habits of the subjects; where drinking status was classified according to the subject’s weekly alcohol consumption in the previous month as moderate drinking, light drinking and never or small drinking; smoking status was defined as never, previous and current smoking; for exercise habits, subjects were considered to have exercise habits if they had at least one physical activity of any form per week. BMI was calculated as weight (kg)/[height (m)]2 and ABSI was calculated as 1,000*WC (m)*[weight (kg)] – 2/3*[height (m)]5/6 [22].\nData on laboratory tests, including blood pressure, triglycerides (TG), aspartate aminotransferase (AST), total cholesterol (TC), fasting blood glucose (FPG), gamma-glutamyl transferase (GGT), high-density lipoprotein cholesterol (HDL-C), alanine aminotransferase (ALT), and glycated hemoglobin (HbA1c). The specific steps for obtaining demographic and anthropometric information and data from laboratory tests have been described in detail elsewhere [19]; all procedures in this study fully followed the Declaration of Helsinki. See STROBE statement (S1 Text).\nIn the current study, we used data from 14,251 medical examiners recruited by the NAGALA cohort between 1994 and 2016 [23]. NAGALA is an ongoing longitudinal cohort study initiated by Murakami Memorial Hospital in 1994, using physical examination data from the general population to assess common risk factors for the development of chronic diseases; a detailed description of the NAGALA cohort study design has been previously published and the study data have been uploaded by Professor Okamura to the Dryad database for public access [24]. For the current study, we excluded from the original data set (1) subjects who were diagnosed or self-reported with hepatitis (viral/alcoholic) or diabetes and impaired fasting glucose at baseline (n = 1,547); (2) subjects with alcohol abuse (n = 1,952, males consuming ≥ 210 g of alcohol per week and females consuming ≥ 140 g) [25]; (3) subjects taking any medications at baseline (n = 2,321); and, (4) subjects with missing anthropometric and medical examination data (n = 873) were also excluded. We ultimately included 14,251 eligible subjects, with the inclusion and exclusion criteria shown in Fig. 1. The NAGALA cohort study has been ethically reviewed by the Murakami Memorial Hospital Institutional Ethics Review Committee (IRB2018-09-01) and all subjects signed written informed consent for the use of their data; The current study was a post-hoc analysis of the NAGALA cohort, and the Jiangxi Provincial People’s Hospital Ethics Review Committee reviewed the study design and granted approval (IRB2021-066).\n\nFig. 1Flowchart of the selection process of study subjects\n\nFlowchart of the selection process of study subjects\nIn this study, we extracted the following data from each subject: anthropometric and demographic information including weight, height, sex, age, WC, BMI, ABSI, smoking and drinking status, and exercise habits of the subjects; where drinking status was classified according to the subject’s weekly alcohol consumption in the previous month as moderate drinking, light drinking and never or small drinking; smoking status was defined as never, previous and current smoking; for exercise habits, subjects were considered to have exercise habits if they had at least one physical activity of any form per week. BMI was calculated as weight (kg)/[height (m)]2 and ABSI was calculated as 1,000*WC (m)*[weight (kg)] – 2/3*[height (m)]5/6 [22].\nData on laboratory tests, including blood pressure, triglycerides (TG), aspartate aminotransferase (AST), total cholesterol (TC), fasting blood glucose (FPG), gamma-glutamyl transferase (GGT), high-density lipoprotein cholesterol (HDL-C), alanine aminotransferase (ALT), and glycated hemoglobin (HbA1c). The specific steps for obtaining demographic and anthropometric information and data from laboratory tests have been described in detail elsewhere [19]; all procedures in this study fully followed the Declaration of Helsinki. See STROBE statement (S1 Text).\n[SUBTITLE] Definition and derivation of the combined indicators of BMI with WC and ABSI [SUBSECTION] Based on previous studies, the present study included four combination types of BMI with WC and ABSI: (1) Using the WHO-recommended clinical cut-offs of WC and BMI combined into four obesity phenotypes; (2) constructing the optimal proportional combination of BMI with WC and ABSI, OBMI+WC and OBMI+ABSI; and (3) using the multiplicative combination of WC and ABSI with BMI, BMI*WC, and BMI*ABSI; (4) construction of anthropometric risk index [ARI (BMI, ABSI)] for combined risk of BMI and ABSI using the method recommended by Krakauer NY et al. [26].\nBased on previous studies, the present study included four combination types of BMI with WC and ABSI: (1) Using the WHO-recommended clinical cut-offs of WC and BMI combined into four obesity phenotypes; (2) constructing the optimal proportional combination of BMI with WC and ABSI, OBMI+WC and OBMI+ABSI; and (3) using the multiplicative combination of WC and ABSI with BMI, BMI*WC, and BMI*ABSI; (4) construction of anthropometric risk index [ARI (BMI, ABSI)] for combined risk of BMI and ABSI using the method recommended by Krakauer NY et al. [26].\n[SUBTITLE] Combination using usual cut-offs for BMI and WC [SUBSECTION] In accordance with the World Health Organization (WHO) expert committee cut-off recommendations for Asian populations, we defined BMI ≥ 25 kg/m2 as overweight/obese and BMI < 25 kg/m2 as normal weight [27]. We defined WC ≥ 85 cm for males or WC ≥ 80 cm for females as central obesity and WC < 85 cm for males or WC < 80 cm for females as normal WC [28]. All subjects were assigned to the following four obesity phenotypes according to the baseline WC and BMI: (1) BMIN/WCN: normal weight + normal WC; (2) BMIO/WCN: overweight/obese + normal WC; (3) BMIN/WCO: normal weight + central obesity; (4) BMIO/WCO: overweight/obese + central obesity.\nIn accordance with the World Health Organization (WHO) expert committee cut-off recommendations for Asian populations, we defined BMI ≥ 25 kg/m2 as overweight/obese and BMI < 25 kg/m2 as normal weight [27]. We defined WC ≥ 85 cm for males or WC ≥ 80 cm for females as central obesity and WC < 85 cm for males or WC < 80 cm for females as normal WC [28]. All subjects were assigned to the following four obesity phenotypes according to the baseline WC and BMI: (1) BMIN/WCN: normal weight + normal WC; (2) BMIO/WCN: overweight/obese + normal WC; (3) BMIN/WCO: normal weight + central obesity; (4) BMIO/WCO: overweight/obese + central obesity.\n[SUBTITLE] Optimal proportional combinations OBMI+WC and OBMI+ABSI, multiplicative combination indicators BMI*ABSI and BMI*WC, and ARI [SUBSECTION] The optimal proportional combination indicator OBMI+WC was calculated as OBMI+WC= nWC*WC + nBMI*BMI. The optimal proportion coefficients for WC (nWC) and BMI (nBMI) were calculated as nWC = βWC / (βBMI + βWC) and nBMI = βBMI / (βBMI + βWC), respectively; where βWC and βBMI were the regression coefficients of WC and BMI in the multivariable logistic regression model, respectively. To acquire regression coefficients for WC and BMI, we included the presence or absence of NAFLD as the outcome event of interest, WC and BMI as variables in a multivariate-adjusted logistic regression model, and age, height, ALT, AST, systolic blood pressure (SBP), TG, GGT, HDL-C, HbA1c, TC, FPG, and drinking status were adjusted as covariates. Similarly, we used the same method to obtain OBMI+ABSI. Since NAFLD risk was positively correlated with BMI, WC, and ABSI, and there was no correlation between BMI and ABSI, which is consistent with the construction of an ARI (BMI, ABSI) for the combined risk of BMI and ABSI, and we calculated ARI (BMI, ABSI) using the method provided by Krakauer NY et al. [26]. In addition, we calculated the products BMI*WC and BMI* ABSI for further analysis.\nThe optimal proportional combination indicator OBMI+WC was calculated as OBMI+WC= nWC*WC + nBMI*BMI. The optimal proportion coefficients for WC (nWC) and BMI (nBMI) were calculated as nWC = βWC / (βBMI + βWC) and nBMI = βBMI / (βBMI + βWC), respectively; where βWC and βBMI were the regression coefficients of WC and BMI in the multivariable logistic regression model, respectively. To acquire regression coefficients for WC and BMI, we included the presence or absence of NAFLD as the outcome event of interest, WC and BMI as variables in a multivariate-adjusted logistic regression model, and age, height, ALT, AST, systolic blood pressure (SBP), TG, GGT, HDL-C, HbA1c, TC, FPG, and drinking status were adjusted as covariates. Similarly, we used the same method to obtain OBMI+ABSI. Since NAFLD risk was positively correlated with BMI, WC, and ABSI, and there was no correlation between BMI and ABSI, which is consistent with the construction of an ARI (BMI, ABSI) for the combined risk of BMI and ABSI, and we calculated ARI (BMI, ABSI) using the method provided by Krakauer NY et al. [26]. In addition, we calculated the products BMI*WC and BMI* ABSI for further analysis.\n[SUBTITLE] Diagnosis of NAFLD [SUBSECTION] Specialized gastroenterologists, blinded to the subjects scored each abdominal ultrasound for NAFLD risk based on four criteria: (1) changes in the intensity of liver blood flow signals (0–4 points); (2) the clarity of liver blood vessels (0–1 points); (3) liver and kidney echo contrast (0–4 points); (4) deep liver echo attenuation (0–2 points). NAFLD was diagnosed if the sum of the above four scores was greater than 2 [29].\nSpecialized gastroenterologists, blinded to the subjects scored each abdominal ultrasound for NAFLD risk based on four criteria: (1) changes in the intensity of liver blood flow signals (0–4 points); (2) the clarity of liver blood vessels (0–1 points); (3) liver and kidney echo contrast (0–4 points); (4) deep liver echo attenuation (0–2 points). NAFLD was diagnosed if the sum of the above four scores was greater than 2 [29].\n[SUBTITLE] Statistical analysis [SUBSECTION] We performed all statistical analysis steps in the current study using Empower (R) version 2.2 and R language version 3.4.3. Significance was defined as P < 0.05 in all comparisons (two-sided). Given the significant differences in body composition, fat deposition patterns, and prevalence of NAFLD between males and females [30], all analyses in the present study were performed separately for female and male subjects and the specific steps were as follows:\nFirst, we calculated the quartile categories of BMI*ABSI using the quartile function and subsequently described baseline information by grouping all subjects based on the quartile categories of BMI*ABSI and whether they had NAFLD. For the measurement data, QQ plots were first used to determine the pattern of data distribution, and the data with a normal and a skewed distribution were described as mean (standard deviation) and median (interquartile range), respectively. T-test or one-way ANOVA was used for group comparisons of normal data, and Mann-Whitney rank-sum test or Kruskal-Wallis rank-sum test was used for group comparisons of skewed data. Count data were described as frequencies (%), using the Pearson chi-square test for comparison between groups.\nSecond, before exploring the association of each obesity indicator with NAFLD, we first analyzed the correlations between several anthropometric measures (height, BMI, WC, ABSI), and it is worth mentioning that a combination of two basic anthropometric indicators of obesity which have smaller correlations is considered more appropriate. Additionally, we also performed a collinearity screening for all covariates [31], where the weight and diastolic blood pressure (DBP) had variance inflation factors (VIF) greater than 5 and were therefore treated as collinear variables and not included in the subsequent model adjustment (Supplementary Table 1). We developed one univariate and three multivariable logistic regression models for assessing the associations between the continuous variables BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and the categorical variable obesity phenotypes and risk of NAFLD, and recorded the corresponding Odds ratio (OR) and 95% confidence interval (CI), respectively. Due to the large numerical span of BMI*WC (689.56-6490.01) and BMI*ABSI (961.13-3630.94), the change in effect value caused by the change of one unit was small, so the BMI*WC and BMI*ABSI were Z-score transformed and included in the regression models. In the model adjustment of multivariable logistic regression, model 1 considered the effect of anthropometric parameters of age and height on NAFLD; model 2 adjusted drinking status and important lipid parameters (TC, TG, HDL-C) on the basis of model 1; model 3 further considered the effects of liver enzyme indicators (ALT, AST, GGT), blood glucose and blood pressure parameters (FPG, HbA1c, SBP) on the basis of model 2. In addition, to further explore the correlation between BMI*ABSI and NAFLD, we also included BMI*ABSI as a categorical variable in the logistic regression models and calculated the trend of the association between the median of each category of BMI*ABSI and NAFLD.\nThird, based on the results of the correlation analysis between each obesity indicator and NAFLD, we also constructed receiver operating characteristic (ROC) curves to evaluate the ability of continuous obesity indicator BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC to identify the risk of NAFLD. The area under the curve (AUC) and Delong test were used to compare the differences in the ability to identify NAFLD risk of each obesity indicator in both sexes and different age subgroups of both sexes, and the optimal diagnostic threshold for each indicator was calculated using Youden’s index.\nWe performed all statistical analysis steps in the current study using Empower (R) version 2.2 and R language version 3.4.3. Significance was defined as P < 0.05 in all comparisons (two-sided). Given the significant differences in body composition, fat deposition patterns, and prevalence of NAFLD between males and females [30], all analyses in the present study were performed separately for female and male subjects and the specific steps were as follows:\nFirst, we calculated the quartile categories of BMI*ABSI using the quartile function and subsequently described baseline information by grouping all subjects based on the quartile categories of BMI*ABSI and whether they had NAFLD. For the measurement data, QQ plots were first used to determine the pattern of data distribution, and the data with a normal and a skewed distribution were described as mean (standard deviation) and median (interquartile range), respectively. T-test or one-way ANOVA was used for group comparisons of normal data, and Mann-Whitney rank-sum test or Kruskal-Wallis rank-sum test was used for group comparisons of skewed data. Count data were described as frequencies (%), using the Pearson chi-square test for comparison between groups.\nSecond, before exploring the association of each obesity indicator with NAFLD, we first analyzed the correlations between several anthropometric measures (height, BMI, WC, ABSI), and it is worth mentioning that a combination of two basic anthropometric indicators of obesity which have smaller correlations is considered more appropriate. Additionally, we also performed a collinearity screening for all covariates [31], where the weight and diastolic blood pressure (DBP) had variance inflation factors (VIF) greater than 5 and were therefore treated as collinear variables and not included in the subsequent model adjustment (Supplementary Table 1). We developed one univariate and three multivariable logistic regression models for assessing the associations between the continuous variables BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and the categorical variable obesity phenotypes and risk of NAFLD, and recorded the corresponding Odds ratio (OR) and 95% confidence interval (CI), respectively. Due to the large numerical span of BMI*WC (689.56-6490.01) and BMI*ABSI (961.13-3630.94), the change in effect value caused by the change of one unit was small, so the BMI*WC and BMI*ABSI were Z-score transformed and included in the regression models. In the model adjustment of multivariable logistic regression, model 1 considered the effect of anthropometric parameters of age and height on NAFLD; model 2 adjusted drinking status and important lipid parameters (TC, TG, HDL-C) on the basis of model 1; model 3 further considered the effects of liver enzyme indicators (ALT, AST, GGT), blood glucose and blood pressure parameters (FPG, HbA1c, SBP) on the basis of model 2. In addition, to further explore the correlation between BMI*ABSI and NAFLD, we also included BMI*ABSI as a categorical variable in the logistic regression models and calculated the trend of the association between the median of each category of BMI*ABSI and NAFLD.\nThird, based on the results of the correlation analysis between each obesity indicator and NAFLD, we also constructed receiver operating characteristic (ROC) curves to evaluate the ability of continuous obesity indicator BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC to identify the risk of NAFLD. The area under the curve (AUC) and Delong test were used to compare the differences in the ability to identify NAFLD risk of each obesity indicator in both sexes and different age subgroups of both sexes, and the optimal diagnostic threshold for each indicator was calculated using Youden’s index.", "In the current study, we used data from 14,251 medical examiners recruited by the NAGALA cohort between 1994 and 2016 [23]. NAGALA is an ongoing longitudinal cohort study initiated by Murakami Memorial Hospital in 1994, using physical examination data from the general population to assess common risk factors for the development of chronic diseases; a detailed description of the NAGALA cohort study design has been previously published and the study data have been uploaded by Professor Okamura to the Dryad database for public access [24]. For the current study, we excluded from the original data set (1) subjects who were diagnosed or self-reported with hepatitis (viral/alcoholic) or diabetes and impaired fasting glucose at baseline (n = 1,547); (2) subjects with alcohol abuse (n = 1,952, males consuming ≥ 210 g of alcohol per week and females consuming ≥ 140 g) [25]; (3) subjects taking any medications at baseline (n = 2,321); and, (4) subjects with missing anthropometric and medical examination data (n = 873) were also excluded. We ultimately included 14,251 eligible subjects, with the inclusion and exclusion criteria shown in Fig. 1. The NAGALA cohort study has been ethically reviewed by the Murakami Memorial Hospital Institutional Ethics Review Committee (IRB2018-09-01) and all subjects signed written informed consent for the use of their data; The current study was a post-hoc analysis of the NAGALA cohort, and the Jiangxi Provincial People’s Hospital Ethics Review Committee reviewed the study design and granted approval (IRB2021-066).\n\nFig. 1Flowchart of the selection process of study subjects\n\nFlowchart of the selection process of study subjects\nIn this study, we extracted the following data from each subject: anthropometric and demographic information including weight, height, sex, age, WC, BMI, ABSI, smoking and drinking status, and exercise habits of the subjects; where drinking status was classified according to the subject’s weekly alcohol consumption in the previous month as moderate drinking, light drinking and never or small drinking; smoking status was defined as never, previous and current smoking; for exercise habits, subjects were considered to have exercise habits if they had at least one physical activity of any form per week. BMI was calculated as weight (kg)/[height (m)]2 and ABSI was calculated as 1,000*WC (m)*[weight (kg)] – 2/3*[height (m)]5/6 [22].\nData on laboratory tests, including blood pressure, triglycerides (TG), aspartate aminotransferase (AST), total cholesterol (TC), fasting blood glucose (FPG), gamma-glutamyl transferase (GGT), high-density lipoprotein cholesterol (HDL-C), alanine aminotransferase (ALT), and glycated hemoglobin (HbA1c). The specific steps for obtaining demographic and anthropometric information and data from laboratory tests have been described in detail elsewhere [19]; all procedures in this study fully followed the Declaration of Helsinki. See STROBE statement (S1 Text).", "Based on previous studies, the present study included four combination types of BMI with WC and ABSI: (1) Using the WHO-recommended clinical cut-offs of WC and BMI combined into four obesity phenotypes; (2) constructing the optimal proportional combination of BMI with WC and ABSI, OBMI+WC and OBMI+ABSI; and (3) using the multiplicative combination of WC and ABSI with BMI, BMI*WC, and BMI*ABSI; (4) construction of anthropometric risk index [ARI (BMI, ABSI)] for combined risk of BMI and ABSI using the method recommended by Krakauer NY et al. [26].", "In accordance with the World Health Organization (WHO) expert committee cut-off recommendations for Asian populations, we defined BMI ≥ 25 kg/m2 as overweight/obese and BMI < 25 kg/m2 as normal weight [27]. We defined WC ≥ 85 cm for males or WC ≥ 80 cm for females as central obesity and WC < 85 cm for males or WC < 80 cm for females as normal WC [28]. All subjects were assigned to the following four obesity phenotypes according to the baseline WC and BMI: (1) BMIN/WCN: normal weight + normal WC; (2) BMIO/WCN: overweight/obese + normal WC; (3) BMIN/WCO: normal weight + central obesity; (4) BMIO/WCO: overweight/obese + central obesity.", "The optimal proportional combination indicator OBMI+WC was calculated as OBMI+WC= nWC*WC + nBMI*BMI. The optimal proportion coefficients for WC (nWC) and BMI (nBMI) were calculated as nWC = βWC / (βBMI + βWC) and nBMI = βBMI / (βBMI + βWC), respectively; where βWC and βBMI were the regression coefficients of WC and BMI in the multivariable logistic regression model, respectively. To acquire regression coefficients for WC and BMI, we included the presence or absence of NAFLD as the outcome event of interest, WC and BMI as variables in a multivariate-adjusted logistic regression model, and age, height, ALT, AST, systolic blood pressure (SBP), TG, GGT, HDL-C, HbA1c, TC, FPG, and drinking status were adjusted as covariates. Similarly, we used the same method to obtain OBMI+ABSI. Since NAFLD risk was positively correlated with BMI, WC, and ABSI, and there was no correlation between BMI and ABSI, which is consistent with the construction of an ARI (BMI, ABSI) for the combined risk of BMI and ABSI, and we calculated ARI (BMI, ABSI) using the method provided by Krakauer NY et al. [26]. In addition, we calculated the products BMI*WC and BMI* ABSI for further analysis.", "Specialized gastroenterologists, blinded to the subjects scored each abdominal ultrasound for NAFLD risk based on four criteria: (1) changes in the intensity of liver blood flow signals (0–4 points); (2) the clarity of liver blood vessels (0–1 points); (3) liver and kidney echo contrast (0–4 points); (4) deep liver echo attenuation (0–2 points). NAFLD was diagnosed if the sum of the above four scores was greater than 2 [29].", "We performed all statistical analysis steps in the current study using Empower (R) version 2.2 and R language version 3.4.3. Significance was defined as P < 0.05 in all comparisons (two-sided). Given the significant differences in body composition, fat deposition patterns, and prevalence of NAFLD between males and females [30], all analyses in the present study were performed separately for female and male subjects and the specific steps were as follows:\nFirst, we calculated the quartile categories of BMI*ABSI using the quartile function and subsequently described baseline information by grouping all subjects based on the quartile categories of BMI*ABSI and whether they had NAFLD. For the measurement data, QQ plots were first used to determine the pattern of data distribution, and the data with a normal and a skewed distribution were described as mean (standard deviation) and median (interquartile range), respectively. T-test or one-way ANOVA was used for group comparisons of normal data, and Mann-Whitney rank-sum test or Kruskal-Wallis rank-sum test was used for group comparisons of skewed data. Count data were described as frequencies (%), using the Pearson chi-square test for comparison between groups.\nSecond, before exploring the association of each obesity indicator with NAFLD, we first analyzed the correlations between several anthropometric measures (height, BMI, WC, ABSI), and it is worth mentioning that a combination of two basic anthropometric indicators of obesity which have smaller correlations is considered more appropriate. Additionally, we also performed a collinearity screening for all covariates [31], where the weight and diastolic blood pressure (DBP) had variance inflation factors (VIF) greater than 5 and were therefore treated as collinear variables and not included in the subsequent model adjustment (Supplementary Table 1). We developed one univariate and three multivariable logistic regression models for assessing the associations between the continuous variables BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and the categorical variable obesity phenotypes and risk of NAFLD, and recorded the corresponding Odds ratio (OR) and 95% confidence interval (CI), respectively. Due to the large numerical span of BMI*WC (689.56-6490.01) and BMI*ABSI (961.13-3630.94), the change in effect value caused by the change of one unit was small, so the BMI*WC and BMI*ABSI were Z-score transformed and included in the regression models. In the model adjustment of multivariable logistic regression, model 1 considered the effect of anthropometric parameters of age and height on NAFLD; model 2 adjusted drinking status and important lipid parameters (TC, TG, HDL-C) on the basis of model 1; model 3 further considered the effects of liver enzyme indicators (ALT, AST, GGT), blood glucose and blood pressure parameters (FPG, HbA1c, SBP) on the basis of model 2. In addition, to further explore the correlation between BMI*ABSI and NAFLD, we also included BMI*ABSI as a categorical variable in the logistic regression models and calculated the trend of the association between the median of each category of BMI*ABSI and NAFLD.\nThird, based on the results of the correlation analysis between each obesity indicator and NAFLD, we also constructed receiver operating characteristic (ROC) curves to evaluate the ability of continuous obesity indicator BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC to identify the risk of NAFLD. The area under the curve (AUC) and Delong test were used to compare the differences in the ability to identify NAFLD risk of each obesity indicator in both sexes and different age subgroups of both sexes, and the optimal diagnostic threshold for each indicator was calculated using Youden’s index.", "[SUBTITLE] Demographic and clinical characteristics of subjects [SUBSECTION] The study included 14,251 subjects, of whom 7,411 (52%) were males, with a mean age of 43.82 (8.99) years, and 2,029 (27.37%) were diagnosed with NAFLD; 6,840 were (48%) females, with mean age 43.22 (8.78) years, and 478 (6.99%) were diagnosed with NAFLD. Table 1 is stratified by gender and describes the differences in baseline characteristics between NAFLD patients and non-NAFLD subjects. We found that NAFLD patients consistently had higher age, weight, TC, SBP, DBP, HbA1c, BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, ALT, AST, GGT, FPG, and TG levels in both sexes, but with lower HDL-C levels. Notably, non-NAFLD subjects of both sexes tended to have greater alcohol consumption, which paradoxically appeared to be a protective factor for NAFLD. Additionally, the comparison between the NAFLD group and the non-NAFLD group showed that physical exercise habits were only significantly different in males, height was only significantly different in females, and smoking habits were not significantly different in both sexes.\n\nTable 1Characteristics of 14,251 subjects stratified by NAFLD and genderMaleFemaleVariablesNon-NAFLDn = 5382 (72.61%)NAFLDn = 2029 (27.39%)P-valueNon-NAFLDn = 6362 (93.01%)NAFLDn = 478 (6.99%)P-valueAge, years43.7 (9.3)44.1 (8.2)0.00242.9 (8.7)47.6 (8.3)< 0.001Height, cm170.9 (6.0)170.6 (5.9)0.084158.4 (5.4)157.0 (5.3)< 0.001Weight, kg64.6 (8.3)74.3 (10.6)< 0.00151.9 (7.1)63.2 (10.0)< 0.001TC, mmol/L5.1 (0.8)5.4 (0.9)< 0.0015.1 (0.9)5.6 (0.9)< 0.001HDL-C, mmol/L1.3 (0.3)1.1 (0.3)< 0.0011.7 (0.4)1.4 (0.3)< 0.001SBP, mmHg116.0 (13.2)124.0 (14.5)< 0.001108.4 (13.8)120.7 (16.0)< 0.001FPG, mmol/L5.3 (0.4)5.4 (0.3)< 0.0015.0 (0.4)5.3 (0.4)< 0.001DBP, mmHg72.9 (9.3)78.4 (10.1)< 0.00167.0 (9.5)75.1 (10.2)< 0.001HbA1c, %5.1 (0.3)5.3 (0.3)< 0.0015.2 (0.3)5.4 (0.3)< 0.001BMI, kg/m222.1 (2.4)25.5 (3.0)< 0.00120.7 (2.6)25.6 (3.6)< 0.001WC, cm78.0 (6.8)86.6 (7.4)< 0.00170.8 (7.3)83.3 (8.9)< 0.001ABSI75.8 (3.3)76.7 (3.1)< 0.00174.8 (4.6)76.8 (4.5)< 0.001ARI13.1 (0.9)14.3 (1.0)< 0.00112.2 (1.0)14.0 (1.3)< 0.001OBMI+WC34.4 (3.2)38.9 (3.8)< 0.00131.7 (3.4)38.3 (4.5)< 0.001OBMI+ABSI32.0 (2.1)34.9 (2.4)< 0.00128.9 (2.3)33.4 (3.0)< 0.001BMI*WC1714.2 (1507.7-1939.1)2152.9 (1919.2-2457.9)< 0.0011423.8 (1249.1-1645.8)2070.2 (1795.3-2396.7)< 0.001BMI*ABSI1669.3 (1538.1–1806.0)1922.5 (1795.9-2082.3)< 0.0011518.4 (1390.8-1673.5)1917.8 (1756.7-2131.4)< 0.001ALT, U/L18.0 (14.0–23.0)29.0 (22.0–41.0)< 0.00113.0 (11.0–17.0)19.0 (15.0–26.0)< 0.001AST, U/L17.0 (14.0–21.0)21.0 (17.0–26.0)< 0.00116.0 (13.0–19.0)18.0 (15.0–22.0)< 0.001GGT, U/L17.0 (14.0–24.0)24.0 (18.0–35.0)< 0.00112.0 (9.0–14.0)15.0 (12.0–20.0)< 0.001TG, mmol/L0.8 (0.6–1.2)1.3 (0.9–1.9)< 0.0010.5 (0.4–0.8)1.0 (0.7–1.4)< 0.001Habit of exercise, No. (%)< 0.0010.335No4300 (79.9%)1720 (84.8%)5351 (84.1%)410 (85.8%)Yes1082 (20.1%)309 (15.2%)1011 (15.9%)68 (14.2%)Drinking status, No. (%)< 0.0010.004Non or small3731 (69.3%)1623 (80.0%)5986 (94.1%)465 (97.3%)Light1096 (20.4%)273 (13.5%)376 (5.9%)13 (2.7%)Moderate555 (10.3%)133 (6.6%)Smoking status, No. (%)0.0670.664Non1952 (36.3%)758 (37.4%)5609 (88.2%)427 (89.3%)Past1538 (28.6%)615 (30.3%)382 (6.0%)24 (5.0%)Current1892 (35.2%)656 (32.3%)371 (5.8%)27 (5.6%)Values were expressed as mean (standard deviation) or medians (quartile interval) or n (%). Abbreviations: BMI: body mass index; WC: waist circumference; ALT: alanine aminotransferase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FPG: fasting plasma glucose; HbA1c: hemoglobin A1c; SBP: systolic blood pressure; DBP: diastolic blood pressure; NAFLD: non-alcoholic fatty liver disease; ABSI: A body shape index; OBMI+ABSI: Optimal proportional combination of BMI and ABSI; OBMI+WC: Optimal proportional combination of BMI and WC; ARI: Anthropometric risk index\n\nCharacteristics of 14,251 subjects stratified by NAFLD and gender\nValues were expressed as mean (standard deviation) or medians (quartile interval) or n (%). Abbreviations: BMI: body mass index; WC: waist circumference; ALT: alanine aminotransferase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FPG: fasting plasma glucose; HbA1c: hemoglobin A1c; SBP: systolic blood pressure; DBP: diastolic blood pressure; NAFLD: non-alcoholic fatty liver disease; ABSI: A body shape index; OBMI+ABSI: Optimal proportional combination of BMI and ABSI; OBMI+WC: Optimal proportional combination of BMI and WC; ARI: Anthropometric risk index\nBaseline characteristics for all subjects according to the quartile category of BMI*ABSI are shown in Table 2. We found that with the increase of BMI*ABSI quartiles, all baseline metrics of subjects showed a trend of change (all P < 0.001). Of these, a greater proportion of male subjects were in the higher BMI*ABSI quartiles, with a male-to-female ratio of almost 7:3 in Q4, and there were more subjects with smoking and drinking habits and with less physical exercise in Q4. Moreover, subjects’ HDL-C levels decreased with increasing BMI*ABSI quartiles, while age, BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*WC, height, weight, TC, SBP, HbA1c, ALT, AST, DBP, FPG, GGT, TG levels, and the prevalence of NAFLD increased (Q4: 48.5%).\n\nTable 2Characteristics of 14,251 subjects stratified according to the BMI*ABSI quartilesBMI*ABSI quartilesvariablesQ1 (961.1-1477.2)Q2 (1477.4-1645.5)Q3 (1645.6-1827.4)Q4 (1827.5-3630.9)P-valueNo. of subjects3563356235633563Male, No. (%)867 (24.3%)1666 (46.8%)2269 (63.7%)2609 (73.2%)Age, years41.0 (8.7)43.1 (8.7)44.5 (8.6)45.6 (8.9)< 0.001BMI, kg/m218.7 (1.3)20.9 (1.2)22.7 (1.2)26.0 (2.5)< 0.001WC, cm65.4 (3.7)72.8 (2.9)78.8 (2.9)87.7 (5.8)< 0.001ABSI73.1 (3.8)75.1 (3.8)76.5 (3.5)77.5 (3.5)< 0.001ARI11.4 (0.4)12.4 (0.3)13.2 (0.3)14.4 (0.8)< 0.001OBMI+WC29.0 (1.5)32.3 (1.0)35.0 (1.1)39.6 (3.0)< 0.001OBMI+ABSI27.4 (1.3)29.9 (1.1)31.9 (1.1)35.0 (2.0)< 0.001BMI*WC1241.2 (1147.1-1318.4)1514.2 (1447.2-1587.4)1782.6 (1703.2-1865.8)2194.4 (2051.7-2436.9)< 0.001Height, cm161.7 (7.7)164.3 (8.3)166.1 (8.5)167.1 (8.4)< 0.001Weight, kg49.0 (5.7)56.4 (6.5)62.7 (7.3)72.9 (10.3)< 0.001TC, mmol/L4.9 (0.8)5.0 (0.9)5.2 (0.9)5.4 (0.9)< 0.001HDL-C, mmol/L1.7 (0.39)1.5 (0.4)1.4 (0.4)1.2 (0.3)< 0.001SBP, mmHg105.6 (12.5)111.1 (12.8)115.9 (13.3)123.1 (14.8)< 0.001FPG, mmol/L4.9 (0.4)5.1 (0.4)5.2 (0.4)5.3 (0.4)< 0.001DBP, mmHg65.5 (8.5)69.0 (9.1)72.5 (9.6)77.6 (10.2)< 0.001HbA1c, %5.1 (0.3)5.1 (0.3)5.2 (0.3)5.3 (0.3)< 0.001ALT, U/L14.0 (11.0–17.0)15.0 (12.0–19.0)18.0 (13.0–23.0)23.0 (17.0–33.0)< 0.001AST, U/L16.0 (13.0–19.0)16.00 (14.0–20.0)17.0 (14.0–21.0)19.0 (16.0–24.0)< 0.001GGT, U/L12.0 (10.0–15.0)13.00 (11.0–18.0)16.0 (12.0–22.0)21.0 (15.0–30.0)< 0.001TG, mmol/L0.51 (0.4–0.7)0.6 (0.5–0.9)0.8 (0.6–1.2)1.1 (0.8–1.6)< 0.001Habit of exercise, No. (%)< 0.001No2963 (83.2%)2858 (80.2%)2920 (82.0%)3040 (85.3%)Yes600 (16.8%)704 (19.8%)643 (18.1%)523 (14.7%)Drinking status, No. (%)< 0.001Non or small3228 (90.6%)2937 (82.5%)2848 (79.9%)2792 (78.4%)Light279 (7.8%)467 (13.1%)507 (14.2%)505 (14.2%)Moderate56 (1.6%)158 (4.4%)208 (5.8%)266 (7.5%)Smoking status, No. (%)< 0.001Non2736 (76.8%)2322 (65.2%)1957 (54.9%)1731 (48.6%)Past337 (9.5%)560 (15.7%)785 (22.0%)877 (24.6%)Current490 (13.8%)680 (19.1%)821 (23.0%)955 (26.8%)NAFLD, No. (%)16 (0.5%)150 (4.2%)611 (17.2%)1730 (48.5%)< 0.001Abbreviations as in Table 1\n\nCharacteristics of 14,251 subjects stratified according to the BMI*ABSI quartiles\nAbbreviations as in Table 1\nThe study included 14,251 subjects, of whom 7,411 (52%) were males, with a mean age of 43.82 (8.99) years, and 2,029 (27.37%) were diagnosed with NAFLD; 6,840 were (48%) females, with mean age 43.22 (8.78) years, and 478 (6.99%) were diagnosed with NAFLD. Table 1 is stratified by gender and describes the differences in baseline characteristics between NAFLD patients and non-NAFLD subjects. We found that NAFLD patients consistently had higher age, weight, TC, SBP, DBP, HbA1c, BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, ALT, AST, GGT, FPG, and TG levels in both sexes, but with lower HDL-C levels. Notably, non-NAFLD subjects of both sexes tended to have greater alcohol consumption, which paradoxically appeared to be a protective factor for NAFLD. Additionally, the comparison between the NAFLD group and the non-NAFLD group showed that physical exercise habits were only significantly different in males, height was only significantly different in females, and smoking habits were not significantly different in both sexes.\n\nTable 1Characteristics of 14,251 subjects stratified by NAFLD and genderMaleFemaleVariablesNon-NAFLDn = 5382 (72.61%)NAFLDn = 2029 (27.39%)P-valueNon-NAFLDn = 6362 (93.01%)NAFLDn = 478 (6.99%)P-valueAge, years43.7 (9.3)44.1 (8.2)0.00242.9 (8.7)47.6 (8.3)< 0.001Height, cm170.9 (6.0)170.6 (5.9)0.084158.4 (5.4)157.0 (5.3)< 0.001Weight, kg64.6 (8.3)74.3 (10.6)< 0.00151.9 (7.1)63.2 (10.0)< 0.001TC, mmol/L5.1 (0.8)5.4 (0.9)< 0.0015.1 (0.9)5.6 (0.9)< 0.001HDL-C, mmol/L1.3 (0.3)1.1 (0.3)< 0.0011.7 (0.4)1.4 (0.3)< 0.001SBP, mmHg116.0 (13.2)124.0 (14.5)< 0.001108.4 (13.8)120.7 (16.0)< 0.001FPG, mmol/L5.3 (0.4)5.4 (0.3)< 0.0015.0 (0.4)5.3 (0.4)< 0.001DBP, mmHg72.9 (9.3)78.4 (10.1)< 0.00167.0 (9.5)75.1 (10.2)< 0.001HbA1c, %5.1 (0.3)5.3 (0.3)< 0.0015.2 (0.3)5.4 (0.3)< 0.001BMI, kg/m222.1 (2.4)25.5 (3.0)< 0.00120.7 (2.6)25.6 (3.6)< 0.001WC, cm78.0 (6.8)86.6 (7.4)< 0.00170.8 (7.3)83.3 (8.9)< 0.001ABSI75.8 (3.3)76.7 (3.1)< 0.00174.8 (4.6)76.8 (4.5)< 0.001ARI13.1 (0.9)14.3 (1.0)< 0.00112.2 (1.0)14.0 (1.3)< 0.001OBMI+WC34.4 (3.2)38.9 (3.8)< 0.00131.7 (3.4)38.3 (4.5)< 0.001OBMI+ABSI32.0 (2.1)34.9 (2.4)< 0.00128.9 (2.3)33.4 (3.0)< 0.001BMI*WC1714.2 (1507.7-1939.1)2152.9 (1919.2-2457.9)< 0.0011423.8 (1249.1-1645.8)2070.2 (1795.3-2396.7)< 0.001BMI*ABSI1669.3 (1538.1–1806.0)1922.5 (1795.9-2082.3)< 0.0011518.4 (1390.8-1673.5)1917.8 (1756.7-2131.4)< 0.001ALT, U/L18.0 (14.0–23.0)29.0 (22.0–41.0)< 0.00113.0 (11.0–17.0)19.0 (15.0–26.0)< 0.001AST, U/L17.0 (14.0–21.0)21.0 (17.0–26.0)< 0.00116.0 (13.0–19.0)18.0 (15.0–22.0)< 0.001GGT, U/L17.0 (14.0–24.0)24.0 (18.0–35.0)< 0.00112.0 (9.0–14.0)15.0 (12.0–20.0)< 0.001TG, mmol/L0.8 (0.6–1.2)1.3 (0.9–1.9)< 0.0010.5 (0.4–0.8)1.0 (0.7–1.4)< 0.001Habit of exercise, No. (%)< 0.0010.335No4300 (79.9%)1720 (84.8%)5351 (84.1%)410 (85.8%)Yes1082 (20.1%)309 (15.2%)1011 (15.9%)68 (14.2%)Drinking status, No. (%)< 0.0010.004Non or small3731 (69.3%)1623 (80.0%)5986 (94.1%)465 (97.3%)Light1096 (20.4%)273 (13.5%)376 (5.9%)13 (2.7%)Moderate555 (10.3%)133 (6.6%)Smoking status, No. (%)0.0670.664Non1952 (36.3%)758 (37.4%)5609 (88.2%)427 (89.3%)Past1538 (28.6%)615 (30.3%)382 (6.0%)24 (5.0%)Current1892 (35.2%)656 (32.3%)371 (5.8%)27 (5.6%)Values were expressed as mean (standard deviation) or medians (quartile interval) or n (%). Abbreviations: BMI: body mass index; WC: waist circumference; ALT: alanine aminotransferase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FPG: fasting plasma glucose; HbA1c: hemoglobin A1c; SBP: systolic blood pressure; DBP: diastolic blood pressure; NAFLD: non-alcoholic fatty liver disease; ABSI: A body shape index; OBMI+ABSI: Optimal proportional combination of BMI and ABSI; OBMI+WC: Optimal proportional combination of BMI and WC; ARI: Anthropometric risk index\n\nCharacteristics of 14,251 subjects stratified by NAFLD and gender\nValues were expressed as mean (standard deviation) or medians (quartile interval) or n (%). Abbreviations: BMI: body mass index; WC: waist circumference; ALT: alanine aminotransferase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FPG: fasting plasma glucose; HbA1c: hemoglobin A1c; SBP: systolic blood pressure; DBP: diastolic blood pressure; NAFLD: non-alcoholic fatty liver disease; ABSI: A body shape index; OBMI+ABSI: Optimal proportional combination of BMI and ABSI; OBMI+WC: Optimal proportional combination of BMI and WC; ARI: Anthropometric risk index\nBaseline characteristics for all subjects according to the quartile category of BMI*ABSI are shown in Table 2. We found that with the increase of BMI*ABSI quartiles, all baseline metrics of subjects showed a trend of change (all P < 0.001). Of these, a greater proportion of male subjects were in the higher BMI*ABSI quartiles, with a male-to-female ratio of almost 7:3 in Q4, and there were more subjects with smoking and drinking habits and with less physical exercise in Q4. Moreover, subjects’ HDL-C levels decreased with increasing BMI*ABSI quartiles, while age, BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*WC, height, weight, TC, SBP, HbA1c, ALT, AST, DBP, FPG, GGT, TG levels, and the prevalence of NAFLD increased (Q4: 48.5%).\n\nTable 2Characteristics of 14,251 subjects stratified according to the BMI*ABSI quartilesBMI*ABSI quartilesvariablesQ1 (961.1-1477.2)Q2 (1477.4-1645.5)Q3 (1645.6-1827.4)Q4 (1827.5-3630.9)P-valueNo. of subjects3563356235633563Male, No. (%)867 (24.3%)1666 (46.8%)2269 (63.7%)2609 (73.2%)Age, years41.0 (8.7)43.1 (8.7)44.5 (8.6)45.6 (8.9)< 0.001BMI, kg/m218.7 (1.3)20.9 (1.2)22.7 (1.2)26.0 (2.5)< 0.001WC, cm65.4 (3.7)72.8 (2.9)78.8 (2.9)87.7 (5.8)< 0.001ABSI73.1 (3.8)75.1 (3.8)76.5 (3.5)77.5 (3.5)< 0.001ARI11.4 (0.4)12.4 (0.3)13.2 (0.3)14.4 (0.8)< 0.001OBMI+WC29.0 (1.5)32.3 (1.0)35.0 (1.1)39.6 (3.0)< 0.001OBMI+ABSI27.4 (1.3)29.9 (1.1)31.9 (1.1)35.0 (2.0)< 0.001BMI*WC1241.2 (1147.1-1318.4)1514.2 (1447.2-1587.4)1782.6 (1703.2-1865.8)2194.4 (2051.7-2436.9)< 0.001Height, cm161.7 (7.7)164.3 (8.3)166.1 (8.5)167.1 (8.4)< 0.001Weight, kg49.0 (5.7)56.4 (6.5)62.7 (7.3)72.9 (10.3)< 0.001TC, mmol/L4.9 (0.8)5.0 (0.9)5.2 (0.9)5.4 (0.9)< 0.001HDL-C, mmol/L1.7 (0.39)1.5 (0.4)1.4 (0.4)1.2 (0.3)< 0.001SBP, mmHg105.6 (12.5)111.1 (12.8)115.9 (13.3)123.1 (14.8)< 0.001FPG, mmol/L4.9 (0.4)5.1 (0.4)5.2 (0.4)5.3 (0.4)< 0.001DBP, mmHg65.5 (8.5)69.0 (9.1)72.5 (9.6)77.6 (10.2)< 0.001HbA1c, %5.1 (0.3)5.1 (0.3)5.2 (0.3)5.3 (0.3)< 0.001ALT, U/L14.0 (11.0–17.0)15.0 (12.0–19.0)18.0 (13.0–23.0)23.0 (17.0–33.0)< 0.001AST, U/L16.0 (13.0–19.0)16.00 (14.0–20.0)17.0 (14.0–21.0)19.0 (16.0–24.0)< 0.001GGT, U/L12.0 (10.0–15.0)13.00 (11.0–18.0)16.0 (12.0–22.0)21.0 (15.0–30.0)< 0.001TG, mmol/L0.51 (0.4–0.7)0.6 (0.5–0.9)0.8 (0.6–1.2)1.1 (0.8–1.6)< 0.001Habit of exercise, No. (%)< 0.001No2963 (83.2%)2858 (80.2%)2920 (82.0%)3040 (85.3%)Yes600 (16.8%)704 (19.8%)643 (18.1%)523 (14.7%)Drinking status, No. (%)< 0.001Non or small3228 (90.6%)2937 (82.5%)2848 (79.9%)2792 (78.4%)Light279 (7.8%)467 (13.1%)507 (14.2%)505 (14.2%)Moderate56 (1.6%)158 (4.4%)208 (5.8%)266 (7.5%)Smoking status, No. (%)< 0.001Non2736 (76.8%)2322 (65.2%)1957 (54.9%)1731 (48.6%)Past337 (9.5%)560 (15.7%)785 (22.0%)877 (24.6%)Current490 (13.8%)680 (19.1%)821 (23.0%)955 (26.8%)NAFLD, No. (%)16 (0.5%)150 (4.2%)611 (17.2%)1730 (48.5%)< 0.001Abbreviations as in Table 1\n\nCharacteristics of 14,251 subjects stratified according to the BMI*ABSI quartiles\nAbbreviations as in Table 1\n[SUBTITLE] Calculation of the optimal proportional combination of BMI with WC and ABSI [SUBSECTION] In the logistic regression equation constructed to calculate the optimal proportional combination OBMI+WC, the regression coefficient βBMI for BMI was 0.218 (0.154–0.282), and βWC for WC was 0.063 (0.038–0.089) in females; in males, βBMI was 0.177 (0.124–0.299), and βWC was 0.068 (0.048–0.089). Therefore, according to the calculation formula of the optimal proportional coefficients, the values ​​of nBMI and nWC in females were 0.776 and 0.224, respectively, and the values ​​of nBMI and nWC in males were 0.722 and 0.278, respectively. Similarly, in the logistic regression equation constructed to calculate the optimal proportional combination OBMI+ABSI, βBMI and βABSI were 0.356 and 0.064, respectively, in females, resulting in nBMI and nABSI values ​​of 0.848 and 0.152, respectively; in males, βBMI and βABSI were 0.334 and 0.075, respectively, and nBMI and nABSI values ​​were 0.817 and 0.183, respectively. Table 3 shows the calculation formulas of OBMI+WC and OBMI+ABSI.\n\nTable 3Optimal combination equations of BMI with WC and ABSI.Logistic regression derived equationsMaleFemaleequations for BMI and WC0.722BMI + 0.278WC0.776BMI + 0.224WCequations for BMI and ABSI0.817BMI + 0.183ABSI0.848BMI + 0.152ABSIAbbreviations as in Table 1\n\nOptimal combination equations of BMI with WC and ABSI.\nAbbreviations as in Table 1\nIn the logistic regression equation constructed to calculate the optimal proportional combination OBMI+WC, the regression coefficient βBMI for BMI was 0.218 (0.154–0.282), and βWC for WC was 0.063 (0.038–0.089) in females; in males, βBMI was 0.177 (0.124–0.299), and βWC was 0.068 (0.048–0.089). Therefore, according to the calculation formula of the optimal proportional coefficients, the values ​​of nBMI and nWC in females were 0.776 and 0.224, respectively, and the values ​​of nBMI and nWC in males were 0.722 and 0.278, respectively. Similarly, in the logistic regression equation constructed to calculate the optimal proportional combination OBMI+ABSI, βBMI and βABSI were 0.356 and 0.064, respectively, in females, resulting in nBMI and nABSI values ​​of 0.848 and 0.152, respectively; in males, βBMI and βABSI were 0.334 and 0.075, respectively, and nBMI and nABSI values ​​were 0.817 and 0.183, respectively. Table 3 shows the calculation formulas of OBMI+WC and OBMI+ABSI.\n\nTable 3Optimal combination equations of BMI with WC and ABSI.Logistic regression derived equationsMaleFemaleequations for BMI and WC0.722BMI + 0.278WC0.776BMI + 0.224WCequations for BMI and ABSI0.817BMI + 0.183ABSI0.848BMI + 0.152ABSIAbbreviations as in Table 1\n\nOptimal combination equations of BMI with WC and ABSI.\nAbbreviations as in Table 1\n[SUBTITLE] Association of various obesity indicators with NAFLD [SUBSECTION] Correlation analysis of the basic anthropometric measures showed a strong correlation between BMI and WC in both sexes (r: male 0.8796; female 0.8191), while BMI was barely correlated with ABSI (r < 0.05) (Supplementary Table 2). In contrast, the combination of ABSI and BMI may be more appropriate. Table 4 presents the associations between BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC and the risk of NAFLD in both sexes. In multivariate-adjusted logistic regression models, BMI and WC, and ABSI were positively correlated with NAFLD risk. In the current study, the combined indicators ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC were positively correlated with NAFLD risk in all four logistic regression models, and the OR values ​​in the univariate logistic regression model ranged from 1.44 to 5.02; after further adjustment for age, height, GGT, TC, AST, HDL-C, drinking status, ALT, TG, FPG, HbA1c, and SBP (Model 3), the results did not change significantly. The risk of NAFLD increased by 33% (OR 1.33, 95%CI 1.29, 1.37) and 52% (OR 1.52, 95%CI 1.45, 1.60) and 172% (OR 2.72, 95%CI 2.43, 3.04), in females, for each unit increase in OBMI+WC and OBMI+ABSI and ARI respectively. Similarly, in males, NAFLD risk was increased by 30% (OR 1.30, 95%CI 1.27, 1.33) and 50% (OR 1.50, 95%CI 1.45, 1.56) and 172% (OR 2.72, 95%CI 2.48, 2.98), respectively. Each standard deviation increase in BMI*WC and BMI*ABSI in females was associated with a 213% (OR 3.13, 95%CI 2.74,3,56) and 209% (OR 3.09, 95%CI 2.72, 3.52) increased risk for NAFLD respectively, and for males 185% (OR 2.85, 95%CI 2.59, 3.15) and 197% (OR 2.97, 95%CI 2.68, 3.28), respectively. Furthermore, the association between BMI*ABSI and NAFLD remained unchanged after treating BMI*ABSI as a categorical variable; taking the first quartile as the control group, the NAFLD risk increased with the increase of BMI*ABSI quartiles (all P for trend < 0.001). Moreover, we also analyzed the risk of NAFLD for each obesity phenotype in multivariable logistic regression models (Supplementary Table 3), which in model 3 showed that compared with the BMIN/WCN phenotype, male and female BMIO/ WCN, BMIN/WCO, and BMIO/WCO phenotypes all had a significantly higher risk of NAFLD. The risk of NAFLD associated with the BMIO/ WCN, BMIN/WCO, and BMIO/WCO phenotypes had ORs of 3.67, 3.77, and 9.78 in females and 2.76, 2.74, and 4.19 in males, respectively. Clearly, subjects with the BMIO/WCO phenotype consistently had the highest risk of developing NAFLD.\n\nTable 4Odds ratios and 95% confidence intervals of BMI, WC, ABSI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and ARI for NAFLD in female and male subjectsOdds ratios (95% confidence interval)Crude modelModel 1Model 2Model 3FemaleBMI1.58 (1.53, 1.64)*1.58 (1.52, 1.63)*1.49 (1.44, 1.55)*1.42 (1.36, 1.47)*WC1.19 (1.17, 1.21)*1.19 (1.17, 1.21)*1.17 (1.15, 1.18)*1.14 (1.12, 1.16)*ABSI1.09 (1.07, 1.12)*1.07 (1.05, 1.09)*1.06 (1.04, 1.08)*1.05 (1.02, 1.07)*ARI3.70 (3.36, 4.07)*3.61 (3.28, 3.98)*3.11 (2.80, 3.44)*2.72 (2.43, 3.04)*OBMI+ABSI1.73 (1.66, 1.80)*1.71 (1.65, 1.79)*1.61 (1.54, 1.68)*1.52 (1.45, 1.60)*OBMI+WC1.44 (1.41, 1.48)*1.44 (1.40, 1.48)*1.38 (1.34, 1.42)*1.33 (1.29, 1.37)*BMI*ABSI (Per SD)4.49 (4.02, 5.02)*4.33 (3.87, 4.84)*3.63 (3.23, 4.09)*3.09 (2.72, 3.52)*BMI*WC (Per SD)4.50 (4.03, 5.03)*4.41 (3.94, 4.93)*3.68 (3.27, 4.15)*3.13 (2.74, 3.56)*BMI*ABSI (quartiles)Quartile 11.01.01.01.0Quartile 210.90 (4.64, 25.59)*10.32 (4.39, 24.24)*8.42 (3.57, 19.86)*7.47 (3.16, 17.65)*Quartile 343.73 (19.20, 99.63)*39.09 (17.13, 89.20)*25.14 (10.94, 57.76)*19.49 (8.44, 44.98)*Quartile 4217.88 (96.72, 490.84)*188.51 (83.50, 425.59)*105.60 (46.43, 240.20)*63.97 (27.87, 146.85)*P for trend< 0.0001< 0.0001< 0.0001< 0.0001MaleBMI1.62 (1.58, 1.67)*1.62 (1.58, 1.67)*1.52 (1.48, 1.57)*1.38 (1.34, 1.42)*WC1.19 (1.18, 1.20)*1.21 (1.20, 1.22)*1.18 (1.17, 1.19)*1.13 (1.12, 1.15)*ABSI1.09 (1.07, 1.10)*1.10 (1.08, 1.12)*1.06 (1.04, 1.08)*1.05 (1.02, 1.07)*ARI4.39 (4.06, 4.75)*4.40 (4.07, 4.77)*3.68 (3.38, 4.00)*2.72 (2.48, 2.98)*OBMI+ABSI1.83 (1.77, 1.89)*1.83 (1.77, 1.89)*1.70 (1.65, 1.76)*1.50 (1.45, 1.56)*OBMI+WC1.47 (1.44, 1.50)*1.47 (1.44, 1.50)*1.41 (1.38, 1.44)*1.30 (1.27, 1.33)*BMI*ABSI (Per SD)5.02 (4.61, 5.48)*5.08 (4.66, 5.55)*4.16 (3.80, 4.56)*2.97 (2.68, 3.28)*BMI*WC (Per SD)4.72 (4.34, 5.13)*4.83 (4.44, 5.26)*3.95 (3.62, 4.32)*2.85 (2.59, 3.15)*BMI*ABSI (quartiles)Quartile 11.01.01.01.0Quartile 25.76 (3.00, 11.09)*5.89 (3.06, 11.33)*4.68 (2.42, 9.02)*3.97 (2.03, 7.78)*Quartile 323.97 (12.75, 45.07)*25.00 (13.29, 47.03)*15.83 (8.38, 29.90)*10.27 (5.35, 19.72)*Quartile 4102.03 (54.44, 191.23)*108.71 (57.95, 203.93)*58.44 (30.99, 110.21)*26.15 (13.61, 50.25)*P for trend< 0.0001< 0.0001< 0.0001< 0.0001*P < 0.0001; Abbreviation: SD: standard deviation; other abbreviations as in Table 1Model 1 adjusted for age and heightModel 2 adjusted for model 1 plus TC, TG, HDL-C, drinking statusModel 3 adjusted for model 2 plus ALT, AST, GGT, FPG, HbA1c, and SBP.\n\nOdds ratios and 95% confidence intervals of BMI, WC, ABSI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and ARI for NAFLD in female and male subjects\n*P < 0.0001; Abbreviation: SD: standard deviation; other abbreviations as in Table 1\nModel 1 adjusted for age and height\nModel 2 adjusted for model 1 plus TC, TG, HDL-C, drinking status\nModel 3 adjusted for model 2 plus ALT, AST, GGT, FPG, HbA1c, and SBP.\nCorrelation analysis of the basic anthropometric measures showed a strong correlation between BMI and WC in both sexes (r: male 0.8796; female 0.8191), while BMI was barely correlated with ABSI (r < 0.05) (Supplementary Table 2). In contrast, the combination of ABSI and BMI may be more appropriate. Table 4 presents the associations between BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC and the risk of NAFLD in both sexes. In multivariate-adjusted logistic regression models, BMI and WC, and ABSI were positively correlated with NAFLD risk. In the current study, the combined indicators ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC were positively correlated with NAFLD risk in all four logistic regression models, and the OR values ​​in the univariate logistic regression model ranged from 1.44 to 5.02; after further adjustment for age, height, GGT, TC, AST, HDL-C, drinking status, ALT, TG, FPG, HbA1c, and SBP (Model 3), the results did not change significantly. The risk of NAFLD increased by 33% (OR 1.33, 95%CI 1.29, 1.37) and 52% (OR 1.52, 95%CI 1.45, 1.60) and 172% (OR 2.72, 95%CI 2.43, 3.04), in females, for each unit increase in OBMI+WC and OBMI+ABSI and ARI respectively. Similarly, in males, NAFLD risk was increased by 30% (OR 1.30, 95%CI 1.27, 1.33) and 50% (OR 1.50, 95%CI 1.45, 1.56) and 172% (OR 2.72, 95%CI 2.48, 2.98), respectively. Each standard deviation increase in BMI*WC and BMI*ABSI in females was associated with a 213% (OR 3.13, 95%CI 2.74,3,56) and 209% (OR 3.09, 95%CI 2.72, 3.52) increased risk for NAFLD respectively, and for males 185% (OR 2.85, 95%CI 2.59, 3.15) and 197% (OR 2.97, 95%CI 2.68, 3.28), respectively. Furthermore, the association between BMI*ABSI and NAFLD remained unchanged after treating BMI*ABSI as a categorical variable; taking the first quartile as the control group, the NAFLD risk increased with the increase of BMI*ABSI quartiles (all P for trend < 0.001). Moreover, we also analyzed the risk of NAFLD for each obesity phenotype in multivariable logistic regression models (Supplementary Table 3), which in model 3 showed that compared with the BMIN/WCN phenotype, male and female BMIO/ WCN, BMIN/WCO, and BMIO/WCO phenotypes all had a significantly higher risk of NAFLD. The risk of NAFLD associated with the BMIO/ WCN, BMIN/WCO, and BMIO/WCO phenotypes had ORs of 3.67, 3.77, and 9.78 in females and 2.76, 2.74, and 4.19 in males, respectively. Clearly, subjects with the BMIO/WCO phenotype consistently had the highest risk of developing NAFLD.\n\nTable 4Odds ratios and 95% confidence intervals of BMI, WC, ABSI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and ARI for NAFLD in female and male subjectsOdds ratios (95% confidence interval)Crude modelModel 1Model 2Model 3FemaleBMI1.58 (1.53, 1.64)*1.58 (1.52, 1.63)*1.49 (1.44, 1.55)*1.42 (1.36, 1.47)*WC1.19 (1.17, 1.21)*1.19 (1.17, 1.21)*1.17 (1.15, 1.18)*1.14 (1.12, 1.16)*ABSI1.09 (1.07, 1.12)*1.07 (1.05, 1.09)*1.06 (1.04, 1.08)*1.05 (1.02, 1.07)*ARI3.70 (3.36, 4.07)*3.61 (3.28, 3.98)*3.11 (2.80, 3.44)*2.72 (2.43, 3.04)*OBMI+ABSI1.73 (1.66, 1.80)*1.71 (1.65, 1.79)*1.61 (1.54, 1.68)*1.52 (1.45, 1.60)*OBMI+WC1.44 (1.41, 1.48)*1.44 (1.40, 1.48)*1.38 (1.34, 1.42)*1.33 (1.29, 1.37)*BMI*ABSI (Per SD)4.49 (4.02, 5.02)*4.33 (3.87, 4.84)*3.63 (3.23, 4.09)*3.09 (2.72, 3.52)*BMI*WC (Per SD)4.50 (4.03, 5.03)*4.41 (3.94, 4.93)*3.68 (3.27, 4.15)*3.13 (2.74, 3.56)*BMI*ABSI (quartiles)Quartile 11.01.01.01.0Quartile 210.90 (4.64, 25.59)*10.32 (4.39, 24.24)*8.42 (3.57, 19.86)*7.47 (3.16, 17.65)*Quartile 343.73 (19.20, 99.63)*39.09 (17.13, 89.20)*25.14 (10.94, 57.76)*19.49 (8.44, 44.98)*Quartile 4217.88 (96.72, 490.84)*188.51 (83.50, 425.59)*105.60 (46.43, 240.20)*63.97 (27.87, 146.85)*P for trend< 0.0001< 0.0001< 0.0001< 0.0001MaleBMI1.62 (1.58, 1.67)*1.62 (1.58, 1.67)*1.52 (1.48, 1.57)*1.38 (1.34, 1.42)*WC1.19 (1.18, 1.20)*1.21 (1.20, 1.22)*1.18 (1.17, 1.19)*1.13 (1.12, 1.15)*ABSI1.09 (1.07, 1.10)*1.10 (1.08, 1.12)*1.06 (1.04, 1.08)*1.05 (1.02, 1.07)*ARI4.39 (4.06, 4.75)*4.40 (4.07, 4.77)*3.68 (3.38, 4.00)*2.72 (2.48, 2.98)*OBMI+ABSI1.83 (1.77, 1.89)*1.83 (1.77, 1.89)*1.70 (1.65, 1.76)*1.50 (1.45, 1.56)*OBMI+WC1.47 (1.44, 1.50)*1.47 (1.44, 1.50)*1.41 (1.38, 1.44)*1.30 (1.27, 1.33)*BMI*ABSI (Per SD)5.02 (4.61, 5.48)*5.08 (4.66, 5.55)*4.16 (3.80, 4.56)*2.97 (2.68, 3.28)*BMI*WC (Per SD)4.72 (4.34, 5.13)*4.83 (4.44, 5.26)*3.95 (3.62, 4.32)*2.85 (2.59, 3.15)*BMI*ABSI (quartiles)Quartile 11.01.01.01.0Quartile 25.76 (3.00, 11.09)*5.89 (3.06, 11.33)*4.68 (2.42, 9.02)*3.97 (2.03, 7.78)*Quartile 323.97 (12.75, 45.07)*25.00 (13.29, 47.03)*15.83 (8.38, 29.90)*10.27 (5.35, 19.72)*Quartile 4102.03 (54.44, 191.23)*108.71 (57.95, 203.93)*58.44 (30.99, 110.21)*26.15 (13.61, 50.25)*P for trend< 0.0001< 0.0001< 0.0001< 0.0001*P < 0.0001; Abbreviation: SD: standard deviation; other abbreviations as in Table 1Model 1 adjusted for age and heightModel 2 adjusted for model 1 plus TC, TG, HDL-C, drinking statusModel 3 adjusted for model 2 plus ALT, AST, GGT, FPG, HbA1c, and SBP.\n\nOdds ratios and 95% confidence intervals of BMI, WC, ABSI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and ARI for NAFLD in female and male subjects\n*P < 0.0001; Abbreviation: SD: standard deviation; other abbreviations as in Table 1\nModel 1 adjusted for age and height\nModel 2 adjusted for model 1 plus TC, TG, HDL-C, drinking status\nModel 3 adjusted for model 2 plus ALT, AST, GGT, FPG, HbA1c, and SBP.\n[SUBTITLE] Accuracy of various obesity indicators to identify NAFLD [SUBSECTION] ROC curves were drawn to assess the accuracy of BMI, WC, ABSI, ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC for identifying NAFLD in both sexes (Fig. 2). Table 5 summarizes the AUC and optimal diagnostic thresholds for these obesity indicators. In contrast, ARI and OBMI+ABSI had the same and highest AUC in both female and male subjects, with AUC and optimal diagnostic thresholds of 0.8270, 33.2569 and 0.8912, 30.4143 for ARI in males and females, respectively. The Delong test further showed that both the combined indicators of BMI and WC (OBMI+WC and BMI*WC) and BMI and ABSI (OBMI+ABSI and BMI*ABSI and ARI) were significantly better at identifying the risk of NAFLD than BMI or WC or ABSI alone (Delong test P < 0.05).\n\nFig. 2ROC curve analysis of BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC in females and males\n\nROC curve analysis of BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC in females and males\n\nTable 5The best threshold, sensitivities, specificities, Youden’s index, and area under the curve of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI in different gendersAUC95%CI low95%CI upBest thresholdSpecificitySensitivityYouden’s indexFemaleBMI**0.87990.86480.895022.75650.81770.79500.6127WC**0.86950.85450.884474.25000.70500.87450.5804ABSI**ARI**0.61710.89120.59160.87750.64260.904875.792512.78250.59260.77350.59620.85770.18880.6312OBMI+ABSI**0.89120.87750.904830.41430.77330.85770.6310OBMI+WC0.88880.87500.902734.12340.77160.85560.6272BMI*ABSI0.88840.87480.90211704.21000.79220.82850.6207BMI*WC0.88880.87500.90271674.11830.77330.85560.6286MaleBMI**0.81600.80550.826423.55550.73820.73090.4691WC**0.81020.79980.820780.65000.66740.80340.4708ABSI**0.57950.56530.593674.91280.40520.72400.1292ARI**0.82700.81700.836913.60960.74560.74570.4913OBMI+ABSI**0.82700.81700.836933.25690.74580.74570.4915OBMI+WC0.82420.81410.834339.83130.74380.74470.4885BMI*ABSI0.82570.81570.83561758.75900.67610.81170.4878BMI*WC0.82440.81430.83451944.04330.75510.73290.4880Abbreviations as in Table 1**P < 0.05 compared with BMI*WC in each gender\n\nThe best threshold, sensitivities, specificities, Youden’s index, and area under the curve of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI in different genders\nABSI**\nARI**\n0.6171\n0.8912\n0.5916\n0.8775\n0.6426\n0.9048\n75.7925\n12.7825\n0.5926\n0.7735\n0.5962\n0.8577\n0.1888\n0.6312\nAbbreviations as in Table 1\n**P < 0.05 compared with BMI*WC in each gender\nTo explore the changes in BMI, WC, ABSI, ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC on the risk identification ability of NAFLD in different age subgroups of both sexes, we constructed ROC curves for 20–39, 40–59, and ≥ 60 years in both sexes, respectively; and the corresponding AUC and optimal diagnostic thresholds were summarized in Table 6. We found that ARI and OBMI+ABSI exhibited considerably higher AUCs in all age subgroups for both sexes except in the female ≥ 60-year age group; the AUC of OBMI+ABSI in the female 20–39 age group was 0.9523, the highest value among all age subgroups of both sexes. In the male 20-59-year-old group and the female 40-59-year-old group, the AUC values ​​of ARI and OBMI+ABSI were slightly higher than those of BMI*WC, but all were significantly higher than those of BMI or WC or ABSI alone, while in the female 20–39 years group and the female ≥ 60 years group AUC values for all combined obesity indicators were significantly higher than that of WC and ABSI; in addition, in the male ≥ 60-year-old group, the seven indicators of obesity had similar NAFLD identification abilities, only ABSI underperformed. Therefore, the combined indicators ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC can significantly improve the ability of simple obesity parameters BMI and WC and ABSI to identify NAFLD in middle-aged females and young and middle-aged males and had the highest diagnostic performance in young females.\n\nTable 6The area under the curve, best threshold, sensitivities, specificities, and Youden’s index of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI as a predictor of NAFLD in the different age groups of both gendersAUC95%CI low95%CI upBest thresholdSpecificitySensitivityYouden’s indexFemaleAge 20–39 yearsBMI0.95110.93360.968523.12660.89810.87230.7704WC***0.93190.90790.955976.60000.85600.88300.7390ABSI***0.56860.50850.628875.57420.61220.53190.1441ARI0.95220.93370.970812.79220.84980.93620.7860BMI*WC0.95160.93310.97011675.47840.84360.92550.7691BMI*ABSI0.94860.92890.96821701.34860.86140.92550.7869OBMI+ABSI0.95230.93370.970830.43680.84980.93620.7860OBMI+WC0.95160.93300.970134.12960.84240.92550.7679Age 40–59 yearsBMI***0.84840.82820.868622.75270.78410.77440.5585WC***0.84230.82300.861674.25000.66300.86910.5321ABSI***0.61860.58970.647675.79250.58080.60720.1880ARI***0.86220.84380.880513.02830.80770.77440.5821BMI*WC0.86030.84170.87881674.11830.73320.83840.5716BMI*ABSI0.86020.84200.87851704.21000.75460.81340.5680OBMI+ABSI***0.86220.84380.880530.99950.80790.77440.5823OBMI+WC0.86020.84170.878835.03630.80930.76320.5725Age ≥ 60 yearsBMI0.83080.74580.915923.33340.84390.76000.6039WC***0.76960.67780.861379.75000.77070.68000.4507ABSI***0.56600.45510.677078.18360.50240.72000.2224ARI0.81950.73910.899912.92520.70240.84000.5424BMI*WC0.81990.73960.90021686.12800.69760.84000.5376BMI*ABSI0.80140.71860.88411659.09940.56100.96000.5210OBMI+ABSI0.81950.73920.899830.75250.70240.84000.5424OBMI+WC0.82010.73930.900934.33930.70240.84000.5424MaleAge 20–39 yearsBMI***0.84390.82750.860223.47820.74950.76650.5160WC***0.84150.82550.857580.45000.72190.80090.5228ABSI***0.60630.58260.630074.50900.48710.69910.1862ARI***0.85490.83960.870213.50260.76000.78440.5444BMI*WC0.85130.83570.86691862.54150.72190.81140.5333BMI*ABSI***0.85520.83990.87041766.28860.75570.78740.5431OBMI+ABSI***0.85490.83960.870233.00760.76140.78290.5443OBMI+WC0.85140.83580.866939.14530.72900.80840.5374Age 40–59 yearsBMI***0.80070.78670.814723.25050.67790.76390.4418WC***0.79080.77660.805181.25000.65830.77950.4378ABSI***0.56150.54290.580175.42940.38970.70050.0902ARI***0.81040.79690.824013.61420.72320.74510.4683BMI*WC0.80800.79430.82171955.12030.74480.71770.4625BMI*ABSI0.80860.79500.82221816.10160.73340.72870.4621OBMI+ABSI***0.81050.79690.824033.27330.72390.74430.4682OBMI+WC0.80750.79380.821240.11920.74750.71850.4660Age ≥ 60 yearsBMI0.79030.73630.844423.74200.81110.69510.5062WC0.78040.72820.832680.50000.59440.86590.4603ABSI***0.58040.51190.649075.85420.29410.86590.1600ARI0.79490.74370.846213.58190.67800.81710.4951BMI*WC0.80290.75130.85451932.61210.73680.79270.5295BMI*ABSI0.79040.73930.84141800.36700.65940.82930.4887OBMI+ABSI0.79500.74380.846233.18800.67800.81710.4951OBMI+WC0.80230.75090.853639.74760.71210.81710.5292Abbreviations as in Table 1***P < 0.05 compared with BMI*WC in each age group of both genders\n\nThe area under the curve, best threshold, sensitivities, specificities, and Youden’s index of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI as a predictor of NAFLD in the different age groups of both genders\nAbbreviations as in Table 1\n***P < 0.05 compared with BMI*WC in each age group of both genders\nROC curves were drawn to assess the accuracy of BMI, WC, ABSI, ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC for identifying NAFLD in both sexes (Fig. 2). Table 5 summarizes the AUC and optimal diagnostic thresholds for these obesity indicators. In contrast, ARI and OBMI+ABSI had the same and highest AUC in both female and male subjects, with AUC and optimal diagnostic thresholds of 0.8270, 33.2569 and 0.8912, 30.4143 for ARI in males and females, respectively. The Delong test further showed that both the combined indicators of BMI and WC (OBMI+WC and BMI*WC) and BMI and ABSI (OBMI+ABSI and BMI*ABSI and ARI) were significantly better at identifying the risk of NAFLD than BMI or WC or ABSI alone (Delong test P < 0.05).\n\nFig. 2ROC curve analysis of BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC in females and males\n\nROC curve analysis of BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC in females and males\n\nTable 5The best threshold, sensitivities, specificities, Youden’s index, and area under the curve of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI in different gendersAUC95%CI low95%CI upBest thresholdSpecificitySensitivityYouden’s indexFemaleBMI**0.87990.86480.895022.75650.81770.79500.6127WC**0.86950.85450.884474.25000.70500.87450.5804ABSI**ARI**0.61710.89120.59160.87750.64260.904875.792512.78250.59260.77350.59620.85770.18880.6312OBMI+ABSI**0.89120.87750.904830.41430.77330.85770.6310OBMI+WC0.88880.87500.902734.12340.77160.85560.6272BMI*ABSI0.88840.87480.90211704.21000.79220.82850.6207BMI*WC0.88880.87500.90271674.11830.77330.85560.6286MaleBMI**0.81600.80550.826423.55550.73820.73090.4691WC**0.81020.79980.820780.65000.66740.80340.4708ABSI**0.57950.56530.593674.91280.40520.72400.1292ARI**0.82700.81700.836913.60960.74560.74570.4913OBMI+ABSI**0.82700.81700.836933.25690.74580.74570.4915OBMI+WC0.82420.81410.834339.83130.74380.74470.4885BMI*ABSI0.82570.81570.83561758.75900.67610.81170.4878BMI*WC0.82440.81430.83451944.04330.75510.73290.4880Abbreviations as in Table 1**P < 0.05 compared with BMI*WC in each gender\n\nThe best threshold, sensitivities, specificities, Youden’s index, and area under the curve of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI in different genders\nABSI**\nARI**\n0.6171\n0.8912\n0.5916\n0.8775\n0.6426\n0.9048\n75.7925\n12.7825\n0.5926\n0.7735\n0.5962\n0.8577\n0.1888\n0.6312\nAbbreviations as in Table 1\n**P < 0.05 compared with BMI*WC in each gender\nTo explore the changes in BMI, WC, ABSI, ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC on the risk identification ability of NAFLD in different age subgroups of both sexes, we constructed ROC curves for 20–39, 40–59, and ≥ 60 years in both sexes, respectively; and the corresponding AUC and optimal diagnostic thresholds were summarized in Table 6. We found that ARI and OBMI+ABSI exhibited considerably higher AUCs in all age subgroups for both sexes except in the female ≥ 60-year age group; the AUC of OBMI+ABSI in the female 20–39 age group was 0.9523, the highest value among all age subgroups of both sexes. In the male 20-59-year-old group and the female 40-59-year-old group, the AUC values ​​of ARI and OBMI+ABSI were slightly higher than those of BMI*WC, but all were significantly higher than those of BMI or WC or ABSI alone, while in the female 20–39 years group and the female ≥ 60 years group AUC values for all combined obesity indicators were significantly higher than that of WC and ABSI; in addition, in the male ≥ 60-year-old group, the seven indicators of obesity had similar NAFLD identification abilities, only ABSI underperformed. Therefore, the combined indicators ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC can significantly improve the ability of simple obesity parameters BMI and WC and ABSI to identify NAFLD in middle-aged females and young and middle-aged males and had the highest diagnostic performance in young females.\n\nTable 6The area under the curve, best threshold, sensitivities, specificities, and Youden’s index of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI as a predictor of NAFLD in the different age groups of both gendersAUC95%CI low95%CI upBest thresholdSpecificitySensitivityYouden’s indexFemaleAge 20–39 yearsBMI0.95110.93360.968523.12660.89810.87230.7704WC***0.93190.90790.955976.60000.85600.88300.7390ABSI***0.56860.50850.628875.57420.61220.53190.1441ARI0.95220.93370.970812.79220.84980.93620.7860BMI*WC0.95160.93310.97011675.47840.84360.92550.7691BMI*ABSI0.94860.92890.96821701.34860.86140.92550.7869OBMI+ABSI0.95230.93370.970830.43680.84980.93620.7860OBMI+WC0.95160.93300.970134.12960.84240.92550.7679Age 40–59 yearsBMI***0.84840.82820.868622.75270.78410.77440.5585WC***0.84230.82300.861674.25000.66300.86910.5321ABSI***0.61860.58970.647675.79250.58080.60720.1880ARI***0.86220.84380.880513.02830.80770.77440.5821BMI*WC0.86030.84170.87881674.11830.73320.83840.5716BMI*ABSI0.86020.84200.87851704.21000.75460.81340.5680OBMI+ABSI***0.86220.84380.880530.99950.80790.77440.5823OBMI+WC0.86020.84170.878835.03630.80930.76320.5725Age ≥ 60 yearsBMI0.83080.74580.915923.33340.84390.76000.6039WC***0.76960.67780.861379.75000.77070.68000.4507ABSI***0.56600.45510.677078.18360.50240.72000.2224ARI0.81950.73910.899912.92520.70240.84000.5424BMI*WC0.81990.73960.90021686.12800.69760.84000.5376BMI*ABSI0.80140.71860.88411659.09940.56100.96000.5210OBMI+ABSI0.81950.73920.899830.75250.70240.84000.5424OBMI+WC0.82010.73930.900934.33930.70240.84000.5424MaleAge 20–39 yearsBMI***0.84390.82750.860223.47820.74950.76650.5160WC***0.84150.82550.857580.45000.72190.80090.5228ABSI***0.60630.58260.630074.50900.48710.69910.1862ARI***0.85490.83960.870213.50260.76000.78440.5444BMI*WC0.85130.83570.86691862.54150.72190.81140.5333BMI*ABSI***0.85520.83990.87041766.28860.75570.78740.5431OBMI+ABSI***0.85490.83960.870233.00760.76140.78290.5443OBMI+WC0.85140.83580.866939.14530.72900.80840.5374Age 40–59 yearsBMI***0.80070.78670.814723.25050.67790.76390.4418WC***0.79080.77660.805181.25000.65830.77950.4378ABSI***0.56150.54290.580175.42940.38970.70050.0902ARI***0.81040.79690.824013.61420.72320.74510.4683BMI*WC0.80800.79430.82171955.12030.74480.71770.4625BMI*ABSI0.80860.79500.82221816.10160.73340.72870.4621OBMI+ABSI***0.81050.79690.824033.27330.72390.74430.4682OBMI+WC0.80750.79380.821240.11920.74750.71850.4660Age ≥ 60 yearsBMI0.79030.73630.844423.74200.81110.69510.5062WC0.78040.72820.832680.50000.59440.86590.4603ABSI***0.58040.51190.649075.85420.29410.86590.1600ARI0.79490.74370.846213.58190.67800.81710.4951BMI*WC0.80290.75130.85451932.61210.73680.79270.5295BMI*ABSI0.79040.73930.84141800.36700.65940.82930.4887OBMI+ABSI0.79500.74380.846233.18800.67800.81710.4951OBMI+WC0.80230.75090.853639.74760.71210.81710.5292Abbreviations as in Table 1***P < 0.05 compared with BMI*WC in each age group of both genders\n\nThe area under the curve, best threshold, sensitivities, specificities, and Youden’s index of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI as a predictor of NAFLD in the different age groups of both genders\nAbbreviations as in Table 1\n***P < 0.05 compared with BMI*WC in each age group of both genders", "The study included 14,251 subjects, of whom 7,411 (52%) were males, with a mean age of 43.82 (8.99) years, and 2,029 (27.37%) were diagnosed with NAFLD; 6,840 were (48%) females, with mean age 43.22 (8.78) years, and 478 (6.99%) were diagnosed with NAFLD. Table 1 is stratified by gender and describes the differences in baseline characteristics between NAFLD patients and non-NAFLD subjects. We found that NAFLD patients consistently had higher age, weight, TC, SBP, DBP, HbA1c, BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, ALT, AST, GGT, FPG, and TG levels in both sexes, but with lower HDL-C levels. Notably, non-NAFLD subjects of both sexes tended to have greater alcohol consumption, which paradoxically appeared to be a protective factor for NAFLD. Additionally, the comparison between the NAFLD group and the non-NAFLD group showed that physical exercise habits were only significantly different in males, height was only significantly different in females, and smoking habits were not significantly different in both sexes.\n\nTable 1Characteristics of 14,251 subjects stratified by NAFLD and genderMaleFemaleVariablesNon-NAFLDn = 5382 (72.61%)NAFLDn = 2029 (27.39%)P-valueNon-NAFLDn = 6362 (93.01%)NAFLDn = 478 (6.99%)P-valueAge, years43.7 (9.3)44.1 (8.2)0.00242.9 (8.7)47.6 (8.3)< 0.001Height, cm170.9 (6.0)170.6 (5.9)0.084158.4 (5.4)157.0 (5.3)< 0.001Weight, kg64.6 (8.3)74.3 (10.6)< 0.00151.9 (7.1)63.2 (10.0)< 0.001TC, mmol/L5.1 (0.8)5.4 (0.9)< 0.0015.1 (0.9)5.6 (0.9)< 0.001HDL-C, mmol/L1.3 (0.3)1.1 (0.3)< 0.0011.7 (0.4)1.4 (0.3)< 0.001SBP, mmHg116.0 (13.2)124.0 (14.5)< 0.001108.4 (13.8)120.7 (16.0)< 0.001FPG, mmol/L5.3 (0.4)5.4 (0.3)< 0.0015.0 (0.4)5.3 (0.4)< 0.001DBP, mmHg72.9 (9.3)78.4 (10.1)< 0.00167.0 (9.5)75.1 (10.2)< 0.001HbA1c, %5.1 (0.3)5.3 (0.3)< 0.0015.2 (0.3)5.4 (0.3)< 0.001BMI, kg/m222.1 (2.4)25.5 (3.0)< 0.00120.7 (2.6)25.6 (3.6)< 0.001WC, cm78.0 (6.8)86.6 (7.4)< 0.00170.8 (7.3)83.3 (8.9)< 0.001ABSI75.8 (3.3)76.7 (3.1)< 0.00174.8 (4.6)76.8 (4.5)< 0.001ARI13.1 (0.9)14.3 (1.0)< 0.00112.2 (1.0)14.0 (1.3)< 0.001OBMI+WC34.4 (3.2)38.9 (3.8)< 0.00131.7 (3.4)38.3 (4.5)< 0.001OBMI+ABSI32.0 (2.1)34.9 (2.4)< 0.00128.9 (2.3)33.4 (3.0)< 0.001BMI*WC1714.2 (1507.7-1939.1)2152.9 (1919.2-2457.9)< 0.0011423.8 (1249.1-1645.8)2070.2 (1795.3-2396.7)< 0.001BMI*ABSI1669.3 (1538.1–1806.0)1922.5 (1795.9-2082.3)< 0.0011518.4 (1390.8-1673.5)1917.8 (1756.7-2131.4)< 0.001ALT, U/L18.0 (14.0–23.0)29.0 (22.0–41.0)< 0.00113.0 (11.0–17.0)19.0 (15.0–26.0)< 0.001AST, U/L17.0 (14.0–21.0)21.0 (17.0–26.0)< 0.00116.0 (13.0–19.0)18.0 (15.0–22.0)< 0.001GGT, U/L17.0 (14.0–24.0)24.0 (18.0–35.0)< 0.00112.0 (9.0–14.0)15.0 (12.0–20.0)< 0.001TG, mmol/L0.8 (0.6–1.2)1.3 (0.9–1.9)< 0.0010.5 (0.4–0.8)1.0 (0.7–1.4)< 0.001Habit of exercise, No. (%)< 0.0010.335No4300 (79.9%)1720 (84.8%)5351 (84.1%)410 (85.8%)Yes1082 (20.1%)309 (15.2%)1011 (15.9%)68 (14.2%)Drinking status, No. (%)< 0.0010.004Non or small3731 (69.3%)1623 (80.0%)5986 (94.1%)465 (97.3%)Light1096 (20.4%)273 (13.5%)376 (5.9%)13 (2.7%)Moderate555 (10.3%)133 (6.6%)Smoking status, No. (%)0.0670.664Non1952 (36.3%)758 (37.4%)5609 (88.2%)427 (89.3%)Past1538 (28.6%)615 (30.3%)382 (6.0%)24 (5.0%)Current1892 (35.2%)656 (32.3%)371 (5.8%)27 (5.6%)Values were expressed as mean (standard deviation) or medians (quartile interval) or n (%). Abbreviations: BMI: body mass index; WC: waist circumference; ALT: alanine aminotransferase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FPG: fasting plasma glucose; HbA1c: hemoglobin A1c; SBP: systolic blood pressure; DBP: diastolic blood pressure; NAFLD: non-alcoholic fatty liver disease; ABSI: A body shape index; OBMI+ABSI: Optimal proportional combination of BMI and ABSI; OBMI+WC: Optimal proportional combination of BMI and WC; ARI: Anthropometric risk index\n\nCharacteristics of 14,251 subjects stratified by NAFLD and gender\nValues were expressed as mean (standard deviation) or medians (quartile interval) or n (%). Abbreviations: BMI: body mass index; WC: waist circumference; ALT: alanine aminotransferase; AST: aspartate aminotransferase; GGT: gamma-glutamyl transferase; HDL-C: high-density lipoprotein cholesterol; TC: total cholesterol; TG: triglyceride; FPG: fasting plasma glucose; HbA1c: hemoglobin A1c; SBP: systolic blood pressure; DBP: diastolic blood pressure; NAFLD: non-alcoholic fatty liver disease; ABSI: A body shape index; OBMI+ABSI: Optimal proportional combination of BMI and ABSI; OBMI+WC: Optimal proportional combination of BMI and WC; ARI: Anthropometric risk index\nBaseline characteristics for all subjects according to the quartile category of BMI*ABSI are shown in Table 2. We found that with the increase of BMI*ABSI quartiles, all baseline metrics of subjects showed a trend of change (all P < 0.001). Of these, a greater proportion of male subjects were in the higher BMI*ABSI quartiles, with a male-to-female ratio of almost 7:3 in Q4, and there were more subjects with smoking and drinking habits and with less physical exercise in Q4. Moreover, subjects’ HDL-C levels decreased with increasing BMI*ABSI quartiles, while age, BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*WC, height, weight, TC, SBP, HbA1c, ALT, AST, DBP, FPG, GGT, TG levels, and the prevalence of NAFLD increased (Q4: 48.5%).\n\nTable 2Characteristics of 14,251 subjects stratified according to the BMI*ABSI quartilesBMI*ABSI quartilesvariablesQ1 (961.1-1477.2)Q2 (1477.4-1645.5)Q3 (1645.6-1827.4)Q4 (1827.5-3630.9)P-valueNo. of subjects3563356235633563Male, No. (%)867 (24.3%)1666 (46.8%)2269 (63.7%)2609 (73.2%)Age, years41.0 (8.7)43.1 (8.7)44.5 (8.6)45.6 (8.9)< 0.001BMI, kg/m218.7 (1.3)20.9 (1.2)22.7 (1.2)26.0 (2.5)< 0.001WC, cm65.4 (3.7)72.8 (2.9)78.8 (2.9)87.7 (5.8)< 0.001ABSI73.1 (3.8)75.1 (3.8)76.5 (3.5)77.5 (3.5)< 0.001ARI11.4 (0.4)12.4 (0.3)13.2 (0.3)14.4 (0.8)< 0.001OBMI+WC29.0 (1.5)32.3 (1.0)35.0 (1.1)39.6 (3.0)< 0.001OBMI+ABSI27.4 (1.3)29.9 (1.1)31.9 (1.1)35.0 (2.0)< 0.001BMI*WC1241.2 (1147.1-1318.4)1514.2 (1447.2-1587.4)1782.6 (1703.2-1865.8)2194.4 (2051.7-2436.9)< 0.001Height, cm161.7 (7.7)164.3 (8.3)166.1 (8.5)167.1 (8.4)< 0.001Weight, kg49.0 (5.7)56.4 (6.5)62.7 (7.3)72.9 (10.3)< 0.001TC, mmol/L4.9 (0.8)5.0 (0.9)5.2 (0.9)5.4 (0.9)< 0.001HDL-C, mmol/L1.7 (0.39)1.5 (0.4)1.4 (0.4)1.2 (0.3)< 0.001SBP, mmHg105.6 (12.5)111.1 (12.8)115.9 (13.3)123.1 (14.8)< 0.001FPG, mmol/L4.9 (0.4)5.1 (0.4)5.2 (0.4)5.3 (0.4)< 0.001DBP, mmHg65.5 (8.5)69.0 (9.1)72.5 (9.6)77.6 (10.2)< 0.001HbA1c, %5.1 (0.3)5.1 (0.3)5.2 (0.3)5.3 (0.3)< 0.001ALT, U/L14.0 (11.0–17.0)15.0 (12.0–19.0)18.0 (13.0–23.0)23.0 (17.0–33.0)< 0.001AST, U/L16.0 (13.0–19.0)16.00 (14.0–20.0)17.0 (14.0–21.0)19.0 (16.0–24.0)< 0.001GGT, U/L12.0 (10.0–15.0)13.00 (11.0–18.0)16.0 (12.0–22.0)21.0 (15.0–30.0)< 0.001TG, mmol/L0.51 (0.4–0.7)0.6 (0.5–0.9)0.8 (0.6–1.2)1.1 (0.8–1.6)< 0.001Habit of exercise, No. (%)< 0.001No2963 (83.2%)2858 (80.2%)2920 (82.0%)3040 (85.3%)Yes600 (16.8%)704 (19.8%)643 (18.1%)523 (14.7%)Drinking status, No. (%)< 0.001Non or small3228 (90.6%)2937 (82.5%)2848 (79.9%)2792 (78.4%)Light279 (7.8%)467 (13.1%)507 (14.2%)505 (14.2%)Moderate56 (1.6%)158 (4.4%)208 (5.8%)266 (7.5%)Smoking status, No. (%)< 0.001Non2736 (76.8%)2322 (65.2%)1957 (54.9%)1731 (48.6%)Past337 (9.5%)560 (15.7%)785 (22.0%)877 (24.6%)Current490 (13.8%)680 (19.1%)821 (23.0%)955 (26.8%)NAFLD, No. (%)16 (0.5%)150 (4.2%)611 (17.2%)1730 (48.5%)< 0.001Abbreviations as in Table 1\n\nCharacteristics of 14,251 subjects stratified according to the BMI*ABSI quartiles\nAbbreviations as in Table 1", "In the logistic regression equation constructed to calculate the optimal proportional combination OBMI+WC, the regression coefficient βBMI for BMI was 0.218 (0.154–0.282), and βWC for WC was 0.063 (0.038–0.089) in females; in males, βBMI was 0.177 (0.124–0.299), and βWC was 0.068 (0.048–0.089). Therefore, according to the calculation formula of the optimal proportional coefficients, the values ​​of nBMI and nWC in females were 0.776 and 0.224, respectively, and the values ​​of nBMI and nWC in males were 0.722 and 0.278, respectively. Similarly, in the logistic regression equation constructed to calculate the optimal proportional combination OBMI+ABSI, βBMI and βABSI were 0.356 and 0.064, respectively, in females, resulting in nBMI and nABSI values ​​of 0.848 and 0.152, respectively; in males, βBMI and βABSI were 0.334 and 0.075, respectively, and nBMI and nABSI values ​​were 0.817 and 0.183, respectively. Table 3 shows the calculation formulas of OBMI+WC and OBMI+ABSI.\n\nTable 3Optimal combination equations of BMI with WC and ABSI.Logistic regression derived equationsMaleFemaleequations for BMI and WC0.722BMI + 0.278WC0.776BMI + 0.224WCequations for BMI and ABSI0.817BMI + 0.183ABSI0.848BMI + 0.152ABSIAbbreviations as in Table 1\n\nOptimal combination equations of BMI with WC and ABSI.\nAbbreviations as in Table 1", "Correlation analysis of the basic anthropometric measures showed a strong correlation between BMI and WC in both sexes (r: male 0.8796; female 0.8191), while BMI was barely correlated with ABSI (r < 0.05) (Supplementary Table 2). In contrast, the combination of ABSI and BMI may be more appropriate. Table 4 presents the associations between BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC and the risk of NAFLD in both sexes. In multivariate-adjusted logistic regression models, BMI and WC, and ABSI were positively correlated with NAFLD risk. In the current study, the combined indicators ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC were positively correlated with NAFLD risk in all four logistic regression models, and the OR values ​​in the univariate logistic regression model ranged from 1.44 to 5.02; after further adjustment for age, height, GGT, TC, AST, HDL-C, drinking status, ALT, TG, FPG, HbA1c, and SBP (Model 3), the results did not change significantly. The risk of NAFLD increased by 33% (OR 1.33, 95%CI 1.29, 1.37) and 52% (OR 1.52, 95%CI 1.45, 1.60) and 172% (OR 2.72, 95%CI 2.43, 3.04), in females, for each unit increase in OBMI+WC and OBMI+ABSI and ARI respectively. Similarly, in males, NAFLD risk was increased by 30% (OR 1.30, 95%CI 1.27, 1.33) and 50% (OR 1.50, 95%CI 1.45, 1.56) and 172% (OR 2.72, 95%CI 2.48, 2.98), respectively. Each standard deviation increase in BMI*WC and BMI*ABSI in females was associated with a 213% (OR 3.13, 95%CI 2.74,3,56) and 209% (OR 3.09, 95%CI 2.72, 3.52) increased risk for NAFLD respectively, and for males 185% (OR 2.85, 95%CI 2.59, 3.15) and 197% (OR 2.97, 95%CI 2.68, 3.28), respectively. Furthermore, the association between BMI*ABSI and NAFLD remained unchanged after treating BMI*ABSI as a categorical variable; taking the first quartile as the control group, the NAFLD risk increased with the increase of BMI*ABSI quartiles (all P for trend < 0.001). Moreover, we also analyzed the risk of NAFLD for each obesity phenotype in multivariable logistic regression models (Supplementary Table 3), which in model 3 showed that compared with the BMIN/WCN phenotype, male and female BMIO/ WCN, BMIN/WCO, and BMIO/WCO phenotypes all had a significantly higher risk of NAFLD. The risk of NAFLD associated with the BMIO/ WCN, BMIN/WCO, and BMIO/WCO phenotypes had ORs of 3.67, 3.77, and 9.78 in females and 2.76, 2.74, and 4.19 in males, respectively. Clearly, subjects with the BMIO/WCO phenotype consistently had the highest risk of developing NAFLD.\n\nTable 4Odds ratios and 95% confidence intervals of BMI, WC, ABSI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and ARI for NAFLD in female and male subjectsOdds ratios (95% confidence interval)Crude modelModel 1Model 2Model 3FemaleBMI1.58 (1.53, 1.64)*1.58 (1.52, 1.63)*1.49 (1.44, 1.55)*1.42 (1.36, 1.47)*WC1.19 (1.17, 1.21)*1.19 (1.17, 1.21)*1.17 (1.15, 1.18)*1.14 (1.12, 1.16)*ABSI1.09 (1.07, 1.12)*1.07 (1.05, 1.09)*1.06 (1.04, 1.08)*1.05 (1.02, 1.07)*ARI3.70 (3.36, 4.07)*3.61 (3.28, 3.98)*3.11 (2.80, 3.44)*2.72 (2.43, 3.04)*OBMI+ABSI1.73 (1.66, 1.80)*1.71 (1.65, 1.79)*1.61 (1.54, 1.68)*1.52 (1.45, 1.60)*OBMI+WC1.44 (1.41, 1.48)*1.44 (1.40, 1.48)*1.38 (1.34, 1.42)*1.33 (1.29, 1.37)*BMI*ABSI (Per SD)4.49 (4.02, 5.02)*4.33 (3.87, 4.84)*3.63 (3.23, 4.09)*3.09 (2.72, 3.52)*BMI*WC (Per SD)4.50 (4.03, 5.03)*4.41 (3.94, 4.93)*3.68 (3.27, 4.15)*3.13 (2.74, 3.56)*BMI*ABSI (quartiles)Quartile 11.01.01.01.0Quartile 210.90 (4.64, 25.59)*10.32 (4.39, 24.24)*8.42 (3.57, 19.86)*7.47 (3.16, 17.65)*Quartile 343.73 (19.20, 99.63)*39.09 (17.13, 89.20)*25.14 (10.94, 57.76)*19.49 (8.44, 44.98)*Quartile 4217.88 (96.72, 490.84)*188.51 (83.50, 425.59)*105.60 (46.43, 240.20)*63.97 (27.87, 146.85)*P for trend< 0.0001< 0.0001< 0.0001< 0.0001MaleBMI1.62 (1.58, 1.67)*1.62 (1.58, 1.67)*1.52 (1.48, 1.57)*1.38 (1.34, 1.42)*WC1.19 (1.18, 1.20)*1.21 (1.20, 1.22)*1.18 (1.17, 1.19)*1.13 (1.12, 1.15)*ABSI1.09 (1.07, 1.10)*1.10 (1.08, 1.12)*1.06 (1.04, 1.08)*1.05 (1.02, 1.07)*ARI4.39 (4.06, 4.75)*4.40 (4.07, 4.77)*3.68 (3.38, 4.00)*2.72 (2.48, 2.98)*OBMI+ABSI1.83 (1.77, 1.89)*1.83 (1.77, 1.89)*1.70 (1.65, 1.76)*1.50 (1.45, 1.56)*OBMI+WC1.47 (1.44, 1.50)*1.47 (1.44, 1.50)*1.41 (1.38, 1.44)*1.30 (1.27, 1.33)*BMI*ABSI (Per SD)5.02 (4.61, 5.48)*5.08 (4.66, 5.55)*4.16 (3.80, 4.56)*2.97 (2.68, 3.28)*BMI*WC (Per SD)4.72 (4.34, 5.13)*4.83 (4.44, 5.26)*3.95 (3.62, 4.32)*2.85 (2.59, 3.15)*BMI*ABSI (quartiles)Quartile 11.01.01.01.0Quartile 25.76 (3.00, 11.09)*5.89 (3.06, 11.33)*4.68 (2.42, 9.02)*3.97 (2.03, 7.78)*Quartile 323.97 (12.75, 45.07)*25.00 (13.29, 47.03)*15.83 (8.38, 29.90)*10.27 (5.35, 19.72)*Quartile 4102.03 (54.44, 191.23)*108.71 (57.95, 203.93)*58.44 (30.99, 110.21)*26.15 (13.61, 50.25)*P for trend< 0.0001< 0.0001< 0.0001< 0.0001*P < 0.0001; Abbreviation: SD: standard deviation; other abbreviations as in Table 1Model 1 adjusted for age and heightModel 2 adjusted for model 1 plus TC, TG, HDL-C, drinking statusModel 3 adjusted for model 2 plus ALT, AST, GGT, FPG, HbA1c, and SBP.\n\nOdds ratios and 95% confidence intervals of BMI, WC, ABSI, OBMI+WC, OBMI+ABSI, BMI*ABSI, BMI*WC, and ARI for NAFLD in female and male subjects\n*P < 0.0001; Abbreviation: SD: standard deviation; other abbreviations as in Table 1\nModel 1 adjusted for age and height\nModel 2 adjusted for model 1 plus TC, TG, HDL-C, drinking status\nModel 3 adjusted for model 2 plus ALT, AST, GGT, FPG, HbA1c, and SBP.", "ROC curves were drawn to assess the accuracy of BMI, WC, ABSI, ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC for identifying NAFLD in both sexes (Fig. 2). Table 5 summarizes the AUC and optimal diagnostic thresholds for these obesity indicators. In contrast, ARI and OBMI+ABSI had the same and highest AUC in both female and male subjects, with AUC and optimal diagnostic thresholds of 0.8270, 33.2569 and 0.8912, 30.4143 for ARI in males and females, respectively. The Delong test further showed that both the combined indicators of BMI and WC (OBMI+WC and BMI*WC) and BMI and ABSI (OBMI+ABSI and BMI*ABSI and ARI) were significantly better at identifying the risk of NAFLD than BMI or WC or ABSI alone (Delong test P < 0.05).\n\nFig. 2ROC curve analysis of BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC in females and males\n\nROC curve analysis of BMI, WC, ABSI, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC in females and males\n\nTable 5The best threshold, sensitivities, specificities, Youden’s index, and area under the curve of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI in different gendersAUC95%CI low95%CI upBest thresholdSpecificitySensitivityYouden’s indexFemaleBMI**0.87990.86480.895022.75650.81770.79500.6127WC**0.86950.85450.884474.25000.70500.87450.5804ABSI**ARI**0.61710.89120.59160.87750.64260.904875.792512.78250.59260.77350.59620.85770.18880.6312OBMI+ABSI**0.89120.87750.904830.41430.77330.85770.6310OBMI+WC0.88880.87500.902734.12340.77160.85560.6272BMI*ABSI0.88840.87480.90211704.21000.79220.82850.6207BMI*WC0.88880.87500.90271674.11830.77330.85560.6286MaleBMI**0.81600.80550.826423.55550.73820.73090.4691WC**0.81020.79980.820780.65000.66740.80340.4708ABSI**0.57950.56530.593674.91280.40520.72400.1292ARI**0.82700.81700.836913.60960.74560.74570.4913OBMI+ABSI**0.82700.81700.836933.25690.74580.74570.4915OBMI+WC0.82420.81410.834339.83130.74380.74470.4885BMI*ABSI0.82570.81570.83561758.75900.67610.81170.4878BMI*WC0.82440.81430.83451944.04330.75510.73290.4880Abbreviations as in Table 1**P < 0.05 compared with BMI*WC in each gender\n\nThe best threshold, sensitivities, specificities, Youden’s index, and area under the curve of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI in different genders\nABSI**\nARI**\n0.6171\n0.8912\n0.5916\n0.8775\n0.6426\n0.9048\n75.7925\n12.7825\n0.5926\n0.7735\n0.5962\n0.8577\n0.1888\n0.6312\nAbbreviations as in Table 1\n**P < 0.05 compared with BMI*WC in each gender\nTo explore the changes in BMI, WC, ABSI, ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC on the risk identification ability of NAFLD in different age subgroups of both sexes, we constructed ROC curves for 20–39, 40–59, and ≥ 60 years in both sexes, respectively; and the corresponding AUC and optimal diagnostic thresholds were summarized in Table 6. We found that ARI and OBMI+ABSI exhibited considerably higher AUCs in all age subgroups for both sexes except in the female ≥ 60-year age group; the AUC of OBMI+ABSI in the female 20–39 age group was 0.9523, the highest value among all age subgroups of both sexes. In the male 20-59-year-old group and the female 40-59-year-old group, the AUC values ​​of ARI and OBMI+ABSI were slightly higher than those of BMI*WC, but all were significantly higher than those of BMI or WC or ABSI alone, while in the female 20–39 years group and the female ≥ 60 years group AUC values for all combined obesity indicators were significantly higher than that of WC and ABSI; in addition, in the male ≥ 60-year-old group, the seven indicators of obesity had similar NAFLD identification abilities, only ABSI underperformed. Therefore, the combined indicators ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, and OBMI+WC can significantly improve the ability of simple obesity parameters BMI and WC and ABSI to identify NAFLD in middle-aged females and young and middle-aged males and had the highest diagnostic performance in young females.\n\nTable 6The area under the curve, best threshold, sensitivities, specificities, and Youden’s index of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI as a predictor of NAFLD in the different age groups of both gendersAUC95%CI low95%CI upBest thresholdSpecificitySensitivityYouden’s indexFemaleAge 20–39 yearsBMI0.95110.93360.968523.12660.89810.87230.7704WC***0.93190.90790.955976.60000.85600.88300.7390ABSI***0.56860.50850.628875.57420.61220.53190.1441ARI0.95220.93370.970812.79220.84980.93620.7860BMI*WC0.95160.93310.97011675.47840.84360.92550.7691BMI*ABSI0.94860.92890.96821701.34860.86140.92550.7869OBMI+ABSI0.95230.93370.970830.43680.84980.93620.7860OBMI+WC0.95160.93300.970134.12960.84240.92550.7679Age 40–59 yearsBMI***0.84840.82820.868622.75270.78410.77440.5585WC***0.84230.82300.861674.25000.66300.86910.5321ABSI***0.61860.58970.647675.79250.58080.60720.1880ARI***0.86220.84380.880513.02830.80770.77440.5821BMI*WC0.86030.84170.87881674.11830.73320.83840.5716BMI*ABSI0.86020.84200.87851704.21000.75460.81340.5680OBMI+ABSI***0.86220.84380.880530.99950.80790.77440.5823OBMI+WC0.86020.84170.878835.03630.80930.76320.5725Age ≥ 60 yearsBMI0.83080.74580.915923.33340.84390.76000.6039WC***0.76960.67780.861379.75000.77070.68000.4507ABSI***0.56600.45510.677078.18360.50240.72000.2224ARI0.81950.73910.899912.92520.70240.84000.5424BMI*WC0.81990.73960.90021686.12800.69760.84000.5376BMI*ABSI0.80140.71860.88411659.09940.56100.96000.5210OBMI+ABSI0.81950.73920.899830.75250.70240.84000.5424OBMI+WC0.82010.73930.900934.33930.70240.84000.5424MaleAge 20–39 yearsBMI***0.84390.82750.860223.47820.74950.76650.5160WC***0.84150.82550.857580.45000.72190.80090.5228ABSI***0.60630.58260.630074.50900.48710.69910.1862ARI***0.85490.83960.870213.50260.76000.78440.5444BMI*WC0.85130.83570.86691862.54150.72190.81140.5333BMI*ABSI***0.85520.83990.87041766.28860.75570.78740.5431OBMI+ABSI***0.85490.83960.870233.00760.76140.78290.5443OBMI+WC0.85140.83580.866939.14530.72900.80840.5374Age 40–59 yearsBMI***0.80070.78670.814723.25050.67790.76390.4418WC***0.79080.77660.805181.25000.65830.77950.4378ABSI***0.56150.54290.580175.42940.38970.70050.0902ARI***0.81040.79690.824013.61420.72320.74510.4683BMI*WC0.80800.79430.82171955.12030.74480.71770.4625BMI*ABSI0.80860.79500.82221816.10160.73340.72870.4621OBMI+ABSI***0.81050.79690.824033.27330.72390.74430.4682OBMI+WC0.80750.79380.821240.11920.74750.71850.4660Age ≥ 60 yearsBMI0.79030.73630.844423.74200.81110.69510.5062WC0.78040.72820.832680.50000.59440.86590.4603ABSI***0.58040.51190.649075.85420.29410.86590.1600ARI0.79490.74370.846213.58190.67800.81710.4951BMI*WC0.80290.75130.85451932.61210.73680.79270.5295BMI*ABSI0.79040.73930.84141800.36700.65940.82930.4887OBMI+ABSI0.79500.74380.846233.18800.67800.81710.4951OBMI+WC0.80230.75090.853639.74760.71210.81710.5292Abbreviations as in Table 1***P < 0.05 compared with BMI*WC in each age group of both genders\n\nThe area under the curve, best threshold, sensitivities, specificities, and Youden’s index of BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, BMI, WC, ABSI, and ARI as a predictor of NAFLD in the different age groups of both genders\nAbbreviations as in Table 1\n***P < 0.05 compared with BMI*WC in each age group of both genders", "Our main findings showed that BMI, WC, ABSI, ARI, BMI*WC, BMI*ABSI, OBMI+ABSI, OBMI+WC, and the obesity phenotypes were all significantly and positively correlated with NAFLD risk; and for the first time in the general population, it was found that BMI combined with WC and ABSI can significantly improve the ability of the basic anthropometric measures to identify NAFLD, especially in middle-aged females and young and middle-aged males. Of significant mention, compared with other age stages in both sexes, all obesity indicators in this study have the highest diagnostic efficiency among young females.\nIn recent years, the rapid global economic development, dietary patterns and lifestyles of the residents have changed dramatically, and a high-calorie, high-fat daily diet and a sedentary lifestyle have become the main themes of today’s society; therefore, the prevalence of metabolic syndrome, obesity, and other obesity-related diseases has increased greatly [2, 32, 33]. NAFLD is considered the hepatic manifestation of the metabolic syndrome and encompasses a complex spectrum of liver pathologies ranging from simple hepatic steatosis to non-alcoholic steatohepatitis, and also the most common chronic liver disease and the leading cause of liver-related death worldwide [1, 34]. The occurrence and development of NAFLD are closely related to the global epidemic of obesity [6]. Recent epidemiological surveys have shown that the incidence of NAFLD in obese patients can reach 80%, and in severely obese patients who require bariatric surgery, the incidence of NAFLD is as high as 90% [4].\nIndependent associations between NAFLD and traditional obesity indicators BMI and WC have been demonstrated in many observational studies, but there is much debate as to which indicator is better at identifying obesity-related NAFLD risk [7–10, 35–39]. Several recent studies have demonstrated that BMI was a stronger indicator of obesity for identifying and predicting NAFLD risk, with BMI having the largest AUC compared with other obesity-related parameters, showing better diagnostic performance than WC and other obesity indicators [37–39]. Of course, there were other studies that expressed the opposite standpoints; two studies of NAFLD in obese adolescents and children found that both WC and BMI independently predicted NAFLD risk, with WC, in particular, having the highest predictive accuracy [35, 36]. Furthermore, a meta-analysis study by Qing et al. found that WC remained the strongest anthropometric predictor of NAFLD even after adjusting for a large number of confounding factors, including BMI, and that central obesity was a more dangerous pattern of body fat distribution than general obesity [7]. Clearly, both BMI and WC were independent risk factors for NAFLD, but neither BMI nor WC alone appeared to be an adequate measure of obesity-related NAFLD risk. In a study by Janssen et al. [40], it was found that either BMI or WC alone was an independent predictor of total fat, non-abdominal fat, abdominal subcutaneous fat, and visceral fat, but 20–40% of the variation in fat mass in these fat pools remained unexplained by BMI or WC alone; after they further combining BMI with WC, found that the variability of fat content in these fat pools decreased significantly. Given that the available evidence generally supports both abdominal subcutaneous fat and visceral fat as independent predictors of insulin resistance [41, 42], we hypothesized that the combination of WC and BMI may be a better predictor of metabolic disease. This speculation has been confirmed in studies of metabolic diseases such as cardiovascular disease, obesity-related hypertension, type 2 diabetes, and risk of all-cause mortality [14–17]. Furthermore, in a recent study, Wang et al. also found that BMI combined with WC was more strongly associated with NAFLD in a diabetic population than either BMI or WC alone [18]. However, their study subjects were limited to the diabetic population and did not explore the value of combined BMI and WC in the identification of NAFLD. In addition, it is also worth noting that in the above-mentioned studies related to BMI combined with WC, almost all researchers ignored the statistical artifacts that may result from the strong correlation between BMI and WC. A recent study by Christakoudi S et al. found a traditional U-shaped association between BMI alone and all-cause mortality risk, but when BMI was combined with WC, which had a strong correlation, a predominantly negative association was found between BMI and all-cause mortality, i.e., the higher the BMI the lower the mortality risk [19]. It means that the combination of WC and BMI may not simply supplement BMI, but even changed the pattern of associations between BMI and outcome events, which may be a statistical artifact caused by the strong correlation between WC and BMI. In addition, when they combined BMI with the moderately correlated waist index waist-to-height ratio found a similar but more modest change in the pattern of association between BMI and all-cause mortality. Therefore, combining BMI with a correlated abdominal obesity index may lead to biased and possibly even misleading risk estimates and ineffective risk stratification [19]. As expected, when the researchers further combined BMI with the unrelated abdominal obesity index ABSI and hip circumference, they found that the pattern of association between BMI and all-cause mortality did not change and that more fine-grained risk stratification was achieved for ABSI in each BMI stratum. ABSI is a reliable predictor of diseases such as cardiovascular disease and all-cause mortality and a valid proxy for identifying sarcopenic obesity, independent of BMI by design, and is a useful complement to BMI for risk estimation and risk stratification [22, 43–45].\nIn this study, we also analyzed the correlations between BMI and WC and ABSI, and in agreement with the results of previous studies [19], there was a strong correlation between BMI and WC and almost no correlation between BMI and ABSI (r < 0.05); in addition, in the association analysis of single indicators with NAFLD risk we found that BMI, WC, and ABSI were all significantly and positively associated with the risk of NAFLD (Table 4). Therefore, the combination of BMI and ABSI may be more appropriate than the combination of BMI and WC. Nevertheless, to fully explore the value of combining BMI and the abdominal obesity index to assess the risk of NAFLD and to circumvent possible statistical artifacts, we still combined BMI with WC and ABSI in 4 separate ways based on previous studies: First, the combination of BMI and WC into four obesity phenotypes, BMIN/WCN, BMIO/WCN, BMIN/WCO, and BMIO/WCO, based on the overweight cut-offs for WC and BMI in Asian populations recommended by the WHO Expert Committee [27, 28]. Second, constructing optimal proportional combinations OBMI+WC and OBMI+ABSI based on the regression coefficients of BMI, WC, and ABSI in multivariate logistic regression models. Third, multiplied BMI with WC and ABSI directly to obtain BMI*WC and BMI*ABSI. Fourth, a predictor ARI (BMI, ABSI) was calculated using the method developed by Krakauer NY et al. [26]. In the analysis of the association between each obesity phenotype and NAFLD we found that after adjusting for a large number of confounders (Model 3), subjects with normal BMI and WC in both sexes had the lowest risk of NAFLD, both the BMIO/WCN phenotype and the BMIN/WCO phenotype increased the risk of NAFLD, and subjects with the BMIO/WCO phenotype had the highest risk of NAFLD (male: OR 4.19, 95%CI 3.54, 4.96; female: OR 9.78, 95%CI 7.35, 13.02); these results were in accord with previous studies [18]. Obviously, comprehensive consideration of the effects of BMI and WC on NAFLD in the general population could explain the obesity-related NAFLD risk to a greater extent, identifying more populations at risk for developing NAFLD. Additionally, all other combined indicators in this study were significantly and positively correlated with NAFLD, with BMI*ABSI having the strongest correlation among males and BMI*WC having the strongest correlation with NAFLD among females. The results also did not change after treating BMI*ABSI as a categorical variable, but it is worth mentioning that the Q4 category has a much higher risk of NAFLD in females than in males compared with the Q1 category in BMI*ABSI (OR: female 63.97 vs. male 26.15). Consistently, in the obesity phenotypes, female subjects with BMIO/WCO phenotype also had a significantly higher risk of NAFLD than males (OR: female 9.78 vs. male 4.19). This suggested that elevated BMI and WC and ABSI may be more important risk factors for NAFLD in females, and females should pay more attention to maintaining normal body weight and body shape to prevent the occurrence of NAFLD.\nOur current study also compared the ability of five continuous combined indicators, ARI, OBMI+WC, OBMI+ABSI, BMI*ABSI, and BMI*WC, to identify NAFLD with BMI and WC and ABSI alone. According to the results of ROC analysis, ARI and OBMI+ABSI had the strongest NAFLD identification ability in both sexes, and although there were only minor differences between the two and OBMI+WC, BMI*WC and BMI*ABSI, they were all significantly higher than BMI and WC and ABSI alone. From Table 5 we found that the optimal proportional combination indicators (OBMI+WC and OBMI+ABSI) have extremely similar diagnostic efficacy to the multiplicative indicators (BMI*WC and BMI*ABSI) and ARI, which was an interesting result. The optimal proportional combination indicators in this study were calculated after directly obtaining the proportional coefficients based on the ratio of the regression coefficients of BMI and WC or ABSI in model 3, which considered the relative weights of the effects of BMI and WC or ABSI on NAFLD and could fully reflect the effects of BMI combined with WC and ABSI on NAFLD. However, the construction of the calculation formula for the optimal proportional combination was relatively complicated, which may be affected by different adjusted confounding factors and changes in the correlation between BMI, WC, ABSI and NAFLD in different test groups. In contrast, the ARI (BMI, ABSI) was obtained by multiplying the logarithms of ORs of BMI and ABSI by the corresponding BMI and ABSI values ​​of each subject and then summing them together [26] and the multiplicative combination indicators were obtained by directly multiplying BMI with WC and ABSI, which are easy to calculate and had a high diagnostic performance in both sexes. In summary, ARI and multiplicative combination indicators (BMI*WC and BMI*ABSI) may be better obesity indicators for clinical screening of NAFLD. Additionally, the results of the ROC analysis after further grouping the sexes by different age groups showed that all combination indicators of BMI and ABSI or WC significantly improved the ability of a single indicator to identify NAFLD in middle-aged females and young and middle-aged males. It is worth mentioning that the highest diagnostic efficacy for NAFLD was found in the young female population for both combined and single obesity indicators; this may be related to the protective effect of large amounts of estrogen on NAFLD in young females. As we all know, the reproductive status of females is closely related to their health status. Experimental studies have shown that estrogens can exert antioxidant, anti-steatosis, and anti-fibrotic effects in the liver [46, 47]. Whereas, a significant decrease in estrogen levels and an increase in circulating androgen levels in postmenopausal females may lead to disorders of lipid metabolism and the development of metabolic syndrome [48]. In addition, the decline in the body’s metabolic status with aging is also an important risk factor for NAFLD [49]. Thus, younger females have a healthier metabolic profile and fewer confounding factors that affect the risk of NAFLD identified by obesity indicators compared with postmenopausal females and males.\nOur findings have important implications for both clinical screening and epidemiological studies of NAFLD. Due to the insidious onset of NAFLD, early-stage patients may be asymptomatic, primary care workers lack attention to NAFLD, and public awareness and treatment rates are very low [50, 51]; moreover, there are no approved treatments for NAFLD, hence the mainstay of prevention and treatment of NAFLD remains healthy lifestyle and weight control [1]. In the current study, ARI and BMI*WC and BMI*ABSI had greater than 80% diagnostic accuracy in all age groups of both sexes and up to 95.22% in young women, and ARI and BMI*WC and BMI*ABSI were easily calculated without additional measurements or equipment requirements. Therefore, we recommend that the ARI and BMI*ABSI and BMI*WC should be incorporated into the clinical screening plan for NAFLD, which will greatly improve the detection rate of NAFLD.\n[SUBTITLE] Advantages and limitations of research [SUBSECTION] Several advantages of the current study need to be mentioned: (1) The current study demonstrated for the first time in a large sample of the general population that BMI combined with WC and ABSI had a stronger ability to identify NAFLD than a basic anthropometric obesity parameter. (2) After combining BMI with WC and ABSI in various ways, we found that the ARI and multiplicative combination indicators (BMI*WC and BMI*ABSI) may be the more appropriate obesity index for clinical screening of NAFLD. These findings provided an important reference for the application of BMI combined with the abdominal obesity index for NAFLD screening in the general population.\nOf course, it is undeniable that the current study has several limitations: (1) The current study was a cross-sectional study, so we can only demonstrate that BMI combined with the abdominal obesity index was stronger than a single indicator for identifying NAFLD in the general population, but whether the predictive performance for NAFLD risk is stronger needs to be validated in a longitudinal cohort study. (2) The diagnosis of NAFLD, the main outcome in this study, was based on ultrasonography, which may have missed some patients with NAFLD [1]. However, in the setting of fewer NAFLD cases, we still demonstrated that BMI combined with the abdominal obesity index had a higher diagnostic performance for NAFLD than a single indicator. (3) Although we systematically adjusted for known NAFLD risk factors, there may be residual confounding due to the limitations of retrospective studies where some NAFLD risk factors could not be measured and obtained.\nSeveral advantages of the current study need to be mentioned: (1) The current study demonstrated for the first time in a large sample of the general population that BMI combined with WC and ABSI had a stronger ability to identify NAFLD than a basic anthropometric obesity parameter. (2) After combining BMI with WC and ABSI in various ways, we found that the ARI and multiplicative combination indicators (BMI*WC and BMI*ABSI) may be the more appropriate obesity index for clinical screening of NAFLD. These findings provided an important reference for the application of BMI combined with the abdominal obesity index for NAFLD screening in the general population.\nOf course, it is undeniable that the current study has several limitations: (1) The current study was a cross-sectional study, so we can only demonstrate that BMI combined with the abdominal obesity index was stronger than a single indicator for identifying NAFLD in the general population, but whether the predictive performance for NAFLD risk is stronger needs to be validated in a longitudinal cohort study. (2) The diagnosis of NAFLD, the main outcome in this study, was based on ultrasonography, which may have missed some patients with NAFLD [1]. However, in the setting of fewer NAFLD cases, we still demonstrated that BMI combined with the abdominal obesity index had a higher diagnostic performance for NAFLD than a single indicator. (3) Although we systematically adjusted for known NAFLD risk factors, there may be residual confounding due to the limitations of retrospective studies where some NAFLD risk factors could not be measured and obtained.", "Several advantages of the current study need to be mentioned: (1) The current study demonstrated for the first time in a large sample of the general population that BMI combined with WC and ABSI had a stronger ability to identify NAFLD than a basic anthropometric obesity parameter. (2) After combining BMI with WC and ABSI in various ways, we found that the ARI and multiplicative combination indicators (BMI*WC and BMI*ABSI) may be the more appropriate obesity index for clinical screening of NAFLD. These findings provided an important reference for the application of BMI combined with the abdominal obesity index for NAFLD screening in the general population.\nOf course, it is undeniable that the current study has several limitations: (1) The current study was a cross-sectional study, so we can only demonstrate that BMI combined with the abdominal obesity index was stronger than a single indicator for identifying NAFLD in the general population, but whether the predictive performance for NAFLD risk is stronger needs to be validated in a longitudinal cohort study. (2) The diagnosis of NAFLD, the main outcome in this study, was based on ultrasonography, which may have missed some patients with NAFLD [1]. However, in the setting of fewer NAFLD cases, we still demonstrated that BMI combined with the abdominal obesity index had a higher diagnostic performance for NAFLD than a single indicator. (3) Although we systematically adjusted for known NAFLD risk factors, there may be residual confounding due to the limitations of retrospective studies where some NAFLD risk factors could not be measured and obtained.", "Taken together, our findings confirmed that BMI combined with ABSI and WC can better explain obesity-related NAFLD risk than a single indicator in the general population, and identified the ARI and the multiplicative combination indicators BMI*ABSI and BMI*WC as the simplest and very effective combination indicators for diagnosing the risk of NAFLD. These new findings underscored the importance of the combined application of BMI and the abdominal obesity index in clinical practice to assess NAFLD risk in the general population and provided a cost-effective tool for precise preventive screening and treatment monitoring for NAFLD.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\n\nSupplementary Material 2\n\nSupplementary Material 2" ]
[ null, null, null, null, null, null, null, null, "results", null, null, null, null, "discussion", null, "conclusion", "supplementary-material", null ]
[ "Body mass index", "Waist circumference", "ABSI", "ARI", "Receiver operating characteristic analysis", "Non-alcoholic fatty liver disease" ]
The blacksmith approach: a strategy for teaching and learning in the medical anatomy course (a qualitative study).
36266705
Anatomy is a symbolic, essential core topic and one of the fundamental pillars of medical and paramedical knowledge. Nevertheless, few exploratory data analyses have focused on how students approach learning anatomy. This study examined how students perceive their learning experience during anatomy lessons and how to make a model which promotes their meaningful learning and professional identity.
BACKGROUND
Using purposive sampling with maximum variation, we conducted a qualitative content analysis at the Shiraz University of Medical Sciences in Iran (2020 to 2021). Twenty-four medical students and twelve faculty members of Iran's medical science universities were enrolled in the study. The data were collected through semi-structured interviews and analyzed according to the theme.
METHODS
A conceptual model emerged from the data analysis with the main theme called the blacksmith approach, which included Three sub-themes: (1) making a new forge (adequate preparation and mindful beginning), (2) heating the students' hearts (considering supporting systems that learners need) and (3) using Sledgehammer's approach (teaching anatomy by using more active methods and engaging all neuroanatomical regions) and (Using fun for enjoyable learning). All the concepts were related to each other.
RESULTS
Medical students experience a challenging fundamental evolution into professional doctors. Educational systems focus primarily on teaching and learning, while students' transition can be facilitated by a three-step model called the Blacksmith Approach. It best serves as an educational framework for any pivotal, preclinical course capable of helping students acquire new roles and tackle challenges. Further research should be conducted to confirm how hard work leads to satisfying results with the opportunity to create enjoyable learning.
CONCLUSION
[ "Humans", "Learning", "Students, Medical", "Education, Medical, Undergraduate", "Qualitative Research", "Curriculum", "Teaching", "Anatomy" ]
9584281
Introduction
Anatomy, as a branch of biological and medical sciences, is one of the introductory courses in the learning experience of undergraduate medical students, which is essential for impending clinical practice and clinical proficiency [1, 2]. A challenging question facing medical education is how to change some young students into responsible and resilient professional physicians. Most of the answers are being placed in clinical years; however, we can begin the transition process earlier with a promising tool called anatomy [3, 4]. Despite being one of the fundamental pillars of medical knowledge, anatomy has been pulled back as a simple course in the preclinical period. Historically, ancient medical schools have relied heavily on philosophical and intuitive systems of thought [5]. Experimental investigations by dissection helped physicians actualize the human body as a combination of multiple organ systems. [6]. We can assume that anatomy science has led to reform in how medicine views the human being from a spiritual, holistic being into a biopsychosocial person. However, anatomy has been underestimated in current medical pedagogy [7, 8]. Recent evidence indicates that we can reform anatomy courses making them as influential as they can be. [9–11]. Anatomy and dissection sessions can act as students’ first encounters with the humanized face of medicine, where medical students can learn teamwork, morality, coping strategies, and communication skills. In addition, anatomy can be perceived as a way of internalizing self-awareness, medical epistemology, empathy, and medical ethics [6, 12, 13]. While a myriad of teaching approaches are available for anatomy education, they have changed according to the needs of learners or global crises like covid-19. The primary approaches include lecture, dissection, and demonstration of cadavers. In addition, many schools incorporated radiological imaging as a learning tool for teaching anatomy in the living body. Considering the shortcomings of traditional methods, medical schools started implementing more innovative techniques that enhanced active learning. While Problem-based learning, flipped classroom, and reflective writing activities on the cadaveric dissection fostered communication skills and self-directed learning [12, 14, 15], other methods focused on students’ learning styles: drawing and whiteboarding, three-dimensional printing (3DP), and digital models for visual learners; play-doh, role modeling and peer physical exam for tactile learning; singing, dancing, yoga, and pilates for kinaesthetic learners [16–18]. The covid pandemic disrupted anatomy education as the number of donated cadavers decreased substantially, and social distancing prevented in-person classes or labs [17–19]. Accordingly, the teaching approaches relied, more than ever, on technology leading to the widespread use of computer-assisted learning, web-based learning via social media (esp. Facebook and youtube), and a more prominent role for Augmented-Reality and Virtual-Reality [20, 21]. The main focus behind all these methods and strategies is better teaching anatomy for its sake. However, such an approach does not allow us to use the full potential of anatomy science as a core element in changing young students into self-confident and resilient learners, which leads to the creation of future professional physicians [22, 23]. Physicians need professional competencies, a set of which essential competencies, including professional resilience, is and emotional competencies, to achieve goals. As a capacity to endure difficulties and quickly improve from a stress-inducing experience, resilience helps one to make valuable adaptations to problems and can utilize as a protective defense against adversities, thus facilitating welfare [23, 24]. Resilience is referred to as one’s capability to maintain and improve their well-being when faced with life challenges. Resilience can be considered a behavior acquired during training and various courses, including the main course of anatomy. Resilience includes cognitive processes and has four dimensions, including self-efficacy. Planning, Self-control, Commitment, and perseverance include the following: young doctors must be present in various educational environments and support clinical and educational supervisors in enduring difficulties to help them develop personal and professional resilience [24, 25]. In Iran and at Shiraz University, anatomy courses entail three steps. Initially, a topic is introduced through face-to-face, online or offline lectures with a slide or a short video clip. Next, students work on anatomical modeling and, finally, on a cadaver [26, 27]. Today, not only the medical students must bear a significant load of details in anatomy courses, which can reduce their motivation, but they should also face the educational consequences of the covid pandemic, which include loss of hands-on learning activities and limited access to tools like cadavers, models, pathology specimens, and skeletons [28–34]. Losing face-to-face contact and direct interaction with peers and teachers may hinder the students’ growth as future physicians [6, 13]. Therefore, finding ways to improve the learning experience is more challenging than ever.
null
null
Results
[SUBTITLE] Participants’ characteristics [SUBSECTION] In this study, semi-structured interviews were performed with 24 medical students from year 1 to senior interns, 15 (62.5% were males), 9 (37.5% were females), and 12 faculty members, 6 (50% were males), 6 (50% were females), with various academic ranks and experience from 9 (Shiraz, Tehran, Fasa, Jahrom, Bandarabas, Yasuj, Gerash, Larestan, and Bushehr) Iranian medical schools. The participants’ age ranged from 19 to 62 years old, with a mean of 33.4 ± 8.4 years. Based on the results, 12 students (50%) were in the clinical training period as students, externs, and interns, and 12 participants (50%) were in the basic sciences period. Accordingly, five participants (20%) had a history of being top students, and four (16.7%) had a history of dropout in anatomy courses. Furthermore, the work experience of the faculty members ranged from 5 to 35 years in teaching anatomy; 9(75%) anatomy faculty members and 3(25%) experts in medical education; faculty member participants, 3(25%) were single, and 9 (75%) were married. (Table 1) Table 1Demographic Characteristics of the Study ParticipantsParticipantsCharacteristics students Gender NoPercent (%) Mean Age (y ± SD) Male1562.522.3 ± 8.4Female937.5 Degree Faculty membersassistant professors,32537.4 ± 11.7associated professors541.67full professors433.34 Demographic Characteristics of the Study Participants From all the interviews, 334 codes were extracted. After the dismissal of similar codes and their integration, the experience of medical students was conceptualized into a central theme called the blacksmith approach, which included Three sub-themes: (1) making a new forge (Adequate preparation and mindful beginning), (2) heating the students’ hearts (considering supporting systems that learners need) and (3) using a Sledgehammer’s approach (teaching Anatomy thorough engaging all neuroanatomical regions in the Age of Pandemics). All the concepts were related to each other and resulted in a pattern revealing the experience of students and faculty members on strategies for successful learning and teaching of anatomy lessons. The main themes, as well as subordinate themes, are listed in Table 2. Table 2The Blacksmith Approach: Best strategy for teaching and learning in the medical anatomy course. Themes, sub-themes, and codesMain ThemesSub Themescodecods Blacksmith Approach Redesigning Educational Mindset (Making a New Forge) Creating a Vision for Students √ Anatomy is hard, bulky, should be taken seriously√ I can help myself more than anyone√ integrated curriculum√ self-study / Self-confidencescarcely happens in the real world!√ professional identity√ prospective physicians Mindful Clinical View Role Modeling and Support (Heating up the Students’ Hearts) Joyful Teaching (Telling Stories/Games/ Humor) √ every trick must be done√ enjoy class√ follow me in the classroom Student Support System √ susceptible students√ assistance√ Vulnerable√ right supportersSitting and talking to them Sledgehammer Approach (Thorough Engagement in the Age of Pandemics) Study Guide √ Preparedness√ Road map Reference Autonomy √ self-learning√ self-direction Neuroanatomical Engagement √ Visual Learning√ Auditory Learning√ Sensory Learning√ Brain HemispheresLong-term Memorization Mindful Dissection, Better Transition √ It smells so bad, but we need the observation√ We got used to it√ ethically important√ respect the dead body√ respect to the living body Drawing √ easy to learn√ Draw a lot Problem-Based Learning √ Look at the patient’s family√ Deep worry√ What is the next step of the problem? Peer-Assisted Learning √ Peer to the peer group√ Discussing the problem The Blacksmith Approach: Best strategy for teaching and learning in the medical anatomy course. Themes, sub-themes, and codes √ Anatomy is hard, bulky, should be taken seriously √ I can help myself more than anyone √ integrated curriculum √ self-study / Self-confidence scarcely happens in the real world! √ professional identity √ prospective physicians √ every trick must be done √ enjoy class √ follow me in the classroom √ susceptible students √ assistance √ Vulnerable √ right supporters Sitting and talking to them √ Preparedness √ Road map √ self-learning √ self-direction √ Visual Learning √ Auditory Learning √ Sensory Learning √ Brain Hemispheres Long-term Memorization √ It smells so bad, but we need the observation √ We got used to it √ ethically important √ respect the dead body √ respect to the living body √ easy to learn √ Draw a lot √ Look at the patient’s family √ Deep worry √ What is the next step of the problem? √ Peer to the peer group √ Discussing the problem In this study, semi-structured interviews were performed with 24 medical students from year 1 to senior interns, 15 (62.5% were males), 9 (37.5% were females), and 12 faculty members, 6 (50% were males), 6 (50% were females), with various academic ranks and experience from 9 (Shiraz, Tehran, Fasa, Jahrom, Bandarabas, Yasuj, Gerash, Larestan, and Bushehr) Iranian medical schools. The participants’ age ranged from 19 to 62 years old, with a mean of 33.4 ± 8.4 years. Based on the results, 12 students (50%) were in the clinical training period as students, externs, and interns, and 12 participants (50%) were in the basic sciences period. Accordingly, five participants (20%) had a history of being top students, and four (16.7%) had a history of dropout in anatomy courses. Furthermore, the work experience of the faculty members ranged from 5 to 35 years in teaching anatomy; 9(75%) anatomy faculty members and 3(25%) experts in medical education; faculty member participants, 3(25%) were single, and 9 (75%) were married. (Table 1) Table 1Demographic Characteristics of the Study ParticipantsParticipantsCharacteristics students Gender NoPercent (%) Mean Age (y ± SD) Male1562.522.3 ± 8.4Female937.5 Degree Faculty membersassistant professors,32537.4 ± 11.7associated professors541.67full professors433.34 Demographic Characteristics of the Study Participants From all the interviews, 334 codes were extracted. After the dismissal of similar codes and their integration, the experience of medical students was conceptualized into a central theme called the blacksmith approach, which included Three sub-themes: (1) making a new forge (Adequate preparation and mindful beginning), (2) heating the students’ hearts (considering supporting systems that learners need) and (3) using a Sledgehammer’s approach (teaching Anatomy thorough engaging all neuroanatomical regions in the Age of Pandemics). All the concepts were related to each other and resulted in a pattern revealing the experience of students and faculty members on strategies for successful learning and teaching of anatomy lessons. The main themes, as well as subordinate themes, are listed in Table 2. Table 2The Blacksmith Approach: Best strategy for teaching and learning in the medical anatomy course. Themes, sub-themes, and codesMain ThemesSub Themescodecods Blacksmith Approach Redesigning Educational Mindset (Making a New Forge) Creating a Vision for Students √ Anatomy is hard, bulky, should be taken seriously√ I can help myself more than anyone√ integrated curriculum√ self-study / Self-confidencescarcely happens in the real world!√ professional identity√ prospective physicians Mindful Clinical View Role Modeling and Support (Heating up the Students’ Hearts) Joyful Teaching (Telling Stories/Games/ Humor) √ every trick must be done√ enjoy class√ follow me in the classroom Student Support System √ susceptible students√ assistance√ Vulnerable√ right supportersSitting and talking to them Sledgehammer Approach (Thorough Engagement in the Age of Pandemics) Study Guide √ Preparedness√ Road map Reference Autonomy √ self-learning√ self-direction Neuroanatomical Engagement √ Visual Learning√ Auditory Learning√ Sensory Learning√ Brain HemispheresLong-term Memorization Mindful Dissection, Better Transition √ It smells so bad, but we need the observation√ We got used to it√ ethically important√ respect the dead body√ respect to the living body Drawing √ easy to learn√ Draw a lot Problem-Based Learning √ Look at the patient’s family√ Deep worry√ What is the next step of the problem? Peer-Assisted Learning √ Peer to the peer group√ Discussing the problem The Blacksmith Approach: Best strategy for teaching and learning in the medical anatomy course. Themes, sub-themes, and codes √ Anatomy is hard, bulky, should be taken seriously √ I can help myself more than anyone √ integrated curriculum √ self-study / Self-confidence scarcely happens in the real world! √ professional identity √ prospective physicians √ every trick must be done √ enjoy class √ follow me in the classroom √ susceptible students √ assistance √ Vulnerable √ right supporters Sitting and talking to them √ Preparedness √ Road map √ self-learning √ self-direction √ Visual Learning √ Auditory Learning √ Sensory Learning √ Brain Hemispheres Long-term Memorization √ It smells so bad, but we need the observation √ We got used to it √ ethically important √ respect the dead body √ respect to the living body √ easy to learn √ Draw a lot √ Look at the patient’s family √ Deep worry √ What is the next step of the problem? √ Peer to the peer group √ Discussing the problem [SUBTITLE] Central theme: blacksmith approach [SUBSECTION] According to the participants, in the student’s learning and maturity process, the teacher and the student act rationally and consciously and are aware of the factors that motivate and control them. Thus, they accept the risks, specific conditions, and hardships that must be tolerated. The student should handle this hardship to acquire a capacity to change, and the teacher should facilitate the change and their primary goal of teaching. In a sense, this is the same approach as the Blacksmiths. Blacksmithing symbolizes endurance, honor, and work under challenging conditions in our culture. (Fig. 1) We took the central theme from an interview with a professor who said: Fig. 1The role of the blacksmith approach in the stages of teaching and learning anatomy in medical students is based on the study’s results. First, the blacksmith (educational system) must prepare a new blacksmith (redesign the educational mentality and create a new perspective on anatomical knowledge in students) and prepare a new forge; then, he/she must heat the metal (supporting young students and having good role-modeling) and then shape the metal with a sledgehammer (the sledgehammer approach to using different teaching strategies and methods) to create a sword or self-confident and knowledgeable learner in medicine (professionalism) ”…The Sledgehammer should be heavy. Its nature is heavy; otherwise, it will not be capable of smoothing any iron. The nature of some courses, such as anatomy, is challenging, but we must make it enjoyable for the student….“ (FM no.6). The role of the blacksmith approach in the stages of teaching and learning anatomy in medical students is based on the study’s results. First, the blacksmith (educational system) must prepare a new blacksmith (redesign the educational mentality and create a new perspective on anatomical knowledge in students) and prepare a new forge; then, he/she must heat the metal (supporting young students and having good role-modeling) and then shape the metal with a sledgehammer (the sledgehammer approach to using different teaching strategies and methods) to create a sword or self-confident and knowledgeable learner in medicine (professionalism) ”…The Sledgehammer should be heavy. Its nature is heavy; otherwise, it will not be capable of smoothing any iron. The nature of some courses, such as anatomy, is challenging, but we must make it enjoyable for the student….“ (FM no.6). According to the participants, in the student’s learning and maturity process, the teacher and the student act rationally and consciously and are aware of the factors that motivate and control them. Thus, they accept the risks, specific conditions, and hardships that must be tolerated. The student should handle this hardship to acquire a capacity to change, and the teacher should facilitate the change and their primary goal of teaching. In a sense, this is the same approach as the Blacksmiths. Blacksmithing symbolizes endurance, honor, and work under challenging conditions in our culture. (Fig. 1) We took the central theme from an interview with a professor who said: Fig. 1The role of the blacksmith approach in the stages of teaching and learning anatomy in medical students is based on the study’s results. First, the blacksmith (educational system) must prepare a new blacksmith (redesign the educational mentality and create a new perspective on anatomical knowledge in students) and prepare a new forge; then, he/she must heat the metal (supporting young students and having good role-modeling) and then shape the metal with a sledgehammer (the sledgehammer approach to using different teaching strategies and methods) to create a sword or self-confident and knowledgeable learner in medicine (professionalism) ”…The Sledgehammer should be heavy. Its nature is heavy; otherwise, it will not be capable of smoothing any iron. The nature of some courses, such as anatomy, is challenging, but we must make it enjoyable for the student….“ (FM no.6). The role of the blacksmith approach in the stages of teaching and learning anatomy in medical students is based on the study’s results. First, the blacksmith (educational system) must prepare a new blacksmith (redesign the educational mentality and create a new perspective on anatomical knowledge in students) and prepare a new forge; then, he/she must heat the metal (supporting young students and having good role-modeling) and then shape the metal with a sledgehammer (the sledgehammer approach to using different teaching strategies and methods) to create a sword or self-confident and knowledgeable learner in medicine (professionalism) ”…The Sledgehammer should be heavy. Its nature is heavy; otherwise, it will not be capable of smoothing any iron. The nature of some courses, such as anatomy, is challenging, but we must make it enjoyable for the student….“ (FM no.6). [SUBTITLE] Sub theme1: redesigning educational mindset (making a new forge) [SUBSECTION] Creating an educational context is the first step for young students to begin their transition in the learning process. This context was found to have two aspects as follows: Creating a Vision for Students. Creating a Vision for Students. Some students mentioned a change in their mindset from the beginning with the idea that they have a difficult task ahead and should know what is helpful or not. In addition, realizing that the student is the most important person to help themselves seemed to provide a better sense of success. According to the participants, anatomy is one of the most voluminous and complex courses in medicine, which students should take seriously from the beginning.“Anatomy is hard, bulky, and should be taken seriously.“ (Student no. 9) “Anatomy is hard, bulky, and should be taken seriously.“ (Student no. 9) Even though many have oversimplified it, successful students have attempted to learn it through vision and self-awareness.“… After entering the school, my mentor advised me to study anatomy as soon as possible. An anatomy lesson is not something you would say I will study and comprehend the concepts overnight or for a week. You should attend the classes regularly. Students should provide accessible learning resources and not seek shortcuts…” " (Student no. 1). “… After entering the school, my mentor advised me to study anatomy as soon as possible. An anatomy lesson is not something you would say I will study and comprehend the concepts overnight or for a week. You should attend the classes regularly. Students should provide accessible learning resources and not seek shortcuts…” " (Student no. 1). Participants believed that successful students have good self-confidence and leadership in their learning process. Some students emphasized that the teachers can first build self-confidence and make them self-centered and self-regulated in learning. This concept requires the teachers to listen to them.“... If I realized the concepts satisfactorily, I would get a lot out of my effort. First, I accepted that anatomy is complicated, but I can help myself more than anyone. I started self-study. More action boosts self-confidence, which is required to become a doctor.“(Student no. 6) “... If I realized the concepts satisfactorily, I would get a lot out of my effort. First, I accepted that anatomy is complicated, but I can help myself more than anyone. I started self-study. More action boosts self-confidence, which is required to become a doctor.“(Student no. 6) Finally, the participants emphasized the importance of integration. Integration is simply defined as merging relevant content or subject areas. Teachers mention that the goal is to integrate and create a general perspective rather than a fragmented perspective of concepts. Some students believed that while multidisciplinary integration was provided, professors’ art of education, such as mastery of the teaching and learning process, generated the necessary harmony between the content presented. If well-coordinated, all aspects mutually reinforce each other, promoting learning in an integrated way.“... I enjoyed our integrated curriculum because that led to a better understanding of medicine. For example, discussing the anatomy of the gastrointestinal system gave me invaluable and integrated knowledge, and I enjoyed it ... “(Student no. 4) “... I enjoyed our integrated curriculum because that led to a better understanding of medicine. For example, discussing the anatomy of the gastrointestinal system gave me invaluable and integrated knowledge, and I enjoyed it ... “(Student no. 4) Anatomy professors acknowledged that integration with clinical courses had changed their perspective, reflected in their teaching.“…During the integration process and while I was listening to the viewpoints of the clinical colleagues, I realized what I thought to be the crucial thing that students should all be aware of scarcely happens in the real world!...“ (FM no. 2) “…During the integration process and while I was listening to the viewpoints of the clinical colleagues, I realized what I thought to be the crucial thing that students should all be aware of scarcely happens in the real world!...“ (FM no. 2) 2)Mindful Clinical View. Mindful Clinical View. Mindfulness is defined as awareness of what is happening at the present moment. Some professors believed that they must be aware of the ultimate goal while teaching at each moment. Therefore, mindfulness is influenced by one’s dreams.“…I try to teach anatomy in a way that our current students and prospective physicians can diagnose heart enlargement in the future…” (FM no. 7) “…I try to teach anatomy in a way that our current students and prospective physicians can diagnose heart enlargement in the future…” (FM no. 7) If anatomy teachers gain a conservative view, the students can achieve integrity and professional identity as doctors.“... The student’s learning and professional identity depend largely on knowing and comprehending anatomy. Teachers can play the role of a good teacher in any situation, whether a crisis (like the Covid-19 pandemic) or not. A good teacher always applies the right approach....“ (FM no. 1). “... The student’s learning and professional identity depend largely on knowing and comprehending anatomy. Teachers can play the role of a good teacher in any situation, whether a crisis (like the Covid-19 pandemic) or not. A good teacher always applies the right approach....“ (FM no. 1). Creating an educational context is the first step for young students to begin their transition in the learning process. This context was found to have two aspects as follows: Creating a Vision for Students. Creating a Vision for Students. Some students mentioned a change in their mindset from the beginning with the idea that they have a difficult task ahead and should know what is helpful or not. In addition, realizing that the student is the most important person to help themselves seemed to provide a better sense of success. According to the participants, anatomy is one of the most voluminous and complex courses in medicine, which students should take seriously from the beginning.“Anatomy is hard, bulky, and should be taken seriously.“ (Student no. 9) “Anatomy is hard, bulky, and should be taken seriously.“ (Student no. 9) Even though many have oversimplified it, successful students have attempted to learn it through vision and self-awareness.“… After entering the school, my mentor advised me to study anatomy as soon as possible. An anatomy lesson is not something you would say I will study and comprehend the concepts overnight or for a week. You should attend the classes regularly. Students should provide accessible learning resources and not seek shortcuts…” " (Student no. 1). “… After entering the school, my mentor advised me to study anatomy as soon as possible. An anatomy lesson is not something you would say I will study and comprehend the concepts overnight or for a week. You should attend the classes regularly. Students should provide accessible learning resources and not seek shortcuts…” " (Student no. 1). Participants believed that successful students have good self-confidence and leadership in their learning process. Some students emphasized that the teachers can first build self-confidence and make them self-centered and self-regulated in learning. This concept requires the teachers to listen to them.“... If I realized the concepts satisfactorily, I would get a lot out of my effort. First, I accepted that anatomy is complicated, but I can help myself more than anyone. I started self-study. More action boosts self-confidence, which is required to become a doctor.“(Student no. 6) “... If I realized the concepts satisfactorily, I would get a lot out of my effort. First, I accepted that anatomy is complicated, but I can help myself more than anyone. I started self-study. More action boosts self-confidence, which is required to become a doctor.“(Student no. 6) Finally, the participants emphasized the importance of integration. Integration is simply defined as merging relevant content or subject areas. Teachers mention that the goal is to integrate and create a general perspective rather than a fragmented perspective of concepts. Some students believed that while multidisciplinary integration was provided, professors’ art of education, such as mastery of the teaching and learning process, generated the necessary harmony between the content presented. If well-coordinated, all aspects mutually reinforce each other, promoting learning in an integrated way.“... I enjoyed our integrated curriculum because that led to a better understanding of medicine. For example, discussing the anatomy of the gastrointestinal system gave me invaluable and integrated knowledge, and I enjoyed it ... “(Student no. 4) “... I enjoyed our integrated curriculum because that led to a better understanding of medicine. For example, discussing the anatomy of the gastrointestinal system gave me invaluable and integrated knowledge, and I enjoyed it ... “(Student no. 4) Anatomy professors acknowledged that integration with clinical courses had changed their perspective, reflected in their teaching.“…During the integration process and while I was listening to the viewpoints of the clinical colleagues, I realized what I thought to be the crucial thing that students should all be aware of scarcely happens in the real world!...“ (FM no. 2) “…During the integration process and while I was listening to the viewpoints of the clinical colleagues, I realized what I thought to be the crucial thing that students should all be aware of scarcely happens in the real world!...“ (FM no. 2) 2)Mindful Clinical View. Mindful Clinical View. Mindfulness is defined as awareness of what is happening at the present moment. Some professors believed that they must be aware of the ultimate goal while teaching at each moment. Therefore, mindfulness is influenced by one’s dreams.“…I try to teach anatomy in a way that our current students and prospective physicians can diagnose heart enlargement in the future…” (FM no. 7) “…I try to teach anatomy in a way that our current students and prospective physicians can diagnose heart enlargement in the future…” (FM no. 7) If anatomy teachers gain a conservative view, the students can achieve integrity and professional identity as doctors.“... The student’s learning and professional identity depend largely on knowing and comprehending anatomy. Teachers can play the role of a good teacher in any situation, whether a crisis (like the Covid-19 pandemic) or not. A good teacher always applies the right approach....“ (FM no. 1). “... The student’s learning and professional identity depend largely on knowing and comprehending anatomy. Teachers can play the role of a good teacher in any situation, whether a crisis (like the Covid-19 pandemic) or not. A good teacher always applies the right approach....“ (FM no. 1). [SUBTITLE] Subtheme 2: role modeling and student support (heating up the students’ hearts) [SUBSECTION] The professors’ experiences in educating successful students during anatomy courses show that they should reduce unnecessary complaints and criticisms to penetrate their hearts and light the torch of knowledge in their minds. Feelings for the learners should be expressed, and love for them should be shown. There should be a role model for them in academia. If the love of science enlightens students, they are strengthened to endure hardships and problems. One of the professors said in this regard:“…As an ancient Persian anecdote, “to attract the heart of a friend, we must do every trick.“ I sat down during soccer World Cup games and watched part of the game with the students…” (FM no. 8) “…As an ancient Persian anecdote, “to attract the heart of a friend, we must do every trick.“ I sat down during soccer World Cup games and watched part of the game with the students…” (FM no. 8) The professors’ experiences in educating successful students during anatomy courses show that they should reduce unnecessary complaints and criticisms to penetrate their hearts and light the torch of knowledge in their minds. Feelings for the learners should be expressed, and love for them should be shown. There should be a role model for them in academia. If the love of science enlightens students, they are strengthened to endure hardships and problems. One of the professors said in this regard:“…As an ancient Persian anecdote, “to attract the heart of a friend, we must do every trick.“ I sat down during soccer World Cup games and watched part of the game with the students…” (FM no. 8) “…As an ancient Persian anecdote, “to attract the heart of a friend, we must do every trick.“ I sat down during soccer World Cup games and watched part of the game with the students…” (FM no. 8) [SUBTITLE] Joyful teaching (telling stories/games/humor) [SUBSECTION] According to the participants, turning hard work into enjoyable teaching and learning is the art of medical teachers. A few faculty members believe that humor and mixing lessons with fun and games can make students aware of their superior position as a professor. They find icebreaking essential at the beginning of each discussion.“…If students enjoy the class and assume that learning is a plausible activity, they will not waste their time. Most of the wasted time and students’ naughtiness occur because of the attractiveness of the classroom and teacher….“ (FM no. 10) “…If students enjoy the class and assume that learning is a plausible activity, they will not waste their time. Most of the wasted time and students’ naughtiness occur because of the attractiveness of the classroom and teacher….“ (FM no. 10) Such an approach strengthens the bond between the students and teachers and leads to effective role modeling.“... If the student does not follow me in the classroom, what is the benefit of my teaching class?“ (FM no..8) “... If the student does not follow me in the classroom, what is the benefit of my teaching class?“ (FM no..8) Students can understand whether their teacher can be a trusted and knowledgeable role model and respond better if they encounter a fun and supportive teacher.“…good teachers are fully aware of all their behavior and movements and know what to teach in the classroom. They avoid traditional teaching methods and teach enjoyable for both students and teachers. Students will not be tired and enjoy the learning process….“ (Student no. 10) “…good teachers are fully aware of all their behavior and movements and know what to teach in the classroom. They avoid traditional teaching methods and teach enjoyable for both students and teachers. Students will not be tired and enjoy the learning process….“ (Student no. 10) According to the participants, turning hard work into enjoyable teaching and learning is the art of medical teachers. A few faculty members believe that humor and mixing lessons with fun and games can make students aware of their superior position as a professor. They find icebreaking essential at the beginning of each discussion.“…If students enjoy the class and assume that learning is a plausible activity, they will not waste their time. Most of the wasted time and students’ naughtiness occur because of the attractiveness of the classroom and teacher….“ (FM no. 10) “…If students enjoy the class and assume that learning is a plausible activity, they will not waste their time. Most of the wasted time and students’ naughtiness occur because of the attractiveness of the classroom and teacher….“ (FM no. 10) Such an approach strengthens the bond between the students and teachers and leads to effective role modeling.“... If the student does not follow me in the classroom, what is the benefit of my teaching class?“ (FM no..8) “... If the student does not follow me in the classroom, what is the benefit of my teaching class?“ (FM no..8) Students can understand whether their teacher can be a trusted and knowledgeable role model and respond better if they encounter a fun and supportive teacher.“…good teachers are fully aware of all their behavior and movements and know what to teach in the classroom. They avoid traditional teaching methods and teach enjoyable for both students and teachers. Students will not be tired and enjoy the learning process….“ (Student no. 10) “…good teachers are fully aware of all their behavior and movements and know what to teach in the classroom. They avoid traditional teaching methods and teach enjoyable for both students and teachers. Students will not be tired and enjoy the learning process….“ (Student no. 10) [SUBTITLE] Student Support System [SUBSECTION] Both faculty members and students mentioned the student support system as influential. All the participants believe that student support is helpful for vulnerable students, those with learning disabilities, and successful and elite students. The faculty members referred them to counseling, helped them solve their educational and emotional problems, and supported their families emotionally and financially.“…Student support is essential, but it is not just for high-risk and susceptible students. Sometimes elites need even more assistance, and I have seen and experienced how sometimes talented kids are vulnerable...“ (FM no.8). “…Student support is essential, but it is not just for high-risk and susceptible students. Sometimes elites need even more assistance, and I have seen and experienced how sometimes talented kids are vulnerable...“ (FM no.8). Such support, mainly when anatomy professors are referred to as the first role models for medical students, motivates students to improve over the past.“…I had the support of the family and could work harder and enjoy learning; they did not hesitate to do anything for me. The teachers and faculty members were sometimes right supporters and solved my problem….“ (Student no. 3) “…I had the support of the family and could work harder and enjoy learning; they did not hesitate to do anything for me. The teachers and faculty members were sometimes right supporters and solved my problem….“ (Student no. 3) Both faculty members and students mentioned the student support system as influential. All the participants believe that student support is helpful for vulnerable students, those with learning disabilities, and successful and elite students. The faculty members referred them to counseling, helped them solve their educational and emotional problems, and supported their families emotionally and financially.“…Student support is essential, but it is not just for high-risk and susceptible students. Sometimes elites need even more assistance, and I have seen and experienced how sometimes talented kids are vulnerable...“ (FM no.8). “…Student support is essential, but it is not just for high-risk and susceptible students. Sometimes elites need even more assistance, and I have seen and experienced how sometimes talented kids are vulnerable...“ (FM no.8). Such support, mainly when anatomy professors are referred to as the first role models for medical students, motivates students to improve over the past.“…I had the support of the family and could work harder and enjoy learning; they did not hesitate to do anything for me. The teachers and faculty members were sometimes right supporters and solved my problem….“ (Student no. 3) “…I had the support of the family and could work harder and enjoy learning; they did not hesitate to do anything for me. The teachers and faculty members were sometimes right supporters and solved my problem….“ (Student no. 3) [SUBTITLE] Subtheme 3: sledgehammer approach (thorough engagement in the age of pandemics) [SUBSECTION] While anatomy is perceived as one of the most challenging courses for preclinical students, the advent of the Covid-19 pandemic adds to the challenge. Successful anatomy teachers state that they must apply a combination of educational theories quickly, seamlessly, and effectively in the classroom or online. They believe that teaching anatomy in pandemic situations should be accompanied by joy and vivacity and increase the moments of discovery and intuition for learning. The faculty members seem to struggle to get the most involvement from students. Teaching approaches that trigger student involvement include a study guide, reference autonomy, neuroanatomical engagement, drawing, problem-based learning, and peer-based learning. While anatomy is perceived as one of the most challenging courses for preclinical students, the advent of the Covid-19 pandemic adds to the challenge. Successful anatomy teachers state that they must apply a combination of educational theories quickly, seamlessly, and effectively in the classroom or online. They believe that teaching anatomy in pandemic situations should be accompanied by joy and vivacity and increase the moments of discovery and intuition for learning. The faculty members seem to struggle to get the most involvement from students. Teaching approaches that trigger student involvement include a study guide, reference autonomy, neuroanatomical engagement, drawing, problem-based learning, and peer-based learning. [SUBTITLE] Study guide [SUBSECTION] Most participants believe preparedness is an essential strategy for success in learning and teaching. The best way is to provide a friendly study guide. The study guide is like a travel guide designed by an instructor to guide learners. It can increase the student’s insight into the subject. Some professors had the experience of contributing to making “road map” books for anatomy. They believe that such a study guide is also a studying resource. Others found the study guide essential for managing the students’ learning.“... Student guidance is an important issue that needs to be planned and implemented. We need more insight and hard work in pandemic conditions. Our “road map” book that primarily serves as a reference of anatomy is a study guide as well…” (FM no. 3) “... Student guidance is an important issue that needs to be planned and implemented. We need more insight and hard work in pandemic conditions. Our “road map” book that primarily serves as a reference of anatomy is a study guide as well…” (FM no. 3) Students assume that having a study guide is an insightful experience with more potential than it seems. They argue that a good stud guide must include common mistakes and guide them in all learning experiences, especially in cadaver dissection sessions.“... The study guide helps a lot in learning, and it is excellent if students’ common mistakes are made in this lesson, for example, in the lab and when we dissect the cadaver ...“ (FM no. 6) “... The study guide helps a lot in learning, and it is excellent if students’ common mistakes are made in this lesson, for example, in the lab and when we dissect the cadaver ...“ (FM no. 6) Most participants believe preparedness is an essential strategy for success in learning and teaching. The best way is to provide a friendly study guide. The study guide is like a travel guide designed by an instructor to guide learners. It can increase the student’s insight into the subject. Some professors had the experience of contributing to making “road map” books for anatomy. They believe that such a study guide is also a studying resource. Others found the study guide essential for managing the students’ learning.“... Student guidance is an important issue that needs to be planned and implemented. We need more insight and hard work in pandemic conditions. Our “road map” book that primarily serves as a reference of anatomy is a study guide as well…” (FM no. 3) “... Student guidance is an important issue that needs to be planned and implemented. We need more insight and hard work in pandemic conditions. Our “road map” book that primarily serves as a reference of anatomy is a study guide as well…” (FM no. 3) Students assume that having a study guide is an insightful experience with more potential than it seems. They argue that a good stud guide must include common mistakes and guide them in all learning experiences, especially in cadaver dissection sessions.“... The study guide helps a lot in learning, and it is excellent if students’ common mistakes are made in this lesson, for example, in the lab and when we dissect the cadaver ...“ (FM no. 6) “... The study guide helps a lot in learning, and it is excellent if students’ common mistakes are made in this lesson, for example, in the lab and when we dissect the cadaver ...“ (FM no. 6) [SUBTITLE] Neuroanatomical engagement [SUBSECTION] Participants believed that people learned new abstract concepts better when presented in visual and auditory ways. Professors believed that due to the functions of the brain hemisphere, the information process occurred through two separate systems in the brain. The right half is dedicated to the image, and the left half is word processing. Visual learning complements auditory learning. Sensory learning, which happens by working on the cadaver, complements student learning. Such a multidimensional approach leads to long-term memorization.“…I always use fascinating slides and video clips. If necessary, I explain my experience through the film, which enhances the understanding and facilitates long-term memorization…” (FM no. 5) “…I always use fascinating slides and video clips. If necessary, I explain my experience through the film, which enhances the understanding and facilitates long-term memorization…” (FM no. 5) Participants believed that people learned new abstract concepts better when presented in visual and auditory ways. Professors believed that due to the functions of the brain hemisphere, the information process occurred through two separate systems in the brain. The right half is dedicated to the image, and the left half is word processing. Visual learning complements auditory learning. Sensory learning, which happens by working on the cadaver, complements student learning. Such a multidimensional approach leads to long-term memorization.“…I always use fascinating slides and video clips. If necessary, I explain my experience through the film, which enhances the understanding and facilitates long-term memorization…” (FM no. 5) “…I always use fascinating slides and video clips. If necessary, I explain my experience through the film, which enhances the understanding and facilitates long-term memorization…” (FM no. 5) [SUBTITLE] Reference autonomy [SUBSECTION] According to the participants, appropriate resources play an essential role in learning. A good resource should encourage the students to become more active and reinforce their thinking, reasoning, and comprehension. Resources must be dynamic to adapt in times of crisis (pandemic) and change to meet the needs of 21st-century learners using current technologies. There seems to be a demand for respecting autonomy in both student and teacher experiences. They react better when students feel free and supported to choose their resources.“…For anatomy, I use Gray’s Anatomy book. However, recently, one of my professors introduced Moore’s Anatomy book. I felt this book was better in the field of …” (Student no. 2) “…For anatomy, I use Gray’s Anatomy book. However, recently, one of my professors introduced Moore’s Anatomy book. I felt this book was better in the field of …” (Student no. 2) Teachers argue that if provided with a dynamic curriculum design that encompasses clear goals, they can select from all the available resources and provide better ones. They believe that students should be taught to search and find valuable resources when they are highly interested in topics. When teachers accept the principle of self-learning and self-guidance, they listen to their students feel more confident, leading to more enthusiasm and thus creating future lifelong learners.“…I had a student who came up to me and asked me to explain how to learn anatomy. I was surprised because I was unfamiliar with the question. I told him to get two books, look at them, and return them to me the day after. He came while he got many valuable points, and now he is looking for further resources. Now, he is one of my most successful students, and one of my most important duties is introducing excellent resources to students...“ (FM no. 4) “…I had a student who came up to me and asked me to explain how to learn anatomy. I was surprised because I was unfamiliar with the question. I told him to get two books, look at them, and return them to me the day after. He came while he got many valuable points, and now he is looking for further resources. Now, he is one of my most successful students, and one of my most important duties is introducing excellent resources to students...“ (FM no. 4) According to the participants, appropriate resources play an essential role in learning. A good resource should encourage the students to become more active and reinforce their thinking, reasoning, and comprehension. Resources must be dynamic to adapt in times of crisis (pandemic) and change to meet the needs of 21st-century learners using current technologies. There seems to be a demand for respecting autonomy in both student and teacher experiences. They react better when students feel free and supported to choose their resources.“…For anatomy, I use Gray’s Anatomy book. However, recently, one of my professors introduced Moore’s Anatomy book. I felt this book was better in the field of …” (Student no. 2) “…For anatomy, I use Gray’s Anatomy book. However, recently, one of my professors introduced Moore’s Anatomy book. I felt this book was better in the field of …” (Student no. 2) Teachers argue that if provided with a dynamic curriculum design that encompasses clear goals, they can select from all the available resources and provide better ones. They believe that students should be taught to search and find valuable resources when they are highly interested in topics. When teachers accept the principle of self-learning and self-guidance, they listen to their students feel more confident, leading to more enthusiasm and thus creating future lifelong learners.“…I had a student who came up to me and asked me to explain how to learn anatomy. I was surprised because I was unfamiliar with the question. I told him to get two books, look at them, and return them to me the day after. He came while he got many valuable points, and now he is looking for further resources. Now, he is one of my most successful students, and one of my most important duties is introducing excellent resources to students...“ (FM no. 4) “…I had a student who came up to me and asked me to explain how to learn anatomy. I was surprised because I was unfamiliar with the question. I told him to get two books, look at them, and return them to me the day after. He came while he got many valuable points, and now he is looking for further resources. Now, he is one of my most successful students, and one of my most important duties is introducing excellent resources to students...“ (FM no. 4) [SUBTITLE] Mindful dissection, better transition [SUBSECTION] The first official and scientific encounter of young students with the human body occurs in dissection sessions. The first sessions familiarize the students with the disgusting aspects of gross medical procedures in which teachers play a facilitating role in such transition.“…I did not feel good on the first day, and many of my friends could not see the cadavers. It smelled so bad, but when we got used to it and started learning, we had better feelings, especially when we had a practical examination …” (Student no.14) “…I did not feel good on the first day, and many of my friends could not see the cadavers. It smelled so bad, but when we got used to it and started learning, we had better feelings, especially when we had a practical examination …” (Student no.14) Anatomy teachers are the game changers who can seed respect towards the human body in students’ minds.“… A cadaver is the most important teaching tool in the anatomy course and serves as a teacher’s blackboard. It is full of teaching points for medical students. I think this is ethically important as well. They should learn to respect the dead body highly to show respect to the living body in the future….“ (FM no.12) “… A cadaver is the most important teaching tool in the anatomy course and serves as a teacher’s blackboard. It is full of teaching points for medical students. I think this is ethically important as well. They should learn to respect the dead body highly to show respect to the living body in the future….“ (FM no.12) The first official and scientific encounter of young students with the human body occurs in dissection sessions. The first sessions familiarize the students with the disgusting aspects of gross medical procedures in which teachers play a facilitating role in such transition.“…I did not feel good on the first day, and many of my friends could not see the cadavers. It smelled so bad, but when we got used to it and started learning, we had better feelings, especially when we had a practical examination …” (Student no.14) “…I did not feel good on the first day, and many of my friends could not see the cadavers. It smelled so bad, but when we got used to it and started learning, we had better feelings, especially when we had a practical examination …” (Student no.14) Anatomy teachers are the game changers who can seed respect towards the human body in students’ minds.“… A cadaver is the most important teaching tool in the anatomy course and serves as a teacher’s blackboard. It is full of teaching points for medical students. I think this is ethically important as well. They should learn to respect the dead body highly to show respect to the living body in the future….“ (FM no.12) “… A cadaver is the most important teaching tool in the anatomy course and serves as a teacher’s blackboard. It is full of teaching points for medical students. I think this is ethically important as well. They should learn to respect the dead body highly to show respect to the living body in the future….“ (FM no.12) [SUBTITLE] Drawing [SUBSECTION] Teachers and students have valuable experiences in learning and memorizing through drawing. Drawing helps them to understand the topic, consolidate what they have learned, and remember it in long-term memory.“…Anatomy has a regional nature. It means that, for instance, everything in the chest area should be learned simultaneously and as a whole. I use drawing to show the adjacency of structures so that students can better learn and consolidate the topic...“ (FM no. 3) “…Anatomy has a regional nature. It means that, for instance, everything in the chest area should be learned simultaneously and as a whole. I use drawing to show the adjacency of structures so that students can better learn and consolidate the topic...“ (FM no. 3) For some learners, drawing becomes a ritual; one of the talented students said:“…I always tell other students to draw pictures to understand anatomy correctly. Draw a lot! Anatomy cannot be understood without painting. No matter how strong you are in abstract thinking, still, draw! I had a notebook in which I was painting...“ (Student no.5) “…I always tell other students to draw pictures to understand anatomy correctly. Draw a lot! Anatomy cannot be understood without painting. No matter how strong you are in abstract thinking, still, draw! I had a notebook in which I was painting...“ (Student no.5) Teachers and students have valuable experiences in learning and memorizing through drawing. Drawing helps them to understand the topic, consolidate what they have learned, and remember it in long-term memory.“…Anatomy has a regional nature. It means that, for instance, everything in the chest area should be learned simultaneously and as a whole. I use drawing to show the adjacency of structures so that students can better learn and consolidate the topic...“ (FM no. 3) “…Anatomy has a regional nature. It means that, for instance, everything in the chest area should be learned simultaneously and as a whole. I use drawing to show the adjacency of structures so that students can better learn and consolidate the topic...“ (FM no. 3) For some learners, drawing becomes a ritual; one of the talented students said:“…I always tell other students to draw pictures to understand anatomy correctly. Draw a lot! Anatomy cannot be understood without painting. No matter how strong you are in abstract thinking, still, draw! I had a notebook in which I was painting...“ (Student no.5) “…I always tell other students to draw pictures to understand anatomy correctly. Draw a lot! Anatomy cannot be understood without painting. No matter how strong you are in abstract thinking, still, draw! I had a notebook in which I was painting...“ (Student no.5) [SUBTITLE] Problem-based learning [SUBSECTION] Students will feel more satisfied if they are involved in future career problems. By using this approach, students are also emotionally involved in future challenges and are taught to enjoy solving future issues. Those problems that include detailed scene descriptions in a socio-emotional context and are related to cultural, moral, and social values seem to engage students better, help long-term memorization, and facilitate transition into future doctors. One of the professors said:“… I will show the students a picture of the patient and tell them that the person has subarachnoid hemorrhage. Look at the patient’s family. You can see their deep worry. Tell me where the bleeding source is and what is the first step?... " (FM no. 6) “… I will show the students a picture of the patient and tell them that the person has subarachnoid hemorrhage. Look at the patient’s family. You can see their deep worry. Tell me where the bleeding source is and what is the first step?... " (FM no. 6) Students will feel more satisfied if they are involved in future career problems. By using this approach, students are also emotionally involved in future challenges and are taught to enjoy solving future issues. Those problems that include detailed scene descriptions in a socio-emotional context and are related to cultural, moral, and social values seem to engage students better, help long-term memorization, and facilitate transition into future doctors. One of the professors said:“… I will show the students a picture of the patient and tell them that the person has subarachnoid hemorrhage. Look at the patient’s family. You can see their deep worry. Tell me where the bleeding source is and what is the first step?... " (FM no. 6) “… I will show the students a picture of the patient and tell them that the person has subarachnoid hemorrhage. Look at the patient’s family. You can see their deep worry. Tell me where the bleeding source is and what is the first step?... " (FM no. 6) [SUBTITLE] Peer-assisted learning [SUBSECTION] Students stated that learning in small groups was attractive because they acquired interpersonal skills like listening, speaking, discussing, and group leadership. Besides, it promotes higher cognitive abilities such as reasoning and problem-solving.“...For me, the best learning tool is a peer to peer group work. I have good experience in all subjects from literature to anatomy utilizing this method…” (Student no.5) “...For me, the best learning tool is a peer to peer group work. I have good experience in all subjects from literature to anatomy utilizing this method…” (Student no.5) Students stated that learning in small groups was attractive because they acquired interpersonal skills like listening, speaking, discussing, and group leadership. Besides, it promotes higher cognitive abilities such as reasoning and problem-solving.“...For me, the best learning tool is a peer to peer group work. I have good experience in all subjects from literature to anatomy utilizing this method…” (Student no.5) “...For me, the best learning tool is a peer to peer group work. I have good experience in all subjects from literature to anatomy utilizing this method…” (Student no.5)
Conclusion
Medical students experience a fundamental evolution into responsible and lifelong learners qualified as professional doctors. Educational systems focus primarily on teaching and learning, while students’ transition can be facilitated by a three-step model called the Blacksmith Approach, based on the study results. Initially, the blacksmith (educational system) must prepare a new forge. It means anatomy teachers should redesign the academic mentality of students by delivering messages about role change, difficulties ahead, and the importance of self-guidance, along with implementing clinical scenarios in active learning methods. Then, he/she must heat the metal. It implies that anatomy teachers, as gatekeepers of medicine, should adopt amusing teaching methods while embracing their role modeling and actively applying support strategies. Finally, he/she may shape the metal with a sledgehammer. Teachers need to use three components to create a sword (self-confident, accountable, resilient professional); thorough engagement by using active and engaging teaching techniques, reframing the situation with problem-based clinical scenarios, and joyful teaching. And selective information seeking via study guide and reference autonomy. In conclusion, an active and integrated strategy for teaching and learning anatomy is suggested to be applied in other courses and areas of medical education in the current medical curriculum.
[ "Objectives", "Design", "Setting", "Sampling", "Data collection", "Data analysis", "Participants’ characteristics", "Central theme: blacksmith approach", "Sub theme1: redesigning educational mindset (making a new forge)", "Subtheme 2: role modeling and student support (heating up the students’ hearts)", "Joyful teaching (telling stories/games/humor)", "Student Support System", "Subtheme 3: sledgehammer approach (thorough engagement in the age of pandemics)", "Study guide", "Neuroanatomical engagement", "Reference autonomy", "Mindful dissection, better transition", "Drawing", "Problem-based learning", "Peer-assisted learning", "First Step: A new forge (mindset)", "Second step", "Third step: Sledgehammer’s approach", "Study limitation", "" ]
[ "In this study, we aimed to identify the themes that explain the strategy for teaching and learning and foster meaningful learning and professional identity in medical anatomy courses among medical students in Iran. Also, we aimed to develop a conceptual framework to explain this strategy development based on the experience of both students and faculty members(FM) through a qualitative study.", "A qualitative approach was used by inductive content analysis in the first step [35]. Then we formulated the discovered themes into a comprehensive pedagogical approach.", "This study was conducted at Shiraz University of Medical Sciences (SUMS). The university currently includes more than 10,000 students, 200 majors, 782 faculty members, 54 research centers, 13 educational hospitals, a history of 70 years, and an assertive group teaching anatomy with fourteen faculty members in different academic ranks (assistant professors, associated professors, and full professors).\nMedical students start an anatomy course as they enter medical school with an integrated organ-based curriculum. They pass 15 academic and practical units of this course, including complete systems: blood circulation, respiration, endocrine, musculoskeletal, and urinary, in integration with other classes such as physiology, biochemistry, and clinical courses on the same topics. Students participate in formative and summative assessment tests such as quizzes and final examinations to improve learning and obtain end-of-semester grades. Students should reveal in-depth, thoughtful principles of anatomy through multiple-choice questions(MCQ) tests. Also, Laboratory work examination includes dissecting preserved specimens, microscopic study, and computer simulations in objective structured clinical examination (OSCE) situations to reveal their competence in the anatomy courses [28, 36].", "We used purposive and snowball sampling with maximum variation. Accordingly, we selected 24 medical students from year 1 to senior interns and 12 FMs within a wide range of academic rank and experience from 9 Iranian Universities of Medical Sciences in 2020–2021.\nThe inclusion criteria for medical students were studying in the first year of basic science to senior interns based on their willingness to participate. The inclusion criteria for faculty members in the anatomy or medical education field were a minimum of five-year teaching experience and a desire to participate. The exclusion criterion was the unwillingness to participate in the study. Initially, we collected data from a medical teacher well known for his high-quality teaching; then, we continued data gathering from medical students, anatomy faculty members, and medical education experts until data saturation was obtained and when no new code was obtained during interviews and repetition of the previous categories and codes [37–40]. Sampling and data coding continued till data saturation when no new code was obtained during interviews and repetition of the previous categories and codes.", "We utilized semi-structured interviews. At first, we contacted participants by telephone to explain the purpose of the study and research questions. The interviews were done in a quiet place, at an appropriate time, face-to-face, and individually. The interview questions focused on participants’ experiences of learning and teaching anatomy. The interviews started with questions as follows:\n\nWhat is your description of successful teaching/learning in an anatomy course?Tell me about your experiences in learning/teaching anatomy and different situations, including the dissection laboratory.\n\nWhat is your description of successful teaching/learning in an anatomy course?\nTell me about your experiences in learning/teaching anatomy and different situations, including the dissection laboratory.\nThen, according to the participants’ answers, we asked exploratory questions. We searched for signs of success in teaching and learning, the factors that lead to failure, and the characteristics that can change the behavior and vision of medical students. Each interview took 25 to 80 min, with an average of 45 min.\nMoreover, we gathered some field notes from the laboratory’s educational atmosphere. Data were collected and analyzed using Microsoft One Note2010, Microsoft, Redmond campus, US.", "We listened to each recorded interview to get an overall understanding. Then we analyzed verbatim and in-depth using inductive content analysis before the following interview, so each interview guided the next.\nThe meaning units consisted of words and sentences abstracted and labeled with codes. Twenty-six participants were interviewed in one session, and two or three sessions were conducted for six subjects.\nThe thematic content analysis includes six stages: [1] familiarizing with data, [2] generating the initial codes, [3] searching for themes, [4] reviewing the themes, (the various codes were compared based on similarities and differences in meaning and were categorized together) [5] defining and naming themes, and [6] preparing the report [41, 42].\nAfter conducting iterative and comparative line-by-line coding procedures, we sorted similar codes into subcategories. Finally, we rearranged similar subcategories and domains into major categories [39].\nIn qualitative research, data quality and accuracy are used instead of validity and reliability. Lincoln and Guba have proposed four methods for validating data that many qualitative researchers have used: credibility, dependability, confirmability, and transferability, which will be discussed later [43, 44].\nWe used various methods to validate the study, such as prolonged involvement (15 months from July 2020 to Nov 2021) with the participants, data, and subjects. Additionally, we performed member checks and expert checks for coding. For the member check, we sent the results of each analysis to the very participant, who would confirm them based on the interview and their own experiences.\nThe analysis results and categories were shared, approved by the supervisors, and approved by an experienced qualitative researcher.\nTransferability is defined by the generalizability of the results to fully describe the subject area and the characteristics of the participants with maximum variation in sampling. The generalization of the results was left to the reader to decide on the given information [45].", "In this study, semi-structured interviews were performed with 24 medical students from year 1 to senior interns, 15 (62.5% were males), 9 (37.5% were females), and 12 faculty members, 6 (50% were males), 6 (50% were females), with various academic ranks and experience from 9 (Shiraz, Tehran, Fasa, Jahrom, Bandarabas, Yasuj, Gerash, Larestan, and Bushehr) Iranian medical schools. The participants’ age ranged from 19 to 62 years old, with a mean of 33.4 ± 8.4 years. Based on the results, 12 students (50%) were in the clinical training period as students, externs, and interns, and 12 participants (50%) were in the basic sciences period. Accordingly, five participants (20%) had a history of being top students, and four (16.7%) had a history of dropout in anatomy courses. Furthermore, the work experience of the faculty members ranged from 5 to 35 years in teaching anatomy; 9(75%) anatomy faculty members and 3(25%) experts in medical education; faculty member participants, 3(25%) were single, and 9 (75%) were married. (Table 1)\n\nTable 1Demographic Characteristics of the Study ParticipantsParticipantsCharacteristics\nstudents\n\nGender\nNoPercent (%)\nMean Age (y ± SD)\nMale1562.522.3 ± 8.4Female937.5\nDegree\nFaculty membersassistant professors,32537.4 ± 11.7associated professors541.67full professors433.34\n\nDemographic Characteristics of the Study Participants\nFrom all the interviews, 334 codes were extracted. After the dismissal of similar codes and their integration, the experience of medical students was conceptualized into a central theme called the blacksmith approach, which included Three sub-themes: (1) making a new forge (Adequate preparation and mindful beginning), (2) heating the students’ hearts (considering supporting systems that learners need) and (3) using a Sledgehammer’s approach (teaching Anatomy thorough engaging all neuroanatomical regions in the Age of Pandemics). All the concepts were related to each other and resulted in a pattern revealing the experience of students and faculty members on strategies for successful learning and teaching of anatomy lessons. The main themes, as well as subordinate themes, are listed in Table 2.\n\nTable 2The Blacksmith Approach: Best strategy for teaching and learning in the medical anatomy course. Themes, sub-themes, and codesMain ThemesSub Themescodecods\nBlacksmith Approach\n\nRedesigning Educational Mindset (Making a New Forge)\n\nCreating a Vision for Students\n√ Anatomy is hard, bulky, should be taken seriously√ I can help myself more than anyone√ integrated curriculum√ self-study / Self-confidencescarcely happens in the real world!√ professional identity√ prospective physicians\nMindful Clinical View\n\nRole Modeling and Support (Heating up the Students’ Hearts)\n\nJoyful Teaching (Telling Stories/Games/ Humor)\n√ every trick must be done√ enjoy class√ follow me in the classroom\nStudent Support System\n√ susceptible students√ assistance√ Vulnerable√ right supportersSitting and talking to them\nSledgehammer Approach (Thorough Engagement in the Age of Pandemics)\n\nStudy Guide\n√ Preparedness√ Road map\nReference Autonomy\n√ self-learning√ self-direction\nNeuroanatomical Engagement\n√ Visual Learning√ Auditory Learning√ Sensory Learning√ Brain HemispheresLong-term Memorization\nMindful Dissection, Better Transition\n√ It smells so bad, but we need the observation√ We got used to it√ ethically important√ respect the dead body√ respect to the living body\nDrawing\n√ easy to learn√ Draw a lot\nProblem-Based Learning\n√ Look at the patient’s family√ Deep worry√ What is the next step of the problem?\nPeer-Assisted Learning\n√ Peer to the peer group√ Discussing the problem\n\nThe Blacksmith Approach: Best strategy for teaching and learning in the medical anatomy course. Themes, sub-themes, and codes\n√ Anatomy is hard, bulky, should be taken seriously\n√ I can help myself more than anyone\n√ integrated curriculum\n√ self-study / Self-confidence\nscarcely happens in the real world!\n√ professional identity\n√ prospective physicians\n√ every trick must be done\n√ enjoy class\n√ follow me in the classroom\n√ susceptible students\n√ assistance\n√ Vulnerable\n√ right supporters\nSitting and talking to them\n√ Preparedness\n√ Road map\n√ self-learning\n√ self-direction\n√ Visual Learning\n√ Auditory Learning\n√ Sensory Learning\n√ Brain Hemispheres\nLong-term Memorization\n√ It smells so bad, but we need the observation\n√ We got used to it\n√ ethically important\n√ respect the dead body\n√ respect to the living body\n√ easy to learn\n√ Draw a lot\n√ Look at the patient’s family\n√ Deep worry\n√ What is the next step of the problem?\n√ Peer to the peer group\n√ Discussing the problem", "According to the participants, in the student’s learning and maturity process, the teacher and the student act rationally and consciously and are aware of the factors that motivate and control them. Thus, they accept the risks, specific conditions, and hardships that must be tolerated. The student should handle this hardship to acquire a capacity to change, and the teacher should facilitate the change and their primary goal of teaching. In a sense, this is the same approach as the Blacksmiths. Blacksmithing symbolizes endurance, honor, and work under challenging conditions in our culture. (Fig. 1) We took the central theme from an interview with a professor who said:\n\nFig. 1The role of the blacksmith approach in the stages of teaching and learning anatomy in medical students is based on the study’s results. First, the blacksmith (educational system) must prepare a new blacksmith (redesign the educational mentality and create a new perspective on anatomical knowledge in students) and prepare a new forge; then, he/she must heat the metal (supporting young students and having good role-modeling) and then shape the metal with a sledgehammer (the sledgehammer approach to using different teaching strategies and methods) to create a sword or self-confident and knowledgeable learner in medicine (professionalism)\n”…The Sledgehammer should be heavy. Its nature is heavy; otherwise, it will not be capable of smoothing any iron. The nature of some courses, such as anatomy, is challenging, but we must make it enjoyable for the student….“ (FM no.6).\n\nThe role of the blacksmith approach in the stages of teaching and learning anatomy in medical students is based on the study’s results. First, the blacksmith (educational system) must prepare a new blacksmith (redesign the educational mentality and create a new perspective on anatomical knowledge in students) and prepare a new forge; then, he/she must heat the metal (supporting young students and having good role-modeling) and then shape the metal with a sledgehammer (the sledgehammer approach to using different teaching strategies and methods) to create a sword or self-confident and knowledgeable learner in medicine (professionalism)\n”…The Sledgehammer should be heavy. Its nature is heavy; otherwise, it will not be capable of smoothing any iron. The nature of some courses, such as anatomy, is challenging, but we must make it enjoyable for the student….“ (FM no.6).", "Creating an educational context is the first step for young students to begin their transition in the learning process. This context was found to have two aspects as follows:\n\nCreating a Vision for Students.\n\nCreating a Vision for Students.\nSome students mentioned a change in their mindset from the beginning with the idea that they have a difficult task ahead and should know what is helpful or not. In addition, realizing that the student is the most important person to help themselves seemed to provide a better sense of success.\nAccording to the participants, anatomy is one of the most voluminous and complex courses in medicine, which students should take seriously from the beginning.“Anatomy is hard, bulky, and should be taken seriously.“ (Student no. 9)\n“Anatomy is hard, bulky, and should be taken seriously.“ (Student no. 9)\nEven though many have oversimplified it, successful students have attempted to learn it through vision and self-awareness.“… After entering the school, my mentor advised me to study anatomy as soon as possible. An anatomy lesson is not something you would say I will study and comprehend the concepts overnight or for a week. You should attend the classes regularly. Students should provide accessible learning resources and not seek shortcuts…” \" (Student no. 1).\n“… After entering the school, my mentor advised me to study anatomy as soon as possible. An anatomy lesson is not something you would say I will study and comprehend the concepts overnight or for a week. You should attend the classes regularly. Students should provide accessible learning resources and not seek shortcuts…” \" (Student no. 1).\nParticipants believed that successful students have good self-confidence and leadership in their learning process. Some students emphasized that the teachers can first build self-confidence and make them self-centered and self-regulated in learning. This concept requires the teachers to listen to them.“... If I realized the concepts satisfactorily, I would get a lot out of my effort. First, I accepted that anatomy is complicated, but I can help myself more than anyone. I started self-study. More action boosts self-confidence, which is required to become a doctor.“(Student no. 6)\n“... If I realized the concepts satisfactorily, I would get a lot out of my effort. First, I accepted that anatomy is complicated, but I can help myself more than anyone. I started self-study. More action boosts self-confidence, which is required to become a doctor.“(Student no. 6)\nFinally, the participants emphasized the importance of integration. Integration is simply defined as merging relevant content or subject areas. Teachers mention that the goal is to integrate and create a general perspective rather than a fragmented perspective of concepts. Some students believed that while multidisciplinary integration was provided, professors’ art of education, such as mastery of the teaching and learning process, generated the necessary harmony between the content presented. If well-coordinated, all aspects mutually reinforce each other, promoting learning in an integrated way.“... I enjoyed our integrated curriculum because that led to a better understanding of medicine. For example, discussing the anatomy of the gastrointestinal system gave me invaluable and integrated knowledge, and I enjoyed it ... “(Student no. 4)\n“... I enjoyed our integrated curriculum because that led to a better understanding of medicine. For example, discussing the anatomy of the gastrointestinal system gave me invaluable and integrated knowledge, and I enjoyed it ... “(Student no. 4)\nAnatomy professors acknowledged that integration with clinical courses had changed their perspective, reflected in their teaching.“…During the integration process and while I was listening to the viewpoints of the clinical colleagues, I realized what I thought to be the crucial thing that students should all be aware of scarcely happens in the real world!...“ (FM no. 2)\n“…During the integration process and while I was listening to the viewpoints of the clinical colleagues, I realized what I thought to be the crucial thing that students should all be aware of scarcely happens in the real world!...“ (FM no. 2)\n\n2)Mindful Clinical View.\n\nMindful Clinical View.\nMindfulness is defined as awareness of what is happening at the present moment. Some professors believed that they must be aware of the ultimate goal while teaching at each moment. Therefore, mindfulness is influenced by one’s dreams.“…I try to teach anatomy in a way that our current students and prospective physicians can diagnose heart enlargement in the future…” (FM no. 7)\n“…I try to teach anatomy in a way that our current students and prospective physicians can diagnose heart enlargement in the future…” (FM no. 7)\nIf anatomy teachers gain a conservative view, the students can achieve integrity and professional identity as doctors.“... The student’s learning and professional identity depend largely on knowing and comprehending anatomy. Teachers can play the role of a good teacher in any situation, whether a crisis (like the Covid-19 pandemic) or not. A good teacher always applies the right approach....“ (FM no. 1).\n“... The student’s learning and professional identity depend largely on knowing and comprehending anatomy. Teachers can play the role of a good teacher in any situation, whether a crisis (like the Covid-19 pandemic) or not. A good teacher always applies the right approach....“ (FM no. 1).", "The professors’ experiences in educating successful students during anatomy courses show that they should reduce unnecessary complaints and criticisms to penetrate their hearts and light the torch of knowledge in their minds. Feelings for the learners should be expressed, and love for them should be shown. There should be a role model for them in academia. If the love of science enlightens students, they are strengthened to endure hardships and problems. One of the professors said in this regard:“…As an ancient Persian anecdote, “to attract the heart of a friend, we must do every trick.“ I sat down during soccer World Cup games and watched part of the game with the students…” (FM no. 8)\n“…As an ancient Persian anecdote, “to attract the heart of a friend, we must do every trick.“ I sat down during soccer World Cup games and watched part of the game with the students…” (FM no. 8)", "According to the participants, turning hard work into enjoyable teaching and learning is the art of medical teachers. A few faculty members believe that humor and mixing lessons with fun and games can make students aware of their superior position as a professor. They find icebreaking essential at the beginning of each discussion.“…If students enjoy the class and assume that learning is a plausible activity, they will not waste their time. Most of the wasted time and students’ naughtiness occur because of the attractiveness of the classroom and teacher….“ (FM no. 10)\n“…If students enjoy the class and assume that learning is a plausible activity, they will not waste their time. Most of the wasted time and students’ naughtiness occur because of the attractiveness of the classroom and teacher….“ (FM no. 10)\nSuch an approach strengthens the bond between the students and teachers and leads to effective role modeling.“... If the student does not follow me in the classroom, what is the benefit of my teaching class?“ (FM no..8)\n“... If the student does not follow me in the classroom, what is the benefit of my teaching class?“ (FM no..8)\nStudents can understand whether their teacher can be a trusted and knowledgeable role model and respond better if they encounter a fun and supportive teacher.“…good teachers are fully aware of all their behavior and movements and know what to teach in the classroom. They avoid traditional teaching methods and teach enjoyable for both students and teachers. Students will not be tired and enjoy the learning process….“ (Student no. 10)\n“…good teachers are fully aware of all their behavior and movements and know what to teach in the classroom. They avoid traditional teaching methods and teach enjoyable for both students and teachers. Students will not be tired and enjoy the learning process….“ (Student no. 10)", "Both faculty members and students mentioned the student support system as influential. All the participants believe that student support is helpful for vulnerable students, those with learning disabilities, and successful and elite students. The faculty members referred them to counseling, helped them solve their educational and emotional problems, and supported their families emotionally and financially.“…Student support is essential, but it is not just for high-risk and susceptible students. Sometimes elites need even more assistance, and I have seen and experienced how sometimes talented kids are vulnerable...“ (FM no.8).\n“…Student support is essential, but it is not just for high-risk and susceptible students. Sometimes elites need even more assistance, and I have seen and experienced how sometimes talented kids are vulnerable...“ (FM no.8).\nSuch support, mainly when anatomy professors are referred to as the first role models for medical students, motivates students to improve over the past.“…I had the support of the family and could work harder and enjoy learning; they did not hesitate to do anything for me. The teachers and faculty members were sometimes right supporters and solved my problem….“ (Student no. 3)\n“…I had the support of the family and could work harder and enjoy learning; they did not hesitate to do anything for me. The teachers and faculty members were sometimes right supporters and solved my problem….“ (Student no. 3)", "While anatomy is perceived as one of the most challenging courses for preclinical students, the advent of the Covid-19 pandemic adds to the challenge. Successful anatomy teachers state that they must apply a combination of educational theories quickly, seamlessly, and effectively in the classroom or online. They believe that teaching anatomy in pandemic situations should be accompanied by joy and vivacity and increase the moments of discovery and intuition for learning. The faculty members seem to struggle to get the most involvement from students.\nTeaching approaches that trigger student involvement include a study guide, reference autonomy, neuroanatomical engagement, drawing, problem-based learning, and peer-based learning.", "Most participants believe preparedness is an essential strategy for success in learning and teaching. The best way is to provide a friendly study guide. The study guide is like a travel guide designed by an instructor to guide learners. It can increase the student’s insight into the subject. Some professors had the experience of contributing to making “road map” books for anatomy. They believe that such a study guide is also a studying resource. Others found the study guide essential for managing the students’ learning.“... Student guidance is an important issue that needs to be planned and implemented. We need more insight and hard work in pandemic conditions. Our “road map” book that primarily serves as a reference of anatomy is a study guide as well…” (FM no. 3)\n“... Student guidance is an important issue that needs to be planned and implemented. We need more insight and hard work in pandemic conditions. Our “road map” book that primarily serves as a reference of anatomy is a study guide as well…” (FM no. 3)\nStudents assume that having a study guide is an insightful experience with more potential than it seems. They argue that a good stud guide must include common mistakes and guide them in all learning experiences, especially in cadaver dissection sessions.“... The study guide helps a lot in learning, and it is excellent if students’ common mistakes are made in this lesson, for example, in the lab and when we dissect the cadaver ...“ (FM no. 6)\n“... The study guide helps a lot in learning, and it is excellent if students’ common mistakes are made in this lesson, for example, in the lab and when we dissect the cadaver ...“ (FM no. 6)", "Participants believed that people learned new abstract concepts better when presented in visual and auditory ways. Professors believed that due to the functions of the brain hemisphere, the information process occurred through two separate systems in the brain. The right half is dedicated to the image, and the left half is word processing. Visual learning complements auditory learning. Sensory learning, which happens by working on the cadaver, complements student learning. Such a multidimensional approach leads to long-term memorization.“…I always use fascinating slides and video clips. If necessary, I explain my experience through the film, which enhances the understanding and facilitates long-term memorization…” (FM no. 5)\n“…I always use fascinating slides and video clips. If necessary, I explain my experience through the film, which enhances the understanding and facilitates long-term memorization…” (FM no. 5)", "According to the participants, appropriate resources play an essential role in learning. A good resource should encourage the students to become more active and reinforce their thinking, reasoning, and comprehension. Resources must be dynamic to adapt in times of crisis (pandemic) and change to meet the needs of 21st-century learners using current technologies. There seems to be a demand for respecting autonomy in both student and teacher experiences. They react better when students feel free and supported to choose their resources.“…For anatomy, I use Gray’s Anatomy book. However, recently, one of my professors introduced Moore’s Anatomy book. I felt this book was better in the field of …” (Student no. 2)\n“…For anatomy, I use Gray’s Anatomy book. However, recently, one of my professors introduced Moore’s Anatomy book. I felt this book was better in the field of …” (Student no. 2)\nTeachers argue that if provided with a dynamic curriculum design that encompasses clear goals, they can select from all the available resources and provide better ones. They believe that students should be taught to search and find valuable resources when they are highly interested in topics.\nWhen teachers accept the principle of self-learning and self-guidance, they listen to their students feel more confident, leading to more enthusiasm and thus creating future lifelong learners.“…I had a student who came up to me and asked me to explain how to learn anatomy. I was surprised because I was unfamiliar with the question. I told him to get two books, look at them, and return them to me the day after. He came while he got many valuable points, and now he is looking for further resources. Now, he is one of my most successful students, and one of my most important duties is introducing excellent resources to students...“ (FM no. 4)\n“…I had a student who came up to me and asked me to explain how to learn anatomy. I was surprised because I was unfamiliar with the question. I told him to get two books, look at them, and return them to me the day after. He came while he got many valuable points, and now he is looking for further resources. Now, he is one of my most successful students, and one of my most important duties is introducing excellent resources to students...“ (FM no. 4)", "The first official and scientific encounter of young students with the human body occurs in dissection sessions. The first sessions familiarize the students with the disgusting aspects of gross medical procedures in which teachers play a facilitating role in such transition.“…I did not feel good on the first day, and many of my friends could not see the cadavers. It smelled so bad, but when we got used to it and started learning, we had better feelings, especially when we had a practical examination …” (Student no.14)\n“…I did not feel good on the first day, and many of my friends could not see the cadavers. It smelled so bad, but when we got used to it and started learning, we had better feelings, especially when we had a practical examination …” (Student no.14)\nAnatomy teachers are the game changers who can seed respect towards the human body in students’ minds.“… A cadaver is the most important teaching tool in the anatomy course and serves as a teacher’s blackboard. It is full of teaching points for medical students. I think this is ethically important as well. They should learn to respect the dead body highly to show respect to the living body in the future….“ (FM no.12)\n“… A cadaver is the most important teaching tool in the anatomy course and serves as a teacher’s blackboard. It is full of teaching points for medical students. I think this is ethically important as well. They should learn to respect the dead body highly to show respect to the living body in the future….“ (FM no.12)", "Teachers and students have valuable experiences in learning and memorizing through drawing. Drawing helps them to understand the topic, consolidate what they have learned, and remember it in long-term memory.“…Anatomy has a regional nature. It means that, for instance, everything in the chest area should be learned simultaneously and as a whole. I use drawing to show the adjacency of structures so that students can better learn and consolidate the topic...“ (FM no. 3)\n“…Anatomy has a regional nature. It means that, for instance, everything in the chest area should be learned simultaneously and as a whole. I use drawing to show the adjacency of structures so that students can better learn and consolidate the topic...“ (FM no. 3)\nFor some learners, drawing becomes a ritual; one of the talented students said:“…I always tell other students to draw pictures to understand anatomy correctly. Draw a lot! Anatomy cannot be understood without painting. No matter how strong you are in abstract thinking, still, draw! I had a notebook in which I was painting...“ (Student no.5)\n“…I always tell other students to draw pictures to understand anatomy correctly. Draw a lot! Anatomy cannot be understood without painting. No matter how strong you are in abstract thinking, still, draw! I had a notebook in which I was painting...“ (Student no.5)", "Students will feel more satisfied if they are involved in future career problems. By using this approach, students are also emotionally involved in future challenges and are taught to enjoy solving future issues. Those problems that include detailed scene descriptions in a socio-emotional context and are related to cultural, moral, and social values seem to engage students better, help long-term memorization, and facilitate transition into future doctors.\nOne of the professors said:“… I will show the students a picture of the patient and tell them that the person has subarachnoid hemorrhage. Look at the patient’s family. You can see their deep worry. Tell me where the bleeding source is and what is the first step?... \" (FM no. 6)\n“… I will show the students a picture of the patient and tell them that the person has subarachnoid hemorrhage. Look at the patient’s family. You can see their deep worry. Tell me where the bleeding source is and what is the first step?... \" (FM no. 6)", "Students stated that learning in small groups was attractive because they acquired interpersonal skills like listening, speaking, discussing, and group leadership. Besides, it promotes higher cognitive abilities such as reasoning and problem-solving.“...For me, the best learning tool is a peer to peer group work. I have good experience in all subjects from literature to anatomy utilizing this method…” (Student no.5)\n“...For me, the best learning tool is a peer to peer group work. I have good experience in all subjects from literature to anatomy utilizing this method…” (Student no.5)", "must be built for the students. This step is in line with the first stage of the four S model, i.e., Situation awareness. Here, the students become aware of what is happening. The teachers here have two roles. First, they should deliver the following messages to the students: (1) you are supposed to actualize a new identity (role change), (2) anatomy (as a pillar of medicine) provides a problematic task ahead (stress), and (3) you can help yourself more than anyone else. (control) Furthermore, the students should try self-study to increase their self-confidence (trigger). Secondly, the teachers should acquire a mindful clinical standpoint, i.e., developing good clinical knowledge and adopting active learning methods based on teamwork and clinical scenarios that encourage active participation and self-directed learning as if they are future doctors. This strategy has been shown to reduce student stress [11, 13] and help them react better as future physicians [3, 8, 48].", "The students’ hearts must be warmed up through support and role modeling. This step is compatible with the following two stages in the 4 S model, i.e., self and support. The students evaluate their demographic and psychological characteristics, and then they need to be aware of support systems and be able to get help [27, 29]. Here again, the role of anatomy teachers is critical. As medical students see anatomy as a gate to medicine, the anatomy teacher can be known as the gatekeeper. They will inevitably actualize their teachers as wise role models who are righteous supporters. The anatomy faculty member should be aware of their role modeling. Role modeling can help students foster resilience for future change, which leads to professionalism and integrity [7, 18, 49–52]. The teachers should adopt ice-breaking methods, use their sense of humor, tell stories, design games, and have friendly chats with students outside the classroom. In addition, anatomy teachers should gain the skills and knowledge for counseling and get familiar with support strategies to become prominent supporters of students [53–56].", "We found that, currently, preclinical students are facing two overwhelming stressors. On the one hand, the covid crisis has detached the students from their teachers and resources [15, 18, 57]. On the other hand, students need to handle their transition into knowledgeable medical professionals. Therefore we need to incorporate teaching methods that assist our detached students in coping with their stressors. The last stage in the 4 S model also deals with coping strategies. Students may use several coping strategies like information seeking, selective ignoring, and reframing the challenge [58].\nAccordingly, the third step in our model includes three components; thorough engagement, reframing the situation, and selective information seeking. Regarding the first component, we encourage meticulously choosing more active and engaging learning techniques. Problem-based learning and Peer-based discussions in small groups, drawing, mind mapping, body painting, using play-doh, and interactive video content, are advised. According to Pandey and Zimitat (2007), a practical approach to anatomy education positively correlates with teaching and reflective learning quality. Successful anatomy learning requires balancing memorization with understanding and visualization[59]. Insightful experiences in teaching anatomy with this strategy are found in Australia (head and neck course) [15] and India (musculoskeletal anatomy) [13]. As Goodman et al. (2006) noted in their previous book, those with higher self-esteem and mental mastery are better able to cope with challenges. [60] A successful learning experience gives students a sense of control over their course, while applying a study guide, reference autonomy, and support system can help them gain self-esteem [61–63].\nReframing the situation, As the second component, is where the students need to redefine their roles and activities. Two sets of strategies can help students to reframe. 1) Using problem-based and peer-assisted learning with a flavor of sociocultural contexts can prepare students for better learning in a positive, non-intimidating learning environment to redefine themselves as future doctors for diagnosing and curing patients. Socio-emotional context and bringing drama elements into case scenarios have been recently emphasized to induce resilience and professionalism in medical students (2.14, 64).\nRecent evidence indicates that gamification leads to positive learning outcomes, higher motivation, and engagement. However, several downsides should also be considered, including increased competition and task evaluation difficulties. Besides, it depends highly on the context in which the gamification is being implemented and its users [65–68]. Hamari et al. (2014) have reviewed empirical literature, showing that gamification has proven effective in many fields, most notably education. Studies in education and learning contexts illustrate how the gamification of learning outcomes has been positive by increasing motivation and engagement in learning and enjoyment tasks over time. However, the studies also highlighted some negative points that require attention, such as the effects of increased competition, task evaluation difficulties, and design features. The review indicates that gamification provides positive effects; however, the effects greatly depend on the context in which the gamification is being implemented and the users who use it [66]. Recent studies suggest that joy and humor facilitate a person’s transition into more socially severe roles (like becoming a doctor) [34]. Hence, teachers should use joyful techniques such as painting, ice braking, storytelling, gamification, dance, and music [4, 9, 15, 53].\nThe last component deals with selective information seeking. Students should have a study guide and be free to choose their desirable resources (anatomy reference books, digital resources, or web-based social media videos). We advise the medical school to design a dynamic curriculum encompassing clear goals. In this setting, the study guide can act as a tool for transferring core knowledge in anatomical sciences. As a result, the tedious aspect of bulky content in anatomy courses can be avoided, and the educational efficiency of synchronous and asynchronous learning can be increased in the pandemic era [55, 56, 61, 69, 70].\nThis research is in line with constructive learning theory that emphasizes student-centered learning, which states that students should be actively involved in education and contribute to knowledge creation [71]. In a special effort to create a modern educational curriculum at Stanford University, Prober et al. recommended building a framework of core knowledge, embedding it into rich interactive formats, and encouraging students to pursue knowledge in some deep, but not all, domains [63]. The Blacksmith approach facilitates personal and in-depth pursuits that help transfer students into lifelong learners and adapt to challenging situations [63, 64]. This statement is consistent with Wald and Monteverde (2021). They report that educational resilience is vital in supporting and sustaining healthcare professional identity development and facilitating the progress of students’ moral resilience and complexity, especially in times of pandemic [25].\nAccording to the participants’ viewpoints, the applicability of the blacksmith approach is one of the most important aspects of improving the quality of anatomy teaching and learning in any situation, such as current critical conditions.\nOur university’s online programs were adequately integrated into the curriculum design and used to guide students on a learning journey rather than merely as another resource; perhaps, these tools could be beneficial. However, in such circumstances, the epidemic and sanctions of Iran and the lack of resources, students and professors should take measures to optimize education to facilitate the teaching and learning of this course. Accordingly, from the above point, through a qualitative study, it was assumed that there were signs of successful teaching and learning and internal change in deep search among students and faculty members.\nRevolution in medical education is a global program, and many medical schools have perceived investigation and experienced challenges [63, 64]. Correspondingly, an active and integrated strategy for teaching and learning anatomy has been emphasized in other medical education courses and areas, including a contemporary medical curriculum. Managers, practitioners, and professors can use the elements of this strategy to increase the efficiency of medical education, improve students’ knowledge, skills, and attitudes, and ultimately maintain and promote understanding of anatomy and other courses. Also, from the participant’s perspective, the applicability of students learning experiences and faculty members’ teaching experiences in the context and workplace conditions was one of the factors that changed the view of anatomy as an introductory course in medicine.", "As this study was conducted in one country, it is suggested that more studies and insights be obtained from other participants to increase the generalizability of findings. Besides, further work should be done to provide more practical guidance in the educational system regarding the transition of medical students in teaching and learning anatomy in the current pandemic crisis.", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Introduction", "Objectives", "Materials and methods", "Design", "Setting", "Sampling", "Data collection", "Data analysis", "Results", "Participants’ characteristics", "Central theme: blacksmith approach", "Sub theme1: redesigning educational mindset (making a new forge)", "Subtheme 2: role modeling and student support (heating up the students’ hearts)", "Joyful teaching (telling stories/games/humor)", "Student Support System", "Subtheme 3: sledgehammer approach (thorough engagement in the age of pandemics)", "Study guide", "Neuroanatomical engagement", "Reference autonomy", "Mindful dissection, better transition", "Drawing", "Problem-based learning", "Peer-assisted learning", "Discussion", "First Step: A new forge (mindset)", "Second step", "Third step: Sledgehammer’s approach", "Study limitation", "Conclusion", "Electronic supplementary material", "" ]
[ "Anatomy, as a branch of biological and medical sciences, is one of the introductory courses in the learning experience of undergraduate medical students, which is essential for impending clinical practice and clinical proficiency [1, 2]. A challenging question facing medical education is how to change some young students into responsible and resilient professional physicians. Most of the answers are being placed in clinical years; however, we can begin the transition process earlier with a promising tool called anatomy [3, 4].\nDespite being one of the fundamental pillars of medical knowledge, anatomy has been pulled back as a simple course in the preclinical period. Historically, ancient medical schools have relied heavily on philosophical and intuitive systems of thought [5]. Experimental investigations by dissection helped physicians actualize the human body as a combination of multiple organ systems. [6]. We can assume that anatomy science has led to reform in how medicine views the human being from a spiritual, holistic being into a biopsychosocial person. However, anatomy has been underestimated in current medical pedagogy [7, 8].\nRecent evidence indicates that we can reform anatomy courses making them as influential as they can be. [9–11]. Anatomy and dissection sessions can act as students’ first encounters with the humanized face of medicine, where medical students can learn teamwork, morality, coping strategies, and communication skills. In addition, anatomy can be perceived as a way of internalizing self-awareness, medical epistemology, empathy, and medical ethics [6, 12, 13].\nWhile a myriad of teaching approaches are available for anatomy education, they have changed according to the needs of learners or global crises like covid-19. The primary approaches include lecture, dissection, and demonstration of cadavers. In addition, many schools incorporated radiological imaging as a learning tool for teaching anatomy in the living body. Considering the shortcomings of traditional methods, medical schools started implementing more innovative techniques that enhanced active learning. While Problem-based learning, flipped classroom, and reflective writing activities on the cadaveric dissection fostered communication skills and self-directed learning [12, 14, 15], other methods focused on students’ learning styles: drawing and whiteboarding, three-dimensional printing (3DP), and digital models for visual learners; play-doh, role modeling and peer physical exam for tactile learning; singing, dancing, yoga, and pilates for kinaesthetic learners [16–18]. The covid pandemic disrupted anatomy education as the number of donated cadavers decreased substantially, and social distancing prevented in-person classes or labs [17–19]. Accordingly, the teaching approaches relied, more than ever, on technology leading to the widespread use of computer-assisted learning, web-based learning via social media (esp. Facebook and youtube), and a more prominent role for Augmented-Reality and Virtual-Reality [20, 21]. The main focus behind all these methods and strategies is better teaching anatomy for its sake. However, such an approach does not allow us to use the full potential of anatomy science as a core element in changing young students into self-confident and resilient learners, which leads to the creation of future professional physicians [22, 23].\nPhysicians need professional competencies, a set of which essential competencies, including professional resilience, is and emotional competencies, to achieve goals. As a capacity to endure difficulties and quickly improve from a stress-inducing experience, resilience helps one to make valuable adaptations to problems and can utilize as a protective defense against adversities, thus facilitating welfare [23, 24]. Resilience is referred to as one’s capability to maintain and improve their well-being when faced with life challenges. Resilience can be considered a behavior acquired during training and various courses, including the main course of anatomy. Resilience includes cognitive processes and has four dimensions, including self-efficacy. Planning, Self-control, Commitment, and perseverance include the following: young doctors must be present in various educational environments and support clinical and educational supervisors in enduring difficulties to help them develop personal and professional resilience [24, 25].\nIn Iran and at Shiraz University, anatomy courses entail three steps. Initially, a topic is introduced through face-to-face, online or offline lectures with a slide or a short video clip. Next, students work on anatomical modeling and, finally, on a cadaver [26, 27].\nToday, not only the medical students must bear a significant load of details in anatomy courses, which can reduce their motivation, but they should also face the educational consequences of the covid pandemic, which include loss of hands-on learning activities and limited access to tools like cadavers, models, pathology specimens, and skeletons [28–34]. Losing face-to-face contact and direct interaction with peers and teachers may hinder the students’ growth as future physicians [6, 13]. Therefore, finding ways to improve the learning experience is more challenging than ever.", "In this study, we aimed to identify the themes that explain the strategy for teaching and learning and foster meaningful learning and professional identity in medical anatomy courses among medical students in Iran. Also, we aimed to develop a conceptual framework to explain this strategy development based on the experience of both students and faculty members(FM) through a qualitative study.", "[SUBTITLE] Design [SUBSECTION] A qualitative approach was used by inductive content analysis in the first step [35]. Then we formulated the discovered themes into a comprehensive pedagogical approach.\nA qualitative approach was used by inductive content analysis in the first step [35]. Then we formulated the discovered themes into a comprehensive pedagogical approach.\n[SUBTITLE] Setting [SUBSECTION] This study was conducted at Shiraz University of Medical Sciences (SUMS). The university currently includes more than 10,000 students, 200 majors, 782 faculty members, 54 research centers, 13 educational hospitals, a history of 70 years, and an assertive group teaching anatomy with fourteen faculty members in different academic ranks (assistant professors, associated professors, and full professors).\nMedical students start an anatomy course as they enter medical school with an integrated organ-based curriculum. They pass 15 academic and practical units of this course, including complete systems: blood circulation, respiration, endocrine, musculoskeletal, and urinary, in integration with other classes such as physiology, biochemistry, and clinical courses on the same topics. Students participate in formative and summative assessment tests such as quizzes and final examinations to improve learning and obtain end-of-semester grades. Students should reveal in-depth, thoughtful principles of anatomy through multiple-choice questions(MCQ) tests. Also, Laboratory work examination includes dissecting preserved specimens, microscopic study, and computer simulations in objective structured clinical examination (OSCE) situations to reveal their competence in the anatomy courses [28, 36].\nThis study was conducted at Shiraz University of Medical Sciences (SUMS). The university currently includes more than 10,000 students, 200 majors, 782 faculty members, 54 research centers, 13 educational hospitals, a history of 70 years, and an assertive group teaching anatomy with fourteen faculty members in different academic ranks (assistant professors, associated professors, and full professors).\nMedical students start an anatomy course as they enter medical school with an integrated organ-based curriculum. They pass 15 academic and practical units of this course, including complete systems: blood circulation, respiration, endocrine, musculoskeletal, and urinary, in integration with other classes such as physiology, biochemistry, and clinical courses on the same topics. Students participate in formative and summative assessment tests such as quizzes and final examinations to improve learning and obtain end-of-semester grades. Students should reveal in-depth, thoughtful principles of anatomy through multiple-choice questions(MCQ) tests. Also, Laboratory work examination includes dissecting preserved specimens, microscopic study, and computer simulations in objective structured clinical examination (OSCE) situations to reveal their competence in the anatomy courses [28, 36].\n[SUBTITLE] Sampling [SUBSECTION] We used purposive and snowball sampling with maximum variation. Accordingly, we selected 24 medical students from year 1 to senior interns and 12 FMs within a wide range of academic rank and experience from 9 Iranian Universities of Medical Sciences in 2020–2021.\nThe inclusion criteria for medical students were studying in the first year of basic science to senior interns based on their willingness to participate. The inclusion criteria for faculty members in the anatomy or medical education field were a minimum of five-year teaching experience and a desire to participate. The exclusion criterion was the unwillingness to participate in the study. Initially, we collected data from a medical teacher well known for his high-quality teaching; then, we continued data gathering from medical students, anatomy faculty members, and medical education experts until data saturation was obtained and when no new code was obtained during interviews and repetition of the previous categories and codes [37–40]. Sampling and data coding continued till data saturation when no new code was obtained during interviews and repetition of the previous categories and codes.\nWe used purposive and snowball sampling with maximum variation. Accordingly, we selected 24 medical students from year 1 to senior interns and 12 FMs within a wide range of academic rank and experience from 9 Iranian Universities of Medical Sciences in 2020–2021.\nThe inclusion criteria for medical students were studying in the first year of basic science to senior interns based on their willingness to participate. The inclusion criteria for faculty members in the anatomy or medical education field were a minimum of five-year teaching experience and a desire to participate. The exclusion criterion was the unwillingness to participate in the study. Initially, we collected data from a medical teacher well known for his high-quality teaching; then, we continued data gathering from medical students, anatomy faculty members, and medical education experts until data saturation was obtained and when no new code was obtained during interviews and repetition of the previous categories and codes [37–40]. Sampling and data coding continued till data saturation when no new code was obtained during interviews and repetition of the previous categories and codes.\n[SUBTITLE] Data collection [SUBSECTION] We utilized semi-structured interviews. At first, we contacted participants by telephone to explain the purpose of the study and research questions. The interviews were done in a quiet place, at an appropriate time, face-to-face, and individually. The interview questions focused on participants’ experiences of learning and teaching anatomy. The interviews started with questions as follows:\n\nWhat is your description of successful teaching/learning in an anatomy course?Tell me about your experiences in learning/teaching anatomy and different situations, including the dissection laboratory.\n\nWhat is your description of successful teaching/learning in an anatomy course?\nTell me about your experiences in learning/teaching anatomy and different situations, including the dissection laboratory.\nThen, according to the participants’ answers, we asked exploratory questions. We searched for signs of success in teaching and learning, the factors that lead to failure, and the characteristics that can change the behavior and vision of medical students. Each interview took 25 to 80 min, with an average of 45 min.\nMoreover, we gathered some field notes from the laboratory’s educational atmosphere. Data were collected and analyzed using Microsoft One Note2010, Microsoft, Redmond campus, US.\nWe utilized semi-structured interviews. At first, we contacted participants by telephone to explain the purpose of the study and research questions. The interviews were done in a quiet place, at an appropriate time, face-to-face, and individually. The interview questions focused on participants’ experiences of learning and teaching anatomy. The interviews started with questions as follows:\n\nWhat is your description of successful teaching/learning in an anatomy course?Tell me about your experiences in learning/teaching anatomy and different situations, including the dissection laboratory.\n\nWhat is your description of successful teaching/learning in an anatomy course?\nTell me about your experiences in learning/teaching anatomy and different situations, including the dissection laboratory.\nThen, according to the participants’ answers, we asked exploratory questions. We searched for signs of success in teaching and learning, the factors that lead to failure, and the characteristics that can change the behavior and vision of medical students. Each interview took 25 to 80 min, with an average of 45 min.\nMoreover, we gathered some field notes from the laboratory’s educational atmosphere. Data were collected and analyzed using Microsoft One Note2010, Microsoft, Redmond campus, US.\n[SUBTITLE] Data analysis [SUBSECTION] We listened to each recorded interview to get an overall understanding. Then we analyzed verbatim and in-depth using inductive content analysis before the following interview, so each interview guided the next.\nThe meaning units consisted of words and sentences abstracted and labeled with codes. Twenty-six participants were interviewed in one session, and two or three sessions were conducted for six subjects.\nThe thematic content analysis includes six stages: [1] familiarizing with data, [2] generating the initial codes, [3] searching for themes, [4] reviewing the themes, (the various codes were compared based on similarities and differences in meaning and were categorized together) [5] defining and naming themes, and [6] preparing the report [41, 42].\nAfter conducting iterative and comparative line-by-line coding procedures, we sorted similar codes into subcategories. Finally, we rearranged similar subcategories and domains into major categories [39].\nIn qualitative research, data quality and accuracy are used instead of validity and reliability. Lincoln and Guba have proposed four methods for validating data that many qualitative researchers have used: credibility, dependability, confirmability, and transferability, which will be discussed later [43, 44].\nWe used various methods to validate the study, such as prolonged involvement (15 months from July 2020 to Nov 2021) with the participants, data, and subjects. Additionally, we performed member checks and expert checks for coding. For the member check, we sent the results of each analysis to the very participant, who would confirm them based on the interview and their own experiences.\nThe analysis results and categories were shared, approved by the supervisors, and approved by an experienced qualitative researcher.\nTransferability is defined by the generalizability of the results to fully describe the subject area and the characteristics of the participants with maximum variation in sampling. The generalization of the results was left to the reader to decide on the given information [45].\nWe listened to each recorded interview to get an overall understanding. Then we analyzed verbatim and in-depth using inductive content analysis before the following interview, so each interview guided the next.\nThe meaning units consisted of words and sentences abstracted and labeled with codes. Twenty-six participants were interviewed in one session, and two or three sessions were conducted for six subjects.\nThe thematic content analysis includes six stages: [1] familiarizing with data, [2] generating the initial codes, [3] searching for themes, [4] reviewing the themes, (the various codes were compared based on similarities and differences in meaning and were categorized together) [5] defining and naming themes, and [6] preparing the report [41, 42].\nAfter conducting iterative and comparative line-by-line coding procedures, we sorted similar codes into subcategories. Finally, we rearranged similar subcategories and domains into major categories [39].\nIn qualitative research, data quality and accuracy are used instead of validity and reliability. Lincoln and Guba have proposed four methods for validating data that many qualitative researchers have used: credibility, dependability, confirmability, and transferability, which will be discussed later [43, 44].\nWe used various methods to validate the study, such as prolonged involvement (15 months from July 2020 to Nov 2021) with the participants, data, and subjects. Additionally, we performed member checks and expert checks for coding. For the member check, we sent the results of each analysis to the very participant, who would confirm them based on the interview and their own experiences.\nThe analysis results and categories were shared, approved by the supervisors, and approved by an experienced qualitative researcher.\nTransferability is defined by the generalizability of the results to fully describe the subject area and the characteristics of the participants with maximum variation in sampling. The generalization of the results was left to the reader to decide on the given information [45].", "A qualitative approach was used by inductive content analysis in the first step [35]. Then we formulated the discovered themes into a comprehensive pedagogical approach.", "This study was conducted at Shiraz University of Medical Sciences (SUMS). The university currently includes more than 10,000 students, 200 majors, 782 faculty members, 54 research centers, 13 educational hospitals, a history of 70 years, and an assertive group teaching anatomy with fourteen faculty members in different academic ranks (assistant professors, associated professors, and full professors).\nMedical students start an anatomy course as they enter medical school with an integrated organ-based curriculum. They pass 15 academic and practical units of this course, including complete systems: blood circulation, respiration, endocrine, musculoskeletal, and urinary, in integration with other classes such as physiology, biochemistry, and clinical courses on the same topics. Students participate in formative and summative assessment tests such as quizzes and final examinations to improve learning and obtain end-of-semester grades. Students should reveal in-depth, thoughtful principles of anatomy through multiple-choice questions(MCQ) tests. Also, Laboratory work examination includes dissecting preserved specimens, microscopic study, and computer simulations in objective structured clinical examination (OSCE) situations to reveal their competence in the anatomy courses [28, 36].", "We used purposive and snowball sampling with maximum variation. Accordingly, we selected 24 medical students from year 1 to senior interns and 12 FMs within a wide range of academic rank and experience from 9 Iranian Universities of Medical Sciences in 2020–2021.\nThe inclusion criteria for medical students were studying in the first year of basic science to senior interns based on their willingness to participate. The inclusion criteria for faculty members in the anatomy or medical education field were a minimum of five-year teaching experience and a desire to participate. The exclusion criterion was the unwillingness to participate in the study. Initially, we collected data from a medical teacher well known for his high-quality teaching; then, we continued data gathering from medical students, anatomy faculty members, and medical education experts until data saturation was obtained and when no new code was obtained during interviews and repetition of the previous categories and codes [37–40]. Sampling and data coding continued till data saturation when no new code was obtained during interviews and repetition of the previous categories and codes.", "We utilized semi-structured interviews. At first, we contacted participants by telephone to explain the purpose of the study and research questions. The interviews were done in a quiet place, at an appropriate time, face-to-face, and individually. The interview questions focused on participants’ experiences of learning and teaching anatomy. The interviews started with questions as follows:\n\nWhat is your description of successful teaching/learning in an anatomy course?Tell me about your experiences in learning/teaching anatomy and different situations, including the dissection laboratory.\n\nWhat is your description of successful teaching/learning in an anatomy course?\nTell me about your experiences in learning/teaching anatomy and different situations, including the dissection laboratory.\nThen, according to the participants’ answers, we asked exploratory questions. We searched for signs of success in teaching and learning, the factors that lead to failure, and the characteristics that can change the behavior and vision of medical students. Each interview took 25 to 80 min, with an average of 45 min.\nMoreover, we gathered some field notes from the laboratory’s educational atmosphere. Data were collected and analyzed using Microsoft One Note2010, Microsoft, Redmond campus, US.", "We listened to each recorded interview to get an overall understanding. Then we analyzed verbatim and in-depth using inductive content analysis before the following interview, so each interview guided the next.\nThe meaning units consisted of words and sentences abstracted and labeled with codes. Twenty-six participants were interviewed in one session, and two or three sessions were conducted for six subjects.\nThe thematic content analysis includes six stages: [1] familiarizing with data, [2] generating the initial codes, [3] searching for themes, [4] reviewing the themes, (the various codes were compared based on similarities and differences in meaning and were categorized together) [5] defining and naming themes, and [6] preparing the report [41, 42].\nAfter conducting iterative and comparative line-by-line coding procedures, we sorted similar codes into subcategories. Finally, we rearranged similar subcategories and domains into major categories [39].\nIn qualitative research, data quality and accuracy are used instead of validity and reliability. Lincoln and Guba have proposed four methods for validating data that many qualitative researchers have used: credibility, dependability, confirmability, and transferability, which will be discussed later [43, 44].\nWe used various methods to validate the study, such as prolonged involvement (15 months from July 2020 to Nov 2021) with the participants, data, and subjects. Additionally, we performed member checks and expert checks for coding. For the member check, we sent the results of each analysis to the very participant, who would confirm them based on the interview and their own experiences.\nThe analysis results and categories were shared, approved by the supervisors, and approved by an experienced qualitative researcher.\nTransferability is defined by the generalizability of the results to fully describe the subject area and the characteristics of the participants with maximum variation in sampling. The generalization of the results was left to the reader to decide on the given information [45].", "[SUBTITLE] Participants’ characteristics [SUBSECTION] In this study, semi-structured interviews were performed with 24 medical students from year 1 to senior interns, 15 (62.5% were males), 9 (37.5% were females), and 12 faculty members, 6 (50% were males), 6 (50% were females), with various academic ranks and experience from 9 (Shiraz, Tehran, Fasa, Jahrom, Bandarabas, Yasuj, Gerash, Larestan, and Bushehr) Iranian medical schools. The participants’ age ranged from 19 to 62 years old, with a mean of 33.4 ± 8.4 years. Based on the results, 12 students (50%) were in the clinical training period as students, externs, and interns, and 12 participants (50%) were in the basic sciences period. Accordingly, five participants (20%) had a history of being top students, and four (16.7%) had a history of dropout in anatomy courses. Furthermore, the work experience of the faculty members ranged from 5 to 35 years in teaching anatomy; 9(75%) anatomy faculty members and 3(25%) experts in medical education; faculty member participants, 3(25%) were single, and 9 (75%) were married. (Table 1)\n\nTable 1Demographic Characteristics of the Study ParticipantsParticipantsCharacteristics\nstudents\n\nGender\nNoPercent (%)\nMean Age (y ± SD)\nMale1562.522.3 ± 8.4Female937.5\nDegree\nFaculty membersassistant professors,32537.4 ± 11.7associated professors541.67full professors433.34\n\nDemographic Characteristics of the Study Participants\nFrom all the interviews, 334 codes were extracted. After the dismissal of similar codes and their integration, the experience of medical students was conceptualized into a central theme called the blacksmith approach, which included Three sub-themes: (1) making a new forge (Adequate preparation and mindful beginning), (2) heating the students’ hearts (considering supporting systems that learners need) and (3) using a Sledgehammer’s approach (teaching Anatomy thorough engaging all neuroanatomical regions in the Age of Pandemics). All the concepts were related to each other and resulted in a pattern revealing the experience of students and faculty members on strategies for successful learning and teaching of anatomy lessons. The main themes, as well as subordinate themes, are listed in Table 2.\n\nTable 2The Blacksmith Approach: Best strategy for teaching and learning in the medical anatomy course. Themes, sub-themes, and codesMain ThemesSub Themescodecods\nBlacksmith Approach\n\nRedesigning Educational Mindset (Making a New Forge)\n\nCreating a Vision for Students\n√ Anatomy is hard, bulky, should be taken seriously√ I can help myself more than anyone√ integrated curriculum√ self-study / Self-confidencescarcely happens in the real world!√ professional identity√ prospective physicians\nMindful Clinical View\n\nRole Modeling and Support (Heating up the Students’ Hearts)\n\nJoyful Teaching (Telling Stories/Games/ Humor)\n√ every trick must be done√ enjoy class√ follow me in the classroom\nStudent Support System\n√ susceptible students√ assistance√ Vulnerable√ right supportersSitting and talking to them\nSledgehammer Approach (Thorough Engagement in the Age of Pandemics)\n\nStudy Guide\n√ Preparedness√ Road map\nReference Autonomy\n√ self-learning√ self-direction\nNeuroanatomical Engagement\n√ Visual Learning√ Auditory Learning√ Sensory Learning√ Brain HemispheresLong-term Memorization\nMindful Dissection, Better Transition\n√ It smells so bad, but we need the observation√ We got used to it√ ethically important√ respect the dead body√ respect to the living body\nDrawing\n√ easy to learn√ Draw a lot\nProblem-Based Learning\n√ Look at the patient’s family√ Deep worry√ What is the next step of the problem?\nPeer-Assisted Learning\n√ Peer to the peer group√ Discussing the problem\n\nThe Blacksmith Approach: Best strategy for teaching and learning in the medical anatomy course. Themes, sub-themes, and codes\n√ Anatomy is hard, bulky, should be taken seriously\n√ I can help myself more than anyone\n√ integrated curriculum\n√ self-study / Self-confidence\nscarcely happens in the real world!\n√ professional identity\n√ prospective physicians\n√ every trick must be done\n√ enjoy class\n√ follow me in the classroom\n√ susceptible students\n√ assistance\n√ Vulnerable\n√ right supporters\nSitting and talking to them\n√ Preparedness\n√ Road map\n√ self-learning\n√ self-direction\n√ Visual Learning\n√ Auditory Learning\n√ Sensory Learning\n√ Brain Hemispheres\nLong-term Memorization\n√ It smells so bad, but we need the observation\n√ We got used to it\n√ ethically important\n√ respect the dead body\n√ respect to the living body\n√ easy to learn\n√ Draw a lot\n√ Look at the patient’s family\n√ Deep worry\n√ What is the next step of the problem?\n√ Peer to the peer group\n√ Discussing the problem\nIn this study, semi-structured interviews were performed with 24 medical students from year 1 to senior interns, 15 (62.5% were males), 9 (37.5% were females), and 12 faculty members, 6 (50% were males), 6 (50% were females), with various academic ranks and experience from 9 (Shiraz, Tehran, Fasa, Jahrom, Bandarabas, Yasuj, Gerash, Larestan, and Bushehr) Iranian medical schools. The participants’ age ranged from 19 to 62 years old, with a mean of 33.4 ± 8.4 years. Based on the results, 12 students (50%) were in the clinical training period as students, externs, and interns, and 12 participants (50%) were in the basic sciences period. Accordingly, five participants (20%) had a history of being top students, and four (16.7%) had a history of dropout in anatomy courses. Furthermore, the work experience of the faculty members ranged from 5 to 35 years in teaching anatomy; 9(75%) anatomy faculty members and 3(25%) experts in medical education; faculty member participants, 3(25%) were single, and 9 (75%) were married. (Table 1)\n\nTable 1Demographic Characteristics of the Study ParticipantsParticipantsCharacteristics\nstudents\n\nGender\nNoPercent (%)\nMean Age (y ± SD)\nMale1562.522.3 ± 8.4Female937.5\nDegree\nFaculty membersassistant professors,32537.4 ± 11.7associated professors541.67full professors433.34\n\nDemographic Characteristics of the Study Participants\nFrom all the interviews, 334 codes were extracted. After the dismissal of similar codes and their integration, the experience of medical students was conceptualized into a central theme called the blacksmith approach, which included Three sub-themes: (1) making a new forge (Adequate preparation and mindful beginning), (2) heating the students’ hearts (considering supporting systems that learners need) and (3) using a Sledgehammer’s approach (teaching Anatomy thorough engaging all neuroanatomical regions in the Age of Pandemics). All the concepts were related to each other and resulted in a pattern revealing the experience of students and faculty members on strategies for successful learning and teaching of anatomy lessons. The main themes, as well as subordinate themes, are listed in Table 2.\n\nTable 2The Blacksmith Approach: Best strategy for teaching and learning in the medical anatomy course. Themes, sub-themes, and codesMain ThemesSub Themescodecods\nBlacksmith Approach\n\nRedesigning Educational Mindset (Making a New Forge)\n\nCreating a Vision for Students\n√ Anatomy is hard, bulky, should be taken seriously√ I can help myself more than anyone√ integrated curriculum√ self-study / Self-confidencescarcely happens in the real world!√ professional identity√ prospective physicians\nMindful Clinical View\n\nRole Modeling and Support (Heating up the Students’ Hearts)\n\nJoyful Teaching (Telling Stories/Games/ Humor)\n√ every trick must be done√ enjoy class√ follow me in the classroom\nStudent Support System\n√ susceptible students√ assistance√ Vulnerable√ right supportersSitting and talking to them\nSledgehammer Approach (Thorough Engagement in the Age of Pandemics)\n\nStudy Guide\n√ Preparedness√ Road map\nReference Autonomy\n√ self-learning√ self-direction\nNeuroanatomical Engagement\n√ Visual Learning√ Auditory Learning√ Sensory Learning√ Brain HemispheresLong-term Memorization\nMindful Dissection, Better Transition\n√ It smells so bad, but we need the observation√ We got used to it√ ethically important√ respect the dead body√ respect to the living body\nDrawing\n√ easy to learn√ Draw a lot\nProblem-Based Learning\n√ Look at the patient’s family√ Deep worry√ What is the next step of the problem?\nPeer-Assisted Learning\n√ Peer to the peer group√ Discussing the problem\n\nThe Blacksmith Approach: Best strategy for teaching and learning in the medical anatomy course. Themes, sub-themes, and codes\n√ Anatomy is hard, bulky, should be taken seriously\n√ I can help myself more than anyone\n√ integrated curriculum\n√ self-study / Self-confidence\nscarcely happens in the real world!\n√ professional identity\n√ prospective physicians\n√ every trick must be done\n√ enjoy class\n√ follow me in the classroom\n√ susceptible students\n√ assistance\n√ Vulnerable\n√ right supporters\nSitting and talking to them\n√ Preparedness\n√ Road map\n√ self-learning\n√ self-direction\n√ Visual Learning\n√ Auditory Learning\n√ Sensory Learning\n√ Brain Hemispheres\nLong-term Memorization\n√ It smells so bad, but we need the observation\n√ We got used to it\n√ ethically important\n√ respect the dead body\n√ respect to the living body\n√ easy to learn\n√ Draw a lot\n√ Look at the patient’s family\n√ Deep worry\n√ What is the next step of the problem?\n√ Peer to the peer group\n√ Discussing the problem\n[SUBTITLE] Central theme: blacksmith approach [SUBSECTION] According to the participants, in the student’s learning and maturity process, the teacher and the student act rationally and consciously and are aware of the factors that motivate and control them. Thus, they accept the risks, specific conditions, and hardships that must be tolerated. The student should handle this hardship to acquire a capacity to change, and the teacher should facilitate the change and their primary goal of teaching. In a sense, this is the same approach as the Blacksmiths. Blacksmithing symbolizes endurance, honor, and work under challenging conditions in our culture. (Fig. 1) We took the central theme from an interview with a professor who said:\n\nFig. 1The role of the blacksmith approach in the stages of teaching and learning anatomy in medical students is based on the study’s results. First, the blacksmith (educational system) must prepare a new blacksmith (redesign the educational mentality and create a new perspective on anatomical knowledge in students) and prepare a new forge; then, he/she must heat the metal (supporting young students and having good role-modeling) and then shape the metal with a sledgehammer (the sledgehammer approach to using different teaching strategies and methods) to create a sword or self-confident and knowledgeable learner in medicine (professionalism)\n”…The Sledgehammer should be heavy. Its nature is heavy; otherwise, it will not be capable of smoothing any iron. The nature of some courses, such as anatomy, is challenging, but we must make it enjoyable for the student….“ (FM no.6).\n\nThe role of the blacksmith approach in the stages of teaching and learning anatomy in medical students is based on the study’s results. First, the blacksmith (educational system) must prepare a new blacksmith (redesign the educational mentality and create a new perspective on anatomical knowledge in students) and prepare a new forge; then, he/she must heat the metal (supporting young students and having good role-modeling) and then shape the metal with a sledgehammer (the sledgehammer approach to using different teaching strategies and methods) to create a sword or self-confident and knowledgeable learner in medicine (professionalism)\n”…The Sledgehammer should be heavy. Its nature is heavy; otherwise, it will not be capable of smoothing any iron. The nature of some courses, such as anatomy, is challenging, but we must make it enjoyable for the student….“ (FM no.6).\nAccording to the participants, in the student’s learning and maturity process, the teacher and the student act rationally and consciously and are aware of the factors that motivate and control them. Thus, they accept the risks, specific conditions, and hardships that must be tolerated. The student should handle this hardship to acquire a capacity to change, and the teacher should facilitate the change and their primary goal of teaching. In a sense, this is the same approach as the Blacksmiths. Blacksmithing symbolizes endurance, honor, and work under challenging conditions in our culture. (Fig. 1) We took the central theme from an interview with a professor who said:\n\nFig. 1The role of the blacksmith approach in the stages of teaching and learning anatomy in medical students is based on the study’s results. First, the blacksmith (educational system) must prepare a new blacksmith (redesign the educational mentality and create a new perspective on anatomical knowledge in students) and prepare a new forge; then, he/she must heat the metal (supporting young students and having good role-modeling) and then shape the metal with a sledgehammer (the sledgehammer approach to using different teaching strategies and methods) to create a sword or self-confident and knowledgeable learner in medicine (professionalism)\n”…The Sledgehammer should be heavy. Its nature is heavy; otherwise, it will not be capable of smoothing any iron. The nature of some courses, such as anatomy, is challenging, but we must make it enjoyable for the student….“ (FM no.6).\n\nThe role of the blacksmith approach in the stages of teaching and learning anatomy in medical students is based on the study’s results. First, the blacksmith (educational system) must prepare a new blacksmith (redesign the educational mentality and create a new perspective on anatomical knowledge in students) and prepare a new forge; then, he/she must heat the metal (supporting young students and having good role-modeling) and then shape the metal with a sledgehammer (the sledgehammer approach to using different teaching strategies and methods) to create a sword or self-confident and knowledgeable learner in medicine (professionalism)\n”…The Sledgehammer should be heavy. Its nature is heavy; otherwise, it will not be capable of smoothing any iron. The nature of some courses, such as anatomy, is challenging, but we must make it enjoyable for the student….“ (FM no.6).\n[SUBTITLE] Sub theme1: redesigning educational mindset (making a new forge) [SUBSECTION] Creating an educational context is the first step for young students to begin their transition in the learning process. This context was found to have two aspects as follows:\n\nCreating a Vision for Students.\n\nCreating a Vision for Students.\nSome students mentioned a change in their mindset from the beginning with the idea that they have a difficult task ahead and should know what is helpful or not. In addition, realizing that the student is the most important person to help themselves seemed to provide a better sense of success.\nAccording to the participants, anatomy is one of the most voluminous and complex courses in medicine, which students should take seriously from the beginning.“Anatomy is hard, bulky, and should be taken seriously.“ (Student no. 9)\n“Anatomy is hard, bulky, and should be taken seriously.“ (Student no. 9)\nEven though many have oversimplified it, successful students have attempted to learn it through vision and self-awareness.“… After entering the school, my mentor advised me to study anatomy as soon as possible. An anatomy lesson is not something you would say I will study and comprehend the concepts overnight or for a week. You should attend the classes regularly. Students should provide accessible learning resources and not seek shortcuts…” \" (Student no. 1).\n“… After entering the school, my mentor advised me to study anatomy as soon as possible. An anatomy lesson is not something you would say I will study and comprehend the concepts overnight or for a week. You should attend the classes regularly. Students should provide accessible learning resources and not seek shortcuts…” \" (Student no. 1).\nParticipants believed that successful students have good self-confidence and leadership in their learning process. Some students emphasized that the teachers can first build self-confidence and make them self-centered and self-regulated in learning. This concept requires the teachers to listen to them.“... If I realized the concepts satisfactorily, I would get a lot out of my effort. First, I accepted that anatomy is complicated, but I can help myself more than anyone. I started self-study. More action boosts self-confidence, which is required to become a doctor.“(Student no. 6)\n“... If I realized the concepts satisfactorily, I would get a lot out of my effort. First, I accepted that anatomy is complicated, but I can help myself more than anyone. I started self-study. More action boosts self-confidence, which is required to become a doctor.“(Student no. 6)\nFinally, the participants emphasized the importance of integration. Integration is simply defined as merging relevant content or subject areas. Teachers mention that the goal is to integrate and create a general perspective rather than a fragmented perspective of concepts. Some students believed that while multidisciplinary integration was provided, professors’ art of education, such as mastery of the teaching and learning process, generated the necessary harmony between the content presented. If well-coordinated, all aspects mutually reinforce each other, promoting learning in an integrated way.“... I enjoyed our integrated curriculum because that led to a better understanding of medicine. For example, discussing the anatomy of the gastrointestinal system gave me invaluable and integrated knowledge, and I enjoyed it ... “(Student no. 4)\n“... I enjoyed our integrated curriculum because that led to a better understanding of medicine. For example, discussing the anatomy of the gastrointestinal system gave me invaluable and integrated knowledge, and I enjoyed it ... “(Student no. 4)\nAnatomy professors acknowledged that integration with clinical courses had changed their perspective, reflected in their teaching.“…During the integration process and while I was listening to the viewpoints of the clinical colleagues, I realized what I thought to be the crucial thing that students should all be aware of scarcely happens in the real world!...“ (FM no. 2)\n“…During the integration process and while I was listening to the viewpoints of the clinical colleagues, I realized what I thought to be the crucial thing that students should all be aware of scarcely happens in the real world!...“ (FM no. 2)\n\n2)Mindful Clinical View.\n\nMindful Clinical View.\nMindfulness is defined as awareness of what is happening at the present moment. Some professors believed that they must be aware of the ultimate goal while teaching at each moment. Therefore, mindfulness is influenced by one’s dreams.“…I try to teach anatomy in a way that our current students and prospective physicians can diagnose heart enlargement in the future…” (FM no. 7)\n“…I try to teach anatomy in a way that our current students and prospective physicians can diagnose heart enlargement in the future…” (FM no. 7)\nIf anatomy teachers gain a conservative view, the students can achieve integrity and professional identity as doctors.“... The student’s learning and professional identity depend largely on knowing and comprehending anatomy. Teachers can play the role of a good teacher in any situation, whether a crisis (like the Covid-19 pandemic) or not. A good teacher always applies the right approach....“ (FM no. 1).\n“... The student’s learning and professional identity depend largely on knowing and comprehending anatomy. Teachers can play the role of a good teacher in any situation, whether a crisis (like the Covid-19 pandemic) or not. A good teacher always applies the right approach....“ (FM no. 1).\nCreating an educational context is the first step for young students to begin their transition in the learning process. This context was found to have two aspects as follows:\n\nCreating a Vision for Students.\n\nCreating a Vision for Students.\nSome students mentioned a change in their mindset from the beginning with the idea that they have a difficult task ahead and should know what is helpful or not. In addition, realizing that the student is the most important person to help themselves seemed to provide a better sense of success.\nAccording to the participants, anatomy is one of the most voluminous and complex courses in medicine, which students should take seriously from the beginning.“Anatomy is hard, bulky, and should be taken seriously.“ (Student no. 9)\n“Anatomy is hard, bulky, and should be taken seriously.“ (Student no. 9)\nEven though many have oversimplified it, successful students have attempted to learn it through vision and self-awareness.“… After entering the school, my mentor advised me to study anatomy as soon as possible. An anatomy lesson is not something you would say I will study and comprehend the concepts overnight or for a week. You should attend the classes regularly. Students should provide accessible learning resources and not seek shortcuts…” \" (Student no. 1).\n“… After entering the school, my mentor advised me to study anatomy as soon as possible. An anatomy lesson is not something you would say I will study and comprehend the concepts overnight or for a week. You should attend the classes regularly. Students should provide accessible learning resources and not seek shortcuts…” \" (Student no. 1).\nParticipants believed that successful students have good self-confidence and leadership in their learning process. Some students emphasized that the teachers can first build self-confidence and make them self-centered and self-regulated in learning. This concept requires the teachers to listen to them.“... If I realized the concepts satisfactorily, I would get a lot out of my effort. First, I accepted that anatomy is complicated, but I can help myself more than anyone. I started self-study. More action boosts self-confidence, which is required to become a doctor.“(Student no. 6)\n“... If I realized the concepts satisfactorily, I would get a lot out of my effort. First, I accepted that anatomy is complicated, but I can help myself more than anyone. I started self-study. More action boosts self-confidence, which is required to become a doctor.“(Student no. 6)\nFinally, the participants emphasized the importance of integration. Integration is simply defined as merging relevant content or subject areas. Teachers mention that the goal is to integrate and create a general perspective rather than a fragmented perspective of concepts. Some students believed that while multidisciplinary integration was provided, professors’ art of education, such as mastery of the teaching and learning process, generated the necessary harmony between the content presented. If well-coordinated, all aspects mutually reinforce each other, promoting learning in an integrated way.“... I enjoyed our integrated curriculum because that led to a better understanding of medicine. For example, discussing the anatomy of the gastrointestinal system gave me invaluable and integrated knowledge, and I enjoyed it ... “(Student no. 4)\n“... I enjoyed our integrated curriculum because that led to a better understanding of medicine. For example, discussing the anatomy of the gastrointestinal system gave me invaluable and integrated knowledge, and I enjoyed it ... “(Student no. 4)\nAnatomy professors acknowledged that integration with clinical courses had changed their perspective, reflected in their teaching.“…During the integration process and while I was listening to the viewpoints of the clinical colleagues, I realized what I thought to be the crucial thing that students should all be aware of scarcely happens in the real world!...“ (FM no. 2)\n“…During the integration process and while I was listening to the viewpoints of the clinical colleagues, I realized what I thought to be the crucial thing that students should all be aware of scarcely happens in the real world!...“ (FM no. 2)\n\n2)Mindful Clinical View.\n\nMindful Clinical View.\nMindfulness is defined as awareness of what is happening at the present moment. Some professors believed that they must be aware of the ultimate goal while teaching at each moment. Therefore, mindfulness is influenced by one’s dreams.“…I try to teach anatomy in a way that our current students and prospective physicians can diagnose heart enlargement in the future…” (FM no. 7)\n“…I try to teach anatomy in a way that our current students and prospective physicians can diagnose heart enlargement in the future…” (FM no. 7)\nIf anatomy teachers gain a conservative view, the students can achieve integrity and professional identity as doctors.“... The student’s learning and professional identity depend largely on knowing and comprehending anatomy. Teachers can play the role of a good teacher in any situation, whether a crisis (like the Covid-19 pandemic) or not. A good teacher always applies the right approach....“ (FM no. 1).\n“... The student’s learning and professional identity depend largely on knowing and comprehending anatomy. Teachers can play the role of a good teacher in any situation, whether a crisis (like the Covid-19 pandemic) or not. A good teacher always applies the right approach....“ (FM no. 1).\n[SUBTITLE] Subtheme 2: role modeling and student support (heating up the students’ hearts) [SUBSECTION] The professors’ experiences in educating successful students during anatomy courses show that they should reduce unnecessary complaints and criticisms to penetrate their hearts and light the torch of knowledge in their minds. Feelings for the learners should be expressed, and love for them should be shown. There should be a role model for them in academia. If the love of science enlightens students, they are strengthened to endure hardships and problems. One of the professors said in this regard:“…As an ancient Persian anecdote, “to attract the heart of a friend, we must do every trick.“ I sat down during soccer World Cup games and watched part of the game with the students…” (FM no. 8)\n“…As an ancient Persian anecdote, “to attract the heart of a friend, we must do every trick.“ I sat down during soccer World Cup games and watched part of the game with the students…” (FM no. 8)\nThe professors’ experiences in educating successful students during anatomy courses show that they should reduce unnecessary complaints and criticisms to penetrate their hearts and light the torch of knowledge in their minds. Feelings for the learners should be expressed, and love for them should be shown. There should be a role model for them in academia. If the love of science enlightens students, they are strengthened to endure hardships and problems. One of the professors said in this regard:“…As an ancient Persian anecdote, “to attract the heart of a friend, we must do every trick.“ I sat down during soccer World Cup games and watched part of the game with the students…” (FM no. 8)\n“…As an ancient Persian anecdote, “to attract the heart of a friend, we must do every trick.“ I sat down during soccer World Cup games and watched part of the game with the students…” (FM no. 8)\n[SUBTITLE] Joyful teaching (telling stories/games/humor) [SUBSECTION] According to the participants, turning hard work into enjoyable teaching and learning is the art of medical teachers. A few faculty members believe that humor and mixing lessons with fun and games can make students aware of their superior position as a professor. They find icebreaking essential at the beginning of each discussion.“…If students enjoy the class and assume that learning is a plausible activity, they will not waste their time. Most of the wasted time and students’ naughtiness occur because of the attractiveness of the classroom and teacher….“ (FM no. 10)\n“…If students enjoy the class and assume that learning is a plausible activity, they will not waste their time. Most of the wasted time and students’ naughtiness occur because of the attractiveness of the classroom and teacher….“ (FM no. 10)\nSuch an approach strengthens the bond between the students and teachers and leads to effective role modeling.“... If the student does not follow me in the classroom, what is the benefit of my teaching class?“ (FM no..8)\n“... If the student does not follow me in the classroom, what is the benefit of my teaching class?“ (FM no..8)\nStudents can understand whether their teacher can be a trusted and knowledgeable role model and respond better if they encounter a fun and supportive teacher.“…good teachers are fully aware of all their behavior and movements and know what to teach in the classroom. They avoid traditional teaching methods and teach enjoyable for both students and teachers. Students will not be tired and enjoy the learning process….“ (Student no. 10)\n“…good teachers are fully aware of all their behavior and movements and know what to teach in the classroom. They avoid traditional teaching methods and teach enjoyable for both students and teachers. Students will not be tired and enjoy the learning process….“ (Student no. 10)\nAccording to the participants, turning hard work into enjoyable teaching and learning is the art of medical teachers. A few faculty members believe that humor and mixing lessons with fun and games can make students aware of their superior position as a professor. They find icebreaking essential at the beginning of each discussion.“…If students enjoy the class and assume that learning is a plausible activity, they will not waste their time. Most of the wasted time and students’ naughtiness occur because of the attractiveness of the classroom and teacher….“ (FM no. 10)\n“…If students enjoy the class and assume that learning is a plausible activity, they will not waste their time. Most of the wasted time and students’ naughtiness occur because of the attractiveness of the classroom and teacher….“ (FM no. 10)\nSuch an approach strengthens the bond between the students and teachers and leads to effective role modeling.“... If the student does not follow me in the classroom, what is the benefit of my teaching class?“ (FM no..8)\n“... If the student does not follow me in the classroom, what is the benefit of my teaching class?“ (FM no..8)\nStudents can understand whether their teacher can be a trusted and knowledgeable role model and respond better if they encounter a fun and supportive teacher.“…good teachers are fully aware of all their behavior and movements and know what to teach in the classroom. They avoid traditional teaching methods and teach enjoyable for both students and teachers. Students will not be tired and enjoy the learning process….“ (Student no. 10)\n“…good teachers are fully aware of all their behavior and movements and know what to teach in the classroom. They avoid traditional teaching methods and teach enjoyable for both students and teachers. Students will not be tired and enjoy the learning process….“ (Student no. 10)\n[SUBTITLE] Student Support System [SUBSECTION] Both faculty members and students mentioned the student support system as influential. All the participants believe that student support is helpful for vulnerable students, those with learning disabilities, and successful and elite students. The faculty members referred them to counseling, helped them solve their educational and emotional problems, and supported their families emotionally and financially.“…Student support is essential, but it is not just for high-risk and susceptible students. Sometimes elites need even more assistance, and I have seen and experienced how sometimes talented kids are vulnerable...“ (FM no.8).\n“…Student support is essential, but it is not just for high-risk and susceptible students. Sometimes elites need even more assistance, and I have seen and experienced how sometimes talented kids are vulnerable...“ (FM no.8).\nSuch support, mainly when anatomy professors are referred to as the first role models for medical students, motivates students to improve over the past.“…I had the support of the family and could work harder and enjoy learning; they did not hesitate to do anything for me. The teachers and faculty members were sometimes right supporters and solved my problem….“ (Student no. 3)\n“…I had the support of the family and could work harder and enjoy learning; they did not hesitate to do anything for me. The teachers and faculty members were sometimes right supporters and solved my problem….“ (Student no. 3)\nBoth faculty members and students mentioned the student support system as influential. All the participants believe that student support is helpful for vulnerable students, those with learning disabilities, and successful and elite students. The faculty members referred them to counseling, helped them solve their educational and emotional problems, and supported their families emotionally and financially.“…Student support is essential, but it is not just for high-risk and susceptible students. Sometimes elites need even more assistance, and I have seen and experienced how sometimes talented kids are vulnerable...“ (FM no.8).\n“…Student support is essential, but it is not just for high-risk and susceptible students. Sometimes elites need even more assistance, and I have seen and experienced how sometimes talented kids are vulnerable...“ (FM no.8).\nSuch support, mainly when anatomy professors are referred to as the first role models for medical students, motivates students to improve over the past.“…I had the support of the family and could work harder and enjoy learning; they did not hesitate to do anything for me. The teachers and faculty members were sometimes right supporters and solved my problem….“ (Student no. 3)\n“…I had the support of the family and could work harder and enjoy learning; they did not hesitate to do anything for me. The teachers and faculty members were sometimes right supporters and solved my problem….“ (Student no. 3)\n[SUBTITLE] Subtheme 3: sledgehammer approach (thorough engagement in the age of pandemics) [SUBSECTION] While anatomy is perceived as one of the most challenging courses for preclinical students, the advent of the Covid-19 pandemic adds to the challenge. Successful anatomy teachers state that they must apply a combination of educational theories quickly, seamlessly, and effectively in the classroom or online. They believe that teaching anatomy in pandemic situations should be accompanied by joy and vivacity and increase the moments of discovery and intuition for learning. The faculty members seem to struggle to get the most involvement from students.\nTeaching approaches that trigger student involvement include a study guide, reference autonomy, neuroanatomical engagement, drawing, problem-based learning, and peer-based learning.\nWhile anatomy is perceived as one of the most challenging courses for preclinical students, the advent of the Covid-19 pandemic adds to the challenge. Successful anatomy teachers state that they must apply a combination of educational theories quickly, seamlessly, and effectively in the classroom or online. They believe that teaching anatomy in pandemic situations should be accompanied by joy and vivacity and increase the moments of discovery and intuition for learning. The faculty members seem to struggle to get the most involvement from students.\nTeaching approaches that trigger student involvement include a study guide, reference autonomy, neuroanatomical engagement, drawing, problem-based learning, and peer-based learning.\n[SUBTITLE] Study guide [SUBSECTION] Most participants believe preparedness is an essential strategy for success in learning and teaching. The best way is to provide a friendly study guide. The study guide is like a travel guide designed by an instructor to guide learners. It can increase the student’s insight into the subject. Some professors had the experience of contributing to making “road map” books for anatomy. They believe that such a study guide is also a studying resource. Others found the study guide essential for managing the students’ learning.“... Student guidance is an important issue that needs to be planned and implemented. We need more insight and hard work in pandemic conditions. Our “road map” book that primarily serves as a reference of anatomy is a study guide as well…” (FM no. 3)\n“... Student guidance is an important issue that needs to be planned and implemented. We need more insight and hard work in pandemic conditions. Our “road map” book that primarily serves as a reference of anatomy is a study guide as well…” (FM no. 3)\nStudents assume that having a study guide is an insightful experience with more potential than it seems. They argue that a good stud guide must include common mistakes and guide them in all learning experiences, especially in cadaver dissection sessions.“... The study guide helps a lot in learning, and it is excellent if students’ common mistakes are made in this lesson, for example, in the lab and when we dissect the cadaver ...“ (FM no. 6)\n“... The study guide helps a lot in learning, and it is excellent if students’ common mistakes are made in this lesson, for example, in the lab and when we dissect the cadaver ...“ (FM no. 6)\nMost participants believe preparedness is an essential strategy for success in learning and teaching. The best way is to provide a friendly study guide. The study guide is like a travel guide designed by an instructor to guide learners. It can increase the student’s insight into the subject. Some professors had the experience of contributing to making “road map” books for anatomy. They believe that such a study guide is also a studying resource. Others found the study guide essential for managing the students’ learning.“... Student guidance is an important issue that needs to be planned and implemented. We need more insight and hard work in pandemic conditions. Our “road map” book that primarily serves as a reference of anatomy is a study guide as well…” (FM no. 3)\n“... Student guidance is an important issue that needs to be planned and implemented. We need more insight and hard work in pandemic conditions. Our “road map” book that primarily serves as a reference of anatomy is a study guide as well…” (FM no. 3)\nStudents assume that having a study guide is an insightful experience with more potential than it seems. They argue that a good stud guide must include common mistakes and guide them in all learning experiences, especially in cadaver dissection sessions.“... The study guide helps a lot in learning, and it is excellent if students’ common mistakes are made in this lesson, for example, in the lab and when we dissect the cadaver ...“ (FM no. 6)\n“... The study guide helps a lot in learning, and it is excellent if students’ common mistakes are made in this lesson, for example, in the lab and when we dissect the cadaver ...“ (FM no. 6)\n[SUBTITLE] Neuroanatomical engagement [SUBSECTION] Participants believed that people learned new abstract concepts better when presented in visual and auditory ways. Professors believed that due to the functions of the brain hemisphere, the information process occurred through two separate systems in the brain. The right half is dedicated to the image, and the left half is word processing. Visual learning complements auditory learning. Sensory learning, which happens by working on the cadaver, complements student learning. Such a multidimensional approach leads to long-term memorization.“…I always use fascinating slides and video clips. If necessary, I explain my experience through the film, which enhances the understanding and facilitates long-term memorization…” (FM no. 5)\n“…I always use fascinating slides and video clips. If necessary, I explain my experience through the film, which enhances the understanding and facilitates long-term memorization…” (FM no. 5)\nParticipants believed that people learned new abstract concepts better when presented in visual and auditory ways. Professors believed that due to the functions of the brain hemisphere, the information process occurred through two separate systems in the brain. The right half is dedicated to the image, and the left half is word processing. Visual learning complements auditory learning. Sensory learning, which happens by working on the cadaver, complements student learning. Such a multidimensional approach leads to long-term memorization.“…I always use fascinating slides and video clips. If necessary, I explain my experience through the film, which enhances the understanding and facilitates long-term memorization…” (FM no. 5)\n“…I always use fascinating slides and video clips. If necessary, I explain my experience through the film, which enhances the understanding and facilitates long-term memorization…” (FM no. 5)\n[SUBTITLE] Reference autonomy [SUBSECTION] According to the participants, appropriate resources play an essential role in learning. A good resource should encourage the students to become more active and reinforce their thinking, reasoning, and comprehension. Resources must be dynamic to adapt in times of crisis (pandemic) and change to meet the needs of 21st-century learners using current technologies. There seems to be a demand for respecting autonomy in both student and teacher experiences. They react better when students feel free and supported to choose their resources.“…For anatomy, I use Gray’s Anatomy book. However, recently, one of my professors introduced Moore’s Anatomy book. I felt this book was better in the field of …” (Student no. 2)\n“…For anatomy, I use Gray’s Anatomy book. However, recently, one of my professors introduced Moore’s Anatomy book. I felt this book was better in the field of …” (Student no. 2)\nTeachers argue that if provided with a dynamic curriculum design that encompasses clear goals, they can select from all the available resources and provide better ones. They believe that students should be taught to search and find valuable resources when they are highly interested in topics.\nWhen teachers accept the principle of self-learning and self-guidance, they listen to their students feel more confident, leading to more enthusiasm and thus creating future lifelong learners.“…I had a student who came up to me and asked me to explain how to learn anatomy. I was surprised because I was unfamiliar with the question. I told him to get two books, look at them, and return them to me the day after. He came while he got many valuable points, and now he is looking for further resources. Now, he is one of my most successful students, and one of my most important duties is introducing excellent resources to students...“ (FM no. 4)\n“…I had a student who came up to me and asked me to explain how to learn anatomy. I was surprised because I was unfamiliar with the question. I told him to get two books, look at them, and return them to me the day after. He came while he got many valuable points, and now he is looking for further resources. Now, he is one of my most successful students, and one of my most important duties is introducing excellent resources to students...“ (FM no. 4)\nAccording to the participants, appropriate resources play an essential role in learning. A good resource should encourage the students to become more active and reinforce their thinking, reasoning, and comprehension. Resources must be dynamic to adapt in times of crisis (pandemic) and change to meet the needs of 21st-century learners using current technologies. There seems to be a demand for respecting autonomy in both student and teacher experiences. They react better when students feel free and supported to choose their resources.“…For anatomy, I use Gray’s Anatomy book. However, recently, one of my professors introduced Moore’s Anatomy book. I felt this book was better in the field of …” (Student no. 2)\n“…For anatomy, I use Gray’s Anatomy book. However, recently, one of my professors introduced Moore’s Anatomy book. I felt this book was better in the field of …” (Student no. 2)\nTeachers argue that if provided with a dynamic curriculum design that encompasses clear goals, they can select from all the available resources and provide better ones. They believe that students should be taught to search and find valuable resources when they are highly interested in topics.\nWhen teachers accept the principle of self-learning and self-guidance, they listen to their students feel more confident, leading to more enthusiasm and thus creating future lifelong learners.“…I had a student who came up to me and asked me to explain how to learn anatomy. I was surprised because I was unfamiliar with the question. I told him to get two books, look at them, and return them to me the day after. He came while he got many valuable points, and now he is looking for further resources. Now, he is one of my most successful students, and one of my most important duties is introducing excellent resources to students...“ (FM no. 4)\n“…I had a student who came up to me and asked me to explain how to learn anatomy. I was surprised because I was unfamiliar with the question. I told him to get two books, look at them, and return them to me the day after. He came while he got many valuable points, and now he is looking for further resources. Now, he is one of my most successful students, and one of my most important duties is introducing excellent resources to students...“ (FM no. 4)\n[SUBTITLE] Mindful dissection, better transition [SUBSECTION] The first official and scientific encounter of young students with the human body occurs in dissection sessions. The first sessions familiarize the students with the disgusting aspects of gross medical procedures in which teachers play a facilitating role in such transition.“…I did not feel good on the first day, and many of my friends could not see the cadavers. It smelled so bad, but when we got used to it and started learning, we had better feelings, especially when we had a practical examination …” (Student no.14)\n“…I did not feel good on the first day, and many of my friends could not see the cadavers. It smelled so bad, but when we got used to it and started learning, we had better feelings, especially when we had a practical examination …” (Student no.14)\nAnatomy teachers are the game changers who can seed respect towards the human body in students’ minds.“… A cadaver is the most important teaching tool in the anatomy course and serves as a teacher’s blackboard. It is full of teaching points for medical students. I think this is ethically important as well. They should learn to respect the dead body highly to show respect to the living body in the future….“ (FM no.12)\n“… A cadaver is the most important teaching tool in the anatomy course and serves as a teacher’s blackboard. It is full of teaching points for medical students. I think this is ethically important as well. They should learn to respect the dead body highly to show respect to the living body in the future….“ (FM no.12)\nThe first official and scientific encounter of young students with the human body occurs in dissection sessions. The first sessions familiarize the students with the disgusting aspects of gross medical procedures in which teachers play a facilitating role in such transition.“…I did not feel good on the first day, and many of my friends could not see the cadavers. It smelled so bad, but when we got used to it and started learning, we had better feelings, especially when we had a practical examination …” (Student no.14)\n“…I did not feel good on the first day, and many of my friends could not see the cadavers. It smelled so bad, but when we got used to it and started learning, we had better feelings, especially when we had a practical examination …” (Student no.14)\nAnatomy teachers are the game changers who can seed respect towards the human body in students’ minds.“… A cadaver is the most important teaching tool in the anatomy course and serves as a teacher’s blackboard. It is full of teaching points for medical students. I think this is ethically important as well. They should learn to respect the dead body highly to show respect to the living body in the future….“ (FM no.12)\n“… A cadaver is the most important teaching tool in the anatomy course and serves as a teacher’s blackboard. It is full of teaching points for medical students. I think this is ethically important as well. They should learn to respect the dead body highly to show respect to the living body in the future….“ (FM no.12)\n[SUBTITLE] Drawing [SUBSECTION] Teachers and students have valuable experiences in learning and memorizing through drawing. Drawing helps them to understand the topic, consolidate what they have learned, and remember it in long-term memory.“…Anatomy has a regional nature. It means that, for instance, everything in the chest area should be learned simultaneously and as a whole. I use drawing to show the adjacency of structures so that students can better learn and consolidate the topic...“ (FM no. 3)\n“…Anatomy has a regional nature. It means that, for instance, everything in the chest area should be learned simultaneously and as a whole. I use drawing to show the adjacency of structures so that students can better learn and consolidate the topic...“ (FM no. 3)\nFor some learners, drawing becomes a ritual; one of the talented students said:“…I always tell other students to draw pictures to understand anatomy correctly. Draw a lot! Anatomy cannot be understood without painting. No matter how strong you are in abstract thinking, still, draw! I had a notebook in which I was painting...“ (Student no.5)\n“…I always tell other students to draw pictures to understand anatomy correctly. Draw a lot! Anatomy cannot be understood without painting. No matter how strong you are in abstract thinking, still, draw! I had a notebook in which I was painting...“ (Student no.5)\nTeachers and students have valuable experiences in learning and memorizing through drawing. Drawing helps them to understand the topic, consolidate what they have learned, and remember it in long-term memory.“…Anatomy has a regional nature. It means that, for instance, everything in the chest area should be learned simultaneously and as a whole. I use drawing to show the adjacency of structures so that students can better learn and consolidate the topic...“ (FM no. 3)\n“…Anatomy has a regional nature. It means that, for instance, everything in the chest area should be learned simultaneously and as a whole. I use drawing to show the adjacency of structures so that students can better learn and consolidate the topic...“ (FM no. 3)\nFor some learners, drawing becomes a ritual; one of the talented students said:“…I always tell other students to draw pictures to understand anatomy correctly. Draw a lot! Anatomy cannot be understood without painting. No matter how strong you are in abstract thinking, still, draw! I had a notebook in which I was painting...“ (Student no.5)\n“…I always tell other students to draw pictures to understand anatomy correctly. Draw a lot! Anatomy cannot be understood without painting. No matter how strong you are in abstract thinking, still, draw! I had a notebook in which I was painting...“ (Student no.5)\n[SUBTITLE] Problem-based learning [SUBSECTION] Students will feel more satisfied if they are involved in future career problems. By using this approach, students are also emotionally involved in future challenges and are taught to enjoy solving future issues. Those problems that include detailed scene descriptions in a socio-emotional context and are related to cultural, moral, and social values seem to engage students better, help long-term memorization, and facilitate transition into future doctors.\nOne of the professors said:“… I will show the students a picture of the patient and tell them that the person has subarachnoid hemorrhage. Look at the patient’s family. You can see their deep worry. Tell me where the bleeding source is and what is the first step?... \" (FM no. 6)\n“… I will show the students a picture of the patient and tell them that the person has subarachnoid hemorrhage. Look at the patient’s family. You can see their deep worry. Tell me where the bleeding source is and what is the first step?... \" (FM no. 6)\nStudents will feel more satisfied if they are involved in future career problems. By using this approach, students are also emotionally involved in future challenges and are taught to enjoy solving future issues. Those problems that include detailed scene descriptions in a socio-emotional context and are related to cultural, moral, and social values seem to engage students better, help long-term memorization, and facilitate transition into future doctors.\nOne of the professors said:“… I will show the students a picture of the patient and tell them that the person has subarachnoid hemorrhage. Look at the patient’s family. You can see their deep worry. Tell me where the bleeding source is and what is the first step?... \" (FM no. 6)\n“… I will show the students a picture of the patient and tell them that the person has subarachnoid hemorrhage. Look at the patient’s family. You can see their deep worry. Tell me where the bleeding source is and what is the first step?... \" (FM no. 6)\n[SUBTITLE] Peer-assisted learning [SUBSECTION] Students stated that learning in small groups was attractive because they acquired interpersonal skills like listening, speaking, discussing, and group leadership. Besides, it promotes higher cognitive abilities such as reasoning and problem-solving.“...For me, the best learning tool is a peer to peer group work. I have good experience in all subjects from literature to anatomy utilizing this method…” (Student no.5)\n“...For me, the best learning tool is a peer to peer group work. I have good experience in all subjects from literature to anatomy utilizing this method…” (Student no.5)\nStudents stated that learning in small groups was attractive because they acquired interpersonal skills like listening, speaking, discussing, and group leadership. Besides, it promotes higher cognitive abilities such as reasoning and problem-solving.“...For me, the best learning tool is a peer to peer group work. I have good experience in all subjects from literature to anatomy utilizing this method…” (Student no.5)\n“...For me, the best learning tool is a peer to peer group work. I have good experience in all subjects from literature to anatomy utilizing this method…” (Student no.5)", "In this study, semi-structured interviews were performed with 24 medical students from year 1 to senior interns, 15 (62.5% were males), 9 (37.5% were females), and 12 faculty members, 6 (50% were males), 6 (50% were females), with various academic ranks and experience from 9 (Shiraz, Tehran, Fasa, Jahrom, Bandarabas, Yasuj, Gerash, Larestan, and Bushehr) Iranian medical schools. The participants’ age ranged from 19 to 62 years old, with a mean of 33.4 ± 8.4 years. Based on the results, 12 students (50%) were in the clinical training period as students, externs, and interns, and 12 participants (50%) were in the basic sciences period. Accordingly, five participants (20%) had a history of being top students, and four (16.7%) had a history of dropout in anatomy courses. Furthermore, the work experience of the faculty members ranged from 5 to 35 years in teaching anatomy; 9(75%) anatomy faculty members and 3(25%) experts in medical education; faculty member participants, 3(25%) were single, and 9 (75%) were married. (Table 1)\n\nTable 1Demographic Characteristics of the Study ParticipantsParticipantsCharacteristics\nstudents\n\nGender\nNoPercent (%)\nMean Age (y ± SD)\nMale1562.522.3 ± 8.4Female937.5\nDegree\nFaculty membersassistant professors,32537.4 ± 11.7associated professors541.67full professors433.34\n\nDemographic Characteristics of the Study Participants\nFrom all the interviews, 334 codes were extracted. After the dismissal of similar codes and their integration, the experience of medical students was conceptualized into a central theme called the blacksmith approach, which included Three sub-themes: (1) making a new forge (Adequate preparation and mindful beginning), (2) heating the students’ hearts (considering supporting systems that learners need) and (3) using a Sledgehammer’s approach (teaching Anatomy thorough engaging all neuroanatomical regions in the Age of Pandemics). All the concepts were related to each other and resulted in a pattern revealing the experience of students and faculty members on strategies for successful learning and teaching of anatomy lessons. The main themes, as well as subordinate themes, are listed in Table 2.\n\nTable 2The Blacksmith Approach: Best strategy for teaching and learning in the medical anatomy course. Themes, sub-themes, and codesMain ThemesSub Themescodecods\nBlacksmith Approach\n\nRedesigning Educational Mindset (Making a New Forge)\n\nCreating a Vision for Students\n√ Anatomy is hard, bulky, should be taken seriously√ I can help myself more than anyone√ integrated curriculum√ self-study / Self-confidencescarcely happens in the real world!√ professional identity√ prospective physicians\nMindful Clinical View\n\nRole Modeling and Support (Heating up the Students’ Hearts)\n\nJoyful Teaching (Telling Stories/Games/ Humor)\n√ every trick must be done√ enjoy class√ follow me in the classroom\nStudent Support System\n√ susceptible students√ assistance√ Vulnerable√ right supportersSitting and talking to them\nSledgehammer Approach (Thorough Engagement in the Age of Pandemics)\n\nStudy Guide\n√ Preparedness√ Road map\nReference Autonomy\n√ self-learning√ self-direction\nNeuroanatomical Engagement\n√ Visual Learning√ Auditory Learning√ Sensory Learning√ Brain HemispheresLong-term Memorization\nMindful Dissection, Better Transition\n√ It smells so bad, but we need the observation√ We got used to it√ ethically important√ respect the dead body√ respect to the living body\nDrawing\n√ easy to learn√ Draw a lot\nProblem-Based Learning\n√ Look at the patient’s family√ Deep worry√ What is the next step of the problem?\nPeer-Assisted Learning\n√ Peer to the peer group√ Discussing the problem\n\nThe Blacksmith Approach: Best strategy for teaching and learning in the medical anatomy course. Themes, sub-themes, and codes\n√ Anatomy is hard, bulky, should be taken seriously\n√ I can help myself more than anyone\n√ integrated curriculum\n√ self-study / Self-confidence\nscarcely happens in the real world!\n√ professional identity\n√ prospective physicians\n√ every trick must be done\n√ enjoy class\n√ follow me in the classroom\n√ susceptible students\n√ assistance\n√ Vulnerable\n√ right supporters\nSitting and talking to them\n√ Preparedness\n√ Road map\n√ self-learning\n√ self-direction\n√ Visual Learning\n√ Auditory Learning\n√ Sensory Learning\n√ Brain Hemispheres\nLong-term Memorization\n√ It smells so bad, but we need the observation\n√ We got used to it\n√ ethically important\n√ respect the dead body\n√ respect to the living body\n√ easy to learn\n√ Draw a lot\n√ Look at the patient’s family\n√ Deep worry\n√ What is the next step of the problem?\n√ Peer to the peer group\n√ Discussing the problem", "According to the participants, in the student’s learning and maturity process, the teacher and the student act rationally and consciously and are aware of the factors that motivate and control them. Thus, they accept the risks, specific conditions, and hardships that must be tolerated. The student should handle this hardship to acquire a capacity to change, and the teacher should facilitate the change and their primary goal of teaching. In a sense, this is the same approach as the Blacksmiths. Blacksmithing symbolizes endurance, honor, and work under challenging conditions in our culture. (Fig. 1) We took the central theme from an interview with a professor who said:\n\nFig. 1The role of the blacksmith approach in the stages of teaching and learning anatomy in medical students is based on the study’s results. First, the blacksmith (educational system) must prepare a new blacksmith (redesign the educational mentality and create a new perspective on anatomical knowledge in students) and prepare a new forge; then, he/she must heat the metal (supporting young students and having good role-modeling) and then shape the metal with a sledgehammer (the sledgehammer approach to using different teaching strategies and methods) to create a sword or self-confident and knowledgeable learner in medicine (professionalism)\n”…The Sledgehammer should be heavy. Its nature is heavy; otherwise, it will not be capable of smoothing any iron. The nature of some courses, such as anatomy, is challenging, but we must make it enjoyable for the student….“ (FM no.6).\n\nThe role of the blacksmith approach in the stages of teaching and learning anatomy in medical students is based on the study’s results. First, the blacksmith (educational system) must prepare a new blacksmith (redesign the educational mentality and create a new perspective on anatomical knowledge in students) and prepare a new forge; then, he/she must heat the metal (supporting young students and having good role-modeling) and then shape the metal with a sledgehammer (the sledgehammer approach to using different teaching strategies and methods) to create a sword or self-confident and knowledgeable learner in medicine (professionalism)\n”…The Sledgehammer should be heavy. Its nature is heavy; otherwise, it will not be capable of smoothing any iron. The nature of some courses, such as anatomy, is challenging, but we must make it enjoyable for the student….“ (FM no.6).", "Creating an educational context is the first step for young students to begin their transition in the learning process. This context was found to have two aspects as follows:\n\nCreating a Vision for Students.\n\nCreating a Vision for Students.\nSome students mentioned a change in their mindset from the beginning with the idea that they have a difficult task ahead and should know what is helpful or not. In addition, realizing that the student is the most important person to help themselves seemed to provide a better sense of success.\nAccording to the participants, anatomy is one of the most voluminous and complex courses in medicine, which students should take seriously from the beginning.“Anatomy is hard, bulky, and should be taken seriously.“ (Student no. 9)\n“Anatomy is hard, bulky, and should be taken seriously.“ (Student no. 9)\nEven though many have oversimplified it, successful students have attempted to learn it through vision and self-awareness.“… After entering the school, my mentor advised me to study anatomy as soon as possible. An anatomy lesson is not something you would say I will study and comprehend the concepts overnight or for a week. You should attend the classes regularly. Students should provide accessible learning resources and not seek shortcuts…” \" (Student no. 1).\n“… After entering the school, my mentor advised me to study anatomy as soon as possible. An anatomy lesson is not something you would say I will study and comprehend the concepts overnight or for a week. You should attend the classes regularly. Students should provide accessible learning resources and not seek shortcuts…” \" (Student no. 1).\nParticipants believed that successful students have good self-confidence and leadership in their learning process. Some students emphasized that the teachers can first build self-confidence and make them self-centered and self-regulated in learning. This concept requires the teachers to listen to them.“... If I realized the concepts satisfactorily, I would get a lot out of my effort. First, I accepted that anatomy is complicated, but I can help myself more than anyone. I started self-study. More action boosts self-confidence, which is required to become a doctor.“(Student no. 6)\n“... If I realized the concepts satisfactorily, I would get a lot out of my effort. First, I accepted that anatomy is complicated, but I can help myself more than anyone. I started self-study. More action boosts self-confidence, which is required to become a doctor.“(Student no. 6)\nFinally, the participants emphasized the importance of integration. Integration is simply defined as merging relevant content or subject areas. Teachers mention that the goal is to integrate and create a general perspective rather than a fragmented perspective of concepts. Some students believed that while multidisciplinary integration was provided, professors’ art of education, such as mastery of the teaching and learning process, generated the necessary harmony between the content presented. If well-coordinated, all aspects mutually reinforce each other, promoting learning in an integrated way.“... I enjoyed our integrated curriculum because that led to a better understanding of medicine. For example, discussing the anatomy of the gastrointestinal system gave me invaluable and integrated knowledge, and I enjoyed it ... “(Student no. 4)\n“... I enjoyed our integrated curriculum because that led to a better understanding of medicine. For example, discussing the anatomy of the gastrointestinal system gave me invaluable and integrated knowledge, and I enjoyed it ... “(Student no. 4)\nAnatomy professors acknowledged that integration with clinical courses had changed their perspective, reflected in their teaching.“…During the integration process and while I was listening to the viewpoints of the clinical colleagues, I realized what I thought to be the crucial thing that students should all be aware of scarcely happens in the real world!...“ (FM no. 2)\n“…During the integration process and while I was listening to the viewpoints of the clinical colleagues, I realized what I thought to be the crucial thing that students should all be aware of scarcely happens in the real world!...“ (FM no. 2)\n\n2)Mindful Clinical View.\n\nMindful Clinical View.\nMindfulness is defined as awareness of what is happening at the present moment. Some professors believed that they must be aware of the ultimate goal while teaching at each moment. Therefore, mindfulness is influenced by one’s dreams.“…I try to teach anatomy in a way that our current students and prospective physicians can diagnose heart enlargement in the future…” (FM no. 7)\n“…I try to teach anatomy in a way that our current students and prospective physicians can diagnose heart enlargement in the future…” (FM no. 7)\nIf anatomy teachers gain a conservative view, the students can achieve integrity and professional identity as doctors.“... The student’s learning and professional identity depend largely on knowing and comprehending anatomy. Teachers can play the role of a good teacher in any situation, whether a crisis (like the Covid-19 pandemic) or not. A good teacher always applies the right approach....“ (FM no. 1).\n“... The student’s learning and professional identity depend largely on knowing and comprehending anatomy. Teachers can play the role of a good teacher in any situation, whether a crisis (like the Covid-19 pandemic) or not. A good teacher always applies the right approach....“ (FM no. 1).", "The professors’ experiences in educating successful students during anatomy courses show that they should reduce unnecessary complaints and criticisms to penetrate their hearts and light the torch of knowledge in their minds. Feelings for the learners should be expressed, and love for them should be shown. There should be a role model for them in academia. If the love of science enlightens students, they are strengthened to endure hardships and problems. One of the professors said in this regard:“…As an ancient Persian anecdote, “to attract the heart of a friend, we must do every trick.“ I sat down during soccer World Cup games and watched part of the game with the students…” (FM no. 8)\n“…As an ancient Persian anecdote, “to attract the heart of a friend, we must do every trick.“ I sat down during soccer World Cup games and watched part of the game with the students…” (FM no. 8)", "According to the participants, turning hard work into enjoyable teaching and learning is the art of medical teachers. A few faculty members believe that humor and mixing lessons with fun and games can make students aware of their superior position as a professor. They find icebreaking essential at the beginning of each discussion.“…If students enjoy the class and assume that learning is a plausible activity, they will not waste their time. Most of the wasted time and students’ naughtiness occur because of the attractiveness of the classroom and teacher….“ (FM no. 10)\n“…If students enjoy the class and assume that learning is a plausible activity, they will not waste their time. Most of the wasted time and students’ naughtiness occur because of the attractiveness of the classroom and teacher….“ (FM no. 10)\nSuch an approach strengthens the bond between the students and teachers and leads to effective role modeling.“... If the student does not follow me in the classroom, what is the benefit of my teaching class?“ (FM no..8)\n“... If the student does not follow me in the classroom, what is the benefit of my teaching class?“ (FM no..8)\nStudents can understand whether their teacher can be a trusted and knowledgeable role model and respond better if they encounter a fun and supportive teacher.“…good teachers are fully aware of all their behavior and movements and know what to teach in the classroom. They avoid traditional teaching methods and teach enjoyable for both students and teachers. Students will not be tired and enjoy the learning process….“ (Student no. 10)\n“…good teachers are fully aware of all their behavior and movements and know what to teach in the classroom. They avoid traditional teaching methods and teach enjoyable for both students and teachers. Students will not be tired and enjoy the learning process….“ (Student no. 10)", "Both faculty members and students mentioned the student support system as influential. All the participants believe that student support is helpful for vulnerable students, those with learning disabilities, and successful and elite students. The faculty members referred them to counseling, helped them solve their educational and emotional problems, and supported their families emotionally and financially.“…Student support is essential, but it is not just for high-risk and susceptible students. Sometimes elites need even more assistance, and I have seen and experienced how sometimes talented kids are vulnerable...“ (FM no.8).\n“…Student support is essential, but it is not just for high-risk and susceptible students. Sometimes elites need even more assistance, and I have seen and experienced how sometimes talented kids are vulnerable...“ (FM no.8).\nSuch support, mainly when anatomy professors are referred to as the first role models for medical students, motivates students to improve over the past.“…I had the support of the family and could work harder and enjoy learning; they did not hesitate to do anything for me. The teachers and faculty members were sometimes right supporters and solved my problem….“ (Student no. 3)\n“…I had the support of the family and could work harder and enjoy learning; they did not hesitate to do anything for me. The teachers and faculty members were sometimes right supporters and solved my problem….“ (Student no. 3)", "While anatomy is perceived as one of the most challenging courses for preclinical students, the advent of the Covid-19 pandemic adds to the challenge. Successful anatomy teachers state that they must apply a combination of educational theories quickly, seamlessly, and effectively in the classroom or online. They believe that teaching anatomy in pandemic situations should be accompanied by joy and vivacity and increase the moments of discovery and intuition for learning. The faculty members seem to struggle to get the most involvement from students.\nTeaching approaches that trigger student involvement include a study guide, reference autonomy, neuroanatomical engagement, drawing, problem-based learning, and peer-based learning.", "Most participants believe preparedness is an essential strategy for success in learning and teaching. The best way is to provide a friendly study guide. The study guide is like a travel guide designed by an instructor to guide learners. It can increase the student’s insight into the subject. Some professors had the experience of contributing to making “road map” books for anatomy. They believe that such a study guide is also a studying resource. Others found the study guide essential for managing the students’ learning.“... Student guidance is an important issue that needs to be planned and implemented. We need more insight and hard work in pandemic conditions. Our “road map” book that primarily serves as a reference of anatomy is a study guide as well…” (FM no. 3)\n“... Student guidance is an important issue that needs to be planned and implemented. We need more insight and hard work in pandemic conditions. Our “road map” book that primarily serves as a reference of anatomy is a study guide as well…” (FM no. 3)\nStudents assume that having a study guide is an insightful experience with more potential than it seems. They argue that a good stud guide must include common mistakes and guide them in all learning experiences, especially in cadaver dissection sessions.“... The study guide helps a lot in learning, and it is excellent if students’ common mistakes are made in this lesson, for example, in the lab and when we dissect the cadaver ...“ (FM no. 6)\n“... The study guide helps a lot in learning, and it is excellent if students’ common mistakes are made in this lesson, for example, in the lab and when we dissect the cadaver ...“ (FM no. 6)", "Participants believed that people learned new abstract concepts better when presented in visual and auditory ways. Professors believed that due to the functions of the brain hemisphere, the information process occurred through two separate systems in the brain. The right half is dedicated to the image, and the left half is word processing. Visual learning complements auditory learning. Sensory learning, which happens by working on the cadaver, complements student learning. Such a multidimensional approach leads to long-term memorization.“…I always use fascinating slides and video clips. If necessary, I explain my experience through the film, which enhances the understanding and facilitates long-term memorization…” (FM no. 5)\n“…I always use fascinating slides and video clips. If necessary, I explain my experience through the film, which enhances the understanding and facilitates long-term memorization…” (FM no. 5)", "According to the participants, appropriate resources play an essential role in learning. A good resource should encourage the students to become more active and reinforce their thinking, reasoning, and comprehension. Resources must be dynamic to adapt in times of crisis (pandemic) and change to meet the needs of 21st-century learners using current technologies. There seems to be a demand for respecting autonomy in both student and teacher experiences. They react better when students feel free and supported to choose their resources.“…For anatomy, I use Gray’s Anatomy book. However, recently, one of my professors introduced Moore’s Anatomy book. I felt this book was better in the field of …” (Student no. 2)\n“…For anatomy, I use Gray’s Anatomy book. However, recently, one of my professors introduced Moore’s Anatomy book. I felt this book was better in the field of …” (Student no. 2)\nTeachers argue that if provided with a dynamic curriculum design that encompasses clear goals, they can select from all the available resources and provide better ones. They believe that students should be taught to search and find valuable resources when they are highly interested in topics.\nWhen teachers accept the principle of self-learning and self-guidance, they listen to their students feel more confident, leading to more enthusiasm and thus creating future lifelong learners.“…I had a student who came up to me and asked me to explain how to learn anatomy. I was surprised because I was unfamiliar with the question. I told him to get two books, look at them, and return them to me the day after. He came while he got many valuable points, and now he is looking for further resources. Now, he is one of my most successful students, and one of my most important duties is introducing excellent resources to students...“ (FM no. 4)\n“…I had a student who came up to me and asked me to explain how to learn anatomy. I was surprised because I was unfamiliar with the question. I told him to get two books, look at them, and return them to me the day after. He came while he got many valuable points, and now he is looking for further resources. Now, he is one of my most successful students, and one of my most important duties is introducing excellent resources to students...“ (FM no. 4)", "The first official and scientific encounter of young students with the human body occurs in dissection sessions. The first sessions familiarize the students with the disgusting aspects of gross medical procedures in which teachers play a facilitating role in such transition.“…I did not feel good on the first day, and many of my friends could not see the cadavers. It smelled so bad, but when we got used to it and started learning, we had better feelings, especially when we had a practical examination …” (Student no.14)\n“…I did not feel good on the first day, and many of my friends could not see the cadavers. It smelled so bad, but when we got used to it and started learning, we had better feelings, especially when we had a practical examination …” (Student no.14)\nAnatomy teachers are the game changers who can seed respect towards the human body in students’ minds.“… A cadaver is the most important teaching tool in the anatomy course and serves as a teacher’s blackboard. It is full of teaching points for medical students. I think this is ethically important as well. They should learn to respect the dead body highly to show respect to the living body in the future….“ (FM no.12)\n“… A cadaver is the most important teaching tool in the anatomy course and serves as a teacher’s blackboard. It is full of teaching points for medical students. I think this is ethically important as well. They should learn to respect the dead body highly to show respect to the living body in the future….“ (FM no.12)", "Teachers and students have valuable experiences in learning and memorizing through drawing. Drawing helps them to understand the topic, consolidate what they have learned, and remember it in long-term memory.“…Anatomy has a regional nature. It means that, for instance, everything in the chest area should be learned simultaneously and as a whole. I use drawing to show the adjacency of structures so that students can better learn and consolidate the topic...“ (FM no. 3)\n“…Anatomy has a regional nature. It means that, for instance, everything in the chest area should be learned simultaneously and as a whole. I use drawing to show the adjacency of structures so that students can better learn and consolidate the topic...“ (FM no. 3)\nFor some learners, drawing becomes a ritual; one of the talented students said:“…I always tell other students to draw pictures to understand anatomy correctly. Draw a lot! Anatomy cannot be understood without painting. No matter how strong you are in abstract thinking, still, draw! I had a notebook in which I was painting...“ (Student no.5)\n“…I always tell other students to draw pictures to understand anatomy correctly. Draw a lot! Anatomy cannot be understood without painting. No matter how strong you are in abstract thinking, still, draw! I had a notebook in which I was painting...“ (Student no.5)", "Students will feel more satisfied if they are involved in future career problems. By using this approach, students are also emotionally involved in future challenges and are taught to enjoy solving future issues. Those problems that include detailed scene descriptions in a socio-emotional context and are related to cultural, moral, and social values seem to engage students better, help long-term memorization, and facilitate transition into future doctors.\nOne of the professors said:“… I will show the students a picture of the patient and tell them that the person has subarachnoid hemorrhage. Look at the patient’s family. You can see their deep worry. Tell me where the bleeding source is and what is the first step?... \" (FM no. 6)\n“… I will show the students a picture of the patient and tell them that the person has subarachnoid hemorrhage. Look at the patient’s family. You can see their deep worry. Tell me where the bleeding source is and what is the first step?... \" (FM no. 6)", "Students stated that learning in small groups was attractive because they acquired interpersonal skills like listening, speaking, discussing, and group leadership. Besides, it promotes higher cognitive abilities such as reasoning and problem-solving.“...For me, the best learning tool is a peer to peer group work. I have good experience in all subjects from literature to anatomy utilizing this method…” (Student no.5)\n“...For me, the best learning tool is a peer to peer group work. I have good experience in all subjects from literature to anatomy utilizing this method…” (Student no.5)", "We first scrutinized the experience of teaching and learning anatomy through qualitative analysis. Next, we conceptualized a new approach to help medical students in anatomy courses have more satisfactory learning outcomes and a better transition into resilient future doctors. We called it “the Blacksmith Approach.“ building a sword (prosperous and resilient learner) takes three steps. 1. Creating a new forge (adequate preparation and mindful beginning) 2; heating students’ hearts (support and role modeling) 3. Sledgehammer’s approach (thorough engagement in the age of pandemics). (Fig. 1)\nWhile other proposed educational methods in teaching anatomy focus mainly on learning outcomes during anatomy courses [14–19], the blacksmith approach tries to reach two goals, i.e., better teaching-learning outcomes and helping young students transition into future medical professionals. In fact, this is not another teaching method. It best serves as an educational framework for any pivotal, preclinical course capable of helping students acquire new roles and tackle challenges. In this sense, medical educators can adopt all prior teaching techniques as long as they help students reach better learning outcomes or have better transition experiences.\nThe blacksmith approach can be an educational implication of Schlossberg’s 4 S model for transition. Transition is a process that enables individuals to incorporate change into their life. The 4 S theory argues that an individual goes through a cyclic experience of 4 stages during the transition, i.e., situation, self, support, and strategies [46]. We encounter two types of transition in an educational setting. One happens within the learners and one within the educators. Although more attention is put on the former, we cannot achieve desired outcomes if we neglect the latter. While in the 4 S model, these four stages happen within the individual, the Blacksmith Approach creates a compatible, three-step learning environment for students and teachers to transition by creating a new attitude and mindset for both groups. As Browne et al. stated, individual motivation, background and circumstances, a sense of control, organizational support, effective networking, and information-seeking behavior contributed to a successful transition into and maintaining a strong self-identity as a medical educator [47].\n[SUBTITLE] First Step: A new forge (mindset) [SUBSECTION] must be built for the students. This step is in line with the first stage of the four S model, i.e., Situation awareness. Here, the students become aware of what is happening. The teachers here have two roles. First, they should deliver the following messages to the students: (1) you are supposed to actualize a new identity (role change), (2) anatomy (as a pillar of medicine) provides a problematic task ahead (stress), and (3) you can help yourself more than anyone else. (control) Furthermore, the students should try self-study to increase their self-confidence (trigger). Secondly, the teachers should acquire a mindful clinical standpoint, i.e., developing good clinical knowledge and adopting active learning methods based on teamwork and clinical scenarios that encourage active participation and self-directed learning as if they are future doctors. This strategy has been shown to reduce student stress [11, 13] and help them react better as future physicians [3, 8, 48].\nmust be built for the students. This step is in line with the first stage of the four S model, i.e., Situation awareness. Here, the students become aware of what is happening. The teachers here have two roles. First, they should deliver the following messages to the students: (1) you are supposed to actualize a new identity (role change), (2) anatomy (as a pillar of medicine) provides a problematic task ahead (stress), and (3) you can help yourself more than anyone else. (control) Furthermore, the students should try self-study to increase their self-confidence (trigger). Secondly, the teachers should acquire a mindful clinical standpoint, i.e., developing good clinical knowledge and adopting active learning methods based on teamwork and clinical scenarios that encourage active participation and self-directed learning as if they are future doctors. This strategy has been shown to reduce student stress [11, 13] and help them react better as future physicians [3, 8, 48].\n[SUBTITLE] Second step [SUBSECTION] The students’ hearts must be warmed up through support and role modeling. This step is compatible with the following two stages in the 4 S model, i.e., self and support. The students evaluate their demographic and psychological characteristics, and then they need to be aware of support systems and be able to get help [27, 29]. Here again, the role of anatomy teachers is critical. As medical students see anatomy as a gate to medicine, the anatomy teacher can be known as the gatekeeper. They will inevitably actualize their teachers as wise role models who are righteous supporters. The anatomy faculty member should be aware of their role modeling. Role modeling can help students foster resilience for future change, which leads to professionalism and integrity [7, 18, 49–52]. The teachers should adopt ice-breaking methods, use their sense of humor, tell stories, design games, and have friendly chats with students outside the classroom. In addition, anatomy teachers should gain the skills and knowledge for counseling and get familiar with support strategies to become prominent supporters of students [53–56].\nThe students’ hearts must be warmed up through support and role modeling. This step is compatible with the following two stages in the 4 S model, i.e., self and support. The students evaluate their demographic and psychological characteristics, and then they need to be aware of support systems and be able to get help [27, 29]. Here again, the role of anatomy teachers is critical. As medical students see anatomy as a gate to medicine, the anatomy teacher can be known as the gatekeeper. They will inevitably actualize their teachers as wise role models who are righteous supporters. The anatomy faculty member should be aware of their role modeling. Role modeling can help students foster resilience for future change, which leads to professionalism and integrity [7, 18, 49–52]. The teachers should adopt ice-breaking methods, use their sense of humor, tell stories, design games, and have friendly chats with students outside the classroom. In addition, anatomy teachers should gain the skills and knowledge for counseling and get familiar with support strategies to become prominent supporters of students [53–56].\n[SUBTITLE] Third step: Sledgehammer’s approach [SUBSECTION] We found that, currently, preclinical students are facing two overwhelming stressors. On the one hand, the covid crisis has detached the students from their teachers and resources [15, 18, 57]. On the other hand, students need to handle their transition into knowledgeable medical professionals. Therefore we need to incorporate teaching methods that assist our detached students in coping with their stressors. The last stage in the 4 S model also deals with coping strategies. Students may use several coping strategies like information seeking, selective ignoring, and reframing the challenge [58].\nAccordingly, the third step in our model includes three components; thorough engagement, reframing the situation, and selective information seeking. Regarding the first component, we encourage meticulously choosing more active and engaging learning techniques. Problem-based learning and Peer-based discussions in small groups, drawing, mind mapping, body painting, using play-doh, and interactive video content, are advised. According to Pandey and Zimitat (2007), a practical approach to anatomy education positively correlates with teaching and reflective learning quality. Successful anatomy learning requires balancing memorization with understanding and visualization[59]. Insightful experiences in teaching anatomy with this strategy are found in Australia (head and neck course) [15] and India (musculoskeletal anatomy) [13]. As Goodman et al. (2006) noted in their previous book, those with higher self-esteem and mental mastery are better able to cope with challenges. [60] A successful learning experience gives students a sense of control over their course, while applying a study guide, reference autonomy, and support system can help them gain self-esteem [61–63].\nReframing the situation, As the second component, is where the students need to redefine their roles and activities. Two sets of strategies can help students to reframe. 1) Using problem-based and peer-assisted learning with a flavor of sociocultural contexts can prepare students for better learning in a positive, non-intimidating learning environment to redefine themselves as future doctors for diagnosing and curing patients. Socio-emotional context and bringing drama elements into case scenarios have been recently emphasized to induce resilience and professionalism in medical students (2.14, 64).\nRecent evidence indicates that gamification leads to positive learning outcomes, higher motivation, and engagement. However, several downsides should also be considered, including increased competition and task evaluation difficulties. Besides, it depends highly on the context in which the gamification is being implemented and its users [65–68]. Hamari et al. (2014) have reviewed empirical literature, showing that gamification has proven effective in many fields, most notably education. Studies in education and learning contexts illustrate how the gamification of learning outcomes has been positive by increasing motivation and engagement in learning and enjoyment tasks over time. However, the studies also highlighted some negative points that require attention, such as the effects of increased competition, task evaluation difficulties, and design features. The review indicates that gamification provides positive effects; however, the effects greatly depend on the context in which the gamification is being implemented and the users who use it [66]. Recent studies suggest that joy and humor facilitate a person’s transition into more socially severe roles (like becoming a doctor) [34]. Hence, teachers should use joyful techniques such as painting, ice braking, storytelling, gamification, dance, and music [4, 9, 15, 53].\nThe last component deals with selective information seeking. Students should have a study guide and be free to choose their desirable resources (anatomy reference books, digital resources, or web-based social media videos). We advise the medical school to design a dynamic curriculum encompassing clear goals. In this setting, the study guide can act as a tool for transferring core knowledge in anatomical sciences. As a result, the tedious aspect of bulky content in anatomy courses can be avoided, and the educational efficiency of synchronous and asynchronous learning can be increased in the pandemic era [55, 56, 61, 69, 70].\nThis research is in line with constructive learning theory that emphasizes student-centered learning, which states that students should be actively involved in education and contribute to knowledge creation [71]. In a special effort to create a modern educational curriculum at Stanford University, Prober et al. recommended building a framework of core knowledge, embedding it into rich interactive formats, and encouraging students to pursue knowledge in some deep, but not all, domains [63]. The Blacksmith approach facilitates personal and in-depth pursuits that help transfer students into lifelong learners and adapt to challenging situations [63, 64]. This statement is consistent with Wald and Monteverde (2021). They report that educational resilience is vital in supporting and sustaining healthcare professional identity development and facilitating the progress of students’ moral resilience and complexity, especially in times of pandemic [25].\nAccording to the participants’ viewpoints, the applicability of the blacksmith approach is one of the most important aspects of improving the quality of anatomy teaching and learning in any situation, such as current critical conditions.\nOur university’s online programs were adequately integrated into the curriculum design and used to guide students on a learning journey rather than merely as another resource; perhaps, these tools could be beneficial. However, in such circumstances, the epidemic and sanctions of Iran and the lack of resources, students and professors should take measures to optimize education to facilitate the teaching and learning of this course. Accordingly, from the above point, through a qualitative study, it was assumed that there were signs of successful teaching and learning and internal change in deep search among students and faculty members.\nRevolution in medical education is a global program, and many medical schools have perceived investigation and experienced challenges [63, 64]. Correspondingly, an active and integrated strategy for teaching and learning anatomy has been emphasized in other medical education courses and areas, including a contemporary medical curriculum. Managers, practitioners, and professors can use the elements of this strategy to increase the efficiency of medical education, improve students’ knowledge, skills, and attitudes, and ultimately maintain and promote understanding of anatomy and other courses. Also, from the participant’s perspective, the applicability of students learning experiences and faculty members’ teaching experiences in the context and workplace conditions was one of the factors that changed the view of anatomy as an introductory course in medicine.\nWe found that, currently, preclinical students are facing two overwhelming stressors. On the one hand, the covid crisis has detached the students from their teachers and resources [15, 18, 57]. On the other hand, students need to handle their transition into knowledgeable medical professionals. Therefore we need to incorporate teaching methods that assist our detached students in coping with their stressors. The last stage in the 4 S model also deals with coping strategies. Students may use several coping strategies like information seeking, selective ignoring, and reframing the challenge [58].\nAccordingly, the third step in our model includes three components; thorough engagement, reframing the situation, and selective information seeking. Regarding the first component, we encourage meticulously choosing more active and engaging learning techniques. Problem-based learning and Peer-based discussions in small groups, drawing, mind mapping, body painting, using play-doh, and interactive video content, are advised. According to Pandey and Zimitat (2007), a practical approach to anatomy education positively correlates with teaching and reflective learning quality. Successful anatomy learning requires balancing memorization with understanding and visualization[59]. Insightful experiences in teaching anatomy with this strategy are found in Australia (head and neck course) [15] and India (musculoskeletal anatomy) [13]. As Goodman et al. (2006) noted in their previous book, those with higher self-esteem and mental mastery are better able to cope with challenges. [60] A successful learning experience gives students a sense of control over their course, while applying a study guide, reference autonomy, and support system can help them gain self-esteem [61–63].\nReframing the situation, As the second component, is where the students need to redefine their roles and activities. Two sets of strategies can help students to reframe. 1) Using problem-based and peer-assisted learning with a flavor of sociocultural contexts can prepare students for better learning in a positive, non-intimidating learning environment to redefine themselves as future doctors for diagnosing and curing patients. Socio-emotional context and bringing drama elements into case scenarios have been recently emphasized to induce resilience and professionalism in medical students (2.14, 64).\nRecent evidence indicates that gamification leads to positive learning outcomes, higher motivation, and engagement. However, several downsides should also be considered, including increased competition and task evaluation difficulties. Besides, it depends highly on the context in which the gamification is being implemented and its users [65–68]. Hamari et al. (2014) have reviewed empirical literature, showing that gamification has proven effective in many fields, most notably education. Studies in education and learning contexts illustrate how the gamification of learning outcomes has been positive by increasing motivation and engagement in learning and enjoyment tasks over time. However, the studies also highlighted some negative points that require attention, such as the effects of increased competition, task evaluation difficulties, and design features. The review indicates that gamification provides positive effects; however, the effects greatly depend on the context in which the gamification is being implemented and the users who use it [66]. Recent studies suggest that joy and humor facilitate a person’s transition into more socially severe roles (like becoming a doctor) [34]. Hence, teachers should use joyful techniques such as painting, ice braking, storytelling, gamification, dance, and music [4, 9, 15, 53].\nThe last component deals with selective information seeking. Students should have a study guide and be free to choose their desirable resources (anatomy reference books, digital resources, or web-based social media videos). We advise the medical school to design a dynamic curriculum encompassing clear goals. In this setting, the study guide can act as a tool for transferring core knowledge in anatomical sciences. As a result, the tedious aspect of bulky content in anatomy courses can be avoided, and the educational efficiency of synchronous and asynchronous learning can be increased in the pandemic era [55, 56, 61, 69, 70].\nThis research is in line with constructive learning theory that emphasizes student-centered learning, which states that students should be actively involved in education and contribute to knowledge creation [71]. In a special effort to create a modern educational curriculum at Stanford University, Prober et al. recommended building a framework of core knowledge, embedding it into rich interactive formats, and encouraging students to pursue knowledge in some deep, but not all, domains [63]. The Blacksmith approach facilitates personal and in-depth pursuits that help transfer students into lifelong learners and adapt to challenging situations [63, 64]. This statement is consistent with Wald and Monteverde (2021). They report that educational resilience is vital in supporting and sustaining healthcare professional identity development and facilitating the progress of students’ moral resilience and complexity, especially in times of pandemic [25].\nAccording to the participants’ viewpoints, the applicability of the blacksmith approach is one of the most important aspects of improving the quality of anatomy teaching and learning in any situation, such as current critical conditions.\nOur university’s online programs were adequately integrated into the curriculum design and used to guide students on a learning journey rather than merely as another resource; perhaps, these tools could be beneficial. However, in such circumstances, the epidemic and sanctions of Iran and the lack of resources, students and professors should take measures to optimize education to facilitate the teaching and learning of this course. Accordingly, from the above point, through a qualitative study, it was assumed that there were signs of successful teaching and learning and internal change in deep search among students and faculty members.\nRevolution in medical education is a global program, and many medical schools have perceived investigation and experienced challenges [63, 64]. Correspondingly, an active and integrated strategy for teaching and learning anatomy has been emphasized in other medical education courses and areas, including a contemporary medical curriculum. Managers, practitioners, and professors can use the elements of this strategy to increase the efficiency of medical education, improve students’ knowledge, skills, and attitudes, and ultimately maintain and promote understanding of anatomy and other courses. Also, from the participant’s perspective, the applicability of students learning experiences and faculty members’ teaching experiences in the context and workplace conditions was one of the factors that changed the view of anatomy as an introductory course in medicine.", "must be built for the students. This step is in line with the first stage of the four S model, i.e., Situation awareness. Here, the students become aware of what is happening. The teachers here have two roles. First, they should deliver the following messages to the students: (1) you are supposed to actualize a new identity (role change), (2) anatomy (as a pillar of medicine) provides a problematic task ahead (stress), and (3) you can help yourself more than anyone else. (control) Furthermore, the students should try self-study to increase their self-confidence (trigger). Secondly, the teachers should acquire a mindful clinical standpoint, i.e., developing good clinical knowledge and adopting active learning methods based on teamwork and clinical scenarios that encourage active participation and self-directed learning as if they are future doctors. This strategy has been shown to reduce student stress [11, 13] and help them react better as future physicians [3, 8, 48].", "The students’ hearts must be warmed up through support and role modeling. This step is compatible with the following two stages in the 4 S model, i.e., self and support. The students evaluate their demographic and psychological characteristics, and then they need to be aware of support systems and be able to get help [27, 29]. Here again, the role of anatomy teachers is critical. As medical students see anatomy as a gate to medicine, the anatomy teacher can be known as the gatekeeper. They will inevitably actualize their teachers as wise role models who are righteous supporters. The anatomy faculty member should be aware of their role modeling. Role modeling can help students foster resilience for future change, which leads to professionalism and integrity [7, 18, 49–52]. The teachers should adopt ice-breaking methods, use their sense of humor, tell stories, design games, and have friendly chats with students outside the classroom. In addition, anatomy teachers should gain the skills and knowledge for counseling and get familiar with support strategies to become prominent supporters of students [53–56].", "We found that, currently, preclinical students are facing two overwhelming stressors. On the one hand, the covid crisis has detached the students from their teachers and resources [15, 18, 57]. On the other hand, students need to handle their transition into knowledgeable medical professionals. Therefore we need to incorporate teaching methods that assist our detached students in coping with their stressors. The last stage in the 4 S model also deals with coping strategies. Students may use several coping strategies like information seeking, selective ignoring, and reframing the challenge [58].\nAccordingly, the third step in our model includes three components; thorough engagement, reframing the situation, and selective information seeking. Regarding the first component, we encourage meticulously choosing more active and engaging learning techniques. Problem-based learning and Peer-based discussions in small groups, drawing, mind mapping, body painting, using play-doh, and interactive video content, are advised. According to Pandey and Zimitat (2007), a practical approach to anatomy education positively correlates with teaching and reflective learning quality. Successful anatomy learning requires balancing memorization with understanding and visualization[59]. Insightful experiences in teaching anatomy with this strategy are found in Australia (head and neck course) [15] and India (musculoskeletal anatomy) [13]. As Goodman et al. (2006) noted in their previous book, those with higher self-esteem and mental mastery are better able to cope with challenges. [60] A successful learning experience gives students a sense of control over their course, while applying a study guide, reference autonomy, and support system can help them gain self-esteem [61–63].\nReframing the situation, As the second component, is where the students need to redefine their roles and activities. Two sets of strategies can help students to reframe. 1) Using problem-based and peer-assisted learning with a flavor of sociocultural contexts can prepare students for better learning in a positive, non-intimidating learning environment to redefine themselves as future doctors for diagnosing and curing patients. Socio-emotional context and bringing drama elements into case scenarios have been recently emphasized to induce resilience and professionalism in medical students (2.14, 64).\nRecent evidence indicates that gamification leads to positive learning outcomes, higher motivation, and engagement. However, several downsides should also be considered, including increased competition and task evaluation difficulties. Besides, it depends highly on the context in which the gamification is being implemented and its users [65–68]. Hamari et al. (2014) have reviewed empirical literature, showing that gamification has proven effective in many fields, most notably education. Studies in education and learning contexts illustrate how the gamification of learning outcomes has been positive by increasing motivation and engagement in learning and enjoyment tasks over time. However, the studies also highlighted some negative points that require attention, such as the effects of increased competition, task evaluation difficulties, and design features. The review indicates that gamification provides positive effects; however, the effects greatly depend on the context in which the gamification is being implemented and the users who use it [66]. Recent studies suggest that joy and humor facilitate a person’s transition into more socially severe roles (like becoming a doctor) [34]. Hence, teachers should use joyful techniques such as painting, ice braking, storytelling, gamification, dance, and music [4, 9, 15, 53].\nThe last component deals with selective information seeking. Students should have a study guide and be free to choose their desirable resources (anatomy reference books, digital resources, or web-based social media videos). We advise the medical school to design a dynamic curriculum encompassing clear goals. In this setting, the study guide can act as a tool for transferring core knowledge in anatomical sciences. As a result, the tedious aspect of bulky content in anatomy courses can be avoided, and the educational efficiency of synchronous and asynchronous learning can be increased in the pandemic era [55, 56, 61, 69, 70].\nThis research is in line with constructive learning theory that emphasizes student-centered learning, which states that students should be actively involved in education and contribute to knowledge creation [71]. In a special effort to create a modern educational curriculum at Stanford University, Prober et al. recommended building a framework of core knowledge, embedding it into rich interactive formats, and encouraging students to pursue knowledge in some deep, but not all, domains [63]. The Blacksmith approach facilitates personal and in-depth pursuits that help transfer students into lifelong learners and adapt to challenging situations [63, 64]. This statement is consistent with Wald and Monteverde (2021). They report that educational resilience is vital in supporting and sustaining healthcare professional identity development and facilitating the progress of students’ moral resilience and complexity, especially in times of pandemic [25].\nAccording to the participants’ viewpoints, the applicability of the blacksmith approach is one of the most important aspects of improving the quality of anatomy teaching and learning in any situation, such as current critical conditions.\nOur university’s online programs were adequately integrated into the curriculum design and used to guide students on a learning journey rather than merely as another resource; perhaps, these tools could be beneficial. However, in such circumstances, the epidemic and sanctions of Iran and the lack of resources, students and professors should take measures to optimize education to facilitate the teaching and learning of this course. Accordingly, from the above point, through a qualitative study, it was assumed that there were signs of successful teaching and learning and internal change in deep search among students and faculty members.\nRevolution in medical education is a global program, and many medical schools have perceived investigation and experienced challenges [63, 64]. Correspondingly, an active and integrated strategy for teaching and learning anatomy has been emphasized in other medical education courses and areas, including a contemporary medical curriculum. Managers, practitioners, and professors can use the elements of this strategy to increase the efficiency of medical education, improve students’ knowledge, skills, and attitudes, and ultimately maintain and promote understanding of anatomy and other courses. Also, from the participant’s perspective, the applicability of students learning experiences and faculty members’ teaching experiences in the context and workplace conditions was one of the factors that changed the view of anatomy as an introductory course in medicine.", "As this study was conducted in one country, it is suggested that more studies and insights be obtained from other participants to increase the generalizability of findings. Besides, further work should be done to provide more practical guidance in the educational system regarding the transition of medical students in teaching and learning anatomy in the current pandemic crisis.", "Medical students experience a fundamental evolution into responsible and lifelong learners qualified as professional doctors. Educational systems focus primarily on teaching and learning, while students’ transition can be facilitated by a three-step model called the Blacksmith Approach, based on the study results. Initially, the blacksmith (educational system) must prepare a new forge. It means anatomy teachers should redesign the academic mentality of students by delivering messages about role change, difficulties ahead, and the importance of self-guidance, along with implementing clinical scenarios in active learning methods. Then, he/she must heat the metal. It implies that anatomy teachers, as gatekeepers of medicine, should adopt amusing teaching methods while embracing their role modeling and actively applying support strategies. Finally, he/she may shape the metal with a sledgehammer. Teachers need to use three components to create a sword (self-confident, accountable, resilient professional); thorough engagement by using active and engaging teaching techniques, reframing the situation with problem-based clinical scenarios, and joyful teaching. And selective information seeking via study guide and reference autonomy.\nIn conclusion, an active and integrated strategy for teaching and learning anatomy is suggested to be applied in other courses and areas of medical education in the current medical curriculum.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ "introduction", null, "materials|methods", null, null, null, null, null, "results", null, null, null, null, null, null, null, null, null, null, null, null, null, null, "discussion", null, null, null, null, "conclusion", "supplementary-material", null ]
[ "Anatomy education", "Teaching", "Learning", "Medical faculty", "Medical students", "Qualitative" ]
Prevalence and factors associated with postpartum pelvic girdle pain among women in Poland: a prospective, observational study.
36266709
Pelvic girdle pain (PGP) is a type of pregnancy-related lumbopelvic pain. This study aimed to examine the prevalence, severity, and factors associated with postpartum PGP in a selected group of postpartum women in Poland.
BACKGROUND
This was a prospective, observational study. In phase 1, 411 women were recruited 24-72 h postpartum. The prevalence of PGP was assessed by a physiotherapist using a series of dedicated tests. Pelvic floor muscle function and presence of diastasis recti were assessed via palpation examination. Age, education, parity, mode of delivery, infant body mass, body mass gain during pregnancy, the use of anesthesia during delivery and were recorded. In a phase 2, 6 weeks postpartum, the prevalence of PGP and its severity were assessed via a self-report.
METHODS
In phase 1 (shortly postpartum), PGP was diagnosed in 9% (n = 37) of women. In phase 2 (6 weeks postpartum), PGP was reported by 15.70% of women (n = 42). The univariable analyses showed a higher likelihood of PGP shortly postpartum in women who declared PGP during pregnancy (OR 14.67, 95% CI 4.43-48.61) and among women with abdominal midline doming (OR 2.05, 95% CI 1.04-4.06). The multivariable regression analysis showed significant associations in women with increased age (OR 1.12, 95% CI 1.01-1.21) and declaring PGP during pregnancy (OR 14.83, 95% CI 4.34-48.72).
RESULTS
Although the prevalence of postpartum PGP among women in Poland is lower than reported in other countries, it is experienced by almost every tenth women shortly postpartum and every sixth can report similar symptoms 6 weeks later. Age, PGP during pregnancy and abdominal midline doming were associated with experiencing PGP shortly postpartum.
CONCLUSION
[ "Humans", "Pregnancy", "Female", "Pelvic Girdle Pain", "Prevalence", "Prospective Studies", "Poland", "Postpartum Period", "Pregnancy Complications" ]
9585777
null
null
null
null
Results
A total of 415 women were invited to participate, 4 of whom did not consent to join the study. Thus, 411 women were included in phase 1 (shortly postpartum). Table 1. presents the characteristics of the participants. In phase 2 (“follow-up”), 268 women replied to the text message (65.2% of the initial group). No statistically significant differences were found between those who responded to the message and those who did not in terms of variables assessed shortly postpartum (in phase 1): BMI, age, presence of PGP postpartum, parity, DRA severity, and ability to activate pelvic floor muscles. Table 1Characteristics of the study group, n = 411All participantsn = 411With PGP n = 37Without PGP n = 374 Age 31.17 ± 4.0132.35 ± 4.3331.06 ± 3.96Height [cm],167.46 ± 5.69167.00 ± 6.64167.51 ± 5.60 BMI before pregnancy 22.14 ± 3.5722.46 ± 3.2022.10 ± 3.60 Body mass gain in pregnancy [kg] 14.30 ± 4.9414.92 ± 4.2814.24 ± 4.99Education, n (%) †vocational education2 (0.5)11secondary education43 (10.5)242university education366 (89)35331 Parity, n (%) 1.72 ± 0.961.95 ± 1.101.70 ± 0.941209 (50.80)2143 (34.80)337 (9.00)412 (3.00)58 (2.00)61 (0.20)71 (0.20) Mode of the last delivery, n (%) vaginal387 (94.20)37 (100)350 (93.60)cesarean ‡18 (4.30)018 (4.80)vacuum extractor ‡6 (1.50)06 (1.60) Perineal injury during recent delivery, n (%) none162 (39.50)18 (48.60)144 (38.50)1st grade138 (33.50)9 (24.30)129 (34.50)episiotomy111 (27)10 (27)101 (27) Anesthesia during last delivery, n (%) none238 (57.91)24 (64.86)214 (57.22)epidural154 (37.47)12 (32.43)142 (37.97)spinal19 (4.62)1 (2.70)18 (4.81) Infant body mass ≥ 4000 g yes336 (81.75)6 (16.22)330 (88.24)no75 (18.25)31 (83.78)44 (11.76)† Participants with vocational education were excluded (from this calculations) because of the low number of records; ‡ no statistical analysis due to low number of observations; χ² Chi2 tet value; U Manna Whitney test value Characteristics of the study group, n = 411 † Participants with vocational education were excluded (from this calculations) because of the low number of records; ‡ no statistical analysis due to low number of observations; χ² Chi2 tet value; U Manna Whitney test value Among the patients in phase 1 of the study (shortly postpartum), 47.9% (n = 197) reported PGP symptoms during pregnancy. In phase 1, PGP was diagnosed in 9.0% (n = 37) of women at the early postpartum stage. In phase 2 (6 weeks postpartum), PGP was reported by 15.7% of women (n = 42). Table 2 presents the prevalence and type of PGP across different time points, and Table 3 shows the severity of pain and functional disturbances in both phases of the study. Table 2Prevalence and types of PGP across the time points.TimepointDuring pregnancyEarly postpartum6 weeks postpartum Assessment form self-reported, retrospective clinical assessment self-reported N % N % N % PGP 197/41147.9337/411942/26815.70 PGP type Posterior Pelvic Pain 86/19743.709/3724.3016/4238.10 Unilateral pain 17/1978.602/375.402/424.80 Symphyseal pain 44/19722.309/3724.3012/4228.60 Pelvic Girdle Syndrome 47/19723.9016/3743.2011/4226.20 Unilateral pain + symphyseal pain 3/1971.501/372.701/422.40 Prevalence and types of PGP across the time points. PGP type Table 3The severity of pain and functional limitations at phase 1 and 2 of the studyTimepointEarlyPostpartum6 weekspostpartum Mean (SD) (min-max) N Mean (SD) (min-max) N NRS 5.34 (1.74)(3–9)37/374.63 (2.04)(1–8)24/42 PGQ [%] 48.87 (16.40)(23.61–82.61)32/3725.80 (14.90)(6.67–56.94)13/42The values of the PGQ questionnaire from the early postpartum stage were missing in five cases – they were left blank or only partially completed. Six weeks after delivery, the pain intensity value on the NRS was reported by 24 women, and the PGQ questionnaire was completed only by 13 The severity of pain and functional limitations at phase 1 and 2 of the study 5.34 (1.74) (3–9) 4.63 (2.04) (1–8) 48.87 (16.40) (23.61–82.61) 25.80 (14.90) (6.67–56.94) The values of the PGQ questionnaire from the early postpartum stage were missing in five cases – they were left blank or only partially completed. Six weeks after delivery, the pain intensity value on the NRS was reported by 24 women, and the PGQ questionnaire was completed only by 13 The univariable analyses showed a higher likelihood of PGP shortly postpartum in women who declared PGP during pregnancy and among women with doming at the abdominal midline in the projection of linea alba (Table 4.). Table 4Univariable analysis of factors associated with pelvic girdle pain (PGP) shortly postpartum (phase 1)PGP (+)n = 37PGP (-)N = 374OR (95% CI)p-value PGP during pregnancy (yes) n (%) 34 (91.90)163 (43.60)14.67 (4.43–48.61) < 0.01 Age mean (SD) 32.35(4.33)31.06 (3.96)1.08 (0.99–1.18)0.06 Body mass gain during pregnancy mean (SD) 32.68 (4.60)31.59 (4.37)1.03 (0.96–1.10)0.42 BMI before pregnancy mean (SD) 22.46 (3.20)22.1 (3.60)1.03 (0.94–1.12)0.56 Number of previous deliveries mean (SD) 1.95 (1.10)1.7 (0.94)1.27 (0.93–1.71)0.14 Urinary incontinence during pregnancy or before (yes) n (%) 15 (40.50)205 (54.80)0.56 (0.28–1.12)0.10 Abdominal midline doming (yes), n (%) 20 (54.10)134 (36.40)2.05 (1.04–4.06) 0.04 DR severity n (%) none, IRD < 2 9 (24.30)149 (40.50)1.27 (0.95–1.69)0.11mild, IRD 2;< 3 8 (21.60)72 (19.60) moderate, IRD 3;<4 12 (32.40)75 (20.40) severe, IRD > 4 8 (21.60)72 (19.60) Oxford scale Mean (SD) 2.27(0.65)2.28 (0.90)0.99 (0.67–1.45)0.96 Reissing scale mean (SD) 0.32 (1.11)-0.53 (0.96)1.25 (0.88–1.77)0.21 Correct activation of pelvic floor (yes), n (%) 13 (35.10)134 (35.80)1.36 (0.69–2.70)0.37 Univariable analysis of factors associated with pelvic girdle pain (PGP) shortly postpartum (phase 1) PGP during pregnancy (yes) n (%) Age mean (SD) Body mass gain during pregnancy mean (SD) BMI before pregnancy mean (SD) Urinary incontinence during pregnancy or before (yes) n (%) DR severity n (%) mild, IRD 2;< 3 Oxford scale Mean (SD) Reissing scale mean (SD) Based on the obtained prevalence of postpartum PGP, we could include up to 4 variables into the multivariable model [38]. The final model, in which we obtained statistically significant results for all included items, consisted of 3 variables. This multivariable regression analysis showed that the odds of having PGP shortly postpartum were higher in women with increased age (10% higher likelihood with every year of age), declaring PGP during pregnancy, and with higher values of Reissing scale. To enhance the interpretability, we then analyzed the Reissing scale in the following categories: hypotonus (range − 3 to -1) and increased muscle tone (range 1 to 3), with the reference value being normotonus (0). However, after this procedure, the Reissing scale became not statistically significant (Table 5). The calculated Nagelkerke R-square was 0.2. Table 5Multivariable analysis of factors associated with pelvic girdle pain (PGP) shortly postpartum (phase 1)OR (95% CI)p-value Presence of PGP during pregnancy (yes) 14.83 (4.340-48.721) < 0.0001 Age 1.12 (1.009–1.214) 0.032 Reissing scale Reissing scale – not grouped 1.43 (1.003–2.046) 0.048 Reissing scale when grouped normal tone (0) referencereference decreased tone (range − 3 to -1) 0.53 (0.227–1.220)0.134 increased tone (range 1 to 3) 0.37 (0.594–4.078)0.368 Multivariable analysis of factors associated with pelvic girdle pain (PGP) shortly postpartum (phase 1) decreased tone (range − 3 to -1) increased tone (range 1 to 3)
Conclusion
The findings presented in this study suggest that every tenth Polish woman may experience PGP during the first days postpartum and every sixth can report similar symptoms 6 weeks later. The pain intensity and functional limitations tend to subside over time: from moderate pain intensity and functional limitation shortly postpartum to mild/low 6 weeks later. Nevertheless, postpartum PGP should not be ignored, especially in the context of the observed continued increase in chronic pain syndromes and their associated consequences. Older age, PGP during pregnancy, and doming of the abdominal midline at the level of linea alba were associated with the experience of PGP within the first days postpartum. Our study showed no association between pelvic floor function and PGP shortly postpartum. However, this may be due to the chosen methodology (assessment shortly postpartum).
[ "Background", "Methods", "Setting", "Participants", "Procedure", "Measures/Variables", "PGP prevalence and severity", "Pelvic floor measurements", "DRA measurements", "Statistical analysis", "Strengths and limitations", "Implications" ]
[ "Symptoms of pelvic girdle pain (PGP) are commonly reported to healthcare providers by pregnant or postpartum women. This musculoskeletal disorder is a form of low back pain experienced between the posterior iliac crests and the lower edge of the gluteal folds, most commonly in the vicinity of the sacroiliac joints. PGP also includes pain in the pubic symphysis, occurring in isolation or in conjunction with other pelvic joints. Patients with PGP have reduced tolerance to standing, walking, sitting, and changing positions [1]. Pregnancy-related PGP may appear as early as the first trimester of pregnancy or can be delayed up to 3 weeks postpartum [2]. PGP is associated with significantly more pain and functional limitations than lower back pain [3]. The pain often subsides after delivery, but some women continue to have persistent symptoms postpartum. Among women reporting PGP during pregnancy, 1 in 10 will suffer from it up to 11 years later [4]. This significantly impacts their quality of life [5]. In Poland, the prevalence of PGP during pregnancy was reported by 42% of women [6]. To our knowledge, there are no studies reporting on the prevalence of postpartum PGP in Poland or any other country in the central-eastern region of Europe.\nAssessment of muscle impairments has been recommended in Clinical Practice Guidelines for PGP in the postpartum population [7]. To date, several studies investigated musculoskeletal factors that could be associated with pregnancy-related PGP, including the function of pelvic floor muscles and diastasis rectus abdominis (DRA). Coordination between lumbopelvic and abdominal muscles and fascia was suggested to play a significant role in postural stabilization [8]. It was hypothesized that insufficient motor control could give rise to pain from impaired load transfer throughout the pelvic girdle [9], and that pelvic floor and abdominal muscles play an important role in the stabilization and motor control of the pelvis [8, 10]. However, there is still little evidence to support these associations. While some studies confirm the relationship between the DRA, linea alba dysfunction, and postpartum PGP [11, 12], others do not [13–15]. A recent systematic review concluded that DRA presence might be associated with decreased abdominal muscle strength and severity of low back pain and suggested further studies rigorously assessing this association [16]. A literature review investigating the relationship between perineal characteristics and PGP suggested that overactivity and increased tension of pelvic floor muscles are more common in women with PGP [17].\nThis study aimed to assess the prevalence and severity of PGP among women in Poland early postpartum and 6 weeks after delivery. Additionally, we aimed to identify factors associated with early postpartum PGP, including also DRA and pelvic floor function.", "This was a prospective, observational study. Ethics approval was received from the Bioethics Committee of the Medical University of Warsaw (KB/136/2017) and the study was registered at https://www.anzctr.org.au/ under the number ACTRN 12,618,000,764,235. Data was collected between 1.12.2017 and 12.03.2020. All participants provided written informed consent prior to commencing any of the study procedures. The study was supported by the Department of Midwifery at the Centre of Postgraduate Medical Education Research Program for 2020. The STROBE checklist was followed to ensure proper reporting of this study [18].\n[SUBTITLE] Setting [SUBSECTION] St. Sophia Specialist Hospital in Warsaw, Poland served as a recruitment site for this study. It is a tertiary, publicly funded hospital with over 6500 births annually. A free consultation with a pelvic health physiotherapist is part of standard care for every woman after delivery at this hospital. The participants were recruited from among the women attending the consultation.\nSt. Sophia Specialist Hospital in Warsaw, Poland served as a recruitment site for this study. It is a tertiary, publicly funded hospital with over 6500 births annually. A free consultation with a pelvic health physiotherapist is part of standard care for every woman after delivery at this hospital. The participants were recruited from among the women attending the consultation.\n[SUBTITLE] Participants [SUBSECTION] Women between 18 and 45 years old who attended free physiotherapy consultation 24–72 h postpartum were invited to participate in the study. The main exclusion criteria were additional comorbidities potentially causing PGP-like symptoms (rheumatoid arthritis, ankylosing spondylitis, Scheuermann disease, Ehlers-Danlos syndrome, spinal surgeries, nerve root compression, spondylolisthesis), contraindications for pelvic floor examination (puerperal genital hematoma, diffuse perineal edema, perineal wound dehiscence, bladder catheterization) and severe postpartum complications (internal bleeding, femoral artery embolism, pelvic fracture). To limit the sampling bias and ensure that patients were randomly included in the study, every third participant of the postpartum physiotherapy consultation was invited, and recruitment for the study took place every third day.\nWomen between 18 and 45 years old who attended free physiotherapy consultation 24–72 h postpartum were invited to participate in the study. The main exclusion criteria were additional comorbidities potentially causing PGP-like symptoms (rheumatoid arthritis, ankylosing spondylitis, Scheuermann disease, Ehlers-Danlos syndrome, spinal surgeries, nerve root compression, spondylolisthesis), contraindications for pelvic floor examination (puerperal genital hematoma, diffuse perineal edema, perineal wound dehiscence, bladder catheterization) and severe postpartum complications (internal bleeding, femoral artery embolism, pelvic fracture). To limit the sampling bias and ensure that patients were randomly included in the study, every third participant of the postpartum physiotherapy consultation was invited, and recruitment for the study took place every third day.\n[SUBTITLE] Procedure [SUBSECTION] The study was conducted in two phases. Phase 1 was carried out at the hospital. Participants who met the inclusion criteria and gave informed consent were included in the study. Age, education, parity (defined as previous deliveries > 24 weeks gestation), delivery type (vaginal, forceps/vacuum extractor, cesarean), infant body mass (< 4000 g/4000 g or more), height, body mass gain during pregnancy, the use of anesthesia during delivery were recorded from the patient medical record and confirmed via self-report. The women were asked if they had experienced PGP during the last pregnancy (yes/no and if yes - in what location) and urinary incontinence during or before the pregnancy (yes/no). The presence and severity of PGP were assessed by the physiotherapist. Additionally, each woman received an examination of the pelvic floor and abdominal muscles. All examinations were performed by the same registered pelvic health physiotherapist who had completed advanced training in urogynaecology and was certified by the Polish Urogynecological Society to digitally examine the pelvic floor muscles.\nIn phase 2 (“follow-up”), all women who participated in phase 1 were contacted via text message 6 weeks postpartum. In the message, they were asked if they experienced PGP. In case of no response within 48 h, another message was sent. No response to the second message meant a loss to follow-up.\nThe study was conducted in two phases. Phase 1 was carried out at the hospital. Participants who met the inclusion criteria and gave informed consent were included in the study. Age, education, parity (defined as previous deliveries > 24 weeks gestation), delivery type (vaginal, forceps/vacuum extractor, cesarean), infant body mass (< 4000 g/4000 g or more), height, body mass gain during pregnancy, the use of anesthesia during delivery were recorded from the patient medical record and confirmed via self-report. The women were asked if they had experienced PGP during the last pregnancy (yes/no and if yes - in what location) and urinary incontinence during or before the pregnancy (yes/no). The presence and severity of PGP were assessed by the physiotherapist. Additionally, each woman received an examination of the pelvic floor and abdominal muscles. All examinations were performed by the same registered pelvic health physiotherapist who had completed advanced training in urogynaecology and was certified by the Polish Urogynecological Society to digitally examine the pelvic floor muscles.\nIn phase 2 (“follow-up”), all women who participated in phase 1 were contacted via text message 6 weeks postpartum. In the message, they were asked if they experienced PGP. In case of no response within 48 h, another message was sent. No response to the second message meant a loss to follow-up.\n[SUBTITLE] Measures/Variables [SUBSECTION] [SUBTITLE] PGP prevalence and severity [SUBSECTION] During the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.\nDuring the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.\n[SUBTITLE] Pelvic floor measurements [SUBSECTION] A pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.\nA pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.\n[SUBTITLE] DRA measurements [SUBSECTION] The width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].\nThe width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].\n[SUBTITLE] PGP prevalence and severity [SUBSECTION] During the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.\nDuring the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.\n[SUBTITLE] Pelvic floor measurements [SUBSECTION] A pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.\nA pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.\n[SUBTITLE] DRA measurements [SUBSECTION] The width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].\nThe width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].\n[SUBTITLE] Statistical analysis [SUBSECTION] The sample size was determined based on a priori calculations, based on the reports of Wu et al., in which the incidence of postpartum low back pain and /or PGP is estimated at 25% [35]. The following formula was used:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$n=\\frac{{Z}^{2}P(1-P)}{{d}^{2}},$$\\end{document}\nwhere n is the sample size, the Z value (corresponding to the significance level p 0.05) is 1.96. P value is the expected occurrence of pain, and the d value is 0.05 (recommended value for an expected prevalence between 10 and 90% [36]). Based on this calculation, the minimum sample size was 288. A high drop-out rate was also expected due to the specificity of the postpartum period, as observed by other authors [37]. To secure the minimal number of participants for phase 2 of the study (“follow-up”), the sample size was increased to a minimum of 400.\nFor continuous variables, mean and SD were calculated. Categorical data are presented as numbers and percentages. The prevalence of PGP was calculated by dividing the number of women classified with PGP by the total number of women who participated in the study. To assess the possible differences between women who responded 6 weeks postpartum and those who were lost to follow-up, Student t-test, the Mann-Whitney U and the the χ2 test were used depending on the type and normality of data.\nUnivariable logistic regression analyses were fitted to test individual factors for association with PGP shortly postpartum (phase 1). Multivariable logistic models were used to identify associations with more than one factor included in the model. The selection of factors to include in the multivariable models was informed by the univariable results. Variables with p-values less than or equal to 0.1 were included in the multivariable regression model. The number of possible investigated factors in the multivariable model was calculated assuming 10 participants per potential associated factor with division by the obtained prevalence rate [38]. The best subset of explanatory variables was selected manually by excluding the variables with the smallest contribution to the model. The predictive power of the model was calculated by Nagelkerke R-square (R²). Missing data were not included in the analysis. Alpha was set at 0.05. Statistical analyses were performed using PQStat ver. 1.8.2.166.\nThe sample size was determined based on a priori calculations, based on the reports of Wu et al., in which the incidence of postpartum low back pain and /or PGP is estimated at 25% [35]. The following formula was used:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$n=\\frac{{Z}^{2}P(1-P)}{{d}^{2}},$$\\end{document}\nwhere n is the sample size, the Z value (corresponding to the significance level p 0.05) is 1.96. P value is the expected occurrence of pain, and the d value is 0.05 (recommended value for an expected prevalence between 10 and 90% [36]). Based on this calculation, the minimum sample size was 288. A high drop-out rate was also expected due to the specificity of the postpartum period, as observed by other authors [37]. To secure the minimal number of participants for phase 2 of the study (“follow-up”), the sample size was increased to a minimum of 400.\nFor continuous variables, mean and SD were calculated. Categorical data are presented as numbers and percentages. The prevalence of PGP was calculated by dividing the number of women classified with PGP by the total number of women who participated in the study. To assess the possible differences between women who responded 6 weeks postpartum and those who were lost to follow-up, Student t-test, the Mann-Whitney U and the the χ2 test were used depending on the type and normality of data.\nUnivariable logistic regression analyses were fitted to test individual factors for association with PGP shortly postpartum (phase 1). Multivariable logistic models were used to identify associations with more than one factor included in the model. The selection of factors to include in the multivariable models was informed by the univariable results. Variables with p-values less than or equal to 0.1 were included in the multivariable regression model. The number of possible investigated factors in the multivariable model was calculated assuming 10 participants per potential associated factor with division by the obtained prevalence rate [38]. The best subset of explanatory variables was selected manually by excluding the variables with the smallest contribution to the model. The predictive power of the model was calculated by Nagelkerke R-square (R²). Missing data were not included in the analysis. Alpha was set at 0.05. Statistical analyses were performed using PQStat ver. 1.8.2.166.", "St. Sophia Specialist Hospital in Warsaw, Poland served as a recruitment site for this study. It is a tertiary, publicly funded hospital with over 6500 births annually. A free consultation with a pelvic health physiotherapist is part of standard care for every woman after delivery at this hospital. The participants were recruited from among the women attending the consultation.", "Women between 18 and 45 years old who attended free physiotherapy consultation 24–72 h postpartum were invited to participate in the study. The main exclusion criteria were additional comorbidities potentially causing PGP-like symptoms (rheumatoid arthritis, ankylosing spondylitis, Scheuermann disease, Ehlers-Danlos syndrome, spinal surgeries, nerve root compression, spondylolisthesis), contraindications for pelvic floor examination (puerperal genital hematoma, diffuse perineal edema, perineal wound dehiscence, bladder catheterization) and severe postpartum complications (internal bleeding, femoral artery embolism, pelvic fracture). To limit the sampling bias and ensure that patients were randomly included in the study, every third participant of the postpartum physiotherapy consultation was invited, and recruitment for the study took place every third day.", "The study was conducted in two phases. Phase 1 was carried out at the hospital. Participants who met the inclusion criteria and gave informed consent were included in the study. Age, education, parity (defined as previous deliveries > 24 weeks gestation), delivery type (vaginal, forceps/vacuum extractor, cesarean), infant body mass (< 4000 g/4000 g or more), height, body mass gain during pregnancy, the use of anesthesia during delivery were recorded from the patient medical record and confirmed via self-report. The women were asked if they had experienced PGP during the last pregnancy (yes/no and if yes - in what location) and urinary incontinence during or before the pregnancy (yes/no). The presence and severity of PGP were assessed by the physiotherapist. Additionally, each woman received an examination of the pelvic floor and abdominal muscles. All examinations were performed by the same registered pelvic health physiotherapist who had completed advanced training in urogynaecology and was certified by the Polish Urogynecological Society to digitally examine the pelvic floor muscles.\nIn phase 2 (“follow-up”), all women who participated in phase 1 were contacted via text message 6 weeks postpartum. In the message, they were asked if they experienced PGP. In case of no response within 48 h, another message was sent. No response to the second message meant a loss to follow-up.", "[SUBTITLE] PGP prevalence and severity [SUBSECTION] During the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.\nDuring the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.\n[SUBTITLE] Pelvic floor measurements [SUBSECTION] A pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.\nA pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.\n[SUBTITLE] DRA measurements [SUBSECTION] The width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].\nThe width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].", "During the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.", "A pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.", "The width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].", "The sample size was determined based on a priori calculations, based on the reports of Wu et al., in which the incidence of postpartum low back pain and /or PGP is estimated at 25% [35]. The following formula was used:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$n=\\frac{{Z}^{2}P(1-P)}{{d}^{2}},$$\\end{document}\nwhere n is the sample size, the Z value (corresponding to the significance level p 0.05) is 1.96. P value is the expected occurrence of pain, and the d value is 0.05 (recommended value for an expected prevalence between 10 and 90% [36]). Based on this calculation, the minimum sample size was 288. A high drop-out rate was also expected due to the specificity of the postpartum period, as observed by other authors [37]. To secure the minimal number of participants for phase 2 of the study (“follow-up”), the sample size was increased to a minimum of 400.\nFor continuous variables, mean and SD were calculated. Categorical data are presented as numbers and percentages. The prevalence of PGP was calculated by dividing the number of women classified with PGP by the total number of women who participated in the study. To assess the possible differences between women who responded 6 weeks postpartum and those who were lost to follow-up, Student t-test, the Mann-Whitney U and the the χ2 test were used depending on the type and normality of data.\nUnivariable logistic regression analyses were fitted to test individual factors for association with PGP shortly postpartum (phase 1). Multivariable logistic models were used to identify associations with more than one factor included in the model. The selection of factors to include in the multivariable models was informed by the univariable results. Variables with p-values less than or equal to 0.1 were included in the multivariable regression model. The number of possible investigated factors in the multivariable model was calculated assuming 10 participants per potential associated factor with division by the obtained prevalence rate [38]. The best subset of explanatory variables was selected manually by excluding the variables with the smallest contribution to the model. The predictive power of the model was calculated by Nagelkerke R-square (R²). Missing data were not included in the analysis. Alpha was set at 0.05. Statistical analyses were performed using PQStat ver. 1.8.2.166.", "This was the first large-scale study conducted in Poland using the recommended guidelines for classifying and investigating postpartum PGP prevalence with the use of screening palpation examination of the pelvic floor and abdominal muscles. To our knowledge, this is also the first study in the central-eastern region in Europe. Considering that this region is inhabited mainly by Caucasian women with a similar physiognomy, our results may estimate postpartum PGP prevalence in this part of Europe.\nThe main limitation of this work is the high drop-out rate in phase 2 (“follow-up”). For this reason, the prevalence of PGP 6 weeks postpartum may be underestimated. Time constraints, lack of trust, and low awareness of clinical trials are the main barriers to participation in research projects [56]. Additionally, the first weeks after delivery are challenging for many women, and research obligations may not be their priority. High drop-out rates were also reported by another study investigating PGP 6 weeks postpartum via SMS where the response rate of 43% was recorded 6 weeks postpartum [37]. Another limitation could be caused by the assessment of PGP 6 weeks via self-reports. However, this method was used in previous PGP research [37, 42] and a study by Rejano-Campo et al. [57] showed that self-reported PGP was verified by specific clinical tests in nearly all cases.\nFinally, we cannot exclude potential selection bias. Although we have made an effort to adopt random recruitment for phase 1 (shortly postpartum), we have included mainly highly educated women from only one center located in the capital city of Poland. This should be taken into account while inferring results from our sample to the general population.", "Obtained results with regard to other recently published matched case-control studies suggest that the relationship between PGP and DRA-related factors is multidimensional and not straightforward as previously suggested. Assessment of DRA-related factors seems not to be a crucial part of the screening for postpartum PGP but may be of greater importance when assessing individuals with postpartum PGP. Future research should further investigate the possible, multidimensional interactions between PGP and the whole abdominal wall complex (not restricted to only IRD as recommended by Delphi Consensus Study for the conservative management of pregnancy-related DRA [58]), and whether the DRA-related dysfunctions “only” co-exist with PGP or play a role in it. In that case, future studies focusing on creating adequate tension through the abdominal wall during PGP rehabilitation may be feasible. It should all be adjusted for psychosocial factors, which weren’t taken into account in our study. However, they are related to central pain mechanisms observed in individuals with persistent postpartum PGP and could be important factors filling the gaps in our current understanding of postpartum PGP [7, 59]." ]
[ null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Setting", "Participants", "Procedure", "Measures/Variables", "PGP prevalence and severity", "Pelvic floor measurements", "DRA measurements", "Statistical analysis", "Results", "Discussion", "Strengths and limitations", "Implications", "Conclusion" ]
[ "Symptoms of pelvic girdle pain (PGP) are commonly reported to healthcare providers by pregnant or postpartum women. This musculoskeletal disorder is a form of low back pain experienced between the posterior iliac crests and the lower edge of the gluteal folds, most commonly in the vicinity of the sacroiliac joints. PGP also includes pain in the pubic symphysis, occurring in isolation or in conjunction with other pelvic joints. Patients with PGP have reduced tolerance to standing, walking, sitting, and changing positions [1]. Pregnancy-related PGP may appear as early as the first trimester of pregnancy or can be delayed up to 3 weeks postpartum [2]. PGP is associated with significantly more pain and functional limitations than lower back pain [3]. The pain often subsides after delivery, but some women continue to have persistent symptoms postpartum. Among women reporting PGP during pregnancy, 1 in 10 will suffer from it up to 11 years later [4]. This significantly impacts their quality of life [5]. In Poland, the prevalence of PGP during pregnancy was reported by 42% of women [6]. To our knowledge, there are no studies reporting on the prevalence of postpartum PGP in Poland or any other country in the central-eastern region of Europe.\nAssessment of muscle impairments has been recommended in Clinical Practice Guidelines for PGP in the postpartum population [7]. To date, several studies investigated musculoskeletal factors that could be associated with pregnancy-related PGP, including the function of pelvic floor muscles and diastasis rectus abdominis (DRA). Coordination between lumbopelvic and abdominal muscles and fascia was suggested to play a significant role in postural stabilization [8]. It was hypothesized that insufficient motor control could give rise to pain from impaired load transfer throughout the pelvic girdle [9], and that pelvic floor and abdominal muscles play an important role in the stabilization and motor control of the pelvis [8, 10]. However, there is still little evidence to support these associations. While some studies confirm the relationship between the DRA, linea alba dysfunction, and postpartum PGP [11, 12], others do not [13–15]. A recent systematic review concluded that DRA presence might be associated with decreased abdominal muscle strength and severity of low back pain and suggested further studies rigorously assessing this association [16]. A literature review investigating the relationship between perineal characteristics and PGP suggested that overactivity and increased tension of pelvic floor muscles are more common in women with PGP [17].\nThis study aimed to assess the prevalence and severity of PGP among women in Poland early postpartum and 6 weeks after delivery. Additionally, we aimed to identify factors associated with early postpartum PGP, including also DRA and pelvic floor function.", "This was a prospective, observational study. Ethics approval was received from the Bioethics Committee of the Medical University of Warsaw (KB/136/2017) and the study was registered at https://www.anzctr.org.au/ under the number ACTRN 12,618,000,764,235. Data was collected between 1.12.2017 and 12.03.2020. All participants provided written informed consent prior to commencing any of the study procedures. The study was supported by the Department of Midwifery at the Centre of Postgraduate Medical Education Research Program for 2020. The STROBE checklist was followed to ensure proper reporting of this study [18].\n[SUBTITLE] Setting [SUBSECTION] St. Sophia Specialist Hospital in Warsaw, Poland served as a recruitment site for this study. It is a tertiary, publicly funded hospital with over 6500 births annually. A free consultation with a pelvic health physiotherapist is part of standard care for every woman after delivery at this hospital. The participants were recruited from among the women attending the consultation.\nSt. Sophia Specialist Hospital in Warsaw, Poland served as a recruitment site for this study. It is a tertiary, publicly funded hospital with over 6500 births annually. A free consultation with a pelvic health physiotherapist is part of standard care for every woman after delivery at this hospital. The participants were recruited from among the women attending the consultation.\n[SUBTITLE] Participants [SUBSECTION] Women between 18 and 45 years old who attended free physiotherapy consultation 24–72 h postpartum were invited to participate in the study. The main exclusion criteria were additional comorbidities potentially causing PGP-like symptoms (rheumatoid arthritis, ankylosing spondylitis, Scheuermann disease, Ehlers-Danlos syndrome, spinal surgeries, nerve root compression, spondylolisthesis), contraindications for pelvic floor examination (puerperal genital hematoma, diffuse perineal edema, perineal wound dehiscence, bladder catheterization) and severe postpartum complications (internal bleeding, femoral artery embolism, pelvic fracture). To limit the sampling bias and ensure that patients were randomly included in the study, every third participant of the postpartum physiotherapy consultation was invited, and recruitment for the study took place every third day.\nWomen between 18 and 45 years old who attended free physiotherapy consultation 24–72 h postpartum were invited to participate in the study. The main exclusion criteria were additional comorbidities potentially causing PGP-like symptoms (rheumatoid arthritis, ankylosing spondylitis, Scheuermann disease, Ehlers-Danlos syndrome, spinal surgeries, nerve root compression, spondylolisthesis), contraindications for pelvic floor examination (puerperal genital hematoma, diffuse perineal edema, perineal wound dehiscence, bladder catheterization) and severe postpartum complications (internal bleeding, femoral artery embolism, pelvic fracture). To limit the sampling bias and ensure that patients were randomly included in the study, every third participant of the postpartum physiotherapy consultation was invited, and recruitment for the study took place every third day.\n[SUBTITLE] Procedure [SUBSECTION] The study was conducted in two phases. Phase 1 was carried out at the hospital. Participants who met the inclusion criteria and gave informed consent were included in the study. Age, education, parity (defined as previous deliveries > 24 weeks gestation), delivery type (vaginal, forceps/vacuum extractor, cesarean), infant body mass (< 4000 g/4000 g or more), height, body mass gain during pregnancy, the use of anesthesia during delivery were recorded from the patient medical record and confirmed via self-report. The women were asked if they had experienced PGP during the last pregnancy (yes/no and if yes - in what location) and urinary incontinence during or before the pregnancy (yes/no). The presence and severity of PGP were assessed by the physiotherapist. Additionally, each woman received an examination of the pelvic floor and abdominal muscles. All examinations were performed by the same registered pelvic health physiotherapist who had completed advanced training in urogynaecology and was certified by the Polish Urogynecological Society to digitally examine the pelvic floor muscles.\nIn phase 2 (“follow-up”), all women who participated in phase 1 were contacted via text message 6 weeks postpartum. In the message, they were asked if they experienced PGP. In case of no response within 48 h, another message was sent. No response to the second message meant a loss to follow-up.\nThe study was conducted in two phases. Phase 1 was carried out at the hospital. Participants who met the inclusion criteria and gave informed consent were included in the study. Age, education, parity (defined as previous deliveries > 24 weeks gestation), delivery type (vaginal, forceps/vacuum extractor, cesarean), infant body mass (< 4000 g/4000 g or more), height, body mass gain during pregnancy, the use of anesthesia during delivery were recorded from the patient medical record and confirmed via self-report. The women were asked if they had experienced PGP during the last pregnancy (yes/no and if yes - in what location) and urinary incontinence during or before the pregnancy (yes/no). The presence and severity of PGP were assessed by the physiotherapist. Additionally, each woman received an examination of the pelvic floor and abdominal muscles. All examinations were performed by the same registered pelvic health physiotherapist who had completed advanced training in urogynaecology and was certified by the Polish Urogynecological Society to digitally examine the pelvic floor muscles.\nIn phase 2 (“follow-up”), all women who participated in phase 1 were contacted via text message 6 weeks postpartum. In the message, they were asked if they experienced PGP. In case of no response within 48 h, another message was sent. No response to the second message meant a loss to follow-up.\n[SUBTITLE] Measures/Variables [SUBSECTION] [SUBTITLE] PGP prevalence and severity [SUBSECTION] During the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.\nDuring the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.\n[SUBTITLE] Pelvic floor measurements [SUBSECTION] A pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.\nA pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.\n[SUBTITLE] DRA measurements [SUBSECTION] The width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].\nThe width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].\n[SUBTITLE] PGP prevalence and severity [SUBSECTION] During the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.\nDuring the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.\n[SUBTITLE] Pelvic floor measurements [SUBSECTION] A pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.\nA pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.\n[SUBTITLE] DRA measurements [SUBSECTION] The width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].\nThe width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].\n[SUBTITLE] Statistical analysis [SUBSECTION] The sample size was determined based on a priori calculations, based on the reports of Wu et al., in which the incidence of postpartum low back pain and /or PGP is estimated at 25% [35]. The following formula was used:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$n=\\frac{{Z}^{2}P(1-P)}{{d}^{2}},$$\\end{document}\nwhere n is the sample size, the Z value (corresponding to the significance level p 0.05) is 1.96. P value is the expected occurrence of pain, and the d value is 0.05 (recommended value for an expected prevalence between 10 and 90% [36]). Based on this calculation, the minimum sample size was 288. A high drop-out rate was also expected due to the specificity of the postpartum period, as observed by other authors [37]. To secure the minimal number of participants for phase 2 of the study (“follow-up”), the sample size was increased to a minimum of 400.\nFor continuous variables, mean and SD were calculated. Categorical data are presented as numbers and percentages. The prevalence of PGP was calculated by dividing the number of women classified with PGP by the total number of women who participated in the study. To assess the possible differences between women who responded 6 weeks postpartum and those who were lost to follow-up, Student t-test, the Mann-Whitney U and the the χ2 test were used depending on the type and normality of data.\nUnivariable logistic regression analyses were fitted to test individual factors for association with PGP shortly postpartum (phase 1). Multivariable logistic models were used to identify associations with more than one factor included in the model. The selection of factors to include in the multivariable models was informed by the univariable results. Variables with p-values less than or equal to 0.1 were included in the multivariable regression model. The number of possible investigated factors in the multivariable model was calculated assuming 10 participants per potential associated factor with division by the obtained prevalence rate [38]. The best subset of explanatory variables was selected manually by excluding the variables with the smallest contribution to the model. The predictive power of the model was calculated by Nagelkerke R-square (R²). Missing data were not included in the analysis. Alpha was set at 0.05. Statistical analyses were performed using PQStat ver. 1.8.2.166.\nThe sample size was determined based on a priori calculations, based on the reports of Wu et al., in which the incidence of postpartum low back pain and /or PGP is estimated at 25% [35]. The following formula was used:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$n=\\frac{{Z}^{2}P(1-P)}{{d}^{2}},$$\\end{document}\nwhere n is the sample size, the Z value (corresponding to the significance level p 0.05) is 1.96. P value is the expected occurrence of pain, and the d value is 0.05 (recommended value for an expected prevalence between 10 and 90% [36]). Based on this calculation, the minimum sample size was 288. A high drop-out rate was also expected due to the specificity of the postpartum period, as observed by other authors [37]. To secure the minimal number of participants for phase 2 of the study (“follow-up”), the sample size was increased to a minimum of 400.\nFor continuous variables, mean and SD were calculated. Categorical data are presented as numbers and percentages. The prevalence of PGP was calculated by dividing the number of women classified with PGP by the total number of women who participated in the study. To assess the possible differences between women who responded 6 weeks postpartum and those who were lost to follow-up, Student t-test, the Mann-Whitney U and the the χ2 test were used depending on the type and normality of data.\nUnivariable logistic regression analyses were fitted to test individual factors for association with PGP shortly postpartum (phase 1). Multivariable logistic models were used to identify associations with more than one factor included in the model. The selection of factors to include in the multivariable models was informed by the univariable results. Variables with p-values less than or equal to 0.1 were included in the multivariable regression model. The number of possible investigated factors in the multivariable model was calculated assuming 10 participants per potential associated factor with division by the obtained prevalence rate [38]. The best subset of explanatory variables was selected manually by excluding the variables with the smallest contribution to the model. The predictive power of the model was calculated by Nagelkerke R-square (R²). Missing data were not included in the analysis. Alpha was set at 0.05. Statistical analyses were performed using PQStat ver. 1.8.2.166.", "St. Sophia Specialist Hospital in Warsaw, Poland served as a recruitment site for this study. It is a tertiary, publicly funded hospital with over 6500 births annually. A free consultation with a pelvic health physiotherapist is part of standard care for every woman after delivery at this hospital. The participants were recruited from among the women attending the consultation.", "Women between 18 and 45 years old who attended free physiotherapy consultation 24–72 h postpartum were invited to participate in the study. The main exclusion criteria were additional comorbidities potentially causing PGP-like symptoms (rheumatoid arthritis, ankylosing spondylitis, Scheuermann disease, Ehlers-Danlos syndrome, spinal surgeries, nerve root compression, spondylolisthesis), contraindications for pelvic floor examination (puerperal genital hematoma, diffuse perineal edema, perineal wound dehiscence, bladder catheterization) and severe postpartum complications (internal bleeding, femoral artery embolism, pelvic fracture). To limit the sampling bias and ensure that patients were randomly included in the study, every third participant of the postpartum physiotherapy consultation was invited, and recruitment for the study took place every third day.", "The study was conducted in two phases. Phase 1 was carried out at the hospital. Participants who met the inclusion criteria and gave informed consent were included in the study. Age, education, parity (defined as previous deliveries > 24 weeks gestation), delivery type (vaginal, forceps/vacuum extractor, cesarean), infant body mass (< 4000 g/4000 g or more), height, body mass gain during pregnancy, the use of anesthesia during delivery were recorded from the patient medical record and confirmed via self-report. The women were asked if they had experienced PGP during the last pregnancy (yes/no and if yes - in what location) and urinary incontinence during or before the pregnancy (yes/no). The presence and severity of PGP were assessed by the physiotherapist. Additionally, each woman received an examination of the pelvic floor and abdominal muscles. All examinations were performed by the same registered pelvic health physiotherapist who had completed advanced training in urogynaecology and was certified by the Polish Urogynecological Society to digitally examine the pelvic floor muscles.\nIn phase 2 (“follow-up”), all women who participated in phase 1 were contacted via text message 6 weeks postpartum. In the message, they were asked if they experienced PGP. In case of no response within 48 h, another message was sent. No response to the second message meant a loss to follow-up.", "[SUBTITLE] PGP prevalence and severity [SUBSECTION] During the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.\nDuring the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.\n[SUBTITLE] Pelvic floor measurements [SUBSECTION] A pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.\nA pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.\n[SUBTITLE] DRA measurements [SUBSECTION] The width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].\nThe width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].", "During the examination by the physiotherapist in phase 1 (shortly postpartum), every patient was asked the question: “Do you have pelvic girdle pain in the places marked in the figure (Fig. 1), which aggravates during activities such as standing up, walking, or rolling from side to side?“ The participants reported their pain by indicating its location on a body chart. This was confirmed by pointing to the site of the pain in their body. The physiotherapist then carried out a further clinical examination to confirm the presence of PGP. For PGP classification, existing guidelines and previous reports were used [1, 3, 4]. Firstly, the lumbar spine examination was performed (flexion/extension movements, lateral rotations, lateral bends, Laseque test) to exclude lumbar causes of pain in the pelvic girdle region. This was followed by the tests dedicated to the pelvic girdle: Posterior Pelvic Pain Provocation (P4) test, distraction test, compression test, palpation of the pubic symphysis, modified Trendelenburg test, active straight leg raise test (ASLR). At least two tests had to be positive for PGP to be confirmed. For the ASLR test, the scores on both sides were added, and the total score ranged from 0 to 10. An ASLR total score of 4 and above was considered positive.\n\nFig. 1Pain maps used for assessing the presence of PGP\n\nPain maps used for assessing the presence of PGP\nPatients with confirmed PGP rated their mean pain intensity during a day on a Numerical Rating Scale (NRS) from 0 to 10, where 0 meant no pain and 10 meant the worst pain imaginable [19]. We designated the categorical cut-off points for the NRS as mild (1–4), moderate (5–6), and severe (7 to 10) [20]. Functional limitations were assessed with the Polish version of The Pelvic Girdle Questionnaire (PGQ) [21]. Values 0–28 were interpreted as low, 28–62 as moderate, and > 62 as high. [22]\nAssessment of PGP prevalence 6 weeks postpartum was carried out via text message. To increase the certainty of our results, we have provided the figure with marked pain locations, and also a short description of PGP-related symptoms. The women were asked: “Are you currently experiencing pelvic/sacrum/coccyx/pubic symphysis pain (Fig. 1) with or without a feeling of instability in these areas, arising or worsening with changes in position or movement?”. The message was accompanied by an image with marked pain locations (Fig. 1). Women were classified as having pelvic girdle syndrome if they indicated 1st and 3rd location. If the response was positive, the patient was asked to rate the pain using NRS and functional limitations by filling out and returning the PGQ via email or MMS.", "A pelvic floor assessment was performed using the palpation examination. We decided this would be the only appropriate method to be used in the early postpartum period as it is fast, painless, non-invasive, and requires no additional equipment. All patients were assessed in a crook lying position. The PERFECT scheme [23] with the 6-point OXFORD scale (0–5) was used to evaluate a maximal voluntary contraction (MVC) of the pelvic floor. The OXFORD scale is a reliable measure of the MVC with acceptable intra-observer and test-retest reliability [24]. The existing research showed that the palpation assessment using the OXFORD scale was consistent with the ultrasound examination results [25].\nA seven-point scale proposed by Reising et al. was used to assess muscle tone with values ranging from − 3 (very hypotonic muscles) to + 3 (very hypertonic muscles) with 0 stating normal tone. This scale has been studied for its reliability showing fair-to-moderate interrater reliability with correlation coefficients of 0.2–0.5 [26]. Weak-to-fair associations between the Reissing scale, dynamometry, and ultrasound imaging with correlation coefficients of 0.2–0.4 have been previously shown [27].\nAdditionally, possible activation of synergistic muscles (gluteal muscles, adductors, and abdomen), and the ability to activate the pelvic floor muscles without breath-holding were observed during the pelvic floor examination. That was done in order to determine the correctness of pelvic floor activation: isolated pelvic floor contraction without breath-holding was considered correct.", "The width of rectus abdominus muscle bellies (inter-recti distance, IRD) was determined by palpation using the procedure reported in previous studies [28]. The women were asked to lie in a standard supine position with their knees bent. Then, they were asked to perform the abdominal curl-up by raising the head and upper torso until the shoulder blades left the examination bed (Fig. 2). Measurements were taken at the navel level and 4.5 cm above and below it [29, 30]. As done in previous studies, the point of largest width was selected for the analysis [13–15]. Mota et al. showed good intra-rater reliability (Kw>0.7) in terms of palpation measurements of IRD [28] and Benjamin et al. showed moderate to very good correlation of IRD palpation with ultrasound (r = 0.75–0.98) [31]. According to other studies implementing this method [13, 15, 29, 30] DRA was considered when the IRD value was ≥ 2 fingerbreadths. The participants in this study were divided into four categories depending on the largest palpation measurement (number of fingers) in one of three locations: no DRA (IRD < 2 fingerbreadths), mild DRA (IRD 2;< 3), moderate DR (IRD 3;<4), severe DRA (IRD > 4) [13].\n\nFig. 2DRA and linea alba assessment\n\nDRA and linea alba assessment\nIt has been suggested that the integrity of linea alba may influence the capacity to stabilize the pelvis and the lumbar spine, and the ability of the linea alba to transmit forces across the midline may have a more significant impact on function than the magnitude of the IRD [32]. Therefore, in the context of the assessment of DRA and linea alba dysfunction, not only IRD but also the stiffness and distortion/bulging at the level of linea alba could play an important role [32, 33]. To investigate any possible association with PGP, we have adopted a simplified method of assessing the doming of the abdominal midline. It was defined as “abdominal midline doming” (yes/no). During the curl-up test, the physiotherapist observed whether the abdominal midline bulged. Although this method does not allow to determine which structures are bulging (e.g., linea alba or “just” subcutaneous tissue), it may give us a simplified estimation of pressure management in the abdominal cavity. A similar assessment has recently been used in another research [34].", "The sample size was determined based on a priori calculations, based on the reports of Wu et al., in which the incidence of postpartum low back pain and /or PGP is estimated at 25% [35]. The following formula was used:\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$n=\\frac{{Z}^{2}P(1-P)}{{d}^{2}},$$\\end{document}\nwhere n is the sample size, the Z value (corresponding to the significance level p 0.05) is 1.96. P value is the expected occurrence of pain, and the d value is 0.05 (recommended value for an expected prevalence between 10 and 90% [36]). Based on this calculation, the minimum sample size was 288. A high drop-out rate was also expected due to the specificity of the postpartum period, as observed by other authors [37]. To secure the minimal number of participants for phase 2 of the study (“follow-up”), the sample size was increased to a minimum of 400.\nFor continuous variables, mean and SD were calculated. Categorical data are presented as numbers and percentages. The prevalence of PGP was calculated by dividing the number of women classified with PGP by the total number of women who participated in the study. To assess the possible differences between women who responded 6 weeks postpartum and those who were lost to follow-up, Student t-test, the Mann-Whitney U and the the χ2 test were used depending on the type and normality of data.\nUnivariable logistic regression analyses were fitted to test individual factors for association with PGP shortly postpartum (phase 1). Multivariable logistic models were used to identify associations with more than one factor included in the model. The selection of factors to include in the multivariable models was informed by the univariable results. Variables with p-values less than or equal to 0.1 were included in the multivariable regression model. The number of possible investigated factors in the multivariable model was calculated assuming 10 participants per potential associated factor with division by the obtained prevalence rate [38]. The best subset of explanatory variables was selected manually by excluding the variables with the smallest contribution to the model. The predictive power of the model was calculated by Nagelkerke R-square (R²). Missing data were not included in the analysis. Alpha was set at 0.05. Statistical analyses were performed using PQStat ver. 1.8.2.166.", "A total of 415 women were invited to participate, 4 of whom did not consent to join the study. Thus, 411 women were included in phase 1 (shortly postpartum). Table 1. presents the characteristics of the participants. In phase 2 (“follow-up”), 268 women replied to the text message (65.2% of the initial group). No statistically significant differences were found between those who responded to the message and those who did not in terms of variables assessed shortly postpartum (in phase 1): BMI, age, presence of PGP postpartum, parity, DRA severity, and ability to activate pelvic floor muscles.\n\nTable 1Characteristics of the study group, n = 411All participantsn = 411With PGP n = 37Without PGP n = 374\nAge\n31.17 ± 4.0132.35 ± 4.3331.06 ± 3.96Height [cm],167.46 ± 5.69167.00 ± 6.64167.51 ± 5.60\nBMI before pregnancy\n22.14 ± 3.5722.46 ± 3.2022.10 ± 3.60\nBody mass gain in pregnancy [kg]\n14.30 ± 4.9414.92 ± 4.2814.24 ± 4.99Education, n (%) †vocational education2 (0.5)11secondary education43 (10.5)242university education366 (89)35331\nParity, n (%)\n1.72 ± 0.961.95 ± 1.101.70 ± 0.941209 (50.80)2143 (34.80)337 (9.00)412 (3.00)58 (2.00)61 (0.20)71 (0.20)\nMode of the last delivery, n (%)\nvaginal387 (94.20)37 (100)350 (93.60)cesarean ‡18 (4.30)018 (4.80)vacuum extractor ‡6 (1.50)06 (1.60)\nPerineal injury during recent delivery, n (%)\nnone162 (39.50)18 (48.60)144 (38.50)1st grade138 (33.50)9 (24.30)129 (34.50)episiotomy111 (27)10 (27)101 (27)\nAnesthesia during last delivery, n (%)\nnone238 (57.91)24 (64.86)214 (57.22)epidural154 (37.47)12 (32.43)142 (37.97)spinal19 (4.62)1 (2.70)18 (4.81)\nInfant body mass ≥ 4000 g\nyes336 (81.75)6 (16.22)330 (88.24)no75 (18.25)31 (83.78)44 (11.76)† Participants with vocational education were excluded (from this calculations) because of the low number of records; ‡ no statistical analysis due to low number of observations; χ² Chi2 tet value; U Manna Whitney test value\n\nCharacteristics of the study group, n = 411\n† Participants with vocational education were excluded (from this calculations) because of the low number of records; ‡ no statistical analysis due to low number of observations; χ² Chi2 tet value; U Manna Whitney test value\nAmong the patients in phase 1 of the study (shortly postpartum), 47.9% (n = 197) reported PGP symptoms during pregnancy. In phase 1, PGP was diagnosed in 9.0% (n = 37) of women at the early postpartum stage. In phase 2 (6 weeks postpartum), PGP was reported by 15.7% of women (n = 42). Table 2 presents the prevalence and type of PGP across different time points, and Table 3 shows the severity of pain and functional disturbances in both phases of the study.\n\nTable 2Prevalence and types of PGP across the time points.TimepointDuring pregnancyEarly postpartum6 weeks postpartum\nAssessment form\n\nself-reported, retrospective\n\nclinical assessment\n\nself-reported\n\nN\n\n%\n\nN\n\n%\n\nN\n\n%\n\nPGP\n197/41147.9337/411942/26815.70\nPGP\n\ntype\n\nPosterior Pelvic Pain\n86/19743.709/3724.3016/4238.10\nUnilateral pain\n17/1978.602/375.402/424.80\nSymphyseal pain\n44/19722.309/3724.3012/4228.60\nPelvic Girdle Syndrome\n47/19723.9016/3743.2011/4226.20\nUnilateral pain + symphyseal pain\n3/1971.501/372.701/422.40\n\nPrevalence and types of PGP across the time points.\n\nPGP\n\n\ntype\n\n\nTable 3The severity of pain and functional limitations at phase 1 and 2 of the studyTimepointEarlyPostpartum6 weekspostpartum\nMean (SD)\n\n(min-max)\n\nN\n\nMean (SD)\n\n(min-max)\n\nN\n\nNRS\n5.34 (1.74)(3–9)37/374.63 (2.04)(1–8)24/42\nPGQ [%]\n48.87 (16.40)(23.61–82.61)32/3725.80 (14.90)(6.67–56.94)13/42The values of the PGQ questionnaire from the early postpartum stage were missing in five cases – they were left blank or only partially completed. Six weeks after delivery, the pain intensity value on the NRS was reported by 24 women, and the PGQ questionnaire was completed only by 13\n\nThe severity of pain and functional limitations at phase 1 and 2 of the study\n5.34 (1.74)\n(3–9)\n4.63 (2.04)\n(1–8)\n48.87 (16.40)\n(23.61–82.61)\n25.80 (14.90)\n(6.67–56.94)\nThe values of the PGQ questionnaire from the early postpartum stage were missing in five cases – they were left blank or only partially completed. Six weeks after delivery, the pain intensity value on the NRS was reported by 24 women, and the PGQ questionnaire was completed only by 13\nThe univariable analyses showed a higher likelihood of PGP shortly postpartum in women who declared PGP during pregnancy and among women with doming at the abdominal midline in the projection of linea alba (Table 4.).\n\nTable 4Univariable analysis of factors associated with pelvic girdle pain (PGP) shortly postpartum (phase 1)PGP (+)n = 37PGP (-)N = 374OR (95% CI)p-value\nPGP during pregnancy (yes)\n\nn (%)\n34 (91.90)163 (43.60)14.67 (4.43–48.61)\n< 0.01\n\nAge\n\nmean (SD)\n32.35(4.33)31.06 (3.96)1.08 (0.99–1.18)0.06\nBody mass gain during pregnancy\n\nmean (SD)\n32.68 (4.60)31.59 (4.37)1.03 (0.96–1.10)0.42\nBMI before pregnancy\n\nmean (SD)\n22.46 (3.20)22.1 (3.60)1.03 (0.94–1.12)0.56\nNumber of previous deliveries mean (SD)\n1.95 (1.10)1.7 (0.94)1.27 (0.93–1.71)0.14\nUrinary incontinence during pregnancy or before\n\n(yes) n (%)\n15 (40.50)205 (54.80)0.56 (0.28–1.12)0.10\nAbdominal midline doming (yes), n (%)\n20 (54.10)134 (36.40)2.05 (1.04–4.06)\n0.04\n\nDR severity\n\nn (%)\n\nnone, IRD < 2\n9 (24.30)149 (40.50)1.27 (0.95–1.69)0.11mild,\nIRD 2;< 3\n8 (21.60)72 (19.60)\nmoderate, IRD 3;<4\n12 (32.40)75 (20.40)\nsevere, IRD > 4\n8 (21.60)72 (19.60)\nOxford scale\n\nMean (SD)\n2.27(0.65)2.28 (0.90)0.99 (0.67–1.45)0.96\nReissing scale\n\nmean (SD)\n0.32 (1.11)-0.53 (0.96)1.25 (0.88–1.77)0.21\nCorrect activation of pelvic floor (yes), n (%)\n13 (35.10)134 (35.80)1.36 (0.69–2.70)0.37\n\nUnivariable analysis of factors associated with pelvic girdle pain (PGP) shortly postpartum (phase 1)\n\nPGP during pregnancy (yes)\n\n\nn (%)\n\n\nAge\n\n\nmean (SD)\n\n\nBody mass gain during pregnancy\n\n\nmean (SD)\n\n\nBMI before pregnancy\n\n\nmean (SD)\n\n\nUrinary incontinence during pregnancy or before\n\n\n(yes) n (%)\n\n\nDR severity\n\n\nn (%)\n\nmild,\n\nIRD 2;< 3\n\n\nOxford scale\n\n\nMean (SD)\n\n\nReissing scale\n\n\nmean (SD)\n\nBased on the obtained prevalence of postpartum PGP, we could include up to 4 variables into the multivariable model [38]. The final model, in which we obtained statistically significant results for all included items, consisted of 3 variables. This multivariable regression analysis showed that the odds of having PGP shortly postpartum were higher in women with increased age (10% higher likelihood with every year of age), declaring PGP during pregnancy, and with higher values of Reissing scale. To enhance the interpretability, we then analyzed the Reissing scale in the following categories: hypotonus (range − 3 to -1) and increased muscle tone (range 1 to 3), with the reference value being normotonus (0). However, after this procedure, the Reissing scale became not statistically significant (Table 5). The calculated Nagelkerke R-square was 0.2.\n\nTable 5Multivariable analysis of factors associated with pelvic girdle pain (PGP) shortly postpartum (phase 1)OR (95% CI)p-value\nPresence of PGP during pregnancy (yes)\n14.83 (4.340-48.721)\n< 0.0001\n\nAge\n1.12 (1.009–1.214)\n0.032\n\nReissing scale\n\nReissing scale – not grouped\n1.43 (1.003–2.046)\n0.048\n\nReissing scale when grouped\n\nnormal tone (0)\nreferencereference\ndecreased tone \n\n(range − 3 to -1)\n0.53 (0.227–1.220)0.134\nincreased tone \n\n(range 1 to 3)\n0.37 (0.594–4.078)0.368\n\nMultivariable analysis of factors associated with pelvic girdle pain (PGP) shortly postpartum (phase 1)\n\ndecreased tone \n\n\n(range − 3 to -1)\n\n\nincreased tone \n\n\n(range 1 to 3)\n", "Our study showed that nearly 10% of women were diagnosed with PGP during the first days postpartum, and almost 16% reported similar symptoms 6 weeks later. The mean pain intensity and functional limitations within the first days postpartum were moderate, with values corresponding to mild/low 6 weeks postpartum. The likelihood of experiencing PGP shortly after delivery increased with age and reporting PGP during pregnancy. The doming of the abdominal midline was significantly associated with PGP shortly postpartum only in the univariable analysis. The remaining variables related to diastasis recti or pelvic floor function were not associated with PGP shortly after delivery (24–72 h postpartum).\nThe reported prevalence of postpartum PGP around 12 weeks after delivery ranges from 3.4 to 43.0% [39–46] with the majority of studies [41–45] showing a higher prevalence than demonstrated in this research. This variation may be due to several reasons. Firstly, diverse diagnoses and terminology were used in the mentioned studies, which could lead to discrepancies in prevalence. In our research, postpartum PGP was defined as a pain that persisted postpartum or occurred within the first weeks after delivery [2]. However, in the study of Stomp van den Berg et al. [44] 25% of the 234 women who had PGP at 12 weeks postpartum had no PGP between 0 and 6 weeks after delivery. Secondly, cultural and ethnic factors can play a role in the processes related to pain perception [47]. Although PGP is prevalent worldwide, it is not recognized by health care systems in some countries. Our previous study has shown that PGP during pregnancy was more common in Norwegian than Polish women [6]. In Norway, PGP is one of the most common causes of sick leave among pregnant women [48]. In Poland, PGP is not commonly recognized, and the term ‘pelvic girdle pain’ is not widely used within health care services. Lower social awareness about this condition could lead to lower reporting. The possible role of ethnicity was noticed in another PGP study [49] indicating a more detailed investigation encompassing cultural and ethnic influences associated with PGP is needed.\nWe could observe similar discrepancies when analyzing the values related to pain intensity and functional limitations, possibly related to the same reasons as those mentioned above. For instance, in the study by Mukkanavar et al. [41] among Indian postpartum women, as many as 84.5% participants with PGP between the 3rd and 18th week after delivery rated their symptoms as greater than 60 mm on the VAS scale. Stomp van de Berg [44] reported median pain intensity 6 weeks postpartum at 4.3 of NRS scale. In the study of Dunn et al. [42] the mean pain intensity values measured on VAS scale were between 22.5 and 55, depending on the location of PGP and co-existing dysfunctions. Our results seem to be in line with those of Sakamoto et al. [50] who also measured functional limitations with PGQ. In the second day postpartum the mean values were oscillating around 47% (95%CI 40–54), while 4 weeks after − 19% (95%CI 12–25).\nOur results showing higher PGP prevalence 6 weeks postpartum when compared to early postpartum period may seem contradictory to previous reports [37, 51]. However, it has to be noted that the cited studies followed women experiencing PGP already during pregnancy. The occurrence of pregnancy-related PGP may be delayed up to the first weeks postpartum [2]. By following all women (with and without pain), our study could capture those individuals that developed pain after the initial examination, 24-72 h postpartum. Additionally, early postpartum period is associated with more bed rest when compared to 6 weeks postpartum when PGP symptoms could be more noticeable and bothersome.\nWhen it comes to factors associated with postpartum PGP, our results are in line with previous reports. A recent systematic review by Wiezer et al. [52] confirmed PGP during pregnancy as a risk factor for persistent postpartum pain. These findings suggest that asking women whether they have experienced PGP during pregnancy may help identify women at risk of persistent postpartum pain. The association between postpartum PGP and age has also been previously shown. Gausel et al. [46] reported age 30 and above as the risk factors for persistent postpartum pain. In European countries, primiparous women are becoming older. In Poland, the mean age of women having their first baby in 2019 was 27.4 and, although constantly increasing, is still one of the lowest in Europe [53]. Increasing maternal age may have several consequences. In accordance with previously mentioned studies, our results indicate that pregnant women who deliver past a certain age should receive special physiotherapy care.\nIn our study, the doming of the abdominal wall in the projection of linea alba was a statistically significant factor only in univariable analysis and there were no associations between the DRA severity (size of IRD) and the presence of PGP shortly postpartum. This is different when compared to our recently published matched-case control studies [54, 55]. However, in mentioned reports participants were matched according to age and parity, mode of delivery and time postpartum. This may suggest that although DRA features and postpartum PGP may co-exist, those associations are not straightforward and there are possibly other factors that may mediate this relationship.\nOur study did not reveal any associations between pelvic floor function and PGP shortly postpartum, despite previous reports [17, 55]. This may be due to the timing of phase 1 of the study when the measurements were taken - some differences in the pelvic floor function may be too subtle to be detected using screening palpation examination in the early postpartum period. Our other hypothesis is that there are no differences in the pelvic floor muscle function between women with and without postpartum PGP shortly after delivery. They may be more visible with time, while pain persists and the adaptive changes in the pelvic floor occur, which could be supported by our other PGP study [55].\n[SUBTITLE] Strengths and limitations [SUBSECTION] This was the first large-scale study conducted in Poland using the recommended guidelines for classifying and investigating postpartum PGP prevalence with the use of screening palpation examination of the pelvic floor and abdominal muscles. To our knowledge, this is also the first study in the central-eastern region in Europe. Considering that this region is inhabited mainly by Caucasian women with a similar physiognomy, our results may estimate postpartum PGP prevalence in this part of Europe.\nThe main limitation of this work is the high drop-out rate in phase 2 (“follow-up”). For this reason, the prevalence of PGP 6 weeks postpartum may be underestimated. Time constraints, lack of trust, and low awareness of clinical trials are the main barriers to participation in research projects [56]. Additionally, the first weeks after delivery are challenging for many women, and research obligations may not be their priority. High drop-out rates were also reported by another study investigating PGP 6 weeks postpartum via SMS where the response rate of 43% was recorded 6 weeks postpartum [37]. Another limitation could be caused by the assessment of PGP 6 weeks via self-reports. However, this method was used in previous PGP research [37, 42] and a study by Rejano-Campo et al. [57] showed that self-reported PGP was verified by specific clinical tests in nearly all cases.\nFinally, we cannot exclude potential selection bias. Although we have made an effort to adopt random recruitment for phase 1 (shortly postpartum), we have included mainly highly educated women from only one center located in the capital city of Poland. This should be taken into account while inferring results from our sample to the general population.\nThis was the first large-scale study conducted in Poland using the recommended guidelines for classifying and investigating postpartum PGP prevalence with the use of screening palpation examination of the pelvic floor and abdominal muscles. To our knowledge, this is also the first study in the central-eastern region in Europe. Considering that this region is inhabited mainly by Caucasian women with a similar physiognomy, our results may estimate postpartum PGP prevalence in this part of Europe.\nThe main limitation of this work is the high drop-out rate in phase 2 (“follow-up”). For this reason, the prevalence of PGP 6 weeks postpartum may be underestimated. Time constraints, lack of trust, and low awareness of clinical trials are the main barriers to participation in research projects [56]. Additionally, the first weeks after delivery are challenging for many women, and research obligations may not be their priority. High drop-out rates were also reported by another study investigating PGP 6 weeks postpartum via SMS where the response rate of 43% was recorded 6 weeks postpartum [37]. Another limitation could be caused by the assessment of PGP 6 weeks via self-reports. However, this method was used in previous PGP research [37, 42] and a study by Rejano-Campo et al. [57] showed that self-reported PGP was verified by specific clinical tests in nearly all cases.\nFinally, we cannot exclude potential selection bias. Although we have made an effort to adopt random recruitment for phase 1 (shortly postpartum), we have included mainly highly educated women from only one center located in the capital city of Poland. This should be taken into account while inferring results from our sample to the general population.\n[SUBTITLE] Implications [SUBSECTION] Obtained results with regard to other recently published matched case-control studies suggest that the relationship between PGP and DRA-related factors is multidimensional and not straightforward as previously suggested. Assessment of DRA-related factors seems not to be a crucial part of the screening for postpartum PGP but may be of greater importance when assessing individuals with postpartum PGP. Future research should further investigate the possible, multidimensional interactions between PGP and the whole abdominal wall complex (not restricted to only IRD as recommended by Delphi Consensus Study for the conservative management of pregnancy-related DRA [58]), and whether the DRA-related dysfunctions “only” co-exist with PGP or play a role in it. In that case, future studies focusing on creating adequate tension through the abdominal wall during PGP rehabilitation may be feasible. It should all be adjusted for psychosocial factors, which weren’t taken into account in our study. However, they are related to central pain mechanisms observed in individuals with persistent postpartum PGP and could be important factors filling the gaps in our current understanding of postpartum PGP [7, 59].\nObtained results with regard to other recently published matched case-control studies suggest that the relationship between PGP and DRA-related factors is multidimensional and not straightforward as previously suggested. Assessment of DRA-related factors seems not to be a crucial part of the screening for postpartum PGP but may be of greater importance when assessing individuals with postpartum PGP. Future research should further investigate the possible, multidimensional interactions between PGP and the whole abdominal wall complex (not restricted to only IRD as recommended by Delphi Consensus Study for the conservative management of pregnancy-related DRA [58]), and whether the DRA-related dysfunctions “only” co-exist with PGP or play a role in it. In that case, future studies focusing on creating adequate tension through the abdominal wall during PGP rehabilitation may be feasible. It should all be adjusted for psychosocial factors, which weren’t taken into account in our study. However, they are related to central pain mechanisms observed in individuals with persistent postpartum PGP and could be important factors filling the gaps in our current understanding of postpartum PGP [7, 59].", "This was the first large-scale study conducted in Poland using the recommended guidelines for classifying and investigating postpartum PGP prevalence with the use of screening palpation examination of the pelvic floor and abdominal muscles. To our knowledge, this is also the first study in the central-eastern region in Europe. Considering that this region is inhabited mainly by Caucasian women with a similar physiognomy, our results may estimate postpartum PGP prevalence in this part of Europe.\nThe main limitation of this work is the high drop-out rate in phase 2 (“follow-up”). For this reason, the prevalence of PGP 6 weeks postpartum may be underestimated. Time constraints, lack of trust, and low awareness of clinical trials are the main barriers to participation in research projects [56]. Additionally, the first weeks after delivery are challenging for many women, and research obligations may not be their priority. High drop-out rates were also reported by another study investigating PGP 6 weeks postpartum via SMS where the response rate of 43% was recorded 6 weeks postpartum [37]. Another limitation could be caused by the assessment of PGP 6 weeks via self-reports. However, this method was used in previous PGP research [37, 42] and a study by Rejano-Campo et al. [57] showed that self-reported PGP was verified by specific clinical tests in nearly all cases.\nFinally, we cannot exclude potential selection bias. Although we have made an effort to adopt random recruitment for phase 1 (shortly postpartum), we have included mainly highly educated women from only one center located in the capital city of Poland. This should be taken into account while inferring results from our sample to the general population.", "Obtained results with regard to other recently published matched case-control studies suggest that the relationship between PGP and DRA-related factors is multidimensional and not straightforward as previously suggested. Assessment of DRA-related factors seems not to be a crucial part of the screening for postpartum PGP but may be of greater importance when assessing individuals with postpartum PGP. Future research should further investigate the possible, multidimensional interactions between PGP and the whole abdominal wall complex (not restricted to only IRD as recommended by Delphi Consensus Study for the conservative management of pregnancy-related DRA [58]), and whether the DRA-related dysfunctions “only” co-exist with PGP or play a role in it. In that case, future studies focusing on creating adequate tension through the abdominal wall during PGP rehabilitation may be feasible. It should all be adjusted for psychosocial factors, which weren’t taken into account in our study. However, they are related to central pain mechanisms observed in individuals with persistent postpartum PGP and could be important factors filling the gaps in our current understanding of postpartum PGP [7, 59].", "The findings presented in this study suggest that every tenth Polish woman may experience PGP during the first days postpartum and every sixth can report similar symptoms 6 weeks later. The pain intensity and functional limitations tend to subside over time: from moderate pain intensity and functional limitation shortly postpartum to mild/low 6 weeks later. Nevertheless, postpartum PGP should not be ignored, especially in the context of the observed continued increase in chronic pain syndromes and their associated consequences. Older age, PGP during pregnancy, and doming of the abdominal midline at the level of linea alba were associated with the experience of PGP within the first days postpartum. Our study showed no association between pelvic floor function and PGP shortly postpartum. However, this may be due to the chosen methodology (assessment shortly postpartum)." ]
[ null, null, null, null, null, null, null, null, null, null, "results", "discussion", null, null, "conclusion" ]
[ "Pelvic girdle pain", "Postpartum period", "Prevalence", "Pelvic floor", "Pelvic floor disorders", "rectus abdominis" ]
Innovative approach to monitor performance of integrated disease surveillance and response after the Ebola outbreak in Sierra Leone: lessons from the field.
36266711
Supervision of healthcare workers improves performance if done in a supportive and objective manner. Regular supervision is a support function of Integrated Disease Surveillance and Response (IDSR) strategy and allows systematic monitoring of IDSR implementation. Starting 2015, WHO and other development partners supported the Ministry of Health and Sanitation (MoHS) to revitalize IDSR in Sierra Leone and to monitor progress through supportive supervision assessments. We report on the findings of these assessments.
BACKGROUND
This was a cross-sectional study where six longitudinal assessments were conducted in randomly selected health facilities. Health facilities assessed were 71 in February 2016, 99 in July 2016, 101 in May 2017, 126 in August 2018, 139 in February 2019 and 156 in August 2021. An electronic checklist based on selected core functions of IDSR was developed and uploaded onto tablets using the Open Data Kit (ODK) platform. Supervision teams interviewed health care workers, reviewed documents and made observations in health facilities. Supervision books were used to record feedback and corrective actions. Data from the supervisory visits was downloaded from ODK platform, cleaned and analysed. Categorical data was summarized using frequencies and proportions while means and medians were used for continuous variables. Z test was used to test for differences in proportions.
METHODS
Completeness of IDSR reporting improved from 84.5% in 2016 to 96% in 2021 (11.5% points; 95% CI 3.6, 21.9; P-value 0.003). Timeliness of IDSR reports improved from 80.3 to 92% (11.7% points; 95% CI 2.4, 22.9; P-value 0.01). There was significant improvement in health worker knowledge of IDSR concepts and tools, in availability of IDSR standard case definition posters and reporting tools and in data analysis practices. Availability of vaccines and temperature monitoring tools in health facilities also improved significantly but some indicators dropped such as availability of IDSR technical guidelines and malaria testing kits and drugs.
RESULTS
Supervision using electronic tool contributed to health systems strengthening through longitudinal tracking of core IDSR indicators and other program indicators such as essential malaria commodities and availability and status of routine vaccines. Supervision using electronic tools should be extended to other programs.
CONCLUSION
[ "Humans", "Hemorrhagic Fever, Ebola", "Public Health Surveillance", "Sierra Leone", "Cross-Sectional Studies", "Disease Outbreaks" ]
9584265
null
null
null
null
Results
[SUBTITLE] Coordination of IDSR activities in health facilities [SUBSECTION] IDSR focal persons were present in most health facilities during all assessments. When compared to the baseline assessment, the proportion of IDSR focal persons who participated in the monthly district management team meetings increased significantly from 74.6% to 2016 (baseline) to 96.1% at end line in 2021 (increase of 21.5% points; 95% CI (11.8, 32.9); P-value < 0.0001). Conversely, availability of IDSR technical guidelines at the health facilities declined from 97.2% to 2016 to 82.1% in 2021 (decrease of 15.1% points; 95% CI (6.5, 22.2); P-value 0.002) (Table 2). The decline in availability of IDSR technical guidelines was mainly due to loss without replacement. Table 2Comparison of IDSR indicators in selected health facilities, Sierra Leone, 2016–2021Core FunctionIndicatorsFeb_2016July_2016May_2017Aug_2018Feb_2019Aug_2021End Line vs. BaselineP valueN = 71n (%)N = 99n (%)N = 101n (%)N = 126n (%)N = 139n (%)N = 156n (%)Difference (95 CI)CoordinationHealth facility (HF) has IDSR focal person69 (97.2)97 (98.0)100 (99.0)121 (96.0)135 (97.1)153 (98.1)0.9 (-3.2, 7.9)0.67HF IDSR focal person attends district monthly meetings53 (74.6)87 (87.9)93 (92.1)118 (93.7)132 (95.0)150 (96.1)21.5 (11.8, 32.9) < 0.0001 HF has IDSR Technical guidelines69 (97.2)89 (89.9)101 (100.0)102 (81.0)123 (88.5)128 (82.1)15.1 (6.5, 22.2) 0.002 Case IdentificationStandard Case definition poster displayed in at least one location in the health facility54 (76.1)84 (84.8)89 (88.1)114 (90.5)127 (91.4)148 (94.9)18.8 (9.2, 30.2) < 0.0001 IDSR focal person conducts active case search at least once a week56 (78.9)54 (54.5)34 (33.7)100 (79.4)75 (54.0)123 (78.8)0.1 (-12.1, 10.7)0.99Reporting of Priority diseasesHCW correctly defines epidemiologic week51 (71.8)93 (93.9)99 (98.0)108 (85.7)132 (95)151 (96.8)25 (15.0, 36.5) < 0.0001 HCW correctly defines zero reporting67 (94.4)94 (94.9)99 (98.0)121 (96.0)138 (99.3)147 (92.2)2.2 (-6.4, 8.5))0.55HCW knows the deadline for submitting weekly IDSR reports65 (91.5)96 (97.0)100 (99.0)115 (91.3)138 (99.3)151 (96.8)5.3 (-0.8, 14.3)0.09Weekly reporting forms available in HF63 (88.7)89 (89.9)96 (95.0)119 (94.4)127 (91.4)150 (96.1)7.4 (0.4, 17.1) 0.03 Line listing forms available in HF60 (84.5)78 (78.8)81 (80.2)95 (75.4)114 (82.0)122 (78.2)6.3 (-5.4, 16.0)0.27Case based forms available in HF66 (93.0)79 (79.8)91 (90.1)110 (87.3)114 (82.0)139 (89.1)3.9 (-5.4, 11.0)0.36Rumor logs available in HF39 (54.9)55 (55.6)52 (51.5)78 (61.9)84 (60.4)97 (62.1)7.2 (-6.3, 20.8)0.31Data Analysis and UseHF conducts data basic data analysis28 (39.4)49 (49.5)41 (40.6)51 (40.5)63 (45.3)99 (63.4)24.0 (10.0, 36.7) 0.0008 HF have current line graphs showing trends of priority diseases5 (7.0)11 (11.1)16 (15.8)29 (23.0)43 (30.9)72(46.1)39.1 (27.8, 47.9) < 0.0001 Outbreak notification and investigationHF reported outbreak within 12 months12 (16.9)45 (45.5)32 (31.7)10 (7.9)7 (5.0)17(10.9)6.0 (-4.2, 16.1)0.31Outbreak notified to DHMT within 48 h*11 (91.7)29 (64.4)29 (90.6)9 (90.0)5 (71.4)15(88.2%)3.5 (-24.7, 27.1)0.09Monitoring and CommunicationSupervisory visits from DHMT (at least once every three months)44 (62.0)91 (91.9)78 (77.2)79 (62.7)70 (50.4)144 (92.3)30.3(18.6, 42.4) < 0.0001 Mobile network available in HF67 (94.4)79 (79.8)91 (90.1)120 (95.2)126 (90.6)144 (92.3)2.1 (-6.5, 8.4)0.57*Denominator is number of outbreaks reported in the preceding 12 monthsHF: Health Facility; IDSR: Integrated Disease Surveillance and Response; HCW: Healthcare worker; DHMT: District Health Management Team Comparison of IDSR indicators in selected health facilities, Sierra Leone, 2016–2021 *Denominator is number of outbreaks reported in the preceding 12 months HF: Health Facility; IDSR: Integrated Disease Surveillance and Response; HCW: Healthcare worker; DHMT: District Health Management Team IDSR focal persons were present in most health facilities during all assessments. When compared to the baseline assessment, the proportion of IDSR focal persons who participated in the monthly district management team meetings increased significantly from 74.6% to 2016 (baseline) to 96.1% at end line in 2021 (increase of 21.5% points; 95% CI (11.8, 32.9); P-value < 0.0001). Conversely, availability of IDSR technical guidelines at the health facilities declined from 97.2% to 2016 to 82.1% in 2021 (decrease of 15.1% points; 95% CI (6.5, 22.2); P-value 0.002) (Table 2). The decline in availability of IDSR technical guidelines was mainly due to loss without replacement. Table 2Comparison of IDSR indicators in selected health facilities, Sierra Leone, 2016–2021Core FunctionIndicatorsFeb_2016July_2016May_2017Aug_2018Feb_2019Aug_2021End Line vs. BaselineP valueN = 71n (%)N = 99n (%)N = 101n (%)N = 126n (%)N = 139n (%)N = 156n (%)Difference (95 CI)CoordinationHealth facility (HF) has IDSR focal person69 (97.2)97 (98.0)100 (99.0)121 (96.0)135 (97.1)153 (98.1)0.9 (-3.2, 7.9)0.67HF IDSR focal person attends district monthly meetings53 (74.6)87 (87.9)93 (92.1)118 (93.7)132 (95.0)150 (96.1)21.5 (11.8, 32.9) < 0.0001 HF has IDSR Technical guidelines69 (97.2)89 (89.9)101 (100.0)102 (81.0)123 (88.5)128 (82.1)15.1 (6.5, 22.2) 0.002 Case IdentificationStandard Case definition poster displayed in at least one location in the health facility54 (76.1)84 (84.8)89 (88.1)114 (90.5)127 (91.4)148 (94.9)18.8 (9.2, 30.2) < 0.0001 IDSR focal person conducts active case search at least once a week56 (78.9)54 (54.5)34 (33.7)100 (79.4)75 (54.0)123 (78.8)0.1 (-12.1, 10.7)0.99Reporting of Priority diseasesHCW correctly defines epidemiologic week51 (71.8)93 (93.9)99 (98.0)108 (85.7)132 (95)151 (96.8)25 (15.0, 36.5) < 0.0001 HCW correctly defines zero reporting67 (94.4)94 (94.9)99 (98.0)121 (96.0)138 (99.3)147 (92.2)2.2 (-6.4, 8.5))0.55HCW knows the deadline for submitting weekly IDSR reports65 (91.5)96 (97.0)100 (99.0)115 (91.3)138 (99.3)151 (96.8)5.3 (-0.8, 14.3)0.09Weekly reporting forms available in HF63 (88.7)89 (89.9)96 (95.0)119 (94.4)127 (91.4)150 (96.1)7.4 (0.4, 17.1) 0.03 Line listing forms available in HF60 (84.5)78 (78.8)81 (80.2)95 (75.4)114 (82.0)122 (78.2)6.3 (-5.4, 16.0)0.27Case based forms available in HF66 (93.0)79 (79.8)91 (90.1)110 (87.3)114 (82.0)139 (89.1)3.9 (-5.4, 11.0)0.36Rumor logs available in HF39 (54.9)55 (55.6)52 (51.5)78 (61.9)84 (60.4)97 (62.1)7.2 (-6.3, 20.8)0.31Data Analysis and UseHF conducts data basic data analysis28 (39.4)49 (49.5)41 (40.6)51 (40.5)63 (45.3)99 (63.4)24.0 (10.0, 36.7) 0.0008 HF have current line graphs showing trends of priority diseases5 (7.0)11 (11.1)16 (15.8)29 (23.0)43 (30.9)72(46.1)39.1 (27.8, 47.9) < 0.0001 Outbreak notification and investigationHF reported outbreak within 12 months12 (16.9)45 (45.5)32 (31.7)10 (7.9)7 (5.0)17(10.9)6.0 (-4.2, 16.1)0.31Outbreak notified to DHMT within 48 h*11 (91.7)29 (64.4)29 (90.6)9 (90.0)5 (71.4)15(88.2%)3.5 (-24.7, 27.1)0.09Monitoring and CommunicationSupervisory visits from DHMT (at least once every three months)44 (62.0)91 (91.9)78 (77.2)79 (62.7)70 (50.4)144 (92.3)30.3(18.6, 42.4) < 0.0001 Mobile network available in HF67 (94.4)79 (79.8)91 (90.1)120 (95.2)126 (90.6)144 (92.3)2.1 (-6.5, 8.4)0.57*Denominator is number of outbreaks reported in the preceding 12 monthsHF: Health Facility; IDSR: Integrated Disease Surveillance and Response; HCW: Healthcare worker; DHMT: District Health Management Team Comparison of IDSR indicators in selected health facilities, Sierra Leone, 2016–2021 *Denominator is number of outbreaks reported in the preceding 12 months HF: Health Facility; IDSR: Integrated Disease Surveillance and Response; HCW: Healthcare worker; DHMT: District Health Management Team [SUBTITLE] Identification of priority IDSR diseases, conditions and events [SUBSECTION] The proportion of health facilities that displayed standard case definition posters in consultation rooms for use by health workers in identifying cases increased significantly from 76.1% to 2016 to 94.9% in 2021 (increase of 18.8% points; 95% CI 9.2, 30.2); P- value 0.002). The proportion of IDSR focal persons who conducted weekly active case search for priority diseases in the health facilities remained the same at 79% for both baseline and end line although it fluctuated across the years. There was positive trend in adequate knowledge of standard case definitions among health workers for five priority diseases that were assessed as shown in Fig. 2 (Acute flaccid paralysis, Neonatal tetanus, Measles, Cholera and Ebola Virus Disease). Additionally, COVID-19 was added in 2021 and 61% of the health workers had adequate knowledge on its case definition. Fig. 2Knowledge of standard case definitions among interviewed healthcare workers, Sierra Leone, 2016 and 2021 Knowledge of standard case definitions among interviewed healthcare workers, Sierra Leone, 2016 and 2021 The proportion of health facilities that displayed standard case definition posters in consultation rooms for use by health workers in identifying cases increased significantly from 76.1% to 2016 to 94.9% in 2021 (increase of 18.8% points; 95% CI 9.2, 30.2); P- value 0.002). The proportion of IDSR focal persons who conducted weekly active case search for priority diseases in the health facilities remained the same at 79% for both baseline and end line although it fluctuated across the years. There was positive trend in adequate knowledge of standard case definitions among health workers for five priority diseases that were assessed as shown in Fig. 2 (Acute flaccid paralysis, Neonatal tetanus, Measles, Cholera and Ebola Virus Disease). Additionally, COVID-19 was added in 2021 and 61% of the health workers had adequate knowledge on its case definition. Fig. 2Knowledge of standard case definitions among interviewed healthcare workers, Sierra Leone, 2016 and 2021 Knowledge of standard case definitions among interviewed healthcare workers, Sierra Leone, 2016 and 2021 [SUBTITLE] Awareness and adherence to IDSR reporting requirements [SUBSECTION] Knowledge of IDSR reporting requirements was high and by the end line assessment in 2021, almost all interviewed health care workers correctly defined the epidemiologic week, zero reporting and reporting deadlines (Table 2). There was a significant improvement in the proportion of health facilities that submitted all the required surveillance reports (completeness of reporting) from 84.5% to 2016 to 96% in 2021(increase of 11.5% points; 95% CI 3.6, 21.9; P-value 0.003). During the same period, timeliness of IDSR reports improved from 80.3 to 92% (increase of 11.7% points; 95% CI 2.4, 22.9; P-value 0.01). Knowledge of IDSR reporting requirements was high and by the end line assessment in 2021, almost all interviewed health care workers correctly defined the epidemiologic week, zero reporting and reporting deadlines (Table 2). There was a significant improvement in the proportion of health facilities that submitted all the required surveillance reports (completeness of reporting) from 84.5% to 2016 to 96% in 2021(increase of 11.5% points; 95% CI 3.6, 21.9; P-value 0.003). During the same period, timeliness of IDSR reports improved from 80.3 to 92% (increase of 11.7% points; 95% CI 2.4, 22.9; P-value 0.01). [SUBTITLE] Availability of IDSR reporting tools [SUBSECTION] Availability of the weekly IDSR reporting tool improved from 88.7% to 2016 to 96.1% in 2021 (increase of 7.4% points; 95% CI 0.4, 17.1; P-value 0.03). Availability of other reporting tools did not change significantly and were at 78%, 89% and 62% in 2021 for line listing forms, case-based reporting forms and rumour logbooks, respectively (Table 2). Availability of the weekly IDSR reporting tool improved from 88.7% to 2016 to 96.1% in 2021 (increase of 7.4% points; 95% CI 0.4, 17.1; P-value 0.03). Availability of other reporting tools did not change significantly and were at 78%, 89% and 62% in 2021 for line listing forms, case-based reporting forms and rumour logbooks, respectively (Table 2). [SUBTITLE] Data analysis and use at health facility level [SUBSECTION] There was significant improvement in data analysis and use in health facilities over the years. The proportion of health facilities that conducted basic data analysis improved from 39% at baseline to 63% at end line (increase of 24% points; 95% CI 10.0, 36.7; P-value 0.0008). The proportion of health facilities with current line graphs showing trends in occurrence of priority diseases increased significantly from 7 to 46.1% (increase of 39.1% points; 95% CI 27.8, 47.9; P-value < 0.0001) (Table 2). There was significant improvement in data analysis and use in health facilities over the years. The proportion of health facilities that conducted basic data analysis improved from 39% at baseline to 63% at end line (increase of 24% points; 95% CI 10.0, 36.7; P-value 0.0008). The proportion of health facilities with current line graphs showing trends in occurrence of priority diseases increased significantly from 7 to 46.1% (increase of 39.1% points; 95% CI 27.8, 47.9; P-value < 0.0001) (Table 2). [SUBTITLE] Outbreak detection and notification [SUBSECTION] The proportion of health facilities that had identified an outbreak within 12 months of the assessment did not change significantly and was 16.9% at baseline compared to 10.9% at end line (P-value 0.31). This was mostly because there were no outbreaks to detect and not for lack of detection capacity. The proportion of identified outbreaks that were notified to the District Health Management Teams within 48 h did not change significantly and were 91.7% at baseline compared to 88.2% at end line (P-value 0.09) (Table 2). The proportion of health facilities that had identified an outbreak within 12 months of the assessment did not change significantly and was 16.9% at baseline compared to 10.9% at end line (P-value 0.31). This was mostly because there were no outbreaks to detect and not for lack of detection capacity. The proportion of identified outbreaks that were notified to the District Health Management Teams within 48 h did not change significantly and were 91.7% at baseline compared to 88.2% at end line (P-value 0.09) (Table 2). [SUBTITLE] Feedback, monitoring and communication [SUBSECTION] Quarterly supervisory visits by DHMTs to the health facilities improved from 62% at baseline to 92% (P-value < 000.1) at end line although it fluctuated for other visits (Table 2). Mobile network connectivity remained high throughout the period and 92% of the health facilities assessed at end line had connectivity. Quarterly supervisory visits by DHMTs to the health facilities improved from 62% at baseline to 92% (P-value < 000.1) at end line although it fluctuated for other visits (Table 2). Mobile network connectivity remained high throughout the period and 92% of the health facilities assessed at end line had connectivity. [SUBTITLE] Availability of routine immunization services and commodities for management of Malaria [SUBSECTION] The proportion of health facilities providing routine immunization services was above 90% in all the assessments with the end line being 96%. The number of immunizing health facilities with functional refrigerators significantly improved from 49.2% at baseline to 66.7% (P-value 0.02) although it fluctuated for other visits (Table 3). Immunizing health facilities with updated temperature monitoring charts significantly improved from 34 to 81% (P-value < 0.0001) while health facilities with all required basic antigens (vaccines) available at the time of the assessment improved from 48 to 83% (P-value < 0.0001). At the end line assessment, there were only 6.7% health facilities with Vaccine Vial Monitor (VVM) at stage three or four which is considered as excessive exposure to high temperatures (Table 3). Malaria management commodities including first line anti-malarial drugs, rapid diagnostic test kits and insecticide treated nets were available in most of the health facilities throughout the assessment period although there was a significant drop for first line anti-malarial drugs and rapid diagnostic test kits at end line assessment in August 2021, mostly due to disruptions associated with COVID-19 (Table 3). Table 3Assessing routine immunization, malaria commodities and diagnostic capacity in health facilities, Sierra Leone, 2016–2021Programmatic AreaVariableFeb_2016July_2016May_2017Aug_2018Feb_2019Aug_2021End Line vs. BaselineP-value(N = 71)n (%)(N = 99)n (%)(N = 101)n (%)(N = 126)n (%)(N = 139)n (%)(N = 156)n (%)Difference (95 CI) Vaccination Health facilities providing routine Immunization Services65 (91.5)95 (96.0)96 (95.0)120 (95.2)135 (97.1)150 (96.2)4.7 (-1.5, 13.7)0.14Health facilities with functional refrigerator†32 (49.2)70 (73.7)72 (75.0)71 (59.2)88 (65.2)100 (66.7)17.5 (3.3, 31.2) 0.0157 Health facilities with updated temperature monitoring chart††11 (34.4)16 (22.9)32 (44.4)29 (40.8)37 (42.0)81 (81)46.6 (27.2,62.0) < 0.0001 Health facilities with all required basic antigens available at the time of the assessment†31(47.7)55(57.9)60(62.5)86 (71.7)103 (76.3)125 (83.3)35.6 (21.9, 48.3) < 0.0001 Heath facilities with expired antigens†2 (3.1)3 (3.2)1 (1)1 (0.8)2 (1.5)2 (1.3)1.8 (-2.3, 9.3)0.37Health facilities with Vaccine Vial Monitor at stage three and four†14 (21.5)16 (16.8)14 (14.6)8 (6.7)9 (6.7)10 (6.7)14.8 (5.1, 26.6) 0.002 Malaria Commodities Health facilities with 1st line anti-malaria drugs69 (97.2)96 (97.0)99 (98.0)120 (95.2)135 (97.1)120 (76.9)20.3 (11.2, 27.7) 0.0002 Health Facilities with rapid diagnostic kits for diagnosis of malaria67 (94.4)89 (89.9)98 (97.0)120 (95.2)134 (96.4)119 (76.3)18.1 (8.1, 26.1) 0.001 Health facilities with insecticide treated nets55 (77.5)92 (92.9)97 (96.0)111 (88.1)132 (95.0)139 (89.1)11.6 (1.6, 23.3) 0.0218 †Denominator is number of health facilities providing immunization services†† Denominator is number of health facilities with a functional fridge Assessing routine immunization, malaria commodities and diagnostic capacity in health facilities, Sierra Leone, 2016–2021 †Denominator is number of health facilities providing immunization services †† Denominator is number of health facilities with a functional fridge The proportion of health facilities providing routine immunization services was above 90% in all the assessments with the end line being 96%. The number of immunizing health facilities with functional refrigerators significantly improved from 49.2% at baseline to 66.7% (P-value 0.02) although it fluctuated for other visits (Table 3). Immunizing health facilities with updated temperature monitoring charts significantly improved from 34 to 81% (P-value < 0.0001) while health facilities with all required basic antigens (vaccines) available at the time of the assessment improved from 48 to 83% (P-value < 0.0001). At the end line assessment, there were only 6.7% health facilities with Vaccine Vial Monitor (VVM) at stage three or four which is considered as excessive exposure to high temperatures (Table 3). Malaria management commodities including first line anti-malarial drugs, rapid diagnostic test kits and insecticide treated nets were available in most of the health facilities throughout the assessment period although there was a significant drop for first line anti-malarial drugs and rapid diagnostic test kits at end line assessment in August 2021, mostly due to disruptions associated with COVID-19 (Table 3). Table 3Assessing routine immunization, malaria commodities and diagnostic capacity in health facilities, Sierra Leone, 2016–2021Programmatic AreaVariableFeb_2016July_2016May_2017Aug_2018Feb_2019Aug_2021End Line vs. BaselineP-value(N = 71)n (%)(N = 99)n (%)(N = 101)n (%)(N = 126)n (%)(N = 139)n (%)(N = 156)n (%)Difference (95 CI) Vaccination Health facilities providing routine Immunization Services65 (91.5)95 (96.0)96 (95.0)120 (95.2)135 (97.1)150 (96.2)4.7 (-1.5, 13.7)0.14Health facilities with functional refrigerator†32 (49.2)70 (73.7)72 (75.0)71 (59.2)88 (65.2)100 (66.7)17.5 (3.3, 31.2) 0.0157 Health facilities with updated temperature monitoring chart††11 (34.4)16 (22.9)32 (44.4)29 (40.8)37 (42.0)81 (81)46.6 (27.2,62.0) < 0.0001 Health facilities with all required basic antigens available at the time of the assessment†31(47.7)55(57.9)60(62.5)86 (71.7)103 (76.3)125 (83.3)35.6 (21.9, 48.3) < 0.0001 Heath facilities with expired antigens†2 (3.1)3 (3.2)1 (1)1 (0.8)2 (1.5)2 (1.3)1.8 (-2.3, 9.3)0.37Health facilities with Vaccine Vial Monitor at stage three and four†14 (21.5)16 (16.8)14 (14.6)8 (6.7)9 (6.7)10 (6.7)14.8 (5.1, 26.6) 0.002 Malaria Commodities Health facilities with 1st line anti-malaria drugs69 (97.2)96 (97.0)99 (98.0)120 (95.2)135 (97.1)120 (76.9)20.3 (11.2, 27.7) 0.0002 Health Facilities with rapid diagnostic kits for diagnosis of malaria67 (94.4)89 (89.9)98 (97.0)120 (95.2)134 (96.4)119 (76.3)18.1 (8.1, 26.1) 0.001 Health facilities with insecticide treated nets55 (77.5)92 (92.9)97 (96.0)111 (88.1)132 (95.0)139 (89.1)11.6 (1.6, 23.3) 0.0218 †Denominator is number of health facilities providing immunization services†† Denominator is number of health facilities with a functional fridge Assessing routine immunization, malaria commodities and diagnostic capacity in health facilities, Sierra Leone, 2016–2021 †Denominator is number of health facilities providing immunization services †† Denominator is number of health facilities with a functional fridge
Conclusion
The IDSR system is now well established in Sierra Leone. The support supervision visits that were done using an integrated electronic tool contributed to health systems strengthening through longitudinal tracking of core IDSR indicators and other program indicators such as essential malaria commodities and availability and status of routine vaccines. The feedback that was provided to all levels of healthcare including the national program and development partners supported to address the gaps identified and hence improved performance over the years. MoHS should entrench the supportive supervision approach by the national and district teams using electronic tools to assure sustained monitoring of IDSR and other programs.
[ "Background", "The effect on Ebola Virus Disease outbreak on the health workforce in Sierra Leone", "Improving public health surveillance after the Ebola outbreak", "Methods", "Study setting and design", "Selection of health facilities", "Data collection", "Data analysis", "Coordination of IDSR activities in health facilities", "Identification of priority IDSR diseases, conditions and events", "Awareness and adherence to IDSR reporting requirements", "Availability of IDSR reporting tools", "Data analysis and use at health facility level", "Outbreak detection and notification", "Feedback, monitoring and communication", "Availability of routine immunization services and commodities for management of Malaria" ]
[ "Supportive supervision is a unique approach to monitoring performance through face to face interactions between a skilled health care worker and a less skilled worker [1]. First promoted in the era of primary health care, supportive supervision sought to improve performance of healthcare workers offering essential services such as immunization in remote areas [2]. However, this approach can be impeded by poor coordination, lack of motivation, geographical barriers, competing activities, high costs and armed conflicts [1, 3].\nInitially, a top-down approach to supervision was used with inspection and correction of junior workers done by a senior officer. There has been a shift to a more collaborative approach (support supervision) with focus on identifying challenges in service delivery and involving the provider in finding solutions. This approach improves motivation and ownership by the health care providers [4]. Records reviews, direct observations, use of objective indicators, collaborative problem solving and targeted on job training are additional collaborative approaches used [1, 5].\nIn 1998, the World Health Organization (WHO), Africa Regional Office adopted the Integrated Disease Surveillance (IDS) strategy, later renamed Integrated Disease Surveillance and Response (IDSR) strategy to strengthen public health surveillance and response in member states [6]. Implementation of the IDSR strategy varies, with the initial momentum exhibited in the first years of implementation declining over time due to several challenges such as non-sustainable financial resources, poor coordination, inadequate training, high turnover of staff, inadequate supervision from the next level, erratic feedback, weak laboratory capacities, unavailability of job aids and poor availability of communication and transport systems particularly at the periphery [7–9]. Considering the growing threat of epidemics especially from new and emerging pathogens, the need for strengthened surveillance systems has never been greater. Ministries of Health in WHO member states are urged to monitor implementation of IDSR as this directly contributes to fulfillment of the International Health Regulations (2005) [10]. National level health staff utilize supervision visits to monitor the implementation of Integrated Disease Surveillance and Response (IDSR) activities and identify key areas that need improvement [11]. While training of sub- national health care workers on IDSR provides them with basic understanding of public health surveillance, supportive supervision reinforces concepts, and identifies and resolves challenges affecting implementation [12].\n[SUBTITLE] The effect on Ebola Virus Disease outbreak on the health workforce in Sierra Leone [SUBSECTION] Health care workers are a high-risk group for highly infectious diseases such as Ebola Virus Disease (EVD). Out of 304 health care workers infected with Ebola virus in Sierra Leone in 2014–2016, two hundred and twenty-one (221) died and several others resigned out of fear of getting infected. This reduced the ratio of skilled health workers from 17.2/ 10,000 to < 4 /10,000 population [13–15]. After the outbreak, the Ministry of Health and Sanitation (MoHS) embarked on strengthening health systems, improving public health surveillance and cross border surveillance [14].\nHealth care workers are a high-risk group for highly infectious diseases such as Ebola Virus Disease (EVD). Out of 304 health care workers infected with Ebola virus in Sierra Leone in 2014–2016, two hundred and twenty-one (221) died and several others resigned out of fear of getting infected. This reduced the ratio of skilled health workers from 17.2/ 10,000 to < 4 /10,000 population [13–15]. After the outbreak, the Ministry of Health and Sanitation (MoHS) embarked on strengthening health systems, improving public health surveillance and cross border surveillance [14].\n[SUBTITLE] Improving public health surveillance after the Ebola outbreak [SUBSECTION] Towards the end of the Ebola Virus Disease outbreak, the Sierra Leone Ministry of Health and Sanitation in collaboration with World Health Organization country office (WCO) and other health sector development partners, embarked on revitalization of IDSR. This involved adaptation of the 2010 WHO-Africa region IDSR technical guidelines and training modules, training of healthcare workers and providing inputs and infrastructure to support IDSR implementation [16].\n\nPreviously, paper-based checklists were used during supportive supervision in Sierra Leone and health facilities were selected purposively based on ease of access. Thus, it was likely that health facilities in remote areas were omitted from supervisory visits. Collation and analysis of data from the supervisory visits was seldom done as it required abstraction of data from paper-based checklists to a computer and this often required hiring of data clerks due to staff shortage. To overcome these issues, we adopted an electronic supervision checklist and an open source platform to collect and manage data. This paper describes the innovative, integrated approach to IDSR supportive supervision. We share the lessons learnt, hopeful that they can provide insight for improving monitoring of public health surveillance programs particularly for countries implementing IDSR.\n\nThe integrated support supervision process described in this paper is considered an innovative way of monitoring IDSR performance indicators for several reasons. First, routine supervision visits were used to collect comprehensive and representative data to monitor key IDSR indicators from 2016 to 2021. During the visits, verbal and written feedback on performance was then given to the health facilities and district health management teams which ensured that corrective action was taken where necessary. Second, an integrated questionnaire was used which not only collected surveillance data, but also data from other related programs such as laboratory, immunization and malaria. Third, the use of electronic supervision checklist and an open source data collection platform to collect and manage the data ensured secure storage and rapid retrieval of data for analysis which enabled monitoring of performance over time.\nTowards the end of the Ebola Virus Disease outbreak, the Sierra Leone Ministry of Health and Sanitation in collaboration with World Health Organization country office (WCO) and other health sector development partners, embarked on revitalization of IDSR. This involved adaptation of the 2010 WHO-Africa region IDSR technical guidelines and training modules, training of healthcare workers and providing inputs and infrastructure to support IDSR implementation [16].\n\nPreviously, paper-based checklists were used during supportive supervision in Sierra Leone and health facilities were selected purposively based on ease of access. Thus, it was likely that health facilities in remote areas were omitted from supervisory visits. Collation and analysis of data from the supervisory visits was seldom done as it required abstraction of data from paper-based checklists to a computer and this often required hiring of data clerks due to staff shortage. To overcome these issues, we adopted an electronic supervision checklist and an open source platform to collect and manage data. This paper describes the innovative, integrated approach to IDSR supportive supervision. We share the lessons learnt, hopeful that they can provide insight for improving monitoring of public health surveillance programs particularly for countries implementing IDSR.\n\nThe integrated support supervision process described in this paper is considered an innovative way of monitoring IDSR performance indicators for several reasons. First, routine supervision visits were used to collect comprehensive and representative data to monitor key IDSR indicators from 2016 to 2021. During the visits, verbal and written feedback on performance was then given to the health facilities and district health management teams which ensured that corrective action was taken where necessary. Second, an integrated questionnaire was used which not only collected surveillance data, but also data from other related programs such as laboratory, immunization and malaria. Third, the use of electronic supervision checklist and an open source data collection platform to collect and manage the data ensured secure storage and rapid retrieval of data for analysis which enabled monitoring of performance over time.", "Health care workers are a high-risk group for highly infectious diseases such as Ebola Virus Disease (EVD). Out of 304 health care workers infected with Ebola virus in Sierra Leone in 2014–2016, two hundred and twenty-one (221) died and several others resigned out of fear of getting infected. This reduced the ratio of skilled health workers from 17.2/ 10,000 to < 4 /10,000 population [13–15]. After the outbreak, the Ministry of Health and Sanitation (MoHS) embarked on strengthening health systems, improving public health surveillance and cross border surveillance [14].", "Towards the end of the Ebola Virus Disease outbreak, the Sierra Leone Ministry of Health and Sanitation in collaboration with World Health Organization country office (WCO) and other health sector development partners, embarked on revitalization of IDSR. This involved adaptation of the 2010 WHO-Africa region IDSR technical guidelines and training modules, training of healthcare workers and providing inputs and infrastructure to support IDSR implementation [16].\n\nPreviously, paper-based checklists were used during supportive supervision in Sierra Leone and health facilities were selected purposively based on ease of access. Thus, it was likely that health facilities in remote areas were omitted from supervisory visits. Collation and analysis of data from the supervisory visits was seldom done as it required abstraction of data from paper-based checklists to a computer and this often required hiring of data clerks due to staff shortage. To overcome these issues, we adopted an electronic supervision checklist and an open source platform to collect and manage data. This paper describes the innovative, integrated approach to IDSR supportive supervision. We share the lessons learnt, hopeful that they can provide insight for improving monitoring of public health surveillance programs particularly for countries implementing IDSR.\n\nThe integrated support supervision process described in this paper is considered an innovative way of monitoring IDSR performance indicators for several reasons. First, routine supervision visits were used to collect comprehensive and representative data to monitor key IDSR indicators from 2016 to 2021. During the visits, verbal and written feedback on performance was then given to the health facilities and district health management teams which ensured that corrective action was taken where necessary. Second, an integrated questionnaire was used which not only collected surveillance data, but also data from other related programs such as laboratory, immunization and malaria. Third, the use of electronic supervision checklist and an open source data collection platform to collect and manage the data ensured secure storage and rapid retrieval of data for analysis which enabled monitoring of performance over time.", "[SUBTITLE] Study setting and design [SUBSECTION] This cross-sectional study was part of operational research to monitor implementation of IDSR program in Sierra Leone post Ebola outbreak. Data was collected through supportive supervision visits to health facilities across all districts in Sierra Leone from 2016 to 2021. The data collected was used to monitor and improve IDSR implementation in the country.\nThis cross-sectional study was part of operational research to monitor implementation of IDSR program in Sierra Leone post Ebola outbreak. Data was collected through supportive supervision visits to health facilities across all districts in Sierra Leone from 2016 to 2021. The data collected was used to monitor and improve IDSR implementation in the country.\n[SUBTITLE] Selection of health facilities [SUBSECTION] The total number of health facilities sampled nationally (sample size) per each support supervision visit was purposively selected based on available resources. There were about 1425 health facilities in the country in 2021, distributed by type as follows, in order of size and scope of services offered (from largest to smallest): Hospital 4%, Community Health Centres (CHC) 19%, Community Health Posts (CHP) 30%, Maternal and Child Health Posts (MCHP) 43% and Clinic 3%. About 10% of the total existing facilities per year was sampled in each visit. To ensure representativeness, health facilities were selected from each type of health facility (except clinics which are mostly private) in all the districts. Selection of health facilities in each district was done randomly from a list of health facilities stratified by health facility type as indicated above. First, health facilities were stratified according to the type of health facility. Thereafter, random sampling, using computer generated numbers was done to identify at least one hospital, three CHC, three CHP and three MCHP in each district. Where available, major private health facilities equivalent to a hospital were included.\nOver the six-year period, six national level supportive supervision visits were conducted covering 71 health facilities in February 2016, 99 in July 2016, 101 in May 2017, 126 in August 2018, 139 in February 2019 and 156 in August 2021 (Fig. 1). No national level supervisory visit was carried out in 2020 due to COVID-19 pandemic. Apart from the national supervision visits, the District Health Management Teams conducted two to four supervision visits per year for the facilities under their jurisdiction. However, this data was captured in a separate database and is therefore not included in this article.\n\nFig. 1Map of Sierra Leone showing health facilities visited during support supervision, 2016–2021\n\nMap of Sierra Leone showing health facilities visited during support supervision, 2016–2021\nThe total number of health facilities sampled nationally (sample size) per each support supervision visit was purposively selected based on available resources. There were about 1425 health facilities in the country in 2021, distributed by type as follows, in order of size and scope of services offered (from largest to smallest): Hospital 4%, Community Health Centres (CHC) 19%, Community Health Posts (CHP) 30%, Maternal and Child Health Posts (MCHP) 43% and Clinic 3%. About 10% of the total existing facilities per year was sampled in each visit. To ensure representativeness, health facilities were selected from each type of health facility (except clinics which are mostly private) in all the districts. Selection of health facilities in each district was done randomly from a list of health facilities stratified by health facility type as indicated above. First, health facilities were stratified according to the type of health facility. Thereafter, random sampling, using computer generated numbers was done to identify at least one hospital, three CHC, three CHP and three MCHP in each district. Where available, major private health facilities equivalent to a hospital were included.\nOver the six-year period, six national level supportive supervision visits were conducted covering 71 health facilities in February 2016, 99 in July 2016, 101 in May 2017, 126 in August 2018, 139 in February 2019 and 156 in August 2021 (Fig. 1). No national level supervisory visit was carried out in 2020 due to COVID-19 pandemic. Apart from the national supervision visits, the District Health Management Teams conducted two to four supervision visits per year for the facilities under their jurisdiction. However, this data was captured in a separate database and is therefore not included in this article.\n\nFig. 1Map of Sierra Leone showing health facilities visited during support supervision, 2016–2021\n\nMap of Sierra Leone showing health facilities visited during support supervision, 2016–2021\n[SUBTITLE] Data collection [SUBSECTION] The assessment teams comprised of national level staff from the surveillance and laboratory programs of MOHS as well as development partners like World Health Organization who were supporting the supervision missions technically and financially. Respondents in the assessments were a team of healthcare workers involved in surveillance and administrative functions in the health facilities. These included the IDSR focal person, health facility in charge, laboratory technician and the nurse in charge of vaccination activities.\nOne week prior to each supportive supervisory visit, the MoHS through the National Disease Surveillance Program contacted the District Medical Officers (DMO) and District Surveillance Officers (DSOs) to inform them of the upcoming supervisory visits. On the first day of the visit, the supervisory team comprising of officials from MoHS and supporting development partners met with the District Health Management Team (DHMT) members to discuss the purpose of the supervisory visits and to select health facilities for the assessment. The selected health facilities were not informed of the visit in advance in order to avoid pre-emptive interference with data tools. Upon reaching a health facility, the supervisory team captured the geographic coordinates of the health facilities using automatically generated geo- coordinates. Recording of geo-coordinates for each facility ensured that there was evidence of representativeness in the distribution of selected health facilities. Next the team conducted interviews, verified documents and confirmed availability of IDSR reporting tools.\nData was collected in real-time using a structured electronic questionnaire created using Open Data Kit platform and loaded onto tablets. Using the electronic tool eased data collection and management, making it possible to analyse data, provide timely feedback to the health facilities staff, and also track performance over time. The questionnaire covered six out of eight core functions of IDSR as well as an assessment on related programmatic areas such as available laboratory services, maternal deaths, malaria commodities and routine immunization services. The questionnaire collected information on (1) case identification; (2) reporting of priority diseases; (3) data analysis and interpretation; (4) investigation and confirmation of reported cases and outbreaks; (5) feedback between the district management teams and health facilities; and (6) monitoring and evaluation (Table 1).\n\nTable 1IDSR Core Functions Assessed During Integrated Support Supervision, Sierra Leone, 2016–2021Core FunctionAssessment methodsMain focus areas1Identification of priority diseases, conditions and eventsInterview, observation, review of registersAssessed the ability of healthcare workers to use standard case definition to identify priority diseases through records review and active case search in the community2Reporting of Priority diseasesInterview, documents review, observationChecked if HCWs are aware of reporting requirements for priority diseases and the availability and use of reporting tools3Data analysis and interpretationInterview and observationChecked if data analysis and interpretation was done at the facility4Investigation and confirmation of outbreaksInterviewAssessed if HF are able to detect outbreaks from various sources, notify the district and participate in outbreak investigations5FeedbackInterviewConfirmed if the HF gets feedback on IDSR performance from the DHMT6Monitoring and EvaluationInterview, document reviewAssessed the frequency of supervision done by the DHMT and mechanisms for monitoring IDSR performance in the HFIDSR: Integrated Disease Surveillance and Response; HCW: Healthcare workers; HF: Health Facility; DHMT: District Health Management Team\n\nIDSR Core Functions Assessed During Integrated Support Supervision, Sierra Leone, 2016–2021\nIDSR: Integrated Disease Surveillance and Response; HCW: Healthcare workers; HF: Health Facility; DHMT: District Health Management Team\nThe teams began by assessing availability of IDSR focal persons in the health facilities and their participation in district monthly review meetings. This is because the focal persons are important in coordinating IDSR activities. On case identification, supervisors checked if IDSR posters with case definitions were available and displayed in the consultation rooms. They then interviewed the respondents to gauge their knowledge of case definitions for five selected IDSR priority diseases/conditions (Acute flaccid paralysis, Neonatal tetanus, Measles, Cholera and Ebola Virus Disease). Additionally, COVID-19 was added in the 2021 assessment. A healthcare worker who mentioned all signs and symptoms correctly was classified as having adequate knowledge, while one who mentioned some of the signs and symptoms was classified as having limited knowledge. A healthcare worker who did not mention any of the appropriate signs and symptoms was classified as having no knowledge.\nThe section on reporting of priority diseases assessed knowledge of and compliance with reporting requirements for priority diseases. Copies of IDSR weekly reports for the previous four weeks were checked to confirm if all reports were available and if they had been submitted on time. The team also checked the availability of case-based reporting forms, weekly reporting forms, line listing forms, and rumour logbooks for recording suspected outbreaks/events. The section on data analysis assessed the capacity and type of data analysis conducted to monitor trends of priority diseases at the health facilities. The section on investigation captured information on case investigation and confirmation of reported outbreaks. The section on feedback assessed feedback mechanisms from the district to the health facilities and vice versa. It included a review of previous supervision reports to elicit frequency of supportive supervision by DHMT. The section on monitoring and evaluation assessed mechanisms for monitoring IDSR performance in the health facility.\nAdditional questions on immunization, malaria treatment commodities and maternal mortality were also included in the questionnaire. The questionnaire had compulsory fields and intrinsic data validation mechanisms to enhance data accuracy and completeness. Finally, at the end of the data collection process, the teams used supervision books available in each health facility to record strengths, gaps, challenges and recommendations agreed upon during the supervisory visit. These records were useful for follow up during subsequent supervisory visits. Supervision also provided an opportunity for on the job training and distribution of IDSR reference materials.\nThe assessment teams comprised of national level staff from the surveillance and laboratory programs of MOHS as well as development partners like World Health Organization who were supporting the supervision missions technically and financially. Respondents in the assessments were a team of healthcare workers involved in surveillance and administrative functions in the health facilities. These included the IDSR focal person, health facility in charge, laboratory technician and the nurse in charge of vaccination activities.\nOne week prior to each supportive supervisory visit, the MoHS through the National Disease Surveillance Program contacted the District Medical Officers (DMO) and District Surveillance Officers (DSOs) to inform them of the upcoming supervisory visits. On the first day of the visit, the supervisory team comprising of officials from MoHS and supporting development partners met with the District Health Management Team (DHMT) members to discuss the purpose of the supervisory visits and to select health facilities for the assessment. The selected health facilities were not informed of the visit in advance in order to avoid pre-emptive interference with data tools. Upon reaching a health facility, the supervisory team captured the geographic coordinates of the health facilities using automatically generated geo- coordinates. Recording of geo-coordinates for each facility ensured that there was evidence of representativeness in the distribution of selected health facilities. Next the team conducted interviews, verified documents and confirmed availability of IDSR reporting tools.\nData was collected in real-time using a structured electronic questionnaire created using Open Data Kit platform and loaded onto tablets. Using the electronic tool eased data collection and management, making it possible to analyse data, provide timely feedback to the health facilities staff, and also track performance over time. The questionnaire covered six out of eight core functions of IDSR as well as an assessment on related programmatic areas such as available laboratory services, maternal deaths, malaria commodities and routine immunization services. The questionnaire collected information on (1) case identification; (2) reporting of priority diseases; (3) data analysis and interpretation; (4) investigation and confirmation of reported cases and outbreaks; (5) feedback between the district management teams and health facilities; and (6) monitoring and evaluation (Table 1).\n\nTable 1IDSR Core Functions Assessed During Integrated Support Supervision, Sierra Leone, 2016–2021Core FunctionAssessment methodsMain focus areas1Identification of priority diseases, conditions and eventsInterview, observation, review of registersAssessed the ability of healthcare workers to use standard case definition to identify priority diseases through records review and active case search in the community2Reporting of Priority diseasesInterview, documents review, observationChecked if HCWs are aware of reporting requirements for priority diseases and the availability and use of reporting tools3Data analysis and interpretationInterview and observationChecked if data analysis and interpretation was done at the facility4Investigation and confirmation of outbreaksInterviewAssessed if HF are able to detect outbreaks from various sources, notify the district and participate in outbreak investigations5FeedbackInterviewConfirmed if the HF gets feedback on IDSR performance from the DHMT6Monitoring and EvaluationInterview, document reviewAssessed the frequency of supervision done by the DHMT and mechanisms for monitoring IDSR performance in the HFIDSR: Integrated Disease Surveillance and Response; HCW: Healthcare workers; HF: Health Facility; DHMT: District Health Management Team\n\nIDSR Core Functions Assessed During Integrated Support Supervision, Sierra Leone, 2016–2021\nIDSR: Integrated Disease Surveillance and Response; HCW: Healthcare workers; HF: Health Facility; DHMT: District Health Management Team\nThe teams began by assessing availability of IDSR focal persons in the health facilities and their participation in district monthly review meetings. This is because the focal persons are important in coordinating IDSR activities. On case identification, supervisors checked if IDSR posters with case definitions were available and displayed in the consultation rooms. They then interviewed the respondents to gauge their knowledge of case definitions for five selected IDSR priority diseases/conditions (Acute flaccid paralysis, Neonatal tetanus, Measles, Cholera and Ebola Virus Disease). Additionally, COVID-19 was added in the 2021 assessment. A healthcare worker who mentioned all signs and symptoms correctly was classified as having adequate knowledge, while one who mentioned some of the signs and symptoms was classified as having limited knowledge. A healthcare worker who did not mention any of the appropriate signs and symptoms was classified as having no knowledge.\nThe section on reporting of priority diseases assessed knowledge of and compliance with reporting requirements for priority diseases. Copies of IDSR weekly reports for the previous four weeks were checked to confirm if all reports were available and if they had been submitted on time. The team also checked the availability of case-based reporting forms, weekly reporting forms, line listing forms, and rumour logbooks for recording suspected outbreaks/events. The section on data analysis assessed the capacity and type of data analysis conducted to monitor trends of priority diseases at the health facilities. The section on investigation captured information on case investigation and confirmation of reported outbreaks. The section on feedback assessed feedback mechanisms from the district to the health facilities and vice versa. It included a review of previous supervision reports to elicit frequency of supportive supervision by DHMT. The section on monitoring and evaluation assessed mechanisms for monitoring IDSR performance in the health facility.\nAdditional questions on immunization, malaria treatment commodities and maternal mortality were also included in the questionnaire. The questionnaire had compulsory fields and intrinsic data validation mechanisms to enhance data accuracy and completeness. Finally, at the end of the data collection process, the teams used supervision books available in each health facility to record strengths, gaps, challenges and recommendations agreed upon during the supervisory visit. These records were useful for follow up during subsequent supervisory visits. Supervision also provided an opportunity for on the job training and distribution of IDSR reference materials.\n[SUBTITLE] Data analysis [SUBSECTION] Data was extracted from the Open Data Kit (ODK) platform first onto an MS Excel database, merged, checked for duplicates and completeness and then exported to Epi Info for analysis. We calculated means and medians for continuous variables and proportions for categorical data. Two proportion Z-test was used to compare differences in performance recorded during the baseline in February 2016 and the end line in August 2021. District weekly report completeness was defined as the proportion of health facilities that submitted required reports to the district health office. This was verified by checking if all copies of the IDSR weekly reports for the preceding four weeks were available. Timeliness was defined as the proportion of health facilities that submitted IDSR weekly reports to the district by 12 pm every Monday.\nData was extracted from the Open Data Kit (ODK) platform first onto an MS Excel database, merged, checked for duplicates and completeness and then exported to Epi Info for analysis. We calculated means and medians for continuous variables and proportions for categorical data. Two proportion Z-test was used to compare differences in performance recorded during the baseline in February 2016 and the end line in August 2021. District weekly report completeness was defined as the proportion of health facilities that submitted required reports to the district health office. This was verified by checking if all copies of the IDSR weekly reports for the preceding four weeks were available. Timeliness was defined as the proportion of health facilities that submitted IDSR weekly reports to the district by 12 pm every Monday.", "This cross-sectional study was part of operational research to monitor implementation of IDSR program in Sierra Leone post Ebola outbreak. Data was collected through supportive supervision visits to health facilities across all districts in Sierra Leone from 2016 to 2021. The data collected was used to monitor and improve IDSR implementation in the country.", "The total number of health facilities sampled nationally (sample size) per each support supervision visit was purposively selected based on available resources. There were about 1425 health facilities in the country in 2021, distributed by type as follows, in order of size and scope of services offered (from largest to smallest): Hospital 4%, Community Health Centres (CHC) 19%, Community Health Posts (CHP) 30%, Maternal and Child Health Posts (MCHP) 43% and Clinic 3%. About 10% of the total existing facilities per year was sampled in each visit. To ensure representativeness, health facilities were selected from each type of health facility (except clinics which are mostly private) in all the districts. Selection of health facilities in each district was done randomly from a list of health facilities stratified by health facility type as indicated above. First, health facilities were stratified according to the type of health facility. Thereafter, random sampling, using computer generated numbers was done to identify at least one hospital, three CHC, three CHP and three MCHP in each district. Where available, major private health facilities equivalent to a hospital were included.\nOver the six-year period, six national level supportive supervision visits were conducted covering 71 health facilities in February 2016, 99 in July 2016, 101 in May 2017, 126 in August 2018, 139 in February 2019 and 156 in August 2021 (Fig. 1). No national level supervisory visit was carried out in 2020 due to COVID-19 pandemic. Apart from the national supervision visits, the District Health Management Teams conducted two to four supervision visits per year for the facilities under their jurisdiction. However, this data was captured in a separate database and is therefore not included in this article.\n\nFig. 1Map of Sierra Leone showing health facilities visited during support supervision, 2016–2021\n\nMap of Sierra Leone showing health facilities visited during support supervision, 2016–2021", "The assessment teams comprised of national level staff from the surveillance and laboratory programs of MOHS as well as development partners like World Health Organization who were supporting the supervision missions technically and financially. Respondents in the assessments were a team of healthcare workers involved in surveillance and administrative functions in the health facilities. These included the IDSR focal person, health facility in charge, laboratory technician and the nurse in charge of vaccination activities.\nOne week prior to each supportive supervisory visit, the MoHS through the National Disease Surveillance Program contacted the District Medical Officers (DMO) and District Surveillance Officers (DSOs) to inform them of the upcoming supervisory visits. On the first day of the visit, the supervisory team comprising of officials from MoHS and supporting development partners met with the District Health Management Team (DHMT) members to discuss the purpose of the supervisory visits and to select health facilities for the assessment. The selected health facilities were not informed of the visit in advance in order to avoid pre-emptive interference with data tools. Upon reaching a health facility, the supervisory team captured the geographic coordinates of the health facilities using automatically generated geo- coordinates. Recording of geo-coordinates for each facility ensured that there was evidence of representativeness in the distribution of selected health facilities. Next the team conducted interviews, verified documents and confirmed availability of IDSR reporting tools.\nData was collected in real-time using a structured electronic questionnaire created using Open Data Kit platform and loaded onto tablets. Using the electronic tool eased data collection and management, making it possible to analyse data, provide timely feedback to the health facilities staff, and also track performance over time. The questionnaire covered six out of eight core functions of IDSR as well as an assessment on related programmatic areas such as available laboratory services, maternal deaths, malaria commodities and routine immunization services. The questionnaire collected information on (1) case identification; (2) reporting of priority diseases; (3) data analysis and interpretation; (4) investigation and confirmation of reported cases and outbreaks; (5) feedback between the district management teams and health facilities; and (6) monitoring and evaluation (Table 1).\n\nTable 1IDSR Core Functions Assessed During Integrated Support Supervision, Sierra Leone, 2016–2021Core FunctionAssessment methodsMain focus areas1Identification of priority diseases, conditions and eventsInterview, observation, review of registersAssessed the ability of healthcare workers to use standard case definition to identify priority diseases through records review and active case search in the community2Reporting of Priority diseasesInterview, documents review, observationChecked if HCWs are aware of reporting requirements for priority diseases and the availability and use of reporting tools3Data analysis and interpretationInterview and observationChecked if data analysis and interpretation was done at the facility4Investigation and confirmation of outbreaksInterviewAssessed if HF are able to detect outbreaks from various sources, notify the district and participate in outbreak investigations5FeedbackInterviewConfirmed if the HF gets feedback on IDSR performance from the DHMT6Monitoring and EvaluationInterview, document reviewAssessed the frequency of supervision done by the DHMT and mechanisms for monitoring IDSR performance in the HFIDSR: Integrated Disease Surveillance and Response; HCW: Healthcare workers; HF: Health Facility; DHMT: District Health Management Team\n\nIDSR Core Functions Assessed During Integrated Support Supervision, Sierra Leone, 2016–2021\nIDSR: Integrated Disease Surveillance and Response; HCW: Healthcare workers; HF: Health Facility; DHMT: District Health Management Team\nThe teams began by assessing availability of IDSR focal persons in the health facilities and their participation in district monthly review meetings. This is because the focal persons are important in coordinating IDSR activities. On case identification, supervisors checked if IDSR posters with case definitions were available and displayed in the consultation rooms. They then interviewed the respondents to gauge their knowledge of case definitions for five selected IDSR priority diseases/conditions (Acute flaccid paralysis, Neonatal tetanus, Measles, Cholera and Ebola Virus Disease). Additionally, COVID-19 was added in the 2021 assessment. A healthcare worker who mentioned all signs and symptoms correctly was classified as having adequate knowledge, while one who mentioned some of the signs and symptoms was classified as having limited knowledge. A healthcare worker who did not mention any of the appropriate signs and symptoms was classified as having no knowledge.\nThe section on reporting of priority diseases assessed knowledge of and compliance with reporting requirements for priority diseases. Copies of IDSR weekly reports for the previous four weeks were checked to confirm if all reports were available and if they had been submitted on time. The team also checked the availability of case-based reporting forms, weekly reporting forms, line listing forms, and rumour logbooks for recording suspected outbreaks/events. The section on data analysis assessed the capacity and type of data analysis conducted to monitor trends of priority diseases at the health facilities. The section on investigation captured information on case investigation and confirmation of reported outbreaks. The section on feedback assessed feedback mechanisms from the district to the health facilities and vice versa. It included a review of previous supervision reports to elicit frequency of supportive supervision by DHMT. The section on monitoring and evaluation assessed mechanisms for monitoring IDSR performance in the health facility.\nAdditional questions on immunization, malaria treatment commodities and maternal mortality were also included in the questionnaire. The questionnaire had compulsory fields and intrinsic data validation mechanisms to enhance data accuracy and completeness. Finally, at the end of the data collection process, the teams used supervision books available in each health facility to record strengths, gaps, challenges and recommendations agreed upon during the supervisory visit. These records were useful for follow up during subsequent supervisory visits. Supervision also provided an opportunity for on the job training and distribution of IDSR reference materials.", "Data was extracted from the Open Data Kit (ODK) platform first onto an MS Excel database, merged, checked for duplicates and completeness and then exported to Epi Info for analysis. We calculated means and medians for continuous variables and proportions for categorical data. Two proportion Z-test was used to compare differences in performance recorded during the baseline in February 2016 and the end line in August 2021. District weekly report completeness was defined as the proportion of health facilities that submitted required reports to the district health office. This was verified by checking if all copies of the IDSR weekly reports for the preceding four weeks were available. Timeliness was defined as the proportion of health facilities that submitted IDSR weekly reports to the district by 12 pm every Monday.", "IDSR focal persons were present in most health facilities during all assessments. When compared to the baseline assessment, the proportion of IDSR focal persons who participated in the monthly district management team meetings increased significantly from 74.6% to 2016 (baseline) to 96.1% at end line in 2021 (increase of 21.5% points; 95% CI (11.8, 32.9); P-value < 0.0001). Conversely, availability of IDSR technical guidelines at the health facilities declined from 97.2% to 2016 to 82.1% in 2021 (decrease of 15.1% points; 95% CI (6.5, 22.2); P-value 0.002) (Table 2). The decline in availability of IDSR technical guidelines was mainly due to loss without replacement.\n\nTable 2Comparison of IDSR indicators in selected health facilities, Sierra Leone, 2016–2021Core FunctionIndicatorsFeb_2016July_2016May_2017Aug_2018Feb_2019Aug_2021End Line vs. BaselineP valueN = 71n (%)N = 99n (%)N = 101n (%)N = 126n (%)N = 139n (%)N = 156n (%)Difference (95 CI)CoordinationHealth facility (HF) has IDSR focal person69 (97.2)97 (98.0)100 (99.0)121 (96.0)135 (97.1)153 (98.1)0.9 (-3.2, 7.9)0.67HF IDSR focal person attends district monthly meetings53 (74.6)87 (87.9)93 (92.1)118 (93.7)132 (95.0)150 (96.1)21.5 (11.8, 32.9)\n< 0.0001\nHF has IDSR Technical guidelines69 (97.2)89 (89.9)101 (100.0)102 (81.0)123 (88.5)128 (82.1)15.1 (6.5, 22.2)\n0.002\nCase IdentificationStandard Case definition poster displayed in at least one location in the health facility54 (76.1)84 (84.8)89 (88.1)114 (90.5)127 (91.4)148 (94.9)18.8 (9.2, 30.2)\n< 0.0001\nIDSR focal person conducts active case search at least once a week56 (78.9)54 (54.5)34 (33.7)100 (79.4)75 (54.0)123 (78.8)0.1 (-12.1, 10.7)0.99Reporting of Priority diseasesHCW correctly defines epidemiologic week51 (71.8)93 (93.9)99 (98.0)108 (85.7)132 (95)151 (96.8)25 (15.0, 36.5)\n< 0.0001\nHCW correctly defines zero reporting67 (94.4)94 (94.9)99 (98.0)121 (96.0)138 (99.3)147 (92.2)2.2 (-6.4, 8.5))0.55HCW knows the deadline for submitting weekly IDSR reports65 (91.5)96 (97.0)100 (99.0)115 (91.3)138 (99.3)151 (96.8)5.3 (-0.8, 14.3)0.09Weekly reporting forms available in HF63 (88.7)89 (89.9)96 (95.0)119 (94.4)127 (91.4)150 (96.1)7.4 (0.4, 17.1)\n0.03\nLine listing forms available in HF60 (84.5)78 (78.8)81 (80.2)95 (75.4)114 (82.0)122 (78.2)6.3 (-5.4, 16.0)0.27Case based forms available in HF66 (93.0)79 (79.8)91 (90.1)110 (87.3)114 (82.0)139 (89.1)3.9 (-5.4, 11.0)0.36Rumor logs available in HF39 (54.9)55 (55.6)52 (51.5)78 (61.9)84 (60.4)97 (62.1)7.2 (-6.3, 20.8)0.31Data Analysis and UseHF conducts data basic data analysis28 (39.4)49 (49.5)41 (40.6)51 (40.5)63 (45.3)99 (63.4)24.0 (10.0, 36.7)\n0.0008\nHF have current line graphs showing trends of priority diseases5 (7.0)11 (11.1)16 (15.8)29 (23.0)43 (30.9)72(46.1)39.1 (27.8, 47.9)\n< 0.0001\nOutbreak notification and investigationHF reported outbreak within 12 months12 (16.9)45 (45.5)32 (31.7)10 (7.9)7 (5.0)17(10.9)6.0 (-4.2, 16.1)0.31Outbreak notified to DHMT within 48 h*11 (91.7)29 (64.4)29 (90.6)9 (90.0)5 (71.4)15(88.2%)3.5 (-24.7, 27.1)0.09Monitoring and CommunicationSupervisory visits from DHMT (at least once every three months)44 (62.0)91 (91.9)78 (77.2)79 (62.7)70 (50.4)144 (92.3)30.3(18.6, 42.4)\n< 0.0001\nMobile network available in HF67 (94.4)79 (79.8)91 (90.1)120 (95.2)126 (90.6)144 (92.3)2.1 (-6.5, 8.4)0.57*Denominator is number of outbreaks reported in the preceding 12 monthsHF: Health Facility; IDSR: Integrated Disease Surveillance and Response; HCW: Healthcare worker; DHMT: District Health Management Team\n\nComparison of IDSR indicators in selected health facilities, Sierra Leone, 2016–2021\n*Denominator is number of outbreaks reported in the preceding 12 months\nHF: Health Facility; IDSR: Integrated Disease Surveillance and Response; HCW: Healthcare worker; DHMT: District Health Management Team", "The proportion of health facilities that displayed standard case definition posters in consultation rooms for use by health workers in identifying cases increased significantly from 76.1% to 2016 to 94.9% in 2021 (increase of 18.8% points; 95% CI 9.2, 30.2); P- value 0.002). The proportion of IDSR focal persons who conducted weekly active case search for priority diseases in the health facilities remained the same at 79% for both baseline and end line although it fluctuated across the years. There was positive trend in adequate knowledge of standard case definitions among health workers for five priority diseases that were assessed as shown in Fig. 2 (Acute flaccid paralysis, Neonatal tetanus, Measles, Cholera and Ebola Virus Disease). Additionally, COVID-19 was added in 2021 and 61% of the health workers had adequate knowledge on its case definition.\n\nFig. 2Knowledge of standard case definitions among interviewed healthcare workers, Sierra Leone, 2016 and 2021\n\nKnowledge of standard case definitions among interviewed healthcare workers, Sierra Leone, 2016 and 2021", "Knowledge of IDSR reporting requirements was high and by the end line assessment in 2021, almost all interviewed health care workers correctly defined the epidemiologic week, zero reporting and reporting deadlines (Table 2). There was a significant improvement in the proportion of health facilities that submitted all the required surveillance reports (completeness of reporting) from 84.5% to 2016 to 96% in 2021(increase of 11.5% points; 95% CI 3.6, 21.9; P-value 0.003). During the same period, timeliness of IDSR reports improved from 80.3 to 92% (increase of 11.7% points; 95% CI 2.4, 22.9; P-value 0.01).", "Availability of the weekly IDSR reporting tool improved from 88.7% to 2016 to 96.1% in 2021 (increase of 7.4% points; 95% CI 0.4, 17.1; P-value 0.03). Availability of other reporting tools did not change significantly and were at 78%, 89% and 62% in 2021 for line listing forms, case-based reporting forms and rumour logbooks, respectively (Table 2).", "There was significant improvement in data analysis and use in health facilities over the years. The proportion of health facilities that conducted basic data analysis improved from 39% at baseline to 63% at end line (increase of 24% points; 95% CI 10.0, 36.7; P-value 0.0008). The proportion of health facilities with current line graphs showing trends in occurrence of priority diseases increased significantly from 7 to 46.1% (increase of 39.1% points; 95% CI 27.8, 47.9; P-value < 0.0001) (Table 2).", "The proportion of health facilities that had identified an outbreak within 12 months of the assessment did not change significantly and was 16.9% at baseline compared to 10.9% at end line (P-value 0.31). This was mostly because there were no outbreaks to detect and not for lack of detection capacity. The proportion of identified outbreaks that were notified to the District Health Management Teams within 48 h did not change significantly and were 91.7% at baseline compared to 88.2% at end line (P-value 0.09) (Table 2).", "Quarterly supervisory visits by DHMTs to the health facilities improved from 62% at baseline to 92% (P-value < 000.1) at end line although it fluctuated for other visits (Table 2). Mobile network connectivity remained high throughout the period and 92% of the health facilities assessed at end line had connectivity.", "The proportion of health facilities providing routine immunization services was above 90% in all the assessments with the end line being 96%. The number of immunizing health facilities with functional refrigerators significantly improved from 49.2% at baseline to 66.7% (P-value 0.02) although it fluctuated for other visits (Table 3). Immunizing health facilities with updated temperature monitoring charts significantly improved from 34 to 81% (P-value < 0.0001) while health facilities with all required basic antigens (vaccines) available at the time of the assessment improved from 48 to 83% (P-value < 0.0001). At the end line assessment, there were only 6.7% health facilities with Vaccine Vial Monitor (VVM) at stage three or four which is considered as excessive exposure to high temperatures (Table 3). Malaria management commodities including first line anti-malarial drugs, rapid diagnostic test kits and insecticide treated nets were available in most of the health facilities throughout the assessment period although there was a significant drop for first line anti-malarial drugs and rapid diagnostic test kits at end line assessment in August 2021, mostly due to disruptions associated with COVID-19 (Table 3).\n\nTable 3Assessing routine immunization, malaria commodities and diagnostic capacity in health facilities, Sierra Leone, 2016–2021Programmatic AreaVariableFeb_2016July_2016May_2017Aug_2018Feb_2019Aug_2021End Line vs. BaselineP-value(N = 71)n (%)(N = 99)n (%)(N = 101)n (%)(N = 126)n (%)(N = 139)n (%)(N = 156)n (%)Difference (95 CI)\nVaccination\nHealth facilities providing routine Immunization Services65 (91.5)95 (96.0)96 (95.0)120 (95.2)135 (97.1)150 (96.2)4.7 (-1.5, 13.7)0.14Health facilities with functional refrigerator†32 (49.2)70 (73.7)72 (75.0)71 (59.2)88 (65.2)100 (66.7)17.5 (3.3, 31.2)\n0.0157\nHealth facilities with updated temperature monitoring chart††11 (34.4)16 (22.9)32 (44.4)29 (40.8)37 (42.0)81 (81)46.6 (27.2,62.0)\n< 0.0001\nHealth facilities with all required basic antigens available at the time of the assessment†31(47.7)55(57.9)60(62.5)86 (71.7)103 (76.3)125 (83.3)35.6 (21.9, 48.3)\n< 0.0001\nHeath facilities with expired antigens†2 (3.1)3 (3.2)1 (1)1 (0.8)2 (1.5)2 (1.3)1.8 (-2.3, 9.3)0.37Health facilities with Vaccine Vial Monitor at stage three and four†14 (21.5)16 (16.8)14 (14.6)8 (6.7)9 (6.7)10 (6.7)14.8 (5.1, 26.6)\n0.002\n\nMalaria Commodities\nHealth facilities with 1st line anti-malaria drugs69 (97.2)96 (97.0)99 (98.0)120 (95.2)135 (97.1)120 (76.9)20.3 (11.2, 27.7)\n0.0002\nHealth Facilities with rapid diagnostic kits for diagnosis of malaria67 (94.4)89 (89.9)98 (97.0)120 (95.2)134 (96.4)119 (76.3)18.1 (8.1, 26.1)\n0.001\nHealth facilities with insecticide treated nets55 (77.5)92 (92.9)97 (96.0)111 (88.1)132 (95.0)139 (89.1)11.6 (1.6, 23.3)\n0.0218\n†Denominator is number of health facilities providing immunization services†† Denominator is number of health facilities with a functional fridge\n\nAssessing routine immunization, malaria commodities and diagnostic capacity in health facilities, Sierra Leone, 2016–2021\n†Denominator is number of health facilities providing immunization services\n†† Denominator is number of health facilities with a functional fridge" ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "The effect on Ebola Virus Disease outbreak on the health workforce in Sierra Leone", "Improving public health surveillance after the Ebola outbreak", "Methods", "Study setting and design", "Selection of health facilities", "Data collection", "Data analysis", "Results", "Coordination of IDSR activities in health facilities", "Identification of priority IDSR diseases, conditions and events", "Awareness and adherence to IDSR reporting requirements", "Availability of IDSR reporting tools", "Data analysis and use at health facility level", "Outbreak detection and notification", "Feedback, monitoring and communication", "Availability of routine immunization services and commodities for management of Malaria", "Discussion", "Conclusion" ]
[ "Supportive supervision is a unique approach to monitoring performance through face to face interactions between a skilled health care worker and a less skilled worker [1]. First promoted in the era of primary health care, supportive supervision sought to improve performance of healthcare workers offering essential services such as immunization in remote areas [2]. However, this approach can be impeded by poor coordination, lack of motivation, geographical barriers, competing activities, high costs and armed conflicts [1, 3].\nInitially, a top-down approach to supervision was used with inspection and correction of junior workers done by a senior officer. There has been a shift to a more collaborative approach (support supervision) with focus on identifying challenges in service delivery and involving the provider in finding solutions. This approach improves motivation and ownership by the health care providers [4]. Records reviews, direct observations, use of objective indicators, collaborative problem solving and targeted on job training are additional collaborative approaches used [1, 5].\nIn 1998, the World Health Organization (WHO), Africa Regional Office adopted the Integrated Disease Surveillance (IDS) strategy, later renamed Integrated Disease Surveillance and Response (IDSR) strategy to strengthen public health surveillance and response in member states [6]. Implementation of the IDSR strategy varies, with the initial momentum exhibited in the first years of implementation declining over time due to several challenges such as non-sustainable financial resources, poor coordination, inadequate training, high turnover of staff, inadequate supervision from the next level, erratic feedback, weak laboratory capacities, unavailability of job aids and poor availability of communication and transport systems particularly at the periphery [7–9]. Considering the growing threat of epidemics especially from new and emerging pathogens, the need for strengthened surveillance systems has never been greater. Ministries of Health in WHO member states are urged to monitor implementation of IDSR as this directly contributes to fulfillment of the International Health Regulations (2005) [10]. National level health staff utilize supervision visits to monitor the implementation of Integrated Disease Surveillance and Response (IDSR) activities and identify key areas that need improvement [11]. While training of sub- national health care workers on IDSR provides them with basic understanding of public health surveillance, supportive supervision reinforces concepts, and identifies and resolves challenges affecting implementation [12].\n[SUBTITLE] The effect on Ebola Virus Disease outbreak on the health workforce in Sierra Leone [SUBSECTION] Health care workers are a high-risk group for highly infectious diseases such as Ebola Virus Disease (EVD). Out of 304 health care workers infected with Ebola virus in Sierra Leone in 2014–2016, two hundred and twenty-one (221) died and several others resigned out of fear of getting infected. This reduced the ratio of skilled health workers from 17.2/ 10,000 to < 4 /10,000 population [13–15]. After the outbreak, the Ministry of Health and Sanitation (MoHS) embarked on strengthening health systems, improving public health surveillance and cross border surveillance [14].\nHealth care workers are a high-risk group for highly infectious diseases such as Ebola Virus Disease (EVD). Out of 304 health care workers infected with Ebola virus in Sierra Leone in 2014–2016, two hundred and twenty-one (221) died and several others resigned out of fear of getting infected. This reduced the ratio of skilled health workers from 17.2/ 10,000 to < 4 /10,000 population [13–15]. After the outbreak, the Ministry of Health and Sanitation (MoHS) embarked on strengthening health systems, improving public health surveillance and cross border surveillance [14].\n[SUBTITLE] Improving public health surveillance after the Ebola outbreak [SUBSECTION] Towards the end of the Ebola Virus Disease outbreak, the Sierra Leone Ministry of Health and Sanitation in collaboration with World Health Organization country office (WCO) and other health sector development partners, embarked on revitalization of IDSR. This involved adaptation of the 2010 WHO-Africa region IDSR technical guidelines and training modules, training of healthcare workers and providing inputs and infrastructure to support IDSR implementation [16].\n\nPreviously, paper-based checklists were used during supportive supervision in Sierra Leone and health facilities were selected purposively based on ease of access. Thus, it was likely that health facilities in remote areas were omitted from supervisory visits. Collation and analysis of data from the supervisory visits was seldom done as it required abstraction of data from paper-based checklists to a computer and this often required hiring of data clerks due to staff shortage. To overcome these issues, we adopted an electronic supervision checklist and an open source platform to collect and manage data. This paper describes the innovative, integrated approach to IDSR supportive supervision. We share the lessons learnt, hopeful that they can provide insight for improving monitoring of public health surveillance programs particularly for countries implementing IDSR.\n\nThe integrated support supervision process described in this paper is considered an innovative way of monitoring IDSR performance indicators for several reasons. First, routine supervision visits were used to collect comprehensive and representative data to monitor key IDSR indicators from 2016 to 2021. During the visits, verbal and written feedback on performance was then given to the health facilities and district health management teams which ensured that corrective action was taken where necessary. Second, an integrated questionnaire was used which not only collected surveillance data, but also data from other related programs such as laboratory, immunization and malaria. Third, the use of electronic supervision checklist and an open source data collection platform to collect and manage the data ensured secure storage and rapid retrieval of data for analysis which enabled monitoring of performance over time.\nTowards the end of the Ebola Virus Disease outbreak, the Sierra Leone Ministry of Health and Sanitation in collaboration with World Health Organization country office (WCO) and other health sector development partners, embarked on revitalization of IDSR. This involved adaptation of the 2010 WHO-Africa region IDSR technical guidelines and training modules, training of healthcare workers and providing inputs and infrastructure to support IDSR implementation [16].\n\nPreviously, paper-based checklists were used during supportive supervision in Sierra Leone and health facilities were selected purposively based on ease of access. Thus, it was likely that health facilities in remote areas were omitted from supervisory visits. Collation and analysis of data from the supervisory visits was seldom done as it required abstraction of data from paper-based checklists to a computer and this often required hiring of data clerks due to staff shortage. To overcome these issues, we adopted an electronic supervision checklist and an open source platform to collect and manage data. This paper describes the innovative, integrated approach to IDSR supportive supervision. We share the lessons learnt, hopeful that they can provide insight for improving monitoring of public health surveillance programs particularly for countries implementing IDSR.\n\nThe integrated support supervision process described in this paper is considered an innovative way of monitoring IDSR performance indicators for several reasons. First, routine supervision visits were used to collect comprehensive and representative data to monitor key IDSR indicators from 2016 to 2021. During the visits, verbal and written feedback on performance was then given to the health facilities and district health management teams which ensured that corrective action was taken where necessary. Second, an integrated questionnaire was used which not only collected surveillance data, but also data from other related programs such as laboratory, immunization and malaria. Third, the use of electronic supervision checklist and an open source data collection platform to collect and manage the data ensured secure storage and rapid retrieval of data for analysis which enabled monitoring of performance over time.", "Health care workers are a high-risk group for highly infectious diseases such as Ebola Virus Disease (EVD). Out of 304 health care workers infected with Ebola virus in Sierra Leone in 2014–2016, two hundred and twenty-one (221) died and several others resigned out of fear of getting infected. This reduced the ratio of skilled health workers from 17.2/ 10,000 to < 4 /10,000 population [13–15]. After the outbreak, the Ministry of Health and Sanitation (MoHS) embarked on strengthening health systems, improving public health surveillance and cross border surveillance [14].", "Towards the end of the Ebola Virus Disease outbreak, the Sierra Leone Ministry of Health and Sanitation in collaboration with World Health Organization country office (WCO) and other health sector development partners, embarked on revitalization of IDSR. This involved adaptation of the 2010 WHO-Africa region IDSR technical guidelines and training modules, training of healthcare workers and providing inputs and infrastructure to support IDSR implementation [16].\n\nPreviously, paper-based checklists were used during supportive supervision in Sierra Leone and health facilities were selected purposively based on ease of access. Thus, it was likely that health facilities in remote areas were omitted from supervisory visits. Collation and analysis of data from the supervisory visits was seldom done as it required abstraction of data from paper-based checklists to a computer and this often required hiring of data clerks due to staff shortage. To overcome these issues, we adopted an electronic supervision checklist and an open source platform to collect and manage data. This paper describes the innovative, integrated approach to IDSR supportive supervision. We share the lessons learnt, hopeful that they can provide insight for improving monitoring of public health surveillance programs particularly for countries implementing IDSR.\n\nThe integrated support supervision process described in this paper is considered an innovative way of monitoring IDSR performance indicators for several reasons. First, routine supervision visits were used to collect comprehensive and representative data to monitor key IDSR indicators from 2016 to 2021. During the visits, verbal and written feedback on performance was then given to the health facilities and district health management teams which ensured that corrective action was taken where necessary. Second, an integrated questionnaire was used which not only collected surveillance data, but also data from other related programs such as laboratory, immunization and malaria. Third, the use of electronic supervision checklist and an open source data collection platform to collect and manage the data ensured secure storage and rapid retrieval of data for analysis which enabled monitoring of performance over time.", "[SUBTITLE] Study setting and design [SUBSECTION] This cross-sectional study was part of operational research to monitor implementation of IDSR program in Sierra Leone post Ebola outbreak. Data was collected through supportive supervision visits to health facilities across all districts in Sierra Leone from 2016 to 2021. The data collected was used to monitor and improve IDSR implementation in the country.\nThis cross-sectional study was part of operational research to monitor implementation of IDSR program in Sierra Leone post Ebola outbreak. Data was collected through supportive supervision visits to health facilities across all districts in Sierra Leone from 2016 to 2021. The data collected was used to monitor and improve IDSR implementation in the country.\n[SUBTITLE] Selection of health facilities [SUBSECTION] The total number of health facilities sampled nationally (sample size) per each support supervision visit was purposively selected based on available resources. There were about 1425 health facilities in the country in 2021, distributed by type as follows, in order of size and scope of services offered (from largest to smallest): Hospital 4%, Community Health Centres (CHC) 19%, Community Health Posts (CHP) 30%, Maternal and Child Health Posts (MCHP) 43% and Clinic 3%. About 10% of the total existing facilities per year was sampled in each visit. To ensure representativeness, health facilities were selected from each type of health facility (except clinics which are mostly private) in all the districts. Selection of health facilities in each district was done randomly from a list of health facilities stratified by health facility type as indicated above. First, health facilities were stratified according to the type of health facility. Thereafter, random sampling, using computer generated numbers was done to identify at least one hospital, three CHC, three CHP and three MCHP in each district. Where available, major private health facilities equivalent to a hospital were included.\nOver the six-year period, six national level supportive supervision visits were conducted covering 71 health facilities in February 2016, 99 in July 2016, 101 in May 2017, 126 in August 2018, 139 in February 2019 and 156 in August 2021 (Fig. 1). No national level supervisory visit was carried out in 2020 due to COVID-19 pandemic. Apart from the national supervision visits, the District Health Management Teams conducted two to four supervision visits per year for the facilities under their jurisdiction. However, this data was captured in a separate database and is therefore not included in this article.\n\nFig. 1Map of Sierra Leone showing health facilities visited during support supervision, 2016–2021\n\nMap of Sierra Leone showing health facilities visited during support supervision, 2016–2021\nThe total number of health facilities sampled nationally (sample size) per each support supervision visit was purposively selected based on available resources. There were about 1425 health facilities in the country in 2021, distributed by type as follows, in order of size and scope of services offered (from largest to smallest): Hospital 4%, Community Health Centres (CHC) 19%, Community Health Posts (CHP) 30%, Maternal and Child Health Posts (MCHP) 43% and Clinic 3%. About 10% of the total existing facilities per year was sampled in each visit. To ensure representativeness, health facilities were selected from each type of health facility (except clinics which are mostly private) in all the districts. Selection of health facilities in each district was done randomly from a list of health facilities stratified by health facility type as indicated above. First, health facilities were stratified according to the type of health facility. Thereafter, random sampling, using computer generated numbers was done to identify at least one hospital, three CHC, three CHP and three MCHP in each district. Where available, major private health facilities equivalent to a hospital were included.\nOver the six-year period, six national level supportive supervision visits were conducted covering 71 health facilities in February 2016, 99 in July 2016, 101 in May 2017, 126 in August 2018, 139 in February 2019 and 156 in August 2021 (Fig. 1). No national level supervisory visit was carried out in 2020 due to COVID-19 pandemic. Apart from the national supervision visits, the District Health Management Teams conducted two to four supervision visits per year for the facilities under their jurisdiction. However, this data was captured in a separate database and is therefore not included in this article.\n\nFig. 1Map of Sierra Leone showing health facilities visited during support supervision, 2016–2021\n\nMap of Sierra Leone showing health facilities visited during support supervision, 2016–2021\n[SUBTITLE] Data collection [SUBSECTION] The assessment teams comprised of national level staff from the surveillance and laboratory programs of MOHS as well as development partners like World Health Organization who were supporting the supervision missions technically and financially. Respondents in the assessments were a team of healthcare workers involved in surveillance and administrative functions in the health facilities. These included the IDSR focal person, health facility in charge, laboratory technician and the nurse in charge of vaccination activities.\nOne week prior to each supportive supervisory visit, the MoHS through the National Disease Surveillance Program contacted the District Medical Officers (DMO) and District Surveillance Officers (DSOs) to inform them of the upcoming supervisory visits. On the first day of the visit, the supervisory team comprising of officials from MoHS and supporting development partners met with the District Health Management Team (DHMT) members to discuss the purpose of the supervisory visits and to select health facilities for the assessment. The selected health facilities were not informed of the visit in advance in order to avoid pre-emptive interference with data tools. Upon reaching a health facility, the supervisory team captured the geographic coordinates of the health facilities using automatically generated geo- coordinates. Recording of geo-coordinates for each facility ensured that there was evidence of representativeness in the distribution of selected health facilities. Next the team conducted interviews, verified documents and confirmed availability of IDSR reporting tools.\nData was collected in real-time using a structured electronic questionnaire created using Open Data Kit platform and loaded onto tablets. Using the electronic tool eased data collection and management, making it possible to analyse data, provide timely feedback to the health facilities staff, and also track performance over time. The questionnaire covered six out of eight core functions of IDSR as well as an assessment on related programmatic areas such as available laboratory services, maternal deaths, malaria commodities and routine immunization services. The questionnaire collected information on (1) case identification; (2) reporting of priority diseases; (3) data analysis and interpretation; (4) investigation and confirmation of reported cases and outbreaks; (5) feedback between the district management teams and health facilities; and (6) monitoring and evaluation (Table 1).\n\nTable 1IDSR Core Functions Assessed During Integrated Support Supervision, Sierra Leone, 2016–2021Core FunctionAssessment methodsMain focus areas1Identification of priority diseases, conditions and eventsInterview, observation, review of registersAssessed the ability of healthcare workers to use standard case definition to identify priority diseases through records review and active case search in the community2Reporting of Priority diseasesInterview, documents review, observationChecked if HCWs are aware of reporting requirements for priority diseases and the availability and use of reporting tools3Data analysis and interpretationInterview and observationChecked if data analysis and interpretation was done at the facility4Investigation and confirmation of outbreaksInterviewAssessed if HF are able to detect outbreaks from various sources, notify the district and participate in outbreak investigations5FeedbackInterviewConfirmed if the HF gets feedback on IDSR performance from the DHMT6Monitoring and EvaluationInterview, document reviewAssessed the frequency of supervision done by the DHMT and mechanisms for monitoring IDSR performance in the HFIDSR: Integrated Disease Surveillance and Response; HCW: Healthcare workers; HF: Health Facility; DHMT: District Health Management Team\n\nIDSR Core Functions Assessed During Integrated Support Supervision, Sierra Leone, 2016–2021\nIDSR: Integrated Disease Surveillance and Response; HCW: Healthcare workers; HF: Health Facility; DHMT: District Health Management Team\nThe teams began by assessing availability of IDSR focal persons in the health facilities and their participation in district monthly review meetings. This is because the focal persons are important in coordinating IDSR activities. On case identification, supervisors checked if IDSR posters with case definitions were available and displayed in the consultation rooms. They then interviewed the respondents to gauge their knowledge of case definitions for five selected IDSR priority diseases/conditions (Acute flaccid paralysis, Neonatal tetanus, Measles, Cholera and Ebola Virus Disease). Additionally, COVID-19 was added in the 2021 assessment. A healthcare worker who mentioned all signs and symptoms correctly was classified as having adequate knowledge, while one who mentioned some of the signs and symptoms was classified as having limited knowledge. A healthcare worker who did not mention any of the appropriate signs and symptoms was classified as having no knowledge.\nThe section on reporting of priority diseases assessed knowledge of and compliance with reporting requirements for priority diseases. Copies of IDSR weekly reports for the previous four weeks were checked to confirm if all reports were available and if they had been submitted on time. The team also checked the availability of case-based reporting forms, weekly reporting forms, line listing forms, and rumour logbooks for recording suspected outbreaks/events. The section on data analysis assessed the capacity and type of data analysis conducted to monitor trends of priority diseases at the health facilities. The section on investigation captured information on case investigation and confirmation of reported outbreaks. The section on feedback assessed feedback mechanisms from the district to the health facilities and vice versa. It included a review of previous supervision reports to elicit frequency of supportive supervision by DHMT. The section on monitoring and evaluation assessed mechanisms for monitoring IDSR performance in the health facility.\nAdditional questions on immunization, malaria treatment commodities and maternal mortality were also included in the questionnaire. The questionnaire had compulsory fields and intrinsic data validation mechanisms to enhance data accuracy and completeness. Finally, at the end of the data collection process, the teams used supervision books available in each health facility to record strengths, gaps, challenges and recommendations agreed upon during the supervisory visit. These records were useful for follow up during subsequent supervisory visits. Supervision also provided an opportunity for on the job training and distribution of IDSR reference materials.\nThe assessment teams comprised of national level staff from the surveillance and laboratory programs of MOHS as well as development partners like World Health Organization who were supporting the supervision missions technically and financially. Respondents in the assessments were a team of healthcare workers involved in surveillance and administrative functions in the health facilities. These included the IDSR focal person, health facility in charge, laboratory technician and the nurse in charge of vaccination activities.\nOne week prior to each supportive supervisory visit, the MoHS through the National Disease Surveillance Program contacted the District Medical Officers (DMO) and District Surveillance Officers (DSOs) to inform them of the upcoming supervisory visits. On the first day of the visit, the supervisory team comprising of officials from MoHS and supporting development partners met with the District Health Management Team (DHMT) members to discuss the purpose of the supervisory visits and to select health facilities for the assessment. The selected health facilities were not informed of the visit in advance in order to avoid pre-emptive interference with data tools. Upon reaching a health facility, the supervisory team captured the geographic coordinates of the health facilities using automatically generated geo- coordinates. Recording of geo-coordinates for each facility ensured that there was evidence of representativeness in the distribution of selected health facilities. Next the team conducted interviews, verified documents and confirmed availability of IDSR reporting tools.\nData was collected in real-time using a structured electronic questionnaire created using Open Data Kit platform and loaded onto tablets. Using the electronic tool eased data collection and management, making it possible to analyse data, provide timely feedback to the health facilities staff, and also track performance over time. The questionnaire covered six out of eight core functions of IDSR as well as an assessment on related programmatic areas such as available laboratory services, maternal deaths, malaria commodities and routine immunization services. The questionnaire collected information on (1) case identification; (2) reporting of priority diseases; (3) data analysis and interpretation; (4) investigation and confirmation of reported cases and outbreaks; (5) feedback between the district management teams and health facilities; and (6) monitoring and evaluation (Table 1).\n\nTable 1IDSR Core Functions Assessed During Integrated Support Supervision, Sierra Leone, 2016–2021Core FunctionAssessment methodsMain focus areas1Identification of priority diseases, conditions and eventsInterview, observation, review of registersAssessed the ability of healthcare workers to use standard case definition to identify priority diseases through records review and active case search in the community2Reporting of Priority diseasesInterview, documents review, observationChecked if HCWs are aware of reporting requirements for priority diseases and the availability and use of reporting tools3Data analysis and interpretationInterview and observationChecked if data analysis and interpretation was done at the facility4Investigation and confirmation of outbreaksInterviewAssessed if HF are able to detect outbreaks from various sources, notify the district and participate in outbreak investigations5FeedbackInterviewConfirmed if the HF gets feedback on IDSR performance from the DHMT6Monitoring and EvaluationInterview, document reviewAssessed the frequency of supervision done by the DHMT and mechanisms for monitoring IDSR performance in the HFIDSR: Integrated Disease Surveillance and Response; HCW: Healthcare workers; HF: Health Facility; DHMT: District Health Management Team\n\nIDSR Core Functions Assessed During Integrated Support Supervision, Sierra Leone, 2016–2021\nIDSR: Integrated Disease Surveillance and Response; HCW: Healthcare workers; HF: Health Facility; DHMT: District Health Management Team\nThe teams began by assessing availability of IDSR focal persons in the health facilities and their participation in district monthly review meetings. This is because the focal persons are important in coordinating IDSR activities. On case identification, supervisors checked if IDSR posters with case definitions were available and displayed in the consultation rooms. They then interviewed the respondents to gauge their knowledge of case definitions for five selected IDSR priority diseases/conditions (Acute flaccid paralysis, Neonatal tetanus, Measles, Cholera and Ebola Virus Disease). Additionally, COVID-19 was added in the 2021 assessment. A healthcare worker who mentioned all signs and symptoms correctly was classified as having adequate knowledge, while one who mentioned some of the signs and symptoms was classified as having limited knowledge. A healthcare worker who did not mention any of the appropriate signs and symptoms was classified as having no knowledge.\nThe section on reporting of priority diseases assessed knowledge of and compliance with reporting requirements for priority diseases. Copies of IDSR weekly reports for the previous four weeks were checked to confirm if all reports were available and if they had been submitted on time. The team also checked the availability of case-based reporting forms, weekly reporting forms, line listing forms, and rumour logbooks for recording suspected outbreaks/events. The section on data analysis assessed the capacity and type of data analysis conducted to monitor trends of priority diseases at the health facilities. The section on investigation captured information on case investigation and confirmation of reported outbreaks. The section on feedback assessed feedback mechanisms from the district to the health facilities and vice versa. It included a review of previous supervision reports to elicit frequency of supportive supervision by DHMT. The section on monitoring and evaluation assessed mechanisms for monitoring IDSR performance in the health facility.\nAdditional questions on immunization, malaria treatment commodities and maternal mortality were also included in the questionnaire. The questionnaire had compulsory fields and intrinsic data validation mechanisms to enhance data accuracy and completeness. Finally, at the end of the data collection process, the teams used supervision books available in each health facility to record strengths, gaps, challenges and recommendations agreed upon during the supervisory visit. These records were useful for follow up during subsequent supervisory visits. Supervision also provided an opportunity for on the job training and distribution of IDSR reference materials.\n[SUBTITLE] Data analysis [SUBSECTION] Data was extracted from the Open Data Kit (ODK) platform first onto an MS Excel database, merged, checked for duplicates and completeness and then exported to Epi Info for analysis. We calculated means and medians for continuous variables and proportions for categorical data. Two proportion Z-test was used to compare differences in performance recorded during the baseline in February 2016 and the end line in August 2021. District weekly report completeness was defined as the proportion of health facilities that submitted required reports to the district health office. This was verified by checking if all copies of the IDSR weekly reports for the preceding four weeks were available. Timeliness was defined as the proportion of health facilities that submitted IDSR weekly reports to the district by 12 pm every Monday.\nData was extracted from the Open Data Kit (ODK) platform first onto an MS Excel database, merged, checked for duplicates and completeness and then exported to Epi Info for analysis. We calculated means and medians for continuous variables and proportions for categorical data. Two proportion Z-test was used to compare differences in performance recorded during the baseline in February 2016 and the end line in August 2021. District weekly report completeness was defined as the proportion of health facilities that submitted required reports to the district health office. This was verified by checking if all copies of the IDSR weekly reports for the preceding four weeks were available. Timeliness was defined as the proportion of health facilities that submitted IDSR weekly reports to the district by 12 pm every Monday.", "This cross-sectional study was part of operational research to monitor implementation of IDSR program in Sierra Leone post Ebola outbreak. Data was collected through supportive supervision visits to health facilities across all districts in Sierra Leone from 2016 to 2021. The data collected was used to monitor and improve IDSR implementation in the country.", "The total number of health facilities sampled nationally (sample size) per each support supervision visit was purposively selected based on available resources. There were about 1425 health facilities in the country in 2021, distributed by type as follows, in order of size and scope of services offered (from largest to smallest): Hospital 4%, Community Health Centres (CHC) 19%, Community Health Posts (CHP) 30%, Maternal and Child Health Posts (MCHP) 43% and Clinic 3%. About 10% of the total existing facilities per year was sampled in each visit. To ensure representativeness, health facilities were selected from each type of health facility (except clinics which are mostly private) in all the districts. Selection of health facilities in each district was done randomly from a list of health facilities stratified by health facility type as indicated above. First, health facilities were stratified according to the type of health facility. Thereafter, random sampling, using computer generated numbers was done to identify at least one hospital, three CHC, three CHP and three MCHP in each district. Where available, major private health facilities equivalent to a hospital were included.\nOver the six-year period, six national level supportive supervision visits were conducted covering 71 health facilities in February 2016, 99 in July 2016, 101 in May 2017, 126 in August 2018, 139 in February 2019 and 156 in August 2021 (Fig. 1). No national level supervisory visit was carried out in 2020 due to COVID-19 pandemic. Apart from the national supervision visits, the District Health Management Teams conducted two to four supervision visits per year for the facilities under their jurisdiction. However, this data was captured in a separate database and is therefore not included in this article.\n\nFig. 1Map of Sierra Leone showing health facilities visited during support supervision, 2016–2021\n\nMap of Sierra Leone showing health facilities visited during support supervision, 2016–2021", "The assessment teams comprised of national level staff from the surveillance and laboratory programs of MOHS as well as development partners like World Health Organization who were supporting the supervision missions technically and financially. Respondents in the assessments were a team of healthcare workers involved in surveillance and administrative functions in the health facilities. These included the IDSR focal person, health facility in charge, laboratory technician and the nurse in charge of vaccination activities.\nOne week prior to each supportive supervisory visit, the MoHS through the National Disease Surveillance Program contacted the District Medical Officers (DMO) and District Surveillance Officers (DSOs) to inform them of the upcoming supervisory visits. On the first day of the visit, the supervisory team comprising of officials from MoHS and supporting development partners met with the District Health Management Team (DHMT) members to discuss the purpose of the supervisory visits and to select health facilities for the assessment. The selected health facilities were not informed of the visit in advance in order to avoid pre-emptive interference with data tools. Upon reaching a health facility, the supervisory team captured the geographic coordinates of the health facilities using automatically generated geo- coordinates. Recording of geo-coordinates for each facility ensured that there was evidence of representativeness in the distribution of selected health facilities. Next the team conducted interviews, verified documents and confirmed availability of IDSR reporting tools.\nData was collected in real-time using a structured electronic questionnaire created using Open Data Kit platform and loaded onto tablets. Using the electronic tool eased data collection and management, making it possible to analyse data, provide timely feedback to the health facilities staff, and also track performance over time. The questionnaire covered six out of eight core functions of IDSR as well as an assessment on related programmatic areas such as available laboratory services, maternal deaths, malaria commodities and routine immunization services. The questionnaire collected information on (1) case identification; (2) reporting of priority diseases; (3) data analysis and interpretation; (4) investigation and confirmation of reported cases and outbreaks; (5) feedback between the district management teams and health facilities; and (6) monitoring and evaluation (Table 1).\n\nTable 1IDSR Core Functions Assessed During Integrated Support Supervision, Sierra Leone, 2016–2021Core FunctionAssessment methodsMain focus areas1Identification of priority diseases, conditions and eventsInterview, observation, review of registersAssessed the ability of healthcare workers to use standard case definition to identify priority diseases through records review and active case search in the community2Reporting of Priority diseasesInterview, documents review, observationChecked if HCWs are aware of reporting requirements for priority diseases and the availability and use of reporting tools3Data analysis and interpretationInterview and observationChecked if data analysis and interpretation was done at the facility4Investigation and confirmation of outbreaksInterviewAssessed if HF are able to detect outbreaks from various sources, notify the district and participate in outbreak investigations5FeedbackInterviewConfirmed if the HF gets feedback on IDSR performance from the DHMT6Monitoring and EvaluationInterview, document reviewAssessed the frequency of supervision done by the DHMT and mechanisms for monitoring IDSR performance in the HFIDSR: Integrated Disease Surveillance and Response; HCW: Healthcare workers; HF: Health Facility; DHMT: District Health Management Team\n\nIDSR Core Functions Assessed During Integrated Support Supervision, Sierra Leone, 2016–2021\nIDSR: Integrated Disease Surveillance and Response; HCW: Healthcare workers; HF: Health Facility; DHMT: District Health Management Team\nThe teams began by assessing availability of IDSR focal persons in the health facilities and their participation in district monthly review meetings. This is because the focal persons are important in coordinating IDSR activities. On case identification, supervisors checked if IDSR posters with case definitions were available and displayed in the consultation rooms. They then interviewed the respondents to gauge their knowledge of case definitions for five selected IDSR priority diseases/conditions (Acute flaccid paralysis, Neonatal tetanus, Measles, Cholera and Ebola Virus Disease). Additionally, COVID-19 was added in the 2021 assessment. A healthcare worker who mentioned all signs and symptoms correctly was classified as having adequate knowledge, while one who mentioned some of the signs and symptoms was classified as having limited knowledge. A healthcare worker who did not mention any of the appropriate signs and symptoms was classified as having no knowledge.\nThe section on reporting of priority diseases assessed knowledge of and compliance with reporting requirements for priority diseases. Copies of IDSR weekly reports for the previous four weeks were checked to confirm if all reports were available and if they had been submitted on time. The team also checked the availability of case-based reporting forms, weekly reporting forms, line listing forms, and rumour logbooks for recording suspected outbreaks/events. The section on data analysis assessed the capacity and type of data analysis conducted to monitor trends of priority diseases at the health facilities. The section on investigation captured information on case investigation and confirmation of reported outbreaks. The section on feedback assessed feedback mechanisms from the district to the health facilities and vice versa. It included a review of previous supervision reports to elicit frequency of supportive supervision by DHMT. The section on monitoring and evaluation assessed mechanisms for monitoring IDSR performance in the health facility.\nAdditional questions on immunization, malaria treatment commodities and maternal mortality were also included in the questionnaire. The questionnaire had compulsory fields and intrinsic data validation mechanisms to enhance data accuracy and completeness. Finally, at the end of the data collection process, the teams used supervision books available in each health facility to record strengths, gaps, challenges and recommendations agreed upon during the supervisory visit. These records were useful for follow up during subsequent supervisory visits. Supervision also provided an opportunity for on the job training and distribution of IDSR reference materials.", "Data was extracted from the Open Data Kit (ODK) platform first onto an MS Excel database, merged, checked for duplicates and completeness and then exported to Epi Info for analysis. We calculated means and medians for continuous variables and proportions for categorical data. Two proportion Z-test was used to compare differences in performance recorded during the baseline in February 2016 and the end line in August 2021. District weekly report completeness was defined as the proportion of health facilities that submitted required reports to the district health office. This was verified by checking if all copies of the IDSR weekly reports for the preceding four weeks were available. Timeliness was defined as the proportion of health facilities that submitted IDSR weekly reports to the district by 12 pm every Monday.", "[SUBTITLE] Coordination of IDSR activities in health facilities [SUBSECTION] IDSR focal persons were present in most health facilities during all assessments. When compared to the baseline assessment, the proportion of IDSR focal persons who participated in the monthly district management team meetings increased significantly from 74.6% to 2016 (baseline) to 96.1% at end line in 2021 (increase of 21.5% points; 95% CI (11.8, 32.9); P-value < 0.0001). Conversely, availability of IDSR technical guidelines at the health facilities declined from 97.2% to 2016 to 82.1% in 2021 (decrease of 15.1% points; 95% CI (6.5, 22.2); P-value 0.002) (Table 2). The decline in availability of IDSR technical guidelines was mainly due to loss without replacement.\n\nTable 2Comparison of IDSR indicators in selected health facilities, Sierra Leone, 2016–2021Core FunctionIndicatorsFeb_2016July_2016May_2017Aug_2018Feb_2019Aug_2021End Line vs. BaselineP valueN = 71n (%)N = 99n (%)N = 101n (%)N = 126n (%)N = 139n (%)N = 156n (%)Difference (95 CI)CoordinationHealth facility (HF) has IDSR focal person69 (97.2)97 (98.0)100 (99.0)121 (96.0)135 (97.1)153 (98.1)0.9 (-3.2, 7.9)0.67HF IDSR focal person attends district monthly meetings53 (74.6)87 (87.9)93 (92.1)118 (93.7)132 (95.0)150 (96.1)21.5 (11.8, 32.9)\n< 0.0001\nHF has IDSR Technical guidelines69 (97.2)89 (89.9)101 (100.0)102 (81.0)123 (88.5)128 (82.1)15.1 (6.5, 22.2)\n0.002\nCase IdentificationStandard Case definition poster displayed in at least one location in the health facility54 (76.1)84 (84.8)89 (88.1)114 (90.5)127 (91.4)148 (94.9)18.8 (9.2, 30.2)\n< 0.0001\nIDSR focal person conducts active case search at least once a week56 (78.9)54 (54.5)34 (33.7)100 (79.4)75 (54.0)123 (78.8)0.1 (-12.1, 10.7)0.99Reporting of Priority diseasesHCW correctly defines epidemiologic week51 (71.8)93 (93.9)99 (98.0)108 (85.7)132 (95)151 (96.8)25 (15.0, 36.5)\n< 0.0001\nHCW correctly defines zero reporting67 (94.4)94 (94.9)99 (98.0)121 (96.0)138 (99.3)147 (92.2)2.2 (-6.4, 8.5))0.55HCW knows the deadline for submitting weekly IDSR reports65 (91.5)96 (97.0)100 (99.0)115 (91.3)138 (99.3)151 (96.8)5.3 (-0.8, 14.3)0.09Weekly reporting forms available in HF63 (88.7)89 (89.9)96 (95.0)119 (94.4)127 (91.4)150 (96.1)7.4 (0.4, 17.1)\n0.03\nLine listing forms available in HF60 (84.5)78 (78.8)81 (80.2)95 (75.4)114 (82.0)122 (78.2)6.3 (-5.4, 16.0)0.27Case based forms available in HF66 (93.0)79 (79.8)91 (90.1)110 (87.3)114 (82.0)139 (89.1)3.9 (-5.4, 11.0)0.36Rumor logs available in HF39 (54.9)55 (55.6)52 (51.5)78 (61.9)84 (60.4)97 (62.1)7.2 (-6.3, 20.8)0.31Data Analysis and UseHF conducts data basic data analysis28 (39.4)49 (49.5)41 (40.6)51 (40.5)63 (45.3)99 (63.4)24.0 (10.0, 36.7)\n0.0008\nHF have current line graphs showing trends of priority diseases5 (7.0)11 (11.1)16 (15.8)29 (23.0)43 (30.9)72(46.1)39.1 (27.8, 47.9)\n< 0.0001\nOutbreak notification and investigationHF reported outbreak within 12 months12 (16.9)45 (45.5)32 (31.7)10 (7.9)7 (5.0)17(10.9)6.0 (-4.2, 16.1)0.31Outbreak notified to DHMT within 48 h*11 (91.7)29 (64.4)29 (90.6)9 (90.0)5 (71.4)15(88.2%)3.5 (-24.7, 27.1)0.09Monitoring and CommunicationSupervisory visits from DHMT (at least once every three months)44 (62.0)91 (91.9)78 (77.2)79 (62.7)70 (50.4)144 (92.3)30.3(18.6, 42.4)\n< 0.0001\nMobile network available in HF67 (94.4)79 (79.8)91 (90.1)120 (95.2)126 (90.6)144 (92.3)2.1 (-6.5, 8.4)0.57*Denominator is number of outbreaks reported in the preceding 12 monthsHF: Health Facility; IDSR: Integrated Disease Surveillance and Response; HCW: Healthcare worker; DHMT: District Health Management Team\n\nComparison of IDSR indicators in selected health facilities, Sierra Leone, 2016–2021\n*Denominator is number of outbreaks reported in the preceding 12 months\nHF: Health Facility; IDSR: Integrated Disease Surveillance and Response; HCW: Healthcare worker; DHMT: District Health Management Team\nIDSR focal persons were present in most health facilities during all assessments. When compared to the baseline assessment, the proportion of IDSR focal persons who participated in the monthly district management team meetings increased significantly from 74.6% to 2016 (baseline) to 96.1% at end line in 2021 (increase of 21.5% points; 95% CI (11.8, 32.9); P-value < 0.0001). Conversely, availability of IDSR technical guidelines at the health facilities declined from 97.2% to 2016 to 82.1% in 2021 (decrease of 15.1% points; 95% CI (6.5, 22.2); P-value 0.002) (Table 2). The decline in availability of IDSR technical guidelines was mainly due to loss without replacement.\n\nTable 2Comparison of IDSR indicators in selected health facilities, Sierra Leone, 2016–2021Core FunctionIndicatorsFeb_2016July_2016May_2017Aug_2018Feb_2019Aug_2021End Line vs. BaselineP valueN = 71n (%)N = 99n (%)N = 101n (%)N = 126n (%)N = 139n (%)N = 156n (%)Difference (95 CI)CoordinationHealth facility (HF) has IDSR focal person69 (97.2)97 (98.0)100 (99.0)121 (96.0)135 (97.1)153 (98.1)0.9 (-3.2, 7.9)0.67HF IDSR focal person attends district monthly meetings53 (74.6)87 (87.9)93 (92.1)118 (93.7)132 (95.0)150 (96.1)21.5 (11.8, 32.9)\n< 0.0001\nHF has IDSR Technical guidelines69 (97.2)89 (89.9)101 (100.0)102 (81.0)123 (88.5)128 (82.1)15.1 (6.5, 22.2)\n0.002\nCase IdentificationStandard Case definition poster displayed in at least one location in the health facility54 (76.1)84 (84.8)89 (88.1)114 (90.5)127 (91.4)148 (94.9)18.8 (9.2, 30.2)\n< 0.0001\nIDSR focal person conducts active case search at least once a week56 (78.9)54 (54.5)34 (33.7)100 (79.4)75 (54.0)123 (78.8)0.1 (-12.1, 10.7)0.99Reporting of Priority diseasesHCW correctly defines epidemiologic week51 (71.8)93 (93.9)99 (98.0)108 (85.7)132 (95)151 (96.8)25 (15.0, 36.5)\n< 0.0001\nHCW correctly defines zero reporting67 (94.4)94 (94.9)99 (98.0)121 (96.0)138 (99.3)147 (92.2)2.2 (-6.4, 8.5))0.55HCW knows the deadline for submitting weekly IDSR reports65 (91.5)96 (97.0)100 (99.0)115 (91.3)138 (99.3)151 (96.8)5.3 (-0.8, 14.3)0.09Weekly reporting forms available in HF63 (88.7)89 (89.9)96 (95.0)119 (94.4)127 (91.4)150 (96.1)7.4 (0.4, 17.1)\n0.03\nLine listing forms available in HF60 (84.5)78 (78.8)81 (80.2)95 (75.4)114 (82.0)122 (78.2)6.3 (-5.4, 16.0)0.27Case based forms available in HF66 (93.0)79 (79.8)91 (90.1)110 (87.3)114 (82.0)139 (89.1)3.9 (-5.4, 11.0)0.36Rumor logs available in HF39 (54.9)55 (55.6)52 (51.5)78 (61.9)84 (60.4)97 (62.1)7.2 (-6.3, 20.8)0.31Data Analysis and UseHF conducts data basic data analysis28 (39.4)49 (49.5)41 (40.6)51 (40.5)63 (45.3)99 (63.4)24.0 (10.0, 36.7)\n0.0008\nHF have current line graphs showing trends of priority diseases5 (7.0)11 (11.1)16 (15.8)29 (23.0)43 (30.9)72(46.1)39.1 (27.8, 47.9)\n< 0.0001\nOutbreak notification and investigationHF reported outbreak within 12 months12 (16.9)45 (45.5)32 (31.7)10 (7.9)7 (5.0)17(10.9)6.0 (-4.2, 16.1)0.31Outbreak notified to DHMT within 48 h*11 (91.7)29 (64.4)29 (90.6)9 (90.0)5 (71.4)15(88.2%)3.5 (-24.7, 27.1)0.09Monitoring and CommunicationSupervisory visits from DHMT (at least once every three months)44 (62.0)91 (91.9)78 (77.2)79 (62.7)70 (50.4)144 (92.3)30.3(18.6, 42.4)\n< 0.0001\nMobile network available in HF67 (94.4)79 (79.8)91 (90.1)120 (95.2)126 (90.6)144 (92.3)2.1 (-6.5, 8.4)0.57*Denominator is number of outbreaks reported in the preceding 12 monthsHF: Health Facility; IDSR: Integrated Disease Surveillance and Response; HCW: Healthcare worker; DHMT: District Health Management Team\n\nComparison of IDSR indicators in selected health facilities, Sierra Leone, 2016–2021\n*Denominator is number of outbreaks reported in the preceding 12 months\nHF: Health Facility; IDSR: Integrated Disease Surveillance and Response; HCW: Healthcare worker; DHMT: District Health Management Team\n[SUBTITLE] Identification of priority IDSR diseases, conditions and events [SUBSECTION] The proportion of health facilities that displayed standard case definition posters in consultation rooms for use by health workers in identifying cases increased significantly from 76.1% to 2016 to 94.9% in 2021 (increase of 18.8% points; 95% CI 9.2, 30.2); P- value 0.002). The proportion of IDSR focal persons who conducted weekly active case search for priority diseases in the health facilities remained the same at 79% for both baseline and end line although it fluctuated across the years. There was positive trend in adequate knowledge of standard case definitions among health workers for five priority diseases that were assessed as shown in Fig. 2 (Acute flaccid paralysis, Neonatal tetanus, Measles, Cholera and Ebola Virus Disease). Additionally, COVID-19 was added in 2021 and 61% of the health workers had adequate knowledge on its case definition.\n\nFig. 2Knowledge of standard case definitions among interviewed healthcare workers, Sierra Leone, 2016 and 2021\n\nKnowledge of standard case definitions among interviewed healthcare workers, Sierra Leone, 2016 and 2021\nThe proportion of health facilities that displayed standard case definition posters in consultation rooms for use by health workers in identifying cases increased significantly from 76.1% to 2016 to 94.9% in 2021 (increase of 18.8% points; 95% CI 9.2, 30.2); P- value 0.002). The proportion of IDSR focal persons who conducted weekly active case search for priority diseases in the health facilities remained the same at 79% for both baseline and end line although it fluctuated across the years. There was positive trend in adequate knowledge of standard case definitions among health workers for five priority diseases that were assessed as shown in Fig. 2 (Acute flaccid paralysis, Neonatal tetanus, Measles, Cholera and Ebola Virus Disease). Additionally, COVID-19 was added in 2021 and 61% of the health workers had adequate knowledge on its case definition.\n\nFig. 2Knowledge of standard case definitions among interviewed healthcare workers, Sierra Leone, 2016 and 2021\n\nKnowledge of standard case definitions among interviewed healthcare workers, Sierra Leone, 2016 and 2021\n[SUBTITLE] Awareness and adherence to IDSR reporting requirements [SUBSECTION] Knowledge of IDSR reporting requirements was high and by the end line assessment in 2021, almost all interviewed health care workers correctly defined the epidemiologic week, zero reporting and reporting deadlines (Table 2). There was a significant improvement in the proportion of health facilities that submitted all the required surveillance reports (completeness of reporting) from 84.5% to 2016 to 96% in 2021(increase of 11.5% points; 95% CI 3.6, 21.9; P-value 0.003). During the same period, timeliness of IDSR reports improved from 80.3 to 92% (increase of 11.7% points; 95% CI 2.4, 22.9; P-value 0.01).\nKnowledge of IDSR reporting requirements was high and by the end line assessment in 2021, almost all interviewed health care workers correctly defined the epidemiologic week, zero reporting and reporting deadlines (Table 2). There was a significant improvement in the proportion of health facilities that submitted all the required surveillance reports (completeness of reporting) from 84.5% to 2016 to 96% in 2021(increase of 11.5% points; 95% CI 3.6, 21.9; P-value 0.003). During the same period, timeliness of IDSR reports improved from 80.3 to 92% (increase of 11.7% points; 95% CI 2.4, 22.9; P-value 0.01).\n[SUBTITLE] Availability of IDSR reporting tools [SUBSECTION] Availability of the weekly IDSR reporting tool improved from 88.7% to 2016 to 96.1% in 2021 (increase of 7.4% points; 95% CI 0.4, 17.1; P-value 0.03). Availability of other reporting tools did not change significantly and were at 78%, 89% and 62% in 2021 for line listing forms, case-based reporting forms and rumour logbooks, respectively (Table 2).\nAvailability of the weekly IDSR reporting tool improved from 88.7% to 2016 to 96.1% in 2021 (increase of 7.4% points; 95% CI 0.4, 17.1; P-value 0.03). Availability of other reporting tools did not change significantly and were at 78%, 89% and 62% in 2021 for line listing forms, case-based reporting forms and rumour logbooks, respectively (Table 2).\n[SUBTITLE] Data analysis and use at health facility level [SUBSECTION] There was significant improvement in data analysis and use in health facilities over the years. The proportion of health facilities that conducted basic data analysis improved from 39% at baseline to 63% at end line (increase of 24% points; 95% CI 10.0, 36.7; P-value 0.0008). The proportion of health facilities with current line graphs showing trends in occurrence of priority diseases increased significantly from 7 to 46.1% (increase of 39.1% points; 95% CI 27.8, 47.9; P-value < 0.0001) (Table 2).\nThere was significant improvement in data analysis and use in health facilities over the years. The proportion of health facilities that conducted basic data analysis improved from 39% at baseline to 63% at end line (increase of 24% points; 95% CI 10.0, 36.7; P-value 0.0008). The proportion of health facilities with current line graphs showing trends in occurrence of priority diseases increased significantly from 7 to 46.1% (increase of 39.1% points; 95% CI 27.8, 47.9; P-value < 0.0001) (Table 2).\n[SUBTITLE] Outbreak detection and notification [SUBSECTION] The proportion of health facilities that had identified an outbreak within 12 months of the assessment did not change significantly and was 16.9% at baseline compared to 10.9% at end line (P-value 0.31). This was mostly because there were no outbreaks to detect and not for lack of detection capacity. The proportion of identified outbreaks that were notified to the District Health Management Teams within 48 h did not change significantly and were 91.7% at baseline compared to 88.2% at end line (P-value 0.09) (Table 2).\nThe proportion of health facilities that had identified an outbreak within 12 months of the assessment did not change significantly and was 16.9% at baseline compared to 10.9% at end line (P-value 0.31). This was mostly because there were no outbreaks to detect and not for lack of detection capacity. The proportion of identified outbreaks that were notified to the District Health Management Teams within 48 h did not change significantly and were 91.7% at baseline compared to 88.2% at end line (P-value 0.09) (Table 2).\n[SUBTITLE] Feedback, monitoring and communication [SUBSECTION] Quarterly supervisory visits by DHMTs to the health facilities improved from 62% at baseline to 92% (P-value < 000.1) at end line although it fluctuated for other visits (Table 2). Mobile network connectivity remained high throughout the period and 92% of the health facilities assessed at end line had connectivity.\nQuarterly supervisory visits by DHMTs to the health facilities improved from 62% at baseline to 92% (P-value < 000.1) at end line although it fluctuated for other visits (Table 2). Mobile network connectivity remained high throughout the period and 92% of the health facilities assessed at end line had connectivity.\n[SUBTITLE] Availability of routine immunization services and commodities for management of Malaria [SUBSECTION] The proportion of health facilities providing routine immunization services was above 90% in all the assessments with the end line being 96%. The number of immunizing health facilities with functional refrigerators significantly improved from 49.2% at baseline to 66.7% (P-value 0.02) although it fluctuated for other visits (Table 3). Immunizing health facilities with updated temperature monitoring charts significantly improved from 34 to 81% (P-value < 0.0001) while health facilities with all required basic antigens (vaccines) available at the time of the assessment improved from 48 to 83% (P-value < 0.0001). At the end line assessment, there were only 6.7% health facilities with Vaccine Vial Monitor (VVM) at stage three or four which is considered as excessive exposure to high temperatures (Table 3). Malaria management commodities including first line anti-malarial drugs, rapid diagnostic test kits and insecticide treated nets were available in most of the health facilities throughout the assessment period although there was a significant drop for first line anti-malarial drugs and rapid diagnostic test kits at end line assessment in August 2021, mostly due to disruptions associated with COVID-19 (Table 3).\n\nTable 3Assessing routine immunization, malaria commodities and diagnostic capacity in health facilities, Sierra Leone, 2016–2021Programmatic AreaVariableFeb_2016July_2016May_2017Aug_2018Feb_2019Aug_2021End Line vs. BaselineP-value(N = 71)n (%)(N = 99)n (%)(N = 101)n (%)(N = 126)n (%)(N = 139)n (%)(N = 156)n (%)Difference (95 CI)\nVaccination\nHealth facilities providing routine Immunization Services65 (91.5)95 (96.0)96 (95.0)120 (95.2)135 (97.1)150 (96.2)4.7 (-1.5, 13.7)0.14Health facilities with functional refrigerator†32 (49.2)70 (73.7)72 (75.0)71 (59.2)88 (65.2)100 (66.7)17.5 (3.3, 31.2)\n0.0157\nHealth facilities with updated temperature monitoring chart††11 (34.4)16 (22.9)32 (44.4)29 (40.8)37 (42.0)81 (81)46.6 (27.2,62.0)\n< 0.0001\nHealth facilities with all required basic antigens available at the time of the assessment†31(47.7)55(57.9)60(62.5)86 (71.7)103 (76.3)125 (83.3)35.6 (21.9, 48.3)\n< 0.0001\nHeath facilities with expired antigens†2 (3.1)3 (3.2)1 (1)1 (0.8)2 (1.5)2 (1.3)1.8 (-2.3, 9.3)0.37Health facilities with Vaccine Vial Monitor at stage three and four†14 (21.5)16 (16.8)14 (14.6)8 (6.7)9 (6.7)10 (6.7)14.8 (5.1, 26.6)\n0.002\n\nMalaria Commodities\nHealth facilities with 1st line anti-malaria drugs69 (97.2)96 (97.0)99 (98.0)120 (95.2)135 (97.1)120 (76.9)20.3 (11.2, 27.7)\n0.0002\nHealth Facilities with rapid diagnostic kits for diagnosis of malaria67 (94.4)89 (89.9)98 (97.0)120 (95.2)134 (96.4)119 (76.3)18.1 (8.1, 26.1)\n0.001\nHealth facilities with insecticide treated nets55 (77.5)92 (92.9)97 (96.0)111 (88.1)132 (95.0)139 (89.1)11.6 (1.6, 23.3)\n0.0218\n†Denominator is number of health facilities providing immunization services†† Denominator is number of health facilities with a functional fridge\n\nAssessing routine immunization, malaria commodities and diagnostic capacity in health facilities, Sierra Leone, 2016–2021\n†Denominator is number of health facilities providing immunization services\n†† Denominator is number of health facilities with a functional fridge\nThe proportion of health facilities providing routine immunization services was above 90% in all the assessments with the end line being 96%. The number of immunizing health facilities with functional refrigerators significantly improved from 49.2% at baseline to 66.7% (P-value 0.02) although it fluctuated for other visits (Table 3). Immunizing health facilities with updated temperature monitoring charts significantly improved from 34 to 81% (P-value < 0.0001) while health facilities with all required basic antigens (vaccines) available at the time of the assessment improved from 48 to 83% (P-value < 0.0001). At the end line assessment, there were only 6.7% health facilities with Vaccine Vial Monitor (VVM) at stage three or four which is considered as excessive exposure to high temperatures (Table 3). Malaria management commodities including first line anti-malarial drugs, rapid diagnostic test kits and insecticide treated nets were available in most of the health facilities throughout the assessment period although there was a significant drop for first line anti-malarial drugs and rapid diagnostic test kits at end line assessment in August 2021, mostly due to disruptions associated with COVID-19 (Table 3).\n\nTable 3Assessing routine immunization, malaria commodities and diagnostic capacity in health facilities, Sierra Leone, 2016–2021Programmatic AreaVariableFeb_2016July_2016May_2017Aug_2018Feb_2019Aug_2021End Line vs. BaselineP-value(N = 71)n (%)(N = 99)n (%)(N = 101)n (%)(N = 126)n (%)(N = 139)n (%)(N = 156)n (%)Difference (95 CI)\nVaccination\nHealth facilities providing routine Immunization Services65 (91.5)95 (96.0)96 (95.0)120 (95.2)135 (97.1)150 (96.2)4.7 (-1.5, 13.7)0.14Health facilities with functional refrigerator†32 (49.2)70 (73.7)72 (75.0)71 (59.2)88 (65.2)100 (66.7)17.5 (3.3, 31.2)\n0.0157\nHealth facilities with updated temperature monitoring chart††11 (34.4)16 (22.9)32 (44.4)29 (40.8)37 (42.0)81 (81)46.6 (27.2,62.0)\n< 0.0001\nHealth facilities with all required basic antigens available at the time of the assessment†31(47.7)55(57.9)60(62.5)86 (71.7)103 (76.3)125 (83.3)35.6 (21.9, 48.3)\n< 0.0001\nHeath facilities with expired antigens†2 (3.1)3 (3.2)1 (1)1 (0.8)2 (1.5)2 (1.3)1.8 (-2.3, 9.3)0.37Health facilities with Vaccine Vial Monitor at stage three and four†14 (21.5)16 (16.8)14 (14.6)8 (6.7)9 (6.7)10 (6.7)14.8 (5.1, 26.6)\n0.002\n\nMalaria Commodities\nHealth facilities with 1st line anti-malaria drugs69 (97.2)96 (97.0)99 (98.0)120 (95.2)135 (97.1)120 (76.9)20.3 (11.2, 27.7)\n0.0002\nHealth Facilities with rapid diagnostic kits for diagnosis of malaria67 (94.4)89 (89.9)98 (97.0)120 (95.2)134 (96.4)119 (76.3)18.1 (8.1, 26.1)\n0.001\nHealth facilities with insecticide treated nets55 (77.5)92 (92.9)97 (96.0)111 (88.1)132 (95.0)139 (89.1)11.6 (1.6, 23.3)\n0.0218\n†Denominator is number of health facilities providing immunization services†† Denominator is number of health facilities with a functional fridge\n\nAssessing routine immunization, malaria commodities and diagnostic capacity in health facilities, Sierra Leone, 2016–2021\n†Denominator is number of health facilities providing immunization services\n†† Denominator is number of health facilities with a functional fridge", "IDSR focal persons were present in most health facilities during all assessments. When compared to the baseline assessment, the proportion of IDSR focal persons who participated in the monthly district management team meetings increased significantly from 74.6% to 2016 (baseline) to 96.1% at end line in 2021 (increase of 21.5% points; 95% CI (11.8, 32.9); P-value < 0.0001). Conversely, availability of IDSR technical guidelines at the health facilities declined from 97.2% to 2016 to 82.1% in 2021 (decrease of 15.1% points; 95% CI (6.5, 22.2); P-value 0.002) (Table 2). The decline in availability of IDSR technical guidelines was mainly due to loss without replacement.\n\nTable 2Comparison of IDSR indicators in selected health facilities, Sierra Leone, 2016–2021Core FunctionIndicatorsFeb_2016July_2016May_2017Aug_2018Feb_2019Aug_2021End Line vs. BaselineP valueN = 71n (%)N = 99n (%)N = 101n (%)N = 126n (%)N = 139n (%)N = 156n (%)Difference (95 CI)CoordinationHealth facility (HF) has IDSR focal person69 (97.2)97 (98.0)100 (99.0)121 (96.0)135 (97.1)153 (98.1)0.9 (-3.2, 7.9)0.67HF IDSR focal person attends district monthly meetings53 (74.6)87 (87.9)93 (92.1)118 (93.7)132 (95.0)150 (96.1)21.5 (11.8, 32.9)\n< 0.0001\nHF has IDSR Technical guidelines69 (97.2)89 (89.9)101 (100.0)102 (81.0)123 (88.5)128 (82.1)15.1 (6.5, 22.2)\n0.002\nCase IdentificationStandard Case definition poster displayed in at least one location in the health facility54 (76.1)84 (84.8)89 (88.1)114 (90.5)127 (91.4)148 (94.9)18.8 (9.2, 30.2)\n< 0.0001\nIDSR focal person conducts active case search at least once a week56 (78.9)54 (54.5)34 (33.7)100 (79.4)75 (54.0)123 (78.8)0.1 (-12.1, 10.7)0.99Reporting of Priority diseasesHCW correctly defines epidemiologic week51 (71.8)93 (93.9)99 (98.0)108 (85.7)132 (95)151 (96.8)25 (15.0, 36.5)\n< 0.0001\nHCW correctly defines zero reporting67 (94.4)94 (94.9)99 (98.0)121 (96.0)138 (99.3)147 (92.2)2.2 (-6.4, 8.5))0.55HCW knows the deadline for submitting weekly IDSR reports65 (91.5)96 (97.0)100 (99.0)115 (91.3)138 (99.3)151 (96.8)5.3 (-0.8, 14.3)0.09Weekly reporting forms available in HF63 (88.7)89 (89.9)96 (95.0)119 (94.4)127 (91.4)150 (96.1)7.4 (0.4, 17.1)\n0.03\nLine listing forms available in HF60 (84.5)78 (78.8)81 (80.2)95 (75.4)114 (82.0)122 (78.2)6.3 (-5.4, 16.0)0.27Case based forms available in HF66 (93.0)79 (79.8)91 (90.1)110 (87.3)114 (82.0)139 (89.1)3.9 (-5.4, 11.0)0.36Rumor logs available in HF39 (54.9)55 (55.6)52 (51.5)78 (61.9)84 (60.4)97 (62.1)7.2 (-6.3, 20.8)0.31Data Analysis and UseHF conducts data basic data analysis28 (39.4)49 (49.5)41 (40.6)51 (40.5)63 (45.3)99 (63.4)24.0 (10.0, 36.7)\n0.0008\nHF have current line graphs showing trends of priority diseases5 (7.0)11 (11.1)16 (15.8)29 (23.0)43 (30.9)72(46.1)39.1 (27.8, 47.9)\n< 0.0001\nOutbreak notification and investigationHF reported outbreak within 12 months12 (16.9)45 (45.5)32 (31.7)10 (7.9)7 (5.0)17(10.9)6.0 (-4.2, 16.1)0.31Outbreak notified to DHMT within 48 h*11 (91.7)29 (64.4)29 (90.6)9 (90.0)5 (71.4)15(88.2%)3.5 (-24.7, 27.1)0.09Monitoring and CommunicationSupervisory visits from DHMT (at least once every three months)44 (62.0)91 (91.9)78 (77.2)79 (62.7)70 (50.4)144 (92.3)30.3(18.6, 42.4)\n< 0.0001\nMobile network available in HF67 (94.4)79 (79.8)91 (90.1)120 (95.2)126 (90.6)144 (92.3)2.1 (-6.5, 8.4)0.57*Denominator is number of outbreaks reported in the preceding 12 monthsHF: Health Facility; IDSR: Integrated Disease Surveillance and Response; HCW: Healthcare worker; DHMT: District Health Management Team\n\nComparison of IDSR indicators in selected health facilities, Sierra Leone, 2016–2021\n*Denominator is number of outbreaks reported in the preceding 12 months\nHF: Health Facility; IDSR: Integrated Disease Surveillance and Response; HCW: Healthcare worker; DHMT: District Health Management Team", "The proportion of health facilities that displayed standard case definition posters in consultation rooms for use by health workers in identifying cases increased significantly from 76.1% to 2016 to 94.9% in 2021 (increase of 18.8% points; 95% CI 9.2, 30.2); P- value 0.002). The proportion of IDSR focal persons who conducted weekly active case search for priority diseases in the health facilities remained the same at 79% for both baseline and end line although it fluctuated across the years. There was positive trend in adequate knowledge of standard case definitions among health workers for five priority diseases that were assessed as shown in Fig. 2 (Acute flaccid paralysis, Neonatal tetanus, Measles, Cholera and Ebola Virus Disease). Additionally, COVID-19 was added in 2021 and 61% of the health workers had adequate knowledge on its case definition.\n\nFig. 2Knowledge of standard case definitions among interviewed healthcare workers, Sierra Leone, 2016 and 2021\n\nKnowledge of standard case definitions among interviewed healthcare workers, Sierra Leone, 2016 and 2021", "Knowledge of IDSR reporting requirements was high and by the end line assessment in 2021, almost all interviewed health care workers correctly defined the epidemiologic week, zero reporting and reporting deadlines (Table 2). There was a significant improvement in the proportion of health facilities that submitted all the required surveillance reports (completeness of reporting) from 84.5% to 2016 to 96% in 2021(increase of 11.5% points; 95% CI 3.6, 21.9; P-value 0.003). During the same period, timeliness of IDSR reports improved from 80.3 to 92% (increase of 11.7% points; 95% CI 2.4, 22.9; P-value 0.01).", "Availability of the weekly IDSR reporting tool improved from 88.7% to 2016 to 96.1% in 2021 (increase of 7.4% points; 95% CI 0.4, 17.1; P-value 0.03). Availability of other reporting tools did not change significantly and were at 78%, 89% and 62% in 2021 for line listing forms, case-based reporting forms and rumour logbooks, respectively (Table 2).", "There was significant improvement in data analysis and use in health facilities over the years. The proportion of health facilities that conducted basic data analysis improved from 39% at baseline to 63% at end line (increase of 24% points; 95% CI 10.0, 36.7; P-value 0.0008). The proportion of health facilities with current line graphs showing trends in occurrence of priority diseases increased significantly from 7 to 46.1% (increase of 39.1% points; 95% CI 27.8, 47.9; P-value < 0.0001) (Table 2).", "The proportion of health facilities that had identified an outbreak within 12 months of the assessment did not change significantly and was 16.9% at baseline compared to 10.9% at end line (P-value 0.31). This was mostly because there were no outbreaks to detect and not for lack of detection capacity. The proportion of identified outbreaks that were notified to the District Health Management Teams within 48 h did not change significantly and were 91.7% at baseline compared to 88.2% at end line (P-value 0.09) (Table 2).", "Quarterly supervisory visits by DHMTs to the health facilities improved from 62% at baseline to 92% (P-value < 000.1) at end line although it fluctuated for other visits (Table 2). Mobile network connectivity remained high throughout the period and 92% of the health facilities assessed at end line had connectivity.", "The proportion of health facilities providing routine immunization services was above 90% in all the assessments with the end line being 96%. The number of immunizing health facilities with functional refrigerators significantly improved from 49.2% at baseline to 66.7% (P-value 0.02) although it fluctuated for other visits (Table 3). Immunizing health facilities with updated temperature monitoring charts significantly improved from 34 to 81% (P-value < 0.0001) while health facilities with all required basic antigens (vaccines) available at the time of the assessment improved from 48 to 83% (P-value < 0.0001). At the end line assessment, there were only 6.7% health facilities with Vaccine Vial Monitor (VVM) at stage three or four which is considered as excessive exposure to high temperatures (Table 3). Malaria management commodities including first line anti-malarial drugs, rapid diagnostic test kits and insecticide treated nets were available in most of the health facilities throughout the assessment period although there was a significant drop for first line anti-malarial drugs and rapid diagnostic test kits at end line assessment in August 2021, mostly due to disruptions associated with COVID-19 (Table 3).\n\nTable 3Assessing routine immunization, malaria commodities and diagnostic capacity in health facilities, Sierra Leone, 2016–2021Programmatic AreaVariableFeb_2016July_2016May_2017Aug_2018Feb_2019Aug_2021End Line vs. BaselineP-value(N = 71)n (%)(N = 99)n (%)(N = 101)n (%)(N = 126)n (%)(N = 139)n (%)(N = 156)n (%)Difference (95 CI)\nVaccination\nHealth facilities providing routine Immunization Services65 (91.5)95 (96.0)96 (95.0)120 (95.2)135 (97.1)150 (96.2)4.7 (-1.5, 13.7)0.14Health facilities with functional refrigerator†32 (49.2)70 (73.7)72 (75.0)71 (59.2)88 (65.2)100 (66.7)17.5 (3.3, 31.2)\n0.0157\nHealth facilities with updated temperature monitoring chart††11 (34.4)16 (22.9)32 (44.4)29 (40.8)37 (42.0)81 (81)46.6 (27.2,62.0)\n< 0.0001\nHealth facilities with all required basic antigens available at the time of the assessment†31(47.7)55(57.9)60(62.5)86 (71.7)103 (76.3)125 (83.3)35.6 (21.9, 48.3)\n< 0.0001\nHeath facilities with expired antigens†2 (3.1)3 (3.2)1 (1)1 (0.8)2 (1.5)2 (1.3)1.8 (-2.3, 9.3)0.37Health facilities with Vaccine Vial Monitor at stage three and four†14 (21.5)16 (16.8)14 (14.6)8 (6.7)9 (6.7)10 (6.7)14.8 (5.1, 26.6)\n0.002\n\nMalaria Commodities\nHealth facilities with 1st line anti-malaria drugs69 (97.2)96 (97.0)99 (98.0)120 (95.2)135 (97.1)120 (76.9)20.3 (11.2, 27.7)\n0.0002\nHealth Facilities with rapid diagnostic kits for diagnosis of malaria67 (94.4)89 (89.9)98 (97.0)120 (95.2)134 (96.4)119 (76.3)18.1 (8.1, 26.1)\n0.001\nHealth facilities with insecticide treated nets55 (77.5)92 (92.9)97 (96.0)111 (88.1)132 (95.0)139 (89.1)11.6 (1.6, 23.3)\n0.0218\n†Denominator is number of health facilities providing immunization services†† Denominator is number of health facilities with a functional fridge\n\nAssessing routine immunization, malaria commodities and diagnostic capacity in health facilities, Sierra Leone, 2016–2021\n†Denominator is number of health facilities providing immunization services\n†† Denominator is number of health facilities with a functional fridge", "Through the support supervision data collected and analysed after each visit to the districts, the country was able to monitor trends of key IDSR indicators. In general, health facilities performed well over the years in several assessment areas (Tables 2 and 3). However, there were also some other areas that did not improve much due to several challenges including inadequate funding and COVID-19 pandemic. The funding challenge was partly resolved by sharing the supervision feedback with stakeholders who could provide financial support to address some of the challenges. For example, WHO and other development partners supported the MoHS to print and distribute surveillance tools to health facilities and to conduct refresher trainings for health workers focusing on case definitions of priority diseases, conditions and events listed in the IDSR technical guidelines. However, as the findings of the availability of surveillance tools shows (Table 2), only the weekly reporting tool had become more available over the years compared to the other tools (line listing forms, case-based reporting forms and rumour logbooks). This is because the weekly tool is the one more frequently used compared to the others and hence development partners were more likely to support in its printing.\nSurveillance data is more accurate in measuring burden of disease if it is representative and timely [9]. IDSR report completeness (proportion of health facilities submitting reports) and timeliness improved during the review period and may be partly attributed to availability of IDSR focal persons in most health facilities who were familiar with IDSR reporting obligations and were equipped with the tools necessary for reporting. Lack of IDSR focal persons and unavailability of technical guidelines are often associated with low weekly IDSR reporting rates and timeliness [17]. Gradual migration from paper based to electronic reporting of IDSR data in Sierra Leone at the health facility level starting mid-2017 may have improved timeliness of IDSR reports by reducing report transmission time [18]. Reporting completeness and timeliness rates in these assessments are comparable to those found in a similar assessment in Uganda [19].\nOver the years, there was overall improvement in knowledge of case definitions for priority conditions among health workers which could have been due to increased availability of case definition posters in health facilities. However, the proportion of health workers with adequate knowledge of the case definitions was less than 80% in all the diseases sampled which means that not all health workers were using the case definition posters (Fig. 2). For Ebola Virus Disease, the suboptimal knowledge of the case definition may have been due to changes in case definitions (from outbreak case definition to a routine surveillance case definition). Low knowledge on the case definition for Cholera was likely because the country had not recorded any case since the last outbreak in 2013.\nSyndromic surveillance is important in low resource settings where laboratory confirmation is not always readily available as is currently the case in Sierra Leone. Inadequate knowledge on case definitions leads to low levels of suspicion or misclassification of cases. Hence, it is important that healthcare workers use clinical signs and symptoms as depicted in the standard case definition posters to suspect cases and initiate investigation. The Ministry of Health and Sanitation must therefore do more to engage the health workers in all health facilities to ensure that they read and master the case definitions for the various diseases since the posters are available in most health facilities.\nData analysis at the health facilities improved markedly over time from 2016 to 2021 (Table 2) and the reasons given by the health workers for this improvement were mainly training on data analysis skills, provision of data analysis graphs and tablets for reporting. In deed these are important elements in data analysis as lack of computers and technical capacity has been reported in other countries as being responsible for poor data analysis [19]. Limited laboratory diagnostic capacity was observed even for organisms such as Vibrio cholerae that have caused large magnitude epidemics in Sierra Leone in the past [20]. This limits the contribution of laboratories to outbreak detection, guiding case management and monitoring trends of priority diseases as specimen referral increases turnaround time for results.\nWhile routine vaccination services were available in most health facilities, gaps were observed in maintenance of cold chain that could have compromised the quality of vaccines. It is worth noting that the gaps reduced in subsequent assessments with significant improvement in the number of health facilities with all basic vaccines and functional cold chain equipment. Availability of first line antimalarial drugs and rapid diagnostic kits for malaria detection dropped during the end line assessment in 2021 and this could have been attributed to the COVID-19 pandemic which has caused constraint on available resources including for malaria [21].\nThis study had a few limitations. Even though two national IDSR support supervision visits were planned each year, only one was conducted annually except in 2016 when two were made. This was mostly due to competing priorities or resource constraints. While two visits per year would have provided us with better trends, it did not affect the quality of the data. Support supervision was however conducted by the districts on a quarterly basis although the comprehensive electronic checklist was not always used and this data could therefore not be included. Additionally, due to the COVID-19 pandemic, no national IDSR support supervision was conducted in 2020 due to travel restrictions to the districts for most part of the year. However, there were other visits made to the districts as part of the response to the pandemic.", "The IDSR system is now well established in Sierra Leone. The support supervision visits that were done using an integrated electronic tool contributed to health systems strengthening through longitudinal tracking of core IDSR indicators and other program indicators such as essential malaria commodities and availability and status of routine vaccines. The feedback that was provided to all levels of healthcare including the national program and development partners supported to address the gaps identified and hence improved performance over the years. MoHS should entrench the supportive supervision approach by the national and district teams using electronic tools to assure sustained monitoring of IDSR and other programs." ]
[ null, null, null, null, null, null, null, null, "results", null, null, null, null, null, null, null, null, "discussion", "conclusion" ]
[ "Public Health", "Integrated Disease Surveillance and Response", "Monitoring", "Technology" ]
Hyperuricaemia and its association with other risk factors for cardiovascular diseases: A population-based study.
36266776
Cardiovascular diseases are very common in the general population, and several factors play a role in their development. The purpose of this study was to investigate the relationship between hyperuricaemia and other cardiovascular disease risk factors.
INTRODUCTION
This cross-sectional study was conducted on 1008 people over the 15-year-old general population in Kerman, Iran. The blood samples of all patients were analysed for the uric acid serum level, and they completed a checklist including physical activity, previous history of hypertension and diabetes, smoking and opium.
METHODS
A number of 1008 cases of people were entered into the study. According to the results of this study, 254 patients had uric acid levels above the 75th percentile (6 mg/dl in males, and 5 mg/dl in females). No significant difference was observed between gender (p = .249) and age groups (p = .125) of people with and without hyperuricaemia. The prevalence of overweight/obesity (p < .001), hypertension (p = .004) and low physical activity (p = .033) was significantly higher in patients with hyperuricaemia. The duration of hypertension was significantly higher in hyperuricaemic individuals (p = .022). Overweight/obesity (OR = 2.67; 95% CI = 1.87-3.82) and hypertension (OR = 1.40; 95% CI = 1.02-1.93) were two significant independent factors that contributed to the increased risk of hyperuricaemia in the subjects.
RESULTS
The uric acid serum level is higher in people with hypertension and overweight/obesity. Hyperuricaemia increases the risk of cardiovascular events, which can be prevented by determining the appropriate strategy for the early diagnosis and treatment of this metabolic disorder.
CONCLUSION
[ "Male", "Female", "Humans", "Adolescent", "Hyperuricemia", "Uric Acid", "Cardiovascular Diseases", "Cross-Sectional Studies", "Overweight", "Risk Factors", "Hypertension", "Obesity" ]
9659661
null
null
null
null
RESULTS
A number of 1008 cases were entered into the study. The relationship between the studied variables and the prevalence of hyperuricaemia is shown in Table 1. According to the results of this study, 254 patients had uric acid levels above the 75th percentile (more than 6 mg/dl in males, and 5 mg/dl in females). No significant difference was seen between gender (p = .249) and age groups (p = .125) of people with and without hyperuricaemia. The prevalence of overweight/obesity was significantly higher in patients with hyperuricaemia (p < .001). The prevalence of low physical activity in patients with hyperuricaemia was significantly higher than in those with normal uric acid levels (p = .033). The prevalence of smoking (p = 406), tobacco consumption (p = .669) and opium addiction (p = .261) in people with and without hyperuricaemia did not show a significant difference. The frequency of hypertension was significantly higher in people with high uric acid levels compared to people with normal uric acid levels (31.9% and 22.8%, respectively; p = .004). The duration of hypertension was significantly higher in hyperuricaemic individuals (p = .022). The prevalence of diabetes (p = .794) and the duration of diabetes (p = .371) in people with and without hyperuricaemia did not show a significant difference. The duration of hypertension and diabetes was converted into categorized variables according to their median (48 and 72 months, respectively). The prevalence of a longer period of hypertension (more than 48 months) in people with hyperuricaemia was significantly higher than in those without hyperuricaemia (p = .025). The prevalence of a longer duration of diabetes (more than 72 months) between the two groups of people with and without hyperuricaemia was similar (p = .986). The relationship between the studied variables and the presence of hyperuricaemia Abbreviations: BMI: Body mass index, MET: Metabolic Equivalent (The amount of energy consumed while sitting (equivalent to 1 kcal/kg/h)). Chi‐square test. Mann–Whitney test. In order to determine the potential predictors of hyperuricaemia, univariable and multivariable survey logistic regression models were used on variables whose frequency was significantly different in individuals with and without hyperuricaemia (Table 2). Based on the univariate logistic regression model, the presence of overweight/obesity, low physical activity and hypertension increased the chances of hyperuricaemia in the subjects by 2.85, 1.91, compared to those with intense physical activity and 1.58, respectively (p < .001, p = .019 and p = .004, respectively). According to the results of a multivariate analysis on the three variables, the amount of physical activity did not have a significant effect on increasing the chances of hyperuricaemia and the two variables of overweight/obesity (OR = 2.67; 95% CI = 1.87–3.82) and hypertension (OR = 1.40; 95% CI = 1.02–1.93) significantly increased the chances of hyperuricaemia in the subjects. Univariate and multivariate logistic regression models for identifying determinants of hyperuricaemia Abbreviations: BMI: Body mass index, MET: Metabolic Equivalent (The amount of energy consumed while sitting (equivalent to 1 kcal/kg/h)).
CONCLUSION
Our study showed that two independent factors for the development of hyperuricaemia are hypertension and overweight/obesity. Since hyperuricaemia is one of the influential factors in cardiovascular events, these events can be prevented by determining the appropriate strategy for early diagnosis and treatment of this metabolic disorder. Considering that our study was conducted in an urban population and environmental and genetic factors can also be involved in the relationship between hyperuricaemia and other cardiovascular disease risk factors, cohort studies with a larger sample size are needed in this field.
[ "INTRODUCTION", "Sample size estimation", "Statistical analysis", "AUTHOR CONTRIBUTIONS", "FUNDING INFORMATION", "ETHIC STATEMENT" ]
[ "Cardiovascular diseases are one of the most important causes of death in the world, and their pathogenesis is such that they are affected by several risk factors such as obesity, diabetes and hypertension.\n1\n Increased serum uric acid levels can lead to endothelial dysfunction resulting in atherosclerosis by releasing reactive oxygen species and is reported as one of the risk factors for cardiovascular diseases.\n2\n\n\nIn the process of purine decomposition, uric acid is the final product.\n3\n A condition in which serum uric acid concentration is high but there are no signs or symptoms of uric acid deposition is called asymptomatic hyperuricaemia. Although gout can occur in a person with any rate of uric acid, about two‐thirds of hyperuricaemic people remain asymptomatic.\n4\n Different studies in different races have had different results on the prevalence of hyperuricaemia in people's age and gender.\n5\n, \n6\n\n\nPrevalence of hyperuricaemia is different in viewpoints of race, age and gender groups, and this serum factor has been mentioned as a risk factor for coronary artery diseases; in this study, the relationship between cardiovascular risk factors and hyperuricaemia was investigated.", "To determine the sample size based on the study of Zhu et al., the prevalence of hyperuricaemia was considered to be 21.4%, and the minimum required sample size was calculated using the following formula\n9\n and considering Z = 1.96, p = .21, and D = 0.03:\nn=Z2P1−Pd2=1.9620.211−0.210.032=708\n\n\nTo increase the power of our regression, we investigated 1008 people in our study.", "To determine the potential predictors of hyperuricaemia, univariable and multivariable survey logistic regression models were used. Variables significantly different in individuals with and without hyperuricaemia were included in the regression models, and the odds ratios (AOR) were reported.", "Farzaneh Yazdi, Mohammad Reza Shakibi, Hamid Najafipour, Amir Baniasad, Mohammad Javad Najafzadeh and Samira Sistani designed the study and collected data, Amir Baniasad, Mohammad Javad Najafzadeh wrote the first draft of the paper and submitted the manuscript, Fatemeh Yazdi contributed to writing and revision of the manuscript. All authors contributed to finalizing the manuscript.", "This research project was funded by the Kerman University of Medical Sciences.", "The study protocol was reviewed and approved by the ethics committee of Kerman University of Medical Sciences (ethic code: IR.KMU.REC.1394.59). Informed consent was obtained from all participants in the study." ]
[ null, null, null, null, null, null ]
[ "INTRODUCTION", "MATERIALS AND METHODS", "Sample size estimation", "Statistical analysis", "RESULTS", "DISCUSSION", "CONCLUSION", "AUTHOR CONTRIBUTIONS", "FUNDING INFORMATION", "CONFLICT OF INTEREST", "ETHIC STATEMENT" ]
[ "Cardiovascular diseases are one of the most important causes of death in the world, and their pathogenesis is such that they are affected by several risk factors such as obesity, diabetes and hypertension.\n1\n Increased serum uric acid levels can lead to endothelial dysfunction resulting in atherosclerosis by releasing reactive oxygen species and is reported as one of the risk factors for cardiovascular diseases.\n2\n\n\nIn the process of purine decomposition, uric acid is the final product.\n3\n A condition in which serum uric acid concentration is high but there are no signs or symptoms of uric acid deposition is called asymptomatic hyperuricaemia. Although gout can occur in a person with any rate of uric acid, about two‐thirds of hyperuricaemic people remain asymptomatic.\n4\n Different studies in different races have had different results on the prevalence of hyperuricaemia in people's age and gender.\n5\n, \n6\n\n\nPrevalence of hyperuricaemia is different in viewpoints of race, age and gender groups, and this serum factor has been mentioned as a risk factor for coronary artery diseases; in this study, the relationship between cardiovascular risk factors and hyperuricaemia was investigated.", "This study was a sub‐analyse of collected data about the risk factors of coronary artery diseases (i.e., Kerman Coronary Artery Disease Risk Factor Study; KERCADRS). This cross‐sectional and descriptive study was conducted in Kerman city on 1008 people who were over 15 years old. The sampling method of the studied people was in the form of a cluster from the entire population of the city. These people were selected from 720,000 Kerman residents, which is the biggest city in Iran's southeast, and they are the representative of this population. In the first phase of the KERCADRS, 250 postal codes (called seeds) were randomly selected from the list of city residents registered in the city post office. First, the location of each seed was determined on the map, and then, each of them was contacted. After identifying the number of people in each family, people over 15 years old were invited to participate in the study. If people do not answer despite calling twice, their neighbour was replacing them based on their seed address. In the first phase of the KERCADRS study, the collection of samples continued until 24 people was collected in each cluster. In the second phase of this study, people were called again, and 1008 people who met the criteria for entering our study were included.\nThe study protocol was reviewed and approved by the ethics committee of Kerman University of Medical Sciences (ethic code: IR.KMU.REC.1394.59). All study participants gave informed consent. The method of data collection has already been published in the study of Najafipour et al.\n7\n\n\nSome risk factors, including gender, age, physical activity, smoking status, using tobacco products, body mass index (BMI), opium addiction and history of hypertension or diabetes, the duration of hypertension, and duration of diabetes disease, were reviewed in our study.\nPeople's height was measured in a standing position without shoes. All participants in the study were weighed with minimal clothing and without shoes on a digital scale (Seca, model 707, Germany). In order to calculate the body mass index (BMI) of the participants, the weight (kg) of the patients was divided by the square of their height (m2). Patients with a BMI between 25–29.9 Kg/m2 were considered overweight, and patients over 30 Kg/m2 were considered obese.\nAccording to the guidelines of the World Health Organization, patients' blood pressure was measured with a standard manometer (mercury manometer RICHTER, Germany) after a 10‐min rest, and if it was abnormal, it was measured again after 1 h.\nWHO Global Physical Activity Questionnaire (GPAQ) and Metabolic Equivalent (MET) were used to evaluate the amount and intensity of daily physical activity, respectively. A MET is defined as the amount of energy consumed while sitting (equivalent to 1 kcal/kg/h). Moderate to severe physical activity is defined as four times and eight times calorie intake compared to sitting quietly. The amount of activity more than 3000 MET and less than 1500 MET per week was considered intense and low physical activity, respectively.\n8\n\n\nA trained interviewer asked participants about smoking, tobacco use and opium addiction. People who routinely smoked cigarettes during data collection were considered smokers. Also, people who continuously used tobacco products or opium during data collection were considered positive for tobacco use and opium addiction, respectively.\nIn addition to determining demographic features of the studied people, being informed of this laboratory finding and drug treatment in the case of pre‐notice, a sample of fasting blood was taken (after 12–14 h of fasting) to determine the level of serum uric acid and fasting plasma glucose (FPG) for each person.\nFPG (KIMIA Kit, code 890410, Iran) and uric acid (measured by the colorimetric method.) were measured for all patients. In our study, the hyperuricaemia cut‐off was considered to be greater than the 75th percentile, which was 6 mg/dl in males and 5 mg/dl in females.\nThe senior researcher trained all the interviewees prior to data collection, and he also continuously evaluated the validity of the data collected. Although agreement indices were not measured in this study, careful quality control was performed to minimize errors.\nSPSS software version 16 (SPSS Inc.) was used to analyse data. Categorical variables were reported as numbers and percentages, and a chi‐square test was used to compare them between the two groups. Continuous variables were reported as medians (with interquartile ranges [IQRs]) based on their abnormal distribution according to the Kolmogorov–Smirnov test, and the Mann–Whitney test was used to compare them between individuals with and without hyperuricaemia. p < .05 was considered as significant.\n[SUBTITLE] Sample size estimation [SUBSECTION] To determine the sample size based on the study of Zhu et al., the prevalence of hyperuricaemia was considered to be 21.4%, and the minimum required sample size was calculated using the following formula\n9\n and considering Z = 1.96, p = .21, and D = 0.03:\nn=Z2P1−Pd2=1.9620.211−0.210.032=708\n\n\nTo increase the power of our regression, we investigated 1008 people in our study.\nTo determine the sample size based on the study of Zhu et al., the prevalence of hyperuricaemia was considered to be 21.4%, and the minimum required sample size was calculated using the following formula\n9\n and considering Z = 1.96, p = .21, and D = 0.03:\nn=Z2P1−Pd2=1.9620.211−0.210.032=708\n\n\nTo increase the power of our regression, we investigated 1008 people in our study.\n[SUBTITLE] Statistical analysis [SUBSECTION] To determine the potential predictors of hyperuricaemia, univariable and multivariable survey logistic regression models were used. Variables significantly different in individuals with and without hyperuricaemia were included in the regression models, and the odds ratios (AOR) were reported.\nTo determine the potential predictors of hyperuricaemia, univariable and multivariable survey logistic regression models were used. Variables significantly different in individuals with and without hyperuricaemia were included in the regression models, and the odds ratios (AOR) were reported.", "To determine the sample size based on the study of Zhu et al., the prevalence of hyperuricaemia was considered to be 21.4%, and the minimum required sample size was calculated using the following formula\n9\n and considering Z = 1.96, p = .21, and D = 0.03:\nn=Z2P1−Pd2=1.9620.211−0.210.032=708\n\n\nTo increase the power of our regression, we investigated 1008 people in our study.", "To determine the potential predictors of hyperuricaemia, univariable and multivariable survey logistic regression models were used. Variables significantly different in individuals with and without hyperuricaemia were included in the regression models, and the odds ratios (AOR) were reported.", "A number of 1008 cases were entered into the study. The relationship between the studied variables and the prevalence of hyperuricaemia is shown in Table 1. According to the results of this study, 254 patients had uric acid levels above the 75th percentile (more than 6 mg/dl in males, and 5 mg/dl in females). No significant difference was seen between gender (p = .249) and age groups (p = .125) of people with and without hyperuricaemia. The prevalence of overweight/obesity was significantly higher in patients with hyperuricaemia (p < .001). The prevalence of low physical activity in patients with hyperuricaemia was significantly higher than in those with normal uric acid levels (p = .033). The prevalence of smoking (p = 406), tobacco consumption (p = .669) and opium addiction (p = .261) in people with and without hyperuricaemia did not show a significant difference. The frequency of hypertension was significantly higher in people with high uric acid levels compared to people with normal uric acid levels (31.9% and 22.8%, respectively; p = .004). The duration of hypertension was significantly higher in hyperuricaemic individuals (p = .022). The prevalence of diabetes (p = .794) and the duration of diabetes (p = .371) in people with and without hyperuricaemia did not show a significant difference. The duration of hypertension and diabetes was converted into categorized variables according to their median (48 and 72 months, respectively). The prevalence of a longer period of hypertension (more than 48 months) in people with hyperuricaemia was significantly higher than in those without hyperuricaemia (p = .025). The prevalence of a longer duration of diabetes (more than 72 months) between the two groups of people with and without hyperuricaemia was similar (p = .986).\nThe relationship between the studied variables and the presence of hyperuricaemia\nAbbreviations: BMI: Body mass index, MET: Metabolic Equivalent (The amount of energy consumed while sitting (equivalent to 1 kcal/kg/h)).\nChi‐square test.\nMann–Whitney test.\nIn order to determine the potential predictors of hyperuricaemia, univariable and multivariable survey logistic regression models were used on variables whose frequency was significantly different in individuals with and without hyperuricaemia (Table 2). Based on the univariate logistic regression model, the presence of overweight/obesity, low physical activity and hypertension increased the chances of hyperuricaemia in the subjects by 2.85, 1.91, compared to those with intense physical activity and 1.58, respectively (p < .001, p = .019 and p = .004, respectively). According to the results of a multivariate analysis on the three variables, the amount of physical activity did not have a significant effect on increasing the chances of hyperuricaemia and the two variables of overweight/obesity (OR = 2.67; 95% CI = 1.87–3.82) and hypertension (OR = 1.40; 95% CI = 1.02–1.93) significantly increased the chances of hyperuricaemia in the subjects.\nUnivariate and multivariate logistic regression models for identifying determinants of hyperuricaemia\nAbbreviations: BMI: Body mass index, MET: Metabolic Equivalent (The amount of energy consumed while sitting (equivalent to 1 kcal/kg/h)).", "According to the results of our study, overweight/obesity and hypertension increase the risk of hyperuricaemia in the general population. Due to the increasing prevalence of hyperuricaemia, it should be considered as an important health issue. So far, several studies have emphasized the relationship between the presence of hyperuricaemia and cardiovascular risk factors, including hypertension and metabolic syndrome\n10\n, \n11\n, \n12\n as in a recent cross‐sectional study; Yazdi et al.\n13\n showed that the ratio of uric acid to high‐density lipoprotein (HDL) cholesterol is significantly higher in people with metabolic syndrome than in healthy individuals. Despite the similarity of the results of the present study with previous studies, there are also differences. Different study designs and different criteria for confirming hyperuricaemia in patients can be the main cause of these differences.\nIn previous studies, inflammatory response, oxidative stress, insulin resistance, endothelial dysfunction and endoplasmic reticulum stress have been mentioned as the main mechanisms of hyperuricaemia's effect on the development of cardiovascular diseases.\n14\n In our study, hypertension was associated with increased uric acid levels in patients. In line with our study in Zhang et al.,\n15\n a prospective cohort study of 6424 patients, hypertension was also associated with an increased risk of hyperuricaemia. In another cross‐sectional study of 3119 people over the age of 50 in a rural population in China, high blood pressure increased the risk of high uric acid levels.\n10\n Although the mechanism of association between hyperuricaemia and hypertension is still unclear, according to the findings of in vitro studies, uric acid causes damage to endothelial cells, vasoconstriction and high blood pressure by inhibiting the release of nitric oxide from endothelial cells, activating the renin‐angiotensin system and oxidative stress.\n16\n Chen et al.’s\n17\n study of animal models and generally healthy early Parkinson's disease patients found no association between uric acid levels and blood pressure. Another possible cause of elevated uric acid in hypertensive patients is the use of certain drugs, including diuretics, β‐blockers, angiotensin‐converting enzyme inhibitors and non‐losartan angiotensin II receptor blockers.\n18\n In our study, the duration of hypertension in patients with hyperuricaemia was significantly longer, which could be due to the long‐term use of antihypertensive drugs, so more studies should be performed on new cases of hypertension to study the relationship between hypertension and uric acid levels.\nIn our study, overweight/obesity was associated with hyperuricaemia. In a study by Choi et al.,\n19\n BMI higher than 25 was significantly associated with an increased risk of gout in patients, and weight loss was a protective factor against gout. The link between obesity and hyperuricaemia can be due to insulin resistance caused by oxidative stress and inflammatory processes. Hyperinsulinaemia is associated with insulin resistance and increases the activity and expression of urate transporter protein 1 (URAT1) and, consequently, uric acid reabsorption through renal proximal tubular cells.\n14\n, \n20\n However, in our study, no association was found between hyperuricaemia and diabetes. In a study by Krishnan et al.,\n5\n elevated serum urate levels were an independent risk factor for type 2 diabetes in non‐obese patients. In a study by Van der Schaft et al.,\n6\n serum uric acid was associated with incidental prediabetes in patients with normal blood sugar but not with the development of type 2 diabetes in prediabetic subjects. In a study conducted on mice by Lu et al.,\n21\n there was no association between high uric acid levels and diabetes, and they concluded that high uric acid levels did not cause diabetes but could accelerate it by damaging glucose tolerance.\nIn our study, although less physical activity significantly increased the risk of hyperuricaemia in univariate analysis, the relationship between physical activity and hyperuricaemia was not significant in multivariate analysis after adjustment of the effect of overweight/obesity and hypertension variables. Dong et al.,\n22\n in a study of 38,855 rural China populations, showed that regular physical activity independently reduced serum uric acid levels and the prevalence of hyperuricaemia. However, in the regression analysis performed in the study of Dong et al.,\n22\n adjustment was done for BMI but not for hypertension. In the study by Qi et al.,\n10\n low physical activity in men was associated with an increased risk of high uric acid levels. It seems that racial differences in the relationship between physical activity and hyperuricaemia need to be further evaluated.\nIn this study, the prevalence of hyperuricaemia has no significant relationship with age groups, smoking, tobacco consumption and opium addiction. Despite our study, Hanna et al.\n23\n showed that uric acid serum level is significantly lower in smoking people, but these people did not suffer from hyperuricaemia in our study. It was shown in another study conducted by Tomita et al.\n24\n that, like our study, uric acid serum levels and hyperuricaemia in smoking people have no significant difference with people who have never smoked statistically. In this study, the majority of cases belonged to the age group of 41 to 60 (45.1%) years, which probably people aged 41–60 had a higher tendency to participate in the study due to their awareness of the benefits of the study.\nOne of the strengths of our study was its population‐based and appropriate sample size. The sampling method in our study was cluster. Among the advantages of this method is that it is economical in terms of cost and time compared to probability sampling methods. But the accuracy of this method is lower than simple random sampling. Our study also had limitations, including the fact that many factors, including smoking and the duration of hypertension, were self‐reported, and there was a possibility of bias in these variables. Another limitation of the study was the possible effects of the drugs used and their lack of evaluation, including diuretics, aspirin and dietary habits on patients' uric acid levels separately. Another limitation of our study was that, unfortunately, the dietary profile of the participants was not recorded at the time of sampling. Despite all the limitations mentioned, our findings are helpful in determining the appropriate strategy to identify people who are most at risk for hyperuricaemia for the prevention and early treatment of this disorder.", "Our study showed that two independent factors for the development of hyperuricaemia are hypertension and overweight/obesity. Since hyperuricaemia is one of the influential factors in cardiovascular events, these events can be prevented by determining the appropriate strategy for early diagnosis and treatment of this metabolic disorder.\nConsidering that our study was conducted in an urban population and environmental and genetic factors can also be involved in the relationship between hyperuricaemia and other cardiovascular disease risk factors, cohort studies with a larger sample size are needed in this field.", "Farzaneh Yazdi, Mohammad Reza Shakibi, Hamid Najafipour, Amir Baniasad, Mohammad Javad Najafzadeh and Samira Sistani designed the study and collected data, Amir Baniasad, Mohammad Javad Najafzadeh wrote the first draft of the paper and submitted the manuscript, Fatemeh Yazdi contributed to writing and revision of the manuscript. All authors contributed to finalizing the manuscript.", "This research project was funded by the Kerman University of Medical Sciences.", "The authors declare that they have no conflict of interest.", "The study protocol was reviewed and approved by the ethics committee of Kerman University of Medical Sciences (ethic code: IR.KMU.REC.1394.59). Informed consent was obtained from all participants in the study." ]
[ null, "materials-and-methods", null, null, "results", "discussion", "conclusions", null, null, "COI-statement", null ]
[ "diabetes", "hypertension", "hyperuricaemia", "obesity", "overweight", "physical activity" ]
Second Malignant Tumors and Non-Tumor Causes of Death for Patients With Penile Cancer During Their Survivorship.
36267038
The aim was to evaluate the causes of death for patients with localized, regional and metastatic penile cancer (PeCa) after diagnosis.
BACKGROUND
PeCa patients diagnosed during 2004-2018 in the Surveillance, Epidemiology, and End Results program database were identified. Causes of deaths including PeCa, second malignant tumors (SMTs) and non-tumor diseases were analyzed, as well as the standardized mortality ratio (SMR) of each cause.
METHODS
For localized PeCa, 800 of 2155 patients died during the follow-up. 24.9% of all deaths were due to PeCa. 18.0% and 57.1% deaths were due to SMTs and non-tumor causes. Main SMTs included cancers of lung and bronchus (n = 40) and skin (n = 11) with significantly increased SMRs of 1.71 (1.22-2.33) and 4.82 (2.41-8.63). Mortality risks of other SMTs were mostly similar with the general populations. Main causes of non-tumor diseases included diseases of heart [n = 172, SMR: 1.66 (1.42-1.93)], COPD and allied cond [n = 38, SMR: 1.63 (1.15-2.24)], and cerebrovascular diseases [n = 33, SMR: 1.71 (1.17-2.4)]. For regional PeCa, 679 of 1310 patients died including 43.5% PeCa, 14.8% SMTs and 26.6% non-tumor causes. The mortality risks of cancers from lung and bronchus [SMR: 2.41 (1.53-3.62)], skin [SMR: 6.41 (2.35-13.95)] and testis [SMR: 149.35 (18.09-539.5)] were significantly increased. Main non-tumor causes of death included diseases of heart [n = 71, SMR: 1.77 (1.38-2.23)], COPD and allied cond [n = 17, SMR: 1.85 (1.08-2.95)] and diabetes mellitus [n = 16, SMR: 3.62 (2.07-5.88)]. For distant diseases, 109 of 132 patients died including 76 (69.7%) died for PeCa itself, 24 (22.0%) died for SMTs and 9 (8.3%) died for non-tumor diseases. The majority of PeCa deaths (67.1%) and SMTs deaths (79.2%) occurred within 1 year after the diagnosis of PeCa.
RESULTS
We firstly analyzed the SMTs and non-tumor causes of death and morality risks of each cause for PeCa patients, which provided valuable information for PeCa patients on disease prevention and health care during their survivorship.
CONCLUSIONS
[ "Male", "Humans", "Cause of Death", "Penile Neoplasms", "Survivorship", "Risk Factors", "Pulmonary Disease, Chronic Obstructive" ]
9597479
Introduction
Penile cancer (PeCa) is a rare malignancy worldwide, accounting for less than 1% of all malignancies in men.1 The overall incidence rate is approximately .69 cases per 100 000 persons in the US.2 It was estimated that the number of new PeCa patients was 34 475 and deaths cases was 15 138 each year worldwide.3 The survival rates still vary greatly with the stages of patients’ disease. It was reported that about 40% of new cases were diagnosed with localized PeCa, with a 5-year overall survival rate of approximately 90%.4 The survival rates decrease dramatically once patients are in the regional or distant metastasis stage. The 5-year overall survival rate is approximately 80% with unilateral inguinal lymph node involvement, 10%-20% with bilateral or pelvic lymph node involvement, and <10% with extranodal extension.4 Since the 1990s, substantial progress has been made in the prevention, diagnosis and treatment of PeCa. The prognosis has been greatly improved in PeCa patients, especially those who have not progressed to the metastatic stage. With the prolonged survival time, the proportion of other causes of death such as second malignant tumors (SMTs) and non-tumor diseases has been increasing among all deaths of PeCa patients. Since few previous studies have focused exclusively on other causes of death of PeCa patients, an analysis of causes of death among PeCa patients is warranted. Our study aimed to evaluate the main causes of death including SMTs and non-cancer causes for PeCa patients, and calculate the standardized mortality ratio (SMR) for each reason of death compared with the normal population.
Methods
[SUBTITLE] Data Sources [SUBSECTION] The data of this study was derived from the Surveillance, Epidemiology, and End Results (SEER) program 18 registries, which is an authoritative source of information on cancer incidence and survival in the United States. SEER currently collects and publishes cancer incidence and survival data from population-based cancer registries covering approximately 27.8% of the U.S. population, and it collects data concerning patient demographics, tumor morphology, stage at diagnosis, primary tumor site, the first course of treatment, and follow-up for vital status. Meanwhile, the data for the general population comes from the Centers for Disease Control and Prevention (CDC) of the United States. All data in this study has been removed identifiable information and can be available freely online. Thus, this study was regarded as exempt research by the institutional review. The data of this study was derived from the Surveillance, Epidemiology, and End Results (SEER) program 18 registries, which is an authoritative source of information on cancer incidence and survival in the United States. SEER currently collects and publishes cancer incidence and survival data from population-based cancer registries covering approximately 27.8% of the U.S. population, and it collects data concerning patient demographics, tumor morphology, stage at diagnosis, primary tumor site, the first course of treatment, and follow-up for vital status. Meanwhile, the data for the general population comes from the Centers for Disease Control and Prevention (CDC) of the United States. All data in this study has been removed identifiable information and can be available freely online. Thus, this study was regarded as exempt research by the institutional review. [SUBTITLE] Study Population and Study Variables [SUBSECTION] Patients with PeCa as the first primary malignancy and clear stage information according to the definition of Summary Stage 2000 (1998+) between 2004 and 2018 were included. The patients who had unclear follow-up time, living status at the end of follow-up and detailed causes of death were excluded. Patients were divided into 3 groups by their stages (localized, regional and distant metastasis). The following variables of all patients and the dead patients were analyzed, including age (15-54, 55-64, 65-74, 75-84, and >85 years), year of diagnosis (2004-2008, 2009-2013, and 2014-2018), race (white, black. American Indian/Alaska native and Asian or Pacific Islander), grade (well-differentiated, moderately differentiated, poorly differentiated, and undifferentiated), main pathological type, Radiotherapy (beam radiation, radioactive implants/brachytherapy and none/unknown) and Chemotherapy (yes and no/unknown). Patients with PeCa as the first primary malignancy and clear stage information according to the definition of Summary Stage 2000 (1998+) between 2004 and 2018 were included. The patients who had unclear follow-up time, living status at the end of follow-up and detailed causes of death were excluded. Patients were divided into 3 groups by their stages (localized, regional and distant metastasis). The following variables of all patients and the dead patients were analyzed, including age (15-54, 55-64, 65-74, 75-84, and >85 years), year of diagnosis (2004-2008, 2009-2013, and 2014-2018), race (white, black. American Indian/Alaska native and Asian or Pacific Islander), grade (well-differentiated, moderately differentiated, poorly differentiated, and undifferentiated), main pathological type, Radiotherapy (beam radiation, radioactive implants/brachytherapy and none/unknown) and Chemotherapy (yes and no/unknown). [SUBTITLE] Outcome Assessments [SUBSECTION] The causes of death after the diagnosis of PeCa included PeCa deaths, SMTs, and non-cancer deaths were regarded as the main outcomes. The standardized mortality ratio (SMR) with the 95% confidence interval (95% CI) of each cause was also calculated to evaluate the mortality risks compared with the general population. The last follow-up time was December 31, 2018, which was the latest data updated in the SEER database. The causes of death after the diagnosis of PeCa included PeCa deaths, SMTs, and non-cancer deaths were regarded as the main outcomes. The standardized mortality ratio (SMR) with the 95% confidence interval (95% CI) of each cause was also calculated to evaluate the mortality risks compared with the general population. The last follow-up time was December 31, 2018, which was the latest data updated in the SEER database. [SUBTITLE] Statistics Analyses [SUBSECTION] For the baseline characteristics of 3 groups of patients with different PeCa stages, we analyzed the number of all included patients and observed deaths stratified by the above variables. The SMRs for each variable were calculated by comparing the observed number of deaths to the expected number. The expected number of deaths was based on the total number of patient-year and the excess risk of the general population. The causes of death and related SMRs and 95% CI were also analyzed in localized, regional, and distant metastasis PeCa patients. Statistical significance for SMR is a two-sided test of P < .05. All these analyses were performed using SEER*Stat version 8.4.0.1. Pie charts of the proportion of different causes of death were drawn with Microsoft Excel 2016. For the baseline characteristics of 3 groups of patients with different PeCa stages, we analyzed the number of all included patients and observed deaths stratified by the above variables. The SMRs for each variable were calculated by comparing the observed number of deaths to the expected number. The expected number of deaths was based on the total number of patient-year and the excess risk of the general population. The causes of death and related SMRs and 95% CI were also analyzed in localized, regional, and distant metastasis PeCa patients. Statistical significance for SMR is a two-sided test of P < .05. All these analyses were performed using SEER*Stat version 8.4.0.1. Pie charts of the proportion of different causes of death were drawn with Microsoft Excel 2016.
Results
A total of 3597 PeCa patients were included, 1588 (44%) of them died during the follow-up. The majority of patients was in the localized stage (n = 2,155, 60%) and regional stage (n = 1310, 36%), whereas only 4% (n = 132) had distant disease. For patients with localized PeCa, 800 (37.1%) of them died with a SMR of 2.12 (1.97-2.27). 679 (51.8%) regional PeCa patients [SMR: 4.61 (4.27-4.97)] and 109 (82.6%) patients with distant metastatic PeCa [SMR: 27.68 (22.73-33.39)] died during the follow-up. Baseline characteristics of included patients and the dead cases were presented in Table 1. The trend of new diagnosed PeCa patients from 2000 to 2018 were revealed in Figure 1.Table 1.Baseline Characteristics of All Patients and Dead Cases During Follow-Up With Localized, Regional, and Distant Penile Cancer.CharacteristicLocalizedRegionalDistantPatients, nDeaths, nSMR (95% CI)Patients, nDeaths, nSMR (95% CI)Patients, nDeaths, nSMR (95% CI)Total21558002.12a (1.97-2.27)13106794.61a (4.27-4.97)13210927.68a (22.73-33.39)Age 15-54 years470804.41a (3.5-5.49)32912116.05a (13.32-19.18)3630106.43a (71.81-151.93) 55-64 years4991282.86a (2.39-3.4)3371537.48a (6.34-8.77)322748.63a (32.05-70.75) 65-74 years5652102.21a (1.92-2.53)3301834.67a (4.01-5.39)332635.53a (23.21-52.06) 75-84 years4102221.55a (1.35-1.76)2171432.61a (2.2-3.08)262110.51a (6.51-16.07) 85+ years2111602.10a (1.79-2.45)97793.10a (2.45-3.86)5513.45a (4.37-31.39)Year of diagnosis 2004-20087301432.65a (2.23-3.12)3532343.11a (2.73-3.54)302717.20a (11.34-25.03) 2009-20137442842.05a (1.82-2.3)4422585.32a (4.69-6.01)484232.68a (23.55-44.17) 2014-20186813732.01a (1.81-2.23)5151877.88a (6.79-9.1)544036.91a (26.37-50.27)Race White18066782.08a (1.92-2.24)11205774.35a (4-4.72)1058326.90a (21.43-33.35) Black222842.48a(1.98-3.07)116727.28a (5.7-9.17)161520.92a (11.71-34.51) American Indian/Alaska native2183.79a (1.64-7.47)1137.25a (1.49-21.18)33586.91a (121.03-1715.19) Asian or Pacific islander106301.95a (1.31-2.78)63276.22a (4.1-9.05)8861.02a (26.35-120.24)Grade Well differentiated6312181.78a (1.55-2.03)188863.01a (2.41-3.71)7620.27a (7.44-44.12) Moderately differentiated7063042.55a (2.27-2.85)5743175.07a (4.52-5.65)383219.92a (13.62-28.11) Poorly differentiated2571302.93a (2.44-3.47)3171925.02a (4.34-5.79)444135.67a (25.6-48.39) Undifferentiated92.7 (.08-2.52)1175.63a (2.27-11.61)329.89a (1.2-35.72)Pathological type 8070/3: Squamous cell carcinoma, NOS14255602.22a (2.04-2.41)8294474.49a (4.08-4.93)846830.34a (23.56-38.46) 8071/3: Squamous cell carcinoma, keratinizing, NOS3471262.26a (1.88-2.69)3321655.54a (4.73-6.46)191414.96a (8.18-25.1) 8051/3: Verrucous carcinoma, NOS169491.50a (1.11-1.98)39101.54 (.74-2.84)2298.29a (11.9-355.06) 8083/3: Basaloid squamous cell carcinoma41122.96a (1.53-5.18)34165.50a(3.14-8.93)6523.04a (7.48-53.77)Radiotherapy Beam radiation68301.90a (1.28-2.72)1981288.26a(6.89-9.83)372945.93a(30.76-65.96) Radioactive implants/brachytherapy214.79 (.12-26.7)3112.38 (.31-68.98)000 None/unknown20857692.13a (1.98-2.28)11095504.17a(3.83-4.54)958024.19a(19.18-30.11)Chemotherapy  Yes59222.69a (1.69-4.08)29816711.38a(9.72-13.24)745831.87a(24.2-41.21)  No/unknown20967782.10a (1.96-2.26)10125123.86a(3.53-4.21)585124.07a(17.92-31.65)SMR: standardized mortality ratio; 95% CI: 95% Confidence interval; NOS: Not otherwise specified.aStatistical significance with P < .05.Figure 1.The number of newly diagnosed of penile cancer from 2004 to 2018. Baseline Characteristics of All Patients and Dead Cases During Follow-Up With Localized, Regional, and Distant Penile Cancer. SMR: standardized mortality ratio; 95% CI: 95% Confidence interval; NOS: Not otherwise specified. aStatistical significance with P < .05. The number of newly diagnosed of penile cancer from 2004 to 2018. [SUBTITLE] Causes of Death for Patients With Localized Penile Cancer [SUBSECTION] Among the 800 cases of death, 199 (24.9%) died for PeCa, 144 (18.0%) died for other tumor causes, and the rest 457 (57.1%) deaths were due to non-tumor causes. 75.9% of all PeCa deaths were within 3 years after diagnosis. The mortality rate of the general population who died for PeCa was rather low, which led to the extremely high SMR from the ratio of observed and expected. The majority of SMTs deaths (n = 106, 73.6%) occurred with 5 years after the diagnosis of PeCa. The main SMTs were lung and bronchus cancer [n = 40, SMR: 1.71 (1.22-2.33)], all cancers of digestive system [n = 25, SMR: 1.12 (.72-1.65)]. The main non-cancer causes of death were diseases of heart [n = 172, SMR: 1.66 (1.42-1.93)], chronic obstructive pulmonary disease (COPD) and allied cond [n = 38, SMR: 1.63 (1.15-2.24)], cerebrovascular diseases [n = 33, 22% of all deaths, SMR: 1.71 (1.17-2.4)] and diabetes mellitus [n = 26, SMR: 2.30 (1.5-3.37)]. The mortality risks of lung and bronchus cancer, skin tumor, diseases of heart, COPD and allied cond, cerebrovascular diseases, diabetes mellitus, nephritis, nephrotic syndrome and nephrosis, other infectious and parasitic diseases, hypertension, chronic liver disease and cirrhosis and atherosclerosis were significantly increased when compared with the general population. All these results are shown in Table 2. The mortality rate of all causes of death, PeCa, SMTs and non-tumor causes of patients after diagnosis is shown in Figure 2A and the percentage composition of the main causes of death is presented in Figure 3A.Table 2.Main causes of death for patients with localized penile cancer.Causes of deathTotal<1 year<1-5 year5-10 years>10 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death800377.822.12a (1.97-2.27)16758.342.86a (2.44-3.33)422183.122.30a (2.09-2.54)167107.681.55a (1.32-1.8)4428.681.53a (1.11-2.06)All malignant cancers34386.53.97a (3.56-4.41)8013.455.95a (4.72-7.4)20442.354.82a (4.18-5.53)4824.481.96a (1.45-2.6)116.221.77 (.88-3.16) Male genital system21310.7219.86a (17.29-22.72)581.7133.97a (25.79-43.91)1295.224.82a (20.72-29.49)252.998.37a (5.42-12.36)1.831.2 (.03-6.7)  Penis199.082495.58a (2160.87-2867.44)53.014477.48a (3353.94-5856.66)125.043293.53a (2741.5-3924.09)21.02898.28a (556.05-1373.11)0.010 (0-561.23)  Prostate910.59.85 (.39-1.61)21.691.18 (.14-4.28)25.14.39 (.05-1.41)42.951.36 (.37-3.47)1.821.22 (.03-6.78) Urinary system86.291.27 (.55-2.51)3.963.14 (.65-9.16)43.051.31 (.36-3.36)11.81.55 (.01-3.08)0.470 (0-7.81)  Urinary bladder43.81.05 (.29-2.7)2.583.47 (.42-12.54)11.83.55 (.01-3.04)11.1.91 (.02-5.09)0.290 (0-12.7)  Kidney and renal Pelvis12.33.43 (.01-2.39)1.362.81 (.07-15.63)01.140 (0-3.24)0.670 (0-5.54)0.170 (0-21.87)  Other urinary organs3.0933.47a (6.9-97.82)0.010 (0-278.54)3.0469.99a (14.43-204.54)0.030 (0-140.81)0.010 (0-503.78) Digestive system2522.341.12 (.72-1.65)33.41.88 (.18-2.57)1810.891.65 (.98-2.61)36.4.47 (.1-1.37)11.63.61 (.02-3.42)  Liver and intrahepatic bile duct83.712.16 (.93-4.25)0.530 (0-6.92)71.783.93a (1.58-8.09)11.11.9 (.02-5.03)0.280 (0-13.01)  Pancreas75.471.28 (.51-2.63)1.821.22 (.03-6.82)52.651.89 (.61-4.4)11.59.63 (.02-3.5)0.420 (0-8.88)  Esophagus42.931.37 (.37-3.5)0.450 (0-8.21)31.442.08 (.43-6.09)0.840 (0-4.41)1.214.84 (.12-26.98)  Colon and rectum37.41.4 (.08-1.18)21.181.7 (.21-6.13)03.650 (0-1.01)12.06.48 (.01-2.7)0.520 (0-7.13)  Stomach21.831.1 (.13-3.96)0.290 (0-12.81)20.92.23 (.27-8.06)0.510 (0-7.2)0.130 (0-28.41)  Intrahepatic bile duct2.762.63 (.32-9.49)0.110 (0-34.14)1.362.77 (.07-15.41)1.234.36 (.11-24.32)0.060 (0-59.14) Respiratory system4324.311.77a (1.28-2.38)73.871.81 (.73-3.73)2512.072.07a (1.34-3.06)86.761.18 (.51-2.33)31.621.85 (.38-5.41)  Lung and bronchus4023.391.71a (1.22-2.33)73.721.88 (.76-3.87)2411.622.07a (1.32-3.07)66.5.92 (.34-2.01)3z1.93 (.4-5.64) Skin excluding basal and squamous112.284.82a (2.41-8.63)1.352.88 (.07-16.03)61.125.38a (1.97-11.7)3.664.57 (.94-13.37)1.166.17 (.16-34.36) Brain and other nervous system21.761.14 (.14-4.11)0.260 (0-13.99)2.862.34 (.28-8.44)0.510 (0-7.23)0.130 (0-28.57) Lymphoma33.5.86 (.18-2.5)0.540 (0-6.79)11.71.58 (.01-3.25)2.992.01 (.24-7.27)0.250 (0-14.66)  Myeloma21.931.04 (.13-3.75)0.290 (0-12.63)1.931.07 (.03-5.97)1.551.81 (.05-10.08)0.150 (0-24.97) Leukemia33.89.77 (.16-2.25)00.60 (0-6.15)21.91.05 (.13-3.8)11.11.9 (.02-5.02)0.280 (0-13.08) Miscellaneous malignant cancer286.794.12a (2.74-5.96)71.056.64a (2.67-13.67)133.323.91a (2.08-6.69)41.922.08 (.57-5.32)4.498.13#(2.21-20.81)Non-cancer causes Diseases of heart172103.441.66a (1.42-1.93)2916.311.78a (1.19-2.55)9150.311.81a (1.46-2.22)4029.081.38 (.98-1.87)127.741.55 (.8-2.71) COPD and allied cond3823.291.63a (1.15-2.24)83.592.23 (.96-4.4)2011.321.77a (1.08-2.73)96.651.35 (.62-2.57)11.74.57 (.01-3.2) Cerebrovascular diseases3319.341.71#(1.17-2.4)73.042.31 (.93-4.75)109.331.07 (.51-1.97)95.461.65 (.75-3.13)71.514.63#(1.86-9.54) Diabetes mellitus2611.322.30a (1.5-3.37)61.723.49#(1.28-7.6)75.471.28 (.51-2.64)113.253.38a (1.69-6.05)2.882.28 (.28-8.24) Nephritis, nephrotic syndrome and nephrosis168.251.94a (1.11-3.15)31.282.34 (.48-6.83)1142.75a (1.37-4.92)12.33.43 (.01-2.39)1.631.58 (.04-8.79) Pneumonia and influenza149.331.5 (.82-2.52)51.53.34a (1.08-7.79)64.541.32 (.49-2.88)32.61.16 (.24-3.38)00.70 (0-5.3) Other infectious and parasitic diseases including HIV133.134.16a (2.21-7.11)5.4910.17a (3.3-23.74)31.551.93 (.4-5.64)5.885.71a (1.85-13.33)0.210 (0-17.77) Accidents and adverse effects1112.11.91 (.45-1.63)21.821.1 (.13-3.96)45.84.69 (.19-1.75)33.52.85 (.18-2.49)2.932.16 (.26-7.79) Hypertension without heart disease104.222.37#(1.14-4.36)2.623.22 (.39-11.62)11.99.5 (.01-2.8)51.244.04a (1.31-9.42)2.365.52 (.67-19.96) Chronic liver disease and cirrhosis93.82.37a (1.08-4.49)2.573.53 (.43-12.76)41.852.16 (.59-5.53)21.111.8 (.22-6.49)1.273.65 (.09-20.36)AbbreviationsSMR: standardized mortality ratio; CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease.aStatistical significance with P < .05.Figure 2.Mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients diagnosed with localized, regional and distant metastasis penile cancer. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.Figure 3.The percentages of main causes of death in penile cancer patients. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer. Main causes of death for patients with localized penile cancer. AbbreviationsSMR: standardized mortality ratio; CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease. aStatistical significance with P < .05. Mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients diagnosed with localized, regional and distant metastasis penile cancer. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer. The percentages of main causes of death in penile cancer patients. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer. Among the 800 cases of death, 199 (24.9%) died for PeCa, 144 (18.0%) died for other tumor causes, and the rest 457 (57.1%) deaths were due to non-tumor causes. 75.9% of all PeCa deaths were within 3 years after diagnosis. The mortality rate of the general population who died for PeCa was rather low, which led to the extremely high SMR from the ratio of observed and expected. The majority of SMTs deaths (n = 106, 73.6%) occurred with 5 years after the diagnosis of PeCa. The main SMTs were lung and bronchus cancer [n = 40, SMR: 1.71 (1.22-2.33)], all cancers of digestive system [n = 25, SMR: 1.12 (.72-1.65)]. The main non-cancer causes of death were diseases of heart [n = 172, SMR: 1.66 (1.42-1.93)], chronic obstructive pulmonary disease (COPD) and allied cond [n = 38, SMR: 1.63 (1.15-2.24)], cerebrovascular diseases [n = 33, 22% of all deaths, SMR: 1.71 (1.17-2.4)] and diabetes mellitus [n = 26, SMR: 2.30 (1.5-3.37)]. The mortality risks of lung and bronchus cancer, skin tumor, diseases of heart, COPD and allied cond, cerebrovascular diseases, diabetes mellitus, nephritis, nephrotic syndrome and nephrosis, other infectious and parasitic diseases, hypertension, chronic liver disease and cirrhosis and atherosclerosis were significantly increased when compared with the general population. All these results are shown in Table 2. The mortality rate of all causes of death, PeCa, SMTs and non-tumor causes of patients after diagnosis is shown in Figure 2A and the percentage composition of the main causes of death is presented in Figure 3A.Table 2.Main causes of death for patients with localized penile cancer.Causes of deathTotal<1 year<1-5 year5-10 years>10 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death800377.822.12a (1.97-2.27)16758.342.86a (2.44-3.33)422183.122.30a (2.09-2.54)167107.681.55a (1.32-1.8)4428.681.53a (1.11-2.06)All malignant cancers34386.53.97a (3.56-4.41)8013.455.95a (4.72-7.4)20442.354.82a (4.18-5.53)4824.481.96a (1.45-2.6)116.221.77 (.88-3.16) Male genital system21310.7219.86a (17.29-22.72)581.7133.97a (25.79-43.91)1295.224.82a (20.72-29.49)252.998.37a (5.42-12.36)1.831.2 (.03-6.7)  Penis199.082495.58a (2160.87-2867.44)53.014477.48a (3353.94-5856.66)125.043293.53a (2741.5-3924.09)21.02898.28a (556.05-1373.11)0.010 (0-561.23)  Prostate910.59.85 (.39-1.61)21.691.18 (.14-4.28)25.14.39 (.05-1.41)42.951.36 (.37-3.47)1.821.22 (.03-6.78) Urinary system86.291.27 (.55-2.51)3.963.14 (.65-9.16)43.051.31 (.36-3.36)11.81.55 (.01-3.08)0.470 (0-7.81)  Urinary bladder43.81.05 (.29-2.7)2.583.47 (.42-12.54)11.83.55 (.01-3.04)11.1.91 (.02-5.09)0.290 (0-12.7)  Kidney and renal Pelvis12.33.43 (.01-2.39)1.362.81 (.07-15.63)01.140 (0-3.24)0.670 (0-5.54)0.170 (0-21.87)  Other urinary organs3.0933.47a (6.9-97.82)0.010 (0-278.54)3.0469.99a (14.43-204.54)0.030 (0-140.81)0.010 (0-503.78) Digestive system2522.341.12 (.72-1.65)33.41.88 (.18-2.57)1810.891.65 (.98-2.61)36.4.47 (.1-1.37)11.63.61 (.02-3.42)  Liver and intrahepatic bile duct83.712.16 (.93-4.25)0.530 (0-6.92)71.783.93a (1.58-8.09)11.11.9 (.02-5.03)0.280 (0-13.01)  Pancreas75.471.28 (.51-2.63)1.821.22 (.03-6.82)52.651.89 (.61-4.4)11.59.63 (.02-3.5)0.420 (0-8.88)  Esophagus42.931.37 (.37-3.5)0.450 (0-8.21)31.442.08 (.43-6.09)0.840 (0-4.41)1.214.84 (.12-26.98)  Colon and rectum37.41.4 (.08-1.18)21.181.7 (.21-6.13)03.650 (0-1.01)12.06.48 (.01-2.7)0.520 (0-7.13)  Stomach21.831.1 (.13-3.96)0.290 (0-12.81)20.92.23 (.27-8.06)0.510 (0-7.2)0.130 (0-28.41)  Intrahepatic bile duct2.762.63 (.32-9.49)0.110 (0-34.14)1.362.77 (.07-15.41)1.234.36 (.11-24.32)0.060 (0-59.14) Respiratory system4324.311.77a (1.28-2.38)73.871.81 (.73-3.73)2512.072.07a (1.34-3.06)86.761.18 (.51-2.33)31.621.85 (.38-5.41)  Lung and bronchus4023.391.71a (1.22-2.33)73.721.88 (.76-3.87)2411.622.07a (1.32-3.07)66.5.92 (.34-2.01)3z1.93 (.4-5.64) Skin excluding basal and squamous112.284.82a (2.41-8.63)1.352.88 (.07-16.03)61.125.38a (1.97-11.7)3.664.57 (.94-13.37)1.166.17 (.16-34.36) Brain and other nervous system21.761.14 (.14-4.11)0.260 (0-13.99)2.862.34 (.28-8.44)0.510 (0-7.23)0.130 (0-28.57) Lymphoma33.5.86 (.18-2.5)0.540 (0-6.79)11.71.58 (.01-3.25)2.992.01 (.24-7.27)0.250 (0-14.66)  Myeloma21.931.04 (.13-3.75)0.290 (0-12.63)1.931.07 (.03-5.97)1.551.81 (.05-10.08)0.150 (0-24.97) Leukemia33.89.77 (.16-2.25)00.60 (0-6.15)21.91.05 (.13-3.8)11.11.9 (.02-5.02)0.280 (0-13.08) Miscellaneous malignant cancer286.794.12a (2.74-5.96)71.056.64a (2.67-13.67)133.323.91a (2.08-6.69)41.922.08 (.57-5.32)4.498.13#(2.21-20.81)Non-cancer causes Diseases of heart172103.441.66a (1.42-1.93)2916.311.78a (1.19-2.55)9150.311.81a (1.46-2.22)4029.081.38 (.98-1.87)127.741.55 (.8-2.71) COPD and allied cond3823.291.63a (1.15-2.24)83.592.23 (.96-4.4)2011.321.77a (1.08-2.73)96.651.35 (.62-2.57)11.74.57 (.01-3.2) Cerebrovascular diseases3319.341.71#(1.17-2.4)73.042.31 (.93-4.75)109.331.07 (.51-1.97)95.461.65 (.75-3.13)71.514.63#(1.86-9.54) Diabetes mellitus2611.322.30a (1.5-3.37)61.723.49#(1.28-7.6)75.471.28 (.51-2.64)113.253.38a (1.69-6.05)2.882.28 (.28-8.24) Nephritis, nephrotic syndrome and nephrosis168.251.94a (1.11-3.15)31.282.34 (.48-6.83)1142.75a (1.37-4.92)12.33.43 (.01-2.39)1.631.58 (.04-8.79) Pneumonia and influenza149.331.5 (.82-2.52)51.53.34a (1.08-7.79)64.541.32 (.49-2.88)32.61.16 (.24-3.38)00.70 (0-5.3) Other infectious and parasitic diseases including HIV133.134.16a (2.21-7.11)5.4910.17a (3.3-23.74)31.551.93 (.4-5.64)5.885.71a (1.85-13.33)0.210 (0-17.77) Accidents and adverse effects1112.11.91 (.45-1.63)21.821.1 (.13-3.96)45.84.69 (.19-1.75)33.52.85 (.18-2.49)2.932.16 (.26-7.79) Hypertension without heart disease104.222.37#(1.14-4.36)2.623.22 (.39-11.62)11.99.5 (.01-2.8)51.244.04a (1.31-9.42)2.365.52 (.67-19.96) Chronic liver disease and cirrhosis93.82.37a (1.08-4.49)2.573.53 (.43-12.76)41.852.16 (.59-5.53)21.111.8 (.22-6.49)1.273.65 (.09-20.36)AbbreviationsSMR: standardized mortality ratio; CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease.aStatistical significance with P < .05.Figure 2.Mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients diagnosed with localized, regional and distant metastasis penile cancer. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.Figure 3.The percentages of main causes of death in penile cancer patients. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer. Main causes of death for patients with localized penile cancer. AbbreviationsSMR: standardized mortality ratio; CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease. aStatistical significance with P < .05. Mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients diagnosed with localized, regional and distant metastasis penile cancer. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer. The percentages of main causes of death in penile cancer patients. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer. [SUBTITLE] Causes of Death for Patients With Regional Penile Cancer [SUBSECTION] For patients with regional PeCa, 348 (51.3%) of all 679 deaths were due to PeCa itself. There were 118 SMTs deaths and 213 non-cancer deaths, accounting for 17.4% and 31.4% of all deaths. For the PeCa deaths, 89.1% of them occurred within 3 years after diagnosis. With the low PeCa mortality rate of the general population, the SMR of PeCa death reached a terribly high level. The main SMTs were lung and bronchus cancer [n = 23, SMR: 2.41 (1.53-3.62)]. The mortality risks of lung and bronchus cancer, and skin tumor [SMR: 6.41 (2.35-13.95)] were significantly increased. The most common non-cancer cause of death was diseases of heart [n = 71, SMR: 1.77 (1.38-2.23)], COPD and allied cond [n = 17, SMR: 1.85 (1.08-2.95)], diabetes mellitus [n = 16, SMR: 3.62 (2.07-5.88)] and cerebrovascular diseases [n = 13, SMR: 1.77 (.94-3.03)]. The mortality risks of diseases of heart, COPD and allied cond, diabetes mellitus, nephritis, nephrotic syndrome and nephrosis [n = 9, SMR: 2.88 (1.32-5.47)] and septicemia [n = 8, SMR: 3.83 (1.65-7.54)] were significantly increased when compared with the general population. All these results are shown in Table 3. The mortality rate of patients after diagnosis and is shown in Figure 2B and the percentages of the main causes of death is revealed in Figure 3B.Table 3.Main causes of death for patients with regional penile cancer.Causes of deathTotal<1 year1-5 years>5 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death679147.44.61a (4.27-4.97)27527.559.98a (8.84-11.23)31067.934.56a (4.07-5.1)9451.921.81a (1.46-2.22)All malignant cancers46634.6513.45a (12.26-14.73)2136.5632.48a (28.27-37.15)22516.4213.71a (11.97-15.62)2811.672.40a (1.59-3.47) Male genital system3624.0788.89a (79.97-98.54)179.79225.26a (193.47-260.79)1681.8889.54a (76.51-104.15)151.410.70a (5.99-17.65)  Penis348.0311 020.00a (9892.46-12,240.84)173.0129 605.21a (25 357.89-34 360.42)162.0111 182.19a (9526.53-13042.87)13.011155.76a (615.39-1976.38)  Prostate84.021.99 (.86-3.92)2.782.55 (.31-9.21)41.852.16 (.59-5.53)21.381.45 (.18-5.22)  Testis2.01149.35a (18.09-539.5)10370.55a (9.38-2064.6)1.01152.52a (3.86-849.81)000 (0-891.78) Urinary system62.512.39 (.88-5.21)1.462.18 (.06-12.16)41.153.47 (.94-8.87)1.891.12 (.03-6.23)  Kidney and renal Pelvis3.953.17 (.65-9.27)0.180 (0-20.85)2.454.46 (.54-16.13)1.323.12 (.08-17.36)  Urinary bladder21.51.34 (.16-4.83)1.273.71 (.09-20.65)1.681.48 (.04-8.23)0.550 (0-6.72) Digestive system78.97.78 (.31-1.61)11.69.59 (.01-3.3)44.27.94 (.26-2.4)23.01.66 (.08-2.4)  Colon and rectum32.951.02 (.21-2.97)0.570 (0-6.44)21.411.41 (.17-5.11)1.971.03 (.03-5.77)  Esophagus21.211.65 (.2-5.95)0.230 (0-16.24)1.581.72 (.04-9.57)1.412.47 (.06-13.75) Respiratory system239.92.32a (1.47-3.49)41.912.1 (.57-5.37)134.82.71a (1.44-4.63)63.191.88 (.69-4.09)  Lung and bronchus239.532.41a (1.53-3.62)41.842.18 (.59-5.58)134.622.81a (1.5-4.81)63.071.95 (.72-4.25) Skin excluding basal and squamous6.946.41a (2.35-13.95)2.1711.79a (1.43-42.6)4.439.23a (2.51-23.62)0.330 (0-11.07) Lymphoma21.411.42 (.17-5.14)0.260 (0-14.17)2.653.05 (.37-11.03)0.490 (0-7.53) Leukemia21.561.28 (.16-4.63)0.290 (0-12.84)2.722.76 (.33-9.97)0.550 (0-6.74) Miscellaneous malignant cancer512.7218.75a (13.96-24.66)25.5148.95a (31.68-72.26)241.2818.76a (12.02-27.92)2.932.15 (.26-7.77)Non-cancer causes Diseases of heart7140.111.77a (1.38-2.23)207.572.64#(1.61-4.08)3218.451.73a (1.19-2.45)1914.091.35 (.81-2.11) COPD and allied cond179.211.85a (1.08-2.95)41.692.37 (.65-6.08)84.241.89 (.82-3.72)53.291.52 (.49-3.55) Diabetes mellitus164.423.62a (2.07-5.88)5.836.00a (1.95-14.01)62.072.89a (1.06-6.3)51.513.31a (1.08-7.73) Cerebrovascular diseases137.341.77 (.94-3.03)31.392.16 (.44-6.3)63.341.79 (.66-3.91)42.611.53 (.42-3.92) Nephritis, nephrotic syndrome and nephrosis93.132.88a (1.32-5.47)3.595.07a (1.05-14.81)51.443.48a (1.13-8.12)11.1.91 (.02-5.08) Septicemia82.093.83a (1.65-7.54)50.412.66a (4.11-29.53)1.961.04 (.03-5.77)2.732.74 (.33-9.89) Alzheimers84.791.67 (.72-3.29)3.843.56 (.74-10.42)22.02.99 (.12-3.57)31.931.56 (.32-4.55) Chronic liver disease and cirrhosis41.622.47 (.67-6.32)3.319.67a (1.99-28.25)0.790 (0-4.64)1.521.94 (.05-10.8) Accidents and adverse effects64.841.24 (.45-2.7)0.910 (0-4.04)32.261.33 (.27-3.88)31.671.8 (.37-5.25) Suicide and self-inflicted injury41.382.9 (.79-7.42)1.273.71 (.09-20.68)1.671.49 (.04-8.28)2.444.57 (.55-16.51)CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease; SMR: standardized mortality ratio.aStatistical significance with P < .05. Main causes of death for patients with regional penile cancer. CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease; SMR: standardized mortality ratio. aStatistical significance with P < .05. For patients with regional PeCa, 348 (51.3%) of all 679 deaths were due to PeCa itself. There were 118 SMTs deaths and 213 non-cancer deaths, accounting for 17.4% and 31.4% of all deaths. For the PeCa deaths, 89.1% of them occurred within 3 years after diagnosis. With the low PeCa mortality rate of the general population, the SMR of PeCa death reached a terribly high level. The main SMTs were lung and bronchus cancer [n = 23, SMR: 2.41 (1.53-3.62)]. The mortality risks of lung and bronchus cancer, and skin tumor [SMR: 6.41 (2.35-13.95)] were significantly increased. The most common non-cancer cause of death was diseases of heart [n = 71, SMR: 1.77 (1.38-2.23)], COPD and allied cond [n = 17, SMR: 1.85 (1.08-2.95)], diabetes mellitus [n = 16, SMR: 3.62 (2.07-5.88)] and cerebrovascular diseases [n = 13, SMR: 1.77 (.94-3.03)]. The mortality risks of diseases of heart, COPD and allied cond, diabetes mellitus, nephritis, nephrotic syndrome and nephrosis [n = 9, SMR: 2.88 (1.32-5.47)] and septicemia [n = 8, SMR: 3.83 (1.65-7.54)] were significantly increased when compared with the general population. All these results are shown in Table 3. The mortality rate of patients after diagnosis and is shown in Figure 2B and the percentages of the main causes of death is revealed in Figure 3B.Table 3.Main causes of death for patients with regional penile cancer.Causes of deathTotal<1 year1-5 years>5 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death679147.44.61a (4.27-4.97)27527.559.98a (8.84-11.23)31067.934.56a (4.07-5.1)9451.921.81a (1.46-2.22)All malignant cancers46634.6513.45a (12.26-14.73)2136.5632.48a (28.27-37.15)22516.4213.71a (11.97-15.62)2811.672.40a (1.59-3.47) Male genital system3624.0788.89a (79.97-98.54)179.79225.26a (193.47-260.79)1681.8889.54a (76.51-104.15)151.410.70a (5.99-17.65)  Penis348.0311 020.00a (9892.46-12,240.84)173.0129 605.21a (25 357.89-34 360.42)162.0111 182.19a (9526.53-13042.87)13.011155.76a (615.39-1976.38)  Prostate84.021.99 (.86-3.92)2.782.55 (.31-9.21)41.852.16 (.59-5.53)21.381.45 (.18-5.22)  Testis2.01149.35a (18.09-539.5)10370.55a (9.38-2064.6)1.01152.52a (3.86-849.81)000 (0-891.78) Urinary system62.512.39 (.88-5.21)1.462.18 (.06-12.16)41.153.47 (.94-8.87)1.891.12 (.03-6.23)  Kidney and renal Pelvis3.953.17 (.65-9.27)0.180 (0-20.85)2.454.46 (.54-16.13)1.323.12 (.08-17.36)  Urinary bladder21.51.34 (.16-4.83)1.273.71 (.09-20.65)1.681.48 (.04-8.23)0.550 (0-6.72) Digestive system78.97.78 (.31-1.61)11.69.59 (.01-3.3)44.27.94 (.26-2.4)23.01.66 (.08-2.4)  Colon and rectum32.951.02 (.21-2.97)0.570 (0-6.44)21.411.41 (.17-5.11)1.971.03 (.03-5.77)  Esophagus21.211.65 (.2-5.95)0.230 (0-16.24)1.581.72 (.04-9.57)1.412.47 (.06-13.75) Respiratory system239.92.32a (1.47-3.49)41.912.1 (.57-5.37)134.82.71a (1.44-4.63)63.191.88 (.69-4.09)  Lung and bronchus239.532.41a (1.53-3.62)41.842.18 (.59-5.58)134.622.81a (1.5-4.81)63.071.95 (.72-4.25) Skin excluding basal and squamous6.946.41a (2.35-13.95)2.1711.79a (1.43-42.6)4.439.23a (2.51-23.62)0.330 (0-11.07) Lymphoma21.411.42 (.17-5.14)0.260 (0-14.17)2.653.05 (.37-11.03)0.490 (0-7.53) Leukemia21.561.28 (.16-4.63)0.290 (0-12.84)2.722.76 (.33-9.97)0.550 (0-6.74) Miscellaneous malignant cancer512.7218.75a (13.96-24.66)25.5148.95a (31.68-72.26)241.2818.76a (12.02-27.92)2.932.15 (.26-7.77)Non-cancer causes Diseases of heart7140.111.77a (1.38-2.23)207.572.64#(1.61-4.08)3218.451.73a (1.19-2.45)1914.091.35 (.81-2.11) COPD and allied cond179.211.85a (1.08-2.95)41.692.37 (.65-6.08)84.241.89 (.82-3.72)53.291.52 (.49-3.55) Diabetes mellitus164.423.62a (2.07-5.88)5.836.00a (1.95-14.01)62.072.89a (1.06-6.3)51.513.31a (1.08-7.73) Cerebrovascular diseases137.341.77 (.94-3.03)31.392.16 (.44-6.3)63.341.79 (.66-3.91)42.611.53 (.42-3.92) Nephritis, nephrotic syndrome and nephrosis93.132.88a (1.32-5.47)3.595.07a (1.05-14.81)51.443.48a (1.13-8.12)11.1.91 (.02-5.08) Septicemia82.093.83a (1.65-7.54)50.412.66a (4.11-29.53)1.961.04 (.03-5.77)2.732.74 (.33-9.89) Alzheimers84.791.67 (.72-3.29)3.843.56 (.74-10.42)22.02.99 (.12-3.57)31.931.56 (.32-4.55) Chronic liver disease and cirrhosis41.622.47 (.67-6.32)3.319.67a (1.99-28.25)0.790 (0-4.64)1.521.94 (.05-10.8) Accidents and adverse effects64.841.24 (.45-2.7)0.910 (0-4.04)32.261.33 (.27-3.88)31.671.8 (.37-5.25) Suicide and self-inflicted injury41.382.9 (.79-7.42)1.273.71 (.09-20.68)1.671.49 (.04-8.28)2.444.57 (.55-16.51)CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease; SMR: standardized mortality ratio.aStatistical significance with P < .05. Main causes of death for patients with regional penile cancer. CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease; SMR: standardized mortality ratio. aStatistical significance with P < .05. [SUBTITLE] Causes of Death for Patients With Distant Metastatic Penile Cancer [SUBSECTION] For the included 132 patients with distant metastasis PeCa, 109 of them died during the follow-up with a significantly increased SMR of 27.68 (22.73-33.39). 100 deaths (91.7%) were due to malignant cancers including 76 cases (69.7%) dead of PeCa. 67.1% (n = 51) of all PeCa deaths occurred within 1 year and 98.7% (n = 75) dead within 3 years after diagnosis. There were 24 patients (22%) who died for SMTs, mainly including cancers from 4 lung and bronchus, 2 prostate, and 2 skin, 2 lymphoma and 3 other urinary organs. 19 (79.2%) of SMTs deaths occurred within 1 year of the diagnosis of PeCa. As for the non-cancer deaths, only 9 cases of deaths were observed, including 5 deaths from diseases of heart, 1 septicemia death, 1 other infection and parasitic diseases deaths, and 2 other causes of death. These results are presented in Table 4 and Figure 3C. The mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients after diagnosis is shown in Figure 2C.Table 4.Main Causes of Death for Patients With Distant Penile Cancer.Causes of deathTotal<1 year1-3 years>3 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death1093.9427.68a (22.73-33.39)741.8540.10a (31.49-50.34)281.0925.61a (17.02-37.01)717.00a (2.82-14.43)All malignant cancers100.98102.30a (83.23-124.42)70.45154.76a (120.65-195.54)27.2992.21a (60.76-134.16)3.2312.91a (2.66-37.72) Respiratory system4.2814.38a (3.92-36.81)3.1322.93a (4.73-67.01)0.090 (0-42.56)1.0616.48 (.42-91.8)  Lung and bronchus4.2714.98a (4.08-38.34)3.1323.85a (4.92-69.69)0.080 (0-44.28)1.0617.24 (.44-96.06) Soft tissue including heart2.01331.85a (40.19-1198.77)10351.01a (8.89-1955.69)10554.76a (14.05-3090.9)000 (0-2682.36) Skin excluding basal and squamous2.0285.91a (10.4-310.32)1.0187.34a (2.21-486.61)1.01146.53a (3.71-816.42)0.010 (0-736.74)  Male genital system80.12650.98a (516.18-810.2)55.051065.43a (802.63-1386.8)24.03713.33a (457.04-1061.38)1.0426.58 (.67-148.08)  Prostate2.1216.48a (2-59.52)2.0539.26a (4.75-141.82)0.030 (0-111.09)0.040 (0-99.09)  Penis76084 751.04a (66 774.18-106078.56)510126 908.70a (94 491.86-166861.5)24092 697.90a (59 393.29-137,927.13)104237.74a (107.29-23,611.17) Urinary system3.0646.59a (9.61-136.15)2.0364.45a (7.81-232.82)0.020 (0-199.1)1.0167.40a (1.71-375.54) Miscellaneous malignant cancer7.0793.72a (37.68-193.1)7.04198.90a (79.97-409.8)0.020 (0-166.27)0.020 (0-213.09)Non-cancer causes Septicemia1.0616.57 (.42-92.3)1.0337.63 (.95-209.66)0.020 (0-218.26)0.020 (0-218.46) Other infectious and parasitic diseases including HIV1.0426.85 (.68-149.58)1.0257.29a (1.45-319.22)0.010 (0-340.04)0.010 (0-412.29) Diseases of heart51.054.75a (1.54-11.09)1.492.02 (.05-11.26)1.283.52 (.09-19.64)3.2710.95a (2.26-32) Other cause of death2.573.53 (.43-12.75)1.273.69 (.09-20.57)0.150 (0-25.01)1.156.75 (.17-37.6)SMR: standardized mortality ratio; CI: confidence interval.aStatistical significance with P < .05. Main Causes of Death for Patients With Distant Penile Cancer. SMR: standardized mortality ratio; CI: confidence interval. aStatistical significance with P < .05. For the included 132 patients with distant metastasis PeCa, 109 of them died during the follow-up with a significantly increased SMR of 27.68 (22.73-33.39). 100 deaths (91.7%) were due to malignant cancers including 76 cases (69.7%) dead of PeCa. 67.1% (n = 51) of all PeCa deaths occurred within 1 year and 98.7% (n = 75) dead within 3 years after diagnosis. There were 24 patients (22%) who died for SMTs, mainly including cancers from 4 lung and bronchus, 2 prostate, and 2 skin, 2 lymphoma and 3 other urinary organs. 19 (79.2%) of SMTs deaths occurred within 1 year of the diagnosis of PeCa. As for the non-cancer deaths, only 9 cases of deaths were observed, including 5 deaths from diseases of heart, 1 septicemia death, 1 other infection and parasitic diseases deaths, and 2 other causes of death. These results are presented in Table 4 and Figure 3C. The mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients after diagnosis is shown in Figure 2C.Table 4.Main Causes of Death for Patients With Distant Penile Cancer.Causes of deathTotal<1 year1-3 years>3 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death1093.9427.68a (22.73-33.39)741.8540.10a (31.49-50.34)281.0925.61a (17.02-37.01)717.00a (2.82-14.43)All malignant cancers100.98102.30a (83.23-124.42)70.45154.76a (120.65-195.54)27.2992.21a (60.76-134.16)3.2312.91a (2.66-37.72) Respiratory system4.2814.38a (3.92-36.81)3.1322.93a (4.73-67.01)0.090 (0-42.56)1.0616.48 (.42-91.8)  Lung and bronchus4.2714.98a (4.08-38.34)3.1323.85a (4.92-69.69)0.080 (0-44.28)1.0617.24 (.44-96.06) Soft tissue including heart2.01331.85a (40.19-1198.77)10351.01a (8.89-1955.69)10554.76a (14.05-3090.9)000 (0-2682.36) Skin excluding basal and squamous2.0285.91a (10.4-310.32)1.0187.34a (2.21-486.61)1.01146.53a (3.71-816.42)0.010 (0-736.74)  Male genital system80.12650.98a (516.18-810.2)55.051065.43a (802.63-1386.8)24.03713.33a (457.04-1061.38)1.0426.58 (.67-148.08)  Prostate2.1216.48a (2-59.52)2.0539.26a (4.75-141.82)0.030 (0-111.09)0.040 (0-99.09)  Penis76084 751.04a (66 774.18-106078.56)510126 908.70a (94 491.86-166861.5)24092 697.90a (59 393.29-137,927.13)104237.74a (107.29-23,611.17) Urinary system3.0646.59a (9.61-136.15)2.0364.45a (7.81-232.82)0.020 (0-199.1)1.0167.40a (1.71-375.54) Miscellaneous malignant cancer7.0793.72a (37.68-193.1)7.04198.90a (79.97-409.8)0.020 (0-166.27)0.020 (0-213.09)Non-cancer causes Septicemia1.0616.57 (.42-92.3)1.0337.63 (.95-209.66)0.020 (0-218.26)0.020 (0-218.46) Other infectious and parasitic diseases including HIV1.0426.85 (.68-149.58)1.0257.29a (1.45-319.22)0.010 (0-340.04)0.010 (0-412.29) Diseases of heart51.054.75a (1.54-11.09)1.492.02 (.05-11.26)1.283.52 (.09-19.64)3.2710.95a (2.26-32) Other cause of death2.573.53 (.43-12.75)1.273.69 (.09-20.57)0.150 (0-25.01)1.156.75 (.17-37.6)SMR: standardized mortality ratio; CI: confidence interval.aStatistical significance with P < .05. Main Causes of Death for Patients With Distant Penile Cancer. SMR: standardized mortality ratio; CI: confidence interval. aStatistical significance with P < .05.
Conclusion
Our study provided a detailed analysis of the causes of death for patients with locally, regionally, and distant metastatic PeCa after diagnosis. This information could be useful for disease prevention and health care during patients’ survivorship.
[ "Data Sources", "Study Population and Study Variables", "Outcome Assessments", "Statistics Analyses", "Causes of Death for Patients With Localized Penile Cancer", "Causes of Death for Patients With Regional Penile Cancer", "Causes of Death for Patients With Distant Metastatic Penile Cancer" ]
[ "The data of this study was derived from the Surveillance, Epidemiology, and End Results (SEER) program 18 registries, which is an authoritative source of information on cancer incidence and survival in the United States. SEER currently collects and publishes cancer incidence and survival data from population-based cancer registries covering approximately 27.8% of the U.S. population, and it collects data concerning patient demographics, tumor morphology, stage at diagnosis, primary tumor site, the first course of treatment, and follow-up for vital status. Meanwhile, the data for the general population comes from the Centers for Disease Control and Prevention (CDC) of the United States. All data in this study has been removed identifiable information and can be available freely online. Thus, this study was regarded as exempt research by the institutional review.", "Patients with PeCa as the first primary malignancy and clear stage information according to the definition of Summary Stage 2000 (1998+) between 2004 and 2018 were included. The patients who had unclear follow-up time, living status at the end of follow-up and detailed causes of death were excluded.\nPatients were divided into 3 groups by their stages (localized, regional and distant metastasis). The following variables of all patients and the dead patients were analyzed, including age (15-54, 55-64, 65-74, 75-84, and >85 years), year of diagnosis (2004-2008, 2009-2013, and 2014-2018), race (white, black. American Indian/Alaska native and Asian or Pacific Islander), grade (well-differentiated, moderately differentiated, poorly differentiated, and undifferentiated), main pathological type, Radiotherapy (beam radiation, radioactive implants/brachytherapy and none/unknown) and Chemotherapy (yes and no/unknown).", "The causes of death after the diagnosis of PeCa included PeCa deaths, SMTs, and non-cancer deaths were regarded as the main outcomes. The standardized mortality ratio (SMR) with the 95% confidence interval (95% CI) of each cause was also calculated to evaluate the mortality risks compared with the general population. The last follow-up time was December 31, 2018, which was the latest data updated in the SEER database.", "For the baseline characteristics of 3 groups of patients with different PeCa stages, we analyzed the number of all included patients and observed deaths stratified by the above variables. The SMRs for each variable were calculated by comparing the observed number of deaths to the expected number. The expected number of deaths was based on the total number of patient-year and the excess risk of the general population. The causes of death and related SMRs and 95% CI were also analyzed in localized, regional, and distant metastasis PeCa patients. Statistical significance for SMR is a two-sided test of P < .05. All these analyses were performed using SEER*Stat version 8.4.0.1. Pie charts of the proportion of different causes of death were drawn with Microsoft Excel 2016.", "Among the 800 cases of death, 199 (24.9%) died for PeCa, 144 (18.0%) died for other tumor causes, and the rest 457 (57.1%) deaths were due to non-tumor causes. 75.9% of all PeCa deaths were within 3 years after diagnosis. The mortality rate of the general population who died for PeCa was rather low, which led to the extremely high SMR from the ratio of observed and expected. The majority of SMTs deaths (n = 106, 73.6%) occurred with 5 years after the diagnosis of PeCa. The main SMTs were lung and bronchus cancer [n = 40, SMR: 1.71 (1.22-2.33)], all cancers of digestive system [n = 25, SMR: 1.12 (.72-1.65)]. The main non-cancer causes of death were diseases of heart [n = 172, SMR: 1.66 (1.42-1.93)], chronic obstructive pulmonary disease (COPD) and allied cond [n = 38, SMR: 1.63 (1.15-2.24)], cerebrovascular diseases [n = 33, 22% of all deaths, SMR: 1.71 (1.17-2.4)] and diabetes mellitus [n = 26, SMR: 2.30 (1.5-3.37)]. The mortality risks of lung and bronchus cancer, skin tumor, diseases of heart, COPD and allied cond, cerebrovascular diseases, diabetes mellitus, nephritis, nephrotic syndrome and nephrosis, other infectious and parasitic diseases, hypertension, chronic liver disease and cirrhosis and atherosclerosis were significantly increased when compared with the general population. All these results are shown in Table 2. The mortality rate of all causes of death, PeCa, SMTs and non-tumor causes of patients after diagnosis is shown in Figure 2A and the percentage composition of the main causes of death is presented in Figure 3A.Table 2.Main causes of death for patients with localized penile cancer.Causes of deathTotal<1 year<1-5 year5-10 years>10 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death800377.822.12a (1.97-2.27)16758.342.86a (2.44-3.33)422183.122.30a (2.09-2.54)167107.681.55a (1.32-1.8)4428.681.53a (1.11-2.06)All malignant cancers34386.53.97a (3.56-4.41)8013.455.95a (4.72-7.4)20442.354.82a (4.18-5.53)4824.481.96a (1.45-2.6)116.221.77 (.88-3.16) Male genital system21310.7219.86a (17.29-22.72)581.7133.97a (25.79-43.91)1295.224.82a (20.72-29.49)252.998.37a (5.42-12.36)1.831.2 (.03-6.7)  Penis199.082495.58a (2160.87-2867.44)53.014477.48a (3353.94-5856.66)125.043293.53a (2741.5-3924.09)21.02898.28a (556.05-1373.11)0.010 (0-561.23)  Prostate910.59.85 (.39-1.61)21.691.18 (.14-4.28)25.14.39 (.05-1.41)42.951.36 (.37-3.47)1.821.22 (.03-6.78) Urinary system86.291.27 (.55-2.51)3.963.14 (.65-9.16)43.051.31 (.36-3.36)11.81.55 (.01-3.08)0.470 (0-7.81)  Urinary bladder43.81.05 (.29-2.7)2.583.47 (.42-12.54)11.83.55 (.01-3.04)11.1.91 (.02-5.09)0.290 (0-12.7)  Kidney and renal Pelvis12.33.43 (.01-2.39)1.362.81 (.07-15.63)01.140 (0-3.24)0.670 (0-5.54)0.170 (0-21.87)  Other urinary organs3.0933.47a (6.9-97.82)0.010 (0-278.54)3.0469.99a (14.43-204.54)0.030 (0-140.81)0.010 (0-503.78) Digestive system2522.341.12 (.72-1.65)33.41.88 (.18-2.57)1810.891.65 (.98-2.61)36.4.47 (.1-1.37)11.63.61 (.02-3.42)  Liver and intrahepatic bile duct83.712.16 (.93-4.25)0.530 (0-6.92)71.783.93a (1.58-8.09)11.11.9 (.02-5.03)0.280 (0-13.01)  Pancreas75.471.28 (.51-2.63)1.821.22 (.03-6.82)52.651.89 (.61-4.4)11.59.63 (.02-3.5)0.420 (0-8.88)  Esophagus42.931.37 (.37-3.5)0.450 (0-8.21)31.442.08 (.43-6.09)0.840 (0-4.41)1.214.84 (.12-26.98)  Colon and rectum37.41.4 (.08-1.18)21.181.7 (.21-6.13)03.650 (0-1.01)12.06.48 (.01-2.7)0.520 (0-7.13)  Stomach21.831.1 (.13-3.96)0.290 (0-12.81)20.92.23 (.27-8.06)0.510 (0-7.2)0.130 (0-28.41)  Intrahepatic bile duct2.762.63 (.32-9.49)0.110 (0-34.14)1.362.77 (.07-15.41)1.234.36 (.11-24.32)0.060 (0-59.14) Respiratory system4324.311.77a (1.28-2.38)73.871.81 (.73-3.73)2512.072.07a (1.34-3.06)86.761.18 (.51-2.33)31.621.85 (.38-5.41)  Lung and bronchus4023.391.71a (1.22-2.33)73.721.88 (.76-3.87)2411.622.07a (1.32-3.07)66.5.92 (.34-2.01)3z1.93 (.4-5.64) Skin excluding basal and squamous112.284.82a (2.41-8.63)1.352.88 (.07-16.03)61.125.38a (1.97-11.7)3.664.57 (.94-13.37)1.166.17 (.16-34.36) Brain and other nervous system21.761.14 (.14-4.11)0.260 (0-13.99)2.862.34 (.28-8.44)0.510 (0-7.23)0.130 (0-28.57) Lymphoma33.5.86 (.18-2.5)0.540 (0-6.79)11.71.58 (.01-3.25)2.992.01 (.24-7.27)0.250 (0-14.66)  Myeloma21.931.04 (.13-3.75)0.290 (0-12.63)1.931.07 (.03-5.97)1.551.81 (.05-10.08)0.150 (0-24.97) Leukemia33.89.77 (.16-2.25)00.60 (0-6.15)21.91.05 (.13-3.8)11.11.9 (.02-5.02)0.280 (0-13.08) Miscellaneous malignant cancer286.794.12a (2.74-5.96)71.056.64a (2.67-13.67)133.323.91a (2.08-6.69)41.922.08 (.57-5.32)4.498.13#(2.21-20.81)Non-cancer causes Diseases of heart172103.441.66a (1.42-1.93)2916.311.78a (1.19-2.55)9150.311.81a (1.46-2.22)4029.081.38 (.98-1.87)127.741.55 (.8-2.71) COPD and allied cond3823.291.63a (1.15-2.24)83.592.23 (.96-4.4)2011.321.77a (1.08-2.73)96.651.35 (.62-2.57)11.74.57 (.01-3.2) Cerebrovascular diseases3319.341.71#(1.17-2.4)73.042.31 (.93-4.75)109.331.07 (.51-1.97)95.461.65 (.75-3.13)71.514.63#(1.86-9.54) Diabetes mellitus2611.322.30a (1.5-3.37)61.723.49#(1.28-7.6)75.471.28 (.51-2.64)113.253.38a (1.69-6.05)2.882.28 (.28-8.24) Nephritis, nephrotic syndrome and nephrosis168.251.94a (1.11-3.15)31.282.34 (.48-6.83)1142.75a (1.37-4.92)12.33.43 (.01-2.39)1.631.58 (.04-8.79) Pneumonia and influenza149.331.5 (.82-2.52)51.53.34a (1.08-7.79)64.541.32 (.49-2.88)32.61.16 (.24-3.38)00.70 (0-5.3) Other infectious and parasitic diseases including HIV133.134.16a (2.21-7.11)5.4910.17a (3.3-23.74)31.551.93 (.4-5.64)5.885.71a (1.85-13.33)0.210 (0-17.77) Accidents and adverse effects1112.11.91 (.45-1.63)21.821.1 (.13-3.96)45.84.69 (.19-1.75)33.52.85 (.18-2.49)2.932.16 (.26-7.79) Hypertension without heart disease104.222.37#(1.14-4.36)2.623.22 (.39-11.62)11.99.5 (.01-2.8)51.244.04a (1.31-9.42)2.365.52 (.67-19.96) Chronic liver disease and cirrhosis93.82.37a (1.08-4.49)2.573.53 (.43-12.76)41.852.16 (.59-5.53)21.111.8 (.22-6.49)1.273.65 (.09-20.36)AbbreviationsSMR: standardized mortality ratio; CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease.aStatistical significance with P < .05.Figure 2.Mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients diagnosed with localized, regional and distant metastasis penile cancer. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.Figure 3.The percentages of main causes of death in penile cancer patients. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.\nMain causes of death for patients with localized penile cancer.\nAbbreviationsSMR: standardized mortality ratio; CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease.\naStatistical significance with P < .05.\nMortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients diagnosed with localized, regional and distant metastasis penile cancer. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.\nThe percentages of main causes of death in penile cancer patients. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.", "For patients with regional PeCa, 348 (51.3%) of all 679 deaths were due to PeCa itself. There were 118 SMTs deaths and 213 non-cancer deaths, accounting for 17.4% and 31.4% of all deaths. For the PeCa deaths, 89.1% of them occurred within 3 years after diagnosis. With the low PeCa mortality rate of the general population, the SMR of PeCa death reached a terribly high level. The main SMTs were lung and bronchus cancer [n = 23, SMR: 2.41 (1.53-3.62)]. The mortality risks of lung and bronchus cancer, and skin tumor [SMR: 6.41 (2.35-13.95)] were significantly increased. The most common non-cancer cause of death was diseases of heart [n = 71, SMR: 1.77 (1.38-2.23)], COPD and allied cond [n = 17, SMR: 1.85 (1.08-2.95)], diabetes mellitus [n = 16, SMR: 3.62 (2.07-5.88)] and cerebrovascular diseases [n = 13, SMR: 1.77 (.94-3.03)]. The mortality risks of diseases of heart, COPD and allied cond, diabetes mellitus, nephritis, nephrotic syndrome and nephrosis [n = 9, SMR: 2.88 (1.32-5.47)] and septicemia [n = 8, SMR: 3.83 (1.65-7.54)] were significantly increased when compared with the general population. All these results are shown in Table 3. The mortality rate of patients after diagnosis and is shown in Figure 2B and the percentages of the main causes of death is revealed in Figure 3B.Table 3.Main causes of death for patients with regional penile cancer.Causes of deathTotal<1 year1-5 years>5 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death679147.44.61a (4.27-4.97)27527.559.98a (8.84-11.23)31067.934.56a (4.07-5.1)9451.921.81a (1.46-2.22)All malignant cancers46634.6513.45a (12.26-14.73)2136.5632.48a (28.27-37.15)22516.4213.71a (11.97-15.62)2811.672.40a (1.59-3.47) Male genital system3624.0788.89a (79.97-98.54)179.79225.26a (193.47-260.79)1681.8889.54a (76.51-104.15)151.410.70a (5.99-17.65)  Penis348.0311 020.00a (9892.46-12,240.84)173.0129 605.21a (25 357.89-34 360.42)162.0111 182.19a (9526.53-13042.87)13.011155.76a (615.39-1976.38)  Prostate84.021.99 (.86-3.92)2.782.55 (.31-9.21)41.852.16 (.59-5.53)21.381.45 (.18-5.22)  Testis2.01149.35a (18.09-539.5)10370.55a (9.38-2064.6)1.01152.52a (3.86-849.81)000 (0-891.78) Urinary system62.512.39 (.88-5.21)1.462.18 (.06-12.16)41.153.47 (.94-8.87)1.891.12 (.03-6.23)  Kidney and renal Pelvis3.953.17 (.65-9.27)0.180 (0-20.85)2.454.46 (.54-16.13)1.323.12 (.08-17.36)  Urinary bladder21.51.34 (.16-4.83)1.273.71 (.09-20.65)1.681.48 (.04-8.23)0.550 (0-6.72) Digestive system78.97.78 (.31-1.61)11.69.59 (.01-3.3)44.27.94 (.26-2.4)23.01.66 (.08-2.4)  Colon and rectum32.951.02 (.21-2.97)0.570 (0-6.44)21.411.41 (.17-5.11)1.971.03 (.03-5.77)  Esophagus21.211.65 (.2-5.95)0.230 (0-16.24)1.581.72 (.04-9.57)1.412.47 (.06-13.75) Respiratory system239.92.32a (1.47-3.49)41.912.1 (.57-5.37)134.82.71a (1.44-4.63)63.191.88 (.69-4.09)  Lung and bronchus239.532.41a (1.53-3.62)41.842.18 (.59-5.58)134.622.81a (1.5-4.81)63.071.95 (.72-4.25) Skin excluding basal and squamous6.946.41a (2.35-13.95)2.1711.79a (1.43-42.6)4.439.23a (2.51-23.62)0.330 (0-11.07) Lymphoma21.411.42 (.17-5.14)0.260 (0-14.17)2.653.05 (.37-11.03)0.490 (0-7.53) Leukemia21.561.28 (.16-4.63)0.290 (0-12.84)2.722.76 (.33-9.97)0.550 (0-6.74) Miscellaneous malignant cancer512.7218.75a (13.96-24.66)25.5148.95a (31.68-72.26)241.2818.76a (12.02-27.92)2.932.15 (.26-7.77)Non-cancer causes Diseases of heart7140.111.77a (1.38-2.23)207.572.64#(1.61-4.08)3218.451.73a (1.19-2.45)1914.091.35 (.81-2.11) COPD and allied cond179.211.85a (1.08-2.95)41.692.37 (.65-6.08)84.241.89 (.82-3.72)53.291.52 (.49-3.55) Diabetes mellitus164.423.62a (2.07-5.88)5.836.00a (1.95-14.01)62.072.89a (1.06-6.3)51.513.31a (1.08-7.73) Cerebrovascular diseases137.341.77 (.94-3.03)31.392.16 (.44-6.3)63.341.79 (.66-3.91)42.611.53 (.42-3.92) Nephritis, nephrotic syndrome and nephrosis93.132.88a (1.32-5.47)3.595.07a (1.05-14.81)51.443.48a (1.13-8.12)11.1.91 (.02-5.08) Septicemia82.093.83a (1.65-7.54)50.412.66a (4.11-29.53)1.961.04 (.03-5.77)2.732.74 (.33-9.89) Alzheimers84.791.67 (.72-3.29)3.843.56 (.74-10.42)22.02.99 (.12-3.57)31.931.56 (.32-4.55) Chronic liver disease and cirrhosis41.622.47 (.67-6.32)3.319.67a (1.99-28.25)0.790 (0-4.64)1.521.94 (.05-10.8) Accidents and adverse effects64.841.24 (.45-2.7)0.910 (0-4.04)32.261.33 (.27-3.88)31.671.8 (.37-5.25) Suicide and self-inflicted injury41.382.9 (.79-7.42)1.273.71 (.09-20.68)1.671.49 (.04-8.28)2.444.57 (.55-16.51)CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease; SMR: standardized mortality ratio.aStatistical significance with P < .05.\nMain causes of death for patients with regional penile cancer.\nCI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease; SMR: standardized mortality ratio.\naStatistical significance with P < .05.", "For the included 132 patients with distant metastasis PeCa, 109 of them died during the follow-up with a significantly increased SMR of 27.68 (22.73-33.39). 100 deaths (91.7%) were due to malignant cancers including 76 cases (69.7%) dead of PeCa. 67.1% (n = 51) of all PeCa deaths occurred within 1 year and 98.7% (n = 75) dead within 3 years after diagnosis. There were 24 patients (22%) who died for SMTs, mainly including cancers from 4 lung and bronchus, 2 prostate, and 2 skin, 2 lymphoma and 3 other urinary organs. 19 (79.2%) of SMTs deaths occurred within 1 year of the diagnosis of PeCa. As for the non-cancer deaths, only 9 cases of deaths were observed, including 5 deaths from diseases of heart, 1 septicemia death, 1 other infection and parasitic diseases deaths, and 2 other causes of death. These results are presented in Table 4 and Figure 3C. The mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients after diagnosis is shown in Figure 2C.Table 4.Main Causes of Death for Patients With Distant Penile Cancer.Causes of deathTotal<1 year1-3 years>3 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death1093.9427.68a (22.73-33.39)741.8540.10a (31.49-50.34)281.0925.61a (17.02-37.01)717.00a (2.82-14.43)All malignant cancers100.98102.30a (83.23-124.42)70.45154.76a (120.65-195.54)27.2992.21a (60.76-134.16)3.2312.91a (2.66-37.72) Respiratory system4.2814.38a (3.92-36.81)3.1322.93a (4.73-67.01)0.090 (0-42.56)1.0616.48 (.42-91.8)  Lung and bronchus4.2714.98a (4.08-38.34)3.1323.85a (4.92-69.69)0.080 (0-44.28)1.0617.24 (.44-96.06) Soft tissue including heart2.01331.85a (40.19-1198.77)10351.01a (8.89-1955.69)10554.76a (14.05-3090.9)000 (0-2682.36) Skin excluding basal and squamous2.0285.91a (10.4-310.32)1.0187.34a (2.21-486.61)1.01146.53a (3.71-816.42)0.010 (0-736.74)  Male genital system80.12650.98a (516.18-810.2)55.051065.43a (802.63-1386.8)24.03713.33a (457.04-1061.38)1.0426.58 (.67-148.08)  Prostate2.1216.48a (2-59.52)2.0539.26a (4.75-141.82)0.030 (0-111.09)0.040 (0-99.09)  Penis76084 751.04a (66 774.18-106078.56)510126 908.70a (94 491.86-166861.5)24092 697.90a (59 393.29-137,927.13)104237.74a (107.29-23,611.17) Urinary system3.0646.59a (9.61-136.15)2.0364.45a (7.81-232.82)0.020 (0-199.1)1.0167.40a (1.71-375.54) Miscellaneous malignant cancer7.0793.72a (37.68-193.1)7.04198.90a (79.97-409.8)0.020 (0-166.27)0.020 (0-213.09)Non-cancer causes Septicemia1.0616.57 (.42-92.3)1.0337.63 (.95-209.66)0.020 (0-218.26)0.020 (0-218.46) Other infectious and parasitic diseases including HIV1.0426.85 (.68-149.58)1.0257.29a (1.45-319.22)0.010 (0-340.04)0.010 (0-412.29) Diseases of heart51.054.75a (1.54-11.09)1.492.02 (.05-11.26)1.283.52 (.09-19.64)3.2710.95a (2.26-32) Other cause of death2.573.53 (.43-12.75)1.273.69 (.09-20.57)0.150 (0-25.01)1.156.75 (.17-37.6)SMR: standardized mortality ratio; CI: confidence interval.aStatistical significance with P < .05.\nMain Causes of Death for Patients With Distant Penile Cancer.\nSMR: standardized mortality ratio; CI: confidence interval.\naStatistical significance with P < .05." ]
[ null, null, null, null, null, null, null ]
[ "Introduction", "Methods", "Data Sources", "Study Population and Study Variables", "Outcome Assessments", "Statistics Analyses", "Results", "Causes of Death for Patients With Localized Penile Cancer", "Causes of Death for Patients With Regional Penile Cancer", "Causes of Death for Patients With Distant Metastatic Penile Cancer", "Discussion", "Conclusion", "Supplemental Material" ]
[ "Penile cancer (PeCa) is a rare malignancy worldwide, accounting for less than 1% of all malignancies in men.1 The overall incidence rate is approximately .69 cases per 100 000 persons in the US.2 It was estimated that the number of new PeCa patients was 34 475 and deaths cases was 15 138 each year worldwide.3 The survival rates still vary greatly with the stages of patients’ disease. It was reported that about 40% of new cases were diagnosed with localized PeCa, with a 5-year overall survival rate of approximately 90%.4 The survival rates decrease dramatically once patients are in the regional or distant metastasis stage. The 5-year overall survival rate is approximately 80% with unilateral inguinal lymph node involvement, 10%-20% with bilateral or pelvic lymph node involvement, and <10% with extranodal extension.4\nSince the 1990s, substantial progress has been made in the prevention, diagnosis and treatment of PeCa. The prognosis has been greatly improved in PeCa patients, especially those who have not progressed to the metastatic stage. With the prolonged survival time, the proportion of other causes of death such as second malignant tumors (SMTs) and non-tumor diseases has been increasing among all deaths of PeCa patients. Since few previous studies have focused exclusively on other causes of death of PeCa patients, an analysis of causes of death among PeCa patients is warranted. Our study aimed to evaluate the main causes of death including SMTs and non-cancer causes for PeCa patients, and calculate the standardized mortality ratio (SMR) for each reason of death compared with the normal population.", "[SUBTITLE] Data Sources [SUBSECTION] The data of this study was derived from the Surveillance, Epidemiology, and End Results (SEER) program 18 registries, which is an authoritative source of information on cancer incidence and survival in the United States. SEER currently collects and publishes cancer incidence and survival data from population-based cancer registries covering approximately 27.8% of the U.S. population, and it collects data concerning patient demographics, tumor morphology, stage at diagnosis, primary tumor site, the first course of treatment, and follow-up for vital status. Meanwhile, the data for the general population comes from the Centers for Disease Control and Prevention (CDC) of the United States. All data in this study has been removed identifiable information and can be available freely online. Thus, this study was regarded as exempt research by the institutional review.\nThe data of this study was derived from the Surveillance, Epidemiology, and End Results (SEER) program 18 registries, which is an authoritative source of information on cancer incidence and survival in the United States. SEER currently collects and publishes cancer incidence and survival data from population-based cancer registries covering approximately 27.8% of the U.S. population, and it collects data concerning patient demographics, tumor morphology, stage at diagnosis, primary tumor site, the first course of treatment, and follow-up for vital status. Meanwhile, the data for the general population comes from the Centers for Disease Control and Prevention (CDC) of the United States. All data in this study has been removed identifiable information and can be available freely online. Thus, this study was regarded as exempt research by the institutional review.\n[SUBTITLE] Study Population and Study Variables [SUBSECTION] Patients with PeCa as the first primary malignancy and clear stage information according to the definition of Summary Stage 2000 (1998+) between 2004 and 2018 were included. The patients who had unclear follow-up time, living status at the end of follow-up and detailed causes of death were excluded.\nPatients were divided into 3 groups by their stages (localized, regional and distant metastasis). The following variables of all patients and the dead patients were analyzed, including age (15-54, 55-64, 65-74, 75-84, and >85 years), year of diagnosis (2004-2008, 2009-2013, and 2014-2018), race (white, black. American Indian/Alaska native and Asian or Pacific Islander), grade (well-differentiated, moderately differentiated, poorly differentiated, and undifferentiated), main pathological type, Radiotherapy (beam radiation, radioactive implants/brachytherapy and none/unknown) and Chemotherapy (yes and no/unknown).\nPatients with PeCa as the first primary malignancy and clear stage information according to the definition of Summary Stage 2000 (1998+) between 2004 and 2018 were included. The patients who had unclear follow-up time, living status at the end of follow-up and detailed causes of death were excluded.\nPatients were divided into 3 groups by their stages (localized, regional and distant metastasis). The following variables of all patients and the dead patients were analyzed, including age (15-54, 55-64, 65-74, 75-84, and >85 years), year of diagnosis (2004-2008, 2009-2013, and 2014-2018), race (white, black. American Indian/Alaska native and Asian or Pacific Islander), grade (well-differentiated, moderately differentiated, poorly differentiated, and undifferentiated), main pathological type, Radiotherapy (beam radiation, radioactive implants/brachytherapy and none/unknown) and Chemotherapy (yes and no/unknown).\n[SUBTITLE] Outcome Assessments [SUBSECTION] The causes of death after the diagnosis of PeCa included PeCa deaths, SMTs, and non-cancer deaths were regarded as the main outcomes. The standardized mortality ratio (SMR) with the 95% confidence interval (95% CI) of each cause was also calculated to evaluate the mortality risks compared with the general population. The last follow-up time was December 31, 2018, which was the latest data updated in the SEER database.\nThe causes of death after the diagnosis of PeCa included PeCa deaths, SMTs, and non-cancer deaths were regarded as the main outcomes. The standardized mortality ratio (SMR) with the 95% confidence interval (95% CI) of each cause was also calculated to evaluate the mortality risks compared with the general population. The last follow-up time was December 31, 2018, which was the latest data updated in the SEER database.\n[SUBTITLE] Statistics Analyses [SUBSECTION] For the baseline characteristics of 3 groups of patients with different PeCa stages, we analyzed the number of all included patients and observed deaths stratified by the above variables. The SMRs for each variable were calculated by comparing the observed number of deaths to the expected number. The expected number of deaths was based on the total number of patient-year and the excess risk of the general population. The causes of death and related SMRs and 95% CI were also analyzed in localized, regional, and distant metastasis PeCa patients. Statistical significance for SMR is a two-sided test of P < .05. All these analyses were performed using SEER*Stat version 8.4.0.1. Pie charts of the proportion of different causes of death were drawn with Microsoft Excel 2016.\nFor the baseline characteristics of 3 groups of patients with different PeCa stages, we analyzed the number of all included patients and observed deaths stratified by the above variables. The SMRs for each variable were calculated by comparing the observed number of deaths to the expected number. The expected number of deaths was based on the total number of patient-year and the excess risk of the general population. The causes of death and related SMRs and 95% CI were also analyzed in localized, regional, and distant metastasis PeCa patients. Statistical significance for SMR is a two-sided test of P < .05. All these analyses were performed using SEER*Stat version 8.4.0.1. Pie charts of the proportion of different causes of death were drawn with Microsoft Excel 2016.", "The data of this study was derived from the Surveillance, Epidemiology, and End Results (SEER) program 18 registries, which is an authoritative source of information on cancer incidence and survival in the United States. SEER currently collects and publishes cancer incidence and survival data from population-based cancer registries covering approximately 27.8% of the U.S. population, and it collects data concerning patient demographics, tumor morphology, stage at diagnosis, primary tumor site, the first course of treatment, and follow-up for vital status. Meanwhile, the data for the general population comes from the Centers for Disease Control and Prevention (CDC) of the United States. All data in this study has been removed identifiable information and can be available freely online. Thus, this study was regarded as exempt research by the institutional review.", "Patients with PeCa as the first primary malignancy and clear stage information according to the definition of Summary Stage 2000 (1998+) between 2004 and 2018 were included. The patients who had unclear follow-up time, living status at the end of follow-up and detailed causes of death were excluded.\nPatients were divided into 3 groups by their stages (localized, regional and distant metastasis). The following variables of all patients and the dead patients were analyzed, including age (15-54, 55-64, 65-74, 75-84, and >85 years), year of diagnosis (2004-2008, 2009-2013, and 2014-2018), race (white, black. American Indian/Alaska native and Asian or Pacific Islander), grade (well-differentiated, moderately differentiated, poorly differentiated, and undifferentiated), main pathological type, Radiotherapy (beam radiation, radioactive implants/brachytherapy and none/unknown) and Chemotherapy (yes and no/unknown).", "The causes of death after the diagnosis of PeCa included PeCa deaths, SMTs, and non-cancer deaths were regarded as the main outcomes. The standardized mortality ratio (SMR) with the 95% confidence interval (95% CI) of each cause was also calculated to evaluate the mortality risks compared with the general population. The last follow-up time was December 31, 2018, which was the latest data updated in the SEER database.", "For the baseline characteristics of 3 groups of patients with different PeCa stages, we analyzed the number of all included patients and observed deaths stratified by the above variables. The SMRs for each variable were calculated by comparing the observed number of deaths to the expected number. The expected number of deaths was based on the total number of patient-year and the excess risk of the general population. The causes of death and related SMRs and 95% CI were also analyzed in localized, regional, and distant metastasis PeCa patients. Statistical significance for SMR is a two-sided test of P < .05. All these analyses were performed using SEER*Stat version 8.4.0.1. Pie charts of the proportion of different causes of death were drawn with Microsoft Excel 2016.", "A total of 3597 PeCa patients were included, 1588 (44%) of them died during the follow-up. The majority of patients was in the localized stage (n = 2,155, 60%) and regional stage (n = 1310, 36%), whereas only 4% (n = 132) had distant disease. For patients with localized PeCa, 800 (37.1%) of them died with a SMR of 2.12 (1.97-2.27). 679 (51.8%) regional PeCa patients [SMR: 4.61 (4.27-4.97)] and 109 (82.6%) patients with distant metastatic PeCa [SMR: 27.68 (22.73-33.39)] died during the follow-up.\nBaseline characteristics of included patients and the dead cases were presented in Table 1. The trend of new diagnosed PeCa patients from 2000 to 2018 were revealed in Figure 1.Table 1.Baseline Characteristics of All Patients and Dead Cases During Follow-Up With Localized, Regional, and Distant Penile Cancer.CharacteristicLocalizedRegionalDistantPatients, nDeaths, nSMR (95% CI)Patients, nDeaths, nSMR (95% CI)Patients, nDeaths, nSMR (95% CI)Total21558002.12a (1.97-2.27)13106794.61a (4.27-4.97)13210927.68a (22.73-33.39)Age 15-54 years470804.41a (3.5-5.49)32912116.05a (13.32-19.18)3630106.43a (71.81-151.93) 55-64 years4991282.86a (2.39-3.4)3371537.48a (6.34-8.77)322748.63a (32.05-70.75) 65-74 years5652102.21a (1.92-2.53)3301834.67a (4.01-5.39)332635.53a (23.21-52.06) 75-84 years4102221.55a (1.35-1.76)2171432.61a (2.2-3.08)262110.51a (6.51-16.07) 85+ years2111602.10a (1.79-2.45)97793.10a (2.45-3.86)5513.45a (4.37-31.39)Year of diagnosis 2004-20087301432.65a (2.23-3.12)3532343.11a (2.73-3.54)302717.20a (11.34-25.03) 2009-20137442842.05a (1.82-2.3)4422585.32a (4.69-6.01)484232.68a (23.55-44.17) 2014-20186813732.01a (1.81-2.23)5151877.88a (6.79-9.1)544036.91a (26.37-50.27)Race White18066782.08a (1.92-2.24)11205774.35a (4-4.72)1058326.90a (21.43-33.35) Black222842.48a(1.98-3.07)116727.28a (5.7-9.17)161520.92a (11.71-34.51) American Indian/Alaska native2183.79a (1.64-7.47)1137.25a (1.49-21.18)33586.91a (121.03-1715.19) Asian or Pacific islander106301.95a (1.31-2.78)63276.22a (4.1-9.05)8861.02a (26.35-120.24)Grade Well differentiated6312181.78a (1.55-2.03)188863.01a (2.41-3.71)7620.27a (7.44-44.12) Moderately differentiated7063042.55a (2.27-2.85)5743175.07a (4.52-5.65)383219.92a (13.62-28.11) Poorly differentiated2571302.93a (2.44-3.47)3171925.02a (4.34-5.79)444135.67a (25.6-48.39) Undifferentiated92.7 (.08-2.52)1175.63a (2.27-11.61)329.89a (1.2-35.72)Pathological type 8070/3: Squamous cell carcinoma, NOS14255602.22a (2.04-2.41)8294474.49a (4.08-4.93)846830.34a (23.56-38.46) 8071/3: Squamous cell carcinoma, keratinizing, NOS3471262.26a (1.88-2.69)3321655.54a (4.73-6.46)191414.96a (8.18-25.1) 8051/3: Verrucous carcinoma, NOS169491.50a (1.11-1.98)39101.54 (.74-2.84)2298.29a (11.9-355.06) 8083/3: Basaloid squamous cell carcinoma41122.96a (1.53-5.18)34165.50a(3.14-8.93)6523.04a (7.48-53.77)Radiotherapy Beam radiation68301.90a (1.28-2.72)1981288.26a(6.89-9.83)372945.93a(30.76-65.96) Radioactive implants/brachytherapy214.79 (.12-26.7)3112.38 (.31-68.98)000 None/unknown20857692.13a (1.98-2.28)11095504.17a(3.83-4.54)958024.19a(19.18-30.11)Chemotherapy  Yes59222.69a (1.69-4.08)29816711.38a(9.72-13.24)745831.87a(24.2-41.21)  No/unknown20967782.10a (1.96-2.26)10125123.86a(3.53-4.21)585124.07a(17.92-31.65)SMR: standardized mortality ratio; 95% CI: 95% Confidence interval; NOS: Not otherwise specified.aStatistical significance with P < .05.Figure 1.The number of newly diagnosed of penile cancer from 2004 to 2018.\nBaseline Characteristics of All Patients and Dead Cases During Follow-Up With Localized, Regional, and Distant Penile Cancer.\nSMR: standardized mortality ratio; 95% CI: 95% Confidence interval; NOS: Not otherwise specified.\naStatistical significance with P < .05.\nThe number of newly diagnosed of penile cancer from 2004 to 2018.\n[SUBTITLE] Causes of Death for Patients With Localized Penile Cancer [SUBSECTION] Among the 800 cases of death, 199 (24.9%) died for PeCa, 144 (18.0%) died for other tumor causes, and the rest 457 (57.1%) deaths were due to non-tumor causes. 75.9% of all PeCa deaths were within 3 years after diagnosis. The mortality rate of the general population who died for PeCa was rather low, which led to the extremely high SMR from the ratio of observed and expected. The majority of SMTs deaths (n = 106, 73.6%) occurred with 5 years after the diagnosis of PeCa. The main SMTs were lung and bronchus cancer [n = 40, SMR: 1.71 (1.22-2.33)], all cancers of digestive system [n = 25, SMR: 1.12 (.72-1.65)]. The main non-cancer causes of death were diseases of heart [n = 172, SMR: 1.66 (1.42-1.93)], chronic obstructive pulmonary disease (COPD) and allied cond [n = 38, SMR: 1.63 (1.15-2.24)], cerebrovascular diseases [n = 33, 22% of all deaths, SMR: 1.71 (1.17-2.4)] and diabetes mellitus [n = 26, SMR: 2.30 (1.5-3.37)]. The mortality risks of lung and bronchus cancer, skin tumor, diseases of heart, COPD and allied cond, cerebrovascular diseases, diabetes mellitus, nephritis, nephrotic syndrome and nephrosis, other infectious and parasitic diseases, hypertension, chronic liver disease and cirrhosis and atherosclerosis were significantly increased when compared with the general population. All these results are shown in Table 2. The mortality rate of all causes of death, PeCa, SMTs and non-tumor causes of patients after diagnosis is shown in Figure 2A and the percentage composition of the main causes of death is presented in Figure 3A.Table 2.Main causes of death for patients with localized penile cancer.Causes of deathTotal<1 year<1-5 year5-10 years>10 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death800377.822.12a (1.97-2.27)16758.342.86a (2.44-3.33)422183.122.30a (2.09-2.54)167107.681.55a (1.32-1.8)4428.681.53a (1.11-2.06)All malignant cancers34386.53.97a (3.56-4.41)8013.455.95a (4.72-7.4)20442.354.82a (4.18-5.53)4824.481.96a (1.45-2.6)116.221.77 (.88-3.16) Male genital system21310.7219.86a (17.29-22.72)581.7133.97a (25.79-43.91)1295.224.82a (20.72-29.49)252.998.37a (5.42-12.36)1.831.2 (.03-6.7)  Penis199.082495.58a (2160.87-2867.44)53.014477.48a (3353.94-5856.66)125.043293.53a (2741.5-3924.09)21.02898.28a (556.05-1373.11)0.010 (0-561.23)  Prostate910.59.85 (.39-1.61)21.691.18 (.14-4.28)25.14.39 (.05-1.41)42.951.36 (.37-3.47)1.821.22 (.03-6.78) Urinary system86.291.27 (.55-2.51)3.963.14 (.65-9.16)43.051.31 (.36-3.36)11.81.55 (.01-3.08)0.470 (0-7.81)  Urinary bladder43.81.05 (.29-2.7)2.583.47 (.42-12.54)11.83.55 (.01-3.04)11.1.91 (.02-5.09)0.290 (0-12.7)  Kidney and renal Pelvis12.33.43 (.01-2.39)1.362.81 (.07-15.63)01.140 (0-3.24)0.670 (0-5.54)0.170 (0-21.87)  Other urinary organs3.0933.47a (6.9-97.82)0.010 (0-278.54)3.0469.99a (14.43-204.54)0.030 (0-140.81)0.010 (0-503.78) Digestive system2522.341.12 (.72-1.65)33.41.88 (.18-2.57)1810.891.65 (.98-2.61)36.4.47 (.1-1.37)11.63.61 (.02-3.42)  Liver and intrahepatic bile duct83.712.16 (.93-4.25)0.530 (0-6.92)71.783.93a (1.58-8.09)11.11.9 (.02-5.03)0.280 (0-13.01)  Pancreas75.471.28 (.51-2.63)1.821.22 (.03-6.82)52.651.89 (.61-4.4)11.59.63 (.02-3.5)0.420 (0-8.88)  Esophagus42.931.37 (.37-3.5)0.450 (0-8.21)31.442.08 (.43-6.09)0.840 (0-4.41)1.214.84 (.12-26.98)  Colon and rectum37.41.4 (.08-1.18)21.181.7 (.21-6.13)03.650 (0-1.01)12.06.48 (.01-2.7)0.520 (0-7.13)  Stomach21.831.1 (.13-3.96)0.290 (0-12.81)20.92.23 (.27-8.06)0.510 (0-7.2)0.130 (0-28.41)  Intrahepatic bile duct2.762.63 (.32-9.49)0.110 (0-34.14)1.362.77 (.07-15.41)1.234.36 (.11-24.32)0.060 (0-59.14) Respiratory system4324.311.77a (1.28-2.38)73.871.81 (.73-3.73)2512.072.07a (1.34-3.06)86.761.18 (.51-2.33)31.621.85 (.38-5.41)  Lung and bronchus4023.391.71a (1.22-2.33)73.721.88 (.76-3.87)2411.622.07a (1.32-3.07)66.5.92 (.34-2.01)3z1.93 (.4-5.64) Skin excluding basal and squamous112.284.82a (2.41-8.63)1.352.88 (.07-16.03)61.125.38a (1.97-11.7)3.664.57 (.94-13.37)1.166.17 (.16-34.36) Brain and other nervous system21.761.14 (.14-4.11)0.260 (0-13.99)2.862.34 (.28-8.44)0.510 (0-7.23)0.130 (0-28.57) Lymphoma33.5.86 (.18-2.5)0.540 (0-6.79)11.71.58 (.01-3.25)2.992.01 (.24-7.27)0.250 (0-14.66)  Myeloma21.931.04 (.13-3.75)0.290 (0-12.63)1.931.07 (.03-5.97)1.551.81 (.05-10.08)0.150 (0-24.97) Leukemia33.89.77 (.16-2.25)00.60 (0-6.15)21.91.05 (.13-3.8)11.11.9 (.02-5.02)0.280 (0-13.08) Miscellaneous malignant cancer286.794.12a (2.74-5.96)71.056.64a (2.67-13.67)133.323.91a (2.08-6.69)41.922.08 (.57-5.32)4.498.13#(2.21-20.81)Non-cancer causes Diseases of heart172103.441.66a (1.42-1.93)2916.311.78a (1.19-2.55)9150.311.81a (1.46-2.22)4029.081.38 (.98-1.87)127.741.55 (.8-2.71) COPD and allied cond3823.291.63a (1.15-2.24)83.592.23 (.96-4.4)2011.321.77a (1.08-2.73)96.651.35 (.62-2.57)11.74.57 (.01-3.2) Cerebrovascular diseases3319.341.71#(1.17-2.4)73.042.31 (.93-4.75)109.331.07 (.51-1.97)95.461.65 (.75-3.13)71.514.63#(1.86-9.54) Diabetes mellitus2611.322.30a (1.5-3.37)61.723.49#(1.28-7.6)75.471.28 (.51-2.64)113.253.38a (1.69-6.05)2.882.28 (.28-8.24) Nephritis, nephrotic syndrome and nephrosis168.251.94a (1.11-3.15)31.282.34 (.48-6.83)1142.75a (1.37-4.92)12.33.43 (.01-2.39)1.631.58 (.04-8.79) Pneumonia and influenza149.331.5 (.82-2.52)51.53.34a (1.08-7.79)64.541.32 (.49-2.88)32.61.16 (.24-3.38)00.70 (0-5.3) Other infectious and parasitic diseases including HIV133.134.16a (2.21-7.11)5.4910.17a (3.3-23.74)31.551.93 (.4-5.64)5.885.71a (1.85-13.33)0.210 (0-17.77) Accidents and adverse effects1112.11.91 (.45-1.63)21.821.1 (.13-3.96)45.84.69 (.19-1.75)33.52.85 (.18-2.49)2.932.16 (.26-7.79) Hypertension without heart disease104.222.37#(1.14-4.36)2.623.22 (.39-11.62)11.99.5 (.01-2.8)51.244.04a (1.31-9.42)2.365.52 (.67-19.96) Chronic liver disease and cirrhosis93.82.37a (1.08-4.49)2.573.53 (.43-12.76)41.852.16 (.59-5.53)21.111.8 (.22-6.49)1.273.65 (.09-20.36)AbbreviationsSMR: standardized mortality ratio; CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease.aStatistical significance with P < .05.Figure 2.Mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients diagnosed with localized, regional and distant metastasis penile cancer. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.Figure 3.The percentages of main causes of death in penile cancer patients. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.\nMain causes of death for patients with localized penile cancer.\nAbbreviationsSMR: standardized mortality ratio; CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease.\naStatistical significance with P < .05.\nMortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients diagnosed with localized, regional and distant metastasis penile cancer. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.\nThe percentages of main causes of death in penile cancer patients. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.\nAmong the 800 cases of death, 199 (24.9%) died for PeCa, 144 (18.0%) died for other tumor causes, and the rest 457 (57.1%) deaths were due to non-tumor causes. 75.9% of all PeCa deaths were within 3 years after diagnosis. The mortality rate of the general population who died for PeCa was rather low, which led to the extremely high SMR from the ratio of observed and expected. The majority of SMTs deaths (n = 106, 73.6%) occurred with 5 years after the diagnosis of PeCa. The main SMTs were lung and bronchus cancer [n = 40, SMR: 1.71 (1.22-2.33)], all cancers of digestive system [n = 25, SMR: 1.12 (.72-1.65)]. The main non-cancer causes of death were diseases of heart [n = 172, SMR: 1.66 (1.42-1.93)], chronic obstructive pulmonary disease (COPD) and allied cond [n = 38, SMR: 1.63 (1.15-2.24)], cerebrovascular diseases [n = 33, 22% of all deaths, SMR: 1.71 (1.17-2.4)] and diabetes mellitus [n = 26, SMR: 2.30 (1.5-3.37)]. The mortality risks of lung and bronchus cancer, skin tumor, diseases of heart, COPD and allied cond, cerebrovascular diseases, diabetes mellitus, nephritis, nephrotic syndrome and nephrosis, other infectious and parasitic diseases, hypertension, chronic liver disease and cirrhosis and atherosclerosis were significantly increased when compared with the general population. All these results are shown in Table 2. The mortality rate of all causes of death, PeCa, SMTs and non-tumor causes of patients after diagnosis is shown in Figure 2A and the percentage composition of the main causes of death is presented in Figure 3A.Table 2.Main causes of death for patients with localized penile cancer.Causes of deathTotal<1 year<1-5 year5-10 years>10 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death800377.822.12a (1.97-2.27)16758.342.86a (2.44-3.33)422183.122.30a (2.09-2.54)167107.681.55a (1.32-1.8)4428.681.53a (1.11-2.06)All malignant cancers34386.53.97a (3.56-4.41)8013.455.95a (4.72-7.4)20442.354.82a (4.18-5.53)4824.481.96a (1.45-2.6)116.221.77 (.88-3.16) Male genital system21310.7219.86a (17.29-22.72)581.7133.97a (25.79-43.91)1295.224.82a (20.72-29.49)252.998.37a (5.42-12.36)1.831.2 (.03-6.7)  Penis199.082495.58a (2160.87-2867.44)53.014477.48a (3353.94-5856.66)125.043293.53a (2741.5-3924.09)21.02898.28a (556.05-1373.11)0.010 (0-561.23)  Prostate910.59.85 (.39-1.61)21.691.18 (.14-4.28)25.14.39 (.05-1.41)42.951.36 (.37-3.47)1.821.22 (.03-6.78) Urinary system86.291.27 (.55-2.51)3.963.14 (.65-9.16)43.051.31 (.36-3.36)11.81.55 (.01-3.08)0.470 (0-7.81)  Urinary bladder43.81.05 (.29-2.7)2.583.47 (.42-12.54)11.83.55 (.01-3.04)11.1.91 (.02-5.09)0.290 (0-12.7)  Kidney and renal Pelvis12.33.43 (.01-2.39)1.362.81 (.07-15.63)01.140 (0-3.24)0.670 (0-5.54)0.170 (0-21.87)  Other urinary organs3.0933.47a (6.9-97.82)0.010 (0-278.54)3.0469.99a (14.43-204.54)0.030 (0-140.81)0.010 (0-503.78) Digestive system2522.341.12 (.72-1.65)33.41.88 (.18-2.57)1810.891.65 (.98-2.61)36.4.47 (.1-1.37)11.63.61 (.02-3.42)  Liver and intrahepatic bile duct83.712.16 (.93-4.25)0.530 (0-6.92)71.783.93a (1.58-8.09)11.11.9 (.02-5.03)0.280 (0-13.01)  Pancreas75.471.28 (.51-2.63)1.821.22 (.03-6.82)52.651.89 (.61-4.4)11.59.63 (.02-3.5)0.420 (0-8.88)  Esophagus42.931.37 (.37-3.5)0.450 (0-8.21)31.442.08 (.43-6.09)0.840 (0-4.41)1.214.84 (.12-26.98)  Colon and rectum37.41.4 (.08-1.18)21.181.7 (.21-6.13)03.650 (0-1.01)12.06.48 (.01-2.7)0.520 (0-7.13)  Stomach21.831.1 (.13-3.96)0.290 (0-12.81)20.92.23 (.27-8.06)0.510 (0-7.2)0.130 (0-28.41)  Intrahepatic bile duct2.762.63 (.32-9.49)0.110 (0-34.14)1.362.77 (.07-15.41)1.234.36 (.11-24.32)0.060 (0-59.14) Respiratory system4324.311.77a (1.28-2.38)73.871.81 (.73-3.73)2512.072.07a (1.34-3.06)86.761.18 (.51-2.33)31.621.85 (.38-5.41)  Lung and bronchus4023.391.71a (1.22-2.33)73.721.88 (.76-3.87)2411.622.07a (1.32-3.07)66.5.92 (.34-2.01)3z1.93 (.4-5.64) Skin excluding basal and squamous112.284.82a (2.41-8.63)1.352.88 (.07-16.03)61.125.38a (1.97-11.7)3.664.57 (.94-13.37)1.166.17 (.16-34.36) Brain and other nervous system21.761.14 (.14-4.11)0.260 (0-13.99)2.862.34 (.28-8.44)0.510 (0-7.23)0.130 (0-28.57) Lymphoma33.5.86 (.18-2.5)0.540 (0-6.79)11.71.58 (.01-3.25)2.992.01 (.24-7.27)0.250 (0-14.66)  Myeloma21.931.04 (.13-3.75)0.290 (0-12.63)1.931.07 (.03-5.97)1.551.81 (.05-10.08)0.150 (0-24.97) Leukemia33.89.77 (.16-2.25)00.60 (0-6.15)21.91.05 (.13-3.8)11.11.9 (.02-5.02)0.280 (0-13.08) Miscellaneous malignant cancer286.794.12a (2.74-5.96)71.056.64a (2.67-13.67)133.323.91a (2.08-6.69)41.922.08 (.57-5.32)4.498.13#(2.21-20.81)Non-cancer causes Diseases of heart172103.441.66a (1.42-1.93)2916.311.78a (1.19-2.55)9150.311.81a (1.46-2.22)4029.081.38 (.98-1.87)127.741.55 (.8-2.71) COPD and allied cond3823.291.63a (1.15-2.24)83.592.23 (.96-4.4)2011.321.77a (1.08-2.73)96.651.35 (.62-2.57)11.74.57 (.01-3.2) Cerebrovascular diseases3319.341.71#(1.17-2.4)73.042.31 (.93-4.75)109.331.07 (.51-1.97)95.461.65 (.75-3.13)71.514.63#(1.86-9.54) Diabetes mellitus2611.322.30a (1.5-3.37)61.723.49#(1.28-7.6)75.471.28 (.51-2.64)113.253.38a (1.69-6.05)2.882.28 (.28-8.24) Nephritis, nephrotic syndrome and nephrosis168.251.94a (1.11-3.15)31.282.34 (.48-6.83)1142.75a (1.37-4.92)12.33.43 (.01-2.39)1.631.58 (.04-8.79) Pneumonia and influenza149.331.5 (.82-2.52)51.53.34a (1.08-7.79)64.541.32 (.49-2.88)32.61.16 (.24-3.38)00.70 (0-5.3) Other infectious and parasitic diseases including HIV133.134.16a (2.21-7.11)5.4910.17a (3.3-23.74)31.551.93 (.4-5.64)5.885.71a (1.85-13.33)0.210 (0-17.77) Accidents and adverse effects1112.11.91 (.45-1.63)21.821.1 (.13-3.96)45.84.69 (.19-1.75)33.52.85 (.18-2.49)2.932.16 (.26-7.79) Hypertension without heart disease104.222.37#(1.14-4.36)2.623.22 (.39-11.62)11.99.5 (.01-2.8)51.244.04a (1.31-9.42)2.365.52 (.67-19.96) Chronic liver disease and cirrhosis93.82.37a (1.08-4.49)2.573.53 (.43-12.76)41.852.16 (.59-5.53)21.111.8 (.22-6.49)1.273.65 (.09-20.36)AbbreviationsSMR: standardized mortality ratio; CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease.aStatistical significance with P < .05.Figure 2.Mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients diagnosed with localized, regional and distant metastasis penile cancer. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.Figure 3.The percentages of main causes of death in penile cancer patients. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.\nMain causes of death for patients with localized penile cancer.\nAbbreviationsSMR: standardized mortality ratio; CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease.\naStatistical significance with P < .05.\nMortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients diagnosed with localized, regional and distant metastasis penile cancer. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.\nThe percentages of main causes of death in penile cancer patients. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.\n[SUBTITLE] Causes of Death for Patients With Regional Penile Cancer [SUBSECTION] For patients with regional PeCa, 348 (51.3%) of all 679 deaths were due to PeCa itself. There were 118 SMTs deaths and 213 non-cancer deaths, accounting for 17.4% and 31.4% of all deaths. For the PeCa deaths, 89.1% of them occurred within 3 years after diagnosis. With the low PeCa mortality rate of the general population, the SMR of PeCa death reached a terribly high level. The main SMTs were lung and bronchus cancer [n = 23, SMR: 2.41 (1.53-3.62)]. The mortality risks of lung and bronchus cancer, and skin tumor [SMR: 6.41 (2.35-13.95)] were significantly increased. The most common non-cancer cause of death was diseases of heart [n = 71, SMR: 1.77 (1.38-2.23)], COPD and allied cond [n = 17, SMR: 1.85 (1.08-2.95)], diabetes mellitus [n = 16, SMR: 3.62 (2.07-5.88)] and cerebrovascular diseases [n = 13, SMR: 1.77 (.94-3.03)]. The mortality risks of diseases of heart, COPD and allied cond, diabetes mellitus, nephritis, nephrotic syndrome and nephrosis [n = 9, SMR: 2.88 (1.32-5.47)] and septicemia [n = 8, SMR: 3.83 (1.65-7.54)] were significantly increased when compared with the general population. All these results are shown in Table 3. The mortality rate of patients after diagnosis and is shown in Figure 2B and the percentages of the main causes of death is revealed in Figure 3B.Table 3.Main causes of death for patients with regional penile cancer.Causes of deathTotal<1 year1-5 years>5 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death679147.44.61a (4.27-4.97)27527.559.98a (8.84-11.23)31067.934.56a (4.07-5.1)9451.921.81a (1.46-2.22)All malignant cancers46634.6513.45a (12.26-14.73)2136.5632.48a (28.27-37.15)22516.4213.71a (11.97-15.62)2811.672.40a (1.59-3.47) Male genital system3624.0788.89a (79.97-98.54)179.79225.26a (193.47-260.79)1681.8889.54a (76.51-104.15)151.410.70a (5.99-17.65)  Penis348.0311 020.00a (9892.46-12,240.84)173.0129 605.21a (25 357.89-34 360.42)162.0111 182.19a (9526.53-13042.87)13.011155.76a (615.39-1976.38)  Prostate84.021.99 (.86-3.92)2.782.55 (.31-9.21)41.852.16 (.59-5.53)21.381.45 (.18-5.22)  Testis2.01149.35a (18.09-539.5)10370.55a (9.38-2064.6)1.01152.52a (3.86-849.81)000 (0-891.78) Urinary system62.512.39 (.88-5.21)1.462.18 (.06-12.16)41.153.47 (.94-8.87)1.891.12 (.03-6.23)  Kidney and renal Pelvis3.953.17 (.65-9.27)0.180 (0-20.85)2.454.46 (.54-16.13)1.323.12 (.08-17.36)  Urinary bladder21.51.34 (.16-4.83)1.273.71 (.09-20.65)1.681.48 (.04-8.23)0.550 (0-6.72) Digestive system78.97.78 (.31-1.61)11.69.59 (.01-3.3)44.27.94 (.26-2.4)23.01.66 (.08-2.4)  Colon and rectum32.951.02 (.21-2.97)0.570 (0-6.44)21.411.41 (.17-5.11)1.971.03 (.03-5.77)  Esophagus21.211.65 (.2-5.95)0.230 (0-16.24)1.581.72 (.04-9.57)1.412.47 (.06-13.75) Respiratory system239.92.32a (1.47-3.49)41.912.1 (.57-5.37)134.82.71a (1.44-4.63)63.191.88 (.69-4.09)  Lung and bronchus239.532.41a (1.53-3.62)41.842.18 (.59-5.58)134.622.81a (1.5-4.81)63.071.95 (.72-4.25) Skin excluding basal and squamous6.946.41a (2.35-13.95)2.1711.79a (1.43-42.6)4.439.23a (2.51-23.62)0.330 (0-11.07) Lymphoma21.411.42 (.17-5.14)0.260 (0-14.17)2.653.05 (.37-11.03)0.490 (0-7.53) Leukemia21.561.28 (.16-4.63)0.290 (0-12.84)2.722.76 (.33-9.97)0.550 (0-6.74) Miscellaneous malignant cancer512.7218.75a (13.96-24.66)25.5148.95a (31.68-72.26)241.2818.76a (12.02-27.92)2.932.15 (.26-7.77)Non-cancer causes Diseases of heart7140.111.77a (1.38-2.23)207.572.64#(1.61-4.08)3218.451.73a (1.19-2.45)1914.091.35 (.81-2.11) COPD and allied cond179.211.85a (1.08-2.95)41.692.37 (.65-6.08)84.241.89 (.82-3.72)53.291.52 (.49-3.55) Diabetes mellitus164.423.62a (2.07-5.88)5.836.00a (1.95-14.01)62.072.89a (1.06-6.3)51.513.31a (1.08-7.73) Cerebrovascular diseases137.341.77 (.94-3.03)31.392.16 (.44-6.3)63.341.79 (.66-3.91)42.611.53 (.42-3.92) Nephritis, nephrotic syndrome and nephrosis93.132.88a (1.32-5.47)3.595.07a (1.05-14.81)51.443.48a (1.13-8.12)11.1.91 (.02-5.08) Septicemia82.093.83a (1.65-7.54)50.412.66a (4.11-29.53)1.961.04 (.03-5.77)2.732.74 (.33-9.89) Alzheimers84.791.67 (.72-3.29)3.843.56 (.74-10.42)22.02.99 (.12-3.57)31.931.56 (.32-4.55) Chronic liver disease and cirrhosis41.622.47 (.67-6.32)3.319.67a (1.99-28.25)0.790 (0-4.64)1.521.94 (.05-10.8) Accidents and adverse effects64.841.24 (.45-2.7)0.910 (0-4.04)32.261.33 (.27-3.88)31.671.8 (.37-5.25) Suicide and self-inflicted injury41.382.9 (.79-7.42)1.273.71 (.09-20.68)1.671.49 (.04-8.28)2.444.57 (.55-16.51)CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease; SMR: standardized mortality ratio.aStatistical significance with P < .05.\nMain causes of death for patients with regional penile cancer.\nCI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease; SMR: standardized mortality ratio.\naStatistical significance with P < .05.\nFor patients with regional PeCa, 348 (51.3%) of all 679 deaths were due to PeCa itself. There were 118 SMTs deaths and 213 non-cancer deaths, accounting for 17.4% and 31.4% of all deaths. For the PeCa deaths, 89.1% of them occurred within 3 years after diagnosis. With the low PeCa mortality rate of the general population, the SMR of PeCa death reached a terribly high level. The main SMTs were lung and bronchus cancer [n = 23, SMR: 2.41 (1.53-3.62)]. The mortality risks of lung and bronchus cancer, and skin tumor [SMR: 6.41 (2.35-13.95)] were significantly increased. The most common non-cancer cause of death was diseases of heart [n = 71, SMR: 1.77 (1.38-2.23)], COPD and allied cond [n = 17, SMR: 1.85 (1.08-2.95)], diabetes mellitus [n = 16, SMR: 3.62 (2.07-5.88)] and cerebrovascular diseases [n = 13, SMR: 1.77 (.94-3.03)]. The mortality risks of diseases of heart, COPD and allied cond, diabetes mellitus, nephritis, nephrotic syndrome and nephrosis [n = 9, SMR: 2.88 (1.32-5.47)] and septicemia [n = 8, SMR: 3.83 (1.65-7.54)] were significantly increased when compared with the general population. All these results are shown in Table 3. The mortality rate of patients after diagnosis and is shown in Figure 2B and the percentages of the main causes of death is revealed in Figure 3B.Table 3.Main causes of death for patients with regional penile cancer.Causes of deathTotal<1 year1-5 years>5 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death679147.44.61a (4.27-4.97)27527.559.98a (8.84-11.23)31067.934.56a (4.07-5.1)9451.921.81a (1.46-2.22)All malignant cancers46634.6513.45a (12.26-14.73)2136.5632.48a (28.27-37.15)22516.4213.71a (11.97-15.62)2811.672.40a (1.59-3.47) Male genital system3624.0788.89a (79.97-98.54)179.79225.26a (193.47-260.79)1681.8889.54a (76.51-104.15)151.410.70a (5.99-17.65)  Penis348.0311 020.00a (9892.46-12,240.84)173.0129 605.21a (25 357.89-34 360.42)162.0111 182.19a (9526.53-13042.87)13.011155.76a (615.39-1976.38)  Prostate84.021.99 (.86-3.92)2.782.55 (.31-9.21)41.852.16 (.59-5.53)21.381.45 (.18-5.22)  Testis2.01149.35a (18.09-539.5)10370.55a (9.38-2064.6)1.01152.52a (3.86-849.81)000 (0-891.78) Urinary system62.512.39 (.88-5.21)1.462.18 (.06-12.16)41.153.47 (.94-8.87)1.891.12 (.03-6.23)  Kidney and renal Pelvis3.953.17 (.65-9.27)0.180 (0-20.85)2.454.46 (.54-16.13)1.323.12 (.08-17.36)  Urinary bladder21.51.34 (.16-4.83)1.273.71 (.09-20.65)1.681.48 (.04-8.23)0.550 (0-6.72) Digestive system78.97.78 (.31-1.61)11.69.59 (.01-3.3)44.27.94 (.26-2.4)23.01.66 (.08-2.4)  Colon and rectum32.951.02 (.21-2.97)0.570 (0-6.44)21.411.41 (.17-5.11)1.971.03 (.03-5.77)  Esophagus21.211.65 (.2-5.95)0.230 (0-16.24)1.581.72 (.04-9.57)1.412.47 (.06-13.75) Respiratory system239.92.32a (1.47-3.49)41.912.1 (.57-5.37)134.82.71a (1.44-4.63)63.191.88 (.69-4.09)  Lung and bronchus239.532.41a (1.53-3.62)41.842.18 (.59-5.58)134.622.81a (1.5-4.81)63.071.95 (.72-4.25) Skin excluding basal and squamous6.946.41a (2.35-13.95)2.1711.79a (1.43-42.6)4.439.23a (2.51-23.62)0.330 (0-11.07) Lymphoma21.411.42 (.17-5.14)0.260 (0-14.17)2.653.05 (.37-11.03)0.490 (0-7.53) Leukemia21.561.28 (.16-4.63)0.290 (0-12.84)2.722.76 (.33-9.97)0.550 (0-6.74) Miscellaneous malignant cancer512.7218.75a (13.96-24.66)25.5148.95a (31.68-72.26)241.2818.76a (12.02-27.92)2.932.15 (.26-7.77)Non-cancer causes Diseases of heart7140.111.77a (1.38-2.23)207.572.64#(1.61-4.08)3218.451.73a (1.19-2.45)1914.091.35 (.81-2.11) COPD and allied cond179.211.85a (1.08-2.95)41.692.37 (.65-6.08)84.241.89 (.82-3.72)53.291.52 (.49-3.55) Diabetes mellitus164.423.62a (2.07-5.88)5.836.00a (1.95-14.01)62.072.89a (1.06-6.3)51.513.31a (1.08-7.73) Cerebrovascular diseases137.341.77 (.94-3.03)31.392.16 (.44-6.3)63.341.79 (.66-3.91)42.611.53 (.42-3.92) Nephritis, nephrotic syndrome and nephrosis93.132.88a (1.32-5.47)3.595.07a (1.05-14.81)51.443.48a (1.13-8.12)11.1.91 (.02-5.08) Septicemia82.093.83a (1.65-7.54)50.412.66a (4.11-29.53)1.961.04 (.03-5.77)2.732.74 (.33-9.89) Alzheimers84.791.67 (.72-3.29)3.843.56 (.74-10.42)22.02.99 (.12-3.57)31.931.56 (.32-4.55) Chronic liver disease and cirrhosis41.622.47 (.67-6.32)3.319.67a (1.99-28.25)0.790 (0-4.64)1.521.94 (.05-10.8) Accidents and adverse effects64.841.24 (.45-2.7)0.910 (0-4.04)32.261.33 (.27-3.88)31.671.8 (.37-5.25) Suicide and self-inflicted injury41.382.9 (.79-7.42)1.273.71 (.09-20.68)1.671.49 (.04-8.28)2.444.57 (.55-16.51)CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease; SMR: standardized mortality ratio.aStatistical significance with P < .05.\nMain causes of death for patients with regional penile cancer.\nCI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease; SMR: standardized mortality ratio.\naStatistical significance with P < .05.\n[SUBTITLE] Causes of Death for Patients With Distant Metastatic Penile Cancer [SUBSECTION] For the included 132 patients with distant metastasis PeCa, 109 of them died during the follow-up with a significantly increased SMR of 27.68 (22.73-33.39). 100 deaths (91.7%) were due to malignant cancers including 76 cases (69.7%) dead of PeCa. 67.1% (n = 51) of all PeCa deaths occurred within 1 year and 98.7% (n = 75) dead within 3 years after diagnosis. There were 24 patients (22%) who died for SMTs, mainly including cancers from 4 lung and bronchus, 2 prostate, and 2 skin, 2 lymphoma and 3 other urinary organs. 19 (79.2%) of SMTs deaths occurred within 1 year of the diagnosis of PeCa. As for the non-cancer deaths, only 9 cases of deaths were observed, including 5 deaths from diseases of heart, 1 septicemia death, 1 other infection and parasitic diseases deaths, and 2 other causes of death. These results are presented in Table 4 and Figure 3C. The mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients after diagnosis is shown in Figure 2C.Table 4.Main Causes of Death for Patients With Distant Penile Cancer.Causes of deathTotal<1 year1-3 years>3 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death1093.9427.68a (22.73-33.39)741.8540.10a (31.49-50.34)281.0925.61a (17.02-37.01)717.00a (2.82-14.43)All malignant cancers100.98102.30a (83.23-124.42)70.45154.76a (120.65-195.54)27.2992.21a (60.76-134.16)3.2312.91a (2.66-37.72) Respiratory system4.2814.38a (3.92-36.81)3.1322.93a (4.73-67.01)0.090 (0-42.56)1.0616.48 (.42-91.8)  Lung and bronchus4.2714.98a (4.08-38.34)3.1323.85a (4.92-69.69)0.080 (0-44.28)1.0617.24 (.44-96.06) Soft tissue including heart2.01331.85a (40.19-1198.77)10351.01a (8.89-1955.69)10554.76a (14.05-3090.9)000 (0-2682.36) Skin excluding basal and squamous2.0285.91a (10.4-310.32)1.0187.34a (2.21-486.61)1.01146.53a (3.71-816.42)0.010 (0-736.74)  Male genital system80.12650.98a (516.18-810.2)55.051065.43a (802.63-1386.8)24.03713.33a (457.04-1061.38)1.0426.58 (.67-148.08)  Prostate2.1216.48a (2-59.52)2.0539.26a (4.75-141.82)0.030 (0-111.09)0.040 (0-99.09)  Penis76084 751.04a (66 774.18-106078.56)510126 908.70a (94 491.86-166861.5)24092 697.90a (59 393.29-137,927.13)104237.74a (107.29-23,611.17) Urinary system3.0646.59a (9.61-136.15)2.0364.45a (7.81-232.82)0.020 (0-199.1)1.0167.40a (1.71-375.54) Miscellaneous malignant cancer7.0793.72a (37.68-193.1)7.04198.90a (79.97-409.8)0.020 (0-166.27)0.020 (0-213.09)Non-cancer causes Septicemia1.0616.57 (.42-92.3)1.0337.63 (.95-209.66)0.020 (0-218.26)0.020 (0-218.46) Other infectious and parasitic diseases including HIV1.0426.85 (.68-149.58)1.0257.29a (1.45-319.22)0.010 (0-340.04)0.010 (0-412.29) Diseases of heart51.054.75a (1.54-11.09)1.492.02 (.05-11.26)1.283.52 (.09-19.64)3.2710.95a (2.26-32) Other cause of death2.573.53 (.43-12.75)1.273.69 (.09-20.57)0.150 (0-25.01)1.156.75 (.17-37.6)SMR: standardized mortality ratio; CI: confidence interval.aStatistical significance with P < .05.\nMain Causes of Death for Patients With Distant Penile Cancer.\nSMR: standardized mortality ratio; CI: confidence interval.\naStatistical significance with P < .05.\nFor the included 132 patients with distant metastasis PeCa, 109 of them died during the follow-up with a significantly increased SMR of 27.68 (22.73-33.39). 100 deaths (91.7%) were due to malignant cancers including 76 cases (69.7%) dead of PeCa. 67.1% (n = 51) of all PeCa deaths occurred within 1 year and 98.7% (n = 75) dead within 3 years after diagnosis. There were 24 patients (22%) who died for SMTs, mainly including cancers from 4 lung and bronchus, 2 prostate, and 2 skin, 2 lymphoma and 3 other urinary organs. 19 (79.2%) of SMTs deaths occurred within 1 year of the diagnosis of PeCa. As for the non-cancer deaths, only 9 cases of deaths were observed, including 5 deaths from diseases of heart, 1 septicemia death, 1 other infection and parasitic diseases deaths, and 2 other causes of death. These results are presented in Table 4 and Figure 3C. The mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients after diagnosis is shown in Figure 2C.Table 4.Main Causes of Death for Patients With Distant Penile Cancer.Causes of deathTotal<1 year1-3 years>3 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death1093.9427.68a (22.73-33.39)741.8540.10a (31.49-50.34)281.0925.61a (17.02-37.01)717.00a (2.82-14.43)All malignant cancers100.98102.30a (83.23-124.42)70.45154.76a (120.65-195.54)27.2992.21a (60.76-134.16)3.2312.91a (2.66-37.72) Respiratory system4.2814.38a (3.92-36.81)3.1322.93a (4.73-67.01)0.090 (0-42.56)1.0616.48 (.42-91.8)  Lung and bronchus4.2714.98a (4.08-38.34)3.1323.85a (4.92-69.69)0.080 (0-44.28)1.0617.24 (.44-96.06) Soft tissue including heart2.01331.85a (40.19-1198.77)10351.01a (8.89-1955.69)10554.76a (14.05-3090.9)000 (0-2682.36) Skin excluding basal and squamous2.0285.91a (10.4-310.32)1.0187.34a (2.21-486.61)1.01146.53a (3.71-816.42)0.010 (0-736.74)  Male genital system80.12650.98a (516.18-810.2)55.051065.43a (802.63-1386.8)24.03713.33a (457.04-1061.38)1.0426.58 (.67-148.08)  Prostate2.1216.48a (2-59.52)2.0539.26a (4.75-141.82)0.030 (0-111.09)0.040 (0-99.09)  Penis76084 751.04a (66 774.18-106078.56)510126 908.70a (94 491.86-166861.5)24092 697.90a (59 393.29-137,927.13)104237.74a (107.29-23,611.17) Urinary system3.0646.59a (9.61-136.15)2.0364.45a (7.81-232.82)0.020 (0-199.1)1.0167.40a (1.71-375.54) Miscellaneous malignant cancer7.0793.72a (37.68-193.1)7.04198.90a (79.97-409.8)0.020 (0-166.27)0.020 (0-213.09)Non-cancer causes Septicemia1.0616.57 (.42-92.3)1.0337.63 (.95-209.66)0.020 (0-218.26)0.020 (0-218.46) Other infectious and parasitic diseases including HIV1.0426.85 (.68-149.58)1.0257.29a (1.45-319.22)0.010 (0-340.04)0.010 (0-412.29) Diseases of heart51.054.75a (1.54-11.09)1.492.02 (.05-11.26)1.283.52 (.09-19.64)3.2710.95a (2.26-32) Other cause of death2.573.53 (.43-12.75)1.273.69 (.09-20.57)0.150 (0-25.01)1.156.75 (.17-37.6)SMR: standardized mortality ratio; CI: confidence interval.aStatistical significance with P < .05.\nMain Causes of Death for Patients With Distant Penile Cancer.\nSMR: standardized mortality ratio; CI: confidence interval.\naStatistical significance with P < .05.", "Among the 800 cases of death, 199 (24.9%) died for PeCa, 144 (18.0%) died for other tumor causes, and the rest 457 (57.1%) deaths were due to non-tumor causes. 75.9% of all PeCa deaths were within 3 years after diagnosis. The mortality rate of the general population who died for PeCa was rather low, which led to the extremely high SMR from the ratio of observed and expected. The majority of SMTs deaths (n = 106, 73.6%) occurred with 5 years after the diagnosis of PeCa. The main SMTs were lung and bronchus cancer [n = 40, SMR: 1.71 (1.22-2.33)], all cancers of digestive system [n = 25, SMR: 1.12 (.72-1.65)]. The main non-cancer causes of death were diseases of heart [n = 172, SMR: 1.66 (1.42-1.93)], chronic obstructive pulmonary disease (COPD) and allied cond [n = 38, SMR: 1.63 (1.15-2.24)], cerebrovascular diseases [n = 33, 22% of all deaths, SMR: 1.71 (1.17-2.4)] and diabetes mellitus [n = 26, SMR: 2.30 (1.5-3.37)]. The mortality risks of lung and bronchus cancer, skin tumor, diseases of heart, COPD and allied cond, cerebrovascular diseases, diabetes mellitus, nephritis, nephrotic syndrome and nephrosis, other infectious and parasitic diseases, hypertension, chronic liver disease and cirrhosis and atherosclerosis were significantly increased when compared with the general population. All these results are shown in Table 2. The mortality rate of all causes of death, PeCa, SMTs and non-tumor causes of patients after diagnosis is shown in Figure 2A and the percentage composition of the main causes of death is presented in Figure 3A.Table 2.Main causes of death for patients with localized penile cancer.Causes of deathTotal<1 year<1-5 year5-10 years>10 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death800377.822.12a (1.97-2.27)16758.342.86a (2.44-3.33)422183.122.30a (2.09-2.54)167107.681.55a (1.32-1.8)4428.681.53a (1.11-2.06)All malignant cancers34386.53.97a (3.56-4.41)8013.455.95a (4.72-7.4)20442.354.82a (4.18-5.53)4824.481.96a (1.45-2.6)116.221.77 (.88-3.16) Male genital system21310.7219.86a (17.29-22.72)581.7133.97a (25.79-43.91)1295.224.82a (20.72-29.49)252.998.37a (5.42-12.36)1.831.2 (.03-6.7)  Penis199.082495.58a (2160.87-2867.44)53.014477.48a (3353.94-5856.66)125.043293.53a (2741.5-3924.09)21.02898.28a (556.05-1373.11)0.010 (0-561.23)  Prostate910.59.85 (.39-1.61)21.691.18 (.14-4.28)25.14.39 (.05-1.41)42.951.36 (.37-3.47)1.821.22 (.03-6.78) Urinary system86.291.27 (.55-2.51)3.963.14 (.65-9.16)43.051.31 (.36-3.36)11.81.55 (.01-3.08)0.470 (0-7.81)  Urinary bladder43.81.05 (.29-2.7)2.583.47 (.42-12.54)11.83.55 (.01-3.04)11.1.91 (.02-5.09)0.290 (0-12.7)  Kidney and renal Pelvis12.33.43 (.01-2.39)1.362.81 (.07-15.63)01.140 (0-3.24)0.670 (0-5.54)0.170 (0-21.87)  Other urinary organs3.0933.47a (6.9-97.82)0.010 (0-278.54)3.0469.99a (14.43-204.54)0.030 (0-140.81)0.010 (0-503.78) Digestive system2522.341.12 (.72-1.65)33.41.88 (.18-2.57)1810.891.65 (.98-2.61)36.4.47 (.1-1.37)11.63.61 (.02-3.42)  Liver and intrahepatic bile duct83.712.16 (.93-4.25)0.530 (0-6.92)71.783.93a (1.58-8.09)11.11.9 (.02-5.03)0.280 (0-13.01)  Pancreas75.471.28 (.51-2.63)1.821.22 (.03-6.82)52.651.89 (.61-4.4)11.59.63 (.02-3.5)0.420 (0-8.88)  Esophagus42.931.37 (.37-3.5)0.450 (0-8.21)31.442.08 (.43-6.09)0.840 (0-4.41)1.214.84 (.12-26.98)  Colon and rectum37.41.4 (.08-1.18)21.181.7 (.21-6.13)03.650 (0-1.01)12.06.48 (.01-2.7)0.520 (0-7.13)  Stomach21.831.1 (.13-3.96)0.290 (0-12.81)20.92.23 (.27-8.06)0.510 (0-7.2)0.130 (0-28.41)  Intrahepatic bile duct2.762.63 (.32-9.49)0.110 (0-34.14)1.362.77 (.07-15.41)1.234.36 (.11-24.32)0.060 (0-59.14) Respiratory system4324.311.77a (1.28-2.38)73.871.81 (.73-3.73)2512.072.07a (1.34-3.06)86.761.18 (.51-2.33)31.621.85 (.38-5.41)  Lung and bronchus4023.391.71a (1.22-2.33)73.721.88 (.76-3.87)2411.622.07a (1.32-3.07)66.5.92 (.34-2.01)3z1.93 (.4-5.64) Skin excluding basal and squamous112.284.82a (2.41-8.63)1.352.88 (.07-16.03)61.125.38a (1.97-11.7)3.664.57 (.94-13.37)1.166.17 (.16-34.36) Brain and other nervous system21.761.14 (.14-4.11)0.260 (0-13.99)2.862.34 (.28-8.44)0.510 (0-7.23)0.130 (0-28.57) Lymphoma33.5.86 (.18-2.5)0.540 (0-6.79)11.71.58 (.01-3.25)2.992.01 (.24-7.27)0.250 (0-14.66)  Myeloma21.931.04 (.13-3.75)0.290 (0-12.63)1.931.07 (.03-5.97)1.551.81 (.05-10.08)0.150 (0-24.97) Leukemia33.89.77 (.16-2.25)00.60 (0-6.15)21.91.05 (.13-3.8)11.11.9 (.02-5.02)0.280 (0-13.08) Miscellaneous malignant cancer286.794.12a (2.74-5.96)71.056.64a (2.67-13.67)133.323.91a (2.08-6.69)41.922.08 (.57-5.32)4.498.13#(2.21-20.81)Non-cancer causes Diseases of heart172103.441.66a (1.42-1.93)2916.311.78a (1.19-2.55)9150.311.81a (1.46-2.22)4029.081.38 (.98-1.87)127.741.55 (.8-2.71) COPD and allied cond3823.291.63a (1.15-2.24)83.592.23 (.96-4.4)2011.321.77a (1.08-2.73)96.651.35 (.62-2.57)11.74.57 (.01-3.2) Cerebrovascular diseases3319.341.71#(1.17-2.4)73.042.31 (.93-4.75)109.331.07 (.51-1.97)95.461.65 (.75-3.13)71.514.63#(1.86-9.54) Diabetes mellitus2611.322.30a (1.5-3.37)61.723.49#(1.28-7.6)75.471.28 (.51-2.64)113.253.38a (1.69-6.05)2.882.28 (.28-8.24) Nephritis, nephrotic syndrome and nephrosis168.251.94a (1.11-3.15)31.282.34 (.48-6.83)1142.75a (1.37-4.92)12.33.43 (.01-2.39)1.631.58 (.04-8.79) Pneumonia and influenza149.331.5 (.82-2.52)51.53.34a (1.08-7.79)64.541.32 (.49-2.88)32.61.16 (.24-3.38)00.70 (0-5.3) Other infectious and parasitic diseases including HIV133.134.16a (2.21-7.11)5.4910.17a (3.3-23.74)31.551.93 (.4-5.64)5.885.71a (1.85-13.33)0.210 (0-17.77) Accidents and adverse effects1112.11.91 (.45-1.63)21.821.1 (.13-3.96)45.84.69 (.19-1.75)33.52.85 (.18-2.49)2.932.16 (.26-7.79) Hypertension without heart disease104.222.37#(1.14-4.36)2.623.22 (.39-11.62)11.99.5 (.01-2.8)51.244.04a (1.31-9.42)2.365.52 (.67-19.96) Chronic liver disease and cirrhosis93.82.37a (1.08-4.49)2.573.53 (.43-12.76)41.852.16 (.59-5.53)21.111.8 (.22-6.49)1.273.65 (.09-20.36)AbbreviationsSMR: standardized mortality ratio; CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease.aStatistical significance with P < .05.Figure 2.Mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients diagnosed with localized, regional and distant metastasis penile cancer. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.Figure 3.The percentages of main causes of death in penile cancer patients. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.\nMain causes of death for patients with localized penile cancer.\nAbbreviationsSMR: standardized mortality ratio; CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease.\naStatistical significance with P < .05.\nMortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients diagnosed with localized, regional and distant metastasis penile cancer. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.\nThe percentages of main causes of death in penile cancer patients. (A) Patients with localized penile cancer; (B) patients with regional penile cancer; (C) patients with distant metastasis penile cancer.", "For patients with regional PeCa, 348 (51.3%) of all 679 deaths were due to PeCa itself. There were 118 SMTs deaths and 213 non-cancer deaths, accounting for 17.4% and 31.4% of all deaths. For the PeCa deaths, 89.1% of them occurred within 3 years after diagnosis. With the low PeCa mortality rate of the general population, the SMR of PeCa death reached a terribly high level. The main SMTs were lung and bronchus cancer [n = 23, SMR: 2.41 (1.53-3.62)]. The mortality risks of lung and bronchus cancer, and skin tumor [SMR: 6.41 (2.35-13.95)] were significantly increased. The most common non-cancer cause of death was diseases of heart [n = 71, SMR: 1.77 (1.38-2.23)], COPD and allied cond [n = 17, SMR: 1.85 (1.08-2.95)], diabetes mellitus [n = 16, SMR: 3.62 (2.07-5.88)] and cerebrovascular diseases [n = 13, SMR: 1.77 (.94-3.03)]. The mortality risks of diseases of heart, COPD and allied cond, diabetes mellitus, nephritis, nephrotic syndrome and nephrosis [n = 9, SMR: 2.88 (1.32-5.47)] and septicemia [n = 8, SMR: 3.83 (1.65-7.54)] were significantly increased when compared with the general population. All these results are shown in Table 3. The mortality rate of patients after diagnosis and is shown in Figure 2B and the percentages of the main causes of death is revealed in Figure 3B.Table 3.Main causes of death for patients with regional penile cancer.Causes of deathTotal<1 year1-5 years>5 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death679147.44.61a (4.27-4.97)27527.559.98a (8.84-11.23)31067.934.56a (4.07-5.1)9451.921.81a (1.46-2.22)All malignant cancers46634.6513.45a (12.26-14.73)2136.5632.48a (28.27-37.15)22516.4213.71a (11.97-15.62)2811.672.40a (1.59-3.47) Male genital system3624.0788.89a (79.97-98.54)179.79225.26a (193.47-260.79)1681.8889.54a (76.51-104.15)151.410.70a (5.99-17.65)  Penis348.0311 020.00a (9892.46-12,240.84)173.0129 605.21a (25 357.89-34 360.42)162.0111 182.19a (9526.53-13042.87)13.011155.76a (615.39-1976.38)  Prostate84.021.99 (.86-3.92)2.782.55 (.31-9.21)41.852.16 (.59-5.53)21.381.45 (.18-5.22)  Testis2.01149.35a (18.09-539.5)10370.55a (9.38-2064.6)1.01152.52a (3.86-849.81)000 (0-891.78) Urinary system62.512.39 (.88-5.21)1.462.18 (.06-12.16)41.153.47 (.94-8.87)1.891.12 (.03-6.23)  Kidney and renal Pelvis3.953.17 (.65-9.27)0.180 (0-20.85)2.454.46 (.54-16.13)1.323.12 (.08-17.36)  Urinary bladder21.51.34 (.16-4.83)1.273.71 (.09-20.65)1.681.48 (.04-8.23)0.550 (0-6.72) Digestive system78.97.78 (.31-1.61)11.69.59 (.01-3.3)44.27.94 (.26-2.4)23.01.66 (.08-2.4)  Colon and rectum32.951.02 (.21-2.97)0.570 (0-6.44)21.411.41 (.17-5.11)1.971.03 (.03-5.77)  Esophagus21.211.65 (.2-5.95)0.230 (0-16.24)1.581.72 (.04-9.57)1.412.47 (.06-13.75) Respiratory system239.92.32a (1.47-3.49)41.912.1 (.57-5.37)134.82.71a (1.44-4.63)63.191.88 (.69-4.09)  Lung and bronchus239.532.41a (1.53-3.62)41.842.18 (.59-5.58)134.622.81a (1.5-4.81)63.071.95 (.72-4.25) Skin excluding basal and squamous6.946.41a (2.35-13.95)2.1711.79a (1.43-42.6)4.439.23a (2.51-23.62)0.330 (0-11.07) Lymphoma21.411.42 (.17-5.14)0.260 (0-14.17)2.653.05 (.37-11.03)0.490 (0-7.53) Leukemia21.561.28 (.16-4.63)0.290 (0-12.84)2.722.76 (.33-9.97)0.550 (0-6.74) Miscellaneous malignant cancer512.7218.75a (13.96-24.66)25.5148.95a (31.68-72.26)241.2818.76a (12.02-27.92)2.932.15 (.26-7.77)Non-cancer causes Diseases of heart7140.111.77a (1.38-2.23)207.572.64#(1.61-4.08)3218.451.73a (1.19-2.45)1914.091.35 (.81-2.11) COPD and allied cond179.211.85a (1.08-2.95)41.692.37 (.65-6.08)84.241.89 (.82-3.72)53.291.52 (.49-3.55) Diabetes mellitus164.423.62a (2.07-5.88)5.836.00a (1.95-14.01)62.072.89a (1.06-6.3)51.513.31a (1.08-7.73) Cerebrovascular diseases137.341.77 (.94-3.03)31.392.16 (.44-6.3)63.341.79 (.66-3.91)42.611.53 (.42-3.92) Nephritis, nephrotic syndrome and nephrosis93.132.88a (1.32-5.47)3.595.07a (1.05-14.81)51.443.48a (1.13-8.12)11.1.91 (.02-5.08) Septicemia82.093.83a (1.65-7.54)50.412.66a (4.11-29.53)1.961.04 (.03-5.77)2.732.74 (.33-9.89) Alzheimers84.791.67 (.72-3.29)3.843.56 (.74-10.42)22.02.99 (.12-3.57)31.931.56 (.32-4.55) Chronic liver disease and cirrhosis41.622.47 (.67-6.32)3.319.67a (1.99-28.25)0.790 (0-4.64)1.521.94 (.05-10.8) Accidents and adverse effects64.841.24 (.45-2.7)0.910 (0-4.04)32.261.33 (.27-3.88)31.671.8 (.37-5.25) Suicide and self-inflicted injury41.382.9 (.79-7.42)1.273.71 (.09-20.68)1.671.49 (.04-8.28)2.444.57 (.55-16.51)CI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease; SMR: standardized mortality ratio.aStatistical significance with P < .05.\nMain causes of death for patients with regional penile cancer.\nCI: confidence interval; COPD: Chronic Obstructive Pulmonary Disease; SMR: standardized mortality ratio.\naStatistical significance with P < .05.", "For the included 132 patients with distant metastasis PeCa, 109 of them died during the follow-up with a significantly increased SMR of 27.68 (22.73-33.39). 100 deaths (91.7%) were due to malignant cancers including 76 cases (69.7%) dead of PeCa. 67.1% (n = 51) of all PeCa deaths occurred within 1 year and 98.7% (n = 75) dead within 3 years after diagnosis. There were 24 patients (22%) who died for SMTs, mainly including cancers from 4 lung and bronchus, 2 prostate, and 2 skin, 2 lymphoma and 3 other urinary organs. 19 (79.2%) of SMTs deaths occurred within 1 year of the diagnosis of PeCa. As for the non-cancer deaths, only 9 cases of deaths were observed, including 5 deaths from diseases of heart, 1 septicemia death, 1 other infection and parasitic diseases deaths, and 2 other causes of death. These results are presented in Table 4 and Figure 3C. The mortality rate of all causes of death, PeCa, SMTs and non-tumor causes for patients after diagnosis is shown in Figure 2C.Table 4.Main Causes of Death for Patients With Distant Penile Cancer.Causes of deathTotal<1 year1-3 years>3 yearsObserved, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)Observed, nExpected, nSMR (95% CI)All causes of death1093.9427.68a (22.73-33.39)741.8540.10a (31.49-50.34)281.0925.61a (17.02-37.01)717.00a (2.82-14.43)All malignant cancers100.98102.30a (83.23-124.42)70.45154.76a (120.65-195.54)27.2992.21a (60.76-134.16)3.2312.91a (2.66-37.72) Respiratory system4.2814.38a (3.92-36.81)3.1322.93a (4.73-67.01)0.090 (0-42.56)1.0616.48 (.42-91.8)  Lung and bronchus4.2714.98a (4.08-38.34)3.1323.85a (4.92-69.69)0.080 (0-44.28)1.0617.24 (.44-96.06) Soft tissue including heart2.01331.85a (40.19-1198.77)10351.01a (8.89-1955.69)10554.76a (14.05-3090.9)000 (0-2682.36) Skin excluding basal and squamous2.0285.91a (10.4-310.32)1.0187.34a (2.21-486.61)1.01146.53a (3.71-816.42)0.010 (0-736.74)  Male genital system80.12650.98a (516.18-810.2)55.051065.43a (802.63-1386.8)24.03713.33a (457.04-1061.38)1.0426.58 (.67-148.08)  Prostate2.1216.48a (2-59.52)2.0539.26a (4.75-141.82)0.030 (0-111.09)0.040 (0-99.09)  Penis76084 751.04a (66 774.18-106078.56)510126 908.70a (94 491.86-166861.5)24092 697.90a (59 393.29-137,927.13)104237.74a (107.29-23,611.17) Urinary system3.0646.59a (9.61-136.15)2.0364.45a (7.81-232.82)0.020 (0-199.1)1.0167.40a (1.71-375.54) Miscellaneous malignant cancer7.0793.72a (37.68-193.1)7.04198.90a (79.97-409.8)0.020 (0-166.27)0.020 (0-213.09)Non-cancer causes Septicemia1.0616.57 (.42-92.3)1.0337.63 (.95-209.66)0.020 (0-218.26)0.020 (0-218.46) Other infectious and parasitic diseases including HIV1.0426.85 (.68-149.58)1.0257.29a (1.45-319.22)0.010 (0-340.04)0.010 (0-412.29) Diseases of heart51.054.75a (1.54-11.09)1.492.02 (.05-11.26)1.283.52 (.09-19.64)3.2710.95a (2.26-32) Other cause of death2.573.53 (.43-12.75)1.273.69 (.09-20.57)0.150 (0-25.01)1.156.75 (.17-37.6)SMR: standardized mortality ratio; CI: confidence interval.aStatistical significance with P < .05.\nMain Causes of Death for Patients With Distant Penile Cancer.\nSMR: standardized mortality ratio; CI: confidence interval.\naStatistical significance with P < .05.", "With the high and increasing survival rate of PeCa, deaths from other causes account for a very high proportion of total deaths and tend to increase.5 Therefore, other causes of death including SMTs and non-tumor diseases should be taken seriously. Besides, whether these causes have relationships with the diagnosis of PeCa is unclear. In current literature, studies on the SMTs for the primary PeCa were quite rare. Our study, as far as we can know, is the first study focusing on the SMTs and non-cancer causes of death for primary PeCa patients. We also relate these causes to the time period after diagnosis of penile cancer and provide results stratified by tumor stage and patient characteristics to provide valuable insights into the prognosis and management of penile cancer.\nAccording to the basic characteristic of dead patients, people diagnosed with penile cancer between 15 and 54 years had obviously higher SMRs than other age groups. As the number of deaths among 15-54 years old were not obviously higher than other age groups, this result was due to the lower excess risk of the general population. For the general population among 15-54 years, most of them have better physical conditions and a lower rate of all kinds of deaths when compared with the population with elderly age. We also found that the mortality risk continued to decrease with the year of diagnosis from 2.65 in 2004-2008 to 2.01 in 2014-2018 in localized disease. This may be the result of improved cancer treatment and disease management after diagnosis for PeCa over time. However, this result was reversed in regional and distant metastatic disease. White people accounted for a higher proportion of all deaths, but black people had a higher risk of death in localized and regional diseases. Previous studies reported that the mortality rate of blacks was higher than that of whites among American penile cancer patients.6 This may be due to earlier exposure of blacks to HPV through early sexual contact, leading to an earlier diagnosis of penile cancer.6,7\nFor localized and regional diseases, deaths from SMR accounted for a certain percentage of total deaths. A study identified 3641 PeCa patients in the Swedish Family-Cancer Database from 1958 to 2015. They found that SMTs occurred in 16.8% of primary PeCa patients and 45.9% of these patients dead for SMTs rather than PeCa.8 The main SMTs included prostate cancer, colorectum cancer, lung cancer, bladder cancer, skin squamous cell cancer and stomach cancer. These results were similar to ours. This study also reported that SMTs was associated with HPV and smoking.8 Another study with 1634 men with PeCa found a 2-3-fold increased risk of a second human papillomavirus (HPV) associated cancer of the oral cavity, oropharynx and anal canal among PeCa patients.9 For the occurrence of PeCa, there were some important risk factors of PeCa including lack of circumcision, smegma retention, chronic balanitis, lichen sclerosis, obesity, smoking, HPV infection and immune-compromised states.10,11 It may be the consequences of treatment for penile cancer and the more widely spread of HPV.12-14\nFor the non-tumor deaths, it accounted for a large proportion among all deaths in localized and regional stages. A previous study reported that 65.7% of deaths were due to non-cancer causes among PeCa patients without SMTs and only 28.4% were due to penile cancer.8 Cardiovascular disease is still a major cause of death in PeCa patients. Statistics in recent years from a study had shown that more than 20% of PeCa patients died from cardiovascular disease.15 It was also reported that the risk of cancer patients dying from cardiovascular diseases was inversely related to the age at diagnosis.15 For this kind of cancer with a good prognosis but still a high risk of cardiovascular death, patients may benefit from clinical intervention by a cardiologist at the time of diagnosis. There were some relationships between diabetes mellitus and PeCa. Previous studies reported that diabetes was associated with phimosis, which could increase the risk of penile cancer.16-18 Men with diabetes, mainly type 2 diabetes, were associated with an increased incidence of penile squamous cell carcinoma compared with men without diabetes.19 Localized and advanced PeCa and the associated treatments have profound physical and psychological effects on the survivors’ quality of life. Due to potentially crippling surgery, patients with penile cancer increased psychological stress and therefore have an increased need for psychosocial care.20-22 However, in our study, we didn’t find a significantly increased risk of suicide and self-inflicted injury.22\nFor patients with distant metastatic PeCa, the prognosis was poor with a mortality rate of 82.6%. The main cause of death in patients was the penile cancer tumor itself. Other causes of death only account for only a small fraction of all deaths. Therefore, active and effective treatment for metastatic penile cancer is still the most important measure to prevent deaths and prolong survival durations.\nOur study is the first to provide the most comprehensive comparison and description of the risk of death from all causes in PeCa patients using tumor stage and time after diagnosis. However, there were some limitations in our study. Firstly, we used Summary Stage 2000 (1998+) to distinguish local, regional and distant diseases, but these classifications maybe not the same in different periods. Therefore, the results of this study can’t fully represent the current staging results. Secondly, some important information of patients in this database was available, such as the basic illness, detailed treatments and socioeconomic status, etc. The information might have certain impacts on the survival and prognosis of patients. Our results might be influenced by the missing information. Thirdly, the sample size selection was not estimated by power calculations. It was also a limitation of our study.", "Our study provided a detailed analysis of the causes of death for patients with locally, regionally, and distant metastatic PeCa after diagnosis. This information could be useful for disease prevention and health care during patients’ survivorship.", "Click here for additional data file.\nSupplemental Material for Second Malignant Tumors and Non-Tumor Causes of Death for Patients With Penile Cancer During Their Survivorship by Pan Song, Xiaotian Wu, Luchen Yang, Kai Ma, Zhenghuan Liu, Jing Zhou, Junhao Chen, Qing Zhu, and Qiang Dong in Cancer Control" ]
[ "intro", "methods", null, null, null, null, "results", null, null, null, "discussion", "conclusion", "supplementary-material" ]
[ "penile cancer", "causes of death", "second malignant tumors", "non-tumor diseases", "standardized mortality ratio", "SEER" ]
Qualitative Analysis of the Roles of Physicians and Nurses in Providing Decision Support to Patients With Relapsed or Refractory Leukemia and Lymphoma.
36268680
This study examined the roles of hematologists and other professionals in providing decision support to patients with relapsed or refractory leukemia and lymphoma.
INTRODUCTION
This was a qualitative study using in-depth semi-structured interviews involving 11 hematologists in Japan.
METHODS
We identified 7 categories related to the roles of hematologists in providing direct decision support to patients: (1) preparing patients before informed consent, (2) selecting the information to convey, (3) choosing a method for conveying this information, (4) respecting the intentions of patients and their families, (5) directing decision-making and considering fairness, (6) considering the emotional aspects of patients and their families, and (7) providing support after discussing treatment options. We also identified the following 5 subcategories related to the roles of hematologists in multidisciplinary collaboration: (1) communicating with other professionals, (2) gathering information from them, (3) providing information to them, (4) managing the entire medical team, and (5) encouraging nurses to actively participate with patients throughout the decision-making process.
RESULT
Through content analysis, the hematologist's direct role in decision-making was extracted as preparation and consideration in situations where information about decision-making is communicated, and emotional support after the information is communicated. In addition, active participation in discussions, sharing information about the patient's situation and relevant discussions, and emotional support as the hematologist's expected roles in other professions were extracted. The results therefore suggest that a multidisciplinary team is needed to share information and provide multidimensional support to patients.
CONCLUSION
[ "Humans", "Communication", "Physicians", "Lymphoma", "Qualitative Research", "Leukemia", "Decision Making" ]
9597203
Introduction
The prognosis and illness trajectory of leukemia and lymphoma vary greatly depending on the degree of progression and subtype of the disease. However, irrespective of the illness trajectory, patients face the same issues and debates related to decision-making, such as how far to go with aggressive treatment during the relapse or treatment-resistant period and the choice of treatment and place of care.1-3 Even in relapse cases, such patients may have the option to receive high-dose chemotherapy treatments or hematopoietic cell transplantations aimed at remission and achieving long-term survival. Such treatment carries significant risks, including complications such as severe infections and bleeding due to bone marrow suppression and treatment-related death, and may result in transition from invasive treatment aimed at cure to end-of-life (EOL). Even when the disease is refractory to treatment, anticancer therapy aimed at disease control is often administered until the end of life. In such contexts, it is very difficult for patients and their families to make decisions about these complex treatment strategies and the high risks involved in the context of their own life values.4-6 The American Society of Clinical Oncology (ASCO) has published consensus guidelines for clinician-patient communication, specifically recommending that clinicians provide information that is congruent with their patients’ interests and intentions while jointly considering their goals for treatment.7 It is also notable that hematologists’ communication skills have been receiving increasing attention in hematology clinical practice in recent years. An ethnographic study of hematologists' communication patterns when communicating bad news revealed 4 characteristics: (1) technical-defensive patterns, (2) an authoritative pattern, (3) a relational-recursive pattern, and (4) a compassionate sharing pattern, and the paper also suggested that hematologists were not good at expressing caring and empathy.8 In contrast, a qualitative study examining the needs of patients with hematological malignancies when being told bad news reported identifying emotional support from their healthcare providers and information needs about future support systems. It was also noted that healthcare-team involvement is necessary to meet these patients' needs and support their decision-making.9 Therefore, this study sought to clarify the following: (a) the way in which hematologists perceive the roles of doctors in providing decision-making support to patients suffering from relapsed and refractory leukemia as well as lymphoma, (b) the expected roles of hematologists in supporting decision-making via multidisciplinary collaboration, and (c) the roles hematologists expect other healthcare providers to fulfill.
Methods
The purpose of this study was to determine hematologists’ role in decision support for patients with leukemia and lymphoma. Physicians at institutions with hematology wards and hematology outpatient clinics where chemotherapy and bone marrow transplantation are performed were included. The eligibility criteria for physicians included being Board Certified Hematologists and having at least 5 years of experience. Hematologists meeting these criteria were considered to have experience seeing patients with relapsed or refractory leukemia or lymphoma. Subjects were selected using opportunistic sampling, which is used in selecting those with common characteristics and the potential to provide a rich, appropriate, and diverse research agenda. Eligible physicians were asked to recommend a qualified physician who met the inclusion criteria to the head of the hematology department of a hospital in Tokyo. The recommended physicians were informed orally and in writing about the purpose and methods of the study and the voluntary participation clause. Although this is a qualitative study and it is difficult to theoretically calculate the sample size, we chose a sample size of 10 to 15 participants based on previous studies9-11 using the same content analysis method as in this study. The acceptance rate of recommended physicians was 100%. Verbal and written explanations of the research objectives and procedures were provided to all participants, and written consent was obtained. This study was approved by the Institutional Review Board of University of Tokyo (Approval No. 3443). The study was initiated at the University of Tokyo and completed at Kyoto University. To collect the data, we conducted in-depth interviews to identify the attributes and roles of doctors as well as the expected roles of nurses in providing decision-making support to patients with relapsed or refractory leukemia and lymphoma. Specifically, we scheduled face-to-face semi-structured interviews in private rooms between August and December 2011. All the interviews conducted were audio recorded and fully transcribed by 1 researcher (a research nurse), who also took detailed field notes during each interview. Physicians were first asked for their demographic details, including age, the type of facility where they worked, and the number of years they had worked in their area of specialization. We then asked them about their roles in treating patients suffering from relapsed or refractory leukemia and lymphoma. To analyze the data, interview data were transcribed verbatim from the audio recordings. Interview data were then numbered for each participant, separated from personal information, and anonymized. A content analysis was then conducted. First, 1 researcher extracted all statements related to the 3 study topics from each transcript (eg, the role of hematologists, the role of hematologists in multidisciplinary collaboration, and the expectations of other professionals in the context of providing decision-making support). We then carefully conceptualized the text and categorized it into content areas through content analysis using the Klaus Krippendorff12 method. The units, codes, and categories were decided through consensus. Next, another experienced qualitative researcher read the original interviews and classified them into categories and subcategories. The coder read the original interview text and independently determined the subcategory into which the content fell. The rate of agreement between the researchers and coders was 91%. Finally, the coder and the researcher discussed the units they disagreed on until they reached a consensus.
Results
[SUBTITLE] Participant Characteristics [SUBSECTION] We conducted semi-structured interviews involving 11 hematologists. The average participant age was 44 years (range: 35-54 years), whereas the average specialization experience was 16 years (range: 7-26 years). Additional details are listed in Table 1. The average length of the interviews was 27 minutes (range: 17-46 minutes).Table 1.Characteristics of doctors and interview details.HematologistsTotaln = 11Gender—Male10Female1Workplace—University hospital7Cancer center2General hospital2Age (years)43.9 (34-54)*Experience in specialty (years)16.1 (7-26)*∗Mean(Range). Characteristics of doctors and interview details. ∗Mean(Range). We conducted semi-structured interviews involving 11 hematologists. The average participant age was 44 years (range: 35-54 years), whereas the average specialization experience was 16 years (range: 7-26 years). Additional details are listed in Table 1. The average length of the interviews was 27 minutes (range: 17-46 minutes).Table 1.Characteristics of doctors and interview details.HematologistsTotaln = 11Gender—Male10Female1Workplace—University hospital7Cancer center2General hospital2Age (years)43.9 (34-54)*Experience in specialty (years)16.1 (7-26)*∗Mean(Range). Characteristics of doctors and interview details. ∗Mean(Range). [SUBTITLE] The Role of Hematologists in Direct Decision Support [SUBSECTION] As shown in Table 2, we identified 7 categories related to hematologists’ roles in providing direct decision support to patients: (1) preparing patients before informed consent, (2) selecting the information to convey, (3) choosing a method for conveying this information, (4) respecting the intentions of patients and their families, (5) directing decision-making and considering fairness, (6) considering the emotional aspects of patients and their families, and (7) providing support after discussing treatment options.Table 2.Role of hematologists in providing direct decision support (n = 11).CategorySubcategoryCoden1Preparing patients before IC*(1)Gathering preliminary information for explaining issues to patientsObtain information from family members to understand the patient’s preferences in advance3Check if home care is possible1Understand the family background1Talk to the family about what you will tell the patient1(2)Encouraging family members to participate in decision-makingSet up a place to provide information to patients, including family members3Discuss life-prolonging treatments with the patient’s family1(3)Creating an environment for concentrating on ICDiscuss in a private room2Take sufficient time1Leave the PHS*2 with the nurse so that the discussion is not disturbed1(4)Providing advance information on the risk of relapseWhen the patient reaches complete remission, explain the risk of relapse and prepare the patient for relapse12Selecting the information to convey(1)Choice of information to provide to patientsIn many cases, do not give the patient a prognosis9Depending on the patient’s medical condition, I (doctor) may not tell the truth5Modify the message according to the patient’s life stage3(2)Providing patients and families with information to help them make treatment decisionsCommunicate the advantages and disadvantages of treatment options9Tell patients and their families the information they need to make treatment decisions6Inform patients of all treatment options4Communicate not only the bad news, but also ways of improvement3Also convey the option of non-treatment3(3)Providing patients and families with information to help them understand the patients' current situationInform family members about the possibility of sudden changes in the medical condition and specific prognosis9Provide patients with information related to what they imagine will happen in the future5Provide patients with information that will help them understand their current medical condition4Provide patients with a vague idea of their prognosis3Communicate the prognosis to patients using concrete numbers23Choosing how to communicate information(1)Explanations based on the patients' stage of readinessJudge what I (doctor) say and how I (doctor) say it based on long-standing relationship with the patients5Provide information based on the patient’s reaction and acceptance of their medical condition5(2)Respect for the preferences of patients and their familiesProvide easy-to-understand explanations without using technical terms5Provide explanations so that patients/families do not develop misconceptions4Convey information carefully and clearly3Provide a written explanation2Divide bad news into components14Respect for the intentions of patients and families(1)Respect for the will of patients and familiesRespect and support the patient’s will and desired treatment6If the patient’s condition is severe, respect the views of the family and primary caregiver2(2)Setting goals in accordance with the preferences of the patients and familiesListen to the needs of patients and their families and determine actionable treatment goals3Explore the intentions of patients and their families through conversations1If there is no difference between the advantages and disadvantages of a treatment choice, the choice is purely made by the patient15Directing decision-making and considering fairness(1)Considering the direction and fairness of treatment options recommended by doctorsExplain treatment options with some direction while also providing options that the doctor finds beneficial8In some cases, offer personal opinion as a doctor2Dare to treat the patient with paternalism2(2)Consideration for patients to make impartial decisionsCommunicate with the knowledge that the doctor’s manner of speaking influences the patients’ decisions5Explain the opinions of other doctors and the second opinion system16Considering the emotional aspects of the patient and family(1)Explaining with consideration for emotionsTalk in a way that allows the patient to have hope5Convey bad news according to the protocol1Consider that patients do not have to listen to explanations while always looking at bad information1Divulge bad news in stages1(2)Showing a supportive attitude to the patientApproach the patient with a non-abandoning attitude2Show that the team supports the patient1(3)Attitude when facing patients and their familiesInteract with patients on an equal footing2Talk while looking into the patient’s eyes1Interact with the patient’s family on an equal footing17Supporting after discussing treatments(1)Emotional support after discussing treatmentsSupport fluctuations in the patient’s mind after discussing treatments2Call out the family and assess their reactions1*1IC = informed consent; *2PHS = Personal Handyphone System (A phone used by doctors to communicate with staff in the hospital). Role of hematologists in providing direct decision support (n = 11). *1IC = informed consent; *2PHS = Personal Handyphone System (A phone used by doctors to communicate with staff in the hospital). [SUBTITLE] Preparing Patients Before Informed Consent (IC) [SUBSECTION] This category comprised 4 subcategories. Several participants talked about preparing patients and families for IC discussions, including “Gathering preliminary information for explaining issues to patients” and “Encouraging family members to participate in decision-making.” As such, physicians were expected to prepare patients before IC discussions. For instance, they would provide patients with information regarding the risk of relapse. This category comprised 4 subcategories. Several participants talked about preparing patients and families for IC discussions, including “Gathering preliminary information for explaining issues to patients” and “Encouraging family members to participate in decision-making.” As such, physicians were expected to prepare patients before IC discussions. For instance, they would provide patients with information regarding the risk of relapse. [SUBTITLE] Selecting the Information To Convey [SUBSECTION] This category comprised 3 subcategories. In subcategory (1) “Choice of information to provide to patients,” more than half of the participants said that they did not provide their patients with prognoses. More importantly, more than half said they may not even tell their patients the truth, depending on the severity of the illness, to avoid a loss of hope; they were very selective about the information they chose to provide to their patients. In subcategory (2) “Providing patients and families with information to help them make treatment decisions,” more than half of the participants identified “Communicating the advantages and disadvantages of treatment options” and “Telling patients and their families the information they need to make treatment decisions.” They provided information they believed would help patients and their families fully understand all the options before making a final decision. In subcategory (3), “Providing patients and families with information to help them understand the patients’ current situation,” a large number said they would inform family members about the possibility of sudden changes and specific prognoses, with more than half saying they would explain the issues so that patients could visualize what may happen to them in the future. Generally, participants provided information to help patients and their families clearly understand their current situations. This category comprised 3 subcategories. In subcategory (1) “Choice of information to provide to patients,” more than half of the participants said that they did not provide their patients with prognoses. More importantly, more than half said they may not even tell their patients the truth, depending on the severity of the illness, to avoid a loss of hope; they were very selective about the information they chose to provide to their patients. In subcategory (2) “Providing patients and families with information to help them make treatment decisions,” more than half of the participants identified “Communicating the advantages and disadvantages of treatment options” and “Telling patients and their families the information they need to make treatment decisions.” They provided information they believed would help patients and their families fully understand all the options before making a final decision. In subcategory (3), “Providing patients and families with information to help them understand the patients’ current situation,” a large number said they would inform family members about the possibility of sudden changes and specific prognoses, with more than half saying they would explain the issues so that patients could visualize what may happen to them in the future. Generally, participants provided information to help patients and their families clearly understand their current situations. [SUBTITLE] Choosing the Way In Which To Communicate Information [SUBSECTION] This category comprised 2 subcategories. Nearly half of participants expressed the ideas of “I determine what I say and how I say it based on our long-standing relationship with the patients” and “Providing information based on the patient’s reaction and acceptance of their medical condition.” They carefully considered what they discussed and how they discussed it, especially considering their long-standing association with their patients. In this regard, they conveyed information based on how they thought patients would react and their level of acceptance. They also provided explanations according to what they believed patients intended and thought depending on their respective stages of readiness. Moreover, nearly half said that they would explain issues without using jargon, thereby ensuring that the information provided was easy to understand. Essentially, participants remained cognizant of the need to communicate both correctly and appropriately with patients and their families. This category comprised 2 subcategories. Nearly half of participants expressed the ideas of “I determine what I say and how I say it based on our long-standing relationship with the patients” and “Providing information based on the patient’s reaction and acceptance of their medical condition.” They carefully considered what they discussed and how they discussed it, especially considering their long-standing association with their patients. In this regard, they conveyed information based on how they thought patients would react and their level of acceptance. They also provided explanations according to what they believed patients intended and thought depending on their respective stages of readiness. Moreover, nearly half said that they would explain issues without using jargon, thereby ensuring that the information provided was easy to understand. Essentially, participants remained cognizant of the need to communicate both correctly and appropriately with patients and their families. [SUBTITLE] Respect for The Intentions Of Patients And Families [SUBSECTION] This category comprised 2 subcategories. In subcategory (1) “Respect for the will of patients and families,” more than half of the participants said they helped patients and families make decisions by supporting and respecting their wishes and desired treatments. In subcategory (2) “Setting goals in accordance with the preferences of the patients and families,” several participants said that they helped patients and families set feasible goals after listening to their wishes. This category comprised 2 subcategories. In subcategory (1) “Respect for the will of patients and families,” more than half of the participants said they helped patients and families make decisions by supporting and respecting their wishes and desired treatments. In subcategory (2) “Setting goals in accordance with the preferences of the patients and families,” several participants said that they helped patients and families set feasible goals after listening to their wishes. [SUBTITLE] Directing decision-making and considering fairness [SUBSECTION] This category comprised 2 subcategories. In subcategory (1) “Considering the direction and fairness of treatment options recommended by doctors,” more than half of the participants said they explain treatment options with some direction while also providing options they find most beneficial. They talked about giving weight to treatment options and facilitating the decision-making process. Furthermore, they attempted to present information in a fair manner, being aware that the way they talk can affect the way patients perceive their treatment, as shown in subcategory (2). This category comprised 2 subcategories. In subcategory (1) “Considering the direction and fairness of treatment options recommended by doctors,” more than half of the participants said they explain treatment options with some direction while also providing options they find most beneficial. They talked about giving weight to treatment options and facilitating the decision-making process. Furthermore, they attempted to present information in a fair manner, being aware that the way they talk can affect the way patients perceive their treatment, as shown in subcategory (2). [SUBTITLE] Considering the Emotional Aspects Of The Patient And Family [SUBSECTION] This category comprised 3 subcategories. In subcategory (1)“explaining with consideration for emotions,” nearly half of the participants said they would attempt to instill hope among patients, and hence, provide explanations considering the related emotional aspects. In subcategory (2) “showing a supportive attitude to the patient”, several participants also expressed adopting a supportive stance to reassure patients that they were not abandoning them or their families. In subcategory (3) “attitude when facing patients and their families”, several participants talked about the doctor’s attitude toward patients, such as speaking with patients and families as equals and looking them in the eye. This category comprised 3 subcategories. In subcategory (1)“explaining with consideration for emotions,” nearly half of the participants said they would attempt to instill hope among patients, and hence, provide explanations considering the related emotional aspects. In subcategory (2) “showing a supportive attitude to the patient”, several participants also expressed adopting a supportive stance to reassure patients that they were not abandoning them or their families. In subcategory (3) “attitude when facing patients and their families”, several participants talked about the doctor’s attitude toward patients, such as speaking with patients and families as equals and looking them in the eye. [SUBTITLE] Providing support after discussing treatments [SUBSECTION] Several participants said they informed patients about bad news and future treatments, after which they provided emotional support in consideration of any emotional variabilities. Several participants said they informed patients about bad news and future treatments, after which they provided emotional support in consideration of any emotional variabilities. As shown in Table 2, we identified 7 categories related to hematologists’ roles in providing direct decision support to patients: (1) preparing patients before informed consent, (2) selecting the information to convey, (3) choosing a method for conveying this information, (4) respecting the intentions of patients and their families, (5) directing decision-making and considering fairness, (6) considering the emotional aspects of patients and their families, and (7) providing support after discussing treatment options.Table 2.Role of hematologists in providing direct decision support (n = 11).CategorySubcategoryCoden1Preparing patients before IC*(1)Gathering preliminary information for explaining issues to patientsObtain information from family members to understand the patient’s preferences in advance3Check if home care is possible1Understand the family background1Talk to the family about what you will tell the patient1(2)Encouraging family members to participate in decision-makingSet up a place to provide information to patients, including family members3Discuss life-prolonging treatments with the patient’s family1(3)Creating an environment for concentrating on ICDiscuss in a private room2Take sufficient time1Leave the PHS*2 with the nurse so that the discussion is not disturbed1(4)Providing advance information on the risk of relapseWhen the patient reaches complete remission, explain the risk of relapse and prepare the patient for relapse12Selecting the information to convey(1)Choice of information to provide to patientsIn many cases, do not give the patient a prognosis9Depending on the patient’s medical condition, I (doctor) may not tell the truth5Modify the message according to the patient’s life stage3(2)Providing patients and families with information to help them make treatment decisionsCommunicate the advantages and disadvantages of treatment options9Tell patients and their families the information they need to make treatment decisions6Inform patients of all treatment options4Communicate not only the bad news, but also ways of improvement3Also convey the option of non-treatment3(3)Providing patients and families with information to help them understand the patients' current situationInform family members about the possibility of sudden changes in the medical condition and specific prognosis9Provide patients with information related to what they imagine will happen in the future5Provide patients with information that will help them understand their current medical condition4Provide patients with a vague idea of their prognosis3Communicate the prognosis to patients using concrete numbers23Choosing how to communicate information(1)Explanations based on the patients' stage of readinessJudge what I (doctor) say and how I (doctor) say it based on long-standing relationship with the patients5Provide information based on the patient’s reaction and acceptance of their medical condition5(2)Respect for the preferences of patients and their familiesProvide easy-to-understand explanations without using technical terms5Provide explanations so that patients/families do not develop misconceptions4Convey information carefully and clearly3Provide a written explanation2Divide bad news into components14Respect for the intentions of patients and families(1)Respect for the will of patients and familiesRespect and support the patient’s will and desired treatment6If the patient’s condition is severe, respect the views of the family and primary caregiver2(2)Setting goals in accordance with the preferences of the patients and familiesListen to the needs of patients and their families and determine actionable treatment goals3Explore the intentions of patients and their families through conversations1If there is no difference between the advantages and disadvantages of a treatment choice, the choice is purely made by the patient15Directing decision-making and considering fairness(1)Considering the direction and fairness of treatment options recommended by doctorsExplain treatment options with some direction while also providing options that the doctor finds beneficial8In some cases, offer personal opinion as a doctor2Dare to treat the patient with paternalism2(2)Consideration for patients to make impartial decisionsCommunicate with the knowledge that the doctor’s manner of speaking influences the patients’ decisions5Explain the opinions of other doctors and the second opinion system16Considering the emotional aspects of the patient and family(1)Explaining with consideration for emotionsTalk in a way that allows the patient to have hope5Convey bad news according to the protocol1Consider that patients do not have to listen to explanations while always looking at bad information1Divulge bad news in stages1(2)Showing a supportive attitude to the patientApproach the patient with a non-abandoning attitude2Show that the team supports the patient1(3)Attitude when facing patients and their familiesInteract with patients on an equal footing2Talk while looking into the patient’s eyes1Interact with the patient’s family on an equal footing17Supporting after discussing treatments(1)Emotional support after discussing treatmentsSupport fluctuations in the patient’s mind after discussing treatments2Call out the family and assess their reactions1*1IC = informed consent; *2PHS = Personal Handyphone System (A phone used by doctors to communicate with staff in the hospital). Role of hematologists in providing direct decision support (n = 11). *1IC = informed consent; *2PHS = Personal Handyphone System (A phone used by doctors to communicate with staff in the hospital). [SUBTITLE] Preparing Patients Before Informed Consent (IC) [SUBSECTION] This category comprised 4 subcategories. Several participants talked about preparing patients and families for IC discussions, including “Gathering preliminary information for explaining issues to patients” and “Encouraging family members to participate in decision-making.” As such, physicians were expected to prepare patients before IC discussions. For instance, they would provide patients with information regarding the risk of relapse. This category comprised 4 subcategories. Several participants talked about preparing patients and families for IC discussions, including “Gathering preliminary information for explaining issues to patients” and “Encouraging family members to participate in decision-making.” As such, physicians were expected to prepare patients before IC discussions. For instance, they would provide patients with information regarding the risk of relapse. [SUBTITLE] Selecting the Information To Convey [SUBSECTION] This category comprised 3 subcategories. In subcategory (1) “Choice of information to provide to patients,” more than half of the participants said that they did not provide their patients with prognoses. More importantly, more than half said they may not even tell their patients the truth, depending on the severity of the illness, to avoid a loss of hope; they were very selective about the information they chose to provide to their patients. In subcategory (2) “Providing patients and families with information to help them make treatment decisions,” more than half of the participants identified “Communicating the advantages and disadvantages of treatment options” and “Telling patients and their families the information they need to make treatment decisions.” They provided information they believed would help patients and their families fully understand all the options before making a final decision. In subcategory (3), “Providing patients and families with information to help them understand the patients’ current situation,” a large number said they would inform family members about the possibility of sudden changes and specific prognoses, with more than half saying they would explain the issues so that patients could visualize what may happen to them in the future. Generally, participants provided information to help patients and their families clearly understand their current situations. This category comprised 3 subcategories. In subcategory (1) “Choice of information to provide to patients,” more than half of the participants said that they did not provide their patients with prognoses. More importantly, more than half said they may not even tell their patients the truth, depending on the severity of the illness, to avoid a loss of hope; they were very selective about the information they chose to provide to their patients. In subcategory (2) “Providing patients and families with information to help them make treatment decisions,” more than half of the participants identified “Communicating the advantages and disadvantages of treatment options” and “Telling patients and their families the information they need to make treatment decisions.” They provided information they believed would help patients and their families fully understand all the options before making a final decision. In subcategory (3), “Providing patients and families with information to help them understand the patients’ current situation,” a large number said they would inform family members about the possibility of sudden changes and specific prognoses, with more than half saying they would explain the issues so that patients could visualize what may happen to them in the future. Generally, participants provided information to help patients and their families clearly understand their current situations. [SUBTITLE] Choosing the Way In Which To Communicate Information [SUBSECTION] This category comprised 2 subcategories. Nearly half of participants expressed the ideas of “I determine what I say and how I say it based on our long-standing relationship with the patients” and “Providing information based on the patient’s reaction and acceptance of their medical condition.” They carefully considered what they discussed and how they discussed it, especially considering their long-standing association with their patients. In this regard, they conveyed information based on how they thought patients would react and their level of acceptance. They also provided explanations according to what they believed patients intended and thought depending on their respective stages of readiness. Moreover, nearly half said that they would explain issues without using jargon, thereby ensuring that the information provided was easy to understand. Essentially, participants remained cognizant of the need to communicate both correctly and appropriately with patients and their families. This category comprised 2 subcategories. Nearly half of participants expressed the ideas of “I determine what I say and how I say it based on our long-standing relationship with the patients” and “Providing information based on the patient’s reaction and acceptance of their medical condition.” They carefully considered what they discussed and how they discussed it, especially considering their long-standing association with their patients. In this regard, they conveyed information based on how they thought patients would react and their level of acceptance. They also provided explanations according to what they believed patients intended and thought depending on their respective stages of readiness. Moreover, nearly half said that they would explain issues without using jargon, thereby ensuring that the information provided was easy to understand. Essentially, participants remained cognizant of the need to communicate both correctly and appropriately with patients and their families. [SUBTITLE] Respect for The Intentions Of Patients And Families [SUBSECTION] This category comprised 2 subcategories. In subcategory (1) “Respect for the will of patients and families,” more than half of the participants said they helped patients and families make decisions by supporting and respecting their wishes and desired treatments. In subcategory (2) “Setting goals in accordance with the preferences of the patients and families,” several participants said that they helped patients and families set feasible goals after listening to their wishes. This category comprised 2 subcategories. In subcategory (1) “Respect for the will of patients and families,” more than half of the participants said they helped patients and families make decisions by supporting and respecting their wishes and desired treatments. In subcategory (2) “Setting goals in accordance with the preferences of the patients and families,” several participants said that they helped patients and families set feasible goals after listening to their wishes. [SUBTITLE] Directing decision-making and considering fairness [SUBSECTION] This category comprised 2 subcategories. In subcategory (1) “Considering the direction and fairness of treatment options recommended by doctors,” more than half of the participants said they explain treatment options with some direction while also providing options they find most beneficial. They talked about giving weight to treatment options and facilitating the decision-making process. Furthermore, they attempted to present information in a fair manner, being aware that the way they talk can affect the way patients perceive their treatment, as shown in subcategory (2). This category comprised 2 subcategories. In subcategory (1) “Considering the direction and fairness of treatment options recommended by doctors,” more than half of the participants said they explain treatment options with some direction while also providing options they find most beneficial. They talked about giving weight to treatment options and facilitating the decision-making process. Furthermore, they attempted to present information in a fair manner, being aware that the way they talk can affect the way patients perceive their treatment, as shown in subcategory (2). [SUBTITLE] Considering the Emotional Aspects Of The Patient And Family [SUBSECTION] This category comprised 3 subcategories. In subcategory (1)“explaining with consideration for emotions,” nearly half of the participants said they would attempt to instill hope among patients, and hence, provide explanations considering the related emotional aspects. In subcategory (2) “showing a supportive attitude to the patient”, several participants also expressed adopting a supportive stance to reassure patients that they were not abandoning them or their families. In subcategory (3) “attitude when facing patients and their families”, several participants talked about the doctor’s attitude toward patients, such as speaking with patients and families as equals and looking them in the eye. This category comprised 3 subcategories. In subcategory (1)“explaining with consideration for emotions,” nearly half of the participants said they would attempt to instill hope among patients, and hence, provide explanations considering the related emotional aspects. In subcategory (2) “showing a supportive attitude to the patient”, several participants also expressed adopting a supportive stance to reassure patients that they were not abandoning them or their families. In subcategory (3) “attitude when facing patients and their families”, several participants talked about the doctor’s attitude toward patients, such as speaking with patients and families as equals and looking them in the eye. [SUBTITLE] Providing support after discussing treatments [SUBSECTION] Several participants said they informed patients about bad news and future treatments, after which they provided emotional support in consideration of any emotional variabilities. Several participants said they informed patients about bad news and future treatments, after which they provided emotional support in consideration of any emotional variabilities. [SUBTITLE] Role of Hematologists in Multidisciplinary Collaboration and Providing Decision Support [SUBSECTION] We identified the following 5 subcategories related to the second main study topic—the roles of hematologists in multidisciplinary collaboration: (1) communicating with other professionals, (2) gathering information from them, (3) providing information to them, (4) managing the entire medical team, and (5) encouraging nurses to actively participate with patients throughout the decision-making process (Table 3). More than half of the participants recognized the hematologist’s role in communicating with other professionals and obtaining information about the patient (eg, the patient’s background and how to help) throughout this process. Others said that they actively encourage nurses to support the patients’ decisions and provide additional information during the discussions or explain the prognoses and future treatment plans to other professionals.Table 3.The role of hematologists in multidisciplinary collaboration and decision support (n = 11).SubcategoryCode1Communicating with other professionalsValue good communication with other professionals, such as nursesMake opportunities to hold discussions with multiple professionals2Gathering information from other professionalsObtain information from nurses about family background and family supportObtain information about the patients’ thoughts and intentions from other professionalsObtain information about the patients’ social background from other professionalsObtain information about the patient’s ADL status from other professionals3Providing information to other professionalsExplain the patient’s prognosis and future treatment plans to other professionalsExplain the concerns of the patient’s family to other professionals4Managing the entire medical teamManage the medical team, in which hematologists make comprehensive decisions based on information from other professionalsCreate a support system for patients5Encouraging nurses to actively participate in the patient’s decision-makingEncourage nurses to participate in the patients’ decision-makingConfirm whether there is any additional information or thoughts to convey to nurses during discussions about treatment decisions with the patientADL = Activities of Daily Living. The role of hematologists in multidisciplinary collaboration and decision support (n = 11). ADL = Activities of Daily Living. We identified the following 5 subcategories related to the second main study topic—the roles of hematologists in multidisciplinary collaboration: (1) communicating with other professionals, (2) gathering information from them, (3) providing information to them, (4) managing the entire medical team, and (5) encouraging nurses to actively participate with patients throughout the decision-making process (Table 3). More than half of the participants recognized the hematologist’s role in communicating with other professionals and obtaining information about the patient (eg, the patient’s background and how to help) throughout this process. Others said that they actively encourage nurses to support the patients’ decisions and provide additional information during the discussions or explain the prognoses and future treatment plans to other professionals.Table 3.The role of hematologists in multidisciplinary collaboration and decision support (n = 11).SubcategoryCode1Communicating with other professionalsValue good communication with other professionals, such as nursesMake opportunities to hold discussions with multiple professionals2Gathering information from other professionalsObtain information from nurses about family background and family supportObtain information about the patients’ thoughts and intentions from other professionalsObtain information about the patients’ social background from other professionalsObtain information about the patient’s ADL status from other professionals3Providing information to other professionalsExplain the patient’s prognosis and future treatment plans to other professionalsExplain the concerns of the patient’s family to other professionals4Managing the entire medical teamManage the medical team, in which hematologists make comprehensive decisions based on information from other professionalsCreate a support system for patients5Encouraging nurses to actively participate in the patient’s decision-makingEncourage nurses to participate in the patients’ decision-makingConfirm whether there is any additional information or thoughts to convey to nurses during discussions about treatment decisions with the patientADL = Activities of Daily Living. The role of hematologists in multidisciplinary collaboration and decision support (n = 11). ADL = Activities of Daily Living. [SUBTITLE] Expected Roles of Other Professionals in Providing Decision Support [SUBSECTION] We extracted the following 4 categories related to the roles that hematologists expected other professionals to fulfill: (1) sharing patient information and care routines, (2) sharing information from treatment discussions, (3) providing emotional support to patients and their families, and (4) assembling a medical team that understands hematological malignancies (Table 4). Nearly half of the participants indicated that they would like other health care providers to convey the patients’ thoughts and intentions that they knew to the doctors involved. They also expected other professionals to share information regarding their patients and the way in which they cared for their patients.Table 4.Expected roles of other professionals in decision support (n = 11).SubcategoryCodenSharing patient information and care routinesListen to the patients’ thoughts and wishes and communicate them to the doctor5Suggest knowledge that doctors may not know and explain how to care for patients in the right way5Point out the problems patients have with treatment decisions2Sharing information from the treatment discussionAttend treatment discussions and understand the patients’ feelings and situation together4Read records of the process of informed consent and share the information1Providing patients and their families with emotional supportSupport the emotions of patients and their families4Assembling a medical team that understands hematological malignancyWork to ensure that the medical team understands hematological malignancy1 Expected roles of other professionals in decision support (n = 11). We extracted the following 4 categories related to the roles that hematologists expected other professionals to fulfill: (1) sharing patient information and care routines, (2) sharing information from treatment discussions, (3) providing emotional support to patients and their families, and (4) assembling a medical team that understands hematological malignancies (Table 4). Nearly half of the participants indicated that they would like other health care providers to convey the patients’ thoughts and intentions that they knew to the doctors involved. They also expected other professionals to share information regarding their patients and the way in which they cared for their patients.Table 4.Expected roles of other professionals in decision support (n = 11).SubcategoryCodenSharing patient information and care routinesListen to the patients’ thoughts and wishes and communicate them to the doctor5Suggest knowledge that doctors may not know and explain how to care for patients in the right way5Point out the problems patients have with treatment decisions2Sharing information from the treatment discussionAttend treatment discussions and understand the patients’ feelings and situation together4Read records of the process of informed consent and share the information1Providing patients and their families with emotional supportSupport the emotions of patients and their families4Assembling a medical team that understands hematological malignancyWork to ensure that the medical team understands hematological malignancy1 Expected roles of other professionals in decision support (n = 11).
null
null
[ "Participant Characteristics", "The Role of Hematologists in Direct Decision Support", "Preparing Patients Before Informed Consent (IC)", "Selecting the Information To Convey", "Choosing the Way In Which To Communicate Information", "Respect for The Intentions Of Patients And Families", "Directing decision-making and considering fairness", "Considering the Emotional Aspects Of The Patient And Family", "Providing support after discussing treatments", "Role of Hematologists in Multidisciplinary Collaboration and Providing\nDecision Support", "Expected Roles of Other Professionals in Providing Decision Support" ]
[ "We conducted semi-structured interviews involving 11 hematologists. The average\nparticipant age was 44 years (range: 35-54 years), whereas the average\nspecialization experience was 16 years (range: 7-26 years). Additional details\nare listed in Table\n1. The average length of the interviews was 27 minutes (range: 17-46\nminutes).Table\n1.Characteristics of doctors and interview\ndetails.HematologistsTotaln\n= 11Gender—Male10Female1Workplace—University hospital7Cancer center2General\nhospital2Age (years)43.9 (34-54)*Experience in specialty (years)16.1\n(7-26)*∗Mean(Range).\nCharacteristics of doctors and interview\ndetails.\n∗Mean(Range).", "As shown in Table 2,\nwe identified 7 categories related to hematologists’ roles in providing direct\ndecision support to patients: (1) preparing patients before informed consent,\n(2) selecting the information to convey, (3) choosing a method for conveying\nthis information, (4) respecting the intentions of patients and their families,\n(5) directing decision-making and considering fairness, (6) considering the\nemotional aspects of patients and their families, and (7) providing support\nafter discussing treatment options.Table 2.Role of hematologists in providing\ndirect decision support (n =\n11).CategorySubcategoryCoden1Preparing patients before\nIC*(1)Gathering preliminary\ninformation for explaining issues to patientsObtain information from family members to\nunderstand the patient’s preferences in advance3Check if home care is possible1Understand the\nfamily background1Talk to the family about\nwhat you will tell the patient1(2)Encouraging family\nmembers to participate in decision-makingSet up a place to provide information to\npatients, including family members3Discuss\nlife-prolonging treatments with the patient’s\nfamily1(3)Creating an environment for concentrating on\nICDiscuss in a private\nroom2Take sufficient time1Leave the\nPHS*2 with\nthe nurse so that the discussion is not\ndisturbed1(4)Providing advance information on the risk\nof relapseWhen the patient reaches\ncomplete remission, explain the risk of relapse and\nprepare the patient for relapse12Selecting the\ninformation to convey(1)Choice of\ninformation to provide to patientsIn many cases, do not give the patient a\nprognosis9Depending on the\npatient’s medical condition, I (doctor) may not tell the\ntruth5Modify the message according to the\npatient’s life stage3(2)Providing patients and families\nwith information to help them make treatment\ndecisionsCommunicate the\nadvantages and disadvantages of treatment\noptions9Tell patients and their families the\ninformation they need to make treatment\ndecisions6Inform patients of all\ntreatment options4Communicate not only the\nbad news, but also ways of improvement3Also convey the option of non-treatment3(3)Providing patients and families with information to\nhelp them understand the patients' current\nsituationInform family members\nabout the possibility of sudden changes in the medical\ncondition and specific prognosis9Provide\npatients with information related to what they imagine\nwill happen in the future5Provide patients with\ninformation that will help them understand their current\nmedical condition4Provide patients with a\nvague idea of their prognosis3Communicate the\nprognosis to patients using concrete numbers23Choosing\nhow to communicate information(1)Explanations based on the patients' stage of\nreadinessJudge what I (doctor) say\nand how I (doctor) say it based on long-standing\nrelationship with the patients5Provide\ninformation based on the patient’s reaction and\nacceptance of their medical condition5(2)Respect\nfor the preferences of patients and their\nfamiliesProvide easy-to-understand\nexplanations without using technical terms5Provide explanations so that patients/families do not\ndevelop misconceptions4Convey information\ncarefully and clearly3Provide a written\nexplanation2Divide bad news into\ncomponents14Respect for the intentions of\npatients and families(1)Respect for the\nwill of patients and familiesRespect and support the patient’s will and desired\ntreatment6If the patient’s\ncondition is severe, respect the views of the family and\nprimary caregiver2(2)Setting goals in accordance\nwith the preferences of the patients and\nfamiliesListen to the needs of\npatients and their families and determine actionable\ntreatment goals3Explore the intentions of\npatients and their families through\nconversations1If there is no\ndifference between the advantages and disadvantages of a\ntreatment choice, the choice is purely made by the\npatient15Directing decision-making and considering\nfairness(1)Considering the direction and\nfairness of treatment options recommended by\ndoctorsExplain treatment options\nwith some direction while also providing options that\nthe doctor finds beneficial8In some cases,\noffer personal opinion as a doctor2Dare to treat\nthe patient with paternalism2(2)Consideration for\npatients to make impartial decisionsCommunicate with the knowledge that the\ndoctor’s manner of speaking influences the patients’\ndecisions5Explain the opinions of\nother doctors and the second opinion system16Considering the emotional aspects of the patient and\nfamily(1)Explaining with consideration\nfor emotionsTalk in a way that\nallows the patient to have hope5Convey bad news\naccording to the protocol1Consider that patients\ndo not have to listen to explanations while always\nlooking at bad information1Divulge bad news in\nstages1(2)Showing a supportive attitude to the\npatientApproach the patient with a\nnon-abandoning attitude2Show that the team\nsupports the patient1(3)Attitude when facing patients\nand their familiesInteract with\npatients on an equal footing2Talk while\nlooking into the patient’s eyes1Interact with\nthe patient’s family on an equal footing17Supporting after discussing treatments(1)Emotional support after discussing\ntreatmentsSupport fluctuations in\nthe patient’s mind after discussing treatments2Call out the family and assess their reactions1*1IC\n= informed consent; *2PHS = Personal Handyphone\nSystem (A phone used by doctors to communicate with staff in the\nhospital).\nRole of hematologists in providing\ndirect decision support (n =\n11).\n*1IC\n= informed consent; *2PHS = Personal Handyphone\nSystem (A phone used by doctors to communicate with staff in the\nhospital).\n[SUBTITLE] Preparing Patients Before Informed Consent (IC) [SUBSECTION] This category comprised 4 subcategories. Several participants talked about\npreparing patients and families for IC discussions, including “Gathering\npreliminary information for explaining issues to patients” and “Encouraging\nfamily members to participate in decision-making.” As such, physicians were\nexpected to prepare patients before IC discussions. For instance, they would\nprovide patients with information regarding the risk of relapse.\nThis category comprised 4 subcategories. Several participants talked about\npreparing patients and families for IC discussions, including “Gathering\npreliminary information for explaining issues to patients” and “Encouraging\nfamily members to participate in decision-making.” As such, physicians were\nexpected to prepare patients before IC discussions. For instance, they would\nprovide patients with information regarding the risk of relapse.\n[SUBTITLE] Selecting the Information To Convey [SUBSECTION] This category comprised 3 subcategories. In subcategory (1) “Choice of\ninformation to provide to patients,” more than half of the participants said\nthat they did not provide their patients with prognoses. More importantly,\nmore than half said they may not even tell their patients the truth,\ndepending on the severity of the illness, to avoid a loss of hope; they were\nvery selective about the information they chose to provide to their\npatients.\nIn subcategory (2) “Providing patients and families with information to help\nthem make treatment decisions,” more than half of the participants\nidentified “Communicating the advantages and disadvantages of treatment\noptions” and “Telling patients and their families the information they need\nto make treatment decisions.” They provided information they believed would\nhelp patients and their families fully understand all the options before\nmaking a final decision.\nIn subcategory (3), “Providing patients and families with information to help\nthem understand the patients’ current situation,” a large number said they\nwould inform family members about the possibility of sudden changes and\nspecific prognoses, with more than half saying they would explain the issues\nso that patients could visualize what may happen to them in the future.\nGenerally, participants provided information to help patients and their\nfamilies clearly understand their current situations.\nThis category comprised 3 subcategories. In subcategory (1) “Choice of\ninformation to provide to patients,” more than half of the participants said\nthat they did not provide their patients with prognoses. More importantly,\nmore than half said they may not even tell their patients the truth,\ndepending on the severity of the illness, to avoid a loss of hope; they were\nvery selective about the information they chose to provide to their\npatients.\nIn subcategory (2) “Providing patients and families with information to help\nthem make treatment decisions,” more than half of the participants\nidentified “Communicating the advantages and disadvantages of treatment\noptions” and “Telling patients and their families the information they need\nto make treatment decisions.” They provided information they believed would\nhelp patients and their families fully understand all the options before\nmaking a final decision.\nIn subcategory (3), “Providing patients and families with information to help\nthem understand the patients’ current situation,” a large number said they\nwould inform family members about the possibility of sudden changes and\nspecific prognoses, with more than half saying they would explain the issues\nso that patients could visualize what may happen to them in the future.\nGenerally, participants provided information to help patients and their\nfamilies clearly understand their current situations.\n[SUBTITLE] Choosing the Way In Which To Communicate Information [SUBSECTION] This category comprised 2 subcategories. Nearly half of participants\nexpressed the ideas of “I determine what I say and how I say it based on our\nlong-standing relationship with the patients” and “Providing information\nbased on the patient’s reaction and acceptance of their medical condition.”\nThey carefully considered what they discussed and how they discussed it,\nespecially considering their long-standing association with their patients.\nIn this regard, they conveyed information based on how they thought patients\nwould react and their level of acceptance. They also provided explanations\naccording to what they believed patients intended and thought depending on\ntheir respective stages of readiness. Moreover, nearly half said that they\nwould explain issues without using jargon, thereby ensuring that the\ninformation provided was easy to understand. Essentially, participants\nremained cognizant of the need to communicate both correctly and\nappropriately with patients and their families.\nThis category comprised 2 subcategories. Nearly half of participants\nexpressed the ideas of “I determine what I say and how I say it based on our\nlong-standing relationship with the patients” and “Providing information\nbased on the patient’s reaction and acceptance of their medical condition.”\nThey carefully considered what they discussed and how they discussed it,\nespecially considering their long-standing association with their patients.\nIn this regard, they conveyed information based on how they thought patients\nwould react and their level of acceptance. They also provided explanations\naccording to what they believed patients intended and thought depending on\ntheir respective stages of readiness. Moreover, nearly half said that they\nwould explain issues without using jargon, thereby ensuring that the\ninformation provided was easy to understand. Essentially, participants\nremained cognizant of the need to communicate both correctly and\nappropriately with patients and their families.\n[SUBTITLE] Respect for The Intentions Of Patients And Families [SUBSECTION] This category comprised 2 subcategories. In subcategory (1) “Respect for the\nwill of patients and families,” more than half of the participants said they\nhelped patients and families make decisions by supporting and respecting\ntheir wishes and desired treatments. In subcategory (2) “Setting goals in\naccordance with the preferences of the patients and families,” several\nparticipants said that they helped patients and families set feasible goals\nafter listening to their wishes.\nThis category comprised 2 subcategories. In subcategory (1) “Respect for the\nwill of patients and families,” more than half of the participants said they\nhelped patients and families make decisions by supporting and respecting\ntheir wishes and desired treatments. In subcategory (2) “Setting goals in\naccordance with the preferences of the patients and families,” several\nparticipants said that they helped patients and families set feasible goals\nafter listening to their wishes.\n[SUBTITLE] Directing decision-making and considering fairness [SUBSECTION] This category comprised 2 subcategories. In subcategory (1) “Considering the\ndirection and fairness of treatment options recommended by doctors,” more\nthan half of the participants said they explain treatment options with some\ndirection while also providing options they find most beneficial. They\ntalked about giving weight to treatment options and facilitating the\ndecision-making process. Furthermore, they attempted to present information\nin a fair manner, being aware that the way they talk can affect the way\npatients perceive their treatment, as shown in subcategory (2).\nThis category comprised 2 subcategories. In subcategory (1) “Considering the\ndirection and fairness of treatment options recommended by doctors,” more\nthan half of the participants said they explain treatment options with some\ndirection while also providing options they find most beneficial. They\ntalked about giving weight to treatment options and facilitating the\ndecision-making process. Furthermore, they attempted to present information\nin a fair manner, being aware that the way they talk can affect the way\npatients perceive their treatment, as shown in subcategory (2).\n[SUBTITLE] Considering the Emotional Aspects Of The Patient And Family [SUBSECTION] This category comprised 3 subcategories. In subcategory (1)“explaining with\nconsideration for emotions,” nearly half of the participants said they would\nattempt to instill hope among patients, and hence, provide explanations\nconsidering the related emotional aspects. In subcategory (2) “showing a\nsupportive attitude to the patient”, several participants also expressed\nadopting a supportive stance to reassure patients that they were not\nabandoning them or their families. In subcategory (3) “attitude when facing\npatients and their families”, several participants talked about the doctor’s\nattitude toward patients, such as speaking with patients and families as\nequals and looking them in the eye.\nThis category comprised 3 subcategories. In subcategory (1)“explaining with\nconsideration for emotions,” nearly half of the participants said they would\nattempt to instill hope among patients, and hence, provide explanations\nconsidering the related emotional aspects. In subcategory (2) “showing a\nsupportive attitude to the patient”, several participants also expressed\nadopting a supportive stance to reassure patients that they were not\nabandoning them or their families. In subcategory (3) “attitude when facing\npatients and their families”, several participants talked about the doctor’s\nattitude toward patients, such as speaking with patients and families as\nequals and looking them in the eye.\n[SUBTITLE] Providing support after discussing treatments [SUBSECTION] Several participants said they informed patients about bad news and future\ntreatments, after which they provided emotional support in consideration of\nany emotional variabilities.\nSeveral participants said they informed patients about bad news and future\ntreatments, after which they provided emotional support in consideration of\nany emotional variabilities.", "This category comprised 4 subcategories. Several participants talked about\npreparing patients and families for IC discussions, including “Gathering\npreliminary information for explaining issues to patients” and “Encouraging\nfamily members to participate in decision-making.” As such, physicians were\nexpected to prepare patients before IC discussions. For instance, they would\nprovide patients with information regarding the risk of relapse.", "This category comprised 3 subcategories. In subcategory (1) “Choice of\ninformation to provide to patients,” more than half of the participants said\nthat they did not provide their patients with prognoses. More importantly,\nmore than half said they may not even tell their patients the truth,\ndepending on the severity of the illness, to avoid a loss of hope; they were\nvery selective about the information they chose to provide to their\npatients.\nIn subcategory (2) “Providing patients and families with information to help\nthem make treatment decisions,” more than half of the participants\nidentified “Communicating the advantages and disadvantages of treatment\noptions” and “Telling patients and their families the information they need\nto make treatment decisions.” They provided information they believed would\nhelp patients and their families fully understand all the options before\nmaking a final decision.\nIn subcategory (3), “Providing patients and families with information to help\nthem understand the patients’ current situation,” a large number said they\nwould inform family members about the possibility of sudden changes and\nspecific prognoses, with more than half saying they would explain the issues\nso that patients could visualize what may happen to them in the future.\nGenerally, participants provided information to help patients and their\nfamilies clearly understand their current situations.", "This category comprised 2 subcategories. Nearly half of participants\nexpressed the ideas of “I determine what I say and how I say it based on our\nlong-standing relationship with the patients” and “Providing information\nbased on the patient’s reaction and acceptance of their medical condition.”\nThey carefully considered what they discussed and how they discussed it,\nespecially considering their long-standing association with their patients.\nIn this regard, they conveyed information based on how they thought patients\nwould react and their level of acceptance. They also provided explanations\naccording to what they believed patients intended and thought depending on\ntheir respective stages of readiness. Moreover, nearly half said that they\nwould explain issues without using jargon, thereby ensuring that the\ninformation provided was easy to understand. Essentially, participants\nremained cognizant of the need to communicate both correctly and\nappropriately with patients and their families.", "This category comprised 2 subcategories. In subcategory (1) “Respect for the\nwill of patients and families,” more than half of the participants said they\nhelped patients and families make decisions by supporting and respecting\ntheir wishes and desired treatments. In subcategory (2) “Setting goals in\naccordance with the preferences of the patients and families,” several\nparticipants said that they helped patients and families set feasible goals\nafter listening to their wishes.", "This category comprised 2 subcategories. In subcategory (1) “Considering the\ndirection and fairness of treatment options recommended by doctors,” more\nthan half of the participants said they explain treatment options with some\ndirection while also providing options they find most beneficial. They\ntalked about giving weight to treatment options and facilitating the\ndecision-making process. Furthermore, they attempted to present information\nin a fair manner, being aware that the way they talk can affect the way\npatients perceive their treatment, as shown in subcategory (2).", "This category comprised 3 subcategories. In subcategory (1)“explaining with\nconsideration for emotions,” nearly half of the participants said they would\nattempt to instill hope among patients, and hence, provide explanations\nconsidering the related emotional aspects. In subcategory (2) “showing a\nsupportive attitude to the patient”, several participants also expressed\nadopting a supportive stance to reassure patients that they were not\nabandoning them or their families. In subcategory (3) “attitude when facing\npatients and their families”, several participants talked about the doctor’s\nattitude toward patients, such as speaking with patients and families as\nequals and looking them in the eye.", "Several participants said they informed patients about bad news and future\ntreatments, after which they provided emotional support in consideration of\nany emotional variabilities.", "We identified the following 5 subcategories related to the second main study\ntopic—the roles of hematologists in multidisciplinary collaboration: (1)\ncommunicating with other professionals, (2) gathering information from them, (3)\nproviding information to them, (4) managing the entire medical team, and (5)\nencouraging nurses to actively participate with patients throughout the\ndecision-making process (Table 3). More than half of the participants recognized the\nhematologist’s role in communicating with other professionals and obtaining\ninformation about the patient (eg, the patient’s background and how to help)\nthroughout this process. Others said that they actively encourage nurses to\nsupport the patients’ decisions and provide additional information during the\ndiscussions or explain the prognoses and future treatment plans to other\nprofessionals.Table\n3.The role of hematologists in multidisciplinary\ncollaboration and decision support (n =\n11).SubcategoryCode1Communicating with other professionalsValue good communication with other\nprofessionals, such as nursesMake opportunities to hold discussions with\nmultiple professionals2Gathering\ninformation from other professionalsObtain information from nurses about family\nbackground and family supportObtain information about the patients’\nthoughts and intentions from other\nprofessionalsObtain\ninformation about the patients’ social background from\nother professionalsObtain\ninformation about the patient’s ADL status from other\nprofessionals3Providing\ninformation to other professionalsExplain the patient’s prognosis and future treatment\nplans to other professionalsExplain the concerns of the patient’s\nfamily to other professionals4Managing the entire medical teamManage the medical team, in which\nhematologists make comprehensive decisions based on\ninformation from other professionalsCreate a support system for\npatients5Encouraging nurses to actively\nparticipate in the patient’s decision-makingEncourage nurses to participate in the\npatients’ decision-makingConfirm whether there is any additional information or\nthoughts to convey to nurses during discussions about\ntreatment decisions with the\npatientADL\n= Activities of Daily\nLiving.\nThe role of hematologists in multidisciplinary\ncollaboration and decision support (n =\n11).\nADL\n= Activities of Daily\nLiving.", "We extracted the following 4 categories related to the roles that hematologists\nexpected other professionals to fulfill: (1) sharing patient information and\ncare routines, (2) sharing information from treatment discussions, (3) providing\nemotional support to patients and their families, and (4) assembling a medical\nteam that understands hematological malignancies (Table 4). Nearly half of the\nparticipants indicated that they would like other health care providers to\nconvey the patients’ thoughts and intentions that they knew to the doctors\ninvolved. They also expected other professionals to share information regarding\ntheir patients and the way in which they cared for their patients.Table 4.Expected\nroles of other professionals in decision support (n =\n11).SubcategoryCodenSharing patient information and care\nroutinesListen to the patients’\nthoughts and wishes and communicate them to the\ndoctor5Suggest knowledge that doctors may not know\nand explain how to care for patients in the right\nway5Point out the problems patients have with treatment\ndecisions2Sharing information from the\ntreatment discussionAttend\ntreatment discussions and understand the patients’\nfeelings and situation together4Read records of the\nprocess of informed consent and share the\ninformation1Providing patients and their families with\nemotional supportSupport the\nemotions of patients and their families4Assembling\na medical team that understands hematological\nmalignancyWork to ensure that the\nmedical team understands hematological\nmalignancy1\nExpected\nroles of other professionals in decision support (n =\n11)." ]
[ null, null, null, null, null, null, null, null, null, null, null ]
[ "Introduction", "Methods", "Results", "Participant Characteristics", "The Role of Hematologists in Direct Decision Support", "Preparing Patients Before Informed Consent (IC)", "Selecting the Information To Convey", "Choosing the Way In Which To Communicate Information", "Respect for The Intentions Of Patients And Families", "Directing decision-making and considering fairness", "Considering the Emotional Aspects Of The Patient And Family", "Providing support after discussing treatments", "Role of Hematologists in Multidisciplinary Collaboration and Providing\nDecision Support", "Expected Roles of Other Professionals in Providing Decision Support", "Discussion", "Supplemental Material" ]
[ "The prognosis and illness trajectory of leukemia and lymphoma vary greatly depending\non the degree of progression and subtype of the disease. However, irrespective of\nthe illness trajectory, patients face the same issues and debates related to\ndecision-making, such as how far to go with aggressive treatment during the relapse\nor treatment-resistant period and the choice of treatment and place of\ncare.1-3 Even in relapse cases, such\npatients may have the option to receive high-dose chemotherapy treatments or\nhematopoietic cell transplantations aimed at remission and achieving long-term\nsurvival. Such treatment carries significant risks, including complications such as\nsevere infections and bleeding due to bone marrow suppression and treatment-related\ndeath, and may result in transition from invasive treatment aimed at cure to\nend-of-life (EOL). Even when the disease is refractory to treatment, anticancer\ntherapy aimed at disease control is often administered until the end of life. In\nsuch contexts, it is very difficult for patients and their families to make\ndecisions about these complex treatment strategies and the high risks involved in\nthe context of their own life values.4-6\nThe American Society of Clinical Oncology (ASCO) has published consensus guidelines\nfor clinician-patient communication, specifically recommending that clinicians\nprovide information that is congruent with their patients’ interests and intentions\nwhile jointly considering their goals for treatment.7 It is also notable that\nhematologists’ communication skills have been receiving increasing attention in\nhematology clinical practice in recent years. An ethnographic study of\nhematologists' communication patterns when communicating bad news revealed 4\ncharacteristics: (1) technical-defensive patterns, (2) an authoritative pattern, (3)\na relational-recursive pattern, and (4) a compassionate sharing pattern, and the\npaper also suggested that hematologists were not good at expressing caring and\nempathy.8 In contrast, a qualitative study examining the needs of patients\nwith hematological malignancies when being told bad news reported identifying\nemotional support from their healthcare providers and information needs about future\nsupport systems. It was also noted that healthcare-team involvement is necessary to\nmeet these patients' needs and support their decision-making.9\nTherefore, this study sought to clarify the following: (a) the way in which\nhematologists perceive the roles of doctors in providing decision-making support to\npatients suffering from relapsed and refractory leukemia as well as lymphoma, (b)\nthe expected roles of hematologists in supporting decision-making via\nmultidisciplinary collaboration, and (c) the roles hematologists expect other\nhealthcare providers to fulfill.", "The purpose of this study was to determine hematologists’ role in decision support\nfor patients with leukemia and lymphoma. Physicians at institutions with hematology\nwards and hematology outpatient clinics where chemotherapy and bone marrow\ntransplantation are performed were included. The eligibility criteria for physicians\nincluded being Board Certified Hematologists and having at least 5 years of\nexperience. Hematologists meeting these criteria were considered to have experience\nseeing patients with relapsed or refractory leukemia or lymphoma. Subjects were\nselected using opportunistic sampling, which is used in selecting those with common\ncharacteristics and the potential to provide a rich, appropriate, and diverse\nresearch agenda. Eligible physicians were asked to recommend a qualified physician\nwho met the inclusion criteria to the head of the hematology department of a\nhospital in Tokyo. The recommended physicians were informed orally and in writing\nabout the purpose and methods of the study and the voluntary participation clause.\nAlthough this is a qualitative study and it is difficult to theoretically calculate\nthe sample size, we chose a sample size of 10 to 15 participants based on previous\nstudies9-11 using the\nsame content analysis method as in this study. The acceptance rate of recommended\nphysicians was 100%.\nVerbal and written explanations of the research objectives and procedures were\nprovided to all participants, and written consent was obtained. This study was\napproved by the Institutional Review Board of University of Tokyo (Approval No.\n3443). The study was initiated at the University of Tokyo and completed at Kyoto\nUniversity.\nTo collect the data, we conducted in-depth interviews to identify the attributes and\nroles of doctors as well as the expected roles of nurses in providing\ndecision-making support to patients with relapsed or refractory leukemia and\nlymphoma. Specifically, we scheduled face-to-face semi-structured interviews in\nprivate rooms between August and December 2011. All the interviews conducted were\naudio recorded and fully transcribed by 1 researcher (a research nurse), who also\ntook detailed field notes during each interview. Physicians were first asked for\ntheir demographic details, including age, the type of facility where they worked,\nand the number of years they had worked in their area of specialization. We then\nasked them about their roles in treating patients suffering from relapsed or\nrefractory leukemia and lymphoma.\nTo analyze the data, interview data were transcribed verbatim from the audio\nrecordings. Interview data were then numbered for each participant, separated from\npersonal information, and anonymized. A content analysis was then conducted. First,\n1 researcher extracted all statements related to the 3 study topics from each\ntranscript (eg, the role of hematologists, the role of hematologists in\nmultidisciplinary collaboration, and the expectations of other professionals in the\ncontext of providing decision-making support). We then carefully conceptualized the\ntext and categorized it into content areas through content analysis using the Klaus\nKrippendorff12 method. The units, codes, and categories were decided\nthrough consensus. Next, another experienced qualitative researcher read the\noriginal interviews and classified them into categories and subcategories. The coder\nread the original interview text and independently determined the subcategory into\nwhich the content fell. The rate of agreement between the researchers and coders was\n91%. Finally, the coder and the researcher discussed the units they disagreed on\nuntil they reached a consensus.", "[SUBTITLE] Participant Characteristics [SUBSECTION] We conducted semi-structured interviews involving 11 hematologists. The average\nparticipant age was 44 years (range: 35-54 years), whereas the average\nspecialization experience was 16 years (range: 7-26 years). Additional details\nare listed in Table\n1. The average length of the interviews was 27 minutes (range: 17-46\nminutes).Table\n1.Characteristics of doctors and interview\ndetails.HematologistsTotaln\n= 11Gender—Male10Female1Workplace—University hospital7Cancer center2General\nhospital2Age (years)43.9 (34-54)*Experience in specialty (years)16.1\n(7-26)*∗Mean(Range).\nCharacteristics of doctors and interview\ndetails.\n∗Mean(Range).\nWe conducted semi-structured interviews involving 11 hematologists. The average\nparticipant age was 44 years (range: 35-54 years), whereas the average\nspecialization experience was 16 years (range: 7-26 years). Additional details\nare listed in Table\n1. The average length of the interviews was 27 minutes (range: 17-46\nminutes).Table\n1.Characteristics of doctors and interview\ndetails.HematologistsTotaln\n= 11Gender—Male10Female1Workplace—University hospital7Cancer center2General\nhospital2Age (years)43.9 (34-54)*Experience in specialty (years)16.1\n(7-26)*∗Mean(Range).\nCharacteristics of doctors and interview\ndetails.\n∗Mean(Range).\n[SUBTITLE] The Role of Hematologists in Direct Decision Support [SUBSECTION] As shown in Table 2,\nwe identified 7 categories related to hematologists’ roles in providing direct\ndecision support to patients: (1) preparing patients before informed consent,\n(2) selecting the information to convey, (3) choosing a method for conveying\nthis information, (4) respecting the intentions of patients and their families,\n(5) directing decision-making and considering fairness, (6) considering the\nemotional aspects of patients and their families, and (7) providing support\nafter discussing treatment options.Table 2.Role of hematologists in providing\ndirect decision support (n =\n11).CategorySubcategoryCoden1Preparing patients before\nIC*(1)Gathering preliminary\ninformation for explaining issues to patientsObtain information from family members to\nunderstand the patient’s preferences in advance3Check if home care is possible1Understand the\nfamily background1Talk to the family about\nwhat you will tell the patient1(2)Encouraging family\nmembers to participate in decision-makingSet up a place to provide information to\npatients, including family members3Discuss\nlife-prolonging treatments with the patient’s\nfamily1(3)Creating an environment for concentrating on\nICDiscuss in a private\nroom2Take sufficient time1Leave the\nPHS*2 with\nthe nurse so that the discussion is not\ndisturbed1(4)Providing advance information on the risk\nof relapseWhen the patient reaches\ncomplete remission, explain the risk of relapse and\nprepare the patient for relapse12Selecting the\ninformation to convey(1)Choice of\ninformation to provide to patientsIn many cases, do not give the patient a\nprognosis9Depending on the\npatient’s medical condition, I (doctor) may not tell the\ntruth5Modify the message according to the\npatient’s life stage3(2)Providing patients and families\nwith information to help them make treatment\ndecisionsCommunicate the\nadvantages and disadvantages of treatment\noptions9Tell patients and their families the\ninformation they need to make treatment\ndecisions6Inform patients of all\ntreatment options4Communicate not only the\nbad news, but also ways of improvement3Also convey the option of non-treatment3(3)Providing patients and families with information to\nhelp them understand the patients' current\nsituationInform family members\nabout the possibility of sudden changes in the medical\ncondition and specific prognosis9Provide\npatients with information related to what they imagine\nwill happen in the future5Provide patients with\ninformation that will help them understand their current\nmedical condition4Provide patients with a\nvague idea of their prognosis3Communicate the\nprognosis to patients using concrete numbers23Choosing\nhow to communicate information(1)Explanations based on the patients' stage of\nreadinessJudge what I (doctor) say\nand how I (doctor) say it based on long-standing\nrelationship with the patients5Provide\ninformation based on the patient’s reaction and\nacceptance of their medical condition5(2)Respect\nfor the preferences of patients and their\nfamiliesProvide easy-to-understand\nexplanations without using technical terms5Provide explanations so that patients/families do not\ndevelop misconceptions4Convey information\ncarefully and clearly3Provide a written\nexplanation2Divide bad news into\ncomponents14Respect for the intentions of\npatients and families(1)Respect for the\nwill of patients and familiesRespect and support the patient’s will and desired\ntreatment6If the patient’s\ncondition is severe, respect the views of the family and\nprimary caregiver2(2)Setting goals in accordance\nwith the preferences of the patients and\nfamiliesListen to the needs of\npatients and their families and determine actionable\ntreatment goals3Explore the intentions of\npatients and their families through\nconversations1If there is no\ndifference between the advantages and disadvantages of a\ntreatment choice, the choice is purely made by the\npatient15Directing decision-making and considering\nfairness(1)Considering the direction and\nfairness of treatment options recommended by\ndoctorsExplain treatment options\nwith some direction while also providing options that\nthe doctor finds beneficial8In some cases,\noffer personal opinion as a doctor2Dare to treat\nthe patient with paternalism2(2)Consideration for\npatients to make impartial decisionsCommunicate with the knowledge that the\ndoctor’s manner of speaking influences the patients’\ndecisions5Explain the opinions of\nother doctors and the second opinion system16Considering the emotional aspects of the patient and\nfamily(1)Explaining with consideration\nfor emotionsTalk in a way that\nallows the patient to have hope5Convey bad news\naccording to the protocol1Consider that patients\ndo not have to listen to explanations while always\nlooking at bad information1Divulge bad news in\nstages1(2)Showing a supportive attitude to the\npatientApproach the patient with a\nnon-abandoning attitude2Show that the team\nsupports the patient1(3)Attitude when facing patients\nand their familiesInteract with\npatients on an equal footing2Talk while\nlooking into the patient’s eyes1Interact with\nthe patient’s family on an equal footing17Supporting after discussing treatments(1)Emotional support after discussing\ntreatmentsSupport fluctuations in\nthe patient’s mind after discussing treatments2Call out the family and assess their reactions1*1IC\n= informed consent; *2PHS = Personal Handyphone\nSystem (A phone used by doctors to communicate with staff in the\nhospital).\nRole of hematologists in providing\ndirect decision support (n =\n11).\n*1IC\n= informed consent; *2PHS = Personal Handyphone\nSystem (A phone used by doctors to communicate with staff in the\nhospital).\n[SUBTITLE] Preparing Patients Before Informed Consent (IC) [SUBSECTION] This category comprised 4 subcategories. Several participants talked about\npreparing patients and families for IC discussions, including “Gathering\npreliminary information for explaining issues to patients” and “Encouraging\nfamily members to participate in decision-making.” As such, physicians were\nexpected to prepare patients before IC discussions. For instance, they would\nprovide patients with information regarding the risk of relapse.\nThis category comprised 4 subcategories. Several participants talked about\npreparing patients and families for IC discussions, including “Gathering\npreliminary information for explaining issues to patients” and “Encouraging\nfamily members to participate in decision-making.” As such, physicians were\nexpected to prepare patients before IC discussions. For instance, they would\nprovide patients with information regarding the risk of relapse.\n[SUBTITLE] Selecting the Information To Convey [SUBSECTION] This category comprised 3 subcategories. In subcategory (1) “Choice of\ninformation to provide to patients,” more than half of the participants said\nthat they did not provide their patients with prognoses. More importantly,\nmore than half said they may not even tell their patients the truth,\ndepending on the severity of the illness, to avoid a loss of hope; they were\nvery selective about the information they chose to provide to their\npatients.\nIn subcategory (2) “Providing patients and families with information to help\nthem make treatment decisions,” more than half of the participants\nidentified “Communicating the advantages and disadvantages of treatment\noptions” and “Telling patients and their families the information they need\nto make treatment decisions.” They provided information they believed would\nhelp patients and their families fully understand all the options before\nmaking a final decision.\nIn subcategory (3), “Providing patients and families with information to help\nthem understand the patients’ current situation,” a large number said they\nwould inform family members about the possibility of sudden changes and\nspecific prognoses, with more than half saying they would explain the issues\nso that patients could visualize what may happen to them in the future.\nGenerally, participants provided information to help patients and their\nfamilies clearly understand their current situations.\nThis category comprised 3 subcategories. In subcategory (1) “Choice of\ninformation to provide to patients,” more than half of the participants said\nthat they did not provide their patients with prognoses. More importantly,\nmore than half said they may not even tell their patients the truth,\ndepending on the severity of the illness, to avoid a loss of hope; they were\nvery selective about the information they chose to provide to their\npatients.\nIn subcategory (2) “Providing patients and families with information to help\nthem make treatment decisions,” more than half of the participants\nidentified “Communicating the advantages and disadvantages of treatment\noptions” and “Telling patients and their families the information they need\nto make treatment decisions.” They provided information they believed would\nhelp patients and their families fully understand all the options before\nmaking a final decision.\nIn subcategory (3), “Providing patients and families with information to help\nthem understand the patients’ current situation,” a large number said they\nwould inform family members about the possibility of sudden changes and\nspecific prognoses, with more than half saying they would explain the issues\nso that patients could visualize what may happen to them in the future.\nGenerally, participants provided information to help patients and their\nfamilies clearly understand their current situations.\n[SUBTITLE] Choosing the Way In Which To Communicate Information [SUBSECTION] This category comprised 2 subcategories. Nearly half of participants\nexpressed the ideas of “I determine what I say and how I say it based on our\nlong-standing relationship with the patients” and “Providing information\nbased on the patient’s reaction and acceptance of their medical condition.”\nThey carefully considered what they discussed and how they discussed it,\nespecially considering their long-standing association with their patients.\nIn this regard, they conveyed information based on how they thought patients\nwould react and their level of acceptance. They also provided explanations\naccording to what they believed patients intended and thought depending on\ntheir respective stages of readiness. Moreover, nearly half said that they\nwould explain issues without using jargon, thereby ensuring that the\ninformation provided was easy to understand. Essentially, participants\nremained cognizant of the need to communicate both correctly and\nappropriately with patients and their families.\nThis category comprised 2 subcategories. Nearly half of participants\nexpressed the ideas of “I determine what I say and how I say it based on our\nlong-standing relationship with the patients” and “Providing information\nbased on the patient’s reaction and acceptance of their medical condition.”\nThey carefully considered what they discussed and how they discussed it,\nespecially considering their long-standing association with their patients.\nIn this regard, they conveyed information based on how they thought patients\nwould react and their level of acceptance. They also provided explanations\naccording to what they believed patients intended and thought depending on\ntheir respective stages of readiness. Moreover, nearly half said that they\nwould explain issues without using jargon, thereby ensuring that the\ninformation provided was easy to understand. Essentially, participants\nremained cognizant of the need to communicate both correctly and\nappropriately with patients and their families.\n[SUBTITLE] Respect for The Intentions Of Patients And Families [SUBSECTION] This category comprised 2 subcategories. In subcategory (1) “Respect for the\nwill of patients and families,” more than half of the participants said they\nhelped patients and families make decisions by supporting and respecting\ntheir wishes and desired treatments. In subcategory (2) “Setting goals in\naccordance with the preferences of the patients and families,” several\nparticipants said that they helped patients and families set feasible goals\nafter listening to their wishes.\nThis category comprised 2 subcategories. In subcategory (1) “Respect for the\nwill of patients and families,” more than half of the participants said they\nhelped patients and families make decisions by supporting and respecting\ntheir wishes and desired treatments. In subcategory (2) “Setting goals in\naccordance with the preferences of the patients and families,” several\nparticipants said that they helped patients and families set feasible goals\nafter listening to their wishes.\n[SUBTITLE] Directing decision-making and considering fairness [SUBSECTION] This category comprised 2 subcategories. In subcategory (1) “Considering the\ndirection and fairness of treatment options recommended by doctors,” more\nthan half of the participants said they explain treatment options with some\ndirection while also providing options they find most beneficial. They\ntalked about giving weight to treatment options and facilitating the\ndecision-making process. Furthermore, they attempted to present information\nin a fair manner, being aware that the way they talk can affect the way\npatients perceive their treatment, as shown in subcategory (2).\nThis category comprised 2 subcategories. In subcategory (1) “Considering the\ndirection and fairness of treatment options recommended by doctors,” more\nthan half of the participants said they explain treatment options with some\ndirection while also providing options they find most beneficial. They\ntalked about giving weight to treatment options and facilitating the\ndecision-making process. Furthermore, they attempted to present information\nin a fair manner, being aware that the way they talk can affect the way\npatients perceive their treatment, as shown in subcategory (2).\n[SUBTITLE] Considering the Emotional Aspects Of The Patient And Family [SUBSECTION] This category comprised 3 subcategories. In subcategory (1)“explaining with\nconsideration for emotions,” nearly half of the participants said they would\nattempt to instill hope among patients, and hence, provide explanations\nconsidering the related emotional aspects. In subcategory (2) “showing a\nsupportive attitude to the patient”, several participants also expressed\nadopting a supportive stance to reassure patients that they were not\nabandoning them or their families. In subcategory (3) “attitude when facing\npatients and their families”, several participants talked about the doctor’s\nattitude toward patients, such as speaking with patients and families as\nequals and looking them in the eye.\nThis category comprised 3 subcategories. In subcategory (1)“explaining with\nconsideration for emotions,” nearly half of the participants said they would\nattempt to instill hope among patients, and hence, provide explanations\nconsidering the related emotional aspects. In subcategory (2) “showing a\nsupportive attitude to the patient”, several participants also expressed\nadopting a supportive stance to reassure patients that they were not\nabandoning them or their families. In subcategory (3) “attitude when facing\npatients and their families”, several participants talked about the doctor’s\nattitude toward patients, such as speaking with patients and families as\nequals and looking them in the eye.\n[SUBTITLE] Providing support after discussing treatments [SUBSECTION] Several participants said they informed patients about bad news and future\ntreatments, after which they provided emotional support in consideration of\nany emotional variabilities.\nSeveral participants said they informed patients about bad news and future\ntreatments, after which they provided emotional support in consideration of\nany emotional variabilities.\nAs shown in Table 2,\nwe identified 7 categories related to hematologists’ roles in providing direct\ndecision support to patients: (1) preparing patients before informed consent,\n(2) selecting the information to convey, (3) choosing a method for conveying\nthis information, (4) respecting the intentions of patients and their families,\n(5) directing decision-making and considering fairness, (6) considering the\nemotional aspects of patients and their families, and (7) providing support\nafter discussing treatment options.Table 2.Role of hematologists in providing\ndirect decision support (n =\n11).CategorySubcategoryCoden1Preparing patients before\nIC*(1)Gathering preliminary\ninformation for explaining issues to patientsObtain information from family members to\nunderstand the patient’s preferences in advance3Check if home care is possible1Understand the\nfamily background1Talk to the family about\nwhat you will tell the patient1(2)Encouraging family\nmembers to participate in decision-makingSet up a place to provide information to\npatients, including family members3Discuss\nlife-prolonging treatments with the patient’s\nfamily1(3)Creating an environment for concentrating on\nICDiscuss in a private\nroom2Take sufficient time1Leave the\nPHS*2 with\nthe nurse so that the discussion is not\ndisturbed1(4)Providing advance information on the risk\nof relapseWhen the patient reaches\ncomplete remission, explain the risk of relapse and\nprepare the patient for relapse12Selecting the\ninformation to convey(1)Choice of\ninformation to provide to patientsIn many cases, do not give the patient a\nprognosis9Depending on the\npatient’s medical condition, I (doctor) may not tell the\ntruth5Modify the message according to the\npatient’s life stage3(2)Providing patients and families\nwith information to help them make treatment\ndecisionsCommunicate the\nadvantages and disadvantages of treatment\noptions9Tell patients and their families the\ninformation they need to make treatment\ndecisions6Inform patients of all\ntreatment options4Communicate not only the\nbad news, but also ways of improvement3Also convey the option of non-treatment3(3)Providing patients and families with information to\nhelp them understand the patients' current\nsituationInform family members\nabout the possibility of sudden changes in the medical\ncondition and specific prognosis9Provide\npatients with information related to what they imagine\nwill happen in the future5Provide patients with\ninformation that will help them understand their current\nmedical condition4Provide patients with a\nvague idea of their prognosis3Communicate the\nprognosis to patients using concrete numbers23Choosing\nhow to communicate information(1)Explanations based on the patients' stage of\nreadinessJudge what I (doctor) say\nand how I (doctor) say it based on long-standing\nrelationship with the patients5Provide\ninformation based on the patient’s reaction and\nacceptance of their medical condition5(2)Respect\nfor the preferences of patients and their\nfamiliesProvide easy-to-understand\nexplanations without using technical terms5Provide explanations so that patients/families do not\ndevelop misconceptions4Convey information\ncarefully and clearly3Provide a written\nexplanation2Divide bad news into\ncomponents14Respect for the intentions of\npatients and families(1)Respect for the\nwill of patients and familiesRespect and support the patient’s will and desired\ntreatment6If the patient’s\ncondition is severe, respect the views of the family and\nprimary caregiver2(2)Setting goals in accordance\nwith the preferences of the patients and\nfamiliesListen to the needs of\npatients and their families and determine actionable\ntreatment goals3Explore the intentions of\npatients and their families through\nconversations1If there is no\ndifference between the advantages and disadvantages of a\ntreatment choice, the choice is purely made by the\npatient15Directing decision-making and considering\nfairness(1)Considering the direction and\nfairness of treatment options recommended by\ndoctorsExplain treatment options\nwith some direction while also providing options that\nthe doctor finds beneficial8In some cases,\noffer personal opinion as a doctor2Dare to treat\nthe patient with paternalism2(2)Consideration for\npatients to make impartial decisionsCommunicate with the knowledge that the\ndoctor’s manner of speaking influences the patients’\ndecisions5Explain the opinions of\nother doctors and the second opinion system16Considering the emotional aspects of the patient and\nfamily(1)Explaining with consideration\nfor emotionsTalk in a way that\nallows the patient to have hope5Convey bad news\naccording to the protocol1Consider that patients\ndo not have to listen to explanations while always\nlooking at bad information1Divulge bad news in\nstages1(2)Showing a supportive attitude to the\npatientApproach the patient with a\nnon-abandoning attitude2Show that the team\nsupports the patient1(3)Attitude when facing patients\nand their familiesInteract with\npatients on an equal footing2Talk while\nlooking into the patient’s eyes1Interact with\nthe patient’s family on an equal footing17Supporting after discussing treatments(1)Emotional support after discussing\ntreatmentsSupport fluctuations in\nthe patient’s mind after discussing treatments2Call out the family and assess their reactions1*1IC\n= informed consent; *2PHS = Personal Handyphone\nSystem (A phone used by doctors to communicate with staff in the\nhospital).\nRole of hematologists in providing\ndirect decision support (n =\n11).\n*1IC\n= informed consent; *2PHS = Personal Handyphone\nSystem (A phone used by doctors to communicate with staff in the\nhospital).\n[SUBTITLE] Preparing Patients Before Informed Consent (IC) [SUBSECTION] This category comprised 4 subcategories. Several participants talked about\npreparing patients and families for IC discussions, including “Gathering\npreliminary information for explaining issues to patients” and “Encouraging\nfamily members to participate in decision-making.” As such, physicians were\nexpected to prepare patients before IC discussions. For instance, they would\nprovide patients with information regarding the risk of relapse.\nThis category comprised 4 subcategories. Several participants talked about\npreparing patients and families for IC discussions, including “Gathering\npreliminary information for explaining issues to patients” and “Encouraging\nfamily members to participate in decision-making.” As such, physicians were\nexpected to prepare patients before IC discussions. For instance, they would\nprovide patients with information regarding the risk of relapse.\n[SUBTITLE] Selecting the Information To Convey [SUBSECTION] This category comprised 3 subcategories. In subcategory (1) “Choice of\ninformation to provide to patients,” more than half of the participants said\nthat they did not provide their patients with prognoses. More importantly,\nmore than half said they may not even tell their patients the truth,\ndepending on the severity of the illness, to avoid a loss of hope; they were\nvery selective about the information they chose to provide to their\npatients.\nIn subcategory (2) “Providing patients and families with information to help\nthem make treatment decisions,” more than half of the participants\nidentified “Communicating the advantages and disadvantages of treatment\noptions” and “Telling patients and their families the information they need\nto make treatment decisions.” They provided information they believed would\nhelp patients and their families fully understand all the options before\nmaking a final decision.\nIn subcategory (3), “Providing patients and families with information to help\nthem understand the patients’ current situation,” a large number said they\nwould inform family members about the possibility of sudden changes and\nspecific prognoses, with more than half saying they would explain the issues\nso that patients could visualize what may happen to them in the future.\nGenerally, participants provided information to help patients and their\nfamilies clearly understand their current situations.\nThis category comprised 3 subcategories. In subcategory (1) “Choice of\ninformation to provide to patients,” more than half of the participants said\nthat they did not provide their patients with prognoses. More importantly,\nmore than half said they may not even tell their patients the truth,\ndepending on the severity of the illness, to avoid a loss of hope; they were\nvery selective about the information they chose to provide to their\npatients.\nIn subcategory (2) “Providing patients and families with information to help\nthem make treatment decisions,” more than half of the participants\nidentified “Communicating the advantages and disadvantages of treatment\noptions” and “Telling patients and their families the information they need\nto make treatment decisions.” They provided information they believed would\nhelp patients and their families fully understand all the options before\nmaking a final decision.\nIn subcategory (3), “Providing patients and families with information to help\nthem understand the patients’ current situation,” a large number said they\nwould inform family members about the possibility of sudden changes and\nspecific prognoses, with more than half saying they would explain the issues\nso that patients could visualize what may happen to them in the future.\nGenerally, participants provided information to help patients and their\nfamilies clearly understand their current situations.\n[SUBTITLE] Choosing the Way In Which To Communicate Information [SUBSECTION] This category comprised 2 subcategories. Nearly half of participants\nexpressed the ideas of “I determine what I say and how I say it based on our\nlong-standing relationship with the patients” and “Providing information\nbased on the patient’s reaction and acceptance of their medical condition.”\nThey carefully considered what they discussed and how they discussed it,\nespecially considering their long-standing association with their patients.\nIn this regard, they conveyed information based on how they thought patients\nwould react and their level of acceptance. They also provided explanations\naccording to what they believed patients intended and thought depending on\ntheir respective stages of readiness. Moreover, nearly half said that they\nwould explain issues without using jargon, thereby ensuring that the\ninformation provided was easy to understand. Essentially, participants\nremained cognizant of the need to communicate both correctly and\nappropriately with patients and their families.\nThis category comprised 2 subcategories. Nearly half of participants\nexpressed the ideas of “I determine what I say and how I say it based on our\nlong-standing relationship with the patients” and “Providing information\nbased on the patient’s reaction and acceptance of their medical condition.”\nThey carefully considered what they discussed and how they discussed it,\nespecially considering their long-standing association with their patients.\nIn this regard, they conveyed information based on how they thought patients\nwould react and their level of acceptance. They also provided explanations\naccording to what they believed patients intended and thought depending on\ntheir respective stages of readiness. Moreover, nearly half said that they\nwould explain issues without using jargon, thereby ensuring that the\ninformation provided was easy to understand. Essentially, participants\nremained cognizant of the need to communicate both correctly and\nappropriately with patients and their families.\n[SUBTITLE] Respect for The Intentions Of Patients And Families [SUBSECTION] This category comprised 2 subcategories. In subcategory (1) “Respect for the\nwill of patients and families,” more than half of the participants said they\nhelped patients and families make decisions by supporting and respecting\ntheir wishes and desired treatments. In subcategory (2) “Setting goals in\naccordance with the preferences of the patients and families,” several\nparticipants said that they helped patients and families set feasible goals\nafter listening to their wishes.\nThis category comprised 2 subcategories. In subcategory (1) “Respect for the\nwill of patients and families,” more than half of the participants said they\nhelped patients and families make decisions by supporting and respecting\ntheir wishes and desired treatments. In subcategory (2) “Setting goals in\naccordance with the preferences of the patients and families,” several\nparticipants said that they helped patients and families set feasible goals\nafter listening to their wishes.\n[SUBTITLE] Directing decision-making and considering fairness [SUBSECTION] This category comprised 2 subcategories. In subcategory (1) “Considering the\ndirection and fairness of treatment options recommended by doctors,” more\nthan half of the participants said they explain treatment options with some\ndirection while also providing options they find most beneficial. They\ntalked about giving weight to treatment options and facilitating the\ndecision-making process. Furthermore, they attempted to present information\nin a fair manner, being aware that the way they talk can affect the way\npatients perceive their treatment, as shown in subcategory (2).\nThis category comprised 2 subcategories. In subcategory (1) “Considering the\ndirection and fairness of treatment options recommended by doctors,” more\nthan half of the participants said they explain treatment options with some\ndirection while also providing options they find most beneficial. They\ntalked about giving weight to treatment options and facilitating the\ndecision-making process. Furthermore, they attempted to present information\nin a fair manner, being aware that the way they talk can affect the way\npatients perceive their treatment, as shown in subcategory (2).\n[SUBTITLE] Considering the Emotional Aspects Of The Patient And Family [SUBSECTION] This category comprised 3 subcategories. In subcategory (1)“explaining with\nconsideration for emotions,” nearly half of the participants said they would\nattempt to instill hope among patients, and hence, provide explanations\nconsidering the related emotional aspects. In subcategory (2) “showing a\nsupportive attitude to the patient”, several participants also expressed\nadopting a supportive stance to reassure patients that they were not\nabandoning them or their families. In subcategory (3) “attitude when facing\npatients and their families”, several participants talked about the doctor’s\nattitude toward patients, such as speaking with patients and families as\nequals and looking them in the eye.\nThis category comprised 3 subcategories. In subcategory (1)“explaining with\nconsideration for emotions,” nearly half of the participants said they would\nattempt to instill hope among patients, and hence, provide explanations\nconsidering the related emotional aspects. In subcategory (2) “showing a\nsupportive attitude to the patient”, several participants also expressed\nadopting a supportive stance to reassure patients that they were not\nabandoning them or their families. In subcategory (3) “attitude when facing\npatients and their families”, several participants talked about the doctor’s\nattitude toward patients, such as speaking with patients and families as\nequals and looking them in the eye.\n[SUBTITLE] Providing support after discussing treatments [SUBSECTION] Several participants said they informed patients about bad news and future\ntreatments, after which they provided emotional support in consideration of\nany emotional variabilities.\nSeveral participants said they informed patients about bad news and future\ntreatments, after which they provided emotional support in consideration of\nany emotional variabilities.\n[SUBTITLE] Role of Hematologists in Multidisciplinary Collaboration and Providing\nDecision Support [SUBSECTION] We identified the following 5 subcategories related to the second main study\ntopic—the roles of hematologists in multidisciplinary collaboration: (1)\ncommunicating with other professionals, (2) gathering information from them, (3)\nproviding information to them, (4) managing the entire medical team, and (5)\nencouraging nurses to actively participate with patients throughout the\ndecision-making process (Table 3). More than half of the participants recognized the\nhematologist’s role in communicating with other professionals and obtaining\ninformation about the patient (eg, the patient’s background and how to help)\nthroughout this process. Others said that they actively encourage nurses to\nsupport the patients’ decisions and provide additional information during the\ndiscussions or explain the prognoses and future treatment plans to other\nprofessionals.Table\n3.The role of hematologists in multidisciplinary\ncollaboration and decision support (n =\n11).SubcategoryCode1Communicating with other professionalsValue good communication with other\nprofessionals, such as nursesMake opportunities to hold discussions with\nmultiple professionals2Gathering\ninformation from other professionalsObtain information from nurses about family\nbackground and family supportObtain information about the patients’\nthoughts and intentions from other\nprofessionalsObtain\ninformation about the patients’ social background from\nother professionalsObtain\ninformation about the patient’s ADL status from other\nprofessionals3Providing\ninformation to other professionalsExplain the patient’s prognosis and future treatment\nplans to other professionalsExplain the concerns of the patient’s\nfamily to other professionals4Managing the entire medical teamManage the medical team, in which\nhematologists make comprehensive decisions based on\ninformation from other professionalsCreate a support system for\npatients5Encouraging nurses to actively\nparticipate in the patient’s decision-makingEncourage nurses to participate in the\npatients’ decision-makingConfirm whether there is any additional information or\nthoughts to convey to nurses during discussions about\ntreatment decisions with the\npatientADL\n= Activities of Daily\nLiving.\nThe role of hematologists in multidisciplinary\ncollaboration and decision support (n =\n11).\nADL\n= Activities of Daily\nLiving.\nWe identified the following 5 subcategories related to the second main study\ntopic—the roles of hematologists in multidisciplinary collaboration: (1)\ncommunicating with other professionals, (2) gathering information from them, (3)\nproviding information to them, (4) managing the entire medical team, and (5)\nencouraging nurses to actively participate with patients throughout the\ndecision-making process (Table 3). More than half of the participants recognized the\nhematologist’s role in communicating with other professionals and obtaining\ninformation about the patient (eg, the patient’s background and how to help)\nthroughout this process. Others said that they actively encourage nurses to\nsupport the patients’ decisions and provide additional information during the\ndiscussions or explain the prognoses and future treatment plans to other\nprofessionals.Table\n3.The role of hematologists in multidisciplinary\ncollaboration and decision support (n =\n11).SubcategoryCode1Communicating with other professionalsValue good communication with other\nprofessionals, such as nursesMake opportunities to hold discussions with\nmultiple professionals2Gathering\ninformation from other professionalsObtain information from nurses about family\nbackground and family supportObtain information about the patients’\nthoughts and intentions from other\nprofessionalsObtain\ninformation about the patients’ social background from\nother professionalsObtain\ninformation about the patient’s ADL status from other\nprofessionals3Providing\ninformation to other professionalsExplain the patient’s prognosis and future treatment\nplans to other professionalsExplain the concerns of the patient’s\nfamily to other professionals4Managing the entire medical teamManage the medical team, in which\nhematologists make comprehensive decisions based on\ninformation from other professionalsCreate a support system for\npatients5Encouraging nurses to actively\nparticipate in the patient’s decision-makingEncourage nurses to participate in the\npatients’ decision-makingConfirm whether there is any additional information or\nthoughts to convey to nurses during discussions about\ntreatment decisions with the\npatientADL\n= Activities of Daily\nLiving.\nThe role of hematologists in multidisciplinary\ncollaboration and decision support (n =\n11).\nADL\n= Activities of Daily\nLiving.\n[SUBTITLE] Expected Roles of Other Professionals in Providing Decision Support [SUBSECTION] We extracted the following 4 categories related to the roles that hematologists\nexpected other professionals to fulfill: (1) sharing patient information and\ncare routines, (2) sharing information from treatment discussions, (3) providing\nemotional support to patients and their families, and (4) assembling a medical\nteam that understands hematological malignancies (Table 4). Nearly half of the\nparticipants indicated that they would like other health care providers to\nconvey the patients’ thoughts and intentions that they knew to the doctors\ninvolved. They also expected other professionals to share information regarding\ntheir patients and the way in which they cared for their patients.Table 4.Expected\nroles of other professionals in decision support (n =\n11).SubcategoryCodenSharing patient information and care\nroutinesListen to the patients’\nthoughts and wishes and communicate them to the\ndoctor5Suggest knowledge that doctors may not know\nand explain how to care for patients in the right\nway5Point out the problems patients have with treatment\ndecisions2Sharing information from the\ntreatment discussionAttend\ntreatment discussions and understand the patients’\nfeelings and situation together4Read records of the\nprocess of informed consent and share the\ninformation1Providing patients and their families with\nemotional supportSupport the\nemotions of patients and their families4Assembling\na medical team that understands hematological\nmalignancyWork to ensure that the\nmedical team understands hematological\nmalignancy1\nExpected\nroles of other professionals in decision support (n =\n11).\nWe extracted the following 4 categories related to the roles that hematologists\nexpected other professionals to fulfill: (1) sharing patient information and\ncare routines, (2) sharing information from treatment discussions, (3) providing\nemotional support to patients and their families, and (4) assembling a medical\nteam that understands hematological malignancies (Table 4). Nearly half of the\nparticipants indicated that they would like other health care providers to\nconvey the patients’ thoughts and intentions that they knew to the doctors\ninvolved. They also expected other professionals to share information regarding\ntheir patients and the way in which they cared for their patients.Table 4.Expected\nroles of other professionals in decision support (n =\n11).SubcategoryCodenSharing patient information and care\nroutinesListen to the patients’\nthoughts and wishes and communicate them to the\ndoctor5Suggest knowledge that doctors may not know\nand explain how to care for patients in the right\nway5Point out the problems patients have with treatment\ndecisions2Sharing information from the\ntreatment discussionAttend\ntreatment discussions and understand the patients’\nfeelings and situation together4Read records of the\nprocess of informed consent and share the\ninformation1Providing patients and their families with\nemotional supportSupport the\nemotions of patients and their families4Assembling\na medical team that understands hematological\nmalignancyWork to ensure that the\nmedical team understands hematological\nmalignancy1\nExpected\nroles of other professionals in decision support (n =\n11).", "We conducted semi-structured interviews involving 11 hematologists. The average\nparticipant age was 44 years (range: 35-54 years), whereas the average\nspecialization experience was 16 years (range: 7-26 years). Additional details\nare listed in Table\n1. The average length of the interviews was 27 minutes (range: 17-46\nminutes).Table\n1.Characteristics of doctors and interview\ndetails.HematologistsTotaln\n= 11Gender—Male10Female1Workplace—University hospital7Cancer center2General\nhospital2Age (years)43.9 (34-54)*Experience in specialty (years)16.1\n(7-26)*∗Mean(Range).\nCharacteristics of doctors and interview\ndetails.\n∗Mean(Range).", "As shown in Table 2,\nwe identified 7 categories related to hematologists’ roles in providing direct\ndecision support to patients: (1) preparing patients before informed consent,\n(2) selecting the information to convey, (3) choosing a method for conveying\nthis information, (4) respecting the intentions of patients and their families,\n(5) directing decision-making and considering fairness, (6) considering the\nemotional aspects of patients and their families, and (7) providing support\nafter discussing treatment options.Table 2.Role of hematologists in providing\ndirect decision support (n =\n11).CategorySubcategoryCoden1Preparing patients before\nIC*(1)Gathering preliminary\ninformation for explaining issues to patientsObtain information from family members to\nunderstand the patient’s preferences in advance3Check if home care is possible1Understand the\nfamily background1Talk to the family about\nwhat you will tell the patient1(2)Encouraging family\nmembers to participate in decision-makingSet up a place to provide information to\npatients, including family members3Discuss\nlife-prolonging treatments with the patient’s\nfamily1(3)Creating an environment for concentrating on\nICDiscuss in a private\nroom2Take sufficient time1Leave the\nPHS*2 with\nthe nurse so that the discussion is not\ndisturbed1(4)Providing advance information on the risk\nof relapseWhen the patient reaches\ncomplete remission, explain the risk of relapse and\nprepare the patient for relapse12Selecting the\ninformation to convey(1)Choice of\ninformation to provide to patientsIn many cases, do not give the patient a\nprognosis9Depending on the\npatient’s medical condition, I (doctor) may not tell the\ntruth5Modify the message according to the\npatient’s life stage3(2)Providing patients and families\nwith information to help them make treatment\ndecisionsCommunicate the\nadvantages and disadvantages of treatment\noptions9Tell patients and their families the\ninformation they need to make treatment\ndecisions6Inform patients of all\ntreatment options4Communicate not only the\nbad news, but also ways of improvement3Also convey the option of non-treatment3(3)Providing patients and families with information to\nhelp them understand the patients' current\nsituationInform family members\nabout the possibility of sudden changes in the medical\ncondition and specific prognosis9Provide\npatients with information related to what they imagine\nwill happen in the future5Provide patients with\ninformation that will help them understand their current\nmedical condition4Provide patients with a\nvague idea of their prognosis3Communicate the\nprognosis to patients using concrete numbers23Choosing\nhow to communicate information(1)Explanations based on the patients' stage of\nreadinessJudge what I (doctor) say\nand how I (doctor) say it based on long-standing\nrelationship with the patients5Provide\ninformation based on the patient’s reaction and\nacceptance of their medical condition5(2)Respect\nfor the preferences of patients and their\nfamiliesProvide easy-to-understand\nexplanations without using technical terms5Provide explanations so that patients/families do not\ndevelop misconceptions4Convey information\ncarefully and clearly3Provide a written\nexplanation2Divide bad news into\ncomponents14Respect for the intentions of\npatients and families(1)Respect for the\nwill of patients and familiesRespect and support the patient’s will and desired\ntreatment6If the patient’s\ncondition is severe, respect the views of the family and\nprimary caregiver2(2)Setting goals in accordance\nwith the preferences of the patients and\nfamiliesListen to the needs of\npatients and their families and determine actionable\ntreatment goals3Explore the intentions of\npatients and their families through\nconversations1If there is no\ndifference between the advantages and disadvantages of a\ntreatment choice, the choice is purely made by the\npatient15Directing decision-making and considering\nfairness(1)Considering the direction and\nfairness of treatment options recommended by\ndoctorsExplain treatment options\nwith some direction while also providing options that\nthe doctor finds beneficial8In some cases,\noffer personal opinion as a doctor2Dare to treat\nthe patient with paternalism2(2)Consideration for\npatients to make impartial decisionsCommunicate with the knowledge that the\ndoctor’s manner of speaking influences the patients’\ndecisions5Explain the opinions of\nother doctors and the second opinion system16Considering the emotional aspects of the patient and\nfamily(1)Explaining with consideration\nfor emotionsTalk in a way that\nallows the patient to have hope5Convey bad news\naccording to the protocol1Consider that patients\ndo not have to listen to explanations while always\nlooking at bad information1Divulge bad news in\nstages1(2)Showing a supportive attitude to the\npatientApproach the patient with a\nnon-abandoning attitude2Show that the team\nsupports the patient1(3)Attitude when facing patients\nand their familiesInteract with\npatients on an equal footing2Talk while\nlooking into the patient’s eyes1Interact with\nthe patient’s family on an equal footing17Supporting after discussing treatments(1)Emotional support after discussing\ntreatmentsSupport fluctuations in\nthe patient’s mind after discussing treatments2Call out the family and assess their reactions1*1IC\n= informed consent; *2PHS = Personal Handyphone\nSystem (A phone used by doctors to communicate with staff in the\nhospital).\nRole of hematologists in providing\ndirect decision support (n =\n11).\n*1IC\n= informed consent; *2PHS = Personal Handyphone\nSystem (A phone used by doctors to communicate with staff in the\nhospital).\n[SUBTITLE] Preparing Patients Before Informed Consent (IC) [SUBSECTION] This category comprised 4 subcategories. Several participants talked about\npreparing patients and families for IC discussions, including “Gathering\npreliminary information for explaining issues to patients” and “Encouraging\nfamily members to participate in decision-making.” As such, physicians were\nexpected to prepare patients before IC discussions. For instance, they would\nprovide patients with information regarding the risk of relapse.\nThis category comprised 4 subcategories. Several participants talked about\npreparing patients and families for IC discussions, including “Gathering\npreliminary information for explaining issues to patients” and “Encouraging\nfamily members to participate in decision-making.” As such, physicians were\nexpected to prepare patients before IC discussions. For instance, they would\nprovide patients with information regarding the risk of relapse.\n[SUBTITLE] Selecting the Information To Convey [SUBSECTION] This category comprised 3 subcategories. In subcategory (1) “Choice of\ninformation to provide to patients,” more than half of the participants said\nthat they did not provide their patients with prognoses. More importantly,\nmore than half said they may not even tell their patients the truth,\ndepending on the severity of the illness, to avoid a loss of hope; they were\nvery selective about the information they chose to provide to their\npatients.\nIn subcategory (2) “Providing patients and families with information to help\nthem make treatment decisions,” more than half of the participants\nidentified “Communicating the advantages and disadvantages of treatment\noptions” and “Telling patients and their families the information they need\nto make treatment decisions.” They provided information they believed would\nhelp patients and their families fully understand all the options before\nmaking a final decision.\nIn subcategory (3), “Providing patients and families with information to help\nthem understand the patients’ current situation,” a large number said they\nwould inform family members about the possibility of sudden changes and\nspecific prognoses, with more than half saying they would explain the issues\nso that patients could visualize what may happen to them in the future.\nGenerally, participants provided information to help patients and their\nfamilies clearly understand their current situations.\nThis category comprised 3 subcategories. In subcategory (1) “Choice of\ninformation to provide to patients,” more than half of the participants said\nthat they did not provide their patients with prognoses. More importantly,\nmore than half said they may not even tell their patients the truth,\ndepending on the severity of the illness, to avoid a loss of hope; they were\nvery selective about the information they chose to provide to their\npatients.\nIn subcategory (2) “Providing patients and families with information to help\nthem make treatment decisions,” more than half of the participants\nidentified “Communicating the advantages and disadvantages of treatment\noptions” and “Telling patients and their families the information they need\nto make treatment decisions.” They provided information they believed would\nhelp patients and their families fully understand all the options before\nmaking a final decision.\nIn subcategory (3), “Providing patients and families with information to help\nthem understand the patients’ current situation,” a large number said they\nwould inform family members about the possibility of sudden changes and\nspecific prognoses, with more than half saying they would explain the issues\nso that patients could visualize what may happen to them in the future.\nGenerally, participants provided information to help patients and their\nfamilies clearly understand their current situations.\n[SUBTITLE] Choosing the Way In Which To Communicate Information [SUBSECTION] This category comprised 2 subcategories. Nearly half of participants\nexpressed the ideas of “I determine what I say and how I say it based on our\nlong-standing relationship with the patients” and “Providing information\nbased on the patient’s reaction and acceptance of their medical condition.”\nThey carefully considered what they discussed and how they discussed it,\nespecially considering their long-standing association with their patients.\nIn this regard, they conveyed information based on how they thought patients\nwould react and their level of acceptance. They also provided explanations\naccording to what they believed patients intended and thought depending on\ntheir respective stages of readiness. Moreover, nearly half said that they\nwould explain issues without using jargon, thereby ensuring that the\ninformation provided was easy to understand. Essentially, participants\nremained cognizant of the need to communicate both correctly and\nappropriately with patients and their families.\nThis category comprised 2 subcategories. Nearly half of participants\nexpressed the ideas of “I determine what I say and how I say it based on our\nlong-standing relationship with the patients” and “Providing information\nbased on the patient’s reaction and acceptance of their medical condition.”\nThey carefully considered what they discussed and how they discussed it,\nespecially considering their long-standing association with their patients.\nIn this regard, they conveyed information based on how they thought patients\nwould react and their level of acceptance. They also provided explanations\naccording to what they believed patients intended and thought depending on\ntheir respective stages of readiness. Moreover, nearly half said that they\nwould explain issues without using jargon, thereby ensuring that the\ninformation provided was easy to understand. Essentially, participants\nremained cognizant of the need to communicate both correctly and\nappropriately with patients and their families.\n[SUBTITLE] Respect for The Intentions Of Patients And Families [SUBSECTION] This category comprised 2 subcategories. In subcategory (1) “Respect for the\nwill of patients and families,” more than half of the participants said they\nhelped patients and families make decisions by supporting and respecting\ntheir wishes and desired treatments. In subcategory (2) “Setting goals in\naccordance with the preferences of the patients and families,” several\nparticipants said that they helped patients and families set feasible goals\nafter listening to their wishes.\nThis category comprised 2 subcategories. In subcategory (1) “Respect for the\nwill of patients and families,” more than half of the participants said they\nhelped patients and families make decisions by supporting and respecting\ntheir wishes and desired treatments. In subcategory (2) “Setting goals in\naccordance with the preferences of the patients and families,” several\nparticipants said that they helped patients and families set feasible goals\nafter listening to their wishes.\n[SUBTITLE] Directing decision-making and considering fairness [SUBSECTION] This category comprised 2 subcategories. In subcategory (1) “Considering the\ndirection and fairness of treatment options recommended by doctors,” more\nthan half of the participants said they explain treatment options with some\ndirection while also providing options they find most beneficial. They\ntalked about giving weight to treatment options and facilitating the\ndecision-making process. Furthermore, they attempted to present information\nin a fair manner, being aware that the way they talk can affect the way\npatients perceive their treatment, as shown in subcategory (2).\nThis category comprised 2 subcategories. In subcategory (1) “Considering the\ndirection and fairness of treatment options recommended by doctors,” more\nthan half of the participants said they explain treatment options with some\ndirection while also providing options they find most beneficial. They\ntalked about giving weight to treatment options and facilitating the\ndecision-making process. Furthermore, they attempted to present information\nin a fair manner, being aware that the way they talk can affect the way\npatients perceive their treatment, as shown in subcategory (2).\n[SUBTITLE] Considering the Emotional Aspects Of The Patient And Family [SUBSECTION] This category comprised 3 subcategories. In subcategory (1)“explaining with\nconsideration for emotions,” nearly half of the participants said they would\nattempt to instill hope among patients, and hence, provide explanations\nconsidering the related emotional aspects. In subcategory (2) “showing a\nsupportive attitude to the patient”, several participants also expressed\nadopting a supportive stance to reassure patients that they were not\nabandoning them or their families. In subcategory (3) “attitude when facing\npatients and their families”, several participants talked about the doctor’s\nattitude toward patients, such as speaking with patients and families as\nequals and looking them in the eye.\nThis category comprised 3 subcategories. In subcategory (1)“explaining with\nconsideration for emotions,” nearly half of the participants said they would\nattempt to instill hope among patients, and hence, provide explanations\nconsidering the related emotional aspects. In subcategory (2) “showing a\nsupportive attitude to the patient”, several participants also expressed\nadopting a supportive stance to reassure patients that they were not\nabandoning them or their families. In subcategory (3) “attitude when facing\npatients and their families”, several participants talked about the doctor’s\nattitude toward patients, such as speaking with patients and families as\nequals and looking them in the eye.\n[SUBTITLE] Providing support after discussing treatments [SUBSECTION] Several participants said they informed patients about bad news and future\ntreatments, after which they provided emotional support in consideration of\nany emotional variabilities.\nSeveral participants said they informed patients about bad news and future\ntreatments, after which they provided emotional support in consideration of\nany emotional variabilities.", "This category comprised 4 subcategories. Several participants talked about\npreparing patients and families for IC discussions, including “Gathering\npreliminary information for explaining issues to patients” and “Encouraging\nfamily members to participate in decision-making.” As such, physicians were\nexpected to prepare patients before IC discussions. For instance, they would\nprovide patients with information regarding the risk of relapse.", "This category comprised 3 subcategories. In subcategory (1) “Choice of\ninformation to provide to patients,” more than half of the participants said\nthat they did not provide their patients with prognoses. More importantly,\nmore than half said they may not even tell their patients the truth,\ndepending on the severity of the illness, to avoid a loss of hope; they were\nvery selective about the information they chose to provide to their\npatients.\nIn subcategory (2) “Providing patients and families with information to help\nthem make treatment decisions,” more than half of the participants\nidentified “Communicating the advantages and disadvantages of treatment\noptions” and “Telling patients and their families the information they need\nto make treatment decisions.” They provided information they believed would\nhelp patients and their families fully understand all the options before\nmaking a final decision.\nIn subcategory (3), “Providing patients and families with information to help\nthem understand the patients’ current situation,” a large number said they\nwould inform family members about the possibility of sudden changes and\nspecific prognoses, with more than half saying they would explain the issues\nso that patients could visualize what may happen to them in the future.\nGenerally, participants provided information to help patients and their\nfamilies clearly understand their current situations.", "This category comprised 2 subcategories. Nearly half of participants\nexpressed the ideas of “I determine what I say and how I say it based on our\nlong-standing relationship with the patients” and “Providing information\nbased on the patient’s reaction and acceptance of their medical condition.”\nThey carefully considered what they discussed and how they discussed it,\nespecially considering their long-standing association with their patients.\nIn this regard, they conveyed information based on how they thought patients\nwould react and their level of acceptance. They also provided explanations\naccording to what they believed patients intended and thought depending on\ntheir respective stages of readiness. Moreover, nearly half said that they\nwould explain issues without using jargon, thereby ensuring that the\ninformation provided was easy to understand. Essentially, participants\nremained cognizant of the need to communicate both correctly and\nappropriately with patients and their families.", "This category comprised 2 subcategories. In subcategory (1) “Respect for the\nwill of patients and families,” more than half of the participants said they\nhelped patients and families make decisions by supporting and respecting\ntheir wishes and desired treatments. In subcategory (2) “Setting goals in\naccordance with the preferences of the patients and families,” several\nparticipants said that they helped patients and families set feasible goals\nafter listening to their wishes.", "This category comprised 2 subcategories. In subcategory (1) “Considering the\ndirection and fairness of treatment options recommended by doctors,” more\nthan half of the participants said they explain treatment options with some\ndirection while also providing options they find most beneficial. They\ntalked about giving weight to treatment options and facilitating the\ndecision-making process. Furthermore, they attempted to present information\nin a fair manner, being aware that the way they talk can affect the way\npatients perceive their treatment, as shown in subcategory (2).", "This category comprised 3 subcategories. In subcategory (1)“explaining with\nconsideration for emotions,” nearly half of the participants said they would\nattempt to instill hope among patients, and hence, provide explanations\nconsidering the related emotional aspects. In subcategory (2) “showing a\nsupportive attitude to the patient”, several participants also expressed\nadopting a supportive stance to reassure patients that they were not\nabandoning them or their families. In subcategory (3) “attitude when facing\npatients and their families”, several participants talked about the doctor’s\nattitude toward patients, such as speaking with patients and families as\nequals and looking them in the eye.", "Several participants said they informed patients about bad news and future\ntreatments, after which they provided emotional support in consideration of\nany emotional variabilities.", "We identified the following 5 subcategories related to the second main study\ntopic—the roles of hematologists in multidisciplinary collaboration: (1)\ncommunicating with other professionals, (2) gathering information from them, (3)\nproviding information to them, (4) managing the entire medical team, and (5)\nencouraging nurses to actively participate with patients throughout the\ndecision-making process (Table 3). More than half of the participants recognized the\nhematologist’s role in communicating with other professionals and obtaining\ninformation about the patient (eg, the patient’s background and how to help)\nthroughout this process. Others said that they actively encourage nurses to\nsupport the patients’ decisions and provide additional information during the\ndiscussions or explain the prognoses and future treatment plans to other\nprofessionals.Table\n3.The role of hematologists in multidisciplinary\ncollaboration and decision support (n =\n11).SubcategoryCode1Communicating with other professionalsValue good communication with other\nprofessionals, such as nursesMake opportunities to hold discussions with\nmultiple professionals2Gathering\ninformation from other professionalsObtain information from nurses about family\nbackground and family supportObtain information about the patients’\nthoughts and intentions from other\nprofessionalsObtain\ninformation about the patients’ social background from\nother professionalsObtain\ninformation about the patient’s ADL status from other\nprofessionals3Providing\ninformation to other professionalsExplain the patient’s prognosis and future treatment\nplans to other professionalsExplain the concerns of the patient’s\nfamily to other professionals4Managing the entire medical teamManage the medical team, in which\nhematologists make comprehensive decisions based on\ninformation from other professionalsCreate a support system for\npatients5Encouraging nurses to actively\nparticipate in the patient’s decision-makingEncourage nurses to participate in the\npatients’ decision-makingConfirm whether there is any additional information or\nthoughts to convey to nurses during discussions about\ntreatment decisions with the\npatientADL\n= Activities of Daily\nLiving.\nThe role of hematologists in multidisciplinary\ncollaboration and decision support (n =\n11).\nADL\n= Activities of Daily\nLiving.", "We extracted the following 4 categories related to the roles that hematologists\nexpected other professionals to fulfill: (1) sharing patient information and\ncare routines, (2) sharing information from treatment discussions, (3) providing\nemotional support to patients and their families, and (4) assembling a medical\nteam that understands hematological malignancies (Table 4). Nearly half of the\nparticipants indicated that they would like other health care providers to\nconvey the patients’ thoughts and intentions that they knew to the doctors\ninvolved. They also expected other professionals to share information regarding\ntheir patients and the way in which they cared for their patients.Table 4.Expected\nroles of other professionals in decision support (n =\n11).SubcategoryCodenSharing patient information and care\nroutinesListen to the patients’\nthoughts and wishes and communicate them to the\ndoctor5Suggest knowledge that doctors may not know\nand explain how to care for patients in the right\nway5Point out the problems patients have with treatment\ndecisions2Sharing information from the\ntreatment discussionAttend\ntreatment discussions and understand the patients’\nfeelings and situation together4Read records of the\nprocess of informed consent and share the\ninformation1Providing patients and their families with\nemotional supportSupport the\nemotions of patients and their families4Assembling\na medical team that understands hematological\nmalignancyWork to ensure that the\nmedical team understands hematological\nmalignancy1\nExpected\nroles of other professionals in decision support (n =\n11).", "In this study, hematologists were interviewed to clarify their role and expectations\nof other professionals. The categories of hematologists’ direct role in\ndecision-making were extracted, including preparation and consideration of the\nsituation in which information about decision-making is communicated and emotional\nsupport after the information is communicated.\nHematologists presented treatment options based on a variety of factors, including\nthe patient’s goals, emotions, the preparedness for the disease, and the severity of\nthe disease. During this process, the hematologists worked with other professionals\nto understand specific information about the patient and family, including their\nbackground, goals, and intentions. They also said that they would decide what\ninformation to convey and carefully explain the treatment and medical condition,\nwithout using jargon, so that the patient and family could easily understand. ASCO\nguidelines recommend that physicians provide information that takes into account\nindividual patient interests and preferences.7 Although the present results\nare data obtained before these guidelines were developed, the hematologists who\nparticipated in this study were taking the actions recommended in the guidelines.\nThe present results reveal more specific behaviors of hematologists with regard to\nwhat is recommended in the guidelines. For patients facing complex and risky\ntreatment choices, it is important for hematologist to select information according\nto the patient’s wishes and to convey information in easy-to-understand terms.\nHematologists were expected other professionalsto pay attention to the emotional\naspects of the patient and family to minimize their anxiety. In addition, they\nexpected nurses and other professionals to listen to the patient and family’s wishes\nand thoughts about treatment, share them with the physician, and serve as emotional\nsupport for the patient and family. Moreover, hematologists were expected to\nunderstand the patient’s wishes and preferences, explain complex and\ndifficult-to-understand treatments and medical conditions, and present options that\nwere consistent with the patient’s wishes. ASCO guidelines also emphasize the need\nto form a rapport with patients and their families and to assess and support coping\nneeds.7 The hematologists who participated in this study wanted to\nimplement this in collaboration with other professionals. The wishes and intentions\nof patients and their families change as the disease progresses or symptoms worsen.\nTherefore, it is thought that by having physicians and other professions work\ntogether to understand the wishes of patients/families at that time, decision-making\nsupport in accordance with the wishes of patients/families will become possible.\nLeukemia and lymphoma may be treated through chemotherapy, even in cases of relapse\nand during the refractory phase. This makes it difficult to determine when to\ntransition from curative treatments to those more focused on quality of life\n(QOL).13,14 Previous studies have reported that hematologists are reluctant\nto discuss EOL issues while primary disease treatments are still possible.15,16 A number of\nthe hematologists who participated in the study had a variety of ways of explaining\nthe prognosis to patients and their families. For example, in some cases, they\ninformed the family about the possibility of a sudden change in prognosis, including\nspecific figures, while in other cases, depending on the severity of the disease and\nthe stage of life, they did not inform them of the prognosis at all or withheld the\nfact. Furthermore, more than half of the respondents stated that they would explain\nthe treatment that they thought would be most beneficial to the patient.\nA number of the hematologists who participated in this study also had a long history\nof working with patients to understand their individual backgrounds, and what they\ntalked about and how they communicate, while also having a direction that the\nhematologists thought was suitable, and gave the patients some direction as they\nspoke. This suggested that hematologists attempt to tailor their decision-making\nsupport to the needs of their patients. On the other hand, hematologists infer\npatients' needs based on their previous experience, which runs the risk of making\nsuggestions that are not in line with the patients’ original needs. Previous studies\nhave reported that hematologic oncology patients take a passive role in the\ntreatment decision-making process due to the complexity of their treatment and\ndisease17 and expect their physicians to take a paternalistic\nrole.16 When hematologists provide some treatment direction and\npaternalistic response to hematologic oncology patients with these characteristics,\npatients may not fully understand the advantages and disadvantages of treatment and\nmay not make an informed choice. If treatment is responded to, the patient’s quality\nof life improves, but if treatment is not effective, the patient may die in the\nhospital during active treatment. In addition, patients may not be able to live the\nlife they want, with increased risk of infection and prolonged\nhospitalization.1 Hematologists need to have frank discussions with patients,\nkeeping in mind the direction they think is best for the patient, and working with\nother professionals to understand the patient’s wishes.\nHematologists who participated in this study indicated that the role they expected of\nmultidisciplinary professionals was to inform them about patient thoughts,\ninformation, and methods of care that the physicians were unaware of. They also\nexpected other professions to play a role in providing emotional support to patients\nand their families. In a previous study of oncologists, including hematologists,\nlack of information about the patient was the most frequently cited factor hindering\npatient involvement in decision-making.18 Physicians also recognized\nthat having a third party present during decision-making discussions supported\npatient decision-making and facilitated involvement and reflection on treatment\ndecisions.18 In that previous study, physicians emphasized the\nimportance of obtaining information about the patient and the presence of a third\nparty in the decision-making situation, which was similar to the findings of the\npresent study. This confirms the importance of a multidisciplinary approach to\nfacilitate decision-making in accordance with the patient’s wishes. In making\ntreatment decisions, it is necessary to organize information not only from a medical\nperspective, but also based on the patient’s priorities and values. Obtaining such\ninformation is difficult for hematologists alone, and multidisciplinary\ncollaboration is important. By understanding the patient’s needs and organizing the\ninformation necessary for decision-making, it is expected that the discussion\nbetween the attending hematologist and the patient will be deepened.9\nFinally, it is important to note that the survey for this study was conducted in 2011\nand there was not much collaboration with the palliative care team,15 the\nparticipants in this study did not mention the role they expected to play regarding\npalliative care physicians. Recently, however, the effects of early palliative care\nhave been reported in clinical hematologic oncology. A study evaluating the effects\nof early introduction of palliative care and continuous psychological support by a\nmultidisciplinary team in patients with acute leukemia reported a reduction in\nsymptoms due to acute stress reactions compared to the usual care group.19 It has also\nbeen reported that patients undergoing hematopoietic stem cell transplantation who\nreceived regular twice-weekly visits by a palliative care physician had a higher QOL\nat 3 months of hospitalization compared to the group that received usual\ncare.20 In patients with leukemia and lymphoma, where the potential for\nrapid change is high, it is expected that hematologists and palliative care\nspecialists will support patients and begin discussing long-term goals early, in\ncase treatment goals suddenly change.21\nThis study had some limitations. The first is that the data collection and analysis\nwere conducted between 2011 and 2012. Therefore, because treatment outcomes for\nhematologic tumors have improved compared to a decade ago, the data cannot be\ndirectly applied to decision support for patients with such tumors and their\nfamilies today. However, the patients with treatment-resistant hematologic tumors\nhave not changed significantly in their characteristics or treatment\nenvironment,1,22 and it is significant to clarify the hematologist’s role in\ndecision support. Second, the use of the opportunistic sampling makes it difficult\nto generalize the findings. In this study, we collected the opinions of\nhematologists at hospitals where hematologic oncology patients are mainly treated,\nsuch as cancer hospitals and university hospitals in Japan, and we believe that we\nwere able to collect diverse opinions regarding the role of decision support as\nperceived by hematologists. Based on the present study, research is needed to\nclarify the decision support roles of other professionals who care for patients with\nleukemia and lymphoma, including nurses. This would make it possible to construct a\nmodel of shared decision-making support for hematologic oncology patients through\nmultidisciplinary collaboration.", "Click here for additional data file.\nSupplemental Material for Qualitative Analysis of the Roles of Physicians and\nNurses in Providing Decision Support to Patients With Relapsed or Refractory\nLeukemia and Lymphoma by Miharu Morikawa, and Yuki Shirai in Cancer Control" ]
[ "intro", "methods", "results", null, null, null, null, null, null, null, null, null, null, null, "discussion", "supplementary-material" ]
[ "hematologic diseases", "shared decision-making", "patient care team", "physician’s role", "communication" ]
Defective Desmosomal Adhesion Causes Arrhythmogenic Cardiomyopathy by Involving an Integrin-αVβ6/TGF-β Signaling Cascade.
36268721
Arrhythmogenic cardiomyopathy (ACM) is characterized by progressive loss of cardiomyocytes with fibrofatty tissue replacement, systolic dysfunction, and life-threatening arrhythmias. A substantial proportion of ACM is caused by mutations in genes of the desmosomal cell-cell adhesion complex, but the underlying mechanisms are not well understood. In the current study, we investigated the relevance of defective desmosomal adhesion for ACM development and progression.
BACKGROUND
We mutated the binding site of DSG2 (desmoglein-2), a crucial desmosomal adhesion molecule in cardiomyocytes. This DSG2-W2A mutation abrogates the tryptophan swap, a central interaction mechanism of DSG2 on the basis of structural data. Impaired adhesive function of DSG2-W2A was confirmed by cell-cell dissociation assays and force spectroscopy measurements by atomic force microscopy. The DSG2-W2A knock-in mouse model was analyzed by echocardiography, ECG, and histologic and biomolecular techniques including RNA sequencing and transmission electron and superresolution microscopy. The results were compared with ACM patient samples, and their relevance was confirmed in vivo and in cardiac slice cultures by inhibitor studies applying the small molecule EMD527040 or an inhibitory integrin-αVβ6 antibody.
METHODS
The DSG2-W2A mutation impaired binding on molecular level and compromised intercellular adhesive function. Mice bearing this mutation develop a severe cardiac phenotype recalling the characteristics of ACM, including cardiac fibrosis, impaired systolic function, and arrhythmia. A comparison of the transcriptome of mutant mice with ACM patient data suggested deregulated integrin-αVβ6 and subsequent transforming growth factor-β signaling as driver of cardiac fibrosis. Blocking integrin-αVβ6 led to reduced expression of profibrotic markers and reduced fibrosis formation in mutant animals in vivo.
RESULTS
We show that disruption of desmosomal adhesion is sufficient to induce a phenotype that fulfils the clinical criteria to establish the diagnosis of ACM, confirming the dysfunctional adhesion hypothesis. Deregulation of integrin-αVβ6 and transforming growth factor-β signaling was identified as a central step toward fibrosis. A pilot in vivo drug test revealed this pathway as a promising target to ameliorate fibrosis. This highlights the value of this model to discern mechanisms of cardiac fibrosis and to identify and test novel treatment options for ACM.
CONCLUSIONS
[ "Mice", "Animals", "Cardiomyopathies", "Arrhythmias, Cardiac", "Integrins", "Myocytes, Cardiac", "Fibrosis", "Transforming Growth Factor beta", "Transforming Growth Factors", "Arrhythmogenic Right Ventricular Dysplasia" ]
9674449
null
null
Methods
Detailed methods are available in the Expanded Methods in the Supplemental Material. [SUBTITLE] Human Heart Samples [SUBSECTION] Heart samples of patients with ACM and healthy controls were derived from forensic autopsy. This study was conducted according to the tenets of the Declaration of Helsinki and approved by the local ethics committees (license number 494-16 from LMU Munich and license number 152/15 from Goethe University). Samples were fixed and embedded in paraffin according to standard procedures. Heart samples of patients with ACM and healthy controls were derived from forensic autopsy. This study was conducted according to the tenets of the Declaration of Helsinki and approved by the local ethics committees (license number 494-16 from LMU Munich and license number 152/15 from Goethe University). Samples were fixed and embedded in paraffin according to standard procedures. [SUBTITLE] Animal Experiments [SUBSECTION] Mouse experiments were approved by the Cantonal Veterinary Office of Basel-Stadt (license numbers 2973_32878 and 3070_32419). Mice were housed under specific pathogen-free conditions with standard chow and bedding with a 12-hour day/night cycle according to institutional guidelines. Animals of both sexes were applied without bias. For inhibitor treatments, mice from the same litter and sex were paired and allocated randomly to the groups. Mouse experiments were approved by the Cantonal Veterinary Office of Basel-Stadt (license numbers 2973_32878 and 3070_32419). Mice were housed under specific pathogen-free conditions with standard chow and bedding with a 12-hour day/night cycle according to institutional guidelines. Animals of both sexes were applied without bias. For inhibitor treatments, mice from the same litter and sex were paired and allocated randomly to the groups. [SUBTITLE] Statistics and Data Compilation [SUBSECTION] Figures were compiled with Adobe Photoshop CC 2017 and Adobe Illustrator CC 2017. Statistical computations were performed with Prism 8 (GraphPad Software). For comparison of 2 or multiple groups, distribution of data was analyzed by a Shapiro-Wilk normality test, and group variances were analyzed by an F-test or a Brown-Forsythe test, respectively. According to the results of these tests, a parametric or nonparametric test with or without Welch correction for unequal variances was applied. For Kaplan-Meier survival analysis, significance was assessed by the Gehan-Breslow-Wilcoxon test. The statistical test used to compare the respective data sets is described in the corresponding figure legend. Statistical significance was assumed at P<0.05. Unless otherwise stated, data are presented as dot blot, with each dot representing the mean of the respective technical replicates of 1 biological replicate. Each animal or independent seeding of cells was taken as biological replicate, or as indicated in the legend. The mean value of these dots is shown as bar diagram ± SD. Figures were compiled with Adobe Photoshop CC 2017 and Adobe Illustrator CC 2017. Statistical computations were performed with Prism 8 (GraphPad Software). For comparison of 2 or multiple groups, distribution of data was analyzed by a Shapiro-Wilk normality test, and group variances were analyzed by an F-test or a Brown-Forsythe test, respectively. According to the results of these tests, a parametric or nonparametric test with or without Welch correction for unequal variances was applied. For Kaplan-Meier survival analysis, significance was assessed by the Gehan-Breslow-Wilcoxon test. The statistical test used to compare the respective data sets is described in the corresponding figure legend. Statistical significance was assumed at P<0.05. Unless otherwise stated, data are presented as dot blot, with each dot representing the mean of the respective technical replicates of 1 biological replicate. Each animal or independent seeding of cells was taken as biological replicate, or as indicated in the legend. The mean value of these dots is shown as bar diagram ± SD. [SUBTITLE] Data Availability [SUBSECTION] RNA sequencing data were deposited in the public database GEO ( accession number GSE181868). Additional data pertaining to the current article are available from the corresponding author upon request. RNA sequencing data were deposited in the public database GEO ( accession number GSE181868). Additional data pertaining to the current article are available from the corresponding author upon request.
Results
[SUBTITLE] W2A Mutation Abrogates DSG2 Interactions and Impairs Cell–Cell Adhesion In Vitro and In Vivo [SUBSECTION] To study the functional consequences of impaired DSG2 binding, we generated the W2A point mutation (DSG2-W2A) to abrogate the proposed interaction mechanism of the only desmoglein expressed in cardiomyocytes (Figure 1A). First, we investigated in a cell-free setup whether DSG2-W2A is indeed affecting the binding properties of DSG2 interaction by applying single molecule force spectroscopy. Here, molecules are coupled to the tip of an atomic force microscopy probe and the surface of a mica sheet by a polyethylene glycol linker. By measuring the deflection of the probe during repeated approach to and retraction from the surface, binding events can be assessed quantitatively. From these data, properties such as binding frequency, binding force, and bond half-life time can be determined. Constructs were generated for expression of wild-type (WT) and mutant (W2A) DSG2 extracellular domains fused to a human Fc fragment and tested for homotypic (WT:WT, W2A:W2A) and heterotypic (WT:W2A) interaction properties (Figure 1B). These experiments showed a significant reduction of the frequency of W2A:W2A as well as WT:W2A interactions compared with WT:WT (Figure 1C). The frequency of the remaining W2A bindings was comparable to probing Fc alone, which indicates that these are mostly nonspecific interactions. Homotypic WT interactions display a clear binding force peak, whereas the remaining W2A interaction forces were spread widely, again pointing to the nonspecificity of these interactions (Figure 1D). The lifetime of DSG2 interactions under zero force (τ0) was determined by fitting the binding forces detected at different loading rates,11 yielding τ0=1.088s with R=0.993. In contrast, no sufficient fit (R=0.509) was possible for W2A interactions (Figure 1E). Together, these data outline that the tryptophan swap is the major interaction mechanism of DSG2. DSG2 (desmoglein-2) adhesion is mediated by tryptophan swap at position 2. A, Predicted interaction model of DSG2 extracellular domains by exchange of tryptophan residue at position 2 into a hydrophobic pocket of the opposing molecule. Cartoon 3D presentation of Protein Data Bank entry 5ERD9; tryptophan-2 is highlighted by ball and stick presentation. B, Schematic of single molecule force spectroscopy experiments. Recombinant extracellular domains (ECs) of wild-type DSG2 (DSG2-WT) or mutant DSG2 (DSG2-W2A) protein were coupled to a mica surface and atomic force microscopy cantilever by mean of a human Fc-tag (hFc) and a polyethylene glycol (PEG)20 linker and probed as indicated. C, Binding frequency of DSG2-W2A/DSG2-WT heterotypic and homotypic interactions at a pulling speed of 2 µm/s are shown. hFc served as control for nonspecific binding. *P<0.05, 1-way analysis of variance, Dunnett post hoc test. Each independent coating procedure with minimum 625 force curves is taken as biological replicate. D, Histogram of binding forces distribution with peak fit at a pulling speed of 2 µm/s corresponding to data in C. E, Determination of the bond half lifetime by the Bell equation11 of mean loading rates and binding forces analyzed from data of pulling speeds at 0.5, 1, 2, 5, and 7.5 µm/s. Average of values from 4 independent coating procedures with minimum 625 force curves each. R=R2, koff =off-rate constant, τ0=bond half lifetime under zero force. F, Dissociation assays to determine cell–cell adhesion were performed in CaCo2 cells (WT or DSG2 KO background) expressing DSG2-WT-mGFP or DSG2-W2A-mGFP constructs. mGFP empty vector served as control. *P<0.05, 1-way analysis of variance, Sidak post hoc test. Corresponding Western blot analysis confirmed effective expression of DSG2 constructs (*) versus the endogenous protein (arrow) in CaCo2 cells. α-tubulin (TUBA) served as loading control. G, Representative images of monolayer fragmentation from experiments in F. H, Dissociation assays in immortalized keratinocytes isolated from neonatal murine skin of the respective genotype. *P<0.05 or as indicated, Welch analysis of variance, Dunnett post hoc test. I, Macroscopic cardiac phenotype of DSG2-W2A mut/mut mice at age 15 weeks. J, Cardiac hypertrophy was analyzed as mean cross-sectional area of cardiomyocytes in hematoxylin & eosin–stained sections. Scale bar, 30 µm. *P<0.05, unpaired Student t test. K, Representative images of intercalated discs acquired by transmission electron microscopy, 3 mice per genotype. Orange asterisks mark intercellular widening, orange circle marks a ruptured junction. Scale bar, 1 µm. To determine the effect of the W2A mutation on a cellular level, DSG2-WT and DSG2-W2A constructs were stably expressed in the epithelial cell line CaCo2 either in a WT or DSG2-deficient background (CaCo2[WT] or CaCo2[DSG2 KO]).12 Cell–cell dissociation assays were performed, in which a confluent cell monolayer is detached and exposed to defined mechanical stress. Expression of DSG2-W2A in CaCo2(WT) cells significantly reduced intercellular adhesion as indicated by an increased number of fragments (Figure 1F and 1G). Whereas expressing DSG2-WT in the CaCo2(DSG2 KO) line significantly rescued the impaired intercellular adhesion in response to DSG2 loss, the expression of DSG2-W2A had no effect on fragment numbers. Because these data demonstrate the DSG2 tryptophan swap to be the central adhesive mechanism, we next generated a CRISPR/Cas9-based knock-in mouse model for DSG2-W2A to investigate the consequences of impaired DSG2 adhesion in vivo (Figure S1). In line with the studies in human cells, dissociation assays in keratinocyte cell lines generated from DSG2-W2A pups revealed reduced cell–cell adhesion in heterozygous (mut/wt) and homozygous (mut/mut) mutants (Figure 1H). Moreover, DSG2-W2A mice presented with a pronounced cardiac phenotype. Adult mut/mut mice demonstrated ventricular deformation, fibrotic and calcified areas, and hypertrophic cardiomyocytes (Figure 1I and 1J). In line with reduced intercellular adhesion of cardiomyocytes, transmission electron microscopy revealed disturbed ICDs with widened intercellular space and occasionally completely ruptured junctions in mut/mut hearts (Figure 1K). A cardiac phenotype was also detectable during embryogenesis. Here, mating of heterozygous mice revealed a reduction of mut/mut offspring to 2.8% compared with the expected Mendelian ratio of 25%. Embryo dissections showed loss of the mut/mut animals between E12 and E14 (Figure S2A). The mut/mut embryos at day E12.5 appeared macroscopically pale with accumulation of blood in the cardiac area although the heart was still beating (Figure S2B). Cells suggestive for blood precursors were detectable in the pericardial space (Figure S2C). This finding suggests a rupture of the cardiac wall leading to pericardial bleeding and loss of mut/mut animals. Together, these in vitro and in vivo data outline an essential role of the DSG2 tryptophan swap for cell–cell adhesion and demonstrate severe pathologies associated with impaired DSG2 interaction. To study the functional consequences of impaired DSG2 binding, we generated the W2A point mutation (DSG2-W2A) to abrogate the proposed interaction mechanism of the only desmoglein expressed in cardiomyocytes (Figure 1A). First, we investigated in a cell-free setup whether DSG2-W2A is indeed affecting the binding properties of DSG2 interaction by applying single molecule force spectroscopy. Here, molecules are coupled to the tip of an atomic force microscopy probe and the surface of a mica sheet by a polyethylene glycol linker. By measuring the deflection of the probe during repeated approach to and retraction from the surface, binding events can be assessed quantitatively. From these data, properties such as binding frequency, binding force, and bond half-life time can be determined. Constructs were generated for expression of wild-type (WT) and mutant (W2A) DSG2 extracellular domains fused to a human Fc fragment and tested for homotypic (WT:WT, W2A:W2A) and heterotypic (WT:W2A) interaction properties (Figure 1B). These experiments showed a significant reduction of the frequency of W2A:W2A as well as WT:W2A interactions compared with WT:WT (Figure 1C). The frequency of the remaining W2A bindings was comparable to probing Fc alone, which indicates that these are mostly nonspecific interactions. Homotypic WT interactions display a clear binding force peak, whereas the remaining W2A interaction forces were spread widely, again pointing to the nonspecificity of these interactions (Figure 1D). The lifetime of DSG2 interactions under zero force (τ0) was determined by fitting the binding forces detected at different loading rates,11 yielding τ0=1.088s with R=0.993. In contrast, no sufficient fit (R=0.509) was possible for W2A interactions (Figure 1E). Together, these data outline that the tryptophan swap is the major interaction mechanism of DSG2. DSG2 (desmoglein-2) adhesion is mediated by tryptophan swap at position 2. A, Predicted interaction model of DSG2 extracellular domains by exchange of tryptophan residue at position 2 into a hydrophobic pocket of the opposing molecule. Cartoon 3D presentation of Protein Data Bank entry 5ERD9; tryptophan-2 is highlighted by ball and stick presentation. B, Schematic of single molecule force spectroscopy experiments. Recombinant extracellular domains (ECs) of wild-type DSG2 (DSG2-WT) or mutant DSG2 (DSG2-W2A) protein were coupled to a mica surface and atomic force microscopy cantilever by mean of a human Fc-tag (hFc) and a polyethylene glycol (PEG)20 linker and probed as indicated. C, Binding frequency of DSG2-W2A/DSG2-WT heterotypic and homotypic interactions at a pulling speed of 2 µm/s are shown. hFc served as control for nonspecific binding. *P<0.05, 1-way analysis of variance, Dunnett post hoc test. Each independent coating procedure with minimum 625 force curves is taken as biological replicate. D, Histogram of binding forces distribution with peak fit at a pulling speed of 2 µm/s corresponding to data in C. E, Determination of the bond half lifetime by the Bell equation11 of mean loading rates and binding forces analyzed from data of pulling speeds at 0.5, 1, 2, 5, and 7.5 µm/s. Average of values from 4 independent coating procedures with minimum 625 force curves each. R=R2, koff =off-rate constant, τ0=bond half lifetime under zero force. F, Dissociation assays to determine cell–cell adhesion were performed in CaCo2 cells (WT or DSG2 KO background) expressing DSG2-WT-mGFP or DSG2-W2A-mGFP constructs. mGFP empty vector served as control. *P<0.05, 1-way analysis of variance, Sidak post hoc test. Corresponding Western blot analysis confirmed effective expression of DSG2 constructs (*) versus the endogenous protein (arrow) in CaCo2 cells. α-tubulin (TUBA) served as loading control. G, Representative images of monolayer fragmentation from experiments in F. H, Dissociation assays in immortalized keratinocytes isolated from neonatal murine skin of the respective genotype. *P<0.05 or as indicated, Welch analysis of variance, Dunnett post hoc test. I, Macroscopic cardiac phenotype of DSG2-W2A mut/mut mice at age 15 weeks. J, Cardiac hypertrophy was analyzed as mean cross-sectional area of cardiomyocytes in hematoxylin & eosin–stained sections. Scale bar, 30 µm. *P<0.05, unpaired Student t test. K, Representative images of intercalated discs acquired by transmission electron microscopy, 3 mice per genotype. Orange asterisks mark intercellular widening, orange circle marks a ruptured junction. Scale bar, 1 µm. To determine the effect of the W2A mutation on a cellular level, DSG2-WT and DSG2-W2A constructs were stably expressed in the epithelial cell line CaCo2 either in a WT or DSG2-deficient background (CaCo2[WT] or CaCo2[DSG2 KO]).12 Cell–cell dissociation assays were performed, in which a confluent cell monolayer is detached and exposed to defined mechanical stress. Expression of DSG2-W2A in CaCo2(WT) cells significantly reduced intercellular adhesion as indicated by an increased number of fragments (Figure 1F and 1G). Whereas expressing DSG2-WT in the CaCo2(DSG2 KO) line significantly rescued the impaired intercellular adhesion in response to DSG2 loss, the expression of DSG2-W2A had no effect on fragment numbers. Because these data demonstrate the DSG2 tryptophan swap to be the central adhesive mechanism, we next generated a CRISPR/Cas9-based knock-in mouse model for DSG2-W2A to investigate the consequences of impaired DSG2 adhesion in vivo (Figure S1). In line with the studies in human cells, dissociation assays in keratinocyte cell lines generated from DSG2-W2A pups revealed reduced cell–cell adhesion in heterozygous (mut/wt) and homozygous (mut/mut) mutants (Figure 1H). Moreover, DSG2-W2A mice presented with a pronounced cardiac phenotype. Adult mut/mut mice demonstrated ventricular deformation, fibrotic and calcified areas, and hypertrophic cardiomyocytes (Figure 1I and 1J). In line with reduced intercellular adhesion of cardiomyocytes, transmission electron microscopy revealed disturbed ICDs with widened intercellular space and occasionally completely ruptured junctions in mut/mut hearts (Figure 1K). A cardiac phenotype was also detectable during embryogenesis. Here, mating of heterozygous mice revealed a reduction of mut/mut offspring to 2.8% compared with the expected Mendelian ratio of 25%. Embryo dissections showed loss of the mut/mut animals between E12 and E14 (Figure S2A). The mut/mut embryos at day E12.5 appeared macroscopically pale with accumulation of blood in the cardiac area although the heart was still beating (Figure S2B). Cells suggestive for blood precursors were detectable in the pericardial space (Figure S2C). This finding suggests a rupture of the cardiac wall leading to pericardial bleeding and loss of mut/mut animals. Together, these in vitro and in vivo data outline an essential role of the DSG2 tryptophan swap for cell–cell adhesion and demonstrate severe pathologies associated with impaired DSG2 interaction. [SUBTITLE] DSG2-W2A Mutant Mice Resemble the Phenotype of ACM [SUBSECTION] Given the cardiac affection in mutant mice, the notion that a majority of patients with ACM are reported with mutations in desmosomal genes,2 and that mutations specifically of W2 have already been associated with ACM,10 we examined homozygous and heterozygous mice including both sexes with regard to histologic and clinical ACM measures in an age range of 12 to 80 weeks. Histologic analysis showed cardiac fibrosis, a major hallmark of ACM, in the myocardium of the right ventricle (RV) and left ventricle (LV) in mut/mut animals (Figure 2A through 2D), which did not profoundly increase over time. In contrast, heterozygous animals demonstrated a milder phenotype, with a fraction of animals (36%) starting to develop fibrosis in the RV around 6 months, whereas LV was unaffected. To better address differences over time, we pooled animals into groups with a mean age of 4 and 6 months for homozygous and 6 and 12 months for heterozygous mice and performed functional analyses by echocardiography and ECG parallel to histologic evaluation. Moreover, we separated 12-month-old heterozygous animals according to RV fibrosis into a group without and with fibrosis (>10% fibrotic area). Mut/mut animals showed impaired RV systolic function at both time points with reduced tricuspid annular plane systolic excursion as well as fractional shortening (Figure 2E through 2G and Table S1). LV systolic impairment with reduction of the ejection fraction and corresponding parameters worsened over time and became significant after 6 months, although no increase in fibrosis was detectable (Figure 2D and 2H and Table S1). In mut/wt mice, RV fractional shortening was significantly reduced in 12-month-old animals with fibrosis. In ECG recordings of mut/mut mice for 30 minutes under anaesthesia, we noted deformation of the QRS complex with elongated QRS interval and reduced amplitude of the S peak with effects more pronounced in the older group; heterozygous animals with RV fibrosis showed similar effects or trends (Figure 2I through 2K and Table S1). Because the T wave was not clearly detectable in mutants, the J peak amplitude as indicator for early repolarization was determined. This was reduced in the mut/mut groups with same trend for fibrotic mut/wt hearts (Figure 2L). Parallel to these depolarization and repolarization abnormalities, we also noted the occurrence of ventricular arrhythmia, ranging from single premature ventricular contractions (PVCs) to multiple, multifocal PVCs and nonsustained ventricular tachycardia in 1 mouse (Figure 2M through 2O). The fraction of homozygous and heterozygous animals with PVCs and the frequency of these events correlated with age and fibrosis. Moreover, a substantial fraction (16%) of mut/mut animals was found dead during the observation period, which strongly suggests the occurrence of malignant arrhythmia with sudden death (Figure 2P). Investigating sex-related differences, no effect was observed on the extent of fibrosis or echocardiographic parameters. However, in mut/wt mice, PVCs were found in males only and male mut/mut mice were more prone to premature sudden death compared with females (Figure S3). DSG2 (desmoglein-2)–W2A mutant mice develop characteristics of arrhythmogenic cardiomyopathy. A, Cardiac fibrosis detected by picrosirius red collagen staining with representative images of sections from 6-month-old DSG2-W2A mut/mut and 12-month-old wt/wt and mut/wt animals. Scale bar = 1 mm. B through D, Corresponding analysis of the area of collagen in the right ventricle (RV) and left ventricle (LV). Continuous lines indicate the simple linear regression between age and area of collagen. Hearts with >10% of collagen in the RV (gray dotted line) were defined as “with fibrosis.” Each dot represents 1 animal. *P<0.05, mixed effects analysis with LV and RV matched per animal, Sidak post hoc test. Lines indicate median and quartile values. E, Representative echocardiography images for measurements of the tricuspid annular plane systolic excursion (TAPSE) and LV function in 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Left side: 2D images from apical 4-chamber view for TAPSE and parasternal short axis view for LV. M-mode tracings on the right were performed along the line indicated on the left. Scale bars, white, 2 mm; black, 100 ms. Corresponding analysis of (F) TAPSE, *P<0.05, Kruskal-Wallis test with Dunn post hoc test; (G) fractional shortening of the RV, *P<0.05, Kruskal-Wallis test with Dunn post hoc test; and (H) ejection fraction of the LV, *P<0.05, 1-way analysis of variance, Sidak post hoc test. I, ECG recoded in lead II with representative curves from 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Definition of respective peaks is indicated in the wt/wt curve. Scale bars, vertical, 0.5 mV; horizontal, 50 ms. Corresponding analysis of (J) QRS interval, *P<0.05 or as indicated, Kruskal-Wallis test with Dunn post hoc test; (K) amplitude of the S peak, *P<0.05 or as indicated, 1-way analysis of variance, Sidak post hoc test; and (L) amplitude of the J peak (early repolarization), *P<0.05 or as indicated, Kruskal-Wallis test with Dunn post hoc test. M, Ventricular arrythmia detected by ECG during 30 minutes of baseline measurements with example curves from 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Asterisks mark premature ventricular contractions (PVCs); *nsVT indicates a nonsustained ventricular tachycardia detected in 1 mut/mut animal. Scale bars, vertical, 0.5 mV; horizontal, 1 s. Corresponding analysis of (N) percentage of mice presenting with PVCs. Values in bars indicate corresponding absolute number of mice with PVC (colored bars) compared with total number of mice evaluated (empty bar) and (O) PVC burden depicted as number of PVCs per minute, *P<0.05, Kruskal-Wallis test with Dunn post hoc test. The black arrow indicates the animal presenting with nonsustained ventricular tachycardia. P, Kaplan-Meier survival plot of DSG2-W2A mice from an analysis period of 3 years. Vertical lines indicate dropouts because of unrelated elimination (end of experiment, breeding, injuries). Values indicate corresponding absolute number of mice with sudden death compared with total number of mice evaluated. *P<0.05, Gehan-Breslow-Wilcoxon test. Box with color indications of respective groups in the middle applies to the entire figure. In summary, mutant animals show histopathologic features with echocardiography and ECG abnormalities similar to patients with ACM. Within the limits of a murine model, both genotypes fulfil the Padua criteria13 to establish the diagnosis of ACM (Table S2). Here, DSG2-W2A mut/mut animals resemble the phenotype of biventricular ACM, whereas in mut/wt mice, the phenotype is milder with variable penetrance and age-dependent occurrence only in the RV. According to the Padua criteria,13 the heterozygous genotype recalls the characteristics of a right-dominant ventricular ACM, as long as fibrosis was present. Together, these data demonstrate that loss of desmosomal adhesion is sufficient to induce an ACM phenotype and that the DSG2-W2A model may resemble 2 different variants of the disease. Given the cardiac affection in mutant mice, the notion that a majority of patients with ACM are reported with mutations in desmosomal genes,2 and that mutations specifically of W2 have already been associated with ACM,10 we examined homozygous and heterozygous mice including both sexes with regard to histologic and clinical ACM measures in an age range of 12 to 80 weeks. Histologic analysis showed cardiac fibrosis, a major hallmark of ACM, in the myocardium of the right ventricle (RV) and left ventricle (LV) in mut/mut animals (Figure 2A through 2D), which did not profoundly increase over time. In contrast, heterozygous animals demonstrated a milder phenotype, with a fraction of animals (36%) starting to develop fibrosis in the RV around 6 months, whereas LV was unaffected. To better address differences over time, we pooled animals into groups with a mean age of 4 and 6 months for homozygous and 6 and 12 months for heterozygous mice and performed functional analyses by echocardiography and ECG parallel to histologic evaluation. Moreover, we separated 12-month-old heterozygous animals according to RV fibrosis into a group without and with fibrosis (>10% fibrotic area). Mut/mut animals showed impaired RV systolic function at both time points with reduced tricuspid annular plane systolic excursion as well as fractional shortening (Figure 2E through 2G and Table S1). LV systolic impairment with reduction of the ejection fraction and corresponding parameters worsened over time and became significant after 6 months, although no increase in fibrosis was detectable (Figure 2D and 2H and Table S1). In mut/wt mice, RV fractional shortening was significantly reduced in 12-month-old animals with fibrosis. In ECG recordings of mut/mut mice for 30 minutes under anaesthesia, we noted deformation of the QRS complex with elongated QRS interval and reduced amplitude of the S peak with effects more pronounced in the older group; heterozygous animals with RV fibrosis showed similar effects or trends (Figure 2I through 2K and Table S1). Because the T wave was not clearly detectable in mutants, the J peak amplitude as indicator for early repolarization was determined. This was reduced in the mut/mut groups with same trend for fibrotic mut/wt hearts (Figure 2L). Parallel to these depolarization and repolarization abnormalities, we also noted the occurrence of ventricular arrhythmia, ranging from single premature ventricular contractions (PVCs) to multiple, multifocal PVCs and nonsustained ventricular tachycardia in 1 mouse (Figure 2M through 2O). The fraction of homozygous and heterozygous animals with PVCs and the frequency of these events correlated with age and fibrosis. Moreover, a substantial fraction (16%) of mut/mut animals was found dead during the observation period, which strongly suggests the occurrence of malignant arrhythmia with sudden death (Figure 2P). Investigating sex-related differences, no effect was observed on the extent of fibrosis or echocardiographic parameters. However, in mut/wt mice, PVCs were found in males only and male mut/mut mice were more prone to premature sudden death compared with females (Figure S3). DSG2 (desmoglein-2)–W2A mutant mice develop characteristics of arrhythmogenic cardiomyopathy. A, Cardiac fibrosis detected by picrosirius red collagen staining with representative images of sections from 6-month-old DSG2-W2A mut/mut and 12-month-old wt/wt and mut/wt animals. Scale bar = 1 mm. B through D, Corresponding analysis of the area of collagen in the right ventricle (RV) and left ventricle (LV). Continuous lines indicate the simple linear regression between age and area of collagen. Hearts with >10% of collagen in the RV (gray dotted line) were defined as “with fibrosis.” Each dot represents 1 animal. *P<0.05, mixed effects analysis with LV and RV matched per animal, Sidak post hoc test. Lines indicate median and quartile values. E, Representative echocardiography images for measurements of the tricuspid annular plane systolic excursion (TAPSE) and LV function in 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Left side: 2D images from apical 4-chamber view for TAPSE and parasternal short axis view for LV. M-mode tracings on the right were performed along the line indicated on the left. Scale bars, white, 2 mm; black, 100 ms. Corresponding analysis of (F) TAPSE, *P<0.05, Kruskal-Wallis test with Dunn post hoc test; (G) fractional shortening of the RV, *P<0.05, Kruskal-Wallis test with Dunn post hoc test; and (H) ejection fraction of the LV, *P<0.05, 1-way analysis of variance, Sidak post hoc test. I, ECG recoded in lead II with representative curves from 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Definition of respective peaks is indicated in the wt/wt curve. Scale bars, vertical, 0.5 mV; horizontal, 50 ms. Corresponding analysis of (J) QRS interval, *P<0.05 or as indicated, Kruskal-Wallis test with Dunn post hoc test; (K) amplitude of the S peak, *P<0.05 or as indicated, 1-way analysis of variance, Sidak post hoc test; and (L) amplitude of the J peak (early repolarization), *P<0.05 or as indicated, Kruskal-Wallis test with Dunn post hoc test. M, Ventricular arrythmia detected by ECG during 30 minutes of baseline measurements with example curves from 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Asterisks mark premature ventricular contractions (PVCs); *nsVT indicates a nonsustained ventricular tachycardia detected in 1 mut/mut animal. Scale bars, vertical, 0.5 mV; horizontal, 1 s. Corresponding analysis of (N) percentage of mice presenting with PVCs. Values in bars indicate corresponding absolute number of mice with PVC (colored bars) compared with total number of mice evaluated (empty bar) and (O) PVC burden depicted as number of PVCs per minute, *P<0.05, Kruskal-Wallis test with Dunn post hoc test. The black arrow indicates the animal presenting with nonsustained ventricular tachycardia. P, Kaplan-Meier survival plot of DSG2-W2A mice from an analysis period of 3 years. Vertical lines indicate dropouts because of unrelated elimination (end of experiment, breeding, injuries). Values indicate corresponding absolute number of mice with sudden death compared with total number of mice evaluated. *P<0.05, Gehan-Breslow-Wilcoxon test. Box with color indications of respective groups in the middle applies to the entire figure. In summary, mutant animals show histopathologic features with echocardiography and ECG abnormalities similar to patients with ACM. Within the limits of a murine model, both genotypes fulfil the Padua criteria13 to establish the diagnosis of ACM (Table S2). Here, DSG2-W2A mut/mut animals resemble the phenotype of biventricular ACM, whereas in mut/wt mice, the phenotype is milder with variable penetrance and age-dependent occurrence only in the RV. According to the Padua criteria,13 the heterozygous genotype recalls the characteristics of a right-dominant ventricular ACM, as long as fibrosis was present. Together, these data demonstrate that loss of desmosomal adhesion is sufficient to induce an ACM phenotype and that the DSG2-W2A model may resemble 2 different variants of the disease. [SUBTITLE] Integrin-β6 Expression Is Altered in ACM Patient and DSG2-W2A Mouse Samples [SUBSECTION] Next, we applied this mouse model to investigate mechanisms leading to ACM. RNA sequencing was performed from ventricles of mut/mut and wt/wt hearts both before onset of fibrosis (age 5 days) and when the biventricular fibrosis and ACM phenotype was established (after 9 weeks). First, we compared sequencing data from 9-week-old mice with the top differentially expressed genes derived from published transcriptomic data sets of ACM patient hearts (GEO database: GSE107157/GSE10748014 and GSE2981915). Murine samples clustered according to their genotype and mut/mut hearts resembled the gene expression pattern of patients (Figure S4), further supporting the observation that mutant mice faithfully reproduce major aspects of the disease. To identify common genes deregulated during ACM pathogenesis, we compared the differentially expressed genes from patient data with the results from mutant mice. Here, we included data from 5-day-old mice to identify changes already present before onset of secondary effects attributable to fibrosis. Integrin-β6 (Itgb6) was the only transcript consistently deregulated in all data sets (Figure 3A). Its common downregulation in patients with ACM and mutant mice on RNA levels was further confirmed for heterozygous DSG2-W2A mutants compared with WT (Figure 3B). However, protein levels were unaltered in mutant heart samples (Figure 3C). Immunostaining revealed that, in mutant hearts, ITGB6, which mainly localized to a compartment suggestive for the transverse tubules and costameres in wt/wt animals, was enriched at the ICD (Figure 3D and E). Together, these data demonstrate altered ITGB6 mRNA expression as a common feature in ACM patient and DSG2-W2A hearts and an enrichment of the protein at the ICD in mutant murine samples. Integrin-β6 is deregulated in DSG2 (desmoglein-2)–W2A mutants. A, Venn diagram of significantly altered genes from indicated arrhythmogenic cardiomyopathy (ACM) patient data sets (ACM versus healthy control) and DSG2-W2A mice at the age of 5 days and 9 weeks (mut/mut versus wt/wt) highlighting integrin-β6 (Itgb6) as only overlapping gene with same direction of expression in all data sets. Numbers indicate the amount of overlapping genes for the respective overlays. B, RNA expression of Itgb6 analyzed by means of reverse transcription quantitative polymerase chain reaction in adult DSG2-W2A mouse hearts. *P<0.05, unpaired Student t test versus wt/wt. Gapdh served as reference gene. C, Representative Western blot and respective analysis of band intensity of integrin-β6 (ITGB6) in DSG2-W2A hearts. GAPDH served as loading control. 1-way analysis of variance, Dunnett post hoc test. D and E, Immunostaining of ITGB6 (red in overlay) in DSG2-W2A hearts with corresponding analysis of staining intensity in cardiomyocytes in total and ratio of staining intensity at the intercalated disc (ICD) area (orange arrowheads) versus cardiomyocytes’ cytosolic area. DSP (desmoplakin; cyan) marks ICDs, DAPI (blue) nuclei and F-actin (green) the sarcomere system. Lower row shows an overview image of a fibrotic area in mut/mut hearts. Scale bars, 25 µm. *P<0.05; left graph: Kruskal-Wallis test with Dunn post hoc test; right graph: 1-way analysis of variance, Dunnett post hoc test. Box with color indications of respective groups applies to the entire figure. Next, we applied this mouse model to investigate mechanisms leading to ACM. RNA sequencing was performed from ventricles of mut/mut and wt/wt hearts both before onset of fibrosis (age 5 days) and when the biventricular fibrosis and ACM phenotype was established (after 9 weeks). First, we compared sequencing data from 9-week-old mice with the top differentially expressed genes derived from published transcriptomic data sets of ACM patient hearts (GEO database: GSE107157/GSE10748014 and GSE2981915). Murine samples clustered according to their genotype and mut/mut hearts resembled the gene expression pattern of patients (Figure S4), further supporting the observation that mutant mice faithfully reproduce major aspects of the disease. To identify common genes deregulated during ACM pathogenesis, we compared the differentially expressed genes from patient data with the results from mutant mice. Here, we included data from 5-day-old mice to identify changes already present before onset of secondary effects attributable to fibrosis. Integrin-β6 (Itgb6) was the only transcript consistently deregulated in all data sets (Figure 3A). Its common downregulation in patients with ACM and mutant mice on RNA levels was further confirmed for heterozygous DSG2-W2A mutants compared with WT (Figure 3B). However, protein levels were unaltered in mutant heart samples (Figure 3C). Immunostaining revealed that, in mutant hearts, ITGB6, which mainly localized to a compartment suggestive for the transverse tubules and costameres in wt/wt animals, was enriched at the ICD (Figure 3D and E). Together, these data demonstrate altered ITGB6 mRNA expression as a common feature in ACM patient and DSG2-W2A hearts and an enrichment of the protein at the ICD in mutant murine samples. Integrin-β6 is deregulated in DSG2 (desmoglein-2)–W2A mutants. A, Venn diagram of significantly altered genes from indicated arrhythmogenic cardiomyopathy (ACM) patient data sets (ACM versus healthy control) and DSG2-W2A mice at the age of 5 days and 9 weeks (mut/mut versus wt/wt) highlighting integrin-β6 (Itgb6) as only overlapping gene with same direction of expression in all data sets. Numbers indicate the amount of overlapping genes for the respective overlays. B, RNA expression of Itgb6 analyzed by means of reverse transcription quantitative polymerase chain reaction in adult DSG2-W2A mouse hearts. *P<0.05, unpaired Student t test versus wt/wt. Gapdh served as reference gene. C, Representative Western blot and respective analysis of band intensity of integrin-β6 (ITGB6) in DSG2-W2A hearts. GAPDH served as loading control. 1-way analysis of variance, Dunnett post hoc test. D and E, Immunostaining of ITGB6 (red in overlay) in DSG2-W2A hearts with corresponding analysis of staining intensity in cardiomyocytes in total and ratio of staining intensity at the intercalated disc (ICD) area (orange arrowheads) versus cardiomyocytes’ cytosolic area. DSP (desmoplakin; cyan) marks ICDs, DAPI (blue) nuclei and F-actin (green) the sarcomere system. Lower row shows an overview image of a fibrotic area in mut/mut hearts. Scale bars, 25 µm. *P<0.05; left graph: Kruskal-Wallis test with Dunn post hoc test; right graph: 1-way analysis of variance, Dunnett post hoc test. Box with color indications of respective groups applies to the entire figure. [SUBTITLE] DSG2-W2A Mutant Hearts Present With Structurally Impaired ICDs [SUBSECTION] This result together with the transmission electron microscopy data (Figure 1K) suggest structural changes of the ICD in mutant hearts, which we addressed further. Using DSP as marker, we reconstructed the outlines of ICDs in wt/wt and mut/mut hearts from 3D stacks captured with structured illumination microscopy. This analysis yielded an increase in ICD volume, in particular attributable to an enhanced width between 2 adjacent cells (Figure 4A). The majority of desmosomal molecules as well as N-cadherin were regularly distributed at mutant ICDs (Figure S5A and S5B). This was corroborated by RNA sequencing data from 5-day-old and 9-week-old animals, which showed no consistent changes of desmosomal, adherens, gap, or tight junction molecules (Figure S5C through S5E). The only adhesion molecule reduced globally was DSG2 (Figure S5A and S5B). Although DSG2 was still present at the ICD, detailed analysis by structured illumination microscopy revealed reduced number and volume of DSG2 signals (Figure 4B). Moreover, fluorescence recovery after photobleaching experiments in neonatal murine cardiomyocytes transduced with DSG2-WT-mGFP or DSG2-W2A-mGFP showed a higher mobility of the mutant protein at cardiomyocyte junctions (Figure 4C), indicating reduced membrane stability of DSG2-W2A. To further investigate the possibility that the stability and integrity of ICD molecules is compromised, we performed Triton-X-100 separation assays for proteins with unaltered ICD localization in mutant hearts. Cytosolic and unanchored membrane proteins are found in the Triton-X-100 soluble fraction, whereas cytoskeleton-bound molecules are mainly detectable in the nonsoluble fraction. Both desmosomal molecules (PG, PKP2) as well as N-cadherin showed increased levels in the soluble pool, indicating reduced cytoskeletal anchorage and impaired ICD integrity in mutants (Figure 4D). Moreover, the gap junction molecule connexin-43, which is required for electrical coupling of cardiomyocytes, was delocalized to the lateral membrane (Figure 4E), which was described as a feature of disrupted ICDs.8 Disrupted ICD structure in DSG2 (desmoglein-2)–W2A mutant mice. A, Representative z-stack reconstruction of segmented intercalated discs (ICDs) in top and side view acquired with structured illumination microscopy. Overlay of analyzed ICD volume is shown in gray. ICDs are marked by DSP (desmoplakin; magenta). Scheme on top presents segmented area and pictograms on the right display the respective angle of view. Corresponding analysis of ICD volume and width of ICD between adjacent cardiomyocytes below. Scale bar, 5 µm. *P<0.05, Mann-Whitney test. Each dot represents the value of 1 ICD from 4 mice per genotype. B, Representative images of DSG2 (magenta) and filamentary actin (f-actin; white) z-stacks acquired by structured illumination microscopy and presented as maximum intensity projection. Lower row shows color-coded height projection of DSG2 signals in z-stack after signal thresholding as performed for analysis. Related analysis of DSG2 signal volume and number per ICD length is shown below. Pictograms on the right display the respective angle of view. Scale bar, 5 µm. *P<0.05, unpaired Student t test, with Welch correction. Each dot represents the value of 1 ICD from in total 3 mice per genotype. C, Fluorescence recovery after photobleaching analysis of DSG2–wild type (WT) and DSG2-W2A-mGFP fusion proteins at the cell–cell junction of neonatal cardiomyocytes with representative intensity kymographs of bleached areas on the left. Time point 0 = bleach as indicated by the black arrow. Analysis of the mobile fraction of the indicated mGFP-fusion proteins is shown on the right. *P<0.05, unpaired Student t test, with Welch correction. Each dot represents the mean value of 1 heart from in total 3 isolations. D, Representative triton-X-100 assay immunoblot with separation of a soluble (sol), noncytoskeletal bound protein fraction from a nonsoluble (non-sol), cytoskeletal-anchored fraction and corresponding analysis shown below. PG (plakoglobin), PKP2 (plakophilin-2), and N-cadherin (NCAD) were analyzed. Intensity of proteins was normalized to the total amount of protein detected by ponceau staining. GAPDH and DSP served as separation control. *P<0.05 or as indicated, unpaired Student t test (PG, NCAD, PKP2) or Mann-Whitney test (DSP). Each dot represents the result from 1 mouse. E, Immunostaining of connexin-43 (CX43; red in overlay) in DSG2-W2A hearts. NCAD (yellow) marks ICDs, DAPI (blue) nuclei, and F-actin (green) the sarcomere system. Orange arrowheads mark ICD, pink arrowheads highlight lateralization of CX43 staining. Scale bars, 25 µm. Images representative for 5 mice per genotype. Box with color indications of respective groups applies to the entire figure. These data together with the transmission electron microscopy results demonstrate structurally severely altered ICDs in response to the DSG2-W2A mutation and suggest a molecular basis leading to the functional alterations detectable by echocardiography and ECG. This result together with the transmission electron microscopy data (Figure 1K) suggest structural changes of the ICD in mutant hearts, which we addressed further. Using DSP as marker, we reconstructed the outlines of ICDs in wt/wt and mut/mut hearts from 3D stacks captured with structured illumination microscopy. This analysis yielded an increase in ICD volume, in particular attributable to an enhanced width between 2 adjacent cells (Figure 4A). The majority of desmosomal molecules as well as N-cadherin were regularly distributed at mutant ICDs (Figure S5A and S5B). This was corroborated by RNA sequencing data from 5-day-old and 9-week-old animals, which showed no consistent changes of desmosomal, adherens, gap, or tight junction molecules (Figure S5C through S5E). The only adhesion molecule reduced globally was DSG2 (Figure S5A and S5B). Although DSG2 was still present at the ICD, detailed analysis by structured illumination microscopy revealed reduced number and volume of DSG2 signals (Figure 4B). Moreover, fluorescence recovery after photobleaching experiments in neonatal murine cardiomyocytes transduced with DSG2-WT-mGFP or DSG2-W2A-mGFP showed a higher mobility of the mutant protein at cardiomyocyte junctions (Figure 4C), indicating reduced membrane stability of DSG2-W2A. To further investigate the possibility that the stability and integrity of ICD molecules is compromised, we performed Triton-X-100 separation assays for proteins with unaltered ICD localization in mutant hearts. Cytosolic and unanchored membrane proteins are found in the Triton-X-100 soluble fraction, whereas cytoskeleton-bound molecules are mainly detectable in the nonsoluble fraction. Both desmosomal molecules (PG, PKP2) as well as N-cadherin showed increased levels in the soluble pool, indicating reduced cytoskeletal anchorage and impaired ICD integrity in mutants (Figure 4D). Moreover, the gap junction molecule connexin-43, which is required for electrical coupling of cardiomyocytes, was delocalized to the lateral membrane (Figure 4E), which was described as a feature of disrupted ICDs.8 Disrupted ICD structure in DSG2 (desmoglein-2)–W2A mutant mice. A, Representative z-stack reconstruction of segmented intercalated discs (ICDs) in top and side view acquired with structured illumination microscopy. Overlay of analyzed ICD volume is shown in gray. ICDs are marked by DSP (desmoplakin; magenta). Scheme on top presents segmented area and pictograms on the right display the respective angle of view. Corresponding analysis of ICD volume and width of ICD between adjacent cardiomyocytes below. Scale bar, 5 µm. *P<0.05, Mann-Whitney test. Each dot represents the value of 1 ICD from 4 mice per genotype. B, Representative images of DSG2 (magenta) and filamentary actin (f-actin; white) z-stacks acquired by structured illumination microscopy and presented as maximum intensity projection. Lower row shows color-coded height projection of DSG2 signals in z-stack after signal thresholding as performed for analysis. Related analysis of DSG2 signal volume and number per ICD length is shown below. Pictograms on the right display the respective angle of view. Scale bar, 5 µm. *P<0.05, unpaired Student t test, with Welch correction. Each dot represents the value of 1 ICD from in total 3 mice per genotype. C, Fluorescence recovery after photobleaching analysis of DSG2–wild type (WT) and DSG2-W2A-mGFP fusion proteins at the cell–cell junction of neonatal cardiomyocytes with representative intensity kymographs of bleached areas on the left. Time point 0 = bleach as indicated by the black arrow. Analysis of the mobile fraction of the indicated mGFP-fusion proteins is shown on the right. *P<0.05, unpaired Student t test, with Welch correction. Each dot represents the mean value of 1 heart from in total 3 isolations. D, Representative triton-X-100 assay immunoblot with separation of a soluble (sol), noncytoskeletal bound protein fraction from a nonsoluble (non-sol), cytoskeletal-anchored fraction and corresponding analysis shown below. PG (plakoglobin), PKP2 (plakophilin-2), and N-cadherin (NCAD) were analyzed. Intensity of proteins was normalized to the total amount of protein detected by ponceau staining. GAPDH and DSP served as separation control. *P<0.05 or as indicated, unpaired Student t test (PG, NCAD, PKP2) or Mann-Whitney test (DSP). Each dot represents the result from 1 mouse. E, Immunostaining of connexin-43 (CX43; red in overlay) in DSG2-W2A hearts. NCAD (yellow) marks ICDs, DAPI (blue) nuclei, and F-actin (green) the sarcomere system. Orange arrowheads mark ICD, pink arrowheads highlight lateralization of CX43 staining. Scale bars, 25 µm. Images representative for 5 mice per genotype. Box with color indications of respective groups applies to the entire figure. These data together with the transmission electron microscopy results demonstrate structurally severely altered ICDs in response to the DSG2-W2A mutation and suggest a molecular basis leading to the functional alterations detectable by echocardiography and ECG. [SUBTITLE] Integrin-αV/β6 Is Activated in DSG2-W2A Mutant Hearts [SUBSECTION] In contrast to DSG2, we detected ITGB6 to be increased at the ICD (Figure 3D and 3E), which was confirmed by analysis of structured illumination microscopy images (Figure 5A). ITGB6 needs to heterodimerize with integrin-αV (ITGAV) in order to be activated and bind to the extracellular matrix.16–18 Thus, we analyzed the expression of the counterpart ITGAV, which was significantly upregulated on protein level and increased at the ICD and costameres of mutant hearts (Figure 5B and 5C). In contrast, the localization and intensity of integrin-β1, as classical representative of the integrin group, was not altered (Figure S6). Staining with an antibody specifically recognizing the heterodimer of ITGAV/B6 revealed increased amount of dimer formation at the ICD and costameres in mutant mice (Figure 5D). Moreover, increased ITGAV/B6 staining intensity was also detectable in an ACM patient sample with DSP mutation (Figure 5E). To stabilize active integrins, recruitment of the cytoskeletal adapters talin and vinculin is required.19 Accordingly, the expression of talin-2 was increased in DSG2-W2A mut/mut myocardium, with vinculin being enriched at the ICD (Figure 5F and 5G). This effect is similar to what was shown before in response to increased intracellular traction forces on junctions.20 Thus, these data suggest that in response to a dysfunctional, nonadhesive ICD with impaired cytoskeletal anchorage, ITGAV/B6 dimers are recruited and activated at the compromised ICD as well as at the costamere region. Increased activation of ITGB6/AV at ICDs of DSG2 (desmoglein-2)–W2A mutant mice. A, Representative intercalated disc (ICD) reconstruction from z-stacks of integrin-β6 (ITGB6; green) and DSP (magenta) immunostaining acquired by structured illumination microscopy and presented as maximum intensity projection. Related analysis of ITGB6 signal volume and number per ICD length is shown below. Scale bar, 5 µm. *P<0.05 or as indicated, unpaired Student t test with Welch correction (left graph) and Mann-Whitney test (right graph). Each dot represents the value of 1 ICD from in total 4 mice per genotype. B, Representative Western blot and analysis of band intensity of integrin-αV (ITGAV) in DSG2-W2A hearts. GAPDH served as loading control. *P<0.05, unpaired Student t test with Welch correction. C, Immunostaining of ITGAV in DSG2-W2A hearts on the right with respective analysis on the left. DAPI (blue) marks nuclei. Lower row shows an overview image of a fibrotic area in mut/mut hearts. Dotted orange line marks the edge of fibrotic area. Scale bars: upper rows, 20 µm; lower row, 50 µm. *P<0.05, unpaired Student t test. D, Immunostaining of ITGAV/B6 heterodimer in DSG2-W2A mutant hearts with respective analysis of staining intensity on the right. Cyan rectangle marks zoomed area on the right. Scale bars: overview, 50 µm; insert, 10 µm. *P<0.05, unpaired Student t test. E, Representative immunostaining images of ITGAV/B6 heterodimer staining (red) in a patient with arrhythmogenic cardiomyopathy (ACM; DSP-E952X, heterozygous) and healthy control sample. F-actin (green) stains the sarcomere system. For the patient with ACM, 4 different tissue samples were analyzed and compared with 2 tissue samples from 2 healthy controls. Scale bar, 20 µm. F, Immunostaining of VCL (vinculin; magenta) and TLN2 (talin-2; red) in DSG2-W2A hearts on the right with respective analysis of the mean staining intensity in cardiomyocytes and ratio of the staining intensity at the ICD versus cytosolic area on the left. DAPI (blue) marks nuclei. F-actin (green) stains the sarcomere system. Scale bar, 25 µm; *P<0.05, unpaired Student t test. G, Representative Western blot and analysis of band intensity of VCL and TLN2 in DSG2-W2A hearts. GAPDH served as loading control. *P<0.05, unpaired Student t test for TLN2, Mann-Whitney test for VCL. Box with color indications of respective groups applies to the entire figure. In contrast to DSG2, we detected ITGB6 to be increased at the ICD (Figure 3D and 3E), which was confirmed by analysis of structured illumination microscopy images (Figure 5A). ITGB6 needs to heterodimerize with integrin-αV (ITGAV) in order to be activated and bind to the extracellular matrix.16–18 Thus, we analyzed the expression of the counterpart ITGAV, which was significantly upregulated on protein level and increased at the ICD and costameres of mutant hearts (Figure 5B and 5C). In contrast, the localization and intensity of integrin-β1, as classical representative of the integrin group, was not altered (Figure S6). Staining with an antibody specifically recognizing the heterodimer of ITGAV/B6 revealed increased amount of dimer formation at the ICD and costameres in mutant mice (Figure 5D). Moreover, increased ITGAV/B6 staining intensity was also detectable in an ACM patient sample with DSP mutation (Figure 5E). To stabilize active integrins, recruitment of the cytoskeletal adapters talin and vinculin is required.19 Accordingly, the expression of talin-2 was increased in DSG2-W2A mut/mut myocardium, with vinculin being enriched at the ICD (Figure 5F and 5G). This effect is similar to what was shown before in response to increased intracellular traction forces on junctions.20 Thus, these data suggest that in response to a dysfunctional, nonadhesive ICD with impaired cytoskeletal anchorage, ITGAV/B6 dimers are recruited and activated at the compromised ICD as well as at the costamere region. Increased activation of ITGB6/AV at ICDs of DSG2 (desmoglein-2)–W2A mutant mice. A, Representative intercalated disc (ICD) reconstruction from z-stacks of integrin-β6 (ITGB6; green) and DSP (magenta) immunostaining acquired by structured illumination microscopy and presented as maximum intensity projection. Related analysis of ITGB6 signal volume and number per ICD length is shown below. Scale bar, 5 µm. *P<0.05 or as indicated, unpaired Student t test with Welch correction (left graph) and Mann-Whitney test (right graph). Each dot represents the value of 1 ICD from in total 4 mice per genotype. B, Representative Western blot and analysis of band intensity of integrin-αV (ITGAV) in DSG2-W2A hearts. GAPDH served as loading control. *P<0.05, unpaired Student t test with Welch correction. C, Immunostaining of ITGAV in DSG2-W2A hearts on the right with respective analysis on the left. DAPI (blue) marks nuclei. Lower row shows an overview image of a fibrotic area in mut/mut hearts. Dotted orange line marks the edge of fibrotic area. Scale bars: upper rows, 20 µm; lower row, 50 µm. *P<0.05, unpaired Student t test. D, Immunostaining of ITGAV/B6 heterodimer in DSG2-W2A mutant hearts with respective analysis of staining intensity on the right. Cyan rectangle marks zoomed area on the right. Scale bars: overview, 50 µm; insert, 10 µm. *P<0.05, unpaired Student t test. E, Representative immunostaining images of ITGAV/B6 heterodimer staining (red) in a patient with arrhythmogenic cardiomyopathy (ACM; DSP-E952X, heterozygous) and healthy control sample. F-actin (green) stains the sarcomere system. For the patient with ACM, 4 different tissue samples were analyzed and compared with 2 tissue samples from 2 healthy controls. Scale bar, 20 µm. F, Immunostaining of VCL (vinculin; magenta) and TLN2 (talin-2; red) in DSG2-W2A hearts on the right with respective analysis of the mean staining intensity in cardiomyocytes and ratio of the staining intensity at the ICD versus cytosolic area on the left. DAPI (blue) marks nuclei. F-actin (green) stains the sarcomere system. Scale bar, 25 µm; *P<0.05, unpaired Student t test. G, Representative Western blot and analysis of band intensity of VCL and TLN2 in DSG2-W2A hearts. GAPDH served as loading control. *P<0.05, unpaired Student t test for TLN2, Mann-Whitney test for VCL. Box with color indications of respective groups applies to the entire figure. [SUBTITLE] TGF-β Signaling Is Upregulated in DSG2-W2A Mutant Hearts [SUBSECTION] ITGAV/B6 dimers have the ability to activate the profibrotic cytokine transforming growth factor–β (TGF-β) by binding and removal of the latency-associated peptide.17 Gene set enrichment analyses revealed an upregulation of genes associated with TGF-β signaling in both 9-week-old mutants as well as patients with ACM (Figure 6A). Moreover, direct targets of receptor-regulated SMADs involved in TGF-β signaling21 were upregulated in DSG2-W2A mutant hearts (Figure 6B). Accordingly, increased amounts of nuclear SMAD2/3 phosphorylated at the activation sites S465/S467 or S423/S425, respectively, were detected in these hearts (Figure 6C). Increased levels of pSMAD2/3 were found not only in fibroblasts but also in cardiomyocytes, mainly adjacent to fibrotic areas. These data indicate upregulation of the profibrotic TGF-β pathway and its downstream targets, which include extracellular matrix proteins such as collagens, laminin, or fibronectin. Elevated transforming growth factor–β signaling in DSG2 (desmoglein-2)–W2A hearts as result of ITGAV/B6 activity. Barcode plots of gene set enrichment analysis of (A) the KEGG_TGF_BETA_SIGNALING_PATHWAY data set (systematic name: M2642)41 and (B) genes directly regulated by receptor-regulated SMADs (R-SMADs; includes SMAD1/2/3/5/9) as published in the TRRUST database21 in 9-week-old DSG2-W2A (mut/mut vs wt/wt) or arrhythmogenic cardiomyopathy (ACM) patient data set 1 (ACM versus healthy control; GEO: GSE107157/GSE107480). Indicated P values are calculated by function cameraPR of the R package limma. C, Immunostaining of phosphorylated SMAD2/3 (magenta, S465/S467 or S423/S425, respectively) in sections of DSG2-W2A hearts and related analysis of nuclear staining intensity. Nuclei are stained with DAPI (blue), cardiomyocytes are marked with f-actin (green). The dotted orange line marks the edge of the fibrotic area. Scale bar, 50 µm. *P<0.05, unpaired Student t test. D, Schematic of experimental setup for integrin-αVβ6 (ITGAV/B6) blocking experiments in cardiac slice culture with related results in E. Icons are derived from BioRender. E, Reverse transcription quantitative polymerase chain reaction analysis of expression of genes downstream of transforming growth factor–β (TGF-β) signaling in cardiac slice cultures treated with inhibiting anti-ITGAV/B6 (1:15) or 10 µmol/L GW788388, an inhibitor of TGF-β receptor I, for 24 hours. *P< 0.05, paired Student t test versus indicated control condition. F, Schematic of experimental setup for in vivo ITGAV/B6 blocking experiments by injection of 40 mg/kg EMD527040 (EMD) intraperitoneally daily. DMSO was applied as vehicle control. Icons are derived from BioRender. G, Reverse transcription quantitative polymerase chain reaction expression analysis of genes downstream of TGF-β signaling in hearts of mice treated with EMD or respective amount of DMSO for 10 days. *P<0.05, unpaired Student t test versus DMSO. H and I, Cardiac fibrosis detected by picrosirius red collagen staining with representative images and corresponding analysis of the area of collagen in the right ventricle (RV) and left ventricle (LV). Lines indicate littermates. Each dot represents 1 animal. *P<0.05 or as indicated, grouped 2-way repeated-measures analysis of variance with LV/RV and experimental pairs matched, Sidak post hoc test. Scale bar, 1 mm. J through L, ECG recoded in lead II with representative curves shown in J. Corresponding analysis of R, S, and J peak amplitude and QRS interval, *P<0.05 or as indicated. Paired Student t test. Lines indicate littermates. Each dot represents 1 animal. ITGAV/B6 dimers have the ability to activate the profibrotic cytokine transforming growth factor–β (TGF-β) by binding and removal of the latency-associated peptide.17 Gene set enrichment analyses revealed an upregulation of genes associated with TGF-β signaling in both 9-week-old mutants as well as patients with ACM (Figure 6A). Moreover, direct targets of receptor-regulated SMADs involved in TGF-β signaling21 were upregulated in DSG2-W2A mutant hearts (Figure 6B). Accordingly, increased amounts of nuclear SMAD2/3 phosphorylated at the activation sites S465/S467 or S423/S425, respectively, were detected in these hearts (Figure 6C). Increased levels of pSMAD2/3 were found not only in fibroblasts but also in cardiomyocytes, mainly adjacent to fibrotic areas. These data indicate upregulation of the profibrotic TGF-β pathway and its downstream targets, which include extracellular matrix proteins such as collagens, laminin, or fibronectin. Elevated transforming growth factor–β signaling in DSG2 (desmoglein-2)–W2A hearts as result of ITGAV/B6 activity. Barcode plots of gene set enrichment analysis of (A) the KEGG_TGF_BETA_SIGNALING_PATHWAY data set (systematic name: M2642)41 and (B) genes directly regulated by receptor-regulated SMADs (R-SMADs; includes SMAD1/2/3/5/9) as published in the TRRUST database21 in 9-week-old DSG2-W2A (mut/mut vs wt/wt) or arrhythmogenic cardiomyopathy (ACM) patient data set 1 (ACM versus healthy control; GEO: GSE107157/GSE107480). Indicated P values are calculated by function cameraPR of the R package limma. C, Immunostaining of phosphorylated SMAD2/3 (magenta, S465/S467 or S423/S425, respectively) in sections of DSG2-W2A hearts and related analysis of nuclear staining intensity. Nuclei are stained with DAPI (blue), cardiomyocytes are marked with f-actin (green). The dotted orange line marks the edge of the fibrotic area. Scale bar, 50 µm. *P<0.05, unpaired Student t test. D, Schematic of experimental setup for integrin-αVβ6 (ITGAV/B6) blocking experiments in cardiac slice culture with related results in E. Icons are derived from BioRender. E, Reverse transcription quantitative polymerase chain reaction analysis of expression of genes downstream of transforming growth factor–β (TGF-β) signaling in cardiac slice cultures treated with inhibiting anti-ITGAV/B6 (1:15) or 10 µmol/L GW788388, an inhibitor of TGF-β receptor I, for 24 hours. *P< 0.05, paired Student t test versus indicated control condition. F, Schematic of experimental setup for in vivo ITGAV/B6 blocking experiments by injection of 40 mg/kg EMD527040 (EMD) intraperitoneally daily. DMSO was applied as vehicle control. Icons are derived from BioRender. G, Reverse transcription quantitative polymerase chain reaction expression analysis of genes downstream of TGF-β signaling in hearts of mice treated with EMD or respective amount of DMSO for 10 days. *P<0.05, unpaired Student t test versus DMSO. H and I, Cardiac fibrosis detected by picrosirius red collagen staining with representative images and corresponding analysis of the area of collagen in the right ventricle (RV) and left ventricle (LV). Lines indicate littermates. Each dot represents 1 animal. *P<0.05 or as indicated, grouped 2-way repeated-measures analysis of variance with LV/RV and experimental pairs matched, Sidak post hoc test. Scale bar, 1 mm. J through L, ECG recoded in lead II with representative curves shown in J. Corresponding analysis of R, S, and J peak amplitude and QRS interval, *P<0.05 or as indicated. Paired Student t test. Lines indicate littermates. Each dot represents 1 animal. [SUBTITLE] Fibrosis in DSG2-W2A Mutant Animals Is Reduced by Inhibition of ITGAV/B6-Dependent Release of TGF-β [SUBSECTION] On the basis of the known profibrotic role of TGF-β in cardiomyopathies2 and the established function of ITGAV/B6 dimers to activate TGF-β,17 we investigated whether inhibition of ITGAV/B6 was efficient to reduce fibrosis in DSG2-W2A mice. Cardiac slice cultures were generated from the ventricles of heterozygous mice at the age of 40 to 50 weeks and treated for 24 hours with anti-ITGAV/B6, which was shown to neutralize the function of the dimer.22 In comparison, slices were treated with the TGF-β receptor I inhibitor GW788388 to directly block TGF-β signaling (Figure 6D). Significant downregulation in the expression of the profibrotic molecules collagen-I type-α1 (Col1a1), laminin subunit-γ2 (Lamc2), metalloproteinase inhibitor-1 (Timp1), and ID-2 (Id2) was induced in the mutant mice in response to inhibition of ITGAV/B6 (Figure 6E). Other extracellular matrix proteins such as fibronectin (Fn1) or collagens (Col3a1, Col1a2) showed the same tendency. All of these fibrotic markers were significantly upregulated in adult mutant hearts as detected by RNA sequencing (Figure 6A and 6B). Direct inhibition of TGF-β led to a similar downregulation of these markers as the blocking antibody. To investigate the in vivo relevance of this pathway, we applied the small molecule EMD527040 (EMD), which is an established inhibitor of ITGAV/B6-dependent TGF-β release, to the DSG2-W2A mouse model.23 For these experiments, we chose wt/mut mice because they reflect the heterozygosity most commonly found in patients with ACM and develop the phenotype during adulthood, when compound application is feasible. Littermates between the ages of 28 and 38 weeks were sex-matched and injected intraperitoneally daily for 10 days with EMD or DMSO control (Figure 6F). In this short-time approach, myocardial samples of EMD-treated animals showed a similar reduction of profibrotic markers as reported for slice cultures (Figure 6G). Having established an effective dose, we performed in vivo studies for a duration of 60 days during the time period when RV fibrosis establishes. At the end of the treatment period, ECG and histologic analysis were performed. EMD-treated mice demonstrated reduced RV fibrosis (Figure 6H and 6I). Moreover, EMD animals showed a mitigation of ECG abnormalities, with significantly higher S amplitude and trends towards an increased R amplitude and shorter QRS interval compared with the vehicle-treated control. These experiments link ITGAV/B6 dimerization in response to abrogated DSG2 binding and ICD dysfunction to TGF-β–dependent fibrosis generation. The pilot inhibitor studies suggest ITGAV/B6 as a promising treatment target to ameliorate the ACM phenotype. On the basis of the known profibrotic role of TGF-β in cardiomyopathies2 and the established function of ITGAV/B6 dimers to activate TGF-β,17 we investigated whether inhibition of ITGAV/B6 was efficient to reduce fibrosis in DSG2-W2A mice. Cardiac slice cultures were generated from the ventricles of heterozygous mice at the age of 40 to 50 weeks and treated for 24 hours with anti-ITGAV/B6, which was shown to neutralize the function of the dimer.22 In comparison, slices were treated with the TGF-β receptor I inhibitor GW788388 to directly block TGF-β signaling (Figure 6D). Significant downregulation in the expression of the profibrotic molecules collagen-I type-α1 (Col1a1), laminin subunit-γ2 (Lamc2), metalloproteinase inhibitor-1 (Timp1), and ID-2 (Id2) was induced in the mutant mice in response to inhibition of ITGAV/B6 (Figure 6E). Other extracellular matrix proteins such as fibronectin (Fn1) or collagens (Col3a1, Col1a2) showed the same tendency. All of these fibrotic markers were significantly upregulated in adult mutant hearts as detected by RNA sequencing (Figure 6A and 6B). Direct inhibition of TGF-β led to a similar downregulation of these markers as the blocking antibody. To investigate the in vivo relevance of this pathway, we applied the small molecule EMD527040 (EMD), which is an established inhibitor of ITGAV/B6-dependent TGF-β release, to the DSG2-W2A mouse model.23 For these experiments, we chose wt/mut mice because they reflect the heterozygosity most commonly found in patients with ACM and develop the phenotype during adulthood, when compound application is feasible. Littermates between the ages of 28 and 38 weeks were sex-matched and injected intraperitoneally daily for 10 days with EMD or DMSO control (Figure 6F). In this short-time approach, myocardial samples of EMD-treated animals showed a similar reduction of profibrotic markers as reported for slice cultures (Figure 6G). Having established an effective dose, we performed in vivo studies for a duration of 60 days during the time period when RV fibrosis establishes. At the end of the treatment period, ECG and histologic analysis were performed. EMD-treated mice demonstrated reduced RV fibrosis (Figure 6H and 6I). Moreover, EMD animals showed a mitigation of ECG abnormalities, with significantly higher S amplitude and trends towards an increased R amplitude and shorter QRS interval compared with the vehicle-treated control. These experiments link ITGAV/B6 dimerization in response to abrogated DSG2 binding and ICD dysfunction to TGF-β–dependent fibrosis generation. The pilot inhibitor studies suggest ITGAV/B6 as a promising treatment target to ameliorate the ACM phenotype.
null
null
[ "What Is New?", "What Are the Clinical Implications?", "Human Heart Samples", "Animal Experiments", "Statistics and Data Compilation", "W2A Mutation Abrogates DSG2 Interactions and Impairs Cell–Cell Adhesion In Vitro and In Vivo", "DSG2-W2A Mutant Mice Resemble the Phenotype of ACM", "DSG2-W2A Mutant Hearts Present With Structurally Impaired ICDs", "Integrin-αV/β6 Is Activated in DSG2-W2A Mutant Hearts", "TGF-β Signaling Is Upregulated in DSG2-W2A Mutant Hearts", "Fibrosis in DSG2-W2A Mutant Animals Is Reduced by Inhibition of ITGAV/B6-Dependent Release of TGF-β", "Defective Desmosomal Adhesion by DSG2-W2A Mutation Is Inducing an ACM Phenotype", "DSG2-W2A Mice as Model to Study ACM", "DSG2-W2A Leads to Dysfunctional ICDs and ITGAV/B6 Rearrangement", "Activation of TGF-β Signaling by ITGAV/B6 as Potential Therapeutic Target in ACM", "Article Information", "Acknowledgments", "Sources of Funding", "Supplemental Material" ]
[ "The tryptophan residue at position 2 of the desmosomal adhesion molecule DSG2 (desmoglein-2) is central for its binding function and cell–cell adhesion in vitro and in vivo.\nMice with abrogated DSG2 binding function (DSG2-W2A) develop a cardiac phenotype recalling arrhythmogenic cardiomyopathy with fibrosis, impaired systolic function, ECG abnormalities, and ventricular arrhythmia.\nIncreased integrin-αVβ6–dependent transforming growth factor–β signaling was identified as a driver of fibrosis.", "We provide a new mouse model reproducing major features of arrhythmogenic cardiomyopathy that can be applied to study disease mechanisms and test therapeutic approaches.\nThe model suggests that loss of mechanical cardiomyocyte coupling is a central and early event causing arrhythmogenic cardiomyopathy.\nOur pilot in vivo study highlights the applicability and potential to target integrin-αVβ6–dependent transforming growth factor–β release as new therapeutic approach to diminish the development of fibrosis in arrhythmogenic cardiomyopathy.", "Heart samples of patients with ACM and healthy controls were derived from forensic autopsy. This study was conducted according to the tenets of the Declaration of Helsinki and approved by the local ethics committees (license number 494-16 from LMU Munich and license number 152/15 from Goethe University). Samples were fixed and embedded in paraffin according to standard procedures.", "Mouse experiments were approved by the Cantonal Veterinary Office of Basel-Stadt (license numbers 2973_32878 and 3070_32419). Mice were housed under specific pathogen-free conditions with standard chow and bedding with a 12-hour day/night cycle according to institutional guidelines. Animals of both sexes were applied without bias. For inhibitor treatments, mice from the same litter and sex were paired and allocated randomly to the groups.", "Figures were compiled with Adobe Photoshop CC 2017 and Adobe Illustrator CC 2017. Statistical computations were performed with Prism 8 (GraphPad Software). For comparison of 2 or multiple groups, distribution of data was analyzed by a Shapiro-Wilk normality test, and group variances were analyzed by an F-test or a Brown-Forsythe test, respectively. According to the results of these tests, a parametric or nonparametric test with or without Welch correction for unequal variances was applied. For Kaplan-Meier survival analysis, significance was assessed by the Gehan-Breslow-Wilcoxon test. The statistical test used to compare the respective data sets is described in the corresponding figure legend. Statistical significance was assumed at P<0.05. Unless otherwise stated, data are presented as dot blot, with each dot representing the mean of the respective technical replicates of 1 biological replicate. Each animal or independent seeding of cells was taken as biological replicate, or as indicated in the legend. The mean value of these dots is shown as bar diagram ± SD.", "To study the functional consequences of impaired DSG2 binding, we generated the W2A point mutation (DSG2-W2A) to abrogate the proposed interaction mechanism of the only desmoglein expressed in cardiomyocytes (Figure 1A). First, we investigated in a cell-free setup whether DSG2-W2A is indeed affecting the binding properties of DSG2 interaction by applying single molecule force spectroscopy. Here, molecules are coupled to the tip of an atomic force microscopy probe and the surface of a mica sheet by a polyethylene glycol linker. By measuring the deflection of the probe during repeated approach to and retraction from the surface, binding events can be assessed quantitatively. From these data, properties such as binding frequency, binding force, and bond half-life time can be determined. Constructs were generated for expression of wild-type (WT) and mutant (W2A) DSG2 extracellular domains fused to a human Fc fragment and tested for homotypic (WT:WT, W2A:W2A) and heterotypic (WT:W2A) interaction properties (Figure 1B). These experiments showed a significant reduction of the frequency of W2A:W2A as well as WT:W2A interactions compared with WT:WT (Figure 1C). The frequency of the remaining W2A bindings was comparable to probing Fc alone, which indicates that these are mostly nonspecific interactions. Homotypic WT interactions display a clear binding force peak, whereas the remaining W2A interaction forces were spread widely, again pointing to the nonspecificity of these interactions (Figure 1D). The lifetime of DSG2 interactions under zero force (τ0) was determined by fitting the binding forces detected at different loading rates,11 yielding τ0=1.088s with R=0.993. In contrast, no sufficient fit (R=0.509) was possible for W2A interactions (Figure 1E). Together, these data outline that the tryptophan swap is the major interaction mechanism of DSG2.\nDSG2 (desmoglein-2) adhesion is mediated by tryptophan swap at position 2. A, Predicted interaction model of DSG2 extracellular domains by exchange of tryptophan residue at position 2 into a hydrophobic pocket of the opposing molecule. Cartoon 3D presentation of Protein Data Bank entry 5ERD9; tryptophan-2 is highlighted by ball and stick presentation. B, Schematic of single molecule force spectroscopy experiments. Recombinant extracellular domains (ECs) of wild-type DSG2 (DSG2-WT) or mutant DSG2 (DSG2-W2A) protein were coupled to a mica surface and atomic force microscopy cantilever by mean of a human Fc-tag (hFc) and a polyethylene glycol (PEG)20 linker and probed as indicated. C, Binding frequency of DSG2-W2A/DSG2-WT heterotypic and homotypic interactions at a pulling speed of 2 µm/s are shown. hFc served as control for nonspecific binding. *P<0.05, 1-way analysis of variance, Dunnett post hoc test. Each independent coating procedure with minimum 625 force curves is taken as biological replicate. D, Histogram of binding forces distribution with peak fit at a pulling speed of 2 µm/s corresponding to data in C. E, Determination of the bond half lifetime by the Bell equation11 of mean loading rates and binding forces analyzed from data of pulling speeds at 0.5, 1, 2, 5, and 7.5 µm/s. Average of values from 4 independent coating procedures with minimum 625 force curves each. R=R2, koff =off-rate constant, τ0=bond half lifetime under zero force. F, Dissociation assays to determine cell–cell adhesion were performed in CaCo2 cells (WT or DSG2 KO background) expressing DSG2-WT-mGFP or DSG2-W2A-mGFP constructs. mGFP empty vector served as control. *P<0.05, 1-way analysis of variance, Sidak post hoc test. Corresponding Western blot analysis confirmed effective expression of DSG2 constructs (*) versus the endogenous protein (arrow) in CaCo2 cells. α-tubulin (TUBA) served as loading control. G, Representative images of monolayer fragmentation from experiments in F. H, Dissociation assays in immortalized keratinocytes isolated from neonatal murine skin of the respective genotype. *P<0.05 or as indicated, Welch analysis of variance, Dunnett post hoc test. I, Macroscopic cardiac phenotype of DSG2-W2A mut/mut mice at age 15 weeks. J, Cardiac hypertrophy was analyzed as mean cross-sectional area of cardiomyocytes in hematoxylin & eosin–stained sections. Scale bar, 30 µm. *P<0.05, unpaired Student t test. K, Representative images of intercalated discs acquired by transmission electron microscopy, 3 mice per genotype. Orange asterisks mark intercellular widening, orange circle marks a ruptured junction. Scale bar, 1 µm.\nTo determine the effect of the W2A mutation on a cellular level, DSG2-WT and DSG2-W2A constructs were stably expressed in the epithelial cell line CaCo2 either in a WT or DSG2-deficient background (CaCo2[WT] or CaCo2[DSG2 KO]).12 Cell–cell dissociation assays were performed, in which a confluent cell monolayer is detached and exposed to defined mechanical stress. Expression of DSG2-W2A in CaCo2(WT) cells significantly reduced intercellular adhesion as indicated by an increased number of fragments (Figure 1F and 1G). Whereas expressing DSG2-WT in the CaCo2(DSG2 KO) line significantly rescued the impaired intercellular adhesion in response to DSG2 loss, the expression of DSG2-W2A had no effect on fragment numbers.\nBecause these data demonstrate the DSG2 tryptophan swap to be the central adhesive mechanism, we next generated a CRISPR/Cas9-based knock-in mouse model for DSG2-W2A to investigate the consequences of impaired DSG2 adhesion in vivo (Figure S1). In line with the studies in human cells, dissociation assays in keratinocyte cell lines generated from DSG2-W2A pups revealed reduced cell–cell adhesion in heterozygous (mut/wt) and homozygous (mut/mut) mutants (Figure 1H). Moreover, DSG2-W2A mice presented with a pronounced cardiac phenotype. Adult mut/mut mice demonstrated ventricular deformation, fibrotic and calcified areas, and hypertrophic cardiomyocytes (Figure 1I and 1J). In line with reduced intercellular adhesion of cardiomyocytes, transmission electron microscopy revealed disturbed ICDs with widened intercellular space and occasionally completely ruptured junctions in mut/mut hearts (Figure 1K).\nA cardiac phenotype was also detectable during embryogenesis. Here, mating of heterozygous mice revealed a reduction of mut/mut offspring to 2.8% compared with the expected Mendelian ratio of 25%. Embryo dissections showed loss of the mut/mut animals between E12 and E14 (Figure S2A). The mut/mut embryos at day E12.5 appeared macroscopically pale with accumulation of blood in the cardiac area although the heart was still beating (Figure S2B). Cells suggestive for blood precursors were detectable in the pericardial space (Figure S2C). This finding suggests a rupture of the cardiac wall leading to pericardial bleeding and loss of mut/mut animals.\nTogether, these in vitro and in vivo data outline an essential role of the DSG2 tryptophan swap for cell–cell adhesion and demonstrate severe pathologies associated with impaired DSG2 interaction.", "Given the cardiac affection in mutant mice, the notion that a majority of patients with ACM are reported with mutations in desmosomal genes,2 and that mutations specifically of W2 have already been associated with ACM,10 we examined homozygous and heterozygous mice including both sexes with regard to histologic and clinical ACM measures in an age range of 12 to 80 weeks.\nHistologic analysis showed cardiac fibrosis, a major hallmark of ACM, in the myocardium of the right ventricle (RV) and left ventricle (LV) in mut/mut animals (Figure 2A through 2D), which did not profoundly increase over time. In contrast, heterozygous animals demonstrated a milder phenotype, with a fraction of animals (36%) starting to develop fibrosis in the RV around 6 months, whereas LV was unaffected. To better address differences over time, we pooled animals into groups with a mean age of 4 and 6 months for homozygous and 6 and 12 months for heterozygous mice and performed functional analyses by echocardiography and ECG parallel to histologic evaluation. Moreover, we separated 12-month-old heterozygous animals according to RV fibrosis into a group without and with fibrosis (>10% fibrotic area). Mut/mut animals showed impaired RV systolic function at both time points with reduced tricuspid annular plane systolic excursion as well as fractional shortening (Figure 2E through 2G and Table S1). LV systolic impairment with reduction of the ejection fraction and corresponding parameters worsened over time and became significant after 6 months, although no increase in fibrosis was detectable (Figure 2D and 2H and Table S1). In mut/wt mice, RV fractional shortening was significantly reduced in 12-month-old animals with fibrosis. In ECG recordings of mut/mut mice for 30 minutes under anaesthesia, we noted deformation of the QRS complex with elongated QRS interval and reduced amplitude of the S peak with effects more pronounced in the older group; heterozygous animals with RV fibrosis showed similar effects or trends (Figure 2I through 2K and Table S1). Because the T wave was not clearly detectable in mutants, the J peak amplitude as indicator for early repolarization was determined. This was reduced in the mut/mut groups with same trend for fibrotic mut/wt hearts (Figure 2L). Parallel to these depolarization and repolarization abnormalities, we also noted the occurrence of ventricular arrhythmia, ranging from single premature ventricular contractions (PVCs) to multiple, multifocal PVCs and nonsustained ventricular tachycardia in 1 mouse (Figure 2M through 2O). The fraction of homozygous and heterozygous animals with PVCs and the frequency of these events correlated with age and fibrosis. Moreover, a substantial fraction (16%) of mut/mut animals was found dead during the observation period, which strongly suggests the occurrence of malignant arrhythmia with sudden death (Figure 2P). Investigating sex-related differences, no effect was observed on the extent of fibrosis or echocardiographic parameters. However, in mut/wt mice, PVCs were found in males only and male mut/mut mice were more prone to premature sudden death compared with females (Figure S3).\nDSG2 (desmoglein-2)–W2A mutant mice develop characteristics of arrhythmogenic cardiomyopathy. A, Cardiac fibrosis detected by picrosirius red collagen staining with representative images of sections from 6-month-old DSG2-W2A mut/mut and 12-month-old wt/wt and mut/wt animals. Scale bar = 1 mm. B through D, Corresponding analysis of the area of collagen in the right ventricle (RV) and left ventricle (LV). Continuous lines indicate the simple linear regression between age and area of collagen. Hearts with >10% of collagen in the RV (gray dotted line) were defined as “with fibrosis.” Each dot represents 1 animal. *P<0.05, mixed effects analysis with LV and RV matched per animal, Sidak post hoc test. Lines indicate median and quartile values. E, Representative echocardiography images for measurements of the tricuspid annular plane systolic excursion (TAPSE) and LV function in 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Left side: 2D images from apical 4-chamber view for TAPSE and parasternal short axis view for LV. M-mode tracings on the right were performed along the line indicated on the left. Scale bars, white, 2 mm; black, 100 ms. Corresponding analysis of (F) TAPSE, *P<0.05, Kruskal-Wallis test with Dunn post hoc test; (G) fractional shortening of the RV, *P<0.05, Kruskal-Wallis test with Dunn post hoc test; and (H) ejection fraction of the LV, *P<0.05, 1-way analysis of variance, Sidak post hoc test. I, ECG recoded in lead II with representative curves from 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Definition of respective peaks is indicated in the wt/wt curve. Scale bars, vertical, 0.5 mV; horizontal, 50 ms. Corresponding analysis of (J) QRS interval, *P<0.05 or as indicated, Kruskal-Wallis test with Dunn post hoc test; (K) amplitude of the S peak, *P<0.05 or as indicated, 1-way analysis of variance, Sidak post hoc test; and (L) amplitude of the J peak (early repolarization), *P<0.05 or as indicated, Kruskal-Wallis test with Dunn post hoc test. M, Ventricular arrythmia detected by ECG during 30 minutes of baseline measurements with example curves from 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Asterisks mark premature ventricular contractions (PVCs); *nsVT indicates a nonsustained ventricular tachycardia detected in 1 mut/mut animal. Scale bars, vertical, 0.5 mV; horizontal, 1 s. Corresponding analysis of (N) percentage of mice presenting with PVCs. Values in bars indicate corresponding absolute number of mice with PVC (colored bars) compared with total number of mice evaluated (empty bar) and (O) PVC burden depicted as number of PVCs per minute, *P<0.05, Kruskal-Wallis test with Dunn post hoc test. The black arrow indicates the animal presenting with nonsustained ventricular tachycardia. P, Kaplan-Meier survival plot of DSG2-W2A mice from an analysis period of 3 years. Vertical lines indicate dropouts because of unrelated elimination (end of experiment, breeding, injuries). Values indicate corresponding absolute number of mice with sudden death compared with total number of mice evaluated. *P<0.05, Gehan-Breslow-Wilcoxon test. Box with color indications of respective groups in the middle applies to the entire figure.\nIn summary, mutant animals show histopathologic features with echocardiography and ECG abnormalities similar to patients with ACM. Within the limits of a murine model, both genotypes fulfil the Padua criteria13 to establish the diagnosis of ACM (Table S2). Here, DSG2-W2A mut/mut animals resemble the phenotype of biventricular ACM, whereas in mut/wt mice, the phenotype is milder with variable penetrance and age-dependent occurrence only in the RV. According to the Padua criteria,13 the heterozygous genotype recalls the characteristics of a right-dominant ventricular ACM, as long as fibrosis was present. Together, these data demonstrate that loss of desmosomal adhesion is sufficient to induce an ACM phenotype and that the DSG2-W2A model may resemble 2 different variants of the disease.", "This result together with the transmission electron microscopy data (Figure 1K) suggest structural changes of the ICD in mutant hearts, which we addressed further. Using DSP as marker, we reconstructed the outlines of ICDs in wt/wt and mut/mut hearts from 3D stacks captured with structured illumination microscopy. This analysis yielded an increase in ICD volume, in particular attributable to an enhanced width between 2 adjacent cells (Figure 4A). The majority of desmosomal molecules as well as N-cadherin were regularly distributed at mutant ICDs (Figure S5A and S5B). This was corroborated by RNA sequencing data from 5-day-old and 9-week-old animals, which showed no consistent changes of desmosomal, adherens, gap, or tight junction molecules (Figure S5C through S5E). The only adhesion molecule reduced globally was DSG2 (Figure S5A and S5B). Although DSG2 was still present at the ICD, detailed analysis by structured illumination microscopy revealed reduced number and volume of DSG2 signals (Figure 4B). Moreover, fluorescence recovery after photobleaching experiments in neonatal murine cardiomyocytes transduced with DSG2-WT-mGFP or DSG2-W2A-mGFP showed a higher mobility of the mutant protein at cardiomyocyte junctions (Figure 4C), indicating reduced membrane stability of DSG2-W2A. To further investigate the possibility that the stability and integrity of ICD molecules is compromised, we performed Triton-X-100 separation assays for proteins with unaltered ICD localization in mutant hearts. Cytosolic and unanchored membrane proteins are found in the Triton-X-100 soluble fraction, whereas cytoskeleton-bound molecules are mainly detectable in the nonsoluble fraction. Both desmosomal molecules (PG, PKP2) as well as N-cadherin showed increased levels in the soluble pool, indicating reduced cytoskeletal anchorage and impaired ICD integrity in mutants (Figure 4D). Moreover, the gap junction molecule connexin-43, which is required for electrical coupling of cardiomyocytes, was delocalized to the lateral membrane (Figure 4E), which was described as a feature of disrupted ICDs.8\nDisrupted ICD structure in DSG2 (desmoglein-2)–W2A mutant mice. A, Representative z-stack reconstruction of segmented intercalated discs (ICDs) in top and side view acquired with structured illumination microscopy. Overlay of analyzed ICD volume is shown in gray. ICDs are marked by DSP (desmoplakin; magenta). Scheme on top presents segmented area and pictograms on the right display the respective angle of view. Corresponding analysis of ICD volume and width of ICD between adjacent cardiomyocytes below. Scale bar, 5 µm. *P<0.05, Mann-Whitney test. Each dot represents the value of 1 ICD from 4 mice per genotype. B, Representative images of DSG2 (magenta) and filamentary actin (f-actin; white) z-stacks acquired by structured illumination microscopy and presented as maximum intensity projection. Lower row shows color-coded height projection of DSG2 signals in z-stack after signal thresholding as performed for analysis. Related analysis of DSG2 signal volume and number per ICD length is shown below. Pictograms on the right display the respective angle of view. Scale bar, 5 µm. *P<0.05, unpaired Student t test, with Welch correction. Each dot represents the value of 1 ICD from in total 3 mice per genotype. C, Fluorescence recovery after photobleaching analysis of DSG2–wild type (WT) and DSG2-W2A-mGFP fusion proteins at the cell–cell junction of neonatal cardiomyocytes with representative intensity kymographs of bleached areas on the left. Time point 0 = bleach as indicated by the black arrow. Analysis of the mobile fraction of the indicated mGFP-fusion proteins is shown on the right. *P<0.05, unpaired Student t test, with Welch correction. Each dot represents the mean value of 1 heart from in total 3 isolations. D, Representative triton-X-100 assay immunoblot with separation of a soluble (sol), noncytoskeletal bound protein fraction from a nonsoluble (non-sol), cytoskeletal-anchored fraction and corresponding analysis shown below. PG (plakoglobin), PKP2 (plakophilin-2), and N-cadherin (NCAD) were analyzed. Intensity of proteins was normalized to the total amount of protein detected by ponceau staining. GAPDH and DSP served as separation control. *P<0.05 or as indicated, unpaired Student t test (PG, NCAD, PKP2) or Mann-Whitney test (DSP). Each dot represents the result from 1 mouse. E, Immunostaining of connexin-43 (CX43; red in overlay) in DSG2-W2A hearts. NCAD (yellow) marks ICDs, DAPI (blue) nuclei, and F-actin (green) the sarcomere system. Orange arrowheads mark ICD, pink arrowheads highlight lateralization of CX43 staining. Scale bars, 25 µm. Images representative for 5 mice per genotype. Box with color indications of respective groups applies to the entire figure.\nThese data together with the transmission electron microscopy results demonstrate structurally severely altered ICDs in response to the DSG2-W2A mutation and suggest a molecular basis leading to the functional alterations detectable by echocardiography and ECG.", "In contrast to DSG2, we detected ITGB6 to be increased at the ICD (Figure 3D and 3E), which was confirmed by analysis of structured illumination microscopy images (Figure 5A). ITGB6 needs to heterodimerize with integrin-αV (ITGAV) in order to be activated and bind to the extracellular matrix.16–18 Thus, we analyzed the expression of the counterpart ITGAV, which was significantly upregulated on protein level and increased at the ICD and costameres of mutant hearts (Figure 5B and 5C). In contrast, the localization and intensity of integrin-β1, as classical representative of the integrin group, was not altered (Figure S6). Staining with an antibody specifically recognizing the heterodimer of ITGAV/B6 revealed increased amount of dimer formation at the ICD and costameres in mutant mice (Figure 5D). Moreover, increased ITGAV/B6 staining intensity was also detectable in an ACM patient sample with DSP mutation (Figure 5E). To stabilize active integrins, recruitment of the cytoskeletal adapters talin and vinculin is required.19 Accordingly, the expression of talin-2 was increased in DSG2-W2A mut/mut myocardium, with vinculin being enriched at the ICD (Figure 5F and 5G). This effect is similar to what was shown before in response to increased intracellular traction forces on junctions.20 Thus, these data suggest that in response to a dysfunctional, nonadhesive ICD with impaired cytoskeletal anchorage, ITGAV/B6 dimers are recruited and activated at the compromised ICD as well as at the costamere region.\nIncreased activation of ITGB6/AV at ICDs of DSG2 (desmoglein-2)–W2A mutant mice. A, Representative intercalated disc (ICD) reconstruction from z-stacks of integrin-β6 (ITGB6; green) and DSP (magenta) immunostaining acquired by structured illumination microscopy and presented as maximum intensity projection. Related analysis of ITGB6 signal volume and number per ICD length is shown below. Scale bar, 5 µm. *P<0.05 or as indicated, unpaired Student t test with Welch correction (left graph) and Mann-Whitney test (right graph). Each dot represents the value of 1 ICD from in total 4 mice per genotype. B, Representative Western blot and analysis of band intensity of integrin-αV (ITGAV) in DSG2-W2A hearts. GAPDH served as loading control. *P<0.05, unpaired Student t test with Welch correction. C, Immunostaining of ITGAV in DSG2-W2A hearts on the right with respective analysis on the left. DAPI (blue) marks nuclei. Lower row shows an overview image of a fibrotic area in mut/mut hearts. Dotted orange line marks the edge of fibrotic area. Scale bars: upper rows, 20 µm; lower row, 50 µm. *P<0.05, unpaired Student t test. D, Immunostaining of ITGAV/B6 heterodimer in DSG2-W2A mutant hearts with respective analysis of staining intensity on the right. Cyan rectangle marks zoomed area on the right. Scale bars: overview, 50 µm; insert, 10 µm. *P<0.05, unpaired Student t test. E, Representative immunostaining images of ITGAV/B6 heterodimer staining (red) in a patient with arrhythmogenic cardiomyopathy (ACM; DSP-E952X, heterozygous) and healthy control sample. F-actin (green) stains the sarcomere system. For the patient with ACM, 4 different tissue samples were analyzed and compared with 2 tissue samples from 2 healthy controls. Scale bar, 20 µm. F, Immunostaining of VCL (vinculin; magenta) and TLN2 (talin-2; red) in DSG2-W2A hearts on the right with respective analysis of the mean staining intensity in cardiomyocytes and ratio of the staining intensity at the ICD versus cytosolic area on the left. DAPI (blue) marks nuclei. F-actin (green) stains the sarcomere system. Scale bar, 25 µm; *P<0.05, unpaired Student t test. G, Representative Western blot and analysis of band intensity of VCL and TLN2 in DSG2-W2A hearts. GAPDH served as loading control. *P<0.05, unpaired Student t test for TLN2, Mann-Whitney test for VCL. Box with color indications of respective groups applies to the entire figure.", "ITGAV/B6 dimers have the ability to activate the profibrotic cytokine transforming growth factor–β (TGF-β) by binding and removal of the latency-associated peptide.17 Gene set enrichment analyses revealed an upregulation of genes associated with TGF-β signaling in both 9-week-old mutants as well as patients with ACM (Figure 6A). Moreover, direct targets of receptor-regulated SMADs involved in TGF-β signaling21 were upregulated in DSG2-W2A mutant hearts (Figure 6B). Accordingly, increased amounts of nuclear SMAD2/3 phosphorylated at the activation sites S465/S467 or S423/S425, respectively, were detected in these hearts (Figure 6C). Increased levels of pSMAD2/3 were found not only in fibroblasts but also in cardiomyocytes, mainly adjacent to fibrotic areas. These data indicate upregulation of the profibrotic TGF-β pathway and its downstream targets, which include extracellular matrix proteins such as collagens, laminin, or fibronectin.\nElevated transforming growth factor–β signaling in DSG2 (desmoglein-2)–W2A hearts as result of ITGAV/B6 activity. Barcode plots of gene set enrichment analysis of (A) the KEGG_TGF_BETA_SIGNALING_PATHWAY data set (systematic name: M2642)41 and (B) genes directly regulated by receptor-regulated SMADs (R-SMADs; includes SMAD1/2/3/5/9) as published in the TRRUST database21 in 9-week-old DSG2-W2A (mut/mut vs wt/wt) or arrhythmogenic cardiomyopathy (ACM) patient data set 1 (ACM versus healthy control; GEO: GSE107157/GSE107480). Indicated P values are calculated by function cameraPR of the R package limma. C, Immunostaining of phosphorylated SMAD2/3 (magenta, S465/S467 or S423/S425, respectively) in sections of DSG2-W2A hearts and related analysis of nuclear staining intensity. Nuclei are stained with DAPI (blue), cardiomyocytes are marked with f-actin (green). The dotted orange line marks the edge of the fibrotic area. Scale bar, 50 µm. *P<0.05, unpaired Student t test. D, Schematic of experimental setup for integrin-αVβ6 (ITGAV/B6) blocking experiments in cardiac slice culture with related results in E. Icons are derived from BioRender. E, Reverse transcription quantitative polymerase chain reaction analysis of expression of genes downstream of transforming growth factor–β (TGF-β) signaling in cardiac slice cultures treated with inhibiting anti-ITGAV/B6 (1:15) or 10 µmol/L GW788388, an inhibitor of TGF-β receptor I, for 24 hours. *P< 0.05, paired Student t test versus indicated control condition. F, Schematic of experimental setup for in vivo ITGAV/B6 blocking experiments by injection of 40 mg/kg EMD527040 (EMD) intraperitoneally daily. DMSO was applied as vehicle control. Icons are derived from BioRender. G, Reverse transcription quantitative polymerase chain reaction expression analysis of genes downstream of TGF-β signaling in hearts of mice treated with EMD or respective amount of DMSO for 10 days. *P<0.05, unpaired Student t test versus DMSO. H and I, Cardiac fibrosis detected by picrosirius red collagen staining with representative images and corresponding analysis of the area of collagen in the right ventricle (RV) and left ventricle (LV). Lines indicate littermates. Each dot represents 1 animal. *P<0.05 or as indicated, grouped 2-way repeated-measures analysis of variance with LV/RV and experimental pairs matched, Sidak post hoc test. Scale bar, 1 mm. J through L, ECG recoded in lead II with representative curves shown in J. Corresponding analysis of R, S, and J peak amplitude and QRS interval, *P<0.05 or as indicated. Paired Student t test. Lines indicate littermates. Each dot represents 1 animal.", "On the basis of the known profibrotic role of TGF-β in cardiomyopathies2 and the established function of ITGAV/B6 dimers to activate TGF-β,17 we investigated whether inhibition of ITGAV/B6 was efficient to reduce fibrosis in DSG2-W2A mice. Cardiac slice cultures were generated from the ventricles of heterozygous mice at the age of 40 to 50 weeks and treated for 24 hours with anti-ITGAV/B6, which was shown to neutralize the function of the dimer.22 In comparison, slices were treated with the TGF-β receptor I inhibitor GW788388 to directly block TGF-β signaling (Figure 6D). Significant downregulation in the expression of the profibrotic molecules collagen-I type-α1 (Col1a1), laminin subunit-γ2 (Lamc2), metalloproteinase inhibitor-1 (Timp1), and ID-2 (Id2) was induced in the mutant mice in response to inhibition of ITGAV/B6 (Figure 6E). Other extracellular matrix proteins such as fibronectin (Fn1) or collagens (Col3a1, Col1a2) showed the same tendency. All of these fibrotic markers were significantly upregulated in adult mutant hearts as detected by RNA sequencing (Figure 6A and 6B). Direct inhibition of TGF-β led to a similar downregulation of these markers as the blocking antibody.\nTo investigate the in vivo relevance of this pathway, we applied the small molecule EMD527040 (EMD), which is an established inhibitor of ITGAV/B6-dependent TGF-β release, to the DSG2-W2A mouse model.23 For these experiments, we chose wt/mut mice because they reflect the heterozygosity most commonly found in patients with ACM and develop the phenotype during adulthood, when compound application is feasible. Littermates between the ages of 28 and 38 weeks were sex-matched and injected intraperitoneally daily for 10 days with EMD or DMSO control (Figure 6F). In this short-time approach, myocardial samples of EMD-treated animals showed a similar reduction of profibrotic markers as reported for slice cultures (Figure 6G). Having established an effective dose, we performed in vivo studies for a duration of 60 days during the time period when RV fibrosis establishes. At the end of the treatment period, ECG and histologic analysis were performed. EMD-treated mice demonstrated reduced RV fibrosis (Figure 6H and 6I). Moreover, EMD animals showed a mitigation of ECG abnormalities, with significantly higher S amplitude and trends towards an increased R amplitude and shorter QRS interval compared with the vehicle-treated control.\nThese experiments link ITGAV/B6 dimerization in response to abrogated DSG2 binding and ICD dysfunction to TGF-β–dependent fibrosis generation. The pilot inhibitor studies suggest ITGAV/B6 as a promising treatment target to ameliorate the ACM phenotype.", "Because of mutations mainly affecting desmosomal genes and evidence of disrupted ICDs, the hypothesis of dysfunctional desmosomes with loss of cell–cell adhesion as a central pathologic step was adopted in the field.2 However, experimental data on this topic are contradictory.6–8 We show that W2 of DSG2 is central for binding and intercellular adhesion in vitro and in vivo. Disrupted desmosomal adhesion is sufficient to induce an ACM phenotype fulfilling the Padua criteria,13 including ventricular fibrosis, depolarization and repolarization abnormalities, arrhythmia, and impaired ventricular function, which are used to establish the diagnosis. In line with this, 3 mutations were described in patients with ACM directly affecting the DSG2 binding mechanism by exchange of the tryptophan with a serine, leucine, or arginine (ClinVar data bank: VCV000199796, VCV000044282, VCV000420241).10 This indicates a disruption of the tryptophan swap independent from the substituting amino acid and supports defective desmosomal adhesion as important factor in ACM.2,24 Moreover, different published DSG2 mutant mouse models show a phenotype resembling the features of DSG2-W2A mutants. These include a DSG2 mutant in which parts of the 2 outermost DSG2 extracellular domains were deleted (but leaving W2A intact),25 2 mouse lines with loss of DSG2,25,26 and mice overexpressing a patient mutation (DSG2-N271S),27 which also showed that ICD structural aberrations precede functional abnormalities and fibrosis generation. These data fuel the hypothesis that disruption and rearrangement of the ICD in response to impaired adhesion between cardiomyocytes is a crucial initial step, at least in case of mutations in desmosomal molecules. Other mechanisms described in patients or translational models, such as aberrant WNT or Hippo/YAP signaling, or immune cell infiltrations,2,28 may be secondary responses and in part even represent adaptive attempts to rescue the functional consequences of such severe structural aberrations.", "Our results suggest the DSG2-W2A model as a valuable tool to study ACM mechanisms. Within the limitations of a murine model, it reproduces a majority of features found in patients with ACM (Table S2). In homozygous animals, both ventricles are affected from early on in life; heterozygous animals develop a milder phenotype, with fibrosis occurring only in the RV. Whether this milder phenotype would extend to the LV over time or whether a gene dose effect underlies the structural differences is unclear. LV and RV remodel differently in response to loading and injury29 and it is possible that the RV is less able to compensate a partial reduction of cardiomyocyte cohesion, in contrast to the LV. A fraction of mut/wt animals did not develop the phenotype at all within the observation period of up to 80 weeks. Thus, the model might help clarify why patients with the same mutation (ie, in the same family) develop the disease with variable penetrance.1 Similar to patients,1 male mutant mice appear to experience more arrhythmia and sudden death than do female mice.\nWe can interpret from the combined histology, echocardiography, and ECG data that the changes in heart function are secondary to fibrosis generation. This is indicated by the notion that functional measures (eg, ejection fraction, QRS interval) worsened over time in homozygous mutants, whereas no increase was detectable for fibrosis. Moreover, functional changes in mut/wt animals were only observed when fibrosis was present. This is in line with a patient cohort with DSP mutations, in which LV fibrosis preceded systolic dysfunction.30 A different study in DSG2-N271S-expressing ex vivo paced hearts demonstrated conduction velocity impairments before onset of fibrosis.27 More longitudinal studies using different mutational backgrounds and sensitive detection methods (eg, cardiac magnetic resonance imaging and ECG under exercise) are necessary to define the role of fibrosis as cause or consequence of functional changes.\nAs with all murine models, the DSG2-W2A line has limitations in its ability to fully recapitulate the human disease phenotype. Mutant mice do not show pronounced replacement with adipose tissue, which is a common characteristic in patient heart samples, but in general is not fully modeled in mice.24 Moreover, at least in ECGs under anesthesia, not all mutant animals develop arrhythmia, even if fibrosis is present. However, in contrast to most other ACM models, this knock-in model has the advantage of a single amino acid exchange under the endogenous promotor, thus reducing the possibility of secondary unwanted effects attributable to complete protein absence or overexpression. As in patients, the mutation is present in all cell types and not limited to cardiomyocytes.2,24", "DSG2-W2A led to a severely altered ICD structure consistent with impaired cytoskeletal attachment and ruptured junctions. Whereas these changes provide explanations for compromised intercellular adhesion, we also noted alterations in cell–matrix protein distribution. RNA sequencing before and after onset of the disease phenotype and a comparison with ACM patient datasets identified ITGB6 as commonly deregulated in patients with ACM and DSG2-W2A mice. Although we noted reduced mRNA levels in mutant hearts, the overall protein content was unaltered. This suggests pronounced posttranslational regulation of ITGB6, which was already demonstrated in skeletal muscle.31 In mutant hearts, ITGB6 was increased at the ICD together with elevated levels of ITGAV and increased heterodimerization of both molecules. By mechanical force exerted through ITGAV/B6, TGF-β is detached from the latency-associated peptide and can induce signaling by means of TGF-β receptors.17,32 In line with higher intracellular forces, talin-2 and vinculin were increased at ICDs, as binding of these molecules activates and stabilizes integrins. A mutual regulation of both adhesive compartments is well-established. As an example, upon loss of N-cadherin, integrins are activated and induce fibronectin deposition.33 A similar mechanism is conceivable in DSG2-W2A hearts, either by loss of DSG2 or because of reduced N-cadherin anchorage. So far, only limited data are available on the role of integrins in ACM. A recent study showed downregulation of integrin-β1D leading to ventricular arrhythmia.34 Furthermore, knockdown of PKP2 in HL-1 cardiomyocytes was described to deregulate focal adhesions, including integrin-α1.35", "The ITGAV/B6 heterodimer is described as one of the major activators of latent TGF-β1 and TGF-β3.17 Although TGF-β signaling is known as general driver of cardiac fibrosis36 and more specifically was implicated in ACM,2 to our knowledge no data are available on the role of ITGAV/B6 and their regulation of TGF-β signaling in cardiac fibrosis. Uncovering this pathway is of high interest as it offers the possibility to target TGF-β with reduced risk of severe side effects occurring in response to direct inhibition.37 We demonstrate a reduction of profibrotic gene expression under ACM conditions in response to ITGAV/B6 inhibition and our pilot study data suggest that a small molecule blocking ITGAV/B6-dependent TGF-β release diminished the generation of fibrosis. Similar approaches using this small molecule or neutralizing antibodies were shown to be protective in murine models of lung, liver, and biliary fibrosis.23,38,39 However, because of the heterogeneity of the phenotype in heterozygous animals, more detailed studies with larger sample sizes are required to further substantiate this finding.\nWhether fibrosis generation is a contributor to the disease or a protective mechanism to ensure integrity of the heart under conditions of compromised adhesion is unclear. In case of the latter, pharmacologic inhibition of fibrosis generation might be detrimental. Nevertheless, fibrosis is a driver of arrhythmia generation,40 which is also supported by our results that ECG abnormalities were reduced by inhibition of fibrosis. Careful studies using different therapeutic approaches to reduce fibrosis in appropriate model organisms need to address this aspect in detail.\nIn conclusion, we established a new mouse model phenocopying many aspects of ACM, uncovered a novel pathway of fibrosis induction, and identified an approach to target this mechanism with future implication as a potential therapeutic option.", "[SUBTITLE] Acknowledgments [SUBSECTION] The authors thank Alain Brühlhart and the team from the Animal Facility; Dr Cinzia Tiberi (Center for Cellular Imaging and NanoAnalytics); Dr Alexia Loynton-Ferrand (Imaging Core Facility); Dr Diego Calabrese (Histology Core Facility); and Drs Mike Abanto and Beat Erne, P. Lorentz, and E. Bartoszek (Microscopy Core Facility), University of Basel, Switzerland; and Dr Christian Beisel and Philippe Demougin, Genomics Facility Basel (D-BSSE, ETH Zürich, and University of Basel), for support; Nicolas Schlegel (Department of Surgery, University Hospital Würzburg, Germany) for providing CaCo2 cells; Nikola Golenhofen (Institute of Anatomy, University of Ulm, Germany) for providing the Fc-His-pEGFP-N3 plasmid; and Anja Fuchs for technical assistance. Calculations were performed at the sciCORE scientific computing center at University of Basel.\nThe authors thank Alain Brühlhart and the team from the Animal Facility; Dr Cinzia Tiberi (Center for Cellular Imaging and NanoAnalytics); Dr Alexia Loynton-Ferrand (Imaging Core Facility); Dr Diego Calabrese (Histology Core Facility); and Drs Mike Abanto and Beat Erne, P. Lorentz, and E. Bartoszek (Microscopy Core Facility), University of Basel, Switzerland; and Dr Christian Beisel and Philippe Demougin, Genomics Facility Basel (D-BSSE, ETH Zürich, and University of Basel), for support; Nicolas Schlegel (Department of Surgery, University Hospital Würzburg, Germany) for providing CaCo2 cells; Nikola Golenhofen (Institute of Anatomy, University of Ulm, Germany) for providing the Fc-His-pEGFP-N3 plasmid; and Anja Fuchs for technical assistance. Calculations were performed at the sciCORE scientific computing center at University of Basel.\n[SUBTITLE] Sources of Funding [SUBSECTION] This project was supported by the German Research Council (SP1300-3/1 to Dr Spindler), the Swiss National Science Foundation (197764 to Dr Spindler), the Swiss Heart Foundation (FF21098 to Drs Schinner and Spindler), the Research Fund Junior Researchers, University of Basel (3BM1079 to Dr Schinner), and the Theiler-Haag Foundation (Dr Spindler). Dr Sheikh is supported by grants from the National Institutes of Health (grant HL142251) and Department of Defense (grant W81XWH1810380).\nThis project was supported by the German Research Council (SP1300-3/1 to Dr Spindler), the Swiss National Science Foundation (197764 to Dr Spindler), the Swiss Heart Foundation (FF21098 to Drs Schinner and Spindler), the Research Fund Junior Researchers, University of Basel (3BM1079 to Dr Schinner), and the Theiler-Haag Foundation (Dr Spindler). Dr Sheikh is supported by grants from the National Institutes of Health (grant HL142251) and Department of Defense (grant W81XWH1810380).\n[SUBTITLE] Disclosures [SUBSECTION] Dr Sheikh is a cofounder and equity shareholder of Papillon Therapeutics Inc and is a consultant and equity shareholder of LEXEO Therapeutics Inc. The other authors have nothing to disclose.\nDr Sheikh is a cofounder and equity shareholder of Papillon Therapeutics Inc and is a consultant and equity shareholder of LEXEO Therapeutics Inc. The other authors have nothing to disclose.\n[SUBTITLE] Supplemental Material [SUBSECTION] Expanded Methods\nTables S1–S3\nFigures S1–S6\nReferences 42–48\nExpanded Methods\nTables S1–S3\nFigures S1–S6\nReferences 42–48", "The authors thank Alain Brühlhart and the team from the Animal Facility; Dr Cinzia Tiberi (Center for Cellular Imaging and NanoAnalytics); Dr Alexia Loynton-Ferrand (Imaging Core Facility); Dr Diego Calabrese (Histology Core Facility); and Drs Mike Abanto and Beat Erne, P. Lorentz, and E. Bartoszek (Microscopy Core Facility), University of Basel, Switzerland; and Dr Christian Beisel and Philippe Demougin, Genomics Facility Basel (D-BSSE, ETH Zürich, and University of Basel), for support; Nicolas Schlegel (Department of Surgery, University Hospital Würzburg, Germany) for providing CaCo2 cells; Nikola Golenhofen (Institute of Anatomy, University of Ulm, Germany) for providing the Fc-His-pEGFP-N3 plasmid; and Anja Fuchs for technical assistance. Calculations were performed at the sciCORE scientific computing center at University of Basel.", "This project was supported by the German Research Council (SP1300-3/1 to Dr Spindler), the Swiss National Science Foundation (197764 to Dr Spindler), the Swiss Heart Foundation (FF21098 to Drs Schinner and Spindler), the Research Fund Junior Researchers, University of Basel (3BM1079 to Dr Schinner), and the Theiler-Haag Foundation (Dr Spindler). Dr Sheikh is supported by grants from the National Institutes of Health (grant HL142251) and Department of Defense (grant W81XWH1810380).", "Expanded Methods\nTables S1–S3\nFigures S1–S6\nReferences 42–48" ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "What Is New?", "What Are the Clinical Implications?", "Methods", "Human Heart Samples", "Animal Experiments", "Statistics and Data Compilation", "Data Availability", "Results", "W2A Mutation Abrogates DSG2 Interactions and Impairs Cell–Cell Adhesion In Vitro and In Vivo", "DSG2-W2A Mutant Mice Resemble the Phenotype of ACM", "Integrin-β6 Expression Is Altered in ACM Patient and DSG2-W2A Mouse Samples", "DSG2-W2A Mutant Hearts Present With Structurally Impaired ICDs", "Integrin-αV/β6 Is Activated in DSG2-W2A Mutant Hearts", "TGF-β Signaling Is Upregulated in DSG2-W2A Mutant Hearts", "Fibrosis in DSG2-W2A Mutant Animals Is Reduced by Inhibition of ITGAV/B6-Dependent Release of TGF-β", "Discussion", "Defective Desmosomal Adhesion by DSG2-W2A Mutation Is Inducing an ACM Phenotype", "DSG2-W2A Mice as Model to Study ACM", "DSG2-W2A Leads to Dysfunctional ICDs and ITGAV/B6 Rearrangement", "Activation of TGF-β Signaling by ITGAV/B6 as Potential Therapeutic Target in ACM", "Article Information", "Acknowledgments", "Sources of Funding", "Disclosures", "Supplemental Material", "Supplementary Material" ]
[ "The tryptophan residue at position 2 of the desmosomal adhesion molecule DSG2 (desmoglein-2) is central for its binding function and cell–cell adhesion in vitro and in vivo.\nMice with abrogated DSG2 binding function (DSG2-W2A) develop a cardiac phenotype recalling arrhythmogenic cardiomyopathy with fibrosis, impaired systolic function, ECG abnormalities, and ventricular arrhythmia.\nIncreased integrin-αVβ6–dependent transforming growth factor–β signaling was identified as a driver of fibrosis.", "We provide a new mouse model reproducing major features of arrhythmogenic cardiomyopathy that can be applied to study disease mechanisms and test therapeutic approaches.\nThe model suggests that loss of mechanical cardiomyocyte coupling is a central and early event causing arrhythmogenic cardiomyopathy.\nOur pilot in vivo study highlights the applicability and potential to target integrin-αVβ6–dependent transforming growth factor–β release as new therapeutic approach to diminish the development of fibrosis in arrhythmogenic cardiomyopathy.", "Detailed methods are available in the Expanded Methods in the Supplemental Material.\n[SUBTITLE] Human Heart Samples [SUBSECTION] Heart samples of patients with ACM and healthy controls were derived from forensic autopsy. This study was conducted according to the tenets of the Declaration of Helsinki and approved by the local ethics committees (license number 494-16 from LMU Munich and license number 152/15 from Goethe University). Samples were fixed and embedded in paraffin according to standard procedures.\nHeart samples of patients with ACM and healthy controls were derived from forensic autopsy. This study was conducted according to the tenets of the Declaration of Helsinki and approved by the local ethics committees (license number 494-16 from LMU Munich and license number 152/15 from Goethe University). Samples were fixed and embedded in paraffin according to standard procedures.\n[SUBTITLE] Animal Experiments [SUBSECTION] Mouse experiments were approved by the Cantonal Veterinary Office of Basel-Stadt (license numbers 2973_32878 and 3070_32419). Mice were housed under specific pathogen-free conditions with standard chow and bedding with a 12-hour day/night cycle according to institutional guidelines. Animals of both sexes were applied without bias. For inhibitor treatments, mice from the same litter and sex were paired and allocated randomly to the groups.\nMouse experiments were approved by the Cantonal Veterinary Office of Basel-Stadt (license numbers 2973_32878 and 3070_32419). Mice were housed under specific pathogen-free conditions with standard chow and bedding with a 12-hour day/night cycle according to institutional guidelines. Animals of both sexes were applied without bias. For inhibitor treatments, mice from the same litter and sex were paired and allocated randomly to the groups.\n[SUBTITLE] Statistics and Data Compilation [SUBSECTION] Figures were compiled with Adobe Photoshop CC 2017 and Adobe Illustrator CC 2017. Statistical computations were performed with Prism 8 (GraphPad Software). For comparison of 2 or multiple groups, distribution of data was analyzed by a Shapiro-Wilk normality test, and group variances were analyzed by an F-test or a Brown-Forsythe test, respectively. According to the results of these tests, a parametric or nonparametric test with or without Welch correction for unequal variances was applied. For Kaplan-Meier survival analysis, significance was assessed by the Gehan-Breslow-Wilcoxon test. The statistical test used to compare the respective data sets is described in the corresponding figure legend. Statistical significance was assumed at P<0.05. Unless otherwise stated, data are presented as dot blot, with each dot representing the mean of the respective technical replicates of 1 biological replicate. Each animal or independent seeding of cells was taken as biological replicate, or as indicated in the legend. The mean value of these dots is shown as bar diagram ± SD.\nFigures were compiled with Adobe Photoshop CC 2017 and Adobe Illustrator CC 2017. Statistical computations were performed with Prism 8 (GraphPad Software). For comparison of 2 or multiple groups, distribution of data was analyzed by a Shapiro-Wilk normality test, and group variances were analyzed by an F-test or a Brown-Forsythe test, respectively. According to the results of these tests, a parametric or nonparametric test with or without Welch correction for unequal variances was applied. For Kaplan-Meier survival analysis, significance was assessed by the Gehan-Breslow-Wilcoxon test. The statistical test used to compare the respective data sets is described in the corresponding figure legend. Statistical significance was assumed at P<0.05. Unless otherwise stated, data are presented as dot blot, with each dot representing the mean of the respective technical replicates of 1 biological replicate. Each animal or independent seeding of cells was taken as biological replicate, or as indicated in the legend. The mean value of these dots is shown as bar diagram ± SD.\n[SUBTITLE] Data Availability [SUBSECTION] RNA sequencing data were deposited in the public database GEO (\naccession number GSE181868). Additional data pertaining to the current article are available from the corresponding author upon request.\nRNA sequencing data were deposited in the public database GEO (\naccession number GSE181868). Additional data pertaining to the current article are available from the corresponding author upon request.", "Heart samples of patients with ACM and healthy controls were derived from forensic autopsy. This study was conducted according to the tenets of the Declaration of Helsinki and approved by the local ethics committees (license number 494-16 from LMU Munich and license number 152/15 from Goethe University). Samples were fixed and embedded in paraffin according to standard procedures.", "Mouse experiments were approved by the Cantonal Veterinary Office of Basel-Stadt (license numbers 2973_32878 and 3070_32419). Mice were housed under specific pathogen-free conditions with standard chow and bedding with a 12-hour day/night cycle according to institutional guidelines. Animals of both sexes were applied without bias. For inhibitor treatments, mice from the same litter and sex were paired and allocated randomly to the groups.", "Figures were compiled with Adobe Photoshop CC 2017 and Adobe Illustrator CC 2017. Statistical computations were performed with Prism 8 (GraphPad Software). For comparison of 2 or multiple groups, distribution of data was analyzed by a Shapiro-Wilk normality test, and group variances were analyzed by an F-test or a Brown-Forsythe test, respectively. According to the results of these tests, a parametric or nonparametric test with or without Welch correction for unequal variances was applied. For Kaplan-Meier survival analysis, significance was assessed by the Gehan-Breslow-Wilcoxon test. The statistical test used to compare the respective data sets is described in the corresponding figure legend. Statistical significance was assumed at P<0.05. Unless otherwise stated, data are presented as dot blot, with each dot representing the mean of the respective technical replicates of 1 biological replicate. Each animal or independent seeding of cells was taken as biological replicate, or as indicated in the legend. The mean value of these dots is shown as bar diagram ± SD.", "RNA sequencing data were deposited in the public database GEO (\naccession number GSE181868). Additional data pertaining to the current article are available from the corresponding author upon request.", "[SUBTITLE] W2A Mutation Abrogates DSG2 Interactions and Impairs Cell–Cell Adhesion In Vitro and In Vivo [SUBSECTION] To study the functional consequences of impaired DSG2 binding, we generated the W2A point mutation (DSG2-W2A) to abrogate the proposed interaction mechanism of the only desmoglein expressed in cardiomyocytes (Figure 1A). First, we investigated in a cell-free setup whether DSG2-W2A is indeed affecting the binding properties of DSG2 interaction by applying single molecule force spectroscopy. Here, molecules are coupled to the tip of an atomic force microscopy probe and the surface of a mica sheet by a polyethylene glycol linker. By measuring the deflection of the probe during repeated approach to and retraction from the surface, binding events can be assessed quantitatively. From these data, properties such as binding frequency, binding force, and bond half-life time can be determined. Constructs were generated for expression of wild-type (WT) and mutant (W2A) DSG2 extracellular domains fused to a human Fc fragment and tested for homotypic (WT:WT, W2A:W2A) and heterotypic (WT:W2A) interaction properties (Figure 1B). These experiments showed a significant reduction of the frequency of W2A:W2A as well as WT:W2A interactions compared with WT:WT (Figure 1C). The frequency of the remaining W2A bindings was comparable to probing Fc alone, which indicates that these are mostly nonspecific interactions. Homotypic WT interactions display a clear binding force peak, whereas the remaining W2A interaction forces were spread widely, again pointing to the nonspecificity of these interactions (Figure 1D). The lifetime of DSG2 interactions under zero force (τ0) was determined by fitting the binding forces detected at different loading rates,11 yielding τ0=1.088s with R=0.993. In contrast, no sufficient fit (R=0.509) was possible for W2A interactions (Figure 1E). Together, these data outline that the tryptophan swap is the major interaction mechanism of DSG2.\nDSG2 (desmoglein-2) adhesion is mediated by tryptophan swap at position 2. A, Predicted interaction model of DSG2 extracellular domains by exchange of tryptophan residue at position 2 into a hydrophobic pocket of the opposing molecule. Cartoon 3D presentation of Protein Data Bank entry 5ERD9; tryptophan-2 is highlighted by ball and stick presentation. B, Schematic of single molecule force spectroscopy experiments. Recombinant extracellular domains (ECs) of wild-type DSG2 (DSG2-WT) or mutant DSG2 (DSG2-W2A) protein were coupled to a mica surface and atomic force microscopy cantilever by mean of a human Fc-tag (hFc) and a polyethylene glycol (PEG)20 linker and probed as indicated. C, Binding frequency of DSG2-W2A/DSG2-WT heterotypic and homotypic interactions at a pulling speed of 2 µm/s are shown. hFc served as control for nonspecific binding. *P<0.05, 1-way analysis of variance, Dunnett post hoc test. Each independent coating procedure with minimum 625 force curves is taken as biological replicate. D, Histogram of binding forces distribution with peak fit at a pulling speed of 2 µm/s corresponding to data in C. E, Determination of the bond half lifetime by the Bell equation11 of mean loading rates and binding forces analyzed from data of pulling speeds at 0.5, 1, 2, 5, and 7.5 µm/s. Average of values from 4 independent coating procedures with minimum 625 force curves each. R=R2, koff =off-rate constant, τ0=bond half lifetime under zero force. F, Dissociation assays to determine cell–cell adhesion were performed in CaCo2 cells (WT or DSG2 KO background) expressing DSG2-WT-mGFP or DSG2-W2A-mGFP constructs. mGFP empty vector served as control. *P<0.05, 1-way analysis of variance, Sidak post hoc test. Corresponding Western blot analysis confirmed effective expression of DSG2 constructs (*) versus the endogenous protein (arrow) in CaCo2 cells. α-tubulin (TUBA) served as loading control. G, Representative images of monolayer fragmentation from experiments in F. H, Dissociation assays in immortalized keratinocytes isolated from neonatal murine skin of the respective genotype. *P<0.05 or as indicated, Welch analysis of variance, Dunnett post hoc test. I, Macroscopic cardiac phenotype of DSG2-W2A mut/mut mice at age 15 weeks. J, Cardiac hypertrophy was analyzed as mean cross-sectional area of cardiomyocytes in hematoxylin & eosin–stained sections. Scale bar, 30 µm. *P<0.05, unpaired Student t test. K, Representative images of intercalated discs acquired by transmission electron microscopy, 3 mice per genotype. Orange asterisks mark intercellular widening, orange circle marks a ruptured junction. Scale bar, 1 µm.\nTo determine the effect of the W2A mutation on a cellular level, DSG2-WT and DSG2-W2A constructs were stably expressed in the epithelial cell line CaCo2 either in a WT or DSG2-deficient background (CaCo2[WT] or CaCo2[DSG2 KO]).12 Cell–cell dissociation assays were performed, in which a confluent cell monolayer is detached and exposed to defined mechanical stress. Expression of DSG2-W2A in CaCo2(WT) cells significantly reduced intercellular adhesion as indicated by an increased number of fragments (Figure 1F and 1G). Whereas expressing DSG2-WT in the CaCo2(DSG2 KO) line significantly rescued the impaired intercellular adhesion in response to DSG2 loss, the expression of DSG2-W2A had no effect on fragment numbers.\nBecause these data demonstrate the DSG2 tryptophan swap to be the central adhesive mechanism, we next generated a CRISPR/Cas9-based knock-in mouse model for DSG2-W2A to investigate the consequences of impaired DSG2 adhesion in vivo (Figure S1). In line with the studies in human cells, dissociation assays in keratinocyte cell lines generated from DSG2-W2A pups revealed reduced cell–cell adhesion in heterozygous (mut/wt) and homozygous (mut/mut) mutants (Figure 1H). Moreover, DSG2-W2A mice presented with a pronounced cardiac phenotype. Adult mut/mut mice demonstrated ventricular deformation, fibrotic and calcified areas, and hypertrophic cardiomyocytes (Figure 1I and 1J). In line with reduced intercellular adhesion of cardiomyocytes, transmission electron microscopy revealed disturbed ICDs with widened intercellular space and occasionally completely ruptured junctions in mut/mut hearts (Figure 1K).\nA cardiac phenotype was also detectable during embryogenesis. Here, mating of heterozygous mice revealed a reduction of mut/mut offspring to 2.8% compared with the expected Mendelian ratio of 25%. Embryo dissections showed loss of the mut/mut animals between E12 and E14 (Figure S2A). The mut/mut embryos at day E12.5 appeared macroscopically pale with accumulation of blood in the cardiac area although the heart was still beating (Figure S2B). Cells suggestive for blood precursors were detectable in the pericardial space (Figure S2C). This finding suggests a rupture of the cardiac wall leading to pericardial bleeding and loss of mut/mut animals.\nTogether, these in vitro and in vivo data outline an essential role of the DSG2 tryptophan swap for cell–cell adhesion and demonstrate severe pathologies associated with impaired DSG2 interaction.\nTo study the functional consequences of impaired DSG2 binding, we generated the W2A point mutation (DSG2-W2A) to abrogate the proposed interaction mechanism of the only desmoglein expressed in cardiomyocytes (Figure 1A). First, we investigated in a cell-free setup whether DSG2-W2A is indeed affecting the binding properties of DSG2 interaction by applying single molecule force spectroscopy. Here, molecules are coupled to the tip of an atomic force microscopy probe and the surface of a mica sheet by a polyethylene glycol linker. By measuring the deflection of the probe during repeated approach to and retraction from the surface, binding events can be assessed quantitatively. From these data, properties such as binding frequency, binding force, and bond half-life time can be determined. Constructs were generated for expression of wild-type (WT) and mutant (W2A) DSG2 extracellular domains fused to a human Fc fragment and tested for homotypic (WT:WT, W2A:W2A) and heterotypic (WT:W2A) interaction properties (Figure 1B). These experiments showed a significant reduction of the frequency of W2A:W2A as well as WT:W2A interactions compared with WT:WT (Figure 1C). The frequency of the remaining W2A bindings was comparable to probing Fc alone, which indicates that these are mostly nonspecific interactions. Homotypic WT interactions display a clear binding force peak, whereas the remaining W2A interaction forces were spread widely, again pointing to the nonspecificity of these interactions (Figure 1D). The lifetime of DSG2 interactions under zero force (τ0) was determined by fitting the binding forces detected at different loading rates,11 yielding τ0=1.088s with R=0.993. In contrast, no sufficient fit (R=0.509) was possible for W2A interactions (Figure 1E). Together, these data outline that the tryptophan swap is the major interaction mechanism of DSG2.\nDSG2 (desmoglein-2) adhesion is mediated by tryptophan swap at position 2. A, Predicted interaction model of DSG2 extracellular domains by exchange of tryptophan residue at position 2 into a hydrophobic pocket of the opposing molecule. Cartoon 3D presentation of Protein Data Bank entry 5ERD9; tryptophan-2 is highlighted by ball and stick presentation. B, Schematic of single molecule force spectroscopy experiments. Recombinant extracellular domains (ECs) of wild-type DSG2 (DSG2-WT) or mutant DSG2 (DSG2-W2A) protein were coupled to a mica surface and atomic force microscopy cantilever by mean of a human Fc-tag (hFc) and a polyethylene glycol (PEG)20 linker and probed as indicated. C, Binding frequency of DSG2-W2A/DSG2-WT heterotypic and homotypic interactions at a pulling speed of 2 µm/s are shown. hFc served as control for nonspecific binding. *P<0.05, 1-way analysis of variance, Dunnett post hoc test. Each independent coating procedure with minimum 625 force curves is taken as biological replicate. D, Histogram of binding forces distribution with peak fit at a pulling speed of 2 µm/s corresponding to data in C. E, Determination of the bond half lifetime by the Bell equation11 of mean loading rates and binding forces analyzed from data of pulling speeds at 0.5, 1, 2, 5, and 7.5 µm/s. Average of values from 4 independent coating procedures with minimum 625 force curves each. R=R2, koff =off-rate constant, τ0=bond half lifetime under zero force. F, Dissociation assays to determine cell–cell adhesion were performed in CaCo2 cells (WT or DSG2 KO background) expressing DSG2-WT-mGFP or DSG2-W2A-mGFP constructs. mGFP empty vector served as control. *P<0.05, 1-way analysis of variance, Sidak post hoc test. Corresponding Western blot analysis confirmed effective expression of DSG2 constructs (*) versus the endogenous protein (arrow) in CaCo2 cells. α-tubulin (TUBA) served as loading control. G, Representative images of monolayer fragmentation from experiments in F. H, Dissociation assays in immortalized keratinocytes isolated from neonatal murine skin of the respective genotype. *P<0.05 or as indicated, Welch analysis of variance, Dunnett post hoc test. I, Macroscopic cardiac phenotype of DSG2-W2A mut/mut mice at age 15 weeks. J, Cardiac hypertrophy was analyzed as mean cross-sectional area of cardiomyocytes in hematoxylin & eosin–stained sections. Scale bar, 30 µm. *P<0.05, unpaired Student t test. K, Representative images of intercalated discs acquired by transmission electron microscopy, 3 mice per genotype. Orange asterisks mark intercellular widening, orange circle marks a ruptured junction. Scale bar, 1 µm.\nTo determine the effect of the W2A mutation on a cellular level, DSG2-WT and DSG2-W2A constructs were stably expressed in the epithelial cell line CaCo2 either in a WT or DSG2-deficient background (CaCo2[WT] or CaCo2[DSG2 KO]).12 Cell–cell dissociation assays were performed, in which a confluent cell monolayer is detached and exposed to defined mechanical stress. Expression of DSG2-W2A in CaCo2(WT) cells significantly reduced intercellular adhesion as indicated by an increased number of fragments (Figure 1F and 1G). Whereas expressing DSG2-WT in the CaCo2(DSG2 KO) line significantly rescued the impaired intercellular adhesion in response to DSG2 loss, the expression of DSG2-W2A had no effect on fragment numbers.\nBecause these data demonstrate the DSG2 tryptophan swap to be the central adhesive mechanism, we next generated a CRISPR/Cas9-based knock-in mouse model for DSG2-W2A to investigate the consequences of impaired DSG2 adhesion in vivo (Figure S1). In line with the studies in human cells, dissociation assays in keratinocyte cell lines generated from DSG2-W2A pups revealed reduced cell–cell adhesion in heterozygous (mut/wt) and homozygous (mut/mut) mutants (Figure 1H). Moreover, DSG2-W2A mice presented with a pronounced cardiac phenotype. Adult mut/mut mice demonstrated ventricular deformation, fibrotic and calcified areas, and hypertrophic cardiomyocytes (Figure 1I and 1J). In line with reduced intercellular adhesion of cardiomyocytes, transmission electron microscopy revealed disturbed ICDs with widened intercellular space and occasionally completely ruptured junctions in mut/mut hearts (Figure 1K).\nA cardiac phenotype was also detectable during embryogenesis. Here, mating of heterozygous mice revealed a reduction of mut/mut offspring to 2.8% compared with the expected Mendelian ratio of 25%. Embryo dissections showed loss of the mut/mut animals between E12 and E14 (Figure S2A). The mut/mut embryos at day E12.5 appeared macroscopically pale with accumulation of blood in the cardiac area although the heart was still beating (Figure S2B). Cells suggestive for blood precursors were detectable in the pericardial space (Figure S2C). This finding suggests a rupture of the cardiac wall leading to pericardial bleeding and loss of mut/mut animals.\nTogether, these in vitro and in vivo data outline an essential role of the DSG2 tryptophan swap for cell–cell adhesion and demonstrate severe pathologies associated with impaired DSG2 interaction.\n[SUBTITLE] DSG2-W2A Mutant Mice Resemble the Phenotype of ACM [SUBSECTION] Given the cardiac affection in mutant mice, the notion that a majority of patients with ACM are reported with mutations in desmosomal genes,2 and that mutations specifically of W2 have already been associated with ACM,10 we examined homozygous and heterozygous mice including both sexes with regard to histologic and clinical ACM measures in an age range of 12 to 80 weeks.\nHistologic analysis showed cardiac fibrosis, a major hallmark of ACM, in the myocardium of the right ventricle (RV) and left ventricle (LV) in mut/mut animals (Figure 2A through 2D), which did not profoundly increase over time. In contrast, heterozygous animals demonstrated a milder phenotype, with a fraction of animals (36%) starting to develop fibrosis in the RV around 6 months, whereas LV was unaffected. To better address differences over time, we pooled animals into groups with a mean age of 4 and 6 months for homozygous and 6 and 12 months for heterozygous mice and performed functional analyses by echocardiography and ECG parallel to histologic evaluation. Moreover, we separated 12-month-old heterozygous animals according to RV fibrosis into a group without and with fibrosis (>10% fibrotic area). Mut/mut animals showed impaired RV systolic function at both time points with reduced tricuspid annular plane systolic excursion as well as fractional shortening (Figure 2E through 2G and Table S1). LV systolic impairment with reduction of the ejection fraction and corresponding parameters worsened over time and became significant after 6 months, although no increase in fibrosis was detectable (Figure 2D and 2H and Table S1). In mut/wt mice, RV fractional shortening was significantly reduced in 12-month-old animals with fibrosis. In ECG recordings of mut/mut mice for 30 minutes under anaesthesia, we noted deformation of the QRS complex with elongated QRS interval and reduced amplitude of the S peak with effects more pronounced in the older group; heterozygous animals with RV fibrosis showed similar effects or trends (Figure 2I through 2K and Table S1). Because the T wave was not clearly detectable in mutants, the J peak amplitude as indicator for early repolarization was determined. This was reduced in the mut/mut groups with same trend for fibrotic mut/wt hearts (Figure 2L). Parallel to these depolarization and repolarization abnormalities, we also noted the occurrence of ventricular arrhythmia, ranging from single premature ventricular contractions (PVCs) to multiple, multifocal PVCs and nonsustained ventricular tachycardia in 1 mouse (Figure 2M through 2O). The fraction of homozygous and heterozygous animals with PVCs and the frequency of these events correlated with age and fibrosis. Moreover, a substantial fraction (16%) of mut/mut animals was found dead during the observation period, which strongly suggests the occurrence of malignant arrhythmia with sudden death (Figure 2P). Investigating sex-related differences, no effect was observed on the extent of fibrosis or echocardiographic parameters. However, in mut/wt mice, PVCs were found in males only and male mut/mut mice were more prone to premature sudden death compared with females (Figure S3).\nDSG2 (desmoglein-2)–W2A mutant mice develop characteristics of arrhythmogenic cardiomyopathy. A, Cardiac fibrosis detected by picrosirius red collagen staining with representative images of sections from 6-month-old DSG2-W2A mut/mut and 12-month-old wt/wt and mut/wt animals. Scale bar = 1 mm. B through D, Corresponding analysis of the area of collagen in the right ventricle (RV) and left ventricle (LV). Continuous lines indicate the simple linear regression between age and area of collagen. Hearts with >10% of collagen in the RV (gray dotted line) were defined as “with fibrosis.” Each dot represents 1 animal. *P<0.05, mixed effects analysis with LV and RV matched per animal, Sidak post hoc test. Lines indicate median and quartile values. E, Representative echocardiography images for measurements of the tricuspid annular plane systolic excursion (TAPSE) and LV function in 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Left side: 2D images from apical 4-chamber view for TAPSE and parasternal short axis view for LV. M-mode tracings on the right were performed along the line indicated on the left. Scale bars, white, 2 mm; black, 100 ms. Corresponding analysis of (F) TAPSE, *P<0.05, Kruskal-Wallis test with Dunn post hoc test; (G) fractional shortening of the RV, *P<0.05, Kruskal-Wallis test with Dunn post hoc test; and (H) ejection fraction of the LV, *P<0.05, 1-way analysis of variance, Sidak post hoc test. I, ECG recoded in lead II with representative curves from 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Definition of respective peaks is indicated in the wt/wt curve. Scale bars, vertical, 0.5 mV; horizontal, 50 ms. Corresponding analysis of (J) QRS interval, *P<0.05 or as indicated, Kruskal-Wallis test with Dunn post hoc test; (K) amplitude of the S peak, *P<0.05 or as indicated, 1-way analysis of variance, Sidak post hoc test; and (L) amplitude of the J peak (early repolarization), *P<0.05 or as indicated, Kruskal-Wallis test with Dunn post hoc test. M, Ventricular arrythmia detected by ECG during 30 minutes of baseline measurements with example curves from 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Asterisks mark premature ventricular contractions (PVCs); *nsVT indicates a nonsustained ventricular tachycardia detected in 1 mut/mut animal. Scale bars, vertical, 0.5 mV; horizontal, 1 s. Corresponding analysis of (N) percentage of mice presenting with PVCs. Values in bars indicate corresponding absolute number of mice with PVC (colored bars) compared with total number of mice evaluated (empty bar) and (O) PVC burden depicted as number of PVCs per minute, *P<0.05, Kruskal-Wallis test with Dunn post hoc test. The black arrow indicates the animal presenting with nonsustained ventricular tachycardia. P, Kaplan-Meier survival plot of DSG2-W2A mice from an analysis period of 3 years. Vertical lines indicate dropouts because of unrelated elimination (end of experiment, breeding, injuries). Values indicate corresponding absolute number of mice with sudden death compared with total number of mice evaluated. *P<0.05, Gehan-Breslow-Wilcoxon test. Box with color indications of respective groups in the middle applies to the entire figure.\nIn summary, mutant animals show histopathologic features with echocardiography and ECG abnormalities similar to patients with ACM. Within the limits of a murine model, both genotypes fulfil the Padua criteria13 to establish the diagnosis of ACM (Table S2). Here, DSG2-W2A mut/mut animals resemble the phenotype of biventricular ACM, whereas in mut/wt mice, the phenotype is milder with variable penetrance and age-dependent occurrence only in the RV. According to the Padua criteria,13 the heterozygous genotype recalls the characteristics of a right-dominant ventricular ACM, as long as fibrosis was present. Together, these data demonstrate that loss of desmosomal adhesion is sufficient to induce an ACM phenotype and that the DSG2-W2A model may resemble 2 different variants of the disease.\nGiven the cardiac affection in mutant mice, the notion that a majority of patients with ACM are reported with mutations in desmosomal genes,2 and that mutations specifically of W2 have already been associated with ACM,10 we examined homozygous and heterozygous mice including both sexes with regard to histologic and clinical ACM measures in an age range of 12 to 80 weeks.\nHistologic analysis showed cardiac fibrosis, a major hallmark of ACM, in the myocardium of the right ventricle (RV) and left ventricle (LV) in mut/mut animals (Figure 2A through 2D), which did not profoundly increase over time. In contrast, heterozygous animals demonstrated a milder phenotype, with a fraction of animals (36%) starting to develop fibrosis in the RV around 6 months, whereas LV was unaffected. To better address differences over time, we pooled animals into groups with a mean age of 4 and 6 months for homozygous and 6 and 12 months for heterozygous mice and performed functional analyses by echocardiography and ECG parallel to histologic evaluation. Moreover, we separated 12-month-old heterozygous animals according to RV fibrosis into a group without and with fibrosis (>10% fibrotic area). Mut/mut animals showed impaired RV systolic function at both time points with reduced tricuspid annular plane systolic excursion as well as fractional shortening (Figure 2E through 2G and Table S1). LV systolic impairment with reduction of the ejection fraction and corresponding parameters worsened over time and became significant after 6 months, although no increase in fibrosis was detectable (Figure 2D and 2H and Table S1). In mut/wt mice, RV fractional shortening was significantly reduced in 12-month-old animals with fibrosis. In ECG recordings of mut/mut mice for 30 minutes under anaesthesia, we noted deformation of the QRS complex with elongated QRS interval and reduced amplitude of the S peak with effects more pronounced in the older group; heterozygous animals with RV fibrosis showed similar effects or trends (Figure 2I through 2K and Table S1). Because the T wave was not clearly detectable in mutants, the J peak amplitude as indicator for early repolarization was determined. This was reduced in the mut/mut groups with same trend for fibrotic mut/wt hearts (Figure 2L). Parallel to these depolarization and repolarization abnormalities, we also noted the occurrence of ventricular arrhythmia, ranging from single premature ventricular contractions (PVCs) to multiple, multifocal PVCs and nonsustained ventricular tachycardia in 1 mouse (Figure 2M through 2O). The fraction of homozygous and heterozygous animals with PVCs and the frequency of these events correlated with age and fibrosis. Moreover, a substantial fraction (16%) of mut/mut animals was found dead during the observation period, which strongly suggests the occurrence of malignant arrhythmia with sudden death (Figure 2P). Investigating sex-related differences, no effect was observed on the extent of fibrosis or echocardiographic parameters. However, in mut/wt mice, PVCs were found in males only and male mut/mut mice were more prone to premature sudden death compared with females (Figure S3).\nDSG2 (desmoglein-2)–W2A mutant mice develop characteristics of arrhythmogenic cardiomyopathy. A, Cardiac fibrosis detected by picrosirius red collagen staining with representative images of sections from 6-month-old DSG2-W2A mut/mut and 12-month-old wt/wt and mut/wt animals. Scale bar = 1 mm. B through D, Corresponding analysis of the area of collagen in the right ventricle (RV) and left ventricle (LV). Continuous lines indicate the simple linear regression between age and area of collagen. Hearts with >10% of collagen in the RV (gray dotted line) were defined as “with fibrosis.” Each dot represents 1 animal. *P<0.05, mixed effects analysis with LV and RV matched per animal, Sidak post hoc test. Lines indicate median and quartile values. E, Representative echocardiography images for measurements of the tricuspid annular plane systolic excursion (TAPSE) and LV function in 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Left side: 2D images from apical 4-chamber view for TAPSE and parasternal short axis view for LV. M-mode tracings on the right were performed along the line indicated on the left. Scale bars, white, 2 mm; black, 100 ms. Corresponding analysis of (F) TAPSE, *P<0.05, Kruskal-Wallis test with Dunn post hoc test; (G) fractional shortening of the RV, *P<0.05, Kruskal-Wallis test with Dunn post hoc test; and (H) ejection fraction of the LV, *P<0.05, 1-way analysis of variance, Sidak post hoc test. I, ECG recoded in lead II with representative curves from 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Definition of respective peaks is indicated in the wt/wt curve. Scale bars, vertical, 0.5 mV; horizontal, 50 ms. Corresponding analysis of (J) QRS interval, *P<0.05 or as indicated, Kruskal-Wallis test with Dunn post hoc test; (K) amplitude of the S peak, *P<0.05 or as indicated, 1-way analysis of variance, Sidak post hoc test; and (L) amplitude of the J peak (early repolarization), *P<0.05 or as indicated, Kruskal-Wallis test with Dunn post hoc test. M, Ventricular arrythmia detected by ECG during 30 minutes of baseline measurements with example curves from 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Asterisks mark premature ventricular contractions (PVCs); *nsVT indicates a nonsustained ventricular tachycardia detected in 1 mut/mut animal. Scale bars, vertical, 0.5 mV; horizontal, 1 s. Corresponding analysis of (N) percentage of mice presenting with PVCs. Values in bars indicate corresponding absolute number of mice with PVC (colored bars) compared with total number of mice evaluated (empty bar) and (O) PVC burden depicted as number of PVCs per minute, *P<0.05, Kruskal-Wallis test with Dunn post hoc test. The black arrow indicates the animal presenting with nonsustained ventricular tachycardia. P, Kaplan-Meier survival plot of DSG2-W2A mice from an analysis period of 3 years. Vertical lines indicate dropouts because of unrelated elimination (end of experiment, breeding, injuries). Values indicate corresponding absolute number of mice with sudden death compared with total number of mice evaluated. *P<0.05, Gehan-Breslow-Wilcoxon test. Box with color indications of respective groups in the middle applies to the entire figure.\nIn summary, mutant animals show histopathologic features with echocardiography and ECG abnormalities similar to patients with ACM. Within the limits of a murine model, both genotypes fulfil the Padua criteria13 to establish the diagnosis of ACM (Table S2). Here, DSG2-W2A mut/mut animals resemble the phenotype of biventricular ACM, whereas in mut/wt mice, the phenotype is milder with variable penetrance and age-dependent occurrence only in the RV. According to the Padua criteria,13 the heterozygous genotype recalls the characteristics of a right-dominant ventricular ACM, as long as fibrosis was present. Together, these data demonstrate that loss of desmosomal adhesion is sufficient to induce an ACM phenotype and that the DSG2-W2A model may resemble 2 different variants of the disease.\n[SUBTITLE] Integrin-β6 Expression Is Altered in ACM Patient and DSG2-W2A Mouse Samples [SUBSECTION] Next, we applied this mouse model to investigate mechanisms leading to ACM. RNA sequencing was performed from ventricles of mut/mut and wt/wt hearts both before onset of fibrosis (age 5 days) and when the biventricular fibrosis and ACM phenotype was established (after 9 weeks). First, we compared sequencing data from 9-week-old mice with the top differentially expressed genes derived from published transcriptomic data sets of ACM patient hearts (GEO database: GSE107157/GSE10748014 and GSE2981915). Murine samples clustered according to their genotype and mut/mut hearts resembled the gene expression pattern of patients (Figure S4), further supporting the observation that mutant mice faithfully reproduce major aspects of the disease.\nTo identify common genes deregulated during ACM pathogenesis, we compared the differentially expressed genes from patient data with the results from mutant mice. Here, we included data from 5-day-old mice to identify changes already present before onset of secondary effects attributable to fibrosis. Integrin-β6 (Itgb6) was the only transcript consistently deregulated in all data sets (Figure 3A). Its common downregulation in patients with ACM and mutant mice on RNA levels was further confirmed for heterozygous DSG2-W2A mutants compared with WT (Figure 3B). However, protein levels were unaltered in mutant heart samples (Figure 3C). Immunostaining revealed that, in mutant hearts, ITGB6, which mainly localized to a compartment suggestive for the transverse tubules and costameres in wt/wt animals, was enriched at the ICD (Figure 3D and E). Together, these data demonstrate altered ITGB6 mRNA expression as a common feature in ACM patient and DSG2-W2A hearts and an enrichment of the protein at the ICD in mutant murine samples.\nIntegrin-β6 is deregulated in DSG2 (desmoglein-2)–W2A mutants. A, Venn diagram of significantly altered genes from indicated arrhythmogenic cardiomyopathy (ACM) patient data sets (ACM versus healthy control) and DSG2-W2A mice at the age of 5 days and 9 weeks (mut/mut versus wt/wt) highlighting integrin-β6 (Itgb6) as only overlapping gene with same direction of expression in all data sets. Numbers indicate the amount of overlapping genes for the respective overlays. B, RNA expression of Itgb6 analyzed by means of reverse transcription quantitative polymerase chain reaction in adult DSG2-W2A mouse hearts. *P<0.05, unpaired Student t test versus wt/wt. Gapdh served as reference gene. C, Representative Western blot and respective analysis of band intensity of integrin-β6 (ITGB6) in DSG2-W2A hearts. GAPDH served as loading control. 1-way analysis of variance, Dunnett post hoc test. D and E, Immunostaining of ITGB6 (red in overlay) in DSG2-W2A hearts with corresponding analysis of staining intensity in cardiomyocytes in total and ratio of staining intensity at the intercalated disc (ICD) area (orange arrowheads) versus cardiomyocytes’ cytosolic area. DSP (desmoplakin; cyan) marks ICDs, DAPI (blue) nuclei and F-actin (green) the sarcomere system. Lower row shows an overview image of a fibrotic area in mut/mut hearts. Scale bars, 25 µm. *P<0.05; left graph: Kruskal-Wallis test with Dunn post hoc test; right graph: 1-way analysis of variance, Dunnett post hoc test. Box with color indications of respective groups applies to the entire figure.\nNext, we applied this mouse model to investigate mechanisms leading to ACM. RNA sequencing was performed from ventricles of mut/mut and wt/wt hearts both before onset of fibrosis (age 5 days) and when the biventricular fibrosis and ACM phenotype was established (after 9 weeks). First, we compared sequencing data from 9-week-old mice with the top differentially expressed genes derived from published transcriptomic data sets of ACM patient hearts (GEO database: GSE107157/GSE10748014 and GSE2981915). Murine samples clustered according to their genotype and mut/mut hearts resembled the gene expression pattern of patients (Figure S4), further supporting the observation that mutant mice faithfully reproduce major aspects of the disease.\nTo identify common genes deregulated during ACM pathogenesis, we compared the differentially expressed genes from patient data with the results from mutant mice. Here, we included data from 5-day-old mice to identify changes already present before onset of secondary effects attributable to fibrosis. Integrin-β6 (Itgb6) was the only transcript consistently deregulated in all data sets (Figure 3A). Its common downregulation in patients with ACM and mutant mice on RNA levels was further confirmed for heterozygous DSG2-W2A mutants compared with WT (Figure 3B). However, protein levels were unaltered in mutant heart samples (Figure 3C). Immunostaining revealed that, in mutant hearts, ITGB6, which mainly localized to a compartment suggestive for the transverse tubules and costameres in wt/wt animals, was enriched at the ICD (Figure 3D and E). Together, these data demonstrate altered ITGB6 mRNA expression as a common feature in ACM patient and DSG2-W2A hearts and an enrichment of the protein at the ICD in mutant murine samples.\nIntegrin-β6 is deregulated in DSG2 (desmoglein-2)–W2A mutants. A, Venn diagram of significantly altered genes from indicated arrhythmogenic cardiomyopathy (ACM) patient data sets (ACM versus healthy control) and DSG2-W2A mice at the age of 5 days and 9 weeks (mut/mut versus wt/wt) highlighting integrin-β6 (Itgb6) as only overlapping gene with same direction of expression in all data sets. Numbers indicate the amount of overlapping genes for the respective overlays. B, RNA expression of Itgb6 analyzed by means of reverse transcription quantitative polymerase chain reaction in adult DSG2-W2A mouse hearts. *P<0.05, unpaired Student t test versus wt/wt. Gapdh served as reference gene. C, Representative Western blot and respective analysis of band intensity of integrin-β6 (ITGB6) in DSG2-W2A hearts. GAPDH served as loading control. 1-way analysis of variance, Dunnett post hoc test. D and E, Immunostaining of ITGB6 (red in overlay) in DSG2-W2A hearts with corresponding analysis of staining intensity in cardiomyocytes in total and ratio of staining intensity at the intercalated disc (ICD) area (orange arrowheads) versus cardiomyocytes’ cytosolic area. DSP (desmoplakin; cyan) marks ICDs, DAPI (blue) nuclei and F-actin (green) the sarcomere system. Lower row shows an overview image of a fibrotic area in mut/mut hearts. Scale bars, 25 µm. *P<0.05; left graph: Kruskal-Wallis test with Dunn post hoc test; right graph: 1-way analysis of variance, Dunnett post hoc test. Box with color indications of respective groups applies to the entire figure.\n[SUBTITLE] DSG2-W2A Mutant Hearts Present With Structurally Impaired ICDs [SUBSECTION] This result together with the transmission electron microscopy data (Figure 1K) suggest structural changes of the ICD in mutant hearts, which we addressed further. Using DSP as marker, we reconstructed the outlines of ICDs in wt/wt and mut/mut hearts from 3D stacks captured with structured illumination microscopy. This analysis yielded an increase in ICD volume, in particular attributable to an enhanced width between 2 adjacent cells (Figure 4A). The majority of desmosomal molecules as well as N-cadherin were regularly distributed at mutant ICDs (Figure S5A and S5B). This was corroborated by RNA sequencing data from 5-day-old and 9-week-old animals, which showed no consistent changes of desmosomal, adherens, gap, or tight junction molecules (Figure S5C through S5E). The only adhesion molecule reduced globally was DSG2 (Figure S5A and S5B). Although DSG2 was still present at the ICD, detailed analysis by structured illumination microscopy revealed reduced number and volume of DSG2 signals (Figure 4B). Moreover, fluorescence recovery after photobleaching experiments in neonatal murine cardiomyocytes transduced with DSG2-WT-mGFP or DSG2-W2A-mGFP showed a higher mobility of the mutant protein at cardiomyocyte junctions (Figure 4C), indicating reduced membrane stability of DSG2-W2A. To further investigate the possibility that the stability and integrity of ICD molecules is compromised, we performed Triton-X-100 separation assays for proteins with unaltered ICD localization in mutant hearts. Cytosolic and unanchored membrane proteins are found in the Triton-X-100 soluble fraction, whereas cytoskeleton-bound molecules are mainly detectable in the nonsoluble fraction. Both desmosomal molecules (PG, PKP2) as well as N-cadherin showed increased levels in the soluble pool, indicating reduced cytoskeletal anchorage and impaired ICD integrity in mutants (Figure 4D). Moreover, the gap junction molecule connexin-43, which is required for electrical coupling of cardiomyocytes, was delocalized to the lateral membrane (Figure 4E), which was described as a feature of disrupted ICDs.8\nDisrupted ICD structure in DSG2 (desmoglein-2)–W2A mutant mice. A, Representative z-stack reconstruction of segmented intercalated discs (ICDs) in top and side view acquired with structured illumination microscopy. Overlay of analyzed ICD volume is shown in gray. ICDs are marked by DSP (desmoplakin; magenta). Scheme on top presents segmented area and pictograms on the right display the respective angle of view. Corresponding analysis of ICD volume and width of ICD between adjacent cardiomyocytes below. Scale bar, 5 µm. *P<0.05, Mann-Whitney test. Each dot represents the value of 1 ICD from 4 mice per genotype. B, Representative images of DSG2 (magenta) and filamentary actin (f-actin; white) z-stacks acquired by structured illumination microscopy and presented as maximum intensity projection. Lower row shows color-coded height projection of DSG2 signals in z-stack after signal thresholding as performed for analysis. Related analysis of DSG2 signal volume and number per ICD length is shown below. Pictograms on the right display the respective angle of view. Scale bar, 5 µm. *P<0.05, unpaired Student t test, with Welch correction. Each dot represents the value of 1 ICD from in total 3 mice per genotype. C, Fluorescence recovery after photobleaching analysis of DSG2–wild type (WT) and DSG2-W2A-mGFP fusion proteins at the cell–cell junction of neonatal cardiomyocytes with representative intensity kymographs of bleached areas on the left. Time point 0 = bleach as indicated by the black arrow. Analysis of the mobile fraction of the indicated mGFP-fusion proteins is shown on the right. *P<0.05, unpaired Student t test, with Welch correction. Each dot represents the mean value of 1 heart from in total 3 isolations. D, Representative triton-X-100 assay immunoblot with separation of a soluble (sol), noncytoskeletal bound protein fraction from a nonsoluble (non-sol), cytoskeletal-anchored fraction and corresponding analysis shown below. PG (plakoglobin), PKP2 (plakophilin-2), and N-cadherin (NCAD) were analyzed. Intensity of proteins was normalized to the total amount of protein detected by ponceau staining. GAPDH and DSP served as separation control. *P<0.05 or as indicated, unpaired Student t test (PG, NCAD, PKP2) or Mann-Whitney test (DSP). Each dot represents the result from 1 mouse. E, Immunostaining of connexin-43 (CX43; red in overlay) in DSG2-W2A hearts. NCAD (yellow) marks ICDs, DAPI (blue) nuclei, and F-actin (green) the sarcomere system. Orange arrowheads mark ICD, pink arrowheads highlight lateralization of CX43 staining. Scale bars, 25 µm. Images representative for 5 mice per genotype. Box with color indications of respective groups applies to the entire figure.\nThese data together with the transmission electron microscopy results demonstrate structurally severely altered ICDs in response to the DSG2-W2A mutation and suggest a molecular basis leading to the functional alterations detectable by echocardiography and ECG.\nThis result together with the transmission electron microscopy data (Figure 1K) suggest structural changes of the ICD in mutant hearts, which we addressed further. Using DSP as marker, we reconstructed the outlines of ICDs in wt/wt and mut/mut hearts from 3D stacks captured with structured illumination microscopy. This analysis yielded an increase in ICD volume, in particular attributable to an enhanced width between 2 adjacent cells (Figure 4A). The majority of desmosomal molecules as well as N-cadherin were regularly distributed at mutant ICDs (Figure S5A and S5B). This was corroborated by RNA sequencing data from 5-day-old and 9-week-old animals, which showed no consistent changes of desmosomal, adherens, gap, or tight junction molecules (Figure S5C through S5E). The only adhesion molecule reduced globally was DSG2 (Figure S5A and S5B). Although DSG2 was still present at the ICD, detailed analysis by structured illumination microscopy revealed reduced number and volume of DSG2 signals (Figure 4B). Moreover, fluorescence recovery after photobleaching experiments in neonatal murine cardiomyocytes transduced with DSG2-WT-mGFP or DSG2-W2A-mGFP showed a higher mobility of the mutant protein at cardiomyocyte junctions (Figure 4C), indicating reduced membrane stability of DSG2-W2A. To further investigate the possibility that the stability and integrity of ICD molecules is compromised, we performed Triton-X-100 separation assays for proteins with unaltered ICD localization in mutant hearts. Cytosolic and unanchored membrane proteins are found in the Triton-X-100 soluble fraction, whereas cytoskeleton-bound molecules are mainly detectable in the nonsoluble fraction. Both desmosomal molecules (PG, PKP2) as well as N-cadherin showed increased levels in the soluble pool, indicating reduced cytoskeletal anchorage and impaired ICD integrity in mutants (Figure 4D). Moreover, the gap junction molecule connexin-43, which is required for electrical coupling of cardiomyocytes, was delocalized to the lateral membrane (Figure 4E), which was described as a feature of disrupted ICDs.8\nDisrupted ICD structure in DSG2 (desmoglein-2)–W2A mutant mice. A, Representative z-stack reconstruction of segmented intercalated discs (ICDs) in top and side view acquired with structured illumination microscopy. Overlay of analyzed ICD volume is shown in gray. ICDs are marked by DSP (desmoplakin; magenta). Scheme on top presents segmented area and pictograms on the right display the respective angle of view. Corresponding analysis of ICD volume and width of ICD between adjacent cardiomyocytes below. Scale bar, 5 µm. *P<0.05, Mann-Whitney test. Each dot represents the value of 1 ICD from 4 mice per genotype. B, Representative images of DSG2 (magenta) and filamentary actin (f-actin; white) z-stacks acquired by structured illumination microscopy and presented as maximum intensity projection. Lower row shows color-coded height projection of DSG2 signals in z-stack after signal thresholding as performed for analysis. Related analysis of DSG2 signal volume and number per ICD length is shown below. Pictograms on the right display the respective angle of view. Scale bar, 5 µm. *P<0.05, unpaired Student t test, with Welch correction. Each dot represents the value of 1 ICD from in total 3 mice per genotype. C, Fluorescence recovery after photobleaching analysis of DSG2–wild type (WT) and DSG2-W2A-mGFP fusion proteins at the cell–cell junction of neonatal cardiomyocytes with representative intensity kymographs of bleached areas on the left. Time point 0 = bleach as indicated by the black arrow. Analysis of the mobile fraction of the indicated mGFP-fusion proteins is shown on the right. *P<0.05, unpaired Student t test, with Welch correction. Each dot represents the mean value of 1 heart from in total 3 isolations. D, Representative triton-X-100 assay immunoblot with separation of a soluble (sol), noncytoskeletal bound protein fraction from a nonsoluble (non-sol), cytoskeletal-anchored fraction and corresponding analysis shown below. PG (plakoglobin), PKP2 (plakophilin-2), and N-cadherin (NCAD) were analyzed. Intensity of proteins was normalized to the total amount of protein detected by ponceau staining. GAPDH and DSP served as separation control. *P<0.05 or as indicated, unpaired Student t test (PG, NCAD, PKP2) or Mann-Whitney test (DSP). Each dot represents the result from 1 mouse. E, Immunostaining of connexin-43 (CX43; red in overlay) in DSG2-W2A hearts. NCAD (yellow) marks ICDs, DAPI (blue) nuclei, and F-actin (green) the sarcomere system. Orange arrowheads mark ICD, pink arrowheads highlight lateralization of CX43 staining. Scale bars, 25 µm. Images representative for 5 mice per genotype. Box with color indications of respective groups applies to the entire figure.\nThese data together with the transmission electron microscopy results demonstrate structurally severely altered ICDs in response to the DSG2-W2A mutation and suggest a molecular basis leading to the functional alterations detectable by echocardiography and ECG.\n[SUBTITLE] Integrin-αV/β6 Is Activated in DSG2-W2A Mutant Hearts [SUBSECTION] In contrast to DSG2, we detected ITGB6 to be increased at the ICD (Figure 3D and 3E), which was confirmed by analysis of structured illumination microscopy images (Figure 5A). ITGB6 needs to heterodimerize with integrin-αV (ITGAV) in order to be activated and bind to the extracellular matrix.16–18 Thus, we analyzed the expression of the counterpart ITGAV, which was significantly upregulated on protein level and increased at the ICD and costameres of mutant hearts (Figure 5B and 5C). In contrast, the localization and intensity of integrin-β1, as classical representative of the integrin group, was not altered (Figure S6). Staining with an antibody specifically recognizing the heterodimer of ITGAV/B6 revealed increased amount of dimer formation at the ICD and costameres in mutant mice (Figure 5D). Moreover, increased ITGAV/B6 staining intensity was also detectable in an ACM patient sample with DSP mutation (Figure 5E). To stabilize active integrins, recruitment of the cytoskeletal adapters talin and vinculin is required.19 Accordingly, the expression of talin-2 was increased in DSG2-W2A mut/mut myocardium, with vinculin being enriched at the ICD (Figure 5F and 5G). This effect is similar to what was shown before in response to increased intracellular traction forces on junctions.20 Thus, these data suggest that in response to a dysfunctional, nonadhesive ICD with impaired cytoskeletal anchorage, ITGAV/B6 dimers are recruited and activated at the compromised ICD as well as at the costamere region.\nIncreased activation of ITGB6/AV at ICDs of DSG2 (desmoglein-2)–W2A mutant mice. A, Representative intercalated disc (ICD) reconstruction from z-stacks of integrin-β6 (ITGB6; green) and DSP (magenta) immunostaining acquired by structured illumination microscopy and presented as maximum intensity projection. Related analysis of ITGB6 signal volume and number per ICD length is shown below. Scale bar, 5 µm. *P<0.05 or as indicated, unpaired Student t test with Welch correction (left graph) and Mann-Whitney test (right graph). Each dot represents the value of 1 ICD from in total 4 mice per genotype. B, Representative Western blot and analysis of band intensity of integrin-αV (ITGAV) in DSG2-W2A hearts. GAPDH served as loading control. *P<0.05, unpaired Student t test with Welch correction. C, Immunostaining of ITGAV in DSG2-W2A hearts on the right with respective analysis on the left. DAPI (blue) marks nuclei. Lower row shows an overview image of a fibrotic area in mut/mut hearts. Dotted orange line marks the edge of fibrotic area. Scale bars: upper rows, 20 µm; lower row, 50 µm. *P<0.05, unpaired Student t test. D, Immunostaining of ITGAV/B6 heterodimer in DSG2-W2A mutant hearts with respective analysis of staining intensity on the right. Cyan rectangle marks zoomed area on the right. Scale bars: overview, 50 µm; insert, 10 µm. *P<0.05, unpaired Student t test. E, Representative immunostaining images of ITGAV/B6 heterodimer staining (red) in a patient with arrhythmogenic cardiomyopathy (ACM; DSP-E952X, heterozygous) and healthy control sample. F-actin (green) stains the sarcomere system. For the patient with ACM, 4 different tissue samples were analyzed and compared with 2 tissue samples from 2 healthy controls. Scale bar, 20 µm. F, Immunostaining of VCL (vinculin; magenta) and TLN2 (talin-2; red) in DSG2-W2A hearts on the right with respective analysis of the mean staining intensity in cardiomyocytes and ratio of the staining intensity at the ICD versus cytosolic area on the left. DAPI (blue) marks nuclei. F-actin (green) stains the sarcomere system. Scale bar, 25 µm; *P<0.05, unpaired Student t test. G, Representative Western blot and analysis of band intensity of VCL and TLN2 in DSG2-W2A hearts. GAPDH served as loading control. *P<0.05, unpaired Student t test for TLN2, Mann-Whitney test for VCL. Box with color indications of respective groups applies to the entire figure.\nIn contrast to DSG2, we detected ITGB6 to be increased at the ICD (Figure 3D and 3E), which was confirmed by analysis of structured illumination microscopy images (Figure 5A). ITGB6 needs to heterodimerize with integrin-αV (ITGAV) in order to be activated and bind to the extracellular matrix.16–18 Thus, we analyzed the expression of the counterpart ITGAV, which was significantly upregulated on protein level and increased at the ICD and costameres of mutant hearts (Figure 5B and 5C). In contrast, the localization and intensity of integrin-β1, as classical representative of the integrin group, was not altered (Figure S6). Staining with an antibody specifically recognizing the heterodimer of ITGAV/B6 revealed increased amount of dimer formation at the ICD and costameres in mutant mice (Figure 5D). Moreover, increased ITGAV/B6 staining intensity was also detectable in an ACM patient sample with DSP mutation (Figure 5E). To stabilize active integrins, recruitment of the cytoskeletal adapters talin and vinculin is required.19 Accordingly, the expression of talin-2 was increased in DSG2-W2A mut/mut myocardium, with vinculin being enriched at the ICD (Figure 5F and 5G). This effect is similar to what was shown before in response to increased intracellular traction forces on junctions.20 Thus, these data suggest that in response to a dysfunctional, nonadhesive ICD with impaired cytoskeletal anchorage, ITGAV/B6 dimers are recruited and activated at the compromised ICD as well as at the costamere region.\nIncreased activation of ITGB6/AV at ICDs of DSG2 (desmoglein-2)–W2A mutant mice. A, Representative intercalated disc (ICD) reconstruction from z-stacks of integrin-β6 (ITGB6; green) and DSP (magenta) immunostaining acquired by structured illumination microscopy and presented as maximum intensity projection. Related analysis of ITGB6 signal volume and number per ICD length is shown below. Scale bar, 5 µm. *P<0.05 or as indicated, unpaired Student t test with Welch correction (left graph) and Mann-Whitney test (right graph). Each dot represents the value of 1 ICD from in total 4 mice per genotype. B, Representative Western blot and analysis of band intensity of integrin-αV (ITGAV) in DSG2-W2A hearts. GAPDH served as loading control. *P<0.05, unpaired Student t test with Welch correction. C, Immunostaining of ITGAV in DSG2-W2A hearts on the right with respective analysis on the left. DAPI (blue) marks nuclei. Lower row shows an overview image of a fibrotic area in mut/mut hearts. Dotted orange line marks the edge of fibrotic area. Scale bars: upper rows, 20 µm; lower row, 50 µm. *P<0.05, unpaired Student t test. D, Immunostaining of ITGAV/B6 heterodimer in DSG2-W2A mutant hearts with respective analysis of staining intensity on the right. Cyan rectangle marks zoomed area on the right. Scale bars: overview, 50 µm; insert, 10 µm. *P<0.05, unpaired Student t test. E, Representative immunostaining images of ITGAV/B6 heterodimer staining (red) in a patient with arrhythmogenic cardiomyopathy (ACM; DSP-E952X, heterozygous) and healthy control sample. F-actin (green) stains the sarcomere system. For the patient with ACM, 4 different tissue samples were analyzed and compared with 2 tissue samples from 2 healthy controls. Scale bar, 20 µm. F, Immunostaining of VCL (vinculin; magenta) and TLN2 (talin-2; red) in DSG2-W2A hearts on the right with respective analysis of the mean staining intensity in cardiomyocytes and ratio of the staining intensity at the ICD versus cytosolic area on the left. DAPI (blue) marks nuclei. F-actin (green) stains the sarcomere system. Scale bar, 25 µm; *P<0.05, unpaired Student t test. G, Representative Western blot and analysis of band intensity of VCL and TLN2 in DSG2-W2A hearts. GAPDH served as loading control. *P<0.05, unpaired Student t test for TLN2, Mann-Whitney test for VCL. Box with color indications of respective groups applies to the entire figure.\n[SUBTITLE] TGF-β Signaling Is Upregulated in DSG2-W2A Mutant Hearts [SUBSECTION] ITGAV/B6 dimers have the ability to activate the profibrotic cytokine transforming growth factor–β (TGF-β) by binding and removal of the latency-associated peptide.17 Gene set enrichment analyses revealed an upregulation of genes associated with TGF-β signaling in both 9-week-old mutants as well as patients with ACM (Figure 6A). Moreover, direct targets of receptor-regulated SMADs involved in TGF-β signaling21 were upregulated in DSG2-W2A mutant hearts (Figure 6B). Accordingly, increased amounts of nuclear SMAD2/3 phosphorylated at the activation sites S465/S467 or S423/S425, respectively, were detected in these hearts (Figure 6C). Increased levels of pSMAD2/3 were found not only in fibroblasts but also in cardiomyocytes, mainly adjacent to fibrotic areas. These data indicate upregulation of the profibrotic TGF-β pathway and its downstream targets, which include extracellular matrix proteins such as collagens, laminin, or fibronectin.\nElevated transforming growth factor–β signaling in DSG2 (desmoglein-2)–W2A hearts as result of ITGAV/B6 activity. Barcode plots of gene set enrichment analysis of (A) the KEGG_TGF_BETA_SIGNALING_PATHWAY data set (systematic name: M2642)41 and (B) genes directly regulated by receptor-regulated SMADs (R-SMADs; includes SMAD1/2/3/5/9) as published in the TRRUST database21 in 9-week-old DSG2-W2A (mut/mut vs wt/wt) or arrhythmogenic cardiomyopathy (ACM) patient data set 1 (ACM versus healthy control; GEO: GSE107157/GSE107480). Indicated P values are calculated by function cameraPR of the R package limma. C, Immunostaining of phosphorylated SMAD2/3 (magenta, S465/S467 or S423/S425, respectively) in sections of DSG2-W2A hearts and related analysis of nuclear staining intensity. Nuclei are stained with DAPI (blue), cardiomyocytes are marked with f-actin (green). The dotted orange line marks the edge of the fibrotic area. Scale bar, 50 µm. *P<0.05, unpaired Student t test. D, Schematic of experimental setup for integrin-αVβ6 (ITGAV/B6) blocking experiments in cardiac slice culture with related results in E. Icons are derived from BioRender. E, Reverse transcription quantitative polymerase chain reaction analysis of expression of genes downstream of transforming growth factor–β (TGF-β) signaling in cardiac slice cultures treated with inhibiting anti-ITGAV/B6 (1:15) or 10 µmol/L GW788388, an inhibitor of TGF-β receptor I, for 24 hours. *P< 0.05, paired Student t test versus indicated control condition. F, Schematic of experimental setup for in vivo ITGAV/B6 blocking experiments by injection of 40 mg/kg EMD527040 (EMD) intraperitoneally daily. DMSO was applied as vehicle control. Icons are derived from BioRender. G, Reverse transcription quantitative polymerase chain reaction expression analysis of genes downstream of TGF-β signaling in hearts of mice treated with EMD or respective amount of DMSO for 10 days. *P<0.05, unpaired Student t test versus DMSO. H and I, Cardiac fibrosis detected by picrosirius red collagen staining with representative images and corresponding analysis of the area of collagen in the right ventricle (RV) and left ventricle (LV). Lines indicate littermates. Each dot represents 1 animal. *P<0.05 or as indicated, grouped 2-way repeated-measures analysis of variance with LV/RV and experimental pairs matched, Sidak post hoc test. Scale bar, 1 mm. J through L, ECG recoded in lead II with representative curves shown in J. Corresponding analysis of R, S, and J peak amplitude and QRS interval, *P<0.05 or as indicated. Paired Student t test. Lines indicate littermates. Each dot represents 1 animal.\nITGAV/B6 dimers have the ability to activate the profibrotic cytokine transforming growth factor–β (TGF-β) by binding and removal of the latency-associated peptide.17 Gene set enrichment analyses revealed an upregulation of genes associated with TGF-β signaling in both 9-week-old mutants as well as patients with ACM (Figure 6A). Moreover, direct targets of receptor-regulated SMADs involved in TGF-β signaling21 were upregulated in DSG2-W2A mutant hearts (Figure 6B). Accordingly, increased amounts of nuclear SMAD2/3 phosphorylated at the activation sites S465/S467 or S423/S425, respectively, were detected in these hearts (Figure 6C). Increased levels of pSMAD2/3 were found not only in fibroblasts but also in cardiomyocytes, mainly adjacent to fibrotic areas. These data indicate upregulation of the profibrotic TGF-β pathway and its downstream targets, which include extracellular matrix proteins such as collagens, laminin, or fibronectin.\nElevated transforming growth factor–β signaling in DSG2 (desmoglein-2)–W2A hearts as result of ITGAV/B6 activity. Barcode plots of gene set enrichment analysis of (A) the KEGG_TGF_BETA_SIGNALING_PATHWAY data set (systematic name: M2642)41 and (B) genes directly regulated by receptor-regulated SMADs (R-SMADs; includes SMAD1/2/3/5/9) as published in the TRRUST database21 in 9-week-old DSG2-W2A (mut/mut vs wt/wt) or arrhythmogenic cardiomyopathy (ACM) patient data set 1 (ACM versus healthy control; GEO: GSE107157/GSE107480). Indicated P values are calculated by function cameraPR of the R package limma. C, Immunostaining of phosphorylated SMAD2/3 (magenta, S465/S467 or S423/S425, respectively) in sections of DSG2-W2A hearts and related analysis of nuclear staining intensity. Nuclei are stained with DAPI (blue), cardiomyocytes are marked with f-actin (green). The dotted orange line marks the edge of the fibrotic area. Scale bar, 50 µm. *P<0.05, unpaired Student t test. D, Schematic of experimental setup for integrin-αVβ6 (ITGAV/B6) blocking experiments in cardiac slice culture with related results in E. Icons are derived from BioRender. E, Reverse transcription quantitative polymerase chain reaction analysis of expression of genes downstream of transforming growth factor–β (TGF-β) signaling in cardiac slice cultures treated with inhibiting anti-ITGAV/B6 (1:15) or 10 µmol/L GW788388, an inhibitor of TGF-β receptor I, for 24 hours. *P< 0.05, paired Student t test versus indicated control condition. F, Schematic of experimental setup for in vivo ITGAV/B6 blocking experiments by injection of 40 mg/kg EMD527040 (EMD) intraperitoneally daily. DMSO was applied as vehicle control. Icons are derived from BioRender. G, Reverse transcription quantitative polymerase chain reaction expression analysis of genes downstream of TGF-β signaling in hearts of mice treated with EMD or respective amount of DMSO for 10 days. *P<0.05, unpaired Student t test versus DMSO. H and I, Cardiac fibrosis detected by picrosirius red collagen staining with representative images and corresponding analysis of the area of collagen in the right ventricle (RV) and left ventricle (LV). Lines indicate littermates. Each dot represents 1 animal. *P<0.05 or as indicated, grouped 2-way repeated-measures analysis of variance with LV/RV and experimental pairs matched, Sidak post hoc test. Scale bar, 1 mm. J through L, ECG recoded in lead II with representative curves shown in J. Corresponding analysis of R, S, and J peak amplitude and QRS interval, *P<0.05 or as indicated. Paired Student t test. Lines indicate littermates. Each dot represents 1 animal.\n[SUBTITLE] Fibrosis in DSG2-W2A Mutant Animals Is Reduced by Inhibition of ITGAV/B6-Dependent Release of TGF-β [SUBSECTION] On the basis of the known profibrotic role of TGF-β in cardiomyopathies2 and the established function of ITGAV/B6 dimers to activate TGF-β,17 we investigated whether inhibition of ITGAV/B6 was efficient to reduce fibrosis in DSG2-W2A mice. Cardiac slice cultures were generated from the ventricles of heterozygous mice at the age of 40 to 50 weeks and treated for 24 hours with anti-ITGAV/B6, which was shown to neutralize the function of the dimer.22 In comparison, slices were treated with the TGF-β receptor I inhibitor GW788388 to directly block TGF-β signaling (Figure 6D). Significant downregulation in the expression of the profibrotic molecules collagen-I type-α1 (Col1a1), laminin subunit-γ2 (Lamc2), metalloproteinase inhibitor-1 (Timp1), and ID-2 (Id2) was induced in the mutant mice in response to inhibition of ITGAV/B6 (Figure 6E). Other extracellular matrix proteins such as fibronectin (Fn1) or collagens (Col3a1, Col1a2) showed the same tendency. All of these fibrotic markers were significantly upregulated in adult mutant hearts as detected by RNA sequencing (Figure 6A and 6B). Direct inhibition of TGF-β led to a similar downregulation of these markers as the blocking antibody.\nTo investigate the in vivo relevance of this pathway, we applied the small molecule EMD527040 (EMD), which is an established inhibitor of ITGAV/B6-dependent TGF-β release, to the DSG2-W2A mouse model.23 For these experiments, we chose wt/mut mice because they reflect the heterozygosity most commonly found in patients with ACM and develop the phenotype during adulthood, when compound application is feasible. Littermates between the ages of 28 and 38 weeks were sex-matched and injected intraperitoneally daily for 10 days with EMD or DMSO control (Figure 6F). In this short-time approach, myocardial samples of EMD-treated animals showed a similar reduction of profibrotic markers as reported for slice cultures (Figure 6G). Having established an effective dose, we performed in vivo studies for a duration of 60 days during the time period when RV fibrosis establishes. At the end of the treatment period, ECG and histologic analysis were performed. EMD-treated mice demonstrated reduced RV fibrosis (Figure 6H and 6I). Moreover, EMD animals showed a mitigation of ECG abnormalities, with significantly higher S amplitude and trends towards an increased R amplitude and shorter QRS interval compared with the vehicle-treated control.\nThese experiments link ITGAV/B6 dimerization in response to abrogated DSG2 binding and ICD dysfunction to TGF-β–dependent fibrosis generation. The pilot inhibitor studies suggest ITGAV/B6 as a promising treatment target to ameliorate the ACM phenotype.\nOn the basis of the known profibrotic role of TGF-β in cardiomyopathies2 and the established function of ITGAV/B6 dimers to activate TGF-β,17 we investigated whether inhibition of ITGAV/B6 was efficient to reduce fibrosis in DSG2-W2A mice. Cardiac slice cultures were generated from the ventricles of heterozygous mice at the age of 40 to 50 weeks and treated for 24 hours with anti-ITGAV/B6, which was shown to neutralize the function of the dimer.22 In comparison, slices were treated with the TGF-β receptor I inhibitor GW788388 to directly block TGF-β signaling (Figure 6D). Significant downregulation in the expression of the profibrotic molecules collagen-I type-α1 (Col1a1), laminin subunit-γ2 (Lamc2), metalloproteinase inhibitor-1 (Timp1), and ID-2 (Id2) was induced in the mutant mice in response to inhibition of ITGAV/B6 (Figure 6E). Other extracellular matrix proteins such as fibronectin (Fn1) or collagens (Col3a1, Col1a2) showed the same tendency. All of these fibrotic markers were significantly upregulated in adult mutant hearts as detected by RNA sequencing (Figure 6A and 6B). Direct inhibition of TGF-β led to a similar downregulation of these markers as the blocking antibody.\nTo investigate the in vivo relevance of this pathway, we applied the small molecule EMD527040 (EMD), which is an established inhibitor of ITGAV/B6-dependent TGF-β release, to the DSG2-W2A mouse model.23 For these experiments, we chose wt/mut mice because they reflect the heterozygosity most commonly found in patients with ACM and develop the phenotype during adulthood, when compound application is feasible. Littermates between the ages of 28 and 38 weeks were sex-matched and injected intraperitoneally daily for 10 days with EMD or DMSO control (Figure 6F). In this short-time approach, myocardial samples of EMD-treated animals showed a similar reduction of profibrotic markers as reported for slice cultures (Figure 6G). Having established an effective dose, we performed in vivo studies for a duration of 60 days during the time period when RV fibrosis establishes. At the end of the treatment period, ECG and histologic analysis were performed. EMD-treated mice demonstrated reduced RV fibrosis (Figure 6H and 6I). Moreover, EMD animals showed a mitigation of ECG abnormalities, with significantly higher S amplitude and trends towards an increased R amplitude and shorter QRS interval compared with the vehicle-treated control.\nThese experiments link ITGAV/B6 dimerization in response to abrogated DSG2 binding and ICD dysfunction to TGF-β–dependent fibrosis generation. The pilot inhibitor studies suggest ITGAV/B6 as a promising treatment target to ameliorate the ACM phenotype.", "To study the functional consequences of impaired DSG2 binding, we generated the W2A point mutation (DSG2-W2A) to abrogate the proposed interaction mechanism of the only desmoglein expressed in cardiomyocytes (Figure 1A). First, we investigated in a cell-free setup whether DSG2-W2A is indeed affecting the binding properties of DSG2 interaction by applying single molecule force spectroscopy. Here, molecules are coupled to the tip of an atomic force microscopy probe and the surface of a mica sheet by a polyethylene glycol linker. By measuring the deflection of the probe during repeated approach to and retraction from the surface, binding events can be assessed quantitatively. From these data, properties such as binding frequency, binding force, and bond half-life time can be determined. Constructs were generated for expression of wild-type (WT) and mutant (W2A) DSG2 extracellular domains fused to a human Fc fragment and tested for homotypic (WT:WT, W2A:W2A) and heterotypic (WT:W2A) interaction properties (Figure 1B). These experiments showed a significant reduction of the frequency of W2A:W2A as well as WT:W2A interactions compared with WT:WT (Figure 1C). The frequency of the remaining W2A bindings was comparable to probing Fc alone, which indicates that these are mostly nonspecific interactions. Homotypic WT interactions display a clear binding force peak, whereas the remaining W2A interaction forces were spread widely, again pointing to the nonspecificity of these interactions (Figure 1D). The lifetime of DSG2 interactions under zero force (τ0) was determined by fitting the binding forces detected at different loading rates,11 yielding τ0=1.088s with R=0.993. In contrast, no sufficient fit (R=0.509) was possible for W2A interactions (Figure 1E). Together, these data outline that the tryptophan swap is the major interaction mechanism of DSG2.\nDSG2 (desmoglein-2) adhesion is mediated by tryptophan swap at position 2. A, Predicted interaction model of DSG2 extracellular domains by exchange of tryptophan residue at position 2 into a hydrophobic pocket of the opposing molecule. Cartoon 3D presentation of Protein Data Bank entry 5ERD9; tryptophan-2 is highlighted by ball and stick presentation. B, Schematic of single molecule force spectroscopy experiments. Recombinant extracellular domains (ECs) of wild-type DSG2 (DSG2-WT) or mutant DSG2 (DSG2-W2A) protein were coupled to a mica surface and atomic force microscopy cantilever by mean of a human Fc-tag (hFc) and a polyethylene glycol (PEG)20 linker and probed as indicated. C, Binding frequency of DSG2-W2A/DSG2-WT heterotypic and homotypic interactions at a pulling speed of 2 µm/s are shown. hFc served as control for nonspecific binding. *P<0.05, 1-way analysis of variance, Dunnett post hoc test. Each independent coating procedure with minimum 625 force curves is taken as biological replicate. D, Histogram of binding forces distribution with peak fit at a pulling speed of 2 µm/s corresponding to data in C. E, Determination of the bond half lifetime by the Bell equation11 of mean loading rates and binding forces analyzed from data of pulling speeds at 0.5, 1, 2, 5, and 7.5 µm/s. Average of values from 4 independent coating procedures with minimum 625 force curves each. R=R2, koff =off-rate constant, τ0=bond half lifetime under zero force. F, Dissociation assays to determine cell–cell adhesion were performed in CaCo2 cells (WT or DSG2 KO background) expressing DSG2-WT-mGFP or DSG2-W2A-mGFP constructs. mGFP empty vector served as control. *P<0.05, 1-way analysis of variance, Sidak post hoc test. Corresponding Western blot analysis confirmed effective expression of DSG2 constructs (*) versus the endogenous protein (arrow) in CaCo2 cells. α-tubulin (TUBA) served as loading control. G, Representative images of monolayer fragmentation from experiments in F. H, Dissociation assays in immortalized keratinocytes isolated from neonatal murine skin of the respective genotype. *P<0.05 or as indicated, Welch analysis of variance, Dunnett post hoc test. I, Macroscopic cardiac phenotype of DSG2-W2A mut/mut mice at age 15 weeks. J, Cardiac hypertrophy was analyzed as mean cross-sectional area of cardiomyocytes in hematoxylin & eosin–stained sections. Scale bar, 30 µm. *P<0.05, unpaired Student t test. K, Representative images of intercalated discs acquired by transmission electron microscopy, 3 mice per genotype. Orange asterisks mark intercellular widening, orange circle marks a ruptured junction. Scale bar, 1 µm.\nTo determine the effect of the W2A mutation on a cellular level, DSG2-WT and DSG2-W2A constructs were stably expressed in the epithelial cell line CaCo2 either in a WT or DSG2-deficient background (CaCo2[WT] or CaCo2[DSG2 KO]).12 Cell–cell dissociation assays were performed, in which a confluent cell monolayer is detached and exposed to defined mechanical stress. Expression of DSG2-W2A in CaCo2(WT) cells significantly reduced intercellular adhesion as indicated by an increased number of fragments (Figure 1F and 1G). Whereas expressing DSG2-WT in the CaCo2(DSG2 KO) line significantly rescued the impaired intercellular adhesion in response to DSG2 loss, the expression of DSG2-W2A had no effect on fragment numbers.\nBecause these data demonstrate the DSG2 tryptophan swap to be the central adhesive mechanism, we next generated a CRISPR/Cas9-based knock-in mouse model for DSG2-W2A to investigate the consequences of impaired DSG2 adhesion in vivo (Figure S1). In line with the studies in human cells, dissociation assays in keratinocyte cell lines generated from DSG2-W2A pups revealed reduced cell–cell adhesion in heterozygous (mut/wt) and homozygous (mut/mut) mutants (Figure 1H). Moreover, DSG2-W2A mice presented with a pronounced cardiac phenotype. Adult mut/mut mice demonstrated ventricular deformation, fibrotic and calcified areas, and hypertrophic cardiomyocytes (Figure 1I and 1J). In line with reduced intercellular adhesion of cardiomyocytes, transmission electron microscopy revealed disturbed ICDs with widened intercellular space and occasionally completely ruptured junctions in mut/mut hearts (Figure 1K).\nA cardiac phenotype was also detectable during embryogenesis. Here, mating of heterozygous mice revealed a reduction of mut/mut offspring to 2.8% compared with the expected Mendelian ratio of 25%. Embryo dissections showed loss of the mut/mut animals between E12 and E14 (Figure S2A). The mut/mut embryos at day E12.5 appeared macroscopically pale with accumulation of blood in the cardiac area although the heart was still beating (Figure S2B). Cells suggestive for blood precursors were detectable in the pericardial space (Figure S2C). This finding suggests a rupture of the cardiac wall leading to pericardial bleeding and loss of mut/mut animals.\nTogether, these in vitro and in vivo data outline an essential role of the DSG2 tryptophan swap for cell–cell adhesion and demonstrate severe pathologies associated with impaired DSG2 interaction.", "Given the cardiac affection in mutant mice, the notion that a majority of patients with ACM are reported with mutations in desmosomal genes,2 and that mutations specifically of W2 have already been associated with ACM,10 we examined homozygous and heterozygous mice including both sexes with regard to histologic and clinical ACM measures in an age range of 12 to 80 weeks.\nHistologic analysis showed cardiac fibrosis, a major hallmark of ACM, in the myocardium of the right ventricle (RV) and left ventricle (LV) in mut/mut animals (Figure 2A through 2D), which did not profoundly increase over time. In contrast, heterozygous animals demonstrated a milder phenotype, with a fraction of animals (36%) starting to develop fibrosis in the RV around 6 months, whereas LV was unaffected. To better address differences over time, we pooled animals into groups with a mean age of 4 and 6 months for homozygous and 6 and 12 months for heterozygous mice and performed functional analyses by echocardiography and ECG parallel to histologic evaluation. Moreover, we separated 12-month-old heterozygous animals according to RV fibrosis into a group without and with fibrosis (>10% fibrotic area). Mut/mut animals showed impaired RV systolic function at both time points with reduced tricuspid annular plane systolic excursion as well as fractional shortening (Figure 2E through 2G and Table S1). LV systolic impairment with reduction of the ejection fraction and corresponding parameters worsened over time and became significant after 6 months, although no increase in fibrosis was detectable (Figure 2D and 2H and Table S1). In mut/wt mice, RV fractional shortening was significantly reduced in 12-month-old animals with fibrosis. In ECG recordings of mut/mut mice for 30 minutes under anaesthesia, we noted deformation of the QRS complex with elongated QRS interval and reduced amplitude of the S peak with effects more pronounced in the older group; heterozygous animals with RV fibrosis showed similar effects or trends (Figure 2I through 2K and Table S1). Because the T wave was not clearly detectable in mutants, the J peak amplitude as indicator for early repolarization was determined. This was reduced in the mut/mut groups with same trend for fibrotic mut/wt hearts (Figure 2L). Parallel to these depolarization and repolarization abnormalities, we also noted the occurrence of ventricular arrhythmia, ranging from single premature ventricular contractions (PVCs) to multiple, multifocal PVCs and nonsustained ventricular tachycardia in 1 mouse (Figure 2M through 2O). The fraction of homozygous and heterozygous animals with PVCs and the frequency of these events correlated with age and fibrosis. Moreover, a substantial fraction (16%) of mut/mut animals was found dead during the observation period, which strongly suggests the occurrence of malignant arrhythmia with sudden death (Figure 2P). Investigating sex-related differences, no effect was observed on the extent of fibrosis or echocardiographic parameters. However, in mut/wt mice, PVCs were found in males only and male mut/mut mice were more prone to premature sudden death compared with females (Figure S3).\nDSG2 (desmoglein-2)–W2A mutant mice develop characteristics of arrhythmogenic cardiomyopathy. A, Cardiac fibrosis detected by picrosirius red collagen staining with representative images of sections from 6-month-old DSG2-W2A mut/mut and 12-month-old wt/wt and mut/wt animals. Scale bar = 1 mm. B through D, Corresponding analysis of the area of collagen in the right ventricle (RV) and left ventricle (LV). Continuous lines indicate the simple linear regression between age and area of collagen. Hearts with >10% of collagen in the RV (gray dotted line) were defined as “with fibrosis.” Each dot represents 1 animal. *P<0.05, mixed effects analysis with LV and RV matched per animal, Sidak post hoc test. Lines indicate median and quartile values. E, Representative echocardiography images for measurements of the tricuspid annular plane systolic excursion (TAPSE) and LV function in 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Left side: 2D images from apical 4-chamber view for TAPSE and parasternal short axis view for LV. M-mode tracings on the right were performed along the line indicated on the left. Scale bars, white, 2 mm; black, 100 ms. Corresponding analysis of (F) TAPSE, *P<0.05, Kruskal-Wallis test with Dunn post hoc test; (G) fractional shortening of the RV, *P<0.05, Kruskal-Wallis test with Dunn post hoc test; and (H) ejection fraction of the LV, *P<0.05, 1-way analysis of variance, Sidak post hoc test. I, ECG recoded in lead II with representative curves from 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Definition of respective peaks is indicated in the wt/wt curve. Scale bars, vertical, 0.5 mV; horizontal, 50 ms. Corresponding analysis of (J) QRS interval, *P<0.05 or as indicated, Kruskal-Wallis test with Dunn post hoc test; (K) amplitude of the S peak, *P<0.05 or as indicated, 1-way analysis of variance, Sidak post hoc test; and (L) amplitude of the J peak (early repolarization), *P<0.05 or as indicated, Kruskal-Wallis test with Dunn post hoc test. M, Ventricular arrythmia detected by ECG during 30 minutes of baseline measurements with example curves from 6-month-old mut/mut and 12-month-old wt/wt and mut/wt animals. Asterisks mark premature ventricular contractions (PVCs); *nsVT indicates a nonsustained ventricular tachycardia detected in 1 mut/mut animal. Scale bars, vertical, 0.5 mV; horizontal, 1 s. Corresponding analysis of (N) percentage of mice presenting with PVCs. Values in bars indicate corresponding absolute number of mice with PVC (colored bars) compared with total number of mice evaluated (empty bar) and (O) PVC burden depicted as number of PVCs per minute, *P<0.05, Kruskal-Wallis test with Dunn post hoc test. The black arrow indicates the animal presenting with nonsustained ventricular tachycardia. P, Kaplan-Meier survival plot of DSG2-W2A mice from an analysis period of 3 years. Vertical lines indicate dropouts because of unrelated elimination (end of experiment, breeding, injuries). Values indicate corresponding absolute number of mice with sudden death compared with total number of mice evaluated. *P<0.05, Gehan-Breslow-Wilcoxon test. Box with color indications of respective groups in the middle applies to the entire figure.\nIn summary, mutant animals show histopathologic features with echocardiography and ECG abnormalities similar to patients with ACM. Within the limits of a murine model, both genotypes fulfil the Padua criteria13 to establish the diagnosis of ACM (Table S2). Here, DSG2-W2A mut/mut animals resemble the phenotype of biventricular ACM, whereas in mut/wt mice, the phenotype is milder with variable penetrance and age-dependent occurrence only in the RV. According to the Padua criteria,13 the heterozygous genotype recalls the characteristics of a right-dominant ventricular ACM, as long as fibrosis was present. Together, these data demonstrate that loss of desmosomal adhesion is sufficient to induce an ACM phenotype and that the DSG2-W2A model may resemble 2 different variants of the disease.", "Next, we applied this mouse model to investigate mechanisms leading to ACM. RNA sequencing was performed from ventricles of mut/mut and wt/wt hearts both before onset of fibrosis (age 5 days) and when the biventricular fibrosis and ACM phenotype was established (after 9 weeks). First, we compared sequencing data from 9-week-old mice with the top differentially expressed genes derived from published transcriptomic data sets of ACM patient hearts (GEO database: GSE107157/GSE10748014 and GSE2981915). Murine samples clustered according to their genotype and mut/mut hearts resembled the gene expression pattern of patients (Figure S4), further supporting the observation that mutant mice faithfully reproduce major aspects of the disease.\nTo identify common genes deregulated during ACM pathogenesis, we compared the differentially expressed genes from patient data with the results from mutant mice. Here, we included data from 5-day-old mice to identify changes already present before onset of secondary effects attributable to fibrosis. Integrin-β6 (Itgb6) was the only transcript consistently deregulated in all data sets (Figure 3A). Its common downregulation in patients with ACM and mutant mice on RNA levels was further confirmed for heterozygous DSG2-W2A mutants compared with WT (Figure 3B). However, protein levels were unaltered in mutant heart samples (Figure 3C). Immunostaining revealed that, in mutant hearts, ITGB6, which mainly localized to a compartment suggestive for the transverse tubules and costameres in wt/wt animals, was enriched at the ICD (Figure 3D and E). Together, these data demonstrate altered ITGB6 mRNA expression as a common feature in ACM patient and DSG2-W2A hearts and an enrichment of the protein at the ICD in mutant murine samples.\nIntegrin-β6 is deregulated in DSG2 (desmoglein-2)–W2A mutants. A, Venn diagram of significantly altered genes from indicated arrhythmogenic cardiomyopathy (ACM) patient data sets (ACM versus healthy control) and DSG2-W2A mice at the age of 5 days and 9 weeks (mut/mut versus wt/wt) highlighting integrin-β6 (Itgb6) as only overlapping gene with same direction of expression in all data sets. Numbers indicate the amount of overlapping genes for the respective overlays. B, RNA expression of Itgb6 analyzed by means of reverse transcription quantitative polymerase chain reaction in adult DSG2-W2A mouse hearts. *P<0.05, unpaired Student t test versus wt/wt. Gapdh served as reference gene. C, Representative Western blot and respective analysis of band intensity of integrin-β6 (ITGB6) in DSG2-W2A hearts. GAPDH served as loading control. 1-way analysis of variance, Dunnett post hoc test. D and E, Immunostaining of ITGB6 (red in overlay) in DSG2-W2A hearts with corresponding analysis of staining intensity in cardiomyocytes in total and ratio of staining intensity at the intercalated disc (ICD) area (orange arrowheads) versus cardiomyocytes’ cytosolic area. DSP (desmoplakin; cyan) marks ICDs, DAPI (blue) nuclei and F-actin (green) the sarcomere system. Lower row shows an overview image of a fibrotic area in mut/mut hearts. Scale bars, 25 µm. *P<0.05; left graph: Kruskal-Wallis test with Dunn post hoc test; right graph: 1-way analysis of variance, Dunnett post hoc test. Box with color indications of respective groups applies to the entire figure.", "This result together with the transmission electron microscopy data (Figure 1K) suggest structural changes of the ICD in mutant hearts, which we addressed further. Using DSP as marker, we reconstructed the outlines of ICDs in wt/wt and mut/mut hearts from 3D stacks captured with structured illumination microscopy. This analysis yielded an increase in ICD volume, in particular attributable to an enhanced width between 2 adjacent cells (Figure 4A). The majority of desmosomal molecules as well as N-cadherin were regularly distributed at mutant ICDs (Figure S5A and S5B). This was corroborated by RNA sequencing data from 5-day-old and 9-week-old animals, which showed no consistent changes of desmosomal, adherens, gap, or tight junction molecules (Figure S5C through S5E). The only adhesion molecule reduced globally was DSG2 (Figure S5A and S5B). Although DSG2 was still present at the ICD, detailed analysis by structured illumination microscopy revealed reduced number and volume of DSG2 signals (Figure 4B). Moreover, fluorescence recovery after photobleaching experiments in neonatal murine cardiomyocytes transduced with DSG2-WT-mGFP or DSG2-W2A-mGFP showed a higher mobility of the mutant protein at cardiomyocyte junctions (Figure 4C), indicating reduced membrane stability of DSG2-W2A. To further investigate the possibility that the stability and integrity of ICD molecules is compromised, we performed Triton-X-100 separation assays for proteins with unaltered ICD localization in mutant hearts. Cytosolic and unanchored membrane proteins are found in the Triton-X-100 soluble fraction, whereas cytoskeleton-bound molecules are mainly detectable in the nonsoluble fraction. Both desmosomal molecules (PG, PKP2) as well as N-cadherin showed increased levels in the soluble pool, indicating reduced cytoskeletal anchorage and impaired ICD integrity in mutants (Figure 4D). Moreover, the gap junction molecule connexin-43, which is required for electrical coupling of cardiomyocytes, was delocalized to the lateral membrane (Figure 4E), which was described as a feature of disrupted ICDs.8\nDisrupted ICD structure in DSG2 (desmoglein-2)–W2A mutant mice. A, Representative z-stack reconstruction of segmented intercalated discs (ICDs) in top and side view acquired with structured illumination microscopy. Overlay of analyzed ICD volume is shown in gray. ICDs are marked by DSP (desmoplakin; magenta). Scheme on top presents segmented area and pictograms on the right display the respective angle of view. Corresponding analysis of ICD volume and width of ICD between adjacent cardiomyocytes below. Scale bar, 5 µm. *P<0.05, Mann-Whitney test. Each dot represents the value of 1 ICD from 4 mice per genotype. B, Representative images of DSG2 (magenta) and filamentary actin (f-actin; white) z-stacks acquired by structured illumination microscopy and presented as maximum intensity projection. Lower row shows color-coded height projection of DSG2 signals in z-stack after signal thresholding as performed for analysis. Related analysis of DSG2 signal volume and number per ICD length is shown below. Pictograms on the right display the respective angle of view. Scale bar, 5 µm. *P<0.05, unpaired Student t test, with Welch correction. Each dot represents the value of 1 ICD from in total 3 mice per genotype. C, Fluorescence recovery after photobleaching analysis of DSG2–wild type (WT) and DSG2-W2A-mGFP fusion proteins at the cell–cell junction of neonatal cardiomyocytes with representative intensity kymographs of bleached areas on the left. Time point 0 = bleach as indicated by the black arrow. Analysis of the mobile fraction of the indicated mGFP-fusion proteins is shown on the right. *P<0.05, unpaired Student t test, with Welch correction. Each dot represents the mean value of 1 heart from in total 3 isolations. D, Representative triton-X-100 assay immunoblot with separation of a soluble (sol), noncytoskeletal bound protein fraction from a nonsoluble (non-sol), cytoskeletal-anchored fraction and corresponding analysis shown below. PG (plakoglobin), PKP2 (plakophilin-2), and N-cadherin (NCAD) were analyzed. Intensity of proteins was normalized to the total amount of protein detected by ponceau staining. GAPDH and DSP served as separation control. *P<0.05 or as indicated, unpaired Student t test (PG, NCAD, PKP2) or Mann-Whitney test (DSP). Each dot represents the result from 1 mouse. E, Immunostaining of connexin-43 (CX43; red in overlay) in DSG2-W2A hearts. NCAD (yellow) marks ICDs, DAPI (blue) nuclei, and F-actin (green) the sarcomere system. Orange arrowheads mark ICD, pink arrowheads highlight lateralization of CX43 staining. Scale bars, 25 µm. Images representative for 5 mice per genotype. Box with color indications of respective groups applies to the entire figure.\nThese data together with the transmission electron microscopy results demonstrate structurally severely altered ICDs in response to the DSG2-W2A mutation and suggest a molecular basis leading to the functional alterations detectable by echocardiography and ECG.", "In contrast to DSG2, we detected ITGB6 to be increased at the ICD (Figure 3D and 3E), which was confirmed by analysis of structured illumination microscopy images (Figure 5A). ITGB6 needs to heterodimerize with integrin-αV (ITGAV) in order to be activated and bind to the extracellular matrix.16–18 Thus, we analyzed the expression of the counterpart ITGAV, which was significantly upregulated on protein level and increased at the ICD and costameres of mutant hearts (Figure 5B and 5C). In contrast, the localization and intensity of integrin-β1, as classical representative of the integrin group, was not altered (Figure S6). Staining with an antibody specifically recognizing the heterodimer of ITGAV/B6 revealed increased amount of dimer formation at the ICD and costameres in mutant mice (Figure 5D). Moreover, increased ITGAV/B6 staining intensity was also detectable in an ACM patient sample with DSP mutation (Figure 5E). To stabilize active integrins, recruitment of the cytoskeletal adapters talin and vinculin is required.19 Accordingly, the expression of talin-2 was increased in DSG2-W2A mut/mut myocardium, with vinculin being enriched at the ICD (Figure 5F and 5G). This effect is similar to what was shown before in response to increased intracellular traction forces on junctions.20 Thus, these data suggest that in response to a dysfunctional, nonadhesive ICD with impaired cytoskeletal anchorage, ITGAV/B6 dimers are recruited and activated at the compromised ICD as well as at the costamere region.\nIncreased activation of ITGB6/AV at ICDs of DSG2 (desmoglein-2)–W2A mutant mice. A, Representative intercalated disc (ICD) reconstruction from z-stacks of integrin-β6 (ITGB6; green) and DSP (magenta) immunostaining acquired by structured illumination microscopy and presented as maximum intensity projection. Related analysis of ITGB6 signal volume and number per ICD length is shown below. Scale bar, 5 µm. *P<0.05 or as indicated, unpaired Student t test with Welch correction (left graph) and Mann-Whitney test (right graph). Each dot represents the value of 1 ICD from in total 4 mice per genotype. B, Representative Western blot and analysis of band intensity of integrin-αV (ITGAV) in DSG2-W2A hearts. GAPDH served as loading control. *P<0.05, unpaired Student t test with Welch correction. C, Immunostaining of ITGAV in DSG2-W2A hearts on the right with respective analysis on the left. DAPI (blue) marks nuclei. Lower row shows an overview image of a fibrotic area in mut/mut hearts. Dotted orange line marks the edge of fibrotic area. Scale bars: upper rows, 20 µm; lower row, 50 µm. *P<0.05, unpaired Student t test. D, Immunostaining of ITGAV/B6 heterodimer in DSG2-W2A mutant hearts with respective analysis of staining intensity on the right. Cyan rectangle marks zoomed area on the right. Scale bars: overview, 50 µm; insert, 10 µm. *P<0.05, unpaired Student t test. E, Representative immunostaining images of ITGAV/B6 heterodimer staining (red) in a patient with arrhythmogenic cardiomyopathy (ACM; DSP-E952X, heterozygous) and healthy control sample. F-actin (green) stains the sarcomere system. For the patient with ACM, 4 different tissue samples were analyzed and compared with 2 tissue samples from 2 healthy controls. Scale bar, 20 µm. F, Immunostaining of VCL (vinculin; magenta) and TLN2 (talin-2; red) in DSG2-W2A hearts on the right with respective analysis of the mean staining intensity in cardiomyocytes and ratio of the staining intensity at the ICD versus cytosolic area on the left. DAPI (blue) marks nuclei. F-actin (green) stains the sarcomere system. Scale bar, 25 µm; *P<0.05, unpaired Student t test. G, Representative Western blot and analysis of band intensity of VCL and TLN2 in DSG2-W2A hearts. GAPDH served as loading control. *P<0.05, unpaired Student t test for TLN2, Mann-Whitney test for VCL. Box with color indications of respective groups applies to the entire figure.", "ITGAV/B6 dimers have the ability to activate the profibrotic cytokine transforming growth factor–β (TGF-β) by binding and removal of the latency-associated peptide.17 Gene set enrichment analyses revealed an upregulation of genes associated with TGF-β signaling in both 9-week-old mutants as well as patients with ACM (Figure 6A). Moreover, direct targets of receptor-regulated SMADs involved in TGF-β signaling21 were upregulated in DSG2-W2A mutant hearts (Figure 6B). Accordingly, increased amounts of nuclear SMAD2/3 phosphorylated at the activation sites S465/S467 or S423/S425, respectively, were detected in these hearts (Figure 6C). Increased levels of pSMAD2/3 were found not only in fibroblasts but also in cardiomyocytes, mainly adjacent to fibrotic areas. These data indicate upregulation of the profibrotic TGF-β pathway and its downstream targets, which include extracellular matrix proteins such as collagens, laminin, or fibronectin.\nElevated transforming growth factor–β signaling in DSG2 (desmoglein-2)–W2A hearts as result of ITGAV/B6 activity. Barcode plots of gene set enrichment analysis of (A) the KEGG_TGF_BETA_SIGNALING_PATHWAY data set (systematic name: M2642)41 and (B) genes directly regulated by receptor-regulated SMADs (R-SMADs; includes SMAD1/2/3/5/9) as published in the TRRUST database21 in 9-week-old DSG2-W2A (mut/mut vs wt/wt) or arrhythmogenic cardiomyopathy (ACM) patient data set 1 (ACM versus healthy control; GEO: GSE107157/GSE107480). Indicated P values are calculated by function cameraPR of the R package limma. C, Immunostaining of phosphorylated SMAD2/3 (magenta, S465/S467 or S423/S425, respectively) in sections of DSG2-W2A hearts and related analysis of nuclear staining intensity. Nuclei are stained with DAPI (blue), cardiomyocytes are marked with f-actin (green). The dotted orange line marks the edge of the fibrotic area. Scale bar, 50 µm. *P<0.05, unpaired Student t test. D, Schematic of experimental setup for integrin-αVβ6 (ITGAV/B6) blocking experiments in cardiac slice culture with related results in E. Icons are derived from BioRender. E, Reverse transcription quantitative polymerase chain reaction analysis of expression of genes downstream of transforming growth factor–β (TGF-β) signaling in cardiac slice cultures treated with inhibiting anti-ITGAV/B6 (1:15) or 10 µmol/L GW788388, an inhibitor of TGF-β receptor I, for 24 hours. *P< 0.05, paired Student t test versus indicated control condition. F, Schematic of experimental setup for in vivo ITGAV/B6 blocking experiments by injection of 40 mg/kg EMD527040 (EMD) intraperitoneally daily. DMSO was applied as vehicle control. Icons are derived from BioRender. G, Reverse transcription quantitative polymerase chain reaction expression analysis of genes downstream of TGF-β signaling in hearts of mice treated with EMD or respective amount of DMSO for 10 days. *P<0.05, unpaired Student t test versus DMSO. H and I, Cardiac fibrosis detected by picrosirius red collagen staining with representative images and corresponding analysis of the area of collagen in the right ventricle (RV) and left ventricle (LV). Lines indicate littermates. Each dot represents 1 animal. *P<0.05 or as indicated, grouped 2-way repeated-measures analysis of variance with LV/RV and experimental pairs matched, Sidak post hoc test. Scale bar, 1 mm. J through L, ECG recoded in lead II with representative curves shown in J. Corresponding analysis of R, S, and J peak amplitude and QRS interval, *P<0.05 or as indicated. Paired Student t test. Lines indicate littermates. Each dot represents 1 animal.", "On the basis of the known profibrotic role of TGF-β in cardiomyopathies2 and the established function of ITGAV/B6 dimers to activate TGF-β,17 we investigated whether inhibition of ITGAV/B6 was efficient to reduce fibrosis in DSG2-W2A mice. Cardiac slice cultures were generated from the ventricles of heterozygous mice at the age of 40 to 50 weeks and treated for 24 hours with anti-ITGAV/B6, which was shown to neutralize the function of the dimer.22 In comparison, slices were treated with the TGF-β receptor I inhibitor GW788388 to directly block TGF-β signaling (Figure 6D). Significant downregulation in the expression of the profibrotic molecules collagen-I type-α1 (Col1a1), laminin subunit-γ2 (Lamc2), metalloproteinase inhibitor-1 (Timp1), and ID-2 (Id2) was induced in the mutant mice in response to inhibition of ITGAV/B6 (Figure 6E). Other extracellular matrix proteins such as fibronectin (Fn1) or collagens (Col3a1, Col1a2) showed the same tendency. All of these fibrotic markers were significantly upregulated in adult mutant hearts as detected by RNA sequencing (Figure 6A and 6B). Direct inhibition of TGF-β led to a similar downregulation of these markers as the blocking antibody.\nTo investigate the in vivo relevance of this pathway, we applied the small molecule EMD527040 (EMD), which is an established inhibitor of ITGAV/B6-dependent TGF-β release, to the DSG2-W2A mouse model.23 For these experiments, we chose wt/mut mice because they reflect the heterozygosity most commonly found in patients with ACM and develop the phenotype during adulthood, when compound application is feasible. Littermates between the ages of 28 and 38 weeks were sex-matched and injected intraperitoneally daily for 10 days with EMD or DMSO control (Figure 6F). In this short-time approach, myocardial samples of EMD-treated animals showed a similar reduction of profibrotic markers as reported for slice cultures (Figure 6G). Having established an effective dose, we performed in vivo studies for a duration of 60 days during the time period when RV fibrosis establishes. At the end of the treatment period, ECG and histologic analysis were performed. EMD-treated mice demonstrated reduced RV fibrosis (Figure 6H and 6I). Moreover, EMD animals showed a mitigation of ECG abnormalities, with significantly higher S amplitude and trends towards an increased R amplitude and shorter QRS interval compared with the vehicle-treated control.\nThese experiments link ITGAV/B6 dimerization in response to abrogated DSG2 binding and ICD dysfunction to TGF-β–dependent fibrosis generation. The pilot inhibitor studies suggest ITGAV/B6 as a promising treatment target to ameliorate the ACM phenotype.", "In this study, we generated a knock-in mouse model with defective binding function of the adhesion molecule DSG2, which demonstrated that impaired desmosomal adhesion is sufficient to induce a phenotype mimicking the characteristics of ACM. Our data suggest a cascade of defective desmosomal adhesion, disrupted ICD structure, and subsequent activation of ITGAV/B6 with profibrotic TGF-β signaling as important underlying mechanisms leading to this phenotype (Figure 7). Moreover, our pilot study indicates a beneficial effect of ITGAV/B6 inhibition by EMD treatment with regard to fibrosis and several ECG parameters, suggesting that this pathway can be targeted successfully by drug treatment.\nSchematic conclusion of data. DSG2 (desmoglein-2) –W2A mutation with loss of desmosomal adhesion leads to impaired intercalated disc (ICD) structure with deregulation of integrin-β6 (ITGB6) and enhanced heterodimerization with integrin-αV (ITGAV). The dimer efficiently binds to the extracellular matrix and activates transforming growth factor–β (TGF-β) by removal of the latency-associated peptide (LAP). Active TGF-β can then induce profibrotic downstream signaling by means of SMAD molecules. In our experiments, this cascade was blocked by different approaches to inhibit ITGAV/B6.\n[SUBTITLE] Defective Desmosomal Adhesion by DSG2-W2A Mutation Is Inducing an ACM Phenotype [SUBSECTION] Because of mutations mainly affecting desmosomal genes and evidence of disrupted ICDs, the hypothesis of dysfunctional desmosomes with loss of cell–cell adhesion as a central pathologic step was adopted in the field.2 However, experimental data on this topic are contradictory.6–8 We show that W2 of DSG2 is central for binding and intercellular adhesion in vitro and in vivo. Disrupted desmosomal adhesion is sufficient to induce an ACM phenotype fulfilling the Padua criteria,13 including ventricular fibrosis, depolarization and repolarization abnormalities, arrhythmia, and impaired ventricular function, which are used to establish the diagnosis. In line with this, 3 mutations were described in patients with ACM directly affecting the DSG2 binding mechanism by exchange of the tryptophan with a serine, leucine, or arginine (ClinVar data bank: VCV000199796, VCV000044282, VCV000420241).10 This indicates a disruption of the tryptophan swap independent from the substituting amino acid and supports defective desmosomal adhesion as important factor in ACM.2,24 Moreover, different published DSG2 mutant mouse models show a phenotype resembling the features of DSG2-W2A mutants. These include a DSG2 mutant in which parts of the 2 outermost DSG2 extracellular domains were deleted (but leaving W2A intact),25 2 mouse lines with loss of DSG2,25,26 and mice overexpressing a patient mutation (DSG2-N271S),27 which also showed that ICD structural aberrations precede functional abnormalities and fibrosis generation. These data fuel the hypothesis that disruption and rearrangement of the ICD in response to impaired adhesion between cardiomyocytes is a crucial initial step, at least in case of mutations in desmosomal molecules. Other mechanisms described in patients or translational models, such as aberrant WNT or Hippo/YAP signaling, or immune cell infiltrations,2,28 may be secondary responses and in part even represent adaptive attempts to rescue the functional consequences of such severe structural aberrations.\nBecause of mutations mainly affecting desmosomal genes and evidence of disrupted ICDs, the hypothesis of dysfunctional desmosomes with loss of cell–cell adhesion as a central pathologic step was adopted in the field.2 However, experimental data on this topic are contradictory.6–8 We show that W2 of DSG2 is central for binding and intercellular adhesion in vitro and in vivo. Disrupted desmosomal adhesion is sufficient to induce an ACM phenotype fulfilling the Padua criteria,13 including ventricular fibrosis, depolarization and repolarization abnormalities, arrhythmia, and impaired ventricular function, which are used to establish the diagnosis. In line with this, 3 mutations were described in patients with ACM directly affecting the DSG2 binding mechanism by exchange of the tryptophan with a serine, leucine, or arginine (ClinVar data bank: VCV000199796, VCV000044282, VCV000420241).10 This indicates a disruption of the tryptophan swap independent from the substituting amino acid and supports defective desmosomal adhesion as important factor in ACM.2,24 Moreover, different published DSG2 mutant mouse models show a phenotype resembling the features of DSG2-W2A mutants. These include a DSG2 mutant in which parts of the 2 outermost DSG2 extracellular domains were deleted (but leaving W2A intact),25 2 mouse lines with loss of DSG2,25,26 and mice overexpressing a patient mutation (DSG2-N271S),27 which also showed that ICD structural aberrations precede functional abnormalities and fibrosis generation. These data fuel the hypothesis that disruption and rearrangement of the ICD in response to impaired adhesion between cardiomyocytes is a crucial initial step, at least in case of mutations in desmosomal molecules. Other mechanisms described in patients or translational models, such as aberrant WNT or Hippo/YAP signaling, or immune cell infiltrations,2,28 may be secondary responses and in part even represent adaptive attempts to rescue the functional consequences of such severe structural aberrations.\n[SUBTITLE] DSG2-W2A Mice as Model to Study ACM [SUBSECTION] Our results suggest the DSG2-W2A model as a valuable tool to study ACM mechanisms. Within the limitations of a murine model, it reproduces a majority of features found in patients with ACM (Table S2). In homozygous animals, both ventricles are affected from early on in life; heterozygous animals develop a milder phenotype, with fibrosis occurring only in the RV. Whether this milder phenotype would extend to the LV over time or whether a gene dose effect underlies the structural differences is unclear. LV and RV remodel differently in response to loading and injury29 and it is possible that the RV is less able to compensate a partial reduction of cardiomyocyte cohesion, in contrast to the LV. A fraction of mut/wt animals did not develop the phenotype at all within the observation period of up to 80 weeks. Thus, the model might help clarify why patients with the same mutation (ie, in the same family) develop the disease with variable penetrance.1 Similar to patients,1 male mutant mice appear to experience more arrhythmia and sudden death than do female mice.\nWe can interpret from the combined histology, echocardiography, and ECG data that the changes in heart function are secondary to fibrosis generation. This is indicated by the notion that functional measures (eg, ejection fraction, QRS interval) worsened over time in homozygous mutants, whereas no increase was detectable for fibrosis. Moreover, functional changes in mut/wt animals were only observed when fibrosis was present. This is in line with a patient cohort with DSP mutations, in which LV fibrosis preceded systolic dysfunction.30 A different study in DSG2-N271S-expressing ex vivo paced hearts demonstrated conduction velocity impairments before onset of fibrosis.27 More longitudinal studies using different mutational backgrounds and sensitive detection methods (eg, cardiac magnetic resonance imaging and ECG under exercise) are necessary to define the role of fibrosis as cause or consequence of functional changes.\nAs with all murine models, the DSG2-W2A line has limitations in its ability to fully recapitulate the human disease phenotype. Mutant mice do not show pronounced replacement with adipose tissue, which is a common characteristic in patient heart samples, but in general is not fully modeled in mice.24 Moreover, at least in ECGs under anesthesia, not all mutant animals develop arrhythmia, even if fibrosis is present. However, in contrast to most other ACM models, this knock-in model has the advantage of a single amino acid exchange under the endogenous promotor, thus reducing the possibility of secondary unwanted effects attributable to complete protein absence or overexpression. As in patients, the mutation is present in all cell types and not limited to cardiomyocytes.2,24\nOur results suggest the DSG2-W2A model as a valuable tool to study ACM mechanisms. Within the limitations of a murine model, it reproduces a majority of features found in patients with ACM (Table S2). In homozygous animals, both ventricles are affected from early on in life; heterozygous animals develop a milder phenotype, with fibrosis occurring only in the RV. Whether this milder phenotype would extend to the LV over time or whether a gene dose effect underlies the structural differences is unclear. LV and RV remodel differently in response to loading and injury29 and it is possible that the RV is less able to compensate a partial reduction of cardiomyocyte cohesion, in contrast to the LV. A fraction of mut/wt animals did not develop the phenotype at all within the observation period of up to 80 weeks. Thus, the model might help clarify why patients with the same mutation (ie, in the same family) develop the disease with variable penetrance.1 Similar to patients,1 male mutant mice appear to experience more arrhythmia and sudden death than do female mice.\nWe can interpret from the combined histology, echocardiography, and ECG data that the changes in heart function are secondary to fibrosis generation. This is indicated by the notion that functional measures (eg, ejection fraction, QRS interval) worsened over time in homozygous mutants, whereas no increase was detectable for fibrosis. Moreover, functional changes in mut/wt animals were only observed when fibrosis was present. This is in line with a patient cohort with DSP mutations, in which LV fibrosis preceded systolic dysfunction.30 A different study in DSG2-N271S-expressing ex vivo paced hearts demonstrated conduction velocity impairments before onset of fibrosis.27 More longitudinal studies using different mutational backgrounds and sensitive detection methods (eg, cardiac magnetic resonance imaging and ECG under exercise) are necessary to define the role of fibrosis as cause or consequence of functional changes.\nAs with all murine models, the DSG2-W2A line has limitations in its ability to fully recapitulate the human disease phenotype. Mutant mice do not show pronounced replacement with adipose tissue, which is a common characteristic in patient heart samples, but in general is not fully modeled in mice.24 Moreover, at least in ECGs under anesthesia, not all mutant animals develop arrhythmia, even if fibrosis is present. However, in contrast to most other ACM models, this knock-in model has the advantage of a single amino acid exchange under the endogenous promotor, thus reducing the possibility of secondary unwanted effects attributable to complete protein absence or overexpression. As in patients, the mutation is present in all cell types and not limited to cardiomyocytes.2,24\n[SUBTITLE] DSG2-W2A Leads to Dysfunctional ICDs and ITGAV/B6 Rearrangement [SUBSECTION] DSG2-W2A led to a severely altered ICD structure consistent with impaired cytoskeletal attachment and ruptured junctions. Whereas these changes provide explanations for compromised intercellular adhesion, we also noted alterations in cell–matrix protein distribution. RNA sequencing before and after onset of the disease phenotype and a comparison with ACM patient datasets identified ITGB6 as commonly deregulated in patients with ACM and DSG2-W2A mice. Although we noted reduced mRNA levels in mutant hearts, the overall protein content was unaltered. This suggests pronounced posttranslational regulation of ITGB6, which was already demonstrated in skeletal muscle.31 In mutant hearts, ITGB6 was increased at the ICD together with elevated levels of ITGAV and increased heterodimerization of both molecules. By mechanical force exerted through ITGAV/B6, TGF-β is detached from the latency-associated peptide and can induce signaling by means of TGF-β receptors.17,32 In line with higher intracellular forces, talin-2 and vinculin were increased at ICDs, as binding of these molecules activates and stabilizes integrins. A mutual regulation of both adhesive compartments is well-established. As an example, upon loss of N-cadherin, integrins are activated and induce fibronectin deposition.33 A similar mechanism is conceivable in DSG2-W2A hearts, either by loss of DSG2 or because of reduced N-cadherin anchorage. So far, only limited data are available on the role of integrins in ACM. A recent study showed downregulation of integrin-β1D leading to ventricular arrhythmia.34 Furthermore, knockdown of PKP2 in HL-1 cardiomyocytes was described to deregulate focal adhesions, including integrin-α1.35\nDSG2-W2A led to a severely altered ICD structure consistent with impaired cytoskeletal attachment and ruptured junctions. Whereas these changes provide explanations for compromised intercellular adhesion, we also noted alterations in cell–matrix protein distribution. RNA sequencing before and after onset of the disease phenotype and a comparison with ACM patient datasets identified ITGB6 as commonly deregulated in patients with ACM and DSG2-W2A mice. Although we noted reduced mRNA levels in mutant hearts, the overall protein content was unaltered. This suggests pronounced posttranslational regulation of ITGB6, which was already demonstrated in skeletal muscle.31 In mutant hearts, ITGB6 was increased at the ICD together with elevated levels of ITGAV and increased heterodimerization of both molecules. By mechanical force exerted through ITGAV/B6, TGF-β is detached from the latency-associated peptide and can induce signaling by means of TGF-β receptors.17,32 In line with higher intracellular forces, talin-2 and vinculin were increased at ICDs, as binding of these molecules activates and stabilizes integrins. A mutual regulation of both adhesive compartments is well-established. As an example, upon loss of N-cadherin, integrins are activated and induce fibronectin deposition.33 A similar mechanism is conceivable in DSG2-W2A hearts, either by loss of DSG2 or because of reduced N-cadherin anchorage. So far, only limited data are available on the role of integrins in ACM. A recent study showed downregulation of integrin-β1D leading to ventricular arrhythmia.34 Furthermore, knockdown of PKP2 in HL-1 cardiomyocytes was described to deregulate focal adhesions, including integrin-α1.35\n[SUBTITLE] Activation of TGF-β Signaling by ITGAV/B6 as Potential Therapeutic Target in ACM [SUBSECTION] The ITGAV/B6 heterodimer is described as one of the major activators of latent TGF-β1 and TGF-β3.17 Although TGF-β signaling is known as general driver of cardiac fibrosis36 and more specifically was implicated in ACM,2 to our knowledge no data are available on the role of ITGAV/B6 and their regulation of TGF-β signaling in cardiac fibrosis. Uncovering this pathway is of high interest as it offers the possibility to target TGF-β with reduced risk of severe side effects occurring in response to direct inhibition.37 We demonstrate a reduction of profibrotic gene expression under ACM conditions in response to ITGAV/B6 inhibition and our pilot study data suggest that a small molecule blocking ITGAV/B6-dependent TGF-β release diminished the generation of fibrosis. Similar approaches using this small molecule or neutralizing antibodies were shown to be protective in murine models of lung, liver, and biliary fibrosis.23,38,39 However, because of the heterogeneity of the phenotype in heterozygous animals, more detailed studies with larger sample sizes are required to further substantiate this finding.\nWhether fibrosis generation is a contributor to the disease or a protective mechanism to ensure integrity of the heart under conditions of compromised adhesion is unclear. In case of the latter, pharmacologic inhibition of fibrosis generation might be detrimental. Nevertheless, fibrosis is a driver of arrhythmia generation,40 which is also supported by our results that ECG abnormalities were reduced by inhibition of fibrosis. Careful studies using different therapeutic approaches to reduce fibrosis in appropriate model organisms need to address this aspect in detail.\nIn conclusion, we established a new mouse model phenocopying many aspects of ACM, uncovered a novel pathway of fibrosis induction, and identified an approach to target this mechanism with future implication as a potential therapeutic option.\nThe ITGAV/B6 heterodimer is described as one of the major activators of latent TGF-β1 and TGF-β3.17 Although TGF-β signaling is known as general driver of cardiac fibrosis36 and more specifically was implicated in ACM,2 to our knowledge no data are available on the role of ITGAV/B6 and their regulation of TGF-β signaling in cardiac fibrosis. Uncovering this pathway is of high interest as it offers the possibility to target TGF-β with reduced risk of severe side effects occurring in response to direct inhibition.37 We demonstrate a reduction of profibrotic gene expression under ACM conditions in response to ITGAV/B6 inhibition and our pilot study data suggest that a small molecule blocking ITGAV/B6-dependent TGF-β release diminished the generation of fibrosis. Similar approaches using this small molecule or neutralizing antibodies were shown to be protective in murine models of lung, liver, and biliary fibrosis.23,38,39 However, because of the heterogeneity of the phenotype in heterozygous animals, more detailed studies with larger sample sizes are required to further substantiate this finding.\nWhether fibrosis generation is a contributor to the disease or a protective mechanism to ensure integrity of the heart under conditions of compromised adhesion is unclear. In case of the latter, pharmacologic inhibition of fibrosis generation might be detrimental. Nevertheless, fibrosis is a driver of arrhythmia generation,40 which is also supported by our results that ECG abnormalities were reduced by inhibition of fibrosis. Careful studies using different therapeutic approaches to reduce fibrosis in appropriate model organisms need to address this aspect in detail.\nIn conclusion, we established a new mouse model phenocopying many aspects of ACM, uncovered a novel pathway of fibrosis induction, and identified an approach to target this mechanism with future implication as a potential therapeutic option.", "Because of mutations mainly affecting desmosomal genes and evidence of disrupted ICDs, the hypothesis of dysfunctional desmosomes with loss of cell–cell adhesion as a central pathologic step was adopted in the field.2 However, experimental data on this topic are contradictory.6–8 We show that W2 of DSG2 is central for binding and intercellular adhesion in vitro and in vivo. Disrupted desmosomal adhesion is sufficient to induce an ACM phenotype fulfilling the Padua criteria,13 including ventricular fibrosis, depolarization and repolarization abnormalities, arrhythmia, and impaired ventricular function, which are used to establish the diagnosis. In line with this, 3 mutations were described in patients with ACM directly affecting the DSG2 binding mechanism by exchange of the tryptophan with a serine, leucine, or arginine (ClinVar data bank: VCV000199796, VCV000044282, VCV000420241).10 This indicates a disruption of the tryptophan swap independent from the substituting amino acid and supports defective desmosomal adhesion as important factor in ACM.2,24 Moreover, different published DSG2 mutant mouse models show a phenotype resembling the features of DSG2-W2A mutants. These include a DSG2 mutant in which parts of the 2 outermost DSG2 extracellular domains were deleted (but leaving W2A intact),25 2 mouse lines with loss of DSG2,25,26 and mice overexpressing a patient mutation (DSG2-N271S),27 which also showed that ICD structural aberrations precede functional abnormalities and fibrosis generation. These data fuel the hypothesis that disruption and rearrangement of the ICD in response to impaired adhesion between cardiomyocytes is a crucial initial step, at least in case of mutations in desmosomal molecules. Other mechanisms described in patients or translational models, such as aberrant WNT or Hippo/YAP signaling, or immune cell infiltrations,2,28 may be secondary responses and in part even represent adaptive attempts to rescue the functional consequences of such severe structural aberrations.", "Our results suggest the DSG2-W2A model as a valuable tool to study ACM mechanisms. Within the limitations of a murine model, it reproduces a majority of features found in patients with ACM (Table S2). In homozygous animals, both ventricles are affected from early on in life; heterozygous animals develop a milder phenotype, with fibrosis occurring only in the RV. Whether this milder phenotype would extend to the LV over time or whether a gene dose effect underlies the structural differences is unclear. LV and RV remodel differently in response to loading and injury29 and it is possible that the RV is less able to compensate a partial reduction of cardiomyocyte cohesion, in contrast to the LV. A fraction of mut/wt animals did not develop the phenotype at all within the observation period of up to 80 weeks. Thus, the model might help clarify why patients with the same mutation (ie, in the same family) develop the disease with variable penetrance.1 Similar to patients,1 male mutant mice appear to experience more arrhythmia and sudden death than do female mice.\nWe can interpret from the combined histology, echocardiography, and ECG data that the changes in heart function are secondary to fibrosis generation. This is indicated by the notion that functional measures (eg, ejection fraction, QRS interval) worsened over time in homozygous mutants, whereas no increase was detectable for fibrosis. Moreover, functional changes in mut/wt animals were only observed when fibrosis was present. This is in line with a patient cohort with DSP mutations, in which LV fibrosis preceded systolic dysfunction.30 A different study in DSG2-N271S-expressing ex vivo paced hearts demonstrated conduction velocity impairments before onset of fibrosis.27 More longitudinal studies using different mutational backgrounds and sensitive detection methods (eg, cardiac magnetic resonance imaging and ECG under exercise) are necessary to define the role of fibrosis as cause or consequence of functional changes.\nAs with all murine models, the DSG2-W2A line has limitations in its ability to fully recapitulate the human disease phenotype. Mutant mice do not show pronounced replacement with adipose tissue, which is a common characteristic in patient heart samples, but in general is not fully modeled in mice.24 Moreover, at least in ECGs under anesthesia, not all mutant animals develop arrhythmia, even if fibrosis is present. However, in contrast to most other ACM models, this knock-in model has the advantage of a single amino acid exchange under the endogenous promotor, thus reducing the possibility of secondary unwanted effects attributable to complete protein absence or overexpression. As in patients, the mutation is present in all cell types and not limited to cardiomyocytes.2,24", "DSG2-W2A led to a severely altered ICD structure consistent with impaired cytoskeletal attachment and ruptured junctions. Whereas these changes provide explanations for compromised intercellular adhesion, we also noted alterations in cell–matrix protein distribution. RNA sequencing before and after onset of the disease phenotype and a comparison with ACM patient datasets identified ITGB6 as commonly deregulated in patients with ACM and DSG2-W2A mice. Although we noted reduced mRNA levels in mutant hearts, the overall protein content was unaltered. This suggests pronounced posttranslational regulation of ITGB6, which was already demonstrated in skeletal muscle.31 In mutant hearts, ITGB6 was increased at the ICD together with elevated levels of ITGAV and increased heterodimerization of both molecules. By mechanical force exerted through ITGAV/B6, TGF-β is detached from the latency-associated peptide and can induce signaling by means of TGF-β receptors.17,32 In line with higher intracellular forces, talin-2 and vinculin were increased at ICDs, as binding of these molecules activates and stabilizes integrins. A mutual regulation of both adhesive compartments is well-established. As an example, upon loss of N-cadherin, integrins are activated and induce fibronectin deposition.33 A similar mechanism is conceivable in DSG2-W2A hearts, either by loss of DSG2 or because of reduced N-cadherin anchorage. So far, only limited data are available on the role of integrins in ACM. A recent study showed downregulation of integrin-β1D leading to ventricular arrhythmia.34 Furthermore, knockdown of PKP2 in HL-1 cardiomyocytes was described to deregulate focal adhesions, including integrin-α1.35", "The ITGAV/B6 heterodimer is described as one of the major activators of latent TGF-β1 and TGF-β3.17 Although TGF-β signaling is known as general driver of cardiac fibrosis36 and more specifically was implicated in ACM,2 to our knowledge no data are available on the role of ITGAV/B6 and their regulation of TGF-β signaling in cardiac fibrosis. Uncovering this pathway is of high interest as it offers the possibility to target TGF-β with reduced risk of severe side effects occurring in response to direct inhibition.37 We demonstrate a reduction of profibrotic gene expression under ACM conditions in response to ITGAV/B6 inhibition and our pilot study data suggest that a small molecule blocking ITGAV/B6-dependent TGF-β release diminished the generation of fibrosis. Similar approaches using this small molecule or neutralizing antibodies were shown to be protective in murine models of lung, liver, and biliary fibrosis.23,38,39 However, because of the heterogeneity of the phenotype in heterozygous animals, more detailed studies with larger sample sizes are required to further substantiate this finding.\nWhether fibrosis generation is a contributor to the disease or a protective mechanism to ensure integrity of the heart under conditions of compromised adhesion is unclear. In case of the latter, pharmacologic inhibition of fibrosis generation might be detrimental. Nevertheless, fibrosis is a driver of arrhythmia generation,40 which is also supported by our results that ECG abnormalities were reduced by inhibition of fibrosis. Careful studies using different therapeutic approaches to reduce fibrosis in appropriate model organisms need to address this aspect in detail.\nIn conclusion, we established a new mouse model phenocopying many aspects of ACM, uncovered a novel pathway of fibrosis induction, and identified an approach to target this mechanism with future implication as a potential therapeutic option.", "[SUBTITLE] Acknowledgments [SUBSECTION] The authors thank Alain Brühlhart and the team from the Animal Facility; Dr Cinzia Tiberi (Center for Cellular Imaging and NanoAnalytics); Dr Alexia Loynton-Ferrand (Imaging Core Facility); Dr Diego Calabrese (Histology Core Facility); and Drs Mike Abanto and Beat Erne, P. Lorentz, and E. Bartoszek (Microscopy Core Facility), University of Basel, Switzerland; and Dr Christian Beisel and Philippe Demougin, Genomics Facility Basel (D-BSSE, ETH Zürich, and University of Basel), for support; Nicolas Schlegel (Department of Surgery, University Hospital Würzburg, Germany) for providing CaCo2 cells; Nikola Golenhofen (Institute of Anatomy, University of Ulm, Germany) for providing the Fc-His-pEGFP-N3 plasmid; and Anja Fuchs for technical assistance. Calculations were performed at the sciCORE scientific computing center at University of Basel.\nThe authors thank Alain Brühlhart and the team from the Animal Facility; Dr Cinzia Tiberi (Center for Cellular Imaging and NanoAnalytics); Dr Alexia Loynton-Ferrand (Imaging Core Facility); Dr Diego Calabrese (Histology Core Facility); and Drs Mike Abanto and Beat Erne, P. Lorentz, and E. Bartoszek (Microscopy Core Facility), University of Basel, Switzerland; and Dr Christian Beisel and Philippe Demougin, Genomics Facility Basel (D-BSSE, ETH Zürich, and University of Basel), for support; Nicolas Schlegel (Department of Surgery, University Hospital Würzburg, Germany) for providing CaCo2 cells; Nikola Golenhofen (Institute of Anatomy, University of Ulm, Germany) for providing the Fc-His-pEGFP-N3 plasmid; and Anja Fuchs for technical assistance. Calculations were performed at the sciCORE scientific computing center at University of Basel.\n[SUBTITLE] Sources of Funding [SUBSECTION] This project was supported by the German Research Council (SP1300-3/1 to Dr Spindler), the Swiss National Science Foundation (197764 to Dr Spindler), the Swiss Heart Foundation (FF21098 to Drs Schinner and Spindler), the Research Fund Junior Researchers, University of Basel (3BM1079 to Dr Schinner), and the Theiler-Haag Foundation (Dr Spindler). Dr Sheikh is supported by grants from the National Institutes of Health (grant HL142251) and Department of Defense (grant W81XWH1810380).\nThis project was supported by the German Research Council (SP1300-3/1 to Dr Spindler), the Swiss National Science Foundation (197764 to Dr Spindler), the Swiss Heart Foundation (FF21098 to Drs Schinner and Spindler), the Research Fund Junior Researchers, University of Basel (3BM1079 to Dr Schinner), and the Theiler-Haag Foundation (Dr Spindler). Dr Sheikh is supported by grants from the National Institutes of Health (grant HL142251) and Department of Defense (grant W81XWH1810380).\n[SUBTITLE] Disclosures [SUBSECTION] Dr Sheikh is a cofounder and equity shareholder of Papillon Therapeutics Inc and is a consultant and equity shareholder of LEXEO Therapeutics Inc. The other authors have nothing to disclose.\nDr Sheikh is a cofounder and equity shareholder of Papillon Therapeutics Inc and is a consultant and equity shareholder of LEXEO Therapeutics Inc. The other authors have nothing to disclose.\n[SUBTITLE] Supplemental Material [SUBSECTION] Expanded Methods\nTables S1–S3\nFigures S1–S6\nReferences 42–48\nExpanded Methods\nTables S1–S3\nFigures S1–S6\nReferences 42–48", "The authors thank Alain Brühlhart and the team from the Animal Facility; Dr Cinzia Tiberi (Center for Cellular Imaging and NanoAnalytics); Dr Alexia Loynton-Ferrand (Imaging Core Facility); Dr Diego Calabrese (Histology Core Facility); and Drs Mike Abanto and Beat Erne, P. Lorentz, and E. Bartoszek (Microscopy Core Facility), University of Basel, Switzerland; and Dr Christian Beisel and Philippe Demougin, Genomics Facility Basel (D-BSSE, ETH Zürich, and University of Basel), for support; Nicolas Schlegel (Department of Surgery, University Hospital Würzburg, Germany) for providing CaCo2 cells; Nikola Golenhofen (Institute of Anatomy, University of Ulm, Germany) for providing the Fc-His-pEGFP-N3 plasmid; and Anja Fuchs for technical assistance. Calculations were performed at the sciCORE scientific computing center at University of Basel.", "This project was supported by the German Research Council (SP1300-3/1 to Dr Spindler), the Swiss National Science Foundation (197764 to Dr Spindler), the Swiss Heart Foundation (FF21098 to Drs Schinner and Spindler), the Research Fund Junior Researchers, University of Basel (3BM1079 to Dr Schinner), and the Theiler-Haag Foundation (Dr Spindler). Dr Sheikh is supported by grants from the National Institutes of Health (grant HL142251) and Department of Defense (grant W81XWH1810380).", "Dr Sheikh is a cofounder and equity shareholder of Papillon Therapeutics Inc and is a consultant and equity shareholder of LEXEO Therapeutics Inc. The other authors have nothing to disclose.", "Expanded Methods\nTables S1–S3\nFigures S1–S6\nReferences 42–48", "" ]
[ null, null, "methods", null, null, null, "data-availability", "results", null, null, "subjects", null, null, null, null, "discussion", null, null, null, null, null, null, null, "COI-Statement", null, "supplementary-material" ]
[ "cardiomyopathies", "desmosomes", "fibrosis", "integrins", "intercalated disc", "transforming growth factors" ]
End-stage renal disease incidence in a cohort of US firefighters from San Francisco, Chicago, and Philadelphia.
36268894
Firefighters perform strenuous work in hot environments, which may increase their risk of chronic kidney disease. The purpose of this study was to evaluate the risk of end-stage renal disease (ESRD) and types of ESRD among a cohort of US firefighters compared to the US general population, and to examine exposure-response relationships.
BACKGROUND
ESRD from 1977 through 2014 was identified through linkage with Medicare data. ESRD incidence in the cohort compared to the US population was evaluated using life table analyses. Associations of all ESRD, systemic ESRD, hypertensive ESRD, and diabetic ESRD with exposure surrogates (exposed days, fire runs, and fire hours) were examined in Cox proportional hazards models adjusted for attained age (the time scale), race, birth date, fire department, and employment duration.
METHODS
The incidence of all ESRD was less than expected (standardized incidence ratio (SIR) = 0.79; 95% confidence interval = 0.69-0.89, observed = 247). SIRs for ESRD types were not significantly increased. Positive associations of all ESRD, systemic ESRD, and hypertensive ESRD with exposed days were observed: however, 95% confidence intervals included one.
RESULTS
We found little evidence of increased risk of ESRD among this cohort of firefighters. Limitations included the inability to evaluate exposure-response relationships for some ESRD types due to small observed numbers, the limitations of the surrogates of exposure, and the lack of information on more sensitive outcome measures for potential kidney effects.
CONCLUSIONS
[ "Humans", "Aged", "United States", "Firefighters", "Incidence", "Chicago", "Philadelphia", "San Francisco", "Medicare", "Kidney Failure, Chronic" ]
9828160
null
null
METHODS
[SUBTITLE] Cohort description [SUBSECTION] The previous mortality cohort included 29,992 career firefighters employed by the fire departments of Chicago (CFD), Philadelphia (PFD), and San Francisco (SFFD) for at least 1 day between the years 1950 and 2009, as described previously. 32 , 33 The current study cohort includes 26,469 members of the mortality cohort who were alive in 1977, when retrospective ascertainment of incident ESRD began, or later. Firefighters of unknown race (n = 727) were assumed White because 81% of firefighters of known race in the current cohort were White and 70% of firefighters of unknown race were hired before 1970 when minority hiring was lower. The cohort used in the exposure–response analysis, hereafter referred to as the restricted cohort, was further limited to 18,923 male firefighters of known race hired in 1950 or later for at least 1 year. The study received approvals from the Institutional Review Boards of NIOSH and the National Cancer Institute. Informed consent was waived for this records‐based study. The previous mortality cohort included 29,992 career firefighters employed by the fire departments of Chicago (CFD), Philadelphia (PFD), and San Francisco (SFFD) for at least 1 day between the years 1950 and 2009, as described previously. 32 , 33 The current study cohort includes 26,469 members of the mortality cohort who were alive in 1977, when retrospective ascertainment of incident ESRD began, or later. Firefighters of unknown race (n = 727) were assumed White because 81% of firefighters of known race in the current cohort were White and 70% of firefighters of unknown race were hired before 1970 when minority hiring was lower. The cohort used in the exposure–response analysis, hereafter referred to as the restricted cohort, was further limited to 18,923 male firefighters of known race hired in 1950 or later for at least 1 year. The study received approvals from the Institutional Review Boards of NIOSH and the National Cancer Institute. Informed consent was waived for this records‐based study. [SUBTITLE] Exposure assessment [SUBSECTION] Exposure surrogates were calculated for the restricted cohort as described previously. 37 , 38 These surrogates were originally developed to estimate exposure to combustion byproducts of fire for a cancer study, 38 but are used in this study as crude surrogates of occupational heat exposure. Briefly, detailed work histories through 2009 were linked with job exposure matrices based on the job, location, and fire‐fighting apparatus assignments. Data were available to calculate the number of exposed days (i.e., days worked in a job or location with potential exposure) for CFD, PFD, and SFFD firefighters; the number of fire runs for CFD and PFD firefighters; and the number of fire hours (i.e., the time spent at fires) for CFD firefighters. Exposure surrogates were calculated for the restricted cohort as described previously. 37 , 38 These surrogates were originally developed to estimate exposure to combustion byproducts of fire for a cancer study, 38 but are used in this study as crude surrogates of occupational heat exposure. Briefly, detailed work histories through 2009 were linked with job exposure matrices based on the job, location, and fire‐fighting apparatus assignments. Data were available to calculate the number of exposed days (i.e., days worked in a job or location with potential exposure) for CFD, PFD, and SFFD firefighters; the number of fire runs for CFD and PFD firefighters; and the number of fire hours (i.e., the time spent at fires) for CFD firefighters. [SUBTITLE] Ascertainment of vital status and ESRD [SUBSECTION] Vital status was ascertained through 2016 as described previously. 32 , 33 Cohort members with incident ESRD treated with dialysis or transplant from 1977 through 2014 were identified by linking the cohort with Medicare data on persons with ESRD by Social Security number (SSN), name, date of birth, and gender using the Fuzzy Lookup Transformation in Microsoft SQL Server Integration Services. 39 Inexact matches were manually reviewed in descending order of the overall similarity score until they were consistently judged to be false matches. All inexact matches with an exact match on the Social Security number were also manually reviewed. Medicare data on persons with ESRD is maintained by the Centers for Medicare & Medicaid Services (CMS) and includes individuals who received Medicare‐covered renal replacement therapy (dialysis or transplant) in 1977 or later. Since 1973, most persons with ESRD have been entitled to Medicare regardless of age. 40 Between the earliest years, when reporting was incomplete, and 1995, when reporting of all ESRD patients regardless of Medicare eligibility was mandated, 41 approximately 93% of ESRD patients in the United States were included in the data collected. 40 Vital status was ascertained through 2016 as described previously. 32 , 33 Cohort members with incident ESRD treated with dialysis or transplant from 1977 through 2014 were identified by linking the cohort with Medicare data on persons with ESRD by Social Security number (SSN), name, date of birth, and gender using the Fuzzy Lookup Transformation in Microsoft SQL Server Integration Services. 39 Inexact matches were manually reviewed in descending order of the overall similarity score until they were consistently judged to be false matches. All inexact matches with an exact match on the Social Security number were also manually reviewed. Medicare data on persons with ESRD is maintained by the Centers for Medicare & Medicaid Services (CMS) and includes individuals who received Medicare‐covered renal replacement therapy (dialysis or transplant) in 1977 or later. Since 1973, most persons with ESRD have been entitled to Medicare regardless of age. 40 Between the earliest years, when reporting was incomplete, and 1995, when reporting of all ESRD patients regardless of Medicare eligibility was mandated, 41 approximately 93% of ESRD patients in the United States were included in the data collected. 40 [SUBTITLE] Analysis [SUBSECTION] The incidence of ESRD among the overall cohort was compared to that of the US general population in R version 4.1.3 42 using the methods described by Calvert et al. 43 Since the Medicare data on ESRD are incomplete before 1977, cohort members who died before 1977 were excluded from the analyses. Person‐years‐at‐risk (PYAR) were accumulated from the date the cohort criteria were met or January 1, 1977, whichever was later, to the first service date for ESRD, the date of death, the date last observed, or the ending date of the study (December 31, 2014), whichever was earliest. The first service date for ESRD is generally the date on which renal replacement therapy began and was used as a surrogate for the date of onset for ESRD. PYAR were stratified by gender, race (White, other), and 5‐year intervals of age and calendar time and then multiplied by the appropriate US ESRD incidence rates to calculate the expected number of cases for each stratum. The US incidence rates were created from CMS Medicare data and US census data as described elsewhere. 43 The expected number of ESRD cases in each stratum was summed to obtain the total expected number. The standardized incidence ratio (SIR) was calculated as the ratio of the observed to the expected number of treated ESRD cases. SIRs were also calculated for types of ESRD and for the restricted cohort. Ninety‐five percent CIs were computed assuming a Poisson distribution for observed ESRD cases. Types of ESRD and the corresponding International Classification of Diseases, 9th Revision, Clinical Modification codes for the primary cause of renal failure in the CMS data are listed in Supporting Information: Table S1. Exposure–response associations within the restricted cohort were examined with Cox proportional hazards regression using the SAS PHREG procedure. 44 Risk sets comprised all restricted cohort members at risk as of the attained age of the case who also matched the case on race (White, other), birth date (within 5 years), and fire department. Restricted cohort members were followed from the completion of the 1‐year eligibility period or January 1, 1977, whichever was latest. The cumulative exposure metrics were lagged 10 and 20 years because chronic kidney failure generally develops over many years. In categorical models, the cut‐points were selected to obtain an approximately equal number of cases in each exposure quartile. Hazard ratios (HRs) were estimated from the maximum partial likelihood ratio test and two‐sided 95% CIs were based on the profile likelihood. The exposure surrogates were also analyzed as continuous time‐dependent variables in log‐linear models where the HR increases exponentially with exposure or, equivalently, the log of the HR increases linearly with exposure. Because the exposure surrogates were generally right‐skewed and exposure–response relationships in occupational studies are often attenuated at high exposure levels, 45 log‐linear models were also evaluated excluding person‐time in the top decile of exposure. The healthy worker survivor effect was addressed by repeating the analyses also matching on employment duration (<10, 10–<20, 20–<30, and 30+ years). Modeling analyses were restricted to analyses with 30 or more observed cases. The incidence of ESRD among the overall cohort was compared to that of the US general population in R version 4.1.3 42 using the methods described by Calvert et al. 43 Since the Medicare data on ESRD are incomplete before 1977, cohort members who died before 1977 were excluded from the analyses. Person‐years‐at‐risk (PYAR) were accumulated from the date the cohort criteria were met or January 1, 1977, whichever was later, to the first service date for ESRD, the date of death, the date last observed, or the ending date of the study (December 31, 2014), whichever was earliest. The first service date for ESRD is generally the date on which renal replacement therapy began and was used as a surrogate for the date of onset for ESRD. PYAR were stratified by gender, race (White, other), and 5‐year intervals of age and calendar time and then multiplied by the appropriate US ESRD incidence rates to calculate the expected number of cases for each stratum. The US incidence rates were created from CMS Medicare data and US census data as described elsewhere. 43 The expected number of ESRD cases in each stratum was summed to obtain the total expected number. The standardized incidence ratio (SIR) was calculated as the ratio of the observed to the expected number of treated ESRD cases. SIRs were also calculated for types of ESRD and for the restricted cohort. Ninety‐five percent CIs were computed assuming a Poisson distribution for observed ESRD cases. Types of ESRD and the corresponding International Classification of Diseases, 9th Revision, Clinical Modification codes for the primary cause of renal failure in the CMS data are listed in Supporting Information: Table S1. Exposure–response associations within the restricted cohort were examined with Cox proportional hazards regression using the SAS PHREG procedure. 44 Risk sets comprised all restricted cohort members at risk as of the attained age of the case who also matched the case on race (White, other), birth date (within 5 years), and fire department. Restricted cohort members were followed from the completion of the 1‐year eligibility period or January 1, 1977, whichever was latest. The cumulative exposure metrics were lagged 10 and 20 years because chronic kidney failure generally develops over many years. In categorical models, the cut‐points were selected to obtain an approximately equal number of cases in each exposure quartile. Hazard ratios (HRs) were estimated from the maximum partial likelihood ratio test and two‐sided 95% CIs were based on the profile likelihood. The exposure surrogates were also analyzed as continuous time‐dependent variables in log‐linear models where the HR increases exponentially with exposure or, equivalently, the log of the HR increases linearly with exposure. Because the exposure surrogates were generally right‐skewed and exposure–response relationships in occupational studies are often attenuated at high exposure levels, 45 log‐linear models were also evaluated excluding person‐time in the top decile of exposure. The healthy worker survivor effect was addressed by repeating the analyses also matching on employment duration (<10, 10–<20, 20–<30, and 30+ years). Modeling analyses were restricted to analyses with 30 or more observed cases.
RESULTS
Table 1 shows the demographic characteristics of the full and restricted cohorts. Most members of the full cohort were male (96.3%) and White (78.6%). Over three‐fourths (78.1%) of the restricted cohort of male firefighters were also White. The mean employment duration for both cohorts was 21 years. At the end of the follow‐up, 38.2% of the full cohort and 25.3% of the restricted cohort were deceased. Characteristics of the full and restricted cohorts Abbreviation: IQR, interquartile range. Male firefighters of a known race who were hired in 1950 or later and employed for at least 1 year. Vital status at the end of follow‐up (December 31, 2014) or the date of diagnosis, whichever was earlier. Cell size <11 or reporting the exact number allows a cell size <11 to be derived. As shown in Table 2, the incidence of all ESRD was less than expected among the full (SIR = 0.79; 95% CI = 0.69–0.89; observed = 247) and restricted (SIR: 0.73; 95% CI = 0.63–0.85; observed = 177) cohorts. The median age at the time of onset was 69 years (interquartile range = 61–77) and 67 years (interquartile range: 59–73) among the full and restricted cohorts, respectively. SIRs for ESRD due to metabolic disease (full cohort: 2.84; 95% CI = 0.76–7.26; restricted cohort: 1.92; 95% CI = 0.22–6.94) and ESRD due to collagen vascular disease (full cohort: 1.18; 95% CI = 0.32–3.01; restricted cohort: 1.17; 95% CI = 0.24–3.42) were elevated, although the CIs for these ESRD types were wide and included one. The incidence of systemic ESRD and diabetic ESRD was less than expected among the full (systemic ESRD: SIR = 0.76; 95% CI = 0.65–0.88; diabetic ESRD: SIR = 0.66; 95% CI = 0.52–0.82) and restricted (systemic ESRD: SIR = 0.71; 95% CI = 059–0.84; diabetic ESRD: SIR = 0.63; 95% CI = 0.49–0.81) cohorts. The incidence of ESRD caused by glomerulonephritis was also less than expected among the full cohort (SIR = 0.61; 95% CI = 0.35–0.97). The SIR for ESRD caused by glomerulonephritis among the restricted cohort was 0.66 (95% CI = 0.36–1.10). SIRs for ESRD among the full and restricted cohorts, 1977–2014 Abbreviations: AIDS, acquired immunodeficiency syndrome; CI, confidence interval; ESRD, end‐stage renal disease; NR, not reported (cell size <11 or reporting the cell size allows numbers <11 to be derived); Obs, observed; SIR, standardized incidence ratio. ESRD due to AIDS, sickle cell disease, and hereditary nephropathy were omitted because no cases were observed. The results of internal analyses among the restricted cohort using categorical models are shown in Table 3 (20‐year lag) and Supporting Information: Table S2 (10‐year lag). Results reported herein were obtained from the analyses with matching on employment duration, unless stated otherwise. HRs for all ESRD obtained from categorical models increased in the second and third quartiles of exposed days, but then decreased in the top quartile. HRs for systemic ESRD increased with each exposed‐day quartile in 20‐ and 10‐year lagged analyses. HRs for hypertensive ESRD also increased with the exposed‐day quartile in 20‐ and 10‐year lagged analyses with an HR of 4.51 (95% CI = 0.97–20.9) in the top quartile in 20‐year lagged analyses. HRs for diabetic ESRD increased with exposed‐day quartile in 10‐year lagged analyses only. ESRD HRs by 20‐year lagged exposure surrogates obtained from categorical models a Abbreviations: CFD, Chicago fire department; CI, confidence interval; dept, fire departments included in the analysis; ESRD, end‐stage renal disease; HR, hazard ratio; PFD, Philadelphia fire department; SFFD, San Francisco fire department. All models were adjusted for age (time scale) and for race (White/non‐White), date of birth (within 5 years), and fire department by matching. Some models were also adjusted for employment duration (<10, 10–<20, 20–<30, and 30+ years) by matching. All modeling analyses were conducted among the restricted cohort and included 30 or more observed ESRD cases with an approximately equal number of cases in each exposure quartile. The overall numbers of ESRD cases in the analyses of exposed days were 177 for all ESRD, 132 for systemic ESRD, 63 for diabetic ESRD, and 53 for hypertensive ESRD. The results of internal analyses among the restricted cohort using log‐linear models are shown in Table 4 (20‐year lag) and Supporting Information: Table S3 (10‐year lag). The HR for all ESRD was 1.11 (95% CI = 0.99–1.25) and 1.08 (95% CI = 0.98–1.18) per 1000 exposed days in 20‐ and 10‐year lagged analyses, respectively, when person‐time in the top exposure decile was excluded. In analogous linear models including person‐time in the top decile of exposure, the HR for all ESRD was 1.05 (95% CI = 0.96–1.16) and 1.07 (0.99–1.16) per 1000 exposed days in 20‐ and 10‐year lagged analyses, respectively. ESRD HRs by 20‐year lagged exposure surrogates obtained from log‐linear models a Abbreviations: CI, confidence interval; ESRD, end‐stage renal disease; HR, hazard ratio. All models were adjusted for age (time scale) and for race (White/non‐White), date of birth (within 5 years), and fire department by matching. Some models were also adjusted for employment duration (<10, 10–<20, 20–<30, and 30+ years) by matching. All modeling analyses were conducted among the restricted cohort and included 30 or more observed ESRD cases. The overall numbers of ESRD cases in analyses of exposed days were 177 for all ESRD, 132 for systemic ESRD, 63 for diabetic ESRD, and 53 for hypertensive ESRD. Exposed days were available for Chicago, Philadelphia, and San Francisco firefighters; fire runs for Chicago and Philadelphia firefighters; and fire hours for Chicago firefighters. 1000 exposed days, 1000 fire runs, or 1000 fire hours. Systemic ESRD HRs per 1000 exposed days were 1.05 (95% CI = 0.95–1.18) and 1.07 (95% CI = 0.98–1.17) in 20‐ and 10‐year lagged analyses, respectively, when person‐time in the top decile of exposure was included and 1.07 (95% CI = 0.95–1.23) and 1.05 (95% CI = 0.95–1.17) in 20‐ and 10‐year lagged analyses, respectively, when person‐time in the top decile of exposure was excluded. Hypertensive ESRD HRs per 1000 exposed days were 1.09 (95% CI = 0.93–1.29) and 1.10 (95% CI = 0.96–1.27) in 20‐ and 10‐year lagged analyses, respectively. When person‐time in the top exposure decile was excluded, the hypertensive ESRD HRs per 1000 exposed days were 1.13 (95% CI = 0.93–1.40) and 1.09 (95% CI = 0.94–1.29) in 20‐ and 10‐year lagged analyses, respectively. In 10‐year lagged analyses, the diabetic ESRD HR per 1000 exposed days was 1.03 (95% CI = 0.92–1.17) and 1.03 (95% CI = 0.90–1.19), including and excluding person‐time in the top exposure decile, respectively.
null
null
[ "INTRODUCTION", "Cohort description", "Exposure assessment", "Ascertainment of vital status and ESRD", "Analysis", "AUTHOR CONTRIBUTIONS", "DISCLOSURE BY AJIM EDITOR OF RECORD", "ETHICS APPROVAL AND INFORMED CONSENT", "DISCLAIMER" ]
[ "Firefighters may be at increased risk of developing chronic kidney disease because they perform strenuous work in hot environments. Strenuous work with heat stress and dehydration may cause renal impairment from clinical or subclinical rhabdomyolysis, hyperuricemia and urate crystalluria, and hyperosmolality‐induced release of vasopressin and activation of the aldose reductase‐fructokinase pathway.\n1\n, \n2\n, \n3\n, \n4\n, \n5\n, \n6\n, \n7\n, \n8\n, \n9\n, \n10\n, \n11\n Increased concern about these potential heat‐related effects on kidney function has arisen due to an epidemic of chronic kidney disease among sugarcane workers in Central America.\n12\n Chronic kidney disease in these workers has often progressed to end‐stage renal disease (ESRD).\n3\n, \n13\n Chronic kidney disease has also been observed among other heat‐exposed workers and in other regions.\n4\n, \n12\n, \n14\n, \n15\n, \n16\n, \n17\n However, few data exist on the risk of kidney disease among firefighters.\nFirefighters are at risk of heat stress from exposure to heat from fires, ambient heat during hot weather, and metabolic heat generated by strenuous physical work.\n18\n, \n19\n The heavy, impermeable personal protective equipment structural firefighters wear adds to their metabolic workload and impairs evaporative cooling.\n19\n Increased core body temperature and heat‐related illnesses, including rhabdomyolysis and fatal heat stroke, have been documented among firefighters and trainees.\n20\n, \n21\n, \n22\n, \n23\n, \n24\n, \n25\n, \n26\n, \n27\n, \n28\n Firefighters are also at risk of dehydration,\n29\n which can contribute to kidney injury.\n30\n, \n31\n\n\nWe previously evaluated mortality and cancer incidence among a large National Institute for Occupational Safety and Health (NIOSH) cohort of US firefighters from the Chicago, Philadelphia, and San Francisco fire departments and found an increased risk of several cancers including kidney cancer compared to the US general population.\n32\n, \n33\n Mortality from chronic and unspecified nephritis and renal failure and other renal sclerosis, based on the underlying cause of death, was elevated among Philadelphia firefighters (standardized mortality ratio = 1.35; 95% confidence interval [CI] = 1.03–1.73) but not Chicago or San Francisco firefighters.\n33\n However, chronic kidney disease is usually not the underlying cause of death even when mentioned on the death certificate.\n34\n In this study, we evaluate ESRD incidence in the NIOSH firefighter cohort compared to the general population and relationships of ESRD with surrogates of exposure. Chicago and Philadelphia have humid continental climates, whereas San Francisco has a Mediterranean climate.\n35\n From 1981 to 2010, the average annual number of days the temperature reached 32.2°C was 17 in Chicago, 25 in Philadelphia, and 2 in San Francisco.\n36\n\n", "The previous mortality cohort included 29,992 career firefighters employed by the fire departments of Chicago (CFD), Philadelphia (PFD), and San Francisco (SFFD) for at least 1 day between the years 1950 and 2009, as described previously.\n32\n, \n33\n The current study cohort includes 26,469 members of the mortality cohort who were alive in 1977, when retrospective ascertainment of incident ESRD began, or later. Firefighters of unknown race (n = 727) were assumed White because 81% of firefighters of known race in the current cohort were White and 70% of firefighters of unknown race were hired before 1970 when minority hiring was lower. The cohort used in the exposure–response analysis, hereafter referred to as the restricted cohort, was further limited to 18,923 male firefighters of known race hired in 1950 or later for at least 1 year.\nThe study received approvals from the Institutional Review Boards of NIOSH and the National Cancer Institute. Informed consent was waived for this records‐based study.", "Exposure surrogates were calculated for the restricted cohort as described previously.\n37\n, \n38\n These surrogates were originally developed to estimate exposure to combustion byproducts of fire for a cancer study,\n38\n but are used in this study as crude surrogates of occupational heat exposure. Briefly, detailed work histories through 2009 were linked with job exposure matrices based on the job, location, and fire‐fighting apparatus assignments. Data were available to calculate the number of exposed days (i.e., days worked in a job or location with potential exposure) for CFD, PFD, and SFFD firefighters; the number of fire runs for CFD and PFD firefighters; and the number of fire hours (i.e., the time spent at fires) for CFD firefighters.", "Vital status was ascertained through 2016 as described previously.\n32\n, \n33\n Cohort members with incident ESRD treated with dialysis or transplant from 1977 through 2014 were identified by linking the cohort with Medicare data on persons with ESRD by Social Security number (SSN), name, date of birth, and gender using the Fuzzy Lookup Transformation in Microsoft SQL Server Integration Services.\n39\n Inexact matches were manually reviewed in descending order of the overall similarity score until they were consistently judged to be false matches. All inexact matches with an exact match on the Social Security number were also manually reviewed.\nMedicare data on persons with ESRD is maintained by the Centers for Medicare & Medicaid Services (CMS) and includes individuals who received Medicare‐covered renal replacement therapy (dialysis or transplant) in 1977 or later. Since 1973, most persons with ESRD have been entitled to Medicare regardless of age.\n40\n Between the earliest years, when reporting was incomplete, and 1995, when reporting of all ESRD patients regardless of Medicare eligibility was mandated,\n41\n approximately 93% of ESRD patients in the United States were included in the data collected.\n40\n\n", "The incidence of ESRD among the overall cohort was compared to that of the US general population in R version 4.1.3\n42\n using the methods described by Calvert et al.\n43\n Since the Medicare data on ESRD are incomplete before 1977, cohort members who died before 1977 were excluded from the analyses. Person‐years‐at‐risk (PYAR) were accumulated from the date the cohort criteria were met or January 1, 1977, whichever was later, to the first service date for ESRD, the date of death, the date last observed, or the ending date of the study (December 31, 2014), whichever was earliest. The first service date for ESRD is generally the date on which renal replacement therapy began and was used as a surrogate for the date of onset for ESRD. PYAR were stratified by gender, race (White, other), and 5‐year intervals of age and calendar time and then multiplied by the appropriate US ESRD incidence rates to calculate the expected number of cases for each stratum. The US incidence rates were created from CMS Medicare data and US census data as described elsewhere.\n43\n The expected number of ESRD cases in each stratum was summed to obtain the total expected number. The standardized incidence ratio (SIR) was calculated as the ratio of the observed to the expected number of treated ESRD cases. SIRs were also calculated for types of ESRD and for the restricted cohort. Ninety‐five percent CIs were computed assuming a Poisson distribution for observed ESRD cases. Types of ESRD and the corresponding International Classification of Diseases, 9th Revision, Clinical Modification codes for the primary cause of renal failure in the CMS data are listed in Supporting Information: Table S1.\nExposure–response associations within the restricted cohort were examined with Cox proportional hazards regression using the SAS PHREG procedure.\n44\n Risk sets comprised all restricted cohort members at risk as of the attained age of the case who also matched the case on race (White, other), birth date (within 5 years), and fire department. Restricted cohort members were followed from the completion of the 1‐year eligibility period or January 1, 1977, whichever was latest. The cumulative exposure metrics were lagged 10 and 20 years because chronic kidney failure generally develops over many years. In categorical models, the cut‐points were selected to obtain an approximately equal number of cases in each exposure quartile. Hazard ratios (HRs) were estimated from the maximum partial likelihood ratio test and two‐sided 95% CIs were based on the profile likelihood. The exposure surrogates were also analyzed as continuous time‐dependent variables in log‐linear models where the HR increases exponentially with exposure or, equivalently, the log of the HR increases linearly with exposure. Because the exposure surrogates were generally right‐skewed and exposure–response relationships in occupational studies are often attenuated at high exposure levels,\n45\n log‐linear models were also evaluated excluding person‐time in the top decile of exposure. The healthy worker survivor effect was addressed by repeating the analyses also matching on employment duration (<10, 10–<20, 20–<30, and 30+ years). Modeling analyses were restricted to analyses with 30 or more observed cases.", "Lynne E. Pinkerton conceived the study and wrote the first draft of the manuscript. Stephen Bertke conducted the analysis. Travis L. Kubale, James H. Yiin, and Robert D. Daniels acquired the data and assembled the cohort. Matthew M. Dahm led the assessment of the exposure surrogates. All authors participated in the interpretation and presentation of results, approved the final manuscript, and agreed to be accountable for all aspects of the work.", "John Meyer declares that he has no conflict of interest in the review and publication decision regarding this article.", "The study received approvals from the Institutional Review Boards of the National Institute for Occupational Safety and Health and the National Cancer Institute. Informed consent was waived for this records‐based study.", "The findings and conclusions in this report are those of the authors and do not necessarily represent the official position of the National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention." ]
[ null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "METHODS", "Cohort description", "Exposure assessment", "Ascertainment of vital status and ESRD", "Analysis", "RESULTS", "DISCUSSION", "AUTHOR CONTRIBUTIONS", "CONFLICTS OF INTEREST", "DISCLOSURE BY AJIM EDITOR OF RECORD", "ETHICS APPROVAL AND INFORMED CONSENT", "DISCLAIMER", "Supporting information" ]
[ "Firefighters may be at increased risk of developing chronic kidney disease because they perform strenuous work in hot environments. Strenuous work with heat stress and dehydration may cause renal impairment from clinical or subclinical rhabdomyolysis, hyperuricemia and urate crystalluria, and hyperosmolality‐induced release of vasopressin and activation of the aldose reductase‐fructokinase pathway.\n1\n, \n2\n, \n3\n, \n4\n, \n5\n, \n6\n, \n7\n, \n8\n, \n9\n, \n10\n, \n11\n Increased concern about these potential heat‐related effects on kidney function has arisen due to an epidemic of chronic kidney disease among sugarcane workers in Central America.\n12\n Chronic kidney disease in these workers has often progressed to end‐stage renal disease (ESRD).\n3\n, \n13\n Chronic kidney disease has also been observed among other heat‐exposed workers and in other regions.\n4\n, \n12\n, \n14\n, \n15\n, \n16\n, \n17\n However, few data exist on the risk of kidney disease among firefighters.\nFirefighters are at risk of heat stress from exposure to heat from fires, ambient heat during hot weather, and metabolic heat generated by strenuous physical work.\n18\n, \n19\n The heavy, impermeable personal protective equipment structural firefighters wear adds to their metabolic workload and impairs evaporative cooling.\n19\n Increased core body temperature and heat‐related illnesses, including rhabdomyolysis and fatal heat stroke, have been documented among firefighters and trainees.\n20\n, \n21\n, \n22\n, \n23\n, \n24\n, \n25\n, \n26\n, \n27\n, \n28\n Firefighters are also at risk of dehydration,\n29\n which can contribute to kidney injury.\n30\n, \n31\n\n\nWe previously evaluated mortality and cancer incidence among a large National Institute for Occupational Safety and Health (NIOSH) cohort of US firefighters from the Chicago, Philadelphia, and San Francisco fire departments and found an increased risk of several cancers including kidney cancer compared to the US general population.\n32\n, \n33\n Mortality from chronic and unspecified nephritis and renal failure and other renal sclerosis, based on the underlying cause of death, was elevated among Philadelphia firefighters (standardized mortality ratio = 1.35; 95% confidence interval [CI] = 1.03–1.73) but not Chicago or San Francisco firefighters.\n33\n However, chronic kidney disease is usually not the underlying cause of death even when mentioned on the death certificate.\n34\n In this study, we evaluate ESRD incidence in the NIOSH firefighter cohort compared to the general population and relationships of ESRD with surrogates of exposure. Chicago and Philadelphia have humid continental climates, whereas San Francisco has a Mediterranean climate.\n35\n From 1981 to 2010, the average annual number of days the temperature reached 32.2°C was 17 in Chicago, 25 in Philadelphia, and 2 in San Francisco.\n36\n\n", "[SUBTITLE] Cohort description [SUBSECTION] The previous mortality cohort included 29,992 career firefighters employed by the fire departments of Chicago (CFD), Philadelphia (PFD), and San Francisco (SFFD) for at least 1 day between the years 1950 and 2009, as described previously.\n32\n, \n33\n The current study cohort includes 26,469 members of the mortality cohort who were alive in 1977, when retrospective ascertainment of incident ESRD began, or later. Firefighters of unknown race (n = 727) were assumed White because 81% of firefighters of known race in the current cohort were White and 70% of firefighters of unknown race were hired before 1970 when minority hiring was lower. The cohort used in the exposure–response analysis, hereafter referred to as the restricted cohort, was further limited to 18,923 male firefighters of known race hired in 1950 or later for at least 1 year.\nThe study received approvals from the Institutional Review Boards of NIOSH and the National Cancer Institute. Informed consent was waived for this records‐based study.\nThe previous mortality cohort included 29,992 career firefighters employed by the fire departments of Chicago (CFD), Philadelphia (PFD), and San Francisco (SFFD) for at least 1 day between the years 1950 and 2009, as described previously.\n32\n, \n33\n The current study cohort includes 26,469 members of the mortality cohort who were alive in 1977, when retrospective ascertainment of incident ESRD began, or later. Firefighters of unknown race (n = 727) were assumed White because 81% of firefighters of known race in the current cohort were White and 70% of firefighters of unknown race were hired before 1970 when minority hiring was lower. The cohort used in the exposure–response analysis, hereafter referred to as the restricted cohort, was further limited to 18,923 male firefighters of known race hired in 1950 or later for at least 1 year.\nThe study received approvals from the Institutional Review Boards of NIOSH and the National Cancer Institute. Informed consent was waived for this records‐based study.\n[SUBTITLE] Exposure assessment [SUBSECTION] Exposure surrogates were calculated for the restricted cohort as described previously.\n37\n, \n38\n These surrogates were originally developed to estimate exposure to combustion byproducts of fire for a cancer study,\n38\n but are used in this study as crude surrogates of occupational heat exposure. Briefly, detailed work histories through 2009 were linked with job exposure matrices based on the job, location, and fire‐fighting apparatus assignments. Data were available to calculate the number of exposed days (i.e., days worked in a job or location with potential exposure) for CFD, PFD, and SFFD firefighters; the number of fire runs for CFD and PFD firefighters; and the number of fire hours (i.e., the time spent at fires) for CFD firefighters.\nExposure surrogates were calculated for the restricted cohort as described previously.\n37\n, \n38\n These surrogates were originally developed to estimate exposure to combustion byproducts of fire for a cancer study,\n38\n but are used in this study as crude surrogates of occupational heat exposure. Briefly, detailed work histories through 2009 were linked with job exposure matrices based on the job, location, and fire‐fighting apparatus assignments. Data were available to calculate the number of exposed days (i.e., days worked in a job or location with potential exposure) for CFD, PFD, and SFFD firefighters; the number of fire runs for CFD and PFD firefighters; and the number of fire hours (i.e., the time spent at fires) for CFD firefighters.\n[SUBTITLE] Ascertainment of vital status and ESRD [SUBSECTION] Vital status was ascertained through 2016 as described previously.\n32\n, \n33\n Cohort members with incident ESRD treated with dialysis or transplant from 1977 through 2014 were identified by linking the cohort with Medicare data on persons with ESRD by Social Security number (SSN), name, date of birth, and gender using the Fuzzy Lookup Transformation in Microsoft SQL Server Integration Services.\n39\n Inexact matches were manually reviewed in descending order of the overall similarity score until they were consistently judged to be false matches. All inexact matches with an exact match on the Social Security number were also manually reviewed.\nMedicare data on persons with ESRD is maintained by the Centers for Medicare & Medicaid Services (CMS) and includes individuals who received Medicare‐covered renal replacement therapy (dialysis or transplant) in 1977 or later. Since 1973, most persons with ESRD have been entitled to Medicare regardless of age.\n40\n Between the earliest years, when reporting was incomplete, and 1995, when reporting of all ESRD patients regardless of Medicare eligibility was mandated,\n41\n approximately 93% of ESRD patients in the United States were included in the data collected.\n40\n\n\nVital status was ascertained through 2016 as described previously.\n32\n, \n33\n Cohort members with incident ESRD treated with dialysis or transplant from 1977 through 2014 were identified by linking the cohort with Medicare data on persons with ESRD by Social Security number (SSN), name, date of birth, and gender using the Fuzzy Lookup Transformation in Microsoft SQL Server Integration Services.\n39\n Inexact matches were manually reviewed in descending order of the overall similarity score until they were consistently judged to be false matches. All inexact matches with an exact match on the Social Security number were also manually reviewed.\nMedicare data on persons with ESRD is maintained by the Centers for Medicare & Medicaid Services (CMS) and includes individuals who received Medicare‐covered renal replacement therapy (dialysis or transplant) in 1977 or later. Since 1973, most persons with ESRD have been entitled to Medicare regardless of age.\n40\n Between the earliest years, when reporting was incomplete, and 1995, when reporting of all ESRD patients regardless of Medicare eligibility was mandated,\n41\n approximately 93% of ESRD patients in the United States were included in the data collected.\n40\n\n\n[SUBTITLE] Analysis [SUBSECTION] The incidence of ESRD among the overall cohort was compared to that of the US general population in R version 4.1.3\n42\n using the methods described by Calvert et al.\n43\n Since the Medicare data on ESRD are incomplete before 1977, cohort members who died before 1977 were excluded from the analyses. Person‐years‐at‐risk (PYAR) were accumulated from the date the cohort criteria were met or January 1, 1977, whichever was later, to the first service date for ESRD, the date of death, the date last observed, or the ending date of the study (December 31, 2014), whichever was earliest. The first service date for ESRD is generally the date on which renal replacement therapy began and was used as a surrogate for the date of onset for ESRD. PYAR were stratified by gender, race (White, other), and 5‐year intervals of age and calendar time and then multiplied by the appropriate US ESRD incidence rates to calculate the expected number of cases for each stratum. The US incidence rates were created from CMS Medicare data and US census data as described elsewhere.\n43\n The expected number of ESRD cases in each stratum was summed to obtain the total expected number. The standardized incidence ratio (SIR) was calculated as the ratio of the observed to the expected number of treated ESRD cases. SIRs were also calculated for types of ESRD and for the restricted cohort. Ninety‐five percent CIs were computed assuming a Poisson distribution for observed ESRD cases. Types of ESRD and the corresponding International Classification of Diseases, 9th Revision, Clinical Modification codes for the primary cause of renal failure in the CMS data are listed in Supporting Information: Table S1.\nExposure–response associations within the restricted cohort were examined with Cox proportional hazards regression using the SAS PHREG procedure.\n44\n Risk sets comprised all restricted cohort members at risk as of the attained age of the case who also matched the case on race (White, other), birth date (within 5 years), and fire department. Restricted cohort members were followed from the completion of the 1‐year eligibility period or January 1, 1977, whichever was latest. The cumulative exposure metrics were lagged 10 and 20 years because chronic kidney failure generally develops over many years. In categorical models, the cut‐points were selected to obtain an approximately equal number of cases in each exposure quartile. Hazard ratios (HRs) were estimated from the maximum partial likelihood ratio test and two‐sided 95% CIs were based on the profile likelihood. The exposure surrogates were also analyzed as continuous time‐dependent variables in log‐linear models where the HR increases exponentially with exposure or, equivalently, the log of the HR increases linearly with exposure. Because the exposure surrogates were generally right‐skewed and exposure–response relationships in occupational studies are often attenuated at high exposure levels,\n45\n log‐linear models were also evaluated excluding person‐time in the top decile of exposure. The healthy worker survivor effect was addressed by repeating the analyses also matching on employment duration (<10, 10–<20, 20–<30, and 30+ years). Modeling analyses were restricted to analyses with 30 or more observed cases.\nThe incidence of ESRD among the overall cohort was compared to that of the US general population in R version 4.1.3\n42\n using the methods described by Calvert et al.\n43\n Since the Medicare data on ESRD are incomplete before 1977, cohort members who died before 1977 were excluded from the analyses. Person‐years‐at‐risk (PYAR) were accumulated from the date the cohort criteria were met or January 1, 1977, whichever was later, to the first service date for ESRD, the date of death, the date last observed, or the ending date of the study (December 31, 2014), whichever was earliest. The first service date for ESRD is generally the date on which renal replacement therapy began and was used as a surrogate for the date of onset for ESRD. PYAR were stratified by gender, race (White, other), and 5‐year intervals of age and calendar time and then multiplied by the appropriate US ESRD incidence rates to calculate the expected number of cases for each stratum. The US incidence rates were created from CMS Medicare data and US census data as described elsewhere.\n43\n The expected number of ESRD cases in each stratum was summed to obtain the total expected number. The standardized incidence ratio (SIR) was calculated as the ratio of the observed to the expected number of treated ESRD cases. SIRs were also calculated for types of ESRD and for the restricted cohort. Ninety‐five percent CIs were computed assuming a Poisson distribution for observed ESRD cases. Types of ESRD and the corresponding International Classification of Diseases, 9th Revision, Clinical Modification codes for the primary cause of renal failure in the CMS data are listed in Supporting Information: Table S1.\nExposure–response associations within the restricted cohort were examined with Cox proportional hazards regression using the SAS PHREG procedure.\n44\n Risk sets comprised all restricted cohort members at risk as of the attained age of the case who also matched the case on race (White, other), birth date (within 5 years), and fire department. Restricted cohort members were followed from the completion of the 1‐year eligibility period or January 1, 1977, whichever was latest. The cumulative exposure metrics were lagged 10 and 20 years because chronic kidney failure generally develops over many years. In categorical models, the cut‐points were selected to obtain an approximately equal number of cases in each exposure quartile. Hazard ratios (HRs) were estimated from the maximum partial likelihood ratio test and two‐sided 95% CIs were based on the profile likelihood. The exposure surrogates were also analyzed as continuous time‐dependent variables in log‐linear models where the HR increases exponentially with exposure or, equivalently, the log of the HR increases linearly with exposure. Because the exposure surrogates were generally right‐skewed and exposure–response relationships in occupational studies are often attenuated at high exposure levels,\n45\n log‐linear models were also evaluated excluding person‐time in the top decile of exposure. The healthy worker survivor effect was addressed by repeating the analyses also matching on employment duration (<10, 10–<20, 20–<30, and 30+ years). Modeling analyses were restricted to analyses with 30 or more observed cases.", "The previous mortality cohort included 29,992 career firefighters employed by the fire departments of Chicago (CFD), Philadelphia (PFD), and San Francisco (SFFD) for at least 1 day between the years 1950 and 2009, as described previously.\n32\n, \n33\n The current study cohort includes 26,469 members of the mortality cohort who were alive in 1977, when retrospective ascertainment of incident ESRD began, or later. Firefighters of unknown race (n = 727) were assumed White because 81% of firefighters of known race in the current cohort were White and 70% of firefighters of unknown race were hired before 1970 when minority hiring was lower. The cohort used in the exposure–response analysis, hereafter referred to as the restricted cohort, was further limited to 18,923 male firefighters of known race hired in 1950 or later for at least 1 year.\nThe study received approvals from the Institutional Review Boards of NIOSH and the National Cancer Institute. Informed consent was waived for this records‐based study.", "Exposure surrogates were calculated for the restricted cohort as described previously.\n37\n, \n38\n These surrogates were originally developed to estimate exposure to combustion byproducts of fire for a cancer study,\n38\n but are used in this study as crude surrogates of occupational heat exposure. Briefly, detailed work histories through 2009 were linked with job exposure matrices based on the job, location, and fire‐fighting apparatus assignments. Data were available to calculate the number of exposed days (i.e., days worked in a job or location with potential exposure) for CFD, PFD, and SFFD firefighters; the number of fire runs for CFD and PFD firefighters; and the number of fire hours (i.e., the time spent at fires) for CFD firefighters.", "Vital status was ascertained through 2016 as described previously.\n32\n, \n33\n Cohort members with incident ESRD treated with dialysis or transplant from 1977 through 2014 were identified by linking the cohort with Medicare data on persons with ESRD by Social Security number (SSN), name, date of birth, and gender using the Fuzzy Lookup Transformation in Microsoft SQL Server Integration Services.\n39\n Inexact matches were manually reviewed in descending order of the overall similarity score until they were consistently judged to be false matches. All inexact matches with an exact match on the Social Security number were also manually reviewed.\nMedicare data on persons with ESRD is maintained by the Centers for Medicare & Medicaid Services (CMS) and includes individuals who received Medicare‐covered renal replacement therapy (dialysis or transplant) in 1977 or later. Since 1973, most persons with ESRD have been entitled to Medicare regardless of age.\n40\n Between the earliest years, when reporting was incomplete, and 1995, when reporting of all ESRD patients regardless of Medicare eligibility was mandated,\n41\n approximately 93% of ESRD patients in the United States were included in the data collected.\n40\n\n", "The incidence of ESRD among the overall cohort was compared to that of the US general population in R version 4.1.3\n42\n using the methods described by Calvert et al.\n43\n Since the Medicare data on ESRD are incomplete before 1977, cohort members who died before 1977 were excluded from the analyses. Person‐years‐at‐risk (PYAR) were accumulated from the date the cohort criteria were met or January 1, 1977, whichever was later, to the first service date for ESRD, the date of death, the date last observed, or the ending date of the study (December 31, 2014), whichever was earliest. The first service date for ESRD is generally the date on which renal replacement therapy began and was used as a surrogate for the date of onset for ESRD. PYAR were stratified by gender, race (White, other), and 5‐year intervals of age and calendar time and then multiplied by the appropriate US ESRD incidence rates to calculate the expected number of cases for each stratum. The US incidence rates were created from CMS Medicare data and US census data as described elsewhere.\n43\n The expected number of ESRD cases in each stratum was summed to obtain the total expected number. The standardized incidence ratio (SIR) was calculated as the ratio of the observed to the expected number of treated ESRD cases. SIRs were also calculated for types of ESRD and for the restricted cohort. Ninety‐five percent CIs were computed assuming a Poisson distribution for observed ESRD cases. Types of ESRD and the corresponding International Classification of Diseases, 9th Revision, Clinical Modification codes for the primary cause of renal failure in the CMS data are listed in Supporting Information: Table S1.\nExposure–response associations within the restricted cohort were examined with Cox proportional hazards regression using the SAS PHREG procedure.\n44\n Risk sets comprised all restricted cohort members at risk as of the attained age of the case who also matched the case on race (White, other), birth date (within 5 years), and fire department. Restricted cohort members were followed from the completion of the 1‐year eligibility period or January 1, 1977, whichever was latest. The cumulative exposure metrics were lagged 10 and 20 years because chronic kidney failure generally develops over many years. In categorical models, the cut‐points were selected to obtain an approximately equal number of cases in each exposure quartile. Hazard ratios (HRs) were estimated from the maximum partial likelihood ratio test and two‐sided 95% CIs were based on the profile likelihood. The exposure surrogates were also analyzed as continuous time‐dependent variables in log‐linear models where the HR increases exponentially with exposure or, equivalently, the log of the HR increases linearly with exposure. Because the exposure surrogates were generally right‐skewed and exposure–response relationships in occupational studies are often attenuated at high exposure levels,\n45\n log‐linear models were also evaluated excluding person‐time in the top decile of exposure. The healthy worker survivor effect was addressed by repeating the analyses also matching on employment duration (<10, 10–<20, 20–<30, and 30+ years). Modeling analyses were restricted to analyses with 30 or more observed cases.", "Table 1 shows the demographic characteristics of the full and restricted cohorts. Most members of the full cohort were male (96.3%) and White (78.6%). Over three‐fourths (78.1%) of the restricted cohort of male firefighters were also White. The mean employment duration for both cohorts was 21 years. At the end of the follow‐up, 38.2% of the full cohort and 25.3% of the restricted cohort were deceased.\nCharacteristics of the full and restricted cohorts\nAbbreviation: IQR, interquartile range.\nMale firefighters of a known race who were hired in 1950 or later and employed for at least 1 year.\nVital status at the end of follow‐up (December 31, 2014) or the date of diagnosis, whichever was earlier.\nCell size <11 or reporting the exact number allows a cell size <11 to be derived.\nAs shown in Table 2, the incidence of all ESRD was less than expected among the full (SIR = 0.79; 95% CI = 0.69–0.89; observed = 247) and restricted (SIR: 0.73; 95% CI = 0.63–0.85; observed = 177) cohorts. The median age at the time of onset was 69 years (interquartile range = 61–77) and 67 years (interquartile range: 59–73) among the full and restricted cohorts, respectively. SIRs for ESRD due to metabolic disease (full cohort: 2.84; 95% CI = 0.76–7.26; restricted cohort: 1.92; 95% CI = 0.22–6.94) and ESRD due to collagen vascular disease (full cohort: 1.18; 95% CI = 0.32–3.01; restricted cohort: 1.17; 95% CI = 0.24–3.42) were elevated, although the CIs for these ESRD types were wide and included one. The incidence of systemic ESRD and diabetic ESRD was less than expected among the full (systemic ESRD: SIR = 0.76; 95% CI = 0.65–0.88; diabetic ESRD: SIR = 0.66; 95% CI = 0.52–0.82) and restricted (systemic ESRD: SIR = 0.71; 95% CI = 059–0.84; diabetic ESRD: SIR = 0.63; 95% CI = 0.49–0.81) cohorts. The incidence of ESRD caused by glomerulonephritis was also less than expected among the full cohort (SIR = 0.61; 95% CI = 0.35–0.97). The SIR for ESRD caused by glomerulonephritis among the restricted cohort was 0.66 (95% CI = 0.36–1.10).\nSIRs for ESRD among the full and restricted cohorts, 1977–2014\nAbbreviations: AIDS, acquired immunodeficiency syndrome; CI, confidence interval; ESRD, end‐stage renal disease; NR, not reported (cell size <11 or reporting the cell size allows numbers <11 to be derived); Obs, observed; SIR, standardized incidence ratio.\nESRD due to AIDS, sickle cell disease, and hereditary nephropathy were omitted because no cases were observed.\nThe results of internal analyses among the restricted cohort using categorical models are shown in Table 3 (20‐year lag) and Supporting Information: Table S2 (10‐year lag). Results reported herein were obtained from the analyses with matching on employment duration, unless stated otherwise. HRs for all ESRD obtained from categorical models increased in the second and third quartiles of exposed days, but then decreased in the top quartile. HRs for systemic ESRD increased with each exposed‐day quartile in 20‐ and 10‐year lagged analyses. HRs for hypertensive ESRD also increased with the exposed‐day quartile in 20‐ and 10‐year lagged analyses with an HR of 4.51 (95% CI = 0.97–20.9) in the top quartile in 20‐year lagged analyses. HRs for diabetic ESRD increased with exposed‐day quartile in 10‐year lagged analyses only.\nESRD HRs by 20‐year lagged exposure surrogates obtained from categorical models\na\n\n\nAbbreviations: CFD, Chicago fire department; CI, confidence interval; dept, fire departments included in the analysis; ESRD, end‐stage renal disease; HR, hazard ratio; PFD, Philadelphia fire department; SFFD, San Francisco fire department.\nAll models were adjusted for age (time scale) and for race (White/non‐White), date of birth (within 5 years), and fire department by matching. Some models were also adjusted for employment duration (<10, 10–<20, 20–<30, and 30+ years) by matching.\nAll modeling analyses were conducted among the restricted cohort and included 30 or more observed ESRD cases with an approximately equal number of cases in each exposure quartile. The overall numbers of ESRD cases in the analyses of exposed days were 177 for all ESRD, 132 for systemic ESRD, 63 for diabetic ESRD, and 53 for hypertensive ESRD.\nThe results of internal analyses among the restricted cohort using log‐linear models are shown in Table 4 (20‐year lag) and Supporting Information: Table S3 (10‐year lag). The HR for all ESRD was 1.11 (95% CI = 0.99–1.25) and 1.08 (95% CI = 0.98–1.18) per 1000 exposed days in 20‐ and 10‐year lagged analyses, respectively, when person‐time in the top exposure decile was excluded. In analogous linear models including person‐time in the top decile of exposure, the HR for all ESRD was 1.05 (95% CI = 0.96–1.16) and 1.07 (0.99–1.16) per 1000 exposed days in 20‐ and 10‐year lagged analyses, respectively.\nESRD HRs by 20‐year lagged exposure surrogates obtained from log‐linear models\na\n\n\nAbbreviations: CI, confidence interval; ESRD, end‐stage renal disease; HR, hazard ratio.\nAll models were adjusted for age (time scale) and for race (White/non‐White), date of birth (within 5 years), and fire department by matching. Some models were also adjusted for employment duration (<10, 10–<20, 20–<30, and 30+ years) by matching.\nAll modeling analyses were conducted among the restricted cohort and included 30 or more observed ESRD cases. The overall numbers of ESRD cases in analyses of exposed days were 177 for all ESRD, 132 for systemic ESRD, 63 for diabetic ESRD, and 53 for hypertensive ESRD.\nExposed days were available for Chicago, Philadelphia, and San Francisco firefighters; fire runs for Chicago and Philadelphia firefighters; and fire hours for Chicago firefighters.\n1000 exposed days, 1000 fire runs, or 1000 fire hours.\nSystemic ESRD HRs per 1000 exposed days were 1.05 (95% CI = 0.95–1.18) and 1.07 (95% CI = 0.98–1.17) in 20‐ and 10‐year lagged analyses, respectively, when person‐time in the top decile of exposure was included and 1.07 (95% CI = 0.95–1.23) and 1.05 (95% CI = 0.95–1.17) in 20‐ and 10‐year lagged analyses, respectively, when person‐time in the top decile of exposure was excluded. Hypertensive ESRD HRs per 1000 exposed days were 1.09 (95% CI = 0.93–1.29) and 1.10 (95% CI = 0.96–1.27) in 20‐ and 10‐year lagged analyses, respectively. When person‐time in the top exposure decile was excluded, the hypertensive ESRD HRs per 1000 exposed days were 1.13 (95% CI = 0.93–1.40) and 1.09 (95% CI = 0.94–1.29) in 20‐ and 10‐year lagged analyses, respectively. In 10‐year lagged analyses, the diabetic ESRD HR per 1000 exposed days was 1.03 (95% CI = 0.92–1.17) and 1.03 (95% CI = 0.90–1.19), including and excluding person‐time in the top exposure decile, respectively.", "The incidence of all ESRD, systemic ESRD, and diabetic ESRD was less than expected based on rates of ESRD in the general population, which is consistent with a healthy worker effect. Diabetes mellitus is the most common cause of ESRD in the United States with over 45% of incident ESRD attributed to diabetes in 2016.\n41\n Thus, these findings may reflect, in part, differences in the prevalence and control of diabetes mellitus among the cohort compared to the general population. This is supported by the previously reported mortality from diabetes mellitus in the mortality cohort (SMR = 0.73; 95% CI = 0.64–0.83).\n33\n A lower prevalence of hyperglycemia among male US firefighters than the general population has also been reported.\n46\n\n\nIncreasing HRs were observed for systemic and hypertensive ESRD with exposed days in 20‐ and 10‐year lagged analyses and for diabetic ESRD with exposed days in 10‐year lagged analyses using categorical models. In log‐linear models, positive trends were observed for all ESRD, systemic ESRD, and hypertensive ESRD with exposed days, although the CIs included one. These associations with exposed days, but not fire runs and fire hours, may reflect the limitation of using fire runs and fire hours as surrogates for heat exposure. Firefighters are also exposed to occupational heat exposure and experience heat‐related illnesses during other activities such as training.\n21\n, \n27\n For the purposes of this study, we assumed that all three exposure surrogates would estimate occupational heat exposure in addition to exposure to combustion byproducts of fire. However, the exposure surrogates are likely crude measures of exposure in this study because we were unable to include additional information in the job exposure matrices regarding risk factors for heat exposure such as environmental air temperature and humidity. Other potential reasons for observing trends with exposed days but not fire runs or fire hours include the lack of data on fire runs and fire hours for all three fire departments and the potential impact of shift work on kidney function.\n47\n, \n48\n\n\nThe positive trend in hypertensive ESRD with exposed days, although not statistically significant, raises the possibility that the work of firefighters may exacerbate underlying hypertensive kidney disease. This trend could also potentially be due to an association between hypertension and other occupational exposures. Occupational exposures that may increase the risk of hypertension include particulate matter from fire smoke,\n49\n shift work,\n50\n work‐related stress,\n51\n noise,\n52\n, \n53\n and perfluoroalkyl substances.\n54\n, \n55\n In a recent study, 69% of firefighters had hypertension and the prevalence of hypertension was higher among male firefighters than in the general population.\n56\n\n\nIn the epidemic of chronic kidney disease in Central America, kidney disease rarely co‐occurred with diabetes or hypertension.\n57\n Biopsies from affected Central American workers showed interstitial nephritis.\n58\n A limitation of the current study was the inability to evaluate the association of ESRD due to interstitial nephritis with exposure surrogates due to the small number of observed cases.\nThe current study also does not provide information on the risk of chronic kidney disease that does not progress to ESRD. Other limitations include the lack of data on occupational heat exposure, surrogates of prolonged occupational heat exposure, and risk factors for ESRD such as hypertension,\n59\n diabetes mellitus,\n60\n and obesity.\n61\n In addition, cumulative exposure surrogates were negatively associated with leaving employment, which suggests that HRs obtained from analyses using g‐estimation instead of matching on employment duration to account for the healthy worker survivor effect may be lower. The findings are also not generalizable to wildland firefighters because of differences in personal protective equipment (heavy encapsulating turnout gear with self‐contained breathing apparatus vs. light, somewhat breathable fabrics and no respiratory protection), and the typical amount and duration of physical exertion (short bursts of very strenuous work for about 30 min a few times during a 24‐h shift vs. relatively constant strenuous work during a 12‐h shift each day during a 14‐day deployment).\nIn summary, the firefighters in this study were not at an increased risk of ESRD compared to the general population and statistically significant associations of ESRD with exposure surrogates were not observed. Only nonstatistically significant positive associations of all ESRD, systemic ESRD, and hypertensive ESRD with exposed days were observed. Associations of ESRD due to interstitial nephritis with surrogates of exposure could not be assessed due to the small number of observed cases. Additional research using more sensitive outcome measures to evaluate potential kidney effects among firefighters is warranted.", "Lynne E. Pinkerton conceived the study and wrote the first draft of the manuscript. Stephen Bertke conducted the analysis. Travis L. Kubale, James H. Yiin, and Robert D. Daniels acquired the data and assembled the cohort. Matthew M. Dahm led the assessment of the exposure surrogates. All authors participated in the interpretation and presentation of results, approved the final manuscript, and agreed to be accountable for all aspects of the work.", "Lynne E. Pinkerton currently works for Maximus, Inc., which holds contracts with CDC NIOSH for assistance, including the conduct of research studies. The remaining authors declare that there are no conflicts of interest.", "John Meyer declares that he has no conflict of interest in the review and publication decision regarding this article.", "The study received approvals from the Institutional Review Boards of the National Institute for Occupational Safety and Health and the National Cancer Institute. Informed consent was waived for this records‐based study.", "The findings and conclusions in this report are those of the authors and do not necessarily represent the official position of the National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention.", "Supporting information.\nClick here for additional data file." ]
[ null, "methods", null, null, null, null, "results", "discussion", null, "COI-statement", null, null, null, "supplementary-material" ]
[ "end‐stage renal disease", "firefighters", "longitudinal studies" ]
Analysis of phenotype and gene mutation in three pedigrees with inherited antithrombin deficiency.
36268972
Inherited AT deficiency is an autosomal-dominant thrombophilic disorder usually caused by various SERPINC1 defects associated with a high risk of recurrent venous thromboembolism. In this article, the phenotype, gene mutation, and molecular pathogenic mechanisms were determined in three pedigrees with inherited AT deficiency.
BACKGROUND
Coagulation indices were examined on STAGO STA-R-MAX analyzer. The AT:Ag was analyzed by ELISA. All exons and flanking sequences of SERPINC1 were amplified by PCR. AT wild type and three mutant expression plasmids were constructed and then transfected into HEK293FT cells. The expression level of AT protein was analyzed by ELISA and Western blot.
METHODS
The AT:A and AT:Ag of probands 1 and 3 were decreased to 49% and 52 mg/dL, 38% and 44 mg/dL, respectively. The AT:A of proband 2 was decreased to 32%. The SERPINC1 gene analysis indicated that there was a p.Ile421Thr in proband 1, a p.Leu417Gln in proband 2, and a p.Met252Thr in proband 3, respectively. The AT mRNA expression level of the three mutants was not significantly different from AT-WT by qRT-PCR. The results of ELISA and Western blot tests showed that the AT-M252T and AT-I421T mutants had a higher AT expression than the AT wild type (AT-WT), and the AT protein expression of AT-L417Q mutants had no significant difference compared with AT-WT in the cell lysate. The AT expression levels of AT-M252T and AT-I421T mutants were lower than that of AT-WT, and there was no significant difference between AT-L417Q mutant and AT-WT in the supernatant.
RESULTS
The p.I421T and p.M252T mutations affected the secretion of AT protein leading to type I AT deficiency of probands 1 and 3. The p.Leu417Gln mutation was responsible for the impaired or ineffective activity AT protein in proband 2 and caused type II AT deficiency.
CONCLUSION
[ "Humans", "Pedigree", "Antithrombin III Deficiency", "Phenotype", "Mutation", "Antithrombins" ]
9701880
null
null
null
null
RESULTS
[SUBTITLE] Phenotype and genotype [SUBSECTION] In pedigree 1, the AT:A and AT:Ag of proband (II3), father (I1), and daughter (III1) all decreased; the values were 49%, 52 mg/dl, 48%, 42 mg/dl 40%, and 45 mg/dl, respectively, representing a type I AT deficiency. The phenotype results of the three pedigrees with inherited AT deficiency is shown in Table 1. Genetic analysis found that the above three members all carry the c.1358 T > C (Ile421Thr) heterozygous mutation in SERPINC1. In pedigree 2, the AT:A of proband (I1), and the eldest son (II2), second daughter (II4), and granddaughter (III3) decreased, with values 32%, 43%, 52%, and 48%, respectively; however, AT:Ag was within normal range, indicating a type II AT deficiency. They were all heterozygous for the c.1346 T > A (Leu417Gln) mutation. In pedigree 3, the AT:A and AT:Ag of proband (III2), mother (II3), and uncle (II1) showed a simultaneous decrease in AT:A and AT:Ag, with values of 38%, 44 mg/dl, 50%, 48 mg/dl, 40%, and 54 mg/dl, respectively, indicating a type I AT deficiency. Sequencing analysis revealed a heterozygous missense mutation c.851 T > C (p.Met252Thr) in the above three members of pedigree 3. The sequencing diagram is shown in Figure 2. Three corresponding sites in 100 healthy subjects were sequenced and excluded genetic polymorphisms. The FV‐Leiden and prothrombin G20210A of common thrombogenic gene mutations were screened and showed none existing in three probands and their family members. Phenotype results of three pedigrees with inherited AT deficiency Sequencing results: (A) c. 1358 T wild type, (B) c. 1358 T > C heterozygous mutation of proband 1; (C) c. 1346 T wild type, (D) c.1346 T > A heterozygous mutation of proband 2; (E) c.851 T wild type, (F) c.851 T > C heterozygous mutation of proband 3 The results of plasma AT protein from Western blot tests are shown in Figure 3. The plasma AT molecular weight of the three probands was consistent with the molecular weight of the normal mixed plasma control, but the plasma AT levels of probands 1 and 3 were significantly lower than those of the normal control. There was no difference between AT content of proband 2 and the normal control. WB results of Plasma AT protein. The control was human mixed plasma. Results 1, 2, and 3 were proband 1, 2, and 3 respectively In pedigree 1, the AT:A and AT:Ag of proband (II3), father (I1), and daughter (III1) all decreased; the values were 49%, 52 mg/dl, 48%, 42 mg/dl 40%, and 45 mg/dl, respectively, representing a type I AT deficiency. The phenotype results of the three pedigrees with inherited AT deficiency is shown in Table 1. Genetic analysis found that the above three members all carry the c.1358 T > C (Ile421Thr) heterozygous mutation in SERPINC1. In pedigree 2, the AT:A of proband (I1), and the eldest son (II2), second daughter (II4), and granddaughter (III3) decreased, with values 32%, 43%, 52%, and 48%, respectively; however, AT:Ag was within normal range, indicating a type II AT deficiency. They were all heterozygous for the c.1346 T > A (Leu417Gln) mutation. In pedigree 3, the AT:A and AT:Ag of proband (III2), mother (II3), and uncle (II1) showed a simultaneous decrease in AT:A and AT:Ag, with values of 38%, 44 mg/dl, 50%, 48 mg/dl, 40%, and 54 mg/dl, respectively, indicating a type I AT deficiency. Sequencing analysis revealed a heterozygous missense mutation c.851 T > C (p.Met252Thr) in the above three members of pedigree 3. The sequencing diagram is shown in Figure 2. Three corresponding sites in 100 healthy subjects were sequenced and excluded genetic polymorphisms. The FV‐Leiden and prothrombin G20210A of common thrombogenic gene mutations were screened and showed none existing in three probands and their family members. Phenotype results of three pedigrees with inherited AT deficiency Sequencing results: (A) c. 1358 T wild type, (B) c. 1358 T > C heterozygous mutation of proband 1; (C) c. 1346 T wild type, (D) c.1346 T > A heterozygous mutation of proband 2; (E) c.851 T wild type, (F) c.851 T > C heterozygous mutation of proband 3 The results of plasma AT protein from Western blot tests are shown in Figure 3. The plasma AT molecular weight of the three probands was consistent with the molecular weight of the normal mixed plasma control, but the plasma AT levels of probands 1 and 3 were significantly lower than those of the normal control. There was no difference between AT content of proband 2 and the normal control. WB results of Plasma AT protein. The control was human mixed plasma. Results 1, 2, and 3 were proband 1, 2, and 3 respectively [SUBTITLE] Bioformatics [SUBSECTION] The bioinformatics prediction results are shown in Table 2. MutationTaster, PolyPhen‐2, PROVEAN, LRT, and SIFT all predicted the p.Met252Thr, and p.Ile421Thr mutations as “pathogenic and harmful”. For the p. Leu417Gln mutation, MutationTaster, PolyPhen‐2, and LRT suggested that it was harmful; SIFT and PROVEAN software labeled the mutations as “tolerable” and “neutral” respectively. 18 The ACMG guidelines classified p.Met252Thr and p.Ile421Thr mutations as “pathogenic,” and the p. Leu417Gln mutation as “likely pathogenic.” The conservation analysis results showed that the Met252, Leu417, and Ile421 were highly conserved among homologous species (Figure 4). In the p.Ile421Thr wild type, when Ile421 was replaced by Thr421, the original hydrogen bond was unchanged, but new hydrogen bonds were formed between Thr421 and Ile412, and Thr421 and Ile422, as shown in Figure 5. In the wild‐type AT protein, the main chain of Leu417 formed two hydrogen bonds with the side chain of Glu414 and Ser52. In the wild‐type AT model, there were two hydrogen bonds between the main chain of Met252 residues and Met320. When the Met252 was substituted by Thr, the original hydrogen bond remained, and an additional hydrogen bond was formed with Met320 (as shown in Figure 5). Bioinformatics prediction results of mutations Note: Meanings of scores are as follows: Mutation Taster: a probability close to 1 indicates a high predictive value; PolyPhen‐2: scores are evaluated as 0.000 (most probably benign) to 1.000 (most probably damaging); PROVEAN: the predefined threshold of score is 2.5, the score 2.5 or less (deleterious), greater than 2.5 (neutral); LRT: scores 0.01 or less (deleterious), greater than 0.01 (benign); SIFT: scores range from 0 to 1, 0.05 or less (damaging), greater than 0.05 (tolerated). Conservation analysis of homologous species of Met252, Leu417, and Ile421 Mutation model analysis (A) p.Ile421Thr wild type, (B) p.Ile421Thr mutant type, (C) p.Met252Thr wild type, (D) p.Met252Thr mutant type. Hydrogen bonds are indicated by green dotted lines. The bioinformatics prediction results are shown in Table 2. MutationTaster, PolyPhen‐2, PROVEAN, LRT, and SIFT all predicted the p.Met252Thr, and p.Ile421Thr mutations as “pathogenic and harmful”. For the p. Leu417Gln mutation, MutationTaster, PolyPhen‐2, and LRT suggested that it was harmful; SIFT and PROVEAN software labeled the mutations as “tolerable” and “neutral” respectively. 18 The ACMG guidelines classified p.Met252Thr and p.Ile421Thr mutations as “pathogenic,” and the p. Leu417Gln mutation as “likely pathogenic.” The conservation analysis results showed that the Met252, Leu417, and Ile421 were highly conserved among homologous species (Figure 4). In the p.Ile421Thr wild type, when Ile421 was replaced by Thr421, the original hydrogen bond was unchanged, but new hydrogen bonds were formed between Thr421 and Ile412, and Thr421 and Ile422, as shown in Figure 5. In the wild‐type AT protein, the main chain of Leu417 formed two hydrogen bonds with the side chain of Glu414 and Ser52. In the wild‐type AT model, there were two hydrogen bonds between the main chain of Met252 residues and Met320. When the Met252 was substituted by Thr, the original hydrogen bond remained, and an additional hydrogen bond was formed with Met320 (as shown in Figure 5). Bioinformatics prediction results of mutations Note: Meanings of scores are as follows: Mutation Taster: a probability close to 1 indicates a high predictive value; PolyPhen‐2: scores are evaluated as 0.000 (most probably benign) to 1.000 (most probably damaging); PROVEAN: the predefined threshold of score is 2.5, the score 2.5 or less (deleterious), greater than 2.5 (neutral); LRT: scores 0.01 or less (deleterious), greater than 0.01 (benign); SIFT: scores range from 0 to 1, 0.05 or less (damaging), greater than 0.05 (tolerated). Conservation analysis of homologous species of Met252, Leu417, and Ile421 Mutation model analysis (A) p.Ile421Thr wild type, (B) p.Ile421Thr mutant type, (C) p.Met252Thr wild type, (D) p.Met252Thr mutant type. Hydrogen bonds are indicated by green dotted lines. [SUBTITLE] Transfection and expression [SUBSECTION] The AT‐M252T, AT‐I421T, and AT‐L417Q mutant plasmids were successfully constructed by site‐directed mutation without other mutation sites verified by sequencing. The wild type and each mutant plasmid were observed under normal field and fluorescence field after 48 h transfection. The results are shown in Figures S1–S4. The wild‐type and each mutant plasmid were successfully transfected. The efficiency was approximately 85%. The mRNA expression level of each group was detected by the fluorescence quantitative RT‐PCR method; the results are shown in Figure S2. AT gene expression of the AT‐WT plasmid was 100%, AT‐M252T, AT‐I421T, and AT‐L417Q mutant were 94.65%, 94.62%, and 107.6%, respectively. There was no significant difference between these and AT‐WT in mRNA expression. The nontransfected control and samples transfected with empty vector did not express AT mRNA. The AT:Ag of the wild‐type and each mutant plasmid group were measured by ELISA. The cell culture supernatant and lysate of the wild‐type group were defined as 100%, and the calculated results were statistically analyzed by GraphPad 8.0 software as shown in Figure S3. The AT expression levels of AT‐M252T and AT‐I421T mutants in the cell culture supernatant were lower than those of wild‐type WT, while the expression levels of AT‐M252T and AT‐I421T mutants in the cell lysate were higher than that of wild‐type WT. The expression of AT of AT‐L417Q mutant in the cell culture supernatant and lysate was equivalent to that of the wild type, and there was no statistical difference. Semi‐quantitative analyses of AT proteins were performed by Western blot in the cell lysate and culture supernatant. The results are shown in Figure S4, respectively. AT protein of AT‐M252T and AT‐I421T mutants expressed significantly more than the wild‐type in the cell lysate. AT protein expressions of AT‐M252T and AT‐I421T mutant were significantly reduced, compared with the wild type. AT protein expression of AT‐L417Q mutant levels were not much different from wild‐type cells in lysate and culture supernatant. The Western blot results of the three mutants were consistent with the ELISA results. The AT‐M252T, AT‐I421T, and AT‐L417Q mutant plasmids were successfully constructed by site‐directed mutation without other mutation sites verified by sequencing. The wild type and each mutant plasmid were observed under normal field and fluorescence field after 48 h transfection. The results are shown in Figures S1–S4. The wild‐type and each mutant plasmid were successfully transfected. The efficiency was approximately 85%. The mRNA expression level of each group was detected by the fluorescence quantitative RT‐PCR method; the results are shown in Figure S2. AT gene expression of the AT‐WT plasmid was 100%, AT‐M252T, AT‐I421T, and AT‐L417Q mutant were 94.65%, 94.62%, and 107.6%, respectively. There was no significant difference between these and AT‐WT in mRNA expression. The nontransfected control and samples transfected with empty vector did not express AT mRNA. The AT:Ag of the wild‐type and each mutant plasmid group were measured by ELISA. The cell culture supernatant and lysate of the wild‐type group were defined as 100%, and the calculated results were statistically analyzed by GraphPad 8.0 software as shown in Figure S3. The AT expression levels of AT‐M252T and AT‐I421T mutants in the cell culture supernatant were lower than those of wild‐type WT, while the expression levels of AT‐M252T and AT‐I421T mutants in the cell lysate were higher than that of wild‐type WT. The expression of AT of AT‐L417Q mutant in the cell culture supernatant and lysate was equivalent to that of the wild type, and there was no statistical difference. Semi‐quantitative analyses of AT proteins were performed by Western blot in the cell lysate and culture supernatant. The results are shown in Figure S4, respectively. AT protein of AT‐M252T and AT‐I421T mutants expressed significantly more than the wild‐type in the cell lysate. AT protein expressions of AT‐M252T and AT‐I421T mutant were significantly reduced, compared with the wild type. AT protein expression of AT‐L417Q mutant levels were not much different from wild‐type cells in lysate and culture supernatant. The Western blot results of the three mutants were consistent with the ELISA results.
CONCLUSION
We reported three heterozygous missense mutations in the SERPINC1 gene, namely c.1358 T > C (p.Ile421Thr), c.1346 T > A (p.Leu417Gln), and c.851 T > C (p.Met252Thr). The pathogenic mechanisms of the three mutations were preliminarily explored through bioinformatics, protein modeling, and in vitro expression analysis, which enriched the AT genetic mutation database.
[ "INTRODUCTION", "Patients", "Plasma AT assays", "\nAT gene analysis", "Bioformatics analysis", "Construction, transfection, and purification", "Phenotype and genotype", "Bioformatics", "Transfection and expression", "AUTHOR CONTRIBUTIONS", "ETHICS STATEMENT" ]
[ "Antithrombin (AT) is the major serine protease inhibitor (SERPIN) in plasma that regulates the proteolytic activities of coagulation proteases of both intrinsic and extrinsic pathways. AT is synthesized predominantly by hepatocytes, has a half‐life of approximately 2.4 days and a molecular weight of 58 kDa, and contains 432 amino acids.\n1\n AT is the major inhibitor of thrombin, activated coagulation factor IX (FIXa), and activated coagulation factor Х (FXa) in plasma, and is also able to inactivate the other serine proteases of the intrinsic coagulation pathway, activated factors XI and XII (FXIa and FXIIa).\n2\n In the physiologic state, AT circulates in a relatively inefficient inhibitor form. However, the anticoagulant effect of AT is greatly increased (at least 1000‐fold) by the presence of heparin and other heparin‐like glycosaminoglycans. This effect is the basis for the use of heparin and low‐molecular‐weight heparins (LMWH) as anticoagulants in patients with venous thromboembolism (VTE).\n3\n\n\nThe AT gene (SERPINC1) which encodes antithrombin is mapped on chromosome 1q23‐25 and comprises 7 exons encompassing 13.5 kb of genomic DNA.\n4\n The 1392 bp mRNA codes for a 464 amino acid polypeptide chain that undergoes several posttranslational modifications including cleavage of the signal peptide (32 amino acids) and N‐glycosylation.\n5\n, \n6\n Inherited AT deficiency is an autosomal‐dominant disorder caused by a defect in the SERPINC1 gene. It was first described in 1965 as one of the important risk factors for hemostatic diseases characterized by hereditary and recurrent thrombosis. In healthy people, the prevalence of inherited AT deficiency is around 0.02%–0.25%, but the incidence in patients with venous thrombosis is increased around 1%–2%.\n7\n, \n8\n, \n9\n Inherited AT deficiency is divided into type І with a reduction in the antigen levels and functional activity and type II deficiency with reduced antithrombin activity but almost normal antigen level.\n10\n, \n11\n\n\nMost cases with type I defects are heterozygous; homozygous patients have a higher risk of severe venous thromboembolism (VTE) in childhood and may even be fatal in utero.\n9\n For type II AT deficiency, the level of AT in the patient's plasma is not significantly affected; therefore, we speculate that type II has a slight thrombotic effect. Related studies have shown that patients with type II deficiency exhibit extensive and heterogeneous clinical phenotype.\n12\n\n\nInherited AT deficiency is widely considered to be one of the causes of thrombotic diseases. However, there are a few large cohort studies reported. More than 400 different mutations have been registered in the AT Mutation Database (The Human Gene Mutation Database, http://www.hgmd.cf.ac.uk/ac/all.php). Most are missense/nonsense mutations, the remaining being small insertion/deletion, large insertion/deletion, shear mutation, and complex mutation. The SERPINC1 gene mutations are heterogeneous in the population, and there are not many race‐specific mutations.\n13\n, \n14\n, \n15\n\n\nInherited AT deficiency is the main genetic risk factor for deep vein thrombosis (DVT) patients in China.\n16\n Sporadic cases with SERPINC1 gene mutations were reported in the population; however, no mutation hotspots were recognized. In this article, we presented three Chinese pedigrees with inherited AT deficiency, the phenotype, gene mutations, and expression of mutation sites in vitro. The molecular bases of the three pedigrees were analyzed.", "Proband 1 was a 27‐year‐old woman who was admitted to the emergency department of our hospital for a sudden headache during puerperium, accompanied by nausea and vomiting. Magnetic resonance venography (MRV) showed thrombosis in the straight sinus, confluence of sinuses, and left transverse sinus. Seven family members of three generations had no relevant clinical manifestations. The pedigree is shown in Figure 1A.\nThree pedigrees of inherited AT deficiency\nProband 2 was a 74‐year‐old man, sent to our hospital for sudden left limb weakness and no movement of the upper and lower limbs. B‐ultrasound examination of the blood vessels of the lower extremities showed that there was thrombosis in the myenteric venous plexus of the legs. The pedigree is shown in Figure 1B.\nProband 3 was a 30‐year‐old pregnant woman, admitted to our hospital because her left leg was swollen and painful. She had undergone a cesarean section 4 years ago and developed pulmonary embolism (PE) after that delivery. Her mother and uncle had the same history of thrombosis, and her grandfather died of PE at the age of 64, while the other family members had no relevant clinical manifestations. The pedigree is shown in Figure 1C.\nIn order to establish a laboratory reference range, one hundred healthy subjects were randomly selected from our hospital as controls to exclude genetic polymorphisms; they included 50 males and 50 females with an average age of 28 years (from 16 to 52 years), without liver or kidney disease, no history of bleeding and thrombosis. Our study was reviewed and approved by the Ethics Committee of the First Affiliated Hospital of Wenzhou Medical University (China). All participants signed a written informed consent.", "Peripheral blood samples of three family members and 100 healthy subjects were collected in anticoagulant tubes and centrifuged at 2100 g for 10 min, to obtain upper platelet‐poor plasma (PPP) and lower blood cells. The upper plasma was examined by coagulation test, and the lower blood cells were used for genomic DNA extraction.\nProthrombin time (PT), activated partial thromboplastin time (APTT), thrombin time (TT), fibrinogen (FIB), and protein S activity (PS:A) were analyzed using the STAGO STA‐R‐Max automatic blood coagulometer (Diagnostica Stago). D‐dimers were measured by the immunoturbidimetric method, AT:A, and protein C activity (PC:A) by the chromogenic substrate method. All laboratory tests were performed in accordance with the manufacturer's instructions (STAGO Company). The AT:Ag were detected by enzyme‐linked immunosorbent assay (ELISA), using the Human Antithrombin‐III (MIM #613118) ELISA kit (Hangzhou Lianke Biotechnology Co., Ltd). The AT protein content was processed and analyzed by Western blot.", "Peripheral blood genomic DNA of all subjects was extracted from the anticoagulant blood samples using a DNA Extraction Kit (TIANGEN) according to the manufacturer's protocol. Primers for all 7 exons and their flanking sequences of SERPINC1 were designed with NCBI Primer. The specific amplification conditions were performed as previously described.\n17\n The amplified products were identified and sequenced (Shanghai Personal Biotechnology Co., Ltd.). The sequencing results were compared with the SERPINC1 standard sequence published in NCBI (GenBank ID: NG_012462.1), using Chromas software for mutation sites. Primers for R560Q and G20210A mutations which are common risk factors for hereditary venous thromboembolism were designed, and corresponding sites of the probands and their family members were screened.", "The bioinformatics tools, including Mutation Taster (http://www.mutationtaster.org/), PROVEAN (http:// provean.jcvi.org/index.php), LRT (http://varcards.biols.ac.cn/), SIFT (http://sift.jcvi.org), and PolyPhen‐2 (http://genetics.bwh.harvard.edu/pph2/index.shtml), were applied to predict and analyze the possible impact of novel mutation on the structure and function of proteins.\nConservation analysis relied on ClustalX‐2.1‐win (Science Foundation Ireland), a multiple sequence alignment software used to analyze the conservation of affected amino acids with nine homologous species (HomoloGene, http://www.ncbi.nlm.nih. gov/homologene).\nThe PyMol software package (DeLano Scientific) was used to assess local structural effects of mutation in the AT gene, to determine the exact type and position of amino acid substitution in the three‐dimensional structural model. The AT protein crystal structure data were obtained from the Protein Data Bank (PDB, http://www.rcsb.org/pdb/home/home.do, PDB ID: 1ANT).", "The vector used in this experiment was pCDH‐copGFP‐T2A‐Puro vector. Wild ‐type AT plasmid (AT‐WT) and empty vector were purchased from Nanjing Tsingke Biotechnology Co., Ltd. The AT‐WT was used as a template to construct AT‐M252T (p.Met252Thr), AT‐I421T (p.Ile421Thr), and AT‐L417Q (p.Leu417Gln) expression plasmids, according to manufacturer's instructions (QuikChange Lightning Site‐Directed Mutagenesis Kit). Mutant primers were designed using PrimerX online software (http://www.bioinformatics.org/primerx/) and synthesized by Nanjing Tsingke Biotechnology Co., Ltd.\nHEK293FT cells were cultured in Dulbecco's modified Eagle medium (DMEM), which was supplemented with 10% fetal bovine serum (FBS). The wild‐type and mutant plasmids were transfected into human embryonic kidney 293 cells (HEK293FT). After 48 h transfection, total mRNA was extracted from the cells using TRIzol reagent and then reversely transcribed to cDNA. AT mRNA levels were determined using Magic SYBR Green qPCR Mix (Maibo, M223), according to manufacturer's instructions. The AT:Ag expression in cell culture supernatant and cell lysate was collected and analyzed by ELISA. Western blot was used to detect AT protein level in the cell supernatant and lysate.", "In pedigree 1, the AT:A and AT:Ag of proband (II3), father (I1), and daughter (III1) all decreased; the values were 49%, 52 mg/dl, 48%, 42 mg/dl 40%, and 45 mg/dl, respectively, representing a type I AT deficiency. The phenotype results of the three pedigrees with inherited AT deficiency is shown in Table 1. Genetic analysis found that the above three members all carry the c.1358 T > C (Ile421Thr) heterozygous mutation in SERPINC1. In pedigree 2, the AT:A of proband (I1), and the eldest son (II2), second daughter (II4), and granddaughter (III3) decreased, with values 32%, 43%, 52%, and 48%, respectively; however, AT:Ag was within normal range, indicating a type II AT deficiency. They were all heterozygous for the c.1346 T > A (Leu417Gln) mutation. In pedigree 3, the AT:A and AT:Ag of proband (III2), mother (II3), and uncle (II1) showed a simultaneous decrease in AT:A and AT:Ag, with values of 38%, 44 mg/dl, 50%, 48 mg/dl, 40%, and 54 mg/dl, respectively, indicating a type I AT deficiency. Sequencing analysis revealed a heterozygous missense mutation c.851 T > C (p.Met252Thr) in the above three members of pedigree 3. The sequencing diagram is shown in Figure 2. Three corresponding sites in 100 healthy subjects were sequenced and excluded genetic polymorphisms. The FV‐Leiden and prothrombin G20210A of common thrombogenic gene mutations were screened and showed none existing in three probands and their family members.\nPhenotype results of three pedigrees with inherited AT deficiency\nSequencing results: (A) c. 1358 T wild type, (B) c. 1358 T > C heterozygous mutation of proband 1; (C) c. 1346 T wild type, (D) c.1346 T > A heterozygous mutation of proband 2; (E) c.851 T wild type, (F) c.851 T > C heterozygous mutation of proband 3\nThe results of plasma AT protein from Western blot tests are shown in Figure 3. The plasma AT molecular weight of the three probands was consistent with the molecular weight of the normal mixed plasma control, but the plasma AT levels of probands 1 and 3 were significantly lower than those of the normal control. There was no difference between AT content of proband 2 and the normal control.\nWB results of Plasma AT protein. The control was human mixed plasma. Results 1, 2, and 3 were proband 1, 2, and 3 respectively", "The bioinformatics prediction results are shown in Table 2. MutationTaster, PolyPhen‐2, PROVEAN, LRT, and SIFT all predicted the p.Met252Thr, and p.Ile421Thr mutations as “pathogenic and harmful”. For the p. Leu417Gln mutation, MutationTaster, PolyPhen‐2, and LRT suggested that it was harmful; SIFT and PROVEAN software labeled the mutations as “tolerable” and “neutral” respectively.\n18\n The ACMG guidelines classified p.Met252Thr and p.Ile421Thr mutations as “pathogenic,” and the p. Leu417Gln mutation as “likely pathogenic.” The conservation analysis results showed that the Met252, Leu417, and Ile421 were highly conserved among homologous species (Figure 4). In the p.Ile421Thr wild type, when Ile421 was replaced by Thr421, the original hydrogen bond was unchanged, but new hydrogen bonds were formed between Thr421 and Ile412, and Thr421 and Ile422, as shown in Figure 5. In the wild‐type AT protein, the main chain of Leu417 formed two hydrogen bonds with the side chain of Glu414 and Ser52. In the wild‐type AT model, there were two hydrogen bonds between the main chain of Met252 residues and Met320. When the Met252 was substituted by Thr, the original hydrogen bond remained, and an additional hydrogen bond was formed with Met320 (as shown in Figure 5).\nBioinformatics prediction results of mutations\n\nNote: Meanings of scores are as follows: Mutation Taster: a probability close to 1 indicates a high predictive value; PolyPhen‐2: scores are evaluated as 0.000 (most probably benign) to 1.000 (most probably damaging); PROVEAN: the predefined threshold of score is 2.5, the score 2.5 or less (deleterious), greater than 2.5 (neutral); LRT: scores 0.01 or less (deleterious), greater than 0.01 (benign); SIFT: scores range from 0 to 1, 0.05 or less (damaging), greater than 0.05 (tolerated).\nConservation analysis of homologous species of Met252, Leu417, and Ile421\nMutation model analysis (A) p.Ile421Thr wild type, (B) p.Ile421Thr mutant type, (C) p.Met252Thr wild type, (D) p.Met252Thr mutant type. Hydrogen bonds are indicated by green dotted lines.", "The AT‐M252T, AT‐I421T, and AT‐L417Q mutant plasmids were successfully constructed by site‐directed mutation without other mutation sites verified by sequencing. The wild type and each mutant plasmid were observed under normal field and fluorescence field after 48 h transfection. The results are shown in Figures S1–S4. The wild‐type and each mutant plasmid were successfully transfected. The efficiency was approximately 85%.\nThe mRNA expression level of each group was detected by the fluorescence quantitative RT‐PCR method; the results are shown in Figure S2. AT gene expression of the AT‐WT plasmid was 100%, AT‐M252T, AT‐I421T, and AT‐L417Q mutant were 94.65%, 94.62%, and 107.6%, respectively. There was no significant difference between these and AT‐WT in mRNA expression. The nontransfected control and samples transfected with empty vector did not express AT mRNA.\nThe AT:Ag of the wild‐type and each mutant plasmid group were measured by ELISA. The cell culture supernatant and lysate of the wild‐type group were defined as 100%, and the calculated results were statistically analyzed by GraphPad 8.0 software as shown in Figure S3. The AT expression levels of AT‐M252T and AT‐I421T mutants in the cell culture supernatant were lower than those of wild‐type WT, while the expression levels of AT‐M252T and AT‐I421T mutants in the cell lysate were higher than that of wild‐type WT. The expression of AT of AT‐L417Q mutant in the cell culture supernatant and lysate was equivalent to that of the wild type, and there was no statistical difference.\nSemi‐quantitative analyses of AT proteins were performed by Western blot in the cell lysate and culture supernatant. The results are shown in Figure S4, respectively. AT protein of AT‐M252T and AT‐I421T mutants expressed significantly more than the wild‐type in the cell lysate. AT protein expressions of AT‐M252T and AT‐I421T mutant were significantly reduced, compared with the wild type. AT protein expression of AT‐L417Q mutant levels were not much different from wild‐type cells in lysate and culture supernatant. The Western blot results of the three mutants were consistent with the ELISA results.", "MSW and ML designed experiments, analyzed data, wrote the article, and supervised the project. SQL and STJ performed molecular modeling. SQL and YHJ designed experiments, conducted genetic analysis, and coagulation assays. All authors approved the final version of the article.", "Our study was approved by the Ethics Committee of the First Affiliated Hospital of Wenzhou Medical University (China)." ]
[ null, null, null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "MATERIALS AND METHODS", "Patients", "Plasma AT assays", "\nAT gene analysis", "Bioformatics analysis", "Construction, transfection, and purification", "RESULTS", "Phenotype and genotype", "Bioformatics", "Transfection and expression", "DISCUSSION", "CONCLUSION", "AUTHOR CONTRIBUTIONS", "CONFLICT OF INTEREST", "ETHICS STATEMENT", "Supporting information" ]
[ "Antithrombin (AT) is the major serine protease inhibitor (SERPIN) in plasma that regulates the proteolytic activities of coagulation proteases of both intrinsic and extrinsic pathways. AT is synthesized predominantly by hepatocytes, has a half‐life of approximately 2.4 days and a molecular weight of 58 kDa, and contains 432 amino acids.\n1\n AT is the major inhibitor of thrombin, activated coagulation factor IX (FIXa), and activated coagulation factor Х (FXa) in plasma, and is also able to inactivate the other serine proteases of the intrinsic coagulation pathway, activated factors XI and XII (FXIa and FXIIa).\n2\n In the physiologic state, AT circulates in a relatively inefficient inhibitor form. However, the anticoagulant effect of AT is greatly increased (at least 1000‐fold) by the presence of heparin and other heparin‐like glycosaminoglycans. This effect is the basis for the use of heparin and low‐molecular‐weight heparins (LMWH) as anticoagulants in patients with venous thromboembolism (VTE).\n3\n\n\nThe AT gene (SERPINC1) which encodes antithrombin is mapped on chromosome 1q23‐25 and comprises 7 exons encompassing 13.5 kb of genomic DNA.\n4\n The 1392 bp mRNA codes for a 464 amino acid polypeptide chain that undergoes several posttranslational modifications including cleavage of the signal peptide (32 amino acids) and N‐glycosylation.\n5\n, \n6\n Inherited AT deficiency is an autosomal‐dominant disorder caused by a defect in the SERPINC1 gene. It was first described in 1965 as one of the important risk factors for hemostatic diseases characterized by hereditary and recurrent thrombosis. In healthy people, the prevalence of inherited AT deficiency is around 0.02%–0.25%, but the incidence in patients with venous thrombosis is increased around 1%–2%.\n7\n, \n8\n, \n9\n Inherited AT deficiency is divided into type І with a reduction in the antigen levels and functional activity and type II deficiency with reduced antithrombin activity but almost normal antigen level.\n10\n, \n11\n\n\nMost cases with type I defects are heterozygous; homozygous patients have a higher risk of severe venous thromboembolism (VTE) in childhood and may even be fatal in utero.\n9\n For type II AT deficiency, the level of AT in the patient's plasma is not significantly affected; therefore, we speculate that type II has a slight thrombotic effect. Related studies have shown that patients with type II deficiency exhibit extensive and heterogeneous clinical phenotype.\n12\n\n\nInherited AT deficiency is widely considered to be one of the causes of thrombotic diseases. However, there are a few large cohort studies reported. More than 400 different mutations have been registered in the AT Mutation Database (The Human Gene Mutation Database, http://www.hgmd.cf.ac.uk/ac/all.php). Most are missense/nonsense mutations, the remaining being small insertion/deletion, large insertion/deletion, shear mutation, and complex mutation. The SERPINC1 gene mutations are heterogeneous in the population, and there are not many race‐specific mutations.\n13\n, \n14\n, \n15\n\n\nInherited AT deficiency is the main genetic risk factor for deep vein thrombosis (DVT) patients in China.\n16\n Sporadic cases with SERPINC1 gene mutations were reported in the population; however, no mutation hotspots were recognized. In this article, we presented three Chinese pedigrees with inherited AT deficiency, the phenotype, gene mutations, and expression of mutation sites in vitro. The molecular bases of the three pedigrees were analyzed.", "[SUBTITLE] Patients [SUBSECTION] Proband 1 was a 27‐year‐old woman who was admitted to the emergency department of our hospital for a sudden headache during puerperium, accompanied by nausea and vomiting. Magnetic resonance venography (MRV) showed thrombosis in the straight sinus, confluence of sinuses, and left transverse sinus. Seven family members of three generations had no relevant clinical manifestations. The pedigree is shown in Figure 1A.\nThree pedigrees of inherited AT deficiency\nProband 2 was a 74‐year‐old man, sent to our hospital for sudden left limb weakness and no movement of the upper and lower limbs. B‐ultrasound examination of the blood vessels of the lower extremities showed that there was thrombosis in the myenteric venous plexus of the legs. The pedigree is shown in Figure 1B.\nProband 3 was a 30‐year‐old pregnant woman, admitted to our hospital because her left leg was swollen and painful. She had undergone a cesarean section 4 years ago and developed pulmonary embolism (PE) after that delivery. Her mother and uncle had the same history of thrombosis, and her grandfather died of PE at the age of 64, while the other family members had no relevant clinical manifestations. The pedigree is shown in Figure 1C.\nIn order to establish a laboratory reference range, one hundred healthy subjects were randomly selected from our hospital as controls to exclude genetic polymorphisms; they included 50 males and 50 females with an average age of 28 years (from 16 to 52 years), without liver or kidney disease, no history of bleeding and thrombosis. Our study was reviewed and approved by the Ethics Committee of the First Affiliated Hospital of Wenzhou Medical University (China). All participants signed a written informed consent.\nProband 1 was a 27‐year‐old woman who was admitted to the emergency department of our hospital for a sudden headache during puerperium, accompanied by nausea and vomiting. Magnetic resonance venography (MRV) showed thrombosis in the straight sinus, confluence of sinuses, and left transverse sinus. Seven family members of three generations had no relevant clinical manifestations. The pedigree is shown in Figure 1A.\nThree pedigrees of inherited AT deficiency\nProband 2 was a 74‐year‐old man, sent to our hospital for sudden left limb weakness and no movement of the upper and lower limbs. B‐ultrasound examination of the blood vessels of the lower extremities showed that there was thrombosis in the myenteric venous plexus of the legs. The pedigree is shown in Figure 1B.\nProband 3 was a 30‐year‐old pregnant woman, admitted to our hospital because her left leg was swollen and painful. She had undergone a cesarean section 4 years ago and developed pulmonary embolism (PE) after that delivery. Her mother and uncle had the same history of thrombosis, and her grandfather died of PE at the age of 64, while the other family members had no relevant clinical manifestations. The pedigree is shown in Figure 1C.\nIn order to establish a laboratory reference range, one hundred healthy subjects were randomly selected from our hospital as controls to exclude genetic polymorphisms; they included 50 males and 50 females with an average age of 28 years (from 16 to 52 years), without liver or kidney disease, no history of bleeding and thrombosis. Our study was reviewed and approved by the Ethics Committee of the First Affiliated Hospital of Wenzhou Medical University (China). All participants signed a written informed consent.\n[SUBTITLE] Plasma AT assays [SUBSECTION] Peripheral blood samples of three family members and 100 healthy subjects were collected in anticoagulant tubes and centrifuged at 2100 g for 10 min, to obtain upper platelet‐poor plasma (PPP) and lower blood cells. The upper plasma was examined by coagulation test, and the lower blood cells were used for genomic DNA extraction.\nProthrombin time (PT), activated partial thromboplastin time (APTT), thrombin time (TT), fibrinogen (FIB), and protein S activity (PS:A) were analyzed using the STAGO STA‐R‐Max automatic blood coagulometer (Diagnostica Stago). D‐dimers were measured by the immunoturbidimetric method, AT:A, and protein C activity (PC:A) by the chromogenic substrate method. All laboratory tests were performed in accordance with the manufacturer's instructions (STAGO Company). The AT:Ag were detected by enzyme‐linked immunosorbent assay (ELISA), using the Human Antithrombin‐III (MIM #613118) ELISA kit (Hangzhou Lianke Biotechnology Co., Ltd). The AT protein content was processed and analyzed by Western blot.\nPeripheral blood samples of three family members and 100 healthy subjects were collected in anticoagulant tubes and centrifuged at 2100 g for 10 min, to obtain upper platelet‐poor plasma (PPP) and lower blood cells. The upper plasma was examined by coagulation test, and the lower blood cells were used for genomic DNA extraction.\nProthrombin time (PT), activated partial thromboplastin time (APTT), thrombin time (TT), fibrinogen (FIB), and protein S activity (PS:A) were analyzed using the STAGO STA‐R‐Max automatic blood coagulometer (Diagnostica Stago). D‐dimers were measured by the immunoturbidimetric method, AT:A, and protein C activity (PC:A) by the chromogenic substrate method. All laboratory tests were performed in accordance with the manufacturer's instructions (STAGO Company). The AT:Ag were detected by enzyme‐linked immunosorbent assay (ELISA), using the Human Antithrombin‐III (MIM #613118) ELISA kit (Hangzhou Lianke Biotechnology Co., Ltd). The AT protein content was processed and analyzed by Western blot.\n[SUBTITLE] \nAT gene analysis [SUBSECTION] Peripheral blood genomic DNA of all subjects was extracted from the anticoagulant blood samples using a DNA Extraction Kit (TIANGEN) according to the manufacturer's protocol. Primers for all 7 exons and their flanking sequences of SERPINC1 were designed with NCBI Primer. The specific amplification conditions were performed as previously described.\n17\n The amplified products were identified and sequenced (Shanghai Personal Biotechnology Co., Ltd.). The sequencing results were compared with the SERPINC1 standard sequence published in NCBI (GenBank ID: NG_012462.1), using Chromas software for mutation sites. Primers for R560Q and G20210A mutations which are common risk factors for hereditary venous thromboembolism were designed, and corresponding sites of the probands and their family members were screened.\nPeripheral blood genomic DNA of all subjects was extracted from the anticoagulant blood samples using a DNA Extraction Kit (TIANGEN) according to the manufacturer's protocol. Primers for all 7 exons and their flanking sequences of SERPINC1 were designed with NCBI Primer. The specific amplification conditions were performed as previously described.\n17\n The amplified products were identified and sequenced (Shanghai Personal Biotechnology Co., Ltd.). The sequencing results were compared with the SERPINC1 standard sequence published in NCBI (GenBank ID: NG_012462.1), using Chromas software for mutation sites. Primers for R560Q and G20210A mutations which are common risk factors for hereditary venous thromboembolism were designed, and corresponding sites of the probands and their family members were screened.\n[SUBTITLE] Bioformatics analysis [SUBSECTION] The bioinformatics tools, including Mutation Taster (http://www.mutationtaster.org/), PROVEAN (http:// provean.jcvi.org/index.php), LRT (http://varcards.biols.ac.cn/), SIFT (http://sift.jcvi.org), and PolyPhen‐2 (http://genetics.bwh.harvard.edu/pph2/index.shtml), were applied to predict and analyze the possible impact of novel mutation on the structure and function of proteins.\nConservation analysis relied on ClustalX‐2.1‐win (Science Foundation Ireland), a multiple sequence alignment software used to analyze the conservation of affected amino acids with nine homologous species (HomoloGene, http://www.ncbi.nlm.nih. gov/homologene).\nThe PyMol software package (DeLano Scientific) was used to assess local structural effects of mutation in the AT gene, to determine the exact type and position of amino acid substitution in the three‐dimensional structural model. The AT protein crystal structure data were obtained from the Protein Data Bank (PDB, http://www.rcsb.org/pdb/home/home.do, PDB ID: 1ANT).\nThe bioinformatics tools, including Mutation Taster (http://www.mutationtaster.org/), PROVEAN (http:// provean.jcvi.org/index.php), LRT (http://varcards.biols.ac.cn/), SIFT (http://sift.jcvi.org), and PolyPhen‐2 (http://genetics.bwh.harvard.edu/pph2/index.shtml), were applied to predict and analyze the possible impact of novel mutation on the structure and function of proteins.\nConservation analysis relied on ClustalX‐2.1‐win (Science Foundation Ireland), a multiple sequence alignment software used to analyze the conservation of affected amino acids with nine homologous species (HomoloGene, http://www.ncbi.nlm.nih. gov/homologene).\nThe PyMol software package (DeLano Scientific) was used to assess local structural effects of mutation in the AT gene, to determine the exact type and position of amino acid substitution in the three‐dimensional structural model. The AT protein crystal structure data were obtained from the Protein Data Bank (PDB, http://www.rcsb.org/pdb/home/home.do, PDB ID: 1ANT).\n[SUBTITLE] Construction, transfection, and purification [SUBSECTION] The vector used in this experiment was pCDH‐copGFP‐T2A‐Puro vector. Wild ‐type AT plasmid (AT‐WT) and empty vector were purchased from Nanjing Tsingke Biotechnology Co., Ltd. The AT‐WT was used as a template to construct AT‐M252T (p.Met252Thr), AT‐I421T (p.Ile421Thr), and AT‐L417Q (p.Leu417Gln) expression plasmids, according to manufacturer's instructions (QuikChange Lightning Site‐Directed Mutagenesis Kit). Mutant primers were designed using PrimerX online software (http://www.bioinformatics.org/primerx/) and synthesized by Nanjing Tsingke Biotechnology Co., Ltd.\nHEK293FT cells were cultured in Dulbecco's modified Eagle medium (DMEM), which was supplemented with 10% fetal bovine serum (FBS). The wild‐type and mutant plasmids were transfected into human embryonic kidney 293 cells (HEK293FT). After 48 h transfection, total mRNA was extracted from the cells using TRIzol reagent and then reversely transcribed to cDNA. AT mRNA levels were determined using Magic SYBR Green qPCR Mix (Maibo, M223), according to manufacturer's instructions. The AT:Ag expression in cell culture supernatant and cell lysate was collected and analyzed by ELISA. Western blot was used to detect AT protein level in the cell supernatant and lysate.\nThe vector used in this experiment was pCDH‐copGFP‐T2A‐Puro vector. Wild ‐type AT plasmid (AT‐WT) and empty vector were purchased from Nanjing Tsingke Biotechnology Co., Ltd. The AT‐WT was used as a template to construct AT‐M252T (p.Met252Thr), AT‐I421T (p.Ile421Thr), and AT‐L417Q (p.Leu417Gln) expression plasmids, according to manufacturer's instructions (QuikChange Lightning Site‐Directed Mutagenesis Kit). Mutant primers were designed using PrimerX online software (http://www.bioinformatics.org/primerx/) and synthesized by Nanjing Tsingke Biotechnology Co., Ltd.\nHEK293FT cells were cultured in Dulbecco's modified Eagle medium (DMEM), which was supplemented with 10% fetal bovine serum (FBS). The wild‐type and mutant plasmids were transfected into human embryonic kidney 293 cells (HEK293FT). After 48 h transfection, total mRNA was extracted from the cells using TRIzol reagent and then reversely transcribed to cDNA. AT mRNA levels were determined using Magic SYBR Green qPCR Mix (Maibo, M223), according to manufacturer's instructions. The AT:Ag expression in cell culture supernatant and cell lysate was collected and analyzed by ELISA. Western blot was used to detect AT protein level in the cell supernatant and lysate.", "Proband 1 was a 27‐year‐old woman who was admitted to the emergency department of our hospital for a sudden headache during puerperium, accompanied by nausea and vomiting. Magnetic resonance venography (MRV) showed thrombosis in the straight sinus, confluence of sinuses, and left transverse sinus. Seven family members of three generations had no relevant clinical manifestations. The pedigree is shown in Figure 1A.\nThree pedigrees of inherited AT deficiency\nProband 2 was a 74‐year‐old man, sent to our hospital for sudden left limb weakness and no movement of the upper and lower limbs. B‐ultrasound examination of the blood vessels of the lower extremities showed that there was thrombosis in the myenteric venous plexus of the legs. The pedigree is shown in Figure 1B.\nProband 3 was a 30‐year‐old pregnant woman, admitted to our hospital because her left leg was swollen and painful. She had undergone a cesarean section 4 years ago and developed pulmonary embolism (PE) after that delivery. Her mother and uncle had the same history of thrombosis, and her grandfather died of PE at the age of 64, while the other family members had no relevant clinical manifestations. The pedigree is shown in Figure 1C.\nIn order to establish a laboratory reference range, one hundred healthy subjects were randomly selected from our hospital as controls to exclude genetic polymorphisms; they included 50 males and 50 females with an average age of 28 years (from 16 to 52 years), without liver or kidney disease, no history of bleeding and thrombosis. Our study was reviewed and approved by the Ethics Committee of the First Affiliated Hospital of Wenzhou Medical University (China). All participants signed a written informed consent.", "Peripheral blood samples of three family members and 100 healthy subjects were collected in anticoagulant tubes and centrifuged at 2100 g for 10 min, to obtain upper platelet‐poor plasma (PPP) and lower blood cells. The upper plasma was examined by coagulation test, and the lower blood cells were used for genomic DNA extraction.\nProthrombin time (PT), activated partial thromboplastin time (APTT), thrombin time (TT), fibrinogen (FIB), and protein S activity (PS:A) were analyzed using the STAGO STA‐R‐Max automatic blood coagulometer (Diagnostica Stago). D‐dimers were measured by the immunoturbidimetric method, AT:A, and protein C activity (PC:A) by the chromogenic substrate method. All laboratory tests were performed in accordance with the manufacturer's instructions (STAGO Company). The AT:Ag were detected by enzyme‐linked immunosorbent assay (ELISA), using the Human Antithrombin‐III (MIM #613118) ELISA kit (Hangzhou Lianke Biotechnology Co., Ltd). The AT protein content was processed and analyzed by Western blot.", "Peripheral blood genomic DNA of all subjects was extracted from the anticoagulant blood samples using a DNA Extraction Kit (TIANGEN) according to the manufacturer's protocol. Primers for all 7 exons and their flanking sequences of SERPINC1 were designed with NCBI Primer. The specific amplification conditions were performed as previously described.\n17\n The amplified products were identified and sequenced (Shanghai Personal Biotechnology Co., Ltd.). The sequencing results were compared with the SERPINC1 standard sequence published in NCBI (GenBank ID: NG_012462.1), using Chromas software for mutation sites. Primers for R560Q and G20210A mutations which are common risk factors for hereditary venous thromboembolism were designed, and corresponding sites of the probands and their family members were screened.", "The bioinformatics tools, including Mutation Taster (http://www.mutationtaster.org/), PROVEAN (http:// provean.jcvi.org/index.php), LRT (http://varcards.biols.ac.cn/), SIFT (http://sift.jcvi.org), and PolyPhen‐2 (http://genetics.bwh.harvard.edu/pph2/index.shtml), were applied to predict and analyze the possible impact of novel mutation on the structure and function of proteins.\nConservation analysis relied on ClustalX‐2.1‐win (Science Foundation Ireland), a multiple sequence alignment software used to analyze the conservation of affected amino acids with nine homologous species (HomoloGene, http://www.ncbi.nlm.nih. gov/homologene).\nThe PyMol software package (DeLano Scientific) was used to assess local structural effects of mutation in the AT gene, to determine the exact type and position of amino acid substitution in the three‐dimensional structural model. The AT protein crystal structure data were obtained from the Protein Data Bank (PDB, http://www.rcsb.org/pdb/home/home.do, PDB ID: 1ANT).", "The vector used in this experiment was pCDH‐copGFP‐T2A‐Puro vector. Wild ‐type AT plasmid (AT‐WT) and empty vector were purchased from Nanjing Tsingke Biotechnology Co., Ltd. The AT‐WT was used as a template to construct AT‐M252T (p.Met252Thr), AT‐I421T (p.Ile421Thr), and AT‐L417Q (p.Leu417Gln) expression plasmids, according to manufacturer's instructions (QuikChange Lightning Site‐Directed Mutagenesis Kit). Mutant primers were designed using PrimerX online software (http://www.bioinformatics.org/primerx/) and synthesized by Nanjing Tsingke Biotechnology Co., Ltd.\nHEK293FT cells were cultured in Dulbecco's modified Eagle medium (DMEM), which was supplemented with 10% fetal bovine serum (FBS). The wild‐type and mutant plasmids were transfected into human embryonic kidney 293 cells (HEK293FT). After 48 h transfection, total mRNA was extracted from the cells using TRIzol reagent and then reversely transcribed to cDNA. AT mRNA levels were determined using Magic SYBR Green qPCR Mix (Maibo, M223), according to manufacturer's instructions. The AT:Ag expression in cell culture supernatant and cell lysate was collected and analyzed by ELISA. Western blot was used to detect AT protein level in the cell supernatant and lysate.", "[SUBTITLE] Phenotype and genotype [SUBSECTION] In pedigree 1, the AT:A and AT:Ag of proband (II3), father (I1), and daughter (III1) all decreased; the values were 49%, 52 mg/dl, 48%, 42 mg/dl 40%, and 45 mg/dl, respectively, representing a type I AT deficiency. The phenotype results of the three pedigrees with inherited AT deficiency is shown in Table 1. Genetic analysis found that the above three members all carry the c.1358 T > C (Ile421Thr) heterozygous mutation in SERPINC1. In pedigree 2, the AT:A of proband (I1), and the eldest son (II2), second daughter (II4), and granddaughter (III3) decreased, with values 32%, 43%, 52%, and 48%, respectively; however, AT:Ag was within normal range, indicating a type II AT deficiency. They were all heterozygous for the c.1346 T > A (Leu417Gln) mutation. In pedigree 3, the AT:A and AT:Ag of proband (III2), mother (II3), and uncle (II1) showed a simultaneous decrease in AT:A and AT:Ag, with values of 38%, 44 mg/dl, 50%, 48 mg/dl, 40%, and 54 mg/dl, respectively, indicating a type I AT deficiency. Sequencing analysis revealed a heterozygous missense mutation c.851 T > C (p.Met252Thr) in the above three members of pedigree 3. The sequencing diagram is shown in Figure 2. Three corresponding sites in 100 healthy subjects were sequenced and excluded genetic polymorphisms. The FV‐Leiden and prothrombin G20210A of common thrombogenic gene mutations were screened and showed none existing in three probands and their family members.\nPhenotype results of three pedigrees with inherited AT deficiency\nSequencing results: (A) c. 1358 T wild type, (B) c. 1358 T > C heterozygous mutation of proband 1; (C) c. 1346 T wild type, (D) c.1346 T > A heterozygous mutation of proband 2; (E) c.851 T wild type, (F) c.851 T > C heterozygous mutation of proband 3\nThe results of plasma AT protein from Western blot tests are shown in Figure 3. The plasma AT molecular weight of the three probands was consistent with the molecular weight of the normal mixed plasma control, but the plasma AT levels of probands 1 and 3 were significantly lower than those of the normal control. There was no difference between AT content of proband 2 and the normal control.\nWB results of Plasma AT protein. The control was human mixed plasma. Results 1, 2, and 3 were proband 1, 2, and 3 respectively\nIn pedigree 1, the AT:A and AT:Ag of proband (II3), father (I1), and daughter (III1) all decreased; the values were 49%, 52 mg/dl, 48%, 42 mg/dl 40%, and 45 mg/dl, respectively, representing a type I AT deficiency. The phenotype results of the three pedigrees with inherited AT deficiency is shown in Table 1. Genetic analysis found that the above three members all carry the c.1358 T > C (Ile421Thr) heterozygous mutation in SERPINC1. In pedigree 2, the AT:A of proband (I1), and the eldest son (II2), second daughter (II4), and granddaughter (III3) decreased, with values 32%, 43%, 52%, and 48%, respectively; however, AT:Ag was within normal range, indicating a type II AT deficiency. They were all heterozygous for the c.1346 T > A (Leu417Gln) mutation. In pedigree 3, the AT:A and AT:Ag of proband (III2), mother (II3), and uncle (II1) showed a simultaneous decrease in AT:A and AT:Ag, with values of 38%, 44 mg/dl, 50%, 48 mg/dl, 40%, and 54 mg/dl, respectively, indicating a type I AT deficiency. Sequencing analysis revealed a heterozygous missense mutation c.851 T > C (p.Met252Thr) in the above three members of pedigree 3. The sequencing diagram is shown in Figure 2. Three corresponding sites in 100 healthy subjects were sequenced and excluded genetic polymorphisms. The FV‐Leiden and prothrombin G20210A of common thrombogenic gene mutations were screened and showed none existing in three probands and their family members.\nPhenotype results of three pedigrees with inherited AT deficiency\nSequencing results: (A) c. 1358 T wild type, (B) c. 1358 T > C heterozygous mutation of proband 1; (C) c. 1346 T wild type, (D) c.1346 T > A heterozygous mutation of proband 2; (E) c.851 T wild type, (F) c.851 T > C heterozygous mutation of proband 3\nThe results of plasma AT protein from Western blot tests are shown in Figure 3. The plasma AT molecular weight of the three probands was consistent with the molecular weight of the normal mixed plasma control, but the plasma AT levels of probands 1 and 3 were significantly lower than those of the normal control. There was no difference between AT content of proband 2 and the normal control.\nWB results of Plasma AT protein. The control was human mixed plasma. Results 1, 2, and 3 were proband 1, 2, and 3 respectively\n[SUBTITLE] Bioformatics [SUBSECTION] The bioinformatics prediction results are shown in Table 2. MutationTaster, PolyPhen‐2, PROVEAN, LRT, and SIFT all predicted the p.Met252Thr, and p.Ile421Thr mutations as “pathogenic and harmful”. For the p. Leu417Gln mutation, MutationTaster, PolyPhen‐2, and LRT suggested that it was harmful; SIFT and PROVEAN software labeled the mutations as “tolerable” and “neutral” respectively.\n18\n The ACMG guidelines classified p.Met252Thr and p.Ile421Thr mutations as “pathogenic,” and the p. Leu417Gln mutation as “likely pathogenic.” The conservation analysis results showed that the Met252, Leu417, and Ile421 were highly conserved among homologous species (Figure 4). In the p.Ile421Thr wild type, when Ile421 was replaced by Thr421, the original hydrogen bond was unchanged, but new hydrogen bonds were formed between Thr421 and Ile412, and Thr421 and Ile422, as shown in Figure 5. In the wild‐type AT protein, the main chain of Leu417 formed two hydrogen bonds with the side chain of Glu414 and Ser52. In the wild‐type AT model, there were two hydrogen bonds between the main chain of Met252 residues and Met320. When the Met252 was substituted by Thr, the original hydrogen bond remained, and an additional hydrogen bond was formed with Met320 (as shown in Figure 5).\nBioinformatics prediction results of mutations\n\nNote: Meanings of scores are as follows: Mutation Taster: a probability close to 1 indicates a high predictive value; PolyPhen‐2: scores are evaluated as 0.000 (most probably benign) to 1.000 (most probably damaging); PROVEAN: the predefined threshold of score is 2.5, the score 2.5 or less (deleterious), greater than 2.5 (neutral); LRT: scores 0.01 or less (deleterious), greater than 0.01 (benign); SIFT: scores range from 0 to 1, 0.05 or less (damaging), greater than 0.05 (tolerated).\nConservation analysis of homologous species of Met252, Leu417, and Ile421\nMutation model analysis (A) p.Ile421Thr wild type, (B) p.Ile421Thr mutant type, (C) p.Met252Thr wild type, (D) p.Met252Thr mutant type. Hydrogen bonds are indicated by green dotted lines.\nThe bioinformatics prediction results are shown in Table 2. MutationTaster, PolyPhen‐2, PROVEAN, LRT, and SIFT all predicted the p.Met252Thr, and p.Ile421Thr mutations as “pathogenic and harmful”. For the p. Leu417Gln mutation, MutationTaster, PolyPhen‐2, and LRT suggested that it was harmful; SIFT and PROVEAN software labeled the mutations as “tolerable” and “neutral” respectively.\n18\n The ACMG guidelines classified p.Met252Thr and p.Ile421Thr mutations as “pathogenic,” and the p. Leu417Gln mutation as “likely pathogenic.” The conservation analysis results showed that the Met252, Leu417, and Ile421 were highly conserved among homologous species (Figure 4). In the p.Ile421Thr wild type, when Ile421 was replaced by Thr421, the original hydrogen bond was unchanged, but new hydrogen bonds were formed between Thr421 and Ile412, and Thr421 and Ile422, as shown in Figure 5. In the wild‐type AT protein, the main chain of Leu417 formed two hydrogen bonds with the side chain of Glu414 and Ser52. In the wild‐type AT model, there were two hydrogen bonds between the main chain of Met252 residues and Met320. When the Met252 was substituted by Thr, the original hydrogen bond remained, and an additional hydrogen bond was formed with Met320 (as shown in Figure 5).\nBioinformatics prediction results of mutations\n\nNote: Meanings of scores are as follows: Mutation Taster: a probability close to 1 indicates a high predictive value; PolyPhen‐2: scores are evaluated as 0.000 (most probably benign) to 1.000 (most probably damaging); PROVEAN: the predefined threshold of score is 2.5, the score 2.5 or less (deleterious), greater than 2.5 (neutral); LRT: scores 0.01 or less (deleterious), greater than 0.01 (benign); SIFT: scores range from 0 to 1, 0.05 or less (damaging), greater than 0.05 (tolerated).\nConservation analysis of homologous species of Met252, Leu417, and Ile421\nMutation model analysis (A) p.Ile421Thr wild type, (B) p.Ile421Thr mutant type, (C) p.Met252Thr wild type, (D) p.Met252Thr mutant type. Hydrogen bonds are indicated by green dotted lines.\n[SUBTITLE] Transfection and expression [SUBSECTION] The AT‐M252T, AT‐I421T, and AT‐L417Q mutant plasmids were successfully constructed by site‐directed mutation without other mutation sites verified by sequencing. The wild type and each mutant plasmid were observed under normal field and fluorescence field after 48 h transfection. The results are shown in Figures S1–S4. The wild‐type and each mutant plasmid were successfully transfected. The efficiency was approximately 85%.\nThe mRNA expression level of each group was detected by the fluorescence quantitative RT‐PCR method; the results are shown in Figure S2. AT gene expression of the AT‐WT plasmid was 100%, AT‐M252T, AT‐I421T, and AT‐L417Q mutant were 94.65%, 94.62%, and 107.6%, respectively. There was no significant difference between these and AT‐WT in mRNA expression. The nontransfected control and samples transfected with empty vector did not express AT mRNA.\nThe AT:Ag of the wild‐type and each mutant plasmid group were measured by ELISA. The cell culture supernatant and lysate of the wild‐type group were defined as 100%, and the calculated results were statistically analyzed by GraphPad 8.0 software as shown in Figure S3. The AT expression levels of AT‐M252T and AT‐I421T mutants in the cell culture supernatant were lower than those of wild‐type WT, while the expression levels of AT‐M252T and AT‐I421T mutants in the cell lysate were higher than that of wild‐type WT. The expression of AT of AT‐L417Q mutant in the cell culture supernatant and lysate was equivalent to that of the wild type, and there was no statistical difference.\nSemi‐quantitative analyses of AT proteins were performed by Western blot in the cell lysate and culture supernatant. The results are shown in Figure S4, respectively. AT protein of AT‐M252T and AT‐I421T mutants expressed significantly more than the wild‐type in the cell lysate. AT protein expressions of AT‐M252T and AT‐I421T mutant were significantly reduced, compared with the wild type. AT protein expression of AT‐L417Q mutant levels were not much different from wild‐type cells in lysate and culture supernatant. The Western blot results of the three mutants were consistent with the ELISA results.\nThe AT‐M252T, AT‐I421T, and AT‐L417Q mutant plasmids were successfully constructed by site‐directed mutation without other mutation sites verified by sequencing. The wild type and each mutant plasmid were observed under normal field and fluorescence field after 48 h transfection. The results are shown in Figures S1–S4. The wild‐type and each mutant plasmid were successfully transfected. The efficiency was approximately 85%.\nThe mRNA expression level of each group was detected by the fluorescence quantitative RT‐PCR method; the results are shown in Figure S2. AT gene expression of the AT‐WT plasmid was 100%, AT‐M252T, AT‐I421T, and AT‐L417Q mutant were 94.65%, 94.62%, and 107.6%, respectively. There was no significant difference between these and AT‐WT in mRNA expression. The nontransfected control and samples transfected with empty vector did not express AT mRNA.\nThe AT:Ag of the wild‐type and each mutant plasmid group were measured by ELISA. The cell culture supernatant and lysate of the wild‐type group were defined as 100%, and the calculated results were statistically analyzed by GraphPad 8.0 software as shown in Figure S3. The AT expression levels of AT‐M252T and AT‐I421T mutants in the cell culture supernatant were lower than those of wild‐type WT, while the expression levels of AT‐M252T and AT‐I421T mutants in the cell lysate were higher than that of wild‐type WT. The expression of AT of AT‐L417Q mutant in the cell culture supernatant and lysate was equivalent to that of the wild type, and there was no statistical difference.\nSemi‐quantitative analyses of AT proteins were performed by Western blot in the cell lysate and culture supernatant. The results are shown in Figure S4, respectively. AT protein of AT‐M252T and AT‐I421T mutants expressed significantly more than the wild‐type in the cell lysate. AT protein expressions of AT‐M252T and AT‐I421T mutant were significantly reduced, compared with the wild type. AT protein expression of AT‐L417Q mutant levels were not much different from wild‐type cells in lysate and culture supernatant. The Western blot results of the three mutants were consistent with the ELISA results.", "In pedigree 1, the AT:A and AT:Ag of proband (II3), father (I1), and daughter (III1) all decreased; the values were 49%, 52 mg/dl, 48%, 42 mg/dl 40%, and 45 mg/dl, respectively, representing a type I AT deficiency. The phenotype results of the three pedigrees with inherited AT deficiency is shown in Table 1. Genetic analysis found that the above three members all carry the c.1358 T > C (Ile421Thr) heterozygous mutation in SERPINC1. In pedigree 2, the AT:A of proband (I1), and the eldest son (II2), second daughter (II4), and granddaughter (III3) decreased, with values 32%, 43%, 52%, and 48%, respectively; however, AT:Ag was within normal range, indicating a type II AT deficiency. They were all heterozygous for the c.1346 T > A (Leu417Gln) mutation. In pedigree 3, the AT:A and AT:Ag of proband (III2), mother (II3), and uncle (II1) showed a simultaneous decrease in AT:A and AT:Ag, with values of 38%, 44 mg/dl, 50%, 48 mg/dl, 40%, and 54 mg/dl, respectively, indicating a type I AT deficiency. Sequencing analysis revealed a heterozygous missense mutation c.851 T > C (p.Met252Thr) in the above three members of pedigree 3. The sequencing diagram is shown in Figure 2. Three corresponding sites in 100 healthy subjects were sequenced and excluded genetic polymorphisms. The FV‐Leiden and prothrombin G20210A of common thrombogenic gene mutations were screened and showed none existing in three probands and their family members.\nPhenotype results of three pedigrees with inherited AT deficiency\nSequencing results: (A) c. 1358 T wild type, (B) c. 1358 T > C heterozygous mutation of proband 1; (C) c. 1346 T wild type, (D) c.1346 T > A heterozygous mutation of proband 2; (E) c.851 T wild type, (F) c.851 T > C heterozygous mutation of proband 3\nThe results of plasma AT protein from Western blot tests are shown in Figure 3. The plasma AT molecular weight of the three probands was consistent with the molecular weight of the normal mixed plasma control, but the plasma AT levels of probands 1 and 3 were significantly lower than those of the normal control. There was no difference between AT content of proband 2 and the normal control.\nWB results of Plasma AT protein. The control was human mixed plasma. Results 1, 2, and 3 were proband 1, 2, and 3 respectively", "The bioinformatics prediction results are shown in Table 2. MutationTaster, PolyPhen‐2, PROVEAN, LRT, and SIFT all predicted the p.Met252Thr, and p.Ile421Thr mutations as “pathogenic and harmful”. For the p. Leu417Gln mutation, MutationTaster, PolyPhen‐2, and LRT suggested that it was harmful; SIFT and PROVEAN software labeled the mutations as “tolerable” and “neutral” respectively.\n18\n The ACMG guidelines classified p.Met252Thr and p.Ile421Thr mutations as “pathogenic,” and the p. Leu417Gln mutation as “likely pathogenic.” The conservation analysis results showed that the Met252, Leu417, and Ile421 were highly conserved among homologous species (Figure 4). In the p.Ile421Thr wild type, when Ile421 was replaced by Thr421, the original hydrogen bond was unchanged, but new hydrogen bonds were formed between Thr421 and Ile412, and Thr421 and Ile422, as shown in Figure 5. In the wild‐type AT protein, the main chain of Leu417 formed two hydrogen bonds with the side chain of Glu414 and Ser52. In the wild‐type AT model, there were two hydrogen bonds between the main chain of Met252 residues and Met320. When the Met252 was substituted by Thr, the original hydrogen bond remained, and an additional hydrogen bond was formed with Met320 (as shown in Figure 5).\nBioinformatics prediction results of mutations\n\nNote: Meanings of scores are as follows: Mutation Taster: a probability close to 1 indicates a high predictive value; PolyPhen‐2: scores are evaluated as 0.000 (most probably benign) to 1.000 (most probably damaging); PROVEAN: the predefined threshold of score is 2.5, the score 2.5 or less (deleterious), greater than 2.5 (neutral); LRT: scores 0.01 or less (deleterious), greater than 0.01 (benign); SIFT: scores range from 0 to 1, 0.05 or less (damaging), greater than 0.05 (tolerated).\nConservation analysis of homologous species of Met252, Leu417, and Ile421\nMutation model analysis (A) p.Ile421Thr wild type, (B) p.Ile421Thr mutant type, (C) p.Met252Thr wild type, (D) p.Met252Thr mutant type. Hydrogen bonds are indicated by green dotted lines.", "The AT‐M252T, AT‐I421T, and AT‐L417Q mutant plasmids were successfully constructed by site‐directed mutation without other mutation sites verified by sequencing. The wild type and each mutant plasmid were observed under normal field and fluorescence field after 48 h transfection. The results are shown in Figures S1–S4. The wild‐type and each mutant plasmid were successfully transfected. The efficiency was approximately 85%.\nThe mRNA expression level of each group was detected by the fluorescence quantitative RT‐PCR method; the results are shown in Figure S2. AT gene expression of the AT‐WT plasmid was 100%, AT‐M252T, AT‐I421T, and AT‐L417Q mutant were 94.65%, 94.62%, and 107.6%, respectively. There was no significant difference between these and AT‐WT in mRNA expression. The nontransfected control and samples transfected with empty vector did not express AT mRNA.\nThe AT:Ag of the wild‐type and each mutant plasmid group were measured by ELISA. The cell culture supernatant and lysate of the wild‐type group were defined as 100%, and the calculated results were statistically analyzed by GraphPad 8.0 software as shown in Figure S3. The AT expression levels of AT‐M252T and AT‐I421T mutants in the cell culture supernatant were lower than those of wild‐type WT, while the expression levels of AT‐M252T and AT‐I421T mutants in the cell lysate were higher than that of wild‐type WT. The expression of AT of AT‐L417Q mutant in the cell culture supernatant and lysate was equivalent to that of the wild type, and there was no statistical difference.\nSemi‐quantitative analyses of AT proteins were performed by Western blot in the cell lysate and culture supernatant. The results are shown in Figure S4, respectively. AT protein of AT‐M252T and AT‐I421T mutants expressed significantly more than the wild‐type in the cell lysate. AT protein expressions of AT‐M252T and AT‐I421T mutant were significantly reduced, compared with the wild type. AT protein expression of AT‐L417Q mutant levels were not much different from wild‐type cells in lysate and culture supernatant. The Western blot results of the three mutants were consistent with the ELISA results.", "Thrombophilia refers to a high thromboembolic tendency caused by certain risk factors, including hereditary and acquired factors. Genetic factors mainly include congenital defects of anticoagulant proteins such as AT, PC, and PS, mutations of prothrombin G20210A, and coagulation factor V Leiden.\n19\n Acquired risk factors include, but are not limited to, oral contraceptives, trauma, immobilization, and surgery, and other factors.\n20\n In this article, we presented 3 families with inherited AT deficiency and found 3 heterozygous missense variants, namely c.1358 T > C (p.Ile421Thr), c.1346 T > A (p.Leu417Gln), and c.851 T > C (p.Met252Thr). However, these three variants were not found in 100 healthy controls, thus excluding genetic polymorphisms.\nProband 1 had p.Ile421Thr mutation and was diagnosed as a type I Inherited AT deficiency. The results of conservation analysis showed that the Ile421 site was highly conserved in homologous species, suggesting that this site plays an important role in the structure or function of AT protein. When le421 was replaced by Thr421, intermolecular hydrophobic force and hydrogen bonds changed, and the protein structure might have been affected, which was in accordance with the results of bioinformatics analysis. The p.Ile421Thr mutation has only been reported since 1994 by overseas researchers.\n20\n, \n21\n The overseas case presented recurrent venous thrombosis with a family history of thrombosis, and developed DVT and PE after the first pregnancy at the age of 27 years, with similar symptoms after the second and third pregnancies. Similar to our study, proband 1 was 27 years old and had venous thrombosis during puerperium, indicating that the p.Ile421Thr mutation has a higher risk of thrombosis, and when combined with acquired risk factors such as pregnancy and surgery, the p.Ile421Thr mutation has early onset characteristics. The p.Leu417Gln mutation was found in all four members in the second family (proband 2, his son, daughter, and granddaughter) who carried this heterozygous mutation; the details of which had been reported by our group.\n18\n All members in the third family including the proband 3, her mother, and uncle carried the p.Met252Thr heterozygous mutation. Western blot results showed that proband 3's plasma AT level was lower than the normal controls, in accordance with the phenotypic results, and diagnosed as a type I hereditary AT deficiency.\n22\n The bioinformatics software predicted that it would be pathogenic and that the protein structure may be affected. The hydrophobic interaction force and the number of hydrogen bonds between Thr252 and surrounding amino acids had been changed, which had an influence on the structural stability. In family 3, all three members carrying the heterozygous mutation of p.Met252Thr had a history of venous thrombosis; the wild‐type members were asymptomatic, indicating that the mutation had a high correlation with thrombosis. Patients in a South Korean study with p.Met252Arg mutation both had decreased AT activity, antigen level, and venous thrombosis.\n9\n We speculated that Met252 might be a hot spot mutation of the AT gene and carries a higher risk of thrombosis.\nIn order to further investigate the reasons for the reduction in AT levels caused by these three mutations, we ligated the coding sequence (CDS) of the AT gene into the eukaryotic expression vector to construct the AT‐WT plasmid. RT‐PCR results showed that the expression of mutant AT mRNA was not significantly different from that of the wild type, indicating that these three mutations had no significant effect on AT transcription. Western blot results showed that levels of AT protein of AT‐M252T and AT‐I421T in the cell lysates were significantly higher than that of AT‐WT, but the AT levels of both in the culture supernatant were significantly lower than that of AT‐WT, showing that the mutations of p.Met252Thr and p.Ile421Thr affected the normal secretion of AT protein from the inside to the outside of the cell, resulting in AT being retained in the cell, and causing the AT level to decrease. ELISA results showed the same findings, and further verified the retention of AT protein in the cell. Mutations such as AT‐Trp225Cys, AT‐Ala404Asp\n,23\n and AT‐Ala404Thr\n24\n had previously been reported. In vitro expression experiments showed that these mutations also led to impaired secretion of AT protein, type I inherited AT deficiency. For the p.Leu417Gln mutation, both ELISA and Western blot showed that the AT level expressed by the AT‐L417Q mutant was not significantly different from that of the wild type, which was consistent with the phenotypic test result of proband 2. The AT activity level of the patient was reduced, indicating that dysfunctional AT molecules may be generated by the mutation, thus impairing or damaging anticoagulant activity.\n25\n\n\nIn this article, we confirmed that pathogenic molecular mechanisms of p.Met252Thr, p.Ile421Thr, and p.Leu417Gln mutations led to inherited AT defects in three families. However, the genotype is not completely consistent with the clinical phenotype. Among the 25 people in the above‐mentioned three families, ten people carry AT mutations, but only five people displayed clinical manifestations; three probands had venous thrombosis, but only family 3 had a family history of hemorrhagic thrombosis. Family members in Family 1 and Family 2 who carried the same mutations as the proband had no thrombosis or related clinical manifestations, suggesting that patients with simple AT deficiency might not always develop thrombus. Therefore, it is necessary to carry out routine and specific antithrombotic prevention and treatment for such patients.", "We reported three heterozygous missense mutations in the SERPINC1 gene, namely c.1358 T > C (p.Ile421Thr), c.1346 T > A (p.Leu417Gln), and c.851 T > C (p.Met252Thr). The pathogenic mechanisms of the three mutations were preliminarily explored through bioinformatics, protein modeling, and in vitro expression analysis, which enriched the AT genetic mutation database.", "MSW and ML designed experiments, analyzed data, wrote the article, and supervised the project. SQL and STJ performed molecular modeling. SQL and YHJ designed experiments, conducted genetic analysis, and coagulation assays. All authors approved the final version of the article.", "The authors declare no conflicts of interests.", "Our study was approved by the Ethics Committee of the First Affiliated Hospital of Wenzhou Medical University (China).", "\nFigures S1–S4\n\nClick here for additional data file." ]
[ null, "materials-and-methods", null, null, null, null, null, "results", null, null, null, "discussion", "conclusions", null, "COI-statement", null, "supplementary-material" ]
[ "antithrombin", "gene mutation", "in vitro expression", "inherited AT deficiency", "thromboembolism" ]
Serum hsa_circ_0000615 is a prognostic biomarker of sorafenib resistance in hepatocellular carcinoma.
36268976
Circular RNAs (circRNAs) can shape tumor progression and chemoresistance. How specific circRNAs shape hepatocellular carcinoma (HCC) chemoresistance, however, remains to be fully elucidated.
BACKGROUND
In total, serum samples were collected from 202 HCC patients that had completed four sorafenib chemotherapy cycles. Serum hsa_circ_0000615 levels in these patients were quantified via quantitative real-time polymerase chain reaction (qRT-PCR), with demographic details and survival outcomes being recorded for subsequent analyses.
METHODS
We found hsa_circ_0000615 to be significantly upregulated in chemoresistant HCC patients relative to chemosensitive patients, with such upregulation being positively correlated with disease stage. Moreover, the area under the curve (AUC) value for hsa_circ_0000615 was moderately good, and high levels of hsa_circ_0000615 expression were associated with shorter overall survival among chemoresistant HCC patients.
RESULTS
Our results highlight hsa_circ_0000615 as a promising driver of sorafenib resistance in HCC patients, highlighting it as a promising target for the treatment of this deadly cancer type.
CONCLUSION
[ "Humans", "Carcinoma, Hepatocellular", "RNA, Circular", "Sorafenib", "Liver Neoplasms", "Prognosis", "Biomarkers, Tumor" ]
9701853
null
null
null
null
RESULTS
[SUBTITLE] Hepatocellular carcinoma patients exhibit serum hsa_circ_0000615 upregulation [SUBSECTION] We began by assessing the levels of hsa_circ_0000615 in control and sorafenib‐resistant HCC cells. The sorafenib‐resistant Hep G2/sorafenib and Huh 7/sorafenib cell lines exhibited marked upregulation of this circRNA relative to corresponding parental cell lines (Figure 1A). To explore the potential utility of hsa_circ_0000615 as a biomarker of chemoresistance, we then assessed the levels of this circRNA in serum samples from 202 HCC patients and 202 healthy controls. HCC patients exhibited significantly elevated serum hsa_circ_0000615 levels compared with healthy controls (Figure 1B). Moreover, hsa_circ_0000615 expression levels were higher in sorafenib‐resistant patients (n = 122) relative to those in sorafenib‐sensitive individuals (n = 80) (Figure 1C). As such, hsa_circ_0000615 offers potential value as an HCC chemotherapy biomarker. Hepatocellular carcinoma (HCC) patient serum samples exhibit hsa_circ_0000615 upregulation. (A). Hsa_circ_0000615 levels were significantly increased in sorafenib‐resistant HCC cells. (B). Serum hsa_circ_0000615 levels were higher in HCC patients. (C). sorafenib‐resistant patients (n = 122) exhibited higher levels of hsa_circ_0000615 expression compared with sorafenib‐sensitive patients in the before treatment and after treatment (n = 80). *p < .05. We began by assessing the levels of hsa_circ_0000615 in control and sorafenib‐resistant HCC cells. The sorafenib‐resistant Hep G2/sorafenib and Huh 7/sorafenib cell lines exhibited marked upregulation of this circRNA relative to corresponding parental cell lines (Figure 1A). To explore the potential utility of hsa_circ_0000615 as a biomarker of chemoresistance, we then assessed the levels of this circRNA in serum samples from 202 HCC patients and 202 healthy controls. HCC patients exhibited significantly elevated serum hsa_circ_0000615 levels compared with healthy controls (Figure 1B). Moreover, hsa_circ_0000615 expression levels were higher in sorafenib‐resistant patients (n = 122) relative to those in sorafenib‐sensitive individuals (n = 80) (Figure 1C). As such, hsa_circ_0000615 offers potential value as an HCC chemotherapy biomarker. Hepatocellular carcinoma (HCC) patient serum samples exhibit hsa_circ_0000615 upregulation. (A). Hsa_circ_0000615 levels were significantly increased in sorafenib‐resistant HCC cells. (B). Serum hsa_circ_0000615 levels were higher in HCC patients. (C). sorafenib‐resistant patients (n = 122) exhibited higher levels of hsa_circ_0000615 expression compared with sorafenib‐sensitive patients in the before treatment and after treatment (n = 80). *p < .05. [SUBTITLE] Hsa_circ_0000615 levels are related to clinical features in HCC patients [SUBSECTION] Next, we stratified HCC patients into those with high and low levels of serum hsa_circ_0000615 based on the mean expression of this circRNA in this cohort and compared clinical features between these two patient groups. Chi‐squared analyses revealed that hsa_circ_0000615 expression was associated with clinical stage, TNM stage, and lymph node metastasis (Table 1), but was unrelated to age, sex, or histological grade. Kaplan–Meier analyses indicated that patients exhibiting higher levels of hsa_circ_0000615 expression presented with shorter OS relative to patients expressing low hsa_circ_0000615 levels (Figure 2). Correlations between hsa_circ_0000615 levels and hepatocellular carcinoma patient clinicopathological features Hsa_circ_0000615 levels are linked with hepatocellular carcinoma patient survival. Patients exhibiting higher hsa_circ_0000615 expression levels exhibited prolonged overall survival compared with patients with lower levels of this circRNA. Next, we stratified HCC patients into those with high and low levels of serum hsa_circ_0000615 based on the mean expression of this circRNA in this cohort and compared clinical features between these two patient groups. Chi‐squared analyses revealed that hsa_circ_0000615 expression was associated with clinical stage, TNM stage, and lymph node metastasis (Table 1), but was unrelated to age, sex, or histological grade. Kaplan–Meier analyses indicated that patients exhibiting higher levels of hsa_circ_0000615 expression presented with shorter OS relative to patients expressing low hsa_circ_0000615 levels (Figure 2). Correlations between hsa_circ_0000615 levels and hepatocellular carcinoma patient clinicopathological features Hsa_circ_0000615 levels are linked with hepatocellular carcinoma patient survival. Patients exhibiting higher hsa_circ_0000615 expression levels exhibited prolonged overall survival compared with patients with lower levels of this circRNA. [SUBTITLE] Hsa_circ_0000615 is associated with poor chemoresistant hepatocellular carcinoma patient prognosis [SUBSECTION] Through Kaplan–Meier analyses and log‐rank tests, we found chemoresistant HCC patients to exhibit significantly reduced OS and progression‐free survival (PFS) compared with chemosensitive patients (Figure 3). Through Cox proportional hazards regression analyses, we determined that clinical stage, chemoresistance, TNM stage, lymph node metastasis, and hsa_circ_0000615 levels were associated with patient PFS (Table 2) and OS (Table 3), highlighting hsa_circ_0000615 as a promising independent predictor of chemoresistant HCC patient survival. Hsa_circ_0000615 levels were significantly linked to poor outcomes in chemoresistant hepatocellular carcinoma (HCC) patients. Chemoresistant HCC patients exhibited significantly decreased progression‐free survival (A) and overall survival (B) relative to chemosensitive patients. Univariate and multivariate analyses of hepatocellular carcinoma patient progression‐free survival Univariate and multivariate analyses of hepatocellular carcinoma patient overall survival Through Kaplan–Meier analyses and log‐rank tests, we found chemoresistant HCC patients to exhibit significantly reduced OS and progression‐free survival (PFS) compared with chemosensitive patients (Figure 3). Through Cox proportional hazards regression analyses, we determined that clinical stage, chemoresistance, TNM stage, lymph node metastasis, and hsa_circ_0000615 levels were associated with patient PFS (Table 2) and OS (Table 3), highlighting hsa_circ_0000615 as a promising independent predictor of chemoresistant HCC patient survival. Hsa_circ_0000615 levels were significantly linked to poor outcomes in chemoresistant hepatocellular carcinoma (HCC) patients. Chemoresistant HCC patients exhibited significantly decreased progression‐free survival (A) and overall survival (B) relative to chemosensitive patients. Univariate and multivariate analyses of hepatocellular carcinoma patient progression‐free survival Univariate and multivariate analyses of hepatocellular carcinoma patient overall survival [SUBTITLE] Serum hsa_circ_0000615 levels offer diagnostic utility for the detection of hepatocellular carcinoma chemoresistance [SUBSECTION] Previous studies have shown that circRNAs show excellent potential diagnostic utility in various cancers, such as, breast cancer, 20 gastric cancer, 21 and HCC. 22 To assess the potential diagnostic utility of serum hsa_circ_0000615 in patients with HCC, the area under the receiver operating characteristic (ROC) curve (AUC) was determined and found to be 0.9238 (95% CI, 0.8915–0.956, Figure 4, p < .0001), consistent with the value of serum hsa_circ_0000615 as a biomarker capable of differentiating between HCC patients and healthy controls. Serum hsa_circ_0000615 levels offer diagnostic value as predictors of hepatocellular carcinoma (HCC) patient chemoresistance. Receiver‐operating characteristic curves were used to differentiate between chemoresistant HCC patients before and after therapy. Previous studies have shown that circRNAs show excellent potential diagnostic utility in various cancers, such as, breast cancer, 20 gastric cancer, 21 and HCC. 22 To assess the potential diagnostic utility of serum hsa_circ_0000615 in patients with HCC, the area under the receiver operating characteristic (ROC) curve (AUC) was determined and found to be 0.9238 (95% CI, 0.8915–0.956, Figure 4, p < .0001), consistent with the value of serum hsa_circ_0000615 as a biomarker capable of differentiating between HCC patients and healthy controls. Serum hsa_circ_0000615 levels offer diagnostic value as predictors of hepatocellular carcinoma (HCC) patient chemoresistance. Receiver‐operating characteristic curves were used to differentiate between chemoresistant HCC patients before and after therapy.
CONCLUSIONS
In summary, we herein found hsa_circ_0000615 upregulation to be prominent within serum samples from HCC patients, with such upregulation being significantly more pronounced in samples from chemosensitive patients relative to chemoresistant patients. As such, hsa_circ_0000615 is a promising target that warrants further study in an effort to understand the mechanistic basis for HCC patient chemoresistance.
[ "INTRODUCTION", "Cell culture and clinical samples", "Quantitative real‐time polymerase chain reaction", "Statistical analysis", "Hepatocellular carcinoma patients exhibit serum hsa_circ_0000615 upregulation", "Hsa_circ_0000615 levels are related to clinical features in HCC patients", "Hsa_circ_0000615 is associated with poor chemoresistant hepatocellular carcinoma patient prognosis", "Serum hsa_circ_0000615 levels offer diagnostic utility for the detection of hepatocellular carcinoma chemoresistance" ]
[ "Hepatocellular carcinoma (HCC) is the leading cause of cancer‐related death worldwide.\n1\n In most cases, long‐term sorafenib administration is associated with the onset of chemoresistance. As such, there is a clear need to identify the mechanistic basis for sorafenib resistance and to design novel approaches to effectively treat this cancer type.\nCircular RNAs (circRNAs) are a covalently closed looping structure that renders them resistant to degradation and more stable than linear RNAs.\n2\n, \n3\n, \n4\n A growing body of evidence suggests that circRNAs can regulate a range of cancers and other important diseases by influencing cellular proliferation, survival, migration, glucose metabolism, and differentiation.\n5\n, \n6\n, \n7\n, \n8\n, \n9\n As such, circRNAs offer great promise as diagnostic biomarkers or therapeutic targets in cancer patients and individuals with other conditions.\n10\n\n\nRecently, circRNAs have been found to be dysregulated in HCC and to play an important functional role in this pathological context.\n11\n, \n12\n, \n13\n The recently identified circRNA hsa_circ_0000615 has been found to play an oncogenic role in prostate,\n14\n breast,\n15\n gastric,\n16\n and colorectal cancers.\n17\n Moreover, hsa_circ_0000615 has been found to promote HCC cell migration, invasion, stemness, and proliferation.\n18\n, \n19\n How hsa_circ_0000615 functions in the context of tumor chemoresistance, however, remains to be defined.\nHerein, we explored the expression of hsa_circ_0000615 in HCC patient serum and its relationship with patient clinical findings. Overall, we found that sorafenib‐resistant HCC patients exhibited hsa_circ_0000615 upregulation that was related to poorer overall survival (OS) outcomes. Moreover, hsa_circ_0000615 exhibited reasonably good area under the ROC curve (AUC) values, suggesting that it may offer value as a novel prognostic biomarker of sorafenib‐resistant HCC.", "Human Hep G2 and Huh 7 were grown in DMEM (Invitrogen, NY, USA). Hep G2/sorafenib and Huh 7/sorafenib cell lines were established by maintaining Hep G2 and Huh 7 cells at 1 mmol/L sorafenib and gradually increasing it at a rate of 0.5 mmol/L per month (up to 5 mmol/L) more than 10‐month. Serum samples from 202 HCC patients and 202 healthy controls were obtained from the First Affiliated Hospital of Bengbu Medical College. This study was approved by the Ethics Committee of the First Affiliated Hospital of Bengbu Medical College, with all patients having provided written informed consent.", "An RNA Isolation Kit (Vazyme Biotech, Nanjing, China) was used to extract total RNA from 500 μl of patient serum, after which a Prime Script RT reagent Kit (Takara, Dalian, China) was used for cDNA synthesis. Prepared cDNA was then used as input for qPCR reactions performed with SYBR Green (Takara). The U6 small nuclear B noncoding RNA (U6) was used to normalize expression values via the 2−ΔΔCt method, with primers used being as follows: hsa_circ_0000615: F 5′–CAGCGCTATCCTTTGGGA–3′, R 5′–GACCTGCCACATTGGTCAGTA–3′; U6: F 5′–TGCGGGTGCTCGCTTCGGCAGC–3′, R 5′–GTGCAGGGTCCGAGGT–3′.", "Data are means ± standard deviation (SD) and were compared via Student's t tests using GraphPad Prism 7. The Kaplan–Meier method was used for survival analyses, with p < .05 as the threshold of significance.", "We began by assessing the levels of hsa_circ_0000615 in control and sorafenib‐resistant HCC cells. The sorafenib‐resistant Hep G2/sorafenib and Huh 7/sorafenib cell lines exhibited marked upregulation of this circRNA relative to corresponding parental cell lines (Figure 1A). To explore the potential utility of hsa_circ_0000615 as a biomarker of chemoresistance, we then assessed the levels of this circRNA in serum samples from 202 HCC patients and 202 healthy controls. HCC patients exhibited significantly elevated serum hsa_circ_0000615 levels compared with healthy controls (Figure 1B). Moreover, hsa_circ_0000615 expression levels were higher in sorafenib‐resistant patients (n = 122) relative to those in sorafenib‐sensitive individuals (n = 80) (Figure 1C). As such, hsa_circ_0000615 offers potential value as an HCC chemotherapy biomarker.\nHepatocellular carcinoma (HCC) patient serum samples exhibit hsa_circ_0000615 upregulation. (A). Hsa_circ_0000615 levels were significantly increased in sorafenib‐resistant HCC cells. (B). Serum hsa_circ_0000615 levels were higher in HCC patients. (C). sorafenib‐resistant patients (n = 122) exhibited higher levels of hsa_circ_0000615 expression compared with sorafenib‐sensitive patients in the before treatment and after treatment (n = 80). *p < .05.", "Next, we stratified HCC patients into those with high and low levels of serum hsa_circ_0000615 based on the mean expression of this circRNA in this cohort and compared clinical features between these two patient groups. Chi‐squared analyses revealed that hsa_circ_0000615 expression was associated with clinical stage, TNM stage, and lymph node metastasis (Table 1), but was unrelated to age, sex, or histological grade. Kaplan–Meier analyses indicated that patients exhibiting higher levels of hsa_circ_0000615 expression presented with shorter OS relative to patients expressing low hsa_circ_0000615 levels (Figure 2).\nCorrelations between hsa_circ_0000615 levels and hepatocellular carcinoma patient clinicopathological features\nHsa_circ_0000615 levels are linked with hepatocellular carcinoma patient survival. Patients exhibiting higher hsa_circ_0000615 expression levels exhibited prolonged overall survival compared with patients with lower levels of this circRNA.", "Through Kaplan–Meier analyses and log‐rank tests, we found chemoresistant HCC patients to exhibit significantly reduced OS and progression‐free survival (PFS) compared with chemosensitive patients (Figure 3). Through Cox proportional hazards regression analyses, we determined that clinical stage, chemoresistance, TNM stage, lymph node metastasis, and hsa_circ_0000615 levels were associated with patient PFS (Table 2) and OS (Table 3), highlighting hsa_circ_0000615 as a promising independent predictor of chemoresistant HCC patient survival.\nHsa_circ_0000615 levels were significantly linked to poor outcomes in chemoresistant hepatocellular carcinoma (HCC) patients. Chemoresistant HCC patients exhibited significantly decreased progression‐free survival (A) and overall survival (B) relative to chemosensitive patients.\nUnivariate and multivariate analyses of hepatocellular carcinoma patient progression‐free survival\nUnivariate and multivariate analyses of hepatocellular carcinoma patient overall survival", "Previous studies have shown that circRNAs show excellent potential diagnostic utility in various cancers, such as, breast cancer,\n20\n gastric cancer,\n21\n and HCC.\n22\n To assess the potential diagnostic utility of serum hsa_circ_0000615 in patients with HCC, the area under the receiver operating characteristic (ROC) curve (AUC) was determined and found to be 0.9238 (95% CI, 0.8915–0.956, Figure 4, p < .0001), consistent with the value of serum hsa_circ_0000615 as a biomarker capable of differentiating between HCC patients and healthy controls.\nSerum hsa_circ_0000615 levels offer diagnostic value as predictors of hepatocellular carcinoma (HCC) patient chemoresistance. Receiver‐operating characteristic curves were used to differentiate between chemoresistant HCC patients before and after therapy." ]
[ null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "MATERIALS AND METHODS", "Cell culture and clinical samples", "Quantitative real‐time polymerase chain reaction", "Statistical analysis", "RESULTS", "Hepatocellular carcinoma patients exhibit serum hsa_circ_0000615 upregulation", "Hsa_circ_0000615 levels are related to clinical features in HCC patients", "Hsa_circ_0000615 is associated with poor chemoresistant hepatocellular carcinoma patient prognosis", "Serum hsa_circ_0000615 levels offer diagnostic utility for the detection of hepatocellular carcinoma chemoresistance", "DISCUSSION", "CONCLUSIONS", "CONFLICT OF INTEREST" ]
[ "Hepatocellular carcinoma (HCC) is the leading cause of cancer‐related death worldwide.\n1\n In most cases, long‐term sorafenib administration is associated with the onset of chemoresistance. As such, there is a clear need to identify the mechanistic basis for sorafenib resistance and to design novel approaches to effectively treat this cancer type.\nCircular RNAs (circRNAs) are a covalently closed looping structure that renders them resistant to degradation and more stable than linear RNAs.\n2\n, \n3\n, \n4\n A growing body of evidence suggests that circRNAs can regulate a range of cancers and other important diseases by influencing cellular proliferation, survival, migration, glucose metabolism, and differentiation.\n5\n, \n6\n, \n7\n, \n8\n, \n9\n As such, circRNAs offer great promise as diagnostic biomarkers or therapeutic targets in cancer patients and individuals with other conditions.\n10\n\n\nRecently, circRNAs have been found to be dysregulated in HCC and to play an important functional role in this pathological context.\n11\n, \n12\n, \n13\n The recently identified circRNA hsa_circ_0000615 has been found to play an oncogenic role in prostate,\n14\n breast,\n15\n gastric,\n16\n and colorectal cancers.\n17\n Moreover, hsa_circ_0000615 has been found to promote HCC cell migration, invasion, stemness, and proliferation.\n18\n, \n19\n How hsa_circ_0000615 functions in the context of tumor chemoresistance, however, remains to be defined.\nHerein, we explored the expression of hsa_circ_0000615 in HCC patient serum and its relationship with patient clinical findings. Overall, we found that sorafenib‐resistant HCC patients exhibited hsa_circ_0000615 upregulation that was related to poorer overall survival (OS) outcomes. Moreover, hsa_circ_0000615 exhibited reasonably good area under the ROC curve (AUC) values, suggesting that it may offer value as a novel prognostic biomarker of sorafenib‐resistant HCC.", "[SUBTITLE] Cell culture and clinical samples [SUBSECTION] Human Hep G2 and Huh 7 were grown in DMEM (Invitrogen, NY, USA). Hep G2/sorafenib and Huh 7/sorafenib cell lines were established by maintaining Hep G2 and Huh 7 cells at 1 mmol/L sorafenib and gradually increasing it at a rate of 0.5 mmol/L per month (up to 5 mmol/L) more than 10‐month. Serum samples from 202 HCC patients and 202 healthy controls were obtained from the First Affiliated Hospital of Bengbu Medical College. This study was approved by the Ethics Committee of the First Affiliated Hospital of Bengbu Medical College, with all patients having provided written informed consent.\nHuman Hep G2 and Huh 7 were grown in DMEM (Invitrogen, NY, USA). Hep G2/sorafenib and Huh 7/sorafenib cell lines were established by maintaining Hep G2 and Huh 7 cells at 1 mmol/L sorafenib and gradually increasing it at a rate of 0.5 mmol/L per month (up to 5 mmol/L) more than 10‐month. Serum samples from 202 HCC patients and 202 healthy controls were obtained from the First Affiliated Hospital of Bengbu Medical College. This study was approved by the Ethics Committee of the First Affiliated Hospital of Bengbu Medical College, with all patients having provided written informed consent.\n[SUBTITLE] Quantitative real‐time polymerase chain reaction [SUBSECTION] An RNA Isolation Kit (Vazyme Biotech, Nanjing, China) was used to extract total RNA from 500 μl of patient serum, after which a Prime Script RT reagent Kit (Takara, Dalian, China) was used for cDNA synthesis. Prepared cDNA was then used as input for qPCR reactions performed with SYBR Green (Takara). The U6 small nuclear B noncoding RNA (U6) was used to normalize expression values via the 2−ΔΔCt method, with primers used being as follows: hsa_circ_0000615: F 5′–CAGCGCTATCCTTTGGGA–3′, R 5′–GACCTGCCACATTGGTCAGTA–3′; U6: F 5′–TGCGGGTGCTCGCTTCGGCAGC–3′, R 5′–GTGCAGGGTCCGAGGT–3′.\nAn RNA Isolation Kit (Vazyme Biotech, Nanjing, China) was used to extract total RNA from 500 μl of patient serum, after which a Prime Script RT reagent Kit (Takara, Dalian, China) was used for cDNA synthesis. Prepared cDNA was then used as input for qPCR reactions performed with SYBR Green (Takara). The U6 small nuclear B noncoding RNA (U6) was used to normalize expression values via the 2−ΔΔCt method, with primers used being as follows: hsa_circ_0000615: F 5′–CAGCGCTATCCTTTGGGA–3′, R 5′–GACCTGCCACATTGGTCAGTA–3′; U6: F 5′–TGCGGGTGCTCGCTTCGGCAGC–3′, R 5′–GTGCAGGGTCCGAGGT–3′.\n[SUBTITLE] Statistical analysis [SUBSECTION] Data are means ± standard deviation (SD) and were compared via Student's t tests using GraphPad Prism 7. The Kaplan–Meier method was used for survival analyses, with p < .05 as the threshold of significance.\nData are means ± standard deviation (SD) and were compared via Student's t tests using GraphPad Prism 7. The Kaplan–Meier method was used for survival analyses, with p < .05 as the threshold of significance.", "Human Hep G2 and Huh 7 were grown in DMEM (Invitrogen, NY, USA). Hep G2/sorafenib and Huh 7/sorafenib cell lines were established by maintaining Hep G2 and Huh 7 cells at 1 mmol/L sorafenib and gradually increasing it at a rate of 0.5 mmol/L per month (up to 5 mmol/L) more than 10‐month. Serum samples from 202 HCC patients and 202 healthy controls were obtained from the First Affiliated Hospital of Bengbu Medical College. This study was approved by the Ethics Committee of the First Affiliated Hospital of Bengbu Medical College, with all patients having provided written informed consent.", "An RNA Isolation Kit (Vazyme Biotech, Nanjing, China) was used to extract total RNA from 500 μl of patient serum, after which a Prime Script RT reagent Kit (Takara, Dalian, China) was used for cDNA synthesis. Prepared cDNA was then used as input for qPCR reactions performed with SYBR Green (Takara). The U6 small nuclear B noncoding RNA (U6) was used to normalize expression values via the 2−ΔΔCt method, with primers used being as follows: hsa_circ_0000615: F 5′–CAGCGCTATCCTTTGGGA–3′, R 5′–GACCTGCCACATTGGTCAGTA–3′; U6: F 5′–TGCGGGTGCTCGCTTCGGCAGC–3′, R 5′–GTGCAGGGTCCGAGGT–3′.", "Data are means ± standard deviation (SD) and were compared via Student's t tests using GraphPad Prism 7. The Kaplan–Meier method was used for survival analyses, with p < .05 as the threshold of significance.", "[SUBTITLE] Hepatocellular carcinoma patients exhibit serum hsa_circ_0000615 upregulation [SUBSECTION] We began by assessing the levels of hsa_circ_0000615 in control and sorafenib‐resistant HCC cells. The sorafenib‐resistant Hep G2/sorafenib and Huh 7/sorafenib cell lines exhibited marked upregulation of this circRNA relative to corresponding parental cell lines (Figure 1A). To explore the potential utility of hsa_circ_0000615 as a biomarker of chemoresistance, we then assessed the levels of this circRNA in serum samples from 202 HCC patients and 202 healthy controls. HCC patients exhibited significantly elevated serum hsa_circ_0000615 levels compared with healthy controls (Figure 1B). Moreover, hsa_circ_0000615 expression levels were higher in sorafenib‐resistant patients (n = 122) relative to those in sorafenib‐sensitive individuals (n = 80) (Figure 1C). As such, hsa_circ_0000615 offers potential value as an HCC chemotherapy biomarker.\nHepatocellular carcinoma (HCC) patient serum samples exhibit hsa_circ_0000615 upregulation. (A). Hsa_circ_0000615 levels were significantly increased in sorafenib‐resistant HCC cells. (B). Serum hsa_circ_0000615 levels were higher in HCC patients. (C). sorafenib‐resistant patients (n = 122) exhibited higher levels of hsa_circ_0000615 expression compared with sorafenib‐sensitive patients in the before treatment and after treatment (n = 80). *p < .05.\nWe began by assessing the levels of hsa_circ_0000615 in control and sorafenib‐resistant HCC cells. The sorafenib‐resistant Hep G2/sorafenib and Huh 7/sorafenib cell lines exhibited marked upregulation of this circRNA relative to corresponding parental cell lines (Figure 1A). To explore the potential utility of hsa_circ_0000615 as a biomarker of chemoresistance, we then assessed the levels of this circRNA in serum samples from 202 HCC patients and 202 healthy controls. HCC patients exhibited significantly elevated serum hsa_circ_0000615 levels compared with healthy controls (Figure 1B). Moreover, hsa_circ_0000615 expression levels were higher in sorafenib‐resistant patients (n = 122) relative to those in sorafenib‐sensitive individuals (n = 80) (Figure 1C). As such, hsa_circ_0000615 offers potential value as an HCC chemotherapy biomarker.\nHepatocellular carcinoma (HCC) patient serum samples exhibit hsa_circ_0000615 upregulation. (A). Hsa_circ_0000615 levels were significantly increased in sorafenib‐resistant HCC cells. (B). Serum hsa_circ_0000615 levels were higher in HCC patients. (C). sorafenib‐resistant patients (n = 122) exhibited higher levels of hsa_circ_0000615 expression compared with sorafenib‐sensitive patients in the before treatment and after treatment (n = 80). *p < .05.\n[SUBTITLE] Hsa_circ_0000615 levels are related to clinical features in HCC patients [SUBSECTION] Next, we stratified HCC patients into those with high and low levels of serum hsa_circ_0000615 based on the mean expression of this circRNA in this cohort and compared clinical features between these two patient groups. Chi‐squared analyses revealed that hsa_circ_0000615 expression was associated with clinical stage, TNM stage, and lymph node metastasis (Table 1), but was unrelated to age, sex, or histological grade. Kaplan–Meier analyses indicated that patients exhibiting higher levels of hsa_circ_0000615 expression presented with shorter OS relative to patients expressing low hsa_circ_0000615 levels (Figure 2).\nCorrelations between hsa_circ_0000615 levels and hepatocellular carcinoma patient clinicopathological features\nHsa_circ_0000615 levels are linked with hepatocellular carcinoma patient survival. Patients exhibiting higher hsa_circ_0000615 expression levels exhibited prolonged overall survival compared with patients with lower levels of this circRNA.\nNext, we stratified HCC patients into those with high and low levels of serum hsa_circ_0000615 based on the mean expression of this circRNA in this cohort and compared clinical features between these two patient groups. Chi‐squared analyses revealed that hsa_circ_0000615 expression was associated with clinical stage, TNM stage, and lymph node metastasis (Table 1), but was unrelated to age, sex, or histological grade. Kaplan–Meier analyses indicated that patients exhibiting higher levels of hsa_circ_0000615 expression presented with shorter OS relative to patients expressing low hsa_circ_0000615 levels (Figure 2).\nCorrelations between hsa_circ_0000615 levels and hepatocellular carcinoma patient clinicopathological features\nHsa_circ_0000615 levels are linked with hepatocellular carcinoma patient survival. Patients exhibiting higher hsa_circ_0000615 expression levels exhibited prolonged overall survival compared with patients with lower levels of this circRNA.\n[SUBTITLE] Hsa_circ_0000615 is associated with poor chemoresistant hepatocellular carcinoma patient prognosis [SUBSECTION] Through Kaplan–Meier analyses and log‐rank tests, we found chemoresistant HCC patients to exhibit significantly reduced OS and progression‐free survival (PFS) compared with chemosensitive patients (Figure 3). Through Cox proportional hazards regression analyses, we determined that clinical stage, chemoresistance, TNM stage, lymph node metastasis, and hsa_circ_0000615 levels were associated with patient PFS (Table 2) and OS (Table 3), highlighting hsa_circ_0000615 as a promising independent predictor of chemoresistant HCC patient survival.\nHsa_circ_0000615 levels were significantly linked to poor outcomes in chemoresistant hepatocellular carcinoma (HCC) patients. Chemoresistant HCC patients exhibited significantly decreased progression‐free survival (A) and overall survival (B) relative to chemosensitive patients.\nUnivariate and multivariate analyses of hepatocellular carcinoma patient progression‐free survival\nUnivariate and multivariate analyses of hepatocellular carcinoma patient overall survival\nThrough Kaplan–Meier analyses and log‐rank tests, we found chemoresistant HCC patients to exhibit significantly reduced OS and progression‐free survival (PFS) compared with chemosensitive patients (Figure 3). Through Cox proportional hazards regression analyses, we determined that clinical stage, chemoresistance, TNM stage, lymph node metastasis, and hsa_circ_0000615 levels were associated with patient PFS (Table 2) and OS (Table 3), highlighting hsa_circ_0000615 as a promising independent predictor of chemoresistant HCC patient survival.\nHsa_circ_0000615 levels were significantly linked to poor outcomes in chemoresistant hepatocellular carcinoma (HCC) patients. Chemoresistant HCC patients exhibited significantly decreased progression‐free survival (A) and overall survival (B) relative to chemosensitive patients.\nUnivariate and multivariate analyses of hepatocellular carcinoma patient progression‐free survival\nUnivariate and multivariate analyses of hepatocellular carcinoma patient overall survival\n[SUBTITLE] Serum hsa_circ_0000615 levels offer diagnostic utility for the detection of hepatocellular carcinoma chemoresistance [SUBSECTION] Previous studies have shown that circRNAs show excellent potential diagnostic utility in various cancers, such as, breast cancer,\n20\n gastric cancer,\n21\n and HCC.\n22\n To assess the potential diagnostic utility of serum hsa_circ_0000615 in patients with HCC, the area under the receiver operating characteristic (ROC) curve (AUC) was determined and found to be 0.9238 (95% CI, 0.8915–0.956, Figure 4, p < .0001), consistent with the value of serum hsa_circ_0000615 as a biomarker capable of differentiating between HCC patients and healthy controls.\nSerum hsa_circ_0000615 levels offer diagnostic value as predictors of hepatocellular carcinoma (HCC) patient chemoresistance. Receiver‐operating characteristic curves were used to differentiate between chemoresistant HCC patients before and after therapy.\nPrevious studies have shown that circRNAs show excellent potential diagnostic utility in various cancers, such as, breast cancer,\n20\n gastric cancer,\n21\n and HCC.\n22\n To assess the potential diagnostic utility of serum hsa_circ_0000615 in patients with HCC, the area under the receiver operating characteristic (ROC) curve (AUC) was determined and found to be 0.9238 (95% CI, 0.8915–0.956, Figure 4, p < .0001), consistent with the value of serum hsa_circ_0000615 as a biomarker capable of differentiating between HCC patients and healthy controls.\nSerum hsa_circ_0000615 levels offer diagnostic value as predictors of hepatocellular carcinoma (HCC) patient chemoresistance. Receiver‐operating characteristic curves were used to differentiate between chemoresistant HCC patients before and after therapy.", "We began by assessing the levels of hsa_circ_0000615 in control and sorafenib‐resistant HCC cells. The sorafenib‐resistant Hep G2/sorafenib and Huh 7/sorafenib cell lines exhibited marked upregulation of this circRNA relative to corresponding parental cell lines (Figure 1A). To explore the potential utility of hsa_circ_0000615 as a biomarker of chemoresistance, we then assessed the levels of this circRNA in serum samples from 202 HCC patients and 202 healthy controls. HCC patients exhibited significantly elevated serum hsa_circ_0000615 levels compared with healthy controls (Figure 1B). Moreover, hsa_circ_0000615 expression levels were higher in sorafenib‐resistant patients (n = 122) relative to those in sorafenib‐sensitive individuals (n = 80) (Figure 1C). As such, hsa_circ_0000615 offers potential value as an HCC chemotherapy biomarker.\nHepatocellular carcinoma (HCC) patient serum samples exhibit hsa_circ_0000615 upregulation. (A). Hsa_circ_0000615 levels were significantly increased in sorafenib‐resistant HCC cells. (B). Serum hsa_circ_0000615 levels were higher in HCC patients. (C). sorafenib‐resistant patients (n = 122) exhibited higher levels of hsa_circ_0000615 expression compared with sorafenib‐sensitive patients in the before treatment and after treatment (n = 80). *p < .05.", "Next, we stratified HCC patients into those with high and low levels of serum hsa_circ_0000615 based on the mean expression of this circRNA in this cohort and compared clinical features between these two patient groups. Chi‐squared analyses revealed that hsa_circ_0000615 expression was associated with clinical stage, TNM stage, and lymph node metastasis (Table 1), but was unrelated to age, sex, or histological grade. Kaplan–Meier analyses indicated that patients exhibiting higher levels of hsa_circ_0000615 expression presented with shorter OS relative to patients expressing low hsa_circ_0000615 levels (Figure 2).\nCorrelations between hsa_circ_0000615 levels and hepatocellular carcinoma patient clinicopathological features\nHsa_circ_0000615 levels are linked with hepatocellular carcinoma patient survival. Patients exhibiting higher hsa_circ_0000615 expression levels exhibited prolonged overall survival compared with patients with lower levels of this circRNA.", "Through Kaplan–Meier analyses and log‐rank tests, we found chemoresistant HCC patients to exhibit significantly reduced OS and progression‐free survival (PFS) compared with chemosensitive patients (Figure 3). Through Cox proportional hazards regression analyses, we determined that clinical stage, chemoresistance, TNM stage, lymph node metastasis, and hsa_circ_0000615 levels were associated with patient PFS (Table 2) and OS (Table 3), highlighting hsa_circ_0000615 as a promising independent predictor of chemoresistant HCC patient survival.\nHsa_circ_0000615 levels were significantly linked to poor outcomes in chemoresistant hepatocellular carcinoma (HCC) patients. Chemoresistant HCC patients exhibited significantly decreased progression‐free survival (A) and overall survival (B) relative to chemosensitive patients.\nUnivariate and multivariate analyses of hepatocellular carcinoma patient progression‐free survival\nUnivariate and multivariate analyses of hepatocellular carcinoma patient overall survival", "Previous studies have shown that circRNAs show excellent potential diagnostic utility in various cancers, such as, breast cancer,\n20\n gastric cancer,\n21\n and HCC.\n22\n To assess the potential diagnostic utility of serum hsa_circ_0000615 in patients with HCC, the area under the receiver operating characteristic (ROC) curve (AUC) was determined and found to be 0.9238 (95% CI, 0.8915–0.956, Figure 4, p < .0001), consistent with the value of serum hsa_circ_0000615 as a biomarker capable of differentiating between HCC patients and healthy controls.\nSerum hsa_circ_0000615 levels offer diagnostic value as predictors of hepatocellular carcinoma (HCC) patient chemoresistance. Receiver‐operating characteristic curves were used to differentiate between chemoresistant HCC patients before and after therapy.", "Herein, we found that serum samples from HCC patients exhibited significant increases in hsa_circ_0000615 levels compared with those from control individuals. Moreover, the upregulation of this circRNA in samples from sorafenib‐resistant HCC patients relative to those from chemosensitive patients suggested that it may offer value as an independent predictor of patient outcomes.\nA growing body of evidence suggests that circRNAs are functionally important in cancer and may offer value as predictive biomarkers or therapeutic targets. In colorectal cancer, for example, circRNA_0000392 can promote tumor progression via the miR‐193a‐5p/PIK3R3/AKT axis.\n23\n Moreover, in non‐small cell lung cancer, circNDUFB2 can destabilize IGF2BPs and activate anti‐tumor immune responses to suppress tumor progression.\n24\n In HCC, circRNA‐SORE can stabilize YBX1 to drive sorafenib resistance.\n25\n As such, circRNAs function in a tumor‐specific manner.\nNeoadjuvant chemotherapy is a mainstay of treatment for many cancers and has been used with increasing frequency over the past decade, with sorafenib‐based neoadjuvant chemotherapy being a standard of care for HCC patients. Those HCC patients that undergo sorafenib‐based chemotherapy prior to radical cystectomy exhibit better OS outcomes, but a subset of patients fail to attain any benefit from such treatment, with pathological responses to neoadjuvant chemotherapy being predictive of disease‐specific survival outcomes. Identifying reliable biomarkers capable of guiding clinicians to the selection of patients most likely to benefit from chemotherapeutic intervention is thus a critical clinical task.\nIn HCC, circRNAs can function as central regulators of sorafenib‐resistance,\n26\n, \n27\n, \n28\n with hsa_circ_0000615 having previously been shown to drive HCC tumor growth and metastatic progression.\n18\n, \n19\n In this study, we further found hsa_circ_0000615 to be expressed at significantly higher levels in Hep G2/sorafenib and Huh 7/sorafenib cells relative to corresponding parental cell lines. The expression of this circRNA was similarly elevated in sorafenib‐resistant HCC patients compared with their chemosensitive counterparts, suggesting that hsa_circ_0000615 may offer value as a predictor of chemotherapeutic responses. Levels of hsa_circ_0000615 were also related to clinical stage, lymph node metastasis, and T stage in HCC patients, although they were unrelated to tumor histological stage, N stage, M stage, or patient age and gender. Kaplan–Meier analyses indicated that higher levels of hsa_circ_0000615 expression were associated with shorter patient OS compared with low levels of this circRNA. Moreover, chemoresistant HCC patients exhibited shorter OS and PFS compared with chemosensitive patients. Univariate and multivariate analyses further revealed clinical stage, T stage, lymph node metastasis, and chemoresistance to be correlated with OS and PFS outcomes, suggesting hsa_circ_0000615 to be a valuable independent predictor of HCC patient outcomes. In addition, the AUC value for this circRNA in HCC patients was 0.9238, indicating that serum levels of hsa_circ_0000615 can be used to reliably differentiate between HCC patients and healthy individuals.", "In summary, we herein found hsa_circ_0000615 upregulation to be prominent within serum samples from HCC patients, with such upregulation being significantly more pronounced in samples from chemosensitive patients relative to chemoresistant patients. As such, hsa_circ_0000615 is a promising target that warrants further study in an effort to understand the mechanistic basis for HCC patient chemoresistance.", "The authors of this work declare that they have no conflict of interest." ]
[ null, "materials-and-methods", null, null, null, "results", null, null, null, null, "discussion", "conclusions", "COI-statement" ]
[ "hepatocellular carcinoma", "hsa_circ_0000615", "sorafenib resistance" ]
Low plasma growth/differentiation factor 1 levels are associated with liver fibrosis in patients with stable angina.
36268984
Growth differentiation factor 1 (GDF1) is a member of the transforming growth factor-β (TGF-β) superfamily and a protective mediator against the development of post-infarction cardiac remodeling by negatively regulating MEK-ERK1/2 and Smad signaling pathways in the heart. The TGF-β/SMAD pathway has been shown to play a key role in the development of hepatic fibrosis. In addition, fatty liver disease has been associated with reduced MEK/ERK1/2 signaling. However, no previous study has investigated the association between GDF1 and liver fibrosis. Therefore, the aim of this study was to investigate the association between plasma GDF1 and liver fibrosis in patients with stable angina.
BACKGROUND
We included 327 consecutive patients with stable angina. ELISA was used to measure circulating levels of GDF1, and the fibrosis-4 index was used to assess liver fibrosis.
METHODS
The advanced liver fibrosis group had lower median plasma GDF1 levels than those with minimal liver fibrosis. There was a significant negative association between GDF1 plasma level and fibrosis-4 index (r = -0.135, p = 0.019). A lower concentration of GDF1 was significantly and independently associated with an increased risk of liver fibrosis when concentration was analyzed as a continuous variable and by tertile. In addition, fibrosis-4 index, aspartate aminotransferase (AST)-to-platelet ratio index, and AST/alanine aminotransferase ratio were significantly associated with GDF1 concentration.
RESULTS
Our results indicated an association between low plasma GDF1 and liver fibrosis in the enrolled patients. Further investigations into the role of plasma GDF1 in the pathogenesis of liver fibrosis are warranted.
CONCLUSIONS
[ "Humans", "Angina, Stable", "Growth Differentiation Factor 1", "Liver", "Liver Cirrhosis", "Mitogen-Activated Protein Kinase Kinases", "Smad Proteins", "Transforming Growth Factor beta" ]
9701856
null
null
null
null
RESULTS
[SUBTITLE] Characteristics of the patients by liver fibrosis category [SUBSECTION] Table 1 shows the biochemical and clinical characteristics of the 327 enrolled patients according to liver fibrosis category. The prevalence rates of minimal liver fibrosis (FIB‐4 < 1.45), moderate liver fibrosis (FIB‐4 1.45–3.25), and advanced liver fibrosis (FIB‐4 > 3.25) were 24.5%, 43.1%, and 32.4%, respectively. The advanced liver fibrosis group were older, had higher levels of AST, blood urea nitrogen, and creatinine, and lower BMI, eGFR, albumin, platelets, hematocrit, and hemoglobin than those with minimal and moderate liver fibrosis. Furthermore, the advanced liver fibrosis group had higher ALT, white blood cell count, and hs‐CRP, and a lower rate of drinking alcohol, waist circumference, and GDF1 than the minimal liver fibrosis group. Moreover, the advanced liver fibrosis group had a lower triglyceride level than the moderate liver fibrosis group. No significant differences were found in hypertension, male sex, hyperlipidemia, current smoking, diabetes mellitus, DBP, SBP, fasting glucose, HbA1c, total cholesterol, LDL‐C, HDL‐C, Gensini score or number of diseased coronary arteries among the three groups. Baseline characteristics of the study population stratified by severity of liver fibrosis. Note: Data are mean ± SD or frequency (percentage). Abbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; BP, blood pressure; BUN, blood urea nitrogen; eGFR, estimated glomerular filtration rate; FIB, fibrosis; GDF1, growth/differentiation factor 1; HDL, high‐density lipoprotein; Hs‐CRP, high‐sensitivity C‐reactive protein; LDL, low‐density lipoprotein; WBC, white blood cell; WHR, waist‐to‐hip ratio. Significant difference was tested using log‐transformed data. Table 1 shows the biochemical and clinical characteristics of the 327 enrolled patients according to liver fibrosis category. The prevalence rates of minimal liver fibrosis (FIB‐4 < 1.45), moderate liver fibrosis (FIB‐4 1.45–3.25), and advanced liver fibrosis (FIB‐4 > 3.25) were 24.5%, 43.1%, and 32.4%, respectively. The advanced liver fibrosis group were older, had higher levels of AST, blood urea nitrogen, and creatinine, and lower BMI, eGFR, albumin, platelets, hematocrit, and hemoglobin than those with minimal and moderate liver fibrosis. Furthermore, the advanced liver fibrosis group had higher ALT, white blood cell count, and hs‐CRP, and a lower rate of drinking alcohol, waist circumference, and GDF1 than the minimal liver fibrosis group. Moreover, the advanced liver fibrosis group had a lower triglyceride level than the moderate liver fibrosis group. No significant differences were found in hypertension, male sex, hyperlipidemia, current smoking, diabetes mellitus, DBP, SBP, fasting glucose, HbA1c, total cholesterol, LDL‐C, HDL‐C, Gensini score or number of diseased coronary arteries among the three groups. Baseline characteristics of the study population stratified by severity of liver fibrosis. Note: Data are mean ± SD or frequency (percentage). Abbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; BP, blood pressure; BUN, blood urea nitrogen; eGFR, estimated glomerular filtration rate; FIB, fibrosis; GDF1, growth/differentiation factor 1; HDL, high‐density lipoprotein; Hs‐CRP, high‐sensitivity C‐reactive protein; LDL, low‐density lipoprotein; WBC, white blood cell; WHR, waist‐to‐hip ratio. Significant difference was tested using log‐transformed data. [SUBTITLE] Association between plasma GDF1 and liver fibrosis [SUBSECTION] We found a significant association between a lower plasma GDF1 concentration and liver fibrosis, even after controlling for anthropometric factors, ALT, hemoglobin, albumin, creatinine, and hs‐CRP (Table 2). Furthermore, a lower concentration of GDF1 showed a significant linear trend and was independently associated with liver fibrosis, especially when analyzing the concentration both as a continuous variable and by tertile (Tables 2 and 3). In the multiple logistic regression analysis, fully adjusted ORs for liver fibrosis in the second and third tertiles were 2.02 (95% CI, 0.70–6.30) and 5.04 (95% CI, 1.35–25.60), respectively. Association of plasma GDF1 with liver fibrosis in fully adjusted models Note: Results of multivariate logistic regression analysis are presented as the odds ratio (OR) of having a liver fibrosis status and decreased plasma GDF1 level. Abbreviations: ALT, alanine aminotransferase; BMI, body mass index; GDF1, growth/differentiation factor 1; Hs‐CRP, high‐sensitivity C‐reactive protein. Univariate and multivariate analyses of the impact of plasma GDF1 level on liver fibrosis Note: Values shown are cut‐off values of plasma GDF1 levels of all subjects, and odds ratios (ORs) with 95% confidence intervals (CIs). Abbreviation: GDF1, growth/differentiation factor 1. Adjusted for age, sex, body mass index, alanine aminotransferase, hemoglobin, albumin, creatinine, and high‐sensitivity C‐reactive protein. We found a significant association between a lower plasma GDF1 concentration and liver fibrosis, even after controlling for anthropometric factors, ALT, hemoglobin, albumin, creatinine, and hs‐CRP (Table 2). Furthermore, a lower concentration of GDF1 showed a significant linear trend and was independently associated with liver fibrosis, especially when analyzing the concentration both as a continuous variable and by tertile (Tables 2 and 3). In the multiple logistic regression analysis, fully adjusted ORs for liver fibrosis in the second and third tertiles were 2.02 (95% CI, 0.70–6.30) and 5.04 (95% CI, 1.35–25.60), respectively. Association of plasma GDF1 with liver fibrosis in fully adjusted models Note: Results of multivariate logistic regression analysis are presented as the odds ratio (OR) of having a liver fibrosis status and decreased plasma GDF1 level. Abbreviations: ALT, alanine aminotransferase; BMI, body mass index; GDF1, growth/differentiation factor 1; Hs‐CRP, high‐sensitivity C‐reactive protein. Univariate and multivariate analyses of the impact of plasma GDF1 level on liver fibrosis Note: Values shown are cut‐off values of plasma GDF1 levels of all subjects, and odds ratios (ORs) with 95% confidence intervals (CIs). Abbreviation: GDF1, growth/differentiation factor 1. Adjusted for age, sex, body mass index, alanine aminotransferase, hemoglobin, albumin, creatinine, and high‐sensitivity C‐reactive protein. [SUBTITLE] Associations between plasma GDF1 level and clinical laboratory data [SUBSECTION] Simple linear regression analysis showed that plasma GDF1 was negatively associated with the FIB‐4 index, blood urea nitrogen, and creatinine, and positively associated with platelet count and eGFR. In multiple linear regression analysis, plasma GDF1 level was positively associated with platelet count, and negatively associated with the FIB‐4 index, blood urea nitrogen, and creatinine (Table 4). In addition, in analysis of the subjects by tertile of GDF1 concentration, the FIB‐4 index, APRI, and AST/ALT ratio were significantly associated with GDF1 concentration (p for trend < 0.05, Figure 1). Linear regression analysis of variables associated with plasma log‐GDF1 levels in the study subjects. Abbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; BP, blood.pressure; GDF1, growth/differentiation factor 1; GFR, glomerular filtration rate; HDL, high‐density lipoprotein; Hs‐CRP, high‐sensitivity C‐reactive protein; LDL, low‐density lipoprotein. Regression coefficient adjusted for age and sex. Classification of subjects into tertiles according to growth/differentiation factor 1 (GDF1) concentration revealed that the fibrosis‐4 index (A), APRI (B), and AST/ALT ratio (C) were significantly associated with GDF1 concentration (p for trend < 0.05). Bars represent the mean ± SD. ALT, alanine aminotransferase; APRI, aspartate aminotransferase to platelet ratio index; AST, aspartate aminotransferase. Simple linear regression analysis showed that plasma GDF1 was negatively associated with the FIB‐4 index, blood urea nitrogen, and creatinine, and positively associated with platelet count and eGFR. In multiple linear regression analysis, plasma GDF1 level was positively associated with platelet count, and negatively associated with the FIB‐4 index, blood urea nitrogen, and creatinine (Table 4). In addition, in analysis of the subjects by tertile of GDF1 concentration, the FIB‐4 index, APRI, and AST/ALT ratio were significantly associated with GDF1 concentration (p for trend < 0.05, Figure 1). Linear regression analysis of variables associated with plasma log‐GDF1 levels in the study subjects. Abbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; BP, blood.pressure; GDF1, growth/differentiation factor 1; GFR, glomerular filtration rate; HDL, high‐density lipoprotein; Hs‐CRP, high‐sensitivity C‐reactive protein; LDL, low‐density lipoprotein. Regression coefficient adjusted for age and sex. Classification of subjects into tertiles according to growth/differentiation factor 1 (GDF1) concentration revealed that the fibrosis‐4 index (A), APRI (B), and AST/ALT ratio (C) were significantly associated with GDF1 concentration (p for trend < 0.05). Bars represent the mean ± SD. ALT, alanine aminotransferase; APRI, aspartate aminotransferase to platelet ratio index; AST, aspartate aminotransferase.
CONCLUSIONS
Our results indicated that in patients with stable angina, low plasma GDF1 was associated with liver fibrosis. Further studies are warranted to investigate the association between plasma GDF1 and the pathogenesis of liver fibrosis.
[ "INTRODUCTION", "Patient population", "Baseline data collection", "Laboratory measurements", "Measurements of plasma GDF1 and high‐sensitive C‐reactive protein (hs‐CRP)", "Definitions", "Statistical analysis", "Characteristics of the patients by liver fibrosis category", "Association between plasma GDF1 and liver fibrosis", "Associations between plasma GDF1 level and clinical laboratory data", "AUTHOR CONTRIBUTIONS", "FUNDING INFORMATION", "INFORMED CONSENT" ]
[ "Cardiovascular disease (CVD) is the leading cause of death worldwide,\n1\n and is precipitated by cardiometabolic risk factors including chronic liver disease, diabetes, metabolic syndrome, and chronic renal disease.\n1\n, \n2\n, \n3\n Chronic liver disease is a progressive deterioration of liver function, and the most common causes are hepatitis, alcohol abuse, and nonalcoholic fatty liver disease (NAFLD).\n4\n NAFLD is common in patients with CVD, and both diseases share common risks factor such as high cholesterol intake, alcohol consumption, diabetes and other metabolic abnormalities.\n5\n In addition, recent studies have reported associations between liver fibrosis, which is the irreversible progression of NAFLD, and coronary artery disease (CAD), cardiac arrhythmias, and impaired heart function.\n3\n, \n6\n, \n7\n Furthermore, previous study also revealed that higher liver fibrosis scores are associated with increased risks of cardiovascular and all‐cause mortality among patients with CAD. Liver fibrosis scores might play a potential role in CAD prognosis prediction.\n3\n However, the underlying mechanisms and interactions remain unknown.\nGrowth differentiation factors (GDFs) are proteins which belong to the transforming growth factor‐β (TGF‐β) superfamily, and 15 GDFs have been identified to date (GDF1 to GDF15).\n8\n The TGF‐β superfamily comprised structurally related polypeptides which have been shown to regulate cell differentiation and growth in various adult and embryonic tissues. GDF1 has been shown to be specifically expressed in the nervous systems of adult mice and late‐stage embryos.\n9\n, \n10\n A previous study showed that GDF1 had favorable effects on vascular endothelial function, sodium excretion, and post‐infarction cardiac remodeling in an animal study and also in patients with type 2 diabetes and normal individuals.\n11\n The underlying mechanism is believed to be through the negative regulation of MEK‐ERK1/2 and Smad signaling pathways.\n11\n Interestingly, the TGF‐β/SMAD pathway has also been found to play a key role in the development of hepatic fibrosis in subjects with fatty liver disease via suppression of the MEK/ERK1/2 signaling pathway.\n12\n, \n13\n However, few studies have investigated the relationship between plasma GDF1 level and liver fibrosis. Therefore, the aim of this study was to evaluate the relationship between plasma GDF1 level and liver fibrosis in a cohort of Taiwanese patients with stable angina.", "We enrolled a total of 327 patients with stable angina from July 2012 to December 2021 who visited the cardiovascular clinic at E‐Da Hospital. The inclusion criteria were patients: (1) with stable angina pectoris, defined as effort‐related chest pain without evidence of recent deterioration or rest pain in the previous 6 months, and diagnosed by the cardiologist in charge as previously described;\n14\n, \n15\n and (2) who underwent a successful percutaneous coronary intervention, defined as <30% residual stenosis in a final angiogram as assessed using quantitative coronary angiography with no occlusion of the large branch (>1 mm) or dissection limiting flow, and Thrombolysis in Myocardial Infarction grade 3.\n16\n The exclusion criteria were patients: (1) who used steroids; (2) had inflammatory diseases (such as infection/sepsis), malignancy, liver diseases, and collagen diseases; (3) with a history of psychosis; and (4) with heart valve disease, myocardial infarction, or had undergone heart surgery. In addition, we also excluded patients who were unable or unwilling to provide informed consent. All study patients lived in the same area during the study period, and they were all of Han Chinese ethnicity. Each patient provided written informed consent before being enrolled into the study. This cross‐sectional study was approved by the Human Research Ethics Committee of E‐Da Hospital.", "A detailed interview was conducted with each patient about their medical and personal history, as well as their demographic characteristics before the coronary angiography examination. We classified smoking status as follows: never smokers, former smokers (those who had stopped smoking for ≥1 year), or current smokers. We grouped the current and former smokers in the analysis and compared them with the never smokers. A trained nurse performed all blood pressure readings with the patients seated using an automated blood pressure monitor (HEM‐907; Omron) after 5 min of rest. Anthropometric parameters including body mass index (BMI) and waist circumference were also recorded.", "Fasting (8 h) peripheral blood samples were obtained from the antecubital vein before the patients underwent coronary angiography. Levels of high‐density lipoprotein cholesterol (HDL‐C), low‐density lipoprotein cholesterol (LDL‐C), total cholesterol, plasma triglycerides, serum glucose, serum albumin, serum creatinine, and complete blood cell count were measured in all patients using a parallel, multi‐channel analyzer (Hitachi 7170A) as described previously.\n17\n, \n18\n We used high‐performance liquid chromatography (Tosoh Automated Glycohemoglobin Analyzer, HLC‐723G8) to measure hemoglobin A1c (HbA1c). In addition, we measured levels of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) following the methods of the Japan Society of Clinical Chemistry (“Hitachi” Discrete photometric chemistry analyzer for clinical use, LAbOSPECT 008AS), which is compatible to the methods by the International Federation of Clinical Chemistry. Serum creatinine level was measured using the Jaffe method, and estimated glomerular filtration rate (eGFR) was calculated using the Chronic Kidney Disease Epidemiology Collaboration (CKD‐EPI) equation within 3–6 months of admission.\n19\n The fibrosis‐4 (FIB‐4) index was calculated as: FIB‐4 = age (years) × AST (IU/L)/platelet count (109/L) × √ALT (IU/L).\n20\n The aspartate transaminase/platelet ratio index (APRI) was calculated as: APRI = AST/upper limit of normal of AST/platelet count × 100 (where the upper limit of normal of AST = 40 IU/L).", "After obtaining overnight fasting blood samples, the plasma was kept at −80°C until assay. We used a commercial ELISA kit to measure concentrations of plasma GDF1 (Cloud‐Clone Corp.). The standard and dilution curves were parallel, and the inter‐assay and intra‐assay coefficients of variation for the assay were <12% and <10%, respectively (both n = 3). Plasma levels of hs‐CRP were measured using an immunochemistry system (Beckman Coulter IMMAGE), which had a detection of 0.2 mg/L. All measurements were made in duplicate during a single experiment.", "We used the World Health Organization criteria to define type 2 diabetes, as receiving medical therapy for diabetes and/or a history of type 2 diabetes.\n21\n Patients with a systolic/diastolic blood pressure (SBP/DBP) of ≥140/90 mmHg, or a prescription for antihypertensive drugs were defined as having hypertension. We used the Adult Treatment Panel III criteria\n22\n to define hyperlipidemia as: elevated total cholesterol (≥200 mg/dl), and/or elevated LDL‐C (≥130 mg/dl), and/or low HDL‐C (<35 mg/dl in men or < 39 mg/dl in women), and/or elevated triglycerides (≥150 mg/dl), or being treated for a lipid disorder. Liver fibrosis was defined according to FIB‐4 index categories as: minimal fibrosis (FIB‐4 < 1.45), moderate fibrosis (FIB‐4 1.45–3.25), and advanced fibrosis (FIB‐4 > 3.25).\n20\n, \n23\n, \n24\n\n", "Continuous data are presented as mean ± SD or median (interquartile range) as appropriate. Between‐group differences were analyzed with one‐way ANOVA for normally distributed variables followed by Tukey's pairwise test. Categorical data are presented as frequency and percentage, and between‐group comparisons were analyzed using the chi‐square test. Because the distributions of serum AST, ALT, triglycerides, creatinine, plasma hs‐CRP, and GDF1 values were skewed, we used logarithmically transformed values in the analysis. The association between GDF1 and liver fibrosis was evaluated using multivariate logistic regression models: (1) GDF1 and age; (2) GDF1, age, and sex; (3) GDF1, age, sex, and BMI; (4) GDF1, age, sex, BMI, and ALT; (5) GDF1, age, sex, BMI, ALT, hemoglobin, and albumin; and (6) GDF1, age, sex, BMI, ALT, hemoglobin, albumin, creatinine, and hs‐CRP. We then classified the concentration of GDF1 into tertiles, and analyzed trends among the tertiles using general linear and logistic regression analyses. Using the highest tertile as the reference, we also estimated the odds ratio (OR) with 95% confidence interval (CI) of liver fibrosis in each tertile.\nAssociations between plasma GDF1 concentration and other variables were analyzed using simple and multiple linear regression analyses. In addition, the values of FIB‐4 index, AST/ALT ratio and APRI in each GDF1 concentration tertile were analyzed for trends. All tests were 2‐sided, and a p value < 0.05 was considered to be statistically significant. All statistical analyses were performed using JMP version 7.0 for Windows (SAS Institute).", "Table 1 shows the biochemical and clinical characteristics of the 327 enrolled patients according to liver fibrosis category. The prevalence rates of minimal liver fibrosis (FIB‐4 < 1.45), moderate liver fibrosis (FIB‐4 1.45–3.25), and advanced liver fibrosis (FIB‐4 > 3.25) were 24.5%, 43.1%, and 32.4%, respectively. The advanced liver fibrosis group were older, had higher levels of AST, blood urea nitrogen, and creatinine, and lower BMI, eGFR, albumin, platelets, hematocrit, and hemoglobin than those with minimal and moderate liver fibrosis. Furthermore, the advanced liver fibrosis group had higher ALT, white blood cell count, and hs‐CRP, and a lower rate of drinking alcohol, waist circumference, and GDF1 than the minimal liver fibrosis group. Moreover, the advanced liver fibrosis group had a lower triglyceride level than the moderate liver fibrosis group. No significant differences were found in hypertension, male sex, hyperlipidemia, current smoking, diabetes mellitus, DBP, SBP, fasting glucose, HbA1c, total cholesterol, LDL‐C, HDL‐C, Gensini score or number of diseased coronary arteries among the three groups.\nBaseline characteristics of the study population stratified by severity of liver fibrosis.\n\nNote: Data are mean ± SD or frequency (percentage).\nAbbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; BP, blood pressure; BUN, blood urea nitrogen; eGFR, estimated glomerular filtration rate; FIB, fibrosis; GDF1, growth/differentiation factor 1; HDL, high‐density lipoprotein; Hs‐CRP, high‐sensitivity C‐reactive protein; LDL, low‐density lipoprotein; WBC, white blood cell; WHR, waist‐to‐hip ratio.\nSignificant difference was tested using log‐transformed data.", "We found a significant association between a lower plasma GDF1 concentration and liver fibrosis, even after controlling for anthropometric factors, ALT, hemoglobin, albumin, creatinine, and hs‐CRP (Table 2). Furthermore, a lower concentration of GDF1 showed a significant linear trend and was independently associated with liver fibrosis, especially when analyzing the concentration both as a continuous variable and by tertile (Tables 2 and 3). In the multiple logistic regression analysis, fully adjusted ORs for liver fibrosis in the second and third tertiles were 2.02 (95% CI, 0.70–6.30) and 5.04 (95% CI, 1.35–25.60), respectively.\nAssociation of plasma GDF1 with liver fibrosis in fully adjusted models\n\nNote: Results of multivariate logistic regression analysis are presented as the odds ratio (OR) of having a liver fibrosis status and decreased plasma GDF1 level.\nAbbreviations: ALT, alanine aminotransferase; BMI, body mass index; GDF1, growth/differentiation factor 1; Hs‐CRP, high‐sensitivity C‐reactive protein.\nUnivariate and multivariate analyses of the impact of plasma GDF1 level on liver fibrosis\n\nNote: Values shown are cut‐off values of plasma GDF1 levels of all subjects, and odds ratios (ORs) with 95% confidence intervals (CIs).\nAbbreviation: GDF1, growth/differentiation factor 1.\nAdjusted for age, sex, body mass index, alanine aminotransferase, hemoglobin, albumin, creatinine, and high‐sensitivity C‐reactive protein.", "Simple linear regression analysis showed that plasma GDF1 was negatively associated with the FIB‐4 index, blood urea nitrogen, and creatinine, and positively associated with platelet count and eGFR. In multiple linear regression analysis, plasma GDF1 level was positively associated with platelet count, and negatively associated with the FIB‐4 index, blood urea nitrogen, and creatinine (Table 4). In addition, in analysis of the subjects by tertile of GDF1 concentration, the FIB‐4 index, APRI, and AST/ALT ratio were significantly associated with GDF1 concentration (p for trend < 0.05, Figure 1).\nLinear regression analysis of variables associated with plasma log‐GDF1 levels in the study subjects.\nAbbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; BP, blood.pressure; GDF1, growth/differentiation factor 1; GFR, glomerular filtration rate; HDL, high‐density lipoprotein; Hs‐CRP, high‐sensitivity C‐reactive protein; LDL, low‐density lipoprotein.\nRegression coefficient adjusted for age and sex.\nClassification of subjects into tertiles according to growth/differentiation factor 1 (GDF1) concentration revealed that the fibrosis‐4 index (A), APRI (B), and AST/ALT ratio (C) were significantly associated with GDF1 concentration (p for trend < 0.05). Bars represent the mean ± SD. ALT, alanine aminotransferase; APRI, aspartate aminotransferase to platelet ratio index; AST, aspartate aminotransferase.", "All authors contributed to this study. W.‐C.H., W.‐H.T., and C.‐C.H. conceived and designed the study. W.‐C.H., W.‐H.T., Y.‐J.L., and C.‐C.H. provided the methodology. F.‐M.C. performed the formal analysis, and project administration. T.‐H.Y., C.‐C.W., and C.‐C.H. validated the data. T.‐H.Y., C.‐C.W., W.‐C.H., and C.‐P.W. performed the investigation, resources, and data curation. T.‐H.Y., W.‐C.H., C.‐C.W., C.‐P.W., Y.‐C.L., and C.‐T.W. prepared the manuscript. W.‐H.T., T.‐H.Y., W.‐C.H., C.‐C.W., C.‐P.W., Y.‐C.L., and C.‐T.W. reviewed and edited the manuscript. W.‐C.H., W.‐H.T., Y.‐J.L., and C.‐C.H. performed the visualization. W.‐C.H. and C.‐C.H. performed the supervision and funding acquisition. All authors have read and agreed to the published version of the manuscript.", "E‐Da Hospital financially supported this research under Contracts EDAHP109002, EDAHI109002, and EDAHI110001.", "Each patient provided written informed consent before being enrolled into the study." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "MATERIALS AND METHODS", "Patient population", "Baseline data collection", "Laboratory measurements", "Measurements of plasma GDF1 and high‐sensitive C‐reactive protein (hs‐CRP)", "Definitions", "Statistical analysis", "RESULTS", "Characteristics of the patients by liver fibrosis category", "Association between plasma GDF1 and liver fibrosis", "Associations between plasma GDF1 level and clinical laboratory data", "DISCUSSION", "CONCLUSIONS", "AUTHOR CONTRIBUTIONS", "FUNDING INFORMATION", "CONFLICT OF INTEREST", "INFORMED CONSENT" ]
[ "Cardiovascular disease (CVD) is the leading cause of death worldwide,\n1\n and is precipitated by cardiometabolic risk factors including chronic liver disease, diabetes, metabolic syndrome, and chronic renal disease.\n1\n, \n2\n, \n3\n Chronic liver disease is a progressive deterioration of liver function, and the most common causes are hepatitis, alcohol abuse, and nonalcoholic fatty liver disease (NAFLD).\n4\n NAFLD is common in patients with CVD, and both diseases share common risks factor such as high cholesterol intake, alcohol consumption, diabetes and other metabolic abnormalities.\n5\n In addition, recent studies have reported associations between liver fibrosis, which is the irreversible progression of NAFLD, and coronary artery disease (CAD), cardiac arrhythmias, and impaired heart function.\n3\n, \n6\n, \n7\n Furthermore, previous study also revealed that higher liver fibrosis scores are associated with increased risks of cardiovascular and all‐cause mortality among patients with CAD. Liver fibrosis scores might play a potential role in CAD prognosis prediction.\n3\n However, the underlying mechanisms and interactions remain unknown.\nGrowth differentiation factors (GDFs) are proteins which belong to the transforming growth factor‐β (TGF‐β) superfamily, and 15 GDFs have been identified to date (GDF1 to GDF15).\n8\n The TGF‐β superfamily comprised structurally related polypeptides which have been shown to regulate cell differentiation and growth in various adult and embryonic tissues. GDF1 has been shown to be specifically expressed in the nervous systems of adult mice and late‐stage embryos.\n9\n, \n10\n A previous study showed that GDF1 had favorable effects on vascular endothelial function, sodium excretion, and post‐infarction cardiac remodeling in an animal study and also in patients with type 2 diabetes and normal individuals.\n11\n The underlying mechanism is believed to be through the negative regulation of MEK‐ERK1/2 and Smad signaling pathways.\n11\n Interestingly, the TGF‐β/SMAD pathway has also been found to play a key role in the development of hepatic fibrosis in subjects with fatty liver disease via suppression of the MEK/ERK1/2 signaling pathway.\n12\n, \n13\n However, few studies have investigated the relationship between plasma GDF1 level and liver fibrosis. Therefore, the aim of this study was to evaluate the relationship between plasma GDF1 level and liver fibrosis in a cohort of Taiwanese patients with stable angina.", "[SUBTITLE] Patient population [SUBSECTION] We enrolled a total of 327 patients with stable angina from July 2012 to December 2021 who visited the cardiovascular clinic at E‐Da Hospital. The inclusion criteria were patients: (1) with stable angina pectoris, defined as effort‐related chest pain without evidence of recent deterioration or rest pain in the previous 6 months, and diagnosed by the cardiologist in charge as previously described;\n14\n, \n15\n and (2) who underwent a successful percutaneous coronary intervention, defined as <30% residual stenosis in a final angiogram as assessed using quantitative coronary angiography with no occlusion of the large branch (>1 mm) or dissection limiting flow, and Thrombolysis in Myocardial Infarction grade 3.\n16\n The exclusion criteria were patients: (1) who used steroids; (2) had inflammatory diseases (such as infection/sepsis), malignancy, liver diseases, and collagen diseases; (3) with a history of psychosis; and (4) with heart valve disease, myocardial infarction, or had undergone heart surgery. In addition, we also excluded patients who were unable or unwilling to provide informed consent. All study patients lived in the same area during the study period, and they were all of Han Chinese ethnicity. Each patient provided written informed consent before being enrolled into the study. This cross‐sectional study was approved by the Human Research Ethics Committee of E‐Da Hospital.\nWe enrolled a total of 327 patients with stable angina from July 2012 to December 2021 who visited the cardiovascular clinic at E‐Da Hospital. The inclusion criteria were patients: (1) with stable angina pectoris, defined as effort‐related chest pain without evidence of recent deterioration or rest pain in the previous 6 months, and diagnosed by the cardiologist in charge as previously described;\n14\n, \n15\n and (2) who underwent a successful percutaneous coronary intervention, defined as <30% residual stenosis in a final angiogram as assessed using quantitative coronary angiography with no occlusion of the large branch (>1 mm) or dissection limiting flow, and Thrombolysis in Myocardial Infarction grade 3.\n16\n The exclusion criteria were patients: (1) who used steroids; (2) had inflammatory diseases (such as infection/sepsis), malignancy, liver diseases, and collagen diseases; (3) with a history of psychosis; and (4) with heart valve disease, myocardial infarction, or had undergone heart surgery. In addition, we also excluded patients who were unable or unwilling to provide informed consent. All study patients lived in the same area during the study period, and they were all of Han Chinese ethnicity. Each patient provided written informed consent before being enrolled into the study. This cross‐sectional study was approved by the Human Research Ethics Committee of E‐Da Hospital.\n[SUBTITLE] Baseline data collection [SUBSECTION] A detailed interview was conducted with each patient about their medical and personal history, as well as their demographic characteristics before the coronary angiography examination. We classified smoking status as follows: never smokers, former smokers (those who had stopped smoking for ≥1 year), or current smokers. We grouped the current and former smokers in the analysis and compared them with the never smokers. A trained nurse performed all blood pressure readings with the patients seated using an automated blood pressure monitor (HEM‐907; Omron) after 5 min of rest. Anthropometric parameters including body mass index (BMI) and waist circumference were also recorded.\nA detailed interview was conducted with each patient about their medical and personal history, as well as their demographic characteristics before the coronary angiography examination. We classified smoking status as follows: never smokers, former smokers (those who had stopped smoking for ≥1 year), or current smokers. We grouped the current and former smokers in the analysis and compared them with the never smokers. A trained nurse performed all blood pressure readings with the patients seated using an automated blood pressure monitor (HEM‐907; Omron) after 5 min of rest. Anthropometric parameters including body mass index (BMI) and waist circumference were also recorded.\n[SUBTITLE] Laboratory measurements [SUBSECTION] Fasting (8 h) peripheral blood samples were obtained from the antecubital vein before the patients underwent coronary angiography. Levels of high‐density lipoprotein cholesterol (HDL‐C), low‐density lipoprotein cholesterol (LDL‐C), total cholesterol, plasma triglycerides, serum glucose, serum albumin, serum creatinine, and complete blood cell count were measured in all patients using a parallel, multi‐channel analyzer (Hitachi 7170A) as described previously.\n17\n, \n18\n We used high‐performance liquid chromatography (Tosoh Automated Glycohemoglobin Analyzer, HLC‐723G8) to measure hemoglobin A1c (HbA1c). In addition, we measured levels of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) following the methods of the Japan Society of Clinical Chemistry (“Hitachi” Discrete photometric chemistry analyzer for clinical use, LAbOSPECT 008AS), which is compatible to the methods by the International Federation of Clinical Chemistry. Serum creatinine level was measured using the Jaffe method, and estimated glomerular filtration rate (eGFR) was calculated using the Chronic Kidney Disease Epidemiology Collaboration (CKD‐EPI) equation within 3–6 months of admission.\n19\n The fibrosis‐4 (FIB‐4) index was calculated as: FIB‐4 = age (years) × AST (IU/L)/platelet count (109/L) × √ALT (IU/L).\n20\n The aspartate transaminase/platelet ratio index (APRI) was calculated as: APRI = AST/upper limit of normal of AST/platelet count × 100 (where the upper limit of normal of AST = 40 IU/L).\nFasting (8 h) peripheral blood samples were obtained from the antecubital vein before the patients underwent coronary angiography. Levels of high‐density lipoprotein cholesterol (HDL‐C), low‐density lipoprotein cholesterol (LDL‐C), total cholesterol, plasma triglycerides, serum glucose, serum albumin, serum creatinine, and complete blood cell count were measured in all patients using a parallel, multi‐channel analyzer (Hitachi 7170A) as described previously.\n17\n, \n18\n We used high‐performance liquid chromatography (Tosoh Automated Glycohemoglobin Analyzer, HLC‐723G8) to measure hemoglobin A1c (HbA1c). In addition, we measured levels of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) following the methods of the Japan Society of Clinical Chemistry (“Hitachi” Discrete photometric chemistry analyzer for clinical use, LAbOSPECT 008AS), which is compatible to the methods by the International Federation of Clinical Chemistry. Serum creatinine level was measured using the Jaffe method, and estimated glomerular filtration rate (eGFR) was calculated using the Chronic Kidney Disease Epidemiology Collaboration (CKD‐EPI) equation within 3–6 months of admission.\n19\n The fibrosis‐4 (FIB‐4) index was calculated as: FIB‐4 = age (years) × AST (IU/L)/platelet count (109/L) × √ALT (IU/L).\n20\n The aspartate transaminase/platelet ratio index (APRI) was calculated as: APRI = AST/upper limit of normal of AST/platelet count × 100 (where the upper limit of normal of AST = 40 IU/L).\n[SUBTITLE] Measurements of plasma GDF1 and high‐sensitive C‐reactive protein (hs‐CRP) [SUBSECTION] After obtaining overnight fasting blood samples, the plasma was kept at −80°C until assay. We used a commercial ELISA kit to measure concentrations of plasma GDF1 (Cloud‐Clone Corp.). The standard and dilution curves were parallel, and the inter‐assay and intra‐assay coefficients of variation for the assay were <12% and <10%, respectively (both n = 3). Plasma levels of hs‐CRP were measured using an immunochemistry system (Beckman Coulter IMMAGE), which had a detection of 0.2 mg/L. All measurements were made in duplicate during a single experiment.\nAfter obtaining overnight fasting blood samples, the plasma was kept at −80°C until assay. We used a commercial ELISA kit to measure concentrations of plasma GDF1 (Cloud‐Clone Corp.). The standard and dilution curves were parallel, and the inter‐assay and intra‐assay coefficients of variation for the assay were <12% and <10%, respectively (both n = 3). Plasma levels of hs‐CRP were measured using an immunochemistry system (Beckman Coulter IMMAGE), which had a detection of 0.2 mg/L. All measurements were made in duplicate during a single experiment.\n[SUBTITLE] Definitions [SUBSECTION] We used the World Health Organization criteria to define type 2 diabetes, as receiving medical therapy for diabetes and/or a history of type 2 diabetes.\n21\n Patients with a systolic/diastolic blood pressure (SBP/DBP) of ≥140/90 mmHg, or a prescription for antihypertensive drugs were defined as having hypertension. We used the Adult Treatment Panel III criteria\n22\n to define hyperlipidemia as: elevated total cholesterol (≥200 mg/dl), and/or elevated LDL‐C (≥130 mg/dl), and/or low HDL‐C (<35 mg/dl in men or < 39 mg/dl in women), and/or elevated triglycerides (≥150 mg/dl), or being treated for a lipid disorder. Liver fibrosis was defined according to FIB‐4 index categories as: minimal fibrosis (FIB‐4 < 1.45), moderate fibrosis (FIB‐4 1.45–3.25), and advanced fibrosis (FIB‐4 > 3.25).\n20\n, \n23\n, \n24\n\n\nWe used the World Health Organization criteria to define type 2 diabetes, as receiving medical therapy for diabetes and/or a history of type 2 diabetes.\n21\n Patients with a systolic/diastolic blood pressure (SBP/DBP) of ≥140/90 mmHg, or a prescription for antihypertensive drugs were defined as having hypertension. We used the Adult Treatment Panel III criteria\n22\n to define hyperlipidemia as: elevated total cholesterol (≥200 mg/dl), and/or elevated LDL‐C (≥130 mg/dl), and/or low HDL‐C (<35 mg/dl in men or < 39 mg/dl in women), and/or elevated triglycerides (≥150 mg/dl), or being treated for a lipid disorder. Liver fibrosis was defined according to FIB‐4 index categories as: minimal fibrosis (FIB‐4 < 1.45), moderate fibrosis (FIB‐4 1.45–3.25), and advanced fibrosis (FIB‐4 > 3.25).\n20\n, \n23\n, \n24\n\n\n[SUBTITLE] Statistical analysis [SUBSECTION] Continuous data are presented as mean ± SD or median (interquartile range) as appropriate. Between‐group differences were analyzed with one‐way ANOVA for normally distributed variables followed by Tukey's pairwise test. Categorical data are presented as frequency and percentage, and between‐group comparisons were analyzed using the chi‐square test. Because the distributions of serum AST, ALT, triglycerides, creatinine, plasma hs‐CRP, and GDF1 values were skewed, we used logarithmically transformed values in the analysis. The association between GDF1 and liver fibrosis was evaluated using multivariate logistic regression models: (1) GDF1 and age; (2) GDF1, age, and sex; (3) GDF1, age, sex, and BMI; (4) GDF1, age, sex, BMI, and ALT; (5) GDF1, age, sex, BMI, ALT, hemoglobin, and albumin; and (6) GDF1, age, sex, BMI, ALT, hemoglobin, albumin, creatinine, and hs‐CRP. We then classified the concentration of GDF1 into tertiles, and analyzed trends among the tertiles using general linear and logistic regression analyses. Using the highest tertile as the reference, we also estimated the odds ratio (OR) with 95% confidence interval (CI) of liver fibrosis in each tertile.\nAssociations between plasma GDF1 concentration and other variables were analyzed using simple and multiple linear regression analyses. In addition, the values of FIB‐4 index, AST/ALT ratio and APRI in each GDF1 concentration tertile were analyzed for trends. All tests were 2‐sided, and a p value < 0.05 was considered to be statistically significant. All statistical analyses were performed using JMP version 7.0 for Windows (SAS Institute).\nContinuous data are presented as mean ± SD or median (interquartile range) as appropriate. Between‐group differences were analyzed with one‐way ANOVA for normally distributed variables followed by Tukey's pairwise test. Categorical data are presented as frequency and percentage, and between‐group comparisons were analyzed using the chi‐square test. Because the distributions of serum AST, ALT, triglycerides, creatinine, plasma hs‐CRP, and GDF1 values were skewed, we used logarithmically transformed values in the analysis. The association between GDF1 and liver fibrosis was evaluated using multivariate logistic regression models: (1) GDF1 and age; (2) GDF1, age, and sex; (3) GDF1, age, sex, and BMI; (4) GDF1, age, sex, BMI, and ALT; (5) GDF1, age, sex, BMI, ALT, hemoglobin, and albumin; and (6) GDF1, age, sex, BMI, ALT, hemoglobin, albumin, creatinine, and hs‐CRP. We then classified the concentration of GDF1 into tertiles, and analyzed trends among the tertiles using general linear and logistic regression analyses. Using the highest tertile as the reference, we also estimated the odds ratio (OR) with 95% confidence interval (CI) of liver fibrosis in each tertile.\nAssociations between plasma GDF1 concentration and other variables were analyzed using simple and multiple linear regression analyses. In addition, the values of FIB‐4 index, AST/ALT ratio and APRI in each GDF1 concentration tertile were analyzed for trends. All tests were 2‐sided, and a p value < 0.05 was considered to be statistically significant. All statistical analyses were performed using JMP version 7.0 for Windows (SAS Institute).", "We enrolled a total of 327 patients with stable angina from July 2012 to December 2021 who visited the cardiovascular clinic at E‐Da Hospital. The inclusion criteria were patients: (1) with stable angina pectoris, defined as effort‐related chest pain without evidence of recent deterioration or rest pain in the previous 6 months, and diagnosed by the cardiologist in charge as previously described;\n14\n, \n15\n and (2) who underwent a successful percutaneous coronary intervention, defined as <30% residual stenosis in a final angiogram as assessed using quantitative coronary angiography with no occlusion of the large branch (>1 mm) or dissection limiting flow, and Thrombolysis in Myocardial Infarction grade 3.\n16\n The exclusion criteria were patients: (1) who used steroids; (2) had inflammatory diseases (such as infection/sepsis), malignancy, liver diseases, and collagen diseases; (3) with a history of psychosis; and (4) with heart valve disease, myocardial infarction, or had undergone heart surgery. In addition, we also excluded patients who were unable or unwilling to provide informed consent. All study patients lived in the same area during the study period, and they were all of Han Chinese ethnicity. Each patient provided written informed consent before being enrolled into the study. This cross‐sectional study was approved by the Human Research Ethics Committee of E‐Da Hospital.", "A detailed interview was conducted with each patient about their medical and personal history, as well as their demographic characteristics before the coronary angiography examination. We classified smoking status as follows: never smokers, former smokers (those who had stopped smoking for ≥1 year), or current smokers. We grouped the current and former smokers in the analysis and compared them with the never smokers. A trained nurse performed all blood pressure readings with the patients seated using an automated blood pressure monitor (HEM‐907; Omron) after 5 min of rest. Anthropometric parameters including body mass index (BMI) and waist circumference were also recorded.", "Fasting (8 h) peripheral blood samples were obtained from the antecubital vein before the patients underwent coronary angiography. Levels of high‐density lipoprotein cholesterol (HDL‐C), low‐density lipoprotein cholesterol (LDL‐C), total cholesterol, plasma triglycerides, serum glucose, serum albumin, serum creatinine, and complete blood cell count were measured in all patients using a parallel, multi‐channel analyzer (Hitachi 7170A) as described previously.\n17\n, \n18\n We used high‐performance liquid chromatography (Tosoh Automated Glycohemoglobin Analyzer, HLC‐723G8) to measure hemoglobin A1c (HbA1c). In addition, we measured levels of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) following the methods of the Japan Society of Clinical Chemistry (“Hitachi” Discrete photometric chemistry analyzer for clinical use, LAbOSPECT 008AS), which is compatible to the methods by the International Federation of Clinical Chemistry. Serum creatinine level was measured using the Jaffe method, and estimated glomerular filtration rate (eGFR) was calculated using the Chronic Kidney Disease Epidemiology Collaboration (CKD‐EPI) equation within 3–6 months of admission.\n19\n The fibrosis‐4 (FIB‐4) index was calculated as: FIB‐4 = age (years) × AST (IU/L)/platelet count (109/L) × √ALT (IU/L).\n20\n The aspartate transaminase/platelet ratio index (APRI) was calculated as: APRI = AST/upper limit of normal of AST/platelet count × 100 (where the upper limit of normal of AST = 40 IU/L).", "After obtaining overnight fasting blood samples, the plasma was kept at −80°C until assay. We used a commercial ELISA kit to measure concentrations of plasma GDF1 (Cloud‐Clone Corp.). The standard and dilution curves were parallel, and the inter‐assay and intra‐assay coefficients of variation for the assay were <12% and <10%, respectively (both n = 3). Plasma levels of hs‐CRP were measured using an immunochemistry system (Beckman Coulter IMMAGE), which had a detection of 0.2 mg/L. All measurements were made in duplicate during a single experiment.", "We used the World Health Organization criteria to define type 2 diabetes, as receiving medical therapy for diabetes and/or a history of type 2 diabetes.\n21\n Patients with a systolic/diastolic blood pressure (SBP/DBP) of ≥140/90 mmHg, or a prescription for antihypertensive drugs were defined as having hypertension. We used the Adult Treatment Panel III criteria\n22\n to define hyperlipidemia as: elevated total cholesterol (≥200 mg/dl), and/or elevated LDL‐C (≥130 mg/dl), and/or low HDL‐C (<35 mg/dl in men or < 39 mg/dl in women), and/or elevated triglycerides (≥150 mg/dl), or being treated for a lipid disorder. Liver fibrosis was defined according to FIB‐4 index categories as: minimal fibrosis (FIB‐4 < 1.45), moderate fibrosis (FIB‐4 1.45–3.25), and advanced fibrosis (FIB‐4 > 3.25).\n20\n, \n23\n, \n24\n\n", "Continuous data are presented as mean ± SD or median (interquartile range) as appropriate. Between‐group differences were analyzed with one‐way ANOVA for normally distributed variables followed by Tukey's pairwise test. Categorical data are presented as frequency and percentage, and between‐group comparisons were analyzed using the chi‐square test. Because the distributions of serum AST, ALT, triglycerides, creatinine, plasma hs‐CRP, and GDF1 values were skewed, we used logarithmically transformed values in the analysis. The association between GDF1 and liver fibrosis was evaluated using multivariate logistic regression models: (1) GDF1 and age; (2) GDF1, age, and sex; (3) GDF1, age, sex, and BMI; (4) GDF1, age, sex, BMI, and ALT; (5) GDF1, age, sex, BMI, ALT, hemoglobin, and albumin; and (6) GDF1, age, sex, BMI, ALT, hemoglobin, albumin, creatinine, and hs‐CRP. We then classified the concentration of GDF1 into tertiles, and analyzed trends among the tertiles using general linear and logistic regression analyses. Using the highest tertile as the reference, we also estimated the odds ratio (OR) with 95% confidence interval (CI) of liver fibrosis in each tertile.\nAssociations between plasma GDF1 concentration and other variables were analyzed using simple and multiple linear regression analyses. In addition, the values of FIB‐4 index, AST/ALT ratio and APRI in each GDF1 concentration tertile were analyzed for trends. All tests were 2‐sided, and a p value < 0.05 was considered to be statistically significant. All statistical analyses were performed using JMP version 7.0 for Windows (SAS Institute).", "[SUBTITLE] Characteristics of the patients by liver fibrosis category [SUBSECTION] Table 1 shows the biochemical and clinical characteristics of the 327 enrolled patients according to liver fibrosis category. The prevalence rates of minimal liver fibrosis (FIB‐4 < 1.45), moderate liver fibrosis (FIB‐4 1.45–3.25), and advanced liver fibrosis (FIB‐4 > 3.25) were 24.5%, 43.1%, and 32.4%, respectively. The advanced liver fibrosis group were older, had higher levels of AST, blood urea nitrogen, and creatinine, and lower BMI, eGFR, albumin, platelets, hematocrit, and hemoglobin than those with minimal and moderate liver fibrosis. Furthermore, the advanced liver fibrosis group had higher ALT, white blood cell count, and hs‐CRP, and a lower rate of drinking alcohol, waist circumference, and GDF1 than the minimal liver fibrosis group. Moreover, the advanced liver fibrosis group had a lower triglyceride level than the moderate liver fibrosis group. No significant differences were found in hypertension, male sex, hyperlipidemia, current smoking, diabetes mellitus, DBP, SBP, fasting glucose, HbA1c, total cholesterol, LDL‐C, HDL‐C, Gensini score or number of diseased coronary arteries among the three groups.\nBaseline characteristics of the study population stratified by severity of liver fibrosis.\n\nNote: Data are mean ± SD or frequency (percentage).\nAbbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; BP, blood pressure; BUN, blood urea nitrogen; eGFR, estimated glomerular filtration rate; FIB, fibrosis; GDF1, growth/differentiation factor 1; HDL, high‐density lipoprotein; Hs‐CRP, high‐sensitivity C‐reactive protein; LDL, low‐density lipoprotein; WBC, white blood cell; WHR, waist‐to‐hip ratio.\nSignificant difference was tested using log‐transformed data.\nTable 1 shows the biochemical and clinical characteristics of the 327 enrolled patients according to liver fibrosis category. The prevalence rates of minimal liver fibrosis (FIB‐4 < 1.45), moderate liver fibrosis (FIB‐4 1.45–3.25), and advanced liver fibrosis (FIB‐4 > 3.25) were 24.5%, 43.1%, and 32.4%, respectively. The advanced liver fibrosis group were older, had higher levels of AST, blood urea nitrogen, and creatinine, and lower BMI, eGFR, albumin, platelets, hematocrit, and hemoglobin than those with minimal and moderate liver fibrosis. Furthermore, the advanced liver fibrosis group had higher ALT, white blood cell count, and hs‐CRP, and a lower rate of drinking alcohol, waist circumference, and GDF1 than the minimal liver fibrosis group. Moreover, the advanced liver fibrosis group had a lower triglyceride level than the moderate liver fibrosis group. No significant differences were found in hypertension, male sex, hyperlipidemia, current smoking, diabetes mellitus, DBP, SBP, fasting glucose, HbA1c, total cholesterol, LDL‐C, HDL‐C, Gensini score or number of diseased coronary arteries among the three groups.\nBaseline characteristics of the study population stratified by severity of liver fibrosis.\n\nNote: Data are mean ± SD or frequency (percentage).\nAbbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; BP, blood pressure; BUN, blood urea nitrogen; eGFR, estimated glomerular filtration rate; FIB, fibrosis; GDF1, growth/differentiation factor 1; HDL, high‐density lipoprotein; Hs‐CRP, high‐sensitivity C‐reactive protein; LDL, low‐density lipoprotein; WBC, white blood cell; WHR, waist‐to‐hip ratio.\nSignificant difference was tested using log‐transformed data.\n[SUBTITLE] Association between plasma GDF1 and liver fibrosis [SUBSECTION] We found a significant association between a lower plasma GDF1 concentration and liver fibrosis, even after controlling for anthropometric factors, ALT, hemoglobin, albumin, creatinine, and hs‐CRP (Table 2). Furthermore, a lower concentration of GDF1 showed a significant linear trend and was independently associated with liver fibrosis, especially when analyzing the concentration both as a continuous variable and by tertile (Tables 2 and 3). In the multiple logistic regression analysis, fully adjusted ORs for liver fibrosis in the second and third tertiles were 2.02 (95% CI, 0.70–6.30) and 5.04 (95% CI, 1.35–25.60), respectively.\nAssociation of plasma GDF1 with liver fibrosis in fully adjusted models\n\nNote: Results of multivariate logistic regression analysis are presented as the odds ratio (OR) of having a liver fibrosis status and decreased plasma GDF1 level.\nAbbreviations: ALT, alanine aminotransferase; BMI, body mass index; GDF1, growth/differentiation factor 1; Hs‐CRP, high‐sensitivity C‐reactive protein.\nUnivariate and multivariate analyses of the impact of plasma GDF1 level on liver fibrosis\n\nNote: Values shown are cut‐off values of plasma GDF1 levels of all subjects, and odds ratios (ORs) with 95% confidence intervals (CIs).\nAbbreviation: GDF1, growth/differentiation factor 1.\nAdjusted for age, sex, body mass index, alanine aminotransferase, hemoglobin, albumin, creatinine, and high‐sensitivity C‐reactive protein.\nWe found a significant association between a lower plasma GDF1 concentration and liver fibrosis, even after controlling for anthropometric factors, ALT, hemoglobin, albumin, creatinine, and hs‐CRP (Table 2). Furthermore, a lower concentration of GDF1 showed a significant linear trend and was independently associated with liver fibrosis, especially when analyzing the concentration both as a continuous variable and by tertile (Tables 2 and 3). In the multiple logistic regression analysis, fully adjusted ORs for liver fibrosis in the second and third tertiles were 2.02 (95% CI, 0.70–6.30) and 5.04 (95% CI, 1.35–25.60), respectively.\nAssociation of plasma GDF1 with liver fibrosis in fully adjusted models\n\nNote: Results of multivariate logistic regression analysis are presented as the odds ratio (OR) of having a liver fibrosis status and decreased plasma GDF1 level.\nAbbreviations: ALT, alanine aminotransferase; BMI, body mass index; GDF1, growth/differentiation factor 1; Hs‐CRP, high‐sensitivity C‐reactive protein.\nUnivariate and multivariate analyses of the impact of plasma GDF1 level on liver fibrosis\n\nNote: Values shown are cut‐off values of plasma GDF1 levels of all subjects, and odds ratios (ORs) with 95% confidence intervals (CIs).\nAbbreviation: GDF1, growth/differentiation factor 1.\nAdjusted for age, sex, body mass index, alanine aminotransferase, hemoglobin, albumin, creatinine, and high‐sensitivity C‐reactive protein.\n[SUBTITLE] Associations between plasma GDF1 level and clinical laboratory data [SUBSECTION] Simple linear regression analysis showed that plasma GDF1 was negatively associated with the FIB‐4 index, blood urea nitrogen, and creatinine, and positively associated with platelet count and eGFR. In multiple linear regression analysis, plasma GDF1 level was positively associated with platelet count, and negatively associated with the FIB‐4 index, blood urea nitrogen, and creatinine (Table 4). In addition, in analysis of the subjects by tertile of GDF1 concentration, the FIB‐4 index, APRI, and AST/ALT ratio were significantly associated with GDF1 concentration (p for trend < 0.05, Figure 1).\nLinear regression analysis of variables associated with plasma log‐GDF1 levels in the study subjects.\nAbbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; BP, blood.pressure; GDF1, growth/differentiation factor 1; GFR, glomerular filtration rate; HDL, high‐density lipoprotein; Hs‐CRP, high‐sensitivity C‐reactive protein; LDL, low‐density lipoprotein.\nRegression coefficient adjusted for age and sex.\nClassification of subjects into tertiles according to growth/differentiation factor 1 (GDF1) concentration revealed that the fibrosis‐4 index (A), APRI (B), and AST/ALT ratio (C) were significantly associated with GDF1 concentration (p for trend < 0.05). Bars represent the mean ± SD. ALT, alanine aminotransferase; APRI, aspartate aminotransferase to platelet ratio index; AST, aspartate aminotransferase.\nSimple linear regression analysis showed that plasma GDF1 was negatively associated with the FIB‐4 index, blood urea nitrogen, and creatinine, and positively associated with platelet count and eGFR. In multiple linear regression analysis, plasma GDF1 level was positively associated with platelet count, and negatively associated with the FIB‐4 index, blood urea nitrogen, and creatinine (Table 4). In addition, in analysis of the subjects by tertile of GDF1 concentration, the FIB‐4 index, APRI, and AST/ALT ratio were significantly associated with GDF1 concentration (p for trend < 0.05, Figure 1).\nLinear regression analysis of variables associated with plasma log‐GDF1 levels in the study subjects.\nAbbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; BP, blood.pressure; GDF1, growth/differentiation factor 1; GFR, glomerular filtration rate; HDL, high‐density lipoprotein; Hs‐CRP, high‐sensitivity C‐reactive protein; LDL, low‐density lipoprotein.\nRegression coefficient adjusted for age and sex.\nClassification of subjects into tertiles according to growth/differentiation factor 1 (GDF1) concentration revealed that the fibrosis‐4 index (A), APRI (B), and AST/ALT ratio (C) were significantly associated with GDF1 concentration (p for trend < 0.05). Bars represent the mean ± SD. ALT, alanine aminotransferase; APRI, aspartate aminotransferase to platelet ratio index; AST, aspartate aminotransferase.", "Table 1 shows the biochemical and clinical characteristics of the 327 enrolled patients according to liver fibrosis category. The prevalence rates of minimal liver fibrosis (FIB‐4 < 1.45), moderate liver fibrosis (FIB‐4 1.45–3.25), and advanced liver fibrosis (FIB‐4 > 3.25) were 24.5%, 43.1%, and 32.4%, respectively. The advanced liver fibrosis group were older, had higher levels of AST, blood urea nitrogen, and creatinine, and lower BMI, eGFR, albumin, platelets, hematocrit, and hemoglobin than those with minimal and moderate liver fibrosis. Furthermore, the advanced liver fibrosis group had higher ALT, white blood cell count, and hs‐CRP, and a lower rate of drinking alcohol, waist circumference, and GDF1 than the minimal liver fibrosis group. Moreover, the advanced liver fibrosis group had a lower triglyceride level than the moderate liver fibrosis group. No significant differences were found in hypertension, male sex, hyperlipidemia, current smoking, diabetes mellitus, DBP, SBP, fasting glucose, HbA1c, total cholesterol, LDL‐C, HDL‐C, Gensini score or number of diseased coronary arteries among the three groups.\nBaseline characteristics of the study population stratified by severity of liver fibrosis.\n\nNote: Data are mean ± SD or frequency (percentage).\nAbbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; BP, blood pressure; BUN, blood urea nitrogen; eGFR, estimated glomerular filtration rate; FIB, fibrosis; GDF1, growth/differentiation factor 1; HDL, high‐density lipoprotein; Hs‐CRP, high‐sensitivity C‐reactive protein; LDL, low‐density lipoprotein; WBC, white blood cell; WHR, waist‐to‐hip ratio.\nSignificant difference was tested using log‐transformed data.", "We found a significant association between a lower plasma GDF1 concentration and liver fibrosis, even after controlling for anthropometric factors, ALT, hemoglobin, albumin, creatinine, and hs‐CRP (Table 2). Furthermore, a lower concentration of GDF1 showed a significant linear trend and was independently associated with liver fibrosis, especially when analyzing the concentration both as a continuous variable and by tertile (Tables 2 and 3). In the multiple logistic regression analysis, fully adjusted ORs for liver fibrosis in the second and third tertiles were 2.02 (95% CI, 0.70–6.30) and 5.04 (95% CI, 1.35–25.60), respectively.\nAssociation of plasma GDF1 with liver fibrosis in fully adjusted models\n\nNote: Results of multivariate logistic regression analysis are presented as the odds ratio (OR) of having a liver fibrosis status and decreased plasma GDF1 level.\nAbbreviations: ALT, alanine aminotransferase; BMI, body mass index; GDF1, growth/differentiation factor 1; Hs‐CRP, high‐sensitivity C‐reactive protein.\nUnivariate and multivariate analyses of the impact of plasma GDF1 level on liver fibrosis\n\nNote: Values shown are cut‐off values of plasma GDF1 levels of all subjects, and odds ratios (ORs) with 95% confidence intervals (CIs).\nAbbreviation: GDF1, growth/differentiation factor 1.\nAdjusted for age, sex, body mass index, alanine aminotransferase, hemoglobin, albumin, creatinine, and high‐sensitivity C‐reactive protein.", "Simple linear regression analysis showed that plasma GDF1 was negatively associated with the FIB‐4 index, blood urea nitrogen, and creatinine, and positively associated with platelet count and eGFR. In multiple linear regression analysis, plasma GDF1 level was positively associated with platelet count, and negatively associated with the FIB‐4 index, blood urea nitrogen, and creatinine (Table 4). In addition, in analysis of the subjects by tertile of GDF1 concentration, the FIB‐4 index, APRI, and AST/ALT ratio were significantly associated with GDF1 concentration (p for trend < 0.05, Figure 1).\nLinear regression analysis of variables associated with plasma log‐GDF1 levels in the study subjects.\nAbbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; BMI, body mass index; BP, blood.pressure; GDF1, growth/differentiation factor 1; GFR, glomerular filtration rate; HDL, high‐density lipoprotein; Hs‐CRP, high‐sensitivity C‐reactive protein; LDL, low‐density lipoprotein.\nRegression coefficient adjusted for age and sex.\nClassification of subjects into tertiles according to growth/differentiation factor 1 (GDF1) concentration revealed that the fibrosis‐4 index (A), APRI (B), and AST/ALT ratio (C) were significantly associated with GDF1 concentration (p for trend < 0.05). Bars represent the mean ± SD. ALT, alanine aminotransferase; APRI, aspartate aminotransferase to platelet ratio index; AST, aspartate aminotransferase.", "According to a previous clinical study, fatty liver disease is common in patients with CAD, and it can progress to liver fibrosis resulting in higher rates of all‐cause and cardiovascular mortality.\n3\n Similarly, in the current study, we found that among the 327 stable angina patients, more than 75% had moderate to advanced liver fibrosis (Table 1). In the past decade, several studies have evaluated the relationship between fatty liver disease and CAD. Although some chronic liver diseases such as NAFLD share similar risk factors with CAD (such as hyperglycemia, insulin resistance, hypertension, inflammation, dyslipidemia, hyperuricemia, hypoadiponectinemia, renal failure, and obesity),\n25\n, \n26\n, \n27\n the underlying molecular relationship between chronic liver disease and CAD is unknown. Clinically, a meta‐analysis found that NAFLD was associated with the severity of atherosclerosis, including coronary calcification levels, carotid intima‐media thickness, arterial stiffness and endothelial dysfunction.\n28\n Furthermore, the review by Meex et al. in 2017 also showed that many hepatokines such as fetuin A, fetuin B, retinol‐binding protein 4, and selenoprotein P are involved in both fatty liver disease and insulin resistance, which in turn influences the process of atherosclerosis.\n29\n However, no previous report has described the underlying molecular pathogenesis between chronic liver disease and CAD. To the best of our knowledge, this is the first study to show that GDF1, which is involved in both post‐infarction cardiac remodeling and the development of fatty liver disease, was associated with liver fibrosis in patients with stable angina.\nThe cytokine system is very complex. Maintaining a balanced healthy environment for all human cells is difficult, and a factor which is beneficial for certain circumstances may be harmful for another. GDF1 is a member of the TGF‐β superfamily and is known to be a protective mediator against the development of post‐infarction cardiac remodeling via negative regulation of the Smad signaling and MEK‐ERK1/2 pathways in the heart.\n11\n, \n30\n, \n31\n A suppressed MEK/ERK1/2 signaling pathway has been associated with fatty liver disease, liver fibrosis, and poor hepatoma differentiation in recent studies.\n12\n, \n13\n In this study, a lower plasma GDF1 concentration was significantly independently associated with and showed a significant linear trend with liver fibrosis (Tables 2 and 3). It is reasonable that a decrease in GDF1 could enhance the progress of liver fibrosis. In addition, the Gensini score and number of diseased coronary arteries, which was used to evaluate CAD severity, did not show a significant difference among tertiles of GDF1 (data not shown). The role of GDF1 post infarction is known, however it remains unclear in CAD.\nIn analysis of the associations between common risk factors for liver fibrosis, we found no significant differences in hypertension, male sex, hyperlipidemia, current smoking, diabetes mellitus, DBP, SBP, LDL‐C, HDL‐C, total cholesterol, HbA1c or fasting glucose among the liver fibrosis severity groups (Table 1). This suggests that the pathogenesis of liver fibrosis in patients with stable angina may have a different mechanism to that of liver fibrosis, even though they share similar risk factors.\nIn this study, we found that plasma GDF1 level was independently associated and showed a significant linear trend with liver fibrosis after controlling for all anthropometric variables, liver enzymes, albumin, renal function and inflammation markers (Tables 2 and 3). In linear regression analysis, we further showed that plasma GDF1 level was positively associated with renal function and liver function and condition (such as albumin and platelet levels), but negatively associated with the FIB‐4 index (Table 4). More importantly, the common CAD risk factors such as BMI, blood pressure, Hba1c, LDL, and hs‐CRP were not associated with GDF1. These findings imply that the role of GDF1 in liver fibrosis in patients with stable angina may be different from the traditional shared risk factors between CAD and NAFLD. Further studies are warranted to elucidate this issue.\nAfter classifying the subjects into tertiles according to plasma GDF1 concentration, we found that GDF1 was associated with the FIB‐4 index and APRI, which is used to predict fibrosis and cirrhosis in patients with chronic hepatitis (Figure 1).\n23\n, \n32\n The average AST/ALT ratio in our patients was 1.78, neither <1 or >2 (Figure 1). This revealed that a high FIB‐4 index in our angina patients was not strongly related to the causes of alcoholic fatty liver disease or NAFLD.\n33\n, \n34\n, \n35\n With regard to the general demographics, we found no significant differences in age (69.3 ± 13.4 vs. 69.9 ± 13.9 vs. 68.6 ± 13.3 years, p = 0.769) or alcohol consumption (22.9% vs. 22.2% vs. 34.6%, p = 0.068) among the tertiles of GDF1 (data not shown).\nThere are several limitations to this study. First, we lacked liver echo evaluation reports and liver biopsy proof. This is because it is not reasonable to ask all stable angina patients to undergo such examinations when there is no strong evidence of liver disease. Second, the role of GDF1 in coronary arteriosclerosis remains unknown. Third, our results showed only that there was an association between plasma GDF1 levels and liver fibrosis. Further investigations are needed to investigate whether the mechanism is through the MEK/ERK1/2 signaling pathway or other underlying pathways. Fourth, if the study population had different diseases (e.g., acute coronary syndrome and myocardial infarction), the diverse condition and disease severity of the study population may have impacted the results. To avoid selection bias, we chose individuals with stable angina for this study, thus the results of the present study might not be generalizable to other populations. Finally, further investigations are also needed to investigate whether other hepatokines are also involved and interact with GDF1 in the liver fibrosis process in patients with stable angina.", "Our results indicated that in patients with stable angina, low plasma GDF1 was associated with liver fibrosis. Further studies are warranted to investigate the association between plasma GDF1 and the pathogenesis of liver fibrosis.", "All authors contributed to this study. W.‐C.H., W.‐H.T., and C.‐C.H. conceived and designed the study. W.‐C.H., W.‐H.T., Y.‐J.L., and C.‐C.H. provided the methodology. F.‐M.C. performed the formal analysis, and project administration. T.‐H.Y., C.‐C.W., and C.‐C.H. validated the data. T.‐H.Y., C.‐C.W., W.‐C.H., and C.‐P.W. performed the investigation, resources, and data curation. T.‐H.Y., W.‐C.H., C.‐C.W., C.‐P.W., Y.‐C.L., and C.‐T.W. prepared the manuscript. W.‐H.T., T.‐H.Y., W.‐C.H., C.‐C.W., C.‐P.W., Y.‐C.L., and C.‐T.W. reviewed and edited the manuscript. W.‐C.H., W.‐H.T., Y.‐J.L., and C.‐C.H. performed the visualization. W.‐C.H. and C.‐C.H. performed the supervision and funding acquisition. All authors have read and agreed to the published version of the manuscript.", "E‐Da Hospital financially supported this research under Contracts EDAHP109002, EDAHI109002, and EDAHI110001.", "The authors have declared that no competing interest exists.", "Each patient provided written informed consent before being enrolled into the study." ]
[ null, "materials-and-methods", null, null, null, null, null, null, "results", null, null, null, "discussion", "conclusions", null, null, "COI-statement", null ]
[ "growth/differentiation factor 1", "liver fibrosis", "MEK‐ERK1/2", "Smad signaling", "stable angina" ]
Clinical features of patients with talaromycosis marneffei and microbiological characteristics of the causative strains.
36268985
Talaromyces marneffei (T. marneffei) is a temperature-dependent dimorphic fungus that is mainly prevalent in Southeast Asia and South China and often causes disseminated life-threatening infections. This study aimed to investigate the clinical features and improve the early diagnosis of talaromycosis marneffei in nonendemic areas.
BACKGROUND
We retrospectively analyzed the medical records of six cases of T. marneffei infection. We describe the clinical manifestations, laboratory tests, and imaging manifestations of the six patients.
METHODS
Talaromyces marneffei infection was confirmed by sputum culture, blood culture, tissue biopsy, and metagenomic next-generation sequencing (mNGS). In this study, there were five disseminated-type patients and two HIV patients. One patient died within 24 h, and the others demonstrated considerable improvement after definitive diagnosis.
RESULTS
Due to the lack of significant clinical presentations of talaromycosis marneffei, many cases may be easily misdiagnosed in nonendemic areas. It is particularly important to analyze the imaging manifestations and laboratory findings of infected patients. With the rapid development of molecular biology, mNGS may be a rapid and effective diagnostic method.
CONCLUSIONS
[ "Humans", "HIV Infections", "Retrospective Studies", "Mycoses", "China", "Antifungal Agents" ]
9701894
null
null
METHODS
[SUBTITLE] Clinical data [SUBSECTION] This was a retrospective study that used data collected from medical records. Six patients were diagnosed and treated at the Ningbo First Hospital, and the patient's inpatient medical records were reviewed. Related data included demographic information (age and sex), clinical features (with or without underlying disease), laboratory data, imaging findings, treatment, and prognoses. The clinical data of the patients are shown in Table 1. Clinical features and laboratory findings of six patients with Talaromyces marneffei infection Abbreviations: ALT, alanine transaminase; AST, aspartate aminotransferase; CRP, C‐reactive protein; ESR, erythrocyte sedimentation rate. This was a retrospective study that used data collected from medical records. Six patients were diagnosed and treated at the Ningbo First Hospital, and the patient's inpatient medical records were reviewed. Related data included demographic information (age and sex), clinical features (with or without underlying disease), laboratory data, imaging findings, treatment, and prognoses. The clinical data of the patients are shown in Table 1. Clinical features and laboratory findings of six patients with Talaromyces marneffei infection Abbreviations: ALT, alanine transaminase; AST, aspartate aminotransferase; CRP, C‐reactive protein; ESR, erythrocyte sedimentation rate. [SUBTITLE] Laboratory tests [SUBSECTION] Routine blood examination, HIV antibody, C‐reactive protein (CRP), blood coagulation, erythrocyte sedimentation rate, and blood biochemistry tests were performed with standardized testing equipment and kits. Routine blood examination, HIV antibody, C‐reactive protein (CRP), blood coagulation, erythrocyte sedimentation rate, and blood biochemistry tests were performed with standardized testing equipment and kits. [SUBTITLE] Identification methods of T. marneffei [SUBSECTION] There were three methods used for pathogen examination. [SUBTITLE] Pathogen culture [SUBSECTION] Cultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast. Cultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast. [SUBTITLE] Microscopy observation [SUBSECTION] Talaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining. Talaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining. [SUBTITLE] Metagenomic next‐generation sequencing [SUBSECTION] The metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results. The metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results. There were three methods used for pathogen examination. [SUBTITLE] Pathogen culture [SUBSECTION] Cultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast. Cultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast. [SUBTITLE] Microscopy observation [SUBSECTION] Talaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining. Talaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining. [SUBTITLE] Metagenomic next‐generation sequencing [SUBSECTION] The metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results. The metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results.
RESULTS
[SUBTITLE] Clinical manifestation [SUBSECTION] Six patients were diagnosed and treated at the Ningbo First Hospital, all of whom had lived in Ningbo for a long time. There were two female and four male patients, and all had no epidemiological history and no history of exposure to bamboo rats. The median age of the patients was 60 years. The most common clinical symptoms of patients infected with T. marneffei were fever and anemia, followed by fungemia, malaise, respiratory symptoms, and weight loss (Table 2). The primary presentation for one patient was gastrointestinal symptoms (such as abdominal pain, diarrhea, and bloody stools). Five patients exhibited disseminated T. marneffei infections; three patients had primary lesions in the lungs, and two patients presented with subcutaneous nodules or abscesses (Table 1). All had various underlying diseases, including chronic obstructive pulmonary disease, multiple myeloma, diabetes, and HIV. Notably, four cases were initially misdiagnosed as pneumonia or pulmonary tuberculosis. Clinical features of patients with talaromycosis marneffei Note: Cured: the resolution of all symptoms and mycological eradication; Recovered: improvement in most signs and symptoms; Relapsed: no clinical improvement, clinical condition deterioration, or death. Abbreviations: BALF, bronchoalveolar lavage fluid; COPD, chronic obstructive pulmonary disease; F, female; M, male; TM, talaromycosis marneffei. Six patients were diagnosed and treated at the Ningbo First Hospital, all of whom had lived in Ningbo for a long time. There were two female and four male patients, and all had no epidemiological history and no history of exposure to bamboo rats. The median age of the patients was 60 years. The most common clinical symptoms of patients infected with T. marneffei were fever and anemia, followed by fungemia, malaise, respiratory symptoms, and weight loss (Table 2). The primary presentation for one patient was gastrointestinal symptoms (such as abdominal pain, diarrhea, and bloody stools). Five patients exhibited disseminated T. marneffei infections; three patients had primary lesions in the lungs, and two patients presented with subcutaneous nodules or abscesses (Table 1). All had various underlying diseases, including chronic obstructive pulmonary disease, multiple myeloma, diabetes, and HIV. Notably, four cases were initially misdiagnosed as pneumonia or pulmonary tuberculosis. Clinical features of patients with talaromycosis marneffei Note: Cured: the resolution of all symptoms and mycological eradication; Recovered: improvement in most signs and symptoms; Relapsed: no clinical improvement, clinical condition deterioration, or death. Abbreviations: BALF, bronchoalveolar lavage fluid; COPD, chronic obstructive pulmonary disease; F, female; M, male; TM, talaromycosis marneffei. [SUBTITLE] Laboratory tests [SUBSECTION] C‐reactive protein (CRP) and erythrocyte sedimentation rate were significantly elevated in six patients, and the white blood cell count was increased in four patients and decreased in one patient. In addition, five patients had varying degrees of anemia, all below 100 g/L; four patients had varying degrees of decreased albumin, with the lowest value being 22.4 g/L; and three patients were tested for lymphocyte subpopulation, which reflected low immune function in two patients and normal immune function in the other (Table 3). Two patients tested positive for HIV antibody (primary screening) and were diagnosed with HIV infection by the Centers for Disease Control and Prevention. Three patients had no significant abnormalities in liver and kidney function. Analysis of lymphocyte subsets in three patients with talaromycosis marneffei Note: Normal ranges: CD3: 58.4%–81.56%; CD4: 31%–60%; CD8: 13%–41%; CD4/CD8: 0.8–4.2; NK: 14%–40%; CD19: 5%–25%. C‐reactive protein (CRP) and erythrocyte sedimentation rate were significantly elevated in six patients, and the white blood cell count was increased in four patients and decreased in one patient. In addition, five patients had varying degrees of anemia, all below 100 g/L; four patients had varying degrees of decreased albumin, with the lowest value being 22.4 g/L; and three patients were tested for lymphocyte subpopulation, which reflected low immune function in two patients and normal immune function in the other (Table 3). Two patients tested positive for HIV antibody (primary screening) and were diagnosed with HIV infection by the Centers for Disease Control and Prevention. Three patients had no significant abnormalities in liver and kidney function. Analysis of lymphocyte subsets in three patients with talaromycosis marneffei Note: Normal ranges: CD3: 58.4%–81.56%; CD4: 31%–60%; CD8: 13%–41%; CD4/CD8: 0.8–4.2; NK: 14%–40%; CD19: 5%–25%. [SUBTITLE] Imaging and other examinations [SUBSECTION] Among six patients, chest imaging showed abnormal lesions in the lungs of five patients for the first time (Table 4). There were multiple enlarged lymph nodes in the mediastinum and pulmonary hila in two patients (Figure 1A), multiple high‐density plaques and strips in two patients (Figure 1B), multiple patchy ground‐glass opacities in one patient (Figure 1C), and bilateral pleural effusion in three patients (Figure 1D). High‐resolution computed tomography among six patients infected with Talaromyces marneffei. Chest CT images among patients infected with Talaromyces marneffei. (A) The blue arrow indicates enlarged mediastinal lymph nodes (mediastinal window). (B) The blue arrow indicates high‐density plaque and strip. (C) Blue arrows indicate multiple patchy ground‐glass opacities. (D) Blue arrows indicate bilateral pleural effusion. Case 4 was diagnosed with talaromycosis marneffei involving the gastrointestinal tract. A colonoscopy revealed colon erosion, hyperemia, edema, and multiple intestinal mucosal ulcers (Figure 2A). Mucosal biopsy of transverse colon and descending colon tissue was performed (Figure 2B). After a period of antifungal treatment at our hospital, a repeat colonoscopy showed multiple ulcers in the colon in the healing stage, which indicated that the patient's condition had improved. Colonoscopy and histopathological examination. (A) The mucosa had apparent hyperemia and edema, as well as scattered erosion and ulceration, according to a colonoscopy. (B) Colon mucosa was stained with hexamine silver staining, showing several yeast‐like organisms with septate forms (indicated by the blue arrow). Among six patients, chest imaging showed abnormal lesions in the lungs of five patients for the first time (Table 4). There were multiple enlarged lymph nodes in the mediastinum and pulmonary hila in two patients (Figure 1A), multiple high‐density plaques and strips in two patients (Figure 1B), multiple patchy ground‐glass opacities in one patient (Figure 1C), and bilateral pleural effusion in three patients (Figure 1D). High‐resolution computed tomography among six patients infected with Talaromyces marneffei. Chest CT images among patients infected with Talaromyces marneffei. (A) The blue arrow indicates enlarged mediastinal lymph nodes (mediastinal window). (B) The blue arrow indicates high‐density plaque and strip. (C) Blue arrows indicate multiple patchy ground‐glass opacities. (D) Blue arrows indicate bilateral pleural effusion. Case 4 was diagnosed with talaromycosis marneffei involving the gastrointestinal tract. A colonoscopy revealed colon erosion, hyperemia, edema, and multiple intestinal mucosal ulcers (Figure 2A). Mucosal biopsy of transverse colon and descending colon tissue was performed (Figure 2B). After a period of antifungal treatment at our hospital, a repeat colonoscopy showed multiple ulcers in the colon in the healing stage, which indicated that the patient's condition had improved. Colonoscopy and histopathological examination. (A) The mucosa had apparent hyperemia and edema, as well as scattered erosion and ulceration, according to a colonoscopy. (B) Colon mucosa was stained with hexamine silver staining, showing several yeast‐like organisms with septate forms (indicated by the blue arrow). [SUBTITLE] Diagnosis of patients with talaromycosis marneffei [SUBSECTION] Six cases were diagnosed with talaromycosis marneffei by culture of bronchoalveolar lavage fluid and sputum specimens, blood microscopy, bronchoalveolar lavage fluid mNGS, intestinal lesion mNGS, and blood mNGS. [SUBTITLE] Culture and microscopy observation [SUBSECTION] The specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4). Culture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400). Blood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000). The specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4). Culture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400). Blood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000). [SUBTITLE] Metagenomic next‐generation sequencing [SUBSECTION] Among six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5). Microorganism detected by mNGS of two patients Abbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing. Among six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5). Microorganism detected by mNGS of two patients Abbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing. Six cases were diagnosed with talaromycosis marneffei by culture of bronchoalveolar lavage fluid and sputum specimens, blood microscopy, bronchoalveolar lavage fluid mNGS, intestinal lesion mNGS, and blood mNGS. [SUBTITLE] Culture and microscopy observation [SUBSECTION] The specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4). Culture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400). Blood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000). The specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4). Culture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400). Blood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000). [SUBTITLE] Metagenomic next‐generation sequencing [SUBSECTION] Among six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5). Microorganism detected by mNGS of two patients Abbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing. Among six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5). Microorganism detected by mNGS of two patients Abbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing.
null
null
[ "INTRODUCTION", "Clinical data", "Laboratory tests", "Identification methods of T. marneffei\n", "Pathogen culture", "Microscopy observation", "Metagenomic next‐generation sequencing", "Clinical manifestation", "Laboratory tests", "Imaging and other examinations", "Diagnosis of patients with talaromycosis marneffei", "Culture and microscopy observation", "Metagenomic next‐generation sequencing", "TREATMENT AND PROGNOSIS", "AUTHOR CONTRIBUTIONS", "FUNDING INFORMATION" ]
[ "\nTalaromyces marneffei is a rare pathogenic thermally dimorphic fungus that grows as mold at 25°C and as yeast at 37°C.\n1\n The resistance of the host to invasion by T. marneffei may be based on cellular immunity, with the T‐cell‐mediated Thl‐type response pattern playing an important role in the defense of the host against fungal infection.\n2\n It mainly invades the human monocyte–macrophage system and causes infections in patients with immune dysfunction (e.g., immunosuppressed, immunocompromised, or immunodeficient patients). The disease is predominant in Guangxi, Hong Kong, and Taiwan but rare in the city of Ningbo in Zhejiang Province.\n3\n, \n4\n It has become the most common opportunistic fungal infection in Southeast Asia (especially in AIDS patients).\n5\n However, in recent years, there have been increasing reports of cases among non‐HIV‐infected patients.\n6\n In this article, we describe the clinical and laboratory features of six patients.", "This was a retrospective study that used data collected from medical records. Six patients were diagnosed and treated at the Ningbo First Hospital, and the patient's inpatient medical records were reviewed. Related data included demographic information (age and sex), clinical features (with or without underlying disease), laboratory data, imaging findings, treatment, and prognoses. The clinical data of the patients are shown in Table 1.\nClinical features and laboratory findings of six patients with Talaromyces marneffei infection\nAbbreviations: ALT, alanine transaminase; AST, aspartate aminotransferase; CRP, C‐reactive protein; ESR, erythrocyte sedimentation rate.", "Routine blood examination, HIV antibody, C‐reactive protein (CRP), blood coagulation, erythrocyte sedimentation rate, and blood biochemistry tests were performed with standardized testing equipment and kits.", "There were three methods used for pathogen examination.\n[SUBTITLE] Pathogen culture [SUBSECTION] Cultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast.\nCultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast.\n[SUBTITLE] Microscopy observation [SUBSECTION] \nTalaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining.\n\nTalaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining.\n[SUBTITLE] Metagenomic next‐generation sequencing [SUBSECTION] The metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results.\nThe metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results.", "Cultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast.", "\nTalaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining.", "The metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results.", "Six patients were diagnosed and treated at the Ningbo First Hospital, all of whom had lived in Ningbo for a long time. There were two female and four male patients, and all had no epidemiological history and no history of exposure to bamboo rats. The median age of the patients was 60 years.\nThe most common clinical symptoms of patients infected with T. marneffei were fever and anemia, followed by fungemia, malaise, respiratory symptoms, and weight loss (Table 2). The primary presentation for one patient was gastrointestinal symptoms (such as abdominal pain, diarrhea, and bloody stools). Five patients exhibited disseminated T. marneffei infections; three patients had primary lesions in the lungs, and two patients presented with subcutaneous nodules or abscesses (Table 1). All had various underlying diseases, including chronic obstructive pulmonary disease, multiple myeloma, diabetes, and HIV. Notably, four cases were initially misdiagnosed as pneumonia or pulmonary tuberculosis.\nClinical features of patients with talaromycosis marneffei\n\nNote: Cured: the resolution of all symptoms and mycological eradication; Recovered: improvement in most signs and symptoms; Relapsed: no clinical improvement, clinical condition deterioration, or death.\nAbbreviations: BALF, bronchoalveolar lavage fluid; COPD, chronic obstructive pulmonary disease; F, female; M, male; TM, talaromycosis marneffei.", "C‐reactive protein (CRP) and erythrocyte sedimentation rate were significantly elevated in six patients, and the white blood cell count was increased in four patients and decreased in one patient. In addition, five patients had varying degrees of anemia, all below 100 g/L; four patients had varying degrees of decreased albumin, with the lowest value being 22.4 g/L; and three patients were tested for lymphocyte subpopulation, which reflected low immune function in two patients and normal immune function in the other (Table 3). Two patients tested positive for HIV antibody (primary screening) and were diagnosed with HIV infection by the Centers for Disease Control and Prevention. Three patients had no significant abnormalities in liver and kidney function.\nAnalysis of lymphocyte subsets in three patients with talaromycosis marneffei\n\nNote: Normal ranges: CD3: 58.4%–81.56%; CD4: 31%–60%; CD8: 13%–41%; CD4/CD8: 0.8–4.2; NK: 14%–40%; CD19: 5%–25%.", "Among six patients, chest imaging showed abnormal lesions in the lungs of five patients for the first time (Table 4). There were multiple enlarged lymph nodes in the mediastinum and pulmonary hila in two patients (Figure 1A), multiple high‐density plaques and strips in two patients (Figure 1B), multiple patchy ground‐glass opacities in one patient (Figure 1C), and bilateral pleural effusion in three patients (Figure 1D).\nHigh‐resolution computed tomography among six patients infected with Talaromyces marneffei.\nChest CT images among patients infected with Talaromyces marneffei. (A) The blue arrow indicates enlarged mediastinal lymph nodes (mediastinal window). (B) The blue arrow indicates high‐density plaque and strip. (C) Blue arrows indicate multiple patchy ground‐glass opacities. (D) Blue arrows indicate bilateral pleural effusion.\nCase 4 was diagnosed with talaromycosis marneffei involving the gastrointestinal tract. A colonoscopy revealed colon erosion, hyperemia, edema, and multiple intestinal mucosal ulcers (Figure 2A). Mucosal biopsy of transverse colon and descending colon tissue was performed (Figure 2B). After a period of antifungal treatment at our hospital, a repeat colonoscopy showed multiple ulcers in the colon in the healing stage, which indicated that the patient's condition had improved.\nColonoscopy and histopathological examination. (A) The mucosa had apparent hyperemia and edema, as well as scattered erosion and ulceration, according to a colonoscopy. (B) Colon mucosa was stained with hexamine silver staining, showing several yeast‐like organisms with septate forms (indicated by the blue arrow).", "Six cases were diagnosed with talaromycosis marneffei by culture of bronchoalveolar lavage fluid and sputum specimens, blood microscopy, bronchoalveolar lavage fluid mNGS, intestinal lesion mNGS, and blood mNGS.\n[SUBTITLE] Culture and microscopy observation [SUBSECTION] The specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4).\nCulture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400).\nBlood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000).\nThe specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4).\nCulture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400).\nBlood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000).\n[SUBTITLE] Metagenomic next‐generation sequencing [SUBSECTION] Among six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5).\nMicroorganism detected by mNGS of two patients\nAbbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing.\nAmong six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5).\nMicroorganism detected by mNGS of two patients\nAbbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing.", "The specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4).\nCulture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400).\nBlood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000).", "Among six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5).\nMicroorganism detected by mNGS of two patients\nAbbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing.", "Amphotericin B, itraconazole, and voriconazole are routinely utilized for talaromycosis marneffei in clinical treatment. In terms of clinical response and fungicidal action, amphotericin B was superior to itraconazole in the initial treatment.\n7\n Voriconazole was reported to be effective for disseminated T. marneffei infection.\n8\n Thus, most of the cases in this study were treated with intravenous amphotericin B at a dose of 0.6 mg/kg/day for 2 weeks, followed by intravenous voriconazole at a dose of 200 mg/day for maintenance therapy. One mild case was treated with intravenous voriconazole at a dose of 200 mg q12 h for the first 24 h, followed by oral therapy at 200 mg twice daily. To reduce the incidence of adverse effects, liver function, renal function, and body electrolytes were monitored closely and regularly during treatment.\nDuring the treatment period, adverse drug reactions to amphotericin B were observed. Two cases showed elevated creatinine levels. These patients were treated with lower doses of amphotericin B, or the drug was discontinued and replaced with voriconazole therapy. Only one patient did not receive antifungal medication and died within 24 h of admission due to deterioration of the disease. During the 6‐month follow‐up, most of the cases in this study showed improvement after treatment and had a good prognosis. One case is still under treatment and follow‐up.", "All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by Lei Peng and Xing‐bei Weng. The first draft of the article was written by Lei Peng, and all authors commented on previous versions of the article. All authors read and approved the final article.", "This work was supported by the Ningbo Natural Science Foundation (2019A610381) and the Ningbo Public Welfare Foundation (2019C50087)." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "METHODS", "Clinical data", "Laboratory tests", "Identification methods of T. marneffei\n", "Pathogen culture", "Microscopy observation", "Metagenomic next‐generation sequencing", "RESULTS", "Clinical manifestation", "Laboratory tests", "Imaging and other examinations", "Diagnosis of patients with talaromycosis marneffei", "Culture and microscopy observation", "Metagenomic next‐generation sequencing", "TREATMENT AND PROGNOSIS", "DISCUSSION", "AUTHOR CONTRIBUTIONS", "FUNDING INFORMATION", "CONFLICT OF INTEREST" ]
[ "\nTalaromyces marneffei is a rare pathogenic thermally dimorphic fungus that grows as mold at 25°C and as yeast at 37°C.\n1\n The resistance of the host to invasion by T. marneffei may be based on cellular immunity, with the T‐cell‐mediated Thl‐type response pattern playing an important role in the defense of the host against fungal infection.\n2\n It mainly invades the human monocyte–macrophage system and causes infections in patients with immune dysfunction (e.g., immunosuppressed, immunocompromised, or immunodeficient patients). The disease is predominant in Guangxi, Hong Kong, and Taiwan but rare in the city of Ningbo in Zhejiang Province.\n3\n, \n4\n It has become the most common opportunistic fungal infection in Southeast Asia (especially in AIDS patients).\n5\n However, in recent years, there have been increasing reports of cases among non‐HIV‐infected patients.\n6\n In this article, we describe the clinical and laboratory features of six patients.", "[SUBTITLE] Clinical data [SUBSECTION] This was a retrospective study that used data collected from medical records. Six patients were diagnosed and treated at the Ningbo First Hospital, and the patient's inpatient medical records were reviewed. Related data included demographic information (age and sex), clinical features (with or without underlying disease), laboratory data, imaging findings, treatment, and prognoses. The clinical data of the patients are shown in Table 1.\nClinical features and laboratory findings of six patients with Talaromyces marneffei infection\nAbbreviations: ALT, alanine transaminase; AST, aspartate aminotransferase; CRP, C‐reactive protein; ESR, erythrocyte sedimentation rate.\nThis was a retrospective study that used data collected from medical records. Six patients were diagnosed and treated at the Ningbo First Hospital, and the patient's inpatient medical records were reviewed. Related data included demographic information (age and sex), clinical features (with or without underlying disease), laboratory data, imaging findings, treatment, and prognoses. The clinical data of the patients are shown in Table 1.\nClinical features and laboratory findings of six patients with Talaromyces marneffei infection\nAbbreviations: ALT, alanine transaminase; AST, aspartate aminotransferase; CRP, C‐reactive protein; ESR, erythrocyte sedimentation rate.\n[SUBTITLE] Laboratory tests [SUBSECTION] Routine blood examination, HIV antibody, C‐reactive protein (CRP), blood coagulation, erythrocyte sedimentation rate, and blood biochemistry tests were performed with standardized testing equipment and kits.\nRoutine blood examination, HIV antibody, C‐reactive protein (CRP), blood coagulation, erythrocyte sedimentation rate, and blood biochemistry tests were performed with standardized testing equipment and kits.\n[SUBTITLE] Identification methods of T. marneffei\n [SUBSECTION] There were three methods used for pathogen examination.\n[SUBTITLE] Pathogen culture [SUBSECTION] Cultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast.\nCultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast.\n[SUBTITLE] Microscopy observation [SUBSECTION] \nTalaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining.\n\nTalaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining.\n[SUBTITLE] Metagenomic next‐generation sequencing [SUBSECTION] The metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results.\nThe metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results.\nThere were three methods used for pathogen examination.\n[SUBTITLE] Pathogen culture [SUBSECTION] Cultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast.\nCultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast.\n[SUBTITLE] Microscopy observation [SUBSECTION] \nTalaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining.\n\nTalaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining.\n[SUBTITLE] Metagenomic next‐generation sequencing [SUBSECTION] The metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results.\nThe metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results.", "This was a retrospective study that used data collected from medical records. Six patients were diagnosed and treated at the Ningbo First Hospital, and the patient's inpatient medical records were reviewed. Related data included demographic information (age and sex), clinical features (with or without underlying disease), laboratory data, imaging findings, treatment, and prognoses. The clinical data of the patients are shown in Table 1.\nClinical features and laboratory findings of six patients with Talaromyces marneffei infection\nAbbreviations: ALT, alanine transaminase; AST, aspartate aminotransferase; CRP, C‐reactive protein; ESR, erythrocyte sedimentation rate.", "Routine blood examination, HIV antibody, C‐reactive protein (CRP), blood coagulation, erythrocyte sedimentation rate, and blood biochemistry tests were performed with standardized testing equipment and kits.", "There were three methods used for pathogen examination.\n[SUBTITLE] Pathogen culture [SUBSECTION] Cultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast.\nCultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast.\n[SUBTITLE] Microscopy observation [SUBSECTION] \nTalaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining.\n\nTalaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining.\n[SUBTITLE] Metagenomic next‐generation sequencing [SUBSECTION] The metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results.\nThe metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results.", "Cultures of clinical specimens, including blood, sputum, and bronchoalveolar lavage fluid, were established on Sabouraud agar medium at 25°C and 37°C. Identification was based upon the morphology of the colonies. At 25°C, T. marneffei grew as a mold and created a soluble red pigment that diffused into the agar. At 37°C, it developed as a yeast.", "\nTalaromyces marneffei was identified by cytology and histopathology from clinical specimens by hexamine silver staining and Wright–Giemsa staining.", "The metagenomic next‐generation sequencing (mNGS) method is based on the Illumina sequencing platform and polymerase chain reaction (PCR) free library building technology. In this study, clinical specimens were sent to specialized testing institutions for different sample processing procedures. This approach mainly includes specimen pretreatment, nucleic acid extraction, library preparation, online sequencing, database matching, and interpretation of results.", "[SUBTITLE] Clinical manifestation [SUBSECTION] Six patients were diagnosed and treated at the Ningbo First Hospital, all of whom had lived in Ningbo for a long time. There were two female and four male patients, and all had no epidemiological history and no history of exposure to bamboo rats. The median age of the patients was 60 years.\nThe most common clinical symptoms of patients infected with T. marneffei were fever and anemia, followed by fungemia, malaise, respiratory symptoms, and weight loss (Table 2). The primary presentation for one patient was gastrointestinal symptoms (such as abdominal pain, diarrhea, and bloody stools). Five patients exhibited disseminated T. marneffei infections; three patients had primary lesions in the lungs, and two patients presented with subcutaneous nodules or abscesses (Table 1). All had various underlying diseases, including chronic obstructive pulmonary disease, multiple myeloma, diabetes, and HIV. Notably, four cases were initially misdiagnosed as pneumonia or pulmonary tuberculosis.\nClinical features of patients with talaromycosis marneffei\n\nNote: Cured: the resolution of all symptoms and mycological eradication; Recovered: improvement in most signs and symptoms; Relapsed: no clinical improvement, clinical condition deterioration, or death.\nAbbreviations: BALF, bronchoalveolar lavage fluid; COPD, chronic obstructive pulmonary disease; F, female; M, male; TM, talaromycosis marneffei.\nSix patients were diagnosed and treated at the Ningbo First Hospital, all of whom had lived in Ningbo for a long time. There were two female and four male patients, and all had no epidemiological history and no history of exposure to bamboo rats. The median age of the patients was 60 years.\nThe most common clinical symptoms of patients infected with T. marneffei were fever and anemia, followed by fungemia, malaise, respiratory symptoms, and weight loss (Table 2). The primary presentation for one patient was gastrointestinal symptoms (such as abdominal pain, diarrhea, and bloody stools). Five patients exhibited disseminated T. marneffei infections; three patients had primary lesions in the lungs, and two patients presented with subcutaneous nodules or abscesses (Table 1). All had various underlying diseases, including chronic obstructive pulmonary disease, multiple myeloma, diabetes, and HIV. Notably, four cases were initially misdiagnosed as pneumonia or pulmonary tuberculosis.\nClinical features of patients with talaromycosis marneffei\n\nNote: Cured: the resolution of all symptoms and mycological eradication; Recovered: improvement in most signs and symptoms; Relapsed: no clinical improvement, clinical condition deterioration, or death.\nAbbreviations: BALF, bronchoalveolar lavage fluid; COPD, chronic obstructive pulmonary disease; F, female; M, male; TM, talaromycosis marneffei.\n[SUBTITLE] Laboratory tests [SUBSECTION] C‐reactive protein (CRP) and erythrocyte sedimentation rate were significantly elevated in six patients, and the white blood cell count was increased in four patients and decreased in one patient. In addition, five patients had varying degrees of anemia, all below 100 g/L; four patients had varying degrees of decreased albumin, with the lowest value being 22.4 g/L; and three patients were tested for lymphocyte subpopulation, which reflected low immune function in two patients and normal immune function in the other (Table 3). Two patients tested positive for HIV antibody (primary screening) and were diagnosed with HIV infection by the Centers for Disease Control and Prevention. Three patients had no significant abnormalities in liver and kidney function.\nAnalysis of lymphocyte subsets in three patients with talaromycosis marneffei\n\nNote: Normal ranges: CD3: 58.4%–81.56%; CD4: 31%–60%; CD8: 13%–41%; CD4/CD8: 0.8–4.2; NK: 14%–40%; CD19: 5%–25%.\nC‐reactive protein (CRP) and erythrocyte sedimentation rate were significantly elevated in six patients, and the white blood cell count was increased in four patients and decreased in one patient. In addition, five patients had varying degrees of anemia, all below 100 g/L; four patients had varying degrees of decreased albumin, with the lowest value being 22.4 g/L; and three patients were tested for lymphocyte subpopulation, which reflected low immune function in two patients and normal immune function in the other (Table 3). Two patients tested positive for HIV antibody (primary screening) and were diagnosed with HIV infection by the Centers for Disease Control and Prevention. Three patients had no significant abnormalities in liver and kidney function.\nAnalysis of lymphocyte subsets in three patients with talaromycosis marneffei\n\nNote: Normal ranges: CD3: 58.4%–81.56%; CD4: 31%–60%; CD8: 13%–41%; CD4/CD8: 0.8–4.2; NK: 14%–40%; CD19: 5%–25%.\n[SUBTITLE] Imaging and other examinations [SUBSECTION] Among six patients, chest imaging showed abnormal lesions in the lungs of five patients for the first time (Table 4). There were multiple enlarged lymph nodes in the mediastinum and pulmonary hila in two patients (Figure 1A), multiple high‐density plaques and strips in two patients (Figure 1B), multiple patchy ground‐glass opacities in one patient (Figure 1C), and bilateral pleural effusion in three patients (Figure 1D).\nHigh‐resolution computed tomography among six patients infected with Talaromyces marneffei.\nChest CT images among patients infected with Talaromyces marneffei. (A) The blue arrow indicates enlarged mediastinal lymph nodes (mediastinal window). (B) The blue arrow indicates high‐density plaque and strip. (C) Blue arrows indicate multiple patchy ground‐glass opacities. (D) Blue arrows indicate bilateral pleural effusion.\nCase 4 was diagnosed with talaromycosis marneffei involving the gastrointestinal tract. A colonoscopy revealed colon erosion, hyperemia, edema, and multiple intestinal mucosal ulcers (Figure 2A). Mucosal biopsy of transverse colon and descending colon tissue was performed (Figure 2B). After a period of antifungal treatment at our hospital, a repeat colonoscopy showed multiple ulcers in the colon in the healing stage, which indicated that the patient's condition had improved.\nColonoscopy and histopathological examination. (A) The mucosa had apparent hyperemia and edema, as well as scattered erosion and ulceration, according to a colonoscopy. (B) Colon mucosa was stained with hexamine silver staining, showing several yeast‐like organisms with septate forms (indicated by the blue arrow).\nAmong six patients, chest imaging showed abnormal lesions in the lungs of five patients for the first time (Table 4). There were multiple enlarged lymph nodes in the mediastinum and pulmonary hila in two patients (Figure 1A), multiple high‐density plaques and strips in two patients (Figure 1B), multiple patchy ground‐glass opacities in one patient (Figure 1C), and bilateral pleural effusion in three patients (Figure 1D).\nHigh‐resolution computed tomography among six patients infected with Talaromyces marneffei.\nChest CT images among patients infected with Talaromyces marneffei. (A) The blue arrow indicates enlarged mediastinal lymph nodes (mediastinal window). (B) The blue arrow indicates high‐density plaque and strip. (C) Blue arrows indicate multiple patchy ground‐glass opacities. (D) Blue arrows indicate bilateral pleural effusion.\nCase 4 was diagnosed with talaromycosis marneffei involving the gastrointestinal tract. A colonoscopy revealed colon erosion, hyperemia, edema, and multiple intestinal mucosal ulcers (Figure 2A). Mucosal biopsy of transverse colon and descending colon tissue was performed (Figure 2B). After a period of antifungal treatment at our hospital, a repeat colonoscopy showed multiple ulcers in the colon in the healing stage, which indicated that the patient's condition had improved.\nColonoscopy and histopathological examination. (A) The mucosa had apparent hyperemia and edema, as well as scattered erosion and ulceration, according to a colonoscopy. (B) Colon mucosa was stained with hexamine silver staining, showing several yeast‐like organisms with septate forms (indicated by the blue arrow).\n[SUBTITLE] Diagnosis of patients with talaromycosis marneffei [SUBSECTION] Six cases were diagnosed with talaromycosis marneffei by culture of bronchoalveolar lavage fluid and sputum specimens, blood microscopy, bronchoalveolar lavage fluid mNGS, intestinal lesion mNGS, and blood mNGS.\n[SUBTITLE] Culture and microscopy observation [SUBSECTION] The specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4).\nCulture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400).\nBlood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000).\nThe specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4).\nCulture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400).\nBlood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000).\n[SUBTITLE] Metagenomic next‐generation sequencing [SUBSECTION] Among six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5).\nMicroorganism detected by mNGS of two patients\nAbbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing.\nAmong six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5).\nMicroorganism detected by mNGS of two patients\nAbbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing.\nSix cases were diagnosed with talaromycosis marneffei by culture of bronchoalveolar lavage fluid and sputum specimens, blood microscopy, bronchoalveolar lavage fluid mNGS, intestinal lesion mNGS, and blood mNGS.\n[SUBTITLE] Culture and microscopy observation [SUBSECTION] The specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4).\nCulture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400).\nBlood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000).\nThe specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4).\nCulture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400).\nBlood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000).\n[SUBTITLE] Metagenomic next‐generation sequencing [SUBSECTION] Among six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5).\nMicroorganism detected by mNGS of two patients\nAbbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing.\nAmong six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5).\nMicroorganism detected by mNGS of two patients\nAbbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing.", "Six patients were diagnosed and treated at the Ningbo First Hospital, all of whom had lived in Ningbo for a long time. There were two female and four male patients, and all had no epidemiological history and no history of exposure to bamboo rats. The median age of the patients was 60 years.\nThe most common clinical symptoms of patients infected with T. marneffei were fever and anemia, followed by fungemia, malaise, respiratory symptoms, and weight loss (Table 2). The primary presentation for one patient was gastrointestinal symptoms (such as abdominal pain, diarrhea, and bloody stools). Five patients exhibited disseminated T. marneffei infections; three patients had primary lesions in the lungs, and two patients presented with subcutaneous nodules or abscesses (Table 1). All had various underlying diseases, including chronic obstructive pulmonary disease, multiple myeloma, diabetes, and HIV. Notably, four cases were initially misdiagnosed as pneumonia or pulmonary tuberculosis.\nClinical features of patients with talaromycosis marneffei\n\nNote: Cured: the resolution of all symptoms and mycological eradication; Recovered: improvement in most signs and symptoms; Relapsed: no clinical improvement, clinical condition deterioration, or death.\nAbbreviations: BALF, bronchoalveolar lavage fluid; COPD, chronic obstructive pulmonary disease; F, female; M, male; TM, talaromycosis marneffei.", "C‐reactive protein (CRP) and erythrocyte sedimentation rate were significantly elevated in six patients, and the white blood cell count was increased in four patients and decreased in one patient. In addition, five patients had varying degrees of anemia, all below 100 g/L; four patients had varying degrees of decreased albumin, with the lowest value being 22.4 g/L; and three patients were tested for lymphocyte subpopulation, which reflected low immune function in two patients and normal immune function in the other (Table 3). Two patients tested positive for HIV antibody (primary screening) and were diagnosed with HIV infection by the Centers for Disease Control and Prevention. Three patients had no significant abnormalities in liver and kidney function.\nAnalysis of lymphocyte subsets in three patients with talaromycosis marneffei\n\nNote: Normal ranges: CD3: 58.4%–81.56%; CD4: 31%–60%; CD8: 13%–41%; CD4/CD8: 0.8–4.2; NK: 14%–40%; CD19: 5%–25%.", "Among six patients, chest imaging showed abnormal lesions in the lungs of five patients for the first time (Table 4). There were multiple enlarged lymph nodes in the mediastinum and pulmonary hila in two patients (Figure 1A), multiple high‐density plaques and strips in two patients (Figure 1B), multiple patchy ground‐glass opacities in one patient (Figure 1C), and bilateral pleural effusion in three patients (Figure 1D).\nHigh‐resolution computed tomography among six patients infected with Talaromyces marneffei.\nChest CT images among patients infected with Talaromyces marneffei. (A) The blue arrow indicates enlarged mediastinal lymph nodes (mediastinal window). (B) The blue arrow indicates high‐density plaque and strip. (C) Blue arrows indicate multiple patchy ground‐glass opacities. (D) Blue arrows indicate bilateral pleural effusion.\nCase 4 was diagnosed with talaromycosis marneffei involving the gastrointestinal tract. A colonoscopy revealed colon erosion, hyperemia, edema, and multiple intestinal mucosal ulcers (Figure 2A). Mucosal biopsy of transverse colon and descending colon tissue was performed (Figure 2B). After a period of antifungal treatment at our hospital, a repeat colonoscopy showed multiple ulcers in the colon in the healing stage, which indicated that the patient's condition had improved.\nColonoscopy and histopathological examination. (A) The mucosa had apparent hyperemia and edema, as well as scattered erosion and ulceration, according to a colonoscopy. (B) Colon mucosa was stained with hexamine silver staining, showing several yeast‐like organisms with septate forms (indicated by the blue arrow).", "Six cases were diagnosed with talaromycosis marneffei by culture of bronchoalveolar lavage fluid and sputum specimens, blood microscopy, bronchoalveolar lavage fluid mNGS, intestinal lesion mNGS, and blood mNGS.\n[SUBTITLE] Culture and microscopy observation [SUBSECTION] The specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4).\nCulture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400).\nBlood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000).\nThe specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4).\nCulture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400).\nBlood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000).\n[SUBTITLE] Metagenomic next‐generation sequencing [SUBSECTION] Among six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5).\nMicroorganism detected by mNGS of two patients\nAbbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing.\nAmong six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5).\nMicroorganism detected by mNGS of two patients\nAbbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing.", "The specimens were inoculated into Sabouraud agar medium and incubated at 25°C and 37°C. The fungus appeared as a mold with red wine pigment at 25°C (Figure 3A), and curved conidial chains were evident under the microscope. The cultures were stained with lactophenol cotton blue, which revealed smooth transparent branched separated mycelia typical of broom‐like branches, with multiple scattered nonparallel peduncle bases with 2–10 vial peduncles on them and narrowed apically with scattered single chains of conidia (Figure 3B). It grew as yeast without pigment yield at 37°C, and fungal cells under the microscope presented as round or oval‐shaped yeast‐like cells with a diameter of 3 μm (Figure 2B). The fungus was identified by its characteristics. Peripheral blood smear was selected for direct microscopic examination after Wright–Giemsa staining. Round, oval, or sausage‐shaped mounded or scattered organisms could be seen in neutrophils and macrophages (Figure 4).\nCulture and microscopy observation of Talaromyces marneffei. (A) Morphology and wine pigment of T. marneffei colonies from alveolar lavage fluid culture on Sabouraud agar medium for 7 days at 25°C. (B) Typical broom‐shaped, branching septate hyphae of T. marneffei at 25°C with lactophenol cotton blue staining (×400).\nBlood smear examination. Blood smears were stained with Wright‐Giemsa staining, which showed small round‐to‐ovoid yeast cells within the neutrophil cytoplasm. Yeast cells were unevenly stained and showed strong staining at one end (indicated by the blue arrow) (×1000).", "Among six patients, only two patients were diagnosed with talaromycosis marneffei by mNGS. Due to exacerbation of the condition and multiple negative sample cultures, clinicians collected related samples for mNGS. mNGS of the BALF of case 3 revealed 38 T. marneffei reads, and mNGS of the intestinal lesion of case 4 revealed 27,935 T. marneffei reads. In addition, the presence of T. marneffei was also detected in their blood by mNGS (Table 5).\nMicroorganism detected by mNGS of two patients\nAbbreviations: BALF, bronchoalveolar lavage fluid; mNGS, metagenomic next‐generation sequencing.", "Amphotericin B, itraconazole, and voriconazole are routinely utilized for talaromycosis marneffei in clinical treatment. In terms of clinical response and fungicidal action, amphotericin B was superior to itraconazole in the initial treatment.\n7\n Voriconazole was reported to be effective for disseminated T. marneffei infection.\n8\n Thus, most of the cases in this study were treated with intravenous amphotericin B at a dose of 0.6 mg/kg/day for 2 weeks, followed by intravenous voriconazole at a dose of 200 mg/day for maintenance therapy. One mild case was treated with intravenous voriconazole at a dose of 200 mg q12 h for the first 24 h, followed by oral therapy at 200 mg twice daily. To reduce the incidence of adverse effects, liver function, renal function, and body electrolytes were monitored closely and regularly during treatment.\nDuring the treatment period, adverse drug reactions to amphotericin B were observed. Two cases showed elevated creatinine levels. These patients were treated with lower doses of amphotericin B, or the drug was discontinued and replaced with voriconazole therapy. Only one patient did not receive antifungal medication and died within 24 h of admission due to deterioration of the disease. During the 6‐month follow‐up, most of the cases in this study showed improvement after treatment and had a good prognosis. One case is still under treatment and follow‐up.", "\nTalaromyces marneffei is a species of penicillium isolated from the liver of bamboo rats that died unnaturally in Vietnam in 1956. In 1959, the fungus was named Penicillium marneffei in honor of Hubert Marneffe, the director of the Pasteur Institute in Indochina.\n9\n In 2011, based on phylogenetic and phenotypic analysis, the name was changed to T. marneffei.\n10\n Bamboo rats are currently considered natural hosts or carriers of this pathogen.\n11\n Regarding the patients' epidemiological history, it was found that no patients had a history of residence in endemic areas or exposure to bamboo rats. The original route of infection is currently unknown, but it could be through skin lesions or conidia inhalation. It has been documented that T. marneffei enters the blood system by invading the reticuloendothelial system and then spreads to other organs through the lymphatic system and blood circulation.\n12\n, \n13\n\nT. marneffei mainly invades organs and sites related to the monocyte–macrophage reticuloendothelial system. T. marneffei infections can be divided into disseminated and focal types; focal type infections are often confined to the site of invasion, and the clinical manifestations are dominated by the symptoms of the original disease. The disseminated type often involves multiple tissues and organs. Common clinical manifestations of disseminated T. marneffei include fever, respiratory signs, anemia, weight loss, skin lesions, lymphadenopathy, and hepatosplenomegaly.\n3\n In this study, five patients exhibited disseminated T. marneffei infections. These patients presented with fever, cough, lymph node enlargement, anemia, and gastrointestinal symptoms, all of which were nonspecific and led to a misdiagnosis of tuberculosis or other fungal infections. Skin lesions are a clinical feature of disseminated T. marneffei, and characteristic lesions are papules with central necrosis.\n9\n Two patients in our cohort presented with atypical skin lesions, such as subcutaneous nodules or abscesses. In three cases, the lungs were affected by fungal invasion first. In addition, the possibility of gastrointestinal infection was not excluded. Although lymphoid tissue is extensive throughout the digestive tract, talaromycosis marneffei infection of the intestine is uncommon.\n13\n The atypical and nonspecific clinical presentation of fungal infections in T. marneffei often adds to the difficulty of early diagnosis, resulting in diagnostic delay and increased mortality.\nT lymphocyte cells are critical for controlling T. marneffei infection.\n14\n Some experiments have indicated that CD4+ T cells mediate significant pulmonary inflammatory infiltration in mice by recruiting macrophages and granulocytes. CD4 deficiency may lead to the loss of Th2 anti‐inflammatory cytokines (IL‐4, IL‐5, and IL‐10).\n15\n In the innate immune system, activated macrophages can activate some proinflammatory cytokines and chemokines.\n16\n Myeloperoxidase‐dependent neutrophils also have fungicidal activity through the actions of reactive oxygen species.\n17\n In HIV‐positive individuals, T. marneffei‐activated monocyte‐derived dendritic cells (MDDCs) may further accelerate immunosuppression by promoting HIV‐1 transfection of CD4+ T cells.\n18\n Overall, the inflammatory and regulatory responses to a fungal infection are significantly modulated by the innate and acquired immune systems. Lymphocyte subpopulation analysis should be performed in clinical work, especially in immunosuppressed or HIV‐positive patients.\nThe pathogenesis of T. marneffei involves the ability of the fungus to avoid being killed by macrophages and replicate inside them.\n19\n\nT. marneffei can produce melanin or melanin‐like compounds to enhance resistance to phagocytosis by macrophages.\n20\n After phagocytosis by macrophages, T. marneffei (as an intracellular yeast form) induces the production of superoxide dismutase and catalase‐peroxidase in response to oxidative stress.\n19\n Other factors may also be associated with the pathogenicity of T. marneffei, including heat shock proteins (Hsp70 and Hsp30),\n19\n secreted galactomannan protein Mp1p\n21\n and the expression of isocitrate lyase.\n22\n Subsequently, conidial replication and establishment of infection mediate disseminated infections.\nThe incidence of infection in HIV‐negative patients is increasing. For example, in 2012, Yongxuan Hu reviewed 668 cases of talaromycosis marneffei in mainland China from January 1984 to December 2009, and there were 82 (12.3%) HIV‐negative infected patients.\n3\n In 2015, Ye Qiu retrospectively analyzed 109 patients admitted to the First Affiliated Hospital of Guangxi Medical University for talaromycosis marneffei from January 1, 2003, to August 1, 2014, 39.45% of whom were HIV‐negative.\n23\n In HIV‐negative individuals, infection factors are most likely attributed to underlying conditions, including immunodeficiency due to anti‐IFN‐γ autoantibodies, immunosuppressive therapy, malignancy, diabetes mellitus, and organ transplantation.\n24\n Anti‐IFN‐γ autoantibodies can inhibit STAT1 phosphorylation and IL‐12 production, increasing the risk of fungal infection.\n25\n Other predisposing factors may be related to lymphocyte depletion and impairment of lymphocyte proliferation. Interestingly, one of our cases was an immunocompetent patient, and the infection factor was most likely attributed to the patient's diabetes (Table 2).\nIn terms of laboratory tests, all patients had elevated serum CRP levels and erythrocyte sedimentation rates (Table 1). Some patients had liver damage, characterized by elevated aspartate aminotransferase (AST), especially severe patients. HIV‐positive patients were more likely to have leukopenia, low platelet counts, elevation of alanine transaminase, and positive blood cultures. In addition, the chest imaging manifestations of patients with T. marneffei infection were diverse, including multiple patches or large consolidations, ground‐glass opacities, pleural effusion, and hilar and mediastinal lymph node enlargement (Table 1). Infectious diseases such as tuberculosis, Aspergillus pneumonia, lymphoma, lung cancer, and others should be ruled out.\n26\n\n\nThe traditional methods for diagnosis of talaromycosis marneffei include culture of the pathogen and histopathological examination.\n1\n However, the culture positivity rate was not high in patients with early infection. These sample culture results were presented as repeated negative, including a blood culture of case 3 and a stool culture of case 4. Among the six cases, two were diagnosed with talaromycosis marneffei by mNGS. One case was diagnosed with talaromycosis marneffei by examination of a peripheral blood smear (Figure 4). Yeast‐like organisms may be seen on the blood smear, especially in patients with heavy fungemia.\n27\n Blood culture was positive in only two patients in our study (Table 2). Whereas it usually takes 3–10 days for a patient's clinical specimen to be cultured for T. marneffei, the time from admission to diagnosis was long for most patients in our study. During this period, some patients were misdiagnosed with bacterial pneumonia or other diseases. Moreover, they received improper treatment. When none of their conditions improved significantly, physicians began to suspect the initial diagnosis and considered mNGS. Other techniques, such as novel Mp1p enzyme immunoassay and the beta‐D‐glucan test, have a certain role in the auxiliary diagnosis of talaromycosis marneffei.\n28\n, \n29\n However, these approaches have the drawbacks of extensive processes and limited sensitivity. Several real‐time PCR techniques to identify T. marneffei have been developed in response to the demand for faster and more sensitive diagnostics, although these are limited to primer and probe design.\n30\n, \n31\n Under such circumstances, the implementation of NGS in the clinical field might have some undeniable advantages.\nmNGS, a rapid and effective diagnostic approach, can obtain information on microbial species and abundance in samples by high‐throughput sequencing of nucleic acids and bioinformatics analysis.\n32\n It not only comprehensively identifies pathogens but also offers a significantly reduced time requirement for pathogen identification. In this study, clinicians chose mNGS in the following situations: (1) difficulty identifying fungal pathogens despite repeated cultures and (2) a need for early and precise diagnosis in patients with severe pathogenic infections. Multiple studies have reported the use of mNGS in clinical fungal diagnosis, which also reveals the potential merits of this technique in rapid etiological diagnosis.\n32\n, \n33\n, \n34\n, \n35\n, \n36\n However, there are some shortcomings in the widespread application of mNGS, such as high cost and a lack of recognized standards in this area.\nTalaromycosis marneffei is an uncommon disease that causes high mortality rates. A previous study showed that HIV infection, CD4/CD8 < 0.5, a reduced percentage of CD4+ T cells and late diagnosis were potential risk factors for poor prognosis.\n37\n Late diagnosis is the leading cause of death in T. marneffei‐infected individuals.\n38\n It is clear that early and rapid diagnosis is particularly important. Therefore, clinicians should consider talaromycosis marneffei if a patient has suffered recurrent fever; multiple patches, nodules, or masses in the lungs; gastrointestinal symptoms; and characteristic skin lesions, especially if anti‐inflammatory treatment is ineffective. Pathogen cultivation and other relevant laboratory tests are essential. In addition, we recommend the early use of mNGS to reduce mortality and prolong survival time. There are some limitations of this study. First, it is a retrospective cohort study, and the limited number of cases makes it difficult to explore the differences in clinical manifestations between HIV‐positive and HIV‐negative populations. Second, there is a lack of comprehensive analysis of the infection mechanism of T. marneffei in nonendemic areas.\nIn conclusion, we analyzed six cases of talaromycosis marneffei in nonendemic areas in the hope of promoting awareness and familiarity with this rare disease among more clinicians in nonendemic areas and improving the prognosis of patients.", "All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by Lei Peng and Xing‐bei Weng. The first draft of the article was written by Lei Peng, and all authors commented on previous versions of the article. All authors read and approved the final article.", "This work was supported by the Ningbo Natural Science Foundation (2019A610381) and the Ningbo Public Welfare Foundation (2019C50087).", "The authors declare no competing interests." ]
[ null, "methods", null, null, null, null, null, null, "results", null, null, null, null, null, null, null, "discussion", null, null, "COI-statement" ]
[ "disseminated Talaromyces marneffei infection", "laboratory test", "metagenomic next‐generation sequencing", "\nTalaromyces marneffei\n", "talaromycosis marneffei" ]
An International Consensus on Actions to Improve Lung Cancer Survival: A Modified Delphi Method Among Clinical Experts in the International Cancer Benchmarking Partnership.
36269109
Research from the International Cancer Benchmarking Partnership (ICBP) demonstrates that international variation in lung cancer survival persists, particularly within early stage disease. There is a lack of international consensus on the critical contributing components to variation in lung cancer outcomes and the steps needed to optimise lung cancer services. These are needed to improve the quality of options for and equitable access to treatment, and ultimately improve survival.
BACKGROUND
Semi-structured interviews were conducted with 9 key informants from ICBP countries. An international clinical network representing 6 ICBP countries (Australia, Canada, Denmark, England, Ireland, New Zealand, Northern Ireland, Scotland & Wales) was established to share local clinical insights and examples of best practice. Using a modified Delphi consensus model, network members suggested and rated recommendations to optimise the management of lung cancer. Calls to Action were developed via Delphi voting as the most crucial recommendations, with Good Practice Points included to support their implementation.
METHODS
Five Calls to Action and thirteen Good Practice Points applicable to high income, comparable countries were developed and achieved 100% consensus. Calls to Action include (1) Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives; (2) Ensure diagnosis of lung cancer within 30 days of referral; (3) Develop Thoracic Centres of Excellence; (4) Undertake an international audit of lung cancer care; and (5) Recognise improvements in lung cancer care and outcomes as a priority in cancer policy.
RESULTS
The recommendations presented are the voice of an expert international lung cancer clinical network, and signpost key considerations for policymakers in countries within the ICBP but also in other comparable high-income countries. These define a roadmap to help align and focus efforts in improving outcomes and management of lung cancer patients globally.
CONCLUSION
[ "Humans", "Benchmarking", "Lung Neoplasms", "Consensus", "Early Detection of Cancer", "Delphi Technique" ]
9596933
Introduction
Lung cancer accounts for nearly 1.8 million deaths annually – more than colorectal and breast cancers combined.1 Despite significant improvements in diagnosis and treatment, lung cancer is typically associated with low survival internationally.2 The International Cancer Benchmarking Partnership (ICBP) is a global collaboration of clinicians, policymakers, researchers, and cancer data experts. It seeks to benchmark and explain cancer survival, incidence, and mortality differences between high-income countries with comprehensive cancer registry coverage, similar budgetary spending on national health systems and universal access to healthcare. Recent ICBP research has demonstrated that international disparities in lung cancer survival persist, with 5-year survival ranging from 14.7% (UK) to 21.7% (Canada).2 Evidence shows that 3-year survival varies mostly in potentially curative lung cancer, by up to 20 percentage points for localised and regional disease (as per SEER summary staging)3 across the ICBP countries.4 Comparably, survival varied by just 3.4 percentage points in patients diagnosed with distant/metastatic disease across the same countries.4 Due to the inherent complexity of health systems, it is unlikely that one single component of the cancer care continuum can ultimately define why survival variation exists, yet the ability to diagnose and stage lung cancer patients as early as possible will likely influence their treatment options.5 Various work has been undertaken to articulate patient, healthcare professional and disease related factors that influence timeliness of diagnosis and ultimately, patient outcomes.6 This understanding has helped build initiatives across the pathway to address the poorer outcomes in lung cancer, but to differing levels internationally. This starts with preventative initiatives for lung cancer, including support for smoking cessation and lung cancer awareness campaigns. Both areas are of significance, with tobacco being a key factor in the aetiology of lung cancer, and some awareness campaigns influencing stage shift.7,8 Variation in primary care practitioner readiness to refer patients with potential symptoms and in service provision of key diagnostic and staging tools for lung cancer, such as PET-CT, have been demonstrated across ICBP countries with unknown quantifiable impacts upon survival.9,10 This is reflected in the variation seen in lung cancer stage distribution between ICBP countries, but not to the same extent as differences in survival by stage.4 In the diagnostic phase, it is clear that international differences exist for lung cancer patients, yet the evidence for how this affects outcomes is variable and complex.11 Variation within the diagnosis and staging of early stage lung cancer patients is important; unwarranted delays enabling tumour growth may shift patients away from being in a potentially curable stage towards more advanced stages with lower survival.12,13 Curative intent treatment options for lung cancer have increased and become significantly more sophisticated over recent years, with the development and adoption of more precise radiotherapy, and minimally invasive surgery.14 Less invasive options have opened up better opportunities for more complex patients or those with greater comorbidities. Guidance and uptake of these treatment options for early stage patients is likely to vary both within countries and internationally. This is particularly the case in patients deemed to be borderline candidates for curative therapy where there are more variables playing a role in decision-making for treatment.15 Variation in the use of curative intent therapies within early stage patients has been demonstrated in some countries.16 Disease stage, patient fitness/performance status, lung function, staffing skillset, socioeconomic status and comorbidities play key influential roles in the perceived operability of patients and subsequent treatment decisions.16 The organisation of each country’s healthcare system has been established to guide patients from presentation to the decision for treatment, with many informal and formal treatment pathways being established. However, gaps and biases within each system have been identified that undoubtedly affect the management of potentially curable patients.17 There is a current lack of internationally agreed recommendations for improving care of lung cancer patients, with the purpose of being interpreted at a policy level to align efforts to empower those in positions to instigate change. Within this study, we have established an international lung cancer network to share local clinical insights, including locally defined best practices, mitigation of modifiable variables and opportunities for collaboration. We present a series of recommendations to improve lung cancer care, in order to improve survival, based on the consensus of our international clinical network. We hope that these recommendations serve to optimise lung cancer services so as to improve the quality of options for treatment, equitable access to treatment, and ultimately improve survival internationally.
null
null
Results
[SUBTITLE] Initial Interview Results [SUBSECTION] These are reported in the Supplementary Materials. Key themes in the differences reported between countries include: variation in data capture and auditing, composition and use of multidisciplinary teams, access to mitigating programmes for vulnerable populations and access to clinical research. The remainder of Results section focuses on the recommendations developed during this study as these are our core output. These are reported in the Supplementary Materials. Key themes in the differences reported between countries include: variation in data capture and auditing, composition and use of multidisciplinary teams, access to mitigating programmes for vulnerable populations and access to clinical research. The remainder of Results section focuses on the recommendations developed during this study as these are our core output. [SUBTITLE] Recommendations [SUBSECTION] A total of 18 recommendations achieved 100% consensus. These were further grouped in to five Calls to Action (Box 1A) and thirteen Good Practice Points (Box 1B). A total of 18 recommendations achieved 100% consensus. These were further grouped in to five Calls to Action (Box 1A) and thirteen Good Practice Points (Box 1B). [SUBTITLE] Calls to Action Summary [SUBSECTION] Calls to Action should be interpreted as key priorities to inform policy not only across the ICBP countries, but in similar high-income countries globally. It is recognised that different countries will have differing resource, capacity, funding, and population-based needs and should action these recommendations accordingly to their local settings.Box 1A: Calls to ActionImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy [SUBTITLE] Box 1A: Calls to Action [SUBSECTION] Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives Ensure diagnosis of lung cancer within 30 days of referral Develop Thoracic Centres of Excellence Undertake an international audit of lung cancer care Recognise improvements in lung cancer care and outcomes as a priority in cancer policy Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives Ensure diagnosis of lung cancer within 30 days of referral Develop Thoracic Centres of Excellence Undertake an international audit of lung cancer care Recognise improvements in lung cancer care and outcomes as a priority in cancer policy Calls to Action should be interpreted as key priorities to inform policy not only across the ICBP countries, but in similar high-income countries globally. It is recognised that different countries will have differing resource, capacity, funding, and population-based needs and should action these recommendations accordingly to their local settings.Box 1A: Calls to ActionImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy [SUBTITLE] Box 1A: Calls to Action [SUBSECTION] Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives Ensure diagnosis of lung cancer within 30 days of referral Develop Thoracic Centres of Excellence Undertake an international audit of lung cancer care Recognise improvements in lung cancer care and outcomes as a priority in cancer policy Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives Ensure diagnosis of lung cancer within 30 days of referral Develop Thoracic Centres of Excellence Undertake an international audit of lung cancer care Recognise improvements in lung cancer care and outcomes as a priority in cancer policy [SUBTITLE] Good Practice Points Summary [SUBSECTION] The network agreed these points are important considerations for similar high-income countries in optimising management and treatment of lung cancer patients. They should be considered as key in supporting the development and implementation of the Calls to Action.Box 1B: Good Practice PointsPre-Diagnosis• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.Diagnosis• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.Therapeutic• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.All Phases of Care• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. [SUBTITLE] Box 1B: Good Practice Points [SUBSECTION] [SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer. • National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer. • National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. [SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners • Protocols for incorporation into workplans, coordination and chairing of meetings • Appropriate attendance from core stakeholders • Means to record and report treatment decisions locally and centrally • Method to communicate treatment decisions to referring practitioners • Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners • Protocols for incorporation into workplans, coordination and chairing of meetings • Appropriate attendance from core stakeholders • Means to record and report treatment decisions locally and centrally • Method to communicate treatment decisions to referring practitioners • Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. [SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate. • Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised. • Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients. • It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate. • Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised. • Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients. • It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. [SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates. • Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels • Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice. • Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes. • System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates. • Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels • Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice. • Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes. • System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. [SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer. • National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer. • National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. [SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners • Protocols for incorporation into workplans, coordination and chairing of meetings • Appropriate attendance from core stakeholders • Means to record and report treatment decisions locally and centrally • Method to communicate treatment decisions to referring practitioners • Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners • Protocols for incorporation into workplans, coordination and chairing of meetings • Appropriate attendance from core stakeholders • Means to record and report treatment decisions locally and centrally • Method to communicate treatment decisions to referring practitioners • Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. [SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate. • Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised. • Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients. • It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate. • Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised. • Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients. • It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. [SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates. • Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels • Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice. • Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes. • System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates. • Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels • Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice. • Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes. • System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. The network agreed these points are important considerations for similar high-income countries in optimising management and treatment of lung cancer patients. They should be considered as key in supporting the development and implementation of the Calls to Action.Box 1B: Good Practice PointsPre-Diagnosis• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.Diagnosis• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.Therapeutic• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.All Phases of Care• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. [SUBTITLE] Box 1B: Good Practice Points [SUBSECTION] [SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer. • National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer. • National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. [SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners • Protocols for incorporation into workplans, coordination and chairing of meetings • Appropriate attendance from core stakeholders • Means to record and report treatment decisions locally and centrally • Method to communicate treatment decisions to referring practitioners • Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners • Protocols for incorporation into workplans, coordination and chairing of meetings • Appropriate attendance from core stakeholders • Means to record and report treatment decisions locally and centrally • Method to communicate treatment decisions to referring practitioners • Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. [SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate. • Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised. • Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients. • It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate. • Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised. • Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients. • It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. [SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates. • Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels • Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice. • Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes. • System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates. • Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels • Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice. • Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes. • System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. [SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer. • National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer. • National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes. [SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners • Protocols for incorporation into workplans, coordination and chairing of meetings • Appropriate attendance from core stakeholders • Means to record and report treatment decisions locally and centrally • Method to communicate treatment decisions to referring practitioners • Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners • Protocols for incorporation into workplans, coordination and chairing of meetings • Appropriate attendance from core stakeholders • Means to record and report treatment decisions locally and centrally • Method to communicate treatment decisions to referring practitioners • Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation. [SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate. • Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised. • Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients. • It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate. • Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised. • Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients. • It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed. [SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates. • Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels • Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice. • Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes. • System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics. • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates. • Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels • Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice. • Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes. • System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.
Conclusion
The burden of lung cancer is significant and represents a global public health challenge. The Calls to Action and Good Practice Points presented here, reflecting the views of an international expert lung cancer clinical network, signpost the key considerations for policymakers within high-income comparable countries to effective change. They provide a roadmap to help align and focus efforts in improving outcomes and management of lung cancer patients. The Good Practice Points described are key to enable delivery of the Calls to Action, which should be considered the highest priority. Collaboration at local, regional, national, and international levels within the lung cancer community is imperative to empower policymakers to ensure policies are enacted that enhance lung cancer patient treatment and improve quality of life and patient outcomes.
[ "Network Formation", "Topic Guide Development and Initial Interviews", "Roundtable Discussions and Recommendation Development", "Modified Delphi Method", "Refinement of Recommendations", "Initial Interview Results", "Recommendations", "Calls to Action Summary", "Box 1A: Calls to Action", "Good Practice Points Summary", "Box 1B: Good Practice Points", "Pre-Diagnosis", "Diagnosis", "Therapeutic", "All Phases of Care", "Implement Cost-Effective, Clinically Efficacious, and Equitable Lung Cancer Screening Initiatives", "Ensure Diagnosis of Lung Cancer Within 30 days of Referral", "Develop Thoracic Centres of Excellence", "Undertake an International Audit of Lung Cancer Care", "Recognise Improvements in Lung Cancer Care and Outcomes as a Priority in Cancer Policy", "Limitations" ]
[ "Nine network members were recruited from nine ICBP jurisdictions: Australia, Canada, Denmark, England, Ireland, New Zealand, Northern Ireland, Scotland and Wales. Members were identified via purposive sampling based on them having roles in national, regional, and local tiers of health systems with expertise primarily being thoracic surgery, radiation oncology, clinical oncology, or respiratory medicine. Existing ICBP clinical networks and stakeholders were approached and asked for suggestions for the network, and desk-based research was conducted to assess suitability based on the criteria outlined in the previous sentence. Members were initially invited to join the network via email, and if interested, they were sent a Terms of Reference and invited to a telephone call to discuss the aims of the project and the ICBP in more detail. At that point, members were asked if they agreed to be part of the network and involved in the project.", "We initially employed a qualitative approach to understand potential contributing factors to international variation in lung cancer outcomes that could form the basis of the recommendations developed. The overarching focus of this was mostly on non-small cell lung cancer (NSCLC), and for early stage lung cancer. Semi-structured interviews were conducted with the network members to identify key themes and points of variation between the ICBP countries. Interview questions (Supplementary Materials) were developed by the lead study team (Finley, Lynch, Butler, Harrison), alongside an evidence scoping exercise and review of published literature. Interviews were recorded via Microsoft Teams and stored securely at Cancer Research UK. Qualitative data was transcribed by the lead study team and interview transcripts were analysed thematically, drawing on documents developed during processes undertaken to improve existing national guidelines and support for improving lung cancer care, largely the Pan-Canadian Standards for Thoracic Surgery.18 The analysis of these interviews helped generate the recommendations.", "Four roundtable discussions were hosted by the lead study team with all network members. The first and second roundtables discussed the results from the interviews and shared the key themes emerging, which helped structure the discussion to begin generating the recommendations. Network members agreed on key topics for the recommendations to be based on (e.g. pre-diagnosis and screening), and the lead study team articulated these into written recommendations. The third and fourth roundtable meetings were hosted to receive feedback on the Delphi rating method (described below) and further refine the recommendations.", "Network members were asked to rate recommendations using a modified Delphi consensus model (Figure 1).19 In the first round, interviewees were asked to rate 22 recommendations developed from the interview results and initial suggestions from the first and second roundtable discussions with the network members. Options to ‘agree’, ‘disagree’ and provide additional comments and/or suggestions within a free text box were available. A minimum satisfactory consensus of 70% was reached in the first round, with agreement on all recommendations. Subsequent roundtable discussions were hosted to obtain further feedback and consolidate agreement on any recommendations disagreed with or requiring modification. In the next round, 20 revised recommendations were shared with the same participants to rate agreement with the same format (agree, disagree, additional comments/suggestions). The recommendations were then further refined, with a final round of consensus voting resulting in 18 final recommendations.Figure 1.Stepwise modified Delphi Method.\nStepwise modified Delphi Method.", "‘Calls to Action’ for each phase of care were identified by the network as the most crucial recommendations via Delphi consensus voting, requiring the most attention by and focus of policymakers, with ‘Good Practice Points’ being required to support the implementation of these Calls to Action. Good Practice points were further categorised broadly into phases of care (pre-diagnosis, diagnosis, therapeutic and all phases) for ease of implementation.", "These are reported in the Supplementary Materials. Key themes in the differences reported between countries include: variation in data capture and auditing, composition and use of multidisciplinary teams, access to mitigating programmes for vulnerable populations and access to clinical research.\nThe remainder of Results section focuses on the recommendations developed during this study as these are our core output.", "A total of 18 recommendations achieved 100% consensus. These were further grouped in to five Calls to Action (Box 1A) and thirteen Good Practice Points (Box 1B).", "Calls to Action should be interpreted as key priorities to inform policy not only across the ICBP countries, but in similar high-income countries globally. It is recognised that different countries will have differing resource, capacity, funding, and population-based needs and should action these recommendations accordingly to their local settings.Box 1A: Calls to ActionImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\n[SUBTITLE] Box 1A: Calls to Action [SUBSECTION] Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives\nEnsure diagnosis of lung cancer within 30 days of referral\nDevelop Thoracic Centres of Excellence\nUndertake an international audit of lung cancer care\nRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives\nEnsure diagnosis of lung cancer within 30 days of referral\nDevelop Thoracic Centres of Excellence\nUndertake an international audit of lung cancer care\nRecognise improvements in lung cancer care and outcomes as a priority in cancer policy", "Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives\nEnsure diagnosis of lung cancer within 30 days of referral\nDevelop Thoracic Centres of Excellence\nUndertake an international audit of lung cancer care\nRecognise improvements in lung cancer care and outcomes as a priority in cancer policy", "The network agreed these points are important considerations for similar high-income countries in optimising management and treatment of lung cancer patients. They should be considered as key in supporting the development and implementation of the Calls to Action.Box 1B: Good Practice PointsPre-Diagnosis• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.Diagnosis• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.Therapeutic• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.All Phases of Care• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n[SUBTITLE] Box 1B: Good Practice Points [SUBSECTION] [SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n[SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n[SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n[SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n[SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n[SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n[SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n[SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.", "[SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n[SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n[SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n[SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.", "• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.", "• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.", "• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.", "• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.", "Due to variation seen in stage distribution between ICBP countries, it is important to consider where opportunities exist to optimise care and improve survival in the pre-treatment interval. Whilst the evidence is strong for a survival benefit of low dose CT screening in individuals with higher risk of lung cancer, implementation has been a complex issue for policymakers.22-24 Whilst low dose CT screening can support detection of early stage disease and increase the likelihood of curable treatment, several factors must be carefully considered to ensure clinical efficacy and cost effectiveness. Integration into existing infrastructure, target populations, recruitment of population at risk, reduction of harm, resource and cost, and engagement with the public and healthcare professionals need to be defined and implemented. Screening programmes need to be designed to reach a traditionally ‘hard to reach’ population, requiring their design to have greater engagement of those affected. There is no “one size fits all” approach, yet it is the collective opinion of this international clinical network that understanding the impact that screening could have for outcomes and considering all these factors within local contexts is a crucial activity for policymakers. Currently, no ICBP country has fully implemented a national lung cancer screening programme, though a number of countries are at different stages of exploration and implementation. Successful models of assessment and pilot trials have been underway internationally for several years and can serve as a baseline to inform the best implementation plan for screening in each jurisdiction.25-29", "Various temporal targets for key time points within the cancer patient journey exist for different countries, but there has previously been a lack of international consensus on a universal ‘best practice’ target. Unwarranted delays in diagnosis can rapidly shift lung cancer patients from being potentially curative to being difficult to treat, highlighting the importance of timely diagnosis to improve the possibility of treatment with curative intent.12 Additionally, differences in the diagnostic interval for lung cancer has been demonstrated across ICBP countries, supporting the need for greater alignment in international efforts to improve time to diagnosis and treatment parameters.30 Different countries take a more aggressive stance in their time targets with compulsory reporting, and the accountability for these (e.g. Denmark), whilst in others this can be less regimented.31 The network formed in this study have agreed that <30 days from point of referral for possible lung cancer diagnosis should be a minimum requirement to allow best opportunity for timely initiation of treatment. A crucial consideration within this target is the need to ensure timely access to diagnostic and staging tools, with strong coordination between diagnostic and treatment services – the need to improve this from a policy and practice perspective will differ, depending on jurisdictional context.", "Developing Thoracic Centres of Excellence and formally affiliated networks can help provide advanced and comprehensive patient-focused approaches to lung cancer treatment. Implementation of this aspiration in different regions may differ but essential components of Centres of Excellence for lung cancer have been researched and agreed by this international network. Expertise from multiple disciplines dedicated to lung cancer management (including thoracic surgery, oncology, anaesthesia, respiratory medicine, pathology, radiology, pulmonology rehabilitation) should be available within the Centre and its network. This enables seamless and multi-disciplinary care for patients. Critical to this is ensuring that robustly trained healthcare professionals and specialist experts relevant to these disciplines are deployed, with investment in their training and development to support continued improvement. Broader formal networks are required to provide contemporary access for patients, alongside streamlined processes across prevention, diagnosis, treatment, and survivorship care. It is important that Centres provide comprehensive care to all patients, including those first referred to local hospitals. There is some evidence in the UK that first referral to a local hospital is associated with fewer patients being offered curative treatment.32 The explanation for this is not clear but may relate to distances that patients need to travel.32 Centres should support availability of clinical trials to help deliver innovative research advances to treatments and to enhance patient outcomes. Ultimately, Thoracic Centres of Excellence and their networks should be viewed as the backbone of timely integrated care for lung cancer patients.18 Development of such Centres has been underway across ICBP countries, but relies heavily on available resource, funding, capacity, human resources, and coordination. When considering service improvement and expansion, Centres of Excellence should be in the forefront of policy plans and considerations.", "High-quality data capture and timely feedback is crucial to improving care.33 The importance of this intelligence at local, regional, and national levels needs to be recognised across all countries. Embedding data capture and feedback loops into existing healthcare structures and processes in lung cancer care should be prioritised for self-evaluation and to drive service improvements. Whilst some work has been undertaken to better understand variation in delivery of lung cancer care between comparable high-income countries (as described in the Background), a truly comprehensive understanding of the key drivers of better outcomes in certain countries requires a detailed international audit. In order to deliver this vital intelligence, a number of considerations re required. A universal minimum dataset with key metrics (wait time, stage at diagnosis, risk adjusted resection and radiation rates, medical oncology treatments, significant adverse event rates) is required, alongside strong collaborative relationships between participating countries.34 Ensuring the minimum data metrics are collected as part of routine practice should be the first step to enable a successful audit within jurisdictions, in order to inform the delivery of an international audit.35 Few national audits have been undertaken for lung cancer care, but if more countries replicate this, the roadmap to commissioning an international audit will be clearer.36-38 This will allow clear identification of international best practice to facilitate sharing of optimal care and lessons learnt.", "Continuous improvement in lung cancer care and outcomes should be recognised as an expectation by policymakers, requiring ongoing investment. The success of cancer site-specific policies in high-income countries, for example breast cancer, is reflected in enhanced policy focus, driving service development (screening programmes, diagnostic initiatives and treatment options) and underpinning improved survival internationally.39 However, even in higher performing countries, lung cancer survival is still low.2,4 Significant inequities in lung cancer risk, access to care and treatment outcomes have historically existed, driven largely by socioeconomic inequalities, and thus prohibiting improvements in lung cancer survival seen for other cancer sites.40 Recognising and addressing these inequities and inequalities in care collectively needs to be prioritised within cancer policy internationally, facilitating service improvement to optimise management and care of lung cancer patients. In order to drive this improvement, jurisdictions must consider the effectiveness of their leadership (within both clinical and political arenas) to instigate change, prioritise connection to and collaboration with appropriate stakeholders to ensure lung cancer is promoted on political agendas, and have the means to lobby for and disseminate improved lung cancer policy robustly.41 By calling for lung cancer care to be a priority internationally, we aim to encourage policymakers to prioritise the most pressing initiatives required within their respective countries to have the greatest impact upon lung cancer outcomes.", "It is acknowledged by the network and the authors that these recommendations were developed in the context of the ICBP country membership and may not fully represent the diversity of healthcare settings globally. We are aware the interviews undertaken to help inform the development of the recommendations were of a limited number, however we targeted these at stakeholders who would have a breadth of knowledge. Data saturation would therefore be unlikely to occur but we hope this format and level of questioning can now be replicated and built upon in other healthcare systems and countries to further develop our understanding of key factors in improving lung cancer care internationally. Both the network and authors hope for the recommendations developed to be applicable to other comparable high-income countries, who can interpret these in the context of their healthcare system structure and funding, cultures, socioeconomic situation, and other population-based needs. Additionally, the importance of rigorously evaluating new initiatives, policies, and the evidence base to support their implementation within local settings should not be underestimated – it is the aim of the network and the authors to bring these recommendations to the forefront of policy attention, in order for local assessment of the requirements needed to evaluate and implement." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Introduction", "Materials and Methods", "Network Formation", "Topic Guide Development and Initial Interviews", "Roundtable Discussions and Recommendation Development", "Modified Delphi Method", "Refinement of Recommendations", "Results", "Initial Interview Results", "Recommendations", "Calls to Action Summary", "Box 1A: Calls to Action", "Good Practice Points Summary", "Box 1B: Good Practice Points", "Pre-Diagnosis", "Diagnosis", "Therapeutic", "All Phases of Care", "Discussion", "Implement Cost-Effective, Clinically Efficacious, and Equitable Lung Cancer Screening Initiatives", "Ensure Diagnosis of Lung Cancer Within 30 days of Referral", "Develop Thoracic Centres of Excellence", "Undertake an International Audit of Lung Cancer Care", "Recognise Improvements in Lung Cancer Care and Outcomes as a Priority in Cancer Policy", "Limitations", "Conclusion", "Supplemental Material" ]
[ "Lung cancer accounts for nearly 1.8 million deaths annually – more than colorectal and breast cancers combined.1 Despite significant improvements in diagnosis and treatment, lung cancer is typically associated with low survival internationally.2 The International Cancer Benchmarking Partnership (ICBP) is a global collaboration of clinicians, policymakers, researchers, and cancer data experts. It seeks to benchmark and explain cancer survival, incidence, and mortality differences between high-income countries with comprehensive cancer registry coverage, similar budgetary spending on national health systems and universal access to healthcare. Recent ICBP research has demonstrated that international disparities in lung cancer survival persist, with 5-year survival ranging from 14.7% (UK) to 21.7% (Canada).2 Evidence shows that 3-year survival varies mostly in potentially curative lung cancer, by up to 20 percentage points for localised and regional disease (as per SEER summary staging)3 across the ICBP countries.4 Comparably, survival varied by just 3.4 percentage points in patients diagnosed with distant/metastatic disease across the same countries.4\nDue to the inherent complexity of health systems, it is unlikely that one single component of the cancer care continuum can ultimately define why survival variation exists, yet the ability to diagnose and stage lung cancer patients as early as possible will likely influence their treatment options.5 Various work has been undertaken to articulate patient, healthcare professional and disease related factors that influence timeliness of diagnosis and ultimately, patient outcomes.6 This understanding has helped build initiatives across the pathway to address the poorer outcomes in lung cancer, but to differing levels internationally. This starts with preventative initiatives for lung cancer, including support for smoking cessation and lung cancer awareness campaigns. Both areas are of significance, with tobacco being a key factor in the aetiology of lung cancer, and some awareness campaigns influencing stage shift.7,8 Variation in primary care practitioner readiness to refer patients with potential symptoms and in service provision of key diagnostic and staging tools for lung cancer, such as PET-CT, have been demonstrated across ICBP countries with unknown quantifiable impacts upon survival.9,10 This is reflected in the variation seen in lung cancer stage distribution between ICBP countries, but not to the same extent as differences in survival by stage.4 In the diagnostic phase, it is clear that international differences exist for lung cancer patients, yet the evidence for how this affects outcomes is variable and complex.11 Variation within the diagnosis and staging of early stage lung cancer patients is important; unwarranted delays enabling tumour growth may shift patients away from being in a potentially curable stage towards more advanced stages with lower survival.12,13\nCurative intent treatment options for lung cancer have increased and become significantly more sophisticated over recent years, with the development and adoption of more precise radiotherapy, and minimally invasive surgery.14 Less invasive options have opened up better opportunities for more complex patients or those with greater comorbidities. Guidance and uptake of these treatment options for early stage patients is likely to vary both within countries and internationally. This is particularly the case in patients deemed to be borderline candidates for curative therapy where there are more variables playing a role in decision-making for treatment.15 Variation in the use of curative intent therapies within early stage patients has been demonstrated in some countries.16 Disease stage, patient fitness/performance status, lung function, staffing skillset, socioeconomic status and comorbidities play key influential roles in the perceived operability of patients and subsequent treatment decisions.16\nThe organisation of each country’s healthcare system has been established to guide patients from presentation to the decision for treatment, with many informal and formal treatment pathways being established. However, gaps and biases within each system have been identified that undoubtedly affect the management of potentially curable patients.17 There is a current lack of internationally agreed recommendations for improving care of lung cancer patients, with the purpose of being interpreted at a policy level to align efforts to empower those in positions to instigate change.\nWithin this study, we have established an international lung cancer network to share local clinical insights, including locally defined best practices, mitigation of modifiable variables and opportunities for collaboration. We present a series of recommendations to improve lung cancer care, in order to improve survival, based on the consensus of our international clinical network. We hope that these recommendations serve to optimise lung cancer services so as to improve the quality of options for treatment, equitable access to treatment, and ultimately improve survival internationally.", "[SUBTITLE] Network Formation [SUBSECTION] Nine network members were recruited from nine ICBP jurisdictions: Australia, Canada, Denmark, England, Ireland, New Zealand, Northern Ireland, Scotland and Wales. Members were identified via purposive sampling based on them having roles in national, regional, and local tiers of health systems with expertise primarily being thoracic surgery, radiation oncology, clinical oncology, or respiratory medicine. Existing ICBP clinical networks and stakeholders were approached and asked for suggestions for the network, and desk-based research was conducted to assess suitability based on the criteria outlined in the previous sentence. Members were initially invited to join the network via email, and if interested, they were sent a Terms of Reference and invited to a telephone call to discuss the aims of the project and the ICBP in more detail. At that point, members were asked if they agreed to be part of the network and involved in the project.\nNine network members were recruited from nine ICBP jurisdictions: Australia, Canada, Denmark, England, Ireland, New Zealand, Northern Ireland, Scotland and Wales. Members were identified via purposive sampling based on them having roles in national, regional, and local tiers of health systems with expertise primarily being thoracic surgery, radiation oncology, clinical oncology, or respiratory medicine. Existing ICBP clinical networks and stakeholders were approached and asked for suggestions for the network, and desk-based research was conducted to assess suitability based on the criteria outlined in the previous sentence. Members were initially invited to join the network via email, and if interested, they were sent a Terms of Reference and invited to a telephone call to discuss the aims of the project and the ICBP in more detail. At that point, members were asked if they agreed to be part of the network and involved in the project.\n[SUBTITLE] Topic Guide Development and Initial Interviews [SUBSECTION] We initially employed a qualitative approach to understand potential contributing factors to international variation in lung cancer outcomes that could form the basis of the recommendations developed. The overarching focus of this was mostly on non-small cell lung cancer (NSCLC), and for early stage lung cancer. Semi-structured interviews were conducted with the network members to identify key themes and points of variation between the ICBP countries. Interview questions (Supplementary Materials) were developed by the lead study team (Finley, Lynch, Butler, Harrison), alongside an evidence scoping exercise and review of published literature. Interviews were recorded via Microsoft Teams and stored securely at Cancer Research UK. Qualitative data was transcribed by the lead study team and interview transcripts were analysed thematically, drawing on documents developed during processes undertaken to improve existing national guidelines and support for improving lung cancer care, largely the Pan-Canadian Standards for Thoracic Surgery.18 The analysis of these interviews helped generate the recommendations.\nWe initially employed a qualitative approach to understand potential contributing factors to international variation in lung cancer outcomes that could form the basis of the recommendations developed. The overarching focus of this was mostly on non-small cell lung cancer (NSCLC), and for early stage lung cancer. Semi-structured interviews were conducted with the network members to identify key themes and points of variation between the ICBP countries. Interview questions (Supplementary Materials) were developed by the lead study team (Finley, Lynch, Butler, Harrison), alongside an evidence scoping exercise and review of published literature. Interviews were recorded via Microsoft Teams and stored securely at Cancer Research UK. Qualitative data was transcribed by the lead study team and interview transcripts were analysed thematically, drawing on documents developed during processes undertaken to improve existing national guidelines and support for improving lung cancer care, largely the Pan-Canadian Standards for Thoracic Surgery.18 The analysis of these interviews helped generate the recommendations.\n[SUBTITLE] Roundtable Discussions and Recommendation Development [SUBSECTION] Four roundtable discussions were hosted by the lead study team with all network members. The first and second roundtables discussed the results from the interviews and shared the key themes emerging, which helped structure the discussion to begin generating the recommendations. Network members agreed on key topics for the recommendations to be based on (e.g. pre-diagnosis and screening), and the lead study team articulated these into written recommendations. The third and fourth roundtable meetings were hosted to receive feedback on the Delphi rating method (described below) and further refine the recommendations.\nFour roundtable discussions were hosted by the lead study team with all network members. The first and second roundtables discussed the results from the interviews and shared the key themes emerging, which helped structure the discussion to begin generating the recommendations. Network members agreed on key topics for the recommendations to be based on (e.g. pre-diagnosis and screening), and the lead study team articulated these into written recommendations. The third and fourth roundtable meetings were hosted to receive feedback on the Delphi rating method (described below) and further refine the recommendations.\n[SUBTITLE] Modified Delphi Method [SUBSECTION] Network members were asked to rate recommendations using a modified Delphi consensus model (Figure 1).19 In the first round, interviewees were asked to rate 22 recommendations developed from the interview results and initial suggestions from the first and second roundtable discussions with the network members. Options to ‘agree’, ‘disagree’ and provide additional comments and/or suggestions within a free text box were available. A minimum satisfactory consensus of 70% was reached in the first round, with agreement on all recommendations. Subsequent roundtable discussions were hosted to obtain further feedback and consolidate agreement on any recommendations disagreed with or requiring modification. In the next round, 20 revised recommendations were shared with the same participants to rate agreement with the same format (agree, disagree, additional comments/suggestions). The recommendations were then further refined, with a final round of consensus voting resulting in 18 final recommendations.Figure 1.Stepwise modified Delphi Method.\nStepwise modified Delphi Method.\nNetwork members were asked to rate recommendations using a modified Delphi consensus model (Figure 1).19 In the first round, interviewees were asked to rate 22 recommendations developed from the interview results and initial suggestions from the first and second roundtable discussions with the network members. Options to ‘agree’, ‘disagree’ and provide additional comments and/or suggestions within a free text box were available. A minimum satisfactory consensus of 70% was reached in the first round, with agreement on all recommendations. Subsequent roundtable discussions were hosted to obtain further feedback and consolidate agreement on any recommendations disagreed with or requiring modification. In the next round, 20 revised recommendations were shared with the same participants to rate agreement with the same format (agree, disagree, additional comments/suggestions). The recommendations were then further refined, with a final round of consensus voting resulting in 18 final recommendations.Figure 1.Stepwise modified Delphi Method.\nStepwise modified Delphi Method.\n[SUBTITLE] Refinement of Recommendations [SUBSECTION] ‘Calls to Action’ for each phase of care were identified by the network as the most crucial recommendations via Delphi consensus voting, requiring the most attention by and focus of policymakers, with ‘Good Practice Points’ being required to support the implementation of these Calls to Action. Good Practice points were further categorised broadly into phases of care (pre-diagnosis, diagnosis, therapeutic and all phases) for ease of implementation.\n‘Calls to Action’ for each phase of care were identified by the network as the most crucial recommendations via Delphi consensus voting, requiring the most attention by and focus of policymakers, with ‘Good Practice Points’ being required to support the implementation of these Calls to Action. Good Practice points were further categorised broadly into phases of care (pre-diagnosis, diagnosis, therapeutic and all phases) for ease of implementation.", "Nine network members were recruited from nine ICBP jurisdictions: Australia, Canada, Denmark, England, Ireland, New Zealand, Northern Ireland, Scotland and Wales. Members were identified via purposive sampling based on them having roles in national, regional, and local tiers of health systems with expertise primarily being thoracic surgery, radiation oncology, clinical oncology, or respiratory medicine. Existing ICBP clinical networks and stakeholders were approached and asked for suggestions for the network, and desk-based research was conducted to assess suitability based on the criteria outlined in the previous sentence. Members were initially invited to join the network via email, and if interested, they were sent a Terms of Reference and invited to a telephone call to discuss the aims of the project and the ICBP in more detail. At that point, members were asked if they agreed to be part of the network and involved in the project.", "We initially employed a qualitative approach to understand potential contributing factors to international variation in lung cancer outcomes that could form the basis of the recommendations developed. The overarching focus of this was mostly on non-small cell lung cancer (NSCLC), and for early stage lung cancer. Semi-structured interviews were conducted with the network members to identify key themes and points of variation between the ICBP countries. Interview questions (Supplementary Materials) were developed by the lead study team (Finley, Lynch, Butler, Harrison), alongside an evidence scoping exercise and review of published literature. Interviews were recorded via Microsoft Teams and stored securely at Cancer Research UK. Qualitative data was transcribed by the lead study team and interview transcripts were analysed thematically, drawing on documents developed during processes undertaken to improve existing national guidelines and support for improving lung cancer care, largely the Pan-Canadian Standards for Thoracic Surgery.18 The analysis of these interviews helped generate the recommendations.", "Four roundtable discussions were hosted by the lead study team with all network members. The first and second roundtables discussed the results from the interviews and shared the key themes emerging, which helped structure the discussion to begin generating the recommendations. Network members agreed on key topics for the recommendations to be based on (e.g. pre-diagnosis and screening), and the lead study team articulated these into written recommendations. The third and fourth roundtable meetings were hosted to receive feedback on the Delphi rating method (described below) and further refine the recommendations.", "Network members were asked to rate recommendations using a modified Delphi consensus model (Figure 1).19 In the first round, interviewees were asked to rate 22 recommendations developed from the interview results and initial suggestions from the first and second roundtable discussions with the network members. Options to ‘agree’, ‘disagree’ and provide additional comments and/or suggestions within a free text box were available. A minimum satisfactory consensus of 70% was reached in the first round, with agreement on all recommendations. Subsequent roundtable discussions were hosted to obtain further feedback and consolidate agreement on any recommendations disagreed with or requiring modification. In the next round, 20 revised recommendations were shared with the same participants to rate agreement with the same format (agree, disagree, additional comments/suggestions). The recommendations were then further refined, with a final round of consensus voting resulting in 18 final recommendations.Figure 1.Stepwise modified Delphi Method.\nStepwise modified Delphi Method.", "‘Calls to Action’ for each phase of care were identified by the network as the most crucial recommendations via Delphi consensus voting, requiring the most attention by and focus of policymakers, with ‘Good Practice Points’ being required to support the implementation of these Calls to Action. Good Practice points were further categorised broadly into phases of care (pre-diagnosis, diagnosis, therapeutic and all phases) for ease of implementation.", "[SUBTITLE] Initial Interview Results [SUBSECTION] These are reported in the Supplementary Materials. Key themes in the differences reported between countries include: variation in data capture and auditing, composition and use of multidisciplinary teams, access to mitigating programmes for vulnerable populations and access to clinical research.\nThe remainder of Results section focuses on the recommendations developed during this study as these are our core output.\nThese are reported in the Supplementary Materials. Key themes in the differences reported between countries include: variation in data capture and auditing, composition and use of multidisciplinary teams, access to mitigating programmes for vulnerable populations and access to clinical research.\nThe remainder of Results section focuses on the recommendations developed during this study as these are our core output.\n[SUBTITLE] Recommendations [SUBSECTION] A total of 18 recommendations achieved 100% consensus. These were further grouped in to five Calls to Action (Box 1A) and thirteen Good Practice Points (Box 1B).\nA total of 18 recommendations achieved 100% consensus. These were further grouped in to five Calls to Action (Box 1A) and thirteen Good Practice Points (Box 1B).\n[SUBTITLE] Calls to Action Summary [SUBSECTION] Calls to Action should be interpreted as key priorities to inform policy not only across the ICBP countries, but in similar high-income countries globally. It is recognised that different countries will have differing resource, capacity, funding, and population-based needs and should action these recommendations accordingly to their local settings.Box 1A: Calls to ActionImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\n[SUBTITLE] Box 1A: Calls to Action [SUBSECTION] Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives\nEnsure diagnosis of lung cancer within 30 days of referral\nDevelop Thoracic Centres of Excellence\nUndertake an international audit of lung cancer care\nRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives\nEnsure diagnosis of lung cancer within 30 days of referral\nDevelop Thoracic Centres of Excellence\nUndertake an international audit of lung cancer care\nRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nCalls to Action should be interpreted as key priorities to inform policy not only across the ICBP countries, but in similar high-income countries globally. It is recognised that different countries will have differing resource, capacity, funding, and population-based needs and should action these recommendations accordingly to their local settings.Box 1A: Calls to ActionImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\n[SUBTITLE] Box 1A: Calls to Action [SUBSECTION] Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives\nEnsure diagnosis of lung cancer within 30 days of referral\nDevelop Thoracic Centres of Excellence\nUndertake an international audit of lung cancer care\nRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives\nEnsure diagnosis of lung cancer within 30 days of referral\nDevelop Thoracic Centres of Excellence\nUndertake an international audit of lung cancer care\nRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\n[SUBTITLE] Good Practice Points Summary [SUBSECTION] The network agreed these points are important considerations for similar high-income countries in optimising management and treatment of lung cancer patients. They should be considered as key in supporting the development and implementation of the Calls to Action.Box 1B: Good Practice PointsPre-Diagnosis• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.Diagnosis• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.Therapeutic• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.All Phases of Care• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n[SUBTITLE] Box 1B: Good Practice Points [SUBSECTION] [SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n[SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n[SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n[SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n[SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n[SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n[SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n[SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\nThe network agreed these points are important considerations for similar high-income countries in optimising management and treatment of lung cancer patients. They should be considered as key in supporting the development and implementation of the Calls to Action.Box 1B: Good Practice PointsPre-Diagnosis• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.Diagnosis• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.Therapeutic• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.All Phases of Care• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n[SUBTITLE] Box 1B: Good Practice Points [SUBSECTION] [SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n[SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n[SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n[SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n[SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n[SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n[SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n[SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.", "These are reported in the Supplementary Materials. Key themes in the differences reported between countries include: variation in data capture and auditing, composition and use of multidisciplinary teams, access to mitigating programmes for vulnerable populations and access to clinical research.\nThe remainder of Results section focuses on the recommendations developed during this study as these are our core output.", "A total of 18 recommendations achieved 100% consensus. These were further grouped in to five Calls to Action (Box 1A) and thirteen Good Practice Points (Box 1B).", "Calls to Action should be interpreted as key priorities to inform policy not only across the ICBP countries, but in similar high-income countries globally. It is recognised that different countries will have differing resource, capacity, funding, and population-based needs and should action these recommendations accordingly to their local settings.Box 1A: Calls to ActionImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\n[SUBTITLE] Box 1A: Calls to Action [SUBSECTION] Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives\nEnsure diagnosis of lung cancer within 30 days of referral\nDevelop Thoracic Centres of Excellence\nUndertake an international audit of lung cancer care\nRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives\nEnsure diagnosis of lung cancer within 30 days of referral\nDevelop Thoracic Centres of Excellence\nUndertake an international audit of lung cancer care\nRecognise improvements in lung cancer care and outcomes as a priority in cancer policy", "Implement cost-effective, clinically efficacious, and equitable lung cancer screening initiativesEnsure diagnosis of lung cancer within 30 days of referralDevelop Thoracic Centres of ExcellenceUndertake an international audit of lung cancer careRecognise improvements in lung cancer care and outcomes as a priority in cancer policy\nImplement cost-effective, clinically efficacious, and equitable lung cancer screening initiatives\nEnsure diagnosis of lung cancer within 30 days of referral\nDevelop Thoracic Centres of Excellence\nUndertake an international audit of lung cancer care\nRecognise improvements in lung cancer care and outcomes as a priority in cancer policy", "The network agreed these points are important considerations for similar high-income countries in optimising management and treatment of lung cancer patients. They should be considered as key in supporting the development and implementation of the Calls to Action.Box 1B: Good Practice PointsPre-Diagnosis• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.Diagnosis• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.Therapeutic• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.All Phases of Care• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n[SUBTITLE] Box 1B: Good Practice Points [SUBSECTION] [SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n[SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n[SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n[SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n[SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n[SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n[SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n[SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.", "[SUBTITLE] Pre-Diagnosis [SUBSECTION] • Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n[SUBTITLE] Diagnosis [SUBSECTION] • Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n[SUBTITLE] Therapeutic [SUBSECTION] • Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n[SUBTITLE] All Phases of Care [SUBSECTION] • Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.", "• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.\n• Timely and widespread availability for primary care referrals to cross sectional imaging should be prioritised in patients with a suspicion of lung cancer.\n• National coordinated education materials and support should be available to public and health care providers on lung cancer and its management and outcomes.", "• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.\n• Where appropriate, lung malignancies should be discussed in a multidisciplinary format with essential components in place:• Protocols for incorporation into workplans, coordination and chairing of meetings• Appropriate attendance from core stakeholders• Means to record and report treatment decisions locally and centrally• Method to communicate treatment decisions to referring practitioners\n• Protocols for incorporation into workplans, coordination and chairing of meetings\n• Appropriate attendance from core stakeholders\n• Means to record and report treatment decisions locally and centrally\n• Method to communicate treatment decisions to referring practitioners\n• Molecular profiling of tumours should be prioritised where there is a clinical need, with pathology reports presented in a synoptic format and communicated within 2 weeks of biopsy or operation.", "• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.\n• Incorporation of pre-habilitation programmes into routine care for patients undergoing surgical resection should be considered and implemented where appropriate.\n• Curative intent therapy should be prioritised where possible, even in higher risk patients where the challenges in management of significant comorbidities and increased risk of adverse events is recognised.\n• Integrated care teams should be in place and supported by surgeons with sufficient topical expertise to identify preventable adverse events in patients.\n• It is the expectation that when adopting new technologies and techniques active tracking of adverse events and outcomes will be completed.", "• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.\n• Each country should have in place a minimum dataset for the evaluation of lung cancer patients’ diagnosis, treatment, and aftercare. Minimum datasets should have at least the following critical data elements: wait times, stage at diagnosis, risk adjusted resection and radiation rates, and significant adverse event rates.\n• Commissioning of regular national clinical audits should be employed in all countries, with necessary analytical capacity for timely analysis and interrogation of data at local levels\n• Emphasis on quality improvement should be placed in every part of the lung cancer pathway with regular review of audit data and processes in place to facilitate sharing and learning of best practice.\n• Identification of patients at high-risk for negative outcomes should be part of routine care, with the development of appropriate pathways to mitigate poorer outcomes.\n• System and investment planning for future advanced equipment should be undertaken to reflect emerging/new and innovative diagnostic methods and therapeutics.", "Rapid progress in areas of healthcare need can be achieved when international collaborative efforts, research and funding are aligned to a common goal, most recently illustrated by the development of the COVID vaccine.20 The international cancer community recognises the need to significantly improve lung cancer survival.21 The Calls to Action and Good Practice Points generated in this study provide a baseline for reflection, appropriate benchmarking, and identification of opportunities for enhancing lung cancer care. These can be interpreted from local, regional, national, and international perspectives. Many advances have been made in lung cancer prevention, screening, diagnosis, and treatment worldwide – from lower smoking rates, utilisation of CT scans for earlier detection, greater access to innovative medicines, improved operative techniques and more advanced radiation therapy. However, the potential improvements in survival have not yet been achieved. The international network formed, and recommendations developed within this study, are advocating for increased and better alignment of efforts within, and across, countries in order to improve lung cancer care and survival. We outline the importance and considerations to be had when addressing the Calls to Action below.", "Due to variation seen in stage distribution between ICBP countries, it is important to consider where opportunities exist to optimise care and improve survival in the pre-treatment interval. Whilst the evidence is strong for a survival benefit of low dose CT screening in individuals with higher risk of lung cancer, implementation has been a complex issue for policymakers.22-24 Whilst low dose CT screening can support detection of early stage disease and increase the likelihood of curable treatment, several factors must be carefully considered to ensure clinical efficacy and cost effectiveness. Integration into existing infrastructure, target populations, recruitment of population at risk, reduction of harm, resource and cost, and engagement with the public and healthcare professionals need to be defined and implemented. Screening programmes need to be designed to reach a traditionally ‘hard to reach’ population, requiring their design to have greater engagement of those affected. There is no “one size fits all” approach, yet it is the collective opinion of this international clinical network that understanding the impact that screening could have for outcomes and considering all these factors within local contexts is a crucial activity for policymakers. Currently, no ICBP country has fully implemented a national lung cancer screening programme, though a number of countries are at different stages of exploration and implementation. Successful models of assessment and pilot trials have been underway internationally for several years and can serve as a baseline to inform the best implementation plan for screening in each jurisdiction.25-29", "Various temporal targets for key time points within the cancer patient journey exist for different countries, but there has previously been a lack of international consensus on a universal ‘best practice’ target. Unwarranted delays in diagnosis can rapidly shift lung cancer patients from being potentially curative to being difficult to treat, highlighting the importance of timely diagnosis to improve the possibility of treatment with curative intent.12 Additionally, differences in the diagnostic interval for lung cancer has been demonstrated across ICBP countries, supporting the need for greater alignment in international efforts to improve time to diagnosis and treatment parameters.30 Different countries take a more aggressive stance in their time targets with compulsory reporting, and the accountability for these (e.g. Denmark), whilst in others this can be less regimented.31 The network formed in this study have agreed that <30 days from point of referral for possible lung cancer diagnosis should be a minimum requirement to allow best opportunity for timely initiation of treatment. A crucial consideration within this target is the need to ensure timely access to diagnostic and staging tools, with strong coordination between diagnostic and treatment services – the need to improve this from a policy and practice perspective will differ, depending on jurisdictional context.", "Developing Thoracic Centres of Excellence and formally affiliated networks can help provide advanced and comprehensive patient-focused approaches to lung cancer treatment. Implementation of this aspiration in different regions may differ but essential components of Centres of Excellence for lung cancer have been researched and agreed by this international network. Expertise from multiple disciplines dedicated to lung cancer management (including thoracic surgery, oncology, anaesthesia, respiratory medicine, pathology, radiology, pulmonology rehabilitation) should be available within the Centre and its network. This enables seamless and multi-disciplinary care for patients. Critical to this is ensuring that robustly trained healthcare professionals and specialist experts relevant to these disciplines are deployed, with investment in their training and development to support continued improvement. Broader formal networks are required to provide contemporary access for patients, alongside streamlined processes across prevention, diagnosis, treatment, and survivorship care. It is important that Centres provide comprehensive care to all patients, including those first referred to local hospitals. There is some evidence in the UK that first referral to a local hospital is associated with fewer patients being offered curative treatment.32 The explanation for this is not clear but may relate to distances that patients need to travel.32 Centres should support availability of clinical trials to help deliver innovative research advances to treatments and to enhance patient outcomes. Ultimately, Thoracic Centres of Excellence and their networks should be viewed as the backbone of timely integrated care for lung cancer patients.18 Development of such Centres has been underway across ICBP countries, but relies heavily on available resource, funding, capacity, human resources, and coordination. When considering service improvement and expansion, Centres of Excellence should be in the forefront of policy plans and considerations.", "High-quality data capture and timely feedback is crucial to improving care.33 The importance of this intelligence at local, regional, and national levels needs to be recognised across all countries. Embedding data capture and feedback loops into existing healthcare structures and processes in lung cancer care should be prioritised for self-evaluation and to drive service improvements. Whilst some work has been undertaken to better understand variation in delivery of lung cancer care between comparable high-income countries (as described in the Background), a truly comprehensive understanding of the key drivers of better outcomes in certain countries requires a detailed international audit. In order to deliver this vital intelligence, a number of considerations re required. A universal minimum dataset with key metrics (wait time, stage at diagnosis, risk adjusted resection and radiation rates, medical oncology treatments, significant adverse event rates) is required, alongside strong collaborative relationships between participating countries.34 Ensuring the minimum data metrics are collected as part of routine practice should be the first step to enable a successful audit within jurisdictions, in order to inform the delivery of an international audit.35 Few national audits have been undertaken for lung cancer care, but if more countries replicate this, the roadmap to commissioning an international audit will be clearer.36-38 This will allow clear identification of international best practice to facilitate sharing of optimal care and lessons learnt.", "Continuous improvement in lung cancer care and outcomes should be recognised as an expectation by policymakers, requiring ongoing investment. The success of cancer site-specific policies in high-income countries, for example breast cancer, is reflected in enhanced policy focus, driving service development (screening programmes, diagnostic initiatives and treatment options) and underpinning improved survival internationally.39 However, even in higher performing countries, lung cancer survival is still low.2,4 Significant inequities in lung cancer risk, access to care and treatment outcomes have historically existed, driven largely by socioeconomic inequalities, and thus prohibiting improvements in lung cancer survival seen for other cancer sites.40 Recognising and addressing these inequities and inequalities in care collectively needs to be prioritised within cancer policy internationally, facilitating service improvement to optimise management and care of lung cancer patients. In order to drive this improvement, jurisdictions must consider the effectiveness of their leadership (within both clinical and political arenas) to instigate change, prioritise connection to and collaboration with appropriate stakeholders to ensure lung cancer is promoted on political agendas, and have the means to lobby for and disseminate improved lung cancer policy robustly.41 By calling for lung cancer care to be a priority internationally, we aim to encourage policymakers to prioritise the most pressing initiatives required within their respective countries to have the greatest impact upon lung cancer outcomes.", "It is acknowledged by the network and the authors that these recommendations were developed in the context of the ICBP country membership and may not fully represent the diversity of healthcare settings globally. We are aware the interviews undertaken to help inform the development of the recommendations were of a limited number, however we targeted these at stakeholders who would have a breadth of knowledge. Data saturation would therefore be unlikely to occur but we hope this format and level of questioning can now be replicated and built upon in other healthcare systems and countries to further develop our understanding of key factors in improving lung cancer care internationally. Both the network and authors hope for the recommendations developed to be applicable to other comparable high-income countries, who can interpret these in the context of their healthcare system structure and funding, cultures, socioeconomic situation, and other population-based needs. Additionally, the importance of rigorously evaluating new initiatives, policies, and the evidence base to support their implementation within local settings should not be underestimated – it is the aim of the network and the authors to bring these recommendations to the forefront of policy attention, in order for local assessment of the requirements needed to evaluate and implement.", "The burden of lung cancer is significant and represents a global public health challenge. The Calls to Action and Good Practice Points presented here, reflecting the views of an international expert lung cancer clinical network, signpost the key considerations for policymakers within high-income comparable countries to effective change. They provide a roadmap to help align and focus efforts in improving outcomes and management of lung cancer patients. The Good Practice Points described are key to enable delivery of the Calls to Action, which should be considered the highest priority. Collaboration at local, regional, national, and international levels within the lung cancer community is imperative to empower policymakers to ensure policies are enacted that enhance lung cancer patient treatment and improve quality of life and patient outcomes.", "Click here for additional data file.\nSupplemental material for An International Consensus on Actions to Improve Lung Cancer Survival: A Modified Delphi Method Among Clinical Experts in the International Cancer Benchmarking Partnership by Charlotte Lynch, Samantha Harrison, John Butler, David R. Baldwin, Paul Dawkins, Joris van der Horst, Erik Jakobsen, Jonathan McAleese, Annette McWilliams, Karen Redmond, Anand Swaminath, and Christian J. Finley in Cancer Control" ]
[ "intro", "materials|methods", null, null, null, null, null, "results", null, null, null, null, null, null, null, null, null, null, "discussion", null, null, null, null, null, null, "conclusion", "supplementary-material" ]
[ "lung cancer", "health policy", "public health", "epidemiology", "survival" ]
Disinfection of Virtual Reality Devices in Health Care Settings: In Vitro Assessment and Survey Study.
36269222
Virtual reality (VR) devices are increasingly used in health care settings. The use among patients has the potential to unintentionally transmit pathogens between patients and hospital staff. No standard operating procedure for disinfection exists to ensure safe use between patients.
BACKGROUND
Three types of bacteria were inoculated onto porous and nonporous surfaces of 2 VR devices: the Meta Oculus Quest and Meta Oculus Quest 2. Disinfection was performed using either isopropyl alcohol or alcohol-free quaternary ammonium wipes. A quantitative culture was used to assess the adequacy of disinfection. A survey was separately sent out to VR device technicians at other pediatric health care institutes to compare the methods of disinfection and how they were established.
METHODS
Both products achieved adequate disinfection of the treated surfaces; however, a greater log-kill was achieved on nonporous surfaces than on the porous surfaces. Alcohol performed better than quaternary ammonium on porous surfaces. The survey respondents reported a wide variability in disinfection processes with only 1 person reporting an established standard operating procedure.
RESULTS
Disinfection can be achieved through the use of either isopropyl alcohol or quaternary ammonium products. Porous surfaces showed lesser log-kill rates than the nonporous surfaces, indicating that the use of an added barrier may be of benefit and should be a point of future research. Given the variability in the disinfection process across health care systems, a standard operating procedure is proposed.
CONCLUSIONS
[ "Child", "Humans", "Disinfection", "2-Propanol", "Ethanol", "Ammonium Compounds", "Surveys and Questionnaires", "Delivery of Health Care", "Virtual Reality" ]
9756115
Introduction
Virtual reality (VR) devices are increasingly used in health care settings to benefit patients, and the examples include patients with posttraumatic stress disorder, anxiety, complex regional pain syndrome, and distraction therapy [1-4]. Recent data show the benefit expands to the pediatric population as well by reducing pain and anxiety during medical procedures through distraction [5]. VR can also be used to educate health care workers through training and simulation [6]. However, a lack of standardized cleaning and disinfection processes for VR devices has limited VR’s use in health care settings, especially during the COVID-19 pandemic [6]. Nosocomial transmission and outbreaks have been reported with many different types of medical devices in clinical use [7], and establishing standard operating procedures (SOPs) for disinfection of VR devices between patient use is paramount. One of the most common and widely sold VR headset devices worldwide, the Meta Oculus Quest 2, specifically, recommends against the use of alcohol to clean and disinfect the device in favor of antibacterial wipes due to theoretical concerns about affecting the porous material [8]. This poses challenges in clinical settings as isopropyl alcohol (IPA) is one of the most common disinfectants used on medical devices. A protocol to clean and disinfect VR devices used in health care settings has been proposed [6]; however, no studies have quantified the efficacy of hospital-grade disinfectants on different parts of the VR equipment. Additionally, little is known about how these machines are currently disinfected in health care settings. In this mixed methods evaluation, we sought to determine the current disinfection practices in health care settings and how they were established. We also studied the effect of commonly used disinfectant wipes on the disinfection of VR headsets experimentally contaminated with common bacterial pathogens to provide evidence for the creation of an SOP to reduce infections with multipatient VR utilization.
Methods
[SUBTITLE] Survey [SUBSECTION] To learn how health care facilities disinfect VR equipment and whether infection prevention teams are involved, a voluntary Qualtrics survey was sent via an electronic link in a group chat of 50 VR technicians working at children’s hospitals across the United States as a convenience sample. To learn how health care facilities disinfect VR equipment and whether infection prevention teams are involved, a voluntary Qualtrics survey was sent via an electronic link in a group chat of 50 VR technicians working at children’s hospitals across the United States as a convenience sample. [SUBTITLE] Ethical Considerations [SUBSECTION] The survey was approved as exempt by the Yale University institutional review board (study #2000033075). The survey was approved as exempt by the Yale University institutional review board (study #2000033075). [SUBTITLE] Laboratory Disinfection [SUBSECTION] Three types of bacteria, Staphylococcus epidermidis (ATCC 12228), Pseudomonas aeruginosa (laboratory strain PAO1), and Staphylococcus aureus (ATCC 25923), were chosen because of their propensity to be present on the skin and cause infection in children with compromised immune systems. The bacteria were grown overnight in 3 mL of lysogeny broth and serially diluted to quantitate the bacterial inoculum. VR headsets and controllers were inoculated by spreading 10 μL (initial inoculum 4.1×106-4.5×108) onto various sites (Figure 1) and allowed to dry for 30 minutes. This large inoculum was chosen to test whether Environmental Protection Agency (EPA)–approved disinfectants achieved sufficient log-reduction in bacteria as per their instructions for use. Two VR devices were experimentally contaminated: the Oculus Quest headset and the Oculus Quest 2 headset and hand controllers (Reality Labs, Meta Platforms). These devices were chosen as these are the most popular consumer devices, the primary ones used at our institution, and contained different surface types to trial disinfection [9]. Contaminated sites included the outer surface of the headset housing on the top side, the controller buttons, and the headband straps for each device (Figure 1). These sites were chosen as they were thought to be high–touch point areas for the hands and head during patient use. We did not study the facial interface as it is our standard practice to use a disposable barrier between the facial interface and the patient’s skin. Both nonporous (Oculus Quest 1 strap, Oculus Quest 2 headset, and controller) and porous (Oculus Quest 1 headset and Oculus Quest 2 strap) surfaces were contaminated to assess disinfection efficacy. Two products were tested for active disinfection: a 70% IPA wipe (Medium Alcohol Prep Pad, Medline) and an alcohol-free quaternary ammonium wipe (Sani-Cloth AF3 Germicidal Disposable Wipe, PDI). A positive control of inoculation without disinfection was performed for every experiment and cultured after the dry time to account for bacterial cell death from desiccation. Disinfection was performed in accordance with each product’s manufacturer’s instructions for use. For the IPA wipe, a 15-second scrub in a back-and-forth motion followed by a 15-second dry time was performed. For the alcohol-free quaternary ammonium wipe, the experimentally contaminated area was wiped to a point of saturation for 3 minutes and allowed to dry. Following disinfection, the cultures were obtained with sterile cotton swabs dipped in Dey-Engley (D/E) neutralizing broth (Hardy Diagnostics) and wiped across the entirety of the contaminated surface for 5 seconds in a back-and-forth motion. The swab was used to inoculate a D/E agar plate that was then incubated overnight at 37 C. Bacterial colony forming units (CFUs) were counted on the plates the following day. Between experiments, the entirety of the VR devices was disinfected with a 70% IPA spray to the point of saturation of the surface materials and dried overnight. The laboratory experiments satisfied the Yale University 100 CH.9 Clinical Quality Improvement criteria and were exempt from institutional review board approval. Areas where cultures were obtained (shown by red circles) of Oculus Quest (top) and Oculus Quest 2 (bottom) devices. Three types of bacteria, Staphylococcus epidermidis (ATCC 12228), Pseudomonas aeruginosa (laboratory strain PAO1), and Staphylococcus aureus (ATCC 25923), were chosen because of their propensity to be present on the skin and cause infection in children with compromised immune systems. The bacteria were grown overnight in 3 mL of lysogeny broth and serially diluted to quantitate the bacterial inoculum. VR headsets and controllers were inoculated by spreading 10 μL (initial inoculum 4.1×106-4.5×108) onto various sites (Figure 1) and allowed to dry for 30 minutes. This large inoculum was chosen to test whether Environmental Protection Agency (EPA)–approved disinfectants achieved sufficient log-reduction in bacteria as per their instructions for use. Two VR devices were experimentally contaminated: the Oculus Quest headset and the Oculus Quest 2 headset and hand controllers (Reality Labs, Meta Platforms). These devices were chosen as these are the most popular consumer devices, the primary ones used at our institution, and contained different surface types to trial disinfection [9]. Contaminated sites included the outer surface of the headset housing on the top side, the controller buttons, and the headband straps for each device (Figure 1). These sites were chosen as they were thought to be high–touch point areas for the hands and head during patient use. We did not study the facial interface as it is our standard practice to use a disposable barrier between the facial interface and the patient’s skin. Both nonporous (Oculus Quest 1 strap, Oculus Quest 2 headset, and controller) and porous (Oculus Quest 1 headset and Oculus Quest 2 strap) surfaces were contaminated to assess disinfection efficacy. Two products were tested for active disinfection: a 70% IPA wipe (Medium Alcohol Prep Pad, Medline) and an alcohol-free quaternary ammonium wipe (Sani-Cloth AF3 Germicidal Disposable Wipe, PDI). A positive control of inoculation without disinfection was performed for every experiment and cultured after the dry time to account for bacterial cell death from desiccation. Disinfection was performed in accordance with each product’s manufacturer’s instructions for use. For the IPA wipe, a 15-second scrub in a back-and-forth motion followed by a 15-second dry time was performed. For the alcohol-free quaternary ammonium wipe, the experimentally contaminated area was wiped to a point of saturation for 3 minutes and allowed to dry. Following disinfection, the cultures were obtained with sterile cotton swabs dipped in Dey-Engley (D/E) neutralizing broth (Hardy Diagnostics) and wiped across the entirety of the contaminated surface for 5 seconds in a back-and-forth motion. The swab was used to inoculate a D/E agar plate that was then incubated overnight at 37 C. Bacterial colony forming units (CFUs) were counted on the plates the following day. Between experiments, the entirety of the VR devices was disinfected with a 70% IPA spray to the point of saturation of the surface materials and dried overnight. The laboratory experiments satisfied the Yale University 100 CH.9 Clinical Quality Improvement criteria and were exempt from institutional review board approval. Areas where cultures were obtained (shown by red circles) of Oculus Quest (top) and Oculus Quest 2 (bottom) devices. [SUBTITLE] Statistical Analysis [SUBSECTION] The experimental design was a 3-factor crossed design with bacterial CFUs as the outcome variable. The generalized linear model with binomial distribution and logit link was used to compare whether the proportion of trials with observable bacteria counts after disinfection differed by (1) type of disinfection, (2) type of organism, and (3) surface type. All models included the natural log of bacterial count prior to disinfection and used robust standard errors. Achieving disinfection was defined as a greater than a 6-log reduction in bacterial counts. Pairwise comparisons were performed for the type of organism. Analyses were performed in SPSS (version 27; IBM Corp), and statistical significance was set at an α level of .05. The experimental design was a 3-factor crossed design with bacterial CFUs as the outcome variable. The generalized linear model with binomial distribution and logit link was used to compare whether the proportion of trials with observable bacteria counts after disinfection differed by (1) type of disinfection, (2) type of organism, and (3) surface type. All models included the natural log of bacterial count prior to disinfection and used robust standard errors. Achieving disinfection was defined as a greater than a 6-log reduction in bacterial counts. Pairwise comparisons were performed for the type of organism. Analyses were performed in SPSS (version 27; IBM Corp), and statistical significance was set at an α level of .05.
Results
[SUBTITLE] Current VR Disinfection Practices in Pediatric Hospitals [SUBSECTION] A total of 50 VR technicians across the Unites States were invited to participate in the Qualtrics survey with a response rate of 18% (9/50). One person consented and then did not answer any of the questions. The selected results of the survey are shown in Table 1 and highlight the variability of VR use and disinfection practices. Survey of current VRa disinfection practices in health care settings. aVR: virtual reality. bSome respondents gave multiple responses. The number of VR sessions varied from less than once a week to multiple times per day. Most locations used multiple types of VR platforms with the Starlight Children’s VR system most commonly used. The Starlight Children’s VR system is a variation of the Lenovo Mirage Solo VR headset and is made of very similar materials as the Oculus Quest 1 and Oculus Quest 2. All participants noted that disinfection was performed before patient use (n=9, 100%). The methods of disinfection were variable with IPA and quaternary ammonium low-level disinfection wipes used most commonly. Most VR technicians used physical barriers between the VR device and the patient such as combinations of silicon covers, disposable eye masks, wipeable replacement head straps, and hair covers, while 3 (33%) participants did not use barriers (Table 1). Only 1 (11%) institution had an SOP for use and disinfection. Infection prevention teams were involved in assisting with VR disinfection protocols at 2 (22%) sites. A total of 50 VR technicians across the Unites States were invited to participate in the Qualtrics survey with a response rate of 18% (9/50). One person consented and then did not answer any of the questions. The selected results of the survey are shown in Table 1 and highlight the variability of VR use and disinfection practices. Survey of current VRa disinfection practices in health care settings. aVR: virtual reality. bSome respondents gave multiple responses. The number of VR sessions varied from less than once a week to multiple times per day. Most locations used multiple types of VR platforms with the Starlight Children’s VR system most commonly used. The Starlight Children’s VR system is a variation of the Lenovo Mirage Solo VR headset and is made of very similar materials as the Oculus Quest 1 and Oculus Quest 2. All participants noted that disinfection was performed before patient use (n=9, 100%). The methods of disinfection were variable with IPA and quaternary ammonium low-level disinfection wipes used most commonly. Most VR technicians used physical barriers between the VR device and the patient such as combinations of silicon covers, disposable eye masks, wipeable replacement head straps, and hair covers, while 3 (33%) participants did not use barriers (Table 1). Only 1 (11%) institution had an SOP for use and disinfection. Infection prevention teams were involved in assisting with VR disinfection protocols at 2 (22%) sites. [SUBTITLE] Effectiveness of Hospital-Grade Disinfecting Wipes on VR Decontamination [SUBSECTION] A total of 175 experiments were performed to assess disinfection (Table 2). Adequate disinfection was achieved with both the IPA and the alcohol-free quaternary ammonium wipes across all bacterial types and headset material comparing untreated with disinfected surfaces. No bacteria were recovered in 88% (154/175) of experiments. IPA wipes performed better than the quaternary ammonium wipes at reducing overall bacterial counts (P=.001). This difference was most pronounced on porous surfaces, where the mean quantity of bacteria remaining after the alcohol-free quaternary ammonium use was more than after IPA use (Figure 2A). When comparing the disinfection by an organism, there were more CFUs of S aureus and S epidermidis recovered after attempted disinfection than P aeruginosa (P=.05 and P=.03, respectively), with this difference most pronounced on porous surfaces (Figure 2B). There was no significant difference between the recovery of S aureus and S epidermidis (P=.72). Finally, fewer bacteria were recovered from porous surfaces after inoculation but prior to performing disinfection. However, more bacteria were recovered after disinfection from porous surfaces than from nonporous surfaces (P=.01) as shown in Figure 2C, confirming that porous materials inoculated (Figure 1) were more difficult to disinfect because of their availability to absorb the bacteria-containing liquid. Bacterial count by disinfection method, organism, and surface. aThe percentage of times when 0 bacterial colony-forming units were observed. bNonporous surfaces were the Quest 1 Strap, Quest 2 Headset, and controller, while the porous surfaces were the Quest 1 Headset and Quest 2 Strap. The mean (SEM) raw bacterial counts recovered after disinfection according to disinfectant and equipment (A), by organism and equipment (B), and by equipment surface type (C) are displayed. Note: Non-porous surfaces were the Quest 1 Strap, Quest 2 Headset and Controller, while porous surfaces were the Quest 1 Headset and Quest 2 Strap. A total of 175 experiments were performed to assess disinfection (Table 2). Adequate disinfection was achieved with both the IPA and the alcohol-free quaternary ammonium wipes across all bacterial types and headset material comparing untreated with disinfected surfaces. No bacteria were recovered in 88% (154/175) of experiments. IPA wipes performed better than the quaternary ammonium wipes at reducing overall bacterial counts (P=.001). This difference was most pronounced on porous surfaces, where the mean quantity of bacteria remaining after the alcohol-free quaternary ammonium use was more than after IPA use (Figure 2A). When comparing the disinfection by an organism, there were more CFUs of S aureus and S epidermidis recovered after attempted disinfection than P aeruginosa (P=.05 and P=.03, respectively), with this difference most pronounced on porous surfaces (Figure 2B). There was no significant difference between the recovery of S aureus and S epidermidis (P=.72). Finally, fewer bacteria were recovered from porous surfaces after inoculation but prior to performing disinfection. However, more bacteria were recovered after disinfection from porous surfaces than from nonporous surfaces (P=.01) as shown in Figure 2C, confirming that porous materials inoculated (Figure 1) were more difficult to disinfect because of their availability to absorb the bacteria-containing liquid. Bacterial count by disinfection method, organism, and surface. aThe percentage of times when 0 bacterial colony-forming units were observed. bNonporous surfaces were the Quest 1 Strap, Quest 2 Headset, and controller, while the porous surfaces were the Quest 1 Headset and Quest 2 Strap. The mean (SEM) raw bacterial counts recovered after disinfection according to disinfectant and equipment (A), by organism and equipment (B), and by equipment surface type (C) are displayed. Note: Non-porous surfaces were the Quest 1 Strap, Quest 2 Headset and Controller, while porous surfaces were the Quest 1 Headset and Quest 2 Strap.
null
null
[ "Survey", "Ethical Considerations", "Laboratory Disinfection", "Statistical Analysis", "Current VR Disinfection Practices in Pediatric Hospitals", "Effectiveness of Hospital-Grade Disinfecting Wipes on VR Decontamination" ]
[ "To learn how health care facilities disinfect VR equipment and whether infection prevention teams are involved, a voluntary Qualtrics survey was sent via an electronic link in a group chat of 50 VR technicians working at children’s hospitals across the United States as a convenience sample.", "The survey was approved as exempt by the Yale University institutional review board (study #2000033075).", "Three types of bacteria, Staphylococcus epidermidis (ATCC 12228), Pseudomonas aeruginosa (laboratory strain PAO1), and Staphylococcus aureus (ATCC 25923), were chosen because of their propensity to be present on the skin and cause infection in children with compromised immune systems. The bacteria were grown overnight in 3 mL of lysogeny broth and serially diluted to quantitate the bacterial inoculum. VR headsets and controllers were inoculated by spreading 10 μL (initial inoculum 4.1×106-4.5×108) onto various sites (Figure 1) and allowed to dry for 30 minutes. This large inoculum was chosen to test whether Environmental Protection Agency (EPA)–approved disinfectants achieved sufficient log-reduction in bacteria as per their instructions for use. Two VR devices were experimentally contaminated: the Oculus Quest headset and the Oculus Quest 2 headset and hand controllers (Reality Labs, Meta Platforms). These devices were chosen as these are the most popular consumer devices, the primary ones used at our institution, and contained different surface types to trial disinfection [9]. Contaminated sites included the outer surface of the headset housing on the top side, the controller buttons, and the headband straps for each device (Figure 1). These sites were chosen as they were thought to be high–touch point areas for the hands and head during patient use. We did not study the facial interface as it is our standard practice to use a disposable barrier between the facial interface and the patient’s skin. Both nonporous (Oculus Quest 1 strap, Oculus Quest 2 headset, and controller) and porous (Oculus Quest 1 headset and Oculus Quest 2 strap) surfaces were contaminated to assess disinfection efficacy.\nTwo products were tested for active disinfection: a 70% IPA wipe (Medium Alcohol Prep Pad, Medline) and an alcohol-free quaternary ammonium wipe (Sani-Cloth AF3 Germicidal Disposable Wipe, PDI). A positive control of inoculation without disinfection was performed for every experiment and cultured after the dry time to account for bacterial cell death from desiccation. Disinfection was performed in accordance with each product’s manufacturer’s instructions for use. For the IPA wipe, a 15-second scrub in a back-and-forth motion followed by a 15-second dry time was performed. For the alcohol-free quaternary ammonium wipe, the experimentally contaminated area was wiped to a point of saturation for 3 minutes and allowed to dry. Following disinfection, the cultures were obtained with sterile cotton swabs dipped in Dey-Engley (D/E) neutralizing broth (Hardy Diagnostics) and wiped across the entirety of the contaminated surface for 5 seconds in a back-and-forth motion. The swab was used to inoculate a D/E agar plate that was then incubated overnight at 37 C. Bacterial colony forming units (CFUs) were counted on the plates the following day. Between experiments, the entirety of the VR devices was disinfected with a 70% IPA spray to the point of saturation of the surface materials and dried overnight. The laboratory experiments satisfied the Yale University 100 CH.9 Clinical Quality Improvement criteria and were exempt from institutional review board approval.\nAreas where cultures were obtained (shown by red circles) of Oculus Quest (top) and Oculus Quest 2 (bottom) devices.", "The experimental design was a 3-factor crossed design with bacterial CFUs as the outcome variable. The generalized linear model with binomial distribution and logit link was used to compare whether the proportion of trials with observable bacteria counts after disinfection differed by (1) type of disinfection, (2) type of organism, and (3) surface type. All models included the natural log of bacterial count prior to disinfection and used robust standard errors. Achieving disinfection was defined as a greater than a 6-log reduction in bacterial counts. Pairwise comparisons were performed for the type of organism. Analyses were performed in SPSS (version 27; IBM Corp), and statistical significance was set at an α level of .05.", "A total of 50 VR technicians across the Unites States were invited to participate in the Qualtrics survey with a response rate of 18% (9/50). One person consented and then did not answer any of the questions. The selected results of the survey are shown in Table 1 and highlight the variability of VR use and disinfection practices.\nSurvey of current VRa disinfection practices in health care settings.\naVR: virtual reality.\nbSome respondents gave multiple responses.\nThe number of VR sessions varied from less than once a week to multiple times per day. Most locations used multiple types of VR platforms with the Starlight Children’s VR system most commonly used. The Starlight Children’s VR system is a variation of the Lenovo Mirage Solo VR headset and is made of very similar materials as the Oculus Quest 1 and Oculus Quest 2. All participants noted that disinfection was performed before patient use (n=9, 100%). The methods of disinfection were variable with IPA and quaternary ammonium low-level disinfection wipes used most commonly. Most VR technicians used physical barriers between the VR device and the patient such as combinations of silicon covers, disposable eye masks, wipeable replacement head straps, and hair covers, while 3 (33%) participants did not use barriers (Table 1). Only 1 (11%) institution had an SOP for use and disinfection. Infection prevention teams were involved in assisting with VR disinfection protocols at 2 (22%) sites.", "A total of 175 experiments were performed to assess disinfection (Table 2). Adequate disinfection was achieved with both the IPA and the alcohol-free quaternary ammonium wipes across all bacterial types and headset material comparing untreated with disinfected surfaces. No bacteria were recovered in 88% (154/175) of experiments. IPA wipes performed better than the quaternary ammonium wipes at reducing overall bacterial counts (P=.001). This difference was most pronounced on porous surfaces, where the mean quantity of bacteria remaining after the alcohol-free quaternary ammonium use was more than after IPA use (Figure 2A).\nWhen comparing the disinfection by an organism, there were more CFUs of S aureus and S epidermidis recovered after attempted disinfection than P aeruginosa (P=.05 and P=.03, respectively), with this difference most pronounced on porous surfaces (Figure 2B). There was no significant difference between the recovery of S aureus and S epidermidis (P=.72). Finally, fewer bacteria were recovered from porous surfaces after inoculation but prior to performing disinfection. However, more bacteria were recovered after disinfection from porous surfaces than from nonporous surfaces (P=.01) as shown in Figure 2C, confirming that porous materials inoculated (Figure 1) were more difficult to disinfect because of their availability to absorb the bacteria-containing liquid.\nBacterial count by disinfection method, organism, and surface.\naThe percentage of times when 0 bacterial colony-forming units were observed.\nbNonporous surfaces were the Quest 1 Strap, Quest 2 Headset, and controller, while the porous surfaces were the Quest 1 Headset and Quest 2 Strap.\nThe mean (SEM) raw bacterial counts recovered after disinfection according to disinfectant and equipment (A), by organism and equipment (B), and by equipment surface type (C) are displayed. Note: Non-porous surfaces were the Quest 1 Strap, Quest 2 Headset and Controller, while porous surfaces were the Quest 1 Headset and Quest 2 Strap." ]
[ null, null, null, null, null, null ]
[ "Introduction", "Methods", "Survey", "Ethical Considerations", "Laboratory Disinfection", "Statistical Analysis", "Results", "Current VR Disinfection Practices in Pediatric Hospitals", "Effectiveness of Hospital-Grade Disinfecting Wipes on VR Decontamination", "Discussion" ]
[ "Virtual reality (VR) devices are increasingly used in health care settings to benefit patients, and the examples include patients with posttraumatic stress disorder, anxiety, complex regional pain syndrome, and distraction therapy [1-4]. Recent data show the benefit expands to the pediatric population as well by reducing pain and anxiety during medical procedures through distraction [5]. VR can also be used to educate health care workers through training and simulation [6]. However, a lack of standardized cleaning and disinfection processes for VR devices has limited VR’s use in health care settings, especially during the COVID-19 pandemic [6].\nNosocomial transmission and outbreaks have been reported with many different types of medical devices in clinical use [7], and establishing standard operating procedures (SOPs) for disinfection of VR devices between patient use is paramount. One of the most common and widely sold VR headset devices worldwide, the Meta Oculus Quest 2, specifically, recommends against the use of alcohol to clean and disinfect the device in favor of antibacterial wipes due to theoretical concerns about affecting the porous material [8]. This poses challenges in clinical settings as isopropyl alcohol (IPA) is one of the most common disinfectants used on medical devices.\nA protocol to clean and disinfect VR devices used in health care settings has been proposed [6]; however, no studies have quantified the efficacy of hospital-grade disinfectants on different parts of the VR equipment. Additionally, little is known about how these machines are currently disinfected in health care settings. In this mixed methods evaluation, we sought to determine the current disinfection practices in health care settings and how they were established. We also studied the effect of commonly used disinfectant wipes on the disinfection of VR headsets experimentally contaminated with common bacterial pathogens to provide evidence for the creation of an SOP to reduce infections with multipatient VR utilization.", "[SUBTITLE] Survey [SUBSECTION] To learn how health care facilities disinfect VR equipment and whether infection prevention teams are involved, a voluntary Qualtrics survey was sent via an electronic link in a group chat of 50 VR technicians working at children’s hospitals across the United States as a convenience sample.\nTo learn how health care facilities disinfect VR equipment and whether infection prevention teams are involved, a voluntary Qualtrics survey was sent via an electronic link in a group chat of 50 VR technicians working at children’s hospitals across the United States as a convenience sample.\n[SUBTITLE] Ethical Considerations [SUBSECTION] The survey was approved as exempt by the Yale University institutional review board (study #2000033075).\nThe survey was approved as exempt by the Yale University institutional review board (study #2000033075).\n[SUBTITLE] Laboratory Disinfection [SUBSECTION] Three types of bacteria, Staphylococcus epidermidis (ATCC 12228), Pseudomonas aeruginosa (laboratory strain PAO1), and Staphylococcus aureus (ATCC 25923), were chosen because of their propensity to be present on the skin and cause infection in children with compromised immune systems. The bacteria were grown overnight in 3 mL of lysogeny broth and serially diluted to quantitate the bacterial inoculum. VR headsets and controllers were inoculated by spreading 10 μL (initial inoculum 4.1×106-4.5×108) onto various sites (Figure 1) and allowed to dry for 30 minutes. This large inoculum was chosen to test whether Environmental Protection Agency (EPA)–approved disinfectants achieved sufficient log-reduction in bacteria as per their instructions for use. Two VR devices were experimentally contaminated: the Oculus Quest headset and the Oculus Quest 2 headset and hand controllers (Reality Labs, Meta Platforms). These devices were chosen as these are the most popular consumer devices, the primary ones used at our institution, and contained different surface types to trial disinfection [9]. Contaminated sites included the outer surface of the headset housing on the top side, the controller buttons, and the headband straps for each device (Figure 1). These sites were chosen as they were thought to be high–touch point areas for the hands and head during patient use. We did not study the facial interface as it is our standard practice to use a disposable barrier between the facial interface and the patient’s skin. Both nonporous (Oculus Quest 1 strap, Oculus Quest 2 headset, and controller) and porous (Oculus Quest 1 headset and Oculus Quest 2 strap) surfaces were contaminated to assess disinfection efficacy.\nTwo products were tested for active disinfection: a 70% IPA wipe (Medium Alcohol Prep Pad, Medline) and an alcohol-free quaternary ammonium wipe (Sani-Cloth AF3 Germicidal Disposable Wipe, PDI). A positive control of inoculation without disinfection was performed for every experiment and cultured after the dry time to account for bacterial cell death from desiccation. Disinfection was performed in accordance with each product’s manufacturer’s instructions for use. For the IPA wipe, a 15-second scrub in a back-and-forth motion followed by a 15-second dry time was performed. For the alcohol-free quaternary ammonium wipe, the experimentally contaminated area was wiped to a point of saturation for 3 minutes and allowed to dry. Following disinfection, the cultures were obtained with sterile cotton swabs dipped in Dey-Engley (D/E) neutralizing broth (Hardy Diagnostics) and wiped across the entirety of the contaminated surface for 5 seconds in a back-and-forth motion. The swab was used to inoculate a D/E agar plate that was then incubated overnight at 37 C. Bacterial colony forming units (CFUs) were counted on the plates the following day. Between experiments, the entirety of the VR devices was disinfected with a 70% IPA spray to the point of saturation of the surface materials and dried overnight. The laboratory experiments satisfied the Yale University 100 CH.9 Clinical Quality Improvement criteria and were exempt from institutional review board approval.\nAreas where cultures were obtained (shown by red circles) of Oculus Quest (top) and Oculus Quest 2 (bottom) devices.\nThree types of bacteria, Staphylococcus epidermidis (ATCC 12228), Pseudomonas aeruginosa (laboratory strain PAO1), and Staphylococcus aureus (ATCC 25923), were chosen because of their propensity to be present on the skin and cause infection in children with compromised immune systems. The bacteria were grown overnight in 3 mL of lysogeny broth and serially diluted to quantitate the bacterial inoculum. VR headsets and controllers were inoculated by spreading 10 μL (initial inoculum 4.1×106-4.5×108) onto various sites (Figure 1) and allowed to dry for 30 minutes. This large inoculum was chosen to test whether Environmental Protection Agency (EPA)–approved disinfectants achieved sufficient log-reduction in bacteria as per their instructions for use. Two VR devices were experimentally contaminated: the Oculus Quest headset and the Oculus Quest 2 headset and hand controllers (Reality Labs, Meta Platforms). These devices were chosen as these are the most popular consumer devices, the primary ones used at our institution, and contained different surface types to trial disinfection [9]. Contaminated sites included the outer surface of the headset housing on the top side, the controller buttons, and the headband straps for each device (Figure 1). These sites were chosen as they were thought to be high–touch point areas for the hands and head during patient use. We did not study the facial interface as it is our standard practice to use a disposable barrier between the facial interface and the patient’s skin. Both nonporous (Oculus Quest 1 strap, Oculus Quest 2 headset, and controller) and porous (Oculus Quest 1 headset and Oculus Quest 2 strap) surfaces were contaminated to assess disinfection efficacy.\nTwo products were tested for active disinfection: a 70% IPA wipe (Medium Alcohol Prep Pad, Medline) and an alcohol-free quaternary ammonium wipe (Sani-Cloth AF3 Germicidal Disposable Wipe, PDI). A positive control of inoculation without disinfection was performed for every experiment and cultured after the dry time to account for bacterial cell death from desiccation. Disinfection was performed in accordance with each product’s manufacturer’s instructions for use. For the IPA wipe, a 15-second scrub in a back-and-forth motion followed by a 15-second dry time was performed. For the alcohol-free quaternary ammonium wipe, the experimentally contaminated area was wiped to a point of saturation for 3 minutes and allowed to dry. Following disinfection, the cultures were obtained with sterile cotton swabs dipped in Dey-Engley (D/E) neutralizing broth (Hardy Diagnostics) and wiped across the entirety of the contaminated surface for 5 seconds in a back-and-forth motion. The swab was used to inoculate a D/E agar plate that was then incubated overnight at 37 C. Bacterial colony forming units (CFUs) were counted on the plates the following day. Between experiments, the entirety of the VR devices was disinfected with a 70% IPA spray to the point of saturation of the surface materials and dried overnight. The laboratory experiments satisfied the Yale University 100 CH.9 Clinical Quality Improvement criteria and were exempt from institutional review board approval.\nAreas where cultures were obtained (shown by red circles) of Oculus Quest (top) and Oculus Quest 2 (bottom) devices.\n[SUBTITLE] Statistical Analysis [SUBSECTION] The experimental design was a 3-factor crossed design with bacterial CFUs as the outcome variable. The generalized linear model with binomial distribution and logit link was used to compare whether the proportion of trials with observable bacteria counts after disinfection differed by (1) type of disinfection, (2) type of organism, and (3) surface type. All models included the natural log of bacterial count prior to disinfection and used robust standard errors. Achieving disinfection was defined as a greater than a 6-log reduction in bacterial counts. Pairwise comparisons were performed for the type of organism. Analyses were performed in SPSS (version 27; IBM Corp), and statistical significance was set at an α level of .05.\nThe experimental design was a 3-factor crossed design with bacterial CFUs as the outcome variable. The generalized linear model with binomial distribution and logit link was used to compare whether the proportion of trials with observable bacteria counts after disinfection differed by (1) type of disinfection, (2) type of organism, and (3) surface type. All models included the natural log of bacterial count prior to disinfection and used robust standard errors. Achieving disinfection was defined as a greater than a 6-log reduction in bacterial counts. Pairwise comparisons were performed for the type of organism. Analyses were performed in SPSS (version 27; IBM Corp), and statistical significance was set at an α level of .05.", "To learn how health care facilities disinfect VR equipment and whether infection prevention teams are involved, a voluntary Qualtrics survey was sent via an electronic link in a group chat of 50 VR technicians working at children’s hospitals across the United States as a convenience sample.", "The survey was approved as exempt by the Yale University institutional review board (study #2000033075).", "Three types of bacteria, Staphylococcus epidermidis (ATCC 12228), Pseudomonas aeruginosa (laboratory strain PAO1), and Staphylococcus aureus (ATCC 25923), were chosen because of their propensity to be present on the skin and cause infection in children with compromised immune systems. The bacteria were grown overnight in 3 mL of lysogeny broth and serially diluted to quantitate the bacterial inoculum. VR headsets and controllers were inoculated by spreading 10 μL (initial inoculum 4.1×106-4.5×108) onto various sites (Figure 1) and allowed to dry for 30 minutes. This large inoculum was chosen to test whether Environmental Protection Agency (EPA)–approved disinfectants achieved sufficient log-reduction in bacteria as per their instructions for use. Two VR devices were experimentally contaminated: the Oculus Quest headset and the Oculus Quest 2 headset and hand controllers (Reality Labs, Meta Platforms). These devices were chosen as these are the most popular consumer devices, the primary ones used at our institution, and contained different surface types to trial disinfection [9]. Contaminated sites included the outer surface of the headset housing on the top side, the controller buttons, and the headband straps for each device (Figure 1). These sites were chosen as they were thought to be high–touch point areas for the hands and head during patient use. We did not study the facial interface as it is our standard practice to use a disposable barrier between the facial interface and the patient’s skin. Both nonporous (Oculus Quest 1 strap, Oculus Quest 2 headset, and controller) and porous (Oculus Quest 1 headset and Oculus Quest 2 strap) surfaces were contaminated to assess disinfection efficacy.\nTwo products were tested for active disinfection: a 70% IPA wipe (Medium Alcohol Prep Pad, Medline) and an alcohol-free quaternary ammonium wipe (Sani-Cloth AF3 Germicidal Disposable Wipe, PDI). A positive control of inoculation without disinfection was performed for every experiment and cultured after the dry time to account for bacterial cell death from desiccation. Disinfection was performed in accordance with each product’s manufacturer’s instructions for use. For the IPA wipe, a 15-second scrub in a back-and-forth motion followed by a 15-second dry time was performed. For the alcohol-free quaternary ammonium wipe, the experimentally contaminated area was wiped to a point of saturation for 3 minutes and allowed to dry. Following disinfection, the cultures were obtained with sterile cotton swabs dipped in Dey-Engley (D/E) neutralizing broth (Hardy Diagnostics) and wiped across the entirety of the contaminated surface for 5 seconds in a back-and-forth motion. The swab was used to inoculate a D/E agar plate that was then incubated overnight at 37 C. Bacterial colony forming units (CFUs) were counted on the plates the following day. Between experiments, the entirety of the VR devices was disinfected with a 70% IPA spray to the point of saturation of the surface materials and dried overnight. The laboratory experiments satisfied the Yale University 100 CH.9 Clinical Quality Improvement criteria and were exempt from institutional review board approval.\nAreas where cultures were obtained (shown by red circles) of Oculus Quest (top) and Oculus Quest 2 (bottom) devices.", "The experimental design was a 3-factor crossed design with bacterial CFUs as the outcome variable. The generalized linear model with binomial distribution and logit link was used to compare whether the proportion of trials with observable bacteria counts after disinfection differed by (1) type of disinfection, (2) type of organism, and (3) surface type. All models included the natural log of bacterial count prior to disinfection and used robust standard errors. Achieving disinfection was defined as a greater than a 6-log reduction in bacterial counts. Pairwise comparisons were performed for the type of organism. Analyses were performed in SPSS (version 27; IBM Corp), and statistical significance was set at an α level of .05.", "[SUBTITLE] Current VR Disinfection Practices in Pediatric Hospitals [SUBSECTION] A total of 50 VR technicians across the Unites States were invited to participate in the Qualtrics survey with a response rate of 18% (9/50). One person consented and then did not answer any of the questions. The selected results of the survey are shown in Table 1 and highlight the variability of VR use and disinfection practices.\nSurvey of current VRa disinfection practices in health care settings.\naVR: virtual reality.\nbSome respondents gave multiple responses.\nThe number of VR sessions varied from less than once a week to multiple times per day. Most locations used multiple types of VR platforms with the Starlight Children’s VR system most commonly used. The Starlight Children’s VR system is a variation of the Lenovo Mirage Solo VR headset and is made of very similar materials as the Oculus Quest 1 and Oculus Quest 2. All participants noted that disinfection was performed before patient use (n=9, 100%). The methods of disinfection were variable with IPA and quaternary ammonium low-level disinfection wipes used most commonly. Most VR technicians used physical barriers between the VR device and the patient such as combinations of silicon covers, disposable eye masks, wipeable replacement head straps, and hair covers, while 3 (33%) participants did not use barriers (Table 1). Only 1 (11%) institution had an SOP for use and disinfection. Infection prevention teams were involved in assisting with VR disinfection protocols at 2 (22%) sites.\nA total of 50 VR technicians across the Unites States were invited to participate in the Qualtrics survey with a response rate of 18% (9/50). One person consented and then did not answer any of the questions. The selected results of the survey are shown in Table 1 and highlight the variability of VR use and disinfection practices.\nSurvey of current VRa disinfection practices in health care settings.\naVR: virtual reality.\nbSome respondents gave multiple responses.\nThe number of VR sessions varied from less than once a week to multiple times per day. Most locations used multiple types of VR platforms with the Starlight Children’s VR system most commonly used. The Starlight Children’s VR system is a variation of the Lenovo Mirage Solo VR headset and is made of very similar materials as the Oculus Quest 1 and Oculus Quest 2. All participants noted that disinfection was performed before patient use (n=9, 100%). The methods of disinfection were variable with IPA and quaternary ammonium low-level disinfection wipes used most commonly. Most VR technicians used physical barriers between the VR device and the patient such as combinations of silicon covers, disposable eye masks, wipeable replacement head straps, and hair covers, while 3 (33%) participants did not use barriers (Table 1). Only 1 (11%) institution had an SOP for use and disinfection. Infection prevention teams were involved in assisting with VR disinfection protocols at 2 (22%) sites.\n[SUBTITLE] Effectiveness of Hospital-Grade Disinfecting Wipes on VR Decontamination [SUBSECTION] A total of 175 experiments were performed to assess disinfection (Table 2). Adequate disinfection was achieved with both the IPA and the alcohol-free quaternary ammonium wipes across all bacterial types and headset material comparing untreated with disinfected surfaces. No bacteria were recovered in 88% (154/175) of experiments. IPA wipes performed better than the quaternary ammonium wipes at reducing overall bacterial counts (P=.001). This difference was most pronounced on porous surfaces, where the mean quantity of bacteria remaining after the alcohol-free quaternary ammonium use was more than after IPA use (Figure 2A).\nWhen comparing the disinfection by an organism, there were more CFUs of S aureus and S epidermidis recovered after attempted disinfection than P aeruginosa (P=.05 and P=.03, respectively), with this difference most pronounced on porous surfaces (Figure 2B). There was no significant difference between the recovery of S aureus and S epidermidis (P=.72). Finally, fewer bacteria were recovered from porous surfaces after inoculation but prior to performing disinfection. However, more bacteria were recovered after disinfection from porous surfaces than from nonporous surfaces (P=.01) as shown in Figure 2C, confirming that porous materials inoculated (Figure 1) were more difficult to disinfect because of their availability to absorb the bacteria-containing liquid.\nBacterial count by disinfection method, organism, and surface.\naThe percentage of times when 0 bacterial colony-forming units were observed.\nbNonporous surfaces were the Quest 1 Strap, Quest 2 Headset, and controller, while the porous surfaces were the Quest 1 Headset and Quest 2 Strap.\nThe mean (SEM) raw bacterial counts recovered after disinfection according to disinfectant and equipment (A), by organism and equipment (B), and by equipment surface type (C) are displayed. Note: Non-porous surfaces were the Quest 1 Strap, Quest 2 Headset and Controller, while porous surfaces were the Quest 1 Headset and Quest 2 Strap.\nA total of 175 experiments were performed to assess disinfection (Table 2). Adequate disinfection was achieved with both the IPA and the alcohol-free quaternary ammonium wipes across all bacterial types and headset material comparing untreated with disinfected surfaces. No bacteria were recovered in 88% (154/175) of experiments. IPA wipes performed better than the quaternary ammonium wipes at reducing overall bacterial counts (P=.001). This difference was most pronounced on porous surfaces, where the mean quantity of bacteria remaining after the alcohol-free quaternary ammonium use was more than after IPA use (Figure 2A).\nWhen comparing the disinfection by an organism, there were more CFUs of S aureus and S epidermidis recovered after attempted disinfection than P aeruginosa (P=.05 and P=.03, respectively), with this difference most pronounced on porous surfaces (Figure 2B). There was no significant difference between the recovery of S aureus and S epidermidis (P=.72). Finally, fewer bacteria were recovered from porous surfaces after inoculation but prior to performing disinfection. However, more bacteria were recovered after disinfection from porous surfaces than from nonporous surfaces (P=.01) as shown in Figure 2C, confirming that porous materials inoculated (Figure 1) were more difficult to disinfect because of their availability to absorb the bacteria-containing liquid.\nBacterial count by disinfection method, organism, and surface.\naThe percentage of times when 0 bacterial colony-forming units were observed.\nbNonporous surfaces were the Quest 1 Strap, Quest 2 Headset, and controller, while the porous surfaces were the Quest 1 Headset and Quest 2 Strap.\nThe mean (SEM) raw bacterial counts recovered after disinfection according to disinfectant and equipment (A), by organism and equipment (B), and by equipment surface type (C) are displayed. Note: Non-porous surfaces were the Quest 1 Strap, Quest 2 Headset and Controller, while porous surfaces were the Quest 1 Headset and Quest 2 Strap.", "A total of 50 VR technicians across the Unites States were invited to participate in the Qualtrics survey with a response rate of 18% (9/50). One person consented and then did not answer any of the questions. The selected results of the survey are shown in Table 1 and highlight the variability of VR use and disinfection practices.\nSurvey of current VRa disinfection practices in health care settings.\naVR: virtual reality.\nbSome respondents gave multiple responses.\nThe number of VR sessions varied from less than once a week to multiple times per day. Most locations used multiple types of VR platforms with the Starlight Children’s VR system most commonly used. The Starlight Children’s VR system is a variation of the Lenovo Mirage Solo VR headset and is made of very similar materials as the Oculus Quest 1 and Oculus Quest 2. All participants noted that disinfection was performed before patient use (n=9, 100%). The methods of disinfection were variable with IPA and quaternary ammonium low-level disinfection wipes used most commonly. Most VR technicians used physical barriers between the VR device and the patient such as combinations of silicon covers, disposable eye masks, wipeable replacement head straps, and hair covers, while 3 (33%) participants did not use barriers (Table 1). Only 1 (11%) institution had an SOP for use and disinfection. Infection prevention teams were involved in assisting with VR disinfection protocols at 2 (22%) sites.", "A total of 175 experiments were performed to assess disinfection (Table 2). Adequate disinfection was achieved with both the IPA and the alcohol-free quaternary ammonium wipes across all bacterial types and headset material comparing untreated with disinfected surfaces. No bacteria were recovered in 88% (154/175) of experiments. IPA wipes performed better than the quaternary ammonium wipes at reducing overall bacterial counts (P=.001). This difference was most pronounced on porous surfaces, where the mean quantity of bacteria remaining after the alcohol-free quaternary ammonium use was more than after IPA use (Figure 2A).\nWhen comparing the disinfection by an organism, there were more CFUs of S aureus and S epidermidis recovered after attempted disinfection than P aeruginosa (P=.05 and P=.03, respectively), with this difference most pronounced on porous surfaces (Figure 2B). There was no significant difference between the recovery of S aureus and S epidermidis (P=.72). Finally, fewer bacteria were recovered from porous surfaces after inoculation but prior to performing disinfection. However, more bacteria were recovered after disinfection from porous surfaces than from nonporous surfaces (P=.01) as shown in Figure 2C, confirming that porous materials inoculated (Figure 1) were more difficult to disinfect because of their availability to absorb the bacteria-containing liquid.\nBacterial count by disinfection method, organism, and surface.\naThe percentage of times when 0 bacterial colony-forming units were observed.\nbNonporous surfaces were the Quest 1 Strap, Quest 2 Headset, and controller, while the porous surfaces were the Quest 1 Headset and Quest 2 Strap.\nThe mean (SEM) raw bacterial counts recovered after disinfection according to disinfectant and equipment (A), by organism and equipment (B), and by equipment surface type (C) are displayed. Note: Non-porous surfaces were the Quest 1 Strap, Quest 2 Headset and Controller, while porous surfaces were the Quest 1 Headset and Quest 2 Strap.", "Adequate disinfection of VR devices can be achieved through the use of low-level EPA-approved hospital-grade disinfectants commonly used in clinical settings for devices that are exposed to intact skin, including IPA and quaternary ammonium wipes. We found a greater than 6-fold logarithmic reduction from initial bacteria inoculation across all pathogen types and VR device surfaces when using either product. However, we did observe the differences when evaluating raw-bacterial counts after disinfection. Notably, IPA performed better than the quaternary ammonium wipe, particularly for porous surfaces. It is possible that IPA penetrates porous surfaces better than quaternary ammonium products due to the vigorous 15-second scrub, and future studies should evaluate how well different wipes perform on these types of surfaces. We also observed that S aureus and S epidermidis persisted on surfaces at greater densities than P aeruginosa, possibly reflective of a mechanism in strain type or environmental survivability. Finally, and perhaps most critically, we observed lower bacterial counts after inoculation but before disinfection, and greater bacterial counts after disinfection, on porous surfaces when compared to nonporous surfaces. This suggests that bacteria may be entering the pores in the material, potentially reducing exposure to the disinfection material. Additionally, using swabs to recover bacteria from porous surfaces is suboptimal as we do not recover bacteria that have penetrated deeper into the material as well as nonporous surfaces which have better transfer efficiency [10]. Thus, despite consistent recovery after disinfection from porous surfaces in these experiments, we likely have overestimated the efficacy of disinfection for this material. Of note, there was outstanding disinfection of all nonporous surfaces, making it the preferred material for the construction of VR devices in health care. Manufacturers should consider material in the design of both headsets and straps, and our data support the use of nonporous material, particularly in health care settings where persistent bacteria may serve as a nidus for transmission to the next VR device user. If porous surfaces are present, there should be adequate barrier protection to prevent the transmission of microbes. This also then allows for the use of IPA without concern for damage to any porous components of the device.\nNosocomial transmission and outbreaks associated with the use of medical devices are well documented and a primary concern for using VR devices in health care settings [7]. Consequently, facilities may restrict VR devices from patient use out of concern. We found substantial variability between facilities in the frequency of device use, disinfection method, and barrier protection used. Importantly, almost all sites reported that infection prevention teams were not involved in performing a risk assessment for device use during patient care, and SOPs for disinfection were absent in all but one institution. Establishing a standard process that appropriately disinfects VR devices to allow for safe and expanded use in health care settings while avoiding equipment degradation can benefit patients and health care workers alike. It is critical to ensure that when new devices such as VR equipment are introduced into patient care settings, Infection Prevention and other stakeholders are involved prior to the purchase of the devices to ensure there is an acceptable plan for device reprocessing.\nBased on this generated data set, manufacturer’s instructions for use [8], health care infection prevention best practices, and previous literature or expert opinion [6], we propose an SOP for VR use and disinfection in the health care setting (Textbox 1). This is particularly important as the patient population served may be undergoing chemotherapy or other immunosuppressants that can increase the risk of infection.\n\nBefore use\n\nAvoid on patients with nonintact skin or active infections on the head or hands that cannot be covered and might come into contact with the device\nAvoid use on patients known to be colonized with pathogens where specialized disinfection is required, including Clostridioides difficile, Candida auris, Mycobacterium tuberculosis, and nonenveloped viruses\nPatient and staff perform hand hygiene\nA nonporous cover over the face pad, a disposable face pad cover, or both should serve as a barrier between the patient’s face and the device. Hair should also be covered (eg, bouffant and washable cloth surgical cap). Any porous material that makes contact with the patient’s skin or hair should be covered with a barrier\nDevices should be assessed for alcohol compatibility. If the device is not alcohol compatible, a nonalcohol-based disinfectant should be used\nPerform disinfection with a device compatible Environmental Protection Agency–registered product List H [11] according to the manufacturer’s instructions for use, ensuring that all surfaces of the headset (including the strap, the casing, the inner and outer facepieces, and the lens), the controller, and the nonporous, nondisposable face cover are saturated. Do not use wipes on multiple devices.\n\nAfter use\n\nPatient and staff perform hand hygiene\nStaff don appropriate personal protective equipment, which should include nitrile gloves at a minimum unless other personal protective equipment is required per the patient’s transmission-based isolation precautions\nRemove the device from the patient and placed on a clean disposable pad\nDiscard the disposable face cover, if present\nRemove nonporous, nondisposable face cover from the device, if present\nClean all visibly soiled areas with disposable wipes or paper towels\nRepeat disinfection as above\nAllow headset and controllers to dry according to the product instructions for use\nStore the device in a dry space physically separated from nondisinfected devices\nPatient and staff perform hand hygiene\nIn creating the SOP (Textbox 1), we considered VR devices a noncritical item requiring low-level disinfection between patients because they are most commonly exposed to intact skin only. Given disinfection success with the IPA and alcohol-free quaternary ammonium wipes, we suspect other equivalent low-level disinfectant products (eg, combination IPA or quaternary ammonium wipes) would be adequate, especially when applied to nonporous services [12]. We did not evaluate high-level disinfectants or sterilization procedures that may be required in the event of device exposure to nonintact skin or mucous membranes. We suggest avoiding VR device use on patients who have breaks in the skin on the hands or head region that cannot be appropriately covered and could come into contact with the device, thus avoiding the need for a high level of disinfection.\nAs disinfection was successfully achieved for a variety of pathogens, VR use is most likely safe for patients where contact isolation (gowns and gloves) is required in the hospital, including patients colonized with methicillin-resistant S aureus or vancomycin-resistant Enterococci. However, we suggest clinicians exercise caution when using VR devices with patients colonized with harder-to-eradicate pathogens such as Clostridioides difficile, Candida auris, and nonenveloped viruses where sodium hypochlorite or other high-level disinfection methods may be required. The pathogens tested (S aureus, P aeruginosa, and S epidermidis) are very common organisms in health care settings seen in both the adult and pediatric populations, and these results from disinfection are likely to be applicable to most health care settings, regardless of the patient’s age.\nLimitations of this study include the single-site nature limiting generalizability and the poor survey response rate. We also only tested 2 VR devices from the same company and 2 methods of surface disinfection. Preliminary experiments with an ultraviolet C (UVC) device specifically designed to decontaminate VR devices failed to produce adequate disinfection (data not shown). Since UVC disinfection depends on the angle and distance from the surface to the UVC source [13], the geometry of the headsets may make UVC disinfection more challenging. Further studies evaluating alternative disinfection methods, including UVC and other types of VR devices, are ongoing." ]
[ "introduction", "methods", null, null, null, null, "results", null, null, "discussion" ]
[ "disinfection", "healthcare-acquired infection", "healthcare worker", "virtual reality", "disinfect", "occupational health", "occupational safety", "infection control", "infection spread" ]
The psychological consequences of living with coronary heart disease: Are patients' psychological needs served? A mixed-method study in Germany.
36269637
This mixed-method study explores psychological needs, access and barriers in coronary heart disease (CHD) patients with and without mental health issues (MHI) within the German healthcare system.
INTRODUCTION
This study was conducted in three different healthcare settings: two hospitals, two rehabilitation clinics and three cardiology practices in Cologne, Germany. Patients were screened for angiographically documented CHD and other inclusion criteria. In total, 364 CHD patients took part in this study. It consisted of two parts: In the first part, participants filled in a newly developed questionnaire about their psychological needs, access and barriers within the healthcare system and their contact with their doctor in these matters. Then, patients were screened for MHIs with the help of the Hospital Anxiety and Depression Scale (HADS). When a score above seven was scored on the HADS, patients were additionally screened for specific MHIs using the Structured Clinical Interview for DSM-IV Axis I Disorders. In the second part, 20 participants were subsequently interviewed in a semi-structured interview to generate more in-depth findings.
METHODS
The interviews show that CHD patients with and without MHI experienced a cardiac event as life-changing and had an urgent need to talk about CHD with their doctor, mostly the general practitioner (GP). When the GP spoke to the patient shortly after the cardiac event, patients experienced relief and were better able to cope with their illness. Only 9.1% reported being aided in their search for psychotherapeutic treatment or drug treatment (4.1%).
RESULTS
The needs of CHD patients with and without MHI were not adequately satisfied within our sample. Psychological measures are necessary for sufficient improvement, such as training of doctors in doctor-patient communication (e.g., better support in coping with MHI/CHD), improvements in the procedure (more time for conversations during doctor contacts), and improvement of structural requirements (referring patients faster to psychotherapists).
CONCLUSION
[ "Humans", "Coronary Disease", "Research Design", "Germany", "Adaptation, Psychological", "Surveys and Questionnaires" ]
9700170
null
null
null
null
RESULTS
[SUBTITLE] Sociodemographic and clinical characteristics [SUBSECTION] Most participants were male (n = 258, 70.9%). The mean age across genders was M age = 65.9 (SD = 11.4); 72.5% lived together with a partner (n = 264); 27.5% had a left ventricular ejection fraction of <40%; 63.2% had a PCI for a myocardial infarction; 52.5% (n = 191) received a positive HADS score and 28.0% (n = 102) had a positive SCID diagnosis. Twelve patients had a positive HADS score but refused further testing (SCID‐I). The following SCID diagnoses were found: unipolar depression (n = 59, 16.5%), bipolar disorder (n = 2, 0.6%), anxiety disorder (n = 26, 7.2%), substance use/addiction disorder (n = 14, 4.0%), adjustment disorder (n = 11, 3.0%) and posttraumatic stress disorder (n = 4, 1.1%). Table 1 provides an overview of the demographic and clinical characteristics. For more details on the ICD‐10 diagnoses, see Table A1. Sociodemographic and clinical characteristics of the participants Abbreviations: MHI: mental health issue; NYHA; New York Heart Association; PCI, percutaneous coronary intervention. It is possible for patients to have more than one answer. At least one cell was too small for the appropriate analysis. Most participants were male (n = 258, 70.9%). The mean age across genders was M age = 65.9 (SD = 11.4); 72.5% lived together with a partner (n = 264); 27.5% had a left ventricular ejection fraction of <40%; 63.2% had a PCI for a myocardial infarction; 52.5% (n = 191) received a positive HADS score and 28.0% (n = 102) had a positive SCID diagnosis. Twelve patients had a positive HADS score but refused further testing (SCID‐I). The following SCID diagnoses were found: unipolar depression (n = 59, 16.5%), bipolar disorder (n = 2, 0.6%), anxiety disorder (n = 26, 7.2%), substance use/addiction disorder (n = 14, 4.0%), adjustment disorder (n = 11, 3.0%) and posttraumatic stress disorder (n = 4, 1.1%). Table 1 provides an overview of the demographic and clinical characteristics. For more details on the ICD‐10 diagnoses, see Table A1. Sociodemographic and clinical characteristics of the participants Abbreviations: MHI: mental health issue; NYHA; New York Heart Association; PCI, percutaneous coronary intervention. It is possible for patients to have more than one answer. At least one cell was too small for the appropriate analysis. [SUBTITLE] Patients' needs [SUBSECTION] A total of 80.8% with MHI and 74.2% without MHI found it appropriate to have been approached by the doctor (e.g., GP or cardiologist) about MHI; several patients were unsure whether they found it appropriate (5.8% with MHI; 7.5% without MHI). Patients expressed the general wish to be approached proactively by their doctor about MHI (46.5% with MHI; 38.0% without MHI). Approximately a fifth of patients in both groups were unsure about whether they wished to be approached by the doctor about MHI. About 23.4% of MHI patients mentioned a preference for GPs to help them find adequate treatment for drug therapy. For detailed information, see Table 2. Details to access, barriers and needs regarding healthcare in CHD patients with and without mental health issues (MHI) Abbreviation: CHD, coronary heart disease. Multiple‐choice question. All percentages relate to the maximum number of patients who were eligible for the underlying question. At least one cell was too small for the appropriate analysis. A total of 80.8% with MHI and 74.2% without MHI found it appropriate to have been approached by the doctor (e.g., GP or cardiologist) about MHI; several patients were unsure whether they found it appropriate (5.8% with MHI; 7.5% without MHI). Patients expressed the general wish to be approached proactively by their doctor about MHI (46.5% with MHI; 38.0% without MHI). Approximately a fifth of patients in both groups were unsure about whether they wished to be approached by the doctor about MHI. About 23.4% of MHI patients mentioned a preference for GPs to help them find adequate treatment for drug therapy. For detailed information, see Table 2. Details to access, barriers and needs regarding healthcare in CHD patients with and without mental health issues (MHI) Abbreviation: CHD, coronary heart disease. Multiple‐choice question. All percentages relate to the maximum number of patients who were eligible for the underlying question. At least one cell was too small for the appropriate analysis. [SUBTITLE] Access and barriers to healthcare [SUBSECTION] A total of 30 CHD patients with MHI (42.3%) stated that they received an adequate amount of information on the content and accessibility of psychological care. Only a minority of patients obtained information about psychotherapy (7.0%) with a quarter receiving no information at all. About 30% of patients obtained information on the content and accessibility of psychological care through their doctors, while 35% stated that they did not receive further information. When patients did obtain information, half of all patients with MHI got information from the GP or other persons outside of the healthcare system (e.g., family members, friends); 47.8% of MHI patients and 44.1% of patients without MHI wished to get this specific information from the GP. For further information, see Table 2. A total of 30 CHD patients with MHI (42.3%) stated that they received an adequate amount of information on the content and accessibility of psychological care. Only a minority of patients obtained information about psychotherapy (7.0%) with a quarter receiving no information at all. About 30% of patients obtained information on the content and accessibility of psychological care through their doctors, while 35% stated that they did not receive further information. When patients did obtain information, half of all patients with MHI got information from the GP or other persons outside of the healthcare system (e.g., family members, friends); 47.8% of MHI patients and 44.1% of patients without MHI wished to get this specific information from the GP. For further information, see Table 2. [SUBTITLE] Qualitative [SUBSECTION] In total, 65 CHD patients agreed to participate in a qualitative interview. These patients were mostly men (n = 53) and had an ejection fraction of <40% (n = 41). Divided over the three settings, 21.5% of patients came from hospitals, 33.9% from rehabilitation clinics and 44.6% from cardiology practices. From this subpopulation, 20 patients were selected and asked to participate. The mean age was M = 67.2 (SD = 12.6). All patients lived with a partner. For detailed information about the selection process, see Figure 1. In total, 65 CHD patients agreed to participate in a qualitative interview. These patients were mostly men (n = 53) and had an ejection fraction of <40% (n = 41). Divided over the three settings, 21.5% of patients came from hospitals, 33.9% from rehabilitation clinics and 44.6% from cardiology practices. From this subpopulation, 20 patients were selected and asked to participate. The mean age was M = 67.2 (SD = 12.6). All patients lived with a partner. For detailed information about the selection process, see Figure 1. [SUBTITLE] Perception of the cardiac event and approaches by doctors to talk about MHI [SUBSECTION] The following section contains information about how the patients experienced the cardiac event and whether they were really approached by the doctor to talk about MHI. Patients experienced CHD as a decisive turning point that unsettled them permanently. Most respondents felt significant anxiety and uncertainty in the weeks following the cardiac event. After diagnosis, spouses and family were the central persons to whom patients articulated their fears. Only a few patients reported that they were approached about MHI by a doctor as part of the CHD diagnosis. In general, MHI was not discussed until months after the diagnosis. Few patients engaged in a conversation about MHI with a professional. They reported that either the GP or a psychologist in a rehabilitation clinic approached them.No, not really. No one actually asked, no doctor, whether there was still a need for treatment. Patient 16, l. 22–24Of course, the doctor should have to ask more background information. How the patient is doing in other areas after this disease, whether he has psychological issues. I think that most specialists, e.g., cardiologists, proceed very technocratically and reel off their plan and don't care much about other things. I have not met any cardiologist, except for the head physician at the university hospital, who has dealt more intensively with this issue. For them it is only important how the echography is, how the ultrasound examination is, the blood values, etc. That's it. I even believe that cardiologists don't have a heart (laughs). Patient 9, l. 78–85Well, I have the feeling that my cardiologist doesn't get involved with me on an emotional level. It's relatively quick, he does a ECG or an ultrasound or something else and then I get my information and have to leave. It's not something where you have a longer conversation, that's what they do. only the treatment, then you go back to the treating GP. Patient 14, l. 131–137 No, not really. No one actually asked, no doctor, whether there was still a need for treatment. Patient 16, l. 22–24 Of course, the doctor should have to ask more background information. How the patient is doing in other areas after this disease, whether he has psychological issues. I think that most specialists, e.g., cardiologists, proceed very technocratically and reel off their plan and don't care much about other things. I have not met any cardiologist, except for the head physician at the university hospital, who has dealt more intensively with this issue. For them it is only important how the echography is, how the ultrasound examination is, the blood values, etc. That's it. I even believe that cardiologists don't have a heart (laughs). Patient 9, l. 78–85 Well, I have the feeling that my cardiologist doesn't get involved with me on an emotional level. It's relatively quick, he does a ECG or an ultrasound or something else and then I get my information and have to leave. It's not something where you have a longer conversation, that's what they do. only the treatment, then you go back to the treating GP. Patient 14, l. 131–137 The following section contains information about how the patients experienced the cardiac event and whether they were really approached by the doctor to talk about MHI. Patients experienced CHD as a decisive turning point that unsettled them permanently. Most respondents felt significant anxiety and uncertainty in the weeks following the cardiac event. After diagnosis, spouses and family were the central persons to whom patients articulated their fears. Only a few patients reported that they were approached about MHI by a doctor as part of the CHD diagnosis. In general, MHI was not discussed until months after the diagnosis. Few patients engaged in a conversation about MHI with a professional. They reported that either the GP or a psychologist in a rehabilitation clinic approached them.No, not really. No one actually asked, no doctor, whether there was still a need for treatment. Patient 16, l. 22–24Of course, the doctor should have to ask more background information. How the patient is doing in other areas after this disease, whether he has psychological issues. I think that most specialists, e.g., cardiologists, proceed very technocratically and reel off their plan and don't care much about other things. I have not met any cardiologist, except for the head physician at the university hospital, who has dealt more intensively with this issue. For them it is only important how the echography is, how the ultrasound examination is, the blood values, etc. That's it. I even believe that cardiologists don't have a heart (laughs). Patient 9, l. 78–85Well, I have the feeling that my cardiologist doesn't get involved with me on an emotional level. It's relatively quick, he does a ECG or an ultrasound or something else and then I get my information and have to leave. It's not something where you have a longer conversation, that's what they do. only the treatment, then you go back to the treating GP. Patient 14, l. 131–137 No, not really. No one actually asked, no doctor, whether there was still a need for treatment. Patient 16, l. 22–24 Of course, the doctor should have to ask more background information. How the patient is doing in other areas after this disease, whether he has psychological issues. I think that most specialists, e.g., cardiologists, proceed very technocratically and reel off their plan and don't care much about other things. I have not met any cardiologist, except for the head physician at the university hospital, who has dealt more intensively with this issue. For them it is only important how the echography is, how the ultrasound examination is, the blood values, etc. That's it. I even believe that cardiologists don't have a heart (laughs). Patient 9, l. 78–85 Well, I have the feeling that my cardiologist doesn't get involved with me on an emotional level. It's relatively quick, he does a ECG or an ultrasound or something else and then I get my information and have to leave. It's not something where you have a longer conversation, that's what they do. only the treatment, then you go back to the treating GP. Patient 14, l. 131–137 [SUBTITLE] Patients' needs [SUBSECTION] [SUBTITLE] The wish for a conversation about MHI [SUBSECTION] All patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210 I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70 It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222 But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210 All patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210 I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70 It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222 But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210 [SUBTITLE] No wish for a conversation about MHI [SUBSECTION] A minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72 No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72 A minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72 No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72 [SUBTITLE] Access and barriers [SUBSECTION] Limited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96 The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200 Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96 Limited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96 The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200 Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96 [SUBTITLE] Specific age‐related issues [SUBSECTION] Besides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’. Besides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’. [SUBTITLE] The younger patient group and their concerns about their ability to work [SUBSECTION] The needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50 Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51 I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50 The needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50 Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51 I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50 [SUBTITLE] The older patient group and their concerns about successful aging [SUBSECTION] On the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144 ‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144 On the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144 ‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144 [SUBTITLE] The wish for a conversation about MHI [SUBSECTION] All patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210 I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70 It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222 But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210 All patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210 I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70 It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222 But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210 [SUBTITLE] No wish for a conversation about MHI [SUBSECTION] A minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72 No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72 A minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72 No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72 [SUBTITLE] Access and barriers [SUBSECTION] Limited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96 The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200 Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96 Limited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96 The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200 Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96 [SUBTITLE] Specific age‐related issues [SUBSECTION] Besides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’. Besides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’. [SUBTITLE] The younger patient group and their concerns about their ability to work [SUBSECTION] The needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50 Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51 I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50 The needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50 Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51 I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50 [SUBTITLE] The older patient group and their concerns about successful aging [SUBSECTION] On the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144 ‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144 On the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144 ‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144
CONCLUSION
This study found that the needs regarding CHD patients with and without MHI were not adequately satisfied. Furthermore, for CHD patients with MHI, access and barriers to the healthcare system regarding psychotherapeutic and drug therapy were insufficient. CHD patients experienced the cardiac event as life‐changing and had an urgent need to talk about mental problems. Primarily, the GP was named as a partner for conversations about CHD‐related mental problems; however, when the doctor spoke to the patient shortly after the cardiac event, patients felt relieved and coped better with their illness. To improve this, psychological measures such as training of doctors in doctor–patient communication are necessary, as well as improvements in procedural and structural requirements.
[ "INTRODUCTION", "Coronary heart disease", "Mental health issues and their relationships with CHD", "MHIs as barrier to medical adherence", "Current study", "Design", "Patient contribution", "Participants", "Research tools", "Quantitative", "Qualitative", "Procedures", "Quantitative", "Qualitative", "Data analysis", "Quantitative", "Qualitative", "Sociodemographic and clinical characteristics", "Patients' needs", "Access and barriers to healthcare", "Qualitative", "Perception of the cardiac event and approaches by doctors to talk about MHI", "Patients' needs", "The wish for a conversation about MHI", "No wish for a conversation about MHI", "Access and barriers", "Specific age‐related issues", "The younger patient group and their concerns about their ability to work", "The older patient group and their concerns about successful aging", "Perception of the cardiac event and patients' needs", "Access and barriers to healthcare and specific issues of patient groups", "Strengths, limitations and further research", "AUTHOR CONTRIBUTIONS" ]
[ "[SUBTITLE] Coronary heart disease [SUBSECTION] Coronary heart disease (CHD) is the leading cause of mortality, accounting for approximately one‐third of all deaths in individuals over the age of 35 years, with a death rate of 102.6 per 100,000.\n1\n In Germany, the lifetime prevalence of CHD for the age group 40−79 years is 9.3% (8.4−10.3).\n2\n Short‐term mortality has been decreased due to advances in acute CHD treatments. However, at a population level, CHD morbidity has increased in recent years. After an interim decline in smoking, hypercholesterolaemia and hypertension, these currently appear to be on the rise again.\n1\n, \n3\n, \n4\n\n\nCoronary heart disease (CHD) is the leading cause of mortality, accounting for approximately one‐third of all deaths in individuals over the age of 35 years, with a death rate of 102.6 per 100,000.\n1\n In Germany, the lifetime prevalence of CHD for the age group 40−79 years is 9.3% (8.4−10.3).\n2\n Short‐term mortality has been decreased due to advances in acute CHD treatments. However, at a population level, CHD morbidity has increased in recent years. After an interim decline in smoking, hypercholesterolaemia and hypertension, these currently appear to be on the rise again.\n1\n, \n3\n, \n4\n\n\n[SUBTITLE] Mental health issues and their relationships with CHD [SUBSECTION] It has been found that mental health issues (MHIs) are highly prevalent among patients with CHD. MHI is defined as a recognizable set of clinical symptoms and/or behavioural problems that correlate with individual distress and impairment of functioning at the individual level.\n5\n After a myocardial infarction, almost 30% of these patients suffer from depressive symptoms and 20% even fulfil ICD‐10 criteria for a depressive episode.\n6\n Depression is associated with higher CHD‐related morbidity and mortality (relative risk = 1.6–2.4): After a cardiac event such as a myocardial infarction, CHD patients with comorbid depression have twofold greater mortality during the next 2 years.\n7\n Furthermore, mortality risk can increase sixfold when the patient suffers from severe depressive symptoms.\n8\n\n\nIt has been found that mental health issues (MHIs) are highly prevalent among patients with CHD. MHI is defined as a recognizable set of clinical symptoms and/or behavioural problems that correlate with individual distress and impairment of functioning at the individual level.\n5\n After a myocardial infarction, almost 30% of these patients suffer from depressive symptoms and 20% even fulfil ICD‐10 criteria for a depressive episode.\n6\n Depression is associated with higher CHD‐related morbidity and mortality (relative risk = 1.6–2.4): After a cardiac event such as a myocardial infarction, CHD patients with comorbid depression have twofold greater mortality during the next 2 years.\n7\n Furthermore, mortality risk can increase sixfold when the patient suffers from severe depressive symptoms.\n8\n\n\n[SUBTITLE] MHIs as barrier to medical adherence [SUBSECTION] MHI also acts as a strong barrier to treatment adherence and exacerbates a required lifestyle change that is necessary to reduce CHD risk factors—regardless of whether MHI were already pre‐existing or occurred as a consequence of CHD.\n4\n For example, patients with MHI are less likely to exhibit healthy lifestyle behaviour (e.g., quit smoking) and have low medication adherence.\n9\n This emphasizes the drive for more patient involvement in terms of disease management and medical consultations.\n10\n At the same time, this group of patients finds it difficult to understand the course of the consultation and to present their own concerns adequately, resulting in a lack of communication of possible physical and/or MHIs to their doctor.\n11\n Hence, this specific population of CHD patients with MHI may be very vulnerable due to difficulties in disease management, low communication skills in a medical setting and problems communicating possible issues and questions about personal topics. This suggests that the quality of communication between patients and their doctors is important to secure the best course of treatment.\n12\n\n\nConsequently, adequate screening and treatment for MHI in CHD patients during the medical consultation from the doctor's perspective is urgently needed. National and international guidelines have recommended routine screening and treatment for MHI on primary and secondary CHD prevention.\n4\n, \n13\n However, studies have been able to show that this screening is not regularly followed due to lack of time and low occurrence of verbal interventions.\n14\n, \n15\n\n\nMHI also acts as a strong barrier to treatment adherence and exacerbates a required lifestyle change that is necessary to reduce CHD risk factors—regardless of whether MHI were already pre‐existing or occurred as a consequence of CHD.\n4\n For example, patients with MHI are less likely to exhibit healthy lifestyle behaviour (e.g., quit smoking) and have low medication adherence.\n9\n This emphasizes the drive for more patient involvement in terms of disease management and medical consultations.\n10\n At the same time, this group of patients finds it difficult to understand the course of the consultation and to present their own concerns adequately, resulting in a lack of communication of possible physical and/or MHIs to their doctor.\n11\n Hence, this specific population of CHD patients with MHI may be very vulnerable due to difficulties in disease management, low communication skills in a medical setting and problems communicating possible issues and questions about personal topics. This suggests that the quality of communication between patients and their doctors is important to secure the best course of treatment.\n12\n\n\nConsequently, adequate screening and treatment for MHI in CHD patients during the medical consultation from the doctor's perspective is urgently needed. National and international guidelines have recommended routine screening and treatment for MHI on primary and secondary CHD prevention.\n4\n, \n13\n However, studies have been able to show that this screening is not regularly followed due to lack of time and low occurrence of verbal interventions.\n14\n, \n15\n\n\n[SUBTITLE] Current study [SUBSECTION] In the current study, we explore the following research questions: (1) What are the MHI‐related needs of CHD patients with MHI after a cardiac event?, (2) how well do doctors meet their patients' needs? and (3) how do patients experience access and barriers to the healthcare system? It is hypothesized that patients with MHI have a desire for doctors to approach and support them with their MHI. Secondly, it is expected that doctors rarely meet their patients' needs. Lastly, it is hypothesized that, due to Hypotheses 1 and 2, patients receive a lack of access and distinct barriers to the healthcare system, especially related to their MHI. The aim of the study is to improve the quality of care and initiate changes in the structure of healthcare towards more patient‐centred care in the long term.\nIn the current study, we explore the following research questions: (1) What are the MHI‐related needs of CHD patients with MHI after a cardiac event?, (2) how well do doctors meet their patients' needs? and (3) how do patients experience access and barriers to the healthcare system? It is hypothesized that patients with MHI have a desire for doctors to approach and support them with their MHI. Secondly, it is expected that doctors rarely meet their patients' needs. Lastly, it is hypothesized that, due to Hypotheses 1 and 2, patients receive a lack of access and distinct barriers to the healthcare system, especially related to their MHI. The aim of the study is to improve the quality of care and initiate changes in the structure of healthcare towards more patient‐centred care in the long term.", "Coronary heart disease (CHD) is the leading cause of mortality, accounting for approximately one‐third of all deaths in individuals over the age of 35 years, with a death rate of 102.6 per 100,000.\n1\n In Germany, the lifetime prevalence of CHD for the age group 40−79 years is 9.3% (8.4−10.3).\n2\n Short‐term mortality has been decreased due to advances in acute CHD treatments. However, at a population level, CHD morbidity has increased in recent years. After an interim decline in smoking, hypercholesterolaemia and hypertension, these currently appear to be on the rise again.\n1\n, \n3\n, \n4\n\n", "It has been found that mental health issues (MHIs) are highly prevalent among patients with CHD. MHI is defined as a recognizable set of clinical symptoms and/or behavioural problems that correlate with individual distress and impairment of functioning at the individual level.\n5\n After a myocardial infarction, almost 30% of these patients suffer from depressive symptoms and 20% even fulfil ICD‐10 criteria for a depressive episode.\n6\n Depression is associated with higher CHD‐related morbidity and mortality (relative risk = 1.6–2.4): After a cardiac event such as a myocardial infarction, CHD patients with comorbid depression have twofold greater mortality during the next 2 years.\n7\n Furthermore, mortality risk can increase sixfold when the patient suffers from severe depressive symptoms.\n8\n\n", "MHI also acts as a strong barrier to treatment adherence and exacerbates a required lifestyle change that is necessary to reduce CHD risk factors—regardless of whether MHI were already pre‐existing or occurred as a consequence of CHD.\n4\n For example, patients with MHI are less likely to exhibit healthy lifestyle behaviour (e.g., quit smoking) and have low medication adherence.\n9\n This emphasizes the drive for more patient involvement in terms of disease management and medical consultations.\n10\n At the same time, this group of patients finds it difficult to understand the course of the consultation and to present their own concerns adequately, resulting in a lack of communication of possible physical and/or MHIs to their doctor.\n11\n Hence, this specific population of CHD patients with MHI may be very vulnerable due to difficulties in disease management, low communication skills in a medical setting and problems communicating possible issues and questions about personal topics. This suggests that the quality of communication between patients and their doctors is important to secure the best course of treatment.\n12\n\n\nConsequently, adequate screening and treatment for MHI in CHD patients during the medical consultation from the doctor's perspective is urgently needed. National and international guidelines have recommended routine screening and treatment for MHI on primary and secondary CHD prevention.\n4\n, \n13\n However, studies have been able to show that this screening is not regularly followed due to lack of time and low occurrence of verbal interventions.\n14\n, \n15\n\n", "In the current study, we explore the following research questions: (1) What are the MHI‐related needs of CHD patients with MHI after a cardiac event?, (2) how well do doctors meet their patients' needs? and (3) how do patients experience access and barriers to the healthcare system? It is hypothesized that patients with MHI have a desire for doctors to approach and support them with their MHI. Secondly, it is expected that doctors rarely meet their patients' needs. Lastly, it is hypothesized that, due to Hypotheses 1 and 2, patients receive a lack of access and distinct barriers to the healthcare system, especially related to their MHI. The aim of the study is to improve the quality of care and initiate changes in the structure of healthcare towards more patient‐centred care in the long term.", "MenDis‐CHD is a cross‐sectional study with a mixed‐method approach using an explanatory sequential design,\n16\n beginning with quantitative data collection (one questionnaire containing: questions about sociodemographic data, patients' needs and access and barriers to the healthcare system; psychological assessments [Hospital Anxiety and Depression Scale {HADS}, SCID‐I]), followed by a qualitative data acquisition (semi‐structured in‐depth interviews) to enrich the quantitative data. Specific details of the methods used in this study can be found in an earlier paper.\n17\n The questionnaire was fully developed in German. Individual translated items can be provided upon request.", "We received input from CHD patients during the pretests of the questionnaire and the qualitative interviews, as well as from members of CHD self‐help groups. The patient perspective has been continuously taken into account in the project and the interests of the patients have also been considered with regard to the transfer of results (e.g., through public lectures, summaries in plain language and design of a leaflet to share with peers and distribute to patient groups). Patients did not receive any compensation for contributing to this study.", "As inclusion criteria, patients had to have an angiographically documented CHD with stable angina pectoris, acute coronary syndromes, percutaneous coronary intervention (PCI) or bypass surgery, be older than 18 years, have sufficient German language skills and be able to give informed consent. Severe physical (e.g., cancer) or unstable mental problems (e.g., acute suicidal ideation, delirium and moderate to severe dementia) were the exclusion criteria.\nOverall, 753 patients were screened for eligibility, of whom 374 were finally enroled in the study. Ten patients dropped out due to withdrawal of informed consent or incomplete questionnaires with missing answers. Therefore, 364 patients were included in the final data analysis. We recruited 107 patients in hospitals, 157 patients in rehabilitation clinics and 100 patients in cardiology practices. Figure 1 features a flowchart presenting the recruitment process.\nFlowchart of the recruitment procedure. CHD, coronary heart disease; LVEF, left ventricular ejection fraction; MHI, mental health issue\nAll 364 included patients filled in the questionnaire. We used maximum variation as the sampling strategy to increase diversity by age, gender, marital status, employed/retired status, severity of CHD, left ventricular ejection fraction and kind and severity of diagnosis of MHI.\n18\n By explicitly aiming for recruiting a diverse sample, one ensures the generalizability of one's findings across a broader part of the general population. Furthermore, we asked every eligible and interested patient during the recruitment phase to also participate in our in‐depth interviews. In total, 20 patients were selected to participate in the qualitative interviews. The diversity of participants was actively monitored, but due to a significant number of patients who did not want to participate in the interviews, participant diversity was not completely balanced, especially gender and ethnicity. In this study, the sample consisted of approximately 70% males. According to the current literature, males are significantly more likely to develop CHD,\n19\n with a lifetime prevalence of 9.9% (9.0–10.8) than women, with 6.7% (6.0–7.4).\n20\n Thus, this study seems to represent an appropriate sample population. For further details about the distribution of samplings for interview groups, see Figure 1.", "[SUBTITLE] Quantitative [SUBSECTION] Sociodemographic data were assessed via a newly developed questionnaire. Disease severity of CHD patients was assessed by screening for cardiac events in previous medical history (e.g., myocardial infarction), cardiac surgeries (e.g., bypass surgery), congestive heart failure, somatic comorbidity, pre‐existing presence of MHI, left ventricular ejection fraction and New York Heart Association stage (classification scheme of heart diseases according to their degree of severity). All of these characteristics were drawn from patients' medical records; they were not part of the questionnaire. Clinical data were measured with the use of the HADS and SCID‐I.\nAn aforementioned self‐made questionnaire was made of five subsections of which two are used in this paper, namely ‘Patients' needs’ and ‘Access and barriers to healthcare system’. The questionnaire was developed and used in the German language only; all items and section headers provided in this article are translated.\nBoth sections, ‘Patients' needs’ and ‘Access and barriers to the healthcare system,’ were assessed by a 20‐item self‐report questionnaire. Patients without MHI could skip several items that contained questions about an existing MHI. The section ‘Patient's needs’ contained six questions, such as ‘Do you wish to be approached by the doctor on possible mental health issues?’ with binary answer possibilities (Yes/No) or multiple‐choice questions such as ‘Who do you think should give a recommendation for treatment in the case of mental health issues?’. The section ‘Access and barriers to healthcare system’ contained 14 questions, also designed with ‘yes or no’ or multiple‐choice questions such as ‘How often have you sufficiently received information on the content and accessibility of psychological care?’. The questionnaire was developed within this study with the help of four CHD patients with and without MHI and has yet to be validated. More information on the content of the questionnaire and how it was created can be found in our first publication on MHI.\n21\n\n\nThe HADS\n22\n, \n23\n was used as a general screening tool to assess symptoms of depression and anxiety and was filled in by the patients. In line with the advice of several studies,\n24\n a score of 8 and above was considered a ‘positive’ test result for anxiety or depression. In this case, the Structured Clinical Interview for DSM‐IV Axis I Disorders (SCID‐I)\n25\n was conducted as a semi‐structured interview to assess mental symptoms and disorders as defined in the ICD‐10.\n6\n\n\nSociodemographic data were assessed via a newly developed questionnaire. Disease severity of CHD patients was assessed by screening for cardiac events in previous medical history (e.g., myocardial infarction), cardiac surgeries (e.g., bypass surgery), congestive heart failure, somatic comorbidity, pre‐existing presence of MHI, left ventricular ejection fraction and New York Heart Association stage (classification scheme of heart diseases according to their degree of severity). All of these characteristics were drawn from patients' medical records; they were not part of the questionnaire. Clinical data were measured with the use of the HADS and SCID‐I.\nAn aforementioned self‐made questionnaire was made of five subsections of which two are used in this paper, namely ‘Patients' needs’ and ‘Access and barriers to healthcare system’. The questionnaire was developed and used in the German language only; all items and section headers provided in this article are translated.\nBoth sections, ‘Patients' needs’ and ‘Access and barriers to the healthcare system,’ were assessed by a 20‐item self‐report questionnaire. Patients without MHI could skip several items that contained questions about an existing MHI. The section ‘Patient's needs’ contained six questions, such as ‘Do you wish to be approached by the doctor on possible mental health issues?’ with binary answer possibilities (Yes/No) or multiple‐choice questions such as ‘Who do you think should give a recommendation for treatment in the case of mental health issues?’. The section ‘Access and barriers to healthcare system’ contained 14 questions, also designed with ‘yes or no’ or multiple‐choice questions such as ‘How often have you sufficiently received information on the content and accessibility of psychological care?’. The questionnaire was developed within this study with the help of four CHD patients with and without MHI and has yet to be validated. More information on the content of the questionnaire and how it was created can be found in our first publication on MHI.\n21\n\n\nThe HADS\n22\n, \n23\n was used as a general screening tool to assess symptoms of depression and anxiety and was filled in by the patients. In line with the advice of several studies,\n24\n a score of 8 and above was considered a ‘positive’ test result for anxiety or depression. In this case, the Structured Clinical Interview for DSM‐IV Axis I Disorders (SCID‐I)\n25\n was conducted as a semi‐structured interview to assess mental symptoms and disorders as defined in the ICD‐10.\n6\n\n\n[SUBTITLE] Qualitative [SUBSECTION] The qualitative module was designed to build on the quantitative part in a second phase to enrich the quantitative findings and further investigate the research questions of the study.\n16\n The qualitative study design was conducted according to the method of the problem‐centred interview.\n26\n This is a semi‐structured in‐depth interview approach that allows the respondent to speak as freely as possible but is centred on (a) predefined problem(s) to which the interviewer repeatedly refers back during the conversation. Based on the research questions of this study, three main categories were developed: (1) Category ‘Perception of the cardiac event and approaches of doctors to talk about MHI’: Were CHD patients approached by the doctor on MHI and after a cardiac event? How did patients experience this approach? What were their expectations? What were positive and negative aspects of the contact with the doctor? Example questions were ‘Have you been approached by the doctor after a cardiac event and if so, how did you experience it? How long did it take until you were asked? Which aspects have been discussed during your meeting? What were your expectations beforehand and what are your expectations now?’; (2) Category ‘Patient's needs’: Do patients want to talk about MHI? Who do patients want to talk to about MHI? If a doctor is the first choice, what type of doctor? Example questions: ‘Who asked you about MHI? Who would you have liked to be addressed by? What did you need (both physically and mentally) after you received your diagnosis?’; (3) Category ‘Access and barriers’: How exactly do patients experience access and barriers to the health care system? Example questions: ‘What information and offers of help were accessible? How well could then be implemented? What information or offers did you receive, which did you not receive albeit your wish to receive them?’\nThe qualitative module was designed to build on the quantitative part in a second phase to enrich the quantitative findings and further investigate the research questions of the study.\n16\n The qualitative study design was conducted according to the method of the problem‐centred interview.\n26\n This is a semi‐structured in‐depth interview approach that allows the respondent to speak as freely as possible but is centred on (a) predefined problem(s) to which the interviewer repeatedly refers back during the conversation. Based on the research questions of this study, three main categories were developed: (1) Category ‘Perception of the cardiac event and approaches of doctors to talk about MHI’: Were CHD patients approached by the doctor on MHI and after a cardiac event? How did patients experience this approach? What were their expectations? What were positive and negative aspects of the contact with the doctor? Example questions were ‘Have you been approached by the doctor after a cardiac event and if so, how did you experience it? How long did it take until you were asked? Which aspects have been discussed during your meeting? What were your expectations beforehand and what are your expectations now?’; (2) Category ‘Patient's needs’: Do patients want to talk about MHI? Who do patients want to talk to about MHI? If a doctor is the first choice, what type of doctor? Example questions: ‘Who asked you about MHI? Who would you have liked to be addressed by? What did you need (both physically and mentally) after you received your diagnosis?’; (3) Category ‘Access and barriers’: How exactly do patients experience access and barriers to the health care system? Example questions: ‘What information and offers of help were accessible? How well could then be implemented? What information or offers did you receive, which did you not receive albeit your wish to receive them?’", "Sociodemographic data were assessed via a newly developed questionnaire. Disease severity of CHD patients was assessed by screening for cardiac events in previous medical history (e.g., myocardial infarction), cardiac surgeries (e.g., bypass surgery), congestive heart failure, somatic comorbidity, pre‐existing presence of MHI, left ventricular ejection fraction and New York Heart Association stage (classification scheme of heart diseases according to their degree of severity). All of these characteristics were drawn from patients' medical records; they were not part of the questionnaire. Clinical data were measured with the use of the HADS and SCID‐I.\nAn aforementioned self‐made questionnaire was made of five subsections of which two are used in this paper, namely ‘Patients' needs’ and ‘Access and barriers to healthcare system’. The questionnaire was developed and used in the German language only; all items and section headers provided in this article are translated.\nBoth sections, ‘Patients' needs’ and ‘Access and barriers to the healthcare system,’ were assessed by a 20‐item self‐report questionnaire. Patients without MHI could skip several items that contained questions about an existing MHI. The section ‘Patient's needs’ contained six questions, such as ‘Do you wish to be approached by the doctor on possible mental health issues?’ with binary answer possibilities (Yes/No) or multiple‐choice questions such as ‘Who do you think should give a recommendation for treatment in the case of mental health issues?’. The section ‘Access and barriers to healthcare system’ contained 14 questions, also designed with ‘yes or no’ or multiple‐choice questions such as ‘How often have you sufficiently received information on the content and accessibility of psychological care?’. The questionnaire was developed within this study with the help of four CHD patients with and without MHI and has yet to be validated. More information on the content of the questionnaire and how it was created can be found in our first publication on MHI.\n21\n\n\nThe HADS\n22\n, \n23\n was used as a general screening tool to assess symptoms of depression and anxiety and was filled in by the patients. In line with the advice of several studies,\n24\n a score of 8 and above was considered a ‘positive’ test result for anxiety or depression. In this case, the Structured Clinical Interview for DSM‐IV Axis I Disorders (SCID‐I)\n25\n was conducted as a semi‐structured interview to assess mental symptoms and disorders as defined in the ICD‐10.\n6\n\n", "The qualitative module was designed to build on the quantitative part in a second phase to enrich the quantitative findings and further investigate the research questions of the study.\n16\n The qualitative study design was conducted according to the method of the problem‐centred interview.\n26\n This is a semi‐structured in‐depth interview approach that allows the respondent to speak as freely as possible but is centred on (a) predefined problem(s) to which the interviewer repeatedly refers back during the conversation. Based on the research questions of this study, three main categories were developed: (1) Category ‘Perception of the cardiac event and approaches of doctors to talk about MHI’: Were CHD patients approached by the doctor on MHI and after a cardiac event? How did patients experience this approach? What were their expectations? What were positive and negative aspects of the contact with the doctor? Example questions were ‘Have you been approached by the doctor after a cardiac event and if so, how did you experience it? How long did it take until you were asked? Which aspects have been discussed during your meeting? What were your expectations beforehand and what are your expectations now?’; (2) Category ‘Patient's needs’: Do patients want to talk about MHI? Who do patients want to talk to about MHI? If a doctor is the first choice, what type of doctor? Example questions: ‘Who asked you about MHI? Who would you have liked to be addressed by? What did you need (both physically and mentally) after you received your diagnosis?’; (3) Category ‘Access and barriers’: How exactly do patients experience access and barriers to the health care system? Example questions: ‘What information and offers of help were accessible? How well could then be implemented? What information or offers did you receive, which did you not receive albeit your wish to receive them?’", "[SUBTITLE] Quantitative [SUBSECTION] The questionnaire was pre‐tested on two persons with CHD, one with MHI and one without MHI, in July 2017. Participants were recruited between 15 January 2018 and 29 March 2019 in two hospital cardiac departments, three cardiology practices and two rehabilitation clinics in Cologne, Germany, to achieve an appropriate realistic sample of routine healthcare settings. After screening for eligibility through the researchers, all patients' demographic data were documented in our screening log. Those who fulfilled our inclusion criteria and provided written informed consent received the quantitative questionnaires. A second appointment was arranged to perform the SCID‐I if the HADS screening was ‘positive’. All researchers of this study were trained in applying the SCID‐I and experienced in conducting it. In addition, patients were asked to participate in qualitative interviews.\nThe questionnaire was pre‐tested on two persons with CHD, one with MHI and one without MHI, in July 2017. Participants were recruited between 15 January 2018 and 29 March 2019 in two hospital cardiac departments, three cardiology practices and two rehabilitation clinics in Cologne, Germany, to achieve an appropriate realistic sample of routine healthcare settings. After screening for eligibility through the researchers, all patients' demographic data were documented in our screening log. Those who fulfilled our inclusion criteria and provided written informed consent received the quantitative questionnaires. A second appointment was arranged to perform the SCID‐I if the HADS screening was ‘positive’. All researchers of this study were trained in applying the SCID‐I and experienced in conducting it. In addition, patients were asked to participate in qualitative interviews.\n[SUBTITLE] Qualitative [SUBSECTION] The interview guideline was pretested by two CHD patients: one with MHI and the other without. Pretests were conducted as face‐to‐face interviews in September 2018. Out of 65 participants who agreed to take part in a qualitative interview, 20 were randomly selected. The actual 20 interviews were carried out as face‐to‐face interviews between October 2018 and March 2019, either during a home visit or at the setting where the respective patient was recruited. Informed consent was obtained during the quantitative phase, but the interviewer informed all patients again by reading the information sheet and informed consent to them to ensure that they fully understood each element. The interview took 1 h. All interviews were audio‐recorded and transcribed.\nThe interview guideline was pretested by two CHD patients: one with MHI and the other without. Pretests were conducted as face‐to‐face interviews in September 2018. Out of 65 participants who agreed to take part in a qualitative interview, 20 were randomly selected. The actual 20 interviews were carried out as face‐to‐face interviews between October 2018 and March 2019, either during a home visit or at the setting where the respective patient was recruited. Informed consent was obtained during the quantitative phase, but the interviewer informed all patients again by reading the information sheet and informed consent to them to ensure that they fully understood each element. The interview took 1 h. All interviews were audio‐recorded and transcribed.", "The questionnaire was pre‐tested on two persons with CHD, one with MHI and one without MHI, in July 2017. Participants were recruited between 15 January 2018 and 29 March 2019 in two hospital cardiac departments, three cardiology practices and two rehabilitation clinics in Cologne, Germany, to achieve an appropriate realistic sample of routine healthcare settings. After screening for eligibility through the researchers, all patients' demographic data were documented in our screening log. Those who fulfilled our inclusion criteria and provided written informed consent received the quantitative questionnaires. A second appointment was arranged to perform the SCID‐I if the HADS screening was ‘positive’. All researchers of this study were trained in applying the SCID‐I and experienced in conducting it. In addition, patients were asked to participate in qualitative interviews.", "The interview guideline was pretested by two CHD patients: one with MHI and the other without. Pretests were conducted as face‐to‐face interviews in September 2018. Out of 65 participants who agreed to take part in a qualitative interview, 20 were randomly selected. The actual 20 interviews were carried out as face‐to‐face interviews between October 2018 and March 2019, either during a home visit or at the setting where the respective patient was recruited. Informed consent was obtained during the quantitative phase, but the interviewer informed all patients again by reading the information sheet and informed consent to them to ensure that they fully understood each element. The interview took 1 h. All interviews were audio‐recorded and transcribed.", "[SUBTITLE] Quantitative [SUBSECTION] All presented data were analysed using IBM SPSS Statistics 22 software\n27\n and controlled for both missing and impossible values. Identified cases with either missing or implausible values were compared to their equivalent paper versions and corrected. Multivariate analyses of variance and χ\n2 tests were conducted, depending on the curvature of the distribution of the variables' scores.\nAll presented data were analysed using IBM SPSS Statistics 22 software\n27\n and controlled for both missing and impossible values. Identified cases with either missing or implausible values were compared to their equivalent paper versions and corrected. Multivariate analyses of variance and χ\n2 tests were conducted, depending on the curvature of the distribution of the variables' scores.\n[SUBTITLE] Qualitative [SUBSECTION] The accuracy of the transcripts was checked based on the audiotape samples by two of the authors (a social scientist and a clinical psychologist). Data were imported to F4 Analysis\n28\n for content analysis.\n29\n In F4 Analysis, the audiotapes of the interviews were automatically converted into text. Then, both researchers listened to the tapes and checked the transcripts independently. Relevant clusters (content‐wise) were developed and presented to the steering group of MenDis‐CHD for interpretation. Data and coding within the different clusters were further analysed, named and defined as: (1) perception of cardiac event; (2) patient's needs; (3) access and barriers to the healthcare system and (4) specific age‐related issues. These categories reflect topics most important to patients during the semi‐structured interview. They were identified through discussion until an agreement was reached. Besides creating an overview of relevant content per category, this procedure was chosen to verify that patients would indeed talk about the research questions, as it was the aim to gather more qualitative information on them. Regular in‐depth discussion meetings of the steering group of MenDis‐CHD increased the reliability of the research results.\nThe accuracy of the transcripts was checked based on the audiotape samples by two of the authors (a social scientist and a clinical psychologist). Data were imported to F4 Analysis\n28\n for content analysis.\n29\n In F4 Analysis, the audiotapes of the interviews were automatically converted into text. Then, both researchers listened to the tapes and checked the transcripts independently. Relevant clusters (content‐wise) were developed and presented to the steering group of MenDis‐CHD for interpretation. Data and coding within the different clusters were further analysed, named and defined as: (1) perception of cardiac event; (2) patient's needs; (3) access and barriers to the healthcare system and (4) specific age‐related issues. These categories reflect topics most important to patients during the semi‐structured interview. They were identified through discussion until an agreement was reached. Besides creating an overview of relevant content per category, this procedure was chosen to verify that patients would indeed talk about the research questions, as it was the aim to gather more qualitative information on them. Regular in‐depth discussion meetings of the steering group of MenDis‐CHD increased the reliability of the research results.", "All presented data were analysed using IBM SPSS Statistics 22 software\n27\n and controlled for both missing and impossible values. Identified cases with either missing or implausible values were compared to their equivalent paper versions and corrected. Multivariate analyses of variance and χ\n2 tests were conducted, depending on the curvature of the distribution of the variables' scores.", "The accuracy of the transcripts was checked based on the audiotape samples by two of the authors (a social scientist and a clinical psychologist). Data were imported to F4 Analysis\n28\n for content analysis.\n29\n In F4 Analysis, the audiotapes of the interviews were automatically converted into text. Then, both researchers listened to the tapes and checked the transcripts independently. Relevant clusters (content‐wise) were developed and presented to the steering group of MenDis‐CHD for interpretation. Data and coding within the different clusters were further analysed, named and defined as: (1) perception of cardiac event; (2) patient's needs; (3) access and barriers to the healthcare system and (4) specific age‐related issues. These categories reflect topics most important to patients during the semi‐structured interview. They were identified through discussion until an agreement was reached. Besides creating an overview of relevant content per category, this procedure was chosen to verify that patients would indeed talk about the research questions, as it was the aim to gather more qualitative information on them. Regular in‐depth discussion meetings of the steering group of MenDis‐CHD increased the reliability of the research results.", "Most participants were male (n = 258, 70.9%). The mean age across genders was M\nage = 65.9 (SD = 11.4); 72.5% lived together with a partner (n = 264); 27.5% had a left ventricular ejection fraction of <40%; 63.2% had a PCI for a myocardial infarction; 52.5% (n = 191) received a positive HADS score and 28.0% (n = 102) had a positive SCID diagnosis. Twelve patients had a positive HADS score but refused further testing (SCID‐I). The following SCID diagnoses were found: unipolar depression (n = 59, 16.5%), bipolar disorder (n = 2, 0.6%), anxiety disorder (n = 26, 7.2%), substance use/addiction disorder (n = 14, 4.0%), adjustment disorder (n = 11, 3.0%) and posttraumatic stress disorder (n = 4, 1.1%). Table 1 provides an overview of the demographic and clinical characteristics. For more details on the ICD‐10 diagnoses, see Table A1.\nSociodemographic and clinical characteristics of the participants\nAbbreviations: MHI: mental health issue; NYHA; New York Heart Association; PCI, percutaneous coronary intervention.\nIt is possible for patients to have more than one answer.\nAt least one cell was too small for the appropriate analysis.", "A total of 80.8% with MHI and 74.2% without MHI found it appropriate to have been approached by the doctor (e.g., GP or cardiologist) about MHI; several patients were unsure whether they found it appropriate (5.8% with MHI; 7.5% without MHI). Patients expressed the general wish to be approached proactively by their doctor about MHI (46.5% with MHI; 38.0% without MHI). Approximately a fifth of patients in both groups were unsure about whether they wished to be approached by the doctor about MHI. About 23.4% of MHI patients mentioned a preference for GPs to help them find adequate treatment for drug therapy. For detailed information, see Table 2.\nDetails to access, barriers and needs regarding healthcare in CHD patients with and without mental health issues (MHI)\nAbbreviation: CHD, coronary heart disease.\nMultiple‐choice question.\nAll percentages relate to the maximum number of patients who were eligible for the underlying question.\nAt least one cell was too small for the appropriate analysis.", "A total of 30 CHD patients with MHI (42.3%) stated that they received an adequate amount of information on the content and accessibility of psychological care. Only a minority of patients obtained information about psychotherapy (7.0%) with a quarter receiving no information at all. About 30% of patients obtained information on the content and accessibility of psychological care through their doctors, while 35% stated that they did not receive further information. When patients did obtain information, half of all patients with MHI got information from the GP or other persons outside of the healthcare system (e.g., family members, friends); 47.8% of MHI patients and 44.1% of patients without MHI wished to get this specific information from the GP. For further information, see Table 2.", "In total, 65 CHD patients agreed to participate in a qualitative interview. These patients were mostly men (n = 53) and had an ejection fraction of <40% (n = 41). Divided over the three settings, 21.5% of patients came from hospitals, 33.9% from rehabilitation clinics and 44.6% from cardiology practices. From this subpopulation, 20 patients were selected and asked to participate. The mean age was M = 67.2 (SD = 12.6). All patients lived with a partner. For detailed information about the selection process, see Figure 1.", "The following section contains information about how the patients experienced the cardiac event and whether they were really approached by the doctor to talk about MHI. Patients experienced CHD as a decisive turning point that unsettled them permanently. Most respondents felt significant anxiety and uncertainty in the weeks following the cardiac event. After diagnosis, spouses and family were the central persons to whom patients articulated their fears. Only a few patients reported that they were approached about MHI by a doctor as part of the CHD diagnosis. In general, MHI was not discussed until months after the diagnosis. Few patients engaged in a conversation about MHI with a professional. They reported that either the GP or a psychologist in a rehabilitation clinic approached them.No, not really. No one actually asked, no doctor, whether there was still a need for treatment. Patient 16, l. 22–24Of course, the doctor should have to ask more background information. How the patient is doing in other areas after this disease, whether he has psychological issues. I think that most specialists, e.g., cardiologists, proceed very technocratically and reel off their plan and don't care much about other things. I have not met any cardiologist, except for the head physician at the university hospital, who has dealt more intensively with this issue. For them it is only important how the echography is, how the ultrasound examination is, the blood values, etc. That's it. I even believe that cardiologists don't have a heart (laughs). Patient 9, l. 78–85Well, I have the feeling that my cardiologist doesn't get involved with me on an emotional level. It's relatively quick, he does a ECG or an ultrasound or something else and then I get my information and have to leave. It's not something where you have a longer conversation, that's what they do. only the treatment, then you go back to the treating GP. Patient 14, l. 131–137\n\nNo, not really. No one actually asked, no doctor, whether there was still a need for treatment. Patient 16, l. 22–24\nOf course, the doctor should have to ask more background information. How the patient is doing in other areas after this disease, whether he has psychological issues. I think that most specialists, e.g., cardiologists, proceed very technocratically and reel off their plan and don't care much about other things. I have not met any cardiologist, except for the head physician at the university hospital, who has dealt more intensively with this issue. For them it is only important how the echography is, how the ultrasound examination is, the blood values, etc. That's it. I even believe that cardiologists don't have a heart (laughs). Patient 9, l. 78–85\nWell, I have the feeling that my cardiologist doesn't get involved with me on an emotional level. It's relatively quick, he does a ECG or an ultrasound or something else and then I get my information and have to leave. It's not something where you have a longer conversation, that's what they do. only the treatment, then you go back to the treating GP. Patient 14, l. 131–137", "[SUBTITLE] The wish for a conversation about MHI [SUBSECTION] All patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n\nI think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70\nIt has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222\nBut then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\nAll patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n\nI think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70\nIt has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222\nBut then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n[SUBTITLE] No wish for a conversation about MHI [SUBSECTION] A minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n\nNo. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\nA minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n\nNo. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n[SUBTITLE] Access and barriers [SUBSECTION] Limited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n\nThe rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200\nWell, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\nLimited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n\nThe rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200\nWell, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n[SUBTITLE] Specific age‐related issues [SUBSECTION] Besides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’.\nBesides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’.\n[SUBTITLE] The younger patient group and their concerns about their ability to work [SUBSECTION] The needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n\nWell, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51\nI don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\nThe needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n\nWell, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51\nI don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n[SUBTITLE] The older patient group and their concerns about successful aging [SUBSECTION] On the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\n\n‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\nOn the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\n\n‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144", "All patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n\nI think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70\nIt has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222\nBut then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210", "A minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n\nNo. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72", "Limited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n\nThe rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200\nWell, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96", "Besides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’.", "The needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n\nWell, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51\nI don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50", "On the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\n\n‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144", "Both quantitative and qualitative data show in general that for all recruitment centres (hospital cardiac departments, cardiology practices and rehabilitation clinics) patients with and without MHI experience anxiety and uncertainty after a cardiac event. This event was perceived as drastic and life‐changing, and consequently, patients felt a great need to talk about what had happened. They expressed disappointment about the fact that doctors rarely address MHI. Interviewed patients stated that they show little initiative during their medical consultation and usually did not address MHI on their own. A study from Lussier et al.\n11\n developed an online tool to encourage patients to be more active and prepared for their healthcare encounters by providing skills coaching via a website. Although patients reported a positive perception of such coaching and a higher motivational level, barriers such as lack of interest, limited access to technology, lack of time or language skills reduced the success of this approach.\n11\n\n\nHowever, when MHI was discussed with the doctor, patients did report a decrease in symptoms, mainly a reduction in anxiety, and a positive influence on their feeling of being able to cope with the cardiac event. It therefore seems reasonable that doctors, for their part, ask more regularly about MHI during medical consultations to improve the previous deficit in the area of emotional care. For this purpose, it seems necessary to have targeted interventions, such as creating greater awareness for this topic and the vulnerable patient group in everyday medical practice, to ensure rapid detection of MHI and thus adequate care. Further training courses, in the field of basic psychosomatic care and medical interviewing, could be helpful to increase individual communication skills in doctor–patient conversations, especially in an empathic manner during medical conversations. Studies showed that empathy of the doctor perceived by the patient significantly improved patient satisfaction with the received medical care, especially in terms of information exchange, treatment, perceived expertize of the doctor, as well as interpersonal trust and partnership with the doctor.\n31\n A higher empathy‐related behaviour of the doctor might be useful to improve the doctor–patient relationship and might increase the chance of patients daring to speak about MHI to their doctor.", "In general, patients received little support in seeking psychological, psychiatric or psychotherapeutic help. Lack of time and personal opportunities, none to little help in coping with mental stress, and finding an ambulant psychotherapist were found to be barriers for patients and GPs in dealing with their CHD‐related MHI's.\nShortcomings in the early stages of CHD and MHI‐related treatment can lead to demoralization and learned helplessness in patients. At the same time, complaints such as future‐ and health‐related worries, anxiety, depression and fear of incapacity for work continue to exist and further restrict patients' mental and physical health. As mentioned before, MHI and even mental stress are associated with higher CHD‐related morbidity and mortality\n7\n and hinder the required lifestyle changes to reduce CHD risk factors.\n4\n, \n8\n\n\nThese shortcomings in medical healthcare can occur because the current healthcare system defines ‘valuable care’ as health outcomes relative to costs and hence encompasses efficiency. However, cost reduction without regard to the achieved outcomes leads to limited effective healthcare.\n32\n Following Porter,\n33\n\nvalue‐based healthcare has the following definition: value equals health outcomes that matter to patients regarding the costs of delivering the outcomes. Thus, value measures all services and activities that determine success by meeting the patients' needs. These needs are determined by the medical condition of the patient and are an interrelated set of medical circumstances. A medical condition includes the most commonly associated conditions—meaning that, for example, care for CHD patients must consist of care for hypertension, and so forth.\n32\n, \n33\n Regarding the high prevalence of MHI in CHD patients, it might be advisable to include MHI as further interrelated medical circumstances. For example, only CHD patients in rehabilitation clinics reported having access to psychotherapeutic help in general; patients from practices and hospitals lacked this possibility. Rehabilitation clinics seem to work more within a more value‐based healthcare cycle because they are medical units that specialize in several diseases and thus are organized in the same way as an integrated practice unit,\n33\n thus including all the necessary skilled medical practitioners needed for medical conditions and their co‐occurring problems.\nTo deal with the shortcomings found in this study, a more value‐based and patient‐centred approach is necessary. With regard to structural requirements, earlier detection and correct diagnosis of MHI, as well as appropriate and fast treatment, would be examples (i.e., an appointment with an outpatient psychotherapist). In addition, more psychological measures, such as training for doctors (e.g., to increase the level of empathy and better support in coping with CHD and MHI) and improvements in procedural requirements (more conversation time during doctor contacts and more precise agreements between different departments) are needed. To be able to implement such requirements in practice adequately, the deployment of nursing or case managers in GP practices would be advisable. Nursing managers could use low‐threshold interventions to assist CHD patients with MHI in the search for a suitable form of psychotherapy and/or drug therapy and to accompany their implementation. They could act as a link between the various care providers and ensure a timely exchange of information and early personal appointments.\n34\n\n", "MenDis‐CHD provided insights into the needs of CHD patients with and without MHI and their access and barriers to healthcare, clarified important health outcomes for this population and indicated the requirements that a more value‐based care system should focus on to increase quality and efficiency.\nNonetheless, this study has some limitations. With regard to the qualitative part of the study, we chose to conduct 20 interviews. Looking at the pool of eligible patients, our sample size is limited by the participant diversity and the recruitment location (e.g., recruitment only in one city in Germany and particularly in cardiology practices). Because patients were mainly recruited in outpatient settings, it is possible that they were biased with regard to their preference to talk to a GP about their MHI. Patients may have perceived the outpatient setting as a familiar environment, as there has not necessarily been an inpatient stay recently and could therefore adopt a one‐sided view. Another limitation was that some of the quantitative items in our questionnaire were self‐developed and had no statistical testing of validation. Replication with a larger sample size and greater diversity, either across Germany or internationally, would be advantageous to explore possible generalization effects. Other vulnerable population groups that also have a high prevalence of MHI (e.g., schizophrenia) should be examined to explore specific health outcomes and needs that are important to the particular group of patients. With more data, researchers could build a better basis in clinical practice to facilitate a change towards a value‐based healthcare system.", "Samia Peltzer wrote the abstract, introduction, method section, results (incl. analyses) and discussion, designed the figure and tables, references with EndNote, made an overall adaption of the text and included references. Samia Peltzer was a major contributor in writing the manuscript. Ursula Köstler conducted the information for the qualitative analysis, conducted qualitative analysis and summed up the results for the qualitative part. Frank Jessen, Frank Schulz‐Nieswandt and Christian Albus designed and conducted the study as chief scientists. Hendrik Müller, Nadine Scholten, Frank Jessen and Christian Albus critically revised the manuscript. All gave final approval and agreed to be accountable for all aspects of work, ensuring integrity and accuracy." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "INTRODUCTION", "Coronary heart disease", "Mental health issues and their relationships with CHD", "MHIs as barrier to medical adherence", "Current study", "MATERIALS AND METHODS", "Design", "Patient contribution", "Participants", "Research tools", "Quantitative", "Qualitative", "Procedures", "Quantitative", "Qualitative", "Data analysis", "Quantitative", "Qualitative", "RESULTS", "Sociodemographic and clinical characteristics", "Patients' needs", "Access and barriers to healthcare", "Qualitative", "Perception of the cardiac event and approaches by doctors to talk about MHI", "Patients' needs", "The wish for a conversation about MHI", "No wish for a conversation about MHI", "Access and barriers", "Specific age‐related issues", "The younger patient group and their concerns about their ability to work", "The older patient group and their concerns about successful aging", "DISCUSSION", "Perception of the cardiac event and patients' needs", "Access and barriers to healthcare and specific issues of patient groups", "Strengths, limitations and further research", "CONCLUSION", "AUTHOR CONTRIBUTIONS" ]
[ "[SUBTITLE] Coronary heart disease [SUBSECTION] Coronary heart disease (CHD) is the leading cause of mortality, accounting for approximately one‐third of all deaths in individuals over the age of 35 years, with a death rate of 102.6 per 100,000.\n1\n In Germany, the lifetime prevalence of CHD for the age group 40−79 years is 9.3% (8.4−10.3).\n2\n Short‐term mortality has been decreased due to advances in acute CHD treatments. However, at a population level, CHD morbidity has increased in recent years. After an interim decline in smoking, hypercholesterolaemia and hypertension, these currently appear to be on the rise again.\n1\n, \n3\n, \n4\n\n\nCoronary heart disease (CHD) is the leading cause of mortality, accounting for approximately one‐third of all deaths in individuals over the age of 35 years, with a death rate of 102.6 per 100,000.\n1\n In Germany, the lifetime prevalence of CHD for the age group 40−79 years is 9.3% (8.4−10.3).\n2\n Short‐term mortality has been decreased due to advances in acute CHD treatments. However, at a population level, CHD morbidity has increased in recent years. After an interim decline in smoking, hypercholesterolaemia and hypertension, these currently appear to be on the rise again.\n1\n, \n3\n, \n4\n\n\n[SUBTITLE] Mental health issues and their relationships with CHD [SUBSECTION] It has been found that mental health issues (MHIs) are highly prevalent among patients with CHD. MHI is defined as a recognizable set of clinical symptoms and/or behavioural problems that correlate with individual distress and impairment of functioning at the individual level.\n5\n After a myocardial infarction, almost 30% of these patients suffer from depressive symptoms and 20% even fulfil ICD‐10 criteria for a depressive episode.\n6\n Depression is associated with higher CHD‐related morbidity and mortality (relative risk = 1.6–2.4): After a cardiac event such as a myocardial infarction, CHD patients with comorbid depression have twofold greater mortality during the next 2 years.\n7\n Furthermore, mortality risk can increase sixfold when the patient suffers from severe depressive symptoms.\n8\n\n\nIt has been found that mental health issues (MHIs) are highly prevalent among patients with CHD. MHI is defined as a recognizable set of clinical symptoms and/or behavioural problems that correlate with individual distress and impairment of functioning at the individual level.\n5\n After a myocardial infarction, almost 30% of these patients suffer from depressive symptoms and 20% even fulfil ICD‐10 criteria for a depressive episode.\n6\n Depression is associated with higher CHD‐related morbidity and mortality (relative risk = 1.6–2.4): After a cardiac event such as a myocardial infarction, CHD patients with comorbid depression have twofold greater mortality during the next 2 years.\n7\n Furthermore, mortality risk can increase sixfold when the patient suffers from severe depressive symptoms.\n8\n\n\n[SUBTITLE] MHIs as barrier to medical adherence [SUBSECTION] MHI also acts as a strong barrier to treatment adherence and exacerbates a required lifestyle change that is necessary to reduce CHD risk factors—regardless of whether MHI were already pre‐existing or occurred as a consequence of CHD.\n4\n For example, patients with MHI are less likely to exhibit healthy lifestyle behaviour (e.g., quit smoking) and have low medication adherence.\n9\n This emphasizes the drive for more patient involvement in terms of disease management and medical consultations.\n10\n At the same time, this group of patients finds it difficult to understand the course of the consultation and to present their own concerns adequately, resulting in a lack of communication of possible physical and/or MHIs to their doctor.\n11\n Hence, this specific population of CHD patients with MHI may be very vulnerable due to difficulties in disease management, low communication skills in a medical setting and problems communicating possible issues and questions about personal topics. This suggests that the quality of communication between patients and their doctors is important to secure the best course of treatment.\n12\n\n\nConsequently, adequate screening and treatment for MHI in CHD patients during the medical consultation from the doctor's perspective is urgently needed. National and international guidelines have recommended routine screening and treatment for MHI on primary and secondary CHD prevention.\n4\n, \n13\n However, studies have been able to show that this screening is not regularly followed due to lack of time and low occurrence of verbal interventions.\n14\n, \n15\n\n\nMHI also acts as a strong barrier to treatment adherence and exacerbates a required lifestyle change that is necessary to reduce CHD risk factors—regardless of whether MHI were already pre‐existing or occurred as a consequence of CHD.\n4\n For example, patients with MHI are less likely to exhibit healthy lifestyle behaviour (e.g., quit smoking) and have low medication adherence.\n9\n This emphasizes the drive for more patient involvement in terms of disease management and medical consultations.\n10\n At the same time, this group of patients finds it difficult to understand the course of the consultation and to present their own concerns adequately, resulting in a lack of communication of possible physical and/or MHIs to their doctor.\n11\n Hence, this specific population of CHD patients with MHI may be very vulnerable due to difficulties in disease management, low communication skills in a medical setting and problems communicating possible issues and questions about personal topics. This suggests that the quality of communication between patients and their doctors is important to secure the best course of treatment.\n12\n\n\nConsequently, adequate screening and treatment for MHI in CHD patients during the medical consultation from the doctor's perspective is urgently needed. National and international guidelines have recommended routine screening and treatment for MHI on primary and secondary CHD prevention.\n4\n, \n13\n However, studies have been able to show that this screening is not regularly followed due to lack of time and low occurrence of verbal interventions.\n14\n, \n15\n\n\n[SUBTITLE] Current study [SUBSECTION] In the current study, we explore the following research questions: (1) What are the MHI‐related needs of CHD patients with MHI after a cardiac event?, (2) how well do doctors meet their patients' needs? and (3) how do patients experience access and barriers to the healthcare system? It is hypothesized that patients with MHI have a desire for doctors to approach and support them with their MHI. Secondly, it is expected that doctors rarely meet their patients' needs. Lastly, it is hypothesized that, due to Hypotheses 1 and 2, patients receive a lack of access and distinct barriers to the healthcare system, especially related to their MHI. The aim of the study is to improve the quality of care and initiate changes in the structure of healthcare towards more patient‐centred care in the long term.\nIn the current study, we explore the following research questions: (1) What are the MHI‐related needs of CHD patients with MHI after a cardiac event?, (2) how well do doctors meet their patients' needs? and (3) how do patients experience access and barriers to the healthcare system? It is hypothesized that patients with MHI have a desire for doctors to approach and support them with their MHI. Secondly, it is expected that doctors rarely meet their patients' needs. Lastly, it is hypothesized that, due to Hypotheses 1 and 2, patients receive a lack of access and distinct barriers to the healthcare system, especially related to their MHI. The aim of the study is to improve the quality of care and initiate changes in the structure of healthcare towards more patient‐centred care in the long term.", "Coronary heart disease (CHD) is the leading cause of mortality, accounting for approximately one‐third of all deaths in individuals over the age of 35 years, with a death rate of 102.6 per 100,000.\n1\n In Germany, the lifetime prevalence of CHD for the age group 40−79 years is 9.3% (8.4−10.3).\n2\n Short‐term mortality has been decreased due to advances in acute CHD treatments. However, at a population level, CHD morbidity has increased in recent years. After an interim decline in smoking, hypercholesterolaemia and hypertension, these currently appear to be on the rise again.\n1\n, \n3\n, \n4\n\n", "It has been found that mental health issues (MHIs) are highly prevalent among patients with CHD. MHI is defined as a recognizable set of clinical symptoms and/or behavioural problems that correlate with individual distress and impairment of functioning at the individual level.\n5\n After a myocardial infarction, almost 30% of these patients suffer from depressive symptoms and 20% even fulfil ICD‐10 criteria for a depressive episode.\n6\n Depression is associated with higher CHD‐related morbidity and mortality (relative risk = 1.6–2.4): After a cardiac event such as a myocardial infarction, CHD patients with comorbid depression have twofold greater mortality during the next 2 years.\n7\n Furthermore, mortality risk can increase sixfold when the patient suffers from severe depressive symptoms.\n8\n\n", "MHI also acts as a strong barrier to treatment adherence and exacerbates a required lifestyle change that is necessary to reduce CHD risk factors—regardless of whether MHI were already pre‐existing or occurred as a consequence of CHD.\n4\n For example, patients with MHI are less likely to exhibit healthy lifestyle behaviour (e.g., quit smoking) and have low medication adherence.\n9\n This emphasizes the drive for more patient involvement in terms of disease management and medical consultations.\n10\n At the same time, this group of patients finds it difficult to understand the course of the consultation and to present their own concerns adequately, resulting in a lack of communication of possible physical and/or MHIs to their doctor.\n11\n Hence, this specific population of CHD patients with MHI may be very vulnerable due to difficulties in disease management, low communication skills in a medical setting and problems communicating possible issues and questions about personal topics. This suggests that the quality of communication between patients and their doctors is important to secure the best course of treatment.\n12\n\n\nConsequently, adequate screening and treatment for MHI in CHD patients during the medical consultation from the doctor's perspective is urgently needed. National and international guidelines have recommended routine screening and treatment for MHI on primary and secondary CHD prevention.\n4\n, \n13\n However, studies have been able to show that this screening is not regularly followed due to lack of time and low occurrence of verbal interventions.\n14\n, \n15\n\n", "In the current study, we explore the following research questions: (1) What are the MHI‐related needs of CHD patients with MHI after a cardiac event?, (2) how well do doctors meet their patients' needs? and (3) how do patients experience access and barriers to the healthcare system? It is hypothesized that patients with MHI have a desire for doctors to approach and support them with their MHI. Secondly, it is expected that doctors rarely meet their patients' needs. Lastly, it is hypothesized that, due to Hypotheses 1 and 2, patients receive a lack of access and distinct barriers to the healthcare system, especially related to their MHI. The aim of the study is to improve the quality of care and initiate changes in the structure of healthcare towards more patient‐centred care in the long term.", "This study (MenDis‐CHD) is one of three projects carried out within the framework of the Federal Ministry of Education and Research (BMBF)‐funded Cologne Research and Practice Network (CoRe‐Net). MenDis‐CHD was approved by the Ethics Commission of Cologne University's Faculty of Medicine (Committee Reference Number: 17–220) on 26 September 2017.\n[SUBTITLE] Design [SUBSECTION] MenDis‐CHD is a cross‐sectional study with a mixed‐method approach using an explanatory sequential design,\n16\n beginning with quantitative data collection (one questionnaire containing: questions about sociodemographic data, patients' needs and access and barriers to the healthcare system; psychological assessments [Hospital Anxiety and Depression Scale {HADS}, SCID‐I]), followed by a qualitative data acquisition (semi‐structured in‐depth interviews) to enrich the quantitative data. Specific details of the methods used in this study can be found in an earlier paper.\n17\n The questionnaire was fully developed in German. Individual translated items can be provided upon request.\nMenDis‐CHD is a cross‐sectional study with a mixed‐method approach using an explanatory sequential design,\n16\n beginning with quantitative data collection (one questionnaire containing: questions about sociodemographic data, patients' needs and access and barriers to the healthcare system; psychological assessments [Hospital Anxiety and Depression Scale {HADS}, SCID‐I]), followed by a qualitative data acquisition (semi‐structured in‐depth interviews) to enrich the quantitative data. Specific details of the methods used in this study can be found in an earlier paper.\n17\n The questionnaire was fully developed in German. Individual translated items can be provided upon request.\n[SUBTITLE] Patient contribution [SUBSECTION] We received input from CHD patients during the pretests of the questionnaire and the qualitative interviews, as well as from members of CHD self‐help groups. The patient perspective has been continuously taken into account in the project and the interests of the patients have also been considered with regard to the transfer of results (e.g., through public lectures, summaries in plain language and design of a leaflet to share with peers and distribute to patient groups). Patients did not receive any compensation for contributing to this study.\nWe received input from CHD patients during the pretests of the questionnaire and the qualitative interviews, as well as from members of CHD self‐help groups. The patient perspective has been continuously taken into account in the project and the interests of the patients have also been considered with regard to the transfer of results (e.g., through public lectures, summaries in plain language and design of a leaflet to share with peers and distribute to patient groups). Patients did not receive any compensation for contributing to this study.\n[SUBTITLE] Participants [SUBSECTION] As inclusion criteria, patients had to have an angiographically documented CHD with stable angina pectoris, acute coronary syndromes, percutaneous coronary intervention (PCI) or bypass surgery, be older than 18 years, have sufficient German language skills and be able to give informed consent. Severe physical (e.g., cancer) or unstable mental problems (e.g., acute suicidal ideation, delirium and moderate to severe dementia) were the exclusion criteria.\nOverall, 753 patients were screened for eligibility, of whom 374 were finally enroled in the study. Ten patients dropped out due to withdrawal of informed consent or incomplete questionnaires with missing answers. Therefore, 364 patients were included in the final data analysis. We recruited 107 patients in hospitals, 157 patients in rehabilitation clinics and 100 patients in cardiology practices. Figure 1 features a flowchart presenting the recruitment process.\nFlowchart of the recruitment procedure. CHD, coronary heart disease; LVEF, left ventricular ejection fraction; MHI, mental health issue\nAll 364 included patients filled in the questionnaire. We used maximum variation as the sampling strategy to increase diversity by age, gender, marital status, employed/retired status, severity of CHD, left ventricular ejection fraction and kind and severity of diagnosis of MHI.\n18\n By explicitly aiming for recruiting a diverse sample, one ensures the generalizability of one's findings across a broader part of the general population. Furthermore, we asked every eligible and interested patient during the recruitment phase to also participate in our in‐depth interviews. In total, 20 patients were selected to participate in the qualitative interviews. The diversity of participants was actively monitored, but due to a significant number of patients who did not want to participate in the interviews, participant diversity was not completely balanced, especially gender and ethnicity. In this study, the sample consisted of approximately 70% males. According to the current literature, males are significantly more likely to develop CHD,\n19\n with a lifetime prevalence of 9.9% (9.0–10.8) than women, with 6.7% (6.0–7.4).\n20\n Thus, this study seems to represent an appropriate sample population. For further details about the distribution of samplings for interview groups, see Figure 1.\nAs inclusion criteria, patients had to have an angiographically documented CHD with stable angina pectoris, acute coronary syndromes, percutaneous coronary intervention (PCI) or bypass surgery, be older than 18 years, have sufficient German language skills and be able to give informed consent. Severe physical (e.g., cancer) or unstable mental problems (e.g., acute suicidal ideation, delirium and moderate to severe dementia) were the exclusion criteria.\nOverall, 753 patients were screened for eligibility, of whom 374 were finally enroled in the study. Ten patients dropped out due to withdrawal of informed consent or incomplete questionnaires with missing answers. Therefore, 364 patients were included in the final data analysis. We recruited 107 patients in hospitals, 157 patients in rehabilitation clinics and 100 patients in cardiology practices. Figure 1 features a flowchart presenting the recruitment process.\nFlowchart of the recruitment procedure. CHD, coronary heart disease; LVEF, left ventricular ejection fraction; MHI, mental health issue\nAll 364 included patients filled in the questionnaire. We used maximum variation as the sampling strategy to increase diversity by age, gender, marital status, employed/retired status, severity of CHD, left ventricular ejection fraction and kind and severity of diagnosis of MHI.\n18\n By explicitly aiming for recruiting a diverse sample, one ensures the generalizability of one's findings across a broader part of the general population. Furthermore, we asked every eligible and interested patient during the recruitment phase to also participate in our in‐depth interviews. In total, 20 patients were selected to participate in the qualitative interviews. The diversity of participants was actively monitored, but due to a significant number of patients who did not want to participate in the interviews, participant diversity was not completely balanced, especially gender and ethnicity. In this study, the sample consisted of approximately 70% males. According to the current literature, males are significantly more likely to develop CHD,\n19\n with a lifetime prevalence of 9.9% (9.0–10.8) than women, with 6.7% (6.0–7.4).\n20\n Thus, this study seems to represent an appropriate sample population. For further details about the distribution of samplings for interview groups, see Figure 1.\n[SUBTITLE] Research tools [SUBSECTION] [SUBTITLE] Quantitative [SUBSECTION] Sociodemographic data were assessed via a newly developed questionnaire. Disease severity of CHD patients was assessed by screening for cardiac events in previous medical history (e.g., myocardial infarction), cardiac surgeries (e.g., bypass surgery), congestive heart failure, somatic comorbidity, pre‐existing presence of MHI, left ventricular ejection fraction and New York Heart Association stage (classification scheme of heart diseases according to their degree of severity). All of these characteristics were drawn from patients' medical records; they were not part of the questionnaire. Clinical data were measured with the use of the HADS and SCID‐I.\nAn aforementioned self‐made questionnaire was made of five subsections of which two are used in this paper, namely ‘Patients' needs’ and ‘Access and barriers to healthcare system’. The questionnaire was developed and used in the German language only; all items and section headers provided in this article are translated.\nBoth sections, ‘Patients' needs’ and ‘Access and barriers to the healthcare system,’ were assessed by a 20‐item self‐report questionnaire. Patients without MHI could skip several items that contained questions about an existing MHI. The section ‘Patient's needs’ contained six questions, such as ‘Do you wish to be approached by the doctor on possible mental health issues?’ with binary answer possibilities (Yes/No) or multiple‐choice questions such as ‘Who do you think should give a recommendation for treatment in the case of mental health issues?’. The section ‘Access and barriers to healthcare system’ contained 14 questions, also designed with ‘yes or no’ or multiple‐choice questions such as ‘How often have you sufficiently received information on the content and accessibility of psychological care?’. The questionnaire was developed within this study with the help of four CHD patients with and without MHI and has yet to be validated. More information on the content of the questionnaire and how it was created can be found in our first publication on MHI.\n21\n\n\nThe HADS\n22\n, \n23\n was used as a general screening tool to assess symptoms of depression and anxiety and was filled in by the patients. In line with the advice of several studies,\n24\n a score of 8 and above was considered a ‘positive’ test result for anxiety or depression. In this case, the Structured Clinical Interview for DSM‐IV Axis I Disorders (SCID‐I)\n25\n was conducted as a semi‐structured interview to assess mental symptoms and disorders as defined in the ICD‐10.\n6\n\n\nSociodemographic data were assessed via a newly developed questionnaire. Disease severity of CHD patients was assessed by screening for cardiac events in previous medical history (e.g., myocardial infarction), cardiac surgeries (e.g., bypass surgery), congestive heart failure, somatic comorbidity, pre‐existing presence of MHI, left ventricular ejection fraction and New York Heart Association stage (classification scheme of heart diseases according to their degree of severity). All of these characteristics were drawn from patients' medical records; they were not part of the questionnaire. Clinical data were measured with the use of the HADS and SCID‐I.\nAn aforementioned self‐made questionnaire was made of five subsections of which two are used in this paper, namely ‘Patients' needs’ and ‘Access and barriers to healthcare system’. The questionnaire was developed and used in the German language only; all items and section headers provided in this article are translated.\nBoth sections, ‘Patients' needs’ and ‘Access and barriers to the healthcare system,’ were assessed by a 20‐item self‐report questionnaire. Patients without MHI could skip several items that contained questions about an existing MHI. The section ‘Patient's needs’ contained six questions, such as ‘Do you wish to be approached by the doctor on possible mental health issues?’ with binary answer possibilities (Yes/No) or multiple‐choice questions such as ‘Who do you think should give a recommendation for treatment in the case of mental health issues?’. The section ‘Access and barriers to healthcare system’ contained 14 questions, also designed with ‘yes or no’ or multiple‐choice questions such as ‘How often have you sufficiently received information on the content and accessibility of psychological care?’. The questionnaire was developed within this study with the help of four CHD patients with and without MHI and has yet to be validated. More information on the content of the questionnaire and how it was created can be found in our first publication on MHI.\n21\n\n\nThe HADS\n22\n, \n23\n was used as a general screening tool to assess symptoms of depression and anxiety and was filled in by the patients. In line with the advice of several studies,\n24\n a score of 8 and above was considered a ‘positive’ test result for anxiety or depression. In this case, the Structured Clinical Interview for DSM‐IV Axis I Disorders (SCID‐I)\n25\n was conducted as a semi‐structured interview to assess mental symptoms and disorders as defined in the ICD‐10.\n6\n\n\n[SUBTITLE] Qualitative [SUBSECTION] The qualitative module was designed to build on the quantitative part in a second phase to enrich the quantitative findings and further investigate the research questions of the study.\n16\n The qualitative study design was conducted according to the method of the problem‐centred interview.\n26\n This is a semi‐structured in‐depth interview approach that allows the respondent to speak as freely as possible but is centred on (a) predefined problem(s) to which the interviewer repeatedly refers back during the conversation. Based on the research questions of this study, three main categories were developed: (1) Category ‘Perception of the cardiac event and approaches of doctors to talk about MHI’: Were CHD patients approached by the doctor on MHI and after a cardiac event? How did patients experience this approach? What were their expectations? What were positive and negative aspects of the contact with the doctor? Example questions were ‘Have you been approached by the doctor after a cardiac event and if so, how did you experience it? How long did it take until you were asked? Which aspects have been discussed during your meeting? What were your expectations beforehand and what are your expectations now?’; (2) Category ‘Patient's needs’: Do patients want to talk about MHI? Who do patients want to talk to about MHI? If a doctor is the first choice, what type of doctor? Example questions: ‘Who asked you about MHI? Who would you have liked to be addressed by? What did you need (both physically and mentally) after you received your diagnosis?’; (3) Category ‘Access and barriers’: How exactly do patients experience access and barriers to the health care system? Example questions: ‘What information and offers of help were accessible? How well could then be implemented? What information or offers did you receive, which did you not receive albeit your wish to receive them?’\nThe qualitative module was designed to build on the quantitative part in a second phase to enrich the quantitative findings and further investigate the research questions of the study.\n16\n The qualitative study design was conducted according to the method of the problem‐centred interview.\n26\n This is a semi‐structured in‐depth interview approach that allows the respondent to speak as freely as possible but is centred on (a) predefined problem(s) to which the interviewer repeatedly refers back during the conversation. Based on the research questions of this study, three main categories were developed: (1) Category ‘Perception of the cardiac event and approaches of doctors to talk about MHI’: Were CHD patients approached by the doctor on MHI and after a cardiac event? How did patients experience this approach? What were their expectations? What were positive and negative aspects of the contact with the doctor? Example questions were ‘Have you been approached by the doctor after a cardiac event and if so, how did you experience it? How long did it take until you were asked? Which aspects have been discussed during your meeting? What were your expectations beforehand and what are your expectations now?’; (2) Category ‘Patient's needs’: Do patients want to talk about MHI? Who do patients want to talk to about MHI? If a doctor is the first choice, what type of doctor? Example questions: ‘Who asked you about MHI? Who would you have liked to be addressed by? What did you need (both physically and mentally) after you received your diagnosis?’; (3) Category ‘Access and barriers’: How exactly do patients experience access and barriers to the health care system? Example questions: ‘What information and offers of help were accessible? How well could then be implemented? What information or offers did you receive, which did you not receive albeit your wish to receive them?’\n[SUBTITLE] Quantitative [SUBSECTION] Sociodemographic data were assessed via a newly developed questionnaire. Disease severity of CHD patients was assessed by screening for cardiac events in previous medical history (e.g., myocardial infarction), cardiac surgeries (e.g., bypass surgery), congestive heart failure, somatic comorbidity, pre‐existing presence of MHI, left ventricular ejection fraction and New York Heart Association stage (classification scheme of heart diseases according to their degree of severity). All of these characteristics were drawn from patients' medical records; they were not part of the questionnaire. Clinical data were measured with the use of the HADS and SCID‐I.\nAn aforementioned self‐made questionnaire was made of five subsections of which two are used in this paper, namely ‘Patients' needs’ and ‘Access and barriers to healthcare system’. The questionnaire was developed and used in the German language only; all items and section headers provided in this article are translated.\nBoth sections, ‘Patients' needs’ and ‘Access and barriers to the healthcare system,’ were assessed by a 20‐item self‐report questionnaire. Patients without MHI could skip several items that contained questions about an existing MHI. The section ‘Patient's needs’ contained six questions, such as ‘Do you wish to be approached by the doctor on possible mental health issues?’ with binary answer possibilities (Yes/No) or multiple‐choice questions such as ‘Who do you think should give a recommendation for treatment in the case of mental health issues?’. The section ‘Access and barriers to healthcare system’ contained 14 questions, also designed with ‘yes or no’ or multiple‐choice questions such as ‘How often have you sufficiently received information on the content and accessibility of psychological care?’. The questionnaire was developed within this study with the help of four CHD patients with and without MHI and has yet to be validated. More information on the content of the questionnaire and how it was created can be found in our first publication on MHI.\n21\n\n\nThe HADS\n22\n, \n23\n was used as a general screening tool to assess symptoms of depression and anxiety and was filled in by the patients. In line with the advice of several studies,\n24\n a score of 8 and above was considered a ‘positive’ test result for anxiety or depression. In this case, the Structured Clinical Interview for DSM‐IV Axis I Disorders (SCID‐I)\n25\n was conducted as a semi‐structured interview to assess mental symptoms and disorders as defined in the ICD‐10.\n6\n\n\nSociodemographic data were assessed via a newly developed questionnaire. Disease severity of CHD patients was assessed by screening for cardiac events in previous medical history (e.g., myocardial infarction), cardiac surgeries (e.g., bypass surgery), congestive heart failure, somatic comorbidity, pre‐existing presence of MHI, left ventricular ejection fraction and New York Heart Association stage (classification scheme of heart diseases according to their degree of severity). All of these characteristics were drawn from patients' medical records; they were not part of the questionnaire. Clinical data were measured with the use of the HADS and SCID‐I.\nAn aforementioned self‐made questionnaire was made of five subsections of which two are used in this paper, namely ‘Patients' needs’ and ‘Access and barriers to healthcare system’. The questionnaire was developed and used in the German language only; all items and section headers provided in this article are translated.\nBoth sections, ‘Patients' needs’ and ‘Access and barriers to the healthcare system,’ were assessed by a 20‐item self‐report questionnaire. Patients without MHI could skip several items that contained questions about an existing MHI. The section ‘Patient's needs’ contained six questions, such as ‘Do you wish to be approached by the doctor on possible mental health issues?’ with binary answer possibilities (Yes/No) or multiple‐choice questions such as ‘Who do you think should give a recommendation for treatment in the case of mental health issues?’. The section ‘Access and barriers to healthcare system’ contained 14 questions, also designed with ‘yes or no’ or multiple‐choice questions such as ‘How often have you sufficiently received information on the content and accessibility of psychological care?’. The questionnaire was developed within this study with the help of four CHD patients with and without MHI and has yet to be validated. More information on the content of the questionnaire and how it was created can be found in our first publication on MHI.\n21\n\n\nThe HADS\n22\n, \n23\n was used as a general screening tool to assess symptoms of depression and anxiety and was filled in by the patients. In line with the advice of several studies,\n24\n a score of 8 and above was considered a ‘positive’ test result for anxiety or depression. In this case, the Structured Clinical Interview for DSM‐IV Axis I Disorders (SCID‐I)\n25\n was conducted as a semi‐structured interview to assess mental symptoms and disorders as defined in the ICD‐10.\n6\n\n\n[SUBTITLE] Qualitative [SUBSECTION] The qualitative module was designed to build on the quantitative part in a second phase to enrich the quantitative findings and further investigate the research questions of the study.\n16\n The qualitative study design was conducted according to the method of the problem‐centred interview.\n26\n This is a semi‐structured in‐depth interview approach that allows the respondent to speak as freely as possible but is centred on (a) predefined problem(s) to which the interviewer repeatedly refers back during the conversation. Based on the research questions of this study, three main categories were developed: (1) Category ‘Perception of the cardiac event and approaches of doctors to talk about MHI’: Were CHD patients approached by the doctor on MHI and after a cardiac event? How did patients experience this approach? What were their expectations? What were positive and negative aspects of the contact with the doctor? Example questions were ‘Have you been approached by the doctor after a cardiac event and if so, how did you experience it? How long did it take until you were asked? Which aspects have been discussed during your meeting? What were your expectations beforehand and what are your expectations now?’; (2) Category ‘Patient's needs’: Do patients want to talk about MHI? Who do patients want to talk to about MHI? If a doctor is the first choice, what type of doctor? Example questions: ‘Who asked you about MHI? Who would you have liked to be addressed by? What did you need (both physically and mentally) after you received your diagnosis?’; (3) Category ‘Access and barriers’: How exactly do patients experience access and barriers to the health care system? Example questions: ‘What information and offers of help were accessible? How well could then be implemented? What information or offers did you receive, which did you not receive albeit your wish to receive them?’\nThe qualitative module was designed to build on the quantitative part in a second phase to enrich the quantitative findings and further investigate the research questions of the study.\n16\n The qualitative study design was conducted according to the method of the problem‐centred interview.\n26\n This is a semi‐structured in‐depth interview approach that allows the respondent to speak as freely as possible but is centred on (a) predefined problem(s) to which the interviewer repeatedly refers back during the conversation. Based on the research questions of this study, three main categories were developed: (1) Category ‘Perception of the cardiac event and approaches of doctors to talk about MHI’: Were CHD patients approached by the doctor on MHI and after a cardiac event? How did patients experience this approach? What were their expectations? What were positive and negative aspects of the contact with the doctor? Example questions were ‘Have you been approached by the doctor after a cardiac event and if so, how did you experience it? How long did it take until you were asked? Which aspects have been discussed during your meeting? What were your expectations beforehand and what are your expectations now?’; (2) Category ‘Patient's needs’: Do patients want to talk about MHI? Who do patients want to talk to about MHI? If a doctor is the first choice, what type of doctor? Example questions: ‘Who asked you about MHI? Who would you have liked to be addressed by? What did you need (both physically and mentally) after you received your diagnosis?’; (3) Category ‘Access and barriers’: How exactly do patients experience access and barriers to the health care system? Example questions: ‘What information and offers of help were accessible? How well could then be implemented? What information or offers did you receive, which did you not receive albeit your wish to receive them?’\n[SUBTITLE] Procedures [SUBSECTION] [SUBTITLE] Quantitative [SUBSECTION] The questionnaire was pre‐tested on two persons with CHD, one with MHI and one without MHI, in July 2017. Participants were recruited between 15 January 2018 and 29 March 2019 in two hospital cardiac departments, three cardiology practices and two rehabilitation clinics in Cologne, Germany, to achieve an appropriate realistic sample of routine healthcare settings. After screening for eligibility through the researchers, all patients' demographic data were documented in our screening log. Those who fulfilled our inclusion criteria and provided written informed consent received the quantitative questionnaires. A second appointment was arranged to perform the SCID‐I if the HADS screening was ‘positive’. All researchers of this study were trained in applying the SCID‐I and experienced in conducting it. In addition, patients were asked to participate in qualitative interviews.\nThe questionnaire was pre‐tested on two persons with CHD, one with MHI and one without MHI, in July 2017. Participants were recruited between 15 January 2018 and 29 March 2019 in two hospital cardiac departments, three cardiology practices and two rehabilitation clinics in Cologne, Germany, to achieve an appropriate realistic sample of routine healthcare settings. After screening for eligibility through the researchers, all patients' demographic data were documented in our screening log. Those who fulfilled our inclusion criteria and provided written informed consent received the quantitative questionnaires. A second appointment was arranged to perform the SCID‐I if the HADS screening was ‘positive’. All researchers of this study were trained in applying the SCID‐I and experienced in conducting it. In addition, patients were asked to participate in qualitative interviews.\n[SUBTITLE] Qualitative [SUBSECTION] The interview guideline was pretested by two CHD patients: one with MHI and the other without. Pretests were conducted as face‐to‐face interviews in September 2018. Out of 65 participants who agreed to take part in a qualitative interview, 20 were randomly selected. The actual 20 interviews were carried out as face‐to‐face interviews between October 2018 and March 2019, either during a home visit or at the setting where the respective patient was recruited. Informed consent was obtained during the quantitative phase, but the interviewer informed all patients again by reading the information sheet and informed consent to them to ensure that they fully understood each element. The interview took 1 h. All interviews were audio‐recorded and transcribed.\nThe interview guideline was pretested by two CHD patients: one with MHI and the other without. Pretests were conducted as face‐to‐face interviews in September 2018. Out of 65 participants who agreed to take part in a qualitative interview, 20 were randomly selected. The actual 20 interviews were carried out as face‐to‐face interviews between October 2018 and March 2019, either during a home visit or at the setting where the respective patient was recruited. Informed consent was obtained during the quantitative phase, but the interviewer informed all patients again by reading the information sheet and informed consent to them to ensure that they fully understood each element. The interview took 1 h. All interviews were audio‐recorded and transcribed.\n[SUBTITLE] Quantitative [SUBSECTION] The questionnaire was pre‐tested on two persons with CHD, one with MHI and one without MHI, in July 2017. Participants were recruited between 15 January 2018 and 29 March 2019 in two hospital cardiac departments, three cardiology practices and two rehabilitation clinics in Cologne, Germany, to achieve an appropriate realistic sample of routine healthcare settings. After screening for eligibility through the researchers, all patients' demographic data were documented in our screening log. Those who fulfilled our inclusion criteria and provided written informed consent received the quantitative questionnaires. A second appointment was arranged to perform the SCID‐I if the HADS screening was ‘positive’. All researchers of this study were trained in applying the SCID‐I and experienced in conducting it. In addition, patients were asked to participate in qualitative interviews.\nThe questionnaire was pre‐tested on two persons with CHD, one with MHI and one without MHI, in July 2017. Participants were recruited between 15 January 2018 and 29 March 2019 in two hospital cardiac departments, three cardiology practices and two rehabilitation clinics in Cologne, Germany, to achieve an appropriate realistic sample of routine healthcare settings. After screening for eligibility through the researchers, all patients' demographic data were documented in our screening log. Those who fulfilled our inclusion criteria and provided written informed consent received the quantitative questionnaires. A second appointment was arranged to perform the SCID‐I if the HADS screening was ‘positive’. All researchers of this study were trained in applying the SCID‐I and experienced in conducting it. In addition, patients were asked to participate in qualitative interviews.\n[SUBTITLE] Qualitative [SUBSECTION] The interview guideline was pretested by two CHD patients: one with MHI and the other without. Pretests were conducted as face‐to‐face interviews in September 2018. Out of 65 participants who agreed to take part in a qualitative interview, 20 were randomly selected. The actual 20 interviews were carried out as face‐to‐face interviews between October 2018 and March 2019, either during a home visit or at the setting where the respective patient was recruited. Informed consent was obtained during the quantitative phase, but the interviewer informed all patients again by reading the information sheet and informed consent to them to ensure that they fully understood each element. The interview took 1 h. All interviews were audio‐recorded and transcribed.\nThe interview guideline was pretested by two CHD patients: one with MHI and the other without. Pretests were conducted as face‐to‐face interviews in September 2018. Out of 65 participants who agreed to take part in a qualitative interview, 20 were randomly selected. The actual 20 interviews were carried out as face‐to‐face interviews between October 2018 and March 2019, either during a home visit or at the setting where the respective patient was recruited. Informed consent was obtained during the quantitative phase, but the interviewer informed all patients again by reading the information sheet and informed consent to them to ensure that they fully understood each element. The interview took 1 h. All interviews were audio‐recorded and transcribed.\n[SUBTITLE] Data analysis [SUBSECTION] [SUBTITLE] Quantitative [SUBSECTION] All presented data were analysed using IBM SPSS Statistics 22 software\n27\n and controlled for both missing and impossible values. Identified cases with either missing or implausible values were compared to their equivalent paper versions and corrected. Multivariate analyses of variance and χ\n2 tests were conducted, depending on the curvature of the distribution of the variables' scores.\nAll presented data were analysed using IBM SPSS Statistics 22 software\n27\n and controlled for both missing and impossible values. Identified cases with either missing or implausible values were compared to their equivalent paper versions and corrected. Multivariate analyses of variance and χ\n2 tests were conducted, depending on the curvature of the distribution of the variables' scores.\n[SUBTITLE] Qualitative [SUBSECTION] The accuracy of the transcripts was checked based on the audiotape samples by two of the authors (a social scientist and a clinical psychologist). Data were imported to F4 Analysis\n28\n for content analysis.\n29\n In F4 Analysis, the audiotapes of the interviews were automatically converted into text. Then, both researchers listened to the tapes and checked the transcripts independently. Relevant clusters (content‐wise) were developed and presented to the steering group of MenDis‐CHD for interpretation. Data and coding within the different clusters were further analysed, named and defined as: (1) perception of cardiac event; (2) patient's needs; (3) access and barriers to the healthcare system and (4) specific age‐related issues. These categories reflect topics most important to patients during the semi‐structured interview. They were identified through discussion until an agreement was reached. Besides creating an overview of relevant content per category, this procedure was chosen to verify that patients would indeed talk about the research questions, as it was the aim to gather more qualitative information on them. Regular in‐depth discussion meetings of the steering group of MenDis‐CHD increased the reliability of the research results.\nThe accuracy of the transcripts was checked based on the audiotape samples by two of the authors (a social scientist and a clinical psychologist). Data were imported to F4 Analysis\n28\n for content analysis.\n29\n In F4 Analysis, the audiotapes of the interviews were automatically converted into text. Then, both researchers listened to the tapes and checked the transcripts independently. Relevant clusters (content‐wise) were developed and presented to the steering group of MenDis‐CHD for interpretation. Data and coding within the different clusters were further analysed, named and defined as: (1) perception of cardiac event; (2) patient's needs; (3) access and barriers to the healthcare system and (4) specific age‐related issues. These categories reflect topics most important to patients during the semi‐structured interview. They were identified through discussion until an agreement was reached. Besides creating an overview of relevant content per category, this procedure was chosen to verify that patients would indeed talk about the research questions, as it was the aim to gather more qualitative information on them. Regular in‐depth discussion meetings of the steering group of MenDis‐CHD increased the reliability of the research results.\n[SUBTITLE] Quantitative [SUBSECTION] All presented data were analysed using IBM SPSS Statistics 22 software\n27\n and controlled for both missing and impossible values. Identified cases with either missing or implausible values were compared to their equivalent paper versions and corrected. Multivariate analyses of variance and χ\n2 tests were conducted, depending on the curvature of the distribution of the variables' scores.\nAll presented data were analysed using IBM SPSS Statistics 22 software\n27\n and controlled for both missing and impossible values. Identified cases with either missing or implausible values were compared to their equivalent paper versions and corrected. Multivariate analyses of variance and χ\n2 tests were conducted, depending on the curvature of the distribution of the variables' scores.\n[SUBTITLE] Qualitative [SUBSECTION] The accuracy of the transcripts was checked based on the audiotape samples by two of the authors (a social scientist and a clinical psychologist). Data were imported to F4 Analysis\n28\n for content analysis.\n29\n In F4 Analysis, the audiotapes of the interviews were automatically converted into text. Then, both researchers listened to the tapes and checked the transcripts independently. Relevant clusters (content‐wise) were developed and presented to the steering group of MenDis‐CHD for interpretation. Data and coding within the different clusters were further analysed, named and defined as: (1) perception of cardiac event; (2) patient's needs; (3) access and barriers to the healthcare system and (4) specific age‐related issues. These categories reflect topics most important to patients during the semi‐structured interview. They were identified through discussion until an agreement was reached. Besides creating an overview of relevant content per category, this procedure was chosen to verify that patients would indeed talk about the research questions, as it was the aim to gather more qualitative information on them. Regular in‐depth discussion meetings of the steering group of MenDis‐CHD increased the reliability of the research results.\nThe accuracy of the transcripts was checked based on the audiotape samples by two of the authors (a social scientist and a clinical psychologist). Data were imported to F4 Analysis\n28\n for content analysis.\n29\n In F4 Analysis, the audiotapes of the interviews were automatically converted into text. Then, both researchers listened to the tapes and checked the transcripts independently. Relevant clusters (content‐wise) were developed and presented to the steering group of MenDis‐CHD for interpretation. Data and coding within the different clusters were further analysed, named and defined as: (1) perception of cardiac event; (2) patient's needs; (3) access and barriers to the healthcare system and (4) specific age‐related issues. These categories reflect topics most important to patients during the semi‐structured interview. They were identified through discussion until an agreement was reached. Besides creating an overview of relevant content per category, this procedure was chosen to verify that patients would indeed talk about the research questions, as it was the aim to gather more qualitative information on them. Regular in‐depth discussion meetings of the steering group of MenDis‐CHD increased the reliability of the research results.", "MenDis‐CHD is a cross‐sectional study with a mixed‐method approach using an explanatory sequential design,\n16\n beginning with quantitative data collection (one questionnaire containing: questions about sociodemographic data, patients' needs and access and barriers to the healthcare system; psychological assessments [Hospital Anxiety and Depression Scale {HADS}, SCID‐I]), followed by a qualitative data acquisition (semi‐structured in‐depth interviews) to enrich the quantitative data. Specific details of the methods used in this study can be found in an earlier paper.\n17\n The questionnaire was fully developed in German. Individual translated items can be provided upon request.", "We received input from CHD patients during the pretests of the questionnaire and the qualitative interviews, as well as from members of CHD self‐help groups. The patient perspective has been continuously taken into account in the project and the interests of the patients have also been considered with regard to the transfer of results (e.g., through public lectures, summaries in plain language and design of a leaflet to share with peers and distribute to patient groups). Patients did not receive any compensation for contributing to this study.", "As inclusion criteria, patients had to have an angiographically documented CHD with stable angina pectoris, acute coronary syndromes, percutaneous coronary intervention (PCI) or bypass surgery, be older than 18 years, have sufficient German language skills and be able to give informed consent. Severe physical (e.g., cancer) or unstable mental problems (e.g., acute suicidal ideation, delirium and moderate to severe dementia) were the exclusion criteria.\nOverall, 753 patients were screened for eligibility, of whom 374 were finally enroled in the study. Ten patients dropped out due to withdrawal of informed consent or incomplete questionnaires with missing answers. Therefore, 364 patients were included in the final data analysis. We recruited 107 patients in hospitals, 157 patients in rehabilitation clinics and 100 patients in cardiology practices. Figure 1 features a flowchart presenting the recruitment process.\nFlowchart of the recruitment procedure. CHD, coronary heart disease; LVEF, left ventricular ejection fraction; MHI, mental health issue\nAll 364 included patients filled in the questionnaire. We used maximum variation as the sampling strategy to increase diversity by age, gender, marital status, employed/retired status, severity of CHD, left ventricular ejection fraction and kind and severity of diagnosis of MHI.\n18\n By explicitly aiming for recruiting a diverse sample, one ensures the generalizability of one's findings across a broader part of the general population. Furthermore, we asked every eligible and interested patient during the recruitment phase to also participate in our in‐depth interviews. In total, 20 patients were selected to participate in the qualitative interviews. The diversity of participants was actively monitored, but due to a significant number of patients who did not want to participate in the interviews, participant diversity was not completely balanced, especially gender and ethnicity. In this study, the sample consisted of approximately 70% males. According to the current literature, males are significantly more likely to develop CHD,\n19\n with a lifetime prevalence of 9.9% (9.0–10.8) than women, with 6.7% (6.0–7.4).\n20\n Thus, this study seems to represent an appropriate sample population. For further details about the distribution of samplings for interview groups, see Figure 1.", "[SUBTITLE] Quantitative [SUBSECTION] Sociodemographic data were assessed via a newly developed questionnaire. Disease severity of CHD patients was assessed by screening for cardiac events in previous medical history (e.g., myocardial infarction), cardiac surgeries (e.g., bypass surgery), congestive heart failure, somatic comorbidity, pre‐existing presence of MHI, left ventricular ejection fraction and New York Heart Association stage (classification scheme of heart diseases according to their degree of severity). All of these characteristics were drawn from patients' medical records; they were not part of the questionnaire. Clinical data were measured with the use of the HADS and SCID‐I.\nAn aforementioned self‐made questionnaire was made of five subsections of which two are used in this paper, namely ‘Patients' needs’ and ‘Access and barriers to healthcare system’. The questionnaire was developed and used in the German language only; all items and section headers provided in this article are translated.\nBoth sections, ‘Patients' needs’ and ‘Access and barriers to the healthcare system,’ were assessed by a 20‐item self‐report questionnaire. Patients without MHI could skip several items that contained questions about an existing MHI. The section ‘Patient's needs’ contained six questions, such as ‘Do you wish to be approached by the doctor on possible mental health issues?’ with binary answer possibilities (Yes/No) or multiple‐choice questions such as ‘Who do you think should give a recommendation for treatment in the case of mental health issues?’. The section ‘Access and barriers to healthcare system’ contained 14 questions, also designed with ‘yes or no’ or multiple‐choice questions such as ‘How often have you sufficiently received information on the content and accessibility of psychological care?’. The questionnaire was developed within this study with the help of four CHD patients with and without MHI and has yet to be validated. More information on the content of the questionnaire and how it was created can be found in our first publication on MHI.\n21\n\n\nThe HADS\n22\n, \n23\n was used as a general screening tool to assess symptoms of depression and anxiety and was filled in by the patients. In line with the advice of several studies,\n24\n a score of 8 and above was considered a ‘positive’ test result for anxiety or depression. In this case, the Structured Clinical Interview for DSM‐IV Axis I Disorders (SCID‐I)\n25\n was conducted as a semi‐structured interview to assess mental symptoms and disorders as defined in the ICD‐10.\n6\n\n\nSociodemographic data were assessed via a newly developed questionnaire. Disease severity of CHD patients was assessed by screening for cardiac events in previous medical history (e.g., myocardial infarction), cardiac surgeries (e.g., bypass surgery), congestive heart failure, somatic comorbidity, pre‐existing presence of MHI, left ventricular ejection fraction and New York Heart Association stage (classification scheme of heart diseases according to their degree of severity). All of these characteristics were drawn from patients' medical records; they were not part of the questionnaire. Clinical data were measured with the use of the HADS and SCID‐I.\nAn aforementioned self‐made questionnaire was made of five subsections of which two are used in this paper, namely ‘Patients' needs’ and ‘Access and barriers to healthcare system’. The questionnaire was developed and used in the German language only; all items and section headers provided in this article are translated.\nBoth sections, ‘Patients' needs’ and ‘Access and barriers to the healthcare system,’ were assessed by a 20‐item self‐report questionnaire. Patients without MHI could skip several items that contained questions about an existing MHI. The section ‘Patient's needs’ contained six questions, such as ‘Do you wish to be approached by the doctor on possible mental health issues?’ with binary answer possibilities (Yes/No) or multiple‐choice questions such as ‘Who do you think should give a recommendation for treatment in the case of mental health issues?’. The section ‘Access and barriers to healthcare system’ contained 14 questions, also designed with ‘yes or no’ or multiple‐choice questions such as ‘How often have you sufficiently received information on the content and accessibility of psychological care?’. The questionnaire was developed within this study with the help of four CHD patients with and without MHI and has yet to be validated. More information on the content of the questionnaire and how it was created can be found in our first publication on MHI.\n21\n\n\nThe HADS\n22\n, \n23\n was used as a general screening tool to assess symptoms of depression and anxiety and was filled in by the patients. In line with the advice of several studies,\n24\n a score of 8 and above was considered a ‘positive’ test result for anxiety or depression. In this case, the Structured Clinical Interview for DSM‐IV Axis I Disorders (SCID‐I)\n25\n was conducted as a semi‐structured interview to assess mental symptoms and disorders as defined in the ICD‐10.\n6\n\n\n[SUBTITLE] Qualitative [SUBSECTION] The qualitative module was designed to build on the quantitative part in a second phase to enrich the quantitative findings and further investigate the research questions of the study.\n16\n The qualitative study design was conducted according to the method of the problem‐centred interview.\n26\n This is a semi‐structured in‐depth interview approach that allows the respondent to speak as freely as possible but is centred on (a) predefined problem(s) to which the interviewer repeatedly refers back during the conversation. Based on the research questions of this study, three main categories were developed: (1) Category ‘Perception of the cardiac event and approaches of doctors to talk about MHI’: Were CHD patients approached by the doctor on MHI and after a cardiac event? How did patients experience this approach? What were their expectations? What were positive and negative aspects of the contact with the doctor? Example questions were ‘Have you been approached by the doctor after a cardiac event and if so, how did you experience it? How long did it take until you were asked? Which aspects have been discussed during your meeting? What were your expectations beforehand and what are your expectations now?’; (2) Category ‘Patient's needs’: Do patients want to talk about MHI? Who do patients want to talk to about MHI? If a doctor is the first choice, what type of doctor? Example questions: ‘Who asked you about MHI? Who would you have liked to be addressed by? What did you need (both physically and mentally) after you received your diagnosis?’; (3) Category ‘Access and barriers’: How exactly do patients experience access and barriers to the health care system? Example questions: ‘What information and offers of help were accessible? How well could then be implemented? What information or offers did you receive, which did you not receive albeit your wish to receive them?’\nThe qualitative module was designed to build on the quantitative part in a second phase to enrich the quantitative findings and further investigate the research questions of the study.\n16\n The qualitative study design was conducted according to the method of the problem‐centred interview.\n26\n This is a semi‐structured in‐depth interview approach that allows the respondent to speak as freely as possible but is centred on (a) predefined problem(s) to which the interviewer repeatedly refers back during the conversation. Based on the research questions of this study, three main categories were developed: (1) Category ‘Perception of the cardiac event and approaches of doctors to talk about MHI’: Were CHD patients approached by the doctor on MHI and after a cardiac event? How did patients experience this approach? What were their expectations? What were positive and negative aspects of the contact with the doctor? Example questions were ‘Have you been approached by the doctor after a cardiac event and if so, how did you experience it? How long did it take until you were asked? Which aspects have been discussed during your meeting? What were your expectations beforehand and what are your expectations now?’; (2) Category ‘Patient's needs’: Do patients want to talk about MHI? Who do patients want to talk to about MHI? If a doctor is the first choice, what type of doctor? Example questions: ‘Who asked you about MHI? Who would you have liked to be addressed by? What did you need (both physically and mentally) after you received your diagnosis?’; (3) Category ‘Access and barriers’: How exactly do patients experience access and barriers to the health care system? Example questions: ‘What information and offers of help were accessible? How well could then be implemented? What information or offers did you receive, which did you not receive albeit your wish to receive them?’", "Sociodemographic data were assessed via a newly developed questionnaire. Disease severity of CHD patients was assessed by screening for cardiac events in previous medical history (e.g., myocardial infarction), cardiac surgeries (e.g., bypass surgery), congestive heart failure, somatic comorbidity, pre‐existing presence of MHI, left ventricular ejection fraction and New York Heart Association stage (classification scheme of heart diseases according to their degree of severity). All of these characteristics were drawn from patients' medical records; they were not part of the questionnaire. Clinical data were measured with the use of the HADS and SCID‐I.\nAn aforementioned self‐made questionnaire was made of five subsections of which two are used in this paper, namely ‘Patients' needs’ and ‘Access and barriers to healthcare system’. The questionnaire was developed and used in the German language only; all items and section headers provided in this article are translated.\nBoth sections, ‘Patients' needs’ and ‘Access and barriers to the healthcare system,’ were assessed by a 20‐item self‐report questionnaire. Patients without MHI could skip several items that contained questions about an existing MHI. The section ‘Patient's needs’ contained six questions, such as ‘Do you wish to be approached by the doctor on possible mental health issues?’ with binary answer possibilities (Yes/No) or multiple‐choice questions such as ‘Who do you think should give a recommendation for treatment in the case of mental health issues?’. The section ‘Access and barriers to healthcare system’ contained 14 questions, also designed with ‘yes or no’ or multiple‐choice questions such as ‘How often have you sufficiently received information on the content and accessibility of psychological care?’. The questionnaire was developed within this study with the help of four CHD patients with and without MHI and has yet to be validated. More information on the content of the questionnaire and how it was created can be found in our first publication on MHI.\n21\n\n\nThe HADS\n22\n, \n23\n was used as a general screening tool to assess symptoms of depression and anxiety and was filled in by the patients. In line with the advice of several studies,\n24\n a score of 8 and above was considered a ‘positive’ test result for anxiety or depression. In this case, the Structured Clinical Interview for DSM‐IV Axis I Disorders (SCID‐I)\n25\n was conducted as a semi‐structured interview to assess mental symptoms and disorders as defined in the ICD‐10.\n6\n\n", "The qualitative module was designed to build on the quantitative part in a second phase to enrich the quantitative findings and further investigate the research questions of the study.\n16\n The qualitative study design was conducted according to the method of the problem‐centred interview.\n26\n This is a semi‐structured in‐depth interview approach that allows the respondent to speak as freely as possible but is centred on (a) predefined problem(s) to which the interviewer repeatedly refers back during the conversation. Based on the research questions of this study, three main categories were developed: (1) Category ‘Perception of the cardiac event and approaches of doctors to talk about MHI’: Were CHD patients approached by the doctor on MHI and after a cardiac event? How did patients experience this approach? What were their expectations? What were positive and negative aspects of the contact with the doctor? Example questions were ‘Have you been approached by the doctor after a cardiac event and if so, how did you experience it? How long did it take until you were asked? Which aspects have been discussed during your meeting? What were your expectations beforehand and what are your expectations now?’; (2) Category ‘Patient's needs’: Do patients want to talk about MHI? Who do patients want to talk to about MHI? If a doctor is the first choice, what type of doctor? Example questions: ‘Who asked you about MHI? Who would you have liked to be addressed by? What did you need (both physically and mentally) after you received your diagnosis?’; (3) Category ‘Access and barriers’: How exactly do patients experience access and barriers to the health care system? Example questions: ‘What information and offers of help were accessible? How well could then be implemented? What information or offers did you receive, which did you not receive albeit your wish to receive them?’", "[SUBTITLE] Quantitative [SUBSECTION] The questionnaire was pre‐tested on two persons with CHD, one with MHI and one without MHI, in July 2017. Participants were recruited between 15 January 2018 and 29 March 2019 in two hospital cardiac departments, three cardiology practices and two rehabilitation clinics in Cologne, Germany, to achieve an appropriate realistic sample of routine healthcare settings. After screening for eligibility through the researchers, all patients' demographic data were documented in our screening log. Those who fulfilled our inclusion criteria and provided written informed consent received the quantitative questionnaires. A second appointment was arranged to perform the SCID‐I if the HADS screening was ‘positive’. All researchers of this study were trained in applying the SCID‐I and experienced in conducting it. In addition, patients were asked to participate in qualitative interviews.\nThe questionnaire was pre‐tested on two persons with CHD, one with MHI and one without MHI, in July 2017. Participants were recruited between 15 January 2018 and 29 March 2019 in two hospital cardiac departments, three cardiology practices and two rehabilitation clinics in Cologne, Germany, to achieve an appropriate realistic sample of routine healthcare settings. After screening for eligibility through the researchers, all patients' demographic data were documented in our screening log. Those who fulfilled our inclusion criteria and provided written informed consent received the quantitative questionnaires. A second appointment was arranged to perform the SCID‐I if the HADS screening was ‘positive’. All researchers of this study were trained in applying the SCID‐I and experienced in conducting it. In addition, patients were asked to participate in qualitative interviews.\n[SUBTITLE] Qualitative [SUBSECTION] The interview guideline was pretested by two CHD patients: one with MHI and the other without. Pretests were conducted as face‐to‐face interviews in September 2018. Out of 65 participants who agreed to take part in a qualitative interview, 20 were randomly selected. The actual 20 interviews were carried out as face‐to‐face interviews between October 2018 and March 2019, either during a home visit or at the setting where the respective patient was recruited. Informed consent was obtained during the quantitative phase, but the interviewer informed all patients again by reading the information sheet and informed consent to them to ensure that they fully understood each element. The interview took 1 h. All interviews were audio‐recorded and transcribed.\nThe interview guideline was pretested by two CHD patients: one with MHI and the other without. Pretests were conducted as face‐to‐face interviews in September 2018. Out of 65 participants who agreed to take part in a qualitative interview, 20 were randomly selected. The actual 20 interviews were carried out as face‐to‐face interviews between October 2018 and March 2019, either during a home visit or at the setting where the respective patient was recruited. Informed consent was obtained during the quantitative phase, but the interviewer informed all patients again by reading the information sheet and informed consent to them to ensure that they fully understood each element. The interview took 1 h. All interviews were audio‐recorded and transcribed.", "The questionnaire was pre‐tested on two persons with CHD, one with MHI and one without MHI, in July 2017. Participants were recruited between 15 January 2018 and 29 March 2019 in two hospital cardiac departments, three cardiology practices and two rehabilitation clinics in Cologne, Germany, to achieve an appropriate realistic sample of routine healthcare settings. After screening for eligibility through the researchers, all patients' demographic data were documented in our screening log. Those who fulfilled our inclusion criteria and provided written informed consent received the quantitative questionnaires. A second appointment was arranged to perform the SCID‐I if the HADS screening was ‘positive’. All researchers of this study were trained in applying the SCID‐I and experienced in conducting it. In addition, patients were asked to participate in qualitative interviews.", "The interview guideline was pretested by two CHD patients: one with MHI and the other without. Pretests were conducted as face‐to‐face interviews in September 2018. Out of 65 participants who agreed to take part in a qualitative interview, 20 were randomly selected. The actual 20 interviews were carried out as face‐to‐face interviews between October 2018 and March 2019, either during a home visit or at the setting where the respective patient was recruited. Informed consent was obtained during the quantitative phase, but the interviewer informed all patients again by reading the information sheet and informed consent to them to ensure that they fully understood each element. The interview took 1 h. All interviews were audio‐recorded and transcribed.", "[SUBTITLE] Quantitative [SUBSECTION] All presented data were analysed using IBM SPSS Statistics 22 software\n27\n and controlled for both missing and impossible values. Identified cases with either missing or implausible values were compared to their equivalent paper versions and corrected. Multivariate analyses of variance and χ\n2 tests were conducted, depending on the curvature of the distribution of the variables' scores.\nAll presented data were analysed using IBM SPSS Statistics 22 software\n27\n and controlled for both missing and impossible values. Identified cases with either missing or implausible values were compared to their equivalent paper versions and corrected. Multivariate analyses of variance and χ\n2 tests were conducted, depending on the curvature of the distribution of the variables' scores.\n[SUBTITLE] Qualitative [SUBSECTION] The accuracy of the transcripts was checked based on the audiotape samples by two of the authors (a social scientist and a clinical psychologist). Data were imported to F4 Analysis\n28\n for content analysis.\n29\n In F4 Analysis, the audiotapes of the interviews were automatically converted into text. Then, both researchers listened to the tapes and checked the transcripts independently. Relevant clusters (content‐wise) were developed and presented to the steering group of MenDis‐CHD for interpretation. Data and coding within the different clusters were further analysed, named and defined as: (1) perception of cardiac event; (2) patient's needs; (3) access and barriers to the healthcare system and (4) specific age‐related issues. These categories reflect topics most important to patients during the semi‐structured interview. They were identified through discussion until an agreement was reached. Besides creating an overview of relevant content per category, this procedure was chosen to verify that patients would indeed talk about the research questions, as it was the aim to gather more qualitative information on them. Regular in‐depth discussion meetings of the steering group of MenDis‐CHD increased the reliability of the research results.\nThe accuracy of the transcripts was checked based on the audiotape samples by two of the authors (a social scientist and a clinical psychologist). Data were imported to F4 Analysis\n28\n for content analysis.\n29\n In F4 Analysis, the audiotapes of the interviews were automatically converted into text. Then, both researchers listened to the tapes and checked the transcripts independently. Relevant clusters (content‐wise) were developed and presented to the steering group of MenDis‐CHD for interpretation. Data and coding within the different clusters were further analysed, named and defined as: (1) perception of cardiac event; (2) patient's needs; (3) access and barriers to the healthcare system and (4) specific age‐related issues. These categories reflect topics most important to patients during the semi‐structured interview. They were identified through discussion until an agreement was reached. Besides creating an overview of relevant content per category, this procedure was chosen to verify that patients would indeed talk about the research questions, as it was the aim to gather more qualitative information on them. Regular in‐depth discussion meetings of the steering group of MenDis‐CHD increased the reliability of the research results.", "All presented data were analysed using IBM SPSS Statistics 22 software\n27\n and controlled for both missing and impossible values. Identified cases with either missing or implausible values were compared to their equivalent paper versions and corrected. Multivariate analyses of variance and χ\n2 tests were conducted, depending on the curvature of the distribution of the variables' scores.", "The accuracy of the transcripts was checked based on the audiotape samples by two of the authors (a social scientist and a clinical psychologist). Data were imported to F4 Analysis\n28\n for content analysis.\n29\n In F4 Analysis, the audiotapes of the interviews were automatically converted into text. Then, both researchers listened to the tapes and checked the transcripts independently. Relevant clusters (content‐wise) were developed and presented to the steering group of MenDis‐CHD for interpretation. Data and coding within the different clusters were further analysed, named and defined as: (1) perception of cardiac event; (2) patient's needs; (3) access and barriers to the healthcare system and (4) specific age‐related issues. These categories reflect topics most important to patients during the semi‐structured interview. They were identified through discussion until an agreement was reached. Besides creating an overview of relevant content per category, this procedure was chosen to verify that patients would indeed talk about the research questions, as it was the aim to gather more qualitative information on them. Regular in‐depth discussion meetings of the steering group of MenDis‐CHD increased the reliability of the research results.", "[SUBTITLE] Sociodemographic and clinical characteristics [SUBSECTION] Most participants were male (n = 258, 70.9%). The mean age across genders was M\nage = 65.9 (SD = 11.4); 72.5% lived together with a partner (n = 264); 27.5% had a left ventricular ejection fraction of <40%; 63.2% had a PCI for a myocardial infarction; 52.5% (n = 191) received a positive HADS score and 28.0% (n = 102) had a positive SCID diagnosis. Twelve patients had a positive HADS score but refused further testing (SCID‐I). The following SCID diagnoses were found: unipolar depression (n = 59, 16.5%), bipolar disorder (n = 2, 0.6%), anxiety disorder (n = 26, 7.2%), substance use/addiction disorder (n = 14, 4.0%), adjustment disorder (n = 11, 3.0%) and posttraumatic stress disorder (n = 4, 1.1%). Table 1 provides an overview of the demographic and clinical characteristics. For more details on the ICD‐10 diagnoses, see Table A1.\nSociodemographic and clinical characteristics of the participants\nAbbreviations: MHI: mental health issue; NYHA; New York Heart Association; PCI, percutaneous coronary intervention.\nIt is possible for patients to have more than one answer.\nAt least one cell was too small for the appropriate analysis.\nMost participants were male (n = 258, 70.9%). The mean age across genders was M\nage = 65.9 (SD = 11.4); 72.5% lived together with a partner (n = 264); 27.5% had a left ventricular ejection fraction of <40%; 63.2% had a PCI for a myocardial infarction; 52.5% (n = 191) received a positive HADS score and 28.0% (n = 102) had a positive SCID diagnosis. Twelve patients had a positive HADS score but refused further testing (SCID‐I). The following SCID diagnoses were found: unipolar depression (n = 59, 16.5%), bipolar disorder (n = 2, 0.6%), anxiety disorder (n = 26, 7.2%), substance use/addiction disorder (n = 14, 4.0%), adjustment disorder (n = 11, 3.0%) and posttraumatic stress disorder (n = 4, 1.1%). Table 1 provides an overview of the demographic and clinical characteristics. For more details on the ICD‐10 diagnoses, see Table A1.\nSociodemographic and clinical characteristics of the participants\nAbbreviations: MHI: mental health issue; NYHA; New York Heart Association; PCI, percutaneous coronary intervention.\nIt is possible for patients to have more than one answer.\nAt least one cell was too small for the appropriate analysis.\n[SUBTITLE] Patients' needs [SUBSECTION] A total of 80.8% with MHI and 74.2% without MHI found it appropriate to have been approached by the doctor (e.g., GP or cardiologist) about MHI; several patients were unsure whether they found it appropriate (5.8% with MHI; 7.5% without MHI). Patients expressed the general wish to be approached proactively by their doctor about MHI (46.5% with MHI; 38.0% without MHI). Approximately a fifth of patients in both groups were unsure about whether they wished to be approached by the doctor about MHI. About 23.4% of MHI patients mentioned a preference for GPs to help them find adequate treatment for drug therapy. For detailed information, see Table 2.\nDetails to access, barriers and needs regarding healthcare in CHD patients with and without mental health issues (MHI)\nAbbreviation: CHD, coronary heart disease.\nMultiple‐choice question.\nAll percentages relate to the maximum number of patients who were eligible for the underlying question.\nAt least one cell was too small for the appropriate analysis.\nA total of 80.8% with MHI and 74.2% without MHI found it appropriate to have been approached by the doctor (e.g., GP or cardiologist) about MHI; several patients were unsure whether they found it appropriate (5.8% with MHI; 7.5% without MHI). Patients expressed the general wish to be approached proactively by their doctor about MHI (46.5% with MHI; 38.0% without MHI). Approximately a fifth of patients in both groups were unsure about whether they wished to be approached by the doctor about MHI. About 23.4% of MHI patients mentioned a preference for GPs to help them find adequate treatment for drug therapy. For detailed information, see Table 2.\nDetails to access, barriers and needs regarding healthcare in CHD patients with and without mental health issues (MHI)\nAbbreviation: CHD, coronary heart disease.\nMultiple‐choice question.\nAll percentages relate to the maximum number of patients who were eligible for the underlying question.\nAt least one cell was too small for the appropriate analysis.\n[SUBTITLE] Access and barriers to healthcare [SUBSECTION] A total of 30 CHD patients with MHI (42.3%) stated that they received an adequate amount of information on the content and accessibility of psychological care. Only a minority of patients obtained information about psychotherapy (7.0%) with a quarter receiving no information at all. About 30% of patients obtained information on the content and accessibility of psychological care through their doctors, while 35% stated that they did not receive further information. When patients did obtain information, half of all patients with MHI got information from the GP or other persons outside of the healthcare system (e.g., family members, friends); 47.8% of MHI patients and 44.1% of patients without MHI wished to get this specific information from the GP. For further information, see Table 2.\nA total of 30 CHD patients with MHI (42.3%) stated that they received an adequate amount of information on the content and accessibility of psychological care. Only a minority of patients obtained information about psychotherapy (7.0%) with a quarter receiving no information at all. About 30% of patients obtained information on the content and accessibility of psychological care through their doctors, while 35% stated that they did not receive further information. When patients did obtain information, half of all patients with MHI got information from the GP or other persons outside of the healthcare system (e.g., family members, friends); 47.8% of MHI patients and 44.1% of patients without MHI wished to get this specific information from the GP. For further information, see Table 2.\n[SUBTITLE] Qualitative [SUBSECTION] In total, 65 CHD patients agreed to participate in a qualitative interview. These patients were mostly men (n = 53) and had an ejection fraction of <40% (n = 41). Divided over the three settings, 21.5% of patients came from hospitals, 33.9% from rehabilitation clinics and 44.6% from cardiology practices. From this subpopulation, 20 patients were selected and asked to participate. The mean age was M = 67.2 (SD = 12.6). All patients lived with a partner. For detailed information about the selection process, see Figure 1.\nIn total, 65 CHD patients agreed to participate in a qualitative interview. These patients were mostly men (n = 53) and had an ejection fraction of <40% (n = 41). Divided over the three settings, 21.5% of patients came from hospitals, 33.9% from rehabilitation clinics and 44.6% from cardiology practices. From this subpopulation, 20 patients were selected and asked to participate. The mean age was M = 67.2 (SD = 12.6). All patients lived with a partner. For detailed information about the selection process, see Figure 1.\n[SUBTITLE] Perception of the cardiac event and approaches by doctors to talk about MHI [SUBSECTION] The following section contains information about how the patients experienced the cardiac event and whether they were really approached by the doctor to talk about MHI. Patients experienced CHD as a decisive turning point that unsettled them permanently. Most respondents felt significant anxiety and uncertainty in the weeks following the cardiac event. After diagnosis, spouses and family were the central persons to whom patients articulated their fears. Only a few patients reported that they were approached about MHI by a doctor as part of the CHD diagnosis. In general, MHI was not discussed until months after the diagnosis. Few patients engaged in a conversation about MHI with a professional. They reported that either the GP or a psychologist in a rehabilitation clinic approached them.No, not really. No one actually asked, no doctor, whether there was still a need for treatment. Patient 16, l. 22–24Of course, the doctor should have to ask more background information. How the patient is doing in other areas after this disease, whether he has psychological issues. I think that most specialists, e.g., cardiologists, proceed very technocratically and reel off their plan and don't care much about other things. I have not met any cardiologist, except for the head physician at the university hospital, who has dealt more intensively with this issue. For them it is only important how the echography is, how the ultrasound examination is, the blood values, etc. That's it. I even believe that cardiologists don't have a heart (laughs). Patient 9, l. 78–85Well, I have the feeling that my cardiologist doesn't get involved with me on an emotional level. It's relatively quick, he does a ECG or an ultrasound or something else and then I get my information and have to leave. It's not something where you have a longer conversation, that's what they do. only the treatment, then you go back to the treating GP. Patient 14, l. 131–137\n\nNo, not really. No one actually asked, no doctor, whether there was still a need for treatment. Patient 16, l. 22–24\nOf course, the doctor should have to ask more background information. How the patient is doing in other areas after this disease, whether he has psychological issues. I think that most specialists, e.g., cardiologists, proceed very technocratically and reel off their plan and don't care much about other things. I have not met any cardiologist, except for the head physician at the university hospital, who has dealt more intensively with this issue. For them it is only important how the echography is, how the ultrasound examination is, the blood values, etc. That's it. I even believe that cardiologists don't have a heart (laughs). Patient 9, l. 78–85\nWell, I have the feeling that my cardiologist doesn't get involved with me on an emotional level. It's relatively quick, he does a ECG or an ultrasound or something else and then I get my information and have to leave. It's not something where you have a longer conversation, that's what they do. only the treatment, then you go back to the treating GP. Patient 14, l. 131–137\nThe following section contains information about how the patients experienced the cardiac event and whether they were really approached by the doctor to talk about MHI. Patients experienced CHD as a decisive turning point that unsettled them permanently. Most respondents felt significant anxiety and uncertainty in the weeks following the cardiac event. After diagnosis, spouses and family were the central persons to whom patients articulated their fears. Only a few patients reported that they were approached about MHI by a doctor as part of the CHD diagnosis. In general, MHI was not discussed until months after the diagnosis. Few patients engaged in a conversation about MHI with a professional. They reported that either the GP or a psychologist in a rehabilitation clinic approached them.No, not really. No one actually asked, no doctor, whether there was still a need for treatment. Patient 16, l. 22–24Of course, the doctor should have to ask more background information. How the patient is doing in other areas after this disease, whether he has psychological issues. I think that most specialists, e.g., cardiologists, proceed very technocratically and reel off their plan and don't care much about other things. I have not met any cardiologist, except for the head physician at the university hospital, who has dealt more intensively with this issue. For them it is only important how the echography is, how the ultrasound examination is, the blood values, etc. That's it. I even believe that cardiologists don't have a heart (laughs). Patient 9, l. 78–85Well, I have the feeling that my cardiologist doesn't get involved with me on an emotional level. It's relatively quick, he does a ECG or an ultrasound or something else and then I get my information and have to leave. It's not something where you have a longer conversation, that's what they do. only the treatment, then you go back to the treating GP. Patient 14, l. 131–137\n\nNo, not really. No one actually asked, no doctor, whether there was still a need for treatment. Patient 16, l. 22–24\nOf course, the doctor should have to ask more background information. How the patient is doing in other areas after this disease, whether he has psychological issues. I think that most specialists, e.g., cardiologists, proceed very technocratically and reel off their plan and don't care much about other things. I have not met any cardiologist, except for the head physician at the university hospital, who has dealt more intensively with this issue. For them it is only important how the echography is, how the ultrasound examination is, the blood values, etc. That's it. I even believe that cardiologists don't have a heart (laughs). Patient 9, l. 78–85\nWell, I have the feeling that my cardiologist doesn't get involved with me on an emotional level. It's relatively quick, he does a ECG or an ultrasound or something else and then I get my information and have to leave. It's not something where you have a longer conversation, that's what they do. only the treatment, then you go back to the treating GP. Patient 14, l. 131–137\n[SUBTITLE] Patients' needs [SUBSECTION] [SUBTITLE] The wish for a conversation about MHI [SUBSECTION] All patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n\nI think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70\nIt has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222\nBut then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\nAll patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n\nI think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70\nIt has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222\nBut then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n[SUBTITLE] No wish for a conversation about MHI [SUBSECTION] A minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n\nNo. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\nA minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n\nNo. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n[SUBTITLE] Access and barriers [SUBSECTION] Limited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n\nThe rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200\nWell, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\nLimited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n\nThe rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200\nWell, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n[SUBTITLE] Specific age‐related issues [SUBSECTION] Besides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’.\nBesides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’.\n[SUBTITLE] The younger patient group and their concerns about their ability to work [SUBSECTION] The needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n\nWell, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51\nI don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\nThe needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n\nWell, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51\nI don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n[SUBTITLE] The older patient group and their concerns about successful aging [SUBSECTION] On the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\n\n‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\nOn the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\n\n‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\n[SUBTITLE] The wish for a conversation about MHI [SUBSECTION] All patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n\nI think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70\nIt has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222\nBut then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\nAll patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n\nI think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70\nIt has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222\nBut then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n[SUBTITLE] No wish for a conversation about MHI [SUBSECTION] A minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n\nNo. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\nA minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n\nNo. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n[SUBTITLE] Access and barriers [SUBSECTION] Limited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n\nThe rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200\nWell, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\nLimited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n\nThe rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200\nWell, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n[SUBTITLE] Specific age‐related issues [SUBSECTION] Besides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’.\nBesides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’.\n[SUBTITLE] The younger patient group and their concerns about their ability to work [SUBSECTION] The needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n\nWell, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51\nI don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\nThe needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n\nWell, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51\nI don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n[SUBTITLE] The older patient group and their concerns about successful aging [SUBSECTION] On the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\n\n‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\nOn the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\n\n‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144", "Most participants were male (n = 258, 70.9%). The mean age across genders was M\nage = 65.9 (SD = 11.4); 72.5% lived together with a partner (n = 264); 27.5% had a left ventricular ejection fraction of <40%; 63.2% had a PCI for a myocardial infarction; 52.5% (n = 191) received a positive HADS score and 28.0% (n = 102) had a positive SCID diagnosis. Twelve patients had a positive HADS score but refused further testing (SCID‐I). The following SCID diagnoses were found: unipolar depression (n = 59, 16.5%), bipolar disorder (n = 2, 0.6%), anxiety disorder (n = 26, 7.2%), substance use/addiction disorder (n = 14, 4.0%), adjustment disorder (n = 11, 3.0%) and posttraumatic stress disorder (n = 4, 1.1%). Table 1 provides an overview of the demographic and clinical characteristics. For more details on the ICD‐10 diagnoses, see Table A1.\nSociodemographic and clinical characteristics of the participants\nAbbreviations: MHI: mental health issue; NYHA; New York Heart Association; PCI, percutaneous coronary intervention.\nIt is possible for patients to have more than one answer.\nAt least one cell was too small for the appropriate analysis.", "A total of 80.8% with MHI and 74.2% without MHI found it appropriate to have been approached by the doctor (e.g., GP or cardiologist) about MHI; several patients were unsure whether they found it appropriate (5.8% with MHI; 7.5% without MHI). Patients expressed the general wish to be approached proactively by their doctor about MHI (46.5% with MHI; 38.0% without MHI). Approximately a fifth of patients in both groups were unsure about whether they wished to be approached by the doctor about MHI. About 23.4% of MHI patients mentioned a preference for GPs to help them find adequate treatment for drug therapy. For detailed information, see Table 2.\nDetails to access, barriers and needs regarding healthcare in CHD patients with and without mental health issues (MHI)\nAbbreviation: CHD, coronary heart disease.\nMultiple‐choice question.\nAll percentages relate to the maximum number of patients who were eligible for the underlying question.\nAt least one cell was too small for the appropriate analysis.", "A total of 30 CHD patients with MHI (42.3%) stated that they received an adequate amount of information on the content and accessibility of psychological care. Only a minority of patients obtained information about psychotherapy (7.0%) with a quarter receiving no information at all. About 30% of patients obtained information on the content and accessibility of psychological care through their doctors, while 35% stated that they did not receive further information. When patients did obtain information, half of all patients with MHI got information from the GP or other persons outside of the healthcare system (e.g., family members, friends); 47.8% of MHI patients and 44.1% of patients without MHI wished to get this specific information from the GP. For further information, see Table 2.", "In total, 65 CHD patients agreed to participate in a qualitative interview. These patients were mostly men (n = 53) and had an ejection fraction of <40% (n = 41). Divided over the three settings, 21.5% of patients came from hospitals, 33.9% from rehabilitation clinics and 44.6% from cardiology practices. From this subpopulation, 20 patients were selected and asked to participate. The mean age was M = 67.2 (SD = 12.6). All patients lived with a partner. For detailed information about the selection process, see Figure 1.", "The following section contains information about how the patients experienced the cardiac event and whether they were really approached by the doctor to talk about MHI. Patients experienced CHD as a decisive turning point that unsettled them permanently. Most respondents felt significant anxiety and uncertainty in the weeks following the cardiac event. After diagnosis, spouses and family were the central persons to whom patients articulated their fears. Only a few patients reported that they were approached about MHI by a doctor as part of the CHD diagnosis. In general, MHI was not discussed until months after the diagnosis. Few patients engaged in a conversation about MHI with a professional. They reported that either the GP or a psychologist in a rehabilitation clinic approached them.No, not really. No one actually asked, no doctor, whether there was still a need for treatment. Patient 16, l. 22–24Of course, the doctor should have to ask more background information. How the patient is doing in other areas after this disease, whether he has psychological issues. I think that most specialists, e.g., cardiologists, proceed very technocratically and reel off their plan and don't care much about other things. I have not met any cardiologist, except for the head physician at the university hospital, who has dealt more intensively with this issue. For them it is only important how the echography is, how the ultrasound examination is, the blood values, etc. That's it. I even believe that cardiologists don't have a heart (laughs). Patient 9, l. 78–85Well, I have the feeling that my cardiologist doesn't get involved with me on an emotional level. It's relatively quick, he does a ECG or an ultrasound or something else and then I get my information and have to leave. It's not something where you have a longer conversation, that's what they do. only the treatment, then you go back to the treating GP. Patient 14, l. 131–137\n\nNo, not really. No one actually asked, no doctor, whether there was still a need for treatment. Patient 16, l. 22–24\nOf course, the doctor should have to ask more background information. How the patient is doing in other areas after this disease, whether he has psychological issues. I think that most specialists, e.g., cardiologists, proceed very technocratically and reel off their plan and don't care much about other things. I have not met any cardiologist, except for the head physician at the university hospital, who has dealt more intensively with this issue. For them it is only important how the echography is, how the ultrasound examination is, the blood values, etc. That's it. I even believe that cardiologists don't have a heart (laughs). Patient 9, l. 78–85\nWell, I have the feeling that my cardiologist doesn't get involved with me on an emotional level. It's relatively quick, he does a ECG or an ultrasound or something else and then I get my information and have to leave. It's not something where you have a longer conversation, that's what they do. only the treatment, then you go back to the treating GP. Patient 14, l. 131–137", "[SUBTITLE] The wish for a conversation about MHI [SUBSECTION] All patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n\nI think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70\nIt has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222\nBut then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\nAll patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n\nI think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70\nIt has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222\nBut then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n[SUBTITLE] No wish for a conversation about MHI [SUBSECTION] A minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n\nNo. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\nA minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n\nNo. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n[SUBTITLE] Access and barriers [SUBSECTION] Limited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n\nThe rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200\nWell, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\nLimited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n\nThe rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200\nWell, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n[SUBTITLE] Specific age‐related issues [SUBSECTION] Besides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’.\nBesides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’.\n[SUBTITLE] The younger patient group and their concerns about their ability to work [SUBSECTION] The needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n\nWell, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51\nI don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\nThe needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n\nWell, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51\nI don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n[SUBTITLE] The older patient group and their concerns about successful aging [SUBSECTION] On the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\n\n‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\nOn the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\n\n‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144", "All patients reported that they would find it appropriate to be asked by a doctor about MHI and that they wish for a conversation about MHI with their doctors. Patients reported a high level of suffering until they talked about MHI. If patients had a professional talk about MHI, the important counterpart was the GP. The conversation was experienced as very positive and relieving when GPs talked about psychological problems on their initiative. Patients reported that the professional talk about MHI should occur shortly after the CHD diagnosis. Patients deliberately changed the GP to find a doctor who was willing to talk about MHI or even actively approached them about possible MHI. One patient experienced contact with his cardiologist as very technical and rational and switched to a cardiologist where he also experienced emotional care. Few patients remembered that they were approached for MHI during their stay in rehabilitation clinics. Those who had psychological appointments in rehabilitation clinics experienced them as positive. No further psychological treatment was needed in the subsequent treatment. Interventions had positive effects on patients' coping skills and self‐care but also resulted in feelings of relief.I think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70It has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222But then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210\n\nI think that all this belongs to a good doctor. Of course, he should carefully address everything. But he should ask. Everything, all these things, so that a depression can be recognized at an early stage. […] In the course of the conversation the doctor might get the impression that there is a serious problem and I think a good doctor should also address that. Patient 8, l. 68–70\nIt has to be a doctor, a GP, who talks to people. So, we've had a new family doctor for almost two years now, and he talks to me. The previous one didn't talk to me. And I'm very happy that I can go there, where I can just sit for a moment and think about what I still have to talk about, and as far as I know that's not usual. So otherwise, I came in, five minutes,—bang bang—went out of the room, got my prescription and that's it. I just don't want to have things like that anymore. Patient 14, l. 215–222\nBut then there was a doctor in rehab, that was the first one who really listened to me, and he also got my story, really informed himself, he was the one who told me that everything was connected. The depression, the heart, and so on. This man has helped me very much. He acknowledged that. that this was not overreacting. I was really suffering. Patient 8, l. 206–210", "A minority of patients did not want to talk about possible psychological distress and denied it. They reported strictly separate physical and psychological impairments or even denied an interaction between the two. One patient denied a link between physiological events such as CHD and possible psychological consequences.No. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72\n\nNo. For me, everything just continues as before. The heart attack was basically like a car repair. There was a doctor who said I had a constriction in the arteries of the heart where this stent was inserted. And after that, that was it for me, I hardly gave it a second thought. Patient 5, l. 68–72", "Limited resources (e.g., lack of time, staff shortage) were often reported as barriers. Several interviewed patients reported that during a triweekly stay in a rehabilitation clinic, only a few patients were screened and asked about MHI. Patients with MHI reported numerous unsuccessful attempts to have a psychological appointment and failures to find an ambulant psychotherapist. They complained about insufficient support by the healthcare system, lacking opportunities to talk about their MHI on medical appointments and lacking help with the search for an ambulant psychotherapist. Drug therapy was not a sufficient alternative for this group; most of them rejected this alternative. Patients were demoralized by those failures, hence they developed a passive coping style, stopped actively searching for psychotherapeutic support or postponed it for the future.The rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200Well, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96\n\nThe rehabilitation was initiated in the hospital and in the rehab itself there also was a psychologist who was supposed to take care of the processing of these things, but unfortunately she was ill while I was in rehab. And then I was not able to implement this in the rehab. I would have liked to have some appointments with her. Now I had bad luck that the psychological support was cancelled. Maybe that would have changed something. Patient 9, 22–26, l. 197–200\nWell, a drug therapy. I don't believe that this is necessary. Maybe in one case for sure, but I don't think that would help me. Patient 16, l. 94–96", "Besides the three main categories, a fourth important category emerged and was summed up as ‘specific issues of younger and older patient groups’.", "The needs of patients varied according to their life phases. Patients in the age range 50–60 years expressed fears and concerns about their future ability to continue with their work. They were also the subgroup who experienced the most depression and anxiety symptoms and reported the highest level of suffering. Employed patients were more accessible to talk about MHI in general, were more sensitive to handle the situation adequately, showed a high adherence to medical advice, were more informed about possible psychotherapeutic services, were more motivated to be active in their medical treatment and had better problem‐solving mechanisms.Well, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51I don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50\n\nWell, I'm working again now, aren't I? But when it came to the question of when I would go back to work, I was very unhappy and very uncertain and I thought, how can I do that? Because it is … well, it is still that I am anxious. So, I have body sensations that are strange to me. And they frighten me. And then I think, well, I hope I don't fall down and die. Well, that's something like that. And once I wasn't feeling well and my GP said to me: ‘You can live up to a 100 years with the bypass, but if you don't work on yourself or something like that, then it's not good for your heart and you'll get sicker.’ […] I'm concerned about the disabled person's pass. Maybe I should go to a psychiatrist to discuss it. Patient 14, l. 37–51\nI don't know if you can imagine that you would be told overnight that you can no longer participate in the active working life that you had and could have until then. Patient 7, l. 48–50", "On the other side, patients who were already retired (65 years and above) expressed more worries about successful ageing. This type of patient often reported fears about the future quality of life and the responsibility for their own family. Retired patients suffered from more somatic comorbidities and were less active in their own medical treatment. For such patients, social networks functioned as a resistance factor. Other factors were being optimistic, acceptance of the current situation and appreciating the last years of life.‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144\n\n‘I think the family must be informed, especially the closer family circle. Just because I had two heart attacks and I hear badly on the left side. that is a handicap in life that you don't see. […] these are limitations that no one can imagine […] this handling of heart diseases in society … if you are no longer so efficient and so resilient, mentally as well as physically, it really is an impairment. One does not see the disease and it is no problem for an outsider who has no problems with it … (Those people) think to themselves: why can't this man walk five stairs? […] And these are such things that are important for a family, because one must also manage everyday life and have the support to get along again in social life. Patient 16, l. 117–125 and l. 135–144", "This mixed‐method study aimed at exploring needs, access and barriers in the care of CHD patients from the patient's viewpoint. In our study, 28% of CHD patients were identified as having MHI. In accordance with the literature, the MHI in our study were mainly depression and anxiety disorders,\n7\n, \n8\n, \n30\n and women were twice as likely to develop MHI.\n1\n Taking the quantitative and qualitative results together, patients reported unmet needs, decreased access to helpful resources and encountered multiple obstacles in handling their MHI adequately.\n[SUBTITLE] Perception of the cardiac event and patients' needs [SUBSECTION] Both quantitative and qualitative data show in general that for all recruitment centres (hospital cardiac departments, cardiology practices and rehabilitation clinics) patients with and without MHI experience anxiety and uncertainty after a cardiac event. This event was perceived as drastic and life‐changing, and consequently, patients felt a great need to talk about what had happened. They expressed disappointment about the fact that doctors rarely address MHI. Interviewed patients stated that they show little initiative during their medical consultation and usually did not address MHI on their own. A study from Lussier et al.\n11\n developed an online tool to encourage patients to be more active and prepared for their healthcare encounters by providing skills coaching via a website. Although patients reported a positive perception of such coaching and a higher motivational level, barriers such as lack of interest, limited access to technology, lack of time or language skills reduced the success of this approach.\n11\n\n\nHowever, when MHI was discussed with the doctor, patients did report a decrease in symptoms, mainly a reduction in anxiety, and a positive influence on their feeling of being able to cope with the cardiac event. It therefore seems reasonable that doctors, for their part, ask more regularly about MHI during medical consultations to improve the previous deficit in the area of emotional care. For this purpose, it seems necessary to have targeted interventions, such as creating greater awareness for this topic and the vulnerable patient group in everyday medical practice, to ensure rapid detection of MHI and thus adequate care. Further training courses, in the field of basic psychosomatic care and medical interviewing, could be helpful to increase individual communication skills in doctor–patient conversations, especially in an empathic manner during medical conversations. Studies showed that empathy of the doctor perceived by the patient significantly improved patient satisfaction with the received medical care, especially in terms of information exchange, treatment, perceived expertize of the doctor, as well as interpersonal trust and partnership with the doctor.\n31\n A higher empathy‐related behaviour of the doctor might be useful to improve the doctor–patient relationship and might increase the chance of patients daring to speak about MHI to their doctor.\nBoth quantitative and qualitative data show in general that for all recruitment centres (hospital cardiac departments, cardiology practices and rehabilitation clinics) patients with and without MHI experience anxiety and uncertainty after a cardiac event. This event was perceived as drastic and life‐changing, and consequently, patients felt a great need to talk about what had happened. They expressed disappointment about the fact that doctors rarely address MHI. Interviewed patients stated that they show little initiative during their medical consultation and usually did not address MHI on their own. A study from Lussier et al.\n11\n developed an online tool to encourage patients to be more active and prepared for their healthcare encounters by providing skills coaching via a website. Although patients reported a positive perception of such coaching and a higher motivational level, barriers such as lack of interest, limited access to technology, lack of time or language skills reduced the success of this approach.\n11\n\n\nHowever, when MHI was discussed with the doctor, patients did report a decrease in symptoms, mainly a reduction in anxiety, and a positive influence on their feeling of being able to cope with the cardiac event. It therefore seems reasonable that doctors, for their part, ask more regularly about MHI during medical consultations to improve the previous deficit in the area of emotional care. For this purpose, it seems necessary to have targeted interventions, such as creating greater awareness for this topic and the vulnerable patient group in everyday medical practice, to ensure rapid detection of MHI and thus adequate care. Further training courses, in the field of basic psychosomatic care and medical interviewing, could be helpful to increase individual communication skills in doctor–patient conversations, especially in an empathic manner during medical conversations. Studies showed that empathy of the doctor perceived by the patient significantly improved patient satisfaction with the received medical care, especially in terms of information exchange, treatment, perceived expertize of the doctor, as well as interpersonal trust and partnership with the doctor.\n31\n A higher empathy‐related behaviour of the doctor might be useful to improve the doctor–patient relationship and might increase the chance of patients daring to speak about MHI to their doctor.\n[SUBTITLE] Access and barriers to healthcare and specific issues of patient groups [SUBSECTION] In general, patients received little support in seeking psychological, psychiatric or psychotherapeutic help. Lack of time and personal opportunities, none to little help in coping with mental stress, and finding an ambulant psychotherapist were found to be barriers for patients and GPs in dealing with their CHD‐related MHI's.\nShortcomings in the early stages of CHD and MHI‐related treatment can lead to demoralization and learned helplessness in patients. At the same time, complaints such as future‐ and health‐related worries, anxiety, depression and fear of incapacity for work continue to exist and further restrict patients' mental and physical health. As mentioned before, MHI and even mental stress are associated with higher CHD‐related morbidity and mortality\n7\n and hinder the required lifestyle changes to reduce CHD risk factors.\n4\n, \n8\n\n\nThese shortcomings in medical healthcare can occur because the current healthcare system defines ‘valuable care’ as health outcomes relative to costs and hence encompasses efficiency. However, cost reduction without regard to the achieved outcomes leads to limited effective healthcare.\n32\n Following Porter,\n33\n\nvalue‐based healthcare has the following definition: value equals health outcomes that matter to patients regarding the costs of delivering the outcomes. Thus, value measures all services and activities that determine success by meeting the patients' needs. These needs are determined by the medical condition of the patient and are an interrelated set of medical circumstances. A medical condition includes the most commonly associated conditions—meaning that, for example, care for CHD patients must consist of care for hypertension, and so forth.\n32\n, \n33\n Regarding the high prevalence of MHI in CHD patients, it might be advisable to include MHI as further interrelated medical circumstances. For example, only CHD patients in rehabilitation clinics reported having access to psychotherapeutic help in general; patients from practices and hospitals lacked this possibility. Rehabilitation clinics seem to work more within a more value‐based healthcare cycle because they are medical units that specialize in several diseases and thus are organized in the same way as an integrated practice unit,\n33\n thus including all the necessary skilled medical practitioners needed for medical conditions and their co‐occurring problems.\nTo deal with the shortcomings found in this study, a more value‐based and patient‐centred approach is necessary. With regard to structural requirements, earlier detection and correct diagnosis of MHI, as well as appropriate and fast treatment, would be examples (i.e., an appointment with an outpatient psychotherapist). In addition, more psychological measures, such as training for doctors (e.g., to increase the level of empathy and better support in coping with CHD and MHI) and improvements in procedural requirements (more conversation time during doctor contacts and more precise agreements between different departments) are needed. To be able to implement such requirements in practice adequately, the deployment of nursing or case managers in GP practices would be advisable. Nursing managers could use low‐threshold interventions to assist CHD patients with MHI in the search for a suitable form of psychotherapy and/or drug therapy and to accompany their implementation. They could act as a link between the various care providers and ensure a timely exchange of information and early personal appointments.\n34\n\n\nIn general, patients received little support in seeking psychological, psychiatric or psychotherapeutic help. Lack of time and personal opportunities, none to little help in coping with mental stress, and finding an ambulant psychotherapist were found to be barriers for patients and GPs in dealing with their CHD‐related MHI's.\nShortcomings in the early stages of CHD and MHI‐related treatment can lead to demoralization and learned helplessness in patients. At the same time, complaints such as future‐ and health‐related worries, anxiety, depression and fear of incapacity for work continue to exist and further restrict patients' mental and physical health. As mentioned before, MHI and even mental stress are associated with higher CHD‐related morbidity and mortality\n7\n and hinder the required lifestyle changes to reduce CHD risk factors.\n4\n, \n8\n\n\nThese shortcomings in medical healthcare can occur because the current healthcare system defines ‘valuable care’ as health outcomes relative to costs and hence encompasses efficiency. However, cost reduction without regard to the achieved outcomes leads to limited effective healthcare.\n32\n Following Porter,\n33\n\nvalue‐based healthcare has the following definition: value equals health outcomes that matter to patients regarding the costs of delivering the outcomes. Thus, value measures all services and activities that determine success by meeting the patients' needs. These needs are determined by the medical condition of the patient and are an interrelated set of medical circumstances. A medical condition includes the most commonly associated conditions—meaning that, for example, care for CHD patients must consist of care for hypertension, and so forth.\n32\n, \n33\n Regarding the high prevalence of MHI in CHD patients, it might be advisable to include MHI as further interrelated medical circumstances. For example, only CHD patients in rehabilitation clinics reported having access to psychotherapeutic help in general; patients from practices and hospitals lacked this possibility. Rehabilitation clinics seem to work more within a more value‐based healthcare cycle because they are medical units that specialize in several diseases and thus are organized in the same way as an integrated practice unit,\n33\n thus including all the necessary skilled medical practitioners needed for medical conditions and their co‐occurring problems.\nTo deal with the shortcomings found in this study, a more value‐based and patient‐centred approach is necessary. With regard to structural requirements, earlier detection and correct diagnosis of MHI, as well as appropriate and fast treatment, would be examples (i.e., an appointment with an outpatient psychotherapist). In addition, more psychological measures, such as training for doctors (e.g., to increase the level of empathy and better support in coping with CHD and MHI) and improvements in procedural requirements (more conversation time during doctor contacts and more precise agreements between different departments) are needed. To be able to implement such requirements in practice adequately, the deployment of nursing or case managers in GP practices would be advisable. Nursing managers could use low‐threshold interventions to assist CHD patients with MHI in the search for a suitable form of psychotherapy and/or drug therapy and to accompany their implementation. They could act as a link between the various care providers and ensure a timely exchange of information and early personal appointments.\n34\n\n\n[SUBTITLE] Strengths, limitations and further research [SUBSECTION] MenDis‐CHD provided insights into the needs of CHD patients with and without MHI and their access and barriers to healthcare, clarified important health outcomes for this population and indicated the requirements that a more value‐based care system should focus on to increase quality and efficiency.\nNonetheless, this study has some limitations. With regard to the qualitative part of the study, we chose to conduct 20 interviews. Looking at the pool of eligible patients, our sample size is limited by the participant diversity and the recruitment location (e.g., recruitment only in one city in Germany and particularly in cardiology practices). Because patients were mainly recruited in outpatient settings, it is possible that they were biased with regard to their preference to talk to a GP about their MHI. Patients may have perceived the outpatient setting as a familiar environment, as there has not necessarily been an inpatient stay recently and could therefore adopt a one‐sided view. Another limitation was that some of the quantitative items in our questionnaire were self‐developed and had no statistical testing of validation. Replication with a larger sample size and greater diversity, either across Germany or internationally, would be advantageous to explore possible generalization effects. Other vulnerable population groups that also have a high prevalence of MHI (e.g., schizophrenia) should be examined to explore specific health outcomes and needs that are important to the particular group of patients. With more data, researchers could build a better basis in clinical practice to facilitate a change towards a value‐based healthcare system.\nMenDis‐CHD provided insights into the needs of CHD patients with and without MHI and their access and barriers to healthcare, clarified important health outcomes for this population and indicated the requirements that a more value‐based care system should focus on to increase quality and efficiency.\nNonetheless, this study has some limitations. With regard to the qualitative part of the study, we chose to conduct 20 interviews. Looking at the pool of eligible patients, our sample size is limited by the participant diversity and the recruitment location (e.g., recruitment only in one city in Germany and particularly in cardiology practices). Because patients were mainly recruited in outpatient settings, it is possible that they were biased with regard to their preference to talk to a GP about their MHI. Patients may have perceived the outpatient setting as a familiar environment, as there has not necessarily been an inpatient stay recently and could therefore adopt a one‐sided view. Another limitation was that some of the quantitative items in our questionnaire were self‐developed and had no statistical testing of validation. Replication with a larger sample size and greater diversity, either across Germany or internationally, would be advantageous to explore possible generalization effects. Other vulnerable population groups that also have a high prevalence of MHI (e.g., schizophrenia) should be examined to explore specific health outcomes and needs that are important to the particular group of patients. With more data, researchers could build a better basis in clinical practice to facilitate a change towards a value‐based healthcare system.", "Both quantitative and qualitative data show in general that for all recruitment centres (hospital cardiac departments, cardiology practices and rehabilitation clinics) patients with and without MHI experience anxiety and uncertainty after a cardiac event. This event was perceived as drastic and life‐changing, and consequently, patients felt a great need to talk about what had happened. They expressed disappointment about the fact that doctors rarely address MHI. Interviewed patients stated that they show little initiative during their medical consultation and usually did not address MHI on their own. A study from Lussier et al.\n11\n developed an online tool to encourage patients to be more active and prepared for their healthcare encounters by providing skills coaching via a website. Although patients reported a positive perception of such coaching and a higher motivational level, barriers such as lack of interest, limited access to technology, lack of time or language skills reduced the success of this approach.\n11\n\n\nHowever, when MHI was discussed with the doctor, patients did report a decrease in symptoms, mainly a reduction in anxiety, and a positive influence on their feeling of being able to cope with the cardiac event. It therefore seems reasonable that doctors, for their part, ask more regularly about MHI during medical consultations to improve the previous deficit in the area of emotional care. For this purpose, it seems necessary to have targeted interventions, such as creating greater awareness for this topic and the vulnerable patient group in everyday medical practice, to ensure rapid detection of MHI and thus adequate care. Further training courses, in the field of basic psychosomatic care and medical interviewing, could be helpful to increase individual communication skills in doctor–patient conversations, especially in an empathic manner during medical conversations. Studies showed that empathy of the doctor perceived by the patient significantly improved patient satisfaction with the received medical care, especially in terms of information exchange, treatment, perceived expertize of the doctor, as well as interpersonal trust and partnership with the doctor.\n31\n A higher empathy‐related behaviour of the doctor might be useful to improve the doctor–patient relationship and might increase the chance of patients daring to speak about MHI to their doctor.", "In general, patients received little support in seeking psychological, psychiatric or psychotherapeutic help. Lack of time and personal opportunities, none to little help in coping with mental stress, and finding an ambulant psychotherapist were found to be barriers for patients and GPs in dealing with their CHD‐related MHI's.\nShortcomings in the early stages of CHD and MHI‐related treatment can lead to demoralization and learned helplessness in patients. At the same time, complaints such as future‐ and health‐related worries, anxiety, depression and fear of incapacity for work continue to exist and further restrict patients' mental and physical health. As mentioned before, MHI and even mental stress are associated with higher CHD‐related morbidity and mortality\n7\n and hinder the required lifestyle changes to reduce CHD risk factors.\n4\n, \n8\n\n\nThese shortcomings in medical healthcare can occur because the current healthcare system defines ‘valuable care’ as health outcomes relative to costs and hence encompasses efficiency. However, cost reduction without regard to the achieved outcomes leads to limited effective healthcare.\n32\n Following Porter,\n33\n\nvalue‐based healthcare has the following definition: value equals health outcomes that matter to patients regarding the costs of delivering the outcomes. Thus, value measures all services and activities that determine success by meeting the patients' needs. These needs are determined by the medical condition of the patient and are an interrelated set of medical circumstances. A medical condition includes the most commonly associated conditions—meaning that, for example, care for CHD patients must consist of care for hypertension, and so forth.\n32\n, \n33\n Regarding the high prevalence of MHI in CHD patients, it might be advisable to include MHI as further interrelated medical circumstances. For example, only CHD patients in rehabilitation clinics reported having access to psychotherapeutic help in general; patients from practices and hospitals lacked this possibility. Rehabilitation clinics seem to work more within a more value‐based healthcare cycle because they are medical units that specialize in several diseases and thus are organized in the same way as an integrated practice unit,\n33\n thus including all the necessary skilled medical practitioners needed for medical conditions and their co‐occurring problems.\nTo deal with the shortcomings found in this study, a more value‐based and patient‐centred approach is necessary. With regard to structural requirements, earlier detection and correct diagnosis of MHI, as well as appropriate and fast treatment, would be examples (i.e., an appointment with an outpatient psychotherapist). In addition, more psychological measures, such as training for doctors (e.g., to increase the level of empathy and better support in coping with CHD and MHI) and improvements in procedural requirements (more conversation time during doctor contacts and more precise agreements between different departments) are needed. To be able to implement such requirements in practice adequately, the deployment of nursing or case managers in GP practices would be advisable. Nursing managers could use low‐threshold interventions to assist CHD patients with MHI in the search for a suitable form of psychotherapy and/or drug therapy and to accompany their implementation. They could act as a link between the various care providers and ensure a timely exchange of information and early personal appointments.\n34\n\n", "MenDis‐CHD provided insights into the needs of CHD patients with and without MHI and their access and barriers to healthcare, clarified important health outcomes for this population and indicated the requirements that a more value‐based care system should focus on to increase quality and efficiency.\nNonetheless, this study has some limitations. With regard to the qualitative part of the study, we chose to conduct 20 interviews. Looking at the pool of eligible patients, our sample size is limited by the participant diversity and the recruitment location (e.g., recruitment only in one city in Germany and particularly in cardiology practices). Because patients were mainly recruited in outpatient settings, it is possible that they were biased with regard to their preference to talk to a GP about their MHI. Patients may have perceived the outpatient setting as a familiar environment, as there has not necessarily been an inpatient stay recently and could therefore adopt a one‐sided view. Another limitation was that some of the quantitative items in our questionnaire were self‐developed and had no statistical testing of validation. Replication with a larger sample size and greater diversity, either across Germany or internationally, would be advantageous to explore possible generalization effects. Other vulnerable population groups that also have a high prevalence of MHI (e.g., schizophrenia) should be examined to explore specific health outcomes and needs that are important to the particular group of patients. With more data, researchers could build a better basis in clinical practice to facilitate a change towards a value‐based healthcare system.", "This study found that the needs regarding CHD patients with and without MHI were not adequately satisfied. Furthermore, for CHD patients with MHI, access and barriers to the healthcare system regarding psychotherapeutic and drug therapy were insufficient. CHD patients experienced the cardiac event as life‐changing and had an urgent need to talk about mental problems. Primarily, the GP was named as a partner for conversations about CHD‐related mental problems; however, when the doctor spoke to the patient shortly after the cardiac event, patients felt relieved and coped better with their illness. To improve this, psychological measures such as training of doctors in doctor–patient communication are necessary, as well as improvements in procedural and structural requirements.", "Samia Peltzer wrote the abstract, introduction, method section, results (incl. analyses) and discussion, designed the figure and tables, references with EndNote, made an overall adaption of the text and included references. Samia Peltzer was a major contributor in writing the manuscript. Ursula Köstler conducted the information for the qualitative analysis, conducted qualitative analysis and summed up the results for the qualitative part. Frank Jessen, Frank Schulz‐Nieswandt and Christian Albus designed and conducted the study as chief scientists. Hendrik Müller, Nadine Scholten, Frank Jessen and Christian Albus critically revised the manuscript. All gave final approval and agreed to be accountable for all aspects of work, ensuring integrity and accuracy." ]
[ null, null, null, null, null, "materials-and-methods", null, null, null, null, null, null, null, null, null, null, null, null, "results", null, null, null, null, null, null, null, null, null, null, null, null, "discussion", null, null, null, "conclusions", null ]
[ "coronary heart disease", "healthcare system", "mental health issues", "mixed‐method approach", "value‐base healthcare" ]
Failure pattern and suggestions for target volume delineation of carcinoma showing thymus-like differentiation treated with intensity-modulated radiotherapy.
36271328
To review our long-term clinical experience, analyze the failure patterns, and give suggestions for target volume delineation of carcinoma showing thymus-like differentiation (CASTLE) treated with intensity-modulated radiotherapy (IMRT).
BACKGROUND
From April 2008 to May 2019, 30 patients with CASTLE treated by postoperative or radical IMRT in our center were retrospectively reviewed. A total dose of 56-60 Gy in 28-30 fractions was prescribed to patients without residual disease and 66 Gy in 33 fractions for patients with residual or unresectable disease. Survival rates were calculated using the Kaplan-Meier method. Treatment-related toxicities were graded by National Cancer Institute Common Toxicity Criteria (NCI-CTC) version 4.0.
METHODS
Among the 30 patients, 12 (40%) received partial resection or biopsy. Lateral lymph node metastasis was observed in 7 (23.3%) patients. During follow-up, regional lymph node recurrence occurred in 2 patients and distant metastasis in 5 patients. With a median follow-up time of 63.5 months, the 5-year local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastasis-free survival (DMFS), overall survival (OS) and progression-free survival (PFS) rates were 100, 88.9, 78.9, 93.1 and 78.9%, respectively. For patients with no lateral neck node metastasis, prophylactic radiotherapy for lateral neck nodal regions failed to improve RRFS (p = 0.381) and OS (p = 0.153).
RESULTS
Distant metastasis was the major failure pattern for CASTLE after surgery and IMRT. For patients with no lateral neck node metastasis, the omission of irradiation for lateral neck nodal regions seems to be safe and feasible.
CONCLUSION
[ "Humans", "Radiotherapy, Intensity-Modulated", "Retrospective Studies", "Carcinoma", "Radiotherapy Planning, Computer-Assisted", "Lymphatic Metastasis" ]
9585782
null
null
null
null
Results
[SUBTITLE] Patient characteristics [SUBSECTION] A total of 30 patients were enrolled in this study. Patients’ characteristics were summarized in Table 1. The median age was 53 years (range 37–61 years). The ratio of male to female was 1:1. Lymph node metastasis was found in 50% of the patients, among which 53.4% were central and 46.6% were lateral. Tumor extension to adjacent organs, including recurrent laryngeal nerve, great vessels, muscles, esophagus, or trachea was found in 21 (70%) patients. About 53% of the patients received complete resection, and 40% received partial resection or biopsy only. Two patients received cisplatin-based concurrent chemotherapy. After treatment, complete remission (CR) was achieved in 26 (86.7%) patients, and partial remission (PR) in 4 (13.3%) patients. Treatment procedures are detailed in Table 2. Table 1Patient characteristicsNo. (%) of patientsTotal30Median age (range)53 (37–61)GenderMale15 (50)Female15 (50)Tumor locationLeft lobe18 (60)Right lobe11 (36.7)Isthmus1 (3.3)Tumor size (cm)≤ 23 (10)2–415 (50)> 47 (23.3)Unknown5 (16.7)Lymph node metastasisPresent15 (50.0)Absent14 (46.6)Unknown1 (3.3)Location of Lymph node(n = 15)Lateral7 (46.6)Central8 (53.4)Tumor extensionPresent21 (70)Absent7 (23.3)Unknown2 (6.7)STE (n = 21)RLN8 (38.1)Muscles6 (28.6)Esophagus3 (14.3)Great vessels4 (19)Trachea2 (9.5)Thymus1 (4.7)Parathyroid gland1 (4.7)Abbreviations: STE, site of tumor extension; RLN, recurrent laryngeal nerve Patient characteristics Abbreviations: STE, site of tumor extension; RLN, recurrent laryngeal nerve Table 2Treatment procedures and clinical outcomeNo. (%) of patientsType of surgeryR016 (53.3)R12 (6.6)R29 (30)Biopsy3 (10)IMRT dose (Gy)Median (range)60 (56–66)IMRT duration (days)Median (range)44.5 (39–67)Systemic therapyYes vs. No3 (10) vs. 27 (90)Follow-up time (months)Median (range)63.5 (21–161)Treatment responseCR26 (86.7)PR4 (13.3)Local recurrencePresent2 (93.3)Absent28 (6.7)Distant metastasisPresent5 (83.3)Absent25 (16.7)Abbreviations: R0, no residual tumor; R1, microscopic residual tumor; R2, macroscopic residual tumor; IMRT, intensity-modulated radiotherapy; CR, complete remission; PR, partial remission Treatment procedures and clinical outcome Abbreviations: R0, no residual tumor; R1, microscopic residual tumor; R2, macroscopic residual tumor; IMRT, intensity-modulated radiotherapy; CR, complete remission; PR, partial remission Pathology and immunohistochemical (IHC) data were available for 29 out of 30 patients. Among these, 93.1% (27/29) expressed CD5, and 72.4% (21/29) expressed CD117 totally or partially. Instead, 72.4% (21/29) of the cases were negatively expressed for thyroid transcription factor 1 (TTF-1), which was the marker of thyroid follicular cells. A total of 30 patients were enrolled in this study. Patients’ characteristics were summarized in Table 1. The median age was 53 years (range 37–61 years). The ratio of male to female was 1:1. Lymph node metastasis was found in 50% of the patients, among which 53.4% were central and 46.6% were lateral. Tumor extension to adjacent organs, including recurrent laryngeal nerve, great vessels, muscles, esophagus, or trachea was found in 21 (70%) patients. About 53% of the patients received complete resection, and 40% received partial resection or biopsy only. Two patients received cisplatin-based concurrent chemotherapy. After treatment, complete remission (CR) was achieved in 26 (86.7%) patients, and partial remission (PR) in 4 (13.3%) patients. Treatment procedures are detailed in Table 2. Table 1Patient characteristicsNo. (%) of patientsTotal30Median age (range)53 (37–61)GenderMale15 (50)Female15 (50)Tumor locationLeft lobe18 (60)Right lobe11 (36.7)Isthmus1 (3.3)Tumor size (cm)≤ 23 (10)2–415 (50)> 47 (23.3)Unknown5 (16.7)Lymph node metastasisPresent15 (50.0)Absent14 (46.6)Unknown1 (3.3)Location of Lymph node(n = 15)Lateral7 (46.6)Central8 (53.4)Tumor extensionPresent21 (70)Absent7 (23.3)Unknown2 (6.7)STE (n = 21)RLN8 (38.1)Muscles6 (28.6)Esophagus3 (14.3)Great vessels4 (19)Trachea2 (9.5)Thymus1 (4.7)Parathyroid gland1 (4.7)Abbreviations: STE, site of tumor extension; RLN, recurrent laryngeal nerve Patient characteristics Abbreviations: STE, site of tumor extension; RLN, recurrent laryngeal nerve Table 2Treatment procedures and clinical outcomeNo. (%) of patientsType of surgeryR016 (53.3)R12 (6.6)R29 (30)Biopsy3 (10)IMRT dose (Gy)Median (range)60 (56–66)IMRT duration (days)Median (range)44.5 (39–67)Systemic therapyYes vs. No3 (10) vs. 27 (90)Follow-up time (months)Median (range)63.5 (21–161)Treatment responseCR26 (86.7)PR4 (13.3)Local recurrencePresent2 (93.3)Absent28 (6.7)Distant metastasisPresent5 (83.3)Absent25 (16.7)Abbreviations: R0, no residual tumor; R1, microscopic residual tumor; R2, macroscopic residual tumor; IMRT, intensity-modulated radiotherapy; CR, complete remission; PR, partial remission Treatment procedures and clinical outcome Abbreviations: R0, no residual tumor; R1, microscopic residual tumor; R2, macroscopic residual tumor; IMRT, intensity-modulated radiotherapy; CR, complete remission; PR, partial remission Pathology and immunohistochemical (IHC) data were available for 29 out of 30 patients. Among these, 93.1% (27/29) expressed CD5, and 72.4% (21/29) expressed CD117 totally or partially. Instead, 72.4% (21/29) of the cases were negatively expressed for thyroid transcription factor 1 (TTF-1), which was the marker of thyroid follicular cells. [SUBTITLE] Dosimetric data for IMRT [SUBSECTION] Dose-volume histogram (DVH) statistics were showed in Table 3. The average volume of GTV and CTV were 131.3 cc (21.2-387.2) and 323 cc (92.4-1408.2), respectively. Rates of dose coverage were excellent for the target volume. The volume receiving less than 95% of the prescribed dose was 0.3% for GTV and 1.2% for CTV. The mean dose was 67.8 Gy for GTV and 62.7 Gy for CTV. Table 3Dose-volume histograms (DVHs) statistics for IMRTGTVAverage (range)CTVAverage (range)Volume (cc)131.3 (21.2-387.2)323 (92.4-1408.2)Maximum dose (Gy)70.9 (69.2–72.5)67.6 (63.5–72.5)Mean dose (Gy)67.8 (65.9–68.8)62.7 (57.7–66.8)Minimum dose (Gy)56.8 (9.9–65.6)42.1 (38.2–56.3)% volume receiving < 95% of the prescribed dose0.3 (0-1.7)1.2 (0–12)% volume receiving ≥ 100% of the prescribed dose92.1(47.5–100)94.0(77.6–99.7)% volume receiving ≥ 110% of the prescribed dose015.8 (0-67.1)IMRT, intensity-modulated radiotherapy; GTV, gross tumor volume; CTV, clinical target volume Dose-volume histograms (DVHs) statistics for IMRT IMRT, intensity-modulated radiotherapy; GTV, gross tumor volume; CTV, clinical target volume Dose-volume histogram (DVH) statistics were showed in Table 3. The average volume of GTV and CTV were 131.3 cc (21.2-387.2) and 323 cc (92.4-1408.2), respectively. Rates of dose coverage were excellent for the target volume. The volume receiving less than 95% of the prescribed dose was 0.3% for GTV and 1.2% for CTV. The mean dose was 67.8 Gy for GTV and 62.7 Gy for CTV. Table 3Dose-volume histograms (DVHs) statistics for IMRTGTVAverage (range)CTVAverage (range)Volume (cc)131.3 (21.2-387.2)323 (92.4-1408.2)Maximum dose (Gy)70.9 (69.2–72.5)67.6 (63.5–72.5)Mean dose (Gy)67.8 (65.9–68.8)62.7 (57.7–66.8)Minimum dose (Gy)56.8 (9.9–65.6)42.1 (38.2–56.3)% volume receiving < 95% of the prescribed dose0.3 (0-1.7)1.2 (0–12)% volume receiving ≥ 100% of the prescribed dose92.1(47.5–100)94.0(77.6–99.7)% volume receiving ≥ 110% of the prescribed dose015.8 (0-67.1)IMRT, intensity-modulated radiotherapy; GTV, gross tumor volume; CTV, clinical target volume Dose-volume histograms (DVHs) statistics for IMRT IMRT, intensity-modulated radiotherapy; GTV, gross tumor volume; CTV, clinical target volume [SUBTITLE] Survival outcome [SUBSECTION] The median follow-up time was 63.5 months (range 21–161 months). The 5-year LRFS, RRFS, DMFS, OS, and PFS were 100, 88.9, 78.9, 93.1, and 78.9%, respectively (Fig. 2). In the subgroup analysis, we divided patients with no lateral neck node metastasis (23 patients) into two groups: group A (6 patients) with prophylactic irradiation for lateral neck nodal regions (levels II-V), while group B (17 patients) without prophylactic irradiation for lateral neck nodal regions. The results showed that there was no significant difference in RRFS (p = 0.381) and OS (p = 0.153) between the two groups (Fig. 3). Patient characteristics for groups A and B were shown in Supplementary Table 1. Fig. 2Kaplan-Meier curves showing local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastases-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) for patients with carcinoma showing thymus-like differentiation Kaplan-Meier curves showing local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastases-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) for patients with carcinoma showing thymus-like differentiation Fig. 3Kaplan-Meier estimate of (a) regional recurrence-free survival (RRFS), and (b) overall survival (OS) between patients with or without irradiation for lateral neck nodal regions RT, radiotherapy; LR: lateral neck nodal regions Kaplan-Meier estimate of (a) regional recurrence-free survival (RRFS), and (b) overall survival (OS) between patients with or without irradiation for lateral neck nodal regions RT, radiotherapy; LR: lateral neck nodal regions The median follow-up time was 63.5 months (range 21–161 months). The 5-year LRFS, RRFS, DMFS, OS, and PFS were 100, 88.9, 78.9, 93.1, and 78.9%, respectively (Fig. 2). In the subgroup analysis, we divided patients with no lateral neck node metastasis (23 patients) into two groups: group A (6 patients) with prophylactic irradiation for lateral neck nodal regions (levels II-V), while group B (17 patients) without prophylactic irradiation for lateral neck nodal regions. The results showed that there was no significant difference in RRFS (p = 0.381) and OS (p = 0.153) between the two groups (Fig. 3). Patient characteristics for groups A and B were shown in Supplementary Table 1. Fig. 2Kaplan-Meier curves showing local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastases-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) for patients with carcinoma showing thymus-like differentiation Kaplan-Meier curves showing local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastases-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) for patients with carcinoma showing thymus-like differentiation Fig. 3Kaplan-Meier estimate of (a) regional recurrence-free survival (RRFS), and (b) overall survival (OS) between patients with or without irradiation for lateral neck nodal regions RT, radiotherapy; LR: lateral neck nodal regions Kaplan-Meier estimate of (a) regional recurrence-free survival (RRFS), and (b) overall survival (OS) between patients with or without irradiation for lateral neck nodal regions RT, radiotherapy; LR: lateral neck nodal regions [SUBTITLE] Failure patterns and salvage treatment [SUBSECTION] Two patients had regional lymph node recurrence. All the recurrences were in the contralateral supraclavicular fossa and were defined as type E (extraneous) failures. Distant metastasis was found in 5 patients. Common sites for distant metastasis were lung (3 patients), bone (1 patient), and liver (1 patient). Two patients suffered both regional and distant failure. Salvage treatment, including surgery, radiotherapy, and systemic therapy, was given to patients with recurrence or distant metastases. If acceptable, clinical trials were the first choice. For patients with oligometastatic or oligoprogressive tumors, SBRT was implemented. Two patients had regional lymph node recurrence. All the recurrences were in the contralateral supraclavicular fossa and were defined as type E (extraneous) failures. Distant metastasis was found in 5 patients. Common sites for distant metastasis were lung (3 patients), bone (1 patient), and liver (1 patient). Two patients suffered both regional and distant failure. Salvage treatment, including surgery, radiotherapy, and systemic therapy, was given to patients with recurrence or distant metastases. If acceptable, clinical trials were the first choice. For patients with oligometastatic or oligoprogressive tumors, SBRT was implemented. [SUBTITLE] Treatment complications [SUBSECTION] The majority of acute toxicities related to IMRT were grade 1–2. Grade 3 mucositis or dermatitis occurred only in 2 patients. No grade 4 toxicities were observed. Details were showed in Table 4. Three patients suffered treatment interruption due to other diseases, including aspiration pneumonia and herpes zoster. After appropriate treatment, they continued and finished IMRT. There was no late toxicity (≥ grade 2) related to IMRT, such as radiation pneumonitis, neck fibrosis, or esophageal stenosis. Table 4Acute toxicities during IMRTToxicitiesNo. of patients by toxicity grade (%)Grade 1Grade 2Grade 3Grade 4Grade 5Mucositis9 (30)20 (66.7)1 (3.3)00Dermatitis25 (83.3)4 (13.3)1 (3.3)00Skin pigmentation26 (86.7)0000Anemia4 (13.3)1 (3.3)000Leukopenia5 (16.7)1 (3.3)000Thrombocytopenia3 (10)0000IMRT, intensity-modulated radiotherapy Acute toxicities during IMRT IMRT, intensity-modulated radiotherapy The majority of acute toxicities related to IMRT were grade 1–2. Grade 3 mucositis or dermatitis occurred only in 2 patients. No grade 4 toxicities were observed. Details were showed in Table 4. Three patients suffered treatment interruption due to other diseases, including aspiration pneumonia and herpes zoster. After appropriate treatment, they continued and finished IMRT. There was no late toxicity (≥ grade 2) related to IMRT, such as radiation pneumonitis, neck fibrosis, or esophageal stenosis. Table 4Acute toxicities during IMRTToxicitiesNo. of patients by toxicity grade (%)Grade 1Grade 2Grade 3Grade 4Grade 5Mucositis9 (30)20 (66.7)1 (3.3)00Dermatitis25 (83.3)4 (13.3)1 (3.3)00Skin pigmentation26 (86.7)0000Anemia4 (13.3)1 (3.3)000Leukopenia5 (16.7)1 (3.3)000Thrombocytopenia3 (10)0000IMRT, intensity-modulated radiotherapy Acute toxicities during IMRT IMRT, intensity-modulated radiotherapy [SUBTITLE] Prognostic factors [SUBSECTION] Univariate analysis, taking age, gender, tumor size, tumor extension, surgery type, and lymph node metastasis as prognostic factors for RRFS, DMFS, OS, and PFS, was performed. Univariate analysis showed that surgery type (p = 0.026, 0.000, 0.026) and lateral neck node metastasis (p = 0.024, 0.009, 0.024) were potential prognostic factors for DMFS, OS and PFS (Supplementary Table 2). However, the multivariate analysis failed to further validate the differences (Supplementary Table 3). We attributed it to the relatively small sample size and few end-point events. Univariate analysis, taking age, gender, tumor size, tumor extension, surgery type, and lymph node metastasis as prognostic factors for RRFS, DMFS, OS, and PFS, was performed. Univariate analysis showed that surgery type (p = 0.026, 0.000, 0.026) and lateral neck node metastasis (p = 0.024, 0.009, 0.024) were potential prognostic factors for DMFS, OS and PFS (Supplementary Table 2). However, the multivariate analysis failed to further validate the differences (Supplementary Table 3). We attributed it to the relatively small sample size and few end-point events.
Conclusion
Our long-term results showed that surgery combined with IMRT is an effective treatment for patients with CASTLE. Distant metastasis is the major failure pattern. For patients with no lateral neck node metastasis, the omission of irradiation for lateral neck nodal regions seems to be safe and feasible. Further prospective research is warranted.
[ "Background", "Methods", "Patients", "Intensity-modulate radiotherapy", "Patient evaluation", "Definition of failure pattern", "Statistical methods", "Patient characteristics", "Dosimetric data for IMRT", "Survival outcome", "Failure patterns and salvage treatment", "Treatment complications", "Prognostic factors", "" ]
[ "Carcinoma showing thymus-like differentiation (CASTLE) is a rare malignant tumor of the thyroid or adjacent soft tissue in the neck, which accounts for only 0.1–0.15% of all thyroid cancers [1–4]. Until now, fewer than 100 patients have been reported, and most of them are case reports. Optimal treatment for CASTLE remains uncertain. Surgery is generally recommended as the mainstay of treatment [3, 5]. However, due to the lower location and high possibility of extrathyroid invasion, complete resection can be challenging for patients with locally advanced disease [6]. It is reported that the incidence of lymph node metastasis is 50–69% and 60–80% for extrathyroid invasion [5–9]. Therefore, radiotherapy is often used as part of the treatment and the efficiency of radiotherapy has been proved by several studies [6, 7, 10, 11].\nHowever, there is no consensus on the delineation of target volume for CASTLE since very limited data has been published. Accurate target volume delineation is the premise of intensity-modulated radiotherapy (IMRT). Excessive irradiation will increase treatment-related toxicities like dermatitis and neck fibrosis, while insufficiency of target volume may cause tumor recurrence. Hence, in the present study, we reviewed our 11 years of clinical experience, analyzed the failure patterns, and gave suggestions for target volume delineation of CASTLE treated with IMRT. To the best of our knowledge, this is the first study focusing on target volume delineation for this rare disease.", "[SUBTITLE] Patients [SUBSECTION] From April 2008 to May 2019, 30 patients with CASTLE treated by postoperative or radical IMRT in our center were retrospectively reviewed. We collected clinicopathological data, treatment procedures, and clinical outcomes. The patterns of treatment failure were also analyzed. The study was approved by the Institutional Review Board of Fudan University Shanghai Cancer Center. Informed consent was obtained from all patients.\nFrom April 2008 to May 2019, 30 patients with CASTLE treated by postoperative or radical IMRT in our center were retrospectively reviewed. We collected clinicopathological data, treatment procedures, and clinical outcomes. The patterns of treatment failure were also analyzed. The study was approved by the Institutional Review Board of Fudan University Shanghai Cancer Center. Informed consent was obtained from all patients.\n[SUBTITLE] Intensity-modulate radiotherapy [SUBSECTION] IMRT was started 4 to 6 weeks after surgery or early after diagnosis. The techniques of IMRT were detailed in our previously published data [7]. Briefly, a thermoplastic mask of the head and shoulder was used for patient immobilization. Patients received computed tomography (CT) simulation at 5 mm thickness of the head and neck region in the supine position. Image fusion of magnetic resonance imaging (MRI) and CT was recommended for target volume delineation. The gross tumor volume (GTV) was defined as all primary gross tumors and involved lymph nodes determined by imaging and clinical findings. The clinical target volume (CTV) included the GTV or tumor bed plus a 5 to 10 mm margin to encompass any microscopic extension. The planning target volume (PTV) was defined as the CTV plus a 3 to 5 mm margin to encompass setup error. Neck nodal level VI was conventionally included in the CTV. If there was no positive lymph node, neck nodal levels II-V were not included in the CTV. Examples of target delineation were showed in Fig. 1. Positive lymph nodes were defined as follows: (1) pathologically diagnosed; (2) minimum axial diameter ≥ 1 cm; (3) extranodal extension or circular enhancement. The prescribed dose was 60 Gy for patients without residual disease and 66 Gy for patients with residual or unresectable disease. Conventional fractionation (2 Gy per fraction, one fraction per day, five days per week) was used.\n\nFig. 1Examples of target volume delineation for carcinoma showing thymus-like differentiation GTV, gross tumor volume; PTV-G, planning target volume for GTV; CTV, clinical target volume; PTV-C, planning target volume for CTV.\n\nExamples of target volume delineation for carcinoma showing thymus-like differentiation\n GTV, gross tumor volume; PTV-G, planning target volume for GTV; CTV, clinical target volume;\n PTV-C, planning target volume for CTV.\nIMRT was started 4 to 6 weeks after surgery or early after diagnosis. The techniques of IMRT were detailed in our previously published data [7]. Briefly, a thermoplastic mask of the head and shoulder was used for patient immobilization. Patients received computed tomography (CT) simulation at 5 mm thickness of the head and neck region in the supine position. Image fusion of magnetic resonance imaging (MRI) and CT was recommended for target volume delineation. The gross tumor volume (GTV) was defined as all primary gross tumors and involved lymph nodes determined by imaging and clinical findings. The clinical target volume (CTV) included the GTV or tumor bed plus a 5 to 10 mm margin to encompass any microscopic extension. The planning target volume (PTV) was defined as the CTV plus a 3 to 5 mm margin to encompass setup error. Neck nodal level VI was conventionally included in the CTV. If there was no positive lymph node, neck nodal levels II-V were not included in the CTV. Examples of target delineation were showed in Fig. 1. Positive lymph nodes were defined as follows: (1) pathologically diagnosed; (2) minimum axial diameter ≥ 1 cm; (3) extranodal extension or circular enhancement. The prescribed dose was 60 Gy for patients without residual disease and 66 Gy for patients with residual or unresectable disease. Conventional fractionation (2 Gy per fraction, one fraction per day, five days per week) was used.\n\nFig. 1Examples of target volume delineation for carcinoma showing thymus-like differentiation GTV, gross tumor volume; PTV-G, planning target volume for GTV; CTV, clinical target volume; PTV-C, planning target volume for CTV.\n\nExamples of target volume delineation for carcinoma showing thymus-like differentiation\n GTV, gross tumor volume; PTV-G, planning target volume for GTV; CTV, clinical target volume;\n PTV-C, planning target volume for CTV.\n[SUBTITLE] Patient evaluation [SUBSECTION] Patients were assessed weekly during IMRT. After IMRT, patients were follow-up every three months in the first two years, every six months during the years 3–5, and annually after that. Follow-up assessments after treatment included examination of the neck, thyroid function tests, and ultrasound or MRI for the thyroid and neck. Chest CT scan, and ultrasound or CT of the abdomen were performed every 6–12 months. Additional tests were performed when clinically indicated.\nPatients were assessed weekly during IMRT. After IMRT, patients were follow-up every three months in the first two years, every six months during the years 3–5, and annually after that. Follow-up assessments after treatment included examination of the neck, thyroid function tests, and ultrasound or MRI for the thyroid and neck. Chest CT scan, and ultrasound or CT of the abdomen were performed every 6–12 months. Additional tests were performed when clinically indicated.\n[SUBTITLE] Definition of failure pattern [SUBSECTION] The images of MRI or CT scans obtained at the time of recurrence were transferred to the pretreatment planning CT. The dose-volume histogram (DVH) was used to calculate the radiation dose received by the recurrent tumor (GTVrecur) region. The recurrences were classified into five types based on combined spatial and dosimetric criteria [12, 13]:\nType A (central high dose): the mapped centroid of GTVrecur originates in high dose PTV, and ≥ 95% of GTVrecur was within the 95% isodose (high dose PTV);\nType B (peripheral high dose): the mapped centroid of GTVrecur originates in high dose PTV, and < 95% of GTVrecur was within the 95% isodose (high dose PTV);\nType C (central elective dose): the mapped centroid of GTVrecur originates in lower dose PTV, and ≥ 95% of GTVrecur was within the 95% isodose (lower dose PTV);\nType D (peripheral elective dose): the mapped centroid of GTVrecur originates in lower dose PTV, and < 95% of GTVrecur was within the 95% isodose (lower dose PTV);\nType E (extraneous dose): the mapped centroid of GTVrecur originates outside all PTVs.\nThe images of MRI or CT scans obtained at the time of recurrence were transferred to the pretreatment planning CT. The dose-volume histogram (DVH) was used to calculate the radiation dose received by the recurrent tumor (GTVrecur) region. The recurrences were classified into five types based on combined spatial and dosimetric criteria [12, 13]:\nType A (central high dose): the mapped centroid of GTVrecur originates in high dose PTV, and ≥ 95% of GTVrecur was within the 95% isodose (high dose PTV);\nType B (peripheral high dose): the mapped centroid of GTVrecur originates in high dose PTV, and < 95% of GTVrecur was within the 95% isodose (high dose PTV);\nType C (central elective dose): the mapped centroid of GTVrecur originates in lower dose PTV, and ≥ 95% of GTVrecur was within the 95% isodose (lower dose PTV);\nType D (peripheral elective dose): the mapped centroid of GTVrecur originates in lower dose PTV, and < 95% of GTVrecur was within the 95% isodose (lower dose PTV);\nType E (extraneous dose): the mapped centroid of GTVrecur originates outside all PTVs.\n[SUBTITLE] Statistical methods [SUBSECTION] All the statistical analysis was performed using the Statistical Package for Social Sciences (SPSS version 24.0) software. The survival rates were calculated from the day of the first treatment. The Kaplan-Meier method was used to calculate the local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastasis-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) rates. Treatment-related toxicities were graded by National Cancer Institute Common Toxicity Criteria (NCI-CTC) version 4.0.\nAll the statistical analysis was performed using the Statistical Package for Social Sciences (SPSS version 24.0) software. The survival rates were calculated from the day of the first treatment. The Kaplan-Meier method was used to calculate the local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastasis-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) rates. Treatment-related toxicities were graded by National Cancer Institute Common Toxicity Criteria (NCI-CTC) version 4.0.", "From April 2008 to May 2019, 30 patients with CASTLE treated by postoperative or radical IMRT in our center were retrospectively reviewed. We collected clinicopathological data, treatment procedures, and clinical outcomes. The patterns of treatment failure were also analyzed. The study was approved by the Institutional Review Board of Fudan University Shanghai Cancer Center. Informed consent was obtained from all patients.", "IMRT was started 4 to 6 weeks after surgery or early after diagnosis. The techniques of IMRT were detailed in our previously published data [7]. Briefly, a thermoplastic mask of the head and shoulder was used for patient immobilization. Patients received computed tomography (CT) simulation at 5 mm thickness of the head and neck region in the supine position. Image fusion of magnetic resonance imaging (MRI) and CT was recommended for target volume delineation. The gross tumor volume (GTV) was defined as all primary gross tumors and involved lymph nodes determined by imaging and clinical findings. The clinical target volume (CTV) included the GTV or tumor bed plus a 5 to 10 mm margin to encompass any microscopic extension. The planning target volume (PTV) was defined as the CTV plus a 3 to 5 mm margin to encompass setup error. Neck nodal level VI was conventionally included in the CTV. If there was no positive lymph node, neck nodal levels II-V were not included in the CTV. Examples of target delineation were showed in Fig. 1. Positive lymph nodes were defined as follows: (1) pathologically diagnosed; (2) minimum axial diameter ≥ 1 cm; (3) extranodal extension or circular enhancement. The prescribed dose was 60 Gy for patients without residual disease and 66 Gy for patients with residual or unresectable disease. Conventional fractionation (2 Gy per fraction, one fraction per day, five days per week) was used.\n\nFig. 1Examples of target volume delineation for carcinoma showing thymus-like differentiation GTV, gross tumor volume; PTV-G, planning target volume for GTV; CTV, clinical target volume; PTV-C, planning target volume for CTV.\n\nExamples of target volume delineation for carcinoma showing thymus-like differentiation\n GTV, gross tumor volume; PTV-G, planning target volume for GTV; CTV, clinical target volume;\n PTV-C, planning target volume for CTV.", "Patients were assessed weekly during IMRT. After IMRT, patients were follow-up every three months in the first two years, every six months during the years 3–5, and annually after that. Follow-up assessments after treatment included examination of the neck, thyroid function tests, and ultrasound or MRI for the thyroid and neck. Chest CT scan, and ultrasound or CT of the abdomen were performed every 6–12 months. Additional tests were performed when clinically indicated.", "The images of MRI or CT scans obtained at the time of recurrence were transferred to the pretreatment planning CT. The dose-volume histogram (DVH) was used to calculate the radiation dose received by the recurrent tumor (GTVrecur) region. The recurrences were classified into five types based on combined spatial and dosimetric criteria [12, 13]:\nType A (central high dose): the mapped centroid of GTVrecur originates in high dose PTV, and ≥ 95% of GTVrecur was within the 95% isodose (high dose PTV);\nType B (peripheral high dose): the mapped centroid of GTVrecur originates in high dose PTV, and < 95% of GTVrecur was within the 95% isodose (high dose PTV);\nType C (central elective dose): the mapped centroid of GTVrecur originates in lower dose PTV, and ≥ 95% of GTVrecur was within the 95% isodose (lower dose PTV);\nType D (peripheral elective dose): the mapped centroid of GTVrecur originates in lower dose PTV, and < 95% of GTVrecur was within the 95% isodose (lower dose PTV);\nType E (extraneous dose): the mapped centroid of GTVrecur originates outside all PTVs.", "All the statistical analysis was performed using the Statistical Package for Social Sciences (SPSS version 24.0) software. The survival rates were calculated from the day of the first treatment. The Kaplan-Meier method was used to calculate the local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastasis-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) rates. Treatment-related toxicities were graded by National Cancer Institute Common Toxicity Criteria (NCI-CTC) version 4.0.", "A total of 30 patients were enrolled in this study. Patients’ characteristics were summarized in Table 1. The median age was 53 years (range 37–61 years). The ratio of male to female was 1:1. Lymph node metastasis was found in 50% of the patients, among which 53.4% were central and 46.6% were lateral. Tumor extension to adjacent organs, including recurrent laryngeal nerve, great vessels, muscles, esophagus, or trachea was found in 21 (70%) patients. About 53% of the patients received complete resection, and 40% received partial resection or biopsy only. Two patients received cisplatin-based concurrent chemotherapy. After treatment, complete remission (CR) was achieved in 26 (86.7%) patients, and partial remission (PR) in 4 (13.3%) patients. Treatment procedures are detailed in Table 2.\n\nTable 1Patient characteristicsNo. (%) of patientsTotal30Median age (range)53 (37–61)GenderMale15 (50)Female15 (50)Tumor locationLeft lobe18 (60)Right lobe11 (36.7)Isthmus1 (3.3)Tumor size (cm)≤ 23 (10)2–415 (50)> 47 (23.3)Unknown5 (16.7)Lymph node metastasisPresent15 (50.0)Absent14 (46.6)Unknown1 (3.3)Location of Lymph node(n = 15)Lateral7 (46.6)Central8 (53.4)Tumor extensionPresent21 (70)Absent7 (23.3)Unknown2 (6.7)STE (n = 21)RLN8 (38.1)Muscles6 (28.6)Esophagus3 (14.3)Great vessels4 (19)Trachea2 (9.5)Thymus1 (4.7)Parathyroid gland1 (4.7)Abbreviations: STE, site of tumor extension; RLN, recurrent laryngeal nerve\n\nPatient characteristics\nAbbreviations: STE, site of tumor extension; RLN, recurrent laryngeal nerve\n\nTable 2Treatment procedures and clinical outcomeNo. (%) of patientsType of surgeryR016 (53.3)R12 (6.6)R29 (30)Biopsy3 (10)IMRT dose (Gy)Median (range)60 (56–66)IMRT duration (days)Median (range)44.5 (39–67)Systemic therapyYes vs. No3 (10) vs. 27 (90)Follow-up time (months)Median (range)63.5 (21–161)Treatment responseCR26 (86.7)PR4 (13.3)Local recurrencePresent2 (93.3)Absent28 (6.7)Distant metastasisPresent5 (83.3)Absent25 (16.7)Abbreviations: R0, no residual tumor; R1, microscopic residual tumor; R2, macroscopic residual tumor; IMRT, intensity-modulated radiotherapy; CR, complete remission; PR, partial remission\n\nTreatment procedures and clinical outcome\nAbbreviations: R0, no residual tumor; R1, microscopic residual tumor; R2, macroscopic residual tumor; IMRT, intensity-modulated radiotherapy; CR, complete remission; PR, partial remission\nPathology and immunohistochemical (IHC) data were available for 29 out of 30 patients. Among these, 93.1% (27/29) expressed CD5, and 72.4% (21/29) expressed CD117 totally or partially. Instead, 72.4% (21/29) of the cases were negatively expressed for thyroid transcription factor 1 (TTF-1), which was the marker of thyroid follicular cells.", "Dose-volume histogram (DVH) statistics were showed in Table 3. The average volume of GTV and CTV were 131.3 cc (21.2-387.2) and 323 cc (92.4-1408.2), respectively. Rates of dose coverage were excellent for the target volume. The volume receiving less than 95% of the prescribed dose was 0.3% for GTV and 1.2% for CTV. The mean dose was 67.8 Gy for GTV and 62.7 Gy for CTV.\n\nTable 3Dose-volume histograms (DVHs) statistics for IMRTGTVAverage (range)CTVAverage (range)Volume (cc)131.3 (21.2-387.2)323 (92.4-1408.2)Maximum dose (Gy)70.9 (69.2–72.5)67.6 (63.5–72.5)Mean dose (Gy)67.8 (65.9–68.8)62.7 (57.7–66.8)Minimum dose (Gy)56.8 (9.9–65.6)42.1 (38.2–56.3)% volume receiving < 95% of the prescribed dose0.3 (0-1.7)1.2 (0–12)% volume receiving ≥ 100% of the prescribed dose92.1(47.5–100)94.0(77.6–99.7)% volume receiving ≥ 110% of the prescribed dose015.8 (0-67.1)IMRT, intensity-modulated radiotherapy; GTV, gross tumor volume; CTV, clinical target volume\n\nDose-volume histograms (DVHs) statistics for IMRT\nIMRT, intensity-modulated radiotherapy; GTV, gross tumor volume; CTV, clinical target volume", "The median follow-up time was 63.5 months (range 21–161 months). The 5-year LRFS, RRFS, DMFS, OS, and PFS were 100, 88.9, 78.9, 93.1, and 78.9%, respectively (Fig. 2). In the subgroup analysis, we divided patients with no lateral neck node metastasis (23 patients) into two groups: group A (6 patients) with prophylactic irradiation for lateral neck nodal regions (levels II-V), while group B (17 patients) without prophylactic irradiation for lateral neck nodal regions. The results showed that there was no significant difference in RRFS (p = 0.381) and OS (p = 0.153) between the two groups (Fig. 3). Patient characteristics for groups A and B were shown in Supplementary Table 1.\n\nFig. 2Kaplan-Meier curves showing local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastases-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) for patients with carcinoma showing thymus-like differentiation\n\nKaplan-Meier curves showing local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastases-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) for patients with carcinoma showing thymus-like differentiation\n\nFig. 3Kaplan-Meier estimate of (a) regional recurrence-free survival (RRFS), and (b) overall survival (OS) between patients with or without irradiation for lateral neck nodal regions RT, radiotherapy; LR: lateral neck nodal regions\n\nKaplan-Meier estimate of (a) regional recurrence-free survival (RRFS), and (b) overall survival (OS) between patients with or without irradiation for lateral neck nodal regions\n RT, radiotherapy; LR: lateral neck nodal regions", "Two patients had regional lymph node recurrence. All the recurrences were in the contralateral supraclavicular fossa and were defined as type E (extraneous) failures. Distant metastasis was found in 5 patients. Common sites for distant metastasis were lung (3 patients), bone (1 patient), and liver (1 patient). Two patients suffered both regional and distant failure.\nSalvage treatment, including surgery, radiotherapy, and systemic therapy, was given to patients with recurrence or distant metastases. If acceptable, clinical trials were the first choice. For patients with oligometastatic or oligoprogressive tumors, SBRT was implemented.", "The majority of acute toxicities related to IMRT were grade 1–2. Grade 3 mucositis or dermatitis occurred only in 2 patients. No grade 4 toxicities were observed. Details were showed in Table 4. Three patients suffered treatment interruption due to other diseases, including aspiration pneumonia and herpes zoster. After appropriate treatment, they continued and finished IMRT. There was no late toxicity (≥ grade 2) related to IMRT, such as radiation pneumonitis, neck fibrosis, or esophageal stenosis.\n\nTable 4Acute toxicities during IMRTToxicitiesNo. of patients by toxicity grade (%)Grade 1Grade 2Grade 3Grade 4Grade 5Mucositis9 (30)20 (66.7)1 (3.3)00Dermatitis25 (83.3)4 (13.3)1 (3.3)00Skin pigmentation26 (86.7)0000Anemia4 (13.3)1 (3.3)000Leukopenia5 (16.7)1 (3.3)000Thrombocytopenia3 (10)0000IMRT, intensity-modulated radiotherapy\n\nAcute toxicities during IMRT\nIMRT, intensity-modulated radiotherapy", "Univariate analysis, taking age, gender, tumor size, tumor extension, surgery type, and lymph node metastasis as prognostic factors for RRFS, DMFS, OS, and PFS, was performed. Univariate analysis showed that surgery type (p = 0.026, 0.000, 0.026) and lateral neck node metastasis (p = 0.024, 0.009, 0.024) were potential prognostic factors for DMFS, OS and PFS (Supplementary Table 2). However, the multivariate analysis failed to further validate the differences (Supplementary Table 3). We attributed it to the relatively small sample size and few end-point events.", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Patients", "Intensity-modulate radiotherapy", "Patient evaluation", "Definition of failure pattern", "Statistical methods", "Results", "Patient characteristics", "Dosimetric data for IMRT", "Survival outcome", "Failure patterns and salvage treatment", "Treatment complications", "Prognostic factors", "Discussion", "Conclusion", "Electronic supplementary material", "" ]
[ "Carcinoma showing thymus-like differentiation (CASTLE) is a rare malignant tumor of the thyroid or adjacent soft tissue in the neck, which accounts for only 0.1–0.15% of all thyroid cancers [1–4]. Until now, fewer than 100 patients have been reported, and most of them are case reports. Optimal treatment for CASTLE remains uncertain. Surgery is generally recommended as the mainstay of treatment [3, 5]. However, due to the lower location and high possibility of extrathyroid invasion, complete resection can be challenging for patients with locally advanced disease [6]. It is reported that the incidence of lymph node metastasis is 50–69% and 60–80% for extrathyroid invasion [5–9]. Therefore, radiotherapy is often used as part of the treatment and the efficiency of radiotherapy has been proved by several studies [6, 7, 10, 11].\nHowever, there is no consensus on the delineation of target volume for CASTLE since very limited data has been published. Accurate target volume delineation is the premise of intensity-modulated radiotherapy (IMRT). Excessive irradiation will increase treatment-related toxicities like dermatitis and neck fibrosis, while insufficiency of target volume may cause tumor recurrence. Hence, in the present study, we reviewed our 11 years of clinical experience, analyzed the failure patterns, and gave suggestions for target volume delineation of CASTLE treated with IMRT. To the best of our knowledge, this is the first study focusing on target volume delineation for this rare disease.", "[SUBTITLE] Patients [SUBSECTION] From April 2008 to May 2019, 30 patients with CASTLE treated by postoperative or radical IMRT in our center were retrospectively reviewed. We collected clinicopathological data, treatment procedures, and clinical outcomes. The patterns of treatment failure were also analyzed. The study was approved by the Institutional Review Board of Fudan University Shanghai Cancer Center. Informed consent was obtained from all patients.\nFrom April 2008 to May 2019, 30 patients with CASTLE treated by postoperative or radical IMRT in our center were retrospectively reviewed. We collected clinicopathological data, treatment procedures, and clinical outcomes. The patterns of treatment failure were also analyzed. The study was approved by the Institutional Review Board of Fudan University Shanghai Cancer Center. Informed consent was obtained from all patients.\n[SUBTITLE] Intensity-modulate radiotherapy [SUBSECTION] IMRT was started 4 to 6 weeks after surgery or early after diagnosis. The techniques of IMRT were detailed in our previously published data [7]. Briefly, a thermoplastic mask of the head and shoulder was used for patient immobilization. Patients received computed tomography (CT) simulation at 5 mm thickness of the head and neck region in the supine position. Image fusion of magnetic resonance imaging (MRI) and CT was recommended for target volume delineation. The gross tumor volume (GTV) was defined as all primary gross tumors and involved lymph nodes determined by imaging and clinical findings. The clinical target volume (CTV) included the GTV or tumor bed plus a 5 to 10 mm margin to encompass any microscopic extension. The planning target volume (PTV) was defined as the CTV plus a 3 to 5 mm margin to encompass setup error. Neck nodal level VI was conventionally included in the CTV. If there was no positive lymph node, neck nodal levels II-V were not included in the CTV. Examples of target delineation were showed in Fig. 1. Positive lymph nodes were defined as follows: (1) pathologically diagnosed; (2) minimum axial diameter ≥ 1 cm; (3) extranodal extension or circular enhancement. The prescribed dose was 60 Gy for patients without residual disease and 66 Gy for patients with residual or unresectable disease. Conventional fractionation (2 Gy per fraction, one fraction per day, five days per week) was used.\n\nFig. 1Examples of target volume delineation for carcinoma showing thymus-like differentiation GTV, gross tumor volume; PTV-G, planning target volume for GTV; CTV, clinical target volume; PTV-C, planning target volume for CTV.\n\nExamples of target volume delineation for carcinoma showing thymus-like differentiation\n GTV, gross tumor volume; PTV-G, planning target volume for GTV; CTV, clinical target volume;\n PTV-C, planning target volume for CTV.\nIMRT was started 4 to 6 weeks after surgery or early after diagnosis. The techniques of IMRT were detailed in our previously published data [7]. Briefly, a thermoplastic mask of the head and shoulder was used for patient immobilization. Patients received computed tomography (CT) simulation at 5 mm thickness of the head and neck region in the supine position. Image fusion of magnetic resonance imaging (MRI) and CT was recommended for target volume delineation. The gross tumor volume (GTV) was defined as all primary gross tumors and involved lymph nodes determined by imaging and clinical findings. The clinical target volume (CTV) included the GTV or tumor bed plus a 5 to 10 mm margin to encompass any microscopic extension. The planning target volume (PTV) was defined as the CTV plus a 3 to 5 mm margin to encompass setup error. Neck nodal level VI was conventionally included in the CTV. If there was no positive lymph node, neck nodal levels II-V were not included in the CTV. Examples of target delineation were showed in Fig. 1. Positive lymph nodes were defined as follows: (1) pathologically diagnosed; (2) minimum axial diameter ≥ 1 cm; (3) extranodal extension or circular enhancement. The prescribed dose was 60 Gy for patients without residual disease and 66 Gy for patients with residual or unresectable disease. Conventional fractionation (2 Gy per fraction, one fraction per day, five days per week) was used.\n\nFig. 1Examples of target volume delineation for carcinoma showing thymus-like differentiation GTV, gross tumor volume; PTV-G, planning target volume for GTV; CTV, clinical target volume; PTV-C, planning target volume for CTV.\n\nExamples of target volume delineation for carcinoma showing thymus-like differentiation\n GTV, gross tumor volume; PTV-G, planning target volume for GTV; CTV, clinical target volume;\n PTV-C, planning target volume for CTV.\n[SUBTITLE] Patient evaluation [SUBSECTION] Patients were assessed weekly during IMRT. After IMRT, patients were follow-up every three months in the first two years, every six months during the years 3–5, and annually after that. Follow-up assessments after treatment included examination of the neck, thyroid function tests, and ultrasound or MRI for the thyroid and neck. Chest CT scan, and ultrasound or CT of the abdomen were performed every 6–12 months. Additional tests were performed when clinically indicated.\nPatients were assessed weekly during IMRT. After IMRT, patients were follow-up every three months in the first two years, every six months during the years 3–5, and annually after that. Follow-up assessments after treatment included examination of the neck, thyroid function tests, and ultrasound or MRI for the thyroid and neck. Chest CT scan, and ultrasound or CT of the abdomen were performed every 6–12 months. Additional tests were performed when clinically indicated.\n[SUBTITLE] Definition of failure pattern [SUBSECTION] The images of MRI or CT scans obtained at the time of recurrence were transferred to the pretreatment planning CT. The dose-volume histogram (DVH) was used to calculate the radiation dose received by the recurrent tumor (GTVrecur) region. The recurrences were classified into five types based on combined spatial and dosimetric criteria [12, 13]:\nType A (central high dose): the mapped centroid of GTVrecur originates in high dose PTV, and ≥ 95% of GTVrecur was within the 95% isodose (high dose PTV);\nType B (peripheral high dose): the mapped centroid of GTVrecur originates in high dose PTV, and < 95% of GTVrecur was within the 95% isodose (high dose PTV);\nType C (central elective dose): the mapped centroid of GTVrecur originates in lower dose PTV, and ≥ 95% of GTVrecur was within the 95% isodose (lower dose PTV);\nType D (peripheral elective dose): the mapped centroid of GTVrecur originates in lower dose PTV, and < 95% of GTVrecur was within the 95% isodose (lower dose PTV);\nType E (extraneous dose): the mapped centroid of GTVrecur originates outside all PTVs.\nThe images of MRI or CT scans obtained at the time of recurrence were transferred to the pretreatment planning CT. The dose-volume histogram (DVH) was used to calculate the radiation dose received by the recurrent tumor (GTVrecur) region. The recurrences were classified into five types based on combined spatial and dosimetric criteria [12, 13]:\nType A (central high dose): the mapped centroid of GTVrecur originates in high dose PTV, and ≥ 95% of GTVrecur was within the 95% isodose (high dose PTV);\nType B (peripheral high dose): the mapped centroid of GTVrecur originates in high dose PTV, and < 95% of GTVrecur was within the 95% isodose (high dose PTV);\nType C (central elective dose): the mapped centroid of GTVrecur originates in lower dose PTV, and ≥ 95% of GTVrecur was within the 95% isodose (lower dose PTV);\nType D (peripheral elective dose): the mapped centroid of GTVrecur originates in lower dose PTV, and < 95% of GTVrecur was within the 95% isodose (lower dose PTV);\nType E (extraneous dose): the mapped centroid of GTVrecur originates outside all PTVs.\n[SUBTITLE] Statistical methods [SUBSECTION] All the statistical analysis was performed using the Statistical Package for Social Sciences (SPSS version 24.0) software. The survival rates were calculated from the day of the first treatment. The Kaplan-Meier method was used to calculate the local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastasis-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) rates. Treatment-related toxicities were graded by National Cancer Institute Common Toxicity Criteria (NCI-CTC) version 4.0.\nAll the statistical analysis was performed using the Statistical Package for Social Sciences (SPSS version 24.0) software. The survival rates were calculated from the day of the first treatment. The Kaplan-Meier method was used to calculate the local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastasis-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) rates. Treatment-related toxicities were graded by National Cancer Institute Common Toxicity Criteria (NCI-CTC) version 4.0.", "From April 2008 to May 2019, 30 patients with CASTLE treated by postoperative or radical IMRT in our center were retrospectively reviewed. We collected clinicopathological data, treatment procedures, and clinical outcomes. The patterns of treatment failure were also analyzed. The study was approved by the Institutional Review Board of Fudan University Shanghai Cancer Center. Informed consent was obtained from all patients.", "IMRT was started 4 to 6 weeks after surgery or early after diagnosis. The techniques of IMRT were detailed in our previously published data [7]. Briefly, a thermoplastic mask of the head and shoulder was used for patient immobilization. Patients received computed tomography (CT) simulation at 5 mm thickness of the head and neck region in the supine position. Image fusion of magnetic resonance imaging (MRI) and CT was recommended for target volume delineation. The gross tumor volume (GTV) was defined as all primary gross tumors and involved lymph nodes determined by imaging and clinical findings. The clinical target volume (CTV) included the GTV or tumor bed plus a 5 to 10 mm margin to encompass any microscopic extension. The planning target volume (PTV) was defined as the CTV plus a 3 to 5 mm margin to encompass setup error. Neck nodal level VI was conventionally included in the CTV. If there was no positive lymph node, neck nodal levels II-V were not included in the CTV. Examples of target delineation were showed in Fig. 1. Positive lymph nodes were defined as follows: (1) pathologically diagnosed; (2) minimum axial diameter ≥ 1 cm; (3) extranodal extension or circular enhancement. The prescribed dose was 60 Gy for patients without residual disease and 66 Gy for patients with residual or unresectable disease. Conventional fractionation (2 Gy per fraction, one fraction per day, five days per week) was used.\n\nFig. 1Examples of target volume delineation for carcinoma showing thymus-like differentiation GTV, gross tumor volume; PTV-G, planning target volume for GTV; CTV, clinical target volume; PTV-C, planning target volume for CTV.\n\nExamples of target volume delineation for carcinoma showing thymus-like differentiation\n GTV, gross tumor volume; PTV-G, planning target volume for GTV; CTV, clinical target volume;\n PTV-C, planning target volume for CTV.", "Patients were assessed weekly during IMRT. After IMRT, patients were follow-up every three months in the first two years, every six months during the years 3–5, and annually after that. Follow-up assessments after treatment included examination of the neck, thyroid function tests, and ultrasound or MRI for the thyroid and neck. Chest CT scan, and ultrasound or CT of the abdomen were performed every 6–12 months. Additional tests were performed when clinically indicated.", "The images of MRI or CT scans obtained at the time of recurrence were transferred to the pretreatment planning CT. The dose-volume histogram (DVH) was used to calculate the radiation dose received by the recurrent tumor (GTVrecur) region. The recurrences were classified into five types based on combined spatial and dosimetric criteria [12, 13]:\nType A (central high dose): the mapped centroid of GTVrecur originates in high dose PTV, and ≥ 95% of GTVrecur was within the 95% isodose (high dose PTV);\nType B (peripheral high dose): the mapped centroid of GTVrecur originates in high dose PTV, and < 95% of GTVrecur was within the 95% isodose (high dose PTV);\nType C (central elective dose): the mapped centroid of GTVrecur originates in lower dose PTV, and ≥ 95% of GTVrecur was within the 95% isodose (lower dose PTV);\nType D (peripheral elective dose): the mapped centroid of GTVrecur originates in lower dose PTV, and < 95% of GTVrecur was within the 95% isodose (lower dose PTV);\nType E (extraneous dose): the mapped centroid of GTVrecur originates outside all PTVs.", "All the statistical analysis was performed using the Statistical Package for Social Sciences (SPSS version 24.0) software. The survival rates were calculated from the day of the first treatment. The Kaplan-Meier method was used to calculate the local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastasis-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) rates. Treatment-related toxicities were graded by National Cancer Institute Common Toxicity Criteria (NCI-CTC) version 4.0.", "[SUBTITLE] Patient characteristics [SUBSECTION] A total of 30 patients were enrolled in this study. Patients’ characteristics were summarized in Table 1. The median age was 53 years (range 37–61 years). The ratio of male to female was 1:1. Lymph node metastasis was found in 50% of the patients, among which 53.4% were central and 46.6% were lateral. Tumor extension to adjacent organs, including recurrent laryngeal nerve, great vessels, muscles, esophagus, or trachea was found in 21 (70%) patients. About 53% of the patients received complete resection, and 40% received partial resection or biopsy only. Two patients received cisplatin-based concurrent chemotherapy. After treatment, complete remission (CR) was achieved in 26 (86.7%) patients, and partial remission (PR) in 4 (13.3%) patients. Treatment procedures are detailed in Table 2.\n\nTable 1Patient characteristicsNo. (%) of patientsTotal30Median age (range)53 (37–61)GenderMale15 (50)Female15 (50)Tumor locationLeft lobe18 (60)Right lobe11 (36.7)Isthmus1 (3.3)Tumor size (cm)≤ 23 (10)2–415 (50)> 47 (23.3)Unknown5 (16.7)Lymph node metastasisPresent15 (50.0)Absent14 (46.6)Unknown1 (3.3)Location of Lymph node(n = 15)Lateral7 (46.6)Central8 (53.4)Tumor extensionPresent21 (70)Absent7 (23.3)Unknown2 (6.7)STE (n = 21)RLN8 (38.1)Muscles6 (28.6)Esophagus3 (14.3)Great vessels4 (19)Trachea2 (9.5)Thymus1 (4.7)Parathyroid gland1 (4.7)Abbreviations: STE, site of tumor extension; RLN, recurrent laryngeal nerve\n\nPatient characteristics\nAbbreviations: STE, site of tumor extension; RLN, recurrent laryngeal nerve\n\nTable 2Treatment procedures and clinical outcomeNo. (%) of patientsType of surgeryR016 (53.3)R12 (6.6)R29 (30)Biopsy3 (10)IMRT dose (Gy)Median (range)60 (56–66)IMRT duration (days)Median (range)44.5 (39–67)Systemic therapyYes vs. No3 (10) vs. 27 (90)Follow-up time (months)Median (range)63.5 (21–161)Treatment responseCR26 (86.7)PR4 (13.3)Local recurrencePresent2 (93.3)Absent28 (6.7)Distant metastasisPresent5 (83.3)Absent25 (16.7)Abbreviations: R0, no residual tumor; R1, microscopic residual tumor; R2, macroscopic residual tumor; IMRT, intensity-modulated radiotherapy; CR, complete remission; PR, partial remission\n\nTreatment procedures and clinical outcome\nAbbreviations: R0, no residual tumor; R1, microscopic residual tumor; R2, macroscopic residual tumor; IMRT, intensity-modulated radiotherapy; CR, complete remission; PR, partial remission\nPathology and immunohistochemical (IHC) data were available for 29 out of 30 patients. Among these, 93.1% (27/29) expressed CD5, and 72.4% (21/29) expressed CD117 totally or partially. Instead, 72.4% (21/29) of the cases were negatively expressed for thyroid transcription factor 1 (TTF-1), which was the marker of thyroid follicular cells.\nA total of 30 patients were enrolled in this study. Patients’ characteristics were summarized in Table 1. The median age was 53 years (range 37–61 years). The ratio of male to female was 1:1. Lymph node metastasis was found in 50% of the patients, among which 53.4% were central and 46.6% were lateral. Tumor extension to adjacent organs, including recurrent laryngeal nerve, great vessels, muscles, esophagus, or trachea was found in 21 (70%) patients. About 53% of the patients received complete resection, and 40% received partial resection or biopsy only. Two patients received cisplatin-based concurrent chemotherapy. After treatment, complete remission (CR) was achieved in 26 (86.7%) patients, and partial remission (PR) in 4 (13.3%) patients. Treatment procedures are detailed in Table 2.\n\nTable 1Patient characteristicsNo. (%) of patientsTotal30Median age (range)53 (37–61)GenderMale15 (50)Female15 (50)Tumor locationLeft lobe18 (60)Right lobe11 (36.7)Isthmus1 (3.3)Tumor size (cm)≤ 23 (10)2–415 (50)> 47 (23.3)Unknown5 (16.7)Lymph node metastasisPresent15 (50.0)Absent14 (46.6)Unknown1 (3.3)Location of Lymph node(n = 15)Lateral7 (46.6)Central8 (53.4)Tumor extensionPresent21 (70)Absent7 (23.3)Unknown2 (6.7)STE (n = 21)RLN8 (38.1)Muscles6 (28.6)Esophagus3 (14.3)Great vessels4 (19)Trachea2 (9.5)Thymus1 (4.7)Parathyroid gland1 (4.7)Abbreviations: STE, site of tumor extension; RLN, recurrent laryngeal nerve\n\nPatient characteristics\nAbbreviations: STE, site of tumor extension; RLN, recurrent laryngeal nerve\n\nTable 2Treatment procedures and clinical outcomeNo. (%) of patientsType of surgeryR016 (53.3)R12 (6.6)R29 (30)Biopsy3 (10)IMRT dose (Gy)Median (range)60 (56–66)IMRT duration (days)Median (range)44.5 (39–67)Systemic therapyYes vs. No3 (10) vs. 27 (90)Follow-up time (months)Median (range)63.5 (21–161)Treatment responseCR26 (86.7)PR4 (13.3)Local recurrencePresent2 (93.3)Absent28 (6.7)Distant metastasisPresent5 (83.3)Absent25 (16.7)Abbreviations: R0, no residual tumor; R1, microscopic residual tumor; R2, macroscopic residual tumor; IMRT, intensity-modulated radiotherapy; CR, complete remission; PR, partial remission\n\nTreatment procedures and clinical outcome\nAbbreviations: R0, no residual tumor; R1, microscopic residual tumor; R2, macroscopic residual tumor; IMRT, intensity-modulated radiotherapy; CR, complete remission; PR, partial remission\nPathology and immunohistochemical (IHC) data were available for 29 out of 30 patients. Among these, 93.1% (27/29) expressed CD5, and 72.4% (21/29) expressed CD117 totally or partially. Instead, 72.4% (21/29) of the cases were negatively expressed for thyroid transcription factor 1 (TTF-1), which was the marker of thyroid follicular cells.\n[SUBTITLE] Dosimetric data for IMRT [SUBSECTION] Dose-volume histogram (DVH) statistics were showed in Table 3. The average volume of GTV and CTV were 131.3 cc (21.2-387.2) and 323 cc (92.4-1408.2), respectively. Rates of dose coverage were excellent for the target volume. The volume receiving less than 95% of the prescribed dose was 0.3% for GTV and 1.2% for CTV. The mean dose was 67.8 Gy for GTV and 62.7 Gy for CTV.\n\nTable 3Dose-volume histograms (DVHs) statistics for IMRTGTVAverage (range)CTVAverage (range)Volume (cc)131.3 (21.2-387.2)323 (92.4-1408.2)Maximum dose (Gy)70.9 (69.2–72.5)67.6 (63.5–72.5)Mean dose (Gy)67.8 (65.9–68.8)62.7 (57.7–66.8)Minimum dose (Gy)56.8 (9.9–65.6)42.1 (38.2–56.3)% volume receiving < 95% of the prescribed dose0.3 (0-1.7)1.2 (0–12)% volume receiving ≥ 100% of the prescribed dose92.1(47.5–100)94.0(77.6–99.7)% volume receiving ≥ 110% of the prescribed dose015.8 (0-67.1)IMRT, intensity-modulated radiotherapy; GTV, gross tumor volume; CTV, clinical target volume\n\nDose-volume histograms (DVHs) statistics for IMRT\nIMRT, intensity-modulated radiotherapy; GTV, gross tumor volume; CTV, clinical target volume\nDose-volume histogram (DVH) statistics were showed in Table 3. The average volume of GTV and CTV were 131.3 cc (21.2-387.2) and 323 cc (92.4-1408.2), respectively. Rates of dose coverage were excellent for the target volume. The volume receiving less than 95% of the prescribed dose was 0.3% for GTV and 1.2% for CTV. The mean dose was 67.8 Gy for GTV and 62.7 Gy for CTV.\n\nTable 3Dose-volume histograms (DVHs) statistics for IMRTGTVAverage (range)CTVAverage (range)Volume (cc)131.3 (21.2-387.2)323 (92.4-1408.2)Maximum dose (Gy)70.9 (69.2–72.5)67.6 (63.5–72.5)Mean dose (Gy)67.8 (65.9–68.8)62.7 (57.7–66.8)Minimum dose (Gy)56.8 (9.9–65.6)42.1 (38.2–56.3)% volume receiving < 95% of the prescribed dose0.3 (0-1.7)1.2 (0–12)% volume receiving ≥ 100% of the prescribed dose92.1(47.5–100)94.0(77.6–99.7)% volume receiving ≥ 110% of the prescribed dose015.8 (0-67.1)IMRT, intensity-modulated radiotherapy; GTV, gross tumor volume; CTV, clinical target volume\n\nDose-volume histograms (DVHs) statistics for IMRT\nIMRT, intensity-modulated radiotherapy; GTV, gross tumor volume; CTV, clinical target volume\n[SUBTITLE] Survival outcome [SUBSECTION] The median follow-up time was 63.5 months (range 21–161 months). The 5-year LRFS, RRFS, DMFS, OS, and PFS were 100, 88.9, 78.9, 93.1, and 78.9%, respectively (Fig. 2). In the subgroup analysis, we divided patients with no lateral neck node metastasis (23 patients) into two groups: group A (6 patients) with prophylactic irradiation for lateral neck nodal regions (levels II-V), while group B (17 patients) without prophylactic irradiation for lateral neck nodal regions. The results showed that there was no significant difference in RRFS (p = 0.381) and OS (p = 0.153) between the two groups (Fig. 3). Patient characteristics for groups A and B were shown in Supplementary Table 1.\n\nFig. 2Kaplan-Meier curves showing local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastases-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) for patients with carcinoma showing thymus-like differentiation\n\nKaplan-Meier curves showing local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastases-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) for patients with carcinoma showing thymus-like differentiation\n\nFig. 3Kaplan-Meier estimate of (a) regional recurrence-free survival (RRFS), and (b) overall survival (OS) between patients with or without irradiation for lateral neck nodal regions RT, radiotherapy; LR: lateral neck nodal regions\n\nKaplan-Meier estimate of (a) regional recurrence-free survival (RRFS), and (b) overall survival (OS) between patients with or without irradiation for lateral neck nodal regions\n RT, radiotherapy; LR: lateral neck nodal regions\nThe median follow-up time was 63.5 months (range 21–161 months). The 5-year LRFS, RRFS, DMFS, OS, and PFS were 100, 88.9, 78.9, 93.1, and 78.9%, respectively (Fig. 2). In the subgroup analysis, we divided patients with no lateral neck node metastasis (23 patients) into two groups: group A (6 patients) with prophylactic irradiation for lateral neck nodal regions (levels II-V), while group B (17 patients) without prophylactic irradiation for lateral neck nodal regions. The results showed that there was no significant difference in RRFS (p = 0.381) and OS (p = 0.153) between the two groups (Fig. 3). Patient characteristics for groups A and B were shown in Supplementary Table 1.\n\nFig. 2Kaplan-Meier curves showing local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastases-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) for patients with carcinoma showing thymus-like differentiation\n\nKaplan-Meier curves showing local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastases-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) for patients with carcinoma showing thymus-like differentiation\n\nFig. 3Kaplan-Meier estimate of (a) regional recurrence-free survival (RRFS), and (b) overall survival (OS) between patients with or without irradiation for lateral neck nodal regions RT, radiotherapy; LR: lateral neck nodal regions\n\nKaplan-Meier estimate of (a) regional recurrence-free survival (RRFS), and (b) overall survival (OS) between patients with or without irradiation for lateral neck nodal regions\n RT, radiotherapy; LR: lateral neck nodal regions\n[SUBTITLE] Failure patterns and salvage treatment [SUBSECTION] Two patients had regional lymph node recurrence. All the recurrences were in the contralateral supraclavicular fossa and were defined as type E (extraneous) failures. Distant metastasis was found in 5 patients. Common sites for distant metastasis were lung (3 patients), bone (1 patient), and liver (1 patient). Two patients suffered both regional and distant failure.\nSalvage treatment, including surgery, radiotherapy, and systemic therapy, was given to patients with recurrence or distant metastases. If acceptable, clinical trials were the first choice. For patients with oligometastatic or oligoprogressive tumors, SBRT was implemented.\nTwo patients had regional lymph node recurrence. All the recurrences were in the contralateral supraclavicular fossa and were defined as type E (extraneous) failures. Distant metastasis was found in 5 patients. Common sites for distant metastasis were lung (3 patients), bone (1 patient), and liver (1 patient). Two patients suffered both regional and distant failure.\nSalvage treatment, including surgery, radiotherapy, and systemic therapy, was given to patients with recurrence or distant metastases. If acceptable, clinical trials were the first choice. For patients with oligometastatic or oligoprogressive tumors, SBRT was implemented.\n[SUBTITLE] Treatment complications [SUBSECTION] The majority of acute toxicities related to IMRT were grade 1–2. Grade 3 mucositis or dermatitis occurred only in 2 patients. No grade 4 toxicities were observed. Details were showed in Table 4. Three patients suffered treatment interruption due to other diseases, including aspiration pneumonia and herpes zoster. After appropriate treatment, they continued and finished IMRT. There was no late toxicity (≥ grade 2) related to IMRT, such as radiation pneumonitis, neck fibrosis, or esophageal stenosis.\n\nTable 4Acute toxicities during IMRTToxicitiesNo. of patients by toxicity grade (%)Grade 1Grade 2Grade 3Grade 4Grade 5Mucositis9 (30)20 (66.7)1 (3.3)00Dermatitis25 (83.3)4 (13.3)1 (3.3)00Skin pigmentation26 (86.7)0000Anemia4 (13.3)1 (3.3)000Leukopenia5 (16.7)1 (3.3)000Thrombocytopenia3 (10)0000IMRT, intensity-modulated radiotherapy\n\nAcute toxicities during IMRT\nIMRT, intensity-modulated radiotherapy\nThe majority of acute toxicities related to IMRT were grade 1–2. Grade 3 mucositis or dermatitis occurred only in 2 patients. No grade 4 toxicities were observed. Details were showed in Table 4. Three patients suffered treatment interruption due to other diseases, including aspiration pneumonia and herpes zoster. After appropriate treatment, they continued and finished IMRT. There was no late toxicity (≥ grade 2) related to IMRT, such as radiation pneumonitis, neck fibrosis, or esophageal stenosis.\n\nTable 4Acute toxicities during IMRTToxicitiesNo. of patients by toxicity grade (%)Grade 1Grade 2Grade 3Grade 4Grade 5Mucositis9 (30)20 (66.7)1 (3.3)00Dermatitis25 (83.3)4 (13.3)1 (3.3)00Skin pigmentation26 (86.7)0000Anemia4 (13.3)1 (3.3)000Leukopenia5 (16.7)1 (3.3)000Thrombocytopenia3 (10)0000IMRT, intensity-modulated radiotherapy\n\nAcute toxicities during IMRT\nIMRT, intensity-modulated radiotherapy\n[SUBTITLE] Prognostic factors [SUBSECTION] Univariate analysis, taking age, gender, tumor size, tumor extension, surgery type, and lymph node metastasis as prognostic factors for RRFS, DMFS, OS, and PFS, was performed. Univariate analysis showed that surgery type (p = 0.026, 0.000, 0.026) and lateral neck node metastasis (p = 0.024, 0.009, 0.024) were potential prognostic factors for DMFS, OS and PFS (Supplementary Table 2). However, the multivariate analysis failed to further validate the differences (Supplementary Table 3). We attributed it to the relatively small sample size and few end-point events.\nUnivariate analysis, taking age, gender, tumor size, tumor extension, surgery type, and lymph node metastasis as prognostic factors for RRFS, DMFS, OS, and PFS, was performed. Univariate analysis showed that surgery type (p = 0.026, 0.000, 0.026) and lateral neck node metastasis (p = 0.024, 0.009, 0.024) were potential prognostic factors for DMFS, OS and PFS (Supplementary Table 2). However, the multivariate analysis failed to further validate the differences (Supplementary Table 3). We attributed it to the relatively small sample size and few end-point events.", "A total of 30 patients were enrolled in this study. Patients’ characteristics were summarized in Table 1. The median age was 53 years (range 37–61 years). The ratio of male to female was 1:1. Lymph node metastasis was found in 50% of the patients, among which 53.4% were central and 46.6% were lateral. Tumor extension to adjacent organs, including recurrent laryngeal nerve, great vessels, muscles, esophagus, or trachea was found in 21 (70%) patients. About 53% of the patients received complete resection, and 40% received partial resection or biopsy only. Two patients received cisplatin-based concurrent chemotherapy. After treatment, complete remission (CR) was achieved in 26 (86.7%) patients, and partial remission (PR) in 4 (13.3%) patients. Treatment procedures are detailed in Table 2.\n\nTable 1Patient characteristicsNo. (%) of patientsTotal30Median age (range)53 (37–61)GenderMale15 (50)Female15 (50)Tumor locationLeft lobe18 (60)Right lobe11 (36.7)Isthmus1 (3.3)Tumor size (cm)≤ 23 (10)2–415 (50)> 47 (23.3)Unknown5 (16.7)Lymph node metastasisPresent15 (50.0)Absent14 (46.6)Unknown1 (3.3)Location of Lymph node(n = 15)Lateral7 (46.6)Central8 (53.4)Tumor extensionPresent21 (70)Absent7 (23.3)Unknown2 (6.7)STE (n = 21)RLN8 (38.1)Muscles6 (28.6)Esophagus3 (14.3)Great vessels4 (19)Trachea2 (9.5)Thymus1 (4.7)Parathyroid gland1 (4.7)Abbreviations: STE, site of tumor extension; RLN, recurrent laryngeal nerve\n\nPatient characteristics\nAbbreviations: STE, site of tumor extension; RLN, recurrent laryngeal nerve\n\nTable 2Treatment procedures and clinical outcomeNo. (%) of patientsType of surgeryR016 (53.3)R12 (6.6)R29 (30)Biopsy3 (10)IMRT dose (Gy)Median (range)60 (56–66)IMRT duration (days)Median (range)44.5 (39–67)Systemic therapyYes vs. No3 (10) vs. 27 (90)Follow-up time (months)Median (range)63.5 (21–161)Treatment responseCR26 (86.7)PR4 (13.3)Local recurrencePresent2 (93.3)Absent28 (6.7)Distant metastasisPresent5 (83.3)Absent25 (16.7)Abbreviations: R0, no residual tumor; R1, microscopic residual tumor; R2, macroscopic residual tumor; IMRT, intensity-modulated radiotherapy; CR, complete remission; PR, partial remission\n\nTreatment procedures and clinical outcome\nAbbreviations: R0, no residual tumor; R1, microscopic residual tumor; R2, macroscopic residual tumor; IMRT, intensity-modulated radiotherapy; CR, complete remission; PR, partial remission\nPathology and immunohistochemical (IHC) data were available for 29 out of 30 patients. Among these, 93.1% (27/29) expressed CD5, and 72.4% (21/29) expressed CD117 totally or partially. Instead, 72.4% (21/29) of the cases were negatively expressed for thyroid transcription factor 1 (TTF-1), which was the marker of thyroid follicular cells.", "Dose-volume histogram (DVH) statistics were showed in Table 3. The average volume of GTV and CTV were 131.3 cc (21.2-387.2) and 323 cc (92.4-1408.2), respectively. Rates of dose coverage were excellent for the target volume. The volume receiving less than 95% of the prescribed dose was 0.3% for GTV and 1.2% for CTV. The mean dose was 67.8 Gy for GTV and 62.7 Gy for CTV.\n\nTable 3Dose-volume histograms (DVHs) statistics for IMRTGTVAverage (range)CTVAverage (range)Volume (cc)131.3 (21.2-387.2)323 (92.4-1408.2)Maximum dose (Gy)70.9 (69.2–72.5)67.6 (63.5–72.5)Mean dose (Gy)67.8 (65.9–68.8)62.7 (57.7–66.8)Minimum dose (Gy)56.8 (9.9–65.6)42.1 (38.2–56.3)% volume receiving < 95% of the prescribed dose0.3 (0-1.7)1.2 (0–12)% volume receiving ≥ 100% of the prescribed dose92.1(47.5–100)94.0(77.6–99.7)% volume receiving ≥ 110% of the prescribed dose015.8 (0-67.1)IMRT, intensity-modulated radiotherapy; GTV, gross tumor volume; CTV, clinical target volume\n\nDose-volume histograms (DVHs) statistics for IMRT\nIMRT, intensity-modulated radiotherapy; GTV, gross tumor volume; CTV, clinical target volume", "The median follow-up time was 63.5 months (range 21–161 months). The 5-year LRFS, RRFS, DMFS, OS, and PFS were 100, 88.9, 78.9, 93.1, and 78.9%, respectively (Fig. 2). In the subgroup analysis, we divided patients with no lateral neck node metastasis (23 patients) into two groups: group A (6 patients) with prophylactic irradiation for lateral neck nodal regions (levels II-V), while group B (17 patients) without prophylactic irradiation for lateral neck nodal regions. The results showed that there was no significant difference in RRFS (p = 0.381) and OS (p = 0.153) between the two groups (Fig. 3). Patient characteristics for groups A and B were shown in Supplementary Table 1.\n\nFig. 2Kaplan-Meier curves showing local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastases-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) for patients with carcinoma showing thymus-like differentiation\n\nKaplan-Meier curves showing local recurrence-free survival (LRFS), regional recurrence-free survival (RRFS), distant metastases-free survival (DMFS), overall survival (OS), and progression-free survival (PFS) for patients with carcinoma showing thymus-like differentiation\n\nFig. 3Kaplan-Meier estimate of (a) regional recurrence-free survival (RRFS), and (b) overall survival (OS) between patients with or without irradiation for lateral neck nodal regions RT, radiotherapy; LR: lateral neck nodal regions\n\nKaplan-Meier estimate of (a) regional recurrence-free survival (RRFS), and (b) overall survival (OS) between patients with or without irradiation for lateral neck nodal regions\n RT, radiotherapy; LR: lateral neck nodal regions", "Two patients had regional lymph node recurrence. All the recurrences were in the contralateral supraclavicular fossa and were defined as type E (extraneous) failures. Distant metastasis was found in 5 patients. Common sites for distant metastasis were lung (3 patients), bone (1 patient), and liver (1 patient). Two patients suffered both regional and distant failure.\nSalvage treatment, including surgery, radiotherapy, and systemic therapy, was given to patients with recurrence or distant metastases. If acceptable, clinical trials were the first choice. For patients with oligometastatic or oligoprogressive tumors, SBRT was implemented.", "The majority of acute toxicities related to IMRT were grade 1–2. Grade 3 mucositis or dermatitis occurred only in 2 patients. No grade 4 toxicities were observed. Details were showed in Table 4. Three patients suffered treatment interruption due to other diseases, including aspiration pneumonia and herpes zoster. After appropriate treatment, they continued and finished IMRT. There was no late toxicity (≥ grade 2) related to IMRT, such as radiation pneumonitis, neck fibrosis, or esophageal stenosis.\n\nTable 4Acute toxicities during IMRTToxicitiesNo. of patients by toxicity grade (%)Grade 1Grade 2Grade 3Grade 4Grade 5Mucositis9 (30)20 (66.7)1 (3.3)00Dermatitis25 (83.3)4 (13.3)1 (3.3)00Skin pigmentation26 (86.7)0000Anemia4 (13.3)1 (3.3)000Leukopenia5 (16.7)1 (3.3)000Thrombocytopenia3 (10)0000IMRT, intensity-modulated radiotherapy\n\nAcute toxicities during IMRT\nIMRT, intensity-modulated radiotherapy", "Univariate analysis, taking age, gender, tumor size, tumor extension, surgery type, and lymph node metastasis as prognostic factors for RRFS, DMFS, OS, and PFS, was performed. Univariate analysis showed that surgery type (p = 0.026, 0.000, 0.026) and lateral neck node metastasis (p = 0.024, 0.009, 0.024) were potential prognostic factors for DMFS, OS and PFS (Supplementary Table 2). However, the multivariate analysis failed to further validate the differences (Supplementary Table 3). We attributed it to the relatively small sample size and few end-point events.", "CASTLE is an extremely rare malignant tumor, which is thought to arise from ectopic thymus or branchial pouch remnants [14]. The morphological, immunohistochemical, and molecular features of CASTLE are similar to those of thymic carcinoma. The carcinogenesis of CASTLE is the result of combined actions by a series of oncogenes and tumor suppressors. The study from Wang et al. [15] indicated that abnormal expression of p16, Bcl-2, p53, E-cadherin, C-KIT, CA-IX, EGFR, and HER-2 might play a role in the tumorigenesis and development of CASTLE. Patients with HER-2 overexpression showed a worse prognosis, suggesting that HER-2 overexpression may enhance the invasive and metastatic potential of CASTLE. However, the molecular pathological mechanism of CASTLE is still unclear. Further studies are needed to explore the molecular networks of this rare disease.\nAlthough the optimal treatment modality for CASTLE remains uncertain, an increasing number of studies have indicated the important role of radiotherapy in treating CASTLE. Due to the high possibility of extrathyroid invasion and high incidence of lymph node metastasis, adjuvant radiotherapy is often used as part of the treatment [5–9]. And growing evidence has validated the efficiency of adjuvant radiotherapy in recent years [5–7, 10, 11]. Choi et al. [10] reported that adjuvant radiotherapy reduced about 43% of the recurrence for patients with positive lymph nodes. In the study by Gao et al. [6], the median survival time was significantly longer in the surgery and adjuvant radiotherapy group than in the surgery alone group (17.1 vs. 8.8 years, p = 0.034). Moreover, radiotherapy can also play an essential role in tumor control for patients with unresectable disease. Petra et al. [16] recently reported a patient with locally advanced disease which was not suitable for surgery and thus underwent radical IMRT alone. Exciting complete remission was achieved after IMRT at a dose of 70 Gy. In the present study, palliative resection was administered to one of the patients due to the tumor invasion of adjacent great vessels. Then the patient received radical IMRT at a dose of 66 Gy in 33 fractions. Partial remission was achieved. Seven years after IMRT, the patient is still alive with stable disease at our last follow-up. All the above implies the indispensable role of radiotherapy in treating CASTLE.\nThere is no consensus on target volume delineation for CASTLE. Most of the previous studies reported the radiation dose only and with no specification of the target volume. To the best of our knowledge, this is the first study to explore the proper target volume delineation for this rare disease. As we know, neck nodal level VI is the sentinal node for thyroid tumors. Excessive irradiation for the neck can induce dermatitis, especially for the lower neck (as most of the CASTLE arises in the lower part of the thyroid lobe). Severe dermatitis may cause the interruption of radiotherapy and influence the treatment effect. After treatment, it may also cause neck fibrosis, which will seriously affect patients’ long-term quality of life. Hence, in the present study, we compared the effects of prophylactic irradiation and omitting irradiation to lateral neck regions on the survival rates of patients with no lateral neck node metastasis. The results showed that there was no significant difference in RRFS and OS between the two groups, which suggested that it was safe and feasible to omit irradiation to lateral neck regions for patients with no lateral neck node metastasis. Of course, there may be a certain deviation due to the small sample size and few end-point events. Prospective studies are warranted to further verify our conclusion.\nTwo patients had lymph node recurrence, which occurred in the contralateral supraclavicular fossa. Both of the patients underwent R0 resection at first treatment. One of the patients (Pt.1) had positive lymph nodes in level VI and the other patient (Pt.2) had negative lymph node. Pt.1 received adjuvant RT to the tumor bed and neck levels III, IV, and VI. Pt.2 received RT for tumor bed and neck level IV. Recurrences occurred 3 and 5 years respectively after surgery. It’s worth noting that both of the patients suffered synchronous or metachronous lung and mediastinal lymph node metastasis. It seems that the recurrence of the supraclavicular lymph node may be caused by lung and mediastinal lymph node metastasis.\nDistant metastasis is the major failure pattern for CASTLE after surgery and IMRT. However, the role of chemotherapy remains unclear because of the rarity of the disease. Different regimens have been explored, including cisplatin, epirubicin, docetaxel, irinotecan, vincristine, and cyclophosphamide. But the results seem to be heterogeneous. Hanamura et al. [17] reported a good response to platinum-based chemotherapy of a patient with lung metastasis from CASTLE. The author suggested that CASTLE is a chemosensitive tumor, and chemotherapy should be recommended for patients with advanced or metastatic disease. However, in contrast to Hanamura’s study, Roka et al. [18] found no response to three different regimens of a patient with liver metastasis. In the current study, one patient with gross residual disease after surgery received concurrent chemotherapy (docetaxel and cisplatin). However, the chemotherapy was stopped because of the toxicities after one cycle. CR was achieved for the positive lymph node and PR for the primary tumor after IMRT. Unfortunately, the patient suffered lung metastasis 18 months after IMRT. Systematic therapy combined with IMRT may be an effective treatment option for patients with locally advanced diseases. However, the optimal regimen and combination mode needs to be further investigated.\nImmunotherapy is a revolutionary breakthrough in the treatment of cancer. The effectiveness and safeness have been proved in different tumors in the past few years. As to CASTLE, Lorenz et al. [19] reported the first case in 2019. The patient suffered multiple metastatic diseases (lung, mediastinal, hilar, and upper mesenteric lymph nodes and pleura) 10 years after treatment of CASTLE in the parotid gland. The tumor showed a high expression level of PD-L1 (60%), and the PD-L1 inhibitor (pembrolizumab 200 mg every three weeks) was given to the patient. Partial remission was achieved four months after the start of immunotherapy, and treatment-related toxicities were mild and tolerable. The result was exciting. Further research on immunotherapy with or without chemotherapy or radiotherapy for patients with advanced or metastatic CASTLE is warranted.", "Our long-term results showed that surgery combined with IMRT is an effective treatment for patients with CASTLE. Distant metastasis is the major failure pattern. For patients with no lateral neck node metastasis, the omission of irradiation for lateral neck nodal regions seems to be safe and feasible. Further prospective research is warranted.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, null, null, null, null, null, null, "results", null, null, null, null, null, null, "discussion", "conclusion", "supplementary-material", null ]
[ "Intensity-modulated radiotherapy", "IMRT", "Radiotherapy", "Carcinoma showing thymus-like differentiation", "CASTLE" ]
Sulbactam combined with tigecycline improves outcomes in patients with severe multidrug-resistant Acinetobacter baumannii pneumonia.
36271362
The purpose of this study was to review the treatment plan of patients with multidrug-resistant Acinetobacter baumannii (MDR-AB) pneumonia and analyze the factors associated with patient deaths and the medication regimen.
BACKGROUND
We collected 1,823 qualified respiratory specimens that were culture-positive for MDR-AB. 166 patients confirmed to have hospital-acquired MDR-AB pneumonia were selected as the research subjects. The differing clinical characteristics and treatment interventions between the surviving group and death group within 28 days were analyzed.
METHODS
The mortality rate was high for those aged > 75 years (p = 0.001). Patients who underwent invasive catheter placement (p < 0.001) and mechanical ventilation (p = 0.046) had a higher mortality rate. Combination therapy with tigecycline can reduce the mortality rate (p < 0.001) of MDR-AB pneumonia in patients with carbapenem-resistant AB(CRAB). Combination therapy with sulbactam was shown to reduce the mortality rate (p < 0.001), and high-dose sulbactam (> 3 g/day) might be better than low-dose sulbactam (≤ 3 g/day).
RESULTS
Reducing the time of invasive catheter placement and mechanical ventilation in patients in the intensive care unit (ICU), antimicrobial treatment, combined with tigecycline and sulbactam, might help reduce the mortality rate in patients with severe MDR-AB hospital-acquired pneumonia.
CONCLUSION
[ "Humans", "Acinetobacter baumannii", "Sulbactam", "Tigecycline", "Acinetobacter Infections", "Drug Resistance, Multiple, Bacterial", "Carbapenems", "Anti-Bacterial Agents", "Anti-Infective Agents", "Pneumonia", "Retrospective Studies", "Microbial Sensitivity Tests" ]
9585752
null
null
null
null
Results
[SUBTITLE] General clinical conditions [SUBSECTION] Of the 166 patients diagnosed with severe hospital-acquired MDR-AB pneumonia, 111 were males (Table 1). The median age was 69 years (19–94 years). We divided the patients with severe MDR-AB pneumonia into two groups based on the clinical outcome during 28 days of treatment: (1) death group (n = 45) and (2) survival group (n = 112). The death group was mainly concentrated in the age group over 75 years old (73.3%), which was consistent with our analysis. That patients older than 75 years had an increased risk of death (OR 0.921, p = 0.001). The median APACHE II score of the study population was 20. (The APACHE II score was used to assess the condition and prognosis of ICU patients. A score of > 15 was classified as more critically ill. The higher the score, the higher the risk of death during hospitalization.) Common underlying diseases were neurological diseases, heart diseases, and chronic lung diseases. Mechanical ventilation (OR 4.525) and invasive catheterization (OR 48.526) significantly increased the risk of death in severe MDR-AB patients (p < 0.05). Table 1Clinical data of patients with MDR-AB pneumoniaDeath group(n = 45)Survival group(n = 121)P valueDataAge (Y)81 (72.5–85.5)64 (50–75.5)< 0.001Gender29 (64.4%)82 (67.8%)0.686APACHEII Scores20 (20, 22.5)20 (16, 33)0.094Underlying diseasesLiver diseases7 (15.6%)12 (9.9%)0.31Neurovascular disease18 (40.0%)53 (43.8%)0.66Kidney diseases16 (35.6%)14 (11.6%)< 0.001Diabetes11 (24.4%)16 (13.2%)0.082Pulmonary diseases23 (51.1%)37 (30.6%)0.014Heart disease27 (60.0%)41 (33.9%)0.002Immune abnormalities4 (8.9%)14 (11.6%)0.621TreatmentTreatment delay7 (15.6%)19 (15.7%)0.982Mechanical ventilation38 (84.4%)65 (53.7%)< 0.001Invasive catheterization44 (95.6%)77 (63.6%)< 0.001Carbapenems43 (95.6%)92 (76.0%)0.004Tigecycline20 (44.4%)98 (81.0%)< 0.001Sulbactam20 (44.4%)80 (66.1%)0.011APACHEII, Acute Physiology and Chronic Health Evaluation II; MDR-AB, Multi-drug resistant Acinetobacter baumannii Clinical data of patients with MDR-AB pneumonia APACHEII, Acute Physiology and Chronic Health Evaluation II; MDR-AB, Multi-drug resistant Acinetobacter baumannii Of the 166 patients diagnosed with severe hospital-acquired MDR-AB pneumonia, 111 were males (Table 1). The median age was 69 years (19–94 years). We divided the patients with severe MDR-AB pneumonia into two groups based on the clinical outcome during 28 days of treatment: (1) death group (n = 45) and (2) survival group (n = 112). The death group was mainly concentrated in the age group over 75 years old (73.3%), which was consistent with our analysis. That patients older than 75 years had an increased risk of death (OR 0.921, p = 0.001). The median APACHE II score of the study population was 20. (The APACHE II score was used to assess the condition and prognosis of ICU patients. A score of > 15 was classified as more critically ill. The higher the score, the higher the risk of death during hospitalization.) Common underlying diseases were neurological diseases, heart diseases, and chronic lung diseases. Mechanical ventilation (OR 4.525) and invasive catheterization (OR 48.526) significantly increased the risk of death in severe MDR-AB patients (p < 0.05). Table 1Clinical data of patients with MDR-AB pneumoniaDeath group(n = 45)Survival group(n = 121)P valueDataAge (Y)81 (72.5–85.5)64 (50–75.5)< 0.001Gender29 (64.4%)82 (67.8%)0.686APACHEII Scores20 (20, 22.5)20 (16, 33)0.094Underlying diseasesLiver diseases7 (15.6%)12 (9.9%)0.31Neurovascular disease18 (40.0%)53 (43.8%)0.66Kidney diseases16 (35.6%)14 (11.6%)< 0.001Diabetes11 (24.4%)16 (13.2%)0.082Pulmonary diseases23 (51.1%)37 (30.6%)0.014Heart disease27 (60.0%)41 (33.9%)0.002Immune abnormalities4 (8.9%)14 (11.6%)0.621TreatmentTreatment delay7 (15.6%)19 (15.7%)0.982Mechanical ventilation38 (84.4%)65 (53.7%)< 0.001Invasive catheterization44 (95.6%)77 (63.6%)< 0.001Carbapenems43 (95.6%)92 (76.0%)0.004Tigecycline20 (44.4%)98 (81.0%)< 0.001Sulbactam20 (44.4%)80 (66.1%)0.011APACHEII, Acute Physiology and Chronic Health Evaluation II; MDR-AB, Multi-drug resistant Acinetobacter baumannii Clinical data of patients with MDR-AB pneumonia APACHEII, Acute Physiology and Chronic Health Evaluation II; MDR-AB, Multi-drug resistant Acinetobacter baumannii [SUBTITLE] Characteristics of Acinetobacter baumannii strains(abs) [SUBSECTION] Of the 166 patients diagnosed with severe MDR-AB, 157 were confirmed to be carbapenemase-producing. OXA-23 accounted for 98.1% of the genotypes of these CRAB strains, and only 3 patients had NDM. The drug susceptibility results of all Abs in vitro were shown in Table 2. Table 2The susceptibility results of Abs in vitroTotal(n = 166)Death group(n = 45)Survival group(n = 121)SISISIAmpicillin/sulbactam3/1668/1661/450/452/1218/121Piperacillin0/1660/1660/450/450/1210/121Carbapenems*9/1660/1664/450/455/1210/121Piperacillin/Tazobactam1/1663/1660/450/451/1213/121Ceftriaxone0/1664/1660/451/450/1213/121Cefotaxime0/1664/1660/451/450/1213/121Cefepime2/1663/1661/451/451/1212/121Cefoxitin0/1660/1660/450/450/1210/121Gentamicin7/1664/1662/451/455/1213/121Tobramycin15/1662/1663/451/4512/1211/121Amikacin12/1661/1663/450/459/1211/121Ciprofloxacin4/1660/1661/450/453/1210/121Levofloxacin4/1667/1661/450/453/1217/121Moxifloxacin4/1660/1661/450/453/1210/121Tetracycline3/1666/1661/450/452/1216/121Tigecycline161/1665/16644/451/45117/1214/121Trimethoprim/Sulfamethoxazole17/1660/1665/450/4512/1210/121Cefoperazone/Sulbactam3/1667/1661/452/452/1215/121Minocyclline25/16613/1664/452/4521/12111/121Ampicillin1/1660/1660/450/451/1210/121Amoxicillin/Clavulanic6/1660/1662/450/454/1210/121Aztreonam7/1660/1663/450/454/1210/121Cefotetan1/1660/1661/450/450/1210/121Cefazolin0/1660/1660/450/450/1210/121*Carbapenems including Imipenem/Meropenem/Doripenem The susceptibility results of Abs in vitro *Carbapenems including Imipenem/Meropenem/Doripenem Of the 166 patients diagnosed with severe MDR-AB, 157 were confirmed to be carbapenemase-producing. OXA-23 accounted for 98.1% of the genotypes of these CRAB strains, and only 3 patients had NDM. The drug susceptibility results of all Abs in vitro were shown in Table 2. Table 2The susceptibility results of Abs in vitroTotal(n = 166)Death group(n = 45)Survival group(n = 121)SISISIAmpicillin/sulbactam3/1668/1661/450/452/1218/121Piperacillin0/1660/1660/450/450/1210/121Carbapenems*9/1660/1664/450/455/1210/121Piperacillin/Tazobactam1/1663/1660/450/451/1213/121Ceftriaxone0/1664/1660/451/450/1213/121Cefotaxime0/1664/1660/451/450/1213/121Cefepime2/1663/1661/451/451/1212/121Cefoxitin0/1660/1660/450/450/1210/121Gentamicin7/1664/1662/451/455/1213/121Tobramycin15/1662/1663/451/4512/1211/121Amikacin12/1661/1663/450/459/1211/121Ciprofloxacin4/1660/1661/450/453/1210/121Levofloxacin4/1667/1661/450/453/1217/121Moxifloxacin4/1660/1661/450/453/1210/121Tetracycline3/1666/1661/450/452/1216/121Tigecycline161/1665/16644/451/45117/1214/121Trimethoprim/Sulfamethoxazole17/1660/1665/450/4512/1210/121Cefoperazone/Sulbactam3/1667/1661/452/452/1215/121Minocyclline25/16613/1664/452/4521/12111/121Ampicillin1/1660/1660/450/451/1210/121Amoxicillin/Clavulanic6/1660/1662/450/454/1210/121Aztreonam7/1660/1663/450/454/1210/121Cefotetan1/1660/1661/450/450/1210/121Cefazolin0/1660/1660/450/450/1210/121*Carbapenems including Imipenem/Meropenem/Doripenem The susceptibility results of Abs in vitro *Carbapenems including Imipenem/Meropenem/Doripenem [SUBTITLE] Antibiotic medication [SUBSECTION] All patients were prescribed 2 or more antibiotics for CRAB pneumonia. According to the recommendations of the MDR-AB diagnosis and treatment guidelines, most of the patients in the study used a combination treatment plan, and only a few were treated with one drug. Some single-agent treatments were explained in detail below (details were shown in Table 3). Table 3Antibiotic usage and clinical outcomes in patients with CRAB pneumonia. Clinical outcomes of different antibiotic regimens in CRAB pneumoniaAntibioticsNumber of casesNumber of deaths(Deaths/Total*100%)Sulbactam + Carbapenems + Tigecycline5912(20.34%)Sulbactam + Carbapenems243(1.25%)Sulbactam + Tigecycline302(6%)Carbapenems + Tigecycline233(13.04%)Sulbactam00(-)Carbapenems*2121(100%)Colistin21(50%)Not using any of the above three drugs00(-)* These patients were treated with other types of antibiotics, including 12 cases with quinolones, 7 cases with other β-lactams, and 2 cases with tetracyclines Antibiotic usage and clinical outcomes in patients with CRAB pneumonia. Clinical outcomes of different antibiotic regimens in CRAB pneumonia * These patients were treated with other types of antibiotics, including 12 cases with quinolones, 7 cases with other β-lactams, and 2 cases with tetracyclines All patients were prescribed 2 or more antibiotics for CRAB pneumonia. According to the recommendations of the MDR-AB diagnosis and treatment guidelines, most of the patients in the study used a combination treatment plan, and only a few were treated with one drug. Some single-agent treatments were explained in detail below (details were shown in Table 3). Table 3Antibiotic usage and clinical outcomes in patients with CRAB pneumonia. Clinical outcomes of different antibiotic regimens in CRAB pneumoniaAntibioticsNumber of casesNumber of deaths(Deaths/Total*100%)Sulbactam + Carbapenems + Tigecycline5912(20.34%)Sulbactam + Carbapenems243(1.25%)Sulbactam + Tigecycline302(6%)Carbapenems + Tigecycline233(13.04%)Sulbactam00(-)Carbapenems*2121(100%)Colistin21(50%)Not using any of the above three drugs00(-)* These patients were treated with other types of antibiotics, including 12 cases with quinolones, 7 cases with other β-lactams, and 2 cases with tetracyclines Antibiotic usage and clinical outcomes in patients with CRAB pneumonia. Clinical outcomes of different antibiotic regimens in CRAB pneumonia * These patients were treated with other types of antibiotics, including 12 cases with quinolones, 7 cases with other β-lactams, and 2 cases with tetracyclines [SUBTITLE] Tigecycline [SUBSECTION] Among the 166 patients diagnosed with sever MDR-AB pneumonia, the resistance rate to tigecycline was 3% (5/166) in vitro ( Sensitivity to tigecycline was defined as MIC < 2 mg/L). There were 113 cases were prescribed tigecycline, and the 28-day mortality rate was 16.7%. The remaining 48 patients who did not use tigecycline exhibited a higher mortality rate of 53.2% (p < 0.001). There were another five patients were not prescribed tigecycline because of the MIC values of tigecycline higher than 2. Among the 166 patients diagnosed with sever MDR-AB pneumonia, the resistance rate to tigecycline was 3% (5/166) in vitro ( Sensitivity to tigecycline was defined as MIC < 2 mg/L). There were 113 cases were prescribed tigecycline, and the 28-day mortality rate was 16.7%. The remaining 48 patients who did not use tigecycline exhibited a higher mortality rate of 53.2% (p < 0.001). There were another five patients were not prescribed tigecycline because of the MIC values of tigecycline higher than 2. [SUBTITLE] Carbapenem [SUBSECTION] There were 157 cases resistant to carbapenem, of which 127 cases produced carbapenemase. These patients were diagnosed with CRAB pneumonia. The resistance rate to carbapenem was 94.6% (157/166). The enzyme-producing genotypes of these CRABs have been described previously. Except for 3 strains producing NDM, the rest were all OXA-23 type. Although the carbapenem resistance rate was high, there were still 104 cases with carbapenem included in the combination treatment regimen. Based on whether tigecycline (T) or sulbactam (S) was used, they were divided into TS group and TS unused group. As a result, the mortality rate in TS group was 6.7%; however, the mortality rate in TS unused group was 30.7% (p = 0.007). The main combined antibiotics in the TS unused group included quinolones (12 cases), other β-lactam antibiotics (9 cases), and tetracyclines (2 cases). There were 157 cases resistant to carbapenem, of which 127 cases produced carbapenemase. These patients were diagnosed with CRAB pneumonia. The resistance rate to carbapenem was 94.6% (157/166). The enzyme-producing genotypes of these CRABs have been described previously. Except for 3 strains producing NDM, the rest were all OXA-23 type. Although the carbapenem resistance rate was high, there were still 104 cases with carbapenem included in the combination treatment regimen. Based on whether tigecycline (T) or sulbactam (S) was used, they were divided into TS group and TS unused group. As a result, the mortality rate in TS group was 6.7%; however, the mortality rate in TS unused group was 30.7% (p = 0.007). The main combined antibiotics in the TS unused group included quinolones (12 cases), other β-lactam antibiotics (9 cases), and tetracyclines (2 cases). [SUBTITLE] Sulbactam [SUBSECTION] From the above results, sulbactam was effective in the treatment of CRAB in addition to tigecycline. The 28-day mortality of patients with CRAB pneumonia was completely different among the different combination regimens, as shown in Fig. 2. The mortality rate of the regimens that included tigecycline, carbapenem, and sulbactam was much lower than that of tigecycline plus carbapenem alone. The effect of the dose of sulbactam on the efficacy of CRAB should be further clarified. The mortality rate in CRAB pneumonia patients treated with carbapenem combined with sulbactam was 18.1%, whereas the mortality rate was 54.5% (p < 0.001) in those patients who were treated with the combination of carbapenem and other antimicrobial drugs (including aminoglycosides, fluoroquinolones, and tetracyclines). For CRAB pneumonia, the addition of sulbactam may reduce the MIC value of carbapenem through a synergistic effect. These patients who were given carbapenem and sulbactam in combination could be divided into a high-dose group and a low-dose group according to the sulbactam dosage. The high-dose group (30 cases) had a mortality rate of 16.7%, and the low-dose group (53 cases) had a mortality rate of 18.9% (p = 0.802) (Fig. 3). Fig. 2The 28-day survival rate in CRAB patients treated with Carbapenem + Tigecycline or Carbapenem + Tigecycline and Sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii The 28-day survival rate in CRAB patients treated with Carbapenem + Tigecycline or Carbapenem + Tigecycline and Sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii Fig. 3Mortality rate in patients with CRAB pneumonia treated with Carbapenem + Tigecycline alone or Carbapenem + Tigecycline and sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii Mortality rate in patients with CRAB pneumonia treated with Carbapenem + Tigecycline alone or Carbapenem + Tigecycline and sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii From the above results, sulbactam was effective in the treatment of CRAB in addition to tigecycline. The 28-day mortality of patients with CRAB pneumonia was completely different among the different combination regimens, as shown in Fig. 2. The mortality rate of the regimens that included tigecycline, carbapenem, and sulbactam was much lower than that of tigecycline plus carbapenem alone. The effect of the dose of sulbactam on the efficacy of CRAB should be further clarified. The mortality rate in CRAB pneumonia patients treated with carbapenem combined with sulbactam was 18.1%, whereas the mortality rate was 54.5% (p < 0.001) in those patients who were treated with the combination of carbapenem and other antimicrobial drugs (including aminoglycosides, fluoroquinolones, and tetracyclines). For CRAB pneumonia, the addition of sulbactam may reduce the MIC value of carbapenem through a synergistic effect. These patients who were given carbapenem and sulbactam in combination could be divided into a high-dose group and a low-dose group according to the sulbactam dosage. The high-dose group (30 cases) had a mortality rate of 16.7%, and the low-dose group (53 cases) had a mortality rate of 18.9% (p = 0.802) (Fig. 3). Fig. 2The 28-day survival rate in CRAB patients treated with Carbapenem + Tigecycline or Carbapenem + Tigecycline and Sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii The 28-day survival rate in CRAB patients treated with Carbapenem + Tigecycline or Carbapenem + Tigecycline and Sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii Fig. 3Mortality rate in patients with CRAB pneumonia treated with Carbapenem + Tigecycline alone or Carbapenem + Tigecycline and sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii Mortality rate in patients with CRAB pneumonia treated with Carbapenem + Tigecycline alone or Carbapenem + Tigecycline and sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii [SUBTITLE] Colistin [SUBSECTION] There were only 2 patients received colistin for the treatment of CRAB. Patient 1 was automatically discharged from the hospital with colistin for less than 3 days; The second patient had delayed treatment and was diagnosed with invasive Aspergillus pulmonary pneumonia at the same time. Therefore, there were no sufficient data to be included in the evaluation of efficacy regarding colistin or combination regimens of colistin. There were only 2 patients received colistin for the treatment of CRAB. Patient 1 was automatically discharged from the hospital with colistin for less than 3 days; The second patient had delayed treatment and was diagnosed with invasive Aspergillus pulmonary pneumonia at the same time. Therefore, there were no sufficient data to be included in the evaluation of efficacy regarding colistin or combination regimens of colistin.
null
null
[ "Background", "Methods", "Subjects", "Method for detecting carbapenemase-producing genotype", "Clinical information", "Treatment", "Statistical methods", "General clinical conditions", "Characteristics of Acinetobacter baumannii strains(abs)", "Antibiotic medication", "Tigecycline", "Carbapenem", "Sulbactam", "Colistin" ]
[ "Acinetobacter baumannii (AB) is a common pathogen that causes opportunistic infections in hospitals. According to the China Antimicrobial Surveillance Network (CHINET) continuous drug resistance monitoring program, AB is the second most common gram-negative bacterium isolated from respiratory specimens and accounted for about 17.07% of infections in 2020. Important risk factors for AB infection include admission to the ICU, impaired immune function, receiving broad-spectrum antibacterial drugs, and mechanical ventilation [1,2]. Since AB has a high rate of resistance to antimicrobial drugs, the control of multi-drug resistant AB (MDR-AB) is currently one of the main goals of nosocomial infection prevention and control.\nIn Asia, the sensitivity of AB to carbapenems were less than 27% [3]. As such, a number of guidelines recommend the combination of polymyxin, tigecycline, carbapenems, sulbactam, or other antibacterial drugs for the treatment of MDR-AB pneumonia in China or abroad [4, 5]. However, the sensitivity of AB to certain drugs differs in different regions. Thus, it is necessary to design an appropriate combination drug regimen according to the local drug resistance situation and common guidelines and recommendations.\nIt is a challenge for clinicians to choose antibacterial drugs when trying to control severe MDR-AB infection, especially carbapenemase-resistant Acinetobacter baumannii (CRAB). According to an in vitro drug sensitivity test, the resistance rate of AB to carbapenems ranged from 62–100% [3], and the resistance rate to cefoperazone/sulbactam was 46.3% [6]. Antibiotics containing sulbactam were the main drugs for the treatment of Acinetobacter infection in China, partly because of the economic burden. The conventional dose is 4 g/d for MDR-AB, but it can be increased to 6.0 g/d or even 8.0 g/d according to expert consensus on the diagnosis, treatment, prevention, and control of AB infection in China [4]. In addition to sulbactam, tigecycline is the most used for the control of hospital-acquired MDR-AB, but its clinical efficacy is controversial [7]. Studies have shown that even if AB was sensitive to tigecycline in vitro, it could not improve the prognosis of patients with MDR-AB. The main objective of this study was to investigate feasible and effective treatment options for patients with severe MDR-AB pneumonia, from a clinical perspective. Here, we retrospectively identified factors influencing mortality in patients with severe MDR-AB pneumonia and appropriate antibiotic regimens, especially the combined effect of sulbactam and tigecycline.", "[SUBTITLE] Subjects [SUBSECTION] We enrolled 1,823 respiratory tract specimens cultured as MDR-AB from 2016 to 2020 in this study. Out of the 1,823 cases, 166 patients were diagnosed with MDR-AB pneumonia. The screening process was shown in Fig. 1. This retrospective study was approved by the ethics committee of Sichuan Provincial People’s Hospital (Affiliated Hospital of the University of Electronic Science and Technology of China). Approval number: 2019059B.\n\nFig. 1MDR-AB inclusion and exclusion flow chart. *Severe immunodeficiency patients: long-term use of immunosuppressants, solid organ transplant patients, recent use of high-dose methylprednisolone drugs (more than 280 mg/w), hematological malignancies, and patients with severe agranulocytosis. MDR-AB, Multi-drug resistant Acinetobacter baumannii\n\nMDR-AB inclusion and exclusion flow chart. *Severe immunodeficiency patients: long-term use of immunosuppressants, solid organ transplant patients, recent use of high-dose methylprednisolone drugs (more than 280 mg/w), hematological malignancies, and patients with severe agranulocytosis. MDR-AB, Multi-drug resistant Acinetobacter baumannii\nWe enrolled 1,823 respiratory tract specimens cultured as MDR-AB from 2016 to 2020 in this study. Out of the 1,823 cases, 166 patients were diagnosed with MDR-AB pneumonia. The screening process was shown in Fig. 1. This retrospective study was approved by the ethics committee of Sichuan Provincial People’s Hospital (Affiliated Hospital of the University of Electronic Science and Technology of China). Approval number: 2019059B.\n\nFig. 1MDR-AB inclusion and exclusion flow chart. *Severe immunodeficiency patients: long-term use of immunosuppressants, solid organ transplant patients, recent use of high-dose methylprednisolone drugs (more than 280 mg/w), hematological malignancies, and patients with severe agranulocytosis. MDR-AB, Multi-drug resistant Acinetobacter baumannii\n\nMDR-AB inclusion and exclusion flow chart. *Severe immunodeficiency patients: long-term use of immunosuppressants, solid organ transplant patients, recent use of high-dose methylprednisolone drugs (more than 280 mg/w), hematological malignancies, and patients with severe agranulocytosis. MDR-AB, Multi-drug resistant Acinetobacter baumannii\n[SUBTITLE] Method for detecting carbapenemase-producing genotype [SUBSECTION] The carbapenemase detection kits (LFA) was used to detect the 157 CRAB strains, which provided by Beijing Gold Mountainriver Tech Development Co., Ltd. Take the strains out of the − 80 °C freezer 24 h in advance and wait at room temperature to thaw. Dip the mixed strain with an inoculating loop and inoculate it on a blood plate or a MacConkey plate in a four-district line. Place these plates in a constant temperature incubator at 35 °C for 18-24 h. Then, fresh strains of AB grown uniformly over 3 zones on the plates were used for the assays.\nThe carbapenemase detection kits (LFA) was used to detect the 157 CRAB strains, which provided by Beijing Gold Mountainriver Tech Development Co., Ltd. Take the strains out of the − 80 °C freezer 24 h in advance and wait at room temperature to thaw. Dip the mixed strain with an inoculating loop and inoculate it on a blood plate or a MacConkey plate in a four-district line. Place these plates in a constant temperature incubator at 35 °C for 18-24 h. Then, fresh strains of AB grown uniformly over 3 zones on the plates were used for the assays.\n[SUBTITLE] Clinical information [SUBSECTION] For each patient, the following information was collected: age, sex, underlying diseases, initial diagnosis at admission, clinical features, laboratory findings, other microorganism expect AB, radiological findings, clinical course until diagnosis, antibiotic and invasive treatment, Acute Physiology and Chronic Health Evaluation II (APACHE II) score at admission, and in-hospital outcome.\nFor each patient, the following information was collected: age, sex, underlying diseases, initial diagnosis at admission, clinical features, laboratory findings, other microorganism expect AB, radiological findings, clinical course until diagnosis, antibiotic and invasive treatment, Acute Physiology and Chronic Health Evaluation II (APACHE II) score at admission, and in-hospital outcome.\n[SUBTITLE] Treatment [SUBSECTION] Each treatment plan used to treat patients with MDR-AB was collected. Treatment plans included invasive catheterization (tracheal intubation, central venous catheterization, and urinary tract catheterization) and antibiotic use (polymyxin, tigecycline, carbapenem, sulbactam, aminoglycosides, quinolones, and tetracyclines). Preparations containing single sulbactam were divided into a high-dose group (> 3 g/day) and a low-dose group (≤ 3 g/day). Cases with delayed treatment were excluded. Delayed treatment meant that the clinician obtained the MDR-AB culture results from the laboratory but did not give effective antimicrobial treatment within 3 days.\n\nDose and duration of tigecycline: the first dose of tigecycline was 100 mg (2 bottles), and then 50 mg (1 bottle) every 12 h. The intravenous infusion time of tigecycline should be once every 12 h, about 30 ~ 60 min each time. The course of treatment in this study was 5–14 days.Carbapenem uses meropenem, dose and duration: it was prepared with sterile water for injection, containing 250 mg meropenem per 5ml, the concentration was about every 50 mg/ml, and it was administered once every 8 h, 500 mg each time, intravenous drip.Sulbactam preparation used single sulbactam to participate in the combined treatment, dose and duration: 2 ~ 4 g per day. The same amount was given intravenously concomitant infusion with cefoperazone once every 12 h or 8 h for 30 ~ 60 min.\n\nDose and duration of tigecycline: the first dose of tigecycline was 100 mg (2 bottles), and then 50 mg (1 bottle) every 12 h. The intravenous infusion time of tigecycline should be once every 12 h, about 30 ~ 60 min each time. The course of treatment in this study was 5–14 days.\nCarbapenem uses meropenem, dose and duration: it was prepared with sterile water for injection, containing 250 mg meropenem per 5ml, the concentration was about every 50 mg/ml, and it was administered once every 8 h, 500 mg each time, intravenous drip.\nSulbactam preparation used single sulbactam to participate in the combined treatment, dose and duration: 2 ~ 4 g per day. The same amount was given intravenously concomitant infusion with cefoperazone once every 12 h or 8 h for 30 ~ 60 min.\nEach treatment plan used to treat patients with MDR-AB was collected. Treatment plans included invasive catheterization (tracheal intubation, central venous catheterization, and urinary tract catheterization) and antibiotic use (polymyxin, tigecycline, carbapenem, sulbactam, aminoglycosides, quinolones, and tetracyclines). Preparations containing single sulbactam were divided into a high-dose group (> 3 g/day) and a low-dose group (≤ 3 g/day). Cases with delayed treatment were excluded. Delayed treatment meant that the clinician obtained the MDR-AB culture results from the laboratory but did not give effective antimicrobial treatment within 3 days.\n\nDose and duration of tigecycline: the first dose of tigecycline was 100 mg (2 bottles), and then 50 mg (1 bottle) every 12 h. The intravenous infusion time of tigecycline should be once every 12 h, about 30 ~ 60 min each time. The course of treatment in this study was 5–14 days.Carbapenem uses meropenem, dose and duration: it was prepared with sterile water for injection, containing 250 mg meropenem per 5ml, the concentration was about every 50 mg/ml, and it was administered once every 8 h, 500 mg each time, intravenous drip.Sulbactam preparation used single sulbactam to participate in the combined treatment, dose and duration: 2 ~ 4 g per day. The same amount was given intravenously concomitant infusion with cefoperazone once every 12 h or 8 h for 30 ~ 60 min.\n\nDose and duration of tigecycline: the first dose of tigecycline was 100 mg (2 bottles), and then 50 mg (1 bottle) every 12 h. The intravenous infusion time of tigecycline should be once every 12 h, about 30 ~ 60 min each time. The course of treatment in this study was 5–14 days.\nCarbapenem uses meropenem, dose and duration: it was prepared with sterile water for injection, containing 250 mg meropenem per 5ml, the concentration was about every 50 mg/ml, and it was administered once every 8 h, 500 mg each time, intravenous drip.\nSulbactam preparation used single sulbactam to participate in the combined treatment, dose and duration: 2 ~ 4 g per day. The same amount was given intravenously concomitant infusion with cefoperazone once every 12 h or 8 h for 30 ~ 60 min.\n[SUBTITLE] Statistical methods [SUBSECTION] SPSS 23.0 software (SPSS Inc., Chicago, United States) was used to analyze the data. Measurement-type data were tested for normality and homogeneity of variance. The data that conformed to normal distribution and homogeneity of variance were expressed as the mean ± standard deviation (x ± s) and analyzed using a t-test. An adjusted t-test method was used for the data that conformed to normal distribution but did not conform to homogeneity of variance. The data that did not conform to normal distribution were expressed as the median [M (Ql, Qu)], and the rank sum test was used for analysis. A chi-square test was used for counting data. After statistical analysis, the independent variables with statistical significance were selected, and the selected variables were subjected to multi-independent logistic regression analysis. A p-value < 0.05 was regarded as statistically significant.\nSPSS 23.0 software (SPSS Inc., Chicago, United States) was used to analyze the data. Measurement-type data were tested for normality and homogeneity of variance. The data that conformed to normal distribution and homogeneity of variance were expressed as the mean ± standard deviation (x ± s) and analyzed using a t-test. An adjusted t-test method was used for the data that conformed to normal distribution but did not conform to homogeneity of variance. The data that did not conform to normal distribution were expressed as the median [M (Ql, Qu)], and the rank sum test was used for analysis. A chi-square test was used for counting data. After statistical analysis, the independent variables with statistical significance were selected, and the selected variables were subjected to multi-independent logistic regression analysis. A p-value < 0.05 was regarded as statistically significant.", "We enrolled 1,823 respiratory tract specimens cultured as MDR-AB from 2016 to 2020 in this study. Out of the 1,823 cases, 166 patients were diagnosed with MDR-AB pneumonia. The screening process was shown in Fig. 1. This retrospective study was approved by the ethics committee of Sichuan Provincial People’s Hospital (Affiliated Hospital of the University of Electronic Science and Technology of China). Approval number: 2019059B.\n\nFig. 1MDR-AB inclusion and exclusion flow chart. *Severe immunodeficiency patients: long-term use of immunosuppressants, solid organ transplant patients, recent use of high-dose methylprednisolone drugs (more than 280 mg/w), hematological malignancies, and patients with severe agranulocytosis. MDR-AB, Multi-drug resistant Acinetobacter baumannii\n\nMDR-AB inclusion and exclusion flow chart. *Severe immunodeficiency patients: long-term use of immunosuppressants, solid organ transplant patients, recent use of high-dose methylprednisolone drugs (more than 280 mg/w), hematological malignancies, and patients with severe agranulocytosis. MDR-AB, Multi-drug resistant Acinetobacter baumannii", "The carbapenemase detection kits (LFA) was used to detect the 157 CRAB strains, which provided by Beijing Gold Mountainriver Tech Development Co., Ltd. Take the strains out of the − 80 °C freezer 24 h in advance and wait at room temperature to thaw. Dip the mixed strain with an inoculating loop and inoculate it on a blood plate or a MacConkey plate in a four-district line. Place these plates in a constant temperature incubator at 35 °C for 18-24 h. Then, fresh strains of AB grown uniformly over 3 zones on the plates were used for the assays.", "For each patient, the following information was collected: age, sex, underlying diseases, initial diagnosis at admission, clinical features, laboratory findings, other microorganism expect AB, radiological findings, clinical course until diagnosis, antibiotic and invasive treatment, Acute Physiology and Chronic Health Evaluation II (APACHE II) score at admission, and in-hospital outcome.", "Each treatment plan used to treat patients with MDR-AB was collected. Treatment plans included invasive catheterization (tracheal intubation, central venous catheterization, and urinary tract catheterization) and antibiotic use (polymyxin, tigecycline, carbapenem, sulbactam, aminoglycosides, quinolones, and tetracyclines). Preparations containing single sulbactam were divided into a high-dose group (> 3 g/day) and a low-dose group (≤ 3 g/day). Cases with delayed treatment were excluded. Delayed treatment meant that the clinician obtained the MDR-AB culture results from the laboratory but did not give effective antimicrobial treatment within 3 days.\n\nDose and duration of tigecycline: the first dose of tigecycline was 100 mg (2 bottles), and then 50 mg (1 bottle) every 12 h. The intravenous infusion time of tigecycline should be once every 12 h, about 30 ~ 60 min each time. The course of treatment in this study was 5–14 days.Carbapenem uses meropenem, dose and duration: it was prepared with sterile water for injection, containing 250 mg meropenem per 5ml, the concentration was about every 50 mg/ml, and it was administered once every 8 h, 500 mg each time, intravenous drip.Sulbactam preparation used single sulbactam to participate in the combined treatment, dose and duration: 2 ~ 4 g per day. The same amount was given intravenously concomitant infusion with cefoperazone once every 12 h or 8 h for 30 ~ 60 min.\n\nDose and duration of tigecycline: the first dose of tigecycline was 100 mg (2 bottles), and then 50 mg (1 bottle) every 12 h. The intravenous infusion time of tigecycline should be once every 12 h, about 30 ~ 60 min each time. The course of treatment in this study was 5–14 days.\nCarbapenem uses meropenem, dose and duration: it was prepared with sterile water for injection, containing 250 mg meropenem per 5ml, the concentration was about every 50 mg/ml, and it was administered once every 8 h, 500 mg each time, intravenous drip.\nSulbactam preparation used single sulbactam to participate in the combined treatment, dose and duration: 2 ~ 4 g per day. The same amount was given intravenously concomitant infusion with cefoperazone once every 12 h or 8 h for 30 ~ 60 min.", "SPSS 23.0 software (SPSS Inc., Chicago, United States) was used to analyze the data. Measurement-type data were tested for normality and homogeneity of variance. The data that conformed to normal distribution and homogeneity of variance were expressed as the mean ± standard deviation (x ± s) and analyzed using a t-test. An adjusted t-test method was used for the data that conformed to normal distribution but did not conform to homogeneity of variance. The data that did not conform to normal distribution were expressed as the median [M (Ql, Qu)], and the rank sum test was used for analysis. A chi-square test was used for counting data. After statistical analysis, the independent variables with statistical significance were selected, and the selected variables were subjected to multi-independent logistic regression analysis. A p-value < 0.05 was regarded as statistically significant.", "Of the 166 patients diagnosed with severe hospital-acquired MDR-AB pneumonia, 111 were males (Table 1). The median age was 69 years (19–94 years). We divided the patients with severe MDR-AB pneumonia into two groups based on the clinical outcome during 28 days of treatment: (1) death group (n = 45) and (2) survival group (n = 112). The death group was mainly concentrated in the age group over 75 years old (73.3%), which was consistent with our analysis. That patients older than 75 years had an increased risk of death (OR 0.921, p = 0.001). The median APACHE II score of the study population was 20. (The APACHE II score was used to assess the condition and prognosis of ICU patients. A score of > 15 was classified as more critically ill. The higher the score, the higher the risk of death during hospitalization.) Common underlying diseases were neurological diseases, heart diseases, and chronic lung diseases. Mechanical ventilation (OR 4.525) and invasive catheterization (OR 48.526) significantly increased the risk of death in severe MDR-AB patients (p < 0.05).\n\nTable 1Clinical data of patients with MDR-AB pneumoniaDeath group(n = 45)Survival group(n = 121)P valueDataAge (Y)81 (72.5–85.5)64 (50–75.5)< 0.001Gender29 (64.4%)82 (67.8%)0.686APACHEII Scores20 (20, 22.5)20 (16, 33)0.094Underlying diseasesLiver diseases7 (15.6%)12 (9.9%)0.31Neurovascular disease18 (40.0%)53 (43.8%)0.66Kidney diseases16 (35.6%)14 (11.6%)< 0.001Diabetes11 (24.4%)16 (13.2%)0.082Pulmonary diseases23 (51.1%)37 (30.6%)0.014Heart disease27 (60.0%)41 (33.9%)0.002Immune abnormalities4 (8.9%)14 (11.6%)0.621TreatmentTreatment delay7 (15.6%)19 (15.7%)0.982Mechanical ventilation38 (84.4%)65 (53.7%)< 0.001Invasive catheterization44 (95.6%)77 (63.6%)< 0.001Carbapenems43 (95.6%)92 (76.0%)0.004Tigecycline20 (44.4%)98 (81.0%)< 0.001Sulbactam20 (44.4%)80 (66.1%)0.011APACHEII, Acute Physiology and Chronic Health Evaluation II; MDR-AB, Multi-drug resistant Acinetobacter baumannii\n\nClinical data of patients with MDR-AB pneumonia\nAPACHEII, Acute Physiology and Chronic Health Evaluation II; MDR-AB, Multi-drug resistant Acinetobacter baumannii", "Of the 166 patients diagnosed with severe MDR-AB, 157 were confirmed to be carbapenemase-producing. OXA-23 accounted for 98.1% of the genotypes of these CRAB strains, and only 3 patients had NDM. The drug susceptibility results of all Abs in vitro were shown in Table 2.\n\nTable 2The susceptibility results of Abs in vitroTotal(n = 166)Death group(n = 45)Survival group(n = 121)SISISIAmpicillin/sulbactam3/1668/1661/450/452/1218/121Piperacillin0/1660/1660/450/450/1210/121Carbapenems*9/1660/1664/450/455/1210/121Piperacillin/Tazobactam1/1663/1660/450/451/1213/121Ceftriaxone0/1664/1660/451/450/1213/121Cefotaxime0/1664/1660/451/450/1213/121Cefepime2/1663/1661/451/451/1212/121Cefoxitin0/1660/1660/450/450/1210/121Gentamicin7/1664/1662/451/455/1213/121Tobramycin15/1662/1663/451/4512/1211/121Amikacin12/1661/1663/450/459/1211/121Ciprofloxacin4/1660/1661/450/453/1210/121Levofloxacin4/1667/1661/450/453/1217/121Moxifloxacin4/1660/1661/450/453/1210/121Tetracycline3/1666/1661/450/452/1216/121Tigecycline161/1665/16644/451/45117/1214/121Trimethoprim/Sulfamethoxazole17/1660/1665/450/4512/1210/121Cefoperazone/Sulbactam3/1667/1661/452/452/1215/121Minocyclline25/16613/1664/452/4521/12111/121Ampicillin1/1660/1660/450/451/1210/121Amoxicillin/Clavulanic6/1660/1662/450/454/1210/121Aztreonam7/1660/1663/450/454/1210/121Cefotetan1/1660/1661/450/450/1210/121Cefazolin0/1660/1660/450/450/1210/121*Carbapenems including Imipenem/Meropenem/Doripenem\n\nThe susceptibility results of Abs in vitro\n*Carbapenems including Imipenem/Meropenem/Doripenem", "All patients were prescribed 2 or more antibiotics for CRAB pneumonia. According to the recommendations of the MDR-AB diagnosis and treatment guidelines, most of the patients in the study used a combination treatment plan, and only a few were treated with one drug. Some single-agent treatments were explained in detail below (details were shown in Table 3).\n\nTable 3Antibiotic usage and clinical outcomes in patients with CRAB pneumonia. Clinical outcomes of different antibiotic regimens in CRAB pneumoniaAntibioticsNumber of casesNumber of deaths(Deaths/Total*100%)Sulbactam + Carbapenems + Tigecycline5912(20.34%)Sulbactam + Carbapenems243(1.25%)Sulbactam + Tigecycline302(6%)Carbapenems + Tigecycline233(13.04%)Sulbactam00(-)Carbapenems*2121(100%)Colistin21(50%)Not using any of the above three drugs00(-)* These patients were treated with other types of antibiotics, including 12 cases with quinolones, 7 cases with other β-lactams, and 2 cases with tetracyclines\n\nAntibiotic usage and clinical outcomes in patients with CRAB pneumonia. Clinical outcomes of different antibiotic regimens in CRAB pneumonia\n* These patients were treated with other types of antibiotics, including 12 cases with quinolones, 7 cases with other β-lactams, and 2 cases with tetracyclines", "Among the 166 patients diagnosed with sever MDR-AB pneumonia, the resistance rate to tigecycline was 3% (5/166) in vitro ( Sensitivity to tigecycline was defined as MIC < 2 mg/L). There were 113 cases were prescribed tigecycline, and the 28-day mortality rate was 16.7%. The remaining 48 patients who did not use tigecycline exhibited a higher mortality rate of 53.2% (p < 0.001). There were another five patients were not prescribed tigecycline because of the MIC values of tigecycline higher than 2.", "There were 157 cases resistant to carbapenem, of which 127 cases produced carbapenemase. These patients were diagnosed with CRAB pneumonia. The resistance rate to carbapenem was 94.6% (157/166). The enzyme-producing genotypes of these CRABs have been described previously. Except for 3 strains producing NDM, the rest were all OXA-23 type. Although the carbapenem resistance rate was high, there were still 104 cases with carbapenem included in the combination treatment regimen. Based on whether tigecycline (T) or sulbactam (S) was used, they were divided into TS group and TS unused group. As a result, the mortality rate in TS group was 6.7%; however, the mortality rate in TS unused group was 30.7% (p = 0.007). The main combined antibiotics in the TS unused group included quinolones (12 cases), other β-lactam antibiotics (9 cases), and tetracyclines (2 cases).", "From the above results, sulbactam was effective in the treatment of CRAB in addition to tigecycline. The 28-day mortality of patients with CRAB pneumonia was completely different among the different combination regimens, as shown in Fig. 2. The mortality rate of the regimens that included tigecycline, carbapenem, and sulbactam was much lower than that of tigecycline plus carbapenem alone. The effect of the dose of sulbactam on the efficacy of CRAB should be further clarified. The mortality rate in CRAB pneumonia patients treated with carbapenem combined with sulbactam was 18.1%, whereas the mortality rate was 54.5% (p < 0.001) in those patients who were treated with the combination of carbapenem and other antimicrobial drugs (including aminoglycosides, fluoroquinolones, and tetracyclines). For CRAB pneumonia, the addition of sulbactam may reduce the MIC value of carbapenem through a synergistic effect. These patients who were given carbapenem and sulbactam in combination could be divided into a high-dose group and a low-dose group according to the sulbactam dosage. The high-dose group (30 cases) had a mortality rate of 16.7%, and the low-dose group (53 cases) had a mortality rate of 18.9% (p = 0.802) (Fig. 3).\n\nFig. 2The 28-day survival rate in CRAB patients treated with Carbapenem + Tigecycline or Carbapenem + Tigecycline and Sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\n\nThe 28-day survival rate in CRAB patients treated with Carbapenem + Tigecycline or Carbapenem + Tigecycline and Sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\n\nFig. 3Mortality rate in patients with CRAB pneumonia treated with Carbapenem + Tigecycline alone or Carbapenem + Tigecycline and sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\n\nMortality rate in patients with CRAB pneumonia treated with Carbapenem + Tigecycline alone or Carbapenem + Tigecycline and sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii", "There were only 2 patients received colistin for the treatment of CRAB. Patient 1 was automatically discharged from the hospital with colistin for less than 3 days; The second patient had delayed treatment and was diagnosed with invasive Aspergillus pulmonary pneumonia at the same time. Therefore, there were no sufficient data to be included in the evaluation of efficacy regarding colistin or combination regimens of colistin." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Subjects", "Method for detecting carbapenemase-producing genotype", "Clinical information", "Treatment", "Statistical methods", "Results", "General clinical conditions", "Characteristics of Acinetobacter baumannii strains(abs)", "Antibiotic medication", "Tigecycline", "Carbapenem", "Sulbactam", "Colistin", "Discussion" ]
[ "Acinetobacter baumannii (AB) is a common pathogen that causes opportunistic infections in hospitals. According to the China Antimicrobial Surveillance Network (CHINET) continuous drug resistance monitoring program, AB is the second most common gram-negative bacterium isolated from respiratory specimens and accounted for about 17.07% of infections in 2020. Important risk factors for AB infection include admission to the ICU, impaired immune function, receiving broad-spectrum antibacterial drugs, and mechanical ventilation [1,2]. Since AB has a high rate of resistance to antimicrobial drugs, the control of multi-drug resistant AB (MDR-AB) is currently one of the main goals of nosocomial infection prevention and control.\nIn Asia, the sensitivity of AB to carbapenems were less than 27% [3]. As such, a number of guidelines recommend the combination of polymyxin, tigecycline, carbapenems, sulbactam, or other antibacterial drugs for the treatment of MDR-AB pneumonia in China or abroad [4, 5]. However, the sensitivity of AB to certain drugs differs in different regions. Thus, it is necessary to design an appropriate combination drug regimen according to the local drug resistance situation and common guidelines and recommendations.\nIt is a challenge for clinicians to choose antibacterial drugs when trying to control severe MDR-AB infection, especially carbapenemase-resistant Acinetobacter baumannii (CRAB). According to an in vitro drug sensitivity test, the resistance rate of AB to carbapenems ranged from 62–100% [3], and the resistance rate to cefoperazone/sulbactam was 46.3% [6]. Antibiotics containing sulbactam were the main drugs for the treatment of Acinetobacter infection in China, partly because of the economic burden. The conventional dose is 4 g/d for MDR-AB, but it can be increased to 6.0 g/d or even 8.0 g/d according to expert consensus on the diagnosis, treatment, prevention, and control of AB infection in China [4]. In addition to sulbactam, tigecycline is the most used for the control of hospital-acquired MDR-AB, but its clinical efficacy is controversial [7]. Studies have shown that even if AB was sensitive to tigecycline in vitro, it could not improve the prognosis of patients with MDR-AB. The main objective of this study was to investigate feasible and effective treatment options for patients with severe MDR-AB pneumonia, from a clinical perspective. Here, we retrospectively identified factors influencing mortality in patients with severe MDR-AB pneumonia and appropriate antibiotic regimens, especially the combined effect of sulbactam and tigecycline.", "[SUBTITLE] Subjects [SUBSECTION] We enrolled 1,823 respiratory tract specimens cultured as MDR-AB from 2016 to 2020 in this study. Out of the 1,823 cases, 166 patients were diagnosed with MDR-AB pneumonia. The screening process was shown in Fig. 1. This retrospective study was approved by the ethics committee of Sichuan Provincial People’s Hospital (Affiliated Hospital of the University of Electronic Science and Technology of China). Approval number: 2019059B.\n\nFig. 1MDR-AB inclusion and exclusion flow chart. *Severe immunodeficiency patients: long-term use of immunosuppressants, solid organ transplant patients, recent use of high-dose methylprednisolone drugs (more than 280 mg/w), hematological malignancies, and patients with severe agranulocytosis. MDR-AB, Multi-drug resistant Acinetobacter baumannii\n\nMDR-AB inclusion and exclusion flow chart. *Severe immunodeficiency patients: long-term use of immunosuppressants, solid organ transplant patients, recent use of high-dose methylprednisolone drugs (more than 280 mg/w), hematological malignancies, and patients with severe agranulocytosis. MDR-AB, Multi-drug resistant Acinetobacter baumannii\nWe enrolled 1,823 respiratory tract specimens cultured as MDR-AB from 2016 to 2020 in this study. Out of the 1,823 cases, 166 patients were diagnosed with MDR-AB pneumonia. The screening process was shown in Fig. 1. This retrospective study was approved by the ethics committee of Sichuan Provincial People’s Hospital (Affiliated Hospital of the University of Electronic Science and Technology of China). Approval number: 2019059B.\n\nFig. 1MDR-AB inclusion and exclusion flow chart. *Severe immunodeficiency patients: long-term use of immunosuppressants, solid organ transplant patients, recent use of high-dose methylprednisolone drugs (more than 280 mg/w), hematological malignancies, and patients with severe agranulocytosis. MDR-AB, Multi-drug resistant Acinetobacter baumannii\n\nMDR-AB inclusion and exclusion flow chart. *Severe immunodeficiency patients: long-term use of immunosuppressants, solid organ transplant patients, recent use of high-dose methylprednisolone drugs (more than 280 mg/w), hematological malignancies, and patients with severe agranulocytosis. MDR-AB, Multi-drug resistant Acinetobacter baumannii\n[SUBTITLE] Method for detecting carbapenemase-producing genotype [SUBSECTION] The carbapenemase detection kits (LFA) was used to detect the 157 CRAB strains, which provided by Beijing Gold Mountainriver Tech Development Co., Ltd. Take the strains out of the − 80 °C freezer 24 h in advance and wait at room temperature to thaw. Dip the mixed strain with an inoculating loop and inoculate it on a blood plate or a MacConkey plate in a four-district line. Place these plates in a constant temperature incubator at 35 °C for 18-24 h. Then, fresh strains of AB grown uniformly over 3 zones on the plates were used for the assays.\nThe carbapenemase detection kits (LFA) was used to detect the 157 CRAB strains, which provided by Beijing Gold Mountainriver Tech Development Co., Ltd. Take the strains out of the − 80 °C freezer 24 h in advance and wait at room temperature to thaw. Dip the mixed strain with an inoculating loop and inoculate it on a blood plate or a MacConkey plate in a four-district line. Place these plates in a constant temperature incubator at 35 °C for 18-24 h. Then, fresh strains of AB grown uniformly over 3 zones on the plates were used for the assays.\n[SUBTITLE] Clinical information [SUBSECTION] For each patient, the following information was collected: age, sex, underlying diseases, initial diagnosis at admission, clinical features, laboratory findings, other microorganism expect AB, radiological findings, clinical course until diagnosis, antibiotic and invasive treatment, Acute Physiology and Chronic Health Evaluation II (APACHE II) score at admission, and in-hospital outcome.\nFor each patient, the following information was collected: age, sex, underlying diseases, initial diagnosis at admission, clinical features, laboratory findings, other microorganism expect AB, radiological findings, clinical course until diagnosis, antibiotic and invasive treatment, Acute Physiology and Chronic Health Evaluation II (APACHE II) score at admission, and in-hospital outcome.\n[SUBTITLE] Treatment [SUBSECTION] Each treatment plan used to treat patients with MDR-AB was collected. Treatment plans included invasive catheterization (tracheal intubation, central venous catheterization, and urinary tract catheterization) and antibiotic use (polymyxin, tigecycline, carbapenem, sulbactam, aminoglycosides, quinolones, and tetracyclines). Preparations containing single sulbactam were divided into a high-dose group (> 3 g/day) and a low-dose group (≤ 3 g/day). Cases with delayed treatment were excluded. Delayed treatment meant that the clinician obtained the MDR-AB culture results from the laboratory but did not give effective antimicrobial treatment within 3 days.\n\nDose and duration of tigecycline: the first dose of tigecycline was 100 mg (2 bottles), and then 50 mg (1 bottle) every 12 h. The intravenous infusion time of tigecycline should be once every 12 h, about 30 ~ 60 min each time. The course of treatment in this study was 5–14 days.Carbapenem uses meropenem, dose and duration: it was prepared with sterile water for injection, containing 250 mg meropenem per 5ml, the concentration was about every 50 mg/ml, and it was administered once every 8 h, 500 mg each time, intravenous drip.Sulbactam preparation used single sulbactam to participate in the combined treatment, dose and duration: 2 ~ 4 g per day. The same amount was given intravenously concomitant infusion with cefoperazone once every 12 h or 8 h for 30 ~ 60 min.\n\nDose and duration of tigecycline: the first dose of tigecycline was 100 mg (2 bottles), and then 50 mg (1 bottle) every 12 h. The intravenous infusion time of tigecycline should be once every 12 h, about 30 ~ 60 min each time. The course of treatment in this study was 5–14 days.\nCarbapenem uses meropenem, dose and duration: it was prepared with sterile water for injection, containing 250 mg meropenem per 5ml, the concentration was about every 50 mg/ml, and it was administered once every 8 h, 500 mg each time, intravenous drip.\nSulbactam preparation used single sulbactam to participate in the combined treatment, dose and duration: 2 ~ 4 g per day. The same amount was given intravenously concomitant infusion with cefoperazone once every 12 h or 8 h for 30 ~ 60 min.\nEach treatment plan used to treat patients with MDR-AB was collected. Treatment plans included invasive catheterization (tracheal intubation, central venous catheterization, and urinary tract catheterization) and antibiotic use (polymyxin, tigecycline, carbapenem, sulbactam, aminoglycosides, quinolones, and tetracyclines). Preparations containing single sulbactam were divided into a high-dose group (> 3 g/day) and a low-dose group (≤ 3 g/day). Cases with delayed treatment were excluded. Delayed treatment meant that the clinician obtained the MDR-AB culture results from the laboratory but did not give effective antimicrobial treatment within 3 days.\n\nDose and duration of tigecycline: the first dose of tigecycline was 100 mg (2 bottles), and then 50 mg (1 bottle) every 12 h. The intravenous infusion time of tigecycline should be once every 12 h, about 30 ~ 60 min each time. The course of treatment in this study was 5–14 days.Carbapenem uses meropenem, dose and duration: it was prepared with sterile water for injection, containing 250 mg meropenem per 5ml, the concentration was about every 50 mg/ml, and it was administered once every 8 h, 500 mg each time, intravenous drip.Sulbactam preparation used single sulbactam to participate in the combined treatment, dose and duration: 2 ~ 4 g per day. The same amount was given intravenously concomitant infusion with cefoperazone once every 12 h or 8 h for 30 ~ 60 min.\n\nDose and duration of tigecycline: the first dose of tigecycline was 100 mg (2 bottles), and then 50 mg (1 bottle) every 12 h. The intravenous infusion time of tigecycline should be once every 12 h, about 30 ~ 60 min each time. The course of treatment in this study was 5–14 days.\nCarbapenem uses meropenem, dose and duration: it was prepared with sterile water for injection, containing 250 mg meropenem per 5ml, the concentration was about every 50 mg/ml, and it was administered once every 8 h, 500 mg each time, intravenous drip.\nSulbactam preparation used single sulbactam to participate in the combined treatment, dose and duration: 2 ~ 4 g per day. The same amount was given intravenously concomitant infusion with cefoperazone once every 12 h or 8 h for 30 ~ 60 min.\n[SUBTITLE] Statistical methods [SUBSECTION] SPSS 23.0 software (SPSS Inc., Chicago, United States) was used to analyze the data. Measurement-type data were tested for normality and homogeneity of variance. The data that conformed to normal distribution and homogeneity of variance were expressed as the mean ± standard deviation (x ± s) and analyzed using a t-test. An adjusted t-test method was used for the data that conformed to normal distribution but did not conform to homogeneity of variance. The data that did not conform to normal distribution were expressed as the median [M (Ql, Qu)], and the rank sum test was used for analysis. A chi-square test was used for counting data. After statistical analysis, the independent variables with statistical significance were selected, and the selected variables were subjected to multi-independent logistic regression analysis. A p-value < 0.05 was regarded as statistically significant.\nSPSS 23.0 software (SPSS Inc., Chicago, United States) was used to analyze the data. Measurement-type data were tested for normality and homogeneity of variance. The data that conformed to normal distribution and homogeneity of variance were expressed as the mean ± standard deviation (x ± s) and analyzed using a t-test. An adjusted t-test method was used for the data that conformed to normal distribution but did not conform to homogeneity of variance. The data that did not conform to normal distribution were expressed as the median [M (Ql, Qu)], and the rank sum test was used for analysis. A chi-square test was used for counting data. After statistical analysis, the independent variables with statistical significance were selected, and the selected variables were subjected to multi-independent logistic regression analysis. A p-value < 0.05 was regarded as statistically significant.", "We enrolled 1,823 respiratory tract specimens cultured as MDR-AB from 2016 to 2020 in this study. Out of the 1,823 cases, 166 patients were diagnosed with MDR-AB pneumonia. The screening process was shown in Fig. 1. This retrospective study was approved by the ethics committee of Sichuan Provincial People’s Hospital (Affiliated Hospital of the University of Electronic Science and Technology of China). Approval number: 2019059B.\n\nFig. 1MDR-AB inclusion and exclusion flow chart. *Severe immunodeficiency patients: long-term use of immunosuppressants, solid organ transplant patients, recent use of high-dose methylprednisolone drugs (more than 280 mg/w), hematological malignancies, and patients with severe agranulocytosis. MDR-AB, Multi-drug resistant Acinetobacter baumannii\n\nMDR-AB inclusion and exclusion flow chart. *Severe immunodeficiency patients: long-term use of immunosuppressants, solid organ transplant patients, recent use of high-dose methylprednisolone drugs (more than 280 mg/w), hematological malignancies, and patients with severe agranulocytosis. MDR-AB, Multi-drug resistant Acinetobacter baumannii", "The carbapenemase detection kits (LFA) was used to detect the 157 CRAB strains, which provided by Beijing Gold Mountainriver Tech Development Co., Ltd. Take the strains out of the − 80 °C freezer 24 h in advance and wait at room temperature to thaw. Dip the mixed strain with an inoculating loop and inoculate it on a blood plate or a MacConkey plate in a four-district line. Place these plates in a constant temperature incubator at 35 °C for 18-24 h. Then, fresh strains of AB grown uniformly over 3 zones on the plates were used for the assays.", "For each patient, the following information was collected: age, sex, underlying diseases, initial diagnosis at admission, clinical features, laboratory findings, other microorganism expect AB, radiological findings, clinical course until diagnosis, antibiotic and invasive treatment, Acute Physiology and Chronic Health Evaluation II (APACHE II) score at admission, and in-hospital outcome.", "Each treatment plan used to treat patients with MDR-AB was collected. Treatment plans included invasive catheterization (tracheal intubation, central venous catheterization, and urinary tract catheterization) and antibiotic use (polymyxin, tigecycline, carbapenem, sulbactam, aminoglycosides, quinolones, and tetracyclines). Preparations containing single sulbactam were divided into a high-dose group (> 3 g/day) and a low-dose group (≤ 3 g/day). Cases with delayed treatment were excluded. Delayed treatment meant that the clinician obtained the MDR-AB culture results from the laboratory but did not give effective antimicrobial treatment within 3 days.\n\nDose and duration of tigecycline: the first dose of tigecycline was 100 mg (2 bottles), and then 50 mg (1 bottle) every 12 h. The intravenous infusion time of tigecycline should be once every 12 h, about 30 ~ 60 min each time. The course of treatment in this study was 5–14 days.Carbapenem uses meropenem, dose and duration: it was prepared with sterile water for injection, containing 250 mg meropenem per 5ml, the concentration was about every 50 mg/ml, and it was administered once every 8 h, 500 mg each time, intravenous drip.Sulbactam preparation used single sulbactam to participate in the combined treatment, dose and duration: 2 ~ 4 g per day. The same amount was given intravenously concomitant infusion with cefoperazone once every 12 h or 8 h for 30 ~ 60 min.\n\nDose and duration of tigecycline: the first dose of tigecycline was 100 mg (2 bottles), and then 50 mg (1 bottle) every 12 h. The intravenous infusion time of tigecycline should be once every 12 h, about 30 ~ 60 min each time. The course of treatment in this study was 5–14 days.\nCarbapenem uses meropenem, dose and duration: it was prepared with sterile water for injection, containing 250 mg meropenem per 5ml, the concentration was about every 50 mg/ml, and it was administered once every 8 h, 500 mg each time, intravenous drip.\nSulbactam preparation used single sulbactam to participate in the combined treatment, dose and duration: 2 ~ 4 g per day. The same amount was given intravenously concomitant infusion with cefoperazone once every 12 h or 8 h for 30 ~ 60 min.", "SPSS 23.0 software (SPSS Inc., Chicago, United States) was used to analyze the data. Measurement-type data were tested for normality and homogeneity of variance. The data that conformed to normal distribution and homogeneity of variance were expressed as the mean ± standard deviation (x ± s) and analyzed using a t-test. An adjusted t-test method was used for the data that conformed to normal distribution but did not conform to homogeneity of variance. The data that did not conform to normal distribution were expressed as the median [M (Ql, Qu)], and the rank sum test was used for analysis. A chi-square test was used for counting data. After statistical analysis, the independent variables with statistical significance were selected, and the selected variables were subjected to multi-independent logistic regression analysis. A p-value < 0.05 was regarded as statistically significant.", "[SUBTITLE] General clinical conditions [SUBSECTION] Of the 166 patients diagnosed with severe hospital-acquired MDR-AB pneumonia, 111 were males (Table 1). The median age was 69 years (19–94 years). We divided the patients with severe MDR-AB pneumonia into two groups based on the clinical outcome during 28 days of treatment: (1) death group (n = 45) and (2) survival group (n = 112). The death group was mainly concentrated in the age group over 75 years old (73.3%), which was consistent with our analysis. That patients older than 75 years had an increased risk of death (OR 0.921, p = 0.001). The median APACHE II score of the study population was 20. (The APACHE II score was used to assess the condition and prognosis of ICU patients. A score of > 15 was classified as more critically ill. The higher the score, the higher the risk of death during hospitalization.) Common underlying diseases were neurological diseases, heart diseases, and chronic lung diseases. Mechanical ventilation (OR 4.525) and invasive catheterization (OR 48.526) significantly increased the risk of death in severe MDR-AB patients (p < 0.05).\n\nTable 1Clinical data of patients with MDR-AB pneumoniaDeath group(n = 45)Survival group(n = 121)P valueDataAge (Y)81 (72.5–85.5)64 (50–75.5)< 0.001Gender29 (64.4%)82 (67.8%)0.686APACHEII Scores20 (20, 22.5)20 (16, 33)0.094Underlying diseasesLiver diseases7 (15.6%)12 (9.9%)0.31Neurovascular disease18 (40.0%)53 (43.8%)0.66Kidney diseases16 (35.6%)14 (11.6%)< 0.001Diabetes11 (24.4%)16 (13.2%)0.082Pulmonary diseases23 (51.1%)37 (30.6%)0.014Heart disease27 (60.0%)41 (33.9%)0.002Immune abnormalities4 (8.9%)14 (11.6%)0.621TreatmentTreatment delay7 (15.6%)19 (15.7%)0.982Mechanical ventilation38 (84.4%)65 (53.7%)< 0.001Invasive catheterization44 (95.6%)77 (63.6%)< 0.001Carbapenems43 (95.6%)92 (76.0%)0.004Tigecycline20 (44.4%)98 (81.0%)< 0.001Sulbactam20 (44.4%)80 (66.1%)0.011APACHEII, Acute Physiology and Chronic Health Evaluation II; MDR-AB, Multi-drug resistant Acinetobacter baumannii\n\nClinical data of patients with MDR-AB pneumonia\nAPACHEII, Acute Physiology and Chronic Health Evaluation II; MDR-AB, Multi-drug resistant Acinetobacter baumannii\nOf the 166 patients diagnosed with severe hospital-acquired MDR-AB pneumonia, 111 were males (Table 1). The median age was 69 years (19–94 years). We divided the patients with severe MDR-AB pneumonia into two groups based on the clinical outcome during 28 days of treatment: (1) death group (n = 45) and (2) survival group (n = 112). The death group was mainly concentrated in the age group over 75 years old (73.3%), which was consistent with our analysis. That patients older than 75 years had an increased risk of death (OR 0.921, p = 0.001). The median APACHE II score of the study population was 20. (The APACHE II score was used to assess the condition and prognosis of ICU patients. A score of > 15 was classified as more critically ill. The higher the score, the higher the risk of death during hospitalization.) Common underlying diseases were neurological diseases, heart diseases, and chronic lung diseases. Mechanical ventilation (OR 4.525) and invasive catheterization (OR 48.526) significantly increased the risk of death in severe MDR-AB patients (p < 0.05).\n\nTable 1Clinical data of patients with MDR-AB pneumoniaDeath group(n = 45)Survival group(n = 121)P valueDataAge (Y)81 (72.5–85.5)64 (50–75.5)< 0.001Gender29 (64.4%)82 (67.8%)0.686APACHEII Scores20 (20, 22.5)20 (16, 33)0.094Underlying diseasesLiver diseases7 (15.6%)12 (9.9%)0.31Neurovascular disease18 (40.0%)53 (43.8%)0.66Kidney diseases16 (35.6%)14 (11.6%)< 0.001Diabetes11 (24.4%)16 (13.2%)0.082Pulmonary diseases23 (51.1%)37 (30.6%)0.014Heart disease27 (60.0%)41 (33.9%)0.002Immune abnormalities4 (8.9%)14 (11.6%)0.621TreatmentTreatment delay7 (15.6%)19 (15.7%)0.982Mechanical ventilation38 (84.4%)65 (53.7%)< 0.001Invasive catheterization44 (95.6%)77 (63.6%)< 0.001Carbapenems43 (95.6%)92 (76.0%)0.004Tigecycline20 (44.4%)98 (81.0%)< 0.001Sulbactam20 (44.4%)80 (66.1%)0.011APACHEII, Acute Physiology and Chronic Health Evaluation II; MDR-AB, Multi-drug resistant Acinetobacter baumannii\n\nClinical data of patients with MDR-AB pneumonia\nAPACHEII, Acute Physiology and Chronic Health Evaluation II; MDR-AB, Multi-drug resistant Acinetobacter baumannii\n[SUBTITLE] Characteristics of Acinetobacter baumannii strains(abs) [SUBSECTION] Of the 166 patients diagnosed with severe MDR-AB, 157 were confirmed to be carbapenemase-producing. OXA-23 accounted for 98.1% of the genotypes of these CRAB strains, and only 3 patients had NDM. The drug susceptibility results of all Abs in vitro were shown in Table 2.\n\nTable 2The susceptibility results of Abs in vitroTotal(n = 166)Death group(n = 45)Survival group(n = 121)SISISIAmpicillin/sulbactam3/1668/1661/450/452/1218/121Piperacillin0/1660/1660/450/450/1210/121Carbapenems*9/1660/1664/450/455/1210/121Piperacillin/Tazobactam1/1663/1660/450/451/1213/121Ceftriaxone0/1664/1660/451/450/1213/121Cefotaxime0/1664/1660/451/450/1213/121Cefepime2/1663/1661/451/451/1212/121Cefoxitin0/1660/1660/450/450/1210/121Gentamicin7/1664/1662/451/455/1213/121Tobramycin15/1662/1663/451/4512/1211/121Amikacin12/1661/1663/450/459/1211/121Ciprofloxacin4/1660/1661/450/453/1210/121Levofloxacin4/1667/1661/450/453/1217/121Moxifloxacin4/1660/1661/450/453/1210/121Tetracycline3/1666/1661/450/452/1216/121Tigecycline161/1665/16644/451/45117/1214/121Trimethoprim/Sulfamethoxazole17/1660/1665/450/4512/1210/121Cefoperazone/Sulbactam3/1667/1661/452/452/1215/121Minocyclline25/16613/1664/452/4521/12111/121Ampicillin1/1660/1660/450/451/1210/121Amoxicillin/Clavulanic6/1660/1662/450/454/1210/121Aztreonam7/1660/1663/450/454/1210/121Cefotetan1/1660/1661/450/450/1210/121Cefazolin0/1660/1660/450/450/1210/121*Carbapenems including Imipenem/Meropenem/Doripenem\n\nThe susceptibility results of Abs in vitro\n*Carbapenems including Imipenem/Meropenem/Doripenem\nOf the 166 patients diagnosed with severe MDR-AB, 157 were confirmed to be carbapenemase-producing. OXA-23 accounted for 98.1% of the genotypes of these CRAB strains, and only 3 patients had NDM. The drug susceptibility results of all Abs in vitro were shown in Table 2.\n\nTable 2The susceptibility results of Abs in vitroTotal(n = 166)Death group(n = 45)Survival group(n = 121)SISISIAmpicillin/sulbactam3/1668/1661/450/452/1218/121Piperacillin0/1660/1660/450/450/1210/121Carbapenems*9/1660/1664/450/455/1210/121Piperacillin/Tazobactam1/1663/1660/450/451/1213/121Ceftriaxone0/1664/1660/451/450/1213/121Cefotaxime0/1664/1660/451/450/1213/121Cefepime2/1663/1661/451/451/1212/121Cefoxitin0/1660/1660/450/450/1210/121Gentamicin7/1664/1662/451/455/1213/121Tobramycin15/1662/1663/451/4512/1211/121Amikacin12/1661/1663/450/459/1211/121Ciprofloxacin4/1660/1661/450/453/1210/121Levofloxacin4/1667/1661/450/453/1217/121Moxifloxacin4/1660/1661/450/453/1210/121Tetracycline3/1666/1661/450/452/1216/121Tigecycline161/1665/16644/451/45117/1214/121Trimethoprim/Sulfamethoxazole17/1660/1665/450/4512/1210/121Cefoperazone/Sulbactam3/1667/1661/452/452/1215/121Minocyclline25/16613/1664/452/4521/12111/121Ampicillin1/1660/1660/450/451/1210/121Amoxicillin/Clavulanic6/1660/1662/450/454/1210/121Aztreonam7/1660/1663/450/454/1210/121Cefotetan1/1660/1661/450/450/1210/121Cefazolin0/1660/1660/450/450/1210/121*Carbapenems including Imipenem/Meropenem/Doripenem\n\nThe susceptibility results of Abs in vitro\n*Carbapenems including Imipenem/Meropenem/Doripenem\n[SUBTITLE] Antibiotic medication [SUBSECTION] All patients were prescribed 2 or more antibiotics for CRAB pneumonia. According to the recommendations of the MDR-AB diagnosis and treatment guidelines, most of the patients in the study used a combination treatment plan, and only a few were treated with one drug. Some single-agent treatments were explained in detail below (details were shown in Table 3).\n\nTable 3Antibiotic usage and clinical outcomes in patients with CRAB pneumonia. Clinical outcomes of different antibiotic regimens in CRAB pneumoniaAntibioticsNumber of casesNumber of deaths(Deaths/Total*100%)Sulbactam + Carbapenems + Tigecycline5912(20.34%)Sulbactam + Carbapenems243(1.25%)Sulbactam + Tigecycline302(6%)Carbapenems + Tigecycline233(13.04%)Sulbactam00(-)Carbapenems*2121(100%)Colistin21(50%)Not using any of the above three drugs00(-)* These patients were treated with other types of antibiotics, including 12 cases with quinolones, 7 cases with other β-lactams, and 2 cases with tetracyclines\n\nAntibiotic usage and clinical outcomes in patients with CRAB pneumonia. Clinical outcomes of different antibiotic regimens in CRAB pneumonia\n* These patients were treated with other types of antibiotics, including 12 cases with quinolones, 7 cases with other β-lactams, and 2 cases with tetracyclines\nAll patients were prescribed 2 or more antibiotics for CRAB pneumonia. According to the recommendations of the MDR-AB diagnosis and treatment guidelines, most of the patients in the study used a combination treatment plan, and only a few were treated with one drug. Some single-agent treatments were explained in detail below (details were shown in Table 3).\n\nTable 3Antibiotic usage and clinical outcomes in patients with CRAB pneumonia. Clinical outcomes of different antibiotic regimens in CRAB pneumoniaAntibioticsNumber of casesNumber of deaths(Deaths/Total*100%)Sulbactam + Carbapenems + Tigecycline5912(20.34%)Sulbactam + Carbapenems243(1.25%)Sulbactam + Tigecycline302(6%)Carbapenems + Tigecycline233(13.04%)Sulbactam00(-)Carbapenems*2121(100%)Colistin21(50%)Not using any of the above three drugs00(-)* These patients were treated with other types of antibiotics, including 12 cases with quinolones, 7 cases with other β-lactams, and 2 cases with tetracyclines\n\nAntibiotic usage and clinical outcomes in patients with CRAB pneumonia. Clinical outcomes of different antibiotic regimens in CRAB pneumonia\n* These patients were treated with other types of antibiotics, including 12 cases with quinolones, 7 cases with other β-lactams, and 2 cases with tetracyclines\n[SUBTITLE] Tigecycline [SUBSECTION] Among the 166 patients diagnosed with sever MDR-AB pneumonia, the resistance rate to tigecycline was 3% (5/166) in vitro ( Sensitivity to tigecycline was defined as MIC < 2 mg/L). There were 113 cases were prescribed tigecycline, and the 28-day mortality rate was 16.7%. The remaining 48 patients who did not use tigecycline exhibited a higher mortality rate of 53.2% (p < 0.001). There were another five patients were not prescribed tigecycline because of the MIC values of tigecycline higher than 2.\nAmong the 166 patients diagnosed with sever MDR-AB pneumonia, the resistance rate to tigecycline was 3% (5/166) in vitro ( Sensitivity to tigecycline was defined as MIC < 2 mg/L). There were 113 cases were prescribed tigecycline, and the 28-day mortality rate was 16.7%. The remaining 48 patients who did not use tigecycline exhibited a higher mortality rate of 53.2% (p < 0.001). There were another five patients were not prescribed tigecycline because of the MIC values of tigecycline higher than 2.\n[SUBTITLE] Carbapenem [SUBSECTION] There were 157 cases resistant to carbapenem, of which 127 cases produced carbapenemase. These patients were diagnosed with CRAB pneumonia. The resistance rate to carbapenem was 94.6% (157/166). The enzyme-producing genotypes of these CRABs have been described previously. Except for 3 strains producing NDM, the rest were all OXA-23 type. Although the carbapenem resistance rate was high, there were still 104 cases with carbapenem included in the combination treatment regimen. Based on whether tigecycline (T) or sulbactam (S) was used, they were divided into TS group and TS unused group. As a result, the mortality rate in TS group was 6.7%; however, the mortality rate in TS unused group was 30.7% (p = 0.007). The main combined antibiotics in the TS unused group included quinolones (12 cases), other β-lactam antibiotics (9 cases), and tetracyclines (2 cases).\nThere were 157 cases resistant to carbapenem, of which 127 cases produced carbapenemase. These patients were diagnosed with CRAB pneumonia. The resistance rate to carbapenem was 94.6% (157/166). The enzyme-producing genotypes of these CRABs have been described previously. Except for 3 strains producing NDM, the rest were all OXA-23 type. Although the carbapenem resistance rate was high, there were still 104 cases with carbapenem included in the combination treatment regimen. Based on whether tigecycline (T) or sulbactam (S) was used, they were divided into TS group and TS unused group. As a result, the mortality rate in TS group was 6.7%; however, the mortality rate in TS unused group was 30.7% (p = 0.007). The main combined antibiotics in the TS unused group included quinolones (12 cases), other β-lactam antibiotics (9 cases), and tetracyclines (2 cases).\n[SUBTITLE] Sulbactam [SUBSECTION] From the above results, sulbactam was effective in the treatment of CRAB in addition to tigecycline. The 28-day mortality of patients with CRAB pneumonia was completely different among the different combination regimens, as shown in Fig. 2. The mortality rate of the regimens that included tigecycline, carbapenem, and sulbactam was much lower than that of tigecycline plus carbapenem alone. The effect of the dose of sulbactam on the efficacy of CRAB should be further clarified. The mortality rate in CRAB pneumonia patients treated with carbapenem combined with sulbactam was 18.1%, whereas the mortality rate was 54.5% (p < 0.001) in those patients who were treated with the combination of carbapenem and other antimicrobial drugs (including aminoglycosides, fluoroquinolones, and tetracyclines). For CRAB pneumonia, the addition of sulbactam may reduce the MIC value of carbapenem through a synergistic effect. These patients who were given carbapenem and sulbactam in combination could be divided into a high-dose group and a low-dose group according to the sulbactam dosage. The high-dose group (30 cases) had a mortality rate of 16.7%, and the low-dose group (53 cases) had a mortality rate of 18.9% (p = 0.802) (Fig. 3).\n\nFig. 2The 28-day survival rate in CRAB patients treated with Carbapenem + Tigecycline or Carbapenem + Tigecycline and Sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\n\nThe 28-day survival rate in CRAB patients treated with Carbapenem + Tigecycline or Carbapenem + Tigecycline and Sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\n\nFig. 3Mortality rate in patients with CRAB pneumonia treated with Carbapenem + Tigecycline alone or Carbapenem + Tigecycline and sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\n\nMortality rate in patients with CRAB pneumonia treated with Carbapenem + Tigecycline alone or Carbapenem + Tigecycline and sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\nFrom the above results, sulbactam was effective in the treatment of CRAB in addition to tigecycline. The 28-day mortality of patients with CRAB pneumonia was completely different among the different combination regimens, as shown in Fig. 2. The mortality rate of the regimens that included tigecycline, carbapenem, and sulbactam was much lower than that of tigecycline plus carbapenem alone. The effect of the dose of sulbactam on the efficacy of CRAB should be further clarified. The mortality rate in CRAB pneumonia patients treated with carbapenem combined with sulbactam was 18.1%, whereas the mortality rate was 54.5% (p < 0.001) in those patients who were treated with the combination of carbapenem and other antimicrobial drugs (including aminoglycosides, fluoroquinolones, and tetracyclines). For CRAB pneumonia, the addition of sulbactam may reduce the MIC value of carbapenem through a synergistic effect. These patients who were given carbapenem and sulbactam in combination could be divided into a high-dose group and a low-dose group according to the sulbactam dosage. The high-dose group (30 cases) had a mortality rate of 16.7%, and the low-dose group (53 cases) had a mortality rate of 18.9% (p = 0.802) (Fig. 3).\n\nFig. 2The 28-day survival rate in CRAB patients treated with Carbapenem + Tigecycline or Carbapenem + Tigecycline and Sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\n\nThe 28-day survival rate in CRAB patients treated with Carbapenem + Tigecycline or Carbapenem + Tigecycline and Sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\n\nFig. 3Mortality rate in patients with CRAB pneumonia treated with Carbapenem + Tigecycline alone or Carbapenem + Tigecycline and sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\n\nMortality rate in patients with CRAB pneumonia treated with Carbapenem + Tigecycline alone or Carbapenem + Tigecycline and sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\n[SUBTITLE] Colistin [SUBSECTION] There were only 2 patients received colistin for the treatment of CRAB. Patient 1 was automatically discharged from the hospital with colistin for less than 3 days; The second patient had delayed treatment and was diagnosed with invasive Aspergillus pulmonary pneumonia at the same time. Therefore, there were no sufficient data to be included in the evaluation of efficacy regarding colistin or combination regimens of colistin.\nThere were only 2 patients received colistin for the treatment of CRAB. Patient 1 was automatically discharged from the hospital with colistin for less than 3 days; The second patient had delayed treatment and was diagnosed with invasive Aspergillus pulmonary pneumonia at the same time. Therefore, there were no sufficient data to be included in the evaluation of efficacy regarding colistin or combination regimens of colistin.", "Of the 166 patients diagnosed with severe hospital-acquired MDR-AB pneumonia, 111 were males (Table 1). The median age was 69 years (19–94 years). We divided the patients with severe MDR-AB pneumonia into two groups based on the clinical outcome during 28 days of treatment: (1) death group (n = 45) and (2) survival group (n = 112). The death group was mainly concentrated in the age group over 75 years old (73.3%), which was consistent with our analysis. That patients older than 75 years had an increased risk of death (OR 0.921, p = 0.001). The median APACHE II score of the study population was 20. (The APACHE II score was used to assess the condition and prognosis of ICU patients. A score of > 15 was classified as more critically ill. The higher the score, the higher the risk of death during hospitalization.) Common underlying diseases were neurological diseases, heart diseases, and chronic lung diseases. Mechanical ventilation (OR 4.525) and invasive catheterization (OR 48.526) significantly increased the risk of death in severe MDR-AB patients (p < 0.05).\n\nTable 1Clinical data of patients with MDR-AB pneumoniaDeath group(n = 45)Survival group(n = 121)P valueDataAge (Y)81 (72.5–85.5)64 (50–75.5)< 0.001Gender29 (64.4%)82 (67.8%)0.686APACHEII Scores20 (20, 22.5)20 (16, 33)0.094Underlying diseasesLiver diseases7 (15.6%)12 (9.9%)0.31Neurovascular disease18 (40.0%)53 (43.8%)0.66Kidney diseases16 (35.6%)14 (11.6%)< 0.001Diabetes11 (24.4%)16 (13.2%)0.082Pulmonary diseases23 (51.1%)37 (30.6%)0.014Heart disease27 (60.0%)41 (33.9%)0.002Immune abnormalities4 (8.9%)14 (11.6%)0.621TreatmentTreatment delay7 (15.6%)19 (15.7%)0.982Mechanical ventilation38 (84.4%)65 (53.7%)< 0.001Invasive catheterization44 (95.6%)77 (63.6%)< 0.001Carbapenems43 (95.6%)92 (76.0%)0.004Tigecycline20 (44.4%)98 (81.0%)< 0.001Sulbactam20 (44.4%)80 (66.1%)0.011APACHEII, Acute Physiology and Chronic Health Evaluation II; MDR-AB, Multi-drug resistant Acinetobacter baumannii\n\nClinical data of patients with MDR-AB pneumonia\nAPACHEII, Acute Physiology and Chronic Health Evaluation II; MDR-AB, Multi-drug resistant Acinetobacter baumannii", "Of the 166 patients diagnosed with severe MDR-AB, 157 were confirmed to be carbapenemase-producing. OXA-23 accounted for 98.1% of the genotypes of these CRAB strains, and only 3 patients had NDM. The drug susceptibility results of all Abs in vitro were shown in Table 2.\n\nTable 2The susceptibility results of Abs in vitroTotal(n = 166)Death group(n = 45)Survival group(n = 121)SISISIAmpicillin/sulbactam3/1668/1661/450/452/1218/121Piperacillin0/1660/1660/450/450/1210/121Carbapenems*9/1660/1664/450/455/1210/121Piperacillin/Tazobactam1/1663/1660/450/451/1213/121Ceftriaxone0/1664/1660/451/450/1213/121Cefotaxime0/1664/1660/451/450/1213/121Cefepime2/1663/1661/451/451/1212/121Cefoxitin0/1660/1660/450/450/1210/121Gentamicin7/1664/1662/451/455/1213/121Tobramycin15/1662/1663/451/4512/1211/121Amikacin12/1661/1663/450/459/1211/121Ciprofloxacin4/1660/1661/450/453/1210/121Levofloxacin4/1667/1661/450/453/1217/121Moxifloxacin4/1660/1661/450/453/1210/121Tetracycline3/1666/1661/450/452/1216/121Tigecycline161/1665/16644/451/45117/1214/121Trimethoprim/Sulfamethoxazole17/1660/1665/450/4512/1210/121Cefoperazone/Sulbactam3/1667/1661/452/452/1215/121Minocyclline25/16613/1664/452/4521/12111/121Ampicillin1/1660/1660/450/451/1210/121Amoxicillin/Clavulanic6/1660/1662/450/454/1210/121Aztreonam7/1660/1663/450/454/1210/121Cefotetan1/1660/1661/450/450/1210/121Cefazolin0/1660/1660/450/450/1210/121*Carbapenems including Imipenem/Meropenem/Doripenem\n\nThe susceptibility results of Abs in vitro\n*Carbapenems including Imipenem/Meropenem/Doripenem", "All patients were prescribed 2 or more antibiotics for CRAB pneumonia. According to the recommendations of the MDR-AB diagnosis and treatment guidelines, most of the patients in the study used a combination treatment plan, and only a few were treated with one drug. Some single-agent treatments were explained in detail below (details were shown in Table 3).\n\nTable 3Antibiotic usage and clinical outcomes in patients with CRAB pneumonia. Clinical outcomes of different antibiotic regimens in CRAB pneumoniaAntibioticsNumber of casesNumber of deaths(Deaths/Total*100%)Sulbactam + Carbapenems + Tigecycline5912(20.34%)Sulbactam + Carbapenems243(1.25%)Sulbactam + Tigecycline302(6%)Carbapenems + Tigecycline233(13.04%)Sulbactam00(-)Carbapenems*2121(100%)Colistin21(50%)Not using any of the above three drugs00(-)* These patients were treated with other types of antibiotics, including 12 cases with quinolones, 7 cases with other β-lactams, and 2 cases with tetracyclines\n\nAntibiotic usage and clinical outcomes in patients with CRAB pneumonia. Clinical outcomes of different antibiotic regimens in CRAB pneumonia\n* These patients were treated with other types of antibiotics, including 12 cases with quinolones, 7 cases with other β-lactams, and 2 cases with tetracyclines", "Among the 166 patients diagnosed with sever MDR-AB pneumonia, the resistance rate to tigecycline was 3% (5/166) in vitro ( Sensitivity to tigecycline was defined as MIC < 2 mg/L). There were 113 cases were prescribed tigecycline, and the 28-day mortality rate was 16.7%. The remaining 48 patients who did not use tigecycline exhibited a higher mortality rate of 53.2% (p < 0.001). There were another five patients were not prescribed tigecycline because of the MIC values of tigecycline higher than 2.", "There were 157 cases resistant to carbapenem, of which 127 cases produced carbapenemase. These patients were diagnosed with CRAB pneumonia. The resistance rate to carbapenem was 94.6% (157/166). The enzyme-producing genotypes of these CRABs have been described previously. Except for 3 strains producing NDM, the rest were all OXA-23 type. Although the carbapenem resistance rate was high, there were still 104 cases with carbapenem included in the combination treatment regimen. Based on whether tigecycline (T) or sulbactam (S) was used, they were divided into TS group and TS unused group. As a result, the mortality rate in TS group was 6.7%; however, the mortality rate in TS unused group was 30.7% (p = 0.007). The main combined antibiotics in the TS unused group included quinolones (12 cases), other β-lactam antibiotics (9 cases), and tetracyclines (2 cases).", "From the above results, sulbactam was effective in the treatment of CRAB in addition to tigecycline. The 28-day mortality of patients with CRAB pneumonia was completely different among the different combination regimens, as shown in Fig. 2. The mortality rate of the regimens that included tigecycline, carbapenem, and sulbactam was much lower than that of tigecycline plus carbapenem alone. The effect of the dose of sulbactam on the efficacy of CRAB should be further clarified. The mortality rate in CRAB pneumonia patients treated with carbapenem combined with sulbactam was 18.1%, whereas the mortality rate was 54.5% (p < 0.001) in those patients who were treated with the combination of carbapenem and other antimicrobial drugs (including aminoglycosides, fluoroquinolones, and tetracyclines). For CRAB pneumonia, the addition of sulbactam may reduce the MIC value of carbapenem through a synergistic effect. These patients who were given carbapenem and sulbactam in combination could be divided into a high-dose group and a low-dose group according to the sulbactam dosage. The high-dose group (30 cases) had a mortality rate of 16.7%, and the low-dose group (53 cases) had a mortality rate of 18.9% (p = 0.802) (Fig. 3).\n\nFig. 2The 28-day survival rate in CRAB patients treated with Carbapenem + Tigecycline or Carbapenem + Tigecycline and Sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\n\nThe 28-day survival rate in CRAB patients treated with Carbapenem + Tigecycline or Carbapenem + Tigecycline and Sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\n\nFig. 3Mortality rate in patients with CRAB pneumonia treated with Carbapenem + Tigecycline alone or Carbapenem + Tigecycline and sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii\n\nMortality rate in patients with CRAB pneumonia treated with Carbapenem + Tigecycline alone or Carbapenem + Tigecycline and sulbactam. CRAB, carbapenem-resistant Acinetobacter baumannii", "There were only 2 patients received colistin for the treatment of CRAB. Patient 1 was automatically discharged from the hospital with colistin for less than 3 days; The second patient had delayed treatment and was diagnosed with invasive Aspergillus pulmonary pneumonia at the same time. Therefore, there were no sufficient data to be included in the evaluation of efficacy regarding colistin or combination regimens of colistin.", "MDR-AB increases the mortality rate of nosocomial pneumonia. Our study found that admission to the intensive care unit, age > 75 years, invasive catheterization, and mechanical ventilation increased the mortality of patients with severe MDR-AB pneumonia, which was consistent with the results of other studies [9]. Early removal of invasive catheters and reducing the time on mechanical ventilation might reduce the mortality of critically ill patients diagnosed with severe MDR-AB pneumonia. Table 1 shows that there were age differences in clinical outcomes in the study population, but no differences by gender. This may be related to factors such as more complications, more co-morbidities, difficulty in weaning from ventilator, and longer ICU stay in older adults with severe pneumonia.\nIn our study, the main drugs that affect the mortality of patients with severe MDR-AB pneumonia or CRAB pneumonia included tigecycline, carbapenem, and sulbactam. Although Abs had good sensitivity to colistin in vitro, the MIC of colistin was less than 1 in China. But colistin was not widely used in the treatment of CRAB because of its high cost and not being covered by general health insurance in our country. There were only 2 patients were treated with colistin for CRAB, and they could not be included in the study due to uncertainty of clinical data and co-infection of fungal.\nAlthough tigecycline was recommended as a first-line drug for CRAB pneumonia, there were different opinions on whether its use was beneficial to reduce mortality. The resistance rate of tigecycline in vitro was low in China (about 2.9%) [6], which ranges from 0.2 to 74.2% in other countries and regions [3]. The concentration of tigecycline was higher in tissue, and it has suitable antibacterial activity and was safe for use against local tissue infections. Therefore, it has been theorized that tigecycline was an effective antibiotic for the treatment of CRAB and can reduce mortality [10. A previous meta-analysis showed that patients treated with tigecycline for CRAB infection had a higher mortality rate, longer hospital stay, and lower microbial clearance rate as compared with other drugs [11]. Another prospective study demonstrated that tigecycline increases the mortality rate in patients with CRAB infection when the tigecycline MIC > 2 mg/L [12]. The results of our study showed that tigecycline could significantly reduce the mortality rate of hospital-acquired pneumonia when CRAB was the main pathogen and treatment was not delayed. Because of Abs isolated in this study was sensitive to tigecycline (Mean MIC = 1.5 mg/L) in vitro, and the therapeutic effect of tigecycline might be related to drug sensitivity. Even if these patients had higher APACHE II scores, some of them were prescribed tigecycline alone, there were still good clinical outcomes, if tigecycline was sensitive in vitro.\nMost MDR-AB produce carbapenemases in previous study [13]. We also found that the resistance rate of carbapenem reached 94.6%. Continued use of carbapenem did not reduce the mortality of patients with CRAB pneumonia. It was not recommended to use only carbapenem for theses patients, but it might be effective as one of the combined treatment options, especially the MIC of carbapenem was less than 8. It was worth noting that, the combination of other drugs may reduce the carbapenem MIC value. The most common combination was sulbactam and or tigecycline in clinical practice. Carbapenem and sulbactam had been shown to have a synergistic effect and could reduce mortality [14]. Sulbactam, a β-lactamase inhibitor, has a suitable clinical therapeutic effect, and most of Abs has a low rate of resistance to sulbactam. Sulbactam enhanced the antibacterial effect of carbapenem by acting on the penicillin-binding protein. A previous study suggested that, compared with a single drug, carbapenem combined with sulbactam enhanced the antibacterial effect on CRAB in vitro and decreased the MIC value of carbapenem [15]. Our study found that although Abs was resistant to carbapenem, the 28-day mortality rate of patients after the addition of sulbactam was significantly reduced, which might be related to a mechanism by which sulbactam reduced the MIC value of carbapenem.\nIn addition, the combined dose of sulbactam needs to be further explored. A previous study found that when patients were given sulbactam 2 g q6h or 3 g q8h intravenously, the T > MIC (above 60%) was 98.83% and 95.59% respectively, if the sulbactam MIC was 16 µg/ml. Therefore, high-dose sulbactam was beneficial to controlling MDR-AB infection [16]. Another study recommended that a high-dose sulbactam (9 g/d) was also beneficial in controlling MDR-AB infection [17]. Our study found that the mortality rate of patients with CRAB pneumonia was lower in the high-dose sulbactam group as compared with the low-dose group, but the difference was not statistically significant. This might be related to the small number of cases in the high-dose sulbactam group. Thus, future studies regarding the dose of sulbactam need to include a larger number of cases.\nOur research has many limitations. First, we followed 1,823 clinical cases with positive AB culture results. Most of the cases were excluded because clinicians were not sure whether AB was the causative bacterium of severe pneumonia or only colonization in the lower respiratory tract. Especially at the same time, other bacterial and or fungal pathogens were cultivated from the lower respiratory tract. Second, only 166 cases were identified as severe MDR-AB pneumonia. After grouping, some data results did not reach statistical differences, for example, comparison of the clinical effects of high- and low-dose sulbactam for CRAB. Then, further mechanistic studies were needed, including the paper diffusion method or the antibiotic concentration gradient method (E-test method), to detect whether there was a synergistic effect between carbapenem and sulbactam, and different resistance mechanism in these CRAB strains.\nIn summary, despite the above shortcomings, ICU status, an extended period of invasive catheter retention time and mechanical ventilation time, and not using the appropriate antibacterial treatment increased the mortality in patients with severe hospital-acquired MDR-AB pneumonia. Although MDR-AB was resistant to many antibiotics, most of the MDR-AB strains in western China were still sensitive to tigecycline. The inclusion of tigecycline in the combined treatment plan could significantly reduce the 28-day mortality of patients. In addition, the inclusion of sulbactam in the treatment plan could also reduce the mortality of patients with carbapenemase-producing AB pneumonia on the basis of tigecycline. High-dose sulbactam might improve the clinical prognosis of patients by reducing the MIC of carbapenem drugs especially." ]
[ null, null, null, null, null, null, null, "results", null, null, null, null, null, null, null, "discussion" ]
[ "Multi-drug resistant Acinetobacter baumannii (MDR-AB)", "Carbapenem-resistant Acinetobacter baumannii(CRAB)", "Severe pneumonia", "Intensive care unit" ]
Neutrophil activation and NETosis are the predominant drivers of airway inflammation in an OVA/CFA/LPS induced murine model.
36271366
Asthma is one of the most common chronic diseases that affects more than 300 million people worldwide. Though most asthma can be well controlled, individuals with severe asthma experience recurrent exacerbations and impose a substantial economic burden on healthcare system. Neutrophil inflammation often occurs in patients with severe asthma who have poor response to glucocorticoids, increasing the difficulty of clinical treatment.
BACKGROUND
We established several neutrophil-dominated allergic asthma mouse models, and analyzed the airway hyperresponsiveness, airway inflammation and lung pathological changes. Neutrophil extracellular traps (NETs) formation was analyzed using confocal microscopy and western blot.
METHODS
We found that the ovalbumin (OVA)/complete Freund's adjuvant (CFA)/low-dose lipopolysaccharide (LPS)-induced mouse model best recapitulated the complex alterations in the airways of human severe asthmatic patients. We also observed OVA/CFA/LPS-exposed mice produced large quantities of neutrophil extracellular traps (NETs) in lung tissue and bone marrow neutrophils. Furthermore, we found that reducing the production of NETs or increasing the degradation of NETs can reduce airway inflammation and airway hyperresponsiveness.
RESULTS
Our findings identify a novel mouse model of neutrophilic asthma. We have also identified NETs play a significant role in neutrophilic asthma models and contribute to neutrophilic asthma pathogenesis. NETs may serve as a promising therapeutic target for neutrophilic asthma.
CONCLUSION
[ "Mice", "Humans", "Animals", "Ovalbumin", "Lipopolysaccharides", "Neutrophil Activation", "Freund's Adjuvant", "Disease Models, Animal", "Glucocorticoids", "Asthma", "Inflammation", "Respiratory Hypersensitivity" ]
9587569
Introduction
Asthma is a heterogeneous chronic respiratory disease, characterized by airway hyperresponsiveness (AHR) and airway inflammation [1]. There are four main inflammatory phenotypes in asthma based on the proportion of granulocytes in induced sputum: neutrophilic asthma, eosinophilic asthma, paucigranulocytic asthma, and mixed granulocytic asthma [2]. Neutrophilic asthma is one of the main types of severe asthma [3], which exhibits worse lung function, more severe airway inflammation, and worse treatment response. Therefore, it is crucial to investigation the pathogenesis of neutrophilic asthma. Experimental animal models are essential to advance asthma pathophysiological research, and mice are the main subjects. There are classical animal models of eosinophilic asthma, such as ovalbumin (OVA)/ aluminum (Alum)-sensitized and OVA-challenged model of asthma, house dust mite (HDM) extract-induced model of asthma and so on [4–7]. However, to the best of our knowledge, there is no animal model of neutrophilic asthma to be universally accepted. Currently, there are some animal studies of neutrophilic asthma, for example, OVA/complete Freund’s adjuvant (CFA)-sensitized and OVA-challenged model of asthma, OVA/ lipopolysaccharide (LPS)-sensitized and OVA-challenged model of asthma [8–10]. While these models recapitulate some of the features of human neutrophilic asthma, each of these models had certain limitations. Neutrophils are the main effector cells of inflammation and tissue infection [11]. The formation of neutrophil extracellular traps (NETs) is a function of neutrophils [12]. Neutrophils increase when inflammation occurs, and can undergo a process of NETosis, releasing NETs outside the cells [13]. However, whether NETs are generated and function in neutrophilic asthma have not been completely elucidated. Our study demonstrated that the OVA/CFA/LPS-induced murine model is suitable for study as a model of neutrophilic asthma. This kind of model has massive neutrophilic inflammation and severe airway hyperresponsiveness. In addition, we also found that a large number of NETs were produced and correlated with the severity of airway inflammation. Therefore, it is reasonable to hypothesize that NETs play an important functional role in neutrophilic asthma.
null
null
Results
[SUBTITLE] AHR and pathological changes induced by different allergens combined with OVA [SUBSECTION] To establish a suitable model to recapitulate as many clinical features as possible of human severe neutrophilic asthma, we constructed three different type of neutrophilic asthma model in BALB/c mice using CFA/OVA or LPS/OVA. In parallel, we established an eosinophilic asthma model as a control using Alum/OVA (Fig. 1A). Whole-body plethysmography was used to assess the AHR. The 10 µg LPS/OVA (10LPS) group exhibited a drastic increase in Penh upon exposure to increasing concentrations of aerosolized methacholine, as compared to the saline-treated (control) control group and CFA/OVA (CFA) group (Fig. 1B). The 0.1 µg LPS/OVA (0.1LPS) group and Alum/OVA (EOS) group also showed significantly increase in Penh upon exposure to aerosolized methacholine compared with the control group. Meanwhile, from that of normal controls, the CFA group showed increase in Penh but did not reach statistical significance. However, the severity of lung histopathology in the CFA group is not similar to its AHR. The CFA group displayed more severe bronchiolar and perivascular inflammation infiltration as assessed by H&E staining than the other four groups (Fig. 1C and 1E). The inflammatory cell infiltration in the 0.1LPS group and 10LPS group was also more than that in the control group, but lower than that in the CFA group. We assessed mucin secretion in the airway of mice by using PAS staining and also observed similar results to H&E staining (Fig. 1D and F). Fig. 1Different allergens combined with OVA-induced airway hyperresponsiveness (AHR) and pathological changes. (A) Schematic representation of experimental procedure. (B) The AHR was determined by calculating enhanced pause (Penh) values 24 h after the last challenge. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 Different allergens combined with OVA-induced airway hyperresponsiveness (AHR) and pathological changes. (A) Schematic representation of experimental procedure. (B) The AHR was determined by calculating enhanced pause (Penh) values 24 h after the last challenge. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 To establish a suitable model to recapitulate as many clinical features as possible of human severe neutrophilic asthma, we constructed three different type of neutrophilic asthma model in BALB/c mice using CFA/OVA or LPS/OVA. In parallel, we established an eosinophilic asthma model as a control using Alum/OVA (Fig. 1A). Whole-body plethysmography was used to assess the AHR. The 10 µg LPS/OVA (10LPS) group exhibited a drastic increase in Penh upon exposure to increasing concentrations of aerosolized methacholine, as compared to the saline-treated (control) control group and CFA/OVA (CFA) group (Fig. 1B). The 0.1 µg LPS/OVA (0.1LPS) group and Alum/OVA (EOS) group also showed significantly increase in Penh upon exposure to aerosolized methacholine compared with the control group. Meanwhile, from that of normal controls, the CFA group showed increase in Penh but did not reach statistical significance. However, the severity of lung histopathology in the CFA group is not similar to its AHR. The CFA group displayed more severe bronchiolar and perivascular inflammation infiltration as assessed by H&E staining than the other four groups (Fig. 1C and 1E). The inflammatory cell infiltration in the 0.1LPS group and 10LPS group was also more than that in the control group, but lower than that in the CFA group. We assessed mucin secretion in the airway of mice by using PAS staining and also observed similar results to H&E staining (Fig. 1D and F). Fig. 1Different allergens combined with OVA-induced airway hyperresponsiveness (AHR) and pathological changes. (A) Schematic representation of experimental procedure. (B) The AHR was determined by calculating enhanced pause (Penh) values 24 h after the last challenge. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 Different allergens combined with OVA-induced airway hyperresponsiveness (AHR) and pathological changes. (A) Schematic representation of experimental procedure. (B) The AHR was determined by calculating enhanced pause (Penh) values 24 h after the last challenge. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 [SUBTITLE] The ability of CFA to induce neutrophil inflammation is stronger than that of LPS [SUBSECTION] To further validate the inflammatory phenotype of the model, we also performed total and differential cell counts of BALF. Results demonstrated that the total number of cells and the percentage of neutrophils were significantly increased in the three groups of neutrophil models, particularly the CFA group (Fig. 2A). The percentage of eosinophils in the BALF of the three groups of neutrophil models had no significant difference compared with the control group, the EOS group of that were markedly increased (Fig. 2A). Moreover, for more precise evaluate the inflammatory phenotype of each group, we performed flow cytometry on cells of the BALF and single lung-cell suspensions of mice. Neutrophils was gated as viable CD45(+)CD11b(+)Ly6G(+) cells (Fig. 2B). Although the percentage of neutrophils in CD45(+) leukocytes increased in the BALF and single lung-cell suspensions of the three groups of neutrophil models compared to the control group, only the CFA group had a statistically significant difference (Fig. 2C and 2D). Similarly, only the percentage of neutrophils in CD45(+) leukocytes of the CFA group was statistically significant compared to the eosinophil group (Fig. 2 C and D). We next assessed the impact of different allergens on inflammatory cytokines in BALF. As a marker of Th2 polarization, IL-4 was obviously increased in the EOS group, while there was no significant change in the three neutrophil groups. Unlike that, the three neutrophil groups, especially the CFA group, had significantly higher IL-17 A, IL-6 and IL-1β expression than the control or EOS group (Fig. 2E). Fig. 2Neutrophil inflammatory infiltration was more pronounced in the CFA-induced neutrophil mouse models. (A) The number of total cells, eosinophils and neutrophils in BALF were quantified at 48 h after the last challenge. (B) Gating strategy for neutrophils is define as cells with the following characteristics: CD45(+)CD11b(+)Ly6G(+). (C) Neutrophil expression in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images in each group are shown. (D) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of BALF and lung tissue by flow cytometry. (E) The cytokines interleukin (IL)-4, IL-17A, IL-6 and IL-1β levels in BALF were measured by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (E); n = 4 in (C) and (D). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 Neutrophil inflammatory infiltration was more pronounced in the CFA-induced neutrophil mouse models. (A) The number of total cells, eosinophils and neutrophils in BALF were quantified at 48 h after the last challenge. (B) Gating strategy for neutrophils is define as cells with the following characteristics: CD45(+)CD11b(+)Ly6G(+). (C) Neutrophil expression in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images in each group are shown. (D) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of BALF and lung tissue by flow cytometry. (E) The cytokines interleukin (IL)-4, IL-17A, IL-6 and IL-1β levels in BALF were measured by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (E); n = 4 in (C) and (D). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 To further validate the inflammatory phenotype of the model, we also performed total and differential cell counts of BALF. Results demonstrated that the total number of cells and the percentage of neutrophils were significantly increased in the three groups of neutrophil models, particularly the CFA group (Fig. 2A). The percentage of eosinophils in the BALF of the three groups of neutrophil models had no significant difference compared with the control group, the EOS group of that were markedly increased (Fig. 2A). Moreover, for more precise evaluate the inflammatory phenotype of each group, we performed flow cytometry on cells of the BALF and single lung-cell suspensions of mice. Neutrophils was gated as viable CD45(+)CD11b(+)Ly6G(+) cells (Fig. 2B). Although the percentage of neutrophils in CD45(+) leukocytes increased in the BALF and single lung-cell suspensions of the three groups of neutrophil models compared to the control group, only the CFA group had a statistically significant difference (Fig. 2C and 2D). Similarly, only the percentage of neutrophils in CD45(+) leukocytes of the CFA group was statistically significant compared to the eosinophil group (Fig. 2 C and D). We next assessed the impact of different allergens on inflammatory cytokines in BALF. As a marker of Th2 polarization, IL-4 was obviously increased in the EOS group, while there was no significant change in the three neutrophil groups. Unlike that, the three neutrophil groups, especially the CFA group, had significantly higher IL-17 A, IL-6 and IL-1β expression than the control or EOS group (Fig. 2E). Fig. 2Neutrophil inflammatory infiltration was more pronounced in the CFA-induced neutrophil mouse models. (A) The number of total cells, eosinophils and neutrophils in BALF were quantified at 48 h after the last challenge. (B) Gating strategy for neutrophils is define as cells with the following characteristics: CD45(+)CD11b(+)Ly6G(+). (C) Neutrophil expression in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images in each group are shown. (D) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of BALF and lung tissue by flow cytometry. (E) The cytokines interleukin (IL)-4, IL-17A, IL-6 and IL-1β levels in BALF were measured by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (E); n = 4 in (C) and (D). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 Neutrophil inflammatory infiltration was more pronounced in the CFA-induced neutrophil mouse models. (A) The number of total cells, eosinophils and neutrophils in BALF were quantified at 48 h after the last challenge. (B) Gating strategy for neutrophils is define as cells with the following characteristics: CD45(+)CD11b(+)Ly6G(+). (C) Neutrophil expression in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images in each group are shown. (D) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of BALF and lung tissue by flow cytometry. (E) The cytokines interleukin (IL)-4, IL-17A, IL-6 and IL-1β levels in BALF were measured by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (E); n = 4 in (C) and (D). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 [SUBTITLE] The neutrophil mouse models developed obvious NETosis [SUBSECTION] To further explore the mechanism of neutrophils in mouse model of neutrophilic asthma, we first assessed neutrophil infiltration in mouse peripheral blood and bone marrow by flow cytometry. Results revealed that the percentage of neutrophils in the CFA group in both peripheral blood and bone marrow were found to be highest among all five groups. Although the percentage of neutrophils in the 0.1LPS group and the 10LPS group increased significantly relative to the control group, it was not as significant as the CFA group. In the flow cytometry analysis of bone marrow cells, the percentage of CD11b(+)Ly6G(+) neutrophils in the 0.1LPS group and 10LPS group lower than that in the EOS group (Fig. 3A and 3B). As the first line of defense, neutrophils partially mediate host responses by establishing NETs [20]. PMA is a very powerful, but non-physiological stimulus for NET formation [18]. We next purified mouse bone marrow neutrophils and stimulated with PMA (100 nM) in vitro to see whether NETs were released. Confocal microscopy revealed that a large number of NETosis neutrophils and NETs were detected in all three neutrophil models, with the highest amount of that produced in the CFA group. There was a small number of NETs observed in the EOS group (Fig. 3C-F). We also found that only a minority of neutrophils developed NETosis in the absence of stimulation, whether isolated from the bone marrow of the neutrophil models or other models. In addition, we detected the expression levels of MPO and CitH3 using western blot in lung tissue of mice, and the results were consistent with that of immunofluorescence analysis (Fig. 3G and H). Fig. 3Abundant NETs occur in neutrophilic asthma models. (A) Neutrophil expression in CD45(+) leucocytes in mouse peripheral blood and bone marrow were determined by flow cytometry within 48 h after the last challenge. The representative images in each group are shown. (B) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of mouse peripheral blood and bone marrow by flow cytometry. (C) In vitro NET-formation assays with purified neutrophils of mouse bone marrow neutrophils stimulated with PMA (100 nM) or RPMI 1640 for 4 h. Then the neutrophils were stained PI and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (D) Comparison of the percentage of NETosis cells in each group upon PMA stimulation and control (Ctrl) stimulation. (E) After stimulated with PMA, the neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green) and DAPI (nuclear staining, blue) and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (F) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (G, H) Western blot was used to detect the levels of MPO and CitH3 protein in lung tissue of five groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S1 and S4 presents the full-length blots. Data were shown as mean ± SEM; n = 4. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 Abundant NETs occur in neutrophilic asthma models. (A) Neutrophil expression in CD45(+) leucocytes in mouse peripheral blood and bone marrow were determined by flow cytometry within 48 h after the last challenge. The representative images in each group are shown. (B) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of mouse peripheral blood and bone marrow by flow cytometry. (C) In vitro NET-formation assays with purified neutrophils of mouse bone marrow neutrophils stimulated with PMA (100 nM) or RPMI 1640 for 4 h. Then the neutrophils were stained PI and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (D) Comparison of the percentage of NETosis cells in each group upon PMA stimulation and control (Ctrl) stimulation. (E) After stimulated with PMA, the neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green) and DAPI (nuclear staining, blue) and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (F) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (G, H) Western blot was used to detect the levels of MPO and CitH3 protein in lung tissue of five groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S1 and S4 presents the full-length blots. Data were shown as mean ± SEM; n = 4. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 To further explore the mechanism of neutrophils in mouse model of neutrophilic asthma, we first assessed neutrophil infiltration in mouse peripheral blood and bone marrow by flow cytometry. Results revealed that the percentage of neutrophils in the CFA group in both peripheral blood and bone marrow were found to be highest among all five groups. Although the percentage of neutrophils in the 0.1LPS group and the 10LPS group increased significantly relative to the control group, it was not as significant as the CFA group. In the flow cytometry analysis of bone marrow cells, the percentage of CD11b(+)Ly6G(+) neutrophils in the 0.1LPS group and 10LPS group lower than that in the EOS group (Fig. 3A and 3B). As the first line of defense, neutrophils partially mediate host responses by establishing NETs [20]. PMA is a very powerful, but non-physiological stimulus for NET formation [18]. We next purified mouse bone marrow neutrophils and stimulated with PMA (100 nM) in vitro to see whether NETs were released. Confocal microscopy revealed that a large number of NETosis neutrophils and NETs were detected in all three neutrophil models, with the highest amount of that produced in the CFA group. There was a small number of NETs observed in the EOS group (Fig. 3C-F). We also found that only a minority of neutrophils developed NETosis in the absence of stimulation, whether isolated from the bone marrow of the neutrophil models or other models. In addition, we detected the expression levels of MPO and CitH3 using western blot in lung tissue of mice, and the results were consistent with that of immunofluorescence analysis (Fig. 3G and H). Fig. 3Abundant NETs occur in neutrophilic asthma models. (A) Neutrophil expression in CD45(+) leucocytes in mouse peripheral blood and bone marrow were determined by flow cytometry within 48 h after the last challenge. The representative images in each group are shown. (B) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of mouse peripheral blood and bone marrow by flow cytometry. (C) In vitro NET-formation assays with purified neutrophils of mouse bone marrow neutrophils stimulated with PMA (100 nM) or RPMI 1640 for 4 h. Then the neutrophils were stained PI and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (D) Comparison of the percentage of NETosis cells in each group upon PMA stimulation and control (Ctrl) stimulation. (E) After stimulated with PMA, the neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green) and DAPI (nuclear staining, blue) and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (F) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (G, H) Western blot was used to detect the levels of MPO and CitH3 protein in lung tissue of five groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S1 and S4 presents the full-length blots. Data were shown as mean ± SEM; n = 4. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 Abundant NETs occur in neutrophilic asthma models. (A) Neutrophil expression in CD45(+) leucocytes in mouse peripheral blood and bone marrow were determined by flow cytometry within 48 h after the last challenge. The representative images in each group are shown. (B) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of mouse peripheral blood and bone marrow by flow cytometry. (C) In vitro NET-formation assays with purified neutrophils of mouse bone marrow neutrophils stimulated with PMA (100 nM) or RPMI 1640 for 4 h. Then the neutrophils were stained PI and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (D) Comparison of the percentage of NETosis cells in each group upon PMA stimulation and control (Ctrl) stimulation. (E) After stimulated with PMA, the neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green) and DAPI (nuclear staining, blue) and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (F) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (G, H) Western blot was used to detect the levels of MPO and CitH3 protein in lung tissue of five groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S1 and S4 presents the full-length blots. Data were shown as mean ± SEM; n = 4. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 [SUBTITLE] The neutrophilic asthma model induced by 0.1LPS and CFA combined with OVA exhibited significant AHR and severe airway infiltration [SUBSECTION] We summarize the above results and conclude that the CFA group is a relatively more suitable model of neutrophilic asthma, although the AHR of this model is not significant. To overcome this, we attempted to establish a new murine model combining the sensitization methods of the 0.1LPS group and the CFA group (0.1LPS + CFA group) (Fig. 4A). Result shown that the 0.1LPS + CFA group exhibited higher AHR in response to methacholine compared with the 0.1LPS or CFA group, although the differences were not statistically significant (Fig. 4B). Similar to the CFA group, lung histopathological examination in 0.1LPS + CFA group showed obvious inflammatory cells infiltration and airway mucus secretion, some airways showed local airway mucus obstruction (Fig. 4C-F). These results seem to indicate that the 0.1LPS + CFA group serve as a suitable model of mouse to study the pathogenesis of the neutrophilic asthma. Fig. 4The 0.1LPS and CFA combined with OVA-induced neutrophilic asthma model exhibits significant AHR and severe airway infiltration. (A) Schematic diagram of the experiment. (B) The AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used to assess impaired lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 The 0.1LPS and CFA combined with OVA-induced neutrophilic asthma model exhibits significant AHR and severe airway infiltration. (A) Schematic diagram of the experiment. (B) The AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used to assess impaired lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 We summarize the above results and conclude that the CFA group is a relatively more suitable model of neutrophilic asthma, although the AHR of this model is not significant. To overcome this, we attempted to establish a new murine model combining the sensitization methods of the 0.1LPS group and the CFA group (0.1LPS + CFA group) (Fig. 4A). Result shown that the 0.1LPS + CFA group exhibited higher AHR in response to methacholine compared with the 0.1LPS or CFA group, although the differences were not statistically significant (Fig. 4B). Similar to the CFA group, lung histopathological examination in 0.1LPS + CFA group showed obvious inflammatory cells infiltration and airway mucus secretion, some airways showed local airway mucus obstruction (Fig. 4C-F). These results seem to indicate that the 0.1LPS + CFA group serve as a suitable model of mouse to study the pathogenesis of the neutrophilic asthma. Fig. 4The 0.1LPS and CFA combined with OVA-induced neutrophilic asthma model exhibits significant AHR and severe airway infiltration. (A) Schematic diagram of the experiment. (B) The AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used to assess impaired lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 The 0.1LPS and CFA combined with OVA-induced neutrophilic asthma model exhibits significant AHR and severe airway infiltration. (A) Schematic diagram of the experiment. (B) The AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used to assess impaired lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 [SUBTITLE] CFA combined with LPS induced strong neutrophilic inflammation and secretion of proinflammatory factors [SUBSECTION] Immediately after, we performed BALF and lung tissue cells analysis. The total number of BALF in the 0.1LPS + CFA group was significantly higher than that in the control and 0.1LPS groups, and there was no significant difference in total number of BALF between the 0.1LPS + CFA and CFA groups (Fig. 5A). Similar results were observed between the groups in the percentage of neutrophils (Fig. 5A). The percentage of eosinophils in the BALF of the three neutrophil groups had no significant difference compared with each other, while higher than that in the control group (Fig. 5A). Likewise, we performed flow cytometry on BALF cells and single lung-cell suspensions to better understand our newly established neutrophil model. The percentage of neutrophils in CD45(+) leukocytes in the BALF and single lung-cell suspensions in both 0.1LPS + CFA and CFA groups was significantly increased, and the difference was statistically significant (Fig. 5B and C). The 0.1LPS group also shown more percentage of neutrophils in CD45(+) leukocytes compared with the control group, but no statistical significance was achieved (Fig. 5B and C). Next, we quantification the expression of inflammatory cytokines in BALF. As expected, the expression of IL-17A, IL-6 and IL-1β in the 0.1LPS + CFA group and other two neutrophil groups were strongly increased compared with the control group. However, the expression of IL-4 was not significantly different in all four groups (Fig. 5D). Fig. 5Significant neutrophilic inflammation and secretion of proinflammatory factors were observed in CFA combined with LPS induced mouse model. (A) Quantification of total cell counts and differential cell counts (eosinophils and neutrophils) in BALF at 48 h after the last challenge. (B) The percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images of each group are shown. (C) Statistical analysis of the above data shown by flow cytometry. (D) Cytokine (IL-4, IL-6 and IL-1β) concentrations in BALF were quantified by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (D); n = 4 mice in (B) and (C). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 Significant neutrophilic inflammation and secretion of proinflammatory factors were observed in CFA combined with LPS induced mouse model. (A) Quantification of total cell counts and differential cell counts (eosinophils and neutrophils) in BALF at 48 h after the last challenge. (B) The percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images of each group are shown. (C) Statistical analysis of the above data shown by flow cytometry. (D) Cytokine (IL-4, IL-6 and IL-1β) concentrations in BALF were quantified by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (D); n = 4 mice in (B) and (C). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 Immediately after, we performed BALF and lung tissue cells analysis. The total number of BALF in the 0.1LPS + CFA group was significantly higher than that in the control and 0.1LPS groups, and there was no significant difference in total number of BALF between the 0.1LPS + CFA and CFA groups (Fig. 5A). Similar results were observed between the groups in the percentage of neutrophils (Fig. 5A). The percentage of eosinophils in the BALF of the three neutrophil groups had no significant difference compared with each other, while higher than that in the control group (Fig. 5A). Likewise, we performed flow cytometry on BALF cells and single lung-cell suspensions to better understand our newly established neutrophil model. The percentage of neutrophils in CD45(+) leukocytes in the BALF and single lung-cell suspensions in both 0.1LPS + CFA and CFA groups was significantly increased, and the difference was statistically significant (Fig. 5B and C). The 0.1LPS group also shown more percentage of neutrophils in CD45(+) leukocytes compared with the control group, but no statistical significance was achieved (Fig. 5B and C). Next, we quantification the expression of inflammatory cytokines in BALF. As expected, the expression of IL-17A, IL-6 and IL-1β in the 0.1LPS + CFA group and other two neutrophil groups were strongly increased compared with the control group. However, the expression of IL-4 was not significantly different in all four groups (Fig. 5D). Fig. 5Significant neutrophilic inflammation and secretion of proinflammatory factors were observed in CFA combined with LPS induced mouse model. (A) Quantification of total cell counts and differential cell counts (eosinophils and neutrophils) in BALF at 48 h after the last challenge. (B) The percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images of each group are shown. (C) Statistical analysis of the above data shown by flow cytometry. (D) Cytokine (IL-4, IL-6 and IL-1β) concentrations in BALF were quantified by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (D); n = 4 mice in (B) and (C). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 Significant neutrophilic inflammation and secretion of proinflammatory factors were observed in CFA combined with LPS induced mouse model. (A) Quantification of total cell counts and differential cell counts (eosinophils and neutrophils) in BALF at 48 h after the last challenge. (B) The percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images of each group are shown. (C) Statistical analysis of the above data shown by flow cytometry. (D) Cytokine (IL-4, IL-6 and IL-1β) concentrations in BALF were quantified by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (D); n = 4 mice in (B) and (C). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 [SUBTITLE] The OVA/CFA/LPS-induced mice model developed obvious NETosis [SUBSECTION] Likewise, to investigate whether neutrophils in the 0.1LPS + CFA group asthma mouse model also have a similar mechanism to the neutrophil groups described above, we also evaluated neutrophil infiltration in mouse peripheral blood and bone marrow by flow cytometry. Results demonstrate that the percentage of neutrophils in peripheral blood and bone marrow in the 0.1LPS + CFA group was significantly higher than that in the control group, which was comparable to the CFA group. The percentage of neutrophils in the 0.1LPS group was increased compared with the control group, but there was no statistical difference (Fig. 6A and B). Confocal microscopy demonstrated that NETs co-localized with MPO, citH3 and DAPI were detected in all three neutrophil models after PMA stimulation, while the 0.1LPS + CFA group and CFA group could observe the most significant NETs release (Fig. 6C-E). Western blot also showed that the protein expression levels of MPO and CitH3 in the lung tissue of mice in each group were consistent with the results of immunofluorescence (Fig. 6F and G). Fig. 6Neutrophilic mouse model induced by CFA combined with LPS showed enhanced NETs formation capacity. (A) At 48 h after the last challenge, the percentage of neutrophil populations in CD45(+) leukocytes in mouse peripheral blood and bone marrow were determined by flow cytometry. The representative images in each group are shown. (B) Statistical analysis of the percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leukocyte gate of mouse peripheral blood and bone marrow by flow cytometry. (C) Neutrophils were purified from mouse bone marrow and stimulated with PMA (100 nM) or vehicle control for 4 h. Then, neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green), and DNA (DAPI, blue) and confocal by immunofluorescence microscope for analysis. Representative images of NETs immunofluorescence. Scale bar = 20 μm. (D) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (E) Representative z-axis images of the NETs immunofluorescence in 0.1LPS + CFA group. Scale bar = 10 μm. (F, G) Western blot analysis the protein expression level of MPO and CitH3 in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S2 and S5 presents the full-length blots. Data were shown as mean ± SEM; n = 4 or 6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 Neutrophilic mouse model induced by CFA combined with LPS showed enhanced NETs formation capacity. (A) At 48 h after the last challenge, the percentage of neutrophil populations in CD45(+) leukocytes in mouse peripheral blood and bone marrow were determined by flow cytometry. The representative images in each group are shown. (B) Statistical analysis of the percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leukocyte gate of mouse peripheral blood and bone marrow by flow cytometry. (C) Neutrophils were purified from mouse bone marrow and stimulated with PMA (100 nM) or vehicle control for 4 h. Then, neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green), and DNA (DAPI, blue) and confocal by immunofluorescence microscope for analysis. Representative images of NETs immunofluorescence. Scale bar = 20 μm. (D) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (E) Representative z-axis images of the NETs immunofluorescence in 0.1LPS + CFA group. Scale bar = 10 μm. (F, G) Western blot analysis the protein expression level of MPO and CitH3 in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S2 and S5 presents the full-length blots. Data were shown as mean ± SEM; n = 4 or 6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 Likewise, to investigate whether neutrophils in the 0.1LPS + CFA group asthma mouse model also have a similar mechanism to the neutrophil groups described above, we also evaluated neutrophil infiltration in mouse peripheral blood and bone marrow by flow cytometry. Results demonstrate that the percentage of neutrophils in peripheral blood and bone marrow in the 0.1LPS + CFA group was significantly higher than that in the control group, which was comparable to the CFA group. The percentage of neutrophils in the 0.1LPS group was increased compared with the control group, but there was no statistical difference (Fig. 6A and B). Confocal microscopy demonstrated that NETs co-localized with MPO, citH3 and DAPI were detected in all three neutrophil models after PMA stimulation, while the 0.1LPS + CFA group and CFA group could observe the most significant NETs release (Fig. 6C-E). Western blot also showed that the protein expression levels of MPO and CitH3 in the lung tissue of mice in each group were consistent with the results of immunofluorescence (Fig. 6F and G). Fig. 6Neutrophilic mouse model induced by CFA combined with LPS showed enhanced NETs formation capacity. (A) At 48 h after the last challenge, the percentage of neutrophil populations in CD45(+) leukocytes in mouse peripheral blood and bone marrow were determined by flow cytometry. The representative images in each group are shown. (B) Statistical analysis of the percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leukocyte gate of mouse peripheral blood and bone marrow by flow cytometry. (C) Neutrophils were purified from mouse bone marrow and stimulated with PMA (100 nM) or vehicle control for 4 h. Then, neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green), and DNA (DAPI, blue) and confocal by immunofluorescence microscope for analysis. Representative images of NETs immunofluorescence. Scale bar = 20 μm. (D) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (E) Representative z-axis images of the NETs immunofluorescence in 0.1LPS + CFA group. Scale bar = 10 μm. (F, G) Western blot analysis the protein expression level of MPO and CitH3 in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S2 and S5 presents the full-length blots. Data were shown as mean ± SEM; n = 4 or 6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 Neutrophilic mouse model induced by CFA combined with LPS showed enhanced NETs formation capacity. (A) At 48 h after the last challenge, the percentage of neutrophil populations in CD45(+) leukocytes in mouse peripheral blood and bone marrow were determined by flow cytometry. The representative images in each group are shown. (B) Statistical analysis of the percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leukocyte gate of mouse peripheral blood and bone marrow by flow cytometry. (C) Neutrophils were purified from mouse bone marrow and stimulated with PMA (100 nM) or vehicle control for 4 h. Then, neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green), and DNA (DAPI, blue) and confocal by immunofluorescence microscope for analysis. Representative images of NETs immunofluorescence. Scale bar = 20 μm. (D) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (E) Representative z-axis images of the NETs immunofluorescence in 0.1LPS + CFA group. Scale bar = 10 μm. (F, G) Western blot analysis the protein expression level of MPO and CitH3 in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S2 and S5 presents the full-length blots. Data were shown as mean ± SEM; n = 4 or 6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 [SUBTITLE] DNase I or CI-amidine administration protects mice of neutrophilic asthma model [SUBSECTION] To determine whether the presence of NETs contributes to the pathogenesis of neutrophilic asthma model, we treated mice with either DNase I or CI-amidine before each challenge (Fig. 7 A). Previous studies have shown that DNase I can break down NETs [21], while activation of PAD4 is a major driver of NETs formation, and CI-amidine is an irreversible PAD4 inhibitor [22, 23]. Here, we found that DNase I or CI-amidine treatment significantly reduced airway hyperresponsiveness (Fig. 7B) and alleviated airway inflammation in the 0.1LPS + CFA group (Fig. 7C and E), PAS staining showed that airway mucus obstruction disappeared (Fig. 7D and F). Analysis of inflammatory cells in BALF also indicated that both the total number of cells and the percentage of neutrophils were significantly reduced by DNase I or CI-amidine treatment (Fig. 7G). Western blot analysis of lung tissue showed that the expression of MPO and CitH3 were significantly reduced in the OVA/CFA/0.1 LPS + DNase I (DNase I) group or the OVA/CFA/0.1 LPS + CI-amidine (CI-amidine) group (Fig. 7 H and I). Fig. 7DNase I or CI-amidine administration reduced airway hyperresponsiveness and alleviated airway inflammation (A) Experimental assay schematic for in vivo experiments. (B) AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used as an indicator of lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. (G) The total number of cells and the differential number of cells (eosinophils and neutrophils) were quantified 48 h after the last challenge in the BALF. (H, I) Western blot analysis to measure MPO and CitH3 protein expression level in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S3 and S6 presents the full-length blots. Data were shown as mean ± SEM; n = 4–6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 DNase I or CI-amidine administration reduced airway hyperresponsiveness and alleviated airway inflammation (A) Experimental assay schematic for in vivo experiments. (B) AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used as an indicator of lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. (G) The total number of cells and the differential number of cells (eosinophils and neutrophils) were quantified 48 h after the last challenge in the BALF. (H, I) Western blot analysis to measure MPO and CitH3 protein expression level in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S3 and S6 presents the full-length blots. Data were shown as mean ± SEM; n = 4–6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 To determine whether the presence of NETs contributes to the pathogenesis of neutrophilic asthma model, we treated mice with either DNase I or CI-amidine before each challenge (Fig. 7 A). Previous studies have shown that DNase I can break down NETs [21], while activation of PAD4 is a major driver of NETs formation, and CI-amidine is an irreversible PAD4 inhibitor [22, 23]. Here, we found that DNase I or CI-amidine treatment significantly reduced airway hyperresponsiveness (Fig. 7B) and alleviated airway inflammation in the 0.1LPS + CFA group (Fig. 7C and E), PAS staining showed that airway mucus obstruction disappeared (Fig. 7D and F). Analysis of inflammatory cells in BALF also indicated that both the total number of cells and the percentage of neutrophils were significantly reduced by DNase I or CI-amidine treatment (Fig. 7G). Western blot analysis of lung tissue showed that the expression of MPO and CitH3 were significantly reduced in the OVA/CFA/0.1 LPS + DNase I (DNase I) group or the OVA/CFA/0.1 LPS + CI-amidine (CI-amidine) group (Fig. 7 H and I). Fig. 7DNase I or CI-amidine administration reduced airway hyperresponsiveness and alleviated airway inflammation (A) Experimental assay schematic for in vivo experiments. (B) AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used as an indicator of lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. (G) The total number of cells and the differential number of cells (eosinophils and neutrophils) were quantified 48 h after the last challenge in the BALF. (H, I) Western blot analysis to measure MPO and CitH3 protein expression level in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S3 and S6 presents the full-length blots. Data were shown as mean ± SEM; n = 4–6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001 DNase I or CI-amidine administration reduced airway hyperresponsiveness and alleviated airway inflammation (A) Experimental assay schematic for in vivo experiments. (B) AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used as an indicator of lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. (G) The total number of cells and the differential number of cells (eosinophils and neutrophils) were quantified 48 h after the last challenge in the BALF. (H, I) Western blot analysis to measure MPO and CitH3 protein expression level in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S3 and S6 presents the full-length blots. Data were shown as mean ± SEM; n = 4–6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001
Conclusion
In conclusion, our findings identify a novel mouse model of neutrophilic asthma induced by OVA, CFA and LPS. This kind of neutrophilic asthma model is phenotypically more comprehensive, and characterized by AHR, massive infiltration of neutrophils and production of non-Th2 cytokines. Our data also suggest that a large number of NETs are generated in vivo and in vitro in the neutrophilic asthma models, reducing NETs can reverse those changes mentioned above. These findings provide further insight into the asthma model studies and pathogenesis of neutrophilic asthma, targeting NETs is a novel strategy that may be effective for treating neutrophilic asthma in the future.
[ "Animals", "Mouse allergen sensitization and challenge", "Airway hyperresponsiveness measurement", "Bronchoalveolar lavage", "Cytokine analysis", "Lung histopathology staining", "Flow cytometry analysis", "Immunostaining and confocal microscopy", "Western blot", "Statistical analysis", "AHR and pathological changes induced by different allergens combined with OVA", "The ability of CFA to induce neutrophil inflammation is stronger than that of LPS", "The neutrophil mouse models developed obvious NETosis", "The neutrophilic asthma model induced by 0.1LPS and CFA combined with OVA exhibited significant AHR and severe airway infiltration", "CFA combined with LPS induced strong neutrophilic inflammation and secretion of proinflammatory factors", "The OVA/CFA/LPS-induced mice model developed obvious NETosis", "DNase I or CI-amidine administration protects mice of neutrophilic asthma model", "" ]
[ "BALB/c mice (female, 6-8 weeks) were purchased from Hangzhou Medical College (Hangzhou, China). We maintained all the mice in specific pathogen-free (SPF) conditions with a 12 light/12 dark cycle in the Experimental Animal Center, the First Affiliated Hospital, School of Medicine, Zhejiang University.", "Mouse models were induced using modified protocols as previously reported [7–9]. Mice were randomly grouped and sensitized on day 1 and day 8. For the OVA/Alum group of asthma, 25 µg OVA (A5503, Sigma Aldrich, St. Louis, MO, USA) dissolved in 100 µl 0.9% saline was mixed 1:1 with Imject Alum (Pierce, Rockford, IL, USA) by intraperitoneal (IP) injection. For the OVA/CFA group of asthma, 25 µg OVA dissolved in 100 µl 0.9% saline was mixed 1:1 with CFA (F5881, Sigma Aldrich, St. Louis, MO, USA) by IP. injection. For the OVA/LPS group of asthma, mice were lightly anesthetized with isoflurane, and intratracheally injected using a combination of 25 µg OVA with 0.1 or 10 µg LPS (L2630, Sigma Aldrich, St. Louis, MO, USA) in a total volume of 40 µl, with 0.9% saline as the diluent. For the OVA/CFA/0.1 LPS group, the model of sensitization is a combination of the OVA/CFA group and OVA/LPS group. After sensitization, the above-mentioned groups were challenged with aerosolized 1% OVA for 30 min on days 15-17. For the OVA/CFA/0.1 LPS + DNase I group or the OVA/CFA/0.1 LPS + CI-amidine group, on the basis of the OVA/CFA/0.1 LPS group, intravenous injection of DNase I (5 mg per kg body weight) or intraperitoneal injection of Cl-amidine (10 mg per kg body weight) was performed 1 h before each challenge. Mice sensitized and challenged with 0.9% saline were used as controls.", "Airway responsiveness was assessed 24 h after the last OVA challenge as previously described [14]. Briefly, mice were placed in a plethysmograph chamber (EMKA Technologies, Paris, France) for at least 10 min to adaption. Baseline pulmonary parameters were first determined, and then mice were sequentially challenged with aerosolized PBS and methacholine (Mch, A2251, Sigma Aldrich, St. Louis, MO, USA) at increased concentrations (3.125, 6.25, 12.5, 25 and 50 mg/ml in PBS). Enhanced Pause (Penh), an indirect estimate of airway resistance, was used to measure airway resistance to methacholine. After each nebulization, record the Penh value for 3 min and take the average of three consecutive values.", "Mice were euthanized 48 h after the last OVA challenge, serum, bronchoalveolar lavage fluid (BALF), bone marrow and lung tissues were collected for further experiments. As described previously [14], BALF was collected on whole lungs by infusing 1 ml of PBS via a tracheal cannula. Then, the BALF was centrifuged, and the supernatants were stored at-80 °C until analysis, the cell pellet was resuspended in PBS for differential cell counting or flow cytometry analysis.", "Expression of interleukin (IL)-4 (431,104), IL-17 A (432,504), IL-6 (431,304), and IL-1β (432,604) in BALF were quantitated by enzyme-linked immunosorbent assay (ELISA) kit (Biolegend, San Diego, CA, USA) according to the manufacturers’ instructions.", "After the BALF was collected, the right main bronchus was ligated, and the left lung was perfused and fixed with 4% paraformaldehyde for 24 h, followed by paraffin embedded, sectioned, stained with hematoxylin and eosin (H&E) or paraffin acid-Schiff (PAS) staining. Sections were performed in a blinded fashion. The degree of inflammation on H&E-stained lung sections was scored as described previously [15]: 0, normal; 1, few inflammatory cells; 2, a ring of inflammatory cells 1 cell layer deep; 3, a ring of inflammatory cells 2 to 4 cells deep; 4, a ring of inflammatory cells greater than 4 cells deep. Mucus-containing goblet cells on PAS-stained lung sections was also scored previously described [16]: 0, no PAS-positive cells; 1, less than 25%; 2, 25 to 50% PAS-positive cells; 3, 50 to 80% PAS-positive cells; 4, greater than 80% PAS-positive cells.", "Flow cytometry of leukocytes in BALF, lung tissue, peripheral blood and bone marrow. The BALF were collected according to the above procedure. Lung single-cell suspensions were obtained as previously described [17]. The peripheral blood was collected from mice by removed their eyeball and placed in tubes containing EDTA. Red blood cells (RBCs) were lysed with RBC lysis buffer (Biolegend, 420,301, San Diego, CA, USA). The bone marrow cells were harvested from the femora and tibiae by flushing with PBS, then cells filtered through 40 μm strainers (352,340, BD Biosciences, San Diego, CA, USA) to obtain single cell suspension. After a washing step, all single-cell suspensions were incubated with Fixable Viability Stain 780 (FVS780, 565,388, BD Biosciences, San Diego, CA, USA, 1:1000) according to manufacturer’s instructions to gate out dead cells. Subsequently, samples were stained with antibodies against CD45-FITC (157,214, Biolegend, San Diego, CA, USA, 1:100), CD11b-PE (101,207, Biolegend, San Diego, CA, USA, 1:100) and Ly6G-PE/Cy7 (560,601, BD Biosciences, San Diego, CA, USA, 1:100) for 30 min on ice, then cells were analyzed on a Cytoflex LX flow cytometer (Beckman Coulter, CA, USA). All above antibodies were diluted in PBS. Analyses were performed using FlowJo software v10.6.2. Neutrophils were identified as CD45(+)CD11b(+)Ly6G(+).", "Bone marrow cells from mice were harvested as describe above. Subsequently, neutrophils from bone marrow were isolated by density gradients according to the mouse neutrophil isolation kit instructions (TBD2013NM, TBD Science, China). The neutrophils (2 × 105 cells per well in serum free RPMI 1640) were plated on poly-lysine-coated round coverslips and placed in 24-well culture plates. Then, the cells were stimulated with 100 nM Phorbol 12-myristate 13-acetate (PMA, P3681, Sigma Aldrich, St. Louis, MO, USA) or RPMI 1640 (Ctrl) for 4 h. Subsequently, cells were fixed in 4% paraformaldehyde (PFA) for 30 min and permeabilized by incubation in 0.5% Triton X-100 at room temperature (RT) for 1 min. After that some samples were stained with propidium iodide (PI, 5 µg/ml, P4170, Sigma Aldrich, St. Louis, MO, USA) for 30 min, and washed in PBS before confocal microscopic observation. To quantify NET formation, NETosis was defined as neutrophils with flattened nuclei, decondensed chromatin and expulsion of extracellular neutrophils under fluorescence microscopy. Two independent researchers analyzed 200 neutrophils in each sample [18]. The other samples were blocked by blocking solution (phosphate-buffered saline (PBS) containing 1% bovine serum albumin (BSA) and 0.1% Tween-20) for 1 h at RT, samples were stained with rabbit anti-citrullinated histone 3 (CitH3) (ab5103, Abcam, 1:500) and mouse anti- Myeloperoxidase (MPO) (AF3667, R&D, 1:50) overnight at 4 °C. The next day, samples were washed in PBST and incubated with secondary antibodies CoraLite 594-conjugated Goat Anti-Rabbit IgG (H + L) (Proteintech, SA00013-4, 1:400) and CoraLite 488-conjugated Affinipure Goat Anti-Mouse IgG (H + L) (Proteintech, SA00013-1, 1:400). All above antibodies were diluted in blocking solution. The nuclei were stained with 4,6-diamidino-2-phenylindole (DAPI, 422,801, Biolegend 1:10000). The NETs were identified by the colocalization of antibodies (MPO, CitH3 and DAPI) and quantified using Fiji software v2.1.0. To determine the colocalization, we used a ratio by normalizing the percentage CitH3 signal to the percentage MPO signal, analysis of the CitH3/MPO percentage values from four regions was performed [19].", "Protein lysates of lung tissue were prepared. Then, the proteins were separated by 10-12% sodium dodecyl sulfatepolyacrylamide gel electrophoresis (SDS-PAGE), transferred onto polyvinylidene fluoride (PVDF) membranes. Membranes were blocked in 5% skim milk for 1 h RT, follow by incubated with primary antibodies overnight at 4 ℃. The following day, membranes were washed and incubated with HRP-conjugated secondary antibodies for 1 h and proteins were visualized using the ECL reagent. Rabbit anti-CitH3 (ab5103, 1:1000) was purchased from Abcam; Mouse anti-MPO (AF3667, 1:400) was purchased from R&D; beta (β)-Tubulin (66,240–1-Ig, 1:5000) was purchased from Proteintech Technology. All primary antibodies were diluted in primary antibody dilution buffer (Beyotime Biotechnology, China), and secondary antibodies were diluted in 5% skim milk.", "All statistics and graphs were performed using GraphPad Prism software v8.4.0 (GraphPad Software Inc., San Diego, CA, USA). In this study, a one-way ANOVA with Tukey posttests was used for multiple comparisons. Data are presented as means ± standard errors of measurement (SEM). Significant differences are shown as *P < 0.05, **P < 0.01, and ***P < 0.001.", "To establish a suitable model to recapitulate as many clinical features as possible of human severe neutrophilic asthma, we constructed three different type of neutrophilic asthma model in BALB/c mice using CFA/OVA or LPS/OVA. In parallel, we established an eosinophilic asthma model as a control using Alum/OVA (Fig. 1A). Whole-body plethysmography was used to assess the AHR. The 10 µg LPS/OVA (10LPS) group exhibited a drastic increase in Penh upon exposure to increasing concentrations of aerosolized methacholine, as compared to the saline-treated (control) control group and CFA/OVA (CFA) group (Fig. 1B). The 0.1 µg LPS/OVA (0.1LPS) group and Alum/OVA (EOS) group also showed significantly increase in Penh upon exposure to aerosolized methacholine compared with the control group. Meanwhile, from that of normal controls, the CFA group showed increase in Penh but did not reach statistical significance. However, the severity of lung histopathology in the CFA group is not similar to its AHR. The CFA group displayed more severe bronchiolar and perivascular inflammation infiltration as assessed by H&E staining than the other four groups (Fig. 1C and 1E). The inflammatory cell infiltration in the 0.1LPS group and 10LPS group was also more than that in the control group, but lower than that in the CFA group. We assessed mucin secretion in the airway of mice by using PAS staining and also observed similar results to H&E staining (Fig. 1D and F).\n\nFig. 1Different allergens combined with OVA-induced airway hyperresponsiveness (AHR) and pathological changes. (A) Schematic representation of experimental procedure. (B) The AHR was determined by calculating enhanced pause (Penh) values 24 h after the last challenge. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nDifferent allergens combined with OVA-induced airway hyperresponsiveness (AHR) and pathological changes. (A) Schematic representation of experimental procedure. (B) The AHR was determined by calculating enhanced pause (Penh) values 24 h after the last challenge. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "To further validate the inflammatory phenotype of the model, we also performed total and differential cell counts of BALF. Results demonstrated that the total number of cells and the percentage of neutrophils were significantly increased in the three groups of neutrophil models, particularly the CFA group (Fig. 2A). The percentage of eosinophils in the BALF of the three groups of neutrophil models had no significant difference compared with the control group, the EOS group of that were markedly increased (Fig. 2A). Moreover, for more precise evaluate the inflammatory phenotype of each group, we performed flow cytometry on cells of the BALF and single lung-cell suspensions of mice. Neutrophils was gated as viable CD45(+)CD11b(+)Ly6G(+) cells (Fig. 2B). Although the percentage of neutrophils in CD45(+) leukocytes increased in the BALF and single lung-cell suspensions of the three groups of neutrophil models compared to the control group, only the CFA group had a statistically significant difference (Fig. 2C and 2D). Similarly, only the percentage of neutrophils in CD45(+) leukocytes of the CFA group was statistically significant compared to the eosinophil group (Fig. 2 C and D). We next assessed the impact of different allergens on inflammatory cytokines in BALF. As a marker of Th2 polarization, IL-4 was obviously increased in the EOS group, while there was no significant change in the three neutrophil groups. Unlike that, the three neutrophil groups, especially the CFA group, had significantly higher IL-17 A, IL-6 and IL-1β expression than the control or EOS group (Fig. 2E).\n\nFig. 2Neutrophil inflammatory infiltration was more pronounced in the CFA-induced neutrophil mouse models. (A) The number of total cells, eosinophils and neutrophils in BALF were quantified at 48 h after the last challenge. (B) Gating strategy for neutrophils is define as cells with the following characteristics: CD45(+)CD11b(+)Ly6G(+). (C) Neutrophil expression in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images in each group are shown. (D) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of BALF and lung tissue by flow cytometry. (E) The cytokines interleukin (IL)-4, IL-17A, IL-6 and IL-1β levels in BALF were measured by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (E); n = 4 in (C) and (D). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nNeutrophil inflammatory infiltration was more pronounced in the CFA-induced neutrophil mouse models. (A) The number of total cells, eosinophils and neutrophils in BALF were quantified at 48 h after the last challenge. (B) Gating strategy for neutrophils is define as cells with the following characteristics: CD45(+)CD11b(+)Ly6G(+). (C) Neutrophil expression in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images in each group are shown. (D) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of BALF and lung tissue by flow cytometry. (E) The cytokines interleukin (IL)-4, IL-17A, IL-6 and IL-1β levels in BALF were measured by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (E); n = 4 in (C) and (D). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "To further explore the mechanism of neutrophils in mouse model of neutrophilic asthma, we first assessed neutrophil infiltration in mouse peripheral blood and bone marrow by flow cytometry. Results revealed that the percentage of neutrophils in the CFA group in both peripheral blood and bone marrow were found to be highest among all five groups. Although the percentage of neutrophils in the 0.1LPS group and the 10LPS group increased significantly relative to the control group, it was not as significant as the CFA group. In the flow cytometry analysis of bone marrow cells, the percentage of CD11b(+)Ly6G(+) neutrophils in the 0.1LPS group and 10LPS group lower than that in the EOS group (Fig. 3A and 3B). As the first line of defense, neutrophils partially mediate host responses by establishing NETs [20]. PMA is a very powerful, but non-physiological stimulus for NET formation [18]. We next purified mouse bone marrow neutrophils and stimulated with PMA (100 nM) in vitro to see whether NETs were released. Confocal microscopy revealed that a large number of NETosis neutrophils and NETs were detected in all three neutrophil models, with the highest amount of that produced in the CFA group. There was a small number of NETs observed in the EOS group (Fig. 3C-F). We also found that only a minority of neutrophils developed NETosis in the absence of stimulation, whether isolated from the bone marrow of the neutrophil models or other models. In addition, we detected the expression levels of MPO and CitH3 using western blot in lung tissue of mice, and the results were consistent with that of immunofluorescence analysis (Fig. 3G and H).\n\nFig. 3Abundant NETs occur in neutrophilic asthma models. (A) Neutrophil expression in CD45(+) leucocytes in mouse peripheral blood and bone marrow were determined by flow cytometry within 48 h after the last challenge. The representative images in each group are shown. (B) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of mouse peripheral blood and bone marrow by flow cytometry. (C) In vitro NET-formation assays with purified neutrophils of mouse bone marrow neutrophils stimulated with PMA (100 nM) or RPMI 1640 for 4 h. Then the neutrophils were stained PI and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (D) Comparison of the percentage of NETosis cells in each group upon PMA stimulation and control (Ctrl) stimulation. (E) After stimulated with PMA, the neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green) and DAPI (nuclear staining, blue) and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (F) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (G, H) Western blot was used to detect the levels of MPO and CitH3 protein in lung tissue of five groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S1 and S4 presents the full-length blots. Data were shown as mean ± SEM; n = 4. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nAbundant NETs occur in neutrophilic asthma models. (A) Neutrophil expression in CD45(+) leucocytes in mouse peripheral blood and bone marrow were determined by flow cytometry within 48 h after the last challenge. The representative images in each group are shown. (B) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of mouse peripheral blood and bone marrow by flow cytometry. (C) In vitro NET-formation assays with purified neutrophils of mouse bone marrow neutrophils stimulated with PMA (100 nM) or RPMI 1640 for 4 h. Then the neutrophils were stained PI and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (D) Comparison of the percentage of NETosis cells in each group upon PMA stimulation and control (Ctrl) stimulation. (E) After stimulated with PMA, the neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green) and DAPI (nuclear staining, blue) and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (F) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (G, H) Western blot was used to detect the levels of MPO and CitH3 protein in lung tissue of five groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S1 and S4 presents the full-length blots. Data were shown as mean ± SEM; n = 4. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "We summarize the above results and conclude that the CFA group is a relatively more suitable model of neutrophilic asthma, although the AHR of this model is not significant. To overcome this, we attempted to establish a new murine model combining the sensitization methods of the 0.1LPS group and the CFA group (0.1LPS + CFA group) (Fig. 4A). Result shown that the 0.1LPS + CFA group exhibited higher AHR in response to methacholine compared with the 0.1LPS or CFA group, although the differences were not statistically significant (Fig. 4B). Similar to the CFA group, lung histopathological examination in 0.1LPS + CFA group showed obvious inflammatory cells infiltration and airway mucus secretion, some airways showed local airway mucus obstruction (Fig. 4C-F). These results seem to indicate that the 0.1LPS + CFA group serve as a suitable model of mouse to study the pathogenesis of the neutrophilic asthma.\n\nFig. 4The 0.1LPS and CFA combined with OVA-induced neutrophilic asthma model exhibits significant AHR and severe airway infiltration. (A) Schematic diagram of the experiment. (B) The AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used to assess impaired lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nThe 0.1LPS and CFA combined with OVA-induced neutrophilic asthma model exhibits significant AHR and severe airway infiltration. (A) Schematic diagram of the experiment. (B) The AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used to assess impaired lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "Immediately after, we performed BALF and lung tissue cells analysis. The total number of BALF in the 0.1LPS + CFA group was significantly higher than that in the control and 0.1LPS groups, and there was no significant difference in total number of BALF between the 0.1LPS + CFA and CFA groups (Fig. 5A). Similar results were observed between the groups in the percentage of neutrophils (Fig. 5A). The percentage of eosinophils in the BALF of the three neutrophil groups had no significant difference compared with each other, while higher than that in the control group (Fig. 5A). Likewise, we performed flow cytometry on BALF cells and single lung-cell suspensions to better understand our newly established neutrophil model. The percentage of neutrophils in CD45(+) leukocytes in the BALF and single lung-cell suspensions in both 0.1LPS + CFA and CFA groups was significantly increased, and the difference was statistically significant (Fig. 5B and C). The 0.1LPS group also shown more percentage of neutrophils in CD45(+) leukocytes compared with the control group, but no statistical significance was achieved (Fig. 5B and C). Next, we quantification the expression of inflammatory cytokines in BALF. As expected, the expression of IL-17A, IL-6 and IL-1β in the 0.1LPS + CFA group and other two neutrophil groups were strongly increased compared with the control group. However, the expression of IL-4 was not significantly different in all four groups (Fig. 5D).\n\nFig. 5Significant neutrophilic inflammation and secretion of proinflammatory factors were observed in CFA combined with LPS induced mouse model. (A) Quantification of total cell counts and differential cell counts (eosinophils and neutrophils) in BALF at 48 h after the last challenge. (B) The percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images of each group are shown. (C) Statistical analysis of the above data shown by flow cytometry. (D) Cytokine (IL-4, IL-6 and IL-1β) concentrations in BALF were quantified by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (D); n = 4 mice in (B) and (C). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nSignificant neutrophilic inflammation and secretion of proinflammatory factors were observed in CFA combined with LPS induced mouse model. (A) Quantification of total cell counts and differential cell counts (eosinophils and neutrophils) in BALF at 48 h after the last challenge. (B) The percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images of each group are shown. (C) Statistical analysis of the above data shown by flow cytometry. (D) Cytokine (IL-4, IL-6 and IL-1β) concentrations in BALF were quantified by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (D); n = 4 mice in (B) and (C). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "Likewise, to investigate whether neutrophils in the 0.1LPS + CFA group asthma mouse model also have a similar mechanism to the neutrophil groups described above, we also evaluated neutrophil infiltration in mouse peripheral blood and bone marrow by flow cytometry. Results demonstrate that the percentage of neutrophils in peripheral blood and bone marrow in the 0.1LPS + CFA group was significantly higher than that in the control group, which was comparable to the CFA group. The percentage of neutrophils in the 0.1LPS group was increased compared with the control group, but there was no statistical difference (Fig. 6A and B). Confocal microscopy demonstrated that NETs co-localized with MPO, citH3 and DAPI were detected in all three neutrophil models after PMA stimulation, while the 0.1LPS + CFA group and CFA group could observe the most significant NETs release (Fig. 6C-E). Western blot also showed that the protein expression levels of MPO and CitH3 in the lung tissue of mice in each group were consistent with the results of immunofluorescence (Fig. 6F and G).\n\nFig. 6Neutrophilic mouse model induced by CFA combined with LPS showed enhanced NETs formation capacity. (A) At 48 h after the last challenge, the percentage of neutrophil populations in CD45(+) leukocytes in mouse peripheral blood and bone marrow were determined by flow cytometry. The representative images in each group are shown. (B) Statistical analysis of the percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leukocyte gate of mouse peripheral blood and bone marrow by flow cytometry. (C) Neutrophils were purified from mouse bone marrow and stimulated with PMA (100 nM) or vehicle control for 4 h. Then, neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green), and DNA (DAPI, blue) and confocal by immunofluorescence microscope for analysis. Representative images of NETs immunofluorescence. Scale bar = 20 μm. (D) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (E) Representative z-axis images of the NETs immunofluorescence in 0.1LPS + CFA group. Scale bar = 10 μm. (F, G) Western blot analysis the protein expression level of MPO and CitH3 in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S2 and S5 presents the full-length blots. Data were shown as mean ± SEM; n = 4 or 6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nNeutrophilic mouse model induced by CFA combined with LPS showed enhanced NETs formation capacity. (A) At 48 h after the last challenge, the percentage of neutrophil populations in CD45(+) leukocytes in mouse peripheral blood and bone marrow were determined by flow cytometry. The representative images in each group are shown. (B) Statistical analysis of the percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leukocyte gate of mouse peripheral blood and bone marrow by flow cytometry. (C) Neutrophils were purified from mouse bone marrow and stimulated with PMA (100 nM) or vehicle control for 4 h. Then, neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green), and DNA (DAPI, blue) and confocal by immunofluorescence microscope for analysis. Representative images of NETs immunofluorescence. Scale bar = 20 μm. (D) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (E) Representative z-axis images of the NETs immunofluorescence in 0.1LPS + CFA group. Scale bar = 10 μm. (F, G) Western blot analysis the protein expression level of MPO and CitH3 in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S2 and S5 presents the full-length blots. Data were shown as mean ± SEM; n = 4 or 6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "To determine whether the presence of NETs contributes to the pathogenesis of neutrophilic asthma model, we treated mice with either DNase I or CI-amidine before each challenge (Fig. 7 A). Previous studies have shown that DNase I can break down NETs [21], while activation of PAD4 is a major driver of NETs formation, and CI-amidine is an irreversible PAD4 inhibitor [22, 23]. Here, we found that DNase I or CI-amidine treatment significantly reduced airway hyperresponsiveness (Fig. 7B) and alleviated airway inflammation in the 0.1LPS + CFA group (Fig. 7C and E), PAS staining showed that airway mucus obstruction disappeared (Fig. 7D and F). Analysis of inflammatory cells in BALF also indicated that both the total number of cells and the percentage of neutrophils were significantly reduced by DNase I or CI-amidine treatment (Fig. 7G). Western blot analysis of lung tissue showed that the expression of MPO and CitH3 were significantly reduced in the OVA/CFA/0.1 LPS + DNase I (DNase I) group or the OVA/CFA/0.1 LPS + CI-amidine (CI-amidine) group (Fig. 7 H and I).\n\nFig. 7DNase I or CI-amidine administration reduced airway hyperresponsiveness and alleviated airway inflammation (A) Experimental assay schematic for in vivo experiments. (B) AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used as an indicator of lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. (G) The total number of cells and the differential number of cells (eosinophils and neutrophils) were quantified 48 h after the last challenge in the BALF. (H, I) Western blot analysis to measure MPO and CitH3 protein expression level in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S3 and S6 presents the full-length blots. Data were shown as mean ± SEM; n = 4–6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nDNase I or CI-amidine administration reduced airway hyperresponsiveness and alleviated airway inflammation (A) Experimental assay schematic for in vivo experiments. (B) AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used as an indicator of lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. (G) The total number of cells and the differential number of cells (eosinophils and neutrophils) were quantified 48 h after the last challenge in the BALF. (H, I) Western blot analysis to measure MPO and CitH3 protein expression level in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S3 and S6 presents the full-length blots. Data were shown as mean ± SEM; n = 4–6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1Fig. S1. Full length blots of Fig. 3G.Fig. S2. Full length blots of Fig. 6 F.Fig. S3. Full length blots of Fig. 7 H.Fig. S4. Additional bands used for statistics in Fig. 3 H.Fig. S5. Additional bands used for statistics in Fig. 6G.Fig. S6. Additional bands used for statistics in Fig. 7I.\n\nSupplementary Material 1\nFig. S1. Full length blots of Fig. 3G.\nFig. S2. Full length blots of Fig. 6 F.\nFig. S3. Full length blots of Fig. 7 H.\nFig. S4. Additional bands used for statistics in Fig. 3 H.\nFig. S5. Additional bands used for statistics in Fig. 6G.\nFig. S6. Additional bands used for statistics in Fig. 7I." ]
[ null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Introduction", "Materials and methods", "Animals", "Mouse allergen sensitization and challenge", "Airway hyperresponsiveness measurement", "Bronchoalveolar lavage", "Cytokine analysis", "Lung histopathology staining", "Flow cytometry analysis", "Immunostaining and confocal microscopy", "Western blot", "Statistical analysis", "Results", "AHR and pathological changes induced by different allergens combined with OVA", "The ability of CFA to induce neutrophil inflammation is stronger than that of LPS", "The neutrophil mouse models developed obvious NETosis", "The neutrophilic asthma model induced by 0.1LPS and CFA combined with OVA exhibited significant AHR and severe airway infiltration", "CFA combined with LPS induced strong neutrophilic inflammation and secretion of proinflammatory factors", "The OVA/CFA/LPS-induced mice model developed obvious NETosis", "DNase I or CI-amidine administration protects mice of neutrophilic asthma model", "Discussion", "Conclusion", "Electronic supplementary material", "" ]
[ "Asthma is a heterogeneous chronic respiratory disease, characterized by airway hyperresponsiveness (AHR) and airway inflammation [1]. There are four main inflammatory phenotypes in asthma based on the proportion of granulocytes in induced sputum: neutrophilic asthma, eosinophilic asthma, paucigranulocytic asthma, and mixed granulocytic asthma [2]. Neutrophilic asthma is one of the main types of severe asthma [3], which exhibits worse lung function, more severe airway inflammation, and worse treatment response. Therefore, it is crucial to investigation the pathogenesis of neutrophilic asthma.\nExperimental animal models are essential to advance asthma pathophysiological research, and mice are the main subjects. There are classical animal models of eosinophilic asthma, such as ovalbumin (OVA)/ aluminum (Alum)-sensitized and OVA-challenged model of asthma, house dust mite (HDM) extract-induced model of asthma and so on [4–7]. However, to the best of our knowledge, there is no animal model of neutrophilic asthma to be universally accepted. Currently, there are some animal studies of neutrophilic asthma, for example, OVA/complete Freund’s adjuvant (CFA)-sensitized and OVA-challenged model of asthma, OVA/ lipopolysaccharide (LPS)-sensitized and OVA-challenged model of asthma [8–10]. While these models recapitulate some of the features of human neutrophilic asthma, each of these models had certain limitations.\nNeutrophils are the main effector cells of inflammation and tissue infection [11]. The formation of neutrophil extracellular traps (NETs) is a function of neutrophils [12]. Neutrophils increase when inflammation occurs, and can undergo a process of NETosis, releasing NETs outside the cells [13]. However, whether NETs are generated and function in neutrophilic asthma have not been completely elucidated.\nOur study demonstrated that the OVA/CFA/LPS-induced murine model is suitable for study as a model of neutrophilic asthma. This kind of model has massive neutrophilic inflammation and severe airway hyperresponsiveness. In addition, we also found that a large number of NETs were produced and correlated with the severity of airway inflammation. Therefore, it is reasonable to hypothesize that NETs play an important functional role in neutrophilic asthma.", "[SUBTITLE] Animals [SUBSECTION] BALB/c mice (female, 6-8 weeks) were purchased from Hangzhou Medical College (Hangzhou, China). We maintained all the mice in specific pathogen-free (SPF) conditions with a 12 light/12 dark cycle in the Experimental Animal Center, the First Affiliated Hospital, School of Medicine, Zhejiang University.\nBALB/c mice (female, 6-8 weeks) were purchased from Hangzhou Medical College (Hangzhou, China). We maintained all the mice in specific pathogen-free (SPF) conditions with a 12 light/12 dark cycle in the Experimental Animal Center, the First Affiliated Hospital, School of Medicine, Zhejiang University.\n[SUBTITLE] Mouse allergen sensitization and challenge [SUBSECTION] Mouse models were induced using modified protocols as previously reported [7–9]. Mice were randomly grouped and sensitized on day 1 and day 8. For the OVA/Alum group of asthma, 25 µg OVA (A5503, Sigma Aldrich, St. Louis, MO, USA) dissolved in 100 µl 0.9% saline was mixed 1:1 with Imject Alum (Pierce, Rockford, IL, USA) by intraperitoneal (IP) injection. For the OVA/CFA group of asthma, 25 µg OVA dissolved in 100 µl 0.9% saline was mixed 1:1 with CFA (F5881, Sigma Aldrich, St. Louis, MO, USA) by IP. injection. For the OVA/LPS group of asthma, mice were lightly anesthetized with isoflurane, and intratracheally injected using a combination of 25 µg OVA with 0.1 or 10 µg LPS (L2630, Sigma Aldrich, St. Louis, MO, USA) in a total volume of 40 µl, with 0.9% saline as the diluent. For the OVA/CFA/0.1 LPS group, the model of sensitization is a combination of the OVA/CFA group and OVA/LPS group. After sensitization, the above-mentioned groups were challenged with aerosolized 1% OVA for 30 min on days 15-17. For the OVA/CFA/0.1 LPS + DNase I group or the OVA/CFA/0.1 LPS + CI-amidine group, on the basis of the OVA/CFA/0.1 LPS group, intravenous injection of DNase I (5 mg per kg body weight) or intraperitoneal injection of Cl-amidine (10 mg per kg body weight) was performed 1 h before each challenge. Mice sensitized and challenged with 0.9% saline were used as controls.\nMouse models were induced using modified protocols as previously reported [7–9]. Mice were randomly grouped and sensitized on day 1 and day 8. For the OVA/Alum group of asthma, 25 µg OVA (A5503, Sigma Aldrich, St. Louis, MO, USA) dissolved in 100 µl 0.9% saline was mixed 1:1 with Imject Alum (Pierce, Rockford, IL, USA) by intraperitoneal (IP) injection. For the OVA/CFA group of asthma, 25 µg OVA dissolved in 100 µl 0.9% saline was mixed 1:1 with CFA (F5881, Sigma Aldrich, St. Louis, MO, USA) by IP. injection. For the OVA/LPS group of asthma, mice were lightly anesthetized with isoflurane, and intratracheally injected using a combination of 25 µg OVA with 0.1 or 10 µg LPS (L2630, Sigma Aldrich, St. Louis, MO, USA) in a total volume of 40 µl, with 0.9% saline as the diluent. For the OVA/CFA/0.1 LPS group, the model of sensitization is a combination of the OVA/CFA group and OVA/LPS group. After sensitization, the above-mentioned groups were challenged with aerosolized 1% OVA for 30 min on days 15-17. For the OVA/CFA/0.1 LPS + DNase I group or the OVA/CFA/0.1 LPS + CI-amidine group, on the basis of the OVA/CFA/0.1 LPS group, intravenous injection of DNase I (5 mg per kg body weight) or intraperitoneal injection of Cl-amidine (10 mg per kg body weight) was performed 1 h before each challenge. Mice sensitized and challenged with 0.9% saline were used as controls.\n[SUBTITLE] Airway hyperresponsiveness measurement [SUBSECTION] Airway responsiveness was assessed 24 h after the last OVA challenge as previously described [14]. Briefly, mice were placed in a plethysmograph chamber (EMKA Technologies, Paris, France) for at least 10 min to adaption. Baseline pulmonary parameters were first determined, and then mice were sequentially challenged with aerosolized PBS and methacholine (Mch, A2251, Sigma Aldrich, St. Louis, MO, USA) at increased concentrations (3.125, 6.25, 12.5, 25 and 50 mg/ml in PBS). Enhanced Pause (Penh), an indirect estimate of airway resistance, was used to measure airway resistance to methacholine. After each nebulization, record the Penh value for 3 min and take the average of three consecutive values.\nAirway responsiveness was assessed 24 h after the last OVA challenge as previously described [14]. Briefly, mice were placed in a plethysmograph chamber (EMKA Technologies, Paris, France) for at least 10 min to adaption. Baseline pulmonary parameters were first determined, and then mice were sequentially challenged with aerosolized PBS and methacholine (Mch, A2251, Sigma Aldrich, St. Louis, MO, USA) at increased concentrations (3.125, 6.25, 12.5, 25 and 50 mg/ml in PBS). Enhanced Pause (Penh), an indirect estimate of airway resistance, was used to measure airway resistance to methacholine. After each nebulization, record the Penh value for 3 min and take the average of three consecutive values.\n[SUBTITLE] Bronchoalveolar lavage [SUBSECTION] Mice were euthanized 48 h after the last OVA challenge, serum, bronchoalveolar lavage fluid (BALF), bone marrow and lung tissues were collected for further experiments. As described previously [14], BALF was collected on whole lungs by infusing 1 ml of PBS via a tracheal cannula. Then, the BALF was centrifuged, and the supernatants were stored at-80 °C until analysis, the cell pellet was resuspended in PBS for differential cell counting or flow cytometry analysis.\nMice were euthanized 48 h after the last OVA challenge, serum, bronchoalveolar lavage fluid (BALF), bone marrow and lung tissues were collected for further experiments. As described previously [14], BALF was collected on whole lungs by infusing 1 ml of PBS via a tracheal cannula. Then, the BALF was centrifuged, and the supernatants were stored at-80 °C until analysis, the cell pellet was resuspended in PBS for differential cell counting or flow cytometry analysis.\n[SUBTITLE] Cytokine analysis [SUBSECTION] Expression of interleukin (IL)-4 (431,104), IL-17 A (432,504), IL-6 (431,304), and IL-1β (432,604) in BALF were quantitated by enzyme-linked immunosorbent assay (ELISA) kit (Biolegend, San Diego, CA, USA) according to the manufacturers’ instructions.\nExpression of interleukin (IL)-4 (431,104), IL-17 A (432,504), IL-6 (431,304), and IL-1β (432,604) in BALF were quantitated by enzyme-linked immunosorbent assay (ELISA) kit (Biolegend, San Diego, CA, USA) according to the manufacturers’ instructions.\n[SUBTITLE] Lung histopathology staining [SUBSECTION] After the BALF was collected, the right main bronchus was ligated, and the left lung was perfused and fixed with 4% paraformaldehyde for 24 h, followed by paraffin embedded, sectioned, stained with hematoxylin and eosin (H&E) or paraffin acid-Schiff (PAS) staining. Sections were performed in a blinded fashion. The degree of inflammation on H&E-stained lung sections was scored as described previously [15]: 0, normal; 1, few inflammatory cells; 2, a ring of inflammatory cells 1 cell layer deep; 3, a ring of inflammatory cells 2 to 4 cells deep; 4, a ring of inflammatory cells greater than 4 cells deep. Mucus-containing goblet cells on PAS-stained lung sections was also scored previously described [16]: 0, no PAS-positive cells; 1, less than 25%; 2, 25 to 50% PAS-positive cells; 3, 50 to 80% PAS-positive cells; 4, greater than 80% PAS-positive cells.\nAfter the BALF was collected, the right main bronchus was ligated, and the left lung was perfused and fixed with 4% paraformaldehyde for 24 h, followed by paraffin embedded, sectioned, stained with hematoxylin and eosin (H&E) or paraffin acid-Schiff (PAS) staining. Sections were performed in a blinded fashion. The degree of inflammation on H&E-stained lung sections was scored as described previously [15]: 0, normal; 1, few inflammatory cells; 2, a ring of inflammatory cells 1 cell layer deep; 3, a ring of inflammatory cells 2 to 4 cells deep; 4, a ring of inflammatory cells greater than 4 cells deep. Mucus-containing goblet cells on PAS-stained lung sections was also scored previously described [16]: 0, no PAS-positive cells; 1, less than 25%; 2, 25 to 50% PAS-positive cells; 3, 50 to 80% PAS-positive cells; 4, greater than 80% PAS-positive cells.\n[SUBTITLE] Flow cytometry analysis [SUBSECTION] Flow cytometry of leukocytes in BALF, lung tissue, peripheral blood and bone marrow. The BALF were collected according to the above procedure. Lung single-cell suspensions were obtained as previously described [17]. The peripheral blood was collected from mice by removed their eyeball and placed in tubes containing EDTA. Red blood cells (RBCs) were lysed with RBC lysis buffer (Biolegend, 420,301, San Diego, CA, USA). The bone marrow cells were harvested from the femora and tibiae by flushing with PBS, then cells filtered through 40 μm strainers (352,340, BD Biosciences, San Diego, CA, USA) to obtain single cell suspension. After a washing step, all single-cell suspensions were incubated with Fixable Viability Stain 780 (FVS780, 565,388, BD Biosciences, San Diego, CA, USA, 1:1000) according to manufacturer’s instructions to gate out dead cells. Subsequently, samples were stained with antibodies against CD45-FITC (157,214, Biolegend, San Diego, CA, USA, 1:100), CD11b-PE (101,207, Biolegend, San Diego, CA, USA, 1:100) and Ly6G-PE/Cy7 (560,601, BD Biosciences, San Diego, CA, USA, 1:100) for 30 min on ice, then cells were analyzed on a Cytoflex LX flow cytometer (Beckman Coulter, CA, USA). All above antibodies were diluted in PBS. Analyses were performed using FlowJo software v10.6.2. Neutrophils were identified as CD45(+)CD11b(+)Ly6G(+).\nFlow cytometry of leukocytes in BALF, lung tissue, peripheral blood and bone marrow. The BALF were collected according to the above procedure. Lung single-cell suspensions were obtained as previously described [17]. The peripheral blood was collected from mice by removed their eyeball and placed in tubes containing EDTA. Red blood cells (RBCs) were lysed with RBC lysis buffer (Biolegend, 420,301, San Diego, CA, USA). The bone marrow cells were harvested from the femora and tibiae by flushing with PBS, then cells filtered through 40 μm strainers (352,340, BD Biosciences, San Diego, CA, USA) to obtain single cell suspension. After a washing step, all single-cell suspensions were incubated with Fixable Viability Stain 780 (FVS780, 565,388, BD Biosciences, San Diego, CA, USA, 1:1000) according to manufacturer’s instructions to gate out dead cells. Subsequently, samples were stained with antibodies against CD45-FITC (157,214, Biolegend, San Diego, CA, USA, 1:100), CD11b-PE (101,207, Biolegend, San Diego, CA, USA, 1:100) and Ly6G-PE/Cy7 (560,601, BD Biosciences, San Diego, CA, USA, 1:100) for 30 min on ice, then cells were analyzed on a Cytoflex LX flow cytometer (Beckman Coulter, CA, USA). All above antibodies were diluted in PBS. Analyses were performed using FlowJo software v10.6.2. Neutrophils were identified as CD45(+)CD11b(+)Ly6G(+).\n[SUBTITLE] Immunostaining and confocal microscopy [SUBSECTION] Bone marrow cells from mice were harvested as describe above. Subsequently, neutrophils from bone marrow were isolated by density gradients according to the mouse neutrophil isolation kit instructions (TBD2013NM, TBD Science, China). The neutrophils (2 × 105 cells per well in serum free RPMI 1640) were plated on poly-lysine-coated round coverslips and placed in 24-well culture plates. Then, the cells were stimulated with 100 nM Phorbol 12-myristate 13-acetate (PMA, P3681, Sigma Aldrich, St. Louis, MO, USA) or RPMI 1640 (Ctrl) for 4 h. Subsequently, cells were fixed in 4% paraformaldehyde (PFA) for 30 min and permeabilized by incubation in 0.5% Triton X-100 at room temperature (RT) for 1 min. After that some samples were stained with propidium iodide (PI, 5 µg/ml, P4170, Sigma Aldrich, St. Louis, MO, USA) for 30 min, and washed in PBS before confocal microscopic observation. To quantify NET formation, NETosis was defined as neutrophils with flattened nuclei, decondensed chromatin and expulsion of extracellular neutrophils under fluorescence microscopy. Two independent researchers analyzed 200 neutrophils in each sample [18]. The other samples were blocked by blocking solution (phosphate-buffered saline (PBS) containing 1% bovine serum albumin (BSA) and 0.1% Tween-20) for 1 h at RT, samples were stained with rabbit anti-citrullinated histone 3 (CitH3) (ab5103, Abcam, 1:500) and mouse anti- Myeloperoxidase (MPO) (AF3667, R&D, 1:50) overnight at 4 °C. The next day, samples were washed in PBST and incubated with secondary antibodies CoraLite 594-conjugated Goat Anti-Rabbit IgG (H + L) (Proteintech, SA00013-4, 1:400) and CoraLite 488-conjugated Affinipure Goat Anti-Mouse IgG (H + L) (Proteintech, SA00013-1, 1:400). All above antibodies were diluted in blocking solution. The nuclei were stained with 4,6-diamidino-2-phenylindole (DAPI, 422,801, Biolegend 1:10000). The NETs were identified by the colocalization of antibodies (MPO, CitH3 and DAPI) and quantified using Fiji software v2.1.0. To determine the colocalization, we used a ratio by normalizing the percentage CitH3 signal to the percentage MPO signal, analysis of the CitH3/MPO percentage values from four regions was performed [19].\nBone marrow cells from mice were harvested as describe above. Subsequently, neutrophils from bone marrow were isolated by density gradients according to the mouse neutrophil isolation kit instructions (TBD2013NM, TBD Science, China). The neutrophils (2 × 105 cells per well in serum free RPMI 1640) were plated on poly-lysine-coated round coverslips and placed in 24-well culture plates. Then, the cells were stimulated with 100 nM Phorbol 12-myristate 13-acetate (PMA, P3681, Sigma Aldrich, St. Louis, MO, USA) or RPMI 1640 (Ctrl) for 4 h. Subsequently, cells were fixed in 4% paraformaldehyde (PFA) for 30 min and permeabilized by incubation in 0.5% Triton X-100 at room temperature (RT) for 1 min. After that some samples were stained with propidium iodide (PI, 5 µg/ml, P4170, Sigma Aldrich, St. Louis, MO, USA) for 30 min, and washed in PBS before confocal microscopic observation. To quantify NET formation, NETosis was defined as neutrophils with flattened nuclei, decondensed chromatin and expulsion of extracellular neutrophils under fluorescence microscopy. Two independent researchers analyzed 200 neutrophils in each sample [18]. The other samples were blocked by blocking solution (phosphate-buffered saline (PBS) containing 1% bovine serum albumin (BSA) and 0.1% Tween-20) for 1 h at RT, samples were stained with rabbit anti-citrullinated histone 3 (CitH3) (ab5103, Abcam, 1:500) and mouse anti- Myeloperoxidase (MPO) (AF3667, R&D, 1:50) overnight at 4 °C. The next day, samples were washed in PBST and incubated with secondary antibodies CoraLite 594-conjugated Goat Anti-Rabbit IgG (H + L) (Proteintech, SA00013-4, 1:400) and CoraLite 488-conjugated Affinipure Goat Anti-Mouse IgG (H + L) (Proteintech, SA00013-1, 1:400). All above antibodies were diluted in blocking solution. The nuclei were stained with 4,6-diamidino-2-phenylindole (DAPI, 422,801, Biolegend 1:10000). The NETs were identified by the colocalization of antibodies (MPO, CitH3 and DAPI) and quantified using Fiji software v2.1.0. To determine the colocalization, we used a ratio by normalizing the percentage CitH3 signal to the percentage MPO signal, analysis of the CitH3/MPO percentage values from four regions was performed [19].\n[SUBTITLE] Western blot [SUBSECTION] Protein lysates of lung tissue were prepared. Then, the proteins were separated by 10-12% sodium dodecyl sulfatepolyacrylamide gel electrophoresis (SDS-PAGE), transferred onto polyvinylidene fluoride (PVDF) membranes. Membranes were blocked in 5% skim milk for 1 h RT, follow by incubated with primary antibodies overnight at 4 ℃. The following day, membranes were washed and incubated with HRP-conjugated secondary antibodies for 1 h and proteins were visualized using the ECL reagent. Rabbit anti-CitH3 (ab5103, 1:1000) was purchased from Abcam; Mouse anti-MPO (AF3667, 1:400) was purchased from R&D; beta (β)-Tubulin (66,240–1-Ig, 1:5000) was purchased from Proteintech Technology. All primary antibodies were diluted in primary antibody dilution buffer (Beyotime Biotechnology, China), and secondary antibodies were diluted in 5% skim milk.\nProtein lysates of lung tissue were prepared. Then, the proteins were separated by 10-12% sodium dodecyl sulfatepolyacrylamide gel electrophoresis (SDS-PAGE), transferred onto polyvinylidene fluoride (PVDF) membranes. Membranes were blocked in 5% skim milk for 1 h RT, follow by incubated with primary antibodies overnight at 4 ℃. The following day, membranes were washed and incubated with HRP-conjugated secondary antibodies for 1 h and proteins were visualized using the ECL reagent. Rabbit anti-CitH3 (ab5103, 1:1000) was purchased from Abcam; Mouse anti-MPO (AF3667, 1:400) was purchased from R&D; beta (β)-Tubulin (66,240–1-Ig, 1:5000) was purchased from Proteintech Technology. All primary antibodies were diluted in primary antibody dilution buffer (Beyotime Biotechnology, China), and secondary antibodies were diluted in 5% skim milk.\n[SUBTITLE] Statistical analysis [SUBSECTION] All statistics and graphs were performed using GraphPad Prism software v8.4.0 (GraphPad Software Inc., San Diego, CA, USA). In this study, a one-way ANOVA with Tukey posttests was used for multiple comparisons. Data are presented as means ± standard errors of measurement (SEM). Significant differences are shown as *P < 0.05, **P < 0.01, and ***P < 0.001.\nAll statistics and graphs were performed using GraphPad Prism software v8.4.0 (GraphPad Software Inc., San Diego, CA, USA). In this study, a one-way ANOVA with Tukey posttests was used for multiple comparisons. Data are presented as means ± standard errors of measurement (SEM). Significant differences are shown as *P < 0.05, **P < 0.01, and ***P < 0.001.", "BALB/c mice (female, 6-8 weeks) were purchased from Hangzhou Medical College (Hangzhou, China). We maintained all the mice in specific pathogen-free (SPF) conditions with a 12 light/12 dark cycle in the Experimental Animal Center, the First Affiliated Hospital, School of Medicine, Zhejiang University.", "Mouse models were induced using modified protocols as previously reported [7–9]. Mice were randomly grouped and sensitized on day 1 and day 8. For the OVA/Alum group of asthma, 25 µg OVA (A5503, Sigma Aldrich, St. Louis, MO, USA) dissolved in 100 µl 0.9% saline was mixed 1:1 with Imject Alum (Pierce, Rockford, IL, USA) by intraperitoneal (IP) injection. For the OVA/CFA group of asthma, 25 µg OVA dissolved in 100 µl 0.9% saline was mixed 1:1 with CFA (F5881, Sigma Aldrich, St. Louis, MO, USA) by IP. injection. For the OVA/LPS group of asthma, mice were lightly anesthetized with isoflurane, and intratracheally injected using a combination of 25 µg OVA with 0.1 or 10 µg LPS (L2630, Sigma Aldrich, St. Louis, MO, USA) in a total volume of 40 µl, with 0.9% saline as the diluent. For the OVA/CFA/0.1 LPS group, the model of sensitization is a combination of the OVA/CFA group and OVA/LPS group. After sensitization, the above-mentioned groups were challenged with aerosolized 1% OVA for 30 min on days 15-17. For the OVA/CFA/0.1 LPS + DNase I group or the OVA/CFA/0.1 LPS + CI-amidine group, on the basis of the OVA/CFA/0.1 LPS group, intravenous injection of DNase I (5 mg per kg body weight) or intraperitoneal injection of Cl-amidine (10 mg per kg body weight) was performed 1 h before each challenge. Mice sensitized and challenged with 0.9% saline were used as controls.", "Airway responsiveness was assessed 24 h after the last OVA challenge as previously described [14]. Briefly, mice were placed in a plethysmograph chamber (EMKA Technologies, Paris, France) for at least 10 min to adaption. Baseline pulmonary parameters were first determined, and then mice were sequentially challenged with aerosolized PBS and methacholine (Mch, A2251, Sigma Aldrich, St. Louis, MO, USA) at increased concentrations (3.125, 6.25, 12.5, 25 and 50 mg/ml in PBS). Enhanced Pause (Penh), an indirect estimate of airway resistance, was used to measure airway resistance to methacholine. After each nebulization, record the Penh value for 3 min and take the average of three consecutive values.", "Mice were euthanized 48 h after the last OVA challenge, serum, bronchoalveolar lavage fluid (BALF), bone marrow and lung tissues were collected for further experiments. As described previously [14], BALF was collected on whole lungs by infusing 1 ml of PBS via a tracheal cannula. Then, the BALF was centrifuged, and the supernatants were stored at-80 °C until analysis, the cell pellet was resuspended in PBS for differential cell counting or flow cytometry analysis.", "Expression of interleukin (IL)-4 (431,104), IL-17 A (432,504), IL-6 (431,304), and IL-1β (432,604) in BALF were quantitated by enzyme-linked immunosorbent assay (ELISA) kit (Biolegend, San Diego, CA, USA) according to the manufacturers’ instructions.", "After the BALF was collected, the right main bronchus was ligated, and the left lung was perfused and fixed with 4% paraformaldehyde for 24 h, followed by paraffin embedded, sectioned, stained with hematoxylin and eosin (H&E) or paraffin acid-Schiff (PAS) staining. Sections were performed in a blinded fashion. The degree of inflammation on H&E-stained lung sections was scored as described previously [15]: 0, normal; 1, few inflammatory cells; 2, a ring of inflammatory cells 1 cell layer deep; 3, a ring of inflammatory cells 2 to 4 cells deep; 4, a ring of inflammatory cells greater than 4 cells deep. Mucus-containing goblet cells on PAS-stained lung sections was also scored previously described [16]: 0, no PAS-positive cells; 1, less than 25%; 2, 25 to 50% PAS-positive cells; 3, 50 to 80% PAS-positive cells; 4, greater than 80% PAS-positive cells.", "Flow cytometry of leukocytes in BALF, lung tissue, peripheral blood and bone marrow. The BALF were collected according to the above procedure. Lung single-cell suspensions were obtained as previously described [17]. The peripheral blood was collected from mice by removed their eyeball and placed in tubes containing EDTA. Red blood cells (RBCs) were lysed with RBC lysis buffer (Biolegend, 420,301, San Diego, CA, USA). The bone marrow cells were harvested from the femora and tibiae by flushing with PBS, then cells filtered through 40 μm strainers (352,340, BD Biosciences, San Diego, CA, USA) to obtain single cell suspension. After a washing step, all single-cell suspensions were incubated with Fixable Viability Stain 780 (FVS780, 565,388, BD Biosciences, San Diego, CA, USA, 1:1000) according to manufacturer’s instructions to gate out dead cells. Subsequently, samples were stained with antibodies against CD45-FITC (157,214, Biolegend, San Diego, CA, USA, 1:100), CD11b-PE (101,207, Biolegend, San Diego, CA, USA, 1:100) and Ly6G-PE/Cy7 (560,601, BD Biosciences, San Diego, CA, USA, 1:100) for 30 min on ice, then cells were analyzed on a Cytoflex LX flow cytometer (Beckman Coulter, CA, USA). All above antibodies were diluted in PBS. Analyses were performed using FlowJo software v10.6.2. Neutrophils were identified as CD45(+)CD11b(+)Ly6G(+).", "Bone marrow cells from mice were harvested as describe above. Subsequently, neutrophils from bone marrow were isolated by density gradients according to the mouse neutrophil isolation kit instructions (TBD2013NM, TBD Science, China). The neutrophils (2 × 105 cells per well in serum free RPMI 1640) were plated on poly-lysine-coated round coverslips and placed in 24-well culture plates. Then, the cells were stimulated with 100 nM Phorbol 12-myristate 13-acetate (PMA, P3681, Sigma Aldrich, St. Louis, MO, USA) or RPMI 1640 (Ctrl) for 4 h. Subsequently, cells were fixed in 4% paraformaldehyde (PFA) for 30 min and permeabilized by incubation in 0.5% Triton X-100 at room temperature (RT) for 1 min. After that some samples were stained with propidium iodide (PI, 5 µg/ml, P4170, Sigma Aldrich, St. Louis, MO, USA) for 30 min, and washed in PBS before confocal microscopic observation. To quantify NET formation, NETosis was defined as neutrophils with flattened nuclei, decondensed chromatin and expulsion of extracellular neutrophils under fluorescence microscopy. Two independent researchers analyzed 200 neutrophils in each sample [18]. The other samples were blocked by blocking solution (phosphate-buffered saline (PBS) containing 1% bovine serum albumin (BSA) and 0.1% Tween-20) for 1 h at RT, samples were stained with rabbit anti-citrullinated histone 3 (CitH3) (ab5103, Abcam, 1:500) and mouse anti- Myeloperoxidase (MPO) (AF3667, R&D, 1:50) overnight at 4 °C. The next day, samples were washed in PBST and incubated with secondary antibodies CoraLite 594-conjugated Goat Anti-Rabbit IgG (H + L) (Proteintech, SA00013-4, 1:400) and CoraLite 488-conjugated Affinipure Goat Anti-Mouse IgG (H + L) (Proteintech, SA00013-1, 1:400). All above antibodies were diluted in blocking solution. The nuclei were stained with 4,6-diamidino-2-phenylindole (DAPI, 422,801, Biolegend 1:10000). The NETs were identified by the colocalization of antibodies (MPO, CitH3 and DAPI) and quantified using Fiji software v2.1.0. To determine the colocalization, we used a ratio by normalizing the percentage CitH3 signal to the percentage MPO signal, analysis of the CitH3/MPO percentage values from four regions was performed [19].", "Protein lysates of lung tissue were prepared. Then, the proteins were separated by 10-12% sodium dodecyl sulfatepolyacrylamide gel electrophoresis (SDS-PAGE), transferred onto polyvinylidene fluoride (PVDF) membranes. Membranes were blocked in 5% skim milk for 1 h RT, follow by incubated with primary antibodies overnight at 4 ℃. The following day, membranes were washed and incubated with HRP-conjugated secondary antibodies for 1 h and proteins were visualized using the ECL reagent. Rabbit anti-CitH3 (ab5103, 1:1000) was purchased from Abcam; Mouse anti-MPO (AF3667, 1:400) was purchased from R&D; beta (β)-Tubulin (66,240–1-Ig, 1:5000) was purchased from Proteintech Technology. All primary antibodies were diluted in primary antibody dilution buffer (Beyotime Biotechnology, China), and secondary antibodies were diluted in 5% skim milk.", "All statistics and graphs were performed using GraphPad Prism software v8.4.0 (GraphPad Software Inc., San Diego, CA, USA). In this study, a one-way ANOVA with Tukey posttests was used for multiple comparisons. Data are presented as means ± standard errors of measurement (SEM). Significant differences are shown as *P < 0.05, **P < 0.01, and ***P < 0.001.", "[SUBTITLE] AHR and pathological changes induced by different allergens combined with OVA [SUBSECTION] To establish a suitable model to recapitulate as many clinical features as possible of human severe neutrophilic asthma, we constructed three different type of neutrophilic asthma model in BALB/c mice using CFA/OVA or LPS/OVA. In parallel, we established an eosinophilic asthma model as a control using Alum/OVA (Fig. 1A). Whole-body plethysmography was used to assess the AHR. The 10 µg LPS/OVA (10LPS) group exhibited a drastic increase in Penh upon exposure to increasing concentrations of aerosolized methacholine, as compared to the saline-treated (control) control group and CFA/OVA (CFA) group (Fig. 1B). The 0.1 µg LPS/OVA (0.1LPS) group and Alum/OVA (EOS) group also showed significantly increase in Penh upon exposure to aerosolized methacholine compared with the control group. Meanwhile, from that of normal controls, the CFA group showed increase in Penh but did not reach statistical significance. However, the severity of lung histopathology in the CFA group is not similar to its AHR. The CFA group displayed more severe bronchiolar and perivascular inflammation infiltration as assessed by H&E staining than the other four groups (Fig. 1C and 1E). The inflammatory cell infiltration in the 0.1LPS group and 10LPS group was also more than that in the control group, but lower than that in the CFA group. We assessed mucin secretion in the airway of mice by using PAS staining and also observed similar results to H&E staining (Fig. 1D and F).\n\nFig. 1Different allergens combined with OVA-induced airway hyperresponsiveness (AHR) and pathological changes. (A) Schematic representation of experimental procedure. (B) The AHR was determined by calculating enhanced pause (Penh) values 24 h after the last challenge. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nDifferent allergens combined with OVA-induced airway hyperresponsiveness (AHR) and pathological changes. (A) Schematic representation of experimental procedure. (B) The AHR was determined by calculating enhanced pause (Penh) values 24 h after the last challenge. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\nTo establish a suitable model to recapitulate as many clinical features as possible of human severe neutrophilic asthma, we constructed three different type of neutrophilic asthma model in BALB/c mice using CFA/OVA or LPS/OVA. In parallel, we established an eosinophilic asthma model as a control using Alum/OVA (Fig. 1A). Whole-body plethysmography was used to assess the AHR. The 10 µg LPS/OVA (10LPS) group exhibited a drastic increase in Penh upon exposure to increasing concentrations of aerosolized methacholine, as compared to the saline-treated (control) control group and CFA/OVA (CFA) group (Fig. 1B). The 0.1 µg LPS/OVA (0.1LPS) group and Alum/OVA (EOS) group also showed significantly increase in Penh upon exposure to aerosolized methacholine compared with the control group. Meanwhile, from that of normal controls, the CFA group showed increase in Penh but did not reach statistical significance. However, the severity of lung histopathology in the CFA group is not similar to its AHR. The CFA group displayed more severe bronchiolar and perivascular inflammation infiltration as assessed by H&E staining than the other four groups (Fig. 1C and 1E). The inflammatory cell infiltration in the 0.1LPS group and 10LPS group was also more than that in the control group, but lower than that in the CFA group. We assessed mucin secretion in the airway of mice by using PAS staining and also observed similar results to H&E staining (Fig. 1D and F).\n\nFig. 1Different allergens combined with OVA-induced airway hyperresponsiveness (AHR) and pathological changes. (A) Schematic representation of experimental procedure. (B) The AHR was determined by calculating enhanced pause (Penh) values 24 h after the last challenge. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nDifferent allergens combined with OVA-induced airway hyperresponsiveness (AHR) and pathological changes. (A) Schematic representation of experimental procedure. (B) The AHR was determined by calculating enhanced pause (Penh) values 24 h after the last challenge. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n[SUBTITLE] The ability of CFA to induce neutrophil inflammation is stronger than that of LPS [SUBSECTION] To further validate the inflammatory phenotype of the model, we also performed total and differential cell counts of BALF. Results demonstrated that the total number of cells and the percentage of neutrophils were significantly increased in the three groups of neutrophil models, particularly the CFA group (Fig. 2A). The percentage of eosinophils in the BALF of the three groups of neutrophil models had no significant difference compared with the control group, the EOS group of that were markedly increased (Fig. 2A). Moreover, for more precise evaluate the inflammatory phenotype of each group, we performed flow cytometry on cells of the BALF and single lung-cell suspensions of mice. Neutrophils was gated as viable CD45(+)CD11b(+)Ly6G(+) cells (Fig. 2B). Although the percentage of neutrophils in CD45(+) leukocytes increased in the BALF and single lung-cell suspensions of the three groups of neutrophil models compared to the control group, only the CFA group had a statistically significant difference (Fig. 2C and 2D). Similarly, only the percentage of neutrophils in CD45(+) leukocytes of the CFA group was statistically significant compared to the eosinophil group (Fig. 2 C and D). We next assessed the impact of different allergens on inflammatory cytokines in BALF. As a marker of Th2 polarization, IL-4 was obviously increased in the EOS group, while there was no significant change in the three neutrophil groups. Unlike that, the three neutrophil groups, especially the CFA group, had significantly higher IL-17 A, IL-6 and IL-1β expression than the control or EOS group (Fig. 2E).\n\nFig. 2Neutrophil inflammatory infiltration was more pronounced in the CFA-induced neutrophil mouse models. (A) The number of total cells, eosinophils and neutrophils in BALF were quantified at 48 h after the last challenge. (B) Gating strategy for neutrophils is define as cells with the following characteristics: CD45(+)CD11b(+)Ly6G(+). (C) Neutrophil expression in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images in each group are shown. (D) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of BALF and lung tissue by flow cytometry. (E) The cytokines interleukin (IL)-4, IL-17A, IL-6 and IL-1β levels in BALF were measured by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (E); n = 4 in (C) and (D). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nNeutrophil inflammatory infiltration was more pronounced in the CFA-induced neutrophil mouse models. (A) The number of total cells, eosinophils and neutrophils in BALF were quantified at 48 h after the last challenge. (B) Gating strategy for neutrophils is define as cells with the following characteristics: CD45(+)CD11b(+)Ly6G(+). (C) Neutrophil expression in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images in each group are shown. (D) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of BALF and lung tissue by flow cytometry. (E) The cytokines interleukin (IL)-4, IL-17A, IL-6 and IL-1β levels in BALF were measured by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (E); n = 4 in (C) and (D). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\nTo further validate the inflammatory phenotype of the model, we also performed total and differential cell counts of BALF. Results demonstrated that the total number of cells and the percentage of neutrophils were significantly increased in the three groups of neutrophil models, particularly the CFA group (Fig. 2A). The percentage of eosinophils in the BALF of the three groups of neutrophil models had no significant difference compared with the control group, the EOS group of that were markedly increased (Fig. 2A). Moreover, for more precise evaluate the inflammatory phenotype of each group, we performed flow cytometry on cells of the BALF and single lung-cell suspensions of mice. Neutrophils was gated as viable CD45(+)CD11b(+)Ly6G(+) cells (Fig. 2B). Although the percentage of neutrophils in CD45(+) leukocytes increased in the BALF and single lung-cell suspensions of the three groups of neutrophil models compared to the control group, only the CFA group had a statistically significant difference (Fig. 2C and 2D). Similarly, only the percentage of neutrophils in CD45(+) leukocytes of the CFA group was statistically significant compared to the eosinophil group (Fig. 2 C and D). We next assessed the impact of different allergens on inflammatory cytokines in BALF. As a marker of Th2 polarization, IL-4 was obviously increased in the EOS group, while there was no significant change in the three neutrophil groups. Unlike that, the three neutrophil groups, especially the CFA group, had significantly higher IL-17 A, IL-6 and IL-1β expression than the control or EOS group (Fig. 2E).\n\nFig. 2Neutrophil inflammatory infiltration was more pronounced in the CFA-induced neutrophil mouse models. (A) The number of total cells, eosinophils and neutrophils in BALF were quantified at 48 h after the last challenge. (B) Gating strategy for neutrophils is define as cells with the following characteristics: CD45(+)CD11b(+)Ly6G(+). (C) Neutrophil expression in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images in each group are shown. (D) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of BALF and lung tissue by flow cytometry. (E) The cytokines interleukin (IL)-4, IL-17A, IL-6 and IL-1β levels in BALF were measured by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (E); n = 4 in (C) and (D). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nNeutrophil inflammatory infiltration was more pronounced in the CFA-induced neutrophil mouse models. (A) The number of total cells, eosinophils and neutrophils in BALF were quantified at 48 h after the last challenge. (B) Gating strategy for neutrophils is define as cells with the following characteristics: CD45(+)CD11b(+)Ly6G(+). (C) Neutrophil expression in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images in each group are shown. (D) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of BALF and lung tissue by flow cytometry. (E) The cytokines interleukin (IL)-4, IL-17A, IL-6 and IL-1β levels in BALF were measured by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (E); n = 4 in (C) and (D). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n[SUBTITLE] The neutrophil mouse models developed obvious NETosis [SUBSECTION] To further explore the mechanism of neutrophils in mouse model of neutrophilic asthma, we first assessed neutrophil infiltration in mouse peripheral blood and bone marrow by flow cytometry. Results revealed that the percentage of neutrophils in the CFA group in both peripheral blood and bone marrow were found to be highest among all five groups. Although the percentage of neutrophils in the 0.1LPS group and the 10LPS group increased significantly relative to the control group, it was not as significant as the CFA group. In the flow cytometry analysis of bone marrow cells, the percentage of CD11b(+)Ly6G(+) neutrophils in the 0.1LPS group and 10LPS group lower than that in the EOS group (Fig. 3A and 3B). As the first line of defense, neutrophils partially mediate host responses by establishing NETs [20]. PMA is a very powerful, but non-physiological stimulus for NET formation [18]. We next purified mouse bone marrow neutrophils and stimulated with PMA (100 nM) in vitro to see whether NETs were released. Confocal microscopy revealed that a large number of NETosis neutrophils and NETs were detected in all three neutrophil models, with the highest amount of that produced in the CFA group. There was a small number of NETs observed in the EOS group (Fig. 3C-F). We also found that only a minority of neutrophils developed NETosis in the absence of stimulation, whether isolated from the bone marrow of the neutrophil models or other models. In addition, we detected the expression levels of MPO and CitH3 using western blot in lung tissue of mice, and the results were consistent with that of immunofluorescence analysis (Fig. 3G and H).\n\nFig. 3Abundant NETs occur in neutrophilic asthma models. (A) Neutrophil expression in CD45(+) leucocytes in mouse peripheral blood and bone marrow were determined by flow cytometry within 48 h after the last challenge. The representative images in each group are shown. (B) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of mouse peripheral blood and bone marrow by flow cytometry. (C) In vitro NET-formation assays with purified neutrophils of mouse bone marrow neutrophils stimulated with PMA (100 nM) or RPMI 1640 for 4 h. Then the neutrophils were stained PI and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (D) Comparison of the percentage of NETosis cells in each group upon PMA stimulation and control (Ctrl) stimulation. (E) After stimulated with PMA, the neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green) and DAPI (nuclear staining, blue) and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (F) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (G, H) Western blot was used to detect the levels of MPO and CitH3 protein in lung tissue of five groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S1 and S4 presents the full-length blots. Data were shown as mean ± SEM; n = 4. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nAbundant NETs occur in neutrophilic asthma models. (A) Neutrophil expression in CD45(+) leucocytes in mouse peripheral blood and bone marrow were determined by flow cytometry within 48 h after the last challenge. The representative images in each group are shown. (B) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of mouse peripheral blood and bone marrow by flow cytometry. (C) In vitro NET-formation assays with purified neutrophils of mouse bone marrow neutrophils stimulated with PMA (100 nM) or RPMI 1640 for 4 h. Then the neutrophils were stained PI and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (D) Comparison of the percentage of NETosis cells in each group upon PMA stimulation and control (Ctrl) stimulation. (E) After stimulated with PMA, the neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green) and DAPI (nuclear staining, blue) and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (F) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (G, H) Western blot was used to detect the levels of MPO and CitH3 protein in lung tissue of five groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S1 and S4 presents the full-length blots. Data were shown as mean ± SEM; n = 4. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\nTo further explore the mechanism of neutrophils in mouse model of neutrophilic asthma, we first assessed neutrophil infiltration in mouse peripheral blood and bone marrow by flow cytometry. Results revealed that the percentage of neutrophils in the CFA group in both peripheral blood and bone marrow were found to be highest among all five groups. Although the percentage of neutrophils in the 0.1LPS group and the 10LPS group increased significantly relative to the control group, it was not as significant as the CFA group. In the flow cytometry analysis of bone marrow cells, the percentage of CD11b(+)Ly6G(+) neutrophils in the 0.1LPS group and 10LPS group lower than that in the EOS group (Fig. 3A and 3B). As the first line of defense, neutrophils partially mediate host responses by establishing NETs [20]. PMA is a very powerful, but non-physiological stimulus for NET formation [18]. We next purified mouse bone marrow neutrophils and stimulated with PMA (100 nM) in vitro to see whether NETs were released. Confocal microscopy revealed that a large number of NETosis neutrophils and NETs were detected in all three neutrophil models, with the highest amount of that produced in the CFA group. There was a small number of NETs observed in the EOS group (Fig. 3C-F). We also found that only a minority of neutrophils developed NETosis in the absence of stimulation, whether isolated from the bone marrow of the neutrophil models or other models. In addition, we detected the expression levels of MPO and CitH3 using western blot in lung tissue of mice, and the results were consistent with that of immunofluorescence analysis (Fig. 3G and H).\n\nFig. 3Abundant NETs occur in neutrophilic asthma models. (A) Neutrophil expression in CD45(+) leucocytes in mouse peripheral blood and bone marrow were determined by flow cytometry within 48 h after the last challenge. The representative images in each group are shown. (B) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of mouse peripheral blood and bone marrow by flow cytometry. (C) In vitro NET-formation assays with purified neutrophils of mouse bone marrow neutrophils stimulated with PMA (100 nM) or RPMI 1640 for 4 h. Then the neutrophils were stained PI and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (D) Comparison of the percentage of NETosis cells in each group upon PMA stimulation and control (Ctrl) stimulation. (E) After stimulated with PMA, the neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green) and DAPI (nuclear staining, blue) and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (F) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (G, H) Western blot was used to detect the levels of MPO and CitH3 protein in lung tissue of five groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S1 and S4 presents the full-length blots. Data were shown as mean ± SEM; n = 4. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nAbundant NETs occur in neutrophilic asthma models. (A) Neutrophil expression in CD45(+) leucocytes in mouse peripheral blood and bone marrow were determined by flow cytometry within 48 h after the last challenge. The representative images in each group are shown. (B) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of mouse peripheral blood and bone marrow by flow cytometry. (C) In vitro NET-formation assays with purified neutrophils of mouse bone marrow neutrophils stimulated with PMA (100 nM) or RPMI 1640 for 4 h. Then the neutrophils were stained PI and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (D) Comparison of the percentage of NETosis cells in each group upon PMA stimulation and control (Ctrl) stimulation. (E) After stimulated with PMA, the neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green) and DAPI (nuclear staining, blue) and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (F) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (G, H) Western blot was used to detect the levels of MPO and CitH3 protein in lung tissue of five groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S1 and S4 presents the full-length blots. Data were shown as mean ± SEM; n = 4. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n[SUBTITLE] The neutrophilic asthma model induced by 0.1LPS and CFA combined with OVA exhibited significant AHR and severe airway infiltration [SUBSECTION] We summarize the above results and conclude that the CFA group is a relatively more suitable model of neutrophilic asthma, although the AHR of this model is not significant. To overcome this, we attempted to establish a new murine model combining the sensitization methods of the 0.1LPS group and the CFA group (0.1LPS + CFA group) (Fig. 4A). Result shown that the 0.1LPS + CFA group exhibited higher AHR in response to methacholine compared with the 0.1LPS or CFA group, although the differences were not statistically significant (Fig. 4B). Similar to the CFA group, lung histopathological examination in 0.1LPS + CFA group showed obvious inflammatory cells infiltration and airway mucus secretion, some airways showed local airway mucus obstruction (Fig. 4C-F). These results seem to indicate that the 0.1LPS + CFA group serve as a suitable model of mouse to study the pathogenesis of the neutrophilic asthma.\n\nFig. 4The 0.1LPS and CFA combined with OVA-induced neutrophilic asthma model exhibits significant AHR and severe airway infiltration. (A) Schematic diagram of the experiment. (B) The AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used to assess impaired lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nThe 0.1LPS and CFA combined with OVA-induced neutrophilic asthma model exhibits significant AHR and severe airway infiltration. (A) Schematic diagram of the experiment. (B) The AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used to assess impaired lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\nWe summarize the above results and conclude that the CFA group is a relatively more suitable model of neutrophilic asthma, although the AHR of this model is not significant. To overcome this, we attempted to establish a new murine model combining the sensitization methods of the 0.1LPS group and the CFA group (0.1LPS + CFA group) (Fig. 4A). Result shown that the 0.1LPS + CFA group exhibited higher AHR in response to methacholine compared with the 0.1LPS or CFA group, although the differences were not statistically significant (Fig. 4B). Similar to the CFA group, lung histopathological examination in 0.1LPS + CFA group showed obvious inflammatory cells infiltration and airway mucus secretion, some airways showed local airway mucus obstruction (Fig. 4C-F). These results seem to indicate that the 0.1LPS + CFA group serve as a suitable model of mouse to study the pathogenesis of the neutrophilic asthma.\n\nFig. 4The 0.1LPS and CFA combined with OVA-induced neutrophilic asthma model exhibits significant AHR and severe airway infiltration. (A) Schematic diagram of the experiment. (B) The AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used to assess impaired lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nThe 0.1LPS and CFA combined with OVA-induced neutrophilic asthma model exhibits significant AHR and severe airway infiltration. (A) Schematic diagram of the experiment. (B) The AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used to assess impaired lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n[SUBTITLE] CFA combined with LPS induced strong neutrophilic inflammation and secretion of proinflammatory factors [SUBSECTION] Immediately after, we performed BALF and lung tissue cells analysis. The total number of BALF in the 0.1LPS + CFA group was significantly higher than that in the control and 0.1LPS groups, and there was no significant difference in total number of BALF between the 0.1LPS + CFA and CFA groups (Fig. 5A). Similar results were observed between the groups in the percentage of neutrophils (Fig. 5A). The percentage of eosinophils in the BALF of the three neutrophil groups had no significant difference compared with each other, while higher than that in the control group (Fig. 5A). Likewise, we performed flow cytometry on BALF cells and single lung-cell suspensions to better understand our newly established neutrophil model. The percentage of neutrophils in CD45(+) leukocytes in the BALF and single lung-cell suspensions in both 0.1LPS + CFA and CFA groups was significantly increased, and the difference was statistically significant (Fig. 5B and C). The 0.1LPS group also shown more percentage of neutrophils in CD45(+) leukocytes compared with the control group, but no statistical significance was achieved (Fig. 5B and C). Next, we quantification the expression of inflammatory cytokines in BALF. As expected, the expression of IL-17A, IL-6 and IL-1β in the 0.1LPS + CFA group and other two neutrophil groups were strongly increased compared with the control group. However, the expression of IL-4 was not significantly different in all four groups (Fig. 5D).\n\nFig. 5Significant neutrophilic inflammation and secretion of proinflammatory factors were observed in CFA combined with LPS induced mouse model. (A) Quantification of total cell counts and differential cell counts (eosinophils and neutrophils) in BALF at 48 h after the last challenge. (B) The percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images of each group are shown. (C) Statistical analysis of the above data shown by flow cytometry. (D) Cytokine (IL-4, IL-6 and IL-1β) concentrations in BALF were quantified by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (D); n = 4 mice in (B) and (C). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nSignificant neutrophilic inflammation and secretion of proinflammatory factors were observed in CFA combined with LPS induced mouse model. (A) Quantification of total cell counts and differential cell counts (eosinophils and neutrophils) in BALF at 48 h after the last challenge. (B) The percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images of each group are shown. (C) Statistical analysis of the above data shown by flow cytometry. (D) Cytokine (IL-4, IL-6 and IL-1β) concentrations in BALF were quantified by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (D); n = 4 mice in (B) and (C). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\nImmediately after, we performed BALF and lung tissue cells analysis. The total number of BALF in the 0.1LPS + CFA group was significantly higher than that in the control and 0.1LPS groups, and there was no significant difference in total number of BALF between the 0.1LPS + CFA and CFA groups (Fig. 5A). Similar results were observed between the groups in the percentage of neutrophils (Fig. 5A). The percentage of eosinophils in the BALF of the three neutrophil groups had no significant difference compared with each other, while higher than that in the control group (Fig. 5A). Likewise, we performed flow cytometry on BALF cells and single lung-cell suspensions to better understand our newly established neutrophil model. The percentage of neutrophils in CD45(+) leukocytes in the BALF and single lung-cell suspensions in both 0.1LPS + CFA and CFA groups was significantly increased, and the difference was statistically significant (Fig. 5B and C). The 0.1LPS group also shown more percentage of neutrophils in CD45(+) leukocytes compared with the control group, but no statistical significance was achieved (Fig. 5B and C). Next, we quantification the expression of inflammatory cytokines in BALF. As expected, the expression of IL-17A, IL-6 and IL-1β in the 0.1LPS + CFA group and other two neutrophil groups were strongly increased compared with the control group. However, the expression of IL-4 was not significantly different in all four groups (Fig. 5D).\n\nFig. 5Significant neutrophilic inflammation and secretion of proinflammatory factors were observed in CFA combined with LPS induced mouse model. (A) Quantification of total cell counts and differential cell counts (eosinophils and neutrophils) in BALF at 48 h after the last challenge. (B) The percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images of each group are shown. (C) Statistical analysis of the above data shown by flow cytometry. (D) Cytokine (IL-4, IL-6 and IL-1β) concentrations in BALF were quantified by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (D); n = 4 mice in (B) and (C). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nSignificant neutrophilic inflammation and secretion of proinflammatory factors were observed in CFA combined with LPS induced mouse model. (A) Quantification of total cell counts and differential cell counts (eosinophils and neutrophils) in BALF at 48 h after the last challenge. (B) The percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images of each group are shown. (C) Statistical analysis of the above data shown by flow cytometry. (D) Cytokine (IL-4, IL-6 and IL-1β) concentrations in BALF were quantified by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (D); n = 4 mice in (B) and (C). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n[SUBTITLE] The OVA/CFA/LPS-induced mice model developed obvious NETosis [SUBSECTION] Likewise, to investigate whether neutrophils in the 0.1LPS + CFA group asthma mouse model also have a similar mechanism to the neutrophil groups described above, we also evaluated neutrophil infiltration in mouse peripheral blood and bone marrow by flow cytometry. Results demonstrate that the percentage of neutrophils in peripheral blood and bone marrow in the 0.1LPS + CFA group was significantly higher than that in the control group, which was comparable to the CFA group. The percentage of neutrophils in the 0.1LPS group was increased compared with the control group, but there was no statistical difference (Fig. 6A and B). Confocal microscopy demonstrated that NETs co-localized with MPO, citH3 and DAPI were detected in all three neutrophil models after PMA stimulation, while the 0.1LPS + CFA group and CFA group could observe the most significant NETs release (Fig. 6C-E). Western blot also showed that the protein expression levels of MPO and CitH3 in the lung tissue of mice in each group were consistent with the results of immunofluorescence (Fig. 6F and G).\n\nFig. 6Neutrophilic mouse model induced by CFA combined with LPS showed enhanced NETs formation capacity. (A) At 48 h after the last challenge, the percentage of neutrophil populations in CD45(+) leukocytes in mouse peripheral blood and bone marrow were determined by flow cytometry. The representative images in each group are shown. (B) Statistical analysis of the percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leukocyte gate of mouse peripheral blood and bone marrow by flow cytometry. (C) Neutrophils were purified from mouse bone marrow and stimulated with PMA (100 nM) or vehicle control for 4 h. Then, neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green), and DNA (DAPI, blue) and confocal by immunofluorescence microscope for analysis. Representative images of NETs immunofluorescence. Scale bar = 20 μm. (D) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (E) Representative z-axis images of the NETs immunofluorescence in 0.1LPS + CFA group. Scale bar = 10 μm. (F, G) Western blot analysis the protein expression level of MPO and CitH3 in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S2 and S5 presents the full-length blots. Data were shown as mean ± SEM; n = 4 or 6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nNeutrophilic mouse model induced by CFA combined with LPS showed enhanced NETs formation capacity. (A) At 48 h after the last challenge, the percentage of neutrophil populations in CD45(+) leukocytes in mouse peripheral blood and bone marrow were determined by flow cytometry. The representative images in each group are shown. (B) Statistical analysis of the percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leukocyte gate of mouse peripheral blood and bone marrow by flow cytometry. (C) Neutrophils were purified from mouse bone marrow and stimulated with PMA (100 nM) or vehicle control for 4 h. Then, neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green), and DNA (DAPI, blue) and confocal by immunofluorescence microscope for analysis. Representative images of NETs immunofluorescence. Scale bar = 20 μm. (D) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (E) Representative z-axis images of the NETs immunofluorescence in 0.1LPS + CFA group. Scale bar = 10 μm. (F, G) Western blot analysis the protein expression level of MPO and CitH3 in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S2 and S5 presents the full-length blots. Data were shown as mean ± SEM; n = 4 or 6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\nLikewise, to investigate whether neutrophils in the 0.1LPS + CFA group asthma mouse model also have a similar mechanism to the neutrophil groups described above, we also evaluated neutrophil infiltration in mouse peripheral blood and bone marrow by flow cytometry. Results demonstrate that the percentage of neutrophils in peripheral blood and bone marrow in the 0.1LPS + CFA group was significantly higher than that in the control group, which was comparable to the CFA group. The percentage of neutrophils in the 0.1LPS group was increased compared with the control group, but there was no statistical difference (Fig. 6A and B). Confocal microscopy demonstrated that NETs co-localized with MPO, citH3 and DAPI were detected in all three neutrophil models after PMA stimulation, while the 0.1LPS + CFA group and CFA group could observe the most significant NETs release (Fig. 6C-E). Western blot also showed that the protein expression levels of MPO and CitH3 in the lung tissue of mice in each group were consistent with the results of immunofluorescence (Fig. 6F and G).\n\nFig. 6Neutrophilic mouse model induced by CFA combined with LPS showed enhanced NETs formation capacity. (A) At 48 h after the last challenge, the percentage of neutrophil populations in CD45(+) leukocytes in mouse peripheral blood and bone marrow were determined by flow cytometry. The representative images in each group are shown. (B) Statistical analysis of the percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leukocyte gate of mouse peripheral blood and bone marrow by flow cytometry. (C) Neutrophils were purified from mouse bone marrow and stimulated with PMA (100 nM) or vehicle control for 4 h. Then, neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green), and DNA (DAPI, blue) and confocal by immunofluorescence microscope for analysis. Representative images of NETs immunofluorescence. Scale bar = 20 μm. (D) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (E) Representative z-axis images of the NETs immunofluorescence in 0.1LPS + CFA group. Scale bar = 10 μm. (F, G) Western blot analysis the protein expression level of MPO and CitH3 in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S2 and S5 presents the full-length blots. Data were shown as mean ± SEM; n = 4 or 6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nNeutrophilic mouse model induced by CFA combined with LPS showed enhanced NETs formation capacity. (A) At 48 h after the last challenge, the percentage of neutrophil populations in CD45(+) leukocytes in mouse peripheral blood and bone marrow were determined by flow cytometry. The representative images in each group are shown. (B) Statistical analysis of the percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leukocyte gate of mouse peripheral blood and bone marrow by flow cytometry. (C) Neutrophils were purified from mouse bone marrow and stimulated with PMA (100 nM) or vehicle control for 4 h. Then, neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green), and DNA (DAPI, blue) and confocal by immunofluorescence microscope for analysis. Representative images of NETs immunofluorescence. Scale bar = 20 μm. (D) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (E) Representative z-axis images of the NETs immunofluorescence in 0.1LPS + CFA group. Scale bar = 10 μm. (F, G) Western blot analysis the protein expression level of MPO and CitH3 in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S2 and S5 presents the full-length blots. Data were shown as mean ± SEM; n = 4 or 6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n[SUBTITLE] DNase I or CI-amidine administration protects mice of neutrophilic asthma model [SUBSECTION] To determine whether the presence of NETs contributes to the pathogenesis of neutrophilic asthma model, we treated mice with either DNase I or CI-amidine before each challenge (Fig. 7 A). Previous studies have shown that DNase I can break down NETs [21], while activation of PAD4 is a major driver of NETs formation, and CI-amidine is an irreversible PAD4 inhibitor [22, 23]. Here, we found that DNase I or CI-amidine treatment significantly reduced airway hyperresponsiveness (Fig. 7B) and alleviated airway inflammation in the 0.1LPS + CFA group (Fig. 7C and E), PAS staining showed that airway mucus obstruction disappeared (Fig. 7D and F). Analysis of inflammatory cells in BALF also indicated that both the total number of cells and the percentage of neutrophils were significantly reduced by DNase I or CI-amidine treatment (Fig. 7G). Western blot analysis of lung tissue showed that the expression of MPO and CitH3 were significantly reduced in the OVA/CFA/0.1 LPS + DNase I (DNase I) group or the OVA/CFA/0.1 LPS + CI-amidine (CI-amidine) group (Fig. 7 H and I).\n\nFig. 7DNase I or CI-amidine administration reduced airway hyperresponsiveness and alleviated airway inflammation (A) Experimental assay schematic for in vivo experiments. (B) AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used as an indicator of lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. (G) The total number of cells and the differential number of cells (eosinophils and neutrophils) were quantified 48 h after the last challenge in the BALF. (H, I) Western blot analysis to measure MPO and CitH3 protein expression level in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S3 and S6 presents the full-length blots. Data were shown as mean ± SEM; n = 4–6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nDNase I or CI-amidine administration reduced airway hyperresponsiveness and alleviated airway inflammation (A) Experimental assay schematic for in vivo experiments. (B) AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used as an indicator of lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. (G) The total number of cells and the differential number of cells (eosinophils and neutrophils) were quantified 48 h after the last challenge in the BALF. (H, I) Western blot analysis to measure MPO and CitH3 protein expression level in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S3 and S6 presents the full-length blots. Data were shown as mean ± SEM; n = 4–6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\nTo determine whether the presence of NETs contributes to the pathogenesis of neutrophilic asthma model, we treated mice with either DNase I or CI-amidine before each challenge (Fig. 7 A). Previous studies have shown that DNase I can break down NETs [21], while activation of PAD4 is a major driver of NETs formation, and CI-amidine is an irreversible PAD4 inhibitor [22, 23]. Here, we found that DNase I or CI-amidine treatment significantly reduced airway hyperresponsiveness (Fig. 7B) and alleviated airway inflammation in the 0.1LPS + CFA group (Fig. 7C and E), PAS staining showed that airway mucus obstruction disappeared (Fig. 7D and F). Analysis of inflammatory cells in BALF also indicated that both the total number of cells and the percentage of neutrophils were significantly reduced by DNase I or CI-amidine treatment (Fig. 7G). Western blot analysis of lung tissue showed that the expression of MPO and CitH3 were significantly reduced in the OVA/CFA/0.1 LPS + DNase I (DNase I) group or the OVA/CFA/0.1 LPS + CI-amidine (CI-amidine) group (Fig. 7 H and I).\n\nFig. 7DNase I or CI-amidine administration reduced airway hyperresponsiveness and alleviated airway inflammation (A) Experimental assay schematic for in vivo experiments. (B) AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used as an indicator of lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. (G) The total number of cells and the differential number of cells (eosinophils and neutrophils) were quantified 48 h after the last challenge in the BALF. (H, I) Western blot analysis to measure MPO and CitH3 protein expression level in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S3 and S6 presents the full-length blots. Data were shown as mean ± SEM; n = 4–6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nDNase I or CI-amidine administration reduced airway hyperresponsiveness and alleviated airway inflammation (A) Experimental assay schematic for in vivo experiments. (B) AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used as an indicator of lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. (G) The total number of cells and the differential number of cells (eosinophils and neutrophils) were quantified 48 h after the last challenge in the BALF. (H, I) Western blot analysis to measure MPO and CitH3 protein expression level in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S3 and S6 presents the full-length blots. Data were shown as mean ± SEM; n = 4–6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "To establish a suitable model to recapitulate as many clinical features as possible of human severe neutrophilic asthma, we constructed three different type of neutrophilic asthma model in BALB/c mice using CFA/OVA or LPS/OVA. In parallel, we established an eosinophilic asthma model as a control using Alum/OVA (Fig. 1A). Whole-body plethysmography was used to assess the AHR. The 10 µg LPS/OVA (10LPS) group exhibited a drastic increase in Penh upon exposure to increasing concentrations of aerosolized methacholine, as compared to the saline-treated (control) control group and CFA/OVA (CFA) group (Fig. 1B). The 0.1 µg LPS/OVA (0.1LPS) group and Alum/OVA (EOS) group also showed significantly increase in Penh upon exposure to aerosolized methacholine compared with the control group. Meanwhile, from that of normal controls, the CFA group showed increase in Penh but did not reach statistical significance. However, the severity of lung histopathology in the CFA group is not similar to its AHR. The CFA group displayed more severe bronchiolar and perivascular inflammation infiltration as assessed by H&E staining than the other four groups (Fig. 1C and 1E). The inflammatory cell infiltration in the 0.1LPS group and 10LPS group was also more than that in the control group, but lower than that in the CFA group. We assessed mucin secretion in the airway of mice by using PAS staining and also observed similar results to H&E staining (Fig. 1D and F).\n\nFig. 1Different allergens combined with OVA-induced airway hyperresponsiveness (AHR) and pathological changes. (A) Schematic representation of experimental procedure. (B) The AHR was determined by calculating enhanced pause (Penh) values 24 h after the last challenge. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nDifferent allergens combined with OVA-induced airway hyperresponsiveness (AHR) and pathological changes. (A) Schematic representation of experimental procedure. (B) The AHR was determined by calculating enhanced pause (Penh) values 24 h after the last challenge. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "To further validate the inflammatory phenotype of the model, we also performed total and differential cell counts of BALF. Results demonstrated that the total number of cells and the percentage of neutrophils were significantly increased in the three groups of neutrophil models, particularly the CFA group (Fig. 2A). The percentage of eosinophils in the BALF of the three groups of neutrophil models had no significant difference compared with the control group, the EOS group of that were markedly increased (Fig. 2A). Moreover, for more precise evaluate the inflammatory phenotype of each group, we performed flow cytometry on cells of the BALF and single lung-cell suspensions of mice. Neutrophils was gated as viable CD45(+)CD11b(+)Ly6G(+) cells (Fig. 2B). Although the percentage of neutrophils in CD45(+) leukocytes increased in the BALF and single lung-cell suspensions of the three groups of neutrophil models compared to the control group, only the CFA group had a statistically significant difference (Fig. 2C and 2D). Similarly, only the percentage of neutrophils in CD45(+) leukocytes of the CFA group was statistically significant compared to the eosinophil group (Fig. 2 C and D). We next assessed the impact of different allergens on inflammatory cytokines in BALF. As a marker of Th2 polarization, IL-4 was obviously increased in the EOS group, while there was no significant change in the three neutrophil groups. Unlike that, the three neutrophil groups, especially the CFA group, had significantly higher IL-17 A, IL-6 and IL-1β expression than the control or EOS group (Fig. 2E).\n\nFig. 2Neutrophil inflammatory infiltration was more pronounced in the CFA-induced neutrophil mouse models. (A) The number of total cells, eosinophils and neutrophils in BALF were quantified at 48 h after the last challenge. (B) Gating strategy for neutrophils is define as cells with the following characteristics: CD45(+)CD11b(+)Ly6G(+). (C) Neutrophil expression in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images in each group are shown. (D) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of BALF and lung tissue by flow cytometry. (E) The cytokines interleukin (IL)-4, IL-17A, IL-6 and IL-1β levels in BALF were measured by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (E); n = 4 in (C) and (D). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nNeutrophil inflammatory infiltration was more pronounced in the CFA-induced neutrophil mouse models. (A) The number of total cells, eosinophils and neutrophils in BALF were quantified at 48 h after the last challenge. (B) Gating strategy for neutrophils is define as cells with the following characteristics: CD45(+)CD11b(+)Ly6G(+). (C) Neutrophil expression in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images in each group are shown. (D) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of BALF and lung tissue by flow cytometry. (E) The cytokines interleukin (IL)-4, IL-17A, IL-6 and IL-1β levels in BALF were measured by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (E); n = 4 in (C) and (D). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "To further explore the mechanism of neutrophils in mouse model of neutrophilic asthma, we first assessed neutrophil infiltration in mouse peripheral blood and bone marrow by flow cytometry. Results revealed that the percentage of neutrophils in the CFA group in both peripheral blood and bone marrow were found to be highest among all five groups. Although the percentage of neutrophils in the 0.1LPS group and the 10LPS group increased significantly relative to the control group, it was not as significant as the CFA group. In the flow cytometry analysis of bone marrow cells, the percentage of CD11b(+)Ly6G(+) neutrophils in the 0.1LPS group and 10LPS group lower than that in the EOS group (Fig. 3A and 3B). As the first line of defense, neutrophils partially mediate host responses by establishing NETs [20]. PMA is a very powerful, but non-physiological stimulus for NET formation [18]. We next purified mouse bone marrow neutrophils and stimulated with PMA (100 nM) in vitro to see whether NETs were released. Confocal microscopy revealed that a large number of NETosis neutrophils and NETs were detected in all three neutrophil models, with the highest amount of that produced in the CFA group. There was a small number of NETs observed in the EOS group (Fig. 3C-F). We also found that only a minority of neutrophils developed NETosis in the absence of stimulation, whether isolated from the bone marrow of the neutrophil models or other models. In addition, we detected the expression levels of MPO and CitH3 using western blot in lung tissue of mice, and the results were consistent with that of immunofluorescence analysis (Fig. 3G and H).\n\nFig. 3Abundant NETs occur in neutrophilic asthma models. (A) Neutrophil expression in CD45(+) leucocytes in mouse peripheral blood and bone marrow were determined by flow cytometry within 48 h after the last challenge. The representative images in each group are shown. (B) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of mouse peripheral blood and bone marrow by flow cytometry. (C) In vitro NET-formation assays with purified neutrophils of mouse bone marrow neutrophils stimulated with PMA (100 nM) or RPMI 1640 for 4 h. Then the neutrophils were stained PI and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (D) Comparison of the percentage of NETosis cells in each group upon PMA stimulation and control (Ctrl) stimulation. (E) After stimulated with PMA, the neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green) and DAPI (nuclear staining, blue) and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (F) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (G, H) Western blot was used to detect the levels of MPO and CitH3 protein in lung tissue of five groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S1 and S4 presents the full-length blots. Data were shown as mean ± SEM; n = 4. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nAbundant NETs occur in neutrophilic asthma models. (A) Neutrophil expression in CD45(+) leucocytes in mouse peripheral blood and bone marrow were determined by flow cytometry within 48 h after the last challenge. The representative images in each group are shown. (B) Quantification: the percentage of CD11b(+)Ly6G(+) neutrophils in the CD45(+) leucocytes gate of mouse peripheral blood and bone marrow by flow cytometry. (C) In vitro NET-formation assays with purified neutrophils of mouse bone marrow neutrophils stimulated with PMA (100 nM) or RPMI 1640 for 4 h. Then the neutrophils were stained PI and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (D) Comparison of the percentage of NETosis cells in each group upon PMA stimulation and control (Ctrl) stimulation. (E) After stimulated with PMA, the neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green) and DAPI (nuclear staining, blue) and analyzed by immunofluorescence confocal microscopy. Representative immunofluorescence images of NETs, Scale bar = 20 μm. (F) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (G, H) Western blot was used to detect the levels of MPO and CitH3 protein in lung tissue of five groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S1 and S4 presents the full-length blots. Data were shown as mean ± SEM; n = 4. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "We summarize the above results and conclude that the CFA group is a relatively more suitable model of neutrophilic asthma, although the AHR of this model is not significant. To overcome this, we attempted to establish a new murine model combining the sensitization methods of the 0.1LPS group and the CFA group (0.1LPS + CFA group) (Fig. 4A). Result shown that the 0.1LPS + CFA group exhibited higher AHR in response to methacholine compared with the 0.1LPS or CFA group, although the differences were not statistically significant (Fig. 4B). Similar to the CFA group, lung histopathological examination in 0.1LPS + CFA group showed obvious inflammatory cells infiltration and airway mucus secretion, some airways showed local airway mucus obstruction (Fig. 4C-F). These results seem to indicate that the 0.1LPS + CFA group serve as a suitable model of mouse to study the pathogenesis of the neutrophilic asthma.\n\nFig. 4The 0.1LPS and CFA combined with OVA-induced neutrophilic asthma model exhibits significant AHR and severe airway infiltration. (A) Schematic diagram of the experiment. (B) The AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used to assess impaired lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nThe 0.1LPS and CFA combined with OVA-induced neutrophilic asthma model exhibits significant AHR and severe airway infiltration. (A) Schematic diagram of the experiment. (B) The AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used to assess impaired lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung tissue. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. Data were shown as mean ± SEM; n = 5. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "Immediately after, we performed BALF and lung tissue cells analysis. The total number of BALF in the 0.1LPS + CFA group was significantly higher than that in the control and 0.1LPS groups, and there was no significant difference in total number of BALF between the 0.1LPS + CFA and CFA groups (Fig. 5A). Similar results were observed between the groups in the percentage of neutrophils (Fig. 5A). The percentage of eosinophils in the BALF of the three neutrophil groups had no significant difference compared with each other, while higher than that in the control group (Fig. 5A). Likewise, we performed flow cytometry on BALF cells and single lung-cell suspensions to better understand our newly established neutrophil model. The percentage of neutrophils in CD45(+) leukocytes in the BALF and single lung-cell suspensions in both 0.1LPS + CFA and CFA groups was significantly increased, and the difference was statistically significant (Fig. 5B and C). The 0.1LPS group also shown more percentage of neutrophils in CD45(+) leukocytes compared with the control group, but no statistical significance was achieved (Fig. 5B and C). Next, we quantification the expression of inflammatory cytokines in BALF. As expected, the expression of IL-17A, IL-6 and IL-1β in the 0.1LPS + CFA group and other two neutrophil groups were strongly increased compared with the control group. However, the expression of IL-4 was not significantly different in all four groups (Fig. 5D).\n\nFig. 5Significant neutrophilic inflammation and secretion of proinflammatory factors were observed in CFA combined with LPS induced mouse model. (A) Quantification of total cell counts and differential cell counts (eosinophils and neutrophils) in BALF at 48 h after the last challenge. (B) The percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images of each group are shown. (C) Statistical analysis of the above data shown by flow cytometry. (D) Cytokine (IL-4, IL-6 and IL-1β) concentrations in BALF were quantified by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (D); n = 4 mice in (B) and (C). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nSignificant neutrophilic inflammation and secretion of proinflammatory factors were observed in CFA combined with LPS induced mouse model. (A) Quantification of total cell counts and differential cell counts (eosinophils and neutrophils) in BALF at 48 h after the last challenge. (B) The percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leucocytes in BALF and lung tissue was determined by flow cytometry at 48 h after the last challenge. The representative images of each group are shown. (C) Statistical analysis of the above data shown by flow cytometry. (D) Cytokine (IL-4, IL-6 and IL-1β) concentrations in BALF were quantified by enzyme-linked immunosorbent assay (ELISA). Data were shown as mean ± SEM; n = 5 in (A) and (D); n = 4 mice in (B) and (C). Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "Likewise, to investigate whether neutrophils in the 0.1LPS + CFA group asthma mouse model also have a similar mechanism to the neutrophil groups described above, we also evaluated neutrophil infiltration in mouse peripheral blood and bone marrow by flow cytometry. Results demonstrate that the percentage of neutrophils in peripheral blood and bone marrow in the 0.1LPS + CFA group was significantly higher than that in the control group, which was comparable to the CFA group. The percentage of neutrophils in the 0.1LPS group was increased compared with the control group, but there was no statistical difference (Fig. 6A and B). Confocal microscopy demonstrated that NETs co-localized with MPO, citH3 and DAPI were detected in all three neutrophil models after PMA stimulation, while the 0.1LPS + CFA group and CFA group could observe the most significant NETs release (Fig. 6C-E). Western blot also showed that the protein expression levels of MPO and CitH3 in the lung tissue of mice in each group were consistent with the results of immunofluorescence (Fig. 6F and G).\n\nFig. 6Neutrophilic mouse model induced by CFA combined with LPS showed enhanced NETs formation capacity. (A) At 48 h after the last challenge, the percentage of neutrophil populations in CD45(+) leukocytes in mouse peripheral blood and bone marrow were determined by flow cytometry. The representative images in each group are shown. (B) Statistical analysis of the percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leukocyte gate of mouse peripheral blood and bone marrow by flow cytometry. (C) Neutrophils were purified from mouse bone marrow and stimulated with PMA (100 nM) or vehicle control for 4 h. Then, neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green), and DNA (DAPI, blue) and confocal by immunofluorescence microscope for analysis. Representative images of NETs immunofluorescence. Scale bar = 20 μm. (D) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (E) Representative z-axis images of the NETs immunofluorescence in 0.1LPS + CFA group. Scale bar = 10 μm. (F, G) Western blot analysis the protein expression level of MPO and CitH3 in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S2 and S5 presents the full-length blots. Data were shown as mean ± SEM; n = 4 or 6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nNeutrophilic mouse model induced by CFA combined with LPS showed enhanced NETs formation capacity. (A) At 48 h after the last challenge, the percentage of neutrophil populations in CD45(+) leukocytes in mouse peripheral blood and bone marrow were determined by flow cytometry. The representative images in each group are shown. (B) Statistical analysis of the percentage of CD11b(+)Ly6G(+) neutrophils in CD45(+) leukocyte gate of mouse peripheral blood and bone marrow by flow cytometry. (C) Neutrophils were purified from mouse bone marrow and stimulated with PMA (100 nM) or vehicle control for 4 h. Then, neutrophils were stained for myeloperoxidase (MPO, red), citrullinated histone 3 (CitH3, green), and DNA (DAPI, blue) and confocal by immunofluorescence microscope for analysis. Representative images of NETs immunofluorescence. Scale bar = 20 μm. (D) Percentage of NETs area normalized to MPO positive signal in mouse bone marrow neutrophils after PMA stimulation. (E) Representative z-axis images of the NETs immunofluorescence in 0.1LPS + CFA group. Scale bar = 10 μm. (F, G) Western blot analysis the protein expression level of MPO and CitH3 in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S2 and S5 presents the full-length blots. Data were shown as mean ± SEM; n = 4 or 6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "To determine whether the presence of NETs contributes to the pathogenesis of neutrophilic asthma model, we treated mice with either DNase I or CI-amidine before each challenge (Fig. 7 A). Previous studies have shown that DNase I can break down NETs [21], while activation of PAD4 is a major driver of NETs formation, and CI-amidine is an irreversible PAD4 inhibitor [22, 23]. Here, we found that DNase I or CI-amidine treatment significantly reduced airway hyperresponsiveness (Fig. 7B) and alleviated airway inflammation in the 0.1LPS + CFA group (Fig. 7C and E), PAS staining showed that airway mucus obstruction disappeared (Fig. 7D and F). Analysis of inflammatory cells in BALF also indicated that both the total number of cells and the percentage of neutrophils were significantly reduced by DNase I or CI-amidine treatment (Fig. 7G). Western blot analysis of lung tissue showed that the expression of MPO and CitH3 were significantly reduced in the OVA/CFA/0.1 LPS + DNase I (DNase I) group or the OVA/CFA/0.1 LPS + CI-amidine (CI-amidine) group (Fig. 7 H and I).\n\nFig. 7DNase I or CI-amidine administration reduced airway hyperresponsiveness and alleviated airway inflammation (A) Experimental assay schematic for in vivo experiments. (B) AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used as an indicator of lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. (G) The total number of cells and the differential number of cells (eosinophils and neutrophils) were quantified 48 h after the last challenge in the BALF. (H, I) Western blot analysis to measure MPO and CitH3 protein expression level in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S3 and S6 presents the full-length blots. Data were shown as mean ± SEM; n = 4–6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001\n\nDNase I or CI-amidine administration reduced airway hyperresponsiveness and alleviated airway inflammation (A) Experimental assay schematic for in vivo experiments. (B) AHR was measured 24 h after the last challenge, enhanced pause (Penh) values were used as an indicator of lung function. (C) Hematoxylin and eosin (H&E) staining of lung tissue. Scale bar = 50 μm. (D) Paraffin acid-Schiff (PAS) staining of lung. Scale bar = 20 μm. (E) Quantification of inflammation infiltration score of the H&E staining. (F) Quantification of mucus-producing goblet cells of the PAS staining. (G) The total number of cells and the differential number of cells (eosinophils and neutrophils) were quantified 48 h after the last challenge in the BALF. (H, I) Western blot analysis to measure MPO and CitH3 protein expression level in lung tissue of four groups of mice. Expression is relative to β-Tubulin. Cropped blots are shown, and supplementary Fig. S3 and S6 presents the full-length blots. Data were shown as mean ± SEM; n = 4–6. Significance between groups was calculated using one-way ANOVA with Tukey’s post hoc method. *p < 0.05, **p < 0.01 and ***p < 0.001", "Asthma is a heterogeneous disease with multiple phenotypes and endotypes [24, 25]. Approximately 5-25% of asthmatics suffer from severe asthma and do not respond to existing medical treatment, those people account for 50-80% of all asthma-related health care costs [26]. Neutrophilic airway inflammation is one of the main features of severe asthma. As of today, although there are many studies on neutrophilic asthma, the pathogenesis of neutrophilic asthma is poorly understood, and there are still many difficulties in treatment.\nOVA is a classic allergen for establishing allergic asthmatic mouse models, which usually requires co-sensitization with appropriate adjuvants [27]. The type of OVA-induced airway inflammation varies depending on the adjuvant. OVA/CFA or OVA/LPS is often used to develop neutrophilic asthma mouse models by investigators. CFA is known as a powerful inflammatory stimulus and widely used experimental models of arthritis [28]. In recent years, many studies have successfully established neutrophilic asthma model using CFA combined with OVA [8, 10, 29]. This model have higher degrees of airway neutrophilic inflammation, however airway responsiveness is not significantly increased [30]. This could be explained by the fact that despite the association between AHR and airway inflammation, there is evidence that AHR can occur independently of inflammation [31–33]. A similar phenomenon was also found in our study, although Penh in the CFA group increased compared with the control group, it was lower than other neutrophil groups or even the eosinophil group.\nLPS is a major component of the outer membrane of gram-negative bacteria, which stimulates the innate immune response to inflammation [34]. The use of LPS alone cannot establish a mouse model of asthma, and it acts as an adjuvant in the asthma model. Neutrophilic airway inflammation and airway hyperresponsiveness can be induced when LPS is used in combination with OVA [9, 35]. However, in asthma models, the role of LPS remains controversial. LPS is present at high levels in air and dust, some studies indicated that the extent of LPS exposure negatively correlates with the risk of developing asthma [36], while others held the opposite opinion that LPS in the environment probably plays an important role in the occurrence of asthma exacerbations and insensitive responses to corticosteroids in humans [37]. According to Eisenbarth et al. [38], low dose LPS promote Th2 immunity, whereas high doses promote Th1 responses. This view is also supported by a recent study showing that exposure to low-dose LPS (0.1 µg) in BALB/c mice enhanced type 2 allergic asthma, whereas starting with a higher dose of LPS (10 µg) had no significant effect [39]. However, there is also study using 50 µg OVA with 0.1 µg LPS sensitization combined with OVA challenge to successfully establish Th17-dependent neutrophilic airway inflammation [9]. We speculate that different effects of LPS in various studies in asthma are due to differences in time and dose. Following the studies described above, we established the OVA/low-dose (0.1 µg) LPS and OVA/high-dose (10 µg) LPS models. Our work revealed that both the 0.1LPS group and 10LPS group showed significant airway hyperresponsiveness, much higher than the CFA group, while the inflammatory cell infiltration in the 0.1LPS group and 10LPS group was lower than that in the CFA group. Based on the above conclusions, with consideration of high doses of LPS may cause acute lung injury, we combined OVA/CFA/0.1LPS to establish a mouse model and subsequent experimental validations. The results showed that the 0.1LPS + CFA group exhibited marked airway hyperresponsiveness and airway inflammation.\nNETs are composed of histones, neutrophil elastase (NE), MPO and double-stranded (ds) DNA [40, 41]. NETs are important in antibacterial defense, helping to limit systemic infection and maintain host defenses against fungal pathogens [42]. When the body was stimulated by pathogenic or chemical stimuli, the neutrophils use degranulation, phagocytosis, and the production of NETs to control initial infections [43]. However, with our knowledge about NETs have been greatly expanded in recent years, accumulating evidence indicated that NETs are considered to be a double-edged sword in lung disease. Infectious and noninfectious pulmonary diseases led to large-scale neutrophil infiltration into the lungs, and activated neutrophils release substantial amounts of NETs [44–46]. However, excessive NETs generation in noninfectious settings could be damaging for the tissue/organ, excessive NETs formation increases mucus viscosity and fills the lungs, impairing lung function [44, 47]. In the neutrophilic asthma models of our study, the group with more severe airway inflammation also produced more NETs. In the pathological staining of the 0.1LPS + CFA group, we could even see a large amount of mucus occluded the lumen. We speculate that this is related to the oversecretion of NETs. It has been shown that BALF from patients with severe asthma had detectable NETs that were positively correlated with IL-17 levels [48]. IL-1β is a proinflammatory cytokine central to the inflammatory response driven by the IL-6 signaling pathway [49]. Proinflammatory cytokines such as IL-1β, IL-6 are up-regulated when neutrophils undergo NETosis [50]. Also in our work, we observed increased expression of IL17A, IL-1β and IL-6 in the neutrophilic asthma models.\nWe validated our results by increasing NETs degradation or decreasing NETs generation to determine that the presence of NETs exacerbated disease in neutrophilic asthma model. In our previous experiments, we found that airway mucus secretion and airway responsiveness were higher in the 0.1LPS + CFA group than that in the other groups. After treated with DNase I, the airway hyperreactivity and mucus production of the 0.1LPS + CFA group, however, were almost completely eliminated. It has also previously been shown that airway mucus embolism in asthmatic patients can achieve complete lysis within minutes after administration of recombinant human DNase [51]. It now appears that recombinant human DNase can relieve airway mucus embolism, possibly by degrading airway NETs. Activation of PAD4 is likely a major driver of NETosis, and histone citrullination catalyzed by PAD4 appears to be a critical step in NETosis [22, 52]. Similarly, we demonstrate that a PAD4 inhibitor, CI-amidine, can significantly alleviate airway inflammation and airway hyperresponsiveness in neutrophilic asthma model. Both above-mentioned results confirmed NETs is critical to the pathogenesis of neutrophilic asthma model. In addition, we found that DNase I and CI-amidine had no significant impact on AHR of EOS mice (Fig S7). This can be explained as follows: DNase I and CI-amidine play a role in neutrophilic asthma model mainly because they can increase NETs degradation or decrease NETs generation. For NETs that are not highly expressed in the EOS group, DNase I and CI-amidine did not work.\nHowever, the current study did have some shortcomings as well. How NETs is regulated in neutrophilic asthma remains unknown. Meanwhile, we also have recognized our study lacks the support of clinical correlative data. The issues mentioned above will require further study.", "In conclusion, our findings identify a novel mouse model of neutrophilic asthma induced by OVA, CFA and LPS. This kind of neutrophilic asthma model is phenotypically more comprehensive, and characterized by AHR, massive infiltration of neutrophils and production of non-Th2 cytokines. Our data also suggest that a large number of NETs are generated in vivo and in vitro in the neutrophilic asthma models, reducing NETs can reverse those changes mentioned above. These findings provide further insight into the asthma model studies and pathogenesis of neutrophilic asthma, targeting NETs is a novel strategy that may be effective for treating neutrophilic asthma in the future.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1Fig. S1. Full length blots of Fig. 3G.Fig. S2. Full length blots of Fig. 6 F.Fig. S3. Full length blots of Fig. 7 H.Fig. S4. Additional bands used for statistics in Fig. 3 H.Fig. S5. Additional bands used for statistics in Fig. 6G.Fig. S6. Additional bands used for statistics in Fig. 7I.\n\nSupplementary Material 1\nFig. S1. Full length blots of Fig. 3G.\nFig. S2. Full length blots of Fig. 6 F.\nFig. S3. Full length blots of Fig. 7 H.\nFig. S4. Additional bands used for statistics in Fig. 3 H.\nFig. S5. Additional bands used for statistics in Fig. 6G.\nFig. S6. Additional bands used for statistics in Fig. 7I.\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1Fig. S1. Full length blots of Fig. 3G.Fig. S2. Full length blots of Fig. 6 F.Fig. S3. Full length blots of Fig. 7 H.Fig. S4. Additional bands used for statistics in Fig. 3 H.Fig. S5. Additional bands used for statistics in Fig. 6G.Fig. S6. Additional bands used for statistics in Fig. 7I.\n\nSupplementary Material 1\nFig. S1. Full length blots of Fig. 3G.\nFig. S2. Full length blots of Fig. 6 F.\nFig. S3. Full length blots of Fig. 7 H.\nFig. S4. Additional bands used for statistics in Fig. 3 H.\nFig. S5. Additional bands used for statistics in Fig. 6G.\nFig. S6. Additional bands used for statistics in Fig. 7I.", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1Fig. S1. Full length blots of Fig. 3G.Fig. S2. Full length blots of Fig. 6 F.Fig. S3. Full length blots of Fig. 7 H.Fig. S4. Additional bands used for statistics in Fig. 3 H.Fig. S5. Additional bands used for statistics in Fig. 6G.Fig. S6. Additional bands used for statistics in Fig. 7I.\n\nSupplementary Material 1\nFig. S1. Full length blots of Fig. 3G.\nFig. S2. Full length blots of Fig. 6 F.\nFig. S3. Full length blots of Fig. 7 H.\nFig. S4. Additional bands used for statistics in Fig. 3 H.\nFig. S5. Additional bands used for statistics in Fig. 6G.\nFig. S6. Additional bands used for statistics in Fig. 7I." ]
[ "introduction", "materials|methods", null, null, null, null, null, null, null, null, null, null, "results", null, null, null, null, null, null, null, "discussion", "conclusion", "supplementary-material", null ]
[ "Neutrophilic asthma", "Neutrophil", "Neutrophil extracellular traps", "Airway inflammation" ]
Influence of optical zone on myopic correction in small incision lenticule extraction: a short-term study.
36271372
To evaluate the influence of preoperative optical zone on myopic correction in small incision lenticule extraction.
BACKGROUND
In this retrospective clinical study, 581 eyes from 316 patients underwent SMILE were selected, including 117 eyes in the small optical zone group (range from 6.0 to 6.4 mm) and 464 eyes in the large optical zone group (range from 6.5 to 6.8 mm). The measurements included uncorrected distance visual acuity (UDVA), corrected distance visual acuity (CDVA), spherical, and cylinder were measured preoperatively and 3 months postoperatively. Propensity score match (PSM) analysis was performed with age, gender, eye (right/left), keratometry and preoperative spherical equivalent between two different groups. The influence of optical zones on postoperative refractive outcomes were evaluated using univariate regression analysis.
METHODS
In total, 78 pairs of eyes were selected by PSM (match ratio 1:1). There were no differences in the age, gender, eye (right/left), keratometry or preoperative spherical equivalent between the small and large optical zone groups. However, the difference of postoperative spherical equivalent was significantly between groups. Patients with larger optical zones had a trend towards less undercorrection (P = 0.018). Univariate linear regression model analysis found that each millimeter larger optical zone resulted in 8.13% or 0.39D less undercorrection (P < 0.001). The dependency between the optical zones and postoperative spherical equivalent was significant in the higher preoperative myopia group (r = 0.281, P < 0.001), but not significant in the lower myopia group (r = 0.028, P = 0.702).
RESULTS
The diameter of optical zones would affect postoperative refractive outcomes in small incision lenticule extraction. This study indicated that larger optical zones induced less undercorrection, especially in patients with high myopia.
CONCLUSION
[ "Humans", "Lasers, Excimer", "Retrospective Studies", "Myopia", "Refraction, Ocular", "Visual Acuity", "Corneal Stroma", "Treatment Outcome", "Astigmatism", "Microsurgery" ]
9585829
null
null
null
null
Results
Three hundred and sixteen patients (581 eyes) were enrolled, including 165 male and 151 female. The mean age was 23.36 ± 5.48 years (range from 18 to 49 years). The clinical information of subjects is shown in Table 1. Table 1Characteristics of Patients Undergoing SMILEParameterMean ± SDAge (y)23.36 ± 5.48Sphere (D)-4.42 ± 1.52Cylinder (D)-0.78 ± 0.67SE (D)-4.81 ± 1.51IOP (mmHg)17.57 ± 2.55CCT (um)543.85 ± 26.85Ablation depth (um)106.36 ± 19.32Scotopic pupil size (mm)6.75 ± 0.74D = diopters; SE = spherical equivalent; IOP = intraocular pressure; CCT = central corneal thickness Characteristics of Patients Undergoing SMILE D = diopters; SE = spherical equivalent; IOP = intraocular pressure; CCT = central corneal thickness
null
null
[ "Background", "Methods", "Patients", "Surgery", "Clinical examinations", "Statistical analysis", "Safety and efficacy", "Predictability", "Refractive outcomes in small and large optical zone groups", "Effect of optical zones on refractive outcomes" ]
[ "Small incision lenticule extraction (SMILE) is a new minimally invasive corneal refractive surgery for the correction of myopia and myopic astigmatism [1–3]. It creates a lenticule of through a 2.0 -2.5 mm incision based on the use of femtosecond laser [4]. In recent years, SMILE has been established as an effective, predictable, safe and stable refractive surgery solution [5–12]. However, overcorrection and undercorrection still exists after SMILE [1, 13, 14].\nSeveral studies have shown that SMILE refractive outcome could be influenced by the age, gender, keratometry, preoperative spherical equivalent and optical zone [15–20]. The optical zone refers to extracted lenticule size that is designed before SMILE surgery. The scotopic pupil size, original corneal thickness and preoperative spherical equivalent are major factors to influence the clinical decision while planning the treatment zone. A refractive surgeon might prefer to design a larger optical zone to avoid the night vision complaints, such as glare, halo, and ghosting when the scotopic pupil size is relatively large [21]. However, a larger optical zone requires more corneal tissue for a given spherical equivalent refraction correction [22]. For some high myopic patients with thin cornea, a relatively smaller optical zone might be selected to save corneal tissue [23, 24]. Therefore, it’s difficult to design a proper optical zone to strike a balance between the postoperative visual quality and safety in SMILE surgery. The purpose of our study is to evaluate the relationship between optical zones and refractive outcome after SMILE for the treatment of myopia with or without astigmatism.", "[SUBTITLE] Patients [SUBSECTION] This was a comparative, retrospective clinical study. The study followed the tenets of the Declaration of Helsinki and was approved by the ethics committee of the Second Affiliated Hospital of Harbin Medical University. Patients undergoing SMILE were enrolled in this study between 2019 and 2021 in the Second Affiliated Hospital of Harbin Medical University. The inclusion criteria were as follows: 18 years or older, spherical refraction up to -9.00 diopters (D), myopic astigmatism up to 3.00 D; refraction change less than 0.50 D for the past two years, and CDVA of 20/30 or better. The exclusion criteria were the presence of active ocular disease, ocular trauma, suspected keratoconus and the expected residual stromal bed less than 250 μm. Soft contact lenses were discontinued for 2 weeks, and rigid lenses for 4 weeks before surgery.\nThis was a comparative, retrospective clinical study. The study followed the tenets of the Declaration of Helsinki and was approved by the ethics committee of the Second Affiliated Hospital of Harbin Medical University. Patients undergoing SMILE were enrolled in this study between 2019 and 2021 in the Second Affiliated Hospital of Harbin Medical University. The inclusion criteria were as follows: 18 years or older, spherical refraction up to -9.00 diopters (D), myopic astigmatism up to 3.00 D; refraction change less than 0.50 D for the past two years, and CDVA of 20/30 or better. The exclusion criteria were the presence of active ocular disease, ocular trauma, suspected keratoconus and the expected residual stromal bed less than 250 μm. Soft contact lenses were discontinued for 2 weeks, and rigid lenses for 4 weeks before surgery.\n[SUBTITLE] Surgery [SUBSECTION] All surgeries procedures were performed with the VisuMax femtosecond laser (Carl Zeiss Meditec AG) in the Second Affiliated Hospital of Harbin Medical University. The optical zone was 6.0 to 6.8 mm, and the cap diameter was 7.6 mm. The predetermined cap thickness was 100 to 120 μm, and the pulse energy ranged from 125 to 160 nJ. The side cut was placed at the 10-o’clock position of the cornea with an angle of 120 degrees and a circumferential width of 2 mm. The lenticule was extracted through the incision and all patients received 0.3% ofloxacin eye drops (Santen Pharmaceutical Co., Ltd., Osaka, Japan.) four times a day for 1 week, 0.1% fluorometholone eye drops (Santen Pharmaceutical Co., Ltd.) four times a day for 2 weeks, and 0.1% sodium hyaluronate eye drops (Santen Pharmaceutical Co., Ltd.) four times a day for 2–3 months.\nAll surgeries procedures were performed with the VisuMax femtosecond laser (Carl Zeiss Meditec AG) in the Second Affiliated Hospital of Harbin Medical University. The optical zone was 6.0 to 6.8 mm, and the cap diameter was 7.6 mm. The predetermined cap thickness was 100 to 120 μm, and the pulse energy ranged from 125 to 160 nJ. The side cut was placed at the 10-o’clock position of the cornea with an angle of 120 degrees and a circumferential width of 2 mm. The lenticule was extracted through the incision and all patients received 0.3% ofloxacin eye drops (Santen Pharmaceutical Co., Ltd., Osaka, Japan.) four times a day for 1 week, 0.1% fluorometholone eye drops (Santen Pharmaceutical Co., Ltd.) four times a day for 2 weeks, and 0.1% sodium hyaluronate eye drops (Santen Pharmaceutical Co., Ltd.) four times a day for 2–3 months.\n[SUBTITLE] Clinical examinations [SUBSECTION] The following measurements were included preoperatively and 3 months after SMILE for all patients: slit-lamp biomicroscopy, fundus examinations, intraocular pressure, corneal topography via the anterior eye segment analysis system (Sirius, CSO, Italy), UDVA, CDVA, cycloplegic and subjective refractions. In order to evaluate the influence of optical zone on SMILE outcomes, all patients were divided into small optical zone group (range from 6.0 to 6.4 mm) and large optical zone group (range from 6.5 to 6.8 mm).\nThe following measurements were included preoperatively and 3 months after SMILE for all patients: slit-lamp biomicroscopy, fundus examinations, intraocular pressure, corneal topography via the anterior eye segment analysis system (Sirius, CSO, Italy), UDVA, CDVA, cycloplegic and subjective refractions. In order to evaluate the influence of optical zone on SMILE outcomes, all patients were divided into small optical zone group (range from 6.0 to 6.4 mm) and large optical zone group (range from 6.5 to 6.8 mm).\n[SUBTITLE] Statistical analysis [SUBSECTION] Data analyses were performed using SPSS software version 26.0. Normality of the data was confirmed by the Kolmogorov-Smirnov test. Data are expressed as the mean ± standard deviation. The propensity score match (PSM) analysis was used to eliminate preoperative confounding factors between small and large optical zone groups. Unpaired two-tailed t test was performed to determine difference between two groups. Pearson correlation and univariate regression analyses were used to determine the relationship between optical zones and postoperative spherical equivalent. To avoid the influence of preoperative refractive status, the cohort were segregated into thirds based on preoperative spherical equivalent. Pearson correlation and univariate regression analyses were repeated in the lower third (from minimum to percentile 33) and the upper third (from percentile 67 to maximum) of preoperative SE respectively. For all cases, P value < 0.05 was considered to be statistically significant.\nData analyses were performed using SPSS software version 26.0. Normality of the data was confirmed by the Kolmogorov-Smirnov test. Data are expressed as the mean ± standard deviation. The propensity score match (PSM) analysis was used to eliminate preoperative confounding factors between small and large optical zone groups. Unpaired two-tailed t test was performed to determine difference between two groups. Pearson correlation and univariate regression analyses were used to determine the relationship between optical zones and postoperative spherical equivalent. To avoid the influence of preoperative refractive status, the cohort were segregated into thirds based on preoperative spherical equivalent. Pearson correlation and univariate regression analyses were repeated in the lower third (from minimum to percentile 33) and the upper third (from percentile 67 to maximum) of preoperative SE respectively. For all cases, P value < 0.05 was considered to be statistically significant.", "This was a comparative, retrospective clinical study. The study followed the tenets of the Declaration of Helsinki and was approved by the ethics committee of the Second Affiliated Hospital of Harbin Medical University. Patients undergoing SMILE were enrolled in this study between 2019 and 2021 in the Second Affiliated Hospital of Harbin Medical University. The inclusion criteria were as follows: 18 years or older, spherical refraction up to -9.00 diopters (D), myopic astigmatism up to 3.00 D; refraction change less than 0.50 D for the past two years, and CDVA of 20/30 or better. The exclusion criteria were the presence of active ocular disease, ocular trauma, suspected keratoconus and the expected residual stromal bed less than 250 μm. Soft contact lenses were discontinued for 2 weeks, and rigid lenses for 4 weeks before surgery.", "All surgeries procedures were performed with the VisuMax femtosecond laser (Carl Zeiss Meditec AG) in the Second Affiliated Hospital of Harbin Medical University. The optical zone was 6.0 to 6.8 mm, and the cap diameter was 7.6 mm. The predetermined cap thickness was 100 to 120 μm, and the pulse energy ranged from 125 to 160 nJ. The side cut was placed at the 10-o’clock position of the cornea with an angle of 120 degrees and a circumferential width of 2 mm. The lenticule was extracted through the incision and all patients received 0.3% ofloxacin eye drops (Santen Pharmaceutical Co., Ltd., Osaka, Japan.) four times a day for 1 week, 0.1% fluorometholone eye drops (Santen Pharmaceutical Co., Ltd.) four times a day for 2 weeks, and 0.1% sodium hyaluronate eye drops (Santen Pharmaceutical Co., Ltd.) four times a day for 2–3 months.", "The following measurements were included preoperatively and 3 months after SMILE for all patients: slit-lamp biomicroscopy, fundus examinations, intraocular pressure, corneal topography via the anterior eye segment analysis system (Sirius, CSO, Italy), UDVA, CDVA, cycloplegic and subjective refractions. In order to evaluate the influence of optical zone on SMILE outcomes, all patients were divided into small optical zone group (range from 6.0 to 6.4 mm) and large optical zone group (range from 6.5 to 6.8 mm).", "Data analyses were performed using SPSS software version 26.0. Normality of the data was confirmed by the Kolmogorov-Smirnov test. Data are expressed as the mean ± standard deviation. The propensity score match (PSM) analysis was used to eliminate preoperative confounding factors between small and large optical zone groups. Unpaired two-tailed t test was performed to determine difference between two groups. Pearson correlation and univariate regression analyses were used to determine the relationship between optical zones and postoperative spherical equivalent. To avoid the influence of preoperative refractive status, the cohort were segregated into thirds based on preoperative spherical equivalent. Pearson correlation and univariate regression analyses were repeated in the lower third (from minimum to percentile 33) and the upper third (from percentile 67 to maximum) of preoperative SE respectively. For all cases, P value < 0.05 was considered to be statistically significant.", "Three months postoperatively, 563 (97%) eyes had a UDVA of 20/20 or better (Fig. 1). 521 (90%) eyes had a UDVA same or better than preoperative CDVA (Fig. 2). The mean efficacy index (ratio of postoperative UDVA to preoperative CDVA) was 1.18 ± 0.23. The CDVA remained the same in 203 (35%) eyes, whereas 376 (65%) eyes improved and 2 eyes lost one line or more at postoperative month 3 (Fig. 3). The mean safety index (ratio of postoperative CDVA to preoperative CDVA) was 1.22 ± 0.20.\n\nFig. 1Cumulative postoperative uncorrected distance visual acuity (UDVA) and preoperative corrected distance visual acuity (CDVA) 3 months after SMILE\n\nCumulative postoperative uncorrected distance visual acuity (UDVA) and preoperative corrected distance visual acuity (CDVA) 3 months after SMILE\n\nFig. 2Distribution of the change in lines of postoperative uncorrected distance visual acuity (UDVA) to preoperative corrected distance visual acuity (CDVA)\n\nDistribution of the change in lines of postoperative uncorrected distance visual acuity (UDVA) to preoperative corrected distance visual acuity (CDVA)\n\nFig. 3Distribution of the change in lines of corrected distance visual acuity (CDVA) 3 months postoperatively\n\nDistribution of the change in lines of corrected distance visual acuity (CDVA) 3 months postoperatively", "The scatterplot of the attempted versus the achieved change in spherical equivalent refraction at 3 months after SMILE is shown in Fig. 4. The relationship between attempted and achieved correction is high with a correlation coefficient of 0.95. 525 (90%) eyes had an SE within ± 0.50 D and 581 (100%) eyes within ± 1.00D at month 3 postoperatively (Fig. 5).\n\nFig. 4Attempted versus achieved change in spherical equivalent refraction (SEQ) 3 months after SMILE\n\nAttempted versus achieved change in spherical equivalent refraction (SEQ) 3 months after SMILE\n\nFig. 5Distribution of postoperative spherical equivalent (SE) after surgery\n\nDistribution of postoperative spherical equivalent (SE) after surgery", "There were 78 pairs of eyes matched by 1:1 PSM analysis between the small and large optical zone groups. After matching two groups for age, gender, eye (right/left), mean keratometry, and preoperative SE, the difference of postoperative SE was significantly between the small and large optical zone groups. Eyes with larger optical zone had a tendency to undercorrection at 3 months after surgery (P < 0.05) (Table 2).\n\nTable 2Clinical characteristics after matching by PSM between the small and large optical zone groupsParameterSmall groupLarge group\nP\nNo. of eyes7878-Age (y)23.54 ± 5.6323.50 ± 5.940.927Gender (M, %)44%45%0.747Eye (OD, %)47%47%1.000Preoperative SE (D)-5.91 ± 0.92-5.81 ± 0.990.484Mean keratometry (D)43.17 ± 1.3143.21 ± 1.270.859Optical Zone (mm)6.23 ± 0.166.53 ± 0.08< 0.001Range6.0 to 6.46.5 to 6.8Postoperative SE (D)-0.10 ± 0.290.02 ± 0.340.018SE = spherical equivalent; D = diopters\n\nClinical characteristics after matching by PSM between the small and large optical zone groups\nSE = spherical equivalent; D = diopters", "The dependency between the optical zones and postoperative SE was significant (r = 0.247, P < 0.001). The patients with larger optical zones had less undercorrection. The relationship between optical zones and postoperative SE were showed in Fig. 6. Each millimeter larger optical zone resulted in 8.13% or 0.39D less undercorrection (R2 = 0.091, P < 0.001). The difference in postoperative SE between the lower third and upper third was significant (P < 0.001). The dependency between optical zones and postoperative SE was significant in the higher degree of preoperative myopia group (R2 = 0.079, P < 0.001), but not significant in the lower degree of myopia group (R2 = 0.001, P = 0.702) (Table 3). The patients with a higher degree of preoperative myopia were more influenced by optical zones.\n\nFig. 6Scatterplot of the regression analysis between optical zone and postoperative spherical equivalent (SE)\n\nScatterplot of the regression analysis between optical zone and postoperative spherical equivalent (SE)\n\nTable 3Regression analysis between optical zones and postoperative SE in different preoperative refractive statusPreoperative SE (D)\nLower Third\n\nUpper Third\n\nParameter\n\n≤-4.00\n\n≥ -5.38\n\nP\nNo. of eyes193194Preoperative SE (D)Mean ± SD-3.18 ± 0.67-6.50 ± 0.89< 0.001Range-4.00 to -1.50-9.25 to -5.38Postoperative SE (D)Mean ± SD-0.17 ± 0.32-0.11 ± 0.49< 0.001\nr\n0.030.28Linear regression equationY= -0.07*X + 0.63Y = 0.48*X-3.03\nP\n0.702< 0.001SE = spherical equivalent; D = diopters\n\nRegression analysis between optical zones and postoperative SE in different preoperative refractive status\nSE = spherical equivalent; D = diopters" ]
[ null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Patients", "Surgery", "Clinical examinations", "Statistical analysis", "Results", "Safety and efficacy", "Predictability", "Refractive outcomes in small and large optical zone groups", "Effect of optical zones on refractive outcomes", "Discussion" ]
[ "Small incision lenticule extraction (SMILE) is a new minimally invasive corneal refractive surgery for the correction of myopia and myopic astigmatism [1–3]. It creates a lenticule of through a 2.0 -2.5 mm incision based on the use of femtosecond laser [4]. In recent years, SMILE has been established as an effective, predictable, safe and stable refractive surgery solution [5–12]. However, overcorrection and undercorrection still exists after SMILE [1, 13, 14].\nSeveral studies have shown that SMILE refractive outcome could be influenced by the age, gender, keratometry, preoperative spherical equivalent and optical zone [15–20]. The optical zone refers to extracted lenticule size that is designed before SMILE surgery. The scotopic pupil size, original corneal thickness and preoperative spherical equivalent are major factors to influence the clinical decision while planning the treatment zone. A refractive surgeon might prefer to design a larger optical zone to avoid the night vision complaints, such as glare, halo, and ghosting when the scotopic pupil size is relatively large [21]. However, a larger optical zone requires more corneal tissue for a given spherical equivalent refraction correction [22]. For some high myopic patients with thin cornea, a relatively smaller optical zone might be selected to save corneal tissue [23, 24]. Therefore, it’s difficult to design a proper optical zone to strike a balance between the postoperative visual quality and safety in SMILE surgery. The purpose of our study is to evaluate the relationship between optical zones and refractive outcome after SMILE for the treatment of myopia with or without astigmatism.", "[SUBTITLE] Patients [SUBSECTION] This was a comparative, retrospective clinical study. The study followed the tenets of the Declaration of Helsinki and was approved by the ethics committee of the Second Affiliated Hospital of Harbin Medical University. Patients undergoing SMILE were enrolled in this study between 2019 and 2021 in the Second Affiliated Hospital of Harbin Medical University. The inclusion criteria were as follows: 18 years or older, spherical refraction up to -9.00 diopters (D), myopic astigmatism up to 3.00 D; refraction change less than 0.50 D for the past two years, and CDVA of 20/30 or better. The exclusion criteria were the presence of active ocular disease, ocular trauma, suspected keratoconus and the expected residual stromal bed less than 250 μm. Soft contact lenses were discontinued for 2 weeks, and rigid lenses for 4 weeks before surgery.\nThis was a comparative, retrospective clinical study. The study followed the tenets of the Declaration of Helsinki and was approved by the ethics committee of the Second Affiliated Hospital of Harbin Medical University. Patients undergoing SMILE were enrolled in this study between 2019 and 2021 in the Second Affiliated Hospital of Harbin Medical University. The inclusion criteria were as follows: 18 years or older, spherical refraction up to -9.00 diopters (D), myopic astigmatism up to 3.00 D; refraction change less than 0.50 D for the past two years, and CDVA of 20/30 or better. The exclusion criteria were the presence of active ocular disease, ocular trauma, suspected keratoconus and the expected residual stromal bed less than 250 μm. Soft contact lenses were discontinued for 2 weeks, and rigid lenses for 4 weeks before surgery.\n[SUBTITLE] Surgery [SUBSECTION] All surgeries procedures were performed with the VisuMax femtosecond laser (Carl Zeiss Meditec AG) in the Second Affiliated Hospital of Harbin Medical University. The optical zone was 6.0 to 6.8 mm, and the cap diameter was 7.6 mm. The predetermined cap thickness was 100 to 120 μm, and the pulse energy ranged from 125 to 160 nJ. The side cut was placed at the 10-o’clock position of the cornea with an angle of 120 degrees and a circumferential width of 2 mm. The lenticule was extracted through the incision and all patients received 0.3% ofloxacin eye drops (Santen Pharmaceutical Co., Ltd., Osaka, Japan.) four times a day for 1 week, 0.1% fluorometholone eye drops (Santen Pharmaceutical Co., Ltd.) four times a day for 2 weeks, and 0.1% sodium hyaluronate eye drops (Santen Pharmaceutical Co., Ltd.) four times a day for 2–3 months.\nAll surgeries procedures were performed with the VisuMax femtosecond laser (Carl Zeiss Meditec AG) in the Second Affiliated Hospital of Harbin Medical University. The optical zone was 6.0 to 6.8 mm, and the cap diameter was 7.6 mm. The predetermined cap thickness was 100 to 120 μm, and the pulse energy ranged from 125 to 160 nJ. The side cut was placed at the 10-o’clock position of the cornea with an angle of 120 degrees and a circumferential width of 2 mm. The lenticule was extracted through the incision and all patients received 0.3% ofloxacin eye drops (Santen Pharmaceutical Co., Ltd., Osaka, Japan.) four times a day for 1 week, 0.1% fluorometholone eye drops (Santen Pharmaceutical Co., Ltd.) four times a day for 2 weeks, and 0.1% sodium hyaluronate eye drops (Santen Pharmaceutical Co., Ltd.) four times a day for 2–3 months.\n[SUBTITLE] Clinical examinations [SUBSECTION] The following measurements were included preoperatively and 3 months after SMILE for all patients: slit-lamp biomicroscopy, fundus examinations, intraocular pressure, corneal topography via the anterior eye segment analysis system (Sirius, CSO, Italy), UDVA, CDVA, cycloplegic and subjective refractions. In order to evaluate the influence of optical zone on SMILE outcomes, all patients were divided into small optical zone group (range from 6.0 to 6.4 mm) and large optical zone group (range from 6.5 to 6.8 mm).\nThe following measurements were included preoperatively and 3 months after SMILE for all patients: slit-lamp biomicroscopy, fundus examinations, intraocular pressure, corneal topography via the anterior eye segment analysis system (Sirius, CSO, Italy), UDVA, CDVA, cycloplegic and subjective refractions. In order to evaluate the influence of optical zone on SMILE outcomes, all patients were divided into small optical zone group (range from 6.0 to 6.4 mm) and large optical zone group (range from 6.5 to 6.8 mm).\n[SUBTITLE] Statistical analysis [SUBSECTION] Data analyses were performed using SPSS software version 26.0. Normality of the data was confirmed by the Kolmogorov-Smirnov test. Data are expressed as the mean ± standard deviation. The propensity score match (PSM) analysis was used to eliminate preoperative confounding factors between small and large optical zone groups. Unpaired two-tailed t test was performed to determine difference between two groups. Pearson correlation and univariate regression analyses were used to determine the relationship between optical zones and postoperative spherical equivalent. To avoid the influence of preoperative refractive status, the cohort were segregated into thirds based on preoperative spherical equivalent. Pearson correlation and univariate regression analyses were repeated in the lower third (from minimum to percentile 33) and the upper third (from percentile 67 to maximum) of preoperative SE respectively. For all cases, P value < 0.05 was considered to be statistically significant.\nData analyses were performed using SPSS software version 26.0. Normality of the data was confirmed by the Kolmogorov-Smirnov test. Data are expressed as the mean ± standard deviation. The propensity score match (PSM) analysis was used to eliminate preoperative confounding factors between small and large optical zone groups. Unpaired two-tailed t test was performed to determine difference between two groups. Pearson correlation and univariate regression analyses were used to determine the relationship between optical zones and postoperative spherical equivalent. To avoid the influence of preoperative refractive status, the cohort were segregated into thirds based on preoperative spherical equivalent. Pearson correlation and univariate regression analyses were repeated in the lower third (from minimum to percentile 33) and the upper third (from percentile 67 to maximum) of preoperative SE respectively. For all cases, P value < 0.05 was considered to be statistically significant.", "This was a comparative, retrospective clinical study. The study followed the tenets of the Declaration of Helsinki and was approved by the ethics committee of the Second Affiliated Hospital of Harbin Medical University. Patients undergoing SMILE were enrolled in this study between 2019 and 2021 in the Second Affiliated Hospital of Harbin Medical University. The inclusion criteria were as follows: 18 years or older, spherical refraction up to -9.00 diopters (D), myopic astigmatism up to 3.00 D; refraction change less than 0.50 D for the past two years, and CDVA of 20/30 or better. The exclusion criteria were the presence of active ocular disease, ocular trauma, suspected keratoconus and the expected residual stromal bed less than 250 μm. Soft contact lenses were discontinued for 2 weeks, and rigid lenses for 4 weeks before surgery.", "All surgeries procedures were performed with the VisuMax femtosecond laser (Carl Zeiss Meditec AG) in the Second Affiliated Hospital of Harbin Medical University. The optical zone was 6.0 to 6.8 mm, and the cap diameter was 7.6 mm. The predetermined cap thickness was 100 to 120 μm, and the pulse energy ranged from 125 to 160 nJ. The side cut was placed at the 10-o’clock position of the cornea with an angle of 120 degrees and a circumferential width of 2 mm. The lenticule was extracted through the incision and all patients received 0.3% ofloxacin eye drops (Santen Pharmaceutical Co., Ltd., Osaka, Japan.) four times a day for 1 week, 0.1% fluorometholone eye drops (Santen Pharmaceutical Co., Ltd.) four times a day for 2 weeks, and 0.1% sodium hyaluronate eye drops (Santen Pharmaceutical Co., Ltd.) four times a day for 2–3 months.", "The following measurements were included preoperatively and 3 months after SMILE for all patients: slit-lamp biomicroscopy, fundus examinations, intraocular pressure, corneal topography via the anterior eye segment analysis system (Sirius, CSO, Italy), UDVA, CDVA, cycloplegic and subjective refractions. In order to evaluate the influence of optical zone on SMILE outcomes, all patients were divided into small optical zone group (range from 6.0 to 6.4 mm) and large optical zone group (range from 6.5 to 6.8 mm).", "Data analyses were performed using SPSS software version 26.0. Normality of the data was confirmed by the Kolmogorov-Smirnov test. Data are expressed as the mean ± standard deviation. The propensity score match (PSM) analysis was used to eliminate preoperative confounding factors between small and large optical zone groups. Unpaired two-tailed t test was performed to determine difference between two groups. Pearson correlation and univariate regression analyses were used to determine the relationship between optical zones and postoperative spherical equivalent. To avoid the influence of preoperative refractive status, the cohort were segregated into thirds based on preoperative spherical equivalent. Pearson correlation and univariate regression analyses were repeated in the lower third (from minimum to percentile 33) and the upper third (from percentile 67 to maximum) of preoperative SE respectively. For all cases, P value < 0.05 was considered to be statistically significant.", "Three hundred and sixteen patients (581 eyes) were enrolled, including 165 male and 151 female. The mean age was 23.36 ± 5.48 years (range from 18 to 49 years). The clinical information of subjects is shown in Table 1.\n\nTable 1Characteristics of Patients Undergoing SMILEParameterMean ± SDAge (y)23.36 ± 5.48Sphere (D)-4.42 ± 1.52Cylinder (D)-0.78 ± 0.67SE (D)-4.81 ± 1.51IOP (mmHg)17.57 ± 2.55CCT (um)543.85 ± 26.85Ablation depth (um)106.36 ± 19.32Scotopic pupil size (mm)6.75 ± 0.74D = diopters; SE = spherical equivalent; IOP = intraocular pressure; CCT = central corneal thickness\n\nCharacteristics of Patients Undergoing SMILE\nD = diopters; SE = spherical equivalent; IOP = intraocular pressure; CCT = central corneal thickness", "Three months postoperatively, 563 (97%) eyes had a UDVA of 20/20 or better (Fig. 1). 521 (90%) eyes had a UDVA same or better than preoperative CDVA (Fig. 2). The mean efficacy index (ratio of postoperative UDVA to preoperative CDVA) was 1.18 ± 0.23. The CDVA remained the same in 203 (35%) eyes, whereas 376 (65%) eyes improved and 2 eyes lost one line or more at postoperative month 3 (Fig. 3). The mean safety index (ratio of postoperative CDVA to preoperative CDVA) was 1.22 ± 0.20.\n\nFig. 1Cumulative postoperative uncorrected distance visual acuity (UDVA) and preoperative corrected distance visual acuity (CDVA) 3 months after SMILE\n\nCumulative postoperative uncorrected distance visual acuity (UDVA) and preoperative corrected distance visual acuity (CDVA) 3 months after SMILE\n\nFig. 2Distribution of the change in lines of postoperative uncorrected distance visual acuity (UDVA) to preoperative corrected distance visual acuity (CDVA)\n\nDistribution of the change in lines of postoperative uncorrected distance visual acuity (UDVA) to preoperative corrected distance visual acuity (CDVA)\n\nFig. 3Distribution of the change in lines of corrected distance visual acuity (CDVA) 3 months postoperatively\n\nDistribution of the change in lines of corrected distance visual acuity (CDVA) 3 months postoperatively", "The scatterplot of the attempted versus the achieved change in spherical equivalent refraction at 3 months after SMILE is shown in Fig. 4. The relationship between attempted and achieved correction is high with a correlation coefficient of 0.95. 525 (90%) eyes had an SE within ± 0.50 D and 581 (100%) eyes within ± 1.00D at month 3 postoperatively (Fig. 5).\n\nFig. 4Attempted versus achieved change in spherical equivalent refraction (SEQ) 3 months after SMILE\n\nAttempted versus achieved change in spherical equivalent refraction (SEQ) 3 months after SMILE\n\nFig. 5Distribution of postoperative spherical equivalent (SE) after surgery\n\nDistribution of postoperative spherical equivalent (SE) after surgery", "There were 78 pairs of eyes matched by 1:1 PSM analysis between the small and large optical zone groups. After matching two groups for age, gender, eye (right/left), mean keratometry, and preoperative SE, the difference of postoperative SE was significantly between the small and large optical zone groups. Eyes with larger optical zone had a tendency to undercorrection at 3 months after surgery (P < 0.05) (Table 2).\n\nTable 2Clinical characteristics after matching by PSM between the small and large optical zone groupsParameterSmall groupLarge group\nP\nNo. of eyes7878-Age (y)23.54 ± 5.6323.50 ± 5.940.927Gender (M, %)44%45%0.747Eye (OD, %)47%47%1.000Preoperative SE (D)-5.91 ± 0.92-5.81 ± 0.990.484Mean keratometry (D)43.17 ± 1.3143.21 ± 1.270.859Optical Zone (mm)6.23 ± 0.166.53 ± 0.08< 0.001Range6.0 to 6.46.5 to 6.8Postoperative SE (D)-0.10 ± 0.290.02 ± 0.340.018SE = spherical equivalent; D = diopters\n\nClinical characteristics after matching by PSM between the small and large optical zone groups\nSE = spherical equivalent; D = diopters", "The dependency between the optical zones and postoperative SE was significant (r = 0.247, P < 0.001). The patients with larger optical zones had less undercorrection. The relationship between optical zones and postoperative SE were showed in Fig. 6. Each millimeter larger optical zone resulted in 8.13% or 0.39D less undercorrection (R2 = 0.091, P < 0.001). The difference in postoperative SE between the lower third and upper third was significant (P < 0.001). The dependency between optical zones and postoperative SE was significant in the higher degree of preoperative myopia group (R2 = 0.079, P < 0.001), but not significant in the lower degree of myopia group (R2 = 0.001, P = 0.702) (Table 3). The patients with a higher degree of preoperative myopia were more influenced by optical zones.\n\nFig. 6Scatterplot of the regression analysis between optical zone and postoperative spherical equivalent (SE)\n\nScatterplot of the regression analysis between optical zone and postoperative spherical equivalent (SE)\n\nTable 3Regression analysis between optical zones and postoperative SE in different preoperative refractive statusPreoperative SE (D)\nLower Third\n\nUpper Third\n\nParameter\n\n≤-4.00\n\n≥ -5.38\n\nP\nNo. of eyes193194Preoperative SE (D)Mean ± SD-3.18 ± 0.67-6.50 ± 0.89< 0.001Range-4.00 to -1.50-9.25 to -5.38Postoperative SE (D)Mean ± SD-0.17 ± 0.32-0.11 ± 0.49< 0.001\nr\n0.030.28Linear regression equationY= -0.07*X + 0.63Y = 0.48*X-3.03\nP\n0.702< 0.001SE = spherical equivalent; D = diopters\n\nRegression analysis between optical zones and postoperative SE in different preoperative refractive status\nSE = spherical equivalent; D = diopters", "This study designed to evaluate the refractive outcomes in eyes with small optical zone (range from 6.0 to 6.4 mm) and large optical zone (range from 6.5 to 6.8 mm) at 3 months after SMILE. The results showed that the larger optical zones induced less undercorrection, especially in patients with higher myopia.\nThe safety, efficacy, and predictability of SMILE in this study were comparable with most published results [5–8, 11, 18]. For safety, Kim et al. [6] reported 3.3% of eyes experienced a loss of one or more lines, 41% improved one line, and 7.2% improved two lines at 3 months postoperatively. In this study, only two eyes lost one line or more, but 38% improved one line and 27% improved two lines. Comparing efficacy, several studies [11, 15, 16] have reported that 84%, 92%, 94% of eyes had an UDVA of 20/20 or better at 3 months after surgery. In the current study, 97% of eyes had a postoperative distance UDVA of 20/20 or better. Regarding predictability, other studies [15, 16, 18] have reported a predictability of SMILE range from 70 to 100% within ± 0.50 D of target refraction, and 94–100% within ± 1.00 D. These results were similar to our findings of 90% eyes within ± 0.50 D and 100% within ± 1.00 D at the 3-month follow-up.\nPrevious studies [15–19] found that age, gender, eye (right/left), keratometry and preoperative spherical equivalent were the relevant factors influencing the refractive outcomes of SMILE surgery. To more accurately evaluate the influence of preoperative optical zone on SMILE surgery, the PSM analysis was used in this study. There were 78 pairs of eyes matched by 1:1 PSM analysis between the small and large optical zone groups. The differences of age, gender, eye (right/left), mean keratometry, and preoperative spherical equivalent were not significant between two groups (P > 0.05). However, we found that postoperative spherical equivalent was significantly different between two groups, and patients with larger optical zones had a trend towards less undercorrection (P < 0.05). To further verify the influence of optical zone on SMILE refractive outcomes, a univariate regression analysis between optical zone and postoperative spherical equivalent was conducted. The results showed that each millimeter larger optical zone resulted in 8.13% or 0.39D less undercorrection (P < 0.001).\nRegarding the effect of optical zone on laser in situ keratomileusis (LASIK) refractive outcomes, Moshirfar et al. [25] conducted a retrospective cohort study of 1332 eyes underwent LASIK at 12 months postoperatively. They indicated that the 6.0 mm optical zone was more myopic postoperatively compared to the 6.5 mm optical zone in moderate myopia group. In this study, the cohort were segregated into thirds based on level of preoperative spherical equivalent. Our results shown that the effect of optical zones on SMILE refractive outcomes was significantly different on the low and high degree of preoperative myopia (P < 0.001). Larger optical zone resulted in less undercorrection in the upper third with the highest myopia group (P < 0.001). Nevertheless, the eyes with larger optical zones did not have a tendency to less undercorrection in the lower third with the lowest myopia group (P = 0.702). These results indicate that larger optical zone may result in less undercorrection in the eyes with high preoperative myopia, but not evident in the eyes with low preoperative myopia. Therefore, we drew a conclusion that eyes with higher preoperative myopia were more influenced by the preoperative optical zones.\nThe visual quality of patients after SMILE surgery would be greatly influenced by the diameter of optical zone. Although better visual quality is obtained in dark, a larger optical zone requires more corneal tissue for a given spherical equivalent refraction correction [22]. In the case of thin cornea or high myopia, a refractive surgeon would choose a relatively smaller optical zone to avoid the postoperative complication such as corneal ectasia [23, 24]. However, we found that a smaller optical zone resulted in more undercorrection, especially in eyes with high myopia in this study.\nA limitation of the current study is the retrospective nature. Further, a concise follow-up period may not give a definite result while evaluating the effect of optical zone diameter on SMILE refractive outcome. It will be effective to verify the treatment method in long-term follow-ups and problematic patients. In addition, the eye sample size is relatively small in this study. Future studies with a larger sample size are essential to stablish a reliable recommendation for nomogram adjustment in patients with high myopia. Another limitation is that we only make a univariate regression analysis between optical zone and postoperative spherical equivalent. A multivariate regression analysis including more confounding factors as covariates would be beneficial to improve the accuracy of these outcomes in a future study. Moreover, the effect of optical zone on visual quality in the current study could not be assessed owing to a lack of related data. In future research, it would be of clinical importance to compare the SMILE visual quality based on preoperative optical zone diameter. In the end, the biggest shortcoming of this study is that we didn’t elucidate whether the effect of optical zone on SMILE refractive outcome was confounded by the residual stromal bed thickness. A randomized comparative study between patients with different optical zone may be effective to judge whether optical zone influence the refractive outcome independently or is confounded by the residual stromal bed thickness.\nIn conclusion, SMILE is a safe, effective, and predictable refractive surgery for the correction of myopia and myopic astigmatism. Our study demonstrated that the postoperative refractive outcomes of SMILE would be affected by preoperative optical zone, and the eyes with a larger optical zone have a tendency to less undercorrection. The influence of optical zones on SMILE refractive outcomes is significant in eyes with high degree of myopia, but not significant in eyes with low myopia." ]
[ null, null, null, null, null, null, "results", null, null, null, null, "discussion" ]
[ "Small incision lenticule extraction", "Optical zones", "Refractive outcomes", "Myopia" ]
Efficacy and safety of combined targeted therapy and immunotherapy versus targeted monotherapy in unresectable hepatocellular carcinoma: a systematic review and meta-analysis.
36271374
Cancer therapy has evolved from non-specific cytotoxic agents to a selective, mechanism-based approach that includes targeted agents and immunotherapy. Although the response to targeted therapies for unresectable hepatocellular carcinoma (HCC) is acceptable with the improved survival, the high tumor recurrence rate and drug-related side effects continue to be problematic. Given that immune checkpoint inhibitor alone are not robust enough to improve survival in unresectable HCC, growing evidence supports the combination of targeted therapy and immunotherapy with synergistic effect.
BACKGROUND
Online databases including PubMed, EMBASE, Cochrane Library, and Web of Science were searched for the studies that compared targeted monotherapy with the combination therapy of targeted drug and checkpoint inhibitors in unresectable HCC patients. Eligibility criteria were the presence of at least one measurable lesion as defined by the Response Evaluation Criteria in Solid Tumors (version 1.1) for unresectable HCC patients, an Eastern Cooperative Oncology Group performance status of 0-2, and a Child-Pugh score ≤ 7. Outcome measurements include overall survival (OS), progression-free survival (PFS), and treatment-related adverse event (TRAE).
METHODS
Three phase II/III randomized controlled trials were included in this study. The pooled results showed that combination therapy significantly improved survival than targeted monotherapy, in terms of OS (hazard ratio (HR) = 0.67; 95% confidence interval [CI]: 0.50-0.91) and PFS (HR = 0.58; 95% CI: 0.51-0.67), respectively. In the incidence of grade 3-5 TRAEs, the combination therapy was significantly higher than targeted monotherapy (odds ratio = 1.98; 95% CI: 1.13-3.48).
RESULTS
For unresectable HCC, combined targeted drug and immunotherapy significantly improved survival compared with targeted monotherapy. However, the incidences of AEs of combinational therapy were higher than targeted monotherapy.
CONCLUSION
[ "Humans", "Carcinoma, Hepatocellular", "Liver Neoplasms", "Immune Checkpoint Inhibitors", "Neoplasm Recurrence, Local", "Immunotherapy", "Immunologic Factors", "Cytotoxins", "Randomized Controlled Trials as Topic", "Clinical Trials, Phase II as Topic" ]
9587661
Introduction
Cancer is one of the important global health issues, which claims the live of approximately one in six individuals. In 2020, an estimated 19.3 million new cases of cancer and nearly 10 million cancer-related deaths were reported worldwide [1, 2]. For many decades, there have been various options of cancer treatment for patients, including surgery, radiation therapy, and chemotherapy, either alone or in combination. In the last three decades, medical research has advanced substantially in the molecular understanding of cancer biology. From relatively non-specific cytotoxic agents to a specifically selective, mechanism-based approach, including targeted agents and cancer immunotherapy, cancer therapy has evolved [1, 3]. This approach has been used for a wide range of solid tumors including hepatocellular carcinoma (HCC) [4–6]. The mechanistic action of targeted therapy is by interfering with specific molecules, which blocks the growth and spread of cancer. Although the initial response to targeted therapies was acceptable with the improved survival in a proportion of patients, obstacles exist with the high rate of tumor recurrence and drug-related side effects. Targeted therapies remain common in treating patients with unresectable and advanced HCC [3, 7, 8], so the need for more effective and safer alternative therapies is urgently warranted. Immunotherapy aims to stimulate a host immune response that destructs tumor and enhances antitumor responses to inhibit tumor growth or kill cancer cells [1, 9]. In patients with unresectable HCC, monotherapy with immune checkpoint inhibitors was not robust enough to improve overall survival (OS) and/or progression-free survival (PFS) [10, 11]. However, there is growing evidence that the combination of targeted therapy and immunotherapy has the potential to provide synergistic and sustained effects for cancer management [12, 13] and for unresectable HCC [14, 15]. One network meta-analysis compared the efficacy and safety of all first-line systemic therapy in patients with unresectable HCC, and one of the results showed that checkpoint inhibitor plus targeted therapy provided better outcomes of OS and PFS than sorafenib [16]. Therefore, this systematic review and meta-analysis are aimed to evaluate the efficacy and safety of the combination therapy versus targeted monotherapy in patients with unresectable/advanced HCC.
null
null
Results
[SUBTITLE] Study selection and characteristics of the included studies [SUBSECTION] The initial search identified 732 articles in online databases. After the screening process, duplicate and irrelevant studies were excluded. Finally, three articles were included in this meta-analysis (Fig. 1) [14, 15, 17]. Study designs for all studies were phase II/III RCTs. The studies were all published from 2020 to 2022. A total of 1,721 patients were included in the meta-analysis. The mean age was approximately 61 year, with a range from 53 to 66 year (Table 1). Fig. 1PRISMA flow diagram showing screening and selection process. Reference: Page, M. J., McKenzie, J. E., Bossuyt, P. M., Boutron, I., Hoffmann, T. C., Mulrow, C. D., et al. (2021). The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. International Journal of Surgery, 88, 105,906 PRISMA flow diagram showing screening and selection process. Reference: Page, M. J., McKenzie, J. E., Bossuyt, P. M., Boutron, I., Hoffmann, T. C., Mulrow, C. D., et al. (2021). The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. International Journal of Surgery, 88, 105,906 Table 1Baseline characteristics of the included studiesTypes of CancerStudy NameStudy phase/ designArmPtsMedian AgeMale (%)Median OS (m)HR, 95%CIMedian PFS (m)HR, 95%CITRAEs of grade 3–5NCT number.HCCFinn et al.,2020(IMbrave150)III/RCTAtezolizumab + Bevacizumab33664277(82)NE0.58,0.42–0.796.80.59,0.47–0.76201NCT03434379Sorafenib16566137(83)13.24.395Ren et al.,2021(ORIENT-32)III/RCTSintilimab + Bevacizumab biosimilar38053334(88)NR0.57,0.43–0.754.60.56, 0.46–0.70231NCT03794440Sorafenib19154171(90)10.42.868Kelley et al.,2022(COSMIC-312)III/RCTAtezolizumab +Cabozantinib43264360(83)15.40.900.69–1.186.80.630.44–0.91236NCT03755791Sorafenib21764186(86)15.54.268HCC = hepatocellular carcinoma; RCT = randomized control trial; OS = overall survival; CI = confidence interval; PFS = progression-free survival; HR = hazard ratio; AEs = adverse events; NR = not reached; NE = not estimable Baseline characteristics of the included studies Finn et al.,2020 (IMbrave150) 0.58, 0.42–0.79 0.59, 0.47–0.76 Ren et al.,2021 (ORIENT-32) 0.57, 0.43–0.75 0.56, 0.46–0.70 Kelley et al.,2022 (COSMIC-312) Atezolizumab + Cabozantinib 0.90 0.69–1.18 0.63 0.44–0.91 HCC = hepatocellular carcinoma; RCT = randomized control trial; OS = overall survival; CI = confidence interval; PFS = progression-free survival; HR = hazard ratio; AEs = adverse events; NR = not reached; NE = not estimable The initial search identified 732 articles in online databases. After the screening process, duplicate and irrelevant studies were excluded. Finally, three articles were included in this meta-analysis (Fig. 1) [14, 15, 17]. Study designs for all studies were phase II/III RCTs. The studies were all published from 2020 to 2022. A total of 1,721 patients were included in the meta-analysis. The mean age was approximately 61 year, with a range from 53 to 66 year (Table 1). Fig. 1PRISMA flow diagram showing screening and selection process. Reference: Page, M. J., McKenzie, J. E., Bossuyt, P. M., Boutron, I., Hoffmann, T. C., Mulrow, C. D., et al. (2021). The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. International Journal of Surgery, 88, 105,906 PRISMA flow diagram showing screening and selection process. Reference: Page, M. J., McKenzie, J. E., Bossuyt, P. M., Boutron, I., Hoffmann, T. C., Mulrow, C. D., et al. (2021). The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. International Journal of Surgery, 88, 105,906 Table 1Baseline characteristics of the included studiesTypes of CancerStudy NameStudy phase/ designArmPtsMedian AgeMale (%)Median OS (m)HR, 95%CIMedian PFS (m)HR, 95%CITRAEs of grade 3–5NCT number.HCCFinn et al.,2020(IMbrave150)III/RCTAtezolizumab + Bevacizumab33664277(82)NE0.58,0.42–0.796.80.59,0.47–0.76201NCT03434379Sorafenib16566137(83)13.24.395Ren et al.,2021(ORIENT-32)III/RCTSintilimab + Bevacizumab biosimilar38053334(88)NR0.57,0.43–0.754.60.56, 0.46–0.70231NCT03794440Sorafenib19154171(90)10.42.868Kelley et al.,2022(COSMIC-312)III/RCTAtezolizumab +Cabozantinib43264360(83)15.40.900.69–1.186.80.630.44–0.91236NCT03755791Sorafenib21764186(86)15.54.268HCC = hepatocellular carcinoma; RCT = randomized control trial; OS = overall survival; CI = confidence interval; PFS = progression-free survival; HR = hazard ratio; AEs = adverse events; NR = not reached; NE = not estimable Baseline characteristics of the included studies Finn et al.,2020 (IMbrave150) 0.58, 0.42–0.79 0.59, 0.47–0.76 Ren et al.,2021 (ORIENT-32) 0.57, 0.43–0.75 0.56, 0.46–0.70 Kelley et al.,2022 (COSMIC-312) Atezolizumab + Cabozantinib 0.90 0.69–1.18 0.63 0.44–0.91 HCC = hepatocellular carcinoma; RCT = randomized control trial; OS = overall survival; CI = confidence interval; PFS = progression-free survival; HR = hazard ratio; AEs = adverse events; NR = not reached; NE = not estimable [SUBTITLE] Risk of bias [SUBSECTION] Four domains of the included studies were found to have a low risk of bias (random sequence generation, allocation concealment, incomplete outcome data, and selective outcome reporting). All three studies rated the high risk of bias for blinding participants and personnel blinding bias. One study was rated as high risk for the blinding of outcome assessment (Fig. 2). Fig. 2Risk of bias graph: with review of authors’ judgements about each risk of bias item presented as percentages across all included studies Risk of bias graph: with review of authors’ judgements about each risk of bias item presented as percentages across all included studies Four domains of the included studies were found to have a low risk of bias (random sequence generation, allocation concealment, incomplete outcome data, and selective outcome reporting). All three studies rated the high risk of bias for blinding participants and personnel blinding bias. One study was rated as high risk for the blinding of outcome assessment (Fig. 2). Fig. 2Risk of bias graph: with review of authors’ judgements about each risk of bias item presented as percentages across all included studies Risk of bias graph: with review of authors’ judgements about each risk of bias item presented as percentages across all included studies [SUBTITLE] Major outcomes: overall survival and progression-free survival [SUBSECTION] OS and PFS data were available for all three trials. The pooled results showed that patients receiving combination therapy with targeted drug and immunotherapy had significantly better pooled OS than targeted monotherapy (HR = 0.67; 95% CI: 0.50–0.91) (Fig. 3). Fig. 3Forest plot of hazard ratio of overall survival using a random-effects model of hepatocellular carcinoma Forest plot of hazard ratio of overall survival using a random-effects model of hepatocellular carcinoma For PFS, patients receiving combination therapy had significantly better pooled PFS than targeted monotherapy (HR = 0.58; 95% CI: 0.51–0.67) (Fig. 4). Fig. 4Forest plot of hazard ratio of progression-free survival using a random-effects model of hepatocellular carcinoma Forest plot of hazard ratio of progression-free survival using a random-effects model of hepatocellular carcinoma OS and PFS data were available for all three trials. The pooled results showed that patients receiving combination therapy with targeted drug and immunotherapy had significantly better pooled OS than targeted monotherapy (HR = 0.67; 95% CI: 0.50–0.91) (Fig. 3). Fig. 3Forest plot of hazard ratio of overall survival using a random-effects model of hepatocellular carcinoma Forest plot of hazard ratio of overall survival using a random-effects model of hepatocellular carcinoma For PFS, patients receiving combination therapy had significantly better pooled PFS than targeted monotherapy (HR = 0.58; 95% CI: 0.51–0.67) (Fig. 4). Fig. 4Forest plot of hazard ratio of progression-free survival using a random-effects model of hepatocellular carcinoma Forest plot of hazard ratio of progression-free survival using a random-effects model of hepatocellular carcinoma [SUBTITLE] Treatment-related adverse events [SUBSECTION] All trials reported the incidences of grade 3–5 TRAEs. The pooled results showed that the combined therapy was associated with a significantly higher incidence of grade 3–5 TRAEs compared with targeted therapy alone (OR = 1.98; 95% CI: 1.13–3.48) (Fig. 5). Fig. 5Forest plot of odds ratios of treatment-related adverse events using a random-effects model of hepatocellular carcinoma Forest plot of odds ratios of treatment-related adverse events using a random-effects model of hepatocellular carcinoma All trials reported the incidences of grade 3–5 TRAEs. The pooled results showed that the combined therapy was associated with a significantly higher incidence of grade 3–5 TRAEs compared with targeted therapy alone (OR = 1.98; 95% CI: 1.13–3.48) (Fig. 5). Fig. 5Forest plot of odds ratios of treatment-related adverse events using a random-effects model of hepatocellular carcinoma Forest plot of odds ratios of treatment-related adverse events using a random-effects model of hepatocellular carcinoma
Conclusion
Our meta-analysis concluded that compared with targeted monotherapy, targeted therapy in combination with PD-1/PD-L1 inhibitors provided the survival benefits in patients with unresectable HCC. The patients receiving combination therapy had significantly higher incidences of grade 3–5 adverse effects.
[ "Methods", "Data source and literature search strategy", "Study selection", "Data extraction and quality assessment", "Statistical analysis", "Study selection and characteristics of the included studies", "Risk of bias", "Major outcomes: overall survival and progression-free survival", "Treatment-related adverse events" ]
[ "[SUBTITLE] Data source and literature search strategy [SUBSECTION] The search databases, including PubMed, EMBASE, Cochrane Library, and Web of Science, were searched for eligible studies from inception to July 2022. The search terms used to define the therapy included (“molecular targeted therapy” OR “targeted therapy”) AND (“immunotherapy” OR “immune checkpoint inhibitors” OR “programmed death 1 receptor” OR “programmed death 1 ligand 1” OR “PD 1 Inhibitors” OR “PD L1 Inhibitors”). The terms used to define the disease included “liver cell carcinoma” OR “advanced hepatocellular carcinoma” OR “hepatocellular carcinoma cell line.” In addition, we also checked the reference lists of all relevant articles to identify additional studies.\nThe search databases, including PubMed, EMBASE, Cochrane Library, and Web of Science, were searched for eligible studies from inception to July 2022. The search terms used to define the therapy included (“molecular targeted therapy” OR “targeted therapy”) AND (“immunotherapy” OR “immune checkpoint inhibitors” OR “programmed death 1 receptor” OR “programmed death 1 ligand 1” OR “PD 1 Inhibitors” OR “PD L1 Inhibitors”). The terms used to define the disease included “liver cell carcinoma” OR “advanced hepatocellular carcinoma” OR “hepatocellular carcinoma cell line.” In addition, we also checked the reference lists of all relevant articles to identify additional studies.\n[SUBTITLE] Study selection [SUBSECTION] The inclusion criteria were as follows: [1] prospective study and randomized controlled trials (RCTs); [2] study involving patients with advanced/unresectable HCC; [3] intervention and comparison with targeted therapy in combination with PD-1/PD-L1 inhibitors compared with targeted monotherapy; [4] the presence of at least one measurable lesion as defined by the Response Evaluation Criteria in Solid Tumors (version 1.1); 5) Eastern Cooperative Oncology Group performance status of 0–2 in HCC patients; [6] Child–Pugh score ≤ 7; [7] at least one of the following clinical outcomes reported—OS, PFS, and the rate of any grade adverse events (AEs); and [8] studies published in English. The exclusion criteria were as follows: review articles, case reports, and conference abstracts.\nThe inclusion criteria were as follows: [1] prospective study and randomized controlled trials (RCTs); [2] study involving patients with advanced/unresectable HCC; [3] intervention and comparison with targeted therapy in combination with PD-1/PD-L1 inhibitors compared with targeted monotherapy; [4] the presence of at least one measurable lesion as defined by the Response Evaluation Criteria in Solid Tumors (version 1.1); 5) Eastern Cooperative Oncology Group performance status of 0–2 in HCC patients; [6] Child–Pugh score ≤ 7; [7] at least one of the following clinical outcomes reported—OS, PFS, and the rate of any grade adverse events (AEs); and [8] studies published in English. The exclusion criteria were as follows: review articles, case reports, and conference abstracts.\n[SUBTITLE] Data extraction and quality assessment [SUBSECTION] For each eligible study, the following information was extracted: article title, first author, publication year, trial phase, study design, applied agents, combination therapy, sample size, rate of OS, rate of PFS, median time to progression, AEs, and national clinical trial identification number. The risk of bias for individual studies was assessed at the study level based on the Cochrane Collaboration’s tool for randomized trials, which include the following domains: random sequence generation, allocation concealment, blinding, incomplete outcome data, and selective outcome reporting [16]. The evaluation of the risk of bias was conducted by the Review Manager (RevMan, V.5.4.1, Nordic Cochrane Centre, Cochrane, Copenhagen, Denmark).\nFor each eligible study, the following information was extracted: article title, first author, publication year, trial phase, study design, applied agents, combination therapy, sample size, rate of OS, rate of PFS, median time to progression, AEs, and national clinical trial identification number. The risk of bias for individual studies was assessed at the study level based on the Cochrane Collaboration’s tool for randomized trials, which include the following domains: random sequence generation, allocation concealment, blinding, incomplete outcome data, and selective outcome reporting [16]. The evaluation of the risk of bias was conducted by the Review Manager (RevMan, V.5.4.1, Nordic Cochrane Centre, Cochrane, Copenhagen, Denmark).\n[SUBTITLE] Statistical analysis [SUBSECTION] We calculated the pooled hazard ratio (HR) and 95% confidence interval (CI) for PFS and OS, as well as the pooled odds ratio (OR) and 95% CI for grade 3–5 TRAEs. The meta-analysis was conducted using the random-effects model under the assumption of significant heterogeneity. Heterogeneity among studies was quantified by I2 test, and I2 > 50% was considered substantial heterogeneity. p < 0.05 was considered statistically significant. The statistical analysis was conducted using Review Manager (RevMan5.4.1).\nWe calculated the pooled hazard ratio (HR) and 95% confidence interval (CI) for PFS and OS, as well as the pooled odds ratio (OR) and 95% CI for grade 3–5 TRAEs. The meta-analysis was conducted using the random-effects model under the assumption of significant heterogeneity. Heterogeneity among studies was quantified by I2 test, and I2 > 50% was considered substantial heterogeneity. p < 0.05 was considered statistically significant. The statistical analysis was conducted using Review Manager (RevMan5.4.1).", "The search databases, including PubMed, EMBASE, Cochrane Library, and Web of Science, were searched for eligible studies from inception to July 2022. The search terms used to define the therapy included (“molecular targeted therapy” OR “targeted therapy”) AND (“immunotherapy” OR “immune checkpoint inhibitors” OR “programmed death 1 receptor” OR “programmed death 1 ligand 1” OR “PD 1 Inhibitors” OR “PD L1 Inhibitors”). The terms used to define the disease included “liver cell carcinoma” OR “advanced hepatocellular carcinoma” OR “hepatocellular carcinoma cell line.” In addition, we also checked the reference lists of all relevant articles to identify additional studies.", "The inclusion criteria were as follows: [1] prospective study and randomized controlled trials (RCTs); [2] study involving patients with advanced/unresectable HCC; [3] intervention and comparison with targeted therapy in combination with PD-1/PD-L1 inhibitors compared with targeted monotherapy; [4] the presence of at least one measurable lesion as defined by the Response Evaluation Criteria in Solid Tumors (version 1.1); 5) Eastern Cooperative Oncology Group performance status of 0–2 in HCC patients; [6] Child–Pugh score ≤ 7; [7] at least one of the following clinical outcomes reported—OS, PFS, and the rate of any grade adverse events (AEs); and [8] studies published in English. The exclusion criteria were as follows: review articles, case reports, and conference abstracts.", "For each eligible study, the following information was extracted: article title, first author, publication year, trial phase, study design, applied agents, combination therapy, sample size, rate of OS, rate of PFS, median time to progression, AEs, and national clinical trial identification number. The risk of bias for individual studies was assessed at the study level based on the Cochrane Collaboration’s tool for randomized trials, which include the following domains: random sequence generation, allocation concealment, blinding, incomplete outcome data, and selective outcome reporting [16]. The evaluation of the risk of bias was conducted by the Review Manager (RevMan, V.5.4.1, Nordic Cochrane Centre, Cochrane, Copenhagen, Denmark).", "We calculated the pooled hazard ratio (HR) and 95% confidence interval (CI) for PFS and OS, as well as the pooled odds ratio (OR) and 95% CI for grade 3–5 TRAEs. The meta-analysis was conducted using the random-effects model under the assumption of significant heterogeneity. Heterogeneity among studies was quantified by I2 test, and I2 > 50% was considered substantial heterogeneity. p < 0.05 was considered statistically significant. The statistical analysis was conducted using Review Manager (RevMan5.4.1).", "The initial search identified 732 articles in online databases. After the screening process, duplicate and irrelevant studies were excluded. Finally, three articles were included in this meta-analysis (Fig. 1) [14, 15, 17]. Study designs for all studies were phase II/III RCTs. The studies were all published from 2020 to 2022. A total of 1,721 patients were included in the meta-analysis. The mean age was approximately 61 year, with a range from 53 to 66 year (Table 1).\n\nFig. 1PRISMA flow diagram showing screening and selection process. Reference: Page, M. J., McKenzie, J. E., Bossuyt, P. M., Boutron, I., Hoffmann, T. C., Mulrow, C. D., et al. (2021). The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. International Journal of Surgery, 88, 105,906\n\nPRISMA flow diagram showing screening and selection process. Reference: Page, M. J., McKenzie, J. E., Bossuyt, P. M., Boutron, I., Hoffmann, T. C., Mulrow, C. D., et al. (2021). The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. International Journal of Surgery, 88, 105,906\n\nTable 1Baseline characteristics of the included studiesTypes of CancerStudy NameStudy phase/ designArmPtsMedian AgeMale (%)Median OS (m)HR, 95%CIMedian PFS (m)HR, 95%CITRAEs of grade 3–5NCT number.HCCFinn et al.,2020(IMbrave150)III/RCTAtezolizumab + Bevacizumab33664277(82)NE0.58,0.42–0.796.80.59,0.47–0.76201NCT03434379Sorafenib16566137(83)13.24.395Ren et al.,2021(ORIENT-32)III/RCTSintilimab + Bevacizumab biosimilar38053334(88)NR0.57,0.43–0.754.60.56, 0.46–0.70231NCT03794440Sorafenib19154171(90)10.42.868Kelley et al.,2022(COSMIC-312)III/RCTAtezolizumab +Cabozantinib43264360(83)15.40.900.69–1.186.80.630.44–0.91236NCT03755791Sorafenib21764186(86)15.54.268HCC = hepatocellular carcinoma; RCT = randomized control trial; OS = overall survival; CI = confidence interval; PFS = progression-free survival; HR = hazard ratio; AEs = adverse events; NR = not reached; NE = not estimable\n\nBaseline characteristics of the included studies\nFinn et al.,2020\n(IMbrave150)\n0.58,\n0.42–0.79\n0.59,\n0.47–0.76\nRen et al.,2021\n(ORIENT-32)\n0.57,\n0.43–0.75\n0.56, \n0.46–0.70\nKelley et al.,2022\n(COSMIC-312)\nAtezolizumab +\nCabozantinib\n0.90\n0.69–1.18\n0.63\n0.44–0.91\nHCC = hepatocellular carcinoma; RCT = randomized control trial; OS = overall survival; CI = confidence interval; PFS = progression-free survival; HR = hazard ratio; AEs = adverse events; NR = not reached; NE = not estimable", "Four domains of the included studies were found to have a low risk of bias (random sequence generation, allocation concealment, incomplete outcome data, and selective outcome reporting). All three studies rated the high risk of bias for blinding participants and personnel blinding bias. One study was rated as high risk for the blinding of outcome assessment (Fig. 2).\n\nFig. 2Risk of bias graph: with review of authors’ judgements about each risk of bias item presented as percentages across all included studies\n\nRisk of bias graph: with review of authors’ judgements about each risk of bias item presented as percentages across all included studies", "OS and PFS data were available for all three trials. The pooled results showed that patients receiving combination therapy with targeted drug and immunotherapy had significantly better pooled OS than targeted monotherapy (HR = 0.67; 95% CI: 0.50–0.91) (Fig. 3).\n\nFig. 3Forest plot of hazard ratio of overall survival using a random-effects model of hepatocellular carcinoma\n\nForest plot of hazard ratio of overall survival using a random-effects model of hepatocellular carcinoma\nFor PFS, patients receiving combination therapy had significantly better pooled PFS than targeted monotherapy (HR = 0.58; 95% CI: 0.51–0.67) (Fig. 4).\n\nFig. 4Forest plot of hazard ratio of progression-free survival using a random-effects model of hepatocellular carcinoma\n\nForest plot of hazard ratio of progression-free survival using a random-effects model of hepatocellular carcinoma", "All trials reported the incidences of grade 3–5 TRAEs. The pooled results showed that the combined therapy was associated with a significantly higher incidence of grade 3–5 TRAEs compared with targeted therapy alone (OR = 1.98; 95% CI: 1.13–3.48) (Fig. 5).\n\nFig. 5Forest plot of odds ratios of treatment-related adverse events using a random-effects model of hepatocellular carcinoma\n\nForest plot of odds ratios of treatment-related adverse events using a random-effects model of hepatocellular carcinoma" ]
[ null, null, null, null, null, null, null, null, null ]
[ "Introduction", "Methods", "Data source and literature search strategy", "Study selection", "Data extraction and quality assessment", "Statistical analysis", "Results", "Study selection and characteristics of the included studies", "Risk of bias", "Major outcomes: overall survival and progression-free survival", "Treatment-related adverse events", "Discussion", "Conclusion" ]
[ "Cancer is one of the important global health issues, which claims the live of approximately one in six individuals. In 2020, an estimated 19.3 million new cases of cancer and nearly 10 million cancer-related deaths were reported worldwide [1, 2].\nFor many decades, there have been various options of cancer treatment for patients, including surgery, radiation therapy, and chemotherapy, either alone or in combination. In the last three decades, medical research has advanced substantially in the molecular understanding of cancer biology. From relatively non-specific cytotoxic agents to a specifically selective, mechanism-based approach, including targeted agents and cancer immunotherapy, cancer therapy has evolved [1, 3]. This approach has been used for a wide range of solid tumors including hepatocellular carcinoma (HCC) [4–6].\nThe mechanistic action of targeted therapy is by interfering with specific molecules, which blocks the growth and spread of cancer. Although the initial response to targeted therapies was acceptable with the improved survival in a proportion of patients, obstacles exist with the high rate of tumor recurrence and drug-related side effects. Targeted therapies remain common in treating patients with unresectable and advanced HCC [3, 7, 8], so the need for more effective and safer alternative therapies is urgently warranted.\nImmunotherapy aims to stimulate a host immune response that destructs tumor and enhances antitumor responses to inhibit tumor growth or kill cancer cells [1, 9]. In patients with unresectable HCC, monotherapy with immune checkpoint inhibitors was not robust enough to improve overall survival (OS) and/or progression-free survival (PFS) [10, 11]. However, there is growing evidence that the combination of targeted therapy and immunotherapy has the potential to provide synergistic and sustained effects for cancer management [12, 13] and for unresectable HCC [14, 15]. One network meta-analysis compared the efficacy and safety of all first-line systemic therapy in patients with unresectable HCC, and one of the results showed that checkpoint inhibitor plus targeted therapy provided better outcomes of OS and PFS than sorafenib [16]. Therefore, this systematic review and meta-analysis are aimed to evaluate the efficacy and safety of the combination therapy versus targeted monotherapy in patients with unresectable/advanced HCC.", "[SUBTITLE] Data source and literature search strategy [SUBSECTION] The search databases, including PubMed, EMBASE, Cochrane Library, and Web of Science, were searched for eligible studies from inception to July 2022. The search terms used to define the therapy included (“molecular targeted therapy” OR “targeted therapy”) AND (“immunotherapy” OR “immune checkpoint inhibitors” OR “programmed death 1 receptor” OR “programmed death 1 ligand 1” OR “PD 1 Inhibitors” OR “PD L1 Inhibitors”). The terms used to define the disease included “liver cell carcinoma” OR “advanced hepatocellular carcinoma” OR “hepatocellular carcinoma cell line.” In addition, we also checked the reference lists of all relevant articles to identify additional studies.\nThe search databases, including PubMed, EMBASE, Cochrane Library, and Web of Science, were searched for eligible studies from inception to July 2022. The search terms used to define the therapy included (“molecular targeted therapy” OR “targeted therapy”) AND (“immunotherapy” OR “immune checkpoint inhibitors” OR “programmed death 1 receptor” OR “programmed death 1 ligand 1” OR “PD 1 Inhibitors” OR “PD L1 Inhibitors”). The terms used to define the disease included “liver cell carcinoma” OR “advanced hepatocellular carcinoma” OR “hepatocellular carcinoma cell line.” In addition, we also checked the reference lists of all relevant articles to identify additional studies.\n[SUBTITLE] Study selection [SUBSECTION] The inclusion criteria were as follows: [1] prospective study and randomized controlled trials (RCTs); [2] study involving patients with advanced/unresectable HCC; [3] intervention and comparison with targeted therapy in combination with PD-1/PD-L1 inhibitors compared with targeted monotherapy; [4] the presence of at least one measurable lesion as defined by the Response Evaluation Criteria in Solid Tumors (version 1.1); 5) Eastern Cooperative Oncology Group performance status of 0–2 in HCC patients; [6] Child–Pugh score ≤ 7; [7] at least one of the following clinical outcomes reported—OS, PFS, and the rate of any grade adverse events (AEs); and [8] studies published in English. The exclusion criteria were as follows: review articles, case reports, and conference abstracts.\nThe inclusion criteria were as follows: [1] prospective study and randomized controlled trials (RCTs); [2] study involving patients with advanced/unresectable HCC; [3] intervention and comparison with targeted therapy in combination with PD-1/PD-L1 inhibitors compared with targeted monotherapy; [4] the presence of at least one measurable lesion as defined by the Response Evaluation Criteria in Solid Tumors (version 1.1); 5) Eastern Cooperative Oncology Group performance status of 0–2 in HCC patients; [6] Child–Pugh score ≤ 7; [7] at least one of the following clinical outcomes reported—OS, PFS, and the rate of any grade adverse events (AEs); and [8] studies published in English. The exclusion criteria were as follows: review articles, case reports, and conference abstracts.\n[SUBTITLE] Data extraction and quality assessment [SUBSECTION] For each eligible study, the following information was extracted: article title, first author, publication year, trial phase, study design, applied agents, combination therapy, sample size, rate of OS, rate of PFS, median time to progression, AEs, and national clinical trial identification number. The risk of bias for individual studies was assessed at the study level based on the Cochrane Collaboration’s tool for randomized trials, which include the following domains: random sequence generation, allocation concealment, blinding, incomplete outcome data, and selective outcome reporting [16]. The evaluation of the risk of bias was conducted by the Review Manager (RevMan, V.5.4.1, Nordic Cochrane Centre, Cochrane, Copenhagen, Denmark).\nFor each eligible study, the following information was extracted: article title, first author, publication year, trial phase, study design, applied agents, combination therapy, sample size, rate of OS, rate of PFS, median time to progression, AEs, and national clinical trial identification number. The risk of bias for individual studies was assessed at the study level based on the Cochrane Collaboration’s tool for randomized trials, which include the following domains: random sequence generation, allocation concealment, blinding, incomplete outcome data, and selective outcome reporting [16]. The evaluation of the risk of bias was conducted by the Review Manager (RevMan, V.5.4.1, Nordic Cochrane Centre, Cochrane, Copenhagen, Denmark).\n[SUBTITLE] Statistical analysis [SUBSECTION] We calculated the pooled hazard ratio (HR) and 95% confidence interval (CI) for PFS and OS, as well as the pooled odds ratio (OR) and 95% CI for grade 3–5 TRAEs. The meta-analysis was conducted using the random-effects model under the assumption of significant heterogeneity. Heterogeneity among studies was quantified by I2 test, and I2 > 50% was considered substantial heterogeneity. p < 0.05 was considered statistically significant. The statistical analysis was conducted using Review Manager (RevMan5.4.1).\nWe calculated the pooled hazard ratio (HR) and 95% confidence interval (CI) for PFS and OS, as well as the pooled odds ratio (OR) and 95% CI for grade 3–5 TRAEs. The meta-analysis was conducted using the random-effects model under the assumption of significant heterogeneity. Heterogeneity among studies was quantified by I2 test, and I2 > 50% was considered substantial heterogeneity. p < 0.05 was considered statistically significant. The statistical analysis was conducted using Review Manager (RevMan5.4.1).", "The search databases, including PubMed, EMBASE, Cochrane Library, and Web of Science, were searched for eligible studies from inception to July 2022. The search terms used to define the therapy included (“molecular targeted therapy” OR “targeted therapy”) AND (“immunotherapy” OR “immune checkpoint inhibitors” OR “programmed death 1 receptor” OR “programmed death 1 ligand 1” OR “PD 1 Inhibitors” OR “PD L1 Inhibitors”). The terms used to define the disease included “liver cell carcinoma” OR “advanced hepatocellular carcinoma” OR “hepatocellular carcinoma cell line.” In addition, we also checked the reference lists of all relevant articles to identify additional studies.", "The inclusion criteria were as follows: [1] prospective study and randomized controlled trials (RCTs); [2] study involving patients with advanced/unresectable HCC; [3] intervention and comparison with targeted therapy in combination with PD-1/PD-L1 inhibitors compared with targeted monotherapy; [4] the presence of at least one measurable lesion as defined by the Response Evaluation Criteria in Solid Tumors (version 1.1); 5) Eastern Cooperative Oncology Group performance status of 0–2 in HCC patients; [6] Child–Pugh score ≤ 7; [7] at least one of the following clinical outcomes reported—OS, PFS, and the rate of any grade adverse events (AEs); and [8] studies published in English. The exclusion criteria were as follows: review articles, case reports, and conference abstracts.", "For each eligible study, the following information was extracted: article title, first author, publication year, trial phase, study design, applied agents, combination therapy, sample size, rate of OS, rate of PFS, median time to progression, AEs, and national clinical trial identification number. The risk of bias for individual studies was assessed at the study level based on the Cochrane Collaboration’s tool for randomized trials, which include the following domains: random sequence generation, allocation concealment, blinding, incomplete outcome data, and selective outcome reporting [16]. The evaluation of the risk of bias was conducted by the Review Manager (RevMan, V.5.4.1, Nordic Cochrane Centre, Cochrane, Copenhagen, Denmark).", "We calculated the pooled hazard ratio (HR) and 95% confidence interval (CI) for PFS and OS, as well as the pooled odds ratio (OR) and 95% CI for grade 3–5 TRAEs. The meta-analysis was conducted using the random-effects model under the assumption of significant heterogeneity. Heterogeneity among studies was quantified by I2 test, and I2 > 50% was considered substantial heterogeneity. p < 0.05 was considered statistically significant. The statistical analysis was conducted using Review Manager (RevMan5.4.1).", "[SUBTITLE] Study selection and characteristics of the included studies [SUBSECTION] The initial search identified 732 articles in online databases. After the screening process, duplicate and irrelevant studies were excluded. Finally, three articles were included in this meta-analysis (Fig. 1) [14, 15, 17]. Study designs for all studies were phase II/III RCTs. The studies were all published from 2020 to 2022. A total of 1,721 patients were included in the meta-analysis. The mean age was approximately 61 year, with a range from 53 to 66 year (Table 1).\n\nFig. 1PRISMA flow diagram showing screening and selection process. Reference: Page, M. J., McKenzie, J. E., Bossuyt, P. M., Boutron, I., Hoffmann, T. C., Mulrow, C. D., et al. (2021). The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. International Journal of Surgery, 88, 105,906\n\nPRISMA flow diagram showing screening and selection process. Reference: Page, M. J., McKenzie, J. E., Bossuyt, P. M., Boutron, I., Hoffmann, T. C., Mulrow, C. D., et al. (2021). The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. International Journal of Surgery, 88, 105,906\n\nTable 1Baseline characteristics of the included studiesTypes of CancerStudy NameStudy phase/ designArmPtsMedian AgeMale (%)Median OS (m)HR, 95%CIMedian PFS (m)HR, 95%CITRAEs of grade 3–5NCT number.HCCFinn et al.,2020(IMbrave150)III/RCTAtezolizumab + Bevacizumab33664277(82)NE0.58,0.42–0.796.80.59,0.47–0.76201NCT03434379Sorafenib16566137(83)13.24.395Ren et al.,2021(ORIENT-32)III/RCTSintilimab + Bevacizumab biosimilar38053334(88)NR0.57,0.43–0.754.60.56, 0.46–0.70231NCT03794440Sorafenib19154171(90)10.42.868Kelley et al.,2022(COSMIC-312)III/RCTAtezolizumab +Cabozantinib43264360(83)15.40.900.69–1.186.80.630.44–0.91236NCT03755791Sorafenib21764186(86)15.54.268HCC = hepatocellular carcinoma; RCT = randomized control trial; OS = overall survival; CI = confidence interval; PFS = progression-free survival; HR = hazard ratio; AEs = adverse events; NR = not reached; NE = not estimable\n\nBaseline characteristics of the included studies\nFinn et al.,2020\n(IMbrave150)\n0.58,\n0.42–0.79\n0.59,\n0.47–0.76\nRen et al.,2021\n(ORIENT-32)\n0.57,\n0.43–0.75\n0.56, \n0.46–0.70\nKelley et al.,2022\n(COSMIC-312)\nAtezolizumab +\nCabozantinib\n0.90\n0.69–1.18\n0.63\n0.44–0.91\nHCC = hepatocellular carcinoma; RCT = randomized control trial; OS = overall survival; CI = confidence interval; PFS = progression-free survival; HR = hazard ratio; AEs = adverse events; NR = not reached; NE = not estimable\nThe initial search identified 732 articles in online databases. After the screening process, duplicate and irrelevant studies were excluded. Finally, three articles were included in this meta-analysis (Fig. 1) [14, 15, 17]. Study designs for all studies were phase II/III RCTs. The studies were all published from 2020 to 2022. A total of 1,721 patients were included in the meta-analysis. The mean age was approximately 61 year, with a range from 53 to 66 year (Table 1).\n\nFig. 1PRISMA flow diagram showing screening and selection process. Reference: Page, M. J., McKenzie, J. E., Bossuyt, P. M., Boutron, I., Hoffmann, T. C., Mulrow, C. D., et al. (2021). The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. International Journal of Surgery, 88, 105,906\n\nPRISMA flow diagram showing screening and selection process. Reference: Page, M. J., McKenzie, J. E., Bossuyt, P. M., Boutron, I., Hoffmann, T. C., Mulrow, C. D., et al. (2021). The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. International Journal of Surgery, 88, 105,906\n\nTable 1Baseline characteristics of the included studiesTypes of CancerStudy NameStudy phase/ designArmPtsMedian AgeMale (%)Median OS (m)HR, 95%CIMedian PFS (m)HR, 95%CITRAEs of grade 3–5NCT number.HCCFinn et al.,2020(IMbrave150)III/RCTAtezolizumab + Bevacizumab33664277(82)NE0.58,0.42–0.796.80.59,0.47–0.76201NCT03434379Sorafenib16566137(83)13.24.395Ren et al.,2021(ORIENT-32)III/RCTSintilimab + Bevacizumab biosimilar38053334(88)NR0.57,0.43–0.754.60.56, 0.46–0.70231NCT03794440Sorafenib19154171(90)10.42.868Kelley et al.,2022(COSMIC-312)III/RCTAtezolizumab +Cabozantinib43264360(83)15.40.900.69–1.186.80.630.44–0.91236NCT03755791Sorafenib21764186(86)15.54.268HCC = hepatocellular carcinoma; RCT = randomized control trial; OS = overall survival; CI = confidence interval; PFS = progression-free survival; HR = hazard ratio; AEs = adverse events; NR = not reached; NE = not estimable\n\nBaseline characteristics of the included studies\nFinn et al.,2020\n(IMbrave150)\n0.58,\n0.42–0.79\n0.59,\n0.47–0.76\nRen et al.,2021\n(ORIENT-32)\n0.57,\n0.43–0.75\n0.56, \n0.46–0.70\nKelley et al.,2022\n(COSMIC-312)\nAtezolizumab +\nCabozantinib\n0.90\n0.69–1.18\n0.63\n0.44–0.91\nHCC = hepatocellular carcinoma; RCT = randomized control trial; OS = overall survival; CI = confidence interval; PFS = progression-free survival; HR = hazard ratio; AEs = adverse events; NR = not reached; NE = not estimable\n[SUBTITLE] Risk of bias [SUBSECTION] Four domains of the included studies were found to have a low risk of bias (random sequence generation, allocation concealment, incomplete outcome data, and selective outcome reporting). All three studies rated the high risk of bias for blinding participants and personnel blinding bias. One study was rated as high risk for the blinding of outcome assessment (Fig. 2).\n\nFig. 2Risk of bias graph: with review of authors’ judgements about each risk of bias item presented as percentages across all included studies\n\nRisk of bias graph: with review of authors’ judgements about each risk of bias item presented as percentages across all included studies\nFour domains of the included studies were found to have a low risk of bias (random sequence generation, allocation concealment, incomplete outcome data, and selective outcome reporting). All three studies rated the high risk of bias for blinding participants and personnel blinding bias. One study was rated as high risk for the blinding of outcome assessment (Fig. 2).\n\nFig. 2Risk of bias graph: with review of authors’ judgements about each risk of bias item presented as percentages across all included studies\n\nRisk of bias graph: with review of authors’ judgements about each risk of bias item presented as percentages across all included studies\n[SUBTITLE] Major outcomes: overall survival and progression-free survival [SUBSECTION] OS and PFS data were available for all three trials. The pooled results showed that patients receiving combination therapy with targeted drug and immunotherapy had significantly better pooled OS than targeted monotherapy (HR = 0.67; 95% CI: 0.50–0.91) (Fig. 3).\n\nFig. 3Forest plot of hazard ratio of overall survival using a random-effects model of hepatocellular carcinoma\n\nForest plot of hazard ratio of overall survival using a random-effects model of hepatocellular carcinoma\nFor PFS, patients receiving combination therapy had significantly better pooled PFS than targeted monotherapy (HR = 0.58; 95% CI: 0.51–0.67) (Fig. 4).\n\nFig. 4Forest plot of hazard ratio of progression-free survival using a random-effects model of hepatocellular carcinoma\n\nForest plot of hazard ratio of progression-free survival using a random-effects model of hepatocellular carcinoma\nOS and PFS data were available for all three trials. The pooled results showed that patients receiving combination therapy with targeted drug and immunotherapy had significantly better pooled OS than targeted monotherapy (HR = 0.67; 95% CI: 0.50–0.91) (Fig. 3).\n\nFig. 3Forest plot of hazard ratio of overall survival using a random-effects model of hepatocellular carcinoma\n\nForest plot of hazard ratio of overall survival using a random-effects model of hepatocellular carcinoma\nFor PFS, patients receiving combination therapy had significantly better pooled PFS than targeted monotherapy (HR = 0.58; 95% CI: 0.51–0.67) (Fig. 4).\n\nFig. 4Forest plot of hazard ratio of progression-free survival using a random-effects model of hepatocellular carcinoma\n\nForest plot of hazard ratio of progression-free survival using a random-effects model of hepatocellular carcinoma\n[SUBTITLE] Treatment-related adverse events [SUBSECTION] All trials reported the incidences of grade 3–5 TRAEs. The pooled results showed that the combined therapy was associated with a significantly higher incidence of grade 3–5 TRAEs compared with targeted therapy alone (OR = 1.98; 95% CI: 1.13–3.48) (Fig. 5).\n\nFig. 5Forest plot of odds ratios of treatment-related adverse events using a random-effects model of hepatocellular carcinoma\n\nForest plot of odds ratios of treatment-related adverse events using a random-effects model of hepatocellular carcinoma\nAll trials reported the incidences of grade 3–5 TRAEs. The pooled results showed that the combined therapy was associated with a significantly higher incidence of grade 3–5 TRAEs compared with targeted therapy alone (OR = 1.98; 95% CI: 1.13–3.48) (Fig. 5).\n\nFig. 5Forest plot of odds ratios of treatment-related adverse events using a random-effects model of hepatocellular carcinoma\n\nForest plot of odds ratios of treatment-related adverse events using a random-effects model of hepatocellular carcinoma", "The initial search identified 732 articles in online databases. After the screening process, duplicate and irrelevant studies were excluded. Finally, three articles were included in this meta-analysis (Fig. 1) [14, 15, 17]. Study designs for all studies were phase II/III RCTs. The studies were all published from 2020 to 2022. A total of 1,721 patients were included in the meta-analysis. The mean age was approximately 61 year, with a range from 53 to 66 year (Table 1).\n\nFig. 1PRISMA flow diagram showing screening and selection process. Reference: Page, M. J., McKenzie, J. E., Bossuyt, P. M., Boutron, I., Hoffmann, T. C., Mulrow, C. D., et al. (2021). The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. International Journal of Surgery, 88, 105,906\n\nPRISMA flow diagram showing screening and selection process. Reference: Page, M. J., McKenzie, J. E., Bossuyt, P. M., Boutron, I., Hoffmann, T. C., Mulrow, C. D., et al. (2021). The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. International Journal of Surgery, 88, 105,906\n\nTable 1Baseline characteristics of the included studiesTypes of CancerStudy NameStudy phase/ designArmPtsMedian AgeMale (%)Median OS (m)HR, 95%CIMedian PFS (m)HR, 95%CITRAEs of grade 3–5NCT number.HCCFinn et al.,2020(IMbrave150)III/RCTAtezolizumab + Bevacizumab33664277(82)NE0.58,0.42–0.796.80.59,0.47–0.76201NCT03434379Sorafenib16566137(83)13.24.395Ren et al.,2021(ORIENT-32)III/RCTSintilimab + Bevacizumab biosimilar38053334(88)NR0.57,0.43–0.754.60.56, 0.46–0.70231NCT03794440Sorafenib19154171(90)10.42.868Kelley et al.,2022(COSMIC-312)III/RCTAtezolizumab +Cabozantinib43264360(83)15.40.900.69–1.186.80.630.44–0.91236NCT03755791Sorafenib21764186(86)15.54.268HCC = hepatocellular carcinoma; RCT = randomized control trial; OS = overall survival; CI = confidence interval; PFS = progression-free survival; HR = hazard ratio; AEs = adverse events; NR = not reached; NE = not estimable\n\nBaseline characteristics of the included studies\nFinn et al.,2020\n(IMbrave150)\n0.58,\n0.42–0.79\n0.59,\n0.47–0.76\nRen et al.,2021\n(ORIENT-32)\n0.57,\n0.43–0.75\n0.56, \n0.46–0.70\nKelley et al.,2022\n(COSMIC-312)\nAtezolizumab +\nCabozantinib\n0.90\n0.69–1.18\n0.63\n0.44–0.91\nHCC = hepatocellular carcinoma; RCT = randomized control trial; OS = overall survival; CI = confidence interval; PFS = progression-free survival; HR = hazard ratio; AEs = adverse events; NR = not reached; NE = not estimable", "Four domains of the included studies were found to have a low risk of bias (random sequence generation, allocation concealment, incomplete outcome data, and selective outcome reporting). All three studies rated the high risk of bias for blinding participants and personnel blinding bias. One study was rated as high risk for the blinding of outcome assessment (Fig. 2).\n\nFig. 2Risk of bias graph: with review of authors’ judgements about each risk of bias item presented as percentages across all included studies\n\nRisk of bias graph: with review of authors’ judgements about each risk of bias item presented as percentages across all included studies", "OS and PFS data were available for all three trials. The pooled results showed that patients receiving combination therapy with targeted drug and immunotherapy had significantly better pooled OS than targeted monotherapy (HR = 0.67; 95% CI: 0.50–0.91) (Fig. 3).\n\nFig. 3Forest plot of hazard ratio of overall survival using a random-effects model of hepatocellular carcinoma\n\nForest plot of hazard ratio of overall survival using a random-effects model of hepatocellular carcinoma\nFor PFS, patients receiving combination therapy had significantly better pooled PFS than targeted monotherapy (HR = 0.58; 95% CI: 0.51–0.67) (Fig. 4).\n\nFig. 4Forest plot of hazard ratio of progression-free survival using a random-effects model of hepatocellular carcinoma\n\nForest plot of hazard ratio of progression-free survival using a random-effects model of hepatocellular carcinoma", "All trials reported the incidences of grade 3–5 TRAEs. The pooled results showed that the combined therapy was associated with a significantly higher incidence of grade 3–5 TRAEs compared with targeted therapy alone (OR = 1.98; 95% CI: 1.13–3.48) (Fig. 5).\n\nFig. 5Forest plot of odds ratios of treatment-related adverse events using a random-effects model of hepatocellular carcinoma\n\nForest plot of odds ratios of treatment-related adverse events using a random-effects model of hepatocellular carcinoma", "Cancer treatment in unresectable HCC and other malignant solid tumors has been rapidly changing, and the combinational therapy is increasingly favored. We performed a systematic review and meta-analysis to provide targeted therapy in combination with PD-1/PD-L1 inhibitors compared with targeted monotherapy. Our analyses explored clinically relevant efficacy outcomes, including OS, PFS, and grade 3–5 TRAEs. According to the results of the present study, targeted therapy in combination with PD-1/PD-L1 checkpoint inhibitors significantly improved the OS and PFS for unresectable HCC compared with targeted monotherapy showed that for unresectable HCC.\nThree phase III RCTs comparing targeted therapy in combination with PD-1/PD-L1 inhibitors with targeted monotherapy have been published so far. Finn et al. reported the combination therapy of atezolizumab (anti-PD-L1) and bevacizumab, which is a vascular endothelial growth factor A (VEGF-A) inhibitor, as compared to sorafenib targeting anti-angiogenesis multikinase receptor, with statistically significant and clinically meaningful improvement in both OS and PFS in the treatment of unresectable HCC [14]. Ren et al. and Kelley et al., respectively, reported that sintilimab (anti-PD-1) plus bevacizumab biosimilar and atezolizumab plus cabozantinib, which is a tyrosine kinases c-Met and VEGFR2 inhibitor, compared with sorafenib, achieved clinically meaningful improvements in OS and PFS for advanced/unresectable HCC [15, 17].\nIn patients with unresectable/advanced HCC, to the best of our knowledge, this is the first meta-analysis on RCTs to investigate the efficacy of targeted therapy in combination with immunotherapy versus targeted monotherapy. Although several trials are still ongoing, only three RCTs have been published. Compared with sorafenib, significantly better OS and PFS were observed with sintilimab plus bevacizumab (HR = 0.57, 95% CI: 0.43–0.75; HR = 0.56, 95% CI: 0.45–0.69), and atezolizumab plus bevacizumab (HR = 0.58, 95% CI: 0.58–0.80; HR = 0.59, 95% CI: 0.46–0.75), respectively. In terms of grade 3–5 AEs, the uses of lenvatinib (HR = 1.51; 95% CI: 1.14–2.00) and linifanib (HR = 1.94, 95% CI: 1.41–2.66) were higher than sorafenib. More data from updated clinical trials are still needed to confirm the benefit of combination therapy for HCC patients.\nIn the analyses of TRAEs, the results showed that compared with targeted monotherapy, the combination therapy had a significantly higher incidence of grade 3–5 TRAEs. The most common grade 3 or 4 TRAE with atezolizumab + bevacizumab and sintilimab + bevacizumab biosimilar group was hypertension (both 15%), which is consistent with the established safety profile of bevacizumab [18]. Besides, gastrointestinal disorders were the most common reasons for treatment discontinuation (5%) in both groups, as expected in patients with liver cancer and underlying cirrhosis. The most common grade 3 or 4 TRAE was alanine aminotransferase increase (9% in the cabozantinib plus atezolizumab combination treatment group). In one of the included studies, Kelley et al. reported immune-mediated adverse events of any grade requiring immunosuppressive treatment occurred in 31 (7%) of 429 patients in the combination treatment group [17]. The most common ones were hepatitis (4%) and pneumonitis (2%). For these 3 trials, the most common TRAEs from targeted agents were hypertension and elevated alanine aminotransferase. Grade 3 or 4 TRAEs, immune mediated or non-immune mediated, leading to study treatment discontinuation were infrequent in these 3 trials, indicating that these TRAEs were manageable with immunosuppressive drugs or other treatments. Potential candidates for the combination therapy of targeted drug and immunotherapy should be provided with this information.\nUnresectable HCC management is still challenged in patients with cirrhosis and varied degree of impaired liver function. Immunotherapy, such as pembrolizumab and nivolumab, has been a viable and safe option in patients with advanced HCC [19]. Newer systemic drugs like the combination of immune checkpoint inhibitors with biologic therapy, such as ramucirumab plus durvalumab treatment, likely to be promising as a new treatment standard for patients with unresectable HCC [20].\nAtezolizumab is also used for other cancers like non-small cell lung cancer and advanced renal cell carcinoma. The COSMIC-021 study reported the combination therapy with atezolizumab and cabozantinib for advanced renal cell carcinoma [21], which regimen was similar to COSMIC-312 trial [17], appeared to be tolerable with a manageable toxicity profile. Grade 3 or 4 TRAE occurred in 59% of patients, slightly higher than 36% of patients of COSMIC-312 trial. Grade 3 or 4 Immune-mediated events were 30%. TRAEs leading to discontinuation of drug was 19–24% for subgroups. All AEs were managed with dose modifications and supportive care.\nAs for the second-line treatment, regorafenib showed promising results after sorafenib failure in HCC patients [22]. One meta-analysis evaluating the efficacy and safety of regorafenib in unresectable HCC showed that pooled objective response rate was as high as 10.1% and pooled median OS of 11.1 months, as well as TRAE greater than Grade 3 was 50-58.3%. Regorafenib represents a valuable and comparatively safe therapeutic option in patients who progress on sorafenib [23]. HCC scenario is continuously and rapidly changing for decades due to different etiology and treatment advance, including the progressions of patients age, increased non-viral cases and an earlier stage migration [24]. This molecular information should also be integrated in the future to guide us to deal with the cancer more precisely [25].\nThe present study included several limitations. First, this meta-analysis mainly compared combination therapy with targeted monotherapy. These included RCTs, however, used various targeted agents and immunotherapy drugs, which may have biased the data analysis from the dissimilar therapeutic effects and AEs between drugs. The efficacy and TRAEs of individual drug in the combination can be further investigated using indirect comparison in the future. Second, the comparison of combination therapy with targeted monotherapy in patients with advanced/unrespectable HCC included only three RCTs with the limited information. Some other ongoing studies, such as LEAP-002 (lenvatinib plus pembrolizumab versus lenvatinib) [26] may be included in the near future. Third, the cost-effective analysis was insufficient in these trials. Cost-effective issue for HCC might be important because of the higher cost for the combination therapy than targeted monotherapy. More studies, especially those with the cost-effective analysis, are warranted in the future.", "Our meta-analysis concluded that compared with targeted monotherapy, targeted therapy in combination with PD-1/PD-L1 inhibitors provided the survival benefits in patients with unresectable HCC. The patients receiving combination therapy had significantly higher incidences of grade 3–5 adverse effects." ]
[ "introduction", null, null, null, null, null, "results", null, null, null, null, "discussion", "conclusion" ]
[ "Targeted therapy", "Immunotherapy", "Unresectable hepatocellular carcinoma", "Systematic review", "Meta-analysis" ]
Evaluation of anaplastic thyroid carcinoma in the Kurdistan region of Iraq.
36271386
Anaplastic thyroid carcinoma is a rare and lethal disease that accounts for 1-2% of thyroid malignancies. It is an aggressive locoregional disease with a high rate of distant metastasis, a poor prognosis, and a mean survival rate of 3-6 months after diagnosis. This retrospective study aimed to analyse the clinical and pathological features of ATC to assess treatment procedures and its outcome.
BACKGROUND
We analysed data from 22 patients diagnosed with ATC from 2018 to 2021, using the Kaplan-Meier method and log-rank test to determine overall survival.
METHODS
Patients' median age was 64.3 ± 17.1 years. Females were more affected (male/female ratio: 1:1.7); 14 cases occurred in females (63.6.4%), and eight in males (36.4%). The most common manifestations were neck enlargement (81.8%) and dyspnoea (72.27%), and the tumour size was > 4 cm in 17 (77.3%) patients. The percentage of cases that presented in clinical-stage IVA was 36.4%, with 31.8% presenting in clinical-stage IVB and 31.8% presenting in clinical-stage VIB. Among 22 cases, 14 (63.6%) were operable, and 8 (36.4) were inoperable (p = 0.015). Multimodal therapies were associated with better survival (surgery plus radiotherapy without systemic treatment, P = 0.063). The median overall survival was three months (IC 95%, 0.078-5.922). One-year and two-year survival rates were 9% and 4.5%, respectively.
RESULTS
ATC is a rapidly growing cancer that, fortunately, is rare. Early diagnosis and multimodality treatment may provide a better quality of life and survival time for this group of patients.
CONCLUSION
[ "Humans", "Female", "Male", "Middle Aged", "Aged", "Aged, 80 and over", "Thyroid Carcinoma, Anaplastic", "Retrospective Studies", "Quality of Life", "Iraq", "Prognosis", "Thyroid Neoplasms", "Treatment Outcome" ]
9587643
null
null
null
null
Results
A total of 22 patients agreed to participate in this study, comprising 14 females (63.6%) and eight males (36.4%). The male-to-female ratio was 1:1.7. The mean age was 64.3 ± 17.1 years, ranging from 26 to 87 years. The majority (n = 14, 63.6%) were aged ≥ 60 years. The most common clinical presentation was swelling in the anterior neck’s lower part, which was observed in 18 (81.8%) patients. It was bilateral in a third of them (n = 6, 33.3%). More than a third (n = 8, 36.4%) of patients were stage A, and the tumour size was > 4 cm in 17 (77.3%) patients. The majority (n = 20, 90.9%) of patients were diagnosed by FNAC, and the rest (n = 2, 9.1%) by excisional biopsy as ATC. The ATC-related clinical characteristics of participants are shown in Table 1. Table 1Characteristics of patients with ATCNo.(%)Mean (SD)MedianMin.Max. Gender Male8(36.4)Female14(63.6) Age at diagnosis 64.3 (17.1)68.026.087.0< 60 years836.4≥ 60 years1463.6 Clinical presentation Lump18(81.8)Pain3(13.6)Dyspnoea16(72.7)Dysphagia10(45.5)Hoarseness of voice9(40.9)Stridor2(9.1) Site of lump 18Right4(22.2)Left4(22.2)Bilateral6(33.3)Diffuse4(22.2) Staging Stage 4  A. Local infiltration8(36.4)B. Regional involvement of lymph nodes7(31.8)C. Distant metastasis7(31.8) Tumour size 5.8 (2.0)5.7312≤ 4 cm5(22.7)> 4 cm17(77.3) WBC 8911 (4354)8350280018,000 Fine needle aspiration Indeterminate*2(9.1)Malignant (anaplastic)20(90.9)Total22(100.0)*Proved post-operatively by tissue biopsy to be malignant Characteristics of patients with ATC Staging Stage 4 *Proved post-operatively by tissue biopsy to be malignant Different methods were used for the treatment of the 22 included cases, of which 14 (63.6%) were operable. Six patients (27.3% of total cases) underwent total thyroidectomy with neck dissection, while the rest had other techniques of surgery like lobectomy (4 cases), subtotal thyroidectomy (1 case) and debulking (3 cases). The multimodal method was applied only for two cases. Supportive interventions such as tracheostomy were undertaken for 3 (13.6% of total cases) patients due to airway obstruction, and one (4.5%) patient had percutaneous endoscopic gastrostomy. Participants’ therapeutic modalities are described in Table 2. Table 2Treatment modalitiesNo.%)Type of surgeryLobectomy4(18.2)Subtotal thyroidectomy1(4.5)Total thyroidectomy6(27.3)Debulking3(13.6)Inoperable8(36.4)Type of neck dissection (n = 6)Central3(50.0)Central and lateral3(50.0)Type of therapyChemotherapy2(9.1)Radiotherapy1(4.5)Palliative Surgery*4(18.2)*Surgery: - supportive interventions such as tracheostomy were undertaken for three patients, and one patient had percutaneous endoscopic gastrostomy as palliative therapy for those inoperable cases Treatment modalities *Surgery: - supportive interventions such as tracheostomy were undertaken for three patients, and one patient had percutaneous endoscopic gastrostomy as palliative therapy for those inoperable cases The mean survival rate was 5.5 months, and the median was 3.0 months. Two (9%) patients who got thyroidectomy, chemotherapy and radiotherapy lived longer than one year, of whom only one case (4.5%) was alive without any evidence of recurrence of the disease at 40-month follow-up. There was no significant association between survival rate in relation to age (P = 0.805) and gender (P = 0.103) (Table 3). Table 3Survival by age and genderMeanMedian 95% Confidence Interval 95% Confidence Interval P* Estimate SE Lower Bound Upper Bound Estimate SE Lower Bound Upper Bound Age (y)< 605.161.741.758.574.003.670.0011.20≥ 605.711.542.688.743.000.921.184.810.805GenderMale3.161.240.715.611.00Female6.571.543.559.584.001.870.337.660.103Overall5.551.173.247.853.001.490.075.92*By Log Rank (Mantel-Cox) Survival by age and gender *By Log Rank (Mantel-Cox) There was no significant association between the survival of patients with tumour grade (P = 0.441) and White blood cell (WBC) count (P = 0.104), as shown in Table 4; Figs. 1 and 2. Significant associations were found between tumour size (P = 0.006) and operability (P = 0.015) and patient survival (Table 4; Figs. 3 and 4). There was no significant association between all treatment modalities and survival rate (P = 0.063) (Table 4; Fig. 5). Fig. 1Survival by grading Survival by grading Fig. 2Survival by WBC count Survival by WBC count Fig. 3Survival by tumor size Survival by tumor size Fig. 4Survival by intervention Survival by intervention Fig. 5Survival by type of therapy Survival by type of therapy Table 4Survival by characteristics and treatment of ATCMeanMedian1`95% Confidence Interval95% Confidence IntervalP*EstimateSELower BoundEstimateSELower BoundUpper BoundGradingA7.422.173.1711.687.001.304.439.56B3.711.301.156.272.000.431.142.85C5.502.640.3210.672.001.830.005.600.441Size≤ 4 cm1.500.280.932.061.00> 4 cm6.561.363.899.225.002.001.088.920.006WBCs< 10,0006.841.603.699.995.002.390.309.69≥ 10,0003.141.200.785.502.000.391.212.780.104InterventionOperable7.411.734.0010.827.002.302.4711.52Non-operable2.750.641.484.022.000.451.102.890.015Type of therapyChemotherapy3.5002.5000.0008.4001.000Radio-therapy2.0000.0002.0002.0002.000Surgery3.2501.7970.0006.7721.0002.0000.0004.920Chemo and radio3.0000.7071.6144.3862.000Multiple modality9.5007.5000.00024.2002.000Sympto-matic treatment1.0000.0001.0001.0001.000Radio surgery therapy9.5002.1875.21313.7878.0001.2255.60010.4000.063Overall5.5501.1783.2427.8583.0001.4910.0785.922*By Log Rank (Mantel-Cox) Survival by characteristics and treatment of ATC *By Log Rank (Mantel-Cox)
Conclusion
ATC and its treatment in the Kurdistan Region exhibit the same tendencies as elsewhere in the world. This study shows the superiority of early diagnosis and multimodality treatment as two important factors that may provide better survival rates. However, this study is limited by its small number of cases. The retrospective nature of this study limits the approach to many specific issues of concern for such pathology. At the same time, local specifications like the inaccessibility of radiotherapy and chemotherapy shortly after surgery, and the lack of proper awareness and screening programs in the country, make the diagnosis and prognosis of this pathology much worse than they could be. ATC is a rapidly growing cancer that, fortunately, is rare. Early diagnosis and multimodality treatment may provide a better quality of life and survival time for patients, and awareness of this among healthcare decision makers in the Kurdistan Region should be encouraged to increase screening and healthcare-seeking behaviour among the public.
[ "Background", "Methods", "Patients and methods", "Statistical analysis", "" ]
[ "Anaplastic thyroid carcinoma (ATC) is an uncommon but profoundly aggressive thyroid malignancy established in undifferentiated follicular thyroid cells [1, 2]. It accounts for more than 50% of all deaths attributable to thyroid tumours, with a mean survival period of three to four months and disease-specific mortality at one year of about 100% [1, 3]. ATC prevalence varies from one place to another, with higher rates in goitre-endemic areas; in the USA it accounts for 1.7% of all thyroid malignancies, ranging from 1.3 to 9.8% [4]. It is commonly diagnosed in elderly persons (the mean age at diagnosis is 65–70 years), with a slightly higher female prevalence [5]. The yearly frequency of ATC is roughly one-to-two cases per million people.\nRisk factors for the development of ATC include a history of radiation to the neck or chest, long-standing goitre, progressed age, and a history of benign or malignant thyroid disease [6]. Patients with ATC encounter noteworthy nearby compressive symptoms due to a quickly advancing central neck mass (77%), accompanying dysphagia (40%), hoarseness (40%), and stridor (24%). Metastases are well known in 50% of patients at the time of diagnosis, most commonly in the lungs (80%), bone (6–16%), and brain (5–13%) [7, 8].\nThe diagnostic modality for ATC is fine needle aspiration cytology (FNAC). In doubtful cases, the diagnosis is made by histology on a tissue biopsy [9]. Because of its aggressive behaviour and low chance of recovery, ATC is classified according to the American Joint Committee on Cancer TNM (AJCC-8) system as T4 and stage IV [10, 11].\nContrast-enhanced neck computed tomography (CT) is recommended to assess the advancement of tumour attack and lymph node metastasis. For the assessment of far-off metastases, fluorodeoxyglucose (FDG), positron emission tomography (PET) and brain magnetic resonance imaging (MRI) are commonly prescribed [12].\nMultidisciplinary healthcare teams (comprising surgeons, radiation oncologists, medical oncologists, psychologists, and nurses, among others) are inherently involved in ATC treatment, and interdisciplinary communication and collaborative working can represent a significant challenge during ATC management. A significant number of ATC patients are non-operable at the time of diagnosis of their cancer. Within the subset of patients with operable disease at the time of diagnosis, surgery combined with postoperative radiation +/- chemotherapy may be useful [1, 13].\nThe purpose of this study is to retrospectively analyse clinical and pathological features of ATC to assess treatment procedures and outcomes.", "[SUBTITLE] Patients and methods [SUBSECTION] In this retrospective study, 700 cases of thyroid cancer were examined from the 1st of June 2018 to the 31st of December 2021). Of these 700 cases, 22 were included as cases of ATC. The data were obtained from various hospitals and health care centres (Rizgary, Nanakaky, Hewa, and Azadi Teaching Hospitals, with their oncologic units) throughout the main cities of the Kurdistan Region (Erbil, Sulaymaniyah, and Duhok) in northern Iraq. All 22 cases were pathologically diagnosed as ATC, depending on FNAC and excisional biopsy.\nPreoperative investigation documentation such as that concerning laryngoscopy, neck ultrasonography, and CT of the head, neck, chest, and abdomen were examined to detect the extent of tumour invasion. Two conditions comprised exclusion criteria: poorly differentiated thyroid cancer and patients’ refusal to participate. A questionnaire was administered to record participants’ demographic characteristics (age, sex, place of residence) and their clinical presentation, diagnostic procedures, therapeutic approaches and outcomes, and survival. Additional data was collected from patients’ relatives by direct interviewing or calling.\nPrognostic factors like survival rates by age, tumour size, presence of distant metastasis, and treatment were calculated. The guidelines of the American Joint Committee on Cancer were used for staging: tumour (T), nodal (N), and distant metastasis (M).\nThis study is part of an ongoing Ph.D. research project sponsored by the College of Medicine at Hawler Medical University. It was approved by the Ethical Committee in the College of Medicine (meeting code: 6, paper code: 6, date: 27/04/2022).\nIn this retrospective study, 700 cases of thyroid cancer were examined from the 1st of June 2018 to the 31st of December 2021). Of these 700 cases, 22 were included as cases of ATC. The data were obtained from various hospitals and health care centres (Rizgary, Nanakaky, Hewa, and Azadi Teaching Hospitals, with their oncologic units) throughout the main cities of the Kurdistan Region (Erbil, Sulaymaniyah, and Duhok) in northern Iraq. All 22 cases were pathologically diagnosed as ATC, depending on FNAC and excisional biopsy.\nPreoperative investigation documentation such as that concerning laryngoscopy, neck ultrasonography, and CT of the head, neck, chest, and abdomen were examined to detect the extent of tumour invasion. Two conditions comprised exclusion criteria: poorly differentiated thyroid cancer and patients’ refusal to participate. A questionnaire was administered to record participants’ demographic characteristics (age, sex, place of residence) and their clinical presentation, diagnostic procedures, therapeutic approaches and outcomes, and survival. Additional data was collected from patients’ relatives by direct interviewing or calling.\nPrognostic factors like survival rates by age, tumour size, presence of distant metastasis, and treatment were calculated. The guidelines of the American Joint Committee on Cancer were used for staging: tumour (T), nodal (N), and distant metastasis (M).\nThis study is part of an ongoing Ph.D. research project sponsored by the College of Medicine at Hawler Medical University. It was approved by the Ethical Committee in the College of Medicine (meeting code: 6, paper code: 6, date: 27/04/2022).\n[SUBTITLE] Statistical analysis [SUBSECTION] Data were analysed using the SPSS version 25. Numerical variables were summarised by calculating the means and the standard deviations (SDs). Categorical variables were presented in the form of frequencies and proportions. Kaplan-Meier survival analysis was applied, and Log Rank (Mantel-Cox) test was used to compare the survival curves of the studied variables (like age, gender, grading, and size). A P value of ≤ 0.05 was considered statistically significant.\nData were analysed using the SPSS version 25. Numerical variables were summarised by calculating the means and the standard deviations (SDs). Categorical variables were presented in the form of frequencies and proportions. Kaplan-Meier survival analysis was applied, and Log Rank (Mantel-Cox) test was used to compare the survival curves of the studied variables (like age, gender, grading, and size). A P value of ≤ 0.05 was considered statistically significant.", "In this retrospective study, 700 cases of thyroid cancer were examined from the 1st of June 2018 to the 31st of December 2021). Of these 700 cases, 22 were included as cases of ATC. The data were obtained from various hospitals and health care centres (Rizgary, Nanakaky, Hewa, and Azadi Teaching Hospitals, with their oncologic units) throughout the main cities of the Kurdistan Region (Erbil, Sulaymaniyah, and Duhok) in northern Iraq. All 22 cases were pathologically diagnosed as ATC, depending on FNAC and excisional biopsy.\nPreoperative investigation documentation such as that concerning laryngoscopy, neck ultrasonography, and CT of the head, neck, chest, and abdomen were examined to detect the extent of tumour invasion. Two conditions comprised exclusion criteria: poorly differentiated thyroid cancer and patients’ refusal to participate. A questionnaire was administered to record participants’ demographic characteristics (age, sex, place of residence) and their clinical presentation, diagnostic procedures, therapeutic approaches and outcomes, and survival. Additional data was collected from patients’ relatives by direct interviewing or calling.\nPrognostic factors like survival rates by age, tumour size, presence of distant metastasis, and treatment were calculated. The guidelines of the American Joint Committee on Cancer were used for staging: tumour (T), nodal (N), and distant metastasis (M).\nThis study is part of an ongoing Ph.D. research project sponsored by the College of Medicine at Hawler Medical University. It was approved by the Ethical Committee in the College of Medicine (meeting code: 6, paper code: 6, date: 27/04/2022).", "Data were analysed using the SPSS version 25. Numerical variables were summarised by calculating the means and the standard deviations (SDs). Categorical variables were presented in the form of frequencies and proportions. Kaplan-Meier survival analysis was applied, and Log Rank (Mantel-Cox) test was used to compare the survival curves of the studied variables (like age, gender, grading, and size). A P value of ≤ 0.05 was considered statistically significant.", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1: Information Sheet\n\nSupplementary Material 1: Information Sheet\n\nSupplementary Material 2: Ethics committee approval sheet\n\nSupplementary Material 2: Ethics committee approval sheet" ]
[ null, null, null, null, null ]
[ "Background", "Methods", "Patients and methods", "Statistical analysis", "Results", "Discussion", "Conclusion", "Electronic supplementary material", "" ]
[ "Anaplastic thyroid carcinoma (ATC) is an uncommon but profoundly aggressive thyroid malignancy established in undifferentiated follicular thyroid cells [1, 2]. It accounts for more than 50% of all deaths attributable to thyroid tumours, with a mean survival period of three to four months and disease-specific mortality at one year of about 100% [1, 3]. ATC prevalence varies from one place to another, with higher rates in goitre-endemic areas; in the USA it accounts for 1.7% of all thyroid malignancies, ranging from 1.3 to 9.8% [4]. It is commonly diagnosed in elderly persons (the mean age at diagnosis is 65–70 years), with a slightly higher female prevalence [5]. The yearly frequency of ATC is roughly one-to-two cases per million people.\nRisk factors for the development of ATC include a history of radiation to the neck or chest, long-standing goitre, progressed age, and a history of benign or malignant thyroid disease [6]. Patients with ATC encounter noteworthy nearby compressive symptoms due to a quickly advancing central neck mass (77%), accompanying dysphagia (40%), hoarseness (40%), and stridor (24%). Metastases are well known in 50% of patients at the time of diagnosis, most commonly in the lungs (80%), bone (6–16%), and brain (5–13%) [7, 8].\nThe diagnostic modality for ATC is fine needle aspiration cytology (FNAC). In doubtful cases, the diagnosis is made by histology on a tissue biopsy [9]. Because of its aggressive behaviour and low chance of recovery, ATC is classified according to the American Joint Committee on Cancer TNM (AJCC-8) system as T4 and stage IV [10, 11].\nContrast-enhanced neck computed tomography (CT) is recommended to assess the advancement of tumour attack and lymph node metastasis. For the assessment of far-off metastases, fluorodeoxyglucose (FDG), positron emission tomography (PET) and brain magnetic resonance imaging (MRI) are commonly prescribed [12].\nMultidisciplinary healthcare teams (comprising surgeons, radiation oncologists, medical oncologists, psychologists, and nurses, among others) are inherently involved in ATC treatment, and interdisciplinary communication and collaborative working can represent a significant challenge during ATC management. A significant number of ATC patients are non-operable at the time of diagnosis of their cancer. Within the subset of patients with operable disease at the time of diagnosis, surgery combined with postoperative radiation +/- chemotherapy may be useful [1, 13].\nThe purpose of this study is to retrospectively analyse clinical and pathological features of ATC to assess treatment procedures and outcomes.", "[SUBTITLE] Patients and methods [SUBSECTION] In this retrospective study, 700 cases of thyroid cancer were examined from the 1st of June 2018 to the 31st of December 2021). Of these 700 cases, 22 were included as cases of ATC. The data were obtained from various hospitals and health care centres (Rizgary, Nanakaky, Hewa, and Azadi Teaching Hospitals, with their oncologic units) throughout the main cities of the Kurdistan Region (Erbil, Sulaymaniyah, and Duhok) in northern Iraq. All 22 cases were pathologically diagnosed as ATC, depending on FNAC and excisional biopsy.\nPreoperative investigation documentation such as that concerning laryngoscopy, neck ultrasonography, and CT of the head, neck, chest, and abdomen were examined to detect the extent of tumour invasion. Two conditions comprised exclusion criteria: poorly differentiated thyroid cancer and patients’ refusal to participate. A questionnaire was administered to record participants’ demographic characteristics (age, sex, place of residence) and their clinical presentation, diagnostic procedures, therapeutic approaches and outcomes, and survival. Additional data was collected from patients’ relatives by direct interviewing or calling.\nPrognostic factors like survival rates by age, tumour size, presence of distant metastasis, and treatment were calculated. The guidelines of the American Joint Committee on Cancer were used for staging: tumour (T), nodal (N), and distant metastasis (M).\nThis study is part of an ongoing Ph.D. research project sponsored by the College of Medicine at Hawler Medical University. It was approved by the Ethical Committee in the College of Medicine (meeting code: 6, paper code: 6, date: 27/04/2022).\nIn this retrospective study, 700 cases of thyroid cancer were examined from the 1st of June 2018 to the 31st of December 2021). Of these 700 cases, 22 were included as cases of ATC. The data were obtained from various hospitals and health care centres (Rizgary, Nanakaky, Hewa, and Azadi Teaching Hospitals, with their oncologic units) throughout the main cities of the Kurdistan Region (Erbil, Sulaymaniyah, and Duhok) in northern Iraq. All 22 cases were pathologically diagnosed as ATC, depending on FNAC and excisional biopsy.\nPreoperative investigation documentation such as that concerning laryngoscopy, neck ultrasonography, and CT of the head, neck, chest, and abdomen were examined to detect the extent of tumour invasion. Two conditions comprised exclusion criteria: poorly differentiated thyroid cancer and patients’ refusal to participate. A questionnaire was administered to record participants’ demographic characteristics (age, sex, place of residence) and their clinical presentation, diagnostic procedures, therapeutic approaches and outcomes, and survival. Additional data was collected from patients’ relatives by direct interviewing or calling.\nPrognostic factors like survival rates by age, tumour size, presence of distant metastasis, and treatment were calculated. The guidelines of the American Joint Committee on Cancer were used for staging: tumour (T), nodal (N), and distant metastasis (M).\nThis study is part of an ongoing Ph.D. research project sponsored by the College of Medicine at Hawler Medical University. It was approved by the Ethical Committee in the College of Medicine (meeting code: 6, paper code: 6, date: 27/04/2022).\n[SUBTITLE] Statistical analysis [SUBSECTION] Data were analysed using the SPSS version 25. Numerical variables were summarised by calculating the means and the standard deviations (SDs). Categorical variables were presented in the form of frequencies and proportions. Kaplan-Meier survival analysis was applied, and Log Rank (Mantel-Cox) test was used to compare the survival curves of the studied variables (like age, gender, grading, and size). A P value of ≤ 0.05 was considered statistically significant.\nData were analysed using the SPSS version 25. Numerical variables were summarised by calculating the means and the standard deviations (SDs). Categorical variables were presented in the form of frequencies and proportions. Kaplan-Meier survival analysis was applied, and Log Rank (Mantel-Cox) test was used to compare the survival curves of the studied variables (like age, gender, grading, and size). A P value of ≤ 0.05 was considered statistically significant.", "In this retrospective study, 700 cases of thyroid cancer were examined from the 1st of June 2018 to the 31st of December 2021). Of these 700 cases, 22 were included as cases of ATC. The data were obtained from various hospitals and health care centres (Rizgary, Nanakaky, Hewa, and Azadi Teaching Hospitals, with their oncologic units) throughout the main cities of the Kurdistan Region (Erbil, Sulaymaniyah, and Duhok) in northern Iraq. All 22 cases were pathologically diagnosed as ATC, depending on FNAC and excisional biopsy.\nPreoperative investigation documentation such as that concerning laryngoscopy, neck ultrasonography, and CT of the head, neck, chest, and abdomen were examined to detect the extent of tumour invasion. Two conditions comprised exclusion criteria: poorly differentiated thyroid cancer and patients’ refusal to participate. A questionnaire was administered to record participants’ demographic characteristics (age, sex, place of residence) and their clinical presentation, diagnostic procedures, therapeutic approaches and outcomes, and survival. Additional data was collected from patients’ relatives by direct interviewing or calling.\nPrognostic factors like survival rates by age, tumour size, presence of distant metastasis, and treatment were calculated. The guidelines of the American Joint Committee on Cancer were used for staging: tumour (T), nodal (N), and distant metastasis (M).\nThis study is part of an ongoing Ph.D. research project sponsored by the College of Medicine at Hawler Medical University. It was approved by the Ethical Committee in the College of Medicine (meeting code: 6, paper code: 6, date: 27/04/2022).", "Data were analysed using the SPSS version 25. Numerical variables were summarised by calculating the means and the standard deviations (SDs). Categorical variables were presented in the form of frequencies and proportions. Kaplan-Meier survival analysis was applied, and Log Rank (Mantel-Cox) test was used to compare the survival curves of the studied variables (like age, gender, grading, and size). A P value of ≤ 0.05 was considered statistically significant.", "A total of 22 patients agreed to participate in this study, comprising 14 females (63.6%) and eight males (36.4%). The male-to-female ratio was 1:1.7. The mean age was 64.3 ± 17.1 years, ranging from 26 to 87 years. The majority (n = 14, 63.6%) were aged ≥ 60 years.\nThe most common clinical presentation was swelling in the anterior neck’s lower part, which was observed in 18 (81.8%) patients. It was bilateral in a third of them (n = 6, 33.3%). More than a third (n = 8, 36.4%) of patients were stage A, and the tumour size was > 4 cm in 17 (77.3%) patients.\nThe majority (n = 20, 90.9%) of patients were diagnosed by FNAC, and the rest (n = 2, 9.1%) by excisional biopsy as ATC. The ATC-related clinical characteristics of participants are shown in Table 1.\n\nTable 1Characteristics of patients with ATCNo.(%)Mean (SD)MedianMin.Max.\nGender\nMale8(36.4)Female14(63.6)\nAge at diagnosis\n64.3 (17.1)68.026.087.0< 60 years836.4≥ 60 years1463.6\nClinical presentation\nLump18(81.8)Pain3(13.6)Dyspnoea16(72.7)Dysphagia10(45.5)Hoarseness of voice9(40.9)Stridor2(9.1)\nSite of lump\n18Right4(22.2)Left4(22.2)Bilateral6(33.3)Diffuse4(22.2)\nStaging\n\nStage 4\n A. Local infiltration8(36.4)B. Regional involvement of lymph nodes7(31.8)C. Distant metastasis7(31.8)\nTumour size\n5.8 (2.0)5.7312≤ 4 cm5(22.7)> 4 cm17(77.3)\nWBC\n8911 (4354)8350280018,000\nFine needle aspiration\nIndeterminate*2(9.1)Malignant (anaplastic)20(90.9)Total22(100.0)*Proved post-operatively by tissue biopsy to be malignant\n\nCharacteristics of patients with ATC\n\nStaging\n\n\nStage 4\n\n*Proved post-operatively by tissue biopsy to be malignant\nDifferent methods were used for the treatment of the 22 included cases, of which 14 (63.6%) were operable. Six patients (27.3% of total cases) underwent total thyroidectomy with neck dissection, while the rest had other techniques of surgery like lobectomy (4 cases), subtotal thyroidectomy (1 case) and debulking (3 cases). The multimodal method was applied only for two cases. Supportive interventions such as tracheostomy were undertaken for 3 (13.6% of total cases) patients due to airway obstruction, and one (4.5%) patient had percutaneous endoscopic gastrostomy. Participants’ therapeutic modalities are described in Table 2.\n\nTable 2Treatment modalitiesNo.%)Type of surgeryLobectomy4(18.2)Subtotal thyroidectomy1(4.5)Total thyroidectomy6(27.3)Debulking3(13.6)Inoperable8(36.4)Type of neck dissection (n = 6)Central3(50.0)Central and lateral3(50.0)Type of therapyChemotherapy2(9.1)Radiotherapy1(4.5)Palliative Surgery*4(18.2)*Surgery: - supportive interventions such as tracheostomy were undertaken for three patients, and one patient had percutaneous endoscopic gastrostomy as palliative therapy for those inoperable cases\n\nTreatment modalities\n*Surgery: - supportive interventions such as tracheostomy were undertaken for three patients, and one patient had percutaneous endoscopic gastrostomy as palliative therapy for those inoperable cases\nThe mean survival rate was 5.5 months, and the median was 3.0 months. Two (9%) patients who got thyroidectomy, chemotherapy and radiotherapy lived longer than one year, of whom only one case (4.5%) was alive without any evidence of recurrence of the disease at 40-month follow-up. There was no significant association between survival rate in relation to age (P = 0.805) and gender (P = 0.103) (Table 3).\n\nTable 3Survival by age and genderMeanMedian\n95% Confidence Interval\n\n95% Confidence Interval\n\nP*\n\nEstimate\n\nSE\n\nLower Bound\n\nUpper Bound\n\nEstimate\n\nSE\n\nLower Bound\n\nUpper Bound\nAge (y)< 605.161.741.758.574.003.670.0011.20≥ 605.711.542.688.743.000.921.184.810.805GenderMale3.161.240.715.611.00Female6.571.543.559.584.001.870.337.660.103Overall5.551.173.247.853.001.490.075.92*By Log Rank (Mantel-Cox)\n\nSurvival by age and gender\n*By Log Rank (Mantel-Cox)\nThere was no significant association between the survival of patients with tumour grade (P = 0.441) and White blood cell (WBC) count (P = 0.104), as shown in Table 4; Figs. 1 and 2. Significant associations were found between tumour size (P = 0.006) and operability (P = 0.015) and patient survival (Table 4; Figs. 3 and 4). There was no significant association between all treatment modalities and survival rate (P = 0.063) (Table 4; Fig. 5).\n\nFig. 1Survival by grading\n\nSurvival by grading\n\nFig. 2Survival by WBC count\n\nSurvival by WBC count\n\nFig. 3Survival by tumor size\n\nSurvival by tumor size\n\nFig. 4Survival by intervention\n\nSurvival by intervention\n\nFig. 5Survival by type of therapy\n\nSurvival by type of therapy\n\nTable 4Survival by characteristics and treatment of ATCMeanMedian1`95% Confidence Interval95% Confidence IntervalP*EstimateSELower BoundEstimateSELower BoundUpper BoundGradingA7.422.173.1711.687.001.304.439.56B3.711.301.156.272.000.431.142.85C5.502.640.3210.672.001.830.005.600.441Size≤ 4 cm1.500.280.932.061.00> 4 cm6.561.363.899.225.002.001.088.920.006WBCs< 10,0006.841.603.699.995.002.390.309.69≥ 10,0003.141.200.785.502.000.391.212.780.104InterventionOperable7.411.734.0010.827.002.302.4711.52Non-operable2.750.641.484.022.000.451.102.890.015Type of therapyChemotherapy3.5002.5000.0008.4001.000Radio-therapy2.0000.0002.0002.0002.000Surgery3.2501.7970.0006.7721.0002.0000.0004.920Chemo and radio3.0000.7071.6144.3862.000Multiple modality9.5007.5000.00024.2002.000Sympto-matic treatment1.0000.0001.0001.0001.000Radio surgery therapy9.5002.1875.21313.7878.0001.2255.60010.4000.063Overall5.5501.1783.2427.8583.0001.4910.0785.922*By Log Rank (Mantel-Cox)\n\nSurvival by characteristics and treatment of ATC\n*By Log Rank (Mantel-Cox)", "Anaplastic thyroid carcinoma is one of the fastest-growing tumours in humans, with local invasion and a high rate of distant metastasis. It has a poor prognosis, with a mean survival rate of 3–6 months after diagnosis [14, 15]. However, according to the data available, it is one of the uncommon malignancies of humanity. In the current study, the incidence of ATC was 3.15% of all cases of thyroid cancer, and the overall survival rate was 5.5 months.\nFemales are more prone to thyroid disease in general and cancers in particular. In our study, which retrospectively selected ATC patients, females comprised 63.6% of the sample (with a male-to-female ratio of 1:1.7), which agrees with the published data in different national contexts [16–18]. Most patients were over 60 years (63.6%), reflecting the association of age with the oncological process, including ATC [5]. This is related to the effect of tumour suppressor gene inhibition in older age, apparently as part of the natural ageing process [19].\nAccording to published literature, local symptoms of ATC most commonly begin with a rapidly evolving central neck mass (77%), followed by noticeable dysphagia (40%), voice change or hoarseness (40%), and stridor (24%) [7, 8]. Our study showed similar results in the way in which most patients (81.8%) presented with compressive symptoms. The disease progression to surrounding tissues is one of the most important aspects of pathophysiology since it directly impacts the treatment and prognosis. In our observation stage, IVB + IVC was about 63.6%, and studies of rapid extension to surrounding tissues have reported that 80% of ATC patients have disease in both thyroid and nearby tissues at their initial presentation [1, 4–8, 20].\nHowever, the survival duration of our cases with stage IVB + IVC was not so different from group A, who lived for 3–11 months, while those in Groups B and C lived for 2 weeks to 10 months (with a median of 2 months). Three months is commonly reported to be the median survival window [21]. The relatively shorter duration of survival in our study (2 months) could be explained by the delayed diagnosis and modality of treatments in the studied cases.\nParticipants’ tumour size was > 4 cm among the majority (77.3%) of participants. Previous studies reported tumours ranging between 3.6 and 10.5 cm, but only 68.9% of cases were reported to be larger than 4 cm [16, 22]. Even though larger tumour sizes are bad prognostic factors [22], those with larger tumours survived longer than those with smaller ones in the current study. This discrepancy could be explained by the role of large size in increasing patient awareness and motivation to seek treatment at an earlier stage, thereby increasing the scope of treatment and survival prospects.\nOne of the prognostic factors in ATC is the level of WBC. A patient with a WBC level of less than 10,000 /µL has a better prognosis regarding the duration of survival and may live 11.3 months, compared to patients with leucocytosis who average 3.6 months [23, 24]. In our study, the difference between the leucocytosis and the non-leucocytosis group was not statistically significant. However, there was a difference in the survival window (2 weeks to 9 months for non-leucocytosis and 2 weeks to 3 months for leucocytosis patients). Thus, the total body reaction in the form of leucocytosis may adversely affect the survival duration.\nThe incidence rate of thyroid cancer is increased, despite advances in early detection and understanding of the disease. Advanced molecular techniques such as DNA zyme are needed to diagnose accurately. Additionally, they may substantially impact the prognosis categorisation of thyroid cancer and provide a more accurate diagnosis [25, 26]. However, our area still uses traditional methods such as clinical examinations, x-rays, FNAC, and histopathological examinations.\nThe FNAC as a diagnostic tool in ATC has good support for its range of accuracy: 89.71% accuracy in diagnosis for 94.7% of ATC cases diagnosed by FNAC [27]. In our study, FNAC was used for all patients, and it was accurate in 20 (90.9%) cases as a diagnostic tool, given that all our cases were included and based on histopathological reports that assured the accuracy of FNAC for such malignancy.\nRecent studies showed the diagnosis of ATC is difficult due to mimic to other carcinomas in the neck like poorly differentiated thyroid cancer, spindle cell variant of medullary carcinoma, solitary fibrous tumor and spindle epithelial tumor with thymus-like differentiation, leiomyosarcoma of the thyroid gland and different pathological changes that may affect normal ectopic thyroid [28, 29]. In addition to the above factors, the mentality and absence of a central hospital for the management of oncological cases in our region can be regarded as another two factors that may affect the late presentation of cases, missing cases during the diagnosis process, and improper management.\nSurgery is one of the best options for patients to remove the tumour completely or improve its local effects, like airway compression. However, in small, non-extended tumours, it could be considered a curative for a short duration, with potential application as a palliative measure in inoperable patients. However, it could potentially extend survival outcomes in the case of surgery combined with chemoradiation [22]. This study found that surgery showed a better median survival time of 7 months for operated cases, compared to 2 months for non-operable ones. The 9% of patients who lived more than one year (a remarkable duration for ATC patients) were those who had undergone surgery as a part of treatment (in addition to chemoradiotherapy).\nThe main treatment modality in the group of patients with local and distant metastasis was adjuvant therapy, with or without debulking. Trials for different chemotherapy variants do not significantly change the duration of survival in ATC [30]. However, hypo fractionated radiotherapy significantly prolonged the median survival duration in this category of patients [31].\nSingle modality treatment does not show good prognostic effects; although surgery was one of the mainstays of treatment in itself (without additional treatment), it only achieved a 2-month survival rate. Similarly, when used alone, radiotherapy and chemotherapy achieved 2 and 3.5 months of survival, respectively. Conversely, multimodal treatment extended the survival window to 9.5 months – in particular, this was achieved by surgery in combination with radiotherapy (combined without chemotherapy). Combining chemotherapy and radiotherapy without surgery only produced a mean survival time of 3 months.\nIn addition, treatment approaches based on molecular pan-inhibitors of Aurora kinases, such as VX-680/MK-0457, SNS-314, and ZM447439 on human ATC-derived cell lines and urokinase-type plasminogen activator, have been developed [32, 33]. However, the clinical trials of these agents do not yet consider them in the management of ATC in our region.", "ATC and its treatment in the Kurdistan Region exhibit the same tendencies as elsewhere in the world. This study shows the superiority of early diagnosis and multimodality treatment as two important factors that may provide better survival rates. However, this study is limited by its small number of cases. The retrospective nature of this study limits the approach to many specific issues of concern for such pathology. At the same time, local specifications like the inaccessibility of radiotherapy and chemotherapy shortly after surgery, and the lack of proper awareness and screening programs in the country, make the diagnosis and prognosis of this pathology much worse than they could be. ATC is a rapidly growing cancer that, fortunately, is rare. Early diagnosis and multimodality treatment may provide a better quality of life and survival time for patients, and awareness of this among healthcare decision makers in the Kurdistan Region should be encouraged to increase screening and healthcare-seeking behaviour among the public.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1: Information Sheet\n\nSupplementary Material 1: Information Sheet\n\nSupplementary Material 2: Ethics committee approval sheet\n\nSupplementary Material 2: Ethics committee approval sheet\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1: Information Sheet\n\nSupplementary Material 1: Information Sheet\n\nSupplementary Material 2: Ethics committee approval sheet\n\nSupplementary Material 2: Ethics committee approval sheet", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1: Information Sheet\n\nSupplementary Material 1: Information Sheet\n\nSupplementary Material 2: Ethics committee approval sheet\n\nSupplementary Material 2: Ethics committee approval sheet" ]
[ null, null, null, null, "results", "discussion", "conclusion", "supplementary-material", null ]
[ "Anaplastic thyroid carcinoma", "Surgery", "Multimodal therapy", "Prognosis", "Survival", "Kurdistan region Iraq" ]
Association between depression, happiness, and sleep duration: data from the UAE healthy future pilot study.
36271400
The United Arab Emirates Healthy Future Study (UAEHFS) is one of the first large prospective cohort studies and one of the few studies in the region which examines causes and risk factors for chronic diseases among the nationals of the United Arab Emirates (UAE). The aim of this study is to investigate the eight-item Patient Health Questionnaire (PHQ-8) as a screening instrument for depression among the UAEHFS pilot participants.
BACKGROUND
The UAEHFS pilot data were analyzed to examine the relationship between the PHQ-8 and possible confounding factors, such as self-reported happiness, and self-reported sleep duration (hours) after adjusting for age, body mass index (BMI), and gender.
METHODS
Out of 517 participants who met the inclusion criteria, 487 (94.2%) participants filled out the questionnaire and were included in the statistical analysis using 100 multiple imputations. 231 (44.7%) were included in the primary statistical analysis after omitting the missing values. Participants' median age was 32.0 years (Interquartile Range: 24.0, 39.0). In total, 22 (9.5%) of the participant reported depression. Females have shown significantly higher odds of reporting depression than males with an odds ratio = 3.2 (95% CI:1.17, 8.88), and there were approximately 5-fold higher odds of reporting depression for unhappy than for happy individuals. For one interquartile-range increase in age and BMI, the odds ratio of reporting depression was 0.34 (95% CI: 0.1, 1.0) and 1.8 (95% CI: 0.97, 3.32) respectively.
RESULTS
Females are more likely to report depression compared to males. Increasing age may decrease the risk of reporting depression. Unhappy individuals have approximately 5-fold higher odds of reporting depression compared to happy individuals. A higher BMI was associated with a higher risk of reporting depression. In a sensitivity analysis, individuals who reported less than 6 h of sleep per 24 h were more likely to report depression than those who reported 7 h of sleep.
CONCLUSION
[ "Male", "Female", "Humans", "Adult", "Happiness", "Pilot Projects", "Prospective Studies", "Depression", "United Arab Emirates", "Sleep" ]
9587590
Introduction
Depression is defined as a set of disorders ranging from mild to moderate to severe [1]. It is widely recognized as a major public health problem worldwide [2]. Reports from the Global Burden of Diseases declared major depressive disorder as one of the top three causes of disability-adjusted life years [3, 4]. The effect of depression has been extensively studied on the individual’s daily functioning and productivity. This is directly reflected in increased economic costs per capita [3, 4]. For example, in Catalonia in 2006, the average annual cost of an adult with depression was close to 1800 Euros, and the total annual cost of depression was 735.4 million Euros. These costs were linked directly to primary care, mental health specialized care, hospitalization, and pharmacological care, as well as indirect costs due to productivity loss, temporary and permanent disability [4]. To measure the level of depression in non-clinical populations; clinical and epidemiological studies have often used the established and validated eight-item Patient Health Questionnaire scale (PHQ-8) instead of the nine- item Patient Health Questionnaire scale (PHQ-9) [5]. As has been confirmed by previous studies, the scale can detect major depression with sensitivity and a specificity of 88%, to classify subjects into depressed or non-depressed, respectively [6–8]. Regionally, in Jordan, Lebanon, Syria and Afghanistan, the cutoff point of 10 was used [9–12]. Similarly, in UAE, studies have mostly used the cutoff point of 10 [13–16]. Recent studies have focused on exploring the methods of improving individual and environmental effects on disability related to depression by investigating its triggers and associations [2, 10]. Factors such as sleep duration and self-reported happiness are evidenced to be predictors for depression [17, 18]. Sleep duration can be considered a risk factor for lower well-being [19]. The number of sleep hours has also been found to have a causal relationship with depression as well as self-reported happiness [20]. For example, shorter sleep duration might lead to lower positive emotions such as self-reported happiness and showed stronger associations with negative emotional affect [21, 22]. One question that needs to be asked, however, is the nature and strength of the association between these three variables respectively. Sleep duration is determined by how many hours an individual sleeps over 24 h. Individuals with depression often have poor sleep status including abnormal REM (rapid eye movement), and insomnia (difficulty falling asleep or staying asleep). Abnormal REM may contribute to the development of altered emotional processing in depression [23]. It was reported that people with insomnia might have a ten-fold higher risk of developing depression in contrast to people who get a good night’s sleep [24]. Also, 75% of depressed individuals will have trouble falling asleep or staying asleep [25]. A bidirectional relationship between sleep duration and reported depression has been investigated [24, 26]. Such studies are unsatisfactory because they do not explore the association between major depressive disorder and sleep duration based on specific population demographics. In this study we are considering other sociodemographic factors such as age, gender, and marital status. Interest in studying happiness in the context of mental health status (such as depression and anxiety) has been growing recently [27]. The findings of some research papers suggest that self-reported happiness is a potential factor in the prevention and management of depression [27, 34]. Happiness is correlated to a person’s ability to approach situations in a less stressful manner and to an individual’s capacity to perceive and control their own feelings. This indicates that higher happiness levels may have a protective effect on depression [27]. Well-being has been defined as the combination of feeling good and functioning well [28]; conversely, quality of life could be defined as an individual’s satisfaction with his or her actual life compared with his or her ideal life. Evaluation of the quality of life depends on one’s value system [29]. Moreover, studies have shown that individuals use various chronically accessible and stable sources of information when making life satisfaction judgments [30]. Yet, well-being, quality of life and life satisfaction are multidimensional constructs which include complex cognitive evaluation processes and cannot be adequately assessed by using a single item or question [31]. Unlike quality of life, which typically requires detailed assessments to ascertain [32], happiness is easier to evaluate using a single item question [33]. Investigating the association between happiness and depression would add valuable information to public health research as the relationship between happiness and depression is observed to be bidirectional (i.e., one variable can predict the other, and people might not report depression but are more likely to report feeling unhappy) [34]. In addition, there are some factors which confound with sleep duration and self-reported happiness, such as age, gender, and marital status [36, 37]. For example, reviews reported that a prognosis of depression was improved with increasing age [38]. Conversely, another study showed that older age was associated with worsening of depressive symptoms [37]. Moreover, depressive symptoms could worsen among widowed individuals as their age increased [40]. Some research findings reflected that females are more likely to perceive depression compared to males [38, 41]. Other studies showed significant interplay between marital status and depressive symptoms [40, 42]. For example, being unmarried could lead to perceiving and developing depressive symptoms [43], while a worsening in depression symptoms among married individuals could lead to separation or being unmarried [42, 44]. So, further exploration is required to study the interplay of these variables together. The United Arab Emirates (UAE) is a high-income developed country which has undergone a rapid epidemiological transition from a traditional semi-nomadic society to a modern affluent society with a lifestyle characterized by over-consumption of energy-dense foods and low physical activity [47]. The UAE was ranked 15th out of 157 countries included in the WHO World Happiness Report with a score of 7.06 [48]. Despite this, depression has been identified as the third leading cause of disability in the UAE [3]. Nevertheless, there are few studies in the Gulf region examining the relationship between happiness, sleep duration and depression [36, 49, 50]. Therefore, studying the association between depression, happiness, and sleep in the United Arab Emirates (UAE) population would be of interest to the public health field in this part of the world. This study aimed to examine the relationship between depression, self-reported happiness, and self-reported sleep duration after adjusting for age and gender using the UAE Healthy Future Study (UAEHFS) pilot data.
null
null
Results
Of 517 participants who consented to participate in the UAEHF pilot study, 487 (94.2%) had completed questionnaire data [47]. Figure 1 describes all key phases from recruitment up to inclusion in the study and in the statistical analysis. After omitting missing values, 231 (44.7%) participants were included in the main statistical analysis. However, 487 (94.2%) participants were included in the sensitivity analysis (using 100 multiple imputations) [73]. The median age of the UAEHFS pilot data participants was 32.0 years (Interquartile Range: 24.0–39.0). The percentage of females included in the study was 32%, which represented the UAE population well [59]. Therefore, we did not make any adjustments for gender bias. Fig. 1Flow chart of UAEHF pilot study (describes all the key phases from the recruitment up to the inclusion in the study analysis) Flow chart of UAEHF pilot study (describes all the key phases from the recruitment up to the inclusion in the study analysis) Note: This Figure represents the data of the included participants in the main statistical analysis after omitting missing values. The number of observed values (%) of each categorical variable was presented by the PHQ-8 categories in Table 1, where the majority of the study participants (90.5%) had PHQ-8 score less than 10. When categorical groups were compared within the PHQ-8 depression group (< 10 versus ≥ 10), there was only a statistically significant difference between females versus males with a Fisher exact p-value = 0.028. Table 1 shows that there was a statistically significant difference in the age distribution across the depression groups (p-value = 0.012). There was no statistically significant difference in the BMI measurements between the PHQ-8 groups (p-value = 0.44). The result of the primary analysis showed that females have statistically significantly greater odds of reporting depression compared to males OR = 3.2 (95% CI:1.1, 8.9), p-value = 0.024 (Table 2). Furthermore, there was an approximately 5-fold greater odds of reporting depression for unhappy than for happy individuals. However, this was not statistically significant (p-value = 0.10). Similarly, the sleep duration and marital status were both not statistically significant (p-value of 0.284 and 0.205, respectively; Table 2). However, in a sensitivity analysis, people who reported sleep duration of less than 6 h were more likely to report depression as compared to the people who reported sleep of 7 h OR (95% CI) of 2.6 (1.0, 6.4), p-value = 0.040 and 1.136 (1.015, 1.272), p-value = 0.027. Table 2 The results of the primary analysis of the fitted multivariate logistic regression model VariableOR (95% CI)PbHappiness - HappyReferenceHappiness - Unhappy5.5 (0.7, 42.2)0.100Age (linear)0.3 (0.1, 1.0)a0.056BMI (linear)1.8 (1.0, 3.3)a0.064Gender = MalesReferenceGender = Females3.2 (1.2, 8.9)0.024Sleep = 7ReferenceSleep < 64.0 (0.9, 17.4)0.061Sleep = 61.3 (0.2, 6.9)0.775Sleep = 81.5 (0.3, 7.3)0.585Sleep > 81.2 (0.2, 9.2)0.848MarriedReferenceSingle0.9 (0.3, 3.0)0.8Others0.2 (0.02, 3.3)0.3Note: aInterquartile-range odds ratio for age and BMI, which compares the 3rd quartile with the 1st quartile, and simple odds ratios for the categorical predictors (happiness, gender, sleep and marital status) compare each group with the reference group (the largest group) with corresponding 95% confidence interval (95% CI). bWald’s p-values are presented for each variable The results of the primary analysis of the fitted multivariate logistic regression model Note: aInterquartile-range odds ratio for age and BMI, which compares the 3rd quartile with the 1st quartile, and simple odds ratios for the categorical predictors (happiness, gender, sleep and marital status) compare each group with the reference group (the largest group) with corresponding 95% confidence interval (95% CI). bWald’s p-values are presented for each variable To compute Interquartile Range (IQR) odds ratios for continuous variables, the age and BMI variables were divided by their IQR values of 15 and 8.7, respectively [54, 56]. Table 2 shows that for one interquartile-range increase in the age and BMI, the IQR-OR was 0.34 (95% CI: 0.12, 1.0) and 1.8 (95% CI: 0.97, 3.3), respectively, where both results were statistically significant with a p-value of 0.056 and 0.064 individually. The results of the sensitivity analysis using 100 multiple imputations (Table 3) were approximately similar to the result of the multivariate logistic ordinal regression analyses using omitted data (Table 2). However, the happiness variable was statistically significant OR = 5.1 (95%CI: 1.7, 15.7, p-value = 0.005), and there was a statistically significant difference between the sleep variable for less than six hours as compared with seven hours OR = 2.6 (95% CI: 1.0, 6.4, p-value = 0.04). Supplementary Fig. 1 illustrates the percentages of missing values in the variables included in this statistical analysis. Marital status had the lowest number of missing values (5.3%), followed by BMI (12.7%) and the Sleep variable with 12.9% missing values. The Happiness variable had 21.1% missing values. The PHQ-8 variables had 32.6–35.1% missing values. Supplementary Table 1 shows the percentages of the “Prefer not to answer (DA)” and “Do not know (UN)” in the eight PHQ questions, where the percentages vary between 16 and 18.5% between the eight questions. The percentages of “Prefer not to answer (DA)” and “Do not know (UN)” in the happiness variable was 2.5% and 2.3%, respectively, which were also considered missing values, although these were not missing at random and can be correlated with one of the PHQ-8 answers. However, this has been addressed in the multiple imputation analysis. Table 3 Results of the sensitivity analysis using multivariate logistic regression models with a sample size N = 487 VariableOR (95% CI)PbHappiness – HappyReferenceHappiness - Unhappy5.1 (1.7, 15.7)0.004Age (linear)0.5 (0.3, 1.0)a0.060BMI (linear)1.7 (1.1, 2.5)a0.021Gender = MalesReferenceGender = Females1.6 (0.9, 3.0)0.154Sleep = 7ReferenceSleep < 62.6 (1.0, 6.4)0.040Sleep = 60.9 (0.3, 2.6)0.887Sleep = 80.9 (0.3, 2.4)0.779Sleep > 81.1 (0.3, 3.7)0.853MarriedReferenceSingle0.9 (0.4, 2.1)0.885Others0.7 (0.2, 2.9)0.597Note: aInterquartile-range odds ratio for Age and BMI, which compares the 3rd quartile with the 1st quartile, and simple odds ratios for the categorical predictors (happiness, gender, sleep and marital status) compare each group with the reference group (the largest group) with corresponding 95% confidence interval (95% CI). bWald’s p-values are presented for each variable. Results were summarized using Rubin’s rules Results of the sensitivity analysis using multivariate logistic regression models with a sample size N = 487 Note: aInterquartile-range odds ratio for Age and BMI, which compares the 3rd quartile with the 1st quartile, and simple odds ratios for the categorical predictors (happiness, gender, sleep and marital status) compare each group with the reference group (the largest group) with corresponding 95% confidence interval (95% CI). bWald’s p-values are presented for each variable. Results were summarized using Rubin’s rules An additional sensitivity analysis was performed using the ordinal PHQ-8 score as an outcome in a multivariate linear regression model (see supplementary Table 1). The result of this sensitivity analysis was very similar to the sensitivity analysis in Table 3.
Conclusion
The results of this study indicate that females are more likely to report depression compared to males. Older age is associated with a decrease in self-reported depression. Unhappy individuals are approximately 5-fold higher odds to report depression compared to happy individuals. BMI was positively associated with reporting depression. The result of the sensitivity analysis shows that individuals who sleep less than 6 h per day are more likely to report depression compared to those who sleep 7 h per day. The results of this study have a potential value for researchers and public health professionals as it presents novel data on the PHQ-8 score in a healthy UAE population, which has not been explored before. Furthermore, the results of this study can help contribute to the knowledge base on current and potential population mental health impact in the UAE and Gulf Region.
[ "Study design", "Participants’ eligibility criteria and recruitment", "Measures", "Statistical analysis", "Sensitivity analysis", "Strengths and Limitations", "" ]
[ "This was a pilot prospective cohort study conducted from January 2015 to May 2015. The participants were recruited from two health care centers in Abu Dhabi. Participants completed an online questionnaire including questions on demographic data, PHQ-8 score, self-reported sleep duration, and self-reported happiness score. Physical measurements such as Body Mass Index (BMI) were collected during the recruitment visit.", "Seven hundred and sixty-nine UAE nationals aged ≥ 18 years were invited to participate voluntarily in the pilot study. Volunteers from the general population with inclusion criteria of age 18 or greater; able to consent; UAE nationals, resident in Abu Dhabi Emirate. All potential participants were given participant information leaflets in either Arabic or English to read and had the opportunity to ask questions prior to completion of the recruitment process. Participants signed an informed consent and were asked to complete a detailed questionnaire. However, 243 invited subjects did not respond, and their reasons for not participating were recorded [45]. Out of 517 participants who met the inclusion criteria, 487 (94.2%) participants filled out the questionnaire and were included in the statistical analysis [47].", "The PHQ-8 questionnaire was used to measure the participants’ depression levels [6, 39, 51]. This study used the cutoff point of 10 as well based on the common local practice considering that there are no specific related research guidelines in UAE [13–16]. The PHQ-8 score was dichotomized into no-depression (total PHQ-8 < 10) versus depression (≥ 10) [6, 51].\nHappiness was measured using a one-question item that asked participants, “In general, how happy you are?’’ Those who responded as extremely happy, very happy and moderately happy were grouped as “happy” while those responding as moderately unhappy, very unhappy, and extremely unhappy were grouped as “unhappy”.\nDemographic variables such as age, gender, and marital status (single, married, and others) were also included in the reference. Sleep duration data was collected as an ordinal variable (number of hours) by asking how many hours of sleep the participant gets in a 24-hour period, including naps. Sleep duration was categorized into five categories (see Table 1) to avoid the linearity assumption between sleep duration and depression status as well as to be able to compare average sleepers to shorter and longer sleepers. The questionnaire was translated from English into Arabic and back-translated into English to check for linguistic validity.\n\nTable 1\nNumber (percentages) of the analyzed variables and median (IQR) for Age and BMI.\nVariableGroupPHQ-8 < 10PHQ-8 ≥ 10P-valueGenderFemale62 (83.8)12 (16.2)0.028aMale147 (93.6)10 (6.4)Sleep (hours)< 639 (83)8 (17)0.284a= 653 (93)4 (7)= 757 (95)3 (5)= 844 (89.8)5 (10.2)> 816 (88.9)2 (11.1)Marital StatusSingle120 (93)9 (7)0.205aMarried15 (93.8)1 (6.2)Other74 (86)12 (14)HappinessHappy204 (91.1)20 (8.9)0.136aUnhappy5 (71.4)2 (28.6)Total209 (90.5)22 (9.5)Median (IQR)Median (IQR)P-valueAgeyears33.0 (25.0, 40.0)24.5 (22.0, 35.0)0.012bBMIkg/m227.6 (23.4, 31.2)28.8 (24.2, 33.0)0.444bNote: aFisher’s exact test p-values for categorical data and bWilcoxon rank sum test for continuous data\n\n\nNumber (percentages) of the analyzed variables and median (IQR) for Age and BMI.\n\nNote: aFisher’s exact test p-values for categorical data and bWilcoxon rank sum test for continuous data\nBody mass index (BMI) was obtained via physical measurement using Tanita MC-780 MA Segmental Body Composition Analyzer [52]. All physical measurements were collected by a clinical research nurse. Additional details of the study recruitment have been previously described [47].", "All eligible participants 487 (94.2%) were included in a sensitivity analysis (using 100 multiple imputations). After excluding participants with missing values, 231 (44.7%) were included in the primary statistical analysis. The PHQ-8 questionnaire was used in this statistical analysis with two additional possible options to select for each question (i.e., P2A – P2H). These were “Do not know (UN)” and “Prefer not to answer (DA)”, which were treated as missing values in the statistical analysis. Fisher’s exact test was used to investigate the association between depression and categorical variables, such as Sleep (hours), Happiness, Marital Status, and Gender. Wilcoxon rank-sum test was performed to investigate differences in the distribution of age and BMI within the depressed versus non-depressed group respectively.\nTo examine the factors associated with depression, a multivariate logistic regression model was performed with the dichotomized PHQ-8 score as the outcome. The predictors were Happiness score (Happy vs. Unhappy), Age (linear), Gender (Females vs. Males), BMI (linear), and Marital status (categorical). Interquartile-range odds ratios (IQR-OR’s) for continuous predictors and simple odds ratios (OR’s) for categorical predictors with 95% confidence intervals (Cis) were estimated for continuous and categorical variables respectively; corresponding p-values were calculated [53].\nIn sensitivity analysis, a multivariate logistic regression model and a multivariate linear regression model were conducted using multiple imputations (see sensitivity analysis section). All applied statistical tests were two-sided; p < 0.05 were considered statistically significant. No adjustment for multiple comparisons was made. Statistical analyses were performed in R version 4.0.2 [54].", "The primary statistical analysis included subjects with at least one non-missing value. However, in a sensitivity analysis, a multivariate imputation by chained equations (MICE) procedure was applied with Classification and Regression Trees (CART) to impute missing values [55]. 100 multiple imputations were used [56]. Rubin’s rules were used to combine the multiple imputed estimates [57].\nThe pattern of missing values was investigated, and it was found that subjects who “did not want to answer” were not systematically different from those who answered the questionnaire. Therefore, “prefer not to answer” was recorded as missing value in the statistical analysis and was considered a missing variable in the sensitivity analysis [55].", "Missing values were omitted in the primary statistical analysis, which decreased the sample size and can lead to overfitting in the main finding [55]. Therefore, the number of participants with the PHQ-8 ≥ 10 (i.e. – events) was 22 which is a limitation in this data set because it does not allow us to fit a complex multivariate statistical regression model if we would follow the statistical rule of thumb “ten events per predictor” [56, 57, 73]. Thus, sensitivity analysis was performed, and the result of the sensitivity analysis (Table 3) was approximately the same as the main finding (Table 2).\nAlthough some limitations were found in this study, the findings provide future direction to mental health research. Further investigation is needed with a larger sample size using the main UAEHFS data to have a better picture of the role of happiness, marital status, sleep, and social demographic variables in association with depression and mental health disorders.", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, null, null, null, null, null, null ]
[ "Introduction", "Materials and methods", "Study design", "Participants’ eligibility criteria and recruitment", "Measures", "Statistical analysis", "Sensitivity analysis", "Results", "Discussion", "Strengths and Limitations", "Conclusion", "Electronic supplementary material", "" ]
[ "Depression is defined as a set of disorders ranging from mild to moderate to severe [1]. It is widely recognized as a major public health problem worldwide [2]. Reports from the Global Burden of Diseases declared major depressive disorder as one of the top three causes of disability-adjusted life years [3, 4]. The effect of depression has been extensively studied on the individual’s daily functioning and productivity. This is directly reflected in increased economic costs per capita [3, 4]. For example, in Catalonia in 2006, the average annual cost of an adult with depression was close to 1800 Euros, and the total annual cost of depression was 735.4 million Euros. These costs were linked directly to primary care, mental health specialized care, hospitalization, and pharmacological care, as well as indirect costs due to productivity loss, temporary and permanent disability [4].\nTo measure the level of depression in non-clinical populations; clinical and epidemiological studies have often used the established and validated eight-item Patient Health Questionnaire scale (PHQ-8) instead of the nine- item Patient Health Questionnaire scale (PHQ-9) [5]. As has been confirmed by previous studies, the scale can detect major depression with sensitivity and a specificity of 88%, to classify subjects into depressed or non-depressed, respectively [6–8]. Regionally, in Jordan, Lebanon, Syria and Afghanistan, the cutoff point of 10 was used [9–12]. Similarly, in UAE, studies have mostly used the cutoff point of 10 [13–16].\nRecent studies have focused on exploring the methods of improving individual and environmental effects on disability related to depression by investigating its triggers and associations [2, 10]. Factors such as sleep duration and self-reported happiness are evidenced to be predictors for depression [17, 18]. Sleep duration can be considered a risk factor for lower well-being [19]. The number of sleep hours has also been found to have a causal relationship with depression as well as self-reported happiness [20]. For example, shorter sleep duration might lead to lower positive emotions such as self-reported happiness and showed stronger associations with negative emotional affect [21, 22]. One question that needs to be asked, however, is the nature and strength of the association between these three variables respectively.\nSleep duration is determined by how many hours an individual sleeps over 24 h. Individuals with depression often have poor sleep status including abnormal REM (rapid eye movement), and insomnia (difficulty falling asleep or staying asleep). Abnormal REM may contribute to the development of altered emotional processing in depression [23]. It was reported that people with insomnia might have a ten-fold higher risk of developing depression in contrast to people who get a good night’s sleep [24]. Also, 75% of depressed individuals will have trouble falling asleep or staying asleep [25]. A bidirectional relationship between sleep duration and reported depression has been investigated [24, 26]. Such studies are unsatisfactory because they do not explore the association between major depressive disorder and sleep duration based on specific population demographics. In this study we are considering other sociodemographic factors such as age, gender, and marital status.\nInterest in studying happiness in the context of mental health status (such as depression and anxiety) has been growing recently [27]. The findings of some research papers suggest that self-reported happiness is a potential factor in the prevention and management of depression [27, 34]. Happiness is correlated to a person’s ability to approach situations in a less stressful manner and to an individual’s capacity to perceive and control their own feelings. This indicates that higher happiness levels may have a protective effect on depression [27]. Well-being has been defined as the combination of feeling good and functioning well [28]; conversely, quality of life could be defined as an individual’s satisfaction with his or her actual life compared with his or her ideal life. Evaluation of the quality of life depends on one’s value system [29].\nMoreover, studies have shown that individuals use various chronically accessible and stable sources of information when making life satisfaction judgments [30]. Yet, well-being, quality of life and life satisfaction are multidimensional constructs which include complex cognitive evaluation processes and cannot be adequately assessed by using a single item or question [31]. Unlike quality of life, which typically requires detailed assessments to ascertain [32], happiness is easier to evaluate using a single item question [33]. Investigating the association between happiness and depression would add valuable information to public health research as the relationship between happiness and depression is observed to be bidirectional (i.e., one variable can predict the other, and people might not report depression but are more likely to report feeling unhappy) [34].\nIn addition, there are some factors which confound with sleep duration and self-reported happiness, such as age, gender, and marital status [36, 37]. For example, reviews reported that a prognosis of depression was improved with increasing age [38]. Conversely, another study showed that older age was associated with worsening of depressive symptoms [37]. Moreover, depressive symptoms could worsen among widowed individuals as their age increased [40]. Some research findings reflected that females are more likely to perceive depression compared to males [38, 41]. Other studies showed significant interplay between marital status and depressive symptoms [40, 42]. For example, being unmarried could lead to perceiving and developing depressive symptoms [43], while a worsening in depression symptoms among married individuals could lead to separation or being unmarried [42, 44]. So, further exploration is required to study the interplay of these variables together.\nThe United Arab Emirates (UAE) is a high-income developed country which has undergone a rapid epidemiological transition from a traditional semi-nomadic society to a modern affluent society with a lifestyle characterized by over-consumption of energy-dense foods and low physical activity [47]. The UAE was ranked 15th out of 157 countries included in the WHO World Happiness Report with a score of 7.06 [48]. Despite this, depression has been identified as the third leading cause of disability in the UAE [3]. Nevertheless, there are few studies in the Gulf region examining the relationship between happiness, sleep duration and depression [36, 49, 50]. Therefore, studying the association between depression, happiness, and sleep in the United Arab Emirates (UAE) population would be of interest to the public health field in this part of the world. This study aimed to examine the relationship between depression, self-reported happiness, and self-reported sleep duration after adjusting for age and gender using the UAE Healthy Future Study (UAEHFS) pilot data.", "[SUBTITLE] Study design [SUBSECTION] This was a pilot prospective cohort study conducted from January 2015 to May 2015. The participants were recruited from two health care centers in Abu Dhabi. Participants completed an online questionnaire including questions on demographic data, PHQ-8 score, self-reported sleep duration, and self-reported happiness score. Physical measurements such as Body Mass Index (BMI) were collected during the recruitment visit.\nThis was a pilot prospective cohort study conducted from January 2015 to May 2015. The participants were recruited from two health care centers in Abu Dhabi. Participants completed an online questionnaire including questions on demographic data, PHQ-8 score, self-reported sleep duration, and self-reported happiness score. Physical measurements such as Body Mass Index (BMI) were collected during the recruitment visit.\n[SUBTITLE] Participants’ eligibility criteria and recruitment [SUBSECTION] Seven hundred and sixty-nine UAE nationals aged ≥ 18 years were invited to participate voluntarily in the pilot study. Volunteers from the general population with inclusion criteria of age 18 or greater; able to consent; UAE nationals, resident in Abu Dhabi Emirate. All potential participants were given participant information leaflets in either Arabic or English to read and had the opportunity to ask questions prior to completion of the recruitment process. Participants signed an informed consent and were asked to complete a detailed questionnaire. However, 243 invited subjects did not respond, and their reasons for not participating were recorded [45]. Out of 517 participants who met the inclusion criteria, 487 (94.2%) participants filled out the questionnaire and were included in the statistical analysis [47].\nSeven hundred and sixty-nine UAE nationals aged ≥ 18 years were invited to participate voluntarily in the pilot study. Volunteers from the general population with inclusion criteria of age 18 or greater; able to consent; UAE nationals, resident in Abu Dhabi Emirate. All potential participants were given participant information leaflets in either Arabic or English to read and had the opportunity to ask questions prior to completion of the recruitment process. Participants signed an informed consent and were asked to complete a detailed questionnaire. However, 243 invited subjects did not respond, and their reasons for not participating were recorded [45]. Out of 517 participants who met the inclusion criteria, 487 (94.2%) participants filled out the questionnaire and were included in the statistical analysis [47].", "This was a pilot prospective cohort study conducted from January 2015 to May 2015. The participants were recruited from two health care centers in Abu Dhabi. Participants completed an online questionnaire including questions on demographic data, PHQ-8 score, self-reported sleep duration, and self-reported happiness score. Physical measurements such as Body Mass Index (BMI) were collected during the recruitment visit.", "Seven hundred and sixty-nine UAE nationals aged ≥ 18 years were invited to participate voluntarily in the pilot study. Volunteers from the general population with inclusion criteria of age 18 or greater; able to consent; UAE nationals, resident in Abu Dhabi Emirate. All potential participants were given participant information leaflets in either Arabic or English to read and had the opportunity to ask questions prior to completion of the recruitment process. Participants signed an informed consent and were asked to complete a detailed questionnaire. However, 243 invited subjects did not respond, and their reasons for not participating were recorded [45]. Out of 517 participants who met the inclusion criteria, 487 (94.2%) participants filled out the questionnaire and were included in the statistical analysis [47].", "The PHQ-8 questionnaire was used to measure the participants’ depression levels [6, 39, 51]. This study used the cutoff point of 10 as well based on the common local practice considering that there are no specific related research guidelines in UAE [13–16]. The PHQ-8 score was dichotomized into no-depression (total PHQ-8 < 10) versus depression (≥ 10) [6, 51].\nHappiness was measured using a one-question item that asked participants, “In general, how happy you are?’’ Those who responded as extremely happy, very happy and moderately happy were grouped as “happy” while those responding as moderately unhappy, very unhappy, and extremely unhappy were grouped as “unhappy”.\nDemographic variables such as age, gender, and marital status (single, married, and others) were also included in the reference. Sleep duration data was collected as an ordinal variable (number of hours) by asking how many hours of sleep the participant gets in a 24-hour period, including naps. Sleep duration was categorized into five categories (see Table 1) to avoid the linearity assumption between sleep duration and depression status as well as to be able to compare average sleepers to shorter and longer sleepers. The questionnaire was translated from English into Arabic and back-translated into English to check for linguistic validity.\n\nTable 1\nNumber (percentages) of the analyzed variables and median (IQR) for Age and BMI.\nVariableGroupPHQ-8 < 10PHQ-8 ≥ 10P-valueGenderFemale62 (83.8)12 (16.2)0.028aMale147 (93.6)10 (6.4)Sleep (hours)< 639 (83)8 (17)0.284a= 653 (93)4 (7)= 757 (95)3 (5)= 844 (89.8)5 (10.2)> 816 (88.9)2 (11.1)Marital StatusSingle120 (93)9 (7)0.205aMarried15 (93.8)1 (6.2)Other74 (86)12 (14)HappinessHappy204 (91.1)20 (8.9)0.136aUnhappy5 (71.4)2 (28.6)Total209 (90.5)22 (9.5)Median (IQR)Median (IQR)P-valueAgeyears33.0 (25.0, 40.0)24.5 (22.0, 35.0)0.012bBMIkg/m227.6 (23.4, 31.2)28.8 (24.2, 33.0)0.444bNote: aFisher’s exact test p-values for categorical data and bWilcoxon rank sum test for continuous data\n\n\nNumber (percentages) of the analyzed variables and median (IQR) for Age and BMI.\n\nNote: aFisher’s exact test p-values for categorical data and bWilcoxon rank sum test for continuous data\nBody mass index (BMI) was obtained via physical measurement using Tanita MC-780 MA Segmental Body Composition Analyzer [52]. All physical measurements were collected by a clinical research nurse. Additional details of the study recruitment have been previously described [47].", "All eligible participants 487 (94.2%) were included in a sensitivity analysis (using 100 multiple imputations). After excluding participants with missing values, 231 (44.7%) were included in the primary statistical analysis. The PHQ-8 questionnaire was used in this statistical analysis with two additional possible options to select for each question (i.e., P2A – P2H). These were “Do not know (UN)” and “Prefer not to answer (DA)”, which were treated as missing values in the statistical analysis. Fisher’s exact test was used to investigate the association between depression and categorical variables, such as Sleep (hours), Happiness, Marital Status, and Gender. Wilcoxon rank-sum test was performed to investigate differences in the distribution of age and BMI within the depressed versus non-depressed group respectively.\nTo examine the factors associated with depression, a multivariate logistic regression model was performed with the dichotomized PHQ-8 score as the outcome. The predictors were Happiness score (Happy vs. Unhappy), Age (linear), Gender (Females vs. Males), BMI (linear), and Marital status (categorical). Interquartile-range odds ratios (IQR-OR’s) for continuous predictors and simple odds ratios (OR’s) for categorical predictors with 95% confidence intervals (Cis) were estimated for continuous and categorical variables respectively; corresponding p-values were calculated [53].\nIn sensitivity analysis, a multivariate logistic regression model and a multivariate linear regression model were conducted using multiple imputations (see sensitivity analysis section). All applied statistical tests were two-sided; p < 0.05 were considered statistically significant. No adjustment for multiple comparisons was made. Statistical analyses were performed in R version 4.0.2 [54].", "The primary statistical analysis included subjects with at least one non-missing value. However, in a sensitivity analysis, a multivariate imputation by chained equations (MICE) procedure was applied with Classification and Regression Trees (CART) to impute missing values [55]. 100 multiple imputations were used [56]. Rubin’s rules were used to combine the multiple imputed estimates [57].\nThe pattern of missing values was investigated, and it was found that subjects who “did not want to answer” were not systematically different from those who answered the questionnaire. Therefore, “prefer not to answer” was recorded as missing value in the statistical analysis and was considered a missing variable in the sensitivity analysis [55].", "Of 517 participants who consented to participate in the UAEHF pilot study, 487 (94.2%) had completed questionnaire data [47]. Figure 1 describes all key phases from recruitment up to inclusion in the study and in the statistical analysis. After omitting missing values, 231 (44.7%) participants were included in the main statistical analysis. However, 487 (94.2%) participants were included in the sensitivity analysis (using 100 multiple imputations) [73]. The median age of the UAEHFS pilot data participants was 32.0 years (Interquartile Range: 24.0–39.0). The percentage of females included in the study was 32%, which represented the UAE population well [59]. Therefore, we did not make any adjustments for gender bias.\n\nFig. 1Flow chart of UAEHF pilot study (describes all the key phases from the recruitment up to the inclusion in the study analysis)\n\nFlow chart of UAEHF pilot study (describes all the key phases from the recruitment up to the inclusion in the study analysis)\nNote: This Figure represents the data of the included participants in the main statistical analysis after omitting missing values.\nThe number of observed values (%) of each categorical variable was presented by the PHQ-8 categories in Table 1, where the majority of the study participants (90.5%) had PHQ-8 score less than 10. When categorical groups were compared within the PHQ-8 depression group (< 10 versus ≥ 10), there was only a statistically significant difference between females versus males with a Fisher exact p-value = 0.028. Table 1 shows that there was a statistically significant difference in the age distribution across the depression groups (p-value = 0.012). There was no statistically significant difference in the BMI measurements between the PHQ-8 groups (p-value = 0.44).\nThe result of the primary analysis showed that females have statistically significantly greater odds of reporting depression compared to males OR = 3.2 (95% CI:1.1, 8.9), p-value = 0.024 (Table 2). Furthermore, there was an approximately 5-fold greater odds of reporting depression for unhappy than for happy individuals. However, this was not statistically significant (p-value = 0.10). Similarly, the sleep duration and marital status were both not statistically significant (p-value of 0.284 and 0.205, respectively; Table 2). However, in a sensitivity analysis, people who reported sleep duration of less than 6 h were more likely to report depression as compared to the people who reported sleep of 7 h OR (95% CI) of 2.6 (1.0, 6.4), p-value = 0.040 and 1.136 (1.015, 1.272), p-value = 0.027.\n\nTable 2\nThe results of the primary analysis of the fitted multivariate logistic regression model\nVariableOR (95% CI)PbHappiness - HappyReferenceHappiness - Unhappy5.5 (0.7, 42.2)0.100Age (linear)0.3 (0.1, 1.0)a0.056BMI (linear)1.8 (1.0, 3.3)a0.064Gender = MalesReferenceGender = Females3.2 (1.2, 8.9)0.024Sleep = 7ReferenceSleep < 64.0 (0.9, 17.4)0.061Sleep = 61.3 (0.2, 6.9)0.775Sleep = 81.5 (0.3, 7.3)0.585Sleep > 81.2 (0.2, 9.2)0.848MarriedReferenceSingle0.9 (0.3, 3.0)0.8Others0.2 (0.02, 3.3)0.3Note: aInterquartile-range odds ratio for age and BMI, which compares the 3rd quartile with the 1st quartile, and simple odds ratios for the categorical predictors (happiness, gender, sleep and marital status) compare each group with the reference group (the largest group) with corresponding 95% confidence interval (95% CI). bWald’s p-values are presented for each variable\n\n\nThe results of the primary analysis of the fitted multivariate logistic regression model\n\nNote: aInterquartile-range odds ratio for age and BMI, which compares the 3rd quartile with the 1st quartile, and simple odds ratios for the categorical predictors (happiness, gender, sleep and marital status) compare each group with the reference group (the largest group) with corresponding 95% confidence interval (95% CI). bWald’s p-values are presented for each variable\nTo compute Interquartile Range (IQR) odds ratios for continuous variables, the age and BMI variables were divided by their IQR values of 15 and 8.7, respectively [54, 56]. Table 2 shows that for one interquartile-range increase in the age and BMI, the IQR-OR was 0.34 (95% CI: 0.12, 1.0) and 1.8 (95% CI: 0.97, 3.3), respectively, where both results were statistically significant with a p-value of 0.056 and 0.064 individually.\nThe results of the sensitivity analysis using 100 multiple imputations (Table 3) were approximately similar to the result of the multivariate logistic ordinal regression analyses using omitted data (Table 2). However, the happiness variable was statistically significant OR = 5.1 (95%CI: 1.7, 15.7, p-value = 0.005), and there was a statistically significant difference between the sleep variable for less than six hours as compared with seven hours OR = 2.6 (95% CI: 1.0, 6.4, p-value = 0.04). Supplementary Fig. 1 illustrates the percentages of missing values in the variables included in this statistical analysis. Marital status had the lowest number of missing values (5.3%), followed by BMI (12.7%) and the Sleep variable with 12.9% missing values. The Happiness variable had 21.1% missing values. The PHQ-8 variables had 32.6–35.1% missing values. Supplementary Table 1 shows the percentages of the “Prefer not to answer (DA)” and “Do not know (UN)” in the eight PHQ questions, where the percentages vary between 16 and 18.5% between the eight questions. The percentages of “Prefer not to answer (DA)” and “Do not know (UN)” in the happiness variable was 2.5% and 2.3%, respectively, which were also considered missing values, although these were not missing at random and can be correlated with one of the PHQ-8 answers. However, this has been addressed in the multiple imputation analysis.\n\nTable 3\nResults of the sensitivity analysis using multivariate logistic regression models with a sample size N = 487\nVariableOR (95% CI)PbHappiness – HappyReferenceHappiness - Unhappy5.1 (1.7, 15.7)0.004Age (linear)0.5 (0.3, 1.0)a0.060BMI (linear)1.7 (1.1, 2.5)a0.021Gender = MalesReferenceGender = Females1.6 (0.9, 3.0)0.154Sleep = 7ReferenceSleep < 62.6 (1.0, 6.4)0.040Sleep = 60.9 (0.3, 2.6)0.887Sleep = 80.9 (0.3, 2.4)0.779Sleep > 81.1 (0.3, 3.7)0.853MarriedReferenceSingle0.9 (0.4, 2.1)0.885Others0.7 (0.2, 2.9)0.597Note: aInterquartile-range odds ratio for Age and BMI, which compares the 3rd quartile with the 1st quartile, and simple odds ratios for the categorical predictors (happiness, gender, sleep and marital status) compare each group with the reference group (the largest group) with corresponding 95% confidence interval (95% CI). bWald’s p-values are presented for each variable. Results were summarized using Rubin’s rules\n\n\nResults of the sensitivity analysis using multivariate logistic regression models with a sample size N = 487\n\nNote: aInterquartile-range odds ratio for Age and BMI, which compares the 3rd quartile with the 1st quartile, and simple odds ratios for the categorical predictors (happiness, gender, sleep and marital status) compare each group with the reference group (the largest group) with corresponding 95% confidence interval (95% CI). bWald’s p-values are presented for each variable. Results were summarized using Rubin’s rules\nAn additional sensitivity analysis was performed using the ordinal PHQ-8 score as an outcome in a multivariate linear regression model (see supplementary Table 1). The result of this sensitivity analysis was very similar to the sensitivity analysis in Table 3.", "Overall, there is a lack of quantitative research examining the relationship between depression, perceived happiness, and sleep duration and other confounding factors in the Gulf region [35,46, 36, 49, 60]. The UAE Healthy Future Study is the first prospective cohort study of the UAE population and one of the few studies in the region which examines such relationships between happiness, sleep duration and quality, and depression using PHQ-8. The evidence collected from this study confirms what has been published in the literature. For instance, in this study, it has been found that males were less likely to report depression symptoms than females, which is similar to what has been documented by other studies [38, 41]; where women reported more depressive symptoms than men [60]. In addition, the results of this study revealed that older people have a lower odds of reporting depression as compared with younger people, suggesting a possible protective age effect.\nThis study has used the pilot data of the UAE Healthy Future Study, as recruitment into the main cohort study is still ongoing. However, we plan to use the main UAEHFS data to confirm the results and provide further understanding of these findings.\nThe association between sleep duration and depression has been intensively studied in the literature [23, 24]. The findings of this study revealed that individuals who reported less than 6 h of sleep per 24 h were more likely to report being depressed compared to those who reported 7 h of sleep. This is similar to what has been reported in the literature that participants who reported insufficient sleep showed a 62–179% increase in the prevalence of depression versus those sleeping 6 to 8 h per day and reporting sufficient sleep (P < 0.05) [61, 62].\nFurthermore, self-reported happiness has been identified as a potential protective and management factor for depression [27]. The result of this study shows that unhappy individuals have approximately 5-fold higher odds of reporting depression compared to happy individuals, and this aligns with what has been found in the literature [27].\nAdditionally, a small number of studies found that insufficient sleep is associated with lower happiness in healthy adults using a self-reported questionnaire as a single item for measuring happiness [22]. Moreover, some longitudinal studies have found that the next-day happiness is lowered following a shorter sleep duration [63]. However, the associations between sleep and happiness have not been well-explored in adults. Our findings will add to the available evidence and will help to bring novel data from the Gulf Cooperation Council (GCC) countries about the association between sleep duration, self-reported happiness, and depression.\nA further finding is that marital status can contribute to health and self-reported happiness in a bidirectional way [49]. Several studies have found that there is an association between depression and marital status [63, 64]. There is an increased risk of reporting depression for divorced and separated people. It is frequently asserted that marriage is more beneficial for the mental health of men than women, but the evidence for this is far from clear-cut [65]. Single people have a higher level of depression as compared to married people and some studies have found that married people have a better mood than single people considering factors of age, gender, and education level [37, 40, 41, 66]. Research does not yet clarify whether gender differences in the prevalence of anxiety-mood disorders are greater among the married than the never-married or the previously married [65]. However, the mechanisms underlying the relationship between depression and marital status are not yet entirely clear and require further exploration.\nAn association has been reported between BMI categories and depression [67, 68]. Higher BMI is a risk factor for the likelihood of developing depression in individuals [69]. Underweight increases the risk of depression as well [70]. Our study considered the BMI factor in the data analysis of this study. This statistical analysis shows that as BMI increases, the odds of reporting depression also increase. This result is comparable to other research finding that participants with central obesity had an increased chance of depression [70,71,72].\nThe UAEHFS is a unique cohort study in the UAE and Gulf region as it allows researchers to investigate the association between disease outcomes and related risk factors [58]. In this study, we investigated the association between depression and sleep duration, self-reported happiness, BMI, and sociodemographic status using the UAEHFS pilot data, which presents a population that has not been studied. The result of our study needs to be confirmed in the main UAEHFS data.", "Missing values were omitted in the primary statistical analysis, which decreased the sample size and can lead to overfitting in the main finding [55]. Therefore, the number of participants with the PHQ-8 ≥ 10 (i.e. – events) was 22 which is a limitation in this data set because it does not allow us to fit a complex multivariate statistical regression model if we would follow the statistical rule of thumb “ten events per predictor” [56, 57, 73]. Thus, sensitivity analysis was performed, and the result of the sensitivity analysis (Table 3) was approximately the same as the main finding (Table 2).\nAlthough some limitations were found in this study, the findings provide future direction to mental health research. Further investigation is needed with a larger sample size using the main UAEHFS data to have a better picture of the role of happiness, marital status, sleep, and social demographic variables in association with depression and mental health disorders.", "The results of this study indicate that females are more likely to report depression compared to males. Older age is associated with a decrease in self-reported depression. Unhappy individuals are approximately 5-fold higher odds to report depression compared to happy individuals. BMI was positively associated with reporting depression. The result of the sensitivity analysis shows that individuals who sleep less than 6 h per day are more likely to report depression compared to those who sleep 7 h per day.\nThe results of this study have a potential value for researchers and public health professionals as it presents novel data on the PHQ-8 score in a healthy UAE population, which has not been explored before. Furthermore, the results of this study can help contribute to the knowledge base on current and potential population mental health impact in the UAE and Gulf Region.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ "introduction", "materials|methods", null, null, null, null, null, "results", "discussion", null, "conclusion", "supplementary-material", null ]
[ "PHQ-8", "Depression", "Sleep duration", "Happiness", "Self-reported happiness", "Sociodemographic and marital status" ]
Insecticidal effects of some selected plant extracts against Anopheles stephensi (Culicidae: Diptera).
36271447
The use of synthetic insecticides against mosquitoes may lead to resistance development and potential health hazards in humans and the environment. Consequently, a paradigm needs to shift towards the alternative use of botanical insecticides that could strengthen an insecticide resistance management programme. This study aimed to assess the insecticidal effects aqueous, hexane, and methanol crude leaf extracts of Calpurnia aurea, Momordica foetida, and Zehneria scabra on an insectary colony of Anopheles stephensi larvae and adults.
BACKGROUND
Fresh leaves of C. aurea, M. foetida and Z. scabra were collected and dried, then separately ground to powder. Powdered leaves of test plants were extracted using sonication with aqueous, hexane, and methanol solvents. The extracts were concentrated, and a stock solution was prepared. For comparison, Temephos (Abate®) and control solutions (a mixture of water and emulsifier) were used as the positive and negative controls, respectively. Different test concentrations for the larvae and the adults were prepared and tested according to WHO (2005) and CDC (2010) guidelines to determine lethal concentration (LC) values. Mortality was observed after 24 h exposure. The statistical analyses were performed using Statistical Package for the Social Sciences (SPSS) software (Kruskal-Wallis test) and R software (a generalized linear model was used to determine LC50 and LC90 values of the extracts).
METHODS
The lowest LC50 values were observed in aqueous extracts of M. foetida followed by Z. scabra extract and C. aurea leaves at 34.61, 35.85, and 38.69 ppm, respectively, against the larvae. Larval mortality was not observed from the hexane extracts and negative control, while the standard larvicide (temephos) achieved 100% mortality. Further, the adulticidal efficacy was greatest for aqueous extract of Z. scabra with LC50 = 176.20 ppm followed by aqueous extract of C. aurea (LC50 = 297.75 ppm).
RESULTS
The results suggest that the leaf extracts of the three test plants have the potential of being used for the control of vector An. stephensi larvae and adult instead of synthetic mosquitocides. Further studies need to be conducted to identify the active ingredients and their mode of action.
CONCLUSION
[ "Humans", "Animals", "Anopheles", "Insecticides", "Culicidae", "Hexanes", "Culex", "Aedes", "Temefos", "Methanol", "Powders", "Mosquito Vectors", "Larva", "Plant Extracts", "Solvents", "Water", "Plant Leaves" ]
9585825
null
null
null
null
Results
[SUBTITLE] Larvicidal activity of plant extracts against An. stephensi [SUBSECTION] Larval mortalities obtained from bioassays of aqueous and methanol extracts of Calpurnia aurea, Momordica foetida, and Zehneria scabra leaves against An. stephensi larvae after 24 h exposure periods are presented in Tables 1, 2 and 3. All aqueous and methanol test plant extracts showed low, moderate, and high larvicidal activities tested between 25 ppm-300 ppm treatments against the late 3rd to early 4th instar larvae of An. stephensi. Within the same exposure period, larval mortality was not observed for the tests using hexane crude leaf extracts and the negative control. The standard positive control (temephos) achieved 100% larval mortality. Table 1Mean % larval mortality of Calpurnia aurea crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE Solvents Aqueous Methanol 2535.00 ± 2.6Aa23.75 ± 3.65Aa5060.00 ± 3.54Ba35.00 ± 0.00Bb10086.25 ± 2.23Ca90.42 ± 1.4Ca15099.58 ± 0.42Da95.00 ± 2.46DCa200100.00 ± 0.00Da96.67 ± 1.42Da250100.00 ± 0.00Da100.00 ± 0.00Da300100.00 ± 0.00Da100.00 ± 0.00DaTemephos (0.25)100.00 ± 0.00Da100.00 ± 0.00DaControl0.00 ± 0.00Ea0.00 ± 0.00Ea*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p > 0.003) Mean % larval mortality of Calpurnia aurea crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure *Each value of % mean mortality ± SE represents the value of twelve replicates *Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p > 0.003) There was a significant difference and interaction effects between all aqueous and methanol test plant extract against the tested larvae (Kruskal-Wallis test, p-value < 0.05). The mean percent larval mortality between treatments, control, and the standard groups had a statistically significant difference (Kruskal-Wallis test, p-value < 0.05). The mortality effect of the test plant extracts against An. stephensi larvae were dose and extraction solvent dependent. In the C. aurea test plant, the highest (100%) larval mortality was found both in aqueous and methanol extracts at 200 ppm and 250 ppm of treatments, respectively (Table 1). Even at 150 ppm, the larvicidal effects of aqueous and methanol extracts of C. aurea were not significantly different from standard checks (Multiple-Mann Whitney U test, p-value > 0.003; Table 1). Yet, all C. aurea treatments significantly affected larval mortality compared to the negative control (Multiple-Mann Whitney U test; p < 0.003). Aqueous and methanol extracts of the M. foetida test plant showed 100% larval mortality at 200 ppm and 250 ppm of treatments, respectively (Table 2). Moreover, larvicidal activity found from aqueous extracts of M. foetida at 100 ppm and methanol extracts at 200 ppm caused mortality (ranging 95–97.9%) was not significantly different from the mortality achieved in temephos exposed on An. stephensi larvae (Multiple-Mann Whitney U test, p-value > 0.003; Table 2). Statistical differences were observed between all M. foetida treatments and negative control (Multiple-Mann Whitney U test; p-value < 0.003). Table 2Mean % larval mortality of Momordica foetida crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE Solvents Aqueous Methanol 2527.5 ± 2.85Aa4.55 ± 1.4Ab5080.83 ± 4.51Ba20.83 ± 2.53Bb10095.00 ± 1.74Ca45.83 ± 2.20Cb15097.92 ± 0.96Ca71.67 ± 3.39Db200100.00 ± 0.00Ca97.5 ± 2.50Ea250100.00 ± 0.00Ca100.00 ± 0.00Ea300100.00 ± 0.00Ca100.00 ± 0.00EaTemephos (0.25)100.00 ± 0.00Ca100.00 ± 0.00EaControl0.00 ± 0.00Da0.00 ± 0.00Fa*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)Table 3Mean % larval mortality of Zehneria scabra crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE Solvents Aqueous Methanol 2529.17 ± 3.68Aa23.75 ± 2.69Aa5073.33 ± 3.9Ba66.25 ± 5.04Ba10095.42 ± 1.44Ca87.08 ± 2.17Ca15097.08 ± 1.44Ca92.50 ± 1.44Da20099.50 ± 0.42Ca99.17 ± 0.83Ea250100.00 ± 0.00Ca100.00 ± 0.00Ea300100.00 ± 0.00Ca100.00 ± 0.00EaTemephos (0.25)100.00 ± 0.00Ca100.00 ± 0.00EaControl0.00 ± 0.00Da0.00 ± 0.00Fa*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Mean % larval mortality of Momordica foetida crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure *Each value of % mean mortality ± SE represents the value of twelve replicates *Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Mean % larval mortality of Zehneria scabra crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure *Each value of % mean mortality ± SE represents the value of twelve replicates *Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Among the two solvent test concentrations of Z. scabra extracts, statistically, non-significant differences were observed (Multiple-Mann Whitney U test, p-value > 0.003; Table 3). However, their larvicidal activity was significantly different from negative control (Multiple-Mann Whitney U test; p-value < 0.003). At the highest concentrations (250 ppm), both aqueous and methanol Z. scabra, crude leaf extracts resulted in 100% larval mortality (Table 3). Moreover, aqueous extracts of Z. scabra at 100 ppm, and methanol extracts at 200 ppm, each caused more than 95% larval mortality, had no significant effect compared to mortality achieved in temephos treatment (Multiple-Mann Whitney U test; p-value > 0.003;). The lethal toxicity doses of aqueous and methanol crude leaf extracts of C. aurea, M. foetida, and Z. scabra against larvae of An. stephensi are shown in Table 4. Crude aqueous leaf extract of M. foetida showed strong larvicidal activity, having an LC50 value of 34.61 ppm and LC90 value of 57.61 ppm, followed by Z. scabra (LC50 = 35.85 ppm; LC90 = 68.26 ppm) and C. aurea (LC50 = 38.69 ppm; LC90 = 108.28 ppm). Similarly, the methanol leaf extract of Z. scabra had good larvicidal activities with LC50 and LC90 values of 41.32 ppm and 99.07 ppm, followed by C. aurea (LC50 = 43.25 ppm; LC90 = 96.02 ppm). However, the crude methanol leaf extract of M. foetida had lower larvicidal toxicity against the larvae of An. stephensi compared to other test plant extracts (LC50 = 99.50 ppm; LC90 = 188.76 ppm, Table 4). Table 4LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of tests plants against the late third to early fourth instar larvae of Anopheles stephensiTest plantsSolventsLC50 (ppm)95%Confidence limitLC90 (ppm)95%Confidence limitχ2 (df = 5) LCL UCL LCL UCL C. aureaAqueous38. 6935. 5741.82108. 2896. 27120. 302.93Methanol43. 2540. 4946. 0296. 0283. 87108. 184.15 M. foetidaAqueous34. 6132. 8436. 3857. 6153. 2961. 930.71Methanol99. 5094. 56104. 43188. 76177. 70199. 826.90 Z. scabraAqueous35. 8533. 7937. 9168. 2660. 2076. 320.37Methanol41. 3238. 5144. 1299. 0784. 64113. 501.43 LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of tests plants against the late third to early fourth instar larvae of Anopheles stephensi Larval mortalities obtained from bioassays of aqueous and methanol extracts of Calpurnia aurea, Momordica foetida, and Zehneria scabra leaves against An. stephensi larvae after 24 h exposure periods are presented in Tables 1, 2 and 3. All aqueous and methanol test plant extracts showed low, moderate, and high larvicidal activities tested between 25 ppm-300 ppm treatments against the late 3rd to early 4th instar larvae of An. stephensi. Within the same exposure period, larval mortality was not observed for the tests using hexane crude leaf extracts and the negative control. The standard positive control (temephos) achieved 100% larval mortality. Table 1Mean % larval mortality of Calpurnia aurea crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE Solvents Aqueous Methanol 2535.00 ± 2.6Aa23.75 ± 3.65Aa5060.00 ± 3.54Ba35.00 ± 0.00Bb10086.25 ± 2.23Ca90.42 ± 1.4Ca15099.58 ± 0.42Da95.00 ± 2.46DCa200100.00 ± 0.00Da96.67 ± 1.42Da250100.00 ± 0.00Da100.00 ± 0.00Da300100.00 ± 0.00Da100.00 ± 0.00DaTemephos (0.25)100.00 ± 0.00Da100.00 ± 0.00DaControl0.00 ± 0.00Ea0.00 ± 0.00Ea*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p > 0.003) Mean % larval mortality of Calpurnia aurea crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure *Each value of % mean mortality ± SE represents the value of twelve replicates *Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p > 0.003) There was a significant difference and interaction effects between all aqueous and methanol test plant extract against the tested larvae (Kruskal-Wallis test, p-value < 0.05). The mean percent larval mortality between treatments, control, and the standard groups had a statistically significant difference (Kruskal-Wallis test, p-value < 0.05). The mortality effect of the test plant extracts against An. stephensi larvae were dose and extraction solvent dependent. In the C. aurea test plant, the highest (100%) larval mortality was found both in aqueous and methanol extracts at 200 ppm and 250 ppm of treatments, respectively (Table 1). Even at 150 ppm, the larvicidal effects of aqueous and methanol extracts of C. aurea were not significantly different from standard checks (Multiple-Mann Whitney U test, p-value > 0.003; Table 1). Yet, all C. aurea treatments significantly affected larval mortality compared to the negative control (Multiple-Mann Whitney U test; p < 0.003). Aqueous and methanol extracts of the M. foetida test plant showed 100% larval mortality at 200 ppm and 250 ppm of treatments, respectively (Table 2). Moreover, larvicidal activity found from aqueous extracts of M. foetida at 100 ppm and methanol extracts at 200 ppm caused mortality (ranging 95–97.9%) was not significantly different from the mortality achieved in temephos exposed on An. stephensi larvae (Multiple-Mann Whitney U test, p-value > 0.003; Table 2). Statistical differences were observed between all M. foetida treatments and negative control (Multiple-Mann Whitney U test; p-value < 0.003). Table 2Mean % larval mortality of Momordica foetida crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE Solvents Aqueous Methanol 2527.5 ± 2.85Aa4.55 ± 1.4Ab5080.83 ± 4.51Ba20.83 ± 2.53Bb10095.00 ± 1.74Ca45.83 ± 2.20Cb15097.92 ± 0.96Ca71.67 ± 3.39Db200100.00 ± 0.00Ca97.5 ± 2.50Ea250100.00 ± 0.00Ca100.00 ± 0.00Ea300100.00 ± 0.00Ca100.00 ± 0.00EaTemephos (0.25)100.00 ± 0.00Ca100.00 ± 0.00EaControl0.00 ± 0.00Da0.00 ± 0.00Fa*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)Table 3Mean % larval mortality of Zehneria scabra crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE Solvents Aqueous Methanol 2529.17 ± 3.68Aa23.75 ± 2.69Aa5073.33 ± 3.9Ba66.25 ± 5.04Ba10095.42 ± 1.44Ca87.08 ± 2.17Ca15097.08 ± 1.44Ca92.50 ± 1.44Da20099.50 ± 0.42Ca99.17 ± 0.83Ea250100.00 ± 0.00Ca100.00 ± 0.00Ea300100.00 ± 0.00Ca100.00 ± 0.00EaTemephos (0.25)100.00 ± 0.00Ca100.00 ± 0.00EaControl0.00 ± 0.00Da0.00 ± 0.00Fa*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Mean % larval mortality of Momordica foetida crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure *Each value of % mean mortality ± SE represents the value of twelve replicates *Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Mean % larval mortality of Zehneria scabra crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure *Each value of % mean mortality ± SE represents the value of twelve replicates *Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Among the two solvent test concentrations of Z. scabra extracts, statistically, non-significant differences were observed (Multiple-Mann Whitney U test, p-value > 0.003; Table 3). However, their larvicidal activity was significantly different from negative control (Multiple-Mann Whitney U test; p-value < 0.003). At the highest concentrations (250 ppm), both aqueous and methanol Z. scabra, crude leaf extracts resulted in 100% larval mortality (Table 3). Moreover, aqueous extracts of Z. scabra at 100 ppm, and methanol extracts at 200 ppm, each caused more than 95% larval mortality, had no significant effect compared to mortality achieved in temephos treatment (Multiple-Mann Whitney U test; p-value > 0.003;). The lethal toxicity doses of aqueous and methanol crude leaf extracts of C. aurea, M. foetida, and Z. scabra against larvae of An. stephensi are shown in Table 4. Crude aqueous leaf extract of M. foetida showed strong larvicidal activity, having an LC50 value of 34.61 ppm and LC90 value of 57.61 ppm, followed by Z. scabra (LC50 = 35.85 ppm; LC90 = 68.26 ppm) and C. aurea (LC50 = 38.69 ppm; LC90 = 108.28 ppm). Similarly, the methanol leaf extract of Z. scabra had good larvicidal activities with LC50 and LC90 values of 41.32 ppm and 99.07 ppm, followed by C. aurea (LC50 = 43.25 ppm; LC90 = 96.02 ppm). However, the crude methanol leaf extract of M. foetida had lower larvicidal toxicity against the larvae of An. stephensi compared to other test plant extracts (LC50 = 99.50 ppm; LC90 = 188.76 ppm, Table 4). Table 4LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of tests plants against the late third to early fourth instar larvae of Anopheles stephensiTest plantsSolventsLC50 (ppm)95%Confidence limitLC90 (ppm)95%Confidence limitχ2 (df = 5) LCL UCL LCL UCL C. aureaAqueous38. 6935. 5741.82108. 2896. 27120. 302.93Methanol43. 2540. 4946. 0296. 0283. 87108. 184.15 M. foetidaAqueous34. 6132. 8436. 3857. 6153. 2961. 930.71Methanol99. 5094. 56104. 43188. 76177. 70199. 826.90 Z. scabraAqueous35. 8533. 7937. 9168. 2660. 2076. 320.37Methanol41. 3238. 5144. 1299. 0784. 64113. 501.43 LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of tests plants against the late third to early fourth instar larvae of Anopheles stephensi [SUBTITLE] The adulticidal activity of plant extracts against An. stephensi [SUBSECTION] Tables 5, 6 and 7 show the mean percentage mortalities and toxicity effects of adult An. stephensi after 24 h exposures to different solvent extracts of C. aurea, M. foetida, and Z. scabra. Statistically significant differences in mean percentage mortality among the different extracts and the negative control were observed (Kruskal-Wallis test, p < 0.05). The adulticidal data showed that the mortality rate of the vector was directly proportional to the concentration and extraction solvents. All hexane test plant extracts showed lower adulticidal property against An. stephensi (≤ 20% at all treatments tested in the bioassays, Tables 5, 6 and 7). Between the different solvent leaf extracts of C. aurea, only aqueous extract gave 55% adult mortality of An. stephensi at 320 ppm, while other crude solvent leaf extracts had lower adulticidal activity, having adult mean mortalities ranging from 0 to 48% (Table 5). Table 5Mean % adult mortality of Calpurnia aurea crude leaf solvent extracts at different treatments against An. stephensiConcentration (ppm)% Mean mortality ± SE Solvents Aqueous Methanol Hexane 201.67 ± 1.67Aa0.00 ± 0.00Aa0.00 ± 0.00Aa406.67 ± 1.67Aa6.67 ± 1.67Ba0.00 ± 0.00Ab8016.67 ± 1.67Ba16.67 ± 1.67Ca0.00 ± 0.00Ab16021.67 ± 1.67Ba26.67 ± 3.33Da6.67 ± 3.33Bb32055.00 ± 5.77Ca48.33 ± 1.67Ea18.33 ± 1.67CbControl0.00 ± 0.00Da0.00 ± 0.00Aa0.00 ± 0.00Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Mean % adult mortality of Calpurnia aurea crude leaf solvent extracts at different treatments against An. stephensi *Each value of % mean mortality ± SE represents the value of three replicates *Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Different solvent (aqueous, methanol, and hexane) crude leaf extracts of M. foetida resulted in lower adult mortalities of An. stephensi (< 24%) at all treatments tested in the bioassays and were not effective against adults of malaria vector An. stephensi (Table 6). Table 6Mean % adult mortality of Momordica foetida crude leaf solvent extract at different concentrations against Anopheles stephensiConcentration(ppm)% Mean mortality ± SE Solvents Aqueous Methanol Hexane 200.00 ± 0.00ABa0.00 ± 0.00Aa0.00 ± 0.00Aa401.67 ± 1.67Ba0.00 ± 0.00Aa0.00 ± 0.00Ab805.00 ± 2.89Ba1.67 ± 1.67Ba0.00 ± 0.00Ab16013.33 ± 1.67Ca13.33 ± 1.67Ca0.00 ± 0.00Ab32018.33 ± 4.41Ca23.33 ± 1.67Da1.67 ± 1.67BbControl0.00 ± 0.00Aa0.00 ± 0.00Aa0.00 ± 0.00Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Mean % adult mortality of Momordica foetida crude leaf solvent extract at different concentrations against Anopheles stephensi *Each value of % mean mortality ± SE represents the value of three replicates *Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) There were statistically significant differences in mean percentage mortality among different concentrations of aqueous, methanol, and hexane leaf extracts of Z. scabra and with negative control (Kruskal-Wallis test; p-value < 0.05; Table 7). Within the given time intervals adult mortality was recorded for the control treatment. However, the highest adult mortality (84%) at higher concentration 320 ppm against An. stephensi was recorded in aqueous crude leaf extract of Z. scabra followed by its methanol extracts (65%) and their adulticidal potential was statistically different from the negative control (Multiple-Mann Whitney U-test; p-value < 0.003, Table 7).Table 7Mean % adult mortality of Zehneria scabra crude leaf solvent extracts at different concentrations against Anopheles stephensiConcentration (ppm)% Mean mortality ± SE Solvents Aqueous Methanol Hexane 204.33 ± 2.6Aa11.67 ± 3.33Aa0.00 ± 0.00Aa4011.67 ± 1.67Ba20.00 ± 2.89ABa0.00 ± 0.00Ab8026.67 ± 3.33Ca23.33 ± 3.33Ba0.00 ± 0.00Ab16036.67 ± 6.00Ca41.66 ± 4.40Ca15.00 ± 2.89Bb32084.00 ± 2.89Da65.00 ± 5.77Db20.00 ± 2.89BcControl0.67 ± 0.45Ea0.67 ± 0.45Ea0.67 ± 0.45Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Mean % adult mortality of Zehneria scabra crude leaf solvent extracts at different concentrations against Anopheles stephensi *Each value of % mean mortality ± SE represents the value of three replicates *Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) The lethal effects of different crude leaf extracts against adults of An. stephensi are shown in Table 8. All hexane and M. foetida crude leaf extracts had low adulticidal impacts, only aqueous and methanol C. aurea and Z. scabra leaf extracts were subjected to dose-response bioassay to detect the lethal concentrations. From the dose-response curves, the lowest LC50 (= 176.20 ppm and 205.41 ppm) and LC90 (= 425.13 and 883.11 ppm) values were demonstrated from the aqueous and methanol Z. scabra crude leaf extracts, respectively (Table 8), and its relative toxicities against adult An. stephensi was much higher than the rest of the plant extracts. The highest LC50 (= 297.75 ppm and 333.27 ppm) and LC90 (= 762.63 ppm and 1031.74 ppm) values were from the aqueous and methanol C. aurea crude leaf extracts, respectively, and its relative toxicity on An. stephensi was much lower than Z. scabra crude extracts. Table 8LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of Calpurnia aurea and Zehneria scabra against adults of Anopheles stephensiTest plantsSolventsLC5095%Confidence limitLC9095%Confidence limitχ2 (df = 3) LCL UCL LCL UCL C. aureaAqueous297. 75254. 95340. 56762. 63491. 131034. 130.88Methanol333. 27268. 92397. 631031. 74569. 061494. 430.75 Z. scabraAqueous176.20157. 82194. 58425. 13346. 14504. 122.70Methanol205. 41172. 29238. 54883. 11529. 331236. 900.70 LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of Calpurnia aurea and Zehneria scabra against adults of Anopheles stephensi Tables 5, 6 and 7 show the mean percentage mortalities and toxicity effects of adult An. stephensi after 24 h exposures to different solvent extracts of C. aurea, M. foetida, and Z. scabra. Statistically significant differences in mean percentage mortality among the different extracts and the negative control were observed (Kruskal-Wallis test, p < 0.05). The adulticidal data showed that the mortality rate of the vector was directly proportional to the concentration and extraction solvents. All hexane test plant extracts showed lower adulticidal property against An. stephensi (≤ 20% at all treatments tested in the bioassays, Tables 5, 6 and 7). Between the different solvent leaf extracts of C. aurea, only aqueous extract gave 55% adult mortality of An. stephensi at 320 ppm, while other crude solvent leaf extracts had lower adulticidal activity, having adult mean mortalities ranging from 0 to 48% (Table 5). Table 5Mean % adult mortality of Calpurnia aurea crude leaf solvent extracts at different treatments against An. stephensiConcentration (ppm)% Mean mortality ± SE Solvents Aqueous Methanol Hexane 201.67 ± 1.67Aa0.00 ± 0.00Aa0.00 ± 0.00Aa406.67 ± 1.67Aa6.67 ± 1.67Ba0.00 ± 0.00Ab8016.67 ± 1.67Ba16.67 ± 1.67Ca0.00 ± 0.00Ab16021.67 ± 1.67Ba26.67 ± 3.33Da6.67 ± 3.33Bb32055.00 ± 5.77Ca48.33 ± 1.67Ea18.33 ± 1.67CbControl0.00 ± 0.00Da0.00 ± 0.00Aa0.00 ± 0.00Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Mean % adult mortality of Calpurnia aurea crude leaf solvent extracts at different treatments against An. stephensi *Each value of % mean mortality ± SE represents the value of three replicates *Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Different solvent (aqueous, methanol, and hexane) crude leaf extracts of M. foetida resulted in lower adult mortalities of An. stephensi (< 24%) at all treatments tested in the bioassays and were not effective against adults of malaria vector An. stephensi (Table 6). Table 6Mean % adult mortality of Momordica foetida crude leaf solvent extract at different concentrations against Anopheles stephensiConcentration(ppm)% Mean mortality ± SE Solvents Aqueous Methanol Hexane 200.00 ± 0.00ABa0.00 ± 0.00Aa0.00 ± 0.00Aa401.67 ± 1.67Ba0.00 ± 0.00Aa0.00 ± 0.00Ab805.00 ± 2.89Ba1.67 ± 1.67Ba0.00 ± 0.00Ab16013.33 ± 1.67Ca13.33 ± 1.67Ca0.00 ± 0.00Ab32018.33 ± 4.41Ca23.33 ± 1.67Da1.67 ± 1.67BbControl0.00 ± 0.00Aa0.00 ± 0.00Aa0.00 ± 0.00Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Mean % adult mortality of Momordica foetida crude leaf solvent extract at different concentrations against Anopheles stephensi *Each value of % mean mortality ± SE represents the value of three replicates *Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) There were statistically significant differences in mean percentage mortality among different concentrations of aqueous, methanol, and hexane leaf extracts of Z. scabra and with negative control (Kruskal-Wallis test; p-value < 0.05; Table 7). Within the given time intervals adult mortality was recorded for the control treatment. However, the highest adult mortality (84%) at higher concentration 320 ppm against An. stephensi was recorded in aqueous crude leaf extract of Z. scabra followed by its methanol extracts (65%) and their adulticidal potential was statistically different from the negative control (Multiple-Mann Whitney U-test; p-value < 0.003, Table 7).Table 7Mean % adult mortality of Zehneria scabra crude leaf solvent extracts at different concentrations against Anopheles stephensiConcentration (ppm)% Mean mortality ± SE Solvents Aqueous Methanol Hexane 204.33 ± 2.6Aa11.67 ± 3.33Aa0.00 ± 0.00Aa4011.67 ± 1.67Ba20.00 ± 2.89ABa0.00 ± 0.00Ab8026.67 ± 3.33Ca23.33 ± 3.33Ba0.00 ± 0.00Ab16036.67 ± 6.00Ca41.66 ± 4.40Ca15.00 ± 2.89Bb32084.00 ± 2.89Da65.00 ± 5.77Db20.00 ± 2.89BcControl0.67 ± 0.45Ea0.67 ± 0.45Ea0.67 ± 0.45Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) Mean % adult mortality of Zehneria scabra crude leaf solvent extracts at different concentrations against Anopheles stephensi *Each value of % mean mortality ± SE represents the value of three replicates *Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003) The lethal effects of different crude leaf extracts against adults of An. stephensi are shown in Table 8. All hexane and M. foetida crude leaf extracts had low adulticidal impacts, only aqueous and methanol C. aurea and Z. scabra leaf extracts were subjected to dose-response bioassay to detect the lethal concentrations. From the dose-response curves, the lowest LC50 (= 176.20 ppm and 205.41 ppm) and LC90 (= 425.13 and 883.11 ppm) values were demonstrated from the aqueous and methanol Z. scabra crude leaf extracts, respectively (Table 8), and its relative toxicities against adult An. stephensi was much higher than the rest of the plant extracts. The highest LC50 (= 297.75 ppm and 333.27 ppm) and LC90 (= 762.63 ppm and 1031.74 ppm) values were from the aqueous and methanol C. aurea crude leaf extracts, respectively, and its relative toxicity on An. stephensi was much lower than Z. scabra crude extracts. Table 8LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of Calpurnia aurea and Zehneria scabra against adults of Anopheles stephensiTest plantsSolventsLC5095%Confidence limitLC9095%Confidence limitχ2 (df = 3) LCL UCL LCL UCL C. aureaAqueous297. 75254. 95340. 56762. 63491. 131034. 130.88Methanol333. 27268. 92397. 631031. 74569. 061494. 430.75 Z. scabraAqueous176.20157. 82194. 58425. 13346. 14504. 122.70Methanol205. 41172. 29238. 54883. 11529. 331236. 900.70 LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of Calpurnia aurea and Zehneria scabra against adults of Anopheles stephensi
Conclusion
This study showed that crude aqueous and methanol extracts of C. aurea, M. foetida, and Z. scabra leaves could be considered as foreseeable products to be developed as potential larvicides against An. stephensi larvae. In addition, crude leaf extracts of C. aurea and Z. scabra are potential candidate insecticides against the adults. These three plants could be used to develop effective, safe, biodegradable, and cheap botanical insecticides for vector control, potentially leading to improved resistance management targeted against malaria vectors in Ethiopia and elsewhere. Therefore, further chemical analysis studies on the identification, preparation, and formulation of bioactive compounds from plants are essential.
[ "Background", "Methods", "Collection of plant samples", "Preparation of plant samples", "Extraction of the plant samples", "Rearing of Anopheles stephensi", "Preparation of stock solution", "Larvicidal bioassay", "Adulticidal bioassay", "Data analysis", "Larvicidal activity of plant extracts against An. stephensi", "The adulticidal activity of plant extracts against An. stephensi" ]
[ "Mosquitoes are among the most important groups of arthropods with medical significance. They transmit several important parasitic and arboviral diseases, such as malaria, filariasis, dengue, yellow fever, and Rift Valley fever [1]. Malaria, caused by protozoans (Plasmodium spp.), can lead to high mortality and morbidity [2]. In 2019 there were 229 million reported malaria cases [2]. Malaria cases that occurred in 2020 were estimated to be 241 million, with 627,000 deaths reported from 85 countries. Around 95% of the malaria cases and 96% of malaria deaths were found in sub-Saharan Africa, with 80% of all malaria deaths in Africa estimated to be among children under the age of five [3].\n\nAnopheles stephensi is a major malaria vector in South Asia and the Middle East, including the Arabian Peninsula [4], and is known to transmit both the major human malaria parasites Plasmodium falciparum and Plasmodium vivax [5]. In 2012, An. stephensi was first reported from the Horn of Africa as an invasive species in Djibouti. In 2016 and 2019, it was reported from additional countries, including Ethiopia, Somalia, and the Republic of Sudan [6–8]. If the species continues to spread across the continent, it is estimated that an additional 126 million people in Africa will be at risk of malaria [9]. In Ethiopia, this malaria vector was first reported from the Somali region in 2016 [10] and was later found to be widely present in urban areas of northeastern Ethiopia [11, 12]. The spread of this vector in different parts of the country has become a serious concern for malaria prevention and elimination strategies. The World Health Organization (WHO) has issued a warning about the invasion and spread of An. stephensi particularly in urging African national malaria control programmes and their partners to be vigilant in areas of risk and to improve and enhance their monitoring systems to identify and control this invasive mosquito species [13]. Recent evidence reveals that invasive An. stephensi from Ethiopia is a more competent vector for P. vivax than Anopheles arabiensis, the primary malaria vector in Ethiopia, in laboratory experiments [14].\nGlobal malaria cases and deaths have been significantly reduced following the scaling up of long-lasting insecticidal nets (LLINs) and indoor residual spraying (IRS) [15, 16]. However, widespread use of synthetic insecticides in controlling mosquito vectors has resulted in the persistence and accumulation of non-biodegradable chemicals in the ecosystem, development of resistance to insecticides in vectors, and toxic effects in non-target organisms [17–19]. As evidenced in recent studies from different parts of Ethiopia, An. arabiensis and An. stephensi have shown resistance to insecticides belonging to four of the chemical classes approved for IRS and ITNs, including DDT (organochlorine), malathion (organophosphate), bendiocarb and propoxur (carbamates), and alpha-cypermethrin, cyfluthrin, deltamethrin, etofenprox, lambda-cyhalothrin, and permethrin (pyrethroids) [20–23]. Reduced effectiveness of insecticides on these malaria vectors may aid in the invasion and establishment of An. stephensi.\nThe emergence of insecticide resistance necessitates an urgent need to develop new and improved mosquito control methods that are economical and effective and less toxic to non-target organisms and the environment. In this regard, botanicals, namely plant extracts and essential oils with insecticidal potential, are recognized as potent alternatives to replace the synthetic insecticides in mosquito control programmes due to their larvicidal, pupicidal, and adulticidal properties; these have also been shown to have oviposition inhibiting, repellent or insect growth regulatory effects, and may help us to find chemicals that are safe, biodegradable, and target specific [24–27].\nTraditionally, plant-based products have constituted an important source of insecticides and other pharmaceutical drugs for many centuries; Calpurnia aurea, Momordica foetida, and Zehneria scabra are the foremost mentioned in Africa [28, 29]. In Ethiopia, these botanicals have been reported as having medicinal properties to prevent vector-borne diseases [30–32] and protect against insect pests [33, 34]. Moreover, these three medicinal plants are cheap and easily available. However, the bioactivities of these plant extract against invasive An. stephensi in Ethiopia has not been evaluated yet. This study evaluated the larvicidal and adulticidal activities of the aforementioned plant leaf extracts against An. stephensi under laboratory conditions.\n[SUBTITLE] Methods [SUBSECTION] [SUBTITLE] Collection of plant samples [SUBSECTION] The fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.\nThe fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.\n[SUBTITLE] Collection of plant samples [SUBSECTION] The fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.\nThe fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.\n[SUBTITLE] Preparation of plant samples [SUBSECTION] The leaves were washed with water and air-dried separately under shade at room temperature for two weeks in the Insect Sciences Laboratory, Addis Ababa University. Finally, the dried leaves were manually ground by pestle and mortar through a sieve into a fine powder. The leaf powder was kept at room temperature in labeled air-tight plastic bags until used.\nThe leaves were washed with water and air-dried separately under shade at room temperature for two weeks in the Insect Sciences Laboratory, Addis Ababa University. Finally, the dried leaves were manually ground by pestle and mortar through a sieve into a fine powder. The leaf powder was kept at room temperature in labeled air-tight plastic bags until used.\n[SUBTITLE] Extraction of the plant samples [SUBSECTION] The crude extraction processes were conducted in the Organic Chemistry Laboratory of the Department of Chemistry, Addis Ababa University. Seventy-two grams of each powdered material was soaked separately in 720 ml of hexane, methanol, and distilled water at a ratio of 1:10 (W/V) in Erlenmeyer flask. Afterward, the mixtures were sonicated by ultrasonic cleaner apparatus (USC-T, Malaysia) at 20 kHz frequency with 11 W power twice per day for 15 min for two days. The mixtures were left to settle for 10 min and then cooled at room temperature. The supernatant of the hexane and methanol crude extracts were filtered through Whatman No.3 (Whatman International Ltd., Maid stone, England) filter paper, while the aqueous extracts using suction were filtered through a Buchner funnel with Whatman filter paper No.1. The filtrates of hexane and methanol crude extracts were concentrated using a vacuum rotary evaporator (Rota-vapor-RE, Buchi Labortechnik AG, Flawil, Switzerland) under reduced pressure at 40oC, while the aqueous crude extracts were evaporated to dryness using a lyophilizer. The residue obtained from each plant extract was left to cool at room temperature to remove traces of solvent, and then finally, were collected separately in a Wheaton bottle glass containers and were preserved a refrigerated at 4 °C until used for experimentation.\nThe crude extraction processes were conducted in the Organic Chemistry Laboratory of the Department of Chemistry, Addis Ababa University. Seventy-two grams of each powdered material was soaked separately in 720 ml of hexane, methanol, and distilled water at a ratio of 1:10 (W/V) in Erlenmeyer flask. Afterward, the mixtures were sonicated by ultrasonic cleaner apparatus (USC-T, Malaysia) at 20 kHz frequency with 11 W power twice per day for 15 min for two days. The mixtures were left to settle for 10 min and then cooled at room temperature. The supernatant of the hexane and methanol crude extracts were filtered through Whatman No.3 (Whatman International Ltd., Maid stone, England) filter paper, while the aqueous extracts using suction were filtered through a Buchner funnel with Whatman filter paper No.1. The filtrates of hexane and methanol crude extracts were concentrated using a vacuum rotary evaporator (Rota-vapor-RE, Buchi Labortechnik AG, Flawil, Switzerland) under reduced pressure at 40oC, while the aqueous crude extracts were evaporated to dryness using a lyophilizer. The residue obtained from each plant extract was left to cool at room temperature to remove traces of solvent, and then finally, were collected separately in a Wheaton bottle glass containers and were preserved a refrigerated at 4 °C until used for experimentation.\n[SUBTITLE] Rearing of Anopheles stephensi [SUBSECTION] Larvicidal and adulticidal bioassays were conducted with colony larvae and adults of An. stephensi originated from the Awash Arba area, eastern Ethiopia. The mosquito larvae and adults were maintained at 28 ± 3oC temperature with 70 ± 10% relative humidity at the insectary of Aklilu Lemma Institute of Pathobiology, Addis Ababa University. All equipment (cages, trays, incubators) containing mosquitoes have been deployed so that accidental contact and release was minimized.\nThe larvae (67 larvae/cm2) were kept in plastic trays (20 cm × 15 cm × 5 cm) containing de-chlorinated water (1.5 l) and were maintained under 12:12 h light and dark photoperiod cycles in the laboratory following the standard mosquito rearing procedure [35]. The larvae were fed with yeast (Saf-Instant yeast). The media were changed every three days. The pupae formed were collected by a glass beaker and transferred to 24.5 × 24.5 × 24.5 cm cages (Bug dorms) for adult emergence. Newly emerged adults were maintained in mosquito cages at 28 ± 3 °C temperature and 70 ± 10% relative humidity and fed sterile 10% sugar solution soaked in cotton pads within Petri dishes. The cotton was always kept moist with the solution and changed every day. In addition to sugar feeding, female mosquitoes were allowed to take blood meals from a restrained rabbit three times a week for egg development and oviposition. Moist filter papers in cups were placed inside rearing cages for oviposition by gravid female mosquitoes. The eggs were washed off with deionized water onto larval rearing trays and allowed to hatch into neonate larvae in the laboratory. Third to fourth instar larvae and bloodmeal starved 2 to 5 days old adult female An. stephensi were used continuously for larvicidal and adulticidal tests, respectively.\nLarvicidal and adulticidal bioassays were conducted with colony larvae and adults of An. stephensi originated from the Awash Arba area, eastern Ethiopia. The mosquito larvae and adults were maintained at 28 ± 3oC temperature with 70 ± 10% relative humidity at the insectary of Aklilu Lemma Institute of Pathobiology, Addis Ababa University. All equipment (cages, trays, incubators) containing mosquitoes have been deployed so that accidental contact and release was minimized.\nThe larvae (67 larvae/cm2) were kept in plastic trays (20 cm × 15 cm × 5 cm) containing de-chlorinated water (1.5 l) and were maintained under 12:12 h light and dark photoperiod cycles in the laboratory following the standard mosquito rearing procedure [35]. The larvae were fed with yeast (Saf-Instant yeast). The media were changed every three days. The pupae formed were collected by a glass beaker and transferred to 24.5 × 24.5 × 24.5 cm cages (Bug dorms) for adult emergence. Newly emerged adults were maintained in mosquito cages at 28 ± 3 °C temperature and 70 ± 10% relative humidity and fed sterile 10% sugar solution soaked in cotton pads within Petri dishes. The cotton was always kept moist with the solution and changed every day. In addition to sugar feeding, female mosquitoes were allowed to take blood meals from a restrained rabbit three times a week for egg development and oviposition. Moist filter papers in cups were placed inside rearing cages for oviposition by gravid female mosquitoes. The eggs were washed off with deionized water onto larval rearing trays and allowed to hatch into neonate larvae in the laboratory. Third to fourth instar larvae and bloodmeal starved 2 to 5 days old adult female An. stephensi were used continuously for larvicidal and adulticidal tests, respectively.\n[SUBTITLE] Preparation of stock solution [SUBSECTION] One gram of each crude plant extract was placed separately in 250 ml Wheaton glass bottles and dissolved in 100 ml of distilled water for methanol and aqueous extracts, while the hexane extracts were first dissolved in 4 ml of acetone and then added to 96 ml of distilled water to prepare 100 ml of a 1% stock solution. In the stock solution, one drop of emulsifier (Tween 80) was added to each extract at a concentration of 0.05%. From the 1% stock solution, concentrations of 25, 50, 100, 150, 200, 250, and 300 ppm for the larvae, and concentrations of 20, 40, 80, 160, and 320 ppm for the adult were prepared for exposure of the target mosquito.\nOne gram of each crude plant extract was placed separately in 250 ml Wheaton glass bottles and dissolved in 100 ml of distilled water for methanol and aqueous extracts, while the hexane extracts were first dissolved in 4 ml of acetone and then added to 96 ml of distilled water to prepare 100 ml of a 1% stock solution. In the stock solution, one drop of emulsifier (Tween 80) was added to each extract at a concentration of 0.05%. From the 1% stock solution, concentrations of 25, 50, 100, 150, 200, 250, and 300 ppm for the larvae, and concentrations of 20, 40, 80, 160, and 320 ppm for the adult were prepared for exposure of the target mosquito.\n[SUBTITLE] Larvicidal bioassay [SUBSECTION] Larvicidal activities of each leaf crude extract were measured according to WHO standard procedures [35]. For larvicidal bioassay, hexane, methanol and aqueous crude leaf extracts of the test plants were screened at 25, 50, 100, 150, 200, 250, and 300 ppm of test concentrations. Twenty individuals of late third to early fourth instar larvae were kept in a 300 ml enamel cups containing 99 ml of distilled water and 1-ml of desired concentrations of crude solvent extracts of C. aurea, M. foetida and Z. scabra were added. In the same way, 99 ml of distilled water with 1 ml mixture of acetone (for hexane extracts) and Tween 80 were made up to 100 ml to serve as a negative control. In addition, temephos at the rate of 0.25 ppm within a similar volume of test cups was used as a positive control [36]. The experiment was conducted for 24 h at the optimum conditions of 28 ± 3 °C temperature and 70 ± 10% relative humidity under 12:12 light and dark photoperiod in the insectary. During the exposure period, no food was offered to the larvae. Each experiment was run three times on different days along randomly set up with appropriate negative control and standard check. Numbers of dead larvae were counted after 24 h of exposure, and the percentage of mortality was reported from the averages of twelve replicates. The dead larvae included moribunds; those incapable of rising to the surface in each concentration of treatments, and the standards were combined separately and expressed as the average mortality to determine LC50 and LC90 values.\nLarvicidal activities of each leaf crude extract were measured according to WHO standard procedures [35]. For larvicidal bioassay, hexane, methanol and aqueous crude leaf extracts of the test plants were screened at 25, 50, 100, 150, 200, 250, and 300 ppm of test concentrations. Twenty individuals of late third to early fourth instar larvae were kept in a 300 ml enamel cups containing 99 ml of distilled water and 1-ml of desired concentrations of crude solvent extracts of C. aurea, M. foetida and Z. scabra were added. In the same way, 99 ml of distilled water with 1 ml mixture of acetone (for hexane extracts) and Tween 80 were made up to 100 ml to serve as a negative control. In addition, temephos at the rate of 0.25 ppm within a similar volume of test cups was used as a positive control [36]. The experiment was conducted for 24 h at the optimum conditions of 28 ± 3 °C temperature and 70 ± 10% relative humidity under 12:12 light and dark photoperiod in the insectary. During the exposure period, no food was offered to the larvae. Each experiment was run three times on different days along randomly set up with appropriate negative control and standard check. Numbers of dead larvae were counted after 24 h of exposure, and the percentage of mortality was reported from the averages of twelve replicates. The dead larvae included moribunds; those incapable of rising to the surface in each concentration of treatments, and the standards were combined separately and expressed as the average mortality to determine LC50 and LC90 values.\n[SUBTITLE] Adulticidal bioassay [SUBSECTION] Adulticidal activities of each crude solvent extract were performed using the CDC bottle bioassays [37, 38]. Most of the synthetic insecticides available for the control of adult mosquitoes, in particular An. stephensi were reported to be resisted by adults [12, 23], thus, this bioassay did not have any synthetic insecticide as a positive control. In this bioassay, aqueous, hexane and methanol crude leaf extracts of the three plants were screened at 25, 50, 100, 150, 200, 250, and 300 ppm concentrations. Two hundred fifty ml Wheaton bottles with screw lids were properly cleaned and dried, and then they were coated with 1ml solution of the desired concentrations of the tested extracts by swirling assuring complete coating of the bottle and its cap. Similarly, 1ml of acetone and emulsifier (Tween 80) solution was added to the control bottle handled as before.\nAdult mosquitoes (20, aged 2–5 days) fed on 10% sucrose solution were released to each bottle, including the control bottle with the help of mouth aspirator. The opening of each bottle was closed with their lids after introduction of mosquito adults. The exposure time was set to 2 h. After that, the mosquitoes were transferred into 250 ml capacity plastic cups using aspirator, where they were provided with 10% sucrose solution and the mortalities were checked after 24 h. The experiment was done at 28 ± 3 °C temperature and 70 ± 10% relative humidity under 12:12 light and dark photoperiod in the insectary. The percentage of adult mortality was corrected using Abbott’s formula [39] when needed.\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\% \\text{Morality} = \\frac{{\\% \\text{test}\\,\\text{morality} - \\% \\text{control}\\, \\text{morality}}}{{100 - \\% \\text{control}\\, \\text{morality}}} \\times 100. $$\\end{document}%Morality=%testmorality-%controlmorality100-%controlmorality×100.\nAdulticidal activities of each crude solvent extract were performed using the CDC bottle bioassays [37, 38]. Most of the synthetic insecticides available for the control of adult mosquitoes, in particular An. stephensi were reported to be resisted by adults [12, 23], thus, this bioassay did not have any synthetic insecticide as a positive control. In this bioassay, aqueous, hexane and methanol crude leaf extracts of the three plants were screened at 25, 50, 100, 150, 200, 250, and 300 ppm concentrations. Two hundred fifty ml Wheaton bottles with screw lids were properly cleaned and dried, and then they were coated with 1ml solution of the desired concentrations of the tested extracts by swirling assuring complete coating of the bottle and its cap. Similarly, 1ml of acetone and emulsifier (Tween 80) solution was added to the control bottle handled as before.\nAdult mosquitoes (20, aged 2–5 days) fed on 10% sucrose solution were released to each bottle, including the control bottle with the help of mouth aspirator. The opening of each bottle was closed with their lids after introduction of mosquito adults. The exposure time was set to 2 h. After that, the mosquitoes were transferred into 250 ml capacity plastic cups using aspirator, where they were provided with 10% sucrose solution and the mortalities were checked after 24 h. The experiment was done at 28 ± 3 °C temperature and 70 ± 10% relative humidity under 12:12 light and dark photoperiod in the insectary. The percentage of adult mortality was corrected using Abbott’s formula [39] when needed.\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\% \\text{Morality} = \\frac{{\\% \\text{test}\\,\\text{morality} - \\% \\text{control}\\, \\text{morality}}}{{100 - \\% \\text{control}\\, \\text{morality}}} \\times 100. $$\\end{document}%Morality=%testmorality-%controlmorality100-%controlmorality×100.\n[SUBTITLE] Data analysis [SUBSECTION] The mean percentage mortality was analysed using SPSS version 25.0 software (SPSS Inc, Chicago, IL, USA), and dose-dependent mortality also was performed by R software version 4.0.5. The percentage mortality of larvae and adult were checked for normality by 1-Sample Kolmogorov-Smirnov Z test (K-S). When the percent mortality did not conform to the normal distribution, the non-parametric equivalent tests of Kruskal-Wallis were followed. p-values were adjusted with the Bonferroni correction to adjust for the inflation of type I errors when several Mann–Whitney tests are performed [40]. The LC50 and LC90 values were calculated using a generalized linear probit model, considering a 95% confidence interval.\nThe mean percentage mortality was analysed using SPSS version 25.0 software (SPSS Inc, Chicago, IL, USA), and dose-dependent mortality also was performed by R software version 4.0.5. The percentage mortality of larvae and adult were checked for normality by 1-Sample Kolmogorov-Smirnov Z test (K-S). When the percent mortality did not conform to the normal distribution, the non-parametric equivalent tests of Kruskal-Wallis were followed. p-values were adjusted with the Bonferroni correction to adjust for the inflation of type I errors when several Mann–Whitney tests are performed [40]. The LC50 and LC90 values were calculated using a generalized linear probit model, considering a 95% confidence interval.", "[SUBTITLE] Collection of plant samples [SUBSECTION] The fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.\nThe fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.", "The fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.", "The leaves were washed with water and air-dried separately under shade at room temperature for two weeks in the Insect Sciences Laboratory, Addis Ababa University. Finally, the dried leaves were manually ground by pestle and mortar through a sieve into a fine powder. The leaf powder was kept at room temperature in labeled air-tight plastic bags until used.", "The crude extraction processes were conducted in the Organic Chemistry Laboratory of the Department of Chemistry, Addis Ababa University. Seventy-two grams of each powdered material was soaked separately in 720 ml of hexane, methanol, and distilled water at a ratio of 1:10 (W/V) in Erlenmeyer flask. Afterward, the mixtures were sonicated by ultrasonic cleaner apparatus (USC-T, Malaysia) at 20 kHz frequency with 11 W power twice per day for 15 min for two days. The mixtures were left to settle for 10 min and then cooled at room temperature. The supernatant of the hexane and methanol crude extracts were filtered through Whatman No.3 (Whatman International Ltd., Maid stone, England) filter paper, while the aqueous extracts using suction were filtered through a Buchner funnel with Whatman filter paper No.1. The filtrates of hexane and methanol crude extracts were concentrated using a vacuum rotary evaporator (Rota-vapor-RE, Buchi Labortechnik AG, Flawil, Switzerland) under reduced pressure at 40oC, while the aqueous crude extracts were evaporated to dryness using a lyophilizer. The residue obtained from each plant extract was left to cool at room temperature to remove traces of solvent, and then finally, were collected separately in a Wheaton bottle glass containers and were preserved a refrigerated at 4 °C until used for experimentation.", "Larvicidal and adulticidal bioassays were conducted with colony larvae and adults of An. stephensi originated from the Awash Arba area, eastern Ethiopia. The mosquito larvae and adults were maintained at 28 ± 3oC temperature with 70 ± 10% relative humidity at the insectary of Aklilu Lemma Institute of Pathobiology, Addis Ababa University. All equipment (cages, trays, incubators) containing mosquitoes have been deployed so that accidental contact and release was minimized.\nThe larvae (67 larvae/cm2) were kept in plastic trays (20 cm × 15 cm × 5 cm) containing de-chlorinated water (1.5 l) and were maintained under 12:12 h light and dark photoperiod cycles in the laboratory following the standard mosquito rearing procedure [35]. The larvae were fed with yeast (Saf-Instant yeast). The media were changed every three days. The pupae formed were collected by a glass beaker and transferred to 24.5 × 24.5 × 24.5 cm cages (Bug dorms) for adult emergence. Newly emerged adults were maintained in mosquito cages at 28 ± 3 °C temperature and 70 ± 10% relative humidity and fed sterile 10% sugar solution soaked in cotton pads within Petri dishes. The cotton was always kept moist with the solution and changed every day. In addition to sugar feeding, female mosquitoes were allowed to take blood meals from a restrained rabbit three times a week for egg development and oviposition. Moist filter papers in cups were placed inside rearing cages for oviposition by gravid female mosquitoes. The eggs were washed off with deionized water onto larval rearing trays and allowed to hatch into neonate larvae in the laboratory. Third to fourth instar larvae and bloodmeal starved 2 to 5 days old adult female An. stephensi were used continuously for larvicidal and adulticidal tests, respectively.", "One gram of each crude plant extract was placed separately in 250 ml Wheaton glass bottles and dissolved in 100 ml of distilled water for methanol and aqueous extracts, while the hexane extracts were first dissolved in 4 ml of acetone and then added to 96 ml of distilled water to prepare 100 ml of a 1% stock solution. In the stock solution, one drop of emulsifier (Tween 80) was added to each extract at a concentration of 0.05%. From the 1% stock solution, concentrations of 25, 50, 100, 150, 200, 250, and 300 ppm for the larvae, and concentrations of 20, 40, 80, 160, and 320 ppm for the adult were prepared for exposure of the target mosquito.", "Larvicidal activities of each leaf crude extract were measured according to WHO standard procedures [35]. For larvicidal bioassay, hexane, methanol and aqueous crude leaf extracts of the test plants were screened at 25, 50, 100, 150, 200, 250, and 300 ppm of test concentrations. Twenty individuals of late third to early fourth instar larvae were kept in a 300 ml enamel cups containing 99 ml of distilled water and 1-ml of desired concentrations of crude solvent extracts of C. aurea, M. foetida and Z. scabra were added. In the same way, 99 ml of distilled water with 1 ml mixture of acetone (for hexane extracts) and Tween 80 were made up to 100 ml to serve as a negative control. In addition, temephos at the rate of 0.25 ppm within a similar volume of test cups was used as a positive control [36]. The experiment was conducted for 24 h at the optimum conditions of 28 ± 3 °C temperature and 70 ± 10% relative humidity under 12:12 light and dark photoperiod in the insectary. During the exposure period, no food was offered to the larvae. Each experiment was run three times on different days along randomly set up with appropriate negative control and standard check. Numbers of dead larvae were counted after 24 h of exposure, and the percentage of mortality was reported from the averages of twelve replicates. The dead larvae included moribunds; those incapable of rising to the surface in each concentration of treatments, and the standards were combined separately and expressed as the average mortality to determine LC50 and LC90 values.", "Adulticidal activities of each crude solvent extract were performed using the CDC bottle bioassays [37, 38]. Most of the synthetic insecticides available for the control of adult mosquitoes, in particular An. stephensi were reported to be resisted by adults [12, 23], thus, this bioassay did not have any synthetic insecticide as a positive control. In this bioassay, aqueous, hexane and methanol crude leaf extracts of the three plants were screened at 25, 50, 100, 150, 200, 250, and 300 ppm concentrations. Two hundred fifty ml Wheaton bottles with screw lids were properly cleaned and dried, and then they were coated with 1ml solution of the desired concentrations of the tested extracts by swirling assuring complete coating of the bottle and its cap. Similarly, 1ml of acetone and emulsifier (Tween 80) solution was added to the control bottle handled as before.\nAdult mosquitoes (20, aged 2–5 days) fed on 10% sucrose solution were released to each bottle, including the control bottle with the help of mouth aspirator. The opening of each bottle was closed with their lids after introduction of mosquito adults. The exposure time was set to 2 h. After that, the mosquitoes were transferred into 250 ml capacity plastic cups using aspirator, where they were provided with 10% sucrose solution and the mortalities were checked after 24 h. The experiment was done at 28 ± 3 °C temperature and 70 ± 10% relative humidity under 12:12 light and dark photoperiod in the insectary. The percentage of adult mortality was corrected using Abbott’s formula [39] when needed.\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\% \\text{Morality} = \\frac{{\\% \\text{test}\\,\\text{morality} - \\% \\text{control}\\, \\text{morality}}}{{100 - \\% \\text{control}\\, \\text{morality}}} \\times 100. $$\\end{document}%Morality=%testmorality-%controlmorality100-%controlmorality×100.", "The mean percentage mortality was analysed using SPSS version 25.0 software (SPSS Inc, Chicago, IL, USA), and dose-dependent mortality also was performed by R software version 4.0.5. The percentage mortality of larvae and adult were checked for normality by 1-Sample Kolmogorov-Smirnov Z test (K-S). When the percent mortality did not conform to the normal distribution, the non-parametric equivalent tests of Kruskal-Wallis were followed. p-values were adjusted with the Bonferroni correction to adjust for the inflation of type I errors when several Mann–Whitney tests are performed [40]. The LC50 and LC90 values were calculated using a generalized linear probit model, considering a 95% confidence interval.", "Larval mortalities obtained from bioassays of aqueous and methanol extracts of Calpurnia aurea, Momordica foetida, and Zehneria scabra leaves against An. stephensi larvae after 24 h exposure periods are presented in Tables 1, 2 and 3. All aqueous and methanol test plant extracts showed low, moderate, and high larvicidal activities tested between 25 ppm-300 ppm treatments against the late 3rd to early 4th instar larvae of An. stephensi. Within the same exposure period, larval mortality was not observed for the tests using hexane crude leaf extracts and the negative control. The standard positive control (temephos) achieved 100% larval mortality.\n\nTable 1Mean % larval mortality of Calpurnia aurea crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol 2535.00 ± 2.6Aa23.75 ± 3.65Aa5060.00 ± 3.54Ba35.00 ± 0.00Bb10086.25 ± 2.23Ca90.42 ± 1.4Ca15099.58 ± 0.42Da95.00 ± 2.46DCa200100.00 ± 0.00Da96.67 ± 1.42Da250100.00 ± 0.00Da100.00 ± 0.00Da300100.00 ± 0.00Da100.00 ± 0.00DaTemephos (0.25)100.00 ± 0.00Da100.00 ± 0.00DaControl0.00 ± 0.00Ea0.00 ± 0.00Ea*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p > 0.003)\nMean % larval mortality of Calpurnia aurea crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure\n*Each value of % mean mortality ± SE represents the value of twelve replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p > 0.003)\nThere was a significant difference and interaction effects between all aqueous and methanol test plant extract against the tested larvae (Kruskal-Wallis test, p-value < 0.05). The mean percent larval mortality between treatments, control, and the standard groups had a statistically significant difference (Kruskal-Wallis test, p-value < 0.05). The mortality effect of the test plant extracts against An. stephensi larvae were dose and extraction solvent dependent.\nIn the C. aurea test plant, the highest (100%) larval mortality was found both in aqueous and methanol extracts at 200 ppm and 250 ppm of treatments, respectively (Table 1). Even at 150 ppm, the larvicidal effects of aqueous and methanol extracts of C. aurea were not significantly different from standard checks (Multiple-Mann Whitney U test, p-value > 0.003; Table 1). Yet, all C. aurea treatments significantly affected larval mortality compared to the negative control (Multiple-Mann Whitney U test; p < 0.003).\nAqueous and methanol extracts of the M. foetida test plant showed 100% larval mortality at 200 ppm and 250 ppm of treatments, respectively (Table 2). Moreover, larvicidal activity found from aqueous extracts of M. foetida at 100 ppm and methanol extracts at 200 ppm caused mortality (ranging 95–97.9%) was not significantly different from the mortality achieved in temephos exposed on An. stephensi larvae (Multiple-Mann Whitney U test, p-value > 0.003; Table 2). Statistical differences were observed between all M. foetida treatments and negative control (Multiple-Mann Whitney U test; p-value < 0.003).\n\nTable 2Mean % larval mortality of Momordica foetida crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol 2527.5 ± 2.85Aa4.55 ± 1.4Ab5080.83 ± 4.51Ba20.83 ± 2.53Bb10095.00 ± 1.74Ca45.83 ± 2.20Cb15097.92 ± 0.96Ca71.67 ± 3.39Db200100.00 ± 0.00Ca97.5 ± 2.50Ea250100.00 ± 0.00Ca100.00 ± 0.00Ea300100.00 ± 0.00Ca100.00 ± 0.00EaTemephos (0.25)100.00 ± 0.00Ca100.00 ± 0.00EaControl0.00 ± 0.00Da0.00 ± 0.00Fa*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)Table 3Mean % larval mortality of Zehneria scabra crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE\nSolvents\nAqueous \nMethanol 2529.17 ± 3.68Aa23.75 ± 2.69Aa5073.33 ± 3.9Ba66.25 ± 5.04Ba10095.42 ± 1.44Ca87.08 ± 2.17Ca15097.08 ± 1.44Ca92.50 ± 1.44Da20099.50 ± 0.42Ca99.17 ± 0.83Ea250100.00 ± 0.00Ca100.00 ± 0.00Ea300100.00 ± 0.00Ca100.00 ± 0.00EaTemephos (0.25)100.00 ± 0.00Ca100.00 ± 0.00EaControl0.00 ± 0.00Da0.00 ± 0.00Fa*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % larval mortality of Momordica foetida crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure\n*Each value of % mean mortality ± SE represents the value of twelve replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % larval mortality of Zehneria scabra crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure\n*Each value of % mean mortality ± SE represents the value of twelve replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nAmong the two solvent test concentrations of Z. scabra extracts, statistically, non-significant differences were observed (Multiple-Mann Whitney U test, p-value > 0.003; Table 3). However, their larvicidal activity was significantly different from negative control (Multiple-Mann Whitney U test; p-value < 0.003). At the highest concentrations (250 ppm), both aqueous and methanol Z. scabra, crude leaf extracts resulted in 100% larval mortality (Table 3). Moreover, aqueous extracts of Z. scabra at 100 ppm, and methanol extracts at 200 ppm, each caused more than 95% larval mortality, had no significant effect compared to mortality achieved in temephos treatment (Multiple-Mann Whitney U test; p-value > 0.003;).\nThe lethal toxicity doses of aqueous and methanol crude leaf extracts of C. aurea, M. foetida, and Z. scabra against larvae of An. stephensi are shown in Table 4. Crude aqueous leaf extract of M. foetida showed strong larvicidal activity, having an LC50 value of 34.61 ppm and LC90 value of 57.61 ppm, followed by Z. scabra (LC50 = 35.85 ppm; LC90 = 68.26 ppm) and C. aurea (LC50 = 38.69 ppm; LC90 = 108.28 ppm). Similarly, the methanol leaf extract of Z. scabra had good larvicidal activities with LC50 and LC90 values of 41.32 ppm and 99.07 ppm, followed by C. aurea (LC50 = 43.25 ppm; LC90 = 96.02 ppm). However, the crude methanol leaf extract of M. foetida had lower larvicidal toxicity against the larvae of An. stephensi compared to other test plant extracts (LC50 = 99.50 ppm; LC90 = 188.76 ppm, Table 4).\n\nTable 4LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of tests plants against the late third to early fourth instar larvae of Anopheles stephensiTest plantsSolventsLC50 (ppm)95%Confidence limitLC90 (ppm)95%Confidence limitχ2 (df = 5)\nLCL \nUCL \nLCL \nUCL \nC. aureaAqueous38. 6935. 5741.82108. 2896. 27120. 302.93Methanol43. 2540. 4946. 0296. 0283. 87108. 184.15\nM. foetidaAqueous34. 6132. 8436. 3857. 6153. 2961. 930.71Methanol99. 5094. 56104. 43188. 76177. 70199. 826.90\nZ. scabraAqueous35. 8533. 7937. 9168. 2660. 2076. 320.37Methanol41. 3238. 5144. 1299. 0784. 64113. 501.43\nLC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of tests plants against the late third to early fourth instar larvae of Anopheles stephensi", "Tables 5, 6 and 7 show the mean percentage mortalities and toxicity effects of adult An. stephensi after 24 h exposures to different solvent extracts of C. aurea, M. foetida, and Z. scabra. Statistically significant differences in mean percentage mortality among the different extracts and the negative control were observed (Kruskal-Wallis test, p < 0.05). The adulticidal data showed that the mortality rate of the vector was directly proportional to the concentration and extraction solvents. All hexane test plant extracts showed lower adulticidal property against An. stephensi (≤ 20% at all treatments tested in the bioassays, Tables 5, 6 and 7). Between the different solvent leaf extracts of C. aurea, only aqueous extract gave 55% adult mortality of An. stephensi at 320 ppm, while other crude solvent leaf extracts had lower adulticidal activity, having adult mean mortalities ranging from 0 to 48% (Table 5).\n\nTable 5Mean % adult mortality of Calpurnia aurea crude leaf solvent extracts at different treatments against An. stephensiConcentration (ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol \nHexane 201.67 ± 1.67Aa0.00 ± 0.00Aa0.00 ± 0.00Aa406.67 ± 1.67Aa6.67 ± 1.67Ba0.00 ± 0.00Ab8016.67 ± 1.67Ba16.67 ± 1.67Ca0.00 ± 0.00Ab16021.67 ± 1.67Ba26.67 ± 3.33Da6.67 ± 3.33Bb32055.00 ± 5.77Ca48.33 ± 1.67Ea18.33 ± 1.67CbControl0.00 ± 0.00Da0.00 ± 0.00Aa0.00 ± 0.00Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % adult mortality of Calpurnia aurea crude leaf solvent extracts at different treatments against An. stephensi\n*Each value of % mean mortality ± SE represents the value of three replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\n\nDifferent solvent (aqueous, methanol, and hexane) crude leaf extracts of M. foetida resulted in lower adult mortalities of An. stephensi (< 24%) at all treatments tested in the bioassays and were not effective against adults of malaria vector An. stephensi (Table 6).\n\nTable 6Mean % adult mortality of Momordica foetida crude leaf solvent extract at different concentrations against Anopheles stephensiConcentration(ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol \nHexane 200.00 ± 0.00ABa0.00 ± 0.00Aa0.00 ± 0.00Aa401.67 ± 1.67Ba0.00 ± 0.00Aa0.00 ± 0.00Ab805.00 ± 2.89Ba1.67 ± 1.67Ba0.00 ± 0.00Ab16013.33 ± 1.67Ca13.33 ± 1.67Ca0.00 ± 0.00Ab32018.33 ± 4.41Ca23.33 ± 1.67Da1.67 ± 1.67BbControl0.00 ± 0.00Aa0.00 ± 0.00Aa0.00 ± 0.00Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % adult mortality of Momordica foetida crude leaf solvent extract at different concentrations against Anopheles stephensi\n*Each value of % mean mortality ± SE represents the value of three replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nThere were statistically significant differences in mean percentage mortality among different concentrations of aqueous, methanol, and hexane leaf extracts of Z. scabra and with negative control (Kruskal-Wallis test; p-value < 0.05; Table 7). Within the given time intervals adult mortality was recorded for the control treatment. However, the highest adult mortality (84%) at higher concentration 320 ppm against An. stephensi was recorded in aqueous crude leaf extract of Z. scabra followed by its methanol extracts (65%) and their adulticidal potential was statistically different from the negative control (Multiple-Mann Whitney U-test; p-value < 0.003, Table 7).Table 7Mean % adult mortality of Zehneria scabra crude leaf solvent extracts at different concentrations against Anopheles stephensiConcentration (ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol \nHexane 204.33 ± 2.6Aa11.67 ± 3.33Aa0.00 ± 0.00Aa4011.67 ± 1.67Ba20.00 ± 2.89ABa0.00 ± 0.00Ab8026.67 ± 3.33Ca23.33 ± 3.33Ba0.00 ± 0.00Ab16036.67 ± 6.00Ca41.66 ± 4.40Ca15.00 ± 2.89Bb32084.00 ± 2.89Da65.00 ± 5.77Db20.00 ± 2.89BcControl0.67 ± 0.45Ea0.67 ± 0.45Ea0.67 ± 0.45Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % adult mortality of Zehneria scabra crude leaf solvent extracts at different concentrations against Anopheles stephensi\n*Each value of % mean mortality ± SE represents the value of three replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nThe lethal effects of different crude leaf extracts against adults of An. stephensi are shown in Table 8. All hexane and M. foetida crude leaf extracts had low adulticidal impacts, only aqueous and methanol C. aurea and Z. scabra leaf extracts were subjected to dose-response bioassay to detect the lethal concentrations. From the dose-response curves, the lowest LC50 (= 176.20 ppm and 205.41 ppm) and LC90 (= 425.13 and 883.11 ppm) values were demonstrated from the aqueous and methanol Z. scabra crude leaf extracts, respectively (Table 8), and its relative toxicities against adult An. stephensi was much higher than the rest of the plant extracts. The highest LC50 (= 297.75 ppm and 333.27 ppm) and LC90 (= 762.63 ppm and 1031.74 ppm) values were from the aqueous and methanol C. aurea crude leaf extracts, respectively, and its relative toxicity on An. stephensi was much lower than Z. scabra crude extracts.\n\nTable 8LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of Calpurnia aurea and Zehneria scabra against adults of Anopheles stephensiTest plantsSolventsLC5095%Confidence limitLC9095%Confidence limitχ2 (df = 3)\nLCL \nUCL \nLCL \nUCL \nC. aureaAqueous297. 75254. 95340. 56762. 63491. 131034. 130.88Methanol333. 27268. 92397. 631031. 74569. 061494. 430.75\nZ. scabraAqueous176.20157. 82194. 58425. 13346. 14504. 122.70Methanol205. 41172. 29238. 54883. 11529. 331236. 900.70\nLC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of Calpurnia aurea and Zehneria scabra against adults of Anopheles stephensi" ]
[ null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Collection of plant samples", "Preparation of plant samples", "Extraction of the plant samples", "Rearing of Anopheles stephensi", "Preparation of stock solution", "Larvicidal bioassay", "Adulticidal bioassay", "Data analysis", "Results", "Larvicidal activity of plant extracts against An. stephensi", "The adulticidal activity of plant extracts against An. stephensi", "Discussion", "Conclusion" ]
[ "Mosquitoes are among the most important groups of arthropods with medical significance. They transmit several important parasitic and arboviral diseases, such as malaria, filariasis, dengue, yellow fever, and Rift Valley fever [1]. Malaria, caused by protozoans (Plasmodium spp.), can lead to high mortality and morbidity [2]. In 2019 there were 229 million reported malaria cases [2]. Malaria cases that occurred in 2020 were estimated to be 241 million, with 627,000 deaths reported from 85 countries. Around 95% of the malaria cases and 96% of malaria deaths were found in sub-Saharan Africa, with 80% of all malaria deaths in Africa estimated to be among children under the age of five [3].\n\nAnopheles stephensi is a major malaria vector in South Asia and the Middle East, including the Arabian Peninsula [4], and is known to transmit both the major human malaria parasites Plasmodium falciparum and Plasmodium vivax [5]. In 2012, An. stephensi was first reported from the Horn of Africa as an invasive species in Djibouti. In 2016 and 2019, it was reported from additional countries, including Ethiopia, Somalia, and the Republic of Sudan [6–8]. If the species continues to spread across the continent, it is estimated that an additional 126 million people in Africa will be at risk of malaria [9]. In Ethiopia, this malaria vector was first reported from the Somali region in 2016 [10] and was later found to be widely present in urban areas of northeastern Ethiopia [11, 12]. The spread of this vector in different parts of the country has become a serious concern for malaria prevention and elimination strategies. The World Health Organization (WHO) has issued a warning about the invasion and spread of An. stephensi particularly in urging African national malaria control programmes and their partners to be vigilant in areas of risk and to improve and enhance their monitoring systems to identify and control this invasive mosquito species [13]. Recent evidence reveals that invasive An. stephensi from Ethiopia is a more competent vector for P. vivax than Anopheles arabiensis, the primary malaria vector in Ethiopia, in laboratory experiments [14].\nGlobal malaria cases and deaths have been significantly reduced following the scaling up of long-lasting insecticidal nets (LLINs) and indoor residual spraying (IRS) [15, 16]. However, widespread use of synthetic insecticides in controlling mosquito vectors has resulted in the persistence and accumulation of non-biodegradable chemicals in the ecosystem, development of resistance to insecticides in vectors, and toxic effects in non-target organisms [17–19]. As evidenced in recent studies from different parts of Ethiopia, An. arabiensis and An. stephensi have shown resistance to insecticides belonging to four of the chemical classes approved for IRS and ITNs, including DDT (organochlorine), malathion (organophosphate), bendiocarb and propoxur (carbamates), and alpha-cypermethrin, cyfluthrin, deltamethrin, etofenprox, lambda-cyhalothrin, and permethrin (pyrethroids) [20–23]. Reduced effectiveness of insecticides on these malaria vectors may aid in the invasion and establishment of An. stephensi.\nThe emergence of insecticide resistance necessitates an urgent need to develop new and improved mosquito control methods that are economical and effective and less toxic to non-target organisms and the environment. In this regard, botanicals, namely plant extracts and essential oils with insecticidal potential, are recognized as potent alternatives to replace the synthetic insecticides in mosquito control programmes due to their larvicidal, pupicidal, and adulticidal properties; these have also been shown to have oviposition inhibiting, repellent or insect growth regulatory effects, and may help us to find chemicals that are safe, biodegradable, and target specific [24–27].\nTraditionally, plant-based products have constituted an important source of insecticides and other pharmaceutical drugs for many centuries; Calpurnia aurea, Momordica foetida, and Zehneria scabra are the foremost mentioned in Africa [28, 29]. In Ethiopia, these botanicals have been reported as having medicinal properties to prevent vector-borne diseases [30–32] and protect against insect pests [33, 34]. Moreover, these three medicinal plants are cheap and easily available. However, the bioactivities of these plant extract against invasive An. stephensi in Ethiopia has not been evaluated yet. This study evaluated the larvicidal and adulticidal activities of the aforementioned plant leaf extracts against An. stephensi under laboratory conditions.\n[SUBTITLE] Methods [SUBSECTION] [SUBTITLE] Collection of plant samples [SUBSECTION] The fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.\nThe fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.\n[SUBTITLE] Collection of plant samples [SUBSECTION] The fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.\nThe fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.\n[SUBTITLE] Preparation of plant samples [SUBSECTION] The leaves were washed with water and air-dried separately under shade at room temperature for two weeks in the Insect Sciences Laboratory, Addis Ababa University. Finally, the dried leaves were manually ground by pestle and mortar through a sieve into a fine powder. The leaf powder was kept at room temperature in labeled air-tight plastic bags until used.\nThe leaves were washed with water and air-dried separately under shade at room temperature for two weeks in the Insect Sciences Laboratory, Addis Ababa University. Finally, the dried leaves were manually ground by pestle and mortar through a sieve into a fine powder. The leaf powder was kept at room temperature in labeled air-tight plastic bags until used.\n[SUBTITLE] Extraction of the plant samples [SUBSECTION] The crude extraction processes were conducted in the Organic Chemistry Laboratory of the Department of Chemistry, Addis Ababa University. Seventy-two grams of each powdered material was soaked separately in 720 ml of hexane, methanol, and distilled water at a ratio of 1:10 (W/V) in Erlenmeyer flask. Afterward, the mixtures were sonicated by ultrasonic cleaner apparatus (USC-T, Malaysia) at 20 kHz frequency with 11 W power twice per day for 15 min for two days. The mixtures were left to settle for 10 min and then cooled at room temperature. The supernatant of the hexane and methanol crude extracts were filtered through Whatman No.3 (Whatman International Ltd., Maid stone, England) filter paper, while the aqueous extracts using suction were filtered through a Buchner funnel with Whatman filter paper No.1. The filtrates of hexane and methanol crude extracts were concentrated using a vacuum rotary evaporator (Rota-vapor-RE, Buchi Labortechnik AG, Flawil, Switzerland) under reduced pressure at 40oC, while the aqueous crude extracts were evaporated to dryness using a lyophilizer. The residue obtained from each plant extract was left to cool at room temperature to remove traces of solvent, and then finally, were collected separately in a Wheaton bottle glass containers and were preserved a refrigerated at 4 °C until used for experimentation.\nThe crude extraction processes were conducted in the Organic Chemistry Laboratory of the Department of Chemistry, Addis Ababa University. Seventy-two grams of each powdered material was soaked separately in 720 ml of hexane, methanol, and distilled water at a ratio of 1:10 (W/V) in Erlenmeyer flask. Afterward, the mixtures were sonicated by ultrasonic cleaner apparatus (USC-T, Malaysia) at 20 kHz frequency with 11 W power twice per day for 15 min for two days. The mixtures were left to settle for 10 min and then cooled at room temperature. The supernatant of the hexane and methanol crude extracts were filtered through Whatman No.3 (Whatman International Ltd., Maid stone, England) filter paper, while the aqueous extracts using suction were filtered through a Buchner funnel with Whatman filter paper No.1. The filtrates of hexane and methanol crude extracts were concentrated using a vacuum rotary evaporator (Rota-vapor-RE, Buchi Labortechnik AG, Flawil, Switzerland) under reduced pressure at 40oC, while the aqueous crude extracts were evaporated to dryness using a lyophilizer. The residue obtained from each plant extract was left to cool at room temperature to remove traces of solvent, and then finally, were collected separately in a Wheaton bottle glass containers and were preserved a refrigerated at 4 °C until used for experimentation.\n[SUBTITLE] Rearing of Anopheles stephensi [SUBSECTION] Larvicidal and adulticidal bioassays were conducted with colony larvae and adults of An. stephensi originated from the Awash Arba area, eastern Ethiopia. The mosquito larvae and adults were maintained at 28 ± 3oC temperature with 70 ± 10% relative humidity at the insectary of Aklilu Lemma Institute of Pathobiology, Addis Ababa University. All equipment (cages, trays, incubators) containing mosquitoes have been deployed so that accidental contact and release was minimized.\nThe larvae (67 larvae/cm2) were kept in plastic trays (20 cm × 15 cm × 5 cm) containing de-chlorinated water (1.5 l) and were maintained under 12:12 h light and dark photoperiod cycles in the laboratory following the standard mosquito rearing procedure [35]. The larvae were fed with yeast (Saf-Instant yeast). The media were changed every three days. The pupae formed were collected by a glass beaker and transferred to 24.5 × 24.5 × 24.5 cm cages (Bug dorms) for adult emergence. Newly emerged adults were maintained in mosquito cages at 28 ± 3 °C temperature and 70 ± 10% relative humidity and fed sterile 10% sugar solution soaked in cotton pads within Petri dishes. The cotton was always kept moist with the solution and changed every day. In addition to sugar feeding, female mosquitoes were allowed to take blood meals from a restrained rabbit three times a week for egg development and oviposition. Moist filter papers in cups were placed inside rearing cages for oviposition by gravid female mosquitoes. The eggs were washed off with deionized water onto larval rearing trays and allowed to hatch into neonate larvae in the laboratory. Third to fourth instar larvae and bloodmeal starved 2 to 5 days old adult female An. stephensi were used continuously for larvicidal and adulticidal tests, respectively.\nLarvicidal and adulticidal bioassays were conducted with colony larvae and adults of An. stephensi originated from the Awash Arba area, eastern Ethiopia. The mosquito larvae and adults were maintained at 28 ± 3oC temperature with 70 ± 10% relative humidity at the insectary of Aklilu Lemma Institute of Pathobiology, Addis Ababa University. All equipment (cages, trays, incubators) containing mosquitoes have been deployed so that accidental contact and release was minimized.\nThe larvae (67 larvae/cm2) were kept in plastic trays (20 cm × 15 cm × 5 cm) containing de-chlorinated water (1.5 l) and were maintained under 12:12 h light and dark photoperiod cycles in the laboratory following the standard mosquito rearing procedure [35]. The larvae were fed with yeast (Saf-Instant yeast). The media were changed every three days. The pupae formed were collected by a glass beaker and transferred to 24.5 × 24.5 × 24.5 cm cages (Bug dorms) for adult emergence. Newly emerged adults were maintained in mosquito cages at 28 ± 3 °C temperature and 70 ± 10% relative humidity and fed sterile 10% sugar solution soaked in cotton pads within Petri dishes. The cotton was always kept moist with the solution and changed every day. In addition to sugar feeding, female mosquitoes were allowed to take blood meals from a restrained rabbit three times a week for egg development and oviposition. Moist filter papers in cups were placed inside rearing cages for oviposition by gravid female mosquitoes. The eggs were washed off with deionized water onto larval rearing trays and allowed to hatch into neonate larvae in the laboratory. Third to fourth instar larvae and bloodmeal starved 2 to 5 days old adult female An. stephensi were used continuously for larvicidal and adulticidal tests, respectively.\n[SUBTITLE] Preparation of stock solution [SUBSECTION] One gram of each crude plant extract was placed separately in 250 ml Wheaton glass bottles and dissolved in 100 ml of distilled water for methanol and aqueous extracts, while the hexane extracts were first dissolved in 4 ml of acetone and then added to 96 ml of distilled water to prepare 100 ml of a 1% stock solution. In the stock solution, one drop of emulsifier (Tween 80) was added to each extract at a concentration of 0.05%. From the 1% stock solution, concentrations of 25, 50, 100, 150, 200, 250, and 300 ppm for the larvae, and concentrations of 20, 40, 80, 160, and 320 ppm for the adult were prepared for exposure of the target mosquito.\nOne gram of each crude plant extract was placed separately in 250 ml Wheaton glass bottles and dissolved in 100 ml of distilled water for methanol and aqueous extracts, while the hexane extracts were first dissolved in 4 ml of acetone and then added to 96 ml of distilled water to prepare 100 ml of a 1% stock solution. In the stock solution, one drop of emulsifier (Tween 80) was added to each extract at a concentration of 0.05%. From the 1% stock solution, concentrations of 25, 50, 100, 150, 200, 250, and 300 ppm for the larvae, and concentrations of 20, 40, 80, 160, and 320 ppm for the adult were prepared for exposure of the target mosquito.\n[SUBTITLE] Larvicidal bioassay [SUBSECTION] Larvicidal activities of each leaf crude extract were measured according to WHO standard procedures [35]. For larvicidal bioassay, hexane, methanol and aqueous crude leaf extracts of the test plants were screened at 25, 50, 100, 150, 200, 250, and 300 ppm of test concentrations. Twenty individuals of late third to early fourth instar larvae were kept in a 300 ml enamel cups containing 99 ml of distilled water and 1-ml of desired concentrations of crude solvent extracts of C. aurea, M. foetida and Z. scabra were added. In the same way, 99 ml of distilled water with 1 ml mixture of acetone (for hexane extracts) and Tween 80 were made up to 100 ml to serve as a negative control. In addition, temephos at the rate of 0.25 ppm within a similar volume of test cups was used as a positive control [36]. The experiment was conducted for 24 h at the optimum conditions of 28 ± 3 °C temperature and 70 ± 10% relative humidity under 12:12 light and dark photoperiod in the insectary. During the exposure period, no food was offered to the larvae. Each experiment was run three times on different days along randomly set up with appropriate negative control and standard check. Numbers of dead larvae were counted after 24 h of exposure, and the percentage of mortality was reported from the averages of twelve replicates. The dead larvae included moribunds; those incapable of rising to the surface in each concentration of treatments, and the standards were combined separately and expressed as the average mortality to determine LC50 and LC90 values.\nLarvicidal activities of each leaf crude extract were measured according to WHO standard procedures [35]. For larvicidal bioassay, hexane, methanol and aqueous crude leaf extracts of the test plants were screened at 25, 50, 100, 150, 200, 250, and 300 ppm of test concentrations. Twenty individuals of late third to early fourth instar larvae were kept in a 300 ml enamel cups containing 99 ml of distilled water and 1-ml of desired concentrations of crude solvent extracts of C. aurea, M. foetida and Z. scabra were added. In the same way, 99 ml of distilled water with 1 ml mixture of acetone (for hexane extracts) and Tween 80 were made up to 100 ml to serve as a negative control. In addition, temephos at the rate of 0.25 ppm within a similar volume of test cups was used as a positive control [36]. The experiment was conducted for 24 h at the optimum conditions of 28 ± 3 °C temperature and 70 ± 10% relative humidity under 12:12 light and dark photoperiod in the insectary. During the exposure period, no food was offered to the larvae. Each experiment was run three times on different days along randomly set up with appropriate negative control and standard check. Numbers of dead larvae were counted after 24 h of exposure, and the percentage of mortality was reported from the averages of twelve replicates. The dead larvae included moribunds; those incapable of rising to the surface in each concentration of treatments, and the standards were combined separately and expressed as the average mortality to determine LC50 and LC90 values.\n[SUBTITLE] Adulticidal bioassay [SUBSECTION] Adulticidal activities of each crude solvent extract were performed using the CDC bottle bioassays [37, 38]. Most of the synthetic insecticides available for the control of adult mosquitoes, in particular An. stephensi were reported to be resisted by adults [12, 23], thus, this bioassay did not have any synthetic insecticide as a positive control. In this bioassay, aqueous, hexane and methanol crude leaf extracts of the three plants were screened at 25, 50, 100, 150, 200, 250, and 300 ppm concentrations. Two hundred fifty ml Wheaton bottles with screw lids were properly cleaned and dried, and then they were coated with 1ml solution of the desired concentrations of the tested extracts by swirling assuring complete coating of the bottle and its cap. Similarly, 1ml of acetone and emulsifier (Tween 80) solution was added to the control bottle handled as before.\nAdult mosquitoes (20, aged 2–5 days) fed on 10% sucrose solution were released to each bottle, including the control bottle with the help of mouth aspirator. The opening of each bottle was closed with their lids after introduction of mosquito adults. The exposure time was set to 2 h. After that, the mosquitoes were transferred into 250 ml capacity plastic cups using aspirator, where they were provided with 10% sucrose solution and the mortalities were checked after 24 h. The experiment was done at 28 ± 3 °C temperature and 70 ± 10% relative humidity under 12:12 light and dark photoperiod in the insectary. The percentage of adult mortality was corrected using Abbott’s formula [39] when needed.\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\% \\text{Morality} = \\frac{{\\% \\text{test}\\,\\text{morality} - \\% \\text{control}\\, \\text{morality}}}{{100 - \\% \\text{control}\\, \\text{morality}}} \\times 100. $$\\end{document}%Morality=%testmorality-%controlmorality100-%controlmorality×100.\nAdulticidal activities of each crude solvent extract were performed using the CDC bottle bioassays [37, 38]. Most of the synthetic insecticides available for the control of adult mosquitoes, in particular An. stephensi were reported to be resisted by adults [12, 23], thus, this bioassay did not have any synthetic insecticide as a positive control. In this bioassay, aqueous, hexane and methanol crude leaf extracts of the three plants were screened at 25, 50, 100, 150, 200, 250, and 300 ppm concentrations. Two hundred fifty ml Wheaton bottles with screw lids were properly cleaned and dried, and then they were coated with 1ml solution of the desired concentrations of the tested extracts by swirling assuring complete coating of the bottle and its cap. Similarly, 1ml of acetone and emulsifier (Tween 80) solution was added to the control bottle handled as before.\nAdult mosquitoes (20, aged 2–5 days) fed on 10% sucrose solution were released to each bottle, including the control bottle with the help of mouth aspirator. The opening of each bottle was closed with their lids after introduction of mosquito adults. The exposure time was set to 2 h. After that, the mosquitoes were transferred into 250 ml capacity plastic cups using aspirator, where they were provided with 10% sucrose solution and the mortalities were checked after 24 h. The experiment was done at 28 ± 3 °C temperature and 70 ± 10% relative humidity under 12:12 light and dark photoperiod in the insectary. The percentage of adult mortality was corrected using Abbott’s formula [39] when needed.\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\% \\text{Morality} = \\frac{{\\% \\text{test}\\,\\text{morality} - \\% \\text{control}\\, \\text{morality}}}{{100 - \\% \\text{control}\\, \\text{morality}}} \\times 100. $$\\end{document}%Morality=%testmorality-%controlmorality100-%controlmorality×100.\n[SUBTITLE] Data analysis [SUBSECTION] The mean percentage mortality was analysed using SPSS version 25.0 software (SPSS Inc, Chicago, IL, USA), and dose-dependent mortality also was performed by R software version 4.0.5. The percentage mortality of larvae and adult were checked for normality by 1-Sample Kolmogorov-Smirnov Z test (K-S). When the percent mortality did not conform to the normal distribution, the non-parametric equivalent tests of Kruskal-Wallis were followed. p-values were adjusted with the Bonferroni correction to adjust for the inflation of type I errors when several Mann–Whitney tests are performed [40]. The LC50 and LC90 values were calculated using a generalized linear probit model, considering a 95% confidence interval.\nThe mean percentage mortality was analysed using SPSS version 25.0 software (SPSS Inc, Chicago, IL, USA), and dose-dependent mortality also was performed by R software version 4.0.5. The percentage mortality of larvae and adult were checked for normality by 1-Sample Kolmogorov-Smirnov Z test (K-S). When the percent mortality did not conform to the normal distribution, the non-parametric equivalent tests of Kruskal-Wallis were followed. p-values were adjusted with the Bonferroni correction to adjust for the inflation of type I errors when several Mann–Whitney tests are performed [40]. The LC50 and LC90 values were calculated using a generalized linear probit model, considering a 95% confidence interval.", "[SUBTITLE] Collection of plant samples [SUBSECTION] The fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.\nThe fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.", "The fully developed fresh leaves of C. aurea and M. foetida were collected around Bahir Dar University campus located at 11°34′28.0″N, 37°23′53.4″E, at an altitude of 1801 m, while the leaves of Z. scabra were collected from Akaki District, Addis Ababa (8°49′40.5″N, 38°50′23.6″E, altitude 2117 m). The plant species were authenticated by a plant taxonomist from the National Herbarium in the Department of Plant Biology and Biodiversity Management, Addis Ababa University. The voucher specimens (MM-01 of M. foetida, MM-02 of C. aurea, and MM-03 of Z. scabra, respectively) were deposited at the National Herbarium, College of Natural and Computational Sciences of Addis Ababa University, Ethiopia.", "The leaves were washed with water and air-dried separately under shade at room temperature for two weeks in the Insect Sciences Laboratory, Addis Ababa University. Finally, the dried leaves were manually ground by pestle and mortar through a sieve into a fine powder. The leaf powder was kept at room temperature in labeled air-tight plastic bags until used.", "The crude extraction processes were conducted in the Organic Chemistry Laboratory of the Department of Chemistry, Addis Ababa University. Seventy-two grams of each powdered material was soaked separately in 720 ml of hexane, methanol, and distilled water at a ratio of 1:10 (W/V) in Erlenmeyer flask. Afterward, the mixtures were sonicated by ultrasonic cleaner apparatus (USC-T, Malaysia) at 20 kHz frequency with 11 W power twice per day for 15 min for two days. The mixtures were left to settle for 10 min and then cooled at room temperature. The supernatant of the hexane and methanol crude extracts were filtered through Whatman No.3 (Whatman International Ltd., Maid stone, England) filter paper, while the aqueous extracts using suction were filtered through a Buchner funnel with Whatman filter paper No.1. The filtrates of hexane and methanol crude extracts were concentrated using a vacuum rotary evaporator (Rota-vapor-RE, Buchi Labortechnik AG, Flawil, Switzerland) under reduced pressure at 40oC, while the aqueous crude extracts were evaporated to dryness using a lyophilizer. The residue obtained from each plant extract was left to cool at room temperature to remove traces of solvent, and then finally, were collected separately in a Wheaton bottle glass containers and were preserved a refrigerated at 4 °C until used for experimentation.", "Larvicidal and adulticidal bioassays were conducted with colony larvae and adults of An. stephensi originated from the Awash Arba area, eastern Ethiopia. The mosquito larvae and adults were maintained at 28 ± 3oC temperature with 70 ± 10% relative humidity at the insectary of Aklilu Lemma Institute of Pathobiology, Addis Ababa University. All equipment (cages, trays, incubators) containing mosquitoes have been deployed so that accidental contact and release was minimized.\nThe larvae (67 larvae/cm2) were kept in plastic trays (20 cm × 15 cm × 5 cm) containing de-chlorinated water (1.5 l) and were maintained under 12:12 h light and dark photoperiod cycles in the laboratory following the standard mosquito rearing procedure [35]. The larvae were fed with yeast (Saf-Instant yeast). The media were changed every three days. The pupae formed were collected by a glass beaker and transferred to 24.5 × 24.5 × 24.5 cm cages (Bug dorms) for adult emergence. Newly emerged adults were maintained in mosquito cages at 28 ± 3 °C temperature and 70 ± 10% relative humidity and fed sterile 10% sugar solution soaked in cotton pads within Petri dishes. The cotton was always kept moist with the solution and changed every day. In addition to sugar feeding, female mosquitoes were allowed to take blood meals from a restrained rabbit three times a week for egg development and oviposition. Moist filter papers in cups were placed inside rearing cages for oviposition by gravid female mosquitoes. The eggs were washed off with deionized water onto larval rearing trays and allowed to hatch into neonate larvae in the laboratory. Third to fourth instar larvae and bloodmeal starved 2 to 5 days old adult female An. stephensi were used continuously for larvicidal and adulticidal tests, respectively.", "One gram of each crude plant extract was placed separately in 250 ml Wheaton glass bottles and dissolved in 100 ml of distilled water for methanol and aqueous extracts, while the hexane extracts were first dissolved in 4 ml of acetone and then added to 96 ml of distilled water to prepare 100 ml of a 1% stock solution. In the stock solution, one drop of emulsifier (Tween 80) was added to each extract at a concentration of 0.05%. From the 1% stock solution, concentrations of 25, 50, 100, 150, 200, 250, and 300 ppm for the larvae, and concentrations of 20, 40, 80, 160, and 320 ppm for the adult were prepared for exposure of the target mosquito.", "Larvicidal activities of each leaf crude extract were measured according to WHO standard procedures [35]. For larvicidal bioassay, hexane, methanol and aqueous crude leaf extracts of the test plants were screened at 25, 50, 100, 150, 200, 250, and 300 ppm of test concentrations. Twenty individuals of late third to early fourth instar larvae were kept in a 300 ml enamel cups containing 99 ml of distilled water and 1-ml of desired concentrations of crude solvent extracts of C. aurea, M. foetida and Z. scabra were added. In the same way, 99 ml of distilled water with 1 ml mixture of acetone (for hexane extracts) and Tween 80 were made up to 100 ml to serve as a negative control. In addition, temephos at the rate of 0.25 ppm within a similar volume of test cups was used as a positive control [36]. The experiment was conducted for 24 h at the optimum conditions of 28 ± 3 °C temperature and 70 ± 10% relative humidity under 12:12 light and dark photoperiod in the insectary. During the exposure period, no food was offered to the larvae. Each experiment was run three times on different days along randomly set up with appropriate negative control and standard check. Numbers of dead larvae were counted after 24 h of exposure, and the percentage of mortality was reported from the averages of twelve replicates. The dead larvae included moribunds; those incapable of rising to the surface in each concentration of treatments, and the standards were combined separately and expressed as the average mortality to determine LC50 and LC90 values.", "Adulticidal activities of each crude solvent extract were performed using the CDC bottle bioassays [37, 38]. Most of the synthetic insecticides available for the control of adult mosquitoes, in particular An. stephensi were reported to be resisted by adults [12, 23], thus, this bioassay did not have any synthetic insecticide as a positive control. In this bioassay, aqueous, hexane and methanol crude leaf extracts of the three plants were screened at 25, 50, 100, 150, 200, 250, and 300 ppm concentrations. Two hundred fifty ml Wheaton bottles with screw lids were properly cleaned and dried, and then they were coated with 1ml solution of the desired concentrations of the tested extracts by swirling assuring complete coating of the bottle and its cap. Similarly, 1ml of acetone and emulsifier (Tween 80) solution was added to the control bottle handled as before.\nAdult mosquitoes (20, aged 2–5 days) fed on 10% sucrose solution were released to each bottle, including the control bottle with the help of mouth aspirator. The opening of each bottle was closed with their lids after introduction of mosquito adults. The exposure time was set to 2 h. After that, the mosquitoes were transferred into 250 ml capacity plastic cups using aspirator, where they were provided with 10% sucrose solution and the mortalities were checked after 24 h. The experiment was done at 28 ± 3 °C temperature and 70 ± 10% relative humidity under 12:12 light and dark photoperiod in the insectary. The percentage of adult mortality was corrected using Abbott’s formula [39] when needed.\\documentclass[12pt]{minimal}\n\t\t\t\t\\usepackage{amsmath}\n\t\t\t\t\\usepackage{wasysym} \n\t\t\t\t\\usepackage{amsfonts} \n\t\t\t\t\\usepackage{amssymb} \n\t\t\t\t\\usepackage{amsbsy}\n\t\t\t\t\\usepackage{mathrsfs}\n\t\t\t\t\\usepackage{upgreek}\n\t\t\t\t\\setlength{\\oddsidemargin}{-69pt}\n\t\t\t\t\\begin{document}$$\\% \\text{Morality} = \\frac{{\\% \\text{test}\\,\\text{morality} - \\% \\text{control}\\, \\text{morality}}}{{100 - \\% \\text{control}\\, \\text{morality}}} \\times 100. $$\\end{document}%Morality=%testmorality-%controlmorality100-%controlmorality×100.", "The mean percentage mortality was analysed using SPSS version 25.0 software (SPSS Inc, Chicago, IL, USA), and dose-dependent mortality also was performed by R software version 4.0.5. The percentage mortality of larvae and adult were checked for normality by 1-Sample Kolmogorov-Smirnov Z test (K-S). When the percent mortality did not conform to the normal distribution, the non-parametric equivalent tests of Kruskal-Wallis were followed. p-values were adjusted with the Bonferroni correction to adjust for the inflation of type I errors when several Mann–Whitney tests are performed [40]. The LC50 and LC90 values were calculated using a generalized linear probit model, considering a 95% confidence interval.", "[SUBTITLE] Larvicidal activity of plant extracts against An. stephensi [SUBSECTION] Larval mortalities obtained from bioassays of aqueous and methanol extracts of Calpurnia aurea, Momordica foetida, and Zehneria scabra leaves against An. stephensi larvae after 24 h exposure periods are presented in Tables 1, 2 and 3. All aqueous and methanol test plant extracts showed low, moderate, and high larvicidal activities tested between 25 ppm-300 ppm treatments against the late 3rd to early 4th instar larvae of An. stephensi. Within the same exposure period, larval mortality was not observed for the tests using hexane crude leaf extracts and the negative control. The standard positive control (temephos) achieved 100% larval mortality.\n\nTable 1Mean % larval mortality of Calpurnia aurea crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol 2535.00 ± 2.6Aa23.75 ± 3.65Aa5060.00 ± 3.54Ba35.00 ± 0.00Bb10086.25 ± 2.23Ca90.42 ± 1.4Ca15099.58 ± 0.42Da95.00 ± 2.46DCa200100.00 ± 0.00Da96.67 ± 1.42Da250100.00 ± 0.00Da100.00 ± 0.00Da300100.00 ± 0.00Da100.00 ± 0.00DaTemephos (0.25)100.00 ± 0.00Da100.00 ± 0.00DaControl0.00 ± 0.00Ea0.00 ± 0.00Ea*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p > 0.003)\nMean % larval mortality of Calpurnia aurea crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure\n*Each value of % mean mortality ± SE represents the value of twelve replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p > 0.003)\nThere was a significant difference and interaction effects between all aqueous and methanol test plant extract against the tested larvae (Kruskal-Wallis test, p-value < 0.05). The mean percent larval mortality between treatments, control, and the standard groups had a statistically significant difference (Kruskal-Wallis test, p-value < 0.05). The mortality effect of the test plant extracts against An. stephensi larvae were dose and extraction solvent dependent.\nIn the C. aurea test plant, the highest (100%) larval mortality was found both in aqueous and methanol extracts at 200 ppm and 250 ppm of treatments, respectively (Table 1). Even at 150 ppm, the larvicidal effects of aqueous and methanol extracts of C. aurea were not significantly different from standard checks (Multiple-Mann Whitney U test, p-value > 0.003; Table 1). Yet, all C. aurea treatments significantly affected larval mortality compared to the negative control (Multiple-Mann Whitney U test; p < 0.003).\nAqueous and methanol extracts of the M. foetida test plant showed 100% larval mortality at 200 ppm and 250 ppm of treatments, respectively (Table 2). Moreover, larvicidal activity found from aqueous extracts of M. foetida at 100 ppm and methanol extracts at 200 ppm caused mortality (ranging 95–97.9%) was not significantly different from the mortality achieved in temephos exposed on An. stephensi larvae (Multiple-Mann Whitney U test, p-value > 0.003; Table 2). Statistical differences were observed between all M. foetida treatments and negative control (Multiple-Mann Whitney U test; p-value < 0.003).\n\nTable 2Mean % larval mortality of Momordica foetida crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol 2527.5 ± 2.85Aa4.55 ± 1.4Ab5080.83 ± 4.51Ba20.83 ± 2.53Bb10095.00 ± 1.74Ca45.83 ± 2.20Cb15097.92 ± 0.96Ca71.67 ± 3.39Db200100.00 ± 0.00Ca97.5 ± 2.50Ea250100.00 ± 0.00Ca100.00 ± 0.00Ea300100.00 ± 0.00Ca100.00 ± 0.00EaTemephos (0.25)100.00 ± 0.00Ca100.00 ± 0.00EaControl0.00 ± 0.00Da0.00 ± 0.00Fa*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)Table 3Mean % larval mortality of Zehneria scabra crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE\nSolvents\nAqueous \nMethanol 2529.17 ± 3.68Aa23.75 ± 2.69Aa5073.33 ± 3.9Ba66.25 ± 5.04Ba10095.42 ± 1.44Ca87.08 ± 2.17Ca15097.08 ± 1.44Ca92.50 ± 1.44Da20099.50 ± 0.42Ca99.17 ± 0.83Ea250100.00 ± 0.00Ca100.00 ± 0.00Ea300100.00 ± 0.00Ca100.00 ± 0.00EaTemephos (0.25)100.00 ± 0.00Ca100.00 ± 0.00EaControl0.00 ± 0.00Da0.00 ± 0.00Fa*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % larval mortality of Momordica foetida crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure\n*Each value of % mean mortality ± SE represents the value of twelve replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % larval mortality of Zehneria scabra crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure\n*Each value of % mean mortality ± SE represents the value of twelve replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nAmong the two solvent test concentrations of Z. scabra extracts, statistically, non-significant differences were observed (Multiple-Mann Whitney U test, p-value > 0.003; Table 3). However, their larvicidal activity was significantly different from negative control (Multiple-Mann Whitney U test; p-value < 0.003). At the highest concentrations (250 ppm), both aqueous and methanol Z. scabra, crude leaf extracts resulted in 100% larval mortality (Table 3). Moreover, aqueous extracts of Z. scabra at 100 ppm, and methanol extracts at 200 ppm, each caused more than 95% larval mortality, had no significant effect compared to mortality achieved in temephos treatment (Multiple-Mann Whitney U test; p-value > 0.003;).\nThe lethal toxicity doses of aqueous and methanol crude leaf extracts of C. aurea, M. foetida, and Z. scabra against larvae of An. stephensi are shown in Table 4. Crude aqueous leaf extract of M. foetida showed strong larvicidal activity, having an LC50 value of 34.61 ppm and LC90 value of 57.61 ppm, followed by Z. scabra (LC50 = 35.85 ppm; LC90 = 68.26 ppm) and C. aurea (LC50 = 38.69 ppm; LC90 = 108.28 ppm). Similarly, the methanol leaf extract of Z. scabra had good larvicidal activities with LC50 and LC90 values of 41.32 ppm and 99.07 ppm, followed by C. aurea (LC50 = 43.25 ppm; LC90 = 96.02 ppm). However, the crude methanol leaf extract of M. foetida had lower larvicidal toxicity against the larvae of An. stephensi compared to other test plant extracts (LC50 = 99.50 ppm; LC90 = 188.76 ppm, Table 4).\n\nTable 4LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of tests plants against the late third to early fourth instar larvae of Anopheles stephensiTest plantsSolventsLC50 (ppm)95%Confidence limitLC90 (ppm)95%Confidence limitχ2 (df = 5)\nLCL \nUCL \nLCL \nUCL \nC. aureaAqueous38. 6935. 5741.82108. 2896. 27120. 302.93Methanol43. 2540. 4946. 0296. 0283. 87108. 184.15\nM. foetidaAqueous34. 6132. 8436. 3857. 6153. 2961. 930.71Methanol99. 5094. 56104. 43188. 76177. 70199. 826.90\nZ. scabraAqueous35. 8533. 7937. 9168. 2660. 2076. 320.37Methanol41. 3238. 5144. 1299. 0784. 64113. 501.43\nLC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of tests plants against the late third to early fourth instar larvae of Anopheles stephensi\nLarval mortalities obtained from bioassays of aqueous and methanol extracts of Calpurnia aurea, Momordica foetida, and Zehneria scabra leaves against An. stephensi larvae after 24 h exposure periods are presented in Tables 1, 2 and 3. All aqueous and methanol test plant extracts showed low, moderate, and high larvicidal activities tested between 25 ppm-300 ppm treatments against the late 3rd to early 4th instar larvae of An. stephensi. Within the same exposure period, larval mortality was not observed for the tests using hexane crude leaf extracts and the negative control. The standard positive control (temephos) achieved 100% larval mortality.\n\nTable 1Mean % larval mortality of Calpurnia aurea crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol 2535.00 ± 2.6Aa23.75 ± 3.65Aa5060.00 ± 3.54Ba35.00 ± 0.00Bb10086.25 ± 2.23Ca90.42 ± 1.4Ca15099.58 ± 0.42Da95.00 ± 2.46DCa200100.00 ± 0.00Da96.67 ± 1.42Da250100.00 ± 0.00Da100.00 ± 0.00Da300100.00 ± 0.00Da100.00 ± 0.00DaTemephos (0.25)100.00 ± 0.00Da100.00 ± 0.00DaControl0.00 ± 0.00Ea0.00 ± 0.00Ea*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p > 0.003)\nMean % larval mortality of Calpurnia aurea crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure\n*Each value of % mean mortality ± SE represents the value of twelve replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p > 0.003)\nThere was a significant difference and interaction effects between all aqueous and methanol test plant extract against the tested larvae (Kruskal-Wallis test, p-value < 0.05). The mean percent larval mortality between treatments, control, and the standard groups had a statistically significant difference (Kruskal-Wallis test, p-value < 0.05). The mortality effect of the test plant extracts against An. stephensi larvae were dose and extraction solvent dependent.\nIn the C. aurea test plant, the highest (100%) larval mortality was found both in aqueous and methanol extracts at 200 ppm and 250 ppm of treatments, respectively (Table 1). Even at 150 ppm, the larvicidal effects of aqueous and methanol extracts of C. aurea were not significantly different from standard checks (Multiple-Mann Whitney U test, p-value > 0.003; Table 1). Yet, all C. aurea treatments significantly affected larval mortality compared to the negative control (Multiple-Mann Whitney U test; p < 0.003).\nAqueous and methanol extracts of the M. foetida test plant showed 100% larval mortality at 200 ppm and 250 ppm of treatments, respectively (Table 2). Moreover, larvicidal activity found from aqueous extracts of M. foetida at 100 ppm and methanol extracts at 200 ppm caused mortality (ranging 95–97.9%) was not significantly different from the mortality achieved in temephos exposed on An. stephensi larvae (Multiple-Mann Whitney U test, p-value > 0.003; Table 2). Statistical differences were observed between all M. foetida treatments and negative control (Multiple-Mann Whitney U test; p-value < 0.003).\n\nTable 2Mean % larval mortality of Momordica foetida crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol 2527.5 ± 2.85Aa4.55 ± 1.4Ab5080.83 ± 4.51Ba20.83 ± 2.53Bb10095.00 ± 1.74Ca45.83 ± 2.20Cb15097.92 ± 0.96Ca71.67 ± 3.39Db200100.00 ± 0.00Ca97.5 ± 2.50Ea250100.00 ± 0.00Ca100.00 ± 0.00Ea300100.00 ± 0.00Ca100.00 ± 0.00EaTemephos (0.25)100.00 ± 0.00Ca100.00 ± 0.00EaControl0.00 ± 0.00Da0.00 ± 0.00Fa*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)Table 3Mean % larval mortality of Zehneria scabra crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE\nSolvents\nAqueous \nMethanol 2529.17 ± 3.68Aa23.75 ± 2.69Aa5073.33 ± 3.9Ba66.25 ± 5.04Ba10095.42 ± 1.44Ca87.08 ± 2.17Ca15097.08 ± 1.44Ca92.50 ± 1.44Da20099.50 ± 0.42Ca99.17 ± 0.83Ea250100.00 ± 0.00Ca100.00 ± 0.00Ea300100.00 ± 0.00Ca100.00 ± 0.00EaTemephos (0.25)100.00 ± 0.00Ca100.00 ± 0.00EaControl0.00 ± 0.00Da0.00 ± 0.00Fa*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % larval mortality of Momordica foetida crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure\n*Each value of % mean mortality ± SE represents the value of twelve replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % larval mortality of Zehneria scabra crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure\n*Each value of % mean mortality ± SE represents the value of twelve replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nAmong the two solvent test concentrations of Z. scabra extracts, statistically, non-significant differences were observed (Multiple-Mann Whitney U test, p-value > 0.003; Table 3). However, their larvicidal activity was significantly different from negative control (Multiple-Mann Whitney U test; p-value < 0.003). At the highest concentrations (250 ppm), both aqueous and methanol Z. scabra, crude leaf extracts resulted in 100% larval mortality (Table 3). Moreover, aqueous extracts of Z. scabra at 100 ppm, and methanol extracts at 200 ppm, each caused more than 95% larval mortality, had no significant effect compared to mortality achieved in temephos treatment (Multiple-Mann Whitney U test; p-value > 0.003;).\nThe lethal toxicity doses of aqueous and methanol crude leaf extracts of C. aurea, M. foetida, and Z. scabra against larvae of An. stephensi are shown in Table 4. Crude aqueous leaf extract of M. foetida showed strong larvicidal activity, having an LC50 value of 34.61 ppm and LC90 value of 57.61 ppm, followed by Z. scabra (LC50 = 35.85 ppm; LC90 = 68.26 ppm) and C. aurea (LC50 = 38.69 ppm; LC90 = 108.28 ppm). Similarly, the methanol leaf extract of Z. scabra had good larvicidal activities with LC50 and LC90 values of 41.32 ppm and 99.07 ppm, followed by C. aurea (LC50 = 43.25 ppm; LC90 = 96.02 ppm). However, the crude methanol leaf extract of M. foetida had lower larvicidal toxicity against the larvae of An. stephensi compared to other test plant extracts (LC50 = 99.50 ppm; LC90 = 188.76 ppm, Table 4).\n\nTable 4LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of tests plants against the late third to early fourth instar larvae of Anopheles stephensiTest plantsSolventsLC50 (ppm)95%Confidence limitLC90 (ppm)95%Confidence limitχ2 (df = 5)\nLCL \nUCL \nLCL \nUCL \nC. aureaAqueous38. 6935. 5741.82108. 2896. 27120. 302.93Methanol43. 2540. 4946. 0296. 0283. 87108. 184.15\nM. foetidaAqueous34. 6132. 8436. 3857. 6153. 2961. 930.71Methanol99. 5094. 56104. 43188. 76177. 70199. 826.90\nZ. scabraAqueous35. 8533. 7937. 9168. 2660. 2076. 320.37Methanol41. 3238. 5144. 1299. 0784. 64113. 501.43\nLC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of tests plants against the late third to early fourth instar larvae of Anopheles stephensi\n[SUBTITLE] The adulticidal activity of plant extracts against An. stephensi [SUBSECTION] Tables 5, 6 and 7 show the mean percentage mortalities and toxicity effects of adult An. stephensi after 24 h exposures to different solvent extracts of C. aurea, M. foetida, and Z. scabra. Statistically significant differences in mean percentage mortality among the different extracts and the negative control were observed (Kruskal-Wallis test, p < 0.05). The adulticidal data showed that the mortality rate of the vector was directly proportional to the concentration and extraction solvents. All hexane test plant extracts showed lower adulticidal property against An. stephensi (≤ 20% at all treatments tested in the bioassays, Tables 5, 6 and 7). Between the different solvent leaf extracts of C. aurea, only aqueous extract gave 55% adult mortality of An. stephensi at 320 ppm, while other crude solvent leaf extracts had lower adulticidal activity, having adult mean mortalities ranging from 0 to 48% (Table 5).\n\nTable 5Mean % adult mortality of Calpurnia aurea crude leaf solvent extracts at different treatments against An. stephensiConcentration (ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol \nHexane 201.67 ± 1.67Aa0.00 ± 0.00Aa0.00 ± 0.00Aa406.67 ± 1.67Aa6.67 ± 1.67Ba0.00 ± 0.00Ab8016.67 ± 1.67Ba16.67 ± 1.67Ca0.00 ± 0.00Ab16021.67 ± 1.67Ba26.67 ± 3.33Da6.67 ± 3.33Bb32055.00 ± 5.77Ca48.33 ± 1.67Ea18.33 ± 1.67CbControl0.00 ± 0.00Da0.00 ± 0.00Aa0.00 ± 0.00Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % adult mortality of Calpurnia aurea crude leaf solvent extracts at different treatments against An. stephensi\n*Each value of % mean mortality ± SE represents the value of three replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\n\nDifferent solvent (aqueous, methanol, and hexane) crude leaf extracts of M. foetida resulted in lower adult mortalities of An. stephensi (< 24%) at all treatments tested in the bioassays and were not effective against adults of malaria vector An. stephensi (Table 6).\n\nTable 6Mean % adult mortality of Momordica foetida crude leaf solvent extract at different concentrations against Anopheles stephensiConcentration(ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol \nHexane 200.00 ± 0.00ABa0.00 ± 0.00Aa0.00 ± 0.00Aa401.67 ± 1.67Ba0.00 ± 0.00Aa0.00 ± 0.00Ab805.00 ± 2.89Ba1.67 ± 1.67Ba0.00 ± 0.00Ab16013.33 ± 1.67Ca13.33 ± 1.67Ca0.00 ± 0.00Ab32018.33 ± 4.41Ca23.33 ± 1.67Da1.67 ± 1.67BbControl0.00 ± 0.00Aa0.00 ± 0.00Aa0.00 ± 0.00Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % adult mortality of Momordica foetida crude leaf solvent extract at different concentrations against Anopheles stephensi\n*Each value of % mean mortality ± SE represents the value of three replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nThere were statistically significant differences in mean percentage mortality among different concentrations of aqueous, methanol, and hexane leaf extracts of Z. scabra and with negative control (Kruskal-Wallis test; p-value < 0.05; Table 7). Within the given time intervals adult mortality was recorded for the control treatment. However, the highest adult mortality (84%) at higher concentration 320 ppm against An. stephensi was recorded in aqueous crude leaf extract of Z. scabra followed by its methanol extracts (65%) and their adulticidal potential was statistically different from the negative control (Multiple-Mann Whitney U-test; p-value < 0.003, Table 7).Table 7Mean % adult mortality of Zehneria scabra crude leaf solvent extracts at different concentrations against Anopheles stephensiConcentration (ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol \nHexane 204.33 ± 2.6Aa11.67 ± 3.33Aa0.00 ± 0.00Aa4011.67 ± 1.67Ba20.00 ± 2.89ABa0.00 ± 0.00Ab8026.67 ± 3.33Ca23.33 ± 3.33Ba0.00 ± 0.00Ab16036.67 ± 6.00Ca41.66 ± 4.40Ca15.00 ± 2.89Bb32084.00 ± 2.89Da65.00 ± 5.77Db20.00 ± 2.89BcControl0.67 ± 0.45Ea0.67 ± 0.45Ea0.67 ± 0.45Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % adult mortality of Zehneria scabra crude leaf solvent extracts at different concentrations against Anopheles stephensi\n*Each value of % mean mortality ± SE represents the value of three replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nThe lethal effects of different crude leaf extracts against adults of An. stephensi are shown in Table 8. All hexane and M. foetida crude leaf extracts had low adulticidal impacts, only aqueous and methanol C. aurea and Z. scabra leaf extracts were subjected to dose-response bioassay to detect the lethal concentrations. From the dose-response curves, the lowest LC50 (= 176.20 ppm and 205.41 ppm) and LC90 (= 425.13 and 883.11 ppm) values were demonstrated from the aqueous and methanol Z. scabra crude leaf extracts, respectively (Table 8), and its relative toxicities against adult An. stephensi was much higher than the rest of the plant extracts. The highest LC50 (= 297.75 ppm and 333.27 ppm) and LC90 (= 762.63 ppm and 1031.74 ppm) values were from the aqueous and methanol C. aurea crude leaf extracts, respectively, and its relative toxicity on An. stephensi was much lower than Z. scabra crude extracts.\n\nTable 8LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of Calpurnia aurea and Zehneria scabra against adults of Anopheles stephensiTest plantsSolventsLC5095%Confidence limitLC9095%Confidence limitχ2 (df = 3)\nLCL \nUCL \nLCL \nUCL \nC. aureaAqueous297. 75254. 95340. 56762. 63491. 131034. 130.88Methanol333. 27268. 92397. 631031. 74569. 061494. 430.75\nZ. scabraAqueous176.20157. 82194. 58425. 13346. 14504. 122.70Methanol205. 41172. 29238. 54883. 11529. 331236. 900.70\nLC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of Calpurnia aurea and Zehneria scabra against adults of Anopheles stephensi\nTables 5, 6 and 7 show the mean percentage mortalities and toxicity effects of adult An. stephensi after 24 h exposures to different solvent extracts of C. aurea, M. foetida, and Z. scabra. Statistically significant differences in mean percentage mortality among the different extracts and the negative control were observed (Kruskal-Wallis test, p < 0.05). The adulticidal data showed that the mortality rate of the vector was directly proportional to the concentration and extraction solvents. All hexane test plant extracts showed lower adulticidal property against An. stephensi (≤ 20% at all treatments tested in the bioassays, Tables 5, 6 and 7). Between the different solvent leaf extracts of C. aurea, only aqueous extract gave 55% adult mortality of An. stephensi at 320 ppm, while other crude solvent leaf extracts had lower adulticidal activity, having adult mean mortalities ranging from 0 to 48% (Table 5).\n\nTable 5Mean % adult mortality of Calpurnia aurea crude leaf solvent extracts at different treatments against An. stephensiConcentration (ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol \nHexane 201.67 ± 1.67Aa0.00 ± 0.00Aa0.00 ± 0.00Aa406.67 ± 1.67Aa6.67 ± 1.67Ba0.00 ± 0.00Ab8016.67 ± 1.67Ba16.67 ± 1.67Ca0.00 ± 0.00Ab16021.67 ± 1.67Ba26.67 ± 3.33Da6.67 ± 3.33Bb32055.00 ± 5.77Ca48.33 ± 1.67Ea18.33 ± 1.67CbControl0.00 ± 0.00Da0.00 ± 0.00Aa0.00 ± 0.00Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % adult mortality of Calpurnia aurea crude leaf solvent extracts at different treatments against An. stephensi\n*Each value of % mean mortality ± SE represents the value of three replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\n\nDifferent solvent (aqueous, methanol, and hexane) crude leaf extracts of M. foetida resulted in lower adult mortalities of An. stephensi (< 24%) at all treatments tested in the bioassays and were not effective against adults of malaria vector An. stephensi (Table 6).\n\nTable 6Mean % adult mortality of Momordica foetida crude leaf solvent extract at different concentrations against Anopheles stephensiConcentration(ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol \nHexane 200.00 ± 0.00ABa0.00 ± 0.00Aa0.00 ± 0.00Aa401.67 ± 1.67Ba0.00 ± 0.00Aa0.00 ± 0.00Ab805.00 ± 2.89Ba1.67 ± 1.67Ba0.00 ± 0.00Ab16013.33 ± 1.67Ca13.33 ± 1.67Ca0.00 ± 0.00Ab32018.33 ± 4.41Ca23.33 ± 1.67Da1.67 ± 1.67BbControl0.00 ± 0.00Aa0.00 ± 0.00Aa0.00 ± 0.00Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % adult mortality of Momordica foetida crude leaf solvent extract at different concentrations against Anopheles stephensi\n*Each value of % mean mortality ± SE represents the value of three replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nThere were statistically significant differences in mean percentage mortality among different concentrations of aqueous, methanol, and hexane leaf extracts of Z. scabra and with negative control (Kruskal-Wallis test; p-value < 0.05; Table 7). Within the given time intervals adult mortality was recorded for the control treatment. However, the highest adult mortality (84%) at higher concentration 320 ppm against An. stephensi was recorded in aqueous crude leaf extract of Z. scabra followed by its methanol extracts (65%) and their adulticidal potential was statistically different from the negative control (Multiple-Mann Whitney U-test; p-value < 0.003, Table 7).Table 7Mean % adult mortality of Zehneria scabra crude leaf solvent extracts at different concentrations against Anopheles stephensiConcentration (ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol \nHexane 204.33 ± 2.6Aa11.67 ± 3.33Aa0.00 ± 0.00Aa4011.67 ± 1.67Ba20.00 ± 2.89ABa0.00 ± 0.00Ab8026.67 ± 3.33Ca23.33 ± 3.33Ba0.00 ± 0.00Ab16036.67 ± 6.00Ca41.66 ± 4.40Ca15.00 ± 2.89Bb32084.00 ± 2.89Da65.00 ± 5.77Db20.00 ± 2.89BcControl0.67 ± 0.45Ea0.67 ± 0.45Ea0.67 ± 0.45Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % adult mortality of Zehneria scabra crude leaf solvent extracts at different concentrations against Anopheles stephensi\n*Each value of % mean mortality ± SE represents the value of three replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nThe lethal effects of different crude leaf extracts against adults of An. stephensi are shown in Table 8. All hexane and M. foetida crude leaf extracts had low adulticidal impacts, only aqueous and methanol C. aurea and Z. scabra leaf extracts were subjected to dose-response bioassay to detect the lethal concentrations. From the dose-response curves, the lowest LC50 (= 176.20 ppm and 205.41 ppm) and LC90 (= 425.13 and 883.11 ppm) values were demonstrated from the aqueous and methanol Z. scabra crude leaf extracts, respectively (Table 8), and its relative toxicities against adult An. stephensi was much higher than the rest of the plant extracts. The highest LC50 (= 297.75 ppm and 333.27 ppm) and LC90 (= 762.63 ppm and 1031.74 ppm) values were from the aqueous and methanol C. aurea crude leaf extracts, respectively, and its relative toxicity on An. stephensi was much lower than Z. scabra crude extracts.\n\nTable 8LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of Calpurnia aurea and Zehneria scabra against adults of Anopheles stephensiTest plantsSolventsLC5095%Confidence limitLC9095%Confidence limitχ2 (df = 3)\nLCL \nUCL \nLCL \nUCL \nC. aureaAqueous297. 75254. 95340. 56762. 63491. 131034. 130.88Methanol333. 27268. 92397. 631031. 74569. 061494. 430.75\nZ. scabraAqueous176.20157. 82194. 58425. 13346. 14504. 122.70Methanol205. 41172. 29238. 54883. 11529. 331236. 900.70\nLC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of Calpurnia aurea and Zehneria scabra against adults of Anopheles stephensi", "Larval mortalities obtained from bioassays of aqueous and methanol extracts of Calpurnia aurea, Momordica foetida, and Zehneria scabra leaves against An. stephensi larvae after 24 h exposure periods are presented in Tables 1, 2 and 3. All aqueous and methanol test plant extracts showed low, moderate, and high larvicidal activities tested between 25 ppm-300 ppm treatments against the late 3rd to early 4th instar larvae of An. stephensi. Within the same exposure period, larval mortality was not observed for the tests using hexane crude leaf extracts and the negative control. The standard positive control (temephos) achieved 100% larval mortality.\n\nTable 1Mean % larval mortality of Calpurnia aurea crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol 2535.00 ± 2.6Aa23.75 ± 3.65Aa5060.00 ± 3.54Ba35.00 ± 0.00Bb10086.25 ± 2.23Ca90.42 ± 1.4Ca15099.58 ± 0.42Da95.00 ± 2.46DCa200100.00 ± 0.00Da96.67 ± 1.42Da250100.00 ± 0.00Da100.00 ± 0.00Da300100.00 ± 0.00Da100.00 ± 0.00DaTemephos (0.25)100.00 ± 0.00Da100.00 ± 0.00DaControl0.00 ± 0.00Ea0.00 ± 0.00Ea*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p > 0.003)\nMean % larval mortality of Calpurnia aurea crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure\n*Each value of % mean mortality ± SE represents the value of twelve replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p > 0.003)\nThere was a significant difference and interaction effects between all aqueous and methanol test plant extract against the tested larvae (Kruskal-Wallis test, p-value < 0.05). The mean percent larval mortality between treatments, control, and the standard groups had a statistically significant difference (Kruskal-Wallis test, p-value < 0.05). The mortality effect of the test plant extracts against An. stephensi larvae were dose and extraction solvent dependent.\nIn the C. aurea test plant, the highest (100%) larval mortality was found both in aqueous and methanol extracts at 200 ppm and 250 ppm of treatments, respectively (Table 1). Even at 150 ppm, the larvicidal effects of aqueous and methanol extracts of C. aurea were not significantly different from standard checks (Multiple-Mann Whitney U test, p-value > 0.003; Table 1). Yet, all C. aurea treatments significantly affected larval mortality compared to the negative control (Multiple-Mann Whitney U test; p < 0.003).\nAqueous and methanol extracts of the M. foetida test plant showed 100% larval mortality at 200 ppm and 250 ppm of treatments, respectively (Table 2). Moreover, larvicidal activity found from aqueous extracts of M. foetida at 100 ppm and methanol extracts at 200 ppm caused mortality (ranging 95–97.9%) was not significantly different from the mortality achieved in temephos exposed on An. stephensi larvae (Multiple-Mann Whitney U test, p-value > 0.003; Table 2). Statistical differences were observed between all M. foetida treatments and negative control (Multiple-Mann Whitney U test; p-value < 0.003).\n\nTable 2Mean % larval mortality of Momordica foetida crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol 2527.5 ± 2.85Aa4.55 ± 1.4Ab5080.83 ± 4.51Ba20.83 ± 2.53Bb10095.00 ± 1.74Ca45.83 ± 2.20Cb15097.92 ± 0.96Ca71.67 ± 3.39Db200100.00 ± 0.00Ca97.5 ± 2.50Ea250100.00 ± 0.00Ca100.00 ± 0.00Ea300100.00 ± 0.00Ca100.00 ± 0.00EaTemephos (0.25)100.00 ± 0.00Ca100.00 ± 0.00EaControl0.00 ± 0.00Da0.00 ± 0.00Fa*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)Table 3Mean % larval mortality of Zehneria scabra crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposureConcentration(ppm)% Mean mortality ± SE\nSolvents\nAqueous \nMethanol 2529.17 ± 3.68Aa23.75 ± 2.69Aa5073.33 ± 3.9Ba66.25 ± 5.04Ba10095.42 ± 1.44Ca87.08 ± 2.17Ca15097.08 ± 1.44Ca92.50 ± 1.44Da20099.50 ± 0.42Ca99.17 ± 0.83Ea250100.00 ± 0.00Ca100.00 ± 0.00Ea300100.00 ± 0.00Ca100.00 ± 0.00EaTemephos (0.25)100.00 ± 0.00Ca100.00 ± 0.00EaControl0.00 ± 0.00Da0.00 ± 0.00Fa*Each value of % mean mortality ± SE represents the value of twelve replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % larval mortality of Momordica foetida crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure\n*Each value of % mean mortality ± SE represents the value of twelve replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % larval mortality of Zehneria scabra crude leaf solvent extracts at different rates against the late 3rd and early 4th instar Anopheles stephensi larvae after 24 h of exposure\n*Each value of % mean mortality ± SE represents the value of twelve replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nAmong the two solvent test concentrations of Z. scabra extracts, statistically, non-significant differences were observed (Multiple-Mann Whitney U test, p-value > 0.003; Table 3). However, their larvicidal activity was significantly different from negative control (Multiple-Mann Whitney U test; p-value < 0.003). At the highest concentrations (250 ppm), both aqueous and methanol Z. scabra, crude leaf extracts resulted in 100% larval mortality (Table 3). Moreover, aqueous extracts of Z. scabra at 100 ppm, and methanol extracts at 200 ppm, each caused more than 95% larval mortality, had no significant effect compared to mortality achieved in temephos treatment (Multiple-Mann Whitney U test; p-value > 0.003;).\nThe lethal toxicity doses of aqueous and methanol crude leaf extracts of C. aurea, M. foetida, and Z. scabra against larvae of An. stephensi are shown in Table 4. Crude aqueous leaf extract of M. foetida showed strong larvicidal activity, having an LC50 value of 34.61 ppm and LC90 value of 57.61 ppm, followed by Z. scabra (LC50 = 35.85 ppm; LC90 = 68.26 ppm) and C. aurea (LC50 = 38.69 ppm; LC90 = 108.28 ppm). Similarly, the methanol leaf extract of Z. scabra had good larvicidal activities with LC50 and LC90 values of 41.32 ppm and 99.07 ppm, followed by C. aurea (LC50 = 43.25 ppm; LC90 = 96.02 ppm). However, the crude methanol leaf extract of M. foetida had lower larvicidal toxicity against the larvae of An. stephensi compared to other test plant extracts (LC50 = 99.50 ppm; LC90 = 188.76 ppm, Table 4).\n\nTable 4LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of tests plants against the late third to early fourth instar larvae of Anopheles stephensiTest plantsSolventsLC50 (ppm)95%Confidence limitLC90 (ppm)95%Confidence limitχ2 (df = 5)\nLCL \nUCL \nLCL \nUCL \nC. aureaAqueous38. 6935. 5741.82108. 2896. 27120. 302.93Methanol43. 2540. 4946. 0296. 0283. 87108. 184.15\nM. foetidaAqueous34. 6132. 8436. 3857. 6153. 2961. 930.71Methanol99. 5094. 56104. 43188. 76177. 70199. 826.90\nZ. scabraAqueous35. 8533. 7937. 9168. 2660. 2076. 320.37Methanol41. 3238. 5144. 1299. 0784. 64113. 501.43\nLC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of tests plants against the late third to early fourth instar larvae of Anopheles stephensi", "Tables 5, 6 and 7 show the mean percentage mortalities and toxicity effects of adult An. stephensi after 24 h exposures to different solvent extracts of C. aurea, M. foetida, and Z. scabra. Statistically significant differences in mean percentage mortality among the different extracts and the negative control were observed (Kruskal-Wallis test, p < 0.05). The adulticidal data showed that the mortality rate of the vector was directly proportional to the concentration and extraction solvents. All hexane test plant extracts showed lower adulticidal property against An. stephensi (≤ 20% at all treatments tested in the bioassays, Tables 5, 6 and 7). Between the different solvent leaf extracts of C. aurea, only aqueous extract gave 55% adult mortality of An. stephensi at 320 ppm, while other crude solvent leaf extracts had lower adulticidal activity, having adult mean mortalities ranging from 0 to 48% (Table 5).\n\nTable 5Mean % adult mortality of Calpurnia aurea crude leaf solvent extracts at different treatments against An. stephensiConcentration (ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol \nHexane 201.67 ± 1.67Aa0.00 ± 0.00Aa0.00 ± 0.00Aa406.67 ± 1.67Aa6.67 ± 1.67Ba0.00 ± 0.00Ab8016.67 ± 1.67Ba16.67 ± 1.67Ca0.00 ± 0.00Ab16021.67 ± 1.67Ba26.67 ± 3.33Da6.67 ± 3.33Bb32055.00 ± 5.77Ca48.33 ± 1.67Ea18.33 ± 1.67CbControl0.00 ± 0.00Da0.00 ± 0.00Aa0.00 ± 0.00Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % adult mortality of Calpurnia aurea crude leaf solvent extracts at different treatments against An. stephensi\n*Each value of % mean mortality ± SE represents the value of three replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\n\nDifferent solvent (aqueous, methanol, and hexane) crude leaf extracts of M. foetida resulted in lower adult mortalities of An. stephensi (< 24%) at all treatments tested in the bioassays and were not effective against adults of malaria vector An. stephensi (Table 6).\n\nTable 6Mean % adult mortality of Momordica foetida crude leaf solvent extract at different concentrations against Anopheles stephensiConcentration(ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol \nHexane 200.00 ± 0.00ABa0.00 ± 0.00Aa0.00 ± 0.00Aa401.67 ± 1.67Ba0.00 ± 0.00Aa0.00 ± 0.00Ab805.00 ± 2.89Ba1.67 ± 1.67Ba0.00 ± 0.00Ab16013.33 ± 1.67Ca13.33 ± 1.67Ca0.00 ± 0.00Ab32018.33 ± 4.41Ca23.33 ± 1.67Da1.67 ± 1.67BbControl0.00 ± 0.00Aa0.00 ± 0.00Aa0.00 ± 0.00Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % adult mortality of Momordica foetida crude leaf solvent extract at different concentrations against Anopheles stephensi\n*Each value of % mean mortality ± SE represents the value of three replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nThere were statistically significant differences in mean percentage mortality among different concentrations of aqueous, methanol, and hexane leaf extracts of Z. scabra and with negative control (Kruskal-Wallis test; p-value < 0.05; Table 7). Within the given time intervals adult mortality was recorded for the control treatment. However, the highest adult mortality (84%) at higher concentration 320 ppm against An. stephensi was recorded in aqueous crude leaf extract of Z. scabra followed by its methanol extracts (65%) and their adulticidal potential was statistically different from the negative control (Multiple-Mann Whitney U-test; p-value < 0.003, Table 7).Table 7Mean % adult mortality of Zehneria scabra crude leaf solvent extracts at different concentrations against Anopheles stephensiConcentration (ppm)% Mean mortality ± SE\nSolvents \nAqueous \nMethanol \nHexane 204.33 ± 2.6Aa11.67 ± 3.33Aa0.00 ± 0.00Aa4011.67 ± 1.67Ba20.00 ± 2.89ABa0.00 ± 0.00Ab8026.67 ± 3.33Ca23.33 ± 3.33Ba0.00 ± 0.00Ab16036.67 ± 6.00Ca41.66 ± 4.40Ca15.00 ± 2.89Bb32084.00 ± 2.89Da65.00 ± 5.77Db20.00 ± 2.89BcControl0.67 ± 0.45Ea0.67 ± 0.45Ea0.67 ± 0.45Aa*Each value of % mean mortality ± SE represents the value of three replicates*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nMean % adult mortality of Zehneria scabra crude leaf solvent extracts at different concentrations against Anopheles stephensi\n*Each value of % mean mortality ± SE represents the value of three replicates\n*Means values followed by the same letters across column (Upper case) and row (Lower case) are not statistically significantly different (Multiple-Mann Whitney U-test; p-value > 0.003)\nThe lethal effects of different crude leaf extracts against adults of An. stephensi are shown in Table 8. All hexane and M. foetida crude leaf extracts had low adulticidal impacts, only aqueous and methanol C. aurea and Z. scabra leaf extracts were subjected to dose-response bioassay to detect the lethal concentrations. From the dose-response curves, the lowest LC50 (= 176.20 ppm and 205.41 ppm) and LC90 (= 425.13 and 883.11 ppm) values were demonstrated from the aqueous and methanol Z. scabra crude leaf extracts, respectively (Table 8), and its relative toxicities against adult An. stephensi was much higher than the rest of the plant extracts. The highest LC50 (= 297.75 ppm and 333.27 ppm) and LC90 (= 762.63 ppm and 1031.74 ppm) values were from the aqueous and methanol C. aurea crude leaf extracts, respectively, and its relative toxicity on An. stephensi was much lower than Z. scabra crude extracts.\n\nTable 8LC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of Calpurnia aurea and Zehneria scabra against adults of Anopheles stephensiTest plantsSolventsLC5095%Confidence limitLC9095%Confidence limitχ2 (df = 3)\nLCL \nUCL \nLCL \nUCL \nC. aureaAqueous297. 75254. 95340. 56762. 63491. 131034. 130.88Methanol333. 27268. 92397. 631031. 74569. 061494. 430.75\nZ. scabraAqueous176.20157. 82194. 58425. 13346. 14504. 122.70Methanol205. 41172. 29238. 54883. 11529. 331236. 900.70\nLC50 and LC90 (ppm) of aqueous and methanolic crude leaf extracts of Calpurnia aurea and Zehneria scabra against adults of Anopheles stephensi", "Synthetic insecticides play a vital role in insect vector control programmes. However, due to environmental concerns and the development of resistance to synthetic insecticides, there is a growing effort to explore plants to obtain bioactive compounds that are safe for non-target animals and do not pose residue problems, but are still able to suppress vector populations. Here, the present study evaluated the larvicidal and adulticidal activities of crude leaf solvent extracts of C. aurea, M. foetida, and Z. scabra, native plants to Ethiopia, which were tested against An. stephensi, an invasive malaria vector in Ethiopia. In this study, all hexane crude leaf extracts showed ineffectual insecticidal activity on An. stephensi larvae and adults. However, aqueous and methanol extracts of the test plants gave promising results to be used as mosquitocides. This could suggest the presence of more polar solvent-soluble phytochemicals in leaves of C. aurea, M. foetida, and Z. scabra, which could be responsible for the observed bioactivities against An. stephensi. These findings are in agreement with that of Chore et al. [41], who suggested that solvent polarities used for extraction could influence the mortality of mosquito vectors.\nCrude aqueous leaf extracts of the plants, M. foetida and Z. scabra at 100 ppm possessed good larvicidal properties relative to the positive control (Temephos). Similar trends were also observed in the late third to early fourth instar larvae of An. stephensi with aqueous leaf extract of C. aurea at 150 ppm. Equally, methanol extracts of C. aurea at 150 ppm and M. foetida and Z. scabra at 200 ppm caused more than 95% larval mortality. Those concentrations of the extracts which showed potent effect were high. This requires using a large amount of the plants if such extracts have to be applied in vector control operations. Therefore, this limitation of the study has to be considered in future research on botanical insecticides.\nGhosh et al. [26] reported that insecticidal effects of plant extracts can vary due to the differences in plant species, mosquito species, geographical variation, extraction methodology, and polarity of solvents used during extraction. Similarly, the finding of the current study varies among concentrations and extraction solvents. Therefore, in agreement with this study, all aqueous crude leaf extracts proved to be sufficiently effective on larvae of An. stephensi. Interestingly, the aqueous crude leaf extract of M. foetida was found to have potent larvicidal activity with low LC50 and LC90 values of 34.61 and 57.61 ppm, respectively, compared to the other two plant extracts assayed concurrently. In other studies, aqueous extracts of M. foetida have been shown to have larvicidal activity against An. gambiae and An. coluzzii larvae with LC50 values of 593.96 and 505.19 ppm, respectively [42]. This high difference in larvicidal activities in the current finding could be due to differences in the mosquito species, extraction methods, locality of the plant, preparation of test concentration, and parts of the plant, which are different from those used in this study.\nTo our knowledge, previous reports are not available on the insecticidal activity of Z. scabra crude extracts against mosquito vectors elsewhere. This study demonstrated the effective larvicidal activity from the Z. scabra aqueous (LC50 = 35.85 ppm; LC90 = 68.26 ppm), and methanol (LC50 = 41.32 ppm; LC90 = 99.06 ppm) crude leaf extracts against An. stephensi larvae, where larvicidal activities also showed higher mortality better results than C. aurea extracts. Previous studies also demonstrated that extracts of Z. scabra were found to be effective against stored product insect pest [34]. These indicate that the plant has bioactive secondary metabolites that can be used to control insects. The presence of secondary metabolites claimed to have insecticidal activities such as phenol, tannins, flavonoids, and glycosides have been identified in methanol extract of Z. scabra leaves in previous studies [43, 44]. Such promising larvicidal potency from this plant against An. stephensi larvae may be associated with the presence of these active secondary metabolites.\nIn this study, crude aqueous and methanol extracts of C. aurea leaf were also shown to have high larvicidal activity against An. stephensi larvae with LC50 and LC90 values of (38.69 ppm, 108.28 ppm), and (43.25 ppm, 96.02 ppm), respectively. This result is comparable with the methanol extract of C. aurea leaf extract, which has shown high larval mortality against An. arabiensis with LC50 and LC90 values of 84.85 and 192.29 ppm, respectively, in previous studies [45].\nMethanol extract of M. foetida leaf showed relatively lower larvicidal efficacy than other test extracts against An. stephensi larvae with LC50 = 99.50 and LC90 = 188.76 ppm, in the present study. However, this result is preferable to methanol M. foetida extract against the larvae of An. gambiae and An. coluzzii with LC50 values of 276.32 and 235.31 ppm, respectively [42]. A previous study demonstrated that M. foetida is known for its insecticidal and repellent activities against mosquitoes [33]. Crude aqueous extract of M. foetida leaf was shown to have high phenolic and flavonoid secondary metabolites [46], which might have contributed to its larvicidal activity.\nThe values of LC50 and LC90 obtained by the larvae in this study are lower than those reported by Dey et al. [47], who found that the maximum larvicidal activity from the Piper longum aqueous (LC50 = 133.42 ppm; LC90 = 279.61 ppm) and methanol (LC50 = 134.71 ppm; LC90 = 464.73 ppm) crude leaf extracts against An. stephensi. In another study, Mohankumar et al. [48] showed insecticidal promising from the methanol extract of Annona reticulate leaf against An. stephensi larvae with LC50 and LC90 values of 262.71 and 636.94 ppm, respectively. Therefore, this study suggests that the three plant species could have a beneficial role in the control of mosquitoes.\nPlant extracts can be used either as insecticides for killing larvae or adult mosquitoes or as repellents for protection against mosquito bites, depending on their activity [49]. In this study, the effects of test plant extract on adults of An. stephensi are remarkably less than those recorded on larvae. The adulticidal activity of M. foetida crude leaf extracts showed less effectiveness against An. stephensi and its effects were not significantly different from the control treatment. This low activity might be due to the solvents used to extract the bioactive metabolites, which have lower efficacy against the adults.\nIn the present study, low LC50 values against the adults of An. stephensi was found from the aqueous (LC50 = 176.20 ppm) and methanol (LC50 = 205.41 ppm) crude leaf extracts of Z. scabra compared to other test plant extracts. Previous studies revealed extracts of Z. scabra have effective insecticidal activities against aphids, glowworms, and mill bugs [50]. It also has been reported that Z. scabra crude extracts to have anti-plasmodial effects [51].\nIn contrast, aqueous and methanol extracts of C. aurea showed less mortality against adults of An. stephensi with LC50 values of 297.75 and 333.28 ppm, respectively. Hiruy and Getu [52] reported potent adulticidal activity from the aqueous, ethanol, and acetone extracts of C. aurea against adults of maize weevils, agriculturally important insect species. Although the WHO does not specify any criteria for classifying the larvicidal and adulticidal potentialities of plant products, some authors use LC50 values as a criterion for activity level determination. Specifically, as noted in other studies, the product is very active if LC50 < 50 ppm, the product is active if LC50 < 100 ppm, the product is moderate if LC50 is between 100 ppm-200 ppm, and the product is weakly effective if LC50 is between 200 ppm-750 ppm, while the product is inactive if LC50 > 750 ppm [53, 54]. Accordingly, the adulticidal activity of C. aurea extract could be categorized as weakly effective against An. stephensi.", "This study showed that crude aqueous and methanol extracts of C. aurea, M. foetida, and Z. scabra leaves could be considered as foreseeable products to be developed as potential larvicides against An. stephensi larvae. In addition, crude leaf extracts of C. aurea and Z. scabra are potential candidate insecticides against the adults. These three plants could be used to develop effective, safe, biodegradable, and cheap botanical insecticides for vector control, potentially leading to improved resistance management targeted against malaria vectors in Ethiopia and elsewhere. Therefore, further chemical analysis studies on the identification, preparation, and formulation of bioactive compounds from plants are essential." ]
[ null, null, null, null, null, null, null, null, null, null, "results", null, null, "discussion", "conclusion" ]
[ "Adulticide", "\nAnopheles stephensi\n", "Botanical", "Larvicide", "Plant extract" ]
Stories of paediatric palliative care: a qualitative study exploring health care professionals' understanding of the concept.
36273144
By sharing patient stories, health care professionals (HCPs) may communicate their attitudes, values and beliefs about caring and treatment. Previous qualitative research has shown that HCPs usually associate paediatric palliative care (PPC) with death or dying and that they find the concept challenging to understand and difficult to implement. Attending to HCPs' stories may provide a richer account of their understanding of PPC. Thus, the aim of this study was to explore PPC stories narrated by HCPs to gain increased insight into their understanding of what PPC entails.
BACKGROUND
This qualitative study collected data from four focus group interviews with 21 HCPs from different units in two Norwegian hospitals. Stories told by the HCPs to illustrate their comprehension of PPC were analysed following thematic analysis procedures.
METHODS
Four themes were identified illustrating what PPC meant to the participants: creating spaces for normality, providing tailored support for the family, careful preparations for saying goodbye and experiencing dilemmas and distress. The stories centred on family care, particularly relating to dramatic or affective situations when the death of a child was imminent.
RESULTS
The stories reflect how the HCPs view PPC as a specific field of health care that requires particular professional sensitivity, including good communication, collaboration and planning. Thus, the HCPs in this study demonstrated knowledge about the core qualities needed to succeed in PPC. However, similar to previous research, the stories illustrate that how HCPs speak about PPC is strongly associated with end-of-life care, and by that the HCPs do not capture the breadth of the PPC concept. The findings highlight the importance of increasing knowledge about the meaning and content of PPC among HCPs in order to maintain quality of life for all children with life-limiting or life-threatening conditions throughout their illness trajectory.
CONCLUSION
[ "Child", "Humans", "Palliative Care", "Quality of Life", "Qualitative Research", "Health Personnel", "Terminal Care" ]
9587603
null
null
null
null
Results
Following the analysis, four themes were identified illustrating what PPC entails to the participants: creating spaces for normality, providing tailored support for the family, careful preparations for saying goodbye and experiencing dilemmas and distress (Table 2). Table 2Themes and corresponding codes and examples of quotes from the storiesThemesCodesQuotesCreating spaces for normalityEveryday life and playSupporting normality Like that one time, we hid under the blanket because she did not want to talk about her disease. When the doctor tried, the girl hid under the blanket, and I asked to join her. So we stayed there together. We kind of had an alliance. It is about seeing them exactly where they are. Providing tailored support for the familyProviding knowledge and supportProviding extraordinary careLayer upon layerCreating safety The parents felt safe when coming here, and they felt safe to leave the room. They could breathe a little while they knew she was taken care of. Careful preparations for saying goodbyeMaking arrangements for saying goodbyeBeing togetherCapturing the right moments for difficult conversationsRealizing that death is imminent And we sat there at the outpatient clinic and I thought to myself that I have to tell them…because I do not know how long he would live, so we should talk about that. And at that moment I was given an opportunity to say that if he came back with an infection we might not be able to do…to save him…or he could end up on a ventilator and that is not necessarily a good idea because we cannot get them off. So we talked about that. Experiencing dilemmas and distressConflicting wishesDilemmas relating to treatment It was obvious to everyone else that the child’s condition was very severe and that she would never recover, and then the parent says “well, that is just what you believe!” (…) If the parents want treatment, when we have no treatment to offer, there is always someone that makes promises without scientific foundation. Themes and corresponding codes and examples of quotes from the stories Everyday life and play Supporting normality Providing knowledge and support Providing extraordinary care Layer upon layer Creating safety Making arrangements for saying goodbye Being together Capturing the right moments for difficult conversations Realizing that death is imminent Conflicting wishes Dilemmas relating to treatment [SUBTITLE] Creating spaces for normality [SUBSECTION] The HCPs seemed attentive to having the child with a life-threatening or life-limiting illness at the centre of their care. Several participants spoke about the importance of supporting a sense of normality and mentioned that the parents as well as the child aimed for ordinary days at home, school or kindergarten: “I believe that most parents want their child’s days to be normal and good until the end and that it’s not going to be something else”. (Group 3) Stories were told that illustrate how the HCPs managed to create spaces for play and fun for the child despite working in a hospital and being responsible for clinical procedures. They gave examples of how they used medical equipment as props for play and how these playful situations opened up conversations with the child about sensitive and important topics. In one story, the nurse explained that they made the hospital stay so engaging that the child looked forward to treatment and felt sorry for leaving. Another nurse elaborated on a playful moment with a little girl with a life-limiting illness engaging in imaginary play:…and then the girl says: “At my prom, I want a dress like that”, and I said “Really? Wow, that’s cool”, and then she kind of realizes that she’s never going to a prom and then she says: “Well, well…it would have been cool, right?” and I responded “Yeah, it would have been really cool” (Group 2). …and then the girl says: “At my prom, I want a dress like that”, and I said “Really? Wow, that’s cool”, and then she kind of realizes that she’s never going to a prom and then she says: “Well, well…it would have been cool, right?” and I responded “Yeah, it would have been really cool” (Group 2). The nurse reflected on the importance of being present in the moment and allowing the child to enjoy the thought of adolescence even though her life expectancy was very limited. The HCPs seemed attentive to having the child with a life-threatening or life-limiting illness at the centre of their care. Several participants spoke about the importance of supporting a sense of normality and mentioned that the parents as well as the child aimed for ordinary days at home, school or kindergarten: “I believe that most parents want their child’s days to be normal and good until the end and that it’s not going to be something else”. (Group 3) Stories were told that illustrate how the HCPs managed to create spaces for play and fun for the child despite working in a hospital and being responsible for clinical procedures. They gave examples of how they used medical equipment as props for play and how these playful situations opened up conversations with the child about sensitive and important topics. In one story, the nurse explained that they made the hospital stay so engaging that the child looked forward to treatment and felt sorry for leaving. Another nurse elaborated on a playful moment with a little girl with a life-limiting illness engaging in imaginary play:…and then the girl says: “At my prom, I want a dress like that”, and I said “Really? Wow, that’s cool”, and then she kind of realizes that she’s never going to a prom and then she says: “Well, well…it would have been cool, right?” and I responded “Yeah, it would have been really cool” (Group 2). …and then the girl says: “At my prom, I want a dress like that”, and I said “Really? Wow, that’s cool”, and then she kind of realizes that she’s never going to a prom and then she says: “Well, well…it would have been cool, right?” and I responded “Yeah, it would have been really cool” (Group 2). The nurse reflected on the importance of being present in the moment and allowing the child to enjoy the thought of adolescence even though her life expectancy was very limited. [SUBTITLE] Providing tailored support for the family [SUBSECTION] The participants spoke about different ways of providing support through adjusted information and tailored care. In their stories, they emphasized the importance of sufficient, timely information. One participant described how a mother once said that she did not understand that her child could die while being on a ventilator and that the lungs could still collapse. The participant had carried this mother’s utterance with her ever since, and she pointed out that making sure that parents understand why their child actually dies is significant. Another participant described how important it is to be able to recognize when the parents communicate a need for more knowledge and to provide accurate information when it is called for. However, the participants also referred to challenging situations in which the parents and HCPs were not in step with each other. One participant admitted that if HCPs believe the parents are informed about the child’s situation and suddenly realize that they are not, this may cause delays in procedures and irritation:I try to be observant to this, both in myself and others. It’s important to avoid a moralizing attitude: ‘Do they [the parents] still not understand?’ Because it creates hassle and annoyance when the parents are not sufficiently informed and up to date on the child’s situation. (Group 2) I try to be observant to this, both in myself and others. It’s important to avoid a moralizing attitude: ‘Do they [the parents] still not understand?’ Because it creates hassle and annoyance when the parents are not sufficiently informed and up to date on the child’s situation. (Group 2) The participants gave several examples of how they adjust and tailor their care for families to make them feel safe in acute as well as more stable situations. They spoke about the importance of working as a team around the family. One participant described it as creating layers upon layers of love around the child. This expression derived from a particular story about a child who was going to be placed in foster care but was too sick to leave the hospital. With help from the HCP, the child’s birth mother and foster mother moved into the children’s ward. With support from the multidisciplinary team, they cared for the child together during the last weeks of the child’s life.…and the foster mother stepped aside to let the birth mother hold the child close, day and night (…) and the baby received so many layers of love (…) And the HCPs were amazingly professional and put things aside to let it happen. (Group 2) …and the foster mother stepped aside to let the birth mother hold the child close, day and night (…) and the baby received so many layers of love (…) And the HCPs were amazingly professional and put things aside to let it happen. (Group 2) Later, the birth mother explained that, despite the great tragedy, these had been the finest weeks of her life. This situation was resource-demanding for the unit, but all the HCPs stood out as highly professional and were willing to make an extra effort in the extraordinary situation. Going that extra mile for children and their families was a recurring theme in the HCPs’ stories. Witnessing severe and distressing situations seemed to make the HCPs mobilize to make the days, weeks and months as good as possible for the child, their parents and their siblings. Many of the participants’ stories were about creating safe spaces for families at the hospital, giving the child, parents and siblings moments to breathe. There were also examples of systems failing to support the families adequately and lack of coordination resulting in disorganized care; however, most stories were examples of how HCPs managed to work together, emphasizing the importance of shared information. One participant said:One of the finest moments that I’ve witnessed was when a nurse who knew the family well took the initiative to play with an older sibling of a child who would soon die. The nurse took the sibling outside in the hallway, and they talked and played it out. They had a good relationship. The nurse knew very well what was about to happen because she had been in the room when the message about the child’s impending death was given. The more people working around the family, the more important it is that we have the same information and work as a team. (Group 4) One of the finest moments that I’ve witnessed was when a nurse who knew the family well took the initiative to play with an older sibling of a child who would soon die. The nurse took the sibling outside in the hallway, and they talked and played it out. They had a good relationship. The nurse knew very well what was about to happen because she had been in the room when the message about the child’s impending death was given. The more people working around the family, the more important it is that we have the same information and work as a team. (Group 4) The participants spoke about different ways of providing support through adjusted information and tailored care. In their stories, they emphasized the importance of sufficient, timely information. One participant described how a mother once said that she did not understand that her child could die while being on a ventilator and that the lungs could still collapse. The participant had carried this mother’s utterance with her ever since, and she pointed out that making sure that parents understand why their child actually dies is significant. Another participant described how important it is to be able to recognize when the parents communicate a need for more knowledge and to provide accurate information when it is called for. However, the participants also referred to challenging situations in which the parents and HCPs were not in step with each other. One participant admitted that if HCPs believe the parents are informed about the child’s situation and suddenly realize that they are not, this may cause delays in procedures and irritation:I try to be observant to this, both in myself and others. It’s important to avoid a moralizing attitude: ‘Do they [the parents] still not understand?’ Because it creates hassle and annoyance when the parents are not sufficiently informed and up to date on the child’s situation. (Group 2) I try to be observant to this, both in myself and others. It’s important to avoid a moralizing attitude: ‘Do they [the parents] still not understand?’ Because it creates hassle and annoyance when the parents are not sufficiently informed and up to date on the child’s situation. (Group 2) The participants gave several examples of how they adjust and tailor their care for families to make them feel safe in acute as well as more stable situations. They spoke about the importance of working as a team around the family. One participant described it as creating layers upon layers of love around the child. This expression derived from a particular story about a child who was going to be placed in foster care but was too sick to leave the hospital. With help from the HCP, the child’s birth mother and foster mother moved into the children’s ward. With support from the multidisciplinary team, they cared for the child together during the last weeks of the child’s life.…and the foster mother stepped aside to let the birth mother hold the child close, day and night (…) and the baby received so many layers of love (…) And the HCPs were amazingly professional and put things aside to let it happen. (Group 2) …and the foster mother stepped aside to let the birth mother hold the child close, day and night (…) and the baby received so many layers of love (…) And the HCPs were amazingly professional and put things aside to let it happen. (Group 2) Later, the birth mother explained that, despite the great tragedy, these had been the finest weeks of her life. This situation was resource-demanding for the unit, but all the HCPs stood out as highly professional and were willing to make an extra effort in the extraordinary situation. Going that extra mile for children and their families was a recurring theme in the HCPs’ stories. Witnessing severe and distressing situations seemed to make the HCPs mobilize to make the days, weeks and months as good as possible for the child, their parents and their siblings. Many of the participants’ stories were about creating safe spaces for families at the hospital, giving the child, parents and siblings moments to breathe. There were also examples of systems failing to support the families adequately and lack of coordination resulting in disorganized care; however, most stories were examples of how HCPs managed to work together, emphasizing the importance of shared information. One participant said:One of the finest moments that I’ve witnessed was when a nurse who knew the family well took the initiative to play with an older sibling of a child who would soon die. The nurse took the sibling outside in the hallway, and they talked and played it out. They had a good relationship. The nurse knew very well what was about to happen because she had been in the room when the message about the child’s impending death was given. The more people working around the family, the more important it is that we have the same information and work as a team. (Group 4) One of the finest moments that I’ve witnessed was when a nurse who knew the family well took the initiative to play with an older sibling of a child who would soon die. The nurse took the sibling outside in the hallway, and they talked and played it out. They had a good relationship. The nurse knew very well what was about to happen because she had been in the room when the message about the child’s impending death was given. The more people working around the family, the more important it is that we have the same information and work as a team. (Group 4) [SUBTITLE] Careful preparations for saying goodbye [SUBSECTION] Most of the stories emerged from critical situations or when the child’s illness was worsening, especially about initiating advance care planning with the end of the child’s life in mind. Some described the importance of seizing that particular moment when the parents and the child are ready and receptive to these difficult conversations. The HCPs gave examples of when the parents themselves took the initiative, but in most of the stories, the HCPs took the lead. However, as underlined by one participant, engaging in such dialogue requires that the HCPs are present and that they tread carefully: “If you’ve established a good relationship, moments will occur if you’re observant and willing to enter these conversations. For me, this is part of palliative care”. (Group 2) With the children, such conversations could take place during play, and difficult topics could be touched upon without needing to make eye contact. It became evident through the HCPs’ stories that these conversations were tough and that it was sometimes difficult to control their own feelings.These conversations are really hard, it is as if you are about to start crying yourself. But I think it is really, really important [to talk about it], because we have reached the moment when it might happen. (Group 3) These conversations are really hard, it is as if you are about to start crying yourself. But I think it is really, really important [to talk about it], because we have reached the moment when it might happen. (Group 3) Through their stories, the HCPs emphasized the need for advance care plans when the risk of severe incidents increased. The advantages of such plans are to secure continuity and ensure that all important information is available for all the HCPs involved in a possible emergency situation. However, these plans should also be made for the family members so that they know how to act if critical events occur. In several stories, the HCPs described how the care plans included strategies and measures for staying at home as much as possible. One example was given of how specialist care and primary care cooperated, where the child was provided with advanced home care, and the child and the family shifted between being at the hospital and being at home. The family in this story expressed a strong wish to let the child die at home, and the HCPs made great efforts to accommodate this wish. The participant described how all the necessary equipment was installed in the family’s house:It was arranged for him to stay at home. He received pain relief, and I believe the community nurses cared for him, and he spent his last 2–3 weeks in the living room together with his siblings and parents and they all slept together in the same bed. They were kind of… they were together.” (Group 1) It was arranged for him to stay at home. He received pain relief, and I believe the community nurses cared for him, and he spent his last 2–3 weeks in the living room together with his siblings and parents and they all slept together in the same bed. They were kind of… they were together.” (Group 1) Making practical arrangements to facilitate a proper farewell in a safe environment together was a prioritized task. The parents and siblings could focus on being there for the child and for each other while the medical team provided care and pain relief for the child. Most of the stories emerged from critical situations or when the child’s illness was worsening, especially about initiating advance care planning with the end of the child’s life in mind. Some described the importance of seizing that particular moment when the parents and the child are ready and receptive to these difficult conversations. The HCPs gave examples of when the parents themselves took the initiative, but in most of the stories, the HCPs took the lead. However, as underlined by one participant, engaging in such dialogue requires that the HCPs are present and that they tread carefully: “If you’ve established a good relationship, moments will occur if you’re observant and willing to enter these conversations. For me, this is part of palliative care”. (Group 2) With the children, such conversations could take place during play, and difficult topics could be touched upon without needing to make eye contact. It became evident through the HCPs’ stories that these conversations were tough and that it was sometimes difficult to control their own feelings.These conversations are really hard, it is as if you are about to start crying yourself. But I think it is really, really important [to talk about it], because we have reached the moment when it might happen. (Group 3) These conversations are really hard, it is as if you are about to start crying yourself. But I think it is really, really important [to talk about it], because we have reached the moment when it might happen. (Group 3) Through their stories, the HCPs emphasized the need for advance care plans when the risk of severe incidents increased. The advantages of such plans are to secure continuity and ensure that all important information is available for all the HCPs involved in a possible emergency situation. However, these plans should also be made for the family members so that they know how to act if critical events occur. In several stories, the HCPs described how the care plans included strategies and measures for staying at home as much as possible. One example was given of how specialist care and primary care cooperated, where the child was provided with advanced home care, and the child and the family shifted between being at the hospital and being at home. The family in this story expressed a strong wish to let the child die at home, and the HCPs made great efforts to accommodate this wish. The participant described how all the necessary equipment was installed in the family’s house:It was arranged for him to stay at home. He received pain relief, and I believe the community nurses cared for him, and he spent his last 2–3 weeks in the living room together with his siblings and parents and they all slept together in the same bed. They were kind of… they were together.” (Group 1) It was arranged for him to stay at home. He received pain relief, and I believe the community nurses cared for him, and he spent his last 2–3 weeks in the living room together with his siblings and parents and they all slept together in the same bed. They were kind of… they were together.” (Group 1) Making practical arrangements to facilitate a proper farewell in a safe environment together was a prioritized task. The parents and siblings could focus on being there for the child and for each other while the medical team provided care and pain relief for the child. [SUBTITLE] Experiencing dilemmas and distress [SUBSECTION] While many stories and examples evolved from cases where the HCPs explained that they managed to provide support and good care, there were also stories about conflicts with parents and distressful experiences. Some HCPs referred to conversations about how to handle a dramatic worsening of the child’s condition (e.g. decisions about not putting the child on a ventilator or stopping medications) and how this had provoked anger and frustration among parents. One HCP described one such situation:... and then there were discussions about withdrawing antibiotics, and we had to tell the parents, and one of them was really devastated and said, ‘You’re giving up on my child!’ whereupon the doctor kindly responded, ‘No, we’re not, we are comforting her during her last days’”. (Group 2) ... and then there were discussions about withdrawing antibiotics, and we had to tell the parents, and one of them was really devastated and said, ‘You’re giving up on my child!’ whereupon the doctor kindly responded, ‘No, we’re not, we are comforting her during her last days’”. (Group 2) In some cases, the participants noted that it was the HCPs and not the parents who pushed too hard by suggesting a new treatment, which turned out to result in complications or negative side effects. Another story illustrated the tension that can occur between HCPs and parents when the child is expected to die within a short time. In this case, one HCP accused a parent of hastening the baby’s death by withholding nutrition. This resulted in a deep conflict among the staff, where some supported the nurse and some sympathized with the parents. Eventually, external counselling was required to resolve the dispute among the staff. After the baby died, the parents reported that the whole situation had added even more to their grief. The participant described this as an ethically demanding situation, and, in retrospect, she was struck by how good intentions can lead to conflict and how emotionally involved one may become. Examples were also given of parents who refused to acknowledge that their child had a life-limiting disease and instead of planning for the end spent all their time searching for a cure. The HCPs experienced it as particularly hard to witness how false hope was being sold through experimental treatment and that parents were willing to squander that precious, limited time left of their child’s life:There was a family that I was never in a position to speak with, because their attention was always directed at something else. It was impossible to talk to them about anything, because they were always focusing on going off somewhere to find a cure. (Group 2) There was a family that I was never in a position to speak with, because their attention was always directed at something else. It was impossible to talk to them about anything, because they were always focusing on going off somewhere to find a cure. (Group 2) In these stories, the parents searched for opportunities to travel far with their child to try experimental treatment, resulting in long periods away from home and separation between family members. The participant found it challenging that the parents were never available to make plans for good palliative care. While many stories and examples evolved from cases where the HCPs explained that they managed to provide support and good care, there were also stories about conflicts with parents and distressful experiences. Some HCPs referred to conversations about how to handle a dramatic worsening of the child’s condition (e.g. decisions about not putting the child on a ventilator or stopping medications) and how this had provoked anger and frustration among parents. One HCP described one such situation:... and then there were discussions about withdrawing antibiotics, and we had to tell the parents, and one of them was really devastated and said, ‘You’re giving up on my child!’ whereupon the doctor kindly responded, ‘No, we’re not, we are comforting her during her last days’”. (Group 2) ... and then there were discussions about withdrawing antibiotics, and we had to tell the parents, and one of them was really devastated and said, ‘You’re giving up on my child!’ whereupon the doctor kindly responded, ‘No, we’re not, we are comforting her during her last days’”. (Group 2) In some cases, the participants noted that it was the HCPs and not the parents who pushed too hard by suggesting a new treatment, which turned out to result in complications or negative side effects. Another story illustrated the tension that can occur between HCPs and parents when the child is expected to die within a short time. In this case, one HCP accused a parent of hastening the baby’s death by withholding nutrition. This resulted in a deep conflict among the staff, where some supported the nurse and some sympathized with the parents. Eventually, external counselling was required to resolve the dispute among the staff. After the baby died, the parents reported that the whole situation had added even more to their grief. The participant described this as an ethically demanding situation, and, in retrospect, she was struck by how good intentions can lead to conflict and how emotionally involved one may become. Examples were also given of parents who refused to acknowledge that their child had a life-limiting disease and instead of planning for the end spent all their time searching for a cure. The HCPs experienced it as particularly hard to witness how false hope was being sold through experimental treatment and that parents were willing to squander that precious, limited time left of their child’s life:There was a family that I was never in a position to speak with, because their attention was always directed at something else. It was impossible to talk to them about anything, because they were always focusing on going off somewhere to find a cure. (Group 2) There was a family that I was never in a position to speak with, because their attention was always directed at something else. It was impossible to talk to them about anything, because they were always focusing on going off somewhere to find a cure. (Group 2) In these stories, the parents searched for opportunities to travel far with their child to try experimental treatment, resulting in long periods away from home and separation between family members. The participant found it challenging that the parents were never available to make plans for good palliative care.
Conclusion
In this study, we aimed to explore stories of PPC as narrated by HCPs to enhance our knowledge of their understanding of what PPC is. In our previous paper, we discussed that although the HCPs were hesitant to use the term PPC, they found themselves responsible for delivering PPC [11]. The HCPs’ stories supported this by highlighting key elements of PPC, such as interdisciplinary teamwork family-centred care and advance care planning. However, similar to findings in our first paper [11] and previous research [8–10], the stories illustrated that HCPs’ reflections about PPC did not capture the entire concept of PPC, as they were strongly associated with end-of-life care. This underscores the importance of increasing knowledge about PPC among HCPs in Norway. A consolidated recognition of the meaning and content of PPC is necessary to maintain quality of life for all children living with life-limiting or life-threatening conditions.
[ "Background", "Methods", "Design", "Participants", "Data collection", "Data analysis", "Ethical considerations", "Creating spaces for normality", "Providing tailored support for the family", "Careful preparations for saying goodbye", "Experiencing dilemmas and distress", "Strengths and limitations" ]
[ "Storytelling is a fundamental part of being human. Through stories information can be shared in a way that creates an emotional connection and by that provides a deeper understanding of the storyteller’s experiences. Thus, storytelling has the potential to bring new perspectives to the Table [1]. When telling stories about specific events from patients´ lives, health care professionals (HCPs) not only describe the patients’ medical situations. By sharing patient stories, HCPs also communicate their attitudes, values and beliefs about caring and treatment [2]. The present study used storytelling as a methodological approach to investigate HCPs’ understanding of paediatric palliative care (PPC). According to the World Health Organization (WHO), PPC should be the standard of care for children with life-threatening or life-limiting conditions and should be integrated with prevention, early diagnosis and treatment [3, 4]. PPC is defined as “the active total care of the child’s body, mind and spirit, and [this] also involves giving support to the family” [5]. Children (0–18 years) and their families should be provided with interdisciplinary care aiming to promote their physical, psychological, social and spiritual well-being [4]. Early implementation of PPC through active and integrated support is endorsed as beneficial to the well-being of the child and the family [3, 6, 7]. HCPs may display good knowledge of the principles of palliative care, but their comprehension of the concept, their way of thinking and their reported reasons for referrals to PPC demonstrate that they associate PPC with death and dying [8, 9]. Research reports that HCPs find the concept challenging to understand and implement and that it evokes negative emotions [10–12]. Consequently, early integration of PPC may be delayed and many children are not referred to PPC until late in their illness trajectory [12]. Among the barriers reported for implementing PPC at an early stage are uncertainty and confusion about the concept, as well as stigma related to the term [10, 13]. In our recent study of interdisciplinary HCPs’ understanding and implementation of PPC, we found that the participants viewed the concept as unfamiliar and not meaningful [11]. HCPs associated PPC with death and dying and used words like scary, burdensome, dramatic and lack of active treatment to describe their comprehension of the concept [11].\nAttending to stories of HCPs´ clinical experience with children and their families may provide further insights than solely through answering questions and thus be a valuable approach to gain an even richer account of their understanding of PPC. Thus, the aim of this study was to explore stories of PPC narrated by HCPs to enhance our knowledge of their understanding of what PPC entails.", "[SUBTITLE] Design [SUBSECTION] This study included qualitative data obtained through focus group interviews with HCPs and emerged from a project exploring HCPs’ understanding of PPC [11]. The project was not originally designed to elicit narratives; however, during the focus groups, the HCPs told stories or gave specific examples to illustrate their comprehension of PPC. We decided to subtract these stories and all the utterances made from the initial material to provide examples or illustrate points when reflecting on what PPC is. Thus, the data was not a part of the material analysed in the first article. We explored the stories in more depth, with the assumption that this may provide an enhanced understanding of what HCPs comprehend when they talk about PPC. The reporting of the study was guided by the consolidated criteria for reporting qualitative research [14].\nThis study included qualitative data obtained through focus group interviews with HCPs and emerged from a project exploring HCPs’ understanding of PPC [11]. The project was not originally designed to elicit narratives; however, during the focus groups, the HCPs told stories or gave specific examples to illustrate their comprehension of PPC. We decided to subtract these stories and all the utterances made from the initial material to provide examples or illustrate points when reflecting on what PPC is. Thus, the data was not a part of the material analysed in the first article. We explored the stories in more depth, with the assumption that this may provide an enhanced understanding of what HCPs comprehend when they talk about PPC. The reporting of the study was guided by the consolidated criteria for reporting qualitative research [14].\n[SUBTITLE] Participants [SUBSECTION] HCPs from three paediatric units in two hospitals were recruited using purposeful sampling, including a variety of professions. Both hospitals are located in Eastern Norway. The units include different departments for children aged 0–18 with a wide range of diagnoses. A total of 21 participants, of whom 19 (90%) were female, participated in 4 focus groups including 2–9 participants (Table 1). Most of the participants had worked with children with life-threatening and life-limiting conditions for more than 10 years. Only two had fewer than two years of experience and some had more than 20 years. Without giving any reason, four HCPs from one of the units withdrew their consent, which is why one of the groups ended up including only two participants. The other three groups included four, six and nine participants. We have no information about the HCPs who declined invitations to participate.\n\nTable 1ParticipantsFocus group numberNumber of participantsProfessions14Paediatric nurses (2)Nurses (2)29Clinical social workerOncology nursePaediatric nurses (2)NurseChief PhysicianSpecialist in psychologyPaediatricianPhysiotherapist32PhysiotherapistPaediatric nurse46Nurses (3)Clinical social workerChief physicianMedical doctor\n\nParticipants\nPaediatric nurses (2)\nNurses (2)\nClinical social worker\nOncology nurse\nPaediatric nurses (2)\nNurse\nChief Physician\nSpecialist in psychology\nPaediatrician\nPhysiotherapist\nPhysiotherapist\nPaediatric nurse\nNurses (3)\nClinical social worker\nChief physician\nMedical doctor\nHCPs from three paediatric units in two hospitals were recruited using purposeful sampling, including a variety of professions. Both hospitals are located in Eastern Norway. The units include different departments for children aged 0–18 with a wide range of diagnoses. A total of 21 participants, of whom 19 (90%) were female, participated in 4 focus groups including 2–9 participants (Table 1). Most of the participants had worked with children with life-threatening and life-limiting conditions for more than 10 years. Only two had fewer than two years of experience and some had more than 20 years. Without giving any reason, four HCPs from one of the units withdrew their consent, which is why one of the groups ended up including only two participants. The other three groups included four, six and nine participants. We have no information about the HCPs who declined invitations to participate.\n\nTable 1ParticipantsFocus group numberNumber of participantsProfessions14Paediatric nurses (2)Nurses (2)29Clinical social workerOncology nursePaediatric nurses (2)NurseChief PhysicianSpecialist in psychologyPaediatricianPhysiotherapist32PhysiotherapistPaediatric nurse46Nurses (3)Clinical social workerChief physicianMedical doctor\n\nParticipants\nPaediatric nurses (2)\nNurses (2)\nClinical social worker\nOncology nurse\nPaediatric nurses (2)\nNurse\nChief Physician\nSpecialist in psychology\nPaediatrician\nPhysiotherapist\nPhysiotherapist\nPaediatric nurse\nNurses (3)\nClinical social worker\nChief physician\nMedical doctor\n[SUBTITLE] Data collection [SUBSECTION] The four focus group meetings were convened between November 2019 and February 2020 in a suitable location at the hospital where the participants worked. The focus groups were moderated by pairs of researchers (AL and KR or LGK and EAF): one researcher led the conversations, and the other assisted and took notes during the focus group. All the moderators are female HCPs with a PhD or a master’s degree. None had any prior professional or private relationship with the participants. All the focus groups started with a short presentation of the aim of the study and the researchers’ background and research interests. A semi-structured interview guide was used to focus the conversation and keep it within relevant topics. The guide was developed based on previous research and included open-ended questions related to the concepts of PPC, alleviation and end-of-life care. The participants were also encouraged to illustrate these concepts by sharing their experiences from working with the children and their parents [10]. The interview guide was not piloted, but the questions were discussed with a reference group including HCPs, service user representatives and researchers to facilitate relevant and clear questions. The focus groups lasted from 45 to 90 min and were audio-recorded.\nThe four focus group meetings were convened between November 2019 and February 2020 in a suitable location at the hospital where the participants worked. The focus groups were moderated by pairs of researchers (AL and KR or LGK and EAF): one researcher led the conversations, and the other assisted and took notes during the focus group. All the moderators are female HCPs with a PhD or a master’s degree. None had any prior professional or private relationship with the participants. All the focus groups started with a short presentation of the aim of the study and the researchers’ background and research interests. A semi-structured interview guide was used to focus the conversation and keep it within relevant topics. The guide was developed based on previous research and included open-ended questions related to the concepts of PPC, alleviation and end-of-life care. The participants were also encouraged to illustrate these concepts by sharing their experiences from working with the children and their parents [10]. The interview guide was not piloted, but the questions were discussed with a reference group including HCPs, service user representatives and researchers to facilitate relevant and clear questions. The focus groups lasted from 45 to 90 min and were audio-recorded.\n[SUBTITLE] Data analysis [SUBSECTION] The focus group interviews were transcribed verbatim by a professional transcriber and imported into the qualitative software package NVivo 12 to aid the analysis. The data analysis was undertaken following a thematic analysis procedure [15]. We applied an inductive approach, meaning that the data were coded without trying to fit a pre-existing coding frame [15]. First, the entire data set was screened for stories of PPC. In addition to the stories shared on request, we included all utterances made to provide an example or illustrate a point when reflecting on what PPC is. The transcripts were read by all the authors to obtain an overall first impression of their content. Next, three authors (KR, HH and AW) jointly coded the transcripts section by section aiming to capture the essence of the data before grouping the codes into semantically related themes determined by perceived patterns. A preliminary set of themes with codes was presented to the other authors, who all provided their comments. Based on the feedback, the first and last authors (KR and EAF) reviewed and refined the themes and validated them against the data set. To tell the story of each theme, a detailed analysis of each theme was written. Lastly, the themes were named with the aim of providing an understanding of each theme’s content [15]. Quotations from the focus groups used in the article were translated into English by the researchers in collaboration. Minor alterations were made to the text to ensure anonymity.\nDependability was enhanced by being explicit about the data analysis process and how this was systematically carried out. Credibility was enhanced through investigator triangulation in the data analysis [16]. All the researchers were involved in one or more steps of the analysis, and we challenged each other to provide alternative interpretations and reach intersubjectivity. All the authors are skilled researchers, and most have extensive experience in qualitative research.\nThe focus group interviews were transcribed verbatim by a professional transcriber and imported into the qualitative software package NVivo 12 to aid the analysis. The data analysis was undertaken following a thematic analysis procedure [15]. We applied an inductive approach, meaning that the data were coded without trying to fit a pre-existing coding frame [15]. First, the entire data set was screened for stories of PPC. In addition to the stories shared on request, we included all utterances made to provide an example or illustrate a point when reflecting on what PPC is. The transcripts were read by all the authors to obtain an overall first impression of their content. Next, three authors (KR, HH and AW) jointly coded the transcripts section by section aiming to capture the essence of the data before grouping the codes into semantically related themes determined by perceived patterns. A preliminary set of themes with codes was presented to the other authors, who all provided their comments. Based on the feedback, the first and last authors (KR and EAF) reviewed and refined the themes and validated them against the data set. To tell the story of each theme, a detailed analysis of each theme was written. Lastly, the themes were named with the aim of providing an understanding of each theme’s content [15]. Quotations from the focus groups used in the article were translated into English by the researchers in collaboration. Minor alterations were made to the text to ensure anonymity.\nDependability was enhanced by being explicit about the data analysis process and how this was systematically carried out. Credibility was enhanced through investigator triangulation in the data analysis [16]. All the researchers were involved in one or more steps of the analysis, and we challenged each other to provide alternative interpretations and reach intersubjectivity. All the authors are skilled researchers, and most have extensive experience in qualitative research.\n[SUBTITLE] Ethical considerations [SUBSECTION] The project was reviewed by the Norwegian Data Protection Official for research, who concluded that it was in accordance with the Personal Data Act (reference number 935,944). The local data protection officers at the two participating hospitals approved the study. The study participants were HCPs, and the project did not collect data about health and illness. Thus, the project did not require permission from a regional committee for research ethics. The participating HCPs received information about the aim of the study and the procedure for data collection and gave written consent to participate after being reassured about confidentiality issues and their right to withdraw.\nThe project was reviewed by the Norwegian Data Protection Official for research, who concluded that it was in accordance with the Personal Data Act (reference number 935,944). The local data protection officers at the two participating hospitals approved the study. The study participants were HCPs, and the project did not collect data about health and illness. Thus, the project did not require permission from a regional committee for research ethics. The participating HCPs received information about the aim of the study and the procedure for data collection and gave written consent to participate after being reassured about confidentiality issues and their right to withdraw.", "This study included qualitative data obtained through focus group interviews with HCPs and emerged from a project exploring HCPs’ understanding of PPC [11]. The project was not originally designed to elicit narratives; however, during the focus groups, the HCPs told stories or gave specific examples to illustrate their comprehension of PPC. We decided to subtract these stories and all the utterances made from the initial material to provide examples or illustrate points when reflecting on what PPC is. Thus, the data was not a part of the material analysed in the first article. We explored the stories in more depth, with the assumption that this may provide an enhanced understanding of what HCPs comprehend when they talk about PPC. The reporting of the study was guided by the consolidated criteria for reporting qualitative research [14].", "HCPs from three paediatric units in two hospitals were recruited using purposeful sampling, including a variety of professions. Both hospitals are located in Eastern Norway. The units include different departments for children aged 0–18 with a wide range of diagnoses. A total of 21 participants, of whom 19 (90%) were female, participated in 4 focus groups including 2–9 participants (Table 1). Most of the participants had worked with children with life-threatening and life-limiting conditions for more than 10 years. Only two had fewer than two years of experience and some had more than 20 years. Without giving any reason, four HCPs from one of the units withdrew their consent, which is why one of the groups ended up including only two participants. The other three groups included four, six and nine participants. We have no information about the HCPs who declined invitations to participate.\n\nTable 1ParticipantsFocus group numberNumber of participantsProfessions14Paediatric nurses (2)Nurses (2)29Clinical social workerOncology nursePaediatric nurses (2)NurseChief PhysicianSpecialist in psychologyPaediatricianPhysiotherapist32PhysiotherapistPaediatric nurse46Nurses (3)Clinical social workerChief physicianMedical doctor\n\nParticipants\nPaediatric nurses (2)\nNurses (2)\nClinical social worker\nOncology nurse\nPaediatric nurses (2)\nNurse\nChief Physician\nSpecialist in psychology\nPaediatrician\nPhysiotherapist\nPhysiotherapist\nPaediatric nurse\nNurses (3)\nClinical social worker\nChief physician\nMedical doctor", "The four focus group meetings were convened between November 2019 and February 2020 in a suitable location at the hospital where the participants worked. The focus groups were moderated by pairs of researchers (AL and KR or LGK and EAF): one researcher led the conversations, and the other assisted and took notes during the focus group. All the moderators are female HCPs with a PhD or a master’s degree. None had any prior professional or private relationship with the participants. All the focus groups started with a short presentation of the aim of the study and the researchers’ background and research interests. A semi-structured interview guide was used to focus the conversation and keep it within relevant topics. The guide was developed based on previous research and included open-ended questions related to the concepts of PPC, alleviation and end-of-life care. The participants were also encouraged to illustrate these concepts by sharing their experiences from working with the children and their parents [10]. The interview guide was not piloted, but the questions were discussed with a reference group including HCPs, service user representatives and researchers to facilitate relevant and clear questions. The focus groups lasted from 45 to 90 min and were audio-recorded.", "The focus group interviews were transcribed verbatim by a professional transcriber and imported into the qualitative software package NVivo 12 to aid the analysis. The data analysis was undertaken following a thematic analysis procedure [15]. We applied an inductive approach, meaning that the data were coded without trying to fit a pre-existing coding frame [15]. First, the entire data set was screened for stories of PPC. In addition to the stories shared on request, we included all utterances made to provide an example or illustrate a point when reflecting on what PPC is. The transcripts were read by all the authors to obtain an overall first impression of their content. Next, three authors (KR, HH and AW) jointly coded the transcripts section by section aiming to capture the essence of the data before grouping the codes into semantically related themes determined by perceived patterns. A preliminary set of themes with codes was presented to the other authors, who all provided their comments. Based on the feedback, the first and last authors (KR and EAF) reviewed and refined the themes and validated them against the data set. To tell the story of each theme, a detailed analysis of each theme was written. Lastly, the themes were named with the aim of providing an understanding of each theme’s content [15]. Quotations from the focus groups used in the article were translated into English by the researchers in collaboration. Minor alterations were made to the text to ensure anonymity.\nDependability was enhanced by being explicit about the data analysis process and how this was systematically carried out. Credibility was enhanced through investigator triangulation in the data analysis [16]. All the researchers were involved in one or more steps of the analysis, and we challenged each other to provide alternative interpretations and reach intersubjectivity. All the authors are skilled researchers, and most have extensive experience in qualitative research.", "The project was reviewed by the Norwegian Data Protection Official for research, who concluded that it was in accordance with the Personal Data Act (reference number 935,944). The local data protection officers at the two participating hospitals approved the study. The study participants were HCPs, and the project did not collect data about health and illness. Thus, the project did not require permission from a regional committee for research ethics. The participating HCPs received information about the aim of the study and the procedure for data collection and gave written consent to participate after being reassured about confidentiality issues and their right to withdraw.", "The HCPs seemed attentive to having the child with a life-threatening or life-limiting illness at the centre of their care. Several participants spoke about the importance of supporting a sense of normality and mentioned that the parents as well as the child aimed for ordinary days at home, school or kindergarten: “I believe that most parents want their child’s days to be normal and good until the end and that it’s not going to be something else”. (Group 3)\nStories were told that illustrate how the HCPs managed to create spaces for play and fun for the child despite working in a hospital and being responsible for clinical procedures. They gave examples of how they used medical equipment as props for play and how these playful situations opened up conversations with the child about sensitive and important topics. In one story, the nurse explained that they made the hospital stay so engaging that the child looked forward to treatment and felt sorry for leaving. Another nurse elaborated on a playful moment with a little girl with a life-limiting illness engaging in imaginary play:…and then the girl says: “At my prom, I want a dress like that”, and I said “Really? Wow, that’s cool”, and then she kind of realizes that she’s never going to a prom and then she says: “Well, well…it would have been cool, right?” and I responded “Yeah, it would have been really cool” (Group 2).\n…and then the girl says: “At my prom, I want a dress like that”, and I said “Really? Wow, that’s cool”, and then she kind of realizes that she’s never going to a prom and then she says: “Well, well…it would have been cool, right?” and I responded “Yeah, it would have been really cool” (Group 2).\nThe nurse reflected on the importance of being present in the moment and allowing the child to enjoy the thought of adolescence even though her life expectancy was very limited.", "The participants spoke about different ways of providing support through adjusted information and tailored care. In their stories, they emphasized the importance of sufficient, timely information. One participant described how a mother once said that she did not understand that her child could die while being on a ventilator and that the lungs could still collapse. The participant had carried this mother’s utterance with her ever since, and she pointed out that making sure that parents understand why their child actually dies is significant. Another participant described how important it is to be able to recognize when the parents communicate a need for more knowledge and to provide accurate information when it is called for. However, the participants also referred to challenging situations in which the parents and HCPs were not in step with each other. One participant admitted that if HCPs believe the parents are informed about the child’s situation and suddenly realize that they are not, this may cause delays in procedures and irritation:I try to be observant to this, both in myself and others. It’s important to avoid a moralizing attitude: ‘Do they [the parents] still not understand?’ Because it creates hassle and annoyance when the parents are not sufficiently informed and up to date on the child’s situation. (Group 2)\nI try to be observant to this, both in myself and others. It’s important to avoid a moralizing attitude: ‘Do they [the parents] still not understand?’ Because it creates hassle and annoyance when the parents are not sufficiently informed and up to date on the child’s situation. (Group 2)\nThe participants gave several examples of how they adjust and tailor their care for families to make them feel safe in acute as well as more stable situations. They spoke about the importance of working as a team around the family. One participant described it as creating layers upon layers of love around the child. This expression derived from a particular story about a child who was going to be placed in foster care but was too sick to leave the hospital. With help from the HCP, the child’s birth mother and foster mother moved into the children’s ward. With support from the multidisciplinary team, they cared for the child together during the last weeks of the child’s life.…and the foster mother stepped aside to let the birth mother hold the child close, day and night (…) and the baby received so many layers of love (…) And the HCPs were amazingly professional and put things aside to let it happen. (Group 2)\n…and the foster mother stepped aside to let the birth mother hold the child close, day and night (…) and the baby received so many layers of love (…) And the HCPs were amazingly professional and put things aside to let it happen. (Group 2)\nLater, the birth mother explained that, despite the great tragedy, these had been the finest weeks of her life. This situation was resource-demanding for the unit, but all the HCPs stood out as highly professional and were willing to make an extra effort in the extraordinary situation.\nGoing that extra mile for children and their families was a recurring theme in the HCPs’ stories. Witnessing severe and distressing situations seemed to make the HCPs mobilize to make the days, weeks and months as good as possible for the child, their parents and their siblings. Many of the participants’ stories were about creating safe spaces for families at the hospital, giving the child, parents and siblings moments to breathe. There were also examples of systems failing to support the families adequately and lack of coordination resulting in disorganized care; however, most stories were examples of how HCPs managed to work together, emphasizing the importance of shared information. One participant said:One of the finest moments that I’ve witnessed was when a nurse who knew the family well took the initiative to play with an older sibling of a child who would soon die. The nurse took the sibling outside in the hallway, and they talked and played it out. They had a good relationship. The nurse knew very well what was about to happen because she had been in the room when the message about the child’s impending death was given. The more people working around the family, the more important it is that we have the same information and work as a team. (Group 4)\nOne of the finest moments that I’ve witnessed was when a nurse who knew the family well took the initiative to play with an older sibling of a child who would soon die. The nurse took the sibling outside in the hallway, and they talked and played it out. They had a good relationship. The nurse knew very well what was about to happen because she had been in the room when the message about the child’s impending death was given. The more people working around the family, the more important it is that we have the same information and work as a team. (Group 4)", "Most of the stories emerged from critical situations or when the child’s illness was worsening, especially about initiating advance care planning with the end of the child’s life in mind. Some described the importance of seizing that particular moment when the parents and the child are ready and receptive to these difficult conversations. The HCPs gave examples of when the parents themselves took the initiative, but in most of the stories, the HCPs took the lead. However, as underlined by one participant, engaging in such dialogue requires that the HCPs are present and that they tread carefully: “If you’ve established a good relationship, moments will occur if you’re observant and willing to enter these conversations. For me, this is part of palliative care”. (Group 2)\nWith the children, such conversations could take place during play, and difficult topics could be touched upon without needing to make eye contact. It became evident through the HCPs’ stories that these conversations were tough and that it was sometimes difficult to control their own feelings.These conversations are really hard, it is as if you are about to start crying yourself. But I think it is really, really important [to talk about it], because we have reached the moment when it might happen. (Group 3)\nThese conversations are really hard, it is as if you are about to start crying yourself. But I think it is really, really important [to talk about it], because we have reached the moment when it might happen. (Group 3)\nThrough their stories, the HCPs emphasized the need for advance care plans when the risk of severe incidents increased. The advantages of such plans are to secure continuity and ensure that all important information is available for all the HCPs involved in a possible emergency situation. However, these plans should also be made for the family members so that they know how to act if critical events occur. In several stories, the HCPs described how the care plans included strategies and measures for staying at home as much as possible. One example was given of how specialist care and primary care cooperated, where the child was provided with advanced home care, and the child and the family shifted between being at the hospital and being at home. The family in this story expressed a strong wish to let the child die at home, and the HCPs made great efforts to accommodate this wish. The participant described how all the necessary equipment was installed in the family’s house:It was arranged for him to stay at home. He received pain relief, and I believe the community nurses cared for him, and he spent his last 2–3 weeks in the living room together with his siblings and parents and they all slept together in the same bed. They were kind of… they were together.” (Group 1)\nIt was arranged for him to stay at home. He received pain relief, and I believe the community nurses cared for him, and he spent his last 2–3 weeks in the living room together with his siblings and parents and they all slept together in the same bed. They were kind of… they were together.” (Group 1)\nMaking practical arrangements to facilitate a proper farewell in a safe environment together was a prioritized task. The parents and siblings could focus on being there for the child and for each other while the medical team provided care and pain relief for the child.", "While many stories and examples evolved from cases where the HCPs explained that they managed to provide support and good care, there were also stories about conflicts with parents and distressful experiences. Some HCPs referred to conversations about how to handle a dramatic worsening of the child’s condition (e.g. decisions about not putting the child on a ventilator or stopping medications) and how this had provoked anger and frustration among parents. One HCP described one such situation:... and then there were discussions about withdrawing antibiotics, and we had to tell the parents, and one of them was really devastated and said, ‘You’re giving up on my child!’ whereupon the doctor kindly responded, ‘No, we’re not, we are comforting her during her last days’”. (Group 2)\n... and then there were discussions about withdrawing antibiotics, and we had to tell the parents, and one of them was really devastated and said, ‘You’re giving up on my child!’ whereupon the doctor kindly responded, ‘No, we’re not, we are comforting her during her last days’”. (Group 2)\nIn some cases, the participants noted that it was the HCPs and not the parents who pushed too hard by suggesting a new treatment, which turned out to result in complications or negative side effects. Another story illustrated the tension that can occur between HCPs and parents when the child is expected to die within a short time. In this case, one HCP accused a parent of hastening the baby’s death by withholding nutrition. This resulted in a deep conflict among the staff, where some supported the nurse and some sympathized with the parents. Eventually, external counselling was required to resolve the dispute among the staff. After the baby died, the parents reported that the whole situation had added even more to their grief. The participant described this as an ethically demanding situation, and, in retrospect, she was struck by how good intentions can lead to conflict and how emotionally involved one may become.\nExamples were also given of parents who refused to acknowledge that their child had a life-limiting disease and instead of planning for the end spent all their time searching for a cure. The HCPs experienced it as particularly hard to witness how false hope was being sold through experimental treatment and that parents were willing to squander that precious, limited time left of their child’s life:There was a family that I was never in a position to speak with, because their attention was always directed at something else. It was impossible to talk to them about anything, because they were always focusing on going off somewhere to find a cure. (Group 2)\nThere was a family that I was never in a position to speak with, because their attention was always directed at something else. It was impossible to talk to them about anything, because they were always focusing on going off somewhere to find a cure. (Group 2)\nIn these stories, the parents searched for opportunities to travel far with their child to try experimental treatment, resulting in long periods away from home and separation between family members. The participant found it challenging that the parents were never available to make plans for good palliative care.", "Using stories as a basis for analysis is a strength of the present study. When we tell stories we use language available and familiar to us and to our audience [1]. While HCPs’ understanding and experience of PPC has been investigated through qualitative interview studies, few, if any, have used storytelling as a research tool to explore their insights. Compared to other qualitative research methods, stories can generate more nuanced, contextualized and culturally reflective information [32]. However, participating in focus groups instead of individual interviews may have limited the HCPs’ time and opportunity to elaborate on their stories. In only one of the four focus group interviews did the interviewer explicitly ask the participants to tell a story or give an example from practice. This is an important limitation. If the same request had been given to the remaining groups, we might have collected richer data. Nevertheless, the participants in all the focus groups spontaneously used their experiences to elaborate on their comprehension of PPC through stories and examples, with little prompting required from the interviewers. Some chose to supplement their colleagues’ stories or follow up with their own version of the same case, while at other times listening to a story served as a cue for a new story mirroring or contrasting with the former. The stories stood out as significant to the storyteller, and it was noticeable that they had been told and reflected over before. This underlines the significant potential of stories to capture the participants’ lived experiences. One limitation to be noted is that the sample was predominantly female and thus the results may be less representative for male HCPs." ]
[ null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Methods", "Design", "Participants", "Data collection", "Data analysis", "Ethical considerations", "Results", "Creating spaces for normality", "Providing tailored support for the family", "Careful preparations for saying goodbye", "Experiencing dilemmas and distress", "Discussion", "Strengths and limitations", "Conclusion" ]
[ "Storytelling is a fundamental part of being human. Through stories information can be shared in a way that creates an emotional connection and by that provides a deeper understanding of the storyteller’s experiences. Thus, storytelling has the potential to bring new perspectives to the Table [1]. When telling stories about specific events from patients´ lives, health care professionals (HCPs) not only describe the patients’ medical situations. By sharing patient stories, HCPs also communicate their attitudes, values and beliefs about caring and treatment [2]. The present study used storytelling as a methodological approach to investigate HCPs’ understanding of paediatric palliative care (PPC). According to the World Health Organization (WHO), PPC should be the standard of care for children with life-threatening or life-limiting conditions and should be integrated with prevention, early diagnosis and treatment [3, 4]. PPC is defined as “the active total care of the child’s body, mind and spirit, and [this] also involves giving support to the family” [5]. Children (0–18 years) and their families should be provided with interdisciplinary care aiming to promote their physical, psychological, social and spiritual well-being [4]. Early implementation of PPC through active and integrated support is endorsed as beneficial to the well-being of the child and the family [3, 6, 7]. HCPs may display good knowledge of the principles of palliative care, but their comprehension of the concept, their way of thinking and their reported reasons for referrals to PPC demonstrate that they associate PPC with death and dying [8, 9]. Research reports that HCPs find the concept challenging to understand and implement and that it evokes negative emotions [10–12]. Consequently, early integration of PPC may be delayed and many children are not referred to PPC until late in their illness trajectory [12]. Among the barriers reported for implementing PPC at an early stage are uncertainty and confusion about the concept, as well as stigma related to the term [10, 13]. In our recent study of interdisciplinary HCPs’ understanding and implementation of PPC, we found that the participants viewed the concept as unfamiliar and not meaningful [11]. HCPs associated PPC with death and dying and used words like scary, burdensome, dramatic and lack of active treatment to describe their comprehension of the concept [11].\nAttending to stories of HCPs´ clinical experience with children and their families may provide further insights than solely through answering questions and thus be a valuable approach to gain an even richer account of their understanding of PPC. Thus, the aim of this study was to explore stories of PPC narrated by HCPs to enhance our knowledge of their understanding of what PPC entails.", "[SUBTITLE] Design [SUBSECTION] This study included qualitative data obtained through focus group interviews with HCPs and emerged from a project exploring HCPs’ understanding of PPC [11]. The project was not originally designed to elicit narratives; however, during the focus groups, the HCPs told stories or gave specific examples to illustrate their comprehension of PPC. We decided to subtract these stories and all the utterances made from the initial material to provide examples or illustrate points when reflecting on what PPC is. Thus, the data was not a part of the material analysed in the first article. We explored the stories in more depth, with the assumption that this may provide an enhanced understanding of what HCPs comprehend when they talk about PPC. The reporting of the study was guided by the consolidated criteria for reporting qualitative research [14].\nThis study included qualitative data obtained through focus group interviews with HCPs and emerged from a project exploring HCPs’ understanding of PPC [11]. The project was not originally designed to elicit narratives; however, during the focus groups, the HCPs told stories or gave specific examples to illustrate their comprehension of PPC. We decided to subtract these stories and all the utterances made from the initial material to provide examples or illustrate points when reflecting on what PPC is. Thus, the data was not a part of the material analysed in the first article. We explored the stories in more depth, with the assumption that this may provide an enhanced understanding of what HCPs comprehend when they talk about PPC. The reporting of the study was guided by the consolidated criteria for reporting qualitative research [14].\n[SUBTITLE] Participants [SUBSECTION] HCPs from three paediatric units in two hospitals were recruited using purposeful sampling, including a variety of professions. Both hospitals are located in Eastern Norway. The units include different departments for children aged 0–18 with a wide range of diagnoses. A total of 21 participants, of whom 19 (90%) were female, participated in 4 focus groups including 2–9 participants (Table 1). Most of the participants had worked with children with life-threatening and life-limiting conditions for more than 10 years. Only two had fewer than two years of experience and some had more than 20 years. Without giving any reason, four HCPs from one of the units withdrew their consent, which is why one of the groups ended up including only two participants. The other three groups included four, six and nine participants. We have no information about the HCPs who declined invitations to participate.\n\nTable 1ParticipantsFocus group numberNumber of participantsProfessions14Paediatric nurses (2)Nurses (2)29Clinical social workerOncology nursePaediatric nurses (2)NurseChief PhysicianSpecialist in psychologyPaediatricianPhysiotherapist32PhysiotherapistPaediatric nurse46Nurses (3)Clinical social workerChief physicianMedical doctor\n\nParticipants\nPaediatric nurses (2)\nNurses (2)\nClinical social worker\nOncology nurse\nPaediatric nurses (2)\nNurse\nChief Physician\nSpecialist in psychology\nPaediatrician\nPhysiotherapist\nPhysiotherapist\nPaediatric nurse\nNurses (3)\nClinical social worker\nChief physician\nMedical doctor\nHCPs from three paediatric units in two hospitals were recruited using purposeful sampling, including a variety of professions. Both hospitals are located in Eastern Norway. The units include different departments for children aged 0–18 with a wide range of diagnoses. A total of 21 participants, of whom 19 (90%) were female, participated in 4 focus groups including 2–9 participants (Table 1). Most of the participants had worked with children with life-threatening and life-limiting conditions for more than 10 years. Only two had fewer than two years of experience and some had more than 20 years. Without giving any reason, four HCPs from one of the units withdrew their consent, which is why one of the groups ended up including only two participants. The other three groups included four, six and nine participants. We have no information about the HCPs who declined invitations to participate.\n\nTable 1ParticipantsFocus group numberNumber of participantsProfessions14Paediatric nurses (2)Nurses (2)29Clinical social workerOncology nursePaediatric nurses (2)NurseChief PhysicianSpecialist in psychologyPaediatricianPhysiotherapist32PhysiotherapistPaediatric nurse46Nurses (3)Clinical social workerChief physicianMedical doctor\n\nParticipants\nPaediatric nurses (2)\nNurses (2)\nClinical social worker\nOncology nurse\nPaediatric nurses (2)\nNurse\nChief Physician\nSpecialist in psychology\nPaediatrician\nPhysiotherapist\nPhysiotherapist\nPaediatric nurse\nNurses (3)\nClinical social worker\nChief physician\nMedical doctor\n[SUBTITLE] Data collection [SUBSECTION] The four focus group meetings were convened between November 2019 and February 2020 in a suitable location at the hospital where the participants worked. The focus groups were moderated by pairs of researchers (AL and KR or LGK and EAF): one researcher led the conversations, and the other assisted and took notes during the focus group. All the moderators are female HCPs with a PhD or a master’s degree. None had any prior professional or private relationship with the participants. All the focus groups started with a short presentation of the aim of the study and the researchers’ background and research interests. A semi-structured interview guide was used to focus the conversation and keep it within relevant topics. The guide was developed based on previous research and included open-ended questions related to the concepts of PPC, alleviation and end-of-life care. The participants were also encouraged to illustrate these concepts by sharing their experiences from working with the children and their parents [10]. The interview guide was not piloted, but the questions were discussed with a reference group including HCPs, service user representatives and researchers to facilitate relevant and clear questions. The focus groups lasted from 45 to 90 min and were audio-recorded.\nThe four focus group meetings were convened between November 2019 and February 2020 in a suitable location at the hospital where the participants worked. The focus groups were moderated by pairs of researchers (AL and KR or LGK and EAF): one researcher led the conversations, and the other assisted and took notes during the focus group. All the moderators are female HCPs with a PhD or a master’s degree. None had any prior professional or private relationship with the participants. All the focus groups started with a short presentation of the aim of the study and the researchers’ background and research interests. A semi-structured interview guide was used to focus the conversation and keep it within relevant topics. The guide was developed based on previous research and included open-ended questions related to the concepts of PPC, alleviation and end-of-life care. The participants were also encouraged to illustrate these concepts by sharing their experiences from working with the children and their parents [10]. The interview guide was not piloted, but the questions were discussed with a reference group including HCPs, service user representatives and researchers to facilitate relevant and clear questions. The focus groups lasted from 45 to 90 min and were audio-recorded.\n[SUBTITLE] Data analysis [SUBSECTION] The focus group interviews were transcribed verbatim by a professional transcriber and imported into the qualitative software package NVivo 12 to aid the analysis. The data analysis was undertaken following a thematic analysis procedure [15]. We applied an inductive approach, meaning that the data were coded without trying to fit a pre-existing coding frame [15]. First, the entire data set was screened for stories of PPC. In addition to the stories shared on request, we included all utterances made to provide an example or illustrate a point when reflecting on what PPC is. The transcripts were read by all the authors to obtain an overall first impression of their content. Next, three authors (KR, HH and AW) jointly coded the transcripts section by section aiming to capture the essence of the data before grouping the codes into semantically related themes determined by perceived patterns. A preliminary set of themes with codes was presented to the other authors, who all provided their comments. Based on the feedback, the first and last authors (KR and EAF) reviewed and refined the themes and validated them against the data set. To tell the story of each theme, a detailed analysis of each theme was written. Lastly, the themes were named with the aim of providing an understanding of each theme’s content [15]. Quotations from the focus groups used in the article were translated into English by the researchers in collaboration. Minor alterations were made to the text to ensure anonymity.\nDependability was enhanced by being explicit about the data analysis process and how this was systematically carried out. Credibility was enhanced through investigator triangulation in the data analysis [16]. All the researchers were involved in one or more steps of the analysis, and we challenged each other to provide alternative interpretations and reach intersubjectivity. All the authors are skilled researchers, and most have extensive experience in qualitative research.\nThe focus group interviews were transcribed verbatim by a professional transcriber and imported into the qualitative software package NVivo 12 to aid the analysis. The data analysis was undertaken following a thematic analysis procedure [15]. We applied an inductive approach, meaning that the data were coded without trying to fit a pre-existing coding frame [15]. First, the entire data set was screened for stories of PPC. In addition to the stories shared on request, we included all utterances made to provide an example or illustrate a point when reflecting on what PPC is. The transcripts were read by all the authors to obtain an overall first impression of their content. Next, three authors (KR, HH and AW) jointly coded the transcripts section by section aiming to capture the essence of the data before grouping the codes into semantically related themes determined by perceived patterns. A preliminary set of themes with codes was presented to the other authors, who all provided their comments. Based on the feedback, the first and last authors (KR and EAF) reviewed and refined the themes and validated them against the data set. To tell the story of each theme, a detailed analysis of each theme was written. Lastly, the themes were named with the aim of providing an understanding of each theme’s content [15]. Quotations from the focus groups used in the article were translated into English by the researchers in collaboration. Minor alterations were made to the text to ensure anonymity.\nDependability was enhanced by being explicit about the data analysis process and how this was systematically carried out. Credibility was enhanced through investigator triangulation in the data analysis [16]. All the researchers were involved in one or more steps of the analysis, and we challenged each other to provide alternative interpretations and reach intersubjectivity. All the authors are skilled researchers, and most have extensive experience in qualitative research.\n[SUBTITLE] Ethical considerations [SUBSECTION] The project was reviewed by the Norwegian Data Protection Official for research, who concluded that it was in accordance with the Personal Data Act (reference number 935,944). The local data protection officers at the two participating hospitals approved the study. The study participants were HCPs, and the project did not collect data about health and illness. Thus, the project did not require permission from a regional committee for research ethics. The participating HCPs received information about the aim of the study and the procedure for data collection and gave written consent to participate after being reassured about confidentiality issues and their right to withdraw.\nThe project was reviewed by the Norwegian Data Protection Official for research, who concluded that it was in accordance with the Personal Data Act (reference number 935,944). The local data protection officers at the two participating hospitals approved the study. The study participants were HCPs, and the project did not collect data about health and illness. Thus, the project did not require permission from a regional committee for research ethics. The participating HCPs received information about the aim of the study and the procedure for data collection and gave written consent to participate after being reassured about confidentiality issues and their right to withdraw.", "This study included qualitative data obtained through focus group interviews with HCPs and emerged from a project exploring HCPs’ understanding of PPC [11]. The project was not originally designed to elicit narratives; however, during the focus groups, the HCPs told stories or gave specific examples to illustrate their comprehension of PPC. We decided to subtract these stories and all the utterances made from the initial material to provide examples or illustrate points when reflecting on what PPC is. Thus, the data was not a part of the material analysed in the first article. We explored the stories in more depth, with the assumption that this may provide an enhanced understanding of what HCPs comprehend when they talk about PPC. The reporting of the study was guided by the consolidated criteria for reporting qualitative research [14].", "HCPs from three paediatric units in two hospitals were recruited using purposeful sampling, including a variety of professions. Both hospitals are located in Eastern Norway. The units include different departments for children aged 0–18 with a wide range of diagnoses. A total of 21 participants, of whom 19 (90%) were female, participated in 4 focus groups including 2–9 participants (Table 1). Most of the participants had worked with children with life-threatening and life-limiting conditions for more than 10 years. Only two had fewer than two years of experience and some had more than 20 years. Without giving any reason, four HCPs from one of the units withdrew their consent, which is why one of the groups ended up including only two participants. The other three groups included four, six and nine participants. We have no information about the HCPs who declined invitations to participate.\n\nTable 1ParticipantsFocus group numberNumber of participantsProfessions14Paediatric nurses (2)Nurses (2)29Clinical social workerOncology nursePaediatric nurses (2)NurseChief PhysicianSpecialist in psychologyPaediatricianPhysiotherapist32PhysiotherapistPaediatric nurse46Nurses (3)Clinical social workerChief physicianMedical doctor\n\nParticipants\nPaediatric nurses (2)\nNurses (2)\nClinical social worker\nOncology nurse\nPaediatric nurses (2)\nNurse\nChief Physician\nSpecialist in psychology\nPaediatrician\nPhysiotherapist\nPhysiotherapist\nPaediatric nurse\nNurses (3)\nClinical social worker\nChief physician\nMedical doctor", "The four focus group meetings were convened between November 2019 and February 2020 in a suitable location at the hospital where the participants worked. The focus groups were moderated by pairs of researchers (AL and KR or LGK and EAF): one researcher led the conversations, and the other assisted and took notes during the focus group. All the moderators are female HCPs with a PhD or a master’s degree. None had any prior professional or private relationship with the participants. All the focus groups started with a short presentation of the aim of the study and the researchers’ background and research interests. A semi-structured interview guide was used to focus the conversation and keep it within relevant topics. The guide was developed based on previous research and included open-ended questions related to the concepts of PPC, alleviation and end-of-life care. The participants were also encouraged to illustrate these concepts by sharing their experiences from working with the children and their parents [10]. The interview guide was not piloted, but the questions were discussed with a reference group including HCPs, service user representatives and researchers to facilitate relevant and clear questions. The focus groups lasted from 45 to 90 min and were audio-recorded.", "The focus group interviews were transcribed verbatim by a professional transcriber and imported into the qualitative software package NVivo 12 to aid the analysis. The data analysis was undertaken following a thematic analysis procedure [15]. We applied an inductive approach, meaning that the data were coded without trying to fit a pre-existing coding frame [15]. First, the entire data set was screened for stories of PPC. In addition to the stories shared on request, we included all utterances made to provide an example or illustrate a point when reflecting on what PPC is. The transcripts were read by all the authors to obtain an overall first impression of their content. Next, three authors (KR, HH and AW) jointly coded the transcripts section by section aiming to capture the essence of the data before grouping the codes into semantically related themes determined by perceived patterns. A preliminary set of themes with codes was presented to the other authors, who all provided their comments. Based on the feedback, the first and last authors (KR and EAF) reviewed and refined the themes and validated them against the data set. To tell the story of each theme, a detailed analysis of each theme was written. Lastly, the themes were named with the aim of providing an understanding of each theme’s content [15]. Quotations from the focus groups used in the article were translated into English by the researchers in collaboration. Minor alterations were made to the text to ensure anonymity.\nDependability was enhanced by being explicit about the data analysis process and how this was systematically carried out. Credibility was enhanced through investigator triangulation in the data analysis [16]. All the researchers were involved in one or more steps of the analysis, and we challenged each other to provide alternative interpretations and reach intersubjectivity. All the authors are skilled researchers, and most have extensive experience in qualitative research.", "The project was reviewed by the Norwegian Data Protection Official for research, who concluded that it was in accordance with the Personal Data Act (reference number 935,944). The local data protection officers at the two participating hospitals approved the study. The study participants were HCPs, and the project did not collect data about health and illness. Thus, the project did not require permission from a regional committee for research ethics. The participating HCPs received information about the aim of the study and the procedure for data collection and gave written consent to participate after being reassured about confidentiality issues and their right to withdraw.", "Following the analysis, four themes were identified illustrating what PPC entails to the participants: creating spaces for normality, providing tailored support for the family, careful preparations for saying goodbye and experiencing dilemmas and distress (Table 2).\n\nTable 2Themes and corresponding codes and examples of quotes from the storiesThemesCodesQuotesCreating spaces for normalityEveryday life and playSupporting normality\nLike that one time, we hid under the blanket because she did not want to talk about her disease. When the doctor tried, the girl hid under the blanket, and I asked to join her. So we stayed there together. We kind of had an alliance. It is about seeing them exactly where they are.\nProviding tailored support for the familyProviding knowledge and supportProviding extraordinary careLayer upon layerCreating safety\nThe parents felt safe when coming here, and they felt safe to leave the room. They could breathe a little while they knew she was taken care of.\nCareful preparations for saying goodbyeMaking arrangements for saying goodbyeBeing togetherCapturing the right moments for difficult conversationsRealizing that death is imminent\nAnd we sat there at the outpatient clinic and I thought to myself that I have to tell them…because I do not know how long he would live, so we should talk about that. And at that moment I was given an opportunity to say that if he came back with an infection we might not be able to do…to save him…or he could end up on a ventilator and that is not necessarily a good idea because we cannot get them off. So we talked about that.\nExperiencing dilemmas and distressConflicting wishesDilemmas relating to treatment\nIt was obvious to everyone else that the child’s condition was very severe and that she would never recover, and then the parent says “well, that is just what\nyou\nbelieve!” (…) If the parents want treatment, when we have no treatment to offer, there is always someone that makes promises without scientific foundation.\n\n\nThemes and corresponding codes and examples of quotes from the stories\nEveryday life and play\nSupporting normality\nProviding knowledge and support\nProviding extraordinary care\nLayer upon layer\nCreating safety\nMaking arrangements for saying goodbye\nBeing together\nCapturing the right moments for difficult conversations\nRealizing that death is imminent\nConflicting wishes\nDilemmas relating to treatment\n[SUBTITLE] Creating spaces for normality [SUBSECTION] The HCPs seemed attentive to having the child with a life-threatening or life-limiting illness at the centre of their care. Several participants spoke about the importance of supporting a sense of normality and mentioned that the parents as well as the child aimed for ordinary days at home, school or kindergarten: “I believe that most parents want their child’s days to be normal and good until the end and that it’s not going to be something else”. (Group 3)\nStories were told that illustrate how the HCPs managed to create spaces for play and fun for the child despite working in a hospital and being responsible for clinical procedures. They gave examples of how they used medical equipment as props for play and how these playful situations opened up conversations with the child about sensitive and important topics. In one story, the nurse explained that they made the hospital stay so engaging that the child looked forward to treatment and felt sorry for leaving. Another nurse elaborated on a playful moment with a little girl with a life-limiting illness engaging in imaginary play:…and then the girl says: “At my prom, I want a dress like that”, and I said “Really? Wow, that’s cool”, and then she kind of realizes that she’s never going to a prom and then she says: “Well, well…it would have been cool, right?” and I responded “Yeah, it would have been really cool” (Group 2).\n…and then the girl says: “At my prom, I want a dress like that”, and I said “Really? Wow, that’s cool”, and then she kind of realizes that she’s never going to a prom and then she says: “Well, well…it would have been cool, right?” and I responded “Yeah, it would have been really cool” (Group 2).\nThe nurse reflected on the importance of being present in the moment and allowing the child to enjoy the thought of adolescence even though her life expectancy was very limited.\nThe HCPs seemed attentive to having the child with a life-threatening or life-limiting illness at the centre of their care. Several participants spoke about the importance of supporting a sense of normality and mentioned that the parents as well as the child aimed for ordinary days at home, school or kindergarten: “I believe that most parents want their child’s days to be normal and good until the end and that it’s not going to be something else”. (Group 3)\nStories were told that illustrate how the HCPs managed to create spaces for play and fun for the child despite working in a hospital and being responsible for clinical procedures. They gave examples of how they used medical equipment as props for play and how these playful situations opened up conversations with the child about sensitive and important topics. In one story, the nurse explained that they made the hospital stay so engaging that the child looked forward to treatment and felt sorry for leaving. Another nurse elaborated on a playful moment with a little girl with a life-limiting illness engaging in imaginary play:…and then the girl says: “At my prom, I want a dress like that”, and I said “Really? Wow, that’s cool”, and then she kind of realizes that she’s never going to a prom and then she says: “Well, well…it would have been cool, right?” and I responded “Yeah, it would have been really cool” (Group 2).\n…and then the girl says: “At my prom, I want a dress like that”, and I said “Really? Wow, that’s cool”, and then she kind of realizes that she’s never going to a prom and then she says: “Well, well…it would have been cool, right?” and I responded “Yeah, it would have been really cool” (Group 2).\nThe nurse reflected on the importance of being present in the moment and allowing the child to enjoy the thought of adolescence even though her life expectancy was very limited.\n[SUBTITLE] Providing tailored support for the family [SUBSECTION] The participants spoke about different ways of providing support through adjusted information and tailored care. In their stories, they emphasized the importance of sufficient, timely information. One participant described how a mother once said that she did not understand that her child could die while being on a ventilator and that the lungs could still collapse. The participant had carried this mother’s utterance with her ever since, and she pointed out that making sure that parents understand why their child actually dies is significant. Another participant described how important it is to be able to recognize when the parents communicate a need for more knowledge and to provide accurate information when it is called for. However, the participants also referred to challenging situations in which the parents and HCPs were not in step with each other. One participant admitted that if HCPs believe the parents are informed about the child’s situation and suddenly realize that they are not, this may cause delays in procedures and irritation:I try to be observant to this, both in myself and others. It’s important to avoid a moralizing attitude: ‘Do they [the parents] still not understand?’ Because it creates hassle and annoyance when the parents are not sufficiently informed and up to date on the child’s situation. (Group 2)\nI try to be observant to this, both in myself and others. It’s important to avoid a moralizing attitude: ‘Do they [the parents] still not understand?’ Because it creates hassle and annoyance when the parents are not sufficiently informed and up to date on the child’s situation. (Group 2)\nThe participants gave several examples of how they adjust and tailor their care for families to make them feel safe in acute as well as more stable situations. They spoke about the importance of working as a team around the family. One participant described it as creating layers upon layers of love around the child. This expression derived from a particular story about a child who was going to be placed in foster care but was too sick to leave the hospital. With help from the HCP, the child’s birth mother and foster mother moved into the children’s ward. With support from the multidisciplinary team, they cared for the child together during the last weeks of the child’s life.…and the foster mother stepped aside to let the birth mother hold the child close, day and night (…) and the baby received so many layers of love (…) And the HCPs were amazingly professional and put things aside to let it happen. (Group 2)\n…and the foster mother stepped aside to let the birth mother hold the child close, day and night (…) and the baby received so many layers of love (…) And the HCPs were amazingly professional and put things aside to let it happen. (Group 2)\nLater, the birth mother explained that, despite the great tragedy, these had been the finest weeks of her life. This situation was resource-demanding for the unit, but all the HCPs stood out as highly professional and were willing to make an extra effort in the extraordinary situation.\nGoing that extra mile for children and their families was a recurring theme in the HCPs’ stories. Witnessing severe and distressing situations seemed to make the HCPs mobilize to make the days, weeks and months as good as possible for the child, their parents and their siblings. Many of the participants’ stories were about creating safe spaces for families at the hospital, giving the child, parents and siblings moments to breathe. There were also examples of systems failing to support the families adequately and lack of coordination resulting in disorganized care; however, most stories were examples of how HCPs managed to work together, emphasizing the importance of shared information. One participant said:One of the finest moments that I’ve witnessed was when a nurse who knew the family well took the initiative to play with an older sibling of a child who would soon die. The nurse took the sibling outside in the hallway, and they talked and played it out. They had a good relationship. The nurse knew very well what was about to happen because she had been in the room when the message about the child’s impending death was given. The more people working around the family, the more important it is that we have the same information and work as a team. (Group 4)\nOne of the finest moments that I’ve witnessed was when a nurse who knew the family well took the initiative to play with an older sibling of a child who would soon die. The nurse took the sibling outside in the hallway, and they talked and played it out. They had a good relationship. The nurse knew very well what was about to happen because she had been in the room when the message about the child’s impending death was given. The more people working around the family, the more important it is that we have the same information and work as a team. (Group 4)\nThe participants spoke about different ways of providing support through adjusted information and tailored care. In their stories, they emphasized the importance of sufficient, timely information. One participant described how a mother once said that she did not understand that her child could die while being on a ventilator and that the lungs could still collapse. The participant had carried this mother’s utterance with her ever since, and she pointed out that making sure that parents understand why their child actually dies is significant. Another participant described how important it is to be able to recognize when the parents communicate a need for more knowledge and to provide accurate information when it is called for. However, the participants also referred to challenging situations in which the parents and HCPs were not in step with each other. One participant admitted that if HCPs believe the parents are informed about the child’s situation and suddenly realize that they are not, this may cause delays in procedures and irritation:I try to be observant to this, both in myself and others. It’s important to avoid a moralizing attitude: ‘Do they [the parents] still not understand?’ Because it creates hassle and annoyance when the parents are not sufficiently informed and up to date on the child’s situation. (Group 2)\nI try to be observant to this, both in myself and others. It’s important to avoid a moralizing attitude: ‘Do they [the parents] still not understand?’ Because it creates hassle and annoyance when the parents are not sufficiently informed and up to date on the child’s situation. (Group 2)\nThe participants gave several examples of how they adjust and tailor their care for families to make them feel safe in acute as well as more stable situations. They spoke about the importance of working as a team around the family. One participant described it as creating layers upon layers of love around the child. This expression derived from a particular story about a child who was going to be placed in foster care but was too sick to leave the hospital. With help from the HCP, the child’s birth mother and foster mother moved into the children’s ward. With support from the multidisciplinary team, they cared for the child together during the last weeks of the child’s life.…and the foster mother stepped aside to let the birth mother hold the child close, day and night (…) and the baby received so many layers of love (…) And the HCPs were amazingly professional and put things aside to let it happen. (Group 2)\n…and the foster mother stepped aside to let the birth mother hold the child close, day and night (…) and the baby received so many layers of love (…) And the HCPs were amazingly professional and put things aside to let it happen. (Group 2)\nLater, the birth mother explained that, despite the great tragedy, these had been the finest weeks of her life. This situation was resource-demanding for the unit, but all the HCPs stood out as highly professional and were willing to make an extra effort in the extraordinary situation.\nGoing that extra mile for children and their families was a recurring theme in the HCPs’ stories. Witnessing severe and distressing situations seemed to make the HCPs mobilize to make the days, weeks and months as good as possible for the child, their parents and their siblings. Many of the participants’ stories were about creating safe spaces for families at the hospital, giving the child, parents and siblings moments to breathe. There were also examples of systems failing to support the families adequately and lack of coordination resulting in disorganized care; however, most stories were examples of how HCPs managed to work together, emphasizing the importance of shared information. One participant said:One of the finest moments that I’ve witnessed was when a nurse who knew the family well took the initiative to play with an older sibling of a child who would soon die. The nurse took the sibling outside in the hallway, and they talked and played it out. They had a good relationship. The nurse knew very well what was about to happen because she had been in the room when the message about the child’s impending death was given. The more people working around the family, the more important it is that we have the same information and work as a team. (Group 4)\nOne of the finest moments that I’ve witnessed was when a nurse who knew the family well took the initiative to play with an older sibling of a child who would soon die. The nurse took the sibling outside in the hallway, and they talked and played it out. They had a good relationship. The nurse knew very well what was about to happen because she had been in the room when the message about the child’s impending death was given. The more people working around the family, the more important it is that we have the same information and work as a team. (Group 4)\n[SUBTITLE] Careful preparations for saying goodbye [SUBSECTION] Most of the stories emerged from critical situations or when the child’s illness was worsening, especially about initiating advance care planning with the end of the child’s life in mind. Some described the importance of seizing that particular moment when the parents and the child are ready and receptive to these difficult conversations. The HCPs gave examples of when the parents themselves took the initiative, but in most of the stories, the HCPs took the lead. However, as underlined by one participant, engaging in such dialogue requires that the HCPs are present and that they tread carefully: “If you’ve established a good relationship, moments will occur if you’re observant and willing to enter these conversations. For me, this is part of palliative care”. (Group 2)\nWith the children, such conversations could take place during play, and difficult topics could be touched upon without needing to make eye contact. It became evident through the HCPs’ stories that these conversations were tough and that it was sometimes difficult to control their own feelings.These conversations are really hard, it is as if you are about to start crying yourself. But I think it is really, really important [to talk about it], because we have reached the moment when it might happen. (Group 3)\nThese conversations are really hard, it is as if you are about to start crying yourself. But I think it is really, really important [to talk about it], because we have reached the moment when it might happen. (Group 3)\nThrough their stories, the HCPs emphasized the need for advance care plans when the risk of severe incidents increased. The advantages of such plans are to secure continuity and ensure that all important information is available for all the HCPs involved in a possible emergency situation. However, these plans should also be made for the family members so that they know how to act if critical events occur. In several stories, the HCPs described how the care plans included strategies and measures for staying at home as much as possible. One example was given of how specialist care and primary care cooperated, where the child was provided with advanced home care, and the child and the family shifted between being at the hospital and being at home. The family in this story expressed a strong wish to let the child die at home, and the HCPs made great efforts to accommodate this wish. The participant described how all the necessary equipment was installed in the family’s house:It was arranged for him to stay at home. He received pain relief, and I believe the community nurses cared for him, and he spent his last 2–3 weeks in the living room together with his siblings and parents and they all slept together in the same bed. They were kind of… they were together.” (Group 1)\nIt was arranged for him to stay at home. He received pain relief, and I believe the community nurses cared for him, and he spent his last 2–3 weeks in the living room together with his siblings and parents and they all slept together in the same bed. They were kind of… they were together.” (Group 1)\nMaking practical arrangements to facilitate a proper farewell in a safe environment together was a prioritized task. The parents and siblings could focus on being there for the child and for each other while the medical team provided care and pain relief for the child.\nMost of the stories emerged from critical situations or when the child’s illness was worsening, especially about initiating advance care planning with the end of the child’s life in mind. Some described the importance of seizing that particular moment when the parents and the child are ready and receptive to these difficult conversations. The HCPs gave examples of when the parents themselves took the initiative, but in most of the stories, the HCPs took the lead. However, as underlined by one participant, engaging in such dialogue requires that the HCPs are present and that they tread carefully: “If you’ve established a good relationship, moments will occur if you’re observant and willing to enter these conversations. For me, this is part of palliative care”. (Group 2)\nWith the children, such conversations could take place during play, and difficult topics could be touched upon without needing to make eye contact. It became evident through the HCPs’ stories that these conversations were tough and that it was sometimes difficult to control their own feelings.These conversations are really hard, it is as if you are about to start crying yourself. But I think it is really, really important [to talk about it], because we have reached the moment when it might happen. (Group 3)\nThese conversations are really hard, it is as if you are about to start crying yourself. But I think it is really, really important [to talk about it], because we have reached the moment when it might happen. (Group 3)\nThrough their stories, the HCPs emphasized the need for advance care plans when the risk of severe incidents increased. The advantages of such plans are to secure continuity and ensure that all important information is available for all the HCPs involved in a possible emergency situation. However, these plans should also be made for the family members so that they know how to act if critical events occur. In several stories, the HCPs described how the care plans included strategies and measures for staying at home as much as possible. One example was given of how specialist care and primary care cooperated, where the child was provided with advanced home care, and the child and the family shifted between being at the hospital and being at home. The family in this story expressed a strong wish to let the child die at home, and the HCPs made great efforts to accommodate this wish. The participant described how all the necessary equipment was installed in the family’s house:It was arranged for him to stay at home. He received pain relief, and I believe the community nurses cared for him, and he spent his last 2–3 weeks in the living room together with his siblings and parents and they all slept together in the same bed. They were kind of… they were together.” (Group 1)\nIt was arranged for him to stay at home. He received pain relief, and I believe the community nurses cared for him, and he spent his last 2–3 weeks in the living room together with his siblings and parents and they all slept together in the same bed. They were kind of… they were together.” (Group 1)\nMaking practical arrangements to facilitate a proper farewell in a safe environment together was a prioritized task. The parents and siblings could focus on being there for the child and for each other while the medical team provided care and pain relief for the child.\n[SUBTITLE] Experiencing dilemmas and distress [SUBSECTION] While many stories and examples evolved from cases where the HCPs explained that they managed to provide support and good care, there were also stories about conflicts with parents and distressful experiences. Some HCPs referred to conversations about how to handle a dramatic worsening of the child’s condition (e.g. decisions about not putting the child on a ventilator or stopping medications) and how this had provoked anger and frustration among parents. One HCP described one such situation:... and then there were discussions about withdrawing antibiotics, and we had to tell the parents, and one of them was really devastated and said, ‘You’re giving up on my child!’ whereupon the doctor kindly responded, ‘No, we’re not, we are comforting her during her last days’”. (Group 2)\n... and then there were discussions about withdrawing antibiotics, and we had to tell the parents, and one of them was really devastated and said, ‘You’re giving up on my child!’ whereupon the doctor kindly responded, ‘No, we’re not, we are comforting her during her last days’”. (Group 2)\nIn some cases, the participants noted that it was the HCPs and not the parents who pushed too hard by suggesting a new treatment, which turned out to result in complications or negative side effects. Another story illustrated the tension that can occur between HCPs and parents when the child is expected to die within a short time. In this case, one HCP accused a parent of hastening the baby’s death by withholding nutrition. This resulted in a deep conflict among the staff, where some supported the nurse and some sympathized with the parents. Eventually, external counselling was required to resolve the dispute among the staff. After the baby died, the parents reported that the whole situation had added even more to their grief. The participant described this as an ethically demanding situation, and, in retrospect, she was struck by how good intentions can lead to conflict and how emotionally involved one may become.\nExamples were also given of parents who refused to acknowledge that their child had a life-limiting disease and instead of planning for the end spent all their time searching for a cure. The HCPs experienced it as particularly hard to witness how false hope was being sold through experimental treatment and that parents were willing to squander that precious, limited time left of their child’s life:There was a family that I was never in a position to speak with, because their attention was always directed at something else. It was impossible to talk to them about anything, because they were always focusing on going off somewhere to find a cure. (Group 2)\nThere was a family that I was never in a position to speak with, because their attention was always directed at something else. It was impossible to talk to them about anything, because they were always focusing on going off somewhere to find a cure. (Group 2)\nIn these stories, the parents searched for opportunities to travel far with their child to try experimental treatment, resulting in long periods away from home and separation between family members. The participant found it challenging that the parents were never available to make plans for good palliative care.\nWhile many stories and examples evolved from cases where the HCPs explained that they managed to provide support and good care, there were also stories about conflicts with parents and distressful experiences. Some HCPs referred to conversations about how to handle a dramatic worsening of the child’s condition (e.g. decisions about not putting the child on a ventilator or stopping medications) and how this had provoked anger and frustration among parents. One HCP described one such situation:... and then there were discussions about withdrawing antibiotics, and we had to tell the parents, and one of them was really devastated and said, ‘You’re giving up on my child!’ whereupon the doctor kindly responded, ‘No, we’re not, we are comforting her during her last days’”. (Group 2)\n... and then there were discussions about withdrawing antibiotics, and we had to tell the parents, and one of them was really devastated and said, ‘You’re giving up on my child!’ whereupon the doctor kindly responded, ‘No, we’re not, we are comforting her during her last days’”. (Group 2)\nIn some cases, the participants noted that it was the HCPs and not the parents who pushed too hard by suggesting a new treatment, which turned out to result in complications or negative side effects. Another story illustrated the tension that can occur between HCPs and parents when the child is expected to die within a short time. In this case, one HCP accused a parent of hastening the baby’s death by withholding nutrition. This resulted in a deep conflict among the staff, where some supported the nurse and some sympathized with the parents. Eventually, external counselling was required to resolve the dispute among the staff. After the baby died, the parents reported that the whole situation had added even more to their grief. The participant described this as an ethically demanding situation, and, in retrospect, she was struck by how good intentions can lead to conflict and how emotionally involved one may become.\nExamples were also given of parents who refused to acknowledge that their child had a life-limiting disease and instead of planning for the end spent all their time searching for a cure. The HCPs experienced it as particularly hard to witness how false hope was being sold through experimental treatment and that parents were willing to squander that precious, limited time left of their child’s life:There was a family that I was never in a position to speak with, because their attention was always directed at something else. It was impossible to talk to them about anything, because they were always focusing on going off somewhere to find a cure. (Group 2)\nThere was a family that I was never in a position to speak with, because their attention was always directed at something else. It was impossible to talk to them about anything, because they were always focusing on going off somewhere to find a cure. (Group 2)\nIn these stories, the parents searched for opportunities to travel far with their child to try experimental treatment, resulting in long periods away from home and separation between family members. The participant found it challenging that the parents were never available to make plans for good palliative care.", "The HCPs seemed attentive to having the child with a life-threatening or life-limiting illness at the centre of their care. Several participants spoke about the importance of supporting a sense of normality and mentioned that the parents as well as the child aimed for ordinary days at home, school or kindergarten: “I believe that most parents want their child’s days to be normal and good until the end and that it’s not going to be something else”. (Group 3)\nStories were told that illustrate how the HCPs managed to create spaces for play and fun for the child despite working in a hospital and being responsible for clinical procedures. They gave examples of how they used medical equipment as props for play and how these playful situations opened up conversations with the child about sensitive and important topics. In one story, the nurse explained that they made the hospital stay so engaging that the child looked forward to treatment and felt sorry for leaving. Another nurse elaborated on a playful moment with a little girl with a life-limiting illness engaging in imaginary play:…and then the girl says: “At my prom, I want a dress like that”, and I said “Really? Wow, that’s cool”, and then she kind of realizes that she’s never going to a prom and then she says: “Well, well…it would have been cool, right?” and I responded “Yeah, it would have been really cool” (Group 2).\n…and then the girl says: “At my prom, I want a dress like that”, and I said “Really? Wow, that’s cool”, and then she kind of realizes that she’s never going to a prom and then she says: “Well, well…it would have been cool, right?” and I responded “Yeah, it would have been really cool” (Group 2).\nThe nurse reflected on the importance of being present in the moment and allowing the child to enjoy the thought of adolescence even though her life expectancy was very limited.", "The participants spoke about different ways of providing support through adjusted information and tailored care. In their stories, they emphasized the importance of sufficient, timely information. One participant described how a mother once said that she did not understand that her child could die while being on a ventilator and that the lungs could still collapse. The participant had carried this mother’s utterance with her ever since, and she pointed out that making sure that parents understand why their child actually dies is significant. Another participant described how important it is to be able to recognize when the parents communicate a need for more knowledge and to provide accurate information when it is called for. However, the participants also referred to challenging situations in which the parents and HCPs were not in step with each other. One participant admitted that if HCPs believe the parents are informed about the child’s situation and suddenly realize that they are not, this may cause delays in procedures and irritation:I try to be observant to this, both in myself and others. It’s important to avoid a moralizing attitude: ‘Do they [the parents] still not understand?’ Because it creates hassle and annoyance when the parents are not sufficiently informed and up to date on the child’s situation. (Group 2)\nI try to be observant to this, both in myself and others. It’s important to avoid a moralizing attitude: ‘Do they [the parents] still not understand?’ Because it creates hassle and annoyance when the parents are not sufficiently informed and up to date on the child’s situation. (Group 2)\nThe participants gave several examples of how they adjust and tailor their care for families to make them feel safe in acute as well as more stable situations. They spoke about the importance of working as a team around the family. One participant described it as creating layers upon layers of love around the child. This expression derived from a particular story about a child who was going to be placed in foster care but was too sick to leave the hospital. With help from the HCP, the child’s birth mother and foster mother moved into the children’s ward. With support from the multidisciplinary team, they cared for the child together during the last weeks of the child’s life.…and the foster mother stepped aside to let the birth mother hold the child close, day and night (…) and the baby received so many layers of love (…) And the HCPs were amazingly professional and put things aside to let it happen. (Group 2)\n…and the foster mother stepped aside to let the birth mother hold the child close, day and night (…) and the baby received so many layers of love (…) And the HCPs were amazingly professional and put things aside to let it happen. (Group 2)\nLater, the birth mother explained that, despite the great tragedy, these had been the finest weeks of her life. This situation was resource-demanding for the unit, but all the HCPs stood out as highly professional and were willing to make an extra effort in the extraordinary situation.\nGoing that extra mile for children and their families was a recurring theme in the HCPs’ stories. Witnessing severe and distressing situations seemed to make the HCPs mobilize to make the days, weeks and months as good as possible for the child, their parents and their siblings. Many of the participants’ stories were about creating safe spaces for families at the hospital, giving the child, parents and siblings moments to breathe. There were also examples of systems failing to support the families adequately and lack of coordination resulting in disorganized care; however, most stories were examples of how HCPs managed to work together, emphasizing the importance of shared information. One participant said:One of the finest moments that I’ve witnessed was when a nurse who knew the family well took the initiative to play with an older sibling of a child who would soon die. The nurse took the sibling outside in the hallway, and they talked and played it out. They had a good relationship. The nurse knew very well what was about to happen because she had been in the room when the message about the child’s impending death was given. The more people working around the family, the more important it is that we have the same information and work as a team. (Group 4)\nOne of the finest moments that I’ve witnessed was when a nurse who knew the family well took the initiative to play with an older sibling of a child who would soon die. The nurse took the sibling outside in the hallway, and they talked and played it out. They had a good relationship. The nurse knew very well what was about to happen because she had been in the room when the message about the child’s impending death was given. The more people working around the family, the more important it is that we have the same information and work as a team. (Group 4)", "Most of the stories emerged from critical situations or when the child’s illness was worsening, especially about initiating advance care planning with the end of the child’s life in mind. Some described the importance of seizing that particular moment when the parents and the child are ready and receptive to these difficult conversations. The HCPs gave examples of when the parents themselves took the initiative, but in most of the stories, the HCPs took the lead. However, as underlined by one participant, engaging in such dialogue requires that the HCPs are present and that they tread carefully: “If you’ve established a good relationship, moments will occur if you’re observant and willing to enter these conversations. For me, this is part of palliative care”. (Group 2)\nWith the children, such conversations could take place during play, and difficult topics could be touched upon without needing to make eye contact. It became evident through the HCPs’ stories that these conversations were tough and that it was sometimes difficult to control their own feelings.These conversations are really hard, it is as if you are about to start crying yourself. But I think it is really, really important [to talk about it], because we have reached the moment when it might happen. (Group 3)\nThese conversations are really hard, it is as if you are about to start crying yourself. But I think it is really, really important [to talk about it], because we have reached the moment when it might happen. (Group 3)\nThrough their stories, the HCPs emphasized the need for advance care plans when the risk of severe incidents increased. The advantages of such plans are to secure continuity and ensure that all important information is available for all the HCPs involved in a possible emergency situation. However, these plans should also be made for the family members so that they know how to act if critical events occur. In several stories, the HCPs described how the care plans included strategies and measures for staying at home as much as possible. One example was given of how specialist care and primary care cooperated, where the child was provided with advanced home care, and the child and the family shifted between being at the hospital and being at home. The family in this story expressed a strong wish to let the child die at home, and the HCPs made great efforts to accommodate this wish. The participant described how all the necessary equipment was installed in the family’s house:It was arranged for him to stay at home. He received pain relief, and I believe the community nurses cared for him, and he spent his last 2–3 weeks in the living room together with his siblings and parents and they all slept together in the same bed. They were kind of… they were together.” (Group 1)\nIt was arranged for him to stay at home. He received pain relief, and I believe the community nurses cared for him, and he spent his last 2–3 weeks in the living room together with his siblings and parents and they all slept together in the same bed. They were kind of… they were together.” (Group 1)\nMaking practical arrangements to facilitate a proper farewell in a safe environment together was a prioritized task. The parents and siblings could focus on being there for the child and for each other while the medical team provided care and pain relief for the child.", "While many stories and examples evolved from cases where the HCPs explained that they managed to provide support and good care, there were also stories about conflicts with parents and distressful experiences. Some HCPs referred to conversations about how to handle a dramatic worsening of the child’s condition (e.g. decisions about not putting the child on a ventilator or stopping medications) and how this had provoked anger and frustration among parents. One HCP described one such situation:... and then there were discussions about withdrawing antibiotics, and we had to tell the parents, and one of them was really devastated and said, ‘You’re giving up on my child!’ whereupon the doctor kindly responded, ‘No, we’re not, we are comforting her during her last days’”. (Group 2)\n... and then there were discussions about withdrawing antibiotics, and we had to tell the parents, and one of them was really devastated and said, ‘You’re giving up on my child!’ whereupon the doctor kindly responded, ‘No, we’re not, we are comforting her during her last days’”. (Group 2)\nIn some cases, the participants noted that it was the HCPs and not the parents who pushed too hard by suggesting a new treatment, which turned out to result in complications or negative side effects. Another story illustrated the tension that can occur between HCPs and parents when the child is expected to die within a short time. In this case, one HCP accused a parent of hastening the baby’s death by withholding nutrition. This resulted in a deep conflict among the staff, where some supported the nurse and some sympathized with the parents. Eventually, external counselling was required to resolve the dispute among the staff. After the baby died, the parents reported that the whole situation had added even more to their grief. The participant described this as an ethically demanding situation, and, in retrospect, she was struck by how good intentions can lead to conflict and how emotionally involved one may become.\nExamples were also given of parents who refused to acknowledge that their child had a life-limiting disease and instead of planning for the end spent all their time searching for a cure. The HCPs experienced it as particularly hard to witness how false hope was being sold through experimental treatment and that parents were willing to squander that precious, limited time left of their child’s life:There was a family that I was never in a position to speak with, because their attention was always directed at something else. It was impossible to talk to them about anything, because they were always focusing on going off somewhere to find a cure. (Group 2)\nThere was a family that I was never in a position to speak with, because their attention was always directed at something else. It was impossible to talk to them about anything, because they were always focusing on going off somewhere to find a cure. (Group 2)\nIn these stories, the parents searched for opportunities to travel far with their child to try experimental treatment, resulting in long periods away from home and separation between family members. The participant found it challenging that the parents were never available to make plans for good palliative care.", "As a means of enhancing our knowledge of HCPs’ understanding of what PPC entails, the present study aimed to explore HCPs’ stories about PPC. Our findings are in line with the findings of previous studies that HCPs associate PPC with advanced illness or more acute phases towards the end of the child’s illness trajectory [8, 10, 11]. The HCPs’ stories circled around family care, particularly relating to dramatic or affective situations when the death of a child was imminent. The stories reflected how the HCPs view PPC as a specific field of health care that requires particular professional sensitivity, including good communication, collaboration and planning. Thus, the HCPs in this study demonstrated knowledge about the core qualities needed to succeed in PPC [17].\nThe potentially complicated illness trajectories in children with life-limiting and life-threatening conditions make early integration of palliative care particularly beneficial [3, 4]. The stories in this study rarely touched upon topics related to earlier palliative phases like living everyday life with a life-limiting disease or coordinating follow-up at home or school. Possible consequences for late referral to palliative care are that symptoms may be unaddressed for longer and psychological needs may be neglected [18]. Bearing in mind that the participants in the present study all worked in specialist healthcare, it is reasonable that most stories revolved around acute and life-threatening situations at a hospital. However, according to the WHO’s guidelines and the guidelines for PPC in Norway, specialist health care and primary health care services should be involved in the entire process from diagnosis onwards, assuring overall and coordinated follow-up of the child and the family [3, 4]. The national guidelines emphasize the importance of continuous mapping, evaluation and adjustment to the family’s needs and that this is a shared responsibility between primary and specialist health care [3]. Over the last years interdisciplinary PPC-teams have been established in children’s wards throughout the country, aiming at provision of care regardless of whether the child is at home, in institutions or in hospitals. In some of the larger cities in Norway, the ‘Hospital at home’ provides specialized health care in the child’s home. Many families express a strong wish for home care [19]. Only a small number of the stories included examples of cooperation with community care, enabling the families to stay at home. This indicates that the HCPs either had few such experiences of collaboration outside the hospital or that the collaboration they engaged in did not fall within their conception of PPC and that they did not regard such work as palliative care. This demonstrates a need for clarification of the distinction between basic and specialized PPC. Basic PPC is to some degree integrated into standard care of children with life-limiting and life-threatening conditions in Norway, but specialized PPC is a relatively new field of medical expertise that was brought to public attention with the first national guidelines in 2016 [3].\nWhile few stories in this study exemplified how HCPs work cooperatively outside the hospital, several stories described the necessity and value of working as a team within the hospital. PPC is defined by the WHO as total care of the child’s body, mind and spirit, which also includes care of the family [4]. Thus, interdisciplinary teamwork is essential for the provision of PPC. To evaluate and alleviate the distress of the child and the family and promote their well-being throughout the illness trajectory, multiprofessional teams need to work in an interdisciplinary way. Some participants emphasized the importance of equal access to information and shared understanding of a child’s and family’s situation within the health care team. Although each team member has a particular scope of practice and responsibility, a mutual understanding based on shared experiences and information enables mutual decision-making [20], which can be particularly important in acute or stressful situations. An interdisciplinary team approach, which includes the child and/or the parents, is essential for achieving holistic and comprehensive care encompassing the complex needs of the patient and the family [21]. One study suggests that HCPs in palliative care teams enhance their ability to see the patient from a holistic perspective by sharing impressions and discussing the patient and family’s situation within the team [22].\nEven though the HPCs’ stories were not about early PPC stages, they do illustrate important elements of later phases, as described in national and international guidelines [3, 4]. Many of the stories accompanied the categories identified in our previous paper relating to family-centred care and preparing for the death of a child [11]. The stories show that the participants recognize the importance of having a family-centred care perspective. The HCPs’ attention is very much drawn to how they can best support the family. In palliative care, family-centred care is defined as seamless continuity addressing patient, family and community needs related to terminal conditions through interdisciplinary collaboration [23]. Family-centred care has been described as containing the principles of information sharing, respecting and honouring differences, partnership and collaboration, negotiation and care in the context of family and community [24]. A child’s and family’s needs and preferences vary. Thus, it is not possible to deliver family-centred PPC with a one-size-fits-all approach. Among the stories told were examples of how HCPs stood out as creative, flexible and responsive to the child’s or the parents’ specific needs through adjusting their information and care for the child and the family. Meanwhile, the participants also emphasized the need for structured advance care planning, particularly when death is approaching. Engaging in advance care planning requires that future scenarios be discussed with families [25]. Coming to a realization that death is inevitable and close is a process for those involved, and families as well as HCPs reach this understanding at different times and paces [26, 27]. Several stories evinced that having these conversations is demanding for HCPs, that timing is key and that insufficient communication may have severe consequences for the parties involved. Conversations about future care and end-of-life planning were mostly initiated by HCPs and less often by the parents in the stories we collected, which corresponds with previous research findings [26]. Several frameworks and tools have been developed to address advance care and end-of-life planning, some of which are specifically developed for PPC [28]. However, no such tools were mentioned by the participants. Instead, their stories indicated that they relied on their experience and professional tact when engaging in dialogues about palliative care. This may reflect the status and development of PPC in Norway at the time of this study. Advance care planning as a specific method has just recently been integrated into the guidelines of the Norwegian Association of Paediatricians [29].\nThe overall aim of PPC is to ensure a child’s quality of life throughout the illness trajectory [4]. Cherishing moments of joy and aiming for days of normality for the children were spoken of as important by the participants. The support of HCPs in achieving this, has been shown to be of great importance to parents as well [30]. For children, normality equals playing. The participants’ stories of how they provided children at the hospital with time and opportunities to play illustrate that the participants recognize its value. Playing moves the child out of time, allowing them to be absorbed in the moment and making room for absolute presence while at the same time giving the child an increased understanding of their existence [31]. This was beautifully illustrated in the story about the girl who knew she would not live to experience her prom but still allowed herself to joyfully engage in play about it. As death approaches, time increases in value. Witnessing how children capture time in play may help parents as well as HCPs broaden their understanding of what PPC comprises.", "Using stories as a basis for analysis is a strength of the present study. When we tell stories we use language available and familiar to us and to our audience [1]. While HCPs’ understanding and experience of PPC has been investigated through qualitative interview studies, few, if any, have used storytelling as a research tool to explore their insights. Compared to other qualitative research methods, stories can generate more nuanced, contextualized and culturally reflective information [32]. However, participating in focus groups instead of individual interviews may have limited the HCPs’ time and opportunity to elaborate on their stories. In only one of the four focus group interviews did the interviewer explicitly ask the participants to tell a story or give an example from practice. This is an important limitation. If the same request had been given to the remaining groups, we might have collected richer data. Nevertheless, the participants in all the focus groups spontaneously used their experiences to elaborate on their comprehension of PPC through stories and examples, with little prompting required from the interviewers. Some chose to supplement their colleagues’ stories or follow up with their own version of the same case, while at other times listening to a story served as a cue for a new story mirroring or contrasting with the former. The stories stood out as significant to the storyteller, and it was noticeable that they had been told and reflected over before. This underlines the significant potential of stories to capture the participants’ lived experiences. One limitation to be noted is that the sample was predominantly female and thus the results may be less representative for male HCPs.", "In this study, we aimed to explore stories of PPC as narrated by HCPs to enhance our knowledge of their understanding of what PPC is. In our previous paper, we discussed that although the HCPs were hesitant to use the term PPC, they found themselves responsible for delivering PPC [11]. The HCPs’ stories supported this by highlighting key elements of PPC, such as interdisciplinary teamwork family-centred care and advance care planning. However, similar to findings in our first paper [11] and previous research [8–10], the stories illustrated that HCPs’ reflections about PPC did not capture the entire concept of PPC, as they were strongly associated with end-of-life care. This underscores the importance of increasing knowledge about PPC among HCPs in Norway. A consolidated recognition of the meaning and content of PPC is necessary to maintain quality of life for all children living with life-limiting or life-threatening conditions." ]
[ null, null, null, null, null, null, null, "results", null, null, null, null, "discussion", null, "conclusion" ]
[ "Paediatric palliative care", "Palliative care", "Health care professionals", "Life-threatening conditions", "Life-limiting conditions", "Concept", "Storytelling", "Thematic analysis" ]
Size selection and placement of pedicle screws using robot-assisted versus fluoroscopy-guided techniques for thoracolumbar fractures: possible implications for the screw loosening rate.
36273170
There has been increased development of robotic technologies for the accuracy of percutaneous pedicle screw placement. However, it remains unclear whether the robot really optimize the selection of screw sizes and enhance screw stability. The purpose of this study is to compare the sizes (diameter and length), placement accuracy and the loosening rate of pedicle screws using robotic-assisted versus conventional fluoroscopy approaches for thoracolumbar fractures.
BACKGROUND
A retrospective cohort study was conducted to evaluate 70 consecutive patients [34 cases of robot-assisted percutaneous pedicle screw fixation (RAF) and 36 of conventional fluoroscopy-guided percutaneous pedicle screw fixation (FGF)]. Demographics, clinical characteristics, and radiological features were recorded. Pedicle screw length, diameter, and pedicle screw placement accuracy were assessed. The patients' sagittal kyphosis Cobb angles (KCA), anterior vertebral height ratios (VHA), and screw loosening rate were evaluated by radiographic data 1 year after surgery.
METHODS
There was no significant difference in the mean computed tomography (CT) Hounsfield unit (HU) values, operation duration, or length of hospital stay between the groups. Compared with the FGF group, the RAF group had a lower fluoroscopy frequency [14 (12-18) vs. 21 (16-25), P < 0.001] and a higher "grade A + B" pedicle screw placement rate (96.5% vs. 89.4%, P < 0.05). The mean screw diameter was 6.04 ± 0.55 mm in the RAF group and 5.78 ± 0.50 mm in the FGF group (P < 0.001). The mean screw length was 50.45 ± 4.37 mm in the RAF group and 48.63 ± 3.86 mm in the FGF group (P < 0.001). The correction loss of the KCA and VHR of the RAF group was less than that of the FGT group at the 1-year follow-up [(3.8 ± 1.8° vs. 4.9 ± 4.2°) and (5.5 ± 4.9% vs. 6.4 ± 5.7%)], and screw loosening occurred in 2 out of 34 patients (5.9%) in the RAF group, and 6 out of 36 patients (16.7%) in the FGF group, but there were no significant differences (P > 0.05).
RESULTS
Compared with the fluoroscopy-guided technique, robotic-assisted spine surgery decreased radiation exposure and optimizes screw trajectories and dimensions intraoperatively. Although not statistically significant, the loosening rate of the RAF group was lower that of than the FGT group.
CONCLUSION
[ "Humans", "Pedicle Screws", "Robotics", "Retrospective Studies", "Spinal Fusion", "Lumbar Vertebrae", "Fluoroscopy", "Fractures, Bone", "Kyphosis" ]
9587613
null
null
null
null
Results
Overall, 70 patients who underwent short-segment fixation were included in the study. Patient demographics and clinical characteristics were compared between the RAF and FGF groups. There were no significant differences between the groups in terms of age, sex, BMI, mean HU values, mechanism of injury, fracture level and type, and TLICS score. The baseline data of the two groups were comparable (Table 1). Table 1Baseline characteristics and perioperative parameters of all patientsCharacteristicRAFFGFStatistic (χ2/t) P valueNo. of pts3436––Mean age (years)42.8 ± 8.739.8 ± 10.71.2770.206Sex0.3700.558 Male2526–– Female910––Mean BMI (kg/m2)23.8 ± 2.624.0 ± 2.70.6990.718Mean HU values184.4 ± 66.5202.4 ± 80.4− 1.0260.311Mechanism of injury0.4750.830 Traffic accident2222 Fall1214Fracture level0.8260.972 T1000 T1134 T1288 L11816 L258Fracture type0.2140.543 A32521 A4915TLICS score4.5 ± 0.54.6 ± 0.5− 0.4480.656RAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation, BMI body mass index, HU Hounsfeld Unit, TLICS Score Thoracolumbar Injury Classification and Severity Score Baseline characteristics and perioperative parameters of all patients RAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation, BMI body mass index, HU Hounsfeld Unit, TLICS Score Thoracolumbar Injury Classification and Severity Score
Conclusion
Compared with fluoroscopy-guided techniques, robotic assistance can help a surgeon to optimize screw trajectories and dimensions intraoperatively. Although not statistically significant, these findings are a possible way to increase bone purchase and reduce the screw loosening rate for thoracolumbar fractures. In addition, with increased experience and proficiency in robot-assisted techniques, surgeons should be more confident in their screw selection, especially when using sizes that they may have previously considered too large.
[ "Background", "Patient selection", "Surgical techniques", "RAF group", "FGF group", "Data collection", "Radiographic measurements", "Statistical analysis", "Clinical evaluation", "Pedicle screw dimensions", "Radiographic evaluation", "Limitations" ]
[ "Thoracolumbar fractures account for 30–60% of spinal fractures and are commonly seen in high-energy trauma patients [1, 2]. Burst fractures (AO spine types A3 and A4) represent the majority of thoracolumbar fractures [3]. Timely diagnosis and surgical stabilization are essential to prevent further neurological deficits and to reconstruct the sagittal balance, which can help avoid long-term complications [4–6]. Percutaneous pedicle screw fixation is the most commonly performed surgical procedure for the treatment of thoracolumbar spine fractures without neurological deficits.\nIn general, misplacement and inappropriate choice of pedicle screws negatively affect the internal fixation stability [7, 8]. Inserting the longer and larger screw accurately optimizes fixation strength [8, 9]. However, the benefits of larger screws must be weighed against the risks of pedicle breach or neurovascular injury [10, 11]. Therefore, the key to this technique lies in the determination of an appropriate-sized screw for maximum fixation strength while simultaneously respecting the structural integrity of the vertebral pedicles.\nSome studies have reported that robot-assisted techniques provide more precise screw placement [12–14], and more recent literature shows that robotic assistance allows for the placement of screws with greater screw diameter and length compared with surgical navigation alone in minimally invasive lumbar fusion procedures [15]. However, to the best of our knowledge, few studies have compared the dimension selection and loosening rate of pedicle screws between robot-assisted techniques and fluoroscopy-guided percutaneous techniques for thoracolumbar fractures. In the present study, we compared radiological and clinical parameters from the immediate postoperative time to the 1-year follow-up between patients who underwent robot-assisted and conventional fluoroscopy surgical procedures.", "This was a retrospective cohort study. A consecutive sample of 70 patients with A3 and A4 thoracolumbar fractures from May 2018 to October 2020 was enrolled. Of the 70 patients reviewed, 34 and 36 were treated with robotic-assisted percutaneous pedicle screw fixation and fluoroscopy-guided percutaneous pedicle screw fixation, respectively, based on the following inclusion criteria: (1) thoracolumbar fracture classified as A3 or A4; (2) short-segment fixation with two additional screws at the single fracture vertebra; (3) no other fractures or organ injuries; and (4) absence of neurological deficits. The exclusion criteria were as follows: (1) incomplete clinical information; (2) combined anterior-posterior surgeries; (3) revision surgeries; (4) fracture displacement in the vertebral pedicle; and (5) structural spinal deformity, spinal tumour, or infection. Written informed consent was obtained from all patients. The present study was reviewed and approved by the Ethics Committee and Institutional Review Board of the Second Affiliated Hospital of Soochow University.", "[SUBTITLE] RAF group [SUBSECTION] Patients were placed in the prone position after general anaesthesia. The CT scan of the surgical region was uploaded to the workstation (Mazor Robotics Company, Israel) before surgery, and the perfect trajectories of the vertebrae based on the surgeon’s requests were planned (Fig. 1A–D). After the anteroposterior and oblique plane images were captured by the C-arm, the fluoroscopic images were automatically merged with the preoperative CT. A small (400 g, 9 cm tall, 5 cm diameter), parallel, 6-DOF robotic manipulator was then placed on the bone-mounted platform and aligned with a planned trajectory according to the surgeon’s instructions (Fig. 1E). After the paths of the screws were tapped with a thread tap through the dilated channels, the screws were inserted using handover guide wires (Fig. 1F). Rods were placed percutaneously from either the cephalad or caudal side with the assistance of screw extenders.\n\nFig. 1Perfect trajectories of the vertebrae based on the surgeon’s requests were planned (A–D). The manipulator was placed on the bone-mounted platform, and the appropriate pedicle screws were inserted (E, F)\nPerfect trajectories of the vertebrae based on the surgeon’s requests were planned (A–D). The manipulator was placed on the bone-mounted platform, and the appropriate pedicle screws were inserted (E, F)\nPatients were placed in the prone position after general anaesthesia. The CT scan of the surgical region was uploaded to the workstation (Mazor Robotics Company, Israel) before surgery, and the perfect trajectories of the vertebrae based on the surgeon’s requests were planned (Fig. 1A–D). After the anteroposterior and oblique plane images were captured by the C-arm, the fluoroscopic images were automatically merged with the preoperative CT. A small (400 g, 9 cm tall, 5 cm diameter), parallel, 6-DOF robotic manipulator was then placed on the bone-mounted platform and aligned with a planned trajectory according to the surgeon’s instructions (Fig. 1E). After the paths of the screws were tapped with a thread tap through the dilated channels, the screws were inserted using handover guide wires (Fig. 1F). Rods were placed percutaneously from either the cephalad or caudal side with the assistance of screw extenders.\n\nFig. 1Perfect trajectories of the vertebrae based on the surgeon’s requests were planned (A–D). The manipulator was placed on the bone-mounted platform, and the appropriate pedicle screws were inserted (E, F)\nPerfect trajectories of the vertebrae based on the surgeon’s requests were planned (A–D). The manipulator was placed on the bone-mounted platform, and the appropriate pedicle screws were inserted (E, F)", "Patients were placed in the prone position after general anaesthesia. The CT scan of the surgical region was uploaded to the workstation (Mazor Robotics Company, Israel) before surgery, and the perfect trajectories of the vertebrae based on the surgeon’s requests were planned (Fig. 1A–D). After the anteroposterior and oblique plane images were captured by the C-arm, the fluoroscopic images were automatically merged with the preoperative CT. A small (400 g, 9 cm tall, 5 cm diameter), parallel, 6-DOF robotic manipulator was then placed on the bone-mounted platform and aligned with a planned trajectory according to the surgeon’s instructions (Fig. 1E). After the paths of the screws were tapped with a thread tap through the dilated channels, the screws were inserted using handover guide wires (Fig. 1F). Rods were placed percutaneously from either the cephalad or caudal side with the assistance of screw extenders.\n\nFig. 1Perfect trajectories of the vertebrae based on the surgeon’s requests were planned (A–D). The manipulator was placed on the bone-mounted platform, and the appropriate pedicle screws were inserted (E, F)\nPerfect trajectories of the vertebrae based on the surgeon’s requests were planned (A–D). The manipulator was placed on the bone-mounted platform, and the appropriate pedicle screws were inserted (E, F)", "Patients were placed in the prone position after general anaesthesia. The fractured vertebrae were confirmed by fluoroscopy. Six transpedicular puncture needles were inserted through six small incisions using fluoroscopy, after which guide wires were inserted. The appropriate pedicle screws were then placed through the guide wires. The position of each screw was examined by fluoroscopy, followed by the insertion of titanium rods with the assistance of screw extenders.", "Patient demographics and clinical characteristics were recorded. The mean CT Hounsfield Unit (HU) values were obtained from the measurement of preoperative CT measurements (Fig. 2). The duration of the operation, number of radiation cycles, amount of intraoperative blood loss, length of hospital stay, and cost were compared between the two groups. The surgical data obtained included the levels of instrument, and pedicle screw length, and diameter. Screw dimensions were obtained from the patient’s electronic health records, to which all operative implant records were uploaded. Considering that short-length pedicle screws were used for the fractured vertebral body, only screws in the vertebra above and below the fractured vertebral body were counted.\n\nFig. 2Measurement methods for mean CT Hounsfield Unit (HU) values: a 41-year-old male patient was diagnosed with a vertebral fracture in L1. A The mean CT Hounsfield Unit (HU) values of the T12 vertebral body (VB) were 176, and the right and left pedicles (excluding the cortical bone) were 133 and 136 respectively; B the mean HU values of the right and left pedicles of the L1 (fractured vertebra) were 157 and 144 respectively; C the mean HU values of the L2 VB was 164, and the right and left pedicle were 160 and 147 respectively. The mean HU values of the patient was 152.1 = [(176 + 133 + 136 + 157 + 144 + 164 + 160 + 147)/8]\nMeasurement methods for mean CT Hounsfield Unit (HU) values: a 41-year-old male patient was diagnosed with a vertebral fracture in L1. A The mean CT Hounsfield Unit (HU) values of the T12 vertebral body (VB) were 176, and the right and left pedicles (excluding the cortical bone) were 133 and 136 respectively; B the mean HU values of the right and left pedicles of the L1 (fractured vertebra) were 157 and 144 respectively; C the mean HU values of the L2 VB was 164, and the right and left pedicle were 160 and 147 respectively. The mean HU values of the patient was 152.1 = [(176 + 133 + 136 + 157 + 144 + 164 + 160 + 147)/8]", "Two spine surgeons with more than 10 years of experience completed the measurements independently. The average value of the measured data was the final data. All data were measured using radiology software (Neusoft PACS/RIS) to reduce variability.\nBased on the Gertzbein and Robbins scale [16], the accuracy of the pedicle screw placement was evaluated on postoperative CT images as follows (Fig. 3A–E): Grade A: screw completely within bone; Grade B: cortical breach of < 2 mm; Grade C: cortical breach of ≥ 2 mm and < 4 mm; Grade D: cortical breach of ≥ 4 mm and < 6 mm; and Grade E: cortical breach of ≥ 6 mm.\n\nFig. 3Grading used for pedicle perforation on the axial CT scan and the representative images: A both pedicle screws are completely within the pedicle (Grade A); B Grade B (< 2 mm) perforation of the lateral wall of the right pedicle and Grade A of the left pedicle; C Grade C (2–4 mm) perforation of the medial wall of the left pedicle and Grade A perforation of the right pedicle; D Grade D (> 4 mm) perforation of the lateral wall of both pedicles; E Grade E (> 6 mm) perforation of the medial wall of the right pedicle. The blue lines represent the medial or lateral wall of the vertebral pedicles; the red lines represent the edge of the pedicle screws; and the green lines represent the distance of the perforation\nGrading used for pedicle perforation on the axial CT scan and the representative images: A both pedicle screws are completely within the pedicle (Grade A); B Grade B (< 2 mm) perforation of the lateral wall of the right pedicle and Grade A of the left pedicle; C Grade C (2–4 mm) perforation of the medial wall of the left pedicle and Grade A perforation of the right pedicle; D Grade D (> 4 mm) perforation of the lateral wall of both pedicles; E Grade E (> 6 mm) perforation of the medial wall of the right pedicle. The blue lines represent the medial or lateral wall of the vertebral pedicles; the red lines represent the edge of the pedicle screws; and the green lines represent the distance of the perforation\nThe pre- and postoperative sagittal kyphosis Cobb angles (defined by the upper endplate of the first vertebra above the fractured vertebra and by the lower endplate of the first vertebra below the fractured vertebra) and anterior vertebral height ratios (defined by the percentage of the mean values for the adjacent vertebrae) of the fractured vertebra were measured using radiographic images (Fig. 4).\n\nFig. 4Schematic drawings showing the measurement methods for the sagittal kyphosis Cobb angles (KCA) and anterior vertebral height ratios (VHR). A The KCA was defined as the angle between the superior endplate of the upper vertebra(superior black line) and the inferior endplate of the lower vertebra (inferior black line) in accordance with Cobb’s method; B VHR = 2b/(a + c)\nSchematic drawings showing the measurement methods for the sagittal kyphosis Cobb angles (KCA) and anterior vertebral height ratios (VHR). A The KCA was defined as the angle between the superior endplate of the upper vertebra(superior black line) and the inferior endplate of the lower vertebra (inferior black line) in accordance with Cobb’s method; B VHR = 2b/(a + c)\nPedicle screw loosening was defined as a “double-halo sign” on plain X-ray or a “lucent zone” around on CT images, which were obtained 1 year postoperatively (Fig. 5) [17, 18].\n\nFig. 5A Postoperative 1-year plain X-ray demonstrated left screw loosening at T12, L1, and L2 in a 39-year-old male who underwent short-segment fixation from T12 to L2 (black arrow: double halo sign); B postoperative 1-year CT demonstrated left screw loosening at L1 in a 47-year-old male who underwent short-segment fixation from T11 to L12 (white arrow: lucent zone)\nA Postoperative 1-year plain X-ray demonstrated left screw loosening at T12, L1, and L2 in a 39-year-old male who underwent short-segment fixation from T12 to L2 (black arrow: double halo sign); B postoperative 1-year CT demonstrated left screw loosening at L1 in a 47-year-old male who underwent short-segment fixation from T11 to L12 (white arrow: lucent zone)\n[SUBTITLE] Statistical analysis [SUBSECTION] Descriptive statistics are presented as the mean ± standard deviation for normally distributed continuous variables; the median and interquartile range was used for nonnormally distributed continuous variables and the count (frequency) was used for categorical variables. Comparisons of baseline characteristics, which were stratified by group, were performed using Student’s t tests for normally distributed continuous variables, Wilcoxon rank-sum tests for variables with skewed distributions, and chi-square tests for categorical data. A p value < 0.05 was considered statistically significant. All analyses were performed using SPSS for Mac (IBM SPSS Statistics 23.0, SPSS Inc., Chicago, IL, USA).\nDescriptive statistics are presented as the mean ± standard deviation for normally distributed continuous variables; the median and interquartile range was used for nonnormally distributed continuous variables and the count (frequency) was used for categorical variables. Comparisons of baseline characteristics, which were stratified by group, were performed using Student’s t tests for normally distributed continuous variables, Wilcoxon rank-sum tests for variables with skewed distributions, and chi-square tests for categorical data. A p value < 0.05 was considered statistically significant. All analyses were performed using SPSS for Mac (IBM SPSS Statistics 23.0, SPSS Inc., Chicago, IL, USA).", "Descriptive statistics are presented as the mean ± standard deviation for normally distributed continuous variables; the median and interquartile range was used for nonnormally distributed continuous variables and the count (frequency) was used for categorical variables. Comparisons of baseline characteristics, which were stratified by group, were performed using Student’s t tests for normally distributed continuous variables, Wilcoxon rank-sum tests for variables with skewed distributions, and chi-square tests for categorical data. A p value < 0.05 was considered statistically significant. All analyses were performed using SPSS for Mac (IBM SPSS Statistics 23.0, SPSS Inc., Chicago, IL, USA).", "The RAF group had significantly fewer radiation cycles (P < 0.001) but higher hospitalization costs (P = 0.001) than the FGF group (Table 2). No significant difference was observed between the two groups in terms of surgical time, intraoperative blood loss, or length of hospital stay (Table 2).\n\nTable 2Summary of operation valuesOperative factorsRAFFGFStatistic (t/Z)\nP valueMean operation time (min)114.1 ± 34.4104.0 ± 18.11.5520.125Fluoroscopy frequency (times)16.1 ± 3.122.6 ± 3.9− 7.8150.000**Mean hospital stay (days)7.5 ± 2.48.2 ± 2.5− 1.2790.204Mean blood loss (mL)93.8 ± 24.9102.2 ± 27.8− 0.9630.339Hospitalization expenses (RMB)70529.6 ± 17224.558803.1 ± 9753.43.530.001**RAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation, RMB Renminbi**P < 0.01, statistically significant\nSummary of operation values\nRAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation, RMB Renminbi\n**P < 0.01, statistically significant", "There were 136 screws placed in the RAF group and 144 placed in the FGF group (screws in the vertebra above and below the fractured vertebra). Screws placed in the RAF group had both a larger screw diameter (RAF 6.04 ± 0.55 mm [range 5.0–7.0 mm]; FGF 5.78 ± 0.50 mm [range 5.0–6.5 mm]) and screw length (RAF 50.45 ± 4.37 mm [range 40–55 mm]; FGF 48.63 ± 3.86 mm [range 40–55 mm]) than screws placed in the FGF group (P < 0.001) (Table 3).\n\nTable 3Pedicle screw diameter and lengthVariableRAFFGFStatistic (χ2)\nP valueNo. of screws Total1361445.3090.379 T1026 T112220 T123634 L13238 L23428 L31018Mean screw diameter (range), mm Total6.04 (5.0–7.0)5.78 (5.0–6.5)− 3.0670.000** T105.5 (5.5)5.17 (5.0–5.5)–– T116.09 (5.5–7.0)5.90 (5.5–6.5)-0.9750.330 T126.11 (5.5–7.0)6.04 (5.5–6.5)-0.3320.740 L15.81 (5.0–6.5)5.29 (5.0–6.0)-4.2810.000** L26.12 (5.5–7.0)6.07 (5.5–6.5)-0.2380.812 L36.20 (6.0–6.5)5.94 (5.5–6.5)-2.1430.032*Mean screw length (range), mm Total50.45 (40–55)48.63 (40–55)-3.5610.000** T1050 (50)48.54 (45–50)–– T1150.00 (40–55)49.50 (40–55)− 0.3200.749 T1249.19 (45–55)47.35 (45–55)2.0640.039* L150.65 (40–55)48.96 (40–55)− 1.9660.049* L251.18 (45–55)49.34 (45–55)− 1.3910.164 L353.00 (50–55)48.33 (45–55)− 3.1440.020*RAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation\n*P < 0.05; **P < 0.01, statistically significant\nPedicle screw diameter and length\nRAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation\n\n*P < 0.05; **P < 0.01, statistically significant", "In the RAF group, 91.2% of the 204 screws were well placed (grade A); the remaining screws were graded B (5.4%), C (2.9%), or D (1.0%), but no screw was graded E. In the FGF group, 80.1% of the 216 screws were graded A; the remaining screws were graded B (9.3%), C (6.9%), D (2.8%), or E (0.9%). The RAF group had a significantly higher ratio of clinically acceptable screws (grades A and B, 96.6%) than the FGF group (89.4%) (P < 0.05) (Table 4).\n\nTable 4Pedicle screw accuracy rateVariableRAFFGFStatistic (χ2)\nP valueNo. of screws204216Screw accuracy rate11.7500.019* Grade A186 (91.2%)173 (80.1%) Grade B11 (5.4%)20 (9.3%) Grade A + B197 (96.6%)193 (89.4%) Grade C5 (2.4%)15 (6.9%) Grade D2 (1.0%)6 (2.8%) Grade E02 (0.9%) Grade C + D + E7 (3.4%)23 (10.6%)RAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation\n*P < 0.05 statistically significant\nPedicle screw accuracy rate\nRAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation\n\n*P < 0.05 statistically significant\nThe preoperative kyphosis Cobb angle (KCA) and anterior height ratio of fractured vertebra (VHR) were similar between the two groups. After surgery, the KCA was significantly corrected, and the VHR was well improved in both groups. The correction loss of the KCA (3.8 ± 1.8°) was less than that of the FGT group( 4.9 ± 4.2°), and the VHR of the RAF group (5.5 ± 4.9% ) was less than that of the FGT group (6.4 ± 5.7%) at the 1-year follow-up, but there were no significant differences (Table 5).\n\nTable 5Summary of radiographic measurementsVariableRAFFGFStatistic (χ2/Z/t)\nP value\nNo. of pts\n\n34\n\n36\n\n–\n\n–\n\nKyphosis Cobb angle (KCA) (°)\n Preoperative KCA20.2 ± 5.820.9 ± 4.3− 0.6130.542 Postoperative KCA8.2 ± 4.88.7 ± 3.9− 0.4940.621 Postoperative KCA correction12.0 ± 4.412.2 ± 3.4− 0.2560.798 KCA at 1-y FU11.9 ± 4.7− 9.3 ± 3.9− 1.3740.174 Correction loss at 1-y FU3.8 ± 1.84.9 ± 4.2− 1.4850.138\nAnterior height ratio of fracture vertebral (VHR) (%)\n Preoperative VHR62.0 ± 11.260.7 ± 14.30.4190.676 Postoperative VHR84.6 ± 9.982.5 ± 11.40.8120.420 Postoperative VHR Correction22.6 ± 12.121.9 ± 12.9− 0.1060.910 VHR at 1-y FU79.2 ± 7.976.1 ± 10.71.3560.180 Correction loss at 1-y FU5.5 ± 4.96.4 ± 5.7− 0.9750.329\nScrew loosening rate of pts (%)\n2 (5.9%)6 (16.7%)2.0090.156RAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation, KCA Kyphosis Cobb angle, AVHR anterior vertebral height ratios, 1-y FU 1-year follow-up, pts patient\nSummary of radiographic measurements\nRAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation, KCA Kyphosis Cobb angle, AVHR anterior vertebral height ratios, 1-y FU 1-year follow-up, pts patient\nScrew loosening occurred in 2 out of 34 patients (5.9%) in the RAF group, and 6 out of 36 patients (16.7%) in the FGF group. There was no statistically significant difference in terms of the screw loosening rate between the two groups (Table 5).", "Our study has several limitations. First, the sample size of fracture cases was relatively small, and all patients were recruited from a single centre. Future studies with larger sample sizes are needed to replicate the findings. Second, data on the trajectory angles, the radians and forces of spinal rods, and misaligned spinal rods, all of which can cause screw loosening, were not available. Finally, further experiments are necessary to design an adequate trajectory, which can significantly increase the stability of the pedicle screw." ]
[ null, null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Materials and methods", "Patient selection", "Surgical techniques", "RAF group", "FGF group", "Data collection", "Radiographic measurements", "Statistical analysis", "Results", "Clinical evaluation", "Pedicle screw dimensions", "Radiographic evaluation", "Discussion", "Limitations", "Conclusion" ]
[ "Thoracolumbar fractures account for 30–60% of spinal fractures and are commonly seen in high-energy trauma patients [1, 2]. Burst fractures (AO spine types A3 and A4) represent the majority of thoracolumbar fractures [3]. Timely diagnosis and surgical stabilization are essential to prevent further neurological deficits and to reconstruct the sagittal balance, which can help avoid long-term complications [4–6]. Percutaneous pedicle screw fixation is the most commonly performed surgical procedure for the treatment of thoracolumbar spine fractures without neurological deficits.\nIn general, misplacement and inappropriate choice of pedicle screws negatively affect the internal fixation stability [7, 8]. Inserting the longer and larger screw accurately optimizes fixation strength [8, 9]. However, the benefits of larger screws must be weighed against the risks of pedicle breach or neurovascular injury [10, 11]. Therefore, the key to this technique lies in the determination of an appropriate-sized screw for maximum fixation strength while simultaneously respecting the structural integrity of the vertebral pedicles.\nSome studies have reported that robot-assisted techniques provide more precise screw placement [12–14], and more recent literature shows that robotic assistance allows for the placement of screws with greater screw diameter and length compared with surgical navigation alone in minimally invasive lumbar fusion procedures [15]. However, to the best of our knowledge, few studies have compared the dimension selection and loosening rate of pedicle screws between robot-assisted techniques and fluoroscopy-guided percutaneous techniques for thoracolumbar fractures. In the present study, we compared radiological and clinical parameters from the immediate postoperative time to the 1-year follow-up between patients who underwent robot-assisted and conventional fluoroscopy surgical procedures.", "[SUBTITLE] Patient selection [SUBSECTION] This was a retrospective cohort study. A consecutive sample of 70 patients with A3 and A4 thoracolumbar fractures from May 2018 to October 2020 was enrolled. Of the 70 patients reviewed, 34 and 36 were treated with robotic-assisted percutaneous pedicle screw fixation and fluoroscopy-guided percutaneous pedicle screw fixation, respectively, based on the following inclusion criteria: (1) thoracolumbar fracture classified as A3 or A4; (2) short-segment fixation with two additional screws at the single fracture vertebra; (3) no other fractures or organ injuries; and (4) absence of neurological deficits. The exclusion criteria were as follows: (1) incomplete clinical information; (2) combined anterior-posterior surgeries; (3) revision surgeries; (4) fracture displacement in the vertebral pedicle; and (5) structural spinal deformity, spinal tumour, or infection. Written informed consent was obtained from all patients. The present study was reviewed and approved by the Ethics Committee and Institutional Review Board of the Second Affiliated Hospital of Soochow University.\nThis was a retrospective cohort study. A consecutive sample of 70 patients with A3 and A4 thoracolumbar fractures from May 2018 to October 2020 was enrolled. Of the 70 patients reviewed, 34 and 36 were treated with robotic-assisted percutaneous pedicle screw fixation and fluoroscopy-guided percutaneous pedicle screw fixation, respectively, based on the following inclusion criteria: (1) thoracolumbar fracture classified as A3 or A4; (2) short-segment fixation with two additional screws at the single fracture vertebra; (3) no other fractures or organ injuries; and (4) absence of neurological deficits. The exclusion criteria were as follows: (1) incomplete clinical information; (2) combined anterior-posterior surgeries; (3) revision surgeries; (4) fracture displacement in the vertebral pedicle; and (5) structural spinal deformity, spinal tumour, or infection. Written informed consent was obtained from all patients. The present study was reviewed and approved by the Ethics Committee and Institutional Review Board of the Second Affiliated Hospital of Soochow University.", "This was a retrospective cohort study. A consecutive sample of 70 patients with A3 and A4 thoracolumbar fractures from May 2018 to October 2020 was enrolled. Of the 70 patients reviewed, 34 and 36 were treated with robotic-assisted percutaneous pedicle screw fixation and fluoroscopy-guided percutaneous pedicle screw fixation, respectively, based on the following inclusion criteria: (1) thoracolumbar fracture classified as A3 or A4; (2) short-segment fixation with two additional screws at the single fracture vertebra; (3) no other fractures or organ injuries; and (4) absence of neurological deficits. The exclusion criteria were as follows: (1) incomplete clinical information; (2) combined anterior-posterior surgeries; (3) revision surgeries; (4) fracture displacement in the vertebral pedicle; and (5) structural spinal deformity, spinal tumour, or infection. Written informed consent was obtained from all patients. The present study was reviewed and approved by the Ethics Committee and Institutional Review Board of the Second Affiliated Hospital of Soochow University.", "[SUBTITLE] RAF group [SUBSECTION] Patients were placed in the prone position after general anaesthesia. The CT scan of the surgical region was uploaded to the workstation (Mazor Robotics Company, Israel) before surgery, and the perfect trajectories of the vertebrae based on the surgeon’s requests were planned (Fig. 1A–D). After the anteroposterior and oblique plane images were captured by the C-arm, the fluoroscopic images were automatically merged with the preoperative CT. A small (400 g, 9 cm tall, 5 cm diameter), parallel, 6-DOF robotic manipulator was then placed on the bone-mounted platform and aligned with a planned trajectory according to the surgeon’s instructions (Fig. 1E). After the paths of the screws were tapped with a thread tap through the dilated channels, the screws were inserted using handover guide wires (Fig. 1F). Rods were placed percutaneously from either the cephalad or caudal side with the assistance of screw extenders.\n\nFig. 1Perfect trajectories of the vertebrae based on the surgeon’s requests were planned (A–D). The manipulator was placed on the bone-mounted platform, and the appropriate pedicle screws were inserted (E, F)\nPerfect trajectories of the vertebrae based on the surgeon’s requests were planned (A–D). The manipulator was placed on the bone-mounted platform, and the appropriate pedicle screws were inserted (E, F)\nPatients were placed in the prone position after general anaesthesia. The CT scan of the surgical region was uploaded to the workstation (Mazor Robotics Company, Israel) before surgery, and the perfect trajectories of the vertebrae based on the surgeon’s requests were planned (Fig. 1A–D). After the anteroposterior and oblique plane images were captured by the C-arm, the fluoroscopic images were automatically merged with the preoperative CT. A small (400 g, 9 cm tall, 5 cm diameter), parallel, 6-DOF robotic manipulator was then placed on the bone-mounted platform and aligned with a planned trajectory according to the surgeon’s instructions (Fig. 1E). After the paths of the screws were tapped with a thread tap through the dilated channels, the screws were inserted using handover guide wires (Fig. 1F). Rods were placed percutaneously from either the cephalad or caudal side with the assistance of screw extenders.\n\nFig. 1Perfect trajectories of the vertebrae based on the surgeon’s requests were planned (A–D). The manipulator was placed on the bone-mounted platform, and the appropriate pedicle screws were inserted (E, F)\nPerfect trajectories of the vertebrae based on the surgeon’s requests were planned (A–D). The manipulator was placed on the bone-mounted platform, and the appropriate pedicle screws were inserted (E, F)", "Patients were placed in the prone position after general anaesthesia. The CT scan of the surgical region was uploaded to the workstation (Mazor Robotics Company, Israel) before surgery, and the perfect trajectories of the vertebrae based on the surgeon’s requests were planned (Fig. 1A–D). After the anteroposterior and oblique plane images were captured by the C-arm, the fluoroscopic images were automatically merged with the preoperative CT. A small (400 g, 9 cm tall, 5 cm diameter), parallel, 6-DOF robotic manipulator was then placed on the bone-mounted platform and aligned with a planned trajectory according to the surgeon’s instructions (Fig. 1E). After the paths of the screws were tapped with a thread tap through the dilated channels, the screws were inserted using handover guide wires (Fig. 1F). Rods were placed percutaneously from either the cephalad or caudal side with the assistance of screw extenders.\n\nFig. 1Perfect trajectories of the vertebrae based on the surgeon’s requests were planned (A–D). The manipulator was placed on the bone-mounted platform, and the appropriate pedicle screws were inserted (E, F)\nPerfect trajectories of the vertebrae based on the surgeon’s requests were planned (A–D). The manipulator was placed on the bone-mounted platform, and the appropriate pedicle screws were inserted (E, F)", "Patients were placed in the prone position after general anaesthesia. The fractured vertebrae were confirmed by fluoroscopy. Six transpedicular puncture needles were inserted through six small incisions using fluoroscopy, after which guide wires were inserted. The appropriate pedicle screws were then placed through the guide wires. The position of each screw was examined by fluoroscopy, followed by the insertion of titanium rods with the assistance of screw extenders.", "Patient demographics and clinical characteristics were recorded. The mean CT Hounsfield Unit (HU) values were obtained from the measurement of preoperative CT measurements (Fig. 2). The duration of the operation, number of radiation cycles, amount of intraoperative blood loss, length of hospital stay, and cost were compared between the two groups. The surgical data obtained included the levels of instrument, and pedicle screw length, and diameter. Screw dimensions were obtained from the patient’s electronic health records, to which all operative implant records were uploaded. Considering that short-length pedicle screws were used for the fractured vertebral body, only screws in the vertebra above and below the fractured vertebral body were counted.\n\nFig. 2Measurement methods for mean CT Hounsfield Unit (HU) values: a 41-year-old male patient was diagnosed with a vertebral fracture in L1. A The mean CT Hounsfield Unit (HU) values of the T12 vertebral body (VB) were 176, and the right and left pedicles (excluding the cortical bone) were 133 and 136 respectively; B the mean HU values of the right and left pedicles of the L1 (fractured vertebra) were 157 and 144 respectively; C the mean HU values of the L2 VB was 164, and the right and left pedicle were 160 and 147 respectively. The mean HU values of the patient was 152.1 = [(176 + 133 + 136 + 157 + 144 + 164 + 160 + 147)/8]\nMeasurement methods for mean CT Hounsfield Unit (HU) values: a 41-year-old male patient was diagnosed with a vertebral fracture in L1. A The mean CT Hounsfield Unit (HU) values of the T12 vertebral body (VB) were 176, and the right and left pedicles (excluding the cortical bone) were 133 and 136 respectively; B the mean HU values of the right and left pedicles of the L1 (fractured vertebra) were 157 and 144 respectively; C the mean HU values of the L2 VB was 164, and the right and left pedicle were 160 and 147 respectively. The mean HU values of the patient was 152.1 = [(176 + 133 + 136 + 157 + 144 + 164 + 160 + 147)/8]", "Two spine surgeons with more than 10 years of experience completed the measurements independently. The average value of the measured data was the final data. All data were measured using radiology software (Neusoft PACS/RIS) to reduce variability.\nBased on the Gertzbein and Robbins scale [16], the accuracy of the pedicle screw placement was evaluated on postoperative CT images as follows (Fig. 3A–E): Grade A: screw completely within bone; Grade B: cortical breach of < 2 mm; Grade C: cortical breach of ≥ 2 mm and < 4 mm; Grade D: cortical breach of ≥ 4 mm and < 6 mm; and Grade E: cortical breach of ≥ 6 mm.\n\nFig. 3Grading used for pedicle perforation on the axial CT scan and the representative images: A both pedicle screws are completely within the pedicle (Grade A); B Grade B (< 2 mm) perforation of the lateral wall of the right pedicle and Grade A of the left pedicle; C Grade C (2–4 mm) perforation of the medial wall of the left pedicle and Grade A perforation of the right pedicle; D Grade D (> 4 mm) perforation of the lateral wall of both pedicles; E Grade E (> 6 mm) perforation of the medial wall of the right pedicle. The blue lines represent the medial or lateral wall of the vertebral pedicles; the red lines represent the edge of the pedicle screws; and the green lines represent the distance of the perforation\nGrading used for pedicle perforation on the axial CT scan and the representative images: A both pedicle screws are completely within the pedicle (Grade A); B Grade B (< 2 mm) perforation of the lateral wall of the right pedicle and Grade A of the left pedicle; C Grade C (2–4 mm) perforation of the medial wall of the left pedicle and Grade A perforation of the right pedicle; D Grade D (> 4 mm) perforation of the lateral wall of both pedicles; E Grade E (> 6 mm) perforation of the medial wall of the right pedicle. The blue lines represent the medial or lateral wall of the vertebral pedicles; the red lines represent the edge of the pedicle screws; and the green lines represent the distance of the perforation\nThe pre- and postoperative sagittal kyphosis Cobb angles (defined by the upper endplate of the first vertebra above the fractured vertebra and by the lower endplate of the first vertebra below the fractured vertebra) and anterior vertebral height ratios (defined by the percentage of the mean values for the adjacent vertebrae) of the fractured vertebra were measured using radiographic images (Fig. 4).\n\nFig. 4Schematic drawings showing the measurement methods for the sagittal kyphosis Cobb angles (KCA) and anterior vertebral height ratios (VHR). A The KCA was defined as the angle between the superior endplate of the upper vertebra(superior black line) and the inferior endplate of the lower vertebra (inferior black line) in accordance with Cobb’s method; B VHR = 2b/(a + c)\nSchematic drawings showing the measurement methods for the sagittal kyphosis Cobb angles (KCA) and anterior vertebral height ratios (VHR). A The KCA was defined as the angle between the superior endplate of the upper vertebra(superior black line) and the inferior endplate of the lower vertebra (inferior black line) in accordance with Cobb’s method; B VHR = 2b/(a + c)\nPedicle screw loosening was defined as a “double-halo sign” on plain X-ray or a “lucent zone” around on CT images, which were obtained 1 year postoperatively (Fig. 5) [17, 18].\n\nFig. 5A Postoperative 1-year plain X-ray demonstrated left screw loosening at T12, L1, and L2 in a 39-year-old male who underwent short-segment fixation from T12 to L2 (black arrow: double halo sign); B postoperative 1-year CT demonstrated left screw loosening at L1 in a 47-year-old male who underwent short-segment fixation from T11 to L12 (white arrow: lucent zone)\nA Postoperative 1-year plain X-ray demonstrated left screw loosening at T12, L1, and L2 in a 39-year-old male who underwent short-segment fixation from T12 to L2 (black arrow: double halo sign); B postoperative 1-year CT demonstrated left screw loosening at L1 in a 47-year-old male who underwent short-segment fixation from T11 to L12 (white arrow: lucent zone)\n[SUBTITLE] Statistical analysis [SUBSECTION] Descriptive statistics are presented as the mean ± standard deviation for normally distributed continuous variables; the median and interquartile range was used for nonnormally distributed continuous variables and the count (frequency) was used for categorical variables. Comparisons of baseline characteristics, which were stratified by group, were performed using Student’s t tests for normally distributed continuous variables, Wilcoxon rank-sum tests for variables with skewed distributions, and chi-square tests for categorical data. A p value < 0.05 was considered statistically significant. All analyses were performed using SPSS for Mac (IBM SPSS Statistics 23.0, SPSS Inc., Chicago, IL, USA).\nDescriptive statistics are presented as the mean ± standard deviation for normally distributed continuous variables; the median and interquartile range was used for nonnormally distributed continuous variables and the count (frequency) was used for categorical variables. Comparisons of baseline characteristics, which were stratified by group, were performed using Student’s t tests for normally distributed continuous variables, Wilcoxon rank-sum tests for variables with skewed distributions, and chi-square tests for categorical data. A p value < 0.05 was considered statistically significant. All analyses were performed using SPSS for Mac (IBM SPSS Statistics 23.0, SPSS Inc., Chicago, IL, USA).", "Descriptive statistics are presented as the mean ± standard deviation for normally distributed continuous variables; the median and interquartile range was used for nonnormally distributed continuous variables and the count (frequency) was used for categorical variables. Comparisons of baseline characteristics, which were stratified by group, were performed using Student’s t tests for normally distributed continuous variables, Wilcoxon rank-sum tests for variables with skewed distributions, and chi-square tests for categorical data. A p value < 0.05 was considered statistically significant. All analyses were performed using SPSS for Mac (IBM SPSS Statistics 23.0, SPSS Inc., Chicago, IL, USA).", "Overall, 70 patients who underwent short-segment fixation were included in the study. Patient demographics and clinical characteristics were compared between the RAF and FGF groups. There were no significant differences between the groups in terms of age, sex, BMI, mean HU values, mechanism of injury, fracture level and type, and TLICS score. The baseline data of the two groups were comparable (Table 1).\n\nTable 1Baseline characteristics and perioperative parameters of all patientsCharacteristicRAFFGFStatistic (χ2/t)\nP valueNo. of pts3436––Mean age (years)42.8 ± 8.739.8 ± 10.71.2770.206Sex0.3700.558 Male2526–– Female910––Mean BMI (kg/m2)23.8 ± 2.624.0 ± 2.70.6990.718Mean HU values184.4 ± 66.5202.4 ± 80.4− 1.0260.311Mechanism of injury0.4750.830 Traffic accident2222 Fall1214Fracture level0.8260.972 T1000 T1134 T1288 L11816 L258Fracture type0.2140.543 A32521 A4915TLICS score4.5 ± 0.54.6 ± 0.5− 0.4480.656RAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation, BMI body mass index, HU Hounsfeld Unit, TLICS Score Thoracolumbar Injury Classification and Severity Score\nBaseline characteristics and perioperative parameters of all patients\nRAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation, BMI body mass index, HU Hounsfeld Unit, TLICS Score Thoracolumbar Injury Classification and Severity Score", "The RAF group had significantly fewer radiation cycles (P < 0.001) but higher hospitalization costs (P = 0.001) than the FGF group (Table 2). No significant difference was observed between the two groups in terms of surgical time, intraoperative blood loss, or length of hospital stay (Table 2).\n\nTable 2Summary of operation valuesOperative factorsRAFFGFStatistic (t/Z)\nP valueMean operation time (min)114.1 ± 34.4104.0 ± 18.11.5520.125Fluoroscopy frequency (times)16.1 ± 3.122.6 ± 3.9− 7.8150.000**Mean hospital stay (days)7.5 ± 2.48.2 ± 2.5− 1.2790.204Mean blood loss (mL)93.8 ± 24.9102.2 ± 27.8− 0.9630.339Hospitalization expenses (RMB)70529.6 ± 17224.558803.1 ± 9753.43.530.001**RAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation, RMB Renminbi**P < 0.01, statistically significant\nSummary of operation values\nRAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation, RMB Renminbi\n**P < 0.01, statistically significant", "There were 136 screws placed in the RAF group and 144 placed in the FGF group (screws in the vertebra above and below the fractured vertebra). Screws placed in the RAF group had both a larger screw diameter (RAF 6.04 ± 0.55 mm [range 5.0–7.0 mm]; FGF 5.78 ± 0.50 mm [range 5.0–6.5 mm]) and screw length (RAF 50.45 ± 4.37 mm [range 40–55 mm]; FGF 48.63 ± 3.86 mm [range 40–55 mm]) than screws placed in the FGF group (P < 0.001) (Table 3).\n\nTable 3Pedicle screw diameter and lengthVariableRAFFGFStatistic (χ2)\nP valueNo. of screws Total1361445.3090.379 T1026 T112220 T123634 L13238 L23428 L31018Mean screw diameter (range), mm Total6.04 (5.0–7.0)5.78 (5.0–6.5)− 3.0670.000** T105.5 (5.5)5.17 (5.0–5.5)–– T116.09 (5.5–7.0)5.90 (5.5–6.5)-0.9750.330 T126.11 (5.5–7.0)6.04 (5.5–6.5)-0.3320.740 L15.81 (5.0–6.5)5.29 (5.0–6.0)-4.2810.000** L26.12 (5.5–7.0)6.07 (5.5–6.5)-0.2380.812 L36.20 (6.0–6.5)5.94 (5.5–6.5)-2.1430.032*Mean screw length (range), mm Total50.45 (40–55)48.63 (40–55)-3.5610.000** T1050 (50)48.54 (45–50)–– T1150.00 (40–55)49.50 (40–55)− 0.3200.749 T1249.19 (45–55)47.35 (45–55)2.0640.039* L150.65 (40–55)48.96 (40–55)− 1.9660.049* L251.18 (45–55)49.34 (45–55)− 1.3910.164 L353.00 (50–55)48.33 (45–55)− 3.1440.020*RAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation\n*P < 0.05; **P < 0.01, statistically significant\nPedicle screw diameter and length\nRAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation\n\n*P < 0.05; **P < 0.01, statistically significant", "In the RAF group, 91.2% of the 204 screws were well placed (grade A); the remaining screws were graded B (5.4%), C (2.9%), or D (1.0%), but no screw was graded E. In the FGF group, 80.1% of the 216 screws were graded A; the remaining screws were graded B (9.3%), C (6.9%), D (2.8%), or E (0.9%). The RAF group had a significantly higher ratio of clinically acceptable screws (grades A and B, 96.6%) than the FGF group (89.4%) (P < 0.05) (Table 4).\n\nTable 4Pedicle screw accuracy rateVariableRAFFGFStatistic (χ2)\nP valueNo. of screws204216Screw accuracy rate11.7500.019* Grade A186 (91.2%)173 (80.1%) Grade B11 (5.4%)20 (9.3%) Grade A + B197 (96.6%)193 (89.4%) Grade C5 (2.4%)15 (6.9%) Grade D2 (1.0%)6 (2.8%) Grade E02 (0.9%) Grade C + D + E7 (3.4%)23 (10.6%)RAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation\n*P < 0.05 statistically significant\nPedicle screw accuracy rate\nRAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation\n\n*P < 0.05 statistically significant\nThe preoperative kyphosis Cobb angle (KCA) and anterior height ratio of fractured vertebra (VHR) were similar between the two groups. After surgery, the KCA was significantly corrected, and the VHR was well improved in both groups. The correction loss of the KCA (3.8 ± 1.8°) was less than that of the FGT group( 4.9 ± 4.2°), and the VHR of the RAF group (5.5 ± 4.9% ) was less than that of the FGT group (6.4 ± 5.7%) at the 1-year follow-up, but there were no significant differences (Table 5).\n\nTable 5Summary of radiographic measurementsVariableRAFFGFStatistic (χ2/Z/t)\nP value\nNo. of pts\n\n34\n\n36\n\n–\n\n–\n\nKyphosis Cobb angle (KCA) (°)\n Preoperative KCA20.2 ± 5.820.9 ± 4.3− 0.6130.542 Postoperative KCA8.2 ± 4.88.7 ± 3.9− 0.4940.621 Postoperative KCA correction12.0 ± 4.412.2 ± 3.4− 0.2560.798 KCA at 1-y FU11.9 ± 4.7− 9.3 ± 3.9− 1.3740.174 Correction loss at 1-y FU3.8 ± 1.84.9 ± 4.2− 1.4850.138\nAnterior height ratio of fracture vertebral (VHR) (%)\n Preoperative VHR62.0 ± 11.260.7 ± 14.30.4190.676 Postoperative VHR84.6 ± 9.982.5 ± 11.40.8120.420 Postoperative VHR Correction22.6 ± 12.121.9 ± 12.9− 0.1060.910 VHR at 1-y FU79.2 ± 7.976.1 ± 10.71.3560.180 Correction loss at 1-y FU5.5 ± 4.96.4 ± 5.7− 0.9750.329\nScrew loosening rate of pts (%)\n2 (5.9%)6 (16.7%)2.0090.156RAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation, KCA Kyphosis Cobb angle, AVHR anterior vertebral height ratios, 1-y FU 1-year follow-up, pts patient\nSummary of radiographic measurements\nRAF robot-assisted percutaneous pedicle screw fixation, FGF fluoroscopy-guided percutaneous pedicle screw fixation, KCA Kyphosis Cobb angle, AVHR anterior vertebral height ratios, 1-y FU 1-year follow-up, pts patient\nScrew loosening occurred in 2 out of 34 patients (5.9%) in the RAF group, and 6 out of 36 patients (16.7%) in the FGF group. There was no statistically significant difference in terms of the screw loosening rate between the two groups (Table 5).", "Our study had several main findings as follows. First, the robot-assisted technique provides more precise screw placement in minimally invasive spine surgery than conventional fluoroscopy. Second, robot-assisted techniques allow for the placement of screws with greater screw diameter and length compared with fluoroscopy-guided techniques. That is, robotic assistance may allow for safe placement of the “optimal screw”. Third, we found that robot-assisted technology can reduce intraoperative radiation exposure and increase treatment costs. Finally, although there was no significant difference, there was a lower correction loss at the 1-year follow-up and the loosening rate of the screw in the RAF group.\nIn recent years, there have been many studies on the robot’s advantage in accuracy and selection of pedicle screws, offering benefits for both patients and surgeons [15, 19–21]. Preoperative CT is used to plan screw trajectories, and intraoperative fluoroscopy is applied to register images. The robot then guides the surgeon to the appropriate trajectory, with precise screw placement, less redirection [22, 23], and a lower axial trajectory [7]. Molliqaj et al. [12] reported a clinically acceptable screw placement accuracy under robotic guidance of up to 93.4%. Karim et al. [15]found that surgeons, with the assistance of a robot, tended to choose larger-diameter and longer screws in the minimally invasive surgery-transforaminal lumbar interbody fusion when compared with intraoperative navigation alone, particularly when using sizes that they may have previously thought to be too large. Our results are somewhat similar to these studies. In our study, compared with fluoroscopy-guided techniques, robotic assistance helped surgeons to select the larger screws and ensured accuracy in the screw placement.\nTheoretically, these benefits may reduce the screw loosening rate, maintain the fractured vertebral height, and provide a more stable environment for the healing of the thoracolumbar fracture. Numerous in vitro experiments have demonstrated that perfect central-axis pedicle screws without redirection are significantly more powerful in terms of screw loosening force and axial pull-out strength and that larger-diameter and longer screws have greater resistance to screw pull-out [22, 24–26]. However, these studies have inherent limitations associated with cadaveric studies and/or bone models, with which it is difficult to mimic the actual clinical situation. A previous study showed that the instability of pedicle screw increased the loosening rate and loss of correction in vertebral fractures [27]. Ohba et al. investigated the association between the pull-out length and screw loosening 1 year after surgery, and found 81.8% of patients with loosened pedicle screws had developed the screw pull-out phenomenon postoperatively [7]. Therefore, to determine whether these “optimal” screws may provide a more stable environment, our study compared the kyphosis Cobb angle of fractured segments, the anterior height ratio of fractured vertebra, and the screw loosening rate between the two groups at the 1-year follow-up.\nOur study revealed that the loosening rate of the RAF group was lower than that of the FGT group (5.9% vs. 16.7%), and the correction loss of the KCA (3.8 ± 1.8° vs. 4.9 ± 4.2°) and VHR (5.5 ± 4.9% vs. 6.4 ± 5.7%) of the RAF group was less than that of the FGT group at the 1-year follow-up, although there were no significant differences in the groups. Of note, 5 out of 8 patients with loosened pedicle screws (62.5%) in this study had showed the obvious screw pull-out phenomenon (data not shown). Three possible interpretations were considered to explain why the significant differences were not seen in our study. First, the mechanisms that contribute to pedicle screw loosening are certain to be multifactorial, in addition to the screw placement and sizes mentioned, including high forces of spinal rods, bone mineral density (BMD), postoperative physiological movements, and paraspinal muscle function [28–30]. Previous studies have shown that BMD plays an important role in the stability of screws [31, 32], so BMD was assessed by the CT HU values to make the two groups comparable in our study. The patients’ CT HU values were relatively high in the two groups, because most of these patients were young, which might increase the holding power of screws and avoid loosening. Second, the sample size of fracture cases was relatively small, and all patients were recruited from a single centre. Last, the 1-year follow-up period was short, and most of patients required removal of the internal fixation after fracture healing. Overall, although the advantages of robotic surgery in the terms of screw stability were not confirmed in our study, the robotic technology is one possible way to insert pedicle screws following the trajectory, which has been designed to increase purchase in the bone and obtain adequate fixation, especially in osteoporotic bone.\nMoreover, we also found that robot-assisted technology helped to reduce intraoperative radiation exposure, which is consistent with previous studies [33]. Another clinically relevant finding is that the treatment cost in the RAF group was higher than that in the conventional group, which was due to the additional costs of disposable materials.", "Our study has several limitations. First, the sample size of fracture cases was relatively small, and all patients were recruited from a single centre. Future studies with larger sample sizes are needed to replicate the findings. Second, data on the trajectory angles, the radians and forces of spinal rods, and misaligned spinal rods, all of which can cause screw loosening, were not available. Finally, further experiments are necessary to design an adequate trajectory, which can significantly increase the stability of the pedicle screw.", "Compared with fluoroscopy-guided techniques, robotic assistance can help a surgeon to optimize screw trajectories and dimensions intraoperatively. Although not statistically significant, these findings are a possible way to increase bone purchase and reduce the screw loosening rate for thoracolumbar fractures. In addition, with increased experience and proficiency in robot-assisted techniques, surgeons should be more confident in their screw selection, especially when using sizes that they may have previously considered too large." ]
[ null, "materials|methods", null, null, null, null, null, null, null, "results", null, null, null, "discussion", null, "conclusion" ]
[ "Thoracolumbar fracture", "Robot-assisted surgery", "Minimally invasive surgery", "Screw loosening" ]
Quantitative parameters of dynamic contrast-enhanced magnetic resonance imaging to predict lymphovascular invasion and survival outcome in breast cancer.
36273200
Lymphovascular invasion (LVI) predicts a poor outcome of breast cancer (BC), but LVI can only be postoperatively diagnosed by histopathology. We aimed to determine whether quantitative parameters of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) can preoperatively predict LVI and clinical outcome of BC patients.
BACKGROUND
A total of 189 consecutive BC patients who underwent multiparametric MRI scans were retrospectively evaluated. Quantitative (Ktrans, Ve, Kep) and semiquantitative DCE-MRI parameters (W- in, W- out, TTP), and clinicopathological features were compared between LVI-positive and LVI-negative groups. All variables were calculated by using univariate logistic regression analysis to determine the predictors for LVI. Multivariate logistic regression was used to build a combined-predicted model for LVI-positive status. Receiver operating characteristic (ROC) curves evaluated the diagnostic efficiency of the model and Kaplan-Meier curves showed the relationships with the clinical outcomes. Multivariate analyses with a Cox proportional hazard model were used to analyze the hazard ratio (HR) for recurrence-free survival (RFS) and overall survival (OS).
METHODS
LVI-positive patients had a higher Kep value than LVI-negative patients (0.92 ± 0.30 vs. 0.81 ± 0.23, P = 0.012). N2 stage [odds ratio (OR) = 3.75, P = 0.018], N3 stage (OR = 4.28, P = 0.044), and Kep value (OR = 5.52, P = 0.016) were associated with LVI positivity. The combined-predicted LVI model that incorporated the N stage and Kep yielded an accuracy of 0.735 and a specificity of 0.801. The median RFS was significantly different between the LVI-positive and LVI-negative groups (31.5 vs. 34.0 months, P = 0.010) and between the combined-predicted LVI-positive and LVI-negative groups (31.8 vs. 32.0 months, P = 0.007). The median OS was not significantly different between the LVI-positive and LVI-negative groups (41.5 vs. 44.0 months, P = 0.270) and between the combined-predicted LVI-positive and LVI-negative groups (42.8 vs. 43.5 months, P = 0.970). LVI status (HR = 2.40), N2 (HR = 3.35), and the combined-predicted LVI model (HR = 1.61) were independently associated with disease recurrence.
RESULTS
The quantitative parameter of Kep could predict LVI. LVI status, N stage, and the combined-predicted LVI model were predictors of a poor RFS but not OS.
CONCLUSION
[ "Humans", "Female", "Breast Neoplasms", "Retrospective Studies", "Magnetic Resonance Imaging", "Proportional Hazards Models", "ROC Curve" ]
9587620
null
null
null
null
Results
[SUBTITLE] Patient characteristics [SUBSECTION] Finally, 189 women (mean age: 51.1 years; range:25~95 years) were included in this study. Out of 189 included women, 43 had LVI-positive tumors and 146 had LVI-negative tumors. The characteristics of patients in the LVI-negative and LVI-positive groups are shown in Table E1 in the supplementary materials. Those pertinent features are presented in Table 1. The occurrence rates of lymphatic metastasis was more higher in LVI-positive patients than in LVI-negative patients (P = 0.003). In addition, the LVI-positive patients had higher Kep values than the LVI-negative patients (0.92 ± 0.30 vs. 0.81 ± 0.23, P = 0.012, Fig. 1). No significant differences were found for age, tumor size, ER, PR, HER2, Ki-67 index, tumor grade, molecular subtype, T stage, M stage, AJCC stage, Ktrans, Ve, W-in, W-out, or TTP value between patients with and without LVI (all P > 0.05). Table 1The characteristics of the patients in the LVI-negative and LVI-positive groupsLVI- (n = 146)LVI+ (n = 43) P OR (95% CI) P d Maximum diameter (cm)2.89 ± 1.123.09 ± 1.180.312b1.16 (0.86–1.55)0.312Minimum diameter (cm)1.85 ± 0.651.92 ± 0.720.528b1.17 (0.70–1.92)0.526ER0.837aNegative50 (34.25%)14 (32.56%)1NAPositive96 (65.75%)29 (67.44%)1.08 (0.53–2.27)0.837PR0.523aNegative70 (47.95%)23 (53.49%)1NAPositive76 (52.05%)20 (46.51%)0.80 (0.40–1.58)0.523HER20.845aNegative103 (70.55%)31 (72.09%)1NAPositive43 (29.45%)12 (27.91%)0.93 (0.42–1.94)0.845Ki-670.409a≤20%47 (32.19%)11 (25.58%)1NA>20%99 (67.81%)32 (74.42%)1.38 (0.66–3.08)0.410T stage0.109cT014 (9.59%)0 (0.00%)0.0 (0.0-3.48 × 10 13)0.988T146 (31.51%)10 (23.26%)1NAT284 (57.53%)32 (74.42%)1.75 (0.81–4.05)0.167T31 (0.68%)0 (0.00%)0.0 (NA-Inf)0.997T41 (0.68%)1 (2.33%)4.6 (0.17–123.2)0.295 N stage 0.003 c N091 (62.33%)17 (39.53%)1NAN140 (27.40%)15 (34.88%)2.01 (0.91–4.43)0.083N210 (6.85%)7 (16.28%)3.75 (1.21–11.2) 0.018 N35 (3.42%)4 (9.30%)4.28 (0.98–17.85) 0.044 ParametersKtrans(min− 1)0.21 ± 0.110.22 ± 0.120.766b1.58 (0.07–30.11)0.765Kep(min− 1)0.81 ± 0.230.92 ± 0.30 0.012 b 5.52 (1.42–23.3) 0.016 V e 0.27 ± 0.130.24 ± 0.090.102b0.12 (0.01–2.15)0.163 W-in (min− 1)0.56 ± 0.220.61 ± 0.240.228b2.50 (0.56–11.17)0.228 W-out (min− 1)-0.01 ± 0.02-0.02 ± 0.020.088b0.00 (0.0-9.35)0.090TTP (min)0.69 ± 0.210.64 ± 0.170.165b0.25 (0.03–1.58)0.167Note. Pa: chi-squared test, Pb: Student’s t-test, Pc: Kruskal-Wallis H test, Pd: univariate analysis. Abbreviations: ER = Estrogen receptor; PR = Progesterone receptor;HER2 = Human epidermal growth factor receptor2; TNBC = Triple negative breast cancer; NA = Not available. The characteristics of the patients in the LVI-negative and LVI-positive groups Note. Pa: chi-squared test, Pb: Student’s t-test, Pc: Kruskal-Wallis H test, Pd: univariate analysis. Abbreviations: ER = Estrogen receptor; PR = Progesterone receptor;HER2 = Human epidermal growth factor receptor2; TNBC = Triple negative breast cancer; NA = Not available. Fig. 1Representative Kep images from breast cancer patients with (A) lymphovascular invasion (LVI) and without (B) LVI. Red represents high values of Kep, yellow represents intermediate values of Kep, and blue represents low values of Kep. The Kep values of the tumors were 1.05 min− 1 and 0.757 min− 1, respectively Representative Kep images from breast cancer patients with (A) lymphovascular invasion (LVI) and without (B) LVI. Red represents high values of Kep, yellow represents intermediate values of Kep, and blue represents low values of Kep. The Kep values of the tumors were 1.05 min− 1 and 0.757 min− 1, respectively Finally, 189 women (mean age: 51.1 years; range:25~95 years) were included in this study. Out of 189 included women, 43 had LVI-positive tumors and 146 had LVI-negative tumors. The characteristics of patients in the LVI-negative and LVI-positive groups are shown in Table E1 in the supplementary materials. Those pertinent features are presented in Table 1. The occurrence rates of lymphatic metastasis was more higher in LVI-positive patients than in LVI-negative patients (P = 0.003). In addition, the LVI-positive patients had higher Kep values than the LVI-negative patients (0.92 ± 0.30 vs. 0.81 ± 0.23, P = 0.012, Fig. 1). No significant differences were found for age, tumor size, ER, PR, HER2, Ki-67 index, tumor grade, molecular subtype, T stage, M stage, AJCC stage, Ktrans, Ve, W-in, W-out, or TTP value between patients with and without LVI (all P > 0.05). Table 1The characteristics of the patients in the LVI-negative and LVI-positive groupsLVI- (n = 146)LVI+ (n = 43) P OR (95% CI) P d Maximum diameter (cm)2.89 ± 1.123.09 ± 1.180.312b1.16 (0.86–1.55)0.312Minimum diameter (cm)1.85 ± 0.651.92 ± 0.720.528b1.17 (0.70–1.92)0.526ER0.837aNegative50 (34.25%)14 (32.56%)1NAPositive96 (65.75%)29 (67.44%)1.08 (0.53–2.27)0.837PR0.523aNegative70 (47.95%)23 (53.49%)1NAPositive76 (52.05%)20 (46.51%)0.80 (0.40–1.58)0.523HER20.845aNegative103 (70.55%)31 (72.09%)1NAPositive43 (29.45%)12 (27.91%)0.93 (0.42–1.94)0.845Ki-670.409a≤20%47 (32.19%)11 (25.58%)1NA>20%99 (67.81%)32 (74.42%)1.38 (0.66–3.08)0.410T stage0.109cT014 (9.59%)0 (0.00%)0.0 (0.0-3.48 × 10 13)0.988T146 (31.51%)10 (23.26%)1NAT284 (57.53%)32 (74.42%)1.75 (0.81–4.05)0.167T31 (0.68%)0 (0.00%)0.0 (NA-Inf)0.997T41 (0.68%)1 (2.33%)4.6 (0.17–123.2)0.295 N stage 0.003 c N091 (62.33%)17 (39.53%)1NAN140 (27.40%)15 (34.88%)2.01 (0.91–4.43)0.083N210 (6.85%)7 (16.28%)3.75 (1.21–11.2) 0.018 N35 (3.42%)4 (9.30%)4.28 (0.98–17.85) 0.044 ParametersKtrans(min− 1)0.21 ± 0.110.22 ± 0.120.766b1.58 (0.07–30.11)0.765Kep(min− 1)0.81 ± 0.230.92 ± 0.30 0.012 b 5.52 (1.42–23.3) 0.016 V e 0.27 ± 0.130.24 ± 0.090.102b0.12 (0.01–2.15)0.163 W-in (min− 1)0.56 ± 0.220.61 ± 0.240.228b2.50 (0.56–11.17)0.228 W-out (min− 1)-0.01 ± 0.02-0.02 ± 0.020.088b0.00 (0.0-9.35)0.090TTP (min)0.69 ± 0.210.64 ± 0.170.165b0.25 (0.03–1.58)0.167Note. Pa: chi-squared test, Pb: Student’s t-test, Pc: Kruskal-Wallis H test, Pd: univariate analysis. Abbreviations: ER = Estrogen receptor; PR = Progesterone receptor;HER2 = Human epidermal growth factor receptor2; TNBC = Triple negative breast cancer; NA = Not available. The characteristics of the patients in the LVI-negative and LVI-positive groups Note. Pa: chi-squared test, Pb: Student’s t-test, Pc: Kruskal-Wallis H test, Pd: univariate analysis. Abbreviations: ER = Estrogen receptor; PR = Progesterone receptor;HER2 = Human epidermal growth factor receptor2; TNBC = Triple negative breast cancer; NA = Not available. Fig. 1Representative Kep images from breast cancer patients with (A) lymphovascular invasion (LVI) and without (B) LVI. Red represents high values of Kep, yellow represents intermediate values of Kep, and blue represents low values of Kep. The Kep values of the tumors were 1.05 min− 1 and 0.757 min− 1, respectively Representative Kep images from breast cancer patients with (A) lymphovascular invasion (LVI) and without (B) LVI. Red represents high values of Kep, yellow represents intermediate values of Kep, and blue represents low values of Kep. The Kep values of the tumors were 1.05 min− 1 and 0.757 min− 1, respectively [SUBTITLE] Interobserver agreement [SUBSECTION] The ICC values between two readers for Ktrans, Kep, Ve, W-in, W-out, and TTP were 0.994 (95% CI: 0.992–0.995), 0.858 (95%CI:0.815–0.891), 0.984 (95%CI:0.979–0.988), 0.987 (95%CI: 0.983–0.990), 0.979 (95%CI:0.972–0.984), and 0.961 (95%CI:0.949–0.971), respectively, indicating good or excellent reliability. The coefficient of variation on ICC value is 0.057. The ICC values between two readers for Ktrans, Kep, Ve, W-in, W-out, and TTP were 0.994 (95% CI: 0.992–0.995), 0.858 (95%CI:0.815–0.891), 0.984 (95%CI:0.979–0.988), 0.987 (95%CI: 0.983–0.990), 0.979 (95%CI:0.972–0.984), and 0.961 (95%CI:0.949–0.971), respectively, indicating good or excellent reliability. The coefficient of variation on ICC value is 0.057. [SUBTITLE] Predictors of LVI and model development [SUBSECTION] Univariate logistic regression analysis (Table E1 in the supplementary materials) showed that N2 (OR = 3.75, P = 0.018), N3 (OR = 4.28, P = 0.044), and Kep value (OR = 5.52, P = 0.016) were associated with LVI positivity. ROC curves were generated based on the prediction probability of the regression equation using the above variables (Table 2; Fig. 2). The combined-predicted LVI model that incorporated the N stage and Kep yielded a maximum AUC of 0.669 (0.574–0.765), with a cutoff value of 0.276, accuracy of 0.735, sensitivity of 0.512, and specificity of 0.801. Based on the combined-predicted model results, 51 patients were LVI positive and 138 were LVI negative. Table 2 Performance of the individualized prediction modelsModelsCutoffAUC(95% CI)AccuracySensitivitySpecificityN0.5000.631 (0.542–0.721)0.6190.6050.623 K ep 0.8080.601 (0.502–0.699)0.5450.6740.507Combined0.2760.669 (0.574–0.765)0.7350.5120.801Note: N and Kep indicate the predicted model based on the N stage and Kep, respectively. Combined indicate the predicted model based on the combination of all above parameters. N mean N2/3 stage versus N0/1 stage. Performance of the individualized prediction models Note: N and Kep indicate the predicted model based on the N stage and Kep, respectively. Combined indicate the predicted model based on the combination of all above parameters. N mean N2/3 stage versus N0/1 stage. Fig. 2Receiver operating characteristic (ROC) curve of the model established by N stage, Kep, and combined-predicted model that incorporated N stage and Kep for the prediction of lymphovascular invasion in breast cancer patients Receiver operating characteristic (ROC) curve of the model established by N stage, Kep, and combined-predicted model that incorporated N stage and Kep for the prediction of lymphovascular invasion in breast cancer patients Univariate logistic regression analysis (Table E1 in the supplementary materials) showed that N2 (OR = 3.75, P = 0.018), N3 (OR = 4.28, P = 0.044), and Kep value (OR = 5.52, P = 0.016) were associated with LVI positivity. ROC curves were generated based on the prediction probability of the regression equation using the above variables (Table 2; Fig. 2). The combined-predicted LVI model that incorporated the N stage and Kep yielded a maximum AUC of 0.669 (0.574–0.765), with a cutoff value of 0.276, accuracy of 0.735, sensitivity of 0.512, and specificity of 0.801. Based on the combined-predicted model results, 51 patients were LVI positive and 138 were LVI negative. Table 2 Performance of the individualized prediction modelsModelsCutoffAUC(95% CI)AccuracySensitivitySpecificityN0.5000.631 (0.542–0.721)0.6190.6050.623 K ep 0.8080.601 (0.502–0.699)0.5450.6740.507Combined0.2760.669 (0.574–0.765)0.7350.5120.801Note: N and Kep indicate the predicted model based on the N stage and Kep, respectively. Combined indicate the predicted model based on the combination of all above parameters. N mean N2/3 stage versus N0/1 stage. Performance of the individualized prediction models Note: N and Kep indicate the predicted model based on the N stage and Kep, respectively. Combined indicate the predicted model based on the combination of all above parameters. N mean N2/3 stage versus N0/1 stage. Fig. 2Receiver operating characteristic (ROC) curve of the model established by N stage, Kep, and combined-predicted model that incorporated N stage and Kep for the prediction of lymphovascular invasion in breast cancer patients Receiver operating characteristic (ROC) curve of the model established by N stage, Kep, and combined-predicted model that incorporated N stage and Kep for the prediction of lymphovascular invasion in breast cancer patients [SUBTITLE] Prognostic value of LVI [SUBSECTION] As of February 28, 2021, all patients had completed RFS follow-up, and the median RFS of all patients was 32.0 (23.0-42.8) months, of which 16 of 189 (8.47%) had a tumor recurrence. Among 16 patient with recurrences, 4 patients had locoregional recurrence and 12 patients had distant metastases. Of note, the recurrence rate was 18.6% (8/43) in patients with LVI and 5.48% (8/146) in patients without LVI, with a significant difference between the two groups (P = 0.016). The median RFS was 31.5 (23.0–42.0) months for patients with LVI and 34.0 (22.0-44.8) months for patients without LVI (log-rank test, P = 0.010, Fig. 3 A). Similar results were observed in the combined-predicted LVI model: The median RFS was 31.8 (23.6–58.5) months for patients with combined-predicted LVI presence and 32.0 (21.8–60.0) months for patients with combined-predicted LVI absence (log-rank test, P = 0.007, Fig. 3B). According to the results of the univariate Cox regression analysis (Table E2 in the supplementary materials), LVI (HR = 3.38, P = 0.015), N2 (HR = 6.13, P = 0.007), and the combined-predicted LVI model (HR = 3.61, P = 0.011) were associated with RFS. Multivariate Cox regression analysis showed that N2 (HR = 4.69, P = 0.026) was independent predictors of disease recurrence. A representative case is provided to show the discriminative ability of the combined-predicted model for predicting LVI and RFS (Fig. 4). Fig. 3Recurrence-free survival (RFS) curves scaled by histologic lymphovascular invasion (LVI) status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis Recurrence-free survival (RFS) curves scaled by histologic lymphovascular invasion (LVI) status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis Fig. 4 A 59-year-old woman with invasive ductal carcinoma of the right breast. Dynamic contrast-enhanced magnetic resonance imaging (A) showing a solid mass in the upper inner quadrant of the right breast, with nonhomogeneous obvious enhancement, Kep value of 1.09 min− 1 (B), and lymph node metastasis to the right axilla (C). The LVI risk calculated by the combined model which was built based on the combination of N stage and Kep was 70.7%. She was confirmed to be LVI positive by histopathology and had multiple metastatic lesions in the right and left liver lobe on follow-up CT (D) 32.5 months after surgery  A 59-year-old woman with invasive ductal carcinoma of the right breast. Dynamic contrast-enhanced magnetic resonance imaging (A) showing a solid mass in the upper inner quadrant of the right breast, with nonhomogeneous obvious enhancement, Kep value of 1.09 min− 1 (B), and lymph node metastasis to the right axilla (C). The LVI risk calculated by the combined model which was built based on the combination of N stage and Kep was 70.7%. She was confirmed to be LVI positive by histopathology and had multiple metastatic lesions in the right and left liver lobe on follow-up CT (D) 32.5 months after surgery When 98.4% (186/189) of patients had completed OS follow-up, the median OS of all patients was 43.0 (31.5–56.5) months, and the overall death rate was 3.70% (7/189). The median OS was 41.5 (32.0-55.5) months for patients with LVI and 44.0 (31.0-57.3) months for patients without LVI (log-rank test, P = 0.27, Fig. 5 A), which is consistent with the results of the combined-predicted LVI model: The median OS was 42.8 (32.0-59.5) months for patients with combined-predicted LVI presence and 43.5 (30.8–60.0) months for patients with combined-predicted LVI absence (log-rank test, P = 0.970, Fig. 5B). There were no variables significantly associated with OS in the univariate Cox regression analysis (Table E1 in the supplementary materials, all P > 0.05). Fig. 5Overall survival (OS) curves scaled by histologic LVI status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis Overall survival (OS) curves scaled by histologic LVI status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis As of February 28, 2021, all patients had completed RFS follow-up, and the median RFS of all patients was 32.0 (23.0-42.8) months, of which 16 of 189 (8.47%) had a tumor recurrence. Among 16 patient with recurrences, 4 patients had locoregional recurrence and 12 patients had distant metastases. Of note, the recurrence rate was 18.6% (8/43) in patients with LVI and 5.48% (8/146) in patients without LVI, with a significant difference between the two groups (P = 0.016). The median RFS was 31.5 (23.0–42.0) months for patients with LVI and 34.0 (22.0-44.8) months for patients without LVI (log-rank test, P = 0.010, Fig. 3 A). Similar results were observed in the combined-predicted LVI model: The median RFS was 31.8 (23.6–58.5) months for patients with combined-predicted LVI presence and 32.0 (21.8–60.0) months for patients with combined-predicted LVI absence (log-rank test, P = 0.007, Fig. 3B). According to the results of the univariate Cox regression analysis (Table E2 in the supplementary materials), LVI (HR = 3.38, P = 0.015), N2 (HR = 6.13, P = 0.007), and the combined-predicted LVI model (HR = 3.61, P = 0.011) were associated with RFS. Multivariate Cox regression analysis showed that N2 (HR = 4.69, P = 0.026) was independent predictors of disease recurrence. A representative case is provided to show the discriminative ability of the combined-predicted model for predicting LVI and RFS (Fig. 4). Fig. 3Recurrence-free survival (RFS) curves scaled by histologic lymphovascular invasion (LVI) status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis Recurrence-free survival (RFS) curves scaled by histologic lymphovascular invasion (LVI) status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis Fig. 4 A 59-year-old woman with invasive ductal carcinoma of the right breast. Dynamic contrast-enhanced magnetic resonance imaging (A) showing a solid mass in the upper inner quadrant of the right breast, with nonhomogeneous obvious enhancement, Kep value of 1.09 min− 1 (B), and lymph node metastasis to the right axilla (C). The LVI risk calculated by the combined model which was built based on the combination of N stage and Kep was 70.7%. She was confirmed to be LVI positive by histopathology and had multiple metastatic lesions in the right and left liver lobe on follow-up CT (D) 32.5 months after surgery  A 59-year-old woman with invasive ductal carcinoma of the right breast. Dynamic contrast-enhanced magnetic resonance imaging (A) showing a solid mass in the upper inner quadrant of the right breast, with nonhomogeneous obvious enhancement, Kep value of 1.09 min− 1 (B), and lymph node metastasis to the right axilla (C). The LVI risk calculated by the combined model which was built based on the combination of N stage and Kep was 70.7%. She was confirmed to be LVI positive by histopathology and had multiple metastatic lesions in the right and left liver lobe on follow-up CT (D) 32.5 months after surgery When 98.4% (186/189) of patients had completed OS follow-up, the median OS of all patients was 43.0 (31.5–56.5) months, and the overall death rate was 3.70% (7/189). The median OS was 41.5 (32.0-55.5) months for patients with LVI and 44.0 (31.0-57.3) months for patients without LVI (log-rank test, P = 0.27, Fig. 5 A), which is consistent with the results of the combined-predicted LVI model: The median OS was 42.8 (32.0-59.5) months for patients with combined-predicted LVI presence and 43.5 (30.8–60.0) months for patients with combined-predicted LVI absence (log-rank test, P = 0.970, Fig. 5B). There were no variables significantly associated with OS in the univariate Cox regression analysis (Table E1 in the supplementary materials, all P > 0.05). Fig. 5Overall survival (OS) curves scaled by histologic LVI status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis Overall survival (OS) curves scaled by histologic LVI status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis
Conclusion
In conclusion, the quantitative parameters of Kep from DCE-MRI, and N2 are independent predictors of LVI in BC patients. LVI status, N stage, and the combined-predicted LVI model were confirmed to predict a poor RFS, but no evidence was found that these features were related to OS.
[ "Background", "Basic characteristics", "Definition of recurrence-free survival and follow-up", "MRI examination", "MR image analysis", "Statistical analysis", "Patient characteristics", "Interobserver agreement", "Predictors of LVI and model development", "Prognostic value of LVI", "" ]
[ "Lymphovascular invasion (LVI), defined as the infiltration of tumor cells into lymphatic or blood vessels at the periphery of invasive carcinoma [1–3], has been widely recognized as a negative prognostic factor in multiple cancers, such as breast cancer (BC), gastric cancer, and rectal cancer [4–7]. Even so, LVI has not been included as an important parameter to consider before adjuvant chemotherapy in the National Comprehensive Cancer Network guidelines for BC due to its hard to determine before adjuvant chemotherapy, which can only be postoperatively diagnosed by histopathology. Thus, more evidence is needed to warrant the application of LVI in clinical decision-making.\nAs a significant preoperative examination for BC patients, dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) are effective tools for evaluating tumor microcirculation [2, 8–13]. The correlation between tumor microcirculation and LVI have been demonstrated and assessed by qualitative parameters of breast MRI in several small cohort studies [14–17]. However, few studies have been published on the prediction of LVI using large samples of quantitative DCE-MRI parameters. In addition, the relationship between the prediction model of LVI and the prognosis of BC is unclear.\nThus, the purpose of this retrospective study was to determine whether quantitative parameters of DCE-MRI can preoperatively predict LVI and to investigate the relationship between the prediction model and survival in terms of disease-specific recurrence and mortality in BC patients to guide clinical decision-making.", "This retrospective study was approved by our institutional review, and it waived the informed consent requirement. Between January 2016 and March 2019, 460 consecutive women with BC confirmed by postoperative histopathology had undergone preoperative DCE-MRI of the breast. Their medical charts were reviewed. The patient exclusion criteria were shown as follows: (1) patients with previous neoadjuvant treatment; (2) patients with recurrence of BC; (3) patients without obvious lesions on breast MRI; (4) patients with poor quality MRI images; (5) patients with a maximum tumor diameter < 1.0 cm;6) patients with non-mass-like enhancement lesions; and 7) patients with bilateral lesions or multiple lesions.The patient inclusion and exclusion criteria were shown in Figure E1 in the supplementary materials. The study patients are part of a large retrospective DCE-MRI database, of which 165 patients have been reported in a previously published study [18]. A previous study only explored the ability of DCE-MRI to predict BC receptor status and molecular subtypes, rather than LVI and survival outcome [18]. Cinicopathological data, inculding age, tumor size (maximum, minimum, and effective diameter), progesterone receptor (PR), estrogen receptor (ER), human epidermal growth factor receptor2 (HER2), Ki-67, molecular subtypes, tumor grade, tumor node metastasis(TNM) stage, and postoperative treatment methods (radiation therapy, adjuvant endocrine therapy, and adjuvant chemotherapy) were used to predict LVI. The clinicopathological data except receptor status and age were no used in the previous study. In addition, follow-up information were added for survival analysis of LVI.\nEffective tumor diameter was defined as the mean value of the maximum and minimum tumor diameter. The TNM stage was reclassified based on the 8th edition of the American Joint Committee on Cancer (AJCC) staging manual. LVI was defined as tumor cells present within a definite endothelial-lined space (either lymphatic or blood vessel) that was only visible on microscopy. Additionally, the immunohistochemistry (IHC) technique was used to classify the status of ER, PR, HER2, and Ki-67 [19].The standard of cut off values for the positivity of Ki-67, HER-2, PR, and ER were all based on internationally recognized standards [13].", "According to the follow-up protocol of our hospital, all patients were postoperatively followed up with chest and abdominal CT, and breast mammography every 12 months. Additionally, the patients with mastectomy were postoperatively followed up with breast, abdominal, gynecological, and superficial lymph node ultrasound every 3 months for the first two years. Of the contralateral breast usually followed up with breast mammography and ultrasound. The patients with breast-conserving surgery were postoperatively followed up with breast MRI every 12 months.\nRecurrence-free survival (RFS) was calculated in months from the date of surgery to the first date of local recurrence, distant metastasis, February 28, 2021, or death, whichever came first. Overall survival (OS) was calculated in months from the date of surgery to the date of death or on February 28, 2021, whichever came first.", "Breast MRI examinations were performed on a 3.0 T MR scanner (Magnetom Skyra, Siemens Healthcare, Erlangen, Germany) using a 16-channel bilateral breast coil with the patient in the prone position. Before dynamic scanning, T1-weighted volume interpolated breath-hold examination (VIBE) was first performed at two different flip angles 2° and 15° to calculate the T1-mapping images (TR = 3.78 ms, TE = 1.38 ms, FOV = 340 mm×340 mm, matrix = 205 × 256, slice thickness = 2 mm, voxel resolution = 1.3 mm×1.3 mm×2.0 mm, acquisition time = 84 s), and then dynamic scanning with 34 consecutive phases was performed using a combination of VIBE with controlled aliasing in parallel imaging results in higher acceleration (CAIPIRINHA), view-sharing time-resolved imaging with interleaved stochastic trajectories (TWIST), and Dixon fat suppression (CAIPIRINHA-Dixon-TWIST-VIBE) sequence. CAIPIRINHA-Dixon-TWIST-VIBE was used for DCE MRI acquisition, and 2 echoes were used for Dixon-based fat suppression. The DCE-MRI scanning time was 17.7s after injection.The scanning parameters of DCE-MRI included TR = 6.4 ms, TE = 3.34 ms, FOV = 340 mm×340 mm, matrix = 288 × 384, voxel resolution was 0.9 mm×0.9 mm×2.0 mm, slice thickness = 2 mm, slices = 80, no slice gap, flip angle 9°, PAT factor 4, partial Fourier factor, temporal resolution 8.7 s (for one phase), and acquisition time 305 s. An intravenous bolus injection of gadopentetate dimeglumine (Bayer Pharma AG ) with a dose of 0.2 ml/kg was power injected at a rate of 3.0 ml/s, followed by a 20 ml saline injection.", "The tumor size was measured on the largest section of the tumor on DCE-MRI images. MRI parameters were measured independently by two radiologists (with more than ten years of experience in breast MRI imaging) who were both blinded to the patients’ clinical history and other examination results. All MRI image data were transferred to a workstation, and BC was identified in DCE-MRI images as the prominent area of enhancement area. DCE-derived parametric maps were automatically generated after motion correction, and registration was performed by using Tissue 4D software (Siemens Healthcare). First, three maximum continuous sections of tumor on DCE-MRI images were selected, and then regions of interest (ROIs) with a minimum area of 10 mm2 were manually drawn on those sections, avoiding visible necrosis, obvious bleeding, vessels, calcifications, and cystic appearing areas. The parameters, including the volume transfer constant (Ktrans, min− 1), extracellular-extravascular volume fraction (Ve, min− 1), reverse reflux rate constant (Kep=Ktrans/Ve), rate of contrast enhancement for inflow (W-in, min− 1), rate of contrast decay for outflow (W-out, min− 1), and the time to peak enhancement after injection (TTP, min) were generated for each voxel defined by the ROIs. The pharmacokinetic parameters were analyzed based on Tofts model of Tissue 4D software (Siemens Healthcare) with a population average arterial input function (AIF) (“intermediate” type). The mean values of the parameters were used for further statistical analysis.", "The statistical analysis was performed with R (version 3.6.0). A two-tailed P < 0.05 was considered to reflect statistical significance. Interobserver agreement for the DCE-MRI parameters between two radiologists was assessed using the intraclass correlation coefficient (ICC). An ICC value of 0.75~0.90 indicates good reliability, while an ICC value ≥ 0.90 indicates excellent reliability [2, 20].\nContinuous variables (age, tumor size, Ktrans, Kep, Ve, W-in, W-out, TTP) with a normal distribution are reported as the means ± standard deviation and were compared with the Student’s t-test, while categorical variables (ER,PR, HER2, Ki-67, tumor grade, molecular subtype, TNM stage, AJCC stage) are summarized as frequencies and percentages and were compared with the chi-squared test (for nominal variables) and the Kruskal-Wallis H test (for ordinal variables) between LVI-positive and LVI-negative groups. The odds ratio (OR) with a 95% confidence interval (CI) for all variables was calculated by using univariate logistic regression analysis to determine the predictors for LVI-positive status. In addition, the combined prediction model for LVI-positive status was constructed by using multivariate logistic regression, and receiver operating characteristic (ROC) curve analysis was performed to evaluate the diagnostic efficiency of the model. The area under the curve (AUC), accuracy, sensitivity, and specificity were calculated, and the optimal cutoff point of the variables was determined according to the largest Youden index.\nFor survival analysis, survival curves of LVI and predicted LVI were generated by using the Kaplan-Meier method and compared by the log-rank test. Multivariate analyses with a Cox proportional hazard model were used to analyze the hazard ratio (HR) with 95% CI for RFS and OS by using the statistically significant variables (P < 0.05) from the univariate Cox analysis.", "Finally, 189 women (mean age: 51.1 years; range:25~95 years) were included in this study. Out of 189 included women, 43 had LVI-positive tumors and 146 had LVI-negative tumors. The characteristics of patients in the LVI-negative and LVI-positive groups are shown in Table E1 in the supplementary materials. Those pertinent features are presented in Table 1. The occurrence rates of lymphatic metastasis was more higher in LVI-positive patients than in LVI-negative patients (P = 0.003). In addition, the LVI-positive patients had higher Kep values than the LVI-negative patients (0.92 ± 0.30 vs. 0.81 ± 0.23, P = 0.012, Fig. 1). No significant differences were found for age, tumor size, ER, PR, HER2, Ki-67 index, tumor grade, molecular subtype, T stage, M stage, AJCC stage, Ktrans, Ve, W-in, W-out, or TTP value between patients with and without LVI (all P > 0.05).\n\nTable 1The characteristics of the patients in the LVI-negative and LVI-positive groupsLVI- (n = 146)LVI+ (n = 43)\nP\nOR (95% CI)\nP\nd\nMaximum diameter (cm)2.89 ± 1.123.09 ± 1.180.312b1.16 (0.86–1.55)0.312Minimum diameter (cm)1.85 ± 0.651.92 ± 0.720.528b1.17 (0.70–1.92)0.526ER0.837aNegative50 (34.25%)14 (32.56%)1NAPositive96 (65.75%)29 (67.44%)1.08 (0.53–2.27)0.837PR0.523aNegative70 (47.95%)23 (53.49%)1NAPositive76 (52.05%)20 (46.51%)0.80 (0.40–1.58)0.523HER20.845aNegative103 (70.55%)31 (72.09%)1NAPositive43 (29.45%)12 (27.91%)0.93 (0.42–1.94)0.845Ki-670.409a≤20%47 (32.19%)11 (25.58%)1NA>20%99 (67.81%)32 (74.42%)1.38 (0.66–3.08)0.410T stage0.109cT014 (9.59%)0 (0.00%)0.0 (0.0-3.48 × 10 13)0.988T146 (31.51%)10 (23.26%)1NAT284 (57.53%)32 (74.42%)1.75 (0.81–4.05)0.167T31 (0.68%)0 (0.00%)0.0 (NA-Inf)0.997T41 (0.68%)1 (2.33%)4.6 (0.17–123.2)0.295 N stage\n0.003\nc\nN091 (62.33%)17 (39.53%)1NAN140 (27.40%)15 (34.88%)2.01 (0.91–4.43)0.083N210 (6.85%)7 (16.28%)3.75 (1.21–11.2)\n0.018\nN35 (3.42%)4 (9.30%)4.28 (0.98–17.85)\n0.044\nParametersKtrans(min− 1)0.21 ± 0.110.22 ± 0.120.766b1.58 (0.07–30.11)0.765Kep(min− 1)0.81 ± 0.230.92 ± 0.30\n0.012\nb\n5.52 (1.42–23.3)\n0.016\n\nV\ne\n0.27 ± 0.130.24 ± 0.090.102b0.12 (0.01–2.15)0.163 W-in (min− 1)0.56 ± 0.220.61 ± 0.240.228b2.50 (0.56–11.17)0.228 W-out (min− 1)-0.01 ± 0.02-0.02 ± 0.020.088b0.00 (0.0-9.35)0.090TTP (min)0.69 ± 0.210.64 ± 0.170.165b0.25 (0.03–1.58)0.167Note. Pa: chi-squared test, Pb: Student’s t-test, Pc: Kruskal-Wallis H test, Pd: univariate analysis. Abbreviations: ER = Estrogen receptor; PR = Progesterone receptor;HER2 = Human epidermal growth factor receptor2; TNBC = Triple negative breast cancer; NA = Not available.\n\nThe characteristics of the patients in the LVI-negative and LVI-positive groups\nNote. Pa: chi-squared test, Pb: Student’s t-test, Pc: Kruskal-Wallis H test, Pd: univariate analysis. Abbreviations: ER = Estrogen receptor; PR = Progesterone receptor;HER2 = Human epidermal growth factor receptor2; TNBC = Triple negative breast cancer; NA = Not available.\n\nFig. 1Representative Kep images from breast cancer patients with (A) lymphovascular invasion (LVI) and without (B) LVI. Red represents high values of Kep, yellow represents intermediate values of Kep, and blue represents low values of Kep. The Kep values of the tumors were 1.05 min− 1 and 0.757 min− 1, respectively\n\nRepresentative Kep images from breast cancer patients with (A) lymphovascular invasion (LVI) and without (B) LVI. Red represents high values of Kep, yellow represents intermediate values of Kep, and blue represents low values of Kep. The Kep values of the tumors were 1.05 min− 1 and 0.757 min− 1, respectively", "The ICC values between two readers for Ktrans, Kep, Ve, W-in, W-out, and TTP were 0.994 (95% CI: 0.992–0.995), 0.858 (95%CI:0.815–0.891), 0.984 (95%CI:0.979–0.988), 0.987 (95%CI: 0.983–0.990), 0.979 (95%CI:0.972–0.984), and 0.961 (95%CI:0.949–0.971), respectively, indicating good or excellent reliability. The coefficient of variation on ICC value is 0.057.", "Univariate logistic regression analysis (Table E1 in the supplementary materials) showed that N2 (OR = 3.75, P = 0.018), N3 (OR = 4.28, P = 0.044), and Kep value (OR = 5.52, P = 0.016) were associated with LVI positivity. ROC curves were generated based on the prediction probability of the regression equation using the above variables (Table 2; Fig. 2). The combined-predicted LVI model that incorporated the N stage and Kep yielded a maximum AUC of 0.669 (0.574–0.765), with a cutoff value of 0.276, accuracy of 0.735, sensitivity of 0.512, and specificity of 0.801. Based on the combined-predicted model results, 51 patients were LVI positive and 138 were LVI negative.\n\nTable 2 Performance of the individualized prediction modelsModelsCutoffAUC(95% CI)AccuracySensitivitySpecificityN0.5000.631 (0.542–0.721)0.6190.6050.623\nK\nep\n0.8080.601 (0.502–0.699)0.5450.6740.507Combined0.2760.669 (0.574–0.765)0.7350.5120.801Note: N and Kep indicate the predicted model based on the N stage and Kep, respectively. Combined indicate the predicted model based on the combination of all above parameters. N mean N2/3 stage versus N0/1 stage.\n\n Performance of the individualized prediction models\nNote: N and Kep indicate the predicted model based on the N stage and Kep, respectively. Combined indicate the predicted model based on the combination of all above parameters. N mean N2/3 stage versus N0/1 stage.\n\nFig. 2Receiver operating characteristic (ROC) curve of the model established by N stage, Kep, and combined-predicted model that incorporated N stage and Kep for the prediction of lymphovascular invasion in breast cancer patients\n\nReceiver operating characteristic (ROC) curve of the model established by N stage, Kep, and combined-predicted model that incorporated N stage and Kep for the prediction of lymphovascular invasion in breast cancer patients", "As of February 28, 2021, all patients had completed RFS follow-up, and the median RFS of all patients was 32.0 (23.0-42.8) months, of which 16 of 189 (8.47%) had a tumor recurrence. Among 16 patient with recurrences, 4 patients had locoregional recurrence and 12 patients had distant metastases. Of note, the recurrence rate was 18.6% (8/43) in patients with LVI and 5.48% (8/146) in patients without LVI, with a significant difference between the two groups (P = 0.016). The median RFS was 31.5 (23.0–42.0) months for patients with LVI and 34.0 (22.0-44.8) months for patients without LVI (log-rank test, P = 0.010, Fig. 3 A). Similar results were observed in the combined-predicted LVI model: The median RFS was 31.8 (23.6–58.5) months for patients with combined-predicted LVI presence and 32.0 (21.8–60.0) months for patients with combined-predicted LVI absence (log-rank test, P = 0.007, Fig. 3B). According to the results of the univariate Cox regression analysis (Table E2 in the supplementary materials), LVI (HR = 3.38, P = 0.015), N2 (HR = 6.13, P = 0.007), and the combined-predicted LVI model (HR = 3.61, P = 0.011) were associated with RFS. Multivariate Cox regression analysis showed that N2 (HR = 4.69, P = 0.026) was independent predictors of disease recurrence. A representative case is provided to show the discriminative ability of the combined-predicted model for predicting LVI and RFS (Fig. 4).\n\nFig. 3Recurrence-free survival (RFS) curves scaled by histologic lymphovascular invasion (LVI) status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis\n\nRecurrence-free survival (RFS) curves scaled by histologic lymphovascular invasion (LVI) status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis\n\nFig. 4 A 59-year-old woman with invasive ductal carcinoma of the right breast. Dynamic contrast-enhanced magnetic resonance imaging (A) showing a solid mass in the upper inner quadrant of the right breast, with nonhomogeneous obvious enhancement, Kep value of 1.09 min− 1 (B), and lymph node metastasis to the right axilla (C). The LVI risk calculated by the combined model which was built based on the combination of N stage and Kep was 70.7%. She was confirmed to be LVI positive by histopathology and had multiple metastatic lesions in the right and left liver lobe on follow-up CT (D) 32.5 months after surgery\n\n A 59-year-old woman with invasive ductal carcinoma of the right breast. Dynamic contrast-enhanced magnetic resonance imaging (A) showing a solid mass in the upper inner quadrant of the right breast, with nonhomogeneous obvious enhancement, Kep value of 1.09 min− 1 (B), and lymph node metastasis to the right axilla (C). The LVI risk calculated by the combined model which was built based on the combination of N stage and Kep was 70.7%. She was confirmed to be LVI positive by histopathology and had multiple metastatic lesions in the right and left liver lobe on follow-up CT (D) 32.5 months after surgery\nWhen 98.4% (186/189) of patients had completed OS follow-up, the median OS of all patients was 43.0 (31.5–56.5) months, and the overall death rate was 3.70% (7/189). The median OS was 41.5 (32.0-55.5) months for patients with LVI and 44.0 (31.0-57.3) months for patients without LVI (log-rank test, P = 0.27, Fig. 5 A), which is consistent with the results of the combined-predicted LVI model: The median OS was 42.8 (32.0-59.5) months for patients with combined-predicted LVI presence and 43.5 (30.8–60.0) months for patients with combined-predicted LVI absence (log-rank test, P = 0.970, Fig. 5B). There were no variables significantly associated with OS in the univariate Cox regression analysis (Table E1 in the supplementary materials, all P > 0.05).\n\nFig. 5Overall survival (OS) curves scaled by histologic LVI status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis\n\nOverall survival (OS) curves scaled by histologic LVI status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, null, null, null, null, null, null, null, null, null, null ]
[ "Background", "Materials and methods", "Basic characteristics", "Definition of recurrence-free survival and follow-up", "MRI examination", "MR image analysis", "Statistical analysis", "Results", "Patient characteristics", "Interobserver agreement", "Predictors of LVI and model development", "Prognostic value of LVI", "Discussion", "Conclusion", "Electronic supplementary material", "" ]
[ "Lymphovascular invasion (LVI), defined as the infiltration of tumor cells into lymphatic or blood vessels at the periphery of invasive carcinoma [1–3], has been widely recognized as a negative prognostic factor in multiple cancers, such as breast cancer (BC), gastric cancer, and rectal cancer [4–7]. Even so, LVI has not been included as an important parameter to consider before adjuvant chemotherapy in the National Comprehensive Cancer Network guidelines for BC due to its hard to determine before adjuvant chemotherapy, which can only be postoperatively diagnosed by histopathology. Thus, more evidence is needed to warrant the application of LVI in clinical decision-making.\nAs a significant preoperative examination for BC patients, dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) are effective tools for evaluating tumor microcirculation [2, 8–13]. The correlation between tumor microcirculation and LVI have been demonstrated and assessed by qualitative parameters of breast MRI in several small cohort studies [14–17]. However, few studies have been published on the prediction of LVI using large samples of quantitative DCE-MRI parameters. In addition, the relationship between the prediction model of LVI and the prognosis of BC is unclear.\nThus, the purpose of this retrospective study was to determine whether quantitative parameters of DCE-MRI can preoperatively predict LVI and to investigate the relationship between the prediction model and survival in terms of disease-specific recurrence and mortality in BC patients to guide clinical decision-making.", "[SUBTITLE] Basic characteristics [SUBSECTION] This retrospective study was approved by our institutional review, and it waived the informed consent requirement. Between January 2016 and March 2019, 460 consecutive women with BC confirmed by postoperative histopathology had undergone preoperative DCE-MRI of the breast. Their medical charts were reviewed. The patient exclusion criteria were shown as follows: (1) patients with previous neoadjuvant treatment; (2) patients with recurrence of BC; (3) patients without obvious lesions on breast MRI; (4) patients with poor quality MRI images; (5) patients with a maximum tumor diameter < 1.0 cm;6) patients with non-mass-like enhancement lesions; and 7) patients with bilateral lesions or multiple lesions.The patient inclusion and exclusion criteria were shown in Figure E1 in the supplementary materials. The study patients are part of a large retrospective DCE-MRI database, of which 165 patients have been reported in a previously published study [18]. A previous study only explored the ability of DCE-MRI to predict BC receptor status and molecular subtypes, rather than LVI and survival outcome [18]. Cinicopathological data, inculding age, tumor size (maximum, minimum, and effective diameter), progesterone receptor (PR), estrogen receptor (ER), human epidermal growth factor receptor2 (HER2), Ki-67, molecular subtypes, tumor grade, tumor node metastasis(TNM) stage, and postoperative treatment methods (radiation therapy, adjuvant endocrine therapy, and adjuvant chemotherapy) were used to predict LVI. The clinicopathological data except receptor status and age were no used in the previous study. In addition, follow-up information were added for survival analysis of LVI.\nEffective tumor diameter was defined as the mean value of the maximum and minimum tumor diameter. The TNM stage was reclassified based on the 8th edition of the American Joint Committee on Cancer (AJCC) staging manual. LVI was defined as tumor cells present within a definite endothelial-lined space (either lymphatic or blood vessel) that was only visible on microscopy. Additionally, the immunohistochemistry (IHC) technique was used to classify the status of ER, PR, HER2, and Ki-67 [19].The standard of cut off values for the positivity of Ki-67, HER-2, PR, and ER were all based on internationally recognized standards [13].\nThis retrospective study was approved by our institutional review, and it waived the informed consent requirement. Between January 2016 and March 2019, 460 consecutive women with BC confirmed by postoperative histopathology had undergone preoperative DCE-MRI of the breast. Their medical charts were reviewed. The patient exclusion criteria were shown as follows: (1) patients with previous neoadjuvant treatment; (2) patients with recurrence of BC; (3) patients without obvious lesions on breast MRI; (4) patients with poor quality MRI images; (5) patients with a maximum tumor diameter < 1.0 cm;6) patients with non-mass-like enhancement lesions; and 7) patients with bilateral lesions or multiple lesions.The patient inclusion and exclusion criteria were shown in Figure E1 in the supplementary materials. The study patients are part of a large retrospective DCE-MRI database, of which 165 patients have been reported in a previously published study [18]. A previous study only explored the ability of DCE-MRI to predict BC receptor status and molecular subtypes, rather than LVI and survival outcome [18]. Cinicopathological data, inculding age, tumor size (maximum, minimum, and effective diameter), progesterone receptor (PR), estrogen receptor (ER), human epidermal growth factor receptor2 (HER2), Ki-67, molecular subtypes, tumor grade, tumor node metastasis(TNM) stage, and postoperative treatment methods (radiation therapy, adjuvant endocrine therapy, and adjuvant chemotherapy) were used to predict LVI. The clinicopathological data except receptor status and age were no used in the previous study. In addition, follow-up information were added for survival analysis of LVI.\nEffective tumor diameter was defined as the mean value of the maximum and minimum tumor diameter. The TNM stage was reclassified based on the 8th edition of the American Joint Committee on Cancer (AJCC) staging manual. LVI was defined as tumor cells present within a definite endothelial-lined space (either lymphatic or blood vessel) that was only visible on microscopy. Additionally, the immunohistochemistry (IHC) technique was used to classify the status of ER, PR, HER2, and Ki-67 [19].The standard of cut off values for the positivity of Ki-67, HER-2, PR, and ER were all based on internationally recognized standards [13].\n[SUBTITLE] Definition of recurrence-free survival and follow-up [SUBSECTION] According to the follow-up protocol of our hospital, all patients were postoperatively followed up with chest and abdominal CT, and breast mammography every 12 months. Additionally, the patients with mastectomy were postoperatively followed up with breast, abdominal, gynecological, and superficial lymph node ultrasound every 3 months for the first two years. Of the contralateral breast usually followed up with breast mammography and ultrasound. The patients with breast-conserving surgery were postoperatively followed up with breast MRI every 12 months.\nRecurrence-free survival (RFS) was calculated in months from the date of surgery to the first date of local recurrence, distant metastasis, February 28, 2021, or death, whichever came first. Overall survival (OS) was calculated in months from the date of surgery to the date of death or on February 28, 2021, whichever came first.\nAccording to the follow-up protocol of our hospital, all patients were postoperatively followed up with chest and abdominal CT, and breast mammography every 12 months. Additionally, the patients with mastectomy were postoperatively followed up with breast, abdominal, gynecological, and superficial lymph node ultrasound every 3 months for the first two years. Of the contralateral breast usually followed up with breast mammography and ultrasound. The patients with breast-conserving surgery were postoperatively followed up with breast MRI every 12 months.\nRecurrence-free survival (RFS) was calculated in months from the date of surgery to the first date of local recurrence, distant metastasis, February 28, 2021, or death, whichever came first. Overall survival (OS) was calculated in months from the date of surgery to the date of death or on February 28, 2021, whichever came first.\n[SUBTITLE] MRI examination [SUBSECTION] Breast MRI examinations were performed on a 3.0 T MR scanner (Magnetom Skyra, Siemens Healthcare, Erlangen, Germany) using a 16-channel bilateral breast coil with the patient in the prone position. Before dynamic scanning, T1-weighted volume interpolated breath-hold examination (VIBE) was first performed at two different flip angles 2° and 15° to calculate the T1-mapping images (TR = 3.78 ms, TE = 1.38 ms, FOV = 340 mm×340 mm, matrix = 205 × 256, slice thickness = 2 mm, voxel resolution = 1.3 mm×1.3 mm×2.0 mm, acquisition time = 84 s), and then dynamic scanning with 34 consecutive phases was performed using a combination of VIBE with controlled aliasing in parallel imaging results in higher acceleration (CAIPIRINHA), view-sharing time-resolved imaging with interleaved stochastic trajectories (TWIST), and Dixon fat suppression (CAIPIRINHA-Dixon-TWIST-VIBE) sequence. CAIPIRINHA-Dixon-TWIST-VIBE was used for DCE MRI acquisition, and 2 echoes were used for Dixon-based fat suppression. The DCE-MRI scanning time was 17.7s after injection.The scanning parameters of DCE-MRI included TR = 6.4 ms, TE = 3.34 ms, FOV = 340 mm×340 mm, matrix = 288 × 384, voxel resolution was 0.9 mm×0.9 mm×2.0 mm, slice thickness = 2 mm, slices = 80, no slice gap, flip angle 9°, PAT factor 4, partial Fourier factor, temporal resolution 8.7 s (for one phase), and acquisition time 305 s. An intravenous bolus injection of gadopentetate dimeglumine (Bayer Pharma AG ) with a dose of 0.2 ml/kg was power injected at a rate of 3.0 ml/s, followed by a 20 ml saline injection.\nBreast MRI examinations were performed on a 3.0 T MR scanner (Magnetom Skyra, Siemens Healthcare, Erlangen, Germany) using a 16-channel bilateral breast coil with the patient in the prone position. Before dynamic scanning, T1-weighted volume interpolated breath-hold examination (VIBE) was first performed at two different flip angles 2° and 15° to calculate the T1-mapping images (TR = 3.78 ms, TE = 1.38 ms, FOV = 340 mm×340 mm, matrix = 205 × 256, slice thickness = 2 mm, voxel resolution = 1.3 mm×1.3 mm×2.0 mm, acquisition time = 84 s), and then dynamic scanning with 34 consecutive phases was performed using a combination of VIBE with controlled aliasing in parallel imaging results in higher acceleration (CAIPIRINHA), view-sharing time-resolved imaging with interleaved stochastic trajectories (TWIST), and Dixon fat suppression (CAIPIRINHA-Dixon-TWIST-VIBE) sequence. CAIPIRINHA-Dixon-TWIST-VIBE was used for DCE MRI acquisition, and 2 echoes were used for Dixon-based fat suppression. The DCE-MRI scanning time was 17.7s after injection.The scanning parameters of DCE-MRI included TR = 6.4 ms, TE = 3.34 ms, FOV = 340 mm×340 mm, matrix = 288 × 384, voxel resolution was 0.9 mm×0.9 mm×2.0 mm, slice thickness = 2 mm, slices = 80, no slice gap, flip angle 9°, PAT factor 4, partial Fourier factor, temporal resolution 8.7 s (for one phase), and acquisition time 305 s. An intravenous bolus injection of gadopentetate dimeglumine (Bayer Pharma AG ) with a dose of 0.2 ml/kg was power injected at a rate of 3.0 ml/s, followed by a 20 ml saline injection.\n[SUBTITLE] MR image analysis [SUBSECTION] The tumor size was measured on the largest section of the tumor on DCE-MRI images. MRI parameters were measured independently by two radiologists (with more than ten years of experience in breast MRI imaging) who were both blinded to the patients’ clinical history and other examination results. All MRI image data were transferred to a workstation, and BC was identified in DCE-MRI images as the prominent area of enhancement area. DCE-derived parametric maps were automatically generated after motion correction, and registration was performed by using Tissue 4D software (Siemens Healthcare). First, three maximum continuous sections of tumor on DCE-MRI images were selected, and then regions of interest (ROIs) with a minimum area of 10 mm2 were manually drawn on those sections, avoiding visible necrosis, obvious bleeding, vessels, calcifications, and cystic appearing areas. The parameters, including the volume transfer constant (Ktrans, min− 1), extracellular-extravascular volume fraction (Ve, min− 1), reverse reflux rate constant (Kep=Ktrans/Ve), rate of contrast enhancement for inflow (W-in, min− 1), rate of contrast decay for outflow (W-out, min− 1), and the time to peak enhancement after injection (TTP, min) were generated for each voxel defined by the ROIs. The pharmacokinetic parameters were analyzed based on Tofts model of Tissue 4D software (Siemens Healthcare) with a population average arterial input function (AIF) (“intermediate” type). The mean values of the parameters were used for further statistical analysis.\nThe tumor size was measured on the largest section of the tumor on DCE-MRI images. MRI parameters were measured independently by two radiologists (with more than ten years of experience in breast MRI imaging) who were both blinded to the patients’ clinical history and other examination results. All MRI image data were transferred to a workstation, and BC was identified in DCE-MRI images as the prominent area of enhancement area. DCE-derived parametric maps were automatically generated after motion correction, and registration was performed by using Tissue 4D software (Siemens Healthcare). First, three maximum continuous sections of tumor on DCE-MRI images were selected, and then regions of interest (ROIs) with a minimum area of 10 mm2 were manually drawn on those sections, avoiding visible necrosis, obvious bleeding, vessels, calcifications, and cystic appearing areas. The parameters, including the volume transfer constant (Ktrans, min− 1), extracellular-extravascular volume fraction (Ve, min− 1), reverse reflux rate constant (Kep=Ktrans/Ve), rate of contrast enhancement for inflow (W-in, min− 1), rate of contrast decay for outflow (W-out, min− 1), and the time to peak enhancement after injection (TTP, min) were generated for each voxel defined by the ROIs. The pharmacokinetic parameters were analyzed based on Tofts model of Tissue 4D software (Siemens Healthcare) with a population average arterial input function (AIF) (“intermediate” type). The mean values of the parameters were used for further statistical analysis.\n[SUBTITLE] Statistical analysis [SUBSECTION] The statistical analysis was performed with R (version 3.6.0). A two-tailed P < 0.05 was considered to reflect statistical significance. Interobserver agreement for the DCE-MRI parameters between two radiologists was assessed using the intraclass correlation coefficient (ICC). An ICC value of 0.75~0.90 indicates good reliability, while an ICC value ≥ 0.90 indicates excellent reliability [2, 20].\nContinuous variables (age, tumor size, Ktrans, Kep, Ve, W-in, W-out, TTP) with a normal distribution are reported as the means ± standard deviation and were compared with the Student’s t-test, while categorical variables (ER,PR, HER2, Ki-67, tumor grade, molecular subtype, TNM stage, AJCC stage) are summarized as frequencies and percentages and were compared with the chi-squared test (for nominal variables) and the Kruskal-Wallis H test (for ordinal variables) between LVI-positive and LVI-negative groups. The odds ratio (OR) with a 95% confidence interval (CI) for all variables was calculated by using univariate logistic regression analysis to determine the predictors for LVI-positive status. In addition, the combined prediction model for LVI-positive status was constructed by using multivariate logistic regression, and receiver operating characteristic (ROC) curve analysis was performed to evaluate the diagnostic efficiency of the model. The area under the curve (AUC), accuracy, sensitivity, and specificity were calculated, and the optimal cutoff point of the variables was determined according to the largest Youden index.\nFor survival analysis, survival curves of LVI and predicted LVI were generated by using the Kaplan-Meier method and compared by the log-rank test. Multivariate analyses with a Cox proportional hazard model were used to analyze the hazard ratio (HR) with 95% CI for RFS and OS by using the statistically significant variables (P < 0.05) from the univariate Cox analysis.\nThe statistical analysis was performed with R (version 3.6.0). A two-tailed P < 0.05 was considered to reflect statistical significance. Interobserver agreement for the DCE-MRI parameters between two radiologists was assessed using the intraclass correlation coefficient (ICC). An ICC value of 0.75~0.90 indicates good reliability, while an ICC value ≥ 0.90 indicates excellent reliability [2, 20].\nContinuous variables (age, tumor size, Ktrans, Kep, Ve, W-in, W-out, TTP) with a normal distribution are reported as the means ± standard deviation and were compared with the Student’s t-test, while categorical variables (ER,PR, HER2, Ki-67, tumor grade, molecular subtype, TNM stage, AJCC stage) are summarized as frequencies and percentages and were compared with the chi-squared test (for nominal variables) and the Kruskal-Wallis H test (for ordinal variables) between LVI-positive and LVI-negative groups. The odds ratio (OR) with a 95% confidence interval (CI) for all variables was calculated by using univariate logistic regression analysis to determine the predictors for LVI-positive status. In addition, the combined prediction model for LVI-positive status was constructed by using multivariate logistic regression, and receiver operating characteristic (ROC) curve analysis was performed to evaluate the diagnostic efficiency of the model. The area under the curve (AUC), accuracy, sensitivity, and specificity were calculated, and the optimal cutoff point of the variables was determined according to the largest Youden index.\nFor survival analysis, survival curves of LVI and predicted LVI were generated by using the Kaplan-Meier method and compared by the log-rank test. Multivariate analyses with a Cox proportional hazard model were used to analyze the hazard ratio (HR) with 95% CI for RFS and OS by using the statistically significant variables (P < 0.05) from the univariate Cox analysis.", "This retrospective study was approved by our institutional review, and it waived the informed consent requirement. Between January 2016 and March 2019, 460 consecutive women with BC confirmed by postoperative histopathology had undergone preoperative DCE-MRI of the breast. Their medical charts were reviewed. The patient exclusion criteria were shown as follows: (1) patients with previous neoadjuvant treatment; (2) patients with recurrence of BC; (3) patients without obvious lesions on breast MRI; (4) patients with poor quality MRI images; (5) patients with a maximum tumor diameter < 1.0 cm;6) patients with non-mass-like enhancement lesions; and 7) patients with bilateral lesions or multiple lesions.The patient inclusion and exclusion criteria were shown in Figure E1 in the supplementary materials. The study patients are part of a large retrospective DCE-MRI database, of which 165 patients have been reported in a previously published study [18]. A previous study only explored the ability of DCE-MRI to predict BC receptor status and molecular subtypes, rather than LVI and survival outcome [18]. Cinicopathological data, inculding age, tumor size (maximum, minimum, and effective diameter), progesterone receptor (PR), estrogen receptor (ER), human epidermal growth factor receptor2 (HER2), Ki-67, molecular subtypes, tumor grade, tumor node metastasis(TNM) stage, and postoperative treatment methods (radiation therapy, adjuvant endocrine therapy, and adjuvant chemotherapy) were used to predict LVI. The clinicopathological data except receptor status and age were no used in the previous study. In addition, follow-up information were added for survival analysis of LVI.\nEffective tumor diameter was defined as the mean value of the maximum and minimum tumor diameter. The TNM stage was reclassified based on the 8th edition of the American Joint Committee on Cancer (AJCC) staging manual. LVI was defined as tumor cells present within a definite endothelial-lined space (either lymphatic or blood vessel) that was only visible on microscopy. Additionally, the immunohistochemistry (IHC) technique was used to classify the status of ER, PR, HER2, and Ki-67 [19].The standard of cut off values for the positivity of Ki-67, HER-2, PR, and ER were all based on internationally recognized standards [13].", "According to the follow-up protocol of our hospital, all patients were postoperatively followed up with chest and abdominal CT, and breast mammography every 12 months. Additionally, the patients with mastectomy were postoperatively followed up with breast, abdominal, gynecological, and superficial lymph node ultrasound every 3 months for the first two years. Of the contralateral breast usually followed up with breast mammography and ultrasound. The patients with breast-conserving surgery were postoperatively followed up with breast MRI every 12 months.\nRecurrence-free survival (RFS) was calculated in months from the date of surgery to the first date of local recurrence, distant metastasis, February 28, 2021, or death, whichever came first. Overall survival (OS) was calculated in months from the date of surgery to the date of death or on February 28, 2021, whichever came first.", "Breast MRI examinations were performed on a 3.0 T MR scanner (Magnetom Skyra, Siemens Healthcare, Erlangen, Germany) using a 16-channel bilateral breast coil with the patient in the prone position. Before dynamic scanning, T1-weighted volume interpolated breath-hold examination (VIBE) was first performed at two different flip angles 2° and 15° to calculate the T1-mapping images (TR = 3.78 ms, TE = 1.38 ms, FOV = 340 mm×340 mm, matrix = 205 × 256, slice thickness = 2 mm, voxel resolution = 1.3 mm×1.3 mm×2.0 mm, acquisition time = 84 s), and then dynamic scanning with 34 consecutive phases was performed using a combination of VIBE with controlled aliasing in parallel imaging results in higher acceleration (CAIPIRINHA), view-sharing time-resolved imaging with interleaved stochastic trajectories (TWIST), and Dixon fat suppression (CAIPIRINHA-Dixon-TWIST-VIBE) sequence. CAIPIRINHA-Dixon-TWIST-VIBE was used for DCE MRI acquisition, and 2 echoes were used for Dixon-based fat suppression. The DCE-MRI scanning time was 17.7s after injection.The scanning parameters of DCE-MRI included TR = 6.4 ms, TE = 3.34 ms, FOV = 340 mm×340 mm, matrix = 288 × 384, voxel resolution was 0.9 mm×0.9 mm×2.0 mm, slice thickness = 2 mm, slices = 80, no slice gap, flip angle 9°, PAT factor 4, partial Fourier factor, temporal resolution 8.7 s (for one phase), and acquisition time 305 s. An intravenous bolus injection of gadopentetate dimeglumine (Bayer Pharma AG ) with a dose of 0.2 ml/kg was power injected at a rate of 3.0 ml/s, followed by a 20 ml saline injection.", "The tumor size was measured on the largest section of the tumor on DCE-MRI images. MRI parameters were measured independently by two radiologists (with more than ten years of experience in breast MRI imaging) who were both blinded to the patients’ clinical history and other examination results. All MRI image data were transferred to a workstation, and BC was identified in DCE-MRI images as the prominent area of enhancement area. DCE-derived parametric maps were automatically generated after motion correction, and registration was performed by using Tissue 4D software (Siemens Healthcare). First, three maximum continuous sections of tumor on DCE-MRI images were selected, and then regions of interest (ROIs) with a minimum area of 10 mm2 were manually drawn on those sections, avoiding visible necrosis, obvious bleeding, vessels, calcifications, and cystic appearing areas. The parameters, including the volume transfer constant (Ktrans, min− 1), extracellular-extravascular volume fraction (Ve, min− 1), reverse reflux rate constant (Kep=Ktrans/Ve), rate of contrast enhancement for inflow (W-in, min− 1), rate of contrast decay for outflow (W-out, min− 1), and the time to peak enhancement after injection (TTP, min) were generated for each voxel defined by the ROIs. The pharmacokinetic parameters were analyzed based on Tofts model of Tissue 4D software (Siemens Healthcare) with a population average arterial input function (AIF) (“intermediate” type). The mean values of the parameters were used for further statistical analysis.", "The statistical analysis was performed with R (version 3.6.0). A two-tailed P < 0.05 was considered to reflect statistical significance. Interobserver agreement for the DCE-MRI parameters between two radiologists was assessed using the intraclass correlation coefficient (ICC). An ICC value of 0.75~0.90 indicates good reliability, while an ICC value ≥ 0.90 indicates excellent reliability [2, 20].\nContinuous variables (age, tumor size, Ktrans, Kep, Ve, W-in, W-out, TTP) with a normal distribution are reported as the means ± standard deviation and were compared with the Student’s t-test, while categorical variables (ER,PR, HER2, Ki-67, tumor grade, molecular subtype, TNM stage, AJCC stage) are summarized as frequencies and percentages and were compared with the chi-squared test (for nominal variables) and the Kruskal-Wallis H test (for ordinal variables) between LVI-positive and LVI-negative groups. The odds ratio (OR) with a 95% confidence interval (CI) for all variables was calculated by using univariate logistic regression analysis to determine the predictors for LVI-positive status. In addition, the combined prediction model for LVI-positive status was constructed by using multivariate logistic regression, and receiver operating characteristic (ROC) curve analysis was performed to evaluate the diagnostic efficiency of the model. The area under the curve (AUC), accuracy, sensitivity, and specificity were calculated, and the optimal cutoff point of the variables was determined according to the largest Youden index.\nFor survival analysis, survival curves of LVI and predicted LVI were generated by using the Kaplan-Meier method and compared by the log-rank test. Multivariate analyses with a Cox proportional hazard model were used to analyze the hazard ratio (HR) with 95% CI for RFS and OS by using the statistically significant variables (P < 0.05) from the univariate Cox analysis.", "[SUBTITLE] Patient characteristics [SUBSECTION] Finally, 189 women (mean age: 51.1 years; range:25~95 years) were included in this study. Out of 189 included women, 43 had LVI-positive tumors and 146 had LVI-negative tumors. The characteristics of patients in the LVI-negative and LVI-positive groups are shown in Table E1 in the supplementary materials. Those pertinent features are presented in Table 1. The occurrence rates of lymphatic metastasis was more higher in LVI-positive patients than in LVI-negative patients (P = 0.003). In addition, the LVI-positive patients had higher Kep values than the LVI-negative patients (0.92 ± 0.30 vs. 0.81 ± 0.23, P = 0.012, Fig. 1). No significant differences were found for age, tumor size, ER, PR, HER2, Ki-67 index, tumor grade, molecular subtype, T stage, M stage, AJCC stage, Ktrans, Ve, W-in, W-out, or TTP value between patients with and without LVI (all P > 0.05).\n\nTable 1The characteristics of the patients in the LVI-negative and LVI-positive groupsLVI- (n = 146)LVI+ (n = 43)\nP\nOR (95% CI)\nP\nd\nMaximum diameter (cm)2.89 ± 1.123.09 ± 1.180.312b1.16 (0.86–1.55)0.312Minimum diameter (cm)1.85 ± 0.651.92 ± 0.720.528b1.17 (0.70–1.92)0.526ER0.837aNegative50 (34.25%)14 (32.56%)1NAPositive96 (65.75%)29 (67.44%)1.08 (0.53–2.27)0.837PR0.523aNegative70 (47.95%)23 (53.49%)1NAPositive76 (52.05%)20 (46.51%)0.80 (0.40–1.58)0.523HER20.845aNegative103 (70.55%)31 (72.09%)1NAPositive43 (29.45%)12 (27.91%)0.93 (0.42–1.94)0.845Ki-670.409a≤20%47 (32.19%)11 (25.58%)1NA>20%99 (67.81%)32 (74.42%)1.38 (0.66–3.08)0.410T stage0.109cT014 (9.59%)0 (0.00%)0.0 (0.0-3.48 × 10 13)0.988T146 (31.51%)10 (23.26%)1NAT284 (57.53%)32 (74.42%)1.75 (0.81–4.05)0.167T31 (0.68%)0 (0.00%)0.0 (NA-Inf)0.997T41 (0.68%)1 (2.33%)4.6 (0.17–123.2)0.295 N stage\n0.003\nc\nN091 (62.33%)17 (39.53%)1NAN140 (27.40%)15 (34.88%)2.01 (0.91–4.43)0.083N210 (6.85%)7 (16.28%)3.75 (1.21–11.2)\n0.018\nN35 (3.42%)4 (9.30%)4.28 (0.98–17.85)\n0.044\nParametersKtrans(min− 1)0.21 ± 0.110.22 ± 0.120.766b1.58 (0.07–30.11)0.765Kep(min− 1)0.81 ± 0.230.92 ± 0.30\n0.012\nb\n5.52 (1.42–23.3)\n0.016\n\nV\ne\n0.27 ± 0.130.24 ± 0.090.102b0.12 (0.01–2.15)0.163 W-in (min− 1)0.56 ± 0.220.61 ± 0.240.228b2.50 (0.56–11.17)0.228 W-out (min− 1)-0.01 ± 0.02-0.02 ± 0.020.088b0.00 (0.0-9.35)0.090TTP (min)0.69 ± 0.210.64 ± 0.170.165b0.25 (0.03–1.58)0.167Note. Pa: chi-squared test, Pb: Student’s t-test, Pc: Kruskal-Wallis H test, Pd: univariate analysis. Abbreviations: ER = Estrogen receptor; PR = Progesterone receptor;HER2 = Human epidermal growth factor receptor2; TNBC = Triple negative breast cancer; NA = Not available.\n\nThe characteristics of the patients in the LVI-negative and LVI-positive groups\nNote. Pa: chi-squared test, Pb: Student’s t-test, Pc: Kruskal-Wallis H test, Pd: univariate analysis. Abbreviations: ER = Estrogen receptor; PR = Progesterone receptor;HER2 = Human epidermal growth factor receptor2; TNBC = Triple negative breast cancer; NA = Not available.\n\nFig. 1Representative Kep images from breast cancer patients with (A) lymphovascular invasion (LVI) and without (B) LVI. Red represents high values of Kep, yellow represents intermediate values of Kep, and blue represents low values of Kep. The Kep values of the tumors were 1.05 min− 1 and 0.757 min− 1, respectively\n\nRepresentative Kep images from breast cancer patients with (A) lymphovascular invasion (LVI) and without (B) LVI. Red represents high values of Kep, yellow represents intermediate values of Kep, and blue represents low values of Kep. The Kep values of the tumors were 1.05 min− 1 and 0.757 min− 1, respectively\nFinally, 189 women (mean age: 51.1 years; range:25~95 years) were included in this study. Out of 189 included women, 43 had LVI-positive tumors and 146 had LVI-negative tumors. The characteristics of patients in the LVI-negative and LVI-positive groups are shown in Table E1 in the supplementary materials. Those pertinent features are presented in Table 1. The occurrence rates of lymphatic metastasis was more higher in LVI-positive patients than in LVI-negative patients (P = 0.003). In addition, the LVI-positive patients had higher Kep values than the LVI-negative patients (0.92 ± 0.30 vs. 0.81 ± 0.23, P = 0.012, Fig. 1). No significant differences were found for age, tumor size, ER, PR, HER2, Ki-67 index, tumor grade, molecular subtype, T stage, M stage, AJCC stage, Ktrans, Ve, W-in, W-out, or TTP value between patients with and without LVI (all P > 0.05).\n\nTable 1The characteristics of the patients in the LVI-negative and LVI-positive groupsLVI- (n = 146)LVI+ (n = 43)\nP\nOR (95% CI)\nP\nd\nMaximum diameter (cm)2.89 ± 1.123.09 ± 1.180.312b1.16 (0.86–1.55)0.312Minimum diameter (cm)1.85 ± 0.651.92 ± 0.720.528b1.17 (0.70–1.92)0.526ER0.837aNegative50 (34.25%)14 (32.56%)1NAPositive96 (65.75%)29 (67.44%)1.08 (0.53–2.27)0.837PR0.523aNegative70 (47.95%)23 (53.49%)1NAPositive76 (52.05%)20 (46.51%)0.80 (0.40–1.58)0.523HER20.845aNegative103 (70.55%)31 (72.09%)1NAPositive43 (29.45%)12 (27.91%)0.93 (0.42–1.94)0.845Ki-670.409a≤20%47 (32.19%)11 (25.58%)1NA>20%99 (67.81%)32 (74.42%)1.38 (0.66–3.08)0.410T stage0.109cT014 (9.59%)0 (0.00%)0.0 (0.0-3.48 × 10 13)0.988T146 (31.51%)10 (23.26%)1NAT284 (57.53%)32 (74.42%)1.75 (0.81–4.05)0.167T31 (0.68%)0 (0.00%)0.0 (NA-Inf)0.997T41 (0.68%)1 (2.33%)4.6 (0.17–123.2)0.295 N stage\n0.003\nc\nN091 (62.33%)17 (39.53%)1NAN140 (27.40%)15 (34.88%)2.01 (0.91–4.43)0.083N210 (6.85%)7 (16.28%)3.75 (1.21–11.2)\n0.018\nN35 (3.42%)4 (9.30%)4.28 (0.98–17.85)\n0.044\nParametersKtrans(min− 1)0.21 ± 0.110.22 ± 0.120.766b1.58 (0.07–30.11)0.765Kep(min− 1)0.81 ± 0.230.92 ± 0.30\n0.012\nb\n5.52 (1.42–23.3)\n0.016\n\nV\ne\n0.27 ± 0.130.24 ± 0.090.102b0.12 (0.01–2.15)0.163 W-in (min− 1)0.56 ± 0.220.61 ± 0.240.228b2.50 (0.56–11.17)0.228 W-out (min− 1)-0.01 ± 0.02-0.02 ± 0.020.088b0.00 (0.0-9.35)0.090TTP (min)0.69 ± 0.210.64 ± 0.170.165b0.25 (0.03–1.58)0.167Note. Pa: chi-squared test, Pb: Student’s t-test, Pc: Kruskal-Wallis H test, Pd: univariate analysis. Abbreviations: ER = Estrogen receptor; PR = Progesterone receptor;HER2 = Human epidermal growth factor receptor2; TNBC = Triple negative breast cancer; NA = Not available.\n\nThe characteristics of the patients in the LVI-negative and LVI-positive groups\nNote. Pa: chi-squared test, Pb: Student’s t-test, Pc: Kruskal-Wallis H test, Pd: univariate analysis. Abbreviations: ER = Estrogen receptor; PR = Progesterone receptor;HER2 = Human epidermal growth factor receptor2; TNBC = Triple negative breast cancer; NA = Not available.\n\nFig. 1Representative Kep images from breast cancer patients with (A) lymphovascular invasion (LVI) and without (B) LVI. Red represents high values of Kep, yellow represents intermediate values of Kep, and blue represents low values of Kep. The Kep values of the tumors were 1.05 min− 1 and 0.757 min− 1, respectively\n\nRepresentative Kep images from breast cancer patients with (A) lymphovascular invasion (LVI) and without (B) LVI. Red represents high values of Kep, yellow represents intermediate values of Kep, and blue represents low values of Kep. The Kep values of the tumors were 1.05 min− 1 and 0.757 min− 1, respectively\n[SUBTITLE] Interobserver agreement [SUBSECTION] The ICC values between two readers for Ktrans, Kep, Ve, W-in, W-out, and TTP were 0.994 (95% CI: 0.992–0.995), 0.858 (95%CI:0.815–0.891), 0.984 (95%CI:0.979–0.988), 0.987 (95%CI: 0.983–0.990), 0.979 (95%CI:0.972–0.984), and 0.961 (95%CI:0.949–0.971), respectively, indicating good or excellent reliability. The coefficient of variation on ICC value is 0.057.\nThe ICC values between two readers for Ktrans, Kep, Ve, W-in, W-out, and TTP were 0.994 (95% CI: 0.992–0.995), 0.858 (95%CI:0.815–0.891), 0.984 (95%CI:0.979–0.988), 0.987 (95%CI: 0.983–0.990), 0.979 (95%CI:0.972–0.984), and 0.961 (95%CI:0.949–0.971), respectively, indicating good or excellent reliability. The coefficient of variation on ICC value is 0.057.\n[SUBTITLE] Predictors of LVI and model development [SUBSECTION] Univariate logistic regression analysis (Table E1 in the supplementary materials) showed that N2 (OR = 3.75, P = 0.018), N3 (OR = 4.28, P = 0.044), and Kep value (OR = 5.52, P = 0.016) were associated with LVI positivity. ROC curves were generated based on the prediction probability of the regression equation using the above variables (Table 2; Fig. 2). The combined-predicted LVI model that incorporated the N stage and Kep yielded a maximum AUC of 0.669 (0.574–0.765), with a cutoff value of 0.276, accuracy of 0.735, sensitivity of 0.512, and specificity of 0.801. Based on the combined-predicted model results, 51 patients were LVI positive and 138 were LVI negative.\n\nTable 2 Performance of the individualized prediction modelsModelsCutoffAUC(95% CI)AccuracySensitivitySpecificityN0.5000.631 (0.542–0.721)0.6190.6050.623\nK\nep\n0.8080.601 (0.502–0.699)0.5450.6740.507Combined0.2760.669 (0.574–0.765)0.7350.5120.801Note: N and Kep indicate the predicted model based on the N stage and Kep, respectively. Combined indicate the predicted model based on the combination of all above parameters. N mean N2/3 stage versus N0/1 stage.\n\n Performance of the individualized prediction models\nNote: N and Kep indicate the predicted model based on the N stage and Kep, respectively. Combined indicate the predicted model based on the combination of all above parameters. N mean N2/3 stage versus N0/1 stage.\n\nFig. 2Receiver operating characteristic (ROC) curve of the model established by N stage, Kep, and combined-predicted model that incorporated N stage and Kep for the prediction of lymphovascular invasion in breast cancer patients\n\nReceiver operating characteristic (ROC) curve of the model established by N stage, Kep, and combined-predicted model that incorporated N stage and Kep for the prediction of lymphovascular invasion in breast cancer patients\nUnivariate logistic regression analysis (Table E1 in the supplementary materials) showed that N2 (OR = 3.75, P = 0.018), N3 (OR = 4.28, P = 0.044), and Kep value (OR = 5.52, P = 0.016) were associated with LVI positivity. ROC curves were generated based on the prediction probability of the regression equation using the above variables (Table 2; Fig. 2). The combined-predicted LVI model that incorporated the N stage and Kep yielded a maximum AUC of 0.669 (0.574–0.765), with a cutoff value of 0.276, accuracy of 0.735, sensitivity of 0.512, and specificity of 0.801. Based on the combined-predicted model results, 51 patients were LVI positive and 138 were LVI negative.\n\nTable 2 Performance of the individualized prediction modelsModelsCutoffAUC(95% CI)AccuracySensitivitySpecificityN0.5000.631 (0.542–0.721)0.6190.6050.623\nK\nep\n0.8080.601 (0.502–0.699)0.5450.6740.507Combined0.2760.669 (0.574–0.765)0.7350.5120.801Note: N and Kep indicate the predicted model based on the N stage and Kep, respectively. Combined indicate the predicted model based on the combination of all above parameters. N mean N2/3 stage versus N0/1 stage.\n\n Performance of the individualized prediction models\nNote: N and Kep indicate the predicted model based on the N stage and Kep, respectively. Combined indicate the predicted model based on the combination of all above parameters. N mean N2/3 stage versus N0/1 stage.\n\nFig. 2Receiver operating characteristic (ROC) curve of the model established by N stage, Kep, and combined-predicted model that incorporated N stage and Kep for the prediction of lymphovascular invasion in breast cancer patients\n\nReceiver operating characteristic (ROC) curve of the model established by N stage, Kep, and combined-predicted model that incorporated N stage and Kep for the prediction of lymphovascular invasion in breast cancer patients\n[SUBTITLE] Prognostic value of LVI [SUBSECTION] As of February 28, 2021, all patients had completed RFS follow-up, and the median RFS of all patients was 32.0 (23.0-42.8) months, of which 16 of 189 (8.47%) had a tumor recurrence. Among 16 patient with recurrences, 4 patients had locoregional recurrence and 12 patients had distant metastases. Of note, the recurrence rate was 18.6% (8/43) in patients with LVI and 5.48% (8/146) in patients without LVI, with a significant difference between the two groups (P = 0.016). The median RFS was 31.5 (23.0–42.0) months for patients with LVI and 34.0 (22.0-44.8) months for patients without LVI (log-rank test, P = 0.010, Fig. 3 A). Similar results were observed in the combined-predicted LVI model: The median RFS was 31.8 (23.6–58.5) months for patients with combined-predicted LVI presence and 32.0 (21.8–60.0) months for patients with combined-predicted LVI absence (log-rank test, P = 0.007, Fig. 3B). According to the results of the univariate Cox regression analysis (Table E2 in the supplementary materials), LVI (HR = 3.38, P = 0.015), N2 (HR = 6.13, P = 0.007), and the combined-predicted LVI model (HR = 3.61, P = 0.011) were associated with RFS. Multivariate Cox regression analysis showed that N2 (HR = 4.69, P = 0.026) was independent predictors of disease recurrence. A representative case is provided to show the discriminative ability of the combined-predicted model for predicting LVI and RFS (Fig. 4).\n\nFig. 3Recurrence-free survival (RFS) curves scaled by histologic lymphovascular invasion (LVI) status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis\n\nRecurrence-free survival (RFS) curves scaled by histologic lymphovascular invasion (LVI) status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis\n\nFig. 4 A 59-year-old woman with invasive ductal carcinoma of the right breast. Dynamic contrast-enhanced magnetic resonance imaging (A) showing a solid mass in the upper inner quadrant of the right breast, with nonhomogeneous obvious enhancement, Kep value of 1.09 min− 1 (B), and lymph node metastasis to the right axilla (C). The LVI risk calculated by the combined model which was built based on the combination of N stage and Kep was 70.7%. She was confirmed to be LVI positive by histopathology and had multiple metastatic lesions in the right and left liver lobe on follow-up CT (D) 32.5 months after surgery\n\n A 59-year-old woman with invasive ductal carcinoma of the right breast. Dynamic contrast-enhanced magnetic resonance imaging (A) showing a solid mass in the upper inner quadrant of the right breast, with nonhomogeneous obvious enhancement, Kep value of 1.09 min− 1 (B), and lymph node metastasis to the right axilla (C). The LVI risk calculated by the combined model which was built based on the combination of N stage and Kep was 70.7%. She was confirmed to be LVI positive by histopathology and had multiple metastatic lesions in the right and left liver lobe on follow-up CT (D) 32.5 months after surgery\nWhen 98.4% (186/189) of patients had completed OS follow-up, the median OS of all patients was 43.0 (31.5–56.5) months, and the overall death rate was 3.70% (7/189). The median OS was 41.5 (32.0-55.5) months for patients with LVI and 44.0 (31.0-57.3) months for patients without LVI (log-rank test, P = 0.27, Fig. 5 A), which is consistent with the results of the combined-predicted LVI model: The median OS was 42.8 (32.0-59.5) months for patients with combined-predicted LVI presence and 43.5 (30.8–60.0) months for patients with combined-predicted LVI absence (log-rank test, P = 0.970, Fig. 5B). There were no variables significantly associated with OS in the univariate Cox regression analysis (Table E1 in the supplementary materials, all P > 0.05).\n\nFig. 5Overall survival (OS) curves scaled by histologic LVI status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis\n\nOverall survival (OS) curves scaled by histologic LVI status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis\nAs of February 28, 2021, all patients had completed RFS follow-up, and the median RFS of all patients was 32.0 (23.0-42.8) months, of which 16 of 189 (8.47%) had a tumor recurrence. Among 16 patient with recurrences, 4 patients had locoregional recurrence and 12 patients had distant metastases. Of note, the recurrence rate was 18.6% (8/43) in patients with LVI and 5.48% (8/146) in patients without LVI, with a significant difference between the two groups (P = 0.016). The median RFS was 31.5 (23.0–42.0) months for patients with LVI and 34.0 (22.0-44.8) months for patients without LVI (log-rank test, P = 0.010, Fig. 3 A). Similar results were observed in the combined-predicted LVI model: The median RFS was 31.8 (23.6–58.5) months for patients with combined-predicted LVI presence and 32.0 (21.8–60.0) months for patients with combined-predicted LVI absence (log-rank test, P = 0.007, Fig. 3B). According to the results of the univariate Cox regression analysis (Table E2 in the supplementary materials), LVI (HR = 3.38, P = 0.015), N2 (HR = 6.13, P = 0.007), and the combined-predicted LVI model (HR = 3.61, P = 0.011) were associated with RFS. Multivariate Cox regression analysis showed that N2 (HR = 4.69, P = 0.026) was independent predictors of disease recurrence. A representative case is provided to show the discriminative ability of the combined-predicted model for predicting LVI and RFS (Fig. 4).\n\nFig. 3Recurrence-free survival (RFS) curves scaled by histologic lymphovascular invasion (LVI) status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis\n\nRecurrence-free survival (RFS) curves scaled by histologic lymphovascular invasion (LVI) status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis\n\nFig. 4 A 59-year-old woman with invasive ductal carcinoma of the right breast. Dynamic contrast-enhanced magnetic resonance imaging (A) showing a solid mass in the upper inner quadrant of the right breast, with nonhomogeneous obvious enhancement, Kep value of 1.09 min− 1 (B), and lymph node metastasis to the right axilla (C). The LVI risk calculated by the combined model which was built based on the combination of N stage and Kep was 70.7%. She was confirmed to be LVI positive by histopathology and had multiple metastatic lesions in the right and left liver lobe on follow-up CT (D) 32.5 months after surgery\n\n A 59-year-old woman with invasive ductal carcinoma of the right breast. Dynamic contrast-enhanced magnetic resonance imaging (A) showing a solid mass in the upper inner quadrant of the right breast, with nonhomogeneous obvious enhancement, Kep value of 1.09 min− 1 (B), and lymph node metastasis to the right axilla (C). The LVI risk calculated by the combined model which was built based on the combination of N stage and Kep was 70.7%. She was confirmed to be LVI positive by histopathology and had multiple metastatic lesions in the right and left liver lobe on follow-up CT (D) 32.5 months after surgery\nWhen 98.4% (186/189) of patients had completed OS follow-up, the median OS of all patients was 43.0 (31.5–56.5) months, and the overall death rate was 3.70% (7/189). The median OS was 41.5 (32.0-55.5) months for patients with LVI and 44.0 (31.0-57.3) months for patients without LVI (log-rank test, P = 0.27, Fig. 5 A), which is consistent with the results of the combined-predicted LVI model: The median OS was 42.8 (32.0-59.5) months for patients with combined-predicted LVI presence and 43.5 (30.8–60.0) months for patients with combined-predicted LVI absence (log-rank test, P = 0.970, Fig. 5B). There were no variables significantly associated with OS in the univariate Cox regression analysis (Table E1 in the supplementary materials, all P > 0.05).\n\nFig. 5Overall survival (OS) curves scaled by histologic LVI status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis\n\nOverall survival (OS) curves scaled by histologic LVI status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis", "Finally, 189 women (mean age: 51.1 years; range:25~95 years) were included in this study. Out of 189 included women, 43 had LVI-positive tumors and 146 had LVI-negative tumors. The characteristics of patients in the LVI-negative and LVI-positive groups are shown in Table E1 in the supplementary materials. Those pertinent features are presented in Table 1. The occurrence rates of lymphatic metastasis was more higher in LVI-positive patients than in LVI-negative patients (P = 0.003). In addition, the LVI-positive patients had higher Kep values than the LVI-negative patients (0.92 ± 0.30 vs. 0.81 ± 0.23, P = 0.012, Fig. 1). No significant differences were found for age, tumor size, ER, PR, HER2, Ki-67 index, tumor grade, molecular subtype, T stage, M stage, AJCC stage, Ktrans, Ve, W-in, W-out, or TTP value between patients with and without LVI (all P > 0.05).\n\nTable 1The characteristics of the patients in the LVI-negative and LVI-positive groupsLVI- (n = 146)LVI+ (n = 43)\nP\nOR (95% CI)\nP\nd\nMaximum diameter (cm)2.89 ± 1.123.09 ± 1.180.312b1.16 (0.86–1.55)0.312Minimum diameter (cm)1.85 ± 0.651.92 ± 0.720.528b1.17 (0.70–1.92)0.526ER0.837aNegative50 (34.25%)14 (32.56%)1NAPositive96 (65.75%)29 (67.44%)1.08 (0.53–2.27)0.837PR0.523aNegative70 (47.95%)23 (53.49%)1NAPositive76 (52.05%)20 (46.51%)0.80 (0.40–1.58)0.523HER20.845aNegative103 (70.55%)31 (72.09%)1NAPositive43 (29.45%)12 (27.91%)0.93 (0.42–1.94)0.845Ki-670.409a≤20%47 (32.19%)11 (25.58%)1NA>20%99 (67.81%)32 (74.42%)1.38 (0.66–3.08)0.410T stage0.109cT014 (9.59%)0 (0.00%)0.0 (0.0-3.48 × 10 13)0.988T146 (31.51%)10 (23.26%)1NAT284 (57.53%)32 (74.42%)1.75 (0.81–4.05)0.167T31 (0.68%)0 (0.00%)0.0 (NA-Inf)0.997T41 (0.68%)1 (2.33%)4.6 (0.17–123.2)0.295 N stage\n0.003\nc\nN091 (62.33%)17 (39.53%)1NAN140 (27.40%)15 (34.88%)2.01 (0.91–4.43)0.083N210 (6.85%)7 (16.28%)3.75 (1.21–11.2)\n0.018\nN35 (3.42%)4 (9.30%)4.28 (0.98–17.85)\n0.044\nParametersKtrans(min− 1)0.21 ± 0.110.22 ± 0.120.766b1.58 (0.07–30.11)0.765Kep(min− 1)0.81 ± 0.230.92 ± 0.30\n0.012\nb\n5.52 (1.42–23.3)\n0.016\n\nV\ne\n0.27 ± 0.130.24 ± 0.090.102b0.12 (0.01–2.15)0.163 W-in (min− 1)0.56 ± 0.220.61 ± 0.240.228b2.50 (0.56–11.17)0.228 W-out (min− 1)-0.01 ± 0.02-0.02 ± 0.020.088b0.00 (0.0-9.35)0.090TTP (min)0.69 ± 0.210.64 ± 0.170.165b0.25 (0.03–1.58)0.167Note. Pa: chi-squared test, Pb: Student’s t-test, Pc: Kruskal-Wallis H test, Pd: univariate analysis. Abbreviations: ER = Estrogen receptor; PR = Progesterone receptor;HER2 = Human epidermal growth factor receptor2; TNBC = Triple negative breast cancer; NA = Not available.\n\nThe characteristics of the patients in the LVI-negative and LVI-positive groups\nNote. Pa: chi-squared test, Pb: Student’s t-test, Pc: Kruskal-Wallis H test, Pd: univariate analysis. Abbreviations: ER = Estrogen receptor; PR = Progesterone receptor;HER2 = Human epidermal growth factor receptor2; TNBC = Triple negative breast cancer; NA = Not available.\n\nFig. 1Representative Kep images from breast cancer patients with (A) lymphovascular invasion (LVI) and without (B) LVI. Red represents high values of Kep, yellow represents intermediate values of Kep, and blue represents low values of Kep. The Kep values of the tumors were 1.05 min− 1 and 0.757 min− 1, respectively\n\nRepresentative Kep images from breast cancer patients with (A) lymphovascular invasion (LVI) and without (B) LVI. Red represents high values of Kep, yellow represents intermediate values of Kep, and blue represents low values of Kep. The Kep values of the tumors were 1.05 min− 1 and 0.757 min− 1, respectively", "The ICC values between two readers for Ktrans, Kep, Ve, W-in, W-out, and TTP were 0.994 (95% CI: 0.992–0.995), 0.858 (95%CI:0.815–0.891), 0.984 (95%CI:0.979–0.988), 0.987 (95%CI: 0.983–0.990), 0.979 (95%CI:0.972–0.984), and 0.961 (95%CI:0.949–0.971), respectively, indicating good or excellent reliability. The coefficient of variation on ICC value is 0.057.", "Univariate logistic regression analysis (Table E1 in the supplementary materials) showed that N2 (OR = 3.75, P = 0.018), N3 (OR = 4.28, P = 0.044), and Kep value (OR = 5.52, P = 0.016) were associated with LVI positivity. ROC curves were generated based on the prediction probability of the regression equation using the above variables (Table 2; Fig. 2). The combined-predicted LVI model that incorporated the N stage and Kep yielded a maximum AUC of 0.669 (0.574–0.765), with a cutoff value of 0.276, accuracy of 0.735, sensitivity of 0.512, and specificity of 0.801. Based on the combined-predicted model results, 51 patients were LVI positive and 138 were LVI negative.\n\nTable 2 Performance of the individualized prediction modelsModelsCutoffAUC(95% CI)AccuracySensitivitySpecificityN0.5000.631 (0.542–0.721)0.6190.6050.623\nK\nep\n0.8080.601 (0.502–0.699)0.5450.6740.507Combined0.2760.669 (0.574–0.765)0.7350.5120.801Note: N and Kep indicate the predicted model based on the N stage and Kep, respectively. Combined indicate the predicted model based on the combination of all above parameters. N mean N2/3 stage versus N0/1 stage.\n\n Performance of the individualized prediction models\nNote: N and Kep indicate the predicted model based on the N stage and Kep, respectively. Combined indicate the predicted model based on the combination of all above parameters. N mean N2/3 stage versus N0/1 stage.\n\nFig. 2Receiver operating characteristic (ROC) curve of the model established by N stage, Kep, and combined-predicted model that incorporated N stage and Kep for the prediction of lymphovascular invasion in breast cancer patients\n\nReceiver operating characteristic (ROC) curve of the model established by N stage, Kep, and combined-predicted model that incorporated N stage and Kep for the prediction of lymphovascular invasion in breast cancer patients", "As of February 28, 2021, all patients had completed RFS follow-up, and the median RFS of all patients was 32.0 (23.0-42.8) months, of which 16 of 189 (8.47%) had a tumor recurrence. Among 16 patient with recurrences, 4 patients had locoregional recurrence and 12 patients had distant metastases. Of note, the recurrence rate was 18.6% (8/43) in patients with LVI and 5.48% (8/146) in patients without LVI, with a significant difference between the two groups (P = 0.016). The median RFS was 31.5 (23.0–42.0) months for patients with LVI and 34.0 (22.0-44.8) months for patients without LVI (log-rank test, P = 0.010, Fig. 3 A). Similar results were observed in the combined-predicted LVI model: The median RFS was 31.8 (23.6–58.5) months for patients with combined-predicted LVI presence and 32.0 (21.8–60.0) months for patients with combined-predicted LVI absence (log-rank test, P = 0.007, Fig. 3B). According to the results of the univariate Cox regression analysis (Table E2 in the supplementary materials), LVI (HR = 3.38, P = 0.015), N2 (HR = 6.13, P = 0.007), and the combined-predicted LVI model (HR = 3.61, P = 0.011) were associated with RFS. Multivariate Cox regression analysis showed that N2 (HR = 4.69, P = 0.026) was independent predictors of disease recurrence. A representative case is provided to show the discriminative ability of the combined-predicted model for predicting LVI and RFS (Fig. 4).\n\nFig. 3Recurrence-free survival (RFS) curves scaled by histologic lymphovascular invasion (LVI) status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis\n\nRecurrence-free survival (RFS) curves scaled by histologic lymphovascular invasion (LVI) status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis\n\nFig. 4 A 59-year-old woman with invasive ductal carcinoma of the right breast. Dynamic contrast-enhanced magnetic resonance imaging (A) showing a solid mass in the upper inner quadrant of the right breast, with nonhomogeneous obvious enhancement, Kep value of 1.09 min− 1 (B), and lymph node metastasis to the right axilla (C). The LVI risk calculated by the combined model which was built based on the combination of N stage and Kep was 70.7%. She was confirmed to be LVI positive by histopathology and had multiple metastatic lesions in the right and left liver lobe on follow-up CT (D) 32.5 months after surgery\n\n A 59-year-old woman with invasive ductal carcinoma of the right breast. Dynamic contrast-enhanced magnetic resonance imaging (A) showing a solid mass in the upper inner quadrant of the right breast, with nonhomogeneous obvious enhancement, Kep value of 1.09 min− 1 (B), and lymph node metastasis to the right axilla (C). The LVI risk calculated by the combined model which was built based on the combination of N stage and Kep was 70.7%. She was confirmed to be LVI positive by histopathology and had multiple metastatic lesions in the right and left liver lobe on follow-up CT (D) 32.5 months after surgery\nWhen 98.4% (186/189) of patients had completed OS follow-up, the median OS of all patients was 43.0 (31.5–56.5) months, and the overall death rate was 3.70% (7/189). The median OS was 41.5 (32.0-55.5) months for patients with LVI and 44.0 (31.0-57.3) months for patients without LVI (log-rank test, P = 0.27, Fig. 5 A), which is consistent with the results of the combined-predicted LVI model: The median OS was 42.8 (32.0-59.5) months for patients with combined-predicted LVI presence and 43.5 (30.8–60.0) months for patients with combined-predicted LVI absence (log-rank test, P = 0.970, Fig. 5B). There were no variables significantly associated with OS in the univariate Cox regression analysis (Table E1 in the supplementary materials, all P > 0.05).\n\nFig. 5Overall survival (OS) curves scaled by histologic LVI status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis\n\nOverall survival (OS) curves scaled by histologic LVI status (A) and combined-predicted LVI status (B) with Kaplan-Meier analysis", "In contrast to prior studies, in which simpler qualitative MRI parameters or MRI-based radiomics features were used to evaluate LVI, we developed this novel model based on clinicopathological features and quantitative parameters of DCE-MRI to predict LVI. Our results showed that the N2 stage (OR = 3.75), N3 stage (OR = 4.28), and Kep value (OR = 5.52) were significantly associated with LVI positivity. The combined-predicted model, which was established by Kep and N stage, could preoperatively predict LVI status with an accuracy of 0.735 and a specificity of 0.801. The RFS of the LVI-positive group was significantly lower than that of the LVI-negative group (31.5 vs. 34.0 months, P = 0.010). Similar results were observed in the combined-predicted LVI-positive group and LVI-negative group (31.8 vs. 32.0 months, P = 0.007). The median OS was not significantly different between the LVI-positive and LVI-negative groups (41.5 vs. 44.0 months, P = 0.270) and between the combined-predicted LVI-positive and LVI-negative groups (42.8 vs. 43.5 months, P = 0.970). LVI, N2, and the combined-predicted LVI model were independently associated with disease recurrence.\nThe CDT-VIBE sequence is a combination of volumetric T1-weighted CAIPIRINHA-VIBE imaging, TWIST view-sharing, and Dixon fat separation. The great potential of CDT-VIBE for high tempo-spatial resolution breast DCE-MRI has been evaluated and widely acknowledged [21–25]. The parallel imaging (PI) technology (CAIPIRINHA) can further shorten image acquisition time than conventional PI techniques such as sensitivity encoding, and the image signal-to-noise ratio will not be greatly lost [26]. Before calculating the pharmacokinetic parameters, the image was first subjected to motion correction, which can eliminate motion artifacts such as breathing, and then the motion-corrected images were registered to the structural image to ensure that the corresponding tumor levels in different phases were consistent, thereby ensuring ROI consistency. A typical two-compartment Tofts model was used to calculate the pharmacokinetic parameters, and the arterial input function was selected according to the minimum chi-square value to ensure the accuracy and robustness of the pharmacokinetic parameters.\nThe simpler qualitative parameters, such as internal enhancement pattern, the initial enhancement percentage, maximum enhancement, slope, and kinetic enhancement curve, have been shown significantly correlated with LVI by other investigators, but little attention was paid to the relationship between quantitative parameters of DCE-MRI and LVI. Thus, we developed this novel model to preoperatively predict LVI and clinical outcome of BC patients in this study. Kep, one of the pharmacokinetic parameters of DCE-MRI, is the rate constant of diffusion for contrast agent from the extravascular space into the intravascular space, which depends primarily on the capillary permeability and the permeability surface area [9, 10, 18]. Our study showed that LVI-positive patients had a higher Kep value than LVI-negative patients (0.92 ± 0.30 vs. 0.81 ± 0.23, P = 0.012), and the Kep value was associated with LVI-positive status (OR = 5.52, P = 0.016). This finding is in line with previous findings: LVI strongly correlates with a high peritumoral lymphovascular density and more aggressive neovascularization, and these alterations induce differences in the volume and flow of blood in the tumor microcirculatory environment [2, 6, 9, 27]. However, we found no significant differences in Ktrans or Ve between LVI-positive and LVI-negative patients, both of which are important markers of vascular permeability. A possible explanation may be as follows: Ktrans, Kep, and Ve will be affected by multiple factors, including blood perfusions such as cardiac output, hypertension, the circulatory system of an individual, and other physiologic parameters such as tumor cellularity, capillary bed perfusion, microvessel density of the tumors and their permeability [9, 28]. Moreover, perfusion status, such as cardiac output and blood pressure, can easily cause changes in the flow of contrast agents in the tissue, thereby affecting the stability of pharmacokinetic parameters. Ktrans value measures the joint effect of the plasma flow and tissue permeability properties and is therefore sensitive to these physiological factors. However, Kep tends to be relatively stable in describing actual tumor capillary permeability [28], because it is less sensitive to the absolute value of the contrasts agent concentration. In addition, one thing that probably affect the estimation of Ktrans is the AIF method. Since a population-based AIF is used in this study, it may not be the most accurate one for each individual, and individualized AIF may be more accurate for Ktrans[29]. This may be the reason why we only found a significant difference in Kep but no difference in Ktrans between LVI-positive and LVI-negative groups. Furthermore, another reason for a correlation with Kep but not with Ktrans or Ve in our study could be due to the complexity of the underlying pathophysiology of heterogeneous BC. Thus, our study showed that Kep might be a more stable parameter than Ktrans and Ve, consistent with the findings in a previous study [9]. Also consistent with previous studies, our study showed that LVI is a risk factor for axillary and distant metastasis [14, 15, 29, 30].\nIn the present study, we combined the Kep value and N stage to predict the histopathology LVI status. ROC analysis showed that the combined-predicted LVI model achieved moderate performance in predicting LVI, with an accuracy of 0.735 and a specificity of 0.801. However, the combined-predicted LVI model overestimates the LVI + case (combined-predicted LVI + 51 vs. histological LVI + 43) based on the cutoff point value determined by the largest Youden index. We used the chi-squared test to analyze these data, and the results showed that there were no significant differences (P = 0.405). A similar result was also found in previous studies for a model that predicted LVI in hepatocellular carcinoma (predicted LVI + 176 vs. histological LVI + 115) and gastric cancer patients (predicted LVI + 87 vs. histological LVI + 68) [3, 31]. A potential explanation for this result may be as follow: The cutoff point value determined by the largest Youden index may lead to a higher rate of false-positive for LVI cases. Therefore, more threshold selection strategies and better biomarkers need to be explored to improve the specificity of the prediction model in future research.\nIn this study, the recurrence rate was significantly higher in patients with LVI than in those without LVI (18.6% vs. 5.48, P = 0.016). Multivariate Cox regression analysis showed that LVI (HR = 3.38), N2 (HR = 6.13), and the combined-predicted LVI model (HR = 3.61) were independent predictors of disease recurrence. The median RFS was lower for patients with LVI than for those without LVI (31.5 vs.34.0, P = 0.010), which was also observed in the combined-predicted LVI model (31.8 vs.32.0, P = 0.007). These results were comparable with previous studies. For instance, Ejlertsen et al. [32] found that disease-free survival was statistically significantly associated with LVI within their high-risk group (HR 2.29, P < 0.001). Matsunuma et al. [33] reported that LVI was a significant factor for locoregional recurrence-free survival. However, no significant difference was found for OS between patients with and without LVI or in the combined-predicted LVI model in our study, which was consistent with the results of Rosen et al. [34] and Freedman et al. [35]. Contrary to our findings, Ejlertsen et al. [32] found that OS was statistically significantly associated with LVI. In their study, the cohort had larger and more high-grade tumors, with frequent triple-negative and HER2-positive phenotypes, as compared with our study. These differences might have contributed to the different survival patterns of patients with LVI positive tumors. In addition, we found that N2 but not N3 is an independent predictor associated with RFS, this may be due to fewer patients with N3. In this study, only 9 patients with N3 stage were included in the study, and patients with N3 have a longer median RFS than patients with N2 (32.0 vs. 24.0). Thus, more patients should be included to verify this founding.\nThe present study had some limitations. First, the study results were assessed in a single institution in a relatively large cohort study with 189 patients, and the inclusion of more patients in a multicenter study will make the results more reliable. Second, LVI status was only classified as positive or negative in this study. Our study did not separate LVI into the vascular invasion and lymphatic invasion. A study by Fujii T et al. [36] found that the presence of vascular invasion but not lymphatic invasion could be an indicator of high biological aggressiveness and maybe a valid prognostic factor for BC. Third, we only used 2D ROIs for the evaluation of BC. It should be noted that 3D ROIs may better reflect the perfusion parameters of the tumors. Fourth, the combined model can successfully predict LVI status with high accuracy and specificity, it have the potential to reduce the need test of combined-predicted LVI negative patients. However, low sensitivity of the combined model needed to be addressed in the future according to the different cut-off point to increase sensitivity in further experiments. Furthermore, there are some limitations in term of survival outcome. First, some patients had only 24 months of follow-up, which may affect the strength of prognostic information. Second, LVI status and the combined-predicted LVI model status were predictors of a poor RFS presumably due to reasonable patient numbers and aggressively treat for recurrence patients. The reasons for these parameters were not predictors of OS presumably due to shorter follow-up time, and we will continue to follow up. In addition, there are some technical limitations in our study. First of all, the classical Tofts model of the dedicated software for pharmacokinetic analysis (TISSUE 4D, Siemens Healthcare) is used for the calculation of PKM parameters. Although the Tofts model is one of the most well-known models and widely used in clinical dynamic enhancement studies [37], it has several defects, such as the ignorance of plasma flow in the region of interest. Secondly, Ktrans reflects the result of the combined action of capillary permeability and plasma flow in the tissue [38]. Perfusion status is likely to cause changes in the Ktrans value. Further studies are needed to validate the application of Ktrans and Ve in breast LV I cases. Another technical limitation of our study is the use of a population average arterial input function provided by Tissue 4D. Although we determine the AIF (intermediate type) by the minimum chi-square value of the fitted time concentration curve to ensure the accuracy and robustness of the pharmacokinetic parameters, the population-based AIF is different from the true AIF to some extent, leading to some bias in the results [39].", "In conclusion, the quantitative parameters of Kep from DCE-MRI, and N2 are independent predictors of LVI in BC patients. LVI status, N stage, and the combined-predicted LVI model were confirmed to predict a poor RFS, but no evidence was found that these features were related to OS.", "[SUBTITLE] [SUBSECTION] Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1\nBelow is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1", "Below is the link to the electronic supplementary material.\n\nSupplementary Material 1\n\nSupplementary Material 1" ]
[ null, "materials|methods", null, null, null, null, null, "results", null, null, null, null, "discussion", "conclusion", "supplementary-material", null ]
[ "Neoplasm Invasiveness", "Prognostic factors", "Breast neoplasms", "Dynamic contrast-enhanced magnetic resonance imaging" ]